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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (5,687)

Search Parameters:
Keywords = immune checkpoints

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 6804 KB  
Article
Chemotherapy Liberates a Broadening Repertoire of Tumor Antigens for TLR7/8/9-Mediated Potent Antitumor Immunity
by Cheng Zu, Yiwei Zhong, Shuting Wu and Bin Wang
Cancers 2025, 17(19), 3277; https://doi.org/10.3390/cancers17193277 - 9 Oct 2025
Abstract
Background: Most immunologically “cold” tumors do not respond durably to checkpoint blockade because tumor antigen (TA) release and presentation are insufficient to prime effective T-cell immunity. While prior work demonstrated synergy between cisplatin and a TLR7/8/9 agonist (CR108) in 4T1 tumors, the underlying [...] Read more.
Background: Most immunologically “cold” tumors do not respond durably to checkpoint blockade because tumor antigen (TA) release and presentation are insufficient to prime effective T-cell immunity. While prior work demonstrated synergy between cisplatin and a TLR7/8/9 agonist (CR108) in 4T1 tumors, the underlying mechanism—particularly whether chemotherapy functions as a broad antigen-releasing agent enabling TLR-driven immune amplification—remained undefined. Methods: Using murine models of breast (4T1), melanoma (B16-F10), and colorectal cancer (CT26), we tested multiple chemotherapeutic classes combined with CR108. We quantified intratumoral and systemic soluble TAs, antigen presentation and cross-priming by antigen-presenting cells, tumor-infiltrating lymphocytes, and cytokine production by flow cytometry/ICS. T-cell receptor β (TCRβ) repertoire dynamics in tumor-draining lymph nodes were profiled to assess amplitude and breadth. Tumor microenvironment remodeling was analyzed, and public datasets (e.g., TCGA basal-like breast cancer) were interrogated for expression of genes linked to TA generation/processing and peptide loading. Results: Using cisplatin + CR108 in 4T1 as a benchmark, we demonstrate that diverse chemotherapies—especially platinum agents—broadly increase the repertoire of soluble tumor antigens available for immune recognition. Across regimens, chemotherapy combined with CR108 increased T-cell recognition of candidate TAs and enhanced IFN-γ+ CD8+ responses, with platinum agents producing the largest expansions in soluble TAs. TCRβ sequencing revealed increased clonal amplitude without loss of repertoire breadth, indicating focused yet diverse antitumor T-cell expansion. Notably, therapeutic efficacy was not predicted by canonical damage-associated molecular pattern (DAMP) signatures but instead correlated with antigen availability and processing capacity. In human basal-like breast cancer, higher expression of genes involved in TA generation and antigen processing/presentation correlated with improved survival. Conclusions: Our findings establish an antigen-centric mechanism underlying chemo–TLR agonist synergy: chemotherapy liberates a broadened repertoire of tumor antigens, which CR108 then leverages via innate immune activation to drive potent, T-cell-mediated antitumor immunity. This framework for rational selection of chemotherapy partners for TLR7/8/9 agonism and support clinical evaluation to convert “cold” tumors into immunologically responsive disease. Full article
14 pages, 1356 KB  
Article
Regulation of PD-L1 Protein Expression by the E3 Ubiquitin Ligase GP78
by Madhumita Chatterjee, Julio M. Pimentel, Jun-Ying Zhou, Thamarahansi Mugunamalwaththa, Zhe Yang, Avraham Raz and Gen Sheng Wu
Curr. Issues Mol. Biol. 2025, 47(10), 829; https://doi.org/10.3390/cimb47100829 (registering DOI) - 9 Oct 2025
Abstract
Immune checkpoint inhibitors (ICIs), including PD-L1 inhibitors, have been approved by the FDA for the treatment of cancers; however, only a small number of cancer patients benefit from these ICIs. Furthermore, the development of drug resistance to this type of treatment is often [...] Read more.
Immune checkpoint inhibitors (ICIs), including PD-L1 inhibitors, have been approved by the FDA for the treatment of cancers; however, only a small number of cancer patients benefit from these ICIs. Furthermore, the development of drug resistance to this type of treatment is often inevitable. The mechanisms of resistance to PD-L1 inhibitors can be attributed, in part, to an incomplete understanding of the regulation of PD-L1 protein expression. In this study, we identified the role of the E3 ligase GP78, also known as the Autocrine Motility Factor Receptor (AMFR), in the regulation of PD-L1 protein levels. We show that GP78 physically interacts with PD-L1, which is confirmed by IP and Western blotting and is supported by molecular modelling using AlphaFold2. Our modeling studies predict that the interface amino acids of the Ig1 domain of PD-L1 interact with the RING domain and a β-hairpin preceding the CUE domain of GP78. The crystal structure of the PD-1/PD-L1 complex reveals that the interaction with PD-1 is mediated by the Ig1 domain of PD-L1. Furthermore, proteasomal degradation of PD-L1 has been observed via GP78-mediated K48-linked ubiquitination, indicating a key regulatory role for GP78 in the downregulation of PD-L1. Because GP78 expression is inversely correlated with PD-L1 levels in cancer, these findings may have clinical implications for predicting tumor immune evasion and patient response to PD-1/PD-L1 blockade therapies. Taken together, these findings identify a previously unknown mechanism by which GP78 targets PD-L1 for ubiquitination and subsequent degradation in cancer cells, and suggest that blocking the interaction between PD-L1 and PD-1 by an E3 ligase is a novel strategy to improve immunotherapies for cancer patients. Full article
(This article belongs to the Section Molecular Medicine)
23 pages, 974 KB  
Review
Immune Checkpoint Inhibitors in Merkel Cell Carcinoma of the Skin: A 2025 Comprehensive Review
by Patricia Tai, Omar Alqaisi, Suhair Al-Ghabeesh, Lorent Sijarina, Edward Yu, Aoife Jones Thachuthara, Avi Assouline, Osama Souied, Kimberly Hagel and Kurian Joseph
Cancers 2025, 17(19), 3272; https://doi.org/10.3390/cancers17193272 - 9 Oct 2025
Abstract
Objective: Merkel cell carcinoma (MCC) is a rare and aggressive form of skin cancer. Although immunotherapy has transformed MCC management, published data remain limited. This comprehensive review evaluates current evidence on immune checkpoint inhibitors (ICIs) in MCC, in relation to other treatment modalities [...] Read more.
Objective: Merkel cell carcinoma (MCC) is a rare and aggressive form of skin cancer. Although immunotherapy has transformed MCC management, published data remain limited. This comprehensive review evaluates current evidence on immune checkpoint inhibitors (ICIs) in MCC, in relation to other treatment modalities such as surgery and radiotherapy. Methods: Peer-reviewed articles published between January 2000 and August 2025 were searched manually in four databases: Scopus, ScienceDirect, PubMed and MEDLINE, using the keywords “Merkel cell carcinoma” AND “immunotherapy” AND “immune checkpoint inhibitors”. The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) methodology was employed. Results: ICIs can be given in different settings: (A) Neoadjuvant: The CheckMate 358 trial reported a 54.5% response rate among 33 radiologically evaluable patients treated with nivolumab, each showing over 30% tumor reduction. (B) Adjuvant: (1) The ADMEC-O phase II trial demonstrated improved disease-free survival with adjuvant nivolumab. (2) The ADAM phase III trial evaluates adjuvant avelumab in node-positive patients post-surgery/radiation, with common side effects including nausea, fatigue, and itching. (3) STAMP, a phase III trial, investigates pembrolizumab in stage I–III MCC. Both ADAM and STAMP have completed accrual and results are pending. (C) Primary therapy: KEYNOTE-017 and JAVELIN trials reported a 60% overall response rate and ~40% 3-year progression-free survival with first-line pembrolizumab or avelumab. Both agents also show promise as salvage therapies. Conclusions: ICIs demonstrate encouraging outcomes in MCC across various treatment stages. Continued research is essential to optimize treatment timing and integrate multimodal therapies. Full article
(This article belongs to the Special Issue Combination Immunotherapy for Cancer Treatment)
26 pages, 1116 KB  
Review
Optimizing Anti-PD1 Immunotherapy: An Overview of Pharmacokinetics, Biomarkers, and Therapeutic Drug Monitoring
by Joaquim Faria Monteiro, Alexandrina Fernandes, Diogo Gavina Tato, Elias Moreira, Ricardo Ribeiro, Henrique Reguengo, Jorge Gonçalves and Paula Fresco
Cancers 2025, 17(19), 3262; https://doi.org/10.3390/cancers17193262 - 8 Oct 2025
Abstract
Anti-PD-1 therapies have transformed cancer treatment by restoring antitumor T cell activity. Despite their broad clinical use, variability in treatment response and immune-related adverse events underscore the need for therapeutic optimization. This article provides an integrative overview of the pharmacokinetics (PKs) of anti-PD-1 [...] Read more.
Anti-PD-1 therapies have transformed cancer treatment by restoring antitumor T cell activity. Despite their broad clinical use, variability in treatment response and immune-related adverse events underscore the need for therapeutic optimization. This article provides an integrative overview of the pharmacokinetics (PKs) of anti-PD-1 antibodies—such as nivolumab, pembrolizumab, and cemiplimab—and examines pharmacokinetic–pharmacodynamic (PK-PD) relationships, highlighting the impact of clearance variability on drug exposure, efficacy, and safety. Baseline clearance and its reduction during therapy, together with interindividual variability, emerge as important dynamic biomarkers with potential applicability across different cancer types for guiding individualized dosing strategies. The review also discusses established biomarkers for anti-PD-1 therapies, including tumor PD-L1 expression and immune cell signatures, and their relevance for patient stratification. The evidence supports a shift from traditional weight-based dosing toward adaptive dosing and therapeutic drug monitoring (TDM), especially in long-term responders and cost-containment contexts. Notably, the inclusion of clearance-based biomarkers—such as baseline clearance and its reduction—into therapeutic models represents a key step toward individualized, dynamic immunotherapy. In conclusion, optimizing anti-PD-1 therapy through PK-PD insights and biomarker integration holds promise for improving outcomes and reducing toxicity. Future research should focus on validating PK-based approaches and developing robust algorithms (machine learning models incorporating clearance, tumor burden, and other validated biomarkers) for tailored cancer treatment. Full article
Show Figures

Figure 1

13 pages, 1092 KB  
Article
Real-World Effectiveness of Racotumomab as Maintenance Therapy in Advanced Non-Small Cell Lung Cancer Patients
by Sailyn Alfonso Alemán, Haslen Cáceres Lavernia, Kirenia Camacho Sosa, Soraida C. Acosta Brooks, Orestes Santos Morales, Carmen E. Viada González, Meylán Cepeda Portales, Mayelín Troche Concepción, Loipa Medel Pérez, Leticia Cabrera Benítez, Milagros C. Domecq Salmón, Daymys Estévez Iglesias, Mayra Ramos Suzarte and Tania Crombet Ramos
Vaccines 2025, 13(10), 1035; https://doi.org/10.3390/vaccines13101035 - 7 Oct 2025
Abstract
Background: Advanced non-small cell lung cancer (NSCLC) has limited curative options and poor survival. Racotumomab, an anti-idiotype monoclonal antibody vaccine targeting tumor gangliosides, has shown efficacy in clinical trials. This study evaluated its real-world effectiveness as maintenance therapy following first-line chemotherapy. Materials and [...] Read more.
Background: Advanced non-small cell lung cancer (NSCLC) has limited curative options and poor survival. Racotumomab, an anti-idiotype monoclonal antibody vaccine targeting tumor gangliosides, has shown efficacy in clinical trials. This study evaluated its real-world effectiveness as maintenance therapy following first-line chemotherapy. Materials and Methods: A multi-center observational study was conducted on 162 patients with advanced NSCLC who received racotumomab from 2012 to 2024. Effectiveness was evaluated in the intention-to-treat (ITT) cohort. Overall survival (OS) was estimated, with subgroup analyses conducted according to clinical and demographic factors. Results: The median OS was 14.9 months (95% CI: 11.7–18.1), and the 5-year survival rate reached 20%. Patients diagnosed with stage III disease, those with better Eastern Cooperative Oncology Group (ECOG) performance status, and individuals younger than 65 years experienced significantly longer survival. Racotumomab demonstrated a favorable hazard ratio compared to historical controls (HR 0.44 vs. supportive care; HR 0.55 vs. docetaxel). Conclusions: In the era of immune checkpoint inhibitors, these real-world results indicate a promising role for racotumomab in the maintenance setting for advanced NSCLC. These findings provide a strong rationale for further investigation of racotumomab in the context of modern immunotherapy, particularly in combination trials with other immunomodulatory antibodies, along with the validation of clinical and biologic predictive biomarkers. Full article
(This article belongs to the Section Vaccine Advancement, Efficacy and Safety)
Show Figures

Figure 1

18 pages, 1567 KB  
Article
Development of Clinical-Grade Durvalumab-680LT and Nivolumab-800CW for Multispectral Fluorescent Imaging of the PD-1/PD-L1 Axis of the Immune Checkpoint Pathway
by Henrik K. Huizinga, Wouter T. R. Hooghiemstra, Matthijs D. Linssen, Derk P. Allersma, Bahez Gareb, Bart G. J. Dekkers, Wouter B. Nagengast and Marjolijn N. Lub-de Hooge
Pharmaceuticals 2025, 18(10), 1501; https://doi.org/10.3390/ph18101501 - 7 Oct 2025
Viewed by 47
Abstract
Background: Immune checkpoint inhibitors (ICIs) are effective against various advanced and metastatic cancers, but patient responses vary and can change over time, complicating treatment prediction. Therefore, better tools for patient stratification, response prediction, and response assessment are needed. This study presents the development [...] Read more.
Background: Immune checkpoint inhibitors (ICIs) are effective against various advanced and metastatic cancers, but patient responses vary and can change over time, complicating treatment prediction. Therefore, better tools for patient stratification, response prediction, and response assessment are needed. This study presents the development and clinical translation of a fluorescently labelled ICI tracer pair used to perform multispectral fluorescent molecular imaging and simultaneously gain spatial and temporal insight in both programmed death ligand 1 (PD-L1) and programmed death receptor 1 (PD-1) expression. Methods: We conjugated the anti-PD-L1 antibody durvalumab to IRDye 680LT and the anti-PD-1 antibody nivolumab to IRDye 800CW. Tracers were developed and optimized for conjugation efficiency and purity to allow use in clinical trials. Stability was tested up to 12 months. An extended single-dose toxicity study in mice was performed for durvalumab-680LT and the unconjugated IRDye 680LT to demonstrate safety for first-in-human administration. Results: Durvalumab-680LT and nivolumab-800CW were successfully conjugated and purified. Conjugation optimization resulted in a robust production with labelling efficiencies of ≥88%. Long-term stability study of both tracers showed all parameters within end of shelf-life specifications for at least 12 months at 2–8 °C. No toxic effects were observed in doses up to 1000x the intended human dose for both IRDye 680LT and durvalumab-680LT, which are therefore considered safe for first-in-human use. Conclusions: We succeeded in the development and clinical translation of two novel fluorescent ICI tracers, durvalumab-680LT and nivolumab-800CW. Moreover, we demonstrated for the first time the safety of IRDye 680LT and durvalumab-680LT, enabling first-in-human use. Together, this makes durvalumab-680LT and nivolumab-800CW suitable for phase I/II clinical trials. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

0 pages, 1512 KB  
Review
Triple-Negative Breast Cancer Unveiled: Bridging Science, Treatment Strategy, and Economic Aspects
by Valeriia Lebedeva, Mira Ebbinghaus, José Villacorta Hidalgo, Olaf Hardt and Rita Pfeifer
Int. J. Mol. Sci. 2025, 26(19), 9714; https://doi.org/10.3390/ijms26199714 - 6 Oct 2025
Viewed by 335
Abstract
Triple-negative breast cancer (TNBC) has historically been challenging to treat due to limited therapeutic options. Since 2018, the treatment landscape has evolved substantially with the approval of precision therapies, including immune checkpoint inhibitors, poly (ADP-ribose) polymerase inhibitors, and antibody–drug conjugates. Despite these advancements, [...] Read more.
Triple-negative breast cancer (TNBC) has historically been challenging to treat due to limited therapeutic options. Since 2018, the treatment landscape has evolved substantially with the approval of precision therapies, including immune checkpoint inhibitors, poly (ADP-ribose) polymerase inhibitors, and antibody–drug conjugates. Despite these advancements, the therapeutic benefit remains limited due to various clinical challenges, largely driven by TNBC heterogeneity and an incomplete understanding of drug–tumor crosstalk mechanisms—both contributing to a restricted pool of eligible patients and variable treatment responses. Concurrently, emerging drugs tested in recent pivotal TNBC trials have demonstrated variable outcomes. Additionally, the associated economic burden has become a pressing global concern, as several approved drugs provide insufficient clinical benefit in relation to high expenditures, often driven by drug pricing. The situation is particularly critical in low- and middle-income countries, where TNBC is highly prevalent, yet access to even chemotherapeutic treatment remains limited. These factors collectively hinder real-world patient outcomes. This review provides a comprehensive analysis of TNBC management, integrating clinical advancements with economic perspectives and raising awareness of underdiscussed topics. The overview presented herein highlights the necessity for a global, interdisciplinary approach and patient centered care in TNBC drug development. Full article
(This article belongs to the Special Issue Progress in New Agents to Treat Breast Cancer)
Show Figures

Figure 1

0 pages, 6519 KB  
Review
Fusobacterium Nucleatum in Colorectal Cancer: Relationship Among Immune Modulation, Potential Biomarkers and Therapeutic Implications
by Dalila Incognito, Giuliana Ciappina, Claudia Gelsomino, Antonio Picone, Pierluigi Consolo, Alessandra Scano, Tindara Franchina, Nicola Maurea, Vincenzo Quagliariello, Salvatore Berretta, Alessandro Ottaiano and Massimiliano Berretta
Int. J. Mol. Sci. 2025, 26(19), 9710; https://doi.org/10.3390/ijms26199710 - 6 Oct 2025
Viewed by 184
Abstract
Fusobacterium nucleatum (Fn) has been increasingly recognized as a crucial mediator of colorectal cancer (CRC) biology, particularly in microsatellite-stable (MSS) tumors, where immune checkpoint inhibitors (ICIs) have shown limited efficacy. Rather than representing a passive microbial passenger, Fn actively shapes tumor [...] Read more.
Fusobacterium nucleatum (Fn) has been increasingly recognized as a crucial mediator of colorectal cancer (CRC) biology, particularly in microsatellite-stable (MSS) tumors, where immune checkpoint inhibitors (ICIs) have shown limited efficacy. Rather than representing a passive microbial passenger, Fn actively shapes tumor behavior by adhering to epithelial cells, activating oncogenic signaling, and promoting epithelial–mesenchymal transition (EMT). At the same time, it remodels the tumor microenvironment, driving immune suppression through inhibitory receptor engagement, accumulation of myeloid-derived cells, and metabolic reprogramming of tumor-associated macrophages. These mechanisms converge to impair cytotoxic immunity and contribute to both intrinsic and acquired resistance to ICIs. Beyond immune escape, Fn interferes with conventional chemotherapy by sustaining autophagy and blocking ferroptosis, thereby linking microbial colonization to multidrug resistance. Most of these mechanisms derive from preclinical in vitro and in vivo models, where causal relationships can be inferred. In contrast, human data are mainly observational and provide correlative evidence without proving causality. No interventional clinical studies directly targeting Fn have yet been conducted. Its enrichment across the adenoma–carcinoma sequence and consistent detection in both tumor and fecal samples highlight its potential as a biomarker for early detection and patient stratification. Importantly, multidimensional stool assays that integrate microbial, genetic, and epigenetic markers are emerging as promising non-invasive tools for CRC screening. Therapeutic strategies targeting Fn are also under exploration, ranging from antibiotics and bacteriophages to multifunctional nanodrugs, dietary modulation, and natural microbiota-derived products. These approaches may not only reduce microbial burden but also restore immune competence and enhance the efficacy of immunotherapy in MSS CRC. Altogether, current evidence positions Fn at the intersection of microbial dysbiosis, tumor progression, and therapy resistance. A deeper understanding of its pathogenic role may support the integration of microbial profiling into precision oncology frameworks, paving the way for innovative diagnostic and therapeutic strategies in CRC. Full article
Show Figures

Figure 1

11 pages, 1113 KB  
Article
Incidence of Hypothyroidism and Thyroid Function Monitoring After Immune Checkpoint Inhibitor Therapy Completion for Lung Cancer: A Nationwide Analysis of a Japanese Claims Database
by Hiroaki Ohta, Hinako Tsugane and Takeo Yasu
Curr. Oncol. 2025, 32(10), 558; https://doi.org/10.3390/curroncol32100558 - 4 Oct 2025
Viewed by 228
Abstract
Immune checkpoint inhibitors (ICIs) improve lung cancer prognosis but are associated with immune-related adverse events, most commonly thyroid dysfunction. While prior studies and guidelines have focused on thyroid dysfunction during ICI therapy, data on hypothyroidism and its monitoring after ICI therapy remain limited. [...] Read more.
Immune checkpoint inhibitors (ICIs) improve lung cancer prognosis but are associated with immune-related adverse events, most commonly thyroid dysfunction. While prior studies and guidelines have focused on thyroid dysfunction during ICI therapy, data on hypothyroidism and its monitoring after ICI therapy remain limited. We aimed to investigate hypothyroidism incidence and implementation of thyroid function monitoring after ICI therapy completion in patients with lung cancer. We conducted a retrospective observational study using the DeSC claims database of approximately 12 million individuals in Japan. Patients with lung cancer who received ICI therapy between April 2014 and August 2023 were included; those with a history of thyroid hormone replacement or insufficient follow-up were excluded. Among 6883 eligible patients, 277 (4.0%) developed hypothyroidism requiring hormone replacement post-ICI therapy completion (median onset, 67.0 d). Risk factors included ICI plus bevacizumab therapy and a history of myasthenia gravis, while steroid use for ≥28 d during ICI therapy lowered the risk. Post-ICI therapy completion thyroid monitoring was performed in 73.7% of patients, with test date distribution showing a median of 126.0 d and mode of 21.0 d. Hypothyroidism was frequently found to develop within 2 months post-ICI therapy completion, highlighting the need for continued thyroid monitoring and prospective studies to establish optimal surveillance strategies. Full article
(This article belongs to the Section Thoracic Oncology)
Show Figures

Figure 1

10 pages, 741 KB  
Article
Lack of Association Between Glucose Homeostasis and Immune Checkpoint Inhibitor Outcomes: A Retrospective Institutional Review
by Joy Justice, Hannah Burnette, Rebecca Irlmeier, Fei Ye and Douglas B. Johnson
Cancers 2025, 17(19), 3230; https://doi.org/10.3390/cancers17193230 - 4 Oct 2025
Viewed by 220
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) have revolutionized outcomes for patients with melanoma. As such, it is important to understand factors that may influence response as well as toxicity to these therapies. Impaired glucose control is often a sign of pathologic inflammation and may [...] Read more.
Background/Objectives: Immune checkpoint inhibitors (ICIs) have revolutionized outcomes for patients with melanoma. As such, it is important to understand factors that may influence response as well as toxicity to these therapies. Impaired glucose control is often a sign of pathologic inflammation and may alter immune system regulation, but it is unclear whether glucose control impacts patients with melanoma on ICIs. Methods: After reviewing patients with melanoma treated with ICIs at our institution between 2014 and 2024, we assessed whether longitudinal glucose control is associated with patient outcomes (response, progression-free survival, overall survival, and treatment toxicity) during ICI therapy. Results: There was no significant difference in baseline glucose values between responders and non-responders (102.5 vs. 106.0, p = 0.093). Having a baseline glucose over 200 or any glucose over 200 was not significantly associated with response (p = 0.79, p = 0.20), progression-free survival (p = 0.64, p = 0.45), overall survival (p = 0.56, p = 0.36), or toxicity (p = 0.29, p = 0.11). Although a diagnosis of diabetes mellitus was not significantly associated with response (p = 0.84), progression-free survival (p = 0.12), or toxicity (p = 0.11), it was associated with improved overall survival (p = 0.0034) in the small number of patients with diabetes. Conclusions: Overall, we observed that glucose control was not strongly associated with efficacy or toxicity in patients treated with ICIs. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

25 pages, 3625 KB  
Article
Checkpoint Imbalance in Primary Glomerulopathies: Comparative Insights into IgA Nephropathy and Membranoproliferative Glomerulonephritis
by Sebastian Mertowski, Paulina Mertowska, Milena Czosnek, Iwona Smarz-Widelska, Wojciech Załuska and Ewelina Grywalska
Cells 2025, 14(19), 1551; https://doi.org/10.3390/cells14191551 - 3 Oct 2025
Viewed by 268
Abstract
Introduction: Primary glomerulopathies are immune-driven kidney diseases. IgA nephropathy (IgAN) and membranoproliferative glomerulonephritis (MPGN) are prevalent entities with a risk of chronic progression. Immune checkpoints, such as PD-1/PD-L1, CTLA-4/CD86, and CD200R/CD200, regulate activation and tolerance in T, B, and NK cells, and also [...] Read more.
Introduction: Primary glomerulopathies are immune-driven kidney diseases. IgA nephropathy (IgAN) and membranoproliferative glomerulonephritis (MPGN) are prevalent entities with a risk of chronic progression. Immune checkpoints, such as PD-1/PD-L1, CTLA-4/CD86, and CD200R/CD200, regulate activation and tolerance in T, B, and NK cells, and also exist in soluble forms, reflecting systemic immune balance. Objective: To compare immune checkpoint profiles in IgAN and MPGN versus healthy volunteers (HV) through surface expression, soluble serum levels, and PBMC transcripts, with attention to sex-related differences and diagnostic value assessed by ROC curves. Materials and Methods: Ninety age-matched subjects were studied: IgAN (n = 30), MPGN (n = 30), HV (n = 30). Flow cytometry evaluated checkpoint expression on CD4+/CD8+ T cells, CD19+ B cells, and NK cells. ELISA quantified sPD-1, sPD-L1, sCTLA-4, sCD86, sCD200, sCD200R; PBMC transcript levels were assessed. Group comparisons, sex stratification, and ROC analyses were performed. Results: Lymphocyte distributions were preserved, but IgAN patients showed anemia and impaired renal function, while MPGN patients had greater proteinuria and dyslipidemia. GN patients displayed increased PD-1/PD-L1 and CD200R/CD200, with reduced CTLA-4/CD86, compared to HV. Serum analysis revealed elevated sPD-1, sPD-L1, sCD200, sCD200R and decreased sCTLA-4, sCD86. PBMC transcripts paralleled these trends, with PD-1/PD-L1 mainly increased in MPGN. Sex had minimal impact. ROC analyses showed strong GN vs. HV discrimination by CD19+CTLA-4+, PD-1/PD-L1, and CD200/CD200R, but limited ability to separate IgAN from MPGN. Conclusions: IgAN and MPGN share a sex-independent checkpoint signature: PD-1/PD-L1 and CD200R/CD200 upregulation with CTLA-4/CD86 downregulation. CD19+, CTLA-4+, and soluble PD-1/PD-L1/CD200(R) emerge as promising biomarkers requiring further validation. Full article
(This article belongs to the Special Issue Kidney Disease: The Role of Cellular Mechanisms in Renal Pathology)
Show Figures

Figure 1

19 pages, 2498 KB  
Article
Multi-Modal Biomarker Profiling of Tumor Microenvironment and Genomic Alterations to Enhance Immunotherapy Stratification in Melanoma
by Meshack Bida, Thabiso Victor Miya, Tebogo Marutha, Rodney Hull, Mohammed Alaouna and Zodwa Dlamini
Curr. Issues Mol. Biol. 2025, 47(10), 821; https://doi.org/10.3390/cimb47100821 - 3 Oct 2025
Viewed by 288
Abstract
Tumor mutational burden (TMB) and tumor-infiltrating lymphocytes (TILs) are key biomarkers for predicting immunotherapy responses in cutaneous melanoma. The discordance between brisk TIL morphology and absent cytokine signals complicates immune profiling. We examined the interactions between TMB, TIL patterns, cytokine expression, and genomic [...] Read more.
Tumor mutational burden (TMB) and tumor-infiltrating lymphocytes (TILs) are key biomarkers for predicting immunotherapy responses in cutaneous melanoma. The discordance between brisk TIL morphology and absent cytokine signals complicates immune profiling. We examined the interactions between TMB, TIL patterns, cytokine expression, and genomic alterations to uncover immune escape mechanisms and refine prognostic tools. A structure-based BRAF druggability analysis was performed to anchor the genomic findings in a therapeutic context. Primary cutaneous melanoma cases (N = 205) were classified as brisk (n = 65), non-brisk (n = 60), or absent TILs (n = 80) according to the American association for cancer research (AACR) guidelines. Inter-observer concordance was measured using intraclass correlation. Tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) levels were graded using immunohistochemistry. Eleven brisk TIL cases lacking TNF-α expression were analyzed using the (Illumina TruSight Oncology 500, Illumina-San Diego, CA, USA). Dabrafenib docking to the BRAF ATP site was performed with Glide SP/XP and rescored with Prime MM-GBSA. Brisk TILs lacking cytokine signals suggested post-translational silencing of TNF-α/IFN-γ. Among the 11 profiled cases, eight exhibited high TMB and copy number alterations, with enrichment of nine metastasis/immune regulation genes. Inter-observer concordance was high (absent TILs, 95%; brisk TILs, 90.7%). BRAF docking yielded a canonical type-I pose and strong ATP pocket engagement (ΔG_bind −84.93 kcal·mol−1). Single biomarkers are insufficient for diagnosis. A multiparametric framework combining histology, cytokine immunohistochemistry (IHC), and genomic profiling enhances stratification and reveals immune escape pathways, with BRAF modeling providing a mechanistic anchor for the targeted therapy. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

24 pages, 2737 KB  
Article
Modulation of the Therapeutic Resistance in SK-MEL-24 Human Melanoma Cells by Combined Treatments
by Lucica Madalina Bolovan, Marieta Elena Panait, Mirela Mihaila, Antonela Busca, Adina Elena Stanciu, Marinela Bostan, Lorelei Irina Brasoveanu and Camelia Mia Hotnog
Appl. Sci. 2025, 15(19), 10703; https://doi.org/10.3390/app151910703 - 3 Oct 2025
Viewed by 150
Abstract
Melanoma represents a worldwide public health problem due to its high incidence and mortality rates. Despite the advances in melanoma therapy, not all patients respond to single or combined therapy because of primary or acquired resistance to the anti-tumor agents. Recently, positive results [...] Read more.
Melanoma represents a worldwide public health problem due to its high incidence and mortality rates. Despite the advances in melanoma therapy, not all patients respond to single or combined therapy because of primary or acquired resistance to the anti-tumor agents. Recently, positive results have been reported since the specific monoclonal antibodies, such as Ipilimumab (Ipi) and Nivolumab (Niv), were included in therapeutic protocols as immune checkpoint inhibitors. The evolution of neoplastic diseases and the therapeutic approaches in cancer involve several biological processes, including apoptosis, DNA progression through cell cycle phases, the release of pro-inflammatory cytokines, and changes in the expression of melanoma genes. Therefore, the potential modulation of these processes and associated molecules, due to single or combined treatments with oncolytic drugs like Carboplatin and Paclitaxel, checkpoint inhibitors such as Ipi and Niv, or natural bioactive compounds like Resveratrol or Quercetin, could represent a great benefit in melanoma treatment, contributing to the decrease or even reversal of the drug resistance in melanoma cells. Full article
Show Figures

Figure 1

30 pages, 7258 KB  
Review
Cancer-Induced Cardiac Dysfunction: Mechanisms, Diagnostics, and Emerging Therapeutics in the Era of Onco-Cardiology
by Sarama Saha, Praveen K. Singh, Partha Roy, Vasa Vemuri, Mariusz Z. Ratajczak, Mahavir Singh and Sham S. Kakar
Cancers 2025, 17(19), 3225; https://doi.org/10.3390/cancers17193225 - 3 Oct 2025
Viewed by 174
Abstract
Cancer-induced cardiac dysfunction has become a major clinical challenge as advances in cancer therapies continue to extend patient survival. Once regarded as a secondary concern, cardiotoxicity is now recognized as a leading contributor to morbidity and mortality among cancer patients and survivors. Its [...] Read more.
Cancer-induced cardiac dysfunction has become a major clinical challenge as advances in cancer therapies continue to extend patient survival. Once regarded as a secondary concern, cardiotoxicity is now recognized as a leading contributor to morbidity and mortality among cancer patients and survivors. Its pathophysiology is multifactorial, involving systemic inflammation (e.g., TNF-α, IL-6), oxidative stress driven by reactive oxygen species (ROS), neurohormonal imbalances (e.g., angiotensin II, endothelin-1), and metabolic disturbances. These mechanisms collectively promote cardiomyocyte apoptosis, atrophy, mitochondrial dysfunction, and impaired cardiac output. Cardiac complications may arise directly from cancer itself or as adverse effects of oncologic therapies such as anthracyclines, trastuzumab, and immune checkpoint inhibitors. These agents have been linked to heart failure (HF), systolic dysfunction, and cardiac atrophy, often progressing insidiously and underscoring the importance of early detection and careful monitoring. Current preventive and therapeutic strategies include pharmacological interventions such as ACE inhibitors, beta-blockers, statins, dexrazoxane, and endothelin receptor antagonists like atrasentan. Emerging compounds, particularly Withaferin A (WFA), have shown potential through their anti-inflammatory and cardiac protective properties. In addition, antioxidants and lifestyle modifications may provide supplementary cardioprotective benefits, while interventional cardiology procedures are increasingly considered in selected patients. Despite encouraging progress, standardized treatment protocols and robust long-term outcome data remain limited. Given the heterogeneity of cancer types and cardiovascular responses, a personalized and multidisciplinary approach is essential. Continued research and close collaboration between oncologists, cardiologists, and basic scientists will be the key to advancing care, reducing treatment-related morbidity, and ensuring that improvements in cancer survival are matched by preservation of cardiovascular health. Full article
(This article belongs to the Special Issue Cancer Induced Organ Dysfunctions (Cachexia))
Show Figures

Figure 1

28 pages, 1887 KB  
Review
Design and Efficacy of Oncolytic Viruses and Antitumor Vaccines: A Dead End in the Immunotherapy of Pancreatic Cancer?
by Eduard Achim, Elena Pîrlici, Cecilia Cristea and Mihaela Tertis
Int. J. Mol. Sci. 2025, 26(19), 9640; https://doi.org/10.3390/ijms26199640 - 2 Oct 2025
Viewed by 244
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies, marked by late diagnosis, limited responsiveness to conventional therapies, and an immunosuppressive tumor microenvironment. While immunotherapy has transformed treatment paradigms in several cancers, its efficacy in PDAC has been minimal. Oncolytic viruses and [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies, marked by late diagnosis, limited responsiveness to conventional therapies, and an immunosuppressive tumor microenvironment. While immunotherapy has transformed treatment paradigms in several cancers, its efficacy in PDAC has been minimal. Oncolytic viruses and therapeutic cancer vaccines have emerged as promising immunotherapeutic strategies designed to stimulate robust, tumor-specific immune responses and reshape the immune landscape. However, despite encouraging preclinical data, clinical translation in PDAC has been largely disappointing. This review critically evaluates the design, delivery, and efficacy of oncolytic virotherapy and cancer vaccines in PDAC, examining barriers such as stromal desmoplasia, immune exclusion, and tumor heterogeneity. We also explore combination strategies integrating checkpoint inhibitors, chemotherapy, radiotherapy, and stromal modulation to overcome resistance. Ultimately, the viability of these approaches hinges on a clearer understanding of their mechanistic limitations and the refinement of delivery platforms. These factors will determine whether oncolytic viruses and cancer vaccines can be successfully repositioned within the therapeutic arsenal or warrant reevaluation in the evolving landscape of PDAC treatment. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Back to TopTop