Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
NPJ Precis Oncol ; 7(1): 69, 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37468567

ABSTRACT

The objective was to correlate CD47 gene expression with resistance to immune checkpoint inhibitors (ICI) in tumor tissue of gynecological cancer (GC). Further, we sought to assess the efficacy of targeting CD47 pathway alone and in combination in pre-clinical ovarian cancer (OC) models. We performed transcriptomic analyses in GC treated with ICI. Signaling pathway enrichment analysis was performed using Ingenuity Pathway Analysis. Immune cell abundance was estimated. CD47 expression was correlated with other pathways, objective response, and progression-free survival (PFS). Anti-tumor efficacy of anti-CD47 therapy alone and in combination was investigated both in-vitro and in-vivo using cell-line derived xenograft (CDX) and patient-derived xenograft (PDX) models. High CD47 expression associated with lower response to ICI and trended toward lower PFS in GC patients. Higher CD47 associated negatively with PDL1 and CTLA4 expression, as well as cytotoxic T-cells and dendritic cells but positively with TGF-ß, BRD4 and CXCR4/CXCL12 expression. Anti-CD47 significantly enhanced macrophage-mediated phagocytosis of OC cells in-vitro and exhibited potent anti-tumor activity in-vivo in OC CDX and PDX models. In-vitro treatment with PARPi increased CD47 expression. Anti-CD47 led to significantly enhanced in-vitro phagocytosis, enhanced STING pathway and synergized in-vivo when combined with PARP inhibitors in BRCA-deficient OC models. This study provides insight on the potential role of CD47 in mediating immunotherapy resistance and its association with higher TGF-ß, BRD4 and CXCR4/CXCL12 expression. Anti-CD47 showed potent anti-tumor activity and synergized with PARPi in OC models. These data support clinical development of anti-CD47 therapy with PARPi in OC.

2.
Cancers (Basel) ; 13(23)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34885092

ABSTRACT

Cancer progression in mycosis fungoides, the most common form of cutaneous T-cell lymphoma, occurs in a predictable, sequential pattern that starts from patches and that evolves to plaques and later to tumors. Therefore, unlocking the relationship between the microarchitecture of mycosis fungoides and the clinical counterparts of that microstructure represents important steps for the design of targeted therapies. Using multispectral fluorescent imaging, we show that the progression of mycosis fungoides from plaque to tumor parallels the cutaneous expansion of the malignant CD4+ T cells that express TOX. The density of exhausted BTLA+ CD4+ T cells around malignant CD4+TOX+ cells was higher in tumors than it was in plaques, suggesting that undesired safeguards are in place within the tumor microenvironment that prevent immune activation and subsequent cancer eradication. Overriding the CD47 checkpoint with an intralesional SIRPαFc fusion decoy receptor induced the resolution of mycosis fungoides in patients that paralleled an amplified expansion of NK and CD8+ T cells in addition to a reduction of the exhausted BTLA+ CD4+ T cells that were engaged in promiscuous intercellular interactions. These therapeutic benefits of the CD47 blockade were further unleashed by adjuvant interferon-α, which stimulates cytotoxic cells, underscoring the importance of an inflamed microenvironment in facilitating the response to immunotherapy. Collectively, these findings support CD47 as a therapeutic target in treating mycosis fungoides and demonstrate a synergistic role of interferon-α in exploiting these clinical benefits.

3.
Lancet Haematol ; 8(11): e808-e817, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34627593

ABSTRACT

BACKGROUND: Intravenous TTI-621 (SIRPα-IgG1 Fc) was previously shown to have activity in relapsed or refractory haematological malignancies. This phase 1 study evaluated the safety and activity of TTI-621 in patients with percutaneously accessible relapsed or refractory mycosis fungoides, Sézary syndrome, or solid tumours. Here we report the clinical and translational results among patients with mycosis fungoides or Sézary syndrome. METHODS: This multicentre, open-label, phase 1 study was conducted at five academic health-care and research centres in the USA. Eligible patients were aged 18 years or older; had injectable, histologically or cytologically confirmed relapsed or refractory cutaneous T-cell lymphoma (CTCL) or solid tumours; Eastern Cooperative Oncology Group performance status of 2 or less; and adequate haematological, renal, hepatic, and cardiac function. TTI-621 was injected intralesionally in a sequential dose escalation (cohorts 1-5; single 1 mg, 3 mg, or 10 mg injection or three 10 mg injections weekly for 1 or 2 weeks) and in expansion cohorts (cohorts 6-9; 2 week induction at the maximum tolerated dose; weekly continuation was allowed). In cohort 6, patients were injected with TTI-621 in a single lesion and in cohort 7, they were injected in multiple lesions. In cohort 8, TTI-621 was combined with pembrolizumab 200 mg injections per product labels. In cohort 9, TTI-621 was combined with the standard labelled dose of subcutaneous pegylated interferon alpha-2a 90 µg. The primary endpoint was the incidence and severity of adverse events. The study is registered with ClinicalTrials.gov, NCT02890368, and was closed by the sponsor to focus on intravenous studies with TTI-621. FINDINGS: Between Jan 30, 2017, and March 31, 2020, 66 patients with mycosis fungoides, Sézary syndrome, other CTCL, or solid tumours were screened, 35 of whom with mycosis fungoides or Sézary syndrome were enrolled and received intralesional TTI-621 (escalation, n=13; expansion, n=22). No dose-limiting toxicities occurred; the maximum tolerated dose was not established. In the dose expansion cohorts, the maximally assessed regimen (10 mg thrice weekly for 2 weeks) was used. 25 (71%) patients had treatment-related adverse events; the most common (occurring in ≥10% of patients) were chills (in ten [29%] patients), injection site pain (nine [26%]), and fatigue (eight [23%]). No treatment-related adverse events were grade 3 or more or serious. There were no treatment-related deaths. Rapid responses (median 45 days, IQR 17-66) occurred independently of disease stage or injection frequency. 26 (90%) of 29 evaluable patients had decreased Composite Assessment of Index Lesion Severity (CAILS) scores; ten (34%) had a decrease in CAILS score of 50% or more (CAILS response). CAILS score reductions occurred in adjacent non-injected lesions in eight (80%) of ten patients with paired assessments and in distal non-injected lesions in one additional patient. INTERPRETATION: Intralesional TTI-621 was well tolerated and had activity in adjacent or distal non-injected lesions in patients with relapsed or refractory mycosis fungoides or Sézary syndrome, suggesting it has systemic and locoregional abscopal effects and potential as an immunotherapy for these conditions. FUNDING: Trillium Therapeutics.


Subject(s)
CD47 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Immunoglobulin G/therapeutic use , Mycosis Fungoides/drug therapy , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Aged , CD47 Antigen/immunology , Female , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immunoglobulin G/administration & dosage , Immunoglobulin G/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Mycosis Fungoides/immunology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Sezary Syndrome/immunology , Skin Neoplasms/immunology
4.
Science ; 363(6427): 639-644, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30733420

ABSTRACT

Although widely studied as a neurotransmitter, T cell-derived acetylcholine (ACh) has recently been reported to play an important role in regulating immunity. However, the role of lymphocyte-derived ACh in viral infection is unknown. Here, we show that the enzyme choline acetyltransferase (ChAT), which catalyzes the rate-limiting step of ACh production, is robustly induced in both CD4+ and CD8+ T cells during lymphocytic choriomeningitis virus (LCMV) infection in an IL-21-dependent manner. Deletion of Chat within the T cell compartment in mice ablated vasodilation in response to infection, impaired the migration of antiviral T cells into infected tissues, and ultimately compromised the control of chronic LCMV clone 13 infection. Our results reveal a genetic proof of function for ChAT in T cells during viral infection and identify a pathway of T cell migration that sustains antiviral immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Choline O-Acetyltransferase/immunology , Interleukins/immunology , Lymphocytic Choriomeningitis/immunology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/enzymology , Cell Movement , Choline O-Acetyltransferase/genetics , Female , Lymphocyte Activation , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vasodilation
5.
PLoS One ; 12(10): e0187262, 2017.
Article in English | MEDLINE | ID: mdl-29084248

ABSTRACT

Tumor-associated macrophages (TAMs) are heterogeneous and can adopt a spectrum of activation states between pro-inflammatory and pro-tumorigenic in response to the microenvironment. We have previously shown that TTI-621, a soluble SIRPαFc fusion protein that blocks the CD47 "do-not-eat" signal, promotes tumor cell phagocytosis by IFN-γ-primed macrophages. To assess the impact of CD47 blockade on diverse types of macrophages that are found within the tumor microenvironment, six different polarized human macrophage subsets (M(-), M(IFN-γ), M(IFN-γ+LPS), M(IL-4), M(HAGG+IL-1ß), M(IL-10 + TGFß)) with distinct cell surface markers and cytokine profiles were generated. Blockade of CD47 using TTI-621 significantly increased phagocytosis of lymphoma cells by all macrophage subsets, with M(IFN-γ), M(IFN-γ+LPS) and M(IL-10 + TGFß) macrophages having the highest phagocytic response. TTI-621-mediated phagocytosis involves macrophage expression of both the low- and high-affinity Fcγ receptors II (CD32) and I (CD64), respectively. Moreover, macrophages with lower phagocytic capabilities (M(-), M(IL-4), M(HAGG+IL-1ß)) could readily be re-polarized into highly phagocytic macrophages using various cytokines or TLR agonists. In line with the in vitro study, we further demonstrate that TTI-621 can trigger phagocytosis of tumor cells by diverse subsets of isolated mouse TAMs ex vivo. These data suggest that TTI-621 may be efficacious in triggering the destruction of cancer cells by a diverse population of TAMs found in vivo and support possible combination approaches to augment the activity of CD47 blockade.


Subject(s)
Antigens, Differentiation/physiology , Cell Polarity , Immunoglobulin Fc Fragments/physiology , Lymphoma, Large B-Cell, Diffuse/immunology , Macrophages/immunology , Phagocytosis/physiology , Receptors, Immunologic/physiology , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Female , Humans , Immunophenotyping , Mice , Mice, Hairless
6.
J Immunol Methods ; 450: 81-89, 2017 11.
Article in English | MEDLINE | ID: mdl-28789924

ABSTRACT

4-1BB is a TNFR family member associated with NF-κB mediated survival signaling. 4-1BB is widely expressed on activated cells of the immune system, including activated T cells, NK cells and dendritic cells. Its ligand, 4-1BBL, is transiently expressed on activated antigen presenting cells and at low levels on activated T cells. Although 4-1BBL-deficient mice clearly demonstrate a role for 4-1BBL in CD8 T cell responses to viruses such as influenza, 4-1BBL can be difficult to detect following infection of mice. Here we provide evidence for a constitutive interaction between endogenous 4-1BB and 4-1BBL on LPS activated bone marrow-derived murine dendritic cells that can mask its detection, with implications for measurement of 4-1BBL expression. The masking of 4-1BBL by its receptor results in loss of reactivity to the anti-4-1BBL antibody TKS-1, whereas the 19H3 antibody binds to 4-1BBL in the presence or absence of 4-1BB. Moreover, 4-1BB/4-1BBL interaction can occur in trans between 4-1BB+/+ and 4-1BB-/- dendritic cells in culture. These data suggest that 19H3 is the preferable antibody to use to detect 4-1BBL in the presence of its receptor.


Subject(s)
4-1BB Ligand/immunology , Antibodies/immunology , Bone Marrow Cells/drug effects , Cell Separation/methods , Dendritic Cells/drug effects , Flow Cytometry , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , 4-1BB Ligand/genetics , 4-1BB Ligand/metabolism , Animals , Antibody Specificity , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Binding , Reproducibility of Results , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
7.
Nat Commun ; 8: 14003, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084302

ABSTRACT

T-cell proliferation is regulated by ubiquitination but the underlying molecular mechanism remains obscure. Here we report that Lys-48-linked ubiquitination of the transcription factor KLF4 mediated by the E3 ligase Mule promotes T-cell entry into S phase. Mule is elevated in T cells upon TCR engagement, and Mule deficiency in T cells blocks proliferation because KLF4 accumulates and drives upregulation of its transcriptional targets E2F2 and the cyclin-dependent kinase inhibitors p21 and p27. T-cell-specific Mule knockout (TMKO) mice develop exacerbated experimental autoimmune encephalomyelitis (EAE), show impaired generation of antigen-specific CD8+ T cells with reduced cytokine production, and fail to clear LCMV infections. Thus, Mule-mediated ubiquitination of the novel substrate KLF4 regulates T-cell proliferation, autoimmunity and antiviral immune responses in vivo.


Subject(s)
Cell Proliferation , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
8.
Clin Cancer Res ; 23(4): 1068-1079, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27856600

ABSTRACT

Purpose: The ubiquitously expressed transmembrane glycoprotein CD47 delivers an anti-phagocytic (do not eat) signal by binding signal-regulatory protein α (SIRPα) on macrophages. CD47 is overexpressed in cancer cells and its expression is associated with poor clinical outcomes. TTI-621 (SIRPαFc) is a fully human recombinant fusion protein that blocks the CD47-SIRPα axis by binding to human CD47 and enhancing phagocytosis of malignant cells. Blockade of this inhibitory axis using TTI-621 has emerged as a promising therapeutic strategy to promote tumor cell eradication.Experimental Design: The ability of TTI-621 to promote macrophage-mediated phagocytosis of human tumor cells was assessed using both confocal microscopy and flow cytometry. In vivo antitumor efficacy was evaluated in xenograft and syngeneic models and the role of the Fc region in antitumor activity was evaluated using SIRPαFc constructs with different Fc tails.Results: TTI-621 enhanced macrophage-mediated phagocytosis of both hematologic and solid tumor cells, while sparing normal cells. In vivo, TTI-621 effectively controlled the growth of aggressive AML and B lymphoma xenografts and was efficacious in a syngeneic B lymphoma model. The IgG1 Fc tail of TTI-621 plays a critical role in its antitumor activity, presumably by engaging activating Fcγ receptors on macrophages. Finally, TTI-621 exhibits minimal binding to human erythrocytes, thereby differentiating it from CD47 blocking antibodies.Conclusions: These data indicate that TTI-621 is active across a broad range of human tumors. These results further establish CD47 as a critical regulator of innate immune surveillance and form the basis for clinical development of TTI-621 in multiple oncology indications. Clin Cancer Res; 23(4); 1068-79. ©2016 AACR.


Subject(s)
Antigens, Differentiation/genetics , CD47 Antigen/genetics , Immunoglobulin G/immunology , Neoplasms/drug therapy , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , CD47 Antigen/antagonists & inhibitors , Cell Line, Tumor , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis/drug effects , Protein Binding , Protein Interaction Maps/drug effects , Receptors, Immunologic/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 112(4): 1119-24, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25583492

ABSTRACT

UV radiation resistance-associated gene (UVRAG) encodes a tumor suppressor with putative roles in autophagy, endocytic trafficking, and DNA damage repair but its in vivo role in T cells is unknown. Because conditional homozygous deletion of Uvrag in mice results in early embryonic lethality, we generated T-cell-specific UVRAG-deficient mice that lacked UVRAG expression specifically in T cells. This loss of UVRAG led to defects in peripheral homeostasis that could not be explained by the increased sensitivity to cell death and impaired proliferation observed for other autophagy-related gene knockout mice. Instead, UVRAG-deficient T-cells exhibited normal mitochondrial clearance and activation-induced autophagy, suggesting that UVRAG has an autophagy-independent role that is critical for peripheral naive T-cell homeostatic proliferation. In vivo, T-cell-specific loss of UVRAG dampened CD8(+) T-cell responses to LCMV infection in mice, delayed viral clearance, and impaired memory T-cell generation. Our data provide novel insights into the control of autophagy in T cells and identify UVRAG as a new regulator of naïve peripheral T-cell homeostasis.


Subject(s)
Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Tumor Suppressor Proteins/immunology , Animals , Autophagy/genetics , CD8-Positive T-Lymphocytes/pathology , Gene Deletion , Homeostasis/genetics , Homeostasis/immunology , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Knockout , Tumor Suppressor Proteins/isolation & purification
10.
Diabetes ; 64(1): 90-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25048196

ABSTRACT

Obesity-related insulin resistance is associated with an influx of pathogenic T cells into visceral adipose tissue (VAT), but the mechanisms regulating lymphocyte balance in such tissues are unknown. Here we describe an important role for the immune cytotoxic effector molecule perforin in regulating this process. Perforin-deficient mice (Prf1(null)) show early increased body weight and adiposity, glucose intolerance, and insulin resistance when placed on high-fat diet (HFD). Regulatory effects of perforin on glucose tolerance are mechanistically linked to the control of T-cell proliferation and cytokine production in inflamed VAT. HFD-fed Prf1(null) mice have increased accumulation of proinflammatory IFN-γ-producing CD4(+) and CD8(+) T cells and M1-polarized macrophages in VAT. CD8(+) T cells from the VAT of Prf1(null) mice have increased proliferation and impaired early apoptosis, suggesting a role for perforin in the regulation of T-cell turnover during HFD feeding. Transfer of CD8(+) T cells from Prf1(null) mice into CD8-deficient mice (CD8(null)) resulted in worsening of metabolic parameters compared with wild-type donors. Improved metabolic parameters in HFD natural killer (NK) cell-deficient mice (NK(null)) ruled out a role for NK cells as a single source of perforin in regulating glucose homeostasis. The findings support the importance of T-cell function in insulin resistance and suggest that modulation of lymphocyte homeostasis in inflamed VAT is one possible avenue for therapeutic intervention.


Subject(s)
Glucose Intolerance/immunology , Insulin Resistance/immunology , Intra-Abdominal Fat/immunology , Obesity/immunology , Panniculitis/immunology , Perforin/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Diet, High-Fat , Embryonic Stem Cells/cytology , Female , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Insulin/metabolism , Intra-Abdominal Fat/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Panniculitis/genetics , Panniculitis/metabolism , Perforin/genetics , Perforin/metabolism , Spleen/cytology , Spleen/immunology
11.
Proc Natl Acad Sci U S A ; 111(3): 1060-5, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24398517

ABSTRACT

The ability to mount a strong immune response against pathogens is crucial for mammalian survival. However, excessive and uncontrolled immune reactions can lead to autoimmunity. Unraveling how the reactive versus tolerogenic state is controlled might point toward novel therapeutic strategies to treat autoimmune diseases. The surface receptor Toso/Faim3 has been linked to apoptosis, IgM binding, and innate immune responses. In this study, we used Toso-deficient mice to investigate the importance of Toso in tolerance and autoimmunity. We found that Toso(-/-) mice do not develop severe experimental autoimmune encephalomyelitis (EAE), a mouse model for the human disease multiple sclerosis. Toso(-/-) dendritic cells were less sensitive to Toll-like receptor stimulation and induced significantly lower levels of disease-associated inflammatory T-cell responses. Consistent with this observation, the transfer of Toso(-/-) dendritic cells did not induce autoimmune diabetes, indicating their tolerogenic potential. In Toso(-/-) mice subjected to EAE induction, we found increased numbers of regulatory T cells and decreased encephalitogenic cellular infiltrates in the brain. Finally, inhibition of Toso activity in vivo at either an early or late stage of EAE induction prevented further disease progression. Taken together, our data identify Toso as a unique regulator of inflammatory autoimmune responses and an attractive target for therapeutic intervention.


Subject(s)
Carrier Proteins/metabolism , Dendritic Cells/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Proteins/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Cell Differentiation , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Gene Expression Regulation , HEK293 Cells , Humans , Immunohistochemistry , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th17 Cells/cytology , Time Factors
12.
Immunol Rev ; 255(1): 125-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947352

ABSTRACT

Immunity to viruses must be tightly controlled to avoid pathology. Receptors and ligands of the tumor necrosis factor (TNF) family play important roles in controlling lymphocyte activation and survival during an immune response. The role of specific TNF receptor (TNFR) family members in antiviral immunity depends on the stage of the immune response and can vary with the virus type and its virulence. Here, we focus on five members of the TNFR family that are prominently expressed on CD8(+) T cells during viral infections, namely, 4-1BB (CD137), CD27, OX40 (CD134), GITR, and TNFR2. 4-1BB, CD27, OX40, and GITR have primarily prosurvival roles for CD8(+) T cells during viral infection, although under some circumstances 4-1BB, GITR, or CD27 signals can limit immunity. Although TNFR2 can be costimulatory under some circumstances, its main role in CD8(+) T-cell responses during viral infection appears to be in contraction of the response. Several TNF family ligands are being explored as adjuvants for viral vaccines, and agonistic antibodies to TNFR family members are being investigated for immunotherapy of chronic viral infection alone and in combination with checkpoint blockade. Such therapies will require thorough and specific optimization to avoid pathology induced by hyperstimulation of these pathways.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology , Animals , Humans , Immunologic Memory , Protein Binding , Signal Transduction , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
13.
PLoS One ; 8(7): e68911, 2013.
Article in English | MEDLINE | ID: mdl-23874808

ABSTRACT

TNF is an important inflammatory mediator and a target for intervention. TNF is produced by many cell types and is involved in innate inflammation as well as adaptive immune responses. CD8 T cells produce TNF and can also respond to TNF. Deficiency of TNF or TNFR2 has been shown to affect anti-viral immunity. However, as the complete knockout of TNF or its receptors has effects on multiple cell types as well as on lymphoid architecture, it has been difficult to assess the role of TNF directly on T cells during viral infection. Here we have addressed this issue by analyzing the effect of CD8 T cell intrinsic TNF/TNFR2 interactions during respiratory influenza infection in mice, using an adoptive transfer model in which only the T cells lack TNF or TNFR2. During a mild influenza infection, the capacity of the responding CD8 T cells to produce TNF increases from day 6 through day 12, beyond the time of viral clearance. Although T cell intrinsic TNF is dispensable for initial expansion of CD8 T cells up to day 9 post infection, intrinsic TNF/TNFR2 interactions potentiate contraction of the CD8 T cell response in the lung between day 9 and 12 post infection. On the other hand, TNF or TNFR2-deficient CD8 T cells in the lung express lower levels of IFN-γ and CD107a per cell than their wild type counterparts. Comparison of TNF levels on the TNFR2 positive and negative T cells is consistent with TNF/TNFR2 interactions inducing feedback downregulation of TNF production by T cells, with greater effects in the lung compared to spleen. Thus CD8 T cell intrinsic TNF/TNFR2 interactions fine-tune the response to influenza virus in the lung by modestly enhancing effector functions, but at the same time potentiating the contraction of the CD8 T cell response post-viral clearance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Tumor Necrosis Factors/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Degranulation , Cytokines/biosynthesis , Disease Models, Animal , Lung/immunology , Lung/metabolism , Lung/virology , Male , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Phenotype , Receptors, Tumor Necrosis Factor, Type II/genetics , Respiratory Tract Infections/virology , Tumor Necrosis Factors/genetics
14.
J Immunol ; 190(9): 4627-39, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536631

ABSTRACT

The TNFR family member 4-1BB plays a key role in the survival of activated and memory CD8 T cells. However, the mechanisms that regulate 4-1BB re-expression on memory CD8 T cells after Ag clearance are unknown. In unimmunized mice, ∼10% of CD8 CD44(hi) memory T cells in the bone marrow (BM) and liver express 4-1BB, with minimal 4-1BB expression in spleen and lymph node. IL-2, IL-15, and IL-7 are collectively dispensable for 4-1BB expression on the memory CD8 T cells. Rather, T cell-intrinsic glucocorticoid-induced TNFR-related protein (GITR) contributes to 4-1BB expression on CD8 T cells upon their entry into the BM or liver. Consistent with its role in regulation of 4-1BB, GITR is required on memory CD8 T cells for their persistence in vivo. These findings reveal site-specific effects of the BM and liver microenvironment on CD8 memory T cells. Previous work has demonstrated that 4-1BB agonists given to unimmunized mice induce splenomegaly, hepatitis, and other immune system anomalies. Moreover, severe liver pathology has been observed in a subset of anti-4-1BB-treated melanoma patients. Remarkably, the absence of GITR in mice almost completely abrogates cellular expansions, splenomegaly, and liver inflammation associated with anti-4-1BB agonist treatment of unimmunized mice. In contrast, lack of CD8 T cells selectively improves liver pathology, but not splenomegaly in the mice. Thus, the regulation of 4-1BB expression by GITR on CD8 T cells, as well as on other cells, contributes to the pathological effects of anti-4-1BB in unimmunized mice.


Subject(s)
4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , CD8-Positive T-Lymphocytes/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immunologic Memory/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Hepatitis/immunology , Hepatitis/metabolism , Humans , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Interleukins/immunology , Interleukins/metabolism , Liver/immunology , Liver/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Spleen/immunology , Spleen/metabolism , Splenomegaly/immunology , Splenomegaly/metabolism
15.
PLoS One ; 8(2): e56014, 2013.
Article in English | MEDLINE | ID: mdl-23405246

ABSTRACT

NOD2 is an intracellular pattern recognition receptor that provides innate sensing of bacterial muramyl dipeptide by host cells, such as dendritic cells, macrophages and epithelial cells. While NOD2's role as an innate pathogen sensor is well established, NOD2 is also expressed at low levels in T cells and there are conflicting data as to whether NOD2 plays an intrinsic role in T cell function. Here we show that following adoptive transfer into WT hosts, NOD2(-/-) OT-I T cells show a small decrease in the number of OVA-specific CD8 T cells recovered at the peak of the response to respiratory influenza virus infection. On the other hand, no such defect was observed upon intranasal immunization with a replication defective adenovirus carrying the OVA epitope recognized by OT-I, or when OVA was delivered with LPS subcutaneously, or when influenza-OVA was delivered intraperitoneally. Thus we observed a selective defect in NOD2-deficient T cell responses only during a live viral infection. Moreover, there was no apparent defect when NOD2(-/-) OT-I T cells were stimulated in vitro. Finally, this selective defect in recovery of NOD2-deficient CD8 T cells was not observed in a non-transgenic respiratory infection model in which mixed bone marrow chimeras were used such that the NOD2(-/-) T cells were allowed to develop and respond in a NOD2-sufficient host. Taken together our data indicate that T cell intrinsic NOD2 is not required for CD8 T cell responses to antigen delivered under a variety of conditions in vitro and in vivo. However, CD8 T cells that have developed in the absence of NOD2 show a selective and modest impairment in their response to live respiratory influenza infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Influenza A virus/immunology , Nod2 Signaling Adaptor Protein/physiology , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell, alpha-beta/physiology , Spleen/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/virology , Flow Cytometry , Immunization , Lipopolysaccharides/pharmacology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/virology , Ovalbumin/metabolism , Spleen/virology
16.
Eur J Immunol ; 42(11): 2861-74, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22886791

ABSTRACT

The persistence of memory lymphocytes is a critical feature of adaptive immunity. The TNF family ligand 4-1BBL supports the antigen-independent survival of CD8⁺ memory T cells. Here, we show that mice lacking 4-1BB only on αß T cells show a similar defect in CD8⁺ T-cell recall responses, as previously shown in 4-1BBL-deficient mice. We show that 4-1BB is selectively expressed on BM CD8⁺ but not CD4⁺ memory T cells of unimmunized mice. Its ligand, 4-1BBL, is found on VCAM-1⁺ stromal cells, CD11c⁺ cells, and a Gr1(lo) myeloid population in unimmunized mice. Adoptive transfer of in vitro generated memory T cells into mice lacking 4-1BBL only on radioresistant cells recapitulates the defect in CD8⁺ T-cell survival seen in the complete knockout mice, with smaller effects of 4-1BBL on hematopoietic cells. In BM, adoptively transferred DsRed CD8⁺ memory T cells are most often found in proximity to VCAM-1⁺ cells or Gr1⁺ cells, followed by B220⁺ cells and to a much lesser extent near CD11c⁺ cells. Thus, a VCAM-1⁺CD45(-) stromal cell is a plausible candidate for the radioresistant cell that provides 4-1BBL to CD8⁺ memory T cells in the BM.


Subject(s)
4-1BB Ligand/immunology , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Vascular Cell Adhesion Molecule-1/immunology , 4-1BB Ligand/genetics , Adoptive Transfer , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/radiation effects , Female , Flow Cytometry , Immunohistochemistry , Immunologic Memory/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA, Viral/chemistry , RNA, Viral/genetics , Radiation Tolerance/immunology , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Transplantation Chimera
17.
J Immunol ; 188(12): 5915-23, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22581858

ABSTRACT

CD8 memory T cells are enriched in the bone marrow, a site where these cells are thought to receive homeostatic signals. However, the primary site where CD8 memory T cells receive their cytokine-induced homeostatic signals has recently come under debate. In this study, we demonstrate that the bone marrow contains a fraction of CD8 memory phenotype T cells with elevated expression of glucocorticoid-induced TNFR-related protein (GITR). In contrast, splenic and lymph node memory phenotype T cells have GITR levels similar to those on naive T cells. The bone marrow GITR(hi) memory T cells have a phenotype indicative of cytokine activation, with higher CD122 and lower CD127 than do the GITR(basal) memory T cells. Remarkably, these bone marrow-specific GITR(hi) cells are almost completely ablated in the absence of IL-15, whereas TNFR2 and 4-1BB expression on the CD8 memory T cells are IL-15 independent. Furthermore, adoptively transferred splenic CD8 memory phenotype T cells show IL-15-dependent GITR upregulation upon entry into the bone marrow. This result implies that the selective appearance of GITR(hi) memory phenotype T cells in the bone marrow reflects the local microenvironment rather than a different subset of memory T cells. GITR(-/-) mice have a lower frequency of CD8 memory phenotype cells in the bone marrow, yet the GITR(-/-) cells hyperproliferate compared with those in wild-type mice. Taken together, these data suggest that GITR plays a role in the survival of CD8 memory phenotype T cells and that GITR upregulation represents a precise marker of cells that have responded to IL-15.


Subject(s)
Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Immunologic Memory/immunology , Interleukin-15/immunology , Adoptive Transfer , Animals , Bone Marrow/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Homeostasis/immunology , Interleukin-15/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Up-Regulation
18.
Immunol Cell Biol ; 90(7): 743-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21946662

ABSTRACT

Immune complexes combining IL-2 with particular anti-IL-2 antibodies can be used to selectively expand regulatory T cells or memory T cells. Combining IL-2 with anti-IL-2 (Clone S4B6) greatly enhances the biological potency of IL-2 in vivo leading to selective expansion of CD8 memory T cells and NK cells compared with regulatory T cells. Here we show that in vivo administration of IL-2/anti-IL-2 mAb (IL-2/mAb) complexes induces 4-1BB expression on both adoptively transferred antigen-specific memory CD8 T cells as well as on endogenous memory phenotype cells. Remarkably, the accumulation of adoptively transferred memory CD8 T cells following in vivo IL-2/mAb-complex treatment was found to be dependent in part on the presence of 4-1BBL in the host. These effects were independent of IL-2-induced cell division, suggesting that 4-1BBL-induced survival signals contribute to IL-2/mAb-complex-induced T-cell accumulation in vivo.


Subject(s)
4-1BB Ligand/immunology , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/pharmacology , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Flow Cytometry , Immunologic Memory/immunology , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
19.
J Exp Med ; 209(1): 77-91, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22184633

ABSTRACT

The signaling adaptor TNFR-associated factor 1 (TRAF1) is specifically lost from virus-specific CD8 T cells during the chronic phase of infection with HIV in humans or lymphocytic choriomeningitis virus (LCMV) clone 13 in mice. In contrast, TRAF1 is maintained at higher levels in virus-specific T cells of HIV controllers or after acute LCMV infection. TRAF1 expression negatively correlates with programmed death 1 expression and HIV load and knockdown of TRAF1 in CD8 T cells from viral controllers results in decreased HIV suppression ex vivo. Consistent with the desensitization of the TRAF1-binding co-stimulatory receptor 4-1BB, 4-1BBL-deficient mice have defects in viral control early, but not late, in chronic infection. TGFß induces the posttranslational loss of TRAF1, whereas IL-7 restores TRAF1 levels. A combination treatment with IL-7 and agonist anti-4-1BB antibody at 3 wk after LCMV clone 13 infection expands T cells and reduces viral load in a TRAF1-dependent manner. Moreover, transfer of TRAF1(+) but not TRAF1(-) memory T cells at the chronic stage of infection reduces viral load. These findings identify TRAF1 as a potential biomarker of HIV-specific CD8 T cell fitness during the chronic phase of disease and a target for therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Lymphocytic Choriomeningitis/immunology , TNF Receptor-Associated Factor 1/deficiency , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , Adoptive Transfer , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Chloroquine/pharmacology , Chronic Disease , Down-Regulation/genetics , Gene Expression , HIV Infections/genetics , Humans , Immunologic Memory , Interleukin-7/pharmacology , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 1/genetics , Transforming Growth Factor beta/metabolism , Viral Load/immunology
20.
Immunol Rev ; 244(1): 197-217, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22017440

ABSTRACT

GITR [glucocorticoid inducible tumor necrosis factor receptor (TNFR)-related protein] and 4-1BB are costimulatory TNFR family members that are expressed on regulatory and effector T cells as well as on other cells of the immune system. Here we discuss the role of GITR and 4-1BB on T cells during viral infections and in cancer immunotherapy. Systemic treatment with agonistic anti-4-1BB antibody leads to a number of immune system abnormalities, and clinical trials of anti-4-1BB have been terminated. However, other modes of 4-1BB ligation may be less toxic. To date, similar toxicities have not been reported for anti-GITR treatment of mice, although anti-GITR antibodies can exacerbate mouse autoimmune models. Intrinsic effects of GITR and 4-1BB on effector T cells appear to predominate over their effects on other cell types in some models. Despite their similarities in enhancing T-cell survival, 4-1BB and GITR are clearly not redundant, and both pathways are required for maximal CD8(+) T-cell responses and mouse survival following severe respiratory influenza infection. GITR uses TNFR-associated factor (TRAF) 2 and TRAF5, whereas 4-1BB recruits TRAF1 and TRAF2 to mediate survival signaling in T cells. The differential use of signaling adapters combined with their differential expression may explain the non-redundant roles of GITR and 4-1BB in the immune system.


Subject(s)
Antibodies/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Immunity, Innate , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Animals , Antibodies/administration & dosage , Autoimmunity/drug effects , Cell Proliferation/drug effects , Gene Expression/immunology , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Immunity, Innate/drug effects , Immunotherapy/methods , Influenza A virus/immunology , Mice , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...