1
|
Hong SJ, Kim BK. Potent P2Y 12 Inhibitor Monotherapy for Acute Coronary Syndrome. Circ J 2025; 89:272-280. [PMID: 37940598 DOI: 10.1253/circj.cj-23-0750] [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] [Indexed: 11/10/2023]
Abstract
Dual antiplatelet therapy (DAPT), consisting of aspirin and a P2Y12inhibitor, has been the principal antiplatelet therapy after drug-eluting stent (DES) implantation in patients with acute coronary syndrome (ACS) and chronic coronary disease. Particularly in patients with ACS, which presents a higher ischemic risk than chronic coronary artery disease, DAPT for up to 12 months is the recommended standard treatment. However, to decrease bleeding events related to the potency of P2Y12inhibitors and a prolonged duration of DAPT, recent studies have suggested P2Y12inhibitor monotherapy after short-term DAPT (1-3 months), which decreased the bleeding risk without an increased ischemic risk. In this article, we discuss the evidence related to the efficacy of a P2Y12inhibitor as single-antiplatelet therapy after short-term DAPT compared with standard DAPT, with a focus on patients with ACS treated with DES.
Collapse
Affiliation(s)
- Sung-Jin Hong
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine
| | - Byeong-Keuk Kim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine
| |
Collapse
|
2
|
Buchan E, Harbi MH, Rickard JJS, Thomas M, Goldberg Oppenheimer P. Advanced biomolecular spectroscopic profiling of cardiovascular disease macromolecular markers: SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP for rapid in-situ detection and monitoring. Int J Biol Macromol 2025; 284:138115. [PMID: 39608533 DOI: 10.1016/j.ijbiomac.2024.138115] [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: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Cardiovascular disease (CVD) remains a major global health concern and a leading cause of morbidity and mortality worldwide. Early-diagnosis and prompt medical attention are crucial in managing and reducing overall impact on health-and-wellbeing, necessitating the development of innovative diagnostics, which transcend traditional methodologies. Raman spectroscopy uniquely provides molecular fingerprinting and structural information, offering insights into biochemical composition. Integration of Raman spectroscopy with advanced machine learning is established as a powerful clinical adjunct for point-of-care detection of CVDs. A non-invasive, label-free spectroscopic platform coupled with neural network algorithm, 'SKiNET' has been developed to accurately detect the biomolecular changes within plasma of CVD versus healthy cohorts, enabling rapid diagnosis and longer-term monitoring, where the real-time capabilities provide dynamic assessment of progression, aligning treatment strategies with evolving states. CVD has been detected and classified via SKiNET with 88.6 %-accuracy, 92.9 %-specificity and 85.1 %-sensitivity and with 83.8 %-accuracy. The hybrid RS-SKiNET bio-molecularly specific detection signposted a comprehensive panel of CVD-indicative biomarkers, including SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP, offering important insights into disease mechanisms and risk-stratification. This multidimensional technique holds potential for improved patient-and-healthcare management for CVDs, laying the platform toward high-throughput biomolecular profiling of CVD-indicative macromolecular biomarkers, particularly vital for widespread point-of-care diagnostics and monitoring.
Collapse
Affiliation(s)
- Emma Buchan
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Maan H Harbi
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jonathan J S Rickard
- Department of Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Healthcare Technologies Institute, Institute of Translational Medicine, Mindelsohn Way, Birmingham B15 2TH, UK.
| |
Collapse
|
3
|
Xi Y, Min Z, Liu M, Lin X, Yuan ZH. Role and recent progress of P2Y12 receptor in cancer development. Purinergic Signal 2024:10.1007/s11302-024-10027-w. [PMID: 38874752 DOI: 10.1007/s11302-024-10027-w] [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: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
P2Y12 receptor (P2Y12R) is an adenosine-activated G protein-coupled receptor (GPCR) that plays a central role in platelet function, hemostasis, and thrombosis. P2Y12R activation can promote platelet aggregation and adhesion to cancer cells, promote tumor angiogenesis, and affect the tumor immune microenvironment (TIME) and tumor drug resistance, which is conducive to the progression of cancers. Meanwhile, P2Y12R inhibitors can inhibit this effect, suggesting that P2Y12R may be a potential therapeutic target for cancer. P2Y12R is involved in cancer development and metastasis, while P2Y12R inhibitors are effective in inhibiting cancer. However, a new study suggests that long-term use of P2Y12R inhibitors may increase the risk of cancer and the mechanism remains to be explored. In this paper, we reviewed the structural and functional characteristics of P2Y12R and its role in cancer. We explored the role of P2Y12R inhibitors in different tumors and the latest advances by summarizing the basic and clinical studies on the effects of P2Y12R inhibitors on tumors.
Collapse
Affiliation(s)
- Yanni Xi
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Zhenya Min
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Mianxue Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Xueqin Lin
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
| | - Zhao-Hua Yuan
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China.
| |
Collapse
|
4
|
Kaufmann CC, Muthspiel M, Lunzer L, Pogran E, Zweiker D, Burger AL, Wojta J, Huber K. Antiplatelet Therapy and Anticoagulation before, during, and after Acute Coronary Syndrome. J Clin Med 2024; 13:2313. [PMID: 38673585 PMCID: PMC11051414 DOI: 10.3390/jcm13082313] [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: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Acute coronary syndrome (ACS) remains a major challenge in clinical practice, requiring rapid and effective antithrombotic treatment to mitigate adverse ischemic events while minimizing the risk of bleeding. In recent years, results from several clinical trials addressing this issue through various approaches have substantially improved the treatment landscape for patients presenting with ACS. The emergence of new, potent P2Y12 inhibitors has significantly enhanced thrombotic risk reduction and different strategies for de-escalating and shortening dual antiplatelet therapy (DAPT) have demonstrated promising outcomes in reducing bleeding rates. Furthermore, data from ongoing trials focusing on novel therapeutic agents and investigating alternative treatment strategies to optimize outcomes for ACS patients are expected in the next few years. In this review, we summarize the current knowledge and emphasize the critical role of individualized treatment approaches tailored to patient-specific risk factors and individual clinical scenarios.
Collapse
Affiliation(s)
- Christoph C. Kaufmann
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
- Faculty of Medicine, Sigmund Freud University, A-1020 Vienna, Austria
| | - Marie Muthspiel
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
- Faculty of Medicine, Sigmund Freud University, A-1020 Vienna, Austria
| | - Laura Lunzer
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
| | - Edita Pogran
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
- Faculty of Medicine, Sigmund Freud University, A-1020 Vienna, Austria
| | - David Zweiker
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
| | - Achim Leo Burger
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
- Faculty of Medicine, Sigmund Freud University, A-1020 Vienna, Austria
| | - Johann Wojta
- Ludwig Boltzmann Institute for Cardiovascular Research, A-1090 Vienna, Austria;
- Core Facilities, Medical University of Vienna, A-1090 Vienna, Austria
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Kurt Huber
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), A-1160 Vienna, Austria; (M.M.); (L.L.); (E.P.); (D.Z.); (A.L.B.); (K.H.)
- Faculty of Medicine, Sigmund Freud University, A-1020 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, A-1090 Vienna, Austria;
| |
Collapse
|
5
|
Ortega-Paz L, Franchi F, Rollini F, Galli M, Been L, Ghanem G, Shalhoub A, Ossi T, Rivas A, Zhou X, Pineda AM, Suryadevara S, Soffer D, Zenni MM, Jennings LK, Angiolillo DJ. Switching from Dual Antiplatelet Therapy with Aspirin Plus a P2Y12 Inhibitor to Dual Pathway Inhibition with Aspirin Plus Vascular-Dose Rivaroxaban: The Switching Anti-Platelet and Anti-Coagulant Therapy (SWAP-AC) Study. Thromb Haemost 2024; 124:263-273. [PMID: 37224883 DOI: 10.1055/a-2098-6639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
BACKGROUND To date, there are no data on switching to dual pathway inhibition (DPI) patients who have completed a guideline-recommended dual antiplatelet therapy (DAPT) regimen. OBJECTIVES To assess the feasibility of switching from DAPT to DPI and to compare the pharmacodynamic (PD) profiles of these treatments. METHODS This was a prospective, randomized, PD study conducted in 90 patients with chronic coronary syndrome (CCS) on DAPT with aspirin (81 mg/qd) plus a P2Y12 inhibitor (clopidogrel [75 mg/qd; n = 30], ticagrelor [90 mg/bid; n = 30], or prasugrel [10 mg/qd; n = 30]). Patients in each cohort were randomized to maintain DAPT or switch to DPI (aspirin 81 mg/qd plus rivaroxaban 2.5 mg/bid). PD assessments included: VerifyNow P2Y12 reaction units; light transmittance aggregometry following stimuli with adenosine diphosphate (ADP), tissue factor (TF), and a combination of collagen, ADP, and TF (maximum platelet aggregation %); thrombin generation (TG). Assays were performed at baseline and 30 days postrandomization. RESULTS Switching from DAPT to DPI occurred without major side effects. DAPT was associated with enhanced P2Y12 inhibition, while DPI with reduced TG. Platelet-mediated global thrombogenicity (primary endpoint) showed no differences between DAPT and DPI in the ticagrelor (14.5% [0.0-63.0] vs. 20.0% [0.0-70.0]; p = 0.477) and prasugrel (20.0% [0.0-66.0] vs. 4.0% [0.0-70.0]; p = 0.482), but not clopidogrel (27.0% [0.0-68.0] vs. 53.0% [0.0-81.0]; p = 0.011), cohorts. CONCLUSION In patients with CCS, switching from different DAPT regimens to DPI was feasible, showing enhanced P2Y12 inhibition with DAPT and reduced TG with DPI, with no differences in platelet-mediated global thrombogenicity between DPI and ticagrelor- and prasugrel-, but not clopidogrel-, based DAPT. CLINICAL TRIAL REGISTRATION http://www. CLINICALTRIALS gov Unique Identifier: NCT04006288.
