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
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
remove_circle_outline

Search Results (9,625)

Search Parameters:
Keywords = tumor–immune cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 1011 KB  
Article
Influence of Untreated and Microbially Degraded Mangrove Sediment Microplastics on Zebrafish (Danio rerio) Intestinal Histology and Immune and Antioxidant Biomarkers
by Xin-Yu Zheng, Wan Wei, Asim Muhammad, Min Zhang, Yan-Jun Chen, Jia-Hong Xie, Dan-Ju Kang and Jin-Jun Chen
Vet. Sci. 2025, 12(9), 854; https://doi.org/10.3390/vetsci12090854 - 4 Sep 2025
Abstract
MPs are pervasive pollutants in marine ecosystems, posing risks to aquatic organisms due to their small size and bioaccumulation potential. This study investigated the intestinal toxicity of MP particles extracted from mangrove sediments in zebrafish, comparing the effects before and after microbial [...] Read more.
MPs are pervasive pollutants in marine ecosystems, posing risks to aquatic organisms due to their small size and bioaccumulation potential. This study investigated the intestinal toxicity of MP particles extracted from mangrove sediments in zebrafish, comparing the effects before and after microbial degradation. Zebrafish were exposed to either undegraded MPs or microbially degraded MP extracts at concentrations of 0 (control), 2, 10, and 50 mg/L for 21 days in 10 L tanks (stocking density: 10 fish/L), with three replicate tanks per concentration. MPs were dispersed ultrasonically before addition to the water. Intestinal samples were collected on 7, 14, and 21 days for the analysis of immune response (tumor necrosis factor-alpha, TNF-α; interleukin-1 beta, IL-1β; interleukin-6, IL-6; interleukin-8, IL-8) and antioxidant activity (superoxide dismutase, SOD; catalase, CAT). Histopathological analysis revealed intestinal wall thinning, villus damage, and epithelial cell detachment in zebrafish exposed to both undegraded and degraded MP extracts; however, undegraded MPs induced more severe intestinal damage. Results indicated dynamic changes in cytokine expression: TNF-α decreased initially before increasing, while IL-1β and IL-8 first rose then declined. IL-6 peaked on day 7, dropped by day 14, and increased again on day 21. CAT expression decreased, whereas SOD increased only in the pre-degradation group. Microbial degradation reduced intestinal damage severity, with effects intensifying at higher MP exposure levels. These findings demonstrate that MPs can impair zebrafish digestive systems, but microbial degradation mitigates their toxicity. This study underscores the importance of biodegradation as a potential environmental remediation strategy and provides experimental evidence on MPs’ impact on aquatic organisms. Full article
(This article belongs to the Topic Recent Advances in Veterinary Pharmacology and Toxicology)
Show Figures

Figure 1

22 pages, 1447 KB  
Review
Photodynamic Therapy and Tumor Microenvironment-Targeting Strategies: A Novel Synergy at the Frontier of Cancer Treatment
by Stefani Torna, Vasiliki Gkretsi and Andreas Stylianou
Int. J. Mol. Sci. 2025, 26(17), 8588; https://doi.org/10.3390/ijms26178588 (registering DOI) - 3 Sep 2025
Abstract
Despite intensive worldwide research efforts and multiple available therapeutic schemes for cancer treatment, cancer still remains a challenge, rendering the need for the discovery of new therapeutic approaches imperative. Photodynamic therapy (PDT) is a novel, non-invasive anti-cancer treatment that relies on the generation [...] Read more.
Despite intensive worldwide research efforts and multiple available therapeutic schemes for cancer treatment, cancer still remains a challenge, rendering the need for the discovery of new therapeutic approaches imperative. Photodynamic therapy (PDT) is a novel, non-invasive anti-cancer treatment that relies on the generation of reactive oxygen species (ROS) that are cytotoxic to cancer cells. ROS are generated by the interaction between a photosensitizer (PS) drug, a light source (primarily a laser), and oxygen. Although PDT offers the advantage of using non-ionizing radiation and bears great therapeutic potential, it has not yet been widely adopted in clinical practice. This review summarizes the new developments in the use of PDT in combination with chemotherapy, immunotherapy, and radiotherapy, giving emphasis to the combination of PDT with a novel type of therapy that also takes into account the tumor microenvironment (TME) to enhance treatment efficacy. TME-targeting therapies include strategies like hypoxia modulation, vascular normalization, and immune cell reprogramming. Interestingly, when combined with PDT, these therapies can improve therapeutic outcomes while reducing side effects, and nanoparticle-based delivery systems have demonstrated the potential to enhance PDT selectivity and efficiency. This review highlights PDT’s enormous potential in treating various cancer types and underscores the need for continued exploration of combination therapies to maximize its clinical impact. Full article
(This article belongs to the Section Molecular Biophysics)
Show Figures

Figure 1

17 pages, 2862 KB  
Article
Recombinant Oncolytic Vesicular Stomatitis Virus Expressing Mouse Interleukin-12 and Granulocyte-Macrophage Colony-Stimulating Factor (rVSV-dM51-mIL12-mGMCSF) for Immunotherapy of Lung Carcinoma
by Anastasia Ryapolova, Margarita Zinovieva, Kristina Vorona, Bogdan Krapivin, Vasiliy Moroz, Nizami Gasanov, Ilnaz Imatdinov, Almaz Imatdinov, Roman Ivanov, Alexander Karabelsky and Ekaterina Minskaia
Int. J. Mol. Sci. 2025, 26(17), 8567; https://doi.org/10.3390/ijms26178567 (registering DOI) - 3 Sep 2025
Abstract
The unique ability of oncolytic viruses (OVs) to replicate in and destroy malignant cells while leaving healthy cells intact and activating the host immune response makes them powerful targeted anti-cancer therapeutic agents. Vesicular stomatitis virus (VSV) only causes mild and asymptomatic infection, lacks [...] Read more.
The unique ability of oncolytic viruses (OVs) to replicate in and destroy malignant cells while leaving healthy cells intact and activating the host immune response makes them powerful targeted anti-cancer therapeutic agents. Vesicular stomatitis virus (VSV) only causes mild and asymptomatic infection, lacks pre-existing immunity, can be genetically engineered for enhanced efficiency and improved safety, and has a broad cell tropism. VSV can facilitate targeted delivery of immunostimulatory cytokines for an enhanced immune response against cancer cells, thus decreasing the possible toxicity frequently observed as a result of systemic delivery. In this study, the oncolytic potency of the two rVSV versions, rVSV-dM51-GFP, delivering green fluorescent protein (GFP), and rVSV-dM51-mIL12-mGMCSF, delivering mouse interleukin-12 (mIL-12) and granulocyte-macrophage colony-stimulating factor (mGMCSF), was compared on the four murine cancer cell lines of different origin and healthy mesenchymal stem cells (MSCs) at 24 h post-infection by flow cytometry. Lewis lung carcinoma (LL/2) cells were demonstrated to be more susceptible to the lytic effects of both rVSV versions compared to melanoma (B16-F10) cells. Detection of expression levels of antiviral and pro-apoptotic genes in response to the rVSV-dM51-GFP infection by quantitative PCR (qPCR) showed lower levels of IFIT, RIG-I, and N-cadherin and higher levels of IFNβ and p53 in LL/2 cells. Subsequently, C57BL/6 mice, infused subcutaneously with the LL/2 cells, were injected intratumorally with the rVSV-dM51-mIL12-mGMCSF 7 days later to assess the synergistic effect of rVSV and immunostimulatory factors. The in vivo study demonstrated that treatment with two rVSV-dM51-mIL12-mGMCSF doses 3 days apart resulted in a tumor growth inhibition index (TGII) of over 50%. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

17 pages, 10998 KB  
Article
NONO Protein Regulates the Immune Response in Human Triple-Negative Breast Cancer Cells
by Carmelina Antonella Iannuzzi, Iris Maria Forte, Marianna Tomeo, Anna Sfera, Francesco Pagano, Riziero Esposito Abate, Michelino De Laurentiis, Antonio Giordano and Luigi Alfano
Int. J. Mol. Sci. 2025, 26(17), 8542; https://doi.org/10.3390/ijms26178542 - 2 Sep 2025
Abstract
Breast cancer (BC) remains a leading cause of cancer-related mortality worldwide, with limited treatment options for triple-negative breast cancer (TNBC). The RNA-binding protein non-POU domain-containing octamer-binding protein (NONO) has emerged as a critical regulator of tumorigenesis, but its role in immune signaling remains [...] Read more.
Breast cancer (BC) remains a leading cause of cancer-related mortality worldwide, with limited treatment options for triple-negative breast cancer (TNBC). The RNA-binding protein non-POU domain-containing octamer-binding protein (NONO) has emerged as a critical regulator of tumorigenesis, but its role in immune signaling remains unexplored. We analyzed the effect of NONO protein by modulating its expression using short hairpin RNA (shRNA) and a chemical inhibitor (R)-SKBG-1. We demonstrate that NONO depletion in MDA-MB-231 TNBC cells leads to cytoplasmic DNA accumulation, micronuclei formation, and activation of the cyclic GMP-AMP synthase—stimulator of interferon genes (cGAS/STING) pathway, resulting in enhanced modulation of the immune response. NONO-deficient cells showed increased cGAS and STING activation, Tank-binding kinase 1 (TBK1) phosphorylation, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear localization, and transcription of pro-inflammatory genes such as CC Motif Chemokine Ligand 5 (CCL5). These effects were recapitulated by pharmacological inhibition using (R)-SKBG-1, confirming NONO’s immunosuppressive function. Our findings establish NONO as a key modulator of immune activation in TNBC and suggest that its inhibition may enhance anti-tumor immunity. This work paves the way for potential combination strategies involving NONO inhibitors and immune checkpoint blockade, particularly in tumors with homologous recombination deficiencies or limited immune infiltration. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

13 pages, 787 KB  
Article
Evaluating the Predictive Value of Clinical Factors for Pembrolizumab Efficacy and Safety in Advanced NSCLC with High PD-L1 Expression (TPS ≥ 50%)
by Fedja Djordjevic, Katarina Ljujic, Nemanja Stanic, Neda Nikolic, Ivan Markovic and Jelena Spasic
J. Clin. Med. 2025, 14(17), 6200; https://doi.org/10.3390/jcm14176200 - 2 Sep 2025
Abstract
Background: Single-agent pembrolizumab represents a standard of care in the first-line treatment of patients with metastatic non-small cell lung cancer (mNSCLC) with a programmed death-ligand 1 (PD-L1) tumor proportion score (TPS) of ≥50%. Real-world evidence is of increasing importance in oncology, as [...] Read more.
Background: Single-agent pembrolizumab represents a standard of care in the first-line treatment of patients with metastatic non-small cell lung cancer (mNSCLC) with a programmed death-ligand 1 (PD-L1) tumor proportion score (TPS) of ≥50%. Real-world evidence is of increasing importance in oncology, as clinical trial inclusion criteria may not be truly reflective of the patient population seen in daily clinical practice. Methods: We performed a prospective–retrospective single-center study including 121 patients who received pembrolizumab as a first-line therapy for mNSCLC with a PD-L1 TPS ≥ 50%. Our aims were to make a comparison with published clinical trial results by assessing the efficacy and safety of pembrolizumab monotherapy in our population. We collected patient demographics, clinical characteristics of the disease, and treatment outcomes, including efficacy and safety. Results: A total of 121 patients were included, with a median follow-up of 40.77 months. The median progression-free survival in the real world (rwPFS) was 20.73 months (95% CI 12.24–29.22), and the median overall survival (OS) was 29.30 months (95% CI 16.57–42.04). Immune-mediated adverse events (irAEs) occurred in 42% of patients, with serious events (grade 3 or more) occurring in 12%. ECOG PS 2, male gender, squamous histology, pleural and visceral metastases, and treatment with corticosteroids prior to initiation of pembrolizumab were found to be negative predictors for overall survival, while the occurrence of irAEs was the predictor of longer survival. Conclusions: This study provides further real-world insights into the efficacy of pembrolizumab in a heterogeneous patient population with advanced NSCLC in a single center in Serbia. It also confirmed the value of good ECOS PS and the occurrence of irAEs as predictors of favorable outcomes. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

24 pages, 7654 KB  
Article
PSMB9 Orchestrates Tumor Immune Landscape and Serves as a Potent Biomarker for Prognosis and T Cell-Based Immunotherapy Response
by Xinran Ma, Qi Zhu, Zhiqiang Wu and Weidong Han
Curr. Issues Mol. Biol. 2025, 47(9), 712; https://doi.org/10.3390/cimb47090712 - 1 Sep 2025
Viewed by 69
Abstract
Proteasome subunit beta type-9 (PSMB9), a member of the proteasome beta subunit family, encodes the pivotal β1i component of the immunoproteasome. PSMB9 plays a crucial role in antigen processing and presentation; however, its comprehensive role in orchestrating a tumor-immune landscape and regulating the [...] Read more.
Proteasome subunit beta type-9 (PSMB9), a member of the proteasome beta subunit family, encodes the pivotal β1i component of the immunoproteasome. PSMB9 plays a crucial role in antigen processing and presentation; however, its comprehensive role in orchestrating a tumor-immune landscape and regulating the anti-tumor immune responses remains unexplored. Here we investigated the context-dependent functions of PSMB9 by integrating multi-omics data from The Cancer Genome Atlas, Genotype-Tissue Expression database, Human Protein Atlas, Tumor Immunotherapy Gene Expression Resource, and multiple other databases. Moreover, we explored the predictive value of PSMB9 in multiple immunotherapy cohorts and investigated its functional relevance in CAR-T therapy using genome-scale CRISPR/Cas9 screening, gene knockout cell line in vitro, and clinical cohort validation. We found widespread dysregulation in PSMB9 across cancers, predominantly upregulated in most malignancies and associated with advanced pathological stages in specific contexts. PSMB9 was also broadly and negatively correlated with tumor stemness indices. Crucially, PSMB9 expression was robustly linked to anti-tumor immunity by being significantly correlated with immune-pathway activation (e.g., IFN response, cytokine signaling), immune regulatory and immune checkpoint gene expression, and enhanced infiltration of T cells across nearly all tumor types. Consequently, elevated PSMB9 predicted superior response to immune checkpoint inhibitors in multiple cohorts, showing comparable predictive power to established predictive signatures. Furthermore, CRISPR/Cas9 screening identified PSMB9 loss as a novel mechanism of resistance to CD19 CAR T cell therapy, with PSMB9-deficient tumor cells exhibiting a survival advantage under CAR-T pressure, supported by trends in clinical CAR-T outcomes. Our study uncovers PSMB9 as a previously unrecognized critical regulator of the tumor immune landscape in a pan-cancer scope, whose expression orchestrates key immune processes within the tumor microenvironment and serves as a potent biomarker for patient prognosis. Critically, we first established PSMB9 as a novel prognostic indicator for both checkpoint blockade and CAR-T cell therapies, highlighting its dual role as a crucial immune modulator and a promising biomarker for guiding T cell-based immunotherapy strategies across diverse human cancers. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

22 pages, 662 KB  
Review
Endothelial Injury Following CAR-T Cell Immunotherapy for Hematological Malignancies
by Christos Demosthenous, Paschalis Evangelidis, Athanasios Gatsis, Ioannis Mitroulis, Sofia Vakalopoulou, Anna Vardi, Stefania Bountoura, Ioanna Sakellari and Eleni Gavriilaki
Cancers 2025, 17(17), 2876; https://doi.org/10.3390/cancers17172876 - 1 Sep 2025
Viewed by 186
Abstract
Chimeric antigen receptor-T (CAR-T) cell immunotherapy constitutes a cornerstone in the management of patients with relapsed/refractory B-cell lineage lymphoid malignancies. Toxicities such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hematotoxicity (ICAHT) have been recognized in the post-infusion period. [...] Read more.
Chimeric antigen receptor-T (CAR-T) cell immunotherapy constitutes a cornerstone in the management of patients with relapsed/refractory B-cell lineage lymphoid malignancies. Toxicities such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hematotoxicity (ICAHT) have been recognized in the post-infusion period. The initial interplay between CAR-T cells and tumor cells, followed by cytokine release and the bystander activation of the innate immunity cells, result in endothelial cell injury. In the current review, the ongoing research regarding endothelial injury in CAR-T cell recipients is summarized. Various markers of endothelial injury have been investigated in CAR-T cell recipients, including markers of complement activation, such as soluble C5b-9, endothelial dysfunction (angiopoietin-2, VCAM1, ICAM-1), inflammation, and thrombosis (von Willebrand antigen, ADAMTS13, thrombomodulin). The expression level of these endothelial injury markers has been identified as impaired in CAR-T cell recipients, not only when compared with healthy controls but also among patients with severe CRS/ICANS and those with mild toxicities or without toxicities. Furthermore, the Endothelial Activation and Stress Index (EASIX) and modified versions of this score, calculated in the pre- and early post-infusion period, seem to predict development of severe toxicities, ICAHT, and, thus, poor overall survival in CAR-T cell patients. More data concerning the role of these endothelial injury markers and clinical outcomes in CAR-T cell settings are essential. Full article
Show Figures

Figure 1

16 pages, 2125 KB  
Article
Granzyme B PET Imaging Enables Early Assessment of Immunotherapy Response in a Humanized Melanoma Mouse Model
by Priska Summer, Naomi Gallon, Niklas Bulmer, Umar Mahmood and Pedram Heidari
Pharmaceuticals 2025, 18(9), 1309; https://doi.org/10.3390/ph18091309 - 31 Aug 2025
Viewed by 303
Abstract
Background/Objectives: This study evaluated a novel PET tracer, 68Ga-NOTA-CYT-200, which targets human granzyme B (GZB) as a biomarker for cytotoxic T-cell activation in a clinically relevant model of melanoma-bearing mice with a humanized immune system treated with immune checkpoint inhibitor (ICI) [...] Read more.
Background/Objectives: This study evaluated a novel PET tracer, 68Ga-NOTA-CYT-200, which targets human granzyme B (GZB) as a biomarker for cytotoxic T-cell activation in a clinically relevant model of melanoma-bearing mice with a humanized immune system treated with immune checkpoint inhibitor (ICI) therapy. Methods: The binding affinity of the tracer was determined using an enzymatic colorimetric assay. Tumor-bearing humanized NSG mice underwent PET imaging before and during ICI monotherapy or combination therapy to assess 68Ga-NOTA-CYT-200 uptake within tumors and other organs. The tumor growth was carefully monitored. The treatment response was evaluated based on the percentage change in tumor size at days 5 and 15 after the treatment started. A tracer biodistribution study and immunohistochemical staining of the tumors and organs were also performed. Results: The inhibition constant (Ki) of 68Ga-NOTA-CYT-200 was estimated at 4.2 nM. PET imaging showed a significantly higher 68Ga-NOTA-CYT-200 uptake in mice receiving the combination therapy compared to those receiving monotherapy or a vehicle (p < 0.0001 or p = 0.0005, respectively), which correlated with the greatest reduction in tumor size in the combination ICI group. Regardless of treatment, the responders presented with a significantly higher 68Ga-NOTA-CYT-200 uptake at days 4 or 7 after the treatment began (p = 0.0002 and p = 0.0109, respectively). An increased uptake of 68Ga-NOTA-CYT-200, especially in the intestines and liver within the combination ICI group, suggested immune-related adverse events (IrAEs). Conclusions: Our study demonstrates that 68Ga-NOTA-CYT-200 PET imaging can predict the early treatment response in melanoma models treated with ICI and may also help in detecting IrAEs. Full article
Show Figures

Figure 1

29 pages, 3437 KB  
Review
Targeting the Purinergic Axis with Phenolic Compounds to Disrupt the Oxidative-Inflammatory Cycle in Thyroid Cancer
by Júlia Leão Batista Simões and Margarete Dulce Bagatini
Int. J. Mol. Sci. 2025, 26(17), 8474; https://doi.org/10.3390/ijms26178474 - 31 Aug 2025
Viewed by 165
Abstract
Thyroid cancer (TC), the most prevalent endocrine neoplasia, has shown a progressive incidence, highlighting the need for new therapeutic approaches—especially for radioiodine-refractory cases, often associated with mutations in genes such as BRAF, RAS, and TP53. This review proposes a mechanistic [...] Read more.
Thyroid cancer (TC), the most prevalent endocrine neoplasia, has shown a progressive incidence, highlighting the need for new therapeutic approaches—especially for radioiodine-refractory cases, often associated with mutations in genes such as BRAF, RAS, and TP53. This review proposes a mechanistic model that highlights two interrelated characteristics of the tumor microenvironment (TME): redox imbalance and chronic inflammation, key elements in tumor progression and treatment resistance. Thus, natural phenolic compounds, such as curcumin, quercetin, resveratrol, and epigallocatechin gallate (EGCG), function not as simple antioxidants but as pleiotropic agents that reprogram the TME. A central mechanism of action for these compounds is the modulation of the purinergic axis (CD39/CD73/adenosine), a critical immune-metabolic checkpoint. By selectively inducing lethal oxidative stress in tumor cells, suppressing pro-survival inflammatory pathways—such as that mediated by nuclear factor kappa B (NF-κB)—and destabilizing the immunosuppressive shield conferred by adenosine, certain phytochemicals demonstrate the potential to restore immune surveillance and promote tumor apoptosis. In this context, a critical analysis of the evidence related to targeting purinergic signals becomes essential, since pharmacological reinforcement of this pathway, especially when combined with immunotherapies based on immune checkpoint blockade, emerges as a promising strategy for overcoming therapeutic resistance. Full article
(This article belongs to the Special Issue Correlation Between Oxidative Stress and Inflammation)
Show Figures

Figure 1

19 pages, 2263 KB  
Article
T-Cell Epitope-Based SARS-CoV-2 DNA Vaccine Encoding an Antigen Fused with Type 1 Herpes Simplex Virus Glycoprotein D (gD)
by Luana Raposo de Melo Moraes Aps, Aléxia Adrianne Venceslau-Carvalho, Carla Longo de Freitas, Bruna Felício Milazzotto Maldonado Porchia, Mariângela de Oliveira Silva, Lennon Ramos Pereira, Natiely Silva Sales, Guilherme Formoso Pelegrin, Ethiane Segabinazi, Karine Bitencourt Rodrigues, Jamile Ramos da Silva, Bianca da Silva Almeida, Jéssica Pires Farias, Maria Fernanda Castro-Amarante, Paola Marcella Camargo Minoprio, Luís Carlos de Souza Ferreira and Rúbens Prince dos Santos Alves
Viruses 2025, 17(9), 1191; https://doi.org/10.3390/v17091191 - 30 Aug 2025
Viewed by 293
Abstract
Authorized SARS-CoV-2 vaccines elicit both antibody and T-cell responses; however, benchmark correlates and update decisions have largely emphasized neutralizing antibodies. Motivated by the complementary role of cellular immunity, we designed a prototype polyepitope DNA vaccine encoding conserved human and mouse T-cell epitopes from [...] Read more.
Authorized SARS-CoV-2 vaccines elicit both antibody and T-cell responses; however, benchmark correlates and update decisions have largely emphasized neutralizing antibodies. Motivated by the complementary role of cellular immunity, we designed a prototype polyepitope DNA vaccine encoding conserved human and mouse T-cell epitopes from non-structural proteins of the original strain SARS-CoV-2 lineage. Epitope selection was guided by in silico predictions for common HLA class I alleles in the Brazilian population and the mouse H-2Kb haplotype. To enhance immunogenicity, the polyepitope sequences were fused to glycoprotein D (gD) from Herpes Simplex Virus 1 (HSV-1), an immune activator of dendritic cells (DCs), leading to enhanced activation of antigen-specific T-cell responses. Mice were immunized with two doses of the electroporated DNA vaccine encoding the gD-fused polyepitope, which induced robust interferon-gamma– and tumor necrosis factor-alpha–producing T cell responses compared to control mice. In addition, K18-hACE2 transgenic mice showed protection against intranasal challenge with the original SARS-CoV-2 strain, with reduced clinical symptoms, less weight loss, and decreased viral burden in both lung and brain tissues. The results experimentally confirm the protective role of T cells in vaccine-induced protection against SARS-CoV-2 and open perspectives for the development of universal anti-coronavirus vaccines. Full article
(This article belongs to the Special Issue SARS-CoV-2, COVID-19 Pathologies, Long COVID, and Anti-COVID Vaccines)
Show Figures

Graphical abstract

31 pages, 1123 KB  
Review
The Role of Tumor Microenvironment in Triple-Negative Breast Cancer and Its Therapeutic Targeting
by Ana Vuletic, Katarina Mirjacic Martinovic and Vladimir Jurisic
Cells 2025, 14(17), 1353; https://doi.org/10.3390/cells14171353 - 30 Aug 2025
Viewed by 172
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by rapid proliferation and infiltration of immune cells into tumor microenvironment (TME). The treatment of TNBC still remains challenging due to the lack of expression of effective molecular targets pertaining to [...] Read more.
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by rapid proliferation and infiltration of immune cells into tumor microenvironment (TME). The treatment of TNBC still remains challenging due to the lack of expression of effective molecular targets pertaining to the tumor cell itself. In TNBC standard of care, therapies such as chemotherapy, together with recently introduced immunotherapy with checkpoint inhibitors, often do not result in durable clinical response. Therefore, better understanding of complex interactions between tumor cells, immune cells, and stromal cells mediated by multiple cytokines, chemokines, enzymes, and metabolites in TME is crucial for understanding the mechanisms that underlie tumor cell immune evasion strategies. The aim of this review is to give comprehensive overview of immune cell network and their interactions with cells in TME and possibilities for therapeutic targeting of TME in TNBC. We discuss cancer-associated fibroblasts (CAFs) as an important recently characterized player in TNBC with respect to their role in interactions with immune cells and their impact on tumor invasion. Based on the recently accumulated knowledge, therapies targeting immune suppressive mechanisms and CAF-related tumor-promoting mechanisms in TME hold great potential for clinical evaluation in TNBC. Full article
(This article belongs to the Special Issue Tumor Stroma Interaction)
Show Figures

Figure 1

19 pages, 8779 KB  
Article
Bulk and Single-Cell Transcriptomes Reveal Exhausted Signature in Prognosis of Hepatocellular Carcinoma
by Ruixin Chun, Haisen Ni, Ziyi Zhao and Chunlong Zhang
Genes 2025, 16(9), 1034; https://doi.org/10.3390/genes16091034 - 30 Aug 2025
Viewed by 236
Abstract
Background/Objectives: Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with poor prognosis. T cell exhaustion (TEX) is a key factor in tumor immune evasion and therapeutic resistance. In this study, we integrated single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (RNA-seq) data to [...] Read more.
Background/Objectives: Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with poor prognosis. T cell exhaustion (TEX) is a key factor in tumor immune evasion and therapeutic resistance. In this study, we integrated single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (RNA-seq) data to characterize TEX-related transcriptional features in HCC. Methods: We first computed TEX scores for each sample using a curated 65-gene signature and classified them into high-TEX and low-TEX groups by the median score. Differentially expressed genes were identified separately in scRNA-seq and bulk RNA-seq data, then intersected to retain shared candidates. A 26-gene prognostic signature was derived from these candidates via univariate Cox and LASSO regression analysis. Results: The high-TEX group exhibited increased expression of immune checkpoint molecules and antigen presentation molecules, suggesting a tumor microenvironment that is more immunosuppressive but potentially more responsive to immunotherapy. Functional enrichment analysis and protein–protein interaction (PPI) network construction further validated the roles of these genes in immune regulation and tumor progression. Conclusions: This study provides a comprehensive characterization of the TEX landscape in HCC and identifies a robust gene signature associated with prognosis and immune infiltration. These findings highlight the potential of targeting TEX-related genes for personalized immunotherapeutic strategies in HCC. Full article
(This article belongs to the Special Issue AI and Machine Learning in Cancer Genomics)
Show Figures

Figure 1

24 pages, 4005 KB  
Article
Enhancing Antitumor Efficacy of MUC1 mRNA Nano-Vaccine by CTLA-4 siRNA-Mediated Immune Checkpoint Modulation in Triple Negative Breast Cancer Mice Model
by Amir Monfaredan, Sena Şen, Nahideh Karimian Fathi, Didem Taştekin, Alaviyehsadat Hosseininasab, Hamza Uğur Bozbey and Oral Öncül
Int. J. Mol. Sci. 2025, 26(17), 8448; https://doi.org/10.3390/ijms26178448 - 30 Aug 2025
Viewed by 271
Abstract
Immunotherapy, particularly approaches that combine tumor-specific vaccines with immune checkpoint modulation, represents a promising strategy for overcoming tumor immune evasion. While most mRNA-based cancer vaccines focus solely on antigen delivery, there is a need for platforms that simultaneously enhance antigen presentation and modulate [...] Read more.
Immunotherapy, particularly approaches that combine tumor-specific vaccines with immune checkpoint modulation, represents a promising strategy for overcoming tumor immune evasion. While most mRNA-based cancer vaccines focus solely on antigen delivery, there is a need for platforms that simultaneously enhance antigen presentation and modulate the tumor microenvironment to increase therapeutic efficacy. This study presents a novel dual-nanolipid exosome (NLE) platform that simultaneously delivers MUC1 mRNA and CTLA-4-targeted siRNA in a single system. These endogenous lipid-based nanoparticles are structurally designed to mimic exosomes and are modified with mannose to enable selective targeting to dendritic cells (DCs) via mannose receptors. The platform was evaluated both in vitro and in vivo in terms of mRNA encapsulation efficiency, nanoparticle stability, and uptake by DCs. The co-delivery platform significantly enhanced antitumor immune responses compared to monotherapies. Flow cytometry revealed a notable increase in tumor-infiltrating CD8+ T cells (p < 0.01), and ELISPOT assays showed elevated IFN-γ production upon MUC1-specific stimulation. In vivo CTL assays demonstrated enhanced MUC1-specific cytotoxicity. Combined therapy resulted in immune response enhancement compared to vaccine or CTLA-4 siRNA alone. The NLE platform exhibited favorable biodistribution and low systemic toxicity. By combining targeted delivery of dendritic cells, immune checkpoint gene silencing, and efficient antigen expression in a biomimetic nanoparticle system, this study represents a significant advance over current immunotherapy strategies. The NLE platform shows strong potential as a modular and safe approach for RNA-based cancer immunotherapy. Full article
(This article belongs to the Special Issue Biopolymers for Enhanced Health Benefits—2nd Edition)
Show Figures

Figure 1

21 pages, 2295 KB  
Article
Discovery of a Promising Hydroxyamino-Piperidine HDAC6 Inhibitor via Integrated Virtual Screening and Experimental Validation in Multiple Myeloma
by Federica Chiera, Antonio Curcio, Roberta Rocca, Ilenia Valentino, Massimo Gentile, Stefano Alcaro, Nicola Amodio and Anna Artese
Pharmaceuticals 2025, 18(9), 1303; https://doi.org/10.3390/ph18091303 - 29 Aug 2025
Viewed by 233
Abstract
Background: Histone deacetylase 6 (HDAC6) is a unique class IIb HDAC isozyme characterized by two catalytic domains and a zinc finger ubiquitin-binding domain. It plays critical roles in various cellular processes, including protein degradation, autophagy, immune regulation, and cytoskeletal dynamics. Due to its [...] Read more.
Background: Histone deacetylase 6 (HDAC6) is a unique class IIb HDAC isozyme characterized by two catalytic domains and a zinc finger ubiquitin-binding domain. It plays critical roles in various cellular processes, including protein degradation, autophagy, immune regulation, and cytoskeletal dynamics. Due to its multifunctional nature and overexpression in several cancer types, HDAC6 has emerged as a promising therapeutic target. Methods: In this study, we employed a ligand-based pharmacophore modeling approach using a structurally diverse set of known HDAC6 inhibitors. This was followed by the virtual screening of over 140,000 commercially available compounds from both the MolPort and Asinex databases. The screening workflow incorporated pharmacophore filtering, molecular docking, and molecular dynamic (MD) simulations. Binding free energies were estimated using Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analysis to prioritize top candidates. A fluorometric enzymatic assay was used to measure HDAC6 activity, while cell viability assay by Cell Titer Glo was used to assess the anti-tumor activity against drug-sensitive and -resistant multiple myeloma (MM) cells. Western blotting was used to evaluate the acetylation of tubulin or histone H4 after treatment with selected compounds. Results: Three promising compounds were identified based on stable binding conformations and favorable interactions within the HDAC6 catalytic pocket. Among them, Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analysis identified Compound 10 (AKOS030273637) as the top theoretical binder, with a ΔGbind value of −45.41 kcal/mol. In vitro enzymatic assays confirmed its binding to the HDAC6 catalytic domain and inhibitory activity. Functional studies on MM cell lines, including drug-resistant variants, showed that Compound 10 reduced cell viability. Increased acetylation of α-tubulin, a substrate of HDAC6, likely suggested on-target mechanism of action. Conclusions: Compound 10, featuring a benzyl 4-[4-(hydroxyamino)-4-oxobutylidene] piperidine-1-carboxylate scaffold, demonstrates potential drug-like properties and a predicted bidentate zinc ion coordination, supporting its potential as an HDAC6 inhibitor for further development in hematologic malignancies. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

16 pages, 2348 KB  
Article
Novel Application of Ion Mobility Mass Spectrometry Reveals Complex Ganglioside Landscape in Diffuse Astrocytoma Peritumoral Regions
by Raluca Ica, Mirela Sarbu, Roxana Biricioiu, Dragana Fabris, Željka Vukelić and Alina D. Zamfir
Int. J. Mol. Sci. 2025, 26(17), 8433; https://doi.org/10.3390/ijms26178433 - 29 Aug 2025
Viewed by 226
Abstract
Diffuse astrocytoma is a primary brain tumor known for its gradual and diffuse infiltration into the surrounding brain tissue. Given this characteristic, the investigation of the peritumoral region holds potential biological and clinical relevance. In this study, ion mobility spectrometry mass spectrometry (IMS [...] Read more.
Diffuse astrocytoma is a primary brain tumor known for its gradual and diffuse infiltration into the surrounding brain tissue. Given this characteristic, the investigation of the peritumoral region holds potential biological and clinical relevance. In this study, ion mobility spectrometry mass spectrometry (IMS MS) was optimized and applied for the first time for the analysis of gangliosides present in the peritumoral tissue of diffuse astrocytoma. Ganglioside profiling and structural characterization were conducted using high-resolution nanoelectrospray ionization (nanoESI) IMS MS, along with tandem mass spectrometry (MS/MS) via low-energy collision-induced dissociation (CID) in the negative ion mode. Using IMS MS-based separation and screening, we observed a greater diversity of ganglioside species in the peritumoral tissue than previously reported. Notably, an elevated expression was detected for several species, including GT1(d18:1/18:0), GT1(d18:1/20:0), GM2(d18:1/16:2), GD1(d18:1/16:0), GD2(d18:1/20:0), Fuc-GT3(d18:1/24:4), and Fuc-GD1(d18:1/18:2). Although preliminary, these observations prompt consideration of whether these species could be implicated in processes such as microenvironmental modulation, tumor cell infiltration and invasion, maintenance of cellular interactions, or regulation of immune responses. Additionally, their potential utility as biomarkers may merit further exploration. In the subsequent phase of the study, structural analysis using IMS MS, CID tandem MS, and fragmentation data supported the identification of GT1b(d18:1/20:0) isomer in the peritumoral tissue. However, given the exploratory nature of the study and reliance on limited sampling, further investigation across broader sample sets is necessary to extend these findings. Full article
(This article belongs to the Special Issue Invasion and Metastasis in Brain Cancer)
Show Figures

Figure 1

Back to TopTop