Next Article in Journal
CD63-Mediated SARS-CoV-2 RBD Fusion Neoantigen DNA Vaccine Enhances Antitumor Immune Response in a Mouse Panc02 Model via EV-Targeted Delivery
Previous Article in Journal
Long-Term Follow-Up of T Cell Immunity Against Orthopoxviruses in People Living with HIV After Vaccination and Natural Monkeypox Virus Infection
Previous Article in Special Issue
Epitope Profiling of SARS-CoV-2 Spike Antigen Provides a Novel Strategy for Developing ELISAs Specific for Different Spike Protein Variants in Bivalent Vaccine Formulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer

Department of Medicine, Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
*
Author to whom correspondence should be addressed.
Vaccines 2025, 13(9), 976; https://doi.org/10.3390/vaccines13090976
Submission received: 31 July 2025 / Revised: 9 September 2025 / Accepted: 11 September 2025 / Published: 14 September 2025
(This article belongs to the Special Issue New Approaches to Vaccine Development and Delivery)

Abstract

Immunotherapy is now an established therapy for nearly a third of patients with cancer. Most therapies, typically using cytokines or checkpoint blockade therapy, rely on global activation of immune effector cells. The ability of vaccines to activate specific populations of cells has led to a renewed interest in their ability to treat cancers, either alone or with other immune therapies or other conventional therapies. The COVID-19 pandemic sparked a new interest in nucleic acid vaccines with the development of new technologies and the short manufacturing time for vaccine implementation. Nucleic acid-based cancer vaccines have been studied for decades, but have shown modest anti-tumor efficacy as monotherapies, as many of these vaccines encode for shared tumor-associated antigens (TAAs) and must overcome immune tolerance. New developments, technologies, routes of delivery, and combination therapies have paved the way for new approaches and clinical trials involving nucleic acid vaccines for the treatment of cancer. Here we review mRNA and pDNA vaccines for the treatment of cancer, including similarities and differences in their mechanisms of action, an overview of these treatment modalities in preclinical and clinical studies, methods to improve these vaccine strategies, and exciting new combination approaches in development.

1. Introduction to Nucleic Acid Vaccines for the Treatment of Cancer

Cancer incidence and mortality remain a major global concern, with 20 million new cases diagnosed and 9.7 million deaths in the year 2022 alone [1]. For many cancers, the first line of treatment is surgery, followed by radiotherapy and/or chemotherapy. However, it is estimated that approximately 90% of cancers develop resistance [2,3]. Resistance to primary treatment underscores the need for new therapies or combination therapies. One treatment modality that has been gaining popularity in recent years is the field of immunotherapy, and cancer vaccines in particular.
Cancer vaccines are a way to “train” the body’s immune system to recognize and kill the tumor. Cancer vaccines developed as a concept in the late 1800s when Dr. William Coley observed tumor regressions after patients were injected with inactivated bacteria [4,5]. Since then, numerous types of cancer vaccines have been developed and are reviewed elsewhere, including viral vector-based [6], bacterial-based [7], protein or peptide-based [8], cell-based (including transfection of antigen presenting cells with mRNA for cell therapy) [9], and nucleic acid vaccines using messenger RNA (mRNA) or plasmid DNA (pDNA, hereafter referred to simply as “DNA”). Since viral and bacterial-based vectors rely on attenuated pathogens to enter cells and encode proteins, a simpler and safer way to deliver the genetic material to cells is needed. Nucleic acid vectors can serve as an efficient and safe alternative for the delivery of genetic material, which will be the focus of this review article.
Nucleic acid vaccines are advantageous to various cancer vaccine platforms due to their cost, ease of manufacture, and safety profile. The first DNA vaccine to enter human clinical trials was a vaccine targeting HIV in 1998 [10]. Although DNA vaccines were deemed safe from these initial trials, it was unclear whether these vaccines mounted significant clinical efficacy. Similarly, mRNA vaccines have a long history. Unlike DNA, mRNA cannot efficiently be directly delivered to cells, due to the inherent instability of the molecule and the presence of RNases in blood and tissues [11]. Hence, the first “mRNA vaccines” were based on the concept of transfecting and transferring dendritic cells with mRNA into the body for protein translation, with the first human clinical trial for cancer therapy based on this technology conducted in 2003 (NCT00003432) [12]. These initial studies, as well as preclinical studies in mice, eventually paved the way for direct injection of synthetic mRNA using modified nucleotides and lipid nanoparticles to evade immune detection, prevent degradation, and improve efficacy [13]. However, the true advancement of both mRNA and DNA vaccines came with the COVID-19 pandemic, where the first mRNA and DNA vaccines were approved in humans for the prophylactic infectious disease setting [14,15]. Although nucleic acids vaccines have proven highly successful in the infectious disease setting, there are still many challenges and issues that remain to be addressed for these vaccines to have clinical efficacy in the cancer setting.

2. Mechanisms of Action of DNA Versus mRNA Vaccines

2.1. Mechanisms of Cellular Uptake and Antigen Presentation

DNA and mRNA vaccines both elicit immune responses by enabling host cells to produce antigenic proteins that can activate the immune system [16]. However, the cellular mechanisms underlying their delivery, uptake, processing, and antigen presentation differ due to the distinct nature of DNA and RNA molecules. Both DNA and mRNA vaccines rely on delivering genetic material into host cells, often using lipid-based nanoparticles or other vectors to facilitate cellular entry [17].
mRNA vaccines are typically directly introduced into the cytoplasm of target cells using either lipid nanoparticle carriers or electroporation. Following cellular uptake, which commonly occurs via endocytosis of mRNA-LNP complexes, the mRNA is released into the cytoplasm, where it is translated into the target antigen by ribosomes [18]. Ionizable lipids play a crucial role in this process, as they enable the delivery vehicle to escape the endosomal compartment by undergoing a conformational change at the lower pH of the endosome. This “dumping” of mRNA into the cytoplasm ensures efficient protein translation [19]. Once the protein is synthesized in the cytosol, it can be broken down into peptides via the endogenous pathway and presented on MHC class I molecules to activate CD8+ T cells [20]. mRNA can also be taken up by professional antigen-presenting cells (APCs) such as dendritic cells (DCs), which process and present the antigen via both MHC class I and MHC class II pathways, allowing for the activation of both CD8+ and CD4+ cytotoxic and helper T cells [21]. Classical antigen presentation by professional APCs occurs when exogenous antigen is processed and presented on MHC class II molecules, activating CD4+ helper-T cells. However, cross-presentation, a mechanism where exogenously produced antigens can be presented on MHC class I molecules, can occur if the mRNA is taken up and expressed by non-professional APC, and the protein is take up and presented by dendritic cells to activate CD8+ T cells [22].
In contrast, DNA vaccines require an additional step of entering the cell nucleus for transcription [23]. After uptake, typically via endocytosis, DNA must successfully traverse the cytoplasm and cross the nuclear membrane to access the transcriptional machinery [24]. This necessity for nuclear delivery is a key distinction from mRNA vaccines [25]. Once inside the nucleus, the DNA is transcribed into mRNA, which then follows the same pathway as mRNA vaccines—translation in the cytoplasm, antigen production, and subsequent presentation on MHC molecules. However, the need for nuclear entry can pose a barrier, reducing the overall efficiency of antigen production compared to mRNA vaccines [16]. Additionally, the instability of mRNA in vivo may reduce its effectiveness as an immune modulator, whereas DNA is known to be stable for over six months in vivo [26]. mRNA’s instability also causes it to be degraded rapidly, resulting in short bursts of protein expression, potentially causing a need for frequent booster immunizations. One trafficking study using eGFP mRNA reported peak mRNA expression 8 h after immunization, and peak protein expression 24 h after immunization [27].
While both mRNA and DNA vaccines can be taken up by a variety of cell types, including myocytes and fibroblasts at the site of injection, professional APCs, such as dendritic cells, macrophages, and B cells, are the most critical for the initiation of adaptive immune responses [28]. mRNA vaccines are particularly adept at targeting DCs, especially when delivered in nanoparticle formulations designed to enhance uptake by these cells [29]. The rapid translation of mRNA in the cytoplasm makes it a highly efficient platform for inducing robust immune responses, particularly when aimed at activating both cellular and humoral arms of the immune system [30]. In contrast, DNA vaccines may be taken up by a broader range of cell types, but due to their requirement for nuclear entry, the efficiency of antigen presentation may vary, especially in non-dividing cells where nuclear access is more limited [31]. B cells have been shown to be the primary professional APC that can uptake and process the naked DNA plasmid to present the encoding antigen, however B cells require licensing by DCs to enable antigen presentation [32,33]. To enhance uptake by APCs and improve immunogenicity, DNA vaccines often rely on additional strategies, such as electroporation, to facilitate more effective nuclear delivery [34].
A key aspect of both mRNA and DNA vaccine delivery systems is the use of ionizable lipids, which assist in endosomal escape [35]. Ionizable lipids are neutral at physiological pH but become positively charged in the acidic environment of the endosome, destabilizing the endosomal membrane and facilitating the release of their cargo into the cytoplasm [36]. This mechanism is particularly important for mRNA vaccines, where protection from nucleases and release into the cytoplasm are necessary for efficient antigen expression [37]. The same strategy can be applied to DNA vaccines, though the DNA must again be transported to the nucleus after cytoplasmic release, adding an extra step to the antigen production process [38,39].
In summary, the key distinctions between DNA and mRNA vaccines lie in the requirement for DNA to reach the nucleus versus the cytoplasmic localization of mRNA, and in the mechanisms of delivery, processing, and antigen presentation. Both platforms have their unique advantages, but the more direct cytoplasmic pathway of mRNA may offer a more rapid and efficient means of inducing immune responses.

2.2. Mechanisms of Innate Immune Recognition

Cells have inherent processes to recognize mRNA and DNA in the cell and send out “danger signals” when these molecules are recognized, generating an innate immune signaling cascade. The key sensors that recognize foreign RNA in the endosomes are Toll-like receptors (TLR) 3 and TLR 7/8 [40]. There are also cytosolic RNA sensors, such as RIG-1 and MDA5, that play a role in RNA detection and result in an innate immune signaling cascade [40]. Since detection of foreign mRNA and activation of these sensors may result in the destruction of the mRNA molecule, more recent efforts have shifted to allow for modified mRNA that can evade immune detection by these sensors. This has enabled the modern mRNA-LNP vaccines to successfully deliver their cargo without undergoing immune destruction [41,42,43]. In contrast, there is a greater quantity of DNA sensors inside the cell compared to mRNA. In the endosomes, the major DNA sensor is TLR9 [44]. There are also many cytosolic DNA sensors, such as cGAS/STING, AIM2, IFI16, the DExD/H-box helicase family members, RNA polymerase 3, DAI, DNA-PK, and MRE-11 [44]. In general, cellular recognition of foreign DNA leads to a pro-inflammatory response, characterized by the secretion of pro-inflammatory cytokines and type I interferon [44]. While conceivable that these differences in intracellular RNA and DNA sensors could affect the nature of the immune response generated by these different vaccine approaches, this has not been rigorously assessed.

2.3. Types of Immune Responses Generated by Immunization with mRNA or DNA Vaccines

While no direct comparisons of the resulting immune responses generated by mRNA and DNA vaccines have been made, there have been reports of the types of immune responses generated by each of these types of vaccines. In the case of mRNA cancer vaccines, there have been reports of strong cell-mediated immunity, characterized by the generation of CD4+ and CD8+ antigen-specific immune responses [45,46,47,48]. Similarly, cancer DNA vaccines have been reported to generate cellular immune responses across multiple studies [49]. Notably, many studies using DNA vaccines to encode tumor antigens have also demonstrated humoral immune responses [49]. Although more studies and trials have been conducted using DNA vaccines, both DNA and mRNA vaccines appear to elicit cellular and humoral immunity. The magnitude, phenotype, and longevity of these cellular responses are unknown, though. Insights regarding direct comparisons of mRNA and DNA will be beneficial in determining whether these treatment modalities are functioning similarly or whether there may be an advantage to combining DNA vaccines and mRNA vaccines for the treatment of cancer.

3. mRNA and DNA Vaccines in Preclinical Animal Models Encoding the Same Antigens

Targets for tumor vaccines can be categorized into tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs). TAAs are “shared” self-antigens which may be aberrantly expressed or overexpressed in tumor cells. In general, due to mechanisms of central and peripheral tolerance, TAAs are generally characterized by low immunogenicity, and T cell receptors often have low affinity for these antigens [50]. Therefore, nucleic acid vaccines encoding TAAs may require additional methods to enhance their immunogenicity. These strategies include incorporating immunoadjuvants, optimizing administration delivery methods and routes, employing prime-boost regimens, or encoding multiple antigens to target tumor heterogeneity [51]. In contrast, most TSAs result from aberrant antigen expression (e.g., frame-shift mutations or individual point mutations) within the tumor cells resulting in mutation-associated neoantigens (MANA) [52]. Since these antigens would not have undergone thymic tolerance, they tend to elicit slightly stronger immune responses and may be more readily targeted through vaccination, at least in murine models [53].
While many preclinical studies have utilized mRNA and DNA vaccines for the treatment of cancer, no studies have directly compared mRNA and DNA vaccines in the same study. Therefore, here we will focus on studies that have examined either mRNA or DNA encoding the same antigen as a means of comparing their actions. Table 1 highlights those studies that have used either mRNA or DNA targeting the same antigen in similar tumor models. Only preclinical studies involving the direct administration of nucleic acid vaccines into the subjects—excluding those utilizing ex vivo DC loading—were considered. Specific target antigens are described in detail below.

3.1. Mucin 1 (MUC1)

MUC1 is a transmembrane glycoprotein on epithelial cells, with aberrant glycosylation linked to malignant cellular transformation. Aberrant expression of MUC1 has been found in several types of cancer, including gastric, breast, ovarian, and bladder cancers. Therefore, MUC1 has been one of the major targets for cancer vaccine development [75]. Various nucleic acid vaccines encoding MUC1 have been developed to elicit anti-tumor response against MUC1-expressing tumors in murine models. An early study of prophylactic MUC1 plasmid DNA (pMUC1) displayed limited efficacy in inducing tumor protection in a murine colorectal cancer model. Co-administration of a plasmid encoding the murine IL-18 was needed to enhance the tumor protection, memory response, and epitope spreading [54]. However, a therapeutic MUC1 DNA vaccine, pcDNA3.1-MUC1 (20 μg, delivered by gene gun and electroporation), elicited enhanced anti-tumor responses in the murine colorectal model. Immunization promoted IFNγ and granzyme B secretion by CD8+ T cells and enhanced their proliferation and cytotoxicity against the target murine colorectal cells in vitro and in vivo [55].
Efforts to improve the efficacy of DNA vaccines have included strategies such as developing plasmid DNA that encodes multiple tumor antigens to address tumor heterogeneity. A DNA vaccine encoding MUC1 and survivin fusion antigens, VR-MS, increased in vitro killing of MUC1+ survivin+ B16 cells and significantly delayed tumor growth using a 100 μg dose [57]. Additional strategies to improve DNA vaccines have been investigated, such as enhancing the immunogenicity of plasmid DNA by designing fusion plasmids that encode multiple TAAs and immunoadjuvants. CpDV-IL2-MS is a bicistronic DNA plasmid encoding survivin, MUC1, and IL-2 [58]. In a study by Liu et al., BABL/c mice with MUC1+ survivin+ colorectal and lung tumors were immunized with CpDV-IL2-MS, CpMS+IL-2, or PBS to examine immunological and antitumor effects of the fusion plasmid. Mice immunized with 100 μg CpDV-IL2-MS had equivalent IFN-γ secretion, tumor cell cytotoxicity, tumor growth rates and survival compared to mice treated with CpMS+IL-2 [58]. Furthermore, incorporation of soluble PD-1 into the plasmid (CpDV-IL2-sPD1/MS) elicited a greater number of cures in mice bearing colorectal or lung cancers [76]. Similarly, mice receiving 100 μg of a chimeric MUC1 DNA vaccine, encoding the transmembrane- and C-terminal domain-deleted Muc1 gene fused to the human HSP70 gene, had greater anti-tumor effects. Immunization with the MUC1 vaccine in the murine B16/MUC1+ melanoma model induced greater MUC1-specific CD8+ T cell response compared to HSP70 or deletion of the C-terminal domain alone [56].
Although still in its early stages, research using mRNA vaccines encoding MUC1 is also underway. Mice inoculated with 4T1 mammary carcinoma cells had enhanced IFN-γ secretion and MUC1-specific CD8+ T cell killing when immunized with 10 μg of a MUC1 mRNA nanovaccine encapsulated in lipid/calcium/phosphate nanoparticles (LCP) compared to mRNA or LCP alone. However, additional combination of MUC1 LCP-mRNA with an anti-CTLA4 antibody was needed to enhance CD8+ T cell recruitment, inhibit the tumor-promoting STAT3 pathway, and further enhance anti-tumor responses in the murine triple negative breast cancer cells, in vivo [59].

3.2. Melanoma Antigen Family A (MAGE-A)

MAGE-A is a cancer-testis antigen (CTA) found in multiple cancer types such as melanoma, brain, breast, lung and ovarian cancers [77]. Unlike most TAAs, CTAs are considered highly desirable due to their distinct characteristics, including restricted expression in immune-privileged organs, tumor-specific expression, and widespread presence across various types of cancers [78]. Immunization by electroporation with 30 μg of a DNA plasmid encoding MAGE-A3 isoform significantly enhanced Th1 (IgG2a, IFN-γ) and Th2 (IgG1, IL-4) responses, and delayed tumor growth when compared to mice immunized with empty vector in the murine lung cancer LLC model. These responses were further augmented when the plasmid was modified to encode MAGE-A3 and soluble PD-1 [60]. While many studies have targeted MAGE-A3, tumor cells can express two or more MAGE-A antigens [79]. Duperret et al. analyzed the Cancer Genome Atlas and found that patients across tumor types commonly expressed multiple MAGE-A isoforms [61]. Therefore, this group designed a synthetic consensus MAGE-A DNA vaccine encoding murine MAGE-A1, -A2, -A3, -A5, -A6, and -A8. Splenocytes from immunized mice displayed IFN-γ response and exhibited high cytolytic potential (CD107a+ T-bet+ IFN-γ+) when stimulated with MAGE-A peptides in vitro. In an inducible melanoma model, driven by Cre activation in melanocytes via tamoxifen induction, immunization using 25 μg DNA, delivered by electroporation, resulted in delayed tumor growth, reduced tumor invasion depth in the skin, and increased infiltration of CD8+ T cells with an activated phenotype (CD44+ PD-1+) [61].
In a study by Choi et al., investigators immunized groups of mice with MAGE-A3 mRNA encapsulated in various lipid nanoparticles, and inoculated mice with CT26 colorectal cancer cells two weeks later. Prophylactic use of 20 μg of MAGE-A3 mRNA encapsulated in various lipid nanoparticles significantly delayed tumor growth, limited lung and liver metastases, and prolonged survival compared to untreated control mice. Researchers repeated this study but administered lipid nanoparticle encapsulated MAGE-A3 mRNA post-tumor inoculation. Therapeutic vaccination post implantation of CT26 cells also delayed tumor growth, comparable to a standard chemotherapeutic agent doxorubicin, but without weight loss that occurred with chemotherapy. The vaccine enhanced humoral responses (IgG2a and IgG2b), although no significant changes in IFN-γ or IL-4 levels were observed in the sera. In vitro co-culture of splenocytes with tumor lysates did increase IL-4, although IFN-γ remained unchanged [62].

3.3. Human Papillomavirus (HPV)

HPV is one of the most prevalent sexually transmitted infections and accounts for approximately 5% of all cancer cases worldwide. The virus has been associated with cervical, vulvar, vaginal, anal, and head and neck cancers [80]. An FDA-approved prophylactic HPV vaccine is estimated to hold the potential to prevent more than 90% of cancers caused by HPV [81]. However, for patients with existing HPV-positive cancers, therapeutic approaches are being developed to target HPV antigens, particularly the oncogenic viral early protein E6 and E7, using DNA and mRNA vaccines. Mice bearing HPV-associated TC-1 tumors were immunized by gene gun with 16 μg of plasmid DNA encoding HPV16 E7 fused to calreticulin (CRT) to enhance MHC-I presentation. Immunization induced a robust E7-specific CD8+ T cell response, resulting in CD8+ T cell-dependent protection against the tumor and pulmonary nodules. Moreover, the inclusion of CRT triggered antiangiogenic activity, which contributed to the reduction in pulmonary tumor nodules [63]. Similarly, other studies have included a related immunoadjuvant, heat shock protein (HSP) 70, to enhance immunogenicity of the E7 encoding plasmid DNA. While the administration of E7 vaccine alone delayed tumor growth compared to the PBS or empty vector controls, the tumor growth suppression was enhanced when co-administered with a vector encoding HSP70. Immunization increased lymphocyte proliferation and in vitro cell-mediated cytotoxicity [64]. These findings underscore the importance of an effective adjuvant, such as CRT or HSP70, to augment the immune responses of HPV-targeting immunotherapies. To illustrate this, further enhancement of the plasmid DNA design, by encoding a fusion protein product consisting of signal peptide fused to Mycobacterium tuberculosis HSP70 and HPV16 E7 (with a point mutation to eliminate oncogenic potential), increased the number of IFN-γ secreting E7-specific CD8+ T cells in the spleen and induced complete in vivo protection against TC-1 tumors [65]. The plasmid design was also further improved by Peng et al. to optimize codons for expression and encode 4 total HPV antigens: HPV16 E6, HPV16 E7, HPV18 E6, and HPV18 E7. Mice immunized with 25 μg of this vaccine (pBI-11) had an increased number of HPV-specific IFN-γ-secreting CD8+ T cells. Immunization of TC-1 tumor-bearing mice also had reduced tumor growth [66].
HPV vaccines have also been developed using mRNA approaches. Immunization with 10 μg of mRNA-LNP encoding HPV E7 expanded HPV-specific CD8+ T cells in the spleen and blood of mice implanted with syngeneic HPV+ oropharyngeal squamous cell carcinoma (mEERL) cells [67]. Subsequent single-cell RNA sequencing (scRNA-seq) analysis comparing vaccinated mice to those immunized with PBS or an empty vector revealed distinct clustering patterns in T cells of the spleen and tumors of vaccinated mice. Vaccination enhanced innate immune activation in the spleen and promoted both effector differentiation and exhaustion of CD8+ T cells in the tumor microenvironment. Coupled with scTCR-seq, the vaccine induced clonal expansion primarily in tumor-infiltrating effector and exhausted cell subclusters. Therefore, the HPV mRNA-LNP vaccine drove effector cells toward an exhaustion trajectory, marked by elevated expression of both effector and inhibitory markers, which play crucial roles in mediating antitumor immunity [67]. Other HPV-targeting mRNA vaccines also demonstrated similar anti-tumor efficacy. HPV16 E6/E7-based mRNA-LNP immunization led to significant tumor regression within the first week of treatment in mice bearing the HPV16+ cervical cancer model. Suppression of tumor volume and complete responses were found to be dose-dependent [68]. Similarly, bi-weekly immunization of mice with 30 μg of mHTV-02, encoding HPV-16 and HPV-18 E6/E7 mRNA-LNP, induced an antigen-specific immune response, indicated by increased secretion of IFN-γ and IL-2, and frequency of IFN-γ+ and TNF-α+ CD8+ T cells [69]. Immune cell infiltration and anti-tumor effects were examined in mice with HPV16 E6/E7-positive TC-1 tumors that were vaccinated with mHTV-02 weekly. mHTV-02 immunization increased the infiltration of multifunctional (IFN-γ+ GzmB+) CD8+ T cells and effector memory CD4+ and CD8+ T cells to the tumor. Additionally, vaccine-induced regression of tumor growth and prolonged survival was observed [69].

3.4. Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS)

KRAS is a small GTPase that transduces signals from receptor tyrosine kinases to downstream pathways, regulating cell proliferation, differentiation, and transformation. Therefore, KRAS represents one of the commonly mutated proto-oncogenes, especially at the G12 residue, found in lung, pancreatic, and colon cancers [82]. While the KRAS G12C mutation has been a successful target, through small molecule inhibitors such as sotorasib and adagrasib, there has been less success targeting other G12 mutations [83]. However, there have been recent attempts to target the KRAS G12 variant through nucleic acid vaccines. Immunization using a plasmid DNA encoding the inactive mutant KrasG12DN17, a dominant-negative mutant devoid of oncogenic activity, produced significant therapeutic responses against a murine lung adenocarcinoma model (TetO-Kras4bG12D/Scgb1a1-rtTA). Mice were immunized with 100 μg KrasG12D DNA vaccine intramuscularly at five-day intervals, administered a total of ten times. Anti-tumor response was indicated by a decreased number of lung nodules and enhanced expression of IFN-γ, IL-12 and IL-4 in splenocyte s [70]. As an attempt to achieve broad-spectrum anti-tumor immunity, researchers designed an mRNA encoding all prevalent RAS variants (mRNA-1521). Groups of mice were immunized with mRNA-1521 or PBS on days 0, 21, and 49 and then inoculated with CT26 colon cancer cells on day 56. Groups receiving vaccine prophylactically had increased infiltration of CD8+ T cells and granzyme B+ CD8+ T cells, and delayed growth of colorectal cancer CT26 in vivo [72]. A therapeutic study of mRNA similarly encoding multiple KRAS variants increased tumor-infiltration of CD8+ T cells and significantly delayed tumor growth in the syngeneic KRAS G12C-expressing lung cancer LL/2-bearing mice [73].

3.5. Summary of Preclinical Models Encoding the Same Antigens

In summary, while no studies to our knowledge have directly compared mRNA and DNA cancer vaccines, both types of vaccines have been able to successfully elicit cellular and humoral responses in murine preclinical and in vitro models, with increased cytokine secretion, and decreased tumor growth reported in most studies. Of note, higher doses of DNA, administered using alternative delivery methods such as gene gun devices or electroporation, were required to generate sufficient anti-tumor responses. This is not surprising given the requirement for DNA to enter the nucleus to be transcribed prior to translation. In contrast, in general, mRNA could be delivered in lower doses without assisted delivery devices, likely due to the protection of mRNA encapsulated in lipid nanoparticles along with the ability for direct translation in the cytoplasm.

4. Monotherapies of mRNA and DNA Vaccines in Human Clinical Trials

Given the breadth and success of preclinical studies utilizing mRNA or DNA vaccines, many of these therapies have been translated to human clinical trials as cancer treatments. While DNA vaccines have largely been explored in clinical trials of breast, cervical, prostate, and melanoma cancers, mRNA vaccines have been predominantly tested in pancreatic and non-small cell lung cancers. Notably, target antigens of DNA vaccines were mostly TAAs, whereas many mRNA vaccines were designed to target patient-specific MANA or neoantigens such as KRAS G12D, G12V, G12C, or G13D mutations. Because of these distinctions, and the fact that very few clinical trials have been conducted using mRNA or DNA encoding the same antigens, we have focused on the clinical trials that have used mRNA or DNA vaccines as monotherapies, with or without adjuvants. Details of these recent clinical trials are summarized in Table 2.

Summary of Human Clinical Trials Using Monotherapy mRNA or DNA Vaccines

As highlighted in Table 2, many trials have been conducted with single-agent mRNA or DNA vaccines, delivered by different routes or methods, at many different doses, and with or without the use of adjuvants. However, within specific cancer types, none of these trials have used an mRNA or DNA vaccine encoding precisely the same antigen(s). Therefore, only general conclusions can be drawn from these studies in a particular disease.
In one trial, in which patients with melanoma received an mRNA vaccine encoding four shared antigens, NY-ESO-1, MAGE-A3, tyrosinase, and TPTE, 3 out of 25 patients exhibited partial responses and 7 of 25 patients had stable disease [94]. Additionally, a patient receiving monotherapy who had a partial clinical response also had a detectable antigen-specific immune responses [94]. While there is not a DNA vaccine encoding these exact four TAAs, three studies have reported on DNA vaccines encoding tyrosinase. These studies have demonstrated safety and tolerability (NCT00698100), antigen-specific immune responses in 6/15 patients [91], and antigen-specific immune responses in 11/26 patients [92].
In general, whether the vector was mRNA or DNA based, many of these clinical trials exploring monotherapy vaccination noted antigen-specific CD4+ or CD8+ T cell responses. However, while these were mostly early phase trials without robust clinical endpoints, only a few of these clinical trials demonstrated any evidence clinical benefit, regardless of whether the vaccine vector was mRNA or DNA. These underwhelming clinical trial results underscore the need for better optimization of the route of delivery, stability of the molecule, targeting to antigen presenting cells, or the need of combination therapies to overcome mechanisms of immune resistance.

5. Methods to Improve mRNA and DNA Vaccines as Monotherapies

Although nucleic acid vaccines are a promising approach for the treatment of cancer, their efficacy as monotherapies, as demonstrated from Table 2, is largely still lacking, potentially due to failure to mount a sufficient immune response, failure to reach antigen presenting cells, or the development of mechanisms of treatment resistance. Therefore, efforts to improve immunogenicity of nucleic acid vaccines by construct design, enhanced delivery methods, or combination therapies are currently underway and described below.

5.1. Methods to Improve mRNA Vaccine Efficacy

Of recent interest in the mRNA field is self-amplifying mRNA. Self-amplifying mRNA encodes viral replication machinery in addition to the gene of interest, resulting in mRNA that will replicate itself in vivo following uptake [110]. These vectors are considered safer than traditional viral vector-mediated vaccines since the virus itself is not introduced and will not replicate [110]. As previously stated, the instability of mRNA causes it to be rapidly degraded, leaving the need for frequent booster immunizations. Self-amplifying mRNA vaccines have the potential to be delivered less frequently than standard mRNA viruses and in lower doses [110]. Preclinical cancer studies using self-amplifying mRNA and LNPs are somewhat limited; however, one study by Maine et al. developed a self-replicating mRNA vaccine platform (SMAART). CT26 tumor-bearing mice were immunized with 10 μg of LNP SMARRT RNAs on days 3 and 17 after tumor injection [111]. SMARRT vaccination induced polyfunctional CD4+ and CD8+ T cells and reduced tumor growth in mice with colorectal cancer. Additionally, rhesus masques with influenza were immunized with SMARRT encoding HA from different strains of influenza on days 0 and 56 at varying doses (1, 10, and 100 ug). The vaccine generated polyfunctional CD4+ and CD8+ T cells in non-human primates at all doses [111]. Another study examined a single immunization with gDE7 mRNA-LNP in an HPV-16-associated tumor model [112]. Mice were inoculated with TC-1 cells and immunized with 30 μg of gDE7 vaccine 3 days later. The study found that a single immunization with a self-replicating mRNA vaccine could control tumor growth in mice [112]. Although research in this area is still ongoing, one self-amplifying mRNA vaccine was approved for protection against COVID-19 in Japan [113].
Another area that is gaining interest in the mRNA field is the manipulation of lipid nanoparticles to increase targeted delivery to certain tissues. One major problem with standard LNP formulations is that many of these LNPs traffic to the liver, or cells that take up RNA/LNP formulations traffic to the liver. While this may be beneficial for diseases of the liver or cancers with liver metastases, many other malignancies may not benefit from liver trafficking. Therefore, recent efforts have focused on LNP modifications to target specific tissues of interest. The selective organ-targeting lipid nanoparticle (SORT) technique involves adding a fifth molecule to the standard four-component LNP to target specific organs such as lung, spleen, and liver [114]. This technology has been used in preclinical cancer vaccines shown in a study by Chen et al. where researchers found a certain LNP, 113-012B, improved uptake in the draining lymph nodes instead of the liver in comparison to ALC-0315 [115]. Mice were subcutaneously injected at the tail base with 113-012B and luciferase mRNA (mLuc) to track in vivo distribution of expressed protein. The intensity of mLuc six hours after injection was higher in draining lymph nodes of mice that received 113-012B compared to other LNPs.
Finally, several groups have modified LNPs to increase their intrinsic ability to act as adjuvants or by directly incorporating adjuvants into the LNP or mRNA-LNP constructs. These methods have been extensively reviewed elsewhere in the past [116,117,118]. However, new studies have demonstrated that modifying the structures or ratios of the non-ionizable lipid components can affect vaccine immunogenicity and intrinsic adjuvanticity [119,120]. While these studies have largely been conducted in infectious disease models, these methods can conceivably be adapted to mRNA vaccines encoding tumor antigens for the treatment of cancer.

5.2. Methods to Improve DNA Vaccine Efficacy

Plasmid DNA vaccines, in general, have low immunogenicity when they are delivered passively by needle-syringe through intradermal, intramuscular, or subcutaneous routes. Therefore, several groups have focused their efforts on improving delivery methods to enhance DNA entry into the cells rather than rely on passive uptake. One of these methods is electroporation, which incorporates an electric pulse immediately after the needle-syringe delivery, allowing for the creation of pores in cell membranes to deliver DNA [121,122]. Several groups have reported increased transfection using electroporation. Broderick et al. found increased humoral immunity in addition to increased protein expression when utilizing a needleless piezoelectric device for electroporation in guinea pigs [123]. Electroporation has also been evaluated for DNA vaccines in human clinical trials; however, a study by Chudley et al. demonstrated similar immunological responses in patients immunized with or without electroporation [124]. Another study by Low et al. showed increased antibody responses in patients receiving a DNA vaccine plus electroporation in comparison to DNA vaccine alone in a clinical trial of prostate cancer [125]. Although some of these studies showed potential positive therapeutic responses, vaccination with electroporation has been shown to cause increased pain at the injection site [122]. This has led to multiple groups focusing their efforts on improving DNA vaccine delivery methods in other ways such as by gene gun or needless devices.
Needleless immunization methods are increasing in popularity due to better tolerability by patients. Gene gun delivery of plasmid DNA was initially implemented in the 1990s to deliver DNA to plant cells, which are notoriously difficult to transfect [126,127]. This technology involves a device, the gene gun, which delivers metal microparticles coated with nucleic acid with high velocity to penetrate the cell membrane, usually propelled by a gas such as helium [128]. Since the initial studies in plants, gene gun delivery has successfully been implemented in mammalian cell and animal studies [129,130,131,132,133,134]. These promising preclinical studies led to human clinical trials, and DNA vaccines delivered by gene gun have proven to be safe, well-tolerated, and immunogenic, with doses of DNA as low as 1 µg being demonstrated to elicit cellular and humoral immunity [135,136]. Similar safety and immunological efficacy results have been shown in human clinical trials using needless fluid-based “jet” devices [137,138]. The only DNA vaccine approved for use in humans is delivered using a needle-free jet device by PharmaJet, indicating the safety, tolerability, and efficacy profiles of needle-free delivery devices are promising for future plasmid DNA delivery trials [139].

6. Combination Therapies Using mRNA or DNA Vaccines

Due to the safety and immunogenicity observed in most small early-phase clinical trials, but lack of profound clinical efficacy, several groups have explored strategies combining nucleic acid vaccines with commonly used therapies or immune-modulating therapies in preclinical studies or human clinical trials. A summary of ongoing human clinical trials is presented in Table 3.

6.1. Combination Therapies with Radiotherapy

In addition to its direct cytotoxic effects, radiation can indirectly induce immunogenic changes in the tumor microenvironment. As radiation-induced DNA damage accumulates in tumor cells, mutations and genomic instability increase, leading to immunogenic cell death. The release of various DAMPs (e.g., HSP, ATP, CRT, and high mobility group box 1) activates macrophages and dendritic cells to enhance antigen presentation. Additionally, the expression of vascular adhesion molecules increases, and inflammatory cytokines and chemokines such as IL-1β, IL-23, and CXCL-10 are locally released. In surviving cancer cells, radiation can activate the cGAS/STING pathway to release type I IFN, further promoting the release of pro-inflammatory molecules, activation of dendritic cells and lymphocytes, and increased susceptibility to CD8+ T cell-mediated cytotoxicity. This cycle ultimately enhances the priming and activation of cytotoxic CD8+ T cells, leading to increased tumor cell killing [154,155,156,157]. However, radiation can also induce immunosuppressive effects, including the recruitment of Tregs and MDSCs as well as the promotion of M2-like macrophages, which may contribute to resistance to both radiation and immunotherapy [158,159]. Therefore, combining radiation with nucleic acid vaccines may enhance anti-tumor efficacy, but may also require careful consideration of factors such as the radiation type, radiation treatment regimen, and the timing of therapies.
In one combination study, an E7-encoding RNA lipoplex (RNA-LPX) was administered to mice inoculated with HPV-related cancers, followed by 12Gy of local external beam radiation [160]. E7 RNA-LPX immunization increased the frequency of intratumoral E7-specific CD8+ T cells, while radiation reduced tumor mass and hypoxia, enhancing the tumor killing capacity of antigen-specific CD8+ T cells. This combination approach delayed tumor growth and prolonged survival of mice bearing TC-1 and C3 tumors. The response depended on the total radiation dose, as both 12Gy and an equivalent biological effective dose of 3 fractions of 6Gy similarly enhanced anti-tumor immunity when combined with E7 RNA-LPX [160].
Another study using a two-component mRNA cancer vaccine, consisting of free and complexed OVA-encoding mRNA, was tested in combination with external beam radiotherapy in mice bearing E.G7-OVA tumors [161]. Two vaccinations were administered per week, starting on the first day of radiation, with a total radiation dose of 6Gy delivered in three equal fractions over three consecutive days. The combination delayed tumor growth and prolonged survival compared to either treatment alone. A total of 5/7 complete responders were noted when rechallenged with parental OVA negative EL4 cells, suggesting the establishment of broad immune protection potentially through antigenic spread. Gene expression analysis revealed that the combination treatment significantly downregulated genes associated with tumor-associated factors and upregulated tumor suppressor-related genes compared to immunization alone [161].
Using a similar ovalbumin-based model, we have explored radiopharmaceutical therapy (RPT) targeting the lipid rafts of tumor cells, 90Y-NM600, as a strategy to improve the efficacy of anti-tumor DNA vaccines. The adoptive transfer of naïve OVA257-264-specific CD8+ T cells before or after low dose 90Y-NM600 administration did not enhance anti-tumor response in mice bearing E.G7-OVA-PD-L1hi tumors. However, when the tumor-specific CD8+ T cells were activated in vivo using OVA-encoding plasmid DNA in combination with 90Y-NM600, tumor growth was significantly delayed. Notably, DNA vaccine was most effective when delivered prior to 90Y-NM600. Our findings suggest that tumor-specific CD8+ T cells need to ideally be present and activated prior to RPT for greater anti-tumor efficacy, suggesting that anti-tumor vaccination should be delivered prior to RPT [162]. While these studies using artificial OVA models cannot be directly compared due to differences in radiation modalities, they serve as proof of concept that the combination of radiation and nucleic acid vaccines enhances anti-tumor efficacy compared to either treatment alone.
Other studies have explored the combination of radiation with HPV-targeting nucleic acid vaccines. In one study, a single dose of 14Gy of external beam radiation was delivered to mice bearing E7-expressing TC-1 tumors, followed by three doses of CRT/E7(detox), a DNA construct encoding HPV16 E7 (modified to abrogate function but retain conformation) and fused to calreticulin (CRT) to enhance antigen presentation. The combination significantly delayed tumor growth and prolonged survival compared to either treatment alone. Additional timing studies revealed that when radiation was given at the time of the second dose of vaccination, significantly higher numbers of E7-specific IFN-γ+ CD8+ T cells were present in the splenocytes and the tumor [163].
Very few clinical trials are currently exploring the use of radiation therapy in combination with nucleic acid vaccines. However, a clinical trial by Papachristofilou et al. examined combination therapy of an mRNA vaccine (300 μg of CV9202, encoding six non-small cell lung cancer antigens) along with 20Gy local radiation over the course of 57 days. Patients that received CV9202 and local radiation had immune responses to the six encoded antigens and increased antigen-specific CD4+ and CD8+ T cells over time [153]. Collectively, these studies using nucleic acid vaccines with radiation therapy suggest that future preclinical and clinical studies should focus on how radiotherapy parameters (e.g., radiation type, dose, schedule) affect combination strategies with vaccination, as well as assess the mechanisms of potential synergy.

6.2. Combination Therapies with Immune Checkpoint Inhibitors (ICI)

Blocking antibodies against the immune checkpoints PD-1, PD-L1, and/or CTLA-4 have been approved by FDA for the treatment of squamous cell head and neck cancer, melanoma, Merkel cell carcinoma, cutaneous squamous cell carcinoma, hepatocellular carcinoma, renal cell carcinoma, cervical cancer, small cell lung cancer, non-small cell lung cancer, triple negative breast cancer, gastric carcinoma, gastroesophageal junction carcinoma, Hodgkin lymphoma, advanced or metastatic urothelial cancer, primary mediastinal B-cell lymphoma, and cancers bearing microsatellite instability or mismatch repair deficiency [164]. However, despite approval for use in these multiple types of cancer, only a small percentage of these patients respond to immune checkpoint inhibitors as monotherapies. Moreover, response to therapy is generally restricted to those cancers with evidence of pre-existing activated tumor-reactive T cells. This suggests they may be best utilized in combination with other therapies, such as cancer vaccines, that can selectively activate tumor-specific T cells.
Multiple groups are studying mRNA vaccines in combination with immune checkpoint inhibitors. Jin et al. created a targeted mRNA vaccine incorporating a cGAS agonist into the LNP formulation to augment immunogenicity. In a LLC-OVA murine model, mice treated with the mRNA vaccine and PD-L1 blockade had reduced tumor growth, likely due to increased infiltration of cell immune cell types into the tumor and increased pro-inflammatory cytokines [165]. Similarly, Wang et al. encapsulated 9 μg of Trp2 mRNA and 1 μg of PD-L1 siRNA into lipid calcium phosphate nanoparticles. Mice inoculated with B16F10 melanoma tumors were treated once, when tumors were ~300 mm3 in size, with this vaccine or mock encoding controls. Mice receiving antigen-specific mRNA in addition to PD-L1 siRNA had reduced tumor growth [166]. Another group has shown similar trends using a PD-1 blocking antibody in combination with an ovalbumin-encoding mRNA-nanostructured lipid nanocarrier (LNC) vaccine in B16-OVA and E.G7-OVA tumor models. Mice were inoculated with tumors (B16-OVA) on day 0 and were immunized with OVA LNC-mRNA (3 μg) on days 3, 6, and 9. Additionally, mice received anti-PD-1 on days 7, 12, and 17. Combination therapy-treated mice exhibited prolonged survival and a greater number of cures than monotherapy-treated control mice [167]. These successful preclinical models have paved the way for several clinical trials combining mRNA vaccines with immune checkpoint inhibitors, and these have shown signs of efficacy in patients with pancreatic ductal adenocarcinoma [148] and melanoma [140]. In particular, a clinical trial targeting individualized melanoma neoantigens (mRNA-4157, V940) given with PD-1 blockade (pembrolizumab), in comparison to PD-1 blockade alone, showed prolonged recurrence-free survival and improved distant metastasis-free survival in patients with resected high-risk melanoma [140]. These positive study results led to “breakthrough therapy” designation by the FDA for this therapy. Other early-phase clinical trials investigating mRNA vaccines in combination with checkpoint inhibitors are underway (Table 3).
Several preclinical studies evaluating DNA vaccines in combination with ICIs have also been conducted. A study by Lopes et al. showed that combination therapy of a DNA vaccine encoding the P815A tumor antigen plus antibodies blocking PD-1 and CTLA-4 prolonged survival and reduced tumor growth in murine P815 mastocytoma [168]. The combination of vaccine and ICI also promoted IFNγ, IL2, and granzyme B production in the tumor microenvironment, suggesting enhanced activation of antigen-specific T cells [168]. Similar preclinical studies in mice have shown improvement in DNA vaccine efficacy when delivered in combination with checkpoint blocking antibodies against PD-1, CTLA-4, LAG-3, or a combination thereof, in mouse models of melanoma [169,170], multiple myeloma [171], prostate cancer [172], glioblastoma [173], human papillomavirus-positive cancers [61], and colorectal cancers [174,175]. As most of these preclinical studies have been conducted using DNA vaccines, it remains unknown whether there are differences between mRNA and DNA vaccines when combined with ICI blockade or whether one approach is more effective than the other when combined with ICI blockade.
As depicted in Table 3, several clinical trials are investigating the use of DNA vaccines in combination with checkpoint inhibitors targeting PD-1, PD-L1, CTLA-4, and/or LAG-3. Most of these trials remain underway. However, two separate trials have been reported using a DNA vaccine encoding a prostate tumor antigen, prostatic acid phosphatase (PAP, pTVG-HP) in patients with early recurrent prostate cancer or more advanced castration-resistant, metastatic prostate cancer (mCRPC) [144,176]. In the trial of patients with mCRPC, vaccination was given either concurrently with PD-1 blockade (pembrolizumab), or PD-1 blockade was administered after completing a 12-week course of vaccination. Objective responses and PSA declines were observed when treatment was given concurrently but not sequentially, suggesting that vaccination is best delivered with concurrent ICI blockade [143]. Concurrent administration of this DNA vaccine and PD-1 blockade (nivolumab) in patients with early recurrent prostate cancer similarly resulted in PSA declines and prolonged stable disease [144].

6.3. Combination Therapies with Toll-like Receptor Agonists

mRNA and DNA vaccines have an inherent ability to act as self-adjuvants due to the ability to activate pattern-recognition receptors (PRRs), including toll-like receptors (TLRs) and RIG-I-like receptors. However, many studies suggest that there may be additional benefits to adding these innate immune sensor ligands as vaccine adjuvants [177]. While lipid-based nanoparticles have been primarily utilized to prevent mRNA degradation and improve delivery to antigen-presenting cells, some have also been shown to activate TLR4 signaling and thereby increase anti-tumor vaccine efficacy in murine tumor models [178,179]. In one of these studies, an optimized C1 nanoparticle was used to encapsulate ovalbumin-encoding mRNA. In vitro studies using bone marrow derived dendritic cells (BMDCs) revealed increased activation of BMDCs as demonstrated by increased protein expression of CD40, CD80, CD86, as well as increased secretion of IL-1β, IL-6, and IL-12p70 in comparison to controls, following treatment with this nanoparticle. However, BMDCs from Tlr4−/− mice failed to show these effects when treated with C1-mRNA, indicating the BMDC activation was dependent on TLR4 signaling [179]. In a study conducted by Lee et al., a TLR1/2 agonist was incorporated into lipid nanoparticles encapsulating an ovalbumin-targeting mRNA vaccine. This resulted in better suppression of E.G7-OVA tumor growth in mice [180]. Similarly, an ovalbumin-specific mRNA vaccine administered with a TLR 7/8 agonist also remarkably improved the expansion of ovalbumin-specific CD8+ T cells and their infiltration into ovalbumin-expressing murine lymphoma and prostate cancer, resulting in significant suppression of tumor growth [181]. Haabeth et al. demonstrated that combination of TLR9 agonist and an OVA-encoding mRNA vaccine increased cytotoxic T cell responses and led to complete regression of A20-OVA lymphoma in mice [182].
Similarly, co-administration of DNA vaccines with a TLR3 or TLR7 agonist showed a greater expansion of HPV E7-specific CD8+ T cells than DNA treatment alone, and the combination also induced significant regression of TC-1 tumors [183]. Vaccination of mice with a DNA vaccine encoding the ligand-binding domain of the androgen receptor, a prostate tumor associated antigen, and combined with TLR3 and TLR9 agonists, resulted in secretion of type 1 IFN from antigen-presenting cells and suppression of TRAMP-C1 prostate tumor growth [184].
Human clinical trials are also beginning to investigate the use of TLR agonists in combination with nucleic acid vaccines. Notably, in cervical cancer, one study is combining a TLR7 agonist (Imiquimod) with a DNA vaccine encoding E6 and E7 (NCT00788164), based on encouraging preclinical data in murine tumor models [185].

6.4. Combination Therapies with Chemotherapy

Chemotherapy is a standard treatment for many cancers, but it is often associated with immunosuppressive effects [186]. However, certain chemotherapeutic agents have been shown to modulate the immune system in ways that can enhance the efficacy of cancer vaccines, creating an opportunity for combinatorial approaches to overcome the limitations of each treatment [187]. Some chemotherapeutic agents, such as cyclophosphamide and doxorubicin, have immunomodulatory effects that make them suitable for combination with mRNA and DNA vaccines [188,189]. These drugs can deplete immunosuppressive cell populations, increase tumor antigen release and/or enhance antigen presentation. For example, low-dose cyclophosphamide, a DNA alkylating agent, has been shown to selectively reduce populations of regulatory T cells (Tregs) and regulate myeloid-derived suppressor cells (MDSCs) infiltration, both populations known to inhibit the immune response in the tumor microenvironment, in a dose-dependent manner [190,191,192]. Cytotoxic chemotherapies also lead to the destruction of tumor cells, which can cause the release of tumor-associated antigens (TAAs) and neoantigens [193]. Tumor cell death induced by chemotherapy also results in the release of damage-associated molecular patterns (DAMPs), which further stimulate immune responses [194]. Certain chemotherapies can upregulate the expression of MHC class I molecules on tumor cells, making them more susceptible to recognition by cytotoxic T lymphocytes (CTLs) [195]. Additionally, chemotherapy can promote dendritic cell maturation, enhancing their ability to present vaccine-derived antigens to T cells [196].
Preclinical studies combining mRNA or DNA vaccines with chemotherapy have demonstrated enhanced antitumor effects compared to either treatment alone [197,198]. In a murine study using E.G7-OVA tumor model, mice were treated with an mRNA vaccine (encoding ovalbumin, 32 μg) alone or in combination with cisplatin (1.2 mg/kg) or docetaxel (25 mg/kg). The results from this study demonstrated improved tumor control in the combination group when using either of the chemotherapeutic agents [161]. The combination approach resulted in significant tumor regression and extended survival compared to monotherapy treatments alone. In another pre-clinical study using a genital tract TC-1 tumor model, Bialkowski et al. demonstrated that the combination of E7-TriMix mRNA vaccine (2.5 μg of each TriMix component and 5 μg of E7 antigen, administered on day 8 and 13) with cisplatin (4 mg/kg, administered on days 8, 9, 15, and 16) caused an increase in E7-specific T cells within tumors, and secretion of IFNγ, compared to the spleen. Additionally, the E7-targeting mRNA vaccine and chemotherapy led to complete regression of cervical tumors in murine models [199].
In preclinical studies of DNA vaccines, combining DNA vaccination with cyclophosphamide has shown promise. In one study, mice were inoculated with 4T1 breast tumor cells on day 0, followed by 50 μg immunizations of a plasmid DNA vaccine encoding fibroblast activating protein-α (CpVR-FAP) into both the right and left tibia muscles on days 2, 9 and 16 post tumor implantation [200]. Cyclophosphamide was administered (50 mg/kg) on days 1, 8 and 15 post tumor implantation. This study demonstrated that chemotherapy enhanced the recruitment of CD8+ T cells and improved the overall efficacy of DNA vaccines in tumor-bearing mice, as evidenced by decreased tumor growth and prolonged survival in the combination therapy group in comparison to either monotherapy alone. Additionally, the proportions of Tregs in the spleen, and immunosuppressive cytokines such as IL-10 and CXCL-12 in the tumor microenvironment, were decreased [200]. Notably, DNA vaccines in combination with cyclophosphamide led to a more durable immune response, without impairing antigen specific immunity resulting from the vaccination. Similarly, Chen et al. also demonstrated synergistic effects of paclitaxel, a chemotherapeutic drug that acts as a mitotic inhibitor, with a chimeric DNA vaccine encoding E7 antigen and connective tissue growth factor, when delivered in low doses frequently over a long period of time [198]. Mice were inoculated with TC-1 tumor cells, and then immunized weekly with 2 μg of pcDNA3-CTGF/E7, delivered by gene gun. Paclitaxel was administered either daily or two times per week at doses of 3, 6, and 25 mg/kg. Mice that received the combination treatment of DNA vaccine plus either 3 mg/kg or 6 mg/kg paclitaxel resulted in smaller tumor volumes and prolonged survival. Additionally, mice that received DNA vaccine in combination with 6 mg/kg paclitaxel had greater numbers of antigen-specific T cells detected in the spleen in comparison to the other combination treatment doses tested [198]. Another group tested the timing of chemotherapy with respect to DNA vaccine. Mice were inoculated with TC-1 cells on day 0, followed by treatment on day 8 with either PBS, cisplatin alone (10 mg/kg, days 8 and 11), DNA vaccine alone (2 μg of CRT/E7 delivered by gene gun with boosters on days 12 and 16), DNA vaccine followed by cisplatin 4d later (DNA on d8, 12, and 16, with cisplatin on days 20 and 23), or cisplastin followed by DNA vaccine 4d later (Cisplatin on d8 and 11, DNA on d15, 19, and 23). The mice that received cisplatin prior to DNA vaccine, in comparison to the other groups tested, had reduced tumor growth and increased numbers of antigen-specific CD8+ cells in their spleens and tumors [163].
In human clinical trials, studies are underway in breast cancer that combine DNA vaccines with chemotherapy. Although these studies are not yet completed, and preclinical data are limited, these initial studies suggest at least the feasibility of combining mRNA or DNA vaccines with chemotherapy. However, several challenges must be considered for these combination therapies. As demonstrated in the studies above, different timing regimens of the DNA or chemotherapy agents, or the dosing strategy, may affect tumor growth, survival, and antigen-specific T-cell generation. Therefore, future studies will need to refine the timing, dosing, and selection of chemotherapeutic agents to maximize synergy.

7. Concluding Remarks and Future Directions

mRNA and DNA vaccines each have advantages and disadvantages. mRNA is rapidly degraded, has transient protein expression, and generally requires cold storage, which may limit its use in certain countries. DNA has the requirement of needing to reach the nucleus of antigen-presenting cells, which may mean higher doses, electroporators, or alternative delivery methods may need to be used to achieve the immunogenicity required to control tumor growth. Unfortunately, there have been no head-to-head studies in preclinical models or human trials using mRNA or DNA vaccines that encode the same antigen, and thus it remains difficult to determine whether one approach is preferred. Due to differences in uptake, cellular processing, recognition by different intracellular nucleic acid sensors, and antigen expression kinetics, it can be speculated that vaccination with mRNA or DNA may produce some differences in the phenotype and magnitude of immune responses generated. This will be an area of future research, to understand the optimal means to improve the anti-tumor efficacy of each approach.
Notwithstanding, vast improvements in technology, delivery methods, and combination therapies have enabled nucleic acid vaccines to be more immunogenic than when they were first implemented for cancer treatments. It is expected that these newer approaches with greater immunogenicity will lead to greater anti-tumor efficacy in clinical trials. Newer gene delivery methods (including needless delivery methods and the use of self-amplifying constructs), the encoding of multiple antigens, the use of different adjuvants, and the use of nucleic acid vaccines in combination with other agents, are mostly still underway and in early stages of development, but have already shown promising results in a small number of clinical trials. In particular, clinical trials incorporating combination treatments of nucleic acid vaccines with checkpoint blockade and/or chemotherapy have already proven successful across many cancer types as evidenced by detectable antigen-specific T cell responses, prolonged recurrence-free survival, prolonged distant metastasis-free survival, and increased tumor infiltration of T cells (Table 3). Many of these studies have been conducted using PD-1, PD-L1, or CTLA-4 blocking antibodies; however, the field of immune checkpoint blockade is rapidly evolving and new therapies targeting other checkpoints, such as LAG-3, VISTA, TIM-3, or TIGIT, or a combination of these, may have more success when used in combination with nucleic acid vaccines. Furthermore, future directions using strategies to overcome the presence and function of regulatory immune cells in the tumor microenvironment may be necessary to further improve the anti-tumor efficacy of nucleic acid vaccines.

Author Contributions

Conceptualization, J.E.M. and D.G.M.; writing—original draft preparation, J.E.M., D.S., D.J., I.R. and D.G.M.; writing—review and editing, J.E.M., D.S., D.J., I.R., K.M.S. and D.G.M.; supervision, D.G.M.; funding acquisition, D.G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported, in part, by the National Institutes of Health (P30 CA014520 and P50 CA269011). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Conflicts of Interest

D.G.M. is a member of the Scientific Advisory Board for PharmJet. The other authors have no relevant potential conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Wang, X.; Zhang, H.; Chen, X. Drug Resistance and Combating Drug Resistance in Cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef] [PubMed]
  3. Ramos, A.; Sadeghi, S.; Tabatabaeian, H. Battling Chemoresistance in Cancer: Root Causes and Strategies to Uproot Them. Int. J. Mol. Sci. 2021, 22, 9451. [Google Scholar] [CrossRef]
  4. Coley, W.B. The Treatment of Inoperable Sarcoma by Bacterial Toxins (the Mixed Toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc. R. Soc. Med. 1910, 3, 1–48. [Google Scholar] [CrossRef]
  5. McCarthy, E.F. The Toxins of William B. Coley and the Treatment of Bone and Soft-Tissue Sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar]
  6. Larocca, C.; Schlom, J. Viral Vector-Based Therapeutic Cancer Vaccines. Cancer J. 2011, 17, 359–371. [Google Scholar] [CrossRef] [PubMed]
  7. Zhou, M.; Tang, Y.; Xu, W.; Hao, X.; Li, Y.; Huang, S.; Xiang, D.; Wu, J. Bacteria-Based Immunotherapy for Cancer: A Systematic Review of Preclinical Studies. Front. Immunol. 2023, 14, 1140463. [Google Scholar] [CrossRef] [PubMed]
  8. Abd-Aziz, N.; Poh, C.L. Development of Peptide-Based Vaccines for Cancer. J. Oncol. 2022, 2022, 9749363. [Google Scholar] [CrossRef]
  9. Tiwari, A.; Alcover, K.; Carpenter, E.; Thomas, K.; Krum, J.; Nissen, A.; Van Decar, S.; Smolinsky, T.; Valdera, F.; Vreeland, T.; et al. Utility of Cell-Based Vaccines as Cancer Therapy: Systematic Review and Meta-Analysis. Hum. Vaccines Immunother. 2024, 20, 2323256. [Google Scholar] [CrossRef]
  10. MacGregor, R.R.; Boyer, J.D.; Ugen, K.E.; Lacy, K.E.; Gluckman, S.J.; Bagarazzi, M.L.; Chattergoon, M.A.; Baine, Y.; Higgins, T.J.; Ciccarelli, R.B.; et al. First Human Trial of a DNA-Based Vaccine for Treatment of Human Immunodeficiency Virus Type 1 Infection: Safety and Host Response. J. Infect. Dis. 1998, 178, 92–100. [Google Scholar] [CrossRef]
  11. Cheng, F.; Wang, Y.; Bai, Y.; Liang, Z.; Mao, Q.; Liu, D.; Wu, X.; Xu, M. Research Advances on the Stability of mRNA Vaccines. Viruses 2023, 15, 668. [Google Scholar] [CrossRef]
  12. Rosa, S.S.; Prazeres, D.M.F.; Azevedo, A.M.; Marques, M.P.C. mRNA Vaccines Manufacturing: Challenges and Bottlenecks. Vaccine 2021, 39, 2190–2200. [Google Scholar] [CrossRef]
  13. Dolgin, E. The Tangled History of mRNA Vaccines. Nature 2021, 597, 318–324. [Google Scholar] [CrossRef]
  14. Abbasi, J. India’s New COVID-19 DNA Vaccine for Adolescents and Adults Is a First. JAMA 2021, 326, 1365. [Google Scholar] [CrossRef]
  15. Lamb, Y.N. BNT162b2 mRNA COVID-19 Vaccine: First Approval. Drugs 2021, 81, 495–501. [Google Scholar] [CrossRef]
  16. Leitner, W.W.; Ying, H.; Restifo, N.P. DNA and RNA-Based Vaccines: Principles, Progress and Prospects. Vaccine 1999, 18, 765–777. [Google Scholar] [CrossRef]
  17. Bolhassani, A.; Javanzad, S.; Saleh, T.; Hashemi, M.; Aghasadeghi, M.R.; Sadat, S.M. Polymeric Nanoparticles: Potent Vectors for Vaccine Delivery Targeting Cancer and Infectious Diseases. Hum. Vaccines Immunother. 2014, 10, 321–332. [Google Scholar] [CrossRef] [PubMed]
  18. Demongeot, J.; Fougère, C. mRNA COVID-19 Vaccines-Facts and Hypotheses on Fragmentation and Encapsulation. Vaccines 2022, 11, 40. [Google Scholar] [CrossRef] [PubMed]
  19. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles—From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef] [PubMed]
  20. Rock, K.L.; Reits, E.; Neefjes, J. Present Yourself! By MHC Class I and MHC Class II Molecules. Trends Immunol. 2016, 37, 724–737. [Google Scholar] [CrossRef]
  21. Del Prete, A.; Salvi, V.; Soriani, A.; Laffranchi, M.; Sozio, F.; Bosisio, D.; Sozzani, S. Dendritic Cell Subsets in Cancer Immunity and Tumor Antigen Sensing. Cell. Mol. Immunol. 2023, 20, 432–447. [Google Scholar] [CrossRef]
  22. Colbert, J.D.; Cruz, F.M.; Rock, K.L. Cross-Presentation of Exogenous Antigens on MHC I Molecules. Curr. Opin. Immunol. 2020, 64, 1–8. [Google Scholar] [CrossRef]
  23. Kozak, M.; Hu, J. DNA Vaccines: Their Formulations, Engineering and Delivery. Vaccines 2024, 12, 71. [Google Scholar] [CrossRef]
  24. Zhou, R.; Geiger, R.C.; Dean, D.A. Intracellular Trafficking of Nucleic Acids. Expert Opin. Drug Deliv. 2004, 1, 127–140. [Google Scholar] [CrossRef] [PubMed]
  25. Al Fayez, N.; Nassar, M.S.; Alshehri, A.A.; Alnefaie, M.K.; Almughem, F.A.; Alshehri, B.Y.; Alawad, A.O.; Tawfik, E.A. Recent Advancement in mRNA Vaccine Development and Applications. Pharmaceutics 2023, 15, 1972. [Google Scholar] [CrossRef] [PubMed]
  26. Ledwith, B.J.; Manam, S.; Troilo, P.J.; Barnum, A.B.; Pauley, C.J.; Griffiths, T.G.; Harper, L.B.; Beare, C.M.; Bagdon, W.J.; Nichols, W.W. Plasmid DNA Vaccines: Investigation of Integration into Host Cellular DNA Following Intramuscular Injection in Mice. Intervirology 2000, 43, 258–272. [Google Scholar] [CrossRef] [PubMed]
  27. Hassett, K.J.; Rajlic, I.L.; Bahl, K.; White, R.; Cowens, K.; Jacquinet, E.; Burke, K.E. mRNA Vaccine Trafficking and Resulting Protein Expression after Intramuscular Administration. Mol. Ther. Nucleic Acids 2024, 35, 102083. [Google Scholar] [CrossRef]
  28. Restifo, N.P.; Ying, H.; Hwang, L.; Leitner, W.W. The Promise of Nucleic Acid Vaccines. Gene Ther. 2000, 7, 89–92. [Google Scholar] [CrossRef]
  29. Zeng, C.; Zhang, C.; Walker, P.G.; Dong, Y. Formulation and Delivery Technologies for mRNA Vaccines. Curr. Top. Microbiol. Immunol. 2022, 440, 71–110. [Google Scholar] [CrossRef]
  30. Bidram, M.; Zhao, Y.; Shebardina, N.G.; Baldin, A.V.; Bazhin, A.V.; Ganjalikhany, M.R.; Zamyatnin, A.A.; Ganjalikhani-Hakemi, M. mRNA-Based Cancer Vaccines: A Therapeutic Strategy for the Treatment of Melanoma Patients. Vaccines 2021, 9, 1060. [Google Scholar] [CrossRef]
  31. Eusébio, D.; Neves, A.R.; Costa, D.; Biswas, S.; Alves, G.; Cui, Z.; Sousa, Â. Methods to Improve the Immunogenicity of Plasmid DNA Vaccines. Drug Discov. Today 2021, 26, 2575–2592. [Google Scholar] [CrossRef]
  32. Colluru, V.T.; McNeel, D.G. B Lymphocytes as Direct Antigen-Presenting Cells for Anti-Tumor DNA Vaccines. Oncotarget 2016, 7, 67901–67918. [Google Scholar] [CrossRef]
  33. Rastogi, I.; McNeel, D.G. B Cells Require Licensing by Dendritic Cells to Serve as Primary Antigen-Presenting Cells for Plasmid DNA. Oncoimmunology 2023, 12, 2212550. [Google Scholar] [CrossRef]
  34. Lee, J.; Ahn, E.; Kissick, H.T.; Ahmed, R. Reinvigorating Exhausted T Cells by Blockade of the PD-1 Pathway. Onco Ther. 2015, 6, 7–17. [Google Scholar] [CrossRef]
  35. Zheng, L.; Bandara, S.R.; Tan, Z.; Leal, C. Lipid Nanoparticle Topology Regulates Endosomal Escape and Delivery of RNA to the Cytoplasm. Proc. Natl. Acad. Sci. USA 2023, 120, e2301067120. [Google Scholar] [CrossRef] [PubMed]
  36. Schlich, M.; Palomba, R.; Costabile, G.; Mizrahy, S.; Pannuzzo, M.; Peer, D.; Decuzzi, P. Cytosolic Delivery of Nucleic Acids: The Case of Ionizable Lipid Nanoparticles. Bioeng. Transl. Med. 2021, 6, e10213. [Google Scholar] [CrossRef] [PubMed]
  37. Hou, X.; Zaks, T.; Langer, R.; Dong, Y. Lipid Nanoparticles for mRNA Delivery. Nat. Rev. Mater. 2021, 6, 1078–1094. [Google Scholar] [CrossRef]
  38. Cui, L.; Renzi, S.; Quagliarini, E.; Digiacomo, L.; Amenitsch, H.; Masuelli, L.; Bei, R.; Ferri, G.; Cardarelli, F.; Wang, J.; et al. Efficient Delivery of DNA Using Lipid Nanoparticles. Pharmaceutics 2022, 14, 1698. [Google Scholar] [CrossRef]
  39. Algarni, A.; Pilkington, E.H.; Suys, E.J.A.; Al-Wassiti, H.; Pouton, C.W.; Truong, N.P. In Vivo Delivery of Plasmid DNA by Lipid Nanoparticles: The Influence of Ionizable Cationic Lipids on Organ-Selective Gene Expression. Biomater. Sci. 2022, 10, 2940–2952. [Google Scholar] [CrossRef] [PubMed]
  40. Wei, L.; Dong, C.; Zhu, W.; Wang, B.-Z. mRNA Vaccine Nanoplatforms and Innate Immunity. Viruses 2024, 16, 120. [Google Scholar] [CrossRef]
  41. Anderson, B.R.; Muramatsu, H.; Jha, B.K.; Silverman, R.H.; Weissman, D.; Kariko, K. Nucleoside Modifications in RNA Limit Activation of 2′-5′-Oligoadenylate Synthetase and Increase Resistance to Cleavage by RNase L. Nucleic Acids Res. 2011, 39, 9329–9338. [Google Scholar] [CrossRef]
  42. Anderson, B.R.; Muramatsu, H.; Nallagatla, S.R.; Bevilacqua, P.C.; Sansing, L.H.; Weissman, D.; Karikó, K. Incorporation of Pseudouridine into mRNA Enhances Translation by Diminishing PKR Activation. Nucleic Acids Res. 2010, 38, 5884–5892. [Google Scholar] [CrossRef]
  43. Alameh, M.-G.; Weissman, D.; Pardi, N. Messenger RNA-Based Vaccines Against Infectious Diseases. In mRNA Vaccines; Yu, D., Petsch, B., Eds.; Current Topics in Microbiology and Immunology; Springer International Publishing: Cham, Switzerland, 2020; Volume 440, pp. 111–145. ISBN 978-3-031-18069-9. [Google Scholar]
  44. Zahid, A.; Ismail, H.; Li, B.; Jin, T. Molecular and Structural Basis of DNA Sensors in Antiviral Innate Immunity. Front. Immunol. 2020, 11, 613039. [Google Scholar] [CrossRef]
  45. Kreiter, S.; Vormehr, M.; Van De Roemer, N.; Diken, M.; Löwer, M.; Diekmann, J.; Boegel, S.; Schrörs, B.; Vascotto, F.; Castle, J.C.; et al. Mutant MHC Class II Epitopes Drive Therapeutic Immune Responses to Cancer. Nature 2015, 520, 692–696. [Google Scholar] [CrossRef]
  46. Deng, Z.; Tian, Y.; Song, J.; An, G.; Yang, P. mRNA Vaccines: The Dawn of a New Era of Cancer Immunotherapy. Front. Immunol. 2022, 13, 887125. [Google Scholar] [CrossRef]
  47. Li, H.; Hu, Y.; Li, J.; He, J.; Yu, G.; Wang, J.; Lin, X. Intranasal Prime-Boost RNA Vaccination Elicits Potent T Cell Response for Lung Cancer Therapy. Signal Transduct. Target. Ther. 2025, 10, 101. [Google Scholar] [CrossRef]
  48. Fan, T.; Xu, C.; Wu, J.; Cai, Y.; Cao, W.; Shen, H.; Zhang, M.; Zhu, H.; Yang, J.; Zhu, Z.; et al. Lipopolyplex-Formulated mRNA Cancer Vaccine Elicits Strong Neoantigen-Specific T Cell Responses and Antitumor Activity. Sci. Adv. 2024, 10, eadn9961. [Google Scholar] [CrossRef] [PubMed]
  49. Yang, B.; Jeang, J.; Yang, A.; Wu, T.C.; Hung, C.-F. DNA Vaccine for Cancer Immunotherapy. Hum. Vaccines Immunother. 2014, 10, 3153–3164. [Google Scholar] [CrossRef] [PubMed]
  50. Buonaguro, L.; Tagliamonte, M. Selecting Target Antigens for Cancer Vaccine Development. Vaccines 2020, 8, 615. [Google Scholar] [CrossRef] [PubMed]
  51. Fan, T.; Zhang, M.; Yang, J.; Zhu, Z.; Cao, W.; Dong, C. Therapeutic Cancer Vaccines: Advancements, Challenges and Prospects. Signal Transduct. Target. Ther. 2023, 8, 450. [Google Scholar] [CrossRef]
  52. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising Targets for Cancer Therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef]
  53. Zhang, Z.; Lu, M.; Qin, Y.; Gao, W.; Tao, L.; Su, W.; Zhong, J. Neoantigen: A New Breakthrough in Tumor Immunotherapy. Front. Immunol. 2021, 12, 672356. [Google Scholar] [CrossRef]
  54. Snyder, L.A.; Goletz, T.J.; Gunn, G.R.; Shi, F.F.; Harris, M.C.; Cochlin, K.; McCauley, C.; McCarthy, S.G.; Branigan, P.J.; Knight, D.M. A MUC1/IL-18 DNA Vaccine Induces Anti-Tumor Immunity and Increased Survival in MUC1 Transgenic Mice. Vaccine 2006, 24, 3340–3352. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, X.; Miao, Y.; Shen, J.; Li, D.; Deng, X.; Yang, C.; Ji, Y.; Dai, Z.; Ma, Y. Unlocking PD-1 Antibody Resistance: The MUC1 DNA Vaccine Augments CD8+ T Cell Infiltration and Attenuates Tumour Suppression. Scand. J. Immunol. 2024, 99, e13356. [Google Scholar] [CrossRef]
  56. Choi, D.-H.; Woo, J.K.; Choi, Y.; Seo, H.-S.; Kim, C.-W. A Novel Chimeric DNA Vaccine: Enhancement of Preventive and Therapeutic Efficacy of DNA Vaccine by Fusion of Mucin 1 to a Heat Shock Protein 70 Gene. Mol. Med. Rep. 2011, 4, 885–890. [Google Scholar] [CrossRef]
  57. Zhang, H.; Liu, C.; Zhang, F.; Geng, F.; Xia, Q.; Lu, Z.; Xu, P.; Xie, Y.; Wu, H.; Yu, B.; et al. MUC1 and Survivin Combination Tumor Gene Vaccine Generates Specific Immune Responses and Anti-Tumor Effects in a Murine Melanoma Model. Vaccine 2016, 34, 2648–2655. [Google Scholar] [CrossRef]
  58. Liu, C.; Xie, Y.; Sun, B.; Geng, F.; Zhang, F.; Guo, Q.; Wu, H.; Yu, B.; Wu, J.; Yu, X.; 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, 63–72. [Google Scholar] [CrossRef] [PubMed]
  59. Lin, X.; Chen, H.; Xie, Y.; Zhou, X.; Wang, Y.; Zhou, J.; Long, S.; Hu, Z.; Zhang, S.; Qiu, W.; et al. Combination of CTLA-4 Blockade with MUC1 mRNA Nanovaccine Induces Enhanced Anti-Tumor CTL Activity by Modulating Tumor Microenvironment of Triple Negative Breast Cancer. Transl. Oncol. 2022, 15, 101298. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, J.; Chen, L.; Zhang, H.; Quan, Y. Enhancing the Antitumour-Specific Immunity of a Lung DNA Vaccine in Vivo by Fusion Expression of MAGE-A3 and Soluble PD-1. Biotechnol. Biotechnol. Equip. 2017, 31, 1064–1069. [Google Scholar] [CrossRef]
  61. Duperret, E.K.; Liu, S.; Paik, M.; Trautz, A.; Stoltz, R.; Liu, X.; Ze, K.; Perales-Puchalt, A.; Reed, C.; Yan, J.; et al. A Designer Cross-Reactive DNA Immunotherapeutic Vaccine That Targets Multiple MAGE-A Family Members Simultaneously for Cancer Therapy. Clin. Cancer Res. 2018, 24, 6015–6027. [Google Scholar] [CrossRef]
  62. Choi, K.; Jeong, H.; Lee, D.H.; Lee, J.W.; Hong, J.-E.; Baek, J.E.; Park, Y.S. Innovative Cancer Immunotherapy with MAGE-A3 mRNA Cancer Vaccines. Cancers 2024, 16, 3428. [Google Scholar] [CrossRef]
  63. Cheng, W.-F.; Hung, C.-F.; Chai, C.-Y.; Hsu, K.-F.; He, L.; Ling, M.; Wu, T.-C. Tumor-Specific Immunity and Antiangiogenesis Generated by a DNA Vaccine Encoding Calreticulin Linked to a Tumor Antigen. J. Clin. Investig. 2001, 108, 669–678. [Google Scholar] [CrossRef]
  64. Farzanehpour, M.; Soleimanjahi, H.; Hassan, Z.M.; Amanzadeh, A.; Ghaemi, A.; Fazeli, M. HSP70 Modified Response against HPV Based Tumor. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 228–234. [Google Scholar] [PubMed]
  65. Trimble, C.; Lin, C.-T.; Hung, C.-F.; Pai, S.; Juang, J.; He, L.; Gillison, M.; Pardoll, D.; Wu, L.; Wu, T.-C. Comparison of the CD8+ T Cell Responses and Antitumor Effects Generated by DNA Vaccine Administered through Gene Gun, Biojector, and Syringe. Vaccine 2003, 21, 4036–4042. [Google Scholar] [CrossRef] [PubMed]
  66. Peng, S.; Ferrall, L.; Gaillard, S.; Wang, C.; Chi, W.-Y.; Huang, C.-H.; Roden, R.B.S.; Wu, T.-C.; Chang, Y.-N.; Hung, C.-F. Development of DNA Vaccine Targeting E6 and E7 Proteins of Human Papillomavirus 16 (HPV16) and HPV18 for Immunotherapy in Combination with Recombinant Vaccinia Boost and PD-1 Antibody. mBio 2021, 12, e03224-20. [Google Scholar] [CrossRef]
  67. Qiu, K.; Duan, X.; Mao, M.; Song, Y.; Rao, Y.; Cheng, D.; Feng, L.; Shao, X.; Jiang, C.; Huang, H.; et al. mRNA-LNP Vaccination-Based Immunotherapy Augments CD8+ T Cell Responses against HPV-Positive Oropharyngeal Cancer. npj Vaccines 2023, 8, 144. [Google Scholar] [CrossRef]
  68. Zhou, K.; Yuzhakov, O.; Behloul, N.; Wang, D.; Bhagat, L.; Chu, D.; Zhang, X.; Cheng, X.; Fan, L.; Huang, X.; et al. HPV16 E6/E7 -Based mRNA Vaccine Is Therapeutic in Mice Bearing Aggressive HPV-Positive Lesions. Front. Immunol. 2023, 14, 1213285. [Google Scholar] [CrossRef]
  69. Wang, J.; Ma, L.; Chen, Y.; Zhou, R.; Wang, Q.; Zhang, T.; Yi, D.; Liu, Q.; Zhang, Y.; Zhang, W.; et al. Immunogenicity and Effectiveness of an mRNA Therapeutic Vaccine for HPV-Related Malignancies. Life Sci. Alliance 2024, 7, e202302448. [Google Scholar] [CrossRef]
  70. Weng, T.-Y.; Yen, M.-C.; Huang, C.-T.; Hung, J.-J.; Chen, Y.-L.; Chen, W.-C.; Wang, C.-Y.; Chang, J.-Y.; Lai, M.-D. DNA Vaccine Elicits an Efficient Antitumor Response by Targeting the Mutant Kras in a Transgenic Mouse Lung Cancer Model. Gene Ther. 2014, 21, 888–896. [Google Scholar] [CrossRef]
  71. Wang, X.; Wang, W.; Zou, S.; Xu, Z.; Cao, D.; Zhang, S.; Wei, M.; Zhan, Q.; Wen, C.; Li, F.; et al. Combination Therapy of KRAS G12V mRNA Vaccine and Pembrolizumab: Clinical Benefit in Patients with Advanced Solid Tumors. Cell Res. 2024, 34, 661–664. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, R.; Liu, W.; Brown, D.M.; Bong, Y.-S.; He, J.; Shen, D.; Wang, C.Y. Abstract 5738: A Pan-Ras mRNA Vaccine Elicits Specific Immune Responses and Inhibits Tumor Growth in the Mouse Model of Colon Cancer. Cancer Res. 2023, 83, 5738. [Google Scholar] [CrossRef]
  73. Shin, S.-H.; Han, Y.; Lim, C.G.; Heo, Y.H.; Jeong, S.; Kim, Y.-Y.; Choi, I.Y. Abstract LB222: An mRNA-Based Cancer Vaccine Multi-Targeting KRAS Mutations Inhibits Tumor Growth by Increasing Immune Response in KRAS Mutant LL/2 Mouse Model. Cancer Res. 2023, 83, LB222. [Google Scholar] [CrossRef]
  74. Li, C.; Ke, F.; Mao, S.; Montemayor, Z.; Traore, M.D.M.; Balsa, A.D.; Djibo, M.; Karekar, N.; Hu, H.; Wen, H.; et al. SARS-CoV-2 B Epitope-Guided Neoantigen NanoVaccines Enhance Tumor-Specific CD4/CD8 T Cell Immunity through B Cell Antigen Presentation. ACS Nano 2025, 19, 7038–7054. [Google Scholar] [CrossRef]
  75. Gao, T.; Cen, Q.; Lei, H. A Review on Development of MUC1-Based Cancer Vaccine. Biomed. Pharmacother. 2020, 132, 110888. [Google Scholar] [CrossRef]
  76. Liu, C.; Lu, Z.; Xie, Y.; Guo, Q.; Geng, F.; Sun, B.; Wu, H.; Yu, B.; Wu, J.; Zhang, H.; et al. Soluble PD-1-Based Vaccine Targeting MUC1 VNTR and Survivin Improves Anti-Tumor Effect. Immunol. Lett. 2018, 200, 33–42. [Google Scholar] [CrossRef]
  77. Esfandiary, A.; Ghafouri-Fard, S. MAGE-A3: An Immunogenic Target Used in Clinical Practice. Immunotherapy 2015, 7, 683–704. [Google Scholar] [CrossRef]
  78. Gjerstorff, M.F.; Andersen, M.H.; Ditzel, H.J. Oncogenic Cancer/Testis Antigens: Prime Candidates for Immunotherapy. Oncotarget 2015, 6, 15772–15787. [Google Scholar] [CrossRef] [PubMed]
  79. Alsalloum, A.; Shevchenko, J.; Sennikov, S. The Melanoma-Associated Antigen Family A (MAGE-A): A Promising Target for Cancer Immunotherapy? Cancers 2023, 15, 1779. [Google Scholar] [CrossRef]
  80. Monie, A.; Tsen, S.-W.D.; Hung, C.-F.; Wu, T.-C. Therapeutic HPV DNA Vaccines. Expert Rev. Vaccines 2009, 8, 1221–1235. [Google Scholar] [CrossRef] [PubMed]
  81. CDC. HPV Vaccination. Available online: https://www.cdc.gov/hpv/vaccines/index.html (accessed on 4 January 2025).
  82. Asimgil, H.; Ertetik, U.; Çevik, N.C.; Ekizce, M.; Doğruöz, A.; Gökalp, M.; Arık-Sever, E.; Istvanffy, R.; Friess, H.; Ceyhan, G.O.; et al. Targeting the Undruggable Oncogenic KRAS: The Dawn of Hope. JCI Insight 2022, 7, e153688. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS Mutation: From Undruggable to Druggable in Cancer. Signal Transduct. Target. Ther. 2021, 6, 386. [Google Scholar] [CrossRef] [PubMed]
  84. Tiriveedhi, V.; Fleming, T.P.; Goedegebuure, P.S.; Naughton, M.; Ma, C.; Lockhart, C.; Gao, F.; Gillanders, W.E.; Mohanakumar, T. Mammaglobin-A cDNA Vaccination of Breast Cancer Patients Induces Antigen-Specific Cytotoxic CD4+ICOShi T Cells. Breast Cancer Res. Treat. 2013, 138, 109–118. [Google Scholar] [CrossRef]
  85. Tiriveedhi, V.; Tucker, N.; Herndon, J.; Li, L.; Sturmoski, M.; Ellis, M.; Ma, C.; Naughton, M.; Lockhart, A.C.; Gao, F.; et al. Safety and Preliminary Evidence of Biologic Efficacy of a Mammaglobin-A DNA Vaccine in Patients with Stable Metastatic Breast Cancer. Clin. Cancer Res. 2014, 20, 5964–5975. [Google Scholar] [CrossRef] [PubMed]
  86. Stanton, S.E.; Wisinski, K.B.; Gwin, W.R.; Coveler, A.; Liao, J.B.; Burkard, M.; Bailey, H.; Kim, K.; Havinghurst, T.; DeShong, K.; et al. Abstract P2-02-02: Phase I Trial of the Safety and Immunogenicity of a Tri-Antigen Vaccine Targeting HER2, IGFBP-2, and IGF-IR in Patients with Non-Metastatic Breast Cancer. Cancer Res. 2023, 83, P2-02-02. [Google Scholar] [CrossRef]
  87. Disis, M.L.; Guthrie, K.A.; Liu, Y.; Coveler, A.L.; Higgins, D.M.; Childs, J.S.; Dang, Y.; Salazar, L.G. Safety and Outcomes of a Plasmid DNA Vaccine Encoding the ERBB2 Intracellular Domain in Patients With Advanced-Stage ERBB2-Positive Breast Cancer: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol. 2023, 9, 71. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, X.; Goedegebuure, S.P.; Chen, M.Y.; Mishra, R.; Zhang, F.; Yu, Y.Y.; Singhal, K.; Li, L.; Gao, F.; Myers, N.B.; et al. Neoantigen DNA Vaccines Are Safe, Feasible, and Induce Neoantigen-Specific Immune Responses in Triple-Negative Breast Cancer Patients. Genome Med. 2024, 16, 131. [Google Scholar] [CrossRef]
  89. Markowitz, J.; Shamblott, M.; Brohl, A.S.; Sarnaik, A.; Eroglu, Z.; Khushalani, N.I.; Chen, P.-L.; De-Aquino, D.B.; Sondak, V.K.; Tarhini, A.A.; et al. IFx-Hu2.0 Phase I First in Human Study for Unresectable Melanoma for an Intralesional “in-Situ Vaccine” Approach. J. Clin. Oncol. 2022, 40, e21542. [Google Scholar] [CrossRef]
  90. Wolchok, J.D.; Yuan, J.; Houghton, A.N.; Gallardo, H.F.; Rasalan, T.S.; Wang, J.; Zhang, Y.; Ranganathan, R.; Chapman, P.B.; Krown, S.E.; et al. Safety and Immunogenicity of Tyrosinase DNA Vaccines in Patients with Melanoma. Mol. Ther. 2007, 15, 2044–2050. [Google Scholar] [CrossRef]
  91. Yuan, J.; Ku, G.Y.; Adamow, M.; Mu, Z.; Tandon, S.; Hannaman, D.; Chapman, P.; Schwartz, G.; Carvajal, R.; Panageas, K.S.; et al. Immunologic Responses to Xenogeneic Tyrosinase DNA Vaccine Administered by Electroporation in Patients with Malignant Melanoma. J. Immunother. Cancer 2013, 1, 20. [Google Scholar] [CrossRef] [PubMed]
  92. Tagawa, S.T.; Lee, P.; Snively, J.; Boswell, W.; Ounpraseuth, S.; Lee, S.; Hickingbottom, B.; Smith, J.; Johnson, D.; Weber, J.S. Phase I Study of Intranodal Delivery of a Plasmid DNA Vaccine for Patients with Stage IV Melanoma. Cancer 2003, 98, 144–154. [Google Scholar] [CrossRef]
  93. Perales, M.-A.; Yuan, J.; Powel, S.; Gallardo, H.F.; Rasalan, T.S.; Gonzalez, C.; Manukian, G.; Wang, J.; Zhang, Y.; Chapman, P.B.; et al. Phase I/II Study of GM-CSF DNA as an Adjuvant for a Multipeptide Cancer Vaccine in Patients With Advanced Melanoma. Mol. Ther. 2008, 16, 2022–2029. [Google Scholar] [CrossRef]
  94. Sahin, U.; Oehm, P.; Derhovanessian, E.; Jabulowsky, R.A.; Vormehr, M.; Gold, M.; Maurus, D.; Schwarck-Kokarakis, D.; Kuhn, A.N.; Omokoko, T.; et al. An RNA Vaccine Drives Immunity in Checkpoint-Inhibitor-Treated Melanoma. Nature 2020, 585, 107–112. [Google Scholar] [CrossRef] [PubMed]
  95. Miller, M.; Sahin, U.; Derhovanessian, E.; Kloke, B.-P.; Simon, P.; Bukur, V.; Albrecht, C.; Paruzynski, A.; Löwer, M.; Kuhn, A.; et al. IVAC MUTANOME: A First-in-Human Phase I Clinical Trial Targeting Individual Mutant Neoantigens for the Treatment of Melanoma. Ann. Oncol. 2017, 28, xi1–xi2. [Google Scholar] [CrossRef]
  96. McNeel, D.G.; Dunphy, E.J.; Davies, J.G.; Frye, T.P.; Johnson, L.E.; Staab, M.J.; Horvath, D.L.; Straus, J.; Alberti, D.; Marnocha, R.; et al. Safety and Immunological Efficacy of a DNA Vaccine Encoding Prostatic Acid Phosphatase in Patients With Stage D0 Prostate Cancer. J. Clin. Oncol. 2009, 27, 4047–4054. [Google Scholar] [CrossRef]
  97. McNeel, D.G.; Becker, J.T.; Eickhoff, J.C.; Johnson, L.E.; Bradley, E.; Pohlkamp, I.; Staab, M.J.; Liu, G.; Wilding, G.; Olson, B.M. Real-Time Immune Monitoring to Guide Plasmid DNA Vaccination Schedule Targeting Prostatic Acid Phosphatase in Patients with Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2014, 20, 3692–3704. [Google Scholar] [CrossRef]
  98. McNeel, D.G.; Eickhoff, J.C.; Johnson, L.E.; Roth, A.R.; Perk, T.G.; Fong, L.; Antonarakis, E.S.; Wargowski, E.; Jeraj, R.; Liu, G. Phase II Trial of a DNA Vaccine Encoding Prostatic Acid Phosphatase (pTVG-HP [MVI-816]) in Patients With Progressive, Nonmetastatic, Castration-Sensitive Prostate Cancer. J. Clin. Oncol. 2019, 37, 3507–3517. [Google Scholar] [CrossRef]
  99. Kyriakopoulos, C.E.; Eickhoff, J.C.; Ferrari, A.C.; Schweizer, M.T.; Wargowski, E.; Olson, B.M.; McNeel, D.G. Multicenter Phase I Trial of a DNA Vaccine Encoding the Androgen Receptor Ligand-Binding Domain (pTVG-AR, MVI-118) in Patients with Metastatic Prostate Cancer. Clin. Cancer Res. 2020, 26, 5162–5171. [Google Scholar] [CrossRef]
  100. Kübler, H.; Scheel, B.; Gnad-Vogt, U.; Miller, K.; Schultze-Seemann, W.; Vom Dorp, F.; Parmiani, G.; Hampel, C.; Wedel, S.; Trojan, L.; et al. Self-Adjuvanted mRNA Vaccination in Advanced Prostate Cancer Patients: A First-in-Man Phase I/IIa Study. J. Immunother. Cancer 2015, 3, 26. [Google Scholar] [CrossRef] [PubMed]
  101. Eriksson, F.; Tötterman, T.; Maltais, A.-K.; Pisa, P.; Yachnin, J. DNA Vaccine Coding for the Rhesus Prostate Specific Antigen Delivered by Intradermal Electroporation in Patients with Relapsed Prostate Cancer. Vaccine 2013, 31, 3843–3848. [Google Scholar] [CrossRef] [PubMed]
  102. Trimble, C.L.; Morrow, M.P.; Kraynyak, K.A.; Shen, X.; Dallas, M.; Yan, J.; Edwards, L.; Parker, R.L.; Denny, L.; Giffear, M.; 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, 2078–2088. [Google Scholar] [CrossRef]
  103. Kim, T.J.; Jin, H.-T.; Hur, S.-Y.; Yang, H.G.; Seo, Y.B.; Hong, S.R.; Lee, C.-W.; Kim, S.; Woo, J.-W.; Park, K.S.; et al. Clearance of Persistent HPV Infection and Cervical Lesion by Therapeutic DNA Vaccine in CIN3 Patients. Nat. Commun. 2014, 5, 5317. [Google Scholar] [CrossRef]
  104. Hasan, Y.; Spiotto, M.T.; Furtado, L.V.; Tergas, A.I.; Lee, N.K.; Brooks, R.A.; McCall, A.R.; Golden, D.W.; Jolly, S.; Fleming, G.F.; et al. A Phase 1/2A Trial of Synthetic DNA Vaccine Immunotherapy Targeting HPV-16 and -18 after Chemoradiation for Cervical Cancer. J. Clin. Oncol. 2018, 36, 5525. [Google Scholar] [CrossRef]
  105. Hillemanns, P.; Denecke, A.; Woelber, L.; Böhmer, G.; Jentschke, M.; Schjetne, K.W.; Bruins Slot, K.M.H.; Fredriksen, A.B. A Therapeutic Antigen-Presenting Cell-Targeting DNA Vaccine VB10.16 in HPV16-Positive High-Grade Cervical Intraepithelial Neoplasia: Results from a Phase I/IIa Trial. Clin. Cancer Res. 2022, 28, 4885–4892. [Google Scholar] [CrossRef] [PubMed]
  106. Trimble, C.L.; Peng, S.; Kos, F.; Gravitt, P.; Viscidi, R.; Sugar, E.; Pardoll, D.; Wu, T.C. A Phase I Trial of a Human Papillomavirus DNA Vaccine for HPV16+ Cervical Intraepithelial Neoplasia 2/3. Clin. Cancer Res. 2009, 15, 361–367. [Google Scholar] [CrossRef] [PubMed]
  107. Weber, J.S.; Vogelzang, N.J.; Ernstoff, M.S.; Goodman, O.B.; Cranmer, L.D.; Marshall, J.L.; Miles, S.; Rosario, D.; Diamond, D.C.; Qiu, Z.; et al. A Phase 1 Study of a Vaccine Targeting Preferentially Expressed Antigen in Melanoma and Prostate-Specific Membrane Antigen in Patients With Advanced Solid Tumors. J. Immunother. 2011, 34, 556–567. [Google Scholar] [CrossRef]
  108. Sebastian, M.; Schröder, A.; Scheel, B.; Hong, H.S.; Muth, A.; von Boehmer, L.; Zippelius, A.; Mayer, F.; Reck, M.; Atanackovic, D.; et al. A Phase I/IIa Study of the mRNA-Based Cancer Immunotherapy CV9201 in Patients with Stage IIIB/IV Non-Small Cell Lung Cancer. Cancer Immunol. Immunother. 2019, 68, 799–812. [Google Scholar] [CrossRef] [PubMed]
  109. Wick, W.; Wick, A.; Sahm, F.; Riehl, D.; Von Deimling, A.; Bendszus, M.; Kickingereder, P.; Beckhove, P.; Schmitz-Winnenthal, F.H.; Jungk, C.; et al. VXM01 Phase I Study in Patients with Progressive Glioblastoma: Final Results. J. Clin. Oncol. 2018, 36, 2017. [Google Scholar] [CrossRef]
  110. Bloom, K.; Van Den Berg, F.; Arbuthnot, P. Self-Amplifying RNA Vaccines for Infectious Diseases. Gene Ther. 2021, 28, 117–129. [Google Scholar] [CrossRef]
  111. Maine, C.J.; Richard, G.; Spasova, D.S.; Miyake-Stoner, S.J.; Sparks, J.; Moise, L.; Sullivan, R.P.; Garijo, O.; Choz, M.; Crouse, J.M.; et al. Self-Replicating RNAs Drive Protective Anti-Tumor T Cell Responses to Neoantigen Vaccine Targets in a Combinatorial Approach. Mol. Ther. 2021, 29, 1186–1198. [Google Scholar] [CrossRef]
  112. Ramos Da Silva, J.; Bitencourt Rodrigues, K.; Formoso Pelegrin, G.; Silva Sales, N.; Muramatsu, H.; De Oliveira Silva, M.; Porchia, B.F.M.M.; Moreno, A.C.R.; Aps, L.R.M.M.; Venceslau-Carvalho, A.A.; et al. Single Immunizations of Self-Amplifying or Non-Replicating mRNA-LNP Vaccines Control HPV-Associated Tumors in Mice. Sci. Transl. Med. 2023, 15, eabn3464. [Google Scholar] [CrossRef]
  113. First Self-Amplifying mRNA Vaccine Approved. Nat. Biotechnol. 2024, 42, 4. [CrossRef]
  114. Wang, X.; Liu, S.; Sun, Y.; Yu, X.; Lee, S.M.; Cheng, Q.; Wei, T.; Gong, J.; Robinson, J.; Zhang, D.; et al. Preparation of Selective Organ-Targeting (SORT) Lipid Nanoparticles (LNPs) Using Multiple Technical Methods for Tissue-Specific mRNA Delivery. Nat. Protoc. 2023, 18, 265–291. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, J.; Ye, Z.; Huang, C.; Qiu, M.; Song, D.; Li, Y.; Xu, Q. Lipid Nanoparticle-Mediated Lymph Node–Targeting Delivery of mRNA Cancer Vaccine Elicits Robust CD8+ T Cell Response. Proc. Natl. Acad. Sci. USA 2022, 119, e2207841119. [Google Scholar] [CrossRef] [PubMed]
  116. Chilumula, S.; Hanchate, P.; Patri, S.V.; Marepally, S. Influence of Structural Modifications in Synthetic Vectors of Lipid Adjuvants on mRNA Vaccine Delivery. Biomater. Sci. 2025, 13, 4952–4969. [Google Scholar] [CrossRef]
  117. Chatzikleanthous, D.; O’Hagan, D.T.; Adamo, R. Lipid-Based Nanoparticles for Delivery of Vaccine Adjuvants and Antigens: Toward Multicomponent Vaccines. Mol. Pharm. 2021, 18, 2867–2888. [Google Scholar] [CrossRef]
  118. Mochida, Y.; Uchida, S. mRNA Vaccine Designs for Optimal Adjuvanticity and Delivery. RNA Biol. 2024, 21, 422–448. [Google Scholar] [CrossRef]
  119. Kawaguchi, Y.; Kimura, M.; Karaki, T.; Tanaka, H.; Ono, C.; Ishida, T.; Matsuura, Y.; Hirai, T.; Akita, H.; Shimizu, T.; et al. Modulating Immunogenicity and Reactogenicity in mRNA-Lipid Nanoparticle Vaccines through Lipid Component Optimization. ACS Nano 2025, 19, 27977–28001. [Google Scholar] [CrossRef]
  120. Vadovics, M.; Zhao, W.; Daley, E.F.; Lam, K.; Daly, O.; Rashid, K.; Lee, H.R.; Schreiner, P.; Lundgreen, K.A.; Gaudette, B.T.; et al. Tailoring the Adjuvanticity of Lipid Nanoparticles by PEG Lipid Ratio and Phospholipid Modifications. Nat. Nanotechnol. 2025, 1–11. [Google Scholar] [CrossRef]
  121. Sardesai, N.Y.; Weiner, D.B. Electroporation Delivery of DNA Vaccines: Prospects for Success. Curr. Opin. Immunol. 2011, 23, 421–429. [Google Scholar] [CrossRef] [PubMed]
  122. Kisakov, D.N.; Belyakov, I.M.; Kisakova, L.A.; Yakovlev, V.A.; Tigeeva, E.V.; Karpenko, L.I. The Use of Electroporation to Deliver DNA-Based Vaccines. Expert Rev. Vaccines 2024, 23, 102–123. [Google Scholar] [CrossRef]
  123. Broderick, K.E.; Kardos, T.; McCoy, J.R.; Fons, M.P.; Kemmerrer, S.; Sardesai, N.Y. Piezoelectric Permeabilization of Mammalian Dermal Tissue for in Vivo DNA Delivery Leads to Enhanced Protein Expression and Increased Immunogenicity. Hum. Vaccines 2011, 7, 22–28. [Google Scholar] [CrossRef] [PubMed]
  124. Chudley, L.; McCann, K.; Mander, A.; Tjelle, T.; Campos-Perez, J.; Godeseth, R.; Creak, A.; Dobbyn, J.; Johnson, B.; Bass, P.; et al. DNA Fusion-Gene Vaccination in Patients with Prostate Cancer Induces High-Frequency CD8+ T-Cell Responses and Increases PSA Doubling Time. Cancer Immunol. Immunother. 2012, 61, 2161–2170. [Google Scholar] [CrossRef]
  125. Low, L.; Mander, A.; McCann, K.; Dearnaley, D.; Tjelle, T.; Mathiesen, I.; Stevenson, F.; Ottensmeier, C.H. DNA Vaccination with Electroporation Induces Increased Antibody Responses in Patients with Prostate Cancer. Hum. Gene Ther. 2009, 20, 1269–1278. [Google Scholar] [CrossRef] [PubMed]
  126. Klein, T.M.; Wolf, E.D.; Wu, R.; Sanford, J.C. High-Velocity Microprojectiles for Delivering Nucleic Acids into Living Cells. Nature 1987, 327, 70–73. [Google Scholar] [CrossRef]
  127. Sanford, J.C.; Klein, T.M.; Wolf, E.D.; Allen, N. Delivery of substances into cells and tissues using a particle bombardment process. Part. Sci. Technol. 1987, 5, 27–37. [Google Scholar] [CrossRef]
  128. Uchida, M.; Li, X.W.; Mertens, P.; Alpar, H.O. Transfection by Particle Bombardment: Delivery of Plasmid DNA into Mammalian Cells Using Gene Gun. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2009, 1790, 754–764. [Google Scholar] [CrossRef]
  129. Zelenin, A.V.; Titomirov, A.V.; Kolesnikov, V.A. Genetic Transformation of Mouse Cultured Cells with the Help of High-velocity Mechanical DNA Injection. FEBS Lett. 1989, 244, 65–67. [Google Scholar] [CrossRef] [PubMed]
  130. Yang, N.S.; Burkholder, J.; Roberts, B.; Martinell, B.; McCabe, D. In Vivo and in Vitro Gene Transfer to Mammalian Somatic Cells by Particle Bombardment. Proc. Natl. Acad. Sci. USA 1990, 87, 9568–9572. [Google Scholar] [CrossRef]
  131. Williams, R.S.; Johnston, S.A.; Riedy, M.; DeVit, M.J.; McElligott, S.G.; Sanford, J.C. Introduction of Foreign Genes into Tissues of Living Mice by DNA-Coated Microprojectiles. Proc. Natl. Acad. Sci. USA 1991, 88, 2726–2730. [Google Scholar] [CrossRef]
  132. Cheng, L.; Ziegelhoffer, P.R.; Yang, N.S. In Vivo Promoter Activity and Transgene Expression in Mammalian Somatic Tissues Evaluated by Using Particle Bombardment. Proc. Natl. Acad. Sci. USA 1993, 90, 4455–4459. [Google Scholar] [CrossRef]
  133. Sato, H.; Hattori, S.; Kawamoto, S.; Kudoh, I.; Hayashi, A.; Yamamoto, I.; Yoshinari, M.; Minami, M.; Kanno, H. In Vivo Gene Gun-Mediated DNA Delivery into Rodent Brain Tissue. Biochem. Biophys. Res. Commun. 2000, 270, 163–170. [Google Scholar] [CrossRef]
  134. Wang, S.; Zhang, C.; Zhang, L.; Li, J.; Huang, Z.; Lu, S. The Relative Immunogenicity of DNA Vaccines Delivered by the Intramuscular Needle Injection, Electroporation and Gene Gun Methods. Vaccine 2008, 26, 2100–2110. [Google Scholar] [CrossRef] [PubMed]
  135. Roy, M.J.; Wu, M.S.; Barr, L.J.; Fuller, J.T.; Tussey, L.G.; Speller, S.; Culp, J.; Burkholder, J.K.; Swain, W.F.; Dixon, R.M.; et al. Induction of Antigen-Specific CD8+ T Cells, T Helper Cells, and Protective Levels of Antibody in Humans by Particle-Mediated Administration of a Hepatitis B Virus DNA Vaccine. Vaccine 2000, 19, 764–778. [Google Scholar] [CrossRef]
  136. Jones, S.; Evans, K.; McElwaine-Johnn, H.; Sharpe, M.; Oxford, J.; Lambkin-Williams, R.; Mant, T.; Nolan, A.; Zambon, M.; Ellis, J.; et al. DNA Vaccination Protects against an Influenza Challenge in a Double-Blind Randomised Placebo-Controlled Phase 1b Clinical Trial. Vaccine 2009, 27, 2506–2512. [Google Scholar] [CrossRef]
  137. Hooper, J.W.; Kwilas, S.A.; Josleyn, M.; Norris, S.; Hutter, J.N.; Hamer, M.; Livezey, J.; Paolino, K.; Twomey, P.; Koren, M.; et al. Phase 1 Clinical Trial of Hantaan and Puumala Virus DNA Vaccines Delivered by Needle-Free Injection. npj Vaccines 2024, 9, 221. [Google Scholar] [CrossRef]
  138. Khobragade, A.; Bhate, S.; Ramaiah, V.; Deshpande, S.; Giri, K.; Phophle, H.; Supe, P.; Godara, I.; Revanna, R.; Nagarkar, R.; et al. Efficacy, Safety, and Immunogenicity of the DNA SARS-CoV-2 Vaccine (ZyCoV-D): The Interim Efficacy Results of a Phase 3, Randomised, Double-Blind, Placebo-Controlled Study in India. Lancet 2022, 399, 1313–1321. [Google Scholar] [CrossRef]
  139. Sheridan, C. First COVID-19 DNA Vaccine Approved, Others in Hot Pursuit. Nat. Biotechnol. 2021, 39, 1479–1482. [Google Scholar] [CrossRef] [PubMed]
  140. Weber, J.S.; Carlino, M.S.; Khattak, A.; Meniawy, T.; Ansstas, G.; Taylor, M.H.; Kim, K.B.; McKean, M.; Long, G.V.; Sullivan, R.J.; et al. Individualised Neoantigen Therapy mRNA-4157 (V940) plus Pembrolizumab versus Pembrolizumab Monotherapy in Resected Melanoma (KEYNOTE-942): A Randomised, Phase 2b Study. Lancet 2024, 403, 632–644. [Google Scholar] [CrossRef]
  141. Pham, T.; Pereira, L.; Roth, S.; Galletta, L.; Link, E.; Akhurst, T.; Solomon, B.; Michael, M.; Darcy, P.; Sampurno, S.; et al. First-in-Human Phase I Clinical Trial of a Combined Immune Modulatory Approach Using TetMYB Vaccine and Anti-PD-1 Antibody in Patients with Advanced Solid Cancer Including Colorectal or Adenoid Cystic Carcinoma: The MYPHISMO Study Protocol (NCT03287427). Contemp. Clin. Trials Commun. 2019, 16, 100409. [Google Scholar] [CrossRef] [PubMed]
  142. Wargowski, E.; Johnson, L.E.; Eickhoff, J.C.; Delmastro, L.; Staab, M.J.; Liu, G.; McNeel, D.G. Prime-Boost Vaccination Targeting Prostatic Acid Phosphatase (PAP) in Patients with Metastatic Castration-Resistant Prostate Cancer (mCRPC) Using Sipuleucel-T and a DNA Vaccine. J. Immunother. Cancer 2018, 6, 21. [Google Scholar] [CrossRef]
  143. McNeel, D.G.; Eickhoff, J.C.; Wargowski, E.; Zahm, C.; Staab, M.J.; Straus, J.; Liu, G. Concurrent, but Not Sequential, PD-1 Blockade with a DNA Vaccine Elicits Anti-Tumor Responses in Patients with Metastatic, Castration-Resistant Prostate Cancer. Oncotarget 2018, 9, 25586–25596. [Google Scholar] [CrossRef]
  144. McNeel, D.G.; Emamekhoo, H.; Eickhoff, J.C.; Kyriakopoulos, C.E.; Wargowski, E.; Tonelli, T.P.; Johnson, L.E.; Liu, G. Phase 2 Trial of a DNA Vaccine (pTVG-HP) and Nivolumab in Patients with Castration-Sensitive Non-Metastatic (M0) Prostate Cancer. J. Immunother. Cancer 2023, 11, e008067. [Google Scholar] [CrossRef]
  145. Madan, R.A.; Bilusic, M.; Stein, M.N.; Donahue, R.N.; Arlen, P.M.; Karzai, F.; Plimack, E.; Wong, Y.-N.; Geynisman, D.M.; Zibelman, M.; et al. Flutamide With or Without PROSTVAC in Non-Metastatic Castration Resistant (M0) Prostate Cancer. Oncologist 2023, 28, 642–e561. [Google Scholar] [CrossRef]
  146. Reardon, D.A.; Brem, S.; Desai, A.S.; Bagley, S.J.; Kurz, S.C.; De La Fuente, M.I.; Nagpal, S.; Welch, M.R.; Hormigo, A.; Forsyth, P.A.J.; et al. Intramuscular (IM) INO-5401 + INO-9012 with Electroporation (EP) in Combination with Cemiplimab (REGN2810) in Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2022, 40, 2004. [Google Scholar] [CrossRef]
  147. Wick, W.; Wick, A.; Chinot, O.; Sahm, F.; Von Deimling, A.; Jungk, C.; Mansour, M.; Podola, L.; Lubenau, H.; Platten, M. KS05.6.A Oral DNA Vaccination Targeting VEGFR2 Combined with the Anti-PD-L1 Antibody Avelumab in Patients with Progressive Glioblastoma-Final Results. NCT03750071. Neuro-Oncology 2022, 24, ii6. [Google Scholar] [CrossRef]
  148. Rojas, L.A.; Sethna, Z.; Soares, K.C.; Olcese, C.; Pang, N.; Patterson, E.; Lihm, J.; Ceglia, N.; Guasp, P.; Chu, A.; et al. Personalized RNA Neoantigen Vaccines Stimulate T Cells in Pancreatic Cancer. Nature 2023, 618, 144–150. [Google Scholar] [CrossRef] [PubMed]
  149. Sethna, Z.; Guasp, P.; Reiche, C.; Milighetti, M.; Ceglia, N.; Patterson, E.; Lihm, J.; Payne, G.; Lyudovyk, O.; Rojas, L.A.; et al. RNA Neoantigen Vaccines Prime Long-Lived CD8+ T Cells in Pancreatic Cancer. Nature 2025, 639, 1042–1051. [Google Scholar] [CrossRef] [PubMed]
  150. Maldonado, L.; Teague, J.E.; Morrow, M.P.; Jotova, I.; Wu, T.C.; Wang, C.; Desmarais, C.; Boyer, J.D.; Tycko, B.; Robins, H.S.; et al. Intramuscular Therapeutic Vaccination Targeting HPV16 Induces T Cell Responses That Localize in Mucosal Lesions. Sci. Transl. Med. 2014, 6, 221ra13. [Google Scholar] [CrossRef]
  151. Alvarez, R.D.; Huh, W.K.; Bae, S.; Lamb, L.S.; Conner, M.G.; Boyer, J.; Wang, C.; Hung, C.-F.; Sauter, E.; Paradis, M.; et al. A Pilot Study of pNGVL4a-CRT/E7(Detox) for the Treatment of Patients with HPV16 + Cervical Intraepithelial Neoplasia 2/3 (CIN2/3). Gynecol. Oncol. 2016, 140, 245–252. [Google Scholar] [CrossRef]
  152. Krasny, S.; Baranau, Y.; Polyakov, S.; Zharkova, E.; Streltsova, O.; Filimonava, A.; Siarheyeva, V.; Kazlouskaya, S.; Khorau, A.; Gabai, V.; et al. Clinical Efficacy of Plasmid Encoding P62/SQSTM1 (Elenagen) in Combination with Gemcitabine in Patients with Platinum-Resistant Ovarian Cancer: A Randomized Controlled Trial. Front. Oncol. 2024, 14, 1343023. [Google Scholar] [CrossRef]
  153. Papachristofilou, A.; Hipp, M.M.; Klinkhardt, U.; Früh, M.; Sebastian, M.; Weiss, C.; Pless, M.; Cathomas, R.; Hilbe, W.; Pall, G.; et al. Phase Ib Evaluation of a Self-Adjuvanted Protamine Formulated mRNA-Based Active Cancer Immunotherapy, BI1361849 (CV9202), Combined with Local Radiation Treatment in Patients with Stage IV Non-Small Cell Lung Cancer. J. Immunother. Cancer 2019, 7, 38. [Google Scholar] [CrossRef] [PubMed]
  154. Yang, C.; Liang, Y.; Liu, N.; Sun, M. Role of the cGAS-STING Pathway in Radiotherapy for Non-Small Cell Lung Cancer. Radiat. Oncol. 2023, 18, 145. [Google Scholar] [CrossRef]
  155. Anderson, R.; Vallis, K. Radionuclide Therapy and Immunomodulation. In Nuclear Medicine and Immunology; Harsini, S., Alavi, A., Rezaei, N., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 249–266. ISBN 978-3-030-81260-7. [Google Scholar]
  156. Carlson, P.M.; Morris, Z.S. Translational Development and Testing of Theranostics in Combination with Immunotherapies. In Nuclear Medicine and Immunology; Harsini, S., Alavi, A., Rezaei, N., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 267–280. ISBN 978-3-030-81260-7. [Google Scholar]
  157. Sun, Q.; Li, J.; Ding, Z.; Liu, Z. Radiopharmaceuticals Heat Anti-Tumor Immunity. Theranostics 2023, 13, 767–786. [Google Scholar] [CrossRef]
  158. Carvalho, H.d.A.; Villar, R.C. Radiotherapy and Immune Response: The Systemic Effects of a Local Treatment. Clinics 2018, 73, e557s. [Google Scholar] [CrossRef]
  159. Muralidhar, A.; Hernandez, R.; Morris, Z.S.; Comas Rojas, H.; Bio Idrissou, M.; Weichert, J.P.; McNeel, D.G. Myeloid-Derived Suppressor Cells Attenuate the Antitumor Efficacy of Radiopharmaceutical Therapy Using90 Y-NM600 in Combination with Androgen Deprivation Therapy in Murine Prostate Tumors. J. Immunother. Cancer 2024, 12, e008760. [Google Scholar] [CrossRef]
  160. Salomon, N.; Selmi, A.; Grunwitz, C.; Kong, A.; Stanganello, E.; Neumaier, J.; Petschenka, J.; Diken, M.; Kreiter, S.; Türeci, Ö.; et al. Local Radiotherapy and E7 RNA-LPX Vaccination Show Enhanced Therapeutic Efficacy in Preclinical Models of HPV16+ Cancer. Cancer Immunol. Immunother. 2022, 71, 1975–1988. [Google Scholar] [CrossRef]
  161. Fotin-Mleczek, M.; Zanzinger, K.; Heidenreich, R.; Lorenz, C.; Thess, A.; Duchardt, K.M.; Kallen, K.-J. Highly Potent mRNA Based Cancer Vaccines Represent an Attractive Platform for Combination Therapies Supporting an Improved Therapeutic Effect. J. Gene Med. 2012, 14, 428–439. [Google Scholar] [CrossRef] [PubMed]
  162. Shim, D.; Hernandez, R.; Rojas, H.C.; Idrissou, M.B.; McNeel, D.G. 785 Combining Tumor-Specific Vaccination with Radiopharmaceutical Therapy (RPT) Enhances Anti-Tumor Response in OVA-Expressing Tumor Model. J. Immunother. Cancer 2024, 12, A889. [Google Scholar] [CrossRef]
  163. Tseng, C.-W.; Hung, C.-F.; Alvarez, R.D.; Trimble, C.; Huh, W.K.; Kim, D.; Chuang, C.-M.; Lin, C.-T.; Tsai, Y.-C.; He, L.; et al. Pretreatment with Cisplatin Enhances E7-Specific CD8+ T-Cell-Mediated Antitumor Immunity Induced by DNA Vaccination. Clin. Cancer Res. 2008, 14, 3185–3192. [Google Scholar] [CrossRef]
  164. Vaddepally, R.K.; Kharel, P.; Pandey, R.; Garje, R.; Chandra, A.B. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers 2020, 12, 738. [Google Scholar] [CrossRef]
  165. Jin, C.; Zhang, Y.; Li, B.; Gao, T.; Wang, B.; Hua, P. Robust Anti-Tumor Immunity through the Integration of Targeted Lipid Nanoparticle-Based mRNA Nanovaccines with PD-1/PD-L1 Blockade. Mater. Today Bio. 2024, 27, 101136. [Google Scholar] [CrossRef]
  166. Wang, Y.; Zhang, L.; Xu, Z.; Miao, L.; Huang, L. mRNA Vaccine with Antigen-Specific Checkpoint Blockade Induces an Enhanced Immune Response against Established Melanoma. Mol. Ther. 2018, 26, 420–434. [Google Scholar] [CrossRef]
  167. Fournier, C.; Mercey-Ressejac, M.; Derangère, V.; Al Kadi, A.; Rageot, D.; Charrat, C.; Leroy, A.; Vollaire, J.; Josserand, V.; Escudé, M.; et al. Nanostructured Lipid Carriers Based mRNA Vaccine Leads to a T Cell–Inflamed Tumour Microenvironment Favourable for Improving PD-1/PD-L1 Blocking Therapy and Long-Term Immunity in a Cold Tumour Model. eBioMedicine 2025, 112, 105543. [Google Scholar] [CrossRef] [PubMed]
  168. Lopes, A.; Vanvarenberg, K.; Kos, Š.; Lucas, S.; Colau, D.; Van Den Eynde, B.; Préat, V.; Vandermeulen, G. Combination of Immune Checkpoint Blockade with DNA Cancer Vaccine Induces Potent Antitumor Immunity against P815 Mastocytoma. Sci. Rep. 2018, 8, 15732. [Google Scholar] [CrossRef] [PubMed]
  169. Kos, S.; Lopes, A.; Preat, V.; Cemazar, M.; Lampreht Tratar, U.; Ucakar, B.; Vanvarenberg, K.; Sersa, G.; Vandermeulen, G. Intradermal DNA Vaccination Combined with Dual CTLA-4 and PD-1 Blockade Provides Robust Tumor Immunity in Murine Melanoma. PLoS ONE 2019, 14, e0217762. [Google Scholar] [CrossRef]
  170. Gregor, P.D.; Wolchok, J.D.; Ferrone, C.R.; Buchinshky, H.; Guevara-Patiño, J.A.; Perales, M.-A.; Mortazavi, F.; Bacich, D.; Heston, W.; Latouche, J.-B.; et al. CTLA-4 Blockade in Combination with Xenogeneic DNA Vaccines Enhances T-Cell Responses, Tumor Immunity and Autoimmunity to Self Antigens in Animal and Cellular Model Systems. Vaccine 2004, 22, 1700–1708. [Google Scholar] [CrossRef]
  171. Neeli, P.; Maza, P.A.M.A.; Chai, D.; Zhao, D.; Hoi, X.P.; Chan, K.S.; Young, K.H.; Li, Y. DNA Vaccines against GPRC5D Synergize with PD-1 Blockade to Treat Multiple Myeloma. npj Vaccines 2024, 9, 180. [Google Scholar] [CrossRef] [PubMed]
  172. Zahm, C.D.; Moseman, J.E.; Delmastro, L.E.; G Mcneel, D. PD-1 and LAG-3 Blockade Improve Anti-Tumor Vaccine Efficacy. Oncoimmunology 2021, 10, 1912892. [Google Scholar] [CrossRef]
  173. Bausart, M.; Vanvarenberg, K.; Ucakar, B.; Lopes, A.; Vandermeulen, G.; Malfanti, A.; Préat, V. Combination of DNA Vaccine and Immune Checkpoint Blockades Improves the Immune Response in an Orthotopic Unresectable Glioblastoma Model. Pharmaceutics 2022, 14, 1025. [Google Scholar] [CrossRef]
  174. Viborg, N.; Pavlidis, M.A.; Barrio-Calvo, M.; Friis, S.; Trolle, T.; Sørensen, A.B.; Thygesen, C.B.; Kofoed, S.V.; Kleine-Kohlbrecher, D.; Hadrup, S.R.; et al. DNA Based Neoepitope Vaccination Induces Tumor Control in Syngeneic Mouse Models. npj Vaccines 2023, 8, 77. [Google Scholar] [CrossRef]
  175. Salvatori, E.; Lione, L.; Compagnone, M.; Pinto, E.; Conforti, A.; Ciliberto, G.; Aurisicchio, L.; Palombo, F. Neoantigen Cancer Vaccine Augments Anti-CTLA-4 Efficacy. npj Vaccines 2022, 7, 15. [Google Scholar] [CrossRef]
  176. McNeel, D.G.; Eickhoff, J.C.; Wargowski, E.; Johnson, L.E.; Kyriakopoulos, C.E.; Emamekhoo, H.; Lang, J.M.; Brennan, M.J.; Liu, G. Phase 2 Trial of T-Cell Activation Using MVI-816 and Pembrolizumab in Patients with Metastatic, Castration-Resistant Prostate Cancer (mCRPC). J. Immunother. Cancer 2022, 10, e004198. [Google Scholar] [CrossRef]
  177. Jeon, D.; Hill, E.; McNeel, D.G. Toll-like Receptor Agonists as Cancer Vaccine Adjuvants. Hum. Vaccines Immunother. 2024, 20, 2297453. [Google Scholar] [CrossRef]
  178. Verbeke, R.; Lentacker, I.; Wayteck, L.; Breckpot, K.; Van Bockstal, M.; Descamps, B.; Vanhove, C.; De Smedt, S.C.; Dewitte, H. Co-Delivery of Nucleoside-Modified mRNA and TLR Agonists for Cancer Immunotherapy: Restoring the Immunogenicity of Immunosilent mRNA. J. Control. Release 2017, 266, 287–300. [Google Scholar] [CrossRef] [PubMed]
  179. Zhang, H.; You, X.; Wang, X.; Cui, L.; Wang, Z.; Xu, F.; Li, M.; Yang, Z.; Liu, J.; Huang, P.; et al. Delivery of mRNA Vaccine with a Lipid-like Material Potentiates Antitumor Efficacy through Toll-like Receptor 4 Signaling. Proc. Natl. Acad. Sci. USA 2021, 118, e2005191118. [Google Scholar] [CrossRef] [PubMed]
  180. Lee, K.; Kim, S.Y.; Seo, Y.; Kim, M.H.; Chang, J.; Lee, H. Adjuvant Incorporated Lipid Nanoparticles for Enhanced mRNA-Mediated Cancer Immunotherapy. Biomater. Sci. 2020, 8, 1101–1105. [Google Scholar] [CrossRef] [PubMed]
  181. Islam, M.A.; Rice, J.; Reesor, E.; Zope, H.; Tao, W.; Lim, M.; Ding, J.; Chen, Y.; Aduluso, D.; Zetter, B.R.; et al. Adjuvant-Pulsed mRNA Vaccine Nanoparticle for Immunoprophylactic and Therapeutic Tumor Suppression in Mice. Biomaterials 2021, 266, 120431. [Google Scholar] [CrossRef]
  182. Haabeth, O.A.W.; Blake, T.R.; McKinlay, C.J.; Waymouth, R.M.; Wender, P.A.; Levy, R. mRNA Vaccination with Charge-Altering Releasable Transporters Elicits Human T Cell Responses and Cures Established Tumors in Mice. Proc. Natl. Acad. Sci. USA 2018, 115, E9153–E9161. [Google Scholar] [CrossRef]
  183. Sajadian, A.; Tabarraei, A.; Soleimanjahi, H.; Fotouhi, F.; Gorji, A.; Ghaemi, A. Comparing the Effect of Toll-like Receptor Agonist Adjuvants on the Efficiency of a DNA Vaccine. Arch. Virol. 2014, 159, 1951–1960. [Google Scholar] [CrossRef]
  184. Jeon, D.; McNeel, D.G. Toll-like Receptor Agonist Combinations Augment Mouse T-Cell Anti-Tumor Immunity via IL-12- and Interferon ß-Mediated Suppression of Immune Checkpoint Receptor Expression. Oncoimmunology 2022, 11, 2054758. [Google Scholar] [CrossRef]
  185. Soong, R.-S.; Song, L.; Trieu, J.; Knoff, J.; He, L.; Tsai, Y.-C.; Huh, W.; Chang, Y.-N.; Cheng, W.-F.; Roden, R.B.S.; et al. Toll-like Receptor Agonist Imiquimod Facilitates Antigen-Specific CD8+ T-Cell Accumulation in the Genital Tract Leading to Tumor Control through IFNγ. Clin. Cancer Res. 2014, 20, 5456–5467. [Google Scholar] [CrossRef]
  186. Anand, U.; Dey, A.; Chandel, A.K.S.; Sanyal, R.; Mishra, A.; Pandey, D.K.; De Falco, V.; Upadhyay, A.; Kandimalla, R.; Chaudhary, A.; et al. Cancer Chemotherapy and beyond: Current Status, Drug Candidates, Associated Risks and Progress in Targeted Therapeutics. Genes Dis. 2023, 10, 1367–1401. [Google Scholar] [CrossRef] [PubMed]
  187. Weir, G.M.; Liwski, R.S.; Mansour, M. Immune Modulation by Chemotherapy or Immunotherapy to Enhance Cancer Vaccines. Cancers 2011, 3, 3114–3142. [Google Scholar] [CrossRef]
  188. Kerr, M.D.; McBride, D.A.; Chumber, A.K.; Shah, N.J. Combining Therapeutic Vaccines with Chemo- and Immunotherapies in the Treatment of Cancer. Expert Opin. Drug Discov. 2021, 16, 89–99. [Google Scholar] [CrossRef]
  189. Emens, L.A.; Middleton, G. The Interplay of Immunotherapy and Chemotherapy: Harnessing Potential Synergies. Cancer Immunol. Res. 2015, 3, 436–443. [Google Scholar] [CrossRef]
  190. Scurr, M.; Pembroke, T.; Bloom, A.; Roberts, D.; Thomson, A.; Smart, K.; Bridgeman, H.; Adams, R.; Brewster, A.; Jones, R.; et al. Low-Dose Cyclophosphamide Induces Antitumor T-Cell Responses, Which Associate with Survival in Metastatic Colorectal Cancer. Clin. Cancer Res. 2017, 23, 6771–6780. [Google Scholar] [CrossRef]
  191. Becker, J.C.; Schrama, D. The Dark Side of Cyclophosphamide: Cyclophosphamide-Mediated Ablation of Regulatory T Cells. J. Investig. Dermatol. 2013, 133, 1462–1465. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, Z.; Till, B.; Gao, Q. Chemotherapeutic Agent-Mediated Elimination of Myeloid-Derived Suppressor Cells. Oncoimmunology 2017, 6, e1331807. [Google Scholar] [CrossRef]
  193. Ma, W.; Pham, B.; Li, T. Cancer Neoantigens as Potential Targets for Immunotherapy. Clin. Exp. Metastasis 2022, 39, 51–60. [Google Scholar] [CrossRef]
  194. Wang, Y.J.; Fletcher, R.; Yu, J.; Zhang, L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes Dis. 2018, 5, 194–203. [Google Scholar] [CrossRef]
  195. Wan, S.; Pestka, S.; Jubin, R.G.; Lyu, Y.L.; Tsai, Y.-C.; Liu, L.F. Chemotherapeutics and Radiation Stimulate MHC Class I Expression through Elevated Interferon-Beta Signaling in Breast Cancer Cells. PLoS ONE 2012, 7, e32542. [Google Scholar] [CrossRef] [PubMed]
  196. Müller, P.; Martin, K.; Theurich, S.; von Bergwelt-Baildon, M.; Zippelius, A. Cancer Chemotherapy Agents Target Intratumoral Dendritic Cells to Potentiate Antitumor Immunity. Oncoimmunology 2014, 3, e954460. [Google Scholar] [CrossRef]
  197. Kang, T.H.; Mao, C.-P.; Lee, S.Y.; Chen, A.; Lee, J.-H.; Kim, T.W.; Alvarez, R.D.; Roden, R.B.S.; Pardoll, D.; Hung, C.-F.; et al. Chemotherapy Acts as an Adjuvant to Convert the Tumor Microenvironment into a Highly Permissive State for Vaccination-Induced Antitumor Immunity. Cancer Res. 2013, 73, 2493–2504. [Google Scholar] [CrossRef]
  198. Chen, C.-A.; Ho, C.-M.; Chang, M.-C.; Sun, W.-Z.; Chen, Y.-L.; Chiang, Y.-C.; Syu, M.-H.; Hsieh, C.-Y.; Cheng, W.-F. Metronomic Chemotherapy Enhances Antitumor Effects of Cancer Vaccine by Depleting Regulatory T Lymphocytes and Inhibiting Tumor Angiogenesis. Mol. Ther. 2010, 18, 1233–1243. [Google Scholar] [CrossRef]
  199. Bialkowski, L.; van Weijnen, A.; Van der Jeught, K.; Renmans, D.; Daszkiewicz, L.; Heirman, C.; Stangé, G.; Breckpot, K.; Aerts, J.L.; Thielemans, K. Intralymphatic mRNA Vaccine Induces CD8 T-Cell Responses That Inhibit the Growth of Mucosally Located Tumours. Sci. Rep. 2016, 6, 22509. [Google Scholar] [CrossRef]
  200. Xia, Q.; Geng, F.; Zhang, F.-F.; Liu, C.-L.; Xu, P.; Lu, Z.-Z.; Xie, Y.; Sun, B.; Wu, H.; Yu, B.; et al. Cyclophosphamide Enhances Anti-Tumor Effects of a Fibroblast Activation Protein α-Based DNA Vaccine in Tumor-Bearing Mice with Murine Breast Carcinoma. Immunopharmacol. Immunotoxicol. 2017, 39, 37–44. [Google Scholar] [CrossRef] [PubMed]
Table 1. mRNA and DNA vaccines in preclinical animal models encoding the same antigens.
Table 1. mRNA and DNA vaccines in preclinical animal models encoding the same antigens.
AntigenVaccine (Vector)Nucleic AcidRoute and DoseResponseTumor ModelReference
MUC1pMUC1
(Vical 1055)
DNAID: 3 doses, biweekly (10 μg)
Did not induce tumor protection alone
Co-administration of plasmid encoding murine IL-18 was needed to induce tumor protection, memory response, and epitope spreading
Colorectal (MC38/MUC1+)[54]
pcDNA3.1-MUC1 (pcDNA3.1(-))DNAIM: 2 doses, biweekly (20 μg)
Increased IFN-γ and granzyme B production
in vitro and in vivo cytotoxicity
Colorectal
(CT26/MUC1+)
[55]
MUC1-HSP70 (pcDNA3)DNAID: 3 doses, weekly (100 μg)
Increased CD8+ proliferation
Increased IFN-γ secretion
Increased in vitro and in vivo cytotoxicity
Melanoma
(B16/MUC1+)
[56]
VR-MSDNAIM: 3 doses, biweekly (100 μg)
Increased in vitro cytotoxicity
Delayed tumor growth
Melanoma (B16/MUC1+survivin+)[57]
CpDV-IL2-sPD1/MS (pBluescriptIISK (+))DNAIM: 2 doses, biweekly (100 μg)
Increased In vitro cytotoxicity
Increased antibody production
Increased IFN-γ secretion
Promoted tumor-infiltration of CD8+ T cells while reducing Treg infiltration
Delayed tumor growth and prolonged survival
Colorectal (CT26/MUC1+ survivin+)
Lung (Lewis/MUC1+ survivin+)
[58]
MUC1 mRNA (encapsulated in lipid/calcium/phosphate nanoparticle)mRNASC: 2 doses, weekly (10 μg)
Increased IFN-γ secretion
Increased in vivo MUC1-specific CD8+ T cell-dependent killing of tumor cells
Triple negative breast (4T1)[59]
MAGE-ApVAX1-MAGE-A3
(pVAX1)
DNAIM: 3 doses, every week (30 μg) with electroporation
Delayed tumor growth
Increased IFN-γ response by splenocytes
Enhanced Th1 (IgG2A, IFN-γ) and Th2 (IgG1 and IL-4) responses
Augmented anti-tumor responses when plasmid fused encode to soluble PD-1
Lung carcinoma (LLC)[60]
MAGE-A (encoding A1, A2, A3, A5, A6, A8)DNAIM: 3 doses, biweekly for in vitro; 4 doses, weekly for in vivo studies (25 μg) followed by electroporation
Increased IFN-γ-secreting splenocytes
Increased frequency of IFN-γ+, CD107a+ T-bet+ IFN-γ+ and TNFα+ CD8+ T cells
Delayed tumor growth
Decreased tumor invasion depth in the skin
Increased tumor-infiltration of CD44+ PD-1+ CD8+ T cells
Inducible model of melanoma (Tyr::CreER;BrafCa/+;Ptenlox/lox)[61]
MAGE-A3 mRNA-containing LNP (DMKD/PS)mRNAIM: 2 doses, biweekly (20 μg)
Delayed tumor growth and prolonged survival
Enhanced humoral response (IgG2a/b)
Did not change IFN-γ or IL-4 in sera
In vitro co-culture of splenocytes from with tumor increased IL-4 but not IFN-γ
Colorectal (CT26)[62]
HPVHPV16 E7/CRT (pDNA3)DNAGene gun: 4 doses, weekly (16 μg)
CD8+ T cell-dependent tumor protection
Reduced pulmonary tumor nodules and tumor growth
Decreased microvessel density in lung tumor nodules
HPV-associated lung (TC-1)[63]
pcDNA/E7, pcDNA/HSP70DNAIM: 2 doses, biweekly
pcDNA/E7 delayed tumor growth compared to PBS and empty vector
Co-administration of pcDNA/HSP70 further delayed tumor growth, enhanced lymphocyte proliferation, in vitro cytolytic activity of splenocytes of immunized mice
HPV-associated lung (TC-1)[64]
pNGVL4a/Sig/E7(detox)/HSP70DNAIM (syringe needle and needle-free jet injection device): 2 doses, weekly (50 μg)
Gene gun: 2 doses, weekly (2 μg)
Increased the number of IFN-γ-secreting E7-specific CD8+ T cells in the spleen (gene gun generated the highest number while requiring the least dose)
Complete in vivo protection
HPV-associated lung (TC-1)[65]
pBI-11 (codon-optimized pNGVL4a/Sig/E7(detox)/HSP70)DNAIM: 3 doses, 3-day interval (25 μg)
Increased number of HPV-specific IFN-y-secreting CD8+ T cells (splenocytes and PBMC)
Delayed tumor growth
HPV-associated lung (TC-1)[66]
HPV16 E7 mRNA-LNPmRNASC, IV: 3 doses, 5-day interval (10 μg)
Expansion of HPV-specific CD8+ T cells in the spleen and tumor
Spleen CD8 +T cells clustered in the IFN-induced group
Tumor-infiltrating CD8+ T cells clustered in effector memory and exhausted groups
Clonal expansion in tumor-infiltrated effectors and exhausted cell subclusters
HPV-associated oropharyngeal squamous cell carcinoma (mEERL)[67]
HPV16 E6/E7 mRNA-LNPmRNAIM or SC: 3 doses, weekly (10 μg or 20 μg);
1 or 2 doses, 10 day interval (3 μg or 10 μg)
Tumor regression after the first week of treatment
Formation of immunological memory (rejection tumor upon re-challenge)
Responses were not dose-dependent and did not differ by injection route
HPV-associated cervical (C3.43)[68]
mHTV-02mRNAIM, IT, SC, IV, ID: 3 doses, weekly (6.25 μg, 12.5 μg, 25 μg)
Increased secretion of IFN-γ and IL-2
Increased HPV-specific IFN-γ+ and TNF-α+ CD8+ T cells
Increased tumor-infiltration of IFN-γ+ Granzyme B+ CD8+ T cells
Increased memory CD8+ and CD4+
Tumor regression and prolonged survival
IM or IT significantly delayed tumor growth and prolonged survival, while IV did not affect growth or survival
Dose-responses observed for naïve immunized mice, but therapeutic doses as little as 6.25 µg were effective at controlling tumor growth
HPV-associated lung (TC-1)[69]
KRAS G12KrasG12DN17DNAGene gun: 10 doses, 5-day interval (10 μg)
and/or
IM: 10 doses, 5-day interval (100 μg)
Decreased number of lung nodule
Increased expression of IFN-γ, IL-12, IL-4 in the splenocytes of vaccinated mice
Combination of GG and IM immunization methods significantly increased CD8+ T cell infiltration into the lungs
Bi-transgenic inducible, spontaneous lung adenocarcinoma (CCSP-rtTA/Tet-Op-K-Ras4G12D)[70]
KRAS G12V mRNAmRNA3 doses, 5-day interval (10 μg)
Increased the percentage of KRAS G12V-specific CD8+ T cells in the spleen
Increased IFNγ secretion by splenocytes
Delayed the growth of G12V-bearing tumors
Melanoma
(G12V and HLA-A11:01 overexpressing B16F10)
[71]
mRNA-1521mRNAImmunized on day 0, 21, and 49
Tested for prophylactic use only
Delayed tumor growth
Combination with anti-PD-1 further delayed tumor growth
IgG response
Greater infiltration of CD8+ and Granzyme B+ CD8+
Colon carcinoma (CT26)[72]
mRNA encoding transcripts for multiple KRAS mutant agmRNANot specified
Increased tumor infiltration of CD8+ T cells (slight 0.5-fold increase in CD44+ CD8+)
Decreased Treg infiltration
Attenuated tumor growth by 37%
Lung (LL/2)[73]
NeoantigensBSARSTKPC-mRNAVaxmRNA20 µg mRNA; immunized every 4 days for three doses, then every 3 days for two doses; total of 5 doses
Significantly delayed tumor growth
Increased germinal center B cells
Increased TFH cells, antigen-specific CD4 T cells and antigen-specific CD8+ T cells
Increased Granzyme B-producing CD4+ and CD8+ T cells
Pancreatic (KPC 6422)[74]
Table 2. Monotherapy mRNA and DNA vaccines in human clinical trials.
Table 2. Monotherapy mRNA and DNA vaccines in human clinical trials.
Cancer TypeTarget AntigenNucleic AcidRoute and DoseAdjuvantResults or Recruitment StatusReference
BreastMammaglobin-A (Mam-A)DNAIM followed by EP, administered weeks 1, 4, and 8 by jet delivery device; unknown doseNoneIncreased IFNγ-producing CD4+ T cells by vaccinationNCT00807781,
[84]
NoneSignificant increase in the number of antigen-specific CD8+ T cellsNCT00807781,
[85]
IGFBP-2, HER2, and IGF-1RDNAID, administered every 28 days; 150 µg, 300 µg, or 600 µgGM-CSFElevated Th1 responses by vaccination with grade 1/2 adverse events; 300 µg dose elicited persistent immune responses 6mo after vaccine administrationNCT02780401,
[86]
HER2 intracellular domain (ICD)DNAID, administered once a month for three months; 10 µg, 100 µg, or 500 µg GM-CSFElicited robust HER2-specific type 1 T-cell responses; Stronger immune responses in 100 µg and 500 µg doses in comparison to 10 µg doseNCT00436254,
[87]
Several neoantigensDNAIM with electroporation; 4 mg administered 3 times, every 28 daysNoneIncreased IFNγ secreting CD8+ T cellsNCT02348320,
[88]
CD105, Yb-1, SOX2, CDH3, and MDM2DNAIV, administered once a month for 3 months, followed by boosters at 6 and 12 monthsGM-CSFRecruitingNCT05455658
ID, administered once a month for 3 months, with potential booster dosesGM-CSFActive, not recruitingNCT02157051
MelanomaEmm55DNAIntralesion; administered one time in up to 3 lesions; 0.1 mg/lesionNoneSeveral patients exhibited stable diseaseNCT03655756,
[89]
gp75 (TYRP1)DNAIM; administered every three weeks for 5 vaccinations; 0.1 mg to 8 mg dose-escalation studyNoneCompleted (results not provided)NCT00034554
