In vitro assessment and phase I randomized clinical trial of anfibatide a snake venom derived anti-thrombotic agent targeting human platelet GPIbα

Benjamin Xiaoyi Li, Xiangrong Dai, Xiaohong Ruby Xu, Reheman Adili, Miguel Antonio Dias Neves, Xi Lei, Chuanbin Shen, Guangheng Zhu, Yiming Wang, Hui Zhou, Yan Hou, Tiffany Ni, Yfke Pasman, Zhongqiang Yang, Fang Qian, Yanan Zhao, Yongxiang Gao, Jing Liu, Maikun Teng, Alexandra H Marshall, Eric G Cerenzia, Mandy Lokyee Li, Heyu Ni, Benjamin Xiaoyi Li, Xiangrong Dai, Xiaohong Ruby Xu, Reheman Adili, Miguel Antonio Dias Neves, Xi Lei, Chuanbin Shen, Guangheng Zhu, Yiming Wang, Hui Zhou, Yan Hou, Tiffany Ni, Yfke Pasman, Zhongqiang Yang, Fang Qian, Yanan Zhao, Yongxiang Gao, Jing Liu, Maikun Teng, Alexandra H Marshall, Eric G Cerenzia, Mandy Lokyee Li, Heyu Ni

Abstract

The interaction of platelet GPIbα with von Willebrand factor (VWF) is essential to initiate platelet adhesion and thrombosis, particularly under high shear stress conditions. However, no drug targeting GPIbα has been developed for clinical practice. Here we characterized anfibatide, a GPIbα antagonist purified from snake (Deinagkistrodon acutus) venom, and evaluated its interaction with GPIbα by surface plasmon resonance and in silico modeling. We demonstrated that anfibatide interferds with both VWF and thrombin binding, inhibited ristocetin/botrocetin- and low-dose thrombin-induced human platelet aggregation, and decreased thrombus volume and stability in blood flowing over collagen. In a single-center, randomized, and open-label phase I clinical trial, anfibatide was administered intravenously to 94 healthy volunteers either as a single dose bolus, or a bolus followed by a constant rate infusion of anfibatide for 24 h. Anfibatide inhibited VWF-mediated platelet aggregation without significantly altering bleeding time or coagulation. The inhibitory effects disappeared within 8 h after drug withdrawal. No thrombocytopenia or anti-anfibatide antibodies were detected, and no serious adverse events or allergic reactions were observed during the studies. Therefore, anfibatide was well-tolerated among healthy subjects. Interestingly, anfibatide exhibited pharmacologic effects in vivo at concentrations thousand-fold lower than in vitro, a phenomenon which deserves further investigation.Trial registration: Clinicaltrials.gov NCT01588132.

Conflict of interest statement

The authors declare the following competing interests: this research and the work in Lei et al. and Hou et al. was partially funded by Lee’s Pharmaceutical Holdings limited. B.X.L., X.D., Z.Y., F.Q., and M.L.L. are supported by Lee’s Pharmaceutical Holdings Limited and/or Zhaoke Pharmaceutical Co. Limited. The other authors declare they have no actual or potential competing interests.

Figures

Figure 1
Figure 1
Purification and structure of anfibatide with GPIbα. (A) MALDI-TOF mass spectrometry showed a mass to charge ratio (m/z) of 29,799.7. Three-dimensional models of (B) structure of anfibatide (purple) integrated with GPIb (orange) and (C) VWF-A1 domain (green) and GPIbα complex from PDB entry 1SQ0 (https://www.rcsb.org/structure/1SQ0). (D) α-Thrombin (blue) and GPIbα complex from PDB entry 1OOK (https://www.rcsb.org/structure/1OOK). Red arrows point to the sulfotyrosine region of GPIbα, the α-thrombin binding site. Protein-complex figures generated using Schrodinger PyMol 2 software (https://pymol.org/2/).
Figure 2
Figure 2
Anfibatide binding to GPIbα prevented VWF and thrombin binding as well as platelet aggregation induced by ristocetin/VWF and low-dose thrombin, but had no effect on blood clotting. (A) SPR analysis of anfibatide binding to disulfide-linked dimeric GPIbαN-Long (a chimera of human GPIbα residues − 2 to 288 with 133 residues of SV40 large T antigen) either with wild-type (WT) GPIbα sequence or with Tyr to Phe substitution of residues 276, 278 and 279 (3Y/F). GPIbαN-Long was captured onto the SPR chip by an immobilized anti-SV40-T mAb, followed by anfibatide at increasing concentrations. Data are presented as the ratio of anfibatide/GPIbαN-Long mass bound to the SPR chip during the equilibrium phase (prior to dissociation) and are fit to a one-site ligand binding model. Mass ratio ± SD, N = 3, Black: GPIbαN-Long WT; Orange: GPIbαN-Long 3Y/F. (B) Inhibition of α-thrombin (BP-αFIIa), VWF A1 domain or mAb LJ-Ib10 binding to GPIbα on human washed platelets by increasing anfibatide concentrations. Data are presented as % binding, relative to the binding of the ligand at a concentration equal to the KD of the ligand in the absence of anfibatide. The data was fit to the Cheng–Prusoff transformation. Ligand binding % ± SD, N = 3, Black: BP-αFIIa; Orange: LJ-Ib10; Green: rVWF. (C) Platelet aggregation was induced by ristocetin (1.2 mg/mL) (P < 0.001), ADP (20 μM) (P > 0.05), TRAP (500 μM) (P > 0.05), or collagen (10 µg/mL) (P > 0.05) in anfibatide-treated (6 µg/mL) PRP or by thrombin (0.1–1 U/mL) in gel-filtered platelets. Curves are from representative light transmission aggregometry plots. (D) Clot formation was measured by thromboelastography in anfibatide-treated whole blood. Anfibatide (6 µg/mL) did not significantly alter the time to initial clot formation (R time, min ± SEM) or maximum clot strength (MA ± SEM). NS, no significant difference. Red: anfibatide-treated plasma. Black: untreated plasma (N = 6).
Figure 3
Figure 3
Anfibatide inhibited platelet adhesion, aggregation and thrombus formation and dissolved preformed thrombi under flow conditions. Platelets in heparin-anticoagulated whole blood from healthy volunteers were fluorescently labeled with DiOC6 before perfusion over collagen at the wall shear rate of 1500 s−1 (A) or 300 s−1 (B) with or without anfibatide (6 µg/mL). (C) Control blood was first perfused at 1500 s−1 for 4 min to form thrombi; then, perfusion was continued with control or anfibatide-treated (6 µg/mL) blood. Representative images of fluorescent platelets (Left) are shown along with plots of the platelet mean (± SEM) fluorescence intensity as a function of time (Right; N = 12). P < 0.01 between control and treatment groups in all three figures. Two-tailed Student’s t-test was used to test for significant differences between 2 groups.
Figure 4
Figure 4
Study design. All randomly-assigned participants were included in the primary outcome analysis. Dose units are μg per 60 kg body weight. N: number of volunteers per group.
Figure 5
Figure 5
Plasma concentration–time curves of anfibatide in healthy volunteers after single intravenous bolus injection at dose levels of 1, 1.5, 2, 3, 4 and 5 µg/60 kg, respectively (Time = 0–8 h, A; Time = 0–48 h, B). Mean ± SEM.
Figure 6
Figure 6
Anfibatide inhibited ristocetin-induced platelet aggregation. Human platelet aggregation induced by ristocetin was studied by platelet aggregometry in single (A, N = 8–9/each) and multiple dose groups (B, N = 6–12/each). The mean inhibitory rate of anfibatide on platelet aggregation over time has been shown. CRI, constant rate infusion; Mean ± SEM.
Figure 7
Figure 7
Anfibatide had no significant effect on coagulation measured by prothrombin time (A), thrombin time (B), activated thromboplastin time (C), and international normalized ratio (D). anfibatide did not significantly change circulating d-dimers (E), representing fibrinolysis. (CRI indicates constant rate infusion). Mean ± SD.
Figure 8
Figure 8
Anfibatide did not significantly prolong bleeding time. Bleeding time was monitored in both single (A, N = 8–9/each) and multiple dose groups (B, N = 6–12/each), and each bleeding time measured was determined to the nearest 30 s. Majority of the subjects in the single dose groups (except 5 subjects) had a lower bound close to 4 min, while those in the multiple dose groups had a lower bound ranging from 2 to 4 min. None of the subjects in the single and multiple dose groups had a bleeding time of more than 9 min. Bleeding time of all subjects was within the normal range of 2 to 9 min. CRI: constant rate infusion.

References

    1. Virani SS, et al. Heart disease and stroke statistics-2020 update: A report from the American Heart Association. Circulation. 2020;141:e139–e596. doi: 10.1161/CIR.0000000000000757.
    1. Ruggeri ZM. Platelets in atherothrombosis. Nat. Med. 2002;8:1227–1234. doi: 10.1038/nm1102-1227.
    1. Xu XR, et al. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit. Rev. Clin. Lab. Sci. 2016;53:409–430. doi: 10.1080/10408363.2016.1200008.
    1. Xu XR, et al. Apolipoprotein A-IV binds alphaIIbbeta3 integrin and inhibits thrombosis. Nat. Commun. 2018;9:3608. doi: 10.1038/s41467-018-05806-0.
    1. Jackson SP. The growing complexity of platelet aggregation. Blood. 2007;109:5087–5095. doi: 10.1182/blood-2006-12-027698.
    1. Bergmeier W, et al. The role of platelet adhesion receptor GPIbα far exceeds that of its main ligand, von Willebrand factor, in arterial thrombosis. Proc. Natl. Acad. Sci. USA. 2006;103:16900–16905. doi: 10.1073/pnas.0608207103.
    1. Ni H, et al. Persistence of platelet thrombus formation in arterioles of mice lacking both von Willebrand factor and fibrinogen. J. Clin. Investig. 2000;106:385–392. doi: 10.1172/JCI9896.
    1. Yang H, et al. Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo. J. Thromb. Haemost. 2006;4:2230–2237. doi: 10.1111/j.1538-7836.2006.02116.x.
    1. Reheman A, et al. Plasma fibronectin depletion enhances platelet aggregation and thrombus formation in mice lacking fibrinogen and von Willebrand factor. Blood. 2009;113:1809–1817. doi: 10.1182/blood-2008-04-148361.
    1. Wang Y, Gallant RC, Ni H. Extracellular matrix proteins in the regulation of thrombus formation. Curr. Opin. Hematol. 2016;23:280–287. doi: 10.1097/MOH.0000000000000237.
    1. Strony J, Beaudoin A, Brands D, Adelman B. Analysis of shear stress and hemodynamic factors in a model of coronary artery stenosis and thrombosis. Am. J. Physiol. 1993;265:H1787–H1796.
    1. Jackson SP, Schoenwaelder SM. Antiplatelet therapy: In search of the 'magic bullet'. Nat. Rev. Drug Discov. 2003;2:775–789. doi: 10.1038/nrd1198.
    1. Patrono C, Garcia Rodriguez LA, Landolfi R, Baigent C. Low-dose aspirin for the prevention of atherothrombosis. N. Engl. J. Med. 2005;353:2373–2383. doi: 10.1056/NEJMra052717.
    1. Reheman A, Xu X, Reddy EC, Ni H. Targeting activated platelets and fibrinolysis: Hitting two birds with one stone. Circ. Res. 2014;114:1070–1073. doi: 10.1161/CIRCRESAHA.114.303600.
    1. Xu XR, et al. Platelets and platelet adhesion molecules: Novel mechanisms of thrombosis and anti-thrombotic therapies. Thromb. J. 2016;14:29. doi: 10.1186/s12959-016-0100-6.
    1. Coller BS, Shattil SJ. The GPIIb/IIIa (integrin αIIbβ3) odyssey: A technology-driven saga of a receptor with twists, turns, and even a bend. Blood. 2008;112:3011–3025. doi: 10.1182/blood-2008-06-077891.
    1. Adair BD, et al. Structure-guided design of pure orthosteric inhibitors of alphaIIbbeta3 that prevent thrombosis but preserve hemostasis. Nat. Commun. 2020;11:398. doi: 10.1038/s41467-019-13928-2.
    1. McFadyen JD, Schaff M, Peter K. Current and future antiplatelet therapies: Emphasis on preserving haemostasis. Nat. Rev. Cardiol. 2018;15:181–191. doi: 10.1038/nrcardio.2017.206.
    1. Cauwenberghs N, et al. Antithrombotic effect of platelet glycoprotein Ib-blocking monoclonal antibody Fab fragments in nonhuman primates. Arterioscler. Thromb. Vasc. Biol. 2000;20:1347–1353. doi: 10.1161/01.ATV.20.5.1347.
    1. Li J, et al. Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia. Nat. Commun. 2015;6:7737. doi: 10.1038/ncomms8737.
    1. Lei X, et al. Anfibatide, a novel GPIb complex antagonist, inhibits platelet adhesion and thrombus formation in vitro and in vivo in murine models of thrombosis. Thromb. Haemost. 2014;111:279–289. doi: 10.1160/TH13-06-0490.
    1. Andrews RK, Gardiner EE, Shen Y, Berndt MC. Structure-activity relationships of snake toxins targeting platelet receptors, glycoprotein Ib-IX-V and glycoprotein VI. Curr. Med. Chem. Cardiovasc. Hematol. Agents. 2003;1:143–149. doi: 10.2174/1568016033477559.
    1. Clemetson KJ. Snaclecs (snake C-type lectins) that inhibit or activate platelets by binding to receptors. Toxicon. 2010;56:1236–1246. doi: 10.1016/j.toxicon.2010.03.011.
    1. Gao Y, et al. Crystal structure of agkisacucetin, a Gpib-binding snake C-type lectin that inhibits platelet adhesion and aggregation. Proteins. 2012;80:1707–1711. doi: 10.1002/prot.24060.
    1. Cheng X, et al. Purification, characterization, and cDNA cloning of a new fibrinogenlytic venom protein, Agkisacutacin, from Agkistrodon acutus venom. Biochem. Biophys. Res. Commun. 1999;265:530–535. doi: 10.1006/bbrc.1999.1685.
    1. Cheng X, et al. Purification and characterization of a platelet agglutinating inhibiting protein (Agkisacutacin) from Agkistrodon acutus venom. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai) 2000;32:653–656.
    1. Li WF, Chen L, Li XM, Liu J. A C-type lectin-like protein from Agkistrodon acutus venom binds to both platelet glycoprotein Ib and coagulation factor IX/factor X. Biochem. Biophys. Res. Commun. 2005;332:904–912. doi: 10.1016/j.bbrc.2005.05.033.
    1. Li J, et al. Platelet protein disulfide isomerase promotes glycoprotein ibalpha-mediated platelet-neutrophil interactions under thromboinflammatory conditions. Circulation. 2018 doi: 10.1161/CIRCULATIONAHA.118.036323.
    1. Atoda H, Hyuga M, Morita T. The primary structure of coagulation factor IX/factor X-binding protein isolated from the venom of Trimeresurus flavoviridis. Homology with asialoglycoprotein receptors, proteoglycan core protein, tetranectin, and lymphocyte Fc epsilon receptor for immunoglobulin E. J Biol Chem. 1991;266:14903–14911. doi: 10.1016/S0021-9258(18)98563-7.
    1. Mizuno H, et al. Structure of coagulation factors IX/X-binding protein, a heterodimer of C-type lectin domains. [Letter] Nat. Struct. Biol. 1997;4:438–441. doi: 10.1038/nsb0697-438.
    1. Navdaev A, et al. Aggretin, a heterodimeric C-type lectin from Calloselasma rhodostoma (Malayan pit viper), stimulates platelets by binding to alpha2beta1 integrin and glycoprotein Ib, activating Syk and phospholipase Cgamma 2, but does not involve the glycoprotein VI/Fc receptor gamma chain collagen receptor. J. Biol. Chem. 2001;276:20882–20889. doi: 10.1074/jbc.M101585200.
    1. Fujimura Y, et al. Isolation and characterization of jararaca GPIb-BP, a snake venom antagonist specific to platelet glycoprotein Ib. Thromb. Haemost. 1995;74:743–750. doi: 10.1055/s-0038-1649807.
    1. Zha XD, Liu J, Xu KS. cDNA cloning, sequence analysis, and recombinant expression of akitonin beta, a C-type lectin-like protein from Agkistrodon acutus. Acta Pharmacol. Sin. 2004;25:372–377.
    1. Lei X, MacKeigan DT, Ni H. Control of data variations in intravital microscopy thrombosis models. Thromb. Haemost. 2020;18:2823–2825. doi: 10.1111/jth.15062.
    1. Hou Y, et al. The first in vitro and in vivo assessment of anfibatide, a novel glycoprotein Ib antagonist, in mice and in a phase I human clinical trial. Blood. 2013;122:577–577. doi: 10.1182/blood.V122.21.577.577.
    1. De Meyer SF, et al. Development of monoclonal antibodies that inhibit platelet adhesion or aggregation as potential anti-thrombotic drugs. Cardiovasc. Hematol. Disord. Drug Targets. 2006;6:191–207. doi: 10.2174/187152906778249536.
    1. Chen C, et al. Platelet glycoprotein receptor Ib blockade ameliorates experimental cerebral ischemia-reperfusion injury by strengthening the blood-brain barrier function and anti-thrombo-inflammatory property. Brain Behav. Immun. 2018;69:255–263. doi: 10.1016/j.bbi.2017.11.019.
    1. Gong P, et al. Anfibatide preserves blood-brain barrier integrity by inhibiting TLR4/RhoA/ROCK pathway after cerebral ischemia/reperfusion injury in rat. J. Mol. Neurosci. 2020;70:71–83. doi: 10.1007/s12031-019-01402-z.
    1. Li TT, et al. A novel snake venom-derived GPIb antagonist, anfibatide, protects mice from acute experimental ischaemic stroke and reperfusion injury. Br. J. Pharmacol. 2015;172:3904–3916. doi: 10.1111/bph.13178.
    1. Luo SY, Li R, Le ZY, Li QL, Chen ZW. Anfibatide protects against rat cerebral ischemia/reperfusion injury via TLR4/JNK/caspase-3 pathway. Eur. J. Pharmacol. 2017;807:127–137. doi: 10.1016/j.ejphar.2017.04.002.
    1. Zheng L, et al. Therapeutic efficacy of the platelet glycoprotein Ib antagonist anfibatide in murine models of thrombotic thrombocytopenic purpura. Blood Adv. 2016;1:75–83. doi: 10.1182/bloodadvances.2016000711.
    1. Guo Y, et al. Balancing the expression and production of a heterodimeric protein: Recombinant agkisacutacin as a novel antithrombotic drug candidate. Sci. Rep. 2015;5:11730. doi: 10.1038/srep11730.
    1. Zhao YN, et al. An indirect sandwich ELISA for the determination of agkisacutacin in human serum: Application to pharmacokinetic study in Chinese healthy volunteers. J. Pharm. Biomed. Anal. 2012;70:396–400. doi: 10.1016/j.jpba.2012.06.001.
    1. Celikel R, et al. Modulation of alpha-thrombin function by distinct interactions with platelet glycoprotein Ibalpha. Science. 2003;301:218–221. doi: 10.1126/science.1084183.
    1. Dumas JJ, et al. Crystal structure of the wild-type von Willebrand factor A1-glycoprotein Ibα complex reveals conformation differences with a complex bearing von Willebrand disease mutations. J. Biol. Chem. 2004;279:23327–23334. doi: 10.1074/jbc.M401659200.
    1. Zarpellon A, et al. Binding of alpha-thrombin to surface-anchored platelet glycoprotein Ib(alpha) sulfotyrosines through a two-site mechanism involving exosite I. Proc. Natl. Acad. Sci. USA. 2011;108:8628–8633. doi: 10.1073/pnas.1017042108.
    1. Marchese P, et al. Identification of three tyrosine residues of glycoprotein Ib alpha with distinct roles in von Willebrand factor and alpha-thrombin binding. J. Biol. Chem. 1995;270:9571–9578. doi: 10.1074/jbc.270.16.9571.
    1. Ni H, et al. Increased thrombogenesis and embolus formation in mice lacking glycoprotein V. Blood. 2001;98:368–373. doi: 10.1182/blood.v98.2.368.
    1. Li C, et al. The maternal immune response to fetal platelet GPIbalpha causes frequent miscarriage in mice that can be prevented by intravenous IgG and anti-FcRn therapies. J. Clin. Investig. 2011;121:4537–4547. doi: 10.1172/JCI57850.
    1. Wong C, et al. CEACAM1 negatively regulates platelet-collagen interactions and thrombus growth in vitro and in vivo. Blood. 2009;113:1818–1828. doi: 10.1182/blood-2008-06-165043.
    1. Matsui H, et al. Distinct and concerted functions of von Willebrand factor and fibrinogen in mural thrombus growth under high shear flow. Blood. 2002;100:3604–3610. doi: 10.1182/blood-2002-02-0508.
    1. Reheman A, Tasneem S, Ni H, Hayward CP. Mice with deleted multimerin 1 and alpha-synuclein genes have impaired platelet adhesion and impaired thrombus formation that is corrected by multimerin 1. Thromb. Res. 2010;125:e177–183. doi: 10.1016/j.thromres.2010.01.009.
    1. Zhu G, et al. The integrin PSI domain has an endogenous thiol isomerase function and is a novel target for antiplatelet therapy. Blood. 2017;129:1840–1854. doi: 10.1182/blood-2016-07-729400.
    1. Nurden AT. Platelet membrane glycoproteins: A historical review. Semin. Thromb. Hemost. 2014;40:577–584. doi: 10.1055/s-0034-1383826.
    1. Kunishima S, et al. Missense mutations of the glycoprotein (GP) Ib beta gene impairing the GPIb alpha/beta disulfide linkage in a family with giant platelet disorder. Blood. 1997;89:2404–2412. doi: 10.1182/blood.V89.7.2404.
    1. Spiel AO, Gilbert JC, von Jilma B. Willebrand factor in cardiovascular disease: Focus on acute coronary syndromes. Circulation. 2008;117:1449–1459. doi: 10.1161/CIRCULATIONAHA.107.722827.
    1. Kasirer-Friede A, et al. Signaling through GP Ib-IX-V activates alpha IIb beta 3 independently of other receptors. Blood. 2004;103:3403–3411. doi: 10.1182/blood-2003-10-3664.
    1. Gilbert JC, et al. First-in-human evaluation of anti von Willebrand factor therapeutic aptamer ARC1779 in healthy volunteers. Circulation. 2007;116:2678–2686. doi: 10.1161/CIRCULATIONAHA.107.724864.
    1. Bartunek J, et al. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards von Willebrand factor. J. Cardiovasc. Transl. Res. 2013;6:355–363. doi: 10.1007/s12265-012-9435-y.
    1. Peyvandi F, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N. Engl. J. Med. 2016;374:511–522. doi: 10.1056/NEJMoa1505533.
    1. Jurk K, et al. Thrombospondin-1 mediates platelet adhesion at high shear via glycoprotein Ib (GPIb): An alternative/backup mechanism to von Willebrand factor. FASEB J. 2003;17:1490–1492. doi: 10.1096/fj.02-0830fje.
    1. Dunne E, et al. Cadherin 6 has a functional role in platelet aggregation and thrombus formation. Arterioscler. Thromb. Vasc. Biol. 2012;32:1724–1731. doi: 10.1161/ATVBAHA.112.250464.
    1. Hou Y, et al. Platelets in hemostasis and thrombosis: Novel mechanisms of fibrinogen-independent platelet aggregation and fibronectin-mediated protein wave of hemostasis. J. Biomed. Res. 2015 doi: 10.7555/JBR.29.20150121.
    1. Jasuja R, et al. Protein disulfide isomerase inhibitors constitute a new class of antithrombotic agents. J. Clin. Investig. 2012;122:2104–2113. doi: 10.1172/JCI61228.
    1. Mason PJ, Freedman JE, Jacobs AK. Aspirin resistance: Current concepts. Rev. Cardiovasc. Med. 2004;5:156–163. doi: 10.1016/j.carrev.2005.01.001.
    1. Elcioglu OC, et al. Severe thrombocytopenia and alveolar hemorrhage represent two types of bleeding tendency during tirofiban treatment: Case report and literature review. Int. J. Hematol. 2012;96:370–375. doi: 10.1007/s12185-012-1133-7.
    1. Mahaney KB, et al. Risk of hemorrhagic complication associated with ventriculoperitoneal shunt placement in aneurysmal subarachnoid hemorrhage patients on dual antiplatelet therapy. J. Neurosurg. 2013;119:937–942. doi: 10.3171/2013.5.JNS122494.
    1. Hermanides RS, et al. Net clinical benefit of prehospital glycoprotein IIb/IIIa inhibitors in patients with ST-elevation myocardial infarction and high risk of bleeding: Effect of tirofiban in patients at high risk of bleeding using CRUSADE bleeding score. J. Invasive Cardiol. 2012;24:84–89.
    1. Reed GW, et al. Point-of-care platelet function testing predicts bleeding in patients exposed to clopidogrel undergoing coronary artery bypass grafting: Verify pre-op TIMI 45–a pilot study. Clin. Cardiol. 2015;38:92–98. doi: 10.1002/clc.22357.
    1. Lincoff AM, et al. Influence of timing of clopidogrel treatment on the efficacy and safety of bivalirudin in patients with non-ST-segment elevation acute coronary syndromes undergoing percutaneous coronary intervention: An analysis of the ACUITY (Acute Catheterization and Urgent Intervention Triage strategY) trial. JACC Cardiovasc. Interv. 2008;1:639–648. doi: 10.1016/j.jcin.2008.10.004.
    1. Ndrepepa G, et al. One-year clinical outcomes with abciximab vs. placebo in patients with non-ST-segment elevation acute coronary syndromes undergoing percutaneous coronary intervention after pre-treatment with clopidogrel: Results of the ISAR-REACT 2 randomized trial. Eur. Heart J. 2008;29:455–461. doi: 10.1093/eurheartj/ehm562.
    1. Wiviott SD, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N. Engl. J. Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482.
    1. Vanhoorelbeke K, Ulrichts H, Schoolmeester A, Deckmyn H. Inhibition of platelet adhesion to collagen as a new target for antithrombotic drugs. Curr. Drug Targets Cardiovasc. Haematol. Disord. 2003;3:125–140. doi: 10.2174/1568006033481500.
    1. Chen J, et al. N-acetylcysteine reduces the size and activity of von Willebrand factor in human plasma and mice. J. Clin. Investig. 2011;121:593–603. doi: 10.1172/JCI41062.
    1. Anfibatide Phase 1 Clinical Trial in Healthy Volunteers; submitted 2012 Apr 25, posted 2012 Apr 30, (2012).
    1. Dominguez C, Boelens R, Bonvin AM. HADDOCK: A protein-protein docking approach based on biochemical or biophysical information. J. Am. Chem. Soc. 2003;125:1731–1737. doi: 10.1021/ja026939x.
    1. Kanaji S, et al. Humanized GPIbα–von Willebrand factor interaction in the mouse. Blood Adv. 2018;2:2522–2532. doi: 10.1182/bloodadvances.2018023507.
    1. Mazzucato M, et al. Characterization of the initial alpha-thrombin interaction with glycoprotein Ib alpha in relation to platelet activation. J. Biol. Chem. 1998;273:1880–1887. doi: 10.1074/jbc.273.4.1880.
    1. Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 1973;22:3099–3108. doi: 10.1016/0006-2952(73)90196-2.
    1. Reheman A, et al. Vitronectin stabilizes thrombi and vessel occlusion but plays a dual role in platelet aggregation. J. Thromb. Haemost. 2005;3:875–883. doi: 10.1111/j.1538-7836.2005.01217.x.
    1. Yang Y, et al. Plant food delphinidin-3-glucoside significantly inhibits platelet activation and thrombosis: Novel protective roles against cardiovascular diseases. PLoS ONE. 2012;7:e37323. doi: 10.1371/journal.pone.0037323.
    1. Wang Y, et al. Plasma fibronectin supports hemostasis and regulates thrombosis. J. Clin. Investig. 2014;124:4281–4293. doi: 10.1172/JCI74630.

Source: PubMed

3
S'abonner