throbber
Current Topics in Medicinal Chemistry 2001,1, 151-159
`Coagulation Factor Xa Inhibition:
`Biological
`
`151
`
`
`
`Background Rationale
`
`and
`
`Robert J. Leadley, Jr.*
`
`Pfizer Global Research and Development, Ann Arbor Laboratories, 2800 Plymouth Rd., Ann
`Arbor, MI 48105, USA
`
`mm
`•
`leading
`
`the
`
`causes
`
`of
`
`Abstract:
`are
`disease
`cerebrovascular
`and
`Ischemic heart disease
`
`
`treated are by relatively antiquated drugs. However, due
`the world. Surprisingly,
`these diseases
`to our improved understanding of the underlying pathology of these diseases, and a number of
`technological advances in tools for drug discovery and chemical optimization, an exciting new
`wave of antithrombotic compounds is beginning to emerge in clinical trials. These agents,
`referred to as direct coagulation factor Xa inhibitors, appear to provide an enhanced risk-
`benefit margin compared to conventional therapy. Preclinical and early clinical data gathered
`over the past few years suggests that direct fXa inhibitors will provide the necessary
`advancements in efficacy, safety, and ease of use required to displace conventional therapy.
`Whether or not these agents will succeed will be determined as this class of agents advances
`through clinical trials in the near future. This
`review describes
`
`of some the key studies that sparked an interest in fXa as a
`
`therapeutic target, highlighting the findings that provided important rationale for continuing the development of potent
`and selective direct fXa
`inhibitors.
`
`;
`
`•-
`•, ! ^
`^
`
`I
`
`,
`
`INTRODUCTION
`
`antibodies against specific coagulation factors, synthetic
`indirect factor Xa (fXa) inhibitors, and possibly most
`promising, direct
`fXa
`inhibitors.
`
`Many approaches to developing antithrombotic drugs
`that interfere with enzymes in the coagulation system have
`been pursued over the past few decades; however, currently
`approved drags
`for
`thrombotic
`diseases
`
`have for
`in
`quite some
`time. Heparin,
`discovered
`1916
`intravenous anticoagulant of choice for acute thrombotic
`conditions. More recently, fractionated, or low-molecular-
`weight, heparins have demonstrated superiority over
`unfractionated heparin in several thrombotic indications [2,
`3], but remain limited to intravenous and subcutaneous
`administration. And although
`the coumarin derivative
`warfarin was discovered in 1941 and used initially as a
`rodenticide [4] that provides systemic anticoagulation by
`indirect
`inactivation of vitamin-K dependent
`serine
`proteases, it remains the most frequently prescribed oral
`anticoagulant.
`
`BACKGROUND
`FACTOR Xa BIOLOGICAL
`been
`around
`the
`[1]
`remains
`Factor X is the zymogen of fXa, a serine protease which
`
`occupies a pivitol position in the coagulation
`cascade. Factor
`X can be activated by the contact ("intrinsic tenase")
`pathway or by the tissue factor/Vila ("extrinsic tenase")
`pathway of the coagulation system (Fig. 1). Consequently,
`initiation of coagulation by either pathway in response to
`vascular injury activates factor X to fXa, making fXa
`inhibition a desirable intervention point when developing
`novel antithrombitics. Factor Xa and its cofactor, factor Va,
`combine on phospholipid membranes
`to
`form
`the
`"prothrombinase"
`complex, which activates prothrombin to
`thrombin. Thrombin, by cleaving fibrinogen to fibrin, by
`activating platelets, and by converting factor XIII to factor
`coagulation
`the
`of
`components
`Since the principal
`system
`Xllla, is
`the principal enzyme
`involved
`in
`thrombus
`have been known for some time, the lack of superior
`generation, growth,
`and
`stabilization.
`replacements for heparin and Coumadin is rather surprising.
`On the other hand, the redundancy and complexity of the
`hemostasis system
`is extensive and has provided tremendous
`obstacles for discovering potent novel agents that are
`effective, yet safe. Fortunately, recent scientific findings and
`advances in technology and testing have provided tools to
`identify and develop new agents that will hopefully provide
`safer and more effective antithrombotic therapy than are
`currently available. Among these newer agents are direct
`thrombin
`inhibitors,
`tissue
`factor pathway
`inhibitors,
`
`Factor X has long been recognized to play a significant
`role in hemostasis [5]. Factor X deficiency, while extremely
`rare, was first described in the 1950s
`in two families, Prower
`and Stuart (thus the original nomenclature of factor X as
`"Stuart" or "Stuart-Prower"
`
`
`7]). [6, factor These patients had
`
`very low antigen and activity levels of factor X, which were
`manifested in severe bleeding diatheses. Also, in the 1950s,
`studies of serum from these patients led to the identification
`of a deficiency of a specific factor, factor X, that was
`responsible for the bleeding diatheses. Since then, a number
`of variants of factor X deficiency suggest that factor X
`Therapeutics,
`at the Cardiovascular
`
`to correspondence this author
`
`
`*Address
`activity levels must be less than 5% of normal to result in
`Pfizer Global Research and Development, Ann Arbor Laboratories, 2800
`Plymouth Rd., Ann Arbor, Ml 48105, USA; Tel: 734-622-1420: Fax: 734­
`spontaneous bleeding
`tendencies [8]. This
`information
`622-1480;
`E-mail:
`ROBERT.LHADLEY@PFlZER.COM
`suggests that factor X activity can be suppressed markedly
`
`1568-0266/01
`
`S28,00+.00
`
`© 2001 Bentham Science Publishers
`
`Ltd.
`
`MYLAN - EXHIBIT 1018
`
`

`

`152 Current Topics in Medicinal Chemistry, 2001, Vol. I, No. 2
`
`Robert J. Lcadley, Jr.
`
`Coagulation Factor Xa Inhibition
`
`r J \
`
`Jf
`
`FVi
`
`i-'V
`
`m^aa^
`egffi
`
`i
`
`FVIJIa-
`
`FVIfS
`
`f' C i M ' X.
`FV ;'K C,i2\
`• r jro-.ifc- n i b -
`
`i
`
`FACTOR XII la ACTIVATION
`
`^
`
`FIBRIN FORMATION
`
`PLATELET AGGREGATION
`
`Fig. (1). Simplified schematic of the coagulation system and the intervention point for direct Factor Xa inhibitors.
`
`without affecting hemostasis, a characteristic that is desirable
`for antithrombotic agents. That is, an ideal agent would
`prevent
`thrombosis without producing systemic hypo-
`coagulation which would lead to bleeding complications at
`sites other than the intended target (e.g., intracerebral
`hemorrhage in patients treated for coronary artery disease).
`While this ideal approach was appealing, the hypothesis
`remained untested because no direct, selective inhibitors of
`fXa were available until fairly recently.
`
`NATURALLY OCCURING INHIBITORS OF FACTOR
`Xa
`
`In 1987, Tuszynski eta!., published their discovery of an
`anticoagulant, antistasin, which they isolated from extracts of
`the Mexican leech, Haementeria officinalis [9]. The amino
`acid sequence of antistasin was determined soon thereafter
`[10], and enzyme kinetic analysis detennined that antistasin
`is a slow, tight-binding, selective inhibitor of fXa with a Ki
`of 0.3-0.6 nM [11]. At approximately the same time,
`Waxman et ai, reported the discovery of tick anticoagulant
`peptide (TAP), a single chain 60 amino acid peptide that was
`isolated from the extracts of the tick Ornithodoros moubata
`[12]. TAP is a reversible, slow, tight-binding inhibitor of fXa
`with an estimated Ki of 0.5 nM. Molecular biology
`techniques quickly allowed scientists to make recombinant
`forms of these peptides which, particularly in the case of
`TAP, were used to validate fXa as a viable drug target and to
`aid in our understanding of the significance of fXa in
`thrombosis.
