1358 Current Pharmaceutical Design, 2009, 15, 1358-1372 Activation by Extracellular Matrix in Haemostasis and Thrombosis

Steve P. Watson*

Centre for Cardiovascular Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, Univer- sity of Birmingham, Birmingham, B15 2TT, UK

Abstract: The prevention of excessive blood loss to avoid fatal haemorrhage is a pivotal process for all organisms pos- sessing a circulatory system. Increased circulating blood volume and pressure, as required in larger animals, make this process all the more important and challenging. It is essential to have a powerful and rapid system to detect damage and generate an effective seal, and which is also exquisitely regulated to prevent unwanted, excessive or systemic activation so as to avoid blockage of vessels. Thus, a highly specialised and efficient haemostatic system has evolved that consists of cellular () and (coagulation factors) components. Importantly, this is able to support haemostasis in both the low shear environment of the venous system and the high shear environment of the arterial system. Endothelial cells, lining the entire circulation system, play a crucial role in the delicate balance between activation and inhibition of the haemostatic system. An intact and healthy endothelium supports blood flow by preventing attachment of cells and pro- teins which is required for initiation of coagulation and platelet activation. Endothelial cells produce and release the two powerful soluble inhibitors of platelet activation, nitric oxide and prostacyclin, and express high levels of CD39 which rapidly metabolises the major platelet feedback agonist, ADP. This antithrombotic environment however can rapidly change following activation or removal of endothelial cells through injury or rupture of atherosclerotic plaques. Loss of endothelial cells exposes the subendothelial extracellular matrix which creates strong signals for activation of the haemo- static system including powerful platelet adhesion and activation. Quantitative and qualitative changes in the composition of the subendothelial extracellular matrix influence these prothrombotic characteristics with life threatening thrombotic and bleeding complications, as illustrated by formation of atherosclerotic plaques or the disorder Ehler-Danlos syndrome, which is caused by a defect in collagen synthesis and is associated with fragile blood vessels. This review will focus on the role of the subendothelial matrix in haemostasis and thrombosis, highlighting its potential as a target for novel anti- thrombotics.

THE COMPOSITION OF THE EXTRACELLULAR vascular matrix are type I and III fibrillar collagens, as ex- MATRIX emplified in the human aorta where they make up 50% of its dry weight [4,5]. These two fibrillar collagens capture circu- The intima extracellular space is localized between the lating VWF and mediate powerful adhesion and activation of endothelial basement membrane and the internal elastic lam- platelets through GPVI and 21 as discussed be- ina representing the border to tunica media. The vascular low [6,7]. Other important components of the vascular ex- endothelial basement membrane (50-100 nm) provides the tracellular matrix include proteoglycans, hyaluronate, elastin primary physical support for endothelial cells and is the first and vitronectin. Elastins are highly abundant in the vascular matrix constituent to be exposed to the flowing blood fol- extracellular matrix, often making up to as much as 50% of lowing denudation of the endothelium. The basement mem- its dry weight, and are essential to preserve elasticity [8]. In brane is primarily composed of laminin (typically 4 laminin  addition the proteoglycan chondroitin sulphate contributes to subtypes), which is the most abundant protein, fibronectin, the tensile strength of the vessel wall, particularly in the enactin, several proteoglycans (including heparin sulphate aorta. proteoglycan) and collagen type IV [1-3]. Of these, laminin and fibronectin have been shown to support adhesion and The major components within the vascular extracellular activation of platelets (see below). In contrast, the role matrix that interact directly with platelets are laminin, fi- played by collagen type IV in regulating platelet activation is bronectin, vitronectin and collagen. Thus, the molecular ba- unclear (see below). sis of the interaction of these proteins with platelets is dis- cussed in more detail below. Several other structural proteins The extracellular matrix that lies between the basement also have significant roles in thrombus formation and are membrane and the internal elastic lamina is primarily layed recruited to the extracellular matrix upon damage to the en- down by smooth muscle cells and has been termed the vas- dothelium, including VWF [9], fibrinogen, thrombospondin cular extracellular matrix. The major components of the and proteins involved in the coagulation response, and these

are discussed as well. For a representative structure of an *Address correspondene to this author at the Centre for Cardiovascular arterial wall showing the location of extracellular matrix Sciences, Institute for Biomedical Research, College of Medical and Dental proteins, see Fig. (1) in the Chapter by Eble and Niland (this Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Tel: 00 volume). 44 121 4146514; E-mail: [email protected]

1381-6128/09 $55.00+.00 © 2009 Bentham Science Publishers Ltd. Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1359

Fig. (1). Steps of platelet adhesion, activation and thrombus formation. At sites of vessel wall damage where ECM is exposed, initial tethering of platelets occurs via GPIb-VWF interaction. VWF is bound to col- lagen via its A3 domain and interacts with GPIb on the surface of platelets via its A1 domain. Rolling platelets undergo stable adhesion through 21-collagen and IIb3-VWF interaction. At this stage both become activated mainly through inside-out signaling trig- gered by GPVI engagement to collagen. Platelet activation and integrin engagement cause platelet spreading as well as secretion of prothrombotic agents (ADP, TxA2, Fibrinogen). This process recruits additional platelets and stabilizes the growing thrombus. Highly acti- vated platelets within this thrombus provide a ‘procoagulant’ surface leading to massive thrombin generation, which further reinforces plate- let activation but also generates a rigid fibrin mesh and leads to clot retraction, which is based on IIb3-fibrin interaction.

THE FUNCTION OF THE SUBENDOTHELIAL MA- The subendothelial matrix is also involved in the regula- TRIX IN HAEMOSTASIS AND THROMBOSIS tion of endothelial cells [12] through integrin-mediated ‘out- side-in’ signalling in response to components of the vessel The subendothelial extracellular matrix provides struc- wall such as collagen, laminin, fibronectin, vitronectin and tural support to endothelial cells and is therefore important in fibrinogen. Thus, changes in the composition of the subendo- the organization and maintenance of the shape of the vessel. thelial matrix have the potential to modulate composition The cylindrical shape of the vessel is required for unob- and the response of endothelial cells to other stimuli, includ- structed laminar blood flow. The structural function of the ing shear [13,14]. Activation of endothelial cells induces the extracellular matrix is illustrated by the massive remodeling expression of the pro-thrombotic proteins, VWF, P- that occurs in processes such as restenosis or atherosclerotic and , and adhesion receptors, such as E-selectin, plaque development which cause a narrowing of the vessel ICAM-1 and VCAM-1. Expression of VWF and P-selectin lumen [10,11]. This may cause a disturbance to laminar flow on the endothelial surface contribute to the development of and a significant increase in shear force, and eventually lead atherosclerotic plaques through recruitment of platelets and to obstruction of blood flow giving rise to life-threatening leukocytes [15]. Furthermore, once exposed to the flowing thrombotic complications. Indeed, it is noteworthy that blood, the subendothelial matrix plays a critical role in plate- atheroma are frequently found at sites of vessel bifurcation, let adhesion and activation, and thrombus formation, as dis- where flow is disturbed. cussed in further detail below. 1360 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

THE REGULATION OF THROMBUS FORMATION binding to fibronectin and laminin, respectively, although BY THE SUBENDOTHELIAL EXTRACELLULAR this role is redundant with that of integrin IIb3 [23-25]. MATRIX Stable adhesion of matrix proteins to their respective The events underlying thrombus formation at the high integrins serves to increase the net affinity of collagen for shear rates of the arterial system that occur following vascu- GPVI leading to enhancement of signals through the immu- lar damage or rupture of atherosclerotic lesions have been noglobulin receptor. In addition, clustering of the integrins subject to intensive research over a number of years, and themselves generates outside-in signals that serve to rein- have led to the development of a multistep model. The trig- force platelet activation, albeit these are much weaker than ger is the denudation of endothelial cells causing exposure of those of GPVI [21]. Thus, platelet receptors the subendothelial extracellular matrix to the flowing blood. generate a repertoire of signals that mediate platelet adhesion This is followed by capture (or tethering) of platelets, which and spreading at sites of vessel injury, with the latter serving is a specialized process that serves to counter the very high to strengthen platelet adhesion. shear rates of the arterial system, including those found at The secretion of platelet dense granules and activation of sites of stenosis. Tethering of platelets is accomplished by phospholipase A leads to the release of ADP and formation binding of VWF to subendothelial collagen which leads to a 2 of TxA2, respectively, the two major feedback mediators of conformational change that enables it to bind to the GPIb- platelets. These two agonists synergise with each other and IX-V complex on the platelet surface (for review [16]). The with signals from platelet glycoprotein receptors to mediate interaction between VWF and GPIb-IX-V has a very fast on- further platelet activation. Crucially, the two feedback mes- rate of association that leads to the formation of an effective sengers, along with VWF and fibrinogen, mediate formation bond at high shear and therefore platelet capture [17]. Strik- of a platelet aggregate through a continual cycle of platelet ingly, the effectiveness of the VWF conformational change tethering (via VWF), platelet activation (primarily via ADP and subsequent binding to GPIb binding increases with in- and TxA2) and platelet crosslinking (via fibrinogen or VWF creasing shear. The interaction between VWF and GPIb-IX- binding to integrin IIb3). Moreover, this scheme of events V has a very fast off-rate, however, thereby leading only to is further enhanced by generation of thrombin through the the transient capture of platelets [17]. This is illustrated by coagulation cascade which activates platelets through bind- the rolling of platelets in the direction of flow on a mono- ing to the protease activated receptors, PAR1 and PAR4, as layer of VWF, with stable aggregates only being formed well as converting fibrinogen to fibrin, which serves to fur- after several minutes through the weak activation of integrins ther strengthen the growing thrombus. The growing throm- by the GPIb-IX-V complex [18]. bus is also consolidated by low affinity interactions between The tethering of platelets to VWF enables other matrix several platelets surface proteins, including binding of CD40 proteins with a slower on-rate of association to their recep- ligand (CD40L) to integrin IIb3, and binding of ephrins to tors to interact with platelet surface receptors. Crucially, Eph kinases. Furthermore, thrombus formation is completed these interactions would not occur in the absence of VWF by an integrin-driven clot retraction, a process through which [19]. The most important interaction is that of the fibrillar contraction of the actin-myosin cytoskeleton is channelled collagens, type I and III with the low affinity immunoglobu- via integrins to fibrin [26]. lin surface receptor, GPVI. Clustering of GPVI by collagen Alongside the above events, platelets provide a ‘proco- or by laminin, which has an approximate tenfold lower affin- agulant’ surface through exposure of phosphatidylserine ity for the glycoprotein receptor compared with collagen leading to a burst of thrombin generation, which further rein- [20], generates powerful signals that lead to activation of forces platelet activation and generates fibrin, leading to clot platelets, including inside-out activation of platelet integrins, retraction, as described above. A simultaneously activated cytoskeletal remodeling (leading to platelet spreading), gran- fibrinolytic response prevents excessive thrombus formation ule secretion and thromboxane A2 (TxA2) formation [6]. Ac- (for review [27]). tivation of integrins converts the tethering of platelets to sta- ble adhesion. In contrast, the affinity of the interaction be- The above model emphasizes that platelet adhesion to the tween collagen or laminin with GPVI is not sufficient to me- subendothelium represents the first step in thrombus forma- diate stable adhesion. The platelet integrins that interact with tion following damage to the arterial system. The early the extracellular matrix are IIb3, 21, 51, 61 and events in this cascade have attracted much interest for phar- v3. Of these, the most important two are considered to be macological intervention in anticipation that this may over- IIb3, which is expressed at 80,000 - 120,000 copies per come limitations of current ant-platelet regimes. In this re- platelet, and 21, which has 1500 - 3000 copies. Integrin view, we shall discuss what is known about the structural IIb3 mediates stable adhesion through binding to VWF features of the major matrix proteins that mediate platelet and fibronectin, and also mediates platelets aggregation binding and platelet activation, as well as the possibility of through interaction with above ligands and also fibrinogen pharmacological intervention and therapeutic potential. [21]. Integrin 21 mediates stable adhesion through bind- ing to collagen, although this role can be masked by the in- COMPONENTS OF THE VASCULAR EXTRACEL- teraction of VWF with integrin IIb3. Thus, the importance LULAR MATRIX AND PLATELET ACTIVA- for 21 in supporting adhesion to collagen is increased TION/ADHESION under conditions of reduced VWF plasma concentration A). Collagen [22]. Integrins 51 and 61 have also been shown to sup- port stable adhesion of platelets under high shear through There are at least 43 distinct collagen  chains in the , assembling into 28 collagen types [28]. Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1361

Their common feature is the Col-domain, a triple-helical proach based on the use of toolkits of triple-helical peptides core. Three polyproline helices form a superhelical structure, (summarized in [38]). From these very elegant studies, it has characterized by the sequence glycine-X-Y. This sequence become clear that although 21 binds with highest affinity may be repeated more than 300 times, so that a triple-helical to GFOGER (single amino code), it binds with varying af- domain is often more than 1,000 amino acids in length. The finities to more general sequences of GXXGER, thus allow-  chains form a right-handed triple helix and may be identi- ing for a controlled adhesion and cell migration over a colla- cal or different, depending on the type of collagen. The X- gen surface. For GPVI it has become clear that the spacing position is often taken by proline (Pro), while the Y position of the GPO-triplets plays a crucial role in GPVI-collagen is frequently occupied by 4-hydroxyproline (Hyp) [29]. This interaction [39]. The VWF binding sequence on collagen has regular repetition and high glycine content is found in very been identified as GPRGQOGVMGFO [40]. The intact se- few other fibrous proteins. Glycine, being the smallest amino quence is found in collagen types II and III, but not in intact acid, is required at every 3rd position where it occupies the collagen type I. However, VWF binding to type I collagen interior of the helix, since there is no space for side chains. has been shown. It has been suggested that a binding region The rings of Pro and Hyp point outward, thermally stabiliz- for VWF on collagen type I could be formed by contribu- ing the triple helix. More than 90% of the collagen in the tions of both (1 and 2) chains [38]. body is type I, II, III and IV. The first step in platelet adhesion and activation follow- Collagen Types in the ECM ing damage to the vessel wall is the interaction of plasma VWF with subendothelial collagen under shear, as discussed While collagen types I, III, IV, V, VI, VIII, XV, XVIII above [41]. Platelets are tethered by the interaction of the and XIX have been found in the vessel wall, types I, III and GPIb-IX-V complex with the A1 domain of VWF, which is V are the most prominent [30]. Collagen I and III form fi- exposed by shear [42,43]. VWF is immobilized on collagen bres, confer tensile strength as well as flexibility. Collagen V types I and III in the subendothelial matrix via its A3 domain is found as co-polymer in association with other collagens [44,45]. In addition, the A1 domain has also been reported to and may also form filaments by itself. Collagens IV and mediate binding to collagen type VI in the subendothelial VIII, forming 2D or 3D networks, are believed to lie closer matrix [46,47]. Platelets roll over the collagen-VWF surface, to the actual vessel lumen than other collagens [4,31]. Colla- forming new GPIb-VWF bonds at the front while releasing gen type IV is found directly beneath the endothelial cells in bonds at the tail. During rolling under high shear conditions, the vascular endothelial basement membrane, often in asso- platelets also shed parts of their membranes, which are in ciation with laminin around smooth muscle cells. Collagen firm contact with VWF leaving trails of procoagulant mi- type IV is a non-fibrillar collagen and is considerably less croparticles [48]. Firm attachment of platelets to collagen is potent than the fibrillar collagens in supporting activation of achieved via the 21-collagen interaction or indirectly via the major signalling receptor for collagen on platelets, gly- binding of VWF to integrin IIb3. Both integrins have to coprotein VI (GPVI), or supporting adhesion through re- be in an active conformation to mediate binding. Activation cruitment of von Willebrand factor (VWF) [32]. On the other is mediated by binding of talin and kindlin-3 to their cytoso- hand, there are reports that collagen type IV is able to sup- lic tails (inside-out signalling) following agonist-induced port GPVI-mediated platelet aggregation in a low shear envi- platelet activation [49,50]. Collagen is the most powerful ronment, or platelet adhesion and thrombus formation at agonist in the subendothelial matrix mediating activation, arteriolar rates of flow [33,34]. The explanation for these although weak signals from GPIb-IX-V also contribute to contradictory observations may be related to the method of activation. collagen preparation. Thus, the importance of basement membrane collagen type IV in supporting platelet activation The collagen-binding region of 21 is the I-domain, and thrombus formation warrants further investigation. Fur- also called A-domain due to its analogy to the vWF A1- ther into the ECM, collagen VI is found, forming beaded domain [51]. Crystallography, using mutant 2 I-domains, filaments. Following superficial vessel wall damage, these has enabled identification of the critical residues that support sub-endothelial collagens are the first ones to contact patrol- binding of a synthetic collagen triple helix peptide contain- ling platelets. On the other hand, increased damage reaches ing GFOGER [52]. The glutamate coordinates the divalent 2+ the internal elastic lamina and deeper layers of the media, cation (under physiological conditions Mg ) in the integrin exposing collagens type I and III and also tissue factor which metal ion dependent adhesion site (MIDAS). The MIDAS is activates the coagulation cascade. not exposed in the inactive conformation and only becomes accessible to collagen l upon integrin activation [53]. Helix 7 Collagen I is present in abundance in the cap of plaques of the integrin undergoes a dramatic movement during acti- [35]. Remodelling of the vessel wall, including collagen vation suggesting that this is a mechanism for affinity regu- turnover, induced by proteolytic activity of macrophages, is lation of the integrin leading to agonist binding and possibly important for the ability of smooth muscle cells to migrate also outside-in signalling, which involves clustering of the and proliferate within the intima. This contributes to the de- integrin [54,55]. Further regulation of integrin 21 is velopment of plaques. achieved via rearrangement of disulphide bonds, catalyzed Direct and Indirect Binding of Platelets to Collagen by protein disulphide isomerases on the platelet surface [56], although this remains controversial. Studies with recombi- The main collagen receptors on platelets are considered nant collagen peptides have shown that only GFOGER to be the integrin 21 (VLA-2, GPIa/IIa) and GPVI [36,37]. (found in collagen type I, II and IV) is capable of binding to Over the last years the binding regions of collagen receptors the inactive conformation of integrin 21 [57]. The binding on different types of collagen have been analyzed by an ap- to the other GXXGER sequences depends upon inside-out 1362 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson signalling. The main role of integrin 21 in platelet colla- an increased expression of the Lutheran glycoprotein, sickle gen interaction is to support firm adhesion, with GPVI play- cells have been shown to have increased affinity for binding ing a crucial role in platelet activation. to laminin relative to normal red cells, possibly contributing to vasoocclusive episodes in these patients [75,76]. Other Studies using mice deficient in the GPVI signal transduc- laminin receptors include dystroglycan, a heterodimeric tion subunit, the Fc receptor  chain, or treatment of mice polypeptide particularly important for the function and struc- with a GPVI-depleting , JAQ1, suggest ture of muscle fibres, the ‘collagen’ receptor GPVI and an that under certain experimental conditions, GPVI is the only uncharacterised platelet protein of 67 kDa receptor [20,77, collagen receptor of importance in thrombosis [58]. Indeed, 78]. treatment with JAQ1 leads to a complete loss of the throm- boembolism induced by collagen and epinephrine. On the Laminin - Platelet Interactions other hand platelets from 1-deficient mice develop less sta- ble aggregates under flow, pointing to an important role Adhesion of platelets to laminin was first described in of 21 in thrombosis [59]. Experimental and species (e.g. 1983 and was subsequently shown to be due to binding to mouse platelets are much smaller than human) differences integrin 61 (also known as VLA-6) [79-81]. The in- might be one reason for the sometimes conflicting results. volvement of GPVI in laminin- induced platelet activation However, the focus on certain aspects of artificial thrombus was only recently reported [20]. The 67 kDa laminin recep- formation might lead to overestimation of the role of the two tor, that was first identified on human breast carcinoma and receptors, as exemplified in the study of Auger et al. This muscle cells [82,83] is also expressed on platelets, although study suggested two distinct mechanisms underlying stable almost nothing is known about its function [81,84,85]. Thus, adhesion to collagen, namely via VWF bridging or through it seems that the principle laminin receptor on platelet is the direct binding, possibly via integrin 21 to the GFOGER integrin 61, while GPVI functions as a downstream recep- motif, as this does not require inside-out activation of the tor, contributing to laminin induced platelet activation. In the integrin [60]. absence of GPVI platelets are still able to adhere normally to laminin but spreading and lamellipodia formation are B). Laminin strongly reduced [20]. Biacore studies have revealed that the interaction between laminin and GPVI has a ten-fold lower Laminins are heterotrimeric consisting of affinity compared to binding to collagen. This lower affinity three polypeptide chains (, , ) [61,62]. 5 encode for may explain why 61 binding is a prerequisite for GPVI the -chains and 3 each for the - and -chain [62]. The binding and why laminin, in contrast to collagen, is unable to chains can assemble in various combinations and 15 differ- induce significant tyrosine phosphorylation or platelet ag- ent laminin subtypes have been described [2]. The expres- gregation in suspension [20]. The concept of 61 and GPVI sion of the different laminin subtypes is tissue specific. as the platelet laminin receptors is very appealing since it Basement membranes of all blood vessels are characterized mirrors the cooperation of 21 and GPVI in the interaction by the presence of 4 laminin subtypes [61]. of collagen [6]. The central part of the laminin molecule is characterized Again in analogy to platelet-collagen interaction, VWF is by a triple -helical assembly of the , , and  chain [63]. required for platelet adhesion to laminin under flow condi- While the C-terminal parts of the  and -chain are within tions [24]. Anti-VWF antibody treatment blocks platelet ad- the triple-helical assembly, the -chain extends further to hesion to laminin completely under high (1500 s-1) but not form five globular motifs, called g-domains. This domain is under low shear (300 s-1) conditions. Similar results were an important mediator of laminin - cell interaction because obtained with VWF deficient blood (type III VWF disease) of its involvement in binding to various integrins [62,64]. or blood from Bernard-Soulier syndrome patients (GPIb de- The N-terminus of the molecule is not triple helical and con- ficiency) [24]. Therefore, in whole blood under flow condi- sists of short arms of the 3 chains, forming distinctive do- tions, the interaction of VWF with laminin seems to be the mains. These domains are critical for the self-assembly of initial step. Recruitment of platelets by VWF may enable the laminin into the polymer that is found in endothelial integrin 61 and GPVI to interact with laminin and thereby basement membranes [65-67]. support integrin activation giving rise to stable adhesion and Laminin Receptors formation of small aggregates. The interaction of VWF with components of the extracellular matrix other than collagen The integrins 11, 21, 61, 64, 71, 91 and has been suggested for several years [86,87], but laminin is v3 have been shown to interact with laminins, with differ- the first non-collagen to be able to mediate direct binding of ent preferences for the various laminin subtypes. However, VWF, a result that has been confirmed by Biacore [24]. the only reported laminin-binding integrin on the platelet In conclusion, laminin seems to interact with platelets surface, 61, is known to bind all laminins subtypes both indirectly via VWF and directly through integrin 61 [68,69]. On the other hand, 21 does not appear to support and GPVI. The role of the 67kDa protein in the interaction of binding of platelets to laminin [70,71]. laminin with platelets is not known. The sequence of events Several additional cell surface molecules on platelets and that results in adhesion of platelet to a laminin surface under other cell types have also been described to interact with flow conditions is similar to that seen on collagen. The over- laminin. Among them is the Lutheran glycoprotein, a blood all significance of the interaction of platelets with laminin group glycoprotein expressed on erythrocytes [72]. The Lu- however, would appear to be minimal due to collagen expo- theran glycoprotein mediates adhesion of red cells to sure. On the other hand, it can be speculated for example that laminin, specifically to 5-chain laminins [73,74]. Based on the interaction of platelets with laminin plays a key role fol- Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1363 lowing superficial injuries that lead only to disruption of the sequence [110], although additional IIb3-binding sites in endothelial lining or during the final stages of angiogenesis fibronectin have been identified. This multisite interaction of as a mechanism for completing the endothelial monolayer IIb3 with fibronectin seems to be necessary for a tight through release of growth factors. In such cases the deeper, interaction [111], with RGD-mediated integrin binding being underlying collagen may not be exposed. essential for platelet adhesion under shear to fibronectin [112]. Platelets also express two additional fibronectin recep- C). Fibronectin tors 51 and v3 [79,113]. 51 has been shown to have non-redundant role in platelet adhesion to fibronectin, since Structure of Fibronectin antibodies to 5 are able to inhibit platelet deposition under Fibronectin is a homodimeric glycoprotein consisting of flow (300s-1) by approximately 50%. In comparison RGD- 250 kDa subunits linked by two disulphide bonds in their containing peptides or antibodies to IIb3 blocked this in- COOH-termini [88]. In contrast to laminin and collagen, teraction by over 85% [25,112]. fibronectin is present in blood plasma at a concentration of While IIb3 supports platelet adhesion as well as acti- 300-400g/ml (~10% of the fibrinogen concentration) as vation in response to fibronectin, 51 and v3 seem to well as in platelet -granules [89-91]. Fibronectin RNA is  mainly function as adhesion receptors. Platelets adhere and alternatively spliced at three sites generating EIIIA(EDA), spread on fibronectin coated surfaces under static conditions EIIIB(EDB) and V (CS-1) variants. EIIIA and EIIIB variants [80,114,115]. Platelet adhesion to fibronectin induces forma- are mostly found in the extracellular matrix of blood vessels, tion of filopodia and lamellipodia through binding to integrin but are also present in platelet granules [92,93]. In contrast IIb3, whereas binding to integrin 51and v3 induces plasma fibronectin mostly lacks these domains, although the 2+ only filopodia formation [23,116]. Robust Ca mobilization level of EIIIA fibronectin in plasma seems to be increased in is dependent on the interaction of IIb3 with fibronectin disease states associated with an increased thrombotic risk [23], causing outside-in signaling as observed for the binding such as atherosclerosis, diabetes and stroke [94-96]. Plasma of fibrinogen to integrin IIb3 [117]. Interestingly, adhe- fibronectin is a soluble dimer, while extracellular matrix sion of platelets to fibronectin is not inhibited in the absence fibronectin is assembled into insoluble aperiodic matrix fi- of functional IIb3, supporting a role for 51 and v3 in brils. The latter is a cell-mediated process, under involve- this process [23,113]. However, neither 51 nor v3 are ment of integrins and cell surface LAMMs (molecules of able to induce a similar level of spreading to fibronectin in large apparent molecular mass). Cells dedicated to fi- comparison to IIb3, which may be due to an almost two bronectin assembly are fibroblasts, vascular smooth muscle orders of magnitude higher expression level of the latter. cells and endothelial cells but also platelets [97]. Further- more, several binding sites for adhesive proteins such as fi- Although all these studies support a function of purified brin, collagen, VWF and integrins have been identified in the fibronectin in platelet adhesion and spreading, an important fibronectin molecule, underlining its capability to crosslink question is the relative role of fibronectin within the suben- molecules and cells [97]. dothelial extracellular matrix, particularly taking into ac- count the presence of other platelet-regulatory proteins. This Fibronectin Receptors has been addressed using human umbilical artery subendo- Cell surface receptors for fibronectin belong to the in- thelium and antibodies against fibronectin [118]. Incubation tegrin family and several different integrins have been de- of the subendothelium with an anti-fibronectin antibody re- scribed to bind fibronectin (31, 51, 41, 47, sulted in a significant inhibition of platelet adhesion. Inter- IIb 3 and v 3) [98-103]. While most of the above in- estingly, this inhibition is dependent on the wall shear rates     -1 tegrins interact via a characteristic RGD- containing se- used, being most pronounced at shear rates above 800s and -1 quence, 41 and 47 bind to a site characterized by a absent at 400s [118]. In contrast to the situation on collagen LDV motif [104]. Of the above, only 51, IIb3 and or laminin, VWF is not able to bind fibronectin and therefore v3 are expressed on platelets. does not mediate platelet adhesion to fibronectin under flow. The Interaction of Cell Matrix Fibronectin with Platelets Fibronectin regulates platelet activation and thrombus formation at several checkpoints. Soluble, plasma fibronectin The interaction of fibronectin with platelets and a role for significantly contributes to thrombus stability and thrombus subendothelial fibronectin in the regulation of platelet re- growth (see below). Fibronectin within the endothelial ex- sponses has been suggested since the late seventies, although tracellular matrix also supports platelet adhesion and spread- this still remains poorly understood [105,106]. This is in part ing [25]. Interestingly, this interaction seems to be more im- due to the presence of plasma fibronectin and the embryonic portant under high shear conditions [118], typically reflect- lethality of the fibronectin-deficient mice [107], as these ing a pathological situation of a stenosed artery. have hampered investigation of the role of extracellular ma- trix fibronectin in thrombosis and haemostasis. D). Vitronectin The first evidence for the presence of fibronectin recep- Structure of Vitronectin tors on platelets came from the observation that platelets from a patient with Glanzmann’s thrombasthenia showed a Human vitronectin is a 75kDa protein which has a similar strongly reduced binding to fibronectin [100,108]. Subse- distribution to fibronectin [119]. Thus, it can be found in quently, by using chemical cross-linking and monoclonal plasma (200-500g/ml), in the extracellular matrix and in antibodies, IIb3 was shown to bind fibronectin directly platelet -granules where it is taken up from the plasma [109]. Binding of fibronectin to IIb3 involves the RGD [120-122]. 1364 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

Vitronectin Receptors injury and collagen/epinephrine induced thromboembolism, although the fibronectin plasma levels are only 20% of con- Vitronectin binds many of the proteins involved in trol mice. The enhancement of collagen/epinephrine induced thrombosis and haemostasis, including plasminogen activa- thromboembolism strongly supports the role for plasma EI- tor inhibitor-1 (PAI-1), plasminogen and heparin. Vi- IIA fibronectin, since in this model no vessel wall injury is tronectin posseses a RGD site that enables it to bind to involved. Interestingly, venous thrombosis as well as coagu- IIb 3 and the minor platelet integrin, v 3 [123]. How-     lation parameters were unchanged in these mice, strongly ever, the role of integrin v 3 in supporting platelet activa-   suggesting that the prothrombotic effect is based on an al- tion is unclear. Moreover, the ability of vitronectin to bind to tered EIIIA fibronectin - platelet interaction [131]. Although integrin IIb 3 is masked by the presence of other ligands,   the molecular basis of this enhanced thrombosis is not clari- including fibrinogen, fibronectin and VWF. fied, enhanced EIIIA fibronectin plasma levels may repre- The Interaction of Cell Matrix Vitronectin with Platelets sent a risk factor for thrombotic complications in certain pathologies [94-96]. The interaction of platelet IIb3 and v3 with cell matrix associated vitronectin may well have a supportive Thus, it seems that fibronectin present in the plasma function in platelet adhesion to the subendothelium. How- and/or platelet granules contributes to thrombus stabilization ever, experimental data to support this hypothesis are, to our in the presence of VWF and fibrinogen. Possible mecha- knowledge, not available at the moment and studies investi- nisms are the support of platelet - platelet contacts via in- gating the role of vitronectin in thrombosis and haemostasis tegrins [132] as well as stabilizing the fibrin network and are mainly focused on the plasma fraction. cross-linking of platelets to fibrin [133,134]. Another process that may support thrombus formation THE ROLE OF MATRIX PROTEINS PRESENT IN and stabilization is the assembly of plasma fibronectin into PLASMA AND PLATELET -GRANULES fibrillar extracellular matrix arrays, a process observed on VWF and fibrinogen are the two major plasma proteins several cell types including activated platelets [135-137]. supporting platelet adhesion to the extracellular matrix and Thus, activated, adherent platelets are able to assemble platelet thrombus formation through the formation of platelet plasma fibronectin into an insoluble matrix on their surface - platelet contacts. However, unexpectedly mice deficient in which may support platelet cohesion and therefore thrombus both proteins are still able to form occlusive thrombi, al- strength. The importance of this process was shown using though with strongly delayed kinetics [124]. This demon- different proteins specifically interfering with this process. In strates that other plasma proteins or platelet granular compo- flow chamber experiments, inhibition of fibronectin assem- nents are able to substitute for VWF and fibrinogen and may bly resulted in a decrease of thrombus volume and the num- therefore also play a role in thrombus formation. ber of adherent platelets by 50-60% [134]. Plasma fibronectin is clearly an important player in Plasma Fibronectin thrombus formation, although its precise role remains unde- Early experiments investigating platelet thrombus forma- fined. Last, but not least, this is emphasised by the observa- tion under flow indicated a supportive role of plasma fi- tion that increased fibronectin plasma levels are associated bronectin in thrombus formation. Using collagen-coated sur- with an increased risk of coronary artery disease [138,139]. faces, platelet adhesion as well as thrombus formation was reduced in fibronectin-depleted plasma. Interestingly, the Vitronectin diminished thrombus formation in fibronectin-depleted Depending on the in vivo model and/or vascular bed, plasma was present at low and high shear rates [125-127]. In vitronectin-deficiency has a variable effect on thrombus vivo investigations also support a role for fibronectin, as formation. Thus, under certain conditions, there is increased mice deficient in VWF and fibrinogen form occlusive embolization, reduced thrombus growth and stability and thrombi. Fibronectin was proposed as the likely substituting therefore increased time to vessel occlusion in vitronectin- candidate protein because of its structure and its ability to deficient mice [140-142]. Importantly, early platelet adhe- interact with IIb3 [128,129]. Moreover, a role for fi- sion to the injured vessel wall was not altered in these mice bronectin in thrombus formation in vivo was also shown in [140], indicating that extracellular matrix vitronectin is not mice expressing less than < 2% of the plasma fibronectin and involved and that other components of the extracellular ma- < 20% of platelet fibronectin. These animals showed delayed trix are able to compensate. This suggests that the deficiency thrombus formation, increased embolization and an in- of plasma vitronectin is the cause for the phenotype. creased time to occlusion in the presence of normal amounts of fibrinogen and VWF [124]. The importance of fibronectin The major role of plasma vitronectin in regard to haemo- was further substantiated in heterozygous mice, having 50% stasis may be its ability to bind to plasminogen activator in- of the normal fibronectin levels. This reduction was suffi- hibitor (PAI-1). PAI-1 is the major physiological inhibitor of cient to have a profound inhibitory effect on thrombus for- tissue-type plasminogen activator (tPA) and therefore an mation in vivo following arterial injury [130]. In a very re- instrumental regulator of plasmin and fibrinolysis. Vi- cent study Cauhan et al. showed, that the presence of a spe- tronectin binds PAI-1, stabilizes the active confirmation of cific fibronectin splice variant (EIIIA) in plasma may have the inhibitor and thereby increases the activity of PAI-1 strong prothrombotic effects. Mice engineered to express [143-145]. This ultimately leads to a reduced fibrinolytic EIIIA fibronectin only, have strongly accelerated thrombosis response and enhanced thrombus formation. Thus, vi- in two different in vivo thrombosis models, ferric-chloride tronectin deficiency should cause reduced PAI-1 activity and Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1365 enhanced fibrinolysis, which nicely corresponds to the ob- exposure) in the pathway suggest an effective and more served phenotype of the knockout mouse (increased emboli- uniform prevention of thrombus formation. zation, reduced thrombus growth and stability). Interestingly, iii) Thirdly, several mechanisms seem to be more important the thrombotic phenotype of vitronectin and PAI-1 double- under the high shear conditions that are typically found in deficient mice is indistinguishable from vitronectin or PAI-1 stenosed arteries. Thus, selective interference of platelet deficient mice [142]. This strongly suggest that both proteins activation in pathological high shear situations may lead serve the same mechanism, and that other vitronectin func- to only minimal disturbance of normal haemostasis. tions in thrombus formation are not significant, at least under the experimental conditions used. The connection between iv) Early platelet activation and adhesion is instrumental for PAI-1 and vitronectin is further emphasised by their co- the role of platelets in atherosclerosis development. localisation in platelet granules [146,147]. Release of PAI-1 It is important to note, that most of the currently used into the thrombus by activated platelets inhibits fibrinolysis, platelet inhibitors (phosphodiesterase inhibitors, the cy- while simultaneous liberation of vitronectin stabilises this clooxygenease inhibitor aspirin and P2Y12 antagonists) do process based on the mechanism described above. not interfere directly with these early events but are targeting Thus, based on these data, plasma vitronectin is impor- late mechanisms or secondary platelet activation. Although, tant for the regulation of thrombosis, most likely by regula- IIb3 antagonists can affect primary adhesion their main tion of PAI-1. The contribution of vitronectin associated function being the inhibition of the final common pathway of with the extracellular matrix seems to be minor, although platelet aggregation. Thus, several different approaches have this has to be said with caution since few data are available. been investigated to interfere with primary platelet activation and adhesion: PHARMACOLOGICAL INHIBITION OF PLATELET - EXTRACELLULAR MATRIX INTERACTION AS Collagen ANTI-THROMBOTIC MECHANISM The early steps of platelet-collagen interaction are seen The critical role of platelets in arterial thrombosis has as highly promising for anti-thrombotic intervention. None been well established. Furthermore, it becomes more and of the currently marketed anti-thrombotics target these more evident that platelets also play a significant role in events, last not least due to the fact that protein-protein inter- other pathological processes such as atherosclerosis [148]. actions are not easy to inhibit, especially with small mole- Thus, anti-platelet regimes belong to the standard treatment cule compounds. in patients with atherothrombotic disease for acute and pro- Biologics interacting with collagen that show some in- phylactic prevention of thrombotic events. Landmark clinical hibitory effect on platelet binding in model settings include studies for aspirin or clopidogrel have shown the enormous the recombinant leech proteins rLAPP (from Haementeria benefit of anti-platelet agents for the prevention of officinalis) and saratin (from Hirudo medicinalis), the mos- atherothrombotic events [149-151]. Moreover, dual anti- quito salivary gland protein aegyptin (from Aedes aegypti) platelet therapy combining aspirin and the P2Y12 antagonist as well as CTRP-1, a C1qTNF-related protein from plasma clopidogrel has been shown to further reduce the risk of [155-158]. All of them have been shown to act in vivo as atherothrombotic complications, with a 20% relative risk inhibitors of platelet function. The exact collagen binding reduction in patients with unstable angina or non - ST myo- site of LAPP is not known. For saratin there is interference cardial infarction as compared to aspirin mono-therapy seen with both VWF and 2-I-domain binding to collagen, [152]. Despite this significant achievement, a high number of suggesting a certain overlap in binding epitopes on collagen patients experience thrombotic events while under anti- [159]. Recently the 3D structure of saratin was solved. The platelet treatment. Atherothrombosis is still a leading cause data from docking experiments with collagen model peptides of death worldwide and, importantly, the absolute number of lead to the conclusion that despite a similar global fold of deaths from this disease continues to increase [153,154]. LAPP and saratin there is a significant difference in collagen Thus, there is a large unmet clinical need and much effort is binding [160]. CTRP-1 blocks the binding of vWF-A3 do- put into the improvement of existing drugs, e.g. the devel- main to collagen and thus inhibits collagen-induced platelet opment of new P2Y12 inhibitors, but also in the exploration aggregation [157]. Aegyptin has been shown to block the of new platelet (thrombus) - inhibitory mechanisms. In this interaction of the three glycoprotein receptors, GPIb, GPVI regard interference with primary platelet activation and ad- and 21 with collagen, thereby showing that it is distinct hesion to the extracellular matrix is considered to be a poten- from the other molecules described above [156]. Thus it tially attractive approach. This is based on the following as- might serve as a powerful tool to evaluate the relative impor- sumptions and supporting experimental data. tance of all three receptors in the very early steps of platelet i) Inhibition of the initial step of platelet activation prevents adhesion and activation. Targeting collagen itself would formation of activated platelets. This mechanism has the limit the action of an inhibitor to sites where collagen is ex- potential to be very effective in blocking thrombus for- posed but also limit the use of such inhibitors for systemic mation and growth. application. ii) The interaction of GPIb and VWF, and of GPVI and col- An interesting mechanism is the use of the collagen bind- lagen, appear to have less redundancy with other path- ing domain of GPVI as competitor for the binding of platelet ways as compared to inhibitors of secondary platelet ac- associated GPVI. Since only dimeric GPVI has a significant tivation. Little redundancy (in the absence of tissue factor affinity for collagen the therapeutic protein is expressed as a human immunoglobulin Fc domain fusion protein [161]. 1366 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

Such GPVI-Fc fusion proteins have been shown to inhibit a series of molecules (OM) partly having crossreactivity to platelet adhesion to collagen and thrombus formation under the rat protein [172]. Fab fragments of one of the generated flow. Moreover, they prevent platelet adhesion to the injured antibodies, OM4, effectively inhibited thrombus formation in vessel and thrombus formation in vivo (endothelial denuda- rats (modified Folts model of cyclic flow reduction in carotid tion of mouse carotid artery by vigorous ligation) [162]. In artery). Importantly, the Fab fragment did not induce throm- contrast, Gruner et al. showed in the same in vivo thrombosis bocytopenia, GPVI depletion nor did treatment with OM4 model an only partial inhibition at high concentration of the prolong the bleeding [175]. This clearly supports the view, fusion protein, while antibodies against GPVI were highly that anti-GPVI treatment in a competitive manner can be a inhibitory [163]. The reason for this discrepancy is unclear promising antithrombotic mechanism, with an excellent but may be based on slight but significant differences in the benefit - safety window. execution of the animal model. Support for the use of GPVI- Fc as therapeutic protein comes also from a study showing 21 that local delivery of the protein to the site of an injured ca- As the first receptor on platelets mediating firm adhesion rotid artery (ballon injury in rabbits) is able to substantially to collagen, this integrin is also a potential target for phar- attenuate thrombus formation [164,165]. This directed deliv- maceutical intervention. Derivatives of specific 21 inhib- ery is an interesting option since it ensures high local con- iting snake venom components like rhodocetin might serve centrations of the fusion protein plus it contains the anti- as starting points for such antagonists [176]. There are no thrombotic effect to the targeted site, which should be fa- reports of clinical trials with direct-acting 21 antagonists. vourable in terms of bleeding. The clinical development of Eisai is investigating an 21-expression inhibitor, E7820, GPVI-Fc (PR-15) is pursued by Trigen, Munich, Germany. for cancer indications in Phase II because of the role of the A Phase I study has been successfully finalized [166]. integrin in angiogenesis [177]. Small molecule antagonists of 2 1 for cancer and thrombosis have been described [178] GPVI   (Biotie website) but further development has not been re- The crystal structure of the GPVI collagen binding do- ported. main has been recently described, which may open some avenues for the identification of small molecule inhibitors VWF [167]. However, based on the large interaction surface be- As the interaction partner in the first step of platelet ad- tween GPVI and collagen the use of biotherapeutic proteins hesion, VWF is a strong target for consideration of pharma- (antibodies or fragments thereof) seems to be the most prom- cological intervention. Targeting VWF would also have the ising alternative to find potent GPVI inhibitors. Furthermore, advantage of avoiding complications based on potential side- anti GPVI antibodies are able to induce receptor down- effects through targeting of GPIb-IX-V that might lead to regulation, which may reflect an additional anti-thrombotic thrombocytopenia given the critical role of GPIb-IX-V in mechanism. It has been shown in vivo that mice, treated with platelet formation. As discussed above the binding region of anti-GPVI antibodies are rendered GPVI-deficient and have VWF for the GPIb-receptor complex on platelets becomes significant antithrombotic characteristics in several thrombo- only available under conditions of high shear. sis models [168]. This effect of anti-GPVI antibodies nicely explains much earlier observations in patients with autoanti- AJW200 is a humanized anti-VWF-A1 antibody that has bodies against the collagen receptor. These patients also been reported to be well tolerated in a clinical Phase I study. showed a defect in collagen-induced platelet activation and This antibody had been shown to inhibit shear-induced plate- GPVI deficiency on their platelets [169]. However, the asso- let aggregation and to reduce thrombosis in coronary arteries ciation of antibody-induced GPVI down-regulation with par- in a canine coronary artery model [179,180]. Further, it tial platelet activation and transient thrombocytopenia has showed a favorable effect - bleeding ratio versus the com- been described, which makes this approach futile for clinical parator, abciximab (IIb3 inhibitor), suggesting a safer use [168]. Thus, more basic research in the principles of pharmacological profile in dogs. GPVI downregulation is warranted. Nevertheless, a recently The two most advanced approaches in targeting VWF in published elucidation of the signalling pathways involved in clinical testing are an aptamer, a protein-binding oligonu- GPVI downregulation and the description of an antibody cleotide, as well as a nanobody, a “heavy-chain only” anti- which seems to cause a decrease in surface expression of the body derived from camelids (for review see [181,182]). receptor without obvious signs of platelet activation or thrombocytopenia may open new avenues for the clinical use Archemix is investigating ARC1779, a pegylated ap- of this mechanism [170,171]. tamer binding to the VWF-A1 domain [183]. In a Phase I study in healthy volunteers it was well tolerated, with an Another option is the classical competitive inhibition of apparent elimination half-life time of 2 hours and no bleed- the collagen binding site of GPVI. To avoid any platelet ac- ing episodes, while inhibiting platelet function dose- tivation, monovalent proteins such as Fab fragments are the dependently. ARC1779 is currently in Phase II clinical de- molecules of choice. A number of antibodies against human velopment, assessing the safety, pharmacokinetics and GPVI have been generated by several groups from academia pharmacodynamics in patients with VWF-mediated platelet and industry [171-174]. Characterization of these molecules function disorders, including TTP (Thrombotic thrombocy- in vivo has been often hampered by the very narrow cross- topenia purpura), as well as acute coronary syndrome pa- reactivity of anti-human GPVI antibodies generated in mice. tients undergoing PCI (Archemix website). Since TTP is To circumvent this, Matsumoto et al. used GPVI-knock out associated with an increased plasma concentration of ultra- mice for the generation of anti GPVI antibodies, resulting in Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1367 large, highly prothrombotic multimers of VWF, a direct in- CONCLUSIONS hibition of VWF seems to be here particularly promising. The subendothelial extracellular matrix has been shown However, clinical studies will show if these predictions hold to be a powerful activator of platelets. Moreover, the inter- true. play between components of the subendothelial matrix and This is also the case for Ablynx’ ALX-0081, an anti- specific platelet receptors causes a stable adhesion of acti- VWF nanobody directed against VWF. The data from ani- vated platelets to the exposed subendothelium, which initi- mal studies in a modified Folts-model in baboons showed a ates the formation of a haemostatic plug but also localizes more pronounced effect on inhibition of thrombus formation the haemostatic reaction to the site of injury. These proper- than a combination of aspirin, heparin and clopidogrel, with ties underline a critical, life saving role of the subendothelial less bleeding complications [184]. The half-life of ALX- matrix in haemostasis. However, pathological changes 0081 in baboons was similar to the half-life of its target, within the vessel wall which include a remodelling of the VWF-A1 (8 hours). A Phase I study in healthy volunteers subendothelial matrix and a dysfunctional endothelial layer has been successfully concluded and confirmed that ALX- give rise to atherosclerosis and life threatening thrombotic 0081 adopts at least the half-life of it’s target. The nanobody complications. Besides intensive medical treatment athero- showed no immungenicity, was safe and well tolerated at all thrombosis is still a leading cause of death worldwide. doses tested, and no serious adverse events were reported. Therefore, the development of new anti- thrombotic and The anti-thrombotic effect was dose-dependent in a range anti-atherosclerotic mechanisms is needed. Because of the from 2 - 12 mg. A Phase Ib study in patients with stable an- importance of the subendothelial matrix for these processes gina undergoing PCI is ongoing. Phase II is scheduled to several components represent interesting drug targets. Plate- start by the end of 2008 (Ablynx website). lets interact with vessel wall associated laminin, fibronectin, vitronectin and collagen. Of these, collagen and associated GPIb VWF seem, however, of particular importance for platelet The receptor for VWF-A1 on platelets, the GPIb-V-IX activation and adhesion. Thus, blocking the interaction of complex, is another option for anti-thrombotic intervention. platelets with collagen (via its receptors GPVI, 21 or col- Besides VWF it also interacts with TSP, P-selectin, throm- lagen binding peptides) and VWF (GPIb inhibitors or mole- bin, Mac-1, high-molecular weight kininogen, FXI and FXII cules against VWF) seems to represent the most promising (reviewed in [185]). All these interactions may play a role mechanism. Several approaches have been initiated and the under pathophysiological conditions, while their relative first molecules have entered early clinical testing. The very importance is still under discussion. The main concern in promising investigational and (pre)clinical data raise hopes targeting GPIb is thrombocytopenia. Thus it will be crucial that these approaches may significantly improve the treat- to monitor this potentially life-threatening condition closely ment of atherothrombotic diseases. while investigating GPIb inhibitory principles. For in vivo ACKNOWLEDGEMENTS studies, Fab-fragments of anti-GPIb antibodies where suc- cessfully used, without causing thrombocytopenia. One ex- The research in the group of SPW is supported by the ample is the humanized anti GPIb antibody 6B4, which was British Heart Foundation, MRC and Wellcome Trust. SPW applied in a model of arterial thrombosis (modified Folts holds a BHF Chair. I thank Gemma Fuller for help with the model) in baboons [186]. While the bolus injection of 0.6 figure. I would like to express considerable gratitude for the mg/kg or 2 mg/kg 6B4 Fab fragments led to a significant many hours of helpful discussion to Drs Peter Wonerow and anti-thrombotic effect, platelet count and bleeding time were Horst Blum. not significantly changed. When GPIb -/- mice were con- structed they showed prolonged bleeding time, the formation REFERENCES of giant platelets and thrombocytopenia, providing a model [1] Kramer JM. Basement membranes. WormBook 2005; 1-15. of the Bernard-Soulier Syndrome [187]. Therefore, to inves- [2] LeBleu VS, Macdonald B, Kalluri R. Structure and function of tigate the effect of GPIb deficiency without these secondary basement membranes. Exp Biol Med (Maywood) 2007; 232(9): effects, transgenic mice where most of the extracellular part 1121-9. of GPIb was replaced by IL-4R (IL-4R/GPIb-tg mice) [3] Stenman S, Vaheri A. Distribution of a major connective tissue were generated. These mice have platelets of normal size and protein, fibronectin, in normal human tissues. J Exp Med 1978; 147(4): 1054-64. only mild thrombocytopenia with an approx. 30% reduction [4] Plenz GA, Deng MC, Robenek H, Volker W. Vascular collagens: in platelet count. In a FeCl3-induced thrombosis model it was spotlight on the role of type VIII collagen in atherogenesis. Athero- demonstrated that platelet adhesion to ECM in arterioles of sclerosis 2003; 166(1): 1-11. these mice was completely inhibited [188]. [5] Seyama Y, Wachi H. Atherosclerosis and matrix dystrophy. J Athe- roscler Thromb 2004; 11(5): 236-45. Due to the diverse interactions of GPIb as mentioned [6] Nieswandt B, Watson SP. Platelet-collagen interaction: is GPVI the above, the effect of GPIb blockade could be more marked central receptor? Blood 2003; 102(2): 449-61. than the effect of vWF inhibition. However it still has to be [7] Ruggeri ZM. Platelets in atherothrombosis. Nat Med 2002; 8(11): demonstrated in direct comparison in suitable animal models 1227-34. that this holds true and is not associated with thrombocy- [8] Hayden MR, Sowers JR, Tyagi SC. The central role of vascular extracellular matrix and basement membrane remodeling in meta- topenia. Several further approaches for targeting GPIb have bolic syndrome and type 2 diabetes: the matrix preloaded. Cardio- been reported (for review [189,190], but none has yet vasc Diabetol 2005; 4(1): 9. reached phases of clinical testing. [9] Rand JH, Sussman II, Gordon RE, Chu SV, Solomon V. Localiza- tion of factor-VIII-related antigen in human vascular subendothe- lium. Blood 1980; 55(5): 752-6. 1368 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

[10] Garratt KN, Edwards WD, Kaufmann UP, Vlietstra RE, Holmes [32] Jung SM, Takemura Y, Imamura Y, Hayashi T, Adachi E, Moroi DR, Jr. Differential histopathology of primary atherosclerotic and M. Collagen-type specificity of glycoprotein VI as a determinant of restenotic lesions in coronary arteries and saphenous vein bypass platelet adhesion. Platelets 2008; 19(1): 32-42. grafts: analysis of tissue obtained from 73 patients by directional [33] Henrita van ZG, Saelman EU, Schut-Hese KM, Wu YP, Slootweg atherectomy. J Am Coll Cardiol 1991; 17(2): 442-8. PJ, Nieuwenhuis HK, et al. Platelet adhesion to collagen type IV [11] Strauss BH, Chisholm RJ, Keeley FW, Gotlieb AI, Logan RA, under flow conditions. Blood 1996; 88(10): 3862-71. Armstrong PW. Extracellular matrix remodeling after balloon an- [34] Jarvis GE, Atkinson BT, Snell DC, Watson SP. Distinct roles of gioplasty injury in a rabbit model of restenosis. Circ Res 1994; GPVI and integrin alpha(2)beta(1) in platelet shape change and ag- 75(4): 650-8. gregation induced by different collagens. Br J Pharmacol 2002; [12] Davis GE, Senger DR. Endothelial extracellular matrix: biosynthe- 137(1): 107-17. sis, remodeling, and functions during vascular morphogenesis and [35] Penz S, Reininger AJ, Brandl R, Goyal P, Rabie T, Bernlochner I, neovessel stabilization. Circ Res 2005; 97(11): 1093-107. et al. Human atheromatous plaques stimulate thrombus formation [13] Orr AW, Ginsberg MH, Shattil SJ, Deckmyn H, Schwartz MA. by activating platelet glycoprotein VI. FASEB J 2005; 19(8): 898- Matrix-specific suppression of integrin activation in shear stress 909. signaling. Mol Biol Cell 2006; 17(11): 4686-97. [36] Clemetson JM, Polgar J, Magnenat E, Wells TN, Clemetson KJ. [14] Orr AW, Sanders JM, Bevard M, Coleman E, Sarembock IJ, The platelet collagen receptor glycoprotein VI is a member of the Schwartz MA. The subendothelial extracellular matrix modulates immunoglobulin superfamily closely related to FcalphaR and the NF-kappaB activation by flow: a potential role in atherosclerosis. J natural killer receptors. J Biol Chem 1999; 274(41): 29019-24. Cell Biol 2005; 169(1): 191-202. [37] Nieuwenhuis HK, Akkerman JW, Houdijk WP, Sixma JJ. Human [15] Gawaz M, Langer H, May AE. Platelets in inflammation and blood platelets showing no response to collagen fail to express sur- atherogenesis. J Clin Invest 2005; 115(12): 3378-84. face glycoprotein Ia. Nature 1985; 318(6045): 470-2. [16] Ruggeri ZM. The role of von Willebrand factor in thrombus forma- [38] Farndale RW, Lisman T, Bihan D, Hamaia S, Smerling CS, Pugh tion. Thromb Res 2007; 120(Suppl 1): S5-9. N, et al. Cell-collagen interactions: the use of peptide Toolkits to [17] Doggett TA, Girdhar G, Lawshe A, Schmidtke DW, Laurenzi IJ, investigate collagen-receptor interactions. Biochem Soc Trans Diamond SL, et al. Selectin-like kinetics and biomechanics pro- 2008; 36(Pt 2): 241-50. mote rapid platelet adhesion in flow: the GPIb(alpha)-vWF tether [39] Jarvis GE, Raynal N, Langford JP, Onley DJ, Andrews A, bond. Biophys J 2002; 83(1): 194-205. Smethurst PA, et al. Identification of a major GpVI-binding locus [18] Savage B, mus-Jacobs F, Ruggeri ZM. Specific synergy of multiple in human type III collagen. Blood 2008; 111(10): 4986-96. substrate-receptor interactions in platelet thrombus formation under [40] Lisman T, Raynal N, Groeneveld D, Maddox B, Peachey AR, flow. Cell 1998; 94(5): 657-66. Huizinga EG, et al. A single high-affinity binding site for von [19] Denis C, Methia N, Frenette PS, Rayburn H, Ullman-Cullere M, Willebrand factor in collagen III, identified using synthetic triple- Hynes RO, et al. A mouse model of severe von Willebrand disease: helical peptides. Blood 2006; 108(12): 3753-6. defects in hemostasis and thrombosis. Proc Natl Acad Sci USA [41] Sakariassen KS, Bolhuis PA, Sixma JJ. Human blood platelet adhe- 1998; 95(16): 9524-9. sion to artery subendothelium is mediated by factor VIII-Von [20] Inoue O, Suzuki-Inoue K, McCarty OJ, Moroi M, Ruggeri ZM, Willebrand factor bound to the subendothelium. Nature 1979; Kunicki TJ, et al. Laminin stimulates spreading of platelets through 279(5714): 636-8. integrin alpha6beta1-dependent activation of GPVI. Blood 2006; [42] Obert B, Houllier A, Meyer D, Girma JP. Conformational changes 107(4): 1405-12. in the A3 domain of von Willebrand factor modulate the interaction [21] Shattil SJ, Newman PJ. Integrins: dynamic scaffolds for adhesion of the A1 domain with platelet . Blood 1999; 93(6): and signaling in platelets. Blood 2004; 104(6): 1606-15. 1959-68. [22] Sarratt KL, Chen H, Zutter MM, Santoro SA, Hammer DA, Kahn [43] Ruggeri ZM. Structure and function of von Willebrand factor. ML. GPVI and alpha2beta1 play independent critical roles during Thromb Haemost 1999; 82(2): 576-84. platelet adhesion and aggregate formation to collagen under flow. [44] Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet adhesion Blood 2005; 106(4): 1268-77. by arrest onto fibrinogen or translocation on von Willebrand factor. [23] McCarty OJ, Zhao Y, Andrew N, Machesky LM, Staunton D, Cell 1996; 84(2): 289-97. Frampton J, et al. Evaluation of the role of platelet integrins in fi- [45] Romijn RA, Bouma B, Wuyster W, Gros P, Kroon J, Sixma JJ, et bronectin-dependent spreading and adhesion. J Thromb Haemost al. Identification of the collagen-binding site of the von Willebrand 2004; 2(10): 1823-33. factor A3-domain. J Biol Chem 2001; 276(13): 9985-91. [24] Inoue O, Suzuki-Inoue K, Ozaki Y. Redundant mechanism of [46] Denis C, Baruch D, Kielty CM, Ajzenberg N, Christophe O, Meyer platelet adhesion to laminin and collagen under flow: involvement D. Localization of von Willebrand factor binding domains to endo- of von Willebrand factor and glycoprotein Ib-IX-V. J Biol Chem thelial extracellular matrix and to type VI collagen. Arterioscler 2008; 283(24): 16279-82. Thromb 1993; 13(3): 398-406. [25] Gruner S, Prostredna M, Schulte V, Krieg T, Eckes B, Brakebusch [47] Rand JH, Patel ND, Schwartz E, Zhou SL, Potter BJ. 150-kD von C, et al. Multiple integrin-ligand interactions synergize in shear- Willebrand factor binding protein extracted from human vascular resistant platelet adhesion at sites of arterial injury in vivo. Blood subendothelium is type VI collagen. J Clin Invest 1991; 88(1): 253- 2003; 102(12): 4021-7. 9. [26] Brass LF, Zhu L, Stalker TJ. Minding the gaps to promote throm- [48] Reininger AJ, Heijnen HF, Schumann H, Specht HM, Schramm W, bus growth and stability. J Clin Invest 2005; 115(12): 3385-92. Ruggeri ZM. Mechanism of platelet adhesion to von Willebrand [27] Cesarman-Maus G, Hajjar KA. Molecular mechanisms of fibri- factor and microparticle formation under high shear stress. Blood nolysis. Br J Haematol 2005; 129(3): 307-21. 2006; 107(9): 3537-45. [28] Veit G, Kobbe B, Keene DR, Paulsson M, Koch M, Wagener R. [49] Moser M, Nieswandt B, Ussar S, Pozgajova M, Fassler R. Kindlin- Collagen XXVIII, a novel von Willebrand factor A domain- 3 is essential for integrin activation and platelet aggregation. Nat containing protein with many imperfections in the collagenous do- Med 2008; 14(3): 325-30. main. J Biol Chem 2006; 281(6): 3494-504. [50] Tadokoro S, Shattil SJ, Eto K, Tai V, Liddington RC, de Pereda [29] Brodsky B, Persikov AV. Molecular structure of the collagen triple JM, et al. Talin binding to integrin beta tails: a final common step helix. Adv Protein Chem 2005; 70: 301-39. in integrin activation. Science 2003; 302(5642): 103-6. [30] Voss B, Rauterberg J. Localization of collagen types I, III, IV and [51] Tuckwell D, Calderwood DA, Green LJ, Humphries MJ. Integrin V, fibronectin and laminin in human arteries by the indirect im- alpha 2 I-domain is a binding site for collagens. J Cell Sci 1995; munofluorescence method. Pathol Res Pract 1986; 181(5): 568-75. 108 ( Pt 4): 1629-37. [31] Timpl R, Wiedemann H, van D, V, Furthmayr H, Kuhn K. A net- [52] Emsley J, Knight CG, Farndale RW, Barnes MJ, Liddington RC. work model for the organization of type IV collagen molecules in Structural basis of collagen recognition by integrin alpha2beta1. basement membranes. Eur J Biochem 1981; 120(2): 203-11. Cell 2000; 101(1): 47-56. Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1369

[53] Emsley J, King SL, Bergelson JM, Liddington RC. Crystal struc- mouse homologue specifically bind alpha5 chain-containing human ture of the I domain from integrin alpha2beta1. J Biol Chem 1997; laminin with high affinity. Blood 2001; 97(1): 312-20. 272(45): 28512-7. [75] Udani M, Zen Q, Cottman M, Leonard N, Jefferson S, Daymont C, [54] Buensuceso C, de Virgilio M., Shattil SJ. Detection of integrin et al. Basal molecule/lutheran protein. The receptor alpha IIbbeta 3 clustering in living cells. J Biol Chem 2003; critical for sickle cell adhesion to laminin. J Clin Invest 1998; 278(17): 15217-24. 101(11): 2550-8. [55] Shimaoka M, Takagi J, Springer TA. Conformational regulation of [76] Zen Q, Cottman M, Truskey G, Fraser R, Telen MJ. Critical factors integrin structure and function. Annu Rev Biophys Biomol Struct in basal cell adhesion molecule/lutheran-mediated adhesion to 2002; 31: 485-516. laminin. J Biol Chem 1999; 274(2): 728-34. [56] Lahav J, Gofer-Dadosh N, Luboshitz J, Hess O, Shaklai M. Protein [77] Nelson J, McFerran NV, Pivato G, Chambers E, Doherty C, Steele disulfide isomerase mediates integrin-dependent adhesion. FEBS D, et al. The 67 kDa laminin receptor: structure, function and role Lett 2000; 475(2): 89-92. in disease. Biosci Rep 2008; 28(1): 33-48. [57] Siljander PR, Hamaia S, Peachey AR, Slatter DA, Smethurst PA, [78] Sciandra F, Gawlik KI, Brancaccio A, Durbeej M. Dystroglycan: a Ouwehand WH, et al. Integrin activation state determines selectiv- possible mediator for reducing congenital muscular dystrophy? ity for novel recognition sites in fibrillar collagens. J Biol Chem Trends Biotechnol 2007; 25(6): 262-8. 2004; 279(46): 47763-72. [79] Hemler ME, Crouse C, Takada Y, Sonnenberg A. Multiple very [58] Nieswandt B, Brakebusch C, Bergmeier W, Schulte V, Bouvard D, late antigen (VLA) heterodimers on platelets. Evidence for distinct Mokhtari-Nejad R, et al. Glycoprotein VI but not alpha2beta1 in- VLA-2, VLA-5 (fibronectin receptor), and VLA-6 structures. J tegrin is essential for platelet interaction with collagen. EMBO J Biol Chem 1988; 263(16): 7660-5. 2001; 20(9): 2120-30. [80] Ill CR, Engvall E, Ruoslahti E. Adhesion of platelets to laminin in [59] Kuijpers MJ, Schulte V, Bergmeier W, Lindhout T, Brakebusch C, the absence of activation. J Cell Biol 1984; 99(6): 2140-5. Offermanns S, et al. Complementary roles of glycoprotein VI and [81] Sonnenberg A, Modderman PW, Hogervorst F. Laminin receptor alpha2beta1 integrin in collagen-induced thrombus formation in on platelets is the integrin VLA-6. Nature 1988; 336(6198): 487-9. flowing whole blood ex vivo. FASEB J 2003; 17(6): 685-7. [82] Lesot H, Kuhl U, Mark KV. Isolation of a laminin-binding protein [60] Auger JM, Kuijpers MJ, Senis YA, Watson SP, Heemskerk JW. from muscle cell membranes. EMBO J 1983; 2(6): 861-5. Adhesion of human and mouse platelets to collagen under shear: a [83] Terranova VP, Rao CN, Kalebic T, Margulies IM, Liotta LA. unifying model. FASEB J 2005; 19(7): 825-7. Laminin receptor on human breast carcinoma cells. Proc Natl Acad [61] Hallmann R, Horn N, Selg M, Wendler O, Pausch F, Sorokin LM. Sci USA 1983; 80(2): 444-8. Expression and function of laminins in the embryonic and mature [84] Tandon NN, Holland EA, Kralisz U, Kleinman HK, Robey FA, vasculature. Physiol Rev 2005; 85(3): 979-1000. Jamieson GA. Interaction of human platelets with laminin and [62] Tunggal P, Smyth N, Paulsson M, Ott MC. Laminins: structure and identification of the 67 kDa laminin receptor on platelets. Biochem genetic regulation. Microsc Res Tech 2000; 51(3): 214-27. J 1991; 274 ( Pt 2): 535-42. [63] Paulsson M, Deutzmann R, Timpl R, Dalzoppo D, Odermatt E, [85] Hindriks G, Ijsseldijk MJ, Sonnenberg A, Sixma JJ, de Groot PG. Engel J. Evidence for coiled-coil alpha-helical regions in the long Platelet adhesion to laminin: role of Ca2+ and Mg2+ ions, shear arm of laminin. EMBO J 1985; 4(2): 309-16. rate, and platelet membrane glycoproteins. Blood 1992; 79(4): 928- [64] Aumailley M, Timpl R, Sonnenberg A. Antibody to integrin alpha 35. 6 subunit specifically inhibits cell-binding to laminin fragment 8. [86] Matsui T, Kunishima S, Hamako J, Katayama M, Kamiya T, Naoe Exp Cell Res 1990; 188(1): 55-60. T, et al. Interaction of von Willebrand factor with the extracellular [65] Paulsson M. The role of Ca2+ binding in the self-aggregation of matrix and glycocalicin under static conditions. J Biochem 1997; laminin-nidogen complexes. J Biol Chem 1988; 263(11): 5425-30. 121(2): 376-81. [66] Yurchenco PD, Tsilibary EC, Charonis AS, Furthmayr H. Laminin [87] Wagner DD, Urban-Pickering M, Marder VJ. Von Willebrand polymerization in vitro. Evidence for a two-step assembly with protein binds to extracellular matrices independently of collagen. domain specificity. J Biol Chem 1985; 260(12): 7636-7644. Proc Natl Acad Sci USA 1984; 81(2): 471-5. [67] Yurchenco PD, Cheng YS, Colognato H. Laminin forms an inde- [88] Mosher DF. Fibronectin. Prog Hemost Thromb 1980; 5: 111-51. pendent network in basement membranes. J Cell Biol 1992; 117(5): [89] Hynes RO. Fibronectins. Sci Am 1986; 254(6): 42-51. 1119-33. [90] Plow EF, Birdwell C, Ginsberg MH. Identification and quantitation [68] Nishiuchi R, Takagi J, Hayashi M, Ido H, Yagi Y, Sanzen N, et al. of platelet-associated fibronectin antigen. J Clin Invest 1979; 63(3): Ligand-binding specificities of laminin-binding integrins: a com- 540-3. prehensive survey of laminin-integrin interactions using recombi- [91] Zucker MB, Mosesson MW, Broekman MJ, Kaplan KL. Release of nant alpha3beta1, alpha6beta1, alpha7beta1 and alpha6beta4 in- platelet fibronectin (cold-insoluble globulin) from alpha granules tegrins. Matrix Biol 2006; 25(3): 189-97. induced by thrombin or collagen; lack of requirement for plasma [69] Scheele S, Nystrom A, Durbeej M, Talts JF, Ekblom M, Ekblom P. fibronectin in ADP-induced platelet aggregation. Blood 1979; Laminin isoforms in development and disease. J Mol Med 2007; 54(1): 8-12. 85(8): 825-36. [92] Paul JI, Schwarzbauer JE, Tamkun JW, Hynes RO. Cell-type- [70] Cherepanova OA, Kalmykova N, Petrov YP, Blinova M, Pinaev G. specific fibronectin subunits generated by alternative splicing. J Contribution of alpha2beta1, alpha3beta1, alpha6beta4 integrins Biol Chem 1986; 261(26): 12258-65. and 67 kDa laminin receptor to the interaction of epidermoid carci- [93] Peters JH, Chen GE, Hynes RO. Fibronectin isoform distribution in noma A-431 cells with laminin-2/4. Cell Biol Int 2006; 30(10): the mouse. II. Differential distribution of the alternatively spliced 784-92. EIIIB, EIIIA, and V segments in the adult mouse. Cell Adhes [71] Pfaff M, Gohring W, Brown JC, Timpl R. Binding of purified Commun 1996; 4(2): 127-48. collagen receptors (alpha 1 beta 1, alpha 2 beta 1) and RGD- [94] Castellanos M, Leira R, Serena J, Blanco M, Pedraza S, Castillo J, dependent integrins to laminins and laminin fragments. Eur J Bio- et al. Plasma cellular-fibronectin concentration predicts hemor- chem 1994; 225(3): 975-84. rhagic transformation after thrombolytic therapy in acute ischemic [72] Parsons SF, Mallinson G, Judson PA, Anstee DJ, Tanner MJ, Dan- stroke. Stroke 2004; 35(7): 1671-6. iels GL. Evidence that the Lub blood group antigen is located on [95] Kanters SD, Banga JD, Algra A, Frijns RC, Beutler JJ, Fijnheer R. red cell membrane glycoproteins of 85 and 78 kd. Transfusion Plasma levels of cellular fibronectin in diabetes. Diabetes Care 1987; 27(1): 61-3. 2001; 24(2): 323-7. [73] El NW, Gane P, Colin Y, Bony V, Rahuel C, Galacteros F, et al. [96] Tan MH, Sun Z, Opitz SL, Schmidt TE, Peters JH, George EL. The Lutheran blood group glycoproteins, the erythroid receptors Deletion of the alternatively spliced fibronectin EIIIA domain in for laminin, are adhesion molecules. J Biol Chem 1998; 273(27): mice reduces atherosclerosis. Blood 2004; 104(1): 11-8. 16686-93. [97] Magnusson MK, Mosher DF. Fibronectin: structure, assembly, and [74] Parsons SF, Lee G, Spring FA, Willig TN, Peters LL, Gimm JA, et cardiovascular implications. Arterioscler Thromb Vasc Biol 1998; al. Lutheran blood group glycoprotein and its newly characterized 18(9): 1363-70. 1370 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

[98] Charo IF, Nannizzi L, Smith JW, Cheresh DA. The vitronectin binding competent population of Vn within platelet alpha-granules. receptor alpha v beta 3 binds fibronectin and acts in concert with Blood 1996; 88(2): 552-60. alpha 5 beta 1 in promoting cellular attachment and spreading on [122] Preissner KT. Structure and biological role of vitronectin. Annu fibronectin. J Cell Biol 1990; 111(6 Pt 1): 2795-800. Rev Cell Biol 1991; 7: 275-310. [99] Elices MJ, Urry LA, Hemler ME. Receptor functions for the in- [123] Ekmekci OB, Ekmekci H. Vitronectin in atherosclerotic disease. tegrin VLA-3: fibronectin, collagen, and laminin binding are dif- Clin Chim Acta 2006; 368(1-2): 77-83. ferentially influenced by Arg-Gly-Asp peptide and by divalent [124] Ni H, Denis CV, Subbarao S, Degen JL, Sato TN, Hynes RO, et al. cations. J Cell Biol 1991; 112(1): 169-81. Persistence of platelet thrombus formation in arterioles of mice [100] Ginsberg MH, Forsyth J, Lightsey A, Chediak J, Plow EF. Reduced lacking both von Willebrand factor and fibrinogen. J Clin Invest surface expression and binding of fibronectin by thrombin- 2000; 106(3): 385-92. stimulated thrombasthenic platelets. J Clin Invest 1983; 71(3): 619- [125] Houdijk WP, Sakariassen KS, Nievelstein PF, Sixma JJ. Role of 24. factor VIII-von Willebrand factor and fibronectin in the interaction [101] Pytela R, Pierschbacher MD, Ruoslahti E. Identification and isola- of platelets in flowing blood with monomeric and fibrillar human tion of a 140 kd cell surface glycoprotein with properties expected collagen types I and III. J Clin Invest 1985; 75(2): 531-40. of a fibronectin receptor. Cell 1985; 40(1): 191-8. [126] Bastida E, Escolar G, Ordinas A, Sixma JJ. Fibronectin is required [102] Akiyama SK, Yamada KM. The interaction of plasma fibronectin for platelet adhesion and for thrombus formation on subendothe- with fibroblastic cells in suspension. J Biol Chem 1985; 260(7): lium and collagen surfaces. Blood 1987; 70(5): 1437-42. 4492-500. [127] Nievelstein PF, D'Alessio PA, Sixma JJ. Fibronectin in platelet [103] Takada Y, Huang C, Hemler ME. Fibronectin receptor structures in adhesion to human collagen types I and III. Use of nonfibrillar and the VLA family of heterodimers. Nature 1987; 326(6113): 607-9. fibrillar collagen in flowing blood studies. Arteriosclerosis 1988; [104] Lobb RR, Hemler ME. The pathophysiologic role of alpha 4 in- 8(2): 200-6. tegrins in vivo. J Clin Invest 1994; 94(5): 1722-8. [128] Ni H, Yuen PS, Papalia JM, Trevithick JE, Sakai T, Fassler R, et [105] Grinnell F, Feld M, Snell W. The influence of cold insoluble globu- al. Plasma fibronectin promotes thrombus growth and stability in lin on platelet morphological response to substrata. Cell Biol Int injured arterioles. Proc Natl Acad Sci USA 2003; 100(5): 2415-9. Rep 1979; 3(7): 585-92. [129] Yang H, Reheman A, Chen P, Zhu G, Hynes RO, Freedman J, et [106] Bensusan HB, Koh TL, Henry KG, Murray BA, Culp LA. Evi- al. Fibrinogen and von Willebrand factor-independent platelet ag- dence that fibronectin is the collagen receptor on platelet mem- gregation in vitro and in vivo. J Thromb Haemost 2006; 4(10): branes. Proc Natl Acad Sci USA 1978; 75(12): 5864-8. 2230-7. [107] George EL, Georges-Labouesse EN, Patel-King RS, Rayburn H, [130] Matuskova J, Chauhan AK, Cambien B, Astrof S, Dole VS, Piffath Hynes RO. Defects in mesoderm, neural tube and vascular devel- CL, et al. Decreased plasma fibronectin leads to delayed thrombus opment in mouse embryos lacking fibronectin. Development 1993; growth in injured arterioles. Arterioscler Thromb Vasc Biol 2006; 119(4): 1079-91. 26(6): 1391-6. [108] Plow EF, Ginsberg MH. Specific and saturable binding of plasma [131] Chauhan AK, Kisucka J, Cozzi MR, Walsh MT, Moretti FA, Bat- fibronectin to thrombin-stimulated human platelets. J Biol Chem tiston M, et al. Prothrombotic effects of fibronectin isoforms con- 1981; 256(18): 9477-82. taining the EDA domain. Arterioscler Thromb Vasc Biol 2008; [109] Gardner JM, Hynes RO. Interaction of fibronectin with its receptor 28(2): 296-301. on platelets. Cell 1985; 42(2): 439-48. [132] Thurlow PJ, Kenneally DA, Connellan JM. The role of fibronectin [110] Ruoslahti E, Pierschbacher MD. New perspectives in cell adhesion: in platelet aggregation. Br J Haematol 1990; 75(4): 549-56. RGD and integrins. Science 1987; 238(4826): 491-7. [133] Mosher DF. Cross-linking of cold-insoluble globulin by fibrin- [111] Bowditch RD, Halloran CE, Aota S, Obara M, Plow EF, Yamada stabilizing factor. J Biol Chem 1975; 250(16): 6614-21. KM, et al. Integrin alpha IIb beta 3 (platelet GPIIb-IIIa) recognizes [134] Cho J, Mosher DF. Enhancement of thrombogenesis by plasma multiple sites in fibronectin. J Biol Chem 1991; 266(34): 23323-8. fibronectin cross-linked to fibrin and assembled in platelet thrombi. [112] Beumer S, Ijsseldijk MJ, de Groot PG, Sixma JJ. Platelet adhesion Blood 2006; 107(9): 3555-63. to fibronectin in flow: dependence on surface concentration and [135] Olorundare OE, Peyruchaud O, Albrecht RM, Mosher DF. Assem- shear rate, role of platelet membrane glycoproteins GP IIb/IIIa and bly of a fibronectin matrix by adherent platelets stimulated by lys- VLA-5, and inhibition by heparin. Blood 1994; 84(11): 3724-33. ophosphatidic acid and other agonists. Blood 2001; 98(1): 117-24. [113] Piotrowicz RS, Orchekowski RP, Nugent DJ, Yamada KY, Kunicki [136] Hynes RO, Ali IU, Destree AT, Mautner V, Perkins ME, Senger TJ. Glycoprotein Ic-IIa functions as an activation-independent fi- DR, et al. A large glycoprotein lost from the surfaces of trans- bronectin receptor on human platelets. J Cell Biol 1988; 106(4): formed cells. Ann N Y Acad Sci 1978; 312: 317-42. 1359-64. [137] Cho J, Mosher DF. Role of fibronectin assembly in platelet throm- [114] Lahav J, Hynes RO. Involvement of fibronectin, Von Willebrand bus formation. J Thromb Haemost 2006; 4(7): 1461-9. factor, and fibrinogen in platelet interaction with solid substrata. J [138] Song KS, Kim HK, Shim W, Jee SH. Plasma fibronectin levels in Supramol Struct Cell Biochem 1981; 17(4): 299-311. ischemic heart disease. Atherosclerosis 2001; 154(2): 449-53. [115] Grinnell F, Phan TV. Platelet attachment and spreading on polysty- [139] Orem C, Durmus I, Kilinc K, Baykan M, Gokce M, Orem A, et al. rene surfaces: dependence on fibronectin and plasma concentration. Plasma fibronectin level and its association with coronary artery Thromb Res 1985; 39(2): 165-71. disease and carotid intima-media thickness. Coron Artery Dis [116] Grunkemeier JM, Tsai WB, McFarland CD, Horbett TA. The effect 2003; 14(3): 219-24. of adsorbed fibrinogen, fibronectin, von Willebrand factor and vi- [140] Reheman A, Gross P, Yang H, Chen P, Allen D, Leytin V, et al. tronectin on the procoagulant state of adherent platelets. Biomateri- Vitronectin stabilizes thrombi and vessel occlusion but plays a dual als 2000; 21(22): 2243-52. role in platelet aggregation. J Thromb Haemost 2005; 3(5): 875-83. [117] Wonerow P, Pearce AC, Vaux DJ, Watson SP. A critical role for [141] Eitzman DT, Westrick RJ, Nabel EG, Ginsburg D. Plasminogen phospholipase Cgamma2 in alphaIIbbeta3-mediated platelet activator inhibitor-1 and vitronectin promote vascular thrombosis spreading. J Biol Chem 2003; 278(39): 37520-9. in mice. Blood 2000; 95(2): 577-80. [118] Houdijk WP, Sixma JJ. Fibronectin in artery subendothelium is [142] Koschnick S, Konstantinides S, Schafer K, Crain K, Loskutoff DJ. important for platelet adhesion. Blood 1985; 65(3): 598-604. Thrombotic phenotype of mice with a combined deficiency in [119] Schvartz I, Seger D, Shaltiel S. Vitronectin. Int J Biochem Cell plasminogen activator inhibitor 1 and vitronectin. J Thromb Hae- Biol 1999; 31(5): 539-44. most 2005; 3(10): 2290-5. [120] Stockmann A, Hess S, Declerck P, Timpl R, Preissner KT. Mul- [143] Seiffert D, Mimuro J, Schleef RR, Loskutoff DJ. Interactions be- timeric vitronectin. Identification and characterization of conforma- tween type 1 plasminogen activator inhibitor, extracellular matrix tion-dependent self-association of the adhesive protein. J Biol and vitronectin. Cell Differ Dev 1990; 32(3): 287-92. Chem 1993; 268(30): 22874-82. [144] Declerck PJ, De MM, Alessi MC, Baudner S, Paques EP, Preissner [121] Seiffert D, Schleef RR. Two functionally distinct pools of vi- KT, et al. Purification and characterization of a plasminogen acti- tronectin (Vn) in the blood circulation: identification of a heparin- vator inhibitor 1 binding protein from human plasma. Identification Platelet Activation by Extracellular Matrix Proteins Current Pharmaceutical Design, 2009, Vol. 15, No. 12 1371

as a multimeric form of S protein (vitronectin). J Biol Chem 1988; [165] Herdeg C, Goehring-Frischholz K, Zuern C, Geisler T, Hartmann 263(30): 15454-61. U, Hoevelborn T, et al. Local catheter-based delivery of antithrom- [145] Lindahl TL, Sigurdardottir O, Wiman B. Stability of plasminogen botic or antiproliferative drugs: A new concept for prevention of activator inhibitor 1 (PAI-1). Thromb Haemost 1989; 62(2): 748- restenosis. Thromb Res 2008. 51. [166] Combe S, Rosport K, Kennedy.A. PR-15 A NOVEL INTRAVE- [146] Erickson LA, Hekman CM, Loskutoff DJ. The primary plasmino- NOUS INHIBITOR OF PLATELET ADHESION: A PHASE I gen-activator inhibitors in endothelial cells, platelets, serum, and DOSE ASCENDING PHARMACOKINETIC, PHARMACODY- plasma are immunologically related. Proc Natl Acad Sci USA NAMIC, AND TOLERANCE STUDY. J Thromb Haemost 1985; 82(24): 8710-4. 5[Supplement 2]. 2008. [147] Fay WP, Eitzman DT, Shapiro AD, Madison EL, Ginsburg D. [167] Horii K, Kahn ML, Herr AB. Structural basis for platelet collagen Platelets inhibit fibrinolysis in vitro by both plasminogen activator responses by the immune-type receptor glycoprotein VI. Blood inhibitor-1-dependent and -independent mechanisms. Blood 1994; 2006; 108(3): 936-42. 83(2): 351-6. [168] Nieswandt B, Schulte V, Bergmeier W, Mokhtari-Nejad R, Racke- [148] May AE, Seizer P, Gawaz M. Platelets: inflammatory firebugs of brandt K, Cazenave JP, et al. Long-term antithrombotic protection vascular walls. Arterioscler Thromb Vasc Biol 2008; 28(3): s5-10. by in vivo depletion of platelet glycoprotein VI in mice. J Exp Med [149] Collaborative meta-analysis of randomised trials of antiplatelet 2001; 193(4): 459-69. therapy for prevention of death, myocardial infarction, and stroke [169] Sugiyama T, Okuma M, Ushikubi F, Sensaki S, Kanaji K, Uchino in high risk patients. BMJ 2002; 324(7329): 71-86. H. A novel platelet aggregating factor found in a patient with de- [150] Mehta SR, Yusuf S. Short- and long-term oral antiplatelet therapy fective collagen-induced platelet aggregation and autoimmune in acute coronary syndromes and percutaneous coronary interven- thrombocytopenia. Blood 1987; 69(6): 1712-20. tion. J Am Coll Cardiol 2003; 41(4 Suppl S): 79S-88S. [170] Rabie T, Varga-Szabo D, Bender M, Pozgaj R, Lanza F, Saito T, et [151] A randomised, blinded, trial of clopidogrel versus aspirin in pa- al. Diverging signaling events control the pathway of GPVI down- tients at risk of ischaemic events (CAPRIE). CAPRIE Steering regulation in vivo. Blood 2007; 110(2): 529-35. Committee. Lancet 1996; 348(9038): 1329-39. [171] Takayama H, Hosaka Y, Nakayama K, Shirakawa K, Naitoh K, [152] Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK. Matsusue T, et al. A novel antiplatelet antibody therapy that in- Effects of clopidogrel in addition to aspirin in patients with acute duces cAMP-dependent endocytosis of the GPVI/Fc receptor coronary syndromes without ST-segment elevation. N Engl J Med gamma-chain complex. J Clin Invest 2008; 118(5): 1785-95. 2001; 345(7): 494-502. [172] Matsumoto Y, Takizawa H, Gong X, Le S, Lockyer S, Okuyama [153] Jemal A, Ward E, Hao Y, Thun M. Trends in the leading causes of K, et al. Highly potent anti-human GPVI monoclonal antibodies death in the United States, 1970-2002. JAMA 2005; 294(10): 1255- derived from GPVI knockout mouse immunization. Thromb Res 9. 2007; 119(3): 319-29. [154] Mathers CD, Loncar D. Projections of global mortality and burden [173] Lecut C, Feeney LA, Kingsbury G, Hopkins J, Lanza F, Gachet C, of disease from 2002 to 2030. PLoS Med 2006; 3(11): e442. et al. Human platelet glycoprotein VI function is antagonized by [155] Barnes CS, Krafft B, Frech M, Hofmann UR, Papendieck A, monoclonal antibody-derived Fab fragments. J Thromb Haemost Dahlems U, et al. Production and characterization of saratin, an in- 2003; 1(12): 2653-62. hibitor of von Willebrand factor-dependent platelet adhesion to col- [174] Moroi M, Mizuguchi J, Kawashima S, Nagamatsu M, Miura Y, lagen. Semin Thromb Hemost 2001; 27(4): 337-48. Nakagaki T, et al. A new monoclonal antibody, mAb 204-11, that [156] Calvo E, Tokumasu F, Marinotti O, Villeval JL, Ribeiro JM, Fran- influences the binding of platelet GPVI to fibrous collagen. cischetti IM. Aegyptin, a novel mosquito salivary gland protein, Thromb Haemost 2003; 89(6): 996-1003. specifically binds to collagen and prevents its interaction with [175] Li H, Lockyer S, Concepcion A, Gong X, Takizawa H, Guertin M, platelet glycoprotein VI, integrin alpha2beta1, and von Willebrand et al. The Fab fragment of a novel anti-GPVI monoclonal antibody, factor. J Biol Chem 2007; 282(37): 26928-38. OM4, reduces in vivo thrombosis without bleeding risk in rats. Ar- [157] Lasser G, Guchhait P, Ellsworth JL, Sheppard P, Lewis K, Bishop terioscler Thromb Vasc Biol 2007; 27(5): 1199-205. P, et al. C1qTNF-related protein-1 (CTRP-1): a vascular wall pro- [176] Eble JA, Beermann B, Hinz HJ, Schmidt-Hederich A. alpha 2beta 1 tein that inhibits collagen-induced platelet aggregation by blocking integrin is not recognized by rhodocytin but is the specific, high af- VWF binding to collagen. Blood 2006; 107(2): 423-30. finity target of rhodocetin, an RGD-independent disintegrin and po- [158] van Zanten GH, Connolly TM, Schiphorst ME, de GS, Slootweg tent inhibitor of cell adhesion to collagen. J Biol Chem 2001; PJ, Sixma JJ. Recombinant leech antiplatelet protein specifically 276(15): 12274-84. blocks platelet deposition on collagen surfaces under flow condi- [177] Funahashi Y, Sugi NH, Semba T, Yamamoto Y, Hamaoka S, Tsu- tions. Arterioscler Thromb Vasc Biol 1995; 15(9): 1424-31. kahara-Tamai N, et al. Sulfonamide derivative, E7820, is a unique [159] White TC, Berny MA, Robinson DK, Yin H, DeGrado WF, Han- angiogenesis inhibitor suppressing an expression of integrin alpha2 son SR, et al. The leech product saratin is a potent inhibitor of subunit on endothelium. Cancer Res 2002; 62(21): 6116-23. platelet integrin alpha2beta1 and von Willebrand factor binding to [178] Choi S, Vilaire G, Marcinkiewicz C, Winkler JD, Bennett JS, De- collagen. FEBS J 2007; 274(6): 1481-91. Grado WF. Small molecule inhibitors of integrin alpha2beta1. J [160] Gronwald W, Bomke J, Maurer T, Domogalla B, Huber F, Schu- Med Chem 2007; 50(22): 5457-62. mann F, et al. Structure of the leech protein saratin and characteri- [179] Kageyama S, Matsushita J, Yamamoto H. Effect of a humanized zation of its binding to collagen. J Mol Biol 2008; 381(4): 913-27. monoclonal antibody to von Willebrand factor in a canine model of [161] Miura Y, Takahashi T, Jung SM, Moroi M. Analysis of the interac- coronary arterial thrombosis. Eur J Pharmacol 2002; 443(1-3): 143- tion of platelet collagen receptor glycoprotein VI (GPVI) with col- 9. lagen. A dimeric form of GPVI, but not the monomeric form, [180] Kageyama S, Yamamoto H, Nakazawa H, Matsushita J, Kouyama shows affinity to fibrous collagen. J Biol Chem 2002; 277(48): T, Gonsho A, et al. Pharmacokinetics and pharmacodynamics of 46197-204. AJW200, a humanized monoclonal antibody to von Willebrand [162] Massberg S, Konrad I, Bultmann A, Schulz C, Munch G, Peluso factor, in monkeys. Arterioscler Thromb Vasc Biol 2002; 22(1): M, et al. Soluble glycoprotein VI dimer inhibits platelet adhesion 187-92. and aggregation to the injured vessel wall in vivo. FASEB J 2004; [181] Keefe AD, Schaub RG. Aptamers as candidate therapeutics for 18(2): 397-9. cardiovascular indications. Curr Opin Pharmacol 2008; 8(2): 147- [163] Gruner S, Prostredna M, Koch M, Miura Y, Schulte V, Jung SM, et 52. al. Relative antithrombotic effect of soluble GPVI dimer compared [182] Roovers RC, van Dongen GA, en Henegouwen PM. Nanobodies in with anti-GPVI antibodies in mice. Blood 2005; 105(4): 1492-9. therapeutic applications. Curr Opin Mol Ther 2007; 9(4): 327-35. [164] Bultmann A, Herdeg C, Li Z, Munch G, Baumgartner C, Langer H, [183] Gilbert JC, Feo-Fraulini T, Hutabarat RM, Horvath CJ, Merlino et al. Local delivery of soluble platelet collagen receptor glycopro- PG, Marsh HN, et al. First-in-human evaluation of anti von Wille- tein VI inhibits thrombus formation in vivo. Thromb Haemost brand factor therapeutic aptamer ARC1779 in healthy volunteers. 2006; 95(5): 763-6. Circulation 2007; 116(23): 2678-86. 1372 Current Pharmaceutical Design, 2009, Vol. 15, No. 12 Steve P. Watson

[184] Silence K, Jonckheere H, Casteels P, Roodt J, Meiring M, Lamp- [187] Ware J, Russell S, Ruggeri ZM. Generation and rescue of a murine recht S, et al. ALX-081 Nanobody, an Engineered Bivalent Anti- model of platelet dysfunction: the Bernard-Soulier syndrome. Proc Thrombotic Drug Candidate with Improved Efficacy and Safety as Natl Acad Sci USA 2000; 97(6): 2803-8. Compared to the Marketed Drugs. Blood 108[11]. 2006. [188] Bergmeier W, Piffath CL, Goerge T, Cifuni SM, Ruggeri ZM, [185] Clemetson KJ. A short history of platelet glycoprotein Ib complex. Ware J, et al. The role of platelet adhesion receptor GPIbalpha far Thromb Haemost 2007; 98(1): 63-8. exceeds that of its main ligand, von Willebrand factor, in arterial [186] Wu D, Vanhoorelbeke K, Cauwenberghs N, Meiring M, Depraetere thrombosis. Proc Natl Acad Sci USA 2006; 103(45): 16900-5. H, Kotze HF, et al. Inhibition of the von Willebrand (VWF)- [189] Clemetson KJ, Clemetson JM. Platelet GPIb complex as a target collagen interaction by an antihuman VWF monoclonal antibody for anti-thrombotic drug development. Thromb Haemost 2008; results in abolition of in vivo arterial platelet thrombus formation in 99(3): 473-9. baboons. Blood 2002; 99(10): 3623-8. [190] Vanhoorelbeke K, Ulrichts H, Van de WG, Fontayne A, Deckmyn H. Inhibition of platelet glycoprotein Ib and its antithrombotic po- tential. Curr Pharm Des 2007; 13(26): 2684-97.