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

Journals

Article Types

Countries / Regions

Search Results (121)

Search Parameters:
Keywords = cytotoxic T cell antigen 4

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 340 KB  
Communication
Six Weeks of Baker’s Yeast β-Glucan Supplementation Reveals Unique Immune Maturation mRNA Signature: Implications for Immunity?
by Brian K. McFarlin, Anyla L. Paschall, David G. Cooper, Caleb A. Class and Meredith A. McFarlin
Int. J. Mol. Sci. 2026, 27(2), 588; https://doi.org/10.3390/ijms27020588 - 6 Jan 2026
Viewed by 148
Abstract
Baker’s yeast beta-glucan (BYBG) supplementation improves various aspects of immune system function, readiness, and response. The purpose of this study was to determine if the expression of immune maturation mRNA was also changed over the course of 6 weeks of BYBG supplementation at [...] Read more.
Baker’s yeast beta-glucan (BYBG) supplementation improves various aspects of immune system function, readiness, and response. The purpose of this study was to determine if the expression of immune maturation mRNA was also changed over the course of 6 weeks of BYBG supplementation at rest. In this exploratory study, a small group of participants (N = 20) were randomized into two groups: BYBG (weeks 0–2 = 50 mg/d; 2–4 = 125 mg/d; and 4–6 = 250 mg/d) or placebo. Blood samples were collected at 0, 2, 4, and 6 weeks and analyzed for the expression of 785 mRNA (NanoString nCounter platform and Nanotube software; R v3.3.2). A total of 42 mRNAs in 21 annotated pathways (antigen presentation, apoptosis, B cell memory, cell cycle, chemokine signaling, cytotoxicity, DAP12 signaling, hypoxia response, IL-1 signaling, IL-10 signaling, MAPK signaling, myeloid immune response, NF-kB signaling, NK activity, Notch Signaling, PD1 signaling, Senescence/Quiescence, T cell checkpoint signaling, TCR signaling, TLR signaling, and TNF signaling), were significantly affected by BYBG at various time points. It is reasonable to speculate that the observed mRNA and associated pathways may underlie previously reported improvements in immune function with BYBG. Full article
(This article belongs to the Collection Feature Papers in Bioactives and Nutraceuticals)
Show Figures

Figure 1

13 pages, 260 KB  
Review
Health-Related Quality of Life in the Era of Immune Checkpoint Blockade: What Do Patient-Reported Outcomes Reveal?
by Alexandra M. Dunker, Neha Malik, Kathryn J. Krause, Emily Z. Keung, Jason B. Liu, Elise F. Nassif Haddad, Neeta Somaiah, Heather G. Lyu and Christina L. Roland
Cancers 2025, 17(24), 3917; https://doi.org/10.3390/cancers17243917 - 7 Dec 2025
Viewed by 704
Abstract
Background: Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by providing durable survival gains, but understanding their effects on patient health-related quality of life (HRQL) is critical. Methods: We performed a narrative review of cross-sectional surveys, early-phase trials, and large-scale phase II and [...] Read more.
Background: Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by providing durable survival gains, but understanding their effects on patient health-related quality of life (HRQL) is critical. Methods: We performed a narrative review of cross-sectional surveys, early-phase trials, and large-scale phase II and III randomized controlled clinical trials assessing FDA-approved ICIs, including programmed cell death protein 1 (PD-1) inhibitors, programmed death ligand 1 (PD-L1) inhibitors, and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) inhibitors, with emphasis on patient-reported HRQL. Validated HRQL instruments were summarized, and for pivotal trials, the positioning of HRQL outcomes as primary, secondary, or exploratory endpoints was taken from original protocols or primary manuscripts. Results: ICIs generally preserved or improved HRQL in patients with various malignancies compared with chemotherapy, targeted therapies, or observation. PD-1/PD-L1 inhibitors maintained global health and function and delayed symptom progression in patients with lung cancer, melanoma, and renal cell carcinoma. Regimens combining CTLA-4 blockade and PD-1/PD-L1 inhibition (e.g., nivolumab + ipilimumab, durvalumab + tremelimumab) are associated with HRQL outcomes similar or superior to those of targeted therapies. Overall, most immune-related adverse effects were short-term and did not diminish HRQL benefits. Conclusions: ICIs extend survival while preserving, and often enhancing, patient HRQL. These medications represent a shift in oncology, offering not just longer life but also better daily well-being. Continued long-term patient-reported outcome monitoring is essential to guide survivorship care in the immunotherapy era. Full article
(This article belongs to the Special Issue Immune-Related Adverse Events in Cancer Immunotherapy)
22 pages, 667 KB  
Review
The Search for Predictive Biomarkers in Response to Immune Checkpoint Inhibitors and Associated Adverse Events
by Marco Agostini, Pietro Traldi and Mahmoud Hamdan
J. Pers. Med. 2025, 15(12), 596; https://doi.org/10.3390/jpm15120596 - 3 Dec 2025
Viewed by 772
Abstract
The introduction of immune checkpoint inhibitors (ICIs) as a part of immunotherapy represented a therapeutic breakthrough in the landscape of cancer treatment. The action of these inhibitors consists of blocking certain inhibitory receptors in the immune system. Blocking these inhibitory pathways, ICIs induce [...] Read more.
The introduction of immune checkpoint inhibitors (ICIs) as a part of immunotherapy represented a therapeutic breakthrough in the landscape of cancer treatment. The action of these inhibitors consists of blocking certain inhibitory receptors in the immune system. Blocking these inhibitory pathways, ICIs induce an enhanced T cell-mediated response necessary to neutralize tumor cells. Over the last 10 years, programmed death cell protein1 (PD-1), PD ligand 1 (PD-L1), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have been among the inhibitory receptors most targeted by ICIs. Currently, this innovative therapeutic approach faces two major challenges: early identification of cancer patients who are likely to get a significant therapeutic benefit through the use of these inhibitors, and the second challenge is the early prediction of likely immune-related adverse events (irAEs) associated with such therapy. The aim of the present text is to discuss the current research efforts to discover and develop much needed effective biomarkers, which may represent an important step towards more efficient and risk-free immunotherapy. We also highlight the increasing role in clinical analyses of liquid biopsy sampling combined with mass spectrometry-based proteomics and how such combination is contributing to current research efforts to enhance the role of immunotherapy. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Biomarkers for Diagnosis and Prognosis)
Show Figures

Figure 1

17 pages, 8700 KB  
Article
Designing a Novel Multi-Epitope Trivalent Vaccine Against NDV, AIV and FAdV-4 Based on Immunoinformatics Approaches
by Jiashuang Ji, Xiaofeng Dong, Xiangyi Liu, Mengchun Ding, Yating Lin, Yunhang Zhang, Wuchao Zhang, Baishi Lei, Wanzhe Yuan and Kuan Zhao
Microorganisms 2025, 13(12), 2744; https://doi.org/10.3390/microorganisms13122744 - 2 Dec 2025
Viewed by 492
Abstract
The diseases caused by genotype VII Newcastle disease virus (NDV), H9N2 avian influenza virus (AIV), and fowl adenovirus serotype 4 (FAdV-4) continue to threaten the global poultry industry. However, no broad-spectrum vaccines provide simultaneous protection against these three pathogens. This study employed bioinformatics [...] Read more.
The diseases caused by genotype VII Newcastle disease virus (NDV), H9N2 avian influenza virus (AIV), and fowl adenovirus serotype 4 (FAdV-4) continue to threaten the global poultry industry. However, no broad-spectrum vaccines provide simultaneous protection against these three pathogens. This study employed bioinformatics and immunoinformatics approaches to design a multi-epitope vaccine, named NFAF, which consists of B-cell, cytotoxic T lymphocyte (CTL) epitopes, and helper T lymphocyte (HTL) epitopes derived from hemagglutinin-neuraminidase (HN) and fusion (F) proteins of genotype VII NDV, hemagglutinin (HA) protein of H9N2, and Fiber2 protein of FAdV-4. The vaccine candidate was predicted to have non-allergenic properties, non-toxicity, high antigenicity, and favorable solubility. Each of its constituent antigenic epitopes has a high degree of conservation. Molecular docking demonstrated stable binding between NFAF and chicken Toll-like receptor (TLRs) and major histocompatibility complex (MHC) molecules. NFAF was expressed in soluble form in Escherichia coli and purified. Polyclonal antibodies against all three target viruses showed specific binding to NFAF. In vitro experiments revealed that NFAF effectively stimulated chicken peripheral blood mononuclear cells (PBMCs) and induced Th1, Th2, and pro-inflammatory cytokine production, confirming its immunogenicity, and increased the mRNA expression of the key signaling molecules MyD88 and NF-κB. These results suggested that NFAF could therefore be an efficacious multi-epitope vaccine against genotype VII NDV, H9N2, and FAdV-4 infections. Full article
(This article belongs to the Special Issue The Host Response to Animal Virus Infection)
Show Figures

Figure 1

25 pages, 6165 KB  
Article
Immune Checkpoint Signatures in Minimal Change Disease and Membranous Nephropathy: Divergent Pathways of a Shared Imbalance
by Sebastian Mertowski, Paulina Mertowska, Milena Czosnek, Iwona Smarz-Widelska, Wojciech Załuska and Ewelina Grywalska
Int. J. Mol. Sci. 2025, 26(23), 11371; https://doi.org/10.3390/ijms262311371 - 25 Nov 2025
Viewed by 508
Abstract
Primary glomerulopathies share common immune dysregulation but differ in their predominant pathways. We compared immune checkpoint profiles in minimal change disease (MCD) and membranous nephropathy (MN) with those in healthy volunteers (HV). In a cohort of 90 individuals (MCD, n = 30; MN, [...] Read more.
Primary glomerulopathies share common immune dysregulation but differ in their predominant pathways. We compared immune checkpoint profiles in minimal change disease (MCD) and membranous nephropathy (MN) with those in healthy volunteers (HV). In a cohort of 90 individuals (MCD, n = 30; MN, n = 30; HV, n = 30), we performed multiparameter flow cytometry of PBMCs to assess the expression of PD-1/PD-L1, CTLA-4/CD86, and CD200/CD200R on CD4+and CD8+ T cells, CD19+ B cells, and natural killer cells (NK cells). ELISA measured serum soluble checkpoint concentrations, and transcript levels in PBMCs were measured by qPCR. Nonparametric statistics and ROC analysis were used. In MCD, a skewed T cell pattern was observed, characterized by dominant expression of PD-1 and CTLA-4, whereas in MN, a humoral predominance was observed with higher PD-L1 expression and attenuated CD200/CD200R axis. Across diseases, expression profiles and correlations between markers differed between HV and between MCD and MN. Soluble checkpoints (sPD-1, sPD-L1, sCD200, sCD200R) showed potential discriminatory value for GN compared to HV and for differentiating MCD from MN in ROC analyses. These findings indicate that the mechanisms maintaining immune tolerance in primary GN are standard but pathway-specific, consistent with the dominant immunological component of each disease. A significant implication of this study is the need to conduct tissue-level studies to confirm clinical utility and provide insights into personalized immunomodulatory strategies targeting PD-1/PD-L1, CTLA-4/CD86, and CD200/CD200R. Full article
(This article belongs to the Special Issue The Innate Immune System in Renal Diseases)
Show Figures

Figure 1

34 pages, 8250 KB  
Review
From Cytokines to Biomarkers: Mapping the Immunopathology of Inflammatory Bowel Disease
by Sarah Baum, Kamron Hamedi, Caroline Loftus, Gannett Loftus, Emily-Rose Zhou and Sergio Arce
Cells 2025, 14(20), 1589; https://doi.org/10.3390/cells14201589 - 13 Oct 2025
Cited by 1 | Viewed by 2912
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition of the gastrointestinal tract, characterized by dysregulated inflammatory responses throughout the gastrointestinal tract. It includes two major phenotypes, Crohn’s disease (CD) and ulcerative colitis (UC), which present with varying gastrointestinal and systemic symptoms. The [...] Read more.
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition of the gastrointestinal tract, characterized by dysregulated inflammatory responses throughout the gastrointestinal tract. It includes two major phenotypes, Crohn’s disease (CD) and ulcerative colitis (UC), which present with varying gastrointestinal and systemic symptoms. The pathophysiology of IBD is multifactorial including genetic predisposition, mucosal and epithelial dysfunction, environmental injury, and both innate and adaptive immune response abnormalities. Several predisposing genetic factors have been associated with IBD explaining the strong hereditary risk for both CD and UC. For example, Caspase Recruitment Domain 9 (CARD9) variant rs10781499 increases risk for IBD, while other variants are specific to either CD or UC. CD is related to loss-of-function mutations in the nucleotide oligomerization domain containing the protein 2 (NOD2) gene and Autophagy-Related 16-like 1 (ATG16L1) gene. UC risk is increased particularly in Chinese populations by the A-1661G polymorphism of the Cytotoxic T-lymphocyte antigen 4 (CTLA-4) gene. This abnormal CTLA-4 interferes with B- and T-cell responses causing predisposition to autoimmune conditions. Previous studies suggested that IBD results from breakdown of the adaptive immune system, primarily of T-cells. However, new evidence suggests that a primary breakdown of the innate immune system in both CD and UC increases susceptibility to invasion by viruses and bacteria, with a compensatory overactivation of the adaptive immune system as a result. When this viral and microbial invasion continues, further damage is incurred, resulting in a downward cycle of further cytokine activation and epithelial damage. Released biomarkers also affect the permeability of the epithelial membrane, including lactoferrin, nitric oxide (NO), myeloperoxidase (MPO) and its activation of hypochlorous acid, matrix metalloproteinases (MMPs), especially MMP-9, omentin-1, and others. Increased macrophage and dendritic cell dysfunction, increased neutrophil activity, increased numbers of innate lymphoid cells, increased T-cells with decreased regulatory T-cells (Tregs), and changes in B-cell populations and immunoglobulin (Ig) functions are all associated with IBD. Finally, treatment of IBD has typically consisted of medical management (e.g., aminosalicylates and corticosteroids) and lifestyle modification, and surgical intervention in extreme cases. New classes of medications with more favorable side effect profiles include anti-integrin antibodies, vedolizumab, etrolizumab, and carotegrast methyl. Additionally, fecal microbiota transplant (FMT) is a newer area of research for treatment of IBD along with TNF-blockers, JAK inhibitors, and S1PR modulators. However, expense and long preparation time have limited the usefulness of FMT. Full article
Show Figures

Figure 1

20 pages, 4356 KB  
Review
Advanced Immunomodulation in Rheumatoid Arthritis: Immune Checkpoints, microRNAs, and Cell-Based Therapies
by Sandra Pascual-García, Raúl Cobo, José Luis Bolinches, Iván Ortiz, Pedro Viamonte, José Miguel Sempere-Ortells and Pascual Martínez-Peinado
Biomedicines 2025, 13(9), 2186; https://doi.org/10.3390/biomedicines13092186 - 7 Sep 2025
Cited by 2 | Viewed by 2189
Abstract
Background/Objectives: Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by persistent synovial inflammation, progressive joint destruction, and systemic complications. Despite significant progress in targeted therapies, major clinical challenges persist, including heterogeneous treatment responses and therapeutic resistance. This review aims to critically [...] Read more.
Background/Objectives: Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by persistent synovial inflammation, progressive joint destruction, and systemic complications. Despite significant progress in targeted therapies, major clinical challenges persist, including heterogeneous treatment responses and therapeutic resistance. This review aims to critically evaluate emerging immunomodulatory strategies—focusing on immune checkpoints, microRNAs (miRNAs), and cell-based therapies—as potential diagnostic and therapeutic tools. Methods: This non-systematic literature review involved a comprehensive analysis of recent studies to investigate emerging immunomodulatory strategies in RA. Special attention was given to immune checkpoint pathways—cytotoxic T-lymphocyte antigen 4 (CTLA-4); programmed death-1 (PD-1) and its ligand, PD-L1; and inducible T-cell costimulator (ICOS)—as well as cell-based therapies. Additionally, miRNA-based interventions were examined for their diagnostic and therapeutic potential. Results: Immune checkpoint modulation has demonstrated preclinical efficacy in attenuating inflammatory responses and restoring immune tolerance. Concurrently, miRNAs have emerged as both biomarkers and therapeutic agents, with exosome-based delivery systems enhancing their function. Cell-based therapies have shown robust immunoregulatory effects with acceptable safety profiles. Notably, integrative strategies that combine checkpoint inhibitors, cell-based interventions, and miRNA delivery exhibit synergistic effects and offer a promising avenue for personalised treatment, when guided by molecular and transcriptomic profiling. The majority of these approaches remain at the preclinical or early translational stage. Conclusions: Targeted immunomodulation is poised to transform RA management. The integration of cell therapies, checkpoint inhibition, and miRNA manipulation with omics technologies holds promise for enhancing therapeutic precision and safety. Advancing towards personalised immunotherapy will necessitate a multidisciplinary and patient-centred effort. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnostics, and Therapeutics for Rheumatic Diseases)
Show Figures

Graphical abstract

31 pages, 1533 KB  
Review
Immunodynamic Disruption in Sepsis: Mechanisms and Strategies for Personalized Immunomodulation
by Jhan S. Saavedra-Torres, María Virginia Pinzón-Fernández, Humberto Alejandro Nati-Castillo, Valentina Cadena Correa, Luis Carlos Lopez Molina, Juan Estaban Gaitán, Daniel Tenorio-Castro, Diego A. Lucero Guanga, Marlon Arias-Intriago, Andrea Tello-De-la-Torre, Alice Gaibor-Pazmiño and Juan S. Izquierdo-Condoy
Biomedicines 2025, 13(9), 2139; https://doi.org/10.3390/biomedicines13092139 - 2 Sep 2025
Cited by 1 | Viewed by 2884
Abstract
Sepsis is a life-threatening syndrome caused by a dysregulated host response to infection. It follows a dynamic course in which early hyperinflammation coexists and overlaps with progressive immune suppression, a process best described as immunodynamic disruption. Key mechanisms include extensive lymphocyte death, expansion [...] Read more.
Sepsis is a life-threatening syndrome caused by a dysregulated host response to infection. It follows a dynamic course in which early hyperinflammation coexists and overlaps with progressive immune suppression, a process best described as immunodynamic disruption. Key mechanisms include extensive lymphocyte death, expansion of regulatory T cells, impaired antigen presentation, and persistent activation of inhibitory checkpoints such as programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte–associated protein 4 (CTLA-4). These changes reduce immune competence and increase vulnerability to secondary infections. Clinically, reduced expression of Human Leukocyte Antigen–DR (HLA-DR) on monocytes and persistent lymphopenia have emerged as robust biomarkers for patient stratification and timing of immunomodulatory therapies. Beyond the acute phase, many survivors do not achieve full immune recovery but instead develop a Persistent Immune Remnant, defined as long-lasting immune, metabolic, and endothelial dysfunction despite apparent clinical resolution. Recognizing PIR emphasizes the need for long-term monitoring and biomarker-guided interventions to restore immune balance. To integrate these observations, we propose the SIMMP–Sepsis model (Sepsis-Associated Persistent Multiorgan Immunometabolic Syndrome), which links molecular dysfunction to clinical trajectories and provides a framework for developing precision immunotherapies. This perspective reframes sepsis not only as an acute crisis but also as a chronic immunometabolic syndrome, where survival marks the beginning of active immune restoration. Full article
Show Figures

Figure 1

17 pages, 3372 KB  
Article
Analysis of Immune Checkpoints on Peripheral Blood Mononuclear Cells Can Predict Clinical Outcome and Reveal Potential of HVEM-BTLA Axis in Epithelial Ovarian Cancers
by Yen-Ling Lai, Han-Wei Lin, Yu-Wen Huang, Jung Chen, Ming-Chien Tai, Chia-Ying Wu, Tyan-Shin Yang, Valentina Oblin, Kristin Shea and Yu-Li Chen
Pharmaceuticals 2025, 18(9), 1295; https://doi.org/10.3390/ph18091295 - 29 Aug 2025
Cited by 2 | Viewed by 1117
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) do not provide promising benefits to patients with advanced epithelial ovarian cancer (EOC). This study analyzed preoperative peripheral blood mononuclear cells (PBMCs) from these patients to evaluate the prognostic and therapeutic checkpoints. Methods: Preoperative PBMCs of 69 advanced [...] Read more.
Background/Objectives: Immune checkpoint inhibitors (ICIs) do not provide promising benefits to patients with advanced epithelial ovarian cancer (EOC). This study analyzed preoperative peripheral blood mononuclear cells (PBMCs) from these patients to evaluate the prognostic and therapeutic checkpoints. Methods: Preoperative PBMCs of 69 advanced EOC cases were collected to analyze the correlation between IC-expressing immune cells and survivals of patients. Co-expression of various ICs on the T lymphocytes from these patients was examined. Activation potential of programmed cell death 1 (PD-1)+herpes virus entry mediator (HVEM)+ T cells in PBMCs from the healthy donors and tumoricidal abilities of PMBCs treated with various ICIs were evaluated in vitro. Impact of respective ICIs on activation of T cells in PMBCs was investigated. Results: Percentages of PD-1+ CD4+ and CD8+ T cells in the PBMCs of patients could positively correlate with disease-free or overall survival. HVEM was highly co-expressed on these T lymphocytes. Prediction potential for overall survival of patients by the subpopulation of PD-1+ CD4+ or CD8+ T cells was higher than that by other parameters. The PD-1+HVEM+ CD4+ and CD8+ T cells showed characteristics of activated phenotype under activation signals. PBMCs receiving anti-B and T lymphocyte attenuator (BTLA) plus anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) or anti-PD-1 Ab had potent tumor-killing ability. Anti-BTLA Ab can drive T cells in the PBMCs toward an effector status. Conclusions: Percentages of PD-1+ T cells in the PBMCs could predict survival of EOC patients. Targeting HVEM-BTLA axis may be considered for ICI treatment of EOCs. Full article
(This article belongs to the Special Issue Tumor Immunopharmacology)
Show Figures

Figure 1

29 pages, 4115 KB  
Article
In Silico Design of a Multiepitope Vaccine Against Intestinal Pathogenic Escherichia coli Based on the 2011 German O104:H4 Outbreak Strain Using Reverse Vaccinology and an Immunoinformatic Approach
by Eman G. Youssef, Khaled Elnesr and Amro Hanora
Diseases 2025, 13(8), 259; https://doi.org/10.3390/diseases13080259 - 13 Aug 2025
Viewed by 1203
Abstract
Background: While most Escherichia coli strains are harmless members of the gastrointestinal microbiota, certain pathogenic variants can cause severe intestinal and extraintestinal diseases. A notable outbreak of E. coli O104:H4, involving both enteroaggregative (EAEC) and enterohemorrhagic (EHEC) strains, occurred [...] Read more.
Background: While most Escherichia coli strains are harmless members of the gastrointestinal microbiota, certain pathogenic variants can cause severe intestinal and extraintestinal diseases. A notable outbreak of E. coli O104:H4, involving both enteroaggregative (EAEC) and enterohemorrhagic (EHEC) strains, occurred in Europe, resulting in symptoms ranging from bloody diarrhea to life-threatening colitis and hemolytic uremic syndrome (HUS). Since treatment options remain limited and have changed little over the past 40 years, there is an urgent need for an effective vaccine. Such a vaccine would offer major public health and economic benefits by preventing severe infections and reducing outbreak-related costs. A multiepitope vaccine approach, enabled by advances in immunoinformatics, offers a promising strategy for targeting HUS-causing E. coli (O104:H4 and O157:H7 serotypes) with minimal disruption to normal microbiota. This study aimed to design an immunogenic multiepitope vaccine (MEV) construct using bioinformatics and immunoinformatic tools. Methods and Results: Comparative proteomic analysis identified 672 proteins unique to E. coli O104:H4, excluding proteins shared with the nonpathogenic E. coli K-12-MG1655 strain and those shorter than 100 amino acids. Subcellular localization (P-SORTb) identified 17 extracellular or outer membrane proteins. Four proteins were selected as vaccine candidates based on transmembrane domains (TMHMM), antigenicity (VaxiJen), and conservation among EHEC strains. Epitope prediction revealed ten B-cell, four cytotoxic T-cell, and three helper T-cell epitopes. Four MEVs with different adjuvants were designed and assessed for solubility, stability, and antigenicity. Structural refinement (GALAXY) and docking studies confirmed strong interaction with Toll-Like Receptor 4 (TLR4). In silico immune simulations (C-ImmSim) indicated robust humoral and cellular immune responses. In Conclusions, the proposed MEV construct demonstrated promising immunogenicity and warrants further validation in experimental models. Full article
Show Figures

Figure 1

12 pages, 446 KB  
Article
Clinical Impact of CTLA-4 Single-Nucleotide Polymorphism in DLBCL Patients Treated with CAR-T Cell Therapy
by Katja Seipel, Inna Shaforostova, Henning Nilius, Ulrike Bacher and Thomas Pabst
Curr. Oncol. 2025, 32(8), 425; https://doi.org/10.3390/curroncol32080425 - 29 Jul 2025
Cited by 4 | Viewed by 1835
Abstract
FMC63-CAR T cell therapy targeting CD19 protein on malignant B-cells is effective in patients with relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL), with complete response rates of 43–54%. Common germline variants of the immune-checkpoint regulator CTLA-4 may elicit different responses to [...] Read more.
FMC63-CAR T cell therapy targeting CD19 protein on malignant B-cells is effective in patients with relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL), with complete response rates of 43–54%. Common germline variants of the immune-checkpoint regulator CTLA-4 may elicit different responses to CAR-T cell therapy. The CTLA4 gene single-nucleotide polymorphism rs231775 coding threonine or alanine at amino acid position 17 of the CTLA-4 protein was prevalent in 55% of the studied DLBCL patients. In a retrospective comparative analysis of clinical outcome, there were significant differences in CTLA4 A17hom vs. T17Ahet and T17hom carriers with four-year progression-free survival at 77%, 59%, and 30% (p = 0.019), four-year overall survival was 79%, 41%, and 33% (p = 0.049), the relapse rates were 20%, 37%, and 56% (p = 0.025), and the death rates 20%, 54%, and 52% (p = 0.049). Conclusions: CTLA4 rs231775 polymorphism may impact the treatment outcome in FMC63-anti-CD19 CAR-T cell therapy, with an association of the CTLA4 minor allele A17 to favorable treatment outcome. Full article
(This article belongs to the Section Cell Therapy)
Show Figures

Graphical abstract

28 pages, 9321 KB  
Article
In Situ Vaccination with a Vpr-Derived Peptide Elicits Systemic Antitumor Immunity by Improving Tumor Immunogenicity
by Danjie Pan, Ling Du, Jiayang Liu, Kudelaidi Kuerban, Xuan Huang, Yue Wang, Qiuyu Guo, Huaning Chen, Songna Wang, Li Wang, Pinghong Zhou, Zhefeng Meng and Li Ye
Vaccines 2025, 13(7), 710; https://doi.org/10.3390/vaccines13070710 - 30 Jun 2025
Viewed by 1349
Abstract
Background: Cancer vaccines represent a groundbreaking advancement in cancer immunotherapy, utilizing tumor antigens to induce tumor-specific immune responses. However, challenges like tumor-induced immune resistance and technical barriers limit the widespread application of predefined antigen vaccines. Here, we investigated the potential of viral protein [...] Read more.
Background: Cancer vaccines represent a groundbreaking advancement in cancer immunotherapy, utilizing tumor antigens to induce tumor-specific immune responses. However, challenges like tumor-induced immune resistance and technical barriers limit the widespread application of predefined antigen vaccines. Here, we investigated the potential of viral protein R (Vpr) peptides as effective candidates for constructing anonymous antigen vaccines in situ by directly injecting at the tumor site and releasing whole-tumor antigens, inducing robust anti-tumor immune responses to overcome the limitations of predefined antigen vaccines. Methods: The cytotoxic effects of Vpr peptides were evaluated using the CCK8 reagent kit. Membrane penetration ability of Vpr peptides was observed using a confocal laser scanning microscope and quantitatively analyzed using flow cytometry. EGFR levels in the cell culture supernatants of cells treated with Vpr peptides were evaluated using an ELISA. Surface exposure of CRT on the tumor cell surface was observed using a confocal laser scanning microscope and quantitatively analyzed using flow cytometry. The secretion levels of ATP from tumor cells were evaluated using an ATP assay kit. HMGB1 release was evaluated using an ELISA. Mouse (Male C57BL/6 mice aged 4 weeks) MC38 and LLC bilateral subcutaneous tumor models were established to evaluate the therapeutic effects of Vpr peptides through in situ vaccination. Proteomic analysis was performed to explore the mechanism of anti-tumor activity of Vpr peptides. Results: Four Vpr peptides were designed and synthesized, with P1 and P4 exhibiting cytotoxic effects on tumor cells, inducing apoptosis and immunogenic cell death. In mouse tumor models, in situ vaccination with Vpr peptide significantly inhibited tumor growth and activated various immune cells. High-dose P1 monotherapy demonstrated potent anti-tumor effects, activating DCs, T cells, and macrophages. Combining ISV of P1 with a CD47 inhibitor SIRPαFc fusion protein showed potent distant tumor suppression effects. Proteomic analysis suggested that Vpr peptides exerted anti-tumor effects by disrupting tumor cell morphology, movement, and adhesion, and promoting immune cell infiltration. Conclusions: The designed Vpr peptides show promise as candidates for in situ vaccination, with significant anti-tumor effects, immune activation, and favorable safety profiles observed in mouse models. In situ vaccination with Vpr-derived peptides represents a potential approach for cancer immunotherapy. Full article
(This article belongs to the Special Issue New Approaches to Vaccine Development and Delivery)
Show Figures

Figure 1

12 pages, 1739 KB  
Article
Local T-Cell Dysregulation and Immune Checkpoint Expression in Human Papillomavirus-Mediated Recurrent Respiratory Papillomatosis
by Hans N. C. Eckel, Su Ir Lyu, Frederik Faste, Shachi J. Sharma, Anne Nobis, Nora Wuerdemann, Maria Ziogas, Marcel Mayer, Malte C. Suchan, Kerstin Wennhold, Maria A. Garcia-Marquez, Martin Thelen, Elena Hagen, Julia Eßer, Charlotte Klasen, Oliver Siefer, Martin Otte, Hans A. Schloesser, Jens P. Klussmann, Alexander Quaas and Kevin K. Hansenadd Show full author list remove Hide full author list
Cells 2025, 14(13), 985; https://doi.org/10.3390/cells14130985 - 27 Jun 2025
Viewed by 1054
Abstract
Human papillomavirus-mediated recurrent respiratory papillomatosis (RRP) is a premalignant neoplasia of the upper airway characterized by significant dysphonia and respiratory obstruction. Immune checkpoint blockade has emerged as a potential alternative to repeated surgical interventions in RRP. Here, we investigated the intralesional T-cell composition [...] Read more.
Human papillomavirus-mediated recurrent respiratory papillomatosis (RRP) is a premalignant neoplasia of the upper airway characterized by significant dysphonia and respiratory obstruction. Immune checkpoint blockade has emerged as a potential alternative to repeated surgical interventions in RRP. Here, we investigated the intralesional T-cell composition and expression of the immune checkpoints programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) in RRP. We analyzed tissue samples from 30 patients treated at a tertiary care center between 2009 and 2021, including paired samples from individual patients collected at different time points. Immunohistochemical staining was performed for CD4, CD8, CTLA-4, FoxP3, and PD-L1 and correlated with disease severity and previous adjuvant therapies. Overall disease burden and intervention-free survival were not associated with the abundance of CD4+, CD8+, or FoxP3+ T cells, nor with immune checkpoint expression. However, patients with aggressive disease exhibited a higher intralesional FoxP3/CD4 T-cell ratio. Prior intralesional cidofovir treatment was associated with reduced CD4+ T-cell infiltration. These findings suggest that a locally immunosuppressive microenvironment, reflected by an elevated FoxP3/CD4 ratio, contributes to disease severity in RRP. Consistent CTLA-4 expression across all evaluated samples supports further investigation of anti-CTLA-4 therapy, either alone or in combination with other checkpoint inhibitors. Full article
(This article belongs to the Section Cellular Immunology)
Show Figures

Graphical abstract

22 pages, 488 KB  
Systematic Review
Immune Checkpoint Inhibitors for Metastatic Colorectal Cancer: A Systematic Review
by Alice Gilson, Vincent Tan, Thibaud Koessler, Jeremy Meyer, Guillaume Meurette, Émilie Liot, Frédéric Ris and Vaihere Delaune
Cancers 2025, 17(13), 2125; https://doi.org/10.3390/cancers17132125 - 24 Jun 2025
Cited by 2 | Viewed by 3955
Abstract
Background: Colorectal cancer is a significant health concern. Immunotherapy has become a promising approach in colorectal cancer, offering a wider array of therapeutic strategies. This study aims to summarize the current evidence regarding the use of checkpoint inhibitors in metastatic colorectal cancer. Methods: [...] Read more.
Background: Colorectal cancer is a significant health concern. Immunotherapy has become a promising approach in colorectal cancer, offering a wider array of therapeutic strategies. This study aims to summarize the current evidence regarding the use of checkpoint inhibitors in metastatic colorectal cancer. Methods: A systematic review of relevant clinical trials and randomized controlled trials (RCTs) assessing checkpoint inhibitors, published between January 2019 and January 2025, was conducted on Medline, Web of Science, and Cochrane. Progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) were the primary outcomes. Studies focusing on other types of immunotherapy, non-clinical trials, pre-clinical trials, and study protocols were excluded. Results: 48 studies were included. Checkpoint inhibitors demonstrated significant efficacy in microsatellite instability (MSI) metastatic colorectal cancer (mCRC). In microsatellite stability (MSS) mCRC, immunotherapy was less effective, and combination strategies with chemotherapy or targeted therapies yielded mixed results. Grade ≥ 3 treatment-related adverse events (TRAEs) were common in combination regimens. Conclusion: Immunotherapy has revolutionized MSI mCRC treatment while treating MSS CRC with these molecules remains unconvincing. Combination strategies and novel agents may offer potential but require further research to prove efficacy. Full article
Show Figures

Figure 1

43 pages, 1769 KB  
Review
The Role of LAIR1 as a Regulatory Receptor of Antitumor Immune Cell Responses and Tumor Cell Growth and Expansion
by Alessandro Poggi, Serena Matis, Chiara Rosa Maria Uras, Lizzia Raffaghello, Roberto Benelli and Maria Raffaella Zocchi
Biomolecules 2025, 15(6), 866; https://doi.org/10.3390/biom15060866 - 13 Jun 2025
Cited by 1 | Viewed by 2743
Abstract
It is becoming evident that the therapeutic effect of reawakening the immune response is to limit tumor cell growth and expansion. The use of immune checkpoint inhibitors, like blocking antibodies against programmed cell death receptor (PD) 1 and/or cytotoxic T lymphocyte antigen (CTLA) [...] Read more.
It is becoming evident that the therapeutic effect of reawakening the immune response is to limit tumor cell growth and expansion. The use of immune checkpoint inhibitors, like blocking antibodies against programmed cell death receptor (PD) 1 and/or cytotoxic T lymphocyte antigen (CTLA) 4 alone or in combination with other drugs, has led to unexpected positive results in some tumors but not all. Several other molecules inhibiting lymphocyte antitumor effector subsets have been discovered in the last 30 years. Herein, we focus on the leukocyte-associated immunoglobulin (Ig)-like receptor 1 (LAIR1/CD305). LAIR1 represents a typical immunoregulatory molecule expressed on almost all leukocytes, unlike other regulatory receptors expressed on discrete leukocyte subsets. It bears two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in the intracytoplasmic protein domain involved in the downregulation of signals mediated by activating receptors. LAIR1 binds to several ligands, such as collagen I and III, complement component 1Q, surfactant protein D, adiponectin, and repetitive interspersed families of polypeptides expressed by erythrocytes infected with Plasmodium malariae. This would suggest LAIR1 involvement in several cell-to-cell interactions and possibly in metabolic regulation. The presence of both cellular and soluble forms of LAIR would indicate a fine regulation of the immunoregulatory activity, as happens for the soluble/exosome-associated forms of PD1 and CTLA4 molecules. As a consequence, LAIR1 appears to play a role in some autoimmune diseases and the immune response against tumor cells. The finding of LAIR1 expression on hematological malignancies, but also on some solid tumors, could open a rationale for the targeting of this molecule to treat neoplasia, either alone or in combination with other therapeutic options. Full article
Show Figures

Figure 1

Back to TopTop