Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Cell Prolif ; 49(6): 698-709, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27611480

ABSTRACT

OBJECTIVES: Beryllium salts (here, beryllium sulphate) can produce a cytostatic effect in some cell types. The basis for this effect may include increased expression of proliferation inhibitors, reduced expression of proliferation promoters, or both. This study sought to determine the role of p53, the tumour-suppressing transcription factor, in mediating beryllium-induced cytostasis. MATERIALS AND METHODS: Human A172 glioma cells express wild-type TP53 gene. Activity of p53 was experimentally manipulated using siRNA and related approaches. Key elements of the beryllium-response were compared in normal and p53-knockdown A172 cells using RT-PCR and Western blotting. RESULTS: In A172 cells, 10 µm BeSO4 caused 300% increase in CDKN1A (cyclin-dependent kinase inhibitor p21) mRNA and 90% reduction of CCNE2 (cyclin E2) mRNA. The increased p21 mRNA and reduced cyclin E2 mRNA were each dependent on presence of functional p53. For p21, increased mRNA led to commensurately increased protein levels. In contrast, reduction in cyclin E2 mRNA levels did not lead to corresponding reductions in cyclin E2 protein. The proteasomal inhibitor MG-132 caused p53 protein to increase, but it had no effect on cyclin E2 protein levels. Cycloheximide time course studies indicated that the cyclin E2 protein half-life was more than 12 hours in these cells. CONCLUSIONS: Beryllium elicited p53-dependent changes in mRNA levels of key determinants of cell proliferation such as p21 and cyclin E2. However, cyclin E2 protein appeared to be aberrantly regulated in this cell type, as its turnover was unexpectedly slow.


Subject(s)
Antineoplastic Agents/pharmacology , Beryllium/pharmacology , Brain Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Tumor Suppressor Protein p53/genetics , Brain/drug effects , Brain/metabolism , Brain Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytostatic Agents/pharmacology , Down-Regulation/drug effects , Glioblastoma/genetics , Humans , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Up-Regulation/drug effects
2.
Neurology ; 77(12): 1156-64, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21880994

ABSTRACT

OBJECTIVE: This analysis was performed to assess whether antiepileptic drugs (AEDs) modulate the effectiveness of temozolomide radiochemotherapy in patients with newly diagnosed glioblastoma. METHODS: The European Organization for Research and Treatment of Cancer (EORTC) 26981-22981/National Cancer Institute of Canada (NCIC) CE.3 clinical trial database of radiotherapy (RT) with or without temozolomide (TMZ) for newly diagnosed glioblastoma was examined to assess the impact of the interaction between AED use and chemoradiotherapy on survival. Data were adjusted for known prognostic factors. RESULTS: When treatment began, 175 patients (30.5%) were AED-free, 277 (48.3%) were taking any enzyme-inducing AED (EIAED) and 135 (23.4%) were taking any non-EIAED. Patients receiving valproic acid (VPA) only had more grade 3/4 thrombopenia and leukopenia than patients without an AED or patients taking an EIAED only. The overall survival (OS) of patients who were receiving an AED at baseline vs not receiving any AED was similar. Patients receiving VPA alone (97 [16.9%]) appeared to derive more survival benefit from TMZ/RT (hazard ratio [HR] 0.39, 95% confidence interval [CI] 0.24-0.63) than patients receiving an EIAED only (252 [44%]) (HR 0.69, 95% CI 0.53-0.90) or patients not receiving any AED (HR 0.67, 95% CI 0.49-0.93). CONCLUSIONS: VPA may be preferred over an EIAED in patients with glioblastoma who require an AED during TMZ-based chemoradiotherapy. Future studies are needed to determine whether VPA increases TMZ bioavailability or acts as an inhibitor of histone deacetylases and thereby sensitizes for radiochemotherapy in vivo.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/mortality , Valproic Acid/therapeutic use , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Canada/epidemiology , Dacarbazine/therapeutic use , Europe/epidemiology , Female , Humans , Male , Middle Aged , Retrospective Studies , Survival Rate/trends , Temozolomide , Young Adult
3.
Br J Cancer ; 97(3): 302-7, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17609661

ABSTRACT

This is one of the few studies that have explored the value of baseline symptoms and health-related quality of life (HRQOL) in predicting survival in brain cancer patients. Baseline HRQOL scores (from the EORTC QLQ-C30 and the Brain Cancer Module (BN 20)) were examined in 490 newly diagnosed glioblastoma cancer patients for the relationship with overall survival by using Cox proportional hazards regression models. Refined techniques as the bootstrap re-sampling procedure and the computation of C-indexes and R(2)-coefficients were used to try and validate the model. Classical analysis controlled for major clinical prognostic factors selected cognitive functioning (P=0.0001), global health status (P=0.0055) and social functioning (P<0.0001) as statistically significant prognostic factors of survival. However, several issues question the validity of these findings. C-indexes and R(2)-coefficients, which are measures of the predictive ability of the models, did not exhibit major improvements when adding selected or all HRQOL scores to clinical factors. While classical techniques lead to positive results, more refined analyses suggest that baseline HRQOL scores add relatively little to clinical factors to predict survival. These results may have implications for future use of HRQOL as a prognostic factor in cancer patients.


Subject(s)
Brain Neoplasms/physiopathology , Glioblastoma/physiopathology , Quality of Life , Survival Analysis , Female , Humans , Male , Middle Aged , Prognosis , Prospective Studies
4.
J Neurooncol ; 78(3): 311-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16710748

ABSTRACT

PURPOSE: To determine the response rate, time to disease progression, survival, and toxicity of intravenous carboplatin and chronic oral high-dose tamoxifen in patients with recurrent malignant gliomas. PATIENTS AND METHODS: Patients with histological confirmation of recurrent malignant gliomas were eligible for this multicenter phase II trial. Treatment consisted of 400 mg/m2 carboplatin intravenously every 4 weeks and oral high dose chronic tamoxifen (80 mg bid in women and 100 mg bid in men). RESULTS: Twenty seven patients met the eligibility criteria and were evaluable for response. The histological subtypes were: 16 (59%) glioblastoma multiforme (GBM), malignant astrocytoma (5 patients), malignant mixed glioma (5 patients), and glioblastoma/gliosarcoma (1 patient). Twenty-two patients (82%) had an ECOG performance status of 0 or 1. No complete responses were observed, 4 patients (15%) achieved a partial response, and 14 patients (52%) had stable disease. Median time to progression was 3.65 months (95%CI 2.56, 4.83). Median overall survival was 14.09 months (95%CI 7.06, 19.91). One patient with a recurrent GBM had a sustained partial response and is progression free 81 months since starting treatment. Another patient with mixed malignant oligoastrocytoma also had a prolonged partial response (lasting 63 months) and is alive 84 months after treatment for recurrence. The most frequently reported grade 3 or 4 toxicities were fatigue (19%), nausea (11%) and anorexia (11%). CONCLUSIONS: Carboplatin and high dose tamoxifen has similar response rates to other regimens for recurrent malignant gliomas and are probably equivalent to those found using tamoxifen as monotherapy. Long-lasting periods of disease free survival in some patients (particularly those with malignant mixed oligo astrocytomas) were found.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Anorexia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/pathology , Carboplatin/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Fatigue/chemically induced , Female , Glioma/pathology , Headache/chemically induced , Humans , Male , Middle Aged , Nausea/chemically induced , Tamoxifen/administration & dosage , Treatment Outcome
5.
J Clin Oncol ; 22(9): 1583-8, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15051755

ABSTRACT

PURPOSE: To prospectively compare standard radiation therapy (RT) with an abbreviated course of RT in older patients with glioblastoma multiforme (GBM). PATIENTS AND METHODS: One hundred patients with GBM, age 60 years or older, were randomly assigned after surgery to receive either standard RT (60 Gy in 30 fractions over 6 weeks) or a shorter course of RT (40 Gy in 15 fractions over 3 weeks). The primary end point was overall survival. The secondary end points were proportionate survival at 6 months, health-related quality of life (HRQoL), and corticosteroid requirement. HRQoL was assessed using the Karnofsky performance status (KPS) and Functional Assessment of Cancer Therapy-Brain (FACT-Br). RESULTS: All patients had died at the time of analysis. Overall survival times measured from randomization were similar at 5.1 months for standard RT versus 5.6 months for the shorter course (log-rank test, P =.57). The survival probabilities at 6 months were also similar at 44.7% for standard RT versus 41.7% for the shorter course (lower-bound 95% CI, -13.7). KPS scores varied markedly but were not significantly different between the two groups (Wilcoxon test, P =.63). Low completion rates of the FACT-Br (45%) precluded meaningful comparisons between the two groups. Of patients completing RT as planned, 49% of patients (standard RT) versus 23% required an increase in posttreatment corticosteroid dosage (chi(2) test, P =.02). CONCLUSION: There is no difference in survival between patients receiving standard RT or short-course RT. In view of the similar KPS scores, decreased increment in corticosteroid requirement, and reduced treatment time, the abbreviated course of RT seems to be a reasonable treatment option for older patients with GBM.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Adrenal Cortex Hormones/therapeutic use , Age Factors , Aged , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Dose Fractionation, Radiation , Female , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Karnofsky Performance Status , Male , Middle Aged , Prospective Studies , Radiotherapy, Adjuvant , Survival Analysis , Treatment Outcome
6.
Cancer Res ; 61(18): 6713-5, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559541

ABSTRACT

Molecular genetic subsets of anaplastic oligodendroglioma behave in biologically distinct ways, in both their rates of growth and their responses to standard therapies. In a series of 64 cases, we evaluated whether allelic loss of chromosomal arms 1p and 19q, an early molecular event in the genesis of chemosensitive oligodendrogliomas, is related to tumor location and extent of tumor spread in the brain. We observed that tumor genotype was closely associated with tumor location (P < 0.001). Anaplastic oligodendrogliomas located in the frontal, parietal, and occipital lobes were significantly more likely to harbor allelic loss of chromosomal arms 1p and 19q than histologically indistinguishable tumors arising in the temporal lobe, insula, and diencephalon (P < 0.001). In addition, loss of heterozygosity for 1p and 19q was significantly associated with a bilateral pattern of growth (P = 0.037); all seven bilaterally distributed anaplastic oligodendrogliomas had 1p and 19q allelic loss. We conclude, therefore, that molecular subtypes of oligodendrogliomas may arise preferentially in certain lobes of the brain and have differential patterns of growth, with tumors having allelic loss of chromosomes 1p and 19q occurring most frequently in the frontal lobes and having a tendency for widespread growth across the midline. These findings encourage inquiries into the biological basis of such marked differences and already have implications for the current management of these neoplasms.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/drug therapy , Cell Division/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , Oligodendroglioma/drug therapy
8.
Can J Neurol Sci ; 28(3): 215-23, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11513339

ABSTRACT

BACKGROUND: We report survival and pretreatment prognostic factors for survival and chemosensitivity in 53 oligodendrogliomas treated with PCV (procarbazine, lomustine and vincristine) chemotherapy. METHODS: A total of 53 patients with histologically proven oligodendroglioma, anaplastic oligodendroglioma or oligo-astrocytoma and treated with PCV were extracted from the London Regional Cancer Center database. A retrospective review was conducted to evaluate overall survival and pretreatment prognostic factors for survival and chemosensitivity. RESULTS: The median survival time from diagnosis was 123.6 months. The overall five- and ten-year survival rates were 72.7% and 52.7% respectively. Age <40, seizure as an initial symptom, absence of cognitive deficit and presence of a homogeneous hypodense lesion without contrast enhancement on the initial pretreatment CT scan were all factors independently associated with favorable outcome. The presence of increased cellularity, pleomorphism, mitosis, vascular proliferation and grading as an anaplastic lesion using these surrogates on pathological assessment, were all associated with an unfavorable outcome in univariable analysis. In multivariable analysis, only the anaplastic grading and presence of increased cellularity were significant determinants of unfavorable survival. The only factor adversely associated with chemosensitivity was the presence of a focal symptom at presentation. CONCLUSION: Overall survival is significantly longer in oligodendroglial lesions than in fibrillary astrocytic tumors. A two tier grading system using standard morphological features seems accurate in predicting outcome in these patients. The presence of a neoplastic astrocytic component does not seem to impact the outcome. No clinical, radiological or pathological factor could be identified to reliably predict chemotherapy response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Cisplatin/therapeutic use , Oligodendroglioma/drug therapy , Paclitaxel/therapeutic use , Vinblastine/therapeutic use , Adult , Age Factors , Astrocytoma/psychology , Brain Neoplasms/psychology , Cognition Disorders/etiology , Cognition Disorders/psychology , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Multivariate Analysis , Oligodendroglioma/psychology , Prognosis , Survival Analysis , Taxoids , Vinblastine/analogs & derivatives
9.
Am J Pathol ; 159(1): 359-67, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438483

ABSTRACT

Allelic loss of 10q is a common genetic event in malignant gliomas, with three 10q tumor suppressor genes, ERCC6, PTEN, and DMBT1, putatively implicated in the most common type of malignant glioma, glioblastoma. Anaplastic oligodendroglioma, another type of malignant glioma, provides a unique opportunity to study the relevance of particular genetic alterations to chemosensitivity and survival. We therefore analyzed these three genes in 72 anaplastic oligodendrogliomas. Deletion mapping demonstrated 10q loss in 14 of 67 informative cases, with the PTEN and DMBT1 regions involved in all deletions but with the ERCC6 locus spared in two cases. Seven tumors had PTEN gene alterations; two had homozygous DMBT1 deletions, but at least one reflected unmasking of a germline DMBT1 deletion. No mutations were found in ERCC6 exon 2. Chemotherapeutic response occurred in two of the seven tumors with PTEN alterations, but with unexpected short survival times. PTEN gene alterations were not associated with poor therapeutic response in multivariate analysis, but were independently predictive of poor prognosis even after multivariate adjustment for both 10q and 1p loss. In anaplastic oligodendroglioma, therefore, PTEN is a target of 10q loss, and PTEN alterations are associated with poor prognosis, even in chemosensitive cases.


Subject(s)
Agglutinins , Chromosomes, Human, Pair 10/genetics , Mutation/physiology , Oligodendroglioma/genetics , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Proteins , Antineoplastic Agents/therapeutic use , Calcium-Binding Proteins , Chromosome Mapping , DNA Helicases/genetics , DNA Repair Enzymes , DNA-Binding Proteins , Gene Deletion , Homozygote , Humans , Oligodendroglioma/drug therapy , PTEN Phosphohydrolase , Poly-ADP-Ribose Binding Proteins , Prognosis , Receptors, Cell Surface/genetics , Survival Analysis , Treatment Outcome
10.
Cancer Res ; 61(10): 4155-9, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358839

ABSTRACT

We examined the effect of p53 inactivation on the response of U87MG glioma cells to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). These studies were motivated by three observations: (a) some human astrocytomas are sensitive to BCNU and some are resistant; (b) chemosensitive astrocytomas are more likely to be found in young adults whose tumors are more likely to harbor a p53 mutation; and (c) mouse astrocytes lacking the p53 gene are more sensitive to BCNU than wild-type cells. Here, we observed that p53 inactivation by transfection with pCMV-E6 sensitized U87MG cells to BCNU. Compared with control U87MG-neo cells with intact p53 function, the clonogenic survival of U87MG-E6 cells exposed to BCNU was reduced significantly. In U87MG-E6 cells, sensitization to BCNU was associated with failure of p21(WAF1) induction, transient cell cycle arrest in S phase, accumulation of polyploid cells, and significant cell death. In contrast, resistance to BCNU in U87MG-neo cells was associated with up-regulation of p53, prolonged induction of p21(WAF1), sustained cell cycle arrest in S phase, and enhancement of DNA repair. U87MG cells with disrupted p53 function were less able to repair BCNU-induced DNA damage and survive this chemotherapeutic insult. The question arises of whether p53 dysfunction might be a chemosensitizing genetic alteration in human astrocytic gliomas.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Glioma/drug therapy , Glioma/genetics , Tumor Suppressor Protein p53/physiology , Cell Cycle/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA Repair/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioma/pathology , Humans , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
11.
Neuro Oncol ; 3(1): 46-54, 2001 01.
Article in English | MEDLINE | ID: mdl-11305417

ABSTRACT

Therapeutic options for the treatment of malignant brain tumors have been limited, in part, because of the presence of the blood-brain barrier. For this reason, the Sixth Annual Meeting of the Blood-Brain Barrier Disruption Consortium, the focus of which was the "Importance of Dose Intensity in Neuro-Oncology Clinical Trials," was convened in April 2000, at Government Camp, Mount Hood, Oregon. This meeting, which was supported by the National Cancer Institute, the National Institute of Neurological Disorders and Stroke, and the National Institute of Deafness and Other Communication Disorders, brought together clinicians and basic scientists from across the U.S. to discuss the role of dose intensity and enhanced chemotherapy delivery in the treatment of malignant brain tumors and to design multicenter clinical trials. Optimizing chemotherapy delivery to the CNS is crucial, particularly in view of recent progress identifying certain brain tumors as chemosensitive. The discovery that specific constellations of genetic alterations can predict which tumors are chemoresponsive, and can therefore more accurately predict prognosis, has important implications for delivery of intensive, effective chemotherapy regimens with acceptable toxicities. This report summarizes the discussions, future directions, and key questions regarding dose-intensive treatment of primary CNS lymphoma, CNS relapse of systemic non-Hodgkin's lymphoma, anaplastic oligodendroglioma, high-grade glioma, and metastatic cancer of the brain. The promising role of cytoenhancers and chemoprotectants as part of dose-intensive regimens for chemosensitive brain tumors and development of improved gene therapies for malignant gliomas are discussed.


Subject(s)
Antineoplastic Agents/administration & dosage , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Hypertonic Solutions/pharmacology , Meningeal Neoplasms/drug therapy , Adult , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Diseases/chemically induced , Bone Marrow Transplantation , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Buthionine Sulfoximine/pharmacology , Buthionine Sulfoximine/therapeutic use , Child , Clinical Trials as Topic/methods , Clinical Trials, Phase III as Topic , Cognition Disorders/etiology , Combined Modality Therapy , Cranial Irradiation , Dose-Response Relationship, Drug , Drug Synergism , Genetic Therapy , Genetic Vectors/pharmacokinetics , Glioma/drug therapy , Glioma/metabolism , Glutathione/metabolism , Guinea Pigs , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/prevention & control , Hematopoietic Stem Cell Transplantation , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Meningeal Neoplasms/physiopathology , Meningeal Neoplasms/secondary , Meningeal Neoplasms/therapy , Multicenter Studies as Topic/methods , Neuroblastoma/drug therapy , Oligodendroglioma/drug therapy , Permeability/drug effects , Quality of Life , Randomized Controlled Trials as Topic/methods , Treatment Outcome
12.
Clin Cancer Res ; 7(4): 839-45, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309331

ABSTRACT

PURPOSE: In a prior study of anaplastic oligodendrogliomas treated with chemotherapy at diagnosis or at recurrence after radiotherapy, allelic loss of chromosome 1p correlated with better chemotherapeutic response and overall survival. However, in this group of patients in whom therapeutic management was not uniform, loss of 1p did not identify all chemosensitive tumors, nor did all patients whose tumors harbor a 1p loss have long survival. EXPERIMENTAL DESIGN: To clarify the clinical relevance of molecular genetic testing at the time of diagnosis for patients with anaplastic oligodendrogliomas, we studied a larger, more homogeneous group of 50 patients with histologically defined anaplastic oligodendrogliomas treated with a chemotherapeutic regimen as the principal initial therapy. RESULTS: We demonstrate that these tumors can be divided genetically into four therapeutically and prognostically relevant subgroups. Patients whose tumors have combined but isolated losses of 1p and 19q have marked and durable responses to chemotherapy associated with long survival, with or without postoperative radiation therapy. Other tumors with chromosome 1p alterations also respond to chemotherapy, but with shorter duration of response and patient survival. Tumors lacking 1p loss can also be divided into two subgroups: those with TP53 mutations, which may also respond to chemotherapy but recur quickly, and those without TP53 mutations, which are poorly responsive, aggressive tumors that are clinically and genotypically similar to glioblastomas. CONCLUSIONS: These data raise the possibility, for the first time, that therapeutic decisions at the time of diagnosis might be tailored to particular genetic subtypes of anaplastic oligodendroglioma.


Subject(s)
Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , Oligodendroglioma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Chromosome Deletion , Female , Genetic Markers , Humans , Male , Middle Aged , Oligodendroglioma/diagnosis , Oligodendroglioma/mortality , Prognosis , Survival Rate
13.
Semin Radiat Oncol ; 11(2): 170-80, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285555

ABSTRACT

Once thought to be rare, oligodendroglial tumors might actually represent up to 25% of primary glial neoplasms. In recent years, the histologic criteria for the diagnosis of oligodendroglioma have been broadened to include most small cell, monomorphic glial neoplasms. These refinements have led to an increased recognition of oligodendroglial neoplasms, but uniform definitions of pure versus mixed oligodendroglioma as well as the criteria for high-grade (anaplastic) versus low-grade tumors remain elusive. From a prognostic standpoint, the presence of an oligodendroglial component in a malignant glioma predicts longer survivals times for patients treated with surgery, and radiation therapy with or without chemotherapy. High rates of response to PCV (procarbazine, CCNU and Vincristine) chemotherapy also have been noted among patients with anaplastic oligodendroglial neoplasms. Ongoing prospective trials seek to clarify the role of PCV chemotherapy when added to radiation therapy and surgery. In addition, the role of molecular markers as diagnostic aides and guides to therapy and prognosis are being explored for patients with pure and mixed anaplastic oligodendroglial tumors.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Oligodendroglioma/therapy , Adult , Antineoplastic Combined Chemotherapy Protocols , Combined Modality Therapy , Humans , Lomustine , Neoplasm Staging , Procarbazine , Prognosis , Treatment Outcome , Vincristine
14.
Int J Radiat Oncol Biol Phys ; 48(3): 825-30, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11020580

ABSTRACT

INTRODUCTION: Allelic loss of the short arm of chromosome 1 predicts radiographic response to chemotherapy and long overall survival times in patients with anaplastic oligodendrogliomas. Using a database of patients with oligodendrogliomas in whom chromosome 1p status was known, we explored whether allelic loss of 1p also predicted longer duration of tumor control when radiotherapy was part of the initial treatment of these patients. MATERIALS AND METHODS: We measured progression-free survival following radiotherapy in a cohort of patients with World Health Organization (WHO) Grade II and WHO Grade III oligodendrogliomas. The effects on progression-free survival of patient age, Karnofsky performance score (KPS), tumor grade when irradiated and chromosome 1p status were examined by univariate and multivariate statistical analyses. For the subset of patients with newly diagnosed anaplastic oligodendrogliomas, relationships between use of chemotherapy, chromosome 1p status and progression-free survival were also examined. RESULTS: Fifty-five patients (29 male, 26 female; ages 18-75 years; median, 44 years; KPS 50-90, median 80) were irradiated for either a WHO Grade II (n = 19) or Grade III (n = 36) oligodendroglioma. Twenty-eight patients had chemotherapy immediately prior to radiotherapy, and 27 had chemotherapy at progression following radiotherapy. The median radiation dose was 54 Gy in 30 fractions. Loss of heterozygosity (LOH) at chromosome 1p was evident in 36 tumors and absent in 19. Overall median progression-free survival after radiotherapy was 40.4 months. Median progression-free survival was 55.0 months for patients whose tumors harbored 1p loss vs. 6.2 months for those patients whose tumors retained both copies of chromosome 1p (p < 0.001). On both univariate and multivariate analyses, chromosome lp loss was the principal independent predictor of longer progression-free survival for patients with Grade II and III oligodendrogliomas. For Grade III oligodendrogliomas, chemotherapy as an adjunct to radiotherapy prolonged tumor control for those patients whose tumors harbored allelic loss of chromosome 1p (p = 0.004). CONCLUSION: These data suggest allelic loss of chromosome 1p in patients with oligodendroglial neoplasms predicts longer progression-free survival among patients receiving radiotherapy +/- chemotherapy as part of their initial treatment. Chromosome 1p loss may be an important stratification variable in future therapeutic trials of oligodendroglioma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Chromosome Deletion , Chromosomes, Human, Pair 1 , Oligodendroglioma/genetics , Oligodendroglioma/therapy , Adolescent , Adult , Aged , Analysis of Variance , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Female , Humans , Karnofsky Performance Status , Male , Middle Aged , Oligodendroglioma/drug therapy , Oligodendroglioma/radiotherapy
15.
Cancer Res ; 60(17): 4812-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10987291

ABSTRACT

The two principal subtypes of glial neoplasms, astrocytomas and oligodendrogliomas, exhibit striking differences in response to chemotherapy. This differential chemosensitivity might be explained by the specific genetic alterations causing gliomas but could also be attributable to specific properties intrinsic to the cells from which gliomas arise. To examine the possibility that chemosensitivity might be associated with lineage-specific properties of potential ancestors of these tumors, we explored: (a) the expression of drug resistance genes in rat glial cells; (b) the sensitivity of rat glial subtypes to the bifunctional alkylating agent, 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU); and (c) the effect of O6-methylguanine-DNA methyltransferase (MGMT) and glutathione modulation on resistance to BCNU. Astrocytes, O-2A progenitors, and oligodendrocytes each displayed a unique pattern of expression of six drug resistance genes: MGMT, GST mu, GST pi,p53, MDR, and MT. Oligodendrocytes were more sensitive to BCNU than either astrocytes or O-2A progenitors. The increased resistance of astrocytes in comparison to oligodendrocytes was modulated, at least in part, by both O6-benzylguanine (BG) and DL-buthionine-(S,R)-sulfoximine, suggesting a role for both MGMT and glutathione in the resistance of astrocytes to BCNU. The sensitivity of O-2A progenitors to BCNU following BG pretreatment is virtually indistinguishable from that of oligodendrocytes depleted of MGMT, suggesting that the down-regulation of MGMT is sufficient to account for the increased sensitivity of oligodendrocyte lineage cells to BCNU as they differentiate. These experiments provide support for the hypothesis that properties of glial cells retained in gliomas may contribute to the differential chemosensitivity of glial neoplasms.


Subject(s)
Astrocytes/physiology , Astrocytoma/pathology , Drug Resistance, Neoplasm/genetics , Oligodendroglia/physiology , Oligodendroglioma/pathology , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytoma/drug therapy , Carmustine/pharmacology , Cell Death/drug effects , Cell Lineage , Cricetinae , Gene Expression , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Glutathione/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Isoenzymes/biosynthesis , Isoenzymes/genetics , Multidrug Resistance-Associated Proteins , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglioma/drug therapy , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
16.
J Neurosurg ; 92(6): 983-90, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10839259

ABSTRACT

OBJECT: Allelic loss of chromosome 1p is a powerful predictor of tumor chemosensitivity and prolonged survival in patients with anaplastic oligodendrogliomas. Chromosome 1p loss also occurs in astrocytic and oligoastrocytic gliomas, although less commonly than in pure oligodendroglial tumors. This observation raises the possibility investigated in this study that chromosome 1p loss might also provide prognostic information for patients with high-grade gliomas with astrocytic components. METHODS: The authors report on seven patients with high-grade gliomas composed of either pure astrocytic or mixed astrocytic-oligodendroglial phenotypes, who had remarkable neuroradiological responses to therapy or unexpectedly long survivals. All of the tumors from these seven patients demonstrated chromosome 1p loss, whereas other genetic alterations characteristic of high-grade gliomas (p53 gene mutations, EGFR gene amplification, chromosome 10 loss, chromosome 19q loss, or CDKN2A/p16 deletions) were only found in occasional cases. The authors also assessed the frequency of chromosome 1p loss in a series of anonymous high-grade astrocytoma samples obtained from a tumor bank and demonstrate that this genetic change is uncommon, occurring in only 10% of cases. CONCLUSIONS: Although any prognostic importance of chromosome 1p loss in astrocytic or mixed astrocytic-oligodendroglial gliomas can only be determined in larger and prospective series, these findings raise the possibility that some high-grade gliomas with chromosome 1p loss, in addition to pure anaplastic oligodendrogliomas, may follow a more favorable clinical course.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Chromosomes, Human, Pair 1 , Gene Deletion , Glioma/genetics , Glioma/pathology , Adult , Alleles , Female , Gene Frequency , Genetic Markers , Glioblastoma/genetics , Humans , Infant, Newborn , Male , Middle Aged , Phenotype , Prognosis , Retrospective Studies , Survival Analysis
19.
Carcinogenesis ; 20(12): 2361-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10590234

ABSTRACT

We observed previously that wild-type p53 rendered neonatal mouse astrocytes resistant to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) in a gene dose-dependent fashion. This effect of p53 appeared to be unrelated to its cell cycle regulation or apoptotic functions. Because in many cell types O(6)-methylguanine-DNA methyltransferase (MGMT)-mediated DNA repair is an important mechanism of resistance to nitrosoureas, we measured MGMT activity in wild-type, heterozygous and p53 knockout neonatal mouse astrocytes. Wild-type p53 astrocytes had significantly greater MGMT activity than either heterozygous or p53 knockout astrocytes: MGMT activity was approximately 5-fold greater in wild-type p53 astrocytes than in p53 knockout cells. However, despite successful depletion of MGMT activity in wild-type astrocytes by O(6)-benzylguanine (BG), resistance to BCNU persisted unchanged. Moreover, we excluded the possibility that continued resistance to BCNU at the concentrations used could be explained by a compensatory induction of MGMT triggered by exposure to either BCNU or BG. Although these studies support a role for p53 regulation of MGMT in neonatal mouse astrocytes, BCNU resistance in wild-type cells appears to be mediated by a non-MGMT mechanism. Nevertheless, regulation of DNA repair by MGMT may be another mechanism by which alterations of the p53 gene promote tumor initiation or progression.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Astrocytes/drug effects , Carmustine/pharmacology , Genes, p53 , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Base Sequence , DNA Primers , Drug Resistance , Enzyme Induction , Mice , Mice, Knockout , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis
20.
Int J Radiat Oncol Biol Phys ; 45(4): 923-9, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10571199

ABSTRACT

PURPOSE: Three databases were pooled and analyzed to determine which groupings of prognostic factors best predicted overall survival for patients with low-grade gliomas treated with surgery and immediate or delayed radiotherapy. METHODS AND MATERIALS: Databases of patients with low-grade gliomas compiled at the London Regional Cancer Centre (LRCC), the Norwegian Radium Hospital (NRH), and the University of California, San Francisco (UCSF) were merged. Inclusion criteria for the pooled analysis included: age > or =18 years and histologically confirmed low-grade (World Health Organization Grade II) supratentorial fibrillary astrocytoma, oligodendroglioma or mixed oligoastrocytoma. Factors analyzed for prognostic significance included: age at diagnosis, gender, seizures at presentation, presence of enhancement on computed tomography (CT) or magnetic resonance imaging (MRI), Karnofsky Performance Status (KPS) at diagnosis, histology, extent of surgical resection, timing of radiotherapy, and treating institution. Univariate and multivariate analysis of overall survival for these factors was performed. Recursive partitioning was performed to generate prognostic groups using these factors. RESULTS: From the combined databases, 401 patients were eligible for analysis. Median survival for the entire group was 95 months/7.9 years. On univariate analysis age 18-40, presence of seizures at presentation, KPS > or =70, treating institution, and absence of contrast enhancement were associated with improved overall survival. On multivariate analysis, these factors remained independent predictors of improved overall survival. Recursive partitioning analysis yielded four prognostic groups with statistically different median survivals (MS): Group I (n = 41: KPS <70, age >40) MS 12 months; Group II (n = 34: KPS > or =70, age >40, enhancement present) MS 46 months; Group III (n = 138: KPS <70, age 18-40 or KPS > or =70 age >40, no enhancement) MS 87 months; Group IV (n = 188: KPS > or =70, age 18-40) MS 128 months. CONCLUSION: Clusters of pretreatment prognostic factors described subgroups of low-grade glioma patients with divergent overall survivals. Consideration of these prognostic subgroups may be important when considering timing of interventions for these patients and in the stratification of patients for clinical trials.


Subject(s)
Glioma/mortality , Supratentorial Neoplasms/mortality , Adult , Age Factors , Analysis of Variance , Combined Modality Therapy , Databases as Topic , Female , Glioma/pathology , Glioma/therapy , Humans , Male , Prognosis , Sex Factors , Supratentorial Neoplasms/pathology , Supratentorial Neoplasms/therapy , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL