Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Med ; 3(12): 1354-61, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9396605

RESUMEN

Intratumoral implantation of murine cells modified to produce retroviral vectors containing the herpes simplex virus-thymidine kinase (HSV-TK) gene induces regression of experimental brain tumors in rodents after ganciclovir (GCV) administration. We evaluated this approach in 15 patients with progressive growth of recurrent malignant brain tumors. Antitumor activity was detected in five of the smaller tumors (1.4 +/- 0.5 ml). In situ hybridization for HSV-TK demonstrated survival of vector-producing cells (VPCs) at 7 days but indicated limited gene transfer to tumors, suggesting that indirect, "bystander," mechanisms provide local antitumor activity in human tumors. However, the response of only very small tumors in which a high density of vector-producing cells had been placed suggests that techniques to improve delivery and distribution of the therapeutic gene will need to be developed if clinical utility is to be achieved with this approach.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/terapia , Ganciclovir/uso terapéutico , Terapia Genética , Vectores Genéticos , Retroviridae/genética , Timidina Quinasa/genética , Adulto , Animales , Trasplante de Células , Femenino , Técnicas de Transferencia de Gen , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Timidina Quinasa/biosíntesis , Trasplante Heterólogo
2.
Oncol Res ; 9(3): 111-8, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9220496

RESUMEN

Cyclopentenyl cytosine (CPEC) exerts an antiproliferative effect against a wide variety of human and murine tumor lines, including a panel of human gliosarcoma and astrocytoma lines. This effect is produced primarily by the 5'-triphosphate metabolite CPEC-TP, an inhibitor of cytidine-5'-triphosphate (CTP) synthase (EC 6.3.4.2). Because previous studies with human glioma cell lines utilized cells in long-term tissue culture, we have undertaken to determine whether the activity of CPEC in such model systems is also demonstrable in freshly excised human glioblastoma cells. Glioma cells obtained at surgery and in log phase growth were exposed to the drug at levels ranging from 0.01 to 1 microM for 24 h, and CPEC-TP and CTP levels were determined by HPLC. Dose-dependent accumulation of CPEC-TP was accompanied by a concomitant decrease in CTP pools, with 50% depletion of the latter being achieved at a CPEC level of ca. 0.1 microM. Human glioma cell proliferation was inhibited 50% by 24-h exposure to 0.07 microM CPEC. Postexposure decay of CPEC-TP was slow, with a half-time of 30 h. DNA cytometry showed a dose-dependent shift in cell cycle distribution, with an accumulation of cells in S-phase. The pharmacological effects of CPEC on freshly excised glioblastoma cells are quantitatively similar to those seen in a range of established tissue culture lines, including human glioma, colon carcinoma, and MOLT-4 lymphoblasts, supporting the recommendation that the drug may be advantageous for the treatment of human glioblastoma.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Citidina/análogos & derivados , Glioblastoma/patología , Inhibidores de Crecimiento/farmacología , Adulto , División Celular/efectos de los fármacos , Citidina/farmacología , Citidina Trifosfato/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Nucleótidos/metabolismo , Células Tumorales Cultivadas
4.
Neuroreport ; 6(9): 1330-2, 1995 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-7669997

RESUMEN

Results of previous studies have suggested differences in the regulatory mechanisms in striatal striosomes and matrix following interruption of dopaminergic input to the striatum by MPTP in the monkey. In the present study we have investigated the possibility that stimulation of dopamine receptors by apomorphine modifies glucose metabolism differentially in the two striatal compartments of unilaterally MPTP-lesioned monkeys. Apomorphine treatment was found to result in higher rates of glucose utilization in the denervated striatum than in the intact hemisphere. Furthermore, the effect was more robust in the striosomes than in the matrix, thus providing evidence for differential functional and/or metabolic regulation in striatal striosomes and matrix in parkinsonian syndromes.


Asunto(s)
Apomorfina/farmacología , Cuerpo Estriado/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Autorradiografía , Femenino , Glucosa/metabolismo , Macaca mulatta , Masculino , Enfermedad de Parkinson Secundaria/inducido químicamente , Receptores Dopaminérgicos/efectos de los fármacos
5.
Cancer Gene Ther ; 2(2): 87-95, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7621261

RESUMEN

The use of intrathecal, retroviral-mediated transfer of the herpes simplex thymidine kinase (HStk) gene and subsequent ganciclovir (GCV) administration has recently been shown to improve survival in a rat model of leptomeningeal carcinomatosis. Clinical application of this approach is attractive because access to the cerebrospinal fluid (CSF) space is relatively noninvasive and distribution of producer cells and vectors may be facilitated by circulation of CSF, overcoming distribution problems inherent in solid tumors. However, meningeal inflammation, transduction and injury to normal CNS tissue, proliferation of the xenogeneic producer cells in the subarachnoid space, immune-mediated injury, and development of hydrocephalus are possible complications of intraventricular or intrathecal administration of vector-producer cells. In addition, the dynamics of producer cell and vector distribution in the CSF are unknown. To address these issues, we evaluated the safety of this approach for gene delivery and assessed the dynamics of distribution of producer cells and retroviral vectors in rats and non-human primates. In rats, transduction of normal central nervous system (CNS) structures surrounding the subarachnoid space was evaluated after intrathecal and intraventricular injections of beta-galactosidase and HStk vector-producer cells, with and without GCV. In primates, beta-galactosidase and HStk vector-producer cells were injected intraventricularly and GCV was administered either intrathecally or intravenously. Toxicity was evaluated by neurologic examination, serial gadolinium-enhanced MRI scans of the brain, and blood and CSF profiles. A subgroup of monkeys received repeated intraventricular injection of vector-producer cells and intravenous GCV. The titer of retroviral-vector was measured in cisternal and lumbar CSF samples after repeated producer cell injection.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Células 3T3/trasplante , Ganciclovir/toxicidad , Terapia Genética/métodos , Vectores Genéticos , Proteínas Recombinantes de Fusión/metabolismo , Timidina Quinasa/genética , Proteínas Virales/genética , Animales , Encefalopatías/etiología , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/virología , Genes Reporteros , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Vectores Genéticos/toxicidad , Supervivencia de Injerto , Inyecciones Intraventriculares , Inyecciones Espinales , Macaca mulatta/sangre , Macaca mulatta/líquido cefalorraquídeo , Imagen por Resonancia Magnética , Ratones , Virus de la Leucemia Murina de Moloney/genética , Ratas/sangre , Ratas/líquido cefalorraquídeo , Proteínas Recombinantes de Fusión/uso terapéutico , Simplexvirus/enzimología , Simplexvirus/genética , Simplexvirus/inmunología , Espacio Subaracnoideo , Timidina Quinasa/inmunología , Timidina Quinasa/uso terapéutico , Distribución Tisular , Proteínas Virales/inmunología , Proteínas Virales/uso terapéutico , beta-Galactosidasa/biosíntesis
6.
Cancer Res ; 55(6): 1306-9, 1995 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-7882327

RESUMEN

Cyclopentenylcytosine (CPEC; NSC 375575) is a pyrimidine nucleoside analogue that has potent antitumor effects when tested in vitro and also when tested in experimental tumors outside the central nervous system. CPEC exerts its antiproliferative effect through inhibition of CTP synthetase and consequent depletion of CTP and dCTP pools required for cell replication. Due to its poor penetration of the bloodbrain barrier, CPEC has failed to demonstrate therapeutic efficacy in experimental brain tumors after systemic administration. We therefore examined the in vivo activation, distribution, and antitumor effect of CPEC after long-term regional infusion of the drug directly into experimental brain tumors in rats. HPLC analysis of CPEC incubated with homogenized human brain and brain tumor tissue showed minimal degradation of the drug over 24 h. Analysis of rat cerebral 9L gliosarcoma infused with tritium-labeled CPEC demonstrated intratumoral accumulation of the active metabolite CPEC-triphosphate and concomitant depletion of CTP to a much greater extent in tumor tissue than in the adjacent brain. Tumor tissue UTP also decreased, but no significant effects on other ribonucleoside triphosphates were detected. Only trace amounts (< 1%) of CPEC and its metabolites reached peripheral sites, including the liver and kidneys, after intratumoral infusion. Rats treated with continuous intratumoral infusion of CPEC for 4 weeks using s.c. implanted osmotic pumps survived significantly longer than control rats receiving intratumoral saline or i.p. CPEC (P < 0.0001). Long-term intratumoral infusion of CPEC was not associated with any detectable toxicity. Our results support the feasibility of using intratumoral administration of CPEC as a regional therapy for malignant brain tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Citidina/análogos & derivados , Gliosarcoma/tratamiento farmacológico , Animales , Biotransformación , Neoplasias Encefálicas/metabolismo , Citidina/administración & dosificación , Citidina/farmacocinética , Citidina/uso terapéutico , Citidina Trifosfato/metabolismo , Estabilidad de Medicamentos , Gliosarcoma/metabolismo , Ratas , Ratas Endogámicas F344 , Distribución Tisular
7.
J Neurosurg ; 82(1): 70-6, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7815137

RESUMEN

Among the appealing features of adenoviruses as vectors for transfer of genes into the central nervous system (CNS) are that they are not neurotoxic, they can accommodate the insertion of several large genes, they are not associated with the hazards of insertional mutagenesis, and they can be concentrated to a high-titer preparation. The authors evaluated the feasibility of using adenovirally mediated gene transfer into cultured human glioma cells and in rat models of solid brain tumors and meningeal cancer. Replication-deficient adenoviral vector particles carrying a nuclear-localizing lacZ gene were injected into established 9L cerebral gliomas in Fischer rats. In addition, the adenoviral vector was injected into the subarachnoid space, either simultaneously with intrathecal tumor inoculation or after establishing leptomeningeal cancer. The brains and spinal cords were removed at various intervals for histochemical evaluation for beta-galactosidase activity using X-Gal staining. Additional rats received a stereotactic intracerebral injection of the vector into normal brain. No clinical abnormalities were observed in the injected rats. Injection of the adenoviral vector into normal brain resulted in diffuse transduction of astrocytes, microglia, neurons, and endothelial cells at the injection site. Injection of a high-concentration vector preparation into cerebral gliomas resulted in effective tumor transduction. Intrathecal injection of the vector in rats with meningeal cancer resulted in transduction of the infiltrating tumor in the subarachnoid space when injections were given simultaneously with, or 7 days after, tumor inoculation. Transduction rates of both solid and leptomeningeal tumors correlated with the number of injected particles. These results suggest that adenoviral vectors can efficiently transduce solid brain tumors and that the vectors survive in the cerebrospinal fluid for a sufficient period of time to allow leptomeningeal tumor transduction. Adenoviral vector should be evaluated for its potential use in therapeutic gene transfer approaches in malignancies of the CNS.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Técnicas de Transferencia de Gen , Glioma/terapia , Neoplasias Meníngeas/terapia , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Expresión Génica , Glioma/genética , Glioma/patología , Operón Lac , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas , beta-Galactosidasa
8.
Cancer Res ; 54(11): 2923-7, 1994 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8187079

RESUMEN

Phenylacetate is a naturally occurring plasma component that suppresses the growth of tumor cells and induces differentiation in vitro. To evaluate the in vivo potential and preventive and therapeutic antitumor efficacy of sodium phenylacetate against malignant brain tumors, Fischer 344 rats (n = 50) bearing cerebral 9L gliosarcomas received phenylacetate by continuous s.c. release starting on the day of tumor inoculation (n = 10) using s.c. osmotic minipumps (550 mg/kg/day for 28 days). Rats with established brain tumors (n = 12) received continuous s.c. phenylacetate supplemented with additional daily i.p. dose (300 mg/kg). Control rats (n = 25) were treated in a similar way with saline. Rats were sacrificed during treatment for electron microscopic studies of their tumors, in vivo proliferation assays, and measurement of phenylacetate levels in the serum and cerebrospinal fluid. Treatment with phenylacetate extended survival when started on the day of tumor inoculation (P < 0.01) or 7 days after inoculation (P < 0.03) without any associated adverse effects. In the latter group, phenylacetate levels in pooled serum and cerebrospinal fluid samples after 7 days of treatment were in the therapeutic range as determined in vitro (2.45 mM in serum and 3.1 mM in cerebrospinal fluid). Electron microscopy of treated tumors demonstrated marked hypertrophy and organization of the rough endoplasmic reticulum, indicating cell differentiation, in contrast to the scant and randomly distributed endoplasmic reticulum in tumors from untreated animals. In addition, in vitro studies demonstrated dose-dependent inhibition of the rate of tumor proliferation and restoration of anchorage dependency, a marker of phenotypic reversion. Phenylacetate, used at clinically achievable concentrations, prolongs survival of rats with malignant brain tumors through induction of tumor differentiation. Its role in the treatment of brain tumors and other cancers should be explored further.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/prevención & control , Gliosarcoma/mortalidad , Gliosarcoma/prevención & control , Fenilacetatos/uso terapéutico , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/ultraestructura , División Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Gliosarcoma/metabolismo , Gliosarcoma/patología , Gliosarcoma/ultraestructura , Microscopía Electrónica , Trasplante de Neoplasias , Fenilacetatos/sangre , Fenilacetatos/líquido cefalorraquídeo , Ratas , Ratas Endogámicas F344 , Ensayo de Tumor de Célula Madre
9.
Cancer Res ; 54(8): 2141-5, 1994 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8174120

RESUMEN

In meningeal carcinomatosis, retroviral vector-producer cells can be introduced into the thecal sac and circulate in the cerebrospinal fluid to reach malignant tumor cells in the leptomeninges, release vector particles, and selectively infect and transfer a gene of interest to these cells. Gene transfer experiments with the lacZ gene and in vitro retroviral titer measurements showed that retroviral vectors can survive in the cerebrospinal fluid, retain their infectivity, and successfully transduce tumor cells. To examine the potential of intrathecal gene therapy, we evaluated the antitumor efficacy of in situ transduction with the herpes simplex-thymidine kinase gene followed by ganciclovir therapy in a rat model of leptomeningeal neoplasia. Fischer rats were inoculated via a subarachnoid catheter implanted at the upper thoracic level, and thymidine kinase vector-producer cells were injected into the subarachnoid space the day of tumor inoculation. Seven days later, rats received ganciclovir for 14 days by daily i.p. injections (30 mg/kg/ml) or intrathecal injections (25 micrograms/kg or 600 micrograms/kg) for 14 days. To evaluate possible enhancement of tumor eradication by the ability of helper virus to package the vector in the cells and further extend gene transfer, additional rats received thymidine kinase vector-producer cells that had been previously coinfected with a replication-competent retrovirus (4070A). In all groups, control rats received i.p. or intrathecal saline injections. Ganciclovir administration i.p. resulted in significant prolongation of survival in rats given injections of thymidine kinase vector-producer cells. Injection of producer cells coinfected with the 4070A retrovirus did not improve antitumor efficacy. Intrathecal administration of ganciclovir (low and high doses) did not extend survival; histological examination of the spinal cords showed elimination of the infiltrative tumor in the leptomeninges, but residual tumor mass was present at the inoculation site, consistent with limited penetration of topical ganciclovir into the tumor. These results support the potential application of gene therapy using the thymidine kinase/ganciclovir approach for treatment of meningeal carcinomatosis.


Asunto(s)
Ganciclovir/uso terapéutico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Neoplasias Meníngeas/terapia , Simplexvirus/enzimología , Timidina Quinasa/biosíntesis , Células 3T3 , Animales , Células Clonales , Vectores Genéticos , Inyecciones Espinales , Neoplasias Meníngeas/patología , Ratones , Ratas , Ratas Endogámicas F344 , Simplexvirus/genética , Médula Espinal/patología , Timidina Quinasa/genética , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
10.
Neurosci Lett ; 127(2): 155-9, 1991 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-1652716

RESUMEN

Quantitative autoradiography was used to study [3H]naloxone binding in the striatum of normal monkeys and monkeys made hemiparkinsonian by the unilateral infusion of the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The density of [3H]naloxone binding sites was significantly higher in the caudate and putamen on the MPTP-treated side of hemiparkinsonian monkeys, as compared with binding on the untreated side and in the striatum of normal monkeys. A more extensive patchy distribution of binding sites was evident throughout the striatum on the MPTP-treated side than seen in the striatum of the untreated side or in normal striatum.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Cuerpo Estriado/metabolismo , Naloxona/metabolismo , Enfermedad de Parkinson Secundaria/metabolismo , Animales , Femenino , Lateralidad Funcional , Macaca mulatta , Masculino , Receptores Opioides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA