Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 8(6): 1589-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19509253

ABSTRACT

Vorinostat (suberoylanilide hydroxamic acid), a histone deacetylase inhibitor, is currently undergoing clinical evaluation as therapy for cancer. We investigated the effects of vorinostat on tumor cell radiosensitivity in a breast cancer brain metastasis model using MDA-MB-231-BR cells. In vitro radiosensitivity was evaluated using clonogenic assay. Cell cycle distribution and apoptosis was measured using flow cytometry. DNA damage and repair was evaluated using gammaH2AX. Mitotic catastrophe was measured by immunostaining. Growth delay and intracranial xenograft models were used to evaluate the in vivo tumor radiosensitivity. Cells exposed to vorinostat for 16 hours before and maintained in the medium after irradiation had an increase in radiosensitivity with a dose enhancement factor of 1.57. gammaH2AX, as an indicator of double-strand breaks, had significantly more foci per cell in the vorinostat plus irradiation group. Mitotic catastrophe, measured at 72 hours, was significantly increased in cells receiving vorinostat plus irradiation. Irradiation of s.c. MDA-MB-231-BR tumors in mice treated with vorinostat resulted in an increase in radiation-induced tumor growth delay. Most importantly, animals with intracranial tumor implants lived the longest after combination treatment. These results indicate that vorinostat enhances tumor cell radiosensitivity in vitro and in vivo. There was a greater than additive improvement in survival in our intracranial model. Combining vorinostat with radiation may be a potential treatment option for patients with breast cancer who develop brain metastases.


Subject(s)
Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Hydroxamic Acids/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Xenograft Model Antitumor Assays , Acetylation/drug effects , Acetylation/radiation effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Combined Modality Therapy , Enzyme Inhibitors/pharmacology , Female , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Immunoblotting , Kaplan-Meier Estimate , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Nude , Mitotic Index , Tumor Burden/drug effects , Tumor Burden/radiation effects , Vorinostat
2.
Clin Cancer Res ; 15(2): 607-12, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147766

ABSTRACT

PURPOSE: Poly (ADP-ribose) polymerase (PARP) inhibitors are undergoing clinical evaluation for cancer therapy. Because PARP inhibition has been shown to enhance tumor cell sensitivity to radiation, we investigated the in vitro and in vivo effects of the novel PARP inhibitor E7016. EXPERIMENTAL DESIGN: The effect of E7016 on the in vitro radiosensitivity of tumor cell lines was evaluated using clonogenic survival. DNA damage and repair were measured using gammaH2AX foci and neutral comet assay. Mitotic catastrophe was determined by immunostaining. Tumor growth delay was evaluated in mice for the effect of E7016 on in vivo (U251) tumor radiosensitivity. RESULTS: Cell lines exposed to E7016 preirradiation yielded an increase in radiosensitivity with dose enhancement factors at a surviving fraction of 0.1 from 1.4 to 1.7. To assess DNA double-strand breaks repair, gammaH2AX measured at 24 hours postirradiation had significantly more foci per cell in the E7016/irradiation group versus irradiation alone. Neutral comet assay further suggested unrepaired double-strand breaks with significantly greater DNA damage at 6 hours postirradiation in the combination group versus irradiation alone. Mitotic catastrophe staining revealed a significantly greater number of cells staining positive at 24 hours postirradiation in the combination group. In vivo, mice treated with E7016/irradiation/temozolomide had an additional growth delay of six days compared with the combination of temozolomide and irradiation. CONCLUSIONS: These results indicate that E7016 can enhance tumor cell radiosensitivity in vitro and in vivo through the inhibition of DNA repair. Moreover, enhanced growth delay with the addition of E7016 to temozolomide and radiotherapy in a glioma mouse model suggests a potential role for this drug in the treatment of glioblastoma multiforme.


Subject(s)
Brain Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apoptosis , Cell Line, Tumor , Comet Assay , DNA Repair , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Humans , In Vitro Techniques , Mice , Mitosis , Poly(ADP-ribose) Polymerases/metabolism , Radiotherapy/methods , Temozolomide
3.
Clin Cancer Res ; 14(17): 5410-5, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765532

ABSTRACT

PURPOSE: Preclinical studies evaluating histone deacetylase (HDAC) inhibitor-induced radiosensitization have largely focused on the preirradiation setting based on the assumption that enhanced radiosensitivity was mediated by changes in gene expression. Our previous investigations identified maximal radiosensitization when cells were exposed to HDAC inhibitors in both the preradiation and postradiation setting. We now expand on these studies to determine whether postirradiation exposure alone affects radiosensitivity. EXPERIMENTAL DESIGN: The effects of the HDAC inhibitor valproic acid (VA) on postirradiation sensitivity in human glioma cell lines were evaluated using a clonogenic assay, exposing cells to VA up to 24 h after irradiation. DNA damage repair was evaluated using gammaH2AX and 53BP1 foci and cell cycle phase distribution was analyzed by flow cytometry. Western blot of acetylated gammaH2AX was done following histone extraction on AUT gels. RESULTS: VA enhanced radiosensitivity when delivered up to 24 h after irradiation. Cells accumulated in G(2)-M following irradiation, although they returned to baseline at 24 h, mitigating the role of cell cycle redistribution in postirradiation sensitization by VA. At 12 h after irradiation, significant gammaH2AX and 53BP1 foci dispersal was shown in the control, although cells exposed to VA after irradiation maintained foci expression. VA alone had no effect on the acetylation or phosphorylation of H2AX, although it did acetylate radiation-induced gammaH2AX. CONCLUSIONS: These results indicate that VA enhances radiosensitivity at times up to 24 h after irradiation, which has direct clinical application.


Subject(s)
Enzyme Inhibitors/pharmacology , Glioma , Histone Deacetylase Inhibitors , Radiation-Sensitizing Agents/pharmacology , Valproic Acid/pharmacology , Brain Neoplasms , Cell Line, Tumor , Humans , Valproic Acid/administration & dosage
4.
Clin Cancer Res ; 14(3): 931-8, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18245557

ABSTRACT

PURPOSE: Temozolomide, a DNA methylating agent, is currently undergoing clinical evaluation for cancer therapy. Because temozolomide has been shown to increase survival rates of patients with malignant gliomas when given combined with radiation, and there is conflicting preclinical data concerning the radiosensitizing effects of temozolomide, we further investigated the possible temozolomide-induced enhancement of radiosensitivity. EXPERIMENTAL DESIGN: The effects of temozolomide on the in vitro radiosensitivity of U251 (a human glioma) and MDA-MB231BR (a brain-seeking variant of a human breast tumor) cell lines was evaluated using clonogenic assay. DNA damage and repair were evaluated using phosphorylated histone H2AX (gammaH2AX), and mitotic catastrophe was measured using nuclear fragmentation. Growth delay was used to evaluate the effects of temozolomide on in vivo (U251) tumor radiosensitivity. RESULTS: Exposure of each cell line to temozolomide for 1 h before irradiation resulted in an increase in radiosensitivity with dose enhancement factors at a surviving fraction of 0.1 ranging from 1.30 to 1.32. Temozolomide had no effect on radiation-induced apoptosis or on the activation of the G(2) cell cycle checkpoint. As a measure of DNA double strand breaks, gammaH2AX foci were determined as a function of time after the temozolomide + irradiation combination. The number of gammaH2AX foci per cell was significantly greater at 24 h after the combined modality compared with the individual treatments. Mitotic catastrophe, measured at 72 h, was also significantly increased in cells receiving the temozolomide + irradiation combination compared with the single treatments. In vivo studies revealed that temozolomide administration to mice bearing U251 tumor xenografts resulted in a greater than additive increase in radiation-induced tumor growth delay with a dose enhancement factor of 2.8. CONCLUSIONS: These results indicate that temozolomide can enhance tumor cell radiosensitivity in vitro and in vivo and suggest that this effect involves an inhibition of DNA repair leading to an increase in mitotic catastrophe.


Subject(s)
Dacarbazine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Brain Neoplasms , Cell Cycle/drug effects , Cell Line, Tumor , DNA Methylation , DNA Replication/drug effects , Dacarbazine/pharmacology , Glioblastoma , Humans , Mitosis/drug effects , Temozolomide , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...