1
|
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
|
2
|
Kubica J, Adamski P, Dobrzycki S, Gajda R, Gąsior M, Gierlotka M, Jaguszewski M, Legutko J, Lesiak M, Navarese EP, Niezgoda P, Ostrowska M, Pawłowski T, Tycińska A, Umińska JM, Witkowski A, Gil R. Cangrelor - Expanding therapeutic options in patients with acute coronary syndrome. Cardiol J 2023; 31:133-146. [PMID: 37964649 PMCID: PMC10919555 DOI: 10.5603/cj.96076] [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: 06/18/2023] [Revised: 08/24/2023] [Accepted: 09/24/2023] [Indexed: 11/16/2023] Open
Abstract
Cangrelor is the only intravenous P2Y12 receptor antagonist. It is an adenosine triphosphate analog that selectively, directly, and reversibly binds to the platelet P2Y12 receptors exerting its antiaggregatory effect. Cangrelor is characterized by linear, dose-dependent pharmacokinetics and rapid onset of action providing potent platelet inhibition exceeding 90%. Cangrelor is rapidly metabolized by endothelial endonucleotidase; thus, its half-life is 2.9 to 5.5 min, and its antiplatelet effect subsides within 60 to 90 min. Data originating from three pivotal cangrelor trials (CHAMPION PLATFORM, CHAMPION PCI, and CHAMPION PHOENIX) indicate that cangrelor reduces the risk of periprocedural thrombotic complications during percutaneous coronary intervention at the expense of mild bleedings. Its unique pharmacological properties allow it to overcome the limitations of oral P2Y12 receptor inhibitors, mainly related to the delayed and decreased bioavailability and antiplatelet effect of these agents, which are often observed in the setting of acute coronary syndrome. Subgroups of patients who could theoretically benefit the most from cangrelor include those in whom pharmacokinetics and pharmacodynamics of oral P2Y12 receptor antagonists are most disturbed, namely patients with ST-segment elevation myocardial infarction, those treated with opioids, with mild therapeutic hypothermia, or in cardiogenic shock. Cangrelor could also be useful if bridging is required in patients undergoing surgery. According to the current guidelines cangrelor may be considered in P2Y12 receptor inhibitor-naïve patients undergoing percutaneous coronary intervention in both acute and stable settings.
Collapse
Affiliation(s)
- Jacek Kubica
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Piotr Adamski
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland.
| | - Sławomir Dobrzycki
- Department of Invasive Cardiology, Medical University of Bialystok, Poland
| | | | - Mariusz Gąsior
- 3rd Department of Cardiology, Silesian Center for Heart Diseases, Medical University of Silesia, Zabrze, Poland
| | - Marek Gierlotka
- Department of Cardiology, Institute of Medical Sciences, University of Opole, Poland
| | | | - Jacek Legutko
- Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Clinical Department of Interventional Cardiology, John Paul II Hospital, Krakow, Poland
| | - Maciej Lesiak
- Chair and 1st Department of Cardiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Eliano P Navarese
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Piotr Niezgoda
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Małgorzata Ostrowska
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Pawłowski
- Department of Cardiology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| | | | - Julia M Umińska
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Adam Witkowski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Robert Gil
- Department of Cardiology, National Medical Institute of the Ministry of Interior and Administration, Warsaw, Poland
| |
Collapse
|
3
|
Filippin KJ, de Souza KFS, de Araujo Júnior RT, Torquato HFV, Dias DA, Parisotto EB, Ferreira AT, Paredes-Gamero EJ. Involvement of P2 receptors in hematopoiesis and hematopoietic disorders, and as pharmacological targets. Purinergic Signal 2020; 16:1-15. [PMID: 31863258 PMCID: PMC7166233 DOI: 10.1007/s11302-019-09684-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
Several reports have shown the presence of P2 receptors in hematopoietic stem cells (HSCs). These receptors are activated by extracellular nucleotides released from different sources. In the hematopoietic niche, the release of purines and pyrimidines in the milieu by lytic and nonlytic mechanisms has been described. The expression of P2 receptors from HSCs until maturity is still intriguing scientists. Several reports have shown the participation of P2 receptors in events associated with modulation of the immune system, but their participation in other physiological processes is under investigation. The presence of P2 receptors in HSCs and their ability to modulate this population have awakened interest in exploring the involvement of P2 receptors in hematopoiesis and their participation in hematopoietic disorders. Among the P2 receptors, the receptor P2X7 is of particular interest, because of its different roles in hematopoietic cells (e.g., infection, inflammation, cell death and survival, leukemias and lymphomas), making the P2X7 receptor a promising pharmacological target. Additionally, the role of P2Y12 receptor in platelet activation has been well-documented and is the main example of the importance of the pharmacological modulation of P2 receptor activity. In this review, we focus on the role of P2 receptors in the hematopoietic system, addressing these receptors as potential pharmacological targets.
Collapse
Affiliation(s)
- Kelly Juliana Filippin
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, 79070-900, Brazil
| | - Kamylla F S de Souza
- Departamento de Bioquímica, Universidade Federal de São Paulo, R. Três de Maio 100, São Paulo, SP, 04044-020, Brazil
| | | | - Heron Fernandes Vieira Torquato
- Departamento de Bioquímica, Universidade Federal de São Paulo, R. Três de Maio 100, São Paulo, SP, 04044-020, Brazil
- Universidade Braz Cubas, Av. Francisco Rodrigues Filho 1233, Mogi das Cruzes, SP, 08773-380, Brazil
| | - Dhébora Albuquerque Dias
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, 79070-900, Brazil
| | - Eduardo Benedetti Parisotto
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, 79070-900, Brazil
| | - Alice Teixeira Ferreira
- Departamento de Biofísica, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, SP, 04023-062, Brazil.
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Laboratório de Biologia Molecular e Culturas Celulares, Av. Costa e Silva, s/n Bairro Universitário, Campo Grande, MS, CEP: 79070-900, Brazil.
| | - Edgar J Paredes-Gamero
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, 79070-900, Brazil.
- Departamento de Bioquímica, Universidade Federal de São Paulo, R. Três de Maio 100, São Paulo, SP, 04044-020, Brazil.
- Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Laboratório de Biologia Molecular e Culturas Celulares, Av. Costa e Silva, s/n Bairro Universitário, Campo Grande, MS, CEP: 79070-900, Brazil.
| |
Collapse
|
4
|
Abstract
The ferric chloride models of arterial thrombosis are useful tools with which to investigate the cellular and molecular mechanisms that contribute to arterial thrombosis. Recent insights have, however, revealed the complex and multifaceted mechanism by which ferric chloride induces thrombus formation. Here, we discuss the strengths and weaknesses of the ferric chloride models of arterial thrombosis. Particular focus is given to the phenotypes of different knockout mice in the ferric chloride models and how these compare to other models with independent modes of initiation. Further, we discuss the relevance of the ferric chloride models to the human pathology of atherothrombotic disease.
Collapse
Affiliation(s)
- Steven P Grover
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill , Chapel Hill, NC, USA
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill , Chapel Hill, NC, USA
| |
Collapse
|
5
|
Tantry U, Chaudhary R, Kubica J, Bliden K, Gurbel PA. Cangrelor for the treatment of patients with Arterial Thrombosis. Expert Opin Pharmacother 2018; 19:1389-1398. [PMID: 30102083 DOI: 10.1080/14656566.2018.1506767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION All oral P2Y12 receptor blockers are associated with some degree of delayed onset and offset of pharmacodynamic (PD) effects in patients with acute coronary syndromes (ACS) undergoing percutaneous coronary intervention (PCI). Although intravenous glycoprotein IIb/IIIa inhibitors are associated with rapid onset of action, they are also associated with delayed offset and other limitations such as elevated bleeding risk and thrombocytopenia. Areas covered: In this review, the authors focus on cangrelor, an intravenous, reversible P2Y12 receptor blocker with fast onset and offset of effects. The authors also describe the pharmacologic effects of cangrelor and its pharmacologic interaction with other P2Y12 receptor inhibitors. Finally, the authors discuss the large-scale clinical trials that compared the efficacy and safety of cangrelor with clopidogrel. Expert opinion: In ACS patients undergoing PCI, cangrelor is most desirable to effectively prevent periprocedural ischemic events and to avoid excessive bleeding. Indeed, any high-risk patient with ST-segment elevation myocardial infraction or patient who is unable to take oral medications is a potential candidate for intravenous cangrelor therapy. Furthermore, stable patients with coronary artery disease, who are considered for ad hoc PCI following coronary angiography, may be considered for treatment with cangrelor to reduce post-PCI thrombotic events.
Collapse
Affiliation(s)
- Udaya Tantry
- a Inova Center for Thrombosis Research and Drug Development , Inova Heart and Vascular Institute , Falls Church , VA , USA
| | - Rahul Chaudhary
- a Inova Center for Thrombosis Research and Drug Development , Inova Heart and Vascular Institute , Falls Church , VA , USA
| | - Jacek Kubica
- b Cardiovascular Institute, Ludwik Rydygier Collegium Medicum , Nicolaus CopernicusUniversity , Bydgoszcz , Poland
| | - Kevin Bliden
- a Inova Center for Thrombosis Research and Drug Development , Inova Heart and Vascular Institute , Falls Church , VA , USA
| | - Paul A Gurbel
- a Inova Center for Thrombosis Research and Drug Development , Inova Heart and Vascular Institute , Falls Church , VA , USA
| |
Collapse
|
6
|
Luo X, Li M, Zhan K, Yang W, Zhang L, Wang K, Yu P, Zhang L. Selective inhibition of TRPM2 channel by two novel synthesized ADPR analogues. Chem Biol Drug Des 2018; 91:552-566. [PMID: 29034580 PMCID: PMC5813235 DOI: 10.1111/cbdd.13119] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 07/04/2017] [Accepted: 08/21/2017] [Indexed: 01/02/2023]
Abstract
Transient receptor potential melastatin-2 (TRPM2) channel critical for monitoring internal body temperature is implicated in the pathological processes such as neurodegeneration. However, lacking selective and potent TRPM2 inhibitors impedes investigation and validation of the channel as a drug target. To discover novel and selective TRPM2 inhibitors, a series of adenosine 5'-diphosphoribose analogues were synthesized, and their activities and selectivity were evaluated. Whole-cell patch-clamp recordings were employed for screen and evaluation of synthesized compounds. Two compounds, 7i and 8a, were identified as TRPM2 inhibitors with IC50 of 5.7 and 5.4 μm, respectively. Both 7i and 8a inhibited TRPM2 current without affecting TRPM7, TRPM8, TRPV1 and TRPV3. These two TRPM2 inhibitors can serve as new pharmacological tools for further investigation and validation of TRPM2 channel as a drug target, and the summarized structure-activity relationship (SAR) may also provide insights into further improving existing inhibitors as potential lead compounds.
Collapse
Affiliation(s)
- Xiao Luo
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - Meng Li
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
| | - Kaiyu Zhan
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Wei Yang
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - KeWei Wang
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
- Department of PharmacologySchool of PharmacyQingdao UniversityQingdaoChina
| | - Peilin Yu
- Department of ToxicologySchool of Public HealthZhejiang UniversityHangzhouZhejiangChina
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| |
Collapse
|
7
|
Medrano MP, Bejarano CA, Battista AG, Venera GD, Bernabeu RO, Faillace MP. Injury-induced purinergic signalling molecules upregulate pluripotency gene expression and mitotic activity of progenitor cells in the zebrafish retina. Purinergic Signal 2017; 13:443-465. [PMID: 28710541 PMCID: PMC5714835 DOI: 10.1007/s11302-017-9572-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023] Open
Abstract
Damage in fish activates retina repair that restores sight. The purinergic signalling system serves multiple homeostatic functions and has been implicated in cell cycle control of progenitor cells in the developing retina. We examined whether changes in the expression of purinergic molecules were instrumental in the proliferative phase after injury of adult zebrafish retinas with ouabain. P2RY1 messenger RNA (mRNA) increased early after injury and showed maximal levels at the time of peak progenitor cell proliferation. Extracellular nucleotides, mainly ADP, regulate P2RY1 transcriptional and protein expression. The injury-induced upregulation of P2RY1 is mediated by an autoregulated mechanism. After injury, the transcriptional expression of ecto-nucleotidases and ecto-ATPases also increased and ecto-ATPase activity inhibitors decreased Müller glia-derived progenitor cell amplification. Inhibition of P2RY1 endogenous activation prevented progenitor cell proliferation at two intervals after injury: one in which progenitor Müller glia mitotically activates and the second one in which Müller glia-derived progenitor cells amplify. ADPβS induced the expression of lin28a and ascl1a genes in mature regions of uninjured retinas. The expression of these genes, which regulate multipotent Müller glia reprogramming, was significantly inhibited by blocking the endogenous activation of P2RY1 early after injury. We consistently observed that the number of glial fibrillary acidic protein-BrdU-positive Müller cells after injury was larger in the absence than in the presence of the P2RY1 antagonist. Ecto-ATPase activity inhibitors or P2RY1-specific antagonists did not modify apoptotic cell death at the time of peak progenitor cell proliferation. The results suggested that ouabain injury upregulates specific purinergic signals which stimulates multipotent progenitor cell response.
Collapse
Affiliation(s)
- Matías P Medrano
- Instituto de Fisiología y Biofísica Prof. Bernardo Houssay (IFIBIO-Houssay), UBA y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro Paladini, UBA-CONICET, Buenos Aires, Argentina
| | - Claudio A Bejarano
- Instituto de Fisiología y Biofísica Prof. Bernardo Houssay (IFIBIO-Houssay), UBA y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Ariadna G Battista
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro Paladini, UBA-CONICET, Buenos Aires, Argentina
| | - Graciela D Venera
- Instituto Universitario Italiano de Rosario (IUNIR), Santa Fe, Argentina
| | - Ramón O Bernabeu
- Instituto de Fisiología y Biofísica Prof. Bernardo Houssay (IFIBIO-Houssay), UBA y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Maria Paula Faillace
- Instituto de Fisiología y Biofísica Prof. Bernardo Houssay (IFIBIO-Houssay), UBA y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.
- IFIBIO-Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Paraguay 2155 7° piso, C1121ABG, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
8
|
Abstract
Antiplatelet therapy with a P2Y12 inhibitor is a key component of treatment for patients with acute coronary syndromes undergoing percutaneous coronary intervention. Before the development of cangrelor (Kengreal, The Medicines Company, Parsippany, NJ), only oral P2Y12 inhibitors were available. Cangrelor is a reversible P2Y12 inhibitor that is administered as an intravenous infusion, and its quick onset and offset make it an appealing option for antiplatelet therapy, particularly for patients who are unable to take oral medications. Although cangrelor struggled to show benefit in early trials, the positive results of the CHAMPION PHOENIX trial led to its approval for use as an adjunct to percutaneous coronary intervention to reduce the risk of periprocedural myocardial infarction, repeat coronary revascularization, and stent thrombosis in patients who have not been treated with another P2Y12 inhibitor and are not being given a glycoprotein IIb/IIIa inhibitor. Cangrelor has also been evaluated as an option for bridging therapy in patients who must discontinue their oral P2Y12 inhibitor before coronary artery bypass grafting. This review of cangrelor will discuss its mechanism of action, its pharmacodynamics and pharmacokinetics, the clinical trial experience, and its potential place in therapy.
Collapse
|
9
|
Abstract
Cangrelor is the first parenteral antagonist of the platelet P2Y12 receptor. This direct-acting antagonist of the platelet P2Y12 receptor should be considered an adjunct to a percutaneous coronary intervention in patients who have not been adequately pretreated with platelet P2Y12 receptor antagonists at the time of the procedure. The use of cangrelor requires transition to an oral platelet P2Y12 receptor antagonist. Transition strategies have been developed on the basis of pharmacologic characteristics of platelet P2Y12 receptor antagonists, results of pharmacodynamic studies, and results from clinical trials. Cangrelor blocks the binding to the platelet P2Y12 receptor of the active metabolite of the thienopyridines, clopidogrel and prasugrel. The active metabolite of thienopyridines is present in blood for a short interval after administration. For this reason, clopidogrel should be administered after cangrelor is stopped. Prasugrel can be administered at the end of the cangrelor infusion or up to 30 min before cangrelor is stopped. Ticagrelor is also a reversible direct-acting antagonist of the platelet P2Y12 receptor. Because there is no interaction between ticagrelor and cangrelor, ticagrelor can be administered before or during the infusion of cangrelor.
Collapse
|
10
|
Abstract
Cangrelor (Kengrexal(®), Kengreal(™)) is an intravenously administered P2Y12 receptor inhibitor. It is direct-acting and reversible, with a very rapid onset and offset of action. The randomized, double-blind, multinational, phase III CHAMPION PHOENIX trial compared the efficacy of intravenous cangrelor with that of oral clopidogrel in patients requiring percutaneous coronary intervention (PCI) for stable angina pectoris, a non-ST-segment elevation acute coronary syndrome or ST-segment elevation myocardial infarction (MI). The primary composite efficacy endpoint of death from any cause, MI, ischaemia-drive revascularization or stent thrombosis in the 48 h following randomization occurred in significantly fewer cangrelor than clopidogrel recipients. The rate of severe or life-threatening non-coronary artery bypass graft-related, GUSTO-defined bleeding at 48 h did not significantly differ between cangrelor and clopidogrel recipients. In conclusion, intravenous cangrelor is an important new option for use in patients undergoing PCI who have not been treated with oral P2Y12 inhibitors.
Collapse
|
11
|
Lechaftois M, Dreano E, Palmier B, Margaill I, Marchand-Leroux C, Bachelot-Loza C, Lerouet D. Another "string to the bow" of PJ34, a potent poly(ADP-Ribose)polymerase inhibitor: an antiplatelet effect through P2Y12 antagonism? PLoS One 2014; 9:e110776. [PMID: 25329809 PMCID: PMC4203827 DOI: 10.1371/journal.pone.0110776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background Neuro- and vasoprotective effects of poly(ADP-ribose)polymerase (PARP) inhibition have been largely documented in models of cerebral ischemia, particularly with the potent PARP inhibitor PJ34. Furthermore, after ischemic stroke, physicians are faced with incomplete tissue reperfusion and reocclusion, in which platelet activation/aggregation plays a key role. Data suggest that certain PARP inhibitors could act as antiplatelet agents. In that context, the present in vitro study investigated on human blood the potential antiplatelet effect of PJ34 and two structurally different PARP inhibitors, DPQ and INO-1001. Methods and results ADP concentrations were chosen to induce a biphasic aggregation curve resulting from the successive activation of both its receptors P2Y1 and P2Y12. In these experimental conditions, PJ34 inhibited the second phase of aggregation; this effect was reduced by incremental ADP concentrations. In addition, in line with a P2Y12 pathway inhibitory effect, PJ34 inhibited the dephosphorylation of the vasodilator stimulated phosphoprotein (VASP) in a concentration-dependent manner. Besides, PJ34 had no effect on platelet aggregation induced by collagen or PAR1 activating peptide, used at concentrations inducing a strong activation independent on secreted ADP. By contrast, DPQ and INO-1001 were devoid of any effect whatever the platelet agonist used. Conclusions We showed that, in addition to its already demonstrated beneficial effects in in vivo models of cerebral ischemia, the potent PARP inhibitor PJ34 exerts in vitro an antiplatelet effect. Moreover, this is the first study to report that PJ34 could act via a competitive P2Y12 antagonism. Thus, this antiplatelet effect could improve post-stroke reperfusion and/or prevent reocclusion, which reinforces the interest of this drug for stroke treatment.
Collapse
Affiliation(s)
- Marie Lechaftois
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Elise Dreano
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Bruno Palmier
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Isabelle Margaill
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Catherine Marchand-Leroux
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Christilla Bachelot-Loza
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Dominique Lerouet
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Cleator JH, Duvernay MT, Holinstat M, Colowick NE, Hudson WJ, Song Y, Harrell FE, Hamm HE. Racial differences in resistance to P2Y12 receptor antagonists in type 2 diabetic subjects. J Pharmacol Exp Ther 2014; 351:33-43. [PMID: 25052834 PMCID: PMC4165026 DOI: 10.1124/jpet.114.215616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/27/2014] [Indexed: 01/21/2023] Open
Abstract
Although resistance to the P2Y12 antagonist clopidogrel is linked to altered drug metabolism, some studies suggest that these pharmacokinetic abnormalities only partially account for drug resistance. To circumvent pharmacokinetic complications and target P2Y12 receptor function we applied the direct P2Y12 antagonist 2-methylthio-AMP (2-methylthioadenosine 5'-monophosphate triethylammonium salt) to purified platelets ex vivo. Platelets were purified from healthy and type 2 diabetes mellitus (T2DM) patients and stimulated with thrombin or the selective protease-activated receptor agonists, protease-activated receptor 1-activating peptide (PAR1-AP), or PAR4-AP. Platelet activation as measured by αIIbβ3 activation, and P-selectin expression was monitored in 141 subjects. Our results demonstrate that, compared with healthy subjects, platelets from diabetic patients are resistant to inhibition by 2-methylthio-AMP, demonstrating P2Y12 pharmacodynamic defects among diabetic patients. Inhibition of thrombin-mediated αIIbβ3 activation by 2-methylthio-AMP was lower in diabetic platelets versus healthy platelets. Subgroup analysis revealed a racial difference in the resistance to 2-methylthio-AMP. We found no resistance in platelets from diabetic African Americans; they were inhibited by 2-methylthio-AMP equally as well as platelets from healthy African Americans. In contrast, platelets from Caucasian patients with diabetes were resistant to P2Y12 antagonism compared with healthy Caucasians. Multivariable analysis demonstrated that other variables, such as obesity, age, or gender, could not account for the differential resistance to 2-methylthio-AMP among races. These results suggest that in addition to altered drug metabolism, P2Y12 receptor function itself is altered in the Caucasian diabetic population. The racial difference in platelet function in T2DM is a novel finding, which may lead to differences in treatment as well as new targets for antiplatelet therapy.
Collapse
Affiliation(s)
- John H Cleator
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Matthew T Duvernay
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Michael Holinstat
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Nancy E Colowick
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Willie J Hudson
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Yanna Song
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Frank E Harrell
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| | - Heidi E Hamm
- Department of Medicine and Division of Cardiovascular Medicine (J.H.C.), Department of Pharmacology (J.H.C., M.T.D., M.H., N.E.C., W.J.H., H.E.H.), and Department of Biostatistics (Y.S., F.E.H.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania (M.H.)
| |
Collapse
|
13
|
Fuentes E, Palomo I. Regulatory mechanisms of cAMP levels as a multiple target for antiplatelet activity and less bleeding risk. Thromb Res 2014; 134:221-6. [PMID: 24830902 DOI: 10.1016/j.thromres.2014.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 04/22/2014] [Accepted: 04/25/2014] [Indexed: 12/19/2022]
Abstract
Platelet activation is a critical component of atherothrombosis. The multiple pathways of platelet activation limit the effect of specific receptor/pathway inhibitors, resulting in limited clinical efficacy. Recent research has confirmed that combination therapy results in enhanced antithrombotic efficacy without increasing bleeding risk. In this way, the best-known inhibitor and turn off signaling in platelet activation is cAMP. In this article we discuss the mechanisms of regulation of intraplatelet cAMP levels, a) platelet-dependent pathway: Gi/Gs protein-coupled receptors, phosphodiesterase inhibition and activation of PPARs and b) platelet-independent pathway: inhibition of adenosine uptake by erythrocytes. With respect to the association between intraplatelet cAMP levels and bleeding risk it is possible to establish that compounds/drugs with pleitropic effect for increased intraplatelet cAMP level could have an antithrombotic activity with less risk of bleeding.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| |
Collapse
|
14
|
Kubica J, Kozinski M, Navarese EP, Tantry U, Kubica A, Siller-Matula JM, Jeong YH, Fabiszak T, Andruszkiewicz A, Gurbel PA. Cangrelor: an emerging therapeutic option for patients with coronary artery disease. Curr Med Res Opin 2014; 30:813-28. [PMID: 24393016 DOI: 10.1185/03007995.2014.880050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES To perform a systematic up-to-date review and critical discussion of potential clinical applications of cangrelor based on its pharmacologic properties and the main findings from randomized clinical studies. METHODS A database search (PubMed, CENTRAL and Google Scholar) by two independent investigators, including proceedings from scientific sessions of ACC, AHA, ESC, TCT and EuroPCR, from January 1998 through December 2013. RESULTS Cangrelor is a potent, intravenous, direct-acting P2Y12 antagonist with rapid onset and quickly reversible action. In contrast to ticagrelor, cangrelor's interaction with thienopiridines requires termination of cangrelor infusion before switching to clopidogrel or prasugrel. According to randomized trials, a cangrelor-clopidogrel combination is relatively safe and more effective than the standard clopidogrel regimen in both urgent and elective percutaneous coronary intervention (PCI) settings, with the advantage of this drug combination fully evident when the universal definition of myocardial infarction is applied. In contrast to available antiplatelet drugs with delayed onset and offset of action, its favorable properties make cangrelor a desirable agent for ad hoc elective PCI, high risk acute coronary syndromes treated with immediate coronary stenting and for bridging those surgery patients who require periprocedural P2Y12 inhibition. Current evidence on cangrelor therapy is limited by the lack of adequately powered studies assessing cangrelor co-administration either with prasugrel or ticagrelor, suboptimal design of some of the trials favoring cangrelor, potentially attenuated benefits with modern stent design, and finally, by the lack of survival advantage. CONCLUSIONS With its pharmacokinetic and pharmacodynamic advantages, allowing consistent and strong P2Y12 inhibition, and with its rapid onset and swift reversal of action devoid of need for an antidote, cangrelor might improve clinical outcomes in clopidogrel-treated patients by reducing ischemic events, while maintaining a favorable safety profile. However, further studies, addressing the safety and efficacy of cangrelor on top of novel oral P2Y12 inhibitors, are warranted.
Collapse
Affiliation(s)
- Jacek Kubica
- Collegium Medicum, Nicolaus Copernicus University , Bydgoszcz , Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|