Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
1.
Clin Radiol ; 70(10): 1087-95, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26231469

RESUMO

AIM: To investigate whether quantitative dynamic susceptibility-weighted contrast-enhanced (DSC) perfusion magnetic resonance imaging (MRI) metrics are influenced by cellular and genomic expression patterns of glioblastoma angiogenesis. MATERIALS AND METHODS: Twenty-five stereotactic neurosurgical tissue samples were prospectively obtained from enhancing and non-enhancing tumour regions from 10 patients with treatment-naïve glioblastoma. Using monoclonal antibodies, histopathological features of angiogenesis were examined: total microvascular density, vascular morphology, and hypoxia. Angiogenic expression patterns of tissue samples were investigated using RNA microarrays. DSC perfusion MRI metrics were measured from the tissue sampling sites. MRI and histopathological variables were compared using Pearson's correlations. Microarray analysis was performed using false discovery rate (FDR) statistics. RESULTS: Thirteen enhancing and 12 non-enhancing MR image-guided tissue specimens were prospectively obtained. Enhancing tumour regions demonstrated a significant difference in DSC perfusion and histopathological metrics of angiogenesis when compared to non-enhancing regions. Four angiogenic pathways (vascular endothelial growth factor [VEGF], hypoxia inducible factor [HIF], platelet-derived growth factor [PDGF], fibroblast growth factor [FGF]; 25 individual genes) were significantly up-regulated within enhancing regions when compared to non-enhancing regions (adjusted p<0.05, FDR <0.05). A statistically significant correlation was observed between VEGF-A expression, microvascular density, microvascular morphology, and DSC perfusion MRI metrics (p<0.05). CONCLUSION: Pro-angiogenic genomic and cellular expression patterns of treatment-naïve primary glioblastoma significantly influences morphological and physiological DSC perfusion metrics suggesting that expression levels of therapeutically relevant genetic signatures can be quantified using MRI.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Imagem de Difusão por Ressonância Magnética/métodos , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/diagnóstico , Glioblastoma/genética , Neoplasias Encefálicas/irrigação sanguínea , Feminino , Glioblastoma/irrigação sanguínea , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica , Estudos Prospectivos
2.
Br J Cancer ; 106(5): 839-45, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22333598

RESUMO

BACKGROUND: Bcl-2 family genes are frequently amplified in small cell lung cancer (SCLC). A phase I trial was conducted to evaluate the safety of obatoclax, a Bcl-2 family inhibitor, given in combination with standard chemotherapy. METHODS: Eligible patients (3-6 per cohort) had extensive-stage SCLC, measurable disease, ≤ 1 before therapy, Eastern Cooperative Oncology Group performance status 0 or 1, and adequate organ function. Patients were treated with escalating doses of obatoclax, either as a 3- or 24-h infusion, on days 1-3 of a 21-day cycle, in combination with carboplatin (area under the curve 5, day 1 only) and etoposide (100 mg m(-2), days 1-3). The primary endpoint was to determine the maximum tolerated dose of obatoclax. RESULTS: Twenty-five patients (56% male; median age 66 years) were enrolled in three dose cohorts for each schedule. Maximum tolerated dose was established with the 3-h infusion at 30 mg per day and was not reached with the 24-h infusion. Compared with the 24-h cohorts, the 3-h cohorts had higher incidence of central nervous system (CNS) adverse events (AEs); dose-limiting toxicities were somnolence, euphoria, and disorientation. These CNS AEs were transient, resolving shortly after the end of infusion, and without sequelae. The response rate was 81% in the 3-h and 44% in the 24-h infusion cohorts. CONCLUSION: Although associated with a higher incidence of transient CNS AEs than the 24-h infusion, 3-h obatoclax infusion combined with carboplatin-etoposide was generally well tolerated at doses of 30 mg per day. Though patient numbers were small, there was a suggestion of improved efficacy in the 3-h infusion group. Obatoclax 30 mg infused intravenously over 3 h on 3 consecutive days will be utilised in future SCLC studies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/administração & dosagem , Etoposídeo/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Pirróis/administração & dosagem , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carboplatina/efeitos adversos , Sistema Nervoso Central/efeitos dos fármacos , Esquema de Medicação , Etoposídeo/efeitos adversos , Feminino , Humanos , Indóis , Neoplasias Pulmonares/patologia , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Pirróis/efeitos adversos , Carcinoma de Pequenas Células do Pulmão/patologia
3.
Phys Rev Lett ; 108(26): 261603, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-23004960

RESUMO

Using data collected with the D0 detector at the Fermilab Tevatron Collider, corresponding to 5.3 fb(-1) of integrated luminosity, we search for violation of Lorentz invariance by examining the tt[over ¯] production cross section in lepton+jets final states. We quantify this violation using the standard-model extension framework, which predicts a dependence of the tt[over ¯] production cross section on sidereal time as the orientation of the detector changes with the rotation of the Earth. Within this framework, we measure components of the matrices (c(Q))(µν33) and (c(U))(µν33) containing coefficients used to parametrize violation of Lorentz invariance in the top quark sector. Within uncertainties, these coefficients are found to be consistent with zero.

4.
Rev Neurol (Paris) ; 167(10): 648-54, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21903234

RESUMO

In recent years, advances in our understanding of the biology of low-grade gliomas (LGG) and high-grade gliomas (HGG) have driven new paradigms in molecular markers, diagnostic imaging, operative techniques and technologies, and adjuvant therapies. Taken together, these developments are collectively pushing the envelope towards improved quality of life and survival. Here, we review the recent literature to synthesize a comprehensive review of the value of extent of resection for LGGs and HGGs in the modern neurosurgical era.


Assuntos
Neoplasias Encefálicas/cirurgia , Glioma/cirurgia , Procedimentos Neurocirúrgicos , Biomarcadores Tumorais , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica , Quimiorradioterapia Adjuvante , Terapia Combinada , Glioma/patologia , Humanos , Imageamento por Ressonância Magnética , Vias Neurais/patologia , Prognóstico , Resultado do Tratamento
5.
J Neurooncol ; 99(3): 423-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20853019

RESUMO

To formulate Functional Assessment of Cancer Therapy-Meningioma (FACT-MNG), a web-based tumor site-specific outcome instrument for assessing intracranial meningioma patients following surgical resection or stereotactic radiosurgery. We surveyed the relevant literature available on intracranial meningioma surgery and subsequent outcomes (38 papers), making note of which, if any, QOL/outcome instruments were utilized. None of the surgveyed papers included QOL assessment specific to tumor site. We subsequently developed questions that were relevant to the signs and symptoms that characterize each of 11 intracranial meningioma sites, and incorporated them into a modified combination of the Functional Assessment of Cancer Therapy-Brain (FACT-BR) and SF36 outcome instruments, thereby creating a new tumor site-specific outcome instrument, FACT-MNG. With outcomes analysis of surgical and radiosurgical treatments becoming more important, measures of the adequacy and success of treatment are needed. FACT-MNG represents a first effort to formalize such an instrument for meningioma patients. Questions specific to tumor site will allow surgeons to better assess specific quality of life issues not addressed in the past by more general questionnaires.


Assuntos
Internet , Neoplasias Meníngeas/cirurgia , Meningioma/cirurgia , Avaliação de Resultados em Cuidados de Saúde , Qualidade de Vida , Radiocirurgia , Humanos
6.
Brain Lang ; 191: 17-27, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30769167

RESUMO

Human language is organized along two main processing streams connecting posterior temporal cortex and inferior frontal cortex in the left hemisphere, travelling dorsal and ventral to the Sylvian fissure. Some views propose a dorsal motor versus ventral semantic division. Others propose division by combinatorial mechanism, with the dorsal stream responsible for combining elements into a sequence and the ventral stream for forming semantic dependencies independent of sequential order. We acquired data from direct cortical stimulation in the left hemisphere in 17 neurosurgical patients and subcortical resection in a subset of 10 patients as part of awake language mapping. Two language tasks were employed: a sentence generation (SG) task tested the ability to form sequential and semantic dependencies, and a picture-word interference (PWI) task manipulated semantic interference. Results show increased error rates in the SG versus PWI task during subcortical testing in the dorsal stream territory, and high error rates in both tasks in the ventral stream territory. Connectivity maps derived from diffusion imaging and seeded in the tumor sites show that patients with more errors in the SG than in the PWI task had tumor locations associated with a dorsal stream connectivity pattern. Patients with the opposite pattern of results had tumor locations associated with a more ventral stream connectivity pattern. These findings provide initial evidence using fiber tract disruption with electrical stimulation that the dorsal pathways are critical for organizing words in a sequence necessary for sentence generation, and the ventral pathways are critical for processing semantic dependencies.


Assuntos
Córtex Cerebral/fisiologia , Conectoma , Semântica , Adulto , Estimulação Elétrica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vias Neurais , Vigília
7.
Cell Death Differ ; 14(3): 548-58, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16946731

RESUMO

Although autophagy enhances cell survival in nutrient-deprived cells by increasing adenosine triphosphate (ATP) production, it remains unclear if autophagy functions similarly in cells treated with cytotoxic chemotherapy agents. To address this issue, we measured both the ability of DNA damaging agents (Temozolomide, and Etoposide) to induce an autophagy-dependent production of ATP, and the effects of modulation of autophagy on drug-induced cell death. Both drugs induced an autophagy-associated increase in ATP production in multiple glioma cell lines. The drug-induced ATP surge could not be blocked by glucose starvation, but could be blocked by preincubation with the autophagy inhibitor 3-methyladenine (3-MA), an siRNA targeting beclin 1, or the mitochondrial inhibitor oligomycin. Inhibition of autophagy-induced ATP production increased non-apoptotic cell death associated with micronucleation, while restoration of the 3-MA-inhibited ATP surge by addition of pyruvate suppressed cell death. These results show that DNA damaging agents induce an autophagy-associated ATP surge that protects cells and may contribute to drug resistance.


Assuntos
Trifosfato de Adenosina/metabolismo , Autofagia , Dano ao DNA , Dacarbazina/análogos & derivados , Glioma/metabolismo , Trifosfato de Adenosina/fisiologia , Antineoplásicos Alquilantes/farmacologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Morte Celular , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Etoposídeo/farmacologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Oxirredução , Fosforilação , Temozolomida
8.
AJNR Am J Neuroradiol ; 28(6): 1078-84, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17569962

RESUMO

BACKGROUND AND PURPOSE: Glioblastoma multiforme (GBM) and single brain metastasis (MET) are the 2 most common malignant brain tumors that can appear similar on anatomic imaging but require vastly different treatment strategy. The purpose of our study was to determine whether the peak height and the percentage of signal intensity recovery derived from dynamic susceptibility-weighted contrast-enhanced (DSC) perfusion MR imaging could differentiate GBM and MET. MATERIALS AND METHODS: Forty-three patients with histopathologic diagnosis of GBM (n=27) or MET (n=16) underwent DSC perfusion MR imaging in addition to anatomic MR imaging before surgery. Regions of interest were drawn around the nonenhancing peritumoral T2 lesion (PTL) and the contrast-enhancing lesion (CEL). T2* signal intensity-time curves acquired during the first pass of gadolinium contrast material were converted to the changes in relaxation rate to yield T2* relaxivity (Delta R2*) curve. The peak height of maximal signal intensity drop and the percentage of signal intensity recovery at the end of first pass were measured for each voxel in the PTL and CEL regions of the tumor. RESULTS: The average peak height for the PTL was significantly higher (P=.04) in GBM than in MET. The average percentage of signal intensity recovery was significantly reduced in PTL (78.4% versus 82.8%; P=.02) and in CEL (62.5% versus 80.9%, P<.01) regions of MET compared with those regions in the GBM group. CONCLUSIONS: The findings of our study show that the peak height and the percentage of signal intensity recovery derived from the Delta R2* curve of DSC perfusion MR imaging can differentiate GBM and MET.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/secundário , Carcinoma/diagnóstico , Carcinoma/secundário , Glioblastoma/diagnóstico , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Adulto , Idoso , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Perfusão/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
9.
Cancer Res ; 61(5): 1957-63, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280752

RESUMO

Temozolomide (TMZ) is a DNA-methylating agent that has recently been introduced into Phase II and III trials for the treatment of gliomas. TMZ produces O6-methylguanine in DNA, which mispairs with thymine during the next cycle of DNA replication. Subsequent futile cycles of DNA mismatch repair can lead to a p53-associated apoptotic cell death, although this mechanism has been described mostly in hematopoietic neoplasms. We studied the action of TMZ in gliomas and the role p53 might play by using U87 glioma cells that were either p53-wild-type or p53-deficient (by virtue of expression of the viral oncoprotein E6). LN-Z308 cells, in which p53 gene is deleted, were also used. p53-proficient U87 MG cells underwent a prolonged, p53- and p21(Waf1/Cip1)-associated G2-M arrest beginning 2 days after TMZ treatment. Although very few of these cells underwent apoptosis, most underwent senescence over a 10-day period. p53-deficient (E6-transfected U87 and LN-Z308) cells similarly underwent G2-M arrest in response to TMZ, but this arrest was accompanied by only minor changes in p53 or p21(Waf1/Cip1) and was reversed within 7 days of TMZ treatment in association with the appearance of cells with either 8n or subG1 DNA content. These results suggest that glioma cells respond to TMZ by undergoing G2-M arrest. p53 is not necessary for this G2-M arrest to occur but is important in the duration of G2-M arrest and in the ultimate fate of TMZ-treated cells. Therefore, the integrity of the G2-M cell cycle checkpoint may be important in the cytotoxicity of TMZ in glioma cells.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Fase G2/efeitos dos fármacos , Glioblastoma/patologia , Mitose/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Pareamento Incorreto de Bases , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Fase G2/fisiologia , Glioblastoma/tratamento farmacológico , Humanos , Mitose/fisiologia , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Temozolomida , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
10.
Cancer Res ; 61(15): 5843-9, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11479224

RESUMO

Temozolomide (TMZ) produces O(6)-methylguanine in DNA, which in turn mispairs with thymine, triggering futile DNA mismatch repair (MMR) and ultimately cell death. We found previously that in p53-proficient human glioma cells, TMZ-induced futile DNA MMR resulted not in apoptosis but rather in prolonged, p53- and p21-associated G(2)-M arrest and senescence. Additionally, p53-deficient cells were relatively more TMZ resistant than p53-deficient glioma cells, which underwent only transient G(2)-M arrest before death by mitotic catastrophe. These results suggested that prolonged G(2)-M arrest might protect cells from TMZ-induced cytotoxicity. In the present study, we therefore focused on the mechanism by which TMZ induces G(2)-M arrest and on whether inhibition of such G(2)-M arrest might sensitize glioma cells to TMZ-induced toxicity. U87MG glioma cells treated with TMZ underwent G(2)-M arrest associated with Chk1 activation and phosphorylation of both cdc25C and cdc2. These TMZ-induced effects were inhibited by the Chk1 kinase inhibitor UCN-01. Although not in itself toxic, UCN-01 increased the cytotoxicity of TMZ 5-fold, primarily by inhibiting cellular senescence and increasing the percentage of cells bypassing G(2)-M arrest and undergoing mitotic catastrophe. In addition to enhancing TMZ-induced cytotoxicity in p53-proficient cells, UCN-01 also blocked TMZ-induced Chk1 activation and transient G(2)-M arrest in p53-deficient U87MG-E6 cells and similarly enhanced TMZ-induced mitotic catastrophe and cell death. Taken together, these results indicate that Chk1 links TMZ-induced MMR to G(2)-M arrest. Furthermore, inhibition of the cytoprotective G(2) arrest pathway sensitizes cells to TMZ-induced cytotoxicity and may represent a novel, mechanism-based means of increasing TMZ efficacy in both p53 wild-type and p53 mutant glioma cells.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Dacarbazina/toxicidade , Fase G2/fisiologia , Glioblastoma/tratamento farmacológico , Inibidores de Proteínas Quinases , Proteínas Quinases , Proteína Supressora de Tumor p53/fisiologia , Alcaloides/farmacologia , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Dacarbazina/análogos & derivados , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Fase G2/efeitos dos fármacos , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Mitose/efeitos dos fármacos , Mitose/fisiologia , Fosforilação/efeitos dos fármacos , Estaurosporina/análogos & derivados , Temozolomida , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Fosfatases cdc25/metabolismo
11.
Cancer Res ; 56(10): 2405-10, 1996 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8625319

RESUMO

The product of the p16/CDKN2 locus, p16ink4, negatively regulates the cell cycle through binding and inactivation of cyclin-dependent kinases (CDKs) 4 and 6. This locus is frequently targeted for deletion in cell lines and primary tumor tissues. In gliomas, although up to 50% do not have detectable expression of p16/CDKN2 protein or mRNA, often the gene is wild type in sequence. Here, we tested the hypothesis that transcriptional repression of p16/CDKN2 in gliomas may be mediated by aberrant methylation of the CpG island, which is in the 5' region of the locus. Partial rather than complete p16/CDKN2 methylation was detected in 24% (10 of 42) of the gliomas, regardless of tumor grade, but was not observed in normal brain (0 of 10). We tested whether this partial methylation could inhibit expression in a human tumor cell line in which suppressed p16/CDKN2 expression was associated with both methylation and tightly compacted chromatin around the p16/CDKN2 promoter. Exposure of these cells to 5-aza-2-deoxycytidine resulted in a dramatic increase in promoter accessibility and induction of p16/CDKN2 expression, indicating that chromatin structure, CpG island methylation, and p16/CDKN2 expression are intimately associated. Taken together, these data suggest that methylation occurs in only a subset of cells within gliomas and that the methylation-associated inactivation of p16/CDKN2 expression observed in many common human cancers may mechanistically result from structural changes in the chromatin containing the p16/CDKN2 locus.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Transporte/biossíntese , Cromatina/ultraestrutura , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Astrocitoma/genética , Astrocitoma/metabolismo , Astrocitoma/patologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Química Encefálica , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Transporte/genética , Cromatina/genética , Inibidor p16 de Quinase Dependente de Ciclina , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA de Neoplasias/química , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/metabolismo , Glioma/patologia , Humanos , Metilação , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Sequências Reguladoras de Ácido Nucleico , Células Tumorais Cultivadas/efeitos dos fármacos
12.
Cancer Res ; 53(14): 3416-20, 1993 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8324751

RESUMO

We assayed the activity of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) in 60 human brain tumors to assess the effects of tumorigenesis in brain on DNA repair capability. Activity was not detectable (< 0.5 fmol/10(6) cells, i.e., < 300 molecules/cells) in 27% of the tumors. Measurable MGMT varied by more than 2 orders of magnitude, 0.5-104.1 fmol/10(6) cells. Mean tumor MGMT levels did not differ between the sexes but did vary widely between diagnostic groups. A significant inverse correlation was observed between tumor MGMT activity and patient age. We also assayed MGMT activity in overlying, histologically tumor-free brain resected with 25 tumors. Of these samples, 52% had no detectable MGMT activity, and the remainder had activity comparable to that in tumors ranging from 0.7-21.8 fmol/10(6) cells. MGMT activity in normal brain was also inversely correlated with patient age. For 15 of 25 (60%) paired samples, tumor activity was 2 to > 38-fold greater than that of normal brain; for 4 pairs (16%) tumor activity was 2.5 to > 17-fold lower than that of normal brain; the remaining 6 (24%) had no detectable activity in both tumor and normal tissue. These differences in the magnitudes and distributions of activities for tumor versus normal brain tissue were significant (P = 0.02), demonstrating that tumorigenesis in brain is often accompanied by marked elevation of MGMT.


Assuntos
Neoplasias Encefálicas/enzimologia , Encéfalo/enzimologia , Metiltransferases/análise , Adolescente , Adulto , Idoso , Astrocitoma/enzimologia , Criança , Pré-Escolar , Ependimoma/enzimologia , Feminino , Glioma/enzimologia , Humanos , Lactente , Masculino , Meduloblastoma/enzimologia , Pessoa de Meia-Idade , O(6)-Metilguanina-DNA Metiltransferase , Oligodendroglioma/enzimologia
13.
Cancer Res ; 53(23): 5663-8, 1993 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8242621

RESUMO

By altering the accessibility of DNA sequences for alkylation or platination, and/or for subsequent repair, topoisomerase II can potentially affect the level of DNA interstrand cross-links induced in cells by bifunctional agents. In this study, we investigated the extent to which inhibition of topoisomerase II activity in a human glioblastoma multiforme cell line alters the kinetics of both the formation and the repair of total genomic DNA interstrand cross-links, as well as the sensitivity of the tumor cells to cis-diamminedichloroplatinum II (cis-DDP) and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). Cells were incubated with and without 200 microM novobiocin, a known topoisomerase II inhibitor, for 24 h, followed by exposure to 50 microM BCNU and 25 microM cis-DDP. DNA interstrand cross-linking was determined at various time points over 72 h, using a modified ethidium bromide-DNA binding assay. Sensitivity of the cells to cis-DDP and BCNU was also determined with and without novobiocin pretreatment with 200 microM novobiocin. This concentration of novobiocin showed no significant direct cytotoxicity, although it inhibited topoisomerase II activity in tumor cell nuclear extracts by 73%. A significant decrease in the rate of repair of both cis-DDP and BCNU induced DNA interstrand cross-links, with a corresponding decrease in the clonogenic survival of the cells, was observed following novobiocin exposure. Although the peak cross-link indices of novobiocin-treated cells relative to controls were not significantly increased, residual DNA cross-linking in the cells after 72 h was increased by 1.4-fold for BCNU and 3-fold for cells treated with cis-DDP, thus, indicating a greater effect of topoisomerase II on cross-link repair than on cross-link formation. These data suggest that inhibition of topoisomerase II may provide a potentially effective clinical strategy for sensitizing human brain tumors, and possibly other tumors as well, to DNA cross-linking anticancer agents.


Assuntos
Carmustina/farmacologia , Cisplatino/farmacologia , Reparo do DNA/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Novobiocina/farmacologia , Inibidores da Topoisomerase II , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Cancer Res ; 58(5): 1068-73, 1998 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9500473

RESUMO

The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) confers resistance to therapeutic methylating and chloroethylating agents in human brain tumor-derived cell lines. In this work, we assayed MGMT activity in 152 adult gliomas to establish correlates with patient and tumor characteristics. We also assayed MGMT in histologically normal brain adjacent to 87 tumors to characterize changes in activity accompanying neurocarcinogenesis. MGMT activity was detectable in 76% (115 of 152) of tumors, ranging approximately 300-fold from 0.30 to 89 fmol/10(6) cells (180-57,000 molecules/cell). Mean activity was 6.6 +/- 13 fmol/10(6) cells and varied 4-fold among diagnostic groups. The mean for oligodendrogliomas was 2-fold lower (P < 0.03), and for mixed oligodendroglioma-astrocytomas, the mean was 4-fold lower (P < 0.006) than for astroglial tumors. Twenty-five % of gliomas had no detectable MGMT activity (Mer- phenotype; < 0.25 fmol/10(6) cells or 150 molecules/cell). Glioma MGMT was inversely correlated with age (P < 0.01), consistent with the observed age dependence in the progenitor tissue of brain tumors (J. R. Silber et al., Proc. Natl. Acad. Sci. USA, 93: 6941-6946, 1996). Neither MGMT activity nor proportion of Mer- tumors differed by sex. Glioma MGMT was correlated with degree of aneuploidy (P < 0.006) but not with fraction of S-phase cells. Mean activity in tumors was 5-fold higher than in adjacent histologically normal brain (5.0 +/- 7.6 versus 1.1 +/- 1.9 fmol/10(6) cells; P < 0.001). Notably, elevation of tumor activity was observed in 62% of tissue pairs, ranging from 2-fold to > 105-fold. Moreover, 64% of Mer- normal tissue was accompanied by Mer+ tumor. These observations indicate that expression of MGMT activity is frequently activated and/or increased during human neurocarcinogenesis, and that the enhancement is not related to proliferation per se. Significantly, enhanced MGMT activity may heighten the resistance of brain tumors to therapeutic alkylating agents.


Assuntos
Biomarcadores Tumorais , Neoplasias Encefálicas/enzimologia , Glioma/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Divisão Celular , Ativação Enzimática , Feminino , Glioma/patologia , Glioma/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade
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 ; 54(10): 2794-9, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168112

RESUMO

Glioblastomas were examined for abnormalities in fibroblast growth factor receptor (FGFR) expression by polymerase chain reaction and immunocytochemical analysis. Polymerase chain reaction analysis demonstrated that FGFR1 mRNA levels were significantly higher in glioblastomas than in normal brain adjacent to the tumor or in untransformed human brain. These results were consistent with immunocytochemical localization of FGFR1 protein in glioblastomas: glioblastoma cells exhibited intense FGFR1 immunoreactivity in frozen sections of tumor and low to undetectable FGFR1 immunoreactivity in adjacent normal brain or in normal white matter obtained from patients without neoplastic disease. Endothelial cells of capillaries and larger vessels within the tumor were devoid of FGFR1 immunoreactivity. All glioblastomas evaluated in the present study expressed FGFR1 mRNA and FGFR1 immunoreactivity. Examination of the FGFR1 gene by Southern blot analysis indicated that overexpression of FGFR1 mRNA in glioblastomas did not result from gene amplification. These results indicate that glioblastoma cells, in contrast to endothelial cells within the tumor, display increased levels of FGFR1. Therefore, FGFR1 signal transduction may be associated with increased autocrine growth activity of tumor cells and is probably not related to the increased endothelial cell proliferation associated with these tumors.


Assuntos
Química Encefálica , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/química , Glioblastoma/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Sequência de Bases , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Neoplásico/análise , Receptores de Fatores de Crescimento de Fibroblastos/análise
18.
Cancer Res ; 45(8): 3460-4, 1985 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3860285

RESUMO

The 9L-2, 9L-7, and 9L-8 cell lines, derived from the 9L in vivo rat brain tumor, were treated with nitrosoureas that can alkylate and cross-link DNA and carbamoylate intracellular molecules to various extents. Compared to 9L cells, 9L-2 cells were very resistant to the cytotoxic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea, and to 2-[3-(2-chloroethyl)-3-nitrosoureido]-D-deoxyglucopyranose. The sensitivity of 9L-7 and 9L-8 cell lines to these drugs was intermediate between 9L and 9L-2. Treatment of 9L, 9L-2, 9L-7, and 9L-8 cell lines with 1,3-bis(trans-4-hydroxycyclohexyl)-1-nitrosourea produced approximately the same level of cell kill. Compared to 9L cells, 9L-2 cells are 10-fold more resistant to the cytotoxic effects, 34-fold more resistant to the induction of sister chromatid exchanges, and have 40% fewer DNA interstrand cross-links caused by treatment with 3-(4-amino-2-methyl-5-pyrimidinyl)methyl-1-(2-chloroethyl)-1-nitrosourea . In contrast, treatment of 9L and 9L-2 cells with 1-ethylnitrosourea produced approximately the same level of cell kill and induction of sister chromatid exchanges. Our results suggest that the resistance of 9L-2, 9L-7, and 9L-8 cells is related to DNA cross-linking and not to alkylation or carbamoylation. We studied the effects of other agents that form DNA cross-links with structures different from those formed by treatment with chloroethylnitrosoureas (CENUs) in 9L and 9L-2 cells. In contrast to results obtained with CENUs, 9L-2 cells were 2-fold more sensitive to the cytotoxic effects, 2-fold more sensitive to the induction of sister chromatid exchanges, and had 3-fold more cross-links formed than 9L cells treated with nitrogen mustard. However, the amount of cell kill, number of sister chromatid exchanges induced, and the DNA cross-linking were the same for 9L and 9L-2 cells treated with cis-diamminedichlorplatinum(II). Our results indicate that cellular resistance to CENUs is highly specific and that the mechanism of resistance does not allow cross-resistance with other DNA cross-linking agents. These and other results suggest that when DNA repair processes mediate cellular resistance to CENUs, other cross-linking agents will not be cross-resistant unless they form alkylation products that are affected by repair processes that mediate resistance to CENUs.


Assuntos
Antineoplásicos/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , DNA/metabolismo , Compostos de Nitrosoureia/farmacologia , Animais , Carmustina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA , Resistência a Medicamentos , Nimustina , Ratos , Troca de Cromátide Irmã/efeitos dos fármacos
19.
Cancer Res ; 55(9): 1941-5, 1995 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-7728764

RESUMO

P16INK4 is a cell cycle regulator that specifically binds to and inactivates cyclin-dependent kinase 4 (CDK4). Its encoding gene (p16/CDKN2) maps to chromosome 9p21, a region that undergoes frequent loss of heterozygosity in a variety of human tumors. We have analyzed the p16/CDKN2 gene and its expression in a series of primary glioma samples. Although homozygous deletion or mutation of the p16/CDKN2 gene was uncommon in this series and P16INK4 protein was detectable in all grade II tumors, it was present in only 50% of grade III and grade IV samples. Conversely, in some grade IV tumors that level of P16INK4 protein was elevated; in these cases, its target, CDK4, was amplified and overexpressed. These results suggest: (a) the involvement of P16INK4 in glioma progression; (b) that mechanisms other than mutation or deletion can down-regulate expression of the p16/CDKN2 gene; and (c) that the balance between CDK4 and its cognate inhibitor, P16INK4, may confer a cell growth advantage and facilitate tumor progression.


Assuntos
Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteínas de Transporte/genética , Glioblastoma/genética , Glioblastoma/patologia , Sequência de Bases , Western Blotting , Proteínas de Transporte/análise , Inibidor p16 de Quinase Dependente de Ciclina , Progressão da Doença , Deleção de Genes , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor/genética , Humanos , Dados de Sequência Molecular , Mutação , Inibidores de Proteínas Quinases , RNA Mensageiro/análise , RNA Mensageiro/genética , Transcrição Gênica
20.
Cancer Res ; 55(6): 1261-6, 1995 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-7882319

RESUMO

Perturbation of the DNA repair process appears to be responsible for the occurrence of a number of human diseases, which are usually associated with a propensity to develop internal malignancies and/or disorders of the central nervous system. We have been interested in the possibility that a subtle abnormality in DNA repair competency might be associated with the transformation of nonmalignant cells to the malignant state. To study this question, we assayed malignant and nonmalignant brain tissues from 19 individuals for mRNA expression levels of the human DNA repair genes ERCC1, ERCC2, and XPAC and for differential splicing of the ERCC1 transcript. We separately compared expression levels of these genes in the following situations: concordance of expression within malignant tissues; concordance of expression within nonmalignant tissues; concordance between malignant and nonmalignant tissues within individuals of the cohort; and concordance of gene expression between two nonmalignant tissue sites within a single individual. Linear regression analyses of mRNA values obtained suggested orderly concordance of these three DNA repair genes in nonmalignant tissues within the patient cohort and an excellent concordance of these genes between two separate biopsy sites from the same individual. In contrast, malignant tissues showed disruption of concordance between the full-length ERCC1 transcript and ERCC2, which have excision and helicase functions, respectively. Furthermore, within the same individuals, malignant tissues were discordant with nonmalignant tissues for ERCC1 and ERCC2, although concordance for XPAC was preserved. These data suggest that one molecular characteristic of human malignancy may be the disruption of the normal relationship between the excision and the helicase functions of the nucleotide excision repair pathway.


Assuntos
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , DNA Helicases , Reparo do DNA , Endonucleases , Proteínas/genética , RNA Mensageiro/análise , Fatores de Transcrição , Dano ao DNA , Proteínas de Ligação a DNA/genética , Humanos , Reação em Cadeia da Polimerase , Proteína de Xeroderma Pigmentoso Grupo A , Proteína Grupo D do Xeroderma Pigmentoso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA