Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Infect Dis ; 226(1): 109-118, 2022 08 12.
Article in English | MEDLINE | ID: mdl-32604406

ABSTRACT

BACKGROUND: Both the elderly and individuals with comorbidities are at increased risk of developing influenza-related complications. Novel influenza antivirals are required, given limitations of current drugs (eg, resistance emergence and poor efficacy). Pimodivir is a first-in-class antiviral for influenza A under development for these patients. METHODS: Hospitalized patients with influenza A infection were randomized 2:1 to receive pimodivir 600 mg plus oseltamivir 75 mg or placebo plus oseltamivir 75 mg twice daily for 7 days in this phase 2b study. The primary objective was to compare pimodivir pharmacokinetics in elderly (aged 65-85 years) versus nonelderly adults (aged 18-64 years). Secondary end points included time to patient-reported symptom resolution. RESULTS: Pimodivir pharmacokinetic parameters in nonelderly and elderly patients were similar. Time to influenza symptom resolution was numerically shorter with pimodivir (72.45 hours) than placebo (94.15 hours). There was a lower incidence of influenza-related complications in the pimodivir group (7.9%) versus placebo group (15.6%). Treatment was generally well tolerated. CONCLUSIONS: No apparent relationship was observed between pimodivir pharmacokinetics and age. Our data demonstrate the need for a larger study of pimodivir in addition to oseltamivir to test whether it results in a clinically significant decrease in time-to-influenza-symptom alleviation and/or the frequency of influenza complications. CLINICAL TRIALS REGISTRATION: NCT02532283.


Subject(s)
Influenza, Human , Oseltamivir , Adult , Aged , Humans , Antiviral Agents , Influenza, Human/drug therapy , Oseltamivir/therapeutic use , Pyridines/therapeutic use , Pyrroles/pharmacokinetics , Treatment Outcome , Adolescent , Young Adult , Middle Aged , Aged, 80 and over
2.
J Infect Dis ; 219(7): 1026-1034, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30428049

ABSTRACT

BACKGROUND: Pimodivir, a first-in-class inhibitor of influenza virus polymerase basic protein 2, is being developed for hospitalized and high-risk patients with influenza A. METHODS: In this double-blinded phase 2b study, adults with acute uncomplicated influenza A were randomized 1:1:1:1 to receive one of the following treatments twice daily for 5 days: placebo, pimodivir 300 mg or 600 mg, or pimodivir 600 mg plus oseltamivir 75 mg. Antiviral activity, safety, and pharmacokinetics of pimodivir alone or in combination were evaluated. RESULTS: Of 292 patients randomized, 223 were treated and had confirmed influenza A virus infection. The trial was stopped early because the primary end point was met; the area under the curve of the viral load, determined by quantitative reverse transcription-polymerase chain reaction analysis, in nasal secretions from baseline to day 8 significantly decreased in the active treatment groups, compared with the placebo group (300 mg group, -3.6 day*log10 copies/mL [95% confidence interval {CI}, -7.1 to -0.1]; 600 mg group, -4.5 [95%CI -8.0 to -1.0]; and combination group, -8.6 [95% CI, -12.0 to -5.1]). Pimodivir plus oseltamivir yielded a significantly lower viral load titer over time than placebo and a trend for a shorter time to symptom resolution than placebo. Pimodivir plasma concentrations increased in a dose-proportional manner. The most commonly reported adverse event was mild or moderate diarrhea. CONCLUSIONS: Pimodivir (with or without oseltamivir) resulted in significant virologic improvements over placebo, demonstrated trends in clinical improvement, and was well tolerated. Pimodivir 600 mg twice daily is in further development. CLINICAL TRIALS REGISTRATION: NCT02342249, 2014-004068-39, and CR107745.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A virus , Influenza, Human/drug therapy , Oseltamivir/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Acute Disease , Adult , Antiviral Agents/adverse effects , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Diarrhea/chemically induced , Double-Blind Method , Drug Resistance, Viral/genetics , Drug Therapy, Combination , Early Termination of Clinical Trials , Female , Humans , Influenza A virus/genetics , Influenza A virus/physiology , Influenza, Human/virology , Male , Middle Aged , Oseltamivir/blood , Pyridines/adverse effects , Pyridines/blood , Pyridines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/blood , Pyrroles/pharmacokinetics , Time Factors , Viral Load , Viral Proteins/genetics , Virus Shedding
3.
Int J Clin Pharmacol Ther ; 52(10): 889-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25138682

ABSTRACT

AIM: Vorapaxar is a proteaseactivated receptor (PAR)-1 antagonist being developed for the prevention and treatment of thrombotic vascular events. To evaluate race/ethnic differences between Caucasians and Chinese in the pharmacokinetics of vorapaxar and its active metabolite SCH 2046273 (M20) or in the metabolite/parent ratio, we conducted a cross-study comparison on pharmacokinetic data of vorapaxar and M20 obtained from two similarly designed studies: one in healthy Chinese subjects and the other in a healthy Western (United States, [U.S.]) population. METHODS: The pharmacokinetic profiles of vorapaxar and M20 were characterized using open label, two treatment parallel group designs in men and women aged 18 - 45 years. Vorapaxar was administered orally as a single dose of 40 mg in Chinese subjects (n = 14) or 120 mg in U.S. subjects (n = 14), or 2.5 mg QD for 6 weeks in both studies (Chinese, n = 14; U.S., n = 23). RESULTS: Vorapaxar was rapidly absorbed in both Chinese and U.S. subjects. Vorapaxar and M20 had similar elimination half-lives. The range of metabolite/parent ratios after single dose or daily administration was largely overlapped in Chinese and U.S. subjects. Steady state was attained by day 21 for vorapaxar and M20 in both race/ethnic groups. The accumulation ratios for vorapaxar and M20 during daily administration were similar in Chinese and U.S. subjects. Vorapaxar was well-tolerated in Chinese and U.S. subjects. CONCLUSION: The pharmacokinetic profiles of vorapaxar and M20 and the metabolite/parent ratios in healthy Chinese were generally comparable to those in a healthy Western population.


Subject(s)
Lactones/pharmacokinetics , Pyridines/pharmacokinetics , Receptor, PAR-1/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Area Under Curve , Female , Humans , Lactones/administration & dosage , Male , Middle Aged , Pyridines/administration & dosage
4.
Sci Rep ; 14(1): 17515, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39080319

ABSTRACT

The interleukin (IL)-23 pathway is a pathogenic driver in psoriasis, psoriatic arthritis, and inflammatory bowel disease. Currently, no oral therapeutics selectively target this pathway. JNJ-77242113 is a peptide targeting the IL-23 receptor with high affinity (KD: 7.1 pM). In human cells, JNJ-77242113 potently and selectively inhibited proximal IL-23 signaling (IC50: 5.6 pM) without impacting IL-12 signaling. JNJ-77242113 inhibited IL-23-induced interferon (IFN)γ production in NK cells, and in blood from healthy donors and psoriasis patients (IC50: 18.4, 11 and 9 pM, respectively). In a rat trinitrobenzene sulfonic acid-induced colitis model, oral JNJ-77242113 attenuated disease parameters at doses ≥ 0.3 mg/kg/day. Pharmacologic activity beyond the gastrointestinal tract was also demonstrated. In blood from rats receiving oral JNJ-77242113, dose-dependent inhibition of ex vivo IL-23-stimulated IL-17A production was observed. In an IL-23-induced rat skin inflammation model, JNJ-77242113 inhibited IL-23-induced skin thickening and IL-17A, -17F and -22 gene induction. Oral dosing of JNJ-77242113 in healthy human volunteers inhibited ex vivo IL-23-stimulated IFNγ production in whole blood. Thus, JNJ-77242113 provided selective, systemic IL-23 pathway inhibition in preclinical models which translated to pharmacodynamic activity in healthy human volunteers, supporting the potential for JNJ-77242113 as a selective oral therapy for IL-23-driven immune-mediated diseases.


Subject(s)
Interleukin-23 , Receptors, Interleukin , Animals , Humans , Rats , Interleukin-23/metabolism , Administration, Oral , Receptors, Interleukin/metabolism , Male , Signal Transduction/drug effects , Psoriasis/drug therapy , Psoriasis/chemically induced , Colitis/drug therapy , Colitis/chemically induced , Disease Models, Animal , Peptides/pharmacology , Peptides/administration & dosage , Female , Rats, Sprague-Dawley
5.
Eur J Clin Pharmacol ; 68(11): 1509-16, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22476387

ABSTRACT

PURPOSE: Vorapaxar is an orally active protease-activated receptor 1 (PAR-1) antagonist that inhibits thrombin-induced platelet aggregation. This open-label study assessed the pharmacokinetics and pharmacodynamics of single-dose warfarin in the presence/absence of multiple-dose vorapaxar in 12 healthy men. METHODS: Subjects received two treatments separated by ≥ 7-day washout: Treatment A warfarin 25 mg (Day 1); Treatment B vorapaxar 2.5 mg/day on Days 1-6 and vorapaxar 40 mg coadministered with warfarin 25 mg (Day 7). R-warfarin, S-warfarin, and prothrombin time (PT) were assayed predose and up to 120 h postdose. RESULTS: The geometric mean ratio (GMR) as a percentage (warfarin + vorapaxar/warfarin) was calculated. The GMR (90 % CIs) estimates of C(max) were 105 (99, 111) and 105 (99, 112) for R- and S-warfarin, respectively. The GMR (90 % CIs) estimates of AUC(0-∞) were 108 (101, 116) and 105 (96, 115) for R- and S-warfarin, respectively. The GMR (95 % CIs) estimates of AUC(0-120 h) for PT and INR were 97 (95, 98) and 96 (94, 98), respectively. CONCLUSION: Results of this study indicate that vorapaxar has no meaningful effect on the pharmacokinetics or pharmacodynamics of warfarin, suggesting that the coadministration of these two drugs or vorapaxar coadministered with other CYP2C9/CYP2C19 substrates is unlikely to cause a clinically significant pharmacokinetic drug interaction.


Subject(s)
Anticoagulants/pharmacokinetics , Lactones/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Warfarin/pharmacokinetics , Adolescent , Adult , Anticoagulants/adverse effects , Anticoagulants/blood , Anticoagulants/pharmacology , Biological Availability , Drug Interactions , Drug Monitoring , Half-Life , Humans , International Normalized Ratio , Lactones/adverse effects , Male , Middle Aged , Platelet Aggregation Inhibitors/adverse effects , Prothrombin Time , Pyridines/adverse effects , Statistics as Topic , Stereoisomerism , Warfarin/adverse effects , Warfarin/blood , Warfarin/pharmacology , Young Adult
6.
Eur J Clin Pharmacol ; 68(11): 1501-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22527342

ABSTRACT

PURPOSE: To determine whether hepatic impairment has an effect on the pharmacokinetics (PK) of vorapaxar or M20, its main pharmacologically active metabolite. METHODS: This was an open-label study in which a single 40-mg oral dose of vorapaxar was administered to patients with mild (n = 6), moderate (n = 6), and severe (n = 4) hepatic impairment and healthy controls (n = 16) matched for age, gender, weight, and height. Blood samples for vorapaxar and M20 assay were collected predose and at frequent intervals up to 8 weeks postdose. RESULTS: Plasma vorapaxar and M20 PK profiles were similar between patients with impaired liver function and healthy controls. Group mean values for vorapaxar C(max) and AUC(tf) were 206-279 ng/mL and 14,200-18,200 ng·h/mL, respectively, with the lowest values observed in patients with severe impairment. Vorapaxar median T(max) and mean t(1/2) values were 1.00-1.75 h and 298-366 h, respectively. There was no apparent correlation between vorapaxar or M20 exposure or t(1/2) values and disease severity. Vorapaxar was generally well tolerated; one serious adverse event (gastrointestinal bleeding secondary to ruptured esophageal varices) was reported in a patient with severe hepatic impairment. CONCLUSIONS: Hepatic impairment had no clinically relevant effect on the PK of vorapaxar and M20. No dose or dosage adjustment of vorapaxar will be required in patients with mild to moderate hepatic impairment. Although systemic exposure to vorapaxar does not appear to increase in patients with severe hepatic impairment, administration of vorapaxar to such patients is not recommended given their bleeding diathesis.


Subject(s)
Hepatic Insufficiency/metabolism , Lactones/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Pyridines/pharmacokinetics , Receptor, PAR-1/antagonists & inhibitors , Receptors, Thrombin/antagonists & inhibitors , Aged , Biotransformation , Female , Gastrointestinal Hemorrhage/chemically induced , Half-Life , Hepatic Insufficiency/blood , Hepatic Insufficiency/physiopathology , Humans , Intestinal Absorption , Lactones/adverse effects , Lactones/blood , Liver Failure/blood , Liver Failure/metabolism , Liver Failure/physiopathology , Male , Middle Aged , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/blood , Pyridines/adverse effects , Pyridines/blood , Severity of Illness Index
7.
Eur J Clin Pharmacol ; 68(3): 249-58, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21935705

ABSTRACT

PURPOSE: The aim of our study was to evaluate the pharmacology of vorapaxar (SCH 530348), an oral PAR-1 antagonist, in healthy volunteers. METHODS AND RESULTS: In two randomized, placebo-controlled studies, subjects received either single ascending doses of vorapaxar (0.25, 1, 5, 10, 20, or 40 mg; n = 50), multiple ascending doses of vorapaxar (1, 3, or 5 mg/day for 28 days; n = 36), a loading dose (10 or 20 mg) followed by daily maintenance doses (1 mg) for 6 days (n = 12), or placebo. Single 20- and 40-mg doses of vorapaxar completely inhibited thrombin receptor activating peptide (TRAP)-induced platelet aggregation (>80% inhibition) at 1 h and sustained this level of inhibition for ≥72 h. Multiple doses yielded complete inhibition on Day 1 (5 mg/day) and Day 7 (1 and 3 mg/day). Adverse events were generally mild, transient, and unrelated to dose. CONCLUSION: Vorapaxar provided rapid and sustained dose-related inhibition of platelet aggregation without affecting bleeding or clotting times.


Subject(s)
Lactones/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Pyridines/administration & dosage , Receptor, PAR-1/antagonists & inhibitors , Adolescent , Adult , Bleeding Time , Female , Humans , Lactones/blood , Lactones/pharmacokinetics , Male , Middle Aged , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , Young Adult
8.
Eur J Clin Pharmacol ; 68(7): 1049-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22315147

ABSTRACT

PURPOSE: To determine whether impaired renal function alters the pharmacokinetics (PK) of vorapaxar or its ability to inhibit thrombin receptor agonist peptide (TRAP)-induced platelet aggregation. METHODS: This was an open-label study in which 8 patients with end-stage renal disease (ESRD) on hemodialysis and 7 matched (based on age, gender, weight, and height) healthy controls were administered a single 10-mg oral dose of vorapaxar. Blood samples for vorapaxar PK and pharmacodynamic analysis were collected predose and at frequent intervals up to 6 weeks postdose. RESULTS: Mean vorapaxar bioavailability (based on area under the curve of plasma vorapaxar concentration over time) was identical in the two subject groups; the ESRD/healthy geometric mean ratio (GMR, expressed in percent) was 98. Mean maximum observed plasma concentration (77.4-98.2 ng/mL) was numerically lower in patients with ESRD compared with matched controls (GMR=76; 90% confidence interval=48 to 118). Median time of maximum observed plasma concentration was 2 h in both subject groups. The observed means for elimination half-life were 186 and 231 h in the ESRD and control groups, respectively. Inhibition of platelet aggregation was similar in the two groups. Four out of 15 (27%) subjects reported adverse events, all of which were characterized by the investigator as mild and unrelated to treatment. CONCLUSIONS: ESRD had no clinically relevant effect on the PK profile of vorapaxar or its ability to inhibit TRAP-induced platelet aggregation.


Subject(s)
Kidney Failure, Chronic/drug therapy , Lactones/pharmacology , Lactones/pharmacokinetics , Platelet Aggregation/drug effects , Pyridines/pharmacology , Pyridines/pharmacokinetics , Receptor, PAR-1/antagonists & inhibitors , Receptors, Thrombin/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Biological Availability , Data Interpretation, Statistical , Female , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/metabolism , Kidney Function Tests , Lactones/blood , Lactones/therapeutic use , Male , Middle Aged , Pyridines/blood , Pyridines/therapeutic use , Renal Dialysis , Young Adult
9.
Eur J Clin Pharmacol ; 68(3): 291-300, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21969227

ABSTRACT

BACKGROUND: Vorapaxar, a novel antiplatelet agent in advanced clinical development for the prevention and treatment of atherothrombotic disease, is a potent, orally bioavailable thrombin receptor antagonist selective for the protease-activated receptor 1 (PAR-1). METHODS: Since race/ethnicity may affect the safety, efficacy and dosage of drugs, this study was conducted to evaluate potential differences in the pharmacodynamics, pharmacokinetics and safety of vorapaxar after single (5, 10, 20, or 40 mg) or multiple (0.5, 1, or 2.5 mg once daily) doses in healthy Japanese and matched (gender, age, height, and weight) Caucasian volunteers. RESULTS: Vorapaxar was well tolerated in both Japanese and Caucasian subjects. Pharmacodynamic and pharmacokinetic profiles of vorapaxar in the two racial/ethnic groups were similar. In both racial groups, complete inhibition of platelet aggregation was achieved most rapidly with vorapaxar 40 mg and was consistently achieved and maintained with a 2.5 mg daily maintenance dose. CONCLUSION: There were no substantial differences in the safety, pharmacokinetics or pharmacodynamics of vorapaxar between Japanese and Caucasian subjects.


Subject(s)
Lactones/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Adolescent , Adult , Asian People , Female , Humans , Lactones/blood , Lactones/pharmacokinetics , Male , Middle Aged , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Pyridines/blood , Pyridines/pharmacokinetics , White People , Young Adult
10.
Eur J Clin Pharmacol ; 67(5): 483-92, 2011 May.
Article in English | MEDLINE | ID: mdl-21120461

ABSTRACT

BACKGROUND: Efforts to lower plasma lipid levels sometimes require multiple agents with different mechanisms of action to achieve results specified by national treatment guidelines. METHODS: This was an open-label, randomized, three-period, multiple-dose crossover study that assessed the potential for pharmacokinetic interaction between extended-release niacin and ezetimibe/simvastatin and their major metabolites. Eighteen adults received three randomized treatments: (A) extended-release (ER) niacin 1000 mg/day for 2 days, followed by 2000 mg/day for 5 days; (B) ezetimibe/simvastatin 10 mg/20 mg/day; (C) coadministration of Treatments A and B. Treatments were given once a day after a low fat breakfast for a total of 7 days, with a 7-day inter-dose period. RESULTS: There were small (mean ≤35%) increases in drug exposure for all analytes after coadministration of ER niacin and ezetimibe/simvastatin 10 mg/20 mg. The least-square mean between treatment C(max) (maximum plasma concentration) ratios (×100) were 97, 98, and 109% for ezetimibe, simvastatin and niacin, respectively. The corresponding ratios for total ezetimibe, simvastatin acid, and nicotinuric acid were 99, 118, and 110%. The AUC((0-24)) (area under the plasma concentration-time curve from time zero to 24 h after dosing) ratios for ezetimibe, simvastatin, and niacin were 109, 120, and 122%, respectively, and the corresponding ratios for total ezetimibe, simvastatin acid, and nicotinuric acid were 126, 135 and 119%. CONCLUSION: There is a small pharmacokinetic drug interaction between ER niacin and ezetimibe/simvastatin and although this is not considered to be clinically significant, the concomitant use of these drugs should be appropriately monitored, especially during the niacin titration period.


Subject(s)
Azetidines/pharmacokinetics , Niacin/pharmacokinetics , Simvastatin/pharmacokinetics , Adult , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/pharmacokinetics , Azetidines/adverse effects , Cross-Over Studies , Delayed-Action Preparations , Drug Interactions , Drug Therapy, Combination , Ezetimibe , Female , Humans , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/pharmacokinetics , Lipids/blood , Male , Niacin/adverse effects , Nicotinic Acids/pharmacokinetics , Simvastatin/adverse effects , Simvastatin/analogs & derivatives , Tablets/adverse effects , Tablets/pharmacokinetics
11.
Clin Pharmacol Drug Dev ; 7(2): 143-150, 2018 02.
Article in English | MEDLINE | ID: mdl-28403576

ABSTRACT

Vorapaxar is a first-in-class antagonist of the protease-activated receptor-1, the primary thrombin receptor on human platelets, which mediates the downstream effects of thrombin in hemostasis and thrombosis. Prasugrel is a platelet inhibitor that acts as a P2Y12 receptor antagonist through an active metabolite, R-138727. This study investigated the interaction of these 2 platelet antagonists when coadministered. This was a randomized, open-label, multiple-dose study in 54 healthy volunteers consisting of a fixed-sequence crossover and a parallel group design. In sequence 1, 36 subjects received prasugrel 60 mg on day 1 and then prasugrel 10 mg once daily on days 2 to 7, followed by vorapaxar 40 mg and prasugrel 10 mg on day 8 and then vorapaxar 2.5 mg and prasugrel 10 mg orally once daily on days 9 to 28. In sequence 2, 18 subjects received vorapaxar 40 mg on day 1 and then vorapaxar 2.5 mg once daily on days 2 to 21. The geometric mean ratios (90% confidence intervals) for AUCτ and Cmax of coadministration/monotherapy for vorapaxar (0.93 ng·h/mL[0.85-1.02 ng·h/mL] and 0.95 ng/mL [0.86-1.05 ng/mL]) and R-138727 (0.91 ng·h/mL [0.85- 0.99 ng·h/mL] and 1.02 ng/mL [0.89-1.17 ng/mL]) were within prespecified bounds, demonstrating the absence of a pharmacokinetic interaction between vorapaxar and prasugrel. There was no specific safety or tolerability risk associated with multiple-dose coadministration of vorapaxar and prasugrel. In conclusion, in this study in healthy volunteers, there was no pharmacokinetic drug-drug interaction between vorapaxar and prasugrel. Multiple-dose coadministration of the 2 drugs was generally well tolerated.


Subject(s)
Lactones/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Pyridines/pharmacokinetics , Adult , Cross-Over Studies , Drug Administration Schedule , Drug Interactions , Female , Healthy Volunteers , Humans , Lactones/administration & dosage , Lactones/adverse effects , Lactones/blood , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/blood , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/blood , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/blood , Young Adult
12.
Drug Saf ; 30(4): 317-26, 2007.
Article in English | MEDLINE | ID: mdl-17408308

ABSTRACT

The development of corticosteroids that are delivered directly to the nasal mucosa has alleviated much of the concern about the systemic adverse effects associated with oral corticosteroid therapy. However, given the high potency of these drugs and their widespread use in the treatment of allergic rhinitis, it is important to ensure that intranasal corticosteroids have a favourable benefit-risk ratio. One agent that typifies the systemic safety found in the majority of intranasal corticosteroids is mometasone furoate nasal spray, a potent and effective treatment for seasonal and perennial allergic rhinitis and nasal polyposis. Mometasone furoate does not reach high systemic concentrations or cause clinically significant adverse effects. Results from pharmacokinetic studies in adults and children suggest that systemic exposure to mometasone furoate after intranasal administration is negligible. This is probably because of the inherently low aqueous solubility of mometasone furoate, which allows only a small fraction of the drug to cross the nasal mucosa and enter the bloodstream, and because a large amount of the administered drug is swallowed and undergoes extensive first-pass metabolism. There is no clinical evidence that mometasone furoate nasal spray suppresses the function of the hypothalamus-pituitary-adrenal axis when the drug is administered at clinically relevant doses (100-200 microg/day); consequently, mometasone furoate nasal spray has not been associated with growth inhibition in children. The safety and tolerability of mometasone furoate nasal spray have been rigorously assessed in clinical trials involving approximately 4,500 patients, with epistaxis, headache and pharyngitis being the most common adverse effects associated with treatment in adolescents and adults. The clinical effectiveness of mometasone furoate nasal spray, coupled with its agreeable safety and tolerability profile, confirms its favourable benefit-risk ratio.


Subject(s)
Pregnadienediols/therapeutic use , Rhinitis, Allergic, Seasonal/drug therapy , Administration, Intranasal , Anti-Allergic Agents/adverse effects , Anti-Allergic Agents/metabolism , Anti-Allergic Agents/therapeutic use , Humans , Mometasone Furoate , Nebulizers and Vaporizers , Pregnadienediols/adverse effects , Pregnadienediols/metabolism , Rhinitis, Allergic, Perennial/drug therapy , Risk Assessment , Risk Factors , Treatment Outcome
13.
J Clin Pharmacol ; 46(3): 321-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16490808

ABSTRACT

This single-center, open-label, 2-period crossover study investigated the effects of multiple-dose ezetimibe (EZE) on a single dose of cyclosporine (CyA). Healthy subjects received 2 treatments in random order with a 14-day washout: (1) CyA 100 mg alone and (2) EZE 20 mg for 7 days with CyA 100 mg coadministered on day 7; EZE 20 mg alone was administered on day 8. AUC(0-last) and Cmax geometric mean ratios (90% confidence interval) for ([CyA + EZE]/CyA alone) were 1.15 (1.07, 1.25) and 1.10 (0.97, 1.26), respectively. Tmax (approximately 1.3 hours) was similar with and without EZE (P >.200). Mean CyA exposure slightly increased (approximately 15%) with multiple-dose EZE 20 mg; however, this value was contained within (0.80, 1.25). The implications for chronic EZE dosing within the usual clinical paradigm of chronic CyA dosing have not been established; caution is recommended when using these agents concomitantly. CyA concentrations should be monitored in patients receiving EZE and CyA.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Adult , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Interactions , Drug Monitoring , Ezetimibe , Female , Humans , Male
14.
J Clin Pharmacol ; 46(3): 328-36, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16490809

ABSTRACT

This open-label, single-period study evaluated the single-dose pharmacokinetics of ezetimibe (EZE) 10 mg in the setting of steady-state cyclosporine (CyA) dosing in renal transplant patients. A single 10-mg dose of EZE was coadministered with the morning dose of CyA (75-150 mg twice a day). Total EZE (sum of unconjugated, parent EZE and EZE-glucuronide; EZE-total) AUC(0-last) and Cmax were compared to values derived from a prespecified database of healthy volunteers. Geometric mean ratios (90% CIs) for (EZE + CyA)/EZE alone for EZE-total AUC((0-last)) and Cmax were 3.41 (2.55, 4.56) and 3.91 (3.13, 4.89), respectively. Compared to healthy controls, EZE-total AUC((0-last)) was 3.4-fold higher in transplant patients receiving CyA; similar exposure levels were seen in a prior multiple-dose study in which EZE 50 mg was administered to healthy volunteers without dose-related toxicity. Because the long-term safety implications of both higher EZE exposures and undetermined effect on CyA are not yet understood, the clinical significance of this interaction is unknown.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Kidney Transplantation , Adult , Aged , Anticholesteremic Agents/blood , Area Under Curve , Azetidines/blood , Dose-Response Relationship, Drug , Drug Interactions , Drug Monitoring , Ezetimibe , Female , Humans , Male , Middle Aged
15.
Clin Pharmacokinet ; 44(5): 467-94, 2005.
Article in English | MEDLINE | ID: mdl-15871634

ABSTRACT

Ezetimibe is the first lipid-lowering drug that inhibits intestinal uptake of dietary and biliary cholesterol without affecting the absorption of fat-soluble nutrients. Following oral administration, ezetimibe is rapidly absorbed and extensively metabolised (>80%) to the pharmacologically active ezetimibe-glucuronide. Total ezetimibe (sum of 'parent' ezetimibe plus ezetimibe-glucuronide) concentrations reach a maximum 1-2 hours post-administration, followed by enterohepatic recycling and slow elimination. The estimated terminal half-life of ezetimibe and ezetimibe-glucuronide is approximately 22 hours. Consistent with the elimination half-life of ezetimibe, an approximate 2-fold accumulation is observed upon repeated once-daily administration. The recommended dose of ezetimibe 10 mg/day can be administered in the morning or evening without regard to food. There are no clinically significant effects of age, sex or race on ezetimibe pharmacokinetics and no dosage adjustment is necessary in patients with mild hepatic impairment or mild-to-severe renal insufficiency. The major metabolic pathway for ezetimibe consists of glucuronidation of the 4-hydroxyphenyl group by uridine 5'-diphosphate-glucuronosyltransferase isoenzymes to form ezetimibe-glucuronide in the intestine and liver. Approximately 78% of the dose is excreted in the faeces predominantly as ezetimibe, with the balance found in the urine mainly as ezetimibe-glucuronide. Overall, ezetimibe has a favourable drug-drug interaction profile, as evidenced by the lack of clinically relevant interactions between ezetimibe and a variety of drugs commonly used in patients with hypercholesterolaemia. Ezetimibe does not have significant effects on plasma levels of HMG-CoA reductase inhibitors commonly known as statins (atorvastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin), fibric acid derivatives (gemfibrozil, fenofibrate), digoxin, glipizide, warfarin and triphasic oral contraceptives (ethinylestradiol and levonorgestrel). Concomitant administration of food, antacids, cimetidine or statins had no significant effect on ezetimibe bioavailability. Although coadministration with gemfibrozil and fenofibrate increased the bioavailability of ezetimibe, the clinical significance is thought to be minor considering the relatively flat dose-response curve of ezetimibe and the lack of dose-related increase in adverse events. In contrast, coadministration with the bile acid binding agent colestyramine significantly decreased ezetimibe oral bioavailability (based on area under the plasma concentration-time curve of total ezetimibe). Hence, ezetimibe and colestyramine should be administered several hours apart to avoid attenuating the efficacy of ezetimibe. Finally, higher ezetimibe exposures were observed in patients receiving concomitant ciclosporin, and ezetimibe caused a small but statistically significant effect on plasma levels of ciclosporin. Because treatment experience in patients receiving ciclosporin is limited, physicians are advised to exercise caution when initiating ezetimibe in the setting of ciclosporin coadministration, and to carefully monitor ciclosporin levels.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Animals , Azetidines/pharmacology , Biological Availability , Cholestyramine Resin/pharmacology , Contraceptives, Oral/pharmacology , Drug Interactions , Ezetimibe , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Warfarin/pharmacology
16.
Curr Med Res Opin ; 21(8): 1171-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16083526

ABSTRACT

OBJECTIVE: The objective of this study was to evaluate the pharmacodynamic effects and safety of the co-administration of ezetimibe and fluvastatin in healthy hypercholesterolemic subjects at clinically-relevant doses and to evaluate the potential for a pharmacokinetic drug interaction between ezetimibe and fluvastatin. METHODS: In a single-center, evaluator-blind, placebo-controlled, multiple-dose, parallel-group study 32 healthy subjects with hypercholesterolemia were randomized to 4 treatments administered once daily for 14 days: ezetimibe 10 mg plus ezetimibe placebo, fluvastatin 20 mg plus ezetimibe placebo, fluvastatin 20 mg plus ezetimibe 10 mg, and ezetimibe placebo. Blood samples were collected to measure serum lipids and to determine steady-state pharmacokinetics. RESULTS: Ezetimibe 10 mg significantly (p < or = 0.01) decreased total-cholesterol and low-density lipoprotein cholesterol (LDL-C) concentrations compared to placebo at Day 14. Fluvastatin 20 mg also caused a significant (p = 0.01) reduction in total-cholesterol and a decrease in LDL-C at Day 14 compared to placebo, however, the decrease in LDL-C did not reach statistical significance (p = 0.08). The coadministration of ezetimibe 10 mg and fluvastatin 20 mg caused significantly (p < or = 0.01) greater mean percent reductions in LDL-C and total-cholesterol than fluvastatin 20 mg alone or placebo at Day 14. Fluvastatin had no clinically significant effect on the pharmacokinetics of ezetimibe. On average, ezetimibe appeared to decrease the rate and extent of fluvastatin bioavailability. CONCLUSION: Coadministration of ezetimibe and fluvastatin was safe and well tolerated and caused significant incremental reductions in LDL-C and total cholesterol compared to fluvastatin administered alone. The pharmacokinetics of ezetimibe were not affected by coadministration with fluvastatin. The apparent decrease in fluvastatin exposure on administration with ezetimibe was likely to be due to the parallel study design and two pharmacokinetic outliers and is considered of no clinical significance.


Subject(s)
Anticholesteremic Agents/therapeutic use , Azetidines/pharmacokinetics , Fatty Acids, Monounsaturated/pharmacokinetics , Hypercholesterolemia/drug therapy , Indoles/pharmacokinetics , Adult , Anticholesteremic Agents/pharmacokinetics , Azetidines/therapeutic use , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Drug Synergism , Drug Therapy, Combination , Ezetimibe , Fatty Acids, Monounsaturated/therapeutic use , Female , Fluvastatin , Humans , Indoles/therapeutic use , Male , Middle Aged
17.
Clin Pharmacol Drug Dev ; 4(1): 56-62, 2015 01.
Article in English | MEDLINE | ID: mdl-27128003

ABSTRACT

PURPOSE: To evaluate the potential effects of vorapaxar on the pharmacokinetics and safety of rosiglitazone. METHODS: This was an open-label, two-period, two-treatment, fixed-sequence study in 18 healthy subjects. On Day 1, Period 1, subjects received a single dose of rosiglitazone 8 mg. In Period 2, subjects received vorapaxar 40 mg on Day 1, vorapaxar 7.5 mg once-daily on Days 2-7, and a single dose of rosiglitazone 8 mg on Day 7. Rosiglitazone and N-desmethylrosiglitazone pharmacokinetics were assessed alone (Period 1) and after coadministration with vorapaxar (Period 2). Vorapaxar and its M20 metabolite pharmacokinetics were assessed on Day 7, Period 2. Safety and tolerability were assessed throughout the study. RESULTS: Coadministration of rosiglitazone with vorapaxar had no effect on rosiglitazone or N-desmethylrosiglitazone pharmacokinetics. The ratio of geometric means (GMR) and 90% confidence intervals (CI) of the coadministration versus monotherapy for Cmax (GMR 95; 90% CI 88, 103) and AUC0-24 h (GMR 103; 90% CI 98, 108) were within the 80-125% bioequivalence criteria. The metabolite-to-parent exposure ratio with and without vorapaxar was unaltered. Coadministration of vorapaxar with rosiglitazone was generally well tolerated. CONCLUSION: Coadministration of vorapaxar with rosiglitazone or drugs metabolized via CYP2C8 is unlikely to cause a significant pharmacokinetic interaction.


Subject(s)
Cytochrome P-450 CYP2C8 Inhibitors/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Lactones/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Pyridines/administration & dosage , Receptor, PAR-1/antagonists & inhibitors , Thiazolidinediones/pharmacokinetics , Adolescent , Adult , Area Under Curve , Biotransformation , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C8 Inhibitors/adverse effects , Drug Interactions , Female , Half-Life , Healthy Volunteers , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/blood , Lactones/adverse effects , Male , Metabolic Clearance Rate , Middle Aged , North Dakota , Platelet Aggregation Inhibitors/adverse effects , Pyridines/adverse effects , Receptor, PAR-1/metabolism , Rosiglitazone , Therapeutic Equivalency , Thiazolidinediones/administration & dosage , Thiazolidinediones/adverse effects , Thiazolidinediones/blood , Young Adult
18.
Curr Med Res Opin ; 20(9): 1493-500, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15383199

ABSTRACT

BACKGROUND: Ezetimibe (Zetia) is a novel inhibitor of intestinal absorption of cholesterol that is approved for the treatment of primary hypercholesterolemia. In a separate pilot study, co-administration of ezetimibe and lovastatin resulted in a significant pharmacodynamic interaction, leading to an additive reduction in LDL-C. The current study was designed to further investigate the potential for pharmacokinetic interaction between ezetimibe and lovastatin. METHODS: This was a randomized, open-label, 3-way crossover study in 18 healthy adult volunteers. All subjects received the following treatments orally once daily for 7 days: ezetimibe 10 mg, lovastatin 20 mg, or ezetimibe 10 mg plus lovastatin 20 mg. Plasma samples obtained on day 7 were evaluated for steady-state pharmacokinetics of ezetimibe (unconjugated), total ezetimibe (ezetimibe and ezetimibe-glucuronide conjugate), lovastatin, and beta-hydroxylovastatin. RESULTS: Co-administration of ezetimibe with lovastatin did not affect the pharmacokinetics of ezetimibe. There were no significant differences in the exposure to total ezetimibe, ezetimibe-glucuronide and ezetimibe after co-administration with lovastatin vs. ezetimibe given alone. Co-administration of ezetimibe with lovastatin had no significant effect on the exposure to either lovastatin or beta-hydroxylovastatin. The point estimates based on the log-transformed Cmax and AUC values for lovastatin and beta-hydroxylovastatin were 113% and 119%, respectively, for co-administration of ezetimibe with lovastatin vs. lovastatin administration alone. Co-administration therapy with ezetimibe and lovastatin was safe and well tolerated. CONCLUSIONS: Ezetimibe did not significantly affect the pharmacokinetics of lovastatin or beta-hydroxylovastatin and vice versa. Co-administration of ezetimibe and lovastatin is unlikely to cause a clinically significant pharmacokinetic drug interaction.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Lovastatin/pharmacokinetics , Adult , Anticholesteremic Agents/administration & dosage , Azetidines/administration & dosage , Cross-Over Studies , Drug Interactions , Drug Therapy, Combination , Ezetimibe , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Lovastatin/administration & dosage , Male , Reference Values
19.
Curr Med Res Opin ; 20(6): 955-65, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15200755

ABSTRACT

BACKGROUND: Ezetimibe is a cholesterol absorption inhibitor which decreases low-density lipoprotein cholesterol (LDL-C) in patients with hypercholesterolemia. This study investigated the potential for pharmacodynamic and/or pharmacokinetic interactions between ezetimibe and lovastatin. METHODS: In a randomized, evaluator (single)-blind, placebo-controlled, parallel-group study, 48 healthy men with hypercholesterolemia (screening LDL-C >or= 130 mg/dL) who were stabilized and maintained on a National Cholesterol Education Program (NCEP) Step I diet were randomized to one of the following six oral treatments once daily for 14 days: lovastatin 20 mg; lovastatin 20 mg plus ezetimibe 5, 10, or 20 mg; lovastatin 40 mg plus ezetimibe 10mg; or placebo. RESULTS: Reported adverse events were generally mild, nonspecific, and similar among treatments. There were no significant changes in safety laboratory test results, including those for enzymes indicative of muscle or liver injury. Coadministration of ezetimibe and lovastatin did not increase the plasma concentrations of lovastatin or beta-hydroxylovastatin. In this parallel comparison study there was an apparent decrease in lovastatin exposure, however, the reduction in lovastatin or beta-hydroxylovastatin concentrations was not related to the ezetimibe dose and is not considered to be clinically important. Ezetimibe 5, 10, or 20 mg combined with lovastatin 20 mg caused a significantly (p < 0.01) greater reduction in LDL-C than lovastatin 20 mg alone, with no apparent effect on HDL-C or triglycerides. LDL-C was reduced by 51.0% with ezetimibe 10 mg plus lovastatin 20 mg, 56.0% with ezetimibe 10 mg plus lovastatin 40 mg, 33.2% with lovastatin alone, and 17.3% with placebo. CONCLUSIONS: The co-administration of ezetimibe and lovastatin was well tolerated and resulted in a significantly greater percentage reduction in serum LDL-C concentrations than with lovastatin alone, with an average incremental reduction of 16-18%. Ezetimibe 10mg appears to be the optimal dose when co-administered with lovastatin 20mg once daily. Further incremental reductions in LDL-C from the co-administration of ezetimibe and lovastatin are expected only when the dose of lovastatin is increased. The co-administration of ezetimibe and lovastatin has the potential to produce clinically significant reductions in LDL-C compared to either drug alone, with favorable safety and tolerability.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacology , Azetidines/pharmacokinetics , Lovastatin/pharmacology , Lovastatin/pharmacokinetics , Adult , Anticholesteremic Agents/administration & dosage , Azetidines/administration & dosage , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Ezetimibe , Humans , Lovastatin/administration & dosage , Male , Placebos , Reference Values , Single-Blind Method , United States
20.
Curr Med Res Opin ; 20(8): 1185-95, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15324521

ABSTRACT

BACKGROUND: Ezetimibe is a lipid-lowering drug indicated for the treatment of hypercholesterolemia as co-administration with HMG-CoA reductase inhibitors (statins) or as monotherapy. The primary objectives of this study were to evaluate the pharmacodynamic effects and safety of the co-administration of ezetimibe and the new statin rosuvastatin. A secondary objective was to examine the potential for a pharmacokinetic interaction between ezetimibe and rosuvastatin. METHODS: This was a randomized, evaluator (single)-blind, placebo-controlled, parallel-group study in healthy hypercholesterolemic subjects (untreated low-density lipoprotein cholesterol [LDL-C] > or = 130 mg/dL [3.37 mmol/L]). After the outpatient screening and NCEP Step I diet stabilization periods, 40 subjects were randomized to one of the 4 following treatments: rosuvastatin 10 mg plus ezetimibe 10 mg (n = 12); rosuvastatin 10 mg plus placebo (matching ezetimibe 10 mg) (n = 12); ezetimibe 10 mg plus placebo (matching ezetimibe 10 mg) (n = 8); or placebo (2 tablets, matching ezetimibe 10 mg) (n = 8). All study treatments were administered once daily in the morning for 14 days as part of a 16-day inpatient confinement period. Fasting serum lipids were assessed pre-dose on days 1 (baseline), 7, and 14 by direct quantitative assay methods. Safety was evaluated by monitoring laboratory tests and recording adverse events. Blood samples were collected for ezetimibe and rosuvastatin pharmacokinetic evaluation prior to the first and last dose and at frequent intervals after the last dose (day 14) of study treatment. Plasma ezetimibe, total ezetimibe (ezetimibe plus ezetimibe-glucuronide) and rosuvastatin concentrations were determined by validated liquid chromatography with tandem mass spectrometric detection (LC-MS/MS) assay methods. RESULTS: All active treatments caused statistically significant (p < or = 0.02) decreases in LDL-C concentration versus placebo from baseline to day 14. The co-administration of ezetimibe and rosuvastatin caused a significantly (p < 0.01) greater reduction in LDL-C and total cholesterol than either drug alone. In this 2-week inpatient study with restricted physical activity there was no apparent effect of any treatment on high-density lipoprotein cholesterol (HDL-C) or triglycerides. The co-administration of ezetimibe and rosuvastatin caused a significantly (p < 0.01) greater percentage reduction in mean LDL-C (-61.4%) than rosuvastatin alone (-44.9%), with a mean incremental reduction of -16.4% (95%CI -26.3 to -6.53). Reported side effects were generally mild, nonspecific, and similar among treatment groups. There were no significant increases or changes in clinical laboratory tests, particularly those assessing muscle and liver function. There was no significant pharmacokinetic drug interaction between ezetimibe and rosuvastatin. CONCLUSIONS: Co-administration of ezetimibe 10 mg with rosuvastatin 10 mg daily caused a significant incremental reduction in LDL-C compared with rosuvastatin alone. Moreover, co-administering ezetimibe and rosuvastatin was well tolerated in patients with hypercholesterolemia.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Fluorobenzenes/pharmacokinetics , Hypercholesterolemia/drug therapy , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Anticholesteremic Agents/therapeutic use , Azetidines/therapeutic use , Drug Synergism , Drug Therapy, Combination , Ezetimibe , Female , Fluorobenzenes/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Male , Middle Aged , Pilot Projects , Pyrimidines/therapeutic use , Rosuvastatin Calcium , Single-Blind Method , Sulfonamides/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL