Cancer Vaccines for Treatment: Breaking Through Scientific Barriers in 2025

Introduction
Cancer vaccines have emerged as a transformative innovation in oncology, offering a new layer of precision in cancer treatment. Recent clinical trials demonstrate their potential, particularly personalized therapeutic vaccines, to improve outcomes across multiple tumor types.
A landmark Phase 2b trial involving a personalized mRNA vaccine (mRNA-4157/V940) for melanoma showed a 44% reduction in the risk of recurrence or death when combined with pembrolizumab, compared to pembrolizumab alone. The recurrence-free survival rate reached 79%, versus 62% with immunotherapy alone. These findings point to the powerful synergy between personalized vaccines and existing immunotherapies.
Since the 2010 FDA approval of sipuleucel-T (Provenge) for advanced prostate cancer, the cancer vaccine landscape has expanded significantly. The advent of mRNA technology, accelerated by the COVID-19 pandemic, has made rapid development and personalization of cancer vaccines increasingly feasible. By tailoring vaccines to the unique neoantigen profile of an individual’s tumor, these therapies aim to generate a targeted immune response with fewer off-target effects.
Promising developments continue across various cancers. For instance, the autogene cevumeran mRNA vaccine, designed for pancreatic ductal adenocarcinoma, has elicited strong neoantigen-specific T-cell responses, which are associated with delayed disease recurrence.
Despite these advances, several challenges remain:
- Tumor-induced immunosuppression, which can blunt vaccine efficacy
- High treatment costs, such as the ~$100,000 price tag for sipuleucel-T
- Limited accessibility, particularly in low-resource settings
This article explores the current landscape of therapeutic cancer vaccines, including:
- Vaccine classifications (e.g., peptide-based, dendritic cell, DNA/mRNA-based)
- Mechanisms of action
- Clinical integration with checkpoint inhibitors
- Scientific and logistical barriers
As research progresses, cancer vaccines are increasingly positioned to become a core component of personalized oncology, shifting treatment paradigms toward more durable, targeted responses.
Classification of Therapeutic Cancer Vaccines in 2025
As of 2025, therapeutic cancer vaccines are evolving across multiple platforms to enhance anti-tumor immune responses, particularly through the activation of T cells and, in some cases, antibody-mediated immunity. These vaccines target tumor-associated or tumor-specific antigens and can be categorized by their composition and delivery methods. Key platforms include cellular vaccines, peptide/protein-based vaccines, gene-based vaccines, and monovalent versus polyvalent strategies, each offering unique advantages in addressing tumor heterogeneity and immune escape.
Cellular Vaccines: Dendritic Cell and Tumor Lysate Approaches
Dendritic cell (DC) vaccines are among the most established cellular platforms. Sipuleucel-T, the first FDA-approved therapeutic cancer vaccine, uses autologous DCs to stimulate immune responses in prostate cancer. These vaccines function by presenting tumor antigens directly to T cells. In early-phase trials, autologous tumor lysate-pulsed DC vaccines for glioblastoma have shown promising results: 6 of 10 patients demonstrated strong IFN-γ-driven cytotoxicity, and 4 of 9 showed expansion of CD8+ T-cell clones targeting antigens such as MAGE-1, gp100, and HER-2.
Advancements in tumor lysate approaches now include allogeneic preparations, offering a more standardized source of tumor antigens. The TLPLDC vaccine, combining dendritic cells with autologous tumor lysate, has shown potential in reducing recurrence in patients with resected stage III/IV melanoma in Phase IIb trials. Personalized approaches are also emerging; PGV001, a neoantigen-based multi-peptide vaccine from Mount Sinai, has demonstrated both safety and early efficacy in patients at high risk of recurrence.
Peptide and Protein-Based Vaccines: NY-ESO-1, MAGE-A3
Peptide-based vaccines target specific tumor antigens like NY-ESO-1, MAGE-A3, survivin, and Wilms’ Tumor 1 (WT1). These vaccines work by stimulating T cells to recognize cancer-associated antigens presented by MHC molecules on antigen-presenting cells. The efficacy of NY-ESO-1 antigen vaccines has been tested using various formulations, including DC vaccines, peptide vaccines, protein vaccines, and viral vaccines. Though generally well-tolerated and capable of eliciting immune responses, peptide vaccines as standalone treatments have shown limited effectiveness.
