Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Nucl Med ; 57(8): 1276-81, 2016 08.
Article in English | MEDLINE | ID: mdl-27056620

ABSTRACT

UNLABELLED: Recently, companion diagnostics with nuclear medicine techniques have been anticipated as more suitable means than biopsy for predicting treatment efficacy. The anticancer effect of capecitabine, an orally administered chemotherapeutic agent activated by thymidine phosphorylase (TP), is positively associated with tumor TP expression levels. This study aimed to assess whether TP imaging using a radiolabeled uracil derivative, (123)I-5-iodo-6-[(2-iminoimidazolidinyl)methyl]uracil ((123)I-IIMU), could predict the efficacy of capecitabine treatment. METHODS: Sensitivity to doxifluridine, a metabolite of capecitabine and direct substrate for TP, was assessed by water-soluble tetrazolium salt assays in vitro for 3 human colon cancer cell lines with different TP expression profiles. The intracellular uptake and retention of (123)I-IIMU were evaluated. Mice inoculated with each cell line were treated with capecitabine for 2 wk, and tumor growth was compared. In vivo distribution studies and SPECT/CT imaging of (123)I-IIMU were performed in inoculated mice. RESULTS: In vitro experiments showed a positive relation between TP expression levels and doxifluridine sensitivity. In vitro studies revealed that intracellular uptake and retention of (123)I-IIMU were dependent on TP expression levels. In vivo experiments in inoculated mice showed that (123)I-IIMU accumulation in tumor tissue was in line with TP expression levels and susceptibility to capecitabine treatment. Moreover, SPECT/CT imaging of (123)I-IIMU in tumor-inoculated mice showed that (123)I-IIMU reflects TP expression levels in tumor tissues. CONCLUSION: (123)I-IIMU could be used as an in vivo companion diagnostic for predicting the efficacy of capecitabine treatment.


Subject(s)
5'-Nucleotidase/metabolism , Capecitabine/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Drug Monitoring/methods , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Capecitabine/pharmacokinetics , Cell Line, Tumor , Colonic Neoplasms/diagnostic imaging , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging/methods , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
2.
Cancer Lett ; 347(1): 151-8, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24530512

ABSTRACT

High atomic number molecules, such as gold and platinum, are known to enhance the biological effect of X-irradiation. This study was aimed to determine the radiosensitizing potential of PEGylated nanogel containing gold nanoparticles (GNG) and the cellular mechanism involved. GNG pretreatment increased the levels of reproductive cell death and apoptosis induced by X-irradiation. GNG accumulated in cytoplasm and increased the expression of endoplasmic reticulum (ER) stress-related protein. GNG suppressed the repair capacity of DNA after X-irradiation by down-regulating DNA repair-related proteins. Our results suggest that GNG radiosensitized cells by enhancing apoptosis and impairing DNA repair capacity via ER stress induction.


Subject(s)
Endoplasmic Reticulum/metabolism , Gels , Gold/chemistry , Metal Nanoparticles , Polyethylene Glycols/chemistry , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Cricetinae , Cricetulus , Mice , X-Rays
3.
FEBS Lett ; 587(20): 3348-53, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24021650

ABSTRACT

In this study, we provide evidence that endoplasmic reticulum (ER) stress suppresses DNA double-strand break (DSB) repair and increases radiosensitivity of tumor cells by altering Rad51 levels. We show that the ER stress inducer tunicamycin stimulates selective degradation of Rad51 via the 26S proteasome, impairing DSB repair and enhancing radiosensitivity in human lung cancer A549 cells. We also found that glucose deprivation, which is a physiological inducer of ER stress, triggered similar events. These findings suggest that ER stress caused by the intratumoral environment influences tumor radiosensitivity, and that it has potential as a novel target to improve cancer radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Repair/radiation effects , Proteasome Endopeptidase Complex/metabolism , Rad51 Recombinase/metabolism , Radiation, Ionizing , Blotting, Western , Cell Line, Tumor , DNA Repair/genetics , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/radiation effects , Humans , Rad51 Recombinase/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
BMC Cancer ; 13: 106, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23496909

ABSTRACT

BACKGROUND: Glioblastoma is one of the intractable cancers and is highly resistant to ionizing radiation. This radioresistance is partly due to the presence of a hypoxic region which is widely found in advanced malignant gliomas. In the present study, we evaluated the effectiveness of the hypoxic cell sensitizer doranidazole (PR-350) using the C6 rat glioblastoma model, focusing on the status of blood brain barrier (BBB). METHODS: Reproductive cell death in the rat C6 glioma cell line was determined by means of clonogenic assay. An intracranial C6 glioma model was established for the in vivo experiments. To investigate the status of the BBB in C6 glioma bearing brain, we performed the Evans blue extravasation test. Autoradiography with [(14)C]-doranidazole was performed to examine the distribution of doranidazole in the glioma tumor. T2-weighted MRI was employed to examine the effects of X-irradiation and/or doranidazole on tumor growth. RESULTS: Doranidazole significantly enhanced radiation-induced reproductive cell death in vitro under hypoxia, but not under normoxia. The BBB in C6-bearing brain was completely disrupted and [(14)C]-doranidazole specifically penetrated the tumor regions. Combined treatment with X-irradiation and doranidazole significantly inhibited the growth of C6 gliomas. CONCLUSIONS: Our results revealed that BBB disruption in glioma enables BBB-impermeable radiosensitizers to penetrate and distribute in the target region. This study is the first to propose that in malignant glioma the administration of hydrophilic hypoxic radiosensitizers could be a potent strategy for improving the clinical outcome of radiotherapy without side effects.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Imidazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Disease Models, Animal , Glioblastoma/pathology , Prospective Studies , Rats
5.
BMC Cancer ; 12: 469, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23057787

ABSTRACT

BACKGROUND: Anti-cancer drugs are widely used in cancer treatment frequently combined with surgical therapy and/or radiation therapy. Although surgery and radiation have been suggested to facilitate invasion and metastasis of tumor cells in some cases, there is so far little information about the effect of anti-cancer drugs on cellular invasive ability and metastasis. In this study, using four different anti-cancer drugs (vincristine, paclitaxel, cisplatin and etoposide), we examined whether these drugs influence the invasive ability of tumor cells. METHODS: Human gastric adenocarcinoma MKN45 cells were used to evaluate the effect of anti-cancer drugs. After drug treatment, cellular invasive ability was assessed using the Matrigel invasion chamber. Cytoskeletal changes after treatment were examined microscopically with F-actin staining. In addition, we monitored cellular motility in 3D matrigel environment by time-lapse microscopic analysis. The drug-induced activation of RhoA and ROCK was evaluated by pull-down assay and Western blotting using an antibody against phosphorylated myosin light chain (MLC), respectively. Where necessary, a ROCK inhibitor Y27632 and siRNA for guanine nucleotide exchange factor-H1 (GEF-H1) were applied. RESULTS: Among all drugs tested, only vincristine stimulated the invasive ability of MKN45 cells. Microscopic analysis revealed that vincristine induced the formation of non-apoptotic membrane blebs and amoeboid-like motility. Vincristine significantly enhanced RhoA activity and MLC phosphorylation, suggesting the involvement of RhoA/ROCK pathway in the vincristine-induced cytoskeletal reorganization and cellular invasion. Furthermore, we found that Y27632 as well as the siRNA for GEF-H1, a RhoA-specific activator, attenuated MLC phosphorylation, the formation of membrane blebs and the invasive ability after vincristine treatment. CONCLUSIONS: These results indicate that vincristine activates GEF-H1/RhoA/ROCK/MLC signaling, thereby promoting amoeboid-like motility and the invasive ability of MKN45 cells.


Subject(s)
Adenocarcinoma/pathology , Cell Movement/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Myosin Light Chains/metabolism , Stomach Neoplasms/pathology , Vincristine/pharmacology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cisplatin/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Etoposide/pharmacology , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Paclitaxel/pharmacology , Phosphorylation/drug effects , Rho Guanine Nucleotide Exchange Factors , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism
6.
J Radiat Res ; 52(4): 456-63, 2011.
Article in English | MEDLINE | ID: mdl-21785234

ABSTRACT

The combination of a chemotherapeutic agent and radiation is widely applied to enhance cell death in solid tumor cells in cancer treatment. The purine analogue 8-aminoadenosine (8-NH(2)-Ado) is known to be a transcription inhibitor that has proved very effective in multiple myeloma cell lines and primary indolent leukemia cells. In this report, to examine whether 8-NH(2)-Ado had the ability to enhance the radiation-induced cell killing in solid tumor cells, human lung adenocarcinoma A549 cells were irradiated in the presence and absence of 8-NH(2)-Ado. 8-NH(2)-Ado significantly increased reproductive cell death and apoptosis in A549 cells exposed to X-rays. When peptide inhibitors against caspase-3, -8, and -9 were utilized to evaluate the involvement of caspases, all inhibitors suppressed the enhancement of radiation-induced apoptosis, suggesting that not only mitochondria-mediated apoptotic signal transduction pathways but also death receptor-mediated pathways were involved in this enhancement of apoptosis. In addition, in the cells exposed to the treatment combining X-irradiation and 8-NH(2)-Ado, reduction of the intracellular ATP concentration was essential for survival, and down-regulation of the expression of antiapoptotic proteins such as survivin and XIAP was observed. These results indicate that 8-NH(2)-Ado has potential not only as an anti-tumor drug for leukemia and lymphoma but also as a radiosensitizing agent for solid tumors.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Adenosine/analogs & derivatives , Multiple Myeloma/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Adenosine/pharmacology , Adenosine Triphosphate/chemistry , Apoptosis , Caspase Inhibitors , Cell Death , Cell Line, Tumor , Dose-Response Relationship, Radiation , Humans , Microscopy, Fluorescence/methods , Multiple Myeloma/drug therapy , Peptides/antagonists & inhibitors , Propidium/pharmacology , Signal Transduction , X-Rays
7.
Mol Cancer ; 10: 92, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798026

ABSTRACT

BACKGROUND: A novel anticancer drug 1-(3-C-ethynyl-ß-D-ribo-pentofuranosyl)cytosine (ECyd, TAS106) has been shown to radiosensitize tumor cells and to improve the therapeutic efficiency of X-irradiation. However, the effect of TAS106 on cellular DNA repair capacity has not been elucidated. Our aim in this study was to examine whether TAS106 modified the repair capacity of DNA double-strand breaks (DSBs) in tumor cells. METHODS: Various cultured cell lines treated with TAS106 were irradiated and then survival fraction was examined by the clonogenic survival assays. Repair of sublethal damage (SLD), which indicates DSBs repair capacity, was measured as an increase of surviving cells after split dose irradiation with an interval of incubation. To assess the effect of TAS106 on the DSBs repair activity, the time courses of γ-H2AX and 53BP1 foci formation were examined by using immunocytochemistry. The expression of DNA-repair-related proteins was also examined by Western blot analysis and semi-quantitative RT-PCR analysis. RESULTS: In clonogenic survival assays, pretreatment of TAS106 showed radiosensitizing effects in various cell lines. TAS106 inhibited SLD repair and delayed the disappearance of γ-H2AX and 53BP1 foci, suggesting that DSB repair occurred in A549 cells. Western blot analysis demonstrated that TAS106 down-regulated the expression of BRCA2 and Rad51, which are known as keys among DNA repair proteins in the homologous recombination (HR) pathway. Although a significant radiosensitizing effect of TAS106 was observed in the parental V79 cells, pretreatment with TAS106 did not induce any radiosensitizing effects in BRCA2-deficient V-C8 cells. CONCLUSIONS: Our results indicate that TAS106 induces the down-regulation of BRCA2 and the subsequent abrogation of the HR pathway, leading to a radiosensitizing effect. Therefore, this study suggests that inhibition of the HR pathway may be useful to improve the therapeutic efficiency of radiotherapy for solid tumors.


Subject(s)
BRCA2 Protein/genetics , Cytidine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , BRCA2 Protein/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Cytidine/pharmacology , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination/drug effects , Homologous Recombination/genetics , Homologous Recombination/radiation effects , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/radiotherapy , Nucleosides/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics
8.
Cancer Lett ; 277(1): 64-71, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19117669

ABSTRACT

Mitochondria in mammalian cells are well-known to play an important role in the intrinsic pathway of genotoxic-agent-induced apoptosis by releasing cytochrome c into cytosol and to be a major source of reactive oxygen species (ROS). The aim of this study was to examine whether mitochondrial ROS are involved in radiation-induced apoptotic signaling in A549 cells. Post-irradiation treatment with N-acetyl-L-cysteine (NAC) inhibited cytochrome c release from mitochondria but did not affect expression levels of Bcl-2, Bcl-X(L) and Bax, suggesting that late production of ROS triggered cytochrome c release. Experiments using DCFDA (a classical ROS fluorescence probe) and MitoAR (a novel mitochondrial ROS probe) demonstrated that intracellular and mitochondrial ROS were enhanced 6h after X irradiation. Furthermore, the O(2)(-*) production ability of mitochondria isolated from A549 cells was evaluated by ESR spectroscopy combined with a spin-trapping reagent (CYPMPO). When isolated mitochondria were incubated with NADH, succinate and CYPMPO, an ESR spectrum due to CYPMPO-OOH was detected. This NADH/succinate-dependent O(2)(-*) production from mitochondria of irradiated cells was significantly increased in comparison with that of unirradiated cells. These results indicate that ionizing radiation enhances O(2)(-*) production from mitochondria to trigger cytochrome c release in A549 cells.


Subject(s)
Cytochromes c/metabolism , Lung Neoplasms/radiotherapy , Mitochondria/radiation effects , Acetylcysteine/pharmacology , Apoptosis/radiation effects , Caspases/metabolism , Cell Line, Tumor , Electron Spin Resonance Spectroscopy , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/enzymology , Oxidation-Reduction , Proto-Oncogene Proteins c-bcl-2/analysis , Reactive Oxygen Species/metabolism , Superoxides/metabolism , bcl-2-Associated X Protein/analysis , bcl-X Protein/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...