Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (370)

Search Parameters:
Keywords = neoantigens

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 3822 KB  
Article
NeoPAIR-T: Functional Mapping of Neoantigen–TCR Pairs Using a CRISPR-Engineered Jurkat Reporter System
by Koji Nagaoka, Yukari Kobayashi and Kazuhiro Kakimi
Cells 2025, 14(22), 1789; https://doi.org/10.3390/cells14221789 - 14 Nov 2025
Abstract
Targeting mutation-derived neoantigens is a promising strategy for personalized immunotherapies. However, identifying true neoantigens and cognate T cell receptors (TCRs) remains challenging because computational prediction of neoantigen peptides is uncertain and most tumor-infiltrating lymphocytes are bystanders rather than tumor-reactive, necessitating functional validation. Here, [...] Read more.
Targeting mutation-derived neoantigens is a promising strategy for personalized immunotherapies. However, identifying true neoantigens and cognate T cell receptors (TCRs) remains challenging because computational prediction of neoantigen peptides is uncertain and most tumor-infiltrating lymphocytes are bystanders rather than tumor-reactive, necessitating functional validation. Here, we developed NeoPAIR-T (Neoantigen–TCR Pairing Assay using reporter T cells), a functional assay based on co-culture of TCR–T reporter cells and autologous antigen-presenting cells (APCs) to screen neoantigen–TCR pairs. Reporter T cells are Jurkat-derived cells engineered to express a luciferase/eGFP dual reporter, providing quantitative readouts of TCR activation, while APCs are immortalized autologous cells transfected with tandem minigenes (TMGs) encoding predicted neoantigens, bypassing peptide synthesis. NeoPAIR-T also includes TCRα-knockout with targeted knock-in of candidate TCRs at the TCRβ locus to prevent mispairing and enables parallel testing of multiple reporter T cell clones co-cultured with the same APCs for efficient identification of functional pairs. Using lung cancer samples, whole-exome and RNA sequencing predicted 63 candidate peptides assembled into three TMGs. Single-cell RNA/TCR sequencing identified eight TCR clonotypes, introduced into reporter T cells and tested in parallel. Co-culture with TMG-expressing APCs revealed two functional neoantigen–TCR pairs validated by peptide assays (EC50: 10−9.2–10−6.7 M). Collectively, NeoPAIR-T streamlines neoantigen–TCR identification for vaccine and TCR-T applications. Full article
(This article belongs to the Section Cellular Immunology)
19 pages, 1167 KB  
Review
mRNA-Based Neoantigen Vaccines in Pancreatic Ductal Adenocarcinoma (PDAC)—A Promising Avenue in Cancer Immunotherapy
by Jacek Kabut, Małgorzata Stopyra, Natalia Nafalska, Grzegorz J. Stępień, Michał Miciak, Marcin Jezierzański, Tomasz Furgoł, Krzysztof Feret and Iwona Gisterek-Grocholska
Int. J. Mol. Sci. 2025, 26(22), 10988; https://doi.org/10.3390/ijms262210988 - 13 Nov 2025
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies, with 5-year survival rates consistently below 5% despite advances in surgery, chemotherapy, and targeted therapy. Worldwide, PDAC remains highly lethal, with 458,918 new cases and 432,242 deaths in 2018—about a 94% mortality-to-incidence [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies, with 5-year survival rates consistently below 5% despite advances in surgery, chemotherapy, and targeted therapy. Worldwide, PDAC remains highly lethal, with 458,918 new cases and 432,242 deaths in 2018—about a 94% mortality-to-incidence ratio. The limited therapeutic efficacy is largely attributed to the pronounced heterogeneity of the disease, late clinical presentation, and the strongly immunosuppressive tumor microenvironment. In recent years, mRNA-based vaccines encoding patient-specific neoantigens have emerged as a promising immunotherapeutic modality. By delivering tailored antigenic sequences, these vaccines are capable of eliciting potent cytotoxic T-cell responses against tumor-restricted epitopes, thereby enhancing tumor immunogenicity while minimizing off-target effects. This review summarizes the biological rationale underlying mRNA vaccination in PDAC, recent progress in preclinical and early clinical trials, and key obstacles related to antigen selection, delivery platforms, and the immunosuppressive stroma. The potential integration of neoantigen mRNA vaccines into multimodal therapeutic strategies, including immune checkpoint inhibition and chemotherapy, is also discussed, underscoring their prospective role in overcoming resistance mechanisms and improving clinical outcomes in PDAC. However, most current data come from early-phase trials, with long-term benefits yet unproven. Definitive conclusions on efficacy and survival await results from ongoing randomized studies expected by 2028–2029. Further progress in neoantigen identification, delivery systems, and combination strategies is crucial to fully harness mRNA vaccine potential in PDAC. Full article
(This article belongs to the Special Issue Molecular Research for Cancer Immunotherapy)
Show Figures

Figure 1

20 pages, 8550 KB  
Article
Integrative Neoepitope Discovery in Glioblastoma via HLA Class I Profiling and AlphaFold2-Multimer
by Raquel Francés, Jenny Bonifacio-Mundaca, Íñigo Casafont, Christophe Desterke and Jorge Mata-Garrido
Biomedicines 2025, 13(11), 2715; https://doi.org/10.3390/biomedicines13112715 - 5 Nov 2025
Viewed by 421
Abstract
Background/Objectives: Glioblastoma multiforme (GBM) is an aggressive primary brain tumor with limited therapeutic options. Neoantigen-based immunotherapy offers a promising avenue, but its efficacy primarily depends on the ability of somatic mutations to generate immunogenic peptides effectively presented by HLA class I molecules and [...] Read more.
Background/Objectives: Glioblastoma multiforme (GBM) is an aggressive primary brain tumor with limited therapeutic options. Neoantigen-based immunotherapy offers a promising avenue, but its efficacy primarily depends on the ability of somatic mutations to generate immunogenic peptides effectively presented by HLA class I molecules and recognized by cytotoxic T cells, in concert with innate immune mechanisms such as NK-cell activation and DAMP/PAMP signaling. This study aimed to characterize the MHC-I binding diversity of peptides derived from GBM-associated somatic variants, with a particular focus on interactions involving HLA-A68:01 and HLA-B15:01 alleles. These alleles were selected based on their ethnic prevalence and potential structural compatibility with neoepitopes. Methods: Somatic missense variants from TCGA-GBM were filtered using high-confidence genomic databases, including dbSNP, COSMIC, and MANE. Neoepitope prediction was performed across multiple HLA class I alleles using binding affinity algorithms (MHCflurry2). Peptide–HLA interactions were characterized through motif analysis and anchor residue enrichment. Structural modeling of peptide–HLA complexes was conducted using ColabFold (AlphaFold2-multimer v3) to evaluate conformational stability. The population frequency of selected HLA alleles was examined through epidemiological comparisons. Results: Canonical GBM driver mutations (e.g., EGFR, TP53, PIK3R1) are recurrent and biologically relevant, although pharmacological inhibition of EGFR alone has not consistently improved patient outcomes, underscoring the complex signaling redundancy in glioblastoma. HLA-A68:01 exhibited high binding affinity and favorable motif compatibility, supporting its potential for effective neoantigen presentation. HLA-B15:01 was identified as a viable presenter for the EGFR p.Arg108Lys variant. Structural modeling confirmed stable peptide insertion into the MHC-I binding groove, with high-confidence folding and preserved interface integrity. Ethnic distribution analysis revealed varying GBM incidence across populations expressing these alleles. Conclusions: This integrative analysis identified structurally validated, immunogenically promising neoantigens derived from GBM mutations, particularly for HLA-A68:01 and HLA-B15:01. These findings support allele-informed neoepitope prioritization in personalized immunotherapy, especially for patient populations with corresponding HLA genotypes and MHC-I presentation capacity. Full article
(This article belongs to the Special Issue Advanced Research in Neuroprotection)
Show Figures

Figure 1

18 pages, 741 KB  
Review
Immunopathogenesis and Therapeutic Implications in Basal Cell Carcinoma: Current Concepts and Future Directions
by Helen C. Steel, Theresa M. Rossouw, Ronald Anderson, Lindsay Anderson, Daniel van Tonder, Teresa Smit and Bernardo Leon Rapoport
Medicina 2025, 61(11), 1914; https://doi.org/10.3390/medicina61111914 - 25 Oct 2025
Viewed by 318
Abstract
This review is focused on understanding the reasons why basal cell carcinoma (BCC), the most common, increasingly prevalent cancer, is classified as an “immune excluded” malignancy. It is, despite manifesting one of the highest tumor mutational burdens of any solid human malignancy, considered [...] Read more.
This review is focused on understanding the reasons why basal cell carcinoma (BCC), the most common, increasingly prevalent cancer, is classified as an “immune excluded” malignancy. It is, despite manifesting one of the highest tumor mutational burdens of any solid human malignancy, considered to be a biomarker of enhanced tumor immunogenicity and efficacy of tumor-targeted immunotherapy. Following a brief clinical overview, the balance of the review addresses important translational issues based on recent insights into the mechanisms underpinning immune exclusion/evasion in BCC. These include, firstly, the role of infectious agents and non-infectious potential causes of predisposition for and/or exacerbation of disease development and progression. Secondly, an overview of existing and emerging novel therapeutic strategies to ameliorate immune exclusion in BCC based on targeting several key immunosuppressive mechanisms. These are (i) inappropriate activation of the hedgehog signaling pathway (HHSP) due to formation of key driver mutations; (ii) interference with the presentation of tumor-specific antigens/neoantigens to cytotoxic T-cells; (iii) attenuation of the influx of anti-tumor natural killer cells; (iv) the recruitment and activation of immune suppressive regulatory T-cells; and (v) localized and systemic immune dysfunction achieved via elevated levels of soluble co-inhibitory immune checkpoint proteins (ICPs). The final section is focused on current and emerging pharmacologic and immune-based therapies. Full article
Show Figures

Figure 1

30 pages, 915 KB  
Review
Personalizing DNA Cancer Vaccines
by Annie A. Wu, Kaiqi Peng, Melanie Vukovich, Michelle Zhu, Yuki Lin, Arindam Bagga, TC Wu and Chien-Fu Hung
J. Pers. Med. 2025, 15(10), 474; https://doi.org/10.3390/jpm15100474 - 2 Oct 2025
Viewed by 1110
Abstract
Recent progress in tumor immunotherapy highlights the important role of the immune system in combating various cancers. Traditionally designed to protect against infectious diseases, vaccines are now being adapted to stimulate immune responses against tumor-specific neoantigens. Both preclinical studies and clinical trials have [...] Read more.
Recent progress in tumor immunotherapy highlights the important role of the immune system in combating various cancers. Traditionally designed to protect against infectious diseases, vaccines are now being adapted to stimulate immune responses against tumor-specific neoantigens. Both preclinical studies and clinical trials have explored innovative approaches for identifying neoantigens and optimizing vaccine design, advancing the field of personalized oncology. Among these, DNA-based vaccines have become a particularly attractive approach for cancer immunotherapy. This evolution has been driven by improvements in molecular biology techniques, including more precise methods for detecting tumor-specific mutations, computational tools for predicting immunogenic antigens, and novel platforms for delivering nucleic acid vaccines. Personalized DNA vaccines are typically developed through a complex, multi-step process that involves sequencing a patient’s tumor, computational analysis to identify potential targets, and custom vaccine production. In this review, we examine the use of both shared tumor antigens and individualized neoantigens in cancer vaccine development. We outline strategies for neoantigen identification that provide insights into tumor-specific alterations. Furthermore, we highlight recent advances in DNA vaccine technologies, address the current limitations facing cancer vaccines, propose strategies to overcome these challenges, and consider key clinical and technical factors for successful implementation. Full article
(This article belongs to the Special Issue Cancer Immunotherapy: Current Advancements and Future Perspectives)
Show Figures

Graphical abstract

15 pages, 434 KB  
Review
Dendritic Cell Immunotherapy for Solid Tumors: Advances in Translational Research and Clinical Application
by Mi Eun Kim and Jun Sik Lee
Curr. Issues Mol. Biol. 2025, 47(10), 806; https://doi.org/10.3390/cimb47100806 - 1 Oct 2025
Viewed by 1943
Abstract
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based [...] Read more.
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based vaccines for solid tumors. Clinical studies across melanoma, glioblastoma, prostate cancer, and non-small cell lung cancer have demonstrated safety and immunogenicity, with encouraging signals of clinical efficacy, particularly when DC vaccination is combined with immune checkpoint blockade or personalized neoantigen approaches. However, translational barriers remain, including the immunosuppressive tumor microenvironment, inefficient DC migration, and variability in manufacturing protocols. Developing solutions such as in vivo DC targeting, biomaterials-based delivery systems, high-resolution single-cell analyses, and artificial intelligence-driven epitope prediction are controlled to overcome these challenges. Together, these innovations highlight the evolving role of DC immunotherapy as a foundation of precision oncology, offering the potential to integrate personalized vaccination strategies into standard treatment paradigms for solid tumors. Therefore, in this review, we specifically focus on these advances in dendritic cell immunotherapy for solid tumors and their translational implications. Full article
(This article belongs to the Special Issue Future Challenges of Targeted Therapy of Cancers: 2nd Edition)
Show Figures

Figure 1

13 pages, 1475 KB  
Article
Long-Acting Recombinant IL-7 (rhIL-7-hyFc) Enhances the Primary and Memory Neoantigen-Specific Immune Response to Breast Cancer Personalized Cancer Vaccines
by Michael Chen, Thomas Kane, Ina Chen, Suangson Supabphol, Xiuli Zhang, Alexandra A. Wolfarth, Donghoon Choi, Lijin Li, S. Peter Goedegebuure and William E. Gillanders
Cancers 2025, 17(19), 3177; https://doi.org/10.3390/cancers17193177 - 30 Sep 2025
Viewed by 550
Abstract
Background: Personalized cancer vaccines (PCVs) are a promising form of cancer immunotherapy, capable of eliciting robust neoantigen-specific immune responses. However, cancer neoantigens are variable in terms of immunogenicity, and PCVs may be less effective when targeting weak neoantigens. Strong and durable immune responses [...] Read more.
Background: Personalized cancer vaccines (PCVs) are a promising form of cancer immunotherapy, capable of eliciting robust neoantigen-specific immune responses. However, cancer neoantigens are variable in terms of immunogenicity, and PCVs may be less effective when targeting weak neoantigens. Strong and durable immune responses are also likely to be critical for vaccine efficacy. Interleukin-7 (IL-7) is a common gamma-chain cytokine known to support T cell development and survival, and a long-acting form of recombinant human IL-7 fused with hybrid Fc (rhIL-7-hyFc) has shown potential to enhance immune responses in early-stage clinical trials. Methods: In this study, we evaluated the ability of rhIL-7-hyFc to serve as a molecular adjuvant to a DNA PCV in the E0771 murine breast cancer model. Results: We found that the combination of rhIL-7-hyFc and DNA PCV treatment prolonged neoantigen-specific CD8+ T cell responses, improved functional memory as measured based on in vivo cytotoxicity, and increased the number of neoantigen-specific tumor-infiltrating lymphocytes (TILs), resulting in improved prophylactic tumor protection and durable memory responses. Conclusions: Our findings support the potential of rhIL-7-hyFc to enhance the efficacy of PCVs and suggest clinical utility for adjuvant rhIL-7-hyFc in cancer immunotherapy. Full article
(This article belongs to the Special Issue Combination Immunotherapy for Cancer Treatment)
Show Figures

Figure 1

16 pages, 823 KB  
Review
Diverse Biological Processes Contribute to Transforming Growth Factor β-Mediated Cancer Drug Resistance
by James P. Heiserman and Rosemary J. Akhurst
Cells 2025, 14(19), 1518; https://doi.org/10.3390/cells14191518 - 28 Sep 2025
Cited by 1 | Viewed by 1286
Abstract
Therapy resistance is a major obstacle to cancer treatment, and transforming growth factor-beta (TGF-β) signaling has emerged as a major instigator across many cancer types and therapeutic regimens. Solid tumors overexpress TGF-β ligands, and canonical and non-canonical TGF-β signaling pathways drive molecular changes [...] Read more.
Therapy resistance is a major obstacle to cancer treatment, and transforming growth factor-beta (TGF-β) signaling has emerged as a major instigator across many cancer types and therapeutic regimens. Solid tumors overexpress TGF-β ligands, and canonical and non-canonical TGF-β signaling pathways drive molecular changes in most cell types within the tumor to hijack therapeutic responses. Cancer therapies further stimulate TGF-β release to potentiate this problem. Molecular mechanisms of TGF-β action supporting resistance include upregulation of drug efflux pumps, enhanced DNA Damage Repair, elaboration of stiffened extracellular matrix, and decreased neoantigen presentation. TGF-β also activates pro-survival pathways, such as epidermal growth factor receptor, B-cell lymphoma-2 expression, and AKT-mTOR signaling. TGF-β-induced epithelial-to-mesenchymal transformation leads to tumor heterogeneity and acquisition of stem-like states. In the tumor microenvironment, TGF-β induces extracellular matrix production, contractility, and secretion of immunosuppressive cytokines by cancer-associated fibroblasts that contribute to drug resistance. TGF-β also blunts cytotoxic T and NK cell activities and stimulates recruitment and differentiation of immunosuppressive cells, including T-regulatory cells, M2 macrophages, and myeloid-derived suppressor cells. The importance of TGF-β signaling in development of drug resistance cannot be understated and should be further explored mechanistically to identify novel molecular approaches and combinatorial drug dosing strategies to prevent drug-resistance. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

18 pages, 331 KB  
Review
A Review of Immunotherapy in Renal Cell Carcinoma: Current Landscape and Future Directions
by Supriya Peshin, Adit Dharia, Nagaishwarya Moka and William Paul Skelton
Cancers 2025, 17(19), 3139; https://doi.org/10.3390/cancers17193139 - 26 Sep 2025
Viewed by 1877
Abstract
Renal cell carcinoma (RCC) is a biologically diverse malignancy with a rising global incidence and a high propensity for metastasis, particularly in its clear cell subtype. While traditional treatments have centered on surgery and targeted therapies, immunotherapy has emerged as a transformative approach [...] Read more.
Renal cell carcinoma (RCC) is a biologically diverse malignancy with a rising global incidence and a high propensity for metastasis, particularly in its clear cell subtype. While traditional treatments have centered on surgery and targeted therapies, immunotherapy has emerged as a transformative approach in the management of advanced RCC. This review explores the evolution of immunotherapy in RCC, from early cytokine-based treatments to the advent of immune checkpoint inhibitors (ICIs) and their integration with tyrosine kinase inhibitors (TKIs). We detail the immunologic mechanisms underpinning these therapies, the rationale from preclinical models, and the pivotal clinical trials that redefined standard of care. Challenges such as immune resistance, tumor heterogeneity, and immune escape mechanisms are critically analyzed, highlighting tumor-intrinsic and microenvironmental factors. Lastly, we examine ongoing innovations including novel immune checkpoints, cytokine modulators, CAR-T therapies, and neoantigen-based vaccines, offering a forward-looking perspective on enhancing immunotherapeutic efficacy and personalization in RCC management. Full article
(This article belongs to the Special Issue Immune Landscape of Renal Cell Carcinoma)
28 pages, 15303 KB  
Article
Spotlight on FAM72B: Pan-Cancer Expression Profiles and Its Potential as a Prognostic and Immunotherapeutic Biomarker
by Anran Chu and Yuchan Wang
Genes 2025, 16(10), 1140; https://doi.org/10.3390/genes16101140 - 26 Sep 2025
Viewed by 597
Abstract
Background/Objectives: FAM72B (Family with sequence similarity 72 member B) is a gene whose function is not yet fully elucidated and which belongs to the FAM72 gene family. Recent studies have indicated that it is involved in the regulation of stem cell proliferation [...] Read more.
Background/Objectives: FAM72B (Family with sequence similarity 72 member B) is a gene whose function is not yet fully elucidated and which belongs to the FAM72 gene family. Recent studies have indicated that it is involved in the regulation of stem cell proliferation and DNA repair and serves as a valuable prognostic biomarker for a few types of cancer. This study aimed to systematically investigate the expression profile of FAM72B in pan-cancer, its role in the tumor immune microenvironment, and its potential as a prognostic and immunotherapeutic biomarker. Methods: Using bioinformatics tools such as SangerBox3.0, GEPIA2.0, Kaplan–Meier Plotter, and cBioPortal, we systematically analyzed the correlation of FAM72B expression levels with various cancer types, clinical pathological parameters, prognostic value, genetic mutations, genomic heterogeneity, immune checkpoint genes, immune cell infiltration levels, and single-cell-level characteristics. Results:FAM72B was found to be overexpressed in most cancers and significantly associated with poor prognosis, although it may exert a protective effect in some cancers like thymoma (THYM). Its expression level was positively correlated with tumor mutation burden (TMB), microsatellite instability (MSI), neoantigen (NEO) levels, and expression of immune checkpoint genes in most cancers, suggesting that patients with high FAM72B expression may respond better to immune checkpoint inhibitors. Moreover, FAM72B expression was significantly correlated with the infiltration levels of various immune cells in the tumor immune microenvironment across pan-cancer. Single-cell sequencing results also demonstrated a significant correlation between FAM72B and the biological functional states of multiple cancers. Conclusions:FAM72B holds promise as a potential pan-cancer prognostic biomarker and therapeutic target, providing a novel basis for the development of personalized treatment strategies. Full article
(This article belongs to the Section Bioinformatics)
Show Figures

Figure 1

17 pages, 1030 KB  
Review
Next-Generation mRNA Vaccines in Melanoma: Advances in Delivery and Combination Strategies
by Stefano Zoroddu and Luigi Bagella
Cells 2025, 14(18), 1476; https://doi.org/10.3390/cells14181476 - 22 Sep 2025
Cited by 1 | Viewed by 3291
Abstract
Messenger RNA (mRNA) vaccines have redefined cancer immunotherapy, offering unparalleled flexibility to encode tumor-specific antigens and to be adapted to individual mutational landscapes. Melanoma, with its high mutational burden and responsiveness to immune checkpoint blockade, has become the leading model for translating these [...] Read more.
Messenger RNA (mRNA) vaccines have redefined cancer immunotherapy, offering unparalleled flexibility to encode tumor-specific antigens and to be adapted to individual mutational landscapes. Melanoma, with its high mutational burden and responsiveness to immune checkpoint blockade, has become the leading model for translating these advances into clinical benefit. Recent innovations in delivery—ranging from lipid nanoparticles and polymeric carriers to biomimetic hybrids and intratumoral administration—are dismantling long-standing barriers of stability, targeting, and immunogenicity. Clinical milestones, including the randomized phase IIb KEYNOTE-942, show that adding the personalized neoantigen vaccine mRNA-4157 (V940) to pembrolizumab prolonged recurrence-free survival versus pembrolizumab alone (HR 0.561, 95% CI 0.309–1.017; 18-month RFS 79% vs. 62%), with the ASCO 3-year update reporting 2.5-year RFS 74.8% vs. 55.6% and sustained distant metastasis-free survival benefit in resected high-risk melanoma. Parallel preclinical studies highlight the potential of multifunctional platforms co-delivering cytokines or innate agonists to reshape the tumor microenvironment and achieve durable systemic immunity. As artificial intelligence drives epitope selection and modular manufacturing accelerates personalization, mRNA vaccines may have the potential to transition from adjuncts to main therapies in melanoma and beyond. Full article
Show Figures

Figure 1

19 pages, 934 KB  
Review
Individualized mRNA Vaccines in Melanoma—Where Do We Stand?
by Ioanna Gazouli, Dimitrios Bafaloukos, Christos Koutserimpas and George Samonis
Vaccines 2025, 13(9), 986; https://doi.org/10.3390/vaccines13090986 - 21 Sep 2025
Viewed by 1691
Abstract
Immunotherapy, consisting mainly of immune checkpoint inhibitors, has been successfully employed in the treatment of early and advanced-stage melanoma for more than ten years. Personalized mRNA vaccines represent the next evolutionary step, offering patients a treatment unique to them and their tumor, whilst [...] Read more.
Immunotherapy, consisting mainly of immune checkpoint inhibitors, has been successfully employed in the treatment of early and advanced-stage melanoma for more than ten years. Personalized mRNA vaccines represent the next evolutionary step, offering patients a treatment unique to them and their tumor, whilst putting recent, significant technological and scientific advances into practice. Clinical and preclinical data about mRNA vaccines are now emerging, further encouraging research and spreading enthusiasm among patients and physicians. Nonetheless, a lot remains to be discovered about mRNA vaccines’ mechanisms of action, their actual effect on the immune cells of the patient, and successful mRNA delivery to the host. Full article
Show Figures

Figure 1

17 pages, 648 KB  
Article
Somatic Mutations in DNA Mismatch Repair Genes, Mutation Rate and Neoantigen Load in Acute Lymphoblastic Leukemia
by Diana Karen Mendiola-Soto, Laura Gómez-Romero, Juan Carlos Núñez-Enríquez, Janet Flores-Lujano, Elva Jiménez-Hernández, Aurora Medina-Sansón, Vilma Carolina Bekker-Méndez, Minerva Mata-Rocha, María Luisa Pérez-Saldívar, David Aldebarán Duarte-Rodríguez, José Refugio Torres-Nava, José Gabriel Peñaloza-González, Luz Victoria Flores-Villegas, Raquel Amador-Sánchez, Martha Margarita Velázquez-Aviña, Jorge Alfonso Martín-Trejo, Laura Elizabeth Merino-Pasaye, Karina Anastacia Solís-Labastida, Rosa Martha Espinosa-Elizondo, Carlos Jhovani Pérez-Amado, Didier Ismael May-Hau, Omar Alejandro Sepúlveda-Robles, Haydee Rosas-Vargas, Juan Manuel Mejía-Aranguré and Silvia Jiménez-Moralesadd Show full author list remove Hide full author list
Pharmaceuticals 2025, 18(9), 1405; https://doi.org/10.3390/ph18091405 - 18 Sep 2025
Viewed by 785
Abstract
Background/Objectives: During cancer development, tumor cells accumulate somatic mutations, which could generate tumor-specific neoantigens. The aberrant protein can be recognized by the immune system as no-self, triggering an immune response against cells expressing this aberrant protein which could mediate tumor control or [...] Read more.
Background/Objectives: During cancer development, tumor cells accumulate somatic mutations, which could generate tumor-specific neoantigens. The aberrant protein can be recognized by the immune system as no-self, triggering an immune response against cells expressing this aberrant protein which could mediate tumor control or rejection. Since the expression of this mutated protein is exclusive to tumor cells, great efforts are being made to identify neoantigens of relevance in the development of new cancer treatment strategies. In comparison to adulthood tumors, pediatric malignancies present fewer mutations and thus fewer potential neoantigens. Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy worldwide that can be benefited by the identification of neoantigens for immunotherapy approaches, the landscape of neoantigens in ALL is not well known, therefore the aim of our study was to identify potential neoantigens in ALL pediatric patients. Methods: To identify neoantigens in ALL, whole-exome sequencing of matched tumor-normal cells from pediatric cases was performed, with these data HLA-I alleles predicted and somatic mutations identified to propose potential neoantigens based on binding affinity of mutated peptide-HLA-I. Results: We found a strong correlation between tumor mutational burden (TMB) and neoantigen load (p < 0.001) but no correlation with prognosis. Furthermore, TMB and neoantigens were greater in ALL patients with at least one mutated DNA mismatch repair gene (p < 0.001). Also, differences between B- and T-cell ALL were found but statistical significance did not remain after permutation. Conclusions: The presence of neoantigens in pediatric cases with ALL makes the neoantigen-based immunotherapy a promising new strategy for the treatment of this malignancy, especially for patients with relapse. Full article
(This article belongs to the Special Issue Immunogenomics for Drug Discovery in Leukemia)
Show Figures

Graphical abstract

13 pages, 891 KB  
Review
Advances in Non-Small Cell Lung Cancer Cellular Immunotherapy: A Progress in Dendritic Cell, T-Cell, and NK Cell Vaccines
by Mirza Masroor Ali Beg, Mohammad Aslam, Asma Ayaz, Muhammad Saeed Akhtar and Wajid Zaman
Cells 2025, 14(18), 1453; https://doi.org/10.3390/cells14181453 - 16 Sep 2025
Viewed by 1490
Abstract
Over the past decade, cellular immunotherapy has emerged as a transformative strategy for non-small cell lung cancer (NSCLC), with dendritic-cell (DC) vaccines, T-cell vaccines, and natural killer (NK)-cell therapies demonstrating distinct mechanisms and clinical potential. DC vaccines capitalize on antigen presentation to prime [...] Read more.
Over the past decade, cellular immunotherapy has emerged as a transformative strategy for non-small cell lung cancer (NSCLC), with dendritic-cell (DC) vaccines, T-cell vaccines, and natural killer (NK)-cell therapies demonstrating distinct mechanisms and clinical potential. DC vaccines capitalize on antigen presentation to prime tumor-specific T-cell responses, showing excellent safety profiles limited mainly to injection-site reactions and flu-like symptoms. While monotherapy has shown limited efficacy, combinations with checkpoint inhibitors or chemotherapy enhance immune activation and survival outcomes. Recent innovations, including neoantigen-loaded, mRNA-electroporated, and exosome-pulsed DCs, demonstrate improved immunogenicity and personalized approaches. T-cell vaccines, designed to activate cytotoxic CD8+ T-cell responses, have been tested across multiple platforms, including peptide-based (MAGE-A3), viral vector (TG4010/MUC1), and mRNA (CV9201/92) formulations. While the phase III MAGRIT trial presented no disease-free survival (DFS) benefit with adjuvant MAGE-A3 vaccination, the TG4010 vaccine improved progression-free survival (PFS; HR 0.66) and overall survival (OS; HR 0.67) in MUC1-positive NSCLC when combined with chemotherapy. Current strategies focus on personalized neoantigen vaccines and KRAS-targeted approaches (e.g., ELI-002), with ongoing phase III trials evaluating their potential in resectable NSCLC. NK-cell therapies have also shown promise, with early trials establishing the feasibility of autologous and allogeneic infusions, while engineered CAR-NK cells enhance tumor-specific targeting. Combination strategies with checkpoint inhibitors significantly improve response rates and PFS, revealing synergies between innate and adaptive immunity. Recent advances include cytokine-enhanced, memory-like NK cells to overcome immunosuppression and “off-the-shelf” products for broader clinical use. Together, these cellular immunotherapies represent a versatile and evolving frontier in NSCLC treatment, with ongoing research optimizing combinations, delivery platforms, and patient selection to maximize therapeutic benefit. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

22 pages, 7902 KB  
Article
CD63-Mediated SARS-CoV-2 RBD Fusion Neoantigen DNA Vaccine Enhances Antitumor Immune Response in a Mouse Panc02 Model via EV-Targeted Delivery
by Guang Liu, Ziqing Yuan, Ziyi Wu, Qiyv Yang, Tingbo Ding, Ker Yu and Jibin Dong
Vaccines 2025, 13(9), 977; https://doi.org/10.3390/vaccines13090977 - 16 Sep 2025
Viewed by 902
Abstract
Background: Although DNA vaccines offer a flexible platform for tumor immunotherapy, their weak immunogenicity remains a key limitation. This study aimed to improve the immunogenicity of DNA vaccines by enhancing the efficiency of tumor neoantigen delivery through extracellular vesicles (EVs), thereby promoting stronger [...] Read more.
Background: Although DNA vaccines offer a flexible platform for tumor immunotherapy, their weak immunogenicity remains a key limitation. This study aimed to improve the immunogenicity of DNA vaccines by enhancing the efficiency of tumor neoantigen delivery through extracellular vesicles (EVs), thereby promoting stronger dendritic cell (DC) activation and antitumor responses. Methods: A novel DNA vaccine (pCSP) was engineered by fusing tumor-specific neoantigens to the EV-associated protein CD63 and incorporating a SARS-CoV-2 receptor-binding domain (RBD) fragment to facilitate EV uptake by DCs. The resulting EVs were expected to carry neoantigens into the immunoproteasome for major histocompatibility complex I (MHC-I) presentation. The immunological and antitumor effects of pCSP were assessed through in vitro functional assays and in vivo experiments in a murine pancreatic cancer model. Safety was evaluated through histological and biochemical analyses. Results: In vitro, pCSP significantly promoted EV internalization by DCs by approximately twofold and enhanced their immune activation, as evidenced by elevated cytokine production. In vivo, pCSP markedly suppressed tumor growth with a decrease in volume by over 70% relative to controls, boosted CD8+ T cell responses, and increased immune infiltration into the tumor microenvironment. Safety assessments revealed that while liver/kidney function markers were within physiological ranges, mild inflammatory infiltrates were consistently observed in the lungs, indicating a localized safety concern that warrants further monitoring. Conclusions: The pCSP vaccine enhances the immunogenicity of neoantigen DNA vaccines by improving EV uptake and immune activation in DCs. These findings provide a potential strategy for improving DNA vaccine efficacy in the context of cancer immunotherapy while maintaining acceptable safety. Full article
(This article belongs to the Special Issue Advances in ImmunoTherapy of Cancer)
Show Figures

Figure 1

Back to TopTop