Collapse
Affiliation(s)
- Luis Ortega-Paz
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Mattia Galli
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
- Departmet of Cardiology, Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Latonya Been
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Ghussan Ghanem
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Awss Shalhoub
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Tiffany Ossi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Andrea Rivas
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Xuan Zhou
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Andres M Pineda
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Siva Suryadevara
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Daniel Soffer
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Martin M Zenni
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | | | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| |
Collapse
|
6
|
Parker WAE, Storey RF. The role of platelet P2Y 12 receptors in inflammation. Br J Pharmacol 2024; 181:515-531. [PMID: 37771103 DOI: 10.1111/bph.16256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Inflammation is a complex pathophysiological process underlying many clinical conditions. Platelets contribute to the thrombo-inflammatory response. Platelet P2Y12 receptors amplify platelet activation, potentiating platelet aggregation, degranulation and shape change. The contents of platelet alpha granules, in particular, act directly on leucocytes, including mediating platelet-leucocyte aggregation and activation via platelet P-selectin. Much evidence for the role of platelet P2Y12 receptors in inflammation comes from studies using antagonists of these receptors, such as the thienopyridines clopidogrel and prasugrel, and the cyclopentyltriazolopyrimidine ticagrelor, in animal and human experimental models. These suggest that antagonism of P2Y12 receptors decreases markers of inflammation with some evidence that this reduces incidence of adverse clinical sequelae during inflammatory conditions. Interpretation is complicated by pleiotropic effects such as those of the thienopyridines on circulating leucocyte numbers and of ticagrelor on adenosine reuptake. The available evidence suggests that P2Y12 receptors are prominent mediators of inflammation and P2Y12 receptor antagonism as a potentially powerful strategy in a broad range of inflammatory conditions. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- William A E Parker
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Robert F Storey
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
7
|
Pan D, Ladds G, Rahman KM, Pitchford SC. Exploring bias in platelet P2Y 1 signalling: Host defence versus haemostasis. Br J Pharmacol 2024; 181:580-592. [PMID: 37442808 PMCID: PMC10952580 DOI: 10.1111/bph.16191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are necessary for maintaining haemostasis. Separately, platelets are important for the propagation of inflammation during the host immune response against infection. The activation of platelets also causes inappropriate inflammation in various disease pathologies, often in the absence of changes to haemostasis. The separate functions of platelets during inflammation compared with haemostasis are therefore varied and this will be reflected in distinct pathways of activation. The activation of platelets by the nucleotide adenosine diphosphate (ADP) acting on P2Y1 and P2Y12 receptors is important for the development of platelet thrombi during haemostasis. However, P2Y1 stimulation of platelets is also important during the inflammatory response and paradoxically in scenarios where no changes to haemostasis and platelet aggregation occur. In these events, Rho-GTPase signalling, rather than the canonical phospholipase Cβ (PLCβ) signalling pathway, is necessary. We describe our current understanding of these differences, reflecting on recent advances in knowledge of P2Y1 structure, and the possibility of biased agonism occurring from activation via other endogenous nucleotides compared with ADP. Knowledge arising from these different pathways of P2Y1 stimulation of platelets during inflammation compared with haemostasis may help therapeutic control of platelet function during inflammation or infection, while preserving essential haemostasis. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Dingxin Pan
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Graham Ladds
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | - Khondaker Miraz Rahman
- Chemical Biology Group, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Simon C. Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
8
|
Alenazy FO, Harbi MH, Kavanagh DP, Price J, Brady P, Hargreaves O, Harrison P, Slater A, Tiwari A, Nicolson PLR, Connolly DL, Kirchhof P, Kalia N, Jandrot-Perrus M, Mangin PH, Watson SP, Thomas MR. Amplified inhibition of atherosclerotic plaque-induced platelet activation by glenzocimab with dual antiplatelet therapy. J Thromb Haemost 2023; 21:3236-3251. [PMID: 37541591 DOI: 10.1016/j.jtha.2023.07.018] [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: 01/22/2023] [Revised: 06/23/2023] [Accepted: 07/16/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Aspirin and platelet P2Y12 inhibitors, such as ticagrelor, suboptimally inhibit microvascular thrombosis during ST-elevation myocardial infarction. Glycoprotein (GP) IIb/IIIa inhibitors may further inhibit this but cause excessive bleeding. OBJECTIVES We investigated whether combination of glenzocimab, a GPVI inhibitor, with aspirin and ticagrelor provides additional antithrombotic effects, as GPVI has a critical role in atherothrombosis but minimal involvement in hemostasis. METHODS We investigated the effects of glenzocimab (monoclonal antibody Fab fragment) using blood from healthy donors and patients with acute coronary syndrome treated with aspirin and ticagrelor. Platelets were stimulated with multiple agonists, including atherosclerotic plaque, from patients undergoing carotid endarterectomy. RESULTS Aspirin and ticagrelor partially inhibited atherosclerotic plaque-induced platelet aggregation by 48% compared with control (34 ± 3 vs 65 ± 4 U; P < .001). Plaque-induced platelet aggregation, adhesion, secretion, and activation were critically dependent on GPVI activation. Glenzocimab alone reduced plaque-induced aggregation by 75% compared with control (16 ± 4 vs 65 ± 4 U; P < .001) and by >95% when combined with aspirin and ticagrelor (3 ± 1 vs 65 ± 4 U; P < .001). Glenzocimab reduced platelet aggregation, adhesion, and thrombin generation when added to blood of aspirin- and ticagrelor-treated patients with acute coronary syndrome. Glenzocimab shared several antithrombotic effects with the GPIIb/IIIa inhibitor eptifibatide with less effect on general hemostasis assessed by rotational thromboelastometry. In a murine intravital model of ST-elevation myocardial infarction, genetic depletion of GPVI reduced microvascular thrombosis. CONCLUSION Addition of glenzocimab to aspirin and ticagrelor enhances platelet inhibition via multiple mechanisms of atherothrombosis. Compared with a GPIIb/IIIa inhibitor, glenzocimab shares multiple antithrombotic effects, with less inhibition of mechanisms involved in general hemostasis.
Collapse
Affiliation(s)
- Fawaz O Alenazy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Maan H Harbi
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Dean P Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joshua Price
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Brady
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Oscar Hargreaves
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alok Tiwari
- Department of Vascular Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Derek L Connolly
- Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, University Heart and Vascular Center (UKE) Hamburg, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Neena Kalia
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Pierre H Mangin
- UMR_S1255, INSERM, Etablissement Francais du Sang-Alsace, Strasbourg, France
| | - Steve P Watson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom; Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom.
| |
Collapse
|
9
|
Parker WAE, Angiolillo DJ, Rollini F, Franchi F, Bonaca MP, Bhatt DL, Steg PG, Orme RC, Thomas MR, Judge HM, Sabatine MS, Storey RF. Influence of body weight and body mass index on the chronic pharmacokinetic and pharmacodynamic responses to clinically available doses of ticagrelor in patients with chronic coronary syndromes. Vascul Pharmacol 2023; 149:107145. [PMID: 36720377 DOI: 10.1016/j.vph.2023.107145] [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: 10/31/2022] [Accepted: 01/25/2023] [Indexed: 01/30/2023]
Abstract
Ticagrelor has multiple indications, including for some patients with chronic coronary syndromes (CCS) at high risk of ischaemic events. Body mass can potentially affect pharmacodynamics (PD) and pharmacokinetics (PK). We investigated the influence of body mass (range 53-172 kg, 20.8-46.9 kg/m2) on PD/PK in 221 CCS patients receiving ticagrelor 60 mg or 90 mg twice-daily (BD) during two randomised-controlled trials. Correlations between body weight (BW) or body mass index (BMI) and PD/PK measurements obtained during maintenance treatment at trough ('pre-dose') and peak effect ('post-dose') were assessed. BW and BMI correlated with P2Y12 reactivity units at pre-dose (e.g. BW:R = 0.26, p = 0.008) but not post-dose timepoints. BW affected ticagrelor active metabolite (TAM) levels (e.g. 60 mg BD, post-dose:R = -0.39, p < 0.0001) and there was evidence of an inverse power law relationship between BW and TAM-to-ticagrelor ratio. PK with ticagrelor 60 mg correlated significantly with BMI. BW and BMI did not affect the chance of high platelet reactivity, which remained very low across the whole cohort. There was no difference in PRU between the two doses of ticagrelor within each weight or BMI group. Body mass has modest effects on the PK/PD response to ticagrelor in patients with CCS but currently-used regimens appear adequate across the range of BW/BMI studied.
Collapse
Affiliation(s)
- William A E Parker
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Fabiana Rollini
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Francesco Franchi
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Marc P Bonaca
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deepak L Bhatt
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM UMR1148/LVTS, AP-HP, Hôpital Bichat, Paris, France
| | - Rachel C Orme
- Department of Cardiology, St John of God Hospital, Southwest Health Campus, Bunbury, WA, Australia
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Heather M Judge
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Marc S Sabatine
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert F Storey
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| |
Collapse
|
10
|
Abstract
In addition to the key role in hemostasis and thrombosis, platelets have also been wildly acknowledged as immune regulatory cells and involving in the pathogenesis of inflammation-related diseases. Since purine receptor P2Y12 plays a crucial role in platelet activation, P2Y12 antagonists such as clopidogrel, prasugrel, and ticagrelor have been widely used in cardiovascular diseases worldwide in recent decades due to their potent antiplatelet and antithrombotic effects. Meanwhile, the role of P2Y12 in inflammatory diseases has also been extensively studied. Relatively, there are few studies on the regulation of P2Y12. This review first summarizes the various roles of P2Y12 in the process of platelet activation, as well as downstream effects and signaling pathways; then introduces the effects of P2Y12 in inflammatory diseases such as sepsis, atherosclerosis, cancer, autoimmune diseases, and asthma; and finally reviews the current researches on P2Y12 regulation.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Infectious Diseases, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China
| | | | - Xia Cao
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China.
| |
Collapse
|
11
|
Wadowski PP, Panzer B, Józkowicz A, Kopp CW, Gremmel T, Panzer S, Koppensteiner R. Microvascular Thrombosis as a Critical Factor in Severe COVID-19. Int J Mol Sci 2023; 24:2492. [PMID: 36768817 PMCID: PMC9916726 DOI: 10.3390/ijms24032492] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Platelet-endothelial interactions have a critical role in microcirculatory function, which maintains tissue homeostasis. The subtle equilibrium between platelets and the vessel wall is disturbed by the coronavirus disease 2019 (COVID-19), which affects all three components of Virchow's triad (endothelial injury, stasis and a hypercoagulable state). Endotheliitis, vasculitis, glycocalyx degradation, alterations in blood flow and viscosity, neutrophil extracellular trap formation and microparticle shedding are only few pathomechanisms contributing to endothelial damage and microthrombosis resulting in capillary plugging and tissue ischemia. In the following opinion paper, we discuss major pathological processes leading to microvascular endothelial activation and thrombosis formation as a possible major adverse factor driving the deterioration of patient disease course in severe COVID-19.
Collapse
Affiliation(s)
- Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Benjamin Panzer
- Department of Cardiology, Wilhelminenspital, 1160 Vienna, Austria
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Gremmel
- Institute of Antithrombotic Therapy in Cardiovascular Disease, Karl Landsteiner Society, 3100 St. Pölten, Austria
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
| | - Simon Panzer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
12
|
P2Y 12 inhibitor monotherapy in patients undergoing percutaneous coronary intervention. Nat Rev Cardiol 2022; 19:829-844. [PMID: 35697777 DOI: 10.1038/s41569-022-00725-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 12/15/2022]
Abstract
For 20 years, dual antiplatelet therapy (DAPT), consisting of the combination of aspirin and a platelet P2Y12 receptor inhibitor, has been the gold standard of antithrombotic pharmacology after percutaneous coronary intervention (PCI). In the past 5 years, several investigations have challenged this paradigm by testing the efficacy and safety of P2Y12 inhibitor monotherapy (that is, without aspirin) following a short course of DAPT. Collectively, these studies suggested a reduction in the risk of major bleeding and no significant increase in thrombotic or ischaemic events compared with guideline-recommended DAPT. Current recommendations are evolving to inform clinical practice on the ideal candidates for P2Y12 inhibitor monotherapy after PCI. Generalizing the results of studies of P2Y12 inhibitor monotherapy requires a thorough understanding of their design, populations, interventions, comparators and results. In this Review, we provide an up-to-date overview on the use of P2Y12 inhibitor monotherapy after PCI, including supporting pharmacodynamic and clinical evidence, practical recommendations and future directions.
Collapse
|
13
|
Pons V, Garcia C, Tidten-Luksch N, Mac Sweeney A, Caroff E, Galés C, Riederer MA. Inverse agonist efficacy of selatogrel blunts constitutive P2Y12 receptor signaling by inducing the inactive receptor conformation. Biochem Pharmacol 2022; 206:115291. [DOI: 10.1016/j.bcp.2022.115291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/02/2022]
|
14
|
Bulla SC, Badial PR, Bulla C. Canine Cancer Cells Activate Platelets via the Platelet P2Y12 Receptor. J Comp Pathol 2022; 192:41-49. [PMID: 35305713 DOI: 10.1016/j.jcpa.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/09/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022]
Abstract
In addition to their well-known functions in haemostasis, anucleated platelets have a critical role in cancer biology. Many human and non-human cancer types can directly interact with and activate platelets, promoting cancer malignancy and progression. Although naturally occurring canine neoplastic diseases mimic the biologically complex conditions of human cancers more closely than laboratory-bred mice, studies evaluating the relationship between cancer cells and platelets in dogs are scarce, and the effects of tumour cells on platelets in these animals are unknown. To evaluate whether cancer cells could activate canine platelets, we assessed the response of platelet-rich plasma to cultured canine cancer cells using light transmittance aggregometry. Similar to human and murine cancer cell research, we demonstrated that both canine osteosarcoma and mammary carcinoma cells activated canine platelets in vitro, resulting in platelet aggregation. The degree of aggregation was most pronounced at a cancer cell to platelet ratio of 1:200 for most cell lines. Mechanistic studies revealed that the platelet adenosine diphosphate (ADP) receptor P2Y12 is essential for canine platelet aggregation induced by canine cancer. ADP receptor blockage on platelets inhibited >50% of cancer cell-induced maximum platelet aggregation in all cell lines evaluated. As in other species, our results suggest that canine cancers may activate canine platelets in vivo. This mechanism is likely relevant for the biology and progression of cancer in the dog.
Collapse
Affiliation(s)
- Sandra C Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Peres R Badial
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Camilo Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA.
| |
Collapse
|
15
|
Li L, Liu S, Tan J, Wei L, Wu D, Gao S, Weng Y, Chen J. Recent advance in treatment of atherosclerosis: Key targets and plaque-positioned delivery strategies. J Tissue Eng 2022; 13:20417314221088509. [PMID: 35356091 PMCID: PMC8958685 DOI: 10.1177/20417314221088509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory disease of vascular wall, is a progressive pathophysiological process with lipids oxidation/depositing initiation and innate/adaptive immune responses. The coordination of multi systems covering oxidative stress, dysfunctional endothelium, diseased lipid uptake, cell apoptosis, thrombotic and pro-inflammatory responding as well as switched SMCs contributes to plaque growth. In this circumstance, inevitably, targeting these processes is considered to be effective for treating atherosclerosis. Arriving, retention and working of payload candidates mediated by targets in lesion direct ultimate therapeutic outcomes. Accumulating a series of scientific studies and clinical practice in the past decades, lesion homing delivery strategies including stent/balloon/nanoparticle-based transportation worked as the potent promotor to ensure a therapeutic effect. The objective of this review is to achieve a very brief summary about the effective therapeutic methods cooperating specifical targets and positioning-delivery strategies in atherosclerosis for better outcomes.
Collapse
Affiliation(s)
- Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Lai Wei
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Dimeng Wu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Shuai Gao
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| |
Collapse
|
16
|
Butt JH, Fosbøl EL, Gerds TA, Iversen K, Bundgaard H, Bruun NE, Larsen AR, Petersen A, Andersen PS, Skov RL, Østergaard L, Havers-Borgersen E, Gislason GH, Torp-Pedersen C, Køber L, Olesen JB. Ticagrelor and the risk of Staphylococcus aureus bacteraemia and other infections. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2022; 8:13-19. [PMID: 32750138 DOI: 10.1093/ehjcvp/pvaa099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
AIMS To investigate the 1-year risks of Staphylococcus aureus bacteraemia (SAB), sepsis, and pneumonia in patients who underwent percutaneous coronary intervention and were treated with ticagrelor vs. clopidogrel. METHODS AND RESULTS In this nationwide observational cohort study, 26 606 patients who underwent urgent or emergent percutaneous coronary intervention (January 2011-December 2017) and initiated treatment with ticagrelor [N = 20 073 (75.5%); median age 64 years (25th-75th percentile 55-72 years); 74.8% men] or clopidogrel [N = 6533 (24.5%); median age 68 years (25th-75th percentile 58-77 years); 70.2% men] were identified using Danish nationwide registries. The 1-year standardized absolute risks of outcomes was calculated based on cause-specific Cox regression models, and average treatment effects between treatment groups were obtained as standardized differences in absolute 1-year risks. The absolute 1-year risk of SAB was 0.10% [95% confidence interval (CI), 0.05-0.15%] in the ticagrelor group and 0.29% (95% CI, 0.17-0.42%) in the clopidogrel group. Compared with clopidogrel, treatment with ticagrelor was associated with a significantly lower absolute 1-year risk of SAB [absolute risk difference -0.19% (95% CI, -0.32% to -0.05%), P value 0.006]. Likewise, treatment with ticagrelor was associated with a significantly lower absolute 1-year risk of sepsis [0.99% (95% CI, 0.83-1.14%) vs. 1.49% (95% CI, 1.17-1.80%); absolute risk difference -0.50% (95% CI, -0.86% to -0.14%), P value 0.007] and pneumonia [3.13% (95% CI, 2.86-3.39%) vs. 4.56% (95% CI, 4.03-5.08%); absolute risk difference -1.43% (95% CI, -2.03% to -0.82%), P value < 0.001] compared with clopidogrel. CONCLUSION Treatment with ticagrelor was associated with a significantly lower 1-year risk of SAB, sepsis, and pneumonia compared with clopidogrel.
Collapse
Affiliation(s)
- Jawad H Butt
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil L Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas A Gerds
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark.,The Danish Heart Foundation, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels Eske Bruun
- Department of Cardiology, Zealand University Hospital, Roskilde, Denmark.,Institute of Clinical Medicine, Copenhagen University, Copenhagen, Denmark.,Clinical Institute, Aalborg University, Aalborg, Denmark
| | | | | | | | | | - Lauge Østergaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eva Havers-Borgersen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gunnar H Gislason
- The Danish Heart Foundation, Copenhagen, Denmark.,Department of Cardiology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Christian Torp-Pedersen
- Department of Cardiology and Clinical Investigation, Nordsjællands Hospital, Hillerød, Denmark.,Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jonas B Olesen
- Department of Cardiology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| |
Collapse
|
17
|
Elevated platelet-leukocyte complexes are associated with, but dispensable for myocardial ischemia-reperfusion injury. Basic Res Cardiol 2022; 117:61. [PMID: 36383299 PMCID: PMC9668925 DOI: 10.1007/s00395-022-00970-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/18/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
AIMS P-selectin is an activatable adhesion molecule on platelets promoting platelet aggregation, and platelet-leukocyte complex (PLC) formation. Increased numbers of PLC are circulating in the blood of patients shortly after acute myocardial infarction and predict adverse outcomes. These correlations led to speculations about whether PLC may represent novel therapeutic targets. We therefore set out to elucidate the pathomechanistic relevance of PLC in myocardial ischemia and reperfusion injury. METHODS AND RESULTS By generating P-selectin deficient bone marrow chimeric mice, the post-myocardial infarction surge in PLC numbers in blood was prevented. Yet, intravital microscopy, flow cytometry and immunohistochemical staining, echocardiography, and gene expression profiling showed unequivocally that leukocyte adhesion to the vessel wall, leukocyte infiltration, and myocardial damage post-infarction were not altered in response to the lack in PLC. CONCLUSION We conclude that myocardial infarction associated sterile inflammation triggers PLC formation, reminiscent of conserved immunothrombotic responses, but without PLC influencing myocardial ischemia and reperfusion injury in return. Our experimental data do not support a therapeutic concept of selectively targeting PLC formation in myocardial infarction.
Collapse
|
18
|
Shrestha B, Katz D, Kelley J, Menzies D, Hong MK. Cangrelor in STEMI as a bridge to CABG- a mini-case series. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 13:100122. [PMID: 38560057 PMCID: PMC10978215 DOI: 10.1016/j.ahjo.2022.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 04/04/2024]
Abstract
Background Patients with STEMI receive dual antiplatelet therapy as soon as possible with aspirin and a P2Y12 receptor antagonist prior to PCI. A fraction of these patients may have multi-vessel disease needing emergent CABG surgery. The choice of a P2Y12 receptor antagonist plays a role in the timing of CABG surgery as it poses a bleeding risk until it is completely eliminated from the system. Oral P2Y12 receptor antagonists have a long duration of platelet inhibition which is difficult to reverse. Cangrelor is an intravenous P2Y12 receptor antagonist with a short half-life and rapid cessation of its effect after discontinuation. Methods Three patients who presented to our emergency department with STEMI were started on cangrelor infusion prior to cardiac catheterization instead of other P2Y12 receptor antagonists like clopidogrel or ticagrelor. The study received ethical approval as it is part of the current standard of care for STEMI patients. Results All three patients were found to have multi-vessel disease during coronary angiography requiring CABG surgery. As cangrelor was used in these patients they were able to have their surgery within 24-48 h. Intravenous cangrelor was stopped about an hour before surgery. No bleeding complications occurred and all three patients made a speedy recovery in the ICU. Conclusion Cangrelor is a potent P2Y12 receptor antagonist which can be used in patients presenting with STEMI as one of the two anti-platelet agents along with aspirin without any dilemma that it would cause a delay in CABG surgery if the patients need one.
Collapse
Affiliation(s)
- Bishesh Shrestha
- Department of Cardiology, Bassett Medical Center, Cooperstown, NY, United States of America
| | - Daniel Katz
- Department of Cardiology, Bassett Medical Center, Cooperstown, NY, United States of America
| | - John Kelley
- Department of Cardiothoracic Surgery, Bassett Medical Center, Cooperstown, NY, United States of America
| | - Dhananjai Menzies
- Department of Cardiology, Bassett Medical Center, Cooperstown, NY, United States of America
| | - Mun K. Hong
- Department of Cardiology, Bassett Medical Center, Cooperstown, NY, United States of America
| |
Collapse
|
19
|
Schrottmaier WC, Mussbacher M, Salzmann M, Kral-Pointner JB, Assinger A. PI3K Isoform Signalling in Platelets. Curr Top Microbiol Immunol 2022; 436:255-285. [PMID: 36243848 DOI: 10.1007/978-3-031-06566-8_11] [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] [Indexed: 06/16/2023]
Abstract
Platelets are unique anucleated blood cells that constantly patrol the vasculature to seal and prevent injuries in a process termed haemostasis. Thereby they rapidly adhere to the subendothelial matrix and recruit further platelets, resulting in platelet aggregates. Apart from their central role in haemostasis, they also kept some of their features inherited by their evolutionary ancestor-the haemocyte, which was also involved in immune defences. Together with leukocytes, platelets fight pathogenic invaders and guide many immune processes. In addition, they rely on several signalling pathways which are also relevant to immune cells. Among these, one of the central signalling hubs is the PI3K pathway. Signalling processes in platelets are unique as they lack a nucleus and therefore transcriptional regulation is absent. As a result, PI3K subclasses fulfil distinct roles in platelets compared to other cells. In contrast to leukocytes, the central PI3K subclass in platelet signalling is PI3K class Iβ, which underlines the uniqueness of this cell type and opens new ways for potential platelet-specific pharmacologic inhibition. An overview of platelet function and signalling with emphasis on PI3K subclasses and their respective inhibitors is given in this chapter.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marion Mussbacher
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA
| | - Manuel Salzmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Julia B Kral-Pointner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
20
|
Crescence L, Kramberg M, Baumann M, Rey M, Roux S, Panicot-Dubois L, Dubois C, Riederer MA. The P2Y12 Receptor Antagonist Selatogrel Dissolves Preformed Platelet Thrombi In Vivo. J Clin Med 2021; 10:jcm10225349. [PMID: 34830631 PMCID: PMC8619398 DOI: 10.3390/jcm10225349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022] Open
Abstract
Selatogrel, a potent and reversible antagonist of the P2Y12 receptor, inhibited FeCl3-induced thrombosis in rats. Here, we report the anti-thrombotic effect of selatogrel after subcutaneous applications in guinea pigs and mice. Selatogrel inhibited platelet function only 10 min after subcutaneous application in mice. In addition, in a modified Folts thrombosis model in guinea pigs, selatogrel prevented a decrease in blood-flow, indicative of the inhibition of ongoing thrombosis, approximately 10 min after subcutaneous injection. Selatogrel fully normalised blood flow; therefore, we speculate that it may not only prevent, but also dissolve, platelet thrombi. Thrombus dissolution was investigated using real-time intravital microscopy in mice. The infusion of selatogrel during ongoing platelet thrombus formation stopped growth and induced the dissolution of the preformed platelet thrombus. In addition, platelet-rich thrombi were given 30 min to consolidate in vivo. The infusion of selatogrel dissolved the preformed and consolidated platelet thrombi. Dissolution was limited to the disintegration of the occluding part of the platelet thrombi, leaving small mural platelet aggregates to seal the blood vessel. Therefore, our experiments uncovered a novel advantage of selatogrel: the dissolution of pre-formed thrombi without the disintegration of haemostatic seals, suggesting a bipartite benefit of the early application of selatogrel in patients with acute thrombosis.
Collapse
Affiliation(s)
- Lydie Crescence
- Aix Marseille Université, INSERM 1263, INRAE 1260, C2VN, 27 Boulevard Jean Moulin, 13385 Marseille, France; (L.C.); (L.P.-D.); (C.D.)
| | - Markus Kramberg
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland; (M.K.); (M.B.); (M.R.); (S.R.)
| | - Martine Baumann
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland; (M.K.); (M.B.); (M.R.); (S.R.)
| | - Markus Rey
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland; (M.K.); (M.B.); (M.R.); (S.R.)
| | - Sebastien Roux
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland; (M.K.); (M.B.); (M.R.); (S.R.)
| | - Laurence Panicot-Dubois
- Aix Marseille Université, INSERM 1263, INRAE 1260, C2VN, 27 Boulevard Jean Moulin, 13385 Marseille, France; (L.C.); (L.P.-D.); (C.D.)
| | - Christophe Dubois
- Aix Marseille Université, INSERM 1263, INRAE 1260, C2VN, 27 Boulevard Jean Moulin, 13385 Marseille, France; (L.C.); (L.P.-D.); (C.D.)
| | - Markus A. Riederer
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland; (M.K.); (M.B.); (M.R.); (S.R.)
- Correspondence: ; Tel.: +41-588-440-885
| |
Collapse
|
21
|
Barrett TJ, Cornwell M, Myndzar K, Rolling CC, Xia Y, Drenkova K, Biebuyck A, Fields AT, Tawil M, Luttrell-Williams E, Yuriditsky E, Smith G, Cotzia P, Neal MD, Kornblith LZ, Pittaluga S, Rapkiewicz AV, Burgess HM, Mohr I, Stapleford KA, Voora D, Ruggles K, Hochman J, Berger JS. Platelets amplify endotheliopathy in COVID-19. SCIENCE ADVANCES 2021; 7:eabh2434. [PMID: 34516880 PMCID: PMC8442885 DOI: 10.1126/sciadv.abh2434] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/19/2021] [Indexed: 05/08/2023]
Abstract
Given the evidence for a hyperactive platelet phenotype in COVID-19, we investigated effector cell properties of COVID-19 platelets on endothelial cells (ECs). Integration of EC and platelet RNA sequencing revealed that platelet-released factors in COVID-19 promote an inflammatory hypercoagulable endotheliopathy. We identified S100A8 and S100A9 as transcripts enriched in COVID-19 platelets and were induced by megakaryocyte infection with SARS-CoV-2. Consistent with increased gene expression, the heterodimer protein product of S100A8/A9, myeloid-related protein (MRP) 8/14, was released to a greater extent by platelets from COVID-19 patients relative to controls. We demonstrate that platelet-derived MRP8/14 activates ECs, promotes an inflammatory hypercoagulable phenotype, and is a significant contributor to poor clinical outcomes in COVID-19 patients. Last, we present evidence that targeting platelet P2Y12 represents a promising candidate to reduce proinflammatory platelet-endothelial interactions. Together, these findings demonstrate a previously unappreciated role for platelets and their activation-induced endotheliopathy in COVID-19.
Collapse
Affiliation(s)
- Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - MacIntosh Cornwell
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Khrystyna Myndzar
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Yuhe Xia
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Kamelia Drenkova
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Antoine Biebuyck
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Alexander T. Fields
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael Tawil
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Eugene Yuriditsky
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Grace Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paolo Cotzia
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Center for Biospecimen Research, New York University Grossman School of Medicine, New York, NY, USA
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lucy Z. Kornblith
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Stefania Pittaluga
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Amy V. Rapkiewicz
- Department of Pathology, NYU Langone Long Island Hospital, New York University Langone Health, Mineola, NY, USA
| | - Hannah M. Burgess
- Department of Microbiology, New York University Langone Health, New York, NY, USA
| | - Ian Mohr
- Department of Microbiology, New York University Langone Health, New York, NY, USA
| | | | - Deepak Voora
- Department of Medicine, Duke Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Kelly Ruggles
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Judith Hochman
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Department of Surgery, New York University Langone Health, New York, NY, USA
| |
Collapse
|
22
|
Adali MK, Buber I, Kilic O, Turkoz A, Yilmaz S. Ticagrelor improves systemic immune-inflammation index in acute coronary syndrome patients. Acta Cardiol 2021; 77:632-638. [PMID: 34493170 DOI: 10.1080/00015385.2021.1973770] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Inflammation plays a critical role in atherosclerosis. This study examines the effects of ticagrelor and clopidogrel on inflammatory parameters, obtained from complete blood count (CBC) and biochemical measurements, in patients with acute coronary syndrome. METHODS AND RESULTS One hundred acute coronary syndrome (ACS) patients were included in the study and grouped according to clopidogrel (n = 50) or ticagrelor (n = 50) usage as an anti-aggregant (with acetylsalicylic acid). All patients underwent percutaneous coronary intervention. On admission, at third- and sixth-month after ACS, white blood cell (WBC) count, neutrophil-to-lymphocyte ratio (NLR), monocyte-to-high-density lipoprotein ratio (MHR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) were calculated from the CBC and biochemical measurements. NLR, PLR, and SII were significantly lower in the ticagrelor group (p < 0.001, at 3rd and 6th month). Also, MHR was lower in the ticagrelor group (p < 0.05). Conversely, WBC count was higher in the ticagrelor group (p < 0.001). CONCLUSIONS NLR, MHR, PLR, and SII levels were lower in ACS patients treating with ticagrelor. Ticagrelor may improve these inflammatory parameters in percutaneous coronary intervention (PCI)-treated ACS patients compared to clopidogrel.
Collapse
Affiliation(s)
- Mehmet Koray Adali
- Department of Cardiology, Pamukkale University, School of Medicine, Denizli, Turkey
| | - Ipek Buber
- Department of Cardiology, Pamukkale University, School of Medicine, Denizli, Turkey
| | - Oguz Kilic
- Department of Cardiology, Simav Doc Dr. Ismail Karakuyu State Hospital, Kutahya, Turkey
| | - Anil Turkoz
- Department of Cardiology, Pamukkale University, School of Medicine, Denizli, Turkey
| | - Samet Yilmaz
- Department of Cardiology, Pamukkale University, School of Medicine, Denizli, Turkey
| |
Collapse
|
23
|
The Molecular Aspects of Disturbed Platelet Activation through ADP/P2Y 12 Pathway in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22126572. [PMID: 34207429 PMCID: PMC8234174 DOI: 10.3390/ijms22126572] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies confirm a high risk of ischemic events in secondary-progressive multiple sclerosis (SP MS) patients, directly associated with an increased level of pro-thrombotic activity of platelets. Our work aimed to verify potential molecular abnormalities of the platelet P2Y12 receptor expression and functionality as a cause of an increased risk of thromboembolism observed in the course of MS. We have demonstrated an enhanced platelet reactivity in response to adenosine diphosphate (ADP) in SP MS relative to controls. We have also shown an increased mRNA expression for the P2RY12 gene in both platelets and megakaryocytes, as well as enhanced density of these receptors on the platelet surface. We postulate that one of the reasons for the elevated risk of ischemic events observed in MS may be a genetically or phenotypically reinforced expression of the platelet P2Y12 receptor. In order to analyze the effect of the PAR1 (protease activated receptor type 1) signaling pathway on the expression level of P2Y12, we also analyzed the correlation parameters between P2Y12 expression and the markers of platelet activation in MS induced by selective PAR1 agonist (thrombin receptor activating peptide-6, TRAP-6). Identifying the molecular base responsible for the enlarged pro-thrombotic activity of platelets in SP MS could contribute to the implementation of prevention and targeted treatment, reducing the development of cardiovascular disorders in the course of the disease.
Collapse
|
24
|
Zhang L, Li Z, Ye X, Chen Z, Chen ZS. Mechanisms of thrombosis and research progress on targeted antithrombotic drugs. Drug Discov Today 2021; 26:2282-2302. [PMID: 33895314 DOI: 10.1016/j.drudis.2021.04.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/26/2022]
Abstract
Globally, the incidence of thromboembolic diseases has increased in recent years, accompanied by an increase in patient mortality. Currently, several targeting delivery strategies have been developed to treat thromboembolic diseases. In this review, we discuss the mechanisms of thrombolysis and current anticoagulant drugs, particularly those with targeting capability, highlighting advances in the accurate treatment of thrombolysis with fewer adverse effects. Such approaches include magnetic drug-loading systems combined with molecular imaging to recanalize blood vessels and systems based on chimeric Arg-Gly-Asp (RGD) sequences that can target platelet glycoprotein receptor. With such progress in targeted antithrombotic drugs, targeted thrombolysis treatment shows significant potential benefit for patients.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Li
- Fujian Cancer Hospital, Fujian Provincial Cancer Hospital of Fujian Medical University, Fuzhou 350014, China
| | - Xianren Ye
- Fujian Cancer Hospital, Fujian Provincial Cancer Hospital of Fujian Medical University, Fuzhou 350014, China.
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY 11439, USA.
| |
Collapse
|
25
|
Gąsecka A, Borovac JA, Guerreiro RA, Giustozzi M, Parker W, Caldeira D, Chiva-Blanch G. Thrombotic Complications in Patients with COVID-19: Pathophysiological Mechanisms, Diagnosis, and Treatment. Cardiovasc Drugs Ther 2021; 35:215-229. [PMID: 33074525 PMCID: PMC7569200 DOI: 10.1007/s10557-020-07084-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Emerging evidence points to an association between severe clinical presentation of COVID-19 and increased risk of thromboembolism. One-third of patients hospitalized due to severe COVID-19 develops macrovascular thrombotic complications, including venous thromboembolism, myocardial injury/infarction and stroke. Concurrently, the autopsy series indicate multiorgan damage pattern consistent with microvascular injury. PROPHYLAXIS, DIAGNOSIS AND TREATMENT COVID-19 associated coagulopathy has distinct features, including markedly elevated D-dimers concentration with nearly normal activated partial thromboplastin time, prothrombin time and platelet count. The diagnosis may be challenging due to overlapping features between pulmonary embolism and severe COVID-19 disease, such as dyspnoea, high concentration of D-dimers, right ventricle with dysfunction or enlargement, and acute respiratory distress syndrome. Both macro- and microvascular complications are associated with an increased risk of in-hospital mortality. Therefore, early recognition of coagulation abnormalities among hospitalized COVID-19 patients are critical measures to identify patients with poor prognosis, guide antithrombotic prophylaxis or treatment, and improve patients' clinical outcomes. RECOMMENDATIONS FOR CLINICIANS Most of the guidelines and consensus documents published on behalf of professional societies focused on thrombosis and hemostasis advocate the use of anticoagulants in all patients hospitalized with COVID-19, as well as 2-6 weeks post hospital discharge in the absence of contraindications. However, since there is no guidance for deciding the intensity and duration of anticoagulation, the decision-making process should be made in individual-case basis. CONCLUSIONS Here, we review the mechanistic relationships between inflammation and thrombosis, discuss the macrovascular and microvascular complications and summarize the prophylaxis, diagnosis and treatment of thromboembolism in patients affected by COVID-19.
Collapse
Affiliation(s)
- Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland.
| | - Josip A Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split, Croatia
| | | | - Michela Giustozzi
- Internal Vascular and Emergency Medicine and Stroke Unit, University of Perugia, Perugia, Italy
| | - William Parker
- Cardiovascular Research Unit, University of Sheffield, Sheffield, UK
| | - Daniel Caldeira
- Centro Cardiovascular da Universidade de Lisboa (CCUL), Faculdade de Medicina, Univerisdade de Lisboa, Lisbon, Portugal
- Cardiology Department, Hospital Universitário de Santa Maria (CHULN), Avenida Professor Egas Moniz, 1649-028, Lisbon, Portugal
| | - Gemma Chiva-Blanch
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Madrid, Spain
| |
Collapse
|
26
|
Özdemir ZC, Düzenli Kar Y, Bör Ö. Whole Blood miR-210, miR-122, miR-223 Expression Levels and Their Relationship With Iron Status Parameters and Hypercoagulability Indices in Children With Iron Deficiency Anemia. J Pediatr Hematol Oncol 2021; 43:e328-e335. [PMID: 33710119 DOI: 10.1097/mph.0000000000002127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022]
Abstract
MicroRNAs have the potential to regulate systemic and cellular iron homeostasis at multiple points. In iron deficiency anemia (IDA), hypoxia, platelet reactivity, and potentially microRNAs play a role in the development of hypercoagulability. A total of 57 children diagnosed with IDA between October 2016 and October 2017 and 48 healthy children were included in this cross-sectional study. Blood count parameters, serum iron, transferrin saturation, ferritin level, maximum clot firmness (MCF), and clot formation time index, which are indicators of hypercoagulability in rotational thromboelastometry test, of the IDA and control groups obtained in our previous study were recorded. miR-210, miR-122, and miR-223 levels were analyzed. There was no difference in the miR-210, miR-122, and miR-223 levels between the IDA and control groups. Patients with hemoglobin (Hb) <8 g/dL had higher miR-210 levels than patients with Hb>8 g/dL (P<0.05). There was a negative correlation between miR-210 and Hb and ferritin levels, a positive correlation between miR-122 and ferritin levels, and a negative correlation between miR-223 and MCF index. In IDA, there is a close relationship between the severity of anemia and miR-210, and miR-210 expression is slightly increased in those with severe anemia. miR-210 and miR-122 collectively play a role in maintaining the iron balance. The correlation between miR-223, a platelet function regulator, and the MCF index, suggested that miR-223 has a role in the development of hypercoagulability in IDA.
Collapse
Affiliation(s)
- Zeynep C Özdemir
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | | | | |
Collapse
|
27
|
Lee SB, Ji HD, Lee IK, Kim KS, Lee J, Lee SW, Jeon YH. Visualization of platelet recruitment to tumor lesions using highly sensitive and stable radioiodine studded gold nanoprobes. J Mater Chem B 2021; 9:2931-2936. [PMID: 33885648 DOI: 10.1039/d0tb02265a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In vivo imaging of platelets will provide a better understanding of their critical roles in arterial cardiovascular disease, hemostasis, inflammation, and cancer. Here, we demonstrate the feasibility of using radioiodine studded gold nanoprobes (RIS-GNPs) as a platelet tracker for nuclear medicine imaging in tumor-bearing mice using positron emission tomography and computed tomography (PET/CT). Platelet labeling with RIS-GNPs did not alter the platelet functions, such as cellular proliferation and aggregation. PET/CT imaging clearly revealed the migration of platelets into tumor sites at 1 to 5 h post-transfer of RIS-GNP-labeled platelets, which was consistent with the biodistribution data. Our findings suggest that the imaging approach using RIS-GNPs makes it feasible to visualize the biological behavior of platelets in living organisms with cancer.
Collapse
Affiliation(s)
- Sang Bong Lee
- Vaccine Commercialization Center, Gyeongbuk Institute for Bio industry, 88 Saneopdanji-gil, pungsan-eup, Andong-si, Gyeongbuk, 33618, South Korea
| | | | | | | | | | | | | |
Collapse
|
28
|
Crescence L, Darbousset R, Caroff E, Hubler F, Riederer MA, Panicot-Dubois L, Dubois C. Selatogrel, a reversible P2Y12 receptor antagonist, has reduced off-target interference with haemostatic factors in a mouse thrombosis model. Thromb Res 2021; 200:133-140. [PMID: 33610885 DOI: 10.1016/j.thromres.2021.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Selatogrel is a reversible antagonist of the P2Y12 receptor. In rat thrombosis/haemostasis models, selatogrel was associated with lower blood loss than clopidogrel or ticagrelor at equivalent anti-thrombotic effect. MATERIAL AND METHODS We sought to elucidate the mechanism underlying the observed differences in blood loss, using real-time intravital microscopy in mouse. RESULTS Selatogrel, ticagrelor and clopidogrel dose-dependently inhibited laser-induced platelet thrombus formation. At maximal antithrombotic effect, only small mural platelets aggregates, corresponding to hemostatic seals, were present. The phenotype of these hemostatic seals was dependent on the type of P2Y12 receptor antagonist. In the presence of clopidogrel and ticagrelor, detachment of platelets from the hemostatic seals was increased, indicative of reduced stability. In contrast, in the presence of selatogrel, platelet detachment was not increased. Moreover, equivalent antithrombotic dosing regimens of ticagrelor and clopidogrel reduced laser-induced calcium mobilization in the endothelium, restricted neutrophil adhesion and subsequent fibrin formation and thus reduced fibrin-mediated stabilization of the hemostatic seals. The effects of ticagrelor were also observed in P2Y12 receptor deficient mice, indicating that the effects are off-target and independent of the P2Y12 receptor. In contrast, selatogrel did not interfere with these elements of haemostasis in wild-type or in P2Y12 receptor deficient mice. CONCLUSION In the presence of selatogrel the stability of hemostatic seals was unperturbed, translating to an improved blood loss profile. Our data suggest that the mechanism underlying the differences in blood loss profiles of P2Y12 receptor antagonists is by off-target interference with endothelial activation, neutrophil function and thus, fibrin-mediated stabilization of haemostatic seals.
Collapse
Affiliation(s)
- Lydie Crescence
- Aix Marseille Université, INSERM 1263, INRAE 1260 27 Boulevard Jean Moulin, Marseille, France
| | - Roxane Darbousset
- Aix Marseille Université, INSERM 1263, INRAE 1260 27 Boulevard Jean Moulin, Marseille, France
| | - Eva Caroff
- Idorsia Pharmaceuticals Ltd. DD Chemistry, Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Francis Hubler
- Idorsia Pharmaceuticals Ltd. DD Chemistry, Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland
| | - Markus A Riederer
- Idorsia Pharmaceuticals Ltd. DD Biology, Hegenheimermattweg 91, CH-4123 Allschwil, Switzerland.
| | - Laurence Panicot-Dubois
- Aix Marseille Université, INSERM 1263, INRAE 1260 27 Boulevard Jean Moulin, Marseille, France
| | - Christophe Dubois
- Aix Marseille Université, INSERM 1263, INRAE 1260 27 Boulevard Jean Moulin, Marseille, France
| |
Collapse
|
29
|
Baumann M, Lack B, Guillaumat I, Murphy MJ, Riederer MA. The potency of selatogrel, a reversible antagonist of the P2Y12 receptor, is affected by calcium concentration. Platelets 2021; 33:147-156. [PMID: 33427002 DOI: 10.1080/09537104.2020.1869711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Here, we report the in vitro characterization of the P2Y12 receptor antagonist selatogrel (ACT-246475). Binding studies with radiolabeled selatogrel demonstrated that selatogrel is a competitive antagonist of ADP binding to the P2Y12 receptor with a fast onset of action. Consequently, selatogrel was confirmed to be a potent inhibitor of P2Y12-mediated intra-platelet signaling and ADP-induced platelet activation. Characterization of selatogrel in platelet-rich plasma in vitro demonstrated that the mode of anti-coagulation affected the anti-platelet potency. Specifically, in platelet-rich plasma containing physiological calcium concentration (anticoagulated with a direct thrombin inhibitor), selatogrel achieved half-maximal inhibition of ADP-induced platelet aggregation at a 3-fold lower concentration than in conditions with low calcium concentration (anticoagulated with citrate). Furthermore, calcium-dependent reduction in selatogrel potency was observed in whole blood platelet aggregation using the VerifyNow™ system with a 3.7-fold potency loss in low calcium conditions. A comparable potency loss was also observed with the reversible P2Y12 receptor antagonists ticagrelor, cangrelor and elinogrel. Furthermore, receptor-binding experiments using radiolabeled selatogrel confirmed a 3-fold lowering of selatogrel binding affinity to the P2Y12 receptor in low calcium conditions. In conclusion, our data suggest that in low calcium conditions (i.e., citrate-anticoagulated blood), there is a risk of underestimating the potency of reversible P2Y12 receptor antagonists. To avoid overdosing, and a potential increase in bleeding risk, we propose that the ex vivo evaluation of reversible P2Y12 receptor antagonists should be performed with platelet assay systems containing physiological calcium concentration.
Collapse
Affiliation(s)
- Martine Baumann
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Benoît Lack
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Isabelle Guillaumat
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Mark J Murphy
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Markus A Riederer
- Drug Discovery Biology, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| |
Collapse
|
30
|
Hennigan BW, Good R, Adamson C, Parker WAE, Martin L, Anderson L, Campbell M, Serruys PW, Storey RF, Oldroyd KG. Recovery of platelet reactivity following cessation of either aspirin or ticagrelor in patients treated with dual antiplatelet therapy following percutaneous coronary intervention: a GLOBAL LEADERS substudy. Platelets 2020; 33:141-146. [PMID: 33356730 DOI: 10.1080/09537104.2020.1863937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cessation of one component of dual antiplatelet therapy (DAPT) following percutaneous coronary intervention (PCI) has been associated with increased risk of ischemic events but it is uncertain whether discontinuation of aspirin is preferable to discontinuation of the oral P2Y12 inhibitor. The GLOBAL LEADERS study compared two antiplatelet strategies following PCI, cessation of aspirin at 1 month with continued ticagrelor monotherapy for 23 months versus standard DAPT for 12 months followed by aspirin monotherapy for a further 12 months. We assessed recovery of platelet reactivity after withdrawal of either aspirin or ticagrelor at 1 month and 12 months, respectively, in this study. Platelet aggregation (PA) was assessed before cessation of DAPT ('baseline') and after 2, 7, and 14 days post-cessation using Multiplate whole-blood aggregometry with collagen, thrombin-receptor-activating peptide (TRAP), adenosine diphosphate (ADP) and arachidonic acid (AA) as agonists. Following cessation of aspirin at 1 month, there was marked recovery of PA induced by AA (baseline [mean ± SD]: 11.1 ± 7.4 U vs. 14 days: 64.9 ± 19.6 U, p < .0001) and collagen (37.4 ± 22.9 U vs. 79.8 ± 13.8 U, p < .0001), whereas PA induced by ADP (18.6 ± 6.6 vs. 69.1 ± 20.5, p < .0001) and collagen (34.4 ± 18.7 U vs. 43.0 ± 21.0, p = .0018) recovered following cessation of ticagrelor at 12 months. There were no significant changes in TRAP-induced PA in either group. In conclusion, cessation of either component of DAPT leads to substantial increase in platelet reactivity with differential effects on different pathways of platelet activation when aspirin or the P2Y12 inhibitor is stopped. Further work is required to determine which patients receive net benefit from long-term continuation of DAPT.
Collapse
Affiliation(s)
- Barry W Hennigan
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - Richard Good
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - Carly Adamson
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - William A E Parker
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Lynn Martin
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - Lynne Anderson
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - Michael Campbell
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| | - Patrick W Serruys
- Department of Cardiology, National University of Ireland, University Road, Galway, Ireland.,National Heart and Lung Institute, Imperial College London, London, UK
| | - Robert F Storey
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Keith G Oldroyd
- Department of Cardiology, Golden Jubilee National Hospital, Glasgow, UK
| |
Collapse
|
31
|
Anderson R, Theron AJ, Steel HC, Nel JG, Tintinger GR. ADP-Mediated Upregulation of Expression of CD62P on Human Platelets Is Critically Dependent on Co-Activation of P2Y1 and P2Y12 Receptors. Pharmaceuticals (Basel) 2020; 13:ph13120420. [PMID: 33255391 PMCID: PMC7760858 DOI: 10.3390/ph13120420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
This study probed the differential utilization of P2Y1 and P2Y12 receptors in mobilizing CD62P (P-selectin) from intracellular granules following activation of human platelets with adenosine 5′-diphosphate (ADP, 100 µmol·L−1) Platelet-rich plasma (PRP) was prepared from the blood of adult humans. CD62P was measured by flow cytometry following activation of PRP with ADP in the absence and presence of the selective antagonists of P2Y1 and P2Y12 receptors, MRS2500 and PSB0739 (both 0.155–10 µmol·L−1), respectively. Effects of the test agents on ADP-activated, CD62P-dependent formation of neutrophil:platelet (NP) aggregates were also measured by flow cytometry, while phosphatidylinositol 3-kinase (PI3K) activity was measured according to Akt1 phosphorylation in platelet lysates. Treatment with MRS2500 or PSB0739 at 10 µmol·L−1 almost completely attenuated (94.6% and 86% inhibition, respectively) ADP-activated expression of CD62P and also inhibited NP aggregate formation. To probe the mechanisms involved in P2Y1/P2Y12 receptor-mediated expression of CD62P, PRP was pre-treated with U73122 (phospholipase C (PLC) inhibitor), 2-aminoethoxy-diphenyl borate (2-APB, inositol triphosphate receptor antagonist), calmidazolium chloride (calmodulin inhibitor), or wortmannin (PI3K inhibitor). U73122, 2-APB, and wortmannin caused almost complete inhibition of ADP-activated expression of CD62P, while calmidazolium chloride caused statistically significant, partial inhibition. PSB0739, but not MRS2500, caused potent inhibition of PI3K-mediated phosphorylation of Akt1. Optimal mobilization of CD62P by ADP-stimulated platelets is critically dependent on the co-activation of platelet P2Y1 and P2Y12 receptors. P2Y12 receptor activation is the key event in activation of PI3K, while activation of the P2Y1 receptor appears to create a high cytosolic Ca2+ environment conducive to optimum PI3K activity.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
- Correspondence: ; Tel.: +27-12-318-2425; Fax: +27-12-323-0732
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Jan G. Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Tshwane Academic Division of the National Laboratory Health Service of South Africa, Pretoria 0001, South Africa
| | - Gregory R. Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
32
|
Affiliation(s)
- William Ae Parker
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.,Directorate of Cardiology and Cardiothoracic Surgery, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Robert F Storey
- Cardiovascular Research Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK .,Directorate of Cardiology and Cardiothoracic Surgery, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
33
|
Pan Y, Wangqin R, Li H, Wang Y, Meng X, Johnston SC, Simon T, Lin J, Zhao X, Liu L, Wang D, Wang Y. F2R Polymorphisms and Clopidogrel Efficacy and Safety in Patients With Minor Stroke or TIA. Neurology 2020; 96:e1-e9. [PMID: 33093222 DOI: 10.1212/wnl.0000000000011078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/06/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the association between protease-activated receptor-1 (PAR-1) gene F2R polymorphisms and efficacy of clopidogrel for minor stroke or TIA. METHODS Three single nucleotide polymorphisms (CYP2C19*2 [681G>A, rs4244285], CYP2C19*3 [636G>A, rs4986893], and F2R [IVSn-14 A/T, rs168753]) were genotyped among 2,924 patients randomized to clopidogrel plus aspirin (n = 1,461) or aspirin alone (n = 1,463). The primary efficacy outcome was new stroke (ischemic or hemorrhagic) and the safety outcome was any bleeding. RESULTS Overall, 859 (29.4%) were AA homozygotes, 1,479 (50.6%) were AT heterozygotes, and 586 (20.0%) were TT homozygotes for F2R IVSn-14 polymorphisms; 1,716 (58.7%) were carriers of at least 1 CYP2C19 loss-of-function allele (*2 or *3). Compared with aspirin alone, patients with clopidogrel-aspirin treatment had a low risk of new stroke in patients with AT genotype (7.6% vs 11.3%; hazard ratio [HR], 0.63; 95% confidence interval [CI], 0.44-0.89) and TT genotype (5.8% vs 11.6%; HR, 0.46; 95% CI, 0.25-0.82) but not in carriers of the AA genotype (10.8% vs 11.6%; HR, 0.95; 95% CI, 0.63-1.44) (p = 0.03 for interaction). The association between F2R IVSn-14 A/T polymorphism and clopidogrel response was present regardless of the carrier status of the CYP2C19 loss-of-function alleles. The F2R IVSn-14 genotypes were not associated with the risk of any bleeding for clopidogrel-aspirin treatment (p = 0.66 for interaction). CONCLUSIONS Among patients with minor ischemic stroke or TIA who were receiving clopidogrel and aspirin, those carrying the F2R IVSn-14 T allele had a lower rate of recurrent stroke than those who were not. CLINICALTRIALSGOV IDENTIFIER NCT00979589.
Collapse
Affiliation(s)
- Yuesong Pan
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Runqi Wangqin
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Hao Li
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Yilong Wang
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ.
| | - Xia Meng
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - S Claiborne Johnston
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Tabassome Simon
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Jinxi Lin
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Xingquan Zhao
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Liping Liu
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - David Wang
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ.
| | - Yongjun Wang
- From the Department of Neurology (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (Y.P., H.L., Y.W., X.M., J.L., X.Z., L.L., Y.W.), Beijing, China; Department of Neurology (R.W.), Duke University Medical Center, Durham, NC; Dell Medical School (S.C.J.), University of Texas at Austin; Department of Clinical Pharmacology and Clinical Research Platform of East of Paris (URCEST-CRC-CRB) (T.S.), Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine (APHP.SU); Sorbonne Université (T.S.); FACT (French Alliance for Cardiovascular Clinical Trials) (T.S.), Paris, France; and Department of Neurology (D.W.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ.
| |
Collapse
|
34
|
Cervo A, Ferrari F, Barchetti G, Quilici L, Piano M, Boccardi E, Pero G. Use of Cangrelor in Cervical and Intracranial Stenting for the Treatment of Acute Ischemic Stroke: A "Real Life" Single-Center Experience. AJNR Am J Neuroradiol 2020; 41:2094-2099. [PMID: 33033047 DOI: 10.3174/ajnr.a6785] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE In cases of large-vessel-occlusion strokes due to an underlying tandem internal carotid artery occlusion or intracranial atherosclerotic disease, concomitant stent placement may be needed. Immediate platelet inhibition is necessary, but to date, a standardized approach for antiplatelet inhibition in acute settings is still missing. Here we report our single-center experience about the safety and efficacy of periprocedural administration of cangrelor in patients with acute ischemic stroke due to intracranial or cervical artery occlusion undergoing stent placement. MATERIALS AND METHODS We retrospectively evaluated all cases of acute ischemic stroke that needed acute stent implantation and were treated with periprocedural administration of cangrelor between January 2019 and April 2020 at our institution. All patients who needed either extracranial or intracranial artery stent placement (in either the anterior or posterior circulation) were included. RESULTS We evaluated 38 patients in whom cangrelor was administered IV periprocedurally. Their mean age was 64 years (range, 26-85 years), with 25/38 male subjects and 13/38 female patients. In 26 patients (68.4%), a tandem occlusion was present and was treated with carotid artery stent placement, while 12 patients (31.6%) required an intracranial stent implantation. In 4 subjects (10.5%), an intracerebral hemorrhage occurred after the procedure. All patients in the series were alive 1 week after the procedure. CONCLUSIONS Although larger, multicentric randomized studies are strongly warranted, our results support the hypothesis of a possible role of cangrelor as a valuable therapeutic option in the management of platelet inhibition in acute ischemic stroke procedures after intra- or extracranial stent placement.
Collapse
Affiliation(s)
- A Cervo
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - F Ferrari
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Biology and Biotecnology (F.F.), University of Pavia, Pavia, Italy
| | - G Barchetti
- Department of Neuroradiology (G.B.), Sapienza University, Rome, Italy
| | - L Quilici
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - M Piano
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - E Boccardi
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - G Pero
- From the Department of Neuroradiology (A.C., F.F., L.Q., M.P., E.B., G.P.), Azienda Socio-Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
35
|
Tomaniak M, Chichareon P, Onuma Y, Deliargyris EN, Takahashi K, Kogame N, Modolo R, Chang CC, Rademaker-Havinga T, Storey RF, Dangas GD, Bhatt DL, Angiolillo DJ, Hamm C, Valgimigli M, Windecker S, Steg PG, Vranckx P, Serruys PW. Benefit and Risks of Aspirin in Addition to Ticagrelor in Acute Coronary Syndromes: A Post Hoc Analysis of the Randomized GLOBAL LEADERS Trial. JAMA Cardiol 2020; 4:1092-1101. [PMID: 31557763 DOI: 10.1001/jamacardio.2019.3355] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Importance The role of aspirin as part of antiplatelet regimens in acute coronary syndromes (ACS) needs to be clarified in the context of newer potent P2Y12 antagonists. Objective To evaluate the benefit and risks of aspirin in addition to ticagrelor among patients with ACS beyond 1 month after percutaneous coronary intervention (PCI). Design, Setting, and Participants This is a nonprespecified, post hoc analysis of GLOBAL LEADERS, a randomized, open-label superiority trial comparing 2 antiplatelet treatment strategies after PCI. The trial included 130 secondary/tertiary care hospitals in different countries, with 15 991 unselected patients with stable coronary artery disease or ACS undergoing PCI. Patients had outpatient visits at 1, 3, 6, 12, 18, and 24 months after index procedure. Interventions The experimental group received aspirin plus ticagrelor for 1 month followed by 23-month ticagrelor monotherapy; the reference group received aspirin plus either clopidogrel (stable coronary artery disease) or ticagrelor (ACS) for 12 months, followed by 12-month aspirin monotherapy. In this analysis, we examined the clinical outcomes occurring between 31 days and 365 days after randomization, specifically in patients with ACS who, within this time frame, were assigned to receive either ticagrelor alone or ticagrelor and aspirin. Main Outcomes and Measures The primary outcome was the composite of all-cause death or new Q-wave myocardial infarction. Results Of 15 968 participants, there were 7487 patients with ACS enrolled; 3750 patients were assigned to the experimental group and 3737 patients to the reference group. Between 31 and 365 days after randomization, the primary outcome occurred in 55 patients (1.5%) in the experimental group and in 75 patients (2.0%) in the reference group (hazard ratio [HR], 0.73; 95% CI, 0.51-1.03; P = .07); investigator-reported Bleeding Academic Research Consortium-defined bleeding type 3 or 5 occurred in 28 patients (0.8%) in the experimental group and in 54 patients (1.5%) in the reference arm (HR, 0.52; 95% CI, 0.33-0.81; P = .004). Conclusions and Relevance Between 1 month and 12 months after PCI in ACS, aspirin was associated with increased bleeding risk and appeared not to add to the benefit of ticagrelor on ischemic events. These findings should be interpreted as exploratory and hypothesis generating; however, they pave the way for further trials evaluating aspirin-free antiplatelet strategies after PCI. Trial Registration ClinicalTrials.gov identifier: NCT01813435.
Collapse
Affiliation(s)
- Mariusz Tomaniak
- Department of Cardiology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands.,First Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Ply Chichareon
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Yoshinobu Onuma
- Department of Cardiology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands.,Cardialysis Core Laboratories and Clinical Trial Management, Rotterdam, the Netherlands
| | | | | | - Norihiro Kogame
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rodrigo Modolo
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Internal Medicine, Cardiology Division, University of Campinas, Campinas, Brazil
| | - Chun Ching Chang
- Department of Cardiology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands
| | | | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, England
| | - George D Dangas
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deepak L Bhatt
- Department of Medicine, Brigham and Women's Hospital Heart and Vascular Center, Boston, Massachusetts
| | | | | | - Marco Valgimigli
- Department of Cardiology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Stephan Windecker
- Department of Cardiology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Philippe Gabriel Steg
- French Alliance for Cardiovascular Trials, Université Paris Diderot, Hôpital Bichat, Assistance Publique Hôpitaux de Paris, and INSERM U-1148, Paris, France
| | - Pascal Vranckx
- Department of Cardiology and Critical Care Medicine, Hartcentrum Hasselt, Jessa Ziekenhuis, Hasselt, Belgium
| | - Patrick W Serruys
- National Heart and Lung Institute, Imperial College London, London, England
| | | |
Collapse
|
36
|
Chiva-Blanch G, Sala-Vila A, Crespo J, Ros E, Estruch R, Badimon L. The Mediterranean diet decreases prothrombotic microvesicle release in asymptomatic individuals at high cardiovascular risk. Clin Nutr 2020; 39:3377-3384. [PMID: 32147198 DOI: 10.1016/j.clnu.2020.02.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Circulating microvesicles (cMV) are small phospholipid-rich vesicles that contribute to the atherothrombotic process, and are biomarkers of cardiovascular disease (CVD) burden and progression. Diet is a cornerstone for CVD prevention, but dietary effects on cMV shedding are poorly characterized. We aimed at assessing the long term effects of a Mediterranean diet compared to a low-fat diet (LFD) on MV shedding by cells of the blood and vascular compartments in patients at high cardiovascular risk treated as per guidelines. METHODS A total of 155 participants from the PREDIMED trial free of cardiovascular events after a mean follow-up of 5 years (n = 53 from the Mediterranean diet supplemented with extra-virgin olive oil -EVOO-; n = 49 from the Mediterranean diet supplemented with mixed nuts -Nuts-; and n = 53 from the LFD) were included in the study. At baseline and after one-year intervention, cMV were quantified and characterized by flow cytometry to identify their activated parental cell origin and prothrombotic potential by Annexin V (AV) binding. RESULTS After one year of dietary intervention, platelet-derived PAC-1+/AV+ and CD62P+/AV+ cMV concentrations were lower in the Nuts group compared with the LFD and EVOO interventions (P = 0.036 and 0.003, respectively). In addition, prothrombotic cMV carrying tissue factor (CD142+/AV+) and CD11a+/AV+ cMV derived from activated cells, were significantly lower in both Mediterranean diet (EVOO and Nuts) interventions compared to one year of LFD (P < 0.0001 and 0.028, respectively). SMAα+/AV- cMV were lower in the LFD compared to the Nuts group after one year of intervention (P = 0.038). CONCLUSIONS cMV are markers of cell activation and vascular injury that appear to be sensitive to dietary changes. Following a Mediterranean diet rich in EVOO or nuts is associated with lower cell activation towards a pro-atherothrombotic phenotype, suggesting a delay in the development of CV complications.
Collapse
Affiliation(s)
- Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau - IIB Sant Pau, Barcelona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Aleix Sala-Vila
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Lipid Clinic, Endocrinology and Nutrition Service, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau - IIB Sant Pau, Barcelona, Spain
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Lipid Clinic, Endocrinology and Nutrition Service, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Internal Medicine, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca Hospital Santa Creu i Sant Pau - IIB Sant Pau, Barcelona, Spain; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
37
|
The association of body mass index with long-term clinical outcomes after ticagrelor monotherapy following abbreviated dual antiplatelet therapy in patients undergoing percutaneous coronary intervention: a prespecified sub-analysis of the GLOBAL LEADERS Trial. Clin Res Cardiol 2020; 109:1125-1139. [PMID: 32006156 PMCID: PMC7449952 DOI: 10.1007/s00392-020-01604-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Background The efficacy of antiplatelet therapies following percutaneous coronary intervention (PCI) may be affected by body mass index (BMI). Methods and results This is a prespecified subgroup analysis of the GLOBAL LEADERS trial, a prospective, multicenter, open-label, randomized controlled trial in an all-comer population undergoing PCI, comparing the experimental strategy (23-month ticagrelor monotherapy following 1-month dual antiplatelet therapy [DAPT]) with a reference regimen (12-month aspirin monotherapy following 12-month DAPT). A total of 15,968 patients were stratified by baseline BMI with prespecified threshold of 27 kg/m2. Of those, 6973 (43.7%) patients with a BMI < 27 kg/m2 had a higher risk of all-cause mortality at 2 years than those with BMI ≥ 27 kg/m2 (adjusted HR 1.24, 95% CI 1.02–1.49). At 2 years, the rates of the primary endpoint (all-cause mortality or new Q-wave myocardial infarction) were similar between treatment strategies in either BMI group (pinteraction = 0.51). In acute coronary syndrome, however, the experimental strategy was associated with significant reduction of the primary endpoint compared to the reference strategy in patients with BMI < 27 kg/m2 (HR 0.69, 95% CI 0.51–0.94), but not in the ones with BMI ≥ 27 kg/m2 (pinteraction = 0.047). In chronic coronary syndrome, there was no between-group difference in the efficacy and safety of the two antiplatelet strategies. Conclusions Overall, BMI did not influence the treatment effect seen with ticagrelor monotherapy; however, a beneficial effect of ticagrelor monotherapy was seen in ACS patients with BMI < 27 kg/m2. Trial registration The trial has been registered with ClinicalTrials.gov, Number NCT01813435. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s00392-020-01604-1) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Abstract
Aspirin and P2Y12 inhibitors remain commonly prescribed antiplatelet drugs in the treatment of atherothrombotic conditions. Despite established benefits of dual antiplatelet therapy (DAPT) in the setting of acute coronary syndromes, there remains residual ischemic risk in this group and the problem of bleeding complications is an ongoing issue. DAPT with aspirin and ticagrelor has now been studied in other patient groups such as those with concurrent diabetes and stable coronary artery disease, and those undergoing elective percutaneous coronary intervention (PCI). Recent trials of ticagrelor monotherapy have suggested this may have benefits over standard-of-care in some settings, such as PCI, but not in others such as peripheral arterial disease or stroke. A novel subcutaneously administered P2Y12 inhibitor, selatogrel, has shown powerful, rapid and consistent effect in a phase 2 study. Aspirin dosing remains an area of investigation, particularly in the setting of DAPT. A novel regimen of very-low-dose twice-daily aspirin has hypothetical advantages in pharmacodynamic and pharmacokinetic effects, maintaining antiplatelet effect whilst reducing potentially harmful peak-trough variation.
Collapse
Affiliation(s)
- William A E Parker
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield , Sheffield, UK.,South Yorkshire Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust , Sheffield, UK
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield , Sheffield, UK.,South Yorkshire Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust , Sheffield, UK
| |
Collapse
|
39
|
Tomaniak M, Storey RF, Serruys PW. Aspirin-free antiplatelet regimens after PCI: when is it best to stop aspirin and who could ultimately benefit? EUROINTERVENTION 2020; 15:1125-1129. [DOI: 10.4244/eijy19m10_01] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
De-escalation of anti-platelet therapy in patients with acute coronary syndromes undergoing percutaneous coronary intervention: a narrative review. Chin Med J (Engl) 2019; 132:197-210. [PMID: 30614864 PMCID: PMC6365275 DOI: 10.1097/cm9.0000000000000047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 receptor inhibitor is the cornerstone of treatment in patients with acute coronary syndromes (ACS) and in those undergoing percutaneous coronary intervention (PCI). In current clinical situation, availability of different oral P2Y12 inhibitors (clopidogrel, prasugrel, and ticagrelor) has enabled physicians to switch among therapies owing to specific clinical scenarios. Although optimum time, loading dose and interval of transition between P2Y12 inhibitors is still controversial and needs further evidence, switching between oral inhibitors frequently occurs in clinical practice for several reasons. Data sources: This review was based on data in articles published in PubMed up to June 2018, with the following keywords “antiplatelet therapy”, “ACS”, “PCI”, “ticagrelor”, and “clopidogrel”. Study selection: Original articles and critical reviews on de-escalation strategy in ACS patients after PCI were selected. References of the retrieved articles were also screened to search for potentially relevant papers. Results: Safety concerns associated with switching between antiplatelet agents, has prompted the use of clopidogrel for patients with ACS especially after PCI as a de-escalation strategy. Practical considerations for de-escalating therapies in patients with ACS such as reducing dose of P2Y12 inhibitors or shortening duration of DAPT (followed by aspirin or P2Y12 receptor inhibitor monotherapy) as potential options are yet to be standardized and validated. Conclusions: Current review will provide an overview of the pharmacology of common P2Y12 inhibitors, definitions of de-escalation and different de-escalating strategies and its outcomes, along with possible direction to be explored in de-escalation.
Collapse
|
41
|
Yandrapalli S, Andries G, Gupta S, Dajani AR, Aronow WS. Investigational drugs for the treatment of acute myocardial infarction: focus on antiplatelet and anticoagulant agents. Expert Opin Investig Drugs 2018; 28:223-234. [PMID: 30580647 DOI: 10.1080/13543784.2019.1559814] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Srikanth Yandrapalli
- Division of Cardiology, Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla,
NY, USA
| | - Gabriela Andries
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla,
NY, USA
| | - Shashvat Gupta
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla,
NY, USA
| | - Abdel Rahman Dajani
- Department of Medicine, Norwalk Hospital affiliated to Yale University, Norwalk,
CT, USA
| | - Wilbert S. Aronow
- Division of Cardiology, Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla,
NY, USA
| |
Collapse
|
42
|
Abstract
Platelet-derived microvesicles (pMVs) are small, heterogeneous vesicles released from platelet membranes as a result of activation. These microvesicles possess a wide range of properties, including prothrombotic, proatherogenic, proinflammatory, immunomodulatory, and even anticoagulant activity. The elevated release of these microvesicles has been observed in various metabolic, inflammatory, thrombotic, and vascular diseases, including ischemic heart disease, stroke, hypertension, diabetes, and connective tissue disease. Modulation of both pMV generation and the expression of their surface molecules may have beneficial clinical implications and could become a novel therapeutic target. However, mechanisms by which pharmacological agents can modify pMV formation are elusive. The purpose of this review is to discuss the effects of drugs routinely used in primary and secondary prevention of vascular disease on the release of pMV and expression of their surface procoagulant and proinflammatory molecules.
Collapse
Affiliation(s)
- Justyna Rosińska
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
43
|
Capodanno D, Mehran R, Valgimigli M, Baber U, Windecker S, Vranckx P, Dangas G, Rollini F, Kimura T, Collet JP, Gibson CM, Steg PG, Lopes RD, Gwon HC, Storey RF, Franchi F, Bhatt DL, Serruys PW, Angiolillo DJ. Aspirin-free strategies in cardiovascular disease and cardioembolic stroke prevention. Nat Rev Cardiol 2018; 15:480-496. [DOI: 10.1038/s41569-018-0049-1] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
44
|
Parker WAE, Eriksson N, Becker RC, Voora D, Åkerblom A, Himmelmann A, James SK, Wallentin L, Storey RF. Equilibrative nucleoside transporter 1 gene polymorphisms and clinical outcomes following acute coronary syndromes: findings from the PLATelet inhibition and patient Outcomes (PLATO) study. Platelets 2018; 30:579-588. [DOI: 10.1080/09537104.2018.1478404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- William A. E. Parker
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Richard C. Becker
- Division of Cardiovascular Health and Disease, Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Deepak Voora
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - Axel Åkerblom
- Department of Medical Sciences, Cardiology and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | | | - Stefan K. James
- Department of Medical Sciences, Cardiology and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Robert F. Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | | |
Collapse
|
45
|
Massolt ET, Meima ME, Swagemakers SMA, Leeuwenburgh S, van den Hout-van Vroonhoven MCGM, Brigante G, Kam BLR, van der Spek PJ, van IJcken WFJ, Visser TJ, Peeters RP, Visser WE. Thyroid State Regulates Gene Expression in Human Whole Blood. J Clin Endocrinol Metab 2018; 103:169-178. [PMID: 29069456 DOI: 10.1210/jc.2017-01144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/16/2017] [Indexed: 02/03/2023]
Abstract
CONTEXT Despite the well-recognized clinical features resulting from insufficient or excessive thyroid hormone (TH) levels in humans, it is largely unknown which genes are regulated by TH in human tissues. OBJECTIVE To study the effect of TH on human gene expression profiles in whole blood, mainly consisting of T3 receptor (TR) α-expressing cells. METHODS We performed next-generation RNA sequencing on whole blood samples from eight athyroid patients (four females) on and after 4 weeks off levothyroxine replacement. Gene expression changes were analyzed through paired differential expression analysis and confirmed in a validation cohort. Weighted gene coexpression network analysis (WGCNA) was applied to identify thyroid state-related networks. RESULTS We detected 486 differentially expressed genes (fold-change >1.5; multiple testing corrected P value < 0.05), of which 76% were positively and 24% were negatively regulated. Gene ontology (GO) enrichment analysis revealed that three biological processes were significantly overrepresented, of which the process translational elongation showed the highest fold enrichment (7.3-fold, P = 1.8 × 10-6). WGCNA analysis independently identified various gene clusters that correlated with thyroid state. Further GO analysis suggested that thyroid state affects platelet function. CONCLUSIONS Changes in thyroid state regulate numerous genes in human whole blood, predominantly TRα-expressing leukocytes. In addition, TH may regulate gene transcripts in platelets.
Collapse
Affiliation(s)
- Elske T Massolt
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| | - Marcel E Meima
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| | | | - Selmar Leeuwenburgh
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| | | | - Giulia Brigante
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Boen L R Kam
- Department of Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | | | - Theo J Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
- Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
46
|
Coordination of platelet agonist signaling during the hemostatic response in vivo. Blood Adv 2017; 1:2767-2775. [PMID: 29296928 DOI: 10.1182/bloodadvances.2017009498] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/23/2017] [Indexed: 11/20/2022] Open
Abstract
The local microenvironment within an evolving hemostatic plug shapes the distribution of soluble platelet agonists, resulting in a gradient of platelet activation. We previously showed that thrombin activity at a site of vascular injury is spatially restricted, resulting in robust activation of a subpopulation of platelets in the hemostatic plug core. In contrast, adenosine 5'-diphosphate (ADP)/P2Y12 signaling contributes to the accumulation of partially activated, loosely packed platelets in a shell overlying the core. The contribution of the additional platelet agonists thromboxane A2 (TxA2) and epinephrine to this hierarchical organization was not previously shown. Using a combination of genetic and pharmacologic approaches coupled with real-time intravital imaging, we show that TxA2 signaling is critical and nonredundant with ADP/P2Y12 for platelet accumulation in the shell region but not required for full platelet activation in the hemostatic plug core, where thrombin activity is highest. In contrast, epinephrine signaling is dispensable even in the presence of a P2Y12 antagonist. Finally, dual P2Y12 and thrombin inhibition does not substantially inhibit hemostatic plug core formation any more than thrombin inhibition alone, providing further evidence that thrombin is the primary driver of platelet activation in this region. Taken together, these studies show for the first time how thrombin, P2Y12, and TxA2 signaling are coordinated during development of a hierarchical organization of hemostatic plugs in vivo and provide novel insights into the impact of dual antiplatelet therapy on hemostasis and thrombosis.
Collapse
|
47
|
Grove EL, Hossain R, Storey RF. Platelet function testing and prediction of procedural bleeding risk. Thromb Haemost 2017; 109:817-24. [DOI: 10.1160/th12-11-0806] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 02/16/2013] [Indexed: 11/05/2022]
Abstract
SummaryThe essential role of platelets in haemostasis underlies the relationship between platelet function and spontaneous or procedure-related bleeding, which has important prognostic implications. Although not routinely undertaken, platelet function testing offers the potential to tailor antiplatelet therapy for individual patients. However, uncertainties remain about how well platelet function testing may predict haemostasis and guide management of bleeding risk. Studies of aspirin, P2Y12 inhibitors and other antiplatelet drugs clearly demonstrate how inhibition of platelet function increases bleeding risk. More potent antiplatelet drugs are associated with higher bleeding rates, consistent with the levels of platelet inhibition achieved by these drugs. Studies of patients treated with clopidogrel, which is associated with wide inter-individual variation in antiplatelet effect, suggest that platelet function testing may predict bleeding risk related to coronary artery bypass grafting (CABG) surgery and potentially guide the timing of surgery following discontinuation of clopidogrel. Similarly, some studies have demonstrated a relationship between clopidogrel response and bleeding in patients undergoing percutaneous coronary intervention (PCI), although other studies have not supported this. Carriage of the *17 allele of cytochrome P450 2C19, which is associated with gain of function and enhanced response to clopidogrel, seems to be associated with increased bleeding risk, although studies showing lack of apparent effect of loss-of-function alleles provide contradictory evidence. Further large studies are needed to guide best practice in the application of platelet function testing in the clinical management of patients treated with antiplatelet drugs in order to optimise individual care.
Collapse
|
48
|
Role of New Antiplatelet Drugs on Cardiovascular Disease: Update on Cangrelor. Curr Atheroscler Rep 2017; 18:66. [PMID: 27714642 DOI: 10.1007/s11883-016-0617-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dual therapy with a P2Y12 receptor antagonist in addition to aspirin is the antiplatelet treatment of choice in patients with acute coronary syndromes or undergoing percutaneous coronary intervention (PCI). However, available oral P2Y12 antagonists have several limitations, mostly due to their pharmacological profile, which can affect outcomes in certain clinical settings. Cangrelor is an intravenous, direct-acting, potent P2Y12 inhibitor with rapid onset and offset of action, which has been recently approved for clinical use in patients undergoing PCI. In clinical trials, cangrelor has demonstrated greater efficacy than clopidogrel with a favorable safety profile among PCI patients not receiving pretreatment with oral P2Y12 antagonists. However, its definitive role in contemporary practice is yet to be determined. This review aims to provide a comprehensive overview of the current status of knowledge on cangrelor, focusing on its pharmacological properties, clinical development, and the potential applications of this newly available agent.
Collapse
|
49
|
Amison R, Arnold S, O'Shaughnessy B, Cleary S, Ofoedu J, Idzko M, Page C, Pitchford S. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm Pharmacol Ther 2017; 45:62-68. [DOI: 10.1016/j.pupt.2017.05.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/04/2017] [Accepted: 05/06/2017] [Indexed: 11/29/2022]
|
50
|
The potential role of platelets in the consensus molecular subtypes of colorectal cancer. Cancer Metastasis Rev 2017; 36:273-288. [DOI: 10.1007/s10555-017-9678-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|