G. Burnstock and V. Ralevic, Purinergic Signaling and Blood Vessels in Health and Disease, Pharmacol. Rev, vol.66, pp.102-192, 2013.

I. Von-kügelgen, Pharmacology of P2Y receptors, Brain Res. Bull, vol.151, pp.12-24, 2019.

M. Cattaneo, P2Y12 receptors: Structure and function, J. Thromb. Haemost, vol.13, pp.10-16, 2015.

C. Gachet, P2Y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells, Purinergic Signal, vol.8, pp.609-619, 2012.

B. Hechler and C. Gachet, The P2 Receptors. Platelets in Thrombotic and Non-Thrombotic Disorders; Pathophysiology, Pharmacology and Therapeutics: An Update, pp.187-202, 2017.

Y. Baqi and C. E. Müller, Antithrombotic P2Y12 receptor antagonists: Recent developments in drug discovery, Drug Discov. Today, vol.24, pp.325-333, 2019.

B. Hechler and C. Gachet, Purinergic Receptors in Thrombosis and Inflammation, Arterioscler. Thromb. Vasc Biol, vol.35, pp.2307-2315, 2015.

M. R. Thomas and R. F. Storey, The role of platelets in inflammation, Thromb. Haemost, vol.114, pp.449-458, 2015.

A. Palacios-acedo, D. Mège, L. Crescence, F. Dignat-george, C. Dubois et al., Thrombo-Inflammation, and Cancer: Collaborating With the Enemy, Front. Immunol, vol.10, p.1805, 2019.

F. L. Zhang, L. Luo, E. Gustafson, J. Lachowicz, M. Smith et al., ADP Is the Cognate Ligand for the Orphan G Protein-coupled Receptor SP1999, J. Biol. Chem, vol.276, pp.8608-8615, 2000.

G. Hollopeter, H. M. Jantzen, D. Vincent, G. Li, L. England et al., Identification of the platelet ADP receptor targeted by antithrombotic drugs, Nature, vol.409, pp.202-207, 2001.

K. Zhang, J. Zhang, Z. G. Gao, D. Zhang, L. Zhu et al., Structure of the human P2Y12 receptor in complex with an antithrombotic drug, Nature, vol.509, pp.115-118, 2014.

M. Cattaneo, The platelet P2 receptor, Platelets, pp.259-277, 2019.

I. Von-kügelgen, Pharmacological profiles of cloned mammalian P2Y-receptor subtypes, vol.110, pp.415-432, 2006.

P. Ohlmann, A. Lecchi, A. El-tayeb, C. E. Müller, M. Cattaneo et al., The platelet P2Y(12) receptor under normal and pathological conditions. Assessment with the radiolabeled selective antagonist, Purinerg. Signal, vol.9, issue.3, pp.59-66, 2012.

S. E. Haynes, G. Hollopeter, G. Yang, D. Kurpius, M. E. Dailey et al., The P2Y12 receptor regulates microglial activation by extracellular nucleotides, Nat. Neurosci, vol.9, pp.1512-1519, 2006.

B. H. Rauch, A. C. Rosenkranz, S. Ermler, A. Böhm, J. Driessen et al., 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, vol.30, pp.2434-2442, 2010.

A. K. Wihlborg, L. Wang, O. Braun, A. Eyjolfsson, R. Gustafsson et al., ADP Receptor P2Y 12 Is Expressed in Vascular Smooth Muscle Cells and Stimulates Contraction in Human Blood Vessels, Arterioscler. Thromb. Vasc Biol, vol.24, pp.1810-1815, 2004.

A. Addi, D. Cammarata, P. B. Conley, J. M. Boeynaems, and B. Robaye, Role of the P2Y 12 Receptor in the Modulation of Murine Dendritic Cell Function by ADP, J. Immunol, vol.185, pp.5900-5906, 2010.

S. Paruchuri, H. Tashimo, C. Feng, A. Maekawa, W. Xing et al., Leukotriene E4-induced pulmonary inflammation is mediated by the P2Y12 receptor, J. Exp. Med, vol.206, pp.2543-2555, 2009.

V. S. Muniz, R. Baptista-dos-reis, C. F. Benjamim, H. A. Mata-santos, A. S. Pyrrho et al., Purinergic P2Y12 Receptor Activation in Eosinophils and the Schistosomal Host Response, PLoS ONE, vol.10, 2015.

J. Micklewright, J. Layhadi, and S. Fountain, P2Y12receptor modulation of ADP-evoked intracellular Ca2+signalling in THP-1 human monocytic cells, Brit. J. Pharmacol, vol.175, pp.2483-2491, 2018.

H. Vemulapalli, S. Albayati, V. C. Patwa, D. G. Tilley, A. Y. Tsygankov et al., ADP exerts P2Y12 -dependent and P2Y12 -independent effects on primary human T cell responses to stimulation, J. Cell Commun. Signal, vol.2019, pp.1-16

L. Wang, S. W. Jacobsen, and A. Bengtsson, Erlinge, D. P2 receptor mRNA expression profiles in human lymphocytes, monocytes and CD34+ stem and progenitor cells, BMC Immunol, vol.5, 2004.

M. Kronlage, J. Song, L. Sorokin, K. Isfort, T. Schwerdtle et al., Autocrine Purinergic Receptor Signaling Is Essential for Macrophage Chemotaxis, Sci. Signal, vol.3, p.55, 2010.

X. Su, D. H. Floyd, A. Hughes, J. Xiang, J. G. Schneider et al., The ADP receptor P2RY12 regulates osteoclast function and pathologic bone remodeling, J. Clin. Investig, vol.122, pp.3579-3592, 2012.

P. Ballerini, M. Dovizio, A. Bruno, S. Tacconelli, and P. Patrignani, P2Y12 Receptors in Tumorigenesis and Metastasis, Front. Pharmacol, vol.9, p.66, 2018.

M. F. Reiner, A. Akhmedov, S. Stivala, S. Keller, D. S. Gaul et al., but not clopidogrel, reduces arterial thrombosis via endothelial tissue factor suppression, Cardiovasc. Res, vol.113, pp.61-69, 2016.

H. Haberstock-debic, P. Andre, S. Mills, D. R. Phillips, and P. B. Conley, 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, vol.339, pp.54-61, 2011.

M. Cattaneo, M. Canciani, A. Lecchi, R. Kinlough-rathbone, M. Packham et al., Released adenosine diphosphate stabilizes thrombin-induced human platelet aggregates, Blood, vol.75, pp.1081-1086, 1990.

A. Eckly, J. L. Gendrault, B. Hechler, J. P. Cazenave, and C. Gachet, Differential Involvement of the P2Y1 and P2YT Receptors in the Morphological Changes of Platelet Aggregation, Thromb. Haemost, vol.85, pp.694-701, 2001.

M. Cattaneo, Bleeding manifestations of congenital and drug-induced defects of the platelet P2Y12 receptor for adenosine diphosphate, Thromb. Haemost, vol.105, pp.67-74, 2011.

J. D. Mcfadyen, M. Schaff, and K. Peter, Current and future antiplatelet therapies: Emphasis on preserving haemostasis, Nat. Rev. Cardiol, vol.15, pp.181-191, 2018.

L. Stefanini, C. R. Roden, and W. Bergmeier, CalDAG-GEFI is at the nexus of calcium-dependent platelet activation, Blood, vol.114, pp.2506-2514, 2009.

P. Ohlmann, K. Laugwitz, B. Nürnberg, K. Spicher, G. Schultz et al., The human platelet ADP receptor activates G i2 proteins, Biochem. J, vol.312, pp.775-779, 1995.

J. Daniel, C. Dangelmaier, J. Jin, Y. Kim, and S. Kunapuli, Role of Intracellular Signaling Events in ADP-induced Platelet Aggregation, Thromb. Haemost, vol.82, pp.1322-1326, 1999.

B. Aleil, C. Ravanat, J. Cazenave, G. Rochoux, A. Heitz et al., Flow cytometric analysis of intraplatelet VASP phosphorylation for the detection of clopidogrel resistance in patients with ischemic cardiovascular diseases, J. Thromb. Haemost, vol.3, pp.85-92, 2005.

M. Laine, V. Panagides, C. Frère, T. Cuisset, C. Gouarne et al., Platelet reactivity inhibition following ticagrelor loading dose in patients undergoing percutaneous coronary intervention for acute coronary syndrome, J. Thromb. Haemost, vol.17, pp.2188-2195, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02393751

M. P. Gratacap, J. P. Hérault, C. Viala, A. Ragab, P. Savi et al., Fc?RIIA requires a Gi-dependent pathway for an efficient stimulation of phosphoinositide 3-kinase, calcium mobilization, and platelet aggregation, Blood, vol.96, pp.3439-3446, 2000.

C. Trumel, B. Payrastre, M. Plantavid, B. Hechler, C. Viala et al., 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, vol.94, pp.4156-4165, 1999.

S. Jackson, C. Yap, and K. Anderson, Phosphoinositide 3-kinases and the regulation of platelet function, Biochem. Soc. T, vol.32, pp.387-392, 2004.

M. P. Gratacap, J. Guillermet-guibert, V. Martin, G. Chicanne, H. Tronchère et al., Regulation and roles of PI3K?, a major actor in platelet signaling and functions, Adv. Enzym. Regul, vol.51, pp.106-116, 2010.

C. Dangelmaier, J. Jin, B. Smith, and S. Kunapuli, Potentiation of Thromboxane A2-induced Platelet Secretion by Gi Signaling through the Phosphoinositide-3 Kinase Pathway, Thromb. Haemost, vol.85, pp.341-348, 2001.

C. Leon, M. Alex, A. Klocke, E. Morgenstern, C. Moosbauer et al., Platelet ADP receptors contribute to the initiation of intravascular coagulation, Blood, vol.103, pp.594-600, 2004.

K. Takano, N. Asazuma, K. Satoh, Y. Yatomi, and Y. Ozaki, Collagen-induced generation of platelet-derived microparticles in whole blood is dependent on ADP released from red blood cells and calcium ions, Platelets, vol.15, pp.223-229, 2004.

R. Dorsam, M. Tuluc, and S. Kunapuli, Role of protease-activated and ADP receptor subtypes in thrombin generation on human platelets, J. Thromb. Haemost, vol.2, pp.804-812, 2004.

P. Van-der-meijden, M. Feijge, P. Giesen, M. Huijberts, L. Van-raak et al., Platelet P2Y12 receptors enhance signalling towards procoagulant activity and thrombin generation, Thromb. Haemost, vol.93, pp.1128-1136, 2005.

E. Liverani, M. C. Rico, A. Y. Tsygankov, L. E. Kilpatrick, and S. P. Kunapuli, P2Y12 Receptor Modulates Sepsis-Induced Inflammation, Arterioscler. Thromb. Vasc. Biol, vol.36, pp.961-971, 2016.

G. Lesyk and P. Jurasz, Advances in Platelet Subpopulation Research. Front. Cardiovasc. Med, vol.6, 2019.

Y. Kotova, F. Ataullakhanov, and M. Panteleev, Formation of coated platelets is regulated by the dense granule secretion of adenosine 5 diphosphate acting via the P2Y12 receptor, J. Thromb. Haemost, vol.6, pp.1603-1605, 2008.

C. L. Kempton, M. Hoffman, A. Lenkowski, H. R. Roberts, and D. M. Monroe, COAT Platelet Formation Is P2Y12-Dependent, Blood, vol.104, p.1569, 2004.

Y. Gao, C. Yu, S. Pi, L. Mao, and B. Hu, The role of P2Y12 receptor in ischemic stroke of atherosclerotic origin, Cell. Mol. Life Sci, vol.76, pp.341-354, 2019.

C. Lee, I. Hwang, C. S. Park, H. Lee, D. W. Park et al., Comparison of Differential Expression of P2Y12 Receptor in Culprit Coronary Plaques in Patients With Acute Myocardial Infarction Versus Stable Angina Pectoris, Am. J. Cardiol, vol.108, pp.799-803, 2011.

H. Satonaka, D. Nagata, M. Takahashi, A. Kiyosue, M. Myojo et al., Involvement of P2Y 12 receptor in vascular smooth muscle inflammatory changes via MCP-1 upregulation and monocyte adhesion, Am. J. Physiol Heart C, vol.308, pp.853-861, 2015.

X. Niu, S. Pi, S. Baral, Y. Xia, Q. He et al., P2Y 12 Promotes Migration of Vascular Smooth Muscle Cells Through Cofilin Dephosphorylation During Atherogenesis, Arterioscler. Thromb. Vasc Biol, vol.37, pp.515-524, 2017.

C. Cserép, B. Pósfai, N. Lénárt, R. Fekete, Z. I. László et al., Microglia monitor and protect neuronal function via specialized somatic purinergic junctions, Science, vol.367, pp.528-537, 2020.

T. A. Patente, M. P. Pinho, A. A. Oliveira, G. C. Evangelista, P. C. Bergami-santos et al., Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy, Front. Immunol, vol.9, p.3176, 2019.

L. Liao, Y. Guo, X. Zhuang, W. Li, J. Zou et al., Immunosuppressive Effect of Ticagrelor on Dendritic Cell Function: A New Therapeutic Target of Antiplatelet Agents in Cardiovascular Disease, J. Biomed. Nanotechnol, vol.14, pp.1665-1673, 2018.

P. Diehl, C. Olivier, C. Halscheid, T. Helbing, C. Bode et al., Clopidogrel affects leukocyte dependent platelet aggregation by P2Y12 expressing leukocytes, Basic Res. Cardiol, vol.105, pp.379-387, 2009.

D. H. Suh, H. Trinh, J. N. Liu, L. Pham, S. Park et al., P2Y12 antagonist attenuates eosinophilic inflammation and airway hyperresponsiveness in a mouse model of asthma, J. Cell. Mol. Med, vol.20, pp.333-341, 2016.

R. Czajkowski, L. Lei, P. Sabala, and J. Baranska, 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, vol.513, pp.179-183, 2002.

J. Jin, W. Tomlinson, I. P. Kirk, Y. B. Kim, R. G. Humphries et al., The C6-2B glioma cell P2Y AC receptor is pharmacologically and molecularly identical to the platelet P2Y 12 receptor, Brit. J. Pharmacol, vol.133, pp.521-528, 2001.

S. Sarangi, A. Pandey, A. L. Papa, P. Sengupta, J. Kopparam et al., P2Y12 receptor inhibition augments cytotoxic effects of cisplatin in breast cancer, Med Oncol, vol.30, 2013.

A. I. Fahmy, M. A. Mekkawy, and A. Abou-ali, 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, vol.57, pp.175-181, 2019.

R. C. Becker, J. Bassand, A. Budaj, D. M. Wojdyla, S. K. James et al., Bleeding complications with the P2Y12 receptor antagonists clopidogrel and ticagrelor in the PLATelet inhibition and patient Outcomes (PLATO) trial, Eur. Heart J, vol.32, pp.2933-2944, 2011.

A. Godier, P. Fontana, S. Motte, A. Steib, F. Bonhomme 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, vol.37, pp.379-389, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02342074

M. Cattaneo and . Antagonists, Platelets, pp.937-956, 2019.
URL : https://hal.archives-ouvertes.fr/hal-00478626

L. Gross, D. Aradi, and D. Sibbing, Pharmacology: Inhibitors of P2Y12. Platelets in Thrombotic and Non-Thrombotic Disorders, pp.1253-1267, 2017.

L. Wallentin, P2Y(12) inhibitors: Differences in properties and mechanisms of action and potential consequences for clinical use, Eur. Heart J, vol.30, 1964.

P. Sharis, C. Cannon, and J. Loscalzo, The Antiplatelet Effects of Ticlopidine and Clopidogrel, Ann. Intern. Med, vol.129, 1998.

P. A. Gurbel, K. P. Bliden, K. Butler, U. S. Tantry, T. Gesheff 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, vol.120, pp.2577-2585, 2009.

S. E. Husted, R. F. Storey, K. Bliden, U. S. Tantry, L. Høimark et al., Pharmacokinetics and Pharmacodynamics of Ticagrelor in Patients with Stable Coronary Artery Disease, Clin. Pharm, vol.51, pp.397-409, 2012.

S. Husted, H. Emanuelsson, S. Heptinstall, P. Sandset, M. Wickens et al., 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, vol.27, pp.1038-1047, 2006.

R. F. Storey, S. Husted, R. A. Harrington, S. Heptinstall, R. G. Wilcox 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, vol.50, pp.1852-1856, 2007.

P. P. Dobesh and J. H. Oestreich, Ticagrelor: Pharmacokinetics, Pharmacodynamics, Clinical Efficacy, and Safety, Pharm. J. Hum. Pharmacol. Drug Ther, vol.34, pp.1077-1090, 2014.

F. Franchi, F. Rollini, A. Muñiz-lozano, J. Cho, and D. J. Angiolillo, Cangrelor: A review on pharmacology and clinical trial development, Expert Rev. Cardiovasc. Ther, vol.11, pp.1279-1291, 2013.

N. A. Farid, A. Kurihara, and S. A. Wrighton, Metabolism and Disposition of the Thienopyridine Antiplatelet Drugs Ticlopidine, Clopidogrel, and Prasugrel in Humans, J. Clin. Pharmacol, vol.50, pp.126-142, 2010.

J. S. Hulot, A. Bura, E. Villard, M. Azizi, V. Remones et al., Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects, Blood, vol.108, pp.2244-2247, 2006.

K. Hagihara, M. Kazui, A. Kurihara, H. Iwabuchi, M. Ishikawa et al., Biotransformation of prasugrel, a novel thienopyridine antiplatelet agent, to the pharmacologically active metabolite, Drug Metab. Dispos. Biol. Fate Chem, vol.38, pp.898-904, 2010.

L. Wallentin, C. Varenhorst, S. James, D. Erlinge, O. Braun et al., 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, vol.29, pp.21-30, 2007.

A. Sugidachi, T. Ogawa, A. Kurihara, K. Hagihara, J. Jakubowski et al., 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, vol.5, pp.1545-1551, 2007.

K. Hoffmann, D. Lutz, J. Straßburger, Y. Baqi, C. Müller et al., Competitive mode and site of interaction of ticagrelor at the human platelet P2Y12 -receptor, J. Thromb. Haemost, vol.12, pp.1898-1905, 2014.

V. J. Giezen, L. Nilsson, P. Betsson, B. Wissing, F. Giordanetto et al., Ticagrelor binds to human P2Y 12 independently from ADP but antagonizes ADP-induced receptor signaling and platelet aggregation, J. Thromb. Haemost, vol.7, pp.1556-1565, 2009.

R. Teng, S. Oliver, M. A. Hayes, and K. Butler, Absorption, Distribution, Metabolism, and Excretion of Ticagrelor in Healthy Subjects, Drug Metab. Dispos, vol.38, pp.1514-1521, 2010.

M. Löffler, J. C. Morote-garcia, S. A. Eltzschig, I. R. Coe, and H. K. Eltzschig, Physiological Roles of Vascular Nucleoside Transporters, Arterioscler. Thromb. Vasc Biol, vol.27, pp.1004-1013, 2007.

R. Aungraheeta, A. Conibear, M. Butler, E. Kelly, S. Nylander et al., Inverse agonism at the P2Y12 receptor and ENT1 transporter blockade contribute to platelet inhibition by ticagrelor, Blood, vol.128, pp.2717-2728, 2016.

M. Cattaneo, R. Schulz, and S. Nylander, Adenosine-mediated effects of ticagrelor: Evidence and potential clinical relevance, J. Am. Coll. Cardiol, vol.63, pp.2503-2509, 2014.

K. Ow, W. A. Parker, M. M. Porter, J. Hanson, H. M. Judge et al., Offset of ticagrelor prior to coronary artery bypass graft surgery for acute coronary syndromes: Effects on platelet function and cellular adenosine uptake, Platelets, vol.2020, pp.1-7

L. Ortega-paz, S. Brugaletta, S. Ariotti, M. K. Akkerhuis, A. Karagiannis et al., On behalf of the investigators. Adenosine and Ticagrelor Plasma Levels in Patients With and Without Ticagrelor-Related Dyspnea, Circulation, vol.138, pp.646-648, 2018.

C. Garcia, A. Maurel-ribes, M. Nauze, D. N'guyen, L. O. Martinez et al., Deciphering biased inverse agonism of cangrelor and ticagrelor at P2Y12 receptor, Cell. Mol. Life Sci, vol.76, pp.561-576, 2019.

P. Lancellotti, L. Musumeci, N. Jacques, L. Servais, E. Goffin et al., Antibacterial Activity of Ticagrelor in Conventional Antiplatelet Dosages Against Antibiotic-Resistant Gram-Positive Bacteria, JAMA Cardiol, vol.4, pp.596-599, 2019.

D. J. Angiolillo, M. S. Firstenberg, M. J. Price, P. E. Tummala, M. Hutyra et al., Bridging antiplatelet therapy with cangrelor in patients undergoing cardiac surgery: A randomized controlled trial, JAMA J. Am. Med Assoc, vol.307, pp.265-274, 2012.

P. Steg, D. L. Bhatt, C. W. Hamm, G. W. Stone, M. C. Gibson et al., Effect of cangrelor on periprocedural outcomes in percutaneous coronary interventions: A pooled analysis of patient-level data, Lancet Lond Engl, vol.382, 1981.

L. Ma and A. Dorling, The roles of thrombin and protease-activated receptors in inflammation, Semin. Immunopathol, vol.34, pp.63-72, 2012.

M. Levi, T. Van-der-poll, and H. R. Büller, Bidirectional Relation between Inflammation and Coagulation, Circulation, vol.109, pp.2698-2704, 2004.

A. Naldini, D. H. Carney, A. Pucci, A. Pasquali, and F. Carraro, Thrombin regulates the expression of proangiogenic cytokines via proteolytic activation of protease-activated receptor-1, Gen. Pharmacol. Vasc. Syst, vol.35, pp.255-259, 2000.

A. Naldini, D. H. Carney, V. Bocci, K. D. Klimpel, M. Asuncion et al., Thrombin Enhances T Cell Proliferative Responses and Cytokine Production, Cell. Immunol, vol.147, pp.367-377, 1993.

K. Johnson, Y. Choi, E. Degroot, I. Samuels, A. Creasey et al., Potential mechanisms for a proinflammatory vascular cytokine response to coagulation activation, J. Immunol. Baltim. Md, vol.160, pp.5130-5135, 19501998.

J. L. Strande and S. A. Phillips, Thrombin increases inflammatory cytokine and angiogenic growth factor secretion in human adipose cells in vitro, J. Inflamm, vol.6, 2009.

R. Shankar, C. De-la-motte, E. Poptic, and P. Dicorleto, 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, vol.269, pp.13936-13941, 1994.

T. Iba and J. H. Levy, Inflammation and thrombosis: Roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis, J. Thromb. Haemost, vol.16, pp.231-241, 2018.

N. B. Norgard, C. L. Hann, and G. L. Dale, Cangrelor Attenuates Coated-Platelet Formation, Clin. Appl. Thromb. Hemost, vol.15, pp.177-182, 2008.

N. B. Norgard, S. Saya, C. Hann, T. Hennebry, E. Schechter et al., Clopidogrel attenuates coated-platelet production in patients undergoing elective coronary catheterization, J. Cardiovasc. Pharm, vol.52, pp.536-539, 2008.

P. J. Hérault, F. Dol, C. Gaich, A. Bernat, and M. J. Herbert, Effect of Clopidogrel on Thrombin Generation in Platelet-Rich Plasma in the Rat, Thromb. Haemost, vol.81, pp.957-960, 1999.

X. Wang, H. Deng, T. Li, S. Miao, Z. Xiao et al., Clopidogrel reduces lipopolysaccharide-induced inflammation and neutrophil-platelet aggregates in an experimental endotoxemic model, J. Biochem. Mol. Toxicol, vol.33, 2019.

M. D. Turner, B. Nedjai, T. Hurst, and D. J. Pennington, Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease, Biochim. Biophys. Acta Mol. Cell Res, vol.1843, pp.2563-2582, 2014.

J. Bester and E. Pretorius, Effects of IL-1?, IL-6 and IL-8 on erythrocytes, platelets and clot viscoelasticity. Sci Rep. UK, vol.6, 2016.

J. A. Lumadue, S. M. Lanzkron, S. D. Kennedy, D. T. Kuhl, and T. S. Kickler, Cytokine Induction of Platelet Activation. Am. J. Clin. Pathol, vol.106, pp.795-798, 1996.

A. Assinger, W. C. Schrottmaier, M. Salzmann, and J. Rayes, Platelets in Sepsis: An Update on Experimental Models and Clinical Data, Front. Immunol, vol.10, 1687.

I. Slaba and P. Kubes, Platelets in Thrombotic and Non-Thrombotic Disorders, pp.489-512, 2017.

V. Sanguigni, D. Ferro, P. Pignatelli, M. Ben, T. Nadia et al., CD40 ligand enhances monocyte tissue factor expression and thrombin generation via oxidative stress in patients with hypercholesterolemia, J. Am. Coll. Cardiol, vol.45, pp.35-42, 2005.

E. Liverani, L. E. Kilpatrick, A. Y. Tsygankov, and S. P. Kunapuli, The role of P2Y 12 receptor and activated platelets during inflammation, Curr. Drug Targets, vol.15, pp.720-728, 2014.

T. R. Sexton, G. Zhang, T. E. Macaulay, L. A. Callahan, R. Charnigo et al., Ticagrelor Reduces Thromboinflammatory Markers in Patients With Pneumonia, JACC. Basic Transl. Sci, vol.3, pp.435-449, 2018.

S. Hagiwara, H. Iwasaka, A. Hasegawa, M. Oyama, R. Imatomi et al., Adenosine Diphosphate Receptor Antagonist Clopidogrel Sulfate Attenuates LPS-Induced Systemic Inflammation in a Rat Model, Shock, vol.35, pp.289-292, 2011.

M. R. Thomas, S. N. Outteridge, R. A. Ajjan, F. Phoenix, G. K. Sangha et al., Platelet P2Y12 Inhibitors Reduce Systemic Inflammation and Its Prothrombotic Effects in an Experimental Human Model, Arterioscler. Thromb. Vasc Biol, vol.35, pp.2562-2570, 2015.

M. R. Thomas and R. F. Storey, Effect of P2Y12 inhibitors on inflammation and immunity, Thromb. Haemost, vol.114, pp.490-497, 2015.

J. Rossaint, A. Margraf, and A. Zarbock, Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation, Front. Immunol, vol.9, p.2712, 2018.

Z. Li, S. S. Smyth, and . Platelets, , pp.295-310, 2019.

V. Evangelista, L. Totani, A. A. Manfredi, and N. Maugeri, Platelets in Thrombotic and Non-Thrombotic Disorders, pp.407-433, 2017.

V. Papayannopoulos, Neutrophil extracellular traps in immunity and disease, Nat. Rev. Immunol, vol.18, pp.134-147, 2018.

O. Elaskalani, N. Razak, and P. Metharom, Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones, Cell Commun. Signal, vol.16, 2018.

J. Etulain, K. Martinod, S. Wong, S. M. Cifuni, M. Schattner et al., P-selectin promotes neutrophil extracellular trap formation in mice, Blood, vol.126, pp.242-246, 2015.

A. Carestia, T. Kaufman, and M. Schattner, Platelets: New Bricks in the Building of, Neutrophil Extracellular Traps. Front. Immunol, 2016.

A. Z. Zucoloto and C. N. Jenne, Platelet-Neutrophil Interplay: Insights Into Neutrophil Extracellular Trap (NET)-Driven Coagulation in Infection, Front. Cardiovasc. Med, vol.6, 2019.

N. M. Kazzaz, G. Sule, and J. S. Knight, Intercellular Interactions as Regulators of NETosis, Front. Immunol, vol.7, p.453, 2016.

M. Rahman, D. Gustafsson, Y. Wang, H. Thorlacius, and O. Braun, Ticagrelor reduces neutrophil recruitment and lung damage in abdominal sepsis, Platelets, vol.25, pp.257-263, 2014.

V. Evangelista, S. Manarini, G. Dell'elba, N. Martelli, E. Napoleone et al., Clopidogrel inhibits platelet-leukocyte adhesion and plateletdependent leukocyte activation, Thromb. Haemost, vol.94, pp.568-577, 2005.

H. M. Judge, R. J. Buckland, A. Sugidachi, J. A. Jakubowski, and R. F. Storey, The active metabolite of prasugrel effectively blocks the platelet P2Y12 receptor and inhibits procoagulant and pro-inflammatory platelet responses, Platelets, vol.19, pp.125-133, 2008.

R. Storey, H. Judge, R. Wilcox, and S. Heptinstall, 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, vol.88, pp.488-494, 2002.

U. Klinkhardt, Clopidogrel but not aspirin reduces P-selectin expression and formation of platelet-leukocyte aggregates in patients with atherosclerotic vascular disease, Clin. Pharmacol. Ther, vol.73, pp.232-241, 2003.

Z. Xiao and P. Théroux, Clopidogrel inhibits platelet-leukocyte interactions and thrombin receptor agonist peptide-induced platelet activation in patients with an acute coronary syndrome, J. Am. Coll. Cardiol, vol.43, 1982.

S. Vigano, D. Alatzoglou, M. Irving, C. Ménétrier-caux, C. Caux et al., Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function, Front. Immunol, vol.10, 2019.

N. Wolska and M. Rozalski, Blood Platelet Adenosine Receptors as Potential Targets for Anti-Platelet Therapy, Int. J. Mol. Sci, vol.20, 2019.

M. Boncler, J. Wzorek, N. Wolska, D. Polak, C. Watala et al., Adenosine receptor agonists deepen the inhibition of platelet aggregation by P2Y12 antagonists, Vasc. Pharmacol, vol.113, pp.47-56, 2018.

J. Winning, J. Reichel, Y. Eisenhut, J. Hamacher, M. Kohl et al., Anti-platelet drugs and outcome in severe infection: Clinical impact and underlying mechanisms, Platelets, vol.20, pp.50-57, 2009.

R. F. Storey, S. K. James, A. Siegbahn, C. Varenhorst, C. Held et al., Lower mortality following pulmonary adverse events and sepsis with ticagrelor compared to clopidogrel in the PLATO study, Platelets, vol.25, pp.517-525, 2013.

M. J. Tsai, S. M. Ou, C. J. Shih, P. Chao, L. F. Wang et al., Association of prior antiplatelet agents with mortality in sepsis patients: A nationwide population-based cohort study, Intensive Care Med, vol.41, pp.806-813, 2015.

T. M. Laidlaw, K. N. Cahill, J. Cardet, K. Murphy, J. Cui et al., A trial of P2Y12 receptor inhibition with prasugrel identifies a potentially distinct endotype of patients with aspirin-exacerbated respiratory disease, J. Allergy Clin. Immun, vol.143, pp.316-324, 2018.

F. Lussana, D. F. Marco, S. Terraneo, M. Parati, C. Razzari et al., Effect of prasugrel in patients with asthma: Results of PRINA, a randomized, double-blind, placebo-controlled, cross-over study, J. Thromb. Haemost, vol.13, pp.136-141, 2015.

S. Raposeiras-roubin, E. Abu-assi, I. Muñoz-pousa, M. Cespón-fernández, R. Cobas-paz et al., Risk of cancer after an acute coronary syndrome according to the type of P2Y12 inhibitor, Thromb. Res, vol.174, pp.51-58, 2019.

A. Leader, R. Zelikson-saporta, D. Pereg, G. Spectre, U. Rozovski et al., The Effect of Combined Aspirin and Clopidogrel Treatment on Cancer Incidence, Am. J. Med, vol.130, pp.826-832, 2017.

B. M. Hicks, L. J. Murray, C. Hughes, and C. R. Cardwell, Clopidogrel use and cancer-specific mortality: A population-based cohort study of colorectal, breast and prostate cancer patients, Pharmacoepidemiol. Drug Saf, vol.24, pp.830-840, 2015.

S. Elmariah, G. Doros, O. R. Benavente, D. L. Bhatt, S. J. Connolly 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, vol.11, p.5795, 2018.

A. Rodríguez-miguel, L. A. García-rodríguez, M. Gil, H. Montoya, S. Rodríguez-martín et al., Clopidogrel and Low-Dose Aspirin, Alone or Together, Reduce Risk of Colorectal Cancer, Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc, vol.17, pp.2024-2033, 2019.

R. S. Hotchkiss, L. L. Moldawer, S. M. Opal, K. Reinhart, I. R. Turnbull et al., Sepsis and septic shock, Nat. Rev. Dis. Primers, vol.2, 2016.

T. Van-der-poll, F. L. Van-de-veerdonk, B. P. Scicluna, and M. G. Netea, The immunopathology of sepsis and potential therapeutic targets, Nat. Rev. Immunol, vol.17, pp.407-420, 2017.

F. Vardon-bounes, S. Ruiz, M. P. Gratacap, C. Garcia, B. Payrastre et al., Platelets Are Critical Key Players in Sepsis, Int. J. Mol. Sci, vol.20, 2019.

Y. Wang, Y. Ouyang, B. Liu, X. Ma, and R. Ding, Platelet activation and antiplatelet therapy in sepsis: A narrative review, Thromb. Res, vol.166, pp.28-36, 2018.

M. A. Matthay, R. L. Zemans, G. A. Zimmerman, Y. M. Arabi, J. R. Beitler et al., Acute respiratory distress syndrome, Nat. Rev. Dis. Primers, vol.5, 2019.

E. A. Middleton, M. T. Rondina, H. Schwertz, and G. A. Zimmerman, Amicus or Adversary Revisited: Platelets in Acute Lung Injury and Acute Respiratory Distress Syndrome, Am. J. Resp. Cell Mol, vol.59, pp.18-35, 2018.

F. A. Bozza, A. Ah, A. S. Weyrich, and G. A. Zimmerman, Amicus or Adversary. Am. J. Resp. Cell Mol, vol.40, pp.123-134, 2009.

L. Wallentin, R. C. Becker, A. Budaj, C. P. Cannon, H. Emanuelsson et al., Ticagrelor versus clopidogrel in patients with acute coronary syndromes, N. Engl. J. Med, vol.361, pp.1045-1057, 2009.

T. Takeda, H. Morita, H. Saito, K. Matsumoto, and A. Matsuda, Recent advances in understanding the roles of blood platelets in the pathogenesis of allergic inflammation and bronchial asthma, Allergol. Int, vol.67, pp.326-333, 2017.

M. Idzko, S. Pitchford, and C. Page, Role of platelets in allergic airway inflammation, J. Allergy Clin. Immun, vol.135, pp.1416-1423, 2015.

K. Kowal, A. Pampuch, O. Kowal-bielecka, L. Dubuske, and A. Bodzenta-?ukaszyk, Platelet activation in allergic asthma patients during allergen challenge with Dermatophagoides pteronyssinus, Clin. Exp. Allergy, vol.36, pp.426-432, 2006.

M. Gawaz and O. Borst, 26 the Role of Platelets in Atherothrombosis, Platelets, pp.459-467, 2019.

D. Lievens and P. Von-hundelshausen, Platelets in atherosclerosis, Thromb. Haemost, vol.106, pp.827-838, 2011.

H. Lebas, K. Yahiaoui, R. Martos, and Y. Boulaftali, Platelets Are at the Nexus of Vascular Diseases, Front. Cardiovasc. Med, vol.6, 2019.

D. Li, Y. Wang, L. Zhang, X. Luo, J. Li 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, vol.32, pp.81-89, 2012.

C. Heim, J. Gebhardt, M. Ramsperger-gleixner, J. Jacobi, M. Weyand et al., Clopidogrel significantly lowers the development of atherosclerosis in ApoE-deficient mice in vivo, vol.31, pp.783-794, 2015.

A. Afek, E. Kogan, S. Maysel-auslender, A. Mor, E. Regev et al., Clopidogrel attenuates atheroma formation and induces a stable plaque phenotype in apolipoprotein E knockout mice, Microvasc. Res, vol.77, pp.364-369, 2009.

X. Xia, J. Li, X. Liang, S. Zhang, T. Liu et al., Ticagrelor suppresses oxidized low-density lipoprotein-induced endothelial cell apoptosis and alleviates atherosclerosis in ApoE-/-mice via downregulation of PCSK9, Mol. Med. Rep, vol.19, pp.1453-1462, 2018.

B. Ganbaatar, D. Fukuda, H. Salim, S. Nishimoto, K. Tanaka et al., Ticagrelor, a P2Y12 antagonist, attenuates vascular dysfunction and inhibits atherogenesis in apolipoprotein-E-deficient mice, Atherosclerosis, vol.275, pp.124-132, 2018.

L. E. West, T. Steiner, H. M. Judge, S. E. Francis, and R. F. Storey, Vessel wall, not platelet, P2Y12 potentiates early atherogenesis, Cardiovasc. Res, vol.102, pp.429-435, 2014.

F. R. Greten and S. I. Grivennikov, Inflammation and Cancer: Triggers, Mechanisms, and Consequences, Immunity, vol.51, pp.27-41, 2019.

M. Haemmerle, R. L. Stone, D. G. Menter, V. Afshar-kharghan, and A. K. Sood, The Platelet Lifeline to Cancer: Challenges and Opportunities, Cancer Cell, vol.33, pp.965-983, 2018.

M. Cho, K. Noh, M. Haemmerle, D. Li, H. Park et al., Role of ADP receptors on platelets in the growth of ovarian cancer, vol.130, pp.1235-1242, 2017.

A. J. Gareau, C. Brien, S. Gebremeskel, R. S. Liwski, B. Johnston et al., Ticagrelor inhibits platelet-tumor cell interactions and metastasis in human and murine breast cancer, Clin. Exp. Metastasis, vol.35, pp.25-35, 2018.

S. Gebremeskel, T. Levatte, R. S. Liwski, B. Johnston, and M. Bezuhly, The reversible P2Y12 inhibitor ticagrelor inhibits metastasis and improves survival in mouse models of cancer, Int. J. Cancer, vol.136, pp.234-240, 2015.

Y. Wang, Y. Sun, D. Li, L. Zhang, K. Wang et al., Platelet P2Y12 is involved in murine pulmonary metastasis, PLoS ONE, vol.8, 2013.

M. Geranpayehvaghei, Q. Shi, B. Zhao, S. Li, J. Xu et al., Targeting Delivery of Platelets Inhibitor to Prevent Tumor Metastasis, Bioconjug. Chem, vol.30, pp.2349-2357, 2019.

A. M. Algra and P. M. Rothwell, Effects of regular aspirin on long-term cancer incidence and metastasis: A systematic comparison of evidence from observational studies versus randomised trials, Lancet Oncol, vol.13, pp.518-527, 2012.

. Caprie-steering-committee, A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE), Lancet, vol.348, pp.1329-1339, 1996.

D. L. Bhatt, K. A. Fox, W. Hacke, P. B. Berger, H. R. Black et al., Clopidogrel and Aspirin versus Aspirin Alone for the Prevention of Atherothrombotic Events, N. Engl. J. Med, vol.354, pp.1706-1717, 2006.

S. D. Wiviott, E. Braunwald, C. H. Mccabe, G. Montalescot, W. Ruzyllo et al., Prasugrel versus Clopidogrel in Patients with Acute Coronary Syndromes, N. Engl. J. Med, vol.357, 2001.

M. T. Roe, P. W. Armstrong, K. A. Fox, H. D. White, D. Prabhakaran et al., Prasugrel versus Clopidogrel for Acute Coronary Syndromes without Revascularization, N. Engl J. Med, vol.367, pp.1297-1309, 2012.

L. Mauri, D. J. Kereiakes, R. W. Yeh, P. Driscoll-shempp, D. E. Cutlip et al., Twelve or 30 months of dual antiplatelet therapy after drug-eluting stents, N. Engl. J. Med, vol.371, pp.2155-2166, 2014.

M. P. Bonaca, D. L. Bhatt, M. Cohen, P. Steg, R. F. Storey et al., Long-Term Use of Ticagrelor in Patients with Prior Myocardial Infarction, N. Engl. J. Med, vol.372, pp.1791-1800, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01934996

V. L. Serebruany, Ticagrelor shift from PLATO to PEGASUS: Vanished mortality benefit, excess cancer deaths, massive discontinuations, and overshooting target events, Int. J. Cardiol, vol.201, pp.508-512, 2015.

, FDA Review Finds Long-Term Treatment with Blood-Thinning Medicine Plavix (Clopidogrel) Does not Change Risk of Death, vol.FDA, 2014.

V. Serebruany, M. Kim, C. Thevathasan, and T. Marciniak, 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, vol.2, pp.28-32, 2018.

R. Kotronias, C. Kwok, C. Wong, T. Kinnaird, A. Zaman et al., Cancer Event Rate and Mortality with Thienopyridines: A Systematic Review and Meta-Analysis, © 2020 by the authors. Licensee MDPI, vol.40, pp.229-240, 2016.