Next Article in Journal
The Cost-Effectiveness of the Human Papilloma Virus Vaccination in Asia Pacific Countries: What Lessons Can Indonesia Learn?—A Systematic Review
Previous Article in Journal
Overcoming HPV Vaccine Hesitancy in Japan: A Narrative Review of Safety Evidence, Risk Communication, and Policy Approaches
Previous Article in Special Issue
A Spike-Based mRNA Vaccine Encapsulated in Phospholipid 1,2-Dioleoyl-sn-Glycero-3-PhosphoEthanolamine Containing Lipid Nanoparticles Induced Potent B- and T-Cell Responses Associated with Protection Against SARS-CoV-2 Infection and COVID-19-like Symptoms in Hamsters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer Vaccines: A Promising Therapeutic Strategy in Advanced Solid Tumors

by
Simona Caridi
1,†,
Valeria Maccauro
1,†,
Lucia Cerrito
1,
Gianluca Ianiro
2,3,
Maria Pallozzi
1,
Leonardo Stella
1,
Antonio Gasbarrini
1,3 and
Francesca Romana Ponziani
1,3,*
1
Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
2
Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
3
Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2025, 13(6), 591; https://doi.org/10.3390/vaccines13060591
Submission received: 15 March 2025 / Revised: 22 May 2025 / Accepted: 25 May 2025 / Published: 30 May 2025
(This article belongs to the Special Issue Role of Next Generation Vaccines in Immunotherapeutics)

Abstract

Recent advancements in understanding how cancer cells evade immune recognition have led to significant progress in cancer immunotherapy. Therapeutic cancer vaccines hold great promise due to their safety, specificity, and ability to establish lasting immune memory, serving as an effective immunotherapy either alone or in combination with other treatments in clinical research. Cancer vaccines aim to restore the host’s innate and adaptive anti-cancer immune responses by stimulating antigen-presenting processes and reversing the immunosuppressive environment that facilitates tumor immune evasion and metastasis. Although in clinical studies cancer vaccines have been observed to not effectively induce tumor regression, they can enhance local immune responses in combination with other immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), thus delaying cancer recurrence and prolonging overall survival in advanced tumor settings.

1. Introduction

Nowadays, solid tumors are diagnosed in advanced metastatic stages, as surgery and chemotherapy may not be suitable therapeutic options to prolong patient overall survival. In this regard, immunotherapy, an innovative technique specifically targeting cancer cells by fostering the patient’s own defenses, has been developed with promising results.
Cancer immunotherapy enhances the activity of the patient’s immune cells against tumors using innovative approaches, including adoptive cell therapy (ACT), immune checkpoint inhibition (ICIs), and cancer vaccines [1].
Tumors often downregulate immune responses through the tumor microenvironment (TME), which involves immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Re-activating the immune system typically requires exogenous antigen-presenting cells (APCs), which present tumor-associated antigens to cytotoxic (CD8+) and helper (CD4+) T cells via major histocompatibility complex (MHC) molecules [2].
Once tumor-associated antigens are recognized by dendritic cells (DCs) at the tumor site, these cells migrate through the lymphatic vessels to the T-cell zones of lymphoid tissues. There, they stimulate T lymphocytes by simultaneously expressing costimulatory molecules. DC-primed CD8+ cytotoxic T lymphocytes (CTLs) can identify tumor-associated antigens presented on the surface of cancer cells via MHC class I molecules and subsequently destroy these cells. Meanwhile, naïve CD4+ T cells differentiate into effector cells, initiating a B cell-mediated, TAA-specific antibody response [3] (Figure 1).
Solid tumors, however, have developed various mechanisms to evade the immune response. Factors contributing to the immunosuppressive TME include hypoxia, an altered extracellular matrix, neoangiogenesis, cancer-associated fibroblasts, Tregs, MDSCs, and tumor-associated M2 macrophages (TAMs). These elements collectively impair the effectiveness of immunotherapy [4]. To counteract this mechanism, new strategies are being developed to restore the anti-cancer immune response and enhance the efficacy of conventional immunotherapies.
Cancer vaccines utilize cancer-specific antigens to stimulate the immune system to counteract tumors by inducing a robust and long-lasting antigen-specific expansion of both CD4+ and CD8+ T cells [5]. These vaccines not only reinvigorate pre-existing immune responses, but also initiate new immune reactions against tumors [6].
In this review we aim to analyze the most important cancer vaccine types currently or potentially used as immunotherapy agents alone or in combination with other immunomodulants for advanced solid tumors.

2. Cancer Vaccines as “Antigen-Specific Target Therapy”

The effectiveness of cancer vaccines is based on the stimulation of an antigen-specific immune response targeted to induce tumor regression and to produce a sustainable memory immune response [7]. These vaccines are designed to activate the host’s T cells against specific antigens on cancer cells surfaces, which can be divided into tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs, also called “neoantigens”); while the first ones are aberrant molecules self-expressed on cancer cells’ surfaces, the second ones are, instead, exclusively produced by tumor cells from non-driver genomic mutations unique to tumor cells [8,9].
As an example of TAAs, melanocyte differentiation proteins (e.g., gp100, tyrosinase) [10] and Human epidermal growth factor receptor 2 (HER2) [11], as well as Cancer-testis antigens, including New York esophageal squamous cell carcinoma 1 (NY-ESO-1) and the melanoma antigen gene (MAGE) protein family [12], have been used as cancer vaccines in many clinical trials to elicit responses against multiple class I and class II MHC-restricted epitopes.
However, despite the fact that cancer vaccines could effectively foster the host adaptive immune response against specific tumor epitopes, a low magnitude of CD8+ T cells and humoral agents was observed in clinical trials, which were ineffective in targeting tumor spread. This phenomenon may be explained via the cancer immunoediting processes, including antigen loss and the selection of non-immunogenic epitope variants (immunoselection), stimulated by vaccines targeting single specific epitopes on tumor cells [13,14].
The most important limitation of cancer vaccines stems largely from the challenge of identifying TAAs and neoantigens [9,15,16]. Tumor antigens can originate from various sources, including autologous modified tumor cells (known as autologous whole-cell vaccines) [17], killed tumor cells, tumor cell lysates, tumor-derived exosomes, tumor peptides and proteins, and neoantigens derived from genetic mutations in tumor-specific molecules, which trigger a cytotoxic T lymphocyte response [18]. Cancers characterized by a high tumor mutation burden (TMB) and a greater frequency of tumor neoantigens tend to exhibit higher rates of tumor infiltrating lymphocytes (TILs). These so-called “hot tumors” are generally more responsive to cytolytic T cell activity and immune checkpoint inhibitors [19,20]. However, the heterogeneity of cancer cells often results in the simultaneous expression of various antigen types, necessitating a combination of neoantigens to elicit an effective immune response.
Tumor cells can employ several mechanisms to evade immune recognition while protecting healthy tissues from autoimmune damage. These include the inefficient processing and presentation of tumor antigens, the upregulation of negative costimulatory ligands that induce T cell anergy and expand the regulatory cell populations, the production of immunosuppressive molecules, such as Fas-L, TGF-β, CTLA-4, and PD-1/PD-L1, the increased expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase, the downregulation of MHC-I molecules on the cell surface, and the impairment of antigen presentation [21]. Combining neoantigen vaccines with ICIs has been proposed to enhance immunotherapy outcomes. Neoantigen-based vaccines can amplify neoAg-specific TIL populations, which, when reactivated through the checkpoint blockade, can effectively infiltrate the TME and destroy cancer cells.
In addition, a critical role of tumor-cognate CD4+ T cells has been hypothesized. In fact, they are supposed to either promote antigen-specific CD8+ T cell responses by enhancing dendritic cell activity as antigen-presenting cells, or mediate direct tumor killing independent of CD8+ T cells and stimulate other innate immune cells’ antitumor responses. Thus, the lack of tumor-cognate CD4+ T-cell responses may explain the limited anti-tumoral efficacy observed in vaccines restricted to epitopes stimulating CD8+T cells [22,23].
On the other hand, neoantigens represent a new field of interest for developing anti-cancer vaccines that overcome the limitation of central immune tolerance, especially when combined with immune checkpoint inhibitors in advanced solid different tumors (e.g., bladder cancer, melanoma, Non-Squamous-Cell Lung Cancer) [24,25]. In particular, this can involve the formulation of shared neoantigens, representing driver mutations expressed by all cells within a patient’s tumor, such as KRAS. Nevertheless, we still require future studies seeking to create vaccines based on multiepitope shared neoantigens [26].

3. Types of Cancer Vaccines

Cancer vaccines are categorized as either preventive or therapeutic. Preventive vaccines aim to protect individuals from certain cancers by inducing immune memory against specific cancer antigens. In contrast, therapeutic vaccines stimulate the immune system to enhance the activity of endogenous T cells against the immunogenic antigens expressed on tumor cell surfaces [5]. A distinct subgroup, known as immune-modulatory vaccines, works by activating anti-regulatory T cells (anti-Tregs) to counteract the immunosuppression mediated by regulatory immune cells [27].
Cancer vaccines can be developed through various approaches [28], and can be distinguished into the following categories (Table 1):
  • Cell-based vaccines. These involve immune cells pre-loaded with cancer-specific antigens and subsequently administered to patients, either alone or in combination with other anti-cancer therapies;
  • Protein/polypeptide vaccines. These utilize modified molecules capable of eliciting specific immune responses;
  • Nucleic acid vaccines. These include DNA and RNA vaccines designed to express tumor antigens, as well as nanovaccines and tumor exosome-based vaccines.

3.1. Cell-Based Vaccines

Cell-based vaccines can be derived from autologous or allogeneic human cancer cell lines or tumor lysates, either loaded on dendritic cells or administered in adjuvants to promote dendritic cell activation in order to stimulate specific CD8+ and CD4+ T cell responses [7].
In the first subgroup, Melacine and Canvaxin have been used as two early allogeneic tumor cell-based vaccines for melanoma. For Melacine, a significant benefit in overall survival (OS) and an improvement in recurrence-free survival (RFS) within a subgroup expressing HLA-A2 and HLA-Cw3 were observed, thus imparting the hypothesis that genetic predisposition might influence the response to cancer vaccines [35].
For Canvaxin, composed of a mixture of irradiated whole-cell melanoma lines with bacillus Calmette–Guérin (BCG) as a vaccine adjuvant, no clinical benefit versus placebo has been observed in patients with advanced melanoma [36].
On the other hand, in the second subgroup of this category, DC vaccines have demonstrated the ability to stimulate antitumor defenses through a dual mechanism—by priming effector T cells against tumor antigens, and by enhancing natural killer (NK) cell activity via cytokine release and the expression of adhesion molecules such as CD155 [37]. The use of DC vaccines has shown potential in enhancing the efficacy of cytokine-induced killer cell immunotherapy for solid tumors, leading to prolonged survival and a strengthened immune response without inducing specific autoimmune side effects [38].
A notable example was represented by Sipuleucel-T, a DC vaccine containing prostate acid phosphatase (PAP) combined with granulocyte–macrophage colony-stimulating factor (GM-CSF), which has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of metastatic castration-resistant prostate cancer [39]. In a neoadjuvant setting prior to radical prostatectomy, Sipuleucel-T significantly increased tumor-immune infiltration in surgical specimens compared to pre-treatment biopsies, and improved OS by four months compared to placebo [39,40]. Despite its effectiveness, this drug is no longer available; in fact, it has been retired due to its high price, complex way of administration, and uncertainty regarding its reimbursement. Furthermore, the vaccine’s supply was limited during the first year of launch due to limited manufacturing capacity [41,42]. Even though Sipuleucel-T is not currently used in clinical practice, the idea of targeting cancer tissue’s differentiated antigens despite the presence of nonmutated ones has been attracting scientific interest, as it could overcome the limit of cancer immunoediting and avoid off-target effects in future trials [39].
Further research on DC vaccines in triple-negative breast cancer has shown an enhanced immune response within the tumor microenvironment, characterized by increased tumor-infiltrating lymphocytes (TILs), elevated PD-L1 expression, and heightened peripheral immune activity, including enhanced lymphocyte proliferation, infiltration, IFN-γ production, and humoral responses [43].
Additionally, a phase I clinical trial involving patients with stage I-II cervical cancer evaluated subcutaneous injections of a DC vaccine carrying keyhole limpet hemocyanin and full-length HPV16/18 E7 antigens. The study demonstrated increased CD4+ T cell responses and a cervical cancer-specific humoral immune response [44].
Recent studies in metastatic melanoma have reported promising results derived from the combination of DC vaccines and ICIs, such as anti-PD-1 and anti-CTLA-4 antibodies, even in patients refractory to checkpoint blockade therapies [45,46,47]. Despite these advances, the impact of this combination on OS has not been consistently significant, and further evidence is required to validate the clinical application of this approach [48].

3.2. Protein/Polypeptide Vaccines

Peptide-based vaccines are specific, safe, and effective in producing a strong, adaptive anti-cancer immune response. The peptide-MHC and T cell receptor interactions are highly immunogenic and result in less ‘off-target’ toxicity and central tolerance. As an example, the Synthetic Long Peptide (SLP) vaccines are subunit vaccines made from peptides mimicking cancer antigens’ epitopes. They contain class-I and class-II MHC- restricted neoepitopes that help to stimulate both specific CD8+ T cell and CD4+ T cell responses. Immunogenicity increases with vaccines containing a huge amount of epitopes and recognition motifs. In fact, the use of long synthetic peptides of tumor antigens instead of short peptides has been observed to foster antigen presentation and tumor responses [14]. Short peptides (~9 aminoacid residues), instead, can be either easily digested by host’s enzymes, or loaded onto MHC-I molecules of non-professional APCs, leading to a wicker T cell response. Longer peptides (25–35 aminoacid residues), instead, are usually loaded onto professional APCs, in order to stimulate specific MHC-II T cell responses [29]. Two important clinical examples of this subgroup are a protein vaccine targeting the parathyroid hormone-related protein (PTHrP) in hypercalcemic solid tumors, and a combination of cancer peptides, called TAS0313, derived from eight disregulated cancer genes (EGFR, KUA, LCK, MRP3, PTHRP, SART2, SART3, and WHSC2) in glioblastoma [49]. Currently, a novel proteogenomic approach is under development to identify potential protein antigens (e.g., 19 tumor-specific peptides) for cancer vaccines in advanced colorectal cancer, but further evidence is needed before applying them in clinical practice [50].

3.3. Nucleic Acid-Based Vaccines

mRNA-based vaccines have gained significant attention due to their versatility and favorable safety profile. Unlike viral vector vaccines, mRNA vaccines are non-integrative, non-infectious, and readily modifiable. They are delivered via carrier molecules (often lipid nanoparticles) and do not elicit anti-vector immunity [30,51]. Upon administration at tumor sites, mRNA vaccines are recognized by Toll-like receptors (TLR7/8), which stimulate type I interferon (IFN) production, thereby enhancing DC-mediated antigen presentation to T cells and IL 12-expression [51]. Another advantage is that mRNA-encoded proteins can undergo post-translation modifications such as glycosylation, acetylation and methylation to produce tumor-specific folded proteins, which could in turn become antigens for cancer vaccines [14].
The clinical application of mRNA vaccines has shown remarkable promise, in particular in stage III and IV melanoma, as personalized RNA-based vaccines combined with PD-1 blockade therapy reduced metastasis rates and improved progression-free survival [52]. A phase Ib trial demonstrated durable neoantigen-specific CD4+ and CD8+ T cell responses with significant tumor reduction in metastatic melanoma lesions [24]. In addition, a nanoparticulate liposomal mRNA vaccine with sequences encoding for four class I MHC-restricted TAAs, with tetanus toxoid epitopes and targeting signal sequences intended for reaching immature dendritic cells in the lymphoid tissue, in combination with ICIs therapy in patients with resected and unresected advanced melanoma demonstrated a more effective CD4+ T cell-dominant responses, tumor killing, and a longer tumor-specific memory T-cell formation [31].
Finally, in resected pancreatic adenocarcinoma, an mRNA-based neoepitope vaccine combined with atezolizumab and chemotherapy improved recurrence-free survival and achieved a 50% overall response rate in a phase I study [53].
Another kind of nucleic acid-based vaccines is represented by DNA vaccines. These are made by closed circular DNA plasmids of bacterial origin encoding proteic antigens, and they are able to promote humoral and cellular responses restricted to target antigens, as well as to stimulate TLRs or membrane-bound receptors critical in DC priming and in B cell and natural killer cell activation, thus fostering local inflammatory reactions [14]. For example, a DNA-based vaccine based on ErbB2, an epidermal growth factor often highly expressed in some colorectal, pancreatic, endometrial, gastric, and breast cancers, has been tested in mouse models, eliciting a more powerful anti-cancer immune response compared with controls [54]. However, DNA vaccines’ delivery is more complex than that of RNA vaccines due to the higher dimensions and the nuclear localization, and thus further studies on strategies for ameliorating transfection are still needed [14].

4. Chimeric Proteins Stimulating the Immune System Against Tumors

An alternative approach in cancer treatment is represented by the injection of soluble molecules capable of stimulating specific immune pathways. For instance, the administration of a chimeric protein (BLS-MICA)—composed of the MICA domain fused with a molecule derived from Brucella spp. (BLS)—has shown promising results. This fusion protein induces the production of high titers of anti-MICA antibodies, which, in turn, neutralize MICA ligands expressed on the surface of tumor cells. This neutralization triggers Antibody-Dependent Cellular Cytotoxicity (ADCC), leading to effective tumor cell destruction [55].

5. Vaccines as Adjuvant to Other Immunotherapy Treatments

To overcome the challenge of delivering exogenous tumor antigens into the tumor milieu, researchers have adopted strategies to generate a large amount of endogenous cancer antigens within the tumors, thus creating “in situ cancer vaccines”. This can be achieved by inducing immunogenic cell death, which involves the apoptosis of tumor cells through chemotherapy (e.g., anthracyclines) and radiotherapy, and by upregulating the presentation of the antigens released by APCs in a proinflammatory milieu driven by a cytokine storm in the TME, which in turn facilitates the recruitment of APCs and T effector cells [18].
Nevertheless, immunogenic cell death is often insufficient to overcome the immunosuppressive TME, and adjuvant agents are needed to improve antigen presentation, repolarize immunosuppressive M2 macrophages and MDSCs, increase the CD8+/Treg ratio, and stimulate the differentiation of effector memory T cells [18].
Moreover, vaccines may upregulate the expression of PD-L1 in the tumor microenvironment, thereby turning “cold” tumors into “hot” tumors, and boosting the anticancer effects of PD-1/PD-L1 inhibitors [52].
In this regard, in situ cancer vaccines, composed of various biomaterials such as oncolytic viruses, bacterial products, and nanomaterials, have been engineered to induce both cancer cell death and intratumoral inflammation, thereby enhancing the efficacy of concomitant anticancer treatments (Figure 2).

5.1. Oncolytic Virotherapy

Oncolytic virotherapy (OV) is a new type of immunotherapy using a viral vector selectively infecting and lysing tumor cells, which results in releasing tumor antigens to promote an anti-tumor adaptative immune response, while preserving normal tissues. Therefore, the combination of oncolytic viruses with other intratumoral therapies to directly alter the tumor microenvironment, thus fostering in situ vaccination effects, may be a promising future anti-cancer treatment [32].
The anti-cancer effects here are due to direct oncolysis, the induction of innate and adaptive immune responses, the alteration of the immunosuppressive TME, and the disruption of tumor blood vessels [56].
Therefore, it can be used to increase T cell migration, survival, and expansion at tumor sites, enhance APC function, reverse T cell exhaustion, and convert “cold” tumors into “hot” tumors, thereby improving the efficacy of other immunotherapy treatments [57].
Nevertheless, although oncolytic viruses have demonstrated high effectiveness against cancer, their lack of tumor specificity results in substantial off-target replication and toxicity; moreover, they may bind to various cell types, potentially preventing them from reaching the target tumor in sufficient numbers. Consequently, it is necessary either to select viruses with natural oncotropism or to genetically engineer viruses for enhanced tumor specificity, as well as to use cell carriers with tumor-homing capabilities [58].
In addition, OV exerts immunomodulatory anti-cancer activity by attracting cytotoxic T lymphocytes to the tumor and upregulating PD-L1 expression on both cancer and immune cells [59]. It also directly induces tumor cell apoptosis, thereby releasing tumor-associated antigens that are subsequently processed by APCs to stimulate a cytotoxic T lymphocyte (CTL) anti-tumor response [60].
Oncolytic viruses have been approved for various tumors such as melanoma, glioblastoma, prostate cancer, and head and neck cancer, and they could play a promising role in enhancing the effects of immunotherapy, but detailing the overall knowledge in this field lies outside the scope of this review.

5.2. Nanomaterial-Based Vaccines

Nanomaterial-based vaccines are increasingly used as adjuvants to enhance the efficacy of other anti-cancer immunotherapies. Among these, liposomes are the most widely employed carriers. They can be pre-loaded with chemotherapeutic agents (such as doxorubicin) and adjuvants to generate tumor antigens in situ, thereby amplifying immune responses within the TME. When combined with conventional immunotherapies, such as anti-PD-1 or anti-CTLA-4 treatments, liposomal vaccines have shown promise in preclinical models of colon cancer and melanoma [33]. Liposomal formulations can also be engineered to include molecules that counteract the immunosuppressive TME and improve local antigen presentation, such as Toll-like receptor agonists, cyclic GMP-AMP (cGAMP), and bacterial membrane components [61].
Other notable nanomaterial-based carriers include inorganic nanoparticles such as silica and alum. These materials stimulate local inflammatory responses by delivering cytokines such as IL-12, IL-2, IL-15, and IFN-γ, thereby promoting dendritic cell (DC) recruitment and T cell priming [62,63]. They can also be pre-loaded with chemotherapeutics or radiation particles to induce immunogenic cell death, further enhancing the immune-stimulatory effects of other therapies [64].
Another novel anti-cancer vaccines approach uses soluble antigens mixture released into cell culture supernatants of several melanoma cell lines, to induce both antibody and T cell responses to the antigens in the vaccines. Even though a small randomized placebo-controlled phase II trial showed significant improvement in recurrent free survival, no phase III trials are currently available that support this hypothesis [65].

6. Other Kinds of Vaccines in Immunotherapy Adjuvant Setting

Exosomes are extracellular vesicles capable of transferring various intracellular molecules—including antigens and MHC complexes—to adjacent or distant cells. Exosomes play a dual role: they facilitate communication between immune cells and present tumor-specific antigens to the immune system, thereby counteracting the immune evasion mechanisms employed by cancer cells [4].
Exosomes derived from immune cells within the TME have distinct functions. NK cell-derived exosomes contain cytotoxic proteins (e.g., perforin, Fas ligand, granzyme A/B, and granulysin) that directly kill cancer cells. They also transport cytokines like IL-15 and tumor-suppressive microRNAs that inhibit tumor growth. M1 macrophage-derived exosomes enhance local inflammation and promote the conversion of M0 macrophages into pro-inflammatory, anti-tumor phenotypes by releasing cytokines such as IL-6, IL-12, and TNF-α [4]. Conversely, tumor-derived exosomes can facilitate immune escape by increasing the expressions of PD-L1 and CD73 and by secreting immunosuppressive molecules (e.g., lactic acid), which downregulate costimulatory molecules (CD28, CD80) on T cells and promote immunotherapy resistance [66,67]. Despite these complexities, tumor-derived exosomes hold promise as vaccines. By loading MHC-I and tumor antigens onto their surface, tumor-derived exosomes can effectively induce T cell-mediated anti-tumor responses. Preclinical studies in melanoma and colon cancer models have shown that TDE-based vaccines prolong survival, delay tumor onset, provide long-term immune memory, and reduce metastasis [68].
A specialized subgroup of exosomes, known as dexosomes, are secreted by DCs. Dexosomes contribute to antigen presentation by incorporating MHC–antigen complexes, which are subsequently transferred to naïve DCs, triggering both innate and adaptive immune responses [3]. Once administered, dexosome vaccines efficiently reach lymph nodes, where they are internalized by resident DCs. This uptake stimulates T cells through the expression of costimulatory molecules (CD54, CD80, CD86) and integrins (CD11b/CD18, CD11a/CD18), while also delivering DC-modulating microRNAs (miRNAs) [3]. In addition to activating T cells, dexosomes can stimulate B cells to produce antigen-specific antibodies, thereby coordinating both humoral and cellular immune responses [69]. Second-generation dexosome therapies have further enhanced anti-tumor effects by promoting NK cell function via NKp30 receptor engagement and BAG6 ligand expression, which together trigger TNF-α and IFN-γ secretion [70].
Emerging platforms such as antigen self-presentation and immunosuppression reverse (ASPIRE) leverage DC-derived membranes loaded with multiple costimulatory signals (CD80/CD86) and anti-PD-1 antibodies to strengthen T cell-mediated immunity [71]. Combination therapies involving dexosomes and immune checkpoint blockades (e.g., PD-1/PD-L1 or CTLA-4 inhibitors) are under investigation for their potential to overcome TME immunosuppression and enhance T cell activation. However, the optimization of molecular and subcellular components remains ongoing, with only two clinical studies completed to date showing promising preliminary results [70,72].

Immune-Modulatory Vaccines (IMVs)

Immune-modulatory vaccines (IMVs) work by activating antiregulatory T cells (anti-Tregs), a subgroup of T cells that specifically target regulatory immune cells, thereby mitigating the immunosuppressive TME. IMVs not only facilitate the direct cytotoxic targeting of tumor cells, but also remodel the TME into a proinflammatory environment that restricts tumor progression. This remodeling includes reprogramming TAMs from an immunosuppressive M2 phenotype to an M1-like phenotype, which supports anti-tumor T cell responses and enhances antigen presentation. Additionally, IMVs can convert cancer-associated fibroblasts (CAFs) into immunocompetent fibroblasts, further promoting anti-tumor immunity.
One example is a TGF-β-based IMV developed to control tumor growth in murine models of pancreatic cancer. This vaccine targets both immunosuppression and fibrosis within the TME. Vaccine-induced T cells not only directly kill tumor cells, but also indirectly modulate the immunosuppressive environment by targeting M2 macrophages and CAFs [27,73]. Another promising approach involves indoleamine 2,3-dioxygenase (IDO)-based vaccines. A phase I clinical trial demonstrated that IDO-targeting monotherapy resulted in a median OS of 26 months and prolonged disease progression-free survival of up to six years in lung cancer patients when used as maintenance therapy [74].
The combination of IMVs with ICIs is emerging as a particularly appealing strategy. IMVs can induce novel T cell activation, foster local inflammation, and modulate the TME to create an immune-favorable environment that enhances the anti-tumor efficacy of ICIs. This combination approach was evaluated in a phase II clinical trial involving patients with metastatic melanoma, wherein the therapy achieved an 80% objective response rate [34,75]. An ongoing multicenter phase III trial (NCT05155254) is investigating the efficacy of an IDO/PD-L1 vaccine in combination with pembrolizumab compared to pembrolizumab alone. Preliminary results indicate promising improvements in progression-free survival and safety profiles, consistent with outcomes observed in preclinical models.
Beyond ICIs, combining IMVs with other immunotherapies—such as ACT or traditional cancer vaccines—represents a promising avenue for enhancing anti-tumor immunity. While in vitro models have shown encouraging results, further in vivo studies are necessary to fully assess the clinical applicability of these approaches [27] (Figure 2).

7. Limitations of Cancer Vaccines as Immunotherapy

Cancer vaccines face several limitations that affect their clinical applicability. These challenges include peripheral and central immune tolerance to tumor antigens, the risk of inducing autoimmunity against normal tissues, and the rapid mutational evolution of tumors, which allows them to evade T cell-mediated immune responses.
Regarding immune tolerance, neoantigens—which are tumor-specific antigens—are less likely to induce immune tolerance or trigger autoimmune reactions due to their absence on normal cells. However, the development of neoantigen-based vaccines remains challenging because of the limited and often variable expression of these antigens on the surface of cancer cells [27].
In addition, off-target effects arising from antigens shared on both tumor and normal cells, dominant antigens that promote tumor immune escape or limit T cell responses, and the suboptimal selection of vaccine adjuvants for antigen processing and presentation, represent other important limitations to the clinical effectiveness of cancer vaccines [76].
Furthermore, various vaccine platforms—such as peptide-based, cell-based, RNA, DNA, and viral vector-based vaccines—present inherent challenges that limit their clinical utility, such as rapid degradation by host enzymes within the TME, which can reduce their efficacy [77]; low intrinsic immunogenicity leading to naturally poor immunogenicity, necessitating high doses of genomic material or the use of potent adjuvants to elicit sufficient immune responses [27]; and other mechanisms of resistance [78]. Key resistance mechanisms include the following:
  • The downregulation of HLA molecules, reducing antigen presentation to T cells;
  • Decreased neoantigen load, limiting the number of recognizable tumor antigens;
  • Aberrations in key signaling pathways, affecting immune activation;
  • Adaptive immune system modifications that impair long-term immune responses;
  • Innate immune evasion mechanisms, leading to immune cell dysfunction and destruction.
Given all these limitations, the use of vaccines as a cancer prevention strategy for solid tumors in high-risk individuals, such as genetically predisposed subjects or pre-malignant lesion carriers, in clinical settings may be important to optimize vaccine efficacy [7].

8. Economic Evaluation of Cancer Immunotherapy

The rising prominence of immunotherapies for solid tumors, including monoclonal antibodies (mAbs) and therapeutic cancer vaccines, has prompted critical evaluations of their economic impact; so far, most of the data regarding solid tumors have been reported for monoclonal antibodies. The systematic review by Geynisman et al. is the only one to have analyzed the cost-effectiveness of a cancer vaccine (e.g., Sipuleucel-T) that was available in 2014 for the treatment of castration-resistant prostate cancer (now unavailable in clinical practice due to the aforementioned reasons), and remarked on an incremental cost–effectiveness ratio (ICER) of nearly USD 290,000 per quality-adjusted life (QALY), raising concerns about its affordability despite its modest survival benefits. Conversely, rituximab (a monoclonal antibody) was associated with favorable ICERs in multiple studies, reinforcing its role as a benchmark for cost-effective immunotherapy, particularly in the treatment of non-Hodgkin lymphoma. Other agents showed limited cost-effectiveness due to high ICERs that exceeded widely accepted thresholds (e.g., USD 50,000–100,000 per QALY).
A central theme emerging from the literature is the lack of predictive biomarkers to guide patient selection, which limits the clinical and economic efficiency of these therapies. Inappropriate patient selection not only reduces efficacy, but also exacerbates healthcare costs. Moreover, the pricing strategies of pharmaceutical companies—often based on the highest-priced competitor—contribute to unsustainable cost escalations.
Despite the financial burden, the clinical value of immunotherapies remains significant, particularly for patients who achieve durable responses.
In conclusion, while cancer immunotherapies represent a paradigm shift in oncologic care, their cost-effectiveness remains variable and context-dependent. Moreover, further analyses are needed in the field of cancer vaccines, which are still under-investigated from the point of view of their economic impact on the healthcare system. Future research should prioritize the development of predictive tools, rational pricing models, and real-world data to refine the economic framework guiding immunotherapy deployment [79].

9. Conclusions

Several studies have shown that a low pretreatment lymphocyte count is associated with poor survival outcomes in patients with solid tumors undergoing immune-based therapies [80,81]. In contrast, patients with preexisting immunity to tumor neoantigens exhibit improved clinical responses to immunotherapy [82]. Based on these findings, combining ICIs with agents that induce a sustained neoantigen-specific T cell response represents a promising strategy for treating advanced solid tumors that are refractory to conventional therapies.
Cancer vaccines, in this context, may emerge as key adjuvant immunotherapies, as they can recruit and activate T cells specific to tumor-associated antigens. This capability allows vaccines to overcome the limitations of ICIs, especially in targeting the “cold” tumors—those with poor immunogenicity—commonly observed in advanced and metastatic solid cancers. As previously discussed, these vaccines enhance the host’s immune defense by promoting local tumor-neoantigen presentation, which helps restore the population of effector T cells and bolsters the efficacy of other immunotherapies in preclinical models.
Importantly, the neoantigen-specific T cells induced by vaccines are more likely to persist, circumvent physiological immune tolerance, and minimize off-target effects, thereby improving overall safety [83]. Additionally, cancer vaccines may enhance the antitumor activity of ICIs by reversing the immunosuppressive TME, as demonstrated with certain immunomodulatory vaccines [27].
However, several challenges remain in optimizing vaccine efficacy, as the selection of the most immunogenic and tumor-specific tumor antigens remains a key hurdle, as do the choices of biomaterials and delivery methods: there are several ongoing trials exploring this issue (Table 2). The ideal delivery platform should maximize immune activation while ensuring safety and tolerability. In addition, reliable radiological and biochemical markers for evaluating objective response rates and the proportion of activated effector T cells in combination therapies (ICIs plus vaccines) are still underdeveloped [16]. Importantly, host factors, including sex, age, and the gut microbiome, can contribute to modulate cancer vaccines effectiveness. None of these have been adequately characterized, and no biomarkers for vaccine efficacy have been formally validated. Therefore, future studies regarding patients’ features are needed to elaborate scores to predict the effectiveness or failure of cancer vaccines in advanced tumor settings [7].
Addressing these challenges is essential for translating these innovative therapies into effective clinical solutions for patients with hard-to-treat cancers.

Author Contributions

S.C., V.M. conceptualization, writing; L.C., methodology; L.C., G.I., M.P., L.S., A.G. and F.R.P., review and editing, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Thanks to Fondazione Roma for their invaluable support for scientific research—FRCEMAD 21-25.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript: checkpoint inhibitors (ICIs), adoptive cell therapy (ACT), tumor microenvironment (TME), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), antigen-presenting cells (APCs), major histocompatibility complex (MHC), dendritic cells (DCs), cytotoxic T lymphocytes (CTLs), tumor-associated M2 macrophages (TAMs), anti-regulatory T cells (anti-Tregs), natural killer (NK), prostate acid phosphatase (PAP), Food and Drug Administration (FDA), tumor-infiltrating lymphocytes (TILs), tumor-specific antigens (TSAs), tumor mutation burden (TMB), interferon (IFN), oncolytic viruses (OVs), Talimogene laherparepvec (T-Vec), herpes simplex virus type 1 (HSV-1), prostate-specific antigen (PSA), oncolytic myxoma virus (vMyx), measles virus (MeV), hepatocellular carcinoma (HCC), cyclic GMP-AMP (cGAMP), microRNAs (miRNAs), antigen self-presentation and immunosuppression reverse (ASPIRE), immune-modulatory vaccines (IMVs), cancer-associated fibroblasts (CAFs), indoleamine 2,3-dioxygenase (IDO), granulocyte–macrophage colony-stimulating factor (GM-CSF), human telomerase reverse transcriptase (hTERT), T cell receptor (TCR), tumor-associated antigen (TAA), messenger RNA (mRNA).

References

  1. Abakushina, E.V.; Popova, L.I.; Zamyatnin, A.A.; Werner, J.; Mikhailovsky, N.V.; Bazhin, A.V. The Advantages and Challenges of Anticancer Dendritic Cell Vaccines and NK Cells in Adoptive Cell Immunotherapy. Vaccines 2021, 9, 1363. [Google Scholar] [CrossRef] [PubMed]
  2. Fujiwara, Y.; Mittra, A.; Naqash, A.R.; Takebe, N. A review of mechanisms of resistance to immune checkpoint inhibitors and potential strategies for therapy. Cancer Drug Resist. 2020, 3, 252. [Google Scholar] [CrossRef] [PubMed]
  3. Nikfarjam, S.; Rezaie, J.; Kashanchi, F.; Jafari, R. Dexosomes as a cell-free vaccine for cancer immunotherapy. J. Exp. Clin. Cancer Res. 2020, 39, 258. [Google Scholar] [CrossRef] [PubMed]
  4. Lyu, C.; Sun, H.; Sun, Z.; Liu, Y.; Wang, Q. Roles of exosomes in immunotherapy for solid cancers. Cell Death Dis. 2024, 15, 106. [Google Scholar] [CrossRef]
  5. Saxena, M.; Van Der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 2021, 21, 360–378. [Google Scholar] [CrossRef]
  6. Han, J.; Zhang, B.; Zheng, S.; Jiang, Y.; Zhang, X.; Mao, K. The Progress and Prospects of Immune Cell Therapy for the Treatment of Cancer. Cell Transplant. 2024, 33, 09636897241231892. [Google Scholar] [CrossRef]
  7. Ninmer, E.K.; Xu, F.; Slingluff, C.L. The Landmark Series: Cancer Vaccines for Solid Tumors. Ann. Surg. Oncol. 2025, 32, 1443–1452. [Google Scholar] [CrossRef]
  8. Hollingsworth, R.E.; Jansen, K. Turning the corner on therapeutic cancer vaccines. npj Vaccines 2019, 4, 7. [Google Scholar] [CrossRef]
  9. 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]
  10. Rosenberg, S.A.; Sherry, R.M.; Morton, K.E.; Scharfman, W.J.; Yang, J.C.; Topalian, S.L.; Royal, R.E.; Kammula, U.; Restifo, N.P.; Hughes, M.S.; et al. Tumor Progression Can Occur despite the Induction of Very High Levels of Self/Tumor Antigen-Specific CD8+ T Cells in Patients with Melanoma. J. Immunol. 2005, 175, 6169–6176. [Google Scholar] [CrossRef]
  11. Mittendorf, E.A.; Lu, B.; Melisko, M.; Price Hiller, J.; Bondarenko, I.; Brunt, A.M.; Sergii, G.; Petrakova, K.; Peoples, G.E. Efficacy and Safety Analysis of Nelipepimut-S Vaccine to Prevent Breast Cancer Recurrence: A Randomized, Multicenter, Phase III Clinical Trial. Clin. Cancer Res. 2019, 25, 4248–4254. [Google Scholar] [CrossRef] [PubMed]
  12. Dreno, B.; Thompson, J.F.; Smithers, B.M.; Santinami, M.; Jouary, T.; Gutzmer, R.; Levchenko, E.; Rutkowski, P.; Grob, J.-J.; Korovin, S.; et al. MAGE-A3 immunotherapeutic as adjuvant therapy for patients with resected, MAGE-A3-positive, stage III melanoma (DERMA): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2018, 19, 916–929. [Google Scholar] [CrossRef] [PubMed]
  13. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef] [PubMed]
  14. Sobhani, N.; Scaggiante, B.; Morris, R.; Chai, D.; Catalano, M.; Tardiel-Cyril, D.R.; Neeli, P.; Roviello, G.; Mondani, G.; Li, Y. Therapeutic cancer vaccines: From biological mechanisms and engineering to ongoing clinical trials. Cancer Treat. Rev. 2022, 109, 102429. [Google Scholar] [CrossRef]
  15. Leko, V.; Rosenberg, S.A. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020, 38, 454–472. [Google Scholar] [CrossRef]
  16. Wu, D.-W.; Jia, S.-P.; Xing, S.-J.; Ma, H.; Wang, X.; Tang, Q.-Y.; Li, Z.-W.; Wu, Q.; Bai, M.; Zhang, X.-Y.; et al. Personalized neoantigen cancer vaccines: Current progression, challenges and a bright future. Clin. Exp. Med. 2024, 24, 229. [Google Scholar] [CrossRef]
  17. Bastin, D.J.; Montroy, J.; Kennedy, M.A.; Martel, A.B.; Shorr, R.; Ghiasi, M.; Boucher, D.M.; Wong, B.; Gresham, L.; Diallo, J.-S.; et al. Safety and efficacy of autologous cell vaccines in solid tumors: A systematic review and meta-analysis of randomized control trials. Sci. Rep. 2023, 13, 3347. [Google Scholar] [CrossRef]
  18. Bo, Y.; Wang, H. Biomaterial-Based In Situ Cancer Vaccines. Adv. Mater. 2024, 36, 2210452. [Google Scholar] [CrossRef]
  19. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
  20. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
  21. Hawlina, S.; Chowdhury, H.H.; Smrkolj, T.; Zorec, R. Dendritic cell-based vaccine prolongs survival and time to next therapy independently of the vaccine cell number. Biol. Direct 2022, 17, 5. [Google Scholar] [CrossRef] [PubMed]
  22. Speiser, D.E.; Chijioke, O.; Schaeuble, K.; Münz, C. CD4+ T cells in cancer. Nat. Cancer 2023, 4, 317–329. [Google Scholar] [CrossRef] [PubMed]
  23. Kruse, B.; Buzzai, A.C.; Shridhar, N.; Braun, A.D.; Gellert, S.; Knauth, K.; Pozniak, J.; Peters, J.; Dittmann, P.; Mengoni, M.; et al. CD4+ T cell-induced inflammatory cell death controls immune-evasive tumours. Nature 2023, 618, 1033–1040. [Google Scholar] [CrossRef] [PubMed]
  24. Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
  25. 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]
  26. Rappaport, A.R.; Kyi, C.; Lane, M.; Hart, M.G.; Johnson, M.L.; Henick, B.S.; Liao, C.-Y.; Mahipal, A.; Shergill, A.; Spira, A.I.; et al. A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: Phase 1 trial interim results. Nat. Med. 2024, 30, 1013–1022. [Google Scholar] [CrossRef]
  27. Strum, S.; Andersen, M.H.; Svane, I.M.; Siu, L.L.; Weber, J.S. State-Of-The-Art Advancements on Cancer Vaccines and Biomarkers. Am. Soc. Clin. Oncol. Educ. Book 2024, 44, e438592. [Google Scholar] [CrossRef]
  28. Thomas, S.; Prendergast, G.C. Cancer Vaccines: A Brief Overview. In Vaccine Design; Thomas, S., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2016; Volume 1403, pp. 755–761. [Google Scholar]
  29. Diao, L.; Meibohm, B. Pharmacokinetics and Pharmacokinetic–Pharmacodynamic Correlations of Therapeutic Peptides. Clin. Pharmacokinet. 2013, 52, 855–868. [Google Scholar] [CrossRef]
  30. Kim, S.C.; Sekhon, S.S.; Shin, W.-R.; Ahn, G.; Cho, B.-K.; Ahn, J.-Y.; Kim, Y.-H. Modifications of mRNA vaccine structural elements for improving mRNA stability and translation efficiency. Mol. Cell. Toxicol. 2022, 18, 1–8. [Google Scholar] [CrossRef]
  31. 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]
  32. Shalhout, S.Z.; Miller, D.M.; Emerick, K.S.; Kaufman, H.L. Therapy with oncolytic viruses: Progress and challenges. Nat. Rev. Clin. Oncol. 2023, 20, 160–177. [Google Scholar] [CrossRef] [PubMed]
  33. Kuai, R.; Ochyl, L.J.; Bahjat, K.S.; Schwendeman, A.; Moon, J.J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 2017, 16, 489–496. [Google Scholar] [CrossRef] [PubMed]
  34. Kjeldsen, J.W.; Lorentzen, C.L.; Martinenaite, E.; Ellebaek, E.; Donia, M.; Holmstroem, R.B.; Klausen, T.W.; Madsen, C.O.; Ahmed, S.M.; Weis-Banke, S.E.; et al. Author Correction: A phase 1/2 trial of an immune-modulatory vaccine against IDO/PD-L1 in combination with nivolumab in metastatic melanoma. Nat. Med. 2022, 28, 871. [Google Scholar] [CrossRef] [PubMed]
  35. Sondak, V.K.; Sosman, J.; Unger, J.M.; Liu, P.Y.; Thompson, J.; Tuthill, R.; Kempf, R.; Flaherty, L. Significant impact of HLA class I allele expression on outcome in melanoma patients treated with an allogeneic melanoma cell lysate vaccine. Final analysis of SWOG-9035. J. Clin. Oncol. 2004, 22, 7501. [Google Scholar] [CrossRef]
  36. Morton, D.L.; Mozzillo, N.; Thompson, J.F.; Kelley, M.C.; Faries, M.; Wagner, J.; Schneebaum, S.; Schuchter, L.; Gammon, G.; Elashoff, R. An international, randomized, phase III trial of bacillus Calmette-Guerin (BCG) plus allogeneic melanoma vaccine (MCV) or placebo after complete resection of melanoma metastatic to regional or distant sites. J. Clin. Oncol. 2007, 25, 8508. [Google Scholar] [CrossRef]
  37. O’Donnell, J.S.; Madore, J.; Li, X.-Y.; Smyth, M.J. Tumor intrinsic and extrinsic immune functions of CD155. Semin. Cancer Biol. 2020, 65, 189–196. [Google Scholar] [CrossRef]
  38. Zhou, X.; Mo, X.; Qiu, J.; Zhao, J.; Wang, S.; Zhou, C.; Su, Y.; Lin, Z.; Ma, H. Chemotherapy combined with dendritic cell vaccine and cytokine-induced killer cells in the treatment of colorectal carcinoma: A meta-analysis. Cancer Manag. Res. 2018, 10, 5363–5372. [Google Scholar] [CrossRef]
  39. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef]
  40. Fong, L.; Carroll, P.; Weinberg, V.; Chan, S.; Lewis, J.; Corman, J.; Amling, C.L.; Stephenson, R.A.; Simko, J.; Sheikh, N.A.; et al. Activated Lymphocyte Recruitment Into the Tumor Microenvironment Following Preoperative Sipuleucel-T for Localized Prostate Cancer. J. Natl. Cancer Inst. 2014, 106, dju268. [Google Scholar] [CrossRef]
  41. Thara, E.; Dorff, T.B.; Pinski, J.K.; Quinn, D.I. Vaccine therapy with sipuleucel-T (Provenge) for prostate cancer. Maturitas 2011, 69, 296–303. [Google Scholar] [CrossRef]
  42. Jarosławski, S.; Toumi, M. Sipuleucel-T (Provenge®)—Autopsy of an Innovative Paradigm Change in Cancer Treatment: Why a Single-Product Biotech Company Failed to Capitalize on its Breakthrough Invention. BioDrugs 2015, 29, 301–307. [Google Scholar] [CrossRef] [PubMed]
  43. Mejías Sosa, L.; López-Janeiro, Á.; Córdoba Iturriagagoitia, A.; Sala, P.; Solans, B.P.; Hato, L.; Inogés, S.; López-Díaz De Cerio, A.; Guillén-Grima, F.; Espinós, J.; et al. Modification of Breast Cancer Milieu with Chemotherapy plus Dendritic Cell Vaccine: An Approach to Select Best Therapeutic Strategies. Biomedicines 2023, 11, 238. [Google Scholar] [CrossRef] [PubMed]
  44. Ferrall, L.; Lin, K.Y.; Roden, R.B.S.; Hung, C.-F.; Wu, T.-C. Cervical Cancer Immunotherapy: Facts and Hopes. Clin. Cancer Res. 2021, 27, 4953–4973. [Google Scholar] [CrossRef] [PubMed]
  45. Van Willigen, W.W.; Bloemendal, M.; Boers-Sonderen, M.J.; De Groot, J.W.B.; Koornstra, R.H.T.; Van Der Veldt, A.A.M.; Haanen, J.B.A.G.; Boudewijns, S.; Schreibelt, G.; Gerritsen, W.R.; et al. Response and survival of metastatic melanoma patients treated with immune checkpoint inhibition for recurrent disease on adjuvant dendritic cell vaccination. OncoImmunology 2020, 9, 1738814. [Google Scholar] [CrossRef]
  46. Tijtgat, J.; Geeraerts, X.; Boisson, A.; Stevens, L.; Vounckx, M.; Dirven, I.; Schwarze, J.K.; Raeymaeckers, S.; Forsyth, R.; Van Riet, I.; et al. Intratumoral administration of the immunologic adjuvant AS01B in combination with autologous CD1c (BDCA-1)+ /CD141 (BDCA-3)+ myeloid dendritic cells plus ipilimumab and intravenous nivolumab in patients with refractory advanced melanoma. J. Immunother. Cancer 2024, 12, e008148. [Google Scholar] [CrossRef]
  47. Schwarze, J.K.; Tijtgat, J.; Awada, G.; Cras, L.; Vasaturo, A.; Bagnall, C.; Forsyth, R.; Dufait, I.; Tuyaerts, S.; Van Riet, I.; et al. Intratumoral administration of CD1c (BDCA-1)+ and CD141 (BDCA-3)+ myeloid dendritic cells in combination with talimogene laherparepvec in immune checkpoint blockade refractory advanced melanoma patients: A phase I clinical trial. J. Immunother. Cancer 2022, 10, e005141. [Google Scholar] [CrossRef]
  48. Hato, L.; Vizcay, A.; Eguren, I.; Pérez-Gracia, J.L.; Rodríguez, J.; Gállego Pérez-Larraya, J.; Sarobe, P.; Inogés, S.; Díaz De Cerio, A.L.; Santisteban, M. Dendritic Cells in Cancer Immunology and Immunotherapy. Cancers 2024, 16, 981. [Google Scholar] [CrossRef]
  49. Arakawa, Y.; Okita, Y.; Narita, Y. Efficacy finding cohort of a cancer peptide vaccine, TAS0313, in treating recurrent glioblastoma. J. Clin. Oncol. 2021, 39, 2038. [Google Scholar] [CrossRef]
  50. Cleyle, J.; Hardy, M.-P.; Minati, R.; Courcelles, M.; Durette, C.; Lanoix, J.; Laverdure, J.-P.; Vincent, K.; Perreault, C.; Thibault, P. Immunopeptidomic Analyses of Colorectal Cancers With and Without Microsatellite Instability. Mol. Cell. Proteomics 2022, 21, 100228. [Google Scholar] [CrossRef]
  51. Chandra, S.; Wilson, J.C.; Good, D.; Wei, M.Q. mRNA vaccines: A new era in vaccine development. Oncol. Res. 2024, 32, 1543–1564. [Google Scholar] [CrossRef]
  52. Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.-P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. Lopes, A.; Vandermeulen, G.; Préat, V. Cancer DNA vaccines: Current preclinical and clinical developments and future perspectives. J. Exp. Clin. Cancer Res. 2019, 38, 146. [Google Scholar] [CrossRef] [PubMed]
  55. Torres, N.; Regge, M.V.; Secchiari, F.; Friedrich, A.D.; Spallanzani, R.G.; Raffo Iraolagoitia, X.L.; Núñez, S.Y.; Sierra, J.M.; Ziblat, A.; Santilli, M.C.; et al. Restoration of antitumor immunity through anti-MICA antibodies elicited with a chimeric protein. J. Immunother. Cancer 2020, 8, e000233. [Google Scholar] [CrossRef] [PubMed]
  56. Ma, R.; Li, Z.; Chiocca, E.A.; Caligiuri, M.A.; Yu, J. The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer 2023, 9, 122–139. [Google Scholar] [CrossRef]
  57. Wang, G.; Kang, X.; Chen, K.S.; Jehng, T.; Jones, L.; Chen, J.; Huang, X.F.; Chen, S.-Y. An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses. Nat. Commun. 2020, 11, 1395. [Google Scholar] [CrossRef]
  58. Leber, M.F.; Neault, S.; Jirovec, E.; Barkley, R.; Said, A.; Bell, J.C.; Ungerechts, G. Engineering and combining oncolytic measles virus for cancer therapy. Cytokine Growth Factor Rev. 2020, 56, 39–48. [Google Scholar] [CrossRef]
  59. Hardcastle, J.; Mills, L.; Malo, C.S.; Jin, F.; Kurokawa, C.; Geekiyanage, H.; Schroeder, M.; Sarkaria, J.; Johnson, A.J.; Galanis, E. Immunovirotherapy with measles virus strains in combination with anti–PD-1 antibody blockade enhances antitumor activity in glioblastoma treatment. Neuro-Oncology 2016, 19, 493–502. [Google Scholar] [CrossRef]
  60. Geils, C.; Kathrein, K.L. Augmentation of Solid Tumor Immunotherapy With IL-12. J. Gene Med. 2024, 26, e70000. [Google Scholar] [CrossRef]
  61. Chen, J.; Qiu, M.; Ye, Z.; Nyalile, T.; Li, Y.; Glass, Z.; Zhao, X.; Yang, L.; Chen, J.; Xu, Q. In situ cancer vaccination using lipidoid nanoparticles. Sci. Adv. 2021, 7, eabf1244. [Google Scholar] [CrossRef]
  62. Agarwal, Y.; Milling, L.E.; Chang, J.Y.H.; Santollani, L.; Sheen, A.; Lutz, E.A.; Tabet, A.; Stinson, J.; Ni, K.; Rodrigues, K.A.; et al. Intratumourally injected alum-tethered cytokines elicit potent and safer local and systemic anticancer immunity. Nat. Biomed. Eng. 2022, 6, 129–143. [Google Scholar] [CrossRef] [PubMed]
  63. Lutz, E.A.; Agarwal, Y.; Momin, N.; Cowles, S.C.; Palmeri, J.R.; Duong, E.; Hornet, V.; Sheen, A.; Lax, B.M.; Rothschilds, A.M.; et al. Alum-anchored intratumoral retention improves the tolerability and antitumor efficacy of type I interferon therapies. Proc. Natl. Acad. Sci. USA 2022, 119, e2205983119. [Google Scholar] [CrossRef] [PubMed]
  64. Yu, X.; Wang, X.; Yamazaki, A.; Li, X. Tumor microenvironment-regulated nanoplatforms for the inhibition of tumor growth and metastasis in chemo-immunotherapy. J. Mater. Chem. B 2022, 10, 3637–3647. [Google Scholar] [CrossRef] [PubMed]
  65. Bystryn, J.C.; Zeleniuch-Jacquotte, A.; Oratz, R.; Shapiro, R.L.; Harris, M.N.; Roses, D.F. Double-blind trial of a polyvalent, shed-antigen, melanoma vaccine. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2001, 7, 1882–1887. [Google Scholar]
  66. Zhao, X.; Yuan, C.; Wangmo, D.; Subramanian, S. Tumor-Secreted Extracellular Vesicles Regulate T-Cell Costimulation and Can Be Manipulated To Induce Tumor-Specific T-Cell Responses. Gastroenterology 2021, 161, 560–574.e11. [Google Scholar] [CrossRef]
  67. Turiello, R.; Capone, M.; Morretta, E.; Monti, M.C.; Madonna, G.; Azzaro, R.; Del Gaudio, P.; Simeone, E.; Sorrentino, A.; Ascierto, P.A.; et al. Exosomal CD73 from serum of patients with melanoma suppresses lymphocyte functions and is associated with therapy resistance to anti-PD-1 agents. J. Immunother. Cancer 2022, 10, e004043. [Google Scholar] [CrossRef]
  68. Xu, Z.; Zeng, S.; Gong, Z.; Yan, Y. Exosome-based immunotherapy: A promising approach for cancer treatment. Mol. Cancer 2020, 19, 160. [Google Scholar] [CrossRef]
  69. Li, J.; Li, J.; Peng, Y.; Du, Y.; Yang, Z.; Qi, X. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J. Control. Release 2023, 353, 423–433. [Google Scholar] [CrossRef]
  70. Besse, B.; Charrier, M.; Lapierre, V.; Dansin, E.; Lantz, O.; Planchard, D.; Le Chevalier, T.; Livartoski, A.; Barlesi, F.; Laplanche, A.; et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. OncoImmunology 2016, 5, e1071008. [Google Scholar] [CrossRef]
  71. Liu, C.; Liu, X.; Xiang, X.; Pang, X.; Chen, S.; Zhang, Y.; Ren, E.; Zhang, L.; Liu, X.; Lv, P.; et al. A nanovaccine for antigen self-presentation and immunosuppression reversal as a personalized cancer immunotherapy strategy. Nat. Nanotechnol. 2022, 17, 531–540. [Google Scholar] [CrossRef]
  72. Escudier, B.; Dorval, T.; Chaput, N.; André, F.; Caby, M.-P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of thefirst phase I clinical trial. J. Transl. Med. 2005, 3, 10. [Google Scholar] [CrossRef] [PubMed]
  73. Perez-Penco, M.; Weis-Banke, S.E.; Schina, A.; Siersbæk, M.; Hübbe, M.L.; Jørgensen, M.A.; Lecoq, I.; Lara De La Torre, L.; Bendtsen, S.K.; Martinenaite, E.; et al. TGFβ-derived immune modulatory vaccine: Targeting the immunosuppressive and fibrotic tumor microenvironment in a murine model of pancreatic cancer. J. Immunother. Cancer 2022, 10, e005491. [Google Scholar] [CrossRef] [PubMed]
  74. Kjeldsen, J.W.; Iversen, T.Z.; Engell-Noerregaard, L.; Mellemgaard, A.; Andersen, M.H.; Svane, I.M. Durable Clinical Responses and Long-Term Follow-Up of Stage III–IV Non-Small-Cell Lung Cancer (NSCLC) Patients Treated With IDO Peptide Vaccine in a Phase I Study—A Brief Research Report. Front. Immunol. 2018, 9, 2145. [Google Scholar] [CrossRef] [PubMed]
  75. Lorentzen, C.L.; Kjeldsen, J.W.; Ehrnrooth, E.; Andersen, M.H.; Marie Svane, I. Long-term follow-up of anti-PD-1 naïve patients with metastatic melanoma treated with IDO/PD-L1 targeting peptide vaccine and nivolumab. J. Immunother. Cancer 2023, 11, e006755. [Google Scholar] [CrossRef]
  76. Pérez-Baños, A.; Gleisner, M.A.; Flores, I.; Pereda, C.; Navarrete, M.; Araya, J.P.; Navarro, G.; Quezada-Monrás, C.; Tittarelli, A.; Salazar-Onfray, F. Whole tumour cell-based vaccines: Tuning the instruments to orchestrate an optimal antitumour immune response. Br. J. Cancer 2023, 129, 572–585. [Google Scholar] [CrossRef]
  77. 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]
  78. Piper, M.; Kluger, H.; Ruppin, E.; Hu-Lieskovan, S. Immune Resistance Mechanisms and the Road to Personalized Immunotherapy. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e390290. [Google Scholar] [CrossRef]
  79. Geynisman, D.M.; Chien, C.-R.; Smieliauskas, F.; Shen, C.; Shih, Y.-C.T. Economic evaluation of therapeutic cancer vaccines and immunotherapy: A systematic review. Hum. Vaccines Immunother. 2014, 10, 3415–3424. [Google Scholar] [CrossRef]
  80. Zhao, J.; Huang, W.; Wu, Y.; Luo, Y.; Wu, B.; Cheng, J.; Chen, J.; Liu, D.; Li, C. Prognostic role of pretreatment blood lymphocyte count in patients with solid tumors: A systematic review and meta-analysis. Cancer Cell Int. 2020, 20, 15. [Google Scholar] [CrossRef]
  81. Mier, P.D.; van den Hurk, J.J. Lysosomal hydrolases of the epidermis. I. Glycosidases. Br. J. Dermatol. 1975, 93, 1–10. [Google Scholar] [CrossRef]
  82. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef]
  83. Shemesh, C.S.; Hsu, J.C.; Hosseini, I.; Shen, B.-Q.; Rotte, A.; Twomey, P.; Girish, S.; Wu, B. Personalized Cancer Vaccines: Clinical Landscape, Challenges, and Opportunities. Mol. Ther. 2021, 29, 555–570. [Google Scholar] [CrossRef]
Figure 1. Cancer vaccines and their mechanisms of action. Cancer vaccines can stimulate local host adaptive anti-tumor immune responses. Vaccines carrying tumor-associated antigens are recognized by DCs at the tumor site. Then, activated DCs migrate through the lymphatic vessels to the T-cell zones of lymphoid tissues. There, they stimulate T lymphocytes by simultaneously expressing costimulatory molecules, thus creating CTLs specific for tumor-associated antigens presented on the surfaces of cancer cells via MHC class I molecules to destroy tumor cells. Meanwhile, naïve CD4+ T cells differentiate into effector cells, initiating a B cell-mediated, TAA/TSA-specific antibody response. Abbreviations: antigen-presenting cell (APC), major histocompatibility complex (MHC), dendritic cell (DC), tumor-infiltrating lymphocyte (TIL), T cell receptor (TCR), tumor-associated antigen (TAA), messenger RNA (mRNA).
Figure 1. Cancer vaccines and their mechanisms of action. Cancer vaccines can stimulate local host adaptive anti-tumor immune responses. Vaccines carrying tumor-associated antigens are recognized by DCs at the tumor site. Then, activated DCs migrate through the lymphatic vessels to the T-cell zones of lymphoid tissues. There, they stimulate T lymphocytes by simultaneously expressing costimulatory molecules, thus creating CTLs specific for tumor-associated antigens presented on the surfaces of cancer cells via MHC class I molecules to destroy tumor cells. Meanwhile, naïve CD4+ T cells differentiate into effector cells, initiating a B cell-mediated, TAA/TSA-specific antibody response. Abbreviations: antigen-presenting cell (APC), major histocompatibility complex (MHC), dendritic cell (DC), tumor-infiltrating lymphocyte (TIL), T cell receptor (TCR), tumor-associated antigen (TAA), messenger RNA (mRNA).
Vaccines 13 00591 g001
Figure 2. Cancer vaccines can be used in combination with other immunotherapy treatments (e.g., checkpoint inhibitors, adoptive T cells and CAR-T) to overcome cancer immune evasion mechanisms, which in turn could be used to determine tumor progression despite ongoing immune treatment. As explained in the text (in the sixth paragraph), different type of cancer vaccines have been examined in clinical trials to ameliorate patients’ objective response rates in combination with other immunotherapy medications in a sort of synergistic anti-cancer therapy. Abbreviations: chimeric antigen receptor T-cell (CAR-T), immune-modulatory vaccines (IMVs).
Figure 2. Cancer vaccines can be used in combination with other immunotherapy treatments (e.g., checkpoint inhibitors, adoptive T cells and CAR-T) to overcome cancer immune evasion mechanisms, which in turn could be used to determine tumor progression despite ongoing immune treatment. As explained in the text (in the sixth paragraph), different type of cancer vaccines have been examined in clinical trials to ameliorate patients’ objective response rates in combination with other immunotherapy medications in a sort of synergistic anti-cancer therapy. Abbreviations: chimeric antigen receptor T-cell (CAR-T), immune-modulatory vaccines (IMVs).
Vaccines 13 00591 g002
Table 1. Cancer vaccines’ composition and application in clinical studies.
Table 1. Cancer vaccines’ composition and application in clinical studies.
CompositionEffectClinical ExampleTarget TumorClinical Application
Exosomes [3,4]
-
enhancing APC processing
-
reversing immunosuppressive TME
T-VecMelanomaAdjuvant to ICIs
Cell-based [7]
-
priming effector T cells
DCsProstate cancerPrimary immunotherapy alone
Protein-based [29]
-
enhancing natural killer activity
Irradiated tumor cellsMelanomaPrimary immunotherapy alone
Nucleic acid-based [30]
-
enhancing APC processing
Neoantigen vCervical cancerPrimary immunotherapy alone
Soluble molecules [31]
-
induction of anticancer antibodies
mRNAHigh tumor mutational burden cancers(ongoing trials)
Oncolytic viruses [32]
-
direct cancer cells oncolysis
-
induction of innate and adaptive immune responses
-
reversing immunosuppressive TME
KRASmelanomaAdjuvant to ACT
Nanomaterial-based [33]
-
increasing local tumor antigen release
-
stimulate local inflammation
hTERTPancreatic cancerAdjuvant to ICIs
Immune-modulatory vaccines [34]
-
reversing immunosuppressive TME
PROSTVACProstate cancerAdjuvant to ICIs
Abbreviations: dendritic cell (DC), messenger RNA (mRNA), antigen-presenting cell (APC), Kirsten Rat Sarcoma viral oncogene homolog (KRAS), adoptive cell therapy (ACT), human telomerase reverse transcriptase (hTERT), checkpoint inhibitor (ICI), talimogene laherparepvec (T-Vec).
Table 2. Future perspectives in clinical research on cancer vaccines.
Table 2. Future perspectives in clinical research on cancer vaccines.
Clinical Trial IDTarget (Peptide)IndicationPhaseStudy Completion Date
NCT03559413Patient-individualized peptideAcute lymphoblastic leukemia1, 22023
NCT04688385Multipeptide vaccineLeukemia12024
NCT05475106Neoantigen peptideDifferent type of cancer12024
NCT04842513MultipeptideGlioblastoma12025
NCT04270149ESR1 peptide VaccineBreast cancer 12026
NCT06949410Peptide VaccineBreast cancer12030
NCT06932861Personalized VaccineRhabdomyosarcomaEarly 12028
NCT06789198Peptide VaccineFibrolamellar hepatocellular carcinoma12028
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

Caridi, S.; Maccauro, V.; Cerrito, L.; Ianiro, G.; Pallozzi, M.; Stella, L.; Gasbarrini, A.; Ponziani, F.R. Cancer Vaccines: A Promising Therapeutic Strategy in Advanced Solid Tumors. Vaccines 2025, 13, 591. https://doi.org/10.3390/vaccines13060591

AMA Style

Caridi S, Maccauro V, Cerrito L, Ianiro G, Pallozzi M, Stella L, Gasbarrini A, Ponziani FR. Cancer Vaccines: A Promising Therapeutic Strategy in Advanced Solid Tumors. Vaccines. 2025; 13(6):591. https://doi.org/10.3390/vaccines13060591

Chicago/Turabian Style

Caridi, Simona, Valeria Maccauro, Lucia Cerrito, Gianluca Ianiro, Maria Pallozzi, Leonardo Stella, Antonio Gasbarrini, and Francesca Romana Ponziani. 2025. "Cancer Vaccines: A Promising Therapeutic Strategy in Advanced Solid Tumors" Vaccines 13, no. 6: 591. https://doi.org/10.3390/vaccines13060591

APA Style

Caridi, S., Maccauro, V., Cerrito, L., Ianiro, G., Pallozzi, M., Stella, L., Gasbarrini, A., & Ponziani, F. R. (2025). Cancer Vaccines: A Promising Therapeutic Strategy in Advanced Solid Tumors. Vaccines, 13(6), 591. https://doi.org/10.3390/vaccines13060591

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