Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 911
Filter
1.
Cell Death Dis ; 15(6): 428, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890285

ABSTRACT

Neuroblastoma (NB) is a highly aggressive pediatric cancer that originates from immature nerve cells, presenting significant treatment challenges due to therapy resistance. Despite intensive treatment, approximately 50% of high-risk NB cases exhibit therapy resistance or experience relapse, resulting in poor outcomes often associated with tumor immune evasion. B7-H3 is an immune checkpoint protein known to inhibit immune responses. MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation. Our study aims to explore the impact of miRNAs on B7-H3 regulation, the anti-tumor immune response, and tumorigenicity in NB. Analysis of NB patients and patient-derived xenograft tumors revealed a correlation between higher B7-H3 expression and poorer patient survival. Notably, deceased patients exhibited a depletion of miR-29 family members (miR-29a, miR-29b, and miR-29c), which displayed an inverse association with B7-H3 expression in NB patients. Overexpression and knockdown experiments demonstrated that these miRNAs degrade B7-H3 mRNA, resulting in enhanced NK cell activation and cytotoxicity. In vivo, experiments provided further evidence that miR-29 family members reduce tumorigenicity, macrophage infiltration, and microvessel density, promote infiltration and activation of NK cells, and induce tumor cell apoptosis. These findings offer a rationale for developing more effective combination treatments that leverage miRNAs to target B7-H3 in NB patients.


Subject(s)
B7 Antigens , Killer Cells, Natural , MicroRNAs , Neuroblastoma , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , B7 Antigens/metabolism , B7 Antigens/genetics , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice, Nude , Female , Male , Lymphocyte Activation
2.
Diagn Pathol ; 19(1): 77, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858715

ABSTRACT

Although most differentiated thyroid carcinoma has a clinically favorable prognosis, some of specific types of thyroid cancer (such as anaplastic thyroid carcinoma and advanced papillary thyroid carcinoma) show fatal outcomes and require novel treatments. Immunotherapy is a promising avenue for the treatment of advanced thyroid carcinoma. B7-H3 (B7 homolog 3 protein) and ICAM-1 (intercellular adhesion molecule 1), as two important immune checkpoints (ICPs), is becoming hopeful target spots for immunotherapy. A growing amount of evidence has suggested that B7-H3 and ICAM-1 are upregulated in papillary thyroid carcinoma. However, their expression level in specific types of thyroid cancer remains largely unclear. In the present study, we explored the expression level of B7-H3 and ICAM-1 in different types of thyroid carcinoma. In the groups of the TCGA cohort, both B7-H3 and ICAM-1 mRNA were highly expressed in thyroid carcinoma. Furthermore, the patients with Stage2, 61-80y, Follicular thyroid papillary carcinoma and N0 had lower B7-H3 and ICAM-1 mRNA expression. In the groups of our cohort, PTCs and ATCs showed frequently moderate to strong expression of B7-H3 and ICAM-1 protein expression. The significant relevance of B7-H3 staining score with ICAM-1 staining score was observed in TCGA database and our cohort, which might open avenues for the combination therapy in advanced thyroid cancer.


Subject(s)
B7 Antigens , Intercellular Adhesion Molecule-1 , Thyroid Neoplasms , Humans , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/therapy , Thyroid Neoplasms/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , B7 Antigens/metabolism , B7 Antigens/genetics , Male , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Aged , Aged, 80 and over , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/therapy , Thyroid Cancer, Papillary/metabolism , Adult
3.
Arch Dermatol Res ; 316(7): 382, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850312

ABSTRACT

Cutaneous squamous cell carcinoma (CSCC) is the second most common malignant tumor of the skin. B7 homolog 4 (B7-H4) and B7-H5 (B7 homolog 5) are associated with a variety of tumors. Investigate the potential role of B7-H4 and B7-H5 in regulating the tumorigenesis and progression of CSCC. B7-H4 and B7-H5 transcriptome data were collected from GEO and TCGA databases and subjected to bioinformatical analysis by protein-protein interaction (PPI) network, functional enrichment analysis, immune analysis, and drug-gene interaction prediction analysis. We characterized the expression of B7-H4 and B7-H5 in carcinoma tissues of CSCC patients by immunohistochemistry. Meanwhile, the clinical correlation of B7-H4 and B7-H5 in CSCC was explored by statistical analysis. B7-H4 and B7-H5 genes were under-expressed in CSCC and correlated with tumor staging. According to GO and KEGG Pathway enrichment analysis, B7-H4, and B7-H5 can regulate the proliferation and activation of T cells, lymphocytes, and monocytes, and the expression of cytokines, such as IL-6 and IL-10, in CSCC. B7-H4 and B7-H5 are also jointly involved in the occurrence and development of CSCC via the JAK-STAT and Notch signaling pathways. We found that B7-H4 and B7-H5 proteins were abnormally highly expressed in CSCC tissue and correlated with tumor size and stage. Our findings offer new insights into the pathogenesis of CSCC and suggest that B7-H4 and B7-H5 are novel tissue biomarkers and promising therapeutic targets for CSCC.


Subject(s)
Carcinoma, Squamous Cell , Gene Expression Regulation, Neoplastic , Skin Neoplasms , V-Set Domain-Containing T-Cell Activation Inhibitor 1 , Aged , Female , Humans , Male , Middle Aged , B7 Antigens/metabolism , B7 Antigens/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Immunoglobulins , Neoplasm Staging , Protein Interaction Maps , Signal Transduction , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism
4.
Cancer Lett ; 596: 217018, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38844062

ABSTRACT

Relapse and treatment resistance pose significant challenges in the management of pediatric B cell acute lymphoblastic leukemia (B-ALL) and acute myeloid leukemia (AML). The efficacy of immunotherapy in leukemia remains limited due to factors such as the immunosuppressive tumor microenvironment (TME) and lack of suitable immunotherapeutic targets. Thus, an in-depth characterization of the TME in pediatric leukemia is warranted to improve the efficacy of immunotherapy. Here, we used single-cell RNA sequencing (scRNA-seq) to characterize the TME of pediatric B-ALL and AML, focusing specifically on bone-marrow-derived T cells. Moreover, we investigated the transcriptome changes during the initiation, remission, and relapse stages of pediatric AML. Our findings revealed that specific functional expression programs correlated with fluctuations in various T cell subsets, which may be associated with AML progression and relapse. Furthermore, our analysis of cellular communication networks led to the identification of VISTA, CD244, and TIM3 as potential immunotherapeutic targets in pediatric AML. Finally, we detected elevated proportions of γδ T cells and associated functional genes in samples from pediatric patients diagnosed with B-ALL and AML, which could inform the development of novel therapeutic approaches, potentially focusing on γδ T cells.


Subject(s)
Leukemia, Myeloid, Acute , Single-Cell Analysis , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Single-Cell Analysis/methods , Child , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Transcriptome , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Gene Expression Profiling/methods , Child, Preschool , Male , Female , B7 Antigens/genetics , Adolescent , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Gene Expression Regulation, Leukemic
5.
Cancer Immunol Immunother ; 73(8): 136, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833004

ABSTRACT

A checkpoint protein called the V-domain Ig suppressor of T cell activation (VISTA) is important for controlling immune responses. Immune cells that interact with VISTA have molecules, or receptors, known as VISTA receptors. Immune system activity can be modified by the interaction between VISTA and its receptors. Since targeting VISTA or its receptors may be beneficial in certain conditions, VISTA has been studied in relation to immunotherapy for cancer and autoimmune illnesses. The purpose of this study was to examine the expression levels and interactions between VISTA and its receptors, VSIG3 and PSGL-1, in breast cancer tissues. IHC analysis revealed higher levels of proteins within the VISTA/VSIG3/PSGL-1 axis in cancer tissues than in the reference samples (mastopathies). VISTA was found in breast cancer cells and intratumoral immune cells, with membranous and cytoplasmic staining patterns. VISTA was also linked with pathological grade and VSIG3 and PSGL-1 levels. Furthermore, we discovered that the knockdown of one axis member boosted the expression of the other partners. This highlights the significance of VISTA/VSIG3/PSGL-1 in tumor stroma and microenvironment remodeling. Our findings indicate the importance of the VISTA/VSIG3/PSGL-1 axis in the molecular biology of cancer cells and the immune microenvironment.


Subject(s)
B7 Antigens , Breast Neoplasms , Carcinoma, Ductal, Breast , Membrane Glycoproteins , Humans , Female , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/immunology , B7 Antigens/metabolism , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/metabolism , Tumor Microenvironment/immunology , Middle Aged
6.
Sci Rep ; 14(1): 14207, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902359

ABSTRACT

This study aimed to confirm urinary protein fragments in relation to the presence of pancreatic ductal adenocarcinoma (PDAC) via a C-terminal proteomics strategy using exploratory and validation cohorts. Urinary fragments were examined by iTRAQ-labelling of tryptic peptides and concentrations of C-terminal fragments were evaluated. Only the urinary CD276 fragment showed a fold change (FC) of > 1.5 with a significant difference of P < 0.01 between healthy (H) and PDAC participants in both the exploratory (H, n = 42; PDAC, n = 39) and validation cohorts (H, n = 36; resectable PDAC, n = 28). The sensitivity and specificity of the CD276 fragment for diagnosing resectable PDAC were 75% and 89%, respectively, in the validation cohort. Postoperative urinary levels of the CD276 fragment were low as compared to those before surgery (n = 18, P < 0.01). Comprehensive C-terminus proteomics identified an increase in the urinary CD276 fragment level as a feature of patients with PDAC. The urinary CD276 fragment is a potential biomarker for detecting resectable PDAC.


Subject(s)
B7 Antigens , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Proteomics , Humans , Pancreatic Neoplasms/urine , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/diagnosis , Proteomics/methods , Female , Male , Biomarkers, Tumor/urine , Aged , B7 Antigens/urine , B7 Antigens/metabolism , Middle Aged , Carcinoma, Pancreatic Ductal/urine , Carcinoma, Pancreatic Ductal/surgery , Carcinoma, Pancreatic Ductal/diagnosis
7.
Sci Rep ; 14(1): 13552, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866941

ABSTRACT

Antibodies have been extensively used in numerous applications within proteomics-based technologies, requiring high sensitivity, specificity, a broad dynamic range for detection, and precise, reproducible quantification. Seeking alternatives to antibodies due to several inherent limitations of antibodies is an area of active research of tremendous importance. Recently, aptamers have been receiving increasing attention, because they not only have all of the advantages of antibodies, but also have unique advantages, such as thermal stability, low cost, and unlimited applications. Aptamers are gaining importance in immunological studies and can potentially replace antibodies in immunoassays. B7H3, an immunoregulatory protein belonging to the B7 family, is an attractive and promising target due to its overexpression in several tumor tissues while exhibiting limited expression in normal tissues. This study employed hybrid-SELEX with next-generation sequencing to select ssDNA aptamers specifically binding to the B7H3 protein. These aptamers demonstrated versatility across various assays, including flow cytometry, dot-blot, and immunohistochemistry. Effective performance in sandwich dot-blot assays and western blot analysis suggests their potential for diagnostic applications and demonstrates their adaptability and cost-effectiveness in diverse protein detection techniques.


Subject(s)
Aptamers, Nucleotide , B7 Antigens , SELEX Aptamer Technique , B7 Antigens/immunology , B7 Antigens/metabolism , Humans , SELEX Aptamer Technique/methods , High-Throughput Nucleotide Sequencing/methods , Cell Line, Tumor , Antibodies/immunology
8.
Sci Immunol ; 9(95): eadj7970, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701193

ABSTRACT

Understanding the mechanisms that regulate T cell immunity is critical for the development of effective therapies for diseases associated with T cell dysfunction, including autoimmune diseases, chronic infections, and cancer. Co-inhibitory "checkpoint molecules," such as programmed cell death protein-1, balance excessive or prolonged immune activation by T cell-intrinsic signaling. Here, by screening for mediators of natural killer (NK) cell recognition on T cells, we identified the immunoglobulin superfamily ligand B7H6 to be highly expressed by activated T cells, including patient-infused CD19-targeting chimeric antigen receptor (CAR) T cells. Unlike other checkpoint molecules, B7H6 mediated NKp30-dependent recognition and subsequent cytolysis of activated T cells by NK cells. B7H6+ T cells were prevalent in the tissue and blood of several diseases, and their abundance in tumor tissue positively correlated with clinical response in a cohort of patients with immune checkpoint inhibitor-treated esophageal cancer. In humanized mouse models, NK cell surveillance via B7H6 limited the persistence and antitumor activity of CAR T cells, and its genetic deletion enhanced T cell proliferation and persistence. Together, we provide evidence of B7H6 protein expression by activated T cells and suggest the B7H6-NKp30 axis as a therapeutically actionable NK cell-dependent immune checkpoint that regulates human T cell function.


Subject(s)
B7 Antigens , Killer Cells, Natural , T-Lymphocytes , Humans , Killer Cells, Natural/immunology , Animals , Mice , B7 Antigens/immunology , T-Lymphocytes/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Lymphocyte Activation/immunology , Female , Esophageal Neoplasms/immunology
9.
Front Immunol ; 15: 1391954, 2024.
Article in English | MEDLINE | ID: mdl-38765008

ABSTRACT

Sarcomas are rare and heterogeneous malignancies that are difficult to treat. Approximately 50% of patients diagnosed with sarcoma develop metastatic disease with so far very limited treatment options. The transmembrane protein B7-H3 reportedly is expressed in various malignancies, including different sarcoma subtypes. In several cancer entities B7-H3 expression is associated with poor prognosis. In turn, B7-H3 is considered a promising target for immunotherapeutic approaches. We here report on the preclinical characterization of a B7-H3xCD3 bispecific antibody in an IgG-based format, termed CC-3, for treatment of different sarcoma subtypes. We found B7-H3 to be expressed on all sarcoma cells tested and expression on sarcoma patients correlated with decreased progression-free and overall survival. CC-3 was found to elicit robust T cell responses against multiple sarcoma subtypes, resulting in significant activation, release of cytokines and effector molecules. In addition, CC-3 promoted T cell proliferation and differentiation, resulting in the generation of memory T cell subsets. Finally, CC-3 induced potent target cell lysis in a target cell restricted manner. Based on these results, a clinical trial evaluating CC-3 in soft tissue sarcoma is currently in preparation.


Subject(s)
Antibodies, Bispecific , B7 Antigens , Sarcoma , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Sarcoma/immunology , Sarcoma/drug therapy , B7 Antigens/immunology , B7 Antigens/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Female , Male , Animals , Lymphocyte Activation/immunology , Middle Aged , CD3 Complex/immunology , Aged , Cell Proliferation , Adult
10.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791382

ABSTRACT

The identification of targets that are expressed on the cell membrane is a main goal in cancer research. The Lymphocyte Antigen 6 Family Member G6D (LY6G6D) gene codes for a protein that is mainly present on the surface of colorectal cancer (CRC) cells. Therapeutic strategies against this protein like the development of T cell engagers (TCE) are currently in the early clinical stage. In the present work, we interrogated public genomic datasets including TCGA to evaluate the genomic and immunologic cell profile present in tumors with high expression of LY6G6D. We used data from TCGA, among others, and the Tumor Immune Estimation Resource (TIMER2.0) platform for immune cell estimations and Spearman correlation tests. LY6G6D expression was exclusively present in CRC, particularly in the microsatellite stable (MSS) subtype, and was associated with left-side tumors and the canonical genomic subgroup. Tumors with mutations of APC and p53 expressed elevated levels of LY6G6D. This protein was expressed in tumors with an inert immune microenvironment with an absence of immune cells and co-inhibitory molecules. In conclusion, we described clinical, genomic and immune-pathologic characteristics that can be used to optimize the clinical development of agents against this target. Future studies should be performed to confirm these findings and potentially explore the suggested clinical development options.


Subject(s)
Colorectal Neoplasms , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Female , Male , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Middle Aged , Aged , Biomarkers, Tumor/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Antigens, Ly/metabolism , Antigens, Ly/genetics , B7 Antigens/genetics , B7 Antigens/metabolism
11.
Front Immunol ; 15: 1388018, 2024.
Article in English | MEDLINE | ID: mdl-38698855

ABSTRACT

Natural killer (NK) cells are key effectors in cancer immunosurveillance, eliminating a broad spectrum of cancer cells without major histocompatibility complex (MHC) specificity and graft-versus-host diseases (GvHD) risk. The use of allogeneic NK cell therapies from healthy donors has demonstrated favorable clinical efficacies in treating diverse cancers, particularly hematologic malignancies, but it requires cytokines such as IL-2 to primarily support NK cell persistence and expansion. However, the role of IL-2 in the regulation of activating receptors and the function of NK cells expanded for clinical trials is poorly understood and needs clarification for the full engagement of NK cells in cancer immunotherapy. Here, we demonstrated that IL-2 deprivation significantly impaired the cytotoxicity of primary expanded NK cells by preferentially downregulating NKp30 but not NKp46 despite their common adaptor requirement for expression and function. Using NK92 and IL-2-producing NK92MI cells, we observed that NKp30-mediated cytotoxicity against myeloid leukemia cells such as K562 and THP-1 cells expressing B7-H6, a ligand for NKp30, was severely impaired by IL-2 deprivation. Furthermore, IL-2 deficiency-mediated NK cell dysfunction was overcome by the ectopic overexpression of an immunostimulatory NKp30 isoform such as NKp30a or NKp30b. In particular, NKp30a overexpression in NK92 cells improved the clearance of THP-1 cells in vivo without IL-2 supplementation. Collectively, our results highlight the distinct role of IL-2 in the regulation of NKp30 compared to that of NKp46 and suggest NKp30 upregulation, as shown here by ectopic overexpression, as a viable modality to harness NK cells in cancer immunotherapy, possibly in combination with IL-2 immunocytokines.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-2 , Killer Cells, Natural , Natural Cytotoxicity Triggering Receptor 3 , Humans , Natural Cytotoxicity Triggering Receptor 3/immunology , Natural Cytotoxicity Triggering Receptor 3/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Interleukin-2/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , K562 Cells , THP-1 Cells , B7 Antigens/genetics , B7 Antigens/metabolism , B7 Antigens/immunology
12.
BMC Cancer ; 24(1): 650, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802739

ABSTRACT

OBJECTIVE: This study aimed to explore the effect of CD276 expression on the sunitinib sensitivity of clear cell renal cell carcinoma (ccRCC) cell and animal models and the potential mechanisms involved. METHODS: CD276 expression levels of ccRCC and normal samples were analyzed via online databases and real-time quantitative PCR (RT-qPCR). CD276 was knocked down in ccRCC cell models (sunitinib-resistant 786-O/R cells and sunitinib-sensitive 786-O cells) using shRNA transfection, and the cells were exposed to a sunitinib (2 µM) environment. Cells proliferation was then analyzed using MTT assay and colony formation experiment. Alkaline comet assay, immunofluorescent staining, and western blot experiments were conducted to assess the DNA damage repair ability of the cells. Western blot was also used to observe the activation of FAK-MAPK pathway within the cells. Finally, a nude mouse xenograft model was established and the nude mice were orally administered sunitinib (40 mg/kg/d) to evaluate the in vivo effects of CD276 knockdown on the therapeutic efficacy of sunitinib against ccRCC. RESULTS: CD276 was significantly upregulated in both ccRCC clinical tissue samples and cell models. In vitro experiments showed that knocking down CD276 reduced the survival rate, IC50 value, and colony-forming ability of ccRCC cells. Knocking down CD276 increased the comet tail moment (TM) values and γH2AX foci number, and reduced BRCA1 and RAD51 protein levels. Knocking down CD276 also decreased the levels of p-FAK, p-MEK, and p-ERK proteins. CONCLUSION: Knocking down CD276 effectively improved the sensitivity of ccRCC cell and animal models to sunitinib treatment.


Subject(s)
Carcinoma, Renal Cell , DNA Damage , DNA Repair , Drug Resistance, Neoplasm , Kidney Neoplasms , Mice, Nude , Sunitinib , Xenograft Model Antitumor Assays , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Humans , Sunitinib/pharmacology , Sunitinib/therapeutic use , Animals , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Mice , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , DNA Damage/drug effects , MAP Kinase Signaling System/drug effects , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Gene Knockdown Techniques , Male , B7 Antigens
13.
Cancer Res Commun ; 4(5): 1369-1379, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38709075

ABSTRACT

B7-H3 (CD276) is a transmembrane glycoprotein of the B7 immune checkpoint superfamily that has emerged as a promising therapeutic target. To better understand the applicability of B7-H3-directed therapies, we analyzed 156,791 samples comprising 50 cancer types to interrogate the clinical, genomic, transcriptomic, and immunologic correlates of B7-H3 mRNA expression. DNA (592-gene/whole-exome) and RNA (whole-transcriptome) sequencing was performed from samples submitted to Caris Life Sciences. B7-H3 high versus low expression was based on top and bottom quartiles for each cancer type. Patients' overall survival was determined from insurance claims data. Pathway analysis was performed using gene set enrichment analyses. Immune cell fractions were inferred using quanTIseq. B7-H3 is expressed across several human malignancies including prostate, pancreatic, ovarian, and lung cancers. High B7-H3 expression is associated with differences in overall survival, possibly indicating a prognostic role of B7-H3 for some cancers. When examining molecular features across all cancer types, we did not identify recurrent associations between B7-H3 expression and genetic alterations in TP53, RB1, and KRAS. However, we find consistent enrichment of epithelial-to-mesenchymal transition, Wnt, TGFß, and Notch signaling pathways. In addition, tumors with high B7-H3 expression are associated with greater proportions of M1 macrophages, but lower fractions of CD8+ T cells. We have begun to define the genomic, transcriptomic, clinical, and immunologic features associated with B7-H3 expression in 50 cancer types. We report novel clinical and molecular features of B7-H3-high tumors which may inform how current B7-H3 therapeutics should be deployed and prioritized. SIGNIFICANCE: B7-H3-targeting therapeutics have shown promising results in initial clinical trials. In this pan-cancer analysis of B7-H3 mRNA expression, we found that B7-H3 exhibits robust expression in many common cancer types. These results may inform further development of B7-H3-targeting therapeutics and may guide clinical decisions for patients with limited treatment options.


Subject(s)
B7 Antigens , Neoplasms , Humans , B7 Antigens/genetics , B7 Antigens/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/therapy , Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Prognosis , Male , Female
14.
Cells ; 13(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38786018

ABSTRACT

Cancer immunotherapy is a rapidly developing field of medicine that aims to use the host's immune mechanisms to inhibit and eliminate cancer cells. Antibodies targeting CTLA-4, PD-1, and its ligand PD-L1 are used in various cancer therapies. However, the most thoroughly researched pathway targeting PD-1/PD-L1 has many limitations, and multiple malignancies resist its effects. Human endogenous retrovirus-H Long repeat-associating 2 (HHLA2, known as B7H5/B7H7/B7y) is the youngest known molecule from the B7 family. HHLA2/TMIGD2/KIRD3DL3 is one of the critical pathways in modulating the immune response. Recent studies have demonstrated that HHLA2 has a double effect in modulating the immune system. The connection of HHLA2 with TMIGD2 induces T cell growth and cytokine production via an AKT-dependent signaling cascade. On the other hand, the binding of HHLA2 and KIR3DL3 leads to the inhibition of T cells and mediates tumor resistance against NK cells. This review aimed to summarize novel information about HHLA2, focusing on immunological mechanisms and clinical features of the HHLA2/KIR3DL3/TMIGD2 pathway in the context of potential strategies for malignancy treatment.


Subject(s)
Neoplasms , Humans , Neoplasms/immunology , Neoplasms/metabolism , Immunotherapy/methods , B7 Antigens/metabolism , Animals , Signal Transduction , Immunoglobulins
15.
J Cell Mol Med ; 28(10): e18360, 2024 May.
Article in English | MEDLINE | ID: mdl-38785199

ABSTRACT

Neuroblastoma (NB), a common solid tumour in young children originating from the sympathetic nervous system during embryonic development, poses challenges despite therapeutic advances like high-dose chemotherapy and immunotherapy. Some survivors still grapple with severe side effects and drug resistance. The role of lncRNA NUTM2A-AS1 has been explored in various cancers, but its function in drug-resistant NB progression is unclear. Our study found that NUTM2A-AS1 expression in cisplatin-resistant NB cells increased in a time- and dose-dependent manner. Knockdown of NUTM2A-AS1 significantly improved NB cell sensitivity to cisplatin and inhibited metastatic abilities. Additionally, we identified B7-H3, an immune checkpoint-related protein, as a NUTM2A-AS1-associated protein in NB cells. NUTM2A-AS1 was shown to inhibit the protein degradation of B7-H3. Moreover, NUTM2A-AS1 modulated immune evasion in cisplatin-resistant NB cells through B7-H3. Furthermore, NUTM2A-AS1 expression in cisplatin-resistant NB cells was transactivated by NR1D1. In summary, our results unveil the molecular or biological relationship within the NR1D1/NUTM2A-AS1/B7-H3 axis in NB cells under cisplatin treatment, providing an intriguing avenue for fundamental research into cisplatin-resistant NB.


Subject(s)
B7 Antigens , Cisplatin , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Neuroblastoma , RNA, Long Noncoding , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Drug Resistance, Neoplasm/genetics , B7 Antigens/metabolism , B7 Antigens/genetics , RNA, Long Noncoding/genetics , Cisplatin/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Immune Evasion , Animals , Proteolysis/drug effects , Mice
16.
Sci Immunol ; 9(95): eadi7418, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758807

ABSTRACT

Immune checkpoint blockade is a promising approach to activate antitumor immunity and improve the survival of patients with cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint target; however, the downstream signaling mechanisms are elusive. Here, we identify leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) as a VISTA binding partner, which acts as an inhibitory receptor by engaging VISTA and suppressing T cell receptor signaling pathways. Mice with T cell-specific LRIG1 deletion developed superior antitumor responses because of expansion of tumor-specific cytotoxic T lymphocytes (CTLs) with increased effector function and survival. Sustained tumor control was associated with a reduction of quiescent CTLs (TCF1+ CD62Lhi PD-1low) and a reciprocal increase in progenitor and memory-like CTLs (TCF1+ PD-1+). In patients with melanoma, elevated LRIG1 expression on tumor-infiltrating CD8+ CTLs correlated with resistance to immunotherapies. These results delineate the role of LRIG1 as an inhibitory immune checkpoint receptor and propose a rationale for targeting the VISTA/LRIG1 axis for cancer immunotherapy.


Subject(s)
B7 Antigens , CD8-Positive T-Lymphocytes , Membrane Glycoproteins , Mice, Inbred C57BL , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/genetics , Humans , B7 Antigens/immunology , B7 Antigens/genetics , Mice, Knockout , Cell Line, Tumor , Female , Membrane Proteins , Nerve Tissue Proteins
17.
Cancer Res Commun ; 4(6): 1410-1429, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38717140

ABSTRACT

Encouraged by the observations of significant B7-H3 protein overexpression in many human solid tumors compared to healthy tissues, we directed our focus towards targeting B7-H3 using chimeric antigen receptor (CAR) T cells. We utilized a nanobody as the B7-H3-targeting domain in our CAR construct to circumvent the stability issues associated with single-chain variable fragment-based domains. In efforts to expand patient access to CAR T-cell therapy, we engineered our nanobody-based CAR into human Epstein-Barr virus-specific T cells (EBVST), offering a readily available off-the-shelf treatment. B7H3.CAR-armored EBVSTs demonstrated potent in vitro and in vivo activities against multiple B7-H3-positive human tumor cell lines and patient-derived xenograft models. Murine T cells expressing a murine equivalent of our B7H3.CAR exhibited no life-threatening toxicities in immunocompetent mice bearing syngeneic tumors. Further in vitro evaluation revealed that while human T, B, and natural killer cells were unaffected by B7H3.CAR EBVSTs, monocytes were targeted because of upregulation of B7-H3. Such targeting of myeloid cells, which are key mediators of cytokine release syndrome (CRS), contributed to a low incidence of CRS in humanized mice after B7H3.CAR EBVST treatment. Notably, we showed that B7H3.CAR EBVSTs can target B7-H3-expressing myeloid-derived suppressor cells (MDSC), thereby mitigating MDSC-driven immune suppression. In summary, our data demonstrate that our nanobody-based B7H3.CAR EBVSTs are effective as an off-the-shelf therapy for B7-H3-positive solid tumors. These cells also offer an avenue to modulate the immunosuppressive tumor microenvironment, highlighting their promising clinical potential in targeting solid tumors. SIGNIFICANCE: Clinical application of EBVSTs armored with B7-H3-targeting CARs offer an attractive solution to translate off-the-shelf CAR T cells as therapy for solid tumors.


Subject(s)
B7 Antigens , Herpesvirus 4, Human , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Xenograft Model Antitumor Assays , Animals , Humans , B7 Antigens/immunology , B7 Antigens/metabolism , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Herpesvirus 4, Human/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/immunology , Female , Single-Domain Antibodies/immunology
18.
Int Immunopharmacol ; 136: 112365, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38820964

ABSTRACT

Growing evidence had showed that tumor-associated macrophages (TAMs) have a tumor-promoting M2 phenotype which could drive pathological phenomena. In breast cancer, TAMs are abundantly present and may play an important role in the development of breast cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel inhibitory checkpoint and immunotherapy target for tumor through regulating immune response. However, its effects on macrophages have not been investigated, which was also the focus of this study. Here, the scRNA-seq data further revealed that VISTA was highly expressed in multiple macrophage subclusters. In vitro experiments showed that the absence of VISTA enhanced the M1 polarization of macrophages, inhibited the M2 polarization of macrophages and the proliferation and phagocytosis of 4 T1 cells induced by M2-CM. VISTA regulated the activation of STAT1 and STAT6 signaling pathways in the process of macrophage polarization. In vivo experiments demonstrated that VISTA deficient mice exhibited reduced tumor growth, possibly due to the increase of M1 macrophages and the decrease of M2 macrophages. In summary, our study is the first to reveal the effect of VISTA on macrophages in breast cancer, which showed that VISTA affects tumor growth by critically regulating the macrophage polarization through the STAT pathway.


Subject(s)
Breast Neoplasms , STAT1 Transcription Factor , STAT6 Transcription Factor , Tumor-Associated Macrophages , Animals , Female , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Mice , STAT6 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Humans , B7 Antigens/genetics , B7 Antigens/metabolism , Mice, Knockout , Cell Line, Tumor , Macrophages/immunology , Signal Transduction , Macrophage Activation , Phagocytosis , Cell Proliferation , Mice, Inbred C57BL , Membrane Proteins
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 435-442, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38790100

ABSTRACT

Objective To elucidate the expression characteristics and clinical significance of B7 homolog 3(B7-H3) in colorectal cancer (CRC) and to explore its associations with tumor glycolysis and immune cell infiltration in the tumor microenvironment. Methods The transcriptomic and clinicopathological data of CRC were obtained from the TCGA database and analyzed to determine the expression and clinical relevance of B7-H3. Correlations between glycolysis-related genes and B7-H3 expression were assessed based on TCGA data. The associations between B7-H3 expression and the infiltration of 22 types of immune cells were analyzed using the CIBERSORT algorithm. Immunohistochemical staining was used to verify the results of database analysis in surgical specimens and adjacent normal tissue specimens of 51 CRC patients.Results B7-H3 was significantly upregulated in CRC tissues and demonstrated strong correlations with tumor invasion depth, advanced TNM stage, and poor prognosis. Additionally, B7-H3 expression was closely associated with the upregulation of glycolysis-related genes in CRC. Immune cell infiltration analysis revealed that a total of 16 types of immune cells were significantly correlated with B7-H3 expression. Furthermore, an inverse relationship between B7-H3 expression and CD8+T cell infiltration was identified at the transcriptional level but not at the protein level. Conclusion B7-H3 is highly expressed in CRC tissues and is significantly correlated with disease progression and poor prognosis. Furthermore, the association of B7-H3 expression with glycolysis-related genes and immune cell infiltration suggests a pivotal role of B7-H3 in the regulation of tumor glycolysis and cancer immunity.


Subject(s)
B7 Antigens , Colorectal Neoplasms , Humans , B7 Antigens/genetics , B7 Antigens/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Male , Female , Middle Aged , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Prognosis , Aged , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Clinical Relevance
20.
J Immunother Cancer ; 12(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38724465

ABSTRACT

BACKGROUND: CD276 (B7-H3), a pivotal immune checkpoint, facilitates tumorigenicity, invasiveness, and metastasis by escaping immune surveillance in a variety of tumors; however, the underlying mechanisms facilitating immune escape in esophageal squamous cell carcinoma (ESCC) remain enigmatic. METHODS: We investigated the expression of CD276 in ESCC tissues from patients by using immunohistochemistry (IHC) assays. In vivo, we established a 4-nitroquinoline 1-oxide (4NQO)-induced CD276 knockout (CD276wKO) and K14cre; CD276 conditional knockout (CD276cKO) mouse model of ESCC to study the functional role of CD276 in ESCC. Furthermore, we used the 4NQO-induced mouse model to evaluate the effects of anti-CXCL1 antibodies, anti-Ly6G antibodies, anti-NK1.1 antibodies, and GSK484 inhibitors on tumor growth. Moreover, IHC, flow cytometry, and immunofluorescence techniques were employed to measure immune cell proportions in ESCC. In addition, we conducted single-cell RNA sequencing analysis to examine the alterations in tumor microenvironment following CD276 depletion. RESULTS: In this study, we elucidate that CD276 is markedly upregulated in ESCC, correlating with poor prognosis. In vivo, our results indicate that depletion of CD276 inhibits tumorigenesis and progression of ESCC. Furthermore, conditional knockout of CD276 in epithelial cells engenders a significant downregulation of CXCL1, consequently reducing the formation of neutrophil extracellular trap networks (NETs) via the CXCL1-CXCR2 signaling axis, while simultaneously augmenting natural killer (NK) cells. In addition, overexpression of CD276 promotes tumorigenesis via increasing NETs' formation and reducing NK cells in vivo. CONCLUSIONS: This study successfully elucidates the functional role of CD276 in ESCC. Our comprehensive analysis uncovers the significant role of CD276 in modulating immune surveillance mechanisms in ESCC, thereby suggesting that targeting CD276 might serve as a potential therapeutic approach for ESCC treatment.


Subject(s)
B7 Antigens , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Extracellular Traps , Animals , Female , Humans , Male , Mice , B7 Antigens/metabolism , Chemokine CXCL1/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Extracellular Traps/metabolism , Mice, Knockout , Receptors, Interleukin-8B/metabolism , Tumor Escape , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...