Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Clin Cancer Res ; 26(2): 465-476, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31767564

RESUMO

PURPOSE: Pancreatic cancer remains one of the most lethal cancers, and late detection renders most tumors refractory to conventional therapies. Development of cancer prophylaxis may be the most realistic option for improving mortality associated with this disease. Here, we develop a novel individualized prophylactic and therapeutic vaccination regimen using induced pluripotent stem cells (iPSC), gene editing, and tumor-targeted replicating oncolytic viruses. EXPERIMENTAL DESIGN: We created a Virus-Infected, Reprogrammed Somatic cell-derived Tumor cell (VIReST) regime. iPSCs from healthy cells were induced to pancreatic tumor cells using in situ gene editing via stable provision of KRas G12D and p53 R172H tumor driver mutations. These cells were preinfected with oncolytic Adenovirus (AdV) as prime or Vaccinia virus (VV) as boost, to improve vaccine immunogenicity, prior to delivery of vaccines in a sequential regime to young KPC transgenic mice, genetically programmed to develop pancreatic cancer, to prevent and delay disease development. RESULTS: Tumor cells preinfected with oncolytic AdV as prime or VV as boost were the best regime to induce tumor-specific immunity. iPSC-derived tumor cells were highly related in antigen repertoire to pancreatic cancer cells of KPC transgenic mice, suggesting that an individual's stem cells can provide an antigenically matched whole tumor cell vaccine. The VIReST vaccination primed tumor-specific T-cell responses, resulting in delayed disease emergence and progression and significantly prolonged survival of KPC transgenic mice. Importantly, this regime was well-tolerated and nontoxic. CONCLUSIONS: These results provide both proof of concept and a robust technology platform for the development of personalized prophylactic cancer vaccines to prevent pancreatic malignancies in at-risk individuals.


Assuntos
Vacinas Anticâncer/administração & dosagem , Células-Tronco Pluripotentes Induzidas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Experimentais/prevenção & controle , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Neoplasias Pancreáticas/prevenção & controle , Animais , Vacinas Anticâncer/imunologia , Chlorocebus aethiops , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Taxa de Sobrevida , Linfócitos T/imunologia , Resultado do Tratamento
3.
Methods Mol Biol ; 1316: 25-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25967050

RESUMO

For structural, biochemical or pharmacological studies, it is required to have pure RNA in large quantities. We previously devised a generic approach that allows efficient in vivo expression of recombinant RNA in Escherichia coli. We have extended the "tRNA scaffold" method to RNA/protein co-expression in order to express and purify RNA by affinity in native condition. As a proof-of-concept, we present the expression and the purification of the AtRNA-mala in complex with the MS2 coat protein.


Assuntos
Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Proteínas/genética , Proteínas/isolamento & purificação , RNA/genética , RNA/isolamento & purificação , Conformação de Ácido Nucleico , Ligação Proteica , Proteínas/metabolismo , RNA/química , RNA/metabolismo , RNA de Transferência/química , RNA de Transferência/genética , RNA de Transferência/isolamento & purificação , RNA de Transferência/metabolismo
4.
Clin Cancer Res ; 21(2): 405-16, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416195

RESUMO

PURPOSE: Vaccinia virus has strong potential as a novel therapeutic agent for treatment of pancreatic cancer. We investigated whether arming vaccinia virus with interleukin-10 (IL10) could enhance the antitumor efficacy with the view that IL10 might dampen the host immunity to the virus, increasing viral persistence, thus maximizing the oncolytic effect and antitumor immunity associated with vaccinia virus. EXPERIMENTAL DESIGN: The antitumor efficacy of IL10-armed vaccinia virus (VVLΔTK-IL10) and control VVΔTK was assessed in pancreatic cancer cell lines, mice bearing subcutaneous pancreatic cancer tumors and a pancreatic cancer transgenic mouse model. Viral persistence within the tumors was examined and immune depletion experiments as well as immunophenotyping of splenocytes were carried out to dissect the functional mechanisms associated with the viral efficacy. RESULTS: Compared with unarmed VVLΔTK, VVLΔTK-IL10 had a similar level of cytotoxicity and replication in vitro in murine pancreatic cancer cell lines, but rendered a superior antitumor efficacy in the subcutaneous pancreatic cancer model and a K-ras-p53 mutant-transgenic pancreatic cancer model after systemic delivery, with induction of long-term antitumor immunity. The antitumor efficacy of VVLΔTK-IL10 was dependent on CD4(+) and CD8(+), but not NK cells. Clearance of VVLΔTK-IL10 was reduced at early time points compared with the control virus. Treatment with VVLΔTK-IL10 resulted in a reduction in virus-specific, but not tumor-specific CD8(+) cells compared with VVLΔTK. CONCLUSIONS: These results suggest that VVLΔTK-IL10 has strong potential as an antitumor therapeutic for pancreatic cancer.


Assuntos
Interleucina-10/genética , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/terapia , Vaccinia virus/genética , Animais , Linhagem Celular Tumoral , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Terapia Viral Oncolítica , Neoplasias Pancreáticas/imunologia , Replicação Viral
5.
Eur J Immunol ; 43(10): 2730-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23839924

RESUMO

Type I interferons (IFNs) have the dual ability to promote the development of the immune response and exert an anti-inflammatory activity. We analyzed the integrated effect of IFN-α, TCR signal strength, and CD28 costimulation on human CD4⁺ T-cell differentiation into cell subsets producing the anti- and proinflammatory cytokines IL-10 and IFN-γ. We show that IFN-α boosted TCR-induced IL-10 expression in activated peripheral CD45RA⁺CD4⁺ T cells and in whole blood cultures. The functional cooperation between TCR and IFN-α efficiently occurred at low engagement of receptors. Moreover, IFN-α rapidly cooperated with anti-CD3 stimulation alone. IFN-α, but not IL-10, drove the early development of type I regulatory T cells that were mostly IL-10⁺ Foxp3⁻ IFN-γ⁻ and favored IL-10 expression in a fraction of Foxp3⁺ T cells. Our data support a model in which IFN-α costimulates TCR toward the production of IL-10 whose level can be amplified via an autocrine feedback loop.


Assuntos
Interferon-alfa/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Antígenos CD28/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Retroalimentação Fisiológica , Fatores de Transcrição Forkhead/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Ativação Linfocitária , Ligação Proteica , Receptor Cross-Talk/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...