1
|
Demir EA, Gonder O. Ticagrelor-related dyspnea beyond adenosine: Insights into retrotrapezoid hyperactivity. Respir Physiol Neurobiol 2025; 331:104349. [PMID: 39293566 DOI: 10.1016/j.resp.2024.104349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Ticagrelor, a P2Y12 receptor antagonist, has been demonstrated to induce dyspnea, which is not associated with cardiac or pulmonary alterations, or metabolic disturbances. The attribution of ticagrelor-related dyspnea to excess adenosine has been widely proposed, yet is not supported by experimental data. In this paper, we put forth a novel hypothesis that the hyperactivity of the retrotrapezoid nucleus, a group of ventral medullary neurons involved in respiratory modulation, is the underlying cause of ticagrelor-related dyspnea. This hypothesis offers a theoretical resolution to the discrepancies and controversies present in previous theories.
Collapse
Affiliation(s)
- Enver Ahmet Demir
- Department of Cardiology, Ankara Etlik City Hospital, Ankara, Republic of Turkey.
| | - Okan Gonder
- Department of Cardiology, Ankara Etlik City Hospital, Ankara, Republic of Turkey
| |
Collapse
|
2
|
Sidiropoulou S, Gatsiou A, Hansson KM, Tsouka AN, Stellos K, Tselepis AD. Ticagrelor Induces Angiogenesis in Progenitor and Mature Endothelial Cells In Vitro: Investigation of the Possible Role of Adenosine. Int J Mol Sci 2024; 25:13343. [PMID: 39769108 PMCID: PMC11727715 DOI: 10.3390/ijms252413343] [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: 09/30/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Ticagrelor, a reversible platelet P2Y12 receptor antagonist, exerts various pleiotropic actions, some of which are at least partially mediated through adenosine. We studied the ticagrelor and adenosine effect on the angiogenic properties of progenitor CD34+-derived endothelial colony-forming cells (ECFCs). Angiogenesis studies were performed in vitro using capillary-like tube formation and spheroid-based angiogenesis assays. The effects of adenosine receptor antagonists, including DPCPX (A1 antagonist), SCH58621 (A2A antagonist), MRS1706 (A2B inverse agonist and antagonist), MRS1220 (A3 antagonist) and adenosine deaminase (ADA), were also investigated. Ticagrelor, adenosine, and their combination increased capillary-like tube formation and spheroid sprout formation by ECFCs in a dose-dependent manner. This effect was significantly reduced by SCH58621, MRS1706, and their combination, as well as by ADA. By contrast, DPCPX and MRS1220 did not exhibit any inhibitory effects. Similar results were obtained when mature human umbilical vein endothelial cells (HUVECs) were studied. These results show that ticagrelor stimulates angiogenesis by progenitor and mature endothelial cells in an adenosine-dependent pathway in which the adenosine receptors A2A and A2B play major roles. The significance of these results at the clinical level in patients with atherothrombotic events and treated with ticagrelor needs to be investigated.
Collapse
Affiliation(s)
- Sofia Sidiropoulou
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 451 10 Ioannina, Greece; (S.S.); (A.N.T.)
| | - Aikaterini Gatsiou
- Cardiovascular Disease Prevention Hub, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.G.); (K.S.)
| | - Kenny M. Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden;
| | - Aikaterini N. Tsouka
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 451 10 Ioannina, Greece; (S.S.); (A.N.T.)
| | - Konstantinos Stellos
- Cardiovascular Disease Prevention Hub, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.G.); (K.S.)
- Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne NE7 7DN, UK
- Department of Cardiology, University Hospital Mannheim, University of Heidelberg, 69117 Mannheim, Germany
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany
| | - Alexandros D. Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 451 10 Ioannina, Greece; (S.S.); (A.N.T.)
| |
Collapse
|
3
|
Tsiafoutis I, Zografos T, Karelas D, Varelas P, Manousopoulos K, Nenekidis I, Koutouzis M, Lagadinos P, Koudounis P, Agelaki M, Katsanou K, Oikonomou E, Siasos G, Katsivas A. Ticagrelor potentiates cardioprotection by remote ischemic preconditioning: the ticagrelor in remote ischemic preconditioning (TRIP) randomized clinical trial. Hellenic J Cardiol 2024:S1109-9666(24)00133-7. [PMID: 38950885 DOI: 10.1016/j.hjc.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/20/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVE Remote ischemic preconditioning (RIPC) reduces periprocedural myocardial injury (PMI) after percutaneous coronary intervention (PCI) through various pathways, including an adenosine-triggered pathway. Ticagrelor inhibits adenosine uptake, thus may potentiate the effects of RIPC. This randomized trial tested the hypothesis that ticagrelor potentiates the effect of RIPC and reduces PMI, assessed by post-procedural troponin release. METHODS Patients undergoing PCI for non-ST elevation acute coronary syndromes were 1:1 randomized to ticagrelor (TG-Group) or clopidogrel (CL-Group). Within each treatment, patients were 1:1 randomized to a RIPC (RIPC-Group) or a control group (CTRL-Group). The primary endpoint was the difference between post- and pre-procedural troponin at 24 h following PCI, termed deltaTnI. RESULTS During a 12-month period, 138 patients were included in the study (34 in the CL-CTRL group, 34 in the TG-CTRL group, 35 in the CL-RIPC group, and 35 in the TG-CTRL group). There was a significant difference in deltaTnI between the study groups [ TG-RIPC:0.04 (0-0.16), CL-CTRL:0.10 (0.03-0.43), CLRIPC:0.11 (0.03-0.89), and TG-CTRL:0.24 (0.06-0.47); p = 0.007]. Eight patients (22.9%) in the TG-RIPC group developed type 4a myocardial infarction (MI), compared to 14 (40%) in the CL-RIPC group, 13 (38.2%) in the CL-CTRL group, and 19 (55.9%) in the TG-CTRL group (p = 0.048). A significant interaction between antiplatelet group allocation and RIPC on deltaTnI was observed [F (1,134) = 7.509; p = 0.007]. In multivariate analysis, the interaction between RIPC and ticagrelor treatment was independently associated with a lower incidence of Type 4a MI. CONCLUSION Our results demonstrate an interaction between ticagrelor and RIPC, which may potentiate the cardioprotective effects of RIPC during PCI by reducing PMI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Athens 11527, Greece
| | | |
Collapse
|
4
|
Akkaif MA, Ng ML, Sk Abdul Kader MA, Daud NAA, Sha'aban A, Ibrahim B. A review of the effects of ticagrelor on adenosine concentration and its clinical significance. Pharmacol Rep 2021; 73:1551-1564. [PMID: 34283374 DOI: 10.1007/s43440-021-00309-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Ticagrelor is an oral antiplatelet drug that can reversibly bind to the platelet P2Y12 receptor. Ticagrelor is metabolized mainly by CYP3A4 and produces a rapid blood concentration-dependent platelet inhibitory effect. Unlike other P2Y12 receptor antagonists, many clinical features of ticagrelor are not related to P2Y12 receptor antagonism. PURPOSE This review aims to gather existing literature on the clinical effects of ticagrelor after inhibiting adenosine uptake. METHODOLOGY The current study reviewed literature related to the effects of ticagrelor on adenosine metabolism. The review also examined the drug's biological effects and clinical characteristics to see how it could be used in a clinical setting. RESULTS Many studies have shown that ticagrelor can inhibit equilibrative nucleoside transporter 1 (ENT1). This inhibition leads to intracellular adenosine uptake, increased adenosine half-life and plasma concentration levels and an enhanced adenosine-mediated biological effect. CONCLUSIONS Based on the studies reviewed, it was found that ticagrelor essentially inhibits adenosine absorption of adenosine into cells through ENT1, which increases the concentration in the blood and subsequently increases the protection of the heart muscle by adenosine. It also prevents platelet aggregation, and extends the biological effects of coronary arteries. Moreover, it leads to a lower mortality rate in acute coronary syndrome (ACS) patients.
Collapse
Affiliation(s)
- Mohammed Ahmed Akkaif
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Mei Li Ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Gelugor, Penang, Malaysia
| | | | - Nur Aizati Athirah Daud
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Abubakar Sha'aban
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Baharudin Ibrahim
- Faculty of Pharmacy, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
Hjortbak MV, Olesen KKW, Seefeldt JM, Lassen TR, Jensen RV, Perkins A, Dodd M, Clayton T, Yellon D, Hausenloy DJ, Bøtker HE. Translation of experimental cardioprotective capability of P2Y 12 inhibitors into clinical outcome in patients with ST-elevation myocardial infarction. Basic Res Cardiol 2021; 116:36. [PMID: 34037861 DOI: 10.1007/s00395-021-00870-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/13/2021] [Indexed: 11/25/2022]
Abstract
We studied the translational cardioprotective potential of P2Y12 inhibitors against acute myocardial ischemia/reperfusion injury (IRI) in an animal model of acute myocardial infarction and in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI). P2Y12 inhibitors may have pleiotropic effects to induce cardioprotection against acute myocardial IRI beyond their inhibitory effects on platelet aggregation. We compared the cardioprotective effects of clopidogrel, prasugrel, and ticagrelor on infarct size in an in vivo rat model of acute myocardial IRI, and investigated the effects of the P2Y12 inhibitors on enzymatic infarct size (48-h area-under-the-curve (AUC) troponin T release) and clinical outcomes in a retrospective study of STEMI patients from the CONDI-2/ERIC-PPCI trial using propensity score analyses. Loading with ticagrelor in rats reduced infarct size after acute myocardial IRI compared to controls (37 ± 11% vs 52 ± 8%, p < 0.01), whereas clopidogrel and prasugrel did not (50 ± 11%, p > 0.99 and 49 ± 9%, p > 0.99, respectively). Correspondingly, troponin release was reduced in STEMI patients treated with ticagrelor compared to clopidogrel (adjusted 48-h AUC ratio: 0.67, 95% CI 0.47-0.94). Compared to clopidogrel, the composite endpoint of cardiac death or hospitalization for heart failure within 12 months was reduced in STEMI patients loaded with ticagrelor (HR 0.63; 95% CI 0.42-0.94) but not prasugrel (HR 0.84, 95% CI 0.43-1.63), prior to PPCI. Major adverse cardiovascular events did not differ between clopidogrel, ticagrelor, or prasugrel. The cardioprotective effects of ticagrelor in reducing infarct size may contribute to the clinical benefit observed in STEMI patients undergoing PPCI.
Collapse
Affiliation(s)
- Marie V Hjortbak
- Department of Clinical Medicine, Cardiology, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark.
| | - Kevin K W Olesen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob M Seefeldt
- Department of Clinical Medicine, Cardiology, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Thomas R Lassen
- Department of Clinical Medicine, Cardiology, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Rebekka V Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander Perkins
- London School of Hygiene and Tropical Medicine, Clinical Trials Unit, London, UK
| | - Matthew Dodd
- London School of Hygiene and Tropical Medicine, Clinical Trials Unit, London, UK
| | - Tim Clayton
- London School of Hygiene and Tropical Medicine, Clinical Trials Unit, London, UK
| | - Derek Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Hearts Centre, Singapore Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Singapore, Singapore
| | - Hans Erik Bøtker
- Department of Clinical Medicine, Cardiology, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark.,Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
6
|
Abstract
Nucleosides play central roles in all facets of life, from metabolism to cellular signaling. Because of their physiochemical properties, nucleosides are lipid bilayer impermeable and thus rely on dedicated transport systems to cross biological membranes. In humans, two unrelated protein families mediate nucleoside membrane transport: the concentrative and equilibrative nucleoside transporter families. The objective of this review is to provide a broad outlook on the current status of nucleoside transport research. We will discuss the role played by nucleoside transporters in human health and disease, with emphasis placed on recent structural advancements that have revealed detailed molecular principles of these important cellular transport systems and exploitable pharmacological features.
Collapse
Affiliation(s)
- Nicholas J. Wright
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
- Correspondence and requests for materials should be addressed to: S.-Y. Lee., , tel: 919-684-1005, fax: 919-684-8885
| |
Collapse
|
7
|
The pleiotropic effects of antithrombotic drugs in the metabolic-cardiovascular-neurodegenerative disease continuum: impact beyond reduced clotting. Clin Sci (Lond) 2021; 135:1015-1051. [PMID: 33881143 DOI: 10.1042/cs20201445] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
Antithrombotic drugs are widely used for primary and secondary prevention, as well as treatment of many cardiovascular disorders. Over the past few decades, major advances in the pharmacology of these agents have been made with the introduction of new drug classes as novel therapeutic options. Accumulating evidence indicates that the beneficial outcomes of some of these antithrombotic agents are not solely related to their ability to reduce thrombosis. Here, we review the evidence supporting established and potential pleiotropic effects of four novel classes of antithrombotic drugs, adenosine diphosphate (ADP) P2Y12-receptor antagonists, Glycoprotein IIb/IIIa receptor Inhibitors, and Direct Oral Anticoagulants (DOACs), which include Direct Factor Xa (FXa) and Direct Thrombin Inhibitors. Specifically, we discuss the molecular evidence supporting such pleiotropic effects in the context of cardiovascular disease (CVD) including endothelial dysfunction (ED), atherosclerosis, cardiac injury, stroke, and arrhythmia. Importantly, we highlight the role of DOACs in mitigating metabolic dysfunction-associated cardiovascular derangements. We also postulate that DOACs modulate perivascular adipose tissue inflammation and thus, may reverse cardiovascular dysfunction early in the course of the metabolic syndrome. In this regard, we argue that some antithrombotic agents can reverse the neurovascular damage in Alzheimer's and Parkinson's brain and following traumatic brain injury (TBI). Overall, we attempt to provide an up-to-date comprehensive review of the less-recognized, beneficial molecular aspects of antithrombotic therapy beyond reduced thrombus formation. We also make a solid argument for the need of further mechanistic analysis of the pleiotropic effects of antithrombotic drugs in the future.
Collapse
|
8
|
Wernly B, Erlinge D, Pernow J, Zhou Z. Ticagrelor: a cardiometabolic drug targeting erythrocyte-mediated purinergic signaling? Am J Physiol Heart Circ Physiol 2020; 320:H90-H94. [PMID: 33095055 DOI: 10.1152/ajpheart.00570.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiometabolic diseases lead to vascular complications, which cause increasing morbidity and mortality worldwide. The underlying mechanisms are multifactorial and complex but may involve altered purinergic signaling that significantly contributes to cardiovascular dysfunction. Ticagrelor is a successful purinergic drug directly targeting ADP-mediated P2Y12R signaling for platelet aggregation and is widely used in patients with acute coronary syndrome. In addition, ticagrelor can target red blood cells (RBCs) to release ATP and inhibit adenosine uptake by RBCs, which subsequently activate purinergic signaling. This involvement in purinergic signaling may allow ticagrelor to mediate pleiotropic effects and contribute to the beneficial cardiovascular outcomes observed in clinical studies. Recent studies have established a novel function of RBCs, which is that RBCs act as disease mediators for the development of cardiovascular complications in type 2 diabetes (T2D). RBC-released ATP is defective in T2D, which has implications for the induction of vascular dysfunction by dysregulating purinergic signaling. Ticagrelor might target RBCs and restore the bioavailability of ATP and adenosine, thereby attenuating cardiovascular complications. The present perspective discusses the pleiotropic effect of ticagrelor, with a focus on the possibility of ticagrelor for the treatment of cardiometabolic complications by targeting RBCs and initiating purinergic activation. A better understanding of the proposed cardiometabolic effects could support novel clinical indications for ticagrelor application.
Collapse
Affiliation(s)
- Bernhard Wernly
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - David Erlinge
- Department of Clinical Sciences, Cardiology, Lund University Hospital, Lund, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Furtado RHM, Venkateswaran RV, Nicolau JC, Gurmu Y, Bhatt DL, Storey RF, Steg PG, Magnani G, Goto S, Dellborg M, Kamensky G, Isaza D, Aylward P, Johanson P, Bonaca MP. Caffeinated Beverage Intake, Dyspnea With Ticagrelor, and Cardiovascular Outcomes: Insights From the PEGASUS-TIMI 54 Trial. J Am Heart Assoc 2020; 9:e015785. [PMID: 32410485 PMCID: PMC7660882 DOI: 10.1161/jaha.119.015785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background A proposed cause of dyspnea induced by ticagrelor is an increase in adenosine blood levels. Because caffeine is an adenosine antagonist, it can potentially improve drug tolerability with regard to dyspnea. Furthermore, association between caffeine and cardiovascular events is of clinical interest. Methods and Results This prespecified analysis used data from the PEGASUS TIMI 54 (Prevention of Cardiovascular Events in Patients With Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin–Thrombolysis in Myocardial Infarction 54) trial, which randomized 21 162 patients with prior myocardial infarction to ticagrelor 60 mg or 90 mg or matching placebo (twice daily). Baseline caffeine intake in cups per week was prospectively collected for 9694 patients. Outcomes of interest included dyspnea, major adverse cardiovascular events (ie, the composite of cardiovascular death, myocardial infarction, or stroke), and arrhythmias. Dyspnea analyses considered the pooled ticagrelor group, whereas cardiovascular outcome analyses included patients from the 3 randomized arms. After adjustment, caffeine intake, compared with no intake, was not associated with lower rates of dyspnea in patients taking ticagrelor (adjusted hazard ratio (HR), 0.91; 95% CI, 0.76–1.10; P=0.34). There was no excess risk with caffeine for major adverse cardiovascular events (adjusted HR, 0.78; 95% CI, 0.63–0.98; P=0.031), sudden cardiac death (adjusted HR, 0.98; 95% CI, 0.57–1.70; P=0.95), or atrial fibrillation (adjusted odds ratio, 1.07; 95% CI, 0.56–2.04; P=0.84). Conclusions In patients taking ticagrelor for secondary prevention after myocardial infarction, caffeine intake at baseline was not associated with lower rates of dyspnea compared with no intake. Otherwise, caffeine appeared to be safe in this population, with no apparent increase in atherothrombotic events or clinically significant arrhythmias. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT01225562.
Collapse
Affiliation(s)
- Remo H M Furtado
- TIMI Study Group Brigham and Women's Hospital Harvard Medical School Boston MA.,Instituto do Coracao (InCor) Hospital das Clinicas da Faculdade de Medicina Universidade de Sao Paulo Brazil.,Hospital Albert Einstein Sao Paulo Brazil
| | | | - Jose C Nicolau
- Instituto do Coracao (InCor) Hospital das Clinicas da Faculdade de Medicina Universidade de Sao Paulo Brazil
| | - Yared Gurmu
- TIMI Study Group Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Deepak L Bhatt
- TIMI Study Group Brigham and Women's Hospital Harvard Medical School Boston MA
| | | | - P Gabriel Steg
- Université de Paris, and Assistance Publique-Hôpitaux de Paris Paris France
| | | | - Shinya Goto
- Department of Medicine (Cardiology) Tokai University Hospital Isehara Japan
| | | | - Gabriel Kamensky
- Department of Non-invasive Cardiovascular Diagnostics University Hospital Bratislava Bratislava Slovakia
| | - Daniel Isaza
- Fundacion Cardioinfantil Instituto de Cardiologia Bogotá Colombia
| | - Philip Aylward
- South Australian Health and Medical Research Institute Flinders University and Medical Centre Adelaide Australia
| | | | - Marc P Bonaca
- TIMI Study Group Brigham and Women's Hospital Harvard Medical School Boston MA.,CPC Clinical Research and Vascular Research Unity University of Colorado Denver CO
| |
Collapse
|
10
|
Sabbah M, Nepper-Christensen L, Køber L, Høfsten DE, Ahtarovski KA, Göransson C, Kyhl K, Ghotbi AA, Schoos MM, Sadjadieh G, Kelbæk H, Lønborg J, Engstrøm T. Infarct size following loading with Ticagrelor/Prasugrel versus Clopidogrel in ST-segment elevation myocardial infarction. Int J Cardiol 2020; 314:7-12. [PMID: 32389767 DOI: 10.1016/j.ijcard.2020.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/14/2020] [Accepted: 05/04/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Treatment with newer direct-acting anti-platelet drugs (Ticagrelor and Prasugrel) prior to primary percutaneous coronary intervention (PCI) is associated with improved outcome in patients with ST-segment elevation myocardial infarction (STEMI) when compared with Clopidogrel. We compared infarct size following treatment with Ticagrelor/Prasugrel versus Clopidogrel in the DANish trial in Acute Myocardial Infarction (DANAMI-3) population of STEMI patients treated with primary PCI. METHODS AND RESULTS Patients were loaded with Clopidogrel, Ticagrelor or Prasugrel in the ambulance before primary PCI. Infarct size and myocardial salvage index were calculated using cardiac magnetic resonance (CMR) during index admission and at three-month follow-up. Six-hundred-and-ninety-three patients were included in this analysis. Clopidogrel was given to 351 patients and Ticagrelor/Prasugrel to 342 patients. The groups were generally comparable in terms of baseline and procedural characteristics. Median infarct size at three-month follow-up was 12.9% vs 10.0%, in patients treated with Clopidogrel and Ticagrelor/ Prasugrel respectively (p < 0.001), and myocardial salvage index was 66% vs 71% (p < 0.001). Results remained significant in a multiple regression model (p < 0.001). CONCLUSIONS Pre-hospital loading with Ticagrelor or Prasugrel compared to Clopidogrel, was associated with smaller infarct size and larger myocardial salvage index at three-month follow-up in patients with STEMI treated with primary PCI.
Collapse
Affiliation(s)
- Muhammad Sabbah
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark.
| | - Lars Nepper-Christensen
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Lars Køber
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Dan Eik Høfsten
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | | | - Christoffer Göransson
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Kasper Kyhl
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Adam Ali Ghotbi
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Mikkel Malby Schoos
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Golnaz Sadjadieh
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Henning Kelbæk
- Zealand University Hospital, Department of Cardiology, Roskilde, Denmark
| | - Jacob Lønborg
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| | - Thomas Engstrøm
- Rigshospitalet - Copenhagen University Hospital, Department of Cardiology, Copenhagen, Denmark
| |
Collapse
|
11
|
Ticagrelor Increases SIRT1 and HES1 mRNA Levels in Peripheral Blood Cells from Patients with Stable Coronary Artery Disease and Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2020; 21:ijms21051576. [PMID: 32106619 PMCID: PMC7084534 DOI: 10.3390/ijms21051576] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/13/2022] Open
Abstract
Ticagrelor is a powerful P2Y12 inhibitor with pleiotropic effects in the cardiovascular system. Consistently, we have reported that in patients with stable coronary artery disease (CAD) and concomitant chronic obstructive pulmonary disease (COPD) who underwent percutaneous coronary intervention (PCI), 1-month treatment with ticagrelor was superior in improving biological markers of endothelial function, compared with clopidogrel. The objective of this study was to investigate the mechanisms underlying these beneficial effects of ticagrelor by conducting molecular analyses of RNA isolated from peripheral blood cells of these patients. We determined mRNAs levels of markers of inflammation and oxidative stress, such as RORγt (T helper 17 cells marker), FoxP3 (regulatory T cells marker), NLRP3, ICAM1, SIRT1, Notch ligands JAG1 and DLL4, and HES1, a Notch target gene. We found that 1-month treatment with ticagrelor, but not clopidogrel, led to increased levels of SIRT1 and HES1 mRNAs. In patients treated with ticagrelor or clopidogrel, we observed a negative correlation among changes in both SIRT1 and HES1 mRNA and serum levels of Epidermal Growth Factor (EGF), a marker of endothelial dysfunction found to be reduced by ticagrelor treatment in our previous study. In conclusion, we report that in stable CAD/COPD patients ticagrelor positively regulates HES1 and SIRT1, two genes playing a protective role in the context of inflammation and oxidative stress. Our observations confirm and expand previous studies showing that the beneficial effects of ticagrelor in stable CAD/COPD patients may be, at least in part, mediated by its capacity to reduce systemic inflammation and oxidative stress.
Collapse
|
12
|
Ticagrelor Enhances Release of Anti-Hypoxic Cardiac Progenitor Cell-Derived Exosomes Through Increasing Cell Proliferation In Vitro. Sci Rep 2020; 10:2494. [PMID: 32051439 PMCID: PMC7016113 DOI: 10.1038/s41598-020-59225-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the widespread clinical use of cardioprotection by long-term direct antagonism of P2Y12 receptor, underlying mechanisms are unclear. Here, we identify how release of pro-survival exosomes from human cardiac-derived mesenchymal progenitor cells (hCPCs) is regulated by clinically relevant dose of ticagrelor (1 μM), an oral selective and reversible non-thienopyridine P2Y12 inhibitor. Ticagrelor-induced enhancement of exosome levels is related to increased mitotic activity of hCPCs. We show a drug-response threshold above which the effects on hCPCs are lost due to higher dose of ticagrelor and larger adenosine levels. While it is known that pan-Aurora kinase inhibitor halts cell proliferation through dephosphorylation of histone H3 residue Ser10, we demonstrate that it also prevents ticagrelor-induced effects on release of cardiac progenitor cell-derived exosomes delivering anti-apoptotic HSP70. Indeed, sustained pre-treatment of cardiomyocytes with exosomes released from explant-derived hCPCs exposed to low-dose ticagrelor attenuated hypoxia-induced apoptosis through acute phosphorylation of ERK42/44. Our data indicate that ticagrelor can be leveraged to modulate release of anti-hypoxic exosomes from resident hCPCs.
Collapse
|
13
|
Brown DG, Smith GF, Wobst HJ. Promiscuity of in Vitro Secondary Pharmacology Assays and Implications for Lead Optimization Strategies. J Med Chem 2019; 63:6251-6275. [PMID: 31714773 DOI: 10.1021/acs.jmedchem.9b01625] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We conducted an analysis on screening data generated from 1445 compounds against a panel of 130 enzymes, ion channels, and receptors to assess secondary pharmacological risks. Hit rates of these targets as well as physicochemical properties for those hits were evaluated. A majority of targets yielded hits with higher clogP, molecular weight, and more basic character than inactive compounds. Although most targets favored lipophilic hits, the average clogP of hits at a given target did not correlate with its hit rate. Furthermore, a matched pair analysis was completed to determine structural changes that impacted off-target activities. A correlation of binding assays used in this analysis illustrated that some pharmacologically related binding assays are highly correlative and may be substituted for a smaller set of surrogate assays.
Collapse
Affiliation(s)
- Dean G Brown
- Hit Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Graham F Smith
- Data Science and Artificial Intelligence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Heike J Wobst
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| |
Collapse
|
14
|
van Leeuwen MAH, van der Hoeven NW, Janssens GN, Everaars H, Nap A, Lemkes JS, de Waard GA, van de Ven PM, van Rossum AC, Ten Cate TJF, Piek JJ, von Birgelen C, Escaned J, Valgimigli M, Diletti R, Riksen NP, van Mieghem NM, Nijveldt R, van Royen N. Evaluation of Microvascular Injury in Revascularized Patients With ST-Segment-Elevation Myocardial Infarction Treated With Ticagrelor Versus Prasugrel. Circulation 2019; 139:636-646. [PMID: 30586720 DOI: 10.1161/circulationaha.118.035931] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Despite successful restoration of epicardial vessel patency with primary percutaneous coronary intervention, coronary microvascular injury occurs in a large proportion of patients with ST-segment-elevation myocardial infarction, adversely affecting clinical and functional outcome. Ticagrelor has been reported to increase plasma adenosine levels, which might have a protective effect on the microcirculation. We investigated whether ticagrelor maintenance therapy after revascularized ST-segment-elevation myocardial infarction is associated with less coronary microvascular injury compared to prasugrel maintenance therapy. METHODS A total of 110 patients with ST-segment-elevation myocardial infarction received a loading dose of ticagrelor and were randomized to maintenance therapy of ticagrelor (n=56) or prasugrel (n=54) after primary percutaneous coronary intervention. The primary outcome was coronary microvascular injury at 1 month, as determined with the index of microcirculatory resistance in the infarct-related artery. Cardiovascular magnetic resonance imaging was performed during the acute phase and at 1 month. RESULTS The primary outcome of index of microcirculatory resistance was not superior in ticagrelor- or prasugrel-treated patients (ticagrelor, 21 [interquartile range, 15-39] U; prasugrel, 18 [interquartile range, 11-29] U; P=0.08). Recovery of microcirculatory resistance over time was not better in patients with ticagrelor versus prasugrel (ticagrelor, -13.9 U; prasugrel, -13.5 U; P=0.96). Intramyocardial hemorrhage was observed less frequently in patients receiving ticagrelor (23% versus 43%; P=0.04). At 1 month, no difference in infarct size was observed (ticagrelor, 7.6 [interquartile range, 3.7-14.4] g, prasugrel 9.9 [interquartile range, 5.7-16.6] g; P=0.17). The occurrence of microvascular obstruction was not different in patients on ticagrelor (28%) or prasugrel (41%; P=0.35). Plasma adenosine concentrations were not different during the index procedure and during maintenance therapy with ticagrelor or prasugrel. CONCLUSIONS In patients with ST-segment-elevation myocardial infarction, ticagrelor maintenance therapy was not superior to prasugrel in preventing coronary microvascular injury in the infarct-related territory as assessed by the index of microcirculatory resistance, and this resulted in a comparable infarct size at 1 month. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov . Unique identifier: NCT02422888.
Collapse
Affiliation(s)
- Maarten A H van Leeuwen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.).,Department of Cardiology, Isala Heart Centre, Zwolle, The Netherlands (M.A.H.v.L.)
| | - Nina W van der Hoeven
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Gladys N Janssens
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Henk Everaars
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Alexander Nap
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Jorrit S Lemkes
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Guus A de Waard
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Peter M van de Ven
- Department of Epidemiology and Biostatistics, VU University, Amsterdam, The Netherlands (P.M.v.d.V.)
| | - Albert C van Rossum
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.)
| | - Tim J F Ten Cate
- Department of Cardiology (T.J.F.t.C., R.N., N.v.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan J Piek
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands (J.J.P.)
| | - Clemens von Birgelen
- Department of Cardiology, Medisch Spectrum Twente, Enschede, The Netherlands (C.v.B.)
| | - Javier Escaned
- Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (J.E.)
| | - Marco Valgimigli
- Department of Cardiology, Bern University Hospital, Switzerland (M.V.)
| | - Roberto Diletti
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands (R.D., N.M.v.M.)
| | - Niels P Riksen
- Department of Internal Medicine (N.P.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Robin Nijveldt
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.).,Department of Cardiology (T.J.F.t.C., R.N., N.v.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niels van Royen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (M.A.H.v.L., N.W.v.d.H., G.N.J., H.E., A.N., J.S.L., G.A.d.W., A.C.v.R., R.N., N.v.R.).,Department of Cardiology (T.J.F.t.C., R.N., N.v.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
15
|
Reiss AB, Grossfeld D, Kasselman LJ, Renna HA, Vernice NA, Drewes W, Konig J, Carsons SE, DeLeon J. Adenosine and the Cardiovascular System. Am J Cardiovasc Drugs 2019; 19:449-464. [PMID: 30972618 PMCID: PMC6773474 DOI: 10.1007/s40256-019-00345-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenosine is an endogenous nucleoside with a short half-life that regulates many physiological functions involving the heart and cardiovascular system. Among the cardioprotective properties of adenosine are its ability to improve cholesterol homeostasis, impact platelet aggregation and inhibit the inflammatory response. Through modulation of forward and reverse cholesterol transport pathways, adenosine can improve cholesterol balance and thereby protect macrophages from lipid overload and foam cell transformation. The function of adenosine is controlled through four G-protein coupled receptors: A1, A2A, A2B and A3. Of these four, it is the A2A receptor that is in a large part responsible for the anti-inflammatory effects of adenosine as well as defense against excess cholesterol accumulation. A2A receptor agonists are the focus of efforts by the pharmaceutical industry to develop new cardiovascular therapies, and pharmacological actions of the atheroprotective and anti-inflammatory drug methotrexate are mediated via release of adenosine and activation of the A2A receptor. Also relevant are anti-platelet agents that decrease platelet activation and adhesion and reduce thrombotic occlusion of atherosclerotic arteries by antagonizing adenosine diphosphate-mediated effects on the P2Y12 receptor. The purpose of this review is to discuss the effects of adenosine on cell types found in the arterial wall that are involved in atherosclerosis, to describe use of adenosine and its receptor ligands to limit excess cholesterol accumulation and to explore clinically applied anti-platelet effects. Its impact on electrophysiology and use as a clinical treatment for myocardial preservation during infarct will also be covered. Results of cell culture studies, animal experiments and human clinical trials are presented. Finally, we highlight future directions of research in the application of adenosine as an approach to improving outcomes in persons with cardiovascular disease.
Collapse
|
16
|
Lavi S, Alemayehu M, Puka K, Wall S, Lavi R. Association Between Administration of Ticagrelor and Microvascular Endothelial Function. JAMA Cardiol 2019; 2:1042-1044. [PMID: 28746721 DOI: 10.1001/jamacardio.2017.2378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Shahar Lavi
- London Health Sciences Centre, London, Ontario, Canada.,Western University, London, Ontario, Canada
| | | | | | - Sabrina Wall
- London Health Sciences Centre, London, Ontario, Canada
| | - Ronit Lavi
- London Health Sciences Centre, London, Ontario, Canada.,Western University, London, Ontario, Canada
| |
Collapse
|
17
|
|
18
|
Máchal J, Hlinomaz O. Efficacy of P2Y12 Receptor Blockers After Myocardial Infarction and Genetic Variability of their Metabolic Pathways. Curr Vasc Pharmacol 2018; 17:35-40. [DOI: 10.2174/1570161116666180206110657] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/18/2017] [Accepted: 11/07/2017] [Indexed: 01/15/2023]
Abstract
Background: Various antiplatelet drugs are used following Acute Coronary Syndromes
(ACS). Of them, adenosine diphosphate receptor P2Y12 inhibitors clopidogrel, prasugrel and ticagrelor
are currently used for post-ACS long-term treatment. Although they act on the same receptor, they differ
in pharmacodynamics and pharmacokinetics. Several enzymes and transporters involved in the metabolism
of P2Y12 inhibitors show genetic variability with functional impact. This includes Pglycoprotein,
carboxylesterase 1 and, most notably, CYP2C19 that is important in clopidogrel activation.
Common gain-of-function or loss-of-function alleles of CYP2C19 gene are associated with lower
or higher platelet reactivity that may impact clinical outcomes of clopidogrel treatment. Prasugrel is
considered to be less dependent on CYP2C19 variability as it is also metabolized by other CYP450 isoforms.
Some studies, however, showed the relevance of CYP2C19 variants for platelet reactivity during
prasugrel treatment as well. Ticagrelor is metabolized mainly by CYP3A4, which does not show functionally
relevant genetic variability. Its concentrations may be modified by the variants of Pglycoprotein
gene ABCB1. While no substantial difference between the clinical efficacy of prasugrel
and ticagrelor has been documented, both of them have been shown to be superior to clopidogrel in
post-ACS treatment. This can be partially explained by lower variability at each step of their metabolism.
It is probable that factors influencing the pharmacokinetics of both drugs, including genetic factors,
may predict the clinical efficacy of antiplatelet treatment in personalized medicine.
</P><P>
Conclusion: We summarize the pharmacokinetics and pharmacogenetics of P2Y12 inhibitors with respect
to their clinical effects in post-myocardial infarction treatment.
Collapse
Affiliation(s)
- Jan Máchal
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Ota Hlinomaz
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
19
|
Moulias A, Xanthopoulou I, Alexopoulos D. Does Ticagrelor Improve Endothelial Function? J Cardiovasc Pharmacol Ther 2018; 24:11-17. [DOI: 10.1177/1074248418786936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ticagrelor is a P2Y12 receptor antagonist with proven clinical benefit in patients with acute coronary syndrome. Apart from its principal antiplatelet action, pleiotropic effects have been implicated in the clinical profile of ticagrelor, including a potentially beneficial impact on endothelial function. In light of the common presence and prognostic value of endothelial dysfunction in patients with coronary artery disease, several clinical studies have investigated the postulated effect of ticagrelor on endothelial function, yielding conflicting results. Limitations of the relevant studies as well as substantial differences in patient population, study design, and methods may account for these controversial findings. Most of these studies, however, support a beneficial impact of ticagrelor on endothelial function, which seems to be significant in the higher risk patients. In order to elucidate this effect, further research efforts should aim to clarify how quickly does endothelial function respond to ticagrelor, how sustained this response is during the dosing intervals and in the long term, which mechanisms are implicated, and whether this pleiotropic action is clinically significant. Future studies should include larger and diverse populations of patients, assess endothelial function at several time points after treatment initiation, and use multiple methods of endothelial function measurement, while implementing strict methodology. Nevertheless, the extent of the clinical benefit of ticagrelor attributable to actions beyond its potent and consistent antiplatelet effect remains uncertain.
Collapse
Affiliation(s)
| | | | - Dimitrios Alexopoulos
- Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
20
|
Vieceli Dalla Sega F, Fortini F, Aquila G, Pavasini R, Biscaglia S, Bernucci D, Del Franco A, Tonet E, Rizzo P, Ferrari R, Campo G. Ticagrelor Improves Endothelial Function by Decreasing Circulating Epidermal Growth Factor (EGF). Front Physiol 2018; 9:337. [PMID: 29686623 PMCID: PMC5900783 DOI: 10.3389/fphys.2018.00337] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/19/2018] [Indexed: 12/13/2022] Open
Abstract
Ticagrelor is one of the most powerful P2Y12 inhibitor. We have recently reported that, in patients with concomitant Stable Coronary Artery Disease (SCAD) and Chronic Obstructive Pulmonary Disease (COPD) undergoing percutaneous coronary intervention (PCI), treatment with ticagrelor, as compared to clopidogrel, is associated with an improvement of the endothelial function (Clinical Trial NCT02519608). In the present study, we showed that, in the same population, after 1 month treatment with ticagrelor, but not with clopidogrel, there is a decrease of the circulating levels of epidermal growth factor (EGF) and that these changes in circulating levels of EGF correlate with on-treatment platelet reactivity. Furthermore, in human umbilical vein endothelial cells (HUVEC) incubated with sera of the patients treated with ticagrelor, but not with clopidogrel there is an increase of p-eNOS levels. Finally, analyzing the changes in EGF and p-eNOS levels after treatment, we observed an inverse correlation between p-eNOS and EGF changes only in the ticagrelor group. Causality between EGF and eNOS activation was assessed in vitro in HUVEC where we showed that EGF decreases eNOS activity in a dose dependent manner. Taken together our data indicate that ticagrelor improves endothelial function by lowering circulating EGF that results in the activation of eNOS in the vascular endothelium.
Collapse
Affiliation(s)
- Francesco Vieceli Dalla Sega
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.,Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Francesca Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.,Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Rita Pavasini
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Simone Biscaglia
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Davide Bernucci
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Annamaria Del Franco
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Elisabetta Tonet
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.,Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Gianluca Campo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| |
Collapse
|
21
|
Kubisa MJ, Jezewski MP, Gasecka A, Siller-Matula JM, Postuła M. Ticagrelor - toward more efficient platelet inhibition and beyond. Ther Clin Risk Manag 2018; 14:129-140. [PMID: 29398917 PMCID: PMC5775739 DOI: 10.2147/tcrm.s152369] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Novel antiplatelet drugs, including ticagrelor, are being successively introduced into the therapy of atherothrombotic conditions due to their superiority over a standard combination of clopidogrel with acetylsalicylic acid in patients with acute coronary syndromes (ACS). A P2Y12 receptor antagonist, ticagrelor, is unique among antiplatelet drugs, because ticagrelor inhibits the platelet P2Y12 receptor in a reversible manner, and because it demonstrates a wide palette of advantageous pleiotropic effects associated with the increased concentration of adenosine. The pleiotropic effects of ticagrelor comprise cardioprotection, restoration of the myocardium after an ischemic event, promotion of the release of anticoagulative factors and, eventually, anti-inflammatory effects. Beyond the advantageous effects, the increased concentration of adenosine is responsible for some of ticagrelor's adverse effects, including dyspnea and bradycardia. Large-scale clinical trials demonstrated that both standard 12-month therapy and long-term use of ticagrelor reduce the risk of cardiovascular events in patients with ACS, but at the expense of a higher risk of major bleeding. Further trials focused on the use of ticagrelor in conditions other than ACS, including ischemic stroke, peripheral artery disease and status after coronary artery bypass grafting. The results of these trials suggest comparable efficacy and safety of ticagrelor and clopidogrel in extra-coronary indications, but firm conclusions are anticipated from currently ongoing studies. Here, we summarize current evidence on the superiority of ticagrelor over other P2Y12 antagonists in ACS, discuss the mechanism underlying the drug-drug interactions and pleiotropic effects of ticagrelor, and present future perspectives of non-coronary indications for ticagrelor.
Collapse
Affiliation(s)
- Michał J Kubisa
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Mateusz P Jezewski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| | - Aleksandra Gasecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Vesicle Observation Centre, Laboratory of Experimental Clinical Chemistry, Academic Medical Centre, University of Amsterdam, the Netherlands
| | | | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT)
| |
Collapse
|
22
|
Xanthopoulou I, Bei I, Bampouri T, Barampoutis N, Moulias A, Davlouros P, Alexopoulos D. Absence of differential effect of ticagrelor versus prasugrel maintenance dose on endothelial function in patients with stable coronary artery disease. Hellenic J Cardiol 2017; 59:338-343. [PMID: 29292242 DOI: 10.1016/j.hjc.2017.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Endothelial function may be improved by ticagrelor through adenosine-mediated mechanisms. We aimed to assess the effect of ticagrelor versus prasugrel on endothelial function in patients with stable coronary artery disease (CAD). METHODS In a prospective, randomized, crossover study, 22 stable CAD patients under prasugrel 10 mg once daily maintenance dose (MD) for at least 3 months were randomized to either ticagrelor 90 mg twice daily or prasugrel 10 mg once daily for 15 days with a direct treatment-crossover for another 15 days. Endothelial function was assessed by peripheral arterial tonometry (EndoPAT 2000 system, Itamar Medical, Caesarea, Israel) at Day 0 (randomization), Day 15, and Day 30. Reactive Hyperemia Index (RHI) was calculated by using an automated software, and endothelial dysfunction (ED) was defined as RHI <1.67. RHI at the end of the two treatment periods did not differ between ticagrelor and prasugrel MD treatments. Least squares estimates (95% confidence interval) were 1.78 (1.58-1.99) versus 1.88 (1.67-2.08), with a fixed estimate of -0.099 (95% CI: -0.45 to 0.25) for the difference between them (p = 0.5). ED rate did not differ significantly between ticagrelor and prasugrel (45.5% vs 45.5%, p = 0.6). CONCLUSIONS In CAD patients, we have failed to find evidence of alteration of endothelial function following ticagrelor compared to prasugrel MD treatment, as assessed by peripheral arterial tonometry. CLINICALTRIALS. GOV UNIQUE IDENTIFIER NCT01957540.
Collapse
Affiliation(s)
| | - Ilianna Bei
- Department of Cardiology, Patras University Hospital, Rion, Patras, Greece
| | - Theodora Bampouri
- Department of Cardiology, Patras University Hospital, Rion, Patras, Greece
| | | | - Athanasios Moulias
- Department of Cardiology, Patras University Hospital, Rion, Patras, Greece
| | - Periklis Davlouros
- Department of Cardiology, Patras University Hospital, Rion, Patras, Greece
| | | |
Collapse
|
23
|
Danielak D, Karaźniewicz-Łada M, Główka F. Ticagrelor in modern cardiology - an up-to-date review of most important aspects of ticagrelor pharmacotherapy. Expert Opin Pharmacother 2017; 19:103-112. [DOI: 10.1080/14656566.2017.1421634] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Dorota Danielak
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland
| | - Marta Karaźniewicz-Łada
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland
| | - Franciszek Główka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
24
|
Kiers D, van der Heijden WA, van Ede L, Gerretsen J, de Mast Q, van der Ven AJ, El Messaoudi S, Rongen GA, Gomes M, Kox M, Pickkers P, Riksen NP. A randomised trial on the effect of anti-platelet therapy on the systemic inflammatory response in human endotoxaemia. Thromb Haemost 2017; 117:1798-1807. [PMID: 28692111 DOI: 10.1160/th16-10-0799] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/25/2017] [Indexed: 01/09/2023]
Abstract
The use of acetylsalicylic acid (ASA) is associated with improved outcome in patients with sepsis, and P2Y12 inhibitors have been suggested to also have immunomodulatory effects. Therefore, we evaluated the effects of clinically relevant combinations of antiplatelet therapy on the immune response in experimental endotoxaemia in humans in vivo. Forty healthy subjects were randomised to seven days of placebo, placebo with ASA, ticagrelor and ASA, or clopidogrel and ASA treatment. Systemic inflammation was elicited at day seven by intravenous administration of Escherichia coli endotoxin. ASA treatment profoundly augmented the plasma concentration of pro-inflammatory cytokines, but did not affect anti-inflammatory cytokines. Addition of either P2Y12 antagonist to ASA did not affect any of the circulating cytokines, except for an attenuation of the ASA-induced increase in TNFα by ticagrelor. Systemic inflammation increased plasma adenosine, without differences between groups, and although P2Y12 inhibition impaired platelet reactivity, there was no correlation with cytokine responses.
Collapse
Affiliation(s)
- Dorien Kiers
- Prof. N. P. Riksen, Dept. of Internal Medicine 463, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, The Netherlands, Tel.: +31 24 3618819, Fax: +31 24 3616519, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rosenbrier Ribeiro L, Ian Storer R. A semi-quantitative translational pharmacology analysis to understand the relationship between in vitro ENT1 inhibition and the clinical incidence of dyspnoea and bronchospasm. Toxicol Appl Pharmacol 2016; 317:41-50. [PMID: 28041785 DOI: 10.1016/j.taap.2016.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/22/2016] [Accepted: 12/27/2016] [Indexed: 01/17/2023]
Abstract
Adenosine contributes to the pathophysiology of respiratory disease, and adenosine challenge leads to bronchospasm and dyspnoea in patients. The equilibrative nucleoside transporter 1 (ENT1) terminates the action of adenosine by removal from the extracellular environment. Therefore, it is proposed that inhibition of ENT1 in respiratory disease patients leads to increased adenosine concentrations, triggering bronchospasm and dyspnoea. This study aims to assess the translation of in vitro ENT1 inhibition to the clinical incidence of bronchospasm and dyspnoea in respiratory disease, cardiovascular disease and healthy volunteer populations. Four marketed drugs with ENT1 activity were assessed; dipyridamole, ticagrelor, draflazine, cilostazol. For each patient population, the relationship between in vitro ENT1 [3H]-NBTI binding affinity (Ki) and [3H]-adenosine uptake (IC50) to the incidence of: (1) bronchospasm/severe dyspnoea; (2) tolerated dyspnoea and; (3) no adverse effects, was evaluated. A high degree of ENT1 inhibition (≥13.3x Ki, ≥4x IC50) associated with increased incidence of bronchospasm/severe dyspnoea for patients with respiratory disease only, whereas a lower degree of ENT1 inhibition (≥0.1x Ki, ≥0.05x IC50) associated with a tolerable level of dyspnoea in both respiratory and cardiovascular disease patients. ENT1 inhibition had no effect in healthy volunteers. Furthermore, physicochemical properties correlative with ENT1 binding were assessed using a set of 1625 diverse molecules. Binding to ENT1 was relatively promiscuous (22% compounds Ki<1μM) especially for neutral or basic molecules, and greater incidence tracked with higher lipophilicity (clogP >5). This study rationalises inclusion of an assessment of ENT1 activity during early safety profiling for programs targeting respiratory disorders.
Collapse
Affiliation(s)
- Lyn Rosenbrier Ribeiro
- Discovery Safety, Drug Safety and Metabolism, AstraZeneca, Cambridge Science Park, Cambridge, United Kingdom.
| | - R Ian Storer
- Discovery Safety, Drug Safety and Metabolism, AstraZeneca, Cambridge Science Park, Cambridge, United Kingdom
| |
Collapse
|