Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 193(4): 2024-33, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25031460

RESUMO

We established a method to generate a large quantity of myeloid lineage cells from mouse embryonic stem (ES) cells, termed ES cell-derived proliferating myeloid cell lines (ES-ML). ES-ML continuously proliferated in the presence of M-CSF and GM-CSF. ES-ML genetically modified to express an anti-HER2 (neu) mAb single-chain V region fragment reduced the number of cocultured mouse Colon-26 cancer cells expressing HER2. Stimulation of ES-ML with IFN-γ plus LPS or TNF resulted in almost complete killing of the Colon-26 cells by the ES-ML, and the cytotoxicity was mediated, in part, by NO produced by ES-ML. When ES-ML were injected into mice with i.p. established Colon-26 tumors, they efficiently infiltrated the tumor tissues. Injection of ES-ML with rIFN-γ and LPS inhibited cancer progression in the mouse peritoneal cavity. Coinjection of TNF-transfected or untransfected ES-ML with rIFN-γ inhibited cancer growth and resulted in prolonged survival of the treated mice. In this experiment, transporter associated with Ag processing (TAP)1-deficient ES-ML exhibited therapeutic activity in MHC-mismatched allogeneic recipient mice. Despite the proliferative capacity of ES-ML, malignancy never developed from the transferred ES-ML in the recipient mice. In summary, TAP-deficient ES-ML with anticancer properties exhibited a therapeutic effect in allogeneic recipients, suggesting the possible use of TAP-deficient human-induced pluripotent stem cell-derived proliferating myeloid cell lines in cancer therapy.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Neoplasias do Colo/terapia , Células-Tronco Embrionárias/imunologia , Macrófagos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Citotoxicidade Imunológica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interferon gama/farmacologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/imunologia , Receptor ErbB-2/imunologia , Anticorpos de Cadeia Única/imunologia , Transplante Homólogo
2.
Oncoimmunology ; 3(1): e27927, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24800175

RESUMO

We established a method to produce a large quantity of myeloid cells from human inducible pluripotent stem cells (iPSCs). When injected intraperitoneally into mice carrying established peritoneal tumors, iPSC-derived myeloid cells (iPS-MCs) efficiently accumulated within neoplastic lesions. The intraperitoneal injection of iPS-MCs expressing interferon ß significantly inhibited the growth of human gastric and pancreatic cancers implanted in the peritoneal cavity of immunocompromised mice.

3.
PLoS One ; 8(6): e67567, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826321

RESUMO

We recently developed a method to generate myeloid cells with proliferation capacity from human iPS cells. iPS-ML (iPS-cell-derived myeloid/macrophage line), generated by introducing proliferation and anti-senescence factors into iPS-cell-derived myeloid cells, grew continuously in an M-CSF-dependent manner. A large number of cells exhibiting macrophage-like properties can be readily obtained by using this technology. In the current study, we evaluated the possible application of iPS-ML in anti-cancer therapy. We established a model of peritoneally disseminated gastric cancer by intraperitoneally injecting NUGC-4 human gastric cancer cells into SCID mice. When iPS-ML were injected intraperitoneally into the mice with pre-established peritoneal NUGC-4 tumors, iPS-ML massively accumulated and infiltrated into the tumor tissues. iPS-ML expressing IFN-ß (iPS-ML/IFN-ß) significantly inhibited the intra-peritoneal growth of NUGC-4 cancer. Furthermore, iPS-ML/IFN-ß also inhibited the growth of human pancreatic cancer MIAPaCa-2 in a similar model. iPS-ML are therefore a promising treatment agent for peritoneally disseminated cancers, for which no standard treatment is currently available.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Interferon beta/metabolismo , Células Mieloides/metabolismo , Células Mieloides/transplante , Neoplasias Peritoneais/terapia , Transplante de Células-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/farmacologia , Humanos , Injeções Intraperitoneais , Macrófagos/patologia , Camundongos , Camundongos SCID , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Neoplasias Peritoneais/secundário , Peritônio/metabolismo , Peritônio/patologia , Receptor ErbB-2/metabolismo , Anticorpos de Cadeia Única/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA