Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neurooncol ; 126(1): 37-45, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26376657

RESUMEN

Historically, intra-arterial (IA) drug administration for malignant brain tumors including glioblastoma multiforme (GBM) was performed as an attempt to improve drug delivery. With the advent of percutaneous neuorovascular techniques and modern microcatheters, intracranial drug delivery is readily feasible; however, the question remains whether IA administration is safe and more effective compared to other delivery modalities such as intravenous (IV) or oral administrations. Preclinical large animal models allow for comparisons between treatment routes and to test novel agents, but can be expensive and difficult to generate large numbers and rapid results. Accordingly, we developed a murine model of IA drug delivery for GBM that is reproducible with clear readouts of tumor response and neurotoxicities. Herein, we describe a novel mouse model of IA drug delivery accessing the internal carotid artery to treat ipsilateral implanted GBM tumors that is consistent and reproducible with minimal experience. The intent of establishing this unique platform is to efficiently interrogate targeted anti-tumor agents that may be designed to take advantage of a directed, regional therapy approach for brain tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Infusiones Intraarteriales , Animales , Encéfalo/patología , Neoplasias Encefálicas/patología , Línea Celular , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/instrumentación , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Examen Neurológico , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Neurooncol ; 95(1): 37-48, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19415456

RESUMEN

Despite the male preponderance for developing glial tumors and a body of published literature that suggests a female gender advantage for long term survival in both human and animal studies, there have been relatively few rigorous investigations into the hormonal effects on glial tumor growth. In a previous study, we concluded that estrogen played a major role in the female survival bias seen in an intracerebral nude rat model of glioblastoma multiforme. Here we explore the potential therapeutic effect of exogenous estradiol delivery in nude rats with orthotopic glioblastoma tumors and examine the mechanism of action of estradiol on reducing tumor growth in this animal model. We administered estradiol, in several dosing regimens, to male, female and ovariectomized nude rats in a survival study. Brain sections, taken at various time points in tumor progression, were analyzed for estrogen receptor protein, proliferative index and apoptotic index. Estradiol increased survival of male, female and ovariectomized nude rats with intracerebral U87MG tumors, in a gender specific manner. The estradiol mediated effect occurred early in tumor progression, and appeared to be caused in-part by an increase in apoptotic activity. It remains unclear if estradiol's effect is direct or indirect and if it is estrogen receptor mediated. Estradiol-based or adjunctive therapy may be beneficial in treating GBM and further study is clearly warranted.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Estradiol/farmacología , Glioblastoma/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Humanos , Inmunoglobulina G/farmacología , Masculino , Melfalán/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Trasplante de Neoplasias , Ovariectomía , Ratas , Ratas Desnudas , Receptores de Estrógenos/metabolismo , Análisis de Supervivencia , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Clin Neurosci ; 68: 117-122, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31340886

RESUMEN

Brain metastasis (BM) is a common neurologic complication of cancers such as lung, breast, and melanoma. Recently, there has been a shift in treatment of BM from whole brain radiation therapy to stereotactic radiosurgery (SRS) and the success is dependent on tumor volume. While most metastases grow over time, data on growth rate is lacking. Therefore, we document volume changes of metastases before treatment. We retrospectively reviewed MRI imaging records of 82 patients with a total of 294 BMs, treated in our cancer center by one neurosurgeon and one radiation oncologist with Gamma Knife SRS over a three-year period. We measured tumor volume at the time of diagnosis and compared with tumor volume on the day of treatment. Volumes were compared using the Wilcoxon signed-rank test. Lung, melanoma and breast made up the majority of metastases diagnosed. More than 75% of tumors grew and these changes in volume and percent changes in volume were statistically significant. Thirty percent of tumors doubled in size before treatment. Patients with the largest mean pretreatment tumor size were urgently treated within 6 days, yet still demonstrated the largest change in volume. This study is one of the first to document volume changes of brain metastases from the time of diagnosis to SRS treatment. Our results indicate that brain metastases can grow rapidly and it is imperative that we streamline patient management processes to minimize delays in treating patients with SRS, since outcomes are dependent on tumor size.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/cirugía , Metástasis de la Neoplasia/patología , Radiocirugia/métodos , Tiempo de Tratamiento , Adulto , Anciano , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/terapia , Estudios Retrospectivos , Carga Tumoral
4.
Clin Cancer Res ; 24(11): 2642-2652, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29540489

RESUMEN

Purpose: Survivin is an inhibitor of apoptosis protein (IAP) that is highly expressed in many cancers and represents an attractive molecule for targeted cancer therapy. Although primarily regarded as an intracellular protein with diverse actions, survivin has also been identified in association with circulating tumor exosomes.Experimental Design: We have reported that active, specific vaccination with a long peptide survivin immunogen leads to the development of survivin-specific CD8-mediated tumor cell lysis and prolongation of survival in tumor-bearing mice. In addition to cellular antitumor responses, circulating anti-survivin antibodies are detected in the serum of mice and human glioblastoma patients following vaccination with the survivin immunogen.Results: Here we demonstrate that survivin is present on the outer cell membrane of a wide variety of cancer cell types, including both murine and human glioma cells. In addition, antibodies to survivin that are derived from the immunogen display antitumor activity against murine GL261 gliomas in both flank and intracranial tumor models and against B16 melanoma as well.Conclusions: In addition to immunogen-induced, CD8-mediated tumor cell lysis, antibodies to the survivin immunogen have antitumor activity in vivo Cell-surface survivin could provide a specific target for antibody-mediated tumor immunotherapeutic approaches. Clin Cancer Res; 24(11); 2642-52. ©2018 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Biomarcadores de Tumor , Membrana Celular/metabolismo , Survivin/antagonistas & inhibidores , Animales , Afinidad de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Masculino , Melanoma Experimental , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ratones , Péptidos/antagonistas & inhibidores , Péptidos/inmunología , Proteínas Recombinantes de Fusión , Survivin/química , Survivin/genética , Survivin/metabolismo
5.
Neuro Oncol ; 19(2): 186-196, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27370399

RESUMEN

Background: The survival rate for patients with glioblastoma (GBM) remains dismal. New therapies targeting molecular pathways dysregulated in GBM are needed. One such clinical-stage drug candidate, CBL0137, is a curaxin, small molecules which simultaneously downregulate nuclear factor-kappaB (NF-ĸB) and activate p53 by inactivating the chromatin remodeling complex, Facilitates Chromatin Transcription (FACT). Methods: We used publicly available databases to establish levels of FACT subunit expression in GBM. In vitro, we evaluated the toxicity and effect of CBL0137 on FACT, p53, and NF-ĸB on U87MG and A1207 human GBM cells. In vivo, we implanted the cells orthotopically in nude mice and administered CBL0137 in various dosing regimens to assess brain and tumor accumulation of CBL0137, its effect on tumor cell proliferation and apoptosis, and on survival of mice with and without temozolomide (TMZ). Results: FACT subunit expression was elevated in GBM compared with normal brain. CBL0137 induced loss of chromatin-unbound FACT, activated p53, inhibited NF-ĸB-dependent transcription, and was toxic to GBM cells. The drug penetrated the blood-brain barrier and accumulated in orthotopic tumors significantly more than normal brain tissue. It increased apoptosis and suppressed proliferation in both U87MG and A1207 tumors. Intravenous administration of CBL0137 significantly increased survival in models of early- through late-stage TMZ-responsive and -resistant GBM, with a trend toward significantly increasing the effect of TMZ in TMZ-responsive U87MG tumors. Conclusion: CBL0137 targets GBM according to its proposed mechanism of action, crosses the blood-brain barrier, and is efficacious in both TMZ-responsive and -resistant orthotopic models, making it an attractive new therapy for GBM.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Carbazoles/uso terapéutico , Proteínas de Unión al ADN/antagonistas & inhibidores , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Factores de Elongación Transcripcional/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Dacarbazina/farmacología , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Temozolomida , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Neurosci ; 24(7): 1726-33, 2004 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-14973255

RESUMEN

The mammalian brain contains neural stem cells (NSCs) that allow continued neurogenesis throughout the life of the animal. However, neurogenesis is known to decline during aging and, to the extent that neurogenesis is required for normal CNS function, this may contribute to neurodegenerative disease. Decreased neurogenesis could result from loss of NSCs or dysfunction at some later step, and distinguishing these possibilities is important for understanding the cause of the decline. However, because of the inability to distinguish NSCs from their rapidly dividing progeny in situ, it has not been possible to quantitatively assess the NSC populations in young and old animals. In this report we show that the G1 phase-specific expression of the replication factor Mcm2 is a useful marker for detecting slowly cycling putative NSCs in situ and confirm the identity of these cells using both cytosine beta-D-arabinofuranoside (Ara-C) treatment and a double nucleoside analog-labeling technique. The ability to distinguish NSCs from proliferative progenitors has allowed characterization of the expression of several markers including Nestin, Musashi, and GFAP in these different cell types. Furthermore, comparison of the NSC populations in the subventricular zones of young (2-4 months) and old (24-26 months) mice demonstrates an approximately twofold reduction in the older mice. A similar twofold reduction is also observed in the number of neurospheres recovered in culture from old relative to young animals. The reduction in the neural stem cell population documented here is sufficient to account for the reduced level of neurogenesis in old animals.


Asunto(s)
Envejecimiento/metabolismo , Desoxiuridina/análogos & derivados , Ventrículos Laterales/citología , Proteínas del Tejido Nervioso , Neuronas/citología , Células Madre/citología , Administración Oral , Animales , Antígenos de Diferenciación/biosíntesis , Bromodesoxiuridina/farmacocinética , Recuento de Células , Ciclo Celular/fisiología , División Celular , Células Cultivadas , Citarabina/farmacología , Desoxiuridina/administración & dosificación , Desoxiuridina/metabolismo , Desoxiuridina/farmacocinética , Proteína Ácida Fibrilar de la Glía/biosíntesis , Idoxuridina/administración & dosificación , Idoxuridina/metabolismo , Idoxuridina/farmacocinética , Proteínas de Filamentos Intermediarios/biosíntesis , Masculino , Ratones , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Nestina , Neuronas/metabolismo , Proteínas Nucleares/biosíntesis , Células Madre/efectos de los fármacos , Células Madre/metabolismo
7.
Cancer Lett ; 213(1): 57-65, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15312684

RESUMEN

2-Methoxyestradiol (2ME), a metabolite of estradiol (E), inhibits proliferation of various tumor cells. In this study we determined the effect of 2ME on human glioblastoma cell lines, in vitro. We compared these cells with cultured astrocytes obtained from traumatized adult rat striatum. Exposure to 2ME had a strong antiproliferative effect on human glioblastoma and caused an increase in the population of apoptotic cells, detected by flow cytometry, in some of the investigated cell lines. A significant number of cells were blocked in the G2/M phase of the cell cycle. Concurrently, the population of cells in the G1 phase decreased in all glioblastoma cell lines. Staining with Hoechst 33258 revealed abnormal nuclear morphology in the proliferating cells treated with 2ME. Treatment with 2ME induced upregulation of wild type p53 in one of the human glioblastoma cell lines as well as in proliferating adult rat astrocytes. We conclude that 2ME inhibits the growth of human glioblastoma cell lines and induces apoptosis, in vitro. This compound deserves further investigation as a treatment for gliomas.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Glioblastoma/patología , 2-Metoxiestradiol , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/veterinaria , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/veterinaria , Humanos , Neoplasias Experimentales , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba
8.
Brain Res Mol Brain Res ; 122(1): 62-70, 2004 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-14992816

RESUMEN

Divalent metal transporter 1 (DMT1), expressed in many different tissues, is responsible for the transport of a broad range of divalent metal ions. DMT1 exists in at least, four distinct isoforms which differ in both the C-terminus (termed here (-)IRE and (+)IRE) and the N-terminus (transcription proceeds from two different promoters). In the rat, two of the forms possess an additional 31 amino acids in the N-terminus (termed exon 1A) whereas the shorter forms lack this sequence (termed exon 2). Studies were performed to compare differences in expression and localization of these isoforms in low density and confluent cultures of rat astrocytes obtained from traumatized striatum and in rat C6 astrocytoma and human U87 glioblastoma. Results of these experiments reveal the presence of both the (+/-)IRE forms of DMT1 in all cultured cells examined. Western blots using affinity purified antibodies, which differentially recognize the two C-terminal species of DMT1, indicate a strong upregulation of the (+)IRE form in low density astrocyte cultures when compared to confluent cultures. Previously we reported that the (-)IRE form was present in both the nucleus and cytoplasm in neurons and neuronal like cells whereas the (+)IRE form was exclusively cytoplasmic. Similar results were found with the (-)IRE species in astrocytes and astrocytomas, i.e. nuclear and cytoplasmic distribution. This form of DMT1 also colocalizes with the early endosomal marker, EEA, suggesting that (-)IRE species may function in the transport of divalent metals. In contrast to our previous findings, however, the (+)IRE form was found predominantly localized in nucleus in both the primary and neoplastic glial cells. Interestingly, neither form of DMT1 colocalizes with the transferrin receptor. These data suggest that selective compartmentalization of specific isoforms of DMT1 imparts distinct and specialized functions that meet the changing needs of essential divalent transition metals as cofactors within cells.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Astrocitos/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteínas de Unión a Hierro/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Western Blotting/métodos , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente/métodos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioblastoma , Humanos , Masculino , Microscopía Confocal/métodos , Fragmentos de Péptidos/metabolismo , Lectinas de Plantas/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores de Transferrina/metabolismo
9.
Anticancer Res ; 30(12): 4851-60, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21187462

RESUMEN

BACKGROUND: To study the neuropathology and selected tumour markers of malignant gliomas, an animal glioma model was developed using the implantation of U87 human glioblastoma cells into chick chorioallantoic membrane. The immunohistochemical characteristics were studied and compared with an orthotopic rodent model. MATERIALS AND METHODS: The U87 cell suspension was inoculated onto the chick chorioallantoic membrane on embryonic day seven and into the brain of nude rats. Brain tumour sections were examined for various known tumour markers by routine haematoxylin and eosin staining and immunohistochemical analyses. RESULTS: The immunohistochemical analyses showed that S100 protein, glial fibrillary acidic protein and synaptophysin expressions, initially present in tissue culture, were lost in both models. Persistent kallikrein, CD68 and vimentin expressions in U87 cells, as well as in both animal tumour models, were detected. The percentage of p53-positive nuclei, which was higher in the tumours grown on the chick chorioallantoic membrane than in rats, did not correlate with the Ki-67 labelling index. Strong cathepsin expression was maintained from the cell culture to both tumour models. CD3-positive cells and numerous leukocytes, but no CD20-positive cells were detected in any of the animal samples, indicating the immunological response of the host to be primarily cellular. Stronger immune reaction for vascular endothelial growth factor in rats correlated with an observed increase in vascular proliferation in these tumours. CONCLUSION: A simple, fast-growing, cheap and well-defined chick chorioallantoic membrane model of glioma was established, providing a basis for further experimental studies of genetic and protein expression during human glioma tumourigenesis. This model may possibly replace some rodent models for selective studies.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Membrana Corioalantoides/patología , Modelos Animales de Enfermedad , Glioblastoma/patología , Adulto , Animales , Neoplasias Encefálicas/metabolismo , Embrión de Pollo , Membrana Corioalantoides/metabolismo , Femenino , Glioblastoma/metabolismo , Humanos , Inmunohistoquímica , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Esferoides Celulares , Trasplante Heterólogo
10.
Cancer Immunol Immunother ; 57(12): 1827-35, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18438666

RESUMEN

Survivin is a tumor-associated antigen (TAA) that has significant potential for use as a cancer vaccine target. To identify survivin epitopes that might serve as targets for CTL-mediated, anti-tumor responses, we evaluated a series of survivin peptides with predicted binding to mouse H2-K(b) and human HLA-A*0201 antigens in peptide-loaded dendritic cell (DC) vaccines. H2-K(b)-positive, C57BL/6 mice were vaccinated using syngeneic, peptide-loaded DC2.4 cells. Splenocytes from vaccinated mice were screened by flow cytometry for binding of dimeric H2-K(b):Ig to peptide-specific CD8+ T cells. Two survivin peptides (SVN(57-64) and SVN(82-89)) generated specific CD8+ T cells. We chose to focus on the SVN(57-64) peptide because that region of the molecule is 100% homologous to human survivin. A larger peptide (SVN(53-67)), containing multiple class I epitopes, and a potential class II ligand, was able to elicit both CD8+ CTL and CD4+ T cell help. We tested the SVN(53-67) 15-mer peptide in a therapeutic model using a peptide-loaded DC vaccine in C57BL/6 mice with survivin-expressing GL261 cerebral gliomas. This vaccine produced significant CTL responses and helper T cell-associated cytokine production, resulting in a significant prolongation of survival. The SVN(53-67) vaccine was significantly more effective than the SVN(57-64) core epitope as a cancer vaccine, emphasizing the potential benefit of incorporating multiple class I epitopes and associated cytokine support within a single peptide.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Glioma/terapia , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Glioma/inmunología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/inmunología , Proteínas Inhibidoras de la Apoptosis , Masculino , Ratones , Ratones Endogámicos C57BL , Survivin , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/uso terapéutico
11.
J Neurooncol ; 86(3): 285-96, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17928956

RESUMEN

Osteopontin (OPN) is a pleotrophic molecule that has been associated with multiple disorders of the central nervous system (CNS). Its roles in CNS malignancy are unclear but suggest that higher levels of OPN expression correlate with increased tumor grade and increased migratory capacity of tumor cells. In this study OPN cDNA was cloned into a retroviral vector and used to infect F98 Fischer rat-derived glioma cells and U87 human-derived glioblastoma multiforme (GBM) cells in vitro. Cells expressing high levels of OPN migrated less distance than control cells in vitro. This effect was not RGD mediated, but was reversed in the presence of c-Jun N-terminal kinase (JNK) inhibitor suggesting that JNK1 is an essential component of a negative feedback loop affecting OPN activated signaling cascades. Implantation of tumor cells expressing high levels of OPN into adult Fischer rats and nude rats resulted in morphologically distinct tumors and prolonged host survival relative to controls. We propose that local produced, high level OPN expression limits the malignant character of glioma cells and that the downstream mechanisms involved represent pathways that may have therapeutic value in the treatment of human CNS malignancy.


Asunto(s)
Neoplasias Encefálicas , Movimiento Celular/fisiología , Glioblastoma , Glioma , Osteopontina/metabolismo , Animales , Apoptosis/fisiología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/fisiopatología , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/fisiopatología , Glioma/metabolismo , Glioma/mortalidad , Glioma/fisiopatología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Osteopontina/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas , Análisis de Supervivencia , Factores de Tiempo , Transfección , Trasplantes
12.
Cancer Immunol Immunother ; 55(8): 918-27, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16187082

RESUMEN

The role that transforming growth factor beta1 (TGF-beta1) plays in influencing growth of glioma cells is somewhat controversial. To further understand the potential growth-regulatory effects of TGF-beta1,we constructed an animal astroglial tumor model by injecting either wild-type or virally transduced human U-87 glioblastoma cells into nude rat brains. Wild type U-87 cells produced very low amounts of TGF-beta1 and were highly tumorigenic. In contrast, U-87 cells transduced to express high levels of TGF-beta1 showed reduced tumor size in vivo, in a dose-dependent manner. This reduction in tumor size was not due to either decreased vascularity or increased apoptosis. To test whether TGF-beta1 overproduction inhibited tumor growth through an autocrine mechanism, the highest TGF-beta1 producing cells were then double transduced with a vector expressing the kinase-truncated type II TGF-beta receptor. Cells expressing high levels of truncated TGF-beta receptor were less sensitive to TGF-beta1 mediated growth inhibition in vitro and produced more aggressive tumors in vivo. The data suggest that the degree of tumorigenicity of the U-87 high-grade glioblastoma cell line may be associated with correspondingly low level of production of TGF-beta1. These results also would tend to support the possibility that TGF-beta1 may be useful in treating some high-grade gliomas.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Glioblastoma/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Northern Blotting , Western Blotting , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Fragmentación del ADN , Modelos Animales de Enfermedad , Glioblastoma/patología , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratas , Ratas Desnudas , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Transducción Genética
13.
Cancer Immunol Immunother ; 55(12): 1491-503, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16485128

RESUMEN

Survivin is a member of the inhibitor of apoptosis protein family. Gliomas and many other tumors express survivin at high levels; whereas, normal fully differentiated cells generally do not. Therefore, survivin represents a tumor-specific target for cancer vaccine therapy. It has been shown that it is possible to produce a MHC-I-restricted cellular immunologic response to survivin vaccines. To study differences in immunogenicity between murine and human survivin proteins, we vaccinated C57BL/6 mice with bone marrow dendritic cells (BMDC) transfected with expression vectors containing the murine and human survivin genes. Mice vaccinated with BMDCs expressing a truncated human survivin protein developed cytotoxic T lymphocyte to subcutaneous GL261 glioma cells and exhibited prolonged tumor-free survival compared to mice vaccinated with BMDCs transfected with vector alone (P<0.01). While mice challenged with intracerebral GL261 cells had increased survival, no cures were observed. In contrast, vaccinated mice that fully resisted subcutaneous tumor challenge were rendered resistant to intracerebral GL261 re-challenge. BMDCs transfected with the full-length human survivin molecule were significantly more effective at prolonging survival than BMDCs expressing the full-length murine survivin gene (P=0.0175). Therefore, xenogeneic differences between human and murine sequences might be exploited to develop more immunogenic tumor vaccines.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Glioma/tratamiento farmacológico , Inmunoterapia Adoptiva , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas de Neoplasias/inmunología , Animales , Antígenos Heterófilos/genética , Apoptosis , Autoinmunidad , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Vacunas contra el Cáncer/síntesis química , Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Proteínas Represoras , Survivin , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA