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.
Nitric Oxide ; 30: 17-25, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23370169

ABSTRACT

Nitric oxide (NO) released from NO donors can be cytotoxic in tumor cells and can enhance the transport of drugs into brain tumors by altering blood-tumor barrier permeability. The NO donor JS-K [O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate] releases NO upon enzymatic activation selectively in cells overexpressing glutathione-S-transferases (GSTs) such as gliomas. Thus, JS-K-dependent NO effects - especially on cell viability and vascular permeability - were investigated in U87 glioma cells in vitro and in an orthotopic U87 xenograft model in vivo by magnetic resonance imaging (MRI). In vitro experiments showed dose-dependent antiproliferative and cytotoxic effects in U87 cells. In addition, treatment of U87 cells with JS-K resulted in a dose-dependent activation of soluble guanylate cyclase and intracellular accumulation of cyclic guanosine monophosphate (cGMP) which was irreversibly inhibited by the selective inhibitor of soluble guanylate cyclase ODQ (1H-[1,2,4]oxadiazolo(4,3a)quinoxaline-1-one). Using dynamic contrast enhanced MRI (DCE-MRI) as a minimally invasive technique, we demonstrated for the first time a significant increase in the DCE-MRI read-out initial area under the concentration curve (iAUC60) indicating an acute increase in blood-tumor barrier permeability after i.v. treatment with JS-K. Repeated MR imaging of animals with intracranial U87 gliomas under treatment with JS-K (3.5 µmol/kg JS-K 3×/week) and of untreated controls on day 12 and 19 after tumor inoculation revealed no significant changes in tumor growth, edema formation or tumor perfusion. Immunohistochemical workup of the brains showed a significant antiproliferative effect of JS-K in the gliomas. Taken together, in vitro and in vivo data suggest that JS-K has antiproliferative effects in U87 gliomas and opens the blood-tumor barrier by activation of the NO/cGMP signaling pathway. This might be a novel approach to facilitate entry of therapeutic drugs into brain tumors. DCE-MRI is a non-invasive, repeatable imaging modality to monitor biological effects of NO donors and other experimental therapeutics in intracranial tumor models.


Subject(s)
Azo Compounds/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Magnetic Resonance Imaging/methods , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic GMP/metabolism , Glioma/blood supply , Glioma/metabolism , Humans , Immunohistochemistry , Rats , Rats, Nude , Xenograft Model Antitumor Assays
2.
Int J Cancer ; 130(5): 1184-94, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21455987

ABSTRACT

Glutathione-S-transferases (GSTs) are upregulated in malignant gliomas and contribute to their chemoresistance. The nitric oxide (NO) donor PABA/NO (O(2) -{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl} 1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate) generates NO upon selective enzymatic activation by GST-π-inducing selective biological effects in tumors. Tumor cell killing and chemosensitization were observed in a variety of tumors after exposure to GST-activated NO donor drugs. In our project, cytotoxic and chemosensitizing effects of PABA/NO in combination with carboplatin (CPT) and temozolomide (TMZ) were studied in human U87 glioma cells in vitro and in vivo. U87 glioma cells were exposed to PABA/NO alone or in combination with CPT or TMZ for 24 hr. Cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after 24-hr incubation and 48 hr after drug removal. The antiproliferative effect of PABA/NO was assessed in an intracranial U87 glioma nude rat model comparing subcutaneous administration and intratumoral delivery by convection-enhanced delivery. PABA/NO monotherapy showed a strong dose-dependent growth-inhibitory effect in U87 glioma cells in vitro, and a strong synergistic effect was observed after concomitant treatment with TMZ, but not with CPT. Systemic and intratumoral PABA/NO administration significantly reduced cell proliferation, but this did not result in prolonged survival in nude rats with intracranial U87 gliomas. PABA/NO has potent antiproliferative effects, sensitizes U87 glioma cells to TMZ in vitro and shows some in vivo efficacy. Further studies are still required to consolidate the role of NO donor therapy in glioma treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azo Compounds/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Glutathione S-Transferase pi/pharmacology , Nitric Oxide Donors/therapeutic use , para-Aminobenzoates , 4-Aminobenzoic Acid/administration & dosage , 4-Aminobenzoic Acid/therapeutic use , Animals , Azo Compounds/administration & dosage , Brain Neoplasms/mortality , Carboplatin/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Evaluation, Preclinical , Enzyme Activation , Glioma/mortality , Growth Inhibitors/therapeutic use , Humans , Rats , Rats, Nude , Temozolomide
3.
Neurosurgery ; 70(2): 497-510; discussion 510, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21849924

ABSTRACT

BACKGROUND: Glutathione S-transferases (GSTs) control multidrug resistance and are upregulated in many cancers, including malignant gliomas. The diazeniumdiolate JS-K generates nitric oxide (NO) on enzymatic activation by glutathione and GST, showing promising NO-based anticancer efficacy. OBJECTIVE: To evaluate the role of NO-based antitumor therapy with JS-K in U87 gliomas in vitro and in vivo. METHODS: U87 glioma cells and primary glioblastoma cell lines were exposed to JS-K and a variety of inhibitors to study cell death by necrosis, apoptosis, and other mechanisms. GST expression was evaluated by immunocytochemistry, polymerase chain reaction, and Western blot, and NO release from JS-K was studied with a NO assay. The growth-inhibitory effect of JS-K was studied in a U87 xenograft model in vivo. RESULTS: Dose-dependent inhibition of cell proliferation was observed in human U87 glioma cells and primary glioblastoma cells in vitro. Cell death was partially induced by caspase-dependent apoptosis, which could be blocked by Z-VAD-FMK and Q-VD-OPH. Inhibition of GST by sulfasalazine, cGMP inhibition by ODQ, and MEK1/2 inhibition by UO126 attenuated the antiproliferative effect of JS-K, suggesting the involvement of various intracellular death signaling pathways. Response to JS-K correlated with mRNA and protein expression of GST and the amount of NO released by the glioma cells. Growth of U87 xenografts was reduced significantly, with immunohistochemical evidence for increased necrosis and apoptosis and reduced proliferation. CONCLUSION: Our data show for the first time the potent antiproliferative effect of JS-K in gliomas in vitro and in vivo. These findings warrant further investigation of this novel NO-releasing prodrug in gliomas.


Subject(s)
Antineoplastic Agents/pharmacology , Azo Compounds/pharmacology , Glioma/metabolism , Neoplasms, Experimental/drug therapy , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Neoplasms, Experimental/metabolism , Nitric Oxide/biosynthesis , Rats , Rats, Nude , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
4.
J Neurosurg ; 110(1): 128-36, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18991497

ABSTRACT

OBJECT: Exogenous nitric oxide (NO) from NO donors has cytotoxic, chemosensitizing, and radiosensitizing effects, and increases vascular permeability and blood flow in tumors. Yet little is known about whether these cytotoxic and chemosensitizing effects can be observed in glioma cells at doses that alter tumor physiological characteristics in vivo and whether these effects are tumor selective. METHODS: The effect of NO released from proline NONOate, diethylamine NONOate, spermine NONOate, and sodium nitrite on cell proliferation, apoptosis, and chemosensitivity to carboplatin of cultured glioma cells was studied in C6, U87 glioma cells, human glioblastoma cells, and human astrocytes and fibroblasts. RESULTS: Although proline NONOate failed to induce cell death, the other NO donors induced growth arrest when present in high concentrations (10(-2) M) in all cell lines. Chemosensitization was observed after concomitant incubation with spermine NONOate and carboplatin in C6 and human glioblastoma cells. There is strong evidence that cell death occurs primarily by necrosis and to a lesser degree by apoptosis. The NO doses, which altered tumor physiology in vivo, were not cytotoxic, indicating that NO alters vascular permeability and cell viability in vivo by different mechanisms. CONCLUSIONS: The authors found that NO-generating agents at high concentrations are potent growth inhibitors and might also be useful as chemosensitizers in glioma cells. These data corroborate the theory that the use of NOgenerating agents may play a role in the multimodal treatment of malignant gliomas but that the NO release must be targeted more specifically to tumor cells to improve selectivity and efficacy.


Subject(s)
Antineoplastic Agents , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioma/drug therapy , Glioma/pathology , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Apoptosis/drug effects , Astrocytes/drug effects , Carboplatin/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coloring Agents , Dose-Response Relationship, Drug , Drug Synergism , Fibroblasts/drug effects , Humans , Immunohistochemistry , Nitric Oxide/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide Synthase/biosynthesis , Trypan Blue
SELECTION OF CITATIONS
SEARCH DETAIL
...