Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Curr Pain Headache Rep ; 22(9): 60, 2018 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-29987680

RESUMO

Ultrasound plays a significant role in the diagnosis and treatment of pain, with significant literature reaching back many years, especially with regard to diagnostic ultrasound and its use for guiding needle-based delivery of drugs. Advances in ultrasound over at least the last decade have opened up new areas of inquiry and potential clinical efficacy in the context of pain diagnosis and treatment. Here we offer an overview of the recent literature associated with ultrasound and pain in order to highlight some promising frontiers at the intersection of these two subjects. We focus first on peripheral application of ultrasound, for which there is a relatively rich, though still young, literature. We then move to central application of ultrasound, for which there is little literature but much promise.


Assuntos
Manejo da Dor , Dor/diagnóstico por imagem , Sistema Nervoso Periférico/cirurgia , Ultrassonografia , Animais , Encéfalo/cirurgia , Humanos , Resultado do Tratamento , Ultrassonografia/métodos
2.
BBA Clin ; 3: 1-10, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25558448

RESUMO

BACKGROUND: CpG methylation in the O6-methylguanine-DNA methyltransferase (MGMT) promoter is associated with better outcome following alkylating agent chemotherapy in glioblastoma (GBM) and anaplastic glioma (AG). To what extent improved response reflects low or absent MGMT activity in glioma tissue has not been unequivocally assessed. This information is central to developing anti-resistance therapies. METHODS: We examined the relationship of MGMT activity in 91 GBMs and 84 AGs with progression-free survival (PFS) following alkylator therapy and with promoter methylation status determined by methylation-specific PCR (MSP). RESULTS: Cox regression analysis revealed that GBMs with high activity had a significantly greater risk for progression in dichotomous (P ≤ 0.001) and continuous (P ≤ 0.003) models, an association observed for different alkylator regimens, including concurrent chemo-radiation with temozolomide. Analysis of MGMT promoter methylation status in 47 of the GBMs revealed that methylated tumors had significantly lower activity (P ≤ 0.005) and longer PFS (P ≤ 0.036) compared to unmethylated tumors, despite overlapping activities. PFS was also significantly greater in methylated vs. unmethylated GBMs with comparable activity (P ≤ 0.005), and among unmethylated tumors with less than median activity (P ≤ 0.026), suggesting that mechanisms in addition to MGMT promote alkylator resistance. Similar associations of MGMT activity with PFS and promoter methylation status were observed for AGs. CONCLUSIONS: Our results provide strong support for the hypotheses that MGMT activity promotes alkylator resistance and reflects promoter methylation status in malignant gliomas. GENERAL SIGNIFICANCE: MGMT activity is an attractive target for anti-resistance therapy regardless of methylation status.

3.
Chem Res Toxicol ; 26(1): 156-68, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23234400

RESUMO

Derivatives of methyl 3-(1-methyl-5-(1-methyl-5-(propylcarbamoyl)-1H-pyrrol-3-ylcarbamoyl)-1H-pyrrol-3-ylamino)-3-oxopropane-1-sulfonate (1), a peptide-based DNA minor groove binding methylating agent, were synthesized and characterized. In all cases, the N-terminus was appended with an O-methyl sulfonate ester, while the C-terminus group was varied with nonpolar and polar side chains. In addition, the number of pyrrole rings was varied from 2 (dipeptide) to 3 (tripeptide). The ability of the different analogues to efficiently generate N3-methyladenine was demonstrated as was their selectivity for minor groove (N3-methyladenine) versus major groove (N7-methylguanine) methylation. Induced circular dichroism studies were used to measure the DNA equilibrium binding properties of the stable sulfone analogues; the tripeptide binds with affinity that is >10-fold higher than that of the dipeptide. The toxicities of the compounds were evaluated in alkA/tag glycosylase mutant E. coli and in human WT glioma cells and in cells overexpressing and under-expressing N-methylpurine-DNA glycosylase, which excises N3-methyladenine from DNA. The results show that equilibrium binding correlates with the levels of N3-methyladenine produced and cellular toxicity. The toxicity of 1 was inversely related to the expression of MPG in both the bacterial and mammalian cell lines. The enhanced toxicity parallels the reduced activation of PARP and the diminished rate of formation of aldehyde reactive sites observed in the MPG knockdown cells. It is proposed that unrepaired N3-methyladenine is toxic due to its ability to directly block DNA polymerization.


Assuntos
Alquilantes/síntese química , DNA/química , Adenina/análogos & derivados , Adenina/química , Alquilantes/química , Alquilantes/toxicidade , Animais , Bovinos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Glicosilases/química , DNA Glicosilases/metabolismo , Metilação de DNA , Escherichia coli/enzimologia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Humanos , Peptídeos/química , Peptídeos/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Termodinâmica
4.
Front Oncol ; 2: 176, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23230562

RESUMO

Alkylating agents have long played a central role in the adjuvant therapy of glioblastoma (GBM). More recently, inclusion of temozolomide (TMZ), an orally administered methylating agent with low systemic toxicity, during and after radiotherapy has markedly improved survival. Extensive in vitro and in vivo evidence has shown that TMZ-induced O(6)-methylguanine (O(6)-meG) mediates GBM cell killing. Moreover, low or absent expression of O(6)-methylguanine-DNA methyltransferase (MGMT), the sole human repair protein that removes O(6)-meG from DNA, is frequently associated with longer survival in GBMs treated with TMZ, promoting interest in developing inhibitors of MGMT to counter resistance. However, the clinical efficacy of TMZ is unlikely to be due solely to O(6)-meG, as the agent produces approximately a dozen additional DNA adducts, including cytotoxic N3-methyladenine (3-meA) and abasic sites. Repair of 3-meA and abasic sites, both of which are produced in greater abundance than O(6)-meG, is mediated by the base excision repair (BER) pathway, and occurs independently of removal of O(6)-meG. These observations indicate that BER activities are also potential targets for strategies to potentiate TMZ cytotoxicity. Here we review the evidence that 3-meA and abasic sites mediate killing of GBM cells. We also present in vitro and in vivo evidence that alkyladenine-DNA glycosylase, the sole repair activity that excises 3-meA from DNA, and Ape1, the major human abasic site endonuclease, mediate TMZ resistance in GBMs and represent potential anti-resistance targets.

5.
Biochim Biophys Acta ; 1826(1): 71-82, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22244911

RESUMO

Gliomas are the most frequent adult primary brain tumor, and are invariably fatal. The most common diagnosis glioblastoma multiforme (GBM) afflicts 12,500 new patients in the U.S. annually, and has a median survival of approximately one year when treated with the current standard of care. Alkylating agents have long been central in the chemotherapy of GBM and other gliomas. The DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT), the principal human activity that removes cytotoxic O(6)-alkylguanine adducts from DNA, promotes resistance to anti-glioma alkylators, including temozolomide and BCNU, in GBM cell lines and xenografts. Moreover, MGMT expression assessed by immunohistochemistry, biochemical activity or promoter CpG methylation status is associated with the response of GBM to alkylator-based therapies, providing evidence that MGMT promotes clinical resistance to alkylating agents. These observations suggest a role for MGMT in directing adjuvant therapy of GBM and other gliomas. Promoter methylation status is the most clinically tractable measure of MGMT, and there is considerable enthusiasm for exploring its utility as a marker to assign therapy to individual patients. Here, we provide an overview of the biochemical, genetic and biological characteristics of MGMT as they relate to glioma therapy. We consider current methods to assess MGMT expression and discuss their utility as predictors of treatment response. Particular emphasis is given to promoter methylation status and the methodological and conceptual impediments that limit its use to direct treatment. We conclude by considering approaches that may improve the utility of MGMT methylation status in planning optimal therapies tailored to individual patients.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Glioma/tratamento farmacológico , Glioma/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Neoplasias Encefálicas/genética , Glioma/genética , Humanos , Terapia de Alvo Molecular , O(6)-Metilguanina-DNA Metiltransferase/genética
6.
Int J Cancer ; 129(10): 2370-9, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21207372

RESUMO

Apurinic/apyrimidinic endonuclease (Ap endo) is a key DNA repair activity that confers radiation resistance in human cells. Here we examined the association between Ap endo activity and response to radiotherapy in pediatric ependymomas, tumors for which treatment options are limited and survival rates are only about 50%. We assayed Ap endo activity in 36 ependymomas and expression of Ape1/Ref-1, the predominant Ap endo activity in humans, in 44 tumors by immunostaining. Cox proportional hazards regression models were used to analyze the association of activity or expression with progression-free survival or with overall survival. Activity varied 13-fold and was not associated with tumor or patient characteristics. In univariate models with Ap endo activity entered as a continuous variable, the hazard ratio for progression increased by a factor of 2.18 for every 0.01 unit increase in activity (p ≤ 0.003) in 24 grade II ependymomas. Risk for death increased by a factor of 1.89 (p ≤ 0.02) in the same population. The fraction of Ape1/Ref-1 immunopositive cells varied widely within individual tumors and was not associated with either progression-free or with overall survival. Suppressing Ap endo activity in pediatric ependymoma cells significantly increased radiation sensitivity, suggesting that the association of activity with radiation response reflected, at least in part, repair of radiation-induced DNA lesions. Our data indicate that Ap endo activity is predictive of outcome following radiotherapy, and suggest that Ape1/Ref-1 promotes radiation resistance in pediatric ependymomas. Our findings support the use of inhibitors of Ap endo activity to overcome resistance.


Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/radioterapia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Ependimoma/enzimologia , Ependimoma/radioterapia , Adolescente , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Criança , Intervalo Livre de Doença , Ependimoma/imunologia , Ependimoma/mortalidade , Feminino , Humanos , Lactente , Masculino , Tolerância a Radiação
7.
Mol Cancer Ther ; 9(5): 1208-18, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20457618

RESUMO

Concurrent treatment with the methylating agent temozolomide during radiotherapy has yielded the first significant improvement in the survival of adult glioblastomas (GBM) in the last three decades. However, improved survival is observed in a minority of patients, most frequently those whose tumors display CpG methylation of the O(6)-methylguanine (O(6)-meG)-DNA methyltransferase (MGMT) promoter, and adult GBMs remain invariably fatal. Some, although not all, preclinical studies have shown that temozolomide can increase radiosensitivity in GBM cells that lack MGMT, the sole activity in human cells that removes O(6)-meG from DNA. Here, we systematically examined the temozolomide dose dependence of radiation killing in established GBM cell lines that differ in ability to remove O(6)-meG or tolerate its lethality. Our results show that minimally cytotoxic doses of temozolomide can produce dose-dependent radiosensitization in MGMT-deficient cells, MGMT-proficient cells, and MGMT-deficient cells that lack mismatch repair, a process that renders cells tolerant of the lethality of O(6)-meG. In cells that either possess or lack MGMT activity, radiosensitization requires exposure to temozolomide before but not after radiation and is accompanied by formation of double-strand breaks within 45 minutes of radiation. Moreover, suppressing alkyladenine-DNA glycosylase, the only activity in human cells that excises 3-methyladenine from DNA, reduces the temozolomide dose dependence of radiosensitization, indicating that radiosensitization is mediated by 3-methyladenine as well as by O(6)-meG. These results provide novel information on which to base further mechanistic study of radiosensitization by temozolomide in human GBM cells and to develop strategies to improve the outcome of concurrent temozolomide radiotherapy.


Assuntos
Neoplasias Encefálicas/radioterapia , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Dacarbazina/análogos & derivados , Glioblastoma/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neoplasias Encefálicas/genética , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA/genética , Reparo de Erro de Pareamento de DNA/efeitos da radiação , Metilases de Modificação do DNA/metabolismo , Metilases de Modificação do DNA/fisiologia , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/fisiologia , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Raios gama , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , Dose Máxima Tolerável , Temozolomida , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia
8.
Childs Nerv Syst ; 25(1): 39-45, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18946672

RESUMO

BACKGROUND: Resistance to drug is a major cause of treatment failure in pediatric brain cancer. The multidrug resistance (MDR) phenotype can be mediated by the superfamily of adenosine triphosphate-binding cassette (ABC) transporters. The dynamics of expression of the MDR genes after exposure to chemotherapy, especially the comparison between pediatric brain tumors of different histology, is poorly described. OBJECTIVE: To compare the expression profiles of the multidrug resistance genes ABCB1, ABCC1, and ABCG2 in different neuroepithelial pediatric brain tumor cell lines prior and following short-term culture with vinblastine. METHODS: Immortalized lineages from pilocytic astrocytoma (R286), anaplasic astrocytoma (UW467), glioblastoma (SF188), and medulloblastoma (UW3) were exposed to vinblastine sulphate at different schedules (10 and 60 nM for 24 and 72 h). Relative amounts of mRNA expression were analyzed by real-time quantitative polymerase chain reaction. Protein expression was assessed by immunohistochemistry for ABCB1, ABCC1, and ABCG2. RESULTS: mRNA expression of ABCB1 increased together with augmenting concentration and time of exposure to vinblastine for R286, UW467, and UW3 cell lines. Interestingly, ABCB1 levels of expression diminished in SF188. Following chemotherapy, mRNA expression of ABCC1 decreased in all cell lines other than glioblastoma. ABCG2 expression was influenced by vinblastine only for UW3. The mRNA levels showed consistent association to protein expression in the selected sets of cell lines analyzed. CONCLUSIONS: The pediatric glioblastoma cell line SF188 shows different pattern of expression of multidrug resistance genes when exposed to vinblastine. These preliminary findings may be useful in determining novel strategies of treatment for neuroepithelial pediatric brain tumors.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Vimblastina/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Criança , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
DNA Repair (Amst) ; 6(8): 1127-33, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17500046

RESUMO

The DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT) is a cardinal defense against the mutagenic and carcinogenic effects of alkylating agents. We have reported evidence that absence of detectable MGMT activity (MGMT(-) phenotype) in human brain is a predisposing factor for primary brain tumors that affects ca. 12% of individuals [J.R. Silber, A. Blank, M.S. Bobola, B.A. Mueller, D.D. Kolstoe, G.A. Ojemann, M.S. Berger, Lack of the DNA repair protein O(6)-methylguanine-DNA methyltransferase in histologically normal brain adjacent to primary brain tumors, Proc. Natl. Acad. Sci. U.S.A. 93 (1996) 6941-6946]. We report here that MGMT(-) phenotype in the brain of children and adults, and the apparent increase in risk of neurocarcinogenesis, may arise during gestation. We found that MGMT activity in 71 brain specimens at 6-19 weeks post-conception was positively correlated with gestational age (P

Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/etiologia , Encéfalo/embriologia , Encéfalo/enzimologia , Metilases de Modificação do DNA/deficiência , Enzimas Reparadoras do DNA/deficiência , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Proteínas Supressoras de Tumor/deficiência , Adulto , Encéfalo/crescimento & desenvolvimento , Neoplasias Encefálicas/genética , Criança , Reparo do DNA , Feto/enzimologia , Idade Gestacional , Humanos , Mutação , Fenótipo
10.
Clin Cancer Res ; 13(2 Pt 1): 612-20, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17255284

RESUMO

PURPOSE: Defining the cytotoxicity of individual adducts in DNA is necessary for mechanistic understanding of human brain tumor resistance to therapeutic alkylating agents and for design of DNA repair-related antiresistance strategies. Our purpose is to characterize the sensitivity of human glioma cells to methyl-lexitropsin (Me-lex), a sequence-specific alkylator that produces 3-methyladenine (3-meA) as the predominant (>90%) DNA lesion. EXPERIMENTAL DESIGN: We quantitated the Me-lex cytotoxicity of 10 human glioma cell lines that differ in O(6)-methylguanine (O(6)-meG)-DNA methyltransferase (MGMT) and mismatch repair activity. We used antisense suppression of alkyladenine DNA glycosylase (AAG) and Ape1 to assess the contribution of 3-meA and abasic sites to lethality and measured abasic sites. RESULTS: (a) The LD(10) for Me-lex varied widely among the cell lines. (b) MGMT-proficient lines were more resistant than MGMT-deficient lines, an unexpected finding because Me-lex produces very little O(6)-meG. (c) Suppression of AAG increased Me-lex killing and reduced abasic site content. (d) Suppression of Ape1 increased Me-lex killing and increased abasic site content. (e) Ablation of MGMT had no effect on Me-lex cytotoxicity. CONCLUSIONS: (a) Me-lex is cytotoxic in human glioma cells and AAG promotes resistance, indicating that 3-meA is a lethal lesion in these cells. (b) Abasic sites resulting from 3-meA repair are cytotoxic and Ape1 promotes resistance to these derivative lesions. (c) A factor(s) associated with MGMT expression, other than repair of O(6)-meG, contributes to Me-lex resistance. (d) Me-lex may have clinical utility in the adjuvant therapy of gliomas. (e) AAG and Ape1 inhibitors may be useful in targeting alkylating agent resistance.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/patologia , Glioma/patologia , Netropsina/análogos & derivados , Linhagem Celular Tumoral , Adutos de DNA/metabolismo , Reparo de Erro de Pareamento de DNA , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Humanos , Netropsina/farmacologia , Oligonucleotídeos Antissenso/química , Fatores de Tempo , Células Tumorais Cultivadas
11.
Clin Cancer Res ; 11(20): 7405-14, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16243814

RESUMO

PURPOSE: Apurinic/apyrimidinic endonuclease (Ap endo) is a key DNA repair activity that confers resistance to radiation- and alkylator-induced cytotoxic abasic sites in human cells. We assayed apurinic/apyrimidinic endonuclease activity in medulloblastomas and primitive neuroectodermal tumors (PNET) to establish correlates with tumor and patient characteristics and with response to adjuvant radiation plus multiagent chemotherapy. EXPERIMENTAL DESIGN: Ap endo activity was assayed in 52 medulloblastomas and 10 PNETs from patients 0.4 to 21 years old. Ape1/Ref-1, the predominant human Ap endo activity, was measured in 42 medulloblastomas by immunostaining. Cox proportional hazards regression models were used to analyze the association of activity with time to tumor progression (TTP). RESULTS: Tumor Ap endo activity varied 180-fold and was significantly associated with age and gender. Tumor Ape1/Ref-1 was detected almost exclusively in nuclei. In a multivariate model, with Ap endo activity entered as a continuous variable, the hazard ratio for progression after adjuvant treatment in 46 medulloblastomas and four PNETs increased by a factor of 1.073 for every 0.01 unit increase in activity (P < or = 0.001) and was independent of age and gender. Suppressing Ap endo activity in a human medulloblastoma cell line significantly increased sensitivity to 1,3-bis(2-chlororethyl)-1-nitrosourea and temozolomide, suggesting that the association of tumor activity with TTP reflected, at least in part, abasic site repair. CONCLUSIONS: Our data (a) suggest that Ap endo activity promotes resistance to radiation plus chemotherapy in medulloblastomas/PNETs, (b) provide a potential marker of treatment outcome, and (c) suggest clinical use of Ap endo inhibitors to overcome resistance.


Assuntos
Neoplasias Encefálicas/enzimologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Meduloblastoma/enzimologia , Tumores Neuroectodérmicos Primitivos/enzimologia , Adolescente , Adulto , Antineoplásicos Alquilantes/farmacologia , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/efeitos da radiação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Carmustina/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Criança , Pré-Escolar , Terapia Combinada , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Masculino , Meduloblastoma/patologia , Meduloblastoma/terapia , Análise Multivariada , Tumores Neuroectodérmicos Primitivos/patologia , Tumores Neuroectodérmicos Primitivos/terapia , Oligonucleotídeos Antissenso/genética , RNA Interferente Pequeno/genética , Fatores de Tempo , Transfecção
12.
Clin Cancer Res ; 11(7): 2747-55, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15814657

RESUMO

PURPOSE: Primary brain tumors are the leading cause of cancer death in children. Our purpose is (a) to assess the contribution of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) to the resistance of pediatric brain tumor cell lines to clinical alkylating agents and (b) to evaluate variables for maximal potentiation of cell killing by the MGMT inhibitor O6-benzylguanine, currently in clinical trials. Few such data for pediatric glioma lines, particularly those from low-grade tumors, are currently available. EXPERIMENTAL DESIGN: We used clonogenic assays of proliferative survival to quantitate cytoxicity of the chloroethylating agent 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and the methylating agent temozolomide in 11 glioma and five medulloblastoma lines. Twelve lines are newly established and characterized here, nine of them from low-grade gliomas including pilocytic astrocytomas. RESULTS: (a) MGMT is a major determinant of BCNU resistance and the predominant determinant of temozolomide resistance in both our glioma and medulloblastoma lines. On average, O(6)-benzylguanine reduced LD10 for BCNU and temozolomide, 2.6- and 26-fold, respectively, in 15 MGMT-expressing lines. (b) O6-Benzylguanine reduced DT (the threshold dose for killing) for BCNU and temozolomide, 3.3- and 138-fold, respectively. DT was decreased from levels higher than, to levels below, clinically achievable plasma doses for both alkylators. (c) Maximal potentiation by O6-benzylguanine required complete and prolonged suppression of MGMT. CONCLUSIONS: Our results support the use of O6-benzylguanine to achieve full benefit of alkylating agents, particularly temozolomide, in the chemotherapy of pediatric brain tumors.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Guanina/análogos & derivados , Guanina/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Adolescente , Encéfalo/enzimologia , Encéfalo/patologia , Carmustina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Criança , Pré-Escolar , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Masculino , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Temozolomida , Fatores de Tempo , Células Tumorais Cultivadas
13.
Clin Cancer Res ; 10(23): 7875-83, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15585620

RESUMO

PURPOSE: Apurinic/apyrimidinic endonuclease (Ap endo) is a key DNA repair enzyme that cleaves DNA at cytotoxic abasic sites caused by alkylating agents and radiation. We have observed that human glioma cells deficient in Ap endo activity are hypersensitive to clinically used alkylators (Silber et al., Clin Cancer Res 2002;8:3008.). Here we examine the association of glioma Ap endo activity with clinical response after alkylating agent-based chemotherapy or after radiotherapy. EXPERIMENTAL DESIGN: Cox proportional hazards regression models were used to analyze the relationship of Ap endo activity with time to tumor progression (TTP). RESULTS: In a univariate model with Ap endo activity entered as a continuous variable, the hazard ratio (HR) for progression after alkylator therapy in 30 grade III gliomas increased by a factor of 1.061 for every 0.01 increase in activity (P = 0.013). Adjusting for age, gender, extent of resection, and prior treatment strengthened slightly the association (HR = 1.094; P = 0.003). Similarly, the HR for progression after radiotherapy in 44 grade II and III tumors increased by a factor of 1.069 (P = 0.008). Adjusting for the aforementioned variables had little effect on the association. In contrast, we observed no association between activity and TTP in grade IV gliomas after either alkylator therapy in 34 tumors or radiotherapy in 26 tumors. CONCLUSIONS: Our data suggest that Ap endo activity mediates resistance to alkylating agents and radiation and may be a useful predictor of progression after adjuvant therapy in a subset of gliomas.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Astrocitoma , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Oligodendroglioma , Adulto , Astrocitoma/tratamento farmacológico , Astrocitoma/enzimologia , Astrocitoma/radioterapia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/radioterapia , Progressão da Doença , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Masculino , Estadiamento de Neoplasias , Oligodendroglioma/tratamento farmacológico , Oligodendroglioma/enzimologia , Oligodendroglioma/radioterapia , Tolerância a Radiação , Dosagem Radioterapêutica , Taxa de Sobrevida , Fatores de Tempo
14.
DNA Repair (Amst) ; 3(6): 629-38, 2004 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15135730

RESUMO

The Werner syndrome (WS) protein (WRN), a DNA helicase/exonuclease, is required for genomic stability and avoidance of cancer. Current evidence suggests that WRN is involved in the resolution of stalled and/or collapsed replication forks. This function is indicated, in part, by replication defects in WS cells and by hypersensitivity to agents causing major structural aberrations in DNA that block replication. We show here that antisense suppression of WRN in two human glioma cell lines reproduces hallmarks of the drug cytotoxicity profile of WS cells, namely, hypersensitivity to 4-nitroquinoline 1-oxide, camptothecin and hydroxyurea. We also show that antisense-treated cells are hypersensitive to methyl-lexitropsin, a site-specific alkylating agent that produces mainly N3-methyladenine, a cytotoxic and replication-blocking lesion. Antisense-treated cells are hypersensitive to O(6)-methylguanine adducts as well, but only when repair by O(6)-methylguanine-DNA methyltransferase is lacking. Our results illustrate the drug sensitivity caused by deficiency of WRN in a uniform genetic background. They extend the WRN DNA damage sensitivity spectrum to methyl base adducts that can result in blocked replication, and suggest that WRN may be required for resumption of processive replication when incomplete repair of DNA damage leaves blocking lesions at forks. The evidence that highly disparate lesions fall within the purview of WRN, and that abrogating DNA repair can reveal dependence on WRN, suggests that WRN may protect the genome from the lethal, mutagenic and carcinogenic effects of widely diverse DNA damage arising from endogenous processes and environmental agents.


Assuntos
Apoptose/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , DNA Helicases/fisiologia , Replicação do DNA/efeitos dos fármacos , Resistência a Medicamentos , Glioma/patologia , Guanina/análogos & derivados , Netropsina/análogos & derivados , 4-Nitroquinolina-1-Óxido/efeitos adversos , Adenina/análogos & derivados , Adenina/metabolismo , Alquilantes/efeitos adversos , Antineoplásicos/efeitos adversos , Camptotecina/efeitos adversos , Carcinógenos/efeitos adversos , Células Cultivadas , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , Exodesoxirribonucleases , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Glioma/metabolismo , Guanina/efeitos adversos , Humanos , Hidroxiureia/efeitos adversos , Netropsina/efeitos adversos , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Oligonucleotídeos Antissenso/farmacologia , RecQ Helicases , Síndrome de Werner , Helicase da Síndrome de Werner
15.
Clin Cancer Res ; 8(9): 3008-18, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12231548

RESUMO

Alkylating agents are standard components of adjuvant chemotherapy for gliomas. We provide evidence here that Ape1/Ref-1, the major mammalian apurinic/apyrimidinic endonuclease (Ap endo), contributes to alkylating agent resistance in human glioma cells by incising DNA at abasic sites. We show that antisense oligonucleotides directed against Ape1/Ref-1 in SNB19, a human glioma cell line lacking O(6)-methylguanine-DNA-methyltransferase, mediate both reduction in Ape1/Ref-1 protein and Ap endo activity and concurrent reduction in resistance to methyl methanesulfonate and the clinical alkylators temozolomide and 1,3-(2-chloroethyl)-1-nitrosourea. An accompanying increase in the level of abasic sites indicates that the DNA repair activity of Ape1/Ref-1 contributes to resistance. Conversely, we also show that exposure of SNB19 cells to HOCl, a generator of reactive oxygen species (ROS), results in elevated Ape1/Ref-1 protein and Ap endo activity, enhanced alkylator resistance, and reduced levels of abasic sites. Given current evidence that heightened oxidative stress prevails within brain tumors, the finding that ROS increase resistance to clinical alkylators in glioma cells may have significance for the response of gliomas to alkylating agent-based chemotherapy. Our results may also be relevant to the design of therapeutic regimens using concurrent ionizing radiation (a generator of ROS) and alkylating agent-based chemotherapy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/enzimologia , Carbono-Oxigênio Liases/fisiologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/fisiologia , Glioblastoma/enzimologia , Proteínas de Neoplasias/fisiologia , Ácido Apurínico/análise , Neoplasias Encefálicas/patologia , Carbono-Oxigênio Liases/biossíntese , Carmustina/farmacologia , Adutos de DNA , Dano ao DNA , DNA de Neoplasias/análise , DNA de Neoplasias/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Dacarbazina/farmacologia , Indução Enzimática , Glioblastoma/patologia , Humanos , Ácido Hipocloroso/farmacologia , Metanossulfonato de Metila/farmacologia , Proteínas de Neoplasias/deficiência , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Estresse Oxidativo , Espécies Reativas de Oxigênio/farmacologia , Temozolomida , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA