Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 19(8): 2216-23, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444211

ABSTRACT

PURPOSE: The diagnosis of leptomeningeal metastases is usually confirmed by the finding of malignant cells by cytologic examination in the cerebrospinal fluid (CSF). More sensitive and specific cancer biomarkers may improve the detection of tumor cells in the CSF. Promoter methylation of the human telomerase reverse transcriptase (hTERT) gene characterizes most cancer cells. The aim of this study was to develop a sensitive method to detect hTERT methylation and to explore its use as a cancer biomarker in CSF. EXPERIMENTAL DESIGN: In 77 CSF specimens from 67 patients, hTERT promoter methylation was evaluated using real-time methylation-sensitive high-resolution melting (MS-HRM) and real-time TaqMan PCR and MS-HRM in a single-tube assay. RESULTS: Real-time MS-HRM assay was able to detect down to 1% hTERT-methylated DNA in a background of unmethylated DNA. PCR products were obtained from 90% (69/77) of CSF samples. No false positive hTERT was detected in the 21 non-neoplastic control cases, given to the method a specificity of 100%. The sensitivity of the real-time MS-HRM compared with the cytologic gold standard analysis was of 92% (11/12). Twenty-six CSFs from 22 patients with an hTERT-methylated primary tumor showed cytologic results suspicious for malignancy; in 17 (65%) of them, a diagnosis of leptomeningeal metastases could be confirmed by the hTERT methylation test. CONCLUSION: The hTERT real-time MS-HRM approach is fast, specific, sensitive, and could therefore become a valuable tool for diagnosis of leptomeningeal metastases as an adjunct to the traditional examination of CSF.


Subject(s)
DNA Methylation , Neoplasms/genetics , Promoter Regions, Genetic/genetics , Telomerase/genetics , Biomarkers, Tumor/cerebrospinal fluid , Biomarkers, Tumor/genetics , DNA, Neoplasm/cerebrospinal fluid , DNA, Neoplasm/genetics , Female , Humans , Male , Meningeal Neoplasms/diagnosis , Meningeal Neoplasms/genetics , Meningeal Neoplasms/secondary , Neoplasms/enzymology , Neoplasms/pathology , Real-Time Polymerase Chain Reaction/methods , Reproducibility of Results , Sensitivity and Specificity
2.
Ophthalmic Genet ; 33(3): 155-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21526971

ABSTRACT

PURPOSE: To report the lethal course of malignant transformation of retinoma in an adult. METHODS: Case report. A 40-year-old patient presented with retinoma in his right eye and retinoblastoma in his left eye. Enucleation was recommended but refused and the patient received whole eye radiotherapy elsewhere. Five years later he presented again, with temporal hemianopsia of the left eye secondary to chiasmatic invasion. RESULTS: Diagnosis of retinoblastoma infiltration was confirmed by stereotactic biopsy of the chiasmatic lesion. Treatment with intravenous and intrathecal chemotherapy led to partial remission, and was followed by stereotactic irradiation of the chiasmatic mass and right optic nerve. The left eye was enucleated. Death occurred one year later due to cerebrospinal fluid metastases. CONCLUSION: Extraocular extension of retinoblastoma diagnosed in adulthood has never, to our knowledge, been reported. This case stresses the importance of lifelong retinoma monitoring and the necessity for radical treatment in the event of malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/pathology , Optic Chiasm/pathology , Optic Nerve Neoplasms/pathology , Precancerous Conditions/pathology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Adult , Eye Enucleation , Humans , Magnetic Resonance Imaging , Male , Neoplasm Invasiveness/pathology , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Retinoblastoma Protein/genetics , Tomography, X-Ray Computed
3.
J Neurooncol ; 107(1): 29-36, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21979892

ABSTRACT

Brain tumors, benign or malignant, are characterized by a very high degree of vascularization. Recent accumulating evidence suggests that during development the neuronal wiring follows the same routes as the vasculature and that these two systems may share some of the same factors for guidance. Thus, expression of dual angiogenic/neurogenic growth factors was evaluated by in situ hybridization in human primary brain tumors of three different types, i.e., astrocytomas, oligodendrogliomas, and ependymomas, of increasing grades, in relation with the grade and type of the tumor. For this evaluation we selected vascular endothelial growth factor (VEGF-A) and its receptors VEGF-R1 and VEGF-R2 and the neuropilins 1 and 2 (NRP-1 and NRP-2), which have proangiogenic properties, platelet-derived growth factor (PDGF) receptor-beta (PDGF-Rß), which is required for the functional maturation of blood vessels, the ephrins and their Eph receptors, angiotensinogen (AGT) and thrombospondin-2 (TSP-2), which have potential antiangiogenic properties, and netrin-1 (Net-1), which regulates vascular architecture. We show that the expression of the VEGF-NRP system, PDGF-Rß, TSP-2, AGT, and Net-1 are differentially regulated, either increased or decreased, in relation with the type and grade of the tumor, whereas regulation of the ephrinB system does not seem to be relevant in these human brain tumors.


Subject(s)
Angiogenic Proteins/genetics , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/genetics , RNA, Messenger/genetics , Angiotensinogen/genetics , Brain Neoplasms/pathology , Humans , In Situ Hybridization , Neoplasm Grading , Neuropilin-1/genetics , Neuropilin-2/genetics , Oncogene Proteins/genetics , RNA Probes , Receptor, Platelet-Derived Growth Factor beta/genetics , Retrospective Studies , Thrombospondins/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Lancet Oncol ; 10(5): 459-66, 2009 May.
Article in English | MEDLINE | ID: mdl-19269895

ABSTRACT

BACKGROUND: In 2004, a randomised phase III trial by the European Organisation for Research and Treatment of Cancer (EORTC) and National Cancer Institute of Canada Clinical Trials Group (NCIC) reported improved median and 2-year survival for patients with glioblastoma treated with concomitant and adjuvant temozolomide and radiotherapy. We report the final results with a median follow-up of more than 5 years. METHODS: Adult patients with newly diagnosed glioblastoma were randomly assigned to receive either standard radiotherapy or identical radiotherapy with concomitant temozolomide followed by up to six cycles of adjuvant temozolomide. The methylation status of the methyl-guanine methyl transferase gene, MGMT, was determined retrospectively from the tumour tissue of 206 patients. The primary endpoint was overall survival. Analyses were by intention to treat. This trial is registered with Clinicaltrials.gov, number NCT00006353. FINDINGS: Between Aug 17, 2000, and March 22, 2002, 573 patients were assigned to treatment. 278 (97%) of 286 patients in the radiotherapy alone group and 254 (89%) of 287 in the combined-treatment group died during 5 years of follow-up. Overall survival was 27.2% (95% CI 22.2-32.5) at 2 years, 16.0% (12.0-20.6) at 3 years, 12.1% (8.5-16.4) at 4 years, and 9.8% (6.4-14.0) at 5 years with temozolomide, versus 10.9% (7.6-14.8), 4.4% (2.4-7.2), 3.0% (1.4-5.7), and 1.9% (0.6-4.4) with radiotherapy alone (hazard ratio 0.6, 95% CI 0.5-0.7; p<0.0001). A benefit of combined therapy was recorded in all clinical prognostic subgroups, including patients aged 60-70 years. Methylation of the MGMT promoter was the strongest predictor for outcome and benefit from temozolomide chemotherapy. INTERPRETATION: Benefits of adjuvant temozolomide with radiotherapy lasted throughout 5 years of follow-up. A few patients in favourable prognostic categories survive longer than 5 years. MGMT methylation status identifies patients most likely to benefit from the addition of temozolomide. FUNDING: EORTC, NCIC, Nélia and Amadeo Barletta Foundation, Schering-Plough.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Combined Modality Therapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/therapeutic use , Disease Progression , Female , Follow-Up Studies , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Survival Analysis , Survival Rate , Temozolomide , Tumor Suppressor Proteins/genetics
5.
Bioorg Med Chem ; 16(15): 7337-46, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18599296

ABSTRACT

Refining the chemical structure of functionalized pyrrolidine-based inhibitors of Golgi alpha-mannosidase II (GMII) to optimize binding affinity provided a lead molecule that demonstrated nanomolar competitive inhibition of alpha-mannosidases II and an optimal fit in the active site of Drosophila GMII by X-ray crystallography. Esters of this lead compound also inhibited the growth of human glioblastoma and brain-derived endothelial cells more than the growth of non-tumoral human fibroblasts, suggesting their potential for anti-cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , alpha-Mannosidase/antagonists & inhibitors , alpha-Mannosidase/classification , Animals , Binding Sites , Cell Line, Tumor , Drosophila/enzymology , Endothelial Cells/drug effects , Fabaceae/enzymology , Glioblastoma , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
6.
Cancer Invest ; 26(6): 597-609, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18584351

ABSTRACT

The DNA repair and detoxifying enzymes, O(6)-methylguanine-DNA-methyltransferase (MGMT) and glutathione-S-transferase (GST), may be responsible fpr poor response to alkylating agents in glioblastoma treatment. The methylation of MGMT promoter and the expression of MGMT and GST were highly heterogeneous in surgical specimens of human glioblastoma and in established human glioblastoma cells under 2-D and 3-D culture conditions, suggesting an intrinsic property of these cells. MGMT and GST expression did not predict the sensitivity of glioblastoma cells to alkylating agents. Combination of alkylating agents with inhibitors of GST disclosed additive effects, suggesting that inhibition of GST should be considered in glioblastoma therapy.


Subject(s)
Brain Neoplasms/enzymology , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma/enzymology , Glutathione Transferase/metabolism , Tumor Suppressor Proteins/metabolism , Alkylating Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carmustine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , DNA Replication/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ethacrynic Acid/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Glutathione S-Transferase pi/metabolism , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Melphalan/pharmacology , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics
7.
N Engl J Med ; 352(10): 987-96, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15758009

ABSTRACT

BACKGROUND: Glioblastoma, the most common primary brain tumor in adults, is usually rapidly fatal. The current standard of care for newly diagnosed glioblastoma is surgical resection to the extent feasible, followed by adjuvant radiotherapy. In this trial we compared radiotherapy alone with radiotherapy plus temozolomide, given concomitantly with and after radiotherapy, in terms of efficacy and safety. METHODS: Patients with newly diagnosed, histologically confirmed glioblastoma were randomly assigned to receive radiotherapy alone (fractionated focal irradiation in daily fractions of 2 Gy given 5 days per week for 6 weeks, for a total of 60 Gy) or radiotherapy plus continuous daily temozolomide (75 mg per square meter of body-surface area per day, 7 days per week from the first to the last day of radiotherapy), followed by six cycles of adjuvant temozolomide (150 to 200 mg per square meter for 5 days during each 28-day cycle). The primary end point was overall survival. RESULTS: A total of 573 patients from 85 centers underwent randomization. The median age was 56 years, and 84 percent of patients had undergone debulking surgery. At a median follow-up of 28 months, the median survival was 14.6 months with radiotherapy plus temozolomide and 12.1 months with radiotherapy alone. The unadjusted hazard ratio for death in the radiotherapy-plus-temozolomide group was 0.63 (95 percent confidence interval, 0.52 to 0.75; P<0.001 by the log-rank test). The two-year survival rate was 26.5 percent with radiotherapy plus temozolomide and 10.4 percent with radiotherapy alone. Concomitant treatment with radiotherapy plus temozolomide resulted in grade 3 or 4 hematologic toxic effects in 7 percent of patients. CONCLUSIONS: The addition of temozolomide to radiotherapy for newly diagnosed glioblastoma resulted in a clinically meaningful and statistically significant survival benefit with minimal additional toxicity.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Antineoplastic Agents, Alkylating/adverse effects , Brain Neoplasms/mortality , Chemotherapy, Adjuvant , Dacarbazine/adverse effects , Disease Progression , Female , Glioblastoma/mortality , Humans , Male , Middle Aged , Proportional Hazards Models , Radiotherapy, Computer-Assisted/adverse effects , Survival Analysis , Temozolomide
8.
N Engl J Med ; 352(10): 997-1003, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15758010

ABSTRACT

BACKGROUND: Epigenetic silencing of the MGMT (O6-methylguanine-DNA methyltransferase) DNA-repair gene by promoter methylation compromises DNA repair and has been associated with longer survival in patients with glioblastoma who receive alkylating agents. METHODS: We tested the relationship between MGMT silencing in the tumor and the survival of patients who were enrolled in a randomized trial comparing radiotherapy alone with radiotherapy combined with concomitant and adjuvant treatment with temozolomide. The methylation status of the MGMT promoter was determined by methylation-specific polymerase-chain-reaction analysis. RESULTS: The MGMT promoter was methylated in 45 percent of 206 assessable cases. Irrespective of treatment, MGMT promoter methylation was an independent favorable prognostic factor (P<0.001 by the log-rank test; hazard ratio, 0.45; 95 percent confidence interval, 0.32 to 0.61). Among patients whose tumor contained a methylated MGMT promoter, a survival benefit was observed in patients treated with temozolomide and radiotherapy; their median survival was 21.7 months (95 percent confidence interval, 17.4 to 30.4), as compared with 15.3 months (95 percent confidence interval, 13.0 to 20.9) among those who were assigned to only radiotherapy (P=0.007 by the log-rank test). In the absence of methylation of the MGMT promoter, there was a smaller and statistically insignificant difference in survival between the treatment groups. CONCLUSIONS: Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylated MGMT promoter did not have such a benefit.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/genetics , DNA Methylation , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Gene Silencing , Glioblastoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Disease-Free Survival , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/radiotherapy , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , Randomized Controlled Trials as Topic , Survival Analysis , Temozolomide
9.
Cancer Res ; 63(20): 6613-25, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14583454

ABSTRACT

The development of targeted treatment strategies adapted to individual patients requires identification of the different tumor classes according to their biology and prognosis. We focus here on the molecular aspects underlying these differences, in terms of sets of genes that control pathogenesis of the different subtypes of astrocytic glioma. By performing cDNA-array analysis of 53 patient biopsies, comprising low-grade astrocytoma, secondary glioblastoma (respective recurrent high-grade tumors), and newly diagnosed primary glioblastoma, we demonstrate that human gliomas can be differentiated according to their gene expression. We found that low-grade astrocytoma have the most specific and similar expression profiles, whereas primary glioblastoma exhibit much larger variation between tumors. Secondary glioblastoma display features of both other groups. We identified several sets of genes with relatively highly correlated expression within groups that: (a). can be associated with specific biological functions; and (b). effectively differentiate tumor class. One prominent gene cluster discriminating primary versus nonprimary glioblastoma comprises mostly genes involved in angiogenesis, including VEGF fms-related tyrosine kinase 1 but also IGFBP2, that has not yet been directly linked to angiogenesis. In situ hybridization demonstrating coexpression of IGFBP2 and VEGF in pseudopalisading cells surrounding tumor necrosis provided further evidence for a possible involvement of IGFBP2 in angiogenesis. The separating groups of genes were found by the unsupervised coupled two-way clustering method, and their classification power was validated by a supervised construction of a nearly perfect glioma classifier.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Neovascularization, Pathologic/genetics , Adolescent , Adult , Aged , Astrocytoma/blood supply , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Child, Preschool , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Insulin-Like Growth Factor Binding Protein 2/genetics , Male , Middle Aged , Multigene Family , Oligonucleotide Array Sequence Analysis , Reproducibility of Results
10.
Semin Oncol ; 30(6 Suppl 19): 23-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14765381

ABSTRACT

Low-grade gliomas are a heterogenous group of diseases characterized by relatively slow-growing primary brain tumors of astrocytic and/or oligodendroglial origin. Many patients present with easily controlled seizures and remain stable for years, whereas others progress rapidly to higher-grade tumors. Several studies have retrospectively investigated tumor-, patient-, and treatment-related prognostic factors in this patient population. Tumor histology, grade, location, contrast enhancement, and molecular markers have been identified as prognostic factors for survival. Likewise, patient age, performance status, and seizure history are patient-dependent prognostic factors. However, although patients who undergo surgical resection and receive adjuvant radiotherapy tend to have improved survival, treatment-dependent prognostic factors have yet to be definitively identified. Recursive partitioning and multivariant analyses have identified a class of patients with good prognosis. Younger patients with good performance status, non-contrast-enhancing tumors (<5 cm), and tumors of oligodendroglial or mixed-oligoastrocytic subtype have improved survival. The European Organisation for Research and Treatment of Cancer has recently developed a prognostic score based on identified prognostic factors to assist in the management of low-grade gliomas. For patients with a favorable (low-risk) score, treatment with radiotherapy or chemotherapy treatment should be withheld until tumor progression. For patients with a high-risk score, treatment at diagnosis may be indicated. However, other than surgery, the optimal types and sequence of therapies are not yet established. Improvements in defining prognostic factors will assist in low-grade glioma management.


Subject(s)
Brain Neoplasms/mortality , Glioma/mortality , Adolescent , Adult , Age Factors , Aged , Biomarkers, Tumor , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Child , Child, Preschool , Glioma/pathology , Glioma/therapy , Humans , Infant , Infant, Newborn , Middle Aged , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL