<<

Position Paper 569

General mechanisms of and targets of (Section I) Position Paper of the ESC Working Group on – Task Force on Anticoagulants in Heart Disease

Raffaele De Caterina1*; Steen Husted2*; Lars Wallentin3*; Felicita Andreotti4**; Harald Arnesen5**; Fedor Bachmann6**; Colin Baigent7**; Kurt Huber8**; Jørgen Jespersen9**; Steen Dalby Kristensen10**; Gregory Y. H. Lip11**; João Morais12**; Lars Hvilsted Rasmussen13**; Agneta Siegbahn14**; Freek W. A. Verheugt15**; Jeffrey I. Weitz16** 1Cardiovascular Division, Ospedale SS. Annunziata, G. d’Annunzio University, Chieti, Italy; 2Medical-Cardiological Department, Aarhus Sygehus, Aarhus, Denmark; 3Cardiology, Uppsala Clinical Research Centre and Department of Medical Sciences, Uppsala University, Uppsala, Sweden; 4Institute of Cardiology, Catholic University, Rome, Italy; 5Medical Department, Oslo University Hospital, Ulleval, Norway; 6Department of Medicine, University of Lausanne, Lausanne, Switzerland; 7Cardiovascular Science, Oxford University, Oxford, UK; 83rd Department of Medicine, Wilhelminenspital, Vienna, Austria; 9Unit for Thrombosis Research, University of Southern Denmark, Esbjerg, Denmark; 10Department of Cardiology, Aarhus University Hospital, Skejby, Aarhus, Denmark; 11Haemostasis Thrombosis & Vascular Biology Unit, Centre for Cardiovascular Sciences, City Hospital, Birmingham, UK; 12Cardiology, Leiria Hospital, Leiria, Portugal; 13Department of Cardiology, Thrombosis Center Aalborg, Aarhus University Hospital, Aalborg, Denmark; 14Coagulation and Science, Department of Medical Sciences, Uppsala University, Uppsala, Sweden; 15Cardiology, Medical Centre, Radboud University Nijmegen, Nijmegen, Netherlands; 16Thrombosis & Atherosclerosis Research Institute, Hamilton General Hospital, Hamilton, Ontario, Canada

Summary inhibition of coagulation, clot stabilisation and anti-, in pro- Contrary to previous models based on plasma, coagulation processes cesses occurring in the proximity of vessel injury, tightly regulated by a are currently believed to be mostly cell surface-based, including three series of inhibitory mechanisms. “Classical” anticoagulants, including overlapping phases: initiation, when -expressing cells and and vitamin K antagonists, typically target multiple coagu- microparticles are exposed to plasma; amplification, whereby small lation steps. A number of new anticoagulants, already developed or amounts of induce activation and aggregation, and under development, target specific steps in the process, inhibiting a promote activation of factors (F)V, FVIII and FXI on platelet surfaces; single coagulation factor or mimicking natural coagulation inhibitors. and propagation, in which the Xase (tenase) and prothrombinase complexes are formed, producing a burst of thrombin and the cleav- Keywords age of to . Thrombin exerts a number of additional Anticoagulants, coagulation, tissue factor, heart disease, coronary biological actions, including platelet activation, amplification and self- heart disease, heart failure, atrial fibrillation

Correspondence to: Received: October 24, 2012 Raffaele De Caterina, MD, PhD Accepted after minor revision: December 25, 2012 Institute of Cardiology Prepublished online: February 28, 2013 “G. d’Annunzio” University – Chieti doi:10.1160/TH12-10-0772 Ospedale SS. Annunziata Thromb Haemost 2013; 109: 569–579 Via dei Vestini, 66013 Chieti, Italy E-mail: [email protected]

* Coordinating Committee Member, **Task Force Member

Introduction in heart disease (2), and an updated summary on new anticoagu- lants (3). Drugs that interfere with blood coagulation (anticoagulants) are a Section I, presented here, provides mainstay of cardiovascular therapy. Despite their widespread use, • (a) a general overview of coagulation in relation to the patho- there are still many unmet needs in this area, prompting the devel- genesis of thrombosis in heart disease; opment of an unprecedented number of new agents. A Task Force • (b) an overview of current targets of anticoagulants; of coagulation experts and clinical cardiologists appointed by the • (c) epidemiological data on the use of anticoagulants in heart European Society of Cardiology (ESC) Working Group on Throm- disease. bosis will review the entire topic of anticoagulants in heart disease. The project is intended to follow and complement the recent Task Future Sections will deal with parenteral anticoagulants (Section Force document on the use of antiplatelet agents in cardiovascular II), vitamin K antagonists (Section III), new anticoagulants in disease (1), a previous comprehensive document on anticoagulants acute coronary syndromes (Section IV), and special situations (Sec- tion V).

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. 570 ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease

Blood coagulation in relation to heart thrombosis in pigs (10), rabbits (11) and humans (12, 13). Alto- disease gether, these data suggest that inhibition of the initiation of coagu- lation at the level of TF/FVlla may provide a novel approach for Haemostasis prevention of thrombotic events, although the bleeding risk con- Under physiological conditions and with intact blood vessels, the nected with this approach is still largely unknown. haemostatic system maintains circulating blood in a fluid phase. Beyond the role in haemostasis, the binary TF/FVIIa-complex Haemostasis, i.e. the arrest of haemorrhage preventing blood loss and the ternary TF/FVIIa/FXa complex elicit intracellular signal- upon blood vessel damage – rapidly sealing the site of disruption ling events that result in the induction of genes involved in diverse in most cases – occurs through the concerted action of , biological functions that include embryonic development, cell mi- the coagulation system, and fibrinolysis, with the additional con- gration, inflammation, apoptosis and angiogenesis (14-17). tribution of a vasomotor response. Haemostasis occurs through the rapid formation of an impermeable platelet and fibrin plug Circulating TF and tissue factor pathway inhibitor (haemostatic ) at the site of injury. To prevent propa- gation of this platelet-fibrin thrombus into the vascular lumen, the In healthy individuals, TF is present in the bloodstream at very low activation of platelets and coagulation is localized to the site of in- concentrations, mainly localised to monocytes and to TF-bearing jury. In addition, fibrin within the thrombus triggers its own dis- microparticles (MPs) derived from monocytes and platelets (18). solution by -mediated fibrinolysis, which further limits MPs are cell membrane-derived fragments with a diameter of thrombus propagation. Maintenance of blood fluidity within the 0.1-1.0 µm that are released upon cell activation or during apopto- circulation and the ability to prevent blood loss after vessel injury sis (19). These MPs consist of proteins and lipids, and may contain reflects therefore a delicate balance among tightly regulated pla- DNA, mRNA and microRNA. Because they are cell membrane- telet function, coagulation and fibrinolysis (haemostatic balance) derived, MPs express antigens on their surface similar to those of (4). Disturbances in the regulation of the balance may cause the the parent cells from which they originate (20). By exposing phos- formation and deposition of too little fibrin at the site of injury, re- phatidylserine and expressing TF on their surface, MPs can initiate sulting in impaired haemostasis – ultimately manifesting as bleed- and propagate coagulation (21). Increased numbers of TF-bearing ing – or enhanced fibrin formation and deposition – causing MPs have been reported in patients with established cardiovascu- thrombosis (4, 5). lar disease and in those with cardiovascular risk factors, such as diabetes, dyslipidaemia, hypertension (20), as well as in patients The initiation of coagulation: local exposure of tissue with atrial fibrillation (22). Although it is unlikely that neutrophils factor are capable of de novo TF synthesis, TF-positive MPs may transfer TF to neutrophils (23). Coagulation is initiated when tissue factor (TF), normally segre- Alternatively-spliced TF is another form of circulating TF. This gated from the flowing blood, is exposed to plasma, binding co- TF derivative, which is formed upon splicing exon 4 directly to agulation factor (F) VII/VIIa and forming a complex on cellular exon 6, lacks the transmembrane domain (24). Alternatively- surfaces that triggers the coagulation cascade. TF (CD142), a spliced TF is produced by monocyte/macrophages, and has been transmembrane glycoprotein, is a member of the class II cytokine postulated to play a role in atherothrombotic disease (18). How- receptor superfamily, and functions both as receptor and essential ever, without the membrane binding properties of TF, it has been cofactor for FVII and FVIIa. In the vessel wall, TF is constitutively shown to lack procoagulant activity (25), and is therefore unlikely expressed by vascular smooth muscle cells, adventitial fibroblasts to play a part in coagulation. and pericytes, the cells that surround blood vessels and large or- Tissue factor pathway inhibitor (TFPI), a Kunitz type inhibitor, gans. This creates a haemostatic barrier that triggers coagulation is an important regulator of TF/FVIIa-induced coagulation. TFPI when the integrity of the vessel wall is compromised. The ex- functions by neutralising the catalytic activity of FXa and, in the pression of TF can also be induced in monocytes and, to some ex- presence of FXa, by feedback inhibition of the TF/FVIIa complex tent, in endothelial cells in response to various stimuli, including (26). TFPI contains three Kunitz-type domains; the first binds to inflammatory cytokines, endotoxin, growth factors and oxidised/ FVIIa and the second to FXa. The third C-terminal domain is in- modified low-density lipoproteins (LDL). Such expression may volved in binding of TFPI to lipoproteins and to cell surfaces (27). lead or contribute to thrombosis under certain pathological condi- Although the primary site of TFPI synthesis is the vascular en- tions, such as sepsis and disseminated intravascular coagulation dothelium (28), other cell types reported to synthesise TFPI in- (6, 7). Total lethality in homozygous TF knock-out mice embryos clude megakaryocytes/platelets and monocytes. In vivo, only 20% provides convincing evidence that TF is indispensable for life. Dif- of TFPI is present in plasma, where it circulates in complex with ferent animal models have enabled the exploration of the role of low-density lipoproteins (LDL). A major pool of TFPI is associated TF in thrombosis. Mice expressing low TF have reduced thrombo- with the endothelial surface and is rapidly released into the circu- sis in a carotid artery injury model, where the vessel wall, mostly in lation after administration of heparin, or by thrombin or shear the adventitial layer, provides the major source of TF (8). Mice forces (29). serves as a cofactor for TFPI and enhances lacking TF in smooth muscle cells also show reduced carotid arter- the rate of TFPI-mediated inhibition of FXa by 10-fold (30). Be- ial thrombosis (9) and inhibitors of the TF/FVlla complex reduce cause of its high affinity for negatively-charged phospholipids,

Thrombosis and Haemostasis 109.4/2013 © Schattauer 2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease 571 protein S may increase the affinity of TFPI for the surface of acti- ates with FVa to form the prothrombinase complex (▶ Figure 1). vated platelets, thereby increasing the local concentration of TFPI FVa is derived from several sources: it is released from activated (31). Because of its potential to downregulate coagulation, recom- platelets adhering at injury sites, or it can come from plasma, binant TFPI (tifacogin) was tested in patients with severe sepsis in where FV can be activated by thrombin or, less efficiently, by FXa. the OPTIMIST trial. Unfortunately, treatment with tifacogin had The prothrombinase complex cleaves prothrombin to generate no effect on all-cause mortality and was associated with an in- small amounts of thrombin, the responsible for fibrin creased risk of bleeding (32). Nonetheless, tifacogin reduced mor- formation. The relative concentrations of TF/FVIIa complex and tality in patients with a normal international normalised ratio TFPI determine the duration of this initiation phase. When FXa is (INR) at baseline (32), raising the possibility that it may have po- generated, it is bound by TFPI, and a quaternary complex with TF tential in some patients. and FVIIa is then formed, which inhibits VIIa. In contrast to FXa, FIXa is not inhibited by TFPI, and is only slowly inhibited by anti- A cell-based model of coagulation thrombin. FIXa moves from TF-bearing cells to the surface of acti- vated platelets that localise at the injury site. Coagulation has been classically depicted in terms of an extrinsic In the amplification phase, low concentrations of thrombin ac- pathway (initiated by TF/FVIIa), an intrinsic pathway (explaining tivate platelets adhering to the injury site, thereby inducing the re- coagulation occurring when plasma is in contact with negatively lease of FV and FVa from their α-granules. A positive feed-back charged surfaces – contact phase activation), and a common path- loop is initiated, whereby thrombin activates circulating FV and way, proceeding after the activation of FX (33). In a more modern releases FVIII from , and activates it. FVa conception, however, the coagulation process in whole blood in and FVIIIa bind to platelet surfaces and serve as cofactors for the contact with injured blood vessels consists of highly regulated large-scale thrombin generation that occurs during the propa- reactions that take place on cell surfaces (34, 35). Coagulation thus gation phase. Thrombin also activates FXI bound to platelets occurs in three overlapping phases: initiation, amplification and (▶ Figure 1). propagation (36-38). The process starts on TF-exposing cells, and In the propagation phase, the FVIIIa/FIXa complex (termed continues on the surfaces of activated platelets. “intrinsic tenase”) and the FVa/FXa complex (prothrombinase) as- The initiation phase is localised to TF-bearing cells that are ex- semble on the surface of activated platelets and accelerate the gen- posed after endothelial injury or are tethered to endothelial cells eration of FXa and thrombin, respectively. In addition, FXIa via adhesion molecules that are expressed when endothelial cells bound to the platelet surface activates FIX to form additional in- are activated. The proteolytic TF/FVIIa complex activates small trinsic tenase. FXa rapidly associates with FVa on the platelet sur- amounts of FIX and FX. On TF-expressing cells, FXa then associ- face, resulting in a burst of thrombin, which converts fibrinogen to

Figure 1: A scheme of current concepts on the coagulation process. vation and aggregation. On the surface of platelets, thrombin activates FV, The cell surface-based coagulation process includes three overlapping FVIII and FXI. In the propagation phase, FVIIIa forms a complex with FIXa phases. In the initiation phase, upon vascular injury, tissue factor (TF)-ex- (Xase), and FVa forms a complex with FXa (prothrombinase) on the platelet pressing cells and microparticles are exposed to the coagulation factors in surface, which accelerate the generation of FXa and thrombin, respectively. the lumen of the vessel, and thereby initiate thrombosis. Platelets, activated When FXa associates with FVa, it is protected from tissue factor pathway in- by vascular injury such as plaque rupture, are recruited and adhere to the site hibitor (TFPI) and (AT). In the propagation phase, a burst of of injury. The TF/FVIIa complex activates coagulation factors IX to IXa and X thrombin is generated, which is sufficient for the clotting of soluble fibri- to Xa, and trace amounts of thrombin are generated. In the amplification nogen into a fibrin meshwork. A thrombus is thus formed. phase, this small amount of thrombin is a signal for further platelet acti-

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. 572 ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease

fibrin. Soluble fibrin monomers polymerise to form fibrin protofi- stasis, pharmacological inhibition of these coagulation factors may brils, which are stabilised by FXIIIa (which is also activated by offer the exciting possibility of anticoagulation therapies with thrombin), to form a solid fibrin network that in turn stabilises minimal or no bleeding risk (40). Such concepts, however, have platelet aggregates to form a platelet/fibrin thrombus (▶ Figure 1). not yet been translated into human trials. Because coagulation comprises a series of enzymatic processes, thrombin generation is the result of an amplifying cascade, with Natural mechanisms approximately one molecule of FXa generating approximately 1,000 molecules of thrombin (39), thus making upstream in- Thrombin generation and fibrin formation occur rapidly at sites of hibition of coagulation, e.g. at the level of FXa, an attractive phar- vascular injury. To control and localise these processes, a number macological target. of inhibitory mechanisms are in place. Regulation of coagulation is Thrombin serves a number of functions in addition to fibrin exerted at multiple levels, either by enzyme inhibition or by modu- formation (▶ Figure 2), thus expanding the role of coagulation in- lation of the activity of the cofactors. Antithrombin, and hibitors, beyond such interference, to platelet activation and in- protein S are the most important regulators of coagulation. To- flammation (see below). gether with TFPI and the fibrinolytic system, they constitute the main natural anticoagulant and mechanisms in the Role of the contact phase organism. Thus, patients with a familial deficiency in one or the other of these components tend to develop thromboembolic com- Hereditary deficiency of FXII (Hageman factor) or FXI, plasma plications (). Knowledge of natural coagulation in- that initiate the intrinsic pathway of coagulation, has hibitors is guiding the development of several new anticoagulants. long been known to have a minimal impact on haemostasis. How- Most of the generated during activation of coagulation ever it has been recently appreciated that such deficiency impairs are inhibited by the serine- inhibitor antithrombin (AT), thrombus formation and provides protection from vascular oc- previously called AT III. AT preferentially inhibits free enzymes, clusive events (40). As the FXII-FXI pathway contributes to whereas enzymes that are part of the intrinsic tenase or prothrom- thrombus formation to a greater extent than to normal haemo- binase complexes are less accessible for inhibition. AT probably physiologically limits the coagulation process to sites of vascular injury and protects the circulation from liberated enzymes (33, 37). AT is, in itself, an inefficient inhibitor, but heparin and the he- parin-like molecules that are present on the surface of endothelial cells stimulate its activity (see below). Thrombomodulin (TM), a transmembrane molecule expressed on endothelial cells, binds thrombin, and the thrombin/TM com- plex activates protein C, a vitamin K-dependent proenzyme, to an active . The activated protein C (APC) anticoagu- lant system regulates coagulation by modulating the activity of the two cofactors, FVIIIa and FVa (33).The activation rate of throm- bin-mediated protein C activation is slow, but is increased at least 100-fold when thrombin binds to TM. The rate increases another 20-fold when protein C binds to endothelial protein C receptor (EPCR), which presents protein C to the thrombin/TM complex for efficient activation, highlighting a mechanism for endothelial cell localisation of anticoagulation. Thus, thrombin (▶ Figure 2) has the capacity to express both procoagulant and anticoagulant functions depending on the context under which it is generated. At Figure 2: Multiple actions of thrombin. As the final coagulation enzyme, sites of vascular disruption, the procoagulant effects of thrombin thrombin exerts multiple biological actions, only one of which, the best re- are fully expressed. In contrast, with an intact vascular system, cognised over time, is the cleavage of fibrinogen to generate fibrin. In addi- thrombin has an anticoagulant function since it binds to TM and tion, by engaging protease-activated receptors (PARs)-1 and -4 present in activates protein C. platelets and multiple cell types, thrombin promotes platelet activation and Another vitamin K-dependent cofactor protein, protein S, sup- aggregation; and exerts pro-inflammatory actions. Thrombin also amplifies ports the anticoagulant activity of APC. In human plasma, about clotting by activating coagulation FXI and the cofactors FV and FVIII into FVa 30% of protein S is free, the remainder being bound to the comple- and FVIIIa, respectively; and it stabilises clots by activating FXIII. Thrombin also exerts anti-fibrinolytic actions, through the activation of thrombin acti- ment regulatory protein C4b-binding protein. APC and free pro- vatable fibrinolysis inhibitor (TAFI), providing a molecular link between co- tein S form a membrane-bound complex, which can cleave FVIIIa agulation and inhibition of fibrinolysis; thrombin promotes the activation of and FVa, even when these are part of the fully assembled intrinsic protein C and protein S, two natural vitamin K-dependent anticoagulant pro- tenase and prothrombinase complexes. In vivo, APC does not teins that contain the coagulation process by inactivating FVa and FVIIIa. cleave intact FVIII because the binding of FVIII to von Willebrand

Thrombosis and Haemostasis 109.4/2013 © Schattauer 2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease 573 factor prevents it from interacting with the phospholipid mem- nus that serves as a tethered ligand by folding back and interacting branes. In contrast, APC is able to cleave FV, which binds phos- with the body of the receptor. In platelets, this induces platelet acti- pholipids to a similar extent as FVa. The consequence of APC-me- vation, the expression of P-selectin and CD40 ligand (CD40L), and diated cleavage of FV is the generation of anticoagulant FV that the release of inflammatory cytokines and growth factors (49). functions in synergy with protein S as an APC cofactor in the Among its numerous biological functions, thrombin is chaemotac- degradation of FVIIIa. Thus, FV can function as a procoagulant tic for leukocytes and promotes the expression of adhesion mol- and an anticoagulant cofactor. Procoagulant FVa is formed after li- ecules on the surface of these cells (▶ Figure 2). Cross-talk be- mited proteolysis by thrombin or FXa, whereas anticoagulant FV tween cells in platelet-leukocyte complexes occurs via P-selectin activity is expressed by FV that has been cleaved by APC. The anti- and CD40L, and leads to TF expression and further cytokine re- coagulant potential of FV may be particularly important in the lease. PAR-1 may also bind the ternary complex TF/FVIIa/FXa. In regulation of the intrinsic tenase complex by APC and protein S. addition, APC bound to the endothelial protein C receptor The physiological importance of the protein C system is shown by (EPCR) on endothelial cells promotes anti-inflammatory and cy- the severe thromboembolic disease that is associated with toprotective signalling through the activation of endothelial PAR-1 homozygous deficiency of protein C or S in both humans and mice (50). (41). In both cases, the severe lethal thrombotic disease manifests PAR-2 does not bind thrombin, but the TF/FVIIa complex and shortly after birth. Mice lacking a functional TM or EPCR gene FXa can activate this receptor (51). Activation of PARs by the vari- have even more severe disease and die during embryogenesis, even ous coagulation proteases results in the upregulation of genes in- before development of a functional cardiovascular system (42). volved in inflammation, including interleukin (IL)-8 and tumour Recombinant forms of soluble TM (recomodulin and solulin) and factor (TNF)-α. The TF/FVIIa complex also can initiate of APC (drotrecogin) have been developed as anticoagulants (see various intracellular signalling events, such as the activation of mi- below). togen-activated protein kinase (MAPK) pathways and phosphati- In addition to activation of protein C, the thrombin/TM-com- dyl inositol-3 kinase (PI3K)/AKT. TF/FVIIa-induced signalling plex also activates the thrombin activatable fibrinolysis inhibitor events can modulate cell fate and behaviour, rendering cells and (TAFI), a latent carboxypeptidase (43). Once activated, TAFI slows tissues proliferative, pro-migratory, and resistant to apoptosis. the rate of fibrin degradation by removing the C-terminal lysine Based on these findings, PAR inhibitors are under development residues from fibrin. Because these lysine residues serve as binding and PAR-1-targeting drugs have undergone phase III clinical trial sites for tissue- (t-PA) and plasminogen, evaluation (52, 53). their removal renders fibrin more resistant to lysis. In addition to the role of PARs in inflammation, additional Decreased overall fibrinolytic potential, occurring in patients cross-talk occurs at the level of FXa. This concept is highlighted by with congenital plasminogen, alpha2-antiplasmin or t-PA defi- the recent demonstration that lufaxin, a FXa inhibitor from the ciency, or with high plasma levels of TAFI have been associated salivary glands of blood-sucking arthropods, not only inhibits with the risk of venous thrombosis, whereas little evidence exists thrombosis in mice, but also attenuates oedema formation trig- for their role in arterial thrombosis. Increased levels of plasmi- gered by FXa injection into their paws (54). nogen activator inhibitor (PAI)-1 have been conversely associated with arterial thrombosis (44). Several fibrinolytic proteins have activities that extend beyond Variable mechanisms of thrombosis in heart fibrinolysis, such as inflammation, vascular remodelling, and disease atherosclerosis (44). Although thrombosis occurs because of excess activation of pla- Cross-talk between coagulation and inflammation telets and coagulation, distinct mechanisms underpin thrombosis in different heart diseases (55), offering opportunities for targeted Coagulation and inflammation are integrated processes (45, 46). antithrombotic strategies. This cross-talk is highlighted by thrombus formation superim- Arterial thrombosis, the leading cause of , posed on ruptured atherosclerotic plaques, which contain an occurs in the vast majority of cases as a complication of athero- abundance of inflammatory cells, as well as by the increased preva- sclerosis (atherothrombosis) through at least two different mech- lence of atherothrombosis (myocardial infarction) in inflamma- anisms: erosion of the endothelium or plaque rupture (56-58). tory rheumatic diseases (47). Superficial erosion or desquamation of endothelial cells lining the Coagulation proteases modulate inflammation by activating plaque accounts for about 25% of all cases of fatal coronary throm- protease activated receptors (PARs), and by binding to other cell bosis (59), while plaque rupture accounts for most of the re- surface receptors, such as TM and EPCR (48, 49). PARs are a mainder. When plaques rupture, there is an exposure of thrombo- family of G protein-coupled receptors expressed on a variety of genic material from the core of the plaque to the flowing blood. cells, including platelets, endothelial cells and leucocytes. Platelets Exposure of the lipid core, which is rich in TF, and of the underly- express PAR-1 and -4, which serve as thrombin receptors. Throm- ing connective tissue matrix rich in collagen leads to activation of bin binds to the extracellular domain of these receptors, where it platelets and coagulation, and to the release of vasoactive sub- cleaves a specific peptide bond, thereby generating a new N-termi- stances, which induce thrombus formation and vasoconstriction.

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. 574 ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease

Unless these processes are rapidly counteracted or an adequate Contrary to atherothrombosis, where there is a prominent role collateral circulation is present in the heart, myocardial ischaemia of both platelets and coagulation, thrombosis in the left atrium/left and an (ACS) may result. An occlusive atrial appendage in the setting of atrial fibrillation (AF) (60, 61) or thrombosis is most often found in ST-elevation myocardial infarc- in akinetic or dyskinetic areas of the left ventricle in the case of tion (STEMI), which is due to the complete interruption of coron- heart failure (62, 63) appears to be mostly caused by blood stasis ary blood flow and the ensuing ischaemia in the dependant terri- and – to some extent – blood hypercoagulability (64). Conse- tory. In the case of mural thrombosis, there is more often a “wax- quently, these thrombi have a larger fibrin component than platelet ing-and-waning” course of ischaemia, with the prevailing conse- component. Blood stasis is necessary but insufficient to increase quence of non-ST elevation ACS (NSTE-ACS) (56). While STEMI the thromboembolic risk in AF, as demonstrated by the low risk in is characterised by propagation of thrombosis (red thrombus), the absence of risk factors (65). The relative importance of coagu- making it susceptible – in most cases and if given early enough – lation over platelets in AF is highlighted by the fact that to fibrinolytic treatments, in NSTE-ACS there is a minimal propa- produces a greater reduction in in such patients than either gation component and thrombi are platelet-rich and largely resis- or aspirin plus (66). Likewise, also tant to fibrinolytic drugs (56). In the proximity of the ruptured was superior to aspirin for stroke prevention (67). plaque, thrombi possess both a platelet and a fibrin component, thus prompting the use of antiplatelet agents and anticoagulants as therapeutic strategies. Targets of anticoagulants

The targets of anticoagulants in current use or in development are depicted in ▶ Figure 3 and ▶ Figure 4, placing them in the context of the familiar traditional scheme of blood coagulation. [unfractionated heparin (UFH) and low-molecular- weight heparins (LMWH)] and vitamin K antagonists (VKA) are among the oldest anticoagulants in clinical use; their main sites of action are shown in ▶ Figure 3. Heparin consists of a family of highly sulfated polysaccharide chains, ranging in molecular weight from 3,000 to 30,000 Dalton (Da) with a mean of 15,000 Da, which corresponds to about 45 saccharide units (68, 69). Only one third of the heparin chains have anticoagulant activity because they possess the unique pen- tasaccharide sequence that binds AT with high affinity (68, 69). With higher doses, however, heparin chains with or without a pen- tasaccharide sequence can activate heparin cofactor II, a second plasma cofactor (70). Heparin catalyses the inhibition of thrombin by AT by simultaneously binding to both AT (via its pentasacchar- ide sequence) and thrombin. Formation of this ternary heparin/ AT/thrombin complex, which bridges the inhibitor and the enzyme together thereby accelerating their interaction (69), can only occur with heparin chains consisting of 18 or more sacchar- ide units (about 5,400 Da). However, shorter pentasaccharide-con- taining heparin molecules can catalyse FXa inhibition by AT be- Figure 3: Targets of “classical” anticoagulants: heparin and vitamin cause this reaction does not require bridging (69). To some extent, K antagonists (VKA, e.g. warfarin). The sites of action of anticoagulants heparin also catalyses the AT-mediated inhibition of other coagu- are depicted within the classical coagulation model, with the tissue factor lation factors, including FVIIa, FIXa, FXIa, and FXIIa, and has (extrinsic) pathway, the contact phase (intrinsic) pathway (not shown), and other anticoagulant properties, the clinical relevance of which are the common pathway. Although probably not as close to reality as the cellu- uncertain (see Section II of this series for an extensive review). lar-based model depicted in Figure 2, this model allows an easier under- Like UFH, LMWH exert their anticoagulant effects by activat- standing of the sites of action of interfering drugs and the results of coagu- ing AT and accelerating the rate at which it inhibits FXa and lation tests. Unfractionated heparin (UFH) inhibits both thrombin and FXa, as thrombin. Because only pentasaccharide-containing chains well as FIXa, FXIa and FXIIa, through a potentiation of the activity of the composed of at least 18 saccharide units are of sufficient length to natural anticoagulant antithrombin (AT). Low-molecular-weight heparins (LMWH) inhibit FXa to a greater extent than thrombin. VKA exert their anti- bridge AT to thrombin, at least 50% to 75% of LMWH chains are coagulant effect by interfering with the γ-carboxylation and thereby acti- too short to catalyse thrombin inhibition. However, these shorter vation of the vitamin K-dependent coagulation factors II, VII, IX and X, but chains retain the capacity to promote FXa inhibition, which does they also interfere with the activation of the natural anticoagulants protein C not require bridging. Consequently, LMWH preparations have and protein S (here not shown, see text for details). greater capacity to promote FXa inhibition than thrombin in-

Thrombosis and Haemostasis 109.4/2013 © Schattauer 2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease 575 hibition, and have anti-FXa to anti-FIIa activity ratios that range from 2:1 to 4:1 depending on their molecular weight profiles (69). The mechanism of action and clinical applications of UFH and LMWH will be reviewed in detail in Section II of this series. The derivatives VKA exert their anticoagulant effect by interfering with the γ-carboxylation and thereby activation of the vitamin K-dependent coagulation factors II, VII, IX and X, but they also interfere with the activation of the natural anticoagulants protein C and protein S (71). The mechanism of action and clinical applications of VKA will be reviewed in detail in Section III of this series. Drugs that target the initiation phase of coagulation are under evaluation. These agents inhibit the activity of TF/FVIIa-complex and include tifacogin, recombinant nematode anticoagulant pro- tein c2 (NAPc2) and recombinant active site–inhibited FVIIa (rFVIIai or ASIS) (▶ Figure 4). Despite promising results in in vitro or animal models, none of these TF/FVIIa inhibitors has reached clinical testing in heart disease. Drugs that target the propagation phase of coagulation de- crease thrombin generation. They include direct or indirect in- hibitors of the proteases FIXa or FXa. Direct FIX inhibitors include monoclonal antibodies and aptamers, such as pegnivacogin. Rec- ombinant human forms of activated protein C (drotrecogin) and soluble thrombomodulin (recomodulin and solulin) inactivate or promote the inactivation of FVa and FVIIIa. None of the above drugs has reached phase III clinical testing, and these agents will Figure 4: Targets of new anticoagulants. The sites of action of new anti- therefore not be reviewed in the following Sections. coagulants are depicted within the classical coagulation model, with the tis- Synthetic pentasaccharides mediate indirect, antithrombin-de- sue factor (extrinsic) pathway, the contact phase activation (intrinsic) path- pendent, inhibition of FXa. Orally active direct FXa inhibitors way, and the common pathway. Although probably not as close to reality as have been licensed for short- and long-term indications. A poten- the cellular-based model depicted in Figure 2, this model is easier to allow an tial advantage of direct FXa inhibitors over indirect inhibitors is understanding of the sites of action of interfering drugs and the results of co- their capacity to inhibit not only free FXa but also FXa within the agulation tests. Direct thrombin inhibitors bind directly to thrombin and pre- vent fibrin formation as well as thrombin-mediated activation of FV, FVIII, FXI prothrombinase complex. and FXIII. They also prevent thrombin-mediated activation of platelets, of in- Direct thrombin inhibitors (parenteral and oral) bind to throm- flammation, of anti-fibrinolysis, as well as of the anticoagulant protein C / bin and block its interaction with substrates, thereby preventing protein S / thrombomodulin pathway. Parenteral direct thrombin inhibitors the formation of fibrin and activation of platelets, FV, FVIII, FXI, include , and . Oral direct thrombin inhibitors and FXIII. These drugs may also inhibit thrombin-induced intra- are pro-drugs that generate an active compound able to bind directly to the cellular signal transduction pathways, including thrombin-in- catalytic site of thrombin: examples include (withdrawn from duced platelet activation. The direct thrombin inhibitors block development), as well as AZD0837, now under evaluation, and thrombin bound to fibrin in addition to thrombin in plasma (72). etexilate. Drugs that target coagulation proteases involved in the amplifi- The utility of oral direct FXa inhibitors and thrombin inhibitors in cation phase include agents that block FIXa (such as the DNA aptamer peg- nivacogin), FVIIIa (TB-402) or jointly FVa/FVIIIa, cofactors that are critical for heart disease has been reviewed by this Task Force (3) and will also the generation of thrombin (drotrecogin, a recombinant form of human acti- be the subject of Section IV of this series. vated protein C; recomodulin and solulin, both recombinant soluble deriva- A rational classification of currently available anticoagulants, tives of human thrombomodulin). Blockers of the propagation phase include based on their route of administration and their mechanism of ac- FXa inhibitors. At variance from the parenteral indirect FXa inhibitors, such as tion is presented in ▶ Figure 5. UFH, LMWH, and pentasaccharide derivatives (, idrabiotapari- nux), which exert their effects equally on thrombin and on FXa (UFH), preva- lently on FXa (LMWH) or exclusively on FXa (fondaparinux, idrabiotapari- The use of anticoagulant therapy in heart nux), all by potentiating the natural inhibitor antithrombin (AT), direct FXa in- disease – epidemiological data hibitors have direct, non-AT-mediated effects. A number of direct FXa in- hibitors are in clinical trials. To target the initiation of coagulation, inhibitors Coronary heart disease towards the TF/FVIIa complex have been developed, such as recombinant TFPI (tifacogin), recombinant nematode anticoagulant protein c2 (NAPc2), Anticoagulant therapies are an essential component of regimens active site-inhibited recombinant (r) FVIIa (rFVIIai) and monoclonal anti- used for ACS management. Population-based surveys conducted bodies against TF. The arrows depict activation or generation. A line ending in Europe from 1999-2001 have shown that most (>90%) patients with two perpendicular short lines depicts inhibition.

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. 576 ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease

Figure 5: A rational classification of currently available anticoagulants, based on their route of administration (paren- teral vs oral) and their mode of action.

presenting with ACS receive aspirin during hospital admission, in AF patients receiving no therapy (median: 4.45/100 person- and that most (~80%) also receive either UFH or a LMWH, with years; range: 0.25-5.90) or antiplatelet-therapy (median: 4.45/100 the proportions receiving these two forms of heparin approxi- person-years; range: 2.0-10.0) compared with VKA-treated pa- mately equal (73-75). The frequency of use of heparin varies little tients who were followed in an anticoagulation clinic (median: according to the presence/absence of ST-elevation on admission, 1.72/100 person-years; range: 0.97-2.00), or were managed in the or to the final diagnosis (Q-wave myocardial infarction [MI], non- community setting (median 1.66/100 person-years; range: 0-4.90) Q-wave MI or unstable angina), but there is considerable variation (79). Interestingly, major bleeding rates in patients receiving anti- in usage among European countries. At hospital discharge, most platelet or no therapy were similar to those in VKA-treated pa- patients (>90%) receive an antithrombotic agent, generally the tients. antiplatelet agent aspirin, but in one survey about 6% were pre- In a recent systematic review of 29 studies of patients with prior scribed warfarin and about 9% were given LMWH (75). stroke/transient ischaemic attack (TIA) who should all have re- In a more recent European survey of prescribing habits in ACS, ceived OAC therapy according to published guidelines, under- the multicentre, prospective, observational Acute Coronary Syn- treatment was reported in 25 studies, with 21 of 29 studies report- dromes Registry analysed data of 11,823 consecutive hospital sur- ing OAC treatment levels below 60% (range 19%-81.3%) (80). In vivors of acute MI. Based on the initial analysis, guideline-adher- the same systematic review, high-risk subjects, essentially those ent secondary prevention drug therapy was associated with an im- with a CHADS2 score ≥2 also were suboptimally treated, with proved one-year survival. On multivariate analysis, chronic oral seven of nine studies reporting treatment levels below 70% (range anticoagulation was the strongest predictor for not receiving as- 39%-92.3%). Thus, there continues to be underuse of OAC therapy pirin (odds ratio [OR]: 19.6, 95% confidence interval [CI]: with VKA, such as warfarin, in AF patients at high risk for stroke. 15.9-24.0) at discharge (76). After adjustment for confounding fac- Generally, the use of VKA therapy for stroke prevention in AF tors, neither prior aspirin nor oral anticoagulant treatment were is increasing, whilst the proportion of untreated patients is de- independent predictors for in-hospital mortality (77). creasing and the proportion of antiplatelet therapy use remains The EUROASPIRE-II study examined treatment of patients static (81). Nonetheless, the percentage of patients receiving no who had undergone a coronary procedure, such as coronary by- therapy still ranges from 4% to 48% (median 18%), antiplatelet pass surgery or percutaneous coronary intervention (PCI), or who therapy from 10% to 56% (median 30%), and VKA therapy from had been hospitalised with an acute MI or myocardial ischaemia 9% to 86% (median 52%), suggesting that many AF patients at (78). On admission, about 50% of such patients were taking an moderate or high risk for stroke still receive suboptimal treatment antiplatelet agent, and 7% an anticoagulant (which was likely to be based on contemporary guidelines. Also, the quality of anticoagu- a VKA in most instances). At discharge, 90% were receiving an lation control is highly variable across centers and countries (81), antiplatelet regimen, and 12% were on an anticoagulant regimen with likely important consequences, as poor time in therapeutic (which could have been a VKA or subcutaneous LMWH). There range can result in outcomes that are worse than if the patient is was evidence of variation in the frequency of use of anticoagulants left untreated (82). among European countries. The Euro Heart Survey on Atrial Fibrillation examined pre- scribing patterns among European cardiology practices in Atrial fibrillation 2003-2004 (83). Most patients surveyed had risk factors for stroke, and hence OAC treatment was indicated. About 80% of those with Oral anticoagulant (OAC) therapy remains the best treatment persistent or permanent AF, and 50% of those with paroxysmal AF strategy for the prevention of cardioembolism in AF. In a system- received OAC treatment. Only 4% of patients with persistent or atic review of “real-world” data, ischaemic stroke rates were higher permanent AF did not receive any antithrombotic therapy. How-

Thrombosis and Haemostasis 109.4/2013 © Schattauer 2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease 577 ever, these rates are likely to overestimate the use of anticoagulants placebo (93). Current evidence, however, does not support their in general practice. routine use in heart failure patients who remain in sinus rhythm, Epidemiological data on the risk of bleeding connected with the as shown in a recently completed randomised controlled trial (94). use of OAC (essentially VKA) have recently become available. One recent survey also did not find any significant – positive or Clearly, the risk of OAC-related bleeding in AF is multifactorial, negative – association of warfarin with mortality and hospitali- and the highest risk period is when OAC treatment is initiated sation (95). (84). The European Heart Rhythm Association recently published a position document, endorsed by the ESC Working Group on Thrombosis, which addresses the epidemiology and scope of the Conclusions problem of bleeding in AF patients and provides an overview of established bleeding risk factors (85). Factors influencing bleeding Blood coagulation is an essential component of haemostasis and are the modality of OAC therapy, i.e. usual care vs anticoagulation thrombosis. Coagulation is mostly a cell surface-based process of- clinic vs self management; age; prior stroke; history of bleeding; fering multiple possibilities of interference. Besides classical anti- anaemia; co-morbidities (hypertension, renal insufficiency, coagulants – heparins and VKA – several new coagulation in- disease); the use of antiplatelet agents, non-steroidal anti-inflam- hibitors, both parenteral and oral, are being developed and intro- matory drugs or drugs affecting the intensity of OAC; and alcohol duced in the market. At variance from classical anticoagulants, abuse (85). Patient values and preferences in balancing the risk of most of the new anticoagulants inhibit only a single step in the co- bleeding against the risk of stroke, as well as awareness of the prog- agulation process. Multiple surveys confirm that parenteral antico- nostic implications of bleeding, are important considerations in agulants are routinely used in ACS with or without PCI. The use of driving therapeutic choices (85). anticoagulants (essentially VKA) for long-term use is mostly re- Despite considerations about bleeding, the low rates of OAC served for the prophylaxis of cardioembolism in AF and with the prescription in patients with AF at increased risk of stroke are a use of prosthetic cardiac valves. Recent surveys show an increased major concern, as recently highlighted by the ESC AF Guidelines use of such drugs in the setting of AF, but still far from the almost (86), and are an important driver for the use of novel oral antico- generalised use recommended by current treatment guidelines, a agulants in this condition (86). deficiency that may be addressed with the availability of the novel oral anticoagulants, which are more convenient to administer than Prosthetic heart valves VKA. OAC (VKA) treatment is widely prescribed and used in patients Conflicts of interest with prosthetic heart valves, but irregularly recommended and Dr. De Caterina receives consultant and speaker fees from Astra- used in patients with rheumatic mitral stenosis who are in sinus Zeneca, Bayer, Boehringer-Ingelheim, Bristol-Myers Squibb, rhythm. There is a paucity of data on the consistency of OAC use Daiichi Sankyo, and Lilly; and research grants from AstraZeneca and on how closely available recommendations are followed in dif- and Boehringer-Ingelheim. Dr. Husted receives advisory board or ferent countries [see (87) and http://americanheart.org/download- speaker fees from AstraZeneca, Bayer, Boehringer Ingelheim, Bris- able/heart/1150461625693ValvularHeartDisease2006.pdf]. tol-Myers Squibb, and Sanofi-Aventis; and research grants from Only a limited number of case series have been published, and AstraZeneca, Bayer, Pfizer, Boehringer Ingelheim, and Bristol- these provide inconclusive information on the pattern of use and Myers Squibb. Dr. Wallentin receives consultant fees from Athera, optimal antithrombotic regimen for patients with prosthetic heart Behring, Evolva, Portola, and Roche Diagnostics; and institutional valves (88). European (89) and North American guidelines (90) research grants from AstraZeneca, Boehringer Ingelheim, Bristol- differ on the recommendations for prescribing anticoagulants Myers Squibb, GlaxoSmithKline, Merck, Pfizer, and Schering- without (89) or with aspirin (90), respectively, likely reflecting dif- Plough. Dr. Andreotti receives consultant or speaker fees from As- ferent patterns of simultaneous use of VKA and aspirin in different traZeneca, Bayer, Bristol-Myers Squibb, Pfizer, Daiichi-Sankyo, parts of the world (see [91] for an in-depth discussion). and Lilly. Dr. Huber receives speaker fees from AstraZeneca, Bayer, Boehringer Ingelheim, Daiichi Sankyo, Eli Lilly, and The Heart failure Medicines Company. Dr. Kristensen receives speaker fees from As- traZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Heart failure is associated with an increased risk of venous throm- Lilly, Merck, Pfizer, and The Medicines Company. Dr. Lip receives boembolism, cardio-embolic stroke and sudden death, and indeed speaker fees from Bayer, Boehringer Ingelheim, Bristol-Myers the latter has been associated with new coronary (thrombotic) oc- Squibb, Pfizer, and Sanofi-Aventis; and consultant fees from Astel- clusions in about 30% of patients (92). In a Cochrane systematic las, AstraZeneca, Bayer, Biotronik, Boehringer Ingelheim, Bristol- review exploring whether long-term oral anticoagulation reduced Myers Squibb, Daiichi Sankyo, Merck, Sanofi-Aventis, Portola, and total deaths and/or major thromboembolic events in patients with Pfizer. Dr. Morais receives consultant fees from AstraZeneca, heart failure, the evidence from randomised, controlled trials and Bayer, Jaba Recordati, MSD, Lilly Portugal, and Merck. Dr. Sieg- observational studies found a reduction in mortality and cardio- bahn receives institutional grants from AstraZeneca and Boehr- vascular events with anticoagulants compared with control or inger Ingelheim. Dr. Verheugt receives consultant fees from Bayer,

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. 578 ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease

Daiichi Sankyo, Pfizer, Eli Lilly, Merck, and The Medicines Com- 22. Choudhury A, Chung I, Blann AD, et al. Elevated platelet microparticle levels in nonvalvular atrial fibrillation: relationship to P-selectin and antithrombotic pany; and educational and research grants from Bayer, Boehringer therapy. Chest 2007; 131: 809-815. Ingelheim, Eli Lilly, and Roche. Dr. Weitz receives consultant fees 23. Østerud B. Tissue factor/TFPI and blood cells. Thromb Res 2012; 129: 274-278. from Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Daiichi 24. Holy EW, Tanner FC. Tissue factor in cardiovascular disease: pathophysiology Sankyo, Johnson & Johnson, Janssen Pharmaceuticals, and Pfizer. and pharmacological intervention. Adv Pharmacol 2010; 59: 259-292. 25. Boing AN, Hau CM, Sturk A, et al. Human alternatively spliced tissue factor is All other authors have reported that they have no relationships rel- not secreted and does not trigger coagulation. J Thromb Haemost 2009; 7: evant to the contents of this paper to disclose. 1423-1426. 26. Crawley JTB, Lane DA. The haemostatic role of tissue factor pathway inhibitor. Arterioscler Thromb Vasc Biol 2008; 28: 233-242. References 27. Lwaleed BA, Bass PS. Tissue factor pathway inhibitor: structure, biology and in- volvement in disease. J Pathol 2006; 208: 327-339. 1. Patrono C, Andreotti F, Arnesen H, et al. Antiplatelet agents for the treatment 28. Bajaj MS, Kuppuswamy MN, Saito H, et al. Cultured normal human hepato- and prevention of atherothrombosis. Expert position paper on the use of anti- cytes do not synthesize lipoprotein-associated coagulation inhibitor: evidence platelet agents by the Task Force of the European Society of Cardiology on the that endothelium is the principal site of its synthesis. Proc Natl Acad Sci USA use of antiplatelet agents for the treatment and prevention of atherothrombosis. 1990; 87: 8869-8873. Eur Heart J 2011; 32: 2922-2932. 29. Sandset PM, Abildgaard U, Larsen ML. Heparin induces release of extrinsic co- 2. De Caterina R, Husted S, Wallentin L, et al. Anticoagulants in heart disease: cur- agulation pathway inhibitor (EPI). Thromb Res 1988; 50: 803-813. rent status and perspectives. Eur Heart J 2007; 28: 880-913. 30. Hackeng TM, Rosing J. Protein S as cofactor for TFPI. Arterioscler Thromb 3. De Caterina R, Husted S, Wallentin L, et al. New oral anticoagulants in atrial fi- Vasc Biol 2009; 29: 2015-2020. brillation and acute coronary syndromes: ESC Working Group on Thrombosis- 31. Hackeng TM, Sére KM, Tans G, et al. Protein S stimulates inhibition of the tis- Task force on Anticoagulants in Heart Disease Position Paper. J Am Coll Car- sue factor pathway by tissue factor pathway inhibitor. Proc Natl Acad Sci USA diol 2012; 59: 1413-1425. 2006; 103: 3106-3111. 4. Astrup T. The haemostatic balance. Thromb Diath Haemorrh 1958; 2: 347-357. 32. Abraham E, Reinhart K, Opal S, et al. Efficacy and safety of tifacogin (recombi- 5. Davie EW, Fujikawa K, Kisiel W. The coagulation cascade: initiation, mainten- nant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled ance, and regulation. Biochemistry 1991; 30: 10363-10370. trial. J Am Med Assoc 2003; 290: 238-247. 6. Owens AP, 3rd, Mackman N. Tissue factor and thrombosis: The clot starts here. 33. Dahlback B. Blood coagulation. Lancet 2000; 355: 1627-1632. Thromb Haemost 2010; 104: 432-439. 34. Hoffman M, Monroe DM, 3rd. A cell-based model of haemostasis. Thromb 7. Pawlinski R, Mackman N. Cellular sources of tissue factor in endotoxemia and Haemost 2001; 85: 958-965. sepsis. Thromb Res 2010; 125 (Suppl 1): S70-73. 35. Monroe DM, Hoffman M, Roberts HR. Platelets and thrombin generation. Ar- 8. Day SM, Reeve JL, Pedersen B, et al. Macrovascular thrombosis is driven by tis- terioscler Thromb Vasc Biol 2002; 22: 1381-1389. sue factor derived primarily from the blood vessel wall. Blood 2005; 105: 36. Mann KG. Thrombin formation. Chest 2003 124: 4S-10S. 192-198. 37. Monroe DM, Hoffman M. What does it take to make the perfect clot? Arte- 9. Wang L, Miller C, Swarthout RF, et al. Vascular smooth muscle-derived tissue rioscler Thromb Vasc Biol 2006; 26: 41-48. factor is critical for arterial thrombosis after ferric chloride-induced injury. 38. Roberts HR, Hoffman M, Monroe DM. A cell-based model of thrombin gener- Blood 2009; 113: 705-713. ation. Semin Thromb Haemost 2006; 32 (Suppl 1): 32-38. 10. Roque M, Reis ED, Fuster V, et al. Inhibition of tissue factor reduces thrombus 39. Mann KG, Brummel K, Butenas S. What is all that thrombin for? J Thromb Hae- formation and intimal hyperplasia after porcine coronary angioplasty. J Am most 2003; 1: 1504-1514. Coll Cardiol 2000; 36: 2303-2310. 40. Müller F, Gailani D, Renné T. Factor XI and XII as antithrombotic targets. Curr 11. Pawashe AB, Golino P, Ambrosio G, et al. A monoclonal antibody against rabbit Opin Hematol 2011; 18: 349-355. tissue factor inhibits thrombus formation in stenotic injured rabbit carotid ar- 41. Jalbert LR, Rosen ED, Moons L, et al. Inactivation of the gene for anticoagulant teries. Circ Res 1994; 74: 56-63. protein C causes lethal perinatal consumptive coagulopathy in mice. J Clin In- 12. Lee AY, Vlasuk GP. Recombinant nematode anticoagulant protein c2 and other vest 1998; 102: 1481-1488. inhibitors targeting blood coagulation factor VIIa/tissue factor. J Intern Med 42. Rosenberg RD. Thrombomodulin gene disruption and mutation in mice. 2003; 254: 313-321. Thromb Haemost 1997; 78: 705-79. 13. Giugliano RP, Wiviott SD, Stone PH, et al. Recombinant nematode anticoagu- 43. Bouma BN, Meijers JC. Thrombin-activatable fibrinolysis inhibitor (TAFI, plas- lant protein c2 in patients with non-ST-segment elevation acute coronary syn- ma procarboxypeptidase B, procarboxypeptidase R, procarboxypeptidase U). J drome: the ANTHEM-TIMI-32 trial. J Am Coll Cardiol 2007; 49: 2398-2407. Thromb Haemost 2003; 1: 1566-1574. 14. Belting M, Dorrell MI, Sandgren S, et al. Regulation of angiogenesis by tissue 44. Meltzer ME, Doggen CJ, de Groot PG, et al. The impact of the fibrinolytic sys- factor cytoplasmic domain signalling. Nat Med 2004 10: 502-509. tem on the risk of venous and arterial thrombosis. Semin Thromb Haemost 15. Rao LVM, Pendurthi UR. Tissue factor-factor VIIa signalling. Arterioscler 2009; 35: 468-477. Thromb Vasc Biol 2005; 25: 47-56. 45. Levi M, van der Poll T. Two-way interactions between inflammation and coagu- 16. Siegbahn A, Johnell M, Sorensen BB, et al. Regulation of chaemotaxis by the cy- lation. Trends Cardiovasc Med 2005; 15: 254-259. toplasmic domain of tissue factor. Thromb Haemost 2005; 93: 27-34. 46. Petäjä J. Inflammation and coagulation. An overview. Thromb Res 2011; 127 17. Siegbahn A, Johnell M, Nordin A, et al. TF/FVIIa transactivate PDGFRβ to (Suppl 2): S34-37. regulate PDGF-BB-induced chaemotaxis in different cell types: involvement of 47. Symmons DP, Gabriel SE. Epidemiology of CVD in rheumatic disease, with a Src and PLC. Arterioscler Thromb Vasc Biol 2008; 28: 135-141. focus on RA and SLE. Nat Rev Rheumatol 2011; 7: 399-408. 18. Owens AP, 3rd, Mackman N. Sources of tissue factor that contribute to throm- 48. Esmon CT. Crosstalk between inflammation and thrombosis. Maturitas 2004; bosis after rupture of an atherosclerotic plaque. Thromb Res 2012; 129 (Suppl 47: 305-314. 2): S30-33. 49. Coughlin SR. Protease-activated receptors in haemostasis, thrombosis and vas- 19. Morel O, Jesel L, Freyssinet JM, et al. Cellular mechanisms underlying the cular biology. J Thromb Haemost 2005; 3: 1800-1814. formation of circulating microparticles. Arterioscler Thromb Vasc Biol 2011; 50. Riewald M, Petrovan RJ, Donner A, et al. Activation of endothelial cell protease 31: 15-26. activated receptor 1 by the protein C pathway. Science 2002; 296: 1880-1882. 20. Rautou PE, Vion AC, Amabile N, et al. Microparticles, vascular function, and 51. Ruf W, Dorfleutner A, Riewald M. Specificity of coagulation factor signalling. J atherothrombosis. Circ Res 2011; 109: 593-606. Thromb Haemost 2003; 1: 1495-1503. 21. Lacroix R, Dignat-George F. Microparticles as a circulating source of procoagu- 52. Morrow DA, Braunwald E, Bonaca MP, et al. in the secondary pre- lant and fibrinolytic activities in the circulation. Thromb Res 2012; 129 (Suppl vention of atherothrombotic events. N Engl J Med 2012; 366: 1404-1413. 2): S27-29.

Thrombosis and Haemostasis 109.4/2013 © Schattauer 2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved. ESC Working Group on Thrombosis – Task Force on Anticoagulants in Heart Disease: Anticoagulants in heart disease 579

53. Tricoci P, Huang Z, Held C, et al. Thrombin-receptor antagonist vorapaxar in 77. Bauer T, Gitt A, Zahn R, et al. Impact of chronic antithrombotic therapy on hos- acute coronary syndromes. N Engl J Med 2012; 366: 20-33. pital course of patients with acute myocardial infarction. The Acute COronary 54. Collin N, Assumpção TC, Mizurini DM, et al. Lufaxin, a Novel Factor Xa In- Syndromes Registry (ACOS) Investigators. Clin Cardiol 2009; 32: 718-723. hibitor From the Salivary Gland of the Sand Fly Lutzomyia longipalpis Blocks 78. Wood DA, for the EUROASPIRE II Study Group. Lifestyle and risk factor man- Protease-Activated Receptor 2 Activation and Inhibits Inflammation and agement and use of drug therapies in coronary patients from 15 countries; Prin- Thrombosis In Vivo. Arterioscler Thromb Vasc Biol 2012; 32: 2185-2198. cipal results from EUROASPIRE II. Euro Heart Survey Programme. Eur Heart J 55. Jerjes-Sanchez C. Venous and arterial thrombosis: a continuous spectrum of the 2001; 22: 554-572. same disease? Eur Heart J 2005; 26: 3-4. 79. Ogilvie IM, Welner SA, Cowell W, et al. Ischaemic stroke and bleeding rates in 56. Fuster V, Moreno PR, Fayad ZA, et al. Atherothrombosis and high-risk plaque: 'real-world' atrial fibrillation patients. Thromb Haemost 2011; 106: 34-44. part I: evolving concepts. J Am Coll Cardiol 2005; 46: 937-954. 80. Ogilvie IM, Newton N, Welner SA, et al. Underuse of oral anticoagulants in at- 57. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Eng rial fibrillation: a systematic review. Am J Med 2010; 123: 638-645. J Med 2005; 352: 1685-1695. 81. Ogilvie IM, Welner SA, Cowell W, et al. Characterisation of the proportion of 58. Lippi G, Franchini M, Targher G. Arterial thrombus formation in cardiovascu- untreated and antiplatelet therapy treated patients with atrial fibrillation. Am J lar disease. Nat Rev Cardiol 2011; 8: 502-512. Cardiol 2011; 108: 151-161. 59. Arbustini E, Dal Bello B, Morbini P, et al. Plaque erosion is a major substrate for 82. Gallagher AM, Setakis E, Plumb JM, et al. Risks of stroke and mortality associ- coronary thrombosis in acute myocardial infarction. Heart 1999 82: 269-272. ated with suboptimal anticoagulation in atrial fibrillation patients. Thromb 60. Becker RC. Thrombogenesis in atrial fibrillation contributing mechanisms and Haemost 2011; 106: 968-977. natural history. J Thromb Haemost 2009; 27: 119-121. 83. Nieuwlaat R, Capucci A, Camm AJ, et al. Atrial fibrillation management: a pros- 61. Watson T, Shantsila E, Lip GY. Mechanisms of thrombogenesis in atrial fibril- pective survey in ESC member countries: The Euro Heart Survey on Atrial Fi- lation: Virchow's triad revisited. Lancet 2009; 373: 155-166. brillation. Eur Heart J 2005; 26: 2422-2434. 62. de Peuter OR, Kok WE, Torp-Pedersen C, et al. Systolic heart failure: a pro- 84. Garcia DA, Lopes RD, Hylek EM. New-onset atrial fibrillation and warfarin thrombotic state. Semin Thromb Haemost 2009; 35: 497-504. initiation: High risk periods and implications for new antithrombotic drugs. 63. Bettari L, Fiuzat M, Becker R, et al. Thromboembolism and antithrombotic Thromb Haemost 2010; 104: 1099-1105. therapy in patients with heart failure in sinus rhythm: current status and future 85. Lip GY, Andreotti F, Fauchier L, et al. Bleeding risk assessment and manage- directions. Circ Heart Fail 2011; 4: 361-368. ment in atrial fibrillation patients: a position document from the European 64. Lopez-Cuenca A, Marin F, Roldan V, et al. Genetic polymorphisms and atrial fi- Heart Rhythm Association, endorsed by the European Society of Cardiology brillation: Insights into the prothrombotic state and thromboembolic risk. Ann Working Group on Thrombosis. Europace 2011; 13: 723-746. Med 2010; 42: 562-575. 86. Camm AJ, Lip GY, De Caterina R, et al. 2012 focused update of the ESC Guide- 65. Olesen JB, Lip GY, Hansen ML, et al. Validation of risk stratification schemes for lines for the management of atrial fibrillation: An update of the 2010 ESC predicting stroke and thromboembolism in patients with atrial fibrillation: na- Guidelines for the management of atrial fibrillation * Developed with the special tionwide cohort study. Br Med J 2011; 342: d124. contribution of the European Heart Rhythm Association. Eur Heart J 2012; 66. Connolly S, Pogue J, Hart R, et al. Clopidogrel plus aspirin versus oral antico- doi:10.1093/eurheartj/ehs253. agulation for atrial fibrillation in the Atrial fibrillation Clopidogrel Trial with Ir- 87. Whitlock RP, Sun JC, Fremes SE, et al. Antithrombotic and thrombolytic ther- besartan for prevention of Vascular Events (ACTIVE W): a randomised con- apy for valvular disease: Antithrombotic Therapy and Prevention of Thrombo- trolled trial. Lancet 2006; 367: 1903-1912. sis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Prac- 67. Connolly SJ, Eikelboom J, Joyner C, et al. Apixaban in patients with atrial fibril- tice Guidelines. Chest 2012; 141: e576S-600S. lation. N Engl J Med 2011; 364: 806-817. 88. Akhtar RP, Abid AR, Zafar H, et al. Aniticoagulation in patients following pros- 68. Hirsh J, Raschke R. Heparin and low-molecular-weight heparin: the Seventh thetic heart valve replacement. Ann Thorac Cardiovasc Surg 2009; 15: 10-17. American College of Chest Physicians Conference on Antithrombotic and 89. Vahanian A, Alfieri O, Andreotti F, et al. Guidelines on the management of val- Thrombolytic Therapy. Chest 2004; 126: 188S-203S. vular heart disease (version 2012): The Joint Task Force on the Management of 69. Garcia DA, Baglin TP, Weitz JI, et al. Parenteral anticoagulants: Antithrombotic Valvular Heart Disease of the European Society of Cardiology (ESC) and the Therapy and Prevention of Thrombosis, 9th ed: American College of Chest European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2012; Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141: 33: 2451-2496. e24S-43S. 90. Bonow RO, Carabello BA, Chatterjee K, et al. 2008 Focused update incorpor- 70. Tollefsen DM, Majerus DW, Blank MK. Heparin cofactor II. Purification and ated into the ACC/AHA 2006 guidelines for the management of patients with properties of a heparin-dependent inhibitor of thrombin in human plasma. J valvular heart disease: a report of the American College of Cardiology/Ameri- Biol Chem 1982; 257: 2162-2169. can Heart Association Task Force on Practice Guidelines (Writing Committee 71. Ageno W, Gallus AS, Wittkowsky A, et al. Oral anticoagulant therapy: Anti- to Revise the 1998 Guidelines for the Management of Patients With Valvular thrombotic Therapy and Prevention of Thrombosis, 9th ed: American College Heart Disease): endorsed by the Society of Cardiovascular Anesthesiologists, of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; Society for Cardiovascular Angiography and Interventions, and Society of Tho- 141: e44S-88S. racic Surgeons. Circulation 2008; 118: e523-661. 72. Weitz JI. Factor Xa and thrombin as targets for new oral anticoagulants. 91. Butchart EG, De Caterina R. Antithrombotic management in patients with Thromb Res 2011; 127 (Suppl 2): S5-S12. prosthetic valves In: Therapeutic Advances in Thrombosis. Wiley-Blackwell, 73. Fox KAA, Cokkinos DV, Deckers J, et al. The ENACT study: a pan-European Hoboken, NJ, USA; 2013; pp. 246-271. survey of acute coronary syndromes. European Network for Acute Coronary 92. Freudenberger RS, Schumaecker MM, Homma S. What is the appropriate ap- Treatment. Eur Heart J 2000; 21: 1440-1449. proach to prevention of thromboembolism in heart failure? Thromb Haemost 74. Fox KAA, Goodman SG, Klein W, et al. Management of acute coronary syn- 2010; 103: 489-495. dromes. Variations in practice and outcome; findings from the Global Registry 93. Lip GH, Gibbs CR. Anticoagulation for heart failure in sinus rhythm. Cochrane of Acute Coronary Events (GRACE). Eur Heart J 2002; 23: 1177-1189. Database Syst Rev 2011; 3: CD003336. 75. Hasdai D, Behar S, Wallentin L, et al. A prospective survey of the characteristics, 94. Homma S, Thompson JL, Pullicino PM, et al. Warfarin and aspirin in patients treatments and outcomes of patients with acute coronary syndromes in Europe with heart failure and sinus rhythm. N Engl J Med 2012; 366: 1859-1869. and the Mediterranean basin; the Euro Heart Survey of Acute Coronary Syn- 95. Mujib M, Rahman AAZ, Desai RV, et al. Warfarin use and outcomes in patients dromes (Euro Heart Survey ACS). Eur Heart J 2002; 23: 1190-1201. with advanced chronic systolic heart failure without atrial fibrillation, prior 76. Bauer T, Gitt AK, Jünger C, et al. Guideline-recommended secondary preven- thromboembolic events, or prosthetic valves. Am J Cardiol 2011; 107: 552-557. tion drug therapy after acute myocardial infarction: predictors and outcomes of nonadherence. Eur J Cardiovasc Prev Rehab 2010; 17: 576-581.

© Schattauer 2013 Thrombosis and Haemostasis 109.4/2013 Downloaded from www.thrombosis-online.com on 2013-06-19 | IP: 2.231.31.96 Note: Uncorrected proof, prepublished online For personal or educational use only. No other uses without permission. All rights reserved.