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.
Mol Ther ; 28(4): 1068-1077, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32101701

RESUMO

Checkpoint-inhibiting antibodies elicit impressive clinical responses, but still face several issues. The current study evaluated whether DNA-based delivery can broaden the application of checkpoint inhibitors, specifically by pursuing cost-efficient in vivo production, facilitating combination therapies, and exploring administration routes that lower immune-related toxicity risks. We therefore optimized plasmid-encoded anti-CTLA-4 and anti-PD-1 antibodies, and studied their pharmacokinetics and pharmacodynamics when delivered alone and in combination via intramuscular or intratumoral electroporation in mice. Intramuscular electrotransfer of these DNA-based antibodies induced complete regressions in a subcutaneous MC38 tumor model, with plasma concentrations up to 4 and 14 µg/mL for anti-CTLA-4 and anti-PD-1 antibodies, respectively, and antibody detection for at least 6 months. Intratumoral antibody gene electrotransfer gave similar anti-tumor responses as the intramuscular approach. Antibody plasma levels, however, were up to 70-fold lower and substantially more transient, potentially improving biosafety of the expressed checkpoint inhibitors. Intratumoral delivery also generated a systemic anti-tumor response, illustrated by moderate abscopal effects and prolonged protection of cured mice against a tumor rechallenge. In conclusion, intramuscular and intratumoral DNA-based delivery of checkpoint inhibitors both enabled long-term anti-tumor responses despite distinct systemic antibody exposure, highlighting the potential of the tumor as delivery site for DNA-based therapeutics.


Assuntos
Anticorpos Monoclonais/genética , Neoplasias do Colo/terapia , Inibidores de Checkpoint Imunológico/administração & dosagem , Plasmídeos/administração & dosagem , Animais , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Sinergismo Farmacológico , Feminino , Inibidores de Checkpoint Imunológico/farmacologia , Injeções Intralesionais , Injeções Intramusculares , Camundongos , Plasmídeos/genética , Receptor de Morte Celular Programada 1/imunologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Gene Ther ; 29(7): 984-992, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34754076

RESUMO

To improve the anti-tumor efficacy of immune checkpoint inhibitors, numerous combination therapies are under clinical evaluation, including with IL-12 gene therapy. The current study evaluated the simultaneous delivery of the cytokine and checkpoint-inhibiting antibodies by intratumoral DNA electroporation in mice. In the MC38 tumor model, combined administration of plasmids encoding IL-12 and an anti-PD-1 antibody induced significant anti-tumor responses, yet similar to the monotherapies. When treatment was expanded with a DNA-based anti-CTLA-4 antibody, this triple combination significantly delayed tumor growth compared to IL-12 alone and the combination of anti-PD-1 and anti-CTLA-4 antibodies. Despite low drug plasma concentrations, the triple combination enabled significant abscopal effects in contralateral tumors, which was not the case for the other treatments. The DNA-based immunotherapies increased T cell infiltration in electroporated tumors, especially of CD8+ T cells, and upregulated the expression of CD8+ effector markers. No general immune activation was detected in spleens following either intratumoral treatment. In B16F10 tumors, evaluation of the triple combination was hampered by a high sensitivity to control plasmids. In conclusion, intratumoral gene electrotransfer allowed effective combined delivery of multiple immunotherapeutics. This approach induced responses in treated and contralateral tumors, while limiting systemic drug exposure and potentially detrimental systemic immunological effects.


Assuntos
Linfócitos T CD8-Positivos , Inibidores de Checkpoint Imunológico , Interleucina-12 , Neoplasias , Animais , Anticorpos Monoclonais/administração & dosagem , Linhagem Celular Tumoral , DNA , Terapia Genética , Inibidores de Checkpoint Imunológico/administração & dosagem , Imunoterapia , Interleucina-12/genética , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia
3.
Sci Rep ; 10(1): 19532, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177564

RESUMO

Intratumoral delivery of drug-encoding plasmid DNA (pDNA) enables localised in vivo expression of biological drugs, offering an attractive alternative to conventional protein treatment. However, this requires physical or chemical methods to enhance the low transfection efficiency of naked pDNA. Electroporation and complexation with the polycation in vivo-jetPEI are both evaluated in the clinic for intratumoral pDNA delivery, but lack head-to-head comparison. This study therefore compared both methods for intratumoral DNA-based reporter gene transfer in a subcutaneous mouse tumour model. Intratumoral electroporation resulted in strong reporter expression that was restricted to the tumour area and persisted for at least ten days. Intratumoral expression after injection of pDNA-jetPEI complexes was two to three logs lower, did not exceed the background in most mice, and lasted less than five days even with repeated dosing. Remarkably, reporter expression was primarily detected in the lungs, presumably due to leakage of pDNA-jetPEI complexes into the systemic circulation. In conclusion, electroporation enabled more efficient, prolonged and tumour-specific reporter expression compared to intratumoral injection of pDNA complexed with in vivo-jetPEI. These results favour the use of electroporation for intratumoral DNA-based gene transfer, and suggest further optimisation of pDNA-jetPEI complexes is needed to improve their efficacy and biosafety.


Assuntos
DNA/administração & dosagem , Eletroporação/métodos , Técnicas de Transferência de Genes , Genes Reporter , Neoplasias Experimentais/genética , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Feminino , Regulação da Expressão Gênica , Luciferases de Vaga-Lume/genética , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/química , Plasmídeos/genética , Polietilenoimina/química , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa