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











Publication year range
1.
Int J Cancer ; 128(8): 1981-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20589679

ABSTRACT

Platelet-derived growth factor receptor (PDGFR) signaling has been implicated in the pathogenesis of glioblastomas and represents a target for the tyrosine kinase inhibitor imatinib. To examine the prognostic or predictive role of PDGFRs in recurrent glioblastomas, expression was examined in tumor samples of 101 patients of CSTI571BDE40, a randomized trial comparing hydroxyurea monotherapy and a combination of hydroxyurea and imatinib. Furthermore, PDGFRα phosphorylation was investigated using in situ proximity ligation assay. PDGFRα protein was expressed in 33% of tumors and was associated with male sex, young age, presence of R132H mutated isocitrate dehydrogenase 1 protein and short median survival (142 vs. 187 days, p = 0.028). Tumor PDGFRα phosphorylation was also associated with short survival (p = 0.030). The subset of patients with PDGFRα positive glioblastoma did not have longer survival on treatment with hydroxyurea and imatinib compared with hydroxyurea monotherapy. In conclusion, both PDGFRα protein expression and phosphorylation status had a prognostic role in recurrent glioblastomas but did not define a group that showed benefit from the combination therapy consisting of hydroxyurea and imatinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Benzamides , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hydroxyurea/administration & dosage , Imatinib Mesylate , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Phosphorylation , Piperazines/administration & dosage , Prognosis , Pyrimidines/administration & dosage , Survival Rate
2.
Brain Pathol ; 20(4): 763-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20030644

ABSTRACT

KIT receptor tyrosine kinase is expressed in tumor endothelial cells of adult glioblastomas, but its expression in pediatric brain tumor endothelial cells is unknown. We assessed expression of KIT, phosphorylated KIT, stem cell factor (SCF) and vascular endothelial growth factor receptor-2 (VEGFR-2) in 35 juvenile pilocytic astrocytomas and 49 other pediatric brain tumors using immunohistochemistry, and KIT messenger RNA (mRNA) using in situ hybridization. KIT and phospho-KIT were moderately or strongly expressed in tumor endothelia of 37% and 35% of pilocytic astrocytomas, respectively, whereas marked SCF and VEGFR-2 expression was uncommon. KIT mRNA was detected in tumor endothelial cells. Tumor endothelial cell KIT expression was strongly (P < 0.01) associated with endothelial cell phospho-KIT and SCF expression, and with tumor KIT (P = 0.0011) and VEGFR-2 expression (P = 0.022). KIT and phospho-KIT were present in endothelia of other pediatric brain tumors, notably ependymomas. Endothelial cell KIT expression was associated with a young age at diagnosis of pilocytic astrocytoma or ependymoma, and it was occasionally present in histologically normal tissue of the fetus and children. We conclude that KIT is commonly present in endothelial cells of juvenile brain tumors and thus may play a role in angiogenesis in these neoplasms.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Endothelial Cells/metabolism , Ependymoma/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Adolescent , Age Factors , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Child, Preschool , Endothelial Cells/pathology , Ependymoma/genetics , Ependymoma/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Infant , Infant, Newborn , Phosphorylation , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Statistics, Nonparametric , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
3.
Histopathology ; 55(5): 544-53, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19912360

ABSTRACT

AIMS: To assess KIT receptor tyrosine kinase and stem cell factor (SCF, KIT ligand) expression in tumour microvessel endothelial cells. METHODS AND RESULTS: KIT and SCF expression were studied in 248 human tumours consisting of 15 different histological types using immunohistochemistry and in situ hybridization for KIT messenger RNA. Moderate to strong intratumoral endothelial cell KIT expression was present in 11 of the 15 tumour types, and was most common in glioblastoma (58%), mixed embryonal carcinoma with teratoma (33%) and renal clear cell carcinoma (29%). Results of in situ hybridization were in line with those obtained with immunohistochemistry in the cases studied (n = 9). Marked SCF expression was uncommon in tumour endothelial cells, but frequent in perinecrotic tumour cells. Patients with glioblastoma with moderate to strong endothelial cell KIT expression had more favourable survival than those whose tumour showed little or no expression (P = 0.024). Glioblastoma patients whose tumour expressed SCF had an unfavourable outcome compared with those with tumour with weak or no expression (P = 0.034). CONCLUSIONS: Intratumoral microvessels of several types of human malignant tumours express KIT. Tumour cell SCF expression and absence of marked endothelial cell KIT expression are novel adverse prognostic features in glioblastoma.


Subject(s)
Biomarkers, Tumor/analysis , Endothelial Cells/metabolism , Neoplasms/metabolism , Stem Cell Factor/biosynthesis , Brain Neoplasms/metabolism , Female , Glioblastoma/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Ki-67 Antigen/biosynthesis , Male , Microvessels/metabolism , Neoplasms/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis
4.
Genes Chromosomes Cancer ; 47(2): 107-17, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17985357

ABSTRACT

Cutaneous T-cell lymphoma (CTCL) patients have an increased risk of certain secondary cancers, the most common of which are lung cancers, especially small cell lung cancer. To reveal the molecular pathogenesis underlying CTCL-associated lung cancer, we analyzed genomic aberrations in CTCL-associated and reference lung cancer samples. DNA derived from microdissected lung cancer cells of five CTCL-associated lung cancers and five reference lung cancers without CTCL association was analyzed by comparative genomic hybridization (CGH). Fluorescent in situ hybridization (FISH), immunohistochemistry (IHC), and loss of heterozygosity (LOH) analysis were performed for selected genes. In CTCL-associated lung cancer, CGH revealed chromosomal aberrations characterizing both lung cancer and CTCL, but also losses of 1p, and 19, and gains of 4q and 7, hallmarks of CTCL. LOH for the CTCL-associated NAV3 gene was detected in two of the four informative primary lung cancers. FISH revealed increased copy number of the KIT gene in 3/4 of CTCL-associated lung cancers and 1/5 of primary lung cancers. PDGFRA and VEGFR2 copy numbers were also increased. IHC showed moderate KIT expression when the gene copy number was increased. CTCL-associated lung cancer shows chromosomal aberrations different from primary lung cancer, especially amplifications of 4q, a chromosome arm frequently deleted in the latter tumor type. Copy numbers and expression of selected genes in chromosome 4 differed between CTCL-associated and reference lung cancers. These preliminary observations warrant further prospective studies to identify the common underlying factors between CTCL and CTCL-associated lung cancer.


Subject(s)
Carcinoma, Small Cell/genetics , Chromosome Aberrations , Lung Neoplasms/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Aged , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Nucleic Acid Hybridization
5.
Int J Cancer ; 121(4): 915-20, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17471559

ABSTRACT

While the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway is known to be activated in multiple sporadic cancers, the role of this pathway in familial tumors is mostly unknown. We searched for alterations in the catalytic domain of PI3K (PIK3CA), PTEN and KRAS, all of which may contribute to PI3K/AKT pathway activation, in a total of 160-familial colorectal (CRC) and endometrial carcinomas (EC), stratified by the presence vs. absence of germline mutations in DNA mismatch repair (MMR) genes. PIK3CA alterations (consisting of point mutations or low-level amplification, which were mutually exclusive with 1 exception) occurred in 10/70 (14%) of CRCs and 19/90 (21%) of ECs. Within ECs, amplification was significantly associated with the subgroup lacking germline mutations in MMR genes (familial site-specific endometrial cancer) (p = 0.015). Decreased or lost PTEN expression was characteristic of endometrial tumourigenesis (51/81, 63%, in EC compared with 24/62, 39%, in CRC, p = 0.004) and KRAS mutations of colorectal tumourigenesis (19/70, 27% in CRC vs. 9/89, 10%, in EC, p = 0.006) regardless of the MMR gene mutation status. PIK3CA alterations frequently coexisted with PTEN or KRAS changes. Combined with published studies on sporadic tumors, our data broaden the understanding of the role for PI3K pathway genes in human tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Endometrial Neoplasms/genetics , Gene Amplification , Mutation , Phosphatidylinositol 3-Kinases/genetics , Aged , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Membrane Proteins/genetics , Middle Aged , PTEN Phosphohydrolase/genetics , Polymorphism, Single-Stranded Conformational , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Reverse Transcriptase Polymerase Chain Reaction , ras Proteins/genetics
6.
Mol Cancer Res ; 4(12): 927-34, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17189383

ABSTRACT

Receptor tyrosine kinase aberrations are implicated in the genesis of gliomas. We investigated expression and amplification of KIT, PDGFRA, VEGFR2, and EGFR in 87 gliomas consisting of astrocytomas, anaplastic astrocytomas, oligodendrogliomas, or oligoastrocytomas in tumor samples collected at the time of the diagnosis and in samples of the same tumors at tumor recurrence. Gene amplifications were investigated using either chromogenic in situ hybridization or fluorescence in situ hybridization, and protein expression using immunohistochemistry. In samples collected at glioma diagnosis, KIT and PDGFRA amplifications were more frequent in anaplastic astrocytomas than in astrocytomas, oligodendrogliomas, and oligoastrocytomas [28% versus 5% (P = 0.012) and 33% versus 2% (P = 0.0008), respectively]. VEGFR2 amplifications occurred in 6% to 17% of the gliomas at diagnosis, and EGFR amplifications in 0% to 12%. Amplified KIT was more frequently present in recurrent gliomas than in newly diagnosed gliomas (P = 0.0066). KIT amplification was associated with KIT protein expression and with presence of PDGFRA and EGFR amplifications both at the time of the first glioma diagnosis and at tumor recurrence, and with VEGFR2 amplification at tumor recurrence. Three (4%) primary gliomas and 10 (14%) recurrent gliomas that were evaluable for coamplification of KIT, PDGFRA, and VEGFR2 showed amplification of at least two of these genes; the amplicon contained amplified KIT in all 13 cases. In conclusion, besides glioblastoma, amplified KIT, PDGFRA, and VEGFR may also occur in lower-grade gliomas and in their recurrent tumors. It is currently not known whether specific tyrosine kinase inhibitors are effective in the treatment of such gliomas.


Subject(s)
Brain Neoplasms/genetics , ErbB Receptors/genetics , Gene Amplification/physiology , Glioma/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression , Humans , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Acta Neuropathol ; 112(4): 483-90, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16823502

ABSTRACT

The gene for RE1-silencing transcription factor (REST) alias neuron-restrictive silencer factor NRSF, acts as a transcriptional repressor in the neuronal differentiation pathways in non-neuronal cells, and plays an important role in neuronal development. Inactivating mutations or overexpression of REST have previously been reported in various types of cancer, but no data is available for the role of REST alterations in gliomas. REST gene was screened for mutations in 161 nervous system tumors consisting of astrocytomas, glioblastomas, oligodendrogliomas, oligoastrocytomas, medulloblastomas, meningiomas and schwannomas. REST exons 1-3 were analyzed using denaturing high-performance liquid chromatography (DHPLC) and direct sequencing. The gene copy numbers of REST were investigated by chromogenic (CISH) and fluorescence in situ hybridization (FISH) techniques. Non-synonymous SNPs (P797L, P815S) were found in eight different brain tumor samples. No truncating or activating novel mutations of REST were discovered. Since REST is located at 4q12, a chromosome region implicated in brain tumorigenesis, we conducted gene copy number analyses in medulloblastomas and gliomas. The majority of gliomas (67%) demonstrated low-level amplifications of REST, and only one oligodendroglioma showed high-level amplification of the gene. In medulloblastomas, 38% of samples were determined as aneuploidic, no high-level amplifications were found. Our data suggests that REST is neither activated nor inactivated via mutations in gliomas, while high-level amplification may rarely occur.


Subject(s)
Glioma/genetics , Nervous System Neoplasms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Chi-Square Distribution , Chromatography, High Pressure Liquid/methods , Exons , Humans , In Situ Hybridization, Fluorescence/methods , Molecular Biology/methods
8.
Cancer Genet Cytogenet ; 167(1): 32-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16682283

ABSTRACT

Both breast and ovarian cancers are associated with HER2 receptor activation, which usually results from receptor overexpression and/or gene amplification. The HER-2 gene harbors a polymorphism at codon 655 (GTC/valine to ATC/isoleucine) in the transmembrane domain region, which has been associated with an elevated risk of breast cancer. The objective of this study was to determine whether the polymorphism is under a selection pressure during breast and ovarian carcinogenesis. The Ile/Val genotype was present in 41% (9/22) of the normal DNA of breast cancer patients. An allelic imbalance in the tumor tissue was found in three breast tumors, with overrepresentation of the Val allele. HER-2 was amplified and overexpressed in these tumors. Half of the eight ovarian tumor patients carried heterozygous Ile/Val genotypes. In contrast to breast tumors, all these ovarian cancer specimens showed the presence of the Ile allele. In our selected set of tumors, the Val allele was overrepresented in the subset of HER2-positive breast cancers and the Ile allele in serous ovarian cancer. Further analyses of tumors with known gene amplifications and overexpression may reveal novel associations between germline polymorphisms and development of sporadic tumors.


Subject(s)
Allelic Imbalance , Breast Neoplasms/genetics , Genes, erbB-2 , Genetic Variation , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , DNA, Neoplasm/analysis , Female , Humans , Middle Aged , Nucleic Acid Amplification Techniques , Polymorphism, Genetic , Selection, Genetic
9.
J Mol Med (Berl) ; 83(12): 976-83, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16133419

ABSTRACT

Mutations that may predict response to adenosine 5'-triphosphate (ATP)-mimetic epidermal growth factor receptor (EGFR) inhibitors occur in the EGFR kinase domain in lung adenocarcinomas and bronchioloalveolar carcinomas (BACs). Data on the frequency of EGFR mutations are sparse in other human tumors. Apart from the deletion mutant EGFRvIII, little is known about the frequency of mutations that encode for the EGFR extracellular domains II and IV that participate in receptor dimerization and formation of the tethered (autoinhibited) receptor conformation. We investigated 566 human neoplasms consisting of various histological types for mutations in exons 6, 7 (encode domain II), 14, 15 (domain IV), 18, 19, and 21 (the kinase domain) using denaturing high-performance liquid chromatography (DHPLC). Approximately 4,500 EGFR exons were screened for the presence of a mutation, and samples with an abnormal finding in DHPLC were sequenced. Only one mutation was found in the extracellular domain IV (glioblastoma), and none in domain II. Eight (11%) out of the 40 lung adenocarcinomas, or 33 BACs, investigated had exon 19 or 21 mutation in the kinase domain, but no mutations were found in other tumor types. Most of the lung cancers with mutated EGFR had three to six copies of the mutated gene in fluorescence in situ hybridization. We conclude that mutations of the EGFR kinase domain and the cysteine-rich extracellular domains are infrequent in most types of human cancer apart from lung adenocarcinoma. Mutated EGFR is usually not amplified in lung cancer.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/genetics , Lung Neoplasms/genetics , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Chromatography, High Pressure Liquid , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Exons , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Molecular Sequence Data , Polymerase Chain Reaction , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Sequence Analysis, DNA
10.
J Pathol ; 207(2): 224-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16021678

ABSTRACT

KIT, platelet-derived growth factor receptors (PDGFRs) and vascular endothelial growth factor receptors (VEGFRs) are important clinical targets for tyrosine kinase inhibitors. The frequency of KIT and VEGFR2 amplification in glioblastomas is not known, and few data are available in any other human tumour type. We investigated 43 primary glioblastomas for KIT, VEGFR2, PDGFRA and EGFR amplification using fluorescence in situ hybridization. KIT was amplified in 47% and VEGFR2 in 39% of the glioblastomas, respectively, and PDGFRA in 29%. Thirty-five (81%) of the tumours had either KIT or EGFR amplification. KIT, PDGFRA and VEGFR2 amplifications were strongly associated (p < 0.0001 for each pairwise comparison), suggesting co-amplification, whereas no significant association was found with EGFR amplification. The four secondary glioblastomas arising from pre-existing lower grade astrocytic tumours investigated had KIT amplification but none had EGFR amplification. No mutations were detected with denaturing high-performance liquid chromatography in KIT exons 9, 11, 13 or 17, PDGFRA exons 12 and 18, or EGFR exons 18, 19 or 21. Glioblastomas with KIT, PDGFR or VEGFR2 amplification were associated with similar outcome to other glioblastomas. We conclude that KIT, PDGFRA and VEGFR2 are commonly amplified in primary glioblastoma and that they may also be amplified in secondary glioblastoma. Amplified kinases may be potential targets for tyrosine kinase inhibitor therapy.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Adult , Aged , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Endothelial Cells/pathology , Exons/genetics , Female , Gene Amplification/genetics , Genes, erbB-1/genetics , Glioblastoma/pathology , Glioblastoma/secondary , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Male , Middle Aged , Mutation , Phosphorylation , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL