Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Mol Ther Oncol ; 32(3): 200839, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39072290

RÉSUMÉ

Targeting immune checkpoint receptors on T cells is a common cancer treatment strategy. Frequently, this is accomplished through antibodies targeting the ligand of inhibitory co-receptors. Blocking the immune checkpoint PD-1 binding to its ligands PD-L1 and PD-L2 prevents downstream signaling and enhances anti-tumor T cell responses. This approach improves cancer patients' outcomes. However, only one-third of the patients respond to these treatments. To better understand the mechanism of anti-PD-1 antibodies, we explored the location of PD-1 within the immune synapse. Surprisingly, we discovered that anti-PD-1 antibodies, besides blocking the interaction between PD-1 and its ligands, also removed PD-1 from the synapse. We demonstrated a correlation between removing PD-1 from the synapse by anti-PD-1 antibodies and the extent of T cell activation. Interestingly, a short version of the anti-PD-1 antibody, F(ab')2, failed to remove PD-1 from the synapse and activate T cells. Using the syngeneic tumor model, we showed a superior anti-tumor effect of the anti-PD-1 antibody over the shorter version of the same antibody. Our data indicate that anti-PD-1 antibodies activate T cells by removing PD-1 from the synapse, and changing the location of PD-1 or other immune receptors within the immune synapse could serve as an alternative, efficient approach to treat cancer.

2.
Cancer Cell ; 41(7): 1207-1221.e12, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37327789

RÉSUMÉ

The cell-autonomous balance of immune-inhibitory and -stimulatory signals is a critical process in cancer immune evasion. Using patient-derived co-cultures, humanized mouse models, and single-cell RNA-sequencing of patient melanomas biopsied before and on immune checkpoint blockade, we find that intact cancer cell-intrinsic expression of CD58 and ligation to CD2 is required for anti-tumor immunity and is predictive of treatment response. Defects in this axis promote immune evasion through diminished T cell activation, impaired intratumoral T cell infiltration and proliferation, and concurrently increased PD-L1 protein stabilization. Through CRISPR-Cas9 and proteomics screens, we identify and validate CMTM6 as critical for CD58 stability and upregulation of PD-L1 upon CD58 loss. Competition between CD58 and PD-L1 for CMTM6 binding determines their rate of endosomal recycling over lysosomal degradation. Overall, we describe an underappreciated yet critical axis of cancer immunity and provide a molecular basis for how cancer cells balance immune inhibitory and stimulatory cues.


Sujet(s)
Antigène CD274 , Mélanome , Souris , Animaux , Antigène CD274/génétique , Lymphocytes T , Antigènes CD58/composition chimique , Antigènes CD58/métabolisme , Mélanome/génétique , Mélanome/métabolisme , Activation des lymphocytes
3.
JCI Insight ; 7(23)2022 12 08.
Article de Anglais | MEDLINE | ID: mdl-36477361

RÉSUMÉ

NK cell deficiencies (NKD) are a type of primary immune deficiency in which the major immunologic abnormality affects NK cell number, maturity, or function. Since NK cells contribute to immune defense against virally infected cells, patients with NKD experience higher susceptibility to chronic, recurrent, and fatal viral infections. An individual with recurrent viral infections and mild hypogammaglobulinemia was identified to have an X-linked damaging variant in the transcription factor gene ELF4. The variant does not decrease expression but disrupts ELF4 protein interactions and DNA binding, reducing transcriptional activation of target genes and selectively impairing ELF4 function. Corroborating previous murine models of ELF4 deficiency (Elf4-/-) and using a knockdown human NK cell line, we determined that ELF4 is necessary for normal NK cell development, terminal maturation, and function. Through characterization of the NK cells of the proband, expression of the proband's variant in Elf4-/- mouse hematopoietic precursor cells, and a human in vitro NK cell maturation model, we established this ELF4 variant as a potentially novel cause of NKD.


Sujet(s)
Facteurs de transcription , Animaux , Humains , Souris , Protéines de liaison à l'ADN/génétique , Cellules tueuses naturelles , Facteurs de transcription/génétique
4.
Mol Ther Methods Clin Dev ; 27: 380-390, 2022 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-36419471

RÉSUMÉ

The transmembrane adaptor phosphoprotein associated with glycosphingolipid-enriched microdomains 1 (PAG) is phosphorylated in T cells downstream of PD-1 signaling and contributes to the resulting functional inhibition of multiple cellular processes. Furthermore, PAG expression is negatively correlated with survival in multiple human tumors and is a driver of murine tumor growth and immune evasion. Here we develop an antibody that targets the extracellular domain of human PAG, with cross-reactivity to murine PAG. We demonstrate that this antibody binds to extracellular PAG on intact cells and affects T cell activation. Finally, we show that administration of anti-PAG monoclonal antibody in combination with anti-PD-1 antibody to mice bearing MC38 tumors limited tumor growth and enhanced T cell infiltration to tumors.

5.
Front Immunol ; 13: 927265, 2022.
Article de Anglais | MEDLINE | ID: mdl-35911672

RÉSUMÉ

Cancer remains the second leading cause of death in the US, accounting for 25% of all deaths nationwide. Immunotherapy techniques bolster the immune cells' ability to target malignant cancer cells and have brought immense improvements in the field of cancer treatments. One important inhibitory protein in T cells, programmed cell death protein 1 (PD-1), has become an invaluable target for cancer immunotherapy. While anti-PD-1 antibody therapy is extremely successful in some patients, in others it fails or even causes further complications, including cancer hyper-progression and immune-related adverse events. Along with countless translational studies of the PD-1 signaling pathway, there are currently close to 5,000 clinical trials for antibodies against PD-1 and its ligand, PD-L1, around 80% of which investigate combinations with other therapies. Nevertheless, more work is needed to better understand the PD-1 signaling pathway and to facilitate new and improved evidence-based combination strategies. In this work, we consolidate recent discoveries of PD-1 signaling mediators and their therapeutic potential in combination with anti-PD-1/PD-L1 agents. We focus on the phosphatases SHP2 and PTPN2; the kinases ITK, VRK2, GSK-3, and CDK4/6; and the signaling adaptor protein PAG. We discuss their biology both in cancer cells and T cells, with a focus on their role in relation to PD-1 to determine their potential in therapeutic combinations. The literature discussed here was obtained from a search of the published literature and ClinicalTrials.gov with the following key terms: checkpoint inhibition, cancer immunotherapy, PD-1, PD-L1, SHP2, PTPN2, ITK, VRK2, CDK4/6, GSK-3, and PAG. Together, we find that all of these proteins are logical and promising targets for combination therapy, and that with a deeper mechanistic understanding they have potential to improve the response rate and decrease adverse events when thoughtfully used in combination with checkpoint inhibitors.


Sujet(s)
Antigène CD274 , Tumeurs , Glycogen Synthase Kinase 3 , Humains , Immunothérapie/méthodes , Tumeurs/thérapie , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Transduction du signal
6.
Sci Rep ; 9(1): 13825, 2019 09 25.
Article de Anglais | MEDLINE | ID: mdl-31554863

RÉSUMÉ

Immune dysfunction due to microgravity remains a hurdle in the next step of human space exploration. Dendritic cells (DC) represent a critical component of immunity, given their role in the detection of invaders and the subsequent task of activating T cells to respond and eliminate the threat. Upon encounter with microbes, DC undergo a process of maturation, whereby the cells upregulate the expression of surface proteins and secrete cytokines, both required for the optimal activation of CD4+ and CD8+ T cells. In this study, DC were cultured from 2-14 days in a rotary cell culture system, which generates a simulated microgravity (SMG) environment, and then the cells were assessed for maturation status and the capacity to activate T cells. Short-term culture (<72 h) of DC in SMG resulted in an increased expression of surface proteins associated with maturation and interleukin-6 production. Subsequently, the SMG exposed DC were superior to Static control DC at activating both CD4+ and CD8+ T cells as measured by interleukin-2 and interferon-γ production, respectively. However, long-term culture (4-14 d) of DC in SMG reduced the expression of maturation markers and the capacity to activate T cells as compared to Static DC controls.


Sujet(s)
Cellules dendritiques/cytologie , Interféron gamma/métabolisme , Interleukine-2/métabolisme , Interleukine-6/métabolisme , Simulation d'apesanteur/instrumentation , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Techniques de culture cellulaire/instrumentation , Techniques de culture cellulaire/méthodes , Lignée cellulaire , Cellules dendritiques/immunologie , Hybridomes , Souris , Modèles animaux , Simulation d'apesanteur/méthodes
7.
JCI Insight ; 2(23)2017 12 07.
Article de Anglais | MEDLINE | ID: mdl-29212951

RÉSUMÉ

NK cell activation has been shown to be metabolically regulated in vitro; however, the role of metabolism during in vivo NK cell responses to infection is unknown. We examined the role of glycolysis in NK cell function during murine cytomegalovirus (MCMV) infection and the ability of IL-15 to prime NK cells during CMV infection. The glucose metabolism inhibitor 2-deoxy-ᴅ-glucose (2DG) impaired both mouse and human NK cell cytotoxicity following priming in vitro. Similarly, MCMV-infected mice treated with 2DG had impaired clearance of NK-specific targets in vivo, which was associated with higher viral burden and susceptibility to infection on the C57BL/6 background. IL-15 priming is known to alter NK cell metabolism and metabolic requirements for activation. Treatment with the IL-15 superagonist ALT-803 rescued mice from otherwise lethal infection in an NK-dependent manner. Consistent with this, treatment of a patient with ALT-803 for recurrent CMV reactivation after hematopoietic cell transplant was associated with clearance of viremia. These studies demonstrate that NK cell-mediated control of viral infection requires glucose metabolism and that IL-15 treatment in vivo can reduce this requirement and may be effective as an antiviral therapy.


Sujet(s)
Cytotoxicité immunologique/immunologie , Infections à Herpesviridae/prévention et contrôle , Cellules tueuses naturelles/immunologie , Muromegalovirus/isolement et purification , Actines/métabolisme , Animaux , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Glycémie/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cytomegalovirus/physiologie , Infections à cytomégalovirus/traitement médicamenteux , Infections à cytomégalovirus/immunologie , Infections à cytomégalovirus/virologie , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Désoxyglucose/pharmacologie , Femelle , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/immunologie , Granzymes/métabolisme , Infections à Herpesviridae/traitement médicamenteux , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Humains , Interféron gamma/biosynthèse , Interleukine-15/agonistes , Interleukine-15/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Protéines/pharmacologie , Protéines/usage thérapeutique , Protéines de fusion recombinantes , Charge virale/effets des médicaments et des substances chimiques , Activation virale , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE