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.
Invest New Drugs ; 25(2): 139-46, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17077998

ABSTRACT

Silibinin is a polyphenolic flavonoid isolated from milk thistle with anti-neoplastic activity in several in vitro and in vivo models of cancer, including prostate cancer. Silybin-phytosome is a commercially available formulation containing silibinin. This trial was designed to assess the toxicity of high-dose silybin-phytosome and recommend a phase II dose. Silybin-phytosome was administered orally to prostate cancer patients, giving 2.5-20 g daily, in three divided doses. Each course was 4 weeks in duration. Thirteen patients received a total of 91 courses of silybin-phytosome. Baseline patient characteristics included: median age of 70 years, median baseline prostate specific antigen (PSA) of 4.3 ng/ml, and a median ECOG performance status of 0. The most prominent adverse event was hyperbilirubinemia, with grade 1-2 bilirubin elevations in 9 of the 13 patients. The only grade 3 toxicity observed was elevation of alanine aminotransferase (ALT) in one patient; no grade 4 toxicity was noted. No objective PSA responses were observed. We conclude that 13 g of oral silybin-phytosome daily, in 3 divided doses, appears to be well tolerated in patients with advanced prostate cancer and is the recommended phase II dose. Asymptomatic liver toxicity is the most commonly seen adverse event.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Prostatic Neoplasms/drug therapy , Silymarin/analogs & derivatives , Antineoplastic Agents, Phytogenic/adverse effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Excipients , Follow-Up Studies , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/epidemiology , Glucuronides/blood , Glucuronides/metabolism , Glucuronides/urine , Humans , Liver Function Tests , Male , Phosphatidylcholines , Silybin , Silymarin/adverse effects , Silymarin/pharmacokinetics , Silymarin/therapeutic use , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
2.
J Pharmacol Exp Ther ; 318(2): 872-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16644900

ABSTRACT

ZD6474 [N-(4-bromo-2-fluorophenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy]-quinazolin-4-amine; Zactima] is a tyrosine kinase inhibitor with antiangiogenic and antitumor activity currently undergoing human trials for cancer treatment. Pharmacokinetic studies in animal models are an important component in the clinical development of this agent to relate preclinical studies to patient treatment. In the studies presented here, the pharmacokinetics of ZD6474 was determined in plasma and tissues of MCF-7 tumor-bearing nude mice following single p.o. doses at 10, 25, and 50 mg/kg. Plasma area under the curve and Cmax were linear, increasing proportionally with dose. Tissue analysis showed that ZD6474 is extensively distributed to tissues, with liver and lung accumulating concentrations of 212 microg/g (approximately 450 microM) and 161 microg/g (approximately 340 microM), respectively. Tumor levels ranged from 27 to 71 microg/g at Cmax levels across the three dose ranges, and ZD6474 was distributed to all of the tissues in a dose-dependent manner. Analysis of putative ZD6474 metabolites in feces found four, with the N-demethyl-piperidinyl-ZD6474 metabolite being the most prominent but still accounting for less than 2% of the total amount of ZD6474 present. The lack of significant metabolism of ZD6474 is consistent with the relatively long half-life in mice (approximately 30 h), as well as that seen in humans (approximately 120 h), and the primary method of drug elimination appears to be unchanged in the feces (approximately 25%). The incorporation of an empirical approach to dosing in mouse models of cancer in preclinical studies may allow for better prediction of clinical efficacy for ZD6474 alone and in combination with other therapeutic modalities based on equivalent drug exposure.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Piperidines/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacokinetics , Animals , Area Under Curve , Chromatography, Liquid , Dose-Response Relationship, Drug , Drug Delivery Systems , Estradiol/administration & dosage , Estradiol/pharmacology , Feces/chemistry , Female , Liver/drug effects , Liver/metabolism , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Nude , Reproducibility of Results , Tissue Distribution , Transplantation, Heterologous
3.
Biochemistry ; 44(42): 13932-40, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16229482

ABSTRACT

The protonation of crotonyl-CoA dienolate following decarboxylation of glutaconyl-CoA by glutaryl-CoA dehydrogenase was investigated. Although it is generally held that the active sites of acyl-CoA dehydrogenases are desolvated when substrate binds, recent evidence has established that water has access to the active site in these binary complexes of glutaryl-CoA dehydrogenase. The present investigation shows that the dehydrogenase catalyzes (a) a rapid exchange of C-4 methyl protons of crotonyl-CoA with bulk solvent and (b) protonation of crotonyl-CoA dienolate by solvent-derived protons under single turnover conditions. Both of the reactions require the catalytic base, Glu370. These findings indicate that decarboxylation proceeds via a dienolate intermediate. The involvement of water in catalysis by glutaryl-CoA dehydrogenase was previously unrecognized and is in conflict with a classically held intramolecular 1,3-prototropic shift for protonation of crotonyl-CoA dienolate.


Subject(s)
Acyl Coenzyme A/chemistry , Glutaryl-CoA Dehydrogenase/chemistry , Solvents/chemistry , Humans , Kinetics , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Protons
4.
J Pharm Biomed Anal ; 39(3-4): 705-11, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15935603

ABSTRACT

ZD6474 (N-(4-Bromo-2-fluorophenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy] quinazolin-4-amine) is a tyrosine kinase inhibitor with anti-angiogenic and anti-tumor activity that is currently undergoing human trials for cancer treatment. Pharmacokinetic studies in animal models are an important component in clinical development of this agent to relate pre-clinical models to patient treatment. A liquid chromatography tandem mass spectrometry method was developed for the determination of ZD6474 levels in mouse plasma and tissues. Plasma (0.05 mL) and tissue homogenates (0.1 mL of 10 mg/mL) were extracted under alkaline conditions with ethyl acetate:pentane (1:1, v/v) after addition of the internal standard (trazodone, 2-[3-[4-(3-chlorophenyl)-1-piperazinyl]propyl]-1,2,4-triazolo[4,3-a]pyridine-3(2H)-one). Separation was achieved on a C18, 50 mm x 2 mm column with quantitation by internal standard reference and multiple reaction monitoring of the ion transitions m/z 475-->112 (ZD6474) and m/z 372-->176 (trazodone). The calibration curve was linear from a range spanning 20-20,000 ng/mL in plasma and 10-320 ng/mg in tissue homogenates. Mean recoveries from plasma and tissue homogenates were 88 and 90%, respectively. The accuracy in plasma was 88% at the lower limit of quantitation (20 ng/mL with a 50 microL plasma sample) with high precision (R.S.D.%<10%). Assay performance in liver and other tissue homogenates is also reported. The assay was applied to a pharmacokinetic study in mice to determine dosing schedules that would approximate therapeutic ZD6474 levels determined in humans.


Subject(s)
Chemistry, Pharmaceutical/methods , Chromatography, Liquid/methods , Drug Screening Assays, Antitumor/methods , Mass Spectrometry/methods , Neoplasms/drug therapy , Piperidines/analysis , Piperidines/pharmacokinetics , Quinazolines/analysis , Quinazolines/pharmacokinetics , Acetates/analysis , Animals , Antineoplastic Agents/pharmacology , Calibration , Chromatography/methods , Enzyme Inhibitors/pharmacology , Female , Humans , Ions , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Models, Chemical , Pentanes/analysis , Piperidines/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/chemistry , Reproducibility of Results , Sensitivity and Specificity , Time Factors , Tissue Distribution , Trazodone/analysis
5.
Cancer Chemother Pharmacol ; 52(2): 159-66, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12759775

ABSTRACT

PURPOSE: Docetaxel is a semisynthetic taxane derived from the needles of the European yew ( Taxus baccata) and it is an important chemotherapeutic agent in the treatment of recurrent ovarian, breast and non-small-cell lung cancers. Traditional dosing regimens with docetaxel involve doses of 60-100 mg/m(2) by infusion every 3 weeks. Now weekly low-dose (30-36 mg/m(2)) regimens are being evaluated in phase I trials. Such low-dose studies require a more sensitive, specific and rapid assay of docetaxel in biological fluids for the determination of pharmacokinetic parameters. Because docetaxel is primarily metabolized by CYP3A4 and is highly protein-bound in the plasma, there is potential for drug-drug interactions and high interpatient variability in pharmacokinetics. Therefore, pharmacokinetic studies are an important component to understanding the therapeutic variability of docetaxel-containing chemotherapeutic regimens. METHODS: To this end, we developed an analytical assay for docetaxel based upon tandem LCMS and paclitaxel as an internal standard. The sensitivity of the new assay allowed us to monitor plasma levels of docetaxel out to 48 h after the end of the infusion in patients enrolled in a phase I trial of exisulind (orally, twice daily) receiving weekly docetaxel doses of 30 or 36 mg/m(2) where plasma docetaxel levels are below the lower limit of quantitation for traditional HPLC/UV-based assays at later time-points. RESULTS: The inclusion of the 48-h time-point had significant effects on the calculated pharmacokinetic parameters when using either a three-compartment or non-compartmental analysis. The terminal half-life was significantly increased when the 48-h time-point was included in the pharmacokinetic analysis, and the use of model parameters derived with the inclusion of the 48-h time-point were able to more accurately predict plasma levels at later times. CONCLUSIONS: The results reflect the importance of accurate and sensitive analytical methods for the determination of pharmacokinetic parameters and the effect of this later time-point on docetaxel pharmacokinetic modeling. Further, with the increased use of weekly docetaxel in combination with other agents, the inclusion of these later sampling time-points and sensitive methods for drug level determinations are important components in the description of pharmacokinetic drug interactions.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Neoplasms/metabolism , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacokinetics , Taxoids , Antineoplastic Agents, Phytogenic/blood , Area Under Curve , Chromatography, Liquid , Docetaxel , Half-Life , Humans , Infusions, Intravenous , Mass Spectrometry , Models, Biological , Neoplasms/drug therapy , Paclitaxel/blood , Reproducibility of Results , Specimen Handling , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...