Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
J Neurooncol ; 126(1): 185-192, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26476729

RESUMO

Survival for glioblastoma (GBM) patients with an unmethyated MGMT promoter in their tumor is generally worse than methylated MGMT tumors, as temozolomide (TMZ) response is limited. How to better treat patients with unmethylated MGMT is unknown. We performed a trial combining erlotinib and bevacizumab in unmethylated GBM patients after completion of radiation (RT) and TMZ. GBM patients with an unmethylated MGMT promoter were trial eligible. Patient received standard RT (60 Gy) and TMZ (75 mg/m2 × 6 weeks) after surgical resection of their tumor. After completion of RT they started erlotinib 150 mg daily and bevacizumab 10 mg/kg every 2 weeks until progression. Imaging evaluations occurred every 8 weeks. The primary endpoint was overall survival. Of the 48 unmethylated patients enrolled, 46 were evaluable (29 men and 17 women); median age was 55.5 years (29-75) and median KPS was 90 (70-100). All patients completed RT with TMZ. The median number of cycles (1 cycle was 4 weeks) was 8 (2-47). Forty-one patients either progressed or died with a median progression free survival of 9.2 months. At a follow up of 33 months the median overall survival was 13.2 months. There were no unexpected toxicities and most observed toxicities were categorized as CTC grade 1 or 2. The combination of erlotinib and bevacizumab is tolerable but did not meet our primary endpoint of increasing survival. Importantly, more trials are needed to find better therapies for GBM patients with an unmethylated MGMT promoter.


Assuntos
Antineoplásicos/uso terapêutico , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Cloridrato de Erlotinib/uso terapêutico , Glioblastoma/tratamento farmacológico , Radioterapia/efeitos adversos , Adulto , Metilação de DNA , Metilases de Modificação do DNA/genética , Dacarbazina/efeitos adversos , Dacarbazina/análogos & derivados , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Temozolomida , Resultado do Tratamento , Adulto Jovem
2.
Br J Cancer ; 111(10): 2014-23, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25314059

RESUMO

BACKGROUND: In a clinical diagnostic laboratory, we evaluated the applicability of the Ion Proton sequencer for screening 409 cancer-related genes in solid tumours. METHODS: DNA was extracted from formalin-fixed, paraffin-embedded (FFPE) tissue biopsy specimens of 55 solid tumours (20 with matched normal tissue) and four cell lines and screened for mutations in 409 genes using the Ion Proton system. The mutation profiles of these samples were known based on prior testing using the Ion Torrent Personal Genome Machine (46-gene hotspot panel), Sanger sequencing, or fluorescence in situ hybridisation (FISH). Concordance with retrospective findings and additional mutations were evaluated. Assay sensitivity and reproducibility were established. Gene copy number variations (CNVs) detected were confirmed by molecular inversion probe (MIP) array. RESULTS: The average Ion Proton (409-gene panel) sequencing output per run was 8 gigabases with 128 million sequencing reads. Of the 15,992 amplicons in the 409-gene panel, 90% achieved a minimum average sequencing depth of 100X. In 59 samples, the Ion Proton detected 100 of 105 expected single-nucleotide variants (SNVs) and all expected deletions (n=8), insertions (n=5), and CNVs (n=7). Five SNVs were not detected due to failed amplification of targeted regions. In 20 tumours with paired normal tissue, Ion Proton detected 37 additional somatic mutations, several in genes of high prognostic or therapeutic significance, such as MET, ALK, TP53, APC, and PTEN. MIP array analysis confirmed all CNVs detected by Ion Proton. CONCLUSIONS: The Ion Proton (409-gene panel) system was found to be well suited for use in a clinical molecular diagnostic laboratory. It can simultaneously screen 409 genes for a variety of sequence variants in multiple samples using a low input of FFPE DNA with high reproducibility and sensitivity.


Assuntos
Variações do Número de Cópias de DNA , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação/genética , Proteínas de Neoplasias/genética , Neoplasias/diagnóstico , Neoplasias/genética , DNA/análise , DNA/genética , Humanos , Hibridização in Situ Fluorescente , Reação em Cadeia da Polimerase , Reprodutibilidade dos Testes
3.
Oncogene ; 31(10): 1264-74, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21804599

RESUMO

The phosphatidylinositol-3-OH kinase (PI3K)-Akt pathway is activated in cancer by genetic or epigenetic events and efforts are under way to develop targeted therapies. phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor is the major brake of the pathway and a common target for inactivation in glioblastoma, one of the most aggressive and therapy-resistant cancers. To achieve potent inhibition of the PI3K-Akt pathway in glioblastoma, we need to understand its mechanism of activation by investigating the interplay between its regulators. We show here that PTEN modulates the PI3K-Akt pathway in glioblastoma within a tumor suppressor network that includes Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) and pleckstrin-homology domain leucine-rich repeat protein phosphatases 1 (PHLPP1). The NHERF1 adaptor, previously characterized by our group as a PTEN ligand and regulator, shows also PTEN-independent Akt-modulating effects that led us to identify the PHLPP1/PHLPP2 Akt phosphatases as NHERF1 ligands. NHERF1 interacts via its PDZ domains with PHLPP1/PHLPP2 and scaffolds heterotrimeric complexes with PTEN. Functionally, PHLPP1 requires NHERF1 for membrane localization and growth-suppressive effects. PHLPP1 loss boosts Akt phosphorylation only in PTEN-negative cells and cooperates with PTEN loss for tumor growth. In a panel of low-grade and high-grade glioma patient samples, we show for the first time a significant disruption of all three members of the PTEN-NHERF1-PHLPP1 tumor suppressor network in high-grade tumors, correlating with Akt activation and patient's abysmal survival. We thus propose a PTEN-NHERF1-PHLPP PI3K-Akt pathway inhibitory network that relies on molecular interactions and can undergo parallel synergistic hits in glioblastoma.


Assuntos
Neoplasias Encefálicas/etiologia , Glioblastoma/etiologia , Proteínas Nucleares/fisiologia , PTEN Fosfo-Hidrolase/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Fosfoproteínas/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Idoso , Humanos , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais
4.
Cell Death Dis ; 2: e121, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21368892

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in malignant cells, including gliomas, and is currently in anticancer clinical trials. However, the full-length and tagged forms of TRAIL, unlike the untagged ligand (soluble TRAIL (sTRAIL)), exhibits toxicity against normal cells. Here, we report the generation and testing of an adenovirus (AdsTRAIL) that expresses untagged sTRAIL in an intracranial xenograft model and a human glioma organotypic slice culture model. AdsTRAIL efficiently induced apoptosis in glioma cell lines, including those resistant to sTRAIL, but not in normal human astrocytes (NHAs). It inhibited anchorage-independent glioma growth and exerted a bystander effect in transwell assays. Intratumoral injections of AdsTRAIL in a rodent intracranial glioma model resulted in reduced tumor growth and improved survival compared with Ad-enhanced green fluorescent protein (EGFP)- or vehicle-treated controls without toxicity. Human glioma organotypic slices treated with AdsTRAIL demonstrated apoptosis induction and caspase activation.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/genética , Glioma/terapia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adenoviridae/metabolismo , Animais , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Secções Congeladas , Terapia Genética/instrumentação , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glioma/metabolismo , Glioma/fisiopatologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico
5.
Oncogene ; 29(31): 4460-72, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20531304

RESUMO

Angiogenesis involves a complex set of cell-cell and cell-extracellular matrix (ECM) interactions that coordinately promote and inhibit blood vessel growth and sprouting. Although many factors that promote angiogenesis have been characterized, the identities and mechanisms of action of endogenous inhibitors of angiogenesis remain unclear. Furthermore, little is known about how cancer cells selectively circumvent the actions of these inhibitors to promote pathological angiogenesis, a requisite event for tumor progression. Using mosaic mouse models of the malignant brain cancer, astrocytoma, we report that tumor cells induce pathological angiogenesis by suppressing expression of the ECM protein receptor alphavbeta8 integrin. Diminished integrin expression in astrocytoma cells leads to reduced activation of latent TGFbetas, resulting in impaired TGFbeta receptor signaling in tumor-associated endothelial cells. These data reveal that astrocytoma cells manipulate their angiogenic balance by selectively suppressing alphavbeta8 integrin expression and function. Finally, these results show that an adhesion and signaling axis normally involved in developmental brain angiogenesis is pathologically exploited in adult brain tumors.


Assuntos
Astrocitoma/irrigação sanguínea , Astrocitoma/genética , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/genética , Integrinas/fisiologia , Neovascularização Patológica/genética , Animais , Animais Recém-Nascidos , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Integrinas/genética , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Mosaicismo , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Transplante Heterólogo , Células Tumorais Cultivadas
6.
Cancer Gene Ther ; 16(3): 256-65, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18927600

RESUMO

The fact that glioblastomas, which are one of the most devastating cancers, frequently express the Delta-EGFR (epithelial growth factor receptor) also called mutant variant III of EGFR (EGFRvIII) suggests that this cancer cell-specific receptor might serve as an ideal target for cancer therapy. To assess its potential as such a target, we constructed an oncolytic adenovirus with Retargeted Infectivity Via EGFR (Delta-24-RIVER) on the backbone of Delta-24. This new oncolytic adenovirus targets, as Delta-24 does, the disrupted Rb pathway in cancer cells; in addition, this adenovirus has also been retargeted through the abrogation of CAR binding (Y477A mutation in adenoviral fiber protein) and insertion of an EGFRvIII-specific binding peptide in the HI loop of the fiber protein. As compared with Delta-24, Delta-24-RIVER induced EGFRvIII-selective cytotoxicity in U-87 MG isogenic cell lines and in tetracycline-inducible EGFRVIII expressing U-251 MG cells. Accordingly, by tittering the viral progeny and examining fiber protein expression in the above cells, we showed that the replication of this new construct also correlated with EGFRvIII expression. Consistently, immunohistochemistry staining of the adenoviral capsid protein hexon in the virus-treated tumors revealed that the virus replicated more efficiently in EGFRvIII-expressing U-87 MG.DeltaEGFR xenografts than in the tumors grown from U-87 MG cells. Importantly, treatment with Delta-24-RIVER prolonged the survival of animals with intracranial xenografts derived from U-87 MG.DeltaEGFR cells. Therefore, our results constitute the first proof of the direct targeting of a cancer-specific receptor using an oncolytic adenovirus.


Assuntos
Adenovírus Humanos/fisiologia , Neoplasias Encefálicas/terapia , Receptores ErbB/antagonistas & inibidores , Vetores Genéticos/uso terapêutico , Glioblastoma/terapia , Proteínas de Neoplasias/antagonistas & inibidores , Terapia Viral Oncolítica , Adenovírus Humanos/genética , Sequência de Aminoácidos , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/transplante , Éxons/genética , Genes do Retinoblastoma , Genes erbB-1 , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Deleção de Sequência , Replicação Viral
7.
J Neurooncol ; 90(1): 89-97, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18581057

RESUMO

Targeting the epidermal growth factor receptor (EGFR) may be effective in a subset of glioblastoma patients. This phase II study assessed the clinical activity of erlotinib plus carboplatin and to determine molecular predictors of response. The primary endpoint was progression free survival (PFS). Patients with recurrent glioblastoma with no more than two prior relapses received carboplatin intravenously on day 1 of every 28-day cycle (target AUC of 6 mg x ml/min). Daily erlotinib at 150 mg/day was dose escalated to 200 mg/day, as tolerated. Clinical and MRI assessments were made every 4 and 8 weeks, respectively. Tumor tissue was evaluated for EGFR, AKT and phosphatase and tensin homolog (PTEN) status. One partial response (PR) was observed out of 43 assessable patients. Twenty patients (47%) had stable disease (SD) for an average of 12 weeks. Median PFS was 9 weeks. The 6-month PFS rate was 14%. Median overall survival (OS) was 30 weeks. This regimen was well tolerated with grade 3/4 toxicities of fatigue, leukopenia, thrombocytopenia and rash requiring dose reductions. A recursive partitioning analysis (RPA) predicted that patients with KPS >or=90 treated with more than 1 prior regimen had the highest OS. No correlation was observed between EGFR, Akt or PTEN expression and either PFS or OS. Carboplatin plus erlotinib is well tolerated but has modest activity in unselected patients. Future trials should be stratified based on optimal molecular or clinical characteristics.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Adulto , Idoso , Biomarcadores Tumorais/análise , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Intervalo Livre de Doença , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Feminino , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/administração & dosagem , Quinazolinas/efeitos adversos
8.
Ann Oncol ; 19(7): 1242-1248, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18334512

RESUMO

BACKGROUND: The purpose of this retrospective study was to determine, in a cohort of patients with breast cancer and central nervous system (CNS) metastases, the effect of trastuzumab in patients with human epidermal growth factor receptor 2 (HER2)-positive disease and to compare this with that of patients with HER2-negative disease. METHODS: Five hundred and ninety-eight patients with invasive breast cancer, CNS metastases and known HER2 status were identified. Time to CNS metastases and survival after CNS metastases were estimated by the Kaplan-Meier method, and Cox models were fitted to determine the association between HER2 status, trastuzumab treatment and outcomes after adjustment for other patient characteristics. RESULTS: In the multivariable model, patients with HER2-negative disease [Hazard ratio (HR) 1.50, 95% confidence interval (CI) 1.15-1.95, P = 0.003] and patients with HER2-positive disease who did not receive trastuzumab (HR 2.13, 95% CI 1.51-3.00, P < 0.0001) had shorter times to CNS metastases compared with patients with HER2-positive disease who had received trastuzumab as first-line therapy for metastases. Furthermore, patients with HER2-negative disease (HR 1.66, 95% CI 1.31-2.12, P < 0.0001) and patients with HER2-positive disease who had never received trastuzumab (HR 1.34, 95% CI 0.78-2.30, P = 0.28) had an increased hazard of death compared with patients with HER2-positive disease who had received trastuzumab before or at the time of CNS metastases diagnosis. CONCLUSION: In our cohort of patients with breast cancer and CNS metastases, patients with HER2-positive disease treated with trastuzumab had longer times to development of and better survival from CNS metastases compared with patients with HER2-positive disease who had never received trastuzumab and patients with HER2-negative breast cancer.


Assuntos
Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Neoplasias do Sistema Nervoso Central/secundário , Receptor ErbB-2/metabolismo , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/terapia , Estudos de Coortes , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Análise Multivariada , Metástase Neoplásica/diagnóstico , Metástase Neoplásica/patologia , Metástase Neoplásica/terapia , Prognóstico , Receptor ErbB-2/genética , Estudos Retrospectivos , Análise de Sobrevida , Fatores de Tempo , Trastuzumab , Resultado do Tratamento
9.
Oncogene ; 26(42): 6212-9, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-17404569

RESUMO

We recently showed that FoxM1 is overexpressed in human glioblastomas and that forced FoxM1B expression in anaplastic astrocytoma cells leads to the formation of highly invasive glioblastoma multiforme (GBM) in nude mice. However, the molecular mechanisms by which FoxM1 enhances glioma invasion are unknown. In this study, we found that FoxM1 overexpression increased matrix metalloproteinase (MMP)-2 expression in glioma cells, whereas blockade of FoxM1 expression suppressed MMP-2 expression. Transfection of FoxM1 into glioma cells directly activated the MMP-2 promoter, whereas inhibition of FoxM1 expression by FoxM1-siRNA suppressed its activation. We identified a FoxM1-binding site in the MMP-2 promoter and demonstrated that FoxM1 protein bound directly to it. Mutation of this FoxM1-binding site significantly attenuated MMP-2 promoter activity. Furthermore, FoxM1 overexpression increased the invasiveness of glioma cells, whereas inhibition of FoxM1 expression suppressed the invasiveness of GBM cells. Inhibition of MMP-2 by a specific MMP-2 inhibitor reversed the invasive phenotype of glioma cells overexpressing FoxM1. Finally, immunohistochemical analysis of 45 human GBM specimens showed a significant correlation between FoxM1 overexpression and elevated MMP-2 expression. Collectively, these findings provide evidence that FoxM1 contributes to glioma progression by enhancing MMP-2 gene transcription and thus tumor-cell invasion.


Assuntos
Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Glioma/patologia , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Regulação para Cima/fisiologia , Linhagem Celular Tumoral , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/fisiologia , Glioma/enzimologia , Glioma/genética , Humanos , Invasividade Neoplásica , Transcrição Gênica/fisiologia
10.
Neurology ; 66(1): 127-30, 2006 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-16401863

RESUMO

Using whole genome expression microarray technology to discover clinically relevant biomarkers for pilocytic astrocytoma (PA), the authors identified matrilin-2 as a unique mRNA overexpressed in PA. Matrilin-2 protein expression was similarly elevated in the majority of sporadic PA, but in only one neurofibromatosis 1-associated PA with an unusually aggressive clinical phenotype. These results suggest that matrilin-2 may be a specific and clinically useful biomarker for discriminating between indolent and clinically aggressive PA.


Assuntos
Astrocitoma/diagnóstico , Astrocitoma/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Astrocitoma/classificação , Transformação Celular Neoplásica/genética , Criança , Cromossomos Humanos Par 8/genética , Análise Mutacional de DNA , Proteínas da Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Testes Genéticos , Glicoproteínas/metabolismo , Humanos , Imuno-Histoquímica , Proteínas Matrilinas , Neurofibromatose 1/complicações , Neurofibromatose 1/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , RNA Mensageiro/genética , Regulação para Cima/genética
11.
Neuropathol Appl Neurobiol ; 29(6): 584-95, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14636165

RESUMO

Haematogenous leucocytes enter the central nervous system (CNS) during diverse disorders of varied aetiologies. Understanding the trafficking cues that mediate CNS leucocyte infiltration might promote the development of flexible and selective means to modulate inflammation to achieve clinical benefit. The trafficking machinery of leucocytes has been elucidated during the past decade and consists of cell-surface adhesion molecules, chemoattractant cytokines (chemokines) and their receptors. Recent work in our laboratory characterized chemokine receptors found on T lymphocytes and monocytes in brain sections from subjects with one pathological subtype of multiple sclerosis (MS), an immune-mediated inflammatory demyelinating disease. In these tissues, the types 1 and 5 CC chemokine receptors (CCR1 and CCR5) were detected on perivascular monocytic cells whereas only CCR5 was present on parenchymal macrophages. The type 3 CXC chemokine receptor (CXCR3) was present on virtually all CD3-positive T cells. In the current study, we evaluated the expression of these receptors on the infiltrating cells present in cases of other inflammatory CNS disorders including those of dysimmune, infectious, neoplastic, and vascular aetiology. Perivascular and parenchymal monocytic cells expressed CCR1 in all cases and CXCR3 was consistently present on a substantial proportion of CD3+ T cells. The occurrence of CCR5 on parenchymal macrophages was much less uniform across the varied disorders. These data implicate CCR1 in monocyte infiltration of the CNS and are consistent with reports of studies in CCR1-deficient mice. CXCR3 is also likely to play a role in accumulation of T cells in the inflamed CNS. By contrast, our findings suggest that regulation of CCR5 on phagocytic macrophages may be contingent on the lesion environment.


Assuntos
Encefalopatias/patologia , Inflamação/patologia , Leucócitos Mononucleares/metabolismo , Receptores de Quimiocinas/metabolismo , Adolescente , Adulto , Idoso , Encefalopatias/imunologia , Complexo CD3/metabolismo , Calgranulina B/metabolismo , Criança , Humanos , Imuno-Histoquímica , Inflamação/imunologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
12.
Glia ; 36(3): 391-405, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11746775

RESUMO

Malignant gliomas (MGs), lethal human central nervous system (CNS) neoplasms, contain tumor infiltrating lymphocytes (TIL). Although MHC class II molecules are frequently detected on MG cells, suggesting that they may be capable of antigen (Ag) presentation to CD4(+) T cells, deficiencies in CD4(+) T-cell activation are associated with these nonimmunogenic tumors. We evaluated regulation of the MHC class II transactivator (CIITA), the key intermediate that controls class II expression, in MG cells and tested whether MG cells could process native Ag. After interferon-gamma (IFN-gamma) stimulation, MG cells upregulated CIITA and class II molecules. IFN-gamma-inducible CIITA expression in MG cells, as well as primary human astrocytes, was directed by two CIITA promoters, pIV, the promoter for IFN-gamma-inducible CIITA expression in nonprofessional antigen-presenting cells (APC), and pIII, the promoter that directs constitutive CIITA expression in B cells. Both pIII and pIV directed CIITA transcription in vivo in MGs and ex vivo in IFN-gamma-activated primary MG cultures. We also demonstrate for the first time that MG cells can process native Ag for presentation to CD4(+) MHC class II-restricted Th1 cells, indicating that MG cells can serve as nonprofessional APC. CIITA may be a key target to modulate MHC class II expression, which could augment immunogenicity, Ag presentation, and CD4(+) T-cell activation in MG therapy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Neoplasias Encefálicas/imunologia , Linfócitos T CD4-Positivos/imunologia , Glioma/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Interferon gama/imunologia , Proteínas Nucleares , Regiões Promotoras Genéticas/imunologia , Transativadores/imunologia , Adulto , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Astrócitos/citologia , Astrócitos/imunologia , Astrócitos/metabolismo , Autoantígenos/imunologia , Autoantígenos/farmacologia , Sequência de Bases/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Éxons/genética , Éxons/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Glioma/metabolismo , Glioma/fisiopatologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imuno-Histoquímica , Interferon gama/metabolismo , Interferon gama/farmacologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteína Básica da Mielina/imunologia , Proteína Básica da Mielina/farmacologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Transativadores/genética , Transativadores/metabolismo , Células Tumorais Cultivadas
13.
J Neuropathol Exp Neurol ; 60(11): 1099-104, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11706939

RESUMO

Although there is much written about the molecular definitions of "primary" glioblastomas (GBM), there is little known about the histological features of this predominant subtype. We hypothesized that the "small cell architecture" would represent a histological feature of most primary GBMs. This was tested by comparing the presence of the small cell phenotype with the presence or absence of amplification of the epidermal growth factor receptor (EGFR), a common event in primary GBMs. After a pilot study that found a correlation between this small cell phenotype and EGFR amplification, we selected 9 pure small cell GBMs (SCGBM) and 12 non-SCGBMs to be studied for EGFR amplification by fluorescence in situ hybridization (FISH). In this set of 21 cases, 8 of 9 SCGBMs and 5 of 12 non-SCGBMs were amplified for EGFR. We then correlated the EGFR status of 79 GBMs unselected for their histological features from a set that had been previously characterized in regard to EGFR amplification. Fourteen of 21 (67%) exclusively small cell neoplasms, 8 of 25 (32%) GBMs with both small cell and non-small cell areas, and 3 of 33 (9%) non-small cell GBMs were amplified for EGFR (p = 0.0004 with an exact test). We conclude that EGFR amplification is associated with a small cell phenotype in GBMs and that SCGBMs are an important component of "primary" GBMs.


Assuntos
Neoplasias Encefálicas/patologia , Receptores ErbB/genética , Glioblastoma/patologia , Neuroglia/patologia , Neoplasias Encefálicas/classificação , Tamanho Celular , Glioblastoma/classificação , Humanos , Hibridização in Situ Fluorescente , Fenótipo
14.
Int J Radiat Oncol Biol Phys ; 51(2): 410-8, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567815

RESUMO

PURPOSE: Recent studies have suggested relative radioresistance in glioblastoma multiforme (GM) tumors in older patients, consistent with their shorter survival. Two common molecular genetic abnormalities in GM are age related: epidermal growth factor receptor (EGFR) overexpression in older patients and p53 mutations in younger patients. We tested whether these abnormalities correlated with clinical heterogeneity in GM response to radiation treatment. METHODS AND MATERIALS: Radiographically assessed radiation response (5-level scale) was correlated with EGFR immunoreactivity, p53 immunoreactivity, and p53 exon 5-8 mutation status in 170 GM patients treated using 2 prospective clinical protocols. Spearman rank correlation and proportional-odds ordinal regression were used for univariate and multivariate analysis. RESULTS: Positive EGFR immunoreactivity predicted poor radiographically assessed radiation response (p = 0.046). Thirty-three percent of tumors with no EGFR immunoreactivity had good radiation responses (>50% reduction in tumor size by CT or MRI), compared to 18% of tumors with intermediate EGFR staining and 9% of tumors with strong staining. There was no significant relationship between p53 immunoreactivity or mutation status and radiation response. Significant relationships were noted between EGFR score and older age and between p53 score or mutation status and younger age. CONCLUSION: The observed relative radioresistance of some GMs is associated with overexpression of EGFR.


Assuntos
Neoplasias Encefálicas/radioterapia , Receptores ErbB/metabolismo , Genes p53/genética , Glioblastoma/radioterapia , Proteínas de Neoplasias/metabolismo , Neoplasias Supratentoriais/radioterapia , Adolescente , Adulto , Fatores Etários , Idoso , Análise de Variância , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Estudos Prospectivos , Dosagem Radioterapêutica , Análise de Regressão , Neoplasias Supratentoriais/genética , Neoplasias Supratentoriais/metabolismo
15.
Cancer Res ; 61(18): 6674-8, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559533

RESUMO

Human malignant gliomas are thought to develop as the result of stepwise accumulations of multiple genetic alterations. Recently, we showed that E6/E7-mediated inactivation of p53/pRb, ras pathway activation (initiated by expression of mutant H-Ras), and expression of human telomerase reverse transcriptase (hTERT) in combination converted normal human astrocytes into cells that formed intracranial tumors resembling human anaplastic astrocytoma (AA). In this study, we created human astrocytes that, in addition to expressing E6/E7, hTERT, and Ras, also expressed a constitutive activated form of Akt intended to mimic the Akt activation noted in grade IV glioblastoma multiforme (GBM). Although these cells grew no differently than astrocytes expressing E6, E7, and H-Ras in vitro or in the first 28 days following s.c. implantation, they ultimately formed tumors four to six times larger than those formed by the E6/E7/hTERT/Ras cells. Unlike the poorly vascularized, necrosis-free AA formed by E6/E7/hTERT/Ras cells, the tumors formed by s.c. or intracranial injection of Akt-expressing cells had large areas of necrosis surrounded by neovascularization and were consistent in appearance with grade IV human GBM. These results show that activation of the Akt pathway is sufficient to allow conversion of human AA to human GBM.


Assuntos
Astrocitoma/enzimologia , Astrocitoma/patologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Glioblastoma/enzimologia , Glioblastoma/patologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Animais , Astrócitos/enzimologia , Astrócitos/patologia , Astrócitos/fisiologia , Astrocitoma/genética , Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Ativação Enzimática , Glioblastoma/genética , Humanos , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Ratos , Transdução de Sinais/fisiologia , Transfecção
16.
Cancer Res ; 61(13): 4956-60, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431323

RESUMO

The formation of human malignant gliomas is thought to involve the accumulation of multiple genetic alterations. To define the function of specific alterations in glioma formation, we serially introduced genetic alterations functionally equivalent to those noted in human malignant gliomas into normal human astrocytes (NHAs). We then monitored the ability of each of these alterations to contribute to the growth of otherwise genetically stable NHAs into intracranial malignant gliomas. Using this model, we show that expression of human telomerase catalytic component (hTERT), but not E7-mediated inactivation of pRb or E6/E7-mediated inactivation of p53/pRb, was sufficient to initiate the tumorigenic process by circumventing cellular senescence in astrocytes. hTERT expression, even in combination with inactivation of p53/pRb, did not transform astrocytes. These alterations together, however, cooperated with ras pathway activation (initiated by expression of mutant H-Ras), but not with phosphatidylinositol 3-kinase pathway activation (initiated by expression of myristoylated Akt) or epidermal growth factor receptor activation, to allow for the formation of intracranial tumors strongly resembling p53/pRb pathway-deficient, telomerase-positive, ras-activated human grade III anaplastic astrocytomas. These results identify four pathways as key in the development of human anaplastic astrocytomas.


Assuntos
Astrócitos/fisiologia , Astrocitoma/genética , Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , RNA , Proteínas Repressoras , Animais , Astrócitos/patologia , Proteínas de Ligação a DNA , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Oncogênicas Virais/biossíntese , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/fisiologia , Proteínas E7 de Papillomavirus , Proteína do Retinoblastoma/antagonistas & inibidores , Transdução de Sinais/genética , Telomerase/biossíntese , Telomerase/genética , Telomerase/fisiologia , Transfecção , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteínas ras/fisiologia
17.
Cancer Res ; 61(10): 3949-54, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358811

RESUMO

In the United States and the San Francisco Bay Area, whites are nearly twice as likely as non-whites to develop brain cancer. To test whether prevalence and types of alterations in the p53 pathway in brain tumor development may explain some of this difference in risk, we have analyzed the p53 status of astrocytic gliomas from a population-based sample of cases within our San Francisco Bay Area Adult Glioma Study. We identified mutations in exons 5-8 of p53 using DNA extracted from formalin-fixed paraffin-embedded tissue blocks from 146 whites and 26 non-whites with astrocytic glioma by PCR-single-strand conformation polymorphism and direct sequencing. Tumor P53 protein (TP53) immunohistochemistry (IHC) available for 164 of these cases showed that tumors from 50% (13 of 26) of non-whites and 32% (44 of 138) of whites contained intense IHC staining for TP53, indicating persistence of TP53 protein. Irrespective of IHC status, tumors from 42% (11 of 26) of non-whites versus 13% (19 of 146) of whites contained p53 mutations (age/gender-adjusted odds ratio, 5.7; 95% confidence interval, 2.2-15.1; P = 0.0004). Patients with p53 mutation-positive tumors were also significantly younger than patients with mutation-negative tumors and somewhat more likely to be female. A higher proportion of tumors from non-whites than from whites had transition mutations, but there were similar proportions of transversion mutations in tumors from whites and non-whites. Whites and non-whites also had similar proportions of tumors with p53 mutations that stained intensely for TP53 (78 and 82%, respectively). Because whites have higher risk for glioma than non-whites in this population, that the gliomas from whites were less likely than those from non-whites to have p53 mutation suggests that whites may be more likely than non-whites to be at risk for the more common type of astrocytic gliomas, which do not contain p53 mutations.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Etnicidade/genética , Genes p53/genética , Glioblastoma/genética , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Feminino , Predisposição Genética para Doença , Glioblastoma/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação , Fatores Sexuais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
J Mol Diagn ; 3(2): 62-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11333301

RESUMO

We amplified various amounts of DNA derived from frozen SF210 and U251NCI human glioblastoma cells, carried out comparative genomic hybridization (CGH) using degenerate oligonucleotide primed-PCR (DOP-PCR) products as test probes, and compared results to analyses performed with probes prepared by standard nick translation. Next we extracted DNA from hematoxylin-eosin (HE)- and methyl green (MG)-stained, microdissected sections of formalin-fixed and paraffin-embedded U251NCI cells, amplified and labeled it by DOP-PCR, and subjected it to CGH. Finally, we used the same methods in multiple samples from a single human mixed glioma tissue. DOP-PCR products from 50 pg to 250 ng of DNA were equally effective in generating the same CGH profiles as the standard method. DOP-PCR products from microdissected pieces of MG-stained cells were effective probes for CGH, but HE-stained samples were not desirable. As the proportion of HE-stained sample increased, CGH profiles deteriorated. DOP-PCR products from microdissected pieces of MG-stained paraffin sections of glioma tissue produced CGH profiles compatible with their histological features. CGH performed with DOP-PCR products from microdissected paraffin blocks allows for the accurate investigation of the cytogenetic characteristics from invasive tumors and of cytogenetic heterogeneity within neoplastic tissue.


Assuntos
Aberrações Cromossômicas , Reação em Cadeia da Polimerase/métodos , Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , DNA/metabolismo , Eletroforese em Gel de Ágar , Humanos , Hibridização de Ácido Nucleico , Células Tumorais Cultivadas
19.
Am J Pathol ; 158(4): 1253-62, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11290543

RESUMO

The combined loss of chromosomes 1p and 19q has recently emerged as a genetic predictor of chemosensitivity in anaplastic oligodendrogliomas. Here, we describe a strategy that uses a novel method of real-time quantitative polymerase chain reaction, quantitative microsatellite analysis (QuMA), for the molecular analysis of 1p and 19q loss in oligodendrogliomas and oligoastrocytomas in archival routinely processed paraffin material. QuMA is performed on the ABI 7700 and based on amplifications of microsatellite loci that contain (CA)n repeats where the repeat itself is the target for hybridization by the fluorescently labeled probe. This single probe can therefore be used to determine copy number of microsatellite loci spread throughout the human genome. In genomic DNA prepared from paraffin-embedded brain tumor specimens, QuMA detected combined loss of 1p and 19q in 64% (21 of 32) of oligodendrogliomas and 67% (6 of 9) of oligoastrocytomas. We validate the use of QuMA as a reliable method to detect copy number by showing concordance between QuMA and fluorescence in situ hybridization at 37 of 45 chromosomal arms tested. These results indicate that QuMA is an accurate, high-throughput assay for the detection of copy number at multiple loci; as many as 31 loci of an individual tumor can be analyzed on a 96-well plate in a single 2-hour run. In addition, it has advantages over standard allelic imbalance/loss of heterozygosity assays in that all loci are potentially informative, paired normal tissue is not required, and gain can be distinguished from loss. QuMA may therefore be a powerful molecular tool to expedite the genotypic analysis of human gliomas in a clinical setting for diagnostic/prognostic purposes.


Assuntos
Neoplasias Encefálicas/genética , Cromossomos Humanos Par 19 , Cromossomos Humanos Par 1 , Deleção de Genes , Repetições de Microssatélites , Oligodendroglioma/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Cromossomos Humanos Par 10 , Sistemas Computacionais , Humanos , Hibridização in Situ Fluorescente , Oligodendroglioma/diagnóstico , Oligodendroglioma/patologia , Reação em Cadeia da Polimerase
20.
Am J Pathol ; 158(3): 1137-43, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238062

RESUMO

Ependymoma occurs most frequently within the central nervous system of children and young adults. We determined relative chromosomal copy-number aberrations in 44 ependymomas using comparative genomic hybridization. The study included 24 intracranial and 20 spinal cord tumors from pediatric and adult patients. Frequent chromosomal aberrations in intracranial tumors were gain of 1q and losses on 6q, 9, and 13. Gain of 1q and loss on 9 were preferentially associated with histological grade 3 tumors. On the other hand, gain on chromosome 7 was recognized almost exclusively in spinal cord tumors, and was associated with various other chromosomal aberrations including frequent loss of 22q. We conclude that cytogenetic analysis of ependymomas may help to classify these tumors and provide leads concerning their initiation and progression. The relationship of these aberrations to patient outcome needs to be addressed.


Assuntos
Neoplasias Encefálicas/classificação , Aberrações Cromossômicas , Ependimoma/classificação , Neoplasias da Medula Espinal/classificação , Adolescente , Adulto , Idoso , Aneuploidia , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Análise Citogenética , Ependimoma/genética , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Neoplasias da Medula Espinal/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...