Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Oncogene ; 29(24): 3501-8, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20418911

ABSTRACT

The migration and invasion inhibitor protein (MIIP, also known as IIp45) was discovered as a negative regulator of cell migration and invasion in glioma. Our previous studies have shown that the MIIP protein was reduced or undetectable in some tissue samples obtained from patients with glioblastoma. The significance of MIIP in gliomagenesis is unknown. In this study, we report that MIIP has an important role in the inhibition of gliomagenesis and attenuation of mitotic transition. Increased MIIP expression levels inhibited colony formation and cell growth of glioma cell lines in vitro, whereas decreased expression by specific small interfering RNA for MIIP resulted in increased cell growth. Expression of MIIP in a glial-specific mouse model blocked glioma development and progression, thus showing that MIIP is an inhibitor of gliomagenesis. Furthermore, we show that MIIP attenuates mitotic transition and results in increased mitotic catastrophe. The biochemical mechanism of MIIP in this process is associated with its regulation of anaphase-promoting complex (APC/C) activity. MIIP interacts directly with Cdc20, and the interaction of MIIP with Cdc20 inhibits APC/C-mediated degradation of cyclin B1. Thus, MIIP attenuates mitotic transition and increases mitotic catastrophe, thereby inhibiting glioma development and progression.


Subject(s)
Carrier Proteins/metabolism , Glioma/metabolism , Glioma/pathology , Mitosis , Animals , Carrier Proteins/genetics , Cdc20 Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin B1/chemistry , Cyclin B1/metabolism , Disease Progression , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Neuroglia/pathology , Organ Specificity , Protein Stability , Ubiquitin-Protein Ligases/metabolism
2.
Apoptosis ; 10(1): 233-43, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15711939

ABSTRACT

Patients with malignant gliomas have a poor prognosis and new treatment paradigms are needed against this disease. TRAIL/Apo2L selectively induces apoptosis in malignant cells sparing normal cells and is hence of interest as a potential therapeutic agent against gliomas. To determine the factors that modulate sensitivity to TRAIL, we examined the differences in TRAIL-activated signaling pathways in glioma cells with variable sensitivities to the agent. Apoptosis in response to TRAIL was unrelated to DR5 expression or endogenous p53 status in a panel of 8 glioma cell lines. TRAIL activated the extrinsic (cleavage of caspase-8, caspase-3 and PARP) and mitochondrial apoptotic pathways and reduced FLIP levels. It also induced caspase-dependent JNK activation, which did not influence TRAIL-induced apoptosis. Because the pro-survival PI3K/Akt pathway is highly relevant to gliomas, we assessed whether Akt could protect against TRAIL-induced apoptosis. Pretreatment with SH-6, a novel Akt inhibitor, enhanced TRAIL-induced apoptosis, suggesting a protective role for Akt. Conversely, TRAIL induced caspase-dependent cleavage of Akt neutralizing its anti-apoptotic effects. These results demonstrate that TRAIL-induced apoptosis in gliomas involves both activation of death pathways and downregulation of survival pathways. Additional studies are warranted to determine the therapeutic potential of TRAIL against gliomas.


Subject(s)
Apoptosis , Glioma/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Glycoproteins/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Apoptosis Regulatory Proteins , Cell Line, Tumor , Enzyme Activation , Humans , MAP Kinase Kinase 4 , Proto-Oncogene Proteins c-akt , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand
3.
J Natl Cancer Inst ; 93(20): 1553-7, 2001 Oct 17.
Article in English | MEDLINE | ID: mdl-11604478

ABSTRACT

BACKGROUND: About 9% of human cancers are brain tumors, of which 90% are gliomas. gamma-Radiation has been identified as a risk factor for brain tumors. In a previous pilot study, we found that lymphocytes from patients with glioma were more sensitive to gamma-radiation than were lymphocytes from matched control subjects. In this larger case-control study, we compared the gamma-radiation sensitivity of lymphocytes from glioma patients with those from control subjects and investigated the association between mutagen sensitivity and the risk for developing glioma. METHODS: We used a mutagen sensitivity assay (an indirect measure of DNA repair activity) to assess chromosomal damage. We gamma-irradiated (1.5 Gy) short-term lymphocyte cultures from 219 case patients with glioma and from 238 healthy control subjects frequency matched by age and sex. After irradiation, cells were cultured for 4 hours, and then Colcemid was added for 1 hour to arrest cells in mitosis. Fifty metaphases were randomly selected for each sample and scored for chromatid breaks. All statistical tests were two-sided. RESULTS: We observed a statistically significantly higher frequency of chromatid breaks per cell from case patients with glioma (mean = 0.55; 95% confidence interval [CI] = 0.50 to 0.59) than from control subjects (mean = 0.44; 95% CI = 0.41 to 0.48) (P<.001). Using 0.40 (the median number of chromatid breaks per cell in control subjects) as the cut point for defining mutagen sensitivity and adjusting for age, sex, and smoking status, we found that mutagen sensitivity was statistically significantly associated with an increased risk for glioma (odds ratio = 2.09; 95% CI = 1.43 to 3.06). When the data were divided into tertiles, the relative risk for glioma increased from the lowest tertile to the highest tertile (trend test, P<.001). CONCLUSION: gamma-Radiation-induced mutagen sensitivity of lymphocytes may be associated with an increased risk for glioma, a result that supports our earlier preliminary findings.


Subject(s)
Brain Neoplasms/genetics , DNA Repair/genetics , Gamma Rays/adverse effects , Glioma/genetics , Neoplasms, Radiation-Induced/genetics , Adult , Animals , Brain Neoplasms/epidemiology , Brain Neoplasms/etiology , Case-Control Studies , Chromatids/radiation effects , Chromatids/ultrastructure , Chromosome Breakage , DNA/radiation effects , DNA Damage , DNA Repair/radiation effects , DNA, Single-Stranded/radiation effects , Demecolcine/pharmacology , Female , Genetic Predisposition to Disease , Glioma/epidemiology , Glioma/etiology , Humans , Lymphocytes/pathology , Lymphocytes/radiation effects , Male , Middle Aged , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Odds Ratio , Radiation Tolerance/genetics , Risk , Smoking/epidemiology
4.
Cancer Res ; 61(11): 4375-81, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389063

ABSTRACT

Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin H synthase, has been implicated in the growth and progression of a variety of human cancers. Although COX-2 overexpression has been observed in human gliomas, the prognostic or clinical relevance of this overexpression has not been investigated to date. In addition, no study has analyzed the relationship between COX-2 expression and other molecular alterations in gliomas. Consequently, we examined COX-2 expression by immunohistochemistry in tumor specimens from 66 patients with low- and high-grade astrocytomas and correlated the percentage of COX-2 expression with patient survival. We also analyzed the relative importance of COX-2 expression in comparison with other clinicopathological features (age and tumor grade) and other molecular alterations commonly found in gliomas (high MIB-1 level, p53 alteration, loss of retinoblastoma (Rb) protein or p16, and high bcl-2 level). Kaplan-Meier analyses demonstrated that high COX-2 expression (>50% of cells stained positive) correlated with poor survival for the study group as a whole (P < 0.0001) and for those with glioblastoma multiforme in particular (P < 0.03). Cox regression analyses demonstrated that COX-2 expression was the strongest predictor of outcome, independent of all other variables. In addition, high COX-2 expression correlated with increasing histological grade but did not correlate with positive p53 immunostaining, bcl-2 expression, loss of p16 or retinoblastoma protein expression, or high MIB-1 expression. These findings indicate that high COX-2 expression in tumor cells is associated with clinically more aggressive gliomas and is a strong predictor of poor survival.


Subject(s)
Astrocytoma/enzymology , Brain Neoplasms/enzymology , Glioblastoma/enzymology , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Adolescent , Adult , Aged , Antigens, Nuclear , Astrocytoma/mortality , Astrocytoma/pathology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclooxygenase 2 , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Immunohistochemistry , Ki-67 Antigen , Membrane Proteins , Middle Aged , Nuclear Proteins/biosynthesis , Prognosis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Retrospective Studies , Survival Rate , Tumor Suppressor Protein p53/biosynthesis
5.
Neurosurgery ; 48(5): 1161-5; discussion 1165-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11334286

ABSTRACT

OBJECTIVE AND IMPORTANCE: A case of metastasis to the pituitary gland from a ductal adenocarcinoma of the salivary gland is presented. Metastasis to this site is rare, and a salivary gland source has never previously been described. CLINICAL PRESENTATION: This patient presented with hypopituitarism, including diabetes insipidus. INTERVENTION: A craniotomy was performed to alleviate visual loss. The histological features of the sellar tumor were identical to those of a tumor removed from the parotid gland 18 months earlier. CONCLUSION: Although intrasellar tumors originating from embryonic rests of salivary gland tissue have been reported, metastasis from a malignant neoplasm arising within a true salivary gland is also possible and should not be excluded from consideration for patients in whom a salivary gland-like tumor is discovered in the sella turcica.


Subject(s)
Carcinoma, Ductal, Breast/secondary , Pituitary Neoplasms/secondary , Salivary Gland Neoplasms/pathology , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/surgery , Combined Modality Therapy , Fatal Outcome , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery
6.
Mutat Res ; 490(1): 35-44, 2001 Jan 25.
Article in English | MEDLINE | ID: mdl-11152970

ABSTRACT

Chromosome instability (CIN) measured as chromosome aberrations has long been suggested as a cancer susceptibility biomarker. Conventional cytogenetic end-points are now being improved by combining molecular methods, which increases the sensitivity, specificity, and precision of the assay. In this study we examined both spontaneous and gamma-ray induced CIN in lymphocyte cultures from 51 previously untreated glioma patients and 51 age-, sex- and ethnicity-matched controls. CIN was assessed using two parallel methods: (1) the mutagen sensitivity (MS) assay and (2) the multicolor fluorescence in situ hybridization (FISH) assay. The frequency of spontaneous breaks was significantly higher in glioma patients (mean+/-S.D., 2.12+/-1.07) than in controls (1.24+/-0.86, P<0.001) when using the FISH assay but not the MS assay (0.019+/-0.02 and 0.019+/-0.01, respectively; P=0.915). Similarly, the frequency of induced chromatid breaks was significantly higher using the FISH assay (3.39+/-1.72) but not the MS assay (0.42+/-0.16) in the patients versus controls (2.08+/-1.18 and 0.37+/-0.15, respectively; P<0.001 and P=0.10, respectively). By using the median number of breaks in the controls as the cutoff value, we observed an odds ratio (ORs) of 5.13 (95% CI=2.23-12.1) for spontaneous and 4.86 (95% CI=2.08-11.4) for induced CIN using the FISH assay, whereas the ORs were 1.32 (95% CI=0.49-3.58) and 1.28 (95% CI=0.59-2.80) for spontaneous and induced CIN using the MS assay. There was also a significant increase in the frequency of hyperdiploid cells in the glioma cases which could only be detected using the FISH assay (OR=4.0, 95% CL=0.9-17.0). By combining both methods an estimated risk of 7.0 (95% CI=1.7-25.6) was observed. There was no correlation between the breaks detected by the two methods suggesting that each method is a measure of a different event. The results indicate that using the multicolor FISH assay for detection of CIN in peripheral blood lymphocytes in glioma patients is a more useful marker for risk assessment.


Subject(s)
Central Nervous System Neoplasms/genetics , Genetic Testing/methods , Glioma/genetics , In Situ Hybridization, Fluorescence/methods , Micronucleus Tests/methods , Adult , Central Nervous System Neoplasms/pathology , Chromosome Aberrations , Female , Glioma/pathology , Humans , Male , Middle Aged
7.
J Neurol ; 248(12): 1030-5, 2001 Dec.
Article in English | MEDLINE | ID: mdl-12013578

ABSTRACT

Oligodendroglial and ependymal tumours are not the most common glial neoplasms; however, they are important subtypes of gliomas with different tumour biologies. Cytogenetic information has suggested that losses of chromosomes 1 p and 19 q are the most frequent genetic alterations in oligodendroglial tumours. Combined loss of these chromosomes has been associated with better chemotherapeutic response and prolonged overall survival. Loss of chromosome 22 is a well defined abnormality in ependymomas. In addition, deletion of chromosome 6 q may be another frequent chromosomic aberration in paediatric ependymomas.


Subject(s)
Brain Neoplasms/genetics , Ependymoma/genetics , Oligodendroglioma/genetics , Brain Neoplasms/pathology , Chromosomes/genetics , Chromosomes/ultrastructure , Ependymoma/pathology , Humans , Oligodendroglioma/pathology
8.
Clin Cancer Res ; 6(10): 3878-84, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11051233

ABSTRACT

Although the efficacy of the nitrosourea-based combination chemotherapy procarbazine, N-(2-chloroethyl)-N'-cyclohexyl-N-nitrosurea, and vincristine (PCV) has been previously demonstrated in the setting of anaplastic/intermediate-grade gliomas, the benefit for glioblastoma patients remains unproven. In the current study, we sought to determine whether the addition of alpha-difluoromethylornithine (eflornithine), an inhibitor of ornithine decarboxylase, which has shown encouraging results in the setting of recurrent glioma patients, to a nitrosourea-based therapy (PCV) would constitute a more effective adjuvant therapy in the treatment of glioblastoma multiforme patients in the postradiation therapy setting. Following conventional radiation therapy, 272 glioblastoma (GBM) patients were randomized to receive either alpha-difluoromethylornithine-PCV (DFMO-PCV; 134 patients) or PCV alone (138 patients), with survival and time to tumor progression being the primary endpoints. The starting dosage of DFMO was 3.0 g/m2 p.o. q8h for 14 days before and after treatment with N-(2-chloroethyl)-N-cyclohexyl-N-nitrosurea; PCV was administered as previously described1. Clinical and radiological (Gadolinium-enhanced MRI) follow-ups were nominally at the end of each 6 or 8 week cycle (PCV at 6 weeks; DFMO-PCV at 8 weeks). Laboratory evaluations for hematologic and other adverse effects were at 2 week intervals. There was no difference in median survival or median time-to-tumor progression between the two treatment groups, as measured from day of commencement of postradiotherapy chemotherapy [MS (months): DFMO-PCV, 10.5; Overall survival, as measured from time of tumor diagnosis at first surgery, was 13.3 and 14.2 months at the median and 6.2 and 8.7% at 5 years, respectively, for the DFMO-PCV and PCV arms. The treatment effect was unchanged after adjustment for age, performance status (KPS), extent of surgery, and other factors using the multivariate Cox proportional hazard model. Adverse effects associated with DFMO consisted of gastrointestinal (diarrhea nausea/vomiting), cytopenias, and minimal ototoxicity (limited to tinnitus) at the dose range tested. The addition of DFMO to the nitrosourea-based PCV regimen in this phase III study demonstrated no additional benefit in glioblastoma patients, underscoring the resistance of glioblastoma multiforme tumors to alkylating agents. For patients with anaplastic (intermediate grade) gliomas, in which the previously demonstrated benefit of post-radiation chemotherapy is more substantial, the evaluation of DFMO-PCV vs. PCV is still ongoing and hopefully will yield more encouraging results.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Eflornithine/administration & dosage , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Lomustine/administration & dosage , Procarbazine/administration & dosage , Vincristine/administration & dosage , Vindesine/administration & dosage , Adolescent , Adult , Aged , Brain Neoplasms/mortality , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Glioblastoma/mortality , Humans , Male , Middle Aged , Prospective Studies , Time Factors
9.
Int J Oncol ; 17(5): 963-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029499

ABSTRACT

The retinoblastoma pathway is a key cell cycle regulatory complex that controls the passage of cells through the G1 checkpoint and is a frequent target of genetic alterations in gliomas. In this study, we examined the expression of Rb and p16 in 170 primary astrocytic gliomas by immunohistochemical techniques, and correlated the expression with overall survival to determine their prognostic value as immunomarkers. There were 130 patients with glioblastoma multiforme (GBM) and 40 with anaplastic astrocytoma (AA). Alterations in the levels of Rb or p16 expression were seen in the majority (>90%) of the gliomas studied. The expression of Rb was completely absent or low in 47.5% of the GBM and 67.5% of the AA. The remainder of the tumors was immunopositive for Rb to varying degrees. Immunoreactivity for p16 was absent in 56% of the GBM and 77.5% of the AA. Kaplan-Meier survival plots (log-rank test) and Cox proportional hazards regression analysis, adjusted for age and histology, showed that neither Rb nor p16 expression independently predicted survival. The results of our study suggest that although genetic alterations of Rb and p16 are common in gliomas, immunohistochemical analysis of these markers correlates poorly with prognosis.


Subject(s)
Astrocytoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Gene Expression Regulation, Neoplastic , Genes, Retinoblastoma , Genes, p16 , Glioblastoma/genetics , Neoplasm Proteins/biosynthesis , Retinoblastoma Protein/biosynthesis , Supratentorial Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytoma/mortality , Astrocytoma/pathology , Child , Child, Preschool , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Life Tables , Middle Aged , Neoplasm Proteins/genetics , Prognosis , Proportional Hazards Models , Supratentorial Neoplasms/mortality , Supratentorial Neoplasms/pathology , Survival Analysis
10.
Histol Histopathol ; 15(3): 971-81, 2000 07.
Article in English | MEDLINE | ID: mdl-10963139

ABSTRACT

As many as 100,000 new cases of brain tumor are diagnosed each year in the United States. About half of these are primary gliomas and the remaining half are metastatic tumors and non-glial primary tumors. Currently, gliomas are classified based on phenotypic characteristics. Recent progress in the elucidation of genetic alterations found in gliomas have raised the exciting possibility of using genetic and molecular analyses to resolve some of the problematic issues currently associated with the histological approach to glioma classification. Recently, immunohistochemical studies using novel proliferation markers have significantly advanced the assessment of tumor growth potential and the grading criteria of some tumor subtypes. Preliminary studies using cDNA array technologies suggest that the profiling of gene expression patterns may provide a novel and meaningful approach to glioma classification and subclassification. Furthermore, cDNA array technologies may also be used to identify candidate genes involved in glioma tumor development, invasion, and progression. This review summarizes current glioma classification schemes that are based on histopathological characteristics and discusses the potential for using cDNA array technology in the molecular classification of gliomas.


Subject(s)
Brain Neoplasms/classification , Glioma/classification , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Expression Profiling , Glioma/genetics , Glioma/pathology , Humans
11.
Anticancer Res ; 20(1A): 65-73, 2000.
Article in English | MEDLINE | ID: mdl-10769636

ABSTRACT

Primitive neuroectodermal tumors constitute a large class of pediatric brain tumors. Despite notable recent advances in improving treatment and survival, the pathogenesis and the molecular genetic bases of these malignancies remain poorly understood. Combined cytogenetic and molecular genetic approaches have been used to identify genomic alterations in different histologic tumor types. Translation of these advances from basic science to clinical application is currently underway. Goals for the future include the development of more efficacious treatment strategies while simultaneously lessening toxicity. The most important cytogenetic and molecular genetic abnormalities documented to date together with their potential prognostic significance are reviewed.


Subject(s)
Aneuploidy , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Chromosome Aberrations , Neuroectodermal Tumors, Primitive/genetics , Brain Neoplasms/classification , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Child , Gene Deletion , Genes, Tumor Suppressor , Goals , Growth Substances/genetics , Humans , Loss of Heterozygosity , Medulloblastoma/genetics , Neuroectodermal Tumors, Primitive/classification , Neuroectodermal Tumors, Primitive/mortality , Neuroectodermal Tumors, Primitive/pathology , Prognosis , Receptors, Growth Factor/genetics , Supratentorial Neoplasms/genetics , Supratentorial Neoplasms/pathology
12.
Int J Radiat Oncol Biol Phys ; 45(1): 127-35, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10477016

ABSTRACT

PURPOSE: To conduct a Phase II study to evaluate the long-term efficacy and safety of high-dose 5'-bromodeoxyuridine (BrdU) and accelerated radiotherapy followed by procarbazine, lomustine (CCNU), and vincristine (PCV) chemotherapy in patients with glioblastoma multiforme. METHODS AND MATERIALS: Between 1994 and 1996, 88 patients were enrolled to receive 1.9 Gy of radiation three times a day for two 5-day cycles separated by 2 weeks; each 5-day cycle was preceded by a continuous 96-hour infusion of BrdU at a dose of 2.1 g/m2/day. After radiotherapy, patients received PCV chemotherapy. RESULTS: Median survival for all 88 patients was 50 weeks. Seventy (79.5 %) received one or more courses of PCV; their median survival was 57 weeks. Covariates predictive of improved survival were gross total versus subtotal resection or biopsy (p = 0.0048) and radiation dose > or = 56 Gy (p = 0.019). While receiving BrdU, 47 patients (53%) suffered grade 3 or 4 thrombocytopenia or leukopenia; 22 patients (25%) suffered grade 3 or 4 dermatologic toxicity. CONCLUSION: Survival was not extended in patients with glioblastoma or gliosarcoma who received BrdU at the dose and administration schedule used in this study. The BrdU dose used in this study resulted in substantial myelosuppressive and dermatologic toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Radiation-Sensitizing Agents/administration & dosage , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Bromodeoxyuridine/administration & dosage , Bromodeoxyuridine/adverse effects , Cognition/drug effects , Cognition/radiation effects , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Female , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Lomustine/administration & dosage , Lomustine/adverse effects , Male , Middle Aged , Procarbazine/administration & dosage , Procarbazine/adverse effects , Radiation-Sensitizing Agents/adverse effects , Reoperation , Vincristine/administration & dosage , Vincristine/adverse effects
13.
Cancer Res ; 59(17): 4228-32, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485462

ABSTRACT

We carried out a gene expression profiling study using cDNA array technology with 24 primary glioma tissues of low-grade (oligodendroglioma), intermediate-grade (anaplastic oligodendroglioma and anaplastic astrocytoma), and high-grade (glioblastomas multiforme) tumors and found that insulin-like growth factor binding protein 2 (IGFBP2) was consistently overexpressed only in glioblastoma multiforme. The cDNA array results were confirmed by Northern and Western blotting. The fact that the IGFBP2 gene, which is normally expressed in fetal cells and turned off in adult cells, becomes reactivated in the most advanced stage of glioma suggests that glioma progression is a result of dedifferentiation or results from a block of differentiation. Identification of IGFBP2 as a gene associated with glioma progression demonstrates the power and utility of high-throughput gene expression profiling in cancer gene discovery.


Subject(s)
Gene Expression Regulation, Enzymologic , Glioblastoma/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Humans , RNA, Messenger/analysis
14.
Carcinogenesis ; 20(5): 811-5, 1999 May.
Article in English | MEDLINE | ID: mdl-10334198

ABSTRACT

Brain tumors exhibit considerable chromosome instability (CIN), suggesting that genetic susceptibility may contribute to brain tumorigenesis. To test this hypothesis, in this pilot study, we examined for CIN in short-term lymphocyte cultures from 25 adult glioma patients and 28 age-, sex- and ethnicity-matched healthy controls (all Caucasian). We evaluated CIN by a multicolor fluorescence in situ hybridization assay using two probes: a classic satellite probe for a large heterochromatin breakage-prone region of chromosome 1 and an alpha satellite probe for a smaller region adjacent to the heterochromatin probe. Our results showed a significant increase in the mean number of spontaneous breaks per 1000 cells in glioma patients (mean +/- SD, 2.4+/-0.8) compared with controls (1.4+/-0.9; P < 0.001). By using the median number of breaks per 1000 cells in the controls as the cutoff value, we observed a crude odds ratio (OR) of 8.5 [95% confidence interval (CI) = 2.05-34.9, P < 0.001] for spontaneous breaks and brain tumor risk. After adjustment for age, sex and smoking status, the adjusted OR was 15.3 (95% CI, 2.71-87.8). A significant increase in cells with chromosome 1 aneuploidy (in the form of hyperdiploidy) (P < 0.001) was also observed in the glioma cases, with an adjusted OR of 6.6 (95% CI = 1.5-30, P < 0.05). These findings suggest that CIN can be detected in the peripheral blood lymphocytes of brain tumor patients and may be a marker for identifying individuals at risk.


Subject(s)
Chromosome Aberrations , Glioma/genetics , Lymphocytes/metabolism , Aneuploidy , Chromosome Breakage , Chromosomes, Human, Pair 1/genetics , Female , Glioma/blood , Humans , In Situ Hybridization, Fluorescence , Lymphocytes/cytology , Male , Middle Aged , Risk Factors
15.
Neuro Oncol ; 1(1): 14-25, 1999 01.
Article in English | MEDLINE | ID: mdl-11554386

ABSTRACT

The Central Brain Tumor Registry of the United States (CBTRUS) obtained 5 years of incidence data (1990-1994)--including reports on all primary brain and CNS tumors--from 11 collaborating state cancer registries. Data were available for 20,765 tumors located in the brain, meninges, and other CNS sites, including the pituitary and pineal glands. The average annual incidence was estimated at 11.5 cases per 100,000 person-years. The higher incidence of tumors in male patients (12.1 per 100,000 person-years) than in female patients (11.0 per 100,000 person-years) was statistically significant (P < 0.05); the higher incidence in whites (11.6 per 100,000 person-years) compared with blacks (7.8 per 100,000 person-years) was statistically significant (P < 0.05). The most frequently reported histologies were meningiomas (24.0%) and glioblastomas (22.6%). Higher rates for glioblastomas, anaplastic astrocytomas, oligodendrogliomas, anaplastic oligodendrogliomas, ependymomas, mixed gliomas, astrocytomas not otherwise specified, medulloblastomas, lymphomas, and germ cell tumors in male than in female patients were statistically significant (P < 0.05), with relative risks (RR) ranging from 1.3 to 3.4. Meningiomas were the only tumors with a significant excess in females (RR = 0.5). We noted higher occurrence rates in whites than in blacks for the following histologies: diffuse astrocytomas, anaplastic astrocytomas, glioblastomas, oligodendrogliomas, ependymomas, mixed gliomas, astrocytomas NOS, medulloblastomas, nerve sheath tumors, hemangioblastomas, and germ cell tumors, with RRs ranging from 1.5 to 3.4. Racial differences in occurrence rates were not observed for predominately benign meningiomas or pituitary tumors. This study represents the largest compilation of data on primary brain and CNS tumors in the United States. Standard reporting definitions and practices must be universally adopted to improve the quality and use of cancer registry data.


Subject(s)
Brain Neoplasms/epidemiology , Spinal Cord Neoplasms/epidemiology , Adolescent , Adult , Age Distribution , Aged , Aged, 80 and over , Child , Child, Preschool , Cranial Nerve Neoplasms/epidemiology , Female , Germinoma/epidemiology , Humans , Incidence , Infant , Infant, Newborn , Lymphoma/epidemiology , Male , Meningeal Neoplasms/epidemiology , Meningioma/epidemiology , Middle Aged , Pinealoma/epidemiology , Pituitary Neoplasms/epidemiology , Racial Groups , Registries , Risk , Sex Distribution , United States
16.
J Neurosurg ; 89(4): 547-51, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9761047

ABSTRACT

OBJECT: In this report the authors describe the epidemiology of craniopharyngioma. METHODS: The incidence of craniopharyngioma in the United States was estimated from two population-based cancer registries that include brain tumors of benign and borderline malignancy: the Central Brain Tumor Registry of the United States (CBTRUS) and the Los Angeles county Cancer Surveillance Program. Information on additional pediatric tumors was available from the Greater Delaware Valley Pediatric Tumor Registry (GDVPTR). The overall incidence of craniopharyngioma was 0.13 per 100,000 person years and did not vary by gender or race. A bimodal distribution by age was noted with peak incidence rates in children (aged 5-14 years) and among older adults (aged 65-74 years in CBTRUS and 50-74 years in Los Angeles county). Survival information was available from GDVPTR and the National Cancer Data Base (NCDB), a hospital-based reporting system. In the NCDB, the 5-year survival rate was 80% and decreased with older age at diagnosis. Survival is higher among children and has improved in recent years. CONCLUSIONS: Craniopharyngioma is a rare brain tumor of uncertain behavior that occurs at a rate of 1.3 per million person years. Approximately 338 cases of this disease are expected to occur annually in the United States, with 96 occurring in children from 0 to 14 years of age.


Subject(s)
Craniopharyngioma/epidemiology , Pituitary Neoplasms/epidemiology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/epidemiology , Child , Child, Preschool , Databases as Topic , Delaware/epidemiology , Female , Humans , Incidence , Infant , Los Angeles/epidemiology , Male , Middle Aged , Population Surveillance , Racial Groups , Registries , Sex Factors , Survival Rate , United States/epidemiology
17.
Endocrine ; 8(1): 7-12, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9666339

ABSTRACT

ATP-sensitive K+ (KATP) channels modulated by sulfonylurea compounds have been previously identified in the anterior pituitary of the rat and have been demonstrated to influence GH release. Recently, a sulfonylurea receptor (SUR) has been cloned from an islet cell tumor and identified as a member of the ATP binding cassette superfamily capable to coupling with inwardly rectifying potassium channels. To determine if the same receptor is expressed in pituitary tumors, SUR mRNA levels were measured in 28 human macroadenoma specimens using an RNase protection assay. All immunonegative, corticotrophin (ACTH), growth hormone (GH), and GH/prolactin (GH/Prl) immunostaining tumors expressed detectable amounts of SUR message. Among these tumors, only the GH and GH/ Prl adenomas were functional. Of the tumors immunostaining for luteinizing hormone (LH), follicle-stimulating hormone (FSH), or both, SUR mRNA was present in small amounts in 5/11. Only 1/3 Prl immunostaining tumors contained SUR mRNA. In summary, we have demonstrated that SUR mRNA expression is common in several types of silent pituitary adenomas and in functional tumors that secrete GH. Lower levels are seen in some gonadotrophin immunostaining tumors.


Subject(s)
ATP-Binding Cassette Transporters , Adenoma/genetics , Gene Expression , Pituitary Neoplasms/genetics , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , RNA, Messenger/analysis , Receptors, Drug/genetics , Adenoma/chemistry , Adrenocorticotropic Hormone/analysis , Follicle Stimulating Hormone/analysis , Human Growth Hormone/analysis , Humans , Immunohistochemistry , Luteinizing Hormone/analysis , Microscopy, Electron , Pituitary Neoplasms/chemistry , Prolactin/analysis , RNA Probes , RNA, Antisense , Ribonucleases , Sulfonylurea Receptors
18.
Genes Chromosomes Cancer ; 22(3): 165-70, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9624527

ABSTRACT

We employed the arbitrarily primed polymerase chain reaction (AP-PCR), which is a PCR-based genomic fingerprinting technique, to detect novel genetic alterations in human astrocytomas. DNA fingerprints generated by arbitrary primers were compared in normal lymphocytes and tumor tissues from the same individuals. We cloned and sequenced an AP-PCR band showing a greatly decreased intensity in tumor tissue DNA, relative to normal. Southern blot showed that this sequence was homozygously deleted in the tumor cell genome. Semiquantitative PCR analysis further showed significant decreases of signals in seven of 24 tumors, consistent with homozygous deletion of this sequence. The deleted sequence was localized to chromosome region 6p21.1 by the fluorescence in situ hybridization method. Microsatellite analysis also showed frequent allelic loss of this locus compared to neighboring loci in astrocytoma tissues. These results suggest the presence of a novel tumor suppressor gene at the 6p21.1 locus.


Subject(s)
Alleles , Astrocytoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 6/genetics , Polymerase Chain Reaction/methods , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Exons , Genetic Markers , Glioma/genetics , Humans , Microsatellite Repeats
19.
Cancer Epidemiol Biomarkers Prev ; 7(6): 553-5, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9641501

ABSTRACT

The role of genetic polymorphisms in modulating susceptibility to carcinogenic exposures has been well explored for tobacco-related neoplasms but not for other neoplasms including gliomas. It is relevant to explore these polymorphisms because certain carcinogenic exposures such as nitrosamines are implicated in the risk of gliomas. We therefore conducted a pilot case-control study to examine the role of polymorphisms in GSTM1, GSTT1, NAT2 (rapid, intermediate, and slow acetylation), and CYP1A1 and risk of glioma. Ninety patients diagnosed with glioma were ascertained as part of an ongoing genetic epidemiological study and were age, gender, and race matched with 90 healthy controls. We used PCR based methodology to determine the prevalence of the above genetic polymorphisms using sequences and PCR conditions directly adapted from studies reported previously. We calculated univariate odds ratios and performed multiple logistic regression to assess interactions between polymorphisms. We found no statistically significant associations between the null genotypes of GSTM1 and GSTT1, and CYP1A1 and risk of gliomas. However, there was an intriguing pattern with NAT2 acetylation status (odds ratios, 1.81, 1.34, and 0.61 for rapid, intermediate, and slow acetylation, respectively; P = 0.10 for trend). It is unlikely that any single polymorphism is sufficiently predictive of risk, and a panel of markers integrated with epidemiological data should be conducted on a large number of study subjects to fully understand the role of genetic polymorphisms and brain tumor risk.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Brain Neoplasms/enzymology , Cytochrome P-450 CYP1A1/genetics , Glioma/enzymology , Glutathione Transferase/genetics , Adult , Brain Neoplasms/genetics , Case-Control Studies , DNA Primers , Female , Glioma/genetics , Humans , Male , Middle Aged , Odds Ratio , Pilot Projects , Polymerase Chain Reaction , Polymorphism, Genetic , Prevalence , Risk , Risk Factors , Texas
20.
J Am Vet Med Assoc ; 212(10): 1572-5, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9604026

ABSTRACT

OBJECTIVE: To determine effect of time of sample collection on serum thyrotropin (canine thyroid-stimulating hormone [cTSH]) concentrations in euthyroid and hypothyroid dogs. DESIGN: Prospective study. ANIMALS: 6 healthy adult euthyroid dogs, 6 adult Beagles with 131iodine-induced hypothyroidism before and during administration of levothyroxine sodium, and 6 adult dogs with naturally developing hypothyroidism. PROCEDURE: Healthy euthyroid dogs were identified. Hypothyroidism was induced by administration of 131sodium iodide and confirmed by thyroid-stimulating hormone testing. These dogs then received levothyroxine for 30 days. Naturally developing hypothyroidism was diagnosed on the basis of clinical signs, low serum thyroxine (T4) concentrations, and high cTSH concentrations or abnormal results on a thyrotropin-releasing hormone response test. Samples for measurement of cTSH and T4 concentrations were obtained at 2-hour intervals from 8 AM to 8 PM. RESULTS: Mean (+/- SD) serum cTSH concentrations for healthy dogs, dogs with induced hypothyroidism before and during treatment, and dogs with naturally developing hypothyroidism were 0.11 +/- 0.08, 3.31 +/- 1.30, 0.08 +/- 0.07, and 0.55 +/- 0.27 ng/ml, respectively. Diurnal variation in cTSH concentrations was not detected. Clinically important random fluctuations in cTSH concentrations were detected for dogs with naturally developing hypothyroidism. CLINICAL IMPLICATIONS: Sample collection time does not appear to predictably influence cTSH concentrations; however, dogs with naturally developing hypothyroidism may have random fluctuations in cTSH concentrations.


Subject(s)
Blood Specimen Collection/veterinary , Dog Diseases/blood , Dogs/blood , Hypothyroidism/veterinary , Thyrotropin/blood , Animals , Circadian Rhythm , Dog Diseases/drug therapy , Dog Diseases/etiology , Hypothyroidism/blood , Hypothyroidism/drug therapy , Iodine Radioisotopes/adverse effects , Male , Reference Values , Thyrotropin-Releasing Hormone , Thyroxine/blood , Thyroxine/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...