Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 38(5-6): 273-288, 2024 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-38589034

RESUMEN

Glioblastoma is universally fatal and characterized by frequent chromosomal copy number alterations harboring oncogenes and tumor suppressors. In this study, we analyzed exome-wide human glioblastoma copy number data and found that cytoband 6q27 is an independent poor prognostic marker in multiple data sets. We then combined CRISPR-Cas9 data, human spatial transcriptomic data, and human and mouse RNA sequencing data to nominate PDE10A as a potential haploinsufficient tumor suppressor in the 6q27 region. Mouse glioblastoma modeling using the RCAS/tv-a system confirmed that Pde10a suppression induced an aggressive glioma phenotype in vivo and resistance to temozolomide and radiation therapy in vitro. Cell culture analysis showed that decreased Pde10a expression led to increased PI3K/AKT signaling in a Pten-independent manner, a response blocked by selective PI3K inhibitors. Single-nucleus RNA sequencing from our mouse gliomas in vivo, in combination with cell culture validation, further showed that Pde10a suppression was associated with a proneural-to-mesenchymal transition that exhibited increased cell adhesion and decreased cell migration. Our results indicate that glioblastoma patients harboring PDE10A loss have worse outcomes and potentially increased sensitivity to PI3K inhibition.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Animales , Ratones , Glioblastoma/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Haploinsuficiencia , Glioma/genética , Fosfohidrolasa PTEN/genética , Hidrolasas Diéster Fosfóricas/genética , Línea Celular Tumoral , Neoplasias Encefálicas/genética
2.
Genes Dev ; 32(7-8): 512-523, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29632085

RESUMEN

Glioblastoma is the most frequently occurring and invariably fatal primary brain tumor in adults. The vast majority of glioblastomas is characterized by chromosomal copy number alterations, including gain of whole chromosome 7 and loss of whole chromosome 10. Gain of whole chromosome 7 is an early event in gliomagenesis that occurs in proneural-like precursor cells, which give rise to all isocitrate dehydrogenase (IDH) wild-type glioblastoma transcriptional subtypes. Platelet-derived growth factor A (PDGFA) is one gene on chromosome 7 known to drive gliomagenesis, but, given its location near the end of 7p, there are likely several other genes located along chromosome 7 that select for its increased whole-chromosome copy number within glioblastoma cells. To identify other potential genes that could select for gain of whole chromosome 7, we developed an unbiased bioinformatics approach that identified homeobox A5 (HOXA5) as a gene whose expression correlated with gain of chromosome 7 and a more aggressive phenotype of the resulting glioma. High expression of HOXA5 in glioblastoma was associated with a proneural gene expression pattern and decreased overall survival in both human proneural and PDGF-driven mouse glioblastoma. Furthermore, HOXA5 overexpression promoted cellular proliferation and potentiated radioresistance. We also found enrichment of HOXA5 expression in recurrent human and mouse glioblastoma at first recurrence after radiotherapy. Overall, this study implicates HOXA5 as a chromosome 7-associated gene-level locus that promotes selection for gain of whole chromosome 7 and an aggressive phenotype in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 7 , Glioblastoma/genética , Proteínas de Homeodominio/metabolismo , Fosfoproteínas/metabolismo , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Proliferación Celular , Duplicación Cromosómica , Glioblastoma/mortalidad , Glioblastoma/patología , Glioblastoma/radioterapia , Proteínas de Homeodominio/genética , Humanos , Isocitrato Deshidrogenasa/genética , Ratones , Recurrencia Local de Neoplasia , Fosfoproteínas/genética , Tolerancia a Radiación , Factores de Transcripción
3.
BMC Cancer ; 24(1): 82, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38225589

