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.
Clin Cancer Res ; 15(11): 3697-704, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19435839

RESUMO

PURPOSE: Epidermal growth factor receptor (EGFR) signal transduction pathways are implicated in malignant glioma aggressiveness and promote tumor cell invasion, proliferation, and angiogenesis. Nevertheless, response to EGFR tyrosine kinase inhibitor gefitinib (Iressa, ZD1839) has been disappointing in clinical trials. One potential explanation may come from the diversity of molecular alterations seen in gliomas. To validate that hypothesis, we have investigated responses to gefitinib on various tumor parameters in human malignant gliomas that exhibited different molecular alterations. EXPERIMENTAL DESIGN: We used a panel of six human malignant gliomas from established xenografts characterized for their genetic (EGFR, PTEN, TP53, and CDKN2A) and molecular (EGFR, PTEN, ERK, and Akt) alterations. Tumors were treated with gefitinib (1 or 10 micromol/L) for prolonged periods (8 or 16 days) in an organotypic brain slice model that allowed quantification of invasion, proliferation, and angiogenesis. RESULTS: In nontreated tumors, EGFR amplification was associated with profuse tumor cell invasion. After treatment, invasion was inhibited in tumors with EGFR amplification in a dose-dependent manner. Treatment had only antiproliferative effect in two of three tumors with EGFR amplification. Tumors with PTEN loss were resistant to treatment. We did not observe shrinkage of the tumors after treatment. None of the tumors had mutations of the EGFR kinase domain. Gefitinib had similar antiangiogenic effect in all of the tumors. CONCLUSIONS: Gefitinib reduces cell invasion in EGFR amplified tumors. PTEN loss of expression seems to be a determinant of resistance. Interestingly, inhibition of angiogenesis by gefitinib seems independent on the EGFR genetic status of the tumors.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Glioma/patologia , Neovascularização Patológica/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gefitinibe , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , Mutação , Invasividade Neoplásica , Neovascularização Patológica/genética , Técnicas de Cultura de Órgãos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Interferência de RNA , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Neuro Oncol ; 9(4): 412-23, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17622648

RESUMO

Angiogenesis inhibitors appear to be promising therapies for highly vascularized tumors such as glioblastoma multiforme (GBM). Sunitinib is an oral multitargeted tyrosine kinase inhibitor with both antiangiogenic and antitumor activities due to selective inhibition of various receptor tyrosine kinases, including those important for angiogenesis (vascular endothelial growth factor receptors and platelet-derived growth factor receptors). Here we evaluated the antitumor activities of sunitinib on orthotopic models of GBM in vitro and in vivo. Sunitinib potently inhibited angiogenesis that was stimulated by implantation of U87MG and GL15 cells into organotypic brain slices at concentrations as low as 10 nM. At high dose (10 microM), sunitinib induced direct antiproliferative and proapoptotic effects on GL15 cells and decreased invasion of these cells implanted into brain slices by 49% (p < 0.001). Treatment was associated with decreases in Src (35%) and focal adhesion kinase (44%) phosphorylation. However, anti-invasive activity was not observed in vivo at the highest dose level utilized (80 mg/kg per day). Survival experiments involving athymic mice bearing intracerebral U87MG GBM demonstrated that oral administration of 80 mg/kg sunitinib (five days on, two days off) improved median survival by 36% (p < 0.0001). Sunitinib treatment caused a 74% reduction in microvessel density (p < 0.05), an increase in tumor necrosis, and a decrease in number of GBM cells positive for MIB antibody. Sunitinib exhibited potent antiangiogenic activity that was associated with a meaningful prolongation of survival of mice bearing intracerebral GBM. These data support the potential utility of sunitinib in the treatment of GBM.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Indóis/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Pirróis/uso terapêutico , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Citometria de Fluxo , Proteína-Tirosina Quinases de Adesão Focal/efeitos dos fármacos , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Técnicas de Cultura de Órgãos , Fosforilação , Sunitinibe , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/efeitos dos fármacos
3.
Mol Biol Cell ; 17(12): 5141-52, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16987961

RESUMO

Phosphoprotein enriched in astrocytes-15 kDa (PEA-15), a phosphoprotein enriched in astrocytes, inhibits both apoptosis and proliferation in normal and cancerous cells. Here, analysis of PEA-15 expression in glioblastoma organotypic cultures revealed low levels of PEA-15 in tumor cells migrating away from the explants, regardless of the expression levels in the originating explants. Because glioblastomas are highly invasive primary brain tumors that can originate from astrocytes, we explored the involvement of PEA-15 in the control of astrocyte migration. PEA-15-/- astrocytes presented an enhanced motility in vitro compared with their wild-type counterparts. Accordingly, NIH-3T3 cells transfected by green fluorescent protein-PEA-15 displayed a reduced migration. Reexpression of PEA-15 restored PEA-15-/- astrocyte motility to wild-type levels. Pharmacological manipulations excluded a participation of extracellular signal-regulated kinase/mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt, and calcium/calmodulin-dependent protein kinase II in this effect of PEA-15. In contrast, treatment by bisindolylmaleimide, Gö6976, and rottlerin, and chronic application of phorbol 12-myristate 13-acetate and/or bryostatin-1 indicated that PKC delta mediated PEA-15 inhibition of astrocyte migration. PEA-15-/- astrocytes constitutively expressed a 40-kDa form of PKC delta that was down-regulated upon PEA-15 reexpression. Together, these data reveal a new function for PEA-15 in the inhibitory control of astrocyte motility through a PKC delta-dependent pathway involving the constitutive expression of a catalytic fragment of PKC delta.


Assuntos
Astrócitos/citologia , Movimento Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C-delta/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Proteínas Reguladoras de Apoptose , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Peso Molecular , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Cicatrização/fisiologia
4.
Bull Cancer ; 92(4): 360-72, 2005 Apr.
Artigo em Francês | MEDLINE | ID: mdl-15888393

RESUMO

The poor prognosis of patients with glioblastoma multiforme in spite of aggressive conventional anticancer therapies has led to the search for new therapeutic strategies. As glioblastomas are highly vascularized and their growth is angiogenesis-dependent, the inhibition of the sprouting of new capillaries from preexisting blood vessels is one of the most promising therapeutic approaches. Different anti-angiogenic strategies have been developed: inhibition of pro-angiogenic factors and/or receptors and/or downstream cell signaling, inactivation of endothelial cells, inhibition of cellular adhesion molecules and/or extracellular matrix remodeling. Inhibitors of angiogenesis are separated into endogenous inhibitors such as angiostatin, trombospondin or alpha interferon and natural or synthetic inhibitors such as thalidomide, antibodies against angiogenic growth factors or inhibitors of tyrosine kinase receptors. In this review, the majority of experimental studies in glioblastoma models in vivo are summarized and clinical perspectives are discussed.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/radioterapia , Quimioterapia Combinada , Glioblastoma/irrigação sanguínea , Glioblastoma/radioterapia , Humanos , Camundongos , Ratos
5.
BMC Cell Biol ; 6(1): 7, 2005 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-15715906

RESUMO

BACKGROUND: Gliomas are "intraparenchymally metastatic" tumors, invading the brain in a non-destructive way that suggests cooperation between glioma cells and their environment. Recent studies using an engineered rodent C6 tumor cell line have pointed to mechanisms of invasion that involved gap junctional communication (GJC), with connexin 43 as a substrate. We explored whether this concept may have clinical relevance by analyzing the participation of GJC in human glioblastoma invasion. RESULTS: Three complementary in vitro assays were used: (i) seeding on collagen IV, to analyze homocellular interactions between tumor cells (ii) co-cultures with astrocytes, to study glioblastoma/astrocytes relationships and (iii) implantation into organotypic brain slice cultures, that mimic the three-dimensional parenchymal environment. Carbenoxolone, a potent blocker of GJC, inhibited cell migration in the two latter models. It paradoxically increased it in the first one. These results showed that homocellular interaction between tumor cells supports intercellular adhesion, whereas heterocellular glioblastoma/astrocytes interactions through functional GJC conversely support tumor cell migration. As demonstrated for the rodent cell line, connexin 43 may be responsible for this heterocellular functional coupling. Its levels of expression, high in astrocytes, correlated positively with invasiveness in biopsied tumors. CONCLUSIONS: our results underscore the potential clinical relevance of the concept put forward by other authors based on experiments with a rodent cell line, that glioblastoma cells use astrocytes as a substrate for their migration by subverting communication through connexin 43-dependent gap junctions.


Assuntos
Astrócitos/patologia , Neoplasias Encefálicas/patologia , Junções Comunicantes/patologia , Glioblastoma/patologia , Invasividade Neoplásica/patologia , Adesão Celular , Comunicação Celular , Movimento Celular , Técnicas de Cocultura , Conexina 43/fisiologia , Humanos , Células Tumorais Cultivadas
6.
Mol Cancer Ther ; 3(2): 129-36, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14985453

RESUMO

Glioblastoma is a therapeutic challenge as a highly infiltrative, proliferative, and resistant tumor. Among novel therapeutic approaches, proteasome inhibition is very promising in controlling cell cycle and inducing apoptosis. This study investigated the effect of ritonavir, a protease inhibitor of the HIV and a proteasome modulator, on glioma cells. The hypothesis was that proteasome modulation, mainly by only inhibiting proteasome chymotrypsin-like activity, could be sufficient to control tumor progression. The experiments were done on a human glioblastoma-derived GL15 cell line and a rat nitrosourea-induced gliosarcoma 9L cell line. Culturing conditions included monolayer cultures, transplantations into brain slices, and transplantations into rat striata. The study demonstrates that ritonavir, by inhibiting the chymotrypsin-like activity of the proteasome, has cytostatic and cytotoxic effects on glioma cells, and can induce resistances in vitro. Ritonavir was unable to control tumor growth in vivo, likely because the therapeutic dose was not reached in the tumor in vivo. Nevertheless, ritonavir might also be beneficial, by decreasing tumor infiltration, in the reduction of the deleterious peritumor edema in glioblastoma.


Assuntos
Inibidores de Cisteína Proteinase/farmacologia , Glioblastoma/enzimologia , Glioblastoma/patologia , Inibidores de Proteassoma , Ritonavir/farmacologia , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quimotripsina/metabolismo , Glioblastoma/metabolismo , Glioblastoma/fisiopatologia , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos
7.
Hum Gene Ther ; 14(9): 883-95, 2003 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12828859

RESUMO

Inhibition of angiogenesis has been considered among the most promising approaches to treat highly vascularized solid tumors such as glioblastoma. In this study, we designed and validated a new in vitro assay system based on the implantation of tumor cells into organotypic brain slice cultures. We evaluated the effects of local production of three endogenous inhibitors of angiogenesis, angiostatin, endostatin, and interferon (IFN)-alpha(1), using stably transfected rat (9L) and human (GL15) glioblastoma cells on tumor vascularization and growth. Despite similar effectiveness of the three proteins in a classic in vitro endothelial cell migration assay, IFN-alpha(1) demonstrated the most potent antiangiogenic effect in organotypic brain slice cultures. In vivo, after intracerebral implantation of such genetically modified glioblastoma cells, IFN-alpha(1) caused a dramatic decrease in tumor volume revealed by magnetic resonance imaging and by postmortem histology. The mechanisms of this antitumor effect were most likely caused by the major antiangiogenic action of the cytokine, because IFN-alpha(1) expression provoked a pronounced decrease in blood vessel density, which was accompanied by extensive necrosis in the body mass of the tumors. The median survival time of rats implanted intracerebrally with IFN-alpha-expressing 9L cells tripled, and was still significantly increased when these constituted only 1% of transplanted tumor cells. A similar effect was seen when 50% of the transplanted cells were replaced by IFN-alpha-expressing bone marrow stromal cells. These data point to the local delivery of IFN-alpha(1) using cell vectors as a potent tool for the inhibition of tumor-induced angiogenesis.


Assuntos
Inibidores da Angiogênese , Neoplasias Encefálicas/terapia , Colágeno/genética , Glioblastoma/terapia , Interferon-alfa/genética , Fragmentos de Peptídeos/genética , Plasminogênio/genética , Inibidores da Angiogênese/análise , Angiostatinas , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Células Cultivadas , Colágeno/análise , Endostatinas , Imunofluorescência , Vetores Genéticos , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Interferon-alfa/análise , Imageamento por Ressonância Magnética , Fragmentos de Peptídeos/análise , Plasminogênio/análise , Engenharia de Proteínas , Ratos , Ratos Sprague-Dawley , Retroviridae/genética , Células Tumorais Cultivadas
8.
J Neurosurg ; 97(1): 169-76, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12134908

RESUMO

OBJECT: The reliable assessment of the invasiveness of gliomas in vitro has proved elusive, because most invasion assays inadequately model in vivo invasion in its complexity. Recently, organotypical brain cultures were successfully used in short-term invasion studies on glioma cell lines. In this paper the authors report that the invasiveness of human glioma biopsy specimens directly implanted into rodent brain slices by using the intraslice implantation system (ISIS) can be quantified with precision. The model was first validated by the demonstration that, in long-term studies, established glioma cells survive in the ISIS and follow pathways of invasion similar to those in vivo. METHODS: Brain slices (400 microm thick) from newborn mice were maintained on millicell membranes for 15 days. Cells from two human and one rodent glioblastoma multiforme (GBM) cell lines injected into the ISIS were detected by immunohistochemistry or after transfection with green fluorescent protein-containing vectors. Preferential migration along blood vessels was identified using confocal and fluorescent microscopy. Freshly isolated (< or = 24 hours after removal) 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate-prelabeled human glioma biopsy specimens were successfully implanted in 19 (83%) of 23 cases, including 12 GBMs and seven lower grade gliomas (LGGs). Morphometric quantification of distance and density of tumor cell invasion showed that the GBMs were two to four times more invasive than the LGGs. Heterogeneity of invasion was also observed among GBMs and LGGs. Directly implanted glioma fragments were more invasive than spheroids derived from the same biopsy specimen. CONCLUSIONS: The ISIS combines a high success rate, technical simplicity, and detailed quantitative measurements and may, therefore, be used to study the invasiveness of biopsy specimens of gliomas of different grades.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Invasividade Neoplásica/patologia , Animais , Biópsia , Técnicas Citológicas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...