Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(1)2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38193532

RESUMEN

Epilepsy has a profound impact on quality of life. Despite the development of new antiseizure medications (ASMs), approximately one-third of affected patients have drug-refractory epilepsy and are nonresponsive to medical treatment. Nearly all currently approved ASMs target neuronal activity through ion channel modulation. Recent human and animal model studies have implicated new immunotherapeutic and metabolomic approaches that may benefit patients with epilepsy. In this Review, we detail the proinflammatory immune landscape of epilepsy and contrast this with the immunosuppressive microenvironment in patients with glioma-related epilepsy. In the tumor setting, excessive neuronal activity facilitates immunosuppression, thereby contributing to subsequent glioma progression. Metabolic modulation of the IDH1-mutant pathway provides a dual pathway for reversing immune suppression and dampening seizure activity. Elucidating the relationship between neurons and immunoreactivity is an area for the prioritization and development of the next era of ASMs.


Asunto(s)
Epilepsia Refractaria , Epilepsia , Glioma , Animales , Humanos , Calidad de Vida , Epilepsia/tratamiento farmacológico , Epilepsia/etiología , Glioma/complicaciones , Glioma/tratamiento farmacológico , Sistema Inmunológico , Microambiente Tumoral
2.
Clin Cancer Res ; 27(20): 5669-5680, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34433651

RESUMEN

PURPOSE: The epigenetic mechanisms involved in transcriptional regulation leading to malignant phenotype in gliomas remains poorly understood. Topoisomerase IIB (TOP2B), an enzyme that decoils and releases torsional forces in DNA, is overexpressed in a subset of gliomas. Therefore, we investigated its role in epigenetic regulation in these tumors. EXPERIMENTAL DESIGN: To investigate the role of TOP2B in epigenetic regulation in gliomas, we performed paired chromatin immunoprecipitation sequencing for TOP2B and RNA-sequencing analysis of glioma cell lines with and without TOP2B inhibition and in human glioma specimens. These experiments were complemented with assay for transposase-accessible chromatin using sequencing, gene silencing, and mouse xenograft experiments to investigate the function of TOP2B and its role in glioma phenotypes. RESULTS: We discovered that TOP2B modulates transcription of multiple oncogenes in human gliomas. TOP2B regulated transcription only at sites where it was enzymatically active, but not at all native binding sites. In particular, TOP2B activity localized in enhancers, promoters, and introns of PDGFRA and MYC, facilitating their expression. TOP2B levels and genomic localization was associated with PDGFRA and MYC expression across glioma specimens, which was not seen in nontumoral human brain tissue. In vivo, TOP2B knockdown of human glioma intracranial implants prolonged survival and downregulated PDGFRA. CONCLUSIONS: Our results indicate that TOP2B activity exerts a pleiotropic role in transcriptional regulation of oncogenes in a subset of gliomas promoting a proliferative phenotype.


Asunto(s)
Neoplasias Encefálicas/genética , ADN-Topoisomerasas de Tipo II/fisiología , Epigénesis Genética/fisiología , Glioma/genética , Intrones/fisiología , Oncogenes/fisiología , Proteínas de Unión a Poli-ADP-Ribosa/fisiología , Regiones Promotoras Genéticas/fisiología , Animales , Neoplasias Encefálicas/enzimología , Regulación Neoplásica de la Expresión Génica , Glioma/enzimología , Humanos , Ratones
3.
Neurooncol Adv ; 3(1): vdab085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34345820

RESUMEN

BACKGROUND: Meningiomas are the most common tumor arising within the cranium of adults. Despite surgical resection and radiotherapy, many meningiomas invade the brain, and many recur, often repeatedly. To date, no chemotherapy has proven effective against such tumors. Thus, there is an urgent need for chemotherapeutic options for treating meningiomas, especially those that enhance radiotherapy. Palbociclib is an inhibitor of cyclin-dependent kinases 4 and 6 that has been shown to enhance radiotherapy in preclinical models of other cancers, is well-tolerated in patients, and is used to treat malignancies elsewhere in the body. We, therefore, sought to determine its therapeutic potential in preclinical models of meningioma. METHODS: Patient-derived meningioma cells were tested in vitro and in vivo with combinations of palbociclib and radiation. Outputs included cell viability, apoptosis, clonogenicity, engrafted mouse survival, and analysis of engrafted tumor tissues after therapy. RESULTS: We found that palbociclib was highly potent against p16-deficient, Rb-intact CH157 and IOMM-Lee meningioma cells in vitro, but was ineffective against p16-intact, Rb-deficient SF8295 meningioma cells. Palbociclib also enhanced the in vitro efficacy of radiotherapy when used against p16-deficient meningioma, as indicated by cell viability and clonogenic assays. In vivo, the combination of palbociclib and radiation extended the survival of mice bearing orthotopic p16 deficient meningioma xenografts, relative to each as a monotherapy. CONCLUSIONS: These data suggest that palbociclib could be repurposed to treat patients with p16-deficient, Rb-intact meningiomas, and that a clinical trial in combination with radiation therapy merits consideration.

4.
Pediatr Blood Cancer ; 68 Suppl 2: e28439, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32827353

RESUMEN

Despite radiation therapy (RT) being an integral part of the treatment of most pediatric cancers and the recent discovery of novel molecular-targeted agents (MTAs) in this era of precision medicine with the potential to improve the therapeutic ratio of modern chemoradiotherapy regimens, there are only a few preclinical trials being conducted to discover novel radiosensitizers and radioprotectors. This has resulted in a paucity of translational clinical trials combining RT and novel MTAs. This report describes the opportunities and challenges of investigating RT together with MTAs in preclinical testing for immunotherapy, brain tumors, and sarcomas in pediatric oncology. We discuss the need for improving the collaboration between radiation oncologists, biologists, and physicists to improve the reliability, reproducibility, and translational potential of RT-based preclinical research. Current translational clinical trials using RT and MTAs for immunotherapy, brain tumors, and sarcomas are described. The technologic advances in experimental RT, availability of novel experimental tumor models, advances in immunology and tumor biology, and the discovery of novel MTAs together hold considerable promise for good quality preclinical and clinical multimodality research to improve the current rates of survival and toxicity in children afflicted with cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/terapia , Quimioradioterapia/métodos , Inmunoterapia/métodos , Terapia Molecular Dirigida , Sarcoma/terapia , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Niño , Humanos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Sarcoma/inmunología , Sarcoma/patología
5.
Neoplasia ; 21(6): 505-515, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31005631

RESUMEN

BACKGROUND: Ependymomas (EPNs) are the third most common brain tumor in children. These tumors are resistant to available chemotherapeutic treatments, therefore new effective targeted therapeutics must be identified. Increasing evidence shows epigenetic alterations including histone posttranslational modifications (PTMs), are associated with malignancy, chemotherapeutic resistance and prognosis for pediatric EPNs. In this study we examined histone PTMs in EPNs and identified potential targets to improve chemotherapeutic efficacy. METHODS: Global histone H3 lysine 4 trimethylation (H3K4me3) levels were detected in pediatric EPN tumor samples with immunohistochemistry and immunoblots. Candidate genes conferring therapeutic resistance were profiled in pediatric EPN tumor samples with micro-array. Promoter H3K4me3 was examined for two candidate genes, CCND1 and ERBB2, with chromatin-immunoprecipitation coupled with real-time PCR (ChIP-PCR). These methods and MTS assay were used to verify a relationship between H3K4me3 levels and CCND1 and ERBB2, and to investigate cell viability in response to chemotherapeutic drugs in primary cultured pediatric EPN cells. RESULTS: H3K4me3 levels positively correlate with WHO grade malignancy in pediatric EPNs and are associated with progression free survival in patients with posterior fossa group A EPNs (PF-EPN-A). Reduction of H3K4me3 by silencing its methyltransferase SETD1A, in primary cultured EPN cells increased cell response to chemotherapy. CONCLUSIONS: Our results support the development of a novel treatment that targets H3K4me3 to increase chemotherapeutic efficacy in pediatric PF-EPN-A tumors.


Asunto(s)
Ciclina D1/genética , Ependimoma/tratamiento farmacológico , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Receptor ErbB-2/genética , Carboplatino/farmacología , Supervivencia Celular/efectos de los fármacos , Preescolar , Resistencia a Antineoplásicos/efectos de los fármacos , Ependimoma/genética , Ependimoma/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Pediatría , Cultivo Primario de Células , Regiones Promotoras Genéticas/efectos de los fármacos , Vincristina/farmacología
6.
Clin Cancer Res ; 23(21): 6650-6660, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28751450

RESUMEN

Purpose: Indoleamine 2,3 dioxygenase 1 (IDO1) mediates potent immunosuppression in multiple preclinical models of cancer. However, the basis for elevated IDO1 expression in human cancer, including the most common primary malignant brain tumor in adults, glioblastoma (GBM), is poorly understood. The major objective of this study is to address this gap in our understanding of how IDO1 expression contributes to the biology of GBM, and whether its level of expression is a determinant of GBM patient outcome.Experimental Design: Patient-resected GBM, The Cancer Genome Atlas, human T-cell:GBM cocultures, as well as nu/nu, NOD-scid, and humanized (NSG-SGM3-BLT) mice-engrafted human GBM form the basis of our investigation.Results:In situ hybridization for IDO1 revealed transcript expression throughout patient-resected GBM, whereas immunohistochemical IDO1 positivity was highly variable. Multivariate statistical analysis revealed that higher levels of IDO1 transcript predict a poor patient prognosis (P = 0.0076). GBM IDO1 mRNA levels positively correlated with increased gene expression for markers of cytolytic and regulatory T cells, in addition to decreased patient survival. Humanized mice intracranially engrafted human GBM revealed an IFNγ-associated T-cell-mediated increase of intratumoral IDO1Conclusions: Our data demonstrate that high intratumoral IDO1 mRNA levels correlate with a poor GBM patient prognosis. It also confirms the positive correlation between increased GBM IDO1 levels and human-infiltrating T cells. Collectively, this study suggests that future efforts aimed at increasing T-cell-mediated effects against GBM should consider combinatorial approaches that coinhibit potential T-cell-mediated IDO1 enhancement during therapy. Clin Cancer Res; 23(21); 6650-60. ©2017 AACR.


Asunto(s)
Glioblastoma/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Linfocitos Infiltrantes de Tumor/metabolismo , Pronóstico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Linfocitos Infiltrantes de Tumor/patología , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Transl Med ; 12: 345, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25464980

RESUMEN

BACKGROUND: GL261 cells are murine glioma cells that demonstrate proliferation, invasion, and angiogenesis when implanted in syngeneic C57BL/6 mice, providing a highly useful immunocompetent animal model of glioblastoma. Modification of tumor cells for luciferase expression enables non-invasive monitoring of orthotopic tumor growth, and has proven useful for studying glioblastoma response to novel therapeutics. However, tumor modification for luciferase has the potential for evoking host immune response against otherwise syngeneic tumor cells, thereby mitigating the tumor cells' value for tumor immunology and immunotherapy studies. METHODS: GL261 cells were infected with lentivirus containing a gene encoding firefly luciferase (GL261.luc). In vitro proliferation of parental (unmodified) GL261 and GL261.luc was measured on days 0, 1, 2, 4, and 7 following plating, and the expression of 82 mouse cytokines and chemokines were analyzed by RT-PCR array. Cell lines were also evaluated for differences in invasion and migration in modified Boyden chambers. GL261 and GL261.luc cells were then implanted intracranially in C57BL/6 mice, with GL261.luc tumor growth monitored by quantitative bioluminescence imaging, and all mice were followed for survival to compare relative malignancy of tumor cells. RESULTS: No difference in proliferation was indicated for GL261 vs. GL261.luc cells (p>0.05). Of the 82 genes examined by RT-PCR array, seven (9%) exhibited statistically significant change after luciferase modification. Of these, only three changed by greater than 2-fold: BMP-2, IL-13, and TGF-ß2. No difference in invasion (p=0.67) or migration (p=0.26) was evident between modified vs. unmodified cells. GL261.luc cell luminescence was detectable in the brains of C57BL/6 mice at day 5 post-implantation, and tumor bioluminescence increased exponentially to day 19. Median overall survival was 20.2 days versus 19.7 days for mice receiving implantation with GL261 and GL261.luc, respectively (p=0.62). Histopathologic analysis revealed no morphological difference between tumors, and immunohistochemical analysis showed no significant difference for staining of CD3, Ki67, or CD31 (p>0.05 for all). CONCLUSIONS: Luciferase expression in GL261 murine glioma cells does not affect GL261 proliferation, invasion, cytokine expression, or in vivo growth. Luciferase modification increases their utility for studying tumor immunology and immunotherapeutic approaches for treating glioblastoma.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Luciferasas/genética , Animales , Neoplasias Encefálicas/inmunología , División Celular , Línea Celular Tumoral , Citocinas/inmunología , Glioma/inmunología , Ratones
8.
J Transl Med ; 12: 107, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24779345

RESUMEN

Glioblastoma remains a lethal diagnosis with a 5-year survival rate of less than 10%. (NEJM 352:987-96, 2005) Although immunotherapy-based approaches are capable of inducing detectable immune responses against tumor-specific antigens, improvements in clinical outcomes are modest, in no small part due to tumor-induced immunosuppressive mechanisms that promote immune escape and immuno-resistance. Immunotherapeutic strategies aimed at bolstering the immune response while neutralizing immunosuppression will play a critical role in improving treatment outcomes for glioblastoma patients. In vivo murine models of glioma provide an invaluable resource to achieving that end, and their use is an essential part of the preclinical workup for novel therapeutics that need to be tested in animal models prior to testing experimental therapies in patients. In this article, we review five contemporary immunocompetent mouse models, GL261 (C57BL/6), GL26 (C57BL/6) CT-2A (C57BL/6), SMA-560 (VM/Dk), and 4C8 (B6D2F1), each of which offer a suitable platform for testing novel immunotherapeutic approaches.


Asunto(s)
Neoplasias Encefálicas/terapia , Modelos Animales de Enfermedad , Glioblastoma/terapia , Inmunocompetencia , Inmunoterapia , Animales , Ratones
9.
BMC Pharmacol Toxicol ; 14: 26, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23668634

RESUMEN

BACKGROUND: Remogliflozin etabonate (RE) is the prodrug of remogliflozin, a selective inhibitor of the renal sodium-dependent glucose transporter 2 (SGLT2), which could increase urine glucose excretion (UGE) and lower plasma glucose in humans. METHODS: This double-blind, randomized, placebo-controlled, single-dose, dose-escalation, crossover study is the first human trial designed to evaluate safety, tolerability, pharmacokinetics (PK) and pharmacodynamics of RE. All subjects received single oral doses of either RE or placebo separated by approximately 2 week intervals. In Part A, 10 healthy subjects participated in 5 dosing periods where they received RE (20 mg, 50 mg, 150 mg, 500 mg, or 1000 mg) or placebo (4:1 active to placebo ratio per treatment period). In Part B, 6 subjects with type 2 diabetes mellitus (T2DM) participated in 3 dose periods where they received RE (50 mg and 500 mg) or placebo (2:1 active to placebo per treatment period). The study protocol was registered with the NIH clinical trials data base with identifier NCT01571661. RESULTS: RE was generally well-tolerated; there were no serious adverse events. In both populations, RE was rapidly absorbed and converted to remogliflozin (time to maximum plasma concentration [Cmax;Tmax] approximately 1 h). Generally, exposure to remogliflozin was proportional to the administered dose. RE was rapidly eliminated (mean T½ of ~25 min; mean plasma T½ for remogliflozin was 120 min) and was independent of dose. All subjects showed dose-dependent increases in 24-hour UGE, which plateaued at approximately 200 to 250 mmol glucose with RE doses ≥150 mg. In T2DM subjects, increased plasma glucose following OGTT was attenuated by RE in a drug-dependent fashion, but there were no clear trends in plasma insulin. There were no apparent effects of treatment on plasma or urine electrolytes. CONCLUSIONS: The results support progression of RE as a potential treatment for T2DM. TRIAL REGISTRATION: ClinicalTrials.gov NCT01571661.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucósidos/uso terapéutico , Pirazoles/uso terapéutico , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Adulto , Área Bajo la Curva , Glucemia/metabolismo , Estudios Cruzados , Diabetes Mellitus Tipo 2/metabolismo , Diarrea/inducido químicamente , Mareo/inducido químicamente , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Electrólitos/orina , Femenino , Glucósidos/efectos adversos , Glucósidos/farmacocinética , Cefalea/inducido químicamente , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/uso terapéutico , Insulina/sangre , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Estructura Molecular , Pirazoles/efectos adversos , Pirazoles/farmacocinética , Transportador 2 de Sodio-Glucosa/metabolismo , Resultado del Tratamiento
10.
BMC Pharmacol Toxicol ; 14: 25, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23631443

RESUMEN

BACKGROUND: The sodium-dependent glucose co-transporter-2 (SGLT2) is expressed in absorptive epithelia of the renal tubules. Remogliflozin etabonate (RE) is the prodrug of remogliflozin, the active entity that inhibits SGLT2. An inhibitor of this pathway would enhance urinary glucose excretion (UGE), and potentially improve plasma glucose concentrations in diabetic patients. RE is intended for use for the treatment of type 2 diabetes mellitus (T2DM) as monotherapy and in combination with existing therapies. Metformin, a dimethylbiguanide, is an effective oral antihyperglycemic agent widely used for the treatment of T2DM. METHODS: This was a randomized, open-label, repeat-dose, two-sequence, cross-over study in 13 subjects with T2DM. Subjects were randomized to one of two treatment sequences in which they received either metformin alone, RE alone, or both over three, 3-day treatment periods separated by two non-treatment intervals of variable duration. On the evening before each treatment period, subjects were admitted and confined to the clinical site for the duration of the 3-day treatment period. Pharmacokinetic, pharmacodynamic (urine glucose and fasting plasma glucose), and safety (adverse events, vital signs, ECG, clinical laboratory parameters including lactic acid) assessments were performed at check-in and throughout the treatment periods. Pharmacokinetic sampling occurred on Day 3 of each treatment period. RESULTS: This study demonstrated the lack of effect of RE on steady state metformin pharmacokinetics. Metformin did not affect the AUC of RE, remogliflozin, or its active metabolite, GSK279782, although Cmax values were slightly lower for remogliflozin and its metabolite after co-administration with metformin compared with administration of RE alone. Metformin did not alter the pharmacodynamic effects (UGE) of RE. Concomitant administration of metformin and RE was well tolerated with minimal hypoglycemia, no serious adverse events, and no increase in lactic acid. CONCLUSIONS: Coadministration of metformin and RE was well tolerated in this study. The results support continued development of RE as a treatment for T2DM. TRIAL REGISTRATION: ClinicalTrials.gov, NCT00376038.


