Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Appl Immunohistochem Mol Morphol ; 25(7): 497-504, 2017 08.
Article in English | MEDLINE | ID: mdl-27153440

ABSTRACT

The O-methylguanine-DNA-methyltranferase (MGMT) status is a powerful predictor of response to temozolomide for newly diagnosed glioblastoma (GBM) patients, and it is commonly assessed by immunohistochemistry (IHC), methylation-specific polymerase chain reaction (MSP), quantitative real-time MSP (qMSP), and/or pyrosequencing (PSQ). This study was to compare their predictive power of prognosis in 121 newly diagnosed GBM patients using multivariate Cox regression with bootstrapping. MGMT status tested by IHC, MSP, qMSP, or PSQ all showed significant correlation with the progression-free survival and overall survival of GBM patients. The predictive power of IHC for progression-free survival and overall survival was lower than those of the methylation assays, but their differences were not significant. Performing additional methylation assay, especially PSQ, could better predict the prognosis of patients with IHC- tumors. MGMT status tested by IHC, MSP, qMSP, or PSQ all showed prognostic significance. An additional MGMT methylation assay, of which PSQ appeared to be the best, could improve the predictive power for GBM patients with MGMT IHC- tumors.


Subject(s)
Brain Neoplasms , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma , Neoplasm Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Brain Neoplasms/enzymology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Disease-Free Survival , Female , Glioblastoma/enzymology , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Survival Rate
2.
J Neurooncol ; 122(1): 179-88, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25575938

ABSTRACT

Methylation-specific polymerase chain reaction (MSP) for the promoter methylation status of O(6)-methylguanine-DNA-methyltranferase (MGMT) gene theoretically provides a positive or negative result. However, the faint MSP product is difficult to interpret. The aim of this study was to evaluate the significance of faint MSP product in glioblastoma (GBM). Critical concentrations of methylated control DNA, i.e., 100, 1, 0.5 and 0 % were run parallel with 116 newly diagnosed GBMs in order to standardize the interpretation and to distinguish positive (+), equivocal (±), and negative (-; unmethylated) results. Cases with the faint MSP product and its intensity between those of 1 and 0.5 % DNA controls were considered equivocal (±). MGMT methylation quantifications were also determined by quantitative real-time MSP (qMSP) and pyrosequencing (PSQ), and protein expression was detected by immunohistochemistry. There were significant correlations between MSP and all the aforementioned studies. The concordance rates between the MSP+ and qMSP+ cases, as well as the MSP- and qMSP- cases were 100 %, and the MSP± cases comprised 76.5 % of qMSP+ cases and 23.5 % of qMSP- cases. PSQ study showed that heterogeneous methylation was more frequently encountered in the MSP± cases. Multivariate analyses disclosed that although the overall survival of the MSP± cases was indistinct from that of the MSP+ cases, its progression free survival was significantly worse and was indistinct from that of the MSP- cases. In conclusion, GBMs with faint MGMT MSP products should be distinguished from MSP+ cases as their behaviors were different.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/mortality , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Polymerase Chain Reaction/methods , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Child , Child, Preschool , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Female , Follow-Up Studies , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate , Tumor Suppressor Proteins/metabolism , Young Adult
3.
Am J Surg Pathol ; 37(2): 264-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23282970

ABSTRACT

We evaluated the predictive value of O6-methylguanine-DNA methyltransferase (MGMT) protein expression and MGMT promoter methylation status in glioblastomas (GBM) treated with temozolomide (TMZ) in a Taiwan medical center. Protein expression by immunohistochemical analysis (IHC) and MGMT promoter methylation detected by methylation-specific polymerase chain reaction (MSP) were performed in a series of 107 newly diagnosed GBMs. We used endothelial cells as an internal reference for IHC staining because the staining intensities of the MGMT-expressing cells in different specimens varied considerably; a positive result was defined as the staining intensity of the majority of tumor cells similar to that of the adjacent endothelial cells. Immunostainings for microglial/endothelial markers were included as part of the MGMT IHC evaluation, and in cases that were difficult to interpret, double-labeling helped to clarify the nature of reactive cells. The MGMT protein expression was reversely associated with MGMT promoter methylation status in 83.7% of cases (MSP/IHC and MSP/IHC; Pearson r=-0.644, P<0.001). Twenty-two of 24 (91.7%) IHC tumors did not respond to TMZ treatment. Combining MSP and IHC results, all the 15 MSP/IHC GBMs were TMZ resistant. The MGMT status detected by either IHC or MSP was significantly correlated with the TMZ treatment response (both P<0.001) and survival of GBM patients (both P<0.05).


Subject(s)
Brain Neoplasms/diagnosis , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Endothelium, Vascular/pathology , Glioblastoma/diagnosis , Histiocytes/pathology , Tumor Suppressor Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/therapeutic use , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Child , Child, Preschool , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Histiocytes/drug effects , Histiocytes/metabolism , Humans , Male , Middle Aged , Predictive Value of Tests , Promoter Regions, Genetic , Staining and Labeling/methods , Survival Rate , Taiwan/epidemiology , Temozolomide , Treatment Outcome , Tumor Suppressor Proteins/genetics , Young Adult
4.
J Cell Physiol ; 227(3): 976-93, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21503893

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor. Patients diagnosed with GBM have a poor prognosis, and it has been reported that tumor malignancy and GBM recurrence are promoted by STAT3 signaling. As resveratrol (RV), a polyphenol in grapes, is reported to be a potent and non-toxic cancer-preventive compound, the aim of this study was to investigate the therapeutic effect and molecular mechanisms of RV on GBM-derived radioresistant tumor initiating cells (TIC). Firstly, our results showed that primary GBM-CD133(+) TIC presented high tumorigenic and radiochemoresistant properties as well as increased protein levels of phosphorylated STAT3. We consistently observed that treatment with shRNA-STAT3 (sh-STAT3) or AG490, a STAT3 inhibitor, significantly inhibited the cancer stem-like cell properties and radioresistance of GBM-CD133(+) in vitro and in vivo. Furthermore, treatment of GBM-CD133(+) with 100 µM RV induced apoptosis and enhanced radiosensitivity by suppressing STAT3 signaling. Microarray results suggested that RV or AG490 inhibited the stemness gene signatures of GBM-CD133(+) and facilitated the differentiation of GBM-CD133(+) into GBM-CD133(-) or astrocytoma cells. Finally, xenotransplant experiments indicated that RV or sh-STAT3 therapy could significantly improve the survival rate and synergistically enhance the radiosensitivity of radiation-treated GBM-TIC. In summary, RV can reduce in vivo tumorigenicity and enhance the sensitivity of GBM-TIC to radiotherapies through the STAT3 pathway.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , STAT3 Transcription Factor/antagonists & inhibitors , Stilbenes/pharmacology , Aged , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Astrocytoma/drug therapy , Astrocytoma/pathology , Astrocytoma/radiotherapy , Brain Neoplasms/pathology , Chemoradiotherapy/methods , Female , Glioblastoma/pathology , Humans , Male , Mice , Mice, SCID , Middle Aged , Resveratrol , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Radiat Oncol ; 6: 7, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21244709

ABSTRACT

BACKGROUND: 1-{4-[Bis(2-chloroethyl)amino]phenyl}-3-[2-methyl-5-(4-methylacridin-9-ylamino)phenyl]urea (BO-1051) is an N-mustard DNA alkylating agent reported to exhibit antitumor activity. Here we further investigate the effects of this compound on radiation responses of human gliomas, which are notorious for the high resistance to radiotherapy. METHODS: The clonogenic assay was used to determine the IC50 and radiosensitivity of human glioma cell lines (U87MG, U251MG and GBM-3) following BO-1051. DNA histogram and propidium iodide-Annexin V staining were used to determine the cell cycle distribution and the apoptosis, respectively. DNA damage and repair state were determined by γ-H2AX foci, and mitotic catastrophe was measure using nuclear fragmentation. Xenograft tumors were measured with a caliper, and the survival rate was determined using Kaplan-Meier method. RESULTS: BO-1051 inhibited growth of human gliomas in a dose- and time-dependent manner. Using the dosage at IC50, BO-1051 significantly enhanced radiosensitivity to different extents [The sensitizer enhancement ratio was between 1.24 and 1.50 at 10% of survival fraction]. The radiosensitive G2/M population was raised by BO-1051, whereas apoptosis and mitotic catastrophe were not affected. γ-H2AX foci was greatly increased and sustained by combined BO-1051 and γ-rays, suggested that DNA damage or repair capacity was impaired during treatment. In vivo studies further demonstrated that BO-1051 enhanced the radiotherapeutic effects on GBM-3-beared xenograft tumors, by which the sensitizer enhancement ratio was 1.97. The survival rate of treated mice was also increased accordingly. CONCLUSIONS: These results indicate that BO-1051 can effectively enhance glioma cell radiosensitivity in vitro and in vivo. It suggests that BO-1051 is a potent radiosensitizer for treating human glioma cells.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Nitrogen Mustard Compounds/pharmacology , Radiation Tolerance/drug effects , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Damage/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Dose-Response Relationship, Drug , Female , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Models, Biological , Radiation-Sensitizing Agents/pharmacology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
6.
J Clin Neurosci ; 12(7): 775-80, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16198917

ABSTRACT

Positron emission tomography (PET) allows examination of a variety of physiological parameter, including blood flow, glucose, amino acid and oxygen metabolism. However, correlation of PET scan findings and the degree of malignancy of intracerebral tumors continues to be controversial. Nine patients with primarily diagnosed intraparenchymal brain tumors were included in this study. We performed 11C-methionine-PET (met-PET) prior to surgical treatment and the differential absorption ratio (DAR) was calculated. All patients underwent open or stereotactic surgery and specimens for pathological diagnosis were obtained. The biological activity of each tumor was determined by calculation of the proliferation index from MIB-1 immunohistochemistry. The DAR of met-PET for individual tumors correlated with the histological diagnosis and degree of malignancy and this was further confirmed by good correlation with the MIB-1 proliferation index. We conclude that met-PET may be a reliable and effective preoperative evaluation to determine the type and malignancy of intraparenchymal brain lesions.


Subject(s)
Brain Neoplasms/diagnostic imaging , Methionine/pharmacokinetics , Positron-Emission Tomography , Aged , Brain Neoplasms/classification , Brain Neoplasms/metabolism , Female , Humans , Immunohistochemistry/methods , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging/methods , Male , Middle Aged , Regression Analysis , Tomography, Emission-Computed/methods , Tritium
7.
J Am Chem Soc ; 126(34): 10640-4, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15327322

ABSTRACT

A chemoenzymatic approach has been developed to synthesize poly(ethylene glycol)-based amphiphilic copolymers under mild reaction conditions that self-assemble in aqueous media to form polymeric nanomicelles in the range of 20-50 nm. The supramolecular organization of polymeric nanomicelles was studied by 1H NMR longitudinal relaxation time (T1) and light scattering techniques (static and dynamic). Interestingly, the enzyme novozyme-435 plays an important role in controlling the polymerization and distribution of polymer chains, which is critical for the formation of nanomicelles with unimodal distributions. The methodology developed is highly flexible as it allows the introduction of various functionalities in the polymeric nanomicelles. These self-organized nanomicelles are highly efficient drug delivery vehicles for hydrophobic and partially hydrophilic drugs, both transdermally and orally, as they have the ability to encapsulate guest molecules during self-organization. In vivo studies by encapsulating anti-inflammatory agents (aspirin and naproxen) in these polymeric nanomicelles and by applying topically resulted in significant reduction in inflammation. The % reduction in inflammation using polymeric nanomicelles containing aspirin and naproxen was 62 and 64%, respectively.


Subject(s)
Aspirin/chemistry , Drug Delivery Systems/methods , Polyesters/chemical synthesis , Polyethylene Glycols/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Aspirin/administration & dosage , Chemistry, Pharmaceutical , Enzymes, Immobilized , Fungal Proteins , Hydrophobic and Hydrophilic Interactions , Inflammation/drug therapy , Lipase/chemistry , Mice , Micelles , Nanotechnology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...