Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
JAMA Dermatol ; 150(12): 1345-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25317746

ABSTRACT

IMPORTANCE The KIT receptor is mutated in approximately 15%of acral, mucosal, and chronic, sun-damaged melanomas. The status of KIT mutations is of interest because they usually are mutually exclusive with N-RAS and B-RAF mutations and because of the availability of KIT kinase inhibitors in the clinic. Some recurrent KIT mutations are well characterized; others are poorly described.OBSERVATIONS We describe a novel KIT mutation in a patient with metastatic melanoma. The mutation, located in exon 13, resulted in S628N substitution in the KIT receptor. Using all-atom molecular dynamics simulations, biochemical assays, and cell-based assays, we showed that the mutation is a bona fide gain-of-function oncogenic mutation. Furthermore,we evaluated the sensitivity of the mutant to imatinib and dasatinib.CONCLUSIONS AND RELEVANCE We report a novel KIT gain-of-function mutation with S628N substitution (exon 13) and show that it is sensitive to imatinib in vitro. Therefore, patients with this mutation may be eligible for KIT kinase inhibitor­based therapy. Further studies are needed to evaluate the clinical benefit of such therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Lung Neoplasms/genetics , Melanoma/genetics , Proto-Oncogene Proteins c-kit/genetics , Skin Neoplasms/genetics , Spinal Neoplasms/genetics , Aged, 80 and over , Animals , Benzamides/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Dasatinib , Fatal Outcome , Female , Fibroblasts/cytology , Humans , Imatinib Mesylate , Lung Neoplasms/secondary , Melanoma/secondary , Mutation, Missense , Phosphorylation/drug effects , Piperazines/pharmacology , Point Mutation , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Rats , Skin Neoplasms/pathology , Spinal Neoplasms/secondary , Thiazoles/pharmacology
2.
PLoS One ; 6(11): e27988, 2011.
Article in English | MEDLINE | ID: mdl-22132187

ABSTRACT

Expression of MYC is deregulated in a wide range of human cancers, and is often associated with aggressive disease and poorly differentiated tumor cells. Identification of compounds with selectivity for cells overexpressing MYC would hence be beneficial for the treatment of these tumors. For this purpose we used cell lines with conditional MYCN or c-MYC expression, to screen a library of 80 conventional cytotoxic compounds for their ability to reduce tumor cell viability and/or growth in a MYC dependent way. We found that 25% of the studied compounds induced apoptosis and/or inhibited proliferation in a MYC-specific manner. The activities of the majority of these were enhanced both by c-MYC or MYCN over-expression. Interestingly, these compounds were acting on distinct cellular targets, including microtubules (paclitaxel, podophyllotoxin, vinblastine) and topoisomerases (10-hydroxycamptothecin, camptothecin, daunorubicin, doxorubicin, etoposide) as well as DNA, RNA and protein synthesis and turnover (anisomycin, aphidicholin, gliotoxin, MG132, methotrexate, mitomycin C). Our data indicate that MYC overexpression sensitizes cells to disruption of specific pathways and that in most cases c-MYC and MYCN overexpression have similar effects on the responses to cytotoxic compounds. Treatment of the cells with topoisomerase I inhibitors led to down-regulation of MYC protein levels, while doxorubicin and the small molecule MYRA-A was found to disrupt MYC-Max interaction. We conclude that the MYC pathway is only targeted by a subset of conventional cytotoxic drugs currently used in the clinic. Elucidating the mechanisms underlying their specificity towards MYC may be of importance for optimizing treatment of tumors with MYC deregulation. Our data also underscores that MYC is an attractive target for novel therapies and that cellular screenings of chemical libraries can be a powerful tool for identifying compounds with a desired biological activity.


Subject(s)
Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Doxorubicin/pharmacology , Humans , Inhibitory Concentration 50 , Neuroblastoma/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Topoisomerase I Inhibitors/pharmacology
3.
Acta Biochim Pol ; 58(2): 265-7, 2011.
Article in English | MEDLINE | ID: mdl-21643560

ABSTRACT

In most cells, the major intracellular redox buffer is glutathione (GSH) and its disulfide-oxidized (GSSG) form. The GSH/GSSG system maintains the intracellular redox balance and the essential thiol status of proteins by thiol disulfide exchange. Topoisomerases are thiol proteins and are a target of thiol-reactive substances. In this study, the inhibitory effect of physiological concentration of GSH and GSSG on topoisomerase IIα activity in vitro was investigated. GSH (0-10 mM) inhibited topoisomerase IIα in a concentration-dependent manner while GSSG (1-100 µM) had no significant effect. These findings suggest that the GSH/GSSG system could have a potential in vivo role in regulating topoisomerase IIα activity.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Glutathione/chemistry , Topoisomerase II Inhibitors/chemistry , Antigens, Neoplasm , DNA Topoisomerases, Type II , Enzyme Assays , Humans , Oxidation-Reduction
4.
Invest New Drugs ; 29(6): 1314-20, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20625795

ABSTRACT

Menadione (Vitamin K3) has anti-tumoral effects against a wide range of cancer cells. Its potential toxicity to normal cells and narrow therapeutic range limit its use as single agent but in combination with radiation or other anti-neoplastic agents can be of therapeutic use. In this paper, we first evaluated the early (within 3 h) effect of menadione on ongoing DNA replication. In normal rat cerebral cortex mini-units menadione showed an age dependent anti-proliferative effect. In tissue mini-units prepared from newborn rats, menadione inhibited ongoing DNA replication with an IC (50) of approximately 10 µM but 50 µM had no effect on mini-units from prepared adult rat tissue. The effect of short (72 h) and prolonged exposure (1-2 weeks) to menadione alone in the DBTRG.05MG human glioma cells line and in combination with vitamin C was studied. After short period of exposure data show that menadione alone or in combination with vitamin C provided similar concentration-response curves (and IC(50) values). Prolonged exposure to these drugs was evaluated by their ability to kill 100% of glioma cells and prevent regrowth when cells are re-incubated in drug-free media. In this long-term assay, menadione:vitamin C at a ratio 1:100 showed higher anti-proliferative activity when compared to each drug alone and allowed to reduce each drug concentration between 2.5 to 5-fold. Similar anti-proliferative effect was demonstrated in 8 patient derived glioblastoma cell cultures. Our data should be able to encourage further advanced studies on animal models to evaluate the potential use of this combination therapy for glioma treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Glioma/drug therapy , Vitamin K 3/pharmacology , Age Factors , Animals , Animals, Newborn , Antineoplastic Agents/administration & dosage , Ascorbic Acid/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Glioma/pathology , Humans , Inhibitory Concentration 50 , Rats , Time Factors , Vitamin K 3/administration & dosage , Vitamins/administration & dosage , Vitamins/pharmacology
5.
Blood ; 116(7): 1114-23, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20484085

ABSTRACT

Compared with adults, pediatric mastocytosis has a relatively favorable prognosis. Interestingly, a difference was also observed in the status of c-kit mutations according to the age of onset. Although most adult patients have a D(816)V mutation in phosphotransferase domain (PTD), we have described that half of the children carry mutations in extracellular domain (ECD). KIT-ECD versus KIT-PTD mutants were introduced into rodent Ba/F3, EML, Rat2, and human TF1 cells to investigate their biologic effect. Both ECD and PTD mutations induced constitutive receptor autophosphorylation and ligand-independent proliferation of the 3 hematopoietic cells. Unlike ECD mutants, PTD mutants enhanced cluster formation and up-regulated several mast cell-related antigens in Ba/F3 cells. PTD mutants failed to support colony formation and erythropoietin-mediated erythroid differentiation. ECD and PTD mutants also displayed distinct whole-genome transcriptional profiles in EML cells. We observed differences in their signaling properties: they both activated STAT, whereas AKT was only activated by ECD mutants. Consistently, AKT inhibitor suppressed ECD mutant-dependent proliferation, clonogenicity, and erythroid differentiation. Expression of myristoylated AKT restored erythroid differentiation in EML-PTD cells, suggesting the differential role of AKT in those mutants. Overall, our study implied different pathogenesis of pediatric versus adult mastocytosis, which might explain their diverse phenotypes.


Subject(s)
Mastocytosis/genetics , Mutation/genetics , Phosphotransferases/genetics , Proto-Oncogene Proteins c-kit/genetics , Adult , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Child , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Lymphocytes/metabolism , Mast Cells/metabolism , Mastocytosis/metabolism , Mastocytosis/pathology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction
6.
Biochem Biophys Res Commun ; 393(1): 174-8, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20117079

ABSTRACT

FES is a cytoplasmic tyrosine kinase activated by several membrane receptors, originally identified as a viral oncogene product. We have recently identified FES as a crucial effector of oncogenic KIT mutant receptor. However, FES implication in wild-type KIT receptor function was not addressed. We report here that FES interacts with KIT and is phosphorylated following activation by its ligand SCF. Unlike in the context of oncogenic KIT mutant, FES is not involved in wild-type KIT proliferation signal, or in cell adhesion. Instead, FES is required for SCF-induced chemotaxis. In conclusion, FES kinase is a mediator of wild-type KIT signalling implicated in cell migration.


Subject(s)
Chemotaxis , Proto-Oncogene Proteins c-fes/metabolism , Stem Cell Factor/metabolism , src Homology Domains , Cell Adhesion , Cell Line, Tumor , Humans , Phosphorylation , Proto-Oncogene Proteins c-fes/genetics , Two-Hybrid System Techniques , Tyrosine
7.
Invest New Drugs ; 28(3): 299-305, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19390782

ABSTRACT

The effects of the cyclin-dependent kinase inhibitors roscovitine and olomoucine on DNA synthesis rate during normal rat brain development were studied by using short time (90 min) incubation. Both purine analogues at 100 microM concentration decreased the DNA synthesis of rat cerebral cortex in an age-dependent manner. The maximum inhibitory effect (approximately 90% for roscovitine, approximately 60% for olomoucine) occurred in rats of 2-13 days postnatal age. In adult rats (> 60 days postnatal age), the effect of both purine analogues was low. Roscovitine even at 200 microM concentration did not inhibit the fraction of DNA synthesis insensitive to hydroxyurea (unscheduled DNA synthesis (UDS)). In addition, in the RG2 rat glioma model, roscovitine produced a strong inhibition of DNA synthesis in glioma cells when compared to adult normal tissue. Since in adult rat brain more than 60% of DNA synthesis is related to DNA repair, usually measured as UDS, our results indicate that roscovitine strongly blocks ongoing DNA synthesis connected with replicative processes.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , DNA/biosynthesis , Glioma/drug therapy , Nucleic Acid Synthesis Inhibitors/therapeutic use , Purines/therapeutic use , Age Factors , Animals , Animals, Newborn , Anticarcinogenic Agents/pharmacology , Cell Line, Tumor , DNA/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Kinetin/pharmacology , Kinetin/therapeutic use , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Roscovitine
8.
Pharmacol Res ; 58(1): 32-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18588979

ABSTRACT

Roscovitine is a cyclin-dependent kinase (Cdk) and signal-regulated kinase (Erk1/2) inhibitor that has been shown to be effective against several cancer types including brain tumors. We have shown previously that roscovitine crosses the blood brain barrier (BBB) and is rapidly eliminated from both plasma and brain in adult rats. However, age-dependent kinetics and its effects on the brain have not been reported. In the present study, we investigated the pharmacokinetics of roscovitine in adult and in 14 days old rats after the administration of a single dose of 25 mg/kg. Moreover, we studied the effect of the drug on Cdk5 and Erk1/2 activities in three brain regions, hippocampus, frontal cortex and cerebellum. The pharmacokinetics of roscovitine followed a two-compartment model in both plasma and brain in both adult and young rats. The terminal elimination half-life was 7 h in brain as well as in plasma in rat pups compared to < 0.5 h observed in adult rats. Brain exposure expressed as AUC brain/AUC plasma was 100% in rat pups compared to 20% found in adult rats. Roscovitine induced a significant Cdk5 inhibition and significant Erk1/2 activation in all studied pups brain regions at 2 h. This is the first study describing age-dependent pharmacokinetics of roscovitine and showing the high brain exposure of infant rats to the drug. Thus, roscovitine may be a promising candidate for the treatment of brain tumors in children.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Brain/enzymology , Cyclin-Dependent Kinase 5/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Purines/pharmacokinetics , Age Factors , Animals , Animals, Newborn , Antineoplastic Agents/blood , Area Under Curve , Cerebellum/enzymology , Enzyme Activation , Female , Frontal Lobe/enzymology , Half-Life , Hippocampus/enzymology , Male , Protein Kinase Inhibitors/blood , Purines/blood , Rats , Roscovitine , Tissue Distribution
9.
Cancer Chemother Pharmacol ; 60(6): 841-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17318617

ABSTRACT

Myelosuppression is one the most frequent side effects of chemotherapy. New agents that more selectively target cancer cells have been developed in attempt to improve the effects and to decrease the side effects of cancer treatment. Roscovitine is a purine analogue and cyclin-dependent kinase inhibitor. Several studies have shown its cytotoxic effect in cancer cell lines in vitro and in xenograft models in vivo. In this study, we investigated the effect of roscovitine on hematopoietic progenitors in vitro and in vivo in mice. The clonogenic capacity of hematopoietic progenitors was studied using burst-forming unit-erythroid (BFU-E), colony-forming unit granulocyte, macrophage (CFU-GM) and colony-forming unit granulocyte, erythroid, macrophage, megakaryocyte (CFU-GEMM). In vitro, bone marrow cells were exposed to roscovitine (25-250 microM) in Iscove's modified Dulbecco's media for 4 h or to roscovitine (1-100 microM) in MethoCult media for 12 days. No effect on colony formation was observed after exposure to roscovitine for 4 h; however, concentration- and cell type-dependent effects were observed after 12 days. Roscovitine in concentration of 100 microM inhibited the growth of all types of colonies, while lower concentrations have shown differential effect on hematopoietic progenitors. The most sensitive were CFU-GEMM, followed by BFU-E and then CFU-GM. In vivo, mice were treated with single dose of roscovitine (50, 100 or 250 mg/kg) and the effect on bone marrow was studied on day 1, 3, 6, 9 or 12 after the treatment. In the second part of experiment, the mice were treated with roscovitine 350 mg/kg/day divided into two daily doses for 4 days. The bone marrow was examined on day 1 and 5 after the last dose of roscovitine. On day 1, BFU-E decreased to less than 50% of the controls (P = 0.019). No decrease in BFU-E formation was observed on day 5. No significant effect was observed on CFU-GM and CFU-GEMM growth after the treatment with multiple doses of roscovitine. Single doses of roscovitine or dimethylsulfoxide did not affect the colony formation. We also studied the distribution of roscovitine to the bone marrow after a dose of 50 mg/kg was administered intraperitoneally. Only 1.5% of the drug was detected in the bone marrow. Thus, the roscovitine effect on hematopoietic progenitors in bone marrow in vivo is only transient. One reason may be that only a small fraction of roscovitine reaches the bone marrow. Another explanation may be the short half-life observed for roscovitine that might not allow enough cell exposure to the drug. However, the toxicity of roscovitine to hematopoietic progenitors in vitro is within the same exposure range as cytotoxicity to cancer cells. Thus, precaution should be taken in clinical trials, especially when combinations with myelosuppressive cytostatics are used.


Subject(s)
Bone Marrow Cells/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Protein Kinase Inhibitors/adverse effects , Purines/adverse effects , Animals , Area Under Curve , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Cell Survival/drug effects , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Drug Administration Schedule , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Female , Granulocytes/drug effects , Granulocytes/metabolism , Half-Life , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Purines/administration & dosage , Purines/pharmacokinetics , Roscovitine , Time Factors , Tissue Distribution
10.
Cell Cycle ; 5(19): 2191-4, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17012843

ABSTRACT

The Myc oncogene is deregulated in a wide variety of human tumors hence the Myc pathway is an attractive target for tailored cancer treatment. We have recently identified two small molecules, MYRAs (Myc-pathway response agents), that induce apoptosis in a Myc-dependent manner and inhibit Myc-driven transformation. Here, we show that these compounds in addition have prominent effects in MYCN overexpressing neuroblastoma cells. A third compound, NSC308848, also induced apoptosis in Myc-overexpressing cells and inhibited Myc-induced cellular transformation. However, in contrast to the MYRAs, NSC308848 treatment resulted in decreased Myc protein levels and gave rise to inhibitory effects also on other transcription factors than Myc. Taken together, our findings suggest that these three small molecules can elicit a similar biological response by interfering with the Myc pathway at different levels.


Subject(s)
Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-myc/drug effects , Animals , Anthracenes/pharmacology , Diamines/pharmacology , Humans , Hydrazones/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Purines/pharmacology
11.
Semin Cancer Biol ; 16(4): 318-30, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16934487

ABSTRACT

Myc expression is deregulated in a wide range of human cancers and is often associated with aggressive, poorly differentiated tumors. The Myc protein is a transcription factor that regulates a variety of cellular processes including cell growth and proliferation, cell-cycle progression, transcription, differentiation, apoptosis, and cell motility. Potential strategies that either inhibit the growth promoting effect of Myc and/or activate its pro-apoptotic function are presently being explored. In this review, we give an overview of Myc activation in human tumors and discuss current strategies aimed at targeting Myc for cancer treatment. Such therapies could have potential in combination with mechanistically different cytotoxic drugs to combat and eradicate tumors cells.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genetic Therapy , Humans , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/physiology , Transcription, Genetic
12.
Article in English | MEDLINE | ID: mdl-15894521

ABSTRACT

In the present investigation, the binding of roscovitine (100, 500 and 1500 ng/mL) to plasma proteins was studied at 25 and 37 degrees C by ultrafiltration and equilibrium dialysis methods. Drug stability in plasma was assessed during a 48 h at 4, 25 and 37 degrees C. The effect of thawing and freezing on drug stability was studied. The pKa of roscovitine was measured using capillary electrophoresis coupled with mass spectrometry. Roscovitine was quantified utilizing liquid chromatography and tandem mass spectrometry. Roscovitine is highly bound to plasma proteins (90%). Binding of roscovitine to human serum albumin was constant (about 90%) within concentration range studied while the binding to alpha1-acid glycoprotein decreased with increasing drug concentration indicating that albumin is more important in clinical settings. However, alpha1-acid glycoprotein might be important when plasma proteins change with disease. Protein binding was higher at 25 degrees C compared to 37 degrees C. The results obtained by equilibrium dialysis were in good agreement with those obtained by ultrafiltration. Roscovitine was stable at all temperatures studied during 48 h. Roscovitine has a pKa of 4.4 showing that the drug mainly acts like a weak mono-base. The results obtained in our studies are important prior to clinical trials and to perform pharmacokinetic studies.


Subject(s)
Blood Proteins/chemistry , Purines/chemistry , Dialysis , Drug Stability , Electrophoresis, Capillary , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Orosomucoid/chemistry , Purines/blood , Roscovitine , Serum Albumin/chemistry , Temperature , Ultrafiltration
13.
Eur J Pharm Sci ; 25(1): 91-103, 2005 May.
Article in English | MEDLINE | ID: mdl-15854805

ABSTRACT

The pharmacokinetics, biodistribution and the metabolic pathway of roscovitine were investigated in Sprague-Dawley rats after a single intravenous dose of 25 mg/kg. Blood, lungs, kidney, liver, testis, adipose tissue, spleen and brain were removed at different time-points. Plasma and tissue samples were analyzed using high performance liquid chromatography. The metabolites were identified using liquid chromatography/tandem mass spectrometry and nuclear magnetic resonance spectroscopy. Roscovitine (MW=354) was cleared rapidly from circulation and highly distributed to the tissues. The elimination half-life of roscovitine in plasma and tissues was short (<30 min). A major metabolite (M1) was observed mainly in plasma and in lower levels in all other tissues. M1 was identified as conversion of the hydroxyl-group at C2 to carboxylic acid (MW=368). A second metabolite (M2) was observed mainly in liver and kidney and identified as a hydroxylation product of the C8 of the purine-ring (MW=370). A third metabolite (M3) was found in several organs and corresponded to N-dealkylation of the N9-isopropyl side-chain (MW=312). Roscovitine concentrations in the brain were 30% of that observed in plasma, however no metabolites were detected in brain. In this investigation, three major metabolites of roscovitine were isolated and identified. Also, it was shown that roscovitine eliminates rapidly from both blood and tissues.


Subject(s)
Purines/pharmacokinetics , Animals , Area Under Curve , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Roscovitine , Tissue Distribution
14.
Article in English | MEDLINE | ID: mdl-15686999

ABSTRACT

Roscovitine, a purine analogue that selectively inhibits cyclin-dependent kinases, has been considered as a potential anti-tumor drug. The determination of roscovitine in plasma and urine was performed using microextraction in packed syringe as on-line sample preparation method with liquid chromatography and tandem mass spectrometry. The sampling sorbent utilized was polystyrene polymer. 2H3-lidocaine was used as internal standard. The limit of detection for roscovitine was as low as 0.5 ng/mL and the lower limit of quantification was 1.0 ng/mL. The accuracy and precision values of quality control samples were between +/-15% and < or =11%, respectively. The calibration curve was obtained within the concentration range 0.5-2000 ng/mL in both plasma and urine. The regression correlation coefficients for plasma and urine samples were > or =0.999 for all runs. The present method is miniaturized and fully automated and can be used for pharmacokinetic and pharmacodynamic studies.


Subject(s)
Chromatography, High Pressure Liquid/methods , Enzyme Inhibitors/pharmacokinetics , Mass Spectrometry/methods , Purines/pharmacokinetics , Calibration , Enzyme Inhibitors/blood , Enzyme Inhibitors/urine , Humans , Purines/blood , Purines/urine , Reference Standards , Reproducibility of Results , Roscovitine , Sensitivity and Specificity
15.
J Pharm Biomed Anal ; 34(2): 425-31, 2004 Feb 04.
Article in English | MEDLINE | ID: mdl-15013157

ABSTRACT

Roscovitine (2-(R)-(1-ethyl-2-hydroxyethylamino)-6-benzylamino-9-isopropylpurine, is a potent and selective inhibitor of cyclin-dependent kinases (CDKs). It inhibits cdc2, cdk2, cdk5 and erk 1 and 2 by competing for the ATP binding domain of the kinases. It inhibits cell proliferation; induces DNA fragmentation and causes cell cycle arrest in S phase. Its stability and toxicity are not fully known. A liquid chromatography method was developed to measure roscovitine in human and rat plasma. The lower limit of quantitation (LLOQ) was 100ng/ml; the intra- and inter-day precision was below 10% at all control levels. Likewise, the accuracy between and within days was lower than 6% at all levels. The drug was stable at room temperature. Twenty-four hours at room temperature has result in a decrease of only 9% of the drug. The recovery of roscovitine from plasma was 84% at 750ng/ml. The present method was used to study the pharmacokinetics of the drug in a rat model. The present investigation, to the authors' knowledge, is the first analytical method reported and the first pharmacokinetics investigation of roscovitine in rat. Roscovitine was administered as a bolus injection (25mg/kg body weight). The pharmacokinetic analysis showed that roscovitine is fitted to a two-compartment open-mode with a biphasic elimination half-life (6 and 26min, respectively). The distribution volume was determined to 3.5l/kg and the clearance (Cl) was 29.5ml/min.


Subject(s)
Purines/analysis , Purines/pharmacokinetics , Animals , Chromatography, High Pressure Liquid/methods , Humans , Purines/chemistry , Rats , Rats, Sprague-Dawley , Roscovitine , Spectrophotometry, Ultraviolet/methods
16.
Cancer Lett ; 180(1): 7-12, 2002 Jun 06.
Article in English | MEDLINE | ID: mdl-11911964

ABSTRACT

The effect of roscovitine, a purine analogue and cyclin dependent kinase inhibitor, on DNA synthesis rate in tissue mini-units obtained from human cervical cancers was investigated. Roscovitine (100 microM) gave a DNA synthesis rate inhibition by 61% (P<0.0001; range 23-93%) within 30 min of incubation. This inhibitory effect was concentration-dependent. The results suggest that the inhibition of tumor DNA synthesis rate is due to a direct effect on the DNA synthesis machinery via presently unknown mechanisms. In addition, the potential application of CDKs inhibitors as preventive agents is discussed.


Subject(s)
DNA/biosynthesis , Purines/pharmacology , Uterine Cervical Neoplasms/metabolism , Adult , Aged , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , Middle Aged , Roscovitine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...