Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(9): e0239595, 2020.
Article in English | MEDLINE | ID: mdl-32970735

ABSTRACT

Immunotherapy checkpoint inhibitors, such as antibodies targeting PD-1 and CTLA-4, have demonstrated the potential of harnessing the immune system to treat cancer. However, despite encouraging results particularly with respect to survival, only a minority of patients benefit from these therapies. In clinical studies aimed at understanding changes in the immune system following immunotherapy treatment, ICOS (Inducible T cell CO-Stimulator) was shown to be significantly up-regulated on CD4+ T cells and this was associated with clinical activity, indicating that ICOS stimulatory activity may be beneficial in the treatment of solid tumors. In this report, we describe the generation of specific, species cross-reactive, agonist antibodies to ICOS, including the humanized clinical candidate, JTX-2011 (vopratelimab). Preclinical studies suggest that the ICOS stimulating antibodies require Fc receptor cross-linking for optimal agonistic activity. Notably, the ICOS antibodies do not exhibit superagonist properties but rather require T cell receptor (TCR)-mediated upregulation of ICOS for agonist activity. Treatment with the ICOS antibodies results in robust anti-tumor benefit and long-term protection in preclinical syngeneic mouse tumor models. Additional benefit is observed when the ICOS antibodies are administered in combination with anti-PD-1 and anti-CTLA-4 therapies. Based on the preclinical data, JTX-2011 is currently being developed in the clinical setting for the treatment of solid tumors.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cross-Priming , Immunotherapy/methods , Inducible T-Cell Co-Stimulator Protein/immunology , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Humans , Jurkat Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Receptors, Fc/immunology
2.
Cell Rep ; 10(9): 1487-1495, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25753415

ABSTRACT

Epidemiologic studies discovered an inverse association between immunoglobulin E (IgE)-mediated allergies and cancer, implying tumor-protective properties of IgE. However, the underlying immunologic mechanisms remain poorly understood. Antigen cross-presentation by dendritic cells (DCs) is of key importance for anti-tumor immunity because it induces the generation of cytotoxic CD8+ T lymphocytes (CTLs) with specificity for tumor antigens. We demonstrate that DCs use IgE and FcεRI, the high-affinity IgE receptor, for cross-presentation and priming of CTLs in response to free soluble antigen at low doses. Importantly, IgE/FcεRI-mediated cross-presentation is a distinct receptor-mediated pathway because it does not require MyD88 signals or IL-12 induction in DCs. Using passive immunization with tumor antigen-specific IgE and DC-based vaccination experiments, we demonstrate that IgE-mediated cross-presentation significantly improves anti-tumor immunity and induces memory responses in vivo. Our findings suggest a cellular mechanism for the tumor-protective features of IgE and expand the known physiological functions of this immunoglobulin.

3.
Nat Med ; 21(1): 71-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485910

ABSTRACT

Activating mutations in genes encoding G protein α (Gα) subunits occur in 4-5% of all human cancers, but oncogenic alterations in Gß subunits have not been defined. Here we demonstrate that recurrent mutations in the Gß proteins GNB1 and GNB2 confer cytokine-independent growth and activate canonical G protein signaling. Multiple mutations in GNB1 affect the protein interface that binds Gα subunits as well as downstream effectors and disrupt Gα interactions with the Gßγ dimer. Different mutations in Gß proteins clustered partly on the basis of lineage; for example, all 11 GNB1 K57 mutations were in myeloid neoplasms, and seven of eight GNB1 I80 mutations were in B cell neoplasms. Expression of patient-derived GNB1 variants in Cdkn2a-deficient mouse bone marrow followed by transplantation resulted in either myeloid or B cell malignancies. In vivo treatment with the dual PI3K-mTOR inhibitor BEZ235 suppressed GNB1-induced signaling and markedly increased survival. In several human tumors, mutations in the gene encoding GNB1 co-occurred with oncogenic kinase alterations, including the BCR-ABL fusion protein, the V617F substitution in JAK2 and the V600K substitution in BRAF. Coexpression of patient-derived GNB1 variants with these mutant kinases resulted in inhibitor resistance in each context. Thus, GNB1 and GNB2 alterations confer transformed and resistance phenotypes across a range of human tumors and may be targetable with inhibitors of G protein signaling.


Subject(s)
Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Proteins/genetics , Lymphoma, B-Cell/genetics , Animals , Cell Line, Tumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Janus Kinase 2/biosynthesis , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Mice , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics
4.
Cancer Immunol Res ; 2(7): 655-67, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24801837

ABSTRACT

Myeloid cells play important regulatory roles within the tumor environment by directly promoting tumor progression and modulating the function of tumor-infiltrating lymphocytes, and as such, they represent a potential therapeutic target for the treatment of cancer. Although distinct subsets of tumor-associated myeloid cells have been identified, a broader analysis of the complete myeloid cell landscape within individual tumors and also across different tumor types has been lacking. By establishing the developmental and transcriptomic signatures of infiltrating myeloid cells from multiple primary tumors, we found that tumor-associated macrophages (TAM) and tumor-associated neutrophils (TAN), while present within all tumors analyzed, exhibited strikingly different frequencies, gene expression profiles, and functions across cancer types. We also evaluated the impact of anatomic location and circulating factors on the myeloid cell composition of tumors. The makeup of the myeloid compartment was determined by the tumor microenvironment rather than the anatomic location of tumor development or tumor-derived circulating factors. Protumorigenic and hypoxia-associated genes were enriched in TAMs and TANs compared with splenic myeloid-derived suppressor cells. Although all TANs had an altered expression pattern of secretory effector molecules, in each tumor type they exhibited a unique cytokine, chemokine, and associated receptor expression profile. One such molecule, haptoglobin, was uniquely expressed by 4T1 TANs and identified as a possible diagnostic biomarker for tumors characterized by the accumulation of myeloid cells. Thus, we have identified considerable cancer-specific diversity in the lineage, gene expression, and function of tumor-infiltrating myeloid cells.


Subject(s)
Myeloid Cells/immunology , Tumor Microenvironment/immunology , Animals , Biomarkers, Tumor/metabolism , Cell Movement/immunology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/immunology , Haptoglobins/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Neutrophils/immunology , Tumor Cells, Cultured , Tumor Microenvironment/genetics
5.
Nature ; 506(7486): 52-7, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24476824

ABSTRACT

Recent clinical trials showed that targeting of inhibitory receptors on T cells induces durable responses in a subset of cancer patients, despite advanced disease. However, the regulatory switches controlling T-cell function in immunosuppressive tumours are not well understood. Here we show that such inhibitory mechanisms can be systematically discovered in the tumour microenvironment. We devised an in vivo pooled short hairpin RNA (shRNA) screen in which shRNAs targeting negative regulators became highly enriched in murine tumours by releasing a block on T-cell proliferation upon tumour antigen recognition. Such shRNAs were identified by deep sequencing of the shRNA cassette from T cells infiltrating tumour or control tissues. One of the target genes was Ppp2r2d, a regulatory subunit of the PP2A phosphatase family. In tumours, Ppp2r2d knockdown inhibited T-cell apoptosis and enhanced T-cell proliferation as well as cytokine production. Key regulators of immune function can therefore be discovered in relevant tissue microenvironments.


Subject(s)
Immunotherapy , Molecular Targeted Therapy , Protein Phosphatase 2/metabolism , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Female , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Protein Phosphatase 2/deficiency , Protein Phosphatase 2/genetics , RNA, Small Interfering/genetics , Reproducibility of Results
6.
Nat Immunol ; 13(11): 1118-28, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023392

ABSTRACT

We assessed gene expression in tissue macrophages from various mouse organs. The diversity in gene expression among different populations of macrophages was considerable. Only a few hundred mRNA transcripts were selectively expressed by macrophages rather than dendritic cells, and many of these were not present in all macrophages. Nonetheless, well-characterized surface markers, including MerTK and FcγR1 (CD64), along with a cluster of previously unidentified transcripts, were distinctly and universally associated with mature tissue macrophages. TCEF3, C/EBP-α, Bach1 and CREG-1 were among the transcriptional regulators predicted to regulate these core macrophage-associated genes. The mRNA encoding other transcription factors, such as Gata6, was associated with single macrophage populations. We further identified how these transcripts and the proteins they encode facilitated distinguishing macrophages from dendritic cells.


Subject(s)
Antigens, CD/genetics , Macrophages/metabolism , RNA, Messenger/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Antigens, CD/immunology , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Variation , Liver/cytology , Liver/immunology , Liver/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Oligonucleotide Array Sequence Analysis , Organ Specificity , RNA, Messenger/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transcription Factors/immunology
7.
Immunity ; 37(2): 276-89, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22884313

ABSTRACT

To initiate adaptive immunity, dendritic cells (DCs) move from parenchymal tissues to lymphoid organs by migrating along stromal scaffolds that display the glycoprotein podoplanin (PDPN). PDPN is expressed by lymphatic endothelial and fibroblastic reticular cells and promotes blood-lymph separation during development by activating the C-type lectin receptor, CLEC-2, on platelets. Here, we describe a role for CLEC-2 in the morphodynamic behavior and motility of DCs. CLEC-2 deficiency in DCs impaired their entry into lymphatics and trafficking to and within lymph nodes, thereby reducing T cell priming. CLEC-2 engagement of PDPN was necessary for DCs to spread and migrate along stromal surfaces and sufficient to induce membrane protrusions. CLEC-2 activation triggered cell spreading via downregulation of RhoA activity and myosin light-chain phosphorylation and triggered F-actin-rich protrusions via Vav signaling and Rac1 activation. Thus, activation of CLEC-2 by PDPN rearranges the actin cytoskeleton in DCs to promote efficient motility along stromal surfaces.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Actins/metabolism , Adaptive Immunity/physiology , Animals , Antigen-Presenting Cells/metabolism , Blood Platelets/metabolism , Cells, Cultured , Dendritic Cells/immunology , Embryo, Mammalian , Endothelial Cells/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Myosin Light Chains/metabolism , Platelet Activation , Pregnancy , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction/physiology , Skin/cytology , Skin/metabolism , Tissue Culture Techniques , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Nat Immunol ; 13(9): 888-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22797772

ABSTRACT

Although much progress has been made in the understanding of the ontogeny and function of dendritic cells (DCs), the transcriptional regulation of the lineage commitment and functional specialization of DCs in vivo remains poorly understood. We made a comprehensive comparative analysis of CD8(+), CD103(+), CD11b(+) and plasmacytoid DC subsets, as well as macrophage DC precursors and common DC precursors, across the entire immune system. Here we characterized candidate transcriptional activators involved in the commitment of myeloid progenitor cells to the DC lineage and predicted regulators of DC functional diversity in tissues. We identified a molecular signature that distinguished tissue DCs from macrophages. We also identified a transcriptional program expressed specifically during the steady-state migration of tissue DCs to the draining lymph nodes that may control tolerance to self tissue antigens.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Dendritic Cells/immunology , Transcription, Genetic , Cell Differentiation/genetics , Dendritic Cells/cytology , Gene Expression Profiling , Humans
9.
Nat Immunol ; 13(5): 499-510, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22466668

ABSTRACT

Lymph node stromal cells (LNSCs) closely regulate immunity and self-tolerance, yet key aspects of their biology remain poorly elucidated. Here, comparative transcriptomic analyses of mouse LNSC subsets demonstrated the expression of important immune mediators, growth factors and previously unknown structural components. Pairwise analyses of ligands and cognate receptors across hematopoietic and stromal subsets suggested a complex web of crosstalk. Fibroblastic reticular cells (FRCs) showed enrichment for higher expression of genes relevant to cytokine signaling, relative to their expression in skin and thymic fibroblasts. LNSCs from inflamed lymph nodes upregulated expression of genes encoding chemokines and molecules involved in the acute-phase response and the antigen-processing and antigen-presentation machinery. Poorly studied podoplanin (gp38)-negative CD31(-) LNSCs showed similarities to FRCs but lacked expression of interleukin 7 (IL-7) and were identified as myofibroblastic pericytes that expressed integrin α(7). Together our data comprehensively describe the transcriptional characteristics of LNSC subsets.


Subject(s)
Gene Expression/immunology , Inflammation/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , Transcriptome , Acute-Phase Reaction/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Cytokines/immunology , Cytokines/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Homeostasis/immunology , Inflammation/genetics , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Lymph Nodes/cytology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Pericytes/immunology , Pericytes/metabolism , Self Tolerance/immunology , Tissue Array Analysis/methods
10.
Cancer Discov ; 1(2): 158-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21984975

ABSTRACT

Initiation of pancreatic ductal adenocarcinoma (PDAC) is driven by oncogenic KRAS mutation, and disease progression is associated with frequent loss of tumor suppressors. In this study, human PDAC genome analyses revealed frequent deletion of the PTEN gene as well as loss of expression in primary tumor specimens. A potential role for PTEN as a haploinsufficient tumor suppressor is further supported by mouse genetic studies. The mouse PDAC driven by oncogenic Kras mutation and Pten deficiency also sustains spontaneous extinction of Ink4a expression and shows prometastatic capacity. Unbiased transcriptomic analyses established that combined oncogenic Kras and Pten loss promotes marked NF-κB activation and its cytokine network, with accompanying robust stromal activation and immune cell infiltration with known tumor-promoting properties. Thus, PTEN/phosphoinositide 3-kinase (PI3K) pathway alteration is a common event in PDAC development and functions in part to strongly activate the NF-κB network, which may serve to shape the PDAC tumor microenvironment.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Cytokines/genetics , NF-kappa B/genetics , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Animals, Genetically Modified , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytokines/metabolism , Genes, Tumor Suppressor , Humans , Mice , Mice, Inbred C57BL , Mutation , NF-kappa B/metabolism , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
11.
Nat Immunol ; 12(11): 1096-104, 2011 Sep 18.
Article in English | MEDLINE | ID: mdl-21926986

ABSTRACT

Fibroblastic reticular cells (FRCs) and lymphatic endothelial cells (LECs) are nonhematopoietic stromal cells of lymphoid organs. They influence the migration and homeostasis of naive T cells; however, their influence on activated T cells remains undescribed. Here we report that FRCs and LECs inhibited T cell proliferation through a tightly regulated mechanism dependent on nitric oxide synthase 2 (NOS2). Expression of NOS2 and production of nitric oxide paralleled the activation of T cells and required a tripartite synergism of interferon-γ, tumor necrosis factor and direct contact with activated T cells. Notably, in vivo expression of NOS2 by FRCs and LECs regulated the size of the activated T cell pool. Our study elucidates an as-yet-unrecognized role for the lymph node stromal niche in controlling T cell responses.


Subject(s)
Clonal Selection, Antigen-Mediated , Endothelium, Lymphatic/metabolism , Nitric Oxide Synthase Type II/metabolism , Stromal Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Growth Processes/genetics , Cell Movement/genetics , Cells, Cultured , Endothelium, Lymphatic/immunology , Endothelium, Lymphatic/pathology , Intercellular Junctions/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Stromal Cells/immunology , Stromal Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transgenes/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 6(8): e23921, 2011.
Article in English | MEDLINE | ID: mdl-21886840

ABSTRACT

Lymphoid organ-resident DC subsets are thought to play unique roles in determining the fate of T cell responses. Recent studies focusing on a single lymphoid organ identified molecular pathways that are differentially operative in each DC subset and led to the assumption that a given DC subset would more or less exhibit the same genomic and functional profiles throughout the body. Whether the local milieu in different anatomical sites can also influence the transcriptome of DC subsets has remained largely unexplored. Here, we interrogated the transcriptional relationships between lymphoid organ-resident DC subsets from spleen, gut- and skin-draining lymph nodes, and thymus of C57BL/6 mice. For this purpose, major resident DC subsets including CD4 and CD8 DCs were sorted at high purity and gene expression profiles were compared using microarray analysis. This investigation revealed that lymphoid organ-resident DC subsets exhibit divergent genomic programs across lymphoid organs. Interestingly, we also found that transcriptional and biochemical properties of a given DC subset can differ between lymphoid organs for lymphoid organ-resident DC subsets, but not plasmacytoid DCs, suggesting that determinants of the tissue milieu program resident DCs for essential site-specific functions.


Subject(s)
Dendritic Cells/metabolism , Lymph Nodes/cytology , Transcriptome , Animals , Intestines , Mice , Mice, Inbred C57BL , Microarray Analysis , Skin , Spleen , Thymus Gland , Tissue Distribution
13.
Blood ; 118(2): 464-72, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21536858

ABSTRACT

Tetraspanin protein CD151 on tumor cells supports invasion and metastasis. In the present study, we show that host animal CD151 also plays a critical role. CD151-null mice showed markedly diminished experimental lung metastasis after injection of Lewis lung carcinoma or B16F10 melanoma cells. Diminished tumor cell residence in the lungs was evident 6-24 hours after injection. Consistent with an endothelial cell deficiency, isolated CD151-null mouse lung endothelial cells showed diminished support for B16F10 adhesion and transendothelial migration, diminished B16F10-induced permeability, and diminished B16F10 adhesion to extracellular matrix deposited by CD151-null mouse lung endothelial cells. However, CD151 deletion did not affect the size of metastatic foci or subcutaneous primary B16F10 tumors, tumor aggregation, tumor clearance from the blood, or tumor-induced immune cell activation and recruitment. Therefore, the effects of host CD151 on metastasis do not involve altered local tumor growth or immune surveillance. VEGF-induced endothelial cell signaling through Src and Akt was diminished in CD151-null endothelial cells. However, deficient signaling was not accompanied by reduced endothelial permeability either in vitro (monolayer permeability assay) or in vivo (VEGF-stimulated Miles assay). In summary, diminished metastasis in CD151-null host animals may be due to impaired tumor-endothelial interactions, with underlying defects in mouse lung endothelial cell extracellular matrix production.


Subject(s)
Antigens, CD/genetics , Neoplasm Metastasis/genetics , Animals , Antigens, CD/physiology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Tetraspanin 24 , Transcellular Cell Migration/genetics , Transendothelial and Transepithelial Migration/genetics
14.
Nat Rev Immunol ; 10(12): 813-25, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21088682

ABSTRACT

T cells encounter their cognate antigens in specialized compartments of secondary lymphoid organs (SLOs). There, dendritic cells (DCs) present self and non-self antigens to T cells, and promote immunity or tolerance depending on the availability of danger signals. Resident stromal cells orchestrate the interaction between T cells and DCs by recruiting them to T cell zones and guiding their migration within SLOs. Recent studies have shown that SLO-resident stromal cells also have a crucial role in tolerance induction in the periphery. In this Review, we discuss the roles of SLO-resident DCs and stromal cells in shaping T cell responses.


Subject(s)
Antigen-Presenting Cells/immunology , Hematopoietic System/immunology , Lymphoid Tissue/immunology , Animals , Antigens/immunology , Cell Movement/immunology , Humans , Immune Tolerance , Mice , Stromal Cells/immunology , T-Lymphocytes/immunology
15.
Proc Natl Acad Sci U S A ; 107(50): 21647-52, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098276

ABSTRACT

Cytotoxic lymphocytes such as natural killer (NK) and CD8 T cells play important roles in immunosurveillance by killing virally infected or malignant cells. The homeostatic cytokine, IL-15, promotes the development, function, and survival of NK and CD8 T cells. IL-15 is normally presented in trans as a surface complex with IL-15 receptor-alpha-chain (IL-15Rα) by dendritic cells (DCs) and monocytes. Signaling by IL-15 occurs via the IL-2/IL-15 receptor ß-chain (CD122) which is expressed primarily by NK1.1(+) cells and CD8 T cells. The use of preformed complexes of IL-15 with soluble IL-15Rα complexes to boost the effector function of CD122(+) cytolytic lymphocytes such as NK and CD8 T cells has recently gained considerable attention. Here we describe the impact of transient and prolonged in vivo stimulation by IL-15/IL-15Rα complexes on NK and CD8 T cells. Whereas transitory stimulation increased the number of activated NK cells and significantly enhanced their effector function, prolonged stimulation by IL-15/IL-15Rα complexes led to a marked accumulation of mature NK cells with considerably impaired activation, cytotoxicity, and proliferative activity, and an altered balance of activating and inhibitory receptors. In contrast to NK cells, CD8 T cells exhibited an activated phenotype and robust T cell receptor stimulation and effector function upon chronic stimulation with IL-15/IL-15Rα complexes. Thus, prolonged stimulation with the strong activating signal leads to a preferential accrual of mature NK cells with altered activation and diminished functional capacity. These findings point to a negative feedback mechanism to preferentially counterbalance excessive NK cell activity and may have important implications for cytokine immunotherapy.


Subject(s)
Interleukin-15 Receptor alpha Subunit/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Animals , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology
16.
Cancer Res ; 70(10): 3945-54, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20406989

ABSTRACT

Therapeutic subunit vaccines based on tumor-associated antigens (TAA) represent an attractive approach for the treatment of cancer. However, poor immunogenicity of TAAs requires potent adjuvants for therapeutic efficacy. We recently proposed the tumor necrosis factor family costimulatory ligands as potential adjuvants for therapeutic vaccines and, hence, generated a soluble form of 4-1BBL chimeric with streptavidin (SA-4-1BBL) that has pleiotropic effects on cells of innate, adaptive, and regulatory immunity. We herein tested whether these effects can translate into effective cancer immunotherapy when SA-4-1BBL was also used as a vehicle to deliver TAAs in vivo to dendritic cells (DCs) constitutively expressing the 4-1BB receptor. SA-4-1BBL was internalized by DCs upon receptor binding and immunization with biotinylated antigens conjugated to SA-4-1BBL resulted in increased antigen uptake and cross-presentation by DCs, leading to the generation of effective T-cell immune responses. Conjugate vaccines containing human papillomavirus 16 E7 oncoprotein or survivin as a self-TAA had potent therapeutic efficacy against TC-1 cervical and 3LL lung carcinoma tumors, respectively. Therapeutic efficacy of the vaccines was associated with increased CD4(+) T and CD8(+) T-cell effector and memory responses and higher intratumoral CD8(+) T effector/CD4(+)CD25(+)Foxp3(+) T regulatory cell ratio. Thus, potent pleiotropic immune functions of SA-4-1BBL combined with its ability to serve as a vehicle to increase the delivery of antigens to DCs in vivo endow this molecule with the potential to serve as an effective immunomodulatory component of therapeutic vaccines against cancer and chronic infections.


Subject(s)
4-1BB Ligand/administration & dosage , Cancer Vaccines/administration & dosage , Drug Delivery Systems , Immunologic Factors/administration & dosage , Lung Neoplasms/therapy , Uterine Cervical Neoplasms/therapy , Animals , Cross-Priming/immunology , Dendritic Cells/immunology , Female , Humans , Inhibitor of Apoptosis Proteins , Lung Neoplasms/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins , Survival Rate , Survivin , T-Lymphocytes/immunology , Tumor Cells, Cultured , Uterine Cervical Neoplasms/immunology
17.
Vaccine ; 28(2): 512-22, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19836479

ABSTRACT

Agonistic Abs to select costimulatory members of CD28 and TNFR family have shown efficacy in various preclinical cancer immunotherapeutic settings. However, the use of agonistic Abs is often associated with severe toxicity due to non-specific activation of lymphocytes. We hypothesized that natural costimulatory ligands may serve as more potent and safer alternative to agonistic Abs for immunotherapy. In this communication, we focused on 4-1BBL as the molecule of choice because of the pleiotropic effects of 4-1BB signaling in the immune system and the demonstrated therapeutic efficacy of 4-1BB agonistic Abs in preclinical cancer and infection models. We report that a novel form of soluble ligand, SA-4-1BBL, delivered more potent and qualitatively different signals to T cells than an agonistic Ab. Importantly, while treatment of naïve mice with the agonistic Ab resulted in severe toxicity, as assessed by enlarged spleen and peripheral LNs, non-specific T cell proliferation, hepatitis, and systemic inflammatory cytokine production, treatment with SA-4-1BBL lacked these immune anomalies. Agonistic Ab treatment produced full toxicity in FcgammaR(-/-) or complement C1q(-/-) or C3(-/-) knockout mice, suggesting lack of involvement of stimulatory FcgammaRs or complement system in the observed toxicity. Naïve and memory T cells served as direct targets of anti-4-1BB Ab-mediated toxicity. Potent immunostimulatory activity combined with lack of toxicity rationalizes further development of soluble SA-4-1BBL as an immunomodulatory component of therapeutic vaccines against cancer and chronic infections.


Subject(s)
4-1BB Ligand/metabolism , Antibodies/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , 4-1BB Ligand/adverse effects , Adjuvants, Immunologic/adverse effects , Animals , Antibodies/adverse effects , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , T-Lymphocytes/immunology
18.
Cancer Res ; 69(10): 4319-26, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435920

ABSTRACT

Vaccines represent an attractive treatment modality for the management of cancer primarily because of their specificity and generation of immunologic memory important for controlling recurrences. However, the efficacy of therapeutic vaccines may require formulations that not only generate effective immune responses but also overcome immune evasion mechanisms employed by progressing tumor. Costimulatory molecules play critical roles in modulating innate, adaptive, and regulatory immunity and have potential to serve as effective immunomodulatory components of therapeutic vaccines. In this study, we tested the function of a novel soluble form of 4-1BB ligand (4-1BBL) costimulatory molecule in modulating innate, adaptive, and regulatory immunity and assessed its therapeutic efficacy in the HPV-16 E7-expressing TC-1 cervical cancer and survivin-expressing 3LL lung carcinoma mouse models. Vaccination with 4-1BBL activated dendritic cells and enhanced antigen uptake, generated CD8(+) T-cell effector/memory responses, and endowed T effector cells refractory to suppression by CD4(+)CD25(+)FoxP3(+) T regulatory cells. Immunization with 4-1BBL in combination with an E7 peptide or survivin protein resulted in eradication of TC-1 and 3LL tumors, respectively. 4-1BBL was more effective than TLR agonists LPS, MPL, and CpG and an agonistic 4-1BB antibody as a component of E7 peptide-based therapeutic vaccine for the generation of immune responses and eradication of TC-1 established tumors in the absence of detectable toxicity. Therapeutic efficacy was associated with reversal of tumor-mediated nonresponsiveness/anergy as well as establishment of long-term CD8(+) T-cell memory. Potent pleiotropic immunomodulatory activities combined with lack of toxicity highlight the potential of 4-1BBL molecule as an effective component of therapeutic cancer vaccines.


Subject(s)
4-1BB Ligand/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Animals , Female , Humans , Immunologic Memory/drug effects , Inhibitor of Apoptosis Proteins , Lung Neoplasms/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/therapeutic use , Repressor Proteins , Survivin , Uterine Cervical Neoplasms/immunology
19.
J Immunol ; 182(4): 2102-12, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201863

ABSTRACT

The B7 family member programmed death-1 ligand (PD-L1) has been shown to play an inhibitory role in the regulation of T cell responses in several organs. However, the role of PD-L1 in regulating tolerance to self-Ags of the small intestine has not been previously addressed. In this study, we investigated the role of PD-L1 in CD8(+) T cell tolerance to an intestinal epithelium-specific Ag using the iFABP-tOVA transgenic mouse model, in which OVA is expressed as a self-Ag throughout the small intestine. Using adoptive transfer of naive OVA-specific CD8(+) T cells, we show that loss of PD-1:PD-L1 signaling, by either Ab-mediated PD-L1 blockade or transfer of PD-1(-/-) T cells, leads to considerable expansion of OVA-specific CD8(+) T cells and their differentiation into effector cells capable of producing proinflammatory cytokines. A fatal CD8(+) T cell-mediated inflammatory response develops rapidly against the small bowel causing destruction of the epithelial barrier, severe blunting of intestinal villi, and recruitment and activation of myeloid cells. This response is highly specific because immune destruction selectively targets the small intestine but not other organs. Collectively, these results indicate that loss of the PD-1:PD-L1 inhibitory pathway breaks CD8(+) T cell tolerance to intestinal self-Ag, thus leading to severe enteric autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , B7-1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Enteritis/immunology , Immune Tolerance/immunology , Immunity, Mucosal/immunology , Membrane Glycoproteins/immunology , Peptides/immunology , Adoptive Transfer , Animals , Antigens, Surface/immunology , Apoptosis Regulatory Proteins/immunology , Autoantigens/immunology , B7-1 Antigen/metabolism , B7-H1 Antigen , Cell Differentiation/immunology , Flow Cytometry , Intestines/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Ovalbumin/immunology , Peptides/metabolism , Programmed Cell Death 1 Receptor
20.
Cancer Res ; 68(8): 2972-83, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413767

ABSTRACT

Tumors often escape immune-mediated destruction by suppressing lymphocyte infiltration or effector function. New approaches are needed that overcome this suppression and thereby augment the tumoricidal capacity of tumor-reactive lymphocytes. The cytokine interleukin-15 (IL-15) promotes proliferation and effector capacity of CD8(+) T cells, natural killer (NK) cells, and NKT cells; however, it has a short half-life and high doses are needed to achieve functional responses in vivo. The biological activity of IL-15 can be dramatically increased by complexing this cytokine to its soluble receptor, IL-15R alpha. Here, we report that in vivo delivery of IL-15/IL-15R alpha complexes triggers rapid and significant regression of established solid tumors in two murine models. Despite a marked expansion of IL-2/IL-15R beta(+) cells in lymphoid organs and peripheral blood following treatment with IL-15/IL-15R alpha complexes, the destruction of solid tumors was orchestrated by tumor-resident rather than newly infiltrating CD8(+) T cells. Our data provide novel insights into the use of IL-15/IL-15R alpha complexes to relieve tumor-resident T cells from functional suppression by the tumor microenvironment and have significant implications for cancer immunotherapy and treatment of chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Neoplasms/immunology , Receptors, Interleukin-15/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Insulinoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/pathology , Pancreatic Neoplasms/immunology , Receptors, Interleukin-15/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...