Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
N Engl J Med ; 370(10): 901-10, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24597865

RESUMO

BACKGROUND: CCR5 is the major coreceptor for human immunodeficiency virus (HIV). We investigated whether site-specific modification of the gene ("gene editing")--in this case, the infusion of autologous CD4 T cells in which the CCR5 gene was rendered permanently dysfunctional by a zinc-finger nuclease (ZFN)--is safe. METHODS: We enrolled 12 patients in an open-label, nonrandomized, uncontrolled study of a single dose of ZFN-modified autologous CD4 T cells. The patients had chronic aviremic HIV infection while they were receiving highly active antiretroviral therapy. Six of them underwent an interruption in antiretroviral treatment 4 weeks after the infusion of 10 billion autologous CD4 T cells, 11 to 28% of which were genetically modified with the ZFN. The primary outcome was safety as assessed by treatment-related adverse events. Secondary outcomes included measures of immune reconstitution and HIV resistance. RESULTS: One serious adverse event was associated with infusion of the ZFN-modified autologous CD4 T cells and was attributed to a transfusion reaction. The median CD4 T-cell count was 1517 per cubic millimeter at week 1, a significant increase from the preinfusion count of 448 per cubic millimeter (P<0.001). The median concentration of CCR5-modified CD4 T cells at 1 week was 250 cells per cubic millimeter. This constituted 8.8% of circulating peripheral-blood mononuclear cells and 13.9% of circulating CD4 T cells. Modified cells had an estimated mean half-life of 48 weeks. During treatment interruption and the resultant viremia, the decline in circulating CCR5-modified cells (-1.81 cells per day) was significantly less than the decline in unmodified cells (-7.25 cells per day) (P=0.02). HIV RNA became undetectable in one of four patients who could be evaluated. The blood level of HIV DNA decreased in most patients. CONCLUSIONS: CCR5-modified autologous CD4 T-cell infusions are safe within the limits of this study. (Funded by the National Institute of Allergy and Infectious Diseases and others; ClinicalTrials.gov number, NCT00842634.).


Assuntos
Linfócitos T CD4-Positivos/transplante , Terapia Genética , Infecções por HIV/terapia , Transfusão de Linfócitos , Receptores CCR5/genética , Adulto , Terapia Antirretroviral de Alta Atividade , Transfusão de Sangue Autóloga , Linfócitos T CD4-Positivos/química , Terapia Combinada , DNA Viral/sangue , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , HIV/genética , HIV/isolamento & purificação , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Humanos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue , Reto/imunologia , Carga Viral
2.
J Immunol ; 179(11): 7376-84, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18025181

RESUMO

NK cells can identify and eliminate emerging tumors due to altered expression of activating and inhibitory ligands on aberrant cells, a process that is greatly enhanced following NK cell activation. As a principal site of both tumor metastases and immature NK cells, the liver represents a unique anatomic location in which activation of the innate immune system could provide substantial therapeutic benefit. We describe here the NK cell-dependent destruction of a primary hepatic tumor following infection with an attenuated intracellular bacterium derived from Listeria monocytogenes. NK cell-mediated immunity correlated with the ordered migration and maturation of NK cells within the liver. Cytolytic activity was partially dependent on NKG2D-mediated tumor cell recognition, but surprisingly was still effective in the absence of type I IFN. Significantly, NK cell-mediated destruction of a primary hepatic tumor in infected mice led to long-lived CD4- and CD8 T cell-dependent tumor-specific adaptive immunity. These findings establish that activation and differentiation of immature NK cells using complex microbial stimuli can elicit potent anti-tumor activity within the liver, promote cross-presentation of tumor-derived Ags leading to long-lived systemic anti-tumor immunity, and suggests a paradigm for clinical intervention of liver metastatic carcinoma.


Assuntos
Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Listeria monocytogenes/imunologia , Neoplasias Hepáticas/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Imunidade Celular , Imunidade Inata , Interferon gama/biossíntese , Ligantes , Listeriose/imunologia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/imunologia , Receptores Imunológicos/uso terapêutico , Receptores de Células Matadoras Naturais
3.
Cancer Res ; 66(2): 1096-104, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424046

RESUMO

Improved immunization and ex vivo T-cell culture strategies can generate larger numbers and more potent tumor-specific effector cells than previously possible. Nonetheless, the capacity of these cells to eliminate established tumors is limited by their ability to efficiently enter tumor-bearing organs and mediate their effector function. In the current study, we show that the administration of an engineered organ-homing microbe selectively targets tumor-specific immune responses to metastases within that organ. Specifically, an attenuated Listeria monocytogenes strain, which preferentially infects the liver following systemic administration, dramatically enhances the activity of a cancer vaccine against liver metastases but not metastases in the lung. This enhanced activity results from both local recruitment of innate immune effectors as well as concentration and increased activation of vaccine-induced antitumor T cells within the liver. These findings show a general approach to focus systemic cancer immunotherapies to specific organs bearing tumor metastases by taking advantage of differential tropisms and the proinflammatory nature of microbes.


Assuntos
Vacinas Anticâncer/imunologia , Engenharia Genética , Listeria monocytogenes/genética , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/terapia , Animais , Feminino , Hepatite/virologia , Humanos , Imunoterapia/métodos , Inflamação , Listeria monocytogenes/patogenicidade , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T
5.
Proc Natl Acad Sci U S A ; 101(38): 13832-7, 2004 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-15365184

RESUMO

The facultative intracellular bacterium Listeria monocytogenes is being developed as a cancer vaccine platform because of its ability to induce potent innate and adaptive immunity. For successful clinical application, it is essential to develop a Listeria platform strain that is safe yet retains the potency of vaccines based on wild-type bacteria. Here, we report the development of a recombinant live-attenuated vaccine platform strain that retains the potency of the fully virulent pathogen, combined with a >1,000-fold reduction in toxicity, as compared with wild-type Listeria. By selectively deleting two virulence factors, ActA (DeltaactA) and Internalin B (DeltainlB), the immunopotency of Listeria was maintained and its toxicity was diminished in vivo, largely by blocking the direct internalin B-mediated infection of nonphagocytic cells, such as hepatocytes, and the indirect ActA-mediated infection by cell-to-cell spread from adjacent phagocytic cells. In contrast, infection of phagocytic cells was not affected, leaving intact the ability of Listeria to stimulate innate immunity and to induce antigenspecific cellular responses. Listeria DeltaactA/DeltainlB-based vaccines were rapidly cleared from mice after immunization and induced potent and durable effector and memory T-cell responses with no measurable liver toxicity. Therapeutic vaccination of BALB/c mice bearing murine CT26 colon tumor lung metastases or palpable s.c. tumors (>100 mm(3)) with recombinant Listeria DeltaactA/DeltainlB expressing an endogenous tumor antigen resulted in breaking of self-tolerance and long-term survival. We propose that recombinant Listeria DeltaactA/DeltainlB expressing human tumor-associated antigens represents an attractive therapeutic strategy for further development and testing in human clinical trials.


Assuntos
Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Neoplasias do Colo/imunologia , Hepatócitos/imunologia , Listeria monocytogenes/imunologia , Neoplasias Pulmonares/imunologia , Monócitos/imunologia , Linfócitos T/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Deleção de Genes , Hepatócitos/efeitos dos fármacos , Humanos , Memória Imunológica , Listeria monocytogenes/genética , Neoplasias Pulmonares/secundário , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , RNA Bacteriano/genética , RNA de Transferência de Arginina/genética , Linfócitos T/efeitos dos fármacos
6.
Cancer Cell ; 4(4): 241-3, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14585348

RESUMO

Selected mutant strains of vesicular stomatitis virus (VSV) are described that are unable to combat endogenous IFN-beta signaling within infected normal cells and as a result are dramatically more selective for productive growth in tumor cells having a defective antiviral response. The VSV mutants may have the potential to be used clinically as a systemic oncolytic agent for the treatment of distal and metastatic cancers.


Assuntos
Interferon beta/metabolismo , Infecções por Vírus de RNA/terapia , Vírus da Estomatite Vesicular Indiana/fisiologia , Vírus/metabolismo , Animais , Humanos , Interferon beta/antagonistas & inibidores , Mutação , Neoplasias/terapia , Neoplasias/virologia , Infecções por Vírus de RNA/virologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...