Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Cancer ; 123(1): 209-16, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18386816

RESUMEN

Malignant gliomas and childhood ependymomas have a high rate of treatment failure. Epidermal growth factor receptor (EGFR) activation has been implicated in the tumorigenesis and radioresistance of many cancers, including brain tumors. Therefore, combining EGFR targeting with irradiation is a potentially attractive therapeutic option. We evaluated the tyrosine kinase inhibitor gefitinib for its antitumor activity and potential to radio-sensitize in vivo in two xenograft models: an EGFR amplified glioma and an EGFR expressing ependymoma, both derived from primary tumors. When administered at 100 mg/kg for 5 consecutive days, gefitinib-induced partial tumor regression in all treated EGFR amplified IGRG88 glioma xenografts. The addition of 1 Gy of irradiation prior to gefitinib administration resulted in 5 complete and 4 partial regressions for the 9 treated tumors as well as a significant tumor growth delay of 33 days for the combined treatment compared to 19 days for each therapy alone, suggesting additive antitumor activity. Tumor regression was associated with inhibition of AKT and MAPK pathways by gefitinib. In contrast, the ependymoma IGREP83 was sensitive to irradiation, but remained resistant to gefitinib. Combined treatment was associated with inhibition of radiation-induced MAPK phosphorylation and significant induction of apoptotic cell death though radiation-induced AKT phosphorylation was maintained. Depending on the scheduling of both therapies, a trend towards superior antitumor activity was observed with combined treatment. Thus, EGFR targeting through tyrosine kinase inhibition appears to be a promising new approach in the treatment of EGFR-driven glioma, particularly in combination with radiation therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Ependimoma/tratamiento farmacológico , Ependimoma/radioterapia , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Glioma/tratamiento farmacológico , Glioma/radioterapia , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Quimioterapia Adyuvante , Niño , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Ependimoma/patología , Receptores ErbB/genética , Femenino , Citometría de Flujo , Fase G1/efectos de los fármacos , Gefitinib , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioma/patología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Radioterapia Adyuvante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Artículo en Inglés | MEDLINE | ID: mdl-16434243

RESUMEN

A liquid chromatography/tandem mass spectrometry assay of glutathione (GSH), glutathione disulfide (GSSG) and of precursors (gamma-glutamyl-cysteine, cysteinyl-glycine, cysteine, cystine, homocysteine and homocystine) was developed to study glutathione synthesis in mice liver. After iodoacetic acid derivatization, the analytes were analyzed using reversed-phase gradient HPLC and detected using multiple reaction monitoring. Linear calibrations were performed over the concentrations range of 100-10,000 ng/mL for the thiol-containing precursors and extended up to 100,000 ng/mL for GSH and GSSG. The method was validated for each compound with inter-day accuracy below 11.9% and with precision below 15%. The method showed low limits of quantitation of 100 ng/mL for each thiol-containing compound and GSSG and of 200 ng/mL for other disulfides.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Glutatión/análisis , Hígado/química , Espectrometría de Masas/métodos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
3.
Cancer Res ; 64(9): 3223-9, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126363

RESUMEN

Topoisomerase I inhibitors, such as CPT-11, are potent anticancer drugs against neuroblastoma (NB). Differentiating agents, such as retinoids, improve the survival of children with metastatic NB. To characterize the biological effects associated with exposure to CPT-11 in vivo, athymic mice bearing a human NB xenograft, named IGR-NB8 and characterized as an immature NB with poor prognostic markers, were treated with CPT-11. Prolonged stable disease was observed, resulting in an overall tumor growth delay of 115 days. During treatment, tumors differentiated into ganglioneuroblastomas (GGNB), which reverted into an immature phenotype when treatment was discontinued. In contrast, 13-cis retinoic acid failed to induce differentiation of IGR-NB8 in vivo. Tumor differentiation was associated with decreased N-myc expression, induction of p73 expression in the perinuclear area and cytoplasm, and a dramatic 35-fold decrease in topoisomerase I (topo I) catalytic activity. The full-length Mr 100,000 topo I protein was present in both pre and post-treatment immature NB xenografts. In contrast, differentiated GGNBs did not contain the Mr 100,000 protein but an intense Mr 48,000 topo I fragment. Furthermore, redistribution of the Mr 48,000 and 68,000 forms to the cytoplasm was observed in differentiated tumors. The same pattern of topo I expression and catalytic activity was observed in NBs and GGNBs obtained from pediatric patients. Our data suggest that prolonged in vivo exposure to CPT-11 induces differentiation of NB xenografts, which is associated with truncation of the topo I enzyme, relocation of the degraded forms to the cytoplasm, and decreased catalytic activity.


Asunto(s)
Camptotecina/análogos & derivados , Camptotecina/farmacología , Inhibidores Enzimáticos/farmacología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/enzimología , Inhibidores de Topoisomerasa I , Animales , Antineoplásicos Fitogénicos/farmacología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Niño , Preescolar , ADN-Topoisomerasas de Tipo I/metabolismo , Diterpenos , Femenino , Humanos , Lactante , Recién Nacido , Irinotecán , Masculino , Ratones , Ratones Desnudos , Neuroblastoma/patología , Retinaldehído/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 62(3): 764-72, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11830531

RESUMEN

Treatment of malignant gliomas remains a major challenge in adults and children because of high treatment failure. The E1B 55 kDa-gene deleted adenovirus, ONYX-015 (ONYX Pharmaceuticals), was demonstrated to replicate selectively in and lyse tumor cells. Currently ongoing clinical trials of ONYX-015 in head and neck tumors are promising. Here, we demonstrate ONYX-015-mediated cell lysis and antitumor activity in three of four s.c. human malignant glioma xenografts deriving from primary tumors. Intratumoral injections of ONYX-015, 1 x 10(8) plaque-forming units daily for 5 consecutive days, yielded significant tumor growth delay in the p53 mutant xenografts IGRG88 and the p53 wild-type IGRG93 and IGRG121 treated at an advanced tumor stage. The p53 wild-type tumors IGRG93 and IGRG121 experienced 45% and 82% complete tumor regressions. Four and 8 of 11 animals, respectively, survived tumor free 4 months after treatment. Widespread intratumoral adenoviral replication was observed in tumor cells of these two xenografts compared with only scattered replication in the p53-mutant tumors. In addition to a fast tumor growth rate, wild-type p53 status was associated with increased antitumor activity of the E1B-attenuated virus, and induction of functional p53 may therefore determine adenoviral cytolysis in tumor cells. In conclusion, ONYX-015 displayed a major antitumor activity in human xenografts derived from primary malignant glioma supporting its development in the treatment of these highly malignant tumors.


Asunto(s)
Adenoviridae/fisiología , Proteínas E1B de Adenovirus/genética , Glioblastoma/terapia , Glioblastoma/virología , Proteína p53 Supresora de Tumor/fisiología , Adenoviridae/genética , Animales , Efecto Citopatogénico Viral , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Receptores Virales/biosíntesis , Activación Transcripcional , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 64(16): 5753-9, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15313916

RESUMEN

Prognosis of malignant glioma is poor, and results of treatment remain mediocre. Conditionally replicative adenoviruses hold promise as alternative anticancer agents for the treatment of malignant glioma. Here, we evaluated the conditionally replicative adenovirus AdDelta24 and its recently developed derivative AdDelta24-p53, which expresses functional p53 tumor suppressor protein while replicating in cancer cells, for treatment of malignant glioma. In comparison to its parent AdDelta24, AdDelta24-p53 killed most malignant glioma cell lines and primary glioblastoma multiforme short-term cultures more effectively, irrespective of their p53 status. Moreover, AdDelta24-p53 caused more frequent regression and more delayed growth of IGRG121 xenografts derived from a glioblastoma multiforme in vivo. Five intratumoral injections of 10(7) pfu AdDelta24 gave 24 days median tumor growth delay (P < 0.01), 30% tumor regressions, and 30% animals surviving >120 days tumor-free or with a minimal tumor residual. The same dose of AdDelta24-p53 caused >113 days of median tumor growth delay (P < 0.001), 70% tumor regressions, and 60% animals surviving >120 days tumor-free or with a minimal tumor residual. Antitumor effects in vivo were associated with extensive conditionally replicative adenovirus replication, apoptosis induction, and tumor morphology changes, including dissociation, inflammatory cell infiltration, and necrosis. We conclude that conditionally replicative adenoviruses expressing p53 are promising new agents for treatment of malignant glioma.


Asunto(s)
Adenoviridae/fisiología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Glioma/terapia , Glioma/virología , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Femenino , Glioblastoma/genética , Glioblastoma/terapia , Glioblastoma/virología , Glioma/genética , Humanos , Ratones , Ratones Desnudos , Proteína p53 Supresora de Tumor/biosíntesis , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Gene Ther ; 10(11): 859-66, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14605672

RESUMEN

Human neuroblastoma (NB) is a highly heterogeneous childhood cancer secreting a high level of vascular endothelial growth factor (VEGF). Its vascularization has been clearly correlated with metastatic progression and poor outcome. Thus, molecules that target the vascular endothelium are regarded as new therapeutics of clinical interest. Angiostatin, an internal fragment of plasminogen containing the first four kringle structures, has been described as a powerful angiogenic inhibitor. We used a recombinant adenovirus encoding the human angiostatin kringle 1-3 directly fused to human serum albumin HSA (AdK3-HSA). Coupling to HSA has been previously shown to increase the in vivo half-life of this angiostatic factor, and to lead to tumor growth inhibition in the MDA-MB-231 carcinoma model. For the assessment of antiangiogenic gene therapy in the human NB IGR-N835 tumor model, 5 x 10(9) PFU of AdK3-HSA were intravenously injected in tumor-bearing athymic mice presenting either of the following experimental settings: early stage, established, and minimal residual tumors. No delay in tumor growth was observed in animals treated with AdK3-HSA as compared to those treated with the empty virus AdCO1. In early-stage tumors, kinetics of tumor occurrence and tumor growth were similar in AdK3-HSA- and AdCO1-treated animals. K3-HSA was found to be expressed at high levels (the mean value for the three experiments being 19.4+/-15.9 microg/ml) in the circulation of all animals up to 21-35 days after virus injection. In addition, IGR-N835 tumors were found to be highly vascularized and to release high amounts of angiogenic factors, in particular VEGF (665+/-370 pg/mg total protein). Thus, in spite of high circulating levels, K3-HSA may be unable to displace the NB proangiogenic switch. In this regard, a more promising target to inhibit NB angiogenesis seems to be the VEGF/VEGFR system.


Asunto(s)
Adenoviridae/genética , Angiostatinas/metabolismo , Técnicas de Transferencia de Gen , Neuroblastoma/genética , Neuroblastoma/patología , Angiostatinas/genética , Animales , Antineoplásicos/farmacología , División Celular/genética , Cisplatino/farmacología , Vectores Genéticos/sangre , Vectores Genéticos/genética , Humanos , Inyecciones Intravenosas , Ratones , Ratones Desnudos , Neuroblastoma/metabolismo , Albúmina Sérica/genética , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Chemother Pharmacol ; 51(5): 385-94, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12736760

RESUMEN

New active drugs are needed for the treatment of primary brain tumors in both children and adults. S16020 is a cytotoxic olivacine derivative that inhibits topoisomerase II. The aim of the study was to determine its antitumor activity in athymic mice bearing subcutaneous medulloblastoma (IGRM33, 34, 57) and glioblastoma (IGRG88, 93, 121) xenografts treated at an advanced stage of tumor growth in comparison with that of doxorubicin. Animals were randomly assigned to receive i.v. S16020 or doxorubicin weekly for three consecutive weeks. The optimal dose was 80 mg/kg per week. S16020 demonstrated a significant antitumor activity in two out of three medulloblastoma xenografts. IGRM57 xenografts were highly sensitive with 100% tumor regressions and a tumor growth delay (TGD) of 102 days, while one of eight IGRM34 xenografts showed a partial regression with a TGD of 16 days. Doxorubicin was significantly more active than S16020 in these two models. IGRM33, a model established from a tumor in relapse after chemotherapy and radiotherapy, was refractory to both drugs. S16020 demonstrated a significant antitumor activity in the three glioblastoma xenografts evaluated. The wild-type p53 IGRG93 xenograft was highly sensitive with 100% tumor regressions and a TGD of 54 days. IGRG121 (wt p53) and IGRG88 (mutant p53) were moderately sensitive with TGDs of 33 and 23 days, respectively. Doxorubicin showed greater activity in two of these models. All six xenografts exhibited low expression of mdr1 as quantitated by RT-PCR, and no correlation was found with the activity of either drug. Conversely, a low activity of the two drugs was significantly associated with a high expression of MRP1 in medulloblastomas. Finally, no relationship was observed between drug sensitivity to either drug and expression of their target, topoisomerase IIalpha. In conclusion, S16020 and doxorubicin showed significant antitumor activity in brain tumor xenografts treated at an advanced stage of tumor growth. Their activity was related to MRP1 expression in medulloblastomas.


Asunto(s)
Carbazoles/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Doxorrubicina/farmacología , Glioblastoma/tratamiento farmacológico , Meduloblastoma/tratamiento farmacológico , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Piridinas/farmacología , Inhibidores de Topoisomerasa II , Animales , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/veterinaria , Quimioterapia Combinada , Femenino , Regulación de la Expresión Génica , Glioblastoma/patología , Glioblastoma/veterinaria , Meduloblastoma/patología , Meduloblastoma/veterinaria , Ratones , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
8.
J Control Release ; 100(1): 29-40, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15491808

RESUMEN

Previous results have shown that PEG-coated poly(hexadecylcyanoacrylate) (PEG-PHDCA) nanospheres displayed a significant accumulation within an orthotopic 9L gliosarcoma model, after i.v. administration to rats. Hence, the aim of the present study was to evaluate in the same model the pre-clinical efficacy of this carrier when loaded with Doxorubicin, an anticancer drug which poorly distributes in the CNS. Free and nanospheres-encapsulated Doxorubicin were administered with a multiple dose treatment. Their maximum tolerated dose (MTD) and increase in life span were respectively assessed in healthy and intracranially 9L-bearing rats. A comparative biodistribution study of Doxorubicin-loaded and unloaded PEG-PHDCA nanospheres was also performed in the tumor-bearing group. The results showed that the cumulative MTD of nanoparticulate doxorubicin was 1.5 times higher than this of free Doxorubicin. Nevertheless, encapsulated Doxorubicin was unable to elicit a better therapeutic response in the 9L gliosarcoma. Biodistribution study revealed that the Doxorubicin-loaded nanospheres accumulated to a 2.5-fold lesser extent in the 9L tumor as compared to the unloaded nanospheres and that they were mainly localized in the lungs and the spleen. Such a typical profile indicated aggregation with plasma proteins as a consequence of the positive surface charge of these loaded particles; this ionic interaction resulting from drug encapsulation was mainly responsible for 9L treatment failure.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Gliosarcoma/tratamiento farmacológico , Nanotubos , Animales , Doxorrubicina/farmacocinética , Inmunohistoquímica , Masculino , Dosis Máxima Tolerada , Ratas , Ratas Endogámicas F344 , Distribución Tisular
9.
Drug Metab Dispos ; 35(2): 306-14, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17132762

RESUMEN

Busulfan is an example of a drug eliminated through glutathione S-transferase (GST)-catalyzed conjugation with reduced glutathione (GSH). We studied the pharmacokinetics and toxicity of busulfan in C57BL6 mice in correlation with liver GST activity and GSH synthesis by accurate determination of precursors, namely, gamma-glutamyl-cysteine and cysteine. A significantly lower incidence of acute toxicity was observed in mice receiving busulfan 16.5 mg/kg twice a day compared with animals receiving 33 mg/kg once a day. In both cases, a total dose of 132 mg/kg was administered over 4 days. The difference in toxicity was explained by pharmacokinetics since a strong induction of clearance was observed only in animals treated twice daily. Induction of metabolism was correlated with an increase in liver cysteine content and enhanced glutathione synthesis rate, whereas GST activity was unchanged. To our knowledge, this is the first time that in vivo flux of GSH synthesis has been shown to be closely related to a drug plasma clearance and toxicity. These results allow hypothesizing that GSH liver synthesis may directly influence busulfan clearance in humans with possible implications in the occurrence of hepatic veno-occlusive disease.


Asunto(s)
Busulfano/toxicidad , Glutatión/biosíntesis , Animales , Trasplante de Médula Ósea , Busulfano/farmacocinética , Interacciones Farmacológicas , Glutatión Transferasa/metabolismo , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Hígado/metabolismo , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL
10.
J Gene Med ; 7(5): 584-94, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15651070

RESUMEN

BACKGROUND: Advanced stage and relapsing neuroblastoma (NB) has a poor prognosis with frequent treatment failures, warranting new treatment options and enhanced local tumor control. Treatment with conditionally replicative adenoviruses (CRAds) has shown effectiveness in various preclinical cancer models, but has not yet been evaluated for local control of NB. Here, we tested the efficacy of the CRAd AdDelta24 and of two AdDelta24 derivatives against NB. Derivative AdDelta24-425S11 infects cells deficient in coxsackie/adenovirus receptor (CAR) via the epidermal growth factor receptor (EGFR). Derivative AdDelta24-p53 expresses the tumor suppressor protein p53 to promote oncolysis. METHODS: Expression of CAR and EGFR, and p53 pathway and DNA damage responses were analyzed in six NB cell lines and two xenografts derived from primary NB using immunohistochemistry, reporter gene transactivation, Western blot and fluorescence-activated cell sorting (FACS) analysis. Efficacy of AdDelta24, AdDelta24-425S11 and AdDelta24-p53 against NB was evaluated in vitro by cell viability analysis and in vivo by monitoring subcutaneous xenograft tumor growth in mice and by histological analysis of treated tumors. RESULTS: Neuroblastoma cell lines were sensitive to oncolysis by AdDelta24, with a higher susceptibility of those with functional p53 and intact DNA damage responses. Compared to AdDelta24, AdDelta24-p53 exhibited enhanced oncolytic potency on all NB cell lines independent of their p53 status and AdDelta24-425S11 was more effective against CAR-low IGR-NB8 cells. Moreover, five daily intratumoral injections of 10(8) plaque-forming units (pfu) of AdDelta24-p53 or AdDelta24-425S11 into subcutaneous IGR-NB8 and IGR-N91 xenografts at an advanced tumor stage yielded significant tumor growth delays (TGD). In contrast, at this dose, AdDelta24 did not cause significant TGD of neuroblastoma xenografts. Injection of AdDelta24-p53 was associated with extensive cell lysis, apoptotic cell death, and fibrous fascicles in the tumors. CONCLUSION: CRAds expressing p53 and targeted towards EGFR appear promising new agents for local control in the treatment of neuroblastoma.


Asunto(s)
Adenoviridae/fisiología , Receptores ErbB/metabolismo , Terapia Genética , Neuroblastoma/terapia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Efecto Citopatogénico Viral/fisiología , Daño del ADN , Femenino , Marcación de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Neuroblastoma/virología , Receptores Virales/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Pharmacol Exp Ther ; 303(3): 928-36, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12438511

RESUMEN

The aim of the present study was to evaluate the tumor accumulation of radiolabeled long-circulating poly(ethylene glycol) (PEG)-coated hexadecylcyanoacrylate nanospheres and non-PEG-coated hexadecylcyanoacrylate nanospheres (used as control), after intravenous injection in Fischer rats bearing intracerebrally well established 9L gliosarcoma. Both types of nanospheres showed an accumulation with a retention effect in the 9L tumor. However, long-circulating nanospheres concentrated 3.1 times higher in the gliosarcoma, compared with non-PEG-coated nanospheres. The tumor-to-brain ratio of pegylated nanospheres was found to be 11, which was in accordance with the ratios reported for other carriers tested for brain tumor targeting such as long-circulating liposomes or labels for magnetic resonance imaging. In addition, a 4- to 8-fold higher accumulation of the PEG-coated carriers was observed in normal brain regions, when compared with control nanospheres. Using a simplified pharmacokinetic model, two different mechanisms were proposed to explain this higher concentration of PEG-coated nanospheres in a tumoral brain. 1) in the 9L tumor, the preferential accumulation of pegylated nanospheres was attributable to their slower plasma clearance, relative to control nanospheres. Diffusion/convection was the proposed mechanism for extravasation of the nanospheres in the 9L interstitium, across the altered blood-brain barrier. 2) In addition, PEG-coated nanospheres displayed an affinity with the brain endothelial cells (normal brain region), which may not be considered as the result of a simple diffusion/convection process. The exact underlying mechanism of such affinity deserves further investigation, since it was observed to be as important as specific interactions described for immunoliposomes with the blood-brain barrier.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Cianoacrilatos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Gliosarcoma/metabolismo , Nanotecnología/métodos , Polietilenglicoles/farmacocinética , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Cianoacrilatos/administración & dosificación , Gliosarcoma/tratamiento farmacológico , Masculino , Microesferas , Polietilenglicoles/administración & dosificación , Ratas , Ratas Endogámicas F344 , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA