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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,315)

Search Parameters:
Keywords = T and NK cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1575 KB  
Review
The Role of IL28B Polymorphism in Regulating Innate and Adaptive Immunity Against Viral Infection Among Allogenic Stem Cells Transplant Recipients
by Mohamed A. Eltokhy, Bhaumik Patel, Marina Curcic, Faizah Alabi, Shadan Modaresahmadi, Omar Eltoukhy, Esraa G. Abdelmageed and Sahar Radwan
Immuno 2025, 5(3), 38; https://doi.org/10.3390/immuno5030038 - 3 Sep 2025
Abstract
Viral infection is a significant cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (Allo-HSCT), largely due to its impact on and interaction with immune reconstitution. Both innate and adaptive immunity are essential for effective viral control, yet their recovery post-transplant [...] Read more.
Viral infection is a significant cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (Allo-HSCT), largely due to its impact on and interaction with immune reconstitution. Both innate and adaptive immunity are essential for effective viral control, yet their recovery post-transplant is often delayed or functionally impaired. Emerging evidence suggests genetic variation, particularly polymorphisms in the IL28B gene (encoding IFN-λ3), as a critical factor influencing the quality and timing of immune responses during the early post-transplant period. This review explores the role of IL28B polymorphisms in shaping antiviral immunity, in general, as well as after Allo-HSCT. IL28B variants have been implicated in modulating interferon-stimulated gene (ISG) expression, natural killer (NK) cell activity, and type I/III interferon signaling, all central components of innate immune defense against viral infections. Furthermore, IL28B polymorphisms, particularly rs12979860, have been shown in both general populations and limited HSCT cohorts to alter T cell response and interferon production, affecting reactivation and clearance of multiple viruses such as cytomegalovirus (CMV), hepatitis B virus (HBV), hepatitis C virus (HCV), Epstein–Barr virus (EBV), COVID-19, and BK polyomavirus (BKPyV) as well as Graft vs. Host disease, thereby affecting adaptive immune reconstitution and long-term viral control. Understanding how IL28B genotype alters immune dynamics in transplant recipients could enhance risk stratification for CMV and other diseases and inform personalized prophylactic or therapeutic strategies. Therefore, this review highlights IL28B as a promising biomarker and potential immunoregulatory target in the management of viral infection post-Allo-HSCT. Full article
Show Figures

Figure 1

15 pages, 2570 KB  
Article
Antibody-Dependent Cellular Cytotoxicity Elicited by the Antibodies Against the E120R Protein of African Swine Fever Virus
by Shengmei Chen, Jing Lan, Zhanhao Lu, Jia Li, Caoyuan Ma, Rui Luo, Qiang Fu, Yuan Sun, Tao Wang and Hua-Ji Qiu
Vaccines 2025, 13(9), 934; https://doi.org/10.3390/vaccines13090934 - 1 Sep 2025
Viewed by 142
Abstract
Background/Objectives: African swine fever (ASF) is a disease of domestic pigs and wild boar caused by African swine fever virus (ASFV), in which infection often leads to high morbidity and mortality. Although subunit and mRNA vaccines based on protective antigens have been explored [...] Read more.
Background/Objectives: African swine fever (ASF) is a disease of domestic pigs and wild boar caused by African swine fever virus (ASFV), in which infection often leads to high morbidity and mortality. Although subunit and mRNA vaccines based on protective antigens have been explored for ASFV, their protective efficacy remains insufficient for practical ASF control, highlighting the need to identify new potential antigens capable of inducing more potent and broadly protective immune responses. Previously, we found that the antibodies against the ASFV E120R protein (pE120R) could significantly inhibit virus replication in primary porcine alveolar macrophages (PAMs). However, it is not yet known whether anti-pE120R antibodies can induce antibody-dependent cellular cytotoxicity (ADCC). Methods: In this study, we analyzed the conservation and immunogenic features of pE120R and established an HEK293T cell line with stable expression of pE120R as target cells (HEK293T-pE120R). Additionally, a co-culture system comprising target cells and peripheral blood mononuclear cells (PBMCs) was established to evaluate the ability of the anti-pE120R antibodies to induce ADCC as measured by lactate dehydrogenase (LDH) release assays. Results: The results showed that pE120R is highly conserved among different ASFV genotypes and contains multiple B-cell and T-cell epitopes. Importantly, LDH release assays demonstrated that anti-pE120R antibodies triggered NK cell-mediated ADCC. Notably, ASFV replication in HEK293T-pE120R cells was not promoted. Conclusions: In summary, pE120R was associated with antibody production in a cytotoxicity assay. The ability of this antigen to induce protective immunity, if any, requires further evaluation in vivo. Full article
(This article belongs to the Special Issue Swine Vaccines and Vaccination)
Show Figures

Figure 1

21 pages, 4640 KB  
Article
Postpartum Uterine Involution in Cows: Quantitative Assessment of Structural Remodeling and Immune Cell Infiltration
by Karine V. Aires, Ana Paula da Silva, Leonardo G. de Andrade, Alexandre Boyer, Gustavo Zamberlam, Valerio M. Portela, Alfredo Q. Antoniazzi and Guillaume St-Jean
Animals 2025, 15(17), 2520; https://doi.org/10.3390/ani15172520 - 27 Aug 2025
Viewed by 290
Abstract
Postpartum uterine involution in cattle involves complex morphological and immunological changes essential for restoring uterine health and fertility. This study evaluated endometrial biopsies collected at four postpartum time points to characterize tissue remodeling and immune cell dynamics during involution. Histology revealed intact luminal [...] Read more.
Postpartum uterine involution in cattle involves complex morphological and immunological changes essential for restoring uterine health and fertility. This study evaluated endometrial biopsies collected at four postpartum time points to characterize tissue remodeling and immune cell dynamics during involution. Histology revealed intact luminal columnar epithelium in 92.98% of samples, with stable stromal architecture. Stromal edema decreased by Day 7 but increased again by Day 35, while endometrial gland numbers significantly rose at Day 35, suggesting glandular recovery linked to resumed cyclicity. Subepithelial collagen deposition peaked on Day 21, indicating active extracellular matrix remodeling. Immunologically, early postpartum was marked by increased PMNs and macrophages, whereas Day 21 showed peak infiltration of natural killer (NK) cells and T and B lymphocytes, sometimes forming lymphoid aggregates. Manual and automated immune cell quantifications correlated well. These findings demonstrate a dynamic shift from acute neutrophil-dominated inflammation to a lymphocyte-rich environment during uterine involution. This immune modulation may contribute to the earlier diagnosis of subclinical endometritis, typically identified at later stages of postpartum period. Overall, this study provides insight into the temporal immunomorphological events supporting uterine recovery, with potential implications for reproductive management in dairy cattle. Full article
(This article belongs to the Special Issue Uterine Homeostasis and Disease in Dairy Cows)
Show Figures

Figure 1

30 pages, 5591 KB  
Article
Expanding the Knowledge of the Molecular Effects and Therapeutic Potential of Incomptine A for the Treatment of Non-Hodgkin Lymphoma: In Vivo and Bioinformatics Studies, Part III
by Normand García-Hernández, Fernando Calzada, Elihú Bautista, José Manuel Sánchez-López, Miguel Valdes, Claudia Velázquez and Elizabeth Barbosa
Pharmaceuticals 2025, 18(9), 1263; https://doi.org/10.3390/ph18091263 - 25 Aug 2025
Viewed by 416
Abstract
Background/Objectives: Non-Hodgkin lymphoma (NHL) is a group of blood cancers that arise in the lymphatic nodes and other tissues after an injury to the DNA of B/T lineage and NK lymphocytes. Recently, we reported that incomptine A (IA) has in [...] Read more.
Background/Objectives: Non-Hodgkin lymphoma (NHL) is a group of blood cancers that arise in the lymphatic nodes and other tissues after an injury to the DNA of B/T lineage and NK lymphocytes. Recently, we reported that incomptine A (IA) has in vivo antilymphoma properties. This research aimed to evaluate the effects of IA in the treatment of NHL using antilymphoma activity, Tandem Mass Tag (TMT), and bioinformatics approaches. Methods: The antilymphoma activity of IA was tested on male Balb/c mice inoculated with U-937 cells. Also, TMT, gene ontology enrichment, Reactome pathway, Kyoto Encyclopedia of Gene and Genomes pathway, molecular docking, toxicoinformatic, and pharmaceutical analyses were performed. Results: By TMT analysis of the altered levels of proteins present in the lymph nodes of Balb/c mice with NHL and treated with IA, we identified 106 significantly differentially expressed proteins (DEPs), including Il1rap, Ifi44, Timd4, Apoa4, and Fabp3 as well as Myh3, Eno 2, and H4c11. Among these, the Fhl1 result was the most important cluster altered and a potential core target of IA for the treatment of NHL. Network pharmacology studies have revealed that DEPs are associated with processes such as muscle contraction, glycolysis, hemostasis, epigenetic regulation of gene expression, transport of small molecules, neutrophil extracellular trap formation, adrenergic signaling in cardiomyocytes, systemic lupus erythematosus, alcoholism, and platelet activation, signaling, and aggregation. Computational studies revealed strong binding affinities with six proteins associated with cancer, positive pharmacokinetic properties, and no toxicity. Conclusions: Our contribution suggests that IA may be a compound with potential therapeutic effects against NHL. Full article
Show Figures

Figure 1

17 pages, 2429 KB  
Article
BCG Vaccine-Induced Innate and Adaptive Pulmonary Immunity Correlating with Protective Efficacy Against Mycobacterium tuberculosis in the Lungs
by Mayank Khanna and Alistair J. Ramsay
Vaccines 2025, 13(8), 876; https://doi.org/10.3390/vaccines13080876 - 19 Aug 2025
Viewed by 504
Abstract
Background/Objectives: Effective prophylaxis for Mycobacterium tuberculosis (Mtb) requires greater understanding of immune correlates of protection. With renewed interest in BCG as an Mtb vaccine, particularly via the intravenous (IV) route, our objective was to characterize both innate and adaptive immune correlates of vaccine-induced [...] Read more.
Background/Objectives: Effective prophylaxis for Mycobacterium tuberculosis (Mtb) requires greater understanding of immune correlates of protection. With renewed interest in BCG as an Mtb vaccine, particularly via the intravenous (IV) route, our objective was to characterize both innate and adaptive immune correlates of vaccine-induced pulmonary immunity as potential biomarkers for protective efficacy in a murine model of Mtb infection. Methods: Mice were given BCG via different routes and some boosted with recombinant virus constructs encoding Mtb Ag85B. Responding innate lymphoid cell (ILC) populations, T cells and B cells were analyzed by fluorescence activated cell sorting (FACS) for surface markers and by intracellular cytokine staining or antibody ELISPOT. Some immunized mice were challenged with aerosolized Mtb and monitored for bacterial growth in the lungs and spleen. Results: BCG given IV, but not intranasally or subcutaneously, resulted in marked increases in IFNγ expression at 72 h by pulmonary CD49+ NK cells, CD69+ ILC1, and two ILC3 populations, NCR-ILC3 and LTi cells, the latter also producing IL-22. Pulmonary ILC2 populations in these mice had significantly increased IL-13 expression at 24 h compared to the other routes. Interestingly, high levels of NK cells and ILC1 expressing IFNγ and/or TNFα were sustained at 8 wk, with sustained expression of IL-17A by pulmonary NCR-ILC3 and pronounced tissue-resident and effector memory CD4+ and CD8+ T cell responses. Intranasal boosting with Ad-Ag85B enhanced these T cell responses and generated Mtb-specific pulmonary IgA and IgG B cells, correlating with significantly reduced bacterial loads following Mtb challenge. Conclusions: BCG given IV primed for both early and persistent pulmonary ILC1/ILC3 responses of a predominantly Th1/Th17-type profile along with local Mtb-specific memory T cell and B cell populations, correlating with enhanced protective efficacy. These are worthy of further study as compartmentalized biomarkers for effective vaccine-induced local immunity against Mtb. Full article
Show Figures

Figure 1

29 pages, 1164 KB  
Review
Induced Pluripotent Stem Cell-Based Cancer Immunotherapy: Strategies and Perspectives
by Xiaodong Xun, Jialing Hao, Qian Cheng and Pengji Gao
Biomedicines 2025, 13(8), 2012; https://doi.org/10.3390/biomedicines13082012 - 19 Aug 2025
Viewed by 649
Abstract
Cellular immunotherapy has emerged as a transformative approach in oncology, revolutionizing cancer treatment paradigms. Since the groundbreaking development of induced pluripotent stem cells (iPSCs) by Yamanaka in 2008, significant progress has been made in generating various iPSCs-derived immunocytes, including T cells, dendritic cells, [...] Read more.
Cellular immunotherapy has emerged as a transformative approach in oncology, revolutionizing cancer treatment paradigms. Since the groundbreaking development of induced pluripotent stem cells (iPSCs) by Yamanaka in 2008, significant progress has been made in generating various iPSCs-derived immunocytes, including T cells, dendritic cells, macrophages, natural killer (NK) cells, and B cells. These engineered immune cells offer unprecedented opportunities for personalized cancer therapy as they can be derived from patients’ own cells to minimize immune rejection. In addition, various new techniques are being used for the induction and amplification of iPSCs-derived immunocytes, such as small-molecule techniques, 3D culture systems, nanotechnology, and animal models for the in vivo amplification of immunocytes. Of course, challenges remain in improving immunocyte characteristics. Targeting efficiency needs enhancement to better distinguish tumor cells from healthy tissue, while biological activity must be optimized for sustained antitumor effects. Safety concerns, particularly regarding potential off-target effects and cytokine release syndrome, require further investigation. The immunosuppressive nature of tumor microenvironment also poses significant hurdles for solid tumor treatment. Ongoing clinical trials are exploring the therapeutic potential of iPSCs-derived immunocytes, with researchers investigating combination therapies and genetic modifications to overcome current limitations. Full article
Show Figures

Figure 1

21 pages, 4323 KB  
Article
Inhibition of the Transcription Factor PU.1 Suppresses Tumor Growth in Mice by Promoting the Recruitment of Cytotoxic Lymphocytes Through the CXCL9-CXCR3 Axis
by Nichita Sleapnicov, Soon-Duck Ha, Shanshan Jenny Zhong, Jackie Duchscher, Sally Ezra, Shawn Shun-Cheng Li and Sung Ouk Kim
Cancers 2025, 17(16), 2684; https://doi.org/10.3390/cancers17162684 - 18 Aug 2025
Viewed by 488
Abstract
Background: Targeting tumor-associated macrophages (TAMs) is a promising immunotherapy for cancers, but current strategies are limited due to strategic caveats. PU.1 is a transcription factor required for macrophage generation and differentiation. To date, the effect of PU.1 inhibition on solid tumors is [...] Read more.
Background: Targeting tumor-associated macrophages (TAMs) is a promising immunotherapy for cancers, but current strategies are limited due to strategic caveats. PU.1 is a transcription factor required for macrophage generation and differentiation. To date, the effect of PU.1 inhibition on solid tumors is unknown. Methods: This study examines the anti-tumor effect of PU.1 inhibition and its mechanism using the small-molecule DB2313 in mouse melanoma and breast tumor models. Results: We found that inhibition of PU.1 by DB2313 suppresses B16-OVA melanoma and 4T1 breast tumor growth in mice. In the melanoma tumor model, DB2313 enhanced tumor recruitment of CD4+ T helper 1 (Th1) and cytotoxic T/natural killer (NK) cells by targeting TAMs. Transcriptome and targeted gene expression analyses revealed that PU.1 inhibition by DB2313 and small-interference RNAs enhances CXCL9 expression in bulk tumors, TAMs, and bone marrow-derived macrophages. The anti-tumor effects of DB2313 were abolished by depleting macrophages with clodronate or inhibiting the CXCL9-CXCR3 chemokine axis using CXCL9- or CXCR3-neutralizing antibodies. Conclusions: These results suggest that pharmacological inhibition of PU.1 suppresses tumor growth by at least promoting the infiltration of lymphocytes into tumors through the CXCL9-CXCR3 chemokine axis. Our study establishes a framework for developing TAM-modulating immunotherapies by targeting the transcriptional factor PU.1. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

10 pages, 518 KB  
Article
A Novel Study of β1- and β2-Adrenergic Receptors Present on PBMCs, T Cells, Monocytes, and NK Cells by Radioligand Method: Quantitation and Correlations
by Mihail. M. Peklo, Ekaterina V. Smolyakova, Lyudmila N. Lipatova, Natal’ya M. Kashirina, Yurij S. Skoblov, Natal’ya A. Skoblova, Mihail A. Slinkin, Igor’ N. Rybalkin, Pavel N. Rutkevich, Olga K. Chusovitina, Elena V. Yanushevskaya, Kirill A. Zykov and Tat’yana N. Vlasik
Int. J. Mol. Sci. 2025, 26(16), 7894; https://doi.org/10.3390/ijms26167894 - 15 Aug 2025
Viewed by 305
Abstract
β-adrenoreceptor (ADRB) ligands are actively used in the therapy of bronchopulmonary and cardiovascular diseases. When using these drugs, it is important to assess changes in ADRB content in different tissues. In most cases, the direct measurement of ADRB content in lung and heart [...] Read more.
β-adrenoreceptor (ADRB) ligands are actively used in the therapy of bronchopulmonary and cardiovascular diseases. When using these drugs, it is important to assess changes in ADRB content in different tissues. In most cases, the direct measurement of ADRB content in lung and heart cells is not possible. ADRB2 content in peripheral blood lymphocytes (or mononuclear cells) was shown to correlate with that in myocardial cells. It has been suggested that blood lymphocytes can be used to monitor ADRB content in solid organs. However, the estimation of ADRB1 content in myocardium from its content in peripheral lymphocytes is not possible due to the low content of ADRB1 in lymphocytes. In the present study, we performed simultaneous determination of ADRB1 and ADRB2 both in the total population of PBMCs and in isolated subpopulations of monocytes, T-lymphocytes, and NK-cells from 23 healthy donors using the modified radioligand method. The highest amount of ADRB2 was detected in NK cells, followed by PBMCs, monocytes, and T cells. The content of these receptors in all blood cell subpopulations was significantly correlated with each other, suggesting the possibility of using PBMCs to monitor ADRB2 in solid organs. For the first time, ADRB1 was detected in monocytes and NK cells. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

23 pages, 8237 KB  
Review
CAR Cell-Derived Exosomes in Cancer Therapy: Biogenesis, Engineering Strategies and Antitumor Mechanisms
by Chaohua Si, Yuanyuan Li, Yunwen Wang, Jianen Gao and Xu Ma
Int. J. Mol. Sci. 2025, 26(16), 7890; https://doi.org/10.3390/ijms26167890 - 15 Aug 2025
Viewed by 764
Abstract
Chimeric antigen receptor (CAR) cell therapy, encompassing CAR T, CAR NK, and CAR macrophage cells, demonstrates high efficacy in tumor treatment, conferring durable and effective responses, notably in hematologic malignancies. However, challenges persist in the manufacture of CAR cells, and treatment is associated [...] Read more.
Chimeric antigen receptor (CAR) cell therapy, encompassing CAR T, CAR NK, and CAR macrophage cells, demonstrates high efficacy in tumor treatment, conferring durable and effective responses, notably in hematologic malignancies. However, challenges persist in the manufacture of CAR cells, and treatment is associated with serious adverse events, notably cytokine release syndrome (CRS), a potentially life-threatening complication. Owing to the inherent properties of exosomes, CAR cell-derived exosomes offer distinct advantages in cancer therapeutics. CAR cells-derived exosomes retain the inherent tumor-killing function of the parent cells while also exhibiting key practical advantages, including wide availability, safety, and ease of storage and transport. Furthermore, CAR cell-derived exosomes can be combined with other tumor therapies; this combinatorial approach significantly enhances efficacy while reducing side effects. To accelerate the clinical translation of CAR cell-derived exosomes in tumor therapy, this paper reviews their biogenesis, engineering strategies, antitumor mechanisms and clinical evidence, including case studies of combination therapies with other antitumor modalities. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

45 pages, 861 KB  
Review
Cytokine Networks in Triple-Negative Breast Cancer: Mechanisms, Therapeutic Targets, and Emerging Strategies
by María Rosado-Sanz, Nuria Martínez-Alarcón, Adrián Abellán-Soriano, Raúl Golfe, Eva M. Trinidad and Jaime Font de Mora
Biomedicines 2025, 13(8), 1945; https://doi.org/10.3390/biomedicines13081945 - 8 Aug 2025
Viewed by 717
Abstract
Triple-negative breast cancer (TNBC) remains a challenging subtype of breast cancer due to its aggressive nature and lack of targeted therapies. Cytokines play a pivotal role in shaping the tumor microenvironment, modulating tumor progression, immune evasion, and therapy resistance. In this review, we [...] Read more.
Triple-negative breast cancer (TNBC) remains a challenging subtype of breast cancer due to its aggressive nature and lack of targeted therapies. Cytokines play a pivotal role in shaping the tumor microenvironment, modulating tumor progression, immune evasion, and therapy resistance. In this review, we discuss the complex cytokine networks involved in TNBC biology, highlighting their contribution to key oncogenic processes, including proliferation, angiogenesis, epithelial–mesenchymal transition, and immunomodulation. We also summarize current and emerging cytokine-targeted therapeutic strategies, including monoclonal antibodies, bispecific antibodies, cell-based therapies, and cytokine-armed CAR-T and CAR-NK cell approaches, with a focus on clinical implications and future directions. Full article
Show Figures

Figure 1

16 pages, 2761 KB  
Article
Persistent Type I Interferon Signaling Impairs Innate Lymphoid Cells During HIV-1 Infection Under Suppressive ART
by Runpeng Han, Haisheng Yu, Guangming Li, Lishan Su and Liang Cheng
Viruses 2025, 17(8), 1099; https://doi.org/10.3390/v17081099 - 8 Aug 2025
Viewed by 818
Abstract
Persistent type I interferon (IFN-I) signaling compromises adaptive anti-HIV-1 T cell immunity and promotes viral reservoir persistence, yet its effects on innate lymphoid cells during chronic infection remain unclear. Through integrated single-cell RNA sequencing and functional validation in HIV-1-infected humanized mice with combination [...] Read more.
Persistent type I interferon (IFN-I) signaling compromises adaptive anti-HIV-1 T cell immunity and promotes viral reservoir persistence, yet its effects on innate lymphoid cells during chronic infection remain unclear. Through integrated single-cell RNA sequencing and functional validation in HIV-1-infected humanized mice with combination antiretroviral therapy (cART) and IFN-I signaling blockade, we reveal IFN-I-induced dysfunction of natural killer (NK) cells and group 3 innate lymphoid cells (ILC3s). Mechanistically, the IFN-I-CD9 axis drives NK cells toward a decidual NK cell-like phenotype, impairing their cytotoxic activity. Furthermore, IFNAR blockade rescues ILC3 functionality, which is critical for IL-17/IL-22-mediated antimicrobial defense and mucosal barrier maintenance. Our study delineates IFN-I-driven immunosuppression across innate lymphocyte compartments and proposes the targeted modulation of this pathway to enhance antiviral and mucosal immunity in HIV-1 management. Full article
(This article belongs to the Special Issue Interferon Signaling in Viral Pathogenesis)
Show Figures

Figure 1

28 pages, 13042 KB  
Article
Anti-Her2 CAR-NK92 Cells and Their Exosomes: Generation, Characterization, and Selective Cytotoxicity Against Her2-Positive Tumor Cells
by Alexandru Tîrziu, Florina Maria Bojin, Oana Isabella Gavriliuc, Roxana Maria Buzan, Lauriana Eunice Zbîrcea, Manuela Grijincu and Virgil Păunescu
Int. J. Mol. Sci. 2025, 26(15), 7648; https://doi.org/10.3390/ijms26157648 - 7 Aug 2025
Viewed by 558
Abstract
Chimeric antigen receptor (CAR)-engineered NK cells are a promising approach for targeted immunotherapy in Her2-positive cancers. This study aimed to generate anti-Her2 CAR-NK92 cells, to evaluate their selective cytotoxicity against Her2-positive cancer cells, and to isolate and characterize their released exosomes. NK92 cells [...] Read more.
Chimeric antigen receptor (CAR)-engineered NK cells are a promising approach for targeted immunotherapy in Her2-positive cancers. This study aimed to generate anti-Her2 CAR-NK92 cells, to evaluate their selective cytotoxicity against Her2-positive cancer cells, and to isolate and characterize their released exosomes. NK92 cells were electroporated with piggyBac transposon vectors encoding anti-Her2 CAR and the helper transposase. Puromycin selection was performed to enrich the transduced cells. CAR and GFP expression were assessed by flow cytometry, and exosomes were isolated and characterized in terms of protein cargo and surface protein expression. Cytotoxicity was evaluated using real-time cell analysis against Her2-positive SK-BR3 cells and Her2-negative MCF-7 cells. Electroporation did not significantly affect NK92 cell viability. Puromycin selection efficiently enriched for CAR-expressing cells, with GFP positivity reaching 99.8% and a 15-fold increase in CAR surface expression compared to wild-type cells. CAR-NK92 cells demonstrated robust, Her2-specific cytotoxicity in a E:T-dependent manner, with the greatest effect observed at a 10:1 effector-to-target ratio. Exosomes derived from CAR-NK92 cells contained CAR molecules and selectively targeted Her2-positive cells. Anti-Her2 CAR-NK92 cells and their exosomes exhibit potent and selective cytotoxicity against Her2-positive cancer cells, supporting their potential as innovative immunotherapeutic agents for solid tumors. Full article
(This article belongs to the Special Issue Chimeric Antigen Receptors Against Cancers and Autoimmune Diseases)
Show Figures

Figure 1

21 pages, 1727 KB  
Review
Immune Evasion in Head and Neck Squamous Cell Carcinoma: Roles of Cancer-Associated Fibroblasts, Immune Checkpoints, and TP53 Mutations in the Tumor Microenvironment
by Chung-Che Tsai, Yi-Chiung Hsu, Tin-Yi Chu, Po-Chih Hsu and Chan-Yen Kuo
Cancers 2025, 17(15), 2590; https://doi.org/10.3390/cancers17152590 - 7 Aug 2025
Viewed by 809
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and extracellular matrix elements, that collectively modulate tumor growth, metastasis, and resistance to therapy. Immune evasion in HNSCC is orchestrated through multiple mechanisms, including the suppression of cytotoxic T lymphocytes, recruitment of immunosuppressive cells, such as regulatory T and myeloid-derived suppressor cells, and upregulation of immune checkpoint molecules (e.g., PD-1/PD-L1 and CTLA-4). Natural killer (NK) cells, which play a crucial role in anti-tumor immunity, are often dysfunctional within the HNSCC TME due to inhibitory signaling and metabolic constraints. Additionally, endothelial cells contribute to tumor angiogenesis and immune suppression, further exacerbating disease progression. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors and NK cell-based strategies, have shown promise in restoring anti-tumor immunity. Moreover, TP53 mutations, frequently observed in HNSCC, influence tumor behavior and therapeutic responses, highlighting the need for personalized treatment approaches. This review provides a comprehensive analysis of the molecular and cellular mechanisms governing immune evasion in HNSCC with a focus on novel therapeutic strategies aimed at improving patient outcomes. Full article
(This article belongs to the Special Issue Oral Cancer: Prevention and Early Detection (2nd Edition))
Show Figures

Figure 1

8 pages, 2701 KB  
Case Report
A Drop of Blood to Lead the Way
by Theodora A. M. Claushuis, Marielle J. Wondergem, Henriette B. Beverloo, Marise R. Heerma van Voss, Remco J. Molenaar, Maud Zwolsman, Fleur M. van der Valk, Hans L. Mooij, Lianne Koens and Sanne H. Tonino
Hematol. Rep. 2025, 17(4), 40; https://doi.org/10.3390/hematolrep17040040 - 5 Aug 2025
Viewed by 311
Abstract
Background and Significances: In patients with Epstein–Barr virus-driven hemophagocytic lymphohistiocytosis (EBV-HLH), identifying the underlying cause poses a significant diagnostic challenge. HLH may precede overt disease, and early directed treatment for HLH can obscure histopathological findings. A liquid biopsy enables the detection of tumor-derived [...] Read more.
Background and Significances: In patients with Epstein–Barr virus-driven hemophagocytic lymphohistiocytosis (EBV-HLH), identifying the underlying cause poses a significant diagnostic challenge. HLH may precede overt disease, and early directed treatment for HLH can obscure histopathological findings. A liquid biopsy enables the detection of tumor-derived DNA from various sources, including cell-free DNA, circulating tumor cells, extracellular vesicles, and tumor-educated platelets, and might aid in this setting. Case Presentation: This case presents a young patient with EBV-HLH, in which genomic analysis of tumor-derived DNA from circulating tumor cells led to the diagnosis of an EBV-positive NK/T-cell lymphoma—where conventional tissue biopsies had failed. Conclusions: This report underscores the potential of the liquid biopsy as a valuable diagnostic tool in complex cases of EBV-HLH. Full article
Show Figures

Figure 1

31 pages, 3657 KB  
Review
Lipid Metabolism Reprogramming in Cancer: Insights into Tumor Cells and Immune Cells Within the Tumor Microenvironment
by Rundong Liu, Chendong Wang, Zhen Tao and Guangyuan Hu
Biomedicines 2025, 13(8), 1895; https://doi.org/10.3390/biomedicines13081895 - 4 Aug 2025
Viewed by 1058
Abstract
This review delves into the characteristics of lipid metabolism reprogramming in cancer cells and immune cells within the tumor microenvironment (TME), discussing its role in tumorigenesis and development and analyzing the value of lipid metabolism-related molecules in tumor diagnosis and prognosis. Cancer cells [...] Read more.
This review delves into the characteristics of lipid metabolism reprogramming in cancer cells and immune cells within the tumor microenvironment (TME), discussing its role in tumorigenesis and development and analyzing the value of lipid metabolism-related molecules in tumor diagnosis and prognosis. Cancer cells support their rapid growth through aerobic glycolysis and lipid metabolism reprogramming. Lipid metabolism plays distinct roles in cancer and immune cells, including energy supply, cell proliferation, angiogenesis, immune suppression, and tumor metastasis. This review focused on shared lipid metabolic enzymes and transporters, lipid metabolism-related oncogenes and non-coding RNAs (ncRNAs) involved in cancer cells, and the influence of lipid metabolism on T cells, dendritic cells (DCs), B cells, tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), and natural killer cells (NKs) within TME. Additionally, the role of lipid metabolism in tumor diagnosis and prognosis was explored, and lipid metabolism-based anti-tumor treatment strategies were summarized, aiming to provide new perspectives for achieving precision medicine. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment: Third Edition)
Show Figures

Graphical abstract

Back to TopTop