Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Oncol ; 8(3): 609-19, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24503218

RESUMO

Nonviral gene therapy represents a realistic option for clinical application in cancer treatment. This preclinical study demonstrates the advantage of using the small-size MIDGE(®) DNA vector for improved transgene expression and therapeutic application. This is caused by significant increase in transcription efficiency, but not by increased intracellular vector copy numbers or gene transfer efficiency. We used the MIDGE-hTNF-alpha vector for high-level expression of hTNF-alpha in vitro and in vivo for a combined gene therapy and vindesine treatment in human melanoma models. The MIDGE vector mediated high-level hTNF-alpha expression leads to sensitization of melanoma cells towards vindesine. The increased efficacy of this combination is mediated by remarkable acceleration and increase of initiator caspase 8 and 9 and effector caspase 3 and 7 activation. In the therapeutic approach, the nonviral intratumoral in vivo jet-injection gene transfer of MIDGE-hTNF-alpha in combination with vindesine causes melanoma growth inhibition in association with increased apoptosis in A375 cell line or patient derived human melanoma xenotransplant (PDX) models. This study represents a proof-of-concept for an anticipated phase I clinical gene therapy trial, in which the MIDGE-hTNF-alpha vector will be used for efficient combined chemo- and nonviral gene therapy of malignant melanoma.


Assuntos
DNA/uso terapêutico , Vetores Genéticos/uso terapêutico , Melanoma/genética , Melanoma/terapia , Fator de Necrose Tumoral alfa/genética , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , DNA/genética , Feminino , Terapia Genética , Vetores Genéticos/genética , Humanos , Melanoma/patologia , Camundongos , Transfecção , Transgenes , Vindesina/uso terapêutico
2.
Oncotarget ; 3(8): 783-97, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22878175

RESUMO

The metastasis-inducing protein S100A4 was found to be a prognostic indicator for the development of metachronous metastases. S100A4 expression levels correlate with the formation of human colorectal cancer metastases and shorter patients' survival. Inhibition of S100A4 expression in patients might therefore result in decreased metastasis formation and prolonged survival. In the present study, we used shRNA expression plasmids to inhibit S100A4 expression in the colorectal cancer cell lines HCT116, SW620 and DLD-1. Cell lines with reduced S100A4 expression showed reduced cell migration and invasion in vitro. The knock-down of S100A4 expression also led to significantly diminished formation of liver metastases when intrasplenically transplanted in mice (P = 0.004). We then focused on the therapeutic potential of systemically applied shRNA expression plasmids acting on S100A4 via repeated hydrodynamics-based tail vein injection of plasmid DNA. Mice, intrasplenically transplanted with HCT116 cells and treated systemically with S100A4­shRNA plasmids, showed a decrease of S100A4 and MMP9 expression levels, resulting in significantly reduced liver metastases (P = 0.005). In summary, we show for the first time the intratumoral knock down of S100A4 via systemic application of S100A4­shRNA plasmid DNA, which restricts metastasis formation in a xenografted mouse model of colorectal cancer.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Metástase Neoplásica , Interferência de RNA , Proteínas S100/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , RNA Interferente Pequeno , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Transplante Heterólogo
3.
Pharm Res ; 29(7): 1949-59, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22399388

RESUMO

PURPOSE: To test targeted liposomes in an effort to improve drug transport across cellular barriers into the brain. METHODS: Therefore we prepared Mitoxantrone (MTO) entrapping, rigid and fluid liposomes, equipped with a 19-mer angiopeptide as ligand for LDL lipoprotein receptor related protein (LRP) targeting. RESULTS: Fluid, ligand bearing liposomes showed in vitro the highest cellular uptake and transcytosis and were significantly better than the corresponding ligand-free liposomes and rigid, ligand-bearing vesicles. Treatment of mice, transplanted with human breast cancer cells subcutaneously and into the brain, with fluid membrane liposomes resulted in a significant reduction in the tumor volume by more than 80% and in a clear reduction in drug toxicity. The improvement was mainly depended on liposome fluidity while the targeting contributed only to a minor degree. Pharmacokinetic parameters were also improved for liposomal MTO formulations in comparison to the free drug. So the area under the curve was increased and t(1/2) was extended for liposomes. CONCLUSION: Our data show that it is possible to significantly improve the therapy of brain metastases if MTO-encapsulating, fluid membrane liposomes are used instead of free MTO. This effect could be further enhanced by fluid, ligand bearing liposomes.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Encéfalo/efeitos dos fármacos , Proteínas Relacionadas a Receptor de LDL/metabolismo , Lipossomos/metabolismo , Mitoxantrona/uso terapêutico , Sequência de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Lipossomos/química , Camundongos , Mitoxantrona/administração & dosagem , Mitoxantrona/farmacocinética , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo
4.
J Natl Cancer Inst ; 103(13): 1018-36, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21685359

RESUMO

BACKGROUND: Metastasis formation in colon cancer severely reduces the survival rate in patients. S100A4, a calcium-binding protein, is implicated in promoting metastasis formation in colon cancer. METHODS: To identify a transcription inhibitor of S100A4, high-throughput screening of 1280 pharmacologically active compounds was performed using a human colon cancer cell line expressing a S100A4 promoter-driven luciferase (LUC) reporter gene construct (HCT116-S1004p-LUC). Niclosamide, an antihelminthic agent, was identified as a potential candidate. Colon cancer cell lines (HCT116, SW620, LS174T, SW480, and DLD-1) were treated with 1 µM niclosamide to analyze the effect on S100A4 mRNA and protein expression by quantitative reverse transcription-polymerase chain reaction and immunoblot assays, and effects on cell migration, invasion, proliferation, and colony formation were also assessed in vitro. The effect of niclosamide on liver metastasis was assessed in a xenograft mouse model of human colon cancer (n = 8 mice) by in vivo imaging. The long-term effect of niclosamide on metastasis formation after discontinued treatment was quantified by scoring, and overall survival (n = 12 mice) was analyzed by Kaplan-Meier method after discontinuation of treatment. All statistical tests were two-sided. RESULTS: Reduced S100A4 mRNA and protein expression, and inhibited cell migration, invasion, proliferation, and colony formation were observed in niclosamide-treated colon cancer cells in vitro. In vivo imaging of niclosamide-treated mice showed reduced liver metastasis compared with solvent-treated control mice (n = 4 mice per group). Compared with the control group, discontinuation of treatment for 26 days showed reduced liver metastasis formation in mice (n = 6 mice per group) (control vs discontinuous treatment, mean metastasis score = 100% vs 34.9%, mean difference = 65.1%; 95% confidence interval [CI] = 18.4% to 111.9%, P < .01) and increased overall survival (n = 6 mice per group; control vs discontinuous treatment, median survival = 24 vs 46.5 days, ratio = 0.52, 95% CI = 0.19 to 0.84, P = .001). CONCLUSION: Niclosamide inhibits S100A4-induced metastasis formation in a mouse model of colon cancer and has therapeutic potential.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias Hepáticas/prevenção & controle , Niclosamida/farmacologia , Proteínas S100/metabolismo , Animais , Anti-Helmínticos/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Neoplasias do Colo/patologia , Progressão da Doença , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Infusões Parenterais , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Niclosamida/administração & dosagem , Niclosamida/química , Prognóstico , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transplante Heterólogo , Ensaio Tumoral de Célula-Tronco , Proteínas Wnt/efeitos dos fármacos , Proteínas Wnt/metabolismo , beta Catenina/efeitos dos fármacos , beta Catenina/metabolismo
5.
Neoplasia ; 13(2): 131-44, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21403839

RESUMO

Colon cancer metastasis is often associated with activation of the Wnt/ß-catenin signaling pathway and high expression of the metastasis mediator S100A4. We previously demonstrated the transcriptional regulation of S100A4 by ß-catenin and the importance of the interconnection of these cellular programs for metastasis. Here we probe the hypothesis that the nonsteroidal anti-inflammatory drug sulindac sulfide can inhibit colon cancer metastasis by intervening in ß-catenin signaling and thereby interdicting S100A4. We treated colon cancer cell lines heterozygous for gain-of-function and wild-type ß-catenin with sulindac. We analyzed sulindac's effects on ß-catenin expression and subcellular localization, ß-catenin binding to the T-cell factor (TCF)/S100A4 promoter complex, S100A4 promoter activity, S100A4 expression, cell motility, and proliferation. Mice intrasplenically transplanted with S100A4-overexpressing colon cancer cells were treated with sulindac. Tumor growth and metastasis, and their ß-catenin and S100A4 expressions, were determined. We report the expression knockdown of ß-catenin by sulindac, leading to its reduced nuclear accumulation. The binding of ß-catenin to TCF was clearly lowered, resulting in reduced S100A4 promoter activity and expression. This correlated well with the inhibition of cell migration and invasion, which could be rescued by ectopic S100A4 expression. In mice, sulindac treatment resulted in reduced tumor growth in the spleen (P = .014) and decreased liver metastasis in a human colon cancer xenograft model (P = .025). Splenic tumors and liver metastases of sulindac-treated mice showed lowered ß-catenin and S100A4 levels. These results suggest that modulators of ß-catenin signaling such as sulindac offer potential as antimetastatic agents by interdicting S100A4 expression.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Proteínas S100/genética , Sulindaco/uso terapêutico , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Fígado/patologia , Camundongos , Metástase Neoplásica , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Transdução de Sinais , Baço/patologia , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Ativação Transcricional , Proteínas Wnt/genética
6.
J Pharm Sci ; 99(5): 2423-33, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20014431

RESUMO

The aim of this study was to investigate the effect of liposomal membrane properties on cellular uptake and transcytosis across a tight Madin-Darby canine kidney (MDCK) cell barrier in vitro. More than 25 small vesicles were prepared by lipid film hydration/extrusion to generate small unilamellar vesicles. The fluorescence marker calcein was encapsulated to mimic hydrophilic drug transport. Marker uptake by MDCK cells seems to be mediated by different mechanisms for the liposomes used. It was mainly depending on membrane fluidity and vesicle charge. Liposomes L2 with a positive charge (325 +/- 3 pmol/well) and vesicles L3 containing the helper lipid dioleylphosphatidylethanolamine (DOPE) in their membrane (216 +/- 42 pmol/well) were taken up to the most. Selected liposomes were tested for their transcytotic transport across a MDCK monolayer. Liposomes L4 containing equimolar DOPE and octadecyl-1,1-dimethylpiperidin-1-ium-4-yl phosphate (OPP) were the most efficient vesicles for transcellular transport resulting in 808 +/- 30 pmol calcein/cm(2) in the basal medium (28.1% of total liposomal marker added). Transcytosis was positively correlated with membrane fluidity in the outer part of the bilayer, as electron paramagnetic resonance measurements revealed. We expect that an increase in membrane fluidity of vesicles should also improve the restricted transport of hydrophilic drugs across the blood-brain barrier.


Assuntos
Portadores de Fármacos/química , Células Epiteliais/metabolismo , Fluidez de Membrana , Nanopartículas/química , Lipossomas Unilamelares/química , Animais , Transporte Biológico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cães , Portadores de Fármacos/farmacocinética , Composição de Medicamentos , Espectroscopia de Ressonância de Spin Eletrônica , Células Epiteliais/efeitos dos fármacos , Fluoresceínas/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Microscopia Confocal , Microscopia de Fluorescência , Lipossomas Unilamelares/farmacocinética
7.
Am J Chin Med ; 37(6): 1153-65, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19938223

RESUMO

The aim of the present work was to explore the anti-hepatoma effects of icariin both in vitro and in vivo and to elucidate its potential mechanism of action. The MTT assay was applied to test the anti-proliferative effects of icariin in vitro. HepG2 bearing NMRI nu/nu mice were used to test the anticancer effects of icariin in vivo. Immunohistochemical assay and flow cytometry assay (FACS) were applied to detect the possible mechanisms of action of icariin. MTT assay illustrated that icariin inhibited the proliferation of HepG2 cells in a concentration dependent manner; meanwhile, icariin inhibited the tumor growth in HepG2 bearing NMRI nu/nu mice. The tumor weight was inhibited by 55.6% and tumor volume was inhibited by 47.2%. Icariin did not influence the spleen and body weights or blood parameters. Immunohistochemical analysis indicated that the expressions of both CD31 and Ki67 in the icariin treated group were significantly lower than those in the control group (p < 0.01). FACS assay showed that icariin dramatically decreased the percentage of CD4+ and CD8+ cells in bone marrow and CD19+ cells in blood on day 8. On day 17, the percentage of CD8+ cells in blood was lower than those in the control group. CD4/CD8 ratio in icariin group was significantly elevated in bone marrow on day 17. Icariin showed anticancer efficacy both in vitro and in vivo. The possible mechanism of action could be related to its anti-angiogenesis and anti-proliferative effects in tumors.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Epimedium/química , Flavonoides/farmacologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Antígenos CD19/sangue , Antineoplásicos Fitogênicos/uso terapêutico , Medula Óssea/imunologia , Relação CD4-CD8 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Flavonoides/uso terapêutico , Citometria de Fluxo , Células Hep G2 , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Nus , Fitoterapia , Extratos Vegetais/uso terapêutico , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Neuro Oncol ; 11(2): 158-66, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18780814

RESUMO

The aim of this study was to determine the efficacy of sagopilone (ZK-EPO), a novel epothilone, compared with other anticancer agents in orthotopic models of human primary and secondary brain tumors. Autoradiography and pharmacokinetic analyses were performed on rats and mice to determine passage across the blood-brain barrier and organ distribution of sagopilone. Mice bearing intracerebral human tumors (U373 or U87 glioblastoma, MDA-MB-435 melanoma, or patient-derived non-small-cell lung cancer [NSCLC]) were treated with sagopilone 5-10 mg/kg, paclitaxel 8-12.5 mg/kg (or temozolomide, 100 mg/kg) or control (vehicle only). Tumor volume was measured to assess antitumor activity. Sagopilone crossed the blood-brain barrier in both rat and mouse models, leading to therapeutically relevant concentrations in the brain with a long half-life. Sagopilone exhibited significant antitumor activity in both the U373 and U87 models of human glioblastoma, while paclitaxel showed a limited effect in the U373 model. Sagopilone significantly inhibited the growth of tumors from CNS metastasis models (MDA-MB-435 melanoma and patient-derived Lu7187 and Lu7466 NSCLC) implanted in the brains of nude mice, in contrast to paclitaxel or temozolomide. Sagopilone has free access to the brain. Sagopilone demonstrated significant antitumor activity in orthotopic models of both glioblastoma and CNS metastases compared with paclitaxel or temozolomide, underlining the value of further research evaluating sagopilone in the treatment of brain tumors. Sagopilone is currently being investigated in a broad phase II clinical trial program, including patients with glioblastoma, NSCLC, breast cancer, and melanoma.


Assuntos
Benzotiazóis/farmacocinética , Barreira Hematoencefálica , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Epotilonas/farmacocinética , Animais , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/uso terapêutico , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/uso terapêutico , Benzotiazóis/uso terapêutico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Epotilonas/uso terapêutico , Feminino , Meia-Vida , Humanos , Masculino , Camundongos , Camundongos Nus , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico , Ratos , Ratos Wistar , Taxa de Sobrevida , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...