TyrosinaseDNAIM; administered every three weeks; 100 µg, 500 µg, or 1500 µg doses NoneDemonstrated vaccine safety and feasibility; CD8+ T cell responses detected in 7 patients; no correlation of immune response with respect to dose or treatment armNCT00698100,
[90]
DNAIM by electroporation; administered every three weeks up to five immunizations; 0.2 mg, 0.5 mg, or 1.5 mg per injectionNoneImmune responses specific to tyrosinase were observed in 6/15 patients; immune responses were only detected in 1.5 mg cohortNCT00471133,
[91]
DNAIN continuously over 96 h every 14 days; 200 µg, 400 µg, or 800 µgNoneImmune responses detected in 11/26 patientsNCT00023647,
[92]
GM-CSF DNA plus Tyrosinase and gp100 peptidesDNASC; administered monthly for a total of three immunizations; 100 µg, 400 µg, or 800 µg of GM-CSF-encoding DNA followed by SC delivery of peptides to the same site on days 5 or 6 post DNA administrationGM-CSFCD8+ T cell responses against melanoma peptides detected in 42% of patients, no correlations between dose and T cell responses were observedNCT00580060,
[93]
Tyrosinase and Melan-A (MART-1)DNAIN; continuous infusion on days 1–4 with boosters every 14 d up to four courses; 500 µg, 1000 µg, or 1500 µgNoneCompleted (results not provided)NCT00033228
NY-ESO-1, MAGE-A3, tyrosinase, and TPTEmRNAIV; administered in 6 injections within 43 days or 8 injections within 64 days; dose escalation study with doses varying from 14.4 µg to 400 µg total RNANoneInduced strong CD4+ and CD8+ T cell immunity against the vaccine antigens; T cells responses were not dose-dependentNCT02410733,
[94]
gp100DNAID or IM; once every 4 weeks up to four dosesIL-2Completed (results not provided)NCT00019448
DNAIM; human gp100 in weeks 1, 4, and 7, followed by mouse gp100 in weeks 10, 13, and 16, or the reverse sequence; dose escalation studyNoneCompleted (results not provided)NCT00104845
Mouse tyrosinase-related protein 2 (TYRP2)DNAIM; every 3 weeks for 6 injections; doses of 500 µg, 2000 µg, 4000 µg, or 8000 µgNoneCompleted (results not provided)NCT00680589
Melanoma-associated antigensmRNAID GM-CSFCompleted (results not provided)NCT00204516
Naked TAAs of melanomamRNAIN; 8 immunizations over 43–51 days; total RNA doses of 100 µg, 200 µg, 600 µg, or 1200 µg containing RBL001 plus RBL002 NoneCompleted (results not provided)NCT01684241
Neoantigens (IVAC MUTANOME1)mRNAINNoneInduced antigen-specific immune responsesNCT02035956,
[95]
ProstatePAP (pTVG-HP)DNAID; 6 immunizations administered biweekly; 100 µg, 500 µg, or 1500 µg dosesGM-CSFPAP-specific T-cell responses detected; responses were not correlated with treatment dosesNCT00582140,
[96]
DNAID; 100 µg; administered biweekly for 6 immunizations, then every 3 months until progression (Arm 1) or biweekly for 6 doses then administered every 2 weeks, 4 weeks, or 3 months as determined by immune responsesGM-CSFNCT00849121,
[97]
DNAID; 100 µg; administered biweekly for 6 immunizations, then every 3 months for two years total treatmentGM-CSFNCT01341652,
[98]
Androgen receptor ligand-binding domain (pTVG-AR)DNAID; administered in biweekly immunizations for 6 doses, then every 3 months up to 12 months (schedule 1) or weeks 0, 2, 12, 14, 24, 26, 36, 38, 48, and 50 (schedule 2)With or without GM-CSFTh1-type immunity to the AR LBD detected; immunological responses in patients treated biweekly for 6 doses followed by boosters every 3 months (schedule 1) were superior to patients treated on the intermittent biweekly schedule (schedule 2)NCT02411786,
[99]
TAAs including PSA, PSCA, PSMA, STEAP1mRNAID; Phase I: 256 µg, 640 µg, 1280 µg; Phase II: 1280 µg; up to 5 immunizationsNoneInduction of antigen-specific CD4+ and CD8+ T cellsEudraCT 2008-003967-37,
[100]
Prostate Specific Antigen (PSA)DNAID with electroporation; administered every 4 weeks for 5 months; doses of 50 µg to 1600 µgNonePSA specific T cell detectedNCT00859729,
[101]
CervicalHPV E6 and E7 (VGX-3100)DNAIM with electroporation; 6 mg; administered weeks 0,4, and 12 NoneBetter histological regression observed compared to placeboNCT01304524,
[102]
HPV E6 and E7 (GX-188E)DNAElectroporation; 1 mg, 2 mg, or 4 mg; administered weeks 0, 4, and 12NoneHPV-specific CD8 T-cell response detected; responses independent of doseNCT01634503,
[103]
DNAElectroporation; 1 mg, 2 mg, or 4 mg dosesNoneUnknown statusNCT02100085
DNAIM with electroporation; weeks 0, 4, and 12; doses of 1 mg or 4 mgNoneCompleted, Results not postedNCT02139267
DNAElectroporation; 1 mg and 4 mg; administered three timesNoneUnknown statusNCT02411019
DNAIM with electroporation; 1 mg administered at weeks 0,4, and 12NoneUnknown statusNCT02596243
HPV E6 and E7DNAIM with electroporation; administered at weeks 0,4,8,12 NoneAntibody responses against HPV oncoproteins detected; antigen specific T cells detectedNCT02172911,
[104]
DNAIM with electroporation; 1 mg, 4 mg or 8 mg dosesNoneRecruitingNCT06276101
HPV E6/E7 fusion proteinDNAIM; 3 immunizations of 3 mg each; administered on weeks 0, 3, and 6 (cohort 1) or weeks 0, 4, and 8 (cohort 2)NoneHPV-specific T-cell responses detected; Stronger T cell responses observed in cohort 1NCT02529930,
[105]
HPV E6, E7, and L2 linked with calreticulinDNAIM with electroporation; 0.3 mg, 1 mg, or 3 mg dosesNoneNot yet recruitingNCT04131413
HPV E7DNAIM; 3 doses at 1 month intervals; 0.5 mg, 1 mg, or 3 mg dosesNoneHPV E7-specific T-cell responses detected; regression instances detected in 3 mg cohortNCT00121173,
[106]
HPV specific antigens (RG002)mRNAIMNoneRecruitingNCT06273553
Multiple advanced solid tumorsNY-ESO-1DNAID by particle-mediated epidermal delivery; administered weeks 1, 5, and 9; 4 µg or 8 µgNoneNY-ESO-1-specific CD4+ and CD8+ T cells detected; no difference in immune responses between 4 µg and 8 µg cohortsNCT00199849
Preferentially expressed antigen in melanoma (PRAME) and prostate-specific membrane antigen (PSMA)DNAID; administered on days 29 and 32 for up to 9 months; 30 µg or 300 µgNoneIncreased PRAME-specific and PSMA-specific T cell responses; no difference in immune responses between low and high peptide dose cohortsNCT00423254,
[107]
 CD105, Yb-1, SOX2, CDH3, and MDM2DNAID; administered on day 14 for 3, 21-day cyclesGM-CSFActive, not recruitingNCT05242965
 NY-ESO-1, MAGE-C1, MAGE-C2, Survivin, and TPBGmRNAID; administered at weeks 1, 3, 7, 11, or at 1, 2, 3, 5, and 7; 400 µg, 800 µg, or 1600 µg totalNoneInduction of immune response against five antigens; dosage did not cause significant differences in immune responseNCT00923312,
[108]
Non-Small Cell LungNeoantigensmRNASCNoneRecruitingNCT03908671
 VEGFR-2DNAOral; administered on days 1, 3, 5, and 7, then 4-weekly single doses every 4 weeks; 106 or 107 CFUNoneVEGFR-2 specific T cell response detectedNCT02718443,
[109]
 IE-1, pp65, gBDNAIM with electroporation; 8 mgNoneCompleted (Results not provided)NCT05698199
 whole tumor mRNA, pp65, LAMPmRNAIV; administered every 2 weeks for 3 doses following radiation, then 12 doses monthlyNoneRecruitingNCT04573140
 pp65mRNAIV; 3 doses administered before or after tumor biopsy/resectionNoneRecruitingNCT06389591
 NeoantigensDNAIM; administered on days 1, 22, and 43 of cycle 1 and then day 1 of each subsequent cyclePlasmid encoded IL-12Active, not recruitingNCT04015700
GlioblastomaTumor-associated antigensmRNAIM; administered on days 1, 8, 15, 29, 43, 57, and 71; 6 µg, 12 µg, 25 µg, 50 µg, 100 µgNoneActive, not recruitingNCT05938387
Table 3. mRNA and DNA vaccines administered as parts of combination therapies in human clinical trials.
Table 3. mRNA and DNA vaccines administered as parts of combination therapies in human clinical trials.
Cancer TypeTarget AntigenNucleic AcidRoute and DoseCombination TherapyResults or Recruitment StatusReference
BreastMammaglobin-A (Mam-A)DNAIM; administered every 28 days for 3 months; 4 mg Neoadjuvant endocrine therapyActive, not recruitingNCT02204098
IGFBP-2, HER2, and IGF-1RDNAID; administered on day 13 and repeated up to 3 times, every 21 days, in the absence of disease progressionPaclitaxel (Chemo), trastuzumab and pertuzumab (anti-HER2)RecruitingNCT04329065
HER2 intracellular domain (ICD)mRNAIV; administered every 2 weeks for 3 injections total; 4 × 108 IUPembrolizumab (anti-PD-1)RecruitingNCT03632941
NeoantigensDNAIM; administered every 28 days ± 7 days, with at least 21 days between injections by electroporation device; 4 mgDurvalumab (anti-PD-L1)Active, not recruitingNCT03199040
 Chemotherapy and Pembrolizumab (anti-PD-1)RecruitingNCT06631092
Shared TAAs and neoantigensmRNAIVSurgery and adjuvant chemotherapyRecruitingNCT02316457
Melanomagp100 and TRP-2DNAIM; administered up to 11 times over 85 weeks using needle-free injection deviceNivolumab (Opdivo) and Ipilimumab (Yervoy)RecruitingNCT04079166
NeoantigensmRNAIM; administered every 21 days up to 9 dosesPembrolizumabProlonged recurrence-free survival in patients receiving vaccine plus Pembrolizumab versus Pembrolizumab alone; greater distant metastasis-free survival in combination therapy treated patientsNCT03897881 [140]
ColorectalOncoprotein MYBDNAID; administered weekly for 6 weeks; 0.1 mg, 0.5 mg, or 1.0 mgTetanus toxoid peptides and anti-PD-1Completed (result not provided)NCT03287427 [141]
ProstatePAP (pTVG-HP)DNAID; administered bi-weekly for 4 weeks; 100 µgSipuleucel-THigher titer antibody responses to PAP detectedNCT01706458 [142]
DNAID; administered every 2 weeks or every 3 weeks; 100 µgPembrolizumab (anti-PD-1)PAP-specific T cells detectedNCT02499835 [143]
DNAID; administered every 2 weeks for 6 vaccinations and then every 4 weeks for 9 vaccinations; 100 µgNivolumab (anti-PD-1)PAP specific T cell responses detected with prolonged time to disease progressionNCT03600350 [144]
PAP (pTVG-HP), Androgen receptor ligand-binding domain (pTVG-AR)DNAID; administered on days 1 and 8 of 21 day cycles; 100 µgPembrolizumab (anti-PD-1)Completed (results not provided)NCT04090528
Androgen receptor ligand-binding domain (pTVG-AR)DNAID; administered weekly for 7 weeks; 100 µgDegarelix (Androgen deprivation therapy), Cemiplimab (anti-PD-1), Fianlimab (anti-LAG-3)RecruitingNCT04989946
PSADNASC; administered monthlyFlutamide (AR blockade)flutamide + vaccine did not further improve patient outcomeNCT00450463 [145]
Three prostate cancer-specific antigens (SL-T10)DNAIM; multiple injections administered; 3 mg or 6 mgGX-I7 (T-cell growth factor), Pembrolizumab (anti-PD-1)RecruitingNCT06344715
NeoantigensDNAIM, followed by EP; administered starting week 21 for a total of 6 treatments every 28 days; 4 mg Nivolumab (anti-PD-1)/ipilimumab (anti-CTLA-4) and PROSTVACCompleted (results not provided)NCT03532217
PSMADNAIM (without EP): 800 µg, 1600 µg, or 3200 µg; IM (with EP): 400 µg, 800 µg, or 1600 µg; administered at weeks 0, 4, 8, 24, and 48Fragment C of tetanus toxinAntigen-specific CD4+ and CD8+ T cells detected; Stronger immune response with IM administration with EP[124]
GlioblastomaWilms tumor gene-1 (WT1), PSMA, hTERTDNAIM followed by EP; administered starting on day 0 and every 3 weeks for 4 doses, then every 9 weeks until disease progression; 3 mgINO-9012 (human IL12), cemiplimab, temozolomide, radiationIncreased antigen specific T cell populationsNCT03491683 [146]
VEGFR-2DNAOral; administered on days 1, 3, 5, 7 every 4 weeks; 106 or 107 CFUanti-PD-L1Increased number of intratumoral CD8+ T-cells detectedNCT03750071 [147]
NeoantigensDNAIM with electroporation; administered once every 28 days for up to 6 dosesRetifanlimab (anti-PD-1)RecruitingNCT05743595
Pancreaticmutant KRASmRNAIMToripalimab (anti-PD-1)RecruitingNCT06577532
NeoantigensmRNAUnknown route; administered 9 weeks post-tumor resectionCamrelizumab (anti-PD-1), Gemcitabine, AbraxaneRecruitingNCT06326736
mRNAUnknown route; administered 12 weeks post-tumor resectionIpilimumab (anti-CTLA-4), gemcitabine, capecitabineNot yet recruitingNCT06353646
mRNAUnknown routeAnti-PD-1RecruitingNCT06496373
mRNAUnknown routeAdebrelimab (anti-PD-L1)Not yet recruitingNCT06156267
 mRNAIV; 25 µg administered weekly beginning 9 weeks post tumor resection plus booster doses at weeks 17 and 46Atezolizumab (Anti-PD-L1), mFOLFIRINOX (chemotherapy)50% of patients had detectable T cell responses to at least one neoantigen; vaccine-induced T cell clonal expansion detected in all immunological responders; longer recurrence free survival in immunological responders; persistent, high-avidity T cell clones detectedNCT04161755 [148,149]
CervicalHPV E6 and E7 (GX-188E)DNAIM with electroporation; administered 3 times; 1 mgGX-17 (IL-7 fused to hyFc), ImiquimodUnknown statusNCT03206138
HPV E6 and E7 (GX-188E)DNAIM; administered on days 1 and 29; 3 mgImiquimod, TA-HPV(recombinant vaccinia virus expressing HPV16/18 E6-E7 fusion proteins)Half of the patients cleared their lesionsNCT00788164
[150]
DNAID: 8 µg or 16 µg
IM: 1 mg or 3 mg
Intralesional: 1 mg or 3 mg
Administered at weeks 0, 4, and 8
ImiquimodIncreased CD8+ T cell infiltration in cervical dysplastic epithelium in intralesional cohortNCT00988559 [151]
Ovarianp62/SQSTM1DNAIM; 2.5 mg; administered weeklyGemcitabineHigher objective response rate in patients receiving combination treatment of DNA vaccine and chemo versus chemo alone; greater number of partial responders and patients with stable disease in combination group; longer progression-free survival in combination treatment groupNCT05979298 [152]
Non-Small Cell LungNeoantigensDNAIM with electroporation; administered once every 4 weeks for 6 cyclesDurvalumab (anti-PD-L1)RecruitingNCT04397003
NY-ESO-1, MAGE-C1, MAGE-C2, survivin, 5T4, and MUC-1mRNAID; administered on days 1, 8, 15, 36, and 57, or days 1, 8, 15, 29, 43, and 57; 320 µgLocal radiationInduction of immune response against six antigens[153]
NY-ESO-1, MAGE-C1, MAGE-C2, survivin, 5T4, and MUC-1mRNAID; each component administered twice for a total of 12 vaccinations; 80 µg Durvalumab (anti-PD-L1), Tremelimumab (anti-CTLA-4)Completed (result not provided)NCT03164772
NeoantigensmRNAUnknown routeAdebrelimab (anti-PD-L1)Not yet recruitingNCT06685653
NeoantigensmRNAUnknown routeAdebrelimab (anti-PD-L1)Not yet recruitingNCT06735508
KRAS mutants (G12D, G12V, G12C, G13D)mRNAIM; administered once every 3 weeks for 9 doses; 1 mgPembrolizumab (anti-PD-1)Completed (results not provided)NCT03948763
Tumor mRNAmRNAIV; administered 7–14 days after last dose of priming vaccine, then every two weeks for 2 doses, and then monthly for 9 dosespp65 RNA-LPNot yet recruitingNCT05660408
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Moseman, J.E.; Shim, D.; Jeon, D.; Rastogi, I.; Schneider, K.M.; McNeel, D.G. Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer. Vaccines 2025, 13, 976. https://doi.org/10.3390/vaccines13090976

AMA Style

Moseman JE, Shim D, Jeon D, Rastogi I, Schneider KM, McNeel DG. Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer. Vaccines. 2025; 13(9):976. https://doi.org/10.3390/vaccines13090976

Chicago/Turabian Style

Moseman, Jena E., Daeun Shim, Donghwan Jeon, Ichwaku Rastogi, Kaitlyn M. Schneider, and Douglas G. McNeel. 2025. "Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer" Vaccines 13, no. 9: 976. https://doi.org/10.3390/vaccines13090976

APA Style

Moseman, J. E., Shim, D., Jeon, D., Rastogi, I., Schneider, K. M., & McNeel, D. G. (2025). Messenger RNA and Plasmid DNA Vaccines for the Treatment of Cancer. Vaccines, 13(9), 976. https://doi.org/10.3390/vaccines13090976

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop