Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 371
Filter
1.
Pharm Dev Technol ; 26(9): 1021-1033, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34435939

ABSTRACT

Prasugrel Hydrochloride (PHCl) is an antiplatelet drug. It is a class II drug with variable bioavailability. The objective of this work was to enhance the solubility and hence the bioavailability and efficacy of PHCl. A Self Nano-Emulsifying Drug Delivery System (SNEDDS) was prepared using Kolliphor El, Maisine 35-1, and Transcutol P as surfactant, oil, and co-surfactant, respectively in a ratio 10:72:18 v/v%. The SNEDDS was converted into solid by adsorption onto Neusilin. In vitro release of the drug from SNEDDS in (pH = 4) at 37 °C and 75 rpm for 45 min was studied. The results were compared to those from the unprocessed PHCl and Lexar® (the commercial drug). In-vivo studies (platelet Aggregation and bleeding time) were conducted using rats as animal models. It was found that the particle size of the SNEDDS ranged between 80 and 155 nm and EE% was in the range of 90.2% ± 0.4. The release from SNEDDS was about 84% compared to around 25% from unprocessed PHCl and 65% from Lexar® after 15 min. The platelet aggregation of the formula was lower than the PHCl, and Lexar® indicating higher bioavailability. In conclusion, SNEDDS with high EE% was prepared and was successful in enhancing the solubility, dissolution rate, and the bioavailability.


Subject(s)
Emulsifying Agents/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Animals , Biological Availability , Dose-Response Relationship, Drug , Emulsifying Agents/blood , Male , Platelet Aggregation Inhibitors/blood , Prasugrel Hydrochloride/blood , Rats , Rats, Sprague-Dawley , Solubility , X-Ray Diffraction/methods
2.
Catheter Cardiovasc Interv ; 95(1): E8-E16, 2020 01.
Article in English | MEDLINE | ID: mdl-30983087

ABSTRACT

OBJECTIVE: The aim of this study was to compare how prasugrel and clopidogrel affect platelet aggregation reactivity, cardiac enzyme release, cardiac remodeling, and the formation of in-stent thrombi after primary percutaneous coronary intervention (PCI). BACKGROUND: The advantages of using prasugrel over clopidogrel in cardiac injury following acute coronary syndrome (ACS) remain unclear. METHODS: A total of 78 ACS patients were randomly allocated into clopidogrel (300 mg loading/75 mg maintenance) or prasugrel (20 mg loading/3.75 mg maintenance) treatment groups, followed by undergoing primary PCI. Platelet reactivity and cardiac enzymes were measured before and after primary PCI. Moreover, cardiac function was measured by ultrasound echocardiography and coronary angioscopic observation was after primary PCI up to 8 months later. RESULTS: Antiplatelet reactivity in the prasugrel treatment group reached optimal levels (P2Y12 reaction units [PRU] less than 262) immediately after the administration and was maintained even at 8 months, independently of the CYP2C19 genotype. Prasugrel treatment significantly suppressed creatine kinase elevation compared to clopidogrel treatment (median value 404 IU/L to 726 IU/L vs. 189 IU/L to 1,736 IU/L, p = 0.018 for maximum values) and reduced left ventricular mass (217.2-168.8 g in prasugrel, p = 0.045; 196.9-176.4 g in clopidogrel, p = 0.061). There were no significant differences in the incidence of in-stent attached thrombi between the two groups. CONCLUSIONS: Compared to clopidogrel, prasugrel produced a stable platelet aggregation inhibitory effect in patients with ACS regardless of CYP2C19 genotype, reduced cardiac enzyme release, and prevented cardiac remodeling after ACS.


Subject(s)
Acute Coronary Syndrome/therapy , Clopidogrel/administration & dosage , Coronary Thrombosis/prevention & control , Drug-Eluting Stents , Percutaneous Coronary Intervention/instrumentation , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Prasugrel Hydrochloride/administration & dosage , Purinergic P2Y Receptor Antagonists/administration & dosage , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnostic imaging , Acute Coronary Syndrome/physiopathology , Aged , Clopidogrel/adverse effects , Clopidogrel/blood , Coronary Thrombosis/diagnostic imaging , Coronary Thrombosis/etiology , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Female , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/blood , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/blood , Prospective Studies , Purinergic P2Y Receptor Antagonists/adverse effects , Purinergic P2Y Receptor Antagonists/blood , Risk Factors , Time Factors , Tokyo , Treatment Outcome
3.
Nitric Oxide ; 97: 20-26, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32006712

ABSTRACT

The study examines the effect of two water-soluble carbon monoxide (CO) donors, CORM-3 and CORM-A1, on selected parameters of oxidative stress and hemostasis in human plasma and blood platelets in vitro. It also compares their activity with that of the lipid-soluble CORM-2. The oxidation of amino acid residues in plasma proteins was evaluated by measuring the amounts of thiol and carbonyl groups. Plasma lipid peroxidation was measured as thiobarbituric acid reactive substance (TBARS) concentration. In addition, three haemostatic parameters of plasma were studied, viz. activated partial thromboplastin time (APTT), prothrombin time (PT) and thrombin time (TT), and one haemostatic parameter of platelets (platelet aggregation). Treatment with CORM-3 and CORM-A1 (all concentrations from 0.1 to 100 µM) decreased thiol group oxidation induced by H2O2/Fe. Incubation with CORM-3 and CORM-A1 also influenced plasma coagulation activity, e.g. CORM-3 and CORM-A1 significantly prolonged TT at the two highest tested concentrations (50 and 100 µM). Only CORM-2 at the highest tested concentration (100 µM) and CORM-3 (50 and 100 µM) reduced platelet aggregation induced by ADP. None of the tested CORMs caused platelet damage. The treatment of various diseases associated with oxidative stress, including cardiovascular diseases, may be enhanced by the administration of CO donors CORM-2 and CORM-3, these being modulators of oxidative stress and hemostasis.


Subject(s)
Anticoagulants/blood , Antioxidants/metabolism , Boranes/blood , Carbonates/blood , Models, Biological , Organometallic Compounds/blood , Platelet Aggregation Inhibitors/blood , Healthy Volunteers , Hemostasis , Humans , Oxidative Stress
4.
Pharm Biol ; 58(1): 630-635, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32633174

ABSTRACT

CONTEXT: Rivaroxaban and ticagrelor are two common drugs for the treatment of atrial fibrillation and acute coronary syndrome. However, the drug-drug interaction between them is still unknown. OBJECTIVE: To investigate the effects of ticagrelor on the pharmacokinetics of rivaroxaban in rats both in vivo and in vitro. MATERIALS AND METHODS: A sensitive and reliable UPLC-MS/MS method was developed for the determination of rivaroxaban in rat plasma. Ten Sprague-Dawley rats were randomly divided into ticagrelor pre-treated group (10 mg/kg/day for 14 days) and control group. The pharmacokinetics of orally administered rivaroxaban (10 mg/kg, single dose) with or without ticagrelor pre-treatment was investigated with developed UPLC-MS/MS method. Additionally, Sprague-Dawley rat liver microsomes were also used to investigate the drug-drug interaction between these two drugs in vitro. RESULTS: The C max (221.34 ± 53.33 vs. 691.18 ± 238.31 ng/mL) and the AUC(0-t) (1060.97 ± 291.21 vs. 3483.03 ± 753.83 µg·h/L) of rivaroxaban increased significantly (p < 0.05) with ticagrelor pre-treatment. The MRT(0-∞) of rivaroxaban increased from 4.41 ± 0.79 to 5.97 ± 1.11 h, while the intrinsic clearance decreased from 9.93 ± 2.55 to 2.89 ± 0.63 L/h/kg (both p < 0.05) after pre-treated with ticagrelor. Enzyme kinetic study indicated that ticagrelor decreased rivaroxaban metabolic clearance with the IC50 value of 14.04 µmol/L. CONCLUSIONS: Our in vivo and in vitro results demonstrated that there is a drug-drug interaction between ticagrelor and rivaroxaban in rats. Further studies need to be carried out to verify whether similar interactions truly apply in humans and whether these interactions have clinical significance.


Subject(s)
Factor Xa Inhibitors/pharmacokinetics , Microsomes, Liver/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Rivaroxaban/pharmacokinetics , Ticagrelor/pharmacokinetics , Animals , Drug Interactions/physiology , Factor Xa Inhibitors/blood , Male , Microsomes, Liver/drug effects , Platelet Aggregation Inhibitors/blood , Rats , Rats, Sprague-Dawley , Rivaroxaban/blood , Ticagrelor/blood
5.
Anal Chem ; 91(16): 10830-10839, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31343155

ABSTRACT

There is a need for scalable automated lab-on-chip systems incorporating precise hemodynamic control that can be applied to high-content screening of new more efficacious antiplatelet therapies. This paper reports on the development and characterization of a novel active micropump-mixer microfluidic to address this need. Using a novel reciprocating elastomeric micropump design, we take advantage of the flexible structural and actuation properties of this framework to manage the hemodynamics for on-chip platelet thrombosis assay on type 1 fibrillar collagen, using whole blood. By characterizing and harnessing the complex three-dimensional hemodynamics of the micropump operation in conjunction with a microvalve controlled reagent injection system we demonstrate that this prototype can act as a real-time assay of antiplatelet drug pharmacokinetics. In a proof-of-concept preclinical application, we utilize this system to investigate the way in which rapid dosing of human whole blood with isoform selective inhibitors of phosphatidylinositol 3-kinase dose dependently modulate platelet thrombus dynamics. This modular system exhibits utility as an automated multiplexable assay system with applications to high-content chemical library screening of new antiplatelet therapies.


Subject(s)
Indomethacin/blood , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Platelet Aggregation Inhibitors/blood , Blood Platelets/drug effects , Hemodynamics , Humans , Indomethacin/pharmacokinetics , Microfluidic Analytical Techniques/instrumentation , Platelet Aggregation Inhibitors/pharmacokinetics
6.
Int J Clin Pharmacol Ther ; 57(2): 82-93, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30431425

ABSTRACT

BACKGROUND AND OBJECTIVES: A significant number of ischemic events occur after acute myocardial infarction (MI), even when adhering to dual antiplatelet therapy including aspirin and clopidogrel. The aim of our study was to investigate the association between the concentration of the prodrug clopidogrel and its intermediary metabolite 2-oxo-clopidogrel plasma as well as demographic and clinical factors, and the long-term clinical outcome in patients with their first acute MI, ST-elevation myocardial infarction (STEMI) or non-ST-elevation myocardial infarction NSTEMI, treated with percutaneous coronary intervention (PCI). MATERIALS AND METHODS: This study included 172 consecutive patients with their first acute MI, 88 STEMI, and 84 NSTEMI, treated with PCI. On the third day of hospitalization, blood samples were collected from each patient to measure the concentration of clopidogrel and its metabolite 2-oxo-clopidogrel using the UHPLC-DAD-MS method. The following clinical outcomes were registered during the 28-month follow-up: mortality from cardiovascular causes, nonfatal MI, nonfatal stroke, and hospitalization for urgent myocardial revascularization or heart failure. RESULTS: Lower dose-adjusted clopidogrel concentrations (p < 0.05) were measured in NSTEMI patients with a composite of the hard clinical endpoint events of cardiovascular mortality, non-fatal MI, or a nonfatal stroke. During the follow-up, there was a 3.4 times higher risk of hard clinical endpoint events (p < 0.05) for each unit decrement of the dose-adjusted clopidogrel plasma concentration. Lower dose-adjusted concentrations of clopidogrel in these patients were associated with lower left ventricular ejection fraction (p < 0.001), and fentanyl (p < 0.001) and pantoprazole administration (p < 0.01) during the acute phase of MI. CONCLUSION: In patients with acute MI treated with PCI, lower dose-adjusted clopidogrel and dose-adjusted 2-oxo-clopidogrel plasma concentrations were associated with an increased risk of ischemic events.
.


Subject(s)
Clopidogrel/adverse effects , Clopidogrel/blood , Ischemia/chemically induced , Myocardial Infarction/drug therapy , Ticlopidine/adverse effects , Ticlopidine/blood , Humans , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/blood , Treatment Outcome
7.
Biomed Chromatogr ; 33(2): e4403, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30276833

ABSTRACT

Beraprost sodium is an oral prostacyclin analog that was first approved in 1992 (Japan) for the treatment of peripheral vascular disorders. It is administered orally as a tablet available in strength 20 µg. In this paper, we described a liquid chromatography tandem mass spectrometry method that was developed for the quantification of beraprost in human plasma with high sensitivity at picogram per milliliter concentration. The method had been validated in terms of selectivity, sensitivity, accuracy and precision, matrix effect, linearity, recovery and carry-over according to the Guideline on Bioanalytical Validation from the European Medicines Agency. The standard calibration curve for beraprost was 9.5-1419 pg/mL. This method has been applied successfully to a bioequivalence study with 60 µg of beraprost (three tablets) in 29 healthy volunteers. The results showed that the two formulations of beraprost are bioequivalent.


Subject(s)
Chromatography, Liquid/methods , Epoprostenol/analogs & derivatives , Tandem Mass Spectrometry/methods , Epoprostenol/blood , Epoprostenol/chemistry , Epoprostenol/pharmacokinetics , Humans , Linear Models , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Therapeutic Equivalency
8.
Luminescence ; 34(1): 64-69, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30537239

ABSTRACT

A spectrofluorimetric method for the determination of eptifibatide is presented based on its native fluorescence. The type of solvent and the wavelength of maximum excitation and emission were carefully selected to optimize the experimental conditions. Under the specified experimental conditions, the linearities obtained between the emission intensity and the corresponding concentrations of eptifibatide were in the range 0.1-2.5 µg/ml for the calibration curve constructed for direct determination of eptifibatide in dosage form and 0.05-2.2 µg/ml for the calibration curve constructed in spiked human plasma with a good correlation coefficient (r > 0.99). The lower limit of quantification for the calibration curve constructed in human plasma was 0.05 µg/ml. Recovery results for eptifibatide in spiked plasma samples and in dosage form, represented as mean ± % RSD, were 95.17 ± 1.94 and 100.29 ± 1.33 respectively. The suggested procedures were validated according to the International Conference on Harmonization (ICH) guidelines for the direct determination of eptifibatide in its pure form and dosage form and United States Food and Drug Administration (US FDA) Guidance for Industry, Bioanalytical Method Validation for the assay of eptifibatide in human plasma.


Subject(s)
Eptifibatide/analysis , Spectrometry, Fluorescence/methods , Calibration , Drug Stability , Eptifibatide/blood , Humans , Limit of Detection , Platelet Aggregation Inhibitors/analysis , Platelet Aggregation Inhibitors/blood , Reproducibility of Results , Sensitivity and Specificity , Solvents/chemistry , Temperature
9.
Biochem Biophys Res Commun ; 503(3): 1740-1746, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30122318

ABSTRACT

Decrease of chloride concentration contributes to cardiovascular diseases, however, whether decrease of chloride concentration is involved in platelet activation remains elusive. In the present study, we found that ACI patients had lower serum chloride which would be rescued after Aspirin administration. ADP induced chloride concentration reduction in platelets. Blockade of chloride channel prevented ADP-induced platelet adhesion, activation and aggregation, however, decreasing the extracellular chloride concentration promoted ADP-induced platelet adhesion and activation. Decrease of the extracellular chloride concentration facilitated the inactivation of Src family kinase Lyn, which was not involved in PI3K/Akt phosphorylation. Nevertheless, low chloride concentration promoted the production of platelet cytosol Gαi2 subunit. This subunit prevents AC from converting ATP into cAMP, which therefore, inhibited the phosphorylation of PKA to promote platelet activation. In conclusion, decreased intracellular chloride promotes ADP induced platelet activation through the Gαi2/cAMP/PKA pathway instead of the Lyn/PI3K/Akt signal pathway.


Subject(s)
Adenosine Diphosphate/metabolism , Chlorides/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP/antagonists & inhibitors , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Chlorides/blood , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation Inhibitors/blood , Proto-Oncogene Proteins c-akt/metabolism , src-Family Kinases/metabolism
10.
Br J Clin Pharmacol ; 84(1): 88-96, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28921624

ABSTRACT

BACKGROUND: Pivotal clinical trials found that ticagrelor reduced ischaemic complications to a greater extent than clopidogrel, and also that the benefit gradually increased with the reduction in creatinine clearance. However, the underlying mechanisms remains poorly explored. METHODS: This was a single-centre, prospective, randomized clinical trial involving 60 hospitalized Adenosine Diphosphate (ADP) P2Y12 receptor inhibitor-naïve patients with chronic kidney disease (CKD) (estimated glomerular filtration rate <60 ml min-1 1.73 m-2 ) and non-ST-elevation acute coronary syndromes (NSTE-ACS). Eligible patients were randomly assigned in a 1:1 ratio to receive ticagrelor (180 mg loading dose, then followed by 90 mg twice daily) or clopidogrel (600 mg loading dose, then followed by 75 mg once daily). The primary endpoint was the P2Y12 reactive unit (PRU) value assessed by VerifyNow at 30 days. The plasma concentrations of ticagrelor and clopidogrel and their active metabolites were measured in the first 10 patients in each group at baseline, and at 1 h, 2 h, 4 h, 8 h, 12 h and 24 h after the loading dose. RESULTS: Baseline characteristics were well matched between the two groups. Our results indicated a markedly lower PRU in patients treated with ticagrelor vs. clopidogrel at 30 days (32.6 ± 11.29 vs. 203.7 ± 17.92; P < 0.001) as well as at 2 h, 8 h and 24 h after the loading dose (P < 0.001). Ticagrelor and its active metabolite AR-C124910XX showed a similar time to reach maximum concentration (Cmax ) of 8 h, with the maximum concentration (Cmax ) of 355 (242.50-522.00) ng ml-1 and 63.20 (50.80-85.15) ng ml-1 , respectively. Both clopidogrel and its active metabolite approached the Cmax at 2 h, with a similar Cmax of 8.67 (6.64-27.75) ng ml-1 vs. 8.53 (6.94-15.93) ng ml-1 . CONCLUSION: Ticagrelor showed much more potent platelet inhibition in comparison with clopidogrel in patients with CKD and NSTE-ACS.


Subject(s)
Acute Coronary Syndrome/drug therapy , Adenosine/analogs & derivatives , Purinergic P2Y Receptor Antagonists/pharmacology , Renal Insufficiency, Chronic/physiopathology , Ticlopidine/analogs & derivatives , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/complications , Adenosine/blood , Adenosine/pharmacology , Adenosine/therapeutic use , Aged , Clopidogrel , Creatinine/blood , Female , Glomerular Filtration Rate , Humans , Kidney/physiopathology , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests , Prospective Studies , Purinergic P2Y Receptor Antagonists/therapeutic use , Renal Elimination , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Ticagrelor , Ticlopidine/blood , Ticlopidine/pharmacology , Ticlopidine/therapeutic use , Treatment Outcome
11.
J Cardiovasc Pharmacol ; 72(5): 252-258, 2018 11.
Article in English | MEDLINE | ID: mdl-30358688

ABSTRACT

Vicagrel, a novel acetate analogue of clopidogrel, exerts more potent antiplatelet effect than clopidogrel in rodents. Relevant evidence indicated that aspirin and vicagrel are the drug substrate for carboxylesterase 2. Accordingly, it is deduced that concomitant use of aspirin could attenuate the bioactivation of and platelet response to vicagrel. To clarify whether there could be such an important drug-drug interaction, the differences in both the formation of vicagrel active metabolite H4 and the inhibition of adenosine diphosphate-induced platelet aggregation by vicagrel were measured and compared between mice treated with vicagrel alone or in combination with aspirin. The plasma H4 concentration was determined by liquid chromatography-tandem mass spectrometry, and the inhibition of platelet aggregation by vicagrel was assessed by whole-blood platelet aggregation. Compared with vicagrel (2.5 mg·kg) alone, concurrent use of aspirin (5, 10, or 20 mg·kg) significantly decreased systemic exposure of H4, an average of 38% and 41% decrease in Cmax and AUC0-∞ in mice when in combination with aspirin at 10 mg·kg, respectively. Furthermore, concomitant use of aspirin (10 mg·kg) and vicagrel (2.5 mg·kg) resulted in an average of 66% reduction in the inhibition of adenosine diphosphate-induced platelet aggregation by vicagrel. We conclude that aspirin significantly attenuates the formation of vicagrel active metabolite H4 and platelet response to vicagrel in mice, and that such an important drug-drug interaction would appear in clinical settings if vicagrel is taken with aspirin concomitantly when marketed in the future.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Phenylacetates/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thiophenes/pharmacology , Activation, Metabolic , Animals , Aspirin/metabolism , Blood Platelets/metabolism , Carboxylesterase , Carboxylic Ester Hydrolases/metabolism , Chromatography, Liquid , Drug Interactions , Male , Mice, Inbred C57BL , Phenylacetates/blood , Phenylacetates/pharmacokinetics , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Tandem Mass Spectrometry , Thiophenes/blood , Thiophenes/pharmacokinetics
12.
Eur J Clin Pharmacol ; 74(9): 1141-1148, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29850937

ABSTRACT

PURPOSE: This single-dose, randomized, open-label, parallel-group, and crossover study assessed pharmacokinetics (PK), pharmacodynamics (PD), and safety of ticagrelor in subjects on hemodialysis versus healthy subjects. METHODS: Hemodialysis subjects were randomized, receiving a single ticagrelor 90-mg dose 1 day post-hemodialysis or just before hemodialysis, with an intervening washout of ≥ 7 days. Healthy subjects (creatinine clearance ≥ 90 mL/min) received a single ticagrelor 90-mg dose. PK, PD (P2Y12 reaction units [PRU], inhibition of platelet aggregation [IPA]), and safety were evaluated. RESULTS: Twenty-seven subjects (14 hemodialysis, 13 healthy) received ticagrelor. The mean maximum plasma concentration (Cmax) and area under the plasma concentration curve from time zero to infinity (AUC0-∞) of ticagrelor were 598.4 ng/mL and 3256.1 ng·h/mL, respectively, in pre-hemodialysis subjects; 560.3 ng/mL and 3015.1 ng·h/mL, respectively, in post-hemodialysis subjects; and 370.8 ng/mL and 2188.8 ng·h/mL, respectively, in healthy subjects. Cmax and AUC0-∞ of AR-C124910XX, the active metabolite, were 152.3 ng/mL and 1144.2 ng·h/mL, respectively, in pre-hemodialysis subjects; 130.8 ng/mL and 1127.8 ng·h/mL, respectively, in post-hemodialysis subjects; and 111.7 ng/mL and 1000.4 ng·h/mL, respectively, in healthy subjects. Mean IPA time curves over 24 h post-dose were almost indistinguishable for all three treatments. The greatest reduction in mean PRU occurred approximately 2 h post-dose for all three treatments. No safety or tolerability issues were identified. CONCLUSION: Hemodialysis resulted in modestly higher exposure to ticagrelor and AR-C124910XX, with no clinically significant effect on PD or tolerability. Accordingly, no dose adjustment is required for hemodialysis patients. Timing of hemodialysis has little impact on ticagrelor PK, or the effect of ticagrelor on IPA.


Subject(s)
Adenosine/analogs & derivatives , Blood Platelets/drug effects , Kidney Diseases/therapy , Kidney/physiopathology , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Renal Dialysis , Adenosine/administration & dosage , Adenosine/blood , Adenosine/pharmacokinetics , Adult , Blood Platelets/metabolism , Cross-Over Studies , Female , Humans , Kidney Diseases/blood , Kidney Diseases/diagnosis , Kidney Diseases/physiopathology , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/blood , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/blood , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/drug effects , Ticagrelor , United States
13.
J Cardiothorac Vasc Anesth ; 32(1): 178-186, 2018 02.
Article in English | MEDLINE | ID: mdl-29107589

ABSTRACT

OBJECTIVE: The purpose of the study was to evaluate the impact of intravenous metamizole on platelet inhibition by aspirin in patients with coronary artery disease early after on-pump coronary artery bypass grafting (CABG). DESIGN: Prospective, single-blind, randomized trial. SETTING: Tertiary referal hospital. PARTICIPANTS: The study comprised 43 patients with multivessel coronary artery disease undergoing CABG. INTERVENTIONS: Patients were randomized to postoperative intravenous metamizole ± opioids (study group; n = 23) or opioids alone (control group; n = 20). Aspirin was withheld at least 7 days before the surgery and reinitiated (300 mg) immediately after the procedure prior to metamizole use, and continued daily thereafter (150 mg). Platelet function was evaluated using multielectrode impedance aggregometry (acid-induced platelet activation [ASPI] and collagen-induced platelet activation [COL] test), P-selectin expression and urinary 11-dehydro-thromboxane B2 (11-DTXB2) level at baseline, postoperative day (POD) 0, POD 1, POD 2, and POD 6. Residual platelet reactivity (RPR) was defined as ASPI test >400 AU*min. MEASUREMENTS AND MAIN RESULTS: In all study participants, postoperative ASPI test value moderately decreased (1058.2 v 966.6 AU*min, p = 0.047), urinary 11-DTXB2 level increased (923.4 v 4367.3 pg/mg, p < 0.001), and P-selectin expression and COL test value remained stable postprocedure. The decreases of ASPI (p = 0.146) and COL test (p = 0.642), and P-selectin expression (p = 0.318) did not differ between both groups. Patients in the control group had higher postoperative increase of urinary 11-DTXB2 level (p = 0.001). The prevalence of RPR was high and comparable between study and control groups (day 1, 95.6% v 100%, p = 0.535; day 6, 100% v 90%, p = 0.21). Multivariate analysis revealed that metamizole use did not predict the fluctuations of ASPI and COL test values and P-selectin expression, yet it independently predicted postoperative change of 11-DTXB2 level (b = -0.518, p = 0.001). CONCLUSIONS: Intravenous metamizole preceded by a loading dose of aspirin did not modify platelet response to aspirin in the postoperative period after CABG.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Coronary Artery Bypass/methods , Coronary Artery Disease/therapy , Dipyrone/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Aged , Anti-Inflammatory Agents, Non-Steroidal/blood , Aspirin/blood , Coronary Artery Bypass/adverse effects , Coronary Artery Disease/blood , Dipyrone/blood , Drug Interactions/physiology , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Platelet Activation/drug effects , Platelet Activation/physiology , Platelet Aggregation Inhibitors/blood , Prospective Studies , Single-Blind Method
14.
Appl Nurs Res ; 39: 241-243, 2018 02.
Article in English | MEDLINE | ID: mdl-29422165

ABSTRACT

This was a retrospective study to assess the effectiveness of Society of Thoracic Surgeons (STS) Guidelines in reducing resource utilization at Washington Adventist Hospital, Takoma Park, Maryland. Pre and post operative stay (days), platelets and other blood product use, along with hospital costs were compared in elective Cardiac Surgical patients receiving P2Y12 inhibitors in the year prior to and year following the implementation of the STS guidelines of preoperative monitoring of platelet inhibition with VerifyNow. Our results demonstrated a significant decrease in length of stay, and although there was no discernible impact on other blood product use, there was a meaningful reduction in platelet use following implementation of the VerifyNow PRUTest assay for routine use in directing the timing of cardiac surgical procedures in this patient population.


Subject(s)
Aftercare/economics , Aftercare/standards , Length of Stay/economics , Length of Stay/statistics & numerical data , Platelet Aggregation Inhibitors/blood , Preoperative Care/economics , Preoperative Care/standards , Adult , Aftercare/methods , Aftercare/statistics & numerical data , Aged , Aged, 80 and over , Female , Health Care Costs/statistics & numerical data , Humans , Male , Maryland , Middle Aged , Practice Guidelines as Topic , Predictive Value of Tests , Preoperative Care/methods , Preoperative Care/statistics & numerical data , Retrospective Studies
15.
Biol Pharm Bull ; 40(1): 88-96, 2017.
Article in English | MEDLINE | ID: mdl-28049954

ABSTRACT

Ticagrelor is a direct-acting P2Y12 receptor antagonist. It is rapidly absorbed and partly metabolized to the active metabolite AR-C124910XX by CYP3A4 and CYP3A5. Three genetic loci (SLCO1B1, CYP3A4, and UGT2B7) were reported to affect ticagrelor pharmacokinetics. This study aimed to investigate the possible effects of SLCO1B1 and CYP3A4/5 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of ticagrelor in healthy Chinese male volunteers. Eighteen healthy male volunteers who participated in pharmacogenetics study of ticagrelor were genotyped for SLCO1B1 rs113681054, SLCO1B1*5 (rs4149056), CYP3A4*1G (rs2242480), and CYP3A5*3 (rs776746). All subjects received a single 180 mg loading dose of ticagrelor and then series blood samples were collected from 0 to 48 h. Plasma concentrations of ticagrelor and AR-C124910XX were determined by the high performance liquid chromatography-tandem mass spectrometry method. Inhibition in platelet aggregation (IPA) was assessed and the area under the time-effect curve (AUEC) for the IPA was calculated as pharmacodynamic parameters. No significant difference in ticagrelor pharmacokinetics among genotypes of the two genes was observed. The AUEC did not differ significantly among genotypes of candidate single nucleotide polymorphisms (SNPs). Our data suggest that common genetic variants in SLCO1B1 and CYP3A4/5 may have no effect on the pharmacokinetics and pharmacodynamics of ticagrelor in healthy Chinese volunteers.


Subject(s)
Adenosine/analogs & derivatives , Cytochrome P-450 CYP3A/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Platelet Aggregation Inhibitors , Purinergic P2 Receptor Antagonists , Adenosine/blood , Adenosine/pharmacokinetics , Adenosine/pharmacology , Area Under Curve , Asian People/genetics , Genotype , Healthy Volunteers , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Purinergic P2 Receptor Antagonists/blood , Purinergic P2 Receptor Antagonists/pharmacokinetics , Purinergic P2 Receptor Antagonists/pharmacology , Ticagrelor
16.
Platelets ; 28(7): 706-711, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28150519

ABSTRACT

The pharmacokinetics after a 180-mg loading dose (LD) of ticagrelor has not been thoroughly investigated in NSTEMI patients. We aimed to compare the ticagrelor uptake and on-treatment platelet reactivity between non-ST-segment elevation myocardial infarction (NSTEMI) patients and a control group of patients with stable coronary artery disease (SCAD) undergoing elective percutaneous coronary intervention. We performed an observational, prospective, single-center study including 40 NSTEMI patients and 20 SCAD controls. Key exclusion criteria included ongoing opioid treatment. Both groups received a 180-mg ticagrelor LD, and blood samples were taken pre-dose and 1, 2, 3, 4, 5, and 6 hours post-LD. Plasma concentrations of ticagrelor and its active metabolite AR-C124910XX were determined by validated methods. Platelet aggregation was tested using ADP-induced multiple electrode aggregometry. The primary endpoint was the time to maximal ticagrelor concentration (Tmax). Clinical trial registration identifier number: NCT02292277. None of the pharmacokinetic variables differed significantly between the groups, including the Tmax of ticagrelor (2.0h [1.0-3.0] versus 2.0h [2.0-3.0], p = 0.393) and the active metabolite AR-C124910XX (3.0 [2.0-4.0] versus 3.0 [2.5-4.0], p = 0.289). High on-treatment platelet reactivity (HPR) was defined as > 46 aggregation units and was at one hour seen in 15% of the NSTEMI patients versus 10% of the controls (p = 1.0). At two hours post the 180-mg ticagrelor LD, 3% of the NSTEMI patients had HPR compared with none of the controls (p = 1.0). In conclusion, the uptake of ticagrelor was not significantly slower in NSTEMI patients not receiving opioids compared with the SCAD controls, leading to adequate onset of platelet inhibition in both groups.


Subject(s)
Adenosine/analogs & derivatives , Coronary Artery Disease/drug therapy , Non-ST Elevated Myocardial Infarction/drug therapy , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Adenosine/blood , Adenosine/pharmacokinetics , Adenosine Diphosphate/pharmacology , Aged , Biotransformation , Blood Platelets/drug effects , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Coronary Artery Disease/surgery , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Non-ST Elevated Myocardial Infarction/blood , Non-ST Elevated Myocardial Infarction/pathology , Non-ST Elevated Myocardial Infarction/surgery , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Prospective Studies , Purinergic P2Y Receptor Antagonists/blood , Sweden , Ticagrelor
17.
Int J Clin Pharmacol Ther ; 55(5): 416-424, 2017 May.
Article in English | MEDLINE | ID: mdl-28139972

ABSTRACT

OBJECTIVE: The population pharmacokinetics of ticagrelor and its active metabolite AR-C124910XX were characterized following ticagrelor 60 mg or 90 mg twice daily oral long-term treatment in 4,426 patients with a history of myocardial infarction. METHODS: The ticagrelor and AR-C124910XX plasma concentration-time data were described by one-compartment models with first-order absorption or metabolite formation and elimination. RESULTS: Systemic exposure to ticagrelor and AR-C124910XX were stable over time. Ticagrelor apparent clearance (CL/F) was 17 L/h for the 60-mg and 15.4 L/h for the 90-mg dose. The CL/F of AR-C124910XX was 11.1 L/h for the 60-mg and 9.95 L/h for the 90-mg dose. Both ticagrelor and AR-C124910XX CL/F were independently influenced by body weight, sex, age, smoking, and Japanese ethnicity. Female sex and age > 75 years were the only categorical covariates, having more than 20% effect on AR-C124910XX CL/F. Ticagrelor CL/F was 6% higher and 11% lower, whereas AR-C124910XX CL/F was 26% higher and 34% lower for patients weighing 110 and 50 kg, respectively, compared with an 83 kg patient. CONCLUSIONS: The small differences in exposure to both ticagrelor and AR-C124910XX between demographic subgroups were in accordance with the consistent efficacy and safety outcomes observed across the population. The results were similar to those observed previously in patients with acute coronary syndromes.
.


Subject(s)
Adenosine/analogs & derivatives , Models, Biological , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Activation, Metabolic , Adenosine/administration & dosage , Adenosine/blood , Adenosine/pharmacokinetics , Administration, Oral , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Gastrointestinal Absorption , Humans , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/blood , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/blood , Ticagrelor
18.
Int J Clin Pharmacol Ther ; 55(2): 177-185, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27936524

ABSTRACT

OBJECTIVE: Clopidogrel is a commonly used antiplatelet aggregation agent. Compared with the reference clopidogrel product, most commercially available generic clopidogrel products contain different crystalline forms of clopidogrel. This study was aimed to compare the pharmacodynamics of a commonly used generic clopidogrel product in Thailand with the reference clopidogrel product under steady state conditions. METHODS: A multiple-dose, randomized 2-way crossover study was conducted in 32 healthy male Thai volunteers. The subjects were assigned to receive 75 mg once daily of the test or the reference product for 7 days with a 2-week wash out period. Blood samples were collected on days 1, 5, 6, and 7 prior to drug administration and at 1, 2, 3, 4, 8, 12, and 24 hours after the last dose administered. The antiplatelet aggregation effects of clopidogrel were determined by using two different ex-vivo platelet aggregation tests including the whole blood impedance assay (WBA) and the VerifyNow® P2Y12 assay. Both pharmacodynamic parameters, the maximal antiplatelet effect (Emax) and the areas under the antiplatelet effect-time curve (AUEC0-24h), were calculated. RESULTS: Neither the mean values of Emax (90.70 ± 15.15 vs. 89.50 ± 10.71% inhibition) nor of AUEC0-24h (1,892.84 ± 657.22 vs. 1,853.58 ± 673.95% inhibition × h) under steady-state conditions obtained using the WBA method of these two clopidogrel products were significantly different. The results obtained using the VerifyNow® P2Y12 assay were consistent with those of the WBA assay. CONCLUSION: This study clearly demonstrated that ex-vivo antiplatelet aggregation effect under steady-state conditions of the test product was not significantly different from the reference product.
.


Subject(s)
Blood Platelets/drug effects , Drugs, Generic/pharmacokinetics , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/administration & dosage , Ticlopidine/analogs & derivatives , Adult , Area Under Curve , Asian People , Blood Platelets/metabolism , Clopidogrel , Cross-Over Studies , Drug Compounding , Drugs, Generic/administration & dosage , Healthy Volunteers , Humans , Male , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Purinergic P2Y Receptor Antagonists/blood , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/drug effects , Thailand , Therapeutic Equivalency , Ticlopidine/administration & dosage , Ticlopidine/blood , Ticlopidine/pharmacokinetics , Young Adult
19.
Eur J Anaesthesiol ; 34(5): 288-296, 2017 05.
Article in English | MEDLINE | ID: mdl-28030443

ABSTRACT

BACKGROUND: Nonopioid analgesic drugs may interfere with platelet inhibition by aspirin. Recent in vitro and clinical studies in patients with cardiovascular disease have suggested that this pharmacodynamic interaction may also occur with dipyrone, a nonopioid analgesic popular in Europe, Asia and South America. OBJECTIVE: Dipyrone is used extensively in acute and chronic pain. This study was undertaken to provide clinical data, so far missing, on its interactions in this group of patients. DESIGN: A case-control study. SETTING: Primary care in one European university hospital centre. PATIENTS: In total, 27 patients with stable cardiovascular, cerebrovascular or peripheral arterial disease and acute or chronic pain were identified and given dipyrone for at least 5 days in combination with low-dose aspirin. In total, 10 comparable patients on low-dose aspirin alone served as controls. MAIN OUTCOME MEASURES: Platelet-rich plasma was prepared to determine arachidonic acid-induced aggregation (aggregometry) and thromboxane formation (immunoassay). Platelet sensitivity to aspirin was examined in vitro. The presence of dipyrone (metabolites) in plasma was confirmed by HPLC. Additional in vitro measurements examined the aspirin/dipyrone interaction in healthy donors. RESULTS: Inhibition of aggregation was observed in only six of 27 patients receiving aspirin with dipyrone, with absence of complete inhibition by antiplatelet therapy showing in 78% of patients. In contrast, aggregation was completely inhibited in nine of 10 control patients (P < 0.001). Platelet thromboxane synthesis was higher in patients receiving dipyrone + aspirin compared with controls (387 ±â€Š89 vs. 7 ±â€Š1 ng ml, P < 0.001). Aspirin added in vitro failed to inhibit aggregation and thromboxane synthesis in platelet-rich plasma from dipyrone-treated patients. In vitro measurements with blood from healthy individuals confirmed that dipyrone dramatically reduces inhibition of platelet thromboxane synthesis by aspirin. CONCLUSIONS: Dipyrone given for 5 days or longer blunts platelet inhibition by low-dose aspirin in the majority of recipients. TRIAL REGISTRATION: German Clinical Trials Register: DRKS ID DRKS00000204. Universal Trial Number (UTN): U1111-1113-3946.


Subject(s)
Acute Pain/blood , Anti-Inflammatory Agents, Non-Steroidal/blood , Aspirin/blood , Chronic Pain/blood , Dipyrone/blood , Platelet Aggregation Inhibitors/blood , Acute Pain/drug therapy , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Blood Platelets/drug effects , Blood Platelets/metabolism , Chronic Pain/drug therapy , Dipyrone/administration & dosage , Drug Interactions/physiology , Female , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage
20.
Drug Metab Dispos ; 44(10): 1622-32, 2016 10.
Article in English | MEDLINE | ID: mdl-27457785

ABSTRACT

Clopidogrel is reported to be associated with cerivastatin-induced rhabdomyolysis, and clopidogrel and its metabolites are capable of inhibiting CYP2C8 and OATP 1B1 in vitro. The objective of the present study was to identify the mechanism of clopidogrel-mediated drug-drug interactions (DDIs) on the pharmacokinetics of OATP1B1 and/or CYP2C8 substrates in vivo. A clinical cassette small-dose study using OATPs, CYP2C8, and OATP1B1/CYP2C8 probe drugs (pitavastatin, pioglitazone, and repaglinide, respectively) with or without the coadministration of either 600 mg rifampicin (an inhibitor for OATPs), 200 mg trimethoprim (an inhibitor for CYP2C8), or 300 mg clopidogrel was performed, and the area under the concentration-time curve (AUC) ratios (AUCRs) for probe substrates were predicted using a static model. Clopidogrel increased the AUC of pioglitazone (2.0-fold) and repaglinide (3.1-fold) but did not significantly change the AUC of pitavastatin (1.1-fold). In addition, the AUC of pioglitazone M4, a CYP2C8-mediated metabolite of pioglitazone, was reduced to 70% of the control by coadministration of clopidogrel. The predicted AUCRs using the mechanism-based inhibition of CYP2C8 by clopidogrel acyl-ß-glucuronide were similar to the observed AUCRs, and the predicted AUCR (1.1) of repaglinide using only the inhibition of OATP1B1 did not reach the observed AUCR (3.1). In conclusion, a single 300 mg of clopidogrel mainly inhibits CYP2C8-mediated metabolism by clopidogrel acyl-ß-glucuronide, but its effect on the pharmacokinetics of OATP1B1 substrates is negligible. Clopidogrel is expected to have an effect not only on CYP2C8 substrates, but also dual CYP2C8/OATP1B1 substrates as seen in the case of repaglinide.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Ticlopidine/analogs & derivatives , Area Under Curve , Clopidogrel , Cytochrome P-450 CYP2C8/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Humans , In Vitro Techniques , Male , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Quinolines/pharmacology , Rifampin/pharmacology , Ticlopidine/blood , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology , Trimethoprim/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL