Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
JCI Insight ; 9(7)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441970

ABSTRACT

Compromised vascular integrity facilitates extravasation of cancer cells and promotes metastatic dissemination. CD93 has emerged as a target for antiangiogenic therapy, but its importance for vascular integrity in metastatic cancers has not been evaluated. Here, we demonstrate that CD93 participates in maintaining the endothelial barrier and reducing metastatic dissemination. Primary melanoma growth was hampered in CD93-/- mice, but metastatic dissemination was increased and associated with disruption of adherens and tight junctions in tumor endothelial cells and elevated expression of matrix metalloprotease 9 at the metastatic site. CD93 directly interacted with vascular endothelial growth factor receptor 2 (VEGFR2) and its absence led to VEGF-induced hyperphosphorylation of VEGFR2 in endothelial cells. Antagonistic anti-VEGFR2 antibody therapy rescued endothelial barrier function and reduced the metastatic burden in CD93-/- mice to wild-type levels. These findings reveal a key role of CD93 in maintaining vascular integrity, which has implications for pathological angiogenesis and endothelial barrier function in metastatic cancer.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Mice , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Cancer Cell ; 42(3): 328-330, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38366590

ABSTRACT

Brain metastases are clinically challenging due to the unique brain microenvironment. In this issue of Cancer Cell, Bejarano et al. use transcriptional profiling and data integration to shed light on the molecular and cellular composition of the vasculature in brain metastases, identifying CD276 as an immunomodulatory target for therapy.


Subject(s)
Brain Neoplasms , Humans , Brain Neoplasms/secondary , Immunomodulation , Tumor Microenvironment , B7 Antigens
3.
Front Immunol ; 14: 1275378, 2023.
Article in English | MEDLINE | ID: mdl-37954592

ABSTRACT

Tertiary lymphoid structures (TLS) are lymph node-like aggregates that can form in association with chronic inflammation or cancer. Mature TLS are organized into B and T cell zones, and are not encapsulated but include all cell types necessary for eliciting an adaptive immune response. TLS have been observed in various cancer types and are generally associated with a positive prognosis as well as increased sensitivity to cancer immunotherapy. However, a comprehensive understanding of the roles of TLS in eliciting anti-tumor immunity as well as the mechanisms involved in their formation and function is still lacking. Further studies in orthotopic, immunocompetent cancer models are necessary to evaluate the influence of TLS on cancer therapies, and to develop new treatments that promote their formation in cancer. Here, we review key insights obtained from functional murine studies, discuss appropriate models that can be used to study cancer-associated TLS, and suggest guidelines on how to identify TLS and distinguish them from other antigen-presenting niches.


Subject(s)
Neoplasms , Tertiary Lymphoid Structures , Animals , Mice , Neoplasms/therapy , Neoplasms/pathology , Prognosis , Lymph Nodes/metabolism , Inflammation/pathology
5.
J Clin Invest ; 133(20)2023 10 16.
Article in English | MEDLINE | ID: mdl-37651195

ABSTRACT

Endothelial phospholipase Cγ (PLCγ) is essential for vascular development; however, its role in healthy, mature, or pathological vessels is unexplored. Here, we show that PLCγ was prominently expressed in vessels of several human cancer forms, notably in renal cell carcinoma (RCC). High PLCγ expression in clear cell RCC correlated with angiogenic activity and poor prognosis, while low expression correlated with immune cell activation. PLCγ was induced downstream of vascular endothelial growth factor receptor 2 (VEGFR2) phosphosite Y1173 (pY1173). Heterozygous Vegfr2Y1173F/+ mice or mice lacking endothelial PLCγ (Plcg1iECKO) exhibited a stabilized endothelial barrier and diminished vascular leakage. Barrier stabilization was accompanied by decreased expression of immunosuppressive cytokines, reduced infiltration of B cells, helper T cells and regulatory T cells, and improved response to chemo- and immunotherapy. Mechanistically, pY1173/PLCγ signaling induced Ca2+/protein kinase C-dependent activation of endothelial nitric oxide synthase (eNOS), required for tyrosine nitration and activation of Src. Src-induced phosphorylation of VE-cadherin at Y685 was accompanied by disintegration of endothelial junctions. This pY1173/PLCγ/eNOS/Src pathway was detected in both healthy and tumor vessels in Vegfr2Y1173F/+ mice, which displayed decreased activation of PLCγ and eNOS and suppressed vascular leakage. Thus, we believe that we have identified a clinically relevant endothelial PLCγ pathway downstream of VEGFR2 pY1173, which destabilizes the endothelial barrier and results in loss of antitumor immunity.


Subject(s)
Capillary Permeability , Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Humans , Mice , Capillary Permeability/genetics , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , CSK Tyrosine-Protein Kinase/metabolism
6.
Nat Commun ; 14(1): 4732, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563127

ABSTRACT

Chimeric antigen receptor (CAR)-T cell therapy is rapidly advancing as cancer treatment, however, designing an optimal CAR remains challenging. A single-chain variable fragment (scFv) is generally used as CAR targeting moiety, wherein the complementarity-determining regions (CDRs) define its specificity. We report here that the CDR loops can cause CAR clustering, leading to antigen-independent tonic signalling and subsequent CAR-T cell dysfunction. We show via CARs incorporating scFvs with identical framework and varying CDR sequences that CARs may cluster on the T cell surface, which leads to antigen-independent CAR-T cell activation, characterized by increased cell size and interferon (IFN)-γ secretion. This results in CAR-T cell exhaustion, activation-induced cell death and reduced responsiveness to target-antigen-expressing tumour cells. CDR mutagenesis confirms that the CAR-clustering is mediated by CDR-loops. In summary, antigen-independent tonic signalling can be induced by CDR-mediated CAR clustering, which could not be predicted from the scFv sequences, but could be tested for by evaluating the activity of unstimulated CAR-T cells.


Subject(s)
Complementarity Determining Regions , Single-Chain Antibodies , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , T-Lymphocytes , Immunotherapy, Adoptive/methods , Signal Transduction , Cell Line, Tumor , Receptors, Antigen, T-Cell/metabolism
8.
Cancer Cell ; 41(6): 1134-1151.e10, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37172581

ABSTRACT

Glioblastomas are aggressive brain tumors that are largely immunotherapy resistant. This is associated with immunosuppression and a dysfunctional tumor vasculature, which hinder T cell infiltration. LIGHT/TNFSF14 can induce high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), suggesting that its therapeutic expression could promote T cell recruitment. Here, we use a brain endothelial cell-targeted adeno-associated viral (AAV) vector to express LIGHT in the glioma vasculature (AAV-LIGHT). We found that systemic AAV-LIGHT treatment induces tumor-associated HEVs and T cell-rich TLS, prolonging survival in αPD-1-resistant murine glioma. AAV-LIGHT treatment reduces T cell exhaustion and promotes TCF1+CD8+ stem-like T cells, which reside in TLS and intratumoral antigen-presenting niches. Tumor regression upon AAV-LIGHT therapy correlates with tumor-specific cytotoxic/memory T cell responses. Our work reveals that altering vascular phenotype through vessel-targeted expression of LIGHT promotes efficient anti-tumor T cell responses and prolongs survival in glioma. These findings have broader implications for treatment of other immunotherapy-resistant cancers.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Mice , Animals , Glioma/genetics , Glioma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/blood supply , Glioblastoma/genetics , Phenotype , Brain , Tumor Microenvironment
9.
FASEB J ; 37(4): e22894, 2023 04.
Article in English | MEDLINE | ID: mdl-36961390

ABSTRACT

Regulation of vascular permeability to plasma is essential for tissue and organ homeostasis and is mediated by endothelial cell-to-cell junctions that tightly regulate the trafficking of molecules between blood and tissue. The single-pass transmembrane glycoprotein CD93 is upregulated in endothelial cells during angiogenesis and controls cytoskeletal dynamics. However, its role in maintaining homeostasis by regulating endothelial barrier function has not been elucidated yet. Here, we demonstrate that CD93 interacts with vascular endothelial (VE)-cadherin and limits its phosphorylation and turnover. CD93 deficiency in vitro and in vivo induces phosphorylation of VE-cadherin under basal conditions, displacing it from endothelial cell-cell contacts. Consistent with this, endothelial junctions are defective in CD93-/- mice, and the blood-brain barrier permeability is enhanced. Mechanistically, CD93 regulates VE-cadherin phosphorylation and turnover at endothelial junctions through the Rho/Rho kinase-dependent pathway. In conclusion, our results identify CD93 as a key regulator of VE-cadherin stability at endothelial junctions, opening up possibilities for therapeutic strategies directed to control vascular permeability.


Subject(s)
Cadherins , Endothelial Cells , Animals , Mice , Phosphorylation , Endothelial Cells/metabolism , Cadherins/genetics , Cadherins/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Cells, Cultured , Adherens Junctions/metabolism
10.
Neuro Oncol ; 25(6): 1073-1084, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36591963

ABSTRACT

BACKGROUND: Systemic delivery of anti-tumor therapeutic agents to brain tumors is thwarted by the blood-brain barrier (BBB), an organotypic specialization of brain endothelial cells (ECs). A failure of pharmacological compounds to cross BBB is one culprit for the dismal prognosis of glioblastoma (GBM) patients. Identification of novel vascular targets to overcome the challenges posed by the BBB in tumors for GBM treatment is urgently needed. METHODS: Temozolomide (TMZ) delivery was investigated in CT2A and PDGFB-driven RCAS/tv-a orthotopic glioma models. Transcriptome analysis was performed on ECs from murine gliomas. Mfsd2a deficient, Cav1 deficient, and Mfsd2a EC-specific inducible mice were developed to study the underlying molecular mechanisms. RESULTS: We demonstrated that inhibiting Wnt signaling by LGK974 could increase TMZ delivery and sensitize glioma to chemotherapy in both murine glioma models. Transcriptome analysis of ECs from murine gliomas revealed that Wnt signaling inhibition enhanced vascular transcytosis as indicated by the upregulation of PLVAP and downregulation of MFSD2A. Mfsd2a deficiency in mice enhances TMZ delivery in tumors, whereas constitutive expression of Mfsd2a in ECs suppresses the enhanced TMZ delivery induced by Wnt pathway inhibition in murine glioma. In addition, Wnt signaling inhibition enhanced caveolin-1 (Cav1)-positive caveolae-mediated transcytosis in tumor ECs. Moreover, Wnt signaling inhibitor or Mfsd2a deficiency fails to enhance TMZ penetration in tumors from Cav1-deficient mice. CONCLUSIONS: These results demonstrated that Wnt signaling regulates MFSD2A-dependent TMZ delivery through a caveolae-mediated EC transcytosis pathway. Our findings identify Wnt signaling as a promising therapeutic target to improve drug delivery for GBM treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Symporters , Mice , Animals , Wnt Signaling Pathway , Endothelial Cells/metabolism , Glioma/drug therapy , Glioma/metabolism , Temozolomide/therapeutic use , Glioblastoma/pathology , Brain Neoplasms/pathology , Transcytosis , Cell Line, Tumor , Drug Resistance, Neoplasm , Symporters/metabolism , Symporters/therapeutic use
11.
Int J Biol Macromol ; 224: 453-464, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36265539

ABSTRACT

Blocking the signaling activated by the plasma membrane receptor CD93 has recently been demonstrated a useful tool in antiangiogenic treatment and oncotherapy. In the proliferating endothelium, CD93 regulates cell adhesion, migration, and vascular maturation, yet it is unclear how CD93 interacts with the extracellular matrix activating signaling pathways involved in the vascular remodeling. Here for the first time we show that in endothelial cells CD93 is structured as a dimer and that this oligomeric form is physiologically instrumental for the binding of CD93 to its ligand Multimerin-2. Crystallographic X-ray analysis of recombinant CD93 reveals the crucial role played by the C-type lectin-like and sushi-like domains in arranging as an antiparallel dimer to achieve a functional binding state, providing key information for the future design of new drugs able to hamper CD93 function in neovascular pathologies.


Subject(s)
Endothelial Cells , Membrane Glycoproteins , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Lectins, C-Type/metabolism , Dimerization
12.
Nat Commun ; 13(1): 4755, 2022 08 13.
Article in English | MEDLINE | ID: mdl-35963857

ABSTRACT

Determining the levels of protein-protein interactions is essential for the analysis of signaling within the cell, characterization of mutation effects, protein function and activation in health and disease, among others. Herein, we describe MolBoolean - a method to detect interactions between endogenous proteins in various subcellular compartments, utilizing antibody-DNA conjugates for identification and signal amplification. In contrast to proximity ligation assays, MolBoolean simultaneously indicates the relative abundances of protein A and B not interacting with each other, as well as the pool of A and B proteins that are proximal enough to be considered an AB complex. MolBoolean is applicable both in fixed cells and tissue sections. The specific and quantifiable data that the method generates provide opportunities for both diagnostic use and medical research.


Subject(s)
Protein Interaction Mapping , Proteins , Protein Interaction Mapping/methods , Proteins/metabolism , Signal Transduction
15.
Cancer Immunol Immunother ; 71(8): 2029-2040, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35018481

ABSTRACT

Galectin-1 (Gal1) is a glycan-binding protein that promotes tumor progression by several distinct mechanisms. Through direct binding to vascular endothelial growth factor (VEGF)-receptor 2, Gal1 is able to induce VEGF-like signaling, which contributes to tumor angiogenesis. Furthermore, several studies have demonstrated an immunosuppressive function of Gal1 through effects on both effector and regulatory T cells. Elevated Gal1 expression and secretion have been shown in many tumor types, and high Gal1 serum levels have been connected to poor prognosis in cancer patients. These findings suggest that therapeutic strategies directed against Gal1 would enable simultaneous targeting of angiogenesis, immune evasion and metastasis. In the current study, we have analyzed the potential of Gal1 as a cancer vaccine target. We show that it is possible to generate high anti-Gal1 antibody levels in mice immunized with a recombinant vaccine protein consisting of bacterial sequences fused to Gal1. Growth of Gal1 expressing melanomas was significantly impaired in the immunized mice compared to the control group. This was associated with improved perfusion of the tumor vasculature, as well as increased infiltration of macrophages and cytotoxic T cells (CTLs). The level of granzyme B, mainly originating from CTLs in our model, was significantly elevated in Gal1 vaccinated mice and correlated with a decrease in tumor burden. We conclude that vaccination against Gal1 is a promising pro-immunogenic approach for cancer therapy that could potentially enhance the effect of other immunotherapeutic strategies due to its ability to promote CTL influx in tumors.


Subject(s)
Cancer Vaccines , Galectin 1 , Melanoma , Tumor Burden , Animals , Cancer Vaccines/immunology , Galectin 1/metabolism , Melanoma/therapy , Mice , Neovascularization, Pathologic , T-Lymphocytes, Cytotoxic/metabolism , Vaccination
16.
Neuro Oncol ; 24(3): 398-411, 2022 03 12.
Article in English | MEDLINE | ID: mdl-34347079

ABSTRACT

BACKGROUND: Tumor vessels in glioma are molecularly and functionally abnormal, contributing to treatment resistance. Proteins differentially expressed in glioma vessels can change vessel phenotype and be targeted for therapy. ELTD1 (Adgrl4) is an orphan member of the adhesion G-protein-coupled receptor family upregulated in glioma vessels and has been suggested as a potential therapeutic target. However, the role of ELTD1 in regulating vessel function in glioblastoma is poorly understood. METHODS: ELTD1 expression in human gliomas and its association with patient survival was determined using tissue microarrays and public databases. The role of ELTD1 in regulating tumor vessel phenotype was analyzed using orthotopic glioma models and ELTD1-/- mice. Endothelial cells isolated from murine gliomas were transcriptionally profiled to determine differentially expressed genes and pathways. The consequence of ELTD1 deletion on glioma immunity was determined by treating tumor-bearing mice with PD-1-blocking antibodies. RESULTS: ELTD1 levels were upregulated in human glioma vessels, increased with tumor malignancy, and were associated with poor patient survival. Progression of orthotopic gliomas was not affected by ELTD1 deletion, however, tumor vascular function was improved in ELTD1-/- mice. Bioinformatic analysis of differentially expressed genes indicated increased inflammatory response and decreased proliferation in tumor endothelium in ELTD1-/- mice. Consistent with an enhanced inflammatory response, ELTD1 deletion improved T-cell infiltration in GL261-bearing mice after PD-1 checkpoint blockade. CONCLUSION: Our data demonstrate that ELTD1 participates in inducing vascular dysfunction in glioma, and suggest that targeting of ELTD1 may normalize the vessels and improve the response to immunotherapy.


Subject(s)
Brain Neoplasms , Glioma , Receptors, G-Protein-Coupled/genetics , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Endothelial Cells/metabolism , Gene Deletion , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/metabolism
17.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830297

ABSTRACT

During angiogenesis, cell adhesion molecules expressed on the endothelial cell surface promote the growth and survival of newly forming vessels. Hence, elucidation of the signaling pathways activated by cell-to-matrix adhesion may assist in the discovery of new targets to be used in antiangiogenic therapy. In proliferating endothelial cells, the single-pass transmembrane glycoprotein CD93 has recently emerged as an important endothelial cell adhesion molecule regulating vascular maturation. In this study, we unveil a signaling pathway triggered by CD93 that regulates actin cytoskeletal dynamics responsible of endothelial cell adhesion. We show that the Src-dependent phosphorylation of CD93 and the adaptor protein Cbl leads to the recruitment of Crk, which works as a downstream integrator in the CD93-mediated signaling. Moreover, confocal microscopy analysis of FRET-based biosensors shows that CD93 drives the coordinated activation of Rac1 and RhoA at the cell edge of spreading cells, thus promoting the establishment of cell polarity and adhesion required for cell motility.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Signal Transduction/genetics , rhoA GTP-Binding Protein/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Polarity/genetics , Cells, Cultured , Humans , Membrane Glycoproteins/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-crk/metabolism , RNA Interference , Receptors, Complement/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
18.
Front Immunol ; 12: 724739, 2021.
Article in English | MEDLINE | ID: mdl-34539661

ABSTRACT

Glioblastoma is the most common and aggressive brain tumor, which is uniformly lethal due to its extreme invasiveness and the absence of curative therapies. Immune checkpoint inhibitors have not yet proven efficacious for glioblastoma patients, due in part to the low prevalence of tumor-reactive T cells within the tumor microenvironment. The priming of tumor antigen-directed T cells in the cervical lymph nodes is complicated by the shortage of dendritic cells and lack of appropriate lymphatic vessels within the brain parenchyma. However, recent data suggest that naive T cells may also be primed within brain tumor-associated tertiary lymphoid structures. Here, we review the current understanding of the formation of these structures within the central nervous system, and hypothesize that promotion of tertiary lymphoid structures could enhance priming of tumor antigen-targeted T cells and sensitize glioblastomas to cancer immunotherapy.


Subject(s)
Brain Neoplasms/immunology , Central Nervous System/immunology , Glioblastoma/immunology , T-Lymphocytes/immunology , Tertiary Lymphoid Structures/immunology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Central Nervous System/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Immunotherapy , Tertiary Lymphoid Structures/pathology , Tumor Microenvironment
19.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34228647

ABSTRACT

Passage of systemically delivered pharmacological agents into the brain is largely blocked by the blood-brain-barrier (BBB), an organotypic specialization of brain endothelial cells (ECs). Tumor vessels in glioblastoma (GBM), the most common malignant brain tumor in humans, are abnormally permeable, but this phenotype is heterogeneous and may differ between the tumor's center and invasive front. Here, through single-cell RNA sequencing (scRNA-seq) of freshly isolated ECs from human glioblastoma and paired tumor peripheral tissues, we have constructed a molecular atlas of human brain ECs providing unprecedented molecular insight into the heterogeneity of the human BBB and its molecular alteration in glioblastoma. We identified 5 distinct EC phenotypes representing different states of EC activation and BBB impairment, and associated with different anatomical locations within and around the tumor. This unique data resource provides key information for designing rational therapeutic regimens and optimizing drug delivery.


Subject(s)
Biological Transport/genetics , Blood-Brain Barrier , Brain Neoplasms , Carrier Proteins/genetics , Cell Membrane Permeability/genetics , Endothelial Cells , Glioblastoma , Biological Variation, Population , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Drug Delivery Systems/methods , Drug Discovery , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
20.
Nat Commun ; 12(1): 4127, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34226552

ABSTRACT

Gliomas are brain tumors characterized by an immunosuppressive microenvironment. Immunostimulatory agonistic CD40 antibodies (αCD40) are in clinical development for solid tumors, but are yet to be evaluated for glioma. Here, we demonstrate that systemic delivery of αCD40 in preclinical glioma models induces the formation of tertiary lymphoid structures (TLS) in proximity of meningeal tissue. In treatment-naïve glioma patients, the presence of TLS correlates with increased T cell infiltration. However, systemic delivery of αCD40 induces hypofunctional T cells and impairs the response to immune checkpoint inhibitors in pre-clinical glioma models. This is associated with a systemic induction of suppressive CD11b+ B cells post-αCD40 treatment, which accumulate in the tumor microenvironment. Our work unveils the pleiotropic effects of αCD40 therapy in glioma and reveals that immunotherapies can modulate TLS formation in the brain, opening up for future opportunities to regulate the immune response.


Subject(s)
CD40 Antigens/immunology , Glioma/drug therapy , Tertiary Lymphoid Structures/immunology , Animals , Antineoplastic Agents/pharmacology , B-Lymphocytes/immunology , Brain Neoplasms/drug therapy , CD11b Antigen , Cell Line, Tumor , Cytokines , Female , Gene Expression , Glioma/pathology , Humans , Immunoglobulin G/genetics , Immunotherapy , Male , Mice , Mice, Inbred C57BL , Myeloid Cells , Phenotype , T-Lymphocytes , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL