1
|
Yang G, Alarcon C, Chanfreau C, Lee NH, Friedman P, Nutescu E, Tuck M, O'Brien T, Gong L, Klein TE, Chang K, Tsao PS, Meltzer DO, Lynch JA, Million Veteran Program, Tuteja S, Perera MA. Investigation of Genomic and Transcriptomic Risk Factors of Clopidogrel Response in African Americans. Clin Pharmacol Ther 2025; 117:1313-1324. [PMID: 39868839 PMCID: PMC11993291 DOI: 10.1002/cpt.3552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025]
Abstract
Clopidogrel, an anti-platelet drug, is used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic events, with African Americans (AA) suffering disproportionately. The aim of this study was to discover novel biomarkers of clopidogrel resistance in African Americans using genome and transcriptome data. We conducted a genome-wide association study (GWAS), including local ancestry adjustment, in 141 AA on clopidogrel to identify genetic associations with high on-treatment platelet reactivity (HTPR), with validation of genome-wide significant and suggestive loci in an independent cohort of AA clopidogrel patients (N = 823) from the Million Veteran's Program (MVP) along with in vitro functional analysis. We performed differential gene expression (DGE) analysis in whole blood to identify transcriptomic predictors of response, followed by functional validation in MEG-01 cells. GWAS identified one signal on Chromosome 7 as significantly associated with increasing risk of HTPR. The lead single-nucleotide polymorphism (SNP), rs7807369, within thrombospondin 7A (THSD7A) was associated with an increased risk of HTPR (odds ratio (OR) = 4.02, P = 4.56 × 10-9). Higher THSD7A gene expression was associated with HTPR in an independent cohort of clopidogrel-treated patients (P = 0.004) and carrying a risk allele showed increased gene expression in primary human endothelial cells. Notably, the CYP2C19*2 variants showed no association with clopidogrel response in the discovery or MVP cohorts. DGE analysis identified an association with decreased LAIR1 and AP3B2 expression to HTPR. LAIR1 knockdown in MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of LAIR1 in the Glycoprotein VI pathway. In summary, these findings suggest that other variants and genes outside of CYP2C19 star alleles play an important role in clopidogrel response in AA.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Center for Applied BioinformaticsSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Catherine Chanfreau
- VA Informatics and Computing Infrastructure (VINCI)VA Salt Lake City Health Care SystemSalt Lake CityUtahUSA
| | - Norman H. Lee
- Department of Pharmacology and PhysiologyGeorge Washington UniversityWashingtonDCUSA
- GW Cancer CenterGeorge Washington UniversityWashington, DCUSA
| | - Paula Friedman
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Edith Nutescu
- Department of Pharmacy PracticeRetzky College of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Matthew Tuck
- Washington DC VA Medical CenterWashingtonDCUSA
- The George Washington UniversityWashingtonDCUSA
| | - Travis O'Brien
- Department of Pharmacology and PhysiologyGeorge Washington UniversityWashingtonDCUSA
| | - Li Gong
- Department of Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - Teri E. Klein
- Department of Biomedical Data Science and Department of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Kyong‐Mi Chang
- Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Philip S. Tsao
- VA Palo Alto Healthcare System and Stanford UniversityPalo AltoCaliforniaUSA
| | - David O. Meltzer
- Section of Hospital Medicine, Department of MedicineUniversity of ChicagoChicagoIllinoisUSA
| | - Julie A. Lynch
- VA Informatics and Computing Infrastructure (VINCI)VA Salt Lake City Health Care SystemSalt Lake CityUtahUSA
| | | | - Sony Tuteja
- Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Minoli A. Perera
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
2
|
Song Q, Zhang C, Wang W, Wang C, Yi C. Exploring the genetic landscape of the brain-heart axis: A comprehensive analysis of pleiotropic effects between heart disease and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111172. [PMID: 39423935 DOI: 10.1016/j.pnpbp.2024.111172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The genetic links between heart disease and psychiatric disorders are complex and not well understood. This study uses genome-wide association studies (GWAS) and advanced multilevel analyses to explore these connections. METHODS We analyzed GWAS data from seven psychiatric disorders and five types of heart disease. Genetic correlations and overlaps were examined using linkage disequilibrium score regression (LDSC), high-definition likelihood (HDL), and Genetic analysis incorporating Pleiotropy and Annotation (GPA). Pleiotropic single-nucleotide variations (SNVs) were identified with pleiotropic analysis under the composite null hypothesis (PLACO) and annotated via Functional mapping and annotation of genetic associations (FUMA). Potential pleiotropic genes were identified using Multi-marker Analysis of GenoMic Annotation (MAGMA) and Summary data-based Mendelian Randomization (SMR). RESULTS Among 35 trait pairs, 32 showed significant genetic correlations or overlaps. PLACO identified 15,077 SNVs, with 287 recognized as pleiotropic loci and 20 colocalization sites. MAGMA and SMR revealed 75 potential pleiotropic genes involved in diverse pathways, including cancer, neurodevelopment, and cellular organization. Mouse Genome Informatics (MGI) queries provided evidence linking multiple genes to heart or psychiatric disorders. CONCLUSIONS This analysis reveals loci and genes with pleiotropic effects between heart disease and psychiatric disorders, highlighting shared biological pathways. These findings illuminate the genetic mechanisms underlying the brain-heart axis and suggest shared biological foundations for these conditions, offering potential targets for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Qifeng Song
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cheng Zhang
- Nanjing Vocational Health College, Nanjing, Jiangsu 210000, China
| | - Wei Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cihan Wang
- Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Kankaria A, Majumdar M, Lee S, Hall RP, Suarez Ferreira SP, Lee I, Patel SS, Jessula S, D'Oria M, Dua A. Platelet function testing and clinical outcomes in peripheral arterial disease: Systematic review and narrative synthesis. J Vasc Surg 2024; 80:269-278. [PMID: 38122860 DOI: 10.1016/j.jvs.2023.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE This systematic review aims to comprehensively assess the contemporary literature on platelet function testing (PFT) in individuals undergoing revascularization therapy for peripheral arterial disease (PAD). The goal is to identify whether PFT can aid in detecting antiplatelet resistance, predicting post-procedural thrombotic complications, and informing tailored treatment strategies. METHODS Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a literature review was conducted using PubMed databases. Search terms included relevant medical subject headings (MeSH) terms. Eligible articles published in English between 1990 and 2023 were analyzed. Studies that examined PFT outcomes in patients with PAD after lower extremity revascularization were included. RESULTS Ten studies met the inclusion criteria. Various PFT methods were used, including thromboelastography with platelet mapping, multiplate analyzer, Cytochrome P450 2C19 testing, VerifyNow, corrected whole blood aggregometry, platelet function analyzer-100, and light transmission aggregometry. PFT identified individuals who were resistant or non-sensitive to antiplatelet therapy, with such patients facing increased risks of graft/stent thrombosis, amputation, and reintervention. However, substantial heterogeneity in surgical procedures, drug regimens, and testing methods was observed among the studies. CONCLUSIONS PFTs can play a crucial role in detecting resistance and non-sensitivity to antiplatelet drugs in patients with PAD post-revascularization. However, heterogeneity of data and methods underlines the need for standardized protocols and consensus-building among PFTs. Enhancing clinical utility and reliability could help optimize antiplatelet thromboprophylaxis, minimize thrombotic complications, and improve treatment strategies in vascular surgery. Further research is necessary to solidify the role of PFTs in guiding antiplatelet therapy post-revascularization in patients with PAD.
Collapse
Affiliation(s)
- Aman Kankaria
- School of Medicine and Biosciences, Kansas City University, Kansas City, MO
| | - Monica Majumdar
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Sonia Lee
- Department of Surgery, Tufts Medical Center, Boston, MA
| | - Ryan P Hall
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Sasha P Suarez Ferreira
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Ivy Lee
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Shiv S Patel
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Samuel Jessula
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Mario D'Oria
- Division of Vascular and Endovascular Surgery, University Hospital of Trieste, Trieste, Italy
| | - Anahita Dua
- Division of Vascular and Endovascular Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA.
| |
Collapse
|
4
|
Yang G, Alarcon C, Chanfreau C, Lee NH, Friedman P, Nutescu E, Tuck M, O'Brien T, Gong L, Klein TE, Chang KM, Tsao PS, Meltzer DO, Tuteja S, Perera MA. Investigation of genomic and transcriptomic risk factors in clopidogrel response in African Americans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.05.23299140. [PMID: 38106031 PMCID: PMC10723512 DOI: 10.1101/2023.12.05.23299140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Clopidogrel, an anti-platelet drug, used to prevent thrombosis after percutaneous coronary intervention. Clopidogrel resistance results in recurring ischemic episodes, with African Americans suffering disproportionately. The aim of this study was to identify biomarkers of clopidogrel resistance in African American patients. We conducted a genome-wide association study, including local ancestry adjustment, in 141 African Americans on clopidogrel to identify associations with high on-treatment platelet reactivity (HTPR). We validated genome-wide and suggestive hits in an independent cohort of African American clopidogrel patients (N = 823) from the Million Veteran's Program (MVP) along with in vitro functional follow up. We performed differential gene expression (DGE) analysis in whole blood with functional follow-up in MEG-01 cells. We identified rs7807369, within thrombospondin 7A (THSD7A), as significantly associated with increasing risk of HTPR (p = 4.56 × 10-9). Higher THSD7A expression was associated with HTPR in an independent gene expression cohort of clopidogrel treated patients (p = 0.004) and supported by increased gene expression on THSD7A in primary human endothelial cells carrying the risk haplotype. Two SNPs (rs1149515 and rs191786) were validated in the MVP cohort. DGE analysis identified an association with decreased LAIR1 expression to HTPR. LAIR1 knockdown in a MEG-01 cells resulted in increased expression of SYK and AKT1, suggesting an inhibitory role of LAIR1 in the Glycoprotein VI pathway. Notably, the CYP2C19 variants showed no association with clopidogrel response in the discovery or MVP cohorts. In summary, these finding suggest that other variants outside of CYP2C19 star alleles play an important role in clopidogrel response in African Americans.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | | | - Norman H Lee
- Department of Pharmacology and Physiology, George Washington University, 2300 I Street NW, Washington, DC, 20037, USA
| | - Paula Friedman
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| | - Edith Nutescu
- Department of Pharmacy Practice and Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois Chicago, College of Pharmacy, Chicago, IL
| | - Matthew Tuck
- Washington DC VA Medical Center, Washington, DC and The George Washington University, Washington, DC
| | - Travis O'Brien
- Department of Pharmacology and Physiology, George Washington University, 2300 I Street NW, Washington, DC, 20037, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, CA
| | - Teri E Klein
- Department of Biomedical Data Science and Department of Medicine, Stanford University, Stanford, CA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Philip S Tsao
- VA Palo Alto Healthcare System and Stanford University, Palo Alto, CA
| | - David O Meltzer
- Section of Hospital Medicine, Department of Medicine, University of Chicago, Chicago, IL
| | - Sony Tuteja
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Minoli A Perera
- Department of Pharmacology, Center for Pharmacogenomics, Fienberg School of Medicine, Northwestern University, Chicago IL
| |
Collapse
|
5
|
Marcucci R, Berteotti M, Gragnano F, Galli M, Cavallari I, Renda G, Capranzano P, Santilli F, Capodanno D, Angiolillo DJ, Cirillo P, Calabrò P, Patti G, De Caterina R. Monitoring antiplatelet therapy: where are we now? J Cardiovasc Med (Hagerstown) 2023; 24:e24-e35. [PMID: 36729588 DOI: 10.2459/jcm.0000000000001406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Single antiplatelet therapy represents the cornerstone of thrombosis prevention in atherosclerotic cardiovascular disease. Dual antiplatelet therapy (DAPT), consisting of aspirin plus a P2Y 12 inhibitor, is the standard of care for patients with acute coronary syndrome or undergoing both coronary and peripheral percutaneous interventions. Recent data suggest the efficacy of DAPT also after minor stroke. In this setting, a large body of evidence has documented that genetic and acquired patients' characteristics may affect the magnitude of platelet inhibition induced by antiplatelet agents. The implementation of tools allowing the identification and prediction of platelet inhibition has recently been shown to improve outcomes, leading to an optimal balance between antithrombotic efficacy and bleeding risk. We are therefore clearly moving towards tailored antiplatelet therapy. The aim of this paper is to summarize the available evidence on the evaluation of platelet inhibition in patients with coronary, peripheral, or cerebrovascular atherosclerosis. We will here focus on antiplatelet therapy based on both aspirin and P2Y 12 inhibitors. In addition, we provide practical insights into the clinical settings in which it appears reasonable to implement antiplatelet therapy monitoring.
Collapse
Affiliation(s)
- Rossella Marcucci
- Department of Clinical and Experimental Medicine, University of Florence, Florence
| | - Martina Berteotti
- Department of Clinical and Experimental Medicine, University of Florence, Florence
| | - Felice Gragnano
- Division of Clinical Cardiology, Azienda Ospedaliera di Rilievo Nazionale 'Sant'Anna e San Sebastiano', Caserta
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
| | - Mattia Galli
- Catholic University of the Sacred Heart, Rome
- Maria Cecilia Hospital, GVM Care & Research, Cotignola
| | | | - Giulia Renda
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), G. d'Annunzio University Chieti-Pescara
| | - Piera Capranzano
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico "G. Rodolico-San Marco", University of Catania, Catania
| | - Francesca Santilli
- Department of Medicine and Aging, and Center for Advanced Studies and Technology (CAST), G. d'Annunzio University Chieti-Pescara, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico "G. Rodolico-San Marco", University of Catania, Catania
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Plinio Cirillo
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples
| | - Paolo Calabrò
- Division of Clinical Cardiology, Azienda Ospedaliera di Rilievo Nazionale 'Sant'Anna e San Sebastiano', Caserta
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
| | - Giuseppe Patti
- Maggiore della Carità Hospital, University of Eastern Piedmont, Novara
| | - Raffaele De Caterina
- Department of Surgical, Medical and Molecular Pathology and of Critical Sciences, University of Pisa, Pisa
- Division of Cardiology, Azienda Ospedaliero-Universitaria Pisana, Pisa
- Fondazione VillaSerena per la Ricerca, Città Sant'Angelo-Pescara, Pescara, Italy
| |
Collapse
|
6
|
Zhang Y, Chou JW, Huang WT, Derry K, Humber D. Platelet reactivity testing in peripheral artery disease. Am J Health Syst Pharm 2022; 79:1312-1322. [PMID: 35381075 DOI: 10.1093/ajhp/zxac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DISCLAIMER In an effort to expedite the publication of articles related to the COVID-19 pandemic, AJHP is posting these manuscripts online as soon as possible after acceptance. Accepted manuscripts have been peer-reviewed and copyedited, but are posted online before technical formatting and author proofing. These manuscripts are not the final version of record and will be replaced with the final article (formatted per AJHP style and proofed by the authors) at a later time. PURPOSE Oral antiplatelet therapy is routinely used to prevent adverse cardiovascular events in patients with peripheral artery disease (PAD). Several laboratory tests are available to quantify the degree of platelet inhibition following antiplatelet therapy. This article aims to provide a review of the literature surrounding platelet functional testing in patients with PAD receiving oral P2Y12 inhibitors and to offer guidance to clinicians for the use and interpretation of these tests. SUMMARY A literature search of PubMed and the Web of Science Core Collection database was conducted. All studies that performed platelet function testing and reported clinical outcomes in patients with PAD were included. Evaluation of the data suggests that, among the available testing strategies, the VerifyNow platelet reactivity unit (PRU) test is the most widely used. Despite numerous investigations attempting to define a laboratory threshold indicating suboptimal response to antiplatelet therapy, controversy exists about which PRU value best correlates with cardiovascular outcomes (ie, mortality, stent thrombosis, etc). In the PAD literature, the most commonly used PRU thresholds are 208 or higher and 235 or higher. Nonetheless, adjusting antiplatelet regimens based on suboptimal P2Y12 reactivity values has yet to be proven useful in reducing the incidence of adverse cardiovascular outcomes. This review examines platelet function testing in patients with PAD and discusses the interpretation and application of these tests when monitoring the safety and efficacy of P2Y12 inhibitors. CONCLUSION Although platelet functional tests may be simple to use, clinical trials thus far have failed to show benefit from therapy adjustments based on test results. Clinicians should be cautioned against relying on this test result alone and should instead consider a combination of laboratory, clinical, and patient-specific factors when adjusting P2Y12 inhibitor therapy in clinical practice.
Collapse
Affiliation(s)
- Youqi Zhang
- Department of Pharmacy, UC San Diego Health, La Jolla, CA, USA
| | - Jennifer W Chou
- Department of Pharmacy, UC San Diego Health, La Jolla, CA, USA
| | - Wan-Ting Huang
- Department of Pharmacy, UC San Diego Health, La Jolla, CA, USA
| | - Katrina Derry
- Department of Pharmacy, UC San Diego Health, La Jolla, CA, USA
| | - Doug Humber
- Department of Pharmacy, UC San Diego Health, La Jolla, CA, USA
| |
Collapse
|
7
|
Busch L, Stern M, M'Pembele R, Dannenberg L, Mourikis P, Gröne M, Özaslan G, Heinen Y, Heiss C, Sansone R, Huhn R, Kelm M, Polzin A. Impact of high on-treatment platelet reactivity after angioplasty in patients with critical limb ischemia. Vascul Pharmacol 2021; 141:106925. [PMID: 34619361 DOI: 10.1016/j.vph.2021.106925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/31/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Dual antiplatelet therapy (DAPT) with aspirin and clopidogrel is standard of care in patients with peripheral artery disease (PAD) after percutaneous transluminal angioplasty (PTA). However, high on treatment platelet reactivity (HTPR) to DAPT is frequent and associated with major adverse limb events (MALE) in PAD patients. Nevertheless, association of MALE and HTPR in patients with critical limb ischemia (CLI) is not known. Moreover, comorbidities might confound response to antiplatelet medication further. Hence, in this trial we analyzed pharmacodynamic responses to DAPT and clinical events in CLI patients post PTA. METHODS In this prospective single center pilot analysis, we included 71 CLI patients. Patients received DAPT after PTA. Antiplatelet effect were measured by light transmission aggregometry (LTA) and vasodilator-stimulated protein phosphorylation assay (VASP). MALE, major adverse cardiac and cerebrovascular events (MACCE) and BARC bleeding within 12 months follow-up were assessed. RESULTS Mean age of patients was 73.37 ± 7.36 years and 47 (66.2%) were male. Overall HTPR appeared in 46 patients (64.8%). MALE and MACCE showed no differences between patients with and patients without HTPR. However, bleeding was higher in patients with sufficient pharmacodynamic response to DAPT (Bleeding - HTPR: 13.4% vs. no HTPR: 36.0%; log-rank HR: 0.32; 95% CI 0.1079 to 0.9396 p = 0.0217). This finding remained robust in multivariate analysis. CONCLUSION HTPR to DAPT is frequent in CLI patients. However, bleeding was higher in patients with sufficient response to DAPT. Ischemic events did not differ. Hence, CLI patients might benefit from an alternative antithrombotic approach.
Collapse
Affiliation(s)
- Lucas Busch
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Manuel Stern
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - René M'Pembele
- Departement of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Lisa Dannenberg
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Philipp Mourikis
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Michael Gröne
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Göksen Özaslan
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Yvonne Heinen
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Christian Heiss
- University of Surrey, Faculty of Health and Medical Sciences, Guildford, United Kingdom
| | - Roberto Sansone
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Ragnar Huhn
- Departement of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Amin Polzin
- Cardiovascular Research Institute Düsseldorf (CARID), Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany.
| |
Collapse
|
8
|
Javalkar V, Kuybu O, Amireh A, Kelley RE. Evolving Approaches to Antithrombotics in Stroke Prevention and Treatment. South Med J 2020; 113:585-592. [PMID: 33140113 DOI: 10.14423/smj.0000000000001173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The optimization of antithrombotic therapy for acute stroke treatment and secondary prevention is an evolving process based on an increasing array of studies that provide an evidence-based approach. Options have increased dramatically with the release of the non-vitamin K oral anticoagulants and with the results of recent randomized clinical trials designed to assess potential benefits versus risks for patients in an individualized fashion. Recent studies have provided important information to guide choice and dosing of antiplatelet agents as well as the length of treatment. Anticoagulant use is particularly pertinent for stroke prevention in patients at higher risk of atrial fibrillation and may have a place in certain other stroke mechanisms. One important focus of study is the potential benefit of combined antiplatelet and anticoagulant therapy. Options for our patients, when the initial choice of therapy does not demonstrate benefit or is not well tolerated, clearly, are valuable. For example, short-term dual antiplatelet therapy for minor stroke and transient ischemic attack is being adopted, but with the recognition that longer-term combined therapy is not worth the increased risk of bleeding. Alternative antiplatelet choices, such as cilostazol and possibly ticagrelor, may be of benefit for refractory patients and this could affect the decision-making process. This review represents an effort to incorporate the information from more recent stroke prevention and treatment studies with information gleaned from prior studies.
Collapse
Affiliation(s)
| | - Okkes Kuybu
- From the Department of Neurology, Louisiana State University Health, Shreveport
| | - Abdallah Amireh
- From the Department of Neurology, Louisiana State University Health, Shreveport
| | - Roger E Kelley
- From the Department of Neurology, Louisiana State University Health, Shreveport
| |
Collapse
|
9
|
Khan H, Gallant R, Jain S, Al-Omran M, De Mestral C, Greco E, Wheatcroft M, Alazonni A, Abdin R, Rand ML, Ni H, Qadura M. Ticagrelor as an Alternative Antiplatelet Therapy in Cardiac Patients Non-Sensitive to Aspirin. ACTA ACUST UNITED AC 2020; 56:medicina56100519. [PMID: 33023261 PMCID: PMC7600331 DOI: 10.3390/medicina56100519] [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] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022]
Abstract
Background and Objectives: Aspirin (acetylsalicylic acid-ASA) is a first-line antiplatelet therapy provided to patients with coronary artery disease (CAD). However, it has been demonstrated that 20-30% of these patients are non-sensitive to their ASA therapy. ASA non-sensitivity is a phenomenon where low-dose ASA (81-325 mg) does not completely inhibit arachidonic-acid-induced platelet aggregation, putting patients at risk of adverse cardio-thrombotic events. Ticagrelor is a P2Y12 receptor inhibitor and alternative antiplatelet that has been approved to reduce the risk of stroke, myocardial infarction, and overall cardiovascular-related death. In this study, we aimed to identify ASA non-sensitive patients and evaluate if they would be sensitive to ticagrelor. Materials and Methods: For this pilot study, thirty-eight patients with CAD taking 81 mg ASA were recruited. Blood samples were collected from each patient and platelet rich plasma (PRP) from each sample was isolated. Light-transmission aggregometry (LTA) was used to determine baseline ASA sensitivity in each patient using 0.5 mg/mL arachidonic acid as a platelet agonist. Patients with ≥20% maximal platelet aggregation after activation were considered ASA non-sensitive. Fresh PRP samples from all patients were then spiked with a clinical dosage of ticagrelor (3 μM-approximately equivalent to a loading dose of 180 mg ticagrelor). Sensitivity was determined using LTA and 5 μM ADP as a platelet agonist. Patients with ≥46% maximal platelet aggregation were considered ticagrelor non-sensitive. Results: Of the 38 CAD patients taking 81 mg ASA, 32% (12/38) were non-sensitive to their 81 mg ASA therapy. All 38 of the recruited patients (100%) were sensitive to ticagrelor ex vivo. In conclusion, we were able to identify ASA non-sensitivity using LTA and determine that ASA non-sensitive patients were sensitive to ticagrelor. Conclusions: Our results suggest that ticagrelor is a promising alternative therapy for patients who are non-sensitive to ASA.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
| | - Reid Gallant
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (R.G.); (H.N.)
| | - Shubha Jain
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
| | - Mohammed Al-Omran
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (R.G.); (H.N.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Charles De Mestral
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (R.G.); (H.N.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Elisa Greco
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Mark Wheatcroft
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ashraf Alazonni
- Division of Cardiology, Scarborough Health Network, Toronto, ON M1P 2T7, Canada;
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Margaret L. Rand
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Departments of Biochemistry and Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
- Translational Medicine, Research Institute; Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Heyu Ni
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (R.G.); (H.N.)
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (S.J.); (M.A.-O.); (C.D.M.); (E.G.); (M.W.)
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (R.G.); (H.N.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Correspondence: ; Tel.: +1-416-864-6047
| |
Collapse
|
10
|
Scudiero F, Valenti R, Marcucci R, Sanna GD, Gori AM, Migliorini A, Vitale R, Giusti B, De Vito E, Corda G, Paniccia R, Zirolia D, Canonico ME, Parodi G. Platelet Reactivity in Hepatitis C Virus-Infected Patients on Dual Antiplatelet Therapy for Acute Coronary Syndrome. J Am Heart Assoc 2020; 9:e016441. [PMID: 32885738 PMCID: PMC7726996 DOI: 10.1161/jaha.120.016441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Coronary artery disease (CAD) has been recognized as a serious and potentially life‐threatening complication of Hepatitis C Virus (HCV) infection. High on‐treatment platelet reactivity has been associated with high risk of ischemic events in patients with CAD, but data regarding the association with HCV infection are still lacking. This post hoc analysis aims to assess high on‐treatment platelet reactivity, severity of CAD, and long‐term outcomes of patients with acute coronary syndrome (ACS) who were infected with HCV. Methods and Results Patients with ACS who were infected with HCV (n=47) were matched to patients with ACS and without HCV (n=137) for age, sex, diabetes mellitus, hypertension, and renal function. HCV‐infected patients with ACS had higher levels of platelet reactivity (ADP10–light transmittance aggregometry, 56±18% versus 44±22% [P=0.002]; arachidonic acid–light transmittance aggregometry, 25±21% versus 16±15% [P=0.011]) and higher rates of high on‐treatment platelet reactivity on clopidogrel and aspirin compared with patients without HCV. Moreover, HCV‐infected patients with ACS had higher rates of multivessel disease (53% versus 30%; P=0.004) and 3‐vessel disease (32% versus 7%; P<0.001) compared with patients without HCV. At long‐term follow‐up, estimated rates of major adverse cardiovascular events (cardiac death, nonfatal myocardial infarction, and ischemia‐driven revascularization) were 57% versus 34% (P=0.005) in HCV‐ and non–HCV‐infected patients with ACS, respectively. In addition, thrombolysis In Myocardial Infarction (TIMI) major bleeding rates were higher in HCV‐infected patients (11% versus 3%; P=0.043) compared with noninfected patients. Multivariable analysis demonstrated that HCV infection was an independent predictor of high on‐treatment platelet reactivity, severity of CAD, and long‐term outcome. Conclusions In this hypothesis‐generating study, patients with ACS and HCV infection showed increased on‐treatment platelet reactivity, more severe CAD, and worse prognosis compared with patients without HCV.
Collapse
Affiliation(s)
- Fernando Scudiero
- Department of Clinical and Experimental Medicine University of Florence Italy.,Cardiology Unit ASST Bergamo est Bolognini Hospital Seriate Italy
| | - Renato Valenti
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Rossella Marcucci
- Department of Clinical and Experimental Medicine University of Florence Italy
| | | | - Anna Maria Gori
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Angela Migliorini
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Raffaele Vitale
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Betti Giusti
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Elena De Vito
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Giulia Corda
- Cardiology Clinic Sassari University Hospital Sassari Italy.,Department of Medical, Surgical and Experimental Sciences Sassari University Sassari Italy
| | - Rita Paniccia
- Department of Clinical and Experimental Medicine University of Florence Italy
| | - Davide Zirolia
- Cardiology Clinic Sassari University Hospital Sassari Italy.,Department of Medical, Surgical and Experimental Sciences Sassari University Sassari Italy
| | - Mario E Canonico
- Cardiology Clinic Sassari University Hospital Sassari Italy.,Department of Medical, Surgical and Experimental Sciences Sassari University Sassari Italy
| | - Guido Parodi
- Cardiology Clinic Sassari University Hospital Sassari Italy.,Department of Medical, Surgical and Experimental Sciences Sassari University Sassari Italy
| |
Collapse
|
11
|
Hulshof AM, Vries M, Verhezen P, Wetzels R, Haartmans M, Olie R, Ten Cate H, Henskens Y. The Influence of Prostaglandin E1 and Use of Inhibitor Percentage on the Correlation between the Multiplate and VerifyNow in Patients on Dual Antiplatelet Therapy. Platelets 2020; 32:463-468. [PMID: 32314928 DOI: 10.1080/09537104.2020.1754378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Platelet function tests (PFT), such as the Multiple Electrode Analyzer (Multiplate) and VerifyNow, show little concordance in patients using antiplatelet drugs. A major difference between these tests is the use of prostaglandin E1 (PGE1) to inhibit P2Y1-platelet-receptor activation in VerifyNow and is proposed to be of influence in the discrepancy between these tests. We aimed to investigate whether the presence of PGE1 could provide an explanation for the moderate correlation and concordance between Multiplate and VerifyNow by adding PGE1 to the Multiplate ADP assay, also known as the ADP-high sensitivity (ADP-HS) assay. We also aimed to investigate whether the difference in baseline platelet function as measured by the VerifyNow and Multiplate could (partly) explain the moderate correlation between the tests, by plotting ADP assay results against baseline function as measured by the corresponding device, which is expressed as the 'inhibitor percentage.' Fifty-one patients who underwent percutaneous coronary intervention (PCI) received dual antiplatelet therapy and were considered to have a high risk of ischemic or bleeding complications were included. The addition of 20 µl PGE1 in the Multiplate resulted in a significant reduction in Arbitrary Aggregation Units, but did not improve correlation with the VerifyNow. The correlation between VerifyNow and Multiplate inhibitor percentage was moderate. Based on these results, we concluded that neither PGE1 nor the calculation of the inhibitor percentage greatly influenced the correlation between PFTs.
Collapse
Affiliation(s)
| | - Minka Vries
- Central Diagnostic Laboratory, Maastricht UMC+, Maastricht, Netherland
| | - Paul Verhezen
- Central Diagnostic Laboratory, Maastricht UMC+, Maastricht, Netherland
| | - Rick Wetzels
- Central Diagnostic Laboratory, Maastricht UMC+, Maastricht, Netherland
| | - Mirella Haartmans
- Department of Orthopedic Surgery, Maastricht UMC+, Maastricht, Netherlands
| | - Renske Olie
- Internal Vascular Medicine, Maastricht UMC+, Maastricht, Netherlands
| | - Hugo Ten Cate
- Internal Vascular Medicine, Maastricht UMC+, Maastricht, Netherlands
| | - Yvonne Henskens
- Central Diagnostic Laboratory, Maastricht UMC+, Maastricht, Netherland
| |
Collapse
|
12
|
Zou X, Deng XL, Wang YM, Li JH, Liu L, Huang X, Liu L, Cao J, Fan L. Genetic polymorphisms of high platelet reactivity in Chinese patients with coronary heart disease under clopidogrel therapy. Int J Clin Pharm 2020; 42:158-166. [PMID: 32253660 DOI: 10.1007/s11096-019-00953-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The variability in the clinical response to clopidogrel treatment has been attributed to genetic factors, but the specific genes and other risk factors remain unclear. OBJECTIVE To investigate the incidence of high on-treatment platelet reactivity in coronary heart disease patients following clopidogrel therapy by analyzing the correlation between genetic polymorphisms and high on-treatment platelet reactivity. SETTING This study was conducted in the Chinese People's Liberation Army (PLA) general hospital. METHOD 578 patients with coronary heart disease undergoing percutaneous transluminal coronary intervention treatment were enrolled. They received dual antiplatelet therapy with aspirin (300 mg) plus clopidogrel (300 mg) over 24 h, or aspirin (100 mg/day) and clopidogrel (75 mg/day) over 3 days. Patients were divided into two groups according to the adenosine diphosphate inhibition rate. The follow-up lasted at least 12 months and adverse endpoint events were recorded. MAIN OUTCOME MEASURE The single nucleotide polymorphisms were detected by MassArray genotyping system. RESULTS The incidence of HTPR was 15.74% in total, being higher in females than in males (24.29% vs. 13.01%, P < 0.01). Diabetes mellitus, homocysteine and high sensitivity C-reactive protein (hs-CRP) levels were significantly higher in the HTPR group than those in the non-HTPR group (P < 0.05). Polymorphisms of rs1057910 (OR 2.90, P = 0.003), rs2246709 (OR 0.69, P = 0.039), and rs776746 (OR 0.66, P = 0.034) were associated with the incidence of high on-treatment platelet reactivity. Female patients were prone to polymorphisms of rs1057910 (OR 3.24, P = 0.004) and rs776746 (OR 0.57, P = 0.025). Compared to non-high on-treatment platelet reactivity group, no differences in high reactivity group were observed with coexisting single nucleotide polymorphisms (14.6% vs. 14.8%, P > 0.05). The adverse endpoint events were significantly higher in the high on-treatment platelet reactivity group than in the non-treatment reactivity group. The survival analysis showed that high on-treatment platelet reactivity was significantly associated with the risk of the endpoint events (P = 0.0219). CONCLUSION Gender (female), diabetes mellitus, high levels of homocysteine and hs-CRP were risk factors for high on-treatment platelet reactivity, and high reactivity was a strong predictor for adverse endpoint events in the coronary heart disease patients. The polymorphism of rs1057910 was a risk factor of high on-treatment platelet reactivity while rs2246709 and rs776746 polymorphisms were protective factors, and coexisting single nucleotide polymorphisms didn't increase the incidence of high on-treatment platelet reactivity.
Collapse
Affiliation(s)
- Xiao Zou
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Xin-Li Deng
- Department of Laboratory, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Yin-Meng Wang
- Department of Respiratory, Clifford Hospital of Guangdong, Panyu, People's Republic of China
| | - Jian-Hua Li
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Lin Liu
- Department of Respiratory, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Xin Huang
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Lu Liu
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Jian Cao
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Li Fan
- Department of Cardiology, National Clinic Research Center for Geriatric Disease, Second Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
13
|
Buchtele N, Herkner H, Schörgenhofer C, Merrelaar A, Laggner R, Gelbenegger G, Spiel AO, Domanovits H, Lang I, Jilma B, Schwameis M. High Platelet Reactivity after Transition from Cangrelor to Ticagrelor in Hypothermic Cardiac Arrest Survivors with ST-Segment Elevation Myocardial Infarction. J Clin Med 2020; 9:jcm9020583. [PMID: 32098088 PMCID: PMC7073541 DOI: 10.3390/jcm9020583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
Transition from cangrelor to oral P2Y12 inhibitors after PCI carries the risk of platelet function recovery and acute stent thrombosis. Whether the recommended transition regimen is appropriate for hypothermic cardiac arrest survivors is unknown. We assessed the rate of high platelet reactivity (HPR) after transition from cangrelor to ticagrelor in hypothermic cardiac arrest survivors. Adult survivors of out-of-hospital cardiac arrest with ST-segment elevation myocardial infarction (STEMI), who were treated for hypothermia (33 °C ± 1) and received intravenous cangrelor during PCI and subsequent oral loading with 180mg ticagrelor were enrolled in this prospective observational cohort study. Platelet function was assessed using whole blood aggregometry. HPR was defined as AUC > 46U. The primary endpoint was the rate of HPR (%) at predefined time points during the first 24 h after cangrelor cessation. Poisson regression was used to estimate the relationship between the overlap time of cangrelor and ticagrelor co-administration and the number of subsequent HPR episodes, expressed as incidence rate ratio (IRR) with 95% confidence interval (95%CI). Between December 2017 and October 2019 16 patients (81% male, 58 years) were enrolled. On average, ticagrelor was administered 39 min (IQR 5-50) before the end of cangrelor infusion. The rate of HPR was highest 90 min after cangrelor cessation and was present in 44% (7/16) of patients. The number of HPR episodes increased significantly with decreasing overlap time of cangrelor and ticagrelor co-administration (IRR 1.03, 95%CI 1.01-1.05; p = 0.005). In this selected cohort of hypothermic cardiac arrest survivors who received cangrelor during PCI, ticagrelor loading within the recommended time frame before cangrelor cessation resulted in a substantial amount of patients with HPR.
Collapse
Affiliation(s)
- Nina Buchtele
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (N.B.); (C.S.); (G.G.); (B.J.)
- Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Harald Herkner
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria; (H.H.); (A.M.); (H.D.); (M.S.)
| | - Christian Schörgenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (N.B.); (C.S.); (G.G.); (B.J.)
| | - Anne Merrelaar
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria; (H.H.); (A.M.); (H.D.); (M.S.)
| | - Roberta Laggner
- Department of Orthopedics and Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (N.B.); (C.S.); (G.G.); (B.J.)
| | - Alexander O. Spiel
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria; (H.H.); (A.M.); (H.D.); (M.S.)
- Correspondence: ; Tel.: +43-1-40-400-39560
| | - Hans Domanovits
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria; (H.H.); (A.M.); (H.D.); (M.S.)
| | - Irene Lang
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (N.B.); (C.S.); (G.G.); (B.J.)
| | - Michael Schwameis
- Department of Emergency Medicine, Medical University of Vienna, 1090 Vienna, Austria; (H.H.); (A.M.); (H.D.); (M.S.)
| |
Collapse
|
14
|
Liu J, Zhang L, Liu M. Mechanisms supporting potential use of bone marrow-derived mesenchymal stem cells in psychocardiology. Am J Transl Res 2019; 11:6717-6738. [PMID: 31814884 PMCID: PMC6895510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Despite great efforts made in recent years, globally cardiovascular disease (CVD) remains the most common and devastating disease. Pharmacological, interventional and surgical treatments have proved to be only partly satisfactory for the majority of patients. A major underlying cause of poor prognosis is a high comorbidity rate between CVD and mental illness, which calls for the approaches of psychocardiology. As psychiatric disorders and CVD can influence each other bidirectionally, it is necessary to develop novel therapies targeting both systems simultaneously. Therefore, innovative stem cell (SC) therapy has become the most promising treatment strategy in psychocardiology. Bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs), among all different types of SCs, have drawn the most attention due to unique advantages in terms of ethical considerations, low immunogenicity and simplicity of preparation. In this review, we survey recent publications and clinical trials to summarize the knowledge and progress gained so far. Moreover, we discuss the feasibility of the clinical application of BM-MSCs in the area of psychocardiology.
Collapse
Affiliation(s)
- Jianyang Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Lijun Zhang
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Meiyan Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| |
Collapse
|
15
|
Halbach M, Baldus S. [Platelet inhibition in patients with coronary, cerebral and peripheral macroangiopathy : What, when and how long?]. Internist (Berl) 2019; 59:288-303. [PMID: 29340741 DOI: 10.1007/s00108-017-0362-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Inhibition of platelet aggregation can reduce the rate of vascular events in patients with coronary artery disease, carotid artery stenosis and symptomatic peripheral arterial disease. The choice of platelet inhibitors in monotherapy and combination therapy as well as the duration of dual platelet inhibition depend on the clinical situation and individual patient characteristics. GOAL The present review summarizes the latest data from clinical trials and recommendations regarding platelet inhibition in coronary, cerebral and peripheral arterial disease. DATA A large number of randomized trials on platelet inhibition in different clinical situations have been performed, allowing evidence-based recommendations on the choice of drugs and duration of treatment. Moreover, new guidelines of European professional societies on platelet inhibition in patients with coronary, cerebral and peripheral arterial disease have been recently published. CONCLUSION Based on latest randomized trials and major society guidelines, a number of recommendations on platelet inhibition in stable coronary artery disease, after stent implantation, after acute coronary syndromes and in cerebral and peripheral arterial disease can be made.
Collapse
Affiliation(s)
- M Halbach
- Klinik III für Innere Medizin, Herzzentrum, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - S Baldus
- Klinik III für Innere Medizin, Herzzentrum, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| |
Collapse
|
16
|
De Hert M, Detraux J, Vancampfort D. The intriguing relationship between coronary heart disease and mental disorders. DIALOGUES IN CLINICAL NEUROSCIENCE 2018. [PMID: 29946209 PMCID: PMC6016051 DOI: 10.31887/dcns.2018.20.1/mdehert] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Coronary heart disease (CHD) and mental illness are among the leading causes of morbidity and mortality worldwide. Decades of research has revealed several, and sometimes surprising, links between CHD and mental illness, and has even suggested that both may actually cause one another. However, the precise nature of these links has not yet been clearly established. The goal of this paper, therefore, is to comprehensively review and discuss the state-of-the-art nature of the epidemiological and pathophysiological aspects of the bidirectional links between mental illness and CHD. This review demonstrates that there exists a large body of epidemiological prospective data showing that people with severe mental illness, including schizophrenia, bipolar disorder, and major depressive disorder, as a group, have an increased risk of developing CHD, compared with controls [adjusted hazard ratio (adjHR)=1.54; 95% CI: 1.30-1.82, P<0.0001]. Anxiety symptoms or disorders (Relative Risk (RR)=1.41, 95% CI: 1.23-1.61, P<0.0001), as well as experiences of persistent or intense stress or posttraumatic stress disorder (PTSD) (adjHR=1.27, 95% CI: 1.08-1.49), although to a lesser degree, may also be independently associated with an increased risk of developing CHD. On the other hand, research also indicates that these symptoms/mental diseases are common in patients with CHD and may be associated with a substantial increase in cardiovascular morbidity and mortality. Finally, mental diseases and CHD appear to have a shared etiology, including biological, behavioral, psychological, and genetic mechanisms.
Collapse
Affiliation(s)
- Marc De Hert
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, Belgium, KU Leuven University of Leuven, Kortenberg, Belgium
| | - Johan Detraux
- Department of Neurosciences, KU Leuven University Psychiatric Centre, Kortenberg, KU Leuven University of Leuven, Kortenberg, Belgium
| | - Davy Vancampfort
- Department of Rehabilitation Sciences, KU Leuven University of Leuven, Leuven, Belgium, KU Leuven University of Leuven, Kortenberg, Belgium
| |
Collapse
|
17
|
On-Treatment Platelet Reactivity is a Predictor of Adverse Events in Peripheral Artery Disease Patients Undergoing Percutaneous Angioplasty. Eur J Vasc Endovasc Surg 2018; 56:545-552. [PMID: 30025662 DOI: 10.1016/j.ejvs.2018.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 06/13/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Few data are available on the association between a different entity of platelet inhibition on antiplatelet treatment and clinical outcomes in patients with peripheral artery disease (PAD). The aim of this study was to evaluate the degree of on-treatment platelet reactivity, and its association with ischaemic and haemorrhagic adverse events at follow up in PAD patients undergoing percutaneous transluminal angioplasty (PTA). METHODS In this observational, prospective, single centre study, 177 consecutive patients with PAD undergoing PTA were enrolled, and treated with dual antiplatelet therapy with aspirin and a P2Y12 inhibitor. Platelet function was assessed on blood samples obtained within 24 h from PTA by light transmission aggregometry (LTA) using arachidonic acid (AA) and adenosine diphosphate (ADP) as agonists of platelet aggregation. High on-treatment platelet reactivity (HPR) was defined by LTA ≥ 20% if induced by AA, and LTA ≥ 70% if induced by ADP. Follow up was performed to record outcomes (death, major amputation, target vessel re-intervention, acute myocardial infarction and/or myocardial revascularisation, stroke/TIA, and bleeding). RESULTS HPR by AA and HPR by ADP were found in 45% and 32% of patients, respectively. During follow up (median duration 23 months) 23 deaths (13%) were recorded; 27 patients (17.5%) underwent target limb revascularisation (TLR), two (1.3%) amputation, and six (3.9%) myocardial revascularisation. Twenty-four patients (15.6%) experienced minor bleeding. On multivariable analysis, HPR by AA and HPR by ADP were independent predictors of death [HR 3.8 (1.2-11.7), p = .023 and HR 4.8 (1.6-14.5), p = .006, respectively]. The median value of LTA by ADP was significantly lower in patients with bleeding complications than in those without [26.5% (22-39.2) vs. 62% (44.5-74), p < .001). LTA by ADP ≤ 41% was independently associated with bleeding HR 14.6 (2.6-24.0), p = .001] on multivariable analysis. CONCLUSIONS In this study a high prevalence of on-clopidogrel and aspirin high platelet reactivity was found, which was significantly associated with the risk of death. Conversely, a low on-clopidogrel platelet reactivity was associated with a higher risk of bleeding. These results document that the entity of platelet inhibition is associated with both thrombotic and bleeding complications in PAD patients.
Collapse
|
18
|
Shatila W, Krajcer Z. Peripheral artery disease: How do genes and pharmacology interplay? Catheter Cardiovasc Interv 2018; 91:1318-1319. [DOI: 10.1002/ccd.27649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Wassim Shatila
- Department of Cardiology; Texas Heart Institute, 6770 Bertner Avenue; Houston Texas
- Division of Cardiology; Baylor College of Medicine, 1 Baylor Plaza; Houston Texas
- Department of Internal Medicine; Baylor College of Medicine, 1 Baylor Plaza; Houston Texas
- CHI St. Luke's Health-Baylor St. Luke's Medical Center; Houston Texas
| | - Zvonimir Krajcer
- Department of Cardiology; Texas Heart Institute, 6770 Bertner Avenue; Houston Texas
- Division of Cardiology; Baylor College of Medicine, 1 Baylor Plaza; Houston Texas
- Department of Internal Medicine; Baylor College of Medicine, 1 Baylor Plaza; Houston Texas
- CHI St. Luke's Health-Baylor St. Luke's Medical Center; Houston Texas
| |
Collapse
|
19
|
Hernandez-Suarez DF, Tomassini-Fernandini JC, Cuevas A, Rosario-Berrios AN, Nuñez-Medina HJ, Padilla-Arroyo D, Rivera N, Liriano J, Vega-Roman RK, Renta JY, Melin K, Duconge J. Clinical Relevant Polymorphisms Affecting Clopidogrel Pharmacokinetics and Pharmacodynamics: Insights from the Puerto Rico Newborn Screening Program. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1115. [PMID: 29848980 PMCID: PMC6025039 DOI: 10.3390/ijerph15061115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 05/28/2018] [Indexed: 12/19/2022]
Abstract
Background: Variations in several clopidogrel-pharmacogenes have been linked to clopidogrel response variability and clinical outcomes. We aimed to determine the frequency distribution of major polymorphisms on CYP2C19, PON1, ABCB1 and P2RY12 pharmacogenes in Puerto Ricans. Methods: This was a cross-sectional, population-based study of 200 unrelated "Guthrie" cards specimens from newborns registered in the Puerto Rican newborn screening program (PRNSP) between 2004 and 2014. Taqman® SNP assay techniques were used for genotyping. Results: Minor allele frequencies (MAF) were 46% for PON1 (rs662), 41% for ABCB1 (rs1045642), 14% for CYP2C19*17, 13% for CYP2C19*2, 12% for P2RY12-H2 and 0.3% for CYP2C19*4. No carriers of the CYP2C19*3 variants were detected. All alleles and genotype proportions were found to be in Hardy⁻Weinberg equilibrium (HWE). Overall, there were no significant differences between MAFs of these variants in Puerto Ricans and the general population (n = 453) of the 1000 Genome project, except when comparisons to each individual parental group were performed (i.e., Africans, Europeans and East-Asians; p < 0.05). As expected, the prevalence of these markers in Puerto Ricans most resembled those in the 181 subjects from reference populations of the Americas. Conclusions: These prevalence data provide a necessary groundwork for future clinical studies of clopidogrel pharmacogenetics in Caribbean Hispanics.
Collapse
Affiliation(s)
- Dagmar F Hernandez-Suarez
- Division of Cardiovascular Medicine, School of Medicine, University of Puerto Rico Medicine Sciences Campus, P.O. Box 365067, San Juan, PR 00936-5067, USA.
| | | | - Angelica Cuevas
- Department of Biology, Natural Sciences, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA.
| | - Anyelis N Rosario-Berrios
- Department of Biology, Natural Sciences, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA.
| | - Héctor J Nuñez-Medina
- Division of Cardiovascular Medicine, School of Medicine, University of Puerto Rico Medicine Sciences Campus, P.O. Box 365067, San Juan, PR 00936-5067, USA.
| | - Dariana Padilla-Arroyo
- Pharmaceutical Sciences Department, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA.
| | - Nannette Rivera
- Department of Biology, Natural Sciences, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA.
| | - Jennifer Liriano
- Department of Biology, Natural Sciences, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA.
| | - Rocio K Vega-Roman
- Department of Biology, Natural Sciences, University of Puerto Rico Bayamon Campus, Bayamon, PR 00959, USA.
| | - Jessicca Y Renta
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA.
| | - Kyle Melin
- Department of Pharmacy Practice, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA.
| | - Jorge Duconge
- Pharmaceutical Sciences Department, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA.
| |
Collapse
|
20
|
Kubisa MJ, Jezewski MP, Gasecka A, Siller-Matula JM, Postuła M. Ticagrelor - toward more efficient platelet inhibition and beyond. Ther Clin Risk Manag 2018; 14:129-140. [PMID: 29398917 PMCID: PMC5775739 DOI: 10.2147/tcrm.s152369] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Novel antiplatelet drugs, including ticagrelor, are being successively introduced into the therapy of atherothrombotic conditions due to their superiority over a standard combination of clopidogrel with acetylsalicylic acid in patients with acute coronary syndromes (ACS). A P2Y12 receptor antagonist, ticagrelor, is unique among antiplatelet drugs, because ticagrelor inhibits the platelet P2Y12 receptor in a reversible manner, and because it demonstrates a wide palette of advantageous pleiotropic effects associated with the increased concentration of adenosine. The pleiotropic effects of ticagrelor comprise cardioprotection, restoration of the myocardium after an ischemic event, promotion of the release of anticoagulative factors and, eventually, anti-inflammatory effects. Beyond the advantageous effects, the increased concentration of adenosine is responsible for some of ticagrelor's adverse effects, including dyspnea and bradycardia. Large-scale clinical trials demonstrated that both standard 12-month therapy and long-term use of ticagrelor reduce the risk of cardiovascular events in patients with ACS, but at the expense of a higher risk of major bleeding. Further trials focused on the use of ticagrelor in conditions other than ACS, including ischemic stroke, peripheral artery disease and status after coronary artery bypass grafting. The results of these trials suggest comparable efficacy and safety of ticagrelor and clopidogrel in extra-coronary indications, but firm conclusions are anticipated from currently ongoing studies. Here, we summarize current evidence on the superiority of ticagrelor over other P2Y12 antagonists in ACS, discuss the mechanism underlying the drug-drug interactions and pleiotropic effects of ticagrelor, and present future perspectives of non-coronary indications for ticagrelor.
Collapse
Affiliation(s)
- Michał J Kubisa
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Mateusz P Jezewski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Aleksandra Gasecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Vesicle Observation Centre, Laboratory of Experimental Clinical Chemistry, Academic Medical Centre, University of Amsterdam, the Netherlands
| | | | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| |
Collapse
|
21
|
Antiplatelet Effect of Different Loading Doses of Ticagrelor in Patients With Non–ST-Elevation Acute Coronary Syndrome Undergoing Percutaneous Coronary Intervention: The APELOT Trial. Can J Cardiol 2017; 33:1675-1682. [DOI: 10.1016/j.cjca.2017.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 12/27/2022] Open
|
22
|
Schoergenhofer C, Hobl EL, Schellongowski P, Heinz G, Speidl WS, Siller-Matula JM, Schmid M, Sunder-Plaßmann R, Stimpfl T, Hackl M, Jilma B. Clopidogrel in Critically Ill Patients. Clin Pharmacol Ther 2017; 103:217-223. [PMID: 28913918 PMCID: PMC5813104 DOI: 10.1002/cpt.878] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/21/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022]
Abstract
Only limited data are available regarding the treatment of critically ill patients with clopidogrel. This trial investigated the effects and the drug concentrations of the cytochrome P450 (CYP450) activated prodrug clopidogrel (n = 43) and the half‐life of the similarly metabolized pantoprazole (n = 16) in critically ill patients. ADP‐induced aggregometry in whole blood classified 74% (95% confidence intervals 59–87%) of critically ill patients as poor responders (n = 43), and 65% (49–79%) responded poorly according to the vasodilator‐stimulated phosphoprotein phosphorylation (VASP‐P) assay. Although the plasma levels of clopidogrel active metabolite normally exceed the inactive prodrug ∼30‐fold, the parent drug levels even exceeded those of the metabolite 2‐fold in critically ill patients. The half‐life of pantoprazole was several‐fold longer in these patients compared with reference populations. The inverse ratio of prodrug/active metabolite indicates insufficient metabolization of clopidogrel, which is independently confirmed by the ∼5‐fold increase in half‐life of pantoprazole. Thus, high‐risk patients may benefit from treatment with alternative platelet inhibitors.
Collapse
Affiliation(s)
| | - Eva-Luise Hobl
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Peter Schellongowski
- Department of Medicine I, Hematology, and Oncology, Medical University of Vienna, Vienna, Austria
| | - Gottfried Heinz
- Department of Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Walter S Speidl
- Department of Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Monika Schmid
- Department of Medicine III, Gastroenterology, and Hepatology, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Stimpfl
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Schoergenhofer C, Hobl EL, Staudinger T, Speidl WS, Heinz G, Siller-Matula J, Zauner C, Reiter B, Kubica J, Jilma B. Prasugrel in critically ill patients. Thromb Haemost 2017; 117:1582-1587. [PMID: 28692105 PMCID: PMC6292180 DOI: 10.1160/th17-03-0154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022]
Abstract
While prasugrel is indicated for the treatment of myocardial infarction, its effects in the most severely affected patients requiring intensive care is unknown, so that we measured the antiplatelet effects and sparse pharmacokinetics of prasugrel in critically ill patients. Twenty-three patients admitted to medical intensive care units, who were treated with 10 mg prasugrel once daily, were included in this prospective trial. Critically ill patients responded poorly to daily prasugrel treatment: adenosine diphosphate (ADP)-induced aggregation in whole blood classified 65 % (95 % confidence intervals (CI) 43-84 %) of patients as having high on treatment platelet reactivity, platelet function under high shear rates even 74 % (95 %CI 52-90 %). There was only limited additional inhibition provided 2 hours after the next dose of prasugrel. In contrast, insufficient inhibition of the target was only seen in 26 % (95 %CI 10-48 %) of patients as measured by the vasodilator-stimulated phosphoprotein phosphorylation (VASP-P) assay. Low effective plasma levels of prasugrel active metabolite were measured at trough [0.5 (quartiles 0.5-1.1) ng/ml at baseline], and 2 hours after intake [5.7 (3.8-9.8) ng/ml], but showed coefficients of variation of ~70 %. In sum, inhibition of platelet aggregation by prasugrel is not uniform but highly variable in critically ill patients, similar to clopidogrel in a general population. The pharmacokinetic measurements indicate that poor absorption/metabolism of prasugrel may partly contribute while inflammation induced heightened intrinsic platelet reactivity may also play a role.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Bernd Jilma
- Bernd Jilma, MD, Währinger Gürtel 18-20, 1090 Vienna, Austria, Tel.: +43 1 40400 29810, Fax: +43 1 40400 29980, E-mail:
| |
Collapse
|
24
|
Schoergenhofer C, Hobl EL, Schwameis M, Gelbenegger G, Staudinger T, Heinz G, Speidl WS, Zauner C, Reiter B, Lang I, Jilma B. Acetylsalicylic acid in critically ill patients: a cross-sectional and a randomized trial. Eur J Clin Invest 2017; 47:504-512. [PMID: 28556061 PMCID: PMC5519937 DOI: 10.1111/eci.12771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/23/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Despite decades of clinical use, the pharmacokinetics and the effects of acetylsalicylic acid (ASA) in critically ill patients remain ill-defined. We aimed to investigate the pharmacokinetics and the effects of different ASA formulations during critical illness. DESIGN A cross-sectional study and a randomized, parallel-group trial were performed. Critically ill patients under chronic oral ASA treatment (100 mg enteric-coated) were screened for high 'on-treatment' platelet reactivity (HTPR) according to arachidonic acid-induced whole-blood aggregometry. Thirty patients with HTPR were randomized to receive 100 mg ASA intravenously, 100 mg enteric-coated ASA bid (bis in die) or 81 mg chewable ASA (n = 10 per group). Serum thromboxane B2 (TXB2) levels, ASA and salicylic acid levels were quantified. RESULTS Of 66 patients, 85% (95% confidence intervals 74-93%) had HTPR. Compared to baseline infusion of 100 mg, ASA significantly reduced platelet aggregation after 24 h to median 80% (Quartiles: 66-84%). Intake of 81 mg chewable ASA significantly reduced platelet aggregation to 75% (54-86%) after four hours, but increased it to 117% after 24 h (81-163%). Treatment with 100 mg enteric-coated ASA bid decreased platelet aggregation after 24 h to median 56% (52-113%). Baseline TXB2 levels were median 0·35 ng/mL (0·07-0·94). Infusion of ASA or intake of 100 mg ASA bid reduced TXB2 levels to 0·07-0·18 ng/mL after 24 h, respectively. Chewable ASA reduced TXB2 levels only transiently. Pharmacokinetic analysis revealed highly variable absorption patterns of oral ASA formulations. CONCLUSION There is a very high prevalence of HTPR in critically ill patients on peroral ASA therapy, caused by an incomplete suppression of TXB2 and/or by impaired absorption of ASA.
Collapse
Affiliation(s)
| | - Eva-Luise Hobl
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Schwameis
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Staudinger
- Department of Internal Medicine I Oncology & Hematology, Medical University of Vienna, Vienna, Austria
| | - Gottfried Heinz
- Department of Internal Medicine II Cardiology, Medical University of Vienna, Vienna, Austria
| | - Walter S Speidl
- Department of Internal Medicine II Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christian Zauner
- Department of Internal Medicine III Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Birgit Reiter
- Clinical Institute of Laboratory Medicine, Forensic Toxicology Unit, Medical University of Vienna, Vienna, Austria
| | - Irene Lang
- Department of Internal Medicine II Cardiology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Mohring A, Piayda K, Dannenberg L, Zako S, Schneider T, Bartkowski K, Levkau B, Zeus T, Kelm M, Hohlfeld T, Polzin A. Thromboxane Formation Assay to Identify High On-Treatment Platelet Reactivity to Aspirin. Pharmacology 2017; 100:127-130. [DOI: 10.1159/000477303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 04/28/2017] [Indexed: 01/29/2023]
Abstract
Platelet inhibition by aspirin is indispensable in the secondary prevention of cardiovascular events. Nevertheless, impaired aspirin antiplatelet effects (high on-treatment platelet reactivity [HTPR]) are frequent. This is associated with an enhanced risk of cardiovascular events. The current gold standard to evaluate platelet hyper-reactivity despite aspirin intake is the light-transmittance aggregometry (LTA). However, pharmacologically, the most specific test is the measurement of arachidonic acid (AA)-induced thromboxane (TX) B2 formation. Currently, the optimal cut-off to define HTPR to aspirin by inhibition of TX formation is not known. Therefore, in this pilot study, we aimed to calculate a TX formation cut-off value to detect HTPR defined by the current gold standard LTA. We measured platelet function in 2,507 samples. AA-induced TX formation by ELISA and AA-induced LTA were used to measure aspirin antiplatelet effects. TX formation correlated nonlinearly with the maximum of aggregation in the AA-induced LTA (Spearman's rho R = 0.7396; 95% CI 0.7208-0.7573, p < 0.0001). Receiver operating characteristic analysis and Youden's J statistics revealed 209.8 ng/mL as the optimal cut-off value to detect HTPR to aspirin with the TX ELISA (area under the curve: 0.92, p < 0.0001, sensitivity of 82.7%, specificity of 90.3%). In summary, TX formation ELISA is reliable in detecting HTPR to aspirin. The calculated cut-off level needs to be tested in trials with clinical end points.
Collapse
|
26
|
Su J, Yu Q, Zhu H, Li X, Cui H, Du W, Ji L, Tong M, Zheng Y, Xu H, Zhang J, Zhu Y, Xia Y, Liu T, Yao Q, Yang J, Chen X, Yu J. The risk of clopidogrel resistance is associated with ABCB1 polymorphisms but not promoter methylation in a Chinese Han population. PLoS One 2017; 12:e0174511. [PMID: 28358842 PMCID: PMC5373545 DOI: 10.1371/journal.pone.0174511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 02/28/2017] [Indexed: 11/19/2022] Open
Abstract
The goal of our study was to investigate the contribution of ABCB1 expression to the risk of clopidogrel resistance (CR). Platelets functions were measured using the Verify-Now P2Y12 assay. Applying Polymerase Chain Reaction–Restriction Fragment Length Polymorphism (PCR-RFLP), the single-nucleotide polymorphisms (SNPs) was tested. Using bisulphite pyrosequencing assay, we investigated the association of the ABCB1 DNA methylation levels and CR. It was shown that female, hypertension, and lower albumin levels increased the risk of CR (P<0.05). If patients did not have hypoproteinaemia or had hypertension, the SNP in rs1045642 was associated with CR (CC vs. TT: albumin ≥35, P = 0.042; hypertension, P = 0.045; C vs. T: albumin ≥35, P = 0.033; hypertension, P = 0.040). Additionally, the platelet inhibition of the CT+TT genotype in rs1128503 was larger than that of the CC genotype (P = 0.021). Multivariate logistic regression analysis showed that male, higher albumin and hsCRP decreased the risk of CR, and the stent size maybe positively correlated with CR. The SNP in rs1045642 was related to all-cause mortality (P = 0.024). We did not find any relationship between the methylation levels of the ABCB1 promoter and CR. In conclusions, our study indicated that ABCB1 polymorphisms might be useful in further evaluating the pathogenesis of CR.
Collapse
Affiliation(s)
- Jia Su
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Qinglin Yu
- Department of Traditional Chinese Internal Medicine, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Hao Zhu
- Department of Anaesthesia, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Xiaojing Li
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Hanbin Cui
- Department of Cardiology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Weiping Du
- Department of Cardiology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Lindan Ji
- Department of Biochemistry, School of Medicine, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Maoqing Tong
- The Key Laboratory of Molecular Medicine, Ningbo No. 1 Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yibo Zheng
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Hongyu Xu
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Jianjiang Zhang
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Yunyun Zhu
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Yezi Xia
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Ting Liu
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
| | - Qi Yao
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
- * E-mail: (QY); (JY); (XC); (JY)
| | - Jun Yang
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
- * E-mail: (QY); (JY); (XC); (JY)
| | - Xiaomin Chen
- Department of Cardiology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
- * E-mail: (QY); (JY); (XC); (JY)
| | - Jingbo Yu
- Department of Gerontology, Ningbo No.1 Hospital, Ningbo, Zhejiang Province, People's Republic of China
- * E-mail: (QY); (JY); (XC); (JY)
| |
Collapse
|
27
|
Ranucci M, Baryshnikova E. The interaction between preoperative platelet count and function and its relationship with postoperative bleeding in cardiac surgery. Platelets 2017; 28:794-798. [DOI: 10.1080/09537104.2017.1280148] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Marco Ranucci
- Department of Cardiothoracic – Vascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | - Ekaterina Baryshnikova
- Department of Cardiothoracic – Vascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
28
|
de Vries MR, Simons KH, Jukema JW, Braun J, Quax PHA. Vein graft failure: from pathophysiology to clinical outcomes. Nat Rev Cardiol 2016; 13:451-70. [PMID: 27194091 DOI: 10.1038/nrcardio.2016.76] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Occlusive arterial disease is a leading cause of morbidity and mortality worldwide. Aside from balloon angioplasty, bypass graft surgery is the most commonly performed revascularization technique for occlusive arterial disease. Coronary artery bypass graft surgery is performed in patients with left main coronary artery disease and three-vessel coronary disease, whereas peripheral artery bypass graft surgery is used to treat patients with late-stage peripheral artery occlusive disease. The great saphenous veins are commonly used conduits for surgical revascularization; however, they are associated with a high failure rate. Therefore, preservation of vein graft patency is essential for long-term surgical success. With the exception of 'no-touch' techniques and lipid-lowering and antiplatelet (aspirin) therapy, no intervention has hitherto unequivocally proven to be clinically effective in preventing vein graft failure. In this Review, we describe both preclinical and clinical studies evaluating the pathophysiology underlying vein graft failure, and the latest therapeutic options to improve patency for both coronary and peripheral grafts.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Karin H Simons
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jerry Braun
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| |
Collapse
|