Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Res ; 1230: 290-302, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18674521

ABSTRACT

The blood-brain tumor barrier (BTB) significantly limits delivery of therapeutic concentrations of chemotherapy to brain tumors. A novel approach to selectively increase drug delivery is pharmacologic modulation of signaling molecules that regulate BTB permeability, such as those in cGMP signaling. Here we show that oral administration of sildenafil (Viagra) and vardenafil (Levitra), inhibitors of cGMP-specific PDE5, selectively increased tumor capillary permeability in 9L gliosarcoma-bearing rats with no significant increase in normal brain capillaries. Tumor-bearing rats treated with the chemotherapy agent, adriamycin, in combination with vardenafil survived significantly longer than rats treated with adriamycin alone. The selective increase in tumor capillary permeability appears to be mediated by a selective increase in tumor cGMP levels and increased vesicular transport through tumor capillaries, and could be attenuated by iberiotoxin, a selective inhibitor for calcium-dependent potassium (K(Ca)) channels, that are effectors in cGMP signaling. The effect by sildenafil could be further increased by simultaneously using another BTB "opener", bradykinin. Collectively, this data demonstrates that oral administration of PDE5 inhibitors selectively increases BTB permeability and enhances anti-tumor efficacy for a chemotherapeutic agent. These findings have significant implications for improving delivery of anti-tumor agents to brain tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioma/drug therapy , Glioma/metabolism , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Animals , Autoradiography , Blood Pressure/drug effects , Brain Chemistry/drug effects , Brain Neoplasms/pathology , Capillaries/pathology , Capillary Permeability/drug effects , Cyclic GMP/blood , Cyclic GMP/metabolism , Female , Glioma/pathology , Imidazoles/pharmacology , Microscopy, Electron, Transmission , Neovascularization, Pathologic/pathology , Piperazines/pharmacology , Purines/pharmacology , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Sucrose/metabolism , Sulfones/pharmacology , Survival Analysis , Tight Junctions/drug effects , Tight Junctions/ultrastructure , Triazines/pharmacology , Vardenafil Dihydrochloride
2.
Brain Res ; 1227: 198-206, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18602898

ABSTRACT

The blood-brain tumor barrier (BTB) significantly limits delivery of effective concentrations of chemotherapeutic drugs to brain tumors. Previous studies suggest that BTB permeability may be modulated via alteration in the activity of potassium channels. In this study, we studied the relationship of BTB permeability increase mediated by potassium channel agonists to channel expression in two rat brain tumor models. Intravenous infusion of KCO912 (K(ATP) agonist), minoxidil sulfate (K(ATP) agonist) or NS1619 (K(Ca) agonist) increased tumor permeability more in the 9L allogeneic brain tumor model than in the syngeneic brain tumor model. Consistently, expression of both K(ATP) and K(Ca) channels in 9L tumors was increased to a significantly greater extent in Wistar rats (allogeneic) as compared to Fischer rats (syngeneic). Furthermore, as a preliminary effort to understand clinical implication of potassium channels in brain tumor treatment, we determined the expression of K(ATP) in surgical specimens. K(ATP) mRNA was detected in glioblastoma multiforme (GBM) from nineteen patients examined, with a wide range of expression levels. Interestingly, in paired GBM tissues from seven patients before and after vaccination therapy, increased levels of K(ATP) were detected in five patients after vaccination that had positive response to chemotherapy after vaccination. The present study indicates that the effects of potassium channel agonists on BTB permeability are different between syngeneic and allogeneic models which have different expression levels of potassium channels. The expression of potassium channels in brain tumors is variable, which may be associated with different tumor permeability to therapeutic agents among patients.


Subject(s)
Brain Neoplasms/physiopathology , Capillary Permeability/drug effects , KATP Channels/agonists , Potassium Channels, Calcium-Activated/agonists , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Blotting, Western , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Female , Glioblastoma/pathology , Glioblastoma/physiopathology , Glioblastoma/therapy , Humans , Immunohistochemistry , Injections, Intravenous , KATP Channels/genetics , KATP Channels/physiology , Microscopy, Confocal , Minoxidil/administration & dosage , Minoxidil/analogs & derivatives , Minoxidil/therapeutic use , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Neoplasms, Experimental/therapy , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Calcium-Activated/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Vaccination/methods
3.
Clin Cancer Res ; 14(12): 4002-9, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559623

ABSTRACT

PURPOSE: The blood-brain tumor barrier (BTB) significantly limits the delivery of chemotherapeutics to brain tumors. Nitric oxide (NO) is involved in the regulation of cerebral vascular permeability. We investigated the effects of NO donors, L-arginine and hydroxyurea, on BTB permeability in 9L gliosarcoma-bearing Fischer rats. EXPERIMENTAL DESIGN: The rats implanted with 9L gliosarcoma were dosed orally with hydroxyurea and L-arginine. BTB permeability, defined by the unidirectional transport constant, Ki, for [14C]sucrose was measured. The expression of neural and endothelial NO synthase (NOS) in tumors and normal brain tissue was examined. Further, the levels of NO, L-citrulline, and cGMP in the tumor and normal brain tissue were measured. RESULTS: Oral administration of l-arginine or hydroxyurea significantly increased BTB permeability when compared with the nontreated control. The selective effects were abolished by iberiotoxin, an antagonist of calcium-dependent potassium (KCa) channel that is a cGMP pathway effector. The expression of endothelial NOS, but not neural NOS, was higher in tumor vessels than in those of normal brain. Moreover, the levels of NO, L-citrulline, a byproduct of NO formation from L-arginine, and cGMP were enhanced in the tumor tissue by oral administration of L-arginine and/or hydroxyurea. CONCLUSIONS: Oral administration of L-arginine or hydroxyurea selectively increased tumor permeability, which is likely mediated by alteration in cGMP levels. The findings suggest that use of oral NO donors may be a strategy to enhance the delivery of chemotherapeutics to malignant brain tumors.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Capillary Permeability/drug effects , Glioma/pathology , Nitric Oxide Donors/pharmacology , Administration, Oral , Animals , Arginine/administration & dosage , Arginine/pharmacology , Blood-Brain Barrier/physiology , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , Capillary Permeability/physiology , Citrulline/metabolism , Cyclic GMP/metabolism , Drug Delivery Systems , Drug Evaluation, Preclinical , Drug Synergism , Female , Glioma/metabolism , Glioma/physiopathology , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
4.
Mol Cancer ; 6: 22, 2007 Mar 14.
Article in English | MEDLINE | ID: mdl-17359538

ABSTRACT

BACKGROUND: The blood-brain tumor barrier (BTB) impedes the delivery of therapeutic agents to brain tumors. While adequate delivery of drugs occurs in systemic tumors, the BTB limits delivery of anti-tumor agents into brain metastases. RESULTS: In this study, we examined the function and regulation of calcium-activated potassium (KCa) channels in a rat metastatic brain tumor model. We showed that intravenous infusion of NS1619, a KCa channel agonist, and bradykinin selectively enhanced BTB permeability in brain tumors, but not in normal brain. Iberiotoxin, a KCa channel antagonist, significantly attenuated NS1619-induced BTB permeability increase. We found KCa channels and bradykinin type 2 receptors (B2R) expressed in cultured human metastatic brain tumor cells (CRL-5904, non-small cell lung cancer, metastasized to brain), human brain microvessel endothelial cells (HBMEC) and human lung cancer brain metastasis tissues. Potentiometric assays demonstrated the activity of KCa channels in metastatic brain tumor cells and HBMEC. Furthermore, we detected higher expression of KCa channels in the metastatic brain tumor tissue and tumor capillary endothelia as compared to normal brain tissue. Co-culture of metastatic brain tumor cells and brain microvessel endothelial cells showed an upregulation of KCa channels, which may contribute to the overexpression of KCa channels in tumor microvessels and selectivity of BTB opening. CONCLUSION: These findings suggest that KCa channels in metastatic brain tumors may serve as an effective target for biochemical modulation of BTB permeability to enhance selective delivery of chemotherapeutic drugs to metastatic brain tumors.


Subject(s)
Blood-Brain Barrier/pathology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Potassium Channels, Calcium-Activated/metabolism , Animals , Brain Neoplasms/metabolism , Coculture Techniques , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Permeability/drug effects , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/genetics , Potentiometry , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Bradykinin B2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 13(3): 1045-52, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17289901

ABSTRACT

PURPOSE: Histone acetylation is one of the main mechanisms involved in regulation of gene expression. During carcinogenesis, tumor-suppressor genes can be silenced by aberrant histone deacetylation. This epigenetic modification has become an important target for tumor therapy. The histone deacetylation inhibitor, suberoylanilide hydroxamic acid (SAHA), can induce growth arrest in transformed cells. The aim of this study is to examine the effects of SAHA on gene expression and growth of glioblastoma multiforme (GBM) cells in vitro and in vivo. EXPERIMENTAL DESIGN: The effect of SAHA on growth of GBM cell lines and explants was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Changes of the cell cycle and relative gene expression were detected by fluorescence-activated cell sorting, real-time reverse transcription-PCR, and Western blotting. After glioma cells were implanted in the brains of mice, the ability of SAHA to decrease tumor growth was studied. RESULTS: Proliferation of GBM cell lines and explants were inhibited in vitro by SAHA (ED50, 2x10(-6) to 2x10(-5) mol/L, 5 days). SAHA exposure of human U87 and T98G glioma cell lines, DA66 and JM94 GBM explants, as well as a murine GL26 GBM cell line resulted in an increased accumulation of cells in G2-M of the cell cycle. Many proapoptotic, antiproliferative genes increased in their expression (DR5, TNFalpha, p21WAF1, p27KIP1), and many antiapoptotic, progrowth genes decreased in their levels (CDK2, CDK4, cyclin D1, cyclin D2) as measured by real-time reverse transcription-PCR and/or Western blot after these GBM cells were cultured with SAHA (2.5x10(-6) mol/L, 1 day). Chromatin immunoprecipitation assay found that acetylation of histone 3 on the p21(WAF1) promoter was markedly increased by SAHA. In vivo murine experiments suggested that SAHA (10 mg/kg, i.v., or 100 mg/kg, i.p.) could cross the blood-brain barrier as shown by prominent increased levels of acetyl-H3 and acetyl-H4 in the brain tissue. Furthermore, the drug significantly (P<0.05) inhibited the proliferation of the GL26 glioma cells growing in the brains of mice and increased their survival. CONCLUSIONS: Taken together, SAHA can slow the growth of GBM in vitro and intracranially in vivo. SAHA may be a welcome addition for the treatment of this devastating disease.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...