Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...