Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Clin Pharmacol ; 40(8): 844-53, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10934668

ABSTRACT

Carvedilol is a relatively new drug with beta- and alpha 1-receptor blocking activity and antioxidant effects recently approved for the treatment of congestive heart failure (CHF). An ascending, multiple-dose study was completed in 20 male patients with stable New York Heart Association (NYHA) Class III or IV CHF. The pharmacokinetics of carvedilol, S(-)-carvedilol, R(+)-carvedilol, and the active metabolites of carvedilol was assessed at steady state after twice-daily oral administration of carvedilol for 7 days at 6.25, 12.5, 25, and 50 mg doses. Carvedilol exhibited stereoselective pharmacokinetics in CHF patients with dose-proportional increases in steady-state plasma concentrations of carvedilol and its enantiomers. Mean AUC and Cmax values for carvedilol were up to twofold higher in patients with Class IV CHF as compared to those with Class III CHF. Steady-state plasma concentrations of the active metabolites also increased in a dose-proportional manner and were typically 10% or less of that observed for carvedilol. In general, carvedilol was adequately tolerated by adult male CHF patients at the dose levels (6.25-50 mg) evaluated in this study as adverse events were consistent with those frequently observed in patients with CHF.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Heart Failure/drug therapy , Propanolamines/pharmacokinetics , Adult , Area Under Curve , Carbazoles/adverse effects , Carvedilol , Heart Failure/metabolism , Humans , Male , Middle Aged , Propanolamines/adverse effects , Stereoisomerism
2.
Am Heart J ; 139(6): 927-33, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10827369

ABSTRACT

BACKGROUND: Platelets play a key role in the pathogenesis of atherosclerosis, thrombosis, and acute coronary and cerebrovascular syndromes. Inhibition of platelet function by acetylsalicylic acid (aspirin) has been shown to reduce the incidence atherothrombotic events in patients with coronary, cerebrovascular, or peripheral vascular disease. Thienopyridine agents, however, including ticlopidine and clopidogrel, inhibit the adenosine diphosphate receptor and have modestly superior effects compared with aspirin on reduction of death, myocardial infarction, and stroke among a broad group of patients with vascular disease. More effective antithrombotic agents are still required to treat patients at high risk for recurrent vascular events. METHODS: Lotrafiban, a selective, nonpeptide antagonist of the human platelet fibrinogen receptor (glycoprotein [GP] IIb/IIIa [alphaIIb/beta3 integrin]), blocks the binding of fibrinogen to the GP IIb/IIIa receptor, which is the final common pathway of platelet aggregation. Lotrafiban at doses of up to 50 mg twice daily was well-tolerated in a 12-week, double-blind, placebo-controlled, dose-ranging study in patients with recent myocardial infarction, unstable angina, transient ischemic attack, or stroke when added to aspirin therapy. On the basis of these results, a dosing regimen was selected for the phase III Blockage of the Glycoprotein IIb/IIIa Receptor to Avoid Vascular Occlusion (BRAVO) trial based on pharmacodynamics and drug tolerability. In the pivotal BRAVO study, lotrafiban therapy is being evaluated in patients who have had a recent myocardial infarction, unstable angina, transient ischemic attack, or ischemic stroke, or who present at any time after a diagnosis of peripheral vascular disease combined with either cardiovascular or cerebrovascular disease. RESULTS: The efficacy evaluation will be based on a composite end point of clinical events (death by any cause, myocardial infarction, stroke, recurrent ischemia requiring hospitalization, or urgent ischemia-driven revascularization). The target enrollment is 9200 patients worldwide. Approximately 700 centers will participate and will be distributed within 30 countries across North America, Europe, Australia, and Asia.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Benzodiazepines , Ischemic Attack, Transient/drug therapy , Myocardial Ischemia/drug therapy , Piperidines , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Administration, Oral , Adolescent , Arterial Occlusive Diseases/blood , Double-Blind Method , Drug Evaluation , Female , Humans , Ischemic Attack, Transient/blood , Male , Myocardial Infarction/blood , Myocardial Infarction/prevention & control , Myocardial Ischemia/blood , Platelet Aggregation Inhibitors/administration & dosage , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Research Design , Secondary Prevention , Stroke/blood , Stroke/prevention & control
3.
Pharmacotherapy ; 19(5): 612-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10331824

ABSTRACT

STUDY OBJECTIVES: To compare eprosartan pharmacokinetics in hemodialysis patients and in volunteers with normal renal function, and to determine the effect of hemodialysis on these values. DESIGN: Open-label, parallel-group, single-dose study. SETTING: Outpatient hemodialysis treatment center and an industry-affiliated clinical pharmacology unit. PATIENTS: Ten healthy volunteers and nine hemodialysis patients. INTERVENTION: A single oral dose of eprosartan 400 mg was administered to volunteers on 1 day and to patients on 2 days (a nondialysis and a dialysis day). Patients underwent high-flux hemodialysis. MEASUREMENTS AND MAIN RESULTS: Concentrations of eprosartan in plasma and dialysate were assayed by high-performance liquid chromatography; plasma protein binding was determined by ultrafiltration. Eprosartan pharmacokinetics showed greater variability in patients than in volunteers. However, six of nine patients had exposures that were within the range observed for volunteers. Mean total AUC(0-t) was increased approximately 60% (95% CI-22, 225) in patients. Total Cmax was similar between groups (PE = 1.01, 95% CI -40, 71). Mean percent fraction unbound (%f(u)) in patients (3.02%) was significantly greater than that in volunteers (1.74%). Unbound AUC(0-t) and unbound Cmax were, on average, approximately 172% (95% CI 28, 479) and 73% (95% CI -1, 199) greater, respectively, in patients. After hemodialysis, the mean %f(u) decreased from 3.19-2.01%. Mean recovery of eprosartan in dialysate was 6.8 mg (range 0-23.1 mg) and hemodialytic clearance was approximately 11 ml/minute, which does not represent a significant portion of total clearance. CONCLUSIONS: Eprosartan was safe and well tolerated in both groups. Based on its known safety profile and because of its exaggerated pharmacokinetic variability in patients undergoing hemodialysis, treatment should be individualized based on tolerability and response. Supplemental doses of eprosartan after hemodialysis are unnecessary.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Renal Dialysis , Renal Insufficiency/metabolism , Thiophenes , Acrylates/adverse effects , Acrylates/blood , Adult , Antihypertensive Agents/adverse effects , Antihypertensive Agents/blood , Female , Humans , Imidazoles/adverse effects , Imidazoles/blood , Male , Middle Aged , Protein Binding
4.
Clin Pharmacol Ther ; 65(5): 473-82, 1999 May.
Article in English | MEDLINE | ID: mdl-10340912

ABSTRACT

OBJECTIVE: To evaluate the pharmacokinetics and pharmacodynamics of an infusion of SB 209670, a non-peptide endothelin-A/endothelin-B receptor antagonist. METHODS: The study was conducted in 2 parts. Part 1 was a placebo-controlled, single-blind, rising-dose crossover evaluation of the pharmacokinetics and safety of SB 209670 infused at doses that ranged from 0.2 to 1.5 mirog kg(-1) for approximately 8 hours in 17 healthy male volunteers. In part 2, renal hemodynamic effects of a 4-hour infusion of SB 209670 were assessed in 10 healthy male volunteers in a 2-period, period-balanced, single-blind, randomized, placebo-controlled crossover study. RESULTS: SB 209670 appeared to display linear kinetics over the dose range from 0.2 to 1.5 microg kg(-1) min(-1). The half-life was approximately 4 to 5 hours. Plasma immunoreactive endothelin-1 increased in an apparent dose-dependent manner. Mean renal hemodynamic responses (para-aminohippurate clearance) increased by approximately 15% relative to placebo (P = .007). Renal sodium excretion was similar during SB 209670 and placebo infusion. CONCLUSION: The pharmacokinetics of intravenous SB 209670 appeared to be linear, and infusion resulted in dose-related increases in immunoreactive endothelin-1. The lack of anti-natriuretic effect and the renal vasodilator response observed in this study indicate that SB 209670 does not possess any partial agonist activity. Further, the renal hemodynamic response supported a potential physiologic role for endogenous endothelin in the maintenance of renal vascular tone in humans.


Subject(s)
Endothelin Receptor Antagonists , Indans/pharmacology , Renal Circulation/drug effects , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Endothelin-1/blood , Humans , Indans/administration & dosage , Indans/pharmacokinetics , Infusions, Intravenous , Male , Middle Aged , Reference Values , Renal Plasma Flow, Effective/drug effects , Single-Blind Method , Vascular Resistance/drug effects
5.
Br J Clin Pharmacol ; 46(3): 267-70, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9764969

ABSTRACT

AIMS: To compare the pharmacokinetics of eprosartan between young (18-45 years) and elderly (65 years) men and between young men and young, premenopausal women (18-45 years). METHODS: Twenty-four subjects (eight subjects/group) received a single 200 mg eprosartan oral dose followed by serial blood sampling over 24 h. RESULTS: Eprosartan was safe and well tolerated. There were no apparent differences in the pharmacokinetics of eprosartan between young females and young males or in the plasma protein binding of eprosartan (98%) for the three groups. On average, AUC (0,infinity) and Cmax values were approximately 2-fold higher in elderly men than young men [AUC (0,infinity) 95% CI: 1.22, 4.34; Cmax 95% CI: 0.98, 4.001. Similarly, unbound AUC (0,infinity) and Cmax values were, on average, approximately 2-fold higher in elderly men than young men [unbound AUC (0,infinity) 95% CI: 1.29, 4.44; unbound Cmax 95% CI: 1.02, 4.12]. tmax was delayed in the elderly men compared with young men, with a median difference of 2.5 h (95% CI: 1.00, 3.01 h). CONCLUSIONS: No gender differences were observed in the pharmacokinetics of eprosartan. There were approximately two fold higher AUC and Cmax values for eprosartan observed in elderly men as compared with young men, most likely due to increased bioavailability of eprosartan in the elderly. Based on the excellent safety profile in the elderly in Phase III clinical trials (doses up to 1200 mg eprosartan) eprosartan can be safely administered to elderly hypertensive patients without an initial dose adjustment. Subsequently, the dose of eprosartan, as for other antihypertensive agents, may be individualized based on tolerability/response.


Subject(s)
Acrylates/pharmacokinetics , Aging/metabolism , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Thiophenes , Acrylates/metabolism , Adult , Aged , Antihypertensive Agents/metabolism , Area Under Curve , Female , Half-Life , Humans , Imidazoles/metabolism , Male , Middle Aged , Premenopause , Protein Binding , Sex Factors
6.
Biopharm Drug Dispos ; 19(6): 351-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9737815

ABSTRACT

Eighteen healthy males received a single 300 mg oral dose of eprosartan as the commercial wet granulation formulation under fasting conditions and following a high-fat breakfast and a single 20 mg intravenous (i.v.) dose. The pharmacokinetics of i.v. eprosartan (mean +/- S.D.) were characterized by a low systemic plasma clearance (131.8 +/- 36.2 mL min(-1)) and a small steady-state volume of distribution (12.6 +/- 2.6 L). Oral bioavailability averaged 13.1%, due to incomplete absorption. In vitro dynamic flow cell dissolution data showed that pH-dependent aqueous solubility of eprosartan is one factor which limits absorption. Eprosartan terminal half-life was shorter after i.v. (approximately 2 h) versus oral (approximately 5-7 h) administration, which may be due to detection of an additional elimination phase or absorption rate-limited elimination following oral administration. Oral administration of eprosartan following a high-fat meal compared with fasting conditions resulted in a similar extent of absorption (based on AUC), but a decreased absorption rate. Cmax was approximately 25% lower, and a median delay of 1.25 h in time to Cmax was observed when eprosartan was administered with food. These minor changes in exposure are unlikely to be of clinical consequence; therefore, eprosartan may be administered without regard to meal times.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Dietary Fats/pharmacology , Food-Drug Interactions , Imidazoles/pharmacokinetics , Thiophenes , Acrylates/administration & dosage , Administration, Oral , Adult , Analysis of Variance , Antihypertensive Agents/administration & dosage , Area Under Curve , Biological Availability , Cross-Over Studies , Dietary Fats/administration & dosage , Half-Life , Humans , Imidazoles/administration & dosage , Injections, Intravenous , Male
7.
J Clin Pharmacol ; 38(7): 649-53, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9702851

ABSTRACT

Eprosartan is an angiotensin II receptor antagonist being developed for the treatment of hypertension and heart failure. The effect of eprosartan on the steady-state anticoagulant activity of warfarin was evaluated in 18 healthy male volunteers. Each subject's daily warfarin dose was titrated over 9 days to achieve a stable international normalized ratio (INR) of 1.3 to 1.6 by day 14. After the 14-day warfarin titration phase, subjects were randomized to receive either eprosartan 300 mg or matching placebo twice a day for 7 days. All subjects continued to take the warfarin dose established during the 14-day titration phase. The anticoagulant activity of warfarin was statistically equivalent when coadministered with eprosartan or with placebo. No serious or unexpected adverse events suggestive of abnormal bleeding occurred during coadministration of eprosartan and warfarin. As measured by the INR, there is no apparent effect of eprosartan on the anticoagulant effect of warfarin.


Subject(s)
Acrylates/pharmacology , Anticoagulants/pharmacology , Antihypertensive Agents/pharmacology , Imidazoles/pharmacology , Thiophenes , Warfarin/pharmacology , Acrylates/adverse effects , Adult , Anticoagulants/adverse effects , Antihypertensive Agents/adverse effects , Double-Blind Method , Drug Interactions , Humans , Imidazoles/adverse effects , International Normalized Ratio , Male , Middle Aged , Warfarin/adverse effects
8.
J Clin Pharmacol ; 38(5): 437-41, 1998 May.
Article in English | MEDLINE | ID: mdl-9602957

ABSTRACT

The effects of antihypertensive agents, including angiotensin II receptor antagonists, on urine uric acid excretion may have important clinical consequences. Therefore, the effects of single and repeated doses of eprosartan on uric acid excretion were evaluated in 57 male patients with mild-to-moderate essential hypertension in a double-blind, randomized, placebo-controlled, repeated dose, dose-rising, two-period, period-balanced, crossover study conducted in two parts. In part 1 (n = 33), the effects of eprosartan dose regimens of 50 mg, 100 mg, and 350 mg once daily and 150 mg every 12 hours on uric acid excretion were assessed. In part 2 (n = 24), the effects of eprosartan dose regimens of 600 mg, 800 mg, and 1,200 mg once daily on uric acid excretion were assessed. Eprosartan was well tolerated. There were no appreciable changes from predose values in fractional excretion of uric acid (FEua), urine uric acid excretion, urine uric acid to creatinine (Uua/Ucr) ratios, or serum uric acid concentrations after single or repeated doses of eprosartan. Mean Uua/Ucr ratios for eprosartan doses of 50 mg, 100 mg, or 350 mg daily or 150 mg every 12 hours were comparable to those for placebo. Mean FEua values and Uua/Ucr ratios for eprosartan doses of 600 mg, 800 mg, or 1,200 mg daily also were comparable to those for placebo. Single and repeated oral doses of eprosartan ranging from 50 mg to 1,200 mg daily had no effect on serum uric acid concentrations or urine uric acid excretion in patients with mild-to-moderate essential hypertension.


Subject(s)
Acrylates/pharmacology , Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Hypertension/metabolism , Imidazoles/pharmacology , Thiophenes , Uric Acid/urine , Acrylates/administration & dosage , Adult , Antihypertensive Agents/administration & dosage , Creatinine/urine , Cross-Over Studies , Double-Blind Method , Humans , Imidazoles/administration & dosage , Male , Middle Aged , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Uric Acid/blood
9.
J Clin Pharmacol ; 38(1): 34-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9597557

ABSTRACT

The present study investigated the proportionality of exposure after single oral doses of 100, 200, 400, and 800 mg of eprosartan, a nonpeptide, nonbiphenyl angiotensin II receptor antagonist, in 23 healthy young men. Eprosartan was safe and well tolerated. Exposure to eprosartan increased with dose but in a less than proportional manner. For each two-fold dose increase, area under the concentration--time curve (AUC) increased an average of 1.6 to 1.8 times and maximum plasma drug concentration (Cmax) increased an average of 1.5 to 1.8 times. For both parameters, the greatest difference from the dose multiple was observed between the 400- and 800-mg doses. Dose proportionality of eprosartan, as assessed by an equivalence-type approach using the 100-mg dose as the reference and a 30% acceptance region (0.70, 1.43), was achieved for the 200- and 400-mg doses for AUC and the 200-mg dose for Cmax. The observed changes in the pharmacokinetics of eprosartan suggest slight saturation of absorption of eprosartan over the 100- to 800-mg dose range, most likely due to the physicochemical properties of the drug (pH-dependent aqueous solubility and lipophilicity).


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Thiophenes , Acrylates/administration & dosage , Acrylates/adverse effects , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male
10.
Clin Pharmacol Ther ; 63(4): 471-81, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9585802

ABSTRACT

STUDY DESIGN: The effects of orally administered eprosartan on changes induced by angiotensin II in blood pressure, renal hemodynamics, and aldosterone secretion were evaluated in healthy men in this double-blind, randomized, single-dose, placebo-controlled crossover study, which was conducted in three parts. Part 1 (n = 12) assessed the onset and duration of the effect of eprosartan 350 mg or placebo; part 2 (n = 14) assessed the dose-response profile of placebo or 10, 30, 50, 70, 100 or 200 mg eprosartan; and part 3 (n = 5) assessed the duration of the effect of 50, 100, or 350 mg eprosartan. RESULTS: In part 1 of the study; 350 mg eprosartan caused complete inhibition of angiotensin II-induced pressor and renal blood flow hemodynamic effects (effects on effective renal plasma flow [ERPF]) and inhibited angiotensin II-induced stimulation of aldosterone secretion from 1 to 3 hours after administration. Eprosartan, 350 mg, inhibited the effects of exogenous angiotensin II by approximately 50% to 70% from 12 to 15 hours after dosing. Eprosartan had no angiotensin II agonistic activity and produced an increase in ERPF starting at 1 to 4 hours after dosing. In study part 2, at 3 hours after single doses of 10, 30, 50, 70, 100, and 200 mg, eprosartan inhibited angiotensin 11-induced decreases in ERPF by 39.1%, 49.9%, 33.0%, 56.0%, 71.0%, and 85.7%, respectively, compared with placebo. In study part 3, 50, 100, and 350 mg eprosartan produced measurable Inhibition of angiotensin II-induced decreases in ERPF from 12 to 15 hours after administration. In parts 2 and 3, the eprosartan angiotensin II antagonism on blood pressure response and aldosterone secretion mirrored the angiotensin II antagonism on ERPF.


Subject(s)
Acrylates/pharmacology , Aldosterone/metabolism , Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Imidazoles/pharmacology , Renal Circulation/drug effects , Thiophenes , Acrylates/administration & dosage , Adult , Aldosterone/blood , Antihypertensive Agents/administration & dosage , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Male , Potassium, Dietary/administration & dosage , Reference Values , Sodium, Dietary/administration & dosage , Time Factors
11.
J Clin Pharmacol ; 38(2): 129-37, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9549643

ABSTRACT

This was an open-label, parallel group study to compare the pharmacokinetics of multiple oral doses of eprosartan in subjects with normal renal function (Clcr > 80 mL/min; n = 8) and patients with mild (Clcr 60-80 mL/min; n = 8), moderate (Clcr 30-59 mL/min; n = 15), or severe (Clcr < 30 mL/min; n = 3) renal insufficiency. Each subject received oral eprosartan 200 mg twice daily for 6 days and a single dose on day 7. Mean total maximum concentration (Cmax) and area under the concentration-time curve from 0 to 12 hours (AUC0-12) were similar for healthy subjects and those with mild renal impairment, but were an average of 25% to 35% and 51% to 55% greater for patients with moderate and severe renal impairment, respectively, compared with healthy subjects. Mean renal clearance (Clr), which was similar for healthy subjects and patients with mild renal impairment, was decreased an average of 41% and 95% in the groups with moderate and severe renal impairment, respectively, compared with normal subjects. Eprosartan was highly bound to plasma proteins in all groups; however, the unbound fraction was increased approximately two-fold in the group with severe renal impairment. Mean unbound Cmax and AUC0-12 were an average of 53% to 61% and 185% to 210% greater for the patients with moderate and severe renal impairment, respectively, compared with healthy subjects. Headache was the most common adverse experience reported in all subgroups. Eprosartan was safe and well tolerated regardless of degree of renal impairment. Cmax and AUC were increased and renal clearance decreased in patients with moderate to severe renal impairment in comparison to healthy subjects and patients with mild renal impairment. However, based on the moderate renal clearance and known safety profile of eprosartan, it is not necessary to adjust the dose of eprosartan in patients with renal insufficiency.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Renal Insufficiency/metabolism , Thiophenes , Acrylates/administration & dosage , Acrylates/blood , Adult , Aged , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Area Under Curve , Humans , Imidazoles/administration & dosage , Imidazoles/blood , Metabolic Clearance Rate , Middle Aged , Renal Insufficiency/blood
12.
Ann Pharmacother ; 32(3): 304-8, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9533060

ABSTRACT

OBJECTIVE: To assess the effect of ranitidine on the pharmacokinetics of eprosartan in healthy male volunteers. DESIGN: Single-center, randomized, open-label, two-period, period-balanced, crossover study. PATIENTS: Seventeen healthy men aged 19 to 43 years. INTERVENTION: In each period (separated by a > or = 7 d washout), subjects received a single 400-mg oral dose of eprosartan alone, or a single oral dose of eprosartan 400 mg and ranitidine 150 mg on day 4 after 3 days of ranitidine 150 mg twice daily. Serial pharmacokinetic samples were obtained for up to 24 hours following eprosartan dosing. MAIN OUTCOME MEASURES: Plasma and urine eprosartan concentrations during each treatment session. RESULTS: Eprosartan maximum concentration (Cmax), the AUC from time-zero to the last quantifiable concentration (AUC0-t), and renal clearance (Cl(r)) values were approximately 7%, 11%, and 4% lower, respectively, when administered with ranitidine compared with eprosartan alone. The 95% CIs for the ratio of eprosartan plus ranitidine compared with eprosartan alone were 0.81 to 1.07, 0.77 to 1.03, and 0.64 to 1.43, for Cmax, AUC0-t, and Cl(r), respectively, indicating no statistically significant difference between regimens. CONCLUSIONS: Repeated doses of ranitidine did not have a marked effect on the single-dose pharmacokinetics of eprosartan.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Histamine H2 Antagonists/pharmacology , Imidazoles/pharmacokinetics , Ranitidine/pharmacology , Thiophenes , Acrylates/blood , Acrylates/urine , Administration, Oral , Adult , Angiotensin II/antagonists & inhibitors , Antihypertensive Agents/blood , Antihypertensive Agents/urine , Area Under Curve , Cross-Over Studies , Histamine H2 Antagonists/adverse effects , Humans , Imidazoles/blood , Imidazoles/urine , Male , Ranitidine/adverse effects
13.
Pharmacotherapy ; 18(1): 42-50, 1998.
Article in English | MEDLINE | ID: mdl-9469680

ABSTRACT

STUDY OBJECTIVE: To evaluate the pharmacokinetics and plasma protein binding of eprosartan in hepatic disease. DESIGN: Single-dose, parallel-group study. SETTING: Oklahoma Foundation for Digestive Research, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. PATIENTS: Eight healthy subjects with normal hepatic function and eight patients with hepatic disease. INTERVENTION: All subjects received a single oral dose of eprosartan 100 mg. MEASUREMENTS AND MAIN RESULTS: Eprosartan plasma concentrations were quantified by high-performance liquid chromatography; plasma protein binding was determined by ultrafiltration. Using analysis of variance, point estimates (PE) and 90% CIs were calculated. Total and unbound maximum concentrations and degree of plasma protein binding were similar for both groups. Total area under the plasma concentration-time curve (AUC0-t) was approximately 40% higher for the group with hepatic disease (PE 1.42, 90% CI 0.94-2.14). Similarly, unbound AUC0-t was approximately 50% higher (PE 1.53, 90% CI 0.98-2.39). CONCLUSION: Eprosartan was safe and well tolerated by both groups. Based on the increase in AUC in patients with hepatic disease compared with those with normal hepatic function, the dosage of eprosartan in patients with hepatic disease should be individualized based on tolerability and response.


Subject(s)
Acrylates/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Liver Diseases/metabolism , Thiophenes , Adult , Area Under Curve , Humans , Male , Metabolic Clearance Rate , Middle Aged
14.
Clin Pharmacol Ther ; 62(4): 417-25, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9357393

ABSTRACT

OBJECTIVE: To investigate the effect of steady-state fluconazole administration on the disposition of eprosartan, losartan, and E-3174. METHODS: Sixteen healthy male subjects received 300 mg eprosartan every 12 hours, and 16 received 100 mg losartan every 24 hours on study days 1 to 20. All 32 subjects received 200 mg fluconazole every 24 hours beginning on day 11 and continuing through day 20. Serial blood samples were collected over one dosing interval on study days 10 and 20 for measurement of plasma concentrations of eprosartan, losartan, and E-3174 (the active metabolite of losartan). RESULTS: There was no significant difference in eprosartan area under the concentration-time curve from time 0 to time of last quantifiable concentration [AUC(0-t)] or maximum concentration (Cmax) when administered alone and with fluconazole. After concomitant administration with fluconazole, losartan AUC(0-t) and Cmax were significantly increased 66% and 30%, respectively, compared with those values for losartan alone. The AUC(0-t) and Cmax for E-3174 were significantly decreased 43% and 56%, respectively, after administration of losartan with fluconazole. CONCLUSIONS: Fluconazole significantly increases the steady-state AUC of losartan and inhibits the formation of the active metabolite of losartan, E-3174. In contrast, fluconazole administration has no effect on the steady-state pharmacokinetics of eprosartan.


Subject(s)
Acrylates/pharmacokinetics , Antifungal Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Fluconazole/pharmacology , Imidazoles/pharmacokinetics , Losartan/pharmacokinetics , Steroid 16-alpha-Hydroxylase , Tetrazoles/pharmacokinetics , Thiophenes , Acrylates/administration & dosage , Adult , Antifungal Agents/administration & dosage , Antihypertensive Agents/administration & dosage , Area Under Curve , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Administration Schedule , Fluconazole/administration & dosage , Humans , Imidazoles/administration & dosage , Losartan/administration & dosage , Male , Middle Aged , Reference Values , Steroid Hydroxylases/drug effects , Steroid Hydroxylases/metabolism , Tetrazoles/administration & dosage , Time Factors
15.
Br J Clin Pharmacol ; 43(6): 661-4, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9205830

ABSTRACT

AIMS: To study the effect of eprosartan, a nonbiphenyl tetrazole angiotensin II receptor antagonist, on digoxin pharmacokinetics in a randomized, open-label, two period, period balanced crossover study in 12 healthy men. METHODS: Each subject received a single 0.6 mg oral dose of digoxin (Lanoxicaps 0.2 mg/capsule, Glaxo Wellcome) alone or following 4 days of dosing with eprosartan 200 mg orally every 12 h. Each study period was separated by a 14 day washout interval. Serial blood samples were obtained for up to 96 h after each digoxin dose for determination of digoxin pharmacokinetics. The effect of eprosartan on digoxin pharmacokinetics was assessed through an equivalence-type approach using AUC(0, t') as the primary endpoint. RESULTS: For AUC(0, t'), the ratio of digoxin+eprosartan: digoxin alone was 0.99 with a 90% confidence interval (CI) of [0.90, 1.09]. For Cmax, the ratio was 1.00 with a 90% CI of [0.86, 1.17]. tmax was similar for both regimens. Both regimens were safe and well tolerated. CONCLUSIONS: Based on AUC and Cmax data, it can be concluded that eprosartan has no effect on the pharmacokinetics of a single oral dose of digoxin.


Subject(s)
Acrylates/pharmacology , Anti-Arrhythmia Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Digoxin/pharmacokinetics , Imidazoles/pharmacology , Thiophenes , Acrylates/administration & dosage , Administration, Oral , Adult , Anti-Arrhythmia Agents/administration & dosage , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Area Under Curve , Cross-Over Studies , Digoxin/administration & dosage , Digoxin/blood , Drug Interactions , Humans , Imidazoles/administration & dosage , Iodine Radioisotopes , Isotope Labeling , Male , Radioimmunoassay , Receptors, Angiotensin/drug effects , Receptors, Angiotensin/metabolism
16.
Am J Health Syst Pharm ; 54(10): 1172-6, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9161624

ABSTRACT

The cardiovascular effects of granisetron given as a 30-second i.v. bolus dose and of granisetron and ondansetron given by currently recommended methods were studied. Healthy adults 18 to 50 years of age were randomly assigned to one of four treatments during each of four study periods: granisetron 10 micrograms/kg (as the hydrochloride salt) i.v. over 5 minutes, granisetron 10 micrograms/kg i.v. over 30 seconds, ondansetron 32 mg (as the hydrochloride salt) i.v. over 15 minutes, and placebo. During each study period, the researchers gave each subject three sequential injections using a double-blind, double-dummy technique. Each subject was to receive all four regimens. Two resting 12-lead electrocardiograms (ECGs) were obtained before the regimen, and one was obtained at the end of each injection and at intervals up to 24 hours after the third injection. Sitting blood pressure and pulse were measured before treatments, immediately after the end of each injection, and at intervals up to 24 hours after injection 3. Safety data were analyzed for 13 subjects, and ECG interval data for 12 of them. The mean postdose QTc interval differed significantly among regimens. There were no other significant regimen-associated differences among the four mean results for any ECG interval. The mean post-dose QTc interval for ondansetron was significantly greater than that for each of the other regimens. The drug regimens were comparable in safety and tolerability. A total of 20 adverse effects, all mild to moderate, were reported in 10 subjects. Changes in vital signs were minimal. There were no clinically important cardiovascular changes associated with the i.v. administration of granisetron 10 micrograms/kg over 30 seconds, granisetron 10 micrograms/kg over 5 minutes, or ondansetron 32 mg over 15 minutes in healthy adults.


Subject(s)
Antiemetics/administration & dosage , Granisetron/administration & dosage , Heart/drug effects , Ondansetron/pharmacology , Adult , Antiemetics/adverse effects , Double-Blind Method , Drug Interactions , Electrocardiography/drug effects , Female , Granisetron/adverse effects , Headache/chemically induced , Heart/physiology , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Ondansetron/adverse effects
17.
J Clin Pharmacol ; 37(2): 155-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9055142

ABSTRACT

The potential for eprosartan, a nonbiphenyl tetrazole angiotensin II receptor antagonist, to affect the 24-hour plasma glucose profiles in type II diabetic patients treated with glyburide was investigated in this randomized, placebo-controlled, double-blind (eprosartan-placebo phase only), two-period, period-balanced, crossover study. All patients received a stable oral dose (3.75-10 mg/day) of glyburide for at least 30 days before the first dose of double-blind study medication was administered. Patients were randomized to receive either 200-mg oral doses of eprosartan twice daily or matching oral placebo doses concomitantly with glyburide for 7 days during each treatment period. After a minimum washout period of 14 days, patients were crossed over to the alternate treatment. Serial samples to measure glucose concentrations in plasma were collected over a 24-hour period on the day before administration of eprosartan or placebo and again on day 7. Mean glucose concentrations were comparable between treatment groups before administration of eprosartan or placebo. The point estimate (90% confidence interval) for the ratio of the average mean 24-hour plasma glucose concentrations of eprosartan + glyburide to placebo + glyburide after 7 days of administration was 0.96 (0.90, 1.01). Eprosartan did not significantly alter the 24-hour plasma glucose profile in patients with type II diabetes mellitus who were previously stabilized on glyburide.


Subject(s)
Acrylates/pharmacology , Antihypertensive Agents/pharmacology , Diabetes Mellitus, Type 2/metabolism , Glyburide/therapeutic use , Hypoglycemic Agents/therapeutic use , Imidazoles/pharmacology , Thiophenes , Acrylates/adverse effects , Angiotensin Receptor Antagonists , Antihypertensive Agents/adverse effects , Blood Glucose/drug effects , Cross-Over Studies , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Drug Interactions , Female , Humans , Imidazoles/adverse effects , Male , Middle Aged , Sweating
18.
J Infect Dis ; 174(3): 463-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8769601

ABSTRACT

Single ascending doses of RSHZ19 (also known as SB 209763), a humanized monoclonal antibody (MAb) directed to the fusion protein of respiratory syncytial virus, were administered to healthy men to evaluate the safety, pharmacokinetics, antigenicity, and fusion inhibition (FI) activity of RSHZ19. Doses of RSHZ19 (0.025-10.0 mg/kg) or placebo were infused over 30 min, and subjects were followed for 10 weeks. Plasma concentrations of RSHZ19 and RSHZ19-specific antibodies were determined by ELISAs. FI titers were used to evaluate the ability of plasma to inhibit virus-induced fusion of VERO cells previously infected with RS Long strain virus. Twenty-six subjects, mean age 24, completed the study. RSHZ19 was safe and well tolerated, and no subject developed antibodies to RSHZ19 during follow-up. RSHZ19 had low plasma clearance and a half-life of approximately 23 days, similar to native IgG. Increases in FI titers relative to pretreatment levels were seen 24 h after MAb administration in all 4 subjects given 10 mg/kg and in 2 of 4 given 5 mg/kg.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Viral/analysis , Respiratory Syncytial Viruses/immunology , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antibodies, Viral/blood , Humans , Male , Middle Aged , Placebos , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/therapy , Single-Blind Method , Viral Fusion Proteins/immunology
19.
J Clin Pharmacol ; 34(12): 1199-207, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7738216

ABSTRACT

The pharmacokinetic profile of penciclovir was determined after a single 500-mg dose of its oral precursor, famciclovir, in 9 healthy volunteers and in 14 patients with chronic hepatic disease. Plasma and urine samples were analyzed for concentrations of penciclovir and 6-deoxy-penciclovir using a reverse-phase high-performance liquid chromatography (HPLC) method. Famciclovir was not quantifiable in patients with hepatic disease, and 6-deoxy-penciclovir was quantifiable in only a limited number of specimens. The extent of systemic availability of penciclovir, as measured by AUC0-infinity, was similar in patients with hepatic disease and in healthy subjects. In contrast, Cmax was significantly lower (average decrease of 43%) in subjects with hepatic disease relative to healthy normal subjects. Median Tmax for subjects with hepatic disease was significantly increased (by 0.75 hours) compared with subjects with normal liver function. These data suggest a decrease in the rate, but not the extent, of systemic availability of penciclovir in patients with hepatic disease. It should be unnecessary to modify the dose of famciclovir for subjects with compensated hepatic disease and normal renal function.


Subject(s)
2-Aminopurine/analogs & derivatives , Acyclovir/analogs & derivatives , Liver Diseases/metabolism , Prodrugs/pharmacokinetics , 2-Aminopurine/administration & dosage , 2-Aminopurine/pharmacokinetics , Acyclovir/blood , Acyclovir/pharmacokinetics , Acyclovir/urine , Administration, Oral , Adolescent , Adult , Biological Availability , Chromatography, High Pressure Liquid , Chronic Disease , Famciclovir , Female , Guanine , Humans , Male , Metabolic Clearance Rate , Middle Aged , Prodrugs/administration & dosage
20.
J Clin Pharmacol ; 34(11): 1098-108, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7876402

ABSTRACT

Nonsteroidal antiinflammatory drugs differ with respect to their effects on prostaglandin metabolism in various tissues, a property that may be partly responsible for some of the differences in the pharmacologic activities and side-effect profiles that are associated with their use. The effects of nabumetone on urinary prostaglandin excretion have not been reported. Fourteen healthy females, age 21-43 years, were treated with nabumetone (NAB) 1000 mg daily, sulindac (SUL) 200 mg every 12 hours, and indomethacin (IND) 50 mg every 12 hours for 7 days in a randomized period-balanced crossover study. The effects of drug treatment on urinary prostaglandin excretion (PGE2, 6-keto-PGF1 alpha, PGF2 alpha, thromboxane [TX] B2) and platelet function (collagen-induced whole blood platelet aggregation [CIPA] and template bleeding time) were determined on day 1 and day 7. For each treatment regimen, mean baseline urinary PG excretion values were comparable for each prostanoid, but the pattern of excretion differed in response to each drug. Treatment with NAB significantly increased the urinary excretion rates of PGE2 and PGF2 alpha, but 6-keto-PGF1 alpha and TXB2 excretion were unchanged. IND treatment did not result in a significant change in PGE2 excretion but did significantly reduce urinary 6-keto-PGF1 alpha and TXB2 excretion rates. Reduced excretion of PGF2 alpha was observed on both study days during treatment with IND and SUL. SUL treatment also resulted in increased urinary PGE2 excretion while significantly reducing 6-keto-PGF1 alpha excretion on day 7. Significant differences were observed between the NAB and SUL regimens with respect to PGF2 alpha excretion and between the NAB and SUL regimens for PGE2, PGF2 alpha, 6-keto-PGF alpha 1 (on day 1 only) and TXB2 (on day 1 only). Neither NAB nor SUL caused inhibition of CIPA or bleeding time although platelet aggregation was inhibited during IND treatment. That NAB treatment was neither associated with alterations in platelet function nor decreases in the urinary excretion of the vasodilatory prostaglandins, PGE2 and 6-keto-PGF1 alpha, suggests that NAB possesses renal sparing properties.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Platelets/drug effects , Butanones/pharmacology , Indomethacin/pharmacology , Prostaglandins/urine , Sulindac/pharmacology , Adult , Bleeding Time , Butanones/pharmacokinetics , Cross-Over Studies , Female , Humans , Nabumetone , Platelet Aggregation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...