Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Thromb Haemost ; 16(6): 1089-1098, 2018 06.
Article in English | MEDLINE | ID: mdl-29582544

ABSTRACT

Essentials Successful outcome of platelet transfusion depends on specific antiplatelet therapy in use. We assessed if ticagrelor, clopidogrel or prasugrel impacts on donor platelet activity ex vivo. Ticagrelor and/or its active metabolite in plasma or bound to platelets can inhibit donor platelets. This might compromise the effectiveness of platelet transfusion therapy. SUMMARY: Background Platelet transfusion is the conventional approach to restore platelet function during acute bleeds or surgery, but successful outcome depends on the specific antiplatelet therapy. Notably ticagrelor is associated with inadequate recovery of platelet function after platelet transfusion. We examined whether plasma and/or platelets from ticagrelor-treated patients influence donor platelet function, in comparison with clopidogrel and prasugrel. Methods Platelet transfusion was mimicked ex vivo by mixing naïve donor platelet-rich plasma (PRP) or gel-filtered platelets (GFP) in defined proportions with PRP, plasma or GFP from cardiovascular patients receiving standard care including medication with prasugrel, clopidogrel or ticagrelor (n = 20 each). Blood was taken 4 h after the previous dose. HLA2/HLA28 haplotyping let us distinguish net (all platelet) and individual patient/donor platelet reactivity in mixtures of patient/donor platelets, measured by flow cytometry analysis of ADP-induced fibrinogen binding and CD62P expression. Results ADP responsiveness of donor platelets was dramatically reduced by even low (10%) concentrations of PRP or plasma from ticagrelor-treated patients. Clopidogrel and prasugrel were associated with more modest donor platelet inhibition. GFP from ticagrelor-treated patients but not patients receiving clopidogrel or prasugrel also suppressed donor GFP function upon mixing, suggesting the transfer of ticagrelor from patient platelets to donor platelets. This transfer did not lead to recovery of ADP responsiveness of patient's platelets. Conclusion Collectively, these observations support the concept that ticagrelor and/or its active metabolite in plasma or bound to platelets can inhibit donor platelets, which might compromise the effectiveness of platelet transfusion therapy.


Subject(s)
Blood Platelets/drug effects , Clopidogrel/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Platelet Transfusion , Platelet-Rich Plasma/drug effects , Prasugrel Hydrochloride/therapeutic use , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Purinergic P2Y12/drug effects , Ticagrelor/therapeutic use , Blood Platelets/metabolism , Clinical Decision-Making , Clopidogrel/adverse effects , Female , Humans , Male , Patient Selection , Platelet Aggregation Inhibitors/adverse effects , Platelet Function Tests , Platelet Transfusion/adverse effects , Prasugrel Hydrochloride/adverse effects , Purinergic P2Y Receptor Antagonists/adverse effects , Receptors, Purinergic P2Y12/blood , Risk Factors , Ticagrelor/adverse effects
3.
J Thromb Haemost ; 15(5): 858-867, 2017 05.
Article in English | MEDLINE | ID: mdl-28092426

ABSTRACT

Essentials Irreversible platelet inhibition persists after reversibly-binding ticagrelor is discontinued. Reversibility of platelet inhibition by ticagrelor and its active metabolite was assessed. Incomplete recovery was observed after prolonged exposure to ticagrelor. Activated GPIIb-IIIa and P-selectin, not platelet reactivity index, showed irreversibility. SUMMARY: Introduction Ticagrelor is described as a reversible P2Y12 antagonist. However, residual platelet inhibition persists after discontinuation of ticagrelor when plasma levels are undetectable. We assessed the reversibility of platelet inhibition by ticagrelor and its active metabolite (T-AM) in comparison with cangrelor and prasugrel's active metabolite (P-AM). Methods Whole blood was treated in vitro with ~ 50% inhibitory concentrations of ticagrelor, T-AM, cangrelor, P-AM and assessed for ADP-stimulated activated GPIIb-IIIa and P-selectin and vasodilator-stimulated phosphoprotein (VASP) platelet reactivity index (PRI) before and after 100-fold dilution. Results Platelets exposed for 30 min to ticagrelor, T-AM or cangrelor showed full recovery of activated GPIIb-IIIa but only partial recovery of P-selectin. Longer exposure (24 h) to the drug decreased reversibility of activated GPIIb-IIIa by ticagrelor (65.1% [49.5-80.6], % of vehicle with 95% confidence interval [CI]) and T-AM (88.8% [79.2-98.3]), but not by cangrelor (101.4% [96.4-106.4]). Compared with 30 min exposure, the reversibility of P-selectin further decreased after 24 h exposure to ticagrelor (from 91.8% [82.1-101.5] to 51.8% [45.5-85.0]), but not T-AM (from 79.0% [67.8-90.3] to 77.4% [61.8-93.1]) or cangrelor (from 76.0% [67.6-84.4] to 76.2% [70.6-81.8]). In contrast, 24 h exposure to ticagrelor, T-AM and cangrelor resulted in full recovery of platelet reactivity as measured by PRI. Platelets exposed to P-AM showed no recovery of ADP reactivity. Conclusions Incomplete recovery after prolonged exposure to ticagrelor, observed by activated GPIIb-IIIa and P-selectin but not upstream VASP signaling, suggests that P2Y12 regains functionality and irreversible changes occur independent of VASP signaling.


Subject(s)
Adenosine/analogs & derivatives , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Adenosine/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Blood Platelets/metabolism , Cell Adhesion Molecules/blood , Dose-Response Relationship, Drug , Humans , Kinetics , Microfilament Proteins/blood , P-Selectin/blood , Phosphoproteins/blood , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Prasugrel Hydrochloride/pharmacology , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/drug effects , Signal Transduction/drug effects , Ticagrelor
4.
TH Open ; 1(2): e92-e100, 2017 Jul.
Article in English | MEDLINE | ID: mdl-31249914

ABSTRACT

Patients with acute coronary syndrome with diabetes mellitus (DM) exhibit an impaired platelet inhibitory response to clopidogrel which is only partially understood. DM was induced by the administration of streptozotocin (STZ) to 9-week-old mice. The antithrombotic effects of clopidogrel (10 mg/kg/d, orally × 5 days) were determined using a FeCl 3 -induced thrombosis model employing wild-type (WT), apolipoprotein E (apoE)-deficient, and diabetic apoE-deficient mice at 21 weeks. Antiplatelet effects were determined using flow cytometry. The antithrombotic effects of clopidogrel were similar in WT and apoE-deficient mice but were attenuated in diabetic apoE-deficient mice with the percent inhibition of thrombus area (µm 2 ) by clopidogrel being 85.5% (WT mice), 75.0% (apoE-deficient mice), and 1.9% (diabetic apoE-deficient mice). The time to first occlusion and lumen stenosis also reflected a significant loss of the antithrombotic effects of clopidogrel in diabetic apoE-deficient mice. Ex vivo platelet activation, which was assessed using ADP-induced expression of activated glycoprotein IIb/IIIa, was completely inhibited by clopidogrel in these three groups of mice. In contrast, the effect of clopidogrel on the ex vivo expression of platelet P-selectin induced by protease-activated receptor 4-activating peptide was diminished in diabetic apoE-deficient mice compared with that in WT and apoE-deficient mice. These data suggest that diabetic apoE-deficient mice may serve as a useful model to better understand the impaired responses to clopidogrel in patients with DM, which may partially reflect a reduction of the effect of clopidogrel on thrombin-induced platelet activation.

5.
Thromb Haemost ; 112(3): 589-97, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25008027

ABSTRACT

CYP2C19 genotype has been shown to impact response to clopidogrel 75-mg but not prasugrel 10-mg. Here, we assessed effects of CYP2C19 metaboliser status on pharmacokinetics (PK) and pharmacodynamic (PD) responses to prasugrel 5-mg and 10-mg and clopidogrel 75-mg using data from two PK/PD studies in stable coronary artery disease (CAD) patients (GENERATIONS and FEATHER). Active metabolite concentrations (area under the curve, AUC[0-tlast]), maximum platelet aggregation (MPA) measured by light transmission aggregometry, vasodilator-stimulated phosphoprotein platelet reactivity index, and VerifyNow P2Y12-platelet reaction units (VN-PRU) were analysed by CYP2C19-predicted phenotype (extensive metaboliser [EM; N=154], *2-*8 non-carriers, vs reduced metaboliser [RM; N=41],*2-*8 carriers/*17 non-carriers). AUC(0-tlast) was unaffected by metaboliser status for prasugrel 5-mg and 10-mg (geometric mean EM/RM ratios 1.00, 95% confidence interval [CI]: 0.86,1.17, p>0.99; and 0.97, 95% CI:0.85,1.12, p=0.71, respectively), but was lower among RMs receiving clopidogrel 75-mg (1.37, 95% CI:1.14,1.65, p<0.001). Platelet reactivity was not significantly affected by CYP2C19 metaboliser status for prasugrel 5-mg, or for prasugrel 10-mg by MPA and VN-PRU, but for clopidogrel 75-mg was significantly higher in reduced metabolisers (all measures p<0.01). Prasugrel 10-mg showed greater antiplatelet effects vs clopidogrel 75-mg (all comparisons p<0.001). Prasugrel 5-mg showed greater antiplatelet effects vs clopidogrel 75-mg in RMs (all p<0.001), and comparable effects in EMs (all p≥0.37). In contrast to clopidogrel, prasugrel active metabolite PK was not influenced by CYP2C19 genotype. Antiplatelet effect for prasugrel 10-mg was greater irrespective of metaboliser status and for prasugrel 5-mg was greater for RMs and comparable for EMs as compared to clopidogrel 75-mg.


Subject(s)
Coronary Artery Disease/drug therapy , Cytochrome P-450 CYP2C19/genetics , Piperazines/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Thiophenes/pharmacokinetics , Ticlopidine/analogs & derivatives , Aged , Clinical Protocols , Clopidogrel , Coronary Artery Disease/genetics , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Piperazines/administration & dosage , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Aggregation Inhibitors/administration & dosage , Polymorphism, Genetic , Prasugrel Hydrochloride , Thiophenes/administration & dosage , Ticlopidine/administration & dosage , Ticlopidine/pharmacokinetics , Treatment Outcome
6.
Thromb Haemost ; 112(2): 311-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24718367

ABSTRACT

High on-treatment platelet reactivity (HPR) has been identified as an independent risk factor for ischaemic events. The randomised, double-blind, TRIPLET trial included a pre-defined comparison of HPR in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI) following a placebo/600-mg clopidogrel loading dose (LD) immediately before a subsequent prasugrel 60-mg or 30-mg LD. Platelet reactivity was assessed using the VerifyNow® P2Y12 assay (P2Y12 Reaction Units, PRU) within 24 hours (h) following the placebo/clopidogrel LD (immediately prior to prasugrel LD), and at 2, 6, 24, 72 h following prasugrel LDs. The impact of CYP2C19 predicted metaboliser phenotype (extensive metabolisers [EM] and reduced metabolisers [RM]) on HPR status was also assessed. HPR (PRU ≥240) following the clopidogrel LD (prior to the prasugrel LD) was 58.5% in the combined clopidogrel LD groups. No significant difference was noted when stratified by time between the clopidogrel and prasugrel LDs (≤6 hs vs>6 h). At 6 h following the 2nd loading dose in the combined prasugrel LD groups, HPR was 7.1%, with 0% HPR by 72 h. There was no significant effect of CYP2C19 genotype on pharmacodynamic (PD) response following either prasugrel LD treatments at any time point, regardless of whether it was preceded by a clopidogrel 600-mg LD. In conclusion, in this study, patients with ACS intended for PCI showed a high prevalence of HPR after clopidogrel 600-mg LD regardless of metaboliser status. When prasugrel LD was added, HPR decreased substantially by 6 h, and was not seen by 72 h.


Subject(s)
Acute Coronary Syndrome/therapy , Blood Platelets/drug effects , Drug Substitution , Percutaneous Coronary Intervention , Piperazines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Thiophenes/administration & dosage , Ticlopidine/analogs & derivatives , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Aged , Blood Platelets/metabolism , Clopidogrel , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Double-Blind Method , Drug Administration Schedule , Drug Resistance , Female , Genotype , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Phenotype , Piperazines/adverse effects , Piperazines/metabolism , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/metabolism , Platelet Function Tests , Prasugrel Hydrochloride , Thiophenes/adverse effects , Thiophenes/metabolism , Ticlopidine/administration & dosage , Ticlopidine/adverse effects , Ticlopidine/metabolism , Time Factors , Treatment Outcome
7.
J Thromb Haemost ; 12(4): 560-3, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24428754

ABSTRACT

BACKGROUND: On-treatment platelet reactivity (OTR) is a predictor of clinical outcomes in patients receiving thienopyridine therapy. OBJECTIVE: To assess whether point-of-care platelet reactivity testing can discriminate between patients who have and have not received a thienopyridine. PATIENTS/METHODS: This was an analysis of a randomized, multicenter, pharmacodynamic trial. Subjects with coronary artery disease treated with aspirin were randomly assigned to clopidogrel 75 mg daily or prasugrel 10 mg daily for 7 days. Platelet reactivity assessment with the VerifyNow P2Y12 test was performed before study drug admistration and 24 h after the final dose. Optimal cut-offs for a detectable drug effect were identified by the use of receiver operating characteristic curve analysis. RESULTS: A total of 54 subjects were enrolled and completed the study. The c-statistic for the identification of a thienopyridine effect was highly significant (0.93, P < 0.001), including for the clopidogrel and prasugrel groups considered separately (P < 0.001 for both). The optimal cut-off was < 213 P2Y12 reaction units (PRU), which provided a sensitivity of 80% and a specificity of 98%. This cut-off provided a sensitivity of 58% and a specificity of 100% for a clopidogrel effect, and a sensitivity of 100% and specificity of 96% for a prasugrel effect. CONCLUSIONS: OTR of < 213 PRU is highly specific for exposure to either clopidogrel or prasugrel. This may be useful in the management of thienoypridine-treated patients who require surgery. Furthermore, this diagnostic cut-off is similar to levels of OTR that have been associated with ischemic events in thienopyridine-treated patients, supporting the contention that a lack of drug effect is the mechanistic basis for the prognostic relationship between OTR and clinical outcomes.


Subject(s)
Blood Platelets/cytology , Coronary Artery Disease/drug therapy , Piperazines/administration & dosage , Pyridines/administration & dosage , Thiophenes/administration & dosage , Ticlopidine/analogs & derivatives , Aged , Aspirin/therapeutic use , Blood Platelets/metabolism , Clopidogrel , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Platelet Activation , Point-of-Care Systems , Prasugrel Hydrochloride , Pyridines/chemistry , ROC Curve , Receptors, Purinergic P2Y12/metabolism , Reproducibility of Results , Sensitivity and Specificity , Ticlopidine/administration & dosage , Time Factors , Treatment Outcome
8.
J Thromb Haemost ; 11(1): 100-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23137352

ABSTRACT

BACKGROUND: Hemostatic benefits of platelet transfusions in thienopyridine-treated acute coronary syndrome (ACS) patients may be compromised by residual metabolite in circulation. OBJECTIVES: To estimate the earliest time after a prasugrel loading-dose when added platelets are no longer inhibited by prasugrel's active metabolite. METHODS: Baseline platelet reactivity of healthy subjects (n=25, 30 ± 5 years, 68% male) on ASA 325 mg was tested using maximum platelet aggregation (MPA, ADP 20 µm) and VerifyNow(®) P2Y12 and was followed by a 60 mg prasugrel loading-dose. At 2, 6, 12 and 24 h post-dose, fresh concentrated platelets from untreated donors were added ex-vivo to subjects' blood, raising platelet counts by 0% (control), 40%, 60% and 80%. To estimate the earliest time when prasugrel's active metabolite's inhibitory effect on the added platelets ceases, platelet function in supplemented samples was compared across time-points to identify the time when effect of supplementation on platelet function stabilized (i.e. the increase in platelet reactivity was statistically similar to that at the next time-point). RESULTS: Supplemented samples showed concentration-dependent increases in platelet reactivity vs. respective controls by both MPA and VerifyNow(®) at all assessment time-points. For each supplementation level, platelet reactivity showed a sharp increase from 2 to 6 h but was stable (P=NS) between 6 and 12 h. CONCLUSIONS: The earliest measured time when supplemented platelets were not inhibited by circulating active metabolite of prasugrel was 6 h after a prasugrel loading-dose. These findings may have important implications for prasugrel-treated ACS patients requiring platelet transfusions during surgery.


Subject(s)
Acute Coronary Syndrome/therapy , Blood Platelets/drug effects , Piperazines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Platelet Transfusion , Thiophenes/administration & dosage , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/drug therapy , Adult , Aspirin/administration & dosage , Biotransformation , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hemostasis/drug effects , Humans , Male , Piperazines/blood , Piperazines/pharmacokinetics , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Count , Prasugrel Hydrochloride , Prospective Studies , Thiophenes/blood , Thiophenes/pharmacokinetics
9.
J Thromb Haemost ; 9(3): 552-61, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21143373

ABSTRACT

BACKGROUND: Aspirin and antagonists of platelet ADP P2Y(12) receptors are often coprescribed for protection against thrombotic events. However, blockade of platelet P2Y(12) receptors can inhibit thromboxane A(2) (TXA(2))-dependent pathways of platelet activation independently of aspirin. OBJECTIVES: To assess in vitro whether aspirin adds additional antiaggregatory effects to strong P2Y(12) receptor blockade. METHODS: With the use of platelet-rich plasma from healthy volunteers, determinations were made in 96-well plates of platelet aggregation, TXA(2) production and ADP/ATP release caused by ADP, arachidonic acid, collagen, epinephrine, TRAP-6 amide and U46619 (six concentrations of each) in the presence of prasugrel active metabolite (PAM; 0.1-10 µmol L(-1)), aspirin (30 µmol L(-1)), PAM + aspirin or vehicle. results: PAM concentration-dependently inhibited aggregation; for example, aggregation in response to all concentrations of ADP and U46619 was inhibited by ≥ 95% by PAM at > 3 µmol L(-1) . In further tests of PAM (3 µmol L(-1)), aspirin (30 µmol L(-1)) and PAM + aspirin, aspirin generally failed to produce more inhibition than PAM or additional inhibition to that caused by PAM. The antiaggregatory effects of PAM were associated with reductions in the platelet release of both TXA(2) and ATP + ADP. Similar effects were found when either citrate or lepirudin were used as anticoagulants, and when traditional light transmission aggregometry was conducted at low stirring speeds. CONCLUSIONS: P2Y(12) receptors are critical to the generation of irreversible aggregation through the TXA(2) -dependent pathway. As a result, strong P2Y(12) receptor blockade alone causes inhibition of platelet aggregation that is little enhanced by aspirin. The clinical relevance of these observations remains to be determined.


Subject(s)
Aspirin/administration & dosage , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/administration & dosage , Receptors, Purinergic P2Y12/blood , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/blood , Adenosine Triphosphate/blood , Adenosine Triphosphate/pharmacology , Arachidonic Acid/pharmacology , Collagen/pharmacology , Drug Synergism , Epinephrine/pharmacology , Humans , In Vitro Techniques , Peptide Fragments/pharmacology , Piperazines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Thromboxane A2/blood
10.
J Thromb Haemost ; 8(8): 1820-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20492465

ABSTRACT

BACKGROUND: The thienopyridine P2Y(12) receptor antagonist clopidogrel reduces the risk of arterial thrombosis and individual pharmacodynamic responses to clopidogrel are believed to reflect the levels of active metabolite (AM) generated. Rifampicin increases the inhibitory effect of clopidogrel on platelet aggregation (PA). We studied the response to clopidogrel before and during administration of rifampicin in order to study the relationship between individual AM levels and P2Y(12) blockade. METHODS: Healthy volunteers received a 600-mg loading dose of clopidogrel followed by 75 mg daily for 7 days and, after a washout period and treatment with rifampicin [300 mg twice a day (b.i.d.)], received the same regimen of clopidogrel. Clopidogrel AM levels were determined over 4 h after the clopidogrel loading dose and unblocked P2Y(12) receptor number was assessed using a (33) P-2MeSADP binding assay. PA was measured by optical aggregometry with ADP and TRAP. RESULTS: Rifampicin enhanced clopidogrel AM production [area-under-the-curve (AUC): clopidogrel 89±22 ng h mL(-1) , clopidogrel+rifampicin 335±86 ng h mL(-1) , P<0.0001], and P2Y(12) blockade (unblocked receptors: clopidogrel 48±24, clopidogrel+rifampicin 4±2, P<0.0001) and reduced PA (5 µmol L(-1) ADP: clopidogrel 20±4, clopidogrel+rifampicin 5±2, P<0.01). Increasing numbers of unblocked receptors were required for an aggregation response with a decreasing concentration of ADP. PA induced by ADP 2 µmol L(-1) was particularly sensitive to low levels of receptor blockade. CONCLUSION: Potentiation of clopidogrel AM production by rifampicin leads to greater P2Y(12) blockade and consequently greater inhibition of PA. PA responses to low concentrations of ADP are more sensitive to P2Y(12) blockade.


Subject(s)
Drug Synergism , Purinergic P2Y Receptor Antagonists/administration & dosage , Receptors, Purinergic P2Y12/chemistry , Rifampin/administration & dosage , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/chemistry , Adult , Area Under Curve , Blood Platelets/drug effects , Clopidogrel , Coronary Artery Disease/drug therapy , Drug Administration Schedule , Humans , Male , Middle Aged , Platelet Aggregation , Ticlopidine/administration & dosage
11.
Curr Med Res Opin ; 25(8): 1821-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19530977

ABSTRACT

OBJECTIVE: Prasugrel is a thienopyridine antiplatelet agent for the prevention of atherothrombotic events in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Since cytochrome P450 enzymes CYP3A4 and CYP2B6 play a major role in prasugrel's active metabolite formation, the effect of potent CYP induction by rifampin on the pharmacokinetics of prasugrel and on the pharmacodynamic response to prasugrel was evaluated in healthy male subjects. RESEARCH DESIGN AND METHODS: This was an open-label, two-period, fixed-sequence study conducted at a single clinical research center. In the first treatment period, subjects received prasugrel as an oral 60-mg loading dose (LD) on the first day followed by ten oral, 10-mg daily maintenance doses. After a 2-week washout period, subjects received oral rifampin alone (600 mg once daily) for 8 days, followed by coadministration of oral rifampin with prasugrel, given as a 60-mg LD on the first day followed by five daily 10-mg MDs. Blood collection for pharmacokinetic and pharmacodynamic analyses occurred after the LD and fifth MD of prasugrel in both periods. CLINICAL TRIAL SYNOPSIS: clinicalstudyresults.org ID #8976 RESULTS: Rifampin coadministration (600 mg daily) did not affect exposure to prasugrel's active metabolite (R-138727). However, at 2 and 4 h after the prasugrel loading dose (60 mg), rifampicin coadministration was associated with a 6-9 percentage point decrease (p < 0.01) in the magnitude of platelet inhibition; similarly, a 5-17 percentage point decrease (p < 0.05) was observed with rifampin coadministration during the prasugrel maintenance dose (10 mg) period. Post hoc in vitro experiments demonstrated a dose-dependent R-138727-rifampin interaction at the P2Y(12) level unrelated to enzyme induction. A limitation of this study is that while results of the in vitro post hoc study indicate a pharmacodynamic interaction with rifampin, the mechanism underlying this interaction has not been elucidated. CONCLUSIONS: Dose adjustment should not be necessary when prasugrel is administered with CYP inducers since formation of prasugrel's active metabolite is not affected by potent enzyme induction with rifampin.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Piperazines/pharmacology , Piperazines/pharmacokinetics , Rifampin/pharmacology , Thiophenes/pharmacology , Thiophenes/pharmacokinetics , Acute Coronary Syndrome , Adolescent , Adult , Antibiotics, Antitubercular/administration & dosage , Drug Interactions , Humans , Male , Middle Aged , Piperazines/administration & dosage , Prasugrel Hydrochloride , Rifampin/administration & dosage , Thiophenes/administration & dosage , Young Adult
12.
J Thromb Haemost ; 6(11): 1908-14, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18752581

ABSTRACT

BACKGROUND: The P2Y(12) receptor plays a crucial role in platelet aggregation and is the target of platelet aggregation inhibitors, including the thienopyridine compound prasugrel. OBJECTIVE: The present study analyzed the effects of R-138727 (2-[1-[2-cyclopropyl-1-(2-fluorophenyl)-2-oxoethyl]-4-mercapto-3-piperidinylidene]acetic acid), the active metabolite of prasugrel, on recombinant wild-type and mutant human P2Y(12) receptors in order to identify the molecular site of action of R-138727. METHODS: The function of wild-type and mutant P2Y(12) receptors stably expressed in Chinese hamster ovary cells was assessed by measuring the 2-methylthio-ADP-mediated inhibition of forskolin-stimulated cellular cAMP production. RESULTS: In cells expressing wild-type receptors, R-138727 potently inhibited receptor function with a half-maximal concentration below 1 microm. The mode of action was irreversible. The same effect of R-138727 was observed in cells expressing Cys17Ala/Cys270Ala constructs. In contrast, in cells expressing either a Cys97Ala construct or a Cys175Ala construct, R-138727 failed to inhibit the response to the agonist. When cells expressing wild-type receptors were pretreated with the P2 receptor antagonists ATP or suramin, no effect of R-138727 was observed. Similar experiments with N-acetylcysteine 10 microm showed no interference of N-acetylcysteine with R-138727. CONCLUSIONS: The experiments demonstrate a potent and irreversible action of R-138727 at the recombinant human P2Y(12) receptor. The data suggest that R-138727 interacts with cysteine 97 (upper portion of the predicted third transmembrane region) and cysteine 175 (second extracellular loop) of the receptor, which are likely to form a disulfide bridge in native receptors. Moreover, the data also suggest that this site of action of R-138727 is close to the ligand-binding site of the receptor.


Subject(s)
Cysteine/metabolism , Piperazines/metabolism , Receptors, Purinergic P2/metabolism , Thiophenes/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Humans , Platelet Aggregation Inhibitors , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Transduction, Genetic
13.
J Thromb Haemost ; 6(7): 1153-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18485086

ABSTRACT

BACKGROUND: Agents that act as antagonists at P2Y(12) ADP receptors on platelets are in use (clopidogrel), and in development for use (cangrelor and prasugrel), in patients with cardiovascular disease. Cangrelor is a direct-acting reversible antagonist being developed for short-term infusion; clopidogrel and prasugrel are oral prodrugs that provide irreversible inhibition via transient formation of active metabolites. At the cessation of cangrelor infusion, patients are likely to receive clopidogrel or prasugrel as a means of maintaining antiplatelet therapy. OBJECTIVES: To apply an experimental in vitro approach to investigate the possibility that cangrelor influences the ability of the active metabolites of clopidogrel and prasugrel to inhibit ADP-mediated platelet function. METHODS: The effects of cangrelor and the active metabolites of clopidogrel (C-AM) and prasugrel (P-AM) on platelet function were assessed by ADP-induced platelet P-selectin expression in whole blood. The method involved rapid removal of the antagonists by dilution, and measurement of residual platelet inhibition. RESULTS: Cangrelor, C-AM and P-AM markedly inhibited P-selectin expression. The effect of cangrelor, but not of C-AM and P-AM, was reversible following antagonist removal. Preincubation of blood with cangrelor prior to addition of C-AM or P-AM reduced the ability of metabolites to irreversibly antagonize P2Y(12). Irreversible inhibition was maintained when blood was preincubated with metabolites prior to cangrelor. CONCLUSIONS: Cangrelor influences the ability of the active metabolites of clopidogrel or prasugrel to inhibit platelet function irreversibly. Careful consideration should be given to the timing of administration of an oral P2Y(12) antagonist following cangrelor infusion.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Piperazines/pharmacology , Platelet Aggregation Inhibitors , Purinergic P2 Receptor Antagonists , Thiophenes/pharmacology , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Blood Platelets/drug effects , Cells, Cultured , Clopidogrel , Drug Interactions , Humans , Piperazines/metabolism , Platelet Function Tests , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Thiophenes/metabolism , Ticlopidine/metabolism , Ticlopidine/pharmacology
14.
J Thromb Haemost ; 6(2): 359-65, 2008 02.
Article in English | MEDLINE | ID: mdl-18021304

ABSTRACT

BACKGROUND: Prasugrel is a novel antiplatelet prodrug of the same thienopyridine class as clopidogrel and ticlopidine. Metabolism of prasugrel generates the active metabolite R-138727, an antagonist of the platelet P2Y(12) adenosine diphosphate (ADP) receptor, leading to inhibition of ADP-mediated platelet activation and aggregation. ADP also enhances the platelet response to collagen, and these two agonists contribute to the generation of platelet procoagulant activity. We therefore examined whether R-138727 inhibits ADP- and collagen-triggered platelet procoagulant activities. METHODS AND RESULTS: As shown by whole blood flow cytometry, R-138727 inhibited surface phosphatidylserine expression on ADP plus collagen-stimulated platelets and tissue factor (TF) expression on ADP-, collagen-, and ADP plus collagen-stimulated monocyte-platelet aggregates. R-138727 reduced monocyte-platelet aggregate formation, thereby further inhibiting TF expression. ADP, collagen, and ADP plus collagen accelerated the kinetics of thrombin generation in recalcified whole blood and R-138727 significantly inhibited this acceleration. Clot strength in a modified thromboelastograph system was also inhibited by R-138727 (IC50 0.7 +/- 0.1 microM). CONCLUSIONS: In addition to its previously known inhibitory effects on platelet activation and aggregation, the active metabolite of prasugrel, R-138727, inhibits platelet procoagulant activity in whole blood (as determined by phosphatidylserine expression on platelets and TF expression on monocyte-platelet aggregates), resulting in the functional consequences of delayed thrombin generation and impaired clot development.


Subject(s)
Blood Coagulation Factors/antagonists & inhibitors , Blood Platelets/drug effects , Clot Retraction/drug effects , Piperazines/pharmacology , Piperazines/pharmacokinetics , Platelet Activation/drug effects , Prodrugs/pharmacokinetics , Purinergic P2 Receptor Antagonists , Thiophenes/pharmacokinetics , Adenosine Diphosphate/pharmacology , Annexin A5/metabolism , Biotransformation , Collagen/pharmacology , Humans , Monocytes/metabolism , Phosphatidylserines/blood , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Thrombelastography , Thrombin/biosynthesis , Thromboplastin/biosynthesis
15.
J Thromb Haemost ; 5(7): 1545-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17456192

ABSTRACT

BACKGROUND AND METHODS: Prasugrel is a novel orally active thienopyridine prodrug with potent and long-lasting antiplatelet effects. Platelet inhibition reflects inhibition of P2Y(12) receptors by its active metabolite (AM). Previous studies have shown that the antiplatelet potency of prasugrel is at least 10 times higher than that of clopidogrel in rats and humans, but the mechanism of its higher potency has not yet been fully elucidated. RESULTS: Oral administration of prasugrel to rats resulted in dose-related and time-related inhibition of ex vivo platelet aggregation, and its effect was about 10 times more potent than that of clopidogrel. The plasma concentration of prasugrel AM was higher than that of clopidogrel AM despite tenfold higher doses of clopidogrel, indicating more efficient in vivo production of prasugrel AM than of clopidogrel AM. In rat platelets, prasugrel AM inhibited in vitro platelet aggregation induced by adenosine 5'-diphosphate (ADP) (10 microm) with an IC(50) value of 1.8 microm. Clopidogrel AM similarly inhibited platelet aggregation with an IC(50) value of 2.4 microm. Similar results were also observed for ADP-induced (10 microm) decreases in prostaglandin E(1)-stimulated rat platelet cAMP levels. These results indicate that both AMs have similar in vitro antiplatelet activities. CONCLUSIONS: The greater in vivo antiplatelet potency of prasugrel as compared to clopidogrel reflects more efficient in vivo generation of its AM, which demonstrates similar in vitro activity to clopidogrel AM.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Piperazines/blood , Piperazines/pharmacology , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacology , Thiophenes/blood , Thiophenes/pharmacology , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Alprostadil/pharmacology , Animals , Clopidogrel , Cyclic AMP/blood , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Male , Piperazines/administration & dosage , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Prasugrel Hydrochloride , Rats , Rats, Sprague-Dawley , Thiophenes/administration & dosage , Ticlopidine/administration & dosage , Ticlopidine/blood , Ticlopidine/pharmacology
16.
Clin Pharmacol Ther ; 81(5): 735-41, 2007 May.
Article in English | MEDLINE | ID: mdl-17361128

ABSTRACT

Prasugrel and clopidogrel inhibit platelet aggregation through active metabolite formation. Prasugrel's active metabolite (R-138727) is formed primarily by cytochrome P450 (CYP) 3A and CYP2B6, with roles for CYP2C9 and CYP2C19. Clopidogrel's activation involves two sequential steps by CYP3A, CYP1A2, CYP2C9, CYP2C19, and/or CYP2B6. In a randomized crossover study, healthy subjects received a loading dose (LD) of prasugrel (60 mg) or clopidogrel (300 mg), followed by five daily maintenance doses (MDs) (15 and 75 mg, respectively) with or without the potent CYP3A inhibitor ketoconazole (400 mg/day). Subjects had a 2-week washout between periods. Ketoconazole decreased R-138727 and clopidogrel active metabolite Cmax (maximum plasma concentration) 34-61% after prasugrel and clopidogrel dosing. Ketoconazole did not affect R-138727 exposure or prasugrel's inhibition of platelet aggregation (IPA). Ketoconazole decreased clopidogrel's active metabolite AUC0-24 (area under the concentration-time curve to 24 h postdose) 22% (LD) to 29% (MD) and reduced IPA 28% (LD) to 33% (MD). We conclude that CYP3A4 and CYP3A5 inhibition by ketoconazole affects formation of clopidogrel's but not prasugrel's active metabolite. The decreased formation of clopidogrel's active metabolite is associated with reduced IPA.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Piperazines/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Thiophenes/pharmacokinetics , Ticlopidine/analogs & derivatives , Adult , Area Under Curve , Clopidogrel , Cross-Over Studies , Double-Blind Method , Drug Interactions , Female , Humans , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Prasugrel Hydrochloride , Thiophenes/administration & dosage , Thiophenes/pharmacology , Ticlopidine/administration & dosage , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
17.
Bioorg Med Chem Lett ; 10(4): 385-9, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10714506

ABSTRACT

6-[4-Amidinobenzoyl]amino]-tetralone-2-acetic acid is a potent antagonist of GPIIb-IIIa. Substitution in the meta position of the benzamidine, or replacement with a heteroaryl amidine was tolerated in this series. Use of an acyl-linked 4-alkyl piperidine as an arginine isostere also provided active compounds. Compounds from this series provided substantial systemic exposure in the rat following oral administration.


Subject(s)
Acetates/metabolism , Amidines/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Tetralones , Adenosine Diphosphate/pharmacology , Animals , Arginine/chemistry , Benzamidines/chemistry , Biological Availability , Drug Evaluation , Enzyme-Linked Immunosorbent Assay , Fibrinogen/metabolism , Humans , Inhibitory Concentration 50 , Models, Molecular , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Protein Binding/drug effects , Protein Structure, Secondary , Rats , Stereoisomerism
18.
Arterioscler Thromb Vasc Biol ; 18(11): 1730-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9812911

ABSTRACT

Because the beta3-antagonist abciximab (c7E3 Fab) has significantly improved late outcomes after coronary angioplasty, the beta3 integrins have been implicated in the arterial response to injury. However, the mechanisms underlying this benefit are unknown. The observation that c7E3 binds beta3 integrins on vascular cells (alphavbeta3) with affinity equal to that for the platelet glycoprotein IIb/IIIa integrin has led to the hypothesis that c7E3 may act directly on the artery wall to prevent restenosis after angioplasty. To test this hypothesis, we studied the effects of c7E3 on structural changes within the artery wall after angioplasty or stent angioplasty in 23 male cynomolgus monkeys with established atherosclerosis. Animals were randomly assigned to receive either a bolus of c7E3 (0.4 mg/kg IV, n=11) followed by a 48-hour infusion (0. 2 microg. kg-1. min-1) or an equal volume of vehicle (n=12). Animals received weight-adjusted aspirin and heparin and then underwent unilateral iliac artery experimental angioplasty and subclavian artery stent angioplasty (Palmaz). Iliac artery lumen diameter (LD) was determined by angiography at baseline (LDPre), after angioplasty (LDPost), and 35 days later (LDDay35). Arteries were then fixed by perfusion and removed for analysis. Lumen, intima, media, and external elastic lamina (EEL) areas were measured in iliac artery cross sections. Values from each injured iliac artery were normalized to the contralateral uninjured iliac artery to control for interanimal variability in baseline artery size and atherosclerosis extent. Intimal area was also measured in subclavian stent cross sections. c7E3 blocked platelet aggregation and prolonged the bleeding time from 2.8+/-1.1 to 19.8+/-2.5 minutes, P<0.001. Experimental angioplasty increased LDPost an average of 28%, and the initial gain was similar in both groups (P=NS). Despite an anti-platelet effect, c7E3 did not inhibit iliac lumen narrowing (LDDay35-LDPost: c7E3, -0.69+/-0.17 versus vehicle, -0.99+/-.17 mm, P=0.35); intimal hyperplasia (neointima area: c7E3, 1.12+/-.28 versus vehicle, 1.22+/-.20 mm2, P=0.77); or decrease in artery wall size (EEL area [percent of uninjured control]: c7E3, 101+/-7% versus vehicle, 121+/-7%). Stent intimal hyperplasia was also unaltered by c7E3 treatment (neointimal area: c7E3, 1.09+/-0.16 versus vehicle, 1. 28+/-0.11 mm2, P=0.36). These results suggest that the benefits of c7E3 treatment in coronary angioplasty were not from inhibition of intimal hyperplasia or improved artery wall remodeling. Alternative mechanisms should be explored to explain improved late outcomes after angioplasty in patients treated with c7E3.


Subject(s)
Angioplasty, Balloon, Coronary , Antibodies, Monoclonal/therapeutic use , Arteriosclerosis/therapy , Immunoglobulin Fab Fragments/therapeutic use , Integrins/antagonists & inhibitors , Platelet Aggregation Inhibitors/therapeutic use , Abciximab , Animals , Arteries , Blood Coagulation/drug effects , Combined Modality Therapy , Drug Evaluation, Preclinical , Hematologic Tests , Hyperplasia/drug therapy , Lipids/blood , Macaca fascicularis , Male , Stents , Treatment Outcome
19.
Thromb Res ; 90(2): 95-100, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9684763

ABSTRACT

The ability of the F(ab')2 fragment of the murine monoclonal antibody 7E3 directed against the platelet glycoprotein IIb-IIIa receptor complex, to cause reperfusion of a totally occluding coronary artery thrombus was examined alone and in combination with aspirin and heparin in a canine model of coronary artery thrombosis. A localized thrombus was produced in the left circumflex coronary artery in open-chest dogs by electrolytic injury of the endothelium. Intravenous administration of a single injection of 5.0 mg/kg aspirin and heparin (80 U/kg bolus plus 30 U/kg/hr x 2 hr) maintained vessel patency for approximately 101 +/- 15 minutes. After vessels had been completely occluded for 5 minutes (in the presence of aspirin + heparin), a single intravenous injection of saline (10 ml) or 0.8 mg/kg 7E3 was administered. Reperfusion was observed in all dogs (6 of 6) receiving 7E3; 4 of 6 dogs maintained vessel patency throughout the course of the 2 hour observation period. Activated partial thromboplastin and thrombin times were elevated 1.4 and 9 fold, respectively, in groups that received heparin. Template bleeding times were significantly elevated in the groups receiving 7E3. In the control group, 2 of 5 dogs reperfused briefly, however neither were patent at the end of the observation period. A third group of 4 dogs which did not receive the aspirin + heparin regimen was allowed to occlude and 5 minutes later received a single intravenous injection of 0.8 mg/kg 7E3. None of the 4 dogs in this group reperfused at any time during the study. There were no significant differences between groups in regards to hematological or hemodynamic measurements during the experiment. We concluded from these findings that the monoclonal antibody, 7E3 can promote the dissolution of friable coronary artery thrombi that evolve during standard anticoagulant and antiplatelet therapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Coronary Thrombosis/therapy , Immunoglobulin Fab Fragments/administration & dosage , Myocardial Reperfusion/methods , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Abciximab , Acute Disease , Animals , Anticoagulants/administration & dosage , Aspirin/administration & dosage , Blood Coagulation/drug effects , Combined Modality Therapy , Coronary Circulation/drug effects , Coronary Thrombosis/drug therapy , Coronary Thrombosis/physiopathology , Disease Models, Animal , Dogs , Female , Heparin/administration & dosage , Injections, Intravenous , Male , Mice , Platelet Aggregation Inhibitors/administration & dosage
20.
J Clin Lab Anal ; 12(4): 191-6, 1998.
Article in English | MEDLINE | ID: mdl-9671169

ABSTRACT

Platelet-membrane surface receptors are important targets for pharmacologic intervention in cardiovascular disease. Among these, glycoprotein (GP) IIb-IIIa is dominant and integrally involved in platelet aggregation and thrombus formation. When activated, GPIIb-IIIa binds soluble fibrinogen (Fb) in a key, early step of this process. New drugs are under development that block Fb binding to GPIIb-IIIa and inhibit platelet aggregation. A thorough understanding of the relationship between circulating drug levels and the extent of GPIIb-IIIa receptor occupancy in humans is crucial for safe and efficacious use of these agents. Described here is the development of a new technique for measurement of GPIIb-IIIa receptor occupancy. In this assay, activated human platelets are incubated with biotinylated fibrinogen (Fb-biotin) followed by antibiotin-FITC.The extent of Fb binding is determined using flow cytometric analysis. Our results indicate that Fb-biotin binds rapidly to activated platelets and its detection is dependent on incubation temperature. Platelets that were pre-incubated with the GPIIb-IIIa antagonist echistatin were inhibited from binding Fb-biotin in a concentration-dependent manner. The fluorescence of processed samples was stable for two weeks in the cold. The assay described here is simple, cost effective, and can be adapted for use in clinical evaluations of these new drugs.


Subject(s)
Blood Platelets/drug effects , Peptides/blood , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Biotin/chemistry , Blood Platelets/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Flow Cytometry/methods , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Peptides/chemistry , Peptides/pharmacology , Spectrometry, Fluorescence , Time Factors , Viper Venoms/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...