Asunto(s)
Glucósidos/administración & dosificación , Hipoglucemiantes/administración & dosificación , Metformina/administración & dosificación , Profármacos/administración & dosificación , Pirazoles/administración & dosificación , Adulto , Glucemia/análisis , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/orina , Femenino , Glucósidos/sangre , Glucósidos/farmacocinética , Glucosuria , Humanos , Hipoglucemiantes/sangre , Hipoglucemiantes/farmacocinética , Masculino , Metformina/sangre , Metformina/farmacocinética , Persona de Mediana Edad , Profármacos/farmacocinética , Pirazoles/sangre , Pirazoles/farmacocinética , Transportador 2 de Sodio-Glucosa , Inhibidores del Cotransportador de Sodio-Glucosa 2
11.
Nanomedicine (Lond) ; 8(12): 1913-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23631502

RESUMEN

AIM: The aim of this work is to evaluate combining targeting strategy and convection-enhanced delivery in brain tumor models by imaging quantum dot-immunoliposome hybrid nanoparticles. MATERIALS & METHODS: An EGF receptor-targeted, quantum dot-immunoliposome hybrid nanoparticle (QD-IL) was synthesized. In vitro uptake was measured by flow cytometry and intracellular localization was imaged by confocal microscopy. In the in vivo study, QD-ILs were delivered to intracranial xenografts via convection-enhanced delivery and fluorescence was monitored noninvasively in real-time. RESULTS: QD-ILs exhibited specific and efficient uptake in vitro and exhibited approximately 1.3- to 5.0-fold higher total fluorescence compared with nontargeted counterpart in intracranial brain tumor xenografts in vivo. CONCLUSION: QD-ILs serve as an effective imaging agent in vitro and in vivo, and the data suggest that ligand-directed liposomal nanoparticles in conjunction with convection-enhanced delivery may offer therapeutic benefits for glioblastoma treatment as a result of specific and efficient uptake by malignant cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/patología , Sistemas de Liberación de Medicamentos , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Liposomas/metabolismo , Puntos Cuánticos/metabolismo , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Convección , Femenino , Glioblastoma/patología , Humanos , Liposomas/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Puntos Cuánticos/análisis
12.
J Neurooncol ; 110(3): 305-13, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22983601

RESUMEN

Diffuse intrinsic pontine gliomas arise almost exclusively in children, and despite advances in treatment, the majority of patients die within 2 years after initial diagnosis. Because of their infiltrative nature and anatomic location in an eloquent area of the brain, most pontine gliomas are treated without a surgical biopsy. The corresponding lack of tissue samples has resulted in a limited understanding of the underlying genetic and molecular biologic abnormalities associated with pontine gliomas, and is a substantial obstacle for the preclinical testing of targeted therapeutic agents for these tumors. We have established a human glioma cell line that originated from surgical biopsy performed on a patient with a pontine glioma. To insure sustainable in vitro propagation, tumor cells were modified with hTERT (human telomerase ribonucleoprotein reverse transcriptase), and with a luciferase reporter to enable non-invasive bioluminescence imaging. The hTERT modified cells are tumorigenic in athymic rodents, and produce brainstem tumors that recapitulate the infiltrative growth of brainstem gliomas in patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias del Tronco Encefálico/patología , Glioma/patología , Puente , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/terapia , Niño , Femenino , Perfilación de la Expresión Génica , Glioma/genética , Glioma/terapia , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Ratas , Ratas Desnudas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
J Neurooncol ; 96(2): 151-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19585223

RESUMEN

Despite the use of radiation and chemotherapy, the prognosis for children with diffuse brainstem gliomas is extremely poor. There is a need for relevant brainstem tumor models that can be used to test new therapeutic agents and delivery systems in pre-clinical studies. We report the development of a brainstem-tumor model in rats and the application of bioluminescence imaging (BLI) for monitoring tumor growth and response to therapy as part of this model. Luciferase-modified human glioblastoma cells from five different tumor cell sources (either cell lines or serially-passaged xenografts) were implanted into the pontine tegmentum of athymic rats using an implantable guide-screw system. Tumor growth was monitored by BLI and tumor volume was calculated by three-dimensional measurements from serial histopathologic sections. To evaluate if this model would allow detection of therapeutic response, rats bearing brainstem U-87 MG or GS2 glioblastoma xenografts were treated with the DNA methylating agent temozolomide (TMZ). For each of the tumor cell sources tested, BLI monitoring revealed progressive tumor growth in all animals, and symptoms caused by tumor burden were evident 26-29 days after implantation of U-87 MG, U-251 MG, GBM6, and GBM14 cells, and 37-47 days after implantation of GS2 cells. Histopathologic analysis revealed tumor growth within the pons in all rats and BLI correlated quantitatively with tumor volume. Variable infiltration was evident among the different tumors, with GS2 tumor cells exhibiting the greatest degree of infiltration. TMZ treatment groups were included for experiments involving U-87 MG and GS2 cells, and in each case TMZ delayed tumor growth, as indicated by BLI monitoring, and significantly extended survival of animal subjects. Our results demonstrate the development of a brainstem tumor model in athymic rats, in which tumor growth and response to therapy can be accurately monitored by BLI. This model is well suited for pre-clinical testing of therapeutics that are being considered for treatment of patients with brainstem tumors.


Asunto(s)
Neoplasias del Tronco Encefálico/diagnóstico , Glioma/diagnóstico , Luciferasas , Sustancias Luminiscentes , Animales , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Línea Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Diagnóstico por Imagen , Modelos Animales de Enfermedad , Glioma/tratamiento farmacológico , Humanos , Etiquetado Corte-Fin in Situ/métodos , Estimación de Kaplan-Meier , Masculino , Trasplante de Neoplasias/métodos , Ratas , Ratas Desnudas , Temozolomida , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
14.
Nat Biotechnol ; 23(2): 209-14, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15685166

RESUMEN

Live attenuated measles viruses of the Edmonston lineage (MV-Edm) have potent anti-tumor activity but are not entirely tumor-specific owing to widespread distribution of their native receptors, CD46 and SLAM. We have therefore developed a pseudoreceptor system that allows rescue and propagation of fully retargeted viruses displaying single-chain antibody fragments. Viruses retargeted to tumor-selective CD38, epidermal growth factor receptor (EGFR) or EGFR mutant vIII (EGFRvIII) efficiently entered cells through their respective targeted receptors in vitro and in vivo, but not through CD46 and SLAM. When administered intratumorally or intravenously to mice bearing human CD38 or EGFR-positive human tumor xenografts, the targeted viruses demonstrated specific receptor-mediated anti-tumor activity. These data provide an in vivo demonstration of antibody-directed tumor destruction by retargeted oncolytic viruses.


Asunto(s)
Terapia Biológica/métodos , Virus del Sarampión/fisiología , Neoplasias/virología , Virus/crecimiento & desarrollo , Virus/genética , Animales , Anticuerpos Antineoplásicos/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Mejoramiento Genético/métodos , Humanos , Ratones , Neoplasias/terapia , Receptores Virales/genética , Receptores Virales/metabolismo , Resultado del Tratamiento , Células Vero , Replicación Viral/genética
15.
Br J Haematol ; 121(2): 287-95, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12694251

RESUMEN

Fluorescence in situ hybridization (FISH) was used to detect 6q-, 11q-, +12, 13q-, 17p- and translocations involving 14q32 in interphase nuclei from blood and/or bone marrow from 113 patients with B-cell chronic lymphocytic leukaemia (B-CLL). A total of 87 patients (77%) had a FISH anomaly: 13q- x 1 was most frequent (64%) followed by 13q- x 2 (28%), +12 (25%), 11q- (15%), 17p- (8%) and 6q- (0%). FISH results for blood and bone marrow cells in 38 patients were similar. Purified CD5+/CD19+ cells from blood were studied in eight patients and results indicate that in some patients not all B cells have FISH anomalies. We used a defined set of hierarchical FISH risk categories to compare FISH results by stable versus progressive disease, age, sex, Rai stage, CD38+ expression and IgVH mutational status. Significant differences in FISH risk distributions were associated with Rai stage, disease status and CD38+, but not by age, sex or IgVH mutational status. To look for baseline factors associated with high-risk disease, multivariate analysis of age, sex, Rai stage, CD38+ and disease status versus FISH risk category was performed. Importantly, only CD38+ was significantly associated with high-risk FISH categories (+12, 11q- and 17p-) after adjustment for the effects of other variables.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 17 , Hibridación Fluorescente in Situ , Leucemia Linfocítica Crónica de Células B/genética , ADP-Ribosil Ciclasa/análisis , ADP-Ribosil Ciclasa 1 , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/análisis , Antígenos CD19/análisis , Linfocitos B/inmunología , Linfocitos B/fisiología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Antígenos CD5/análisis , Distribución de Chi-Cuadrado , Progresión de la Enfermedad , Femenino , Humanos , Región Variable de Inmunoglobulina/genética , Inmunofenotipificación , Interfase , Leucemia Linfocítica Crónica de Células B/inmunología , Masculino , Glicoproteínas de Membrana , Persona de Mediana Edad , Análisis Multivariante , Mutación , Pronóstico , Medición de Riesgo , Estadísticas no Paramétricas
16.
Neurosurgery ; 51(5): 1246-54; discussion 1254-5, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12383370

RESUMEN

OBJECTIVE: Immunostaining for p53 commonly is considered a marker of neoplasia. Previous studies of nonneoplastic processes have yielded conflicting results. METHODS: To test the assumption that p53 immunoreactivity indicates neoplasia, we examined 60 formalin-fixed, paraffin-embedded biopsies of nonneoplastic central nervous system lesions, including gliosis (n = 12), infarction (n = 9), demyelinating disease (n = 23), progressive multifocal leukoencephalopathy (n = 11), and herpes simplex virus encephalitis (n = 5). Diffuse astrocytomas (n = 50) of World Health Organization Grades 2 to 4 also were studied, as were six control autopsy brains. The avidin-biotin-peroxidase complex method was used with commercially available monoclonal antisera to both p53 (clone DO7; Dako, Carpinteria, CA) and mdm2 (Dako), a protein known to stabilize p53. Two samples of each nonneoplastic lesion also were subjected to deoxyribonucleic acid isolation, amplification, and sequencing of exons 5 to 8 of TP53. RESULTS: Although it was low level in most instances, p53 immunoreactivity was noted in all but normal control samples. In reactive lesions, staining was largely observed in astrocytes and histiocytes. Scant oligodendroglia also were labeled in demyelinating disease. The progressive multifocal leukoencephalopathy samples revealed exceptionally strong staining in astrocytes and infected oligodendrocytes. Staining also was noted in occasional endothelial cells and neurons, and in rare lymphocytes. Immunoreactivity for mdm2, studied only in nonneoplastic lesions, was moderate to strong in all cases and limited to reactive astrocytes and histiocytes. No TP53 mutations were noted in the nonneoplastic lesions studied. To some extent, all astrocytomas exhibited p53 immunopositivity, particularly high-grade lesions. CONCLUSION: p53 immunoreactivity is not limited to astrocytomas, but it can be observed in lesions that often are mistaken for glioma. No TP53 mutations accompany p53 expression in nonneoplastic lesions, and mdm2 may be responsible for persistence of p53 expression in these processes.


Asunto(s)
Encefalopatías/metabolismo , Proteínas Nucleares , Proteína p53 Supresora de Tumor/metabolismo , Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Infarto Cerebral/metabolismo , Análisis Mutacional de ADN , Enfermedades Desmielinizantes/metabolismo , Encefalitis por Herpes Simple/metabolismo , Gliosis/metabolismo , Humanos , Inmunohistoquímica , Leucoencefalopatía Multifocal Progresiva/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Valores de Referencia , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA