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
País de afiliação
Intervalo de ano de publicação
1.
Blood ; 122(20): 3461-72, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24016461

RESUMO

Genetically targeted T cells promise to solve the feasibility and efficacy hurdles of adoptive T-cell therapy for cancer. Selecting a target expressed in multiple-tumor types and that is required for tumor growth would widen disease indications and prevent immune escape caused by the emergence of antigen-loss variants. The adhesive receptor CD44 is broadly expressed in hematologic and epithelial tumors, where it contributes to the cancer stem/initiating phenotype. In this study, silencing of its isoform variant 6 (CD44v6) prevented engraftment of human acute myeloid leukemia (AML) and multiple myeloma (MM) cells in immunocompromised mice. Accordingly, T cells targeted to CD44v6 by means of a chimeric antigen receptor containing a CD28 signaling domain mediated potent antitumor effects against primary AML and MM while sparing normal hematopoietic stem cells and CD44v6-expressing keratinocytes. Importantly, in vitro activation with CD3/CD28 beads and interleukin (IL)-7/IL-15 was required for antitumor efficacy in vivo. Finally, coexpressing a suicide gene enabled fast and efficient pharmacologic ablation of CD44v6-targeted T cells and complete rescue from hyperacute xenogeneic graft-versus-host disease modeling early and generalized toxicity. These results warrant the clinical investigation of suicidal CD44v6-targeted T cells in AML and MM.


Assuntos
Antígenos de Neoplasias/imunologia , Receptores de Hialuronatos/imunologia , Imunoterapia Adotiva , Leucemia Mieloide Aguda/terapia , Terapia de Alvo Molecular , Mieloma Múltiplo/terapia , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos CD28/imunologia , Complexo CD3/imunologia , Linhagem Celular Tumoral/imunologia , Linhagem Celular Tumoral/transplante , Citotoxicidade Imunológica , Genes Transgênicos Suicidas , Doença Enxerto-Hospedeiro/terapia , Humanos , Receptores de Hialuronatos/genética , Interleucina-15/imunologia , Interleucina-15/farmacologia , Interleucina-7/imunologia , Interleucina-7/farmacologia , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Leucemia Mielomonocítica Aguda/imunologia , Leucemia Mielomonocítica Aguda/patologia , Leucemia Mielomonocítica Aguda/terapia , Ativação Linfocitária , Camundongos , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Estrutura Terciária de Proteína , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nat Commun ; 12(1): 4559, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34315896

RESUMO

Activating mutations in the BRAF-MAPK pathway have been reported in histiocytoses, hematological inflammatory neoplasms characterized by multi-organ dissemination of pro-inflammatory myeloid cells. Here, we generate a humanized mouse model of transplantation of human hematopoietic stem and progenitor cells (HSPCs) expressing the activated form of BRAF (BRAFV600E). All mice transplanted with BRAFV600E-expressing HSPCs succumb to bone marrow failure, displaying myeloid-restricted hematopoiesis and multi-organ dissemination of aberrant mononuclear phagocytes. At the basis of this aggressive phenotype, we uncover the engagement of a senescence program, characterized by DNA damage response activation and a senescence-associated secretory phenotype, which affects also non-mutated bystander cells. Mechanistically, we identify TNFα as a key determinant of paracrine senescence and myeloid-restricted hematopoiesis and show that its inhibition dampens inflammation, delays disease onset and rescues hematopoietic defects in bystander cells. Our work establishes that senescence in the human hematopoietic system links oncogene-activation to the systemic inflammation observed in histiocytic neoplasms.


Assuntos
Senescência Celular , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Histiocitose/patologia , Inflamação/patologia , Células Mieloides/patologia , Oncogenes , Animais , Medula Óssea/patologia , Pontos de Checagem do Ciclo Celular/genética , Senescência Celular/genética , Doença Crônica , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Histiocitose/complicações , Humanos , Inflamação/complicações , Lentivirus/genética , Camundongos , Mutação/genética , Comunicação Parácrina , Análise de Componente Principal , Proteínas Proto-Oncogênicas B-raf/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
3.
Nat Commun ; 9(1): 2896, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30042420

RESUMO

Immunotherapy is emerging as a new pillar of cancer treatment with potential to cure. However, many patients still fail to respond to these therapies. Among the underlying factors, an immunosuppressive tumor microenvironment (TME) plays a major role. Here we show that monocyte-mediated gene delivery of IFNα inhibits leukemia in a mouse model. IFN gene therapy counteracts leukemia-induced expansion of immunosuppressive myeloid cells and imposes an immunostimulatory program to the TME, as shown by bulk and single-cell transcriptome analyses. This reprogramming promotes T-cell priming and effector function against multiple surrogate tumor-specific antigens, inhibiting leukemia growth in our experimental model. Durable responses are observed in a fraction of mice and are further increased combining gene therapy with checkpoint blockers. Furthermore, IFN gene therapy strongly enhances anti-tumor activity of adoptively transferred T cells engineered with tumor-specific TCR or CAR, overcoming suppressive signals in the leukemia TME. These findings warrant further investigations on the potential development of our gene therapy strategy towards clinical testing.


Assuntos
Antígenos de Neoplasias/imunologia , Terapia Genética/métodos , Imunidade/imunologia , Interferons/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Regulação Leucêmica da Expressão Gênica , Imunidade/genética , Imunoterapia Adotiva/métodos , Interferons/genética , Interferons/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante , Microambiente Tumoral/genética
4.
Front Immunol ; 9: 507, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29619024

RESUMO

Chimeric antigen receptor (CAR)-T cell immunotherapy is at the forefront of innovative cancer therapeutics. However, lack of standardization of cellular products within the same clinical trial and lack of harmonization between different trials have hindered the clear identification of efficacy and safety determinants that should be unveiled in order to advance the field. With the aim of facilitating the isolation and in vivo tracking of CAR-T cells, we here propose the inclusion within the CAR molecule of a novel extracellular spacer based on the low-affinity nerve-growth-factor receptor (NGFR). We screened four different spacer designs using as target antigen the CD44 isoform variant 6 (CD44v6). We successfully generated NGFR-spaced CD44v6 CAR-T cells that could be efficiently enriched with clinical-grade immuno-magnetic beads without negative consequences on subsequent expansion, immuno-phenotype, in vitro antitumor reactivity, and conditional ablation when co-expressing a suicide gene. Most importantly, these cells could be tracked with anti-NGFR monoclonal antibodies in NSG mice, where they expanded, persisted, and exerted potent antitumor effects against both high leukemia and myeloma burdens. Similar results were obtained with NGFR-enriched CAR-T cells specific for CD19 or CEA, suggesting the universality of this strategy. In conclusion, we have demonstrated that the incorporation of the NGFR marker gene within the CAR sequence allows for a single molecule to simultaneously work as a therapeutic and selection/tracking gene. Looking ahead, NGFR spacer enrichment might allow good manufacturing procedures-manufacturing of standardized CAR-T cell products with high therapeutic potential, which could be harmonized in different clinical trials and used in combination with a suicide gene for future application in the allogeneic setting.


Assuntos
Imunoterapia Adotiva , Proteínas do Tecido Nervoso/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de Fator de Crescimento Neural/imunologia , Linfócitos T/imunologia , Timidina Quinase/genética , Animais , Linhagem Celular Tumoral , Genes Transgênicos Suicidas , Receptores de Hialuronatos/imunologia , Leucemia/terapia , Camundongos , Mieloma Múltiplo/terapia , Proteínas do Tecido Nervoso/genética , Receptores de Antígenos Quiméricos/genética , Receptores de Fator de Crescimento Neural/genética
5.
Nat Med ; 24(6): 739-748, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29808007

RESUMO

In the clinic, chimeric antigen receptor-modified T (CAR T) cell therapy is frequently associated with life-threatening cytokine-release syndrome (CRS) and neurotoxicity. Understanding the nature of these pathologies and developing treatments for them are hampered by the lack of appropriate animal models. Herein, we describe a mouse model recapitulating key features of CRS and neurotoxicity. In humanized mice with high leukemia burden, CAR T cell-mediated clearance of cancer triggered high fever and elevated IL-6 levels, which are hallmarks of CRS. Human monocytes were the major source of IL-1 and IL-6 during CRS. Accordingly, the syndrome was prevented by monocyte depletion or by blocking IL-6 receptor with tocilizumab. Nonetheless, tocilizumab failed to protect mice from delayed lethal neurotoxicity, characterized by meningeal inflammation. Instead, the IL-1 receptor antagonist anakinra abolished both CRS and neurotoxicity, resulting in substantially extended leukemia-free survival. These findings offer a therapeutic strategy to tackle neurotoxicity and open new avenues to safer CAR T cell therapies.


Assuntos
Imunoterapia Adotiva/efeitos adversos , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Monócitos/metabolismo , Neurotoxinas/toxicidade , Receptores de Antígenos Quiméricos/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Linhagem Celular Tumoral , Células-Tronco Hematopoéticas/metabolismo , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Leucemia/imunologia , Leucemia/patologia , Camundongos , Síndrome
6.
Methods Mol Biol ; 1393: 127-32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27033222

RESUMO

Hematopoietic stem cell transplantation (HSCT) from an allogeneic donor is an effective form of cancer immunotherapy, especially for acute leukemias. HSCT is however frequently complicated by the occurrence of graft-versus-host disease (GVHD). Immunocompromised mice infused with human T cells often develop a clinical syndrome resembling human GVHD (xenogeneic or X-GVHD). Herein, we describe a method for inducing X-GVHD in a highly reproducible manner. Given the human nature of immune effectors, this xenogeneic model can be routinely adopted for screening the efficacy of new treatments for GVHD.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Leucócitos Mononucleares/imunologia , Animais , Modelos Animais de Doenças , Humanos , Hospedeiro Imunocomprometido , Leucócitos Mononucleares/transplante , Camundongos Endogâmicos NOD , Camundongos SCID
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA