throbber
1522
`
`Current Perspectives
`
`Direct Thrombin Inhibitors
`in Cardiovascular Medicine
`
`Jeffrey Lefkovits, MBBS; Eric J. Topol, MD
`
`Currently used antithrombotics such as heparin
`Abstract
`have a number of potential limitations that may be overcome
`by the new class of agents that directly inhibit thrombin. These
`agents variously block the active catalytic and/or the anion
`binding exosites of the thrombin molecule and are potent and
`specific inhibitors of thrombin's many biological actions, as
`demonstrated by in vitro and animal models of thrombosis.
`Preliminary data indicate that the direct antithrombins are
`safe and efficacious in humans, and their use in acute coronary
`
`syndromes and coronary angioplasty in place of heparin has
`yielded promising early results. Phase III trials in these clinical
`settings are currently under way. Newer antithrombotics that
`inhibit thrombin generation and thrombin activity at various
`strategic points within the coagulation cascade are also in the
`early stages of development. (Circulation. 1994;90:1522-1536.)
`Key Words * antithrombotics * thrombosis *
`coagulation
`current perspectives
`
`*
`
`T he introduction of the direct thrombin inhibi-
`tors - a new class of agents that specifically and
`potently antagonize the actions of thrombin -
`has opened up new and exciting opportunities in the
`regulation of the thrombotic process, which is so impor-
`tant in the field of cardiovascular medicine. It is now
`firmly established that coronary artery thrombosis is
`causal in the development of acute coronary syn-
`dromes.12 Thrombin in turn plays a pivotal role in both
`the platelet activation and the fibrin generation inher-
`ent in this process and offers an ideal target for modu-
`lation of the clinical course of these syndromes. Hepa-
`rin, currently the mainstay of antithrombotic therapy,
`has a number of limitations that may be overcome by
`specific characteristics of the newer thrombin inhibitors.
`This review will focus on these agents, from their
`biochemistry through to the current status of their
`clinical use in the cardiological fields of acute coronary
`syndromes and coronary angioplasty. Their use has
`been evaluated in several other areas, including dissem-
`inated intravascular coagulation,3 hemodialysis,4 micro-
`surgery,5 and deep venous thrombosis,6 but will not be
`covered in this review.
`Limitations of Current Antithrombotics
`The principal antithrombotic action of heparin de-
`pends completely on the presence of cofactors. Inacti-
`vation of thrombin; activated factor X (factor Xa);
`factors XII, XI, and IX; and tissue factor Vlla complex7
`is achieved through the activation and modulation of
`antithrombin 111.8 Heparin may also act by potentiating
`heparin cofactor II-mediated inactivation of thrombin,
`especially at higher doses.9 The therapeutic use of
`
`Received February 23, 1994; revision accepted May 18, 1994.
`From the Department of Cardiology, Center for Thrombosis
`and Vascular Biology, The Cleveland Clinic Foundation, Cleve-
`land, Ohio.
`Correspondence to Eric J. Topol, MD, Department of Cardiol-
`ogy, One Clinic Center, Cleveland Clinic Foundation, Cleveland,
`OH 44195.
`© 1994 American Heart Association, Inc.
`
`heparin mandates intensive and meticulous laboratory
`monitoring of its anticoagulant effect because of the
`marked individual variation in its anticoagulant re-
`sponse. Heparin can be inactivated by platelet factor 4
`and heparinase, both of which are released by activated
`platelets,10 and fibrin monomers have been found to
`protect thrombin from inactivation by the heparin-
`antithrombin III complex in thrombogenic states."
`Binding to vitronectin, fibronectin, and other plasma
`proteins limits the amount of heparin available and can
`decrease its anticoagulant effect,12 whereas heparin
`therapy is ineffective in people with hereditary anti-
`thrombin III deficiency. Most important, however, is the
`inability of the heparin-antithrombin III complex to
`inactivate thrombin already bound to clot.1"12 This is
`most likely due to a conformational change in the
`thrombin molecule induced by its binding to fibrin,
`which, in turn, prevents attachment of the heparin-
`antithrombin III complex.13 The "protected status" of
`clot-bound thrombin is a major potential drawback to
`heparin use, as this thrombin can act as an ongoing
`source of thrombogenesis at sites of pathological throm-
`bus formation, theoretically limiting heparin efficacy.
`In contrast, the actions of direct thrombin inhibitors
`are antithrombin III independent; they do not bind
`plasma proteins to a significant extent, are able to
`inactivate clot-bound thrombin, and prevent thrombin-
`induced platelet activation, thus offering several poten-
`tial advantages over heparin. Their mechanisms of
`action and likely therapeutic benefit in cardiovascular
`disorders may be better understood by reviewing the
`biology of thrombin itself.
`Biology of Thrombin
`Thrombin, the key regulator of the thrombotic pro-
`cess, is a glycosylated, trypsinlike serine protease, with
`lysine- or arginine-directed specificity.14 It is generated
`from prothrombin by the prothrombinase complex-
`factors Xa, Va, calcium, and phospholipids, with factor
`Xa cleaving the prothrombin molecule to form the A
`
`
`
`http://circ.ahajournals.org/Downloaded from
`
` by guest on October 7, 2015
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2007 - 1/16
`
`

`
`Lefkovits and Topol
`
`Direct Thrombin Inhibitors
`
`1523
`
`Simplified overview of the coagulation cas-
`FIG 1.
`cade, illustrating the central roles of factor Xa and
`thrombin in the coagulation process. The sites of
`action of the various antithrombotic agents are also
`shown. ATIII indicates antithrombin 111; LMWH, low
`molecular weight heparins; PC, protein C; APC, (re-
`combinant) activated protein C; TFPI, tissue factor
`pathway inhibitor; TRAP, thrombin receptor antagonist
`peptide; TAP, tick anticoagulant peptide; and TM,
`thrombomodulin.
`
`--
`
`'bxlla
`
`FR
`
`Protein S /
`
`Acved Protein C
`
`Inactvion Va, Villa
`
`and B chains of thrombin, which combine to form
`a-thrombin.
`Thrombin has many and varied biologic functions, but
`its main action is to catalyze the transformation of
`fibrinogen to fibrin, whether the thrombin is soluble in
`plasma or fibrin bound. Thrombin further activates
`factor XIII to cross-link fibrin and stabilize the clot, and
`promotes and amplifies clot formation by activating
`other clotting plasma proteins, including factors V and
`VIII. Of importance, it also is one of the most potent
`agonists for platelet recruitment and aggregation, which
`further reinforces the newly formed clot15 (Fig 1).
`Thrombin also initiates a series of counterregulatory
`reactions to trigger endogenous anticoagulant systems
`to maintain homeostasis. Through its binding to throm-
`bomodulin, thrombin activates protein C, which joins
`with protein S to inactivate factors Va and VIIIa (Fig 1).
`It also stimulates the release of both tissue-type plas-
`minogen activator (TPA) and plasminogen activator
`inhibitor type 1, its natural antagonist, to further regu-
`late endogenous thrombolysis.15
`There are complex interactions between the endothe-
`lium and thrombin, with thrombin promoting vasodila-
`tation where the endothelium is intact, through release
`of prostacyclin and nitric oxide, but causing vasocon-
`striction where endothelium is damaged, through liber-
`ation of endothelin.16 Thrombin also acts as an effector
`molecule, occupying receptors on the endothelium and
`smooth muscle cells to activate cellular proliferation,17
`as well as stimulate the release of platelet-derived
`growth factor (PDGF).18 Leukocyte and monocyte
`chemotaxis19 and release of interleukin-1 from macro-
`phages are also induced by thrombin.20
`The unique structure of thrombin is believed to
`account for its high specificity for its substrates. The
`location of the catalytic binding site in a deep narrow
`canyon on the thrombin molecule surface appears to
`restrict access to other macromolecules by steric hin-
`drance.21 Thrombin, distinct from most other serine
`
`proteases, has an anion-binding exosite, also known as
`the substrate recognition site, that is separate but
`adjacent to the active catalytic site.21 Fibrinogen binds
`at this site, increasing thrombin's specificity for it.
