Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
BMC Neurol ; 21(1): 231, 2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162346

RESUMEN

BACKGROUND: Gliosarcoma (GS) refers to the presence of mesenchymal differentiation (as seen using light microscopy) in the setting of glioblastoma (GB, an astrocytoma, WHO Grade 4). Although the same approach to treatment is typically adopted for GS and GB, there remains some debate as to whether GS should be considered a discrete pathological entity. Differences between these tumors have not been clearly established at the molecular level. METHODS: Patients with GS (n=48) or GB (n=1229) underwent molecular profiling (MP) with a pan-cancer panel of tests as part of their clinical care. The methods employed included next-generation sequencing (NGS) of DNA and RNA, copy number variation (CNV) of DNA and immunohistochemistry (IHC). The MP comprised 1153 tests in total, although results for each test were not available for every tumor profiled. We analyzed this data retrospectively in order to determine if our results were in keeping with what is known about the pathogenesis of GS by contrast with GB. We also sought novel associations between the MP and GS vs. GB which might improve our understanding of pathogenesis of GS. RESULTS: Potentially meaningful associations (p<0.1, Fisher's exact test (FET)) were found for 14 of these tests in GS vs. GB. A novel finding was higher levels of proteins mediating immuno-evasion (PD-1, PD-L1) in GS. All of the differences we observed have been associated with epithelial-to-mesenchymal transition (EMT) in other tumor types. Many of the changes we saw in GS are novel in the setting of glial tumors, including copy number amplification in LYL1 and mutations in PTPN11. CONCLUSIONS: GS shows certain characteristics of EMT, by contrast with GB. Treatments targeting immuno-evasion may be of greater therapeutic value in GS relative to GB.


Asunto(s)
Glioblastoma/patología , Gliosarcoma/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Transición Epitelial-Mesenquimal , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Gliosarcoma/genética , Gliosarcoma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Estudios Retrospectivos , Adulto Joven
2.
Cell Biol Int ; 43(11): 1257-1266, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30839135

RESUMEN

Glioblastoma is the most common malignant primary brain tumor with poor prognosis. Invasion involves pro-inflammatory cytokines and major signaling hubs. Tumor necrosis factor-α (TNF-α) acts as a master switch in establishing an intricate link between inflammation and cancer. The present study attempted to explore the possible implication of MAPK extracellular signaling-regulated kinase kinase (MEK)-extracellular signaling-regulated kinase (ERK) signaling pathway and expression of nuclear factor-κB (NF-κB), signal transducers and activators of transcription-6 (STAT-6), ERK, and phosphorylated-ERK (p-ERK) signaling proteins in TNF-α microenvironment. U0126 and PD98059 were used to inhibit the MEK-ERK1/2 pathway. TNF-α stimulation enhanced invasion in U87MG, U251MG and patient-derived primary glioma cells, whereas cell viability was not altered. Matrix metalloproteinase-2 (MMP-2) activity was increased only in U251MG glioma cells. These data suggest that TNF-α microenvironment plays an important role in the invasion of U251MG, U87MG, and patient-derived primary glioma cells, without any cytotoxic effect. The MMP-2 activity is differentially regulated by TNF-α stimulation in these cells. TNF-α stimulation upregulated the protein expression of ERK-1, ERK-2 and also increased the level of p-ERK1/2. TNF-α stimulation further upregulated the expression of NF-κB1, STAT-6 in tandem with Ras-MEK signaling system in U87MG cells, which emphasized the possible involvement of these signaling hubs in the glioma microenvironment. MEK-ERK inhibitors significantly attenuated the invasion of U87MG cells mediated by the TNF-α stimulation, probably through their inhibitory impact on p-ERK1/2 and ERK-2. This study provides the possible rationale of invasion by glioma cells in a TNF-α-induced pro-inflammatory milieu, which involves direct role of MEK-ERK signaling, with possible implication of NF-κB and STAT-6.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Gliosarcoma/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Línea Celular Tumoral , Movimiento Celular , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Microambiente Tumoral
3.
J Neurooncol ; 137(2): 303-311, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29264835

RESUMEN

Gliosarcoma is a rare histopathologic variant of glioblastoma traditionally associated with a poor prognosis. While gliosarcoma may represent a distinct clinical entity given its unique histologic composition and molecular features, its relative prognostic significance remains uncertain. While treatment of gliosarcoma generally encompasses the same standardized approach used in glioblastoma, supporting evidence is limited given its rarity. Here, we characterized 32 cases of gliosarcoma and retrospectively evaluated survival relative to 451 glioblastoma patients diagnosed during the same era within the same institution. Overall, we identified 22 primary gliosarcomas, representing 4.7% of WHO Grade IV primary glioblastomas, and 10 secondary gliosarcomas. With median age of 62, patients were predominately Caucasian (87.5%) and male (65.6%). Tumors with available molecular profiling were primarily MGMT-unmethylated (87.5%), IDH-1-preserved (100%) and EGFR wild-type (100%). Interestingly, while no significant median survival difference between primary gliosarcoma and glioblastoma was observed across the entire cohort (11.0 vs. 14.8 months, p = 0.269), median survival was worse for gliosarcoma specifically among patients who received modern temozolomide-based (TMZ) chemoradiotherapy (11.0 vs. 17.3 months, p = 0.006). Matched-pair analysis also trended toward worse median survival among gliosarcomas (11.0 vs. 19.6 months, log-rank p = 0.177, Breslow p = 0.010). While adjuvant radiotherapy (HR 0.206, p = 0.035) and TMZ-based chemotherapy (HR 0.531, p = 0.000) appeared protective, gliosarcoma emerged as a significantly poor prognostic factor on multivariate analysis (HR 3.27, p = 0.012). Collectively, our results suggest that gliosarcoma may still portend worse prognosis even with modern trimodality therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Gliosarcoma/metabolismo , Gliosarcoma/patología , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Femenino , Glioblastoma/genética , Glioblastoma/terapia , Gliosarcoma/genética , Gliosarcoma/terapia , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Análisis de Supervivencia
4.
Neuropathology ; 36(5): 448-455, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26932501

RESUMEN

Gliosarcoma (GS) is a rare subtype of glioblastoma (GBM) characterized by both glial and mesenchymal components. Unlike GBM, there are no specific prognostic markers, and optimized treatments for patients with GS do not exist. Recent reports describe BRAFV600E mutation in malignant peripheral nerve sheath tumors, and aberrant Wnt signaling and CTNNB1 (ß-catenin gene) mutations have been described in GBM. We sought to determine whether GS tumors harbor BRAFV600E mutations or aberrant Wnt signaling, as indicated by nuclear localization of ß-catenin, by immunohistochemical detection. Forty-eight (48) cases of primary and secondary adult GS (including recurrent ones) were evaluated by immunohistochemical techniques for the presence of nuclear ß-catenin and the BRAFV600E mutation. A small subset (6/46, 13%) showed nuclear localization of ß-catenin. None of the cases harbored BRAFV600E mutations (0/48). These results are the first to describe the presence of Wnt signaling pathway abnormalities, manifested by nuclear ß-catenin, in a subset, as well as the lack of BRAFV600E mutation in GS. We propose a potential role for Wnt pathway alterations in the pathogenesis of a subset of GS.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Núcleo Celular/metabolismo , Gliosarcoma/genética , Gliosarcoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , beta Catenina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Femenino , Gliosarcoma/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Vía de Señalización Wnt
5.
Int J Clin Oncol ; 20(4): 650-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25407559

RESUMEN

AIMS: To determine the efficacy of methylguanine methyltransferase (MGMT) depletion + BCNU [1,3-bis(2-chloroethyl)-1- nitrosourea: carmustine] therapy and the impact of methylation status in adults with glioblastoma multiforme (GBM) and gliosarcoma. METHODS: Methylation analysis was performed on GBM patients with adequate tissue samples. Patients with newly diagnosed GBM or gliosarcoma were eligible for this Phase III open-label clinical trial. At registration, patients were randomized to Arm 1, which consisted of therapy with O(6)-benzylguanine (O(6)-BG) + BCNU 40 mg/m(2) (reduced dose) + radiation therapy (RT) (O6BG + BCNU arm), or Arm 2, which consisted of therapy with BCNU 200 mg/m(2) + RT (BCNU arm). RESULTS: A total of 183 patients with newly diagnosed GBM or gliosarcoma from 42 U.S. institutions were enrolled in this study. Of these, 90 eligible patients received O(6)-BG + BCNU + RT and 89 received BCNU + RT. The trial was halted at the first interim analysis in accordance with the guidelines for stopping the study due to futility (<40 % improvement among patients on the O6BG + BCNU arm). Following adjustment for stratification factors, there was no significant difference in overall survival (OS) or progression-free survival (PFS) between the two groups (one sided p = 0.94 and p = 0.88, respectively). Median OS was 11 [95 % confidence interval (CI) 8-13] months for patients in the O6BG + BCNU arm and 10 (95 % CI 8-12) months for those in the BCNU arm. PFS was 4 months for patients in each arm. Adverse events were reported in both arms, with significantly more grade 4 and 5 events in the experimental arm. CONCLUSIONS: The addition of O(6)-BG to the standard regimen of radiation and BCNU for the treatment patients with newly diagnosed GBM and gliosarcoma did not provide added benefit and in fact caused additional toxicity.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Gliosarcoma/terapia , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/metabolismo , Carmustina/administración & dosificación , Terapia Combinada , Metilación de ADN , Femenino , Glioblastoma/metabolismo , Gliosarcoma/metabolismo , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Masculino , Persona de Mediana Edad , Radioterapia , Adulto Joven
6.
Mol Imaging ; 132014.
Artículo en Inglés | MEDLINE | ID: mdl-25430958

RESUMEN

Magnetic resonance imaging (MRI) allows for noninvasive, deep tissue imaging with high spatial resolution, making it an attractive modality for in vivo cellular imaging. Since reporter genes can generate magnetic resonance (MR) contrast based on molecular activity, they offer a potentially powerful tool for cellular imaging. The mms6 gene was originally identified in magnetotactic bacteria (MTB), which is known to play a key role in magnetic crystal formation. The purpose of the present work was to investigate the possibility of using mms6 as an MR reporter gene. We established a transgenic mammalian cell line that stably expresses mms6. In vitro experiments show that mms6-expressing cells form clusters of nanoparticles within and outside membrane-enclosed structures and produce changes in MR contrast, most likely by increasing iron uptake of intracellular iron. Additionally, in vivo MRI experiments demonstrate that mms6-expressing tumors can be distinguished from parental tumors not expressing mms6, even in the absence of exogenous iron supplementation. Our results demonstrate that mms6 can function as an MR reporter gene with the potential to monitor gene expression and to visualize the proliferation, migration, and metastasis of tumor cells expressing it.


Asunto(s)
Proteínas Bacterianas/análisis , Neoplasias Encefálicas/patología , Genes Reporteros , Gliosarcoma/patología , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Animales , Proteínas Bacterianas/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Gliosarcoma/genética , Gliosarcoma/metabolismo , Masculino , Nanopartículas/metabolismo , Trasplante de Neoplasias , Ratas
7.
Nanotechnology ; 25(44): 445104, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25325364

RESUMEN

Here, we present a new optical contrast agent based on silver nanoplate clusters embedded inside of a polymer nano matrix. Unlike nanosphere clusters, which have been well studied, nanoplate clusters have unique properties due to the different possible orientations of interaction between the individual plates, resulting in a significant broadening of the absorption spectra. These nanoclusters were immobilized inside of a polymer cladding so as to maintain their stability and optical properties under in vivo conditions. The polymer-coated silver nanoplate clusters show a lower toxicity compared to the uncoated nanoparticles. At high nanoparticle concentrations, cell death occurs mostly due to apoptosis. These nanoparticles were used for targeted fluorescence imaging in a rat glioma cell line by incorporating a fluorescent dye into the matrix, followed by conjugation of a tumor targeting an F3 peptide. We further used these nanoparticles as photoacoustic contrast agents in vivo to enhance the contrast of the vasculature structures in a rat ear model. We observed a contrast enhancement of over 90% following the nanoparticle injection. It is also shown that these NPs can serve as efficient contrast agents, with specific targeting abilities for broadband multimodal imaging that are usable for diagnostic applications and that extend into use as therapeutic agents as well.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Medios de Contraste/síntesis química , Diagnóstico por Imagen/métodos , Gliosarcoma/diagnóstico , Nanopartículas , Animales , Apoptosis , Neoplasias Encefálicas/metabolismo , Gliosarcoma/metabolismo , Nanopartículas/efectos adversos , Nanopartículas/ultraestructura , Estrés Oxidativo , Polímeros/síntesis química , Ratas , Plata
8.
Zhonghua Bing Li Xue Za Zhi ; 43(10): 657-62, 2014 Oct.
Artículo en Zh | MEDLINE | ID: mdl-25567590

RESUMEN

OBJECTIVE: To study the clinical and pathologic features of gliosarcoma of cerebral hemispheres. METHODS: The clinicopathologic features of 10 cases of gliosarcoma involving cerebral hemispheres were reviewed. Immunohistochemical study was carried out using EnVision method. RESULTS: The mean age of the patients was 54 years and the male-to-female ratio was 6 to 4. Clinical symptoms included headache (6/10), nausea/vomiting (5/10), and sensory or motor impairment (4/10). Nine of the cases were primary gliosarcoma, with maximum diameter ranging from 2.4 to 5.5 cm (mean = 4.2 cm). The remaining case represented secondary gliosarcoma involving skull base and extracranial tissues. Histologic examination showed a biphasic pattern in all cases. Regarding the glial component, there were 9 cases of pleomorphic glioblastoma and 1 case of giant cell glioblastoma. Reticulin stain was positive in all cases. Immunohistochemical study showed that the tumor cells variably expressed GFAP (10/10), p16 (4/10), EGFR (1/10), CD68 (1/10) and p53 (6/10). The Ki-67 index ranged from 15% to 70% (mean = 34%). Six patients had follow-up data available. One patient was disease-free for 45 months and 5 patients died of the disease at 3 to 17 months after the operation (mean duration of survival = 9 months). CONCLUSIONS: Gliosarcoma is a highly aggressive tumor, often locates in the deeper part cerebral hemispheres and has a relatively short duration of symptoms. It carries a poor prognosis. GFAP immunostain and reticulin stain are helpful in confirming the diagnosis. p53 and p16 are also expressed in some cases.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Gliosarcoma/patología , Adulto , Neoplasias Encefálicas/metabolismo , Cerebro/patología , Femenino , Glioblastoma/metabolismo , Gliosarcoma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Neuroglía/patología
9.
PLoS One ; 19(2): e0291368, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38306361

RESUMEN

Glioblastoma multiforme (GBM) and the GBM variant gliosarcoma (GS) are among the tumors with the highest morbidity and mortality, providing only palliation. Stem-like glioma cells (SLGCs) are involved in tumor initiation, progression, therapy resistance, and relapse. The identification of general features of SLGCs could contribute to the development of more efficient therapies. Commercially available protein arrays were used to determine the cell surface signature of eight SLGC lines from GBMs, one SLGC line obtained from a xenotransplanted GBM-derived SLGC line, and three SLGC lines from GSs. By means of non-negative matrix factorization expression metaprofiles were calculated. Using the cophenetic correlation coefficient (CCC) five metaprofiles (MPs) were identified, which are characterized by specific combinations of 7-12 factors. Furthermore, the expression of several factors, that are associated with GBM prognosis, GBM subtypes, SLGC differentiation stages, or neural identity was evaluated. The investigation encompassed 24 distinct SLGC lines, four of which were derived from xenotransplanted SLGCs, and included the SLGC lines characterized by the metaprofiles. It turned out that all SLGC lines expressed the epidermal growth factor EGFR and EGFR ligands, often in the presence of additional receptor tyrosine kinases. Moreover, all SLGC lines displayed a neural signature and the IDH1 wildtype, but differed in their p53 and PTEN status. Pearson Correlation analysis identified a positive association between the pluripotency factor Sox2 and the expression of FABP7, Musashi, CD133, GFAP, but not with MGMT or Hif1α. Spherical growth, however, was positively correlated with high levels of Hif1α, CDK4, PTEN, and PDGFRß, whereas correlations with stemness factors or MGMT (MGMT expression and promoter methylation) were low or missing. Factors highly expressed by all SLGC lines, irrespective of their degree of stemness and growth behavior, are Cathepsin-D, CD99, EMMPRIN/CD147, Intß1, the Galectins 3 and 3b, and N-Cadherin.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Gliosarcoma , Humanos , Glioblastoma/metabolismo , Gliosarcoma/genética , Gliosarcoma/metabolismo , Gliosarcoma/patología , Neoplasias Encefálicas/metabolismo , Recurrencia Local de Neoplasia/patología , Glioma/patología , Células Madre Neoplásicas/metabolismo , Receptores ErbB/metabolismo , Línea Celular Tumoral
10.
J Nucl Med ; 65(8): 1217-1223, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38960714

RESUMEN

Despite their unique histologic features, gliosarcomas belong to the group of glioblastomas and are treated according to the same standards. Fibroblast activation protein (FAP) is a component of a tumor-specific subpopulation of fibroblasts that plays a critical role in tumor growth and invasion. Some case studies suggest an elevated expression of FAP in glioblastoma and a particularly strong expression in gliosarcoma attributed to traits of predominant mesenchymal differentiation. However, the prognostic impact of FAP and its diagnostic and therapeutic potential remain unclear. Here, we investigate the clinical relevance of FAP expression in gliosarcoma and glioblastoma and how it correlates with 68Ga-FAP inhibitor (FAPI)-46 PET uptake. Methods: Patients diagnosed with gliosarcoma or glioblastoma without sarcomatous differentiation with an overall survival of less than 2.5 y were enrolled. Histologic examination included immunohistochemistry and semiquantitative scoring of FAP (0-3, with higher values indicating stronger expression). Additionally, 68Ga-FAPI-46 PET scans were performed in a subset of glioblastomas without sarcomatous differentiation patients. The clinical SUVs were correlated with FAP expression levels in surgically derived tumor tissue and relevant prognostic factors. Results: Of the 61 patients who were enrolled, 13 of them had gliosarcoma. Immunohistochemistry revealed significantly more FAP in gliosarcomas than in glioblastomas without sarcomatous differentiation of tumor tissue (P < 0.0001). In the latter, FAP expression was confined to the perivascular space, whereas neoplastic cells additionally expressed FAP in gliosarcoma. A significant correlation of immunohistochemical FAP with SUVmean and SUVpeak of 68Ga-FAPI-46 PET indicates that clinical tracer uptake represents FAP expression of the tumor. Although gliosarcomas express higher levels of FAP than do glioblastomas without sarcomatous differentiation, overall survival does not significantly differ between the groups. Conclusion: The analysis reveals a significant correlation between SUVmean and SUVpeak in 68Ga-FAPI-46 PET and immunohistochemical FAP expression. This study indicates that FAP expression is much more abundant in the gliosarcoma subgroup of glioblastomas. This could open not only a diagnostic but also a therapeutic gap, since FAP could be explored as a theranostic target to enhance survival in a distinct subgroup of high-risk brain tumor patients with poor survival prognosis.


Asunto(s)
Glioblastoma , Gliosarcoma , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Endopeptidasas , Gelatinasas/metabolismo , Glioblastoma/diagnóstico por imagen , Glioblastoma/metabolismo , Glioblastoma/patología , Gliosarcoma/diagnóstico por imagen , Gliosarcoma/metabolismo , Gliosarcoma/patología , Proteínas de la Membrana/metabolismo , Tomografía de Emisión de Positrones , Pronóstico , Quinolinas , Serina Endopeptidasas/metabolismo , Análisis de Supervivencia
11.
Am J Pathol ; 180(5): 1816-23, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22538188

RESUMEN

Gliosarcoma is a rare glioblastoma variant characterized by a biphasic tissue pattern with alternating areas that display either glial (glial fibrillary acidic protein-positive) or mesenchymal (reticulin-positive) differentiation. Previous analyses have shown identical genetic alterations in glial and mesenchymal tumor areas, suggesting that gliosarcomas are genetically monoclonal, and mesenchymal differentiation was considered to reflect the elevated genomic instability of glioblastomas. In the present study, we compared genome-wide chromosomal imbalances using array comparative genomic hybridization in glial and mesenchymal tumor areas of 13 gliosarcomas. The patterns of gain and loss were similar, except that the gain at 13q13.3-q14.1 (log(2) ratio >3.0), containing the STOML3, FREM2, and LHFP genes, which was restricted to the mesenchymal tumor area of a gliosarcoma. Further analyses of 64 cases of gliosarcoma using quantitative PCR showed amplification of the STOML3, FREM2, and LHFP genes in 14 (22%), 10 (16%), and 7 (11%) mesenchymal tumor areas, respectively, but not in glial tumor areas. Results of IHC analysis confirmed that overexpression of STOML3 and FREM2 was more extensive in mesenchymal than in glial tumor areas. These results suggest that the mesenchymal components in a small fraction of gliosarcomas may be derived from glial cells with additional genetic alterations.


Asunto(s)
Amplificación de Genes , Gliosarcoma/genética , Proteínas de Neoplasias/genética , Adulto , Anciano , Anciano de 80 o más Años , Desequilibrio Alélico , Diferenciación Celular/genética , Hibridación Genómica Comparativa/métodos , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genes Relacionados con las Neoplasias/genética , Estudio de Asociación del Genoma Completo/métodos , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Gliosarcoma/metabolismo , Gliosarcoma/patología , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Análisis de Supervivencia , Tetraspaninas
12.
Clin Neuropathol ; 32(6): 508-14, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23863343

RESUMEN

A rare case of gliosarcoma which arose in the temporal lobe of a 39-yearold man was reported. The gliomatous area of the tumor showed ependymal differentiation, and also contained immature neuroectodermal tissue resembling a primitive neuroectodermal tumor (PNET) in addition to an ordinary glioblastomatous component. Tumor cells in the PNET-like component were immunoreactive for synaptophysin, CD99, neurogenin 3, and α-internexin, but not for glial fibrillary acidic protein (GFAP), Class III-ß tubulin, or Neu N. The mesenchymal area exhibited a compact fascicular proliferation of atypical spindle cells invested by fine reticulin fibrils. In addition, these cells were immunoreactive for Slug and Twist - transcription factors which are involved in the "epithelial-mesenchymal transition (EMT)" phenomenon. Gliosarcomas containing an ependymal or PNET-like component are rare, and to our knowledge, the present case is the first to be reported whose glial element exhibited differentiation toward these two components. The diverse differentiation in the glial element suggests that the tumor most likely originated from primitive neuroepithelial progenitor cells rather than from the neometaplasia of a glioblastoma. The immunoreactivity for transcription factors in the mesenchymal element indicated that EMT might be involved in the pathogenesis of this very rare type of gliosarcoma.


Asunto(s)
Neoplasias Encefálicas/patología , Gliosarcoma/patología , Lóbulo Temporal/patología , Adulto , Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/metabolismo , Diferenciación Celular , Epéndimo/patología , Transición Epitelial-Mesenquimal , Gliosarcoma/metabolismo , Humanos , Inmunohistoquímica , Masculino , Tumores Neuroectodérmicos Primitivos/metabolismo , Tumores Neuroectodérmicos Primitivos/patología
13.
Clin Neuropathol ; 32(3): 165-70, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23254140

RESUMEN

Gliosarcoma, a biphasic tumor with both mesenchymal and glial elements, is typically considered a variant of astrocytoma (glioblastoma), WHO Grade IV. A 57-year-old man presented with altered mental status and was found to have a large right frontal mass. Biopsy and subsequent subtotal resection revealed a WHO Grade II oligodendroglioma with classic histological features, expression of IDH1 R132H mutant protein, and chromosome 1p19q co-deletion. Fifteen months later, the patient developed recurrent tumor composed of intersecting fascicles of spindled cells with necrosis and a high mitotic index. The recurrent tumor stained for both mesenchymal and glial elements, consistent with the diagnosis of gliosarcoma, and showed retained IDH1 R132H expression. By FISH analysis, the gliosarcoma showed no evidence of 1p19q co-deletion. We performed SNP arrays and detailed SNP analysis of both the oligodendroglioma and the gliosarcoma. This demonstrated loss of heterozygosity (LOH) of chromosomes 1 and 19 in the gliosarcoma with retention of the same full-length chromosomes 1 and 19 found intact in the oligodendroglioma. Not surprisingly, the gliosarcoma harbored multiple additional alterations, consistent with clonal evolution. There have been only rare reports of sarcomatous transformation of oligodendroglioma ("oligosarcoma") and most were published prior to the development of modern genetic modalities. Here we present a case with detailed genetic evidence that suggests that mesenchymal metaplasia sarcomatous transformation is possible in classic oligodendrogliomas with 1p19q codeletions.


Asunto(s)
Neoplasias Encefálicas/patología , Gliosarcoma/patología , Neoplasias Primarias Secundarias/patología , Oligodendroglioma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Gliosarcoma/genética , Gliosarcoma/metabolismo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/metabolismo , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple
14.
Radiology ; 265(3): 743-52, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22996750

RESUMEN

PURPOSE: To assess the reproducibility of the magnetic resonance (MR) estimate of blood oxygen saturation (sO(2)) in the rat brain, to evaluate the relationship between low MR estimate of sO(2) values and tissue hypoxia in a hypoxic and necrotic glioscarcoma model (9L gliosarcoma cells), and to evaluate the capability of the MR estimate of sO(2) parameter to help identify modifications induced by an antiangiogenic treatment (sorafenib) in 9L gliosarcoma tumors. MATERIALS AND METHODS: Experiments were performed with permits from the French Ministry of Agriculture. Forty-eight male rats bearing a 9L gliosarcoma were randomized in untreated and treated (sorafenib) groups. MR blood volume fraction and MR estimate of sO(2) parameters were estimated 1 day before and 1, 3, 5, and 8 days after the start of the treatment. The in vivo MR estimate of sO(2) measurement was correlated with the ex vivo hypoxia assessment by using pimonidazole staining. Paired and unpaired t tests, as well as parametric Pearson tests, were used for the statistical analyses. RESULTS: In healthy tissues, MR estimate of sO(2) measurements were comparable to literature values and were reproducible (mean across all animals, 68.0% ± 6.5 [standard deviation]). In untreated tumors, MR estimate of sO(2) and immunohistochemical analysis yielded correlated fractional hypoxic-necrotic areas (R(2) = 0.81). In tumors treated with antiangiogenic therapy, tumor MR estimate of sO(2) was decreased with respect to the healthy tissue (P< .001). CONCLUSION: Results of this study suggest that the MR estimate of sO(2) is a reproducible estimate that could be used as an in vivo probe of hypoxia in brain tumors and as a sensitive reporter of the hypoxic effects of antiangiogenic therapies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Bencenosulfonatos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Gliosarcoma/tratamiento farmacológico , Gliosarcoma/metabolismo , Imagen por Resonancia Magnética/métodos , Oxígeno/sangre , Piridinas/farmacología , Análisis de Varianza , Animales , Dextranos/administración & dosificación , Hipoxia Encefálica/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Modelos Lineales , Nanopartículas de Magnetita/administración & dosificación , Masculino , Niacinamida/análogos & derivados , Nitroimidazoles/administración & dosificación , Compuestos de Fenilurea , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Reproducibilidad de los Resultados , Sorafenib
15.
Magn Reson Med ; 68(4): 1074-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22246684

RESUMEN

Chemical exchange saturation transfer MRI is a promising new technique for cellular and molecular imaging. This contrast allows the detection of tumors and ischemia without the use of gadolinium as well as the design of microenvironment-sensitive probes that can be discriminated based on their exchange contrast properties and saturation frequency. Current acquisition schemes to detect and analyze this contrast suffer from sensitivity to spatial B0 inhomogeneity and low contrast-to-noise-ratio, which is an obstacle to widespread adoption of the technology. A new method to detect chemical exchange saturation transfer contrast is proposed here, termed "length and offset varied saturation" which acquires a set of images with the saturation parameters varied so as to modulate the exchange contrast. Either fast fourier transform or the general linear model can be employed to decompose the modulation patterns into separate sources of water signal loss. After transformation, a length and offset varied saturation phase map is generated, which is insensitive to B0 inhomogeneity. When collected on live mice bearing 9L gliosarcomas, and compared to the conventional asymmetry in the magnetization transfer ratio map using offset increment correction, the results show that length and offset varied saturation phase mapping obtains about three to four times contrast-to-noise-ratio and exhibits less B0 artifacts.


Asunto(s)
Algoritmos , Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Gliosarcoma/diagnóstico , Gliosarcoma/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Animales , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Imagen Molecular/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
16.
Stem Cells ; 28(2): 181-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19937755

RESUMEN

Cancer stem cells (CSCs) are characterized by their self-renewing potential and by their ability to differentiate and phenocopy the original tumor in orthotopic xenografts. Long-term propagation of glioblastoma (GBM) cells in serum-containing medium results in loss of the CSCs and outgrowth of cells genetically and biologically divergent from the parental tumors. In contrast, the use of a neurosphere assay, a serum-free culture for selection, and propagation of central nervous system-derived stem cells allows the selection of a subpopulation containing CSCs. Gliosarcoma (GS), a morphological variant comprising approximately 2% of GBMs, present a biphasic growth pattern, composed of glial and metaplastic mesenchymal components. To assess whether the neurosphere assay would allow the amplification of a subpopulation of cells with "gliosarcoma stem cell" properties, capable of propagating both components of this malignancy, we have generated neurospheres and serum cultures from primary GS and GBM surgical specimens. Neurosphere cultures from GBM and GS samples expressed neural stem cell markers Sox2, Musashi1, and Nestin. In contrast to the GBM neurosphere lines, the GS neurospheres were negative for the stem cell marker CD133. All neurosphere lines generated high-grade invasive orthotopic tumor xenografts, with histological features strikingly similar to the parental tumors, demonstrating that these cultures indeed are enriched in CSCs. Remarkably, low-passage GS serum cultures retained the expression of stem cell markers, the ability to form neurospheres, and tumorigenicity. The GS experimental tumors phenocopied the parental tumor, exhibiting biphasic glial and mesenchymal components, constituting a clinically relevant model to investigate mesenchymal differentiation in GBMs.


Asunto(s)
Diferenciación Celular/fisiología , Glioblastoma/patología , Gliosarcoma/patología , Células Madre Mesenquimatosas/fisiología , Células Madre Neoplásicas/citología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Western Blotting , Diferenciación Celular/genética , Glioblastoma/metabolismo , Gliosarcoma/metabolismo , Glicoproteínas/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteínas de Filamentos Intermediarios/metabolismo , Imagen por Resonancia Magnética , Células Madre Mesenquimatosas/citología , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nestina , Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
17.
J Neurooncol ; 105(2): 317-23, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21533839

RESUMEN

Gliosarcoma is a variant of glioblastoma and is characterized by distinct glial and sarcomatous components. Typically, there is no macroscopic boundary between the components and special stains are often required to distinguish the glial and sarcomatous elements. Some studies suggest similar genetic alterations in both components pointing to a common origin. We present an extreme case of gliosarcoma arising as a discrete fibrous nodule adjacent to a typical glioblastoma. A 65 year-old woman presented with progressive weakness, seizures and right-sided hemiparesis. CT scan demonstrated an irregular enhancing left frontal lobe mass and an adjacent discrete nodule with different imaging characteristics. The unique nature of this macroscopically biphasic neoplasm allowed us to compare the molecular characteristics of glial and sarcomatous elements which were strikingly similar except for small losses and gains in Chr 3. Studies are under way to determine the significance of chromosome 3 alterations in gliosarcomas.


Asunto(s)
Neoplasias Encefálicas/patología , Lóbulo Frontal/patología , Glioblastoma/patología , Gliosarcoma/etiología , Gliosarcoma/patología , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/metabolismo , Femenino , Lóbulo Frontal/metabolismo , Glioblastoma/complicaciones , Glioblastoma/metabolismo , Gliosarcoma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Pronóstico
18.
Nat Med ; 2(5): 567-73, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8616717

RESUMEN

Cytosine arabinoside (ara-C) is a cytidine analog that incorporates into replicating DNA and induces lethal DNA strand breaks. Although ara-C is a potent antitumor agent for hematologic malignancies, it has only minimal activity against most solid tumors. The rate-limiting step in intracellular ara-C activation is phosphorylation of the prodrug by deoxycytidine kinase (dCK). The present results demonstrate that both retroviral and adenoviral vector-mediated transduction of the dCK cDNA results in marked sensitization of glioma cells lines to the cytotoxic effects of ara-C in vitro. We also demonstrate that ara-C treatment of established intradermal and intracerebral gliomas transduced with dCK results in significant antitumor effects in vivo. These data suggest that viral vector transduction of the dCK gene followed by treatment with ara-C represents a new chemosensitization strategy for cancer gene therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Citarabina/farmacología , Desoxicitidina Quinasa/metabolismo , Gliosarcoma/tratamiento farmacológico , Profármacos/farmacología , Adenoviridae/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citarabina/metabolismo , Desoxicitidina Quinasa/genética , Relación Dosis-Respuesta a Droga , Vectores Genéticos , Gliosarcoma/genética , Gliosarcoma/metabolismo , Humanos , Masculino , Profármacos/metabolismo , Ratas , Ratas Endogámicas F344 , Sarcoma Experimental/tratamiento farmacológico , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Análisis de Supervivencia , Transfección
19.
J Comput Assist Tomogr ; 35(6): 667-73, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22082533

RESUMEN

OBJECTIVE: To investigate the features of pathologically confirmed gliosarcomas using computed tomography (CT) and magnetic resonance (MR) imaging. METHODS: We retrospectively reviewed the cross-sectional CT and MR images of 54 patients (37 males and 17 females; mean age, 44.5 years; range, 13-74 years) with gliosarcomas confirmed by histopathology. RESULTS: Across all patients, there were 59 lesions. On nonenhanced CT and MR images, tumors were predominantly inhomogeneous. On the postcontrast CT and MR images, 50 (84.7%) irregular lesions had thick walls with a strong rim- and ringlike enhancement, whereas the remaining 9 (15.3%) round or oval lesions had even thin walls with an enhanced peripheral ring. Magnetic resonance spectroscopy showed increased choline and lactate values, along with decreased N-acetylaspartate and creatine values. On diffusion-weighted imaging, the tumor was slightly or markedly hyperintense compared with the white matter. CONCLUSION: A well-demarcated mass located peripherally, with rimlike or ring enhancement, is a common presentation of gliosarcoma on CT and MR images. In addition, magnetic resonance spectroscopy and diffusion-weighted imaging can be used to make a differential diagnosis.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Gliosarcoma/diagnóstico , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X/métodos , Adolescente , Adulto , Anciano , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Colina/metabolismo , Medios de Contraste , Creatina/metabolismo , Imagen de Difusión por Resonancia Magnética , Femenino , Gadolinio DTPA , Gliosarcoma/diagnóstico por imagen , Gliosarcoma/metabolismo , Humanos , Yohexol , Lactatos/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
20.
Clin Neurol Neurosurg ; 207: 106771, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34198223

RESUMEN

BACKGROUND: Captopril is a well-characterized, FDA-approved drug that has demonstrated promise as a repurposed oncology therapeutic. Captopril's known anti-cancer effects include inhibition of Matrix Metalloproteinase-2 (MMP-2), an endopeptidase which selectively breaks down the extracellular matrix to promote cell migration. MMP-2 is a known therapeutic target in gliomas, tumors with significant clinical need. Using an aggressive gliosarcoma model, we assessed captopril's effects on MMP-2 expression in vitro and in vivo as well as its efficacy as an adjuvant in combination therapy regimens in vivo. METHODS: Following captopril treatment, MMP-2 protein expression and migratory capabilities of 9 L gliosarcoma cells were assessed in vitro via western blots and scratch wound assays, respectively. Rats were intracranially implanted with 9 L gliosarcoma tumors, and survival was assessed in the following groups: control; captopril (30 mg/kg/day); temozolomide (TMZ) (50 mg/kg/day), and captopril+TMZ. In vivo experiments were accompanied by immunohistochemistry for MMP-2 from brain tissue. RESULTS: In vitro, captopril decreased MMP-2 protein expression and reduced migratory capacity in 9 L gliosarcoma cells. In a gliosarcoma animal model, captopril decreased MMP-2 protein expression and extended survival as a TMZ adjuvant relative to untreated controls, captopril monotherapy, and TMZ monotherapy groups (27.5 versus 14 (p < 0.001), 16 (p < 0.001), and 23 (p = 0.018) days, respectively). CONCLUSIONS: Captopril decreases gliosarcoma cell migration, which may be mediated by reduction in MMP-2 protein expression. Captopril provided a survival advantage as a TMZ adjuvant in a rat intracranial gliosarcoma model. Captopril may represent a promising potential adjuvant to TMZ therapy in gliosarcoma as a modulator of the MMP-2 pathway.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Captopril/uso terapéutico , Gliosarcoma/tratamiento farmacológico , Animales , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Femenino , Gliosarcoma/metabolismo , Gliosarcoma/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Ratas , Ratas Endogámicas F344 , Temozolomida/uso terapéutico , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA