Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
J Neurooncol ; 164(2): 331-339, 2023 Sep.
Article En | MEDLINE | ID: mdl-37665475

PURPOSE: To report the long-term outcomes in adult patients with grade 2 IDH-mutant astrocytoma treated with temozolomide (TMZ)-based chemoradiation. METHODS: One hundred and three patients with histologically proven grade 2 astrocytoma received radiation therapy (RT), 50.4-54 Gy in 1.8 Gy fractions, and adjuvant TMZ up to 12 cycles. Fifty-two patients received RT at the time of tumor progression and 51 in the early postoperative period for the presence of at least one high-risk feature (age > 40 years, preoperative tumor size > 5 cm, large postoperative residual tumor, tumor crossing the midline, or presence of neurological symptoms). Overall survival (OS) and progression-free survival (PFS) were calculated from the time of diagnosis. RESULTS: With a median follow-up time of 9.0 years (range, 1.3-15 years), median PFS and OS times were 9 years (95%CI, 6.6-10.3) and 11.8 years (95%CI, 9.3-13.4), respectively. Median PFS was 10.6 years in the early treatment group and 6 years in delayed treatment group (hazard ratio (HR) 0.30; 95%CI 0.16-0.59; p = 0.0005); however, OS was not significantly different between groups (12.8 vs. 10.4 years; HR 0.64; 95%CI 0.33-1.25; p = 0.23). Extent of resection, KPS, and small residual disease were associated with OS, with postoperative tumor ≤ 1 cc that emerged as the strongest independent predictor (HR: 0.27; 95%CI 0.08-0.87; p = 0.01). CONCLUSIONS: TMZ-based chemoradiation is associated with survival benefit in patients with grade 2 IDH-mutant astrocytoma. For this group of patients, chemoradiation can be deferred until time of progression in younger patients receiving extensive resection, while early treatment should be recommended in high-risk patients.


Astrocytoma , Brain Neoplasms , Humans , Adult , Temozolomide/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/diagnosis , Astrocytoma/genetics , Astrocytoma/therapy , Astrocytoma/pathology , Treatment Outcome
2.
Commun Biol ; 5(1): 780, 2022 08 02.
Article En | MEDLINE | ID: mdl-35918402

Glioblastoma multiforme (GBM) is the most frequent and aggressive form of primary brain tumor in the adult population; its high recurrence rate and resistance to current therapeutics urgently demand a better therapy. Regulation of protein stability by the ubiquitin proteasome system (UPS) represents an important control mechanism of cell growth. UPS deregulation is mechanistically linked to the development and progression of a variety of human cancers, including GBM. Thus, the UPS represents a potentially valuable target for GBM treatment. Using an integrated approach that includes proteomics, transcriptomics and metabolic profiling, we identify praja2, a RING E3 ubiquitin ligase, as the key component of a signaling network that regulates GBM cell growth and metabolism. Praja2 is preferentially expressed in primary GBM lesions expressing the wild-type isocitrate dehydrogenase 1 gene (IDH1). Mechanistically, we found that praja2 ubiquitylates and degrades the kinase suppressor of Ras 2 (KSR2). As a consequence, praja2 restrains the activity of downstream AMP-dependent protein kinase in GBM cells and attenuates the oxidative metabolism. Delivery in the brain of siRNA targeting praja2 by transferrin-targeted self-assembling nanoparticles (SANPs) prevented KSR2 degradation and inhibited GBM growth, reducing the size of the tumor and prolonging the survival rate of treated mice. These data identify praja2 as an essential regulator of cancer cell metabolism, and as a potential therapeutic target to suppress GBM growth.


Brain Neoplasms , Glioblastoma , Adult , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/metabolism , Humans , Mice , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin
3.
Clin Neuropathol ; 40(2): 108-117, 2021.
Article En | MEDLINE | ID: mdl-33191898

AIM: Diffuse midline glioma (DMG) H3 K27M-mutant is a specific entity that, as the name indicates, tends to occur in midline structures including the thalamus, brainstem, and spinal cord. DMG predominates in children, is an aggressive tumor with poor prognosis, and is considered a WHO grade IV tumor regardless of histological features. The exact frequency of these mutations in adults diagnosed with glioma in the midline is unknown. MATERIALS AND METHODS: We report a series of 6 more adult cases, and we critically review the current literature on adults with DMG H3 K27M-mutant. RESULTS: There were 5 males and 1 female. The age ranged from 26 to 52 years (median 39 years). All cases showed astrocytic differentiation, with positive staining for H3 K27M protein, and loss of H3 K27me in the tumor cells confirming the diagnosis. CONCLUSION: H3 K27M-mutant midline glioma can occur in adults, affecting midline structures. Increasing awareness of the reporting pathologists of this entity might help in a better determination of the frequency of mutant DMG in adults as well as better diagnosis and patient counseling of the outcome.


Brain Neoplasms , Glioma , Adult , Brain Neoplasms/genetics , Female , Glioma/genetics , Histones/genetics , Humans , Male , Middle Aged , Mutation/genetics , Thalamus
4.
Clin Epigenetics ; 11(1): 100, 2019 07 09.
Article En | MEDLINE | ID: mdl-31288858

BACKGROUND: Clustered protocadherins (PCDHs) map in tandem at human chromosome 5q31 and comprise three multi-genes clusters: α-, ß- and γ-PCDH. The expression of this cluster consists of a complex mechanism involving DNA hub formation through DNA-CCTC binding factor (CTCF) interaction. Methylation alterations can affect this interaction, leading to transcriptional dysregulation. In cancer, clustered PCDHs undergo a mechanism of long-range epigenetic silencing by hypermethylation. RESULTS: In this study, we detected frequent methylation alterations at CpG islands associated to these clustered PCDHs in all the solid tumours analysed (colorectal, gastric and biliary tract cancers, pilocytic astrocytoma), but not hematologic neoplasms such as chronic lymphocytic leukemia. Importantly, several altered CpG islands were associated with CTCF binding sites. Interestingly, our analysis revealed a hypomethylation event in pilocytic astrocytoma, suggesting that in neuronal tissue, where PCDHs are highly expressed, these genes become hypomethylated in this type of cancer. On the other hand, in tissues where PCDHs are lowly expressed, these CpG islands are targeted by DNA methylation. In fact, PCDH-associated CpG islands resulted hypermethylated in gastrointestinal tumours. CONCLUSIONS: Our study highlighted a strong alteration of the clustered PCDHs methylation pattern in the analysed solid cancers and suggested these methylation aberrations in the CpG islands associated with PCDH genes as powerful diagnostic biomarkers.


Cadherins/genetics , DNA Methylation , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , CpG Islands , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Multigene Family , Promoter Regions, Genetic , Sequence Analysis, DNA
5.
Cancers (Basel) ; 11(4)2019 Apr 12.
Article En | MEDLINE | ID: mdl-31013784

A link between mucolipin channels and tumors has been recently suggested. Herein, we aim to investigate the transient receptor potential mucolipin (TRPML)-1 relevance in glioblastoma. The expression of this channel was evaluated via qRT-PCR and immunohistochemistry in biopsies from 66 glioblastoma patients and two human glioblastoma cell lines and compared to normal human brain, astrocytes, and epileptic tissues. The subcellular distribution of TRPML-1 was examined via confocal microscopy in the glioma cell lines. Then, to assess the role of TRPML-1, cell viability assays have been conducted in T98 and U251 cell lines treated with the specific TRPML-1 agonist, MK6-83. We found that MK6-83 reduced cell viability and induced caspase-3-dependent apoptosis. Indeed, the TRPML-1 silencing or the blockage of TRPML-1 dependent [Ca2+]i release abrogated these effects. In addition, exposure of glioma cells to the reactive oxygen species (ROS) inducer, carbonyl cyanide m-chlorophenylhydrazone (CCCP), stimulated a TRPML-1-dependent autophagic cell death, as demonstrated by the ability of the autophagic inhibitor bafilomycin A, the TRPML-1 inhibitor sphingomyelin, and the TRPML-1 silencing to completely inhibit the CCCP-mediated effects. To test a possible correlation with patient's survival, Kaplan-Meier, univariate, and multivariate analysis have been performed. Data showed that the loss/reduction of TRPML-1 mRNA expression strongly correlates with short survival in glioblastoma (GBM) patients, suggesting that the reduction of TRPML-1 expression represents a negative prognostic factor in GBM patients.

7.
Oncotarget ; 9(17): 13807-13821, 2018 Mar 02.
Article En | MEDLINE | ID: mdl-29568396

Pilocytic astrocytoma (PA) is the most common glioma in pediatric patients and occurs in different locations. Chromosomal alterations are mostly located at chromosome 7q34 comprising the BRAF oncogene with consequent activation of the mitogen-activated protein kinase pathway. Although genetic and epigenetic alterations characterizing PA from different localizations have been reported, the role of epigenetic alterations in PA development is still not clear. The aim of this study was to investigate whether distinctive methylation patterns may define biologically relevant groups of PAs. Integrated DNA methylation analysis was performed on 20 PAs and 4 normal brain samples by Illumina Infinium HumanMethylation27 BeadChips. We identified distinct methylation profiles characterizing PAs from different locations (infratentorial vs supratentorial) and tumors with onset before and after 3 years of age. These results suggest that PA may be related to the specific brain site where the tumor arises from region-specific cells of origin. We identified and validated in silico the methylation alterations of some CpG islands. Furthermore, we evaluated the expression levels of selected differentially methylated genes and identified two biomarkers, one, IRX2, related to the tumor localization and the other, TOX2, as tumoral biomarker.

8.
Int J Radiat Oncol Biol Phys ; 91(1): 109-15, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25442339

PURPOSE: To evaluate 2 specific radiation schedules, each combined with temozolomide (TMZ), assessing their efficacy and safety in patients aged ≥65 years with newly diagnosed glioblastoma (GBM). METHODS AND MATERIALS: Patients aged ≥65 years with Karnofsky performance status (KPS) ≥60 who received either standard (60 Gy) or short-course (40 Gy) radiation therapy (RT) with concomitant and adjuvant TMZ between June 2004 and October 2013 were retrospectively analyzed. A propensity score analysis was executed for a balanced comparison of treatment outcomes. RESULTS: A total of 127 patients received standard RT-TMZ, whereas 116 patients underwent short-course RT-TMZ. Median overall survival and progression-free survival times were similar: 12 months and 5.6 months for the standard RT-TMZ group and 12.5 months and 6.7 months for the short-course RT-TMZ group, respectively. Radiation schedule was associated with similar survival outcomes in either unadjusted or adjusted analysis. O(6)-methylguanine-DNA methyltransferase promoter methylation was the most favorable prognostic factor (P=.0001). Standard RT-TMZ therapy was associated with a significant rise in grade 2 and 3 neurologic toxicity (P=.01), lowering of KPS scores during the study (P=.01), and higher posttreatment dosing of corticosteroid (P=.02). CONCLUSIONS: In older adults with GBM, survival outcomes of standard and short-course RT-TMZ were similar. An abbreviated course of RT plus TMZ may represent a reasonable therapeutic approach for these patients, without loss of survival benefit and acceptable toxicity.


Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Adrenal Cortex Hormones/therapeutic use , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Chemoradiotherapy/adverse effects , Chemotherapy, Adjuvant/adverse effects , Chemotherapy, Adjuvant/methods , DNA Methylation , Dacarbazine/therapeutic use , Disease-Free Survival , Drug Administration Schedule , Female , Glioblastoma/mortality , Humans , Karnofsky Performance Status , Male , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Propensity Score , Radiotherapy Dosage , Regression Analysis , Retrospective Studies , Temozolomide , Time Factors , Treatment Outcome
9.
J Neurooncol ; 118(2): 377-383, 2014 Jun.
Article En | MEDLINE | ID: mdl-24748470

Several molecular markers have been proposed as predictors of outcome in patients with high grade gliomas. We report a retrospective multicenter study of 97 consecutive adult patients with anaplastic astrocytoma (AA) treated with radiation therapy (RT) plus concomitant and adjuvant temozolomide (TMZ) between October 2004 and March 2012. Correlations between the isocitrate dehydrogenase 1 (IDH1) mutation and O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation with survival outcomes have been analyzed. At a median follow-up time of 46 months (range 12-89 months), median and 5-year overall survival rates were 50.5 months (95 % CI, 37.8-63.2) and 38% (95 % CI, 25.7-50.7%), and median and 5-year progression-free survival rates were 36 months (95% CI, 28.5-44.0) and 22 % (95 % CI, 10-34%), respectively. IDH1 mutation and MGMT promoter methylation were present in 54 and 60% of evaluable patients, respectively. Multivariate Cox proportional hazards regression analysis showed that IDH1 mutation (P = 0.001), MGMT methylation (P = 0.01), age < 50 years (P = 0.02), and extent of resection (P = 0.04) were significantly associated with longer survival. Our study confirms the favorable prognostic value of IDH1 mutation and MGMT methylation in patients with AA treated with RT plus concomitant and adjuvant TMZ. The superiority of combined radiochemotherapy over other treatment modalities remains to be demonstrated.


Astrocytoma/therapy , Brain Neoplasms/therapy , Chemoradiotherapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Isocitrate Dehydrogenase/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Astrocytoma/genetics , Biomarkers , Brain Neoplasms/genetics , DNA Methylation , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mutation , Prognosis , Promoter Regions, Genetic , Prospective Studies , Retrospective Studies , Temozolomide , Young Adult
10.
J Neurooncol ; 116(2): 275-82, 2014 Jan.
Article En | MEDLINE | ID: mdl-24162810

Combination of procarbazine, lomustine and vincristine (PCV) with radiation therapy (RT) has been associated with longer survival in patients with anaplastic oligodendroglioma (AO) and anaplastic oligoastrocytoma (AOA), especially in those with chromosome 1p/19q codeletion. We report a multicenter retrospective study of 84 consecutive adult patients with AO and AOA treated with RT plus concomitant and adjuvant temozolomide (TMZ) between February 2004 and January 2011. Correlations between chromosome 1p/19q codeletion, isocitrate dehydrogenase1 (IDH1) mutation, and O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation with survival outcomes have been analyzed. For all 84 patients the median overall survival (OS) and progression-free survival rates were 55.6 and 45.2 months, respectively. Grade 3 or 4 hematological toxicity occurred in 17 % of patients. Chromosome 1p/19q codeletion was detected in 57 %, IDH1 mutation in 63 %, and MGMT promoter methylation in 74 % of evaluable patients. In multivariate analysis the presence of chromosome 1p/19q codeletion was associated with significant survival benefit (median OS 34 months in noncodeleted tumors and not reached in codeleted tumors; HR 0.16, 95 % CI 0.03-0.45; P = 0.005). IDH1 mutation was also of prognostic significance for longer survival (P = 0.001; HR 0.20, 95 % 0.06-0.41), whereas MGMT promoter methylation was only of borderline significance. The study indicates that RT with concomitant and adjuvant TMZ is a relatively safe treatment associated with longer survival in patients with 1p/19q codeleted and IDH1 mutated tumors. Results from ongoing randomized studies will be essential to clarify if RT plus TMZ may provide survival as good as or better than RT combined with PCV for patients with AO and AOA.


Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Loss of Heterozygosity/genetics , Oligodendroglioma/drug therapy , Oligodendroglioma/radiotherapy , Treatment Outcome , Adult , Aged , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 9/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Disease-Free Survival , Female , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Proportional Hazards Models , Retrospective Studies , Tumor Suppressor Proteins/genetics , Young Adult
11.
Neuro Oncol ; 16(2): 228-40, 2014 Jan.
Article En | MEDLINE | ID: mdl-24305714

BACKGROUND: High-grade gliomas (HGGs) account for 15% of all pediatric brain tumors and are a leading cause of cancer-related mortality and morbidity. Pediatric HGGs (pHGGs) are histologically indistinguishable from their counterpart in adulthood. However, recent investigations indicate that differences occur at the molecular level, thus suggesting that the molecular path to gliomagenesis in childhood is distinct from that of adults. MicroRNAs (miRNAs) have been identified as key molecules in gene expression regulation, both in development and in cancer. miRNAs have been investigated in adult high-grade gliomas (aHGGs), but scant information is available for pHGGs. METHODS: We explored the differences in microRNAs between pHGG and aHGG, in both fresh-frozen and paraffin-embedded tissue, by high-throughput miRNA profiling. We also evaluated the biological effects of miR-17-92 cluster silencing on a pHGG cell line. RESULTS: Comparison of miRNA expression patterns in formalin versus frozen specimens resulted in high correlation between both types of samples. The analysis of miRNA profiling revealed a specific microRNA pattern in pHGG with an overexpression and a proliferative role of the miR-17-92 cluster. Moreover, we highlighted a possible quenching function of miR-17-92 cluster on its target gene PTEN, together with an activation of tumorigenic signaling such as sonic hedgehog in pHGG. CONCLUSIONS: Our results suggest that microRNA profiling represents a tool to distinguishing pediatric from adult HGG and that miR-17-92 cluster sustains pHGG.


Brain Neoplasms/genetics , Gene Expression Profiling , Glioma/genetics , MicroRNAs/genetics , Adolescent , Adult , Aged , Blotting, Western , Brain/metabolism , Brain/pathology , Brain Neoplasms/pathology , Case-Control Studies , Cell Proliferation , Child , Child, Preschool , Female , Follow-Up Studies , Glioma/pathology , Humans , In Situ Hybridization , Male , Middle Aged , Neoplasm Grading , Prognosis , RNA, Long Noncoding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
12.
Int J Radiat Oncol Biol Phys ; 83(1): 93-9, 2012 May 01.
Article En | MEDLINE | ID: mdl-22079725

PURPOSE: Radiotherapy (RT) and chemotherapy may prolong survival in older patients (age ≥70 years) with glioblastoma multiforme (GBM), although the survival benefits remain poor. This Phase II multicenter study was designed to evaluate the efficacy and safety of an abbreviated course of RT plus concomitant and adjuvant temozolomide (TMZ) in older patients with GBM. PATIENTS AND METHODS: Seventy-one eligible patients 70 years of age or older with newly diagnosed GBM and a Karnofsky performance status ≥60 were treated with a short course of RT (40 Gy in 15 fractions over 3 weeks) plus TMZ at the dosage of 75 mg/m(2) per day followed by 12 cycles of adjuvant TMZ (150-200 mg/m(2) for 5 days during each 28-day cycle). The primary endpoint was overall survival (OS). Secondary endpoints included progression-free survival and toxicity. RESULTS: The Median OS was 12.4 months, and the 1-year and 2-year OS rates were 58% and 20%, respectively. The median and 1-year rates of progression-free survival were 6 months and 20%, respectively. All patients completed the planned programme of RT. Grade 3 or 4 adverse events occurred in 16 patients (22%). Grade 3 and 4 neutropenia and/or thrombocytopenia occurred in 10 patients (15%), leading to the interruption of treatment in 6 patients (8%). Nonhematologic Grade 3 toxicity was rare, and included fatigue in 4 patients and cognitive disability in 1 patient. CONCLUSIONS: A combination of an abbreviated course of RT plus concomitant and adjuvant TMZ is well tolerated and may prolong survival in elderly patients with GBM. Future randomized studies need to evaluate the efficacy and toxicity of different schedules of RT in association with chemotherapy.


Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/mortality , Chemoradiotherapy/adverse effects , Chemotherapy, Adjuvant , Dacarbazine/administration & dosage , Dacarbazine/therapeutic use , Disease-Free Survival , Dose Fractionation, Radiation , Drug Administration Schedule , Female , Glioblastoma/mortality , Humans , Karnofsky Performance Status , Male , Neutropenia/etiology , Temozolomide , Thrombocytopenia/etiology
13.
Brain Pathol ; 21(5): 558-63, 2011 Sep.
Article En | MEDLINE | ID: mdl-21261775

Recent gene expression microarray analyses have indicated that claudin-6 is specifically expressed in atypical teratoid rhabdoid tumors (AT/RTs), suggesting a role as a positive diagnostic marker in addition to SMARCB1 (INI1) loss, which is encountered in the majority of AT/RTs. In order to investigate the potential of claudin-6 as a diagnostic marker, expression was investigated in 59 AT/RTs and 60 other primary central nervous system (CNS) tumors, including primitive neuroectodermal tumors, medulloblastomas, choroid plexus tumors, and both pediatric and adult low- and high-grade gliomas using immunohistochemistry. Claudin-6 was expressed in 17/59 AT/RTs (29%), but also in a variety of other primary CNS tumors, including 60% of medulloblastomas and 21% of malignant gliomas. Even though high staining scores (2+ or 3+) were more often encountered in AT/RTs (Chi-square 4.177; P=0.041), the overall frequency of claudin-6 staining was not significantly higher in AT/RTs as compared with the other tumors (17/59 vs. 16/60; Chi-square=0.328; P=0.567). In a subgroup of 43 AT/RT patients, of which follow-up data were available, claudin-6 expression did not show any correlation with survival. In conclusion, claudin-6 immunohistochemistry is of limited sensitivity and specificity for the diagnosis of AT/RT and does not correlate with clinical behavior.


Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Claudins/metabolism , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/metabolism , Austria , Brain Neoplasms/classification , Brain Neoplasms/mortality , Chi-Square Distribution , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Female , Follow-Up Studies , Germany , Humans , Kaplan-Meier Estimate , Male , Pediatrics , Rhabdoid Tumor/classification , Rhabdoid Tumor/mortality , SMARCB1 Protein , Sensitivity and Specificity , Transcription Factors/metabolism
14.
Radiother Oncol ; 97(3): 377-81, 2010 Dec.
Article En | MEDLINE | ID: mdl-20855119

PURPOSE: To analyze the recurrence patterns in patients with newly diagnosed glioblastoma (GBM) treated with conformal radiotherapy (RT) plus concomitant and adjuvant temozolomide (TMZ), and to compare the patterns of failure according to different target volume delineations. METHODS AND MATERIALS: One hundred and five patients with GBM which recurred after three-dimensional (3D) conformal RT plus TMZ were evaluated. The clinical target volume (CTV) used for our treatment planning (S'Andrea plans) consisted of residual tumor and resection cavity plus 2-cm margins according to recent randomized trials of the European Organisation for Research and Treatment of Cancer (EORTC). MRI scans showing tumor recurrences were fused with the planning computed tomography (CT), and the patterns of failure were analyzed dosimetrically using dose-volume histograms. For each patient a theoretical plan based on the addition of postoperative edema plus 2-cm margins according to current guidelines of Radiation Therapy Oncology Group (RTOG) was created and patterns of failure were evaluated. RESULTS: The median overall survival and progression-free survival were 14.2 months and 7.5 months, respectively. Recurrences were central in 79 patients, in-field in 6 patients, marginal in 6 patients, and distant in 14 patients. Analysis of O(6)-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status showed different recurrence patterns of GBMs in patients with MGMT methylated compared with patients with MGMT unmethylated status. Recurrences occurred central/in-field and outside in 64% and 31% of methylated patients, and in 91% and 5.4% of unmethylated patients, respectively (P=0.01). Patterns of failure were similar between the different treatment plans, however the median volume percent of brain irradiated to high doses was significantly smaller for our plans than for RTOG plans (P=0.0001). CONCLUSION: Most of patients treated with RT plus concomitant and adjuvant RT have central recurrences, however distant new lesions may occur in more than 10% of patients. The use of target delineation using postoperative residual tumor and cavity plus 2-cm margins is associated with smaller volumes of normal brain irradiated to high doses as compared with plans including expanded edema, without a significant increase of the risk of marginal recurrences. Future clinical randomized studies need to compare the different planning methods in terms of efficacy and risk of late radiation-induced toxicity.


Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Radiotherapy, Conformal , Adult , Aged , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Chemotherapy, Adjuvant , Combined Modality Therapy , Dacarbazine/therapeutic use , Disease-Free Survival , Female , Glioblastoma/diagnosis , Glioblastoma/mortality , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Survival Rate , Temozolomide , Tomography, X-Ray Computed
15.
Childs Nerv Syst ; 26(8): 1051-6, 2010 Aug.
Article En | MEDLINE | ID: mdl-20552207

INTRODUCTION: In this study, we investigated the prognostic and predictive value of MGMT promoter methylation and protein expression in 30 pediatric high grade gliomas (pHGG). METHODS: MGMT promoter methylation was assayed by methylation-specific polymerase chain reaction (MSP), whereas MGMT protein expression was evaluated by immunohistochemistry (IHC). RESULTS: MGMT promoter methylation was observed in 7/24 (30%) cases, whereas MGMT protein overexpression was found in 19/28 (68%) cases with similar distribution in grade III and grade IV gliomas. Median survival of methylated and unmethylated patients was 16 and 8 months, respectively. Moreover, overall survival and progression-free survival showed a trend toward reduction in patients with unmethylation (p = 0.9 and p = 0.7, respectively). For MGMT protein expression, the median survival was 8.5 and 17 months for patients with MGMT overexpression or low expression, respectively. Although these two groups did not show statistically significant differences in terms of overall survival or progression-free survival (p = 0.8 and p = 0.7, respectively), there was a significant correlation between MGMT protein expression and MGMT promoter methylation status (p = 0.01). CONCLUSIONS: Our findings indicate that, in pHGG, (a) MGMT promoter methylation is less frequent than in adult malignant gliomas, (b) there is a high correlation between MGMT MSP and MGMT IHC, and (c) as in adults, MGMT status is associated with prognosis, although this observation has to be statistically validated on larger series of patients.


Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioma/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Biomarkers, Tumor/analysis , Brain Neoplasms/enzymology , Brain Neoplasms/mortality , Child , Child, Preschool , DNA Methylation/genetics , DNA Modification Methylases/biosynthesis , DNA Repair Enzymes/biosynthesis , Disease-Free Survival , Female , Glioma/enzymology , Glioma/mortality , Humans , Immunohistochemistry , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/biosynthesis
16.
Carcinogenesis ; 31(5): 794-803, 2010 May.
Article En | MEDLINE | ID: mdl-20093382

The aim of this study was to investigate the expression and function of the transient receptor potential vanilloid 2 (TRPV2) in human glioma cells. By Real-Time-PCR and western blot analysis, we found that TRPV2 messenger RNA (mRNA) and protein were expressed in benign astrocyte tissues, and its expression progressively declined in high-grade glioma tissues as histological grade increased (n = 49 cases), and in U87MG cells and in MZC, FCL and FSL primary glioma cells. To investigate the function of TRPV2 in glioma, small RNA interfering was used to silence TRPV2 expression in U87MG cells. As evaluated by RT-Profiler PCR array, siTRPV2-U87MG transfected cells displayed a marked downregulation of Fas and procaspase-8 mRNA expression, associated with upregulation of cyclin E1, cyclin-dependent kinase 2, E2F1 transcriptor factor 1, V-raf-1 murine leukemia viral oncogene homolog 1 and Bcl-2-associated X protein (Bcl-X(L)) mRNA expression. TRPV2 silencing increased U87MG cell proliferation as shown by the increased percentage of cells incorporating 5-bromo-2-deoxyuridine expressing beta(III)-tubulin and rescued glioma cells to Fas-induced apoptosis. These events were dependent on extracellular signal-regulated kinase (ERK) activation: indeed inhibition of ERK activation in siTRPV2-U87MG transfected cells by treatment with PD98059, a specific mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibitor, reduced Bcl-X(L) protein levels, promoted Fas expression, and restored Akt/protein kinase B pathway activation leading to reduced U87MG cell survival and proliferation, and increased sensitivity to Fas-induced apoptosis. In addition, transfection of TRPV2 in MZC glioma cells, by inducing Fas overexpression, resulted in a reduced viability and an increased spontaneous and Fas-induced apoptosis. Overall, our findings indicate that TRPV2 negatively controls glioma cell survival and proliferation, as well as resistance to Fas-induced apoptotic cell death in an ERK-dependent manner.


Apoptosis , Extracellular Signal-Regulated MAP Kinases/physiology , Glioma/pathology , TRPV Cation Channels/physiology , fas Receptor/physiology , Cell Line, Tumor , Cell Proliferation , Flavonoids/pharmacology , Glioma/drug therapy , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/analysis , TRPV Cation Channels/analysis , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , bcl-X Protein/analysis
17.
Hum Pathol ; 40(12): 1738-46, 2009 Dec.
Article En | MEDLINE | ID: mdl-19716156

Caveolin-1 (cav-1) has been proposed as an immunohistochemical marker able to distinguish astroglial from oligodendroglial tumors. In addition, it has been suggested that the reduction of caveolin-1 expression in glioblastoma cells increases their proliferative and invasive potential. Accordingly, the present study investigates caveolin-1 immunoexpression and correlation with the 1p/19q status, histologic grade, proliferation index, epidermal growth factor receptor, and p53 expression in a series of 73 diffuse gliomas. A membranous and cytoplasmic immunolabeling for caveolin-1 was detected in neoplastic cells of 60% of cases. No significant differences in terms of caveolin-1 expression were observed between astrocytomas, oligodendrogliomas, and oligoastrocytomas. In addition, caveolin-1 expression was not correlated with 1p/19q status in oligodendrogliomas and mixed oligoastrocytomas. Caveolin-1 was expressed in most high-grade (World Health Organization III and IV) gliomas. Low caveolin-1 expression correlated with a higher Ki-67 labeling index and the absence of p53 overexpression in glioblastomas, and it was significantly associated with epidermal growth factor receptor overexpression in anaplastic astrocytomas. In conclusion, the present study indicates that caveolin-1 is not useful as diagnostic marker to differentiate grade II astrocytomas from oligodendrogliomas.


Brain Neoplasms/diagnosis , Caveolin 1/biosynthesis , ErbB Receptors/biosynthesis , Glioma/diagnosis , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Biomarkers, Tumor/analysis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Diagnosis, Differential , Glioma/genetics , Glioma/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Ki-67 Antigen/biosynthesis , Middle Aged
18.
Proc Natl Acad Sci U S A ; 105(39): 15118-23, 2008 Sep 30.
Article En | MEDLINE | ID: mdl-18809912

We examined how the endogenous anticonvulsant adenosine might influence gamma-aminobutyric acid type A (GABA(A)) receptor stability and which adenosine receptors (ARs) were involved. Upon repetitive activation (GABA 500 microM), GABA(A) receptors, microtransplanted into Xenopus oocytes from neurosurgically resected epileptic human nervous tissues, exhibited an obvious GABA(A)-current (I(GABA)) run-down, which was consistently and significantly reduced by treatment with the nonselective adenosine receptor antagonist CGS15943 (100 nM) or with adenosine deaminase (ADA) (1 units/ml), that inactivates adenosine. It was also found that selective antagonists of A2B (MRS1706, 10 nM) or A3 (MRS1334, 30 nM) receptors reduced I(GABA) run-down, whereas treatment with the specific A1 receptor antagonist DPCPX (10 nM) was ineffective. The selective A2A receptor antagonist SCH58261 (10 nM) reduced or potentiated I(GABA) run-down in approximately 40% and approximately 20% of tested oocytes, respectively. The ADA-resistant, AR agonist 2-chloroadenosine (2-CA) (10 microM) potentiated I(GABA) run-down but only in approximately 20% of tested oocytes. CGS15943 administration again decreased I(GABA) run-down in patch-clamped neurons from either human or rat neocortex slices. I(GABA) run-down in pyramidal neurons was equivalent in A1 receptor-deficient and wt neurons but much larger in neurons from A2A receptor-deficient mice, indicating that, in mouse cortex, GABA(A)-receptor stability is tonically influenced by A2A but not by A1 receptors. I(GABA) run-down from wt mice was not affected by 2-CA, suggesting maximal ARs activity by endogenous adenosine. Our findings strongly suggest that cortical A2-A3 receptors alter the stability of GABA(A) receptors, which could offer therapeutic opportunities.


Adenosine/metabolism , Anticonvulsants/metabolism , Epilepsy/metabolism , Purinergic P1 Receptor Antagonists , Receptors, GABA-A/metabolism , Adenosine/pharmacology , Adenosine Deaminase/pharmacology , Adult , Animals , Anticonvulsants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , GABA-A Receptor Agonists , Humans , Male , Neurons/metabolism , Oocytes , Pyramidal Tracts/metabolism , Pyrimidines/pharmacology , Quinazolines/pharmacology , Rats , Triazoles/pharmacology , Xanthines/pharmacology , Xenopus , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
19.
Ultrastruct Pathol ; 31(1): 33-44, 2007.
Article En | MEDLINE | ID: mdl-17455096

Gene mutations impairing the functions of the WNT signaling transduction pathway have been found in approximately 15% of human sporadic medulloblastomas. To understand the functional role of the WNT pathway in medulloblastoma, we have investigated the intracellular distribution of beta-catenin in a series of 17 human medulloblastomas to correlate such expression with neuronal differentiation and in cultured cell models following functional silencing of the APC gene by small-interference RNA (siRNA). Transient siRNA transfection resulted in a 50% reduction of the APC gene product levels in both DAOY and D283MED cell lines. In the former, less-differentiated cell line, beta-catenin levels remained unchanged or were slightly reduced, but beta-catenin translocated in the nucleus following APC gene siRNA silencing. In contrast, in the more differentiated D283MED cells, beta-catenin levels increased about twofold while mainly maintaining the cytoplasmic and cell membrane localization. Cytoplasmic/nuclear localization of beta-catenin was present in 12 of 17 cases of medulloblastoma with a prevalent distribution in the classic, 6/7 cases, and large cell/anaplastic variant, 4/4 cases. The nodular/desmoplastic lesions showed strongly positivity in the cell membrane mainly of intranodular cells with advanced neuronal differentiation. These observations support an important functional role of WNT/beta-catenin pathway in neuronal differentiation in medulloblastoma.


Cell Differentiation , Medulloblastoma/metabolism , Neurons/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Genes, APC , Humans , Immunohistochemistry , Medulloblastoma/genetics , Medulloblastoma/ultrastructure , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Neurons/ultrastructure , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Wnt Proteins/metabolism
20.
Acta Neuropathol ; 109(2): 231-4, 2005 Feb.
Article En | MEDLINE | ID: mdl-15549332

We describe a case of ependymoma with neuronal differentiation in form of neuropil-like islands. A 6-year-old boy presented at clinical examination for a short history of headaches and vomiting. Brain computed tomography showed a large, partially cystic, parieto-occipital lesion. The tumor was composed by glial fibrillary acidic protein-positive round cells with a perivascular arrangement and scattered neuropil-like islands, showing intense positivity for synaptophysin. Despite radiotherapy, the tumor recurred, showing frank features of anaplasia, but lacking the neuropil-like islands. The histological features of the tumor are discussed in the light of the concept that neuronal differentiation can occur occasionally in gliomas of different lineage without affecting the expected biological behavior.


Brain Neoplasms/diagnosis , Ependymoma/diagnosis , Neuropil/pathology , Antibodies, Antinuclear/metabolism , Antibodies, Monoclonal/metabolism , Brain Neoplasms/metabolism , Child , Ependymoma/metabolism , Ependymoma/pathology , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry/methods , Male , Microtubule-Associated Proteins/metabolism , Mucin-1/metabolism , Neurofilament Proteins/metabolism , Neuropil/metabolism , Synaptophysin/metabolism
...