`Thrombomodulin and hirudin also attach here, whereas
`heparin appears to have its own basic binding site.2'
`Direct thrombin inhibitors known to block the anion-
`binding site have been shown to inactivate thrombin
`without displacing the thrombin molecule from the
`fibrin surface, providing evidence that thrombin has a
`further and separate binding site for fibrin distinct from
`its fibrinogen binding site.12
`The apolar binding site is located in a hydrophobic
`pocket close to the catalytic site in the fibrinopeptide
`groove22 and is also involved in substrate binding to the
`active catalytic site, as well as thrombin attachment to
`the platelet receptor glycoprotein lb. The main platelet
`receptor for thrombin, however, was recently character-
`ized by Coughlin and colleagues23 and has the unique
`feature of a tethered ligand. After thrombin binds the
`receptor through a hirudin-like anion-binding exosite, it
`cleaves the receptor to reveal a new receptor's amino
`acid terminus, which functions as the tethered ligand to
`activate the receptor. The molecule of thrombin thus
`remains free to activate other receptor sites and prop-
`agate the thrombotic process.23
`The regulation of thrombin is complex, involving
`proteinase inhibitors antithrombin III, heparin cofactor
`II, and a2-macroglobulin. Thrombin generation is con-
`trolled through coagulation amplification via factors
`including V and VIII; interaction with thrombomodulin,
`protein C, and protein S; as well as partitioning in the
`clot itself.24
`
`Direct Thrombin Inhibitors
`
`History
`The therapeutic use of the leech (Hirudo medicinalis)
`probably dates back to the ancient Greeks,25 but it was
`Haycraft who first described the antithrombotic prop-
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on October 7, 2015
`
`ANACOR EX. 2007 - 2/16
`
`

`
`1524
`
`Circulation
`
`Vol 90, No 3
`
`September 1994
`
`<}Cablyfc
`
`|
`
`Thrombin
`
`Thrombl n
`
`pro
`AN
`Unker
`
`Subtrate
`Recogntlon Site
`
`Thrombin
`
`Thrombn
`
`PPACK
`PA
`Argatban
`
`Schematic of the thrombin molecule, illustrating its key binding sites. Thrombin interactions with the direct thrombin inhibitors
`FIG 2.
`hirudin, hirulog, hirugen, PPACK, and argatroban are illustrated.
`
`erty of leech saliva in 1884.26 Hirudin was isolated by
`Markwardt in the 1950s,27 and the structure has subse-
`quently been characterized and cloned.28 Further devel-
`opment based on the structure of hirudin has led to the
`formation of a family of hirudin-like peptides called
`hirugens or hirullins.29 Peptide analogues hirugen and
`hirulog are based on the structure of hirudin, and
`synthetic derivatives argatroban, D-phenylalanyl-L-pro-
`lyl-L-arginyl chloromethylketone (PPACK), and the
`boroarginine derivatives have subsequently been devel-
`oped (Fig 2).
`Structure and Function
`Hirudin
`The prototypic thrombin inhibitor is the naturally
`occurring hirudin, isolated from the leech saliva, and is
`the most potent and specific known inhibitor of throm-
`bin, forming a tight, highly stable noncovalent complex
`with thrombin. It is a 65-amino-acid protein with three
`disulfide bridges and a molecular weight of -7000 d.
`Multiple isoforms of natural hirudin exist, with slightly
`different numbers of amino acids but similar anticoag-
`ulant activities. All have the disulfide bridges and a
`sulfated tyrosine residue in position 63.29
`Recombinant DNA technology has allowed the de-
`velopment of recombinant hirudin (r-hirudin), which
`has an identical amino acid sequence to natural hirudin,
`with or without the sulfated Tyr63.30 The nonsulfated
`molecule results in about a 10-fold reduction in throm-
`bin affinity,31 although binding still remains strong.
`Several companies have now developed proprietary
`versions of r-hirudin, which are at various stages of
`clinical testing. Although direct head-to-head compari-
`sons of these different recombinant have not been
`performed, Longstaff and colleagues,32 in a study de-
`signed to investigate standardization of laboratory hiru-
`din assays, found that four proprietary formulations of
`
`r-hirudin all had similar potencies, using a simple,
`standardized chromogenic assay.
