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

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

Search Results (3,085)

Search Parameters:
Keywords = antitumor immunity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 8340 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
Show Figures

Figure 1

16 pages, 2205 KB  
Article
IL-2 Receptor Expression in Renal Cell Carcinoma Cells: IL-2 Influences Cell Survival and Induces Cell Death
by Sophie Grigolo, Isabelle Fellay and Luis Filgueira
Curr. Issues Mol. Biol. 2025, 47(10), 830; https://doi.org/10.3390/cimb47100830 (registering DOI) - 9 Oct 2025
Abstract
Renal cell carcinoma (RCC) is the most common form of kidney cancer in adults. Immunotherapy, such as the application of interleukin-2 (IL-2), is a crucial treatment. It is known that IL-2 is involved in the upregulation of the anti-tumor immune response; however, a [...] Read more.
Renal cell carcinoma (RCC) is the most common form of kidney cancer in adults. Immunotherapy, such as the application of interleukin-2 (IL-2), is a crucial treatment. It is known that IL-2 is involved in the upregulation of the anti-tumor immune response; however, a direct action of IL-2 on RCC cells has not yet been demonstrated. In this project, we aimed to investigate the expression and the functionality of the IL-2Rα, IL-2Rβ, and IL-2Rγ subunits on the four human RCC cell lines A-498, ACHN, Caki-1, and Caki-2. The expression of the three subunit genes was investigated via PCR, agarose gel of PCR products, Western blot, and flow cytometry. IL-2R functionality was assessed in RCC cells cultured with or without rhIL-2 using MTT and BrdU assays to investigate cell viability and proliferation; LDH assays, Live-or-Dye staining, and Annexin V/PI staining to study cell death; and Western blot to detect apoptotic markers, cleaved PARP, and cleaved caspases 3 and 9. Expression of IL-2Rα, IL-2Rβ, and IL-2Rγ subunits in the four cell lines was observed at the protein level with Western blot. Flow cytometry confirmed the cell-surface expression of IL-2Rα, IL-2Rβ, and IL-2Rγ subunits. In addition, we observed that rhIL-2 influenced cell survival/proliferation and cell death, depending on the cell line. We conclude that IL-2R is functional in RCC cells and that rhIL-2 could be used as a therapeutic option to act directly on RCC cells. However, further studies are required to elucidate the signaling pathways triggered by the IL-2-receptor binding on RCC cells. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

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

14 pages, 1681 KB  
Review
Mechanisms of Oncolytic Virus-Induced Multi-Modal Cell Death and Therapeutic Prospects
by Jinzhou Xu, Chenqian Liu, Ye An, Jianxuan Sun, Shaogang Wang and Qidong Xia
Int. J. Mol. Sci. 2025, 26(19), 9770; https://doi.org/10.3390/ijms26199770 (registering DOI) - 7 Oct 2025
Viewed by 90
Abstract
Cancer is a major challenge to global health, and its incidence rate and mortality are expected to continue to rise in the coming decades. Traditional treatment methods such as surgery, radiotherapy, and chemotherapy have limitations, which has prompted people to explore new treatment [...] Read more.
Cancer is a major challenge to global health, and its incidence rate and mortality are expected to continue to rise in the coming decades. Traditional treatment methods such as surgery, radiotherapy, and chemotherapy have limitations, which has prompted people to explore new treatment strategies. As a promising therapeutic approach, oncolytic viruses can selectively target and lyse tumor cells while avoiding damage to normal tissues. This article systematically reviews the mechanisms by which oncolytic viruses induce various forms of cell death, including apoptosis, autophagy, pyroptosis, necroptosis, and ferroptosis. We explored the direct killing effect of oncolytic viruses and their ability to activate local and systemic antitumor immunity, with a focus on the latest developments in the clinical application of oncolytic viruses, such as the development of novel recombinant viruses. In addition, we also analyzed strategies to enhance the efficacy of oncolytic viruses through gene modification, combination therapy, and targeted delivery systems. A deeper understanding of the multiple mechanisms of action of oncolytic viruses can help us develop more effective and personalized cancer treatment plans. Future research should focus on optimizing oncolytic viruses to overcome tumor drug resistance and improve patient prognosis, making them an important pillar of cancer treatment. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Graphical abstract

16 pages, 3680 KB  
Article
Hsp70 Peptides Induce TREM-1-Dependent and TREM-1-Independent Activation of Cytotoxic Lymphocytes
by Daria M. Yurkina, Elena A. Romanova, Aleksandr S. Chernov, Irina S. Gogleva, Anna V. Tvorogova, Alexey V. Feoktistov, Rustam H. Ziganshin, Denis V. Yashin and Lidia P. Sashchenko
Int. J. Mol. Sci. 2025, 26(19), 9750; https://doi.org/10.3390/ijms26199750 - 7 Oct 2025
Viewed by 99
Abstract
The novel data show that the Hsp70 protein is a potent activator of the immune system. Using limited trypsinolisis, we have identified the epitopes of Hsp70 responsible for TREM-1-dependent and TREM-1-independent cytotoxicity. The 11aa N9 peptide (AMTKDNNLLGR) contains nine amino acids that correspond [...] Read more.
The novel data show that the Hsp70 protein is a potent activator of the immune system. Using limited trypsinolisis, we have identified the epitopes of Hsp70 responsible for TREM-1-dependent and TREM-1-independent cytotoxicity. The 11aa N9 peptide (AMTKDNNLLGR) contains nine amino acids that correspond to the amino acid sequence of the known TKD peptide. Also, like TKD, this peptide does not interact with the TREM-1 receptor but activates CD94+ NK cells that kill tumor cells by secreting granzymes and inducing apoptosis. The 16aa peptide N7 (SDNQPGVLIQVYEGEK) interacts with the TREM-1 receptor and induces the activation of NK cells and cytotoxic T lymphocytes at different time points. T-lymphocytes activated by this peptide induce two alternative processes of cell death in HLA-negative tumor cells, apoptosis and necroptosis, through the interaction of the FasL lymphocyte with the Fas receptor of the tumor cell. A shortened fragment of this peptide, N7.1 (SDNQPGVL), has been identified that inhibits the interaction of TREM-1 with its ligands. This peptide has shown protective effects in the development of sepsis in mice. The results obtained can be used in antitumor and anti-inflammation therapy. Full article
Show Figures

Graphical abstract

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 163
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

26 pages, 1201 KB  
Review
The Tumor Environment in Peritoneal Carcinomatosis and Malignant Pleural Effusions: Implications for Therapy
by Paige O. Mirsky, Patrick L. Wagner, Maja Mandic-Popov, Vera S. Donnenberg and Albert D. Donnenberg
Cancers 2025, 17(19), 3217; https://doi.org/10.3390/cancers17193217 - 2 Oct 2025
Viewed by 430
Abstract
Peritoneal carcinomatosis (PC) and malignant pleural effusions (MPE) are two common complications of cancers metastatic to the respective body cavities. A PC diagnosis indicates metastasis to the tissue lining the abdominal cavity and is most common in patients with gastrointestinal and gynecological cancers. [...] Read more.
Peritoneal carcinomatosis (PC) and malignant pleural effusions (MPE) are two common complications of cancers metastatic to the respective body cavities. A PC diagnosis indicates metastasis to the tissue lining the abdominal cavity and is most common in patients with gastrointestinal and gynecological cancers. It is often accompanied by ascites, an accumulation of serous fluid in the abdomen. MPE presents as the accumulation of fluid in the space between the lungs and chest wall. It is a common terminal event in patients diagnosed with breast cancer, lung cancer, lymphoma, and mesothelial cancers, and less commonly, in a wide variety of other epithelial cancers. Due to the aggressive nature of cavitary tumors, the outcome of current treatments for both PC and MPE remains bleak. Although PC and MPE are characteristically affected by different sets of primary tumors (lung/breast/mesothelioma for MPE and gynecologic/gastrointestinal for PC), their environments share common cytokines and cellular components. Owing to the unique cytokine and chemokine content, this environment promotes aggressive tumor behavior and paradoxically both recruits and suppresses central memory and effector memory T cells. The cellular and secretomic complexity of the cavitary tumor environment renders most currently available therapeutics ineffective but also invites approaches that leverage the robust T-cell infiltrate while addressing the causes of local suppression of anti-tumor immunity. Interactions between the heterogeneous components of the tumor environment are an area of active research. We highlight the roles of the immune cell infiltrate, stromal cells, and tumor cells, and the soluble products that they secrete into their environment. A more comprehensive understanding of the cavitary tumor environment can be expected to lead to better immunotherapeutic approaches to these devastating conditions. Full article
(This article belongs to the Special Issue Recent Advances in Peritoneal Carcinomatosis)
Show Figures

Figure 1

26 pages, 25630 KB  
Article
Constructing a Pan-Cancer Prognostic Model via Machine Learning Based on Immunogenic Cell Death Genes and Identifying NT5E as a Biomarker in Head and Neck Cancer
by Luojin Wu, Qing Sun, Atsushi Kitani, Xiaorong Zhou, Liming Mao and Mengmeng Sang
Curr. Issues Mol. Biol. 2025, 47(10), 812; https://doi.org/10.3390/cimb47100812 - 1 Oct 2025
Viewed by 307
Abstract
Immunogenic cell death (ICD) is a specialized form of cell death that triggers antitumor immune responses. In tumors, ICD promotes the release of tumor-associated and tumor-specific antigens, thereby reshaping the immune microenvironment, restoring antitumor immunity, and facilitating tumor eradication. However, the regulatory mechanisms [...] Read more.
Immunogenic cell death (ICD) is a specialized form of cell death that triggers antitumor immune responses. In tumors, ICD promotes the release of tumor-associated and tumor-specific antigens, thereby reshaping the immune microenvironment, restoring antitumor immunity, and facilitating tumor eradication. However, the regulatory mechanisms of ICD and its immunological effects vary across tumor types, and a comprehensive understanding remains limited. We systematically analyzed the expression of 34 ICD-related regulatory genes across 33 tumor types. Differential expression at the RNA, copy number variation (CNV), and DNA methylation levels was assessed in relation to clinical features. Associations between patient survival and RNA expression, CNVs, single-nucleotide variations (SNVs), and methylation were evaluated. Patients were stratified into immunological subtypes and further divided into high- and low-risk groups based on optimal prognostic models built using a machine learning framework. We explored the relationships between ICD-related genes and immune cell infiltration, stemness, heterogeneity, immune scores, immune checkpoint and regulatory genes, and subtype-specific expression patterns. Moreover, we examined the influence of immunotherapy and anticancer immune responses, applied three machine learning algorithms to identify prognostic biomarkers, and performed drug prediction and molecular docking analyses to nominate therapeutic targets. ICD-related genes were predominantly overexpressed in ESCA, GBM, KIRC, LGG, PAAD, and STAD. RNA expression of most ICD-related genes was associated with poor prognosis, while DNA methylation of these genes showed significant survival correlations in LGG and UVM. Prognostic models were successfully established for 18 cancer types, revealing intrinsic immune regulatory mechanisms of ICD-related genes. Machine learning identified several key prognostic biomarkers across cancers, among which NT5E emerged as a predictive biomarker in head and neck squamous cell carcinoma (HNSC), mediating tumor–immune interactions through multiple ligand–receptor pairs. This study provides a comprehensive view of ICD-related genes across cancers, identifies NT5E as a potential biomarker in HNSC, and highlights novel targets for predicting immunotherapy response and improving clinical outcomes in cancer patients. Full article
(This article belongs to the Special Issue Challenges and Advances in Bioinformatics and Computational Biology)
Show Figures

Figure 1

13 pages, 2422 KB  
Article
Co-Targeting PD-1 and IL-33/ST2 Pathways for Enhanced Acquired Anti-Tumor Immunity in Breast Cancer
by Marina Z. Jovanović, Milena Jurišević, Milan Jovanović, Nevena Gajović, Miodrag Jocić, Marina M. Jovanović, Boško Milev, Krstina Doklestić Vasiljev and Ivan Jovanović
Int. J. Mol. Sci. 2025, 26(19), 9600; https://doi.org/10.3390/ijms26199600 - 1 Oct 2025
Viewed by 239
Abstract
Despite advances in immunotherapy, the treatment of breast cancer still remains a major global problem. In a previous study, we showed that co-blockade of Interleukin-33/ST2 and Programmed death-1/Programmed death-ligand (PD-1/PD-L) signaling pathways strongly slows progression by enhancing the antitumor capacity of natural killer [...] Read more.
Despite advances in immunotherapy, the treatment of breast cancer still remains a major global problem. In a previous study, we showed that co-blockade of Interleukin-33/ST2 and Programmed death-1/Programmed death-ligand (PD-1/PD-L) signaling pathways strongly slows progression by enhancing the antitumor capacity of natural killer (NK) cells. The main aim of this study is to elucidate the exact effect of co-blockade on the T lymphocyte and macrophage effector cells. 4T1 cells were used to induct breast cancer in female BALB/C and BALB/C ST2−/− mice. The mice, both BALB/C and BALB/C ST2−/−, were treated with anti-PD-1 antibody on certain days. After the mice were sacrificed, T cells and macrophages were analyzed using flow cytometry; dual co-blockade increased significantly the percentage of M1 macrophages in the tumor microenvironment, followed by an increase in expression of CD86+ and TNFα+. T cell accumulation was significantly higher in the spleen and within the tumor microenvironment, with elevation in activation markers such as Interleukin-17, CD69, NKG2D, and FasL and a decrease in Interleukin-10 and FoxP3 expression. Co-blockade of the PD-1/PD-L axes and IL-33/ST2 axes shows promising results in reestablishing an effective immune response and offers a new perspective on improving immune response to breast carcinoma. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

29 pages, 3789 KB  
Article
A Novel Early Memory-Enriched Allogeneic NKG2D CAR-T Cell Therapy Based on CRISPR/Cas9 Technology for Solid Tumors
by Cristina Aparicio, Mónica Queipo, Marina Belver, Francisco Espeso, Julia Serna-Pérez, Lucía Enríquez-Rodríguez, Carlos Acebal, Álvaro Martín-Muñoz, Antonio Valeri, Alejandra Leivas, Paula Río, Daniel J. Powell, Rosa Lobo-Valentín, David Arrabal, Joaquín Martínez-López, Ana Sánchez, Miguel Á. de la Fuente and Margarita González-Vallinas
Cancers 2025, 17(19), 3186; https://doi.org/10.3390/cancers17193186 - 30 Sep 2025
Viewed by 449
Abstract
Background/Objectives: Chimeric Antigen Receptor (CAR)-T cell therapy has demonstrated impressive clinical results against hematological malignancies. However, currently commercialized CAR-T therapies are designed for autologous use, which entails some disadvantages, including high costs, manufacturing delays, complex standardization, and frequent production failures due to patient [...] Read more.
Background/Objectives: Chimeric Antigen Receptor (CAR)-T cell therapy has demonstrated impressive clinical results against hematological malignancies. However, currently commercialized CAR-T therapies are designed for autologous use, which entails some disadvantages, including high costs, manufacturing delays, complex standardization, and frequent production failures due to patient T cell dysfunction. Moreover, their CARs target one specific antigen, increasing the probability of antigen-negative tumor relapses. To overcome these limitations, we developed a novel NKG2D CAR-T cell therapy for allogeneic use with broad target specificity, as this CAR targets eight different ligands commonly upregulated in both solid and hematological tumors. Additionally, the manufacturing process was optimized to improve the phenotypic characteristics of the final product. Methods: Multiplex CRISPR/Cas9 technology was applied to eliminate the expression of TCR and HLA class I complexes in healthy donor T cells to reduce the risk of graft-versus-host disease and immune rejection, respectively, as well as lentiviral transduction for introducing the second-generation NKG2D-CAR. Moreover, we sought to optimize this manufacturing process by comparing the effect of different culture interleukin supplementations (IL-2, IL-7/IL-15 or IL-7/IL-15/IL-21) on the phenotypic and functional characteristics of the product obtained. Results: Our results showed that the novel CAR-T cells effectively targeted cervicouterine and colorectal cancer cells, and that those manufactured with IL-7/IL-15/IL-21 supplementation showed the most suitable characteristics among the conditions tested, considering genetic modification efficiency, cell proliferation, antitumor activity and proportion of the stem cell memory T cell subset, which is associated with enhanced in vivo CAR-T cell survival, expansion and long-term persistence. Conclusions: In summary, this new prototype of NKG2D CAR-T cell therapy for allogeneic use represents a promising universal treatment for a wide range of tumor types. Full article
Show Figures

Graphical abstract

22 pages, 4431 KB  
Review
Macrophages—Target and Tool in Tumor Treatment: Insights from Ovarian Cancer
by Małgorzata Górczak and Łukasz Kiraga
Cancers 2025, 17(19), 3182; https://doi.org/10.3390/cancers17193182 - 30 Sep 2025
Viewed by 317
Abstract
Today, science and medicine are striving to develop novel techniques for treating deadly diseases, including a wide range of cancers. Efforts are being made to better understand the molecular and biochemical mechanisms of tumor cell functioning, but a particular emphasis has recently been [...] Read more.
Today, science and medicine are striving to develop novel techniques for treating deadly diseases, including a wide range of cancers. Efforts are being made to better understand the molecular and biochemical mechanisms of tumor cell functioning, but a particular emphasis has recently been given to investigating immune cells residing in the tumor microenvironment, which may lead to revolutionary benefits in the design of new immunotherapies. Among these cells, tumor-associated macrophages (TAMs) are highly abundant and act as critical regulators of ovarian cancer progression, metastasis, and resistance to therapy. Their dual nature—as drivers of malignancy and as potential therapeutic mediators—has positioned them at the forefront of research into next-generation immunotherapies. As therapeutic targets, approaches include blocking macrophage recruitment (e.g., CSF-1/CSF-1R inhibitors), selectively depleting subsets of TAMs (e.g., via Folate Receptor Beta), or reprogramming immunosuppressive M2-like macrophages toward an anti-tumor M1 phenotype. On the other hand, macrophages can also serve as a therapeutic tool—they may be engineered to enhance anti-tumor immunity, as exemplified by the development of Chimeric Antigen Receptor Macrophages (CAR-Ms), or leveraged as delivery vehicles for targeted drug transport into the tumor microenvironment. A particularly innovative strategy involves Macrophage–Drug Conjugates (MDCs), which employs the transfer of iron-binding proteins (TRAIN) mechanism for precise intracellular delivery of therapeutic agents, thereby enhancing drug efficacy while minimizing systemic toxicity. This review integrates current knowledge of TAM biology, highlights emerging therapeutic approaches, and underscores the promise of macrophage-based interventions in ovarian cancer. By integrating macrophage-targeting strategies with advanced immunotherapeutic platforms, novel treatment paradigms may be determined that could substantially improve outcomes for patients with ovarian cancer and other solid tumors. Our work highlights that macrophages should be a particular area of research interest in the context of cancer treatment. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

30 pages, 3330 KB  
Review
Translational Insights into NK Immunophenotyping: Comparative Surface Marker Analysis and Circulating Immune Cell Profiling in Cancer Immunotherapy
by Kirill K. Tsyplenkov, Arina A. Belousova, Marina V. Zinovyeva, Irina V. Alekseenko and Victor V. Pleshkan
Int. J. Mol. Sci. 2025, 26(19), 9547; https://doi.org/10.3390/ijms26199547 - 30 Sep 2025
Viewed by 475
Abstract
Cells of the innate immune system, particularly natural killer (NK) cells, serve as the first line of defense against tumor development and play a critical role in antitumor immunity. Characterizing the immune cell pool and its functional state is essential for understanding immunotherapy [...] Read more.
Cells of the innate immune system, particularly natural killer (NK) cells, serve as the first line of defense against tumor development and play a critical role in antitumor immunity. Characterizing the immune cell pool and its functional state is essential for understanding immunotherapy mechanisms and identifying key cellular players. However, defining NK cell populations in mice, the primary model for cancer immunotherapy, is challenging due to strain-specific marker variability and the absence of a universal NK cell marker, such as human CD56. This study evaluates surface markers of NK and other peripheral blood immune cells in both humans and mice, associating these markers with specific functional profiles. Bioinformatic approaches are employed to visualize these markers, enabling rapid immunoprofiling. We explore the translational relevance of these markers in assessing immunotherapy efficacy, including their gene associations, ligand interactions, and interspecies variations. Markers compatible with rapid flow-cytometry-based detection are prioritized to streamline experimental workflows. We propose a standardized immunoprofiling strategy for monitoring systemic immune status and evaluating the effectiveness of immunotherapy in preclinical and clinical settings. This approach facilitates the design of preclinical studies that aim to identify predictive biomarkers for immunotherapy outcomes by monitoring immune status. Full article
(This article belongs to the Special Issue Recent Advances in Immunosuppressive Therapy)
Show Figures

Figure 1

28 pages, 2696 KB  
Review
Challenges and Opportunities in High-Grade Glioma Management and Imaging-Based Response Monitoring During Novel Immunotherapies
by Carlos A. Gallegos, Benjamin P. Lee, Benjamin B. Kasten, Jack M. Rogers, Carlos E. Cardenas, Jason M. Warram, James M. Markert and Anna G. Sorace
Cancers 2025, 17(19), 3176; https://doi.org/10.3390/cancers17193176 - 30 Sep 2025
Viewed by 296
Abstract
The highly heterogeneous and invasive nature characteristic of high-grade gliomas (HGG) has historically limited the efficacy of standard-of-care approaches, resulting in poor prognosis and treatment outcomes. Novel immunotherapies have shown remarkable potential to promote antitumoral immune responses and allow for long-term tumor remission. [...] Read more.
The highly heterogeneous and invasive nature characteristic of high-grade gliomas (HGG) has historically limited the efficacy of standard-of-care approaches, resulting in poor prognosis and treatment outcomes. Novel immunotherapies have shown remarkable potential to promote antitumoral immune responses and allow for long-term tumor remission. However, the complexity of the HGG tumor microenvironment and the dynamic immunological changes associated with immunotherapy response can limit the diagnostic utility of conventional magnetic resonance imaging (MRI) and positron emission tomography (PET) approaches. Consequently, distinguishing true tumor progression from immunotherapy-related effects often requires prolonged clinical follow-up over several months. To address this, novel quantitative MRI and PET-based approaches are being evaluated in preclinical studies and clinical trials. These advanced imaging methods target key biological features of the tumor microenvironment, including vascularity, cellularity, intratumoral habitats, tracer pharmacokinetics and immune infiltration, and can provide metrics to stratify patient response at earlier timepoints to support clinical decision making and improve treatment outcomes. This review highlights key HGG biological characteristics, describes standard-of-care and emerging therapeutic strategies, and discusses both conventional and advanced imaging methods to characterize immunotherapeutic responses. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

16 pages, 2980 KB  
Article
Grifola frondosa Polysaccharides Alleviated Cyclophosphamide—Induced Intestinal Injury Based on Microbiota, Metabolite and Immune Axis Modulation
by Jindi Wu, Guilu Chen, Dingfeng Chen, Haoran Zhang, Huirong Lv and Zhengshun Wen
Foods 2025, 14(19), 3376; https://doi.org/10.3390/foods14193376 - 29 Sep 2025
Viewed by 346
Abstract
Grifola frondosa polysaccharides (GFP), which possess antitumor properties, can counteract intestinal injury induced by cyclophosphamide (CTX). The objective of this research was to evaluate the efficacy of GFP in protecting the intestinal barrier of mice and investigate the mechanisms behind this effect. Using [...] Read more.
Grifola frondosa polysaccharides (GFP), which possess antitumor properties, can counteract intestinal injury induced by cyclophosphamide (CTX). The objective of this research was to evaluate the efficacy of GFP in protecting the intestinal barrier of mice and investigate the mechanisms behind this effect. Using a CTX-induced intestinal barrier injury model, we found that GFP treatment significantly alleviated body weight loss and organ atrophy, while enhancing serum IgG and IgM levels. Histological analysis showed that GFP effectively repaired the intestinal mucosal structure, increased goblet cell numbers, and led to an upregulation in the gene expression of ZO-1, Occludin, and MUC2. GFP modulated cytokine expression, including IFN-γ, IL-4, IL-10, and IL-22. According to 16S rDNA sequencing results, GFP enhanced the abundance of unclassified_Muribaculaceae while reducing the prevalence of Escherichia_Shigella. Furthermore, GFP elevated the concentrations of several metabolites, including SCFAs and pyridoxal, which are closely related to intestinal barrier protection and mucosal immunity. Overall, this study demonstrated that GFP has strong potential as an immune-enhancing adjuvant and may represent a promising intervention strategy to mitigate chemotherapy-induced intestinal injury. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Graphical abstract

16 pages, 5989 KB  
Article
Leveraging the Immune Response from LIFE Biomaterial and Photon-Flash in Pre-Clinical Pancreatic Cancer Treatment
by Michele Moreau, Katelyn Kelly, Serena Mao, Debarghya China, Girmachew Wasihun, Aditya Pandya, MohammadAli Tajik-Mansoury, Daniel Sforza, Devin Miles, Amol K. Narang, Mohammad Rezaee, Wilfred Ngwa and Kai Ding
Pharmaceutics 2025, 17(10), 1273; https://doi.org/10.3390/pharmaceutics17101273 - 29 Sep 2025
Viewed by 351
Abstract
Pre-clinical animal studies evaluating the ‘flash effect’ caused by ultra-high dose rate (≥40 Gy/s) favorably spares normal tissue from radiation-caused toxicity while maintaining anti-tumor effects like conventional (CONV) radiation. The goal of this study is to leverage an immune response resulting from the [...] Read more.
Pre-clinical animal studies evaluating the ‘flash effect’ caused by ultra-high dose rate (≥40 Gy/s) favorably spares normal tissue from radiation-caused toxicity while maintaining anti-tumor effects like conventional (CONV) radiation. The goal of this study is to leverage an immune response resulting from the treatment combination of flash radiotherapy (Flash-RT) and LIFE (liquid immunogenic fiducial eluter) biomaterial incorporating an anti-mouse CD40 monoclonal antibody to enhance the therapeutic ratio in pancreatic cancer. Methods: A small animal FLASH radiation research platform (FLASH-SARRP) was utilized to deliver both ultra-high and CONV dose-rate irradiation to treat syngeneic subcutaneous pancreatic tumors generated in 8–10-week-old male and female C57BL6 mice. The efficacy of FLASH versus CONV radiotherapy (RT) at varying doses of 5, 8, 10, and 15 Gy delivered in a single fraction was evaluated by assessing tumor growth and mice survival over time or comparing tumor weight at 10 days post-treatment. Results: Similar tumor control capability was observed by the high-dose rate and conventional RT related to the control group. Nevertheless, longer survival was observed for the FLASH group at 5 Gy compared to CONV and control at either 5 Gy, 10 Gy, or 15 Gy doses. Multiplex immunofluorescence and immunohistochemistry results showed higher T-cell infiltration within the combination of RT (either FLASH or CONV) and LIFE biomaterial-treated tumors compared to the control cohort. Conclusions: This animal study serves as an impetus for future studies leveraging the immune response using the combination of FLASH and LIFE Biomaterial to enhance the efficacy of pancreatic cancer treatment. Full article
(This article belongs to the Special Issue Smart Radiotherapy Biomaterials for Cancer Therapy and Imaging)
Show Figures

Graphical abstract

Back to TopTop