Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Immunity ; 56(7): 1631-1648.e10, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392737

ABSTRACT

CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell Differentiation , Cell Proliferation , Receptors, Antigen, T-Cell
2.
Blood ; 139(8): 1160-1176, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35201323

ABSTRACT

Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , Antineoplastic Agents, Immunological/pharmacology , Bone Marrow Cells , Membrane Glycoproteins/antagonists & inhibitors , Multiple Myeloma , Neoplasm Proteins/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , HEK293 Cells , Humans , Membrane Glycoproteins/metabolism , Mice, Transgenic , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Xenograft Model Antitumor Assays
3.
Immunity ; 53(4): 824-839.e10, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053331

ABSTRACT

CD8+ T cells within the tumor microenvironment (TME) are exposed to various signals that ultimately determine functional outcomes. Here, we examined the role of the co-activating receptor CD226 (DNAM-1) in CD8+ T cell function. The absence of CD226 expression identified a subset of dysfunctional CD8+ T cells present in peripheral blood of healthy individuals. These cells exhibited reduced LFA-1 activation, altered TCR signaling, and a distinct transcriptomic program upon stimulation. CD226neg CD8+ T cells accumulated in human and mouse tumors of diverse origin through an antigen-specific mechanism involving the transcriptional regulator Eomesodermin (Eomes). Despite similar expression of co-inhibitory receptors, CD8+ tumor-infiltrating lymphocyte failed to respond to anti-PD-1 in the absence of CD226. Immune checkpoint blockade efficacy was hampered in Cd226-/- mice. Anti-CD137 (4-1BB) agonists also stimulated Eomes-dependent CD226 loss that limited the anti-tumor efficacy of this treatment. Thus, CD226 loss restrains CD8+ T cell function and limits the efficacy of cancer immunotherapy.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , T-Box Domain Proteins/immunology , Animals , Humans , Immune Checkpoint Inhibitors/immunology , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
4.
Blood ; 132(16): 1689-1694, 2018 10 18.
Article in English | MEDLINE | ID: mdl-29986909

ABSTRACT

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , Multiple Myeloma/prevention & control , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/etiology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology
5.
Immunity ; 45(4): 774-787, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27742544

ABSTRACT

The transcription factor Foxo3 plays a crucial role in myeloid cell function but its role in lymphoid cells remains poorly defined. Here, we have shown that Foxo3 expression was increased after T cell receptor engagement and played a specific role in the polarization of CD4+ T cells toward pathogenic T helper 1 (Th1) cells producing interferon-γ (IFN-γ) and granulocyte monocyte colony stimulating factor (GM-CSF). Consequently, Foxo3-deficient mice exhibited reduced susceptibility to experimental autoimmune encephalomyelitis. At the molecular level, we identified Eomes as a direct target gene for Foxo3 in CD4+ T cells and we have shown that lentiviral-based overexpression of Eomes in Foxo3-deficient CD4+ T cells restored both IFN-γ and GM-CSF production. Thus, the Foxo3-Eomes pathway is central to achieve the complete specialized gene program required for pathogenic Th1 cell differentiation and development of neuroinflammation.


Subject(s)
Cell Differentiation/physiology , Forkhead Box Protein O3/metabolism , Interleukin-1/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Th1 Cells/pathology , Transcription Factors/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cell Line , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Forkhead Box Protein O3/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Box Domain Proteins/immunology , Th1 Cells/immunology
6.
Brain ; 139(Pt 5): 1433-46, 2016 05.
Article in English | MEDLINE | ID: mdl-27000832

ABSTRACT

Polyspecific T cells recognizing multiple distinct self-antigens have been identified in multiple sclerosis and other organ-specific autoimmune diseases, but their pathophysiological relevance remains undetermined. Using a mouse model of multiple sclerosis, we show that autoimmune encephalomyelitis induction is strictly dependent on reactivation of pathogenic T cells by a peptide (35-55) derived from myelin oligodendrocyte glycoprotein (MOG). This disease-inducing response wanes after onset. Strikingly, the progression of disease is driven by the in situ activation and expansion of a minority of MOG35-55-specific T cells that also recognize neurofilament-medium (NF-M)15-35, an intermediate filament protein expressed in neurons. This mobilization of bispecific T cells is critical for disease progression as adoptive transfer of NF-M15-35/MOG35-55 bispecific T cell lines caused full-blown disease in wild-type but not NF-M-deficient recipients. Moreover, specific tolerance through injection of NF-M15-35 peptide at the peak of disease halted experimental autoimmune encephalomyelitis progression. Our findings highlight the importance of polyspecific autoreactive T cells in the aggravation and perpetuation of central nervous system autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Peptide Fragments/immunology
7.
J Immunol ; 193(7): 3267-77, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25135834

ABSTRACT

The recognition of multiple ligands by a single TCR is an intrinsic feature of T cell biology, with important consequences for physiological and pathological processes. Polyspecific T cells targeting distinct self-antigens have been identified in healthy individuals as well as in the context of autoimmunity. We have previously shown that the 2D2 TCR recognizes the myelin oligodendrocyte glycoprotein epitope (MOG)35-55 as well as an epitope within the axonal protein neurofilament medium (NF-M15-35) in H-2(b) mice. In this study, we assess whether this cross-reactivity is a common feature of the MOG35-55-specific T cell response. To this end, we analyzed the CD4 T cell response of MOG35-55-immunized C57BL/6 mice for cross-reactivity with NF-M15-35. Using Ag recall responses, we established that an important proportion of MOG35-55-specific CD4 T cells also responded to NF-M15-35 in all mice tested. To study the clonality of this response, we analyzed 22 MOG35-55-specific T cell hybridomas expressing distinct TCR. Seven hybridomas were found to cross-react with NF-M15-35. Using an alanine scan of NF-M18-30 and an in silico predictive model, we dissected the molecular basis of cross-reactivity between MOG35-55 and NF-M15-35. We established that NF-M F24, R26, and V27 proved important TCR contacts. Strikingly, the identified TCR contacts are conserved within MOG38-50. Our data indicate that due to linear sequence homology, part of the MOG35-55-specific T cell repertoire of all C57BL/6 mice also recognizes NF-M15-35, with potential implications for CNS autoimmunity.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Myelin Sheath/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurofilament Proteins/immunology , Receptors, Antigen/immunology , Animals , Autoantigens/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , CD4-Positive T-Lymphocytes/pathology , Cross Reactions/genetics , Cross Reactions/immunology , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin-Oligodendrocyte Glycoprotein/genetics , Neurofilament Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Antigen/genetics
8.
J Immunol ; 189(6): 3140-9, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22904307

ABSTRACT

An increase in IL-17-producing CD8+ T (Tc17) cells has been reported in the peripheral blood of children with recent onset type 1 diabetes (T1D), but their contribution to disease pathogenesis is still unknown. To directly study the pathogenic potential of ß cell-specific Tc17 cells, we used an experimental model of T1D based on the expression of the neo-self Ag hemagglutinin (HA) in the ß cells of the pancreas. When transferred alone, the IL-17-producing HA-specific CD8+ T cells homed to the pancreatic lymph nodes without causing any pancreatic infiltration or tissue destruction. When transferred together with small numbers of diabetogenic HA-specific CD4+ T cells, a strikingly different phenotype developed. Under these conditions, Tc17 cells sustained disease progression, driving the destruction of ß-islet cells, causing hyperglycemia and ultimately death. Disease progression did not correlate with functional or numerical alterations among the HA-specific CD4+ T cells. Rather, the transferred CD8+ T cells accumulated in the pancreatic islets and a considerable fraction converted, under the control of IL-12, to an IFN-γ-producing phenotype. Our data indicate that Tc17 cells are not diabetogenic but can potentiate a Th1-mediated disease. Plasticity of the Tc17 lineage is associated with transition to overt disease in this experimental model of T1D.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Interleukin-17/biosynthesis , Th1 Cells/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Progression , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-17/metabolism , Interleukin-17/physiology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Inbred BALB C , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/pathology , Th17 Cells/transplantation , Up-Regulation/immunology
9.
J Immunol ; 189(4): 1680-8, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22802417

ABSTRACT

The pathogenesis of multiple sclerosis requires the participation of effector neuroantigen-specific T cells. Thus, T cell targeting has been proposed as a promising therapeutic strategy. However, the mechanism underlying effective disease prevention following T cell targeting remains incompletely known. We found, using several TCR-transgenic strains, that CD4 blockade is effective in preventing experimental autoimmune encephalopathy and in treating mice after the disease onset. The mechanism does not rely on direct T cell depletion, but the anti-CD4 mAb prevents the proliferation of naive neuroantigen-specific T cells, as well as acquisition of effector Th1 and Th17 phenotypes. Simultaneously, the mAb favors peripheral conversion of Foxp3(+) regulatory T cells. Pre-existing effector cells, or neuroantigen-specific cells that undergo cell division despite the presence of anti-CD4, are committed to apoptosis. Therefore, protection from experimental autoimmune encephalopathy relies on a combination of dominant mechanisms grounded on regulatory T cell induction and recessive mechanisms based on apoptosis of neuropathogenic cells. We anticipate that the same mechanisms may be implicated in other T cell-mediated autoimmune diseases that can be treated or prevented with Abs targeting T cell molecules, such as CD4 or CD3.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4 Antigens/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Proc Natl Acad Sci U S A ; 106(15): 6238-43, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19325124

ABSTRACT

T cells differentiate into functionally distinct effector subsets in response to pathogen encounter. Cells of the innate immune system direct this process; CD1d-restricted invariant natural killer T (iNKT) cells, for example, can either promote or inhibit Th(1) and Th(2) responses. Recently, a new subset of CD4(+) T helper cells, called Th(17), was identified that is implicated in mucosal immunity and autoimmune disorders. To investigate the influence of iNKT cells on the differentiation of naïve T cells we used an adoptive transfer model of traceable antigen-specific CD4(+) T cells. Transferred naïve CD25(-)CD62L(+) CD4(+) T cells were primed by antigen immunization of the recipient mice, permitting their expansion and Th(17) differentiation. This study establishes that in vivo activation of iNKT cells during T-cell priming impedes the commitment of naïve T cells to the Th(17) lineage. In vivo cytokine neutralization experiments revealed a role for IL-4, IL-10, and IFN-gamma in the iNKT-cell-mediated regulation of T-cell lineage development. Moreover, by comparing IL-17 production by antigen-experienced T cells from unmanipulated wild-type mice and iNKT-cell-deficient mice, we demonstrate an enhanced Th(17) response in mice lacking iNKT cells. This invigorated Th(17) response reverts to physiological levels when iNKT cells are introduced into Jalpha18(-/-) mice by adoptive transfer, indicating that iNKT cells control the Th(17) compartment at steady state. We conclude that iNKT cells play an important role in limiting development of the Th(17) lineage and suggest that iNKT cells provide a natural barrier against Th(17) responses.


Subject(s)
Cell Lineage/immunology , Interleukin-17/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Separation , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-17/biosynthesis , Interleukin-4/biosynthesis , Interleukin-4/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...