v
Search
Advanced

Publications > Journals > Cancer Screening and Prevention> Article Full Text

  • OPEN ACCESS

Personalized Vaccines: Unlocking the Next Era of Medical Innovation in Cancer Immunotherapy

  • Joseph F. Murphy* 
 Author information 

Abstract

Precision medicine represents a paradigm shift in healthcare, emphasizing individualized approaches to disease prevention, diagnosis, and treatment based on a patient’s genetic, proteomic, and immunologic profile. In the field of oncology, this paradigm has gained traction, particularly with the integration of immunotherapeutic modalities. Among the most promising advancements are therapeutic cancer vaccines, which harness the body’s immune system to fight tumors more effectively. This mini-review highlights recent developments in therapeutic vaccine engineering. It also discusses key barriers to clinical translation and summarizes findings from contemporary human clinical trials evaluating personalized cancer vaccines. In addition, it evaluates the growing potential of these therapies to redefine cancer treatment.

Keywords

Cancer prevention, Immunotherapy, Adoptive cell therapy, Personalized medicine, Vaccines, Checkpoint inhibitors

Introduction

Precision medicine, often called personalized or individualized medicine, customizes treatment to a patient’s molecular and immune profile and has become central to modern oncology, particularly immuno-oncology. Personalized cancer immunotherapy is a rapidly evolving sphere of research and development that is moving toward highly individualized treatments, leveraging genetic analysis of patient tumors to develop vaccines that elicit specific immune responses against unique cancer antigens.1

The scope of precision medicine is broadening to include comprehensive strategies, incorporating advancements in early diagnostics, and even integrating traditional and Western medical practices for more effective patient care. Emerging biomarkers such as genomic, proteomic, and imaging markers, coupled with artificial intelligence (AI)-driven biomarker discovery, are transforming personalized medicine by providing detailed information to guide treatment decisions.2

Genomic markers: Comprehensive genomic profiling involves analyzing a tumor’s DNA and RNA to identify specific mutations, copy number variations, and other alterations. This information allows clinicians to match patients with specific targeted therapies, such as those targeting a particular gene mutation. It also studies how genetic variations affect drug response, helping to optimize drug efficacy and minimize adverse drug reactions. Moreover, analyzing circulating tumor DNA from blood samples can provide a non-invasive way to monitor treatment effectiveness, detect recurrence, and identify treatment resistance.

Proteomic markers: Protein analysis involves studying the large-scale set of proteins produced by an organism, which can reveal a tumor’s functional state and its response to therapies. Proteomic data can help identify different molecular subtypes of a disease, leading to more precise diagnoses and treatments. It can also identify markers of resistance to certain drugs, which can inform treatment adjustments.

Imaging markers: Radiomics, coupled with AI-powered image analysis, extracts quantitative features from medical images (computed tomography, magnetic resonance imaging, and histopathology) that are not visible to the human eye. These markers can provide detailed information about tumor heterogeneity, shape, and boundaries, which can be used for diagnosis, staging, and predicting treatment response. AI systems can automate parts of the imaging analysis process, increasing efficiency and consistency for radiologists, and also help predict treatment needs and identify patients who may benefit from specific therapies.3 These emerging biomarkers are transforming personalized medicine by providing detailed information to guide treatment decisions, helping to predict treatment response and disease heterogeneity. This mini review aimed to evaluate the role of precision medicine in oncology, with a particular focus on therapeutic cancer vaccines.

Cancer immunotherapy

Cancer is relentless because it learns to hide, bend, and reshape the immune system to survive. A useful way to frame immune evasion is the “three Cs”: camouflage (reducing antigen visibility), coercion (reprogramming immune cells), and corruption (remodeling the microenvironment to suppress immunity). This scaffolds how tumors resist therapies and why single agents often fall short, as resistance emerges when these escape routes dominate.4 Primary resistance reflects baseline non-inflamed phenotypes (few T cells, low programmed cell death protein-1/ligand (PD-1/PD-L1), antigen presentation deficits), while acquired resistance often involves adaptive upregulation of alternate checkpoints, loss of neoantigens, or IFN signaling defects after an initial response. Biomarkers tied to resistance include low T-cell infiltration, impaired antigen presentation, interferon pathway alterations, and metabolic signatures; these inform rational combinations to restore antitumor immunity. There are multiple strategies currently under investigation.5

Immune checkpoint inhibitors (ICIs): The advent of immune checkpoint inhibitors has revolutionized the field of cancer immunotherapy. ICIs that target Cytotoxic T-lymphocyte associated protein 4 (CTLA-4), PD-1, and PD-L1 have transformed care for multiple cancers by restoring antitumor T-cell activity and producing durable responses in subsets of patients.6 ICIs are monoclonal antibodies that block inhibitory immune checkpoints, releasing constraints on T cells and enabling antitumor immunity.7 Clinically approved examples include ipilimumab (anti–CTLA-4), pembrolizumab and nivolumab (anti–PD-1), and multiple anti–PD-L1 agents; these drugs produce profound and sometimes long-lasting tumor regressions but show variable overall response rates across tumor types and are limited by primary and acquired resistance and immune-related adverse events such as myocarditis.8

Adoptive cell therapies: Adoptive cell transfer comprises approaches that isolate, expand, and/or genetically modify autologous immune cells for reinfusion. TIL therapy isolates tumor-reactive lymphocytes from resected tumors, expands them ex vivo, and reinfuses them to achieve durable responses in selected patients with solid tumors. Two principal engineered T-cell strategies have been developed: chimeric antigen receptor (CAR) T cells, which recognize surface antigens in a human leukocyte antigen (HLA)-independent manner, and T-cell receptor (TCR)-engineered T cells, which recognize peptide–HLA complexes and therefore can target intracellular antigens.9

CAR T therapy: CAR T-cell therapy has achieved landmark clinical successes in hematologic malignancies, leading to multiple approvals by the U.S. Food and Drug Administration (FDA) and durable remissions in subsets of patients with B-cell malignancies and multiple myeloma; however, efficacy in solid tumors remains limited by tumor trafficking, the immunosuppressive tumor microenvironment, antigen heterogeneity, manufacturing complexity, cost, and treatment-related toxicities such as cytokine release syndrome and neurotoxicity.10

TCR-engineered T cells expand the antigenic repertoire to intracellular, HLA-presented peptides, offering opportunities to target more broadly expressed tumor antigens and essential oncogenic proteins. They can demonstrate high antigen sensitivity and physiologic signaling, which may improve persistence and tumor recognition; nevertheless, affinity enhancement and cross-reactivity can produce severe, unexpected off-target toxicities, including fatal cardiotoxicity seen in early affinity-enhanced anti–MAGE-A3 TCR trials due to titin cross-reactivity.11

Natural killer (NK) therapy: NK cells are innate lymphocytes with intrinsic antitumor activity and multiple effector mechanisms, including missing-self recognition and antibody-dependent cellular cytotoxicity. CAR NK therapies aim to combine CAR targeting with NK biology to produce off-the-shelf, allogeneic products with lower graft-versus-host risk and potentially improved safety relative to CAR T cells. CAR NK approaches promise allogeneic availability and reduced alloreactivity but face challenges including efficient gene delivery, in vivo persistence, scalable expansion, and target selection for solid tumors.12

Key limitations across immunotherapies include immune resistance mechanisms, antigen escape, limited penetration into solid tumors, immune-related toxicity, manufacturing complexity, and cost. Current research focuses on combination strategies (ICIs with radiotherapy/targeted therapy or adoptive cell transfer), improved antigen discovery, engineered safety switches and affinity tuning for TCR/CAR constructs, radiomics and other biomarkers for response prediction, and next-generation manufacturing (in vivo CAR approaches and allogeneic platforms) to expand access and reduce time to treatment.13

Vaccines

Vaccines are biological preparations that stimulate the immune system to develop active acquired immunity against infectious agents or malignant cells. Traditionally used for disease prevention, vaccines present defined antigens or antigenic material that the immune system recognizes as non-self, thereby priming adaptive humoral and cellular responses and establishing immunological memory. In oncology, the vaccine concept has been extended beyond prophylaxis to therapeutic applications that aim to induce or augment immune responses against established tumors.

Progress in vaccine engineering—especially in antigen design, delivery platforms, and adjuvant systems—has significantly enhanced the immunogenicity and specificity of these vaccines. Among the most transformative innovations is the advent of messenger RNA (mRNA) vaccine technology, which allows for rapid and scalable production of individualized vaccines encoding multiple neoantigens. mRNA vaccines have demonstrated promising immunologic and clinical responses in early-phase trials across various solid tumors.14

Conventional vaccine platforms

Common conventional vaccine platforms include live attenuated, inactivated, and toxoid vaccines, each with distinct immunological and logistical properties. Live attenuated vaccines use replication-competent but weakened organisms to elicit strong, durable immunity and often require only one or two doses for long-term protection (e.g., measles, mumps, and rubella). Inactivated vaccines contain killed pathogens and typically require multiple doses or boosters to maintain protection because they elicit weaker cellular immunity than live vaccines.1 Toxoid vaccines present inactivated bacterial toxins to induce neutralizing antibodies (classically used for diphtheria and tetanus). Modern platforms also include subunit/protein, viral vector, DNA, and mRNA vaccines, which vary in antigen presentation, safety, manufacturability, and suitability for personalization.15

Therapeutic cancer vaccines: concept and vaccine selection

Therapeutic cancer vaccines are designed to treat existing malignancies by directing the immune system against tumor antigens rather than preventing infection. Vaccine strategies target either shared tumor-associated antigens expressed across patients or tumor-specific neoantigens arising from somatic mutations unique to an individual’s tumor. Personalized pipelines typically involve tumor biopsy, next-generation sequencing, bioinformatic neoantigen prediction, antigen prioritization, and manufacture of a patient-specific vaccine product (peptide, RNA, DNA, or dendritic cell–loaded formulation). Neoantigen-targeted vaccines aim to exploit sequences not subject to central tolerance, improving the likelihood of eliciting high-avidity T-cell responses.16,17

mRNA technology and the rise of personalized vaccines

The rapid acceleration of mRNA vaccine platforms, initially driven by the COVID-19 pandemic, is now significantly impacting cancer therapeutics, offering faster manufacturing and tailored antigen targeting. mRNA vaccine platforms offer rapid, modular, and non-integrating means to encode one or multiple antigens for in vivo expression by host cells. Advantages include short developmental timelines, scalable cell-free manufacturing via in vitro transcription, and flexibility to encode personalized neoantigen repertoires. Early clinical studies and reviews demonstrate that mRNA vaccines can induce polyclonal T-cell responses and are amenable to combination with checkpoint blockade and other immunotherapies, although regulatory approval for mRNA cancer vaccines remains pending.16,18

Mechanisms limiting efficacy and strategies to overcome them

Several biological and practical barriers limit therapeutic vaccine efficacy. Tumors employ immune evasion mechanisms such as antigen loss or heterogeneity, impaired antigen presentation, upregulation of inhibitory pathways (for example, PD-L1), recruitment of immunosuppressive cells, metabolic and enzymatic suppression, and hypoxia within the tumor microenvironment, all of which can blunt vaccine-induced responses.19,20 High intratumoral heterogeneity may allow a vaccine to control the index lesion yet fail against metastatic or antigen-negative subclones. Practical constraints include complex, resource-intensive manufacturing for personalized and cell-based products, long turnaround times, and cost, which hamper broad clinical implementation.21 Current development strategies focus on improved neoantigen prediction, standardized rapid manufacturing, optimized delivery and adjuvant systems, biomarker-guided patient selection, and rational combinations with checkpoint inhibitors or therapies that modulate the tumor microenvironment.22,23

Clinical landscape and regulatory status

Despite decades of research and innovation, relatively few therapeutic cancer vaccines have achieved regulatory approval. This reflects the inherent complexity of designing immunotherapies that can overcome tumor immune evasion, elicit durable responses, and demonstrate consistent efficacy across diverse patient populations. The development timeline—from antigen discovery through preclinical validation, clinical trials, and regulatory review—often spans 10–15 years, with many candidates failing due to limited immunogenicity or safety concerns.

As of 2025, four therapeutic cancer vaccines have received FDA approval and remain active in clinical practice. These agents showcase a range of immunological strategies, from autologous cell-based therapies to oncolytic viruses and gene delivery platforms. Each has carved out a niche in oncology, offering new hope for patients with otherwise limited treatment options.

  • Sipuleucel-T: Approved for metastatic prostate cancer, it is an autologous cellular immunotherapy that stimulates an immune response against prostatic acid phosphatase, an antigen expressed by prostate cancer cells.

  • Bacillus Calmette-Guérin: Used for early-stage bladder cancer, Bacillus Calmette-Guérin works by inducing an inflammatory response within the bladder that helps eliminate cancer cells.

  • Nadofaragene firadenovec: Recently approved for early bladder cancer, this gene therapy delivers a modified adenovirus into the bladder, prompting cells to produce interferon alfa-2b, which fights cancer.

  • T-VEC (Talimogene laherparepvec): Approved for advanced melanoma, T-VEC is an oncolytic viral therapy that directly infects and lyses cancer cells, also releasing tumor antigens that stimulate an antitumor immune response.

These vaccines not only validate the concept of therapeutic immunization but also serve as platforms for future combination strategies, including checkpoint inhibitors, CAR T cells, and personalized neoantigen vaccines. Their success underscores the importance of continued investment in immunogenomics, delivery technologies, and biomarker-driven patient selection. Cancer vaccines are now becoming center stage, offering personalized treatment options for treatment-resistant cancers, with several FDA-approved and investigational vaccines showing continued promise. Some of the successes and challenges of this new treatment modality are summarized in Table 1.24–26

Table 1

Specific successes and challenges in different cancers

Cancer typeKey successesDominant challengesTailored approach signals
MelanomaStrong immunogenicity vaccines combined with PD-1 show personalized mRNA; off-the-shelf immunomodulatory randomized phase dataHeterogeneity of immune escape; durability, sequencing with ICIs, and standardization of endpointsCombination with PD 1/PD L1, next gen delivery, dual targets (e.g., IDO + PD L1)
Pancreatic cancerPersonalized mRNA vaccines induced persistent T cell responses in adjuvant settings; expanding into phase 2Low neoantigen load, immunosuppressive TME, rapid relapseAdjuvant/Minimal residual disease focus, vaccine + checkpoint inhibitors + stroma/targeted agents
Brain tumors (glioblastoma)Early human signals of immune reprogramming; diverse vaccine concepts (mRNA, cell-based)Profound immunosuppression, heterogeneity behind BBB, steroid useLocal/neoantigen-rich delivery, multimodal combinations (RT/chemo/ICI), intratumoral strategies

Emerging landscape of cancer vaccines

The landscape of cancer vaccine development has evolved significantly in recent years due to advances in target identification, formulation, manufacturing, and patient stratification, as previously reported.27 Within the last year, significant developments have been reported in vaccine engineering, personalized therapeutic strategies, and the broader integration of precision approaches across diverse medical fields. These improvements have led to a surge in clinical activity, with over 360 active trials currently investigating therapeutic cancer vaccines.23 Notably, personalized and antigen-specific approaches are demonstrating promising clinical outcomes. One example is VB10.16, a therapeutic DNA vaccine targeting human paoillomavirus (HPV) 16 E6/E7 oncoproteins. It comprises three components: a fusion protein encoding HPV16 E6/E7, a dimerization domain, and a targeting unit that binds antigen-presenting cells. In a phase I/IIa trial, VB10.16 was well tolerated and elicited robust HPV16-specific T-cell responses in patients with high-grade cervical intraepithelial neoplasia.28 Another innovative vaccine, TG4050, utilizes a modified Vaccinia virus Ankara vector to deliver patient-specific neoantigens identified through AI and genomic profiling. In early trials, TG4050 was safe, well-tolerated, and induced T-cell responses in patients with head and neck squamous cell carcinoma, particularly in immunologically “cold” tumors.

Advances in mRNA vaccine platforms

mRNA-based therapeutics have expanded beyond infectious diseases into oncology.1 Early trials have explored applications in cardiovascular and genetic disorders; however, cancer poses unique challenges due to immune suppression and antigenic heterogeneity within the tumor microenvironment.28

Moderna’s phase IIb trial of mRNA-4157/V940, a personalized mRNA vaccine encoding up to 34 neoantigens, demonstrated a 44% reduction in recurrence or death when combined with pembrolizumab in high-risk melanoma patients, regardless of tumor mutational burden.29 The vaccine was tailored to each patient’s tumor DNA and manufactured within eight weeks. Similarly, Memorial Sloan Kettering Cancer Center reported results from a phase I trial using autogene cevumeran, an mRNA vaccine targeting up to 20 neoantigens in pancreatic ductal adenocarcinoma. Patients treated with atezolizumab and cevumeran showed enhanced T-cell responses and longer recurrence-free survival compared with chemotherapy alone. Strand Therapeutics received FDA clearance for STX-001, a programmable mRNA therapy expressing IL-12. This platform promotes immunogenic cell death and recruits T and NK cells to the tumor microenvironment, enhancing checkpoint inhibitor efficacy.29

mRNA vaccines are increasingly recognized for their versatility and rapid development potential. Targeting dendritic cells, key antigen-presenting cells, has emerged as a promising strategy for enhancing vaccine potency. Clinical data suggest that mRNA vaccines act synergistically with ICIs, modulating the immune response and improving outcomes. Self-amplifying RNA is a next-generation platform offering prolonged antigen expression, lower dosing, and reduced adverse effects. Although not yet FDA-approved, Japan has authorized a self-amplifying RNA-based COVID-19 vaccine, paving the way for oncology applications.23

Beyond mRNA, novel approaches include engineered tumor cells using CRISPR-Cas9 to secrete interferon-beta and GM-CSF, which demonstrated tumor clearance and long-term immunity in glioblastoma models. Additionally, BipotentR, a computational tool, has identified 38 immune-metabolic regulators that modulate both cancer metabolism and immune response, offering new targets for immunotherapy-resistant tumors.28 Several groundbreaking cancer vaccine trials are underway, including preventive vaccines such as LungVax for lung cancer and therapeutic vaccines such as mRNA-based personalized vaccines for melanoma and breast cancer. These trials highlight both individualized and “off-the-shelf” approaches, aiming to improve survival, reduce recurrence, and expand access. Personalized cancer vaccines have progressed from small feasibility studies to multicenter phase II programs and randomized trials. A 2025 systematic review cataloged 78 ongoing cancer vaccine clinical trials, underscoring the rapid expansion of the field. These trials reflect diversification across platforms (mRNA, peptide, viral vectors, dendritic cell-based), indications (melanoma, breast, lung, colorectal, pancreatic), and combination strategies—particularly with immune checkpoint inhibitors such as PD-1/PD-L1 and CTLA-4 blockade. This convergence highlights the growing role of cancer vaccines as both standalone immunotherapies and synergistic agents within multimodal treatment regimens.30,31 Some examples of the most recent data from these trials are summarized in Table 2. Together, these approaches are reshaping oncology by combining precision immunotherapy with industrial-scale manufacturing, potentially moving cancer vaccines from niche therapies to mainstream treatment and prevention.

Table 2

Summary of clinical trial outcomes

VaccineTypeCancer targetPhaseKey outcome
Duke breast cancer vaccineTherapeuticBreast cancerLong-term follow-up24 years of disease-free survival
LungVaxPreventitiveLung cancerPhase 1Immune priming against neoantigens
IO112/IO170TherapeuticMultiple cancersPre-clinicalIND planned for 2026
mRNA-4157/V940 + KeytrudaTherapeuticMelanoma, lung, colorectal, pancreaticPhase 2b-344% reduced recurrent death
Regulatory framework

Personalized vaccines represent a novel product class, meaning existing regulatory standards are often insufficient. Developers face uncertainty, as approaches accepted by one agency may not be accepted by another, complicating a global development strategy. As of late 2024/early 2025, the FDA has approved very few personalized vaccines, with Sipuleucel-T for prostate cancer being a notable, though different-platform, example. The European Union has not yet authorized any therapeutic cancer vaccines. The regulatory landscape for personalized cancer vaccines remains complex and time-consuming, requiring robust evidence of safety, efficacy, and quality control, which is challenging to gather for highly individualized, small-batch products.32

For general vaccines such as the COVID-19 shot, the FDA has adopted an annual assessment for strain updates, similar to influenza vaccines, a model that might inform aspects of personalized medicine development. Moreover, ongoing efforts, including those by the World Health Organization, aim to establish universally applicable principles and standardized protocols for genomic data use to promote international collaboration and consistency in regulation.

The reliance on sensitive patient genomic data in personalized vaccine development raises significant ethical considerations. An individual’s genome contains highly personal information with potential for misuse by employers, insurers, or governments. Robust data governance frameworks, encryption, and compliance with privacy regulations (such as the health insurance portability and accountability actHIPAA in the U.S.) are crucial to prevent unauthorized access and discrimination. Personalized vaccines are expected to have high manufacturing costs and logistical challenges, potentially limiting access in resource-limited settings. Regulatory frameworks must incorporate strategies to ensure equitable access and prevent disparities in healthcare delivery.33

Future directions and technologies

Precision medicine extends far beyond therapeutic vaccines, encompassing diagnostic and therapeutic advancements that are shaping a more personalized approach to healthcare. The goal is to tailor medical decisions, treatments, practices, or products to the individual patient based on their predicted response or risk of disease. Early detection and accurate diagnosis are foundational to effective precision medicine. Recent advancements in screening technologies, such as improved methods for pancreatic cancer screening and updated guidelines for ovarian and breast cancer screening, are crucial.34 These diagnostic improvements allow for earlier intervention, which can significantly enhance the effectiveness of subsequent precision therapies, including therapeutic vaccines.

Recent advances in next-generation sequencing, neoantigen prediction algorithms, and nucleic acid-based platforms—particularly mRNA and DNA technologies—have revitalized interest in this approach, enabling the design of vaccines tailored to individual tumor mutational landscapes.35 These innovations, coupled with improved delivery systems such as lipid nanoparticles and viral vectors, have yielded promising results in early-phase clinical trials, demonstrating the potential to elicit robust T-cell responses against tumor-specific antigens.36

Cancer vaccines are transitioning from artisanal production toward industrialized, automated, and decentralized manufacturing models. Advances in platform-ready technologies such as mRNA, viral vectors, and peptide backbones enable rapid personalization with patient-specific antigens, accelerating timelines and improving flexibility.37 Dendritic cell vaccines have matured with standardized protocols for monocyte collection, differentiation, and antigen loading, reducing variability and streamlining release testing. Emerging “off-the-shelf” cellular vaccines, including mass-produced cDC1s, promise scalability and broader accessibility compared with individualized autologous products.

Moreover, digital and computational tools, including AI-enabled pipelines and vector optimization platforms, are compressing the vaccine lifecycle from antigen discovery through robotic manufacturing and in-line analytics, enhancing precision and reducing batch variability. Bioprocess automation—including closed systems, modular skids, and integrated digital monitoring manufacturing execution system/laboratory information management system—supports global manufacturing networks and consistent technology transfer, drawing lessons from cell and gene therapy scale-up. End-to-end digitalization and data-driven quality-by-design approaches further improve reproducibility and cost efficiency.38 Decentralized and near-patient manufacturing models reduce logistical friction associated with centralized hubs, potentially lowering costs, though they require harmonized quality assurance and regulatory oversight.

Limitations

In spite of all the recent advances, personalized immunotherapy is still limited by three main factors:

  • Data sources: Patient data is fragmented, inconsistent, and often biased toward certain populations, reducing reliability.

  • Sample size: Studies frequently involve small, non-diverse cohorts, which weakens statistical power and generalizability.

  • Follow-up duration: Outcomes are usually tracked short-term, missing late effects and making long-term safety and effectiveness uncertain.

  • These constraints hinder the accuracy, scalability, and long-term impact of personalized treatments. Moreover, other major challenges need to be addressed that include:

  • Cost-effectiveness: Prices exceed $100,000 per patient in the U.S., making them unaffordable for many. Effectiveness varies by cancer type and treatment, but predictive biomarkers can improve efficiency by targeting patients most likely to benefit.39

  • Reimbursement hurdles: High upfront costs, limited large-scale trial data, and differing payment models across countries complicate coverage. Innovative approaches like performance-based or risk-sharing agreements are being tested, but navigating regulatory bodies adds further complexity.

  • Global disparities: Access is highly unequal, with low- and middle-income countries lacking infrastructure, workforce, and representation in clinical trials. This worsens cancer outcomes, as patients are often diagnosed late and miss opportunities for advanced care.

While personalized immunotherapies hold promise for long-term savings and better outcomes, their high costs, reimbursement barriers, and unequal global access demand coordinated policy, funding, and infrastructure solutions.40

Conclusions

Precision medicine and immunotherapy have reshaped cancer treatment. While checkpoint inhibitors have revolutionized outcomes for some patients, recent advances in cancer immunotherapy have catalyzed vaccine development as a novel treatment modality, with mRNA-based vaccines offering a personalized and scalable alternative. Therapeutic cancer vaccines, a cornerstone of precision oncology, are designed to elicit targeted immune responses against tumor-specific antigens. These antigens are typically derived from the patient’s own tumor, enabling personalized vaccines that are uniquely tailored to the molecular characteristics of each malignancy. Addressing tumor heterogeneity, optimizing delivery routes, and enhancing immune activation are critical for future success. Integrating mRNA technologies with immune-stimulatory platforms is poised to transform cancer care across multiple indications.

Declarations

Acknowledgement

The author is grateful to Tara Finn for careful reading of this manuscript.

Funding

None.

Conflict of interest

JFM is the Founder and President of ImmunePCS LLC. The author declares no other conflicts of interest.

Authors’ contributions

JFM is the sole author of the manuscript.

References

  1. Murphy JF. Recent Developments of Vaccines as a Precision Medicine Approach to Cancer Immunotherapy. Cancer Screen Prev 2024;3(2):113-117 View Article
  2. Alum EU. AI-driven biomarker discovery: enhancing precision in cancer diagnosis and prognosis. Discov Oncol 2025;16(1):313 View Article PubMed/NCBI
  3. Rajendran A, Rajan RA, Balasubryamanyiam S, Elumalai K. AI-Enhanced Predictive Imaging in Precision Medicine: Advancing Diagnostic Accuracy and Personalized Treatment. iRadiology 2025;3(4):261-278 View Article
  4. Galassi C, Chan TA, Vitale I, Galluzzi L. The hallmarks of cancer immune evasion. Cancer Cell 2024;42(11):1825-1863 View Article PubMed/NCBI
  5. Alsaafeen BH, Ali BR, Elkord E. Combinational therapeutic strategies to overcome resistance to immune checkpoint inhibitors. Front Immunol 2025;16:1546717 View Article PubMed/NCBI
  6. Meng L, Wu H, Wu J, Ding P, He J, Sang M, et al. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis 2024;15(1):3 View Article PubMed/NCBI
  7. Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024;25(10):5490 View Article PubMed/NCBI
  8. Friedman CF, Proverbs-Singh TA, Postow MA. Treatment of the Immune-Related Adverse Effects of Immune Checkpoint Inhibitors: A Review. JAMA Oncol 2016;2(10):1346-1353 View Article PubMed/NCBI
  9. Shafer P, Kelly LM, Hoyos V. Cancer Therapy With TCR-Engineered T Cells: Current Strategies, Challenges, and Prospects. Front Immunol 2022;13:835762 View Article PubMed/NCBI
  10. Peng L, Sferruzza G, Yang L, Zhou L, Chen S. CAR-T and CAR-NK as cellular cancer immunotherapy for solid tumors. Cell Mol Immunol 2024;21(10):1089-1108 View Article PubMed/NCBI
  11. Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 2013;122(6):863-871 View Article PubMed/NCBI
  12. Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025;16:1574742 View Article PubMed/NCBI
  13. Haslam A, Hoeg TB, Prasad V. Estimation of eligibility for and response to CAR-T therapy in the United States. Blood Adv 2024;8(4):1032-1036 View Article PubMed/NCBI
  14. Fierro C, Sanchez-Crespo N, Makrinos D, Zhang W, Sun Y, Rohilla P, et al. Shared clinical and immunologic features of mRNA vaccines: preliminary results from a comparative clinical study. Front Immunol 2025;16:1501275 View Article PubMed/NCBI
  15. Leong KY, Tham SK, Poh CL. Revolutionizing immunization: a comprehensive review of mRNA vaccine technology and applications. Virol J 2025;22(1):71 View Article PubMed/NCBI
  16. Kaczmarek M, Poznańska J, Fechner F, Michalska N, Paszkowska S, Napierała A, et al. Cancer Vaccine Therapeutics: Limitations and Effectiveness-A Literature Review. Cells 2023;12(17):2159 View Article PubMed/NCBI
  17. Gao Y, Yang L, Li Z, Peng X, Li H. mRNA vaccines in tumor targeted therapy: mechanism, clinical application, and development trends. Biomark Res 2024;12(1):93 View Article PubMed/NCBI
  18. Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025;25(5):353-369 View Article PubMed/NCBI
  19. Sayour EJ, Boczkowski D, Mitchell DA, Nair SK. Cancer mRNA vaccines: clinical advances and future opportunities. Nat Rev Clin Oncol 2024;21(7):489-500 View Article PubMed/NCBI
  20. Viborg N, Kleine Kohlbrecher D, Rønø B. Personalized neoantigen DNA cancer vaccines: Current status and future perspectives. J Cell Immunol 2024;6(1):15-24 View Article
  21. Wu DW, Jia SP, Xing SJ, Ma HL, Wang X, Tang QY, et al. Personalized neoantigen cancer vaccines: current progression, challenges and a bright future. Clin Exp Med 2024;24(1):229 View Article PubMed/NCBI
  22. Zhou Y, Wei Y, Tian X, Wei X. Cancer vaccines: current status and future directions. J Hematol Oncol 2025;18(1):18 View Article PubMed/NCBI
  23. Kiełbowski K, Plewa P, Zadworny J, Bakinowska E, Becht R, Pawlik A. Recent Advances in the Development and Efficacy of Anti-Cancer Vaccines-A Narrative Review. Vaccines (Basel) 2025;13(3):237 View Article PubMed/NCBI
  24. Blass E, Keskin DB, Tu CR, Forman C, Vanasse A, Sax HE, et al. A multi-adjuvant personal neoantigen vaccine generates potent immunity in melanoma. Cell 2025;188(19):5125-5141.e27 View Article PubMed/NCBI
  25. Magoola M, Niazi SK. Current Progress and Future Perspectives of RNA-Based Cancer Vaccines: A 2025 Update. Cancers (Basel) 2025;17(11):1882 View Article PubMed/NCBI
  26. Karimi-Sani I, Molavi Z, Naderi S, Mirmajidi SH, Zare I, Naeimzadeh Y, et al. Personalized mRNA vaccines in glioblastoma therapy: from rational design to clinical trials. J Nanobiotechnology 2024;22(1):601 View Article PubMed/NCBI
  27. Murphy JF. What’s next for mRNA vaccines. Nov Res Sci 2021;10(1):NRS.000726
  28. Zaidi N, Jaffee EM, Yarchoan M. Recent advances in therapeutic cancer vaccines. Nat Rev Cancer 2025;25(7):517-533 View Article PubMed/NCBI
  29. Weber JS, Carlino MS, Khattak A, Meniawy T, Ansstas G, Taylor MH, 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(10427):632-644 View Article PubMed/NCBI
  30. Schlom J, Arlen PM, Gulley JL. Cancer vaccines: moving beyond current paradigms. Clin Cancer Res 2007;13(13):3776-3782 View Article PubMed/NCBI
  31. Lei W, Zhou K, Lei Y, Li Q, Zhu H. Cancer vaccines: platforms and current progress. Mol Biomed 2025;6(1):3 View Article PubMed/NCBI
  32. Montin D, Santilli V, Beni A, Costagliola G, Martire B, Mastrototaro MF, et al. Towards personalized vaccines. Front Immunol 2024;15:1436108 View Article PubMed/NCBI
  33. S D, R K. A Review of the Regulatory Challenges of Personalized Medicine. Cureus 2024;16(8):e67891 View Article PubMed/NCBI
  34. Age to Initiate Routine Breast Cancer Screening ACOG Clinical Practice Update. Obstet Gynecol 2025;145(1):e40-e44 View Article PubMed/NCBI
  35. 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(1):450 View Article PubMed/NCBI
  36. Wang C, Hao P. Therapeutic vaccie strategies in cancer: advances, challenges, and future directions. Clin Cancer Bull 2025;4(1):10 View Article
  37. Kong H. Advances in personalized cancer vaccine development: AI applications from neoantigen discovery to mRNA formulation. BioChem 2025;5(2):5 View Article
  38. Isoko K, Cordiner JL, Kis Z, Moghadam PZ. Bioprocessing 4.0: a pragmatic review and future perspectives. Digit Discov 2024;3(9):1662-1681 View Article
  39. Koleva-Kolarova R, Buchanan J, Vellekoop H, Huygens S, Versteegh M, Mölken MR, et al. Financing and Reimbursement Models for Personalised Medicine: A Systematic Review to Identify Current Models and Future Options. Appl Health Econ Health Policy 2022;20(4):501-524 View Article PubMed/NCBI
  40. Dos-Santos-Silva I, Gupta S, Orem J, Shulman LN. Global disparities in access to cancer care. Commun Med (Lond) 2022;2:31 View Article PubMed/NCBI

About this Article

Cite this article
Murphy JF. Personalized Vaccines: Unlocking the Next Era of Medical Innovation in Cancer Immunotherapy. Cancer Screen Prev. Published online: Dec 30, 2025. doi: 10.14218/CSP.2025.00023.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
October 24, 2025 November 13, 2025 December 12, 2025 December 30, 2025
DOI http://dx.doi.org/10.14218/CSP.2025.00023
  • Cancer Screening and Prevention
  • pISSN 2993-6314
  • eISSN 2835-3315
Back to Top

Personalized Vaccines: Unlocking the Next Era of Medical Innovation in Cancer Immunotherapy

Joseph F. Murphy
  • Reset Zoom
  • Download TIFF