Abstract
Cancer immunotherapy activates the host immune system against tumor cells and has the potential to lead to the development of innovative strategies for cancer treatment. Neoantigens are non-self-antigens produced by genetic mutations in tumor cells that induce a strong immune response against tumor cells without central immune tolerance. Along with advances in neoantigen analysis technology, the development of vaccines focusing on neoantigens is being accelerated. Whereas there are various platforms for neoantigen vaccines, combined immuno-therapies are being developed simultaneously with the clinical application of synthetic long peptides and mRNA and dendritic-cell (DC)-based vaccines. Personalized DC-based vaccines not only can load various antigens including neoantigens, but also have the potential to elicit a strong immune response in T cells as antigen-presenting cells. In this review, we describe the properties of neoantigens and the basic characteristics of DCs. We also discuss the clinical applications of neoantigen vaccines, focusing on personalized DC-based vaccines, as well as future research and development directions and challenges.
In recent years, immunotherapy has attracted attention as a novel approach to eliminate malignant tumors. Immune checkpoints on the surfaces of T cells play a crucial role in maintaining the balance of the immune system by acting as molecular brakes during an immune response. However, it has been shown that tumor cells can escape the immune system by utilizing the checkpoint mechanisms themselves (1). This Nobel Prize-winning research finding has led to the development of therapeutic strategies using immune checkpoint inhibitors (ICIs), such as programmed cell death (PD)-1/programmed death-ligand 1(PD-L1) inhibitors and cytotoxic T-lymphocyte antigen (CTLA)-4 inhibitors, which are now widely used in clinical practice. These therapies exert their anti-tumor effects by inhibiting the immune evasion mechanisms of tumor cells and reactivating T-cell attacks on tumors (2).
In cancer immunity cycle, standard cancer therapies, including chemotherapy, radiotherapy or ICIs, may augment the effects of anti-cancer immunity (3). Chemotherapy and radiotherapy are considered essential for combination with immunotherapy to enhance antitumor efficacy by promoting antigen release from tumor cells, releasing immunosuppression in the tumor microenvironment (TME), and inducing T cell immune responses via antigen-presenting cells (APCs) (4). Immune checkpoints exert a potent immunosuppressive effect, blocking the anti-tumor immune response. It has been reported that neoantigen-specific T cells exhibit a PD1+ phenotype and that PD-L1 in tumor cells is up-regulated after neoantigen vaccination (5). Therefore, a therapy combining a neoantigen vaccine with an ICI could have the potential to induce a strong T cell-mediated immune response to kill tumor cells.
Dendritic cells (DCs) play a central role in cancer immunotherapy owing to their potent antigen-presenting capacity. These cells trigger an immune response by presenting antigens in a form recognizable by T cells; the respective antigens presented by major histocompatibility complex (MHC) class I and II molecules are recognized by CD8+ and CD4+ T cells, activating the T cells. The antigen presentation mechanism in DCs has also been reported, with MHC class I molecules presenting endogenous or exogenous antigens and MHC class II molecules presenting exogenous antigens (6). DCs elicit T cell responses both endogenously and after vaccination during the early stages of the cancer immunity cycle. The role of DCs in the TME is not limited to antigen transfer from tumor to drainage lymph node, but also includes the activation and proliferation of antigen-specific T cells in the tumor. To promote the cancer immunity cycle, it is important to maintain T cell activation by regulating DC activation and maturation (3).
Neoantigens are non-self-proteins that result from nonsynonymous mutations in the tumor cell (TC) genome and are specifically recognized by the immune system. Unlike conventional tumor-associated antigens (TAAs), these highly specific antigens are not present in normal tissues and are unique to tumor cells (7). Thus, as cancer-cell-specific mutant peptides, TC-derived neoantigens have potent immunogenicity and high affinity for the MHC molecules, are unaffected by central immune tolerance, and can be effective cancer immunotherapy targets. Since CD4+ T cell responses have been shown to be essential for neoantigen-related tumor regression (8), it is advantageous to use DCs, that is, APCs that can activate CD4+ T cells via MHC class II molecules, as a neoantigen vaccine.
Research on DC-based vaccines targeting neoantigens may lead to a promising therapeutic strategy for specifically identifying and eliminating cancer cells. This approach enhances the immune response to malignancy and opens new possibilities for personalized cancer therapy. In this review, we will discuss the development status and prospects of DC-based vaccines targeting cancer neoantigens.
Cancer Neoantigens as Targets
Tumor cells express mutant proteins that are normally not present owing to genetic and epigenetic alterations, presenting an altered repertoire of MHC-class-I- and MHC-class-II-related peptides. The activity of cytotoxic T cells against these MHC-class-I- and MHC-class-II-presented mutant antigens (i.e., neoantigens) may lead to cancer regression and is the subject of various basic and clinical studies. Neoantigens are produced by various genetic abnormalities, including somatic single-nucleotide variants (SNVs), gene fusions, and mutational frameshift insertions and deletions (indels) (9). In a mouse melanoma model, mutant synthetic long peptides (SLPs) generated by the transcription and translation of a mutant gene elicited strong cellular immune responses both in vitro and in vivo. Some of immunogenic SLPs exhibited a strong mutation-specific immune response with no cross-reactivity to wild-type peptides (10). Because neoantigens are derived from genetic mutations in tumor cells, they are present in tumor cells but not in normal tissues, unlike TAAs, which are present in both normal and tumor tissues (11). Therefore, neoantigens are ideal targets for cancer therapy that elicit a strong tumor-specific T cell immune response.
With the recent advent of next-generation sequencers (NGS), genetic mutations in individual patients can be easily detected, and artificial intelligence can predict antigens derived from genetic mutations, which have a high binding capacity to MHC molecules (10). Identifying neoantigens requires samples from both normal cells and tumor cells from the same patient. Whole exome DNA sequencing, which is used to detect nonsynonymous somatic mutations, can identify important variants in protein-coding regions. In addition, RNA sequencing can be used to identify expressed mutants and more accurately predict potential neoantigens (12). The bioinformatics pipeline for neoantigen prediction shares four major computational modules: HLA (human leukocyte antigen) typing, calling mutant peptides from somatic mutations and splicing variants, HLA binding prediction, and T cell recognition prediction. These technologies enable the design of neoantigen vaccines to elicit tumor-specific immune responses (13). Neoantigens are finally identified through several processes that validate the T cell response to the predicted antigen (Figure 1). The important function of neoantigens depends on three points: (i) tumor mutational burden (TMB) and neoantigens, (ii) presentation of neoantigens by MHC molecules, and (iii) recognition of cancer neoantigens by T cells. A positive correlation was observed between TMB and the number of cancer neoantigens (14). However, not all neoantigens can be further recognized by T cells and induce a subsequent immune response. Conversely, owing to tumor cell heterogeneity, even cases with a small TMB could exhibit stronger than average T cell reactivity (15).
The efficient presentation of mutant peptides by MHC molecules is critical to their recognition by T cells and their subsequent activation, and although SNVs account for 95% of tumor mutations, indel or frameshift mutations generate immunogenic peptides through the juxtaposition of two peptide sequences or new peptide sequences. Owing to changes in an amino acid sequence and a spatial structure caused by indel or frameshift mutations, these mutant peptides have a stronger affinity for MHC molecules than mutant peptides of non-immunogenic SNVs that either bind poorly to MHC class I molecules or do not contain the mutated amino acid in the appropriate position and have been shown to be more likely to be recognized as neoantigens by T cells (16). T cells recognize antigens presented on MHC molecules, and CD8+ T cells are considered key effector cells in anti-tumor immune responses because of their important role in specifically recognizing cancer neoantigens (17). However, CD4+ T cells also play an important role in anti-tumor immunity as they assist in the initial stimulation and maintenance of CD8+ T cell responses (18), and CD4+ T cell responses have been shown to be essential for neoantigen-related tumor regression (19).
Neoantigens are generally classified into two subgroups: shared neoantigens and personalized neoantigens (8). Shared neoantigens may be common in some tumor types, whereas personalized neoantigens are antigens specific to each individual patient’s tumor. Therapy with personalized neoantigens offers a customized approach for each patient and may represent an important advance in cancer therapy, making neoantigen-targeted personalized immunotherapy the mainstream therapy. Neoantigen vaccines have been designed in various forms, including peptide, DNA, RNA, and DC-based vaccines (20). Among the various neoantigen vaccines being developed, peptide and mRNA neoantigen vaccines account for more than half of vaccines used in clinical trials (21).
In clinical trials of peptide vaccines, short-peptide vaccines were used in the early days, but SLP vaccines, which induce both CD4+ T cells and CD8+ T cells, are now the most common. The SLP vaccine platform has important advantages regarding the safety profile and the good manufacturing practice system. In addition, the 20-30 amino acid SLPs in cancer neoantigen vaccines may be rapidly processed and presented by APCs (22) and have the advantage of binding to both MHC class I and II molecules that activate CD8+ and CD4+ T cells, respectively. After cleavage and antigen processing of SLPs by proteasome, CD8+ T cells recognize short peptides of 8-11 amino acids bound to MHC class I molecules. In contrast, longer peptides of 14-18 amino acids bind to MHC class II molecules and are presented to CD4+ T cells (23).
mRNA neoantigen vaccines have unique advantages; for example, they are not incorporated into the host cell genome, and RNA can be extracted from small amounts of tumor tissue to be amplified for vaccine preparation when suitable tumor tissue is not available. Moreover, mRNA modifications and sequence optimization considerably increase the efficiency of protein expression from mRNA templates, making them more stable, durable, and easier to translate into encoded proteins (24). In terms of deliverability, upon intranodal injection of RNA, DCs in lymph nodes selectively uptake these RNA molecules encoding antigens, attract T cells, and induce expansion of antigen-specific CD8+ and CD4+ T cells (25). The ability to rapidly produce vaccines against arbitrary targets in dedicated mRNA production facilities is a major advantage of using mRNA in neoantigen vaccine development.
Principles of DC-based Vaccines
DC-based vaccines play an important role in cancer therapy: they induce the over-expression of MHC I and II and co-stimulatory molecules (such as CD80 and CD86), resulting in the effective antigen presentation to naive T cells (26). Co-culturing peripheral blood mononuclear cells (PBMCs) with granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4) enhances the efficacy of antigen presentation of DCs (27). This protocol allows ex vivo cultured DCs to be used in clinical studies of cancer vaccines combined with various cancer antigens.
Various types of tumor-specific antigens, such as viral antigens and TAAs as well as neoantigens, including peptides, proteins and mRNAs, have been investigated using the DC-based vaccine platform (Figure 1). For DC/TC fusion vaccine, DCs can phagocytose isolated apoptotic TCs and acquire neoantigens upon fusion. In particular, anti-tumor peptides consisting of 9-15 amino acids are presented on MHC class I molecules on the surfaces of DCs and can be effectively stimulated by tumor-specific CD8+ cytotoxic T lymphocytes (CTLs). When extracellular protein antigens are used, antigens are presented via MHC class II and then via MHC class I by cross-presentation. Antigen presentation via MHC class I for DCs depends on antigen formulation for cross-presentation efficiency and the mode of antigen delivery during antigen processing. Antigen structure and loading method to DCs affect the efficiency of antigen presentation (28). However, loading antigens into DCs by electroporation could efficiently induce cross-presentation regardless of the antigen structures. Electroporation has been used for mRNA and protein loading into DCs, which has been shown to effectively induce antigen-specific CD8+ CTLs (29). Once immature DCs take up the antigen, they migrate to secondary lymphoid organs where they present the antigen to helper or effector T cells, triggering specific CTL responses (30).
DC-based vaccines have strong personalized vaccine features and are being investigated for targeting TAAs and/or neoantigens as individualized therapies. This is expected to elicit a strong immune response against patient-specific tumor antigens. DC-based neoantigen vaccines have shown potential as an effective and safe immunotherapy against solid tumors. Antigens containing neoantigens can be loaded into DCs by various methods, including the use of synthetic peptides, incubation with autologous tumor-derived whole mRNA and whole tumor lysates, and fusion with tumor cells. A key feature of many of these methods is the ability to pulse DCs with tumor lysates containing tumor-associated antigens to generate DC-based vaccines that harbor a wide range of tumor antigens, including neoantigens (31). Sipuleucel-T (Provenge®, Dendreon Corp., Seattle, WA, USA), an FDA-approved autologous cellular immunotherapy for the treatment of metastatic hormone-refractory prostate cancer, uses APCs cultured with a recombinant fusion protein containing prostate acid phosphatase (PAP) to stimulate effector T cells (32). Previously, we successfully prepared an autologous tumor lysate-loaded DC vaccine with high cell viability via electroporation. Our study of the autologous tumor lysate-loaded DC vaccine confirmed its safety and feasibility for 41 patients with various solid tumors, such as carcinomas of the lung, colorectum, breast, stomach and liver. In addition, we observed no autoimmune reactions. The anti-tumor effect of the DC-based vaccine resulted in tumor regression or long-term stable disease, with tumor-reactive immunological responses for 13 patients (31.7%) (33). In addition, an autologous glioma lysate DC-based vaccine may be eligible for more glioma patients than a glioma-associated peptide DC-based vaccine since lysate is HLA-independent while peptide is HLA-dependent (34).
Although it is possible to generate a broad range of tumor-based antigens by utilizing fusion hybrid vaccines of DCs and tumor cells (DC/TC) as well as using tumor lysates (35), their indications are limited by the difficulty in obtaining tumor cells. However, in hematologic malignancies where tumor cells are readily available, such as multiple myeloma, DC/TC fusion hybrid vaccines may be able to induce tumor-specific immune responses and tumor regression (36).
mRNA transfection is a simple method to produce antigens in DCs. A Wilms’ tumor 1 protein (WT1) is reported to be over-expressed in the majority of acute myeloid leukemia (AML) cases. In the phase I/II trial of DC vaccines, DCs transfected with full-length WT1 mRNA for AML by electroporation yielded clinical responses that were found to be correlated with a vaccine-associated increase in WT1-specific CD8+ T cell frequency (37). Thus, a vaccine that introduces TAA mRNA by electroporation has already been generated, and specific immune induction and anti-tumor effects of mRNA-based DC vaccines have been confirmed in clinical trials. However, the development of DC vaccines using mRNAs encoding neoantigens predicted by NGS has not yet been reported. Currently, the most versatile neoantigen-specific DC vaccine platforms are those loaded with SLPs as the identified neoantigens (38).
Various attempts have been made to determine which form of neoantigen, such as SLPs, mRNA, protein, or whole tumor lysate is the best to load in DC vaccines, but no conclusions have yet been made. Furthermore, there are still many challenges, including DC maturation, as well as routes of administration to make DCs effective in priming and activating T cells in draining lymph nodes and the TME (3). Further research and clinical trials are needed to address these challenges, but DC-based vaccines hold much promise for cancer immunotherapy.
Personalized DC-based Vaccines Designed Without Neoantigen Identification by NGS
For DC-based vaccines, various tumor antigens are loaded into DCs; many previous clinical trials of DC vaccines have tested various TAAs along with neoantigens as personalized vaccines loaded into DCs. In an early clinical trial of a DC-based vaccine for glioma, patients received three treatments with a DC vaccine pulsed with tumor peptides eluted from primary cultured cells of an MHC-class-I-matched allogeneic glioblastoma. In this trial, the DC vaccine was well tolerated, and the patients showed a positive immune response, but no objective clinical response was observed (39). Although Sipuleucel-T for the treatment of prostate cancer was approved by the FDA in 2010 as an important milestone in the clinical application of DC vaccines, the efficacy of Sipuleucel-T has been limited, as its tumor progression-free effect has been inconsistent (29). In a study comparing the efficacy of an autologous DC vaccine with that of an autologous irradiated tumor cell vaccine (TCV), after five years of follow-up, these vaccine therapies were found to be well tolerated, and the DC vaccine was associated with a longer overall survival and a 70% reduction in risk of death (median survival was 43.4 months vs. 20.5 months for TCV) (40). In the case of treatment with ICT-107, which is an autologous DC vaccine pulsed with six different peptides targeting glioblastoma and cancer stem cells, the progression-free survival (PFS) in the ICT-107 group was significantly longer than that in the control group in HLA-A2+ glioblastoma patients with methylation of the O6-methylguanine-DNA methyltransferase promoter (24.1 vs. 8.5 months) (41). A significant association between immune responses and favorable clinical outcomes were also observed in these ICT-107 group patients.
The DC/TC fusion hybrid vaccine was well tolerated and safe for solid tumors and hematologic malignancies, such as AML and multiple myeloma. In addition, the DC/TC fusion vaccine increased both CD4+ and CD8+ T cells which express intracellular IFN-γ in response to in vitro exposure to tumor lysate. This suggests the broad antigen-presenting ability of the DC/TC fusion vaccine to produce TAAs containing various neoantigens recognized by the immune system (42).
In a study of DC vaccines in which mRNA was introduced by electroporation, DC vaccines loaded with whole tumor mRNA by electroporation improved the survival of immune responders with advanced melanoma, as confirmed by an immunological evaluation of the induction of T cell responses (43). In addition, DC vaccines against malignant melanoma transfected with mRNA encoding CD40L, caTLR4, and CD70 (TriMix mRNA) and mRNA for melanoma-associated antigen have demonstrated TAA-specific CD8+ T-cell skin infiltration (44).
The most important question is whether personalized DC vaccines loaded with tumor lysates or mRNA can induce T cell responses to tumor neoantigens arising from nonsynonymous somatic cell tumor mutations. Tanyi et al. reported that in a clinical trial of a DC vaccine pulsed with whole tumor lysates of ovarian cancer, neoantigens were identified by NGS in tumor samples from six patients, and T cell reactivity to neoantigens was examined for each patient after five DC vaccinations (45). Before DC vaccination, a neoantigen response was observed in only four patients, but after vaccination, it was observed in all six patients and the number of neoantigens to which T cells reacted increased. Neoantigen-specific CD8+ T cells isolated after vaccination were found to be multifunctional, simultaneously producing interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and IL-2 in response to neoantigens. The results of immunological evaluations in this clinical trial demonstrated that the DC vaccine loaded with tumor lysates enhanced the specific response of T cells to neoantigens after personalized DC vaccination without the pre-identification of neoantigen candidates. Personalized DC vaccines loaded with tumor lysate or tumor mRNA, both containing neoantigens and TAAs, have been noted to potentially contribute to improving overall survival in malignant melanoma and glioblastoma (43, 46).
Clinical Applications of Personalized Vaccines Targeting Neoantigens Identified by NGS
Multiple clinical trials have shown that neoantigen-targeted vaccines are safe and induce specific T cell immune responses. The results of various basic studies and clinical trials indicate that future strategies for personalized neoantigen DC vaccines need to be developed. Significant benefits in terms of overall survival are still limited but are improving with the development of combined immunotherapies.
Neoantigen vaccines for SLP or mRNA platforms. Animal model studies have shown that neoantigens determined on the basis of MHC class I binding affinity are recognized by CD4+ T cells. Vaccines with such CD4+ immunogenic mutations have been shown to counteract the immunosuppressive TME and confer potent antitumor activity, providing the rationale for using SLPs that stimulate CD4+ and CD8+ T cells in neoantigen peptide vaccines (18). Individual HLA-binding neoantigen peptides selected on the basis of the predicted binding affinity were identified after surgery in six patients with resectable Stage IIIB or higher untreated high-risk melanoma. A total of seven subcutaneous inoculations of 13-20 SLPs based on the identified neoantigens were administered along with an adjuvant poly-ICLC. An enzyme-linked immunosorbent spot (ELISpot) assay detected multifunctional CD4+ T cells and CD8+ T cells, respectively, targeting 58 (60%) and 15 (16%) of all 97 unique neoantigens across all six patients. Immunological evaluation confirmed that the CD4+ T-cell response rate was higher than the CD8+ T cell response rate, even though neoantigens were prioritized on the basis of their predicted binding affinity to HLA class I molecules in humans. Four patients without metastatic disease had no recurrence after SLP vaccination. Two other patients with metastatic disease had disease progression after vaccination but received anti-PD1 checkpoint inhibitors and achieved tumor regression (47). Immunological responses of CD4+ T cells have been reported for neoantigen SLP vaccines in glioblastoma clinical trials. Thirteen newly diagnosed glioblastoma patients who were HLA-A*02:01- or A*24:02-positive were vaccinated with a conventional TAA (APVAC1), followed by a cancer neoantigen (APVAC2) SLP vaccine with the adjuvants poly-ICLC and GM-CSF. All the patients underwent surgical resection and received standard adjuvant chemotherapy with temozolomide postoperatively. APVAC1 induced a sustained CD8+ T cell response with a central memory phenotype. In contrast, APVAC2 elicited neoantigen-specific CD4+ T cell responses in all the patients, among which 84.7% (11/13) showed multifunctional CD4 responses with the Th1 phenotype (48). Another clinical trial of the neoantigen SLP vaccine for glioblastoma showed that the vaccine significantly increased the number of tumor-infiltrating lymphocytes (TILs) and induced potent multifunctional novel CD4+ and CD8+ T cell responses to neoantigens in patients who did not receive dexamethasone, which is given for brain edema induced in patients with glioblastoma (49).
In an mRNA neoantigen vaccine study, 13 patients with Stage III or IV malignant melanoma were treated with an RNA-based neoantigen vaccine prepared on the basis of an identified neoantigen. The RNA neoantigen vaccine was well tolerated with no serious adverse events. ELISpot assays of CD4+ and CD8+ T cells before and after vaccination showed that the immunoreactivity of T cells to neoantigens was enhanced in one-third of the patients and a novel immune response in the remaining two-thirds. CD4+ T cells, CD8+ T cells, and combinations of both cells were reactive to 57, 17, and 26% of neoantigens, respectively. Detailed analysis of postvaccination resected metastases from two of the three patients confirmed vaccine-induced T-cell tumor infiltration and neoantigen-specific tumor cell death. In addition, a comparison of the recurrence frequency in all patients recorded before and after neoantigen RNA vaccination showed a highly significant reduction in cumulative recurrent metastatic events over time (5).
DC-based neoantigen vaccines. In 2015, the first Phase I clinical trial of the first individualized neoantigen-loaded DC vaccine was conducted (50). The appropriate neoantigen was determined by epitope prediction using whole tumor exome sequencing in three patients with Stage III resected cutaneous melanoma who were previously treated with ipilimumab. Seven neoantigens selected from each patient and two gp100 peptides were loaded, and ex vivo cultured DCs were administered intravenously to the patients three times. After treatment, an enhanced immune response induced by T cells was observed, and all three patients survived. Moreover, no serious side effects were observed. This clinical study showed that the DC vaccine pulsed with individualized neoantigens was safe and well tolerated. In a study of personalized neoantigen-pulsed DC vaccines, 12 patients with advanced lung cancer were enrolled and 13-30 peptide-based personalized neoantigens were isolated and identified from the tumor tissue of each patient. Simultaneously, the selected neoantigens were pulsed to DCs induced to differentiate from each patient’s PBMCs. The personalized neoantigen-pulsed DC vaccine was shown to be well tolerated and to induce specific T-cell immune responses, with an objective response rate of 25% and a disease control rate of 75%. Specifically, patients with lung cancer that metastasized to the bone, pelvis, and adrenal and inferior vena cava lymph nodes received personalized neoantigen-pulsed DC vaccine therapy after failure of three lines of therapy, including the anti-PD-1 antibody therapy. After five doses of this vaccine, the metastatic lymph nodes and adrenal and pelvic lesions shrank, indicating a favorable therapeutic effect of this vaccine (51).
We conducted a phase I study on the safety and feasibility of a DC vaccine loaded with synthetic short peptides of neoantigens, identified by whole exome sequencing and RNA sequencing of solid tumors using NGS (jRCTc030190213). Four neoantigens with high binding affinity to MHC class I molecules were selected for each patient and 9-10 mer short peptides were generated. DCs were cultured for 5 days in AIM-V medium supplemented with IL-4 and GM-CSF from peripheral blood monocytes. For maturation, the DCs were cultured for an additional 2 days in AIM-V medium supplemented with GM-CSF, IL-4, TNF-α, prostaglandin E2, and neoantigen peptides as previously described (33). Zoledronic acid was also added as an adjuvant to the DCs during culture (52). In this study, a total of 25 subcutaneous vaccinations were administered to five patients with solid tumors and, except for a Grade 1 fever in one patient with pancreatic cancer, the neoantigen DC vaccine was well tolerated. Of the four patients who completed the five vaccinations of the protocol treatment, positive delayed-type hypersensitivity (DTH) reactions were observed in two patients (Table I). We observed that T cells reacted to the neoantigen peptide pool after administration in one of the three patients who could be studied before and after vaccination (53) (Figure 2). The neoantigen-specific T cell response was detected in only one patient because the activation of CD8+ T cells is essential but the frequency of CD8+ T cell activation is not always high (5). Indel-based mutations may generate more immunogenic neoantigens compared to SNVs (7). Out of four neoantigens selected for each patient, two Indel-based neoantigens were used for N-01, and one Indel-based neoantigen was used for N-02. For the other three patients, only SNV neoantigens were used. We used more Indel-based neoantigens for N-01, which may be another reason why a strong immune response was elicited.
Neoantigen-vaccine-based combined immunotherapy. In a clinical trial of NEO-PV-01, an anti-PD-1 antibody and neoantigen personalized vaccine, in 82 patients with advanced melanoma, non-small cell lung cancer, or bladder cancer, the combination therapy was safe and well tolerated. CD4+ and CD8+ T cell responses exhibiting a neoantigen-specific and cytotoxic phenotype were observed in all patients after neoantigen vaccination. In addition, after the vaccination, epitope spreading was observed in response to a neoantigen not included in this vaccine (54). Neoantigen-pulsed DC (Neo-MoDC) vaccines in combination with an ICI have been reported for metastatic gastric cancer. Neo-MoDC vaccines alone have the potential to induce neoantigen-specific CD4+ and CD8+ T cell responses, but combination therapy resulted in a stronger immune response and tumor regression (55).
Thus, these findings hold promise for immunotherapies that combine neoantigen-based DC vaccines with other therapeutic strategies to induce a highly patient-specific immune response, leading to a more potent attack against cancer.
Completed or ongoing clinical trials of neoantigen-targeted DC vaccines are shown in Table II. In many studies, neoantigen peptides are loaded into DCs to develop personalized vaccines; the NCT05886439 study is of interest because it is one of the few clinical trials on mRNA-based neoantigen DC vaccines. In addition, the development of combination therapies is being accelerated through studies combining chemotherapy, radiation therapy, and ICI therapy, as well as other therapies, such as effector cell therapy or microwave ablation. Overall, DC vaccines and neoantigen-targeted therapies are making great strides in the field of cancer immunotherapy, but continued comprehensive research and innovation are needed to maximize their potential.
Future Challenges and Prospects
Personalized DC vaccines have shown efficacy in clinical applications, but several research issues must be solved. Although neoantigens are screened for mutations only in the exon region that accounts for 2% of the entire human genome, 99% of tumor-specific mutations occur in noncoding regions of a gene. In leukemia and lung cancer patients, an abundance of both mutated and nonmutated antigens from noncoding regions was discovered using mass spectrometry and RNA sequencing (56). Neoantigens are generated primarily from mutations in coding regions of the genome, but noncoding regions could also generate neoantigens, such as through promoter mutations, intron retention, non-coding RNAs, or mutation-induced novel open reading frames (13). Mass spectrometry has identified noncoding regions that generate large numbers of nonmutated tumor-specific antigens, most of which are derived from epigenetic modifications in atypical translational events (56, 57). Additionally, these tumor-specific antigens in several noncoding regions have been identified as targets for TIL therapy (56). These tumor-specific antigens in noncoding regions are more pervasive than neoantigens in coding regions and may be shared among tumor patients (56, 58). Not only with the personalized neoantigens but also with the shared antigens derived from epigenetic modifications of noncoding regions, future vaccines have the potential to become both personalized and universal cancer therapies. Tumor neoantigen sequencing and screening are complex and time-consuming processes, but in the future, the time and cost of these processes can be significantly reduced. In addition, the production of personalized neoantigen-pulsed DC vaccines requires the individual culture of DCs recovered from each patient by apheresis, and improvements in DC maturation technology during the DC ex vivo culture process may allow for the development of personalized DC vaccines at lower costs (59).
Conclusion
The development of DC vaccines targeting neoantigens still faces many challenges, including excessive costs, long production times, and the control of DC maturation and migration efficiency. In addition, the selection of antigens containing neoantigens that are loaded into DCs, which is a fundamental issue for DC-based cancer immunotherapy, needs further improvement. In particular, advances in whole exome sequencing technology and the optimization of neoantigen prediction algorithms, as well as the elucidation of mechanisms related to the immune tolerance state in the TME, will enable the development of neoantigen-based DC vaccines that efficiently promote cancer recognition and attack by the immune system. Immunotherapy in combination with neoantigen-based DC vaccines and other therapies, such as chemotherapy, molecularly targeted therapies, and ICIs, has been offered to patients with various cancers and has shown therapeutic efficacy. The results indicate that the new immunotherapy strategy in cancer treatment is going in the right direction. More cancer patients may benefit from the development and optimization of new combined immunotherapies with various ICIs, chemotherapy, radiotherapy, oncolytic virus, antibody drug or adoptive cell therapy, such as TILs or chimeric antigen receptor (CAR)-T, in accordance with the cancer immunity cycle (3).
As there are prospects to increase the adaptability of neoantigen-based DC vaccines for various cancer types, it is imperative to develop novel approaches to realizing personalized medicine tailored to the cancer characteristics of individual patients, namely, biomarker research for the determination of therapeutic indications and the prediction of efficacy. Such an exploration will further increase the efficacy of immunotherapy and contribute to the improved survival of cancer patients.
Acknowledgements
This work was carried out (in part) at the Intractable Disease Research Center, Juntendo University Graduate School of Medicine. The Authors thank Ms. Shoko Saiwaki and Ms. Ayumi Yamaguchi for their assistance with immunological analyses. The Authors also thank Ms. Mariko Mitsubori for preparing the figures.
Footnotes
Authors’ Contributions
TK, RT and SG contributed to Data acquisition; Formal analysis; Investigation Resources; Supervision; Validation; Visualization; Funding; Writing of the original draft for this review. SO, HI and EO contributed to Formal analysis; Validation; Supervision; Funding acquisition. All Authors have read and approved the final version of this manuscript.
Conflicts of Interest
The Authors affirm that there are no potential conflicts of interest in relation to this study.
- Received May 21, 2024.
- Revision received July 9, 2024.
- Accepted July 24, 2024.
- Copyright © 2024 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.
This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).