Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
BBA Clin ; 3: 1-10, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25558448

ABSTRACT

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.

2.
Front Oncol ; 2: 176, 2012.
Article in English | MEDLINE | ID: mdl-23230562

ABSTRACT

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.

3.
Mol Cancer Ther ; 9(5): 1208-18, 2010 May.
Article in English | MEDLINE | ID: mdl-20457618

ABSTRACT

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.


Subject(s)
Brain Neoplasms/radiotherapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Glioblastoma/radiotherapy , Radiation Tolerance/drug effects , Tumor Suppressor Proteins/genetics , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , DNA Mismatch Repair/drug effects , DNA Mismatch Repair/genetics , DNA Mismatch Repair/radiation effects , DNA Modification Methylases/metabolism , DNA Modification Methylases/physiology , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/physiology , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gamma Rays , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Maximum Tolerated Dose , Temozolomide , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
4.
Clin Cancer Res ; 13(2 Pt 1): 612-20, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17255284

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioma/pathology , Netropsin/analogs & derivatives , Cell Line, Tumor , DNA Adducts/metabolism , DNA Mismatch Repair , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , Netropsin/pharmacology , Oligonucleotides, Antisense/chemistry , Time Factors , Tumor Cells, Cultured
5.
Clin Cancer Res ; 10(23): 7875-83, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585620

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Astrocytoma , Biomarkers, Tumor/metabolism , Brain Neoplasms , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Oligodendroglioma , Adult , Astrocytoma/drug therapy , Astrocytoma/enzymology , Astrocytoma/radiotherapy , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/radiotherapy , Disease Progression , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Male , Neoplasm Staging , Oligodendroglioma/drug therapy , Oligodendroglioma/enzymology , Oligodendroglioma/radiotherapy , Radiation Tolerance , Radiotherapy Dosage , Survival Rate , Time Factors
6.
Clin Cancer Res ; 8(9): 3008-18, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231548

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/enzymology , Carbon-Oxygen Lyases/physiology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/physiology , Glioblastoma/enzymology , Neoplasm Proteins/physiology , Apurinic Acid/analysis , Brain Neoplasms/pathology , Carbon-Oxygen Lyases/biosynthesis , Carmustine/pharmacology , DNA Adducts , DNA Damage , DNA, Neoplasm/analysis , DNA, Neoplasm/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Dacarbazine/pharmacology , Enzyme Induction , Glioblastoma/pathology , Humans , Hypochlorous Acid/pharmacology , Methyl Methanesulfonate/pharmacology , Neoplasm Proteins/deficiency , O(6)-Methylguanine-DNA Methyltransferase/deficiency , Oxidative Stress , Reactive Oxygen Species/pharmacology , Temozolomide , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...