Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Drug Metab Dispos ; 52(7): 654-661, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38729662

ABSTRACT

The delicate balance between ischemic and bleeding risks is a critical factor in antiplatelet therapy administration. Clopidogrel and prasugrel, belonging to the thienopyridine class of antiplatelet drugs, are known for their variability in individual responsiveness and high incidence of bleeding events, respectively. The present study is centered on the development and assessment of a range of deuterated thienopyridine derivatives, leveraging insights from structure-pharmacokinetic relationships of clopidogrel and prasugrel. Our approaches were grounded in the molecular framework of clopidogrel and incorporated the C2-pharmacophore design from prasugrel. The selection of ester or carbamate substituents at the C2-position facilitated the generation of the 2-oxointermediate through hydrolysis, akin to prasugrel, thereby bypassing the issue of CYP2C19 dependency. The bulky C2-pharmacophore in our approach distinguishes itself from prasugrel's acetyloxy substituent by exhibiting a moderated hydrolysis rate, resulting in a more gradual formation of the active metabolite. Excessive and rapid release of the active metabolite, believed to be linked with an elevated risk of bleeding, is thus mitigated. Our proposed structural modification retains the hydrolysis-sensitive methyl ester of clopidogrel but substitutes it with a deuterated methyl group, shown to effectively reduce metabolic deactivation. Three promising compounds demonstrated a pharmacokinetic profile similar to that of clopidogrel at four times the dose, while also augmenting its antiplatelet activity. SIGNIFICANCE STATEMENT: Inspired by the structure-pharmacokinetic relationship of clopidogrel and prasugrel, a range of clopidogrel derivatives were designed, synthesized, and assessed. Among them, three promising compounds have been identified, striking a delicate balance between efficacy and safety for antiplatelet therapy. Additionally, the ozagrel prodrug conjugate was discovered to exert a synergistic therapeutic effect alongside clopidogrel.


Subject(s)
Clopidogrel , Platelet Aggregation Inhibitors , Prasugrel Hydrochloride , Clopidogrel/pharmacokinetics , Clopidogrel/pharmacology , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/chemistry , Humans , Prasugrel Hydrochloride/pharmacokinetics , Prasugrel Hydrochloride/pharmacology , Cytochrome P-450 CYP2C19/metabolism , Structure-Activity Relationship , Activation, Metabolic , Male , Hydrolysis , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects
2.
Clin Pharmacol Drug Dev ; 12(1): 21-29, 2023 01.
Article in English | MEDLINE | ID: mdl-36251178

ABSTRACT

This study evaluated the pharmacokinetics and pharmacodynamics of the antiplatelet agent prasugrel, and explored its optimal dose regimens via modeling and simulation using NONMEM. We measured platelet aggregation and the serial plasma concentrations of the inactive (R-95913) and active metabolites (R-138727) of prasugrel after a single oral dose of 10-60 mg in 20 healthy adult male volunteers. A pharmacokinetic model for R-95913 and R-138727, and a pharmacodynamic model between the concentration of R-138727 and maximal platelet aggregation measured by light transmittance were constructed. The predictability of the model for platelet aggregation was evaluated by comparing the model prediction values with the observed ones not used in the construction of the model. Pharmacokinetic data were best described by a 3-compartment models for R-95913, a 1-compartment model for R-138727 with transit compartment model for absorption delay, and first-pass metabolic conversion of R-95913 into R-138727 during absorption. The association-dissociation model between R-138727 and its receptor in platelets was applied for the inhibitory effect of prasugrel on platelet aggregation. Prasugrel rapidly inhibited platelet aggregation after oral administration, with a prolonged duration of action; however, the concentration of the active metabolite decreased rapidly, which may have been due to the slow dissociation rate of R-138727 from its target receptor in platelets. The external validation suggests that our model could be used to individualize prasugrel treatment in various clinical situations. Simulation showed rapid onset of inhibitory effect with great magnitude and consistent inhibition after therapeutic dose of prasugrel.


Subject(s)
Platelet Aggregation Inhibitors , Platelet Aggregation , Adult , Male , Humans , Prasugrel Hydrochloride/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Healthy Volunteers
3.
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
4.
J Cardiovasc Med (Hagerstown) ; 22(9): 686-692, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34280175

ABSTRACT

AIM: To compare the degree of platelet inhibition between ticagrelor and prasugrel in patients undergoing percutaneous coronary intervention for acute coronary syndrome. METHODS: Platelet function was assessed by impedance aggregometry after 30-90 days of therapy with acetylsalicylic acid and ticagrelor and over 15 days after switching to prasugrel. High-on-treatment platelet reactivity (HRPR) was defined for ADP test results above the upper limit of normal. RESULTS: A total of 105 patients were included, 81.9% males and 33.3% people with diabetes, with a mean age of 60.8 ±â€Š8.1 years. Mean platelet reactivity was not significantly different between the two antiplatelet strategies, as the prevalence of HRPR (8.6 vs 12.3%, P = 0.50). Switching between the two antiplatelet agents was safe and well tolerated, and effectively reduced platelet reactivity in over 95% of the patients (only 3.8% of the study population displaying ineffective response to both drugs). CONCLUSION: Ticagrelor and prasugrel have a similar effect on platelet reactivity. Switching between the two drugs can be safely done.


Subject(s)
Acute Coronary Syndrome , Drug Substitution , Percutaneous Coronary Intervention/methods , Platelet Aggregation/drug effects , Prasugrel Hydrochloride , Ticagrelor , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/surgery , Cross-Over Studies , Drug Monitoring/methods , Drug Substitution/methods , Drug Substitution/statistics & numerical data , Female , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests/methods , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/pharmacokinetics , Ticagrelor/administration & dosage , Ticagrelor/pharmacokinetics
5.
CPT Pharmacometrics Syst Pharmacol ; 10(7): 735-747, 2021 07.
Article in English | MEDLINE | ID: mdl-33955698

ABSTRACT

Selatogrel is a potent and reversible P2Y12 receptor antagonist developed for subcutaneous self-administration by patients with suspected acute myocardial infarction. After single-dose emergency treatment with selatogrel, patients are switched to long-term treatment with oral P2Y12 receptor antagonists. Selatogrel shows rapid onset and offset of inhibition of platelet aggregation (IPA) to overcome the critical initial time after acute myocardial infarction. Long-term benefit is provided by oral P2Y12 receptor antagonists such as clopidogrel, prasugrel, and ticagrelor. A population pharmacokinetic (PK)/pharmacodynamic (PD) model based on data from 545 subjects in 4 phase I and 2 phase II studies well described the effect of selatogrel on IPA alone and in combination with clopidogrel, prasugrel, and ticagrelor. The PK of selatogrel were described by a three-compartment model. The PD model included a receptor-pool compartment to which all drugs can bind concurrently, reversibly or irreversibly, depending on their mode of action. Furthermore, ticagrelor and its active metabolite can bind to the selatogrel-receptor complex allosterically, releasing selatogrel from the binding site. The model provided a framework for predicting the effect on IPA of selatogrel followed by reversibly and irreversibly binding oral P2Y12 receptor antagonists for sustained effects. Determining the timepoint for switching from emergency to maintenance treatment is critical to achieve sufficient IPA at all times. Simulations based on the interaction model showed that loading doses of clopidogrel and prasugrel administered 15 h and 4.5 h after selatogrel, respectively, provide sustained IPA with clinically negligible drug interaction. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? Selatogrel is a potent reversible P2Y12 receptor antagonist developed for subcutaneous self-administration by patients in case of suspected acute myocardial infarction. Transition to oral P2Y12 receptor antagonists without drug interaction and sufficient inhibition of platelet aggregation must be assured at all times. WHAT QUESTION DID THIS STUDY ADDRESS? The pharmacokinetic/pharmacodynamic model semimechanistically describes the effect of selatogrel on platelet inhibition alone and in combination with the oral P2Y12 receptor antagonists clopidogrel, prasugrel, and ticagrelor. WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? Model-based simulations showed that loading doses of clopidogrel and prasugrel can be administered from 15 h and 4.5 h after selatogrel, respectively. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? These results support guiding the clinical transition from selatogrel emergency treatment to oral maintenance therapy in a safe and efficacious way.


Subject(s)
Models, Biological , Organophosphonates/administration & dosage , Purinergic P2Y Receptor Antagonists/administration & dosage , Pyrimidines/administration & dosage , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clopidogrel/administration & dosage , Clopidogrel/pharmacokinetics , Clopidogrel/pharmacology , Computer Simulation , Drug Interactions , Female , Humans , Male , Middle Aged , Organophosphonates/pharmacokinetics , Organophosphonates/pharmacology , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/pharmacokinetics , Prasugrel Hydrochloride/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Randomized Controlled Trials as Topic , Ticagrelor/administration & dosage , Ticagrelor/pharmacokinetics , Ticagrelor/pharmacology , Time Factors
6.
J Thromb Thrombolysis ; 51(3): 741-747, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33582955

ABSTRACT

Prasugrel and ticagrelor are potent oral platelet P2Y12 inhibitors and are recommended over clopidogrel in patients with acute coronary syndrome (ACS). Oral platelet P2Y12 inhibitors are characterized by varying degrees of pharmacodynamic response profiles as assessed by a variety of commercially available assays. Because of its ease of use, rapid turnaround times and ability to provide results specific to P2Y12 inhibitory effects, VerifyNow has emerged as one of the most commonly utilized platelet function assays. However, reference ranges with VerifyNow have been reported mainly for clopidogrel and there has not yet been any study specifically conducted to provide the expected on treatment reference ranges following administration of prasugrel and ticagrelor. This was a prospective single center investigation conducted in 120 patients with ACS who were treated with prasugrel or ticagrelor as per standard of care. Patients who underwent percutaneous coronary interventions (PCI) were treated with a loading dose of prasugrel (60 mg) or ticagrelor (180 mg), and patients who were on maintenance therapy were taking prasugrel (10 mg qd or 5 mg qd) or ticagrelor (90 mg bid). Platelet function testing was performed using the VerifyNow™ PRUTest™. The overall range of PRUTest values was lower than that observed in studies of patients treated with clopidogrel. The use of a maintenance dose regimen had a wider range of PRUTest values compared to the use of a loading dose for both prasugrel (1-179 vs. 2-128) and ticagrelor (1-196 vs. 1-177). The average PRUTest values in patients on prasugrel and ticagrelor maintenance dosing were 20% and 9% higher those observed in patients treated with a loading dose. PRUTest results following loading dose administration were very similar between drugs, but were 20% higher with prasugrel compared with ticagrelor during maintenance dosing. This study establishes expected PRUTest ranges for patients taking loading and maintenance doses of prasugrel and ticagrelor.Clinical Trial Registration http://www.clinicaltrials.gov Unique Identifier: NCT04492423, registered July 2020 retrospectively registered.


Subject(s)
Myocardial Ischemia , Platelet Function Tests/methods , Point-of-Care Testing , Prasugrel Hydrochloride , Ticagrelor , Dose-Response Relationship, Drug , Drug Monitoring , Female , Humans , Male , Middle Aged , Myocardial Ischemia/blood , Myocardial Ischemia/diagnosis , Myocardial Ischemia/therapy , Outcome and Process Assessment, Health Care , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/methods , Platelet Aggregation/drug effects , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/pharmacokinetics , Prospective Studies , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Ticagrelor/administration & dosage , Ticagrelor/pharmacokinetics
7.
J Cardiovasc Transl Res ; 14(1): 110-119, 2021 02.
Article in English | MEDLINE | ID: mdl-32096064

ABSTRACT

Antithrombotic therapy is a critical component of the management of ST-elevation myocardial infarction (STEMI) patients treated with primary percutaneous coronary intervention (PCI). Rapid and profound inhibition of platelet reactivity has been shown to mitigate the ischemic risks and improve myocardial salvage. High residual platelet reactivity (HRPR) has been reported up to 4 or 6 h after loading dose of prasugrel or ticagrelor; therefore, multiple alternative strategies, including crushed or chewed oral tables or intravenous agents, have been investigated to provide a more rapid and sustained inhibition of platelet function and bridge the initial treatment gap. The FABOLUS FASTER is the first investigator-initiated, multicentre, open-label, prospective, randomized study to directly compare the pharmacodynamics effects of cangrelor, tirofiban, chewed or integer prasugrel. This study will add new insights in the management of antiplatelet therapy in patients with STEMI undergoing primary PCI and might be hypothesis-generating for future clinical trials in this field. The trial is registered on clinicaltrials.gov NCT02978040, and EudraCT 2017-001065-24.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Percutaneous Coronary Intervention , Prasugrel Hydrochloride/administration & dosage , ST Elevation Myocardial Infarction/therapy , Tirofiban/administration & dosage , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/pharmacokinetics , Aged , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Prospective Studies , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/pharmacology , ST Elevation Myocardial Infarction/metabolism , Tirofiban/pharmacokinetics
8.
JACC Cardiovasc Interv ; 13(5): 621-630, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32139220

ABSTRACT

OBJECTIVES: The aim of this study was to evaluate prospectively the clinical impact of routine transmission of CYP2C19 genotype in the management of acute ST-segment elevation myocardial infarction with primary percutaneous coronary intervention. BACKGROUND: Response to clopidogrel differs widely among patients, notably because of CYP2C19 genetic polymorphisms. METHODS: CYP2C19 genotype (6 alleles) was determined centrally and communicated within 4.1 ± 1.9 days of primary percutaneous coronary intervention in 1,445 patients with ST-segment elevation myocardial infarction recruited at 57 centers in France. CYP2C19 metabolic status was predicted from genotype and served to adjust thienopyridine treatment. The primary endpoint was differences in 12-month outcomes (death, myocardial infarction, and stent thrombosis) between patients with the wild-type genotype or gain-of-function allele (class 1, n = 1,118) and those with loss-of-function (LOF) alleles (class 2, n = 272) who received optimized thienopyridine treatment. RESULTS: Detection of LOF alleles resulted in adjustment of P2Y12 inhibition in 85% of patients, with significantly higher use of prasugrel or double-dose clopidogrel. The primary endpoint did not differ between class 1 and class 2 patients (3.31% vs. 3.04%, respectively; p = 0.82). In contrast, carriers of LOF alleles without treatment adjustment had significantly worse outcomes (15.6%; p < 0.05). Bleeding rates were not different between groups. CONCLUSIONS: In a real-world setting, a complete CYPC2C19 genotype can be mostly determined in <7 days using analysis of saliva deoxyribonucleic acid collected during the in-hospital phase among patients with ST-segment elevation myocardial infarction treated with primary percutaneous coronary intervention. Genotype information led to stronger platelet inhibition treatment in the vast majority of LOF allele carriers and to similar clinical outcomes as in patients carrying the wild-type genotype or gain-of-function allele. (Genotyping Infarct Patients to Adjust and Normalize Thienopyridine Treatment [GIANT]; NCT01134380).


Subject(s)
Clopidogrel/administration & dosage , Coronary Thrombosis/prevention & control , Cytochrome P-450 CYP2C19/genetics , Percutaneous Coronary Intervention , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/administration & dosage , Polymorphism, Genetic , Prasugrel Hydrochloride/administration & dosage , ST Elevation Myocardial Infarction/therapy , Aged , Clinical Decision-Making , Clopidogrel/adverse effects , Clopidogrel/pharmacokinetics , Coronary Thrombosis/etiology , Coronary Thrombosis/mortality , Cytochrome P-450 CYP2C19/metabolism , Drug Resistance , Female , France , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/mortality , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/pharmacokinetics , Precision Medicine , Prospective Studies , Recurrence , Risk Assessment , Risk Factors , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/mortality , Time Factors , Treatment Outcome
9.
Clin Pharmacokinet ; 59(5): 545-566, 2020 05.
Article in English | MEDLINE | ID: mdl-32056160

ABSTRACT

Coronary artery disease remains the major cause of mortality worldwide. Antiplatelet drugs such as acetylsalicylic acid and P2Y12 receptor antagonists are cornerstone treatments for the prevention of thrombotic events in patients with coronary artery disease. Clopidogrel has long been the gold standard but has major pharmacological limitations such as a slow onset and long duration of effect, as well as weak platelet inhibition with high inter-individual pharmacokinetic and pharmacodynamic variability. There has been a strong need to develop potent P2Y12 receptor antagonists with more favorable pharmacological properties. Prasugrel and ticagrelor are more potent and have a faster onset of action; however, they have shown an increased bleeding risk compared with clopidogrel. Cangrelor is highly potent and has a very rapid onset and offset of effect; however, its indication is limited to P2Y12 antagonist-naïve patients undergoing percutaneous coronary intervention. Two novel P2Y12 receptor antagonists are currently in clinical development, namely vicagrel and selatogrel. Vicagrel is an analog of clopidogrel with enhanced and more efficient formation of its active metabolite. Selatogrel is characterized by a rapid onset of action following subcutaneous administration and developed for early treatment of a suspected acute myocardial infarction. This review article describes the clinical pharmacology profile of marketed P2Y12 receptor antagonists and those under development focusing on pharmacokinetic, pharmacodynamic, and drug-drug interaction liability.


Subject(s)
Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Acute Coronary Syndrome , Clopidogrel/pharmacokinetics , Humans , Prasugrel Hydrochloride/pharmacokinetics , Ticagrelor/pharmacokinetics
10.
J Thromb Thrombolysis ; 49(1): 10-17, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31643039

ABSTRACT

Prasugrel, a novel P2Y12 receptor antagonist, has been shown to be more effective than clopidogrel for preventing cardiovascular events in patients with acute coronary syndromes undergoing percutaneous coronary intervention. We investigated the dose-response antiplatelet effects of prasugrel compared with clopidogrel in Japanese patients with non-cardioembolic stroke. The influence of cytochrome P450 (CYP) polymorphisms on the antiplatelet effects of both drugs was also compared. In this multicenter randomized active-control comparative study, patients were randomized to receive prasugrel 2.5 mg, 5 mg, or 7.5 mg (double blind) or clopidogrel 75 mg (open label) once daily for 14 days. The primary endpoint was inhibition of platelet aggregation (IPA) in response to adenosine diphosphate 20 µM within 8 h of study drug administration on day 14. Of the 66 patients randomized, data from 63 (prasugrel 2.5 mg, 5 mg, and 7.5 mg groups, n = 14, 16, and 18, respectively; clopidogrel group, n = 15) were used in the pharmacodynamic assessment. IPA (arithmetic mean ± SD) after prasugrel administration increased dose-dependently (33 ± 9%, 44 ± 11%, and 53 ± 14%, at 2.5 mg, 5 mg, and 7.5 mg, respectively) and was higher in these groups than after clopidogrel (23 ± 16%). In a subgroup of CYP2C19 intermediate metabolizers, IPA was higher in the prasugrel 5 mg and 7.5 mg groups than in the clopidogrel group. No death or serious adverse events were reported. Prasugrel was well tolerated at doses up to 7.5 mg/day and had antiplatelet effects higher than those of clopidogrel 75 mg/day. CYP2C19 polymorphisms may have reduced clopidogrel-induced IPA.


Subject(s)
Cytochrome P-450 CYP2C19/genetics , Platelet Aggregation Inhibitors , Platelet Aggregation/drug effects , Polymorphism, Genetic , Prasugrel Hydrochloride , Stroke , Adult , Aged , Clopidogrel/administration & dosage , Clopidogrel/pharmacokinetics , Cytochrome P-450 CYP2C19/administration & dosage , Cytochrome P-450 CYP2C19/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/pharmacokinetics , Stroke/drug therapy , Stroke/enzymology , Stroke/genetics
11.
Eur J Clin Pharmacol ; 76(2): 257-265, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31773191

ABSTRACT

PURPOSE: Use of oral antiplatelets (OAPs) is essential for preventing thrombotic events in patients with acute coronary syndrome (ACS). Effects of clopidogrel, prasugrel, and ticagrelor may be enhanced due to pharmacodynamic interactions, but as CYP substrates, they are prone to pharmacokinetic interactions too. The aim was to study polypharmacy in ACS patients following hospital discharge. METHODS: This observational drug utilization study linked patient-level data from nationwide registers. The study population consisted of adult ACS patients discharged from Finnish hospitals in 2009-2013. Logistic regression was used to model the probability of drug-drug interactions with odd ratios for predefined predictors such as age, gender, and ACS type. RESULTS: In the cohort of 54,416 ACS patients, 91% of those treated with OAP received clopidogrel. Of clopidogrel-treated patients, 12% purchased warfarin at least once while on clopidogrel treatment. Old age, male sex, ST-elevation myocardial infarction as index event, and a history of previous ACS events were associated with an increased risk of warfarin-OAP interaction (p < 0.001 for all). Ibuprofen, and serotonergic drugs tramadol, citalopram, and escitalopram were the next most common drugs causing pharmacodynamic interactions. In general, concomitant use of drugs known to cause pharmacokinetic interactions was rare, but both esomeprazole and omeprazole were prescribed in more than 6% of clopidogrel-treated patients. CONCLUSIONS: Warfarin and ibuprofen were the most commonly used concomitant medications causing pharmacodynamic interactions and potentially increasing the risk of bleeding in OAP-treated patients. Esomeprazole and omeprazole were used in clopidogrel-treated patients although there are alternatives available for gastric protection.


Subject(s)
Acute Coronary Syndrome/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Polypharmacy , Administration, Oral , Adult , Aged , Clopidogrel/administration & dosage , Clopidogrel/adverse effects , Clopidogrel/pharmacokinetics , Cohort Studies , Drug Interactions , Female , Finland , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Outpatients , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/pharmacokinetics , Retrospective Studies , Ticagrelor/administration & dosage , Ticagrelor/adverse effects , Ticagrelor/pharmacokinetics
13.
Am J Cardiovasc Drugs ; 20(3): 259-269, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31586336

ABSTRACT

INTRODUCTION: Real-life data comparing clopidogrel, prasugrel, and ticagrelor for unselected patients undergoing percutaneous coronary intervention (PCI) for acute coronary syndrome (ACS) are lacking, as are data for the temporal distribution of ischemic and bleeding risks. METHODS: A total of 19,825 patients were enrolled from the RENAMI and BleeMACS registries. Both were multicenter, retrospective, observational registries including the data and outcomes of consecutive patients with ACS who underwent primary PCI and were discharged with dual antiplatelet therapy (DAPT). We evaluated the long-term outcome stratified by the different antiplatelet agents. RESULTS: A total of 14,105 patients (71.2%) were treated with clopidogrel, 2364 patients (11.9%) with prasugrel and 3356 patients (16.9%) with ticagrelor. After propensity score matching, at 1 year, prasugrel reduced the incidence of net adverse clinical events (NACE; a composite endpoint of all-cause death, myocardial infarction [MI] and Bleeding Academic Research Consortium [BARC] 3-5 bleeding) (4.2% vs.7.6%, p = 0.002) and of major adverse cardiovascular events (MACE; a composite endpoint of death and MI) compared with clopidogrel (2.6% vs. 5.2%, p = 0.007). Ticagrelor decreased rates of MACE compared with clopidogrel (2.7% vs. 6.2%, p < 0.001), but not of NACE (6.6% vs. 8.7%, p = 0.07). Ticagrelor presented similar performance in terms of MACE compared with prasugrel (2.8% vs. 2.4%, p = 0.56), with a trend towards a reduction in MI (0.2% vs. 0.4%, p = 0.56), but with higher risk of BARC 3-5 bleedings (3.8% vs. 1.7%, p = 0.04). In the daily risk analysis, clopidogrel presented a binomial distribution with a peak of ischemic risk at 3 months, which decreased towards bleedings; prasugrel had a constant equivalence between opposite risks; and ticagrelor constantly reduced recurrent MIs despite higher risk of BARC 3-5 events. CONCLUSION: In real life, ticagrelor is more effective in reducing ischemic events during the first year after ACS, despite an increased risk of major bleedings, while prasugrel assures a better balance between ischemic and bleeding recurrent events.


Subject(s)
Acute Coronary Syndrome/surgery , Clopidogrel , Hemorrhage , Myocardial Infarction , Percutaneous Coronary Intervention , Prasugrel Hydrochloride , Ticagrelor , Acute Coronary Syndrome/epidemiology , Clopidogrel/administration & dosage , Clopidogrel/adverse effects , Clopidogrel/pharmacokinetics , Europe/epidemiology , Female , Hemorrhage/chemically induced , Hemorrhage/epidemiology , Humans , Male , Medication Therapy Management/statistics & numerical data , Middle Aged , Mortality , Myocardial Infarction/epidemiology , Myocardial Infarction/prevention & control , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/methods , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/pharmacokinetics , Registries/statistics & numerical data , Risk Adjustment/methods , Therapeutic Equivalency , Ticagrelor/administration & dosage , Ticagrelor/adverse effects , Ticagrelor/pharmacokinetics
15.
Clin Transl Sci ; 12(6): 564-572, 2019 11.
Article in English | MEDLINE | ID: mdl-31169344

ABSTRACT

In this paper, a special case for bioequivalence evaluation of oral formulations is discussed. Drug formulations with different forms of active moieties (e.g., free base and salt) may yield different dissolution characteristics and, thus, differ in absorption at elevated gastric pH. However, routine bioequivalence trials using subjects with normal gastric pH (i.e., ~ 1) may fail to identify these differences because dissolution/absorption profiles of the two formulations at normal gastric pH are similar. In the case of palbociclib, it is confirmedthat the free base and salt formulations showed different absorption in patients with different gastric pH. Significant reduction in drug absorption was observed only in patients with elevated gastric pH using free base formulation. The discovery that the free base had significantly reduced absorption hinged on the inclusion of enough patients with elevated gastric pH to detect a difference in a bioequivalence trial. This raises a concern, as demonstrated through simulation, that dissolution/absorption differences in other formulations could be missed in routine bioequivalence trials. Aside from differences in active pharmaceutical ingredients (APIs), other factors, such as changes in excipients or manufacturing methods, may also lead to exposure differences between formulations at elevated gastric pH. For formulations containing different forms of the same active moiety or the same API and showing different dissolution profiles at elevated pH (i.e., pH ~ 4-6.8), evaluation of bioequivalence with gastric pH modulators (e.g., a H2 blocker) in addition to routine bioequivalence assessments may help to ensure therapeutic equivalence in patients with elevated gastric pH.


Subject(s)
Antacids/administration & dosage , Chemistry, Pharmaceutical/methods , Drug Liberation , Equivalence Trials as Topic , Gastric Acid/metabolism , Research Design , Administration, Oral , Area Under Curve , Computer Simulation , Drug Compounding/methods , Excipients/chemistry , Gastrointestinal Absorption , Humans , Hydrogen-Ion Concentration , Models, Biological , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Prasugrel Hydrochloride/administration & dosage , Prasugrel Hydrochloride/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Therapeutic Equivalency
16.
Article in English | MEDLINE | ID: mdl-30936195

ABSTRACT

Antiplatelet drugs are widely utilized in the setting of primary stroke prevention, secondary stroke prevention, and neuroendovascular device-related stroke prevention. These medications are effective in general, although significant variability in drug activity exists between patients. Although this variation may be related in part to a multitude of factors, a growing body of evidence suggests that individual genotypes are a main contributor. The PharmGKB database was mined to prioritize genetic variants with potential clinical relevance for response to aspirin, clopidogrel, prasugrel, and ticagrelor. Although variants were reported for all drugs, the highest level of evidence was found in cytochrome P450 (CYP450) genotype variation related to clopidogrel response. Individual genetic influences have an impact on the pharmacodynamics of antiplatelet agents. Current clinical practice for stroke prevention is primarily empiric or guided by functional assays; however, there now exists a third potential pathway to base treatment decisions: genotype-guided treatment.


Subject(s)
Pharmacogenomic Variants , Platelet Aggregation Inhibitors/pharmacokinetics , Stroke/prevention & control , Clopidogrel/pharmacokinetics , Clopidogrel/therapeutic use , Cytochrome P-450 Enzyme System/genetics , Humans , Platelet Aggregation Inhibitors/therapeutic use , Prasugrel Hydrochloride/pharmacokinetics , Prasugrel Hydrochloride/therapeutic use , Precision Medicine , Stroke/genetics , Ticagrelor/pharmacokinetics , Ticagrelor/therapeutic use
18.
Rev. esp. cardiol. (Ed. impr.) ; 72(3): 215-223, mar. 2019. tab, graf
Article in Spanish | IBECS | ID: ibc-182643

ABSTRACT

Introducción y objetivos: La puntuación PARIS permite una estratificación combinada de los riesgos isquémico y hemorrágico de los pacientes con cardiopatía isquémica tratados con stent coronario y tratamiento antiagregante plaquetario doble (TAPD). Se desconoce su utilidad en pacientes con síndrome coronario agudo (SCA) tratados con ticagrelor o prasugrel. Se investiga este aspecto en un registro internacional. Métodos: Estudio retrospectivo multicéntrico con participación voluntaria de 11 centros de 6 países europeos. Se estudio ́a 4.310 pacientes con SCA dados de alta en TAPD con ticagrelor o prasugrel. Se definío evento isquémico como trombosis de stent o infarto de miocardio espontáneo, y evento hemorrágico según BARC (Bleeding Academic Research Consortium) tipo3 o 5. Se calculó la discriminación y la calibración para ambas vertientes de la puntuación PARIS (PARISisquémico y PARIShemorrágico). El beneficio neto isquémico-hemorrágico se obtuvo mediante la diferencia entre las probabilidades predichas de eventos isqueémicos y hemorrágicos. Resultados: Durante 17,2 ± 8,3 meses, hubo 80 eventos isquémicos (el 1,9% anual) y 66 eventos hemorrágicos (el 1,6% anual). PARISisquémico y PARIShemorrágico se asociaron con el riesgo de evetos isquémicos (sHR=1,27; IC95%, 1,16-1,39) y hemorrágicos (sHR = 1,14; IC95%, 1,01-1,30) respectivamente. La discriminación de eventos isquémicos fue discreta (índice C = 0,64) y la de eventos hemorrágicos, pobre (índice C= 0,56), con buena calibración para ambos. El beneficio neto isquémico-hemorrágico resultó negativo (más eventos hemorrágicos) en pacientes con alto riesgo hemorrágico y positivo (más eventos isquémicos) en pacientes con alto riesgo isquémico. Conclusiones: En pacientes con SCA tratados con TAPD conticagrelor o prasugrel, la escala PARIS ayuda a establecer un equilibrio apropiado del riesgo isquémico-hemorrágico


Introduction and objectives: The PARIS score allows combined stratification of ischemic and hemorrhagic risk in patients with ischemic heart disease treated with coronary stenting and dual antiplatelet therapy(DAPT). Its usefulness in patients with acute coronary syndrome (ACS) treated with ticagrelor or prasugrel is unknown. We investigated this issue in an international registry. Methods: Retrospective multicenter study with voluntary participation of 11 centers in 6 European countries. We studied 4310 patients with ACS discharged with DAPT with ticagrelor or prasugrel. Ischemic events were defined as stent thrombosis or spontaneous myocardial infarction, and hemorrhagic events as BARC (Bleeding Academic Research Consortium) type 3 or 5 bleeding. Discrimination and calibration were calculated for both PARIS scores (PARISischemic and PARIShemorrhagic). The ischemic-hemorrhagic net benefit was obtained by the difference between the predicted probabilities of ischemic and bleeding events. Results: During a period of 17.2 ± 8.3 months, there were 80 ischemic events(1.9% per year) and 66 bleeding events (1.6% per year). PARISischemic and PARIShemorrhagic scores were associated with a risk of ischemic events (sHR, 1.27; 95%CI, 1.16-1.39) and bleeding events (sHR, 1.14; 95%CI, 1.01-1.30), respectively. The discrimination for ischemic events was modest (Cindex = 0.64) and was suboptimal for hemorrhagic events (Cindex = 0.56), where as calibration was acceptable for both. The ischemic-hemorrhagic net benefit was negative (more hemorrhagic events) in patients at high hemorrhagic risk, and was positive (more ischemicevents) in patients at high ischemic risk. Conclusions: In patients with ACS treated with DAPT with ticagrelor or prasugrel, the PARIS model helps to properly evaluate the ischemic-hemorrhagic risk


Subject(s)
Humans , Acute Coronary Syndrome/drug therapy , Myocardial Ischemia/drug therapy , Ticagrelor/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Hemorrhage/drug therapy , Myocardial Infarction/drug therapy , Severity of Illness Index , Acute Coronary Syndrome/physiopathology , Retrospective Studies , Recurrence , Platelet Aggregation Inhibitors/therapeutic use , Myocardial Infarction/epidemiology
19.
Curr Vasc Pharmacol ; 17(1): 35-40, 2019.
Article in English | MEDLINE | ID: mdl-29412111

ABSTRACT

BACKGROUND: Various antiplatelet drugs are used following Acute Coronary Syndromes (ACS). Of them, adenosine diphosphate receptor P2Y12 inhibitors clopidogrel, prasugrel and ticagrelor are currently used for post-ACS long-term treatment. Although they act on the same receptor, they differ in pharmacodynamics and pharmacokinetics. Several enzymes and transporters involved in the metabolism of P2Y12 inhibitors show genetic variability with functional impact. This includes Pglycoprotein, carboxylesterase 1 and, most notably, CYP2C19 that is important in clopidogrel activation. Common gain-of-function or loss-of-function alleles of CYP2C19 gene are associated with lower or higher platelet reactivity that may impact clinical outcomes of clopidogrel treatment. Prasugrel is considered to be less dependent on CYP2C19 variability as it is also metabolized by other CYP450 isoforms. Some studies, however, showed the relevance of CYP2C19 variants for platelet reactivity during prasugrel treatment as well. Ticagrelor is metabolized mainly by CYP3A4, which does not show functionally relevant genetic variability. Its concentrations may be modified by the variants of Pglycoprotein gene ABCB1. While no substantial difference between the clinical efficacy of prasugrel and ticagrelor has been documented, both of them have been shown to be superior to clopidogrel in post-ACS treatment. This can be partially explained by lower variability at each step of their metabolism. It is probable that factors influencing the pharmacokinetics of both drugs, including genetic factors, may predict the clinical efficacy of antiplatelet treatment in personalized medicine. CONCLUSION: We summarize the pharmacokinetics and pharmacogenetics of P2Y12 inhibitors with respect to their clinical effects in post-myocardial infarction treatment.


Subject(s)
Blood Platelets/drug effects , Clopidogrel/therapeutic use , Myocardial Infarction/drug therapy , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/therapeutic use , Prasugrel Hydrochloride/therapeutic use , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Purinergic P2Y12/drug effects , Ticagrelor/therapeutic use , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biotransformation/genetics , Blood Platelets/metabolism , Clopidogrel/adverse effects , Clopidogrel/pharmacokinetics , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Drug Resistance/genetics , Humans , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/adverse effects , Prasugrel Hydrochloride/pharmacokinetics , Purinergic P2Y Receptor Antagonists/adverse effects , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Receptors, Purinergic P2Y12/blood , Ticagrelor/adverse effects , Ticagrelor/pharmacokinetics , Treatment Outcome
20.
Clin Pharmacol Ther ; 105(2): 448-457, 2019 02.
Article in English | MEDLINE | ID: mdl-29998574

ABSTRACT

CYP3A enzymes participate in the elimination of ticagrelor and the bioactivation of clopidogrel and prasugrel. We studied the effects of functional CYP3A genetic variants (CYP3A4*22; rs35599367 and CYP3A5*3; rs776746) on the pharmacokinetics and pharmacodynamics of ticagrelor, clopidogrel, and prasugrel. Six healthy volunteers with the CYP3A4*1/*22 and CYP3A5*3/*3 genotype (CYP3A4*22 carriers), eight with the CYP3A4*1/*1 and CYP3A5*1/*3 genotype (CYP3A5 expressors), and 11-13 with the CYP3A4*1/*1 and CYP3A5*3/*3 genotypes (controls) ingested single doses of ticagrelor, clopidogrel, and prasugrel on separate occasions. Ticagrelor area under the plasma concentration-time curve (AUC) was 89% (P = 0.004) higher in CYP3A4*22 carriers than in controls. CYP3A4*22 carriers also showed more pronounced platelet inhibition at 24 hours after ticagrelor ingestion than the controls (43% vs. 21%; P = 0.029). The CYP3A5 genotype did not affect ticagrelor pharmacokinetics. Neither CYP3A5 nor CYP3A4 genotypes significantly affected prasugrel or clopidogrel. In conclusion, the CYP3A4*22 allele markedly impairs ticagrelor elimination enhancing its antiplatelet effect.


Subject(s)
Clopidogrel/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Ticagrelor/pharmacokinetics , Activation, Metabolic/genetics , Adult , Area Under Curve , Clopidogrel/pharmacology , Female , Genetic Variation , Genotype , Healthy Volunteers , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Function Tests , Prasugrel Hydrochloride/pharmacology , Ticagrelor/pharmacology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...