`The hirudin molecule has two distinct domains-an
`NH2 terminal core domain and a COOH terminal tail.27
`The N-terminal binds and inhibits the active catalytic
`site of thrombin,27 whereas the carboxy terminal simul-
`taneously blocks the anion-binding exosite33 (Fig 2).
`This interaction takes place in two steps, with an initial
`ionic interaction and then a rearrangement of the
`hirudin-thrombin complex to form a stronger binding.34
`The apolar-binding site also appears to be involved in
`hirudin binding, since the compound proflavin (which
`binds to the apolar-binding site) is displaced during
`formation of the hirudin-thrombin complex.22 Crystal
`structure studies have further demonstrated that the
`attachment of hirudin to thrombin is not limited to the
`three binding sites but also forms multiple other con-
`tacts, tightly binding thrombin over an extended area of
`the molecule.3536 Overall, this allows hirudin to be
`uniquely specific for thrombin, with no inhibitory action
`on any other serine protease.36
`Hirugen
`Hirugen, a synthetic hirudin derivative, is a dode-
`capeptide comprising the terminal 12 residues of hiru-
`din involved in anion-binding exosite blockade (Fig 2).
`The binding of hirugen to thrombin is similar to the
`C-terminal of hirudin,37 and the molecule contains a
`sulfated tyrosine to increase its thrombin afiinity. Hir-
`ugen was designed to block the interaction of thrombin
`with fibrinogen, leaving the catalytic site of thrombin
`free to interact with antithrombin III, enhancing its
`actions, with or without heparin.38 However, the in vivo
`antithrombotic activity of hirugen is considerably
`weaker compared with hirudin and hirulog,39 and it has
`not progressed to testing in the clinical arena.
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on October 7, 2015
`
`ANACOR EX. 2007 - 3/16
`
`

`
`Hirulog
`A group of peptides called hirulogs were subse-
`quently developed that had the D-Phe-Pro-Arg se-
`quence of the N-terminal of hirudin linked to the
`hirugen molecule by a segment of glycyl residues varying
`from 6 to 18 A in length.40 Like hirudin, this synthetic
`derivative is able to block both the active catalytic site
`and anion-binding site of the thrombin molecule (Fig 2).
`Hirulog-1 has four glycyl residues between the two
`domains and has been found to be a potent thrombin
`inhibitor.40 Although hirulog retains the high specificity
`of hirudin for thrombin, its substrate-type binding to
`thrombin is more typical of other protease inhibitors, in
`contrast to the unique multiple contact binding of
`hirudin.36 However, doubt has arisen regarding the
`contribution of active site inhibition to hirulog's action,
`following the discovery that thrombin itself mediates
`slow cleavage of hirulog at the Arg-Pro bond on the
`amino-terminal extension in vivo,41 effectively convert-
`ing it into a hirugen-like molecule. This may account for
`some of the pharmacological differences with hirudin40
`and has led to the development of hirudin derivatives
`with noncleavable bonds.42
`PPACK and Argatroban
`PPACK is a tripeptide synthetic compound that has a
`structure very close to fibrinopeptide A.43 It contains
`the amino acid sequence corresponding to the cleavage
`site of fibrinogen and acts as an affinity agent to
`thrombin.44 PPACK blocks the active catalytic site by
`alkylating the active site histidine residue and is an
`irreversible inhibitor of thrombin (Fig 2). The com-
`pound has been found to be an effective thrombin
`inhibitor in plasma and in vivo, but the presence of a
`reactive group leads to a relatively rapid loss of activity
`due to reactions with other plasma components.45 Ar-
`gatroban, an arginine derivative, binds thrombin at the
`apolar-binding site adjacent to the active site. Its main
`action is to block thrombin's active catalytic site as does
`PPACK, but unlike this compound, argatroban is a
`competitive antagonist. Comparative studies with other
`thrombin inhibitors have shown this compound to be a
`potent thrombin inhibitor with strong affinity for
`thrombin.44,46
`Other Thrombin Inhibitors
`Many other compounds have been developed that
`have antithrombin activity, but are too toxic for clinical
`use (Table 1). Thrombin inhibition by arginine and
`benzamidine derivatives ranges from weak to strong,44"45
`whereas the boroarginine derivatives such as DuP 714
`are potent antithrombins and have the potential for oral
`bioavailability.47 However, to date use has been limited
`by adverse liver toxicity, believed to be related to the
`boron constituent.
`Several other classes of antithrombins, at various
`stages of development, are aimed at controlling the
`thrombotic process at different strategic points or
`through novel methods. The low molecular weight hep-
`arin (LMWH) preparations are fragments of standard
`heparin obtained by chemical or enzymatic depolymer-
`ization of the polysaccharide chains. Although they are
`antithrombin III dependent and cannot inhibit throm-
`bin that is fibrin bound, they are differentiated from
`
`Lefkovits and Topol
`
`Direct Thrombin Inhibitors
`
`1525
`
`Classification of Antithrombotic Agents
`TABLE 1.
`Antithrombin Ill dependent
`Heparin
`Low molecular weight heparins
`Heparinoids
`Direct thrombin inhibitors
`Hirudin
`Hirugen
`Hirulog
`Argatroban
`PPACK
`DuP 714
`Thrombin aptamers
`Thrombin generation inhibitors
`Factor Xa inhibitors
`Antistasin
`Tick anticoagulant peptide
`Inactivated factor X
`Factor VIl antibody and peptidomimetics
`Tissue factor pathway inhibitor
`Recombinant endogenous anticoagulants
`Activated protein C
`Antithrombin Ill
`Heparin cofactor 11
`Tissue factor pathway inhibitor
`Thrombin receptor blockers
`Thrombin receptor antagonist peptides
`Vitamin K antagonists
`Warfarin sodium
`
`heparin by both their relative and absolute increased
`affinity for factor Xa over thrombin,48 with the degree of
`selectivity for factor Xa varying with different commer-
`cial preparations. The LMWH compounds have lower
`affinities for endothelial cells and plasma proteins than
`standard heparin and are therefore cleared more slowly,
`allowing once-daily dosing and more predictable anti-
`coagulant responses.48 Several trials have now demon-
`strated their clinical use, particularly in the areas of
`deep venous thrombosis prophylaxis and treatment.49
`A novel mechanism for control of the thrombotic
`process has been developed using single-stranded DNA
`oligonucleotides known as aptamers to bind target
`proteins and block their actions. Thrombin aptamers,
`first described by Bock and colleagues,50 bind the fibrin-
`ogen recognition site on the thrombin molecule51 and
`have demonstrated potent antithrombotic effects in
`vitro,50 in animal models of thrombosis,5253 and also
`have the ability to inhibit clot-bound thrombin.53 The 15
`nucleotide aptamer used in a number of studies has the
`distinct characteristics of rapid onset of action and short
`half-life, which may favor its future use in certain acute
`clinical settings.52'53
`Alternative strategies for controlling thrombin activ-
`ity are becoming available through the isolation and
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on October 7, 2015
`
`ANACOR EX. 2007 - 4/16
`
`

`
`1526
`
`Circulation
`
`Vol 90, No 3
`
`September 1994
`
`development of agents that selectively inhibit key en-
`zymes at different points within the coagulation cas-
`cade. From a theoretical standpoint, because the direct
`thrombin inhibitors antagonize thrombin that is pre-
`formed, some thrombin activity may still occur before its
`neutralization. New formation of thrombin remains
`unaffected and may also contribute to a "leakage" of
`thrombin activity despite the presence of an inhibitor.
`Conversely, inhibition of activated factor X (factor Xa)
`can prevent new formation of thrombin and disrupt the
`thrombin feedback loop that autoamplifies thrombin
`generation but has no effect on thrombin that is already
`present. Inhibition of proteins earlier in the pathway,
`such as tissue factor - a key initiator of thrombosis in
`vivo - offers a more mechanistically specific targeting
`for prevention of thrombosis but may have limited
`efficacy because of compensatory augmentation in ac-
`tivity of alternate coagulation pathways such as the
`intrinsic pathway.
`With these theoretical considerations in mind, a
`number of thrombin generation inhibitors have under-
`gone early experimental testing. Local inhibition of
`factor Xa has been achieved by antistasin, a 119-amino-
`acid protein initially isolated from the salivary gland of
`the Mexican leech,54 and tick anticoagulant peptide
`(TAP) derived from the soft tick,55 now produced by
`recombinant technology. Both are potent and highly
`selective antagonists of factor Xa,55,56 and unlike anti-
`thrombin III, can inhibit factor Xa attached to the
`prothrombinase complex as well as free factor Xa.56
`When used as adjuncts to thrombolysis, these agents,
`through the inhibition of thrombin generation, acceler-
`ated clot lysis and prevented reocclusion.5758 This has
`brought into focus the controversial concept that, at
`least in part, it is the continuous generation of new
`thrombin rather than reexposure of preformed clot-
`bound thrombin that is responsible for the phenomenon
`of reocclusion. Indeed, markers of thrombin generation
`such as prothrombin fragment F1.2 have been found to
`increase during and after thrombolytic treatment for
`acute myocardial infarction, suggesting that increased
`thrombin activity associated with thrombolysis is, at
`least in part, due to new thrombin generation.59 This in
`turn may have important implications for the eventual
`place of direct thrombin inhibitors as adjuncts to throm-
`bolysis, as lack of inhibition of new thrombin generation
`represents a potential mechanistic limitation of these
`agents.
`Recombinant forms of naturally occurring endoge-
`nous anticoagulants is yet another strategy that has
`been exploited to control the thrombotic process. Tissue
`factor pathway inhibitor (TFPI) is a multivalent protein-
`ase inhibitor, whose complex actions involve the binding
`and inhibition of factor Xa, producing a factor Xa-TFPI
`complex that causes feedback inhibition on the factor
`VIIa-tissue factor complex responsible for triggering
`factor X activation.6061 Purified and recombinant acti-
`vated Protein C, through their inhibition of factors Va
`and VIIIa, inhibit thrombin formation62 and have also
`been found to inactivate plasminogen activator inhibi-
`tor.63 In vitro and animal studies have demonstrated
`antithrombotic effects of this agent,63,64 and human
`evaluation in phase I trials has already commenced.65 A
`potential use for this compound is as an adjunct to
`thrombolysis, particularly following the discovery that
`
`thrombolytic therapy itself appears to generate acti-
`vated protein C.66 Novel approaches such as monoclonal
`antibodies against or peptidomimetics of factor VII and
`antibodies against the recently characterized platelet
`thrombin receptor are all in the very early stages of
`development.
`Experimental Studies With Direct
`Thrombin Inhibitors
`As expected from their mode of action, direct throm-
`bin inhibitors antagonize the broad spectrum of throm-
`bin's actions. Hirudin has been shown to inhibit fibrin
`formation; prevent activation of factors V, VIII, and
`XIII; and prevent thrombin-mediated platelet activa-
`tion in vitro.67,68 It produces virtual total inhibition of
`thrombus formation ex vivo in both rat and human
`plasma30 and may also augment displacement of factor
`Xa from vascular endothelium, actually associating it-
`self with factor Xa.69 Animal studies have confirmed the
`antithrombotic efficacy of hirudin in a wide range of
`animal venous and arterial models of thrombo-
`sisj27,30,70-73 Hirulog,39,74 argatroban,75,76 and PPACK77
`have also proven to be effective antithrombotics in
`various animal models. Hirudin appears to have addi-
`tional effects on thrombus composed predominantly of
`platelets rather than fibrin,78 although in a rat dissemi-
`nated intravascular coagulation model higher concen-
`trations of hirudin were required to inhibit platelet
`deposition than to prevent fibrin deposition.79
`Direct thrombin inhibitors may have an especially
`important adjunctive role in thrombolysis. Apart from
`their main action to activate plasmin and lyse fibrin,
`thrombolytics also trigger a number of reactions that
`engender a paradoxically "prothrombotic state," includ-
`ing plasmin-mediated activation of factor XII and plate-
`let activation,80 platelet release of plasminogen activator
`inhibitor,81 and consumption of plasma plasminogen,
`which in turn draws clot-bound plasminogen into the
`soluble phase, reducing the substrate for activation and
`attenuating fibrinolytic efficacy (plasminogen steal).82
`Of chief significance, however, is that lytic agents accel-
`erate exposure of thrombin bound to fibrin within the
`clot, as well as increase the amount of free throm-
`bin,83-85 producing a milieu highly conducive to re-
`thrombosis. Experimental models both in vitro and in
`vivo have demonstrated the ability of thrombin inhibi-
`tors to neutralize both clot-bound thrombin and free
`soluble thrombin.1286-88 Hirudin has been shown to
`accelerate thrombolysis following TPA,89 and prevent
`reocclusion in a canine model,90 whereas hirulog accel-
`erated thrombolysis and prevented reocclusion in a rat
`model.74 Argatroban, an agent with less affinity for
`thrombin than either hirudin or hirulog, was able to
`accelerate clot lysis by TPA in a canine stenotic coro-
`nary artery model but required the addition of aspirin
`to reduce the incidence of reocclusion.9'
`There is further compelling experimental evidence to
`support a beneficial role for direct thrombin inhibitors
`in the field of coronary intervention. Platelet aggrega-
`tion and fibrin deposition produced in a pig model of
`carotid angioplasty have been successfully prevented by
`hirudin,78 and the same agent also inhibited restenosis
`at 28 days in a rabbit angioplasty model, suggesting an
`additional mechanism of inhibition of thrombin's effec-
`tor molecule function as a stimulator of smooth muscle
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on October 7, 2015
`
`ANACOR EX. 2007 - 5/16
`
`

`
`proliferation.92 Thrombin inhibitors effectively pre-
`vented thrombin-induced platelet activation,93,94 and
`comparative studies with heparin found hirudin to be
`more effective in reducing both platelet and fibrin
`deposition on coronary stents,95 platelet thrombi in the
`porcine deep arterial injury model,78 and more potently
`inhibited thrombus in rabbit and rat thrombosis
`models.88,96
`Clinical Pharmacology and Phase I Studies
`Relatively little data are available from comparative
`and systematic pharmacological trials, although hirudin
`remains the most extensively studied thrombin inhibitor
`pharmacologically. Recombinant hirudin is poorly ab-
`sorbed orally and is administered parenterally via the
`intravenous, intramuscular, or subcutaneous route. Pri-
`mate studies have demonstrated faster onset and earlier
`return to baseline after intravenous administration. The
`half-life after intravenous use was 40 minutes compared
`with almost 2 hours after subcutaneous injection.70 The
`volume of distribution has variously been estimated
`from 8.9 to 17.2 L97 with significant distribution into the
`extravascular space. Little hepatic metabolism takes
`place with hirudin, and 95% of the compound is renally
`excreted in its active form.70'97 Impaired elimination has
`been demonstrated in patients with renal insufficiency,97
`and dose reduction is recommended in patients with
`renal dysfunction.
`In a recent pharmacological trial with human volun-
`teers,98 hirulog differed from hirudin in that its half-life
`was 15 to 20 minutes shorter after intravenous bolus
`administration and that total body clearance rate was
`more rapid at 0.5 L/min for a 70-kg man compared with
`hirudin's clearance rate of 0.19 L/min.99 However, only
`20% of hirulog was recovered from the urine, suggesting
`that it undergoes more extensive metabolic clearance
`than hirudin, including hepatic metabolism or proteol-
`ysis at other sites.
`Several phase I trials have now been completed with
`the various thrombin inhibitors, demonstrating excel-
`lent safety and anticoagulant efficacy profiles in healthy
`volunteers. Argatroban was well tolerated in healthy
`human volunteers and produced a dose-dependent pro-
`longation of the coagulation parameters activated par-
`tial thromboplastin time (aPTT) and thrombin time
`(TT), with a return to baseline levels 1 hour after
`intravenous administration. No prolongation of skin
`bleeding time was detected, even with concomitant
`administration of aspirin.100
`The biologic effects of sulfated r-hirudin (CGP-39393)
`were comprehensively studied in a recent report by
`Verstraete and colleagues.10' Hirudin was administered
`subcutaneously, as either a single dose or repeated doses,
`in 231 healthy volunteers. The drug was extremely well
`tolerated, with no significant adverse events reported.
`Single injections of 0.1 mg/kg prolonged the aPTT to
`almost twice baseline values in 184 volunteers, and
`increasing doses up to 0.75 mg/kg increased the aPTT
`linearly. The anticoagulant effect after subcutaneous
`administration was noted within 30 minutes, peaking at 4
`to 6 hours, and returning to baseline within 24 hours.
`Similar findings have been reported elsewhere.99102"103
`Repeated subcutaneous doses lengthened aPTT for as
`long as the administration was continued, with no evi-
`dence of cumulative effects even up to 6 days of admin-
`
`Lefkovits and Topol
`
`Direct Thrombin Inhibitors
`
`1527
`
`istration. Bleeding time was not prolonged even up to
`doses of 0.5 mg/kg, suggesting little effect of hirudin on
`primary hemostasis.10'
`Tolerability and anticoagulant activity of hirulog
`were assessed in a recent randomized, placebo-con-
`trolled study of 54 healthy volunteers,98 with both
`intravenous and subcutaneous routes of administration
`studied. Doses ranged from 0.05 to 1.0 mg/kg, and a
`rapid dose-dependent prolongation of the aPTT for
`both intravenous and subcutaneous routes was demon-
`strated. There was no significant bleeding or other
`adverse events reported, and overall there was no
`prolongation of the skin bleeding time. Subcutaneous
`injection resulted in a later peaking of anticoagulant
`effect and a sustained prolongation of aPTT over sev-
`eral hours. Greater anticoagulant activity was achieved
`at lower doses of hirulog by extending the intravenous
`infusion period for 2 hours or more, after steady-state
`levels of the drug were obtained.98
`Clinical (Phase II) Studies
`Several studies have assessed the role of thrombin
`inhibitors in acute ischemic syndromes and PTCA
`(Table 2).
`Acute Myocardial Infarction
`A number of small studies published recently have
`assessed thrombin inhibitors as adjuncts to thromboly-
`sis. Lidon et al'04 randomized 42 patients to heparin or
`hirulog after streptokinase for acute myocardial infarc-
`tion. Hirulog had significantly better 90-minute Throm-
`bolysis in Myocardial Infarction (TIMI)-3 patency rates
`(61% versus 29%, P=.05) and 120-minute patency rates
`(75% versus 36%, P<.02). Bleeding complications were
`the same in the two groups. Tabata et al105 evaluated
`argatroban in the prevention of reocclusion in 24 pa-
`tients following successful reperfusion therapy for myo-
`cardial infarction. Reocclusion at 1 month was 0%
`compared with 15% in 74 patients on heparin control.
`This effect was independent of the type of reperfusion
`therapy or residual stenosis after treatment.
`Results of the Hirudin for the Improvement of
`Thrombolysis (HIT) study were recently published.106
`In this dose-escalation study, three doses of recombi-
`nant hirudin (HBW 023) were used as adjunctive ther-
`apy following front-loaded TPA for myocardial infarc-
`tion in 143 patients. Patency rates were determined at
`30, 60, and 90 minutes; at 36 to 48 hours; and after
`discharge. Ninety-minute TIMI-3 patency rates in-
`creased, and reocclusion rates diminished with increas-
`ing doses of hirudin. This trial did not have a heparin
`control arm, but patency rates compared favorably with
`those previously established for heparin, and the reoc-
`clusion rates were lower than the 10% to 15% rate
`associated with adjunctive heparin therapy. There were
`three spontaneous hemorrhages in the entire study
`group, and an increase in puncture site bleeding was
`noted in the highest dose group (5 of 83 patients).
`Cannon and colleagues107 reported the findings of the
`TIMI-5 trial that randomized 246 patients to either
`hirudin or heparin for 5 days following

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket