Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 16(5): 893-900, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18388927

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Produtos do Gene tat/genética , Técnicas de Transferência de Genes , HIV/metabolismo , Lipossomos/química , Nanopartículas/química , Animais , Encéfalo/metabolismo , DNA/metabolismo , Produtos do Gene tat/fisiologia , Terapia Genética/métodos , Humanos , Transplante de Neoplasias , Peptídeos/química , Plasmídeos/metabolismo , Ratos
2.
Neuro Oncol ; 10(2): 112-20, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18287341

RESUMO

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.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Glioma/tratamento farmacológico , Oligonucleotídeos/administração & dosagem , Telomerase/antagonistas & inibidores , Administração Intranasal , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Masculino , Ratos , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Int J Radiat Oncol Biol Phys ; 67(5): 1538-47, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17394949

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Efeito Espectador/efeitos dos fármacos , Hipóxia Celular/genética , Citosina Desaminase/uso terapêutico , Flucitosina/uso terapêutico , Glioblastoma/terapia , Tolerância a Radiação/genética , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citosina Desaminase/genética , Técnicas de Transferência de Genes , Glioblastoma/fisiopatologia , Glioblastoma/radioterapia , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Pró-Fármacos/uso terapêutico , Tolerância a Radiação/efeitos dos fármacos
4.
J Med Chem ; 50(4): 820-7, 2007 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-17253677

RESUMO

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.


Assuntos
Boranos/síntese química , Porfirinas/síntese química , Animais , Ânions , Boranos/química , Boranos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Cardiopatias/induzido quimicamente , Hemorragia/induzido quimicamente , Nefropatias/induzido quimicamente , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Pneumopatias/induzido quimicamente , Masculino , Dose Máxima Tolerável , Necrose , Porfirinas/química , Porfirinas/toxicidade , Ratos , Ratos Endogâmicos F344 , Trombocitopenia/induzido quimicamente , Aumento de Peso/efeitos dos fármacos
5.
Brain Res ; 1128(1): 181-90, 2007 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-17125752

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Convecção , Sistemas de Liberação de Medicamentos , Substâncias Macromoleculares/metabolismo , Albuminas/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Vias de Administração de Medicamentos , Líquido Extracelular/metabolismo , Glioma/tratamento farmacológico , Glioma/patologia , Lipossomos/metabolismo , Substâncias Macromoleculares/uso terapêutico , Microdiálise/métodos , Ratos , Fatores de Tempo
6.
Int J Radiat Oncol Biol Phys ; 66(1): 263-70, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16904526

RESUMO

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.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Animais , Neoplasias Encefálicas/mortalidade , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Glioblastoma/mortalidade , Humanos , Expectativa de Vida , Masculino , Tolerância a Radiação/fisiologia , Ratos , Ratos Nus , Transplante Heterólogo
7.
J Neurooncol ; 78(2): 113-21, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16554968

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral/metabolismo , Senescência Celular/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas Oncogênicas Virais/metabolismo , Telomerase/genética , Transformação Genética/genética , Técnicas de Cultura de Células/métodos , Senescência Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Desmoplaquinas/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Transferência de Genes , Humanos , Cariotipagem , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Papillomaviridae , Proteínas E7 de Papillomavirus/metabolismo , Telomerase/metabolismo , Transfecção/métodos , Vimentina/metabolismo
8.
J Control Release ; 110(2): 236-259, 2006 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-16318895

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , DNA/administração & dosagem , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Terapia Genética , Animais , Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Humanos , Lipossomos
9.
Genes Chromosomes Cancer ; 45(1): 20-30, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16130123

RESUMO

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.


Assuntos
Neoplasias Encefálicas/genética , Cromossomos Humanos Par 7/fisiologia , Glioblastoma/genética , Tolerância a Radiação/genética , Neoplasias Encefálicas/radioterapia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Mapeamento Cromossômico , Coloração Cromossômica , Cromossomos Humanos Par 7/genética , Glioblastoma/radioterapia , Humanos , Repetições de Microssatélites
10.
Neurosurgery ; 57(5): 1041-7; discussion 1041-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16284574

RESUMO

OBJECTIVE: For patients with gliomas, decreasing the tumor burden with macroscopic surgical resection may affect quality of life, time to tumor progression, and survival. Injection of bromophenol blue (BPB) may enhance intraoperative visualization of an infiltrating tumor and its margins and improve the extent of resection. In this study, we investigated the uptake of BPB in experimental rat brain tumors. METHODS: We first conducted a toxicity study with bolus intravenous injections of 5, 60, and 360 mg/kg doses of BPB in nontumor-bearing Fischer 344 rats. No adverse effects were observed in any of the animals during the 60 day observation period. We then injected 9L tumor cells intracerebrally into Fischer 344 rats and approximately 2 weeks later, administered a bolus intravenous injection of 5 to 360 mg/kg BPB. Fifteen minutes after BPB injection, we sacrificed the animals and removed their brains. In a subsequent study, we injected 180 mg/kg BPB and sacrificed animals at several time points to monitor tumor staining over time. RESULTS: The stain was clearly visible and localized to the tumor for all BPB concentrations 60 mg/kg or greater, and in an additional experiment, we found that tumor staining persisted for at least 8 hours after BPB injection. CONCLUSION: We conclude that BPB helped visualize experimental tumors at time points from a few minutes to several hours after injection. Because BPB also proved to be nontoxic to the animals at effective concentrations, we believe the compound may be potentially useful in helping neurosurgeons visualize brain tumors in humans.


Assuntos
Neoplasias Encefálicas/diagnóstico , Azul de Bromofenol , Glioma/diagnóstico , Animais , Azul de Bromofenol/administração & dosagem , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Indicadores e Reagentes/administração & dosagem , Masculino , Transplante de Neoplasias/métodos , Neoplasias Experimentais/diagnóstico , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo
11.
Cancer Res ; 65(22): 10389-93, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16288029

RESUMO

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.


Assuntos
Expressão Gênica/efeitos da radiação , Glioma/genética , Glioma/patologia , Animais , Processos de Crescimento Celular/genética , Processos de Crescimento Celular/efeitos da radiação , Linhagem Celular Tumoral , Glioma/metabolismo , Humanos , Camundongos , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Int J Radiat Oncol Biol Phys ; 63(1): 247-52, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16111595

RESUMO

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.


Assuntos
Compostos de Boro/farmacocinética , Compostos de Boro/toxicidade , Boro/farmacocinética , Neoplasias Encefálicas/metabolismo , Deuteroporfirinas/farmacocinética , Deuteroporfirinas/toxicidade , Animais , Compostos de Boro/administração & dosagem , Terapia por Captura de Nêutron de Boro , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Deuteroporfirinas/administração & dosagem , Bombas de Infusão Implantáveis , Injeções Intramusculares , Masculino , Ratos , Ratos Endogâmicos F344 , Distribuição Tecidual
13.
Proc Natl Acad Sci U S A ; 102(23): 8287-92, 2005 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-15928080

RESUMO

Defining the molecules that regulate tumor cell survival is an essential prerequisite for the development of targeted approaches to cancer treatment. Whereas many studies aimed at identifying such targets use human tumor cells grown in vitro or as s.c. xenografts, it is unclear whether such experimental models replicate the phenotype of the in situ tumor cell. To begin addressing this issue, we have used microarray analysis to define the gene expression profile of two human glioma cell lines (U251 and U87) when grown in vitro and in vivo as s.c. or as intracerebral (i.c.) xenografts. For each cell line, the gene expression profile generated from tissue culture was significantly different from that generated from the s.c. tumor, which was significantly different from those grown i.c. The disparity between the i.c gene expression profiles and those generated from s.c. xenografts suggests that whereas an in vivo growth environment modulates gene expression, orthotopic growth conditions induce a different set of modifications. In this study the U251 and U87 gene expression profiles generated under the three growth conditions were also compared. As expected, the profiles of the two glioma cell lines were significantly different when grown as monolayer cultures. However, the glioma cell lines had similar gene expression profiles when grown i.c. These results suggest that tumor cell gene expression, and thus phenotype, as defined in vitro is affected not only by in vivo growth but also by orthotopic growth, which may have implications regarding the identification of relevant targets for cancer therapy.


Assuntos
Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos
14.
Radiat Res ; 163(6): 644-53, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15913396

RESUMO

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.


Assuntos
Apoptose/efeitos da radiação , Hipóxia Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Tolerância a Radiação , Adaptação Fisiológica/efeitos da radiação , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Doses de Radiação , Proteínas Recombinantes/metabolismo , Proteína X Associada a bcl-2
15.
Int J Radiat Oncol Biol Phys ; 62(1): 207-12, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15850923

RESUMO

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.


Assuntos
Hipóxia Celular/fisiologia , Tolerância a Radiação/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Transformada/metabolismo , Linhagem Celular Transformada/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos
16.
Cancer Gene Ther ; 12(5): 449-55, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15706354

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Glioblastoma/metabolismo , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Transcrição/genética , Animais , Apoptose , Hipóxia Celular , Feminino , Glioblastoma/genética , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2
17.
Brain Res ; 1035(2): 139-53, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15722054

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Convecção , Sistemas de Liberação de Medicamentos/métodos , Lipossomos/administração & dosagem , Lipossomos/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/instrumentação , Tamanho da Partícula , Ratos , Ratos Nus
18.
Cancer Gene Ther ; 12(3): 276-83, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15650767

RESUMO

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.


Assuntos
Citosina Desaminase/uso terapêutico , Fluoruracila/uso terapêutico , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Neoplasias/terapia , Pró-Fármacos/uso terapêutico , Western Blotting , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Citosina Desaminase/genética , Elementos Facilitadores Genéticos/genética , Eritropoetina/genética , Flucitosina/metabolismo , Fluoruracila/metabolismo , Vetores Genéticos/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Vírus 40 dos Símios , Fatores de Transcrição/genética , Transfecção , Ensaio Tumoral de Célula-Tronco , Leveduras
19.
Neuro Oncol ; 6(3): 218-26, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15279714

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/prevenção & controle , Glioblastoma/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Telomerase/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/enzimologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Glioblastoma/enzimologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oligonucleotídeos/farmacologia , Ratos , Ratos Nus , Telomerase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Mol Pharm ; 1(5): 368-74, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-16026007

RESUMO

Boron neutron capture therapy (BNCT) is an adjuvant therapy that has the potential to control local tumor growth. A selective delivery of sufficient amounts of boron to individual tumor cells, compared to surrounding normal tissues, is the key for successful BNCT. We have designed and synthesized a new highly water-soluble boronated porphyrin, TABP-1, as a possible BNCT agent. When we injected the maximum tolerated dose (MTD: 15 mg/kg) of TABP-1 systemically into the tail vein of athymic rats bearing intracerebral (i.c.) human glioblastoma U-87 MG xenografts, the compound accumulated preferentially in brain tumors compared to normal brain; however, the level of boron in the tumors was less than the 30 microg/g of tissue that is generally considered necessary for BNCT. We next investigated whether convection-enhanced delivery (CED) could improve the boron distribution. The compound was administered directly into i.c. tumors using an osmotic minipump attached to a brain-infusion cannula. TABP-1 doses from 0.25 to 1.0 mg infused locally over 24 h produced tumor boron concentrations greater than those obtained by systemic administration at the MTD. For example, CED administration of 0.5 mg of TABP-1 produced a tumor boron level of 65.4 microg/g of tumor, whereas the serum level was only 0.41 microg/g (tumor to serum ratio of approximately 160:1). CED also produced relatively high tumor to normal brain ratios of approximately 5:1 for ipsilateral brain and approximately 26:1 for contralateral brain tissues at the 0.5 mg dose. Thus, we may be able to achieve therapeutic BNCT efficacy with minimal systemic toxicity or radiation-induced damage to normal tissue by administering TABP-1 using CED.


Assuntos
Compostos de Boro/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Metaloporfirinas/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Compostos de Boro/administração & dosagem , Compostos de Boro/química , Terapia por Captura de Nêutron de Boro/métodos , Neoplasias Encefálicas/química , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Convecção , Vias de Administração de Medicamentos , Sistemas de Liberação de Medicamentos/métodos , Glioblastoma/química , Glioblastoma/metabolismo , Humanos , Masculino , Metaloporfirinas/administração & dosagem , Metaloporfirinas/química , Estrutura Molecular , Ratos , Ratos Endogâmicos Lew , Ratos Nus , Fatores de Tempo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA