Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Ther ; 16(5): 893-900, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18388927

RESUMEN

We evaluated gene transfer using PEGylated bioresponsive nanolipid particles (NLPs) containing plasmid DNA administered by convection-enhanced delivery (CED) into orthotopically implanted U87-MG tumors in rat brain. We hypothesized that attachment of the human immunodeficiency virus trans-acting transcriptional activator peptide (TATp) to pH-sensitive, reduction-sensitive NLPs would increase gene transfer. TATp was attached either directly to a phospholipid (TATp-lipid) or via a 2-kd polyethylene glycol (PEG) to a lipid (TATp-PEG-lipid). Incorporation of 0.3 mol% TATp-PEG into pH-sensitive NLPs improved transfection 100,000-fold compared to NLPs in culture. In the brain or implanted tumors, the TATp-PEG restricted NLP convection to regions adjacent to the infusion catheter. Gene transfer in the brain from TATp-PEG NLPs, measured by green fluorescent protein (GFP) expression, was substantially greater than from NLPs adjacent to the catheter. Gene transfer using TATp-PEG NLPs, measured by luciferase expression, was 8-12-fold greater than from a 1,2-dioleoyl-3-trimethylammonium-propane/cholesterol cationic lipoplex but 13-27-fold less than from the NLPs. Brain luciferase expression was localized in perivascular macrophages. Thus a cationic ligand, such as the TATp-PEG-lipid, can dramatically increase gene expression in culture, in the normal brain, and in implanted tumors; however, restriction of NLP distribution to the vicinity of the infusion catheter reduces the absolute level of gene transfer.


Asunto(s)
Regulación de la Expresión Génica , Productos del Gen tat/genética , Técnicas de Transferencia de Gen , VIH/metabolismo , Liposomas/química , Nanopartículas/química , Animales , Encéfalo/metabolismo , ADN/metabolismo , Productos del Gen tat/fisiología , Terapia Genética/métodos , Humanos , Trasplante de Neoplasias , Péptidos/química , Plásmidos/metabolismo , Ratas
2.
Neuro Oncol ; 10(2): 112-20, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18287341

RESUMEN

The blood-brain barrier is a substantial obstacle for delivering anticancer agents to brain tumors, and new strategies for bypassing it are greatly needed for brain-tumor therapy. Intranasal delivery provides a practical, noninvasive method for delivering therapeutic agents to the brain and could provide an alternative to intravenous injection and convection-enhanced delivery. We treated rats bearing intracerebral human tumor xenografts intranasally with GRN163, an oligonucleotide N3'-->P5'thio-phosphoramidate telomerase inhibitor. 3'-Fuorescein isothiocyanate (FITC)-labeled GRN163 was administered intranasally every 2 min as 6 microl drops into alternating sides of the nasal cavity over 22 min. FITC-labeled GRN163 was present in tumor cells at all time points studied, and accumulation of GRN163 peaked at 4 h after delivery. Moreover, GRN163 delivered intranasally, daily for 12 days, significantly prolonged the median survival from 35 days in the control group to 75.5 days in the GRN163-treated group. Thus, intranasal delivery of GRN163 readily bypassed the blood-brain barrier, exhibited favorable tumor uptake, and inhibited tumor growth, leading to a prolonged lifespan for treated rats compared to controls. This delivery approach appears to kill tumor cells selectively, and no toxic effects were noted in normal brain tissue. These data support further development of intranasal delivery of tumor-specific therapeutic agents for brain tumor patients.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Glioma/tratamiento farmacológico , Oligonucleótidos/administración & dosificación , Telomerasa/antagonistas & inhibidores , Administración Intranasal , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratas , Ratas Desnudas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Radiat Oncol Biol Phys ; 67(5): 1538-47, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17394949

RESUMEN

PURPOSE: Treatment of glioblastoma (GBM) is limited by therapeutic ratio; therefore, successful therapy must be specifically cytotoxic to cancer cells. Hypoxic cells are ubiquitous in GBM, and resistant to radiation and chemotherapy, and, thus, are logical targets for gene therapy. In this study, we investigated whether cytosine deaminase (CD)/5-fluorocytosine (5-FC) enzyme/prodrug treatment induced a bystander effect (BE) and/or radiosensitization in hypoxic GBM cells. METHODS AND MATERIALS: We stably transfected cells with a gene construct consisting of the SV40 minimal promoter, nine copies of a hypoxia-responsive element, and the yeast CD gene. During hypoxia, a hypoxia-responsive element regulates expression of the CD gene and facilitates the conversion of 5-FC to 5-fluorouracil, a highly toxic antimetabolite. We used colony-forming efficiency (CFE) and immunofluorescence assays to assess for BE in co-cultures of CD-expressing clone cells and parent, pNeo- or green fluorescent protein-stably transfected GBM cells. We also investigated the radiosensitivity of CD clone cells treated with 5-FC under hypoxic conditions, and we used flow cytometry to investigate treatment-induced cell cycle changes. RESULTS: Both a large BE and radiosensitization occurred in GBM cells under hypoxic conditions. The magnitude of the BE depended on the number of transfected cells producing CD, the functionality of the CD, the administered concentration of 5-FC, and the sensitivity of cell type to 5-fluorouracil. CONCLUSION: Hypoxia-inducible CD/5-FC therapy in combination with radiation therapy shows both a pronounced BE and a radiosensitizing effect under hypoxic conditions.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Efecto Espectador/efectos de los fármacos , Hipoxia de la Célula/genética , Citosina Desaminasa/uso terapéutico , Flucitosina/uso terapéutico , Glioblastoma/terapia , Tolerancia a Radiación/genética , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citosina Desaminasa/genética , Técnicas de Transferencia de Gen , Glioblastoma/fisiopatología , Glioblastoma/radioterapia , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Profármacos/uso terapéutico , Tolerancia a Radiación/efectos de los fármacos
4.
J Med Chem ; 50(4): 820-7, 2007 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-17253677

RESUMEN

Three structurally similar tetraphenylporphyrins bearing polyhedral borane anions have been synthesized and their toxicological profiles obtained in rats. These conjugates were found to have quite different acute toxicities as manifested at the maximum tolerated dose (MTD). When given at the MTD and observed over 28 days, the most acutely toxic porphyrin was found to be devoid of toxicity, as measured by blood chemistry panels. The remaining two less acutely toxic compounds both elicited significant changes, characterized by moderate to severe thrombocytopenia, failure to gain weight normally and changes in liver enzymes indicative of mild hepatotoxicity. All toxic effects were transient, with platelets rebounding to above normal levels at day 28. We conclude that thrombocytopenia is the dose limiting toxicity for boronated porphyrins in mammals and suggest that these effects may be due to the porphyrin, not the borane or carborane.


Asunto(s)
Boranos/síntesis química , Porfirinas/síntesis química , Animales , Aniones , Boranos/química , Boranos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Cardiopatías/inducido químicamente , Hemorragia/inducido químicamente , Enfermedades Renales/inducido químicamente , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Enfermedades Pulmonares/inducido químicamente , Masculino , Dosis Máxima Tolerada , Necrosis , Porfirinas/química , Porfirinas/toxicidad , Ratas , Ratas Endogámicas F344 , Trombocitopenia/inducido químicamente , Aumento de Peso/efectos de los fármacos
5.
Brain Res ; 1128(1): 181-90, 2007 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-17125752

RESUMEN

A retro-convection enhanced delivery (R-CED) method has been developed to improve the entry of intravenously administered therapeutics within solid brain tumors. R-CED uses an osmotic gradient to withdraw brain interstitial fluid (ISF) in a controlled manner via an implanted microdialysis catheter. Withdrawal of ISF increases the local tissue specific gravity in normal brain and increases twofold the extravasation of intravenous Evans blue (EB) albumin in normal brain and in an orthotopic 9L tumor. R-CED also increases the extravasation of 70 nm fluorescent liposomes fivefold in the 9L tumor. Thus the transmembrane osmotic gradient induces movement of substances in the blood into the tissue parenchyma. Following probe removal, the magnitude of the R-CED effect on EB-albumin extravasation decreases to control values within 1.5 h in normal brain; however, the effect persists beyond 6 h in the tumor. There was no evidence of histologic damage to the neurons at either 6 h or 2 weeks after R-CED. These studies establish the feasibility of applying R-CED to increase the distribution of systemically administered drugs in both the normal tissue-tumor margin as well as in the central tumor core, holding forth the possibility of improved antitumor drug efficacy.


Asunto(s)
Encéfalo/metabolismo , Convección , Sistemas de Liberación de Medicamentos , Sustancias Macromoleculares/metabolismo , Albúminas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Vías de Administración de Medicamentos , Líquido Extracelular/metabolismo , Glioma/tratamiento farmacológico , Glioma/patología , Liposomas/metabolismo , Sustancias Macromoleculares/uso terapéutico , Microdiálisis/métodos , Ratas , Factores de Tiempo
6.
Cancer Res ; 65(22): 10389-93, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16288029

RESUMEN

The effect of radiation on gene expression has been most frequently studied using tissue culture models. To determine the influence of experimental growth condition on radiation-induced changes in gene expression, microarray analysis was done on two human glioma cell lines (U87 and U251) grown in tissue culture and as s.c. or i.c. xenografts. Compared with tissue culture, the number of genes, whose expression was affected by radiation in both cell lines, was increased in the s.c. xenografts and further increased in the orthotopic tumors. Furthermore, in each growth condition, radiation modulated the expression of a different set of genes. In addition, whereas there were few commonly affected genes after irradiation of U87 and U251 in tissue culture, there were 729 common changes after orthotopic irradiation. These results indicate that the influence of the orthotopic environment on radiation-induced modulation of gene expression in glioma cells was both quantitative and qualitative. Moreover, they suggest that investigations of the functional consequence of radiation-induced gene expression will require accounting for experimental growth conditions.


Asunto(s)
Expresión Génica/efectos de la radiación , Glioma/genética , Glioma/patología , Animales , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/efectos de la radiación , Línea Celular Tumoral , Glioma/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Trasplante Heterólogo , Células Tumorales Cultivadas
7.
Int J Radiat Oncol Biol Phys ; 66(1): 263-70, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16904526

RESUMEN

PURPOSE: We investigated the effects of fractionated radiation treatments on the life spans of athymic rats bearing intracerebral brain tumors. METHODS AND MATERIALS: U-251 MG or U-87 MG human glioblastoma cells were implanted into the brains of athymic rats, and the resulting tumors were irradiated once daily with various doses of ionizing radiation for 5 consecutive days or for 10 days with a 2-day break after Day 5. RESULTS: Five daily doses of 1 and 1.5 Gy, and 10 doses of 0.75 and 1 Gy, cured some U-251 MG tumors. However, five daily doses of 0.5 Gy increased the survival time of animals bearing U-251 MG tumors 5 days without curing any animals of their tumors. Ten doses of 0.3 Gy given over 2 weeks extended the lifespan of the host animals 9 days without curing any animals. For U-87 MG tumors, 5 daily doses of 3 Gy produced an increased lifespan of 8 days without curing any animals, and 10 doses of 1 Gy prolonged lifespan 5.5 days without curing any animals. The differences in extension of life span between the 5- and 10-fraction protocols were minor for either tumor type. CONCLUSION: The finding that the U-251 MG tumors are more sensitive than U-87 MG tumors, despite the fact that U-251 MG tumors contain many more hypoxic cells than U-87 MG tumors, suggests the intrinsic cellular radiosensitivities of these cell lines are more important than hypoxia in determining their in vivo radiosensitivities.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Animales , Neoplasias Encefálicas/mortalidad , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Fraccionamiento de la Dosis de Radiación , Glioblastoma/mortalidad , Humanos , Esperanza de Vida , Masculino , Tolerancia a Radiación/fisiología , Ratas , Ratas Desnudas , Trasplante Heterólogo
8.
J Control Release ; 110(2): 236-259, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16318895

RESUMEN

Brain tumor patients face a poor prognosis despite significant advances in tumor imaging, neurosurgery and radiation therapy. Potent chemotherapeutic drugs fail when used to treat brain tumors because biochemical and physiological barriers limit drug delivery into the brain. In the past decade a number of strategies have been introduced to increase drug delivery into the brain parenchyma. In particular, direct drug administration into the brain tumor has shown promising results in both animal models and clinical trials. This technique is well suited for the delivery of liposome and polymer drug carriers, which have the potential to provide a sustained level of drug and to reach cellular targets with improved specificity. We will discuss the current approaches that have been used to increase drug delivery into the brain parenchyma in the context of fluid and solute transport into, through and from the brain, with a focus on liposome and polymer drug carriers.


Asunto(s)
Neoplasias Encefálicas/terapia , ADN/administración & dosificación , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Terapia Genética , Animales , Encéfalo/anatomía & histología , Encéfalo/fisiología , Humanos , Liposomas
9.
Cancer Res ; 63(8): 1962-8, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12702589

RESUMEN

Vascular endothelial growth factor (VEGF) is thought to promote tumor growth and angiogenesis. Whereas VEGF is up-regulated in only a portion of anaplastic astrocytoma (AA), it is overexpressed in most glioblastoma multiforme (GBM), and the level of expression is correlated with grade of glioma. To explore the possibility that VEGF may act as a driving force in the progression of AA to GBM, the VEGF isoforms VEGF(121) and VEGF(165) were overexpressed in genetically modified, mutant H-Ras-transformed human astrocytes that on intracranial implantation form AA-like tumors. The ability of the VEGF isoforms to stimulate growth, angiogenesis, oxygenation, and the formation of necrotic GBM-like tumors was then monitored. The parental mutant H-Ras-modified astrocytes expressed four times more endogenous VEGF than normal human astrocytes, but on intracranial implantation formed hypovascular, hypoxic, small AA-like tumors. Whereas these modest levels of VEGF overexpression were insufficient to drive oxygenation and GBM formation, an additional 8-fold increase in VEGF expression mediated by retroviral infection with constructs encoding either VEGF (121) or VEGF (165) resulted in cells which, after intracranial implantation, formed tumors that were larger, more vascular, and better oxygenated than those formed by the mutant H-ras parental cells. However, the tumors formed by the cells expressing exogenous VEGF (121) or VEGF (165) retained the phenotype of AA, lacking areas of necrosis that are the hallmark of the GBM phenotype. These results suggest that whereas the VEGF(121) and VEGF(165) isoforms can contribute to glioma vascularization, oxygenation, and growth, they do not in and of themselves drive the formation of the GBM phenotype.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Factores de Crecimiento Endotelial/biosíntesis , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Linfocinas/biosíntesis , Oxígeno/metabolismo , Astrocitos/metabolismo , Astrocitos/fisiología , Astrocitoma/irrigación sanguínea , Astrocitoma/genética , Astrocitoma/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , División Celular/fisiología , Progresión de la Enfermedad , Factores de Crecimiento Endotelial/genética , Genes ras , Glioblastoma/irrigación sanguínea , Glioblastoma/genética , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fenotipo , Isoformas de Proteínas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
Cancer Gene Ther ; 12(3): 276-83, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15650767

RESUMEN

One important feature of human solid tumors is the presence of a hypoxic microenvironment. Under hypoxia, genes that contain a hypoxia-response element (HRE) can be activated by the binding of hypoxia-inducible factor-1. To reach the goal of selectively killing tumor cells in a hypoxic microenvironment using a gene therapy approach, we developed a cytosine deaminase (CD) gene construct (pH9YCD2) that contains an HRE gene enhancer. CD is an enzyme that catalyzes the conversion of noncytotoxic 5-fluorocytosine (5-FC) to the cytotoxic and radiosensitizing drug 5-fluorouracil (5-FU). Yeast CD was cloned into an SV40 promoter-based mammalian expression vector, and an HRE enhancer was inserted in front of the promoter. Human glioblastoma U-87 MG cells were transfected with pH9YCD2. Western blots revealed that CD was strongly expressed under hypoxic conditions (0.3-1% O2), whereas only minor CD expression was seen under normoxic conditions. To confirm that the expressed CD enzyme retains catalytic activity, we performed a 5-FC/5-FU-conversion assay in which 5-FC was incubated with the lysates of pH9YCD2-transfected cells. The percentage of conversion from 5-FC to 5-FU was 63% under hypoxia versus 13% under normoxia. In vitro, cell viability and colony-forming efficiency assays demonstrated that the gene construct was able to significantly kill glioblastoma cells in a hypoxia-dependent manner. In addition, 5-FC treatment of hypoxic pH9YCD2-transfected cells produced a marked bystander effect, which could be a distinct advantage for gene therapy. If this construct exhibits antitumor efficacy in vivo, it may have promise as an antitumor agent in humans.


Asunto(s)
Citosina Desaminasa/uso terapéutico , Fluorouracilo/uso terapéutico , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Neoplasias/terapia , Profármacos/uso terapéutico , Western Blotting , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Citosina Desaminasa/genética , Elementos de Facilitación Genéticos/genética , Eritropoyetina/genética , Flucitosina/metabolismo , Fluorouracilo/metabolismo , Vectores Genéticos/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Virus 40 de los Simios , Factores de Transcripción/genética , Transfección , Ensayo de Tumor de Célula Madre , Levaduras
11.
Cancer Gene Ther ; 12(5): 449-55, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15706354

RESUMEN

The effectiveness of radiation therapy for human brain tumors is limited by the presence of radiation-resistant hypoxic cells. In order to improve patient outcomes, therapeutic methods that increase hypoxic cell killing must be developed. To investigate the possibility of using the hypoxic tumor microenvironment itself as a target for gene therapy, we stably transfected U-251 MG human glioblastoma cells with constructs containing the suicide gene Bax under the regulation of a nine-copy concatemer of hypoxia responsive elements (HREs). Previously, we demonstrated that the expression of BAX protein under anoxic conditions in transfected U-251 MG clones leads to increased cell killing in vitro. Our recent studies revealed that HIF-1alpha induction under anoxic conditions occurs prior to the increase in BAX expression, thereby implicating HIF-1 induction as the basis of BAX upregulation. To test the effect of BAX-mediated cell killing in vivo, we implanted five stably transfected clones subcutaneously into the flanks of athymic mice. Compared to nontransfected controls, tumor growth in four of five clones was significantly retarded. Histopathological analysis demonstrated decreased hypoxic fractions and increased amounts of apoptosis in clone-derived tumors. These results suggest that the tumor microenvironment is sufficiently hypoxic to trigger HRE-mediated cell killing via the BAX apoptotic pathway.


Asunto(s)
Proteínas de Unión al ADN/genética , Glioblastoma/metabolismo , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Transcripción/genética , Animales , Apoptosis , Hipoxia de la Célula , Femenino , Glioblastoma/genética , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2
12.
Int J Radiat Oncol Biol Phys ; 63(1): 247-52, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16111595

RESUMEN

PURPOSE: To investigate the toxicity, biodistribution, and convection-enhanced delivery (CED) of a boronated porphyrin (BOPP) that was designed for boron neutron capture therapy and photodynamic therapy. METHODS AND MATERIALS: For the toxicity study, Fischer 344 rats were injected with graded concentrations of BOPP (35-100 mg/kg) into the tail vein. For boron biodistribution studies, 9L tumor-bearing rats received BOPP either systematically (intravenously) or locally. RESULTS: All rats that received 70 mg/kg BOPP and 70% of rats that received < or = 60 mg/kg BOPP i.v. either had to be euthanized or died within 4 days of injection. In the biodistribution study, boron levels were relatively high in liver, kidney, spleen, and adrenal gland tissue, and moderate levels were found in all other organs. The maximum tumor boron concentration was 21.4 mug/g at 48 h after i.v. injection; this concentration of boron in brain tumors is at the low end of the range considered optimal for therapy. In addition, the tumor/blood ratio (approximately 1.2) was not optimal. When BOPP was delivered directly into intracerebral 9L tumors with CED, we obtained tumor boron concentrations much greater than those obtained by i.v. injection. Convection-enhanced delivery of 1.5 mg BOPP produced an average tumor boron level of 519 mug/g and a tumor/blood ratio of approximately 1850:1. CONCLUSIONS: Our study demonstrates that changing the method of BOPP delivery from i.v. to CED significantly enhances the boron concentration in tumors and produces very favorable tumor/brain and tumor/blood ratios.


Asunto(s)
Compuestos de Boro/farmacocinética , Compuestos de Boro/toxicidad , Boro/farmacocinética , Neoplasias Encefálicas/metabolismo , Deuteroporfirinas/farmacocinética , Deuteroporfirinas/toxicidad , Animales , Compuestos de Boro/administración & dosificación , Terapia por Captura de Neutrón de Boro , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Deuteroporfirinas/administración & dosificación , Bombas de Infusión Implantables , Inyecciones Intramusculares , Masculino , Ratas , Ratas Endogámicas F344 , Distribución Tisular
13.
Int J Radiat Oncol Biol Phys ; 62(1): 207-12, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15850923

RESUMEN

PURPOSE: We sought to determine whether hypoxia-induced radioresistance is mediated by the transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha). METHODS AND MATERIALS: We used 2 mouse embryonic fibroblast cell lines transformed with H-ras and TAg, 1 HIF-1alpha+/+ and the other HIF-1alpha-/-. Cell were exposed to either 95% air and 5% CO2 (normoxic conditions) or 0.2% O2, 94.8% N2, and 5% CO2 (hypoxic conditions) for 4 hours. Cells were then irradiated and subjected to clonogenic survival assays. RESULTS: Whereas neither +/+ ras/TAg nor -/- ras/TAg cells expressed HIF-1alpha under normoxic conditions, hypoxia induced expression of HIF-1alpha only in +/+ ras/TAg cells, confirming the absence of HIF-1alpha in -/- ras/TAg cells. Clonogenic survival curves for +/+ ras/TAg and -/- ras/TAg cells under normoxia and hypoxia demonstrated that hypoxia increased radioresistance in both cell lines to the same degree. At 1-log cell kill, the +/+ ras/TAg and -/- ras/TAg cells had an identical oxygen enhancement ratio of 1.28 +/- 0.09 and nearly identical oxygen enhancement ratios at 2-log cell kill. CONCLUSION: In our system of transformed mouse embryonic fibroblasts, hypoxia-mediated radiation resistance is independent of HIF-1alpha.


Asunto(s)
Hipoxia de la Célula/fisiología , Tolerancia a Radiación/fisiología , Factores de Transcripción/metabolismo , Animales , Línea Celular Transformada/metabolismo , Línea Celular Transformada/efectos de la radiación , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones
14.
Radiat Res ; 163(6): 644-53, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15913396

RESUMEN

One major challenge in treating glioblastoma multiforme (GBM) has been the presence of radiation-resistant hypoxic cells. The pro-apoptosis protein BAX has been reported to be a possible radiation sensitizer of cancer cells; however, to our knowledge, no studies have reported on the effects of BAX on radiation sensitivity under hypoxic conditions. Therefore, in this study, we specifically overexpressed murine Bax in hypoxic cells in an attempt to enhance radiation-induced cell killing. We have previously stably transfected U-251 MG and U-87 MG human GBM cells with constructs containing murine Bax under the regulation of nine copies of hypoxia-responsive elements (HREs). During hypoxia, the transcriptional complex hypoxia-inducible factor 1 (HIF1) forms and binds to HRE; this binding facilitates the transcription of downstream genes. In the experiments reported here, two protocols were used. In the first protocol, parent and clone cells were exposed to graded doses of X rays under hypoxic conditions, left hypoxic for 0, 4, 16 or 24 h, and then assayed for clonogenic cell survival. In the second protocol, cells were incubated under hypoxic conditions for 20 h, irradiated with graded doses under hypoxia, then left in hypoxic conditions for 4 h before being assayed for clonogenic cell survival. Western blots showed that we had successfully increased Bax expression in both U-251 MG and U-87 MG Bax clone cells after 16 h of hypoxic incubation, yet dose-response curves showed no difference in radiation-induced cell killing between control non-Bax-expressing pNeo clone cells and the U-251 MG Bax clone cells using either protocol. In U-87 MG cells, the first protocol showed no difference in radiation response between control pNeo clone cells and the Bax clone, similar to the results obtained in U-251 cells. However, the second protocol revealed that Bax overexpression did render these cells more sensitive to radiation under hypoxic conditions. Therefore, we conclude that whether Bax is a radiation enhancer under hypoxia not only is cell line-dependent but also depends on when the Bax overexpression occurs.


Asunto(s)
Apoptosis/efectos de la radiación , Hipoxia de la Célula/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Glioblastoma/metabolismo , Glioblastoma/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tolerancia a Radiación , Adaptación Fisiológica/efectos de la radiación , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Dosis de Radiación , Proteínas Recombinantes/metabolismo , Proteína X Asociada a bcl-2
15.
Brain Res ; 1035(2): 139-53, 2005 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-15722054

RESUMEN

We have investigated the role of diameter, charge, and steric shielding on the brain distribution of liposomes infused by convection enhanced delivery (CED) using both radiolabeled and fluorescent-labeled particles. Liposomes of 40 and 80-nm diameter traveled the same distance but penetrated significantly less than a 10-kDa dextran; whereas 200-nm-diameter liposomes penetrated less than 80 nm liposomes. A neutral liposome shielded by polyethylene glycol (PEG; 2 kDa; 10% by mole) penetrated significantly farther than an unshielded liposome. Even when shielded with PEG, positive surface charge (10% by mole) significantly reduced the penetration radius compared to a neutral or negative charged liposome (10% by mole). A mathematical CED model including a term for liposome cell binding was applied to analyze the radius of particle penetration. Neutral liposomes had a binding constant of k=0.0010+/-0.0002 min-1, whereas for positive charged liposomes k increased 50-fold. The binding constant was independently verified using a degradable lipid radiolabel that eliminated from the brain with a 9.9+/-2.0 h half-life, equivalent to the calculated elimination constant k=0.0012+/-0.0002 min-1. During CED, liposomes accumulated in a subpopulation of perivascular cells within the brain. A non-degradable lipid radiolabel showed that lipid components remained within these perivascular brain cells for at least 2 days. To reduce this uptake, 100-fold molar excess of non-labeled liposomes were co-infused with labeled liposomes, which significantly increased liposome penetration. These studies suggest that optimization of therapeutic CED using particles such as drug-loaded liposomes, polymeric nanoparticles, non-viral DNA complexes, and viruses will require a strategy to overcome particle binding and clearance by cells within the CNS.


Asunto(s)
Encéfalo/metabolismo , Convección , Sistemas de Liberación de Medicamentos/métodos , Liposomas/administración & dosificación , Liposomas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos/instrumentación , Tamaño de la Partícula , Ratas , Ratas Desnudas
16.
Neoplasia ; 5(4): 288-96, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14511400

RESUMEN

We used the standard comet assay successfully to generate in vitro dose-response curves under oxic and hypoxic conditions. We then made mixtures of cells that had been irradiated with 3 and 9 Gy of X-rays to simulate two subpopulations in a tumor, but efforts to accurately detect and quantify the subpopulations using the standard comet assay were unsuccessful. Therefore, we investigated a modified comet assay to determine whether it could be used for measuring hypoxia in our model systems. U251 MG cells were grown as subcutaneous tumors in athymic mice; U251 MG and U87 MG cells were grown as intracerebral (i.c.) tumors in athymic rats. Animals were injected with RSU 1069, irradiated, and euthanized. Tumors and normal brains were removed, and the cells were analyzed using a modified comet assay. Differences in comet tail moment distributions between tumor and contralateral normal brain, using tail moments at either the 25th or 50th percentile in each distribution, were taken as measures of the degree of tumor hypoxia. For U251 MG tumors, there was a positive relationship between tumor size and the degree of hypoxia, whereas preliminary data from U87 MG i.c. tumors showed less hypoxia and no apparent relationship between tumor size and hypoxia.


Asunto(s)
Neoplasias Encefálicas/patología , Ensayo Cometa/métodos , Hipoxia , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Radiación Ionizante , Ratas , Ratas Desnudas
17.
Neuro Oncol ; 6(3): 218-26, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15279714

RESUMEN

Telomerase is a ribonucleoprotein complex that elongates telomeric DNA and appears to play an important role in cellular immortalization of cancers. Because telomerase is expressed in the vast majority of malignant gliomas but not in normal brain tissues, it is a logical target for gliomaspecific therapy. The telomerase inhibitor GRN163, a 13-mer oligonucleotide N3'-->P5' thio-phosphoramidate (Geron Corporation, Menlo Park, Calif.), is complementary to the template region of the human telomerase RNA subunit hTR. When athymic mice bearing U-251 MG human brain tumor xenografts in their flanks were treated intratumorally with GRN163, a significant growth delay in tumor size was observed (P < 0.01 in all groups) as compared to the tumor size in mice receiving a mismatched oligonucleotide or the carrier alone. We also investigated biodistribution of the drug in vivo in an intracerebral rat brain-tumor model. Fluorescein-labeled GRN163 was loaded into an osmotic minipump and infused directly into U-251 MG brain tumors over 7 days. Examination of the brains revealed that GRN163 was present in tumor cells at all time points studied. When GRN163 was infused into intracerebral U-251 MG tumors shortly after their implantation, it prevented their establishment and growth. Lastly, when rats with larger intracerebral tumors were treated with the inhibitor, GRN163 increased animal survival times. Our results demonstrate that the antitelomerase agent GRN163 inhibits growth of glioblastoma in vivo, exhibits favorable intracerebral tumor uptake properties, and prevents the growth of intracerebral tumors. These findings support further development of this compound as a potential anticancer agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/prevención & control , Glioblastoma/tratamiento farmacológico , Oligonucleótidos/uso terapéutico , Telomerasa/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/enzimología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Glioblastoma/enzimología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligonucleótidos/farmacología , Ratas , Ratas Desnudas , Telomerasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
In Vivo ; 16(1): 55-60, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11980362

RESUMEN

BACKGROUND: We have developed xenografts of human glioblastoma (GBM) and established the baseline growth parameters and histopathological features of these tumors. MATERIALS-METHODS: Cells from 4 different human GBM cell lines were injected into the right caudate-putamen of brain in athymic rats. We measured tumor weights and the estimated survival time of each rat. RESULTS-CONCLUSION: U-251 MG and U-87 MG cells produced solid intracerebral tumors with a 100% tumor take rate, while SF-767 and SF-126 cells did not grow in the brains of athymic rats. Under the conditions employed, U-87 MG tumors grew faster than U-251 MG tumors, but both types of tumors exhibited reproducible growth characteristics from animal to animal. There was heterogeneity in the growth characteristics and histologies between the 2 tumor types, indicating that these tumor models might be useful for simulating some of the heterogeneity that occurs between GBM in humans.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Neoplasias Experimentales/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Longevidad , Masculino , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Factores de Tiempo , Células Tumorales Cultivadas
19.
Genes Chromosomes Cancer ; 45(1): 20-30, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16130123

RESUMEN

Glioblastoma multiforme (GM) is the most lethal form of brain tumor, with a median survival of approximately 1 year. Treatment options are limited. Radiation therapy is a common form of treatment, but many tumors are resistant. In earlier studies, we found that gain of chromosome 7 is associated with radiation resistance in human primary GM. In this study, we extend that result to a model system in which we transferred chromosome 7 to recipient cells and confirmed radiation resistance as a function of chromosome 7 gain. We identified three candidate regions on chromosome 7 that conferred radiation resistance in our model system.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 7/fisiología , Glioblastoma/genética , Tolerancia a Radiación/genética , Neoplasias Encefálicas/radioterapia , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Mapeo Cromosómico , Pintura Cromosómica , Cromosomas Humanos Par 7/genética , Glioblastoma/radioterapia , Humanos , Repeticiones de Microsatélite
20.
J Neurooncol ; 78(2): 113-21, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16554968

RESUMEN

Even though meningiomas are the second most common brain tumor in adults, little is known about the molecular basis of their growth and development. The lack of suitable cell culture model systems is an impediment to this understanding. Most studies on meningiomas rely on primary, early passage cell lines that eventually senesce or a few established cell lines that have been derived from aggressive variants of meningiomas. We have isolated three primary meningioma cell lines that are negative for telomerase activity. We can overcome the senescence of a Grade III derived meningioma cell line by expressing the telomerase catalytic subunit (hTERT), whereas Grade I meningioma cell lines require the expression of the human papillomavirus E6 and E7 oncogenes in conjunction with hTERT. Meningioma cell lines, immortalized in this manner, maintain their pre-transfection morphology and form colonies in vitro. We have confirmed the meningothelial origin of these cell lines by assessing expression of vimentin and desmoplakin, characteristic markers for meningiomas. Additionally, we have karyotyped these cell lines using array CGH and shown that they represent a spectrum of the genetic diversity seen in primary meningiomas. Thus, these cell lines represent novel cellular reagents for investigating the molecular oncogenesis of meningiomas.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral/metabolismo , Senescencia Celular/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas Oncogénicas Virales/metabolismo , Telomerasa/genética , Transformación Genética/genética , Técnicas de Cultivo de Célula/métodos , Senescencia Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Desmoplaquinas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Humanos , Cariotipificación , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Papillomaviridae , Proteínas E7 de Papillomavirus/metabolismo , Telomerasa/metabolismo , Transfección/métodos , Vimentina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA