1
|
Gulyás Z, Horváth Z, Hajtman L, Kovács A, Kohut L, Kósa I, Tóth-Zsámboki E, Kiss RG. High platelet adrenergic activity and concomitant activation of the pituitary/medullar axis as alarming laboratory parameters in ACS survivors-the STRESS-AMI study. Front Cardiovasc Med 2024; 11:1338066. [PMID: 38450368 PMCID: PMC10914969 DOI: 10.3389/fcvm.2024.1338066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Kinetics of stress-related biological parameters were determined in acute coronary syndrome (ACS) patients undergoing complex cardiovascular rehabilitation. Methods We determined platelet functionality in the absence/presence of a selective alpha-2 adrenergic receptor inhibitor, atipemazole parallel with salivary cortisol levels at enrolment, and at 3- and 12-months follow-up in 75 ACS patients with percutaneous coronary intervention. Results Pharmacological/non-pharmacological secondary prevention methods have been efficiently applied. Baseline aggregometry indicated platelet hyperactivity, decreasing gradually and being significantly reduced late, at 12 months (p < 0.05). Cortisol levels followed similar kinetics (p < 0.05). Baseline epinephrine-induced aggregations (EIA) significantly correlated with most of the other platelet agonists, even at subsequent time-points. Patients with upper-quartile EIA at enrolment (EIA-UQ) had significantly higher ADP- and collagen-induced aggregations at enrolment, at 3- and 12-months follow-up as well, indicating that high adrenergic response in the acute phase is accompanied by general platelet hyperactivity and predicts sustained platelet activation. In the EIA-UQ group higher cardiac biomarker release, elevated C-reactive protein and cortisol levels, and lower baseline left ventricular ejection fraction were detected.Atipemazole significantly reduced platelet aggregation induced by several platelet agonists, being most potent and comparable to full in vitro P2Y12 inhibition on collagen-induced aggregations (p < 0.05), indicating that catecholamines might serve as promt/long-term modulators of platelet function. Discussion Despite effective CCR programme and dual antiplatelet therapy, prolonged activation of sympathetic neuroendocrine system and general platelet hyperactivity can be detected up to one year in ACS patients with high adrenergic platelet activity. Moreover, initial high adrenergic activity is accompanied by clinical parameters associated to increased cardiovascular risk, therefore early identification of these patients might support complex optimal long-term therapy.
Collapse
Affiliation(s)
- Zalán Gulyás
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| | - Zsófia Horváth
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| | - László Hajtman
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| | - Andrea Kovács
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| | - László Kohut
- Cardiac Rehabilitation Institute, Central Hospital of Northern Pest—Military Hospital, Balatonfüred, Hungary
| | - István Kósa
- Cardiac Rehabilitation Institute, Central Hospital of Northern Pest—Military Hospital, Balatonfüred, Hungary
| | - Emese Tóth-Zsámboki
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| | - Róbert Gábor Kiss
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, Budapest, Hungary
| |
Collapse
|
2
|
Pontis A, Delavenne X, Verdier MC, Hodin S, Andriamaharo A, Gueret P, Nedelec-Gac F, Bachelot-Loza C, Gaussem P, Gouin-Thibault I. Impact of age on in vitro metabolism of clopidogrel: a potential explanation for high on-treatment platelet reactivity in the elderly? Res Pract Thromb Haemost 2023; 7:100014. [PMID: 36891520 PMCID: PMC9986101 DOI: 10.1016/j.rpth.2022.100014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/07/2022] [Indexed: 01/08/2023] Open
Abstract
Background High on-treatment platelet reactivity has been reported in 30% of patients on clopidogrel and 50% in elderly patients; however, little is known about the mechanisms of this biological resistance. One hypothesis is an age-related impaired hepatic metabolism of the prodrug clopidogrel, leading to a lower formation of its active metabolite (clopidogrel-AM). Objectives To compare the levels of clopidogrel-AM formed in vitro using "old" and "young" human liver microsomes (HLMs) and their consequences on platelet functions. Methods We developed an in vitro model using "old" (73.6 ± 2.3 years) and "young" (51.2 ± 8.5 years) HLMs, added to platelet-rich plasma from 21 healthy donors with or without clopidogrel (50 μM) and incubated at 37 °C for 30 (T30) and 45 minutes (T45). Clopidogrel-AM was quantified by liquid chromatography-mass spectrometry/mass spectrometry method. Platelet aggregation was performed by light transmission aggregometry. Results The generation of clopidogrel-AM increased over time and reached concentrations comparable with those reported in treated patients. At T30, mean clopidogrel-AM concentrations were significantly higher with "young" (8.56 μg/L; 95% CI, 5.87-11.24) than with "old" HLMs (7.64 μg/L; 95% CI, 5.14-10.14; P = .002); and at T45, 11.40 μg/L; 95% CI (7.57-15.22) vs 10.63 μg/L, 95% CI (7.10-14.15), P = .02 (n = 21). Despite a significant inhibition of platelet aggregation, no significant difference was found in light transmission aggregometry (adenosine diphosphate, 10 μM) after clopidogrel metabolism by "old" or "young" HLMs, probably because of low sensitivity of the method to small variations of clopidogrel-AM. Conclusion In this original model combining metabolic and functional approaches, less clopidogrel-AM was produced with HLMs from older patients. This provides support for a decreased CYP450 activity that may contribute to high on-treatment platelet reactivity in elderly patients.
Collapse
Affiliation(s)
- Adeline Pontis
- Laboratoire d'Hématologie, Pontchaillou University Hospital, Rennes, France
- INSERM, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, University of Rennes, Rennes, France
| | - Xavier Delavenne
- Laboratoire de Pharmacologie Toxicologie, Saint-Etienne University Hospital, Saint-Etienne, France
- INSERM, UMR_S1059, Université de Lyon, Saint-Etienne, France
| | - Marie-Clémence Verdier
- INSERM, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, University of Rennes, Rennes, France
- Laboratoire de Pharmacologie Clinique, Pontchaillou University Hospital, Rennes, France
| | - Sophie Hodin
- Laboratoire de Pharmacologie Toxicologie, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Annie Andriamaharo
- Laboratoire de Pharmacologie Clinique, Pontchaillou University Hospital, Rennes, France
| | - Pierre Gueret
- Laboratoire d'Hématologie, Pontchaillou University Hospital, Rennes, France
| | | | | | - Pascale Gaussem
- Université de Paris Cité, Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris, France
- Service d'Hématologie Biologique, AP-HP, Université de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Isabelle Gouin-Thibault
- Laboratoire d'Hématologie, Pontchaillou University Hospital, Rennes, France
- INSERM, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, University of Rennes, Rennes, France
- Correspondence Isabelle Gouin-Thibault, Laboratoire d'Hématologie, Pontchaillou University Hospital, INSERM, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, University of Rennes, 35000, Rennes, France.
| |
Collapse
|
3
|
Aluvilu A, Ferro A. Role of platelet function testing in acute coronary syndromes: a meta-analysis. Open Heart 2022; 9:e002129. [PMID: 36581378 PMCID: PMC9806016 DOI: 10.1136/openhrt-2022-002129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE This meta-analysis aimed to evaluate whether using platelet function testing (PFT) in acute coronary syndromes (ACS) to personalise antiplatelet therapy including a P2Y12 antagonist offers any clinical benefits to indicate incorporation into routine practice. METHODS A search was conducted on five databases for randomised controlled trials (RCTs) conducted between 1 January 2000 and 17 July 2022, which included an ADP-specific platelet function assays and P2Y12 antagonists as part of dual antiplatelet therapy (DAPT) and have reported the efficacy and/or safety outcomes. The reported event frequencies were used to calculate the risk ratios (RRs) with a 95% CI. The χ2 heterogeneity statistical test and sensitivity analysis were used for heterogeneity assessment. RESULTS Five RCTs with 7691 patients were included in the analysis. No significant risk reduction was seen in major adverse cardiovascular events (RR=0.95, p=0.42), individual cardiac events (cardiovascular death: RR=0.76, p=0.26; myocardial infarction: RR=0.96, p=0.74; stent thrombosis: RR=0.92, p=0.83; stroke: RR=0.91, p=0.72; target vessel revascularisation: RR=1.06, p=0.47) and overall clinical outcome (RR=0.90, p=0.22). There was also no difference in the rate of bleeding between PFT-guided and standard therapies (major bleeding: RR=0.97, p=0.78, minor bleeding: RR=0.89, p=0.19 and any bleeding: RR=1.04, p=0.33). CONCLUSION Compared with standard DAPT with P2Y12 antagonists, using PFT to adjust antiplatelet therapy does not improve clinical outcomes. Therefore, the positions of key guidelines on routine testing in ACS should remain unchanged. In addition, the study highlights the need for well-designed and powered RCTs and standardised testing methodologies to provide reliable findings and definitive conclusions.
Collapse
Affiliation(s)
- Anastasia Aluvilu
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Albert Ferro
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| |
Collapse
|
4
|
Burban A, Idzik A, Gelo A, Filipiak KJ, Jakimowicz T, Jama K, Grabowski M, Gasecka A, Siniarski A. Platelet function changes in patients undergoing endovascular aortic aneurysm repair: Review of the literature. Front Cardiovasc Med 2022; 9:927995. [PMID: 36035918 PMCID: PMC9417250 DOI: 10.3389/fcvm.2022.927995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Patients with abdominal aortic aneurysm (AAA) have a higher risk of cardiovascular (CV) events, which seems to be associated with disturbed platelet (PLT) function. Endovascular aneurysm repair (EVAR) is an emerging, less-invasive treatment alternative to surgical AAA repair. Both platelet function abnormalities in patients with AAA and the effect of EVAR on platelet function are poorly understood. In this review, we aim to fill the gap regarding the effect of EVAR on PLT function in AAA patients by discussing PLT function disturbances in patients with AAA, PLT function changes after EVAR, evidence from clinical studies regarding PLT function before and after EVAR, and antiplatelet or and antithrombotic treatment in patients undergoing EVAR. The goal of our review is to summarize the contemporary knowledge and initiate further studies to better understand PLT function changes in patients undergoing EVAR, optimize the pharmacotherapy before and after EVAR and further improve outcomes in this group of patients.
Collapse
Affiliation(s)
- Anna Burban
- Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Idzik
- Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Agata Gelo
- Department of Anesthesiology and Intensive Care, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof J Filipiak
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy, Warsaw, Poland
| | - Tomasz Jakimowicz
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Jama
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Grabowski
- Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Gasecka
- Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksander Siniarski
- Department of Coronary Disease and Heart Failure, Institute of Cardiology, Jagiellonian University Medical College, Cracow, Poland
- John Paul II Hospital, Cracow, Poland
| |
Collapse
|
5
|
Gelbenegger G, Grafeneder J, Gager GM, Siller-Matula JM, Schwameis M, Jilma B, Schoergenhofer C. Advanced pharmacodynamics of cangrelor in healthy volunteers: a dose-finding, open-label, pilot trial. Thromb J 2022; 20:19. [PMID: 35422039 PMCID: PMC9008922 DOI: 10.1186/s12959-022-00377-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High on-treatment platelet reactivity (HTPR) remains a major problem in the acute management of ST-elevation myocardial infarction (STEMI), leading to higher rates of stent thrombosis and mortality. We aimed to investigate a novel, prehospital treatment strategy using cangrelor and tested its pharmacodynamic effects in a model using healthy volunteers. METHODS We conducted a dose-finding, open-label, pilot trial including 12 healthy volunteers and tested three ascending bolus infusions of cangrelor (5 mg, 10 mg and 20 mg) and a bolus infusion followed by a continuous infusion via an intravenous (IV) flow regulator. Platelet function was assessed using multiple electrode aggregometry (MEA), vasodilator-stimulated phosphoprotein phosphorylation assay (VASP-P) and the platelet function analyzer. In an ex vivo experiment, epinephrine was used to counteract the antiplatelet effect of cangrelor. RESULTS All cangrelor bolus infusions resulted in immediate and pronounced platelet inhibition. Bolus infusions of cangrelor 20 mg resulted in sufficient platelet inhibition assessed by MEA for 20 min in 90% of subjects. Infusion of cangrelor via the IV flow regulator resulted in sufficient platelet inhibition throughout the course of administration. Ex vivo epinephrine, in concentrations of 200 and 500 ng/mL was able to partially reverse the antiplatelet effect of cangrelor in a dose-dependent manner. CONCLUSIONS Weight-adapted bolus infusions followed by a continuous infusion of cangrelor via IV flow regulator result in immediate and pronounced platelet inhibition in healthy subjects. Cangrelor given as weight-adapted bolus infusion followed by a continuous infusion using an IV flow regulator may be a viable treatment approach for effective and well controllable prehospital platelet inhibition. TRIAL REGISTRATION EC (Medical University of Vienna) 1835/2019 and EudraCT 2019-002792-34 .
Collapse
Affiliation(s)
- Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Juergen Grafeneder
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Gloria M Gager
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Jolanta M Siller-Matula
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Michael Schwameis
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christian Schoergenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
6
|
Moster M, Bolliger D. Perioperative Guidelines on Antiplatelet and Anticoagulant Agents: 2022 Update. CURRENT ANESTHESIOLOGY REPORTS 2022. [DOI: 10.1007/s40140-021-00511-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Abstract
Purpose of Review
Multiple guidelines and recommendations have been written to address the perioperative management of antiplatelet and anticoagulant drugs. In this review, we evaluated the recent guidelines in non-cardiac, cardiac, and regional anesthesia. Furthermore, we focused on unresolved problems and novel approaches for optimized perioperative management.
Recent Findings
Vitamin K antagonists should be stopped 3 to 5 days before surgery. Preoperative laboratory testing is recommended. Bridging therapy does not decrease the perioperative thromboembolic risk and might increase perioperative bleeding risk. In patients on direct-acting oral anticoagulants (DOAC), a discontinuation interval of 24 and 48 h in those scheduled for surgery with low and high bleeding risk, respectively, has been shown to be saved. Several guidelines for regional anesthesia recommend a conservative interruption interval of 72 h for DOACs before neuraxial anesthesia. Finally, aspirin is commonly continued in the perioperative period, whereas potent P2Y12 receptor inhibitors should be stopped, drug-specifically, 3 to 7 days before surgery.
Summary
Many guidelines have been published from various societies. Their applicability is limited in emergent or urgent surgery, where novel approaches might be helpful. However, their evidence is commonly based on small series, case reports, or expert opinions.
Collapse
|
7
|
Gąsecka A, Zawadka M, Burban A, Idzik A, Gelo A, Graczyńska A, Jama K, Filipiak KJ, Jakimowicz T. Pre-operative platelet reactivity is a strong, independent predictor of bleeding complications after branched endovascular thoracoabdominal aortic aneurysm repair. Platelets 2021; 33:577-585. [PMID: 34355639 DOI: 10.1080/09537104.2021.1961708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endovascular aortic repair (EVAR) an alternative to open surgical repair of thoracoabdominal aortic aneurysm (TAAA). The effect of EVAR on platelet reactivity is unknown. We prospectively determined the effect of branched EVAR (bEVAR) on platelet reactivity in patients with TAAA, and evaluated the predictive value of preoperative platelet reactivity for post-operative bleeding in 50 consecutive patients undergoing elective bEVAR (mean age 70.9 ± 5.7 years, 66% male). Blood samples were collected within 24 hours before bEVAR, after bEVAR and at hospital discharge. Platelet reactivity was assessed with impedance aggregometry using ASPI, ADP and TRAP tests. Platelet reactivity decreased within 24 hours after bEVAR compared to the measurement before bEVAR in all tests (p ≤ 0.04), with a further decrease in hospital discharge in the ADP test (p = .004). Twenty-three patients experienced post-operative bleeding complications (transfusion ≥2 red blood cell [RBC] units). Preoperative platelet reactivity below the cutoff value of 30 AUC units predicted post-operative bleeding with 78% sensitivity and 59% specificity (p = .045). In the multivariable analysis, platelet reactivity was the only independent predictor of postoperative bleeding (OR 6.507, 95% CI 1.227-34.506, p = .028). We conclude that platelet reactivity decreases following bEVAR of TAAA and is a strong and independent predictor for postoperative bleeding complications.
Collapse
Affiliation(s)
- Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland.,Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mateusz Zawadka
- 2nd Department of Anesthesia and Intensive Care, Medical University of Warsaw, Warsaw, Poland.,Department of Perioperative Medicine, Barts Heart Centre and St. Bartholomew's Hospital, London, UK
| | - Anna Burban
- 1 Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Idzik
- 1 Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Agata Gelo
- 2nd Department of Anesthesia and Intensive Care, Medical University of Warsaw, Warsaw, Poland
| | - Agata Graczyńska
- 2nd Department of Anesthesia and Intensive Care, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Jama
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof J Filipiak
- 1 Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Jakimowicz
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Czajka P, Fitas A, Jakubik D, Eyileten C, Gasecka A, Wicik Z, Siller-Matula JM, Filipiak KJ, Postula M. MicroRNA as Potential Biomarkers of Platelet Function on Antiplatelet Therapy: A Review. Front Physiol 2021; 12:652579. [PMID: 33935804 PMCID: PMC8081881 DOI: 10.3389/fphys.2021.652579] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs, able to regulate cellular functions by specific gene modifications. Platelets are the major source for circulating miRNAs, with significant regulatory potential on cardiovascular pathophysiology. MiRNAs have been shown to modify the expression of platelet proteins influencing platelet reactivity. Circulating miRNAs can be determined from plasma, serum, or whole blood, and they can be used as diagnostic and prognostic biomarkers of platelet reactivity during antiplatelet therapy as well as novel therapeutic targets in cardiovascular diseases (CVDs). Herein, we review diagnostic and prognostic value of miRNAs levels related to platelet reactivity based on human studies, presenting its interindividual variability as well as the substantial role of genetics. Furthermore, we discuss antiplatelet treatment in the context of miRNAs alterations related to pathways associated with drug response.
Collapse
Affiliation(s)
- Pamela Czajka
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Alex Fitas
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Daniel Jakubik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Aleksandra Gasecka
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland.,Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Paulo, Brazil
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland.,Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Krzysztof J Filipiak
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| |
Collapse
|
9
|
Hofer F, Perkmann T, Gager G, Winter MP, Niessner A, Hengstenberg C, Siller-Matula JM. Comparison of high-sensitivity C-reactive protein vs. C-reactive protein for diagnostic accuracy and prediction of mortality in patients with acute myocardial infarction. Ann Clin Biochem 2021; 58:342-349. [PMID: 33715444 DOI: 10.1177/00045632211004651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The role of chronic inflammation in the pathogenesis of atherosclerosis has been unequivocally proven. However, the prognostic impact of C-reactive protein, a marker of inflammatory response in patients with acute myocardial infarction has not been fully clarified. Furthermore, there is no direct comparison of the diagnostic accuracy of C-reactive protein and high sensitivity C-reactive protein in the acute myocardial infarction population. METHODS In this prospective observational cohort study, 344 patients with acute myocardial infarction were enrolled. All-cause mortality was a primary endpoint. Patients were followed prospectively for a median of six years. RESULTS The correlation between high sensitivity C-reactive protein and C-reactive protein (r = 0.99; P < 0.001) and the diagnostic accuracy (98.6%) was high. The ROC analysis revealed that C-reactive protein and high sensitivity C-reactive protein had a low AUC for prediction of mortality (C-reactive protein: 0.565, 95% CI [0.462-0.669], vs. high sensitivity C-reactive protein: 0.572, 95% CI [0.470-0.675]) or major adverse cardiac events (C-reactive protein: AUC 0.607, 95% CI [0.405-0.660], vs. high sensitivity C-reactive protein: AUC 0.526, 95% CI [0.398-0.653]) when assessed at time point of acute myocardial infarction. In contrast, longitudinal inflammatory risk assessment with serial C-reactive protein measurements in the stable phase of the disease revealed a 100% specificity, 100% negative predictive value, 32% sensitivity and 12% positive predictive value of C-reactive protein to predict long-term mortality. The Kaplan Meier analysis showed a significant survival benefit for patients at low residual inflammatory risk (P = 0.014). CONCLUSION C-reactive protein and high sensitivity C-reactive protein provide a similar diagnostic accuracy, highlighting that C-reactive protein might replace high sensitivity C-reactive protein in routine assessments. Furthermore, low inflammatory status during the stable phase after acute myocardial infarction predicts favourable six-year survival.
Collapse
Affiliation(s)
- Felix Hofer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gloria Gager
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Max-Paul Winter
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Alexander Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Jolanta M Siller-Matula
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.,Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Voetsch A, Pregartner G, Berghold A, Seitelberger R, Schoerghuber M, Toller W, Mahla E. How Do Type of Preoperative P2Y12 Receptor Inhibitor and Withdrawal Time Affect Bleeding? Ann Thorac Surg 2021; 111:77-84. [DOI: 10.1016/j.athoracsur.2020.04.126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/02/2020] [Accepted: 04/23/2020] [Indexed: 12/18/2022]
|
11
|
Lewis JP, Backman JD, Reny JL, Bergmeijer TO, Mitchell BD, Ritchie MD, Déry JP, Pakyz RE, Gong L, Ryan K, Kim EY, Aradi D, Fernandez-Cadenas I, Lee MTM, Whaley RM, Montaner J, Gensini GF, Cleator JH, Chang K, Holmvang L, Hochholzer W, Roden DM, Winter S, Altman RB, Alexopoulos D, Kim HS, Gawaz M, Bliden KP, Valgimigli M, Marcucci R, Campo G, Schaeffeler E, Dridi NP, Wen MS, Shin JG, Fontana P, Giusti B, Geisler T, Kubo M, Trenk D, Siller-Matula JM, Ten Berg JM, Gurbel PA, Schwab M, Klein TE, Shuldiner AR. Pharmacogenomic polygenic response score predicts ischaemic events and cardiovascular mortality in clopidogrel-treated patients. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2020; 6:203-210. [PMID: 31504375 DOI: 10.1093/ehjcvp/pvz045] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/15/2019] [Accepted: 08/29/2019] [Indexed: 01/23/2023]
Abstract
AIMS Clopidogrel is prescribed for the prevention of atherothrombotic events. While investigations have identified genetic determinants of inter-individual variability in on-treatment platelet inhibition (e.g. CYP2C19*2), evidence that these variants have clinical utility to predict major adverse cardiovascular events (CVEs) remains controversial. METHODS AND RESULTS We assessed the impact of 31 candidate gene polymorphisms on adenosine diphosphate (ADP)-stimulated platelet reactivity in 3391 clopidogrel-treated coronary artery disease patients of the International Clopidogrel Pharmacogenomics Consortium (ICPC). The influence of these polymorphisms on CVEs was tested in 2134 ICPC patients (N = 129 events) in whom clinical event data were available. Several variants were associated with on-treatment ADP-stimulated platelet reactivity (CYP2C19*2, P = 8.8 × 10-54; CES1 G143E, P = 1.3 × 10-16; CYP2C19*17, P = 9.5 × 10-10; CYP2B6 1294 + 53 C > T, P = 3.0 × 10-4; CYP2B6 516 G > T, P = 1.0 × 10-3; CYP2C9*2, P = 1.2 × 10-3; and CYP2C9*3, P = 1.5 × 10-3). While no individual variant was associated with CVEs, generation of a pharmacogenomic polygenic response score (PgxRS) revealed that patients who carried a greater number of alleles that associated with increased on-treatment platelet reactivity were more likely to experience CVEs (β = 0.17, SE 0.06, P = 0.01) and cardiovascular-related death (β = 0.43, SE 0.16, P = 0.007). Patients who carried eight or more risk alleles were significantly more likely to experience CVEs [odds ratio (OR) = 1.78, 95% confidence interval (CI) 1.14-2.76, P = 0.01] and cardiovascular death (OR = 4.39, 95% CI 1.35-14.27, P = 0.01) compared to patients who carried six or fewer of these alleles. CONCLUSION Several polymorphisms impact clopidogrel response and PgxRS is a predictor of cardiovascular outcomes. Additional investigations that identify novel determinants of clopidogrel response and validating polygenic models may facilitate future precision medicine strategies.
Collapse
Affiliation(s)
- Joshua P Lewis
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA
| | - Joshua D Backman
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA
| | - Jean-Luc Reny
- Department of Internal Medicine, Béziers Hospital, 2 Rue Valentin Hau, BP 740, Béziers 34525, France.,Department of Medicine, Geneva Platelet Group, University of Geneva School of Medicine, University Hospitals of Geneva, 24 rue du Général-Dufour, Genève 4 CH-1211, Switzerland
| | - Thomas O Bergmeijer
- Department of Cardiology, Antonius Center for Platelet Function Research, St Antonius Hospital, P O Box 2500, Nieuwegein 3432 EM, The Netherlands
| | - Braxton D Mitchell
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA.,Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, 10 N. Greene St., Baltimore, MD 21201, USA
| | - Marylyn D Ritchie
- Center for Translational Bioinformatics, Institute for Biomedical Informatics, University of Pennsylvania, A301 Richards Building, 3700 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Jean-Pierre Déry
- Quebec Heart and Lung Institute, University Laval, 2725 chemin Sainte-Foy, Quebec City G1V 4G5, Canada
| | - Ruth E Pakyz
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, 443 Via Ortega, Room 213, Stanford, CA 94305, USA
| | - Kathleen Ryan
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA
| | - Eun-Young Kim
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Bokji-ro 75, Busangjin-gu, Busan 614-735, South Korea
| | - Daniel Aradi
- Department of Cardiology, Heart Center Balatonfüred, 2 Gyogy Ter, Balatonfured 8230, Hungary
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetic Group, Fundació Docencia i Recerca Mutuaterrassa, 508221 Terrassa, Barcelona 8041, Spain.,Department of Neurology, Vall d'Hebron Institute of Research, Passeig Vall d'Hebron, Barcelona 8035, Spain
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger Health System, 100 N. Academy Ave., Danville, PA 17822, USA
| | - Ryan M Whaley
- Department of Biomedical Data Science, Stanford University, 443 Via Ortega, Room 213, Stanford, CA 94305, USA
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Passeig Vall d'Hebron 119-129, Barcelona 8035, Spain
| | - Gian Franco Gensini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, Florence 50055, Italy
| | - John H Cleator
- Division of Cardiology, Vanderbilt University Medical Center, 2215B Garland Avenue, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University Medical Center, 2215B Garland Avenue, Nashville, TN 37232, USA
| | - Kiyuk Chang
- Department of Internal Medicine, Cardiology Division, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul 6591, South Korea
| | - Lene Holmvang
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmannsvej 7 - 2142, Copenhagen 2100, Denmark
| | - Willibald Hochholzer
- Department of Cardiology and Angiology II, University Heart Center Freiburg, Suedring 15, Bad Krozingen 79189, Germany
| | - Dan M Roden
- Department of Pharmacology, Vanderbilt University Medical Center, 2215B Garland Avenue, Nashville, TN 37232, USA.,Department of Medicine, Vanderbilt University Medical Center, 2215B Garland Avenue, Nashville, TN 37232, USA.,Department of Biomedical Informatics, Vanderbilt University Medical Center, 2215B Garland Avenue, Nashville, TN 37232, USA
| | - Stefan Winter
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, Stuttgart, 70376 Germany
| | - Russ B Altman
- Department of Bioengineering, Genetics, and Medicine, Stanford University, 443 Via Ortega Drive, Shriram Room 209, Stanford, CA 94305, USA
| | | | - Ho-Sook Kim
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Gaegum2-dong 622-165, Busanjin-Gu, Busan 614-735, South Korea
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Otfired-Müller-Straße 10, Tübingen 72076, Germany
| | - Kevin P Bliden
- Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, 3300 Gallows Rd, Falls Church, VA 22042, USA
| | - Marco Valgimigli
- Department of Cardiology, Bern University Hospital, Freiburgstrasse 8, Bern 3010, Switzerland
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, Florence 50055, Italy.,Atherothrombotic Diseases Center, Careggi University Hospital, Largo G. Alessandro Brambilla, Florence 50134, Italy
| | - Gianluca Campo
- Department of Cardiology, University Hospital of Ferrara, Via Aldo Moro 8, Cona (FE), Ferrara 44123, Italy.,GVM Care & Research, Maria Cecilia Hospital, Via Madonna di Genova, 1, Cotignola 48033, Italy
| | - Elke Schaeffeler
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, Stuttgart, 70376 Germany
| | - Nadia P Dridi
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Inge Lehmannsvej 7 - 2142, Copenhagen 2100, Denmark
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and School of Medicine, Chang Gung University, No. 5, Fuxing St, Guishan Dist., Taoyuan City 333, Taiwan
| | - Jae Gook Shin
- Department of Clinical Pharmacology, Inje University, Busan Paik Hospital, Gaegum2-dong 622-165, Busanjin-Gu, Busan 614-735, South Korea
| | - Pierre Fontana
- Department of Medicine, Geneva Platelet Group, University of Geneva School of Medicine, University Hospitals of Geneva, 24 rue du Général-Dufour, Genève 4 CH-1211, Switzerland.,Division of Angiology and Haemostasis, University Hospitals of Geneva, 24 Rue Gabrielle-Perret-Gentil, Geneva 1205, Switzerland
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, Florence 50055, Italy.,Atherothrombotic Diseases Center, Careggi University Hospital, Largo G. Alessandro Brambilla, Florence 50134, Italy
| | - Tobias Geisler
- Department of Cardiology and Angiology, University of Tübingen, Otfired-Müller-Straße 10, Tübingen 72076, Germany
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Dietmar Trenk
- Department of Cardiology and Angiology II, Clinical Pharmacology, University Heart Centre Freiburg, Suedring 15, Bad Krozingen D-79189, Germany
| | - Jolanta M Siller-Matula
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria
| | - Jurriën M Ten Berg
- Department of Cardiology, Antonius Center for Platelet Function Research, St Antonius Hospital, P O Box 2500, Nieuwegein 3432 EM, The Netherlands
| | - Paul A Gurbel
- Department of Cardiology and Angiology, University of Tübingen, Otfired-Müller-Straße 10, Tübingen 72076, Germany
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, Stuttgart, 70376 Germany.,Department of Clinical Pharmacology, University of Tuebingen, Otfried-Mueller-Strasse 10, Tuebingen 72076, Germany.,Department of Pharmacy and Biochemistry, University of Tuebingen, Otfried-Mueller-Strasse 10, Tuebingen 72076, Germany
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford University, 443 Via Ortega, Room 213, Stanford, CA 94305, USA.,Department of Bioengineering, Genetics, and Medicine, Stanford University, 443 Via Ortega Drive, Shriram Room 209, Stanford, CA 94305, USA
| | - Alan R Shuldiner
- Department of Medicine and Program for Personalized and Genomic Medicine, University of Maryland, 670 W. Baltimore St., Baltimore, MD 21201, USA
| | | |
Collapse
|
12
|
Gager GM, Jilma B, Winter M, Hengstenberg C, Lang IM, Toma A, Prüller F, Wallner M, Kolesnik E, von Lewinski D, Siller‐Matula JM. Ticagrelor and prasugrel are independent predictors of improved long-term survival in ACS patients. Eur J Clin Invest 2020; 50:e13304. [PMID: 32506444 PMCID: PMC7685125 DOI: 10.1111/eci.13304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022]
Abstract
AIM To investigate the long-term clinical benefit of dual antiplatelet therapy with potent P2Y12 inhibitors compared to clopidogrel in patients with acute coronary syndrome (ACS). METHODS In this prospective multicenter observational study, we enrolled 708 patients with ACS treated with clopidogrel (n = 137), ticagrelor (n = 260) or prasugrel (n = 311). Major adverse cardiac events (MACE; over 1 year) and long-term mortality (median: 5.6 years; interquartile range [IQR] 4.9-6.5 years) were assessed. Multiple electrode aggregometry (MEA) was used to measure adenosine diphosphate (ADP)- and arachidonic acid (AA)-induced platelet aggregation. RESULTS Type of P2Y12 inhibitor emerged as an independent predictor of long-term mortality and MACE: patients treated with potent platelet inhibitors prasugrel or ticagrelor were at lower risk for long-term mortality (adjusted hazard ratio [HR] = 0.44; 95% CI: 0.22-0.92; P = .028) or MACE (adjusted HR = 0.38; 95% CI: 0.20-0.73; P = .004) than those treated with clopidogrel independent from clinical risk factors. In contrast, the efficacy of clopidogrel decreased with increasing severity of ACS: platelet aggregation was 37% higher in patients with ST segment elevation myocardial infarction (STEMI) and 25% higher in patients with non-ST elevation myocardial infarction (non-STEMI) compared to patients with unstable angina (P = .039). Patients with diabetes achieved less potent ADP- and AA-induced platelet inhibition under clopidogrel, compared to patients without diabetes (P = .045; P = .030, respectively). CONCLUSION In the setting of ACS, treatment with ticagrelor or prasugrel reduced long-term mortality and 1-year MACE as compared to clopidogrel.
Collapse
Affiliation(s)
- Gloria M. Gager
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
| | - Bernd Jilma
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - Max‐Paul Winter
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
| | - Christian Hengstenberg
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
| | - Irene M. Lang
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
| | - Aurel Toma
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Markus Wallner
- Department of CardiologyMedical University of GrazGrazAustria
- Cardiovascular Research CenterLewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
- Center for Biomarker Research in MedicineCBmed GmbHGrazAustria
| | - Ewald Kolesnik
- Department of CardiologyMedical University of GrazGrazAustria
| | | | - Jolanta M. Siller‐Matula
- Department of Internal Medicine IIDivision of CardiologyMedical University of ViennaViennaAustria
- Department of Experimental and Clinical PharmacologyCentre for Preclinical Research and Technology (CEPT)Medical University of WarsawWarsawPoland
| |
Collapse
|
13
|
Shao T, Cheng Y, Jin J, Huang L, Yang D, Luo C, Han Z, Wang Z, Ge W, Xu Y. A comparison of three platelet function tests in ischemic stroke patients with antiplatelet therapy. J Clin Neurosci 2020; 78:91-96. [PMID: 32624366 DOI: 10.1016/j.jocn.2020.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/20/2020] [Accepted: 06/06/2020] [Indexed: 12/31/2022]
Abstract
Predicting the effectiveness of antiplatelet drugs is critical to precision antiplatelet therapy. However, there is a lack of an acceptable method, although there are a variety of methods for detecting platelet function. In this study, we compared three major platelet function tests to assess their performance and found better methods for platelet function evaluation after aspirin or clopidogrel treatment in ischemic stroke patients by comparative study. A total of 249 ischemic stroke patients were enrolled who were treated with aspirin or clopidogrel or both. Three platelet function tests including light transmittance aggregometry (LTA), thromboelastography (TEG), platelet function analyzer (PFA) were performed as well as CYP2C19 genotype determination. Correlation analyses and kappa statistics were used. All three methods were effective in evaluating aspirin function. However, only LTA and TEG had good correlation and consistency (r = -0.37, kappa = 0.634). TEG-ADP was the least sensitive for clopidogrel, as the platelet inhibition ratio did not differ between the clopidogrel-user group and the control (P = 0.074), while LTA and PFA were sensitive (P < 0.001). Correlations between platelet assays were poor for clopidogrel (the absolute value of r range from 0.13 to 0.35) and so was the agreement (Kappa from 0.232 to 0.314). LTA and PFA have a good correlation with CYP2C19 genotyping (P = 0.034 and 0.014). In conclusion, all three tests were able to evaluate aspirin effect, LTA-AA and TEG-AA had a good correlation. TEG perform badly for clopidogrel effect detection. The fair-to-modest agreement among assays indicated further study was indispensable.
Collapse
Affiliation(s)
- Tengfei Shao
- Department of Pharmacy, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yue Cheng
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jiali Jin
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Lili Huang
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Dan Yang
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Caimei Luo
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhou Han
- Department of Pharmacy, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhongyuan Wang
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Weihong Ge
- Department of Pharmacy, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China.
| |
Collapse
|
14
|
Ali Z, Elewa H. The Effect of CYP2C19 and Nongenetic Factors on Clopidogrel Responsiveness in the MENA Region: A Systematic Review. Clin Appl Thromb Hemost 2020; 25:1076029619875520. [PMID: 31512486 PMCID: PMC6829969 DOI: 10.1177/1076029619875520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clopidogrel is the cornerstone antiplatelet used in the treatment and prevention of thrombotic events. Some studies examined the effect of CYP2C19 polymorphism and nongenetic factors on clopidogrel response in the Middle East and North Africa (MENA) region. However, the consistency among these studies is yet unknown. This study aims to estimate the prevalence of CYP2C19 genetic variants in MENA region and to evaluate the effect of these variants as well as the nongenetic factors on clopidogrel responsiveness. A systematic literature search was performed to identify relevant articles. Only observational studies were included. A total of 20 studies in 8 different populations were included. The CYP2C19*2 variant is the most prevalent loss-of-function (LOF) allele in the MENA region (1.7%-35%). The frequency of CYP2C19*17 ranged from 5.3% to 26.9%. Of the 9 studies, 6 found an association between carriers of at least 1 LOF allele and clopidogrel resistance. Older age, high body mass index, females, and the use of calcium channel blockers were associated with clopidogrel resistance as well. Association between the CYP2C19*2 allele and clopidogrel resistance is common among MENA populations. Future studies should focus on having larger sample sizes to detect other minor variant alleles and their effect on bleeding and cardiovascular outcomes.
Collapse
Affiliation(s)
- Zainab Ali
- College of Pharmacy, Qatar University, Doha, Qatar
| | - Hazem Elewa
- College of Pharmacy, Qatar University, Doha, Qatar
- Hazem Elewa, College of Pharmacy, Qatar University,
Doha 2713, Qatar.
| |
Collapse
|
15
|
Gasecka A, Nieuwland R, Budnik M, Dignat‐George F, Eyileten C, Harrison P, Lacroix R, Leroyer A, Opolski G, Pluta K, van der Pol E, Postuła M, Siljander P, Siller‐Matula JM, Filipiak KJ. Ticagrelor attenuates the increase of extracellular vesicle concentrations in plasma after acute myocardial infarction compared to clopidogrel. J Thromb Haemost 2020; 18:609-623. [PMID: 31833175 PMCID: PMC7065161 DOI: 10.1111/jth.14689] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Platelet P2Y12 antagonist ticagrelor reduces mortality after acute myocardial infarction (AMI) compared to clopidogrel, but the underlying mechanism is unknown. Because activated platelets, leukocytes, and endothelial cells release proinflammatory and prothrombotic extracellular vesicles (EVs), we hypothesized that the release of EVs is more efficiently inhibited by ticagrelor compared to clopidogrel. OBJECTIVES We compared EV concentrations and EV procoagulant activity in plasma of patients after AMI treated with ticagrelor or clopidogrel. METHODS After percutaneous coronary intervention, 60 patients with first AMI were randomized to ticagrelor or clopidogrel. Flow cytometry was used to determine concentrations of EVs from activated platelets (CD61+ , CD62p+ ), fibrinogen+ , phosphatidylserine (PS+ ), leukocytes (CD45+ ), endothelial cells (CD31+ , 146+ ), and erythrocytes (CD235a+ ) in plasma at randomization, after 72 hours and 6 months of treatment. A fibrin generation test was used to determine EV procoagulant activity. RESULTS Concentrations of platelet, fibrinogen+ , PS+ , leukocyte, and erythrocyte EVs increased 6 months after AMI compared to the acute phase of AMI (P ≤ .03). Concentrations of platelet EVs were lower on ticagrelor compared to clopidogrel after 6 months (P = .03). Concentrations of fibrinogen+ , PS+ , and leukocyte EVs were lower on ticagrelor compared to clopidogrel both after 72 hours and 6 months (P ≤ .03). Concentrations of endothelial EVs and EV procoagulant activity did not differ between patient groups and over time (P ≥ .17). CONCLUSIONS Ticagrelor attenuates the increase of EV concentrations in plasma after acute myocardial infarction compared to clopidogrel. The ongoing release of EVs despite antiplatelet therapy might explain recurrent thrombotic events after AMI and worse clinical outcomes on clopidogrel compared to ticagrelor.
Collapse
Affiliation(s)
- Aleksandra Gasecka
- First Chair and Department of CardiologyMedical University of WarsawPoland
- Vesicle Observation Centre, and Laboratory of Experimental Clinical ChemistryAmsterdam UMCUniversity of Amsterdamthe Netherlands
| | - Rienk Nieuwland
- Vesicle Observation Centre, and Laboratory of Experimental Clinical ChemistryAmsterdam UMCUniversity of Amsterdamthe Netherlands
| | - Monika Budnik
- First Chair and Department of CardiologyMedical University of WarsawPoland
| | - Françoise Dignat‐George
- INSERMFaculty of PharmacyAix‐Marseille UniversityMarseilleFrance
- Hematology and Vascular Biology DepartmentCHU La ConceptionAP‐HMMarseilleFrance
| | - Ceren Eyileten
- Department of Experimental and Clinical PharmacologyCentre for Preclinical Research and TechnologyMedical University of WarsawWarsawPoland
| | - Paul Harrison
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | - Romaric Lacroix
- INSERMFaculty of PharmacyAix‐Marseille UniversityMarseilleFrance
- Hematology and Vascular Biology DepartmentCHU La ConceptionAP‐HMMarseilleFrance
| | - Aurélie Leroyer
- INSERMFaculty of PharmacyAix‐Marseille UniversityMarseilleFrance
| | - Grzegorz Opolski
- First Chair and Department of CardiologyMedical University of WarsawPoland
| | - Kinga Pluta
- First Chair and Department of CardiologyMedical University of WarsawPoland
| | - Edwin van der Pol
- Vesicle Observation Centre, and Laboratory of Experimental Clinical ChemistryAmsterdam UMCUniversity of Amsterdamthe Netherlands
- Biomedical Engineering & PhysicsAmsterdam UMCUniversity of Amsterdamthe Netherlands
| | - Marek Postuła
- Department of Experimental and Clinical PharmacologyCentre for Preclinical Research and TechnologyMedical University of WarsawWarsawPoland
| | - Pia Siljander
- EV‐group, Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiFinland
| | | | | |
Collapse
|
16
|
Ostrowska M, Kubica J, Adamski P, Kubica A, Eyileten C, Postula M, Toma A, Hengstenberg C, Siller-Matula JM. Stratified Approaches to Antiplatelet Therapies Based on Platelet Reactivity Testing. Front Cardiovasc Med 2019; 6:176. [PMID: 31850373 PMCID: PMC6901499 DOI: 10.3389/fcvm.2019.00176] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/14/2019] [Indexed: 01/17/2023] Open
Abstract
Antiplatelet therapy with P2Y12 receptor inhibitors (clopidogrel, prasugrel, ticagrelor, cangrelor) is a cornerstone of medical therapy after percutaneous coronary interventions. Significant prevalence of high on-treatment platelet reactivity (HTPR) on clopidogrel treatment led to introduction of more potent P2Y12 inhibitors: prasugrel (a third generation thienopyridine), ticagrelor, and cangrelor (cyclopentyl-triazolo-pyrimidines). Nevertheless, more potent platelet inhibition and resulting low on-treatment platelet reactivity (LTPR) has led to increased risk of major bleeding events. These limitations resulted in a need for an individualized antiplatelet therapy approach. This review discusses the current role and future perspectives of diagnostic tools such as platelet function testing to optimize antiplatelet therapy with a focus on deescalating therapies to reduce bleeding risks.
Collapse
Affiliation(s)
- Małgorzata Ostrowska
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Piotr Adamski
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Aldona Kubica
- Department of Health Promotion, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Aurel Toma
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland.,Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Eyileten C, Soplinska A, Pordzik J, Siller‐Matula JM, Postuła M. Effectiveness of Antiplatelet Drugs Under Therapeutic Hypothermia: A Comprehensive Review. Clin Pharmacol Ther 2019; 106:993-1005. [DOI: 10.1002/cpt.1492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Ceren Eyileten
- Department of Experimental and Clinical PharmacologyCenter for Preclinical Research and Technology CEPTMedical University of Warsaw Warsaw Poland
| | - Aleksandra Soplinska
- Department of Experimental and Clinical PharmacologyCenter for Preclinical Research and Technology CEPTMedical University of Warsaw Warsaw Poland
| | - Justyna Pordzik
- Department of Experimental and Clinical PharmacologyCenter for Preclinical Research and Technology CEPTMedical University of Warsaw Warsaw Poland
| | | | - Marek Postuła
- Department of Experimental and Clinical PharmacologyCenter for Preclinical Research and Technology CEPTMedical University of Warsaw Warsaw Poland
| |
Collapse
|
18
|
Gasecka A, Nieuwland R, Budnik M, Dignat-George F, Eyileten C, Harrison P, Huczek Z, Kapłon-Cieślicka A, Lacroix R, Opolski G, Pluta K, van der Pol E, Postuła M, Leroyer A, Siljander P, Sturk A, Filipiak KJ. Randomized controlled trial protocol to investigate the antiplatelet therapy effect on extracellular vesicles (AFFECT EV) in acute myocardial infarction. Platelets 2018; 31:26-32. [PMID: 30585111 DOI: 10.1080/09537104.2018.1557616] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Activated platelets contribute to thrombosis and inflammation by the release of extracellular vesicles (EVs) exposing P-selectin, phosphatidylserine (PS) and fibrinogen. P2Y12 receptor antagonists are routinely administered to inhibit platelet activation in patients after acute myocardial infarction (AMI), being a combined antithrombotic and anti-inflammatory therapy. The more potent P2Y12 antagonist ticagrelor improves cardiovascular outcome in patients after AMI compared to the less potent clopidogrel, suggesting that greater inhibition of platelet aggregation is associated with better prognosis. The effect of ticagrelor and clopidogrel on the release of EVs from platelets and other P2Y12-exposing cells is unknown. This study compares the effects of ticagrelor and clopidogrel on (1) the concentrations of EVs from activated platelets (primary end point), (2) the concentrations of EVs exposing fibrinogen, exposing PS, from leukocytes and from endothelial cells (secondary end points) and (3) the procoagulant activity of plasma EVs (tertiary end points) in 60 consecutive AMI patients. After the percutaneous coronary intervention, patients will be randomized to antiplatelet therapy with ticagrelor (study group) or clopidogrel (control group). Blood will be collected from patients at randomization, 48 hours after randomization and 6 months following the index hospitalization. In addition, 30 age- and gender-matched healthy volunteers will be enrolled in the study to investigate the physiological concentrations and procoagulant activity of EVs using recently standardized protocols and EV-dedicated flow cytometry. Concentrations of EVs will be determined by flow cytometry. Procoagulant activity of EVs will be determined by fibrin generation test. The compliance and response to antiplatelet therapy will be assessed by impedance aggregometry. We expect that plasma from patients treated with ticagrelor (1) contains lower concentrations of EVs from activated platelets, exposing fibrinogen, exposing PS, from leukocytes and from endothelial cells and (2) has lower procoagulant activity, when compared to patients treated with clopidogrel. Antiplatelet therapy effect on EVs may identify a new mechanism of action of ticagrelor, as well as create a basis for future studies to investigate whether lower EV concentrations are associated with improved clinical outcomes in patients treated with P2Y12 antagonists.
Collapse
Affiliation(s)
- Aleksandra Gasecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland.,Vesicle Observation Centre and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Vesicle Observation Centre and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Monika Budnik
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | | | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Warsaw, Poland
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Zenon Huczek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | | | - Romaric Lacroix
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Kinga Pluta
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Edwin van der Pol
- Vesicle Observation Centre and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Biomedical Engineering & Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Warsaw, Poland
| | - Aurélie Leroyer
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| | - Pia Siljander
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland.,Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Auguste Sturk
- Vesicle Observation Centre and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Krzysztof J Filipiak
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
19
|
Alfredsson J, Swahn E, Gustafsson KM, Janzon M, Jonasson L, Logander E, Nilsson L, Lindahl TL. Individual long-term variation of platelet reactivity in patients with dual antiplatelet therapy after myocardial infarction. Platelets 2018; 30:572-578. [DOI: 10.1080/09537104.2018.1479519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Joakim Alfredsson
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Eva Swahn
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Kerstin M Gustafsson
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Magnus Janzon
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lena Jonasson
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Elisabeth Logander
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lennart Nilsson
- Department of Cardiology and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Tomas L. Lindahl
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
20
|
Moon JY, Franchi F, Rollini F, Rios JRR, Kureti M, Cavallari LH, Angiolillo DJ. Role of genetic testing in patients undergoing percutaneous coronary intervention. Expert Rev Clin Pharmacol 2018; 11:151-164. [PMID: 28689434 PMCID: PMC5771818 DOI: 10.1080/17512433.2017.1353909] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Variability in individual response profiles to antiplatelet therapy, in particular clopidogrel, is a well-established phenomenon. Genetic variations of the cytochrome P450 (CYP) 2C19 enzyme, a key determinant in clopidogrel metabolism, have been associated with clopidogrel response profiles. Moreover, the presence of a CYP2C19 loss-of-function allele is associated with an increased risk of atherothrombotic events among clopidogrel-treated patients undergoing percutaneous coronary interventions (PCI), prompting studies evaluating the use of genetic tests to identify patients who may be potential candidates for alternative platelet P2Y12 receptor inhibiting therapies (prasugrel or ticagrelor). Areas covered: The present manuscript provides an overview of genetic factors associated with response profiles to platelet P2Y12 receptor inhibitors and their clinical implications, as well as the most recent developments and future considerations on the role of genetic testing in patients undergoing PCI. Expert commentary: The availability of more user-friendly genetic tests has contributed towards the development of many ongoing clinical trials and personalized medicine programs for patients undergoing PCI. Results of pilot investigations have shown promising results, which however need to be confirmed in larger-scale studies to support the routine use of genetic testing as a strategy to personalize antiplatelet therapy and improve clinical outcomes.
Collapse
Affiliation(s)
- Jae Youn Moon
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Jose R. Rivas Rios
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Megha Kureti
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Center for Pharmacogenomics, University of Florida, Gainesville, FL, USA
- Clinical & Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Dominick J. Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
21
|
Spronk HMH, Padro T, Siland JE, Prochaska JH, Winters J, van der Wal AC, Posthuma JJ, Lowe G, d'Alessandro E, Wenzel P, Coenen DM, Reitsma PH, Ruf W, van Gorp RH, Koenen RR, Vajen T, Alshaikh NA, Wolberg AS, Macrae FL, Asquith N, Heemskerk J, Heinzmann A, Moorlag M, Mackman N, van der Meijden P, Meijers JCM, Heestermans M, Renné T, Dólleman S, Chayouâ W, Ariëns RAS, Baaten CC, Nagy M, Kuliopulos A, Posma JJ, Harrison P, Vries MJ, Crijns HJGM, Dudink EAMP, Buller HR, Henskens YMC, Själander A, Zwaveling S, Erküner O, Eikelboom JW, Gulpen A, Peeters FECM, Douxfils J, Olie RH, Baglin T, Leader A, Schotten U, Scaf B, van Beusekom HMM, Mosnier LO, van der Vorm L, Declerck P, Visser M, Dippel DWJ, Strijbis VJ, Pertiwi K, Ten Cate-Hoek AJ, Ten Cate H. Atherothrombosis and Thromboembolism: Position Paper from the Second Maastricht Consensus Conference on Thrombosis. Thromb Haemost 2018; 118:229-250. [PMID: 29378352 DOI: 10.1160/th17-07-0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.
Collapse
Affiliation(s)
- H M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Padro
- Cardiovascular Research Center (ICCC), Hospital Sant Pau, Barcelona, Spain
| | - J E Siland
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - J H Prochaska
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - J Winters
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A C van der Wal
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - J J Posthuma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - G Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - E d'Alessandro
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Wenzel
- Department of Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - D M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - P H Reitsma
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - W Ruf
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - R H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - R R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - T Vajen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - N A Alshaikh
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - F L Macrae
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - N Asquith
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - J Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Moorlag
- Synapse, Maastricht, The Netherlands
| | - N Mackman
- Department of Medicine, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States
| | - P van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - J C M Meijers
- Department of Plasma Proteins, Sanquin, Amsterdam, The Netherlands
| | - M Heestermans
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - T Renné
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S Dólleman
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - W Chayouâ
- Synapse, Maastricht, The Netherlands
| | - R A S Ariëns
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - C C Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Kuliopulos
- Tufts University School of Graduate Biomedical Sciences, Biochemistry/Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - J J Posma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - M J Vries
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H J G M Crijns
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - E A M P Dudink
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H R Buller
- Department of Vascular Medicine, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - Y M C Henskens
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Själander
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - S Zwaveling
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Synapse, Maastricht, The Netherlands
| | - O Erküner
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J W Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - A Gulpen
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - F E C M Peeters
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J Douxfils
- Department of Pharmacy, Thrombosis and Hemostasis Center, Faculty of Medicine, Namur University, Namur, Belgium
| | - R H Olie
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Baglin
- Department of Haematology, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - A Leader
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Tel Aviv, Israel
| | - U Schotten
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - B Scaf
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - H M M van Beusekom
- Department of Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | | | - P Declerck
- Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | | | - D W J Dippel
- Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | | | - K Pertiwi
- Department of Cardiovascular Pathology, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - A J Ten Cate-Hoek
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
22
|
van der Meijden PEJ, Henskens YMC, ten Cate-Hoek AJ, Cate HT, Vries MJA. Assessment of bleeding risk in patients with coronary artery disease on dual antiplatelet therapy. Thromb Haemost 2017; 115:7-24. [DOI: 10.1160/th15-04-0355] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/29/2015] [Indexed: 12/14/2022]
Abstract
SummaryPatients with coronary artery disease are usually treated with dual antiplatelet therapy (DAPT) after percutaneous coronary intervention. Patients on DAPT are at risk of both ischaemic and bleeding events. Although side-lined for a long time, real-life studies have shown that both the incidence and the associated morbidity and mortality of outof-hospital bleeding are high. This indicates that prevention of (postinterventional) bleeding is as important as prevention of ischaemia. For this purpose it is crucial to reliably identify patients with a high bleeding risk. In order to postulate an algorithm, which could help identifying these patients, we performed a systematic review to determine the value of previously proposed prognostic modalities for bleeding. We searched and appraised the following tools: platelet function tests, genetic tests, bleeding scores and questionnaires and haemostatic tests. Most studies indicated that low on-treatment platelet reactivity (LTPR), as measured by several platelet function tests, and the carriage of CYP2C19*17 allele were independent risk factors for bleeding. A bleeding score also proved to be helpful in identifying patients at risk. No studies on haemostatic tests were retrieved. Several patient characteristics were also identified as independent predictors of bleeding, such as older age, female sex and renal failure. Combining these risk factors we propose an algorithm that would hypothetically facilitate identification of those patients at highest risk, warranting prevention measures for bleeding. This could be a starting point for further research concerning the topic.
Collapse
|
23
|
Platelets redox balance assessment: Current evidence and methodological considerations. Vascul Pharmacol 2017; 93-95:6-13. [DOI: 10.1016/j.vph.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/10/2017] [Accepted: 06/28/2017] [Indexed: 01/22/2023]
|
24
|
Jiang M, You JHS. CYP2C19 LOF and GOF-Guided Antiplatelet Therapy in Patients with Acute Coronary Syndrome: A Cost-Effectiveness Analysis. Cardiovasc Drugs Ther 2017; 31:39-49. [PMID: 27924429 DOI: 10.1007/s10557-016-6705-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE This study aimed to examine the cost-effectiveness of CYP2C19 loss-of-function and gain-of-function allele guided (LOF/GOF-guided) antiplatelet therapy in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI). METHODS A life-long decision-analytic model was designed to simulate outcomes of three strategies: universal clopidogrel (75 mg daily), universal alternative P2Y12 inhibitor (prasugrel 10 mg daily or ticagrelor 90 mg twice daily), and LOF/GOF-guided therapy (LOF/GOF allele carriers receiving alternative P2Y12 inhibitor, wild-type patients receiving clopidogrel). Model outcomes included clinical event rates, quality-adjusted life-years (QALYs) gained and direct medical costs from perspective of US healthcare provider. RESULTS Base-case analysis found nonfatal myocardial infarction (5.62%) and stent thrombosis (1.2%) to be the lowest in universal alternative P2Y12 inhibitor arm, whereas nonfatal stroke (0.72%), cardiovascular death (2.42%), and major bleeding (2.73%) were lowest in LOF/GOF-guided group. LOF/GOF-guided arm gained the highest QALYs (7.5301 QALYs) at lowest life-long cost (USD 76,450). One-way sensitivity analysis showed base-case results were subject to the hazard ratio of cardiovascular death in carriers versus non-carriers of LOF allele and hazard ratio of cardiovascular death in non-carriers of LOF allele versus general patients. In probabilistic sensitivity analysis of 10,000 Monte Carlo simulations, LOF/GOF-guided therapy, universal alternative P2Y12 inhibitor, and universal clopidogrel were the preferred strategy (willingness-to-pay threshold = 50,000 USD/QALY) in 99.07%, 0.04%, and 0.89% of time, respectively. CONCLUSIONS Using both CYP2C19 GOF and LOF alleles to select antiplatelet therapy appears to be the preferred antiplatelet strategy over universal clopidogrel and universal alternative P2Y12 inhibitor therapy for ACS patients with PCI.
Collapse
Affiliation(s)
- Minghuan Jiang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N. T, Hong Kong, China
| | - Joyce H S You
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N. T, Hong Kong, China.
| |
Collapse
|
25
|
Winter MP, Grove EL, De Caterina R, Gorog DA, Ahrens I, Geisler T, Gurbel PA, Tantry U, Navarese EP, Siller-Matula JM. Advocating cardiovascular precision medicine with P2Y12 receptor inhibitors. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2017; 3:221-234. [DOI: 10.1093/ehjcvp/pvw044] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/30/2016] [Indexed: 12/13/2022]
|
26
|
Vries MJ, Bouman HJ, Olie RH, Veenstra LF, Zwaveling S, Verhezen PW, ten Cate-Hoek AJ, ten Cate H, Henskens YM, van der Meijden PE. Determinants of agreement between proposed therapeutic windows of platelet function tests in vulnerable patients. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2016; 3:11-17. [DOI: 10.1093/ehjcvp/pvw026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/18/2022]
|
27
|
Siller-Matula JM, Specht S, Kubica J, Alexopoulos D, De Caterina R, Hobl EL, Jilma B, Christ G, Lang IM. Abciximab as a bridging strategy to overcome morphine-prasugrel interaction in STEMI patients. Br J Clin Pharmacol 2016; 82:1343-1350. [PMID: 27366874 DOI: 10.1111/bcp.13053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 05/18/2016] [Accepted: 06/12/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The present study investigated whether the glycoprotein (GP)IIb/IIIa receptor blocker abciximab might be a successful bridging strategy to achieve adequate levels of platelet inhibition rapidly in cases where prasugrel is used in morphine-pretreated ST-elevation myocardial infarction (STEMI) patients. METHODS In a prospective observational cohort study, 32 patients presenting with STEMI were given prasugrel at a loading dose of 60 mg. Patients were stratified into four groups, according to morphine and/or abciximab use. Adenosine diphosphate (ADP)-induced platelet aggregation was measured at four time points: at baseline, and at 2 h, 1 day and 2 days after prasugrel loading. RESULTS Morphine use was associated with a three-fold higher level of ADP-induced platelet aggregation 2 h after prasugrel loading compared with no morphine/no abciximab (P = 0.019). However, when abciximab was infused in the catheterization laboratory, the effect of morphine on ADP-induced platelet aggregation disappeared (P = 0.884). This interaction was also seen in the presence of high on-treatment platelet reactivity (HTPR) at 2 h; while HTPR was seen in 88% of morphine users/no abciximab users, it was found in only 17-20% in the three other groups (P = 0.003). The effect of morphine disappeared by day 1 - 2. CONCLUSION The infusion of the GPIIb/IIIa receptor blocker abciximab allows immediate and efficient platelet inhibition in STEMI patients concomitantly receiving the oral ADP receptor blocker prasugrel and morphine.
Collapse
Affiliation(s)
| | - Simon Specht
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | | - Raffaele De Caterina
- Institute of Cardiology, "G. d'Annunzio" University - Chieti-Pescara, Chieti, Italy
| | - Eva-Luise Hobl
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Günter Christ
- 5th Medical Department of Cardiology, Kaiser Franz Josef Hospital, Vienna, Austria
| | - Irene M Lang
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Dobrovolsky AB, Laguta PS, Guskova EV, Yarovaya EB, Titaeva EV, Storozhilova AN, Panchenko EP. Effect of Fibrinogen on Platelet Reactivity Measured by the VerifyNow P2Y12 Assay. BIOCHEMISTRY (MOSCOW) 2016; 81:439-44. [PMID: 27297894 DOI: 10.1134/s0006297916050011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The VerifyNow assay is based upon the ability of activated platelets to cross-link beads coated with fibrinogen. However, fibrinogen is an abundant protein of blood, and therefore it may affect test results by competing with fibrinogen of beads for binding to platelets. To test this assumption, we assessed the influence of artificial alteration of fibrinogen level in blood samples obtained from donors (n = 9) and patients on clopidogrel therapy (n = 8) on the results of the VerifyNow P2Y12 assay. Fibrinogen level was altered by adding to blood samples 1/10 volume of fibrinogen solution (10.56 g/liter) or corresponding buffer. Relative to baseline, addition of buffer significantly increased platelet reactivity, whereas addition of fibrinogen decreased it. Analysis of the relationship between change in platelet reactivity values (dBase and dPRU) and change in fibrinogen concentration (dFg) revealed strong negative correlations: dBase = -63.3 × dFg - 27.1 (r = -0.924, p < 0.0005) and dPRU = -54.4 × dFg - 21.8 (r = -0.764, p < 0.0005). Thus, the results of our experiments suggest that: (i) blood fibrinogen strongly influences results of the VerifyNow P2Y12 assay, and (ii) correcting for fibrinogen effect may be needed to improve the accuracy of the test in the measuring of antiplatelet effect of clopidogrel therapy.
Collapse
Affiliation(s)
- A B Dobrovolsky
- Russian Cardiology Research and Production Complex, Ministry of Healthcare, Moscow, 121552, Russia.
| | | | | | | | | | | | | |
Collapse
|
29
|
Impaired P2Y12 inhibition by clopidogrel in kidney transplant recipients: results from a cohort study. BMC Nephrol 2016; 17:58. [PMID: 27278793 PMCID: PMC4899921 DOI: 10.1186/s12882-016-0270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/01/2016] [Indexed: 12/04/2022] Open
Abstract
Background Cardiovascular complications represent a major cause of morbidity and mortality for patients who received kidney transplantation (KT). However, the impact of KT and chronic immunosuppression on platelet response to clopidogrel in patients undergoing coronary or peripheral revascularization procedures remains unclear. This cohort study compares platelet responsiveness to clopidogrel as assessed byvasodilator-stimulated phosphoprotein (VASP) phosphorylation. Methods The study population was divided between chronic kidney disease (CKD) patients who underwent KT (n = 36) and non-transplanted CKD patients (control group, n = 126). Patients were on maintenance antiplatelet therapy with clopidogrel 75 mg daily for at least 8 days. The mean platelet reactivity index (PRI) VASP values and the prevalence of high on-treatment platelet reactivity (HPR, defined as PRI VASP ≥61 %) were compared. Results The mean PRI VASP value was significantly higher in the transplant group (60.1 ± 3 vs 51.2 ± 1.6 %; p=0.014). HPR was significantly more common in the transplant group on clopidogrel maintenance therapy (58 vs. 31 %; p = 0.011). KT was the only independent predictor of HPR (odds ratio: 2.6; 95 % confidence interval: 1.03–6.27, p = 0.03). The effect of treatment with calcineurin inhibitors on clopidogrel response could not be analyzed separately from the kidney transplant status. Conclusions KT is associated with an increased prevalence of HPR. Our results suggest that plateletfunction tests may be clinically useful for the management of this specific population.
Collapse
|
30
|
Jiang M, You JHS. Cost–effectiveness analysis of personalized antiplatelet therapy in patients with acute coronary syndrome. Pharmacogenomics 2016; 17:701-13. [DOI: 10.2217/pgs-2016-0008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed to compare the clinical and economic outcomes of pharmacogenetic-guided (PG-guided) and platelet reactivity testing-guided antiplatelet therapy for patients with acute coronary syndrome undergoing percutaneous coronary intervention. Methods: A decision-analytic model was simulated including four antiplatelet strategies: universal clopidogrel 75 mg daily, universal alternative P2Y12 inhibitor (prasugrel or ticagrelor), PG-guided therapy, and platelet reactivity testing-guided therapy. Results: PG-guided therapy was the preferred option with lowest cost (US$75,208) and highest quality-adjusted life years gained (7.6249 quality-adjusted life years). The base-case results were robust in sensitivity analysis. Conclusion: PG-guided antiplatelet therapy showed the highest probability to be preferred antiplatelet strategy for acute coronary syndrome patients with percutaneous coronary intervention.
Collapse
Affiliation(s)
- Minghuan Jiang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, China SAR
| | - Joyce HS You
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, China SAR
| |
Collapse
|
31
|
Distribution of clinical events across platelet aggregation values in all-comers treated with prasugrel and ticagrelor. Vascul Pharmacol 2016; 79:6-10. [DOI: 10.1016/j.vph.2016.01.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 12/18/2022]
|
32
|
Jilma-Stohlawetz P, Ratzinger F, Schörgenhofer C, Jilma B, Quehenberger P. Effect of preanalytical time-delay on platelet function as measured by multiplate, PFA-100 and VerifyNow. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:249-55. [DOI: 10.3109/00365513.2016.1143115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Komosa A, Siller-Matula J, Lesiak M, Michalak M, Kowal J, Mączyński M, Siniawski A, Mularek-Kubzdela T, Wiśniewski S, Grajek S. Association between high on-treatment platelet reactivity and occurrence of cerebral ischemic events in patients undergoing percutaneous coronary intervention. Thromb Res 2016; 138:49-54. [DOI: 10.1016/j.thromres.2015.12.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/25/2015] [Accepted: 12/23/2015] [Indexed: 11/16/2022]
|
34
|
Weber C, Lip GYH. Editors’ Choice 2015 papers in Thrombosis and Haemostasis. Thromb Haemost 2016; 115:230-2. [DOI: 10.1160/th15-11-0911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 11/05/2022]
|
35
|
Siller-Matula JM, Akca B, Neunteufl T, Maurer G, Lang IM, Kreiner G, Berger R, Delle-Karth G. Inter-patient variability of platelet reactivity in patients treated with prasugrel and ticagrelor. Platelets 2015; 27:373-7. [PMID: 26555925 DOI: 10.3109/09537104.2015.1095874] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study was to evaluate the distribution of platelet reactivity values in patients treated with prasugrel and ticagrelor. This prospective observational study enrolled 200 patients treated with prasugrel or ticagrelor. Platelet aggregation was determined by multiple electrode aggregometry after stimulation with adenosine diphosphate (ADP) in the maintenance phase of treatment with prasugrel or ticagrelor. Only 3% of patients in the prasugrel group and 2% of study participants in the ticagrelor group had high on treatment platelet reactivity (HTPR). The majority of patients displayed low on treatment platelet reactivity (LTPR; prasugrel: 69%; ticagrelor: 64%). The pharmacodynamic effect was similar in patients treated with prasugrel and ticagrelor: the median level of ADP-induced platelet aggregation was 15U (interquartile range IQR 9-21U) under prasugrel treatment and 17U (IQR 8-24U) under ticagrelor treatment (p=0.370). In conclusion, our study suggests that there is some degree of variability in ADP-induced platelet aggregation under treatment with prasugrel and ticagrelor.
Collapse
Affiliation(s)
| | - Betül Akca
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Thomas Neunteufl
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Gerald Maurer
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Irene M Lang
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Gerhard Kreiner
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Rudolf Berger
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| | - Georg Delle-Karth
- a Department of Cardiology , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
36
|
Vries MJA, van der Meijden PEJ, Henskens YMC, ten Cate-Hoek AJ, ten Cate H. Assessment of bleeding risk in patients with coronary artery disease on dual antiplatelet therapy. Thromb Haemost 2015. [DOI: 10.1160/th-15-04-0355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Non-traumatic splenic rupture on dual antiplatelet therapy with aspirin and ticagrelor after stenting for acute coronary syndrome. J Cardiol Cases 2015; 12:65-67. [PMID: 30524542 DOI: 10.1016/j.jccase.2015.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 04/26/2015] [Accepted: 05/10/2015] [Indexed: 12/14/2022] Open
Abstract
We report a case of non-traumatic splenic rupture in a 57-year-old man on dual antiplatelet therapy (DAPT) with aspirin and ticagrelor, seven months after percutaneous coronary intervention and drug-eluting stent implantation for non-ST elevation myocardial infarction. No splenic abnormalities were found at histopathological analysis after splenectomy, and no history of recent trauma was reported. Once restarted, DAPT after splenectomy, assessment of platelet function was performed by light transmittance aggregometry, showing a profound inhibition of platelet function by adenosine diphosphate, arachidonic acid, and collagen. Taking into account the bleeding risk associated with low on-treatment platelet reactivity, and to switch the patient from ticagrelor to a less potent P2Y12 inhibitor such as clopidogrel, cytochrome P450, genetic polymorphisms accounting for clopidogrel response variability were analyzed. The polymorphisms associated with lower response (CYP2C19*2, CYP2C19*3) were absent. Therefore, ticagrelor was withdrawn, and DAPT was continued with aspirin and clopidogrel. Rupture of the spleen may occur in the absence of major trauma or previous splenic diseases, and could be a complication of antithrombotic treatments. Moreover, low on-treatment platelet reactivity during DAPT is emerging as a possible risk factor for bleeding complications, so underlining the usefulness of assessing platelet function in special conditions to ensure that the patient receives the best tailored antiplatelet therapy. <Learning objective: Non-traumatic splenic rupture is a rare event, and is more often associated with pre-existing splenic abnormalities. However, it may be also a complication of medical treatments, especially with antithrombotic drugs. Low on-treatment platelet reactivity is emerging as a possible risk factor for bleeding complications; therefore, assessing platelet function in special conditions could be useful to ensure the patient receives the best-tailored antiplatelet therapy.>.
Collapse
|
38
|
Siller-Matula JM, Petre A, Delle-Karth G, Huber K, Ay C, Lordkipanidzé M, De Caterina R, Kolh P, Mahla E, Gersh BJ. Impact of preoperative use of P2Y12 receptor inhibitors on clinical outcomes in cardiac and non-cardiac surgery: A systematic review and meta-analysis. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2015; 6:753-770. [DOI: 10.1177/2048872615585516] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Alexandra Petre
- Department of Cardiology, Medical University of Vienna, Austria
| | | | - Kurt Huber
- 3rd Medical Department of Cardiology and Emergency Medicine, Wilhelminen Hospital, Vienna, Austria
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Austria
| | - Marie Lordkipanidzé
- Faculty of Pharmacy, University of Montreal; Research Center, Montreal Heart Institute, Canada
| | - Raffaele De Caterina
- Institute of Cardiology, ‘G d’Annunzio’ University – Chieti-Pescara, Chieti, Italy
| | - Philippe Kolh
- Department of Cardiothoracic Surgery, University Hospital of Liege, Belgium
| | - Elisabeth Mahla
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Austria
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, College of Medicine Rochester, USA
| |
Collapse
|
39
|
Personalized antiplatelet therapy with P2Y12 receptor inhibitors: benefits and pitfalls. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2015; 11:259-80. [PMID: 26677375 PMCID: PMC4679793 DOI: 10.5114/pwki.2015.55596] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/07/2023] Open
Abstract
Antiplatelet therapy with P2Y12 receptor inhibitors has become the cornerstone of medical treatment in patients with acute coronary syndrome, after percutaneous coronary intervention and in secondary prevention of atherothrombotic events. Clopidogrel used to be the most broadly prescribed P2Y12 receptor inhibitor with undisputable benefits especially in combination with aspirin, but a considerable number of clopidogrel-treated patients experience adverse thrombotic events in whom insufficient P2Y12-inhibition and a consequential high on-treatment platelet reactivity is a common finding. This clinically relevant limitation of clopidogrel has driven the increased use of new antiplatelet agents. Prasugrel (a third generation thienopyridine) and ticagrelor (a cyclopentyl-triazolo-pyrimidine) feature more potent and predictable P2Y12-inhibition compared to clopidogrel, which translates into improved ischemic outcomes. However, excessive platelet inhibition and consequential low on-treatment platelet reactivity comes at the price of increased risk of major bleeding. The majority of randomized clinical trials failed to demonstrate improved clinical outcomes with platelet function testing and tailored antiplatelet therapy, but results of all recent trials of potent antiplatelets and prolonged antiplatelet durations point towards a need for individualized antiplatelet approach in order to decrease thrombotic events without increasing bleeding. This review focuses on potential strategies for personalizing antiplatelet treatment.
Collapse
|