Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
J Immunol ; 209(8): 1475-1480, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36096643

ABSTRACT

Vγ9Vδ2+ T cell-targeted immunotherapy is of interest to harness its MHC-independent cytotoxic potential against a variety of cancers. Recent studies have identified heterodimeric butyrophilin (BTN) 2A1 and BTN3A1 as the molecular entity providing "signal 1" to the Vγ9Vδ2 TCR, but "signal 2" costimulatory requirements remain unclear. Using a tumor cell-free assay, we demonstrated that a BTN2A1/3A1 heterodimeric fusion protein activated human Vγ9Vδ2+ T cells, but only in the presence of costimulatory signal via CD28 or NK group 2 member D. Nonetheless, addition of a bispecific γδ T cell engager BTN2A1/3A1-Fc-CD19scFv alone enhanced granzyme B-mediated killing of human CD19+ lymphoma cells when cocultured with Vγ9Vδ2+ T cells, suggesting expression of costimulatory ligand(s) on tumor cells is sufficient to satisfy the "signal 2" requirement. These results highlight the parallels of signal 1 and signal 2 requirements in αß and γδ T cell activation and demonstrate the utility of heterodimeric BTNs to promote targeted activation of γδ T cells.


Subject(s)
CD28 Antigens , Receptors, Antigen, T-Cell, gamma-delta , Antigens, CD/metabolism , Butyrophilins/metabolism , Granzymes , Humans , Ligands , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Antigen, T-Cell, gamma-delta/metabolism
2.
J Immunol ; 209(3): 510-525, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35817517

ABSTRACT

Coinhibition of TIGIT (T cell immunoreceptor with Ig and ITIM domains) and PD-1/PD-L1 (PD-1/L1) may improve response rates compared with monotherapy PD-1/L1 blockade in checkpoint naive non-small cell lung cancer with PD-L1 expression >50%. TIGIT mAbs with an effector-competent Fc can induce myeloid cell activation, and some have demonstrated effector T cell depletion, which carries a clinical liability of unknown significance. TIGIT Ab blockade translates to antitumor activity by enabling PVR signaling through CD226 (DNAM-1), which can be directly inhibited by PD-1. Furthermore, DNAM-1 is downregulated on tumor-infiltrating lymphocytes (TILs) in advanced and checkpoint inhibition-resistant cancers. Therefore, broadening clinical responses from TIGIT blockade into PD-L1low or checkpoint inhibition-resistant tumors, may be induced by immune costimulation that operates independently from PD-1/L1 inhibition. TNFSF14 (LIGHT) was identified through genomic screens, in vitro functional analysis, and immune profiling of TILs as a TNF ligand that could provide broad immune activation. Accordingly, murine and human bifunctional fusion proteins were engineered linking the extracellular domain of TIGIT to the extracellular domain of LIGHT, yielding TIGIT-Fc-LIGHT. TIGIT competitively inhibited binding to all PVR ligands. LIGHT directly activated myeloid cells through interactions with LTßR (lymphotoxin ß receptor), without the requirement for a competent Fc domain to engage Fcγ receptors. LIGHT costimulated CD8+ T and NK cells through HVEM (herpes virus entry mediator A). Importantly, HVEM was more widely expressed than DNAM-1 on T memory stem cells and TILs across a range of tumor types. Taken together, the mechanisms of TIGIT-Fc-LIGHT promoted strong antitumor activity in preclinical tumor models of primary and acquired resistance to PD-1 blockade, suggesting that immune costimulation mediated by LIGHT may broaden the clinical utility of TIGIT blockade.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , B7-H1 Antigen/genetics , Humans , Mice , Myeloid Cells/metabolism , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
3.
Cancer Immunol Res ; 8(2): 230-245, 2020 02.
Article in English | MEDLINE | ID: mdl-31852716

ABSTRACT

Disrupting the binding of CD47 to SIRPα has emerged as a promising immunotherapeutic strategy for advanced cancers by potentiating antibody-dependent cellular phagocytosis (ADCP) of targeted antibodies. Preclinically, CD47/SIRPα blockade induces antitumor activity by increasing the phagocytosis of tumor cells by macrophages and enhancing the cross-presentation of tumor antigens to CD8+ T cells by dendritic cells; both of these processes are potentiated by CD40 signaling. Here we generated a novel, two-sided fusion protein incorporating the extracellular domains of SIRPα and CD40L, adjoined by a central Fc domain, termed SIRPα-Fc-CD40L. SIRPα-Fc-CD40L bound CD47 and CD40 with high affinity and activated CD40 signaling in the absence of Fc receptor cross-linking. No evidence of hemolysis, hemagglutination, or thrombocytopenia was observed in vitro or in cynomolgus macaques. Murine SIRPα-Fc-CD40L outperformed CD47 blocking and CD40 agonist antibodies in murine CT26 tumor models and synergized with immune checkpoint blockade of PD-1 and CTLA4. SIRPα-Fc-CD40L activated a type I interferon response in macrophages and potentiated the activity of ADCP-competent targeted antibodies both in vitro and in vivo These data illustrated that whereas CD47/SIRPα inhibition could potentiate tumor cell phagocytosis, CD40-mediated activation of a type I interferon response provided a bridge between macrophage- and T-cell-mediated immunity that significantly enhanced durable tumor control and rejection.


Subject(s)
CD40 Antigens/metabolism , CD47 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Interferon Type I/immunology , Neoplasms/immunology , Neoplasms/therapy , Recombinant Fusion Proteins/pharmacology , Adaptive Immunity , Animals , CD40 Ligand/genetics , CD40 Ligand/immunology , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cell Line, Tumor , Humans , Immunity, Innate , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interferon Type I/metabolism , Macaca fascicularis , Macrophages/immunology , Mice , Mice, Inbred BALB C , Neoplasms/metabolism , Neoplasms/pathology , Random Allocation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
4.
J Immunother Cancer ; 6(1): 149, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30563566

ABSTRACT

Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of the TNFRSF, and is most effective when these functions occur simultaneously. To address this mechanism, we developed a two-sided human fusion protein incorporating the extracellular domains (ECD) of PD-1 and OX40L, adjoined by a central Fc domain, termed PD1-Fc-OX40L. The PD-1 end of the fusion protein binds PD-L1 and PD-L2 with affinities of 2.08 and 1.76 nM, respectively, and the OX40L end binds OX40 with an affinity of 246 pM. High binding affinity on both sides of the construct translated to potent stimulation of OX40 signaling and PD1:PD-L1/L2 blockade, in multiple in vitro assays, including improved potency as compared to pembrolizumab, nivolumab, tavolixizumab and combinations of those antibodies. Furthermore, when activated human T cells were co-cultured with PD-L1 positive human tumor cells, PD1-Fc-OX40L was observed to concentrate to the immune synapse, which enhanced proliferation of T cells and production of IL-2, IFNγ and TNFα, and led to efficient killing of tumor cells. The therapeutic activity of PD1-Fc-OX40L in established murine tumors was significantly superior to either PD1 blocking, OX40 agonist, or combination antibody therapy; and required CD4+ T cells for maximum response. Importantly, all agonist functions of PD1-Fc-OX40L are independent of Fc receptor cross-linking. Collectively, these data demonstrate a highly potent fusion protein that is part of a platform, capable of providing checkpoint blockade and TNFRSF costimulation in a single molecule, which uniquely localizes TNFRSF costimulation to checkpoint ligand positive tumor cells.


Subject(s)
CD40 Ligand/metabolism , Immunoglobulin Fc Fragments , Immunomodulation , Neoplasms/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , Recombinant Fusion Proteins/metabolism , Animals , B7-H1 Antigen/metabolism , Biomarkers, Tumor , CD40 Ligand/chemistry , Cell Line , Cytotoxicity, Immunologic , Humans , Immunoglobulin Fc Fragments/immunology , Lymphocyte Activation/immunology , Mice , Neoplasms/immunology , Neoplasms/mortality , Programmed Cell Death 1 Receptor/chemistry , Protein Binding , Receptors, OX40/metabolism , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Cell Rep ; 5(1): 166-79, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24095736

ABSTRACT

Thymic epithelial cells in the medulla (mTECs) play a critical role in enforcing central tolerance through expression and presentation of tissue-specific antigens (TSAs) and deletion of autoreactive thymocytes. TSA expression requires autoimmune regulator (Aire), a transcriptional activator present in a subset of mTECs characterized by high CD80 and major histocompatibility complex II expression and a lack of potential for differentiation or proliferation. Here, using an Aire-DTR transgenic line, we show that short-term ablation specifically targets Aire(+) mTECs, which quickly undergo RANK-dependent recovery. Repeated ablation also affects Aire(-) mTECs, and using an inducible Aire-Cre fate-mapping system, we find that this results from the loss of a subset of mTECs that showed prior expression of Aire, maintains intermediate TSA expression, and preferentially migrates toward the center of the medulla. These results clearly identify a distinct stage of mTEC development and underscore the diversity of mTECs that play a key role in maintaining tolerance.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cell Differentiation/physiology , Female , Humans , Mice , Mice, Transgenic , Signal Transduction
6.
Immunity ; 39(3): 560-72, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23993652

ABSTRACT

The autoimmune regulator (Aire) is essential for prevention of autoimmunity; its role is best understood in the thymus, where it promotes self-tolerance through tissue-specific antigen (TSA) expression. Recently, extrathymic Aire-expressing cells (eTACs) have been described in murine secondary lymphoid organs, but the identity of such cells and their role in immune tolerance remains unclear. Here we have shown that eTACs are a discrete major histocompatibility complex class II (MHC II)(hi), CD80(lo), CD86(lo), epithelial cell adhesion molecule (EpCAM)(hi), CD45(lo) bone marrow-derived peripheral antigen-presenting cell (APC) population. We also have demonstrated that eTACs can functionally inactivate CD4⁺ T cells through a mechanism that does not require regulatory T cells (Treg) and is resistant to innate inflammatory stimuli. Together, these findings further define eTACs as a distinct tolerogenic cell population in secondary lymphoid organs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Self Tolerance , Transcription Factors/metabolism , Adoptive Transfer , Animals , Antigen Presentation , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/metabolism , Autoimmunity , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Bone Marrow Cells , Cell Adhesion Molecules/metabolism , Epithelial Cell Adhesion Molecule , Histocompatibility Antigens Class II/immunology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred NOD , Transcription Factors/biosynthesis , AIRE Protein
7.
J Immunol ; 188(10): 4906-12, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22490868

ABSTRACT

Chronic inflammatory demyelinating polyneuropathy is a debilitating autoimmune disease characterized by peripheral nerve demyelination and dysfunction. How the autoimmune response is initiated, identity of provoking Ags, and pathogenic effector mechanisms are not well defined. The autoimmune regulator (Aire) plays a critical role in central tolerance by promoting thymic expression of self-Ags and deletion of self-reactive T cells. In this study, we used mice with hypomorphic Aire function and two patients with Aire mutations to define how Aire deficiency results in spontaneous autoimmune peripheral neuropathy. Autoimmunity against peripheral nerves in both mice and humans targets myelin protein zero, an Ag for which expression is Aire-regulated in the thymus. Consistent with a defect in thymic tolerance, CD4(+) T cells are sufficient to transfer disease in mice and produce IFN-γ in infiltrated peripheral nerves. Our findings suggest that defective Aire-mediated central tolerance to myelin protein zero initiates an autoimmune Th1 effector response toward peripheral nerves.


Subject(s)
Immune Tolerance , Myelin P0 Protein/deficiency , Point Mutation , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , Amino Acid Sequence , Animals , Autoantibodies/blood , Disease Models, Animal , Female , Humans , Immune Tolerance/genetics , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mice, SCID , Molecular Sequence Data , Myelin P0 Protein/genetics , Myelin P0 Protein/physiology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/blood , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/genetics , Transcription Factors/physiology , AIRE Protein
8.
J Immunol ; 184(8): 4236-46, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20237294

ABSTRACT

Sjögren's Syndrome (SS) is a human autoimmune disease characterized by immune-mediated destruction of the lacrimal and salivary glands. In this study, we show that the Aire-deficient mouse represents a new tool to investigate autoimmune dacryoadenitis and keratoconjunctivitis sicca, features of SS. Previous work in the Aire-deficient mouse suggested a role for alpha-fodrin, a ubiquitous Ag, in the disease process. Using an unbiased biochemical approach, however, we have identified a novel lacrimal gland autoantigen, odorant binding protein 1a, targeted by the autoimmune response. This novel autoantigen is expressed in the thymus in an Aire-dependent manner. The results from our study suggest that defects in central tolerance may contribute to SS and provide a new and clinically relevant model to investigate the pathogenic mechanisms in lacrimal gland autoimmunity and associated ocular surface sequelae.


Subject(s)
Autoantibodies/biosynthesis , Dry Eye Syndromes/genetics , Dry Eye Syndromes/immunology , Receptors, Odorant/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Autoantibodies/blood , Dacryocystitis/genetics , Dacryocystitis/immunology , Dacryocystitis/pathology , Disease Models, Animal , Dry Eye Syndromes/pathology , Female , Humans , Keratoconjunctivitis Sicca/genetics , Keratoconjunctivitis Sicca/immunology , Keratoconjunctivitis Sicca/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Receptors, Odorant/biosynthesis , Receptors, Odorant/genetics , Sjogren's Syndrome/pathology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology , AIRE Protein
9.
Sci Transl Med ; 1(9): 9ra20, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-20368189

ABSTRACT

Interstitial lung disease (ILD) is a common manifestation of systemic autoimmunity characterized by progressive inflammation or scarring of the lungs. Patients who develop these complications can exhibit significantly impaired gas exchange that may result in hypoxemia, pulmonary hypertension, and even death. Unfortunately, little is understood about how these diseases arise, including the role of specific defects in immune tolerance. Another key question is whether autoimmune responses targeting the lung parenchyma are critical to ILD pathogenesis, including that of isolated idiopathic forms. We show that a specific defect in central tolerance brought about by mutations in the autoimmune regulator gene (Aire) leads to an autoreactive T cell response to a lung antigen named vomeromodulin and the development of ILD. We found that a human patient and mice with defects in Aire develop similar lung pathology, demonstrating that the AIRE-deficient model of autoimmunity is a suitable translational system in which to unravel fundamental mechanisms of ILD pathogenesis.


Subject(s)
Adaptation, Physiological , Autoantigens/analysis , Lung Diseases, Interstitial/immunology , Animals , Lung Diseases, Interstitial/physiopathology , Mice , Transcription Factors/genetics , Transcription Factors/physiology , AIRE Protein
10.
J Immunol ; 181(6): 4072-9, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768863

ABSTRACT

Mutations in the Aire gene result in a clinical phenomenon known as Autoimmune Polyglandular Syndrome (APS) Type I, which classically manifests as a triad of adrenal insufficiency, hypoparathyroidism, and chronic mucocutaneous infections. In addition to this triad, a number of other autoimmune diseases have been observed in APS1 patients including Sjögren's syndrome, vitiligo, alopecia, uveitis, and others. Aire-deficient mice, the animal model for APS1, have highlighted the role of the thymus in the disease process and demonstrated a failure in central tolerance in aire-deficient mice. However, autoantibodies have been observed against multiple organs in both mice and humans, making it unclear what the specific role of B and T cells are in the pathogenesis of disease. Using the aire-deficient mouse as a preclinical model for APS1, we have investigated the relative contribution of specific lymphocyte populations, with the goal of identifying the cell populations which may be targeted for rational therapeutic design. In this study, we show that T cells are indispensable to the breakdown of self-tolerance, in contrast to B cells which play a more limited role in autoimmunity. Th1 polarized CD4(+) T cells, in particular, are major contributors to the autoimmune response. With this knowledge, we go on to use therapies targeted at T cells to investigate their ability to modulate disease in vivo. Depletion of CD4(+) T cells using a neutralizing Ab ameliorated the disease process. Thus, therapies targeted specifically at the CD4(+) T cell subset may help control autoimmune disease in patients with APS1.


Subject(s)
Polyendocrinopathies, Autoimmune/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/pathology , Polyendocrinopathies, Autoimmune/therapy , Syndrome , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL