Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cancer Res ; 66(17): 8722-30, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951188

ABSTRACT

Malignant gliomas are highly proliferative and angiogenic cancers resistant to conventional therapies. Although RAS and RAF mutations are uncommon in gliomas, RAS activity is increased in gliomas. Additionally, vascular endothelial growth factor and its cognate receptors are highly expressed in gliomas. We now report that AAL881, a novel low-molecular weight inhibitor of the kinase activities associated with B-RAF, C-RAF (RAF-1), and VEGF receptor-2 (VEGFR2), showed activity against glioma cell lines and xenografts. In culture, AAL881 inhibited the downstream effectors of RAF in a concentration-dependent manner, with inhibition of proliferation associated with a G(1) cell cycle arrest, induction of apoptosis, and decreased colony formation. AAL881 decreased the proliferation of bovine aortic endothelial cells as well as the tumor cell secretion of vascular endothelial growth factor and inhibited the invasion of glioma cells through an artificial extracellular matrix. Orally administered AAL881 was well tolerated with minimal weight loss in non-tumor-bearing mice. Established s.c. human malignant glioma xenografts grown in immunocompromised mice treated with a 10-day course of oral AAL881 exhibited growth delays relative to control tumors, frequently resulting in long-term complete regressions. AAL881 treatment extended the survival of immunocompromised mice bearing orthotopic glioma xenografts compared with placebo controls. The intraparenchymal portions of orthotopic AAL881-treated tumors underwent widespread necrosis consistent with vascular disruption compared with the subarachnoid elements. These effects are distinct from our prior experience with VEGFR2 inhibitors, suggesting that targeting RAF itself or in combination with VEGFR2 induces profound tumor responses in gliomas and may serve as a novel therapeutic approach in patients with malignant gliomas.


Subject(s)
Cell Division/drug effects , Glioma/pathology , Isoquinolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Aorta , Biopsy , Cattle , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glioma/drug therapy , Humans , raf Kinases/genetics
2.
Clin Cancer Res ; 11(22): 8145-57, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16299247

ABSTRACT

PURPOSE: Primary central nervous system (CNS) tumors represent a diverse group of tumor types with heterogeneous molecular mechanisms that underlie their formation and maintenance. CNS tumors depend on angiogenesis and often display increased activity of ErbB-associated pathways. Current nonspecific therapies frequently have poor efficacy in many of these tumor types, so there is a pressing need for the development of novel targeted therapies. EXPERIMENTAL DESIGN: ZD6474 is a novel, orally available low molecular weight inhibitor of the kinase activities associated with vascular endothelial growth factor receptor-2 and epidermal growth factor receptor. We hypothesized that ZD6474 may provide benefit in the treatment of several CNS tumor types. RESULTS: In mice bearing established s.c. tumor xenografts of CNS tumors (malignant glioma and ependymoma) or rhabdomyosarcoma, a limited course of ZD6474 treatment produced significant tumor growth delays and a high rate of partial tumor regression in most models examined. Mice with i.c. malignant glioma xenografts treated with ZD6474 experienced a significant prolongation of survival. Tumors from mice treated with ZD6474 displayed a lower proliferative index and disrupted tumor vascularity. Notably, some of these models are insensitive to low molecular weight kinase inhibitors targeting only vascular endothelial growth factor receptor-2 or epidermal growth factor receptor functions, suggesting that the combined disruption of both epidermal growth factor receptor and vascular endothelial growth factor receptor-2 activities may significantly increase tumor control. CONCLUSIONS: In conclusion, ZD6474 shows significant activity against xenograft models of several primary human CNS tumor types. Consideration for clinical development in this disease setting seems warranted.


Subject(s)
Central Nervous System Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Piperidines/pharmacology , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Dose-Response Relationship, Drug , Ependymoma/drug therapy , Ependymoma/pathology , ErbB Receptors/metabolism , Glioma/drug therapy , Glioma/pathology , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Phosphorylation/drug effects , Receptors, Vascular Endothelial Growth Factor/metabolism , Xenograft Model Antitumor Assays
3.
Cancer ; 104(8): 1693-9, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16116609

ABSTRACT

BACKGROUND: Allelic losses of 1p and 19q arms correlate with the oligodendroglial phenotype as well as with sensitivity to radiotherapy and chemotherapy. Furthermore, the DNA repair gene, methylguanine methyltransferase (MGMT), is diminished in 80% of oligodendroglial tumors and represents a possible mechanism for this therapeutic sensitivity. However, the authors questioned the relevance of genetic testing and measuring MGMT levels in tumors that were diagnostic of oligodendroglioma. METHODS: The authors performed a retrospective analysis of 1p, 19q, 9p21, TP53, and MGMT status in 46 patients with oligodendrogliomas to address any relations that may exist among these markers with regard to progression-free survival (PFS) and total survival. Methodologies included comparative genomic hybridization; loss of heterozygosity (LOH) on 1p, 19q, and 9p21; TP53 mutational analysis; and immunohistochemistry for MGMT. RESULTS: The authors found that survival among patients with light microscopically diagnosed oligodendroglial tumors demonstrating LOH of 1p and 19q trended toward statistical significance (P = 0.102 and P = 0.058, respectively). 9p21 LOH was significant as a predictor of PFS only among anaplastic oligodendrogliomas in this cohort (P = 0.033). TP53 mutation was found to be significantly predictive of a shorter survival (P = 0.027) among all patients and exhibited a strong trend toward a shorter PFS (P = 0.060). Low-level MGMT labeling index (LI) (< 20%) was noted in 86% of all oligodendroglial tumors. MGMT LI was not found to correlate with an improved PFS or total survival in this cohort, recognizing that median survival was not reached after a median follow-up of 104 months. CONCLUSIONS: 9p21 and TP53 mutational status assisted in developing a stricter subclassification of these tumors with prognostic significance. MGMT levels were decreased in a majority of oligodendrogliomas.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/mortality , O(6)-Methylguanine-DNA Methyltransferase/analysis , Oligodendroglioma/mortality , Tumor Suppressor Protein p53/analysis , Brain Neoplasms/chemistry , Chromosomes, Human/genetics , Cohort Studies , Disease-Free Survival , Humans , In Situ Hybridization , Loss of Heterozygosity , Neoplasm Proteins , Oligodendroglioma/chemistry , Prognosis , Retrospective Studies , Survival Rate
4.
Cancer Res ; 65(10): 4051-8, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899794

ABSTRACT

Despite the strikingly grave prognosis for older patients with glioblastomas, significant variability in patient outcome is experienced. To explore the potential for developing improved prognostic capabilities based on the elucidation of potential biological relationships, we did analyses of genes commonly mutated, amplified, or deleted in glioblastomas and DNA microarray gene expression data from tumors of glioblastoma patients of age >50 for whom survival is known. No prognostic significance was associated with genetic changes in epidermal growth factor receptor (amplified in 17 of 41 patients), TP53 (mutated in 11 of 41 patients), p16INK4A (deleted in 15 of 33 patients), or phosphatase and tensin homologue (mutated in 15 of 41 patients). Statistical analysis of the gene expression data in connection with survival involved exploration of regression models on small subsets of genes, based on computational search over multiple regression models with cross-validation to assess predictive validity. The analysis generated a set of regression models that, when weighted and combined according to posterior probabilities implied by the statistical analysis, identify patterns in expression of a small subset of genes that are associated with survival and have value in assessing survival risks. The dominant genes across such multiple regression models involve three key genes-SPARC (Osteonectin), Doublecortex, and Semaphorin3B-which play key roles in cellular migration processes. Additional analysis, based on statistical graphical association models constructed using similar computational analysis methods, reveals other genes which support the view that multiple mediators of tumor invasion may be important prognostic factor in glioblastomas in older patients.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Aged , Brain Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Doublecortin Domain Proteins , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Gene Expression Profiling , Genetic Markers/genetics , Glioblastoma/metabolism , Humans , Loss of Heterozygosity , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Middle Aged , Neuropeptides/biosynthesis , Neuropeptides/genetics , Osteonectin/biosynthesis , Osteonectin/genetics , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/biosynthesis , Phosphoric Monoester Hydrolases/genetics , Reproducibility of Results , Semaphorins , Survival Rate , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
6.
J Clin Oncol ; 22(1): 133-42, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14638850

ABSTRACT

PURPOSE: To evaluate the efficacy and tolerability of gefitinib (ZD1839, Iressa; AstraZeneca, Wilmington, DE), a novel epidermal growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. PATIENTS AND METHODS: This was an open-label, single-center phase II trial. Fifty-seven patients with first recurrence of a glioblastoma who were previously treated with surgical resection, radiation, and usually chemotherapy underwent an open biopsy or resection at evaluation for confirmation of tumor recurrence. Each patient initially received 500 mg of gefitinib orally once daily; dose escalation to 750 mg then 1,000 mg, if a patient received enzyme-inducing antiepileptic drugs or dexamethasone, was allowed within each patient. RESULTS: Although no objective tumor responses were seen among the 53 assessable patients, only 21% of patients (11 of 53 patients) had measurable disease at treatment initiation. Seventeen percent of patients (nine of 53 patients) underwent at least six 4-week cycles, and the 6-month event-free survival (EFS) was 13% (seven of 53 patients). The median EFS time was 8.1 weeks, and the median overall survival (OS) time from treatment initiation was 39.4 weeks. Adverse events were generally mild (grade 1 or 2) and consisted mainly of skin reactions and diarrhea. Drug-related toxicities were more frequent at higher doses. Withdrawal caused by drug-related adverse events occurred in 6% of patients (three of 53 patients). Although the presence of diarrhea positively predicted favorable OS from treatment initiation, epidermal growth factor receptor expression did not correlate with either EFS or OS. CONCLUSION: Gefitinib is well tolerated and has activity in patients with recurrent glioblastoma. Further study of this agent at higher doses is warranted.


Subject(s)
Brain Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Quinazolines/therapeutic use , Adult , Aged , Brain Neoplasms/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Epidermal Growth Factor/antagonists & inhibitors , Female , Gefitinib , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/administration & dosage , Quinazolines/adverse effects , Treatment Outcome
7.
Acta Neuropathol ; 99(4): 345-51, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10787031

ABSTRACT

Loss of portions of chromosome 17p, usually through the formation of i(17qp) is a well-known finding in medulloblastomas. Loss of heterozygosity (LOH) studies, however, occasionally demonstrate loss of the more distal portions of 17p, a pattern which is more consistent with a terminal deletion. Here we use a combination of routine karyotyping, fluorescence in situ hybridization (FISH) and LOH studies on four medulloblastoma cell lines and one xenograft to demonstrate the spectrum of chromosome 17 abnormalities which occur in these tumors. Cell line D-556 Med showed a typical dicentric i(17q) and cell line D-721 Med showed two normal copies of chromosome 17 by all methods. Cell line D-425 Med showed loss of terminal 17p by LOH, while the karyotype showed what appeared to be an i(17q). FISH and chromosome 17 painting, however, demonstrated that the abnormal chromosome 17 was actually formed through an unbalanced translocation involving two copies of chromosome 17, with breakpoints at p12 and q11-1, an explanation which reconciled the cytogenetic and LOH findings. Cell line D 581 Med had a terminal deletion at 17p11.2. The finding of two cells with i(17q) in this case by interphase FISH suggests that the terminal deletion arose from breakage of an i(17q). Finally, xenograft D 690 Med showed LOH for regions distal to 17p12, whereas karyotyping, FISH using probes on 17p, and chromosome 17 painting showed two intact copies of chromosome 17. This pattern can be explained by homologous recombination. These data support the concept that the critical deletion of 17p can occur through a variety of mechanisms in the medulloblastoma. The losses may occur through typical i(17q), as well as other mechanisms such as terminal deletions, possibly through breakage of i(17q), unbalanced translocations and homologous recombination.


Subject(s)
Cerebellar Neoplasms/genetics , Chromosome Aberrations/genetics , Chromosomes, Human, Pair 17/genetics , Medulloblastoma/genetics , Adolescent , Child , Child, Preschool , Chromosome Banding , Chromosome Disorders , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Tumor Cells, Cultured
8.
Am J Clin Pathol ; 113(1): 135-40, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10631867

ABSTRACT

Several methods have been used to evaluate engraftment after allogeneic bone marrow transplantation (BMT). We assessed the usefulness of a multiple short tandem repeat (STR) amplification kit combined with a capillary electrophoresis unit for DNA identity analysis in the evaluation of engraftment after BMT. For 17 of 18 patients, at least 1 locus showed unique alleles for the donor and the recipient. In all cases, at least 1 locus was informative for the presence of small amounts of recipient DNA. The results from STR analysis were the same as Southern blot analysis in 14 of 17 cases. Differences included mixed chimerism detected only with STR analysis, informative loci present only with STR analysis, and informative loci present only with Southern blot analysis (1 case each). By using mock mixed chimeras, minor populations of 5% were detected routinely in all loci using the kit manufacturer's default protocol. By increasing the amount of amplified DNA, minor populations of 1% were detected in all cases but not in all loci. This single reaction technique provides for faster results, reduced workforce needs, and greater sensitivity than traditional Southern blot.


Subject(s)
Bone Marrow Transplantation , Graft Survival , Hematologic Diseases/therapy , Adolescent , Adult , Blotting, Southern , Child , Child, Preschool , DNA/analysis , DNA Fingerprinting/methods , Electrophoresis, Capillary/methods , Evaluation Studies as Topic , Female , Graft Survival/genetics , Humans , Infant , Male , Minisatellite Repeats/genetics , Polymerase Chain Reaction/methods , Polymorphism, Restriction Fragment Length , Reagent Kits, Diagnostic , Reproducibility of Results , Sensitivity and Specificity , Transplantation, Homologous
9.
Neuro Oncol ; 2(3): 164-73, 2000 07.
Article in English | MEDLINE | ID: mdl-11302337

ABSTRACT

Cytogenetic and molecular genetic studies of glioblastoma multiforme (GBM) have shown that the most frequent alterations are gains of chromosome 7, losses of 9p loci and chromosome 10, and gene amplification, primarily of the epidermal growth factor receptor (EGFR) gene. Although this profile is potentially useful in distinguishing GBM from other tumor types, the techniques used tend to be labor intensive, and some can detect only gains or losses of genetic loci. Comparative genomic hybridization (CGH) is a powerful technique capable of identifying both gains and losses of DNA sequences. The present study compares the CGH evaluation of 22 GBM with classic cytogenetics, loss of heterozygosity by allelotyping, and gene amplification by Southern blot analysis to determine the reliability of CGH in the genetic characterization of GBM. The CGH and karyotypic data were consistent in showing gain of chromosome 7 accompanied by a loss of chromosome 10 as the most frequent abnormality, followed by a loss of 9p in 17 of 22 GBM cases. Loss of heterozygosity of chromosomes 10 (19/22) and 9p (9/22) loci confirmed the underrepresentation by CGH. Genomic amplifications were observed by CGH in 5 of the 10 cases where gene amplification was detected by Southern blot analysis. The data show that CGH is equally reliable, compared with the more established genetic methods, for recognizing the prominent genetic alterations associated with GBM and support its use as a plausible adjunct to glioma classification.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Profiling , Glioblastoma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/classification , Brain Neoplasms/diagnosis , Female , Glioblastoma/classification , Glioblastoma/diagnosis , Humans , Karyotyping , Male , Middle Aged , Molecular Biology , Nucleic Acid Hybridization
10.
Am J Pathol ; 155(2): 375-86, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433931

ABSTRACT

Oligodendroglial neoplasms are a subgroup of gliomas with distinctive morphological characteristics. In the present study we have evaluated a series of these tumors to define their molecular profiles and to determine whether there is a relationship between molecular genetic parameters and histological pattern in this tumor type. Loss of heterozygosity (LOH) for 1p and 19q was seen in 17/23 (74%) well-differentiated oligodendrogliomas, in 18/23 (83%) anaplastic oligodendrogliomas, and in 3/8 (38%) oligoastrocytomas grades II and III. LOH for 17p and/or mutations of the TP53 gene occurred in 14 of these 55 tumors. Only one of the 14 cases with 17p LOH/TP53 gene mutation also had LOH for 1p and 19q, and significant astrocytic elements were seen histologically in the majority of these 14 tumors. LOH for 9p and/or deletion of the CDKN2A gene occurred in 15 of these 55 tumors, and 11 of these cases were among the 24 (42%) anaplastic oligodendrogliomas. Comparative genomic hybridization (CGH) identified the majority of cases with 1p and 19q loss and, in addition, showed frequent loss of chromosomes 4, 14, 15, and 18. These findings demonstrate that oligodendroglial neoplasms usually have loss of 1p and 19q whereas astrocytomas of the progressive type frequently contain mutations of the TP53 gene, and that 9p loss and CDKN2A deletions are associated with progression from well-differentiated to anaplastic oligodendrogliomas.


Subject(s)
Brain Neoplasms/genetics , Loss of Heterozygosity , Nucleic Acid Hybridization/methods , Oligodendroglioma/genetics , Tumor Suppressor Proteins , Adolescent , Adult , Astrocytoma/genetics , Brain Neoplasms/pathology , Child , Chromosome Deletion , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 4 , Female , Genes, p16/genetics , Genes, p53/genetics , Glioblastoma/genetics , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Oligodendroglioma/pathology , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/genetics , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Y Chromosome
11.
Curr Opin Oncol ; 11(3): 162-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10328589

ABSTRACT

De novo glioblastomas develop in older patients without prior clinical history of less malignant tumors. Progressive glioblastomas are common among younger patients and arise through progression from lower-grade astrocytomas. CDKN2A deletions, PTEN alterations, and EGFR amplification are more prevalent among de novo glioblastomas, whereas p53 mutations are more common among progressive glioblastomas. Loss of heterozygosity (LOH) for chromosome 10 is seen uniformly among both de novo and progressive high-grade astrocytomas. The inactivation of the PTEN gene is found in approximately 30% to 40% of astrocytomas with chromosome 10 loss, and LOH pattern in the remaining astrocytomas strongly supports the presence of another yet unidentified tumor suppressor gene telomeric to PTEN. More than 80% of oligodendrogliomas exhibit LOH for 1 p and 19q alleles. Oligoastrocytomas with 1p/19q LOH are related to oligodendrogliomas, and those with p53 mutations are related to astrocytomas.


Subject(s)
Brain Neoplasms/genetics , Chromosome Mapping , Glioma/genetics , Mutation , Tumor Suppressor Proteins , Astrocytoma/genetics , Chromosomes, Human, Pair 10 , Gene Amplification , Gene Deletion , Genes, Tumor Suppressor , Glioblastoma/genetics , Humans , Loss of Heterozygosity , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/genetics
12.
Neuro Oncol ; 1(1): 52-60, 1999 01.
Article in English | MEDLINE | ID: mdl-11550302

ABSTRACT

Morphologic criteria for diagnosing oligodendrogliomas and for classifying them as well-differentiated (World Health Organization grade II) and anaplastic (World Health Organization grade III) are well recognized. Nevertheless, applying these guidelines to specific cases often reveals discrepancies among different observers. In addition, whether a given tumor also contains an astrocytic component may be debatable. Loss of heterozygosity studies have demonstrated that oligodendroglial neoplasms have a high incidence of loss of the 1p and 19q chromosomal arms. Although loss of heterozygosity for portions of 19q are sometimes seen in astrocytic neoplasms, these tumors seldom show complete loss of 19q accompanied by loss of 1p. Loss of 9p or homozygous deletion of the CDKN2 gene or both are associated with anaplastic oligodendrogliomas, whereas loss of 17p or TP53 gene mutations or both are frequent in astrocytomas, but rare in oligodendrogliomas. These observations suggest that molecular genetic parameters could provide an objective, reproducible framework for classifying oligodendroglial neoplasms.


Subject(s)
Brain Neoplasms/pathology , Oligodendroglioma/pathology , Astrocytoma/classification , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/classification , Brain Neoplasms/genetics , Chromosomes, Human/genetics , Chromosomes, Human/ultrastructure , Cyclin-Dependent Kinase Inhibitor p16/physiology , Genes, Tumor Suppressor , Genes, p16 , Genes, p53 , Glioma/classification , Glioma/genetics , Glioma/pathology , Humans , Karyotyping , Loss of Heterozygosity , Nucleic Acid Hybridization , Oligodendroglioma/classification , Oligodendroglioma/genetics , Tumor Suppressor Protein p53/physiology , World Health Organization
13.
Cancer Res ; 57(19): 4187-90, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9331072

ABSTRACT

The PTEN gene, located on 10q23, has recently been implicated as a candidate tumor suppressor gene in brain, breast and prostate tumors. In the present study, 123 brain tumors, including various grades and histological types of gliomas occurring in children and adults, were analyzed for PTEN mutations by SSCP assay and sequencing. Mutations in the PTEN gene were found in 13 of 42 adult glioblastomas and 3 of 13 adult anaplastic astrocytomas, whereas none of the 21 low-grade adult gliomas or the 22 childhood gliomas of all grades showed mutations. The single medulloblastoma with a mutation was a recurrent tumor that also possessed a p53 mutation. High-grade adult gliomas with PTEN mutations included cases that also contained gene amplification or p53 gene mutations, as well as cases that did not contain either of these abnormalities. There was no obvious relationship between presence of PTEN mutation and survival; however, there was a tendency for PTEN mutations to occur in older age group patients. This analysis suggest that PTEN gene mutations are restricted to high-grade adult gliomas and that this abnormality is independent of the presence or absence of gene amplification or p53 gene mutation in these tumors.


Subject(s)
Brain Neoplasms/genetics , Genes, Tumor Suppressor , Glioma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Child , DNA Mutational Analysis , DNA, Neoplasm/genetics , Disease Progression , Female , Genes, p53 , Glioma/pathology , Humans , Loss of Heterozygosity , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Middle Aged , Polymorphism, Single-Stranded Conformational , Sequence Deletion
14.
Cancer Res ; 57(14): 2933-6, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230204

ABSTRACT

A methylator-resistant human glioblastoma multiforme xenograft, D-245 MG (PR), in athymic nude mice was established by serially treating the parent xenograft D-245 MG with procarbazine. D-245 MG xenografts were sensitive to procarbazine, temozolomide, N-methyl-N-nitrosourea, 1,3-bis(2-chloroethyl)-1-nitrosourea, 9-aminocamptothecin, topotecan, CPT-11, cyclophosphamide, and busulfan. D-245 MG (PR) xenografts were resistant to procarbazine, temozolomide, N-methyl-N-nitrosourea, and busulfan, but they were sensitive to the other agents. Both D-245 MG and D-245 MG (PR) xenografts displayed no O6-alkylguanine-DNA alkyltransferase activity, and their levels of glutathione and glutathione-S-transferase were similar. D-245 MG xenografts expressed the human mismatch repair proteins hMSH2 and hMLH1, whereas D-245 MG (PR) expressed hMLH1 but not hMSH2.


Subject(s)
DNA Methylation , DNA Repair , Glioblastoma/drug therapy , Animals , Drug Resistance , Glioblastoma/genetics , Humans , Melanocyte-Stimulating Hormones/analysis , Methyltransferases/metabolism , Mice , Mice, Inbred BALB C , Microsatellite Repeats , Neoplasm Transplantation , O(6)-Methylguanine-DNA Methyltransferase , Transplantation, Heterologous , Tumor Cells, Cultured
15.
J Neuropathol Exp Neurol ; 55(5): 540-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8627345

ABSTRACT

Childhood ependymomas exhibit epidemiologic, anatomic, histologic, and biologic features and distinguish them from other gliomas. Because of their propensity to grow in functionally sensitive regions of the brain, adequate tumor sampling for basic and therapeutic research is limited. We have established xenografts in both subcutaneous and intracranial nude mouse systems (D528 EP-X, D612 EP-X) from the ependymomas of two nonrelated children. Median subcutaneous growth rates (reported in days to grow from 200 mm3 to 1000 mm3) are 82 days for D528 EP-X (n = 10) and 50 days for D612 EP-X (n = 10). D528 EP-X grows intracranially with a median postimplantation survival of 85 days (n = 10); D612 EP produces a median postimplantation survival of 72.5 days (n = 10). Both xenografts grow as well-formed masses with no evidence of infiltration into either brain or subcutaneous tissues. While perivascular pseudopalisading is found in both xenografts, true ependymal rosette formation is absent. Ultrastructurally, neither xenograft exhibits cilia, but both produce abundant intermediate filaments. By light microscopy, the neoplastic cells are immunoreactive for the intermediate filaments glial fibrillary acidic protein, vimentin, and nestin. Karyotypically D528 EP exhibits 46,XX,del(6)(q22q26)/46,XX while D612 EP exhibits 50,XX, +X,t(1;8)(p11;q11),t(1;8)(p11;q11), +1,-4, der(5)t(4;5)(q12;q35), +der(5)t(4;5)(q12;q35),-6, +9, +9,-16, +der(17)t(6;17)(p11;p11), +mar. Restriction fragment length polymorphism studies comparing the primary brain tumor tissue from each patient against multiple passages of the resulting xenografts confirm the origin of both xenografts. These xenografts represent models on which future studies into the oncogenesis, progression and therapy of ependymomas can be performed.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellopontine Angle , Cerebral Ventricle Neoplasms/pathology , Ependymoma/pathology , Neoplasm Transplantation , Transplantation, Heterologous , Animals , Child, Preschool , Female , Humans , Infant , Karyotyping , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron , Nerve Tissue Proteins/genetics , Polymorphism, Restriction Fragment Length , Whole-Body Irradiation
16.
Genes Chromosomes Cancer ; 15(1): 54-63, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8824726

ABSTRACT

Loss of heterozygosity at specific chromosomal locations has been taken as evidence of a tumor suppressor gene located in that area. We performed a genomic allelotyping study on 46 childhood brain tumors of different histopathological types in order to identify and confirm common areas of deletion in different tumor types. Two hundred microsatellite DNA probes equally distributed over the 22 autosomes were applied, covering the genome in steps of approximately 25 cM. Our results confirm frequent loss of heterozygosity of chromosome arms 9q, 10q, 11p, 11q, 16q, and 22q in high-grade gliomas, medulloblastomas, and ependymomas. In addition, we found a new region of loss on chromosome segment 2p21-23 affected predominantly in high-grade gliomas and medulloblastomas.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , DNA, Satellite , Adolescent , Adult , Child , Child, Preschool , Female , Heterozygote , Humans , Infant , Male , Microsatellite Repeats , Polymerase Chain Reaction
17.
Oncogene ; 10(11): 2243-6, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7784070

ABSTRACT

The high incidence of loss of chromosome 10 alleles in glioblastoma multiforme suggests the presence on this chromosome of a tumor suppressor gene that is important in glioma tumorigenesis and progression. Our initial deletion mapping studies using restriction fragment length polymorphism markers indicated a common deletion region in 10q24-qter. In an attempt to localize the deleted region further, we screened a panel of 117 gliomas for loss of heterozygosity for chromosome 10 loci using 10 microsatellite markers. Seventeen tumors showed partial loss of a copy of chromosome 10 and were further analysed using 28 additional microsatellite markers. Of these, 10 had terminal deletion in the q arm, three had deletions in both p and q arms, two contained interstitial deletion in 10q and two carried deletions in 10p. In the 15 tumors with deletions in 10q, the minimal overlapping deletion region was in distal 10q between markers D10S587 and D10S216. Loci D10S587 and D10S216 are approximately mapped to a 5 cM region in 10q25.1.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 10 , Glioma/genetics , Heterozygote , Humans , Tumor Cells, Cultured
19.
Cancer Res ; 54(5): 1324-30, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8118823

ABSTRACT

Glial tumors of all grades and histological types from 72 adults and 48 children were analyzed for mutations of the TP53 gene, loss of heterozygosity (LOH) for 17p, and accumulation of TP53 protein to determine whether the incidence and type of TP53 alterations differ among tumors of different histological type and between tumors from adults and children. These tumors were also evaluated for LOH for chromosome 10 and for amplification of the epidermal growth factor receptor, C-MYC, N-MYC, GLI, platelet-derived growth factor receptor-alpha, and murine double minute 2 genes to determine the patterns of molecular alterations involved in the progression of these neoplasms. Seventeen of the 120 tumors contained mutations of the TP53 gene. One of the tumors with TP53 gene mutation was from one of the 48 patients less than 18 years of age. Twelve of the 17 tumors with mutations occurred among the 27 patients in the 18-45-year age group, while 4 tumors with mutations were among the 45 patients more than 45 years old. There was also an increased incidence of TP53 mutation in patients with anaplastic astrocytoma histology. However, no significant association between presence of TP53 mutation and patient survival was observed. These studies demonstrate that TP53 gene mutations are a common mechanism for glial cell neoplasms in the 18-45-year age group but are unrelated to progression and advanced histological grade. LOH for chromosome 10 and gene amplification, however, occurring in 82 and 40%, respectively, of glioblastoma multiforme, whether seen alone or along with TP53 gene alterations, are related to advanced histological grade of the tumor. In childhood gliomas, in contrast, TP53 gene alterations, LOH for 17p and 10q, and gene amplification are uncommon in tumors of all grades, suggesting that presently unknown mechanisms are responsible for the genesis and progression of these tumors.


Subject(s)
Brain Neoplasms/genetics , Genes, p53/genetics , Glioma/genetics , Mutation/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Child , Child, Preschool , Chromosome Aberrations , Chromosomes, Human, Pair 17 , Female , Gene Amplification/genetics , Gene Deletion , Glioma/mortality , Glioma/pathology , Heterozygote , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Polymerase Chain Reaction
20.
Genes Chromosomes Cancer ; 5(1): 75-82, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1384665

ABSTRACT

Cytogenetic and RFLP studies have shown that chromosome 10 is frequently lost in tumor cells from glioblastomas, suggesting that a suppressor gene important in tumorigenesis is present on this chromosome. Forty-one tumors were examined for loss of heterozygosity at 23 loci on chromosome 10 to determine the smallest common deletion interval on this chromosome. Seven tumors did not lose heterozygosity for any of the markers. Twenty-three tumors lost an allele for all the informative loci. In 11 tumors heterozygosity was maintained at some loci and lost at other loci, indicating partial deletion of chromosome 10. The common region of deletion in these 11 tumors was located in 10q24-q26 between the markers pHUK-8 and pMCT122.2.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 10/ultrastructure , Glioma/genetics , Alleles , DNA Probes , Female , Genetic Markers , Glioblastoma/genetics , Humans , Male , Polymorphism, Restriction Fragment Length
SELECTION OF CITATIONS
SEARCH DETAIL
...