Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Clin Pharmacol Ther ; 115(5): 965-970, 2024 May.
Article in English | MEDLINE | ID: mdl-38251824

ABSTRACT

To further our understanding of the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) E17 guideline and promote effective implementation, a public workshop was held in Japan by regulatory agency and industry representatives. In this workshop, important concepts explained in the ICH E17 guideline, such as intrinsic/extrinsic ethnic factors that influence treatment effects ("effect modifiers") and the holistic evaluation of consistency of treatment effect were actively discussed through case studies. The importance of holistic evaluation of consistency was recognized, and it was concluded that the evaluation and relevant discussion should be shared with regulatory authorities, sponsors, and broader stakeholders.


Subject(s)
Government Agencies , Research Report , Humans , Japan
2.
J Pers Med ; 12(5)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35629092

ABSTRACT

Advances in biotechnology have enabled us to assay human tissue and cells to a depth and resolution that was never possible before, redefining what we know as the "biomarker", and how we define a "disease". This comes along with the shift of focus from a "one-drug-fits-all" to a "personalized approach", placing the drug development industry in a highly dynamic landscape, having to navigate such disruptive trends. In response to this, innovative clinical trial designs have been key in realizing biomarker-driven drug development. Regulatory approvals of cancer genome sequencing panels and associated targeted therapies has brought personalized medicines to the clinic. Increasing availability of sophisticated biotechnologies such as next-generation sequencing (NGS) has also led to a massive outflux of real-world genomic data. This review summarizes the current state of biomarker-driven drug development and highlights examples showing the utility and importance of the application of real-world data in the process. We also propose that all stakeholders in drug development should (1) be conscious of and efficiently utilize real-world evidence and (2) re-vamp the way the industry approaches drug development in this era of personalized medicines.

3.
J Pers Med ; 12(3)2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35330444

ABSTRACT

In recent years, with the advancement of next-generation sequencing (NGS) technology, gene panel tests have been approved in the field of cancer diseases, and approaches to prescribe optimal molecular target drugs to patients are being developed. In the field of rare diseases, whole-genome and whole-exome analysis has been used to identify the causative genes of undiagnosed diseases and to diagnose patients' diseases, and further progress in personalized medicine is expected. In order to promote personalized medicine in the future, we investigated the current status and progress of personalized medicine in disease areas other than cancer and rare diseases, where personalized medicine is most advanced. We selected rheumatoid arthritis and psoriasis as the inflammatory disease, in addition to Alzheimer's disease. These diseases have high unmet needs for personalized medicine from the viewpoints of disease mechanisms, diagnostic biomarkers, therapeutic drugs with diagnostic markers and treatment satisfaction. In rheumatoid arthritis and psoriasis, there are many therapeutic options; however, diagnostic methods have not been developed to select the best treatment for each patient. In addition, there are few effective therapeutic agents in Alzheimer's disease, although clinical trials of many candidate drugs have been conducted. In rheumatoid arthritis and psoriasis, further elucidation of the disease mechanism is desired to enable the selection of appropriate therapeutic agents according to the patient profile. In the case of Alzheimer's disease, progress in preventive medicine is desired through the establishment of an early diagnosis method as well as the research and development of innovative therapeutic agents. To this end, we hope for further research and development of diagnostic markers and new drugs through progress in comprehensive data analysis such as comprehensive genomic and transcriptomic information. Furthermore, new types of markers such as miRNAs and the gut microbiome are desired to be utilized in clinical diagnostics.

4.
Drug Metab Pharmacokinet ; 41: 100422, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34717268

ABSTRACT

Rilpivirine is a non-nucleoside reverse transcriptase inhibitor, used for the treatment of human immunodeficiency virus type-1 infection. An open label study was conducted to investigate the pharmacokinetics (PK) and safety of a single oral dose of rilpivirine 25 mg in Japanese healthy adult subjects. No adverse events were reported. The mean Cmax (144.3 ng/mL) and AUCinf (4542 ng h/mL) in Japanese subjects were approximately 30 % higher than those reported from a similar study in Caucasian healthy subjects, whereas the median tmax and mean t1/2 values were comparable between studies. A simple physiologically based PK model was developed to characterize the rilpivirine PK profile. The model adequately described rilpivirine PK profiles, and well-predicted drug-drug interactions. With exploration using the model, body size and CYP3A4 abundance were identified as factors which explained the observed inter-ethnic difference in rilpivirine exposure. The inter-ethnic difference in rilpivirine exposure was however considered not clinically relevant, since inter-individual variabilities of those intrinsic factors are larger than inter-ethnic ones; and the observed AUCinf in Japanese subjects was within the range of AUCtau associated with efficacy and safety in Phase 3 studies. This study results support the use of rilpivirine without dose modification specific to Japanese patients.


Subject(s)
Ethnicity , Rilpivirine , Adult , Asian People , Healthy Volunteers , Humans , Rilpivirine/adverse effects , White People
5.
Intest Res ; 19(4): 386-397, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33249802

ABSTRACT

BACKGROUND/AIMS: We aimed to evaluate the efficacy and safety of ustekinumab (UST) in the East-Asian population with moderate to severely active ulcerative colitis (UC). METHODS: This sub-analysis was conducted on data from East-Asian patients included in the UNIFI program (NCT02407236). UNIFI consisted of two double-blind, placebo-controlled trials: an 8-week induction study and a 44-week randomized withdrawal maintenance study. RESULTS: Of 133 East-Asian patients (Japanese: 107, Korean: 26) who underwent randomization, 131 completed induction study and 111 entered maintenance study. In the maintenance study, 78 patients were randomized. Patients who received UST 130 mg and UST 6 mg/kg showed numerically higher clinical remission at week 8 in the induction study (5/44 [11.4%] and 5/45 [11.1%], respectively) compared with those who received placebo (0/44, 0%). The proportion of patients achieved clinical remission at week 44 was numerically higher in the UST 90 mg q12w group (10/21, 47.6%), but similar in the UST 90 mg q8w group (5/26, 19.2%) compared to placebo (7/31, 22.6%). Serious adverse events were reported in 1 patient in UST 130 mg group, but no patient in UST 6 mg/kg group through week 8 in the induction study, and 1 patient in UST 90 mg q12w group and 5 patients in the UST 90 mg q8w group in the maintenance study. No deaths were reported in East-Asian patients throughout the study. CONCLUSIONS: UST induction and maintenance treatments were effective in East-Asian patients with moderate to severe UC; the efficacy and safety profiles were consistent with the overall population.

6.
Intest Res ; 15(4): 475-486, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29142515

ABSTRACT

BACKGROUND/AIMS: Efficacy and safety of ustekinumab were evaluated in a Japanese subpopulation with moderately to severely active Crohn's disease (CD) in UNITI-1, UNITI-2 and IM-UNITI studies and results were compared with the overall population. METHODS: Overall, patients in UNITI-1 (Japan, n=56; failed response to tumor necrosis factor antagonist) and UNITI-2 (Japan, n=26; failed response to prior conventional therapy) were randomized to placebo or ustekinumab intravenous induction (130 mg or ~6 mg/kg) at week 0. Responders to ustekinumab induction therapy (Japan, n=21) were randomized to placebo or ustekinumab (90 mg, subcutaneous) maintenance (every 12 weeks [q12w] or 8 weeks [q8w]) in IM-UNITI. The primary endpoint was clinical response at week 6 for induction studies and clinical remission at week 44 for maintenance study. RESULTS: Percentage of patients achieving clinical response at week 6 was greater in ustekinumab 130 mg and ~6 mg/kg groups than in the placebo group (UNITI-1: 36.8% and 31.6% vs. 27.8%, respectively, for Japanese; 34.3% and 33.7% vs. 21.5%, respectively, for overall; UNITI-2: 37.5% and 55.6% vs. 11.1%, respectively, for Japanese; 51.7% and 55.5% vs. 28.7%, respectively, for overall). Clinical remission rate at week 44 during maintenance was greater in the ustekinumab 90 mg SC q12w and q8w groups than in the placebo group (50.0% and 55.6% vs. 25.0%, respectively, for Japanese; 48.8% and 53.1% vs. 35.9%, respectively, for overall). Efficacy and safety results observed in the Japanese subpopulation were generally consistent with those in the overall population. CONCLUSIONS: Ustekinumab could be considered as a new therapeutic option for moderately to severely active CD in Japanese patients. Both ustekinumab induction and maintenance treatments were generally well tolerated (Clinical Trial Registration: NCT01369329, NCT01369342, NCT01369355).

7.
Clin Pharmacol Drug Dev ; 6(4): 398-407, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27652589

ABSTRACT

We evaluated the pharmacokinetics and pharmacodynamics of prasugrel used in combination with aspirin in healthy Japanese subjects. All subjects received aspirin 100 mg/day. Subsequently, in the single-administration study, 23 subjects also received prasugrel 20 or 30 mg, and in the multiple-administration study, 20 subjects received a loading dose of prasugrel 20 or 30 mg on day 1, followed by a maintenance dose of prasugrel 5 or 7.5 mg/day, respectively, on days 2-5. In both studies, the plasma concentration of the active metabolite of prasugrel, R-138727, reached a maximum 0.5 hours after administration and rapidly decreased within 4 hours. In the single-administration study, the inhibitory effect on adenosine diphosphate-induced platelet aggregation was significantly higher in the prasugrel 20- and 30-mg groups than in the placebo group at all times (1-144 hours) after administration. In the multiple-administration study, a similar antiplatelet effect was found after both the loading dose and the maintenance dose and was maintained for 3-6 days after the last administration. There were study drug-related adverse events; however, all were mild, and none was clinically significant.


Subject(s)
Aspirin/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Adult , Area Under Curve , Aspirin/administration & dosage , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Healthy Volunteers , Humans , Japan , Piperazines/blood , Platelet Aggregation Inhibitors/administration & dosage , Platelet Function Tests , Prasugrel Hydrochloride/administration & dosage , Young Adult
8.
Br J Clin Pharmacol ; 82(6): 1591-1600, 2016 12.
Article in English | MEDLINE | ID: mdl-27530188

ABSTRACT

AIMS: Edoxaban, a novel factor Xa inhibitor, is a substrate of cytochrome P450 3 A4 (CYP3A4) and the efflux transporter P-glycoprotein (P-gp). Three edoxaban drug-drug interaction studies examined the effects of P-gp inhibitors with varying degrees of CYP3A4 inhibition. METHODS: In each study, healthy subjects received a single oral dose of 60 mg edoxaban with or without an oral dual P-gp/CYP3A4 inhibitor as follows: ketoconazole 400 mg once daily for 7 days, edoxaban on day 4; erythromycin 500 mg four times daily for 8 days, edoxaban on day 7; or single dose of cyclosporine 500 mg with edoxaban. Serial plasma samples were obtained for pharmacokinetics and pharmacodynamics. Safety was assessed throughout the study. RESULTS: Coadministration of ketoconazole, erythromycin, or cyclosporine increased edoxaban total exposure by 87%, 85%, and 73%, respectively, and the peak concentration by 89%, 68%, and 74%, respectively, compared with edoxaban alone. The half-life did not change appreciably. Exposure of M4, the major active edoxaban metabolite, was consistent when edoxaban was administered alone or with ketoconazole and erythromycin. With cyclosporine, M4 total exposure increased by 6.9-fold and peak exposure by 8.7-fold, suggesting an additional interaction. Pharmacodynamic effects were reflective of increased edoxaban exposure. No clinically significant adverse events were observed. CONCLUSIONS: Administration of dual inhibitors of P-gp and CYP3A4 increased edoxaban exposure by less than two-fold. This effect appears to be primarily due to inhibition of P-gp. The impact of CYP3A4 inhibition appears to be less pronounced, and its contribution to total clearance appears limited in healthy subjects.


Subject(s)
Cyclosporine/pharmacokinetics , Erythromycin/pharmacokinetics , Factor Xa Inhibitors/pharmacokinetics , Ketoconazole/pharmacokinetics , Pyridines/pharmacokinetics , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Adolescent , Adult , Cross-Over Studies , Cyclosporine/administration & dosage , Cyclosporine/blood , Cytochrome P-450 CYP3A/metabolism , Drug Administration Schedule , Drug Interactions , Erythromycin/administration & dosage , Erythromycin/blood , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/blood , Healthy Volunteers , Humans , Ketoconazole/administration & dosage , Ketoconazole/blood , Metabolic Clearance Rate , Middle Aged , Pyridines/administration & dosage , Pyridines/blood , Substrate Specificity , Thiazoles/administration & dosage , Thiazoles/blood , Young Adult
9.
Cancer Chemother Pharmacol ; 77(5): 987-96, 2016 05.
Article in English | MEDLINE | ID: mdl-27017616

ABSTRACT

PURPOSE: The purpose of this analysis was to develop a population pharmacokinetic (PK) model for patritumab, a fully human monoclonal antibody that targets human epidermal growth factor receptor 3. METHODS: A total of 833 serum concentrations were included in this analysis; serum concentrations were obtained from 145 subjects (136 with non-small cell lung cancer, nine with solid tumors) treated with patritumab [9 or 18 mg/kg intravenously every 3 weeks (q3w)] in one phase 1 and one phase 1b/2 study. Data were analyzed by nonlinear mixed-effect modeling. RESULTS: Patritumab PKs were best described through a two-compartment model with first-order elimination and interindividual variability on clearance (CL), volume of the central compartment (V c), distributional clearance, and volume of the peripheral compartment. In the final model, CL and V c were estimated as 0.0238 L/h and 3.62 L, respectively. Body weight (BW) and baseline albumin were found to be covariates for CL and BW was a covariate for V c. Covariates associated with hepatic and renal impairment were not significant on CL. Simulations showed that BW-based dosing reduced interindividual variability in patritumab exposure compared with fixed dosing. CONCLUSIONS: The PK of patritumab was linear at the doses studied and well described by the two-compartment model. Hepatic and renal impairment did not appear to affect PK. Our results support BW-based dosing of patritumab on a q3w schedule.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Models, Biological , Receptor, ErbB-3/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Broadly Neutralizing Antibodies , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Computer Simulation , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Dosage Calculations , Female , Humans , Injections, Intravenous , Lung Neoplasms/blood , Lung Neoplasms/pathology , Male , Middle Aged , Tissue Distribution
10.
J Clin Pharmacol ; 56(4): 484-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26239632

ABSTRACT

Two studies (ROADMAP and ORIENT) evaluating the renoprotective effects of olmesartan medoxomil (OM) in patients with type 2 diabetes suggested OM is associated with increased cardiovascular mortality. We conducted a thorough QTc study to evaluate the effects of OM on cardiac repolarization. A randomized, double-blind, phase 1 study was conducted per E14 Guidance to assess the effects of single doses of OM therapeutic dose (40 mg), OM supratherapeutic dose (160 mg), placebo, or moxifloxacin (MOXI; 400 mg) on QTc in 56 healthy subjects. The primary endpoint was the baseline-adjusted, placebo-corrected QTc interval using Fridericia's formula (ΔΔQTcF) for OM and MOXI. Assay sensitivity was concluded if lower limit of 1-sided 95%CI > 5 milliseconds of ΔΔQTcF for MOXI. No threshold pharmacologic effect for OM was concluded if upper limit of 1-sided 95%CI <10 milliseconds for ΔΔQTcF at any timepoint. Pharmacokinetics, ECGs, and safety were assessed. Assay sensitivity was demonstrated. The largest upper limit of the 1-sided 95%CI for ΔΔQTcF was <5 milliseconds for OM. No clinically significant changes were observed in ECGs. Pharmacokinetics and safety profile were consistent with previous data. Therapeutic and supratherapeutic OM doses had no clinically significant effect on cardiac repolarization and were well tolerated.


Subject(s)
Electrocardiography/drug effects , Heart/drug effects , Long QT Syndrome/chemically induced , Olmesartan Medoxomil/administration & dosage , Adolescent , Adult , Cross-Over Studies , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
12.
Drug Metab Dispos ; 42(3): 326-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24319124

ABSTRACT

(2R,3R,4R)-4-hydroxy-2-(hydroxymethyl)pyrrolidin-3-yl 4-O-(6-deoxy-ß-D-glucopyranosyl)-α-D-glucopyranoside (CS-1036), which is an α-amylase inhibitor, exhibited biphasic and sustained elimination with a long t1/2 (18.4-30.0 hours) in rats and monkeys, but exhibited a short t1/2 (3.7-7.9 hours) in humans. To clarify the species differences in the t1/2, the plasma protein binding of CS-1036 was evaluated by ultrafiltration. A concentration-dependent and saturable plasma protein binding of CS-1036 was observed in rats and monkeys with the dissociation rate constant (KD) of 8.95 and 27.2 nM, and maximal binding capacity (Bmax) of 52.8 and 22.1 nM, respectively. By the assessments of the recombinant amylase and immunoprecipitation, the major binding protein of CS-1036 in rats was identified as salivary amylase (KD 5.64 nM). CS-1036 also showed concentration-dependent and saturable binding to human salivary and pancreatic amylase, with similar binding affinity in rats. However, the protein binding of CS-1036 was constant in human plasma (≤10.2%) due to the lower serum amylase level compared with rats and monkeys. From the calculation of the unbound fraction (fu) in plasma based on in vitro KD and Bmax, the dose-dependent increase in fu after oral administration is speculated to lead to a dose-dependent increase in total body clearance and a high area under the curve/dose at lower doses, such as 0.3 mg/kg in rats.


Subject(s)
Blood Proteins/metabolism , Disaccharides/pharmacology , Enzyme Inhibitors/pharmacology , Pancreatic alpha-Amylases/antagonists & inhibitors , Pyrrolidines/pharmacology , Salivary alpha-Amylases/antagonists & inhibitors , Adult , Animals , Disaccharides/blood , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/blood , Escherichia coli/genetics , Humans , Immunoprecipitation , Macaca fascicularis , Male , Pancreatic alpha-Amylases/blood , Pancreatic alpha-Amylases/genetics , Protein Binding , Pyrrolidines/blood , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Recombinant Proteins , Salivary alpha-Amylases/blood , Salivary alpha-Amylases/genetics , Species Specificity , Ultrafiltration , Young Adult
13.
Am J Cardiovasc Drugs ; 13(5): 331-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23784266

ABSTRACT

BACKGROUND: Edoxaban, an oral direct factor Xa inhibitor, is in development for thromboprophylaxis, including prevention of stroke and systemic embolism in patients with atrial fibrillation (AF). P-glycoprotein (P-gp), an efflux transporter, modulates absorption and excretion of xenobiotics. Edoxaban is a P-gp substrate, and several cardiovascular (CV) drugs have the potential to inhibit P-gp and increase drug exposure. OBJECTIVE: To assess the potential pharmacokinetic interactions of edoxaban and 6 cardiovascular drugs used in the management of AF and known P-gp substrates/inhibitors. METHODS: Drug-drug interaction studies with edoxaban and CV drugs with known P-gp substrate/inhibitor potential were conducted in healthy subjects. In 4 crossover, 2-period, 2-treatment studies, subjects received edoxaban 60 mg alone and coadministered with quinidine 300 mg (n = 42), verapamil 240 mg (n = 34), atorvastatin 80 mg (n = 32), or dronedarone 400 mg (n = 34). Additionally, edoxaban 60 mg alone and coadministered with amiodarone 400 mg (n = 30) or digoxin 0.25 mg (n = 48) was evaluated in a single-sequence study and 2-cohort study, respectively. RESULTS: Edoxaban exposure measured as area under the curve increased for concomitant administration of edoxaban with quinidine (76.7 %), verapamil (52.7 %), amiodarone (39.8 %), and dronedarone (84.5 %), and exposure measured as 24-h concentrations for quinidine (11.8 %), verapamil (29.1 %), and dronedarone (157.6 %) also increased. Administration of edoxaban with amiodarone decreased the 24-h concentration for edoxaban by 25.7 %. Concomitant administration with digoxin or atorvastatin had minimal effects on edoxaban exposure. CONCLUSION: Coadministration of the P-gp inhibitors quinidine, verapamil, and dronedarone increased edoxaban exposure. Modest/minimal effects were observed for amiodarone, atorvastatin, and digoxin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Anticoagulants/pharmacokinetics , Cardiovascular Agents/pharmacology , Pyridines/pharmacokinetics , Thiazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Area Under Curve , Cohort Studies , Cross-Over Studies , Drug Interactions , Factor Xa Inhibitors , Female , Humans , Male , Young Adult
14.
Clin Pharmacol Drug Dev ; 2(4): 358-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-27121940

ABSTRACT

BACKGROUND: Edoxaban is an oral, once-daily, direct factor Xa inhibitor under investigation for stroke prevention in patients with atrial fibrillation and for treatment and secondary prevention of venous thromboembolism. This study evaluated edoxaban absolute bioavailability and effects of the P-glycoprotein inhibitor quinidine on edoxaban pharmacokinetics after intravenous edoxaban administration. METHODS: Healthy volunteers received three treatments in a randomized sequence: single oral 60-mg edoxaban dose, single intravenous 30-mg edoxaban dose, and concomitant single intravenous 30-mg edoxaban dose with quinidine 300 mg every 8 hours for 4 days. The primary objective was to determine absolute bioavailability of edoxaban. Secondary objectives included pharmacokinetics and pharmacodynamics of edoxaban after oral or intravenous administration, quinidine effect on intravenous edoxaban pharmacokinetics, and safety. RESULTS: Thirty-six subjects were randomized; five discontinued (three for adverse events [AEs]). Edoxaban oral absolute bioavailability was 61.8%. With concomitant quinidine, total edoxaban exposure increased ∼35% and total clearance decreased ∼25%. Coagulation parameters increased after edoxaban administration in most subjects, but returned to baseline within 24 hours postdose. No deaths, serious AEs, or bleeding-related AEs occurred. CONCLUSIONS: Absolute bioavailability of edoxaban in healthy volunteers was established (61.8%). Edoxaban, administered orally or intravenously, appeared to be safe and well tolerated.

15.
Thromb Haemost ; 108(1): 166-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22628060

ABSTRACT

Edoxaban is an oral direct factor (F)Xa inhibitor in advanced stages of clinical development. The primary objective of the present study was to assess the pharmacodynamics (PD) and safety of enoxaparin 1 mg/kg followed 12 hours (h) post-dose by edoxaban 60 mg, which is the regimen being used in the phase III study of edoxaban for the treatment of venous thromboembolism (Hokusai-VTE). This was a phase I, open-label, randomised, four-period, four-treatment cross-over study. Treatments were edoxaban alone (EDOX), enoxaparin alone (ENOX), edoxaban plus enoxaparin (EDOX+ENOX), and enoxaparin followed by edoxaban 12 h later (ENOX12-EDOX). Serial blood samples were collected for PD (thrombin generation, anti-FXa) and pharmacokinetic (PK) variables (edoxaban and its principal metabolite M4 by LC-MS/MS, and anti-FIIa as a surrogate of enoxaparin). The highest effect on thrombin AUC (endogenous thrombin potential, or ETP), thrombin (peak), thrombin generation lag time, and velocity index was observed for EDOX+ENOX, followed by ENOX, ENOX12-EDOX, and EDOX. The greatest effect on anti-FXa activity was observed for EDOX+ENOX, followed by ENOX12-EDOX. As expected, neither edoxaban nor enoxaparin significantly altered the PK of the other drug. There were no serious adverse events during the study. It is concluded that a 60-mg dose of edoxaban can be safely administered 12 h following enoxaparin 1 mg/kg.


Subject(s)
Enoxaparin/administration & dosage , Pyridines/administration & dosage , Thiazoles/administration & dosage , Venous Thromboembolism/drug therapy , Adult , Blood Coagulation Tests , Chromatography, Liquid , Drug Interactions , Drug Therapy, Combination , Enoxaparin/adverse effects , Enoxaparin/pharmacokinetics , Factor Xa Inhibitors , Female , Humans , Male , Mass Spectrometry , Pyridines/adverse effects , Pyridines/pharmacokinetics , Thiazoles/adverse effects , Thiazoles/pharmacokinetics , Thrombin/biosynthesis , Venous Thromboembolism/blood
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 856(1-2): 190-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17602900

ABSTRACT

A specific, sensitive and fast method based on high performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) was developed for the determination of olmesartan in human plasma and urine. Solid-phase extraction (SPE) was used to isolate the compounds from biological matrix followed by injection of the extracts onto a C(18) column with isocratic elution. The method was validated over the concentration range of 0.2-1000 and 5-10,000 ng/mL for olmesartan in human plasma and urine, respectively. The method was applied to the pharmacokinetic study of olmesartan medoxomil in healthy Chinese male and female subjects.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Imidazoles/pharmacokinetics , Tandem Mass Spectrometry/methods , Tetrazoles/pharmacokinetics , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II Type 1 Receptor Blockers/urine , Female , Humans , Imidazoles/blood , Imidazoles/urine , Male , Reproducibility of Results , Sensitivity and Specificity , Tetrazoles/blood , Tetrazoles/urine
17.
Br J Clin Pharmacol ; 63(4): 421-30, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17076696

ABSTRACT

AIMS: This double-blind, placebo-controlled trial was designed to evaluate the pharmacodynamics, pharmacokinetics, safety, and tolerability of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y(12) ADP receptor antagonist compared with clopidogrel, during multiple oral dosing in healthy subjects. METHODS: Thirty subjects received placebo, prasugrel 5 mg, 10 mg, or 20 mg, or clopidogrel 75 mg orally, daily for 10 days. Platelet aggregation, bleeding time, and prasugrel metabolites were measured and adverse events were recorded. RESULTS: Inhibition of ADP-induced platelet aggregation reached steady state by day 3 following prasugrel 10 and 20 mg compared with 5 days for clopidogrel 75 mg or prasugrel 5 mg. Compared with placebo, at 24 h after the last dose of study drug, inhibition of platelet aggregation using (20 microm) ADP was significantly higher in the prasugrel 10 mg group (58.2 +/- 4.9% vs. 9.2 +/- 4.0%, P < 0.001) with no difference in the clopidogrel group (15.7 +/- 6.8% vs. 9.2 +/- 4.0%, P = 0.78). With 5 microm ADP, inhibition of platelet aggregation with prasugrel 10 mg and clopidogrel 75 mg was significantly higher than with placebo (prasugrel 10 mg, 70.5 +/- 4.7%; clopidogrel 75 mg, 36.5 +/- 9.0%; vs. placebo, 11.3 +/- 5.1%; P < 0.0001 and P = 0.02). On day 10 at 4 h postdose, bleeding time was prolonged with prasugrel 10 mg (prasugrel 10 mg, 706 +/- 252 s vs. placebo, 221 +/- 38 s, P = 0.05) but not with clopidogrel (283 +/- 56 s, P = 0.98). There were no clinically significant bleeding events, serious adverse events, or discontinuations of the study drug. CONCLUSIONS: Compared with clopidogrel 75 mg, prasugrel 10 mg and 20 mg daily for 10 days resulted in more rapid, more consistent, and higher levels of platelet inhibition.


Subject(s)
Piperazines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Thiophenes/administration & dosage , Ticlopidine/analogs & derivatives , Adolescent , Adult , Clopidogrel , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Humans , Male , Middle Aged , Piperazines/adverse effects , Piperazines/pharmacokinetics , Platelet Aggregation , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride , Receptors, Purinergic P2 , Receptors, Purinergic P2Y12 , Thiophenes/adverse effects , Thiophenes/pharmacokinetics , Ticlopidine/administration & dosage , Ticlopidine/adverse effects , Ticlopidine/pharmacokinetics
18.
J Pharmacokinet Pharmacodyn ; 33(5): 635-55, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16941234

ABSTRACT

The application of D-optimization and the assessment of bias and precision of parameter estimates for four basic pharmacodynamic (PD) indirect response (IDR) models for ascending doses was examined using simulated data. While D-optimization provided four sampling times, each IDR model was used to generate eight data points per dose level. The PD parameters were: input rate constant (k (in)), disposition rate constant (k (out)), capacity constant (I (max) or S (max)), and sensitivity constant (IC (50) or SC (50)). A monoexponential pharmacokinetic function was applied with single doses increased by a factor of 10 to generate responses that vary from weak to fully saturable. For each dose, 100 replications of response data were simulated using independent normally distributed errors of CV = 20%. The original IDR model was fitted and PD parameters estimated. Histograms and descriptive statistics were generated. All parameters exhibited asymmetric distributions with positive coefficients of skewness except for I (max). Higher doses resulted in unbiased estimates of all PD parameters. The precision of parameter estimates improved with increasing doses except for IC (50) and SC (50) indicating that a single dose experimental design cannot be corrected by increasing dose in order to improve precision of estimates of IC (50) or SC (50). Highest variability was for IC (50) and SC (50) parameters. This study provides new insights into optimum study designs and recovery of parameters for basic IDR models.


Subject(s)
Models, Biological , Observer Variation , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Biological Availability , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Metabolic Clearance Rate , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/metabolism , Research Design/trends , Time Factors
19.
Platelets ; 17(4): 218-26, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16769599

ABSTRACT

This double-blind, placebo-controlled trial evaluated the safety, pharmacodynamics, and pharmacokinetics of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y12 ADP receptor antagonist, during multiple oral dosing in healthy subjects. Eighteen subjects received placebo, or prasugrel 2.5 or 10 mg, orally, daily for 10 days. Adverse events were recorded and blood samples for measurement of platelet aggregation, bleeding time, and prasugrel metabolite concentrations were obtained. Two bleeding events were experienced in the prasugrel 10 mg dose group and one in the placebo group. Neither of the events was considered serious. ADP-induced platelet aggregation accumulated over the 10-day study period, reaching a steady state by days 2-4 following administration of prasugrel 10 mg daily. Limited inhibition of platelet aggregation was obtained with prasugrel 2.5 mg. In the 10-mg dose group at day 5, 4 h postdose, with 20 microM ADP as the agonist, inhibition of platelet aggregation was 61.2 +/- 5.6 vs. 17.9 +/- 6.2% in the placebo group (P < 0.01). Recovery of platelet aggregation was observed 48 h after the last dose of prasugrel 10 mg, but was only partial, consistent with irreversible platelet inhibition. The mean bleeding time was prolonged by day 5 at 4 h postdose in all subjects in the prasugrel 10 mg dose group (prasugrel 10 mg, 1058 +/- 412 s vs. placebo, 196 +/- 74 s; P < 0.001). The maximum plasma concentration of metabolites increased with prasugrel dose and there was no accumulation of metabolites over the 10-day study period. This study indicated that prasugrel 10 mg daily for 10 days was well tolerated and at steady-state provided sustained high levels of inhibition of platelet aggregation.


Subject(s)
Piperazines/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation/drug effects , Purinergic P2 Receptor Antagonists , Thiophenes/pharmacokinetics , Administration, Oral , Adolescent , Adult , Bleeding Time , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/adverse effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Thiophenes/administration & dosage , Thiophenes/adverse effects , Time Factors
20.
Platelets ; 17(4): 209-17, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16769598

ABSTRACT

We assessed the tolerability, pharmacodynamics as measured by inhibition of platelet aggregation (IPA), and pharmacokinetics of prasugrel (CS-747, LY640315), a novel thienopyridine antiplatelet agent in healthy volunteers. Twenty-four subjects were randomized into four groups of six in a double-blind, placebo-controlled trial. One subject in each group received placebo and five subjects received prasugrel orally at single doses of 2.5, 10, 30, or 75 mg. The IPA, assessed using 5 and 20 microM ADP, was periodically measured over a 7-day period by light transmission aggregometry. Plasma concentrations for three major metabolites, R-95913, R-106583, and R-100932, were measured. There were no serious adverse events and no clinically significant changes noted in any laboratory or clinical evaluations in any subject. At 1 h after prasugrel 30 and 75 mg, platelet aggregation induced by 20 microM ADP was inhibited by 43.5 +/- 7.8 and 43.2 +/- 15.7%, respectively, and this inhibition was significantly greater than that following placebo (5.9 +/- 3.5%) (P < 0.05 for both doses). The degree of inhibition observed at 2 h was slightly higher with both prasugrel 30 and 75 mg (59.8 +/- 9.9 and 57.0 +/- 7.2%) and was maintained through the subsequent 22 h. At 24 h, maximal platelet aggregation induced by 20 microM ADP was reduced to 0.05 for 2.5 and 10 mg prasugrel vs. placebo). With prasugrel 75 mg at 4 h postdose, there was a significant increase in the mean bleeding time compared to placebo (682 vs. 161 s; P < 0.05). Prasugrel metabolites obeyed linear pharmacokinetics and the three metabolites appeared in the plasma soon after administration, reaching maximum levels at approximately 1 h. In conclusion, prasugrel 30 and 75 mg were well tolerated and achieved a consistently high level of platelet inhibition with a fast onset of action.


Subject(s)
Piperazines/pharmacokinetics , Platelet Aggregation/drug effects , Purinergic P2 Receptor Antagonists , Thiophenes/pharmacokinetics , Adenosine Diphosphate/pharmacology , Administration, Oral , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/blood , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Thiophenes/administration & dosage , Thiophenes/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...