Next Article in Journal
New Insights into the Role of Glutathione in the Mechanism of Fever
Next Article in Special Issue
Primary Hemostasis in Chronic Liver Disease and Cirrhosis: What Did We Learn over the Past Decade?
Previous Article in Journal
Impact of Pituitary Autoimmunity and Genetic Disorders on Growth Hormone Deficiency in Children and Adults
Previous Article in Special Issue
Platelet Biochemistry and Morphology after Cryopreservation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P2Y12 Inhibition beyond Thrombosis: Effects on Inflammation

by
Alexandre Mansour
1,2,3,
Christilla Bachelot-Loza
1,
Nicolas Nesseler
2,3,4,
Pascale Gaussem
1,5,† and
Isabelle Gouin-Thibault
3,6,*,†
1
Université de Paris, Innovative Therapies in Haemostasis, INSERM 1140, F-75006 Paris, France
2
CHU de Rennes, Department of Anesthesiology Critical Care Medicine and Perioperative Medicine, F-35000 Rennes, France
3
INSERM, CIC 1414 (Centre d’Investigation Clinique de Rennes), Université de Rennes, CHU de Rennes, F-35000 Rennes, France
4
INRA, INSERM, Institut NUMECAN–UMR_A 1341, Université de Rennes, CHU de Rennes, UMR_S 1241, F-35000 Rennes, France
5
AP-HP, Hôpital Européen Georges Pompidou, Department of Biological Hematology, F-75015 Paris, France
6
CHU de Rennes, Department of Biological Hematology, F-35000 Rennes, France
*
Author to whom correspondence should be addressed.
P.G. and I.G.T. share equal last authorship.
Int. J. Mol. Sci. 2020, 21(4), 1391; https://doi.org/10.3390/ijms21041391
Submission received: 29 January 2020 / Revised: 14 February 2020 / Accepted: 15 February 2020 / Published: 19 February 2020
(This article belongs to the Special Issue Molecular and Cellular Basis of Thrombotic Diseases)

Abstract

:
The P2Y12 receptor is a key player in platelet activation and a major target for antithrombotic drugs. The beneficial effects of P2Y12 receptor antagonists might, however, not be restricted to the primary and secondary prevention of arterial thrombosis. Indeed, it has been established that platelet activation also has an essential role in inflammation. Additionally, nonplatelet P2Y12 receptors present in immune cells and vascular smooth muscle cells might be effective players in the inflammatory response. This review will investigate the biological and clinical impact of P2Y12 receptor inhibition beyond its platelet-driven antithrombotic effects, focusing on its anti-inflammatory role. We will discuss the potential molecular and cellular mechanisms of P2Y12-mediated inflammation, including cytokine release, platelet–leukocyte interactions and neutrophil extracellular trap formation. Then we will summarize the current evidence on the beneficial effects of P2Y12 antagonists during various clinical inflammatory diseases, especially during sepsis, acute lung injury, asthma, atherosclerosis, and cancer.

1. Introduction

Nucleotides are universal extracellular signaling molecules, acting as intercellular or autocrine messengers that can be passively released by injured cells or secreted by specific mechanisms. Among their countless physiological or pathological functions, purinergic signaling can regulate hemostasis, thrombosis, and inflammation through the costimulation of various cell types, including platelets, leukocytes, endothelial, and vascular smooth muscle cells [1].
The specific plasma membrane receptors for nucleotides are called P2 receptors and are divided into two subgroups: P2X ligand-gated ion channels and P2Y G-protein (guanine nucleotide-binding protein)-coupled receptors. Eight P2Y receptors have been identified and divided according to their preferred agonist: adenine nucleotides (ADP and ATP) for P2Y1, P2Y11, P2Y12, and P2Y13 receptors; uracil nucleotides (UDP and UTP) for P2Y4 and P2Y6; adenine and uracil nucleotides for P2Y2; and UDP and UDP-glucose for P2Y14 [2,3,4,5].
Adenine nucleotide mediated platelet activation is a critical mechanism in both physiological and pathological hemostasis (including thrombosis), and it involves three platelet P2 receptors (P2Y1, P2Y12, and P2X1, the latter one being an ATP channel). Among the P2Y ADP receptors, P2Y12 is a key receptor and the unique P2 target for clinically approved antiplatelet drugs (herein called P2Y12 inhibitors) [6].
Besides their hemostatic capacities, platelets play an emerging and significant role in regulating inflammatory and immune response. Indeed, they are able to interact with immune cells through membrane exposure of P-selectin and CD40 (cluster of differentiation 40) ligand, and to release inflammatory mediators (cytokines, chemokines) [7,8]. Platelets are also involved in tumorigenesis and in the modulation of tumor microenvironment [9]. Moreover, P2Y12 is expressed in other cell types than platelets, including cancer, immune and vascular cells, and binding of ADP to P2Y12 might activate leukocytes and dendritic cells [2].
Therefore, together with antithrombotic effects, P2Y12 inhibitors might also have a role in modulation of inflammation.
After a brief description of P2Y12 function and its current clinical inhibitors, we will review the effects of P2Y12 inhibition on molecular and cellular mechanisms of inflammation and the current evidence for P2Y12 antagonism in clinical inflammatory diseases and syndromes.

2. P2Y12 Receptors

2.1. Structure of P2Y12 Receptors

The P2Y12 receptor was originally described as a platelet ADP receptor inhibited by thienopyridine antiplatelet agents ticlopidine and clopidogrel. It was further identified by cloning in 2001 by Hollopeter and Zhang and maps to chromosome 3q25.1 [10,11].
P2Y12 is a Gi-coupled seven-transmembrane domain receptor composed of 342 amino acids with a molecular weight of 39kDa. Its structure consists of a seven-transmembrane (7-TM) bundle of α-helices connected by three intracellular and three extracellular loops (EL), and a carboxy-terminal helix (H8) that is parallel to the membrane bilayer on the cytoplasmic side [12]. It contains four extracellular cysteines at positions 17, 97, 175, and 270 that form two disulfide bridges (between the N-terminal domain and EL3 and between EL1 and EL2) which are important sites for receptor expression and potential target for active metabolites of thienopyridines [2,13,14].

2.2. Expression of P2Y12 Receptor

The P2Y12 receptor was originally sought to be exclusively expressed on platelets, with about 400 copies of the P2Y12 receptor per cell, and to a lesser extent in subregions of the brain [11,15]. However, further studies demonstrated that P2Y12 has a wider tissue distribution, being expressed and functional in microglial cells [16], vascular smooth muscle cells [17,18], and on several immune cells including dendritic cells [19], mast cells [20], eosinophils [21], monocytes [22], lymphocytes [23,24] and macrophages [25]. The P2Y12 receptor expression has been also recently reported in osteoclasts [26] as well as in brain and breast cancer cell lines [27]. P2Y12 is not expressed by human endothelial cells of aortic, cerebral or cardiac origin [28]. Still, the expression and function of P2Y12 in other cell types remain poorly investigated. Although P2Y12 is expressed on the plasma membrane of resting platelets, an inducible pool of P2Y12 can also be exposed upon platelet activation by strong agonists [29].

2.3. Role of P2Y12 Rreceptor in Platelet Activation Pathways

Among extracellular nucleotides, ADP (adenosine 5′-diphosphate) plays a key role in platelet function and thrombus formation (Figure 1). It is a weak platelet agonist as it only induces reversible responses, including shape change and reversible aggregation. Platelets express two G protein-coupled receptors for ADP: P2Y1, which is coupled to Gq (G-protein q), and P2Y12, which is coupled to Gi (G-protein i) [3]. Despite the fact that ADP alone is not able to induce the release of platelet dense granules, binding of ADP to its platelet receptors amplifies and sustains the secretion and aggregation induced by other strong agonists [3]. Hence, P2Y12 plays an important role in the stabilization of platelet aggregates induced by other agonists such as thrombin and thromboxane A2 (TXA2), as well as in thrombogenesis in vivo [3,13,18,30,31]. This central position explains both the clinical benefit of P2Y12 antagonists and the bleeding diathesis observed in P2Y12 deficient patients [32,33].
Concomitant stimulation of P2Y1 and P2Y12 receptors by ADP is required to generate normal ADP-induced platelet aggregation [2,4]. Binding of ADP to P2Y1 initiates shape change and early reversible aggregation by mobilization of calcium ions from internal stores through the stimulation of phospholipase C and formation of inositol 1,4,5-trisphosphate (IP3) [5,13,34]. P2Y12 activates a G αi2 G protein subtype that mediates the inhibition of adenylate cyclase and therefore cyclic adenosine monophosphate (cAMP) production, leading to impaired protein kinase A (PKA) activation and a subsequent inhibition of downstream effectors such as vasodilator-stimulated phosphoprotein (VASP) [5,13,35]. This cAMP inhibition is not sufficient to induce platelet aggregation by itself but can promote it [36]. VASP phosphorylation state can be used to monitor the effects of anti P2Y12 therapy [37,38]. Binding of ADP to P2Y12 also induces an activation of two isoforms of phosphoinositide-3-kinase-(PI3K) - p110β and p110γ - through the recruitment of βγ subunits of Gi, which is critical for integrin αIIbβ3 (GpIIbIIIa) activation and stabilization of platelet aggregates, especially induced by thrombin or TXA2 [13,39,40,41,42]. This PI3K activation also amplifies platelet secretion induced by other agonists [13,43]. Finally, P2Y12 supports thrombin generation by amplifying membrane exposition of phosphatidylserine, platelet-derived microparticle formation and collagen-induced exposure of tissue factor (TF). Moreover, it contributes to leukocyte activation induced by surface P-selectin exposure and formation of platelet-leukocyte aggregates [13,44,45,46,47,48]. Formation of coated-platelets—which are a subpopulation of platelets characterized by surface retention of procoagulant proteins, expression of phosphatidylserine and strong prothrombinase activity—is dependent on ADP-induced P2Y12 activation [49,50,51].

2.4. Role of Non-Platelet P2Y12 Receptors

Vascular smooth muscle cells (VSMCs) play a key role in the physiological functions of blood vessels, such as vasoconstriction, vasodilatation and extracellular matrix production. They are also involved in the pathogenesis of vascular diseases, especially atherosclerosis, vascular inflammation and restenosis following angioplasty [52]. P2Y12 receptor expression and function in ADP-induced vessel contraction was first described in human internal mammary artery SMCs in 2004 [18]. Activation of P2Y12 is thought to induce an inflammatory state in VSMCs and correlates with atherosclerotic plaque instability [17,52,53,54]. It is associated with increased monocyte chemoattractant protein 1 (MCP-1) expression and monocyte adhesion [54]. P2Y12 is also implicated in the migration VSMCs through cAMP/PKA signaling pathway associated with actin disassembly and therefore an increase in VSMC motility and migration [55].
P2Y12 is also functionally expressed in microglial cells and can play a role in their activation [16] and in microglia-neuron communications and microglial neuroprotection [56]. Thus, as microglial cells are the primary innate immune cells of the brain and play an important role in the pathophysiology of many brain-based conditions, an implication of P2Y12 may be expected in various diseases including multiple sclerosis, Alzheimer’s disease, traumatic brain and spinal cord injury, and brain cancers [55].
Dendritic cells (DC) are considered the most efficient antigen presenting cells and are able to regulate adaptive immunity by inducing naïve T cell activation and effector differentiation [57]. P2Y12 receptor is expressed in murine DCs and its activation enhances specific T cell activation by increasing antigen endocytosis [19]. P2Y12 is also expressed in human DCs and it has been demonstrated that inhibition of P2Y12 mediated PI3K activation induces an immunosuppressive effect on DCs by decreasing antigen uptake [58].
To date, only few studies have evaluated the role of P2Y12 receptor in the purinergic signaling in leukocytes. High amounts of the P2Y12 receptor mRNA were previously described in lymphocytes as well as in CD34+ progenitor cells [24]. In a clinical study involving cardiologic patients, Diehl et al. showed that P2Y12 mRNA was expressed in leukocytes obtained from leukapheresis and that P2Y12 inhibition by clopidogrel decreased leukocyte CD11b (Mac-1) expression [59]. The P2Y12 expression was also reported in human eosinophils in which P2Y12 activation induced the release of eosinophil peroxidase [21] that could be prevented by clopidogrel [60]. Activation of P2Y12 in macrophages induced cell spreading with formation of lamellipodia, and P2Y12 inhibition alleviated chemotaxis [25]. Micklewright et al. demonstrated in human THP-1 monocytic cells that P2Y12 was expressed and positively regulated P2Y6-mediated intracellular Ca2+ signaling through suppression of adenylate cyclase activity [22]. Finally, Vemulapalli et al. recently showed that functional P2Y12 was expressed by T lymphocytes and that it exerted effects on biological responses of T cells when stimulated [23].
Taken together, these results suggest that P2Y12 inhibitors might target immune cell function and contribute to the modulation of inflammatory and immune responses.
Even though P2Y12 inhibitors are being explored as potential anti-cancer agents, expression of P2Y12 receptor in cancer cells has only been reported by a few studies. P2Y12 has been found in glioma and astrocytoma cells [61,62]. P2Y12 expression has been also described in breast cancer cell lines and was upregulated by cell treatment with cisplatin [63]. In those cells, co-administration of P2Y12 inhibitor with cisplatin resulted in significantly higher cytotoxic response [63].

3. P2Y12 Antagonists

3.1. Thienopyridines

As previously described, P2Y12 activation by ADP has a fundamental role in thrombus formation and stabilization. Hence, antiplatelet agents inhibiting P2Y12 form a cornerstone of therapy for patients at risk of major adverse cardiovascular events (MACE), especially those with acute coronary syndrome undergoing percutaneous coronary intervention, as well as in the secondary prevention of cardiovascular events [33]. Besides their antithrombotic benefits, P2Y12 inhibitors also carry a bleeding risk, in both procedural and non-procedural settings [64,65,66].
Pharmacological description of P2Y12 inhibitors is essential to better understand their potential role in the modulation of inflammation, whether by a platelet dependent and/or an independent mechanism.
The P2Y12 inhibitors involve two classes of drugs: the thienotetrahydropyridines or thienopyridines (clopidogrel, prasugrel, and ticlopidine) and the nucleoside–nucleotide derivatives (cangrelor and ticagrelor) [33,67,68,69]. Pharmacological properties of P2Y12 inhibitors are summarized in Table 1.
Thienopyridines are orally administered inactive prodrugs that develop their antiplatelet effects only after in vivo metabolism to active metabolites [6]. Active metabolites of thienopyridines have a very short half-life and selectively interact with P2Y12 by forming a disulfide bond with cysteine-97 and then irreversibly inhibit ADP-induced activation [6]. Consequently, the inhibitory effect of thienopyridines on platelets lasts for the lifespan of a circulating platelet, which is 8 to 12 days on average [6].
The active metabolites of thienopyridines, and particularly for clopidogrel, are produced by a complex multistep hepatic cytochrome P450 (CYP450) dependent activation [6,77]. Although clopidogrel was shown to exhibit a beneficial effect in the prevention of MACE, it suffered from many drawbacks including slow onset of action, high variability of response, drug resistance due to CYP450 polymorphisms, moderate inhibition of platelet aggregation (33–64%) and drug interactions [6,67,68,77,78]. This led to the development of third generation thienopyridine prasugrel. Unlike ticlopidine and clopidogrel, prasugrel is first metabolized by an intestinal esterase and then undergoes a single step CYP450 dependent activation [6,79]. This allows a faster onset of action, more potent inhibition of platelet aggregation (50–80%), fewer resistance and drug interactions [68,77,80,81]. Consequently, prasugrel is associated with a higher bleeding risk [67].

3.2. Direct P2Y12 Inhibitors

By contrast, several reversible direct-acting P2Y12 receptor antagonists were developed using adenine-nucleotides (ADP or ATP) as the primary structure, that do not require hepatic metabolism [6,33,67,68,69]. Altogether, this pharmacological approach allows a faster, more potent and more predictable platelet inhibitory effect than thienopyridines [6,33].
Ticagrelor is an oral direct-acting P2Y12 inhibitor belonging to the cyclopentyl-triazolo-pyrimidines class. It has a rapid onset of action (2h) and produces a dose-dependent and important inhibition of platelet aggregation (60–90%) [68,71,73]. However, whether ticagrelor exerts a competitive or noncompetitive inhibition of ADP-induced P2Y12 activation is still debated [82,83]. Ticagrelor has one active metabolite (AR–C124910XX) produced by hepatic metabolism, which is at least equipotent and accounts for a third of its antiplatelet effects [73,84]. Even if the offset of action is significantly faster than prasugrel, the more potent platelet inhibitory effect induces a rate of offset still equivalent to clopidogrel, with platelet aggregation returning to pretreatment values after 5 days [68,71].
Ticagrelor exhibits PY12-independent pleiotropic effects. In addition to anti-P2Y12 activity, it has been shown to inhibit equilibrative nucleoside transporter 1 (ENT-1) which is a ubiquitous membrane transport protein, notably responsible for adenine uptake by erythrocytes and platelets [85,86,87]. Ticagrelor inhibits adenine uptake in vitro, inducing increased biological effect of exogenous adenosine and increased plasma levels and biological effects of endogenous adenosine [87]. High plasma levels of adenosine can induce platelet inhibition and coronary vasodilatation, reduce inflammatory response and improve ischemia/reperfusion injuries [87]. Thus, ENT-1 inhibition by ticagrelor may account for some of its biological effect, including inhibition of platelet activation. However, ticagrelor-induced increase in adenosine plasma levels in vivo remains very controversial [87,88,89].
It was also established that, together with an inhibition of platelet ENT1, ticagrelor can act as an inverse agonist at the P2Y12 receptor [86]. This inverse agonist effect was further investigated in the study of Garcia, supporting a new concept of P2Y12 receptor constitutive Gi/o-dependent signaling [90]. This effect was inhibited by ticagrelor but not by the thienopyridine inhibitors and led to an increase of cAMP-dependent signaling pathway compared to resting condition [90]. Moreover, Reiner et al. showed that ticagrelor exhibited endothelial-specific antithrombotic properties by reducing tissue factor expression and activity, independently of P2Y12 and ENT-1 [28]. Finally, a recent study by Lancellotti et al. demonstrated that ticagrelor had a bactericidal activity against antibiotic-resistant gram-positive bacteria in vitro and inhibited biofilm growth and dissemination of bacteria on a mouse model of implanted Staphylococcus aureus–preinfected disks [91].
Cangrelor is the only intravenous P2Y12 inhibitor. It is a direct-acting ATP analogue that selectively and reversibly binds to the P2Y12 receptor in a dose-dependent manner, without requiring metabolism. It achieves a high inhibition of platelet aggregation (>80%) with a rapid onset of action (2 min) and a very fast offset of action (30 to 60 min) due to a half-life of 3 to 6 minutes [76]. It is therefore a therapeutic option for patients with coronary artery disease (CAD) undergoing percutaneous coronary intervention (PCI), and to maintain P2Y12 inhibition when oral therapy is interrupted for any reason [92,93].

4. Effects of P2Y12 Inhibitors on Inflammation: Possible Molecular and Cellular Mechanisms

As described above, ADP-mediated activation of P2Y12 seems to be a common activating pathway in many inflammatory and immune cell types including platelets, leukocytes and dendritic cells.
Hemostasis and inflammation are intimately linked, inducing and amplifying each other and this interconnection contributes to many pathological situations including sepsis, acute lung injury, autoimmune diseases, tumorigenesis and metastasis.
Hence, P2Y12 inhibitors might modulate inflammation through many different cellular and molecular mechanisms, summarized in Figure 2 and further detailed in this section.

4.1. Thrombin Generation

Thrombin generation is a critical step during in vivo thrombogenesis that initiates the formation of fibrin clots and platelet activation. Besides, thrombin can mediate direct effects on inflammatory and immune cells through activation of protease-activated receptors (PARs), and notably PAR-1 [94,95]. Thrombin has been shown to induce the secretion of proinflammatory cytokines interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor α (TNFα), and monocyte chemotactic protein MCP-1 from different cell types including vascular endothelial cells and monocytes [96,97,98]. It also enhances angiogenesis by stimulating the expression of angiogenic growth factors including platelet derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) [99,100]. Finally, thrombin has been shown to support T-cell proliferation and to induce leukocyte recruitment through PAR-1 activation [94,97].
Interplay between thrombin generation and inflammation involves positive feedback as leukocyte activation can also trigger thrombin generation, mainly through the release of neutrophil extracellular traps (NETs) and the expression of tissue factor by monocytes, macrophages and leukocyte-derived microparticles [95,101].
As described above, P2Y12 plays a role in thrombin generation and in thrombus formation in vitro. Accordingly, P2Y12 antagonism has been shown to inhibit thrombin generation through the decreased formation of coated-platelet in vitro and in vivo [50,51,102,103]. Clopidogrel can reduce ex vivo thrombin generation triggered by low concentrations of TF in rodent platelet rich plasma [104]. In a murine experimental endotoxemic model, clopidogrel was recently shown to reduce TF expression on leukocytes [105]. Finally, ticagrelor inhibited TF expression and activity in human aortic endothelial cells, independently of P2Y12 and ENT-1, and decreased thrombosis and endothelial TF expression in a photochemical mouse model of arterial thrombosis [28].

4.2. Release of Inflammatory Mediators

Inflammatory mediators, including cytokines, chemokines and growth factors are key modulators of acute and chronic inflammatory processes. Most of them, including IL-1, IL-6, IL-8 and TNFα are essentially produced by macrophages, monocytes and lymphocytes. They play a critical role in the positive feedback between hemostasis and inflammation [95,106]. Indeed, they induce proliferation, activation and recruitment of leukocytes, but also TF expression on cell surface of mononuclear and endothelial cells, as well as platelet activation [95,106,107,108].
Upon activation, platelets secrete a great amount of soluble mediators from their granules, with potent procoagulant and proinflammatory effects [109,110]. Cytokines and chemokines released by platelets include IL-1β, CD40L, platelet factor 4 (PF4, CXCL4), RANTES (CCL5), βTG (CXCL7), and IL-8 (CXCL8) [106,109,110]. These mediators play an important role in paracrine activation of platelets and in immune cell activation, proliferation and chemotaxis [106]. Notably, soluble CD40L activates platelets, induces IL-6 of and monocyte chemoattractant protein (MCP-1) secretion and enhances TF expression by monocytes, leading to increased thrombin generation [111,112].
P2Y12 inhibition by clopidogrel or by gene knockout in murine models of abdominal sepsis or lipopolysaccharide (LPS)-induced inflammation has been shown to decrease proinflammatory mediator levels in plasma, notably IL-6, TNFα, CCL4 (MIP-1β) and IL-1β [48,105,113]. This is consistent with the reduction in IL-6 and TNFα levels, upon clopidogrel treatment, in a rat model of LPS-induced inflammation [114]. In a human experimental model of intravenous LPS-induced inflammation, ticagrelor and clopidogrel were associated with a significant reduction of IL-6, TNFα and CCL2, and an additional reduction of IL-8 was obtained with ticagrelor [115]. However, anti-inflammatory effect of clopidogrel is inconsistently reported in clinical trials enrolling patients with acute coronary syndromes or stable coronary artery disease, with a reduction of CRP and TNFα [116]. Still, the recently published double-blinded randomized XANTHIPPE study comparing ticagrelor to placebo in pneumonia, demonstrated a significant reduction in plasma IL-6 levels in the ticagrelor group [113].

4.3. Platelet-Leukocyte Interactions and Formation of Neutrophil Extracellular Traps (NETs)

The pathophysiology of platelet-leukocyte interaction has recently been extensively reviewed by Rossaint et al. [117]. Resting circulating platelets act as sentinels in the blood. Upon activation, degranulation and membrane expression of platelet receptors enable physical interactions between platelets and leukocytes, especially neutrophils, forming so-called platelet-leukocyte aggregates (PLAs) [117,118,119]. This interaction is critical in both thrombosis and inflammatory situations. It allows leukocyte activation and recruitment to sites of inflammation and induces the release of proinflammatory mediators, reactive oxygen species (ROS) and neutrophil extracellular traps (NETs) by neutrophils [117,118,119].
The first interaction between platelets and leukocytes is established between P-selectin (CD62P) on activated platelets and PSGL-1, constitutively expressed on the surface of neutrophils, monocytes, dendritic cells, and subclasses of lymphocytes. CD62P/PSGL-1 interaction induces the activation of αMβ2 (Mac-1) integrin on leukocyte surface. Platelet leukocyte interaction is then stabilized by direct-binding of Mac-1 to platelet GPIbα and indirect binding to activated platelet αIIbβ3 (GPIIbIIIa) via fibrinogen [117,118,119].
NETs are large, web-like extracellular chromatin structures that are released by neutrophils upon various activating stimuli, including exogenous microorganisms (bacteria, fungi, virus, and parasites), immune complexes and activated platelets [120]. NETs can trap, neutralize and kill microorganisms, but also activate platelets and promote coagulation [121]. They are considered as critical players in many inflammatory conditions, especially sepsis-induced organ injuries, autoimmunity, tumorigenesis and metastasis [120]. NETs also contribute to thrombogenesis by forming a mesh with platelets and fibrin and accumulating coagulation factors such as TF [118,120,122].
The crosstalk between activated platelets and neutrophils is a critical regulator of NETosis [123,124]. Platelet-neutrophil direct binding is an essential step in platelet-driven NETosis and is mainly mediated by P-selectin/PSGL-1 and GPIbα/Mac-1 interactions [120,123,124,125]. NET formation is further stimulated by platelet-released soluble mediators including high mobility group box 1 (HMGB1), PF4 and RANTES [120,123,124,125].
P2Y12 inhibition by clopidogrel, prasugrel, ticagrelor and cangrelor has been consistently associated with inhibition of platelet-leukocyte interaction and platelet P-selectin expression in animal models of inflammation and in vitro studies [48,105,126,127,128,129]. In a human experimental model of intravenous LPS-induced inflammation, both ticagrelor and clopidogrel were associated with a significant reduction of platelet-monocyte aggregates [115]. Moreover, reduction of PLAs and P-selectin expression by clopidogrel was reported in clinical studies of patients with acute coronary syndromes and atherosclerotic vascular disease [130,131]. Finally, the recent XANTHIPPE study demonstrated a reduction in PLAs in the ticagrelor group compared to placebo [113]. To date, no study reported a significant modulation of NETosis by P2Y12 inhibitors, although ticagrelor was recently shown to reduce in vitro NET-induced platelet aggregation, secretion and expression of P-selectin [113,121].

4.4. Adenosine Mediated Effects

The mechanisms of adenosine signaling on immune cells has been extensively reviewed in a recent work by Vigano et al. [132]. Adenosine exerts a powerful modulatory effect on inflammation and innate immune responses through the activation of four different receptors (A1, A2A, A2B and A3) which are expressed in the majority of immune cells, including neutrophils, lymphocytes, macrophages, mast cells and dendritic cells. Adenosine principally induces an anti-inflammatory phenotype characterized by a decreased ability to release inflammatory mediators, an inhibition of immune cell activation, proliferation and chemotaxis. Therefore, adenosine acts as a regulator of immune cells that aims at preserving host integrity and promotes the resolution of inflammation. Moreover, adenosine is a potent inhibitor of platelet aggregation and adenosine receptor agonists have been shown to potentiate antiplatelet effects of P2Y12 antagonists [87,133,134]. Thus, adenosine might have a beneficial effect in the management of severe inflammatory disorders including sepsis.
As described above, ticagrelor has been shown to increase plasma levels of adenosine by inhibiting ENT-1. To date, no study demonstrated an adenosine-mediated anti-inflammatory effect of ticagrelor, although ticagrelor was shown to increase the in vitro inhibitory effect of exogenous adenosine on platelet aggregation [87].

5. Current Evidence for P2Y12 Inhibition in Clinical Inflammatory Diseases and Syndromes

P2Y12 is now considered as a potential target in several inflammatory diseases, including sepsis, asthma, atherosclerosis and cancer. Main clinical studies evaluating the effects of P2Y12 inhibition in those inflammatory diseases are reported in Table 2 and will be further discussed in this section.

5.1. Sepsis and Sepsis-Induced Acute Lung Injury (ALI)

Sepsis is defined as a life-threatening organ dysfunction that is caused by a dysregulated systemic inflammatory and immune host response to infection [145]. The mechanisms underlying the excessive inflammation in sepsis have been extensively reviewed by van der Poll et al. [146]. To summarize, sepsis-induced organ dysfunctions result from the interplay between uncontrolled activation of the complement, coagulation, and inflammatory systems. Platelet-mediated inflammatory response is now recognized as a critical player in the pathophysiology of sepsis, and is especially involved in sepsis-induced experimental acute lung injury (ALI) and clinical life-threatening acute respiratory distress syndrome (ARDS) through several mechanisms, including pathogen sensing, release of inflammatory mediators, recruitment and activation of immune cells and thrombosis [109,147,148,149,150,151].
First report of the potential benefits of P2Y12 inhibition in sepsis came from observational studies of patients receiving antiplatelet agents. A study of 224 consecutive patients admitted for community acquired pneumonia showed lower use of intensive care unit and shorter stay in hospital in patients receiving antiplatelet agents (aspirin and/or thienopyridines) for at least 6 months compared with age-matched controls [135]. More evidence came from a post hoc analysis of the PLATO trial, in which ticagrelor was shown to significantly reduce death from vascular causes, myocardial infarction, or stroke compared to clopidogrel in 18,624 patients with ACS [152]. This analysis on 18,421 patients revealed that, compared to clopidogrel, ticagrelor was associated with a lower mortality risk following pulmonary events and sepsis in acute coronary syndrome [136]. Finally, a recent observational study including 683,421 patients hospitalized for sepsis, showed a lower risk of mortality in patients receiving antiplatelet agents before admission [137]. Using a nested-control study design in 372,748 patients, they showed that both aspirin and P2Y12 inhibitors were associated with a lower adjusted risk of mortality [137].
Those findings were further confirmed in experimental animal models of endotoxemia and abdominal sepsis. In a rat model of LPS-induced endotoxemia, clopidogrel inhibited IL-6 and TNFα secretion and reduced lung and liver histological injuries [114]. Liverani et al. demonstrated in a mouse model of intra-abdominal sepsis that clopidogrel-treated and P2Y12 null mice were refractory to sepsis-induced lung injury and exhibited a decrease in platelet activation, platelet-leukocyte aggregation and release of inflammatory cytokines [48]. Those results were confirmed in another mouse model of intra-abdominal sepsis in which ticagrelor reduced histological findings of sepsis-induced lung injury, pulmonary infiltration of neutrophils, formation of PLAs and platelet activation [126]. To our knowledge, only one experimental study on LPS-treated mice demonstrated a benefit of ticagrelor on mortality [113].
The XANTHIPPE trial (Examining the Effect of Ticagrelor on Platelet Activation, Platelet-Leukocyte Aggregates, and Acute Lung Injury in Pneumonia), was the first double-blinded placebo-controlled randomized study to evaluate the effect of ticagrelor on inflammation, platelet activation and lung function in 60 patients with community or hospital-acquired pneumonia [113]. Ticagrelor administration to patients within 48h of pneumonia diagnosis demonstrated anti-inflammatory effect with reduced PLAs in circulation, lowered IL-6 levels and improved lung function with a decrease in supplemental oxygen requirements [113].

5.2. Asthma

Platelet activation is strongly involved in the pathogenesis of allergic asthma, including bronchial hyperresponsiveness and airway wall inflammation and remodeling [153,154,155]. Among proposed mechanisms, platelet P2Y12 pathway seems to play an important role. In a study of murine leukotriene E4-induced asthma, clopidogrel treatment and P2Y12 gene knock-out reduced IL-13 levels and airway eosinophilia and inflammation [20]. The underlying mechanism remains unknown. Two randomized trials evaluated the effect of P2Y12 inhibition in patients with asthma. In a double-blind placebo-controlled crossover trial of prasugrel in 40 patients with aspirin-exacerbated respiratory disease, prasugrel did not attenuate aspirin-induced symptoms and failed to decrease PLA levels and mast cell activation [138]. Finally, the PRINA (effect of prasugrel on bronchial hyperreactivity and on markers of inflammation in patients with chronic asthma) trial, in which 26 asthmatic patients were randomly and blindly allocated to prasugrel or placebo for 15 days followed by a 15-day wash-out and a cross-over, showed that P2Y12 inhibition by prasugrel significantly decreased airway hyperresponsiveness with improvement of forced expiratory volume [139].

5.3. Atherosclerosis

Atherosclerosis is a chronic inflammatory vascular disease involving cellular and molecular interactions between platelets, endothelial cells, VSMCs and monocytes, interactions that deeply involve the P2Y12 receptor [156,157,158].
The role of P2Y12 in atherosclerosis has been well documented in experimental models. In a mouse model of atherosclerosis by Apolipoprotein E (ApoE) gene knock-out, genetic P2Y12 deletion was associated with reduced lesion area, increased fibrous content at the plaque site and decreased inflammatory cell infiltration [159]. Likewise, clopidogrel and ticagrelor limited the progression of late atherosclerotic lesion and promoted plaque stability in ApoE knockout mice [160,161,162,163]. Unfortunately, differential analysis of the role of platelet and non-platelet P2Y12 receptors was not possible with these studies. Therefore, a study using P2Y12 gene knockout and bone marrow transplantation in ApoE null mice demonstrated that, compared to vessel wall P2Y12, platelet P2Y12 had no effect on early atheroma formation, implying additional role of VSMC P2Y12 receptors [164]. Further studies are necessary to clarify the extent to which vessel wall and platelet P2Y12 influence early and late atherogenesis.

5.4. Cancer—Tumor Growth and Metastasis

The recent advances in the knowledge of cancer have shown that the biology of cancer has evolved from a tumor-centered view to a concept that places cancer cells within a cell network, including fibroblasts, vascular cells and inflammatory immune cells, that form the tumor microenvironment (TME) [165]. Modulation of inflammation within the TME is a critical player during all stages of tumorigenesis and involves reciprocal interactions between cancer cells and surrounding inflammatory cells. These interactions can promote tumor growth by direct proliferative effect on tumor cells and by inducing immunosuppression [165].
The role of platelets in all steps of tumorigenesis including tumor growth, tumor cell extravasation and metastasis has been extensively studied and is now well-recognized [9,166]. Activated platelets not only secrete PF4 but also lipids, microRNAs and numerous growth factors, including TGFβ (transforming growth factor β) and VEGF, which favor tumor cell proliferation, metastasis and angiogenesis. Moreover, the interaction between platelets and cancer cells is largely described in the literature as a mechanism favoring tumor- cell induced platelet activation (TCIPA), and inducing a “shield” of platelets coating the tumor cells and protecting them from the immune response [9].

5.4.1. In Vitro and Preclinical Studies on P2Y12 Inhibition in Cancer

Consequently, most of published data have evidenced a net anti tumoral effect of antiplatelet agents, including aspirin, cilostazole and P2Y12 inhibitors. The rationale to focus on anti-P2Y12 agents came recently first from the discovery of ADP secretion by tumor cells. For instance, Cho et al. nicely showed the secretion of ADP by ovarian cancer cells [167]. Therefore, platelets can be activated directly by tumor cells via cell-to-cell interactions, and indirectly via ADP secreted by tumor cell. Second is the expression of P2Y12 receptors in cells other than platelets, such as VSMCs and tumor cells such as breast cancer cell lines [27]. In Cho’s study, the use of ticagrelor reduced tumor growth by 60% compared to aspirin and 75% versus placebo, inhibited proliferation as assessed by Ki67 positivity, and increased apoptosis of ovarian cancer cells. In P2Y12−/− mice, the growth of ovarian tumors was reduced by over 85% compared to wild-type animals, but not in P2Y1−/− mice, pointing to the essential role of P2Y12 between the two ADP receptors. Finally, the invalidation of ectopyrase gene (CD39, which catabolizes ADP) in cancer cells increased platelet-related cancer cell proliferation.
This finding is not restricted to ovarian tumor cell lines. Gareau et al. showed the beneficial effect of ticagrelor in limiting the interaction between platelets and human mammary carcinoma cells [168].
In other preclinical studies, ticagrelor used at clinical dose (10 mg/kg) inhibited metastasis and improved survival in a mouse model of melanoma metastasis [169].
In a Lewis lung carcinoma spontaneous metastatic mouse model, P2Y12 deficiency reduced pulmonary metastasis and reduced the ability of platelets to secrete active TGFβ1, thus limiting epithelial to mesenchymal transition and invasiveness of tumor cells [170]. Same findings were observed with B16 melanoma metastasis model.
Consequently, targeted therapies start to emerge in the aim to prevent platelet-tumor cell interactions. The tumor-homing peptide Cys-Arg-Glu-Lys-Ala (CREKA) that targets fibrin-fibronectin complexes found on the tumor stroma and vessel wall was linked to ticagrelor. CREKA-ticagrelor inhibited platelet-induced migration of 4T1 tumor cells and prevented tumor-platelet interaction. In vivo, it suppressed lung metastasis in a mouse model injected with breast cancer cells [171]. This finding opens the way to innovative antimetastatic agents.

5.4.2. Are anti P2Y12 Agents Protective or at Risk for Increased Cancer-Related Events?

Numerous studies analyzing the potential effect of antiplatelet agents on cancer converge to a protective effect of low dose aspirin on cancer-related mortality. For instance, in the meta-analysis of Algra and Rothwell including 17 studies, regular use of aspirin was protective from colorectal cancer ([OR] 0.62, 95% CI 0.58–0.67, p < 0.0001) [172]. Given that COX1 inhibition is predominant over COX2 inhibition at low dose aspirin, this effect is likely to result primarily from the antiplatelet effect of aspirin.
By contrast, results from randomized clinical trials on anti P2Y12 agents are conflicting. Whereas CAPRIE and CHARISMA studies of clopidogrel versus aspirin did not report increased cancer development, some data from trials using prolonged anti P2Y12 treatment showed increased rates of cancer-related mortality [173,174]. TRITON-TIMI 38 trial of prasugrel compared to clopidogrel on top of aspirin for 6 to 15 months showed a significantly accelerated cancer progression and increased risk of cancer death in the prasugrel group, particularly with breast, colorectal and prostate cancers [175]. One explanation for this apparent paradoxical effect was that the more potent antiplatelet effect of prasugrel brought more events to medical attention and to an increased number of diagnosed cancers. However, results were different in the TRILOGY trial with no difference in cancer frequency between clopidogrel and prasugrel groups after a median follow-up of 17 months [176]. Clopidogrel and ticagrelor given more than 12 months after drug-eluting stenting in the DAPT trial showed a significant increase in cancer-related deaths [177]. However, deaths related to cancer in this study were relatively low in number. Also concerning ticagrelor, PEGASUS-TIMI 54 trial showed an enhanced cancer risk of ticagrelor administered beyond 1 year, whereas PLATO was negative [152,178,179]. Interestingly Raposeiras-Roubin et al. performed a retrospective study on 4229 consecutive acute coronary syndrome patients with a median follow up of 46 months [140]. They found that ticagrelor resulted in a lower cancer risk than clopidogrel without difference between clopidogrel and prasugrel. Noteworthy, only 311 patients were diagnosed with cancer during the follow up (incidence 2.1 per 100 people per year) and ticagrelor-receiving population was 459 versus 3530 with clopidogrel.
Overall, these clinical randomized trials do not include an untreated comparator arm, and are not powered to detect differences in cancer-related events or mortality. Consequently, the Food and Drug Administration (FDA) reported a two trial-level that “rejected the hypothesis of cancer association in patients on dual anti platelet therapy with clopidogrel, that is, the adverse mortality findings in the DAPT trial were not confirmed” [180]. Moreover, the FDA Adverse Event Reporting system is probably unreliable for adequate assessment of cancer risk during antiplatelet treatment as associated cancers might be unreported and/or missed [181].
The evidence for no cancer risk with P2Y12 inhibitors mostly stems from meta-analysis and cohort studies. The meta-analysis of Kotronias et al included nine studies with more than 282,000 participants [182]. When compared with standard aspirin or placebo, the thienopyridines clopidogrel and prasugrel were not associated with cancer mortality and event rate. The study concluded that there was insufficient evidence to suggest an association between thienopyridine exposure and increased risk of cancer event rate or mortality.
The question of the duration of treatment was also addressed in cohort studies. Leader et al showed a lower risk of cancer in subjects exposed to aspirin compared to non-users, with or without clopidogrel, on long-term follow-up [141]. In a large cohort of 10,359 colorectal cancer, 17,889 breast cancer, and 13,155 prostate cancer patients, Hicks et al evaluated the post-diagnostic use of clopidogrel and cancer-specific mortality during an average follow-up of 5 years [142]. Overall, there was no increase in the rate of cancers in patients receiving clopidogrel, after adjustment for potential confounders. Finally, the meta-analysis of Elmariah et al including more than 48,000 patients from six randomized trials confirmed the absence of impact of prolonged clopidogrel on top of aspirin on mortality or cancer [143]. More recently, Rodriguez-Miguel et al showed in 15,491 cases of colorectal cancer versus 60,000 controls, that low-dose aspirin was associated with a reduced risk of colorectal cancer incidence in patients receiving treatment for more than one year [144]. Same reduction of 20 to 30% was found for clopidogrel alone or in combination with aspirin. In short-term users, there was on the contrary an increased risk for patients on clopidogrel and aspirin. Again, the hypothesis raised was an increased incidence of gastro-intestinal bleedings that led to a greater number of colonoscopies and early diagnosis.
Altogether, if it is challenging to compare the effects of antiplatelet agents on cancer-related death in studies designed to analyze adverse cardiovascular-related events, a head-to-head comparison between molecules is also questionable because their pharmacology differs. The thienopyridine clopidogrel has a less predictable effect than prasugrel or ticagrelor. Clopidogrel is indeed less effective in a subset of around 30% of patients, especially those carrying loss-of-function 2C19*2 variant [78]. Besides, ticagrelor not only has more predictable pharmacokinetics than clopidogrel but has been shown to exert an anti-inflammatory effect due to an increased adenosine extracellular concentration. Finally, most studies have investigated the effect of anti P2Y12 agents on top of aspirin and it is difficult to conclude for one drug over the others.

6. Conclusions

P2Y12-mediated nucleotide signaling is now considered to be a critical player in inflammatory response. Besides platelets, P2Y12 is expressed in many immune and vascular cells and has been shown to modulate inflammatory processes, including the release of inflammatory mediators, platelet–leukocyte interactions, and thrombosis. In addition to their antithrombotic properties, P2Y12 inhibitors can therefore be considered to have valuable pharmacological targets for inflammation, and the beneficial effects of anti-P2Y12 drugs have been reported for several experimental and clinical inflammatory diseases, including sepsis, acute lung injury, asthma, atherosclerosis, and cancer. Still, evidence is missing on the exact role of nonplatelet P2Y12 receptors and on how to distinguish between the P2Y12-mediated and adenosine-mediated effects of ticagrelor. More prospective controlled clinical studies are warranted to determine the potential benefits of P2Y12 inhibition therapy for inflammatory diseases, and pharmacological strategies should be further developed to overcome potential P2Y12-associated bleeding risks.

Author Contributions

All listed authors wrote the MS and prepared the figures. All authors have read and agreed to the published version of the manuscript.

Funding

We thank Promex Stiftung für die Forschung for their financial support.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the review or in the writing of the manuscript.

Abbreviations

ACSAcute coronary syndrome
ADPAdenosine diphosphate
ALIAcute lung injury
APCAntigen-presenting cells
ApoEApolipoprotein E
ARDSAcute respiratory distress syndrome
ATPAdenosine triphosphate
CADCoronary artery disease
cAMPCyclic adenosine monophosphate
CD40Cluster of differentiation 40
COXCyclooxygenase
CREKATumor-homing peptide Cys-Arg-Glu-Lys-Ala
CRPC-reactive protein
CYP450Cytochrome P450
DCDendritic cells
ELExtracellular loop
ENT-1Equilibrative nucleoside transporter 1
FDAFood and Drug Administration
GiG-protein i
G-proteinGuanine nucleotide-binding protein
GqG-protein q
GPIIbIIIaGlycoprotein αIIbβ3
HMGB1High-mobility group box 1
ILInterleukin
IP3Inositol 1,4,5-trisphosphate
LPSLipopolysaccharide
MACEMajor adverse cardiovascular event
MCP-1Monocyte chemoattractant protein 1
NETsNeutrophil extracellular traps
PARProtease-activated receptors
PCIPercutaneous coronary intervention
PDGFPlatelet-derived growth factor
PF4Platelet factor 4
PI3KPhosphoinositide-3-kinase
PKAProtein kinase A
PLAsPlatelet leukocyte aggregates
RANTESRegulated on activation, normal T-cell-expressed and -secreted
ROSReactive oxygen species
TCIPATumor-cell-induced platelet activation
TFTissue factor
TGFTransforming growth factor
TMETumor microenvironment
TNFTumor necrosis factor
TXA2Thromboxane A2
VASPVasodilator-stimulated phosphoprotein
VEGFVascular endothelial growth factor
VSCMsVascular smooth muscle cells

References

  1. Burnstock, G.; Ralevic, V. Purinergic Signaling and Blood Vessels in Health and Disease. Pharmacol. Rev. 2013, 66, 102–192. [Google Scholar] [PubMed]
  2. von Kügelgen, I. Pharmacology of P2Y receptors. Brain Res. Bull. 2019, 151, 12–24. [Google Scholar] [CrossRef] [PubMed]
  3. Cattaneo, M. P2Y12 receptors: Structure and function. J. Thromb. Haemost. 2015, 13, 10–16. [Google Scholar] [CrossRef] [PubMed]
  4. Gachet, C. P2Y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells. Purinergic Signal. 2012, 8, 609–619. [Google Scholar] [CrossRef]
  5. Hechler, B.; Gachet, C. The P2 Receptors. Platelets in Thrombotic and Non-Thrombotic Disorders; Pathophysiology, Pharmacology and Therapeutics: An Update; Springer: Cham, Switzerland, 2017; pp. 187–202. [Google Scholar]
  6. Baqi, Y.; Müller, C.E. Antithrombotic P2Y12 receptor antagonists: Recent developments in drug discovery. Drug Discov. Today 2019, 24, 325–333. [Google Scholar] [CrossRef]
  7. Hechler, B.; Gachet, C. Purinergic Receptors in Thrombosis and Inflammation. Arterioscler. Thromb. Vasc Biol. 2015, 35, 2307–2315. [Google Scholar] [CrossRef] [Green Version]
  8. Thomas, M.R.; Storey, R.F. The role of platelets in inflammation. Thromb. Haemost. 2015, 114, 449–458. [Google Scholar]
  9. Palacios-Acedo, A.; Mège, D.; Crescence, L.; Dignat-George, F.; Dubois, C.; Panicot-Dubois, L. Platelets, Thrombo-Inflammation, and Cancer: Collaborating With the Enemy. Front. Immunol. 2019, 10, 1805. [Google Scholar] [CrossRef] [Green Version]
  10. Zhang, F.L.; Luo, L.; Gustafson, E.; Lachowicz, J.; Smith, M.; Qiao, X.; Liu, Y.H.; Chen, G.; Pramanik, B.; Laz, T.M.; et al. ADP Is the Cognate Ligand for the Orphan G Protein-coupled Receptor SP1999. J. Biol. Chem. 2000, 276, 8608–8615. [Google Scholar] [CrossRef] [Green Version]
  11. Hollopeter, G.; Jantzen, H.M.; Vincent, D.; Li, G.; England, L.; Ramakrishnan, V.; Yang, R.B.; Nurden, P.; Nurden, A.; Julius, D.; et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001, 409, 202–207. [Google Scholar] [CrossRef] [Green Version]
  12. Zhang, K.; Zhang, J.; Gao, Z.G.; Zhang, D.; Zhu, L.; Han, G.; Moss, S.M.; Paoletta, S.; Kiselev, E.; Lu, W.; et al. Structure of the human P2Y12 receptor in complex with an antithrombotic drug. Nature 2014, 509, 115–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cattaneo, M. The platelet P2 receptor. In Platelets, 4th ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 259–277. [Google Scholar]
  14. von Kügelgen, I. Pharmacological profiles of cloned mammalian P2Y-receptor subtypes. Pharmacol. Therapeut. 2006, 110, 415–432. [Google Scholar] [CrossRef] [PubMed]
  15. Ohlmann, P.; Lecchi, A.; El-Tayeb, A.; Müller, C.E.; Cattaneo, M.; Gachet, C. The platelet P2Y(12) receptor under normal and pathological conditions. Assessment with the radiolabeled selective antagonist [(3)H]PSB-0413. Purinerg. Signal. 2012, 9, 59–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Haynes, S.E.; Hollopeter, G.; Yang, G.; Kurpius, D.; Dailey, M.E.; Gan, W.B.; Julius, D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 2006, 9, 1512–1519. [Google Scholar] [CrossRef] [PubMed]
  17. Rauch, B.H.; Rosenkranz, A.C.; Ermler, S.; Böhm, A.; Driessen, J.; Fischer, J.W.; Sugidachi, A.; Jakubowski, J.A.; Schrör, K. Regulation of Functionally Active P2Y12 ADP Receptors by Thrombin in Human Smooth Muscle Cells and the Presence of P2Y12 in Carotid Artery Lesions. Arterioscler. Thromb. Vasc Biol. 2010, 30, 2434–2442. [Google Scholar] [CrossRef] [Green Version]
  18. Wihlborg, A.K.; Wang, L.; Braun, O.; Eyjolfsson, A.; Gustafsson, R.; Gudbjartsson, T.; Erlinge, D. ADP Receptor P2Y 12 Is Expressed in Vascular Smooth Muscle Cells and Stimulates Contraction in Human Blood Vessels. Arterioscler. Thromb. Vasc Biol. 2004, 24, 1810–1815. [Google Scholar] [CrossRef] [Green Version]
  19. Addi, A.; Cammarata, D.; Conley, P.B.; Boeynaems, J.M.; Robaye, B. Role of the P2Y 12 Receptor in the Modulation of Murine Dendritic Cell Function by ADP. J. Immunol. 2010, 185, 5900–5906. [Google Scholar] [CrossRef] [Green Version]
  20. Paruchuri, S.; Tashimo, H.; Feng, C.; Maekawa, A.; Xing, W.; Jiang, Y.; Kanaoka, Y.; Conley, P.; Boyce, J.A. Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor. J. Exp. Med. 2009, 206, 2543–2555. [Google Scholar] [CrossRef] [Green Version]
  21. Muniz, V.S.; Baptista-Dos-Reis, R.; Benjamim, C.F.; Mata-Santos, H.A.; Pyrrho, A.S.; Strauch, M.A.; Melo, P.A.; Vicentino, A.R.; Silva-Paiva, J.; Bandeira-Melo, C.; et al. Purinergic P2Y12 Receptor Activation in Eosinophils and the Schistosomal Host Response. PLoS ONE 2015, 10, e0139805. [Google Scholar] [CrossRef] [Green Version]
  22. Micklewright, J.; Layhadi, J.; Fountain, S. P2Y12receptor modulation of ADP-evoked intracellular Ca2+signalling in THP-1 human monocytic cells. Brit. J. Pharmacol. 2018, 175, 2483–2491. [Google Scholar] [CrossRef] [Green Version]
  23. Vemulapalli, H.; Albayati, S.; Patwa, V.C.; Tilley, D.G.; Tsygankov, A.Y.; Liverani, E. ADP exerts P2Y12 -dependent and P2Y12 -independent effects on primary human T cell responses to stimulation. J. Cell Commun. Signal. 2019, 1–16. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, L.; Jacobsen, S.W.; Bengtsson, A.; Erlinge, D. P2 receptor mRNA expression profiles in human lymphocytes, monocytes and CD34+ stem and progenitor cells. BMC Immunol. 2004, 5, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kronlage, M.; Song, J.; Sorokin, L.; Isfort, K.; Schwerdtle, T.; Leipziger, J.; Robaye, B.; Conley, P.; Kim, C.H.; Sargin, S.; et al. Autocrine Purinergic Receptor Signaling Is Essential for Macrophage Chemotaxis. Sci. Signal. 2010, 3, 55. [Google Scholar] [CrossRef]
  26. Su, X.; Floyd, D.H.; Hughes, A.; Xiang, J.; Schneider, J.G.; Uluckan, O.; Heller, E.; Deng, H.; Zou, W.; Craft, C.S.; et al. The ADP receptor P2RY12 regulates osteoclast function and pathologic bone remodeling. J. Clin. Investig. 2012, 122, 3579–3592. [Google Scholar] [CrossRef] [PubMed]
  27. Ballerini, P.; Dovizio, M.; Bruno, A.; Tacconelli, S.; Patrignani, P. P2Y12 Receptors in Tumorigenesis and Metastasis. Front. Pharmacol. 2018, 9, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Reiner, M.F.; Akhmedov, A.; Stivala, S.; Keller, S.; Gaul, D.S.; Bonetti, N.R.; Savarese, G.; Glanzmann, M.; Zhu, C.; Ruf, W.; et al. Ticagrelor, but not clopidogrel, reduces arterial thrombosis via endothelial tissue factor suppression. Cardiovasc. Res. 2016, 113, 61–69. [Google Scholar] [CrossRef] [Green Version]
  29. Haberstock-Debic, H.; Andre, P.; Mills, S.; Phillips, D.R.; Conley, P.B. A clopidogrel-insensitive inducible pool of P2Y12 receptors contributes to thrombus formation: Inhibition by elinogrel, a direct-acting, reversible P2Y12 antagonist. J. Pharmacol. Exp. Ther. 2011, 339, 54–61. [Google Scholar] [CrossRef] [Green Version]
  30. Cattaneo, M.; Canciani, M.; Lecchi, A.; Kinlough-Rathbone, R.; Packham, M.; Mannucci, P.; Mustard, J. Released adenosine diphosphate stabilizes thrombin-induced human platelet aggregates. Blood 1990, 75, 1081–1086. [Google Scholar] [CrossRef] [Green Version]
  31. Eckly, A.; Gendrault, J.L.; Hechler, B.; Cazenave, J.P.; Gachet, C. Differential Involvement of the P2Y1 and P2YT Receptors in the Morphological Changes of Platelet Aggregation. Thromb. Haemost. 2001, 85, 694–701. [Google Scholar] [CrossRef]
  32. Cattaneo, M. Bleeding manifestations of congenital and drug-induced defects of the platelet P2Y12 receptor for adenosine diphosphate. Thromb. Haemost. 2011, 105, 67–74. [Google Scholar] [CrossRef]
  33. McFadyen, J.D.; Schaff, M.; Peter, K. Current and future antiplatelet therapies: Emphasis on preserving haemostasis. Nat. Rev. Cardiol. 2018, 15, 181–191. [Google Scholar] [CrossRef] [PubMed]
  34. Stefanini, L.; Roden, C.R.; Bergmeier, W. CalDAG-GEFI is at the nexus of calcium-dependent platelet activation. Blood 2009, 114, 2506–2514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ohlmann, P.; Laugwitz, K.; Nürnberg, B.; Spicher, K.; Schultz, G.; Cazenave, J.; Gachet, C. The human platelet ADP receptor activates G i2 proteins. Biochem. J. 1995, 312, 775–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Daniel, J.; Dangelmaier, C.; Jin, J.; Kim, Y.; Kunapuli, S. Role of Intracellular Signaling Events in ADP-induced Platelet Aggregation. Thromb. Haemost. 1999, 82, 1322–1326. [Google Scholar] [CrossRef]
  37. Aleil, B.; Ravanat, C.; Cazenave, J.; Rochoux, G.; Heitz, A.; Gachet, C. Flow cytometric analysis of intraplatelet VASP phosphorylation for the detection of clopidogrel resistance in patients with ischemic cardiovascular diseases. J. Thromb. Haemost. 2005, 3, 85–92. [Google Scholar] [CrossRef]
  38. Laine, M.; Panagides, V.; Frère, C.; Cuisset, T.; Gouarne, C.; Jouve, B.; Thuny, F.; Paganelli, F.; Alessi, M.C.; Mancini, J.; et al. Platelet reactivity inhibition following ticagrelor loading dose in patients undergoing percutaneous coronary intervention for acute coronary syndrome. J. Thromb. Haemost. 2019, 17, 2188–2195. [Google Scholar] [CrossRef]
  39. Gratacap, M.P.; Hérault, J.P.; Viala, C.; Ragab, A.; Savi, P.; Herbert, J.M.; Chap, H.; Plantavid, M.; Payrastre, B. FcγRIIA requires a Gi-dependent pathway for an efficient stimulation of phosphoinositide 3-kinase, calcium mobilization, and platelet aggregation. Blood 2000, 96, 3439–3446. [Google Scholar] [CrossRef]
  40. Trumel, C.; Payrastre, B.; Plantavid, M.; Hechler, B.; Viala, C.; Presek, P.; Martinson, E.A.; Cazenave, J.P.; Chap, H.; Gachet, C. A Key Role of Adenosine Diphosphate in the Irreversible Platelet Aggregation Induced by the PAR1-Activating Peptide Through the Late Activation of Phosphoinositide 3-Kinase. Blood 1999, 94, 4156–4165. [Google Scholar] [CrossRef]
  41. Jackson, S.; Yap, C.; Anderson, K. Phosphoinositide 3-kinases and the regulation of platelet function. Biochem. Soc. T 2004, 32, 387–392. [Google Scholar] [CrossRef]
  42. Gratacap, M.P.; Guillermet-Guibert, J.; Martin, V.; Chicanne, G.; Tronchère, H.; Gaits-Iacovoni, F.; Payrastre, B. Regulation and roles of PI3Kβ, a major actor in platelet signaling and functions. Adv. Enzym. Regul. 2010, 51, 106–116. [Google Scholar] [CrossRef]
  43. Dangelmaier, C.; Jin, J.; Smith, B.; Kunapuli, S. Potentiation of Thromboxane A2-induced Platelet Secretion by Gi Signaling through the Phosphoinositide-3 Kinase Pathway. Thromb. Haemost. 2001, 85, 341–348. [Google Scholar] [CrossRef] [PubMed]
  44. Leon, C.; Alex, M.; Klocke, A.; Morgenstern, E.; Moosbauer, C.; Eckly, A.; Spannagl, M.; Gachet, C.; Engelmann, B. Platelet ADP receptors contribute to the initiation of intravascular coagulation. Blood 2004, 103, 594–600. [Google Scholar] [CrossRef] [PubMed]
  45. Takano, K.; Asazuma, N.; Satoh, K.; Yatomi, Y.; Ozaki, Y. Collagen-induced generation of platelet-derived microparticles in whole blood is dependent on ADP released from red blood cells and calcium ions. Platelets 2004, 15, 223–229. [Google Scholar] [CrossRef] [PubMed]
  46. Dorsam, R.; Tuluc, M.; Kunapuli, S. Role of protease-activated and ADP receptor subtypes in thrombin generation on human platelets. J. Thromb. Haemost. 2004, 2, 804–812. [Google Scholar] [CrossRef]
  47. van der Meijden, P.; Feijge, M.; Giesen, P.; Huijberts, M.; van Raak, L.; Heemskerk, J. Platelet P2Y12 receptors enhance signalling towards procoagulant activity and thrombin generation. Thromb. Haemost. 2005, 93, 1128–1136. [Google Scholar] [CrossRef]
  48. Liverani, E.; Rico, M.C.; Tsygankov, A.Y.; Kilpatrick, L.E.; Kunapuli, S.P. P2Y12 Receptor Modulates Sepsis-Induced Inflammation. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 961–971. [Google Scholar] [CrossRef] [Green Version]
  49. Lesyk, G.; Jurasz, P. Advances in Platelet Subpopulation Research. Front. Cardiovasc. Med. 2019, 6, 138. [Google Scholar] [CrossRef] [Green Version]
  50. Kotova, Y.; Ataullakhanov, F.; Panteleev, M. Formation of coated platelets is regulated by the dense granule secretion of adenosine 5′diphosphate acting via the P2Y12 receptor. J. Thromb. Haemost. 2008, 6, 1603–1605. [Google Scholar] [CrossRef]
  51. Kempton, C.L.; Hoffman, M.; Lenkowski, A.; Roberts, H.R.; Monroe, D.M. COAT Platelet Formation Is P2Y12-Dependent. Blood 2004, 104, 1569. [Google Scholar] [CrossRef]
  52. Gao, Y.; Yu, C.; Pi, S.; Mao, L.; Hu, B. The role of P2Y12 receptor in ischemic stroke of atherosclerotic origin. Cell. Mol. Life Sci. 2019, 76, 341–354. [Google Scholar] [CrossRef]
  53. Lee, C.; Hwang, I.; Park, C.S.; Lee, H.; Park, D.W.; Kang, S.J.; Lee, S.W.; Kim, Y.H.; Park, S.W.; Park, S.J. Comparison of Differential Expression of P2Y12 Receptor in Culprit Coronary Plaques in Patients With Acute Myocardial Infarction Versus Stable Angina Pectoris. Am. J. Cardiol. 2011, 108, 799–803. [Google Scholar] [CrossRef] [PubMed]
  54. Satonaka, H.; Nagata, D.; Takahashi, M.; Kiyosue, A.; Myojo, M.; Fujita, D.; Ishimitsu, T.; Nagano, T.; Nagai, R.; Hirata, Y. Involvement of P2Y 12 receptor in vascular smooth muscle inflammatory changes via MCP-1 upregulation and monocyte adhesion. Am. J. Physiol Heart C 2015, 308, H853–H861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Niu, X.; Pi, S.; Baral, S.; Xia, Y.; He, Q.; Li, Y.; Jin, H.; Li, M.; Wang, M.; Mao, L.; et al. P2Y 12 Promotes Migration of Vascular Smooth Muscle Cells Through Cofilin Dephosphorylation During Atherogenesis. Arterioscler. Thromb. Vasc Biol. 2017, 37, 515–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Cserép, C.; Pósfai, B.; Lénárt, N.; Fekete, R.; László, Z.I.; Lele, Z.; Orsolits, B.; Molnár, G.; Heindl, S.; Schwarcz, A.D.; et al. Microglia monitor and protect neuronal function via specialized somatic purinergic junctions. Science 2020, 367, 528–537. [Google Scholar] [CrossRef] [PubMed]
  57. Patente, T.A.; Pinho, M.P.; Oliveira, A.A.; Evangelista, G.C.; Bergami-Santos, P.C.; Barbuto, J.A. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front. Immunol. 2019, 9, 3176. [Google Scholar] [CrossRef]
  58. Liao, L.; Guo, Y.; Zhuang, X.; Li, W.; Zou, J.; Su, Q.; Zhao, J.; Liu, Y.; Liao, X.; Du, Z.; et al. Immunosuppressive Effect of Ticagrelor on Dendritic Cell Function: A New Therapeutic Target of Antiplatelet Agents in Cardiovascular Disease. J. Biomed. Nanotechnol. 2018, 14, 1665–1673. [Google Scholar] [CrossRef]
  59. Diehl, P.; Olivier, C.; Halscheid, C.; Helbing, T.; Bode, C.; Moser, M. Clopidogrel affects leukocyte dependent platelet aggregation by P2Y12 expressing leukocytes. Basic Res. Cardiol. 2009, 105, 379–387. [Google Scholar] [CrossRef]
  60. Suh, D.H.; Trinh, H.; Liu, J.N.; Pham, L.; Park, S.; Park, H.S.; Shin, Y. P2Y12 antagonist attenuates eosinophilic inflammation and airway hyperresponsiveness in a mouse model of asthma. J. Cell. Mol. Med. 2016, 20, 333–341. [Google Scholar] [CrossRef] [Green Version]
  61. Czajkowski, R.; Lei, L.; Sabal̵a, P.; Barańska, J. ADP-evoked phospholipase C stimulation and adenylyl cyclase inhibition in glioma C6 cells occur through two distinct nucleotide receptors, P2Y 1 and P2Y 12. FEBS Lett. 2002, 513, 179–183. [Google Scholar] [CrossRef] [Green Version]
  62. Jin, J.; Tomlinson, W.; Kirk, I.P.; Kim, Y.B.; Humphries, R.G.; Kunapuli, S.P. The C6-2B glioma cell P2Y AC receptor is pharmacologically and molecularly identical to the platelet P2Y 12 receptor. Brit. J. Pharmacol. 2001, 133, 521–528. [Google Scholar] [CrossRef] [Green Version]
  63. Sarangi, S.; Pandey, A.; Papa, A.L.; Sengupta, P.; Kopparam, J.; Dadwal, U.; Basu, S.; Sengupta, S. P2Y12 receptor inhibition augments cytotoxic effects of cisplatin in breast cancer. Med Oncol. (Northwood Lond. Engl.) 2013, 30, 567. [Google Scholar] [CrossRef] [PubMed]
  64. Fahmy, A.I.; Mekkawy, M.A.; Abou-Ali, A. Evaluation of adverse events involving bleeding associated with oral P2Y12 inhibitors use in the Food and Drug Administration adverse event reporting system. Int. J. Clin. Pharm. Ther. 2019, 57, 175–181. [Google Scholar] [CrossRef] [PubMed]
  65. Becker, R.C.; Bassand, J.; Budaj, A.; Wojdyla, D.M.; James, S.K.; Cornel, J.H.; French, J.; Held, C.; Horrow, J.; Husted, S.; et al. Bleeding complications with the P2Y12 receptor antagonists clopidogrel and ticagrelor in the PLATelet inhibition and patient Outcomes (PLATO) trial. Eur. Heart J. 2011, 32, 2933–2944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Godier, A.; Fontana, P.; Motte, S.; Steib, A.; Bonhomme, F.; Schlumberger, S.; Lecompte, T.; Rosencher, N.; Susen, S.; Vincentelli, A.; et al. Management of antiplatelet therapy in patients undergoing elective invasive procedures. Proposals from the French Working Group on perioperative haemostasis (GIHP) and the French Study Group on thrombosis and haemostasis (GFHT). In collaboration with the French Society for Anaesthesia and Intensive Care Medicine (SFAR). Anaesth. Crit. Care Pain Med. 2018, 37, 379–389. [Google Scholar]
  67. Cattaneo, M. P2Y12 antagonists. In Platelets, 4th ed.; Academic Press, Elsevier: Cambridge, MA, USA, 2019; pp. 937–956. [Google Scholar]
  68. Gross, L.; Aradi, D.; Sibbing, D. Pharmacology: Inhibitors of P2Y12. Platelets in Thrombotic and Non-Thrombotic Disorders; Springer: Cham, Switzerland, 2017; pp. 1253–1267. [Google Scholar]
  69. Wallentin, L. P2Y(12) inhibitors: Differences in properties and mechanisms of action and potential consequences for clinical use. Eur. Heart J. 2009, 30, 1964–1977. [Google Scholar] [CrossRef] [Green Version]
  70. Sharis, P.; Cannon, C.; Loscalzo, J. The Antiplatelet Effects of Ticlopidine and Clopidogrel. Ann. Intern. Med. 1998, 129, 394. [Google Scholar] [CrossRef]
  71. Gurbel, P.A.; Bliden, K.P.; Butler, K.; Tantry, U.S.; Gesheff, T.; Wei, C.; Teng, R.; Antonino, M.J.; Patil, S.B.; Karunakaran, A.; et al. Randomized Double-Blind Assessment of the ONSET and OFFSET of the Antiplatelet Effects of Ticagrelor Versus Clopidogrel in Patients With Stable Coronary Artery Disease. Circulation 2009, 120, 2577–2585. [Google Scholar] [CrossRef] [Green Version]
  72. Husted, S.E.; Storey, R.F.; Bliden, K.; Tantry, U.S.; Høimark, L.; Butler, K.; Wei, C.; Teng, R.; Gurbel, P.A. Pharmacokinetics and Pharmacodynamics of Ticagrelor in Patients with Stable Coronary Artery Disease. Clin. Pharm. 2012, 51, 397–409. [Google Scholar] [CrossRef]
  73. Husted, S.; Emanuelsson, H.; Heptinstall, S.; Sandset, P.; Wickens, M.; Peters, G. Pharmacodynamics, pharmacokinetics, and safety of the oral reversible P2Y12 antagonist AZD6140 with aspirin in patients with atherosclerosis: A double-blind comparison to clopidogrel with aspirin. Eur. Heart J. 2006, 27, 1038–1047. [Google Scholar] [CrossRef] [Green Version]
  74. Storey, R.F.; Husted, S.; Harrington, R.A.; Heptinstall, S.; Wilcox, R.G.; Peters, G.; Wickens, M.; Emanuelsson, H.; Gurbel, P.; Grande, P.; et al. Inhibition of platelet aggregation by AZD6140, a reversible oral P2Y12 receptor antagonist, compared with clopidogrel in patients with acute coronary syndromes. J. Am. Coll. Cardiol. 2007, 50, 1852–1856. [Google Scholar] [CrossRef] [Green Version]
  75. Dobesh, P.P.; Oestreich, J.H. Ticagrelor: Pharmacokinetics, Pharmacodynamics, Clinical Efficacy, and Safety. Pharm. J. Hum. Pharmacol. Drug Ther. 2014, 34, 1077–1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Franchi, F.; Rollini, F.; Muñiz-Lozano, A.; Cho, J.; Angiolillo, D.J. Cangrelor: A review on pharmacology and clinical trial development. Expert Rev. Cardiovasc. Ther. 2013, 11, 1279–1291. [Google Scholar] [CrossRef] [PubMed]
  77. Farid, N.A.; Kurihara, A.; Wrighton, S.A. Metabolism and Disposition of the Thienopyridine Antiplatelet Drugs Ticlopidine, Clopidogrel, and Prasugrel in Humans. J. Clin. Pharmacol. 2010, 50, 126–142. [Google Scholar] [CrossRef] [PubMed]
  78. Hulot, J.S.; Bura, A.; Villard, E.; Azizi, M.; Remones, V.; Goyenvalle, C.; Aiach, M.; Lechat, P.; Gaussem, P. Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects. Blood 2006, 108, 2244–2247. [Google Scholar] [CrossRef]
  79. Hagihara, K.; Kazui, M.; Kurihara, A.; Iwabuchi, H.; Ishikawa, M.; Kobayashi, H.; Tanaka, N.; Okazaki, O.; Farid, N.A.; Ikeda, T. Biotransformation of prasugrel, a novel thienopyridine antiplatelet agent, to the pharmacologically active metabolite. Drug Metab. Dispos. Biol. Fate Chem. 2010, 38, 898–904. [Google Scholar] [CrossRef] [Green Version]
  80. Wallentin, L.; Varenhorst, C.; James, S.; Erlinge, D.; Braun, O.; Jakubowski, J.; Sugidachi, A.; Winters, K.; Siegbahn, A. Prasugrel achieves greater and faster P2Y12receptor-mediated platelet inhibition than clopidogrel due to more efficient generation of its active metabolite in aspirin-treated patients with coronary artery disease. Eur. Heart J. 2007, 29, 21–30. [Google Scholar] [CrossRef]
  81. Sugidachi, A.; Ogawa, T.; Kurihara, A.; Hagihara, K.; Jakubowski, J.; Imoto; Niitsu, Y.; Asai, F. The greater in vivo antiplatelet effects of prasugrel as compared to clopidogrel reflect more efficient generation of its active metabolite with similar antiplatelet activity to that of clopidogrel’s active metabolite. J. Thromb. Haemost. 2007, 5, 1545–1551. [Google Scholar] [CrossRef]
  82. Hoffmann, K.; Lutz, D.; Straßburger, J.; Baqi, Y.; Müller, C.; von Kügelgen, I. Competitive mode and site of interaction of ticagrelor at the human platelet P2Y12 -receptor. J. Thromb. Haemost. 2014, 12, 1898–1905. [Google Scholar] [CrossRef]
  83. Giezen, V.J.; Nilsson, L.; Betsson, P.; Wissing, B.; Giordanetto, F.; Tomlinson, W.; Greasley, P. Ticagrelor binds to human P2Y 12 independently from ADP but antagonizes ADP-induced receptor signaling and platelet aggregation. J. Thromb. Haemost. 2009, 7, 1556–1565. [Google Scholar] [CrossRef]
  84. Teng, R.; Oliver, S.; Hayes, M.A.; Butler, K. Absorption, Distribution, Metabolism, and Excretion of Ticagrelor in Healthy Subjects. Drug Metab. Dispos. 2010, 38, 1514–1521. [Google Scholar] [CrossRef]
  85. Löffler, M.; Morote-Garcia, J.C.; Eltzschig, S.A.; Coe, I.R.; Eltzschig, H.K. Physiological Roles of Vascular Nucleoside Transporters. Arterioscler. Thromb. Vasc Biol. 2007, 27, 1004–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Aungraheeta, R.; Conibear, A.; Butler, M.; Kelly, E.; Nylander, S.; Mumford, A.; Mundell, S.J. Inverse agonism at the P2Y12 receptor and ENT1 transporter blockade contribute to platelet inhibition by ticagrelor. Blood 2016, 128, 2717–2728. [Google Scholar] [CrossRef] [PubMed]
  87. Cattaneo, M.; Schulz, R.; Nylander, S. Adenosine-mediated effects of ticagrelor: Evidence and potential clinical relevance. J. Am. Coll. Cardiol. 2014, 63, 2503–2509. [Google Scholar] [CrossRef] [Green Version]
  88. Ow, K.; Parker, W.A.; Porter, M.M.; Hanson, J.; Judge, H.M.; Briffa, N.P.; Thomas, M.R.; Storey, R.F. Offset of ticagrelor prior to coronary artery bypass graft surgery for acute coronary syndromes: Effects on platelet function and cellular adenosine uptake. Platelets 2020, 1–7. [Google Scholar] [CrossRef]
  89. Ortega-Paz, L.; Brugaletta, S.; Ariotti, S.; Akkerhuis, M.K.; Karagiannis, A.; Windecker, S.; Valgimigli, M.; On behalf of the investigators. Adenosine and Ticagrelor Plasma Levels in Patients With and Without Ticagrelor-Related Dyspnea. Circulation 2018, 138, 646–648. [Google Scholar] [CrossRef] [PubMed]
  90. Garcia, C.; Maurel-Ribes, A.; Nauze, M.; N’Guyen, D.; Martinez, L.O.; Payrastre, B.; Sénard, J.M.; Galés, C.; Pons, V. Deciphering biased inverse agonism of cangrelor and ticagrelor at P2Y12 receptor. Cell. Mol. Life Sci. 2019, 76, 561–576. [Google Scholar] [CrossRef] [PubMed]
  91. Lancellotti, P.; Musumeci, L.; Jacques, N.; Servais, L.; Goffin, E.; Pirotte, B.; Oury, C. Antibacterial Activity of Ticagrelor in Conventional Antiplatelet Dosages Against Antibiotic-Resistant Gram-Positive Bacteria. JAMA Cardiol. 2019, 4, 596–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Angiolillo, D.J.; Firstenberg, M.S.; Price, M.J.; Tummala, P.E.; Hutyra, M.; Welsby, I.J.; Voeltz, M.D.; Chandna, H.; Ramaiah, C.; Brtko, M.; et al. Bridging antiplatelet therapy with cangrelor in patients undergoing cardiac surgery: A randomized controlled trial. JAMA J. Am. Med Assoc. 2012, 307, 265–274. [Google Scholar] [CrossRef] [PubMed]
  93. Steg, P.; Bhatt, D.L.; Hamm, C.W.; Stone, G.W.; Gibson, M.C.; Mahaffey, K.W.; Leonardi, S.; Liu, T.; Skerjanec, S.; Day, J.R.; et al. Effect of cangrelor on periprocedural outcomes in percutaneous coronary interventions: A pooled analysis of patient-level data. Lancet Lond Engl. 2013, 382, 1981–1992. [Google Scholar] [CrossRef]
  94. Ma, L.; Dorling, A. The roles of thrombin and protease-activated receptors in inflammation. Semin. Immunopathol. 2012, 34, 63–72. [Google Scholar] [CrossRef] [Green Version]
  95. Levi, M.; van der Poll, T.; Büller, H.R. Bidirectional Relation between Inflammation and Coagulation. Circulation 2004, 109, 2698–2704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Naldini, A.; Carney, D.H.; Pucci, A.; Pasquali, A.; Carraro, F. Thrombin regulates the expression of proangiogenic cytokines via proteolytic activation of protease-activated receptor-1. Gen. Pharmacol. Vasc. Syst. 2000, 35, 255–259. [Google Scholar] [CrossRef]
  97. Naldini, A.; Carney, D.H.; Bocci, V.; Klimpel, K.D.; Asuncion, M.; Soares, L.E.; Klimpel, G.R. Thrombin Enhances T Cell Proliferative Responses and Cytokine Production. Cell. Immunol. 1993, 147, 367–377. [Google Scholar] [CrossRef] [PubMed]
  98. Johnson, K.; Choi, Y.; DeGroot, E.; Samuels, I.; Creasey, A.; Aarden, L. Potential mechanisms for a proinflammatory vascular cytokine response to coagulation activation. J. Immunol. Baltim. Md. 1950 1998, 160, 5130–5135. [Google Scholar]
  99. Strande, J.L.; Phillips, S.A. Thrombin increases inflammatory cytokine and angiogenic growth factor secretion in human adipose cells in vitro. J. Inflamm. 2009, 6, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Shankar, R.; de la Motte, C.; Poptic, E.; DiCorleto, P. Thrombin receptor-activating peptides differentially stimulate platelet-derived growth factor production, monocytic cell adhesion, and E-selectin expression in human umbilical vein endothelial cells. J. Biological. Chem. 1994, 269, 13936–13941. [Google Scholar]
  101. Iba, T.; Levy, J.H. Inflammation and thrombosis: Roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J. Thromb. Haemost. 2018, 16, 231–241. [Google Scholar] [CrossRef]
  102. Norgard, N.B.; Hann, C.L.; Dale, G.L. Cangrelor Attenuates Coated-Platelet Formation. Clin. Appl. Thromb. Hemost. 2008, 15, 177–182. [Google Scholar] [CrossRef] [Green Version]
  103. Norgard, N.B.; Saya, S.; Hann, C.; Hennebry, T.; Schechter, E.; Dale, G. Clopidogrel attenuates coated-platelet production in patients undergoing elective coronary catheterization. J. Cardiovasc. Pharm. 2008, 52, 536–539. [Google Scholar] [CrossRef]
  104. Hérault, P.J.; Dol, F.; Gaich, C.; Bernat, A.; Herbert, M.J. Effect of Clopidogrel on Thrombin Generation in Platelet-Rich Plasma in the Rat. Thromb. Haemost. 1999, 81, 957–960. [Google Scholar] [CrossRef]
  105. Wang, X.; Deng, H.; Li, T.; Miao, S.; Xiao, Z.; Liu, M.; Liu, K.; Xiao, X. Clopidogrel reduces lipopolysaccharide-induced inflammation and neutrophil-platelet aggregates in an experimental endotoxemic model. J. Biochem. Mol. Toxicol. 2019, 33, 22279. [Google Scholar] [CrossRef] [PubMed]
  106. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 2563–2582. [Google Scholar] [CrossRef] [Green Version]
  107. Bester, J.; Pretorius, E. Effects of IL-1β, IL-6 and IL-8 on erythrocytes, platelets and clot viscoelasticity. Sci Rep. UK 2016, 6, 32188. [Google Scholar] [CrossRef] [PubMed]
  108. Lumadue, J.A.; Lanzkron, S.M.; Kennedy, S.D.; Kuhl, D.T.; Kickler, T.S. Cytokine Induction of Platelet Activation. Am. J. Clin. Pathol. 1996, 106, 795–798. [Google Scholar] [CrossRef] [Green Version]
  109. Assinger, A.; Schrottmaier, W.C.; Salzmann, M.; Rayes, J. Platelets in Sepsis: An Update on Experimental Models and Clinical Data. Front. Immunol. 2019, 10, 1687. [Google Scholar] [CrossRef] [PubMed]
  110. Slaba, I.; Kubes, P. Platelets in Thrombotic and Non-Thrombotic Disorders; Springer: Cham, Switzerland, 2017; pp. 489–512. [Google Scholar]
  111. Sanguigni, V.; Ferro, D.; Pignatelli, P.; Ben, M.; Nadia, T.; Saliola, M.; Sorge, R.; Violi, F. CD40 ligand enhances monocyte tissue factor expression and thrombin generation via oxidative stress in patients with hypercholesterolemia. J. Am. Coll. Cardiol. 2005, 45, 35–42. [Google Scholar] [CrossRef] [PubMed]
  112. Liverani, E.; Kilpatrick, L.E.; Tsygankov, A.Y.; Kunapuli, S.P. The role of P2Y₁₂ receptor and activated platelets during inflammation. Curr. Drug Targets 2014, 15, 720–728. [Google Scholar] [CrossRef] [Green Version]
  113. Sexton, T.R.; Zhang, G.; Macaulay, T.E.; Callahan, L.A.; Charnigo, R.; Vsevolozhskaya, O.A.; Li, Z.; Smyth, S. Ticagrelor Reduces Thromboinflammatory Markers in Patients With Pneumonia. JACC. Basic Transl. Sci. 2018, 3, 435–449. [Google Scholar] [CrossRef]
  114. Hagiwara, S.; Iwasaka, H.; Hasegawa, A.; Oyama, M.; Imatomi, R.; Uchida, T.; Noguchi, T. Adenosine Diphosphate Receptor Antagonist Clopidogrel Sulfate Attenuates LPS-Induced Systemic Inflammation in a Rat Model. Shock (Augusta Ga.) 2011, 35, 289–292. [Google Scholar] [CrossRef]
  115. Thomas, M.R.; Outteridge, S.N.; Ajjan, R.A.; Phoenix, F.; Sangha, G.K.; Faulkner, R.E.; Ecob, R.; Judge, H.M.; Khan, H.; West, L.E.; et al. Platelet P2Y12 Inhibitors Reduce Systemic Inflammation and Its Prothrombotic Effects in an Experimental Human Model. Arterioscler. Thromb. Vasc Biol. 2015, 35, 2562–2570. [Google Scholar] [CrossRef] [Green Version]
  116. Thomas, M.R.; Storey, R.F. Effect of P2Y12 inhibitors on inflammation and immunity. Thromb. Haemost. 2015, 114, 490–497. [Google Scholar] [PubMed] [Green Version]
  117. Rossaint, J.; Margraf, A.; Zarbock, A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front. Immunol. 2018, 9, 2712. [Google Scholar] [CrossRef] [PubMed]
  118. Li, Z.; Smyth, S.S. Platelets, 4th ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 295–310. [Google Scholar]
  119. Evangelista, V.; Totani, L.; Manfredi, A.A.; Maugeri, N. Platelets in Thrombotic and Non-Thrombotic Disorders; Springer: Berlin/Heidelberg, Germany, 2017; pp. 407–433. [Google Scholar]
  120. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
  121. Elaskalani, O.; Razak, N.; Metharom, P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun. Signal. 2018, 16, 24. [Google Scholar] [CrossRef] [Green Version]
  122. Etulain, J.; Martinod, K.; Wong, S.; Cifuni, S.M.; Schattner, M.; Wagner, D.D. P-selectin promotes neutrophil extracellular trap formation in mice. Blood 2015, 126, 242–246. [Google Scholar] [CrossRef] [Green Version]
  123. Carestia, A.; Kaufman, T.; Schattner, M. Platelets: New Bricks in the Building of Neutrophil Extracellular Traps. Front. Immunol. 2016, 7, 271. [Google Scholar] [CrossRef] [Green Version]
  124. Zucoloto, A.Z.; Jenne, C.N. Platelet-Neutrophil Interplay: Insights Into Neutrophil Extracellular Trap (NET)-Driven Coagulation in Infection. Front. Cardiovasc. Med. 2019, 6, 85. [Google Scholar] [CrossRef] [Green Version]
  125. Kazzaz, N.M.; Sule, G.; Knight, J.S. Intercellular Interactions as Regulators of NETosis. Front. Immunol. 2016, 7, 453. [Google Scholar] [CrossRef] [Green Version]
  126. Rahman, M.; Gustafsson, D.; Wang, Y.; Thorlacius, H.; Braun, O. Ticagrelor reduces neutrophil recruitment and lung damage in abdominal sepsis. Platelets 2014, 25, 257–263. [Google Scholar] [CrossRef]
  127. Evangelista, V.; Manarini, S.; Dell’Elba, G.; Martelli, N.; Napoleone, E.; Santo, A.; Savi, P.; Lorenzet, R. Clopidogrel inhibits platelet-leukocyte adhesion and plateletdependent leukocyte activation. Thromb. Haemost. 2005, 94, 568–577. [Google Scholar]
  128. Judge, H.M.; Buckland, R.J.; Sugidachi, A.; Jakubowski, J.A.; Storey, R.F. The active metabolite of prasugrel effectively blocks the platelet P2Y12 receptor and inhibits procoagulant and pro-inflammatory platelet responses. Platelets 2008, 19, 125–133. [Google Scholar] [CrossRef] [PubMed]
  129. Storey, R.; Judge, H.; Wilcox, R.; Heptinstall, S. Inhibition of ADP-induced P-selectin Expression and Platelet-Leukocyte Conjugate Formation by Clopidogrel and the P2Y12 Receptor Antagonist AR-C69931MX but not Aspirin. Thromb. Haemost. 2002, 88, 488–494. [Google Scholar] [PubMed]
  130. Klinkhardt, U. Clopidogrel but not aspirin reduces P-selectin expression and formation of platelet-leukocyte aggregates in patients with atherosclerotic vascular disease. Clin. Pharmacol. Ther. 2003, 73, 232–241. [Google Scholar] [CrossRef] [PubMed]
  131. Xiao, Z.; Théroux, P. Clopidogrel inhibits platelet-leukocyte interactions and thrombin receptor agonist peptide-induced platelet activation in patients with an acute coronary syndrome. J. Am. Coll. Cardiol. 2004, 43, 1982–1988. [Google Scholar] [CrossRef] [Green Version]
  132. Vigano, S.; Alatzoglou, D.; Irving, M.; Ménétrier-Caux, C.; Caux, C.; Romero, P.; Coukos, G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front. Immunol. 2019, 10, 925. [Google Scholar] [CrossRef] [Green Version]
  133. Wolska, N.; Rozalski, M. Blood Platelet Adenosine Receptors as Potential Targets for Anti-Platelet Therapy. Int. J. Mol. Sci. 2019, 20, 5475. [Google Scholar] [CrossRef] [Green Version]
  134. Boncler, M.; Wzorek, J.; Wolska, N.; Polak, D.; Watala, C.; Rozalski, M. Adenosine receptor agonists deepen the inhibition of platelet aggregation by P2Y12 antagonists. Vasc. Pharmacol. 2018, 113, 47–56. [Google Scholar] [CrossRef]
  135. Winning, J.; Reichel, J.; Eisenhut, Y.; Hamacher, J.; Kohl, M.; Deigner, H.; Claus, R.A.; Bauer, M.; Lösche, W. Anti-platelet drugs and outcome in severe infection: Clinical impact and underlying mechanisms. Platelets 2009, 20, 50–57. [Google Scholar] [CrossRef]
  136. Storey, R.F.; James, S.K.; Siegbahn, A.; Varenhorst, C.; Held, C.; Ycas, J.; Husted, S.E.; Cannon, C.P.; Becker, R.C.; Steg, P.; et al. Lower mortality following pulmonary adverse events and sepsis with ticagrelor compared to clopidogrel in the PLATO study. Platelets 2013, 25, 517–525. [Google Scholar] [CrossRef]
  137. Tsai, M.J.; Ou, S.M.; Shih, C.J.; Chao, P.; Wang, L.F.; Shih, Y.N.; Li, S.Y.; Kuo, S.C.; Hsu, Y.T.; Chen, Y.T. Association of prior antiplatelet agents with mortality in sepsis patients: A nationwide population-based cohort study. Intensive Care Med. 2015, 41, 806–813. [Google Scholar] [CrossRef]
  138. Laidlaw, T.M.; Cahill, K.N.; Cardet, J.; Murphy, K.; Cui, J.; Dioneda, B.; Kothari, P.; Raby, B.A.; Israel, E.; Boyce, J. A trial of P2Y12 receptor inhibition with prasugrel identifies a potentially distinct endotype of patients with aspirin-exacerbated respiratory disease. J. Allergy Clin. Immun. 2018, 143, 316–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Lussana, F.; Marco, D.F.; Terraneo, S.; Parati, M.; Razzari, C.; Scavone, M.; Femia, E.; Moro, A.; Centanni, S.; Cattaneo, M. Effect of prasugrel in patients with asthma: Results of PRINA, a randomized, double-blind, placebo-controlled, cross-over study. J. Thromb. Haemost. 2015, 13, 136–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Raposeiras-Roubin, S.; Abu-Assi, E.; Muñoz-Pousa, I.; Cespón-Fernández, M.; Cobas-Paz, R.; Caneiro-Queija, B.; López-Rodríguez, E.; Pérez-Casares, L.; Jamhour-Chelh, K.; Castiñeira-Busto, M.; et al. Risk of cancer after an acute coronary syndrome according to the type of P2Y12 inhibitor. Thromb. Res. 2019, 174, 51–58. [Google Scholar] [CrossRef] [PubMed]
  141. Leader, A.; Zelikson-Saporta, R.; Pereg, D.; Spectre, G.; Rozovski, U.; Raanani, P.; Hermoni, D.; Lishner, M. The Effect of Combined Aspirin and Clopidogrel Treatment on Cancer Incidence. Am. J. Med. 2017, 130, 826–832. [Google Scholar] [CrossRef] [Green Version]
  142. Hicks, B.M.; Murray, L.J.; Hughes, C.; Cardwell, C.R. Clopidogrel use and cancer-specific mortality: A population-based cohort study of colorectal, breast and prostate cancer patients. Pharmacoepidemiol. Drug Saf. 2015, 24, 830–840. [Google Scholar] [CrossRef]
  143. Elmariah, S.; Doros, G.; Benavente, O.R.; Bhatt, D.L.; Connolly, S.J.; Yusuf, S.; Steinhubl, S.R.; Liu, Y.; Hsieh, W.H.; Yeh, R.W.; et al. Impact of Clopidogrel Therapy on Mortality and Cancer in Patients With Cardiovascular and Cerebrovascular Disease: A Patient-Level Meta-Analysis. Circulation. Cardiovasc. Interv. 2018, 11, 005795. [Google Scholar] [CrossRef]
  144. Rodríguez-Miguel, A.; García-Rodríguez, L.A.; Gil, M.; Montoya, H.; Rodríguez-Martín, S.; de Abajo, F.J. Clopidogrel and Low-Dose Aspirin, Alone or Together, Reduce Risk of Colorectal Cancer. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2019, 17, 2024–2033. [Google Scholar] [CrossRef] [Green Version]
  145. Hotchkiss, R.S.; Moldawer, L.L.; Opal, S.M.; Reinhart, K.; Turnbull, I.R.; Vincent, J.L. Sepsis and septic shock. Nat. Rev. Dis. Primers 2016, 2, 16045. [Google Scholar] [CrossRef] [Green Version]
  146. van der Poll, T.; van de Veerdonk, F.L.; Scicluna, B.P.; Netea, M.G. The immunopathology of sepsis and potential therapeutic targets. Nat. Rev. Immunol. 2017, 17, 407–420. [Google Scholar] [CrossRef]
  147. Vardon-Bounes, F.; Ruiz, S.; Gratacap, M.P.; Garcia, C.; Payrastre, B.; Minville, V. Platelets Are Critical Key Players in Sepsis. Int. J. Mol. Sci. 2019, 20, 3494. [Google Scholar] [CrossRef] [Green Version]
  148. Wang, Y.; Ouyang, Y.; Liu, B.; Ma, X.; Ding, R. Platelet activation and antiplatelet therapy in sepsis: A narrative review. Thromb. Res. 2018, 166, 28–36. [Google Scholar] [CrossRef] [PubMed]
  149. Matthay, M.A.; Zemans, R.L.; Zimmerman, G.A.; Arabi, Y.M.; Beitler, J.R.; Mercat, A.; Herridge, M.; Randolph, A.G.; Calfee, C.S. Acute respiratory distress syndrome. Nat. Rev. Dis. Primers 2019, 5, 18. [Google Scholar] [CrossRef] [PubMed]
  150. Middleton, E.A.; Rondina, M.T.; Schwertz, H.; Zimmerman, G.A. Amicus or Adversary Revisited: Platelets in Acute Lung Injury and Acute Respiratory Distress Syndrome. Am. J. Resp. Cell Mol. 2018, 59, 18–35. [Google Scholar] [CrossRef] [PubMed]
  151. Bozza, F.A.; ah, A.; Weyrich, A.S.; Zimmerman, G.A. Amicus or Adversary. Am. J. Resp. Cell Mol. 2009, 40, 123–134. [Google Scholar] [CrossRef] [PubMed]
  152. Wallentin, L.; Becker, R.C.; Budaj, A.; Cannon, C.P.; Emanuelsson, H.; Held, C.; Horrow, J.; Husted, S.; James, S.; Katus, H.; et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N. Engl. J. Med. 2009, 361, 1045–1057. [Google Scholar] [CrossRef]
  153. Takeda, T.; Morita, H.; Saito, H.; Matsumoto, K.; Matsuda, A. Recent advances in understanding the roles of blood platelets in the pathogenesis of allergic inflammation and bronchial asthma. Allergol. Int. 2017, 67, 326–333. [Google Scholar] [CrossRef]
  154. Idzko, M.; Pitchford, S.; Page, C. Role of platelets in allergic airway inflammation. J. Allergy Clin. Immun. 2015, 135, 1416–1423. [Google Scholar] [CrossRef]
  155. Kowal, K.; Pampuch, A.; Kowal-Bielecka, O.; DuBuske, L.; Bodzenta-Łukaszyk, A. Platelet activation in allergic asthma patients during allergen challenge with Dermatophagoides pteronyssinus. Clin. Exp. Allergy 2006, 36, 426–432. [Google Scholar] [CrossRef]
  156. Gawaz, M.; Borst, O. 26 the Role of Platelets in Atherothrombosis. Platelets 2019, 459–467. [Google Scholar]
  157. Lievens, D.; von Hundelshausen, P. Platelets in atherosclerosis. Thromb. Haemost. 2011, 106, 827–838. [Google Scholar]
  158. Lebas, H.; Yahiaoui, K.; Martos, R.; Boulaftali, Y. Platelets Are at the Nexus of Vascular Diseases. Front. Cardiovasc. Med. 2019, 6, 132. [Google Scholar] [CrossRef] [PubMed]
  159. Li, D.; Wang, Y.; Zhang, L.; Luo, X.; Li, J.; Chen, X.; Niu, H.; Wang, K.; Sun, Y.; Wang, X.; et al. Roles of Purinergic Receptor P2Y, G Protein–Coupled 12 in the Development of Atherosclerosis in Apolipoprotein E–Deficient Mice. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 81–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Heim, C.; Gebhardt, J.; Ramsperger-Gleixner, M.; Jacobi, J.; Weyand, M.; Ensminger, S.M. Clopidogrel significantly lowers the development of atherosclerosis in ApoE-deficient mice in vivo. Heart Vessel. 2015, 31, 783–794. [Google Scholar] [CrossRef] [PubMed]
  161. Afek, A.; Kogan, E.; Maysel-Auslender, S.; Mor, A.; Regev, E.; Rubinstein, A.; Keren, G.; George, J. Clopidogrel attenuates atheroma formation and induces a stable plaque phenotype in apolipoprotein E knockout mice. Microvasc. Res. 2009, 77, 364–369. [Google Scholar] [CrossRef] [PubMed]
  162. Xia, X.; Li, J.; Liang, X.; Zhang, S.; Liu, T.; Liu, J.; Arif, M.; Li, G. Ticagrelor suppresses oxidized low-density lipoprotein-induced endothelial cell apoptosis and alleviates atherosclerosis in ApoE-/- mice via downregulation of PCSK9. Mol. Med. Rep. 2018, 19, 1453–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ganbaatar, B.; Fukuda, D.; Salim, H.; Nishimoto, S.; Tanaka, K.; Higashikuni, Y.; Hirata, Y.; Yagi, S.; Soeki, T.; Sata, M. Ticagrelor, a P2Y12 antagonist, attenuates vascular dysfunction and inhibits atherogenesis in apolipoprotein-E-deficient mice. Atherosclerosis 2018, 275, 124–132. [Google Scholar] [CrossRef] [PubMed]
  164. West, L.E.; Steiner, T.; Judge, H.M.; Francis, S.E.; Storey, R.F. Vessel wall, not platelet, P2Y12 potentiates early atherogenesis. Cardiovasc. Res. 2014, 102, 429–435. [Google Scholar] [CrossRef] [Green Version]
  165. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  166. Haemmerle, M.; Stone, R.L.; Menter, D.G.; Afshar-Kharghan, V.; Sood, A.K. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018, 33, 965–983. [Google Scholar] [CrossRef]
  167. Cho, M.; Noh, K.; Haemmerle, M.; Li, D.; Park, H.; Hu, Q.; Hisamatsu, T.; Mitamura, T.; Mak, S.; Kunapuli, S.; et al. Role of ADP receptors on platelets in the growth of ovarian cancer. Blood 2017, 130, 1235–1242. [Google Scholar] [CrossRef]
  168. Gareau, A.J.; Brien, C.; Gebremeskel, S.; Liwski, R.S.; Johnston, B.; Bezuhly, M. Ticagrelor inhibits platelet-tumor cell interactions and metastasis in human and murine breast cancer. Clin. Exp. Metastasis 2018, 35, 25–35. [Google Scholar] [CrossRef]
  169. Gebremeskel, S.; LeVatte, T.; Liwski, R.S.; Johnston, B.; Bezuhly, M. The reversible P2Y12 inhibitor ticagrelor inhibits metastasis and improves survival in mouse models of cancer. Int. J. Cancer 2015, 136, 234–240. [Google Scholar] [CrossRef] [PubMed]
  170. Wang, Y.; Sun, Y.; Li, D.; Zhang, L.; Wang, K.; Zuo, Y.; Gartner, K.T.; Liu, J. Platelet P2Y12 is involved in murine pulmonary metastasis. PLoS ONE 2013, 8, e80780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Geranpayehvaghei, M.; Shi, Q.; Zhao, B.; Li, S.; Xu, J.; Taleb, M.; Qin, H.; Zhang, Y.; Khajeh, K.; Nie, G. Targeting Delivery of Platelets Inhibitor to Prevent Tumor Metastasis. Bioconjug. Chem. 2019, 30, 2349–2357. [Google Scholar] [CrossRef] [PubMed]
  172. Algra, A.M.; Rothwell, P.M. Effects of regular aspirin on long-term cancer incidence and metastasis: A systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 2012, 13, 518–527. [Google Scholar] [CrossRef]
  173. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 1996, 348, 1329–1339. [Google Scholar] [CrossRef]
  174. Bhatt, D.L.; Fox, K.A.; Hacke, W.; Berger, P.B.; Black, H.R.; Boden, W.E.; Cacoub, P.; Cohen, E.A.; Creager, M.A.; Easton, D.J.; et al. Clopidogrel and Aspirin versus Aspirin Alone for the Prevention of Atherothrombotic Events. N. Engl. J. Med. 2006, 354, 1706–1717. [Google Scholar] [CrossRef] [Green Version]
  175. Wiviott, S.D.; Braunwald, E.; McCabe, C.H.; Montalescot, G.; Ruzyllo, W.; Gottlieb, S.; Neumann, F.J.; Ardissino, D.; Servi, S.; Murphy, S.A.; et al. Prasugrel versus Clopidogrel in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 2007, 357, 2001–2015. [Google Scholar] [CrossRef] [Green Version]
  176. Roe, M.T.; Armstrong, P.W.; Fox, K.A.; White, H.D.; Prabhakaran, D.; Goodman, S.G.; Cornel, J.H.; Bhatt, D.L.; Clemmensen, P.; Martinez, F.; et al. Prasugrel versus Clopidogrel for Acute Coronary Syndromes without Revascularization. N. Engl J. Med. 2012, 367, 1297–1309. [Google Scholar] [CrossRef] [Green Version]
  177. Mauri, L.; Kereiakes, D.J.; Yeh, R.W.; Driscoll-Shempp, P.; Cutlip, D.E.; Steg, G.P.; Normand, S.L.T.; Braunwald, E.; Wiviott, S.D.; Cohen, D.J.; et al. Twelve or 30 months of dual antiplatelet therapy after drug-eluting stents. N. Engl. J. Med. 2014, 371, 2155–2166. [Google Scholar] [CrossRef] [Green Version]
  178. Bonaca, M.P.; Bhatt, D.L.; Cohen, M.; Steg, P.; Storey, R.F.; Jensen, E.C.; Magnani, G.; Bansilal, S.; Fish, P.M.; Im, K.; et al. Long-Term Use of Ticagrelor in Patients with Prior Myocardial Infarction. N. Engl. J. Med. 2015, 372, 1791–1800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Serebruany, V.L. Ticagrelor shift from PLATO to PEGASUS: Vanished mortality benefit, excess cancer deaths, massive discontinuations, and overshooting target events. Int. J. Cardiol. 2015, 201, 508–512. [Google Scholar] [CrossRef] [PubMed]
  180. U.S. Food and Drug Administration. U.S. FDA Review Finds Long-Term Treatment with Blood- Thinning Medicine Plavix (Clopidogrel) Does not Change Risk of Death; FDA: Silver Spring, MD, USA, 2014.
  181. Serebruany, V.; Kim, M.; Thevathasan, C.; Marciniak, T. Assessing Cancer Signal during Oral Antiplatelet Therapy in the Food and Drug Administration Adverse Event Reporting System: Mission Impossible. TH Open Companion J. Thromb. Haemost. 2018, 2, 28–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Kotronias, R.; Kwok, C.; Wong, C.; Kinnaird, T.; Zaman, A.; Mamas, M.A. Cancer Event Rate and Mortality with Thienopyridines: A Systematic Review and Meta-Analysis. Drug Saf. 2016, 40, 229–240. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the platelet P2-mediated ADP signaling pathway and its role in platelet activation. ADP can induce platelet activation by interacting with two G-coupled platelet receptors, P2Y1 and P2Y12, and contributes to the stabilization of platelet aggregation induced by other strong agonists such as thrombin and TXA2. ADP binding to P2Y1 induces a mobilization of calcium ions through the stimulation of phospholipase C and therefore initiates early reversible aggregation. ADP binding to P2Y12 induces PI3K activation through the recruitment of βγ subunits of Gi and inhibition of adenylate cyclase through the recruitment of αi subunit, therefore decreasing cAMP levels and leading to impaired PKA activation. These pathways ultimately lead to platelet activation, αIIbβ3 activation and stabilization of platelet aggregates. AC: adenylate cyclase; ADP: adenosine diphosphate; ATP: adenosine triphosphate; PI3K: phosphoinositide-3-kinase; PKA: protein kinase A; PLC: phospholipase C; VASP: vasodilator-stimulated phosphoprotein.
Figure 1. Schematic illustration of the platelet P2-mediated ADP signaling pathway and its role in platelet activation. ADP can induce platelet activation by interacting with two G-coupled platelet receptors, P2Y1 and P2Y12, and contributes to the stabilization of platelet aggregation induced by other strong agonists such as thrombin and TXA2. ADP binding to P2Y1 induces a mobilization of calcium ions through the stimulation of phospholipase C and therefore initiates early reversible aggregation. ADP binding to P2Y12 induces PI3K activation through the recruitment of βγ subunits of Gi and inhibition of adenylate cyclase through the recruitment of αi subunit, therefore decreasing cAMP levels and leading to impaired PKA activation. These pathways ultimately lead to platelet activation, αIIbβ3 activation and stabilization of platelet aggregates. AC: adenylate cyclase; ADP: adenosine diphosphate; ATP: adenosine triphosphate; PI3K: phosphoinositide-3-kinase; PKA: protein kinase A; PLC: phospholipase C; VASP: vasodilator-stimulated phosphoprotein.
Ijms 21 01391 g001
Figure 2. Schematic overview of the role of platelet P2Y12 activation in inflammation. ADP binding to P2Y12 initiates platelet activation and amplifies granule secretion and aggregation induced by ADP-mediated P2Y1 stimulation and by strong agonists such as thrombin (PAR-1 and PAR-4 receptors) and TXA2 (TP receptor). Activated platelets release alpha and dense granules contents, including cytokines, chemokines, coagulation factors and platelet agonists. Cytokines, chemokines and soluble CD40 ligand (sCD40L) can recruit and activate leukocytes, especially neutrophils and monocytes. Expression of P-selectin and CD40L at the surface of activated platelets allow interaction between platelets and leukocytes which is a critical step in platelet-mediated inflammation. P-selectin cross-links platelets and leukocytes through its corresponding ligand P-selectin glycoprotein ligand-1 (PSGL-1) present on monocytes and neutrophils. Platelet-leukocyte aggregates are further stabilized by numerous additional receptor/ligand pairs, especially leukocyte Mac-1 that recognizes platelet CD40L and fibrinogen (Fg) bound to platelet αIIbβ3. Activation by platelet interaction or soluble mediators stimulates monocyte cytokine and chemokine production, expression of tissue factor and thrombin generation. Platelet-bound neutrophils produce inflammatory mediators including reactive oxygen species (ROS), and release neutrophil extracellular traps (NETs) that play a critical role in many inflammatory conditions, especially sepsis-induced organ injuries, autoimmunity, tumorigenesis and metastasis.
Figure 2. Schematic overview of the role of platelet P2Y12 activation in inflammation. ADP binding to P2Y12 initiates platelet activation and amplifies granule secretion and aggregation induced by ADP-mediated P2Y1 stimulation and by strong agonists such as thrombin (PAR-1 and PAR-4 receptors) and TXA2 (TP receptor). Activated platelets release alpha and dense granules contents, including cytokines, chemokines, coagulation factors and platelet agonists. Cytokines, chemokines and soluble CD40 ligand (sCD40L) can recruit and activate leukocytes, especially neutrophils and monocytes. Expression of P-selectin and CD40L at the surface of activated platelets allow interaction between platelets and leukocytes which is a critical step in platelet-mediated inflammation. P-selectin cross-links platelets and leukocytes through its corresponding ligand P-selectin glycoprotein ligand-1 (PSGL-1) present on monocytes and neutrophils. Platelet-leukocyte aggregates are further stabilized by numerous additional receptor/ligand pairs, especially leukocyte Mac-1 that recognizes platelet CD40L and fibrinogen (Fg) bound to platelet αIIbβ3. Activation by platelet interaction or soluble mediators stimulates monocyte cytokine and chemokine production, expression of tissue factor and thrombin generation. Platelet-bound neutrophils produce inflammatory mediators including reactive oxygen species (ROS), and release neutrophil extracellular traps (NETs) that play a critical role in many inflammatory conditions, especially sepsis-induced organ injuries, autoimmunity, tumorigenesis and metastasis.
Ijms 21 01391 g002
Table 1. Pharmacological properties of P2Y12 receptor inhibitors.
Table 1. Pharmacological properties of P2Y12 receptor inhibitors.
DrugTiclopidineClopidogrelPrasugrelTicagrelorCangrelor
TargetP2Y12P2Y12P2Y12P2Y12
ENT-1
P2Y12
P2Y12 receptor binding IrreversibleIrreversibleIrreversibleReversibleReversible
Route of administrationOralOralOralOralIntravenous
MetabolismProdrug
CYP450
Prodrug
Esterase
CYP450
Prodrug
Intestinal
Esterase
CYP450
Direct-Acting and CYP450Direct-Acting
Dephosphorylation
Time to maximum IPA 13–4 days4–5 h2–4h2–4 h2 min
Steady-state IPA 220–30%33–64%43–73%82–95%>80%
Offset of action 311–13 days5–7 days7–10 days3–5 days30–60 min
CYP450: cytochrome P450; IPA: inhibition of platelet aggregation; 1 after a loading dose; 2 percentage inhibition of platelet aggregation measured by light transmission aggregometry; 3 based on return of platelet aggregation and/or bleeding time to baseline values; [67,68,69,70,71,72,73,74,75,76].
Table 2. Clinical studies investigating anti-inflammatory effects of P2Y12 antagonists.
Table 2. Clinical studies investigating anti-inflammatory effects of P2Y12 antagonists.
StudyType of StudyConditionAntiplatelet Drugs EvaluatedEffects of P2Y12 Antagonists
Winning et al. [135]Observational
224 patients
CAPASA and thienopyridinesLower use of ICU and shorter stay in hospital (thienopyridines plus ASA or thienopyridines alone
Storey et al. [136]Observational post hoc analysis18,421 patientsACSTicagrelor vs clopidogrelLower mortality risk following pulmonary events and sepsis
Tsai et al. [137]Observational
683,421 patients
SepsisASA and thienopyridinesLower risk of mortality
XANTHIPPE
Sexton et al. [113]
RCT
60 patients
Pneumonia (CAP, HAP)Ticagrelor vs placeboReduced PLAs and IL-6 levels improved. Decreased supplemental oxygen requirements
Laidlaw et al. [138]Crossover RCT
40 patients
AERDPrasugrel vs placeboNo effect on clinical or inflammatory parameters
PRINA
Lussana et al. [139]
Crossover RCT
26 patients
Chronic asthmaPrasugrel vs placeboDecreased airway hyperresponsiveness
Raposeiras-Roubin et al. [140]Observational
4229 patients
ACSTicagrelor, Clopidogrel and PrasugrelLower cancer risk ticagrelor vs clopidogrelNS clopidogrel vs prasugrel
Leader et al. [141]Observational
3479 patients
ACSASA and clopidogrelLower cancer risk (clopidogrel plus ASA or clopidogrel alone)
Hicks et al. [142]Observational
41,403 patients
Cancer (breast, colorectal, prostate)ClopidogrelNo increase in cancer risk
Elmariah et al. [143]Meta-analysis
48,000 patients
Cardiovascular and cerebrovascular diseaseASA and clopidogrelNo increase in cancer risk
Rodriguez-Miguel et al. [144]Observational
75,491 patients
CRCASA and clopidogrelLower cancer risk (clopidogrel plus ASA or clopidogrel alone)
CAP: community acquired pneumonia, HAP: hospital acquired pneumonia, ASA: acetylsalicylic acid=aspirin, ICU: intensive care unit, ACS: acute coronary syndrome, RCT: randomized control trial, AERD: aspirin-exacerbated respiratory disease, NS: non-significant difference, CRC: colorectal cancer.

Share and Cite

MDPI and ACS Style

Mansour, A.; Bachelot-Loza, C.; Nesseler, N.; Gaussem, P.; Gouin-Thibault, I. P2Y12 Inhibition beyond Thrombosis: Effects on Inflammation. Int. J. Mol. Sci. 2020, 21, 1391. https://doi.org/10.3390/ijms21041391

AMA Style

Mansour A, Bachelot-Loza C, Nesseler N, Gaussem P, Gouin-Thibault I. P2Y12 Inhibition beyond Thrombosis: Effects on Inflammation. International Journal of Molecular Sciences. 2020; 21(4):1391. https://doi.org/10.3390/ijms21041391

Chicago/Turabian Style

Mansour, Alexandre, Christilla Bachelot-Loza, Nicolas Nesseler, Pascale Gaussem, and Isabelle Gouin-Thibault. 2020. "P2Y12 Inhibition beyond Thrombosis: Effects on Inflammation" International Journal of Molecular Sciences 21, no. 4: 1391. https://doi.org/10.3390/ijms21041391

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop