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.
J Clin Pharmacol ; 45(10): 1123-36, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16172177

ABSTRACT

First-time-in-human studies are small, time-lagged dose-escalation studies including volunteer subjects evaluating safety and tolerability. There is little consensus in the design of a first-time-in-human study, and it is difficult to get an overview of studies performed. One hundred five studies comprising 3323 healthy volunteers published in the 5 major clinical pharmacology journals since 1995 were analyzed. The average trial was placebo controlled, double blind including 32 subjects at 5 dose levels but with great variation in cohort size and dose-escalation method. The parallel single-dose design was the most common design, with the crossover designs being more frequent in the early publications. Despite discussions on the optimization of phase I trials, little seems to be happening. The development of study designs and evaluation methods for cancer trials is extensive, but formal statistically based methods and more scientific study designs are unusual in phase I dose-escalation trials in healthy volunteers.


Subject(s)
Clinical Trials, Phase I as Topic/methods , Research Design , Clinical Trials, Phase I as Topic/statistics & numerical data , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Randomized Controlled Trials as Topic
2.
J Clin Pharmacol ; 43(6): 649-60, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12817528

ABSTRACT

The object of this study was to analyze drug interactions between repaglinide, a short-acting insulin secretagogue, and five other drugs interacting with CYP3A4: ketoconazole, rifampicin, ethinyloestradiol/levonorgestrel (in an oral contraceptive), simvastatin, and nifedipine. In two open-label, two-period, randomized crossover studies, healthy subjects received repaglinide alone, repaglinide on day 5 of ketoconazole treatment, or repaglinide on day 7 of rifampicin treatment. In three open-label, three-period, randomized crossover studies, healthy subjects received 5 days of repaglinide alone; 5 days of ethinyloestradiol/levonorgestrel, simvastatin, or nifedipine alone; or 5 days of repaglinide concomitant with ethinyloestradiol/levonorgestrel, simvastatin, or nifedipine. Compared to administration of repaglinide alone, concomitant ketoconazole increased mean AUC0-infinity for repaglinide by 15% and mean Cmax by 7%. Concomitant rifampicin decreased mean AUC0-infinity for repaglinide by 31% and mean Cmax by 26%. Concomitant treatment with CYP3A4 substrates altered mean AUC0-5 h and mean Cmax for repaglinide by 1% and 17% (ethinyloestradiol/levonorgestrel), 2% and 27% (simvastatin), or 11% and 3% (nifedipine). Profiles of blood glucose concentration following repaglinide dosing were altered by less than 8% by both ketoconazole and rifampicin. In all five studies, most adverse events were related to hypoglycemia, as expected in a normal population given a blood glucose regulator. The safety profile of repaglinide was not altered by pretreatment with ketoconazole or rifampicin or by coadministration with ethinyloestradiol/levonorgestrel. The incidence of adverse events increased with coadministration of simvastatin or nifedipine compared to either repaglinide or simvastatin/nifedipine treatment alone. No clinically relevant pharmacokinetic interactions occurred between repaglinide and the CYP3A4 substrates ethinyloestradiol/levonorgestrel, simvastatin, or nifedipine. The pharmacokinetic profile of repaglinide was altered by administration of potent inhibitors or inducers, such as ketoconazole or rifampicin, but to a lesser degree than expected. These results are probably explained by the metabolic pathway of repaglinide that involves other enzymes than CYP3A4, reflected to some extent by a small change in repaglinide pharmacodynamics. Thus, careful monitoring of blood glucose in repaglinide-treated patients receiving strong inhibitors or inducers of CYP3A4 is recommended, and an increase in repaglinide dose may be necessary. No safety concerns were observed, except a higher incidence in adverse events in patients receiving repaglinide and simvastatin or nifedipine.


Subject(s)
Carbamates , Cytochrome P-450 Enzyme System/drug effects , Drug Interactions , Hypoglycemic Agents , Piperidines , Adult , Area Under Curve , Blood Glucose/drug effects , Carbamates/blood , Carbamates/pharmacokinetics , Carbamates/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Female , Half-Life , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Ketoconazole/pharmacology , Male , Piperidines/blood , Piperidines/pharmacokinetics , Piperidines/pharmacology , Rifampin/pharmacology , Simvastatin/pharmacology
3.
J Clin Pharmacol ; 43(5): 470-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12751268

ABSTRACT

A survey of Phase I dose escalation trials published since 1995 shows that there is great disparity in all aspects of the design of the studies, and the cohort sizes range from 2 to 16 subjects with a great variety in the distribution between active and placebo-treated subjects. This study investigates the impact of the cohort size on Type I error and power in Phase I dose escalation trials based on laboratory data, with the hospitalization-induced increase in hepatic enzyme levels taken into consideration. The power of a Phase I dose escalation trial is very low, and only events with a very high probability of occurrence are detectable with acceptable power. For studies with cohort sizes smaller than 6 active subjects, there is much to gain with the inclusion of 1 extra subject, but for more than 10 subjects, little is gained by increasing the cohort size. With increasing cohort sizes, the probability of spontaneous non-drug-related events also increases, and this background rate needs to be considered when evaluating the trial.


Subject(s)
Clinical Trials, Phase I as Topic/methods , Cohort Studies , Pharmaceutical Preparations/administration & dosage , Research Design , Algorithms , Dose-Response Relationship, Drug , Drug Therapy , Humans , Liver/enzymology , Mixed Function Oxygenases/metabolism , Sample Size
4.
Eur J Clin Pharmacol ; 58(10): 683-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12610745

ABSTRACT

OBJECTIVE: NN703 (tabimorelin) is an orally active growth hormone (GH) secretagogue intended for use as an alternative to daily injections of GH. In vitro studies in human liver microsomes have indicated that NN703 is a mechanism-based inhibitor of CYP3A4. The aim of the present study was to investigate in man the effects of NN703 on the pharmacokinetics of midazolam, a substrate of CYP3A4. METHODS: Seventeen adult male subjects were enrolled in the study, and each received an oral dose of midazolam (7.5 mg) on four occasions: at baseline (day 1), after one dose of NN703 (day 3), after 7 days once daily NN703 treatment (day 9) and after a 7-day washout period (day 16). The pharmacokinetics of midazolam and its main metabolite, alpha-hydroxymidazolam, were investigated. RESULTS: Following a single dose of NN703 (day 3), the AUC of both midazolam and alpha-hydroxymidazolam increased by 64% and 34%, respectively (P=0.0001 for both). After repeated NN703 dosing (day 9), NN703 levels reached steady state, and midazolam AUC further increased to 93% relative to baseline (P=0.0001), whereas alpha-hydroxymidazolam AUC decreased slightly and was 11% higher than baseline (n.s.). Following the washout period (day 16), midazolam AUC decreased to values lower than those on day 3 and day 9, but still significantly (45%) higher than baseline levels (P=0.0001). The C(max) values of midazolam and alpha-hydroxymidazolam demonstrated a pattern similar to the AUC, but the effect following repeated NN703 dosing was more pronounced. The t(1/2) of midazolam increased from day 1 to day 3 (P=0.0483) but was essentially unchanged at steady state on day 9. CONCLUSION: This study shows that administration of NN703 and midazolam, a CYP3A4 substrate, leads to a significant increase in exposure of midazolam. This is consistent with NN703 inhibition of CYP3A4 activity.


Subject(s)
Adjuvants, Anesthesia/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors , Dipeptides/pharmacology , Midazolam/analogs & derivatives , Midazolam/pharmacokinetics , Adjuvants, Anesthesia/metabolism , Adult , Area Under Curve , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Dipeptides/adverse effects , Drug Interactions , Half-Life , Humans , Male , Midazolam/metabolism , Middle Aged , Time Factors
5.
Arterioscler Thromb Vasc Biol ; 22(6): 1036-41, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12067917

ABSTRACT

FFR-rFVIIa is an inactivated recombinant factor VIIa (rFVIIa) that inhibits the binding of factor VIIa to tissue factor (TF). It has been shown to prevent TF-induced thrombosis in animals. The present study is a substudy of the Active Site Inhibited Seven (ASIS) trial and examines the antithrombotic effect of 3 doses of FFR-rFVIIa in 24 patients undergoing percutaneous coronary intervention (PCI). Group 1 (n=9) received 400 microg/kg FFR-rFVIIa and 40 to 50 U/kg heparin, group 2 (n=7) received 200 microg/kg FFR-rFVIIa and 100 U/kg heparin, and group 3 (n=8) received 50 microg/kg FFR-rFVIIa and 100 U/kg heparin. Blood thrombogenicity was assessed as total thrombus area and fibrin deposition on the perfusion chamber at shear rate conditions typical of mild-moderate coronary stenosis. Baseline blood thrombogenicity was evaluated a day before PCI, after heparin administration. A second perfusion chamber study was performed just before PCI, 15 minutes after the administration of heparin and FFR-rFVIIa. Thrombus formation at a high shear rate was markedly reduced in groups 1 and 2 after drug administration, by 79% to 84% and 76% to 87%, respectively (P<0.004 [group 1], P<0.04 [group 2]). In group 3, moderate thrombus reduction of 46% to 48% was achieved (P<0.04). Fibrin deposition in all 3 groups was nearly eliminated after drug administration. Our data demonstrate that FFR-rFVIIa has a potent antithrombotic effect at different shear rates and severe arterial injury conditions.


Subject(s)
Factor VIIa/pharmacology , Factor VIIa/therapeutic use , Thromboplastin/antagonists & inhibitors , Thrombosis/prevention & control , Angioplasty, Balloon, Coronary/methods , Animals , Blood Coagulation Tests/methods , Coronary Stenosis/therapy , Double-Blind Method , Drug Administration Schedule , Factor VIIa/administration & dosage , Female , Fibrin/metabolism , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Humans , Injections, Intravenous , Male , Middle Aged , Perfusion/instrumentation , Perfusion/methods , Platelet Function Tests/methods , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...