Skip to main content
Journal of Experimental & Clinical Cancer Research : CR logoLink to Journal of Experimental & Clinical Cancer Research : CR
. 2019 Apr 5;38:146. doi: 10.1186/s13046-019-1154-7

Cancer DNA vaccines: current preclinical and clinical developments and future perspectives

Alessandra Lopes 1, Gaëlle Vandermeulen 1,#, Véronique Préat 1,✉,#
PMCID: PMC6449928  PMID: 30953535

Abstract

The recent developments in immuno-oncology have opened an unprecedented avenue for the emergence of vaccine strategies. Therapeutic DNA cancer vaccines are now considered a very promising strategy to activate the immune system against cancer. In the past, several clinical trials using plasmid DNA vaccines demonstrated a good safety profile and the activation of a broad and specific immune response. However, these vaccines often demonstrated only modest therapeutic effects in clinical trials due to the immunosuppressive mechanisms developed by the tumor. To enhance the vaccine-induced immune response and the treatment efficacy, DNA vaccines could be improved by using two different strategies. The first is to increase their immunogenicity by selecting and optimizing the best antigen(s) to be inserted into the plasmid DNA. The second strategy is to combine DNA vaccines with other complementary therapies that could improve their activity by attenuating immunosuppression in the tumor microenvironment or by increasing the activity/number of immune cells. A growing number of preclinical and clinical studies are adopting these two strategies to better exploit the potential of DNA vaccination. In this review, we analyze the last 5-year preclinical studies and 10-year clinical trials using plasmid DNA vaccines for cancer therapy. We also investigate the strategies that are being developed to overcome the limitations in cancer DNA vaccination, revisiting the rationale for different combinations of therapy and the different possibilities in antigen choice. Finally, we highlight the most promising developments and critical points that need to be addressed to move towards the approval of therapeutic cancer DNA vaccines as part of the standard of cancer care in the future.

Keywords: DNA vaccines, Cancer, Combination therapy, Immunotherapy, Immuno-oncology, Cancer vaccines, Antigens, Neoantigens

Background

Over the last few years, immunotherapy has received increasing attention as a strategy for cancer treatment, and many different approaches are being developed to improve the clinical outcome in cancer patients [1]. The main types of immunotherapy now being used to treat cancer include (i) monoclonal antibodies against specific antigens [2], (ii) immune checkpoint blockade (ICB) to release the “breaks” of T cells [3, 4], (iii) chimeric antigen receptor (CAR) T cell therapy, using a patient’s autologous cells [5], (iv) oncolytic viruses that selectively kill cancer cells and (v) cancer vaccines [68]. Currently, a few immunotherapeutic treatments are commercially available, such as anti-CTLA4, anti-PD1 and anti-PD-L1, CAR T cells against acute lymphoblastic leukemia and B-cell lymphoma, among others. Despite their costs and their immune-related side effects, their success has aroused interest in cancer immunotherapy as a new therapeutic option for cancer patients.

Cancer vaccines represent a promising strategy to induce a specific and long-lasting immune response against tumor antigens (TAs). TAs are mainly proteins overexpressed in the tumor tissue that play a central role in tumor initiation, progression and metastasis [9, 10]. Since the characterization of the first tumor antigen, the melanoma antigen (MAGE) in 1991 [11], a growing number of TAs have been identified. TAs can be classified into 2 main types (Table 1):

  • Mutational antigens. These are derived from mutated self-proteins, which should not be present in normal cells. Some of these genes may be directly related to cancer development (oncogenes and tumor suppressor genes, such as Ras and Bcr-Abl) [12]. In many papers, these antigens are called “tumor-specific antigens” (TSAs). However, this specificity is relative because they can potentially be found in other tumors or even in any altered but nonmalignant cells [13]. Other unique TAs may have or not an association with tumor progression and are the result of the genetic instability of cancer cells. These are classically called “neoantigens”.

  • Tumor-associated antigens (TAAs). These are nonmutated proteins overexpressed or aberrantly expressed in cancer cells [13, 14]. They include products of silent genes, such as oncofetal or cancer/testis antigens, which are not expressed in postnatal tissues or are normally expressed only in placenta and testis; differentiation antigens, which are tissue-specific proteins overexpressed in cancer cells; and universal tumor antigens, which are expressed in low amounts in normal tissues, but overexpressed in cancer [13].

    In the category of TAAs, we can include the oncoviral TAAs, which are non-self TAs and non-human proteins, expressed only by malignant cells transformed after an infection by an oncogenic virus. Examples of oncogenic viruses are human papilloma virus for cervical cancer and Epstein-Barr virus for nasopharyngeal carcinoma [15].

Table 1.

Categories of TAs

TA category TA subtype Examples
Mutational antigens Products of mutated oncogenes (TSA) P53, Ras, Bcr-Abl
Neoantigens Case-specific mutations
Tumor associated antigens (TAAs) Products of silent genes Cancer/testis antigens (a-fetoprotein, MAGE-1, NY-ESO1)
Differentiation antigens Gp100, tyrosinase, Melan-A, MART-1, TRP-1/− 2
Universal tumor antigens Her2/neu, telomerase, survivin
Oncoviral TAAs HPV E6, E7, EBV-latent membrane proteins

In the last few years, different types of cancer vaccines have been developed, i.e., formulations of TAs able to elicit an immune response to arrest the progression of cancer and prevent it from recurring [16]. These include cell-based vaccines, such as dendritic cell vaccines (e.g., Sipuleucel) [17] or whole tumor cells, protein/peptide vaccines [18], viral/bacterial-based vaccines [19, 20] and gene-based vaccines, including RNA and DNA vaccines [7, 21] (Fig. 1).

Fig. 1.

Fig. 1

The different types of cancer vaccines

In this context, DNA vaccination represents a promising strategy for harnessing the immune system. DNA vaccines are plasmids designed to deliver genes encoding TAs, eliciting or augmenting the adaptive immune response towards TA-bearing tumor cells. Cancer DNA vaccines can also induce the innate immune response, acting as powerful “danger signals” that stimulate several DNA-sensing pathways in the cytosol of transfected cells due to the presence of CpG motifs and the double stranded structure itself [22] (Fig. 2).

Fig. 2.

Fig. 2

Innate and adaptive immune activation induced by DNA vaccines

Until now, only one therapeutic cancer vaccine has been approved for human use (DC cancer vaccine, Sipuleucel T) [7], and most of the other cancer vaccines, including DNA vaccines, are still in clinical phase I or II. New developments in plasmid delivery and optimization and the combination with other therapies aim to improve the efficacy of DNA vaccines in preclinical and clinical studies to overcome their low immunogenicity in humans. In this review, we investigate the last 5-year preclinical studies and 10-year clinical studies using non-formulated (naked) DNA vaccines for cancer therapy. We also analyze the strategies that are being developed to overcome the actual limitations in cancer DNA vaccination to predict future trends in this field.

Cancer DNA vaccines advantages and limitations

DNA-mediated immunization began in the 1990s, when a plasmid DNA encoding the influenza A nucleoprotein led to a protective and specific cytotoxic T lymphocyte (CTL) response [23]. Since then, DNA vaccines have been developed to treat a variety of pathologies, including allergies, infectious diseases, autoimmune diseases and cancer. DNA vaccines are based on bacterial plasmids that encode antigens and eventually encoded immunostimulatory molecules (IL-2, GM-CSF, etc.). They can be delivered by a variety of different routes, including intramuscular (IM), intradermal (ID), subcutaneous (SC) and mucosal. The most commonly used delivery strategies are physical methods, such as electroporation [24], sonoporation [25, 26], DNA tattooing [27], or gene gun [28], which are able to overcome the extra and intracellular barriers to transport DNA into the nucleus. Once in the nucleus, the antigen encoded by the DNA vaccine needs to be expressed and presented on major histocompatibility molecules (MHC) for T cell activation. An important advantage of DNA vaccines is that the encoded antigen can be presented by MHC class I and class II, thus activating both CD4 and CD8 T cells and, indirectly, humoral immunity [29]. Furthermore, the intrinsic elements of plasmid DNA can also activate the innate immune response due to the recognition of the double stranded DNA structure by cytosolic sensors [30, 31]. The induction of a protective and specific immune response has been demonstrated in mice against a variety of antigens, including hepatitis B surface and core antigens, HIV Env and Gag antigens, as well as TAs in different cancer models [3235].

The antigen specificity and the safety of cancer DNA vaccines confer some advantages compared to other nonspecific and nontargeted therapies, which could have many side effects and cause extensive damage to normal tissues [9]. Cancer DNA vaccines promote a systemic immune response and thus are also effective on metastases, which are not easily removed by surgical intervention. In addition, unlike antibodies and small molecule inhibitors, DNA vaccines promote immunological memory [36, 37].

However, despite the improvement in the delivery techniques, DNA vaccines revealed poor immunogenicity in human trials [38, 39]. Some molecular approaches have been tested to improve their efficacy, such as codon optimization. This gene engineering technique permits the replacement of synonymous codons to increase protein production and plasmid immunogenicity [37]. Optimized cancer DNA vaccines demonstrated good efficacy in different preclinical models [37], especially in prophylactic models, and showed a great safety profile in humans. Unfortunately, the success in therapeutic vaccination is still limited even in preclinical models [37, 40]. This limitation is mostly due to the different mechanisms of resistance during tumor development, such as a loss or change of epitopes recognized by immune cells, T cell exhaustion, antigen tolerance, and the infiltration of immunosuppressive cells (regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), etc.), which produce immunosuppressive cytokines (TGF-β, IL-10, etc.) and a deprivation of nutrients and oxygen [41, 42]. Hence, new strategies are necessary to completely eradicate tumors.

The analysis of the last 5–10 years of preclinical and clinical trials revealed two main trends. First, a rational choice of the encoded antigen(s) can straighten the DNA vaccine immunogenicity and induce a broad immune response, overcoming the problems linked to antigen loss, modification and tolerance. Second, strategies that combine different therapies to prevent the infiltration of immunosuppressive cells and the production of immunosuppressive cytokines have been implemented to reduce immunosuppression in the tumor microenvironment (TME).

Enhancement of DNA vaccine immunogenicity

Chimeric DNA vaccines

Chimeric DNA vaccines are vaccines that encode xenogeneic antigens. They are proteins or peptides derived from different species in which the sequence is significantly homologous with the self-ortholog [43]. The subtle differences between the epitopes of the orthologue and the native protein elicit T and B cell responses against the xenoantigen [13, 43]. Hence, xenogeneic antigens are recognized as “non-self-antigens”, thus circumventing immune tolerance while preserving an optimal homology to allow T cell recognition [13, 44]. During recent years, different studies have demonstrated the higher efficacy of xenogeneic antigens compared to autologous antigens [44, 45]. A complex DNA vaccine construct that delivers several xenogeneic epitopes dramatically increased the CTL antitumor activity [46]. The efficacy of DNA xenovaccines was also tested in dogs [47], leading to the approval of the first xenogeneic DNA vaccine against human tyrosinase, Oncept, for the treatment of oral malignant melanoma in dogs [43].

It is also possible to design hybrid plasmids, which code for chimeric proteins that include both xenogeneic and homologous antigen domains [13]. In this type of plasmid, the xenogeneic moiety can circumvent immune tolerance and induce a more potent cellular response, while the homologous sequence can stimulate the activation of a broader immune response [43]. Indeed, the chimeric protein produced by transfected cells can be taken up by DCs, thus activating the T cell immune response but it can also be recognized and internalized by B cells [43]. Quaglino et al. found that the plasmid encoding the chimeric neu-Her-2 antigen was superior to both the fully autologous and the fully xenogeneic vaccines in inducing a protective antitumor immune response against ErbB2+ tumors [48]. Starting from these results, other DNA vaccines were constructed by shuffling genes from mouse, rat, human and other species, improving the antigen immunogenicity and vaccine efficacy [4952]. DNA xenovaccination has also been tested in the clinic in melanoma patients, with encouraging results [53, 54], and one clinical study (NCT00096629) using the human and murine prostate-specific membrane antigen is ongoing (Table 2).

Table 2.

Clinical trials in cancer DNA vaccination (2009–2019)

Cancer type Phase Study start DNA vaccine/encoded antigen Combination therapy Treatment schedule DNA delivery References
Breast cancer I 2015 Personalized polyepitope / - Vaccine: 4 mg, at D 1, 29 and 57 IM EP NCT02348320
I 2015 Mammaglobin-A antigen Anastrozole, Letrozole, Tamoxifen, Exemestane, Goserelin, endocrine therapy - Vaccine: 2 injections in the deltoid or lateralis muscles, at D 28, 56 and 84
- Endocrine therapy: to be determined by the treating physicians
IM EP NCT02204098
I 2015 pUMVC3-CD105/Yb-1/SOX2/CDH3/MDM2-polyepitope: mammalian expression vector pUMVC3 + CD105, Y-box binding protein-1, SRY-box 2, cadherin 3, murine double minute 2 rhuGM-CSF, adjuvant therapy - Vaccine: every 28 D for 3 M, then an injection at 6 and 12 M
- rhGM-CSF: ID every 28 D for 3 M
ID NCT02157051
I 2016 pUMVC3-IGFBP2-HER2-IGF1R: pUMVC3 vector + insulin-like growth factor binding protein-2 (IGFBP2), HER2 and insulin-like growth factor 1 receptor precursor (IGF-1R) GM-CSF (Sargramostim), adjuvant therapy Vaccine: D 1 and every 28 D, 3 times
- GM-CSF: the same as the vaccine
ID NCT02780401
I 2018 Neoantigens Durvalumab (anti-PD-L1 antibody), immune therapy - Vaccine: 2 injections in 2 different sites, 3 M after the standard of care (D 1) and then D 29, 57, 85, 113 and 141
- Durvalumab: 1500 mg every 4 W, at D 85
EP NCT03199040
Prostate cancer II 2009 pTGV, encoding prostatic acid phosphatase (PAP) rhGM-CSF, adjuvant therapy - Vaccine: 100 μg, every 2 W for the first 12 W and then every 12 W, according to the immune response
- rhGM-CSF: 200 μg every 3 M
ID NCT00849121
II 2011 pTGV-HP rhGM-CSF, adjuvant therapy - Vaccine: 100 μg, every 2 W for 6 times and then every 3 M for 2 years
- rhGM-CSF: the same as the vaccine, but 208 μg of dose
ID NCT01341652
II 2013 pTGV-HP Sipuleucel-T, autologous peripheral blood mononuclear cells with antigen presenting dendritic cells that have been activated ex vivo with a recombinant fusion protein (PA2024) consisting of PAP linked to GM-CSF, immune therapy - Vaccine: at W 6, 8, 10, 12, M 6 and 12
- Sipuleucel-T: W 0, 2 and 4
/ NCT01706458
I 2015 pTGV-AR GM-CSF, adjuvant therapy - Vaccine: 100 μg, 6 doses every 2 W and then 12, 24, 36 and 48 or 100 μg at W 0, 2, 12, 14, 24, 26, 36, 38, 48, 50
- GM-CSF: 200 μg co-injected with the vaccine
/ NCT02411786
I/II 2015 pTGV-HP Pembrolizumab (anti-PD1 antibody), immune therapy; rhGM-CSF, adjuvant therapy - Vaccine: 100 μg, at D 1, 15, 29, 43, 57 and 71
- Pembrilizumab: 2 mg/kg, IV, every 3 W, at D 1, 22, 43, 64 or D 85, 106, 127 and 148
- rhGM-CSF: 208 μg, ID, every 3 W
ID NCT02499835
II 2018 pTGV-HP Nivolumab,
GM-CSF
- Nivolumab: 240 mg IV every 2 W × 6 beginning D 1, then every 4 W × 9 beginning W 12)
- rhGM-CSF: 208 μg, ID every 2 W × 4 beginning W 4, then every 4 W × 9 beginning W 12
- pTVG-HP: 100 μg, every 2 W × 6, beginning at D 1; then every 4 W × 9, beginning at W 12
ID NCT03600350
I 2018 Neoantigens Nivolumab/Ipilimumab and Prostvac - Priming with Prostavac (1 mg/kg, every 3 W, 2 doses) and the ICB (3 mg/kg, every 3 W, 6 doses)
- Vaccine: 4 mg at 2 different sites, 6 times every 28 D
IM EP NCT03532217
Cervical cancer I/II 2015 VB10.16: composed by E6/E7 antigen of HPV16 + dimerization entity + APC binding protein / - Vaccine: 3 mg, W 0, 3, 6 or W 0, 4, 12 Lateral deltoid mucle NCT02529930
II 2015 GX-188E, encoding E6/E7 fusion protein of HPV 16 and 18, plus the immune-enhancer, Fms-like tyrosine kinase-3 ligand (FLT3L) / - Vaccine: 1 mg, W 0, 4 and 12 IM NCT02596243
I/II 2017 MEDI0457 = INO3112 = VGX-3100 (, encoding E6 and E7 proteins of HPV types 16 and 18) + INO-9012 (hIL-12) Darvalumab (anti PD-L1 antibody), immune therapy - Vaccine: /
- Darvalumab: 1500 mg, IV, every 4 W
IM EP NCT03162224
/ 2017 GX-188E GX-I7 encoding IL7 receptor agonist, Imiquimod, adjuvant therapy - Vaccine: 1 mg, 3 times
- GX-I7: 3 mg, locally on the cervix, 4 times
- Imiquimod: 12.5 mg, administered locally on the cervix, 8 times
IM NCT03206138
III 2017 VGX-3100 / - Vaccine: 1 ml on D 0, W 4 and W 12 IM EP NCT03185013
I/II 2018 GX-188E Pembrolizumab - Vaccine: 1.0 mg/0.5 ml
- Pembrolizumab: 100 mg/4 mL, IV
IM EP NCT03444376
II 2018 VGX-3100 / - Vaccine: over 10 s for 4 doses in W 0, 4, 12, and 24 IM EP NCT03603808
II 2018 VGX-3100 Darvalumab - Vaccine: W 1, 3, 7, and 12
- Darvalumab: W 4, 8, and 12
IM EP NCT03439085
III 2019 VGX-3100 / - Vaccine: 1 ml on D 0, W 4 and W 12 IM EP NCT03721978
Ovarian cancer I 2012 pUMVC3-hIGFBP2 multiepitope: mammalian vector pUMVC3 + human IGFBP2 / - Vaccine: monthly, for 3 M ID NCT01322802
II 2017 pUMVC3-hIGFBP2 multiepitope Carboplatin, Paclitaxel, chemotherapy - Vaccine: 2 W after the chemotherapy and every 3 W
- Chemotherapy: IV, 2 W before the vaccine
ID NCT03029611
Pancreatic cancer I 2018 Personalized neoantigen: pING vector + prioritized neoantigens + mesothelin epitopes Chemotherapy - Vaccine: W 1, 5, 9, 13, 17 and 21
- Chemotherapy: at W 1, 5, 9, 13, 17, 21, 25 and 77, after surgery and before vaccination
IM EP NCT03122106
Glioblastoma I/II 2018 INO-5401 (3 separate DNA plasmids targeting Wilms tumor gene-1 (WT1) antigen, prostate-specific membrane antigen (PSMA) and human telomerase reverse transcriptase (hTERT) genes) Cemiplimab, radiation and chemotherapy; INO-9012 - Vaccine: 3 mg at D 0, every 3 W × 4 doses, and then every 9 W
- INO-9012: 1 mg at the same time of the vaccine
- Cemiplimab: IV, every 3 W at a dose of 350 mg per dose
- Radiation therapy: 42 days after surgical intervention, and should start approximately 2 W after D 0
- Temozolomide: daily during radiation therapy, at a dose of 75 mg/m2
IM EP NCT03491683
Melanoma Early I 2018 IFx-Hu2.0 coding for Emm55 Streptococcal Antigen / - Vaccine: 100 μg in 200 μL per lesion Intralesion NCT03655756
Renal cell carcinoma II 2019 Neoantigens Darvalumab
Tremelimumab
- Durvalumab: IV at a dose of 10 mg/kg over the course of 60 min, every 2 W, 8 doses
- Tremelimumab: at a dose of 1 mg/kg over the course of 60 min, every 4 W, 4 cycles
- Vaccine: D 1 of the first 28 D cycle of treatment with durvalumab and tremelimumab, every 2 W
IM EP NCT03598816
Solid tumors I 2014 hTERT / - Vaccine: 100, 400 and 800 μg as a single agent, every 4 W × 3 cycles ID EP NCT02301754
Anal neoplasm II 2018 VGX-3100 / - Vaccine: 1 ml on D 0, W 4 and W 12, and potentially W 40 IM EP NCT03499795
Urothelial carcinoma I/II 2018 INO-5401 INO-9012, Atezolizumab - Vaccine: 9 mg, every 3 W × 4 doses then every 6 W × 6 additional doses, thereafter every 12 W
- INO-9012: administered with the vaccine
- Atezolizumab: IV infusion every 3 W
IM EP NCT03502785

M = month; W = week; D = day; EP = electroporation; IM = intramuscular; ID = intradermal; IV = intravenous

Neoantigen DNA vaccines and personalized vaccination

Most anticancer DNA vaccines, both past and present, immunize using nonmutated TAs. However, these antigens are often present in normal or germline tissues, which can prevent a strong immune activation because of immune tolerance [55]. Several clinical trials using nonmutated TAs have failed to demonstrate beneficial effects compared with the standard of care treatment [14]. In contrast, neoantigens are the result of tumor-specific DNA alterations that create new epitopes. Due to their specific expression in cancer tissue and the potential lack of side effects, they represent ideal targets against cancer and can be used in the design of cancer vaccines [56, 57]. They can also turn “cold” tumors into “hot” ones and mediate the upregulation of PD-L1 in the TME, thus extending the applicability of the anti-PD-1/PD-L1 immunotherapy [58]. Neoantigens are presented by APCs to CD4+ and CD8+ T cells to activate an immune response. They are highly tumor-specific and, therefore, they represent an attractive immunotherapy target. It is expected that they are not affected by T cell tolerance, as they may be recognized as non-self by the host immune system and, thus, generate a specific anti-tumor response [59, 60]. Their identification starts with exon sequencing from a tumor biopsy. Then, mutations are identified compared to whole exome data from normal tissue. Prediction algorithms select those antigens that are recognized by MHC class I or II. Finally, in vitro and in vivo studies validate their ability to stimulate the CD8+ immune response, especially a CD4 response [61, 62]. However, not all peptides are immunogenic, and identifying which mutations are targeted by the immune system is currently a subject of great interest. Hence, the prediction of the immune response to neoantigens needs to be optimized. Assessing the immunogenicity of each neoepitope is not reasonably applicable on a large scale. Current computational approaches are being refined to improve the accuracy of neoantigen identification and are discussed in detail in [63]. Integrated pipelines will need to be developed beginning with tumor genomic characterization, variant analysis, and the accurate prediction of which mutations are likely to give rise to tumor-specific neoantigens [64]. Other hurdles are associated with the use of personalized neoantigens for cancer immunotherapy, such as the manufacturing time. The median period for the discovery and production of a personalized vaccine is approximately 4.5 months [65]. In particular, the time from the selection of mutations to vaccine release ranges from approximately 89–160 days [66]. This amount of time has to be reduced to cure patients with metastatic disease. Another issue concerns the genetic heterogeneity of tumors [67]. Thus, targeting a unique neoantigen would probably lead to the selection of antigen non-expressing tumor cells. It has been demonstrated that the use of a poly-epitope neoantigen RNA vaccine encoding up to 10 neoantigens was effective in 8/13 melanoma patients who were completely tumor-free after one year [66]. Compared to RNA and peptide vaccines, DNA vaccines seem to elicit a more potent CD8 response against the encoded neoantigens, making them more attractive for cancer vaccination [60, 68]. Hence, once identified, the neoantigen can be cloned into a DNA vaccine. This personalization permits the design of cancer vaccines tailored to each patient.

Polyepitope DNA vaccines

An advantage of DNA vaccines is the possibility of delivering several antigen genes in the same construct, at the same time and with the same delivery method. The presence of immunodominant and unconventional epitopes simultaneously delivered by a polyepitope DNA vaccine can induce a broad CTL response specific to multiple antigens [69]. In this way, it is possible to overcome the antigen mutation or deletion by tumor cells, the variation or absence of the appropriate T cell repertoire and the MHC haplotype in patients [69].

When designing a poly-epitope DNA vaccine, many parameters should be considered. First, the competition for antigen recognition at the surface of the APC and the affinity of the selected epitopes for MHC molecules should be considered [70, 71]. Palmowski et al. demonstrated that the use of an MHC class I polyepitope vaccine leads to the preferential expansion of CTLs with a single immunodominant specificity [72, 73]. In addition, the affinity of the selected epitopes for MHC molecules and transporters could influence the CTL immunodominance and the consequent immune response [70].

Second, although the CD8 T cell response has been considered to be the main protagonist in the antitumor immune response resulting from vaccination, the insertion of an epitope/antigen recognized by CD4 T cells into a DNA vaccine could activate a broader and stronger immune response. Several studies suggest the importance of the CD4 T cell population for cancer immunotherapy [74, 75]. Recently, it has been demonstrated that CD4 T cells recognize a higher number of neoantigens than previously known and can generate potent antitumor responses [62, 76]. Hence, a coordinated CD4 and CD8 response is necessary for the complete eradication of a tumor [76]. T helper (Th) peptides have already been used in combination with DNA vaccines to increase the activation of Th cells, thus further eliciting the CTL immune response [7782]. An example of a Th epitope is the pan DR epitope (PADRE). This synthetic Th epitope, encoded in a DNA vaccine and administered with an antigen-encoding plasmid, increased the number of antigen-specific CD8 T cells, resulting in potent protective and therapeutic antitumor effects [83]. Other studies demonstrated that a PADRE-encoding DNA generated CD4 Th1 cells that play an important role in maintaining long-term memory responses, helping the activity of CD8 T cells [84].

Many techniques have been developed to find new epitopes. These studies led to the identification of NY-ESO-1, MelanA/MART-1, SSX4, MELOE-1 and TRAG-3 in melanoma, EphA2 and MAGE-6 in renal cell carcinoma, CEA, MAGE-3 and telomerase in lung carcinoma, TRAG-3 in breast carcinoma, and NY-ESO-1, p53 and SSX4 in ovarian cancer, among others [85]. Some of these tumor antigens recognized by CD4 T cells belong to the same categories as those recognized by cytotoxic CD8 T cells [75].

Finally, it is important to identify the most immunogenic epitopes derived from tumor antigens. New in silico techniques are being developed to improve the prediction of epitope immunogenicity to design a poly-epitope vaccine. They not only consider the binding affinity to the MHC and the different HLA subtypes but also the conformation and interaction with the HLA, immunodominance vs tolerance, etc. [86]

Many recent preclinical studies have investigated the use of polyepitope DNA vaccines to reach a broad immune response. As a result, an increased IFNg production, a higher Th and CTL response [86, 87], and a general decrease in the tumor growth rate and metastasis formation were observed in different types of cancer models [88, 89]. Some preclinical studies focus on the HPV model, using DNA vaccines encoding E6 and E7 molecules [90], or E7 with a helper epitope [88]. Another example is SCT-KDR2, which encodes the mouse β2microglobulin + KDR2 (VEGFR2 antigen peptide) + MHC class I H-2Db, in a B16 melanoma tumor model [89]. A non-exhaustive list of the most recent preclinical trials (in the last 5 years) can be found in Table 3. Additionally, many clinical trials are testing the safety and efficacy of polyepitope DNA vaccines, such as NCT02348320 and NCT02157051 for breast cancer, NCT02172911 for cervical cancer, and NCT01322802 and NCT03029611 for ovarian cancer. In particular, in the clinical studies NCT02348320 and NCT03199040, a personalized polyepitope vaccine against breast cancer is being used, as well as in the NCT03122106 for pancreatic cancer, and the results will help to establish the relevance of this vaccine strategy. This would address tumor heterogeneity and the loss of immunogenicity associated with TAAs, which accounts for the failure of the current anticancer treatments [58]. A complete list of the ongoing clinical trials could be found in Table 2.

Table 3.

Preclinical studies in cancer DNA vaccination (2015–2018)

Cancer type Animal DNA vaccine Combination therapies Protocol DNA vaccine delivery Results Year, ref.
Cervical cancer (TC-1 cells) C57BL/6 mice HPV plasmid encoding E6 and E7 antigens pVAX1-ISG15 encoding an optimized mouse adjuvant ISG15 Therapeutic vaccination
Tumor: 5 × 104 TC-1 tumor cells, SC
Vaccine and adjuvant: 7 D after tumor implantation, followed by 3 boosts weekly
IM EP, in the tibialis anterior muscle - Strong HPV E7-specific CD8+ T cell immune response
- Increase in INF-γ secretion
- 6/10 mice were tumor-free at D 42
2015 [90]
Cervical cancer (TC-1 cells) C57BL/6 mice pcDNA3.1-E7, encoding E7 antigen of HPV16 Monophosphoryl lipid A (MPL, TLR4 agonist) and α-galactosylceramide (GalCer) Therapeutic vaccination
Tumor: 2 × 105 TC-1 tumor cells, SC
Vaccine: 100 μg, 7 D after tumor implantation, followed by 2 boosts weekly
Adjuvant: 25 μg of MPL and 1 μg of GalCer, SC with the vaccine
SC - CTL-specific cytolytic activity
- Higher INF-γ, IL-4 and IL-12 production
- Decrease in tumor growth if both adjuvants were administered
2016 [136]
Cervical cancer (TC-1 cells) C57BL/6 mice HELP-E7SH, encoding E7 antigen of HPV16 and a helper epitope to stimulate CD4+ response Abs against CD70, CTLA-4, PD-1; agonistic Ab to CD27 Therapeutic vaccination
Tumor: 1 × 105 TC-1 tumor cells, at D 0 before the vaccination, SC
Vaccine: 15 μl of a 2 mg/ml DNA solution, on D 0, 3 and 6
Abs: 100 μg, at D 0, 3 and 6 (CD70 also at D 9), IP
Tattoo (intraepidermal vaccination) - Help epitopes increased E7-specific CD8+ response in lymph nodes and spleen
- CTLA4 and PD-1 did not promote CTL priming, if not combined with CD27 or the vaccine
- CD27 agonism + anti-PD1 improved mice survival, albeit CD27 + anti-CTLA4 further increased the CTL response
2016 [35]
Cervical cancer (TC-1 cells) C57BL/6 mice pVAX1-gDE7, encoding HPV-16 E7 protein fused to HSV-1 gD protein pcDNA3-IL2 encoding murine IL-2; anti-Gr1 Ab Therapeutic vaccination
Tumor: 7.5 × 104 TC-1 tumor cells, at D 0 before the vaccination, SC
Vaccine and adjuvant: 50 μg alone or in combination with 50 μg of the adjuvant, at D 3, 2 doses, weekly
Anti-Gr1: 200 μg, once/W for 6 IP injections, at D 7–10, 5 doses, weekly
IM - Vaccination with the 2 plasmids avoided MDSC accumulation
- Combination of the vaccines and anti-Gr1 antibody increased mice survival, completely eradicating the tumor
2016 [95]
Cervical cancer (TC-1 cells) C57BL/6 mice pcDNA3.1-E7, encoding HPV-16 E7 antigen melatonin Therapeutic vaccination
Tumor: 2 × 105 TC-1 cells at D0, before the vaccination, SC
Vaccine: 90 μg, 3 times, at 7 D interval
Melatonin: 50 or 100 mg/kg
SC - Production of HPV16 E7-specific CTL
- Increase of IFNg and TNFa in the TME
- Tumor volume reduction
2018 [34]
Cervical cancer (TC-1 cells) C57BL/6 mice dbDNA, encoding HPV16 E6 and E7 / Therapeutic vaccination
Tumor: 5 × 104 TC-1 cells, SC, at D0
Vaccine: at D3, 25 μg/hind, boost after 7 W
IM in the anterior tibialis and IM EP in the quadriceps - Delay in the tumor growth
- High levels of IFNg-secreting Th cells
- Production of IgG1
- IL-12 production and low IL10
2018 [137]
Cervical cancer (HPV) BALB/c mice pNGVL4a-hCRTE6E7L2, expressing the HPV16 E6, E7 and L2 antigens / Prophylactic vaccination
Vaccine: 3 injections, biweekly
Tumor: 12 μl of HPV16 PsV, 19 D after the last vaccination
IM EP - Production of Ab against E6, E7 and L2;
- Protection of 3/5 of mice from the challenge, but without a significant difference compared to the control group
2017 [138]
Lobular carcinoma (TUBO cells) BALB/c mice pAmot, coding human p80 Amot (Angiomotin), antiangiogenic / Therapeutic vaccination
Tumor: 105 TUBO cells, SC
Vaccine: 50 μg, at D 7
IM EP in the quadriceps muscle - Delay in tumor progression
- Heterogeneous changes in the tumor region following antiangiogenetic treatment
2015 [139]
Murine breast cancer (D2F2 cells) BALB/c mice pVAX-E2A, encoding Her2/neu antigen pVAX-CCL4, encoding CCL4, chemoattractant for immune effector cells Prophylactic vaccination
Vaccine: 2 × 100 μg, on D 1 and 15
Tumor: 2 × 105 Her2/neu+ cells, on D 25, SC
IM - With the combined therapy, 26% of mice remained tumor-free (CCL4 improved tumor protection)
- CCL4 produced a Th1 anti-Her2/neu response
2016 [140]
Murine breast cancer (4 T1 cells) BALB/c mice CpVR-FAP, encoding fibroblast associated protein (FAP) Cyclophosphamide, chemotherapy agent Therapeutic vaccination
Tumor: 2 × 104 4 T1 cells, at D 0, SC
Vaccine: 100 μg, on D 2, 9 and 16 after tumor injection
Cyclophosphamide: 50 mg/kg, on D 1, 8, 15, IP
IM in the tibialis anterior muscle - Combination therapy increased median survival time of mice
- Suppression of IL-10, VEGFα and CXCL12 mRNA expression
2016 [115]
Murine breast cancer (4 T1 cells) BALB/c mice CpVR-FAP, encoding FAP / Prophylactic and therapeutic vaccination
Tumor: 2 × 104 4 T1 cells, SC
Vaccine: 100 μg, on D 2, 9 and 16 after tumor injection or 3 times every 2 W before tumor injection
IM in the tibialis anterior muscle - Specific CTL response against FAP
- Increased IL-2 production
- Delay of the tumor growth also in therapeutic setting
- Decrease in FAP expression without impairing wound healing
2016 [141]
Murine breast cancer (4 T1 cells) BALB/c mice pVAX1-mCr-1, encoding mouse Cripto-1 oncofetal protein / Prophylactic vaccination
Vaccine: 40 μg
Tumor: 2 × 105 4 T1 mCr-1 cells, W12
ID EP - Humoral response against Cr-1
- Protective immune response against cancer stem cells
- Reduced lung metastasis
2018 [142]
Colon cancer (colon 26/β-gal cells) BALB/c mice pcDNA3/β-gal encoding β-galactosidase pCAGGS/FasL encoding Fas ligand Prophylactic vaccination
Vaccine and adjuvant: 50 μg + 1 μM cardiotoxin to facilitate DNA uptake
Tumor: 106 Colon 26/β-gal cells, 21 D after vaccine injection, SC
/ - The combined therapy decreased tumor growth rate
- Production of Abs anti-β-gal
2015 [143]
Colon cancer (CT26/HER2 cells) BALB/c mice pVAX1-HER2, coding HER2 antigen Gemcitabine, chemotherapy agent; anti-Gr1 antibody; anti-PD-L1 Ab Prophylactic and therapeutic vaccination
Tumor: 3–5 × 105 CT26/HER2 cells, SC
Vaccine: 50 μg
Anti-PD-L1 and Gr-1 Ab: 200 μg and 250 μg, respectively, IP
Gemcitabine: 75 μg/g, 2 times/W, IP
IM EP - In prophylactic vaccination, the combination of vaccine + anti-PD-L1 Ab failed to delay tumor growth
- The addition of anti-Gr1 or gemcitabine delayed tumor growth
2017 [144]
Colon cancer (CT26 cells) Balb/c mice CpVR-MS and CpDV-IL2-MS, encoding a fusion gene of human surviving S8 and human 33 MUC1, plus IL2) Ad-MS (Adenovirus) Therapeutic vaccination
Tumor: 106 CT26 cells, SC
Vaccine: 100 μg, twice
Ad-MS: 108 pfu, D1, 15 and 29
IM - Specific immune response in splenocytes
- Upregulation of CCL-19 and GM-CSF
- Downregulation of PD-L1 and MMP-9
2018 [145]
Colorectal cancer (CT- 26/NIS cells) BALB/c mice pcDNA-hNIS, expressing human sodium/io- dide symporter (hNIS) / Vaccine: 100 μg, 3 times at 2 W intervals
Tumor: 2 W after the final hNIS DNA injection, 5 × 105 (left) or 1 × 105 (right) CT-26/NIS cells, SC
ID - Increase of IgG2a/IgG1 ratio
- Increase of INF-g secreting cells and IFN-g production
- Th1 response
- Slower tumor growth
2018 [146]
Melanoma (B16F10-β-hCG cells) C57BL/6 mice CAVE = pSVK-VEGFR2-GFc-IL12, Semliki Forest Virus expressing VEGFR2 and IL-12 CAVA = SFV replicon DNA vaccine targeting surviving and hCG antigens Prophylactic vaccination
Vaccine: 10 μg of CAVA/CAVE, 3 times at 10 D of interval
Tumor: 7.5 × 104 B16F10-β-hCG cells, 7 D after the last immunization, SC
IM EP - Combination of the 2 vaccines delayed tumor growth more efficiently than the single vaccine and increased mice survival
- CAVE + CAVA decreased microvessel density
2015 [147]
Melanoma (B16F10 cells) C57BL/6 mice pSPD-gp100-CD40L, encoding gp100 inserted between mouse Surfactant Protein D (SPD) and CD40L pIL-12, encoding IL-12p70; pcDNA3.1-GM-CSF encoding GM-CSF Therapeutic vaccination
Tumor: 5 × 104 B16F10 cells, ID
Vaccine and adjuvants: 80 μg vaccine + 20 μg of each adjuvant plasmid, on D 3, 10 and 17
IM in hind quadriceps muscles - Vaccine alone did not delay tumor growth, but the combination with the 2 adjuvants was very effective in increasing mice survival 2015 [94]
Melanoma (B16F10 cells) C57BL/6 mice pVAX1-MUCI, encoding mucin I glycoprotein pVAX1-Flt3L, encoding Fms-like tyrosinase 3-ligand Therapeutic vaccination
Tumor: 1 × 106 B16F10 cells, SC
Vaccine + adjuvant: 50 μg, priming when tumors were palpable, boosts after 7 and 14 D
IM EP - Specific CTL and antibodies
- Tumor growth suppression
2018 [148]
Melanoma (B16 cells) C57BL/6 mice p-mBAP31 and p-LAMP/mBAP31 = p43 and p43- Lysosomal Associated Membrane Protein (LAMP) vectors + mouse B-cell receptor-associated protein (mBAP) / Therapeutic vaccination
Tumor: 5 × 104 B16 cells, at D 0, SC
Vaccine: 50 μg, at D 3, 10, 17 and 24
SC - No evidence of autoimmune disorders
- High INF-γ production, especially using LAMP vaccine
- LAMP vaccine increased the CTL cytotoxicity
- Suppression of tumor growth, especially using LAMP vaccine
2015 [149]
Melanoma Horses Minimalistic immunogenically defined gene expression (MIDGE)-Th1 vector + eqIL12 and IL-1beta receptor antagonist protein (ILRAP)-eqIL18 hgp100MIDGE-Th1; htyrMIDGE-Th1 Therapeutic vaccination
Tumor: horses were already affected by melanoma
Vaccine and adjuvants: 500 μg ID peritumorally and 500 μg IM into the semimembranosus muscle, 3 times
ID and IM - Vaccine was safe and well-tolerated, except an increase in the body temperature on the day after injection and signs of acute inflammation
- Tumor volume was reduced by 28.5%, but without significant differences if adjuvants were added
2015 [150]
Melanoma (B16F10-OVA cells) C57BL/6 mice pVAX2-OVA, encoding ovalbumin; pVAX2-gp100, encoding gp100 pVAX2-HIV-1 Gag Prophylactic and therapeutic vaccination
Vaccine: 1 μg (p-OVA) or 50 μg (p-gp100), at D 2, 9 and 16 (therapeutic) or 3 times every 2 W before the tumor challenge (prophylactic)
Adjuvant: 1 μg, co-administered with the vaccine
Tumor: 1 × 105 B16F10-OVA cells at D 0 (therapeutic) or 2 W after the last vaccine injection (prophylactic)
IM EP - Delay of tumor growth and increase in mice survival
- Codelivery of the adjuvant-encoding plasmid polarized the immune response towards a Th1-like phenotype
2016 [40]
Mastocytoma (P815 cells) DBA/2 mice Differently optimized pVAX2-P1A vaccines, encoding P815A / Prophylactic and therapeutic vaccination
Vaccine: 50 μg, at D 2, 9 and 16 (therapeutic) or 3 times every 2 W before the tumor challenge (prophylactic)
Tumor: 1 × 106 P815 cells at D 0 (therapeutic) or 2 W after the last vaccine injection (prophylactic)
IM EP - Delay of tumor growth and increase in mice survival
Activation of innate immunity related to the different CpG motif amount inside the P1A gene
2017 [37]
Mastocytoma (P815 cells) DBA/2 mice Optimized pVAX2-P1A vaccine, encoding P815A Anti-CTLA4, anti-PD1 Therapeutic vaccination
Tumor: 1 × 106 P815 cells at D 0
Vaccine: 50 μg, at D 2, 9 and 16
Anti-CTLA4, anti-PD1: 100 μg at D 3, 6 and 9
IM EP - Survival reached 90%
- Increase of specific T cell infiltration in the TME
- Increase of IL-12 production
- Decrease of metastasis formation
2018 [109]
Malignant tumor HLA-A2.1/Kb transgenic mice p-GST-YL66, against multiepitope YL66 (from COX2 and MAGE4), linked with membrane permeable Tat-PTD and the universal Th epitope / Not available / - CTL-mediated tumor cell lysis in vitro and in vivo 2017 [87]
Colorectal cancer (MC32 cells) C57BL/6 mice Pc-DNA3-CEA, carcinoembryonic antigen (CEA) Ab 4-1BB Prophylactic and therapeutic vaccination
Tumor: 1–5 × 105 MC32 cells, SC
Vaccine: 50 μg, at 1 W of interval
Ab anti-4-1BB: 50 μg, systemically, after vaccine injection
IM EP - Antigen-specific CTL activity and tumor-protective immune response in prophylactic model
- Ab 4-1BB increased CTL lytic activity
- MC32 cells resisted to CEA DNA vaccination by loss of antigen presentation to CEA-specific CTL in therapeutic model
2015 [151]
Melanoma (B16), carcinoma (3LL) C57BL/6 mice SCT-KDR2, encoding the mouse β2microglobulin + KDR2 (VEGFR2 antigen peptide) + MHC class I H-2Db, subcloned into pdDNA3.1 / Prophylactic vaccination
Vaccine: 50 μg, 3 times, at 1 W of interval
Tumor: 105 B16 cells or 2 × 105 3LL cells, 10 D after the last vaccination, SC
ID - CTL response to VEGFR2
- Inhibition of tumor-induced angiogenesis
- Inhibition of tumor metastasis
2015 [89]
Sarcoma HHDII-DR1 mice SSX2-optimized vaccine, encoding modified cancer testis antigen Ab anti-PD-1/L1 Prophylactic and therapeutic vaccination
Vaccine: 100 μg, 6 times, every 2 W (prophylactic) or weekly the day after the tumor injection (therapeutic)
Tumor: 2 × 104 SSX2-expressing sarcoma cells
An anti-PD1/L1: 100 μg, IP on the day following each vaccination
ID - Optimized vaccine elicited inferior antitumor effect relative to the native vaccine
- Increase of PD-L1 expression on tumor cells
- CTL from immunized mice expressed more PD-1, increasing the antitumor efficacy of the combination with ICB
2015 [152]
Kidney cancer (RenCa cells) BALB/c mice pVAX1-G250-F2A-CTLA4, containing the co-expression gene G250-CTLA4, linked by Furin-2A (F2A) / Therapeutic vaccination
Tumor: 105 RenCa cells, SC
Vaccine: 50 μg, at D 7, 17 and 27
EP - Humoral and cellular-specific immune response against CTLA4 and G250
- Increase in INFγ and IL-4 (Th1/2 response)
- Tumor growth rate decreased
2017 [153]

Keywords search “cancer plasmid DNA vaccine”, from 2015 to 2018. D = day, W = week, M = month

A good option to further optimize the efficacy of cancer DNA vaccination could be the combination of the 3 cited approaches, designing a poly-epitope chimeric vaccine containing specific neoantigens. In the clinic, this could reduce the number of nonresponding patients by developing a stronger and more complete immune response.

Combination of DNA vaccines with other therapies

In the analyzed preclinical (Table 3) and clinical (Table 2) studies, DNA vaccines can delay tumor growth and elicit a strong immune response, especially an antigen-specific CTL response, but are rarely able to completely reject the tumor. These modest gains were reached by optimizing DNA vaccines in several aspects, such as plasmid design and delivery and administration strategies [1, 9, 37, 91] However, DNA vaccines alone are not able to overcome the tumor immune escape caused by the natural selection of tumor cell clones lacking immunogenic antigens or by immunosuppressive cells that are recruited to the TME (MDSCs, Tregs among others), which lead to the exhaustion of T effector cells [7]. Cancer DNA vaccines can reach their optimum efficacy if combined with other strategies that can not only potentiate the antigen response but also silence immunosuppression in the TME [92].

There is evidence that combining therapeutic cancer vaccines with traditional modalities (radiotherapy, chemotherapy, surgical removal) may be synergistic. The combination therapies already tested in clinical and preclinical studies can be summarized as follows:

Cytokines/adjuvants

Immunostimulatory cytokines can increase the effect of the vaccine on effector T cells. They are generally encoded by the antigen-encoding vaccine, by another plasmid or injected as proteins in combination with the vaccine. In recent studies, the most commonly used cytokines include IL-2, IL-12 and GM-CSF. IL-2 is involved in the differentiation of immature T cells into both Tregs and effector T cells. Its great efficacy against metastatic melanoma and metastatic renal cell carcinoma led to its approval by the FDA [7, 93]. IL-12 is another important cytokine involved in T cell activation and effector function, and its combination with a vaccine increases the vaccine’s efficacy [94]. A plasmid encoding IL-12 combined with a DNA vaccine against cervical cancer promoted mouse survival and decreased the number of MDSCs in the TME [95]. GM-CSF is used in many clinical trials (Table 2) for its activity on DC maturation and T cell activation and proliferation. However, this molecule can also attract MDSCs, and it is not clear how this cytokine balances between immune activation and inhibition in vivo. Current clinical studies are seeking to answer this question [7]. Other cytokines could be used in combination with DNA vaccines, e.g., INFγ, IL-15, and IL-7 [7, 9].

The combination with other types of adjuvants could also be tested, such as TLR-activators. Recently, we demonstrated that the insertion of some CpG immunostimulatory motifs inside the antigen gene sequence through codon optimization could enhance cytokine production, thus increasing the efficacy of a DNA vaccine against P815 mastocytoma [37].

Immune checkpoint blockade (ICB)

The signaling mechanism mediated by costimulatory/inhibitory molecules plays an important role in T cell-mediated immunity. Many cells in the TME can express ligands for inhibitory receptors on T cells, leading to their inactivation [96]. Inhibitory receptors include CTLA-4, PD-1, TIM-3, LAG-3, etc. [96] In several studies, the in vivo blockade of CTLA-4 delayed tumor growth in animal models and resulted in tumor rejection in patients affected by melanoma [97, 98]. This effect was mainly due to the inhibition of TGF-β- and IL-10-secreting Tregs and an increased T effector cell activation [99]. Interestingly, this also resulted in immunity against the secondary exposure to tumor cells, suggesting that the memory component of the immune response can be evoked by anti CTLA-4 antibodies [100]. Antagonist antibodies that target PD-1 and its ligand PD-L1 have also achieved impressive and durable results in many solid tumors, leading to their FDA approval for different cancer types [7]. Recently, a relationship between ICB administration and the neoantigen burden has been demonstrated [101]. Snyder et al. sequenced 64 patients with advanced melanoma and showed that the somatic mutation burden was strongly associated with the clinical response to anti-CTLA4 [102]. Similarly, Rizvi et al. demonstrated that the mutation burden was a strong predictor of clinical response in non-small cell lung cancer (NSCLC) patients treated with anti-PD1 therapy, and that this therapy enhances neoantigen-specific T cell reactivity [103]. The higher prevalence of somatic mutations in cancer cell genomes was a common feature among cancers with a higher probability of responding to ICB. Thereafter, the link between the mutation burden and the clinical benefit following ICB immunotherapy was validated multiple times and in multiple tumor types [63]. This is related to the concept that with an increased tumor mutation burden, the probability of a cognate T cell clonally expanding against a specific tumor antigen will increase. In other words, high tumor mutation burden tumors often have more neoantigens that could be recognized by the processes involved in antitumor immunity, making such cancers more likely to respond to ICB therapy [59, 104].

To increase T cell activity in the TME and to broaden the number of patients responding to ICB, combinations of ICB with different strategies were tested for a variety of malignancies in preclinical and clinical studies [96]. Some examples include combination with radiation therapy [105], other antibodies [106], photodynamic therapy [107], and cancer vaccines [108]. Combination with cancer DNA vaccination seems to be promising in coupling the benefits of ICB with the ability of vaccines to prime the antigen-specific CTL response [88, 109]. A potent cancer vaccine that induces a T cell response against tumor-specific antigens could also increase the number of responders to ICB [63]. However, only a minority of patients respond to ICB therapy, suggesting the need for a rational use of ICB based on biomarkers predictive of the immune response to avoid nonresponsiveness to therapy and undesired side effects [110, 111].

Chemotherapy/targeted therapy

In the last few years, it has been reported that anticancer chemotherapy can play a double role in tumor eradication. Many chemotherapeutic drugs, such as gemcitabine [112], paclitaxel [113], cyclophosphamide [114] and others, applied in ultralow (metronomic) noncytotoxic doses, not only target tumor cells inducing TA release but also enhance T cell infiltration/activity in the TME and remove immunosuppressive cells. In a preclinical study, the combination of cyclophosphamide with DNA vaccines enhanced mouse survival and decreased the expression of immunosuppressive cytokines, such as IL-10 and VEGF [115]. Based on preclinical and clinical studies, the combination of the appropriate chemotherapeutic drug and vaccine therapy may play a substantial role in future cancer treatments, especially when patients do not respond to ICB [116]. Indeed, it has been demonstrated that treatment with chemotherapy restored sensitivity to checkpoint blockade through TLR4 simulation [116]. Further clinical studies are necessary to better define the optimal agents and schedule of administration.

DNA vaccines could also be combined with targeted therapies that are able to mediate tumor cell antigen release and enhance T cell priming. Sunitinib, a multitargeted receptor tyrosine kinase inhibitor, was found to decrease Tregs and MDSCs and increase INFγ-producing T cells in renal cell carcinoma patients [117]. The combination of sunitinib with a viral vaccine encoding CEA decreased the tumor volume in a mouse model [118]. Although not already tested with DNA vaccines, other tyrosine kinase inhibitors already approved by FDA, such as pazopanib, axitinib, and cabozantinib, could improve the patient response to vaccination.

Combination with other therapies

Other strategies that can be used in combination with DNA vaccines include endocrine therapy and radiotherapy (RT).

In hormonally driven tumors such as prostate cancer and breast cancer, endocrine therapy is part of the standard of care, and the effect of letrozole in decreasing the Tregs in the TME has already been demonstrated [119]. Furthermore, androgen deprivation in prostate cancer induces thymic regeneration and increases the number of effector T cells [7]. In an ongoing clinical trial (NCT02204098, phase I), the effect of Mam-A vaccine administration in combination with anastrozole, letrozole, tamoxifen, exemestane, and goserelin is under investigation.

Preclinical data have demonstrated the additive effect of RT and vaccines with an enhanced destruction of tumor cells, the release of TAs, an increase in IFNg production, and a global decrease of the tumor volume. T cells specific for other antigens not included in the vaccine were also generated [120, 121]. Since RT is a part of the standard of care, many trials using vaccines after radiation try to evaluate the mutual effects from the 2 therapies (Table 2).

In Fig. 3, the main mechanisms of action of therapies postulated to mediate synergistic effects in combination with DNA vaccines are shown.

Fig. 3.

Fig. 3

Mechanisms of action of therapies postulated to mediate synergistic effects in combination with DNA

Results of completed clinical trials

Many already completed clinical trials tested the efficacy of DNA vaccines against different tumor types, such as breast, cervical, pancreatic and prostate cancers, multiple myeloma, and melanoma. These trials aimed at principally evaluating the safety and immunological response of DNA vaccines. A search for studies with “cancer” and “DNA vaccines” in clinicaltrials.gov [122] revealed 48 studies in the last 10 years with the following criteria: “completed”, “suspended” and “terminated”. Among the trials using DNA vaccines in a therapeutic approach, only a few of them have published results to date. Here, a non-exhaustive list of completed studies using naked DNA vaccines and containing results is described.

The NCT01304524 phase IIb clinical study tested the safety and efficacy of VGX-3100, a DNA vaccine targeting HPV 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia grade 2/3. Six milligrams of the vaccine were delivered by IM EP at 0, 4 and 12 weeks without any severe side effects, but only mild reactions in the injection site, fatigue, nausea and general malaise in some of the patients. The vaccine was generally well tolerated and showed great efficacy against the pathology in almost 50% of the treated patients, as shown in the histopathological and immunological analysis. Indeed, VGX-3100 elicited significantly increased frequencies of antigen-specific activated CD8+ T cells and a higher humoral response compared to the placebo, making it the first therapeutic vaccine to elicit a complete adaptive immune response in patients with preinvasive cervical disease caused by HPV-16 and 18 [123]. Two phase III clinical trials (NCT03185013 and NCT03721978) using VGX-3100 are ongoing, as shown in Table 2.

Recently, Kim et al. published the results of the clinical trial NCT01634503 concerning the safety and efficacy of GX-188E, another plasmid DNA encoding the E6 and E7 proteins of HPV serotypes 16 and 18. The vaccine was injected 3 times (weeks 0, 4 and 12) IM to alternating deltoid muscles, and three different doses were tested (1, 2 or 4 mg). Importantly, 8/9 of the patients exhibited an enhanced polyfunctional HPV-specific CD8 T cell response, and 7/9 of the patients displayed a complete regression of their lesions and viral clearance within 36 weeks of follow-up. The vaccine administration did not elicit serious vaccine-associated adverse events and was estimated to be safe and well tolerated [124].

Other published results show the properties of mammaglobin-A (Mam-A) DNA vaccination for patients with breast cancer. Mam-A is a tumor-specific secretory protein overexpressed in 80% of human breast cancers. In a phase I clinical trial (NCT00807781), 4 mg of a pING-Mam-A DNA vaccine was administered at weeks 1, 4 and 8 IM to patients with metastatic breast cancer. The first results demonstrated the safety of the vaccine, with no significant side effects. The main observations about its efficacy were (i) an increase in the generation of specific Mam-A CD8+ T cells and IFN-γ production; (ii) a decrease in the frequency of Tregs and lower levels of IL-10; and (iii) an improved progression-free survival compared to the control group. These encouraging results suggest that Mam-A DNA vaccination can induce antitumor immunity in breast cancer patients and increase survival time [125127].

In another phase I/II study (NCT00859729), 50–1600 μg of a pVAXrcPSAv531 plasmid coding for the full-length PSA protein were ID injected and electroporated in patients with relapsed prostate cancer. The vaccine followed radiotherapy and endocrine therapy with an LH-RH analogue (leuprorelin). No systemic toxicity was observed, and discomfort from EP did not require the use of topical anesthetics. A general increase in T cell reactivity was observed in most patients, although IM immunization seemed to result in more potent antibody responses [128].

A personalized DNA vaccine was tested in patients with multiple myeloma in a phase I clinical trial. The DNA encoded a patient-specific single chain variable fragment linked to fragment C of the tetanus toxin. Six doses of 1 mg of the vaccine were injected IM after chemotherapy or autologous stem cell transplant. In total, 72% of the patients generated a cell-specific immune response, and the overall survival was 64% after a median follow-up of 85.6 months [129].

A phase II clinical trial (NCT01334060) evaluated the safety and efficacy of a pDOM-WT1–37 and pDOM-WT1–126 DNA fusion gene vaccine encoding the Wilms tumor antigen 1 for leukemia patients. The plasmid was injected using IM EP, with no severe side effects. However, combination strategies to expand T cell responses with immunomodulatory antibodies are in development [130].

Interestingly, Niethammer et al. reported a phase I clinical trial (NCT01486329) using an oral vaccine (VXM01) against the VEGF-Receptor 2 with Salmonella typhimurium as a carrier, in addition to chemotherapy with gemcitabine, in patients with stage IV and locally advanced pancreatic cancer. The doses consisted of a solution containing 106, 108, 109, and 1010 colony forming units of VXM01. VXM01 represents a novel strategy by not targeting a tumor cell-resident antigen but instead targeting a tumor stroma-resident antigen overexpressed by the nonmalignant endothelial cells of the tumor neovasculature, giving the vaccine the potential to target many cancer types [131]. The same vaccine is also being tested in patients with glioblastoma (NCT02718443).

Another 19 studies were found in PubMed using the following criteria: “cancer DNA vaccine”, article type “clinical trial”, starting from 2013 until now. Most of the studies focus on prophylactic immunization with HPV DNA vaccines. Two phase I studies show some results of therapeutic cancer DNA vaccination (NCT00250419 and NCT00647114). Both of them used the HER2/CEA DNA vaccine V930 and showed the instauration of both humoral and cellular immune responses with no detectable immune response against the vaccine itself. As CEA and HER2 are expressed by many solid tumors, patients with different types of cancer were recruited. The vaccination dose was on the order of a few milligrams every 14 days for 5 injections, and the plasmid was injected by IM EP. However, in this case, no evidence of an increase of a HER/2 or CEA-specific response was observed [132].

Overall, vaccination is used after conventional therapies. Completed, terminated and suspended clinical trials reported only minor discomfort after vaccination, no important side effects and, generally, an increased number of CD8+ T cells specific for the antigen encoded by the DNA vaccine. Most of the trials used DNA vaccines encoding TAAs, and only a few tested personalized approaches.

Ongoing human clinical trials using therapeutic cancer DNA vaccinations

In searching all the cancer DNA vaccine interventional clinical studies in the last 10 years with the criteria “not yet recruiting”, “recruiting”, “enrolling by invitation” and “active nonrecruiting”, we found 56 studies. Among them, 27 studies used DNA vaccines as naked plasmids not encapsulated in cells or in virus-like nanoparticles. These studies are listed in Table 2. They are all in clinical phase I or I/II or II, and DNA vaccines are generally administered after the standard of care for each cancer type, including surgical ablation, radiotherapy and/or chemotherapy. The results for these trials are not yet available, except for the trial NCT00849121. This study used a DNA vaccine encoding PAP, with GM-CSF as an adjuvant, administered ID into patients with prostate cancer. Only one of the 17 patients experienced a vaccine-related adverse event of grade 2 or more, more than half had a great PAP-specific CTL response, and in 7/17 patients, the PSA doubling time increased during the treatment period. Twelve of the 17 patients (70%) were metastasis-free after one year of treatment (clinicaltrials.gov).

Another study with the criteria “DNA electroporation” and “cancer” led to 3 more trials (“not yet recruiting”, “recruiting”, “enrolling by invitation” and “active nonrecruiting”) in the last 10 years: NCT03499795, NCT03491683, and NCT02301754. With the criteria “plasmid” and “tumor”, we found 2 additional studies: NCT02531425 and NCT03502785. These are all listed in Table 2.

Of particular interest are the only 2 studies we found in phase III (NCT03721978 and NCT03185013) using VGX-3100 delivered by IM EP against cervical cancer.

Breast, prostate and cervical cancer are the most studied in the trials (Fig. 4a). Most of the vaccines encode well-known TAAs (E6/7 HPV protein for cervical cancer, Mam-A or HER2 for breast cancer, prostatic acid phosphatase (PAP) for prostate cancer, etc.). Only 17% of the clinical trials used personalized/neoantigen vaccines (e.g., NCT02348320 and NCT03122106), as shown in Fig. 4b. This number has increased in recent years: 80% of the trials using neoantigens started in 2018–2019. Generally, more than one epitope is encoded by the DNA vaccines in both TAA and neoantigen vaccines (Fig. 4b).

Fig. 4.

Fig. 4

Ongoing clinical trials of the analyzed studies. a Cancer types using cancer DNA vaccines in clinical trials. b Type of antigens encoded in the DNA vaccine. c Studies combining cancer DNA vaccines with other therapies (endocrine therapy, immunotherapy, chemotherapy, chemoradiotherapy or adjuvants) or using DNA vaccines as a single therapy

DNA vaccines are mostly associated with other therapies: immunotherapies (antibodies anti-HER2, anti-CTLA4, anti-PD1, anti-PD-L1, and cell vaccines), immune adjuvants (GM-CSF, hIL-12, etc.) generally injected with the DNA vaccine or encoded in the vaccine itself, chemotherapy (carboplatin, paclitaxel, cyclophosphamide), and endocrine therapies (anastrozole, letrozole, tamoxifen, exemestane, and goserelin). In recent years, the number of studies using other therapies in combination with DNA vaccines has also increased (Fig. 4c). DNA vaccines are usually injected IM or ID, in rare cases SC or in the lesion/tumor, and electroporated after the injection. The doses can vary from 100 μg to a few mg. The regimen of administration depends on the type of vaccine, but in all trials, vaccines are injected more than once, at 2–4 weeks of intervals, and the therapy lasts for a few months.

Current challenges and future perspectives

Past and ongoing clinical studies investigate DNA vaccines that are optimized using various strategies. The use of a codon-optimized, polyepitopic DNA vaccine encoding TAAs or neoantigens and their combination with other therapies to modulate the immunosuppressive TME seem to be the most relevant options. However, many questions still need to be addressed.

Selection of the encoded antigen(s)

The first question concerns the antigen type: TAAs or neoantigens? Relevant TAAs have been identified for most tumors, but immune tolerance can limit their efficiency. On the other hand, neoantigen identification is time consuming and expensive, and neoantigens do not reflect the tumor heterogeneity in the individual patient (e.g., in metastasis). Second, how many antigens should be encoded in the same plasmid? It is still not clear whether one antigen is superior to another in terms of frequency of immune response or clinical effect [133]. In one study, it has been shown that neoantigens with a predicted high affinity are more immunogenic and that a poly-specific and poly-functional DNA vaccine encoding neoantigens was the most efficient solution to prevent tumor growth in mice [134]. Further studies are needed to generalize these findings. Most of the clinical trials use a relevant antigen for the vaccination (e.g., PAP, E6/E7), but none compare the combination of this antigen with others or with neoantigens to evaluate their effect on immune activation. This point arouses a last question: is it worth mixing TAAs and neoantigens in the same vaccine? Furthermore, should the different antigens be administered in the same plasmid or is it better to encode them in different plasmids administered together? To our knowledge, none of the clinical studies address this question, which could be a further area of investigation in the future.

Selection of the combination therapy and treatment schedule

An important parameter in the combinatorial approach is the choice of the right therapies to be combined. This aspect depends on the tumor type, its presence, and the possibility of detecting patient-specific biomarkers, among others. In addition, the administration schedule of multiple therapies is crucial. Until now, vaccines have been used as a last-line therapy. To integrate them as a standard of care therapy, it is crucial to know when to administer them. This decision should consider the time for the immune system to generate a specific immune response against the delivered antigen, the need for multiple doses of administration and the interaction with the combined therapy. For example, some studies revealed that using ICB after the peptide/RNA vaccine treatment induced a sustained remission with no sign of disease recurrence [66]. However, this should be adapted to the specific therapy and patient. Furthermore, the doses should be consequently adapted to the combination regimen.

Find a good preclinical model

Most of the critical points aroused in cancer DNA vaccination can ultimately be addressed only in clinical practice because translation from animals is extremely difficult. This is due to the different tumor characteristics and the differences in the immune system between humans and animals [135]. To try to overcome this problem, many orthotopic injection models, genetically engineered mice, xenograph and humanized models have been developed. However, they all fail to recapitulate the chaotic way in which malignant transformation occurs during cancer development in human patients. Mouse models provide valuable insight into the mechanisms of action and provide important proof of concept for human studies, but there remains a need for larger animal models encompassing a fully competent immune system. Some researchers suggest the use of canine and porcine models, especially for skin cancers [135]. However, housing, ethical regulation, and breeding difficulties limit the use of big animal models. Furthermore, even these models have limitations, and the idea of a universal model for oncoimmunology currently seems unrealistic.

The variability problem: the necessity of biomarkers and therapy standardization; how far from therapy personalization?

Immunotherapies have a variable response rate from one patient to another and are often associated with side effects. For this reason, it is important to identify biomarkers that could predict the patient response to a specific therapy and to standardize the therapy according to the predicted biomarker. Furthermore, biomarkers may be useful for monitoring treatment response. However, the translation of biological data into predictive or prognostic biomarkers is complicated by the complex interactions between tumors and the immune system and by host and tumor variability. Many studies are using bioinformatics tools and new genomic and proteomic technologies to predict specific tumor signatures, generating complex datasets that give rise to analytical challenges. Currently, we can rely on imperfect biomarkers, such as PD-L1 expression in the tumor or the tumor mutation burden. This gap in knowledge leaves room for further studies that will help treatment selection and design the best combination therapy for each patient.

Conclusions

The analysis of recent preclinical and clinical trials suggests that the current therapeutic cancer vaccines are unlikely to dramatically impact cancer outcomes as a single agent. Many combinations with other strategies have been tested, demonstrating the greater potential of the combination on improving clinical outcomes compared to the single therapy. The personalized approaches both in the vaccine design and in the choice of combination therapy will be crucial for success in the clinic. Furthermore, since DNA vaccines are well tolerated and safe, their combination with other therapies could become part of the standard of care in many malignancies.

We anticipate that, in the future, personalization in the DNA vaccine design will be coupled with personalization in the choice of the most appropriate combined therapy, following the analysis of single patient specificity and biomarkers that can predict the response to a specific agent. This could represent the best approach to increase the efficacy of cancer immunotherapy and reduce the adverse effects linked to a nonspecific treatment.

Acknowledgements

Not applicable.

Funding

Fondation contre le cancer, Belgium.

Availability of data and materials

Not applicable.

Abbreviations

CAR

Chimeric antigen receptor

CTL

Cytotoxic T cell

ICB

Immune checkpoint blockade

ID

Intradermal

IM

Intramuscular

MDSC

Myeloid-derived T cell

MHC

Major histocompatibility complex

PADRE

Pan DR epitope

RT

Radiotherapy

SC

Subcutaneous

TA

Tumor antigen

TAA

Tumor-associated antigen

TAM

Tumor-associated macrophages

Th

T helper

TME

Tumor microenvironment

Treg

Regulatory T cell

TSA

Tumor-specific antigen

Authors’ contributions

All authors planned, read and approved the final manuscript. All wrote the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Contributor Information

Alessandra Lopes, Email: Alessandra.lopes@uclouvain.be.

Gaëlle Vandermeulen, Email: Gaelle.vandermeulen@uclouvain.be.

Véronique Préat, Phone: +3227647309, Email: veronique.preat@uclouvain.be.

References

  • 1.Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, Chlichlia K. DNA vaccines to attack cancer: strategies for improving immunogenicity and efficacy. Pharmacol Ther. 2016;165:32–49. doi: 10.1016/j.pharmthera.2016.05.004. [DOI] [PubMed] [Google Scholar]
  • 2.Gasser M, Waaga-Gasser AM. Therapeutic antibodies in Cancer therapy. In: Böldicke T, editor. Protein targeting compounds: prediction, selection and activity of specific inhibitors. Cham: Springer International Publishing; 2016. pp. 95–120. [Google Scholar]
  • 3.Clarke JM, George DJ, Lisi S, Salama AKS. Immune checkpoint blockade: the new frontier in Cancer treatment. Target Oncol. 2018;13(1):1–20. doi: 10.1007/s11523-017-0549-7. [DOI] [PubMed] [Google Scholar]
  • 4.Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39. doi: 10.1016/j.intimp.2018.06.001. [DOI] [PubMed] [Google Scholar]
  • 5.Fry TJ, Mackall CL. T-cell adoptive immunotherapy for acute lymphoblastic leukemia. Hematology American Society of Hematology Education Program. 2013;2013:348–353. doi: 10.1182/asheducation-2013.1.348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst. 2012;104(8):599–613. doi: 10.1093/jnci/djs033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gatti-Mays ME, Redman JM, Collins JM, Bilusic M. Cancer vaccines: enhanced immunogenic modulation through therapeutic combinations. Human vaccines & immunotherapeutics. 2017;13(11):2561–2574. doi: 10.1080/21645515.2017.1364322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cebon J. Perspective: cancer vaccines in the era of immune checkpoint blockade. Mammalian genome : official journal of the International Mammalian Genome Society. 2018;29(11–12):703–713. doi: 10.1007/s00335-018-9786-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Amara S, Tiriveedhi V. The five immune forces impacting DNA-Based cancer immunotherapeutic strategy. Int J Mol Sci. 2017;18:650. 10.3390/ijms18030650. [DOI] [PMC free article] [PubMed]
  • 10.Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14:135. doi: 10.1038/nrc3670. [DOI] [PubMed] [Google Scholar]
  • 11.Boon T, van der Bruggen P. Human tumor antigens recognized by T lymphocytes. J Exp Med. 1996;183(3):725–729. doi: 10.1084/jem.183.3.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Finn OJ. Human tumor antigens yesterday, today, and tomorrow. Cancer immunology research. 2017;5(5):347–354. doi: 10.1158/2326-6066.CIR-17-0112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Strioga MM, Darinskas A, Pasukoniene V, Mlynska A, Ostapenko V, Schijns V. Xenogeneic therapeutic cancer vaccines as breakers of immune tolerance for clinical application: to use or not to use? Vaccine. 2014;32(32):4015–4024. doi: 10.1016/j.vaccine.2014.05.006. [DOI] [PubMed] [Google Scholar]
  • 14.Li L, Goedegebuure SP, Gillanders WE. Preclinical and clinical development of neoantigen vaccines. Ann Oncol. 2017;28(suppl_12):xii11–xxii7. doi: 10.1093/annonc/mdx681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Alibek K, Baiken Y, Kakpenova A, Mussabekova A, Zhussupbekova S, Akan M, et al. Implication of human herpesviruses in oncogenesis through immune evasion and supression. Infectious agents and cancer. 2014;9(1):3. doi: 10.1186/1750-9378-9-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Martin-Liberal J, Ochoa de Olza M, Hierro C, Gros A, Rodon J, Tabernero J. The expanding role of immunotherapy. Cancer Treat Rev. 2017;54:74–86. doi: 10.1016/j.ctrv.2017.01.008. [DOI] [PubMed] [Google Scholar]
  • 17.Shang N, Figini M, Shangguan J, Wang B, Sun C, Pan L, et al. Dendritic cells based immunotherapy. Am J Cancer Res. 2017;7(10):2091–2102. [PMC free article] [PubMed] [Google Scholar]
  • 18.Obara W, Kanehira M, Katagiri T, Kato R, Kato Y, Takata R. Present status and future perspective of peptide-based vaccine therapy for urological cancer. Cancer Sci. 2018;109(3):550–559. doi: 10.1111/cas.13506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chauchet X, Wang Y, Polack B, Gouëllec AL. Live-attenuated bacteria as a cancer vaccine vector AU - Toussaint, Bertrand. Expert review of vaccines. 2013;12(10):1139–1154. doi: 10.1586/14760584.2013.836914. [DOI] [PubMed] [Google Scholar]
  • 20.Russell SJ, Barber GN. Oncolytic viruses as antigen-agnostic Cancer vaccines. Cancer Cell. 2018;33(4):599–605. doi: 10.1016/j.ccell.2018.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.McNamara MA, Nair SK, Holl EK. RNA-based vaccines in Cancer immunotherapy. J Immunol Res. 2015;2015:794528. doi: 10.1155/2015/794528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Herrada AA, Rojas-Colonelli N, Gonzalez-Figueroa P, Roco J, Oyarce C, Ligtenberg MA, et al. Harnessing DNA-induced immune responses for improving cancer vaccines. Human vaccines & immunotherapeutics. 2012;8(11):1682–1693. doi: 10.4161/hv.22345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yankauckas MA, Morrow JE, Parker SE, Abai A, Rhodes GH, Dwarki VJ, et al. Long-term anti-nucleoprotein cellular and humoral immunity is induced by intramuscular injection of plasmid DNA containing NP gene. DNA Cell Biol. 1993;12(9):771–776. doi: 10.1089/dna.1993.12.771. [DOI] [PubMed] [Google Scholar]
  • 24.Lambricht L, Lopes A, Kos S, Sersa G, Preat V, Vandermeulen G. Clinical potential of electroporation for gene therapy and DNA vaccine delivery. Expert opinion on drug delivery. 2016;13(2):295–310. doi: 10.1517/17425247.2016.1121990. [DOI] [PubMed] [Google Scholar]
  • 25.Liu Y, Yan J, Santangelo PJ, Prausnitz MR. DNA uptake, intracellular trafficking and gene transfection after ultrasound exposure. Journal of controlled release : official journal of the Controlled Release Society. 2016;234:1–9. doi: 10.1016/j.jconrel.2016.05.013. [DOI] [PubMed] [Google Scholar]
  • 26.Miller DL, Pislaru SV, Greenleaf JE. Sonoporation: mechanical DNA delivery by ultrasonic cavitation. Somat Cell Mol Genet. 2002;27(1–6):115–134. doi: 10.1023/A:1022983907223. [DOI] [PubMed] [Google Scholar]
  • 27.van den Berg JH, Oosterhuis K, Schumacher TN, Haanen JB, Bins AD. Intradermal vaccination by DNA tattooing. Methods Mol Biol. 2014;1143:131–140. doi: 10.1007/978-1-4939-0410-5_9. [DOI] [PubMed] [Google Scholar]
  • 28.Bergmann-Leitner ES, Leitner WW. Vaccination using Gene-gun Technology. In: Vaughan A, editor. Malaria vaccines: methods and protocols. New York: Springer New York; 2015. pp. 289–302. [DOI] [PubMed] [Google Scholar]
  • 29.Li L, Petrovsky N. Molecular mechanisms for enhanced DNA vaccine immunogenicity. Expert review of vaccines. 2016;15(3):313–329. doi: 10.1586/14760584.2016.1124762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ori D, Murase M, Kawai T. Cytosolic nucleic acid sensors and innate immune regulation. Int Rev Immunol. 2017;36(2):74–88. doi: 10.1080/08830185.2017.1298749. [DOI] [PubMed] [Google Scholar]
  • 31.Tang CK, Pietersz GA. Intracellular detection and immune signaling pathways of DNA vaccines. Expert review of vaccines. 2009;8(9):1161–1170. doi: 10.1586/erv.09.79. [DOI] [PubMed] [Google Scholar]
  • 32.Boyer J, Ugen K, Wang B, Chattergoon M, Tsai A, Merva M, et al. Induction of a TH1 type cellular immune response to the human immunodeficiency type 1 virus by in vivo DNA inoculation. Dev Biol Stand. 1998;92:169–174. [PubMed] [Google Scholar]
  • 33.Schirmbeck R, Bohm W, Ando K, Chisari FV, Reimann J. Nucleic acid vaccination primes hepatitis B virus surface antigen-specific cytotoxic T lymphocytes in nonresponder mice. J Virol. 1995;69(10):5929–5934. doi: 10.1128/jvi.69.10.5929-5934.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Baghban Rahimi S, Mohebbi A, Vakilzadeh G, Biglari P, Razeghi Jahromi S, Mohebi SR, et al. Enhancement of therapeutic DNA vaccine potency by melatonin through inhibiting VEGF expression and induction of antitumor immunity mediated by CD8+ T cells. Arch Virol. 2018;163(3):587–597. doi: 10.1007/s00705-017-3647-z. [DOI] [PubMed] [Google Scholar]
  • 35.Ahrends T, Bąbała N, Xiao Y, Yagita H, van Eenennaam H, Borst J. CD27 Agonism Plus PD-1 Blockade Recapitulates CD4<sup>+</sup> T-cell Help in Therapeutic Anticancer Vaccination. Cancer Res. 2016;76(10):2921. doi: 10.1158/0008-5472.CAN-15-3130. [DOI] [PubMed] [Google Scholar]
  • 36.Denies S, Cicchelero L, Polis I, Sanders NN. Immunogenicity and safety of xenogeneic vascular endothelial growth factor receptor-2 DNA vaccination in mice and dogs. Oncotarget. 2016;7(10):10905–10916. doi: 10.18632/oncotarget.7265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lopes A, Vanvarenberg K, Preat V, Vandermeulen G. Codon-optimized P1A-encoding DNA vaccine: toward a therapeutic vaccination against P815 Mastocytoma. Molecular therapy Nucleic acids. 2017;8:404–415. doi: 10.1016/j.omtn.2017.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity. Human vaccines & immunotherapeutics. 2017;13(12):2837–2848. doi: 10.1080/21645515.2017.1330236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jorritsma SHT, Gowans EJ, Grubor-Bauk B, Wijesundara DK. Delivery methods to increase cellular uptake and immunogenicity of DNA vaccines. Vaccine. 2016;34(46):5488–5494. doi: 10.1016/j.vaccine.2016.09.062. [DOI] [PubMed] [Google Scholar]
  • 40.Lambricht L, Vanvarenberg K, De Beuckelaer A, Van Hoecke L, Grooten J, Ucakar B, et al. Coadministration of a plasmid encoding HIV-1 gag enhances the efficacy of Cancer DNA vaccines. Molecular therapy : the journal of the American Society of Gene Therapy. 2016;24(9):1686–1696. doi: 10.1038/mt.2016.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Aldous AR, Dong JZ. Personalized neoantigen vaccines: a new approach to cancer immunotherapy. Bioorg Med Chem. 2018;26(10):2842–2849. doi: 10.1016/j.bmc.2017.10.021. [DOI] [PubMed] [Google Scholar]
  • 42.Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Riccardo F, Bolli E, Macagno M, Arigoni M, Cavallo F, Quaglino E. Chimeric DNA vaccines: an effective way to overcome immune tolerance. Curr Top Microbiol Immunol. 2017;405:99–122. doi: 10.1007/82_2014_426. [DOI] [PubMed] [Google Scholar]
  • 44.Soong RS, Trieu J, Lee SY, He L, Tsai YC, Wu TC, et al. Xenogeneic human p53 DNA vaccination by electroporation breaks immune tolerance to control murine tumors expressing mouse p53. PLoS One. 2013;8(2):e56912. doi: 10.1371/journal.pone.0056912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sioud M, Sørensen D. Generation of an effective anti-tumor immunity after immunization with xenogeneic antigens. Eur J Immunol. 2003;33(1):38–45. doi: 10.1002/immu.200390005. [DOI] [PubMed] [Google Scholar]
  • 46.Wei Y, Sun Y, Song C, Li H, Li Y, Zhang K, et al. Enhancement of DNA vaccine efficacy by targeting the xenogeneic human chorionic gonadotropin, survivin and vascular endothelial growth factor receptor 2 combined tumor antigen to the major histocompatibility complex class II pathway. The journal of gene medicine. 2012;14(5):353–362. doi: 10.1002/jgm.2624. [DOI] [PubMed] [Google Scholar]
  • 47.Grosenbaugh DA, Leard AT, Bergman PJ, Klein MK, Meleo K, Susaneck S, et al. Safety and efficacy of a xenogeneic DNA vaccine encoding for human tyrosinase as adjunctive treatment for oral malignant melanoma in dogs following surgical excision of the primary tumor. Am J Vet Res. 2011;72(12):1631–1638. doi: 10.2460/ajvr.72.12.1631. [DOI] [PubMed] [Google Scholar]
  • 48.Quaglino E, Riccardo F, Macagno M, Bandini S, Cojoca R, Ercole E, et al. Chimeric DNA vaccines against ErbB2+ carcinomas: from mice to humans. Cancers. 2011;3(3):3225–3241. doi: 10.3390/cancers3033225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Almajhdi FN, Senger T, Amer HM, Gissmann L, Ohlschlager P. Design of a highly effective therapeutic HPV16 E6/E7-specific DNA vaccine: optimization by different ways of sequence rearrangements (shuffling) PLoS One. 2014;9(11):e113461. doi: 10.1371/journal.pone.0113461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Aurisicchio L, Roscilli G, Marra E, Luberto L, Mancini R, La Monica N, et al. Superior immunologic and therapeutic efficacy of a xenogeneic genetic Cancer vaccine targeting carcinoembryonic human antigen. Hum Gene Ther. 2015;26(6):386–398. doi: 10.1089/hum.2014.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Occhipinti S, Sponton L, Rolla S, Caorsi C, Novarino A, Donadio M, et al. Chimeric rat/human HER2 efficiently circumvents HER2 tolerance in cancer patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014;20(11):2910–2921. doi: 10.1158/1078-0432.CCR-13-2663. [DOI] [PubMed] [Google Scholar]
  • 52.Ruffini PA, Os A, Dolcetti R, Tjonnfjord GE, Munthe LA, Bogen B. Targeted DNA vaccines eliciting crossreactive anti-idiotypic antibody responses against human B cell malignancies in mice. J Transl Med. 2014;12:207. doi: 10.1186/1479-5876-12-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.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. [PMC free article] [PubMed] [Google Scholar]
  • 54.Yuan J, Ku GY, Adamow M, Mu Z, Tandon S, Hannaman D, et al. Immunologic responses to xenogeneic tyrosinase DNA vaccine administered by electroporation in patients with malignant melanoma. Journal for immunotherapy of cancer. 2013;1:20. doi: 10.1186/2051-1426-1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Brennick CA, George MM, Corwin WL, Srivastava PK, Ebrahimi-Nik H. Neoepitopes as cancer immunotherapy targets: key challenges and opportunities. Immunotherapy. 2017;9(4):361–371. doi: 10.2217/imt-2016-0146. [DOI] [PubMed] [Google Scholar]
  • 56.Aurisicchio L, Pallocca M, Ciliberto G, Palombo F. The perfect personalized cancer therapy: cancer vaccines against neoantigens. Journal of experimental & clinical cancer research : CR. 2018;37(1):86. doi: 10.1186/s13046-018-0751-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565(7738):234–239. doi: 10.1038/s41586-018-0792-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sahin U, Tureci O. Personalized vaccines for cancer immunotherapy. Science. 2018;359(6382):1355–1360. doi: 10.1126/science.aar7112. [DOI] [PubMed] [Google Scholar]
  • 59.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. doi: 10.1126/science.aaa4971. [DOI] [PubMed] [Google Scholar]
  • 60.Duperret EK, Perales-Puchalt A, Stoltz R, Hiranjith GH, Mandloi N, Barlow J, et al. A Synthetic DNA, Multi-Neoantigen Vaccine Drives Predominately MHC Class I CD8<sup>+</sup> T-cell Responses, Impacting Tumor Challenge. Cancer Immunology Research. 2019;7(2):174. doi: 10.1158/2326-6066.CIR-18-0283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vasquez M, Tenesaca S, Berraondo. New trends in antitumor vaccines in melanoma. Ann Trans Med. 2017;5(19):384. 10.21037/atm.2017.09.09. [DOI] [PMC free article] [PubMed]
  • 62.Kreiter S, Vormehr M, van de Roemer N, Diken M, Lower M, Diekmann J, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015;520(7549):692–696. doi: 10.1038/nature14426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lee CH, Yelensky R, Jooss K, Chan TA. Update on tumor Neoantigens and their utility: why it is good to be different. Trends Immunol. 2018;39(7):536–548. doi: 10.1016/j.it.2018.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hundal J, Carreno BM, Petti AA, Linette GP, Griffith OL, Mardis ER, et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome medicine. 2016;8(1):11. doi: 10.1186/s13073-016-0264-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hellmann MD, Snyder A. Making it personal: Neoantigen vaccines in metastatic melanoma. Immunity. 2017;47(2):221–223. doi: 10.1016/j.immuni.2017.08.001. [DOI] [PubMed] [Google Scholar]
  • 66.Sahin U, Derhovanessian E, Miller M, Kloke B-P, Simon P, Löwer M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222. doi: 10.1038/nature23003. [DOI] [PubMed] [Google Scholar]
  • 67.Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer. 2012;12:323. doi: 10.1038/nrc3261. [DOI] [PubMed] [Google Scholar]
  • 68.Duperret EK, Perales-Puchalt A, Stoltz R. G HH, Mandloi N, Barlow J, et al. a synthetic DNA, multi-Neoantigen vaccine drives predominately MHC class I CD8(+) T-cell responses, impacting tumor challenge. Cancer immunology research. 2019;7(2):174–182. doi: 10.1158/2326-6066.CIR-18-0283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bae J, Prabhala R, Voskertchian A, Brown A, Maguire C, Richardson P, et al. A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia. 2015;29(1):218–229. doi: 10.1038/leu.2014.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bei R, Scardino A. TAA polyepitope DNA-based vaccines: a potential tool for cancer therapy. J Biomed Biotechnol. 2010;2010:102758. doi: 10.1155/2010/102758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tan AC, La Gruta NL, Zeng W, Jackson DC. Precursor frequency and competition dictate the HLA-A2-restricted CD8+ T cell responses to influenza a infection and vaccination in HLA-A2.1 transgenic mice. J Immunol. 2011;187(4):1895–1902. doi: 10.4049/jimmunol.1100664. [DOI] [PubMed] [Google Scholar]
  • 72.Palmowski M, Salio M, Dunbar RP, Cerundolo V. The use of HLA class I tetramers to design a vaccination strategy for melanoma patients. Immunol Rev. 2002;188:155–163. doi: 10.1034/j.1600-065X.2002.18814.x. [DOI] [PubMed] [Google Scholar]
  • 73.Durantez M, Lopez-Vazquez AB, de Cerio AL, Huarte E, Casares N, Prieto J, et al. Induction of multiepitopic and long-lasting immune responses against tumour antigens by immunization with peptides, DNA and recombinant adenoviruses expressing minigenes. Scand J Immunol. 2009;69(2):80–89. doi: 10.1111/j.1365-3083.2008.02202.x. [DOI] [PubMed] [Google Scholar]
  • 74.Galaine J, Borg C, Godet Y, Adotevi O. Interest of tumor-specific CD4 T helper 1 cells for therapeutic anticancer vaccine. Vaccines. 2015;3(3):490–502. doi: 10.3390/vaccines3030490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Protti MP, Monte LD, Lullo GD. Tumor antigen-specific CD4+ T cells in cancer immunity: from antigen identification to tumor prognosis and development of therapeutic strategies. Tissue Antigens. 2014;83(4):237–246. doi: 10.1111/tan.12329. [DOI] [PubMed] [Google Scholar]
  • 76.Efremova M, Finotello F, Rieder D, Trajanoski Z. Neoantigens generated by individual mutations and their role in Cancer immunity and immunotherapy. Front Immunol. 2017;8:1679. doi: 10.3389/fimmu.2017.01679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Doan T, Herd K, Ramshaw I, Thomson S, Tindle RW. A polytope DNA vaccine elicits multiple effector and memory CTL responses and protects against human papillomavirus 16 E7-expressing tumour. Cancer immunology, immunotherapy : CII. 2005;54(2):157–171. doi: 10.1007/s00262-004-0544-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lund LH, Andersson K, Zuber B, Karlsson A, Engstrom G, Hinkula J, et al. Signal sequence deletion and fusion to tetanus toxoid epitope augment antitumor immune responses to a human carcinoembryonic antigen (CEA) plasmid DNA vaccine in a murine test system. Cancer Gene Ther. 2003;10(5):365–376. doi: 10.1038/sj.cgt.7700574. [DOI] [PubMed] [Google Scholar]
  • 79.Lu Y, Ouyang K, Fang J, Zhang H, Wu G, Ma Y, et al. Improved efficacy of DNA vaccination against prostate carcinoma by boosting with recombinant protein vaccine and by introduction of a novel adjuvant epitope. Vaccine. 2009;27(39):5411–5418. doi: 10.1016/j.vaccine.2009.06.089. [DOI] [PubMed] [Google Scholar]
  • 80.Scardino A, Alimandi M, Correale P, Smith SG, Bei R, Firat H, et al. A polyepitope DNA vaccine targeted to her-2/ErbB-2 elicits a broad range of human and murine CTL effectors to protect against tumor challenge. Cancer Res. 2007;67(14):7028–7036. doi: 10.1158/0008-5472.CAN-06-3998. [DOI] [PubMed] [Google Scholar]
  • 81.Wu A, Zeng Q, Kang TH, Peng S, Roosinovich E, Pai SI, et al. Innovative DNA vaccine for human papillomavirus (HPV)-associated head and neck cancer. Gene Ther. 2011;18(3):304–312. doi: 10.1038/gt.2010.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cho HI, Celis E. Design of immunogenic and effective multi-epitope DNA vaccines for melanoma. Cancer immunology, immunotherapy : CII. 2012;61(3):343–351. doi: 10.1007/s00262-011-1110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hung CF, Tsai YC, He L, Wu TC. DNA vaccines encoding ii-PADRE generates potent PADRE-specific CD4+ T-cell immune responses and enhances vaccine potency. Molecular therapy : the journal of the American Society of Gene Therapy. 2007;15(6):1211–1219. doi: 10.1038/sj.mt.6300121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Park JY, Jin DH, Lee CM, Jang MJ, Lee SY, Shin HS, et al. CD4+ TH1 cells generated by ii-PADRE DNA at prime phase are important to induce effectors and memory CD8+ T cells. J Immunother. 2010;33(5):510–522. doi: 10.1097/CJI.0b013e3181d75cef. [DOI] [PubMed] [Google Scholar]
  • 85.Vigneron N, Stroobant V, Van den Eynde BJ, van der Bruggen P. Database of T cell-defined human tumor antigens: the 2013 update. Cancer Immun. 2013;13:15. [PMC free article] [PubMed] [Google Scholar]
  • 86.Khalili S, Rahbar MR, Dezfulian MH, Jahangiri A. In silico analyses of Wilms′ tumor protein to designing a novel multi-epitope DNA vaccine against cancer. J Theor Biol. 2015;379:66–78. doi: 10.1016/j.jtbi.2015.04.026. [DOI] [PubMed] [Google Scholar]
  • 87.Wu Y, Zhai W, Sun M, Zou Z, Zhou X, Li G, et al. A novel recombinant multi-epitope vaccine could induce specific cytotoxic T lymphocyte response in vitro and in vivo. Protein and peptide letters. 2017;24(6):573–580. doi: 10.2174/0929866524666170419152700. [DOI] [PubMed] [Google Scholar]
  • 88.Ahrends T, Babala N, Xiao Y, Yagita H, van Eenennaam H, Borst J. CD27 Agonism plus PD-1 blockade recapitulates CD4+ T-cell help in therapeutic anticancer vaccination. Cancer Res. 2016;76(10):2921–2931. doi: 10.1158/0008-5472.CAN-15-3130. [DOI] [PubMed] [Google Scholar]
  • 89.Chen R, Wang S, Yao Y, Zhou Y, Zhang C, Fang J, et al. Anti-metastatic effects of DNA vaccine encoding single-chain trimer composed of MHC I and vascular endothelial growth factor receptor 2 peptide. Oncol Rep. 2015;33(5):2269–2276. doi: 10.3892/or.2015.3820. [DOI] [PubMed] [Google Scholar]
  • 90.Villarreal DO, Wise MC, Siefert RJ, Yan J, Wood LM, Weiner DB. Ubiquitin-like molecule ISG15 acts as an immune adjuvant to enhance antigen-specific CD8 T-cell tumor immunity. Mol Ther. 2015;23(10):1653–1662. doi: 10.1038/mt.2015.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Vandermeulen G, Vanvarenberg K, De Beuckelaer A, De Koker S, Lambricht L, Uyttenhove C, et al. The site of administration influences both the type and the magnitude of the immune response induced by DNA vaccine electroporation. Vaccine. 2015;33(28):3179–3185. doi: 10.1016/j.vaccine.2015.05.005. [DOI] [PubMed] [Google Scholar]
  • 92.Melero I, Berman DM, Aznar MA, Korman AJ, Perez Gracia JL, Haanen J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015;15(8):457–472. doi: 10.1038/nrc3973. [DOI] [PubMed] [Google Scholar]
  • 93.McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, et al. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol. 2005;23(1):133–141. doi: 10.1200/JCO.2005.03.206. [DOI] [PubMed] [Google Scholar]
  • 94.Gupta S, Termini JM, Rivas Y, Otero M, Raffa FN, Bhat V, et al. A multi-trimeric fusion of CD40L and gp100 tumor antigen activates dendritic cells and enhances survival in a B16-F10 melanoma DNA vaccine model. Vaccine. 2015;33(38):4798–4806. doi: 10.1016/j.vaccine.2015.07.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Diniz MO, Sales NS, Silva JR, Ferreira LC. Protection against HPV-16-associated tumors requires the activation of CD8+ effector memory T cells and the control of myeloid-derived suppressor cells. Mol Cancer Ther. 2016;15(8):1920–1930. doi: 10.1158/1535-7163.MCT-15-0742. [DOI] [PubMed] [Google Scholar]
  • 96.Cunha LL, Marcello MA, Rocha-Santos V, Ward LS. Immunotherapy against endocrine malignancies: immune checkpoint inhibitors lead the way. Endocr Relat Cancer. 2017;24(12):T261–TT81. doi: 10.1530/ERC-17-0222. [DOI] [PubMed] [Google Scholar]
  • 97.Achkar T, Tarhini AA. The use of immunotherapy in the treatment of melanoma. J Hematol Oncol. 2017;10(1):88. doi: 10.1186/s13045-017-0458-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Puzanov I, Milhem MM, Andtbacka RHI, Minor DR, Hamid O, Li A, et al. Primary analysis of a phase 1b multicenter trial to evaluate safety and efficacy of talimogene laherparepvec (T-VEC) and ipilimumab (ipi) in previously untreated, unresected stage IIIB-IV melanoma. J Clin Oncol. 2014;32(15_suppl):9029. doi: 10.1200/jco.2014.32.15_suppl.9029. [DOI] [Google Scholar]
  • 99.Sandin LC, Eriksson F, Ellmark P, Loskog AS, Totterman TH, Mangsbo SM. Local CTLA4 blockade effectively restrains experimental pancreatic adenocarcinoma growth in vivo. Oncoimmunology. 2014;3(1):e27614. doi: 10.4161/onci.27614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Gao Y, Whitaker-Dowling P, Griffin JA, Barmada MA, Bergman I. Recombinant vesicular stomatitis virus targeted to Her2/neu combined with anti-CTLA4 antibody eliminates implanted mammary tumors. Cancer Gene Ther. 2009;16(1):44–52. doi: 10.1038/cgt.2008.55. [DOI] [PubMed] [Google Scholar]
  • 101.Furness AJ, Quezada SA, Peggs KS. Neoantigen heterogeneity: a key driver of immune response and sensitivity to immune checkpoint blockade? Immunotherapy. 2016;8(7):763–766. doi: 10.2217/imt-2016-0064. [DOI] [PubMed] [Google Scholar]
  • 102.Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–2199. doi: 10.1056/NEJMoa1406498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. doi: 10.1126/science.aaa1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Maleki VS. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunotherapy Cancer. 2018;6(1):157. doi: 10.1186/s40425-018-0479-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hu ZI, Ho AY, McArthur HL. Combined radiation therapy and immune checkpoint blockade therapy for breast Cancer. Int J Radiat Oncol Biol Phys. 2017;99(1):153–164. doi: 10.1016/j.ijrobp.2017.05.029. [DOI] [PubMed] [Google Scholar]
  • 106.Messenheimer DJ, Jensen SM, Afentoulis ME, Wegmann KW, Feng Z, Friedman DJ, et al. Timing of PD-1 blockade is critical to effective combination immunotherapy with anti-OX40. Clinical cancer research : an official journal of the American Association for Cancer Research. 2017;23(20):6165–6177. doi: 10.1158/1078-0432.CCR-16-2677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kleinovink JW, Fransen MF, Lowik CW, Ossendorp F. Photodynamic-immune checkpoint therapy eradicates local and distant tumors by CD8(+) T cells. Cancer immunology research. 2017;5(10):832–838. doi: 10.1158/2326-6066.CIR-17-0055. [DOI] [PubMed] [Google Scholar]
  • 108.Xue W, Metheringham RL, Brentville VA, Gunn B, Symonds P, Yagita H, et al. SCIB2, an antibody DNA vaccine encoding NY-ESO-1 epitopes, induces potent antitumor immunity which is further enhanced by checkpoint blockade. Oncoimmunology. 2016;5(6):e1169353. doi: 10.1080/2162402X.2016.1169353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lopes A, Vanvarenberg K, Kos S, Lucas S, Colau D, Van den Eynde B, et al. Combination of immune checkpoint blockade with DNA cancer vaccine induces potent antitumor immunity against P815 mastocytoma. Sci Rep. 2018;8(1):15732. doi: 10.1038/s41598-018-33933-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Masucci GV, Cesano A, Hawtin R, Janetzki S, Zhang J, Kirsch I, et al. Validation of biomarkers to predict response to immunotherapy in cancer: volume I - pre-analytical and analytical validation. Journal for immunotherapy of cancer. 2016;4:76. doi: 10.1186/s40425-016-0178-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chabanon RM, Pedrero M, Lefebvre C, Marabelle A, Soria JC, Postel-Vinay S. Mutational landscape and sensitivity to immune checkpoint blockers. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016;22(17):4309–4321. doi: 10.1158/1078-0432.CCR-16-0903. [DOI] [PubMed] [Google Scholar]
  • 112.Sasso MS, Lollo G, Pitorre M, Solito S, Pinton L, Valpione S, et al. Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy. Biomaterials. 2016;96:47–62. doi: 10.1016/j.biomaterials.2016.04.010. [DOI] [PubMed] [Google Scholar]
  • 113.Sevko A, Kremer V, Falk C, Umansky L, Shurin MR, Shurin GV, et al. Application of paclitaxel in low non-cytotoxic doses supports vaccination with melanoma antigens in normal mice. J Immunotoxicol. 2012;9(3):275–281. doi: 10.3109/1547691X.2012.655343. [DOI] [PubMed] [Google Scholar]
  • 114.Wu J, Waxman DJ. Metronomic cyclophosphamide eradicates large implanted GL261 gliomas by activating antitumor Cd8(+) T-cell responses and immune memory. Oncoimmunology. 2015;4(4):e1005521. doi: 10.1080/2162402X.2015.1005521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Xia Q, Geng F, Zhang FF, Liu CL, Xu P, Lu ZZ, et al. Cyclophosphamide enhances anti-tumor effects of a fibroblast activation protein alpha-based DNA vaccine in tumor-bearing mice with murine breast carcinoma. Immunopharmacol Immunotoxicol. 2017;39(1):37–44. doi: 10.1080/08923973.2016.1269337. [DOI] [PubMed] [Google Scholar]
  • 116.Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F, et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity. 2016;44(2):343–354. doi: 10.1016/j.immuni.2015.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Finke JH, Rini B, Ireland J, Rayman P, Richmond A, Golshayan A, et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14(20):6674–6682. doi: 10.1158/1078-0432.CCR-07-5212. [DOI] [PubMed] [Google Scholar]
  • 118.Kwilas AR, Donahue RN, Tsang KY, Hodge JW. Immune consequences of tyrosine kinase inhibitors that synergize with cancer immunotherapy. Cancer Cell Microenviron. 2015;2(1):e677. 10.14800/ccm.677. [DOI] [PMC free article] [PubMed]
  • 119.Generali D, Bates G, Berruti A, Brizzi MP, Campo L, Bonardi S, et al. Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15(3):1046–1051. doi: 10.1158/1078-0432.CCR-08-1507. [DOI] [PubMed] [Google Scholar]
  • 120.Ferrara TA, Hodge JW, Gulley JL. Combining radiation and immunotherapy for synergistic antitumor therapy. Curr Opin Mol Ther. 2009;11(1):37–42. [PMC free article] [PubMed] [Google Scholar]
  • 121.Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 2004;64(12):4328–4337. doi: 10.1158/0008-5472.CAN-04-0073. [DOI] [PubMed] [Google Scholar]
  • 122.ClinicalTrials.gov. Accessed 21/02/2019 [21/02/2019]. Available from: https://clinicaltrials.gov/.
  • 123.Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386(10008):2078–2088. doi: 10.1016/S0140-6736(15)00239-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kim TJ, Jin HT, Hur SY, Yang HG, Seo YB, Hong SR, et al. Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat Commun. 2014;5:5317. doi: 10.1038/ncomms6317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tiriveedhi V, Tucker N, Herndon J, Li L, Sturmoski M, Ellis M, et al. Safety and preliminary evidence of biologic efficacy of a mammaglobin-a DNA vaccine in patients with stable metastatic breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014;20(23):5964–5975. doi: 10.1158/1078-0432.CCR-14-0059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tiriveedhi V, Fleming TP, Goedegebuure PS, Naughton M, Ma C, Lockhart C, et al. Mammaglobin-a cDNA vaccination of breast cancer patients induces antigen-specific cytotoxic CD4+ICOShi T cells. Breast Cancer Res Treat. 2013;138(1):109–118. doi: 10.1007/s10549-012-2110-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Soysal SD, Muenst S, Kan-Mitchell J, Huarte E, Zhang X, Wilkinson-Ryan I, et al. Identification and translational validation of novel mammaglobin-a CD8 T cell epitopes. Breast Cancer Res Treat. 2014;147(3):527–537. doi: 10.1007/s10549-014-3129-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Eriksson F, Totterman T, Maltais AK, Pisa P, Yachnin J. DNA vaccine coding for the rhesus prostate specific antigen delivered by intradermal electroporation in patients with relapsed prostate cancer. Vaccine. 2013;31(37):3843–3848. doi: 10.1016/j.vaccine.2013.06.063. [DOI] [PubMed] [Google Scholar]
  • 129.McCann KJ, Godeseth R, Chudley L, Mander A, Di Genova G, Lloyd-Evans P, et al. Idiotypic DNA vaccination for the treatment of multiple myeloma: safety and immunogenicity in a phase I clinical study. Cancer Immunol Immunother. 2015;64(8):1021–1032. doi: 10.1007/s00262-015-1703-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ottensmeier C, Bowers M, Hamid D, Maishman T, Regan S, Wood W, et al. Wilms’ tumour antigen 1 immunity via DNA fusion gene vaccination in haematological malignancies by intramuscular injection followed by intramuscular electroporation: a phase II non-randomised clinical trial (WIN). Efficacy and mechanism evaluation. Southampton: NIHR Journals Library; 2016. [PubMed] [Google Scholar]
  • 131.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. doi: 10.1186/1471-2407-12-361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Diaz CM, Chiappori A, Aurisicchio L, Bagchi A, Clark J, Dubey S, 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. doi: 10.1186/1479-5876-11-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15(17):5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Aurisicchio L, Salvatori E, Lione L, Bandini S, Pallocca M, Maggio R, et al. Poly-specific neoantigen-targeted cancer vaccines delay patient derived tumor growth. Journal of experimental & clinical cancer research : CR. 2019;38(1):78. doi: 10.1186/s13046-019-1084-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Overgaard NH, Fan TM, Schachtschneider KM, Principe DR, Schook LB, Jungersen G. Of mice, dogs, pigs, and men: choosing the appropriate model for Immuno-oncology research. ILAR J. 2018. [DOI] [PubMed]
  • 136.Gableh F, Saeidi M, Hemati S, Hamdi K, Soleimanjahi H, Gorji A, et al. Combination of the toll like receptor agonist and alpha-Galactosylceramide as an efficient adjuvant for cancer vaccine. J Biomed Sci. 2016;23:16. doi: 10.1186/s12929-016-0238-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Allen A, Wang C, Caproni LJ, Sugiyarto G, Harden E, Douglas LR, et al. Linear doggybone DNA vaccine induces similar immunological responses to conventional plasmid DNA independently of immune recognition by TLR9 in a pre-clinical model. Cancer immunology, immunotherapy : CII. 2018;67(4):627–638. doi: 10.1007/s00262-017-2111-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Jiang W, Wang S, Chen H, Ren H, Huang X, Wang G, et al. A bivalent heterologous DNA virus-like-particle prime-boost vaccine elicits broad protection against both group 1 and 2 influenza a viruses. J Virol. 2017;91(9):e02052–e02016. doi: 10.1128/JVI.02052-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Longo DL, Dastru W, Consolino L, Espak M, Arigoni M, Cavallo F, et al. Cluster analysis of quantitative parametric maps from DCE-MRI: application in evaluating heterogeneity of tumor response to antiangiogenic treatment. Magn Reson Imaging. 2015;33(6):725–736. doi: 10.1016/j.mri.2015.03.005. [DOI] [PubMed] [Google Scholar]
  • 140.Nguyen-Hoai T, Pham-Duc M, Gries M, Dorken B, Pezzutto A, Westermann J. CCL4 as an adjuvant for DNA vaccination in a Her2/neu mouse tumor model. Cancer Gene Ther. 2016;23(6):162–167. doi: 10.1038/cgt.2016.9. [DOI] [PubMed] [Google Scholar]
  • 141.Xia Q, Zhang FF, Geng F, Liu CL, Xu P, Lu ZZ, et al. Anti-tumor effects of DNA vaccine targeting human fibroblast activation protein alpha by producing specific immune responses and altering tumor microenvironment in the 4T1 murine breast cancer model. Cancer immunology, immunotherapy. 2016;65(5):613–624. doi: 10.1007/s00262-016-1827-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Witt K, Ligtenberg MA, Conti L, Lanzardo S, Ruiu R, Wallmann T, et al. Cripto-1 plasmid DNA vaccination targets metastasis and cancer stem cells in murine mammary carcinoma. Cancer Immunology Research. 2018. 10.1158/2326-6066.CIR-17-0572. [DOI] [PubMed]
  • 143.Zhong B, Ma G, Sato A, Shimozato O, Liu H, Li Q, et al. Fas ligand DNA enhances a vaccination effect by coadministered DNA encoding a tumor antigen through augmenting production of antibody against the tumor antigen. J Immunol Res. 2015;2015:743828. doi: 10.1155/2015/743828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Danishmalik SN, Sin JI. Therapeutic tumor control of HER2 DNA vaccines is achieved by an alteration of tumor cells and tumor microenvironment by gemcitabine and anti-gr-1 ab treatment in a HER2-expressing tumor model. DNA Cell Biol. 2017;36(9):801–811. doi: 10.1089/dna.2017.3810. [DOI] [PubMed] [Google Scholar]
  • 145.Liu C, Xie Y, Sun B, Geng F, Zhang F, Guo Q, et al. MUC1- and Survivin-based DNA vaccine combining Immunoadjuvants CpG and interleukin-2 in a Bicistronic expression plasmid generates specific immune responses and antitumour effects in a murine colorectal carcinoma model. Scand J Immunol. 2018;87(2):63–72. doi: 10.1111/sji.12633. [DOI] [PubMed] [Google Scholar]
  • 146.Son HY, Apostolopoulos V, Chung JK, Kim CW, Park JU. Protective efficacy of a plasmid DNA vaccine against transgene-specific tumors by Th1 cellular immune responses after intradermal injection. Cell Immunol. 2018;329:17–26. doi: 10.1016/j.cellimm.2018.04.003. [DOI] [PubMed] [Google Scholar]
  • 147.Yin X, Wang W, Zhu X, Wang Y, Wu S, Wang Z, et al. Synergistic antitumor efficacy of combined DNA vaccines targeting tumor cells and angiogenesis. Biochem Biophys Res Commun. 2015;465(2):239–244. doi: 10.1016/j.bbrc.2015.08.003. [DOI] [PubMed] [Google Scholar]
  • 148.Gao FS, Zhan YT, Wang XD, Zhang C. Enhancement of anti-tumor effect of plasmid DNA-carrying MUC1 by the adjuvanticity of FLT3L in mouse model. Immunopharmacol Immunotoxicol. 2018;40(4):353–357. doi: 10.1080/08923973.2018.1498099. [DOI] [PubMed] [Google Scholar]
  • 149.Yu S, Wang F, Fan L, Wei Y, Li H, Sun Y, et al. BAP31, a promising target for the immunotherapy of malignant melanomas. J Exp Clin Cancer Res. 2015;34:36. doi: 10.1186/s13046-015-0153-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Mahlmann K, Feige K, Juhls C, Endmann A, Schuberth HJ, Oswald D, et al. Local and systemic effect of transfection-reagent formulated DNA vectors on equine melanoma. BMC Vet Res. 2015;11:132. doi: 10.1186/s12917-015-0422-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.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 letters. 2015;356(2 Pt B):676–685. doi: 10.1016/j.canlet.2014.10.019. [DOI] [PubMed] [Google Scholar]
  • 152.Rekoske BT, Smith HA, Olson BM, Maricque BB, McNeel DG. PD-1 or PD-L1 blockade restores antitumor efficacy following SSX2 epitope-modified DNA vaccine immunization. Cancer Immunology Research. 2015;3(8):946–955. doi: 10.1158/2326-6066.CIR-14-0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zhao Y, Wei Z, Yang H, Li X, Wang Q, Wang L, et al. Enhance the anti-renca carcinoma effect of a DNA vaccine targeting G250 gene by co-expression with cytotoxic T-lymphocyte associated antigen-4(CTLA-4) Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2017;90:147–152. doi: 10.1016/j.biopha.2017.03.015. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Journal of Experimental & Clinical Cancer Research : CR are provided here courtesy of BMC

RESOURCES