Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 193(8): 3835-41, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25281753

RESUMO

Programmed death ligand 1 (PD-L1, also known as B7 homolog 1 or CD274) is a major obstacle to antitumor immunity because it tolerizes/anergizes tumor-reactive T cells by binding to its receptor programmed death-1 (CD279), renders tumor cells resistant to CD8(+) T cell- and FasL-mediated lysis, and tolerizes T cells by reverse signaling through T cell-expressed CD80. PD-L1 is abundant in the tumor microenvironment, where it is expressed by many malignant cells, as well as by immune cells and vascular endothelial cells. The critical role of PD-L1 in obstructing antitumor immunity has been demonstrated in multiple animal models and in recent clinical trials. This article reviews the mechanisms by which PD-L1 impairs antitumor immunity and discusses established and experimental strategies for maintaining T cell activation in the presence of PD-L1-expressing cells in the tumor microenvironment.


Assuntos
Antígeno B7-1/imunologia , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Apoptose/imunologia , Proteína Ligante Fas/imunologia , Humanos , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia
2.
J Immunol ; 191(5): 2829-36, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23918985

RESUMO

Many tumor cells escape anti-tumor immunity through their expression of programmed death ligand-1 (PDL1 or B7-H1), which interacts with T cell-expressed PD1 and results in T cell apoptosis. We previously reported that transfection of human tumor cells with a membrane-bound form of the human costimulatory molecule CD80 prevented PD1 binding and restored T cell activation. We now report that a membrane-bound form of murine CD80 similarly reduces PDL1-PD1-mediated suppression by mouse tumor cells and that a soluble protein consisting of the extracellular domains of human or mouse CD80 fused to the Fc domain of IgG1 (CD80-Fc) overcomes PDL1-mediated suppression by human and mouse tumor cells, respectively. T cell activation experiments with human and mouse tumor cells indicate that CD80-Fc facilitates T cell activation by binding to PDL1 to inhibit PDL1-PD1 interactions and by costimulating through CD28. CD80-Fc is more effective in preventing PD1-PDL1-mediated suppression and restoring T cell activation compared with treatment with mAb to either PD1 or PDL1. These studies identify CD80-Fc as an alternative and potentially more efficacious therapeutic agent for overcoming PDL1-induced immune suppression and facilitating tumor-specific immunity.


Assuntos
Antígeno B7-1/imunologia , Antígeno B7-H1/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Animais , Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/metabolismo , Linfócitos T/metabolismo , Transfecção
3.
J Immunol ; 186(12): 6822-9, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21555531

RESUMO

Programmed death ligand 1 (PDL1, or B7-H1) is expressed constitutively or is induced by IFN-γ on the cell surface of most human cancer cells and acts as a "molecular shield" by protecting tumor cells from T cell-mediated destruction. Using seven cell lines representing four histologically distinct solid tumors (lung adenocarcinoma, mammary carcinoma, cutaneous melanoma, and uveal melanoma), we demonstrate that transfection of human tumor cells with the gene encoding the costimulatory molecule CD80 prevents PDL1-mediated immune suppression by tumor cells and restores T cell activation. Mechanistically, CD80 mediates its effects through its extracellular domain, which blocks the cell surface expression of PDL1 but does not prevent intracellular expression of PDL1 protein. These studies demonstrate a new role for CD80 in facilitating antitumor immunity and suggest new therapeutic avenues for preventing tumor cell PDL1-induced immune suppression.


Assuntos
Antígenos CD/imunologia , Antígeno B7-1/imunologia , Linfócitos T/imunologia , Antígeno B7-1/administração & dosagem , Antígeno B7-1/genética , Antígeno B7-H1 , Linhagem Celular Tumoral , Expressão Gênica/efeitos dos fármacos , Humanos , Imunoterapia/métodos , Ativação Linfocitária/imunologia , Transfecção
4.
Oncoimmunology ; 9(1): 1757360, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32923113

RESUMO

Recent advances in novel immune strategies, particularly chimeric antigen receptor (CAR)-bearing T-cells, have shown limited efficacy against glioblastoma (GBM) in clinical trials. We currently have an incomplete understanding of how these emerging therapies integrate with the current standard of care, specifically radiation therapy (RT). Additionally, there is an insufficient number of preclinical studies monitoring these therapies with high spatiotemporal resolution. To address these limitations, we report the first longitudinal fluorescence-based intravital microscopy imaging of CAR T-cells within an orthotopic GBM preclinical model to illustrate the necessity of RT for complete therapeutic response. Additionally, we detail the first usage of murine-derived CAR T-cells targeting the disialoganglioside GD2 in an immunocompetent tumor model. Cell culture assays demonstrated substantial GD2 CAR T-cell-mediated killing of murine GBM cell lines SB28 and GL26 induced to overexpress GD2. Complete antitumor response in advanced syngeneic orthotopic models of GBM was achieved only when a single intravenous dose of GD2 CAR T-cells was following either sub-lethal whole-body irradiation or focal RT. Intravital microscopy imaging successfully visualized CAR T-cell homing and T-cell mediated apoptosis of tumor cells in real-time within the tumor stroma. Findings indicate that RT allows for rapid CAR T-cell extravasation from the vasculature and expansion within the tumor microenvironment, leading to a more robust and lasting immunologic response. These exciting results highlight potential opportunities to improve intravenous adoptive T-cell administration in the treatment of GBM through concurrent RT. Additionally, they emphasize the need for advancements in immunotherapeutic homing to and extravasation through the tumor microenvironment.


Assuntos
Glioblastoma , Animais , Linhagem Celular Tumoral , Glioblastoma/radioterapia , Imunoterapia Adotiva , Microscopia Intravital , Camundongos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
5.
SLAS Discov ; 23(7): 603-612, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29634393

RESUMO

High-throughput flow cytometry is an attractive platform for the analysis of adoptive cellular therapies such as chimeric antigen receptor T cell therapy (CAR-T) because it allows for the concurrent measurement of T cell-dependent cellular cytotoxicity (TDCC) and the functional characterization of engineered T cells with respect to percentage of CAR transduction, T cell phenotype, and measurement of T cell function such as activation in a single assay. The use of adherent tumor cell lines can be challenging in these flow-based assays. Here, we present the development of a high-throughput flow-based assay to measure TDCC for a CAR-T construct co-cultured with multiple adherent tumor cell lines. We describe optimal assay conditions (such as adherent cell dissociation techniques to minimize impact on cell viability) that result in robust cytotoxicity assays. In addition, we report on the concurrent use of T cell transduction and activation antibody panels (CD25) that provide further dissection of engineered T cell function. In conclusion, we present the development of a high-throughput flow cytometry method allowing for in vitro interrogation of solid tumor, targeting CAR-T cell-mediated cytotoxicity, CAR transduction, and engineered T cell characterization in a single assay.


Assuntos
Citotoxicidade Imunológica , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo/métodos , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T/imunologia , Transdução Genética
6.
Cancer Immunol Res ; 2(7): 610-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24819296

RESUMO

Tumor cells use various methods of immunosuppression to overcome antitumor immunity. One such method is that of programmed death ligand-1 (PD-L1 or B7-H1), which upon binding its receptor PD-1 on T cells triggers apoptotic death of the activated T cells. Overexpression of the costimulatory molecule CD80 on PD-L1(+) tumor cells, or inclusion of a soluble form of CD80 (CD80-Fc), maintains the activation of PD-1(+)-activated T cells. Using T cells from CD28-deficient mice and antibodies to block CD28 on human T cells, we now report that a soluble form of CD80 mediates this effect by simultaneously neutralizing PD-1-PD-L1-mediated immunosuppression and by providing CD80-CD28 costimulation, and is more effective than antibodies to PD-L1 or PD-1 in maintaining IFNγ production by PD-1(+) activated T cells. Therefore, soluble CD80 may be a more effective therapeutic than these checkpoint antibodies for facilitating the development and maintenance of antitumor immunity because it has the dual functions of preventing PD-L1-mediated immunosuppression and simultaneously delivering the second signal for T-cell activation.


Assuntos
Antígeno B7-1/imunologia , Antígeno B7-H1/imunologia , Melanoma/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD28/deficiência , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas , Evasão Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA