Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 20(1): 121-131, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33277440

RESUMO

TIGIT is an immune checkpoint inhibitor expressed by effector CD4+ and CD8+ T cells, NK cells, and regulatory T cells (Tregs). Inhibition of TIGIT-ligand binding using antagonistic anti-TIGIT mAbs has shown in vitro potential to restore T-cell function and therapeutic efficacy in murine tumor models when combined with an anti-PD(L)-1 antibody. In the current work, we demonstrate broader TIGIT expression than previously reported in healthy donors and patients with cancer with expression on γδ T cells, particularly in CMV-seropositive donors, and on tumor cells from hematologic malignancies. Quantification of TIGIT density revealed tumor-infiltrating Tregs as the population expressing the highest receptor density. Consequently, the therapeutic potential of anti-TIGIT mAbs might be wider than the previously described anti-PD(L)-1-like restoration of αß T-cell function. CD155 also mediated inhibition of γδ T cells, an immune population not previously described to be sensitive to TIGIT inhibition, which could be fully prevented via use of an antagonistic anti-TIGIT mAb (EOS-448). In PBMCs from patients with cancer, as well as in tumor-infiltrating lymphocytes from mice, the higher TIGIT expression in Tregs correlated with strong antibody-dependent killing and preferential depletion of this highly immunosuppressive population. Accordingly, the ADCC/ADCP-enabling format of the anti-TIGIT mAb had superior antitumor activity, which was dependent upon Fcγ receptor engagement. In addition, the anti-TIGIT mAb was able to induce direct killing of TIGIT-expressing tumor cells both in human patient material and in animal models, providing strong rationale for therapeutic intervention in hematologic malignancies. These findings reveal multiple therapeutic opportunities for anti-TIGIT mAbs in cancer therapeutics.


Assuntos
Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/farmacologia , Citotoxicidade Imunológica , Receptores Imunológicos/antagonistas & inibidores , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos CD/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/metabolismo , Depleção Linfocítica , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de IgG/metabolismo , Receptores Imunológicos/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos
2.
Sci Transl Med ; 7(284): 284ra56, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904740

RESUMO

Regulatory T cells (Tregs) are essential to prevent autoimmunity, but excessive Treg function contributes to cancer progression by inhibiting antitumor immune responses. Tregs exert contact-dependent inhibition of immune cells through the production of active transforming growth factor-ß1 (TGF-ß1). On the Treg cell surface, TGF-ß1 is in an inactive form bound to membrane protein GARP and then activated by an unknown mechanism. We demonstrate that GARP is involved in this activation mechanism. Two anti-GARP monoclonal antibodies were generated that block the production of active TGF-ß1 by human Tregs. These antibodies recognize a conformational epitope that requires amino acids GARP137-139 within GARP/TGF-ß1 complexes. A variety of antibodies recognizing other GARP epitopes did not block active TGF-ß1 production by Tregs. In a model of xenogeneic graft-versus-host disease in NSG mice, the blocking antibodies inhibited the immunosuppressive activity of human Tregs. These antibodies may serve as therapeutic tools to boost immune responses to infection or cancer via a mechanism of action distinct from that of currently available immunomodulatory antibodies. Used alone or in combination with tumor vaccines or antibodies targeting the CTLA4 or PD1/PD-L1 pathways, blocking anti-GARP antibodies may improve the efficiency of cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/química , Imunossupressores/química , Proteínas de Membrana/química , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/química , Animais , Autoimunidade , Epitopos/química , Doença Enxerto-Hospedeiro , Humanos , Proteínas de Membrana/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Ligação Proteica , Conformação Proteica , Fator de Crescimento Transformador beta1/metabolismo
3.
PLoS One ; 8(9): e76186, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098777

RESUMO

GARP is a transmembrane protein present on stimulated human regulatory T lymphocytes (Tregs), but not on other T lymphocytes (Th cells). It presents the latent form of TGF-ß1 on the Treg surface. We report here that GARP favors the cleavage of the pro-TGF-ß1 precursor and increases the amount of secreted latent TGF-ß1. Stimulated Tregs, which naturally express GARP, and Th cells transfected with GARP secrete a previously unknown form of latent TGF-ß1 that is disulfide-linked to GARP. These GARP/TGF-ß1 complexes are possibly shed from the T cell surface. Secretion of GARP/TGF-ß1 complexes was not observed with transfected 293 cells and may thus be restricted to the T cell lineage. We conclude that in stimulated human Tregs, GARP not only displays latent TGF-ß1 at the cell surface, but also increases its secretion by forming soluble disulfide-linked complexes. Moreover, we identified six microRNAs (miRNAs) that are expressed at lower levels in Treg than in Th clones and that target a short region of the GARP 3' UTR. In transfected Th cells, the presence of this region decreased GARP levels, cleavage of pro-TGF-ß1, and secretion of latent TGF-ß1.


Assuntos
Proteínas de Membrana/imunologia , MicroRNAs/metabolismo , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/metabolismo , Western Blotting , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Imunoprecipitação , Luciferases , Proteínas de Membrana/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/metabolismo
4.
J Neurochem ; 113(4): 819-25, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20180881

RESUMO

Neuroblastoma is a sympathetic nervous system tumour whose degree of malignancy, prognosis and therapy resistance has been associated with the amplification of MYCN oncogene. However, the molecular pathway responsible for such resistance is unknown. To contribute addressing this issue, in this study, we have compared the vulnerability of four human neuroblastoma cell lines differentially amplifying MYCN, namely SK-N-BE-2 and IMR-32 (MYCN-amplified cells) and SH-SY5Y and SK-N-SH (MCYN-non-amplified cells), to H(2)O(2)-mediated apoptotic death. We found that the high resistance of the MYCN-amplified neuroblastoma cells against oxidative damage can be accounted for by their greater expression of both the mRNA and protein of the catalytic subunit of glutamate-cysteine ligase (GCL(cat)), the rate-limiting step in GSH biosynthesis. Furthermore, we found that MYCN directly binds to an E-box containing GCL(cat) promoter and that over-expression of MYCN in MYCN-non-amplified cells stimulated GCL(cat) expression and provided resistance to oxidative damage; whereas knock down of MYCN in MYCN-amplified cells decreased GCL(cat) expression and sensitized them to oxidative damage. Finally, GCL(cat) knock down enhanced the vulnerability of MYCN-amplified cells to oxidative damage. These results demonstrate that regulation of GCL(cat) by MYCN accounts for the survival of neuroblastoma cells against oxidative damage, and suggest that GCL should be considered a potential therapeutic target for the treatment of MYCN-amplified neuroblastoma.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Glutamato-Cisteína Ligase/genética , Neuroblastoma/enzimologia , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Estresse Oxidativo/genética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação para Baixo/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/química , Humanos , Peróxido de Hidrogênio/toxicidade , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/tratamento farmacológico , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ativação Transcricional/fisiologia , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA