Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38551501

RESUMO

PURPOSE: Medulloblastoma (MB), the most common childhood malignant brain tumor, has a poor prognosis in about 30% of patients. The current standard of care, which includes surgery, radiation and chemotherapy, is often responsible for cognitive, neurologic and endocrine side effects. We investigated whether chimeric antigen receptor (CAR) T-cells directed towards the disialoganglioside GD2 can represent a potentially more effective treatment with reduced long-term side effects. EXPERIMENTAL DESIGN: GD2 expression was evaluated on primary tumor biopsies of MB children by flow-cytometry. GD2 expression in MB cells was evaluated also in response to an EZH2 inhibitor (Tazemetostat). In in vitro, as well as in in vivo models, GD2+MB cells were targeted by a CAR-GD2.CD28.4-1BBζ (CAR.GD2)-T construct, including the suicide gene inducible-caspase-9. RESULTS: GD2 was expressed in 73.17% of MB tumors. The SHH and G4 subtypes expressed the highest levels of GD2, while the WNT subtype the lowest. In in-vitro co-culture assays, CAR.GD2 T-cells were able to kill GD2+MB cells. Pre-treatment with Tazemetostat upregulated GD2 expression, sensitizing GD2dimMB cells to CAR.GD2 T-cells cytotoxic activity. In orthotopic mouse models of MB, intravenously injected CAR.GD2 T-cells significantly controlled tumor growth, prolonging overall survival of treated mice. Moreover, the dimerizing drug AP1903 was able to cross the murine blood brain barrier and to eliminate both blood circulating and tumor infiltrating CAR.GD2 T-cells. CONCLUSIONS: Our experimental data indicate the feasibility of CAR.GD2 T-cell therapy. A phase I/II clinical trial will be conducted to evaluate the safety and therapeutic efficacy of CAR.GD2 therapy in high-risk MB patients.

2.
Nat Commun ; 14(1): 3423, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37296093

RESUMO

Chimeric antigen receptor T (CAR-T) cell therapy may achieve long-lasting remission in patients with B-cell malignancies not responding to conventional therapies. However, potentially severe and hard-to-manage side effects, including cytokine release syndrome (CRS), neurotoxicity and macrophage activation syndrome, and the lack of pathophysiological experimental models limit the applicability and development of this form of therapy. Here we present a comprehensive humanized mouse model, by which we show that IFNγ neutralization by the clinically approved monoclonal antibody, emapalumab, mitigates severe toxicity related to CAR-T cell therapy. We demonstrate that emapalumab reduces the pro-inflammatory environment in the model, thus allowing control of severe CRS and preventing brain damage, characterized by multifocal hemorrhages. Importantly, our in vitro and in vivo experiments show that IFNγ inhibition does not affect the ability of CD19-targeting CAR-T (CAR.CD19-T) cells to eradicate CD19+ lymphoma cells. Thus, our study provides evidence that anti-IFNγ treatment might reduce immune related adverse effect without compromising therapeutic success and provides rationale for an emapalumab-CAR.CD19-T cell combination therapy in humans.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Camundongos , Animais , Humanos , Imunoterapia Adotiva/efeitos adversos , Linfócitos B , Interferon gama , Neoplasias/etiologia , Síndrome da Liberação de Citocina , Antígenos CD19 , Terapia Baseada em Transplante de Células e Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...