Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Curr Pain Headache Rep ; 22(9): 60, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29987680

ABSTRACT

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.


Subject(s)
Pain Management , Pain/diagnostic imaging , Peripheral Nervous System/surgery , Ultrasonography , Animals , Brain/surgery , Humans , Treatment Outcome , Ultrasonography/methods
2.
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.

3.
Chem Res Toxicol ; 26(1): 156-68, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23234400

ABSTRACT

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.


Subject(s)
Alkylating Agents/chemical synthesis , DNA/chemistry , Adenine/analogs & derivatives , Adenine/chemistry , Alkylating Agents/chemistry , Alkylating Agents/toxicity , Animals , Cattle , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Glycosylases/chemistry , DNA Glycosylases/metabolism , DNA Methylation , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Peptides/chemistry , Peptides/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thermodynamics
4.
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.

5.
Biochim Biophys Acta ; 1826(1): 71-82, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22244911

ABSTRACT

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.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Glioma/drug therapy , Glioma/enzymology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Brain Neoplasms/genetics , Glioma/genetics , Humans , Molecular Targeted Therapy , O(6)-Methylguanine-DNA Methyltransferase/genetics
6.
Int J Cancer ; 129(10): 2370-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21207372

ABSTRACT

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.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/radiotherapy , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Ependymoma/enzymology , Ependymoma/radiotherapy , Adolescent , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Child , Disease-Free Survival , Ependymoma/immunology , Ependymoma/mortality , Female , Humans , Infant , Male , Radiation Tolerance
7.
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
8.
Childs Nerv Syst ; 25(1): 39-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18946672

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Vinblastine/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Child , Drug Resistance, Neoplasm/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
DNA Repair (Amst) ; 6(8): 1127-33, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17500046

ABSTRACT

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

Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/etiology , Brain/embryology , Brain/enzymology , DNA Modification Methylases/deficiency , DNA Repair Enzymes/deficiency , O(6)-Methylguanine-DNA Methyltransferase/deficiency , Tumor Suppressor Proteins/deficiency , Adult , Brain/growth & development , Brain Neoplasms/genetics , Child , DNA Repair , Fetus/enzymology , Gestational Age , Humans , Mutation , Phenotype
10.
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
11.
Clin Cancer Res ; 11(20): 7405-14, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16243814

ABSTRACT

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.


Subject(s)
Brain Neoplasms/enzymology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Medulloblastoma/enzymology , Neuroectodermal Tumors, Primitive/enzymology , Adolescent , Adult , Antineoplastic Agents, Alkylating/pharmacology , Blotting, Western , Brain/drug effects , Brain/enzymology , Brain/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Carmustine/pharmacology , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Survival/drug effects , Cell Survival/genetics , Child , Child, Preschool , Combined Modality Therapy , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Progression , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Medulloblastoma/pathology , Medulloblastoma/therapy , Multivariate Analysis , Neuroectodermal Tumors, Primitive/pathology , Neuroectodermal Tumors, Primitive/therapy , Oligonucleotides, Antisense/genetics , RNA, Small Interfering/genetics , Time Factors , Transfection
12.
Clin Cancer Res ; 11(7): 2747-55, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15814657

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Guanine/analogs & derivatives , Guanine/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Adolescent , Brain/enzymology , Brain/pathology , Carmustine/pharmacology , Cell Survival/drug effects , Child , Child, Preschool , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Female , Humans , Male , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Temozolomide , Time Factors , Tumor Cells, Cultured
13.
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
14.
DNA Repair (Amst) ; 3(6): 629-38, 2004 Jun 03.
Article in English | MEDLINE | ID: mdl-15135730

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , DNA Helicases/physiology , DNA Replication/drug effects , Drug Resistance , Glioma/pathology , Guanine/analogs & derivatives , Netropsin/analogs & derivatives , 4-Nitroquinoline-1-oxide/adverse effects , Adenine/analogs & derivatives , Adenine/metabolism , Alkylating Agents/adverse effects , Antineoplastic Agents/adverse effects , Camptothecin/adverse effects , Carcinogens/adverse effects , Cells, Cultured , DNA Helicases/antagonists & inhibitors , DNA Helicases/genetics , Exodeoxyribonucleases , Fibroblasts/cytology , Fibroblasts/drug effects , Glioma/metabolism , Guanine/adverse effects , Humans , Hydroxyurea/adverse effects , Netropsin/adverse effects , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oligonucleotides, Antisense/pharmacology , RecQ Helicases , Werner Syndrome , Werner Syndrome Helicase
15.
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
...