`
`VALIDATION OF FACTOR Xa AS A VIABLE DRUG
`TARGET
`
`A number of animal models of thrombosis were used to
`compare the antithrombotic effect of peptide inhibitors of
`fXa to direct inhibitors of thrombin and to indirect inhibitors
`of thrombin and fXa, such as heparin and low-molecular
`weight heparins [e.g., 13, 14, 15, 16]. These studies provided
`a preponderance of evidence to support the concept that
`direct fXa
`inhibition may provide a more effective
`antithrombotic approach than other mechanistic inhibitors,
`and that there appears to be a larger safety margin for fXa
`inhibitors as well, particularly regarding primary hemostasis
`at wound sites. For example, in canine models of thrombosis
`and thrombolysis, recombinant TAP was shown to promote
`rapid and prolonged vascular reperfiision at doses which
`produced relatively minor elevations in prothrombin time
`(PT), activated partial thromboplastin time (APTT), and
`template bleeding time [13, 14, 16, 17]. Although TAP was
`apparently discontinued from development for undisclosed
`reasons, the preclinical studies demonstrated
`that fXa
`inhibition was, indeed, an attractive drug target for the
`development of small molecule inhibitors of fXa.
`
`RATIONALE FOR DEVELOPING FACTOR Xa
`INHIBITORS
`
`TAP also provided a valuable tool for defining the role of
`fXa in thrombus formation and growth, thereby providing
`
`

`

`Coagulation Factor Xa Inhibition
`
`Current Topics in Medicinal Chemistry, 2001, Vol. I, No. 2 153
`
`further rationale to support the concept of fXa inhibition as a
`desirable drug target. The following discussion highlights
`some of the important findings.
`
`From experiments performed by Eisenberg, et al [18, 19],
`it became clear
`that Xa associated with
`thrombi
`is
`enzymatically active. For example, fXa in thrombi recovered
`from
`injured arteries in vivo is capable of activating
`prothrombin
`to
`thrombin [19]. Furthermore, fXa,
`in
`comparison to thrombin, appears to be primarily responsible
`for clot-associated procoagulant activity [20]. Specifically,
`TAP inhibited clot-associated generation of Fibrinopeptide A
`(a byproduct of thrombin-induced cleavage of fibrinogen to
`fibrin) to the same degree as the direct thrombin inhibitor,
`hirudin, suggesting that the procoagulant activity of thrombi
`is due to the de novo activation of prothrombin to thrombin,
`not merely to the presence of preexisting thrombin. These
`data indicate that fXa is active in thrombi and that direct
`inhibition of thrombus-associated fXa would be an effective,
`highly localized approach to prevention of thrombus growth.
`
`One of the current problems associated with anti-
`thrombin therapy for acute coronary syndromes is the
`phenomenon of "rebound" ischemia shortly after the therapy
`is terminated. This phenomenon was observed with direct
`and
`indirect, antithrombin Ill-dependent
`inhibitors of
`thrombin (argatroban and heparin, respectively [21, 22] and
`is thought to occur because of continual generation of
`thrombin after therapy is discontinued. Presumably, these
`agents were able to block thrombin activity as long as the
`drug was present, but the generation of thrombin by
`thrombus-associated
`fXa continued after
`therapy was
`withdrawn, resulting in recurrence of thrombotic ischemic
`events.
`
`Recent experiments indicate that short-term inhibition of
`fXa by TAP provides sustained inhibition of clot-associated
`procoagulant activity in vitro [23]. These results were
`validated in vivo by examining platelet deposition on dacron
`vascular grafts in baboons during and after a 2 hour
`intravenous infusion of rTAP [24], TAP decreased platelet
`deposition during the infusion and for 2 and 53 hours after
`termination of low dose (10 ng/kg/min) and high dose (25
`Hg/kg/min) rTAP, respectively. In another experiment, CI-
`1031 (ZK-807834), a small molecule, direct inhibitor of fXa,
`was administered as adjunctive treatment during and for 1 hr
`post fibrinolytic therapy to dogs that had experimentally-
`induced occlusive coronary artery
`thrombosis
`[25].
`Compared to heparin plus aspirin, 24-hr vessel patency
`achieved with CI-1031 was markedly enhanced. These
`findings were also supported by a preliminary report on a
`study in which another small molecule inhibitor of fXa,
`FXV673, administered
`to canines over a
`few hours
`prevented thrombosis for three days following electrolytic
`injury-induced thrombosis in the carotid artery [26]. These
`data suggest that the sustained antithrombotic effect after
`short-term drug exposure is not a feature of TAP alone, but
`is a unique and highly desirable effect of direct fXa
`inhibition. This
`feature,
`sometimes
`referred
`to as
`"passivation," may be important to consider when designing
`clinical studies to evaluate novel fXa inhibitors in acute
`thrombotic diseases, especially
`in
`the current medical
`
`environment which mandates short hospital stays for
`interventions such a percuataneous coronary interventions.
`
`inhibition provide sustained
`fXa
`Not only does
`antithrombotic protection, fXa inhibition also disaggregates
`prefonned platelet thrombi in vivo [27]. In experiments using
`anesthetized pigs, the carotid artery was damaged by serial
`hemostat crushes and the resulting thrombus was allowed to
`grow for 30 min prior to drug administration. Despite the
`fact that TAP and hirudin have no intrinsic lytic activity,
`these agents not only prevented further platelet deposition,
`but they actually reduced radiolabelled platelet deposition at
`the site of vascular damage. Worth noting is the observation
`that with TAP, the "dissaggregation" or "dethrombosis"
`occurred at doses of TAP that yielded only modest changes
`in clotting time assays such as APTT. In summary, newly
`fonned thrombi (<6 hours) can disaggregate if exposed to a
`hirudin or TAP. If proven to be true for small molecule
`direct
`inhibitors of
`fXa,
`this
`intriguing
`feature of
`"dethrombosis" may
`be
`an
`important
`therapeutic
`characteristic of direct fXa inhibitors, particularly for the
`acute treatment of thrombotic syndromes.
`
`INSIGHTS PROVIDED BY SYNTHETIC
`NEW
`INHIBITORS OF FACTOR Xa
`
`Based on the favorable preclinical evidence provided by
`recombinant forms of naturally oecuring fXa inhibitors,
`many pharmaceutical companies quickly initiated chemistry
`programs that produced potent and selective small molecule
`inhibitors of DCa, of which DX9065a is the most widely
`characterized agent in the literature [e.g., 28, 29, 30, 31, 32,
`33],
`
`inhibitor
`indirect fXa
`the success of
`In addition,
`heparanoids, such as low-molecular-weight heparins, in
`venous [3] and arterial thrombosis [2] prompted interest in
`continuing the development of more selective indirect
`inhibitors of fXa that were designed based on the minimal
`saccharide sequence of heparin required for antithrombin-III
`binding
`[34], SR90107A/ORG31540
`is
`the
`furthest-
`developed of a family of synthetic pentasaccharides that are
`selective, antithrombin-III dependent fXa inhibitors.
`
`and SR90107A/
`agents, DX-9065a
`two
`These
`ORG31540 have been extensively evaluated in in vivo
`models of thrombosis, exemplified best, perhaps, by their
`efficacy
`in a baboon model of shunt
`thrombosis.
`SR90107A/ORG31540 dose-dependently inhibited platelet
`and fibrin deposition on shunts that represented conditions of
`venous
`thrombosis, but was relatively
`ineffective
`in
`preventing platelet deposition on shunts mimicing arterial,
`platelet-dependent thrombosis [35], Likewise, intravenous or
`oral administration of DX-9065a significantly inhibited
`thrombus fonnation in a venous-type chamber but had no
`significant antithrombotic effect in an arterial-type chamber
`[30]. These results suggest that selective inhibition of fXa
`results
`in specific inhibition of fibrin formation with
`relatively less activity against platelet-dependent thrombi.
`These data appear to directly contradict the data from
`experiments mentioned earlier in which platelet deposition
`on injured porcine arteries was reversed by fXa inhibition
`[27]. Which experimental condition is more comparable to
`
`

`

`154 Current Topics in Medicinal Chemistry, 200], Vol. I, Mo. 2
`
`Robert J. Leadley, Jr.
`
`the situation in patients with arterial thrombosis is still
`uncertain, especially in regard to how these new agents
`perform in the model versus the clinical setting. This
`apparent discrepancy is important to consider because it
`could have a direct impact on the indications targeted for a
`fXa inhibitor and on the design of clinical trials of novel fXa
`inhibitors
`(e.g., are antiplatelet agents
`required as
`concomitant therapy with fXa inhibitors?).
`
`SEPARATION OF ANTITHROMBOTIC EFFICACY
`FROM BLEEDING EFFECTS
`
`As alluded to previously, one of the most intriguing
`advantages of direct fXa inhibitors over conventional therapy
`is
`the
`relatively
`large
`therapeutic window between
`antithrombotic efficacy and bleeding tendency. Numerous
`studies have demonstrated antithrombotic efficacy at doses
`of fXa inhibitors that have little or no effect on markers of
`primary hemostasis such as template bleeding time, tail
`resection bleeding time, or cuticle bleeding time [32, 33, 36,
`37, 38, 39]. Sato et al, [36] demonstrated that the risk-
`benefit ratio (dose that doubled bleeding time/dose that
`produced 50% inhibition in a rat venous thrombosis model)
`for YM-60828 was dramatically higher than the ratio
`measured with argatroban, heparin, and dalteparin (94 versus
`7.4, 2.9, and 5.3, respectively). Similar risk-benefit analysis
`could be applied to the studies sited above, demonstrating a
`favorable safety profile for fXa inhibitors over other types of
`antithrombotic agents.
`
`There are several mechanisms that have been postulated
`to explain
`the minimal bleeding observed during
`administration of antithrombotic doses of fXa inhibitors in
`animal models of thrombosis. First, fXa has no direct effect
`on platelet aggregation and only activates platelets indirectly
`via generation of thrombin when Xa is assembled into the
`prothrombinase complex. Consequently, unlike direct
`thrombin inhibitors or heparinoids, nearly all direct fXa
`inhibitors have no inhibitory effect on thrombin- or thrombin
`receptor
`activator
`peptide
`(TRAP)-induced
`platelet
`aggregation [28, 40, 41]. The only cited exception is YM-
`60828, which inhibits TRAP-induced platelet aggregation in
`plasma with an IC50 of 3.3
`and thrombin-induced
`washed platelet aggregation with an IC50 of 23.4 jiM [42],
`Since thrombin is a potent activator of platelets, and its
`affinity for platelet receptors is 10,000-fold higher than for
`fibrinogen [43, 44] minimal amounts of thrombin may be
`adequate to activate platelets and allow for normal systemic
`hemostasis.
`
`Minimal bleeding with fXa inhibitors may be attributed
`to reduction of thrombin generation, as opposed to blocking
`thrombin's activity once it is fonned. Once incorporated into
`the prothrombinase complex, the reaction rate of fXa
`increases 300,000-fold compared to the rate of free fXa,
`thereby generating an explosive production of thrombin [45],
`Inhibiting fXa directly with potent, selective agents therefore
`provides efficient reduction of thrombin generation at the
`thrombus, without the need for excessively high systemic
`plasma concentration of the antithrombotic drug.
`
`SEPARATION OF ANTITHROMBOTIC EFFICACY
`FROM SYSTEMIC HYPOCOAGULATION
`Several studies also demonstrated that, in comparison to
`other mechanisms of inhibition of thrombus formation (e.g.,
`unfractionated heparin, low-molecular-weight heparin, and
`direct thrombin inhibition), fXa inhibitors produced their
`antithrombotic effect with only modest changes in markers
`of systemic anticoagulation, such as PT, APTT, and
`thrombin time [32, 33, 36, 37, 38, 39, 46) . For example,
`maximally-effective antithrombotic doses of the direct fXa
`inhibitor, C92I-78, heparin, enoxaparin, and PPACK (a
`direct thrombin inhibitor) in a rabbit model of arteriovenous
`shunt thrombosis produced changes in cuticle bleeding times
`of approximately 2- (not significant from baseline), 5.5-, 3.5-,
`and 6-fold, and changes in APTT of 1.6-, >6-, 5-, and 1.3­
`fold over baseline, respectively [39]. Similarly, when
`RPR208566 was compared to heparin and argatroban (a
`direct thrombin inhibitor), PT and APTT were not changed
`significantly (<1.5-fold), while maximally effective doses of
`argatroban and heparin increased PT by 2.5- and 2.5-fold and
`APTT by 3- and 6-fold,
`respectively
`[46]. These
`observations have been made with a number of compounds
`from different chemical series indicating that, although there
`are certainly differences in the pharmacodynamics between
`individual fXa inhibitors, the overall safety advantages of
`fXa inhibition are mechanism-related.
`
`interestingly, the effect of potent selective inhibitors of
`fXa on clotting time assays is highly variable. For example,
`some fXa inhibitors appear to be more potent in the APTT
`assay than in the PT assay (TAP [15]; RPR120844, [47]),
`some are more potent in the PT than in the APTT assay (DX-
`9065a [28]; CI-1031 [38]), and others appear to be equally
`potent in both assays (YM-60828 [42], SKS49 [48]). The
`sensitivity of these tests for fXa inhibitors appears to be
`compound-dependent and may reflect as yet undefined
`differences in enzyme kinetics. Regardless, the APTT, PT,
`and activated clotting time (ACT) assays, which are used
`routinely to monitor heparin and warfarin treatment, are not
`sensitive enough to accurately monitor fXa inhibitors when
`administered at antithrombotic doses in animal models of
`thrombosis. Consequently, other more specific assays such
`as chromogenic fXa activity assays, Russel's viper venom
`clotting assay, the Heptest®, or thrombin generation assays
`might prove to be more sensitive pharmacodynamic markers
`for fXa inhibitors.
`
`Although the explanation for the ability of fXa inhibitors
`to provide antithrombotic protection without increasing
`systemic markers of anticoagulation is not yet clearly
`elucidated, the affinity of these agents for thrombus-
`associated fXa may provide a plausible explanation [49, 50].
`By inhibiting thrombus-bound fXa with high affinity, lower
`plasma drug concentrations may be required to provide
`antithrombotic efficacy. Significant prolongation of APTT
`and PT can be achieved by high-dose administration of fXa
`inhibitors, indicating that systemic hypocoagulation, and the
`susceptibility for bleeding complications, is not avoided
`completely by fXa inhibitors. However, the doses required to
`reach potentially dangerous plasma levels are quite high
`compared
`to doses providing maximal antithrombotic
`efficacy.
`
`

`

`Coagulation Factor Xa Inhibition
`
`Current Topics in Medicinal Chemistry, 2001, Vol 1, No. 2 155
`
`OTHER ADVANTAGES OF FACTOR Xa INHIBI­
`TION OVER CONVENTIONAL THERAPY
`
`Additional rationale for developing fXa inhibitors to
`replace heparin and warfarin are listed in (Table I). The
`considerations for dosing and monitoring heparin therapy
`have been extensively reviewed by Hirsh, et al. [51]. Briefly,
`heparin administration must be monitored carefully to
`maintain plasma drug concentrations within a safe and
`effective window. Monitoring is necessary because heparin
`binds to a number of plasma proteins and its activity is
`neutralized by platelet factor 4 released from activated
`platelets,
`resulting
`in highly variable anticoagulant
`responses. Also, heparin administration causes an immune
`reaction referred to as heparin-induced thrombocytopenia
`(HIT) in approximately 1-3% of patients; LMWHs also
`produce HIT, but at a lower incidence compared
`to
`unfractionated heparin. Low molecular weight heparins have
`eliminated the need for monitoring in most indications, but
`have not substantially replaced heparin in acute coronary
`syndromes due, in part, to the lack of a specific monitoring
`device that can provide rapid feedback for dose adjustment
`before, during, and after percutaneous coronary inter­
`ventions. The limitations of indirect versus direct inhibition
`of coagulation enzymes has been highlighted in studies by
`Weitz et al. [52] and by Herault et al., [49]. As mentioned
`above, fXa and thrombin are both enzymatically active when
`associated with the intravascular thrombus, so it is vitally
`important that an antithrombotic agent be able to inhibit this
`so-called "clot-bound" activity. Indirect inhibitors act by
`catalyzing
`the
`inactivation of
`thrombin or
`fXa by
`antithrombin-III, a large molecule that is not able to
`penetrate the thrombus. Consequently, heparin, LMWHs, or
`the synthetic pentasaccharides are not effective inhibitors of
`thrombus-associated fXa or
`thrombin. Direct fXa and
`thrombin inhibitors, however, are capable of inhibiting
`thrombus-associated fXa and thrombin, making these agents
`more attractive for medical interventions in thrombotic
`diseases. The other features of fXa inhibitors that were
`highlighted above, namely "dethrombosis" and vascular
`"passivation," are important characteristics for agents that
`
`will be administered over a brief period. Emergency
`treatment of acute coronary syndromes and percutaneous
`coronary intervention by intravenous administration of fXa
`inhibitors may provide ideal settings to evaluate whether the
`mechanistic advantages of fXa inhibition will translate into
`superior efficacy and safety over heparin.
`
`Currently, heparinoids are limited to subcutaneous and
`intravenous administration because they are not absorbed
`adequately after oral administration. However, new carriers
`have been developed that have enhanced the bioavailability
`of heparin so that an oral form of this agent is now being
`evaluated
`in clinical
`trials [53]. Regardless,
`the oral
`formulation of heparin will still have the same limitations as
`i.v. heparin, so that by virtue of inherent interindividual
`differences in oral absorption, orally-administered heparin
`will likely be even more difficult to maintain in the safe and
`effective therapeutic plasma concentration range.
`
`The limitations of oral anticoagulation with warfarin are
`well-recognized and have effectively restricted its use in the
`clinic [54], Warfarin acts in the liver by antagonizing the
`vitamin K-dependent carboxylation of glutamic acid residues
`on the amino terminal of coagulation factors II, VII, IX, and
`X. These residues are essential for calcium-dependent
`binding of these enzymes into their appropriate enzyme
`complexes on phospholipid surfaces. The approximate half-
`life of factor 11 (prothrombin) is 50 hours, so several days are
`required before warfarin achieves its full antithrombotic
`effect. Likewise, the reversal of the effect of warfarin can
`take 24 hr or longer. In addition, warfarin has many drug and
`food interactions that complicate its dosing and require
`regular monitoring to maintain safe and effective plasma
`drug concentrations [54, 55]. The significant bleeding
`complications
`and
`difficulty maintaining
`plasma
`concentrations of warfarin within the targetted range has led
`to the labeling of warfarin (Coumadin®) as a "narrow
`therapeutic index drug."
`
`Obviously, a fXa inhibitor that is orally bioavailable and
`does not have many of the drawbacks of warfarin would be
`
`Table I.
`
`Comparison of Direct tXa Inhibitors to Currently Available Anticoagulants
`
`Enzymes
`inhibited
`
`Cofactor
`required?
`
`Inhibition of clot-
`bound enzyme?
`
`Monitoring
`required?
`
`Heparin-induced
`thrombocytopenia?
`
`Route of
`administration
`
`Risk/Benefit
`
`N
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`N
`
`N
`
`N
`
`N
`
`Y
`
`Y
`
`N**
`
`Y
`
`++++
`
`N
`
`N
`
`Y
`
`Y
`
`N
`
`i.v., oral
`
`s.c., i.v.
`
`i.v., s.c.
`
`s.c., i.v.
`
`s.c., i.v.
`
`Oral, i.v.
`
`Agent
`
`Direct fXa
`
`Direct Ha
`
`Heparin
`
`LMWH
`
`fXa
`
`11a
`
`Xa-IJa
`
`Xa>IIa
`
`Pentasaccharide*
`
`Xa»>I]a
`
`Warfarin
`
`I la, Vila, IXa,
`Xa, PC, PS
`
`*
`
`*
`**
`
`Risk/Benefit is based on a qualitative estimate using heparin as a comparator. Hie risk/benefit for iXa inhibitors is speculative, based on preclinical evidence and experience
`with indirect fXa inhibitors.
`The pentasaccharide is not currently approved for use, but is included because the positive results in Phase III clinical trials will likely lead to approval in the near future.
`fn general, monitoring of LMWHs is not required. However, for indications such as percutaneous coronary intervention it is desirable to know the anticoagulation level of the
`patient before, during, and after the procedure.
`PC: Protein C; PS: Protein S
`
`

`

`156 Current Topics in Medicinal Chemistry, 2001, Vol. I, No. 2
`
`Robert J. Lcadley, Jr.
`
`highly desirable pharmaceutical agent. Many of the known
`fXa inhibitors have some degree of oral bioavailability [28,
`37, 41, 42, 56, 57, 58, 59], and several have demonstrated
`antithrombotic efficacy in animal models of thrombosis after
`oral or intraduodenal administration [30, 32, 41, 60].
`Although the hurdles for discovering a fXa inhibitor that has
`the appropriate pharmacokinetic properties are quite
`challenging, these data are encouraging and suggest that
`orally active fXa inhibitors may be coming closer to
`realization.
`
`FACTOR Xa INHIBITORS IN CLINICAL TRIALS
`
`Preclinical studies provided sufficient safety and efficacy
`data to propel fXa inhibitors into clinical trials, the first of
`which were the direct fXa inhibitor, DX-9065a, and the
`indirect
`antithrombin
`Ill-dependent
`fXa
`inhibitor,
`SR90I07A/ORG31540 (for review, see Porcari el al. [61]).
`Early
`safety
`trials
`indicated
`that DX-9065a
`and
`SR90107A/ORG31540 can be administered predictably and
`safely
`to normal volunteers [62, 63] and
`to patients
`undergoing percutaneous transluminal coronary angioplasty
`[64]. More recently, preliminary reports on the use of
`SR90107A/ORG31540
`in patients undergoing major
`orthopedic surgery indicate that SR90107A/ORG31540
`appears to be as safe as enoxaparin (a low-molecular-weight
`heparin), but
`reduces
`the overall
`risk of venous
`thromboembolism by 50% over enoxaparin [65]. These
`results are interesting in light of the limited inhibition of
`prothrombinase
`activity
`by
`SR90107A/ORG31540
`compared to DX9065a in vitro [49]. If indirect inhibition of
`fXa produces such a dramatic improvement in efficacy over
`enoxaparin, it will be very interesting to discover whether
`the potential mechanistic advantages of a potent, direct
`inhibitor of fXa will be manifested in significantly better
`outcomes in this patient population. While several other
`small molecule fXa inhibitors have likely progressed into
`clinical trials, no data is currently available in the literature.
`
`POTENTIAL
`INHIBITORS
`
`INDICATIONS FOR FACTOR Xa
`
`The limitations of currently available agents such as
`heparin and warfarin demand improved acute and chronic
`therapy for thromboembolic diseases. If the mechanistic,
`safety, and efficacy advantages of fXa inhibition ring true,
`the potential therapeutic uses for fXa inhibitors are virtually
`unlimited. Any thrombotic indication that has an underlying
`pathology of
`fibrin deposition or
`thrombin-dependent
`platelet activation and aggregation would certainly benefit
`from fXa
`inhibition. These
`indications
`include acute
`coronary syndromes, adjunctive therapy with thrombolytics
`for acute myocardial infarction, stroke, peripheral vascular
`disease, disseminated intravascular coagulation, and deep
`vein
`thrombosis. In additon, whenever atherosclerotic
`vessels are damaged, either by spontaneous plaque rupture or
`by percutaneous interventions such as balloon angioplasty or
`stent deployment, tissue factor is exposed and fXa is
`generated at the site of the lesion. Consequently, fXa
`inhibition would be desirable in the setting of unstable
`angina with or without percutaneous interventions. With
`
`percutaneous intervention, fXa inhibitors may also prevent
`restenosis after balloon angioplasty or stent placement by
`virtue of
`their antimitogenic effect [66]. Also, when
`thrombolytic therapy is administered, active fXa is exposed
`in the residual thrombus and requires inactivation to prevent
`reocclusion.
`Furthennore,
`by
`preventing
`thrombin
`generation, fXa inhibition may be important in preventing
`platelet
`activation, deposition,
`and
`recruitment, as
`demonstrated by the decrease in platelet deposition on
`damaged vessels
`and
`collagen-coated
`surfaces by
`administration of fXa inhibitors [27, 30, 67]. Finally, fXa
`inhibitors may serve as excellent anticoagulants
`for
`extracorporeal circulation systems, hemodialysis, and even
`for blood sampling tubes and systems (depending on the
`desired test). Overall, the possibilities for antithrombotic
`indications for fXa inhibitors are abundant and diverse, and
`should not be underestimated.
`
`CONCLUSION
`
`The importance of fXa in thrombosis and hemostasis has
`been widely appreciated for many years. This understanding
`logically led to the concept of fXa inhibition as a viable and
`attractive target for antithrombotic therapy. The anticipated
`enhanced efficacy and safety of fXa
`inhibition over
`conventional therapy has been whole-heartedly supported by
`results of preclinical experiments with potent and selective
`inhibitors of fXa. Unf

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