RESUMEN

BACKGROUND: Glioblastoma is the most common and most aggressive malignant primary brain tumor in adults. Glioblastoma cells synthesize and secrete large quantities of the excitatory neurotransmitter glutamate, driving epilepsy, neuronal death, tumor growth and invasion. Moreover, neuronal networks interconnect with glioblastoma cell networks through glutamatergic neuroglial synapses, activation of which induces oncogenic calcium oscillations that are propagated via gap junctions between tumor cells. The primary objective of this study is to explore the efficacy of brain-penetrating anti-glutamatergic drugs to standard chemoradiotherapy in patients with glioblastoma. METHODS/DESIGN: GLUGLIO is a 1:1 randomized phase Ib/II, parallel-group, open-label, multicenter trial of gabapentin, sulfasalazine, memantine and chemoradiotherapy (Arm A) versus chemoradiotherapy alone (Arm B) in patients with newly diagnosed glioblastoma. Planned accrual is 120 patients. The primary endpoint is progression-free survival at 6 months. Secondary endpoints include overall and seizure-free survival, quality of life of patients and caregivers, symptom burden and cognitive functioning. Glutamate levels will be assessed longitudinally by magnetic resonance spectroscopy. Other outcomes of interest include imaging response rate, neuronal hyperexcitability determined by longitudinal electroencephalography, Karnofsky performance status as a global measure of overall performance, anticonvulsant drug use and steroid use. Tumor tissue and blood will be collected for translational research. Subgroup survival analyses by baseline parameters include segregation by age, extent of resection, Karnofsky performance status, O6-methylguanine DNA methyltransferase (MGMT) promotor methylation status, steroid intake, presence or absence of seizures, tumor volume and glutamate levels determined by MR spectroscopy. The trial is currently recruiting in seven centers in Switzerland. TRIAL REGISTRATION: NCT05664464. Registered 23 December 2022.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Quimioradioterapia , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Reposicionamiento de Medicamentos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glutamatos , Estudios Multicéntricos como Asunto , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Esteroides/uso terapéutico
4.
Acta Neuropathol ; 146(2): 173-190, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37368072

RESUMEN

Meningiomas are the most common primary intracranial tumors. Although most symptomatic cases can be managed by surgery and/or radiotherapy, a relevant number of patients experience an unfavorable clinical course and additional treatment options are needed. As meningiomas are often perfused by dural branches of the external carotid artery, which is located outside the blood-brain barrier, they might be an accessible target for immunotherapy. However, the landscape of naturally presented tumor antigens in meningioma is unknown. We here provide a T-cell antigen atlas for meningioma by in-depth profiling of the naturally presented immunopeptidome using LC-MS/MS. Candidate target antigens were selected based on a comparative approach using an extensive immunopeptidome data set of normal tissues. Meningioma-exclusive antigens for HLA class I and II are described here for the first time. Top-ranking targets were further functionally characterized by showing their immunogenicity through in vitro T-cell priming assays. Thus, we provide an atlas of meningioma T-cell antigens which will be publicly available for further research. In addition, we have identified novel actionable targets that warrant further investigation as an immunotherapy option for meningioma.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/terapia , Cromatografía Liquida , Espectrometría de Masas en Tándem , Inmunoterapia , Linfocitos T , Neoplasias Meníngeas/terapia
5.
Adv Exp Med Biol ; 1416: 225-234, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37432631

RESUMEN

Systemic treatment approaches are urgently needed for a subset of meningioma patients who do not achieve local tumor control with surgery and radiotherapy. Classical chemotherapy or anti-angiogenic agents exert only very limited activity in these tumors. Long-term survival of patients with advanced metastatic cancer following treatment with immune checkpoint inhibitors, i.e., monoclonal antibodies designed to unleash suppressed anticancer immune responses, has fostered hopes for benefit from similar approaches in patients with meningiomas that recur after standard local therapy. Moreover, a plethora of immunotherapy approaches beyond these drugs have entered clinical development or clinical practice for other cancer entities, including (i) novel immune checkpoint inhibitors that may act independently of T cell activity, (ii) cancer peptide or dendritic cell vaccines to induce anticancer immunity utilizing cancer-associated antigens, (iii) cellular therapies utilizing genetically modified peripheral blood cells to directly target cancer cells, (iv) T cell engaging recombinant proteins that link tumor antigen-binding sites to effector cell activating or recognition domains, or to immunogenic cytokines, and (v) oncolytic virotherapy utilizing attenuated viral vectors designed to specifically infect cancer cells, seeking to elicit systemic anticancer immunity. This chapter provides an overview of the principles of immunotherapy, summarizes ongoing immunotherapy clinical trials in meningioma patients, and discusses the applicability of established and emerging immunotherapy concepts to meningioma patients.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Virus Oncolíticos , Humanos , Meningioma/terapia , Inhibidores de Puntos de Control Inmunológico , Recurrencia Local de Neoplasia , Inmunoterapia , Antígenos de Histocompatibilidad Clase II , Neoplasias Meníngeas/terapia
6.
Brain ; 144(11): 3322-3327, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33974079

RESUMEN

Pro-tumorigenic electrochemical synapses between neurons and brain tumour cells in preclinical studies suggest unfavourable effects of epilepsy on patient survival. We investigated associations of epilepsy and survival in three cohorts of brain tumour patients (meningioma, glioblastoma and brain metastases). Cohorts were segregated into three groups for comparative analyses: (i) no epilepsy; (ii) epilepsy without status epilepticus; and (iii) status epilepticus. Status epilepticus was considered a surrogate of extensive neuronal hyperexcitability. The main outcome was progression-free survival (meningioma) and overall survival (glioblastoma and brain metastases), adjusted for established prognostic factors and onset of epilepsy by time-dependent multivariate Cox modelling. The primary analysis population comprised 1792 patients (742 meningioma, 249 glioblastoma, 801 brain metastases). Epilepsy was associated with favourable prognostic factors. However, on multivariate analyses, status epilepticus was associated with inferior overall survival of patients with glioblastoma [status epilepticus versus no epilepsy multivariate hazard ratio (HR) 3.72, confidence interval (CI) 1.78-7.76, P < 0.001] and brain metastases (status epilepticus versus no epilepsy HR 2.30, CI 1.10-4.79, P = 0.026). Among brain metastases patients, but not among patients with meningioma or glioblastoma, epilepsy was similarly associated with inferior overall survival (epilepsy versus no epilepsy HR 2.16, CI 1.60-2.93, P < 0.001). We conclude that epilepsy may convey inferior survival of patients with malignant brain tumours.


Asunto(s)
Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/mortalidad , Epilepsia/etiología , Estudios de Cohortes , Femenino , Glioblastoma/complicaciones , Glioblastoma/mortalidad , Humanos , Masculino , Neoplasias Meníngeas/complicaciones , Neoplasias Meníngeas/mortalidad , Meningioma/complicaciones , Meningioma/mortalidad , Pronóstico , Supervivencia sin Progresión
7.
Qual Life Res ; 29(7): 1801-1808, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32128657

RESUMEN

PURPOSE: Long-term impairment of quality of life (QoL) occurs in a subset of meningioma patients, even after curative surgical resection. We sought to explore socioeconomic burden of meningioma surgery and associations with post-operative QoL to identify patients at risk for inferior outcome. METHODS: All patients with histological diagnosis of an intracranial meningioma treated at a single institution 2000-2013 were screened for inclusion in this cross-sectional survey study. Surveys comprised tools to assess socioeconomic status including social deprivation, QoL and symptom burden. Multivariate binary regression models controlling for established prognostic factors were applied to explore associations of socioeconomics with QoL 1 year after surgery. RESULTS: Completed surveys were returned by 249 patients. The median age at diagnosis was 56 years (SD ± 12), 185 patients (74%) were female and 219 (88%) had World Health Organization grade I meningiomas. One year after surgery, there was a 20% decrease in the number of patients working (p < 0.001), 22% of full-time working patients transitioned to part-time work (p < 0.001) and more patients depended on professional care (14% versus 4%, p < 0.001). Patients reported improved QoL, including improved global health (effect: 21%, 95% confidence interval [1] 15-26%), headaches (effect: 19%, CI 13-24%) and seizures (effect: 12%, CI 8-17%). On multivariable analyses, QoL after meningioma surgery was associated with preoperative employment status (odds ratio [OR] 0.41, 95% CI 0.17-0.98) and subjective work ability (OR 0.37, 95% CI 0.15-0.92). CONCLUSION: In a subset of meningioma patients, there is marked socioeconomic burden, which may be associated with inferior patient-reported outcome.


Asunto(s)
Meningioma/epidemiología , Meningioma/psicología , Calidad de Vida/psicología , Factores Socioeconómicos , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Encuestas y Cuestionarios
8.
Acta Neuropathol ; 138(2): 295-308, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31069492

RESUMEN

DNA methylation patterns delineate clinically relevant subgroups of meningioma. We previously established the six meningioma methylation classes (MC) benign 1-3, intermediate A and B, and malignant. Here, we set out to identify subgroup-specific mutational patterns and gene regulation. Whole genome sequencing was performed on 62 samples across all MCs and WHO grades from 62 patients with matched blood control, including 40 sporadic meningiomas and 22 meningiomas arising after radiation (Mrad). RNA sequencing was added for 18 of these cases and chromatin-immunoprecipitation for histone H3 lysine 27 acetylation (H3K27ac) followed by sequencing (ChIP-seq) for 16 samples. Besides the known mutations in meningioma, structural variants were found as the mechanism of NF2 inactivation in a small subset (5%) of sporadic meningiomas, similar to previous reports for Mrad. Aberrations of DMD were found to be enriched in MCs with NF2 mutations, and DMD was among the most differentially upregulated genes in NF2 mutant compared to NF2 wild-type cases. The mutational signature AC3, which has been associated with defects in homologous recombination repair (HRR), was detected in both sporadic meningioma and Mrad, but widely distributed across the genome in sporadic cases and enriched near genomic breakpoints in Mrad. Compared to the other MCs, the number of single nucleotide variants matching the AC3 pattern was significantly higher in the malignant MC, which also exhibited higher genomic instability, determined by the numbers of both large segments affected by copy number alterations and breakpoints between large segments. ChIP-seq analysis for H3K27ac revealed a specific activation of genes regulated by the transcription factor FOXM1 in the malignant MC. This analysis also revealed a super enhancer near the HOXD gene cluster in this MC, which, together with general upregulation of HOX genes in the malignant MC, indicates a role of HOX genes in meningioma aggressiveness. This data elucidates the biological mechanisms rendering different epigenetic subgroups of meningiomas, and suggests leveraging HRR as a novel therapeutic target.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Meníngeas/clasificación , Meningioma/clasificación , Mutación , Inmunoprecipitación de Cromatina , Dosificación de Gen , Inestabilidad Genómica , Humanos , Neoplasias Meníngeas/etiología , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/etiología , Meningioma/genética , Meningioma/patología , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , ARN Neoplásico/genética , Reparación del ADN por Recombinación , Alineación de Secuencia , Factores de Transcripción/fisiología , Transcriptoma , Secuenciación Completa del Genoma
10.
Acta Neuropathol ; 135(6): 955-963, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29627952

RESUMEN

Epigenetic patterns on the level of DNA methylation have already been shown to separate clinically relevant subgroups of meningiomas. We here set out to identify potential prognostic implications of epigenetic modification on the level of histones with focus on H3K27 trimethylation (H3K27me3). H3K27me3 was assessed by immunohistochemistry on 232 meningiomas from 232 patients. In 194 cases, trimethylation was detected in tumor cells. In 25 cases, staining was limited to vessels while all tumor cells were negative. Finally, 13 cases yielded equivocal staining patterns. Reduced abundance of H3K27me3 in cases with staining limited to vessels was confirmed by mass spectrometry on a subset of cases. Lack of staining for H3K27me3 in all tumor cells was significantly associated with more rapid progression (p = 0.009). In line, H3K27me3-negative cases were associated with a DNA methylation pattern of the more aggressive types among the recently introduced DNA methylation groups. Also, NF2 and SUFU mutations were enriched among cases with complete lack of H3K27me3 staining in tumor cells (p < 0.0001 and p = 0.029, respectively). H3K27me3 staining pattern added significant prognostic insight into WHO grade II cases and in the compound subset of WHO grade I and II cases (p = 0.04 and p = 0.007, respectively). However, it did not further stratify within WHO grade III cases. Collectively, these data indicate that epigenetic modifications beyond DNA methylation are involved in the aggressiveness of meningioma. It also suggests that H3K27me3 immunohistochemistry might be a useful adjunct in meningioma diagnostics, particularly for cases with WHO grade II histology or at the borderline between WHO grade I and II.


Asunto(s)
Metilación de ADN , Histonas/metabolismo , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Estudios de Cohortes , Epigénesis Genética , Femenino , Genes de la Neurofibromatosis 2 , Histonas/genética , Humanos , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/genética , Meningioma/patología , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Pronóstico , Supervivencia sin Progresión , Proteínas Represoras/genética
11.
J Neurooncol ; 140(3): 659-667, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30196368

RESUMEN

PURPOSE: Preoperative embolization of radiographically suspected meningiomas is often performed to facilitate tumor resection. Its effects on the subsequent disease course of meningioma patients have not been studied in detail and randomized trials are lacking. The purpose of this study was to explore associations of preoperative meningioma embolization with postoperative outcome. PATIENTS AND METHODS: Patients undergoing resection of an intracranial meningioma at the University Hospital Zurich 2000-2013 (N = 741) were reviewed for the inclusion of pre-operative embolization in the management strategy. Annotations included demographics, radiographic, surgical, histological and hematological parameters, cardiovascular risk factors, pre- and postoperative neurological function and gene methylation-based classification. Binary regression and Cox proportional hazards models were applied to determine factors associated with outcome. RESULTS: Pre-operative embolization was performed in 337 patients (42%). Cardiovascular events after surgery comprised mostly deep vein thrombosis (N = 39) and pulmonary embolisms (N = 64). On multivariate analyses of post-operative cardiovascular adverse events controlling for established risk factors, there were associations with embolization (OR 2.38, 95% CI 1.37-4.00), and with female gender (OR 2.18, 95% CI 1.17-4.08). Recurrence-free survival (RFS) of embolized patients was less favorable among patients with WHO grade II or grade III meningiomas (median RFS: 4.3 vs. 7.0 years, P = 0.029) or in patients with intermediate or malignant gene methylation subtype meningiomas (median RFS: 2.0 vs. 8.2 years, P = 0.005). CONCLUSION: Pre-operative meningioma embolization may cause adverse outcomes. Randomized trials to determine benefit-risk ratios are warranted to clarify the role of pre-operative embolization for the treatment of meningioma patients.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Embolización Terapéutica/efectos adversos , Neoplasias Meníngeas/terapia , Meningioma/terapia , Recurrencia Local de Neoplasia/epidemiología , Cuidados Preoperatorios/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Enfermedades Cardiovasculares/epidemiología , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Neoplasias Meníngeas/complicaciones , Neoplasias Meníngeas/epidemiología , Meningioma/complicaciones , Meningioma/epidemiología , Persona de Mediana Edad , Complicaciones Posoperatorias , Estudios Retrospectivos , Adulto Joven
12.
Ther Umsch ; 75(7): 438-443, 2018.
Artículo en Alemán | MEDLINE | ID: mdl-30935361

RESUMEN

Update on primary brain tumours Abstract. Primary brain tumors comprise a heterogeneous group of clinically, histologically and molecularly distinct entities. The World Health Organisation (WHO) revised the classification of tumors of the central nervous system in 2016 and defined molecular markers for the diagnosis of some entities. For example, co-deletion of chromosome arms 1 p and 19 q now defines oligodendrogliomas, irrespective of their histopathological appearance. Astrocytomas are segregated into such with mutated isocitrate dehydrogenase (IDH) and better prognosis, and such with IDH wildtype, which also comprise the vast majority of glioblastomas, the most common and prognostically least favorable malignant primary brain tumors in adults. Here, we provide an overview of the most common primary brain tumor entities and implications of the 2016 classification for the clinical management. Moreover, we provide an outlook on novel therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas , Glioma , Adulto , Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Humanos , Isocitrato Deshidrogenasa , Pronóstico , Organización Mundial de la Salud
13.
Lancet Oncol ; 18(5): 682-694, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28314689

RESUMEN

BACKGROUND: The WHO classification of brain tumours describes 15 subtypes of meningioma. Nine of these subtypes are allotted to WHO grade I, and three each to grade II and grade III. Grading is based solely on histology, with an absence of molecular markers. Although the existing classification and grading approach is of prognostic value, it harbours shortcomings such as ill-defined parameters for subtypes and grading criteria prone to arbitrary judgment. In this study, we aimed for a comprehensive characterisation of the entire molecular genetic landscape of meningioma to identify biologically and clinically relevant subgroups. METHODS: In this multicentre, retrospective analysis, we investigated genome-wide DNA methylation patterns of meningiomas from ten European academic neuro-oncology centres to identify distinct methylation classes of meningiomas. The methylation classes were further characterised by DNA copy number analysis, mutational profiling, and RNA sequencing. Methylation classes were analysed for progression-free survival outcomes by the Kaplan-Meier method. The DNA methylation-based and WHO classification schema were compared using the Brier prediction score, analysed in an independent cohort with WHO grading, progression-free survival, and disease-specific survival data available, collected at the Medical University Vienna (Vienna, Austria), assessing methylation patterns with an alternative methylation chip. FINDINGS: We retrospectively collected 497 meningiomas along with 309 samples of other extra-axial skull tumours that might histologically mimic meningioma variants. Unsupervised clustering of DNA methylation data clearly segregated all meningiomas from other skull tumours. We generated genome-wide DNA methylation profiles from all 497 meningioma samples. DNA methylation profiling distinguished six distinct clinically relevant methylation classes associated with typical mutational, cytogenetic, and gene expression patterns. Compared with WHO grading, classification by individual and combined methylation classes more accurately identifies patients at high risk of disease progression in tumours with WHO grade I histology, and patients at lower risk of recurrence among WHO grade II tumours (p=0·0096) from the Brier prediction test). We validated this finding in our independent cohort of 140 patients with meningioma. INTERPRETATION: DNA methylation-based meningioma classification captures clinically more homogenous groups and has a higher power for predicting tumour recurrence and prognosis than the WHO classification. The approach presented here is potentially very useful for stratifying meningioma patients to observation-only or adjuvant treatment groups. We consider methylation-based tumour classification highly relevant for the future diagnosis and treatment of meningioma. FUNDING: German Cancer Aid, Else Kröner-Fresenius Foundation, and DKFZ/Heidelberg Institute of Personalized Oncology/Precision Oncology Program.


Asunto(s)
Metilación de ADN , Neoplasias Meníngeas/clasificación , Neoplasias Meníngeas/genética , Meningioma/clasificación , Meningioma/genética , Recurrencia Local de Neoplasia/genética , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Genoma , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Neoplasias Meníngeas/patología , Meningioma/patología , Clasificación del Tumor , Recurrencia Local de Neoplasia/patología , Neurofibromina 2/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-akt/genética , Estudios Retrospectivos , Análisis de Secuencia de ARN , Receptor Smoothened/genética , Tasa de Supervivencia , Factores de Transcripción/genética , Transcriptoma , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
14.
Curr Treat Options Oncol ; 17(10): 51, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27501915

RESUMEN

OPINION STATEMENT: The revised World Health Organization (WHO) classification of tumors of the central nervous system of 2016 combines biology-driven molecular marker diagnostics with classical histological cancer diagnosis. Reclassification of gliomas by molecular similarity beyond histological boundaries improves outcome prediction and will increasingly guide treatment decisions. This change in paradigms implies more personalized and eventually more efficient therapeutic approaches, but the era of molecular targeted therapies for gliomas is yet at its onset. Promising results of molecularly targeted therapies in genetically less complex gliomas with circumscribed growth such as subependymal giant cell astrocytoma or pilocytic astrocytoma support further development of molecularly targeted therapies. In diffuse gliomas, several molecular markers that predict benefit from alkylating agent chemotherapy have been identified in recent years. For example, co-deletion of chromosome arms 1p and 19q predicts benefit from polychemotherapy with procarbazine, CCNU (lomustine), and vincristine (PCV) in patients with anaplastic oligodendroglioma, and the presence of 1p/19q co-deletion was integrated as a defining feature of oligodendroglial tumors in the revised WHO classification. However, the tremendous increase in knowledge of molecular drivers of diffuse gliomas on genomic, epigenetic, and gene expression levels has not yet translated into effective molecular targeted therapies. Multiple reasons account for the failure of early clinical trials of molecularly targeted therapies in diffuse gliomas, including the lack of molecular entry controls as well as pharmacokinetic and pharmacodynamics issues, but the key challenge of specifically targeting the molecular backbone of diffuse gliomas is probably extensive clonal heterogeneity. A more profound understanding of clonal selection, alternative activation of oncogenic signaling pathways, and genomic instability is warranted to identify effective combination treatments and ultimately improve survival.


Asunto(s)
Glioma/diagnóstico , Glioma/terapia , Técnicas de Diagnóstico Molecular , Biomarcadores de Tumor , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/terapia , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Aberraciones Cromosómicas , Metilación de ADN , Manejo de la Enfermedad , Glioma/etiología , Glioma/mortalidad , Histonas/genética , Histonas/metabolismo , Humanos , Terapia Molecular Dirigida , Mutación , Clasificación del Tumor , Estadificación de Neoplasias , Regiones Promotoras Genéticas , Resultado del Tratamiento
15.
Curr Opin Oncol ; 27(6): 502-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26397765

RESUMEN

PURPOSE OF REVIEW: Genetic, epigenetic, and expression analyses have refined the traditional, histopathology-based classification of diffusely infiltrating gliomas. This review summarizes these trends and implications for elderly patients. RECENT FINDINGS: The vast majority of diffusely infiltrating gliomas in elderly patients share an unfavorable molecular phenotype, that is, telomerase reverse transcriptase promoter mutation in the absence of isocitrate dehydrogenase (IDH) mutation and 1p/19q codeletion. Histopathologically, these are mostly astrocytic tumors and treatment is guided by the methylation status of the O6-methylguanine-DNA-methyltransferase (MGMT) promoter. 1p/19q codeletion indicates oligodendroglial histology and benefit from the addition of procarbazine, chlorethyl-cyclohexyl-nitroso-urea/lomustine, and vincristine polychemotherapy to radiotherapy. These tumors are almost exclusively associated with IDH mutations, but their molecular profile is rare in elderly patients. Two large phase III trials, RTOG 0825 and AVAglio, failed to demonstrate an overall survival benefit from antiangiogenic therapy with bevacizumab added to combined chemoradiotherapy (TMZ) in patients with newly diagnosed glioblastoma, but a trend toward improved survival with increasing age can be noted. Ongoing clinical trials in elderly patients with diffusely infiltrating glioma will clarify the role of combined chemoradiotherapy, and of bevacizumab or other antiangiogenic agents as an adjunct to radiotherapy. SUMMARY: The choice of first-line therapy in elderly patients with diffusely infiltrating glioma is between postoperative hypofractionated radiotherapy and chemotherapy, guided by MGMT methylation in most patients.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias del Sistema Nervioso Central/terapia , Glioma/terapia , Terapia Molecular Dirigida/métodos , Anciano , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/metabolismo , Ensayos Clínicos como Asunto , Terapia Combinada , Metilación de ADN , Metilasas de Modificación del ADN/genética , Epigenómica , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Telomerasa/genética
17.
Brain ; 137(Pt 2): 433-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24355709

RESUMEN

Thymosin beta 4 is a pleiotropic actin-sequestering polypeptide that is involved in wound healing and developmental processes. Thymosin beta 4 gene silencing promotes differentiation of neural stem cells whereas thymosin beta 4 overexpression initiates cortical folding of developing brain hemispheres. A role of thymosin beta 4 in malignant gliomas has not yet been investigated. We analysed thymosin beta 4 staining on tissue microarrays and performed interrogations of the REMBRANDT and the Cancer Genome Atlas databases. We investigated thymosin beta 4 expression in seven established glioma cell lines and seven glioma-initiating cell lines and induced or silenced thymosin beta 4 expression by lentiviral transduction in LNT-229, U87MG and GS-2 cells to study the effects of altered thymosin beta 4 expression on gene expression, growth, clonogenicity, migration, invasion, self-renewal and differentiation capacity in vitro, and tumorigenicity in vivo. Thymosin beta 4 expression increased with grade of malignancy in gliomas. Thymosin beta 4 gene silencing in LNT-229 and U87MG glioma cells inhibited migration and invasion, promoted starvation-induced cell death in vitro and enhanced survival of glioma-bearing mice. Thymosin beta 4 gene silencing in GS-2 cells inhibited self-renewal and promoted differentiation in vitro and decreased tumorigenicity in vivo. Gene expression analysis suggested a thymosin beta 4-dependent regulation of mesenchymal signature genes and modulation of TGFß and p53 signalling networks. We conclude that thymosin beta 4 should be explored as a novel molecular target for anti-glioma therapy.


Asunto(s)
Silenciador del Gen , Glioblastoma/genética , Invasividad Neoplásica/genética , Células Madre Neoplásicas/patología , Timosina/antagonistas & inhibidores , Timosina/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Bases de Datos Genéticas , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Células 3T3 NIH , Invasividad Neoplásica/patología , Células Madre Neoplásicas/fisiología , Timosina/biosíntesis
18.
Neurooncol Pract ; 11(2): 132-141, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38496908

RESUMEN

Background: Incidence rates of glioblastoma in very old patients are rising. The standard of care for this cohort is only partially defined and survival remains poor. The aims of this study were to reveal current practice of tumor-specific therapy and supportive care, and to identify predictors for survival in this cohort. Methods: Patients aged 80 years or older at the time of glioblastoma diagnosis were retrospectively identified in 6 clinical centers in Switzerland and France. Demographics, clinical parameters, and survival outcomes were annotated from patient charts. Cox proportional hazards modeling was performed to identify parameters associated with survival. Results: Of 107 patients, 45 were diagnosed by biopsy, 30 underwent subtotal resection, and 25 had gross total resection. In 7 patients, the extent of resection was not specified. Postoperatively, 34 patients did not receive further tumor-specific treatment. Twelve patients received radiotherapy with concomitant temozolomide, but only 2 patients had maintenance temozolomide therapy. Fourteen patients received temozolomide alone, 35 patients received radiotherapy alone, 1 patient received bevacizumab, and 1 took part in a clinical trial. Median progression-free survival (PFS) was 3.3 months and median overall survival (OS) was 4.2 months. Among patients who received any postoperative treatment, median PFS was 3.9 months and median OS was 7.2 months. Karnofsky performance status (KPS) ≥70%, gross total resection, and combination therapy were associated with better outcomes. The median time spent hospitalized was 30 days, accounting for 23% of the median OS. End-of-life care was mostly provided by nursing homes (n = 20; 32%) and palliative care wards (n = 16; 26%). Conclusions: In this cohort of very old patients diagnosed with glioblastoma, a large proportion was treated with best supportive care. Treatment beyond surgery and, in particular, combined modality treatment were associated with longer OS and may be considered for selected patients even at higher ages.

20.
Front Oncol ; 13: 1279933, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38023177

RESUMEN

Purpose: The selection of patients for further therapy after meningioma surgery remains a challenge. Progress has been made in this setting in selecting patients that are more likely to have an aggressive disease course by using molecular tests such as gene panel sequencing and DNA methylation profiling. The aim of this study was to create a preselection tool warranting further molecular work-up. Methods: All patients undergoing surgery for resection or biopsy of a cranial meningioma from January 2013 until December 2018 at the University Hospital Zurich with available tumor histology were included. Various prospectively collected clinical, radiological, histological and immunohistochemical variables were analyzed and used to train a logistic regression model to predict tumor recurrence or progression. Regression coefficients were used to generate a scoring system grading every patient into low, intermediate, and high-risk group for tumor progression or recurrence. Results: Out of a total of 13 variables preselected for this study, previous meningioma surgery, Simpson grade, progesterone receptor staining as well as presence of necrosis and patternless growth on histopathological analysis of 378 patients were included into the final model. Discrimination showed an AUC of 0.81 (95% CI 0.73 - 0.88), the model was well-calibrated. Recurrence-free survival was significantly decreased in patients in intermediate and high-risk score groups (p-value < 0.001). Conclusion: The proposed prediction model showed good discrimination and calibration. This prediction model is based on easily obtainable information and can be used as an adjunct for patient selection for further molecular work-up in a tertiary hospital setting.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA