Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Vaccine ; 42(6): 1300-1310, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38302336

RESUMO

DNA- based vaccines have demonstrated the potential as a safe and effective modality. PlaCCine, a DNA-based vaccine approach described subsequently relies on a synthetic DNA delivery system and is independent of virus or device. The synthetic functionalized polymer combined with DNA demonstrated stability over 12 months at 4C and for one month at 25C. Transfection efficiency compared to naked DNA increased by 5-15-fold in murine skeletal muscle. Studies of DNA vaccines expressing spike proteins from variants D614G (pVAC15), Delta (pVAC16), or a D614G + Delta combination (pVAC17) were conducted. Mice immunized intramuscular injection (IM) with pVAC15, pVAC16 or pVAC17 formulated with functionalized polymer and adjuvant resulted in induction of spike-specific humoral and cellular responses. Antibody responses were observed after one immunization. And endpoint IgG titers increased to greater than 1x 105 two weeks after the second injection. Neutralizing antibodies as determined by a pseudovirus competition assay were observed following vaccination with pVAC15, pVAC16 or pVAC17. Spike specific T cell immune responses were also observed following vaccination and flow cytometry analysis demonstrated the cellular immune responses included both CD4 and CD8 spike specific T cells. The immune responses in vaccinated mice were maintained for up to 14 months after vaccination. In an immunization and challenge study of K18 hACE2 transgenic mice pVAC15, pVAC16 and pVAC17 induced immune responses lead to decreased lung viral loads by greater than 90 % along with improved clinical score. These findings suggest that PlaCCine DNA vaccines are effective and stable and further development against emerging SARS-CoV-2 variants is warranted.


Assuntos
COVID-19 , Vacinas de DNA , Camundongos , Animais , Vacinas contra COVID-19 , COVID-19/prevenção & controle , SARS-CoV-2 , Camundongos Transgênicos , Anticorpos Neutralizantes , DNA , Anticorpos Antivirais , Glicoproteína da Espícula de Coronavírus/genética , Imunogenicidade da Vacina
2.
J Control Release ; 201: 49-55, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25599856

RESUMO

Tumor drug delivery is a complex phenomenon affected by several elements in addition to drug or delivery vehicle's physico-chemical properties. A key factor is tumor microvasculature with complex effects including convective transport, high interstitial pressure and enhanced vascular permeability due to the presence of "leaky vessels". Current in vitro models of the tumor microenvironment for evaluating drug delivery are oversimplified and, as a result, show poor correlation with in vivo performance. In this study, we report on the development of a novel microfluidic platform that models the tumor microenvironment more accurately, with physiologically and morphologically realistic microvasculature including endothelial cell lined leaky capillary vessels along with 3D solid tumors. Endothelial cells and 3D spheroids of cervical tumor cells were co-cultured in the networks. Drug vehicle screening was demonstrated using GFP gene delivery by different formulations of nanopolymers. The synthetic tumor network was successful in predicting in vivo delivery efficiencies of the drug vehicles. The developed assay will have critical applications both in basic research, where it can be used to develop next generation delivery vehicles, and in drug discovery where it can be used to study drug transport and delivery efficacy in realistic tumor microenvironment, thereby enabling drug compound and/or delivery vehicle screening.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias/metabolismo , Microambiente Tumoral , Linhagem Celular , Técnicas de Cocultura , Células Endoteliais , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Microfluídica , Nanopartículas/administração & dosagem , Nanopartículas/química , Plasmídeos , Polímeros/administração & dosagem , Polímeros/química
3.
J Gene Med ; 11(8): 718-28, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19507172

RESUMO

BACKGROUND: The poor prognosis associated with ovarian cancer is primarily the result of delayed diagnosis and the lack of an effective treatment for advanced disease. Use of novel immunotherapy strategies are being evaluated that work to enhance local and systemic immune responses against cancer cells and can possibly work together with traditional cytotoxic chemotherapy regimens to produce more effective treatment options. METHODS: In the present study, we describe a gene-based therapy whereby the anticancer cytokine interleukin-12 gene (pmIL-12) is formulated with a synthetic polymeric delivery vehicle (PPC) and administered intraperitoneally into a mouse model of disseminated ovarian cancer. RESULTS: The administration of pmIL-12/PPC in tumor-bearing mice was associated with a shift towards a Th1 immune state, including significant increases in murine IL-12 (mIL-12) and interferon (IFN)-gamma (mIFN-gamma) in ascites fluid, with little change in systemic levels of these proteins. The mIL-12 protein was detectable for several days and could be reintroduced with subsequent injections. We show that treatment delayed the onset of ascites formation and improved survival in a dose-dependent manner. A significant decrease in vascular endothelial growth factor was associated with pmIL-12/PPC delivery and believed to play a predominant role in inhibiting ascites accumulation. Administration of pmIL-12/PPC was associated with minimal toxicity and, when combined with standard chemotherapies, resulted in additive improvement in survival. CONCLUSIONS: Taken together, these results suggest that pmIL-12/PPC may be an effective strategy for inhibiting progression of disseminated ovarian cancer and may offer a new option for treatment of advanced disease that can be used to complement standard therapies.


Assuntos
Terapia Genética , Interleucina-12/genética , Interleucina-12/uso terapêutico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Animais , Ascite/metabolismo , Contagem de Células Sanguíneas , Peso Corporal/efeitos dos fármacos , Carboplatina/farmacologia , Carboplatina/uso terapêutico , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Interleucina-12/administração & dosagem , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/sangue , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Vírus
4.
Anticancer Drugs ; 19(2): 133-42, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18176109

RESUMO

Interleukin-12 (IL-12) triggers an antitumoral immune response and an antiangiogenic effect against cancer. In this study, we tested a novel polymeric vehicle for IL-12 gene therapy along with adjuvant local biodegradable carmustine (BCNU) chemotherapy for the treatment of malignant glioma. Highly concentrated DNA/PPC (polyethylenimine covalently modified with methoxypolyethyleneglycol and cholesterol) complexes were used to deliver a murine plasmid encoding IL-12 (pmIL-12). For toxicity assessment, mice received intracranial injections with different volumes of pmIL-12/PPC. For efficacy, mice with intracranial GL261 glioma were treated with local delivery of pmIL-12/PPC and/or BCNU-containing polymers. Intracranial injections of 5-10 microl of pmIL-12/PPC were well tolerated and led to IL-12 expression in the brains of treated animals. Treatment with pmIL-12/PPC led to a significant increase in survival compared with untreated mice (median survival 57 days; 25% long-term survival >95 vs. 45 days for control; P<0.05). Treatment with BCNU led to a significant increase in survival compared with untreated mice, with 75% of treated mice having a long-term survival >95 days, (P<0.05). Most importantly, the combination of BCNU and pmIL-12/PPC led to a survival of 100% of the mice for 95 days after treatment (P<0.0001). This novel strategy is safe and effective for the treatment of malignant glioma. The synergy resultant from the combination of locally administered pmIL-12/PPC and BCNU suggests a role for this approach in the treatment of malignant brain tumors.


Assuntos
Terapia Genética/métodos , Glioma/terapia , Interleucina-12/genética , Polímeros/química , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Células COS , Chlorocebus aethiops , Colesterol/química , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/tendências , Glioma/genética , Glioma/patologia , Injeções , Interleucina-12/metabolismo , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/administração & dosagem , Plasmídeos/química , Plasmídeos/genética , Polietilenoglicóis/química , Transfecção , Transgenes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA