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










Publication year range
1.
Clin Pharmacol Ther ; 82(2): 137-42, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17632539

ABSTRACT

As the transition to model-based drug development continues, pharmacometric analysis will have an increasingly important role across the entire life cycle of drug discovery, development, regulatory approval, and commercialization. For this reason, pharmacometrics can--and should--have an integrating function in the transformation to model-based development. This essay describes an approach for formalizing the pharmacometrics process using the disciplines encompassed by enterprise engineering.


Subject(s)
Drug Information Services/statistics & numerical data , Models, Theoretical , Pharmacology, Clinical/statistics & numerical data , Animals , Computer Simulation , Drug Approval/methods , Drug Approval/statistics & numerical data , Drug Design , Drug Information Services/trends , Humans , Pharmacology, Clinical/methods , Pharmacology, Clinical/trends
2.
J Clin Pharmacol ; 40(5): 496-507, 2000 May.
Article in English | MEDLINE | ID: mdl-10806603

ABSTRACT

MK-852, a cyclic heptapeptide, is a potent platelet fibrinogen receptor antagonist. When administered to normal healthy male subjects by 1- and 4-hour constant rate intravenous infusions, it provides a generally well-tolerated and reversible means of inhibition of platelet function. At infusion rates of 1 microgram/kg/min for 1 hour and 0.44 microgram/kg/min for 4 hours, respectively, MK-852 extended baseline bleeding time by greater than 2.2-fold and 2.6-fold, inhibited ADP-induced platelet aggregation by 76% and 69%, and inhibited collagen-induced platelet aggregation by 65% and 67%, respectively. The pharmacokinetics of MK-852 include an elimination half-life of approximately 2 hours, total clearance of about 150 ml/min, and volume of distribution of about 18 liters. Examination of the relationship between MK-852 whole-blood concentration in vitro and inhibition of platelet aggregation showed an EC50 of about 55 ng/ml and a Hill coefficient of 1.55. The infusions were generally well tolerated, with no study drug-related changes in blood counts or biochemical profiles.


Subject(s)
Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Adenosine Diphosphate/pharmacology , Adult , Area Under Curve , Collagen/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Oligopeptides/blood , Oligopeptides/pharmacokinetics , Peptides, Cyclic/blood , Peptides, Cyclic/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Thiazolidines , Time Factors
3.
Acta Neuropathol ; 96(4): 395-400, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9797004

ABSTRACT

The brain of Alzheimer's disease patients contains deposits of the 39-42-amino acid (approximately 4 kDa) amyloid beta-peptide, which is derived from the beta-amyloid precursor protein. These pathological deposits have been shown to consist in part of insoluble 8- and 16-kDa aggregates of the amyloid beta-peptide. This report confirms that the amyloid beta-peptide is a substrate for tissue transglutaminase (TGase) and demonstrates that human brain preparations from Alzheimer's disease patients and control patients form cross-linked dimers from added iodinated amyloid beta-peptide. Immunohistochemical staining for TGase revealed its presence in tissue sections and isolated amyloid plaque cores obtained from brains of patients diagnosed as having Alzheimer's disease. These results provide evidence that the previously described insoluble amyloid deposits in Alzheimer's disease may involve TGase-mediated cross-linked amyloid beta-peptide polymers, and suggest a potential role for TGase in the pathogenesis of this disease.


Subject(s)
Brain/enzymology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins , Plaque, Amyloid/enzymology , Transglutaminases/metabolism , Aged , Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/enzymology , Female , Humans , Immunohistochemistry/methods , Male , Polymers/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Reference Values , Staining and Labeling
4.
Eur J Clin Pharmacol ; 54(3): 243-7, 1998 May.
Article in English | MEDLINE | ID: mdl-9681667

ABSTRACT

OBJECTIVE: The aim of the study was to evaluate the effects of food on the rate and extent of eptastigmine absorption in healthy volunteers. METHODS: The study was carried out according to a double-blind, randomized, placebo-controlled, three-way cross-over design. On three separate occasions, six young subjects received 30 mg eptastigmine after a 12-h overnight fast (reference treatment), 30 mg eptastigmine 15 min after a standard breakfast (test treatment) and placebo 15 min after a standard breakfast (control treatment). Acetylcholinesterase activity in red blood cells was assayed 24 h after drug administration as a biological marker of eptastigmine plasma concentrations. RESULTS: Mean maximum acetylcholinesterase inhibition (Imax) was 39.9% after eptastigmine without food and 33.1% after eptastigmine with food. Maximum inhibitions occurred at 4.75 h and 4.88 h after eptastigmine without and with food, respectively. Areas under the curve of acetylcholinesterase per cent inhibition from 0 to 8 h after drug administration (AUC0-8) were 198% h after eptastigmine without food and 124% h after eptastigmine with food. Ninety per cent confidence intervals of test/reference ratios for AUC0-8 and Imax exceeded the 0.80 to 1.20 limits, thus indicating that the two eptastigmine treatments cannot be considered bioequivalent. Mild and transient adverse events were recorded in three subjects receiving eptastigmine without food, one subject receiving eptastigmine with food and one subject receiving placebo. CONCLUSIONS: The ingestion of food significantly reduces the bioavailability of eptastigmine estimated by the assay of red blood cell acetylcholinesterase activity.


Subject(s)
Cholinesterase Inhibitors/pharmacokinetics , Food , Physostigmine/analogs & derivatives , Acetylcholinesterase/blood , Acetylcholinesterase/drug effects , Administration, Oral , Adolescent , Adult , Area Under Curve , Butyrylcholinesterase/blood , Butyrylcholinesterase/drug effects , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/adverse effects , Cross-Over Studies , Dizziness/chemically induced , Double-Blind Method , Headache/chemically induced , Humans , Male , Physostigmine/administration & dosage , Physostigmine/adverse effects , Physostigmine/pharmacokinetics , Treatment Outcome
5.
J Clin Pharmacol ; 37(6): 480-5, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208354

ABSTRACT

The time of peak concentration after administration of oral drug is an often quoted and used pharmacokinetic parameter. It is not well appreciated, however, that the peak times after a single dose and a dose at steady state during a multiple administration regimen can differ significantly. This article derives the mathematical relationships that determine how a peak time at steady state differs from that after a single or first dose. These relationships are then evaluated using three different approaches: 1) graphic simulations of time courses of drug concentration for three hypothetical drugs; 2) comparisons of predicted and observed peak times using examples from the literature; and 3) comparisons of predicted and simulated peak times based on different sampling schedules for three hypothetical drugs. The key finding is that peak times after a dose at steady state can occur considerably earlier after administration than after a single dose. However, the manner by which peak times are usually determined, that is, the sampling time corresponding to the highest measured drug concentration, imposes significant limitations on the usefulness of this parameter.


Subject(s)
Pharmacokinetics , Administration, Oral , Humans
6.
Life Sci ; 60(25): 2323-32, 1997.
Article in English | MEDLINE | ID: mdl-9194688

ABSTRACT

The brain of Alzheimer's disease (AD) patients contains deposits of amyloid beta-peptide (A beta). Recent studies have shown that A beta is a substrate for tissue transglutaminase (TGase), which induces the formation of cross-linked dimers and polymers, and that tacrine, indomethacin and deferoxamine, which have widely different chemical structures, attenuate the progression of symptoms of AD. This report evaluated the potential of a total of ten different pharmacological agents to inhibit TGase-induced cross-linking of A beta, including known TGase inhibitors (dansylcadaverine, spermine), non-steroidal anti-inflammatory drugs (indomethacin, meclofenamic acid, diflunisal, salicylic acid), monoamine oxidase inhibitors (tranylcypromine, phenelzine), an acetylcholinesterase inhibitor (tacrine), and an iron chelating agent (deferoxamine). All but one (salicylic acid) of these ten agents had an inhibitory effect on TGase-induced A beta cross-linking. These results suggest that inhibition of TGase-induced cross-linking of A beta is a potential pharmacologic target for the treatment of AD. A method is also presented for the determination of percent inhibition of TGase-induced A beta cross-linking based on the separated monomer, dimer and polymer bands on SDS-PAGE gels.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Iron Chelating Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Transglutaminases/antagonists & inhibitors , Animals , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Deferoxamine/pharmacology , Guinea Pigs , Humans , Tacrine/pharmacology
7.
Article in English | MEDLINE | ID: mdl-9179555

ABSTRACT

This article summarizes the practical uses of the six key pharmacokinetic parameters, i.e. elimination half-life, volume of distribution, total clearance, fraction of administered dose eliminated by the kidneys, free fraction in plasma, and bioavailability. Emphasis is on the practical medical uses of these parameters in clinical practice and drug development. Numerous pharmacokinetic simulations are used to illustrate the different practical uses. The principles underlying drug dosage regimen design, drug concentration monitoring and adjustment, and dosage adjustment in renal disease are also discussed.


Subject(s)
Clinical Protocols , Dose-Response Relationship, Drug , Drug Evaluation , Pharmacokinetics , Half-Life , Humans , Models, Biological
9.
J Clin Pharmacol ; 36(8): 669-73, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8877669

ABSTRACT

A systematic classification system has been developed for the categorization of therapeutic effects of individual drugs based on their relationships to the underlying disease processes being treated or prevented, rather than on the pharmacologic or biochemical effects of the individual drugs. This system involves a total of six categories of different therapeutic specificities, involving four categories based on disease process-oriented mechanisms of drug action (Categories I through IV), and two additional ones (Categories 0 and V) to complete the classification system. Category 0 includes drugs or drug uses that prevent the development of a disease when none exists; Category I, those that affect etiologic factors of a disease; Category II, those that affect specific disease processes; Category III, those that affect specific disease manifestations; Category IV, those that affect non-specific disease manifestations or symptoms; and Category V includes drug uses or drugs used to diagnose or facilitate treatment of a disease. Examples are provided for each category as well as for applications of the classification system.


Subject(s)
Drug Therapy/classification , Pharmaceutical Preparations/classification , Humans
10.
J Clin Pharmacol ; 36(5): 462-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8739025

ABSTRACT

This study was conducted to examine the pharmacokinetics and pharmacodynamics of tepoxalin in healthy volunteers, an antiinflammatory compound that inhibits cyclooxygenase and lipoxygenase. Tepoxalin was absorbed after oral administration of single doses from 35 to 300 mg, after which it was rapidly converted to an acidic metabolite, RWJ 20142, which inhibits cyclooxygenase but not lipoxygenase. The areas under the concentration-time curve (AUC) of tepoxalin and RWJ 20142 in plasma increased in a dose-dependent fashion. Administration of the lowest dose of tepoxalin completely inhibited whole blood cyclooxygenase for the entire period of observation. This inhibition correlated closely with that of secretion and aggregation induced by collagen of platelets obtained from these subjects. Similarly, administration of tepoxalin was associated with significant inhibition of lipoxygenase in whole blood. Lipoxygenase was inhibited a maximum of 60% in a time-dependent fashion, and the duration of inhibition was dose-dependent. These studies demonstrate that tepoxalin inhibits whole blood cyclooxygenase, lipoxygenase, and platelet function after oral administration in humans.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacokinetics , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/pharmacokinetics , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Administration, Oral , Adult , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Blood Platelets/drug effects , Blood Platelets/physiology , Cyclooxygenase Inhibitors/adverse effects , Dose-Response Relationship, Drug , Humans , Lipoxygenase Inhibitors/adverse effects , Male , Pyrazoles/adverse effects
11.
Clin Pharmacol Ther ; 58(6): 641-9, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8529329

ABSTRACT

The pharmacokinetics of the angiotensin II receptor antagonist losartan potassium and its active carboxylic acid metabolite EXP3174 were characterized in 18 healthy male subjects after administration of intravenous losartan, intravenous EXP3174, and oral losartan. In these subjects, the average plasma clearance of losartan was 610 ml/min, and the volume of distribution was 34 L. Renal clearance (70 ml/min) accounted for 12% of plasma clearance. Terminal half-life was 2.1 hours. In contrast, the average plasma clearance of EXP3174 was 47 ml/min, and its volume of distribution was 10 L. Renal clearance was 26 ml/min, which accounted for 55% of plasma clearance; terminal half-life was 6.3 hours. After oral administration of losartan, peak concentrations of losartan were reached in 1 hour. Peak concentrations of EXP3174 were reached in 3 1/2 hours. The area under the plasma concentration-time curve of EXP3174 was about four times that of losartan. The oral bioavailability of losartan tablets was 33%. The low bioavailability was mainly attributable to first-pass metabolism. After intravenous or oral administration of losartan the conversion of losartan to the metabolite EXP3174 was 14%.


Subject(s)
Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Imidazoles/pharmacokinetics , Tetrazoles/pharmacokinetics , Administration, Oral , Adult , Antihypertensive Agents/administration & dosage , Biphenyl Compounds/administration & dosage , Cross-Over Studies , Humans , Imidazoles/administration & dosage , Infusions, Intravenous , Losartan , Male , Tetrazoles/administration & dosage
12.
J Clin Pharmacol ; 35(10): 1008-15, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8568008

ABSTRACT

Losartan, on orally active, nonpeptide angiotensin II receptor antagonist is being developed as a therapeutic agent for the treatment of hypertension and heart failure. Many patients requiring anticoagulant therapy with warfarin also may have hypertension or heart failure, and thus, are potential candidates for losartan therapy. This study was designed to investigate whether losartan at likely dosage levels would alter the anticoagulant response to warfarin. In a two-period, placebo-controlled, randomized, crossover study, ten healthy male subjects received a single oral dose of 30 mg warfarin sodium on the seventh day of a 13-day treatment with losartan, 100 mg daily by mouth, or placebo. Multiple plasma samples were collected over a 6-day period after both warfarin doses for the measurements of R- and S-warfarin concentrations and prothrombin times. The pharmacokinetics of R- and S-warfarin were comparable in the absence and presence of losartan (no significant effects of losartan on area under the curve, Cmax, or tmax). Losartan also had no significant effect on the anticoagulant effect of warfarin, as assessed by the area under the prothrombin time versus time curve and the maximum response for prothrombin time. The lack of pharmacokinetic or pharmacodynamic interaction between warfarin and losartan observed in this investigation suggests that a clinically important interaction between these drugs is unlikely to occur in patients requiring concomitant administration of both drugs.


Subject(s)
Angiotensin Receptor Antagonists , Anticoagulants/pharmacology , Antihypertensive Agents/pharmacology , Biphenyl Compounds/pharmacology , Imidazoles/pharmacology , Tetrazoles/pharmacology , Warfarin/pharmacology , Administration, Oral , Adolescent , Adult , Anticoagulants/pharmacokinetics , Antihypertensive Agents/therapeutic use , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/therapeutic use , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Drug Interactions , Humans , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Losartan , Male , Prothrombin Time , Receptors, Angiotensin/metabolism , Stereoisomerism , Tetrazoles/administration & dosage , Tetrazoles/therapeutic use , Warfarin/blood , Warfarin/pharmacokinetics
13.
Am J Obstet Gynecol ; 173(3 Pt 1): 801-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7573247

ABSTRACT

OBJECTIVE: The purpose of this study was to assess the effect of 17 beta-estradiol, progesterone, and testosterone on secretion of plasminogen activators and plasminogen activator inhibitor type 1 by cultured endothelial cells. STUDY DESIGN: Bovine aortic endothelial cells were cultured in medium that contained 17 beta-estradiol, progesterone, or testosterone at various concentrations (10(-13) to 10(-6) mol/L). Plasminogen activator activity in culture medium in the presence of cells was assayed after a 36-hour incubation using chromogenic substrate and iodine 125-labeled fibrin plate assays. Plasminogen activator inhibitor type 1 antigen was detected in conditioned media of bovine aortic endothelial cells by Western blotting analysis. RESULTS: All three steroid hormones exhibited biphasic dose-response effects, characterized by stimulation of plasminogen activator secretion at lower concentrations and inhibition of plasminogen activator secretion at higher concentrations. A significant stimulatory effect on plasminogen activator secretion (74% over control) was observed at a 17 beta-estradiol concentration of 10(-12) mol/L (p < 0.03). At higher concentrations of 17 beta-estradiol, progesterone, and testosterone, inhibition of plasminogen activator secretion was observed (p < 0.05). Decreased levels of plasminogen activator inhibitor type 1 antigen were detected in supernatants treated with either 17 beta-estradiol or progesterone at a concentration of 10(-12) mol/L and were maximal at 10(-7) mol/L 17 beta-estradiol, progesterone, and testosterone. CONCLUSION: The secretion of plasminogen activators and plasminogen activator inhibitor type 1 is regulated in a biphasic dose-dependent manner by sex hormones in bovine aortic endothelial cells.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gonadal Steroid Hormones/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/metabolism , Animals , Aorta , Blotting, Western , Cattle , Culture Media, Conditioned , Estradiol/pharmacology , Progesterone/pharmacology , Testosterone/pharmacology
14.
J Clin Pharmacol ; 35(4): 362-7, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7650224

ABSTRACT

A famotidine wafer that rapidly disperses on the tongue without water is a novel alternative to other histamine2 (H2)-antagonist dosage forms. Benefits associated with such a dosage form include convenience and potentially improved compliance for patients who dislike or have difficulty taking tablets and capsules. This report describes the research of three studies on the famotidine wafer dosage form. In the first trial, the bioequivalence and tolerability of the new 40-mg famotidine wafer and the marketed 40-mg famotidine tablet were studied in a 2-period crossover study (n = 18). The two formulations were bioequivalent as assessed by area under the plasma concentration versus time curve and maximum plasma concentration of famotidine. The plasma concentration of famotidine associated with 50% inhibition of pentagastrin stimulated gastric acid secretion (EC50; 10 ng/mL) was attained on average within 0.5 hours post-dose for the wafer and tablet. In a second trial, the tolerability of the famotidine 20-mg and 40-mg wafers or placebo given twice daily (bid) for 14 days were evaluated (n = 192). Both wafer strengths were well and equally tolerated. In a third trial of 450 subjects, the 40-mg wafer was preferred over tablets by 75% of the subjects, when they were asked to consider the method of administration and flavor. When used as an alternative to tablets and other conventional dosage forms, the wafers have the potential therapeutic benefit of improved compliance. It is concluded that similar systemic exposure, excellent tolerability, palatability, and preference make the famotidine wafer a clinically acceptable and convenient dosage from for patients on H2-antagonist therapy.


Subject(s)
Drug Delivery Systems , Famotidine/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Drug Tolerance , Female , Humans , Male , Middle Aged , Patient Compliance , Single-Blind Method , Therapeutic Equivalency
15.
Hypertension ; 25(1): 37-46, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7843751

ABSTRACT

We investigated the effects of angiotensin II (Ang II) type 1 receptor blockade with losartan on the renin-angiotensin-aldosterone system in hypertensive patients (supine diastolic blood pressure, 95 to 110 mm Hg). Qualifying patients (n = 51) were allocated to placebo, 25 or 100 mg losartan, or 20 mg enalapril. Blood pressure, plasma drug concentrations, and renin-angiotensin-aldosterone system mediators were measured on 4 inpatient days: end of placebo run-in, after first dose, and 2 and 6 weeks of treatment. Plasma drug concentrations were similar after the first and last doses of losartan. At 6 weeks, 100 mg losartan and 20 mg enalapril showed comparable antihypertensive activity. Four hours after dosing, compared with the run-in day, 100 mg losartan increased plasma renin activity 1.7-fold and Ang II 2.5-fold, whereas enalapril increased plasma renin activity 2.8-fold and decreased Ang II 77%. Both drugs decreased plasma aldosterone concentration. For losartan, plasma renin activity and Ang II increases were greater at 2 than at 6 weeks. Effects of losartan were dose related. After the last dose of losartan, plasma renin activity and Ang II changes were similar to placebo changes by 36 hours. These results indicate that long-term blockade of the feedback Ang II receptor in hypertensive patients produces modest increases of plasma renin activity and Ang II that do not appear to affect the antihypertensive response to the antagonist.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Biphenyl Compounds/pharmacology , Hypertension/drug therapy , Imidazoles/pharmacology , Renin-Angiotensin System/drug effects , Tetrazoles/pharmacology , Adult , Aged , Angiotensin II/blood , Biphenyl Compounds/therapeutic use , Double-Blind Method , Enalapril/pharmacology , Female , Humans , Hypertension/physiopathology , Imidazoles/therapeutic use , Losartan , Male , Middle Aged , Norepinephrine/blood , Renin/blood , Tetrazoles/therapeutic use
16.
J Clin Pharmacol ; 34(11): 1046-52, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7876394

ABSTRACT

Simulations of time courses of drug concentrations after oral administration are frequently hampered by the lack of pharmacokinetic parameters after oral dosing. This article presents methods by which to estimate such parameters from pharmacokinetic parameters after intravenous dosing and knowledge of time of peak concentration and bioavailability after oral dosing. The application of these approaches enables the generation of meaningful graphic simulations of time courses of drug concentrations after oral administration that can have educational and illustrative uses.


Subject(s)
Pharmacokinetics , Absorption , Administration, Oral
18.
Br J Clin Pharmacol ; 36(5): 464-6, 1993 Nov.
Article in English | MEDLINE | ID: mdl-12959296

ABSTRACT

The influence of dose and food on the pharmacokinetic profile of orally administered verlukast, a leukotriene D4 receptor antagonist, was investigated in 12 healthy male volunteers. This was an open, four-period, single dose, randomised, crossover design including the following doses: one 75 mg tablet, one 250 mg tablet, 500 mg (2 x 250 mg) and 500 mg immediately following a standard meal. There were dose-related increases in the AUC, although after 500 mg verlukast this was disproportionately greater than with 75 mg (P = 0.04). Similarly, there were dose-related increases in C(max). No differences were observed in the t(max) between treatments. With respect to food, there was a 22% decrease (P = 0.02) in C(max) after 500 mg, and the AUC was 13% less (P = 0.052). The differences in the plasma concentration profiles betweeen fasted and fed states are not considered to be of clinical importance.


Subject(s)
Food , Leukotriene Antagonists , Leukotriene D4 , Propionates/pharmacokinetics , Quinolines/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Double-Blind Method , Humans , Male , Propionates/administration & dosage , Propionates/blood , Quinolines/administration & dosage , Quinolines/blood
19.
J Clin Pharmacol ; 33(6): 581-5, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8366184

ABSTRACT

In 15 non-diabetic Type II hypercholesterolemic patients, the effect of 80 mg lovastatin daily on oral glucose tolerance was investigated. Using a randomized, double-blind, two-panel, parallel design, patients on a low cholesterol diet received lovastatin (n = 7) or placebo (n = 8) for 6 weeks. After 6 weeks of treatment, patients receiving lovastatin had a significant reduction in total cholesterol (30%), LDL-cholesterol (36%), and triglycerides (26%). Time courses of plasma glucose and serum insulin changes from baseline after the oral glucose tolerance test were evaluated by AUC. No statistically significant differences were observed in the AUC of changes from baseline between treatment groups or within either treatment group at prestudy, 6 weeks, and poststudy. No patient had a clinically important laboratory or clinical drug-related adverse effect during the study. This study demonstrated that short-term administration of 80 mg lovastatin daily effectively lowers cholesterol without having adverse effects on oral glucose tolerance.


Subject(s)
Blood Glucose/drug effects , Hypercholesterolemia/blood , Lovastatin/pharmacology , Adult , Aged , Blood Glucose/metabolism , Cholesterol, LDL/blood , Double-Blind Method , Fasting/blood , Female , Glucose Tolerance Test , Humans , Male , Middle Aged , Time Factors , Triglycerides/blood
20.
Hypertension ; 21(5): 704-13, 1993 May.
Article in English | MEDLINE | ID: mdl-8491505

ABSTRACT

Losartan is an orally active, nonpeptide angiotensin II (Ang II) (site-1) receptor antagonist. We conducted a multiple-dose study in healthy male volunteers to investigate the tolerability, blood pressure effects, and changes in plasma renin activity (PRA) and plasma Ang II concentration associated with once-daily administration of 100 mg losartan for a week. Subjects were studied on a standardized sodium diet (24-hour urinary sodium excretion, 98 +/- 37 [SD] mEq per 24 hours on the placebo run-in day). Measurements of blood pressure, heart rate, PRA, Ang II, and aldosterone were taken during a placebo run-in day and after single and multiple (7 days) daily doses of losartan (100 mg, n = 10) or placebo (n = 4). Ang II was measured specifically by high performance liquid chromatography coupled with radioimmunoassay. In subjects given losartan, respective decreases (systolic/diastolic) from run-in in supine blood pressure 6 hours after dosing were (mean +/- SD), compared with the placebo run-in day, first dose: -8.8 +/- 9.6/-6.8 +/- 5.0, last dose: -11.6 +/- 8.9/-7.0 +/- 4.8 mm Hg (p < 0.05 for all changes). At this 6-hour time point, corresponding increases from run-in in PRA were from 1.2 +/- 0.6 to 12.0 +/- 6.3 (first dose) and 9.6 +/- 4.9 (last dose) ng angiotensin I per milliliter per hour and in Ang II were from 4.3 +/- 1.7 to 72.4 +/- 33.3 and 45.7 +/- 14.1 pg/mL. All changes in PRA and Ang II were statistically significant within the losartan-treated group, and the biochemical changes were significantly greater than those in the placebo-treated group. The increment in Ang II was less after the last dose than after the first (p < 0.05). The drug was well tolerated by all subjects. These data indicate that, under the conditions of this study, losartan administration (100 mg/day for eight doses over 9 days) results in treatment-related decreases in blood pressure and increases in PRA and Ang II octapeptide.


Subject(s)
Angiotensin II/blood , Biphenyl Compounds/pharmacology , Blood Pressure/drug effects , Imidazoles/pharmacology , Renin/blood , Tetrazoles/pharmacology , Adolescent , Adult , Angiotensin II/antagonists & inhibitors , Biphenyl Compounds/adverse effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Double-Blind Method , Heart Rate/drug effects , Humans , Imidazoles/adverse effects , Losartan , Male , Osmolar Concentration , Radioimmunoassay , Reference Values , Renin-Angiotensin System/drug effects , Tetrazoles/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...