Several clinical trials evaluating NY-ESO-1 recombinant protein vaccines have been conducted, although none have advanced to phase III. Researchers have redirected attention toward peptide vaccines that bypass the requirement for protein antigen expression and processing, allowing direct loading of epitopes onto MHC-I/MHC-II molecules.
Gene-Based Vaccines: DNA, RNA, and Viral Vectors
DNA vaccines, delivered as circular plasmids encoding tumor antigens, have demonstrated immunogenicity in early trials. A Phase I study of a NY-ESO-1 DNA vaccine reported CD4+ T cell responses in 93% of patients who were previously non-responsive.
mRNA vaccines have gained significant momentum due to their safety profile (non-integration into the genome), efficient manufacturing, and ability to stimulate both innate and adaptive immunity. Their flexibility makes them ideal for personalized cancer vaccines targeting patient-specific neoantigens.
Viral vector vaccines, notably those based on adenoviruses and Modified Vaccinia Ankara (MVA), remain powerful tools for delivering large or multiple transgenes. Prime-boost regimens using Great Ape Adenovirus (GAd) followed by MVA have shown robust T cell activation in multiple clinical settings.
Monovalent vs Polyvalent Vaccine Strategies
The capability of targeting multiple neoantigens simultaneously is vital in addressing tumor antigen heterogeneity. Polyvalent vaccines targeting both dominant and subdominant clones can potentially curtail tumor immune escape. For instance, BNT111, a nano-liposomal mRNA vaccine developed by BioNTech, encodes four tumor-associated antigens: NY-ESO-1, MAGE-A3, tyrosinase, and TPTE.
In contrast, monovalent vaccines target single antigens but may be less effective against heterogeneous tumors. Researchers increasingly favor polyvalent approaches that enhance the breadth of immune responses, particularly in cancers with high mutational burdens where multiple neoantigens can be targeted simultaneously.
Tumor Antigen Targeting and Personalization
The identification of appropriate tumor antigens represents a foundational step in the development of effective therapeutic cancer vaccines. The degree of antigen specificity directly impacts both vaccine efficacy and potential adverse effects, making antigen selection a decisive factor in clinical outcomes.
Tumor-Associated Antigens (TAAs) vs Tumor-Specific Antigens (TSAs)
Tumor antigens are generally classified into two categories based on their expression patterns:
- Tumor-Associated Antigens (TAAs) are overexpressed or aberrantly expressed in tumor cells but may also be present at low levels in normal tissues. These include:
- Overexpressed antigens: EGFR, hTERT, p53
- Differentiation antigens: Tyrosinase, gp100
- Cancer/testis antigens: MAGE family, NY-ESO-1
TAAs were initially favored for vaccine development due to their shared expression across patients, making them compatible with standardized, “off-the-shelf” therapies. However, because TAAs are not entirely tumor-specific, they carry a higher risk of triggering autoimmune reactions.
- Tumor-Specific Antigens (TSAs), on the other hand, are exclusive to tumor cells and absent in normal tissues (with rare exceptions like testicular expression). TSAs are typically derived from:
- Somatic mutations (neoantigens)
- Oncoviral proteins (e.g., HPV E6/E7)
- Non-coding or aberrantly translated genomic regions
Since TSAs are recognized as foreign by the immune system, they bypass central tolerance and elicit stronger, more tumor-specific T cell responses. Despite the technical complexity in identifying them, TSAs are increasingly preferred for personalized cancer vaccines due to their higher safety and immunogenicity profiles.
Neoantigen Discovery via Tumor Exome Sequencing
Modern neoantigen discovery relies predominantly on next-generation sequencing technologies. Whole-exome sequencing (WES) identifies somatic mutations by comparing tumor DNA with normal tissue DNA from the same patient. This approach targets approximately 2-3% of the genome corresponding to protein-coding regions, offering cost-effective mutation identification.
RNA sequencing (RNA-seq) complements WES by confirming whether mutated genes are actually expressed, as only expressed mutations can generate potential neoantigens. RNA-seq additionally reveals other neoantigen sources including gene fusions, alternative splicing isoforms, and RNA editing events.
After identifying mutations, computational algorithms predict which altered peptides will bind to the patient’s specific HLA molecules. These algorithms typically rank candidate neoantigens based on:
- Binding affinity to HLA molecules
- Likelihood of proteasomal processing
- Probability of T cell receptor recognition
- Expression levels of source proteins
Nevertheless, computational prediction alone presents limitations. Primarily, it provides no direct experimental evidence of the predicted neoantigens’ actual presentation on tumor cells. Moreover, tumors with low mutation burdens may yield few targetable neoantigens, potentially limiting vaccine efficacy.
Personalized Vaccine Design Using Immunopeptidomics
Immunopeptidomics is a promising approach that directly identifies peptides naturally presented on tumor cell surfaces through their MHC complexes. This technique involves immunoprecipitation of MHC-peptide complexes followed by mass spectrometry analysis of the eluted peptides.
Unlike computational predictions, immunopeptidomics provides empirical evidence of antigen presentation, thereby enabling identification of multiple antigen types including:
- Mutated neoantigens from somatic mutations
- Tumor-specific antigens from aberrantly expressed genes
- Peptides derived from non-canonical reading frames
- Post-translationally modified peptides
Recent advances include integrated computational frameworks like NeoDisc, which combines genomic, transcriptomic, and immunopeptidomic data to predict clinically relevant antigenic peptides. NeoDisc creates personalized proteome references where mutations and non-canonical expressed transcripts are annotated for downstream HLA binding prediction. Furthermore, this approach facilitates detection of potential defects in antigen presentation machinery, a key mechanism of immune evasion.
Importantly, research shows that peptides with higher coverage in immunopeptidomic databases are five times more likely to induce spontaneous CD8+ T cell responses compared to computationally predicted peptides without such evidence. This insight allows researchers to prioritize candidates most likely to generate robust immune responses in therapeutic cancer vaccines.
Clinical Integration and FDA-Approved Cancer Vaccines
Currently, only two therapeutic vaccines for cancer have received FDA approval, highlighting the challenges in transitioning promising research into standard clinical practice.
Sipuleucel-T for Prostate Cancer: Mechanism and Limitations
Approved in 2010, sipuleucel-T is an autologous cellular immunotherapy for men with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). It involves harvesting the patient’s peripheral blood mononuclear cells via leukapheresis and culturing them with a fusion protein composed of prostatic acid phosphatase (PAP) and GM-CSF. These activated antigen-presenting cells are then reinfused to stimulate an anti-tumor immune response.
In the pivotal IMPACT phase III trial (n=512), sipuleucel-T improved median overall survival to 25.8 months compared to 21.7 months with placebo (HR 0.78; 95% CI: 0.61–0.98; p = 0.03). At 36 months, survival was 31.7% in the treatment group versus 23.0% in controls. Notably, this survival benefit occurred without significant changes in time to progression, highlighting a key limitation in using conventional endpoints like tumor shrinkage to evaluate immunotherapies.
Despite its clinical benefit, sipuleucel-T faces practical barriers: high cost (~$93,000 for the full regimen), complex manufacturing requiring personalized cell processing, and relatively modest response rates.
BCG Vaccine in Bladder Cancer: Historical Success
First reported in 1976 and FDA-approved in 1990, Bacillus Calmette-Guérin (BCG) represents the earliest successful cancer immunotherapy. Originally developed as a tuberculosis vaccine, this attenuated strain of Mycobacterium bovis has become standard treatment for high-risk non-muscle-invasive bladder cancer following transurethral resection.
The treatment protocol involves six weekly intravesical instillations of 5×10^8 colony-forming units. Importantly, maintenance therapy with three weekly treatments at specific intervals (3, 6, 12, 18, 24, 30, and 36 months) substantially reduces recurrence rates. A multicenter randomized trial demonstrated 5-year disease-free survival of 45% for BCG recipients versus 18% for patients receiving doxorubicin.
Unlike previous assumptions, recent research reveals BCG’s efficacy stems from immune responses against tumor neoantigens rather than against BCG antigens themselves. This insight parallels modern checkpoint inhibitor mechanisms.
Checkpoint Inhibitor Combinations with Cancer Vaccines
The clinical experience with sipuleucel-T and BCG has paved the way for vaccine–checkpoint inhibitor combinations, designed to enhance anti-tumor responses. Vaccines can prime and expand tumor-specific T cells, while checkpoint inhibitors such as ipilimumab or atezolizumab help sustain their activity within the immunosuppressive tumor microenvironment.
Ongoing trials are evaluating combinations of sipuleucel-T with checkpoint inhibitors, aiming to improve efficacy without significantly increasing toxicity. Early data suggest manageable safety profiles, although definitive benefits remain under investigation.
Scientific Barriers in Vaccine Delivery and Efficacy
Although therapeutic cancer vaccines have shown encouraging results in clinical trials, several scientific and practical challenges continue to limit their broader clinical use. These obstacles span biological complexity, technical limitations, manufacturing constraints, and patient-specific factors; all of which must be addressed to improve their real-world effectiveness and scalability.
Immunosuppressive Tumor Microenvironment
The tumor microenvironment (TME) actively suppresses anti-tumor immune responses through multiple mechanisms. Cancer cells produce immunosuppressive cytokines like TGF-β and IL-10 that inhibit T-cell activation and function. The TME contains regulatory T cells (Tregs) and myeloid-derived suppressor cells that create a hostile environment for effector T cells. Accordingly, some tumors downregulate expression of MHC molecules, further hindering antigen presentation. Recent findings show that the dynamic interactions between immune cells and tumor cells within the TME directly impact vaccine efficacy.
Low Immunogenicity in Solid Tumors
Solid tumors tend to be poorly immunogenic due to immune tolerance that develops over time. Even when neoantigens are identified, only 15–30% trigger a measurable CD8+ T-cell response, and these responses are often weak. Technical hurdles in tumor sequencing and neoantigen prediction further limit the ability to identify targetable antigens. Many potential neoantigens are either missed during analysis or fail to bind effectively to MHC molecules, reducing their utility in vaccine development.
Cost and Manufacturing Challenges in Personalized Vaccines
Personalized cancer vaccines involve a multi-step production process—starting with a tumor biopsy, followed by genomic sequencing, bioinformatic analysis, and vaccine formulation. This process typically takes 7–16 weeks, which may be too slow for patients with aggressive disease. The personalized nature of these vaccines demands small-batch manufacturing with strict quality control, driving up costs. For example, the FDA-approved vaccine sipuleucel-T costs nearly $93,000 per treatment course, highlighting the economic barriers to widespread use.
Patient Selection and Tumor Burden Considerations
Clinical evidence suggests that therapeutic cancer vaccines show greater efficacy in patients with lower tumor burden or early-stage disease. Primarily, higher tumor burden correlates with increased regulatory T cells and immunosuppressive factors. Multiple prior chemotherapy regimens also negatively impact vaccine response. Therefore, the ideal candidate has slow-growing and/or low-volume disease with minimal prior chemotherapy exposure. Immunotherapy or chemotherapy can substantially suppress antibody responses to vaccines, with anthracycline-based regimens showing the lowest GMR of 0.004 compared to healthy controls.
Ongoing Trials and Breakthroughs in 2025
The cancer vaccine landscape in 2025 has seen unprecedented growth, with multiple promising candidates showing strong clinical efficacy and addressing long-standing limitations. These advances span various tumor types and reflect a shift toward personalized, immune-driven treatment strategies.
mRNA-4157/V940 in Melanoma: Phase 2b Results
Long-term data from the KEYNOTE-942 trial reveals sustained benefits of mRNA-4157/V940 with pembrolizumab in high-risk melanoma. At 34.9 months median follow-up, this combination reduced recurrence or death risk by 49% compared to pembrolizumab alone. Likewise, the risk of distant metastasis decreased by 62%, with 2.5-year recurrence-free survival reaching 74.8% versus 55.6% for monotherapy. Importantly, the exploratory overall survival endpoint showed 96.0% survival at 2.5 years compared to 90.2% with pembrolizumab monotherapy. Based on these results, phase 3 trials have launched across multiple tumor types, including renal cell carcinoma and non-small cell lung cancer.
Autogene Cevumeran in Pancreatic Cancer
In pancreatic ductal adenocarcinoma, autogene cevumeran, an individualized neoantigen vaccine using uridine mRNA-lipoplex nanoparticles, has demonstrated extraordinary durability. Among 16 patients, vaccine-induced T cells persisted up to four years post-treatment. Furthermore, of eight immune responders, six remained cancer-free at three-year follow-up. These vaccine-expanded T cells maintained anti-cancer functionality even after chemotherapy. A global phase 2 trial enrolling approximately 260 patients is presently underway to confirm these findings.
Combination Trials with Checkpoint Inhibitors and Radiation
Ongoing trials increasingly pair cancer vaccines with established treatment modalities. For instance, personalized neoantigen vaccines for kidney cancer combined with checkpoint inhibitors have shown complete anti-cancer immune responses in all nine treated patients with stage III/IV clear cell renal cell carcinoma. Similarly, PGV001, a multi-peptide neoantigen vaccine, is being evaluated with immune checkpoint inhibitors in urothelial cancer.
Emerging Trials in Breast, Lung, and Colorectal Cancers
BNT116, a non-personalized mRNA vaccine targeting six tumor markers in non-small cell lung cancer, is undergoing phase 1 evaluation across seven countries. Concurrently, a KRAS-targeted peptide vaccine for pancreatic and colorectal cancers achieved T-cell responses in 84% of participants, with median overall survival reaching 28.9 months. At least 26 phase II/III trials testing various cancer vaccine approaches are actively recruiting patients in 2025.
Conclusion
Therapeutic cancer vaccines have undergone remarkable evolution since the first FDA approvals, transforming from limited applications to sophisticated personalized treatments. The convergence of next-generation sequencing, computational biology, and immunology has consequently accelerated development of highly targeted vaccines with unprecedented specificity. Evidence from clinical trials demonstrates that cancer vaccines no longer represent theoretical constructs but rather practical therapeutic options with measurable benefits across multiple tumor types.
Despite these advances, challenges remain. The immunosuppressive tumor microenvironment still hampers vaccine effectiveness. Manufacturing complexity and high costs, especially for personalized vaccines, limit scalability and accessibility. However, these barriers are gradually becoming more manageable with the maturation of technology and improvements in production processes.
Recent breakthroughs deserve special attention. The mRNA-4157/V940 melanoma vaccine’s 49% reduction in recurrence risk highlights the potential of mRNA platforms when combined with checkpoint inhibitors. Likewise, autogene cevumeran’s durable responses in pancreatic cancer patients—a malignancy traditionally resistant to immunotherapy—underscore the transformative potential of personalized approaches. These successes reflect a fundamental shift in cancer vaccine development: from generic targets toward precisely defined neoantigens unique to each patient’s tumor.
Future directions will undoubtedly focus on overcoming remaining obstacles through novel delivery systems, improved antigen selection, and strategic combinations with established therapies. The identification of biomarkers predicting vaccine response will certainly help optimize patient selection, while manufacturing innovations should gradually reduce costs and production timelines. Combination strategies pairing vaccines with checkpoint inhibitors, chemotherapy, or radiation therapy offer particularly promising avenues for enhancing efficacy.
The scientific community stands at a pivotal moment in cancer immunotherapy. Though challenges remain, therapeutic cancer vaccines have clearly transcended previous limitations to emerge as vital components of comprehensive cancer treatment strategies. Therefore, as ongoing trials validate preliminary findings and new platforms enter clinical testing, cancer vaccines will likely become standard therapeutic options for an expanding range of malignancies, fundamentally altering treatment paradigms and improving patient outcomes.
Frequently Asked Questions:
FAQs
Q1. Are cancer vaccines expected to be available by 2025? Cancer vaccines are showing promising results in clinical trials, with some breakthroughs expected by 2025. While not all cancers will have vaccines, significant progress is being made in developing personalized and targeted vaccines for various cancer types.
Q2. What are the most promising cancer vaccine developments? Recent breakthroughs include mRNA-4157/V940 for melanoma, which showed a 49% reduction in recurrence risk when combined with immunotherapy. Another promising candidate is autogene cevumeran for pancreatic cancer, demonstrating durable responses in clinical trials.
Q3. How do therapeutic cancer vaccines work? Therapeutic cancer vaccines stimulate the immune system to recognize and attack cancer cells. They can be made from tumor cells, proteins, peptides, or genetic material that teaches the immune system to target specific cancer antigens.
Q4. What are the main challenges in developing effective cancer vaccines? Key challenges include overcoming the immunosuppressive tumor microenvironment, improving vaccine efficacy in solid tumors, reducing manufacturing costs for personalized vaccines, and optimizing patient selection for treatment.
Q5. Are cancer vaccines used alone or in combination with other treatments? Cancer vaccines are increasingly being tested in combination with other treatments, particularly checkpoint inhibitors. These combinations aim to enhance the immune response against cancer cells and improve overall treatment efficacy.
References:
[1] – https://pmc.ncbi.nlm.nih.gov/articles/PMC4159742/
[2] – https://www.sciencedirect.com/science/article/pii/S0264410X2101046X
[3] – https://www.sciencedirect.com/science/article/pii/S1044532323000416
[4] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10486481/
[5] – https://letswinpc.org/research/mrna-vaccine-effective-small-trial/
[6] – https://www.sciencedirect.com/science/article/pii/S1359644623002891
[7] – https://www.thelancet.com/journals/eclinm/article/PIIS2589-5370(22)00338-8/fulltext
[8] – https://jhoonline.biomedcentral.com/articles/10.1186/s13045-025-01670-w
[9] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7352969/
[10] – https://blog.crownbio.com/targeting-tumor-associated-antigens-and-tumor-specific-antigens
[11] – https://www.nature.com/articles/s41587-024-02420-y
[12] – https://www.merckmanuals.com/professional/hematology-and-oncology/tumor-immunology/tumor-antigens
[13] – https://pmc.ncbi.nlm.nih.gov/articles/PMC7600129/
[14] – https://www.nature.com/articles/s41392-022-01270-x
[15] – https://jitc.bmj.com/content/4/1/22
[16] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8989416/
[17] – https://www.science.org/doi/10.1126/sciadv.adv6445
[18] – https://www.ludwigcancerresearch.org/news-releases/an-ai-powered-pipeline-for-personalized-cancer-vaccines/
[19] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8262093/
[20] – https://pmc.ncbi.nlm.nih.gov/articles/PMC6361626/
[21] – https://aacrjournals.org/clincancerres/article/17/11/3520/12151/PROVENGE-Sipuleucel-T-in-Prostate-Cancer-The-First
[22] – https://pubmed.ncbi.nlm.nih.gov/34417051/
[23] – https://www.mskcc.org/news/oldest-approved-immunotherapy-gets-new-explanation
[24] – https://academic.oup.com/jnci/article/112/6/562/5799082
[25] – https://pmc.ncbi.nlm.nih.gov/articles/PMC3447980/
[26] – https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.23273
[27] – https://www.mdpi.com/2076-393X/12/12/1341
[28] – https://www.nature.com/articles/s41392-023-01674-3
[29] – https://www.thelancet.com/journals/lanonc/article/PIIS1470-2045(25)00096-8/fulltext
[30] – https://investors.modernatx.com/news/news-details/2024/Moderna–Merck-Announce-3-Year-Data-For-mRNA-4157-V940-in-Combination-With-KEYTRUDAR-pembrolizumab-Demonstrated-Sustained-Improvement-in-Recurrence-Free-Survival–Distant-Metastasis-Free-Survival-Versus-KEYTRUDA-in-Patients-With-High-Risk-Stage-IIIIV/default.aspx
[31] – https://www.mskcc.org/news/can-mrna-vaccines-fight-pancreatic-cancer-msk-clinical-researchers-are-trying-find-out
[32] – https://www.dana-farber.org/newsroom/news-releases/2025/cancer-vaccine-shows-promise-for-patients-with-stage-iii-and-iv-kidney-cancer
[33] – https://www.mountsinai.org/about/newsroom/2025/personalized-cancer-vaccine-proves-promising-in-a-phase-1-trial-at-mount-sinai
[34] – https://medicine.uky.edu/news/markey-home-two-breakthrough-therapeutic-cancer-2025-01-29t09-11-19
[35] – https://www.cancertodaymag.org/cancer-talk/cancer-vaccines-show-promise-in-early-trials/
[36] – https://aacrjournals.org/cancerres/article/64/14/4973/511479/Vaccination-with-Tumor-Lysate-Pulsed-Dendritic
[37] – https://www.sciencedirect.com/science/article/pii/S2468294224000558
[38] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10508181/
[39] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8233841/
[40] – https://www.nature.com/articles/s41417-023-00587-1
[41] – https://pmc.ncbi.nlm.nih.gov/articles/PMC3207353/
[42] – https://www.mdpi.com/1422-0067/24/23/16591