Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(32): 9978-83, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26216958

RESUMEN

We present a proof of concept study designed to support the clinical development of mass spectrometry imaging (MSI) for the detection of pituitary tumors during surgery. We analyzed by matrix-assisted laser desorption/ionization (MALDI) MSI six nonpathological (NP) human pituitary glands and 45 hormone secreting and nonsecreting (NS) human pituitary adenomas. We show that the distribution of pituitary hormones such as prolactin (PRL), growth hormone (GH), adrenocorticotropic hormone (ACTH), and thyroid stimulating hormone (TSH) in both normal and tumor tissues can be assessed by using this approach. The presence of most of the pituitary hormones was confirmed by using MS/MS and pseudo-MS/MS methods, and subtyping of pituitary adenomas was performed by using principal component analysis (PCA) and support vector machine (SVM). Our proof of concept study demonstrates that MALDI MSI could be used to directly detect excessive hormonal production from functional pituitary adenomas and generally classify pituitary adenomas by using statistical and machine learning analyses. The tissue characterization can be completed in fewer than 30 min and could therefore be applied for the near-real-time detection and delineation of pituitary tumors for intraoperative surgical decision-making.


Asunto(s)
Sistemas de Computación , Imagenología Tridimensional , Neoplasias Hipofisarias/diagnóstico , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Humanos , Proteínas de Neoplasias/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/patología , Análisis de Componente Principal , Reproducibilidad de los Resultados
3.
Oncotarget ; 6(11): 8788-806, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25871395

RESUMEN

Malignant gliomas remain aggressive and lethal primary brain tumors in adults. The epidermal growth factor receptor (EGFR) is frequently overexpressed in the most common malignant glioma, glioblastoma (GBM), and represents an important therapeutic target. GBM stem-like cells (GSCs) present in tumors are felt to be highly tumorigenic and responsible for tumor recurrence. Multifunctional magnetic iron-oxide nanoparticles (IONPs) can be directly imaged by magnetic resonance imaging (MRI) and designed to therapeutically target cancer cells. The targeting effects of IONPs conjugated to the EGFR inhibitor, cetuximab (cetuximab-IONPs), were determined with EGFR- and EGFRvIII-expressing human GBM neurospheres and GSCs. Transmission electron microscopy revealed cetuximab-IONP GBM cell binding and internalization. Fluorescence microscopy and Prussian blue staining showed increased uptake of cetuximab-IONPs by EGFR- as well as EGFRvIII-expressing GSCs and neurospheres in comparison to cetuximab or free IONPs. Treatment with cetuximab-IONPs resulted in a significant antitumor effect that was greater than with cetuximab alone due to more efficient, CD133-independent cellular targeting and uptake, EGFR signaling alterations, EGFR internalization, and apoptosis induction in EGFR-expressing GSCs and neurospheres. A significant increase in survival was found after cetuximab-IONP convection-enhanced delivery treatment of 3 intracranial rodent GBM models employing human EGFR-expressing GBM xenografts.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Cetuximab/farmacología , Glioblastoma/tratamiento farmacológico , Nanopartículas de Magnetita/administración & dosificación , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Animales , Células Cultivadas , Cetuximab/administración & dosificación , Cetuximab/toxicidad , Portadores de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Glioblastoma/patología , Humanos , Nanopartículas de Magnetita/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas de Neoplasias/análisis , Neuronas/efectos de los fármacos , Polietilenglicoles/administración & dosificación , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 6(7): 4704-16, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25609200

RESUMEN

There are no effective medical treatments for WHO grade III (anaplastic) meningioma. Patients with this high-grade malignancy have a median survival of less than two years. Therapeutics that modulate the mechanisms that inhibit local immune responses in the tumor microenvironment are showing significant and durable clinical responses in patients with treatment refractory high-grade tumors. We examined the immune infiltrate of 291 meningiomas including WHO grade I-III meningiomas using immunohistochemistry and we examined the expression of PD-L1 mRNA by RNAscope in situ hybridization and PD-L1 protein by immunohistochemistry. In meningioma, the tumor infiltrating lymphocytes are predominantly T cells. In anaplastic meningioma, there is a sharp decrease in the number of T cells, including the numbers of CD4+ and CD8+ T cells and cells expressing PD-1 and there is also an increase in the number of FOXP3 expressing immunoregulatory (Treg) cells. PD-L1 expression is increased in anaplastic meningioma - both mRNA and protein. Using patient derived meningioma cell, we confirm that PD-L1 is expressed in meningioma cells themselves, and not solely in infiltrating immune cells. This work indicates that high-grade meningioma harbor an immunosuppressive tumor microenviroment and that increased Treg cells and elevated PD-L1 may contribute to the aggressive phenotype of these tumors.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Western Blotting , Linfocitos T CD8-positivos/inmunología , Humanos , Técnicas para Inmunoenzimas , Neoplasias Meníngeas/inmunología , Neoplasias Meníngeas/mortalidad , Neoplasias Meníngeas/patología , Meningioma/mortalidad , Meningioma/patología , Clasificación del Tumor , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia
5.
Cancer Res ; 70(15): 6303-12, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20647323

RESUMEN

The magnetic nanoparticle has emerged as a potential multifunctional clinical tool that can provide cancer cell detection by magnetic resonance imaging (MRI) contrast enhancement as well as targeted cancer cell therapy. A major barrier in the use of nanotechnology for brain tumor applications is the difficulty in delivering nanoparticles to intracranial tumors. Iron oxide nanoparticles (IONP; 10 nm in core size) conjugated to a purified antibody that selectively binds to the epidermal growth factor receptor (EGFR) deletion mutant (EGFRvIII) present on human glioblastoma multiforme (GBM) cells were used for therapeutic targeting and MRI contrast enhancement of experimental glioblastoma, both in vitro and in vivo, after convection-enhanced delivery (CED). A significant decrease in glioblastoma cell survival was observed after nanoparticle treatment and no toxicity was observed with treatment of human astrocytes (P < 0.001). Lower EGFR phosphorylation was found in glioblastoma cells after EGFRvIIIAb-IONP treatment. Apoptosis was determined to be the mode of cell death after treatment of GBM cells and glioblastoma stem cell-containing neurospheres with EGFRvIIIAb-IONPs. MRI-guided CED of EGFRvIIIAb-IONPs allowed for the initial distribution of magnetic nanoparticles within or adjacent to intracranial human xenograft tumors and continued dispersion days later. A significant increase in animal survival was found after CED of magnetic nanoparticles (P < 0.01) in mice implanted with highly tumorigenic glioblastoma xenografts (U87DeltaEGFRvIII). IONPs conjugated to an antibody specific to the EGFRvIII deletion mutant constitutively expressed by human glioblastoma tumors can provide selective MRI contrast enhancement of tumor cells and targeted therapy of infiltrative glioblastoma cells after CED.


Asunto(s)
Neoplasias Encefálicas/terapia , Receptores ErbB/inmunología , Compuestos Férricos/administración & dosificación , Glioblastoma/terapia , Inmunoconjugados/administración & dosificación , Nanopartículas/administración & dosificación , Animales , Especificidad de Anticuerpos , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Compuestos Férricos/química , Glioblastoma/diagnóstico , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Nanopartículas/química , Transducción de Señal , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA