1
|
Dillinger JG, Pezel T, Batias L, Angoulvant D, Goralski M, Ferrari E, Cayla G, Silvain J, Gilard M, Lemesle G, Souteyrand G, Lim P, Roubille F, Georges JL, Bal Dit Sollier C, Petroni T, Morel O, Delarche N, Elbaz M, Puymirat E, Toupin S, Montalescot G, Drouet L, Vicaut E, Henry P. Twice-a-day administration of aspirin in patients with diabetes mellitus or aspirin resistance after acute coronary syndrome: Rationale and design of the randomized ANDAMAN trial. Am Heart J 2025:S0002-8703(25)00132-2. [PMID: 40268181 DOI: 10.1016/j.ahj.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Patients with diabetes mellitus (DM) or aspirin resistance are exposed to recurrent atherothrombotic events after acute coronary syndrome (ACS). Aspirin once-daily can allow the recovery of platelet cyclooxygenase activity before the next intake in these patients. Twice-daily administration provides more stable inhibition of platelet aggregation and may improve prognosis in these patients. AIM To demonstrate the superiority of twice-daily aspirin compared to once daily in reducing major adverse cardiovascular events (MACE) in patients with DM or aspirin resistance after ACS. METHODS The ANDAMAN trial is a randomized, multicenter study including patients (aged ≥18 years) with DM or with aspirin resistance defined as: i) index event occurring under aspirin; ii) body mass index ≥ 27 kg/m2); iii) increased waist circumference (≥ 88 cm for women or ≥ 102 cm for men). The patients will be recruited in 39 centers after an ACS (with or without ST elevation) with at least one significant coronary stenosis and will be randomized before hospital discharge between twice-daily versus once daily low-dose aspirin (100 mg bid versus od). The primary composite endpoint will be the occurrence of MACE including all-cause death, myocardial infarction, stroke, urgent coronary revascularization or acute arterial thrombotic event during a follow-up of 18 months. To achieve a 20% reduction in the relative risk of MACE in the twice-daily aspirin group, a total of 2,574 patients will be included in the trial. The main secondary endpoint will be major bleeding (type 3-5 following BARC classification). CONCLUSIONS The trial will evaluate the prognostic impact of twice-daily aspirin for ACS patients with DM or aspirin resistance and may change the way aspirin is administered to these patients. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02520921.
Collapse
Affiliation(s)
- Jean-Guillaume Dillinger
- Université Paris Cité, Department of Cardiology, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Inserm MASCOT - UMRS 942, Paris, France.; MIRACL.ai laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisiere (AP-HP), 75010, Paris, France.; CREATIF, Centre de Référence et d'Education des antithrombotiques d'Ile de France, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France..
| | - Théo Pezel
- Université Paris Cité, Department of Cardiology, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Inserm MASCOT - UMRS 942, Paris, France.; MIRACL.ai laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisiere (AP-HP), 75010, Paris, France
| | - Laure Batias
- Centre Hospitalier Métropole Savoie, Service de Cardiologie, Place Lucien Biset, 73000 Chambéry, France
| | - Denis Angoulvant
- Cardiology Department, CHU Tours, INSERM Unité 1327 ISCHEMIA, Université de Tours, Tours, France
| | | | - Emile Ferrari
- Cardiology Department, Pasteur University Hospital, 30 avenue de la Voie Romaine, CS 51069, 06001, Nice CEDEX 1, France
| | - Guillaume Cayla
- Cardiology department, Nimes university Hospital, Montpellier University, ACTION group, Nimes, France
| | - Johanne Silvain
- Sorbonne Université, ACTION Group, INSERM UMRS1166, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Martine Gilard
- Chest Diseases, CHU_Brest, INSERM U1304, Univ_Brest, Brest, France
| | - Gilles Lemesle
- Heart and Lung Institute, University hospital of Lille, CHU Lille, F-59000 Lille, France. Univ. Lille, F-59000, France. Institut Pasteur of Lille, Inserm U1011-EGID, F-59000 Lille, France. FACT (French Alliance for Cardiovascular Trials), F-75000 Paris, France
| | - Géraud Souteyrand
- Institut Pascal, Thérapies Guidées par l'Image, CNRS SIGMA UCA UMR 6602 University Hospital Gabriel Montpied, Clermont-Ferrand, France
| | - Pascal Lim
- Service de Cardiologie, Univ Paris Est Créteil, INSERM, IMRB, AP-HP, Hôpital Universitaire Henri-Mondor, F-94010, Créteil, France
| | - François Roubille
- PhyMedExp, Cardiology Department, Université de Montpellier, INSERM, CNRS, INI-CRT, CHU de Montpellier, France
| | - Jean-Louis Georges
- Service de cardiologie, cardiologie interventionnelle, Centre Hospitalier de Versailles, Hôpital André Mignot, Le Chesnay-Rocquencourt, France
| | - Claire Bal Dit Sollier
- CREATIF, Centre de Référence et d'Education des antithrombotiques d'Ile de France, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thibaut Petroni
- Cardiology Department, Clinique Pont de Chaume, Montauban, France
| | - Olivier Morel
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, UR 3074 Translational CardioVascular Medicine CRBS, Strasbourg, France
| | | | - Meier Elbaz
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse 31400, France
| | - Etienne Puymirat
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Department of Cardiology, 75015 Paris, France; Université de Paris Cité, 75006 Paris, France
| | - Solenn Toupin
- Université Paris Cité, Department of Cardiology, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Inserm MASCOT - UMRS 942, Paris, France.; MIRACL.ai laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisiere (AP-HP), 75010, Paris, France
| | - Gilles Montalescot
- Sorbonne Université, ACTION Group, INSERM UMRS1166, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France
| | - Ludovic Drouet
- CREATIF, Centre de Référence et d'Education des antithrombotiques d'Ile de France, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric Vicaut
- Unité de Recherche Clinique, ACTION Group, Hôpital Fernand Widal (AP-HP), Paris, France
| | - Patrick Henry
- Université Paris Cité, Department of Cardiology, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Inserm MASCOT - UMRS 942, Paris, France
| |
Collapse
|
2
|
Kallmes DF, Altschul D. Personalized Platelet Protocols: Progress, Pitfalls, and Potential. Radiology 2025; 314:e250488. [PMID: 40100024 DOI: 10.1148/radiol.250488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Affiliation(s)
- David F Kallmes
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | | |
Collapse
|
3
|
Erdoes G, Milojevic M, Rocca B, Koster A. Clinical guidelines for a dry land: challenges in developing recommendations for DOAC management in emergent cardiac surgery. Eur J Cardiothorac Surg 2025; 67:ezaf015. [PMID: 39873698 DOI: 10.1093/ejcts/ezaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Affiliation(s)
- Gabor Erdoes
- Department of Anesthesiology and Pain Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Milan Milojevic
- Department of Cardiac Surgery and Cardiovascular Research, Dedinje Cardiovascular Institute, Belgrade, Serbia
| | - Bianca Rocca
- Department of Medicine and Surgery, LUM University, Casamassima, Bari, Italy
| | - Andreas Koster
- Clinics of Anesthesiology and Intensive Care Medicine, Sana Heart Center Cottbus, Cottbus, Germany
- Institute of Anesthesiology and Pain Therapy, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
4
|
Kennedy NN, Xia Y, Barrett T, Luttrell-Williams E, Berland T, Cayne N, Garg K, Jacobowitz G, Lamparello PJ, Maldonado TS, Newman J, Sadek M, Smilowitz NR, Rockman C, Berger JS. Dynamic perioperative platelet activity and cardiovascular events in peripheral artery disease. J Vasc Surg 2025; 81:432-440.e3. [PMID: 39362415 DOI: 10.1016/j.jvs.2024.09.028] [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: 05/03/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
OBJECTIVE Patients with peripheral artery disease (PAD) undergo lower extremity revascularization (LER) for symptomatic relief or limb salvage. Despite LER, patients remain at increased risk of platelet-mediated complications, such as major adverse cardiac and limb events (MACLEs). Platelet activity is associated with cardiovascular events, yet little is known about the dynamic nature of platelet activity over time. We, therefore, investigated the change in platelet activity over time and its association with long-term cardiovascular risk. METHODS Patients with PAD undergoing LER were enrolled into the multicenter, prospective Platelet Activity and Cardiovascular Events study. Platelet aggregation was assessed by light transmission aggregometry to submaximal epinephrine (0.4 μmol/L) immediately before LER, and on postoperative day 1 or 2 (POD1 or POD2) and 30 (POD30). A hyperreactive platelet phenotype was defined as >60% aggregation. Patients were followed longitudinally for MACLEs, defined as the composite of death, myocardial infarction, stroke, major lower extremity amputation, or acute limb ischemia leading to reintervention. RESULTS Among 287 patients undergoing LER, the mean age was 70 ± 11 years, 33% were female, 61% were White, and 89% were on baseline antiplatelet therapy. Platelet aggregation to submaximal epinephrine induced a bimodal response; 15.5%, 16.8%, and 16.4% of patients demonstrated a hyperreactive platelet phenotype at baseline, POD1, and POD30, respectively. Platelet aggregation increased by 18.5% (P = .001) from baseline to POD1, which subsequently returned to baseline at POD30. After a median follow-up of 19 months, MACLEs occurred in 165 patients (57%). After adjustment for demographics, clinical risk factors, procedure type, and antiplatelet therapy, platelet hyperreactivity at POD1 was associated with a significant hazard of long-term MACLE (adjusted hazard ratio, 4.61; 95% confidence interval, 2.08-10.20; P < .001). CONCLUSIONS Among patients with severe PAD, platelet activity increases after LER. Platelet hyperreactivity to submaximal epinephrine on POD1 is associated with long-term MACLE. Platelet activity after LER may represent a modifiable biomarker associated with excess cardiovascular risk.
Collapse
Affiliation(s)
- Natalie N Kennedy
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Yuhe Xia
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Tessa Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Elliot Luttrell-Williams
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Todd Berland
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Neal Cayne
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Karan Garg
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Glenn Jacobowitz
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Patrick J Lamparello
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Thomas S Maldonado
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Jonathan Newman
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Mikel Sadek
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Nathaniel R Smilowitz
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY; Cardiology Section, Department of Medicine, VA New York Harbor Healthcare System, New York, NY
| | - Caron Rockman
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Jeffrey S Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY; Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY.
| |
Collapse
|
5
|
Ingraham BS, Valgimigli M, Angiolillo DJ, Capodanno D, Rao SV, Urban P, Singh M. Relevance of High Bleeding Risk and Postdischarge Bleeding in Patients Undergoing Percutaneous Coronary Intervention. Mayo Clin Proc 2025; 100:304-331. [PMID: 39909670 DOI: 10.1016/j.mayocp.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 02/07/2025]
Abstract
Bleeding avoidance strategies are critical in the modern era of percutaneous coronary intervention; however, most efforts are geared toward reducing access-related complications. Improvements in procedural techniques (radial access, improved procedural anticoagulation regimens, etc) and modifications in postdischarge pharmacotherapy (shortened dual antiplatelet therapy, genotype-guided P2Y12 inhibition, etc) that led to a decline in bleeding related to percutaneous procedures were largely offset by increases in complexity and performance of percutaneous coronary intervention in high-risk patients. Among patients presenting with acute coronary syndrome, aggressive antiplatelet regimens with potent P2Y12 inhibitors are typically prescribed for a longer duration, prioritizing reduction in ischemic events over bleeding risk. Because postdischarge bleeding connotes an adverse prognosis similar to an ischemic event, postprocedure freedom from adverse outcomes can be best tailored by individualizing and recognizing the patient's bleeding and ischemic risks. This review of the contemporary and historical literature (PubMed, EMBASE, Cochrane Library) summarizes the available data, provides strategies to navigate these complex decisions, and helps individualize antithrombotic therapy.
Collapse
Affiliation(s)
| | - Marco Valgimigli
- Cardiocentro Ticino Institute and Università della Svizzera italiana, Lugano, Switzerland
| | | | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Catania, Italy
| | - Sunil V Rao
- Division of Cardiology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY
| | | | - Mandeep Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
6
|
Tunehag KR, Pearce AF, Fox LP, Stouffer GA, Solander S, Lee CR. CYP2C19 Genotype-Guided Antiplatelet Therapy and Clinical Outcomes in Patients Undergoing a Neurointerventional Procedure. Clin Transl Sci 2025; 18:e70131. [PMID: 39822142 PMCID: PMC11739457 DOI: 10.1111/cts.70131] [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: 10/29/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 01/19/2025] Open
Abstract
In neurovascular settings, including treatment and prevention of ischemic stroke and prevention of thromboembolic complications after percutaneous neurointerventional procedures, dual antiplatelet therapy with a P2Y12 inhibitor and aspirin is the standard of care. Clopidogrel remains the most commonly prescribed P2Y12 inhibitor for neurovascular indications. However, patients carrying CYP2C19 no-function alleles have diminished capacity for inhibition of platelet reactivity due to reduced formation of clopidogrel's active metabolite. In patients with cardiovascular disease undergoing a percutaneous coronary intervention, CYP2C19 no-function allele carriers treated with clopidogrel experience a higher risk of major adverse cardiovascular outcomes, and multiple large prospective outcomes studies have shown an improvement in clinical outcomes when antiplatelet therapy selection was guided by CYP2C19 genotype. Similarly, accumulating evidence has associated CYP2C19 no-function alleles with poor clinical outcomes in clopidogrel-treated patients in neurovascular settings. However, the utility of implementing a genotype-guided antiplatelet therapy selection strategy in the setting of neurovascular disease and the clinical outcomes evidence in neurointerventional procedures remains unclear. In this review, we will (1) summarize existing evidence and guideline recommendations related to CYP2C19 genotype-guided antiplatelet therapy in the setting of neurovascular disease, (2) evaluate and synthesize the existing evidence on the relationship of clinical outcomes to CYP2C19 genotype and clopidogrel treatment in patients undergoing a percutaneous neurointerventional procedure, and (3) identify knowledge gaps and discuss future research directions.
Collapse
Affiliation(s)
- Kayla R. Tunehag
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Ashton F. Pearce
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Layna P. Fox
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - George A. Stouffer
- Division of Cardiology, Department of Medicine, UNC School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- UNC McAllister Heart InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Sten Solander
- Department of Radiology, UNC School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Division of Cardiology, Department of Medicine, UNC School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- UNC McAllister Heart InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
7
|
Pedersen OB, Nissen PH, Pasalic L, Hvas AM, Kristensen SD, Grove EL. Changes in platelet maturity and reactivity following acute ST-segment elevation myocardial infarction. Res Pract Thromb Haemost 2025; 9:102652. [PMID: 39845649 PMCID: PMC11751529 DOI: 10.1016/j.rpth.2024.102652] [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: 09/13/2024] [Revised: 10/31/2024] [Accepted: 12/03/2024] [Indexed: 01/24/2025] Open
Abstract
Background Reduced effect of antiplatelet therapy has been reported in patients with ST-segment elevation myocardial infarction (STEMI). This could partly be explained by an increase of highly reactive immature platelets. Objectives To investigate changes in platelet maturity and reactivity after acute STEMI. Methods Patients diagnosed with STEMI, admitted for primary percutaneous coronary intervention, and treated according to international guidelines, were included. Blood samples were obtained within 24 hours after admission and at 2- to 3-months follow-up. Platelet maturity and reactivity using multicolor flow cytometry with SYTO-13 to categorize platelet maturity, whole blood platelet aggregation, serum thromboxane B2 levels, and standard immature platelet markers (eg, immature platelet count and fraction, and mean platelet volume) were measured. Results A total of 44 STEMI patients were included. The reactivity of immature platelets was consistently higher at baseline and at follow-up when compared to the entire platelet population and the mature platelet population (all P values < .05). The expression of CD63 (a dense granule marker) in immature platelets was consistently high compared to the entire platelet population and the mature platelet population and did not change from baseline to follow-up (P values > .24). Additionally, a positive significant correlation was found between standard immature platelet markers and the expression of CD63 on platelets both at baseline and follow-up (rho ranging from 0.32 to 0.62, all P values < .05). Conclusion Immature platelets represent a highly reactive platelet subpopulation crucial for the overall platelet reactivity, partly due to a high expression of dense granules. Despite treatment with loading and maintenance doses of antiplatelet therapy, the reactivity of immature platelets remained high in STEMI patients.
Collapse
Affiliation(s)
- Oliver Buchhave Pedersen
- Thrombosis and Haemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Peter H. Nissen
- Thrombosis and Haemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Leonardo Pasalic
- Institute of Clinical Pathology and Medical Research, Westmead Hospital, New South Wales Health Pathology, Sydney, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | | | - Steen Dalby Kristensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Erik Lerkevang Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
8
|
Qamar U, Naeem F, Maqsood MT, Khan MZ, Imtiaz Z, Saeed F, Gupta N, Brohi FZ, Mkpozi C, Sattar Y. Efficacy and safety of ticagrelor monotherapy following a brief DAPT vs. prolonged 12-month DAPT in ACS patients post-PCI: a meta-analysis of RCTs. Eur J Clin Pharmacol 2024; 80:1871-1882. [PMID: 39264445 DOI: 10.1007/s00228-024-03747-w] [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: 05/19/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND As per current guidelines, acute coronary syndrome (ACS) patients who undergo percutaneous coronary intervention (PCI) should be started on dual antiplatelet therapy (DAPT) for a period of 12 months. OBJECTIVE To assess the efficacy and safety of brief DAPT (up to 3 months) succeeded by ticagrelor monotherapy compared with a 12-month DAPT in ACS patients following PCI. METHODS We systematically searched Cochrane, Embase, and PubMed to find relevant randomized clinical trials. Examined outcomes included the incidence of major adverse cerebrovascular and cardiovascular events (MACCE), bleeding events, and the composite incidence of net adverse clinical events (NACE). RESULTS Our primary analysis included 21,927 ACS patients from six RCTs. Our pooled results indicate that following PCI in individuals with ACS, brief DAPT followed by ticagrelor did not increase the risk of MACCE (OR 0.92, 95% CI 0.79-1.07) but significantly reduced the risk of minor or major bleeding (OR 0.52, 95% CI 0.44-0.62) and NACE (OR 0.71, 95% CI 0.59-0.86) compared with a long-term DAPT within a follow-up of 12 months. CONCLUSION Brief DAPT followed by ticagrelor monotherapy is superior to a 12-month DAPT in offering a net clinical advantage in ACS patients following PCI.
Collapse
Affiliation(s)
- Usama Qamar
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Farhan Naeem
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Maleeka Zamurad Khan
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Zeeshan Imtiaz
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Fatima Saeed
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Neelesh Gupta
- Department of Cardiology, Kirk Kerkorian School of Medicine at the, University of Nevada, Las Vegas, USA
| | | | - Celestine Mkpozi
- Department of Internal Medicine and Department of Cardiology, West Virginia University, 1 Medical Ctr Dr., Morgantown, WV, 26506, USA
| | - Yasar Sattar
- Department of Internal Medicine and Department of Cardiology, West Virginia University, 1 Medical Ctr Dr., Morgantown, WV, 26506, USA.
| |
Collapse
|
9
|
Angiolillo DJ, Galli M, Alexopoulos D, Aradi D, Bhatt DL, Bonello L, Capodanno D, Cavallari LH, Collet JP, Cuisset T, Ferreiro JL, Franchi F, Geisler T, Gibson CM, Gorog DA, Gurbel PA, Jeong YH, Marcucci R, Siller-Matula JM, Mehran R, Neumann FJ, Pereira NL, Rizas KD, Rollini F, So DYF, Stone GW, Storey RF, Tantry US, Berg JT, Trenk D, Valgimigli M, Waksman R, Sibbing D. International Consensus Statement on Platelet Function and Genetic Testing in Percutaneous Coronary Intervention: 2024 Update. JACC Cardiovasc Interv 2024; 17:2639-2663. [PMID: 39603778 DOI: 10.1016/j.jcin.2024.08.027] [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: 07/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 11/29/2024]
Abstract
Current evidence indicates that dual antiplatelet therapy with aspirin plus a P2Y12 inhibitor is essential for the prevention of thrombotic events after percutaneous coronary interventions. However, dual antiplatelet therapy is associated with increased bleeding which may outweigh the benefits. This has set the foundations for customizing antiplatelet treatments to the individual patient. However, bleeding and ischemic risks are often present in the same patient, making it difficult to achieve this balance. The fact that oral P2Y12 inhibitors (clopidogrel, prasugrel, and ticagrelor) have diverse pharmacodynamic profiles that affect clinical outcomes supports the rationale for using platelet function and genetic testing to individualize antiplatelet treatment regimens. Indeed, up to one-third of patients treated with clopidogrel, but a minority of those treated with prasugrel or ticagrelor, exhibit high residual platelet reactivity resulting in an increased thrombotic risk. On the other hand, prasugrel and ticagrelor are frequently associated with low platelet reactivity and increased bleeding risk compared with clopidogrel without providing any additional reduction in ischemic events compared with patients who adequately respond to clopidogrel. The use of platelet function and genetic testing may allow for a guided selection of oral P2Y12 inhibitors. However, the nonuniform results of randomized controlled trials have led guidelines to provide limited recommendations on the implementation of these tests in patients undergoing percutaneous coronary intervention. In light of recent advancements in the field, this consensus document by a panel of international experts fills in the guideline gap by providing updates on the latest evidence in the field as well as recommendations for clinical practice.
Collapse
Affiliation(s)
- Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA.
| | - Mattia Galli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Dimitrios Alexopoulos
- 7th Department of Cardiology, Hygeia Hospital, Athens, Greece; State Hospital for Cardiology, Balatonfüred, Hungary
| | - Daniel Aradi
- State Hospital for Cardiology, Balatonfüred, Hungary; Hungary and Heart and Vascular Centre, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laurent Bonello
- Intensive Care Unit, Hopital Universitaire Nord, Aix-Marseille University, Marseille, France
| | - Davide Capodanno
- Azienda Ospedaliero-Universitaria Policlinico G. Rodolico-San Marco, University of Catania, Catania, Italy
| | - Larisa H Cavallari
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Jean-Philippe Collet
- ACTION Study Group, INSERM UMRS 1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Thomas Cuisset
- Department of Cardiology, La Timone Hospital, Marseille, France
| | - Jose Luis Ferreiro
- Department of Cardiology, Joan XXIII University Hospital, IISPV, Rovira i Virgili University, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Tarragona, Spain
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - C Michael Gibson
- Baim Institute of Clinical Research, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Diana A Gorog
- Cardiovascular Division, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Centre for Health Services and Clinical Research, Postgraduate Medical School, University of Hertfordshire, Hertfordshire, United Kingdom
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | - Young-Hoon Jeong
- CAU Thrombosis and Biomarker Center, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, South Korea; Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jolanta M Siller-Matula
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, Icahn School Medicine at Mount Sinai, New York, New York, USA
| | - Franz-Josef Neumann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Konstantinos D Rizas
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Derek Y F So
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Gregg W Stone
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Robert F Storey
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | - Jurrien Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands; Department of Cardiology, University Medical Center Maastricht, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Dietmar Trenk
- Clinical Pharmacology, Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland; University of Bern, Bern, Switzerland
| | - Ron Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Dirk Sibbing
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; Privatklinik Lauterbacher Mühle am Ostsee, Seeshaupt, Germany
| |
Collapse
|
10
|
Elserwey A, Jabbour RJ, Curzen N. Does one size really fit all? The case for personalized antiplatelet therapy in interventional cardiology. Future Cardiol 2024; 20:499-515. [PMID: 39093436 PMCID: PMC11485715 DOI: 10.1080/14796678.2024.2384217] [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: 03/06/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. Dual antiplatelet therapy (DAPT), with aspirin plus a P2Y12 inhibitor, is currently recommended as a default for patients after acute coronary syndrome (ACS) and following percutaneous coronary intervention (PCI). However, controversies arise over the role of aspirin, the optimal duration of DAPT after drug-eluting stent (DES) implantation, the choice of P2Y12 inhibitor and the variability in individual responses to antiplatelet agents. Recent data indicate that monotherapy with a P2Y12 inhibitor may have adequate anti-ischemic effects with lower bleeding risk. Additionally, discrepancies in DAPT duration recommendations and the optimal P2Y12 inhibitor, provides more uncertainty. We ask the question "does one size really fits all?" or should a more personalized strategy should be implemented.
Collapse
Affiliation(s)
- Ahmed Elserwey
- Faculty of Medicine, University of Southampton
- University Hospital Southampton NHS FT
| | | | - Nick Curzen
- Faculty of Medicine, University of Southampton
- University Hospital Southampton NHS FT
| |
Collapse
|
11
|
Masi P, Gendreau S, Moyon Q, Leguyader M, Lebreton G, Ropers J, Dangers L, Sitruk S, Bréchot N, Pineton de Chambrun M, Chommeloux J, Schmidt M, Luyt CE, Leprince P, Combes A, Frere C, Hékimian G. Bleeding complications, coagulation disorders, and their management in acute myocardial infarction-related cardiogenic shock rescued by veno-arterial ECMO: A retrospective cohort study. J Crit Care 2024; 82:154771. [PMID: 38471248 DOI: 10.1016/j.jcrc.2024.154771] [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: 07/19/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE Management of dual antiplatelet therapy (DAPT) in patients on venoarterial-extracorporeal membrane (VA-ECMO) after acute myocardial infarction (AMI) is challenging. Our objective was to describe the frequency, management and outcomes of severe bleeding complications and determine their occurrence risk factors. MATERIAL AND METHODS We conducted a retrospective observational cohort study including post-AMI cardiogenic shock patients requiring VA-ECMO. Severe bleeding was defined based on the Bleeding Academic Research Consortium classification. We calculated multivariable Fine-Gray models to assess factors associated with risk of severe bleeding. RESULTS From January 2015 to July 2019, 176 patients received VA-ECMO after AMI and 132 patients were included. Sixty-five (49%) patients died. Severe bleeding occurred in 39% of cases. Severe thrombocytopenia (< 50 G/L) and hypofibrinogenemia (<1,5 g/L) occurred in respectively 31% and 19% of patients. DAPT was stopped in 32% of patients with a 6% rate of stent thrombosis. Anticoagulation was stopped in 39% of patients. Using a multivariate competing risk model, female sex, time on ECMO, troponin at admission and Impella® implantation were independently associated with severe bleeding. CONCLUSIONS Bleeding complications and coagulation disorders were frequent and severe in patients on VA-ECMO after AMI, leading of antiplatelet therapy withdrawal in one third of patients.
Collapse
Affiliation(s)
- Paul Masi
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010 Créteil, France.
| | - Ségolène Gendreau
- AP-HP, Hôpitaux Universitaires Henri-Mondor, Service de Médecine Intensive Réanimation, F-94010 Créteil, France
| | - Quentin Moyon
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Maxence Leguyader
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France
| | - Guillaume Lebreton
- Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France; Cardiothoracic surgery department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Jacques Ropers
- Assistance Publique Hôpitaux de Paris, Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière -Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Paris, France
| | - Laurence Dangers
- Réanimation Polyvalente, Centre Hospitalier Universitaire Félix Guyon, Saint-Denis, France
| | - Samuel Sitruk
- Assistance Publique Hôpitaux de Paris, Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière -Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Paris, France
| | - Nicolas Bréchot
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Marc Pineton de Chambrun
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Juliette Chommeloux
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Matthieu Schmidt
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Charles Edouard Luyt
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Pascal Leprince
- Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France; Cardiothoracic surgery department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Alain Combes
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| | - Corinne Frere
- Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France; Department of Hematology, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Guillaume Hékimian
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive Réanimation, Paris, France; Sorbonne Université, Inserm, UMRS_1166-ICAN, Institut de Cardiométabolisme et Nutrition (ICAN), Paris, France
| |
Collapse
|
12
|
Romagnoli E, Aurigemma C, Biondi-Zoccai G, Burzotta F. John Steinbeck in cardiology: of guidelines and giants. Minerva Cardiol Angiol 2024; 72:311-312. [PMID: 38934265 DOI: 10.23736/s2724-5683.24.06635-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
| | | | - Giuseppe Biondi-Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy -
- Unit of Cardiology, Santa Maria Goretti Hospital, Latina, Italy
| | | |
Collapse
|
13
|
Sabouret P, Barbato E, Montalescot G, Biondi-Zoccai G, Manzo-Silberman S. Remembering our friend Jean-Philippe Collet: a giant in cardiology, a gentleman in life. Minerva Cardiol Angiol 2024; 72:329-331. [PMID: 38535986 DOI: 10.23736/s2724-5683.24.06559-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Pierre Sabouret
- Heart Institute and Action Group, Pitié-Salpétrière Hospital, Sorbonne University, Paris, France
| | - Emanuele Barbato
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Gilles Montalescot
- ACTION Group, Sorbonne University, Institute of Cardiology (AP-HP), Pitié-Salpêtrière Hospital, Paris, France
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy -
- Mediterranea Cardiocentro, Naples, Italy
| | - Stéphane Manzo-Silberman
- Heart Institute and Action Group, Pitié-Salpétrière Hospital, Sorbonne University, Paris, France
| |
Collapse
|
14
|
Kwon O, Ahn JH, Koh JS, Park Y, Hwang SJ, Tantry US, Gurbel PA, Hwang JY, Jeong YH. Platelet-fibrin clot strength and platelet reactivity predicting cardiovascular events after percutaneous coronary interventions. Eur Heart J 2024; 45:2217-2231. [PMID: 38804262 DOI: 10.1093/eurheartj/ehae296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/26/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND AND AIMS Platelet-fibrin clot strength (PFCS) is linked to major adverse cardiovascular event (MACE) risk. However, the association between PFCS and platelet reactivity and their prognostic implication remains uncertain in patients undergoing percutaneous coronary intervention (PCI). METHODS In PCI-treated patients (n = 2512) from registry data from January 2010 to November 2018 in South Korea, PFCS using thromboelastography and platelet reactivity using VerifyNow were measured. High PFCS (PFCSHigh) was defined as thromboelastography maximal amplitude ≥ 68 mm, and high platelet reactivity (HPR) was defined as >208 P2Y12 reaction units. Patients were stratified into four groups according to maximal amplitude and P2Y12 reaction unit levels: (i) normal platelet reactivity (NPR)-PFCSNormal (31.8%), (ii) HPR-PFCSNormal (29.0%), (iii) NPR-PFCSHigh (18.1%), and (iv) HPR-PFCSHigh (21.1%). Major adverse cardiovascular event (all-cause death, myocardial infarction, or stroke) and major bleeding were followed up to 4 years. RESULTS High platelet reactivity and PFCSHigh showed an additive effect for clinical outcomes (log-rank test, P < .001). Individuals with NPR-PFCSNormal, NPR-PFCSHigh, HPR-PFCSNormal, and HPR-PFCSHigh demonstrated MACE incidences of 7.5%, 12.6%, 13.4%, and 19.3%, respectively. The HPR-PFCSHigh group showed significantly higher risks of MACE compared with the NPR-PFCSNormal group [adjusted hazard ratio (HRadj) 1.89; 95% confidence interval (CI) 1.23-2.91; P = .004] and the HPR-PFCSNormal group (HRadj 1.60; 95% CI 1.12-2.27; P = .009). Similar results were observed for all-cause death. Compared with HPR-PFCSNormal phenotype, NPR-PFCSNormal phenotype was associated with a higher risk of major bleeding (HRadj 3.12; 95% CI 1.30-7.69; P = .010). CONCLUSIONS In PCI patients, PFCS and platelet reactivity demonstrated important relationships in predicting clinical prognosis. Their combined assessment may enhance post-PCI risk stratification for personalized antithrombotic therapy.
Collapse
Affiliation(s)
- Osung Kwon
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong-Hwa Ahn
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Jin-Sin Koh
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, 79, Gangnam-ro, Jinju 52727, Republic of Korea
| | - Yongwhi Park
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Seok Jae Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, 79, Gangnam-ro, Jinju 52727, Republic of Korea
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, 79, Gangnam-ro, Jinju 52727, Republic of Korea
| | - Young-Hoon Jeong
- CAU Thrombosis and Biomarker Center, Chung-Ang University Gwangmyeong Hospital, 110, Deokan-ro, Gwangmyeong 14353, Republic of Korea
- Department of Internal Medicine, Chung-Ang University College of Medicine, 84, Heukseok-ro, Seoul 06974, Republic of Korea
| |
Collapse
|
15
|
Rajachandran M, Lange RA. Role of Cyp2c19 Genotype-Guided Antiplatelet Therapy After Percutaneous Coronary Intervention. Curr Cardiol Rep 2024; 26:675-680. [PMID: 38806977 DOI: 10.1007/s11886-024-02071-0] [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] [Accepted: 05/07/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW Identification of a reliable discriminatory test to accurately stratify patient responses to antiplatelet therapy following coronary revascularization has become increasingly desirable to optimize therapeutic efficacy and safety. RECENT FINDINGS The expansion of platelet function testing to include genotype assessment has been an evolutionary journey, initially fraught with confounding results. However, more recent and rigorous data analysis suggests that genotype testing- guided, tailored antiplatelet therapy may hold promise in optimizing treatment of patients after coronary intervention. Current evidence increasingly supports the use of genotype guided CYP2C19 testing to better match the post coronary intervention patient with the most efficacious and least risky antiplatelet inhibitor. The risk stratification of poor, intermediate, and good metabolizers of these drugs with such testing promises to yield clinical dividends in terms of morbidity, mortality and cost control, in this growing patient population.
Collapse
Affiliation(s)
- Manu Rajachandran
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, 4800 Alberta Avenue, El Paso, TX, 79905, USA.
| | - Richard A Lange
- Division of Cardiology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, 4800 Alberta Avenue, El Paso, TX, 79905, USA
| |
Collapse
|
16
|
Wegerif ECJ, Ünlü Ç, Generaal MI, van den Bor RM, van de Ven PM, Bots ML, de Borst GJ. Rationale and design for the randomized placebo-controlled double-blind trial studying the effect of single antiplatelet therapy (clopidogrel) versus dual antiplatelet therapy (clopidogrel/acetylsalicylic acid) on the occurrence of atherothrombotic events following lower extremity peripheral transluminal angioplasty (CLEAR-PATH). Am Heart J 2024; 273:121-129. [PMID: 38608997 DOI: 10.1016/j.ahj.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
RATIONALE Antiplatelet therapy (APT) is the standard of care after endovascular revascularization (EVR) in patients with peripheral artery disease (PAD). APT aims to prevent both major adverse cardiovascular events (MACE) and major adverse limb events (MALE). Nonetheless, the rates of MACE and MALE after EVR remain high. In coronary artery and cerebrovascular disease, dual APT (DAPT)compared to acetylsalicylic acid alone has been proven to reduce MACE without increasing the risk of major bleeding when applied for a restricted number of weeks. However, within the PAD population, insufficient data are available to understand the potential attributable effect of DAPT over single APT (SAPT). Therefore, prospective randomized studies in targeted study populations are warranted. TRIAL DESIGN CLEAR-PATH is a Dutch multicenter, double-blind, placebo-controlled, randomized trial comparing SAPT (clopidogrel 75 mg plus placebo) with DAPT (clopidogrel 75 mg plus acetylsalicylic acid 80 mg) in patients with PAD undergoing EVR. CLEAR-PATH includes a time-to-event analysis with a follow-up of one year. The primary composite efficacy endpoint consists of all-cause mortality, nonfatal stroke, nonfatal myocardial infarction, severe limb ischemia, (indication for) re-intervention due to any symptomatic restenosis, re-occlusion, or due to acute limb ischemia, and major amputation. The primary safety endpoint contains major bleeding following the Thrombolysis in Myocardial Infarction classification. The enrolment started in August 2022. In total 450 primary efficacy outcome events are required which expectedly amounts to 1696 subjects. Recruitment will take approximately 36 months. CONCLUSION CLEAR-PATH will assess the efficacy and safety of DAPT compared to SAPT following EVR in PAD patients. TRIAL REGISTRATION NUMBER NL80009.041.21.
Collapse
Affiliation(s)
- Emilien C J Wegerif
- Division of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Çağdaş Ünlü
- Division of Vascular Surgery, Northwest Hospital Group, Alkmaar, The Netherlands
| | - Manon I Generaal
- Division of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rutger M van den Bor
- Department of Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Peter M van de Ven
- Department of Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gert J de Borst
- Division of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Montone RA, Ford TJ, Galli M, Rinaldi R, Bland A, Morrow A, Angiolillo DJ, Berry C, Kaski JC, Crea F. Stratified medicine for acute and chronic coronary syndromes: A patient-tailored approach. Prog Cardiovasc Dis 2024; 85:2-13. [PMID: 38936756 DOI: 10.1016/j.pcad.2024.06.003] [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: 06/23/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
The traditional approach to management of cardiovascular disease relies on grouping clinical presentations with common signs and symptoms into pre-specified disease pathways, all uniformly treated according to evidence-based guidelines ("one-size-fits-all"). The goal of precision medicine is to provide the right treatment to the right patients at the right time, combining data from time honoured sources (e.g., history, physical examination, imaging, laboratory) and those provided by multi-omics technologies. In patients with ischemic heart disease, biomarkers and intravascular assessment can be used to identify endotypes with different pathophysiology who may benefit from distinct treatments. This review discusses strategies for the application of stratified management to patients with acute and chronic coronary syndromes.
Collapse
Affiliation(s)
- Rocco A Montone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Thomas J Ford
- Faculty of Medicine - The University of Newcastle, Australia; Gosford Hospital Central Coast Local Health District, NSW Health, Australia; School Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom; NHS Golden Jubilee Hospital, Clydebank, United Kingdom
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Riccardo Rinaldi
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart Rome, Italy
| | - Adam Bland
- Faculty of Medicine - The University of Newcastle, Australia; Gosford Hospital Central Coast Local Health District, NSW Health, Australia
| | - Andrew Morrow
- School Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom; NHS Golden Jubilee Hospital, Clydebank, United Kingdom
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, United States
| | - Colin Berry
- School Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom; NHS Golden Jubilee Hospital, Clydebank, United Kingdom
| | - Juan Carlos Kaski
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| |
Collapse
|
18
|
Mathew RO, Sidhu MS, Rihal CS, Lennon R, El-Hajjar M, Yager N, Lyubarova R, Abdul-Nour K, Weitz S, O'Cochlain DF, Murthy V, Levisay J, Marzo K, Graham J, Dzavik V, So D, Goodman S, Rosenberg YD, Pereira N, Farkouh ME. Safety and Efficacy of CYP2C19 Genotype-Guided Escalation of P2Y 12 Inhibitor Therapy After Percutaneous Coronary Intervention in Chronic Kidney Disease: a Post Hoc Analysis of the TAILOR-PCI Study. Cardiovasc Drugs Ther 2024; 38:447-457. [PMID: 36445624 PMCID: PMC10225474 DOI: 10.1007/s10557-022-07392-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Chronic kidney disease (CKD) is a risk factor for ischemic and bleeding events with dual antiplatelet therapy after percutaneous coronary intervention (PCI). Whether the presence of CYP2C19 loss of function (LOF) alleles modifies this risk, and whether a genotype-guided (GG) escalation of P2Y12 inhibitor therapy post PCI is safe in this population is unclear. METHODS This was a post hoc analysis of randomized patients in TAILOR PCI. Patients were divided into two groups based on estimated glomerular filtration rate (eGFR) threshold of < 60 ml/min/1.73 m2 for CKD (n = 539) and non-CKD (n = 4276). The aggregate of cardiovascular death, stroke, myocardial infarction, stent thrombosis, and severe recurrent coronary ischemia at 12-months post-PCI was assessed as the primary endpoint. Secondary endpoint was major or minor bleeding. RESULTS Mean (standard deviation) eGFR among patients with CKD was 49.5 (8.4) ml/min/1.72 m2. Among all patients, there was no significant interaction between randomized strategy and CKD status for any endpoint. Among LOF carriers, the interaction between randomized strategy and CKD status on composite ischemic outcome was not significant (p = 0.2). GG strategy was not associated with an increased risk of bleeding in either CKD group. CONCLUSIONS In this exploratory analysis, escalation of P2Y12 inhibitor therapy following a GG strategy did not reduce the primary outcome in CKD. However, P2Y12 inhibitor escalation following a GG strategy was not associated with increased bleeding risk in CKD. Larger studies in CKD are needed. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT01742117?term=TAILOR-PCI&draw=2&rank=1 . NCT01742117.
Collapse
Affiliation(s)
- Roy O Mathew
- Department of Medicine, Loma Linda VA Health Care System, 11201 Benton Street, Loma Linda, CA, 92357, USA.
| | - Mandeep S Sidhu
- Department of Medicine, Albany Medical College, 43 New Scotland Avenue Albany, Schenectady, NY, 12208, USA.
| | | | - Ryan Lennon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Mohammed El-Hajjar
- Department of Medicine, Albany Medical College, 43 New Scotland Avenue Albany, Schenectady, NY, 12208, USA
| | - Neil Yager
- Department of Medicine, Albany Medical College, 43 New Scotland Avenue Albany, Schenectady, NY, 12208, USA
| | - Radmila Lyubarova
- Department of Medicine, Albany Medical College, 43 New Scotland Avenue Albany, Schenectady, NY, 12208, USA
| | | | - Steven Weitz
- Cardiology Associates of Schenectady, Schenectady, NY, USA
| | | | - Vishakantha Murthy
- Department of Endocrine and Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Justin Levisay
- Department of Medicine, North Shore University Health System, Evanston, IL, USA
| | - Kevin Marzo
- Department of Medicine, Winthrop University Hospital, Mineola, NY, USA
| | - John Graham
- Department of Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | - Vlad Dzavik
- Department of Medicine, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Derek So
- Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Shaun Goodman
- Department of Medicine, St. Michael's Hospital, Toronto, ON, Canada
| | | | | | - Michael E Farkouh
- Department of Medicine, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
19
|
Pradhan A, Bhandari M, Vishwakarma P, Sethi R. Clopidogrel resistance and its relevance: Current concepts. J Family Med Prim Care 2024; 13:2187-2199. [PMID: 39027844 PMCID: PMC11254075 DOI: 10.4103/jfmpc.jfmpc_1473_23] [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: 09/05/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 07/20/2024] Open
Abstract
Clopidogrel is the most widely used P2Y12 receptor inhibitor (P2Y12i) as a part of dual antiplatelet therapy along with aspirin. Clopidogrel is a pro-drug and is metabolized to its active metabolite by the hepatic enzyme cytochrome P4502C19 (CYP2C19). This active metabolite is responsible for the antiplatelet action of clopidogrel. Recent studies have demonstrated that single nucleotide polymorphisms in the CYP2C19 gene, including CYP2C19*2,*3,*4, and *5 alleles, result in reduced production of the active metabolite of clopidogrel, and hence reduced inhibition of platelet aggregation. This in turn enhances the incidence of stent thrombosis and recurrent cardiovascular (CV) events. We report a case of coronary stent thrombosis due to clopidogrel resistance proven by CYP2C19 genotyping. We then review the literature on clopidogrel resistance and its impact on CV outcomes. Subsequently, we discuss the methods of diagnosis of resistance, evidence from clinical trials for tailoring clopidogrel therapy, the role of potent P2Y12 inhibitors, the current guidelines, and future directions.
Collapse
Affiliation(s)
- Akshyaya Pradhan
- Department of Cardiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Monika Bhandari
- Department of Cardiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Pravesh Vishwakarma
- Department of Cardiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Rishi Sethi
- Department of Cardiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Cha J, Lee S, Park JH, Hong SJ, Ahn TH, Chang K, Park Y, Song YB, Ahn SG, Suh J, Lee SY, Cho JR, Her A, Jeong Y, Kim H, Kim MH, Shin E, Kim B, Lim D. Association of Age- and Body Mass Index-Stratified High On-Treatment Platelet Reactivity With Coronary Intervention Outcomes in East Asian Patients. J Am Heart Assoc 2024; 13:e031819. [PMID: 38639339 PMCID: PMC11179895 DOI: 10.1161/jaha.123.031819] [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: 07/20/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Although age and body mass index (BMI) significantly affect platelet reactivity units and clinical outcomes after percutaneous coronary intervention, there are limited data on the relationship between high on-treatment platelet reactivity (HPR) and clinical outcomes on age and BMI differences. Thus, we investigated the association of HPR with clinical outcomes according to age and BMI. METHODS AND RESULTS The study analyzed 11 714 patients who underwent platelet function tests after percutaneous coronary intervention. The primary end point was the occurrence of major adverse cardiac and cerebrovascular events (MACCEs), whereas the secondary end point was major bleeding. HPR was defined as platelet reactivity units ≥252. Patients were categorized by age (<67 years of age or ≥67 years of age) and BMI (≤22.6 kg/m2 or >22.6 kg/m2). Patients <67 years of age with HPR had increases in both MACCEs (adjusted hazard ratio [HR], 1.436 [95% CI, 1.106-1.867]; P=0.007) and major bleeding (adjusted HR, 1.584 [95% CI, 1.095-2.290]; P=0.015) compared with the those with non-HPR, respectively. In patients ≥67 years of age with HPR, there were no differences in MACCEs, but there was a decrease in major bleeding (adjusted HR, 0.721 [95% CI, 0.542-0.959]; P=0.024). Meanwhile, patients with HPR with BMI >22.6 kg/m2 had increases in MACCEs (adjusted HR, 1.387 [95% CI, 1.140-1.688]; P=0.001). No differences were shown in major bleeding. CONCLUSIONS HPR was linked to an increase in MACCEs or a decrease in major bleeding in patients after percutaneous coronary intervention, depending on age and BMI. This study is the first to observe that clinical outcomes in patients with HPR after percutaneous coronary intervention may vary based on age and BMI. Because the study is observational, the results should be viewed as hypothesis generating and emphasize the need for randomized clinical trials.
Collapse
Affiliation(s)
- Jung‐Joon Cha
- Department of Cardiology, Cardiovascular CenterKorea University Anam Hospital, Korea University College of MedicineSeoulSouth Korea
| | | | - Jae Hyoung Park
- Department of Cardiology, Cardiovascular CenterKorea University Anam Hospital, Korea University College of MedicineSeoulSouth Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular CenterKorea University Anam Hospital, Korea University College of MedicineSeoulSouth Korea
| | - Tae Hoon Ahn
- Cardiovascular CenterChung‐Ang University Gwang‐myeong Hospital, Chung‐Ang University College of MedicineGwang‐myeongSouth Korea
| | - Kiyuk Chang
- Division of Cardiology, Department of Internal Medicine, College of MedicineCatholic University of KoreaSeoulSouth Korea
| | - Yongwhi Park
- Department of Internal MedicineGyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon HospitalChangwonSouth Korea
| | - Young Bin Song
- Division of Cardiology, Department of Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Sung Gyun Ahn
- Department of CardiologyYonsei University Wonju Severance Christian HospitalWonjuSouth Korea
| | - Jung‐Won Suh
- Department of Cardiology, Seoul National University College of MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| | - Sang Yeub Lee
- Cardiovascular CenterChung‐Ang University Gwang‐myeong Hospital, Chung‐Ang University College of MedicineGwang‐myeongSouth Korea
| | - Jung Rae Cho
- Cardiology Division, Department of Internal MedicineKangnam Sacred Heart Hospital, Hallym University College of MedicineSeoulSouth Korea
| | - Ae‐Young Her
- Division of Cardiology, Department of Internal MedicineKangwon National University School of MedicineChuncheonSouth Korea
| | - Young‐Hoon Jeong
- Cardiovascular CenterChung‐Ang University Gwang‐myeong Hospital, Chung‐Ang University College of MedicineGwang‐myeongSouth Korea
| | - Hyo‐Soo Kim
- Department of Internal Medicine and Cardiovascular CenterSeoul National University HospitalSeoulSouth Korea
| | - Moo Hyun Kim
- Department of CardiologyDong‐A University HospitalBusanSouth Korea
| | - Eun‐Seok Shin
- Division of CardiologyUlsan University Hospital, University of Ulsan College of MedicineUlsanSouth Korea
| | | | - Do‐Sun Lim
- Department of Cardiology, Cardiovascular CenterKorea University Anam Hospital, Korea University College of MedicineSeoulSouth Korea
| |
Collapse
|
21
|
Birocchi S, Rocchetti M, Minardi A, Podda GM, Squizzato A, Cattaneo M. Guided Anti-P2Y12 Therapy in Patients Undergoing PCI: Three Systematic Reviews with Meta-analyses of Randomized Controlled Trials with Homogeneous Design. Thromb Haemost 2024; 124:482-496. [PMID: 37549688 DOI: 10.1055/a-2149-4344] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
BACKGROUND The value of guided therapy (GT) with anti-P2Y12 drugs in percutaneous coronary intervention (PCI) is unclear. Meta-analyses lumped together randomized controlled trials (RCTs) with heterogeneous designs, comparing either genotype-GT or platelet function test (PFT)-GT with unguided therapy. Some meta-analysis also included RCTs that did not explore GT, but included the effects of switching patients with high on-treatment platelet reactivity (HTPR) to alternative therapies (HTPR-Therapy). We performed three distinct systematic reviews/meta-analyses, each exploring only RCTs with homogeneous design. METHODS MEDLINE, Embase, and Central databases were searched for RCTs testing genotype-GT, PFT-GT, or HTPR-Therapy in PCI-treated patients, through October 1, 2022. Two reviewers extracted the data. Risk ratios (RRs) (95% confidence intervals) were calculated. Primary outcomes were major bleedings (MBs) and major adverse cardiovascular events (MACE). RESULTS In seven genotype-GT RCTs, RRs were: MB, 1.06 (0.73-1.54; p = 0.76); MACE, 0.65 (0.47-0.91; p = 0.01), but significant risk reduction was observed in RCTs performed in China (0.30, 0.16-0.54; p < 0.0001) and not elsewhere (0.75, 0.48-1.18; p = 0.21). In six PFT-GT RCTs, RRs were: MB, 0.91 (0.64-1.28, p = 0.58); MACE, 0.82 (0.56-1.19; p = 0.30): 0.62 (0.42-0.93; p = 0.02) in China, 1.08 (0.82-1.41; p = 0.53) elsewhere. In eight HTPR-Therapy RCTs, RRs were: MB, 0.71 (0.41-1.23; p = 0.22); MACE, 0.57 (0.44-0.75; p < 0.0001): 0.56 (0.43-0.74, p < 0.0001) in China, 0.58 (0.27-1.23, p = 0.16) elsewhere. CONCLUSION No GT strategy affected MB. Overall, genotype-GT but not PFT-GT reduced MACE. However, genotype-GT and PFT-GT reduced MACE in China, but not elsewhere. PFT-GT performed poorly compared to HTPR-Therapy, likely due to inaccurate identification of HTPR patients by PFT.
Collapse
Affiliation(s)
- Simone Birocchi
- Divisione di Medicina Generale II, Dipartimento di Scienze della Salute, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
| | - Matteo Rocchetti
- Divisione di Cardiologia, Dipartimento di Scienze della Salute, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Minardi
- Divisione di Cardiologia, Dipartimento di Scienze della Salute, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
| | - Gian Marco Podda
- Divisione di Medicina Generale II, Dipartimento di Scienze della Salute, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Squizzato
- Research Center on Thromboembolic Disorders and Antithrombotic Therapies, ASST Lariana, University of Insubria, Como, Italy
| | - Marco Cattaneo
- Divisione di Medicina Generale II, Dipartimento di Scienze della Salute, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
- Fondazione Arianna Anticoagulazione, Bologna, Italy
| |
Collapse
|
22
|
Rantamo A, Gallé C, Numminen J, Virta J, Tanskanen P, Lindroos AC, Resendiz-Nieves J, Lehecka M, Niemelä M, Haeren R, Raj R. Flow diversion of ruptured intracranial aneurysms: a single-center study with a standardized antithrombotic treatment protocol. Acta Neurochir (Wien) 2024; 166:130. [PMID: 38467916 PMCID: PMC10927838 DOI: 10.1007/s00701-024-06029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND The use of antithrombotic medication following acute flow diversion for a ruptured intracranial aneurysm (IA) is challenging with no current guidelines. We investigated the incidence of treatment-related complications and patient outcomes after flow diversion for a ruptured IA before and after the implementation of a standardized antithrombotic medication protocol. METHODS We conducted a single-center retrospective study including consecutive patients treated for acutely ruptured IAs with flow diversion during 2015-2023. We divided the patients into two groups: those treated before the implementation of the protocol (pre-protocol) and those treated after the implementation of the protocol (post-protocol). The primary outcomes were hemorrhagic and ischemic complications. A secondary outcome was clinical outcome using the modified Ranking Scale (mRS). RESULTS Totally 39 patients with 40 ruptured IAs were treated with flow diversion (69% pre-protocol, 31% post-protocol). The patient mean age was 55 years, 62% were female, 63% of aneurysms were in the posterior circulation, 92% of aneurysms were non-saccular, and 44% were in poor grade on admission. Treatment differences included the use of glycoprotein IIb/IIIa inhibitors (pre-group 48% vs. post-group 100%), and the use of early dual antiplatelets (pre-group 44% vs. 92% post-group). The incidence of ischemic complications was 37% and 42% and the incidence of hemorrhagic complications was 30% and 33% in the pre- and post-groups, respectively, with no between-group differences. There were three (11%) aneurysm re-ruptures in the pre-group and none in the post-group. There were no differences in mortality or mRS 0-2 between the groups at 6 months. CONCLUSION We found no major differences in the incidence of ischemic or hemorrhagic complications after the implementation of a standardized antithrombotic protocol for acute flow diversion for ruptured IAs. There is an urgent need for more evidence-based guidelines to optimize antithrombotic treatment after flow diversion in the setting of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Anni Rantamo
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland.
| | - Camille Gallé
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
- Department of Neurosurgery, Maastricht University Medical Center, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Jussi Numminen
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| | - Jyri Virta
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| | - Päivi Tanskanen
- Anaesthesiology and Intensive Care Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Ann-Christine Lindroos
- Anaesthesiology and Intensive Care Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Julio Resendiz-Nieves
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| | - Martin Lehecka
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| | - Roel Haeren
- Department of Neurosurgery, Maastricht University Medical Center, PO box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Rahul Raj
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Po Box 320, 00029 HUS, Helsinki, Finland
| |
Collapse
|
23
|
Kobayashi N, Shibata Y, Kurihara O, Todoroki T, Tsutsumi M, Shirakabe A, Shigihara S, Sawatani T, Kiuchi K, Takano M, Asai K. Clinical Background and Coronary Artery Lesions Characteristics in Japanese Patients With Acute Coronary Syndrome Suffering Major Bleeding. Circ Rep 2024; 6:64-73. [PMID: 38464989 PMCID: PMC10920023 DOI: 10.1253/circrep.cr-24-0003] [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: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 03/12/2024] Open
Abstract
Background: Although the clinical factors that predict major bleeding in Western patients with acute coronary syndrome (ACS) are becoming elucidated, they have not been fully investigated, especially coronary lesion characteristics, in a Japanese population. Methods and Results: ACS patients (n=1,840) were divided into a "bleeding group" and a "no-bleeding group," according to whether they had major bleeding during the 2-year follow-up period, to investigate the prognostic effect of bleeding and the predictive factors of bleeding. Among them, patients who underwent primary percutaneous coronary intervention with optical coherence tomography (OCT) guidance (n=958) were examined to identify the effect of coronary lesion characteristics on bleeding. Of the 1,840 enrolled patients, 124 (6.7%) experienced major bleeding during the 2-year follow-up period. Incidence of cardiovascular death during the 2-year follow-up period was significantly higher among patients with major bleeding (26.4% vs. 8.5%, P=0.001). OCT examination showed that disrupted fibrous cap (DFC: 68% vs. 48%, P=0.014) and calcified plaque (63% vs. 42%, P=0.011) were more prevalent in the bleeding group. DFC was a predictor of major bleeding in the multivariate Cox proportional hazards analyses (hazard ratio 2.135 [95% confidence interval 1.070-4.263], P<0.001). Conclusions: ACS patients with major bleeding had poorer cardiac outcomes. Advanced atherosclerosis at the culprit lesion influences the higher incidence of major bleeding in ACS patients.
Collapse
Affiliation(s)
- Nobuaki Kobayashi
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Yusaku Shibata
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Osamu Kurihara
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Takahiro Todoroki
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Masayuki Tsutsumi
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Akihiro Shirakabe
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Shota Shigihara
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Tomofumi Sawatani
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Kazutaka Kiuchi
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Masamichi Takano
- Department of Cardiology, Nippon Medical School Chiba Hokusoh Hospital Inzai Japan
| | - Kuniya Asai
- Department of Cardiovascular Medicine, Nippon Medical School Tokyo Japan
| |
Collapse
|
24
|
Patel RC, Jones JE, Stouffer GA. P2Y12 Inhibitors in STEMI Patients - One Size Does Not Fit All. Cardiovasc Drugs Ther 2024; 38:5-7. [PMID: 37530947 PMCID: PMC11098026 DOI: 10.1007/s10557-023-07497-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Affiliation(s)
- Rajiv C Patel
- Division of Cardiology and the McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey E Jones
- Division of Cardiology and the McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - George A Stouffer
- Division of Cardiology and the McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
Robaux V, Kautbally S, Ginion A, Dechamps M, Lejeune S, Menghoum N, Bertrand L, Pouleur AC, Horman S, Beauloye C. Dual antiplatelet therapy is associated with high α-tubulin acetylation in circulating platelets from coronary artery disease patients. Platelets 2023; 34:2250002. [PMID: 37700239 DOI: 10.1080/09537104.2023.2250002] [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: 03/07/2023] [Revised: 07/20/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
Platelet inhibition is the main treatment strategy to prevent atherothrombotic complications after acute coronary syndrome or percutaneous coronary intervention. Despite dual antiplatelet therapy (DAPT) combining aspirin and a P2Y12 receptor inhibitor, high on-treatment platelet reactivity (HPR) persists in some patients due to poor response to treatment and is associated with ischemic risk. Tubulin acetylation has been pointed out as a hallmark of stable microtubules responsible for the discoid shape of resting platelets. However, the impact of antiplatelet treatments on this post-translational modification has never been studied. This study investigated whether tubulin acetylation differs according to antiplatelet therapy and on-treatment platelet reactivity. Platelets were isolated from arterial blood samples of 240 patients admitted for coronary angiography, and levels of α-tubulin acetylation on lysine 40 (α-tubulin K40 acetylation) were assessed by western blot. We show that platelet α-tubulin K40 acetylation was significantly increased in DAPT-treated patients. In addition, the proportion of patients with high levels of α-tubulin K40 acetylation was drastically reduced among DAPT-treated patients with HPR. Multivariate logistic regression confirmed that DAPT resulting in adequate platelet inhibition was strongly associated with elevated α-tubulin K40 acetylation. In conclusion, our study highlights the role of elevated platelet α-tubulin K40 acetylation as a marker of platelet inhibition in response to DAPT.Clinical trial registration: https://clinicaltrials.gov - NCT03034148.
Collapse
Affiliation(s)
- Valentine Robaux
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Shakeel Kautbally
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Audrey Ginion
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Mélanie Dechamps
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sibille Lejeune
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Nassiba Menghoum
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Anne-Catherine Pouleur
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
26
|
Silvain J, Lattuca B, Puymirat E, Ducrocq G, Dillinger JG, Lhermusier T, Procopi N, Cachanado M, Drouet E, Abergel H, Danchin N, Montalescot G, Simon T, Steg PG. Impact of transfusion strategy on platelet aggregation and biomarkers in myocardial infarction patients with anemia. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2023; 9:647-657. [PMID: 37609995 DOI: 10.1093/ehjcvp/pvad055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/10/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Higher rates of thrombotic events have been reported in myocardial infarction (MI) patients requiring blood transfusion. The impact of blood transfusion strategy on thrombosis and inflammation is still unknown. OBJECTIVE To compare the impact of a liberal vs. a restrictive transfusion strategy on P2Y12 platelet reactivity and biomarkers in the multicentric randomized REALITY trial. METHODS Patients randomized to a liberal (hemoglobin ≤10 g/dL) or a restrictive (hemoglobin ≤8 g/dL) transfusion strategy had VASP-PRI platelet reactivity measured centrally in a blinded fashion and platelet reactivity unit (PRU) measured locally using encrypted VerifyNow; at baseline and after randomization. Biomarkers of thrombosis (P-selectin, PAI-1, vWF) and inflammation (TNF-α) were also measured. The primary endpoint was the change in the VASP-PRI (difference from baseline and post randomization) between the randomized groups. RESULTS A total of 100 patients randomized were included in this study (n = 50 in each group). Transfused patients received on average 2.4 ± 1.6 units of blood. We found no differences in change of the VASP PRI (difference 1.2% 95% CI (-10.3-12.7%)) or by the PRU (difference 13.0 95% CI (-21.8-47.8)) before and after randomization in both randomized groups. Similar results were found in transfused patients (n = 71) regardless of the randomized group, VASP PRI (difference 1.7%; 95% CI (-9.5-1.7%)) or PRU (difference 27.0; 95% CI (-45.0-0.0)). We did not find an impact of transfusion strategy or transfusion itself in the levels of P-selectin, PAI-1, vWF, and TNF-α. CONCLUSION In this study, we found no impact of a liberal vs. a restrictive transfusion strategy on platelet reactivity and biomarkers in MI patients with anemia. A conclusion that should be tempered due to missing patients with exploitable biological data that has affected our power to show a difference.
Collapse
Affiliation(s)
- Johanne Silvain
- Sorbonne Université, ACTION Group, INSERM UMRS1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Paris 75013, France
| | - Benoit Lattuca
- Cardiology Department, Nîmes University Hospital, Montpellier University, ACTION study group, Nîmes 30900, France
| | - Etienne Puymirat
- Université Paris-Cité, AP-HP, Hôpital Européen Georges Pompidou, French Alliance for Cardiovascular Trials (FACT), Paris 75015, France
| | - Gregory Ducrocq
- Université Paris-Cité, AP-HP, French Alliance for Cardiovascular Trials (FACT), INSERM U1148, Paris 75018, France
| | - Jean-Guillaume Dillinger
- Department of Cardiology, Inserm U942, Hôpital Lariboisière, Assistance Publique - Hôpitaux de Paris, University Paris-Cité, Paris 75010, France
| | | | - Niki Procopi
- Sorbonne Université, ACTION Group, INSERM UMRS1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Paris 75013, France
| | - Marine Cachanado
- Department of Clinical Pharmacology-Clinical Research Platform (URCEST-CRB-CRCEST), AP-HP, Hôpital Saint Antoine, French Alliance for Cardiovascular Trials (FACT), Sorbonne-Université, Paris 75012, France
| | - Elodie Drouet
- Department of Clinical Pharmacology-Clinical Research Platform (URCEST-CRB-CRCEST), AP-HP, Hôpital Saint Antoine, French Alliance for Cardiovascular Trials (FACT), Sorbonne-Université, Paris 75012, France
| | - Helene Abergel
- Université Paris-Cité, AP-HP, French Alliance for Cardiovascular Trials (FACT), INSERM U1148, Paris 75018, France
| | - Nicolas Danchin
- Université Paris-Cité, AP-HP, Hôpital Européen Georges Pompidou, French Alliance for Cardiovascular Trials (FACT), Paris 75015, France
| | - Gilles Montalescot
- Sorbonne Université, ACTION Group, INSERM UMRS1166, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (AP-HP), Paris 75013, France
| | - Tabassome Simon
- Department of Clinical Pharmacology-Clinical Research Platform (URCEST-CRB-CRCEST), AP-HP, Hôpital Saint Antoine, French Alliance for Cardiovascular Trials (FACT), Sorbonne-Université, Paris 75012, France
| | - Philippe Gabriel Steg
- Université Paris-Cité, AP-HP, French Alliance for Cardiovascular Trials (FACT), INSERM U1148, Paris 75018, France
- Institut Universitaire de France, Paris 75005, France
| |
Collapse
|
27
|
Marx N, Federici M, Schütt K, Müller-Wieland D, Ajjan RA, Antunes MJ, Christodorescu RM, Crawford C, Di Angelantonio E, Eliasson B, Espinola-Klein C, Fauchier L, Halle M, Herrington WG, Kautzky-Willer A, Lambrinou E, Lesiak M, Lettino M, McGuire DK, Mullens W, Rocca B, Sattar N. 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023; 44:4043-4140. [PMID: 37622663 DOI: 10.1093/eurheartj/ehad192] [Citation(s) in RCA: 507] [Impact Index Per Article: 253.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/26/2023] Open
|
28
|
Brambilla M, Becchetti A, Rovati GE, Cosentino N, Conti M, Canzano P, Giesen PL, Loffreda A, Bonomi A, Cattaneo M, De Candia E, Podda GM, Trabattoni D, Werba PJ, Campodonico J, Pinna C, Marenzi G, Tremoli E, Camera M. Cell Surface Platelet Tissue Factor Expression: Regulation by P2Y 12 and Link to Residual Platelet Reactivity. Arterioscler Thromb Vasc Biol 2023; 43:2042-2057. [PMID: 37589138 PMCID: PMC10521789 DOI: 10.1161/atvbaha.123.319099] [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: 04/21/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND ADP-induced platelet activation leads to cell surface expression of several proteins, including TF (tissue factor). The role of ADP receptors in platelet TF modulation is still unknown. We aimed to assess the (1) involvement of P2Y1 and P2Y12 receptors in ADP-induced TF exposure; (2) modulation of TFpos-platelets in anti-P2Y12-treated patients with coronary artery disease. Based on the obtained results, we revisited the intracellular localization of TF in platelets. METHODS The effects of P2Y1 or P2Y12 antagonists on ADP-induced TF expression and activity were analyzed in vitro by flow cytometry and thrombin generation assay in blood from healthy subjects, P2Y12-/-, and patients with gray platelet syndrome. Ex vivo, P2Y12 inhibition of TF expression by clopidogrel/prasugrel/ticagrelor, assessed by VASP (vasodilator-stimulated phosphoprotein) platelet reactivity index, was investigated in coronary artery disease (n=238). Inhibition of open canalicular system externalization and electron microscopy (TEM) were used for TF localization. RESULTS In blood from healthy subjects, stimulated in vitro by ADP, the percentage of TFpos-platelets (17.3±5.5%) was significantly reduced in a concentration-dependent manner by P2Y12 inhibition only (-81.7±9.5% with 100 nM AR-C69931MX). In coronary artery disease, inhibition of P2Y12 is paralleled by reduction of ADP-induced platelet TF expression (VASP platelet reactivity index: 17.9±11%, 20.9±11.3%, 40.3±13%; TFpos-platelets: 10.5±4.8%, 9.8±5.9%, 13.6±6.3%, in prasugrel/ticagrelor/clopidogrel-treated patients, respectively). Despite this, 15% of clopidogrel good responders had a level of TFpos-platelets similar to the poor-responder group. Indeed, a stronger P2Y12 inhibition (130-fold) is required to inhibit TF than VASP. Thus, a VASP platelet reactivity index <20% (as in prasugrel/ticagrelor-treated patients) identifies patients with TFpos-platelets <20% (92% sensitivity). Finally, colchicine impaired in vitro ADP-induced TF expression but not α-granule release, suggesting that TF is open canalicular system stored as confirmed by TEM and platelet analysis of patients with gray platelet syndrome. CONCLUSIONS Data show that TF expression is regulated by P2Y12 and not P2Y1; P2Y12 antagonists downregulate the percentage of TFpos-platelets. In clopidogrel good-responder patients, assessment of TFpos-platelets highlights those with residual platelet reactivity. TF is stored in open canalicular system, and its membrane exposure upon activation is prevented by colchicine.
Collapse
Affiliation(s)
- Marta Brambilla
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Alessia Becchetti
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Gian Enrico Rovati
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| | - Nicola Cosentino
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Maria Conti
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Paola Canzano
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | | | - Alessia Loffreda
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy (A.L.)
| | - Alice Bonomi
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Marco Cattaneo
- Unità di Medicina II, ASST Santi Paolo e Carlo, Department of Scienze della Salute (M. Cattaneo, G.M.P.), Università degli Studi di Milano, Italy
| | - Erica De Candia
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy (E.D.C.)
| | - Gian Marco Podda
- Unità di Medicina II, ASST Santi Paolo e Carlo, Department of Scienze della Salute (M. Cattaneo, G.M.P.), Università degli Studi di Milano, Italy
| | - Daniela Trabattoni
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Pablo Josè Werba
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Jeness Campodonico
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Christian Pinna
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | | | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| |
Collapse
|
29
|
Nardin M, Verdoia M, Cao D, Nardin S, Kedhi E, Galasso G, van ‘t Hof AWJ, Condorelli G, De Luca G. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention. J Clin Med 2023; 12:6074. [PMID: 37763014 PMCID: PMC10531614 DOI: 10.3390/jcm12186074] [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: 06/22/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The key role played by platelets in the atherosclerosis physiopathology, especially in the acute setting, is ascertained: they are the main actors during thrombus formation and, thus, one of the major investigated elements related to atherothrombotic process involving coronary arteries. Platelets have been studied from different points of view, according with the technology advances and the improvement in the hemostasis knowledge achieved in the last years. Morphology and reactivity constitute the first aspects investigated related to platelets with a significant body of evidence published linking a number of their values and markers to coronary artery disease and cardiovascular events. Recently, the impact of genetics on platelet activation has been explored with promising findings as additional instrument for patient risk stratification; however, this deserves further confirmations. Moreover, the interplay between immune system and platelets has been partially elucidated in the last years, providing intriguing elements that will be basic components for future research to better understand platelet regulation and improve cardiovascular outcome of patients.
Collapse
Affiliation(s)
- Matteo Nardin
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Third Medicine Division, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13875 Biella, Italy
- Department of Translational Medicine, Eastern Piedmont University, 28100 Novara, Italy
| | - Davide Cao
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Department of Cardiology, Humanitas Gavazzeni Hospital, 24125 Bergamo, Italy
| | - Simone Nardin
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, 16126 Genova, Italy
| | - Elvin Kedhi
- Division of Cardiology, Hopital Erasmus, Universitè Libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Gennaro Galasso
- Division of Cardiology, Ospedale Ruggi D’Aragona, Università di Salerno, 84084 Salerno, Italy
| | - Arnoud W. J. van ‘t Hof
- Department of Cardiology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), 6229 ER Maastricht, The Netherlands
- Department of Cardiology, Zuyderland Medical Center, 6419 PC Heerlen, The Netherlands
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Department of Cardiovascular Medicine, IRCCS-Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant’Ambrogio, 20157 Milan, Italy
| |
Collapse
|
30
|
Capodanno D, Angiolillo DJ. Personalised antiplatelet therapies for coronary artery disease: what the future holds. Eur Heart J 2023; 44:3059-3072. [PMID: 37345589 DOI: 10.1093/eurheartj/ehad362] [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: 03/05/2023] [Revised: 05/07/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
Coronary artery disease (CAD) is one of the leading causes of death globally, and antiplatelet therapy is crucial for both its prevention and treatment. Antiplatelet drugs such as aspirin and P2Y12 inhibitors are commonly used to reduce the risk of thrombotic events, including myocardial infarction, stroke, and stent thrombosis. However, the benefits associated with the use of antiplatelet drugs also come with a risk of bleeding complications. The ever-growing understanding of the poor prognostic implications associated with bleeding has set the foundations for defining strategies that can mitigate such safety concern without any trade-off in antithrombotic protection. To this extent, personalised antiplatelet therapy has emerged as a paradigm that optimizes the balance between safety and efficacy by customizing treatment to the individual patient's needs and risk profile. Accurate risk stratification for both bleeding and thrombosis can aid in selecting the optimal antiplatelet therapy and prevent serious and life-threatening outcomes. Risk stratification has traditionally included clinical and demographic characteristics and has expanded to incorporate angiographic features and laboratory findings. The availability of bedside platelet function testing as well as rapid genotyping assays has also allowed for a more individualized selection of antiplatelet therapy. This review provides a comprehensive overview of the current state of the art and future trends in personalised antiplatelet therapy for patients with CAD, with emphasis on those presenting with an acute coronary syndrome and undergoing percutaneous coronary revascularization. The aim is to provide clinicians with a comprehensive understanding of personalised antiplatelet therapy and facilitate informed clinical decision-making.
Collapse
Affiliation(s)
- Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria 'G. Rodolico - San Marco', University of Catania, Via Santa Sofia, 78 - 95123 Catania, Italy
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine-Jacksonville, 655 West 8th Street, Jacksonville, FL 32209, USA
| |
Collapse
|
31
|
Shpigelman J, Proshkina A, Daly MJ, Cox D. Personalized Dual Antiplatelet Therapy in Acute Coronary Syndromes: Striking a Balance Between Bleeding and Thrombosis. Curr Cardiol Rep 2023; 25:693-710. [PMID: 37261665 PMCID: PMC10307718 DOI: 10.1007/s11886-023-01892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE OF REVIEW Dual antiplatelet therapy (DAPT)-aspirin in conjunction with a P2Y12 inhibitor-is the cornerstone of managing patients with acute coronary syndromes post-revascularization, but the clinical response is highly variable, with potentially devastating consequences. Herein, we review the mechanisms underpinning said variability and explore emerging approaches to normalizing therapeutic benefit. RECENT FINDINGS The potent P2Y12 inhibitors, prasugrel and ticagrelor, exhibit minimal inter-individual variability, replacing clopidogrel in DAPT and achieving greater rates of therapeutic response. However, these benefits decline in later phases when bleeding risk begins to supersede that of ischemia. Guided de-escalation of P2Y12 inhibition as well as shortening DAPT duration have emerged as strategies that retain antithrombotic efficacy while reducing bleeding risk. Aspirin is the other component of DAPT but is also used in isolation for secondary prevention of thrombotic disease. In contrast to the P2Y12 inhibitors, genetic influences on aspirin non-response appear to be outweighed by a triad of clinical factors: non-adherence, enteric aspirin use, and inappropriate dosing according to bodyweight and BMI. Multiple de-escalation strategies for DAPT have been shown to mitigate bleeding risk, but it remains unclear which approach is ideal, necessitating head-to-head investigations to determine which exhibits the most favorable cost-to-benefit ratio. However, there is likely a role for more than one approach in clinical practice, depending on patient risk profile. Our approach to aspirin use is also in need of reassessment: strategies to improve adherence, avoidance of enteric aspirin in cardiac patients, and dose adjustment according to bodyweight and/or BMI are all likely to improve rates of therapeutic response. Moreover, platelet function testing may have a role in identifying patients expected to benefit from primary prophylactic aspirin.
Collapse
Affiliation(s)
| | | | - Michael J Daly
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Cardiology, Connolly Hospital, Blanchardstown, Dublin, Ireland
| | - Dermot Cox
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
32
|
Perla HT, Thomson VS, Attumalil TV, Geevar T, Alex AG, Dave RG, Nair SC, Gowri SM, Mony PK, George P, Joseph G. Randomized, Double-Blind, Active Comparator Pharmacodynamic Study of Platelet Inhibition with Crushed and Integral Formulations of Clopidogrel and Ticagrelor in Acute Coronary Syndrome. Am J Cardiovasc Drugs 2023:10.1007/s40256-023-00591-8. [PMID: 37351814 DOI: 10.1007/s40256-023-00591-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Crushed formulations of specific antiplatelet agents produce earlier and stronger platelet inhibition. We studied the platelet inhibitory effect of crushed clopidogrel in patients with acute coronary syndrome (ACS) and its relative efficacy compared with integral clopidogrel, crushed and integral ticagrelor. OBJECTIVES We aimed to compare the platelet inhibitory effect of crushed and integral formulations of clopidogrel and ticagrelor in patients with acute coronary syndrome (ACS). METHODS Overall, 142 patients with suspected ACS were randomly assigned to receive crushed or integral formulations of clopidogrel or ticagrelor. Platelet inhibition at baseline and 1 and 8 h was assessed using the VerifyNow assay. High on-treatment platelet reactivity (HTPR) ≥ 235 P2Y12 reaction units (PRUs) 1 h after the medication loading dose was also determined. RESULTS The PRU and percentage inhibition median (interquartile range) at 1 h for the different formulations were as follows: crushed clopidogrel: 196.50 (155.50, 246.50), 9.36 (- 1.79, 25.10); integral clopidogrel: 189.50 (159.00, 214.00), 2.32 (- 2.67, 19.89); crushed ticagrelor: 59.00 (10.00, 96.00), 75.53 (49.12, 95.18); and integral ticagrelor: 126.50 (50.00, 168.00), 40.56 (25.59, 78.69). There was no significant difference in PRU or percentage platelet inhibition between the crushed and integral formulations of clopidogrel (p = 0.990, p = 0.479); both formulations of ticagrelor were superior to the clopidogrel formulations (p < 0.05). On paired comparison, crushed ticagrelor showed robust early inhibition of platelets compared with the integral formulation (p = 0.03). Crushed clopidogrel exhibited the maximal HTPR of 34.3%, but was < 3% for both formulations of ticagrelor. CONCLUSIONS The platelet inhibitory effect of crushed clopidogrel is not superior to integral preparation in patients with ACS. Crushed ticagrelor produced maximal platelet inhibition acutely. HTPR rates in ACS are similar and very low with both formulations of ticagrelor, and maximal with crushed clopidogrel. Clinical Trials Registry of India identifier number CTRI/2020/06/025647.
Collapse
Affiliation(s)
- Harsha Teja Perla
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India
| | - Viji Samuel Thomson
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India.
- Salalah Heart Center, Salalah, Oman.
| | - Thomas V Attumalil
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India
| | - Tulasi Geevar
- Department of Immunohematology and Transfusion Medicine, Christian Medical College and Hospital, Vellore, India
| | - Anoop George Alex
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India
| | - Rutvi G Dave
- Department of Immunohematology and Transfusion Medicine, Christian Medical College and Hospital, Vellore, India
| | - Sukesh C Nair
- Department of Immunohematology and Transfusion Medicine, Christian Medical College and Hospital, Vellore, India
| | - S Mahasampath Gowri
- Department of Biostatistics, Christian Medical College and Hospital, Vellore, India
| | - Prem K Mony
- Department of Epidemiology and Population Health, St John's National Academy of Health Sciences, Bangalore, India
| | - Paul George
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India
| | - George Joseph
- Department of Cardiology, Christian Medical College and Hospital, Vellore, India
| |
Collapse
|
33
|
Capodanno D, Mehran R, Krucoff MW, Baber U, Bhatt DL, Capranzano P, Collet JP, Cuisset T, De Luca G, De Luca L, Farb A, Franchi F, Gibson CM, Hahn JY, Hong MK, James S, Kastrati A, Kimura T, Lemos PA, Lopes RD, Magee A, Matsumura R, Mochizuki S, O'Donoghue ML, Pereira NL, Rao SV, Rollini F, Shirai Y, Sibbing D, Smits PC, Steg PG, Storey RF, Ten Berg J, Valgimigli M, Vranckx P, Watanabe H, Windecker S, Serruys PW, Yeh RW, Morice MC, Angiolillo DJ. Defining Strategies of Modulation of Antiplatelet Therapy in Patients With Coronary Artery Disease: A Consensus Document from the Academic Research Consortium. Circulation 2023; 147:1933-1944. [PMID: 37335828 DOI: 10.1161/circulationaha.123.064473] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/24/2023] [Indexed: 06/21/2023]
Abstract
Antiplatelet therapy is the mainstay of pharmacologic treatment to prevent thrombotic or ischemic events in patients with coronary artery disease treated with percutaneous coronary intervention and those treated medically for an acute coronary syndrome. The use of antiplatelet therapy comes at the expense of an increased risk of bleeding complications. Defining the optimal intensity of platelet inhibition according to the clinical presentation of atherosclerotic cardiovascular disease and individual patient factors is a clinical challenge. Modulation of antiplatelet therapy is a medical action that is frequently performed to balance the risk of thrombotic or ischemic events and the risk of bleeding. This aim may be achieved by reducing (ie, de-escalation) or increasing (ie, escalation) the intensity of platelet inhibition by changing the type, dose, or number of antiplatelet drugs. Because de-escalation or escalation can be achieved in different ways, with a number of emerging approaches, confusion arises with terminologies that are often used interchangeably. To address this issue, this Academic Research Consortium collaboration provides an overview and definitions of different strategies of antiplatelet therapy modulation for patients with coronary artery disease, including but not limited to those undergoing percutaneous coronary intervention, and consensus statements on standardized definitions.
Collapse
Affiliation(s)
- Davide Capodanno
- Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy (D.C., P.C.)
| | - Roxana Mehran
- Zena and Michael A. Wiener Cardiovascular Institute (R.M.), Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Usman Baber
- University of Oklahoma Health Sciences Center, Oklahoma City (U.B.)
| | - Deepak L Bhatt
- Mount Sinai Heart (D.L.B.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Piera Capranzano
- Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy (D.C., P.C.)
| | - Jean-Philippe Collet
- Sorbonne Université, ACTION Study Group, Institut de Cardiologie, Hôpital Pitié-Salpêtrière (APHP), Paris, France (J.-P.C.)
| | - Thomas Cuisset
- Interventional Cardiology Unit and Cathlab, Department of Cardiology, University Hospital, La Timone, Marseille, France (T.C.)
| | - Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G Martino," University of Messina, Italy (G.D.L.)
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, Milan, Italy (G.D.L.)
| | - Leonardo De Luca
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy (L.D.L.)
| | - Andrew Farb
- US Food and Drug Administration, Silver Spring, MD (A.F., A.M.)
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville (F.F., F.R., D.J.A.)
| | | | - Joo-Yong Hahn
- Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (J.-Y.H.)
| | - Myeong-Ki Hong
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea (M.-K.H.)
| | - Stefan James
- Department of Medical Sciences and Uppsala Clinical Research Center, Uppsala University, Sweden (S.J.)
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (A.K.)
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK; German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (A.K., D.S.)
| | - Takeshi Kimura
- Department of Cardiology, Hirakata Kohsai Hospital, Osaka, Japan (T.K.)
| | - Pedro A Lemos
- Hospital Israelita Albert Einstein, São Paulo, Brazil (P.A.L.)
| | - Renato D Lopes
- Duke University Medical Center, Durham, NC (M.W.K., R.D.L.)
| | - Adrian Magee
- US Food and Drug Administration, Silver Spring, MD (A.F., A.M.)
| | - Ryosuke Matsumura
- Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (R.M., S.M., Y.S.)
| | - Shuichi Mochizuki
- Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (R.M., S.M., Y.S.)
| | - Michelle L O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.L.O.)
| | - Naveen L Pereira
- Department of Cardiovascular Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN (N.L.P.)
| | - Sunil V Rao
- NYU Langone Health System, New York, NY (S.V.R.)
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville (F.F., F.R., D.J.A.)
| | - Yuko Shirai
- Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (R.M., S.M., Y.S.)
| | - Dirk Sibbing
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK; German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (A.K., D.S.)
- Ludwig-Maximilians University München, Munich, Germany (D.S.)
- Privatklinik Lauterbacher Mühle am Ostsee, Seeshaupt, Germany (D.S.)
| | - Peter C Smits
- Department of Cardiology, Maasstad Hospital, Rotterdam, the Netherlands (P.C.S.)
| | - P Gabriel Steg
- Université Paris-Cité, AP-HP, Paris, France (P.G.S.)
- INSERM U-1148/LVTS, Paris, France (P.G.S.)
- Institut Universitaire de France, Paris (P.G.S.)
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, UK (R.F.S.)
| | - Jurrien Ten Berg
- Cardiovascular Research Institute Maastricht (CARIM), School for Cardiovascular Diseases, Maastricht University Medical Center, the Netherlands (J.t.B.)
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, the Netherlands (J.t.B.)
- Department of Cardiology, University Medical Center Maastricht, the Netherlands (J.t.B.)
| | - Marco Valgimigli
- Cardiocentro Institute, Ente Ospedaliero Cantonale, Università della Svizzera Italiana (USI), Lugano, Switzerland (M.V.)
- University of Bern, Switzerland (M.V.)
| | - Pascal Vranckx
- Department of Cardiology and Critical Care Medicine, Jessa Ziekenhuis, Hasselt, Belgium (P.V.)
- Faculty of Medicine and Life Sciences, University of Hasselt, Belgium (P.V.)
| | | | - Stephan Windecker
- Department of Cardiology, Bern University Hospital, Inselspital (S.W.)
| | | | - Robert W Yeh
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (R.W.Y.)
| | | | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville (F.F., F.R., D.J.A.)
| |
Collapse
|
34
|
Berteotti M, Gori AM, Giusti B, Fortini A, Grossi G, Ciardetti N, Migliorini A, Lotti E, Valenti R, Di Mario C, Marchionni N, Marcucci R. Clinical impact of high platelet reactivity in patients with atrial fibrillation and concomitant percutaneous coronary intervention on dual or triple antithrombotic therapy. J Thromb Thrombolysis 2023; 55:667-679. [PMID: 36905562 PMCID: PMC10147742 DOI: 10.1007/s11239-023-02784-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/05/2023] [Indexed: 03/12/2023]
Abstract
High platelet reactivity (HPR) on clopidogrel is an established thrombotic risk factor after percutaneous coronary intervention (PCI). The introduction of more potent antiplatelet drugs has partially surpassed this issue. However, in the setting of concomitant atrial fibrillation (AF) and PCI clopidogrel is still the most adopted P2Y12 inhibitor. In the present study all consecutive patients with history of AF discharged from our cardiology ward with dual (DAT) or triple (TAT) antithrombotic therapy after a PCI from April 2018 to March 2021 were enrolled in an observational registry. For all subjects, blood serum samples were collected and tested for platelet reactivity by arachidonic acid and ADP (VerifyNow system) and genotyping of the CYP2C19*2 loss-of-function polymorphism. We recorded at 3 and 12-months follow-up: (1) major adverse cardiac and cerebrovascular events (MACCE), (2) major hemorrhagic or clinically relevant non-major bleeding and (3) all-cause mortality. A total of 147 patients were included (91, 62% on TAT). In 93.4% of patients, clopidogrel was chosen as P2Y12 inhibitor. P2Y12 dependent HPR resulted an independent predictor of MACCE both at 3 and 12 months (HR 2.93, 95% C.I. 1.03 to 7.56, p = 0.027 and HR 1.67, 95% C.I. 1.20 to 2.34, p = 0.003, respectively). At 3-months follow-up the presence of CYP2C19*2 polymorphism was independently associated with MACCE (HR 5.21, 95% C.I. 1.03 to 26.28, p = 0.045). In conclusion, in a real-world unselected population on TAT or DAT, the entity of platelet inhibition on P2Y12 inhibitor is a potent predictor of thrombotic risk, suggesting the clinical utility of this laboratory evaluation for a tailored antithrombotic therapy in this high-risk clinical scenario. The present analysis was performed in patients with AF undergoing PCI on dual or triple antithrombotic therapy. At 1 year follow-up MACCE incidence was consistent, and it was not different in different antithrombotic pattern groups. P2Y12 dependent HPR was a potent independent predictor of MACCE both at 3- and 12-months follow-up. In the first 3 months after stenting the carriage of CYP2C19*2 allele was similarly associated with MACCE. Abbreviation: DAT, dual antithrombotic therapy; HPR, high platelet reactivity; MACCE, major adverse cardiac and cerebrovascular events; PRU, P2Y12 reactive unit; TAT, triple antithrombotic therapy. Created with BioRender.com.
Collapse
Affiliation(s)
- M Berteotti
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy.
| | - A M Gori
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - B Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - A Fortini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - G Grossi
- Division of Interventional Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - N Ciardetti
- Division of Interventional Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - A Migliorini
- Division of Interventional Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - E Lotti
- Thrombosis Center, Careggi University Hospital, Florence, Italy
| | - R Valenti
- Division of Interventional Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - C Di Mario
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - N Marchionni
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - R Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| |
Collapse
|
35
|
Peng W, Zhang Y, Lin B, Lin Y. Clinical Outcomes of Individualized Antiplatelet Therapy Based on Platelet Function Test in Patients After Percutaneous Coronary Intervention: A Systematic Review and Meta-analysis. J Cardiovasc Pharmacol 2023; 81:270-279. [PMID: 36651931 DOI: 10.1097/fjc.0000000000001393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/03/2022] [Indexed: 01/19/2023]
Abstract
ABSTRACT Platelet function test (PFT) is universally used to assess platelet reactivity to antiplatelet drugs in patients after percutaneous coronary intervention (PCI). However, it remains controversial whether individualized antiplatelet therapy guided by PFT can improve the prognosis in patients after PCI. This meta-analysis was conducted to explore the efficacy and safety of individualized antiplatelet therapy guided by PFT in patients after PCI. Studies that compared PFT-guided antiplatelet therapy with standard antiplatelet therapy were researched. The risks of major adverse cardiovascular and cerebrovascular events (MACCE) and major bleeding events were assessed. Pooled odds ratios (ORs) with 95% CIs were obtained. Finally, a total of 16,835 patients from 22 studies met the criteria and were included in the meta-analysis. Compared with standard antiplatelet therapy, individualized antiplatelet therapy guided by PFT significantly decreased the risk of MACCE (OR: 0.58, 95% CI: 0.43-0.77) in patients after PCI. There was no significant difference in major bleeding events (OR: 0.85, 95% CI: 0.70-1.05, P = 0.13). This study identified that PFT-guided individualized antiplatelet therapy could reduce the incidence of MACCE without increasing the risk of hemorrhage in patients after PCI.
Collapse
Affiliation(s)
- Wenxing Peng
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
| | - Yunnan Zhang
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Baidi Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Yang Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; and
| |
Collapse
|
36
|
Tscharre M, Gremmel T. Antiplatelet Therapy in Coronary Artery Disease: Now and Then. Semin Thromb Hemost 2023; 49:255-271. [PMID: 36455618 DOI: 10.1055/s-0042-1758821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Cardiovascular disease, particularly coronary artery disease (CAD), remains the leading cause of mortality and morbidity in industrialized countries. Platelet activation and aggregation at the site of endothelial injury play a key role in the processes ultimately resulting in thrombus formation with vessel occlusion and subsequent end-organ damage. Consequently, antiplatelet therapy has become a mainstay in the pharmacological treatment of CAD. Several drug classes have been developed over the last decades and a broad armamentarium of antiplatelet agents is currently available. This review portrays the evolution of antiplatelet therapy, and provides an overview on previous and current antiplatelet drugs and strategies.
Collapse
Affiliation(s)
- Maximilian Tscharre
- Department of Internal Medicine, Cardiology and Nephrology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria.,Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Thomas Gremmel
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, Mistelbach, Austria.,Institute of Antithrombotic Therapy in Cardiovascular Disease, Karl Landsteiner Society, St. Pölten, Austria
| |
Collapse
|
37
|
Huseynov A, Reinhardt J, Chandra L, Dürschmied D, Langer HF. Novel Aspects Targeting Platelets in Atherosclerotic Cardiovascular Disease—A Translational Perspective. Int J Mol Sci 2023; 24:ijms24076280. [PMID: 37047253 PMCID: PMC10093962 DOI: 10.3390/ijms24076280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Platelets are important cellular targets in cardiovascular disease. Based on insights from basic science, translational approaches and clinical studies, a distinguished anti-platelet drug treatment regimen for cardiovascular patients could be established. Furthermore, platelets are increasingly considered as cells mediating effects “beyond thrombosis”, including vascular inflammation, tissue remodeling and healing of vascular and tissue lesions. This review has its focus on the functions and interactions of platelets with potential translational and clinical relevance. The role of platelets for the development of atherosclerosis and therapeutic modalities for primary and secondary prevention of atherosclerotic disease are addressed. Furthermore, novel therapeutic options for inhibiting platelet function and the use of platelets in regenerative medicine are considered.
Collapse
|
38
|
Jin H, Song J, Shen X, Liang Q, Sun G, Yu Y. Multiple genetic mutations increase the risk of thrombosis associated with clopidogrel after percutaneous coronary intervention. Pharmacogenomics 2023; 24:227-237. [PMID: 36891827 DOI: 10.2217/pgs-2022-0167] [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: 03/10/2023] Open
Abstract
Background: The effect of multiple mutations in CYP2C19, PON1 and ABCB1 genes on the effectiveness and safety of dual antiplatelet therapy after percutaneous coronary intervention remains unclear. Methods: In total, 263 Chinese Han patients were enrolled in this study. Platelet aggregation rates and thrombosis risk were used to compare clopidogrel responses and outcomes in patients with different numbers of genetic mutations. Results: Our study demonstrated that 74% of the patients carried more than two genetic mutations. High platelet aggregation rates were associated with genetic mutations in patients receiving clopidogrel and aspirin after percutaneous coronary intervention. Genetic mutations were closely related to the recurrence of thrombotic events, but not bleeding. The number of genes that become dysfunctional in patients is directly correlated with the risk of recurrent thrombosis. Conclusion: Compared with CYP2C19 alone or the platelet aggregation rate, it is more helpful to predict clinical outcomes by considering the polymorphisms of all three genes.
Collapse
Affiliation(s)
- Hui Jin
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Jinfei Song
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Xiaoying Shen
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Qing Liang
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Guangchun Sun
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Yan Yu
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| |
Collapse
|
39
|
Rocca B, Patrono C. Precision antiplatelet therapy. Res Pract Thromb Haemost 2023; 7:100138. [PMID: 37215094 PMCID: PMC10193296 DOI: 10.1016/j.rpth.2023.100138] [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: 01/03/2023] [Revised: 02/23/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
A State of the Art lecture titled "Personalizing Antiplatelet Therapy Based on Platelet Turnover and Metabolic Phenotype" was presented by Bianca Rocca at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2022. Increased variability in drug response may be associated with serious, mechanism-based and off-target side effects, especially in the case of drugs that do not routinely undergo therapeutic drug monitoring, such as antiplatelet drugs or direct oral anticoagulants. Precision pharmacology can be defined as the identification of a drug regimen that maximizes the benefit/risk balance at the level of an individual patient. Key tools for identifying relevant sources of variability and developing precision drug dosing are represented by genetic, biochemical, and pharmacological biomarkers recognized as a valid surrogate or strong predictor of major clinical complications. Pharmacodynamic, pharmacokinetic, and/or disease-related biomarkers are central to identifying the right population to be targeted, characterizing the sources of variability in drug response, guiding precision treatments that maximize benefits and minimize risks, and designing precision dosing trials. Another valuable tool for guiding precision pharmacology is represented by in silico pharmacokinetic/pharmacodynamic models and simulations instructed by real-world data of validated biomarkers. This review critically analyzes the tools for precision dosing and exemplifies conditions in which precision dosing can considerably optimize the efficacy and safety of antiplatelet drugs, namely aspirin and P2Y12 receptor blockers, used alone and in combination. Finally, we summarize relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Bianca Rocca
- Section of Pharmacology, Catholic University School of Medicine and Fondazione Policlinico Universitario Agostino Gemelli and Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| | - Carlo Patrono
- Section of Pharmacology, Catholic University School of Medicine and Fondazione Policlinico Universitario Agostino Gemelli and Istituto di Ricerca e Cura a Carattere Scientifico, Rome, Italy
| |
Collapse
|
40
|
Hartmann J, Curzen N. Modified Thromboelastography for Peri-interventional Assessment of Platelet Function in Cardiology Patients: A Narrative Review. Semin Thromb Hemost 2023; 49:192-200. [PMID: 36252602 PMCID: PMC9894686 DOI: 10.1055/s-0042-1757545] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Viscoelastic testing (VET), such as thromboelastography, can measure whole blood coagulation dynamics in real time and is used across a range of clinical settings, including cardiac surgery, liver transplant, and trauma. The use of modified thromboelastography with platelet function assessment (TEG(R) PlateletMapping(R) Assay) can provide an analysis of platelet contribution to hemostasis, including the contribution of the P2Y12 receptor and thromboxane pathway to platelet function. The TEG PlateletMapping Assay has shown high correlation with the current gold standard test of platelet function, light transmission aggregometry, to measure arachidonic acid and adenosine diphosphate agonist-induced platelet activation. Studies have also shown comparable results with other whole blood platelet function tests. In this review, we explore the clinical applications of modified thromboelastography with platelet function assessment. This includes guiding dual antiplatelet therapy in relation to cardiac procedures, such as percutaneous coronary interventions, transcatheter aortic valve replacement, and left atrial appendage closure. We also explore the developing use of thromboelastography in the emergency care setting of coronavirus disease 2019, which is commonly associated with a hypercoagulable and hypofibrinolytic state. Despite a general lack of high-quality, grade 1 evidence regarding the use of modified thromboelastography with platelet function assessment in these disease areas, the ability of the TEG PlateletMapping Assay to measure global hemostasis and platelet reactivity rapidly and to view and evaluate results at the point of care makes it a promising area for further study for managing patient treatment and optimizing hemostatic therapy.
Collapse
Affiliation(s)
| | - Nick Curzen
- Faculty of Medicine, University of Southampton & Cardiothoracic Department, University Hospital Southampton, United Kingdom
| |
Collapse
|
41
|
Platelet Reactivity and Cardiovascular Mortality Risk in the LURIC Study. J Clin Med 2023; 12:jcm12051913. [PMID: 36902699 PMCID: PMC10003439 DOI: 10.3390/jcm12051913] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/14/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND The clinical and prognostic implications of platelet reactivity (PR) testing in a P2Y12-inhibitor naïve population are poorly understood. OBJECTIVES This explorative study aims to assess the role of PR and explore factors that may modify elevated mortality risk in patients with altered PR. METHODS Platelet ADP-induced CD62P and CD63 expression were measured by flow-cytometry in 1520 patients who were referred for coronary angiography in the Ludwigshafen Risk and Cardiovascular Health Study (LURIC). RESULTS High- and Low-platelet reactivity to ADP were strong predictors of cardiovascular and all-cause mortality and risk equivalent to the presence of coronary artery disease. (High platelet reactivity 1.4 [95% CI 1.1-1.9]; Low platelet reactivity: 1.4 [95% CI 1.0-2.0]). Relative weight analysis indicated glucose control (HbA1c), renal function ([eGFR]), inflammation (high-sensitive C-reactive protein [hsCRP]) and antiplatelet therapy by Aspirin as consistent mortality risk modifiers in patients with Low- and High-platelet reactivity. Pre-specified stratification of patients by risk modifiers HbA1c (<7.0%), eGFR (>60 mL/min/1.73 m2) and CRP (<3 mg/L) was associated with a lower mortality risk, however irrespective of platelet reactivity. Aspirin treatment was associated with reduced mortality in patients with high platelet reactivity only (p for interaction: 0.02 for CV-death [<0.01 for all-cause mortality]. CONCLUSIONS Cardiovascular mortality risk in patients with High- and Low platelet reactivity is equivalent to the presence of coronary artery disease. Targeted glucose control, improved kidney function and lower inflammation are associated with reduced mortality risk, however independent of platelet reactivity. In contrast, only in patients with High-platelet reactivity was Aspirin treatment associated with lower mortality.
Collapse
|
42
|
Abstract
Antiplatelet therapy is used in the treatment of patients with acute coronary syndromes, stroke, and those undergoing percutaneous coronary intervention. Clopidogrel is the most widely used antiplatelet P2Y12 inhibitor in clinical practice. Genetic variation in CYP2C19 may influence its enzymatic activity, resulting in individuals who are carriers of loss-of-function CYP2C19 alleles and thus have reduced active clopidogrel metabolites, high on-treatment platelet reactivity, and increased ischemic risk. Prospective studies have examined the utility of CYP2C19 genetic testing to guide antiplatelet therapy, and more recently published meta-analyses suggest that pharmacogenetics represents a key treatment strategy to individualize antiplatelet therapy. Rapid genetic tests, including bedside genotyping platforms that are validated and have high reproducibility, are available to guide selection of P2Y12 inhibitors in clinical practice. The aim of this review is to provide an overview of the background and rationale for the role of a guided antiplatelet approach to enhance patient care.
Collapse
Affiliation(s)
- Matteo Castrichini
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA;
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan, USA
| | - Naveen Pereira
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA;
| |
Collapse
|
43
|
Pennings GJ, Reddel CJ, Chen VM, Gnanenthiran SR, Kritharides L. Perspective: Collagen induced platelet activation via the GPVI receptor as a primary target of colchicine in cardiovascular disease. Front Cardiovasc Med 2023; 9:1104744. [PMID: 36741844 PMCID: PMC9892722 DOI: 10.3389/fcvm.2022.1104744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Colchicine has been demonstrated to reduce cardiovascular death, myocardial infarction (MI), ischemic stroke, and ischemia-driven coronary revascularization in people with coronary artery disease (CAD). These reductions were observed even in patients already taking antiplatelet therapy. As well as having anti-inflammatory effects, colchicine demonstrates antiplatelet effects. We propose that colchicine's antiplatelet effects primarily target collagen-induced platelet activation via the collagen receptor, glycoprotein (GP)VI, which is critical for arterial thrombosis formation. In settings such as stroke and MI, GPVI signaling is upregulated. We have demonstrated in vitro that therapeutic concentrations of colchicine lead to a decrease in collagen-induced platelet aggregation and alter GPVI signaling. Clinical studies of colchicine given for 6 months lead to a significant reduction in serum GPVI levels in CAD patients, which may ameliorate thrombotic risk. Future evaluation of the effects of colchicine in clinical trials should include assessment of its effects on collagen-mediated platelet activation, and consideration be given to quantifying the contribution of such antiplatelet effects additional to the known anti-inflammatory effects of colchicine.
Collapse
Affiliation(s)
- Gabrielle J. Pennings
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia
| | - Caroline J. Reddel
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia
| | - Vivien M. Chen
- Department of Haematology, Concord Repatriation General Hospital, Concord, NSW, Australia,Platelet, Thrombosis Research Laboratory, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia
| | - Sonali R. Gnanenthiran
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia,The George Institute for Global Health, University of New South Wales, Newtown, NSW, Australia
| | - Leonard Kritharides
- Vascular Biology Group, ANZAC Research Institute, The University of Sydney, Concord, NSW, Australia,Department of Cardiology, Concord Repatriation General Hospital, Concord, NSW, Australia,*Correspondence: Leonard Kritharides ✉
| |
Collapse
|
44
|
Urban L, Ingrid Š, Žolková J, Ján S, Bolek T, Samoš M. High On-Treatment Platelet Reactivity in Patients Undergoing Complex Percutaneous Coronary Interventions. Clin Appl Thromb Hemost 2023; 29:10760296231199089. [PMID: 37697705 PMCID: PMC10498693 DOI: 10.1177/10760296231199089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/03/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023] Open
Abstract
Patient response to P2Y12 inhibitor therapy is heterogeneous, and those with high on-treatment platelet reactivity (HTPR) are at an increased risk of thrombotic complications. The aim of our study was to determine whether selecting a high-risk patient group of individuals after complex percutaneous coronary intervention (PCI) would show the clinical benefit of HTPR testing for preventing thrombotic complications. Blood samples of patients after complex PCI were acquired 1 day and 1 month after the intervention. The samples were tested using vasodilator-stimulated phosphoprotein phosphorylation (VASP-P) and platelet function assay (PFA). The occurrence of clinically significant stent thrombosis with repeated revascularization of the target vessel was observed over a 1-year period. One day after PCI, 37% of patients had HTPR as established by VASP-P. One month after PCI, the percentage of patients with HTPR decreased to 30.9%. According to PFA, 1 day after PCI, 33.3% of patients had HTPR. This percentage declined to 19.8% after 1 month. All measurements identified a significantly higher proportion of HTPR in patients on clopidogrel compared to ticagrelor and prasugrel. Two cases of early stent thrombosis and 1 case of late stent thrombosis were identified. Further study of adenosine diphosphate receptor blocker on-treatment response in patients undergoing complex PCI is necessary.
Collapse
Affiliation(s)
- Lukáš Urban
- Department of Internal Medicine I., Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
- Department of Cardiology, University Hospital Martin, Martin, Slovakia
| | - Škorňová Ingrid
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
| | - Jana Žolková
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
| | - Staško Ján
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
| | - Tomáš Bolek
- Department of Internal Medicine I., Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
| | - Matej Samoš
- Department of Internal Medicine I., Jessenius Faculty of Medicine, Comenius University, University Hospital Martin, Martin, Slovakia
| |
Collapse
|
45
|
Siasos G, Tsigkou V, Bletsa E, Stampouloglou PK, Oikonomou E, Kalogeras K, Katsarou O, Pesiridis T, Vavuranakis M, Tousoulis D. Antithrombotic Treatment in Coronary Artery Disease. Curr Pharm Des 2023; 29:2764-2779. [PMID: 37644793 DOI: 10.2174/1381612829666230830105750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/16/2023] [Accepted: 07/20/2023] [Indexed: 08/31/2023]
Abstract
Coronary artery disease exhibits growing mortality and morbidity worldwide despite the advances in pharmacotherapy and coronary intervention. Coronary artery disease is classified in the acute coronary syndromes and chronic coronary syndromes according to the most recent guidelines of the European Society of Cardiology. Antithrombotic treatment is the cornerstone of therapy in coronary artery disease due to the involvement of atherothrombosis in the pathophysiology of the disease. Administration of antiplatelet agents, anticoagulants and fibrinolytics reduce ischemic risk, which is amplified early post-acute coronary syndromes or post percutaneous coronary intervention; though, antithrombotic treatment increases the risk for bleeding. The balance between ischemic and bleeding risk is difficult to achieve and is affected by patient characteristics, procedural parameters, concomitant medications and pharmacologic characteristics of the antithrombotic agents. Several pharmacological strategies have been evaluated in patients with coronary artery disease, such as the effectiveness and safety of antithrombotic agents, optimal dual antiplatelet treatment schemes and duration, aspirin de-escalation strategies of dual antiplatelet regimens, dual inhibition pathway strategies as well as triple antithrombotic therapy. Future studies are needed in order to investigate the gaps in our knowledge, including special populations.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
- Cardiovascular Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Vasiliki Tsigkou
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Evanthia Bletsa
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Konstantinos Kalogeras
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Ourania Katsarou
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Theodoros Pesiridis
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, School of Medicine, Sotiria General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, School of Medicine, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
46
|
Buntsma N, van der Pol E, Nieuwland R, Gąsecka A. Extracellular Vesicles in Coronary Artery Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:81-103. [PMID: 37603274 DOI: 10.1007/978-981-99-1443-2_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Coronary artery disease (CAD) is the leading cause of death and disability worldwide. Despite recent progress in the diagnosis and treatment of CAD, evidence gaps remain, including pathogenesis, the most efficient diagnostic strategy, prognosis of individual patients, monitoring of therapy, and novel therapeutic strategies. These gaps could all be filled by developing novel, minimally invasive, blood-based biomarkers. Potentially, extracellular vesicles (EVs) could fill such gaps. EVs are lipid membrane particles released from cells into blood and other body fluids. Because the concentration, composition, and functions of EVs change during disease, and because all cell types involved in the development and progression of CAD release EVs, currently available guidelines potentially enable reliable and reproducible measurements of EVs in clinical trials, offering a wide range of opportunities. In this chapter, we provide an overview of the associations reported between EVs and CAD, including (1) the role of EVs in CAD pathogenesis, (2) EVs as biomarkers to diagnose CAD, predict prognosis, and monitor therapy in individual patients, and (3) EVs as new therapeutic targets and/or drug delivery vehicles. In addition, we summarize the challenges encountered in EV isolation and detection, and the lack of standardization, which has hampered real clinical applications of EVs. Since most conclusions are based on animal models and single-center studies, the knowledge and insights into the roles and opportunities of EVs as biomarkers in CAD are still changing, and therefore, the content of this chapter should be seen as a snapshot in time rather than a final and complete compendium of knowledge on EVs in CAD.
Collapse
Affiliation(s)
- Naomi Buntsma
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Vesicle Observation Centre, and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Edwin van der Pol
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- 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
| | - Aleksandra Gąsecka
- Vesicle Observation Centre, and Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
47
|
Zhong PY, Deng JP, Zhao JH, Peng L, Liu T, Wang HY. Guided vs. conventional anti-platelet therapy for patients with acute coronary syndrome: A meta-analysis of randomized controlled trials. Front Cardiovasc Med 2023; 10:1079332. [PMID: 37025677 PMCID: PMC10070678 DOI: 10.3389/fcvm.2023.1079332] [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: 10/25/2022] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Background Whether guided antiplatelet therapy in patients with acute coronary syndrome (ACS) is effective in improving net clinical benefits compared with conventional antiplatelet therapy remains controversial. Therefore, we assessed the safety and efficacy of guided antiplatelet therapy in patients with ACS and undergoing percutaneous coronary intervention. Method We searched PubMed, EMBASE, and Cochrane Library databases to select the relevant randomized controlled trials comparing the guided and conventional antiplatelet therapy in patients with ACS. The primary and safety outcomes are major adverse cardiovascular events (MACE) and major bleeding, respectively. The efficacy outcomes included myocardial infarction, stent thrombosis, all-cause death, and cardiovascular death. We selected the relative risk (RR) and 95% confidence intervals (CIs) as effect size and calculated it using the Review Manager software. In addition, we evaluated the final results by trial sequential analysis (registered by PROSPERO, CRD 42020210912). Results We selected seven randomized controlled trials and included 8,451 patients in this meta-analysis. Guided antiplatelet therapy can significantly reduce the risk of MACE (RR 0.64, 95% CI 0.54-0.76, P < 0.00001), myocardial infarction (RR 0.62, 95% CI 0.49-0.79, P = 0.0001), all-cause death (RR 0.61, 95% CI 0.44-0.85, P = 0.003), and cardiovascular death (RR 0.66, 0.49-0.90, P = 0.009). In addition, there is no significant difference between the two groups in stent thrombosis (RR 0.67, 95% CI 0.44-1.03, P = 0.07) and major bleeding (RR 0.86, 95% CI 0.65-1.13, P = 0.27). The subgroup analysis showed that the guided group based on genotype tests could bring benefits in MACE and myocardial infarction. Conclusions The guided antiplatelet therapy is not only associated with a comparable risk of bleeding but also with a lower risk of MACE, myocardial infarction, all-cause death, cardiovascular death, and stent thrombosis than the conventional strategy in patients with ACS.
Collapse
Affiliation(s)
- Peng-Yu Zhong
- Department of Cardiology, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Jian-Ping Deng
- Department of Cardiology, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Jing-Hong Zhao
- Department of Cardiology, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Lei Peng
- Department of Urology, The Second Hospital of Lanzhou University Medical School, Lanzhou, China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Tao Liu
- Department of Cardiology, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- Correspondence: Tao Liu Hao-Yu Wang
| | - Hao-Yu Wang
- Department of Cardiology, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
- Correspondence: Tao Liu Hao-Yu Wang
| |
Collapse
|
48
|
Davidson S. Monitoring of Antiplatelet Therapy. Methods Mol Biol 2023; 2663:381-402. [PMID: 37204725 DOI: 10.1007/978-1-0716-3175-1_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In the late 1990s, the antithrombotic antiplatelet agent, clopidogrel, a P2Y12 inhibitor, was introduced. Around the same time, there was an increase in a number of new methods to measure platelet function (e.g., PFA-100 in 1995), and this has continued. It became evident that not all patients responded to clopidogrel in the same way and that some patients had a relative "resistance" to therapy, termed "high on-treatment platelet reactivity." This then led to some publications to advocate platelet function testing being used for patients on antiplatelet therapy. Platelet function testing was also suggested for use in patients awaiting cardiac surgery after stopping their antiplatelet therapy as a way of balancing thrombotic risk pre-surgery and bleeding risk perioperatively. This chapter will discuss some of the commonly used platelet function tests used in these settings, particularly those that are sometimes referred to as point-of-care tests or that require minimal laboratory sample manipulation. The latest guidance and recommendations for platelet function testing will be discussed following several clinical trials looking at the usefulness of platelet function testing in these clinical settings.
Collapse
Affiliation(s)
- Simon Davidson
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
49
|
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) 2022; 24:e24-e35. [PMID: 36729588 DOI: 10.2459/jcm.0000000000001406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [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 P2Y12 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 P2Y12 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
|
50
|
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
|