throbber
J Thromb Thrombolysis (2011) 31:478–492
`DOI 10.1007/s11239-011-0551-3
`
`Preclinical discovery of apixaban, a direct and orally bioavailable
`factor Xa inhibitor
`
`Pancras C. Wong • Donald J. P. Pinto •
`Donglu Zhang
`
`Published online: 13 February 2011
`Ó Springer Science+Business Media, LLC 2011
`
`Abstract Apixaban (BMS-562247; 1-(4-methoxyphenyl)-
`7-oxo-6-(4-(2-oxopiperidin-1-yl)phenyl)-4,5,6,7-tetrahydro-
`1H-pyrazolo[3,4-c]pyridine-3-carboxamide),
`a
`direct
`inhibitor of activated factor X (FXa), is in development for
`the prevention and treatment of various thromboembolic
`diseases. With an inhibitory constant of 0.08 nM for human
`FXa, apixaban has greater than 30,000-fold selectivity for
`FXa over other human coagulation proteases. It produces a
`rapid onset of inhibition of FXa with association rate
`constant of 20 lM
`-1/s approximately and inhibits free as
`well as prothrombinase- and clot-bound FXa activity in
`vitro. Apixaban also inhibits FXa from rabbits, rats and
`dogs, an activity which parallels its antithrombotic potency
`in these species. Although apixaban has no direct effects on
`platelet aggregation, it indirectly inhibits this process by
`reducing thrombin generation. Pre-clinical
`studies of
`apixaban in animal models have demonstrated dose-
`dependent antithrombotic efficacy at doses that preserved
`hemostasis. Apixaban improves pre-clinical antithrombotic
`activity, without excessive increases in bleeding times,
`when added on top of aspirin or aspirin plus clopidogrel at
`their
`clinically relevant doses. Apixaban has good
`
`P. C. Wong (&)
`Department of Cardiovascular Biology, Bristol-Myers Squibb
`Company, 311 Pennington-Rocky Hill Road, Pennington,
`NJ 08534, USA
`e-mail: pancras.wong@bms.com
`
`D. J. P. Pinto
`Department of Medicinal Chemistry, Bristol-Myers Squibb
`Company, Princeton, NJ, USA
`
`D. Zhang
`Department of Pharmaceutical Candidate Optimization,
`Bristol-Myers Squibb Company, Princeton, NJ, USA
`
`123
`
`bioavailability, low clearance and a small volume of dis-
`tribution in animals and humans, and a low potential for
`drug–drug interactions. Elimination pathways for apixaban
`include renal excretion, metabolism and biliary/intestinal
`excretion. Although a sulfate conjugate of O-demethyl
`apixaban (O-demethyl apixaban sulfate) has been identified
`as the major circulating metabolite of apixaban in humans,
`it is inactive against human FXa. Together, these non-
`clinical findings have established the favorable pharmaco-
`logical profile of apixaban, and support the potential use of
`apixaban in the clinic for the prevention and treatment of
`various thromboembolic diseases.
`
`Keywords Apixaban Factor Xa Anticoagulants
`Thrombosis Atrial fibrillation
`
`Introduction
`
`Thrombosis is a major cause of morbidity and mortality in
`the Western world and plays a pivotal role in the patho-
`genesis of numerous cardiovascular disorders, including
`acute coronary syndrome (ACS) (i.e. unstable angina and
`myocardial infarction), sudden cardiac death, peripheral
`arterial occlusion, ischemic stroke, deep vein thrombosis
`(DVT) and pulmonary embolism. Despite recent advances
`in interventional and drug therapy for thrombosis,
`the
`burden of thrombotic disease remains unacceptably high
`[1, 2]. There is therefore a significant need for new anti-
`thrombotic therapies that are more effective and provide
`improved safety profile compared with current treatments.
`This review focuses on the pre-clinical discovery of apix-
`aban, a promising new oral antithrombotic agent that spe-
`cifically targets activated factor X (FXa) of the blood
`coagulation cascade.
`
`BMS 2009
`MYLAN v. BMS
`IPR2018-00892
`
`

`

`Preclinical discovery of apixaban
`
`479
`
`Drug discovery strategy—targeting factor Xa
`
`As the last serine protease in the blood coagulation cas-
`cade, thrombin is the key enzyme responsible for physio-
`logical fibrin clot
`formation and platelet activation.
`Thrombin also plays a prominent role in the pathologic
`generation of occlusive thrombi in arteries or veins, a
`process that may lead to arterial or venous thrombotic
`disease. Thus, attenuation of the activity of thrombin—
`either via direct inhibition or via blockade of other prote-
`ases that lie upstream in the coagulation cascade and are
`intimately involved in thrombin generation (e.g. FXa)—
`has been intensively investigated as a novel means to
`prevent and treat thrombotic disease.
`Three key observations supported our hypothesis that
`inhibition of FXa may represent an acceptable approach for
`effective and safe antithrombotic therapy. First, as the
`process of blood coagulation involves sequential activation
`and amplification of coagulation proteins, generation of
`one molecule of FXa can lead to the activation of hundreds
`of thrombin molecules [3]. In principle, therefore, inhibi-
`tion of FXa may represent a more efficient way of reducing
`fibrin clot formation than direct inhibition of thrombin
`activity. This principle is consistent with an in vitro
`observation, suggesting that
`inhibition of FXa but not
`thrombin may result in a more effective sustained reduction
`of thrombus-associated procoagulant activity [4]. Second,
`inhibition of FXa is not thought to affect existing levels of
`thrombin. Further, reversible FXa inhibitors might not
`completely suppress the production of thrombin. These
`small amounts of thrombin might be sufficient to activate
`high affinity platelet thrombin receptors to permit physio-
`logical
`regulation of hemostasis.
`Indeed, experimental
`evidence from animal studies suggests that the antithrom-
`botic efficacy of FXa inhibitors is accompanied by a lower
`risk of bleeding when compared with thrombin inhibitors
`[5–7] (for review, see Hauptmann and Stu¨rzebecher [8] and
`Leadley [9]). Finally, the strongest evidence for FXa as an
`antithrombotic drug target is the clinical proof of concept
`studies of the indirect FXa inhibitor fondaparinux [10].
`Taken together, these observations suggest that inhibition
`of FXa is a potentially attractive antithrombotic strategy.
`We initiated a drug discovery program on small-mole-
`cule direct FXa inhibitors, with the goal of identifying
`novel oral anticoagulants not burdened by the well-known
`limitations of vitamin K antagonists such as warfarin,
`agents that remain the only oral anticoagulants approved
`for long-term use until very recently [11]. (On October 19,
`2010, FDA approved the oral direct thrombin inhibitor
`dabigatran etexilate to prevent stroke and blood clots in
`patients with non-valvular atrial fibrillation [12].) These
`new FXa inhibitors would have the following target profile.
`First, they would be direct, highly selective and reversible
`
`inhibitors of FXa, with a rapid onset of action, and would
`demonstrate a relatively wide therapeutic index and few
`food and drug interactions (thereby avoiding the need for
`frequent coagulation monitoring and dose adjustment).
`Second,
`these FXa inhibitors would have predictable
`pharmacokinetic and pharmacodynamic profiles that allow
`fixed oral dosing, accompanied by low peak-to-trough
`plasma concentrations that provide high levels of efficacy
`and low rates of bleeding. Finally, as the FXa target resides
`in the central or blood compartment, the pharmacokinetic
`profile of these agents would also feature a low volume of
`distribution (to minimize off-target risks) and low systemic
`clearance
`(to
`reduce
`the
`potential
`for
`drug-drug
`interactions).
`Based on many years of research and development, we
`have identified the potent, highly selective and direct FXa
`inhibitor, apixaban (BMS-562247) [13–15]. Apixaban is
`one of the most promising specific, single-target oral
`anticoagulants in late clinical development. In clinical tri-
`als, apixaban has been shown to provide predictable and
`consistent anticoagulation, accompanied by promising
`efficacy and safety profiles in the prevention and treatment
`of various thromboembolic diseases [16–22]. The phar-
`macological and clinical profiles of apixaban suggest that it
`has the potential to address many of the limitations of
`warfarin therapy, currently the standard of care in chronic
`oral anticoagulation. In this review, we summarize the
`chemistry and pre-clinical profile of apixaban.
`
`Chemistry
`
`Apixaban is a small-molecule, selective FXa inhibitor. It is
`chemically described as 1-(4-methoxyphenyl)-7-oxo-6-[4-
`(2-oxopiperidin-1-yl)phenyl]-4,5,6,7-tetrahydro-1H-pyraz-
`olo[3,4-c]pyridine-3-carboxamide. The molecular formula
`for apixaban is C25H25N5O4, which corresponds to a
`molecular weight of 459.5.
`
`Discovery of apixaban
`
`In the early 1990s, DuPont scientists invested a great
`amount of effort in the development of inhibitors of gly-
`coprotein IIb/IIIa. These efforts resulted in several com-
`pounds that were advanced to clinical trials as potential
`anti-platelet agents. By the mid-1990s, scientists at DuPont
`had recognized similarities between the platelet glycopro-
`tein GPIIb/IIIa peptide sequence Arg-Gly-Asp (RGD) and
`the prothrombin substrate FXa sequence, Glu-Gly-Arg
`(EGR). Consequently, a high-throughput lead evaluation
`program was initiated to screen the IIb/IIIa library for FXa
`inhibitory activity. This effort resulted in the identification
`
`123
`
`

`

`P. C. Wong et al.
`
`H2NO2S
`
`MeO2C
`
`O
`
`NH
`
`N O
`
` 2 (SF303)
`
`fXa Ki = 6.3 nM
`
`SO2NH2
`
` 4 (SN429)
`
`fXa Ki = 0.013 nM
`
`H2N
`
`NH
`
`HN
`
`O
`
`H3C
`
`N
`
`N
`
`H2N
`
`NH
`
`Me2
`
`NN
`
`N
`
`F
`
`HN
`
`O
`
`NH2
`
`F3C
`
`N
`
`N
`
`O N
`
` 6 (Razaxaban)
`
` fXa Ki = 0.19 nM
`
`H2N
`
`O
`
`N
`
`N
`
`N
`
`O
`
`NH
`
`HOOC
`
`N
`
`O O
`
`O
`
`N
`
`1
`
`Initial Screening Lead
`fXa Ki = 38.5 μM
`
`H2N
`
`NH
`
`SO2NH2
`
`HN
`
`O
`
`O
`N
`
`H2N
`
`NH
`
` 3
`
`fXa Ki = 0.1 nM
`
`SO2Me
`
`F
`
`HN
`
`O
`
`F3C
`
`N
`
`N
`
`NH2
`
` 5 (DPC423)
`
`fXa Ki = 0.15 nM
`
`OH
`
`N
`
`F3C
`
`N
`
`N
`
`N
`
`O
`
`O N
`
`NH2
` 7 (BMS-740808)
`
` fXa Ki = 0.03 nM
`
` 8
`
`OCH3
`
`fXa Ki = 0.14 nM
`
`H2N
`
`O
`
`N
`
`N
`
`N
`
`O
`
`O
`
`N
`
`OCH3
`
` 9 (BMS-562247, Apixaban)
`
` fXa Ki = 0.08 nM
`
`
`
`480
`
`Fig. 1 The evolution of the
`pyrazole-based FXa inhibitors:
`the discovery of apixaban
`
`of a small number of isoxazoline derivatives such as 1
`(FXa Ki = 38.5 lM) (Fig. 1) [23]. Using molecular mod-
`eling and structure-based design, an optimization strategy
`resulted in the identification of a benzamidine containing
`FXa inhibitor 2 (SF303) with enhanced potency (FXa
`= 6.3 nM) and potent antithrombotic activity in an
`Ki
`experimental model of thrombosis [24–26]. Aside from the
`
`key amidine P1 and the enzyme Asp189 interaction, the
`biarylsulfonamide P4 moiety was designed to neatly stack
`in the S4 hydrophobic box of FXa, which contains the
`residues Tyr99, Phe174 and Trp215, with the terminal
`O-phenylsulfonamide ring making an edge-to-face inter-
`action with Trp215. Subsequent re-optimizations led to
`vicinally substituted isoxazole analogs such as compound
`
`123
`
`

`

`Preclinical discovery of apixaban
`
`481
`
`3, which retained anti-FXa potency (FXa Ki = 0.1 nM)
`[27] and a pyrazole analog 4 (SN429), which demonstrated
`13 pM binding affinity against FXa and good antithrom-
`botic activity in a rabbit model of thrombosis [28, 29]. The
`discovery of SN429 was tremendously important in that it
`set the stage for an optimization strategy that led to the
`important compounds, such as 5
`discovery of several
`(DPC423), a phase I clinical candidate with a long terminal
`half-life of approximately 30 h in humans [6, 28–30], and 6
`(razaxaban) [31, 32], a compound that was advanced to a
`phase II proof-of-principle clinical trial. In fact, razaxaban
`was the first small molecule FXa inhibitor to provide
`clinical validation of the effectiveness of FXa inhibition
`strategies [33].
`Development of razaxaban was quickly followed by the
`identification of a novel bicyclic tetrahydropyrazolo-pyr-
`= 0.03 nM) [34].
`idinone analog 7 (BMS-740808, FXa Ki
`The evolution of the bicyclic pyrazole template allowed for
`the incorporation of a diverse set of P1 groups, the most
`important of which was the p-methoxyphenyl analog 8
`(Ki = 0.14 nM) [13]. Compound 8 retained potent FXa
`affinity and good anticoagulant activity in vitro, was effi-
`cacious in in vivo rabbit antithrombotic models and
`showed high oral bioavailability in dogs. A significant
`breakthrough was subsequently achieved, via the incorpo-
`ration of a pendent P4 lactam group and a carboxamido
`pyrazole moiety, that led to the discovery of 9 (BMS-
`562247, FXa Ki = 0.08 nM) [13], a highly potent and
`selective FXa inhibitor with good efficacy in various ani-
`mal models of thrombosis. Importantly, compound 9 also
`showed an excellent pharmacokinetic profile in dogs, with
`low clearance, low volume of distribution and high oral
`bioavailability [13]. The superior pre-clinical profile dem-
`onstrated by 9 enabled its rapid progression into clinical
`development as apixaban [15]. Figure 2 illustrates the
`X-ray structure of apixaban bound to FXa and shows the
`
`p-methoxyphenyl P1 deeply inserted into the S1 pocket,
`with the aryllactam P4 moiety neatly stacked in the
`hydrophobic S4 pocket.
`
`In vitro pharmacology
`
`Potency, selectivity and kinetic mode of inhibition
`
`Apixaban is a highly potent, reversible, active-site inhibitor
`of human FXa, with a Ki of 0.08 nM at 25°C and 0.25 nM
`at 37°C in the FXa tripeptide substrate (N-a-benzyloxy-
`carbonyl-D-Arg-Gly-Arg-pNA) assay [35]. Analysis of
`enzyme kinetics shows that apixaban acts as a competitive
`inhibitor of FXa versus the synthetic tripeptide substrate,
`indicating that it binds in the active site. Apixaban pro-
`duces a rapid onset of inhibition under a variety of con-
`ditions with association rate constant of 20 (lM
`-1/s
`approximately, and shows competitive inhibition of FXa
`versus the synthetic tripeptide substrate. Reversibility
`of FXa inhibition is demonstrated by the recovery of FXa
`activity at 37°C upon 200-fold dilution of a pre-formed
`FXa:apixaban complex into tripeptide substrate, an effect
`associated with
`a
`dissociation
`rate
`constant
`of
`*0.0113 s
`-1. Unlike indirect inhibitors of thrombin and
`FXa, such as heparin, the low molecular weight heparins
`and fondaparinux, apixaban, a direct FXa inhibitor, does
`not require the presence of antithrombin III to inhibit FXa.
`As shown in Table 1, apixaban has greater than 30,000-
`fold selectivity for FXa relative to other human coagulation
`proteases and structurally related enzymes involved in
`digestion and fibrinolysis [13].
`In the prothrombinase assay, apixaban is an effective
`inhibitor of the action of human FXa on its physiological
`substrate, prothrombin, blocking the action of FXa on
`prothrombin within the prothrombinase complex with a Ki
`
`Table 1 In vitro Ki values for inhibition of human enzymes by
`apixaban at 25°C [13]
`
`Enzyme
`
`Factor Xa
`
`Activated protein C
`
`Chymotrypsin
`
`Factor IXa
`
`Factor VIIa
`
`Plasma kallikrein
`
`Plasmin
`
`Thrombin
`
`Tissue plasminogen activator
`
`Trypsin
`
`Ki (nM)
`
`0.08 ± 0.03
`[30,000
`3,500
`[15,000
`[15,000
`3,700
`[25,000
`3,100
`[40,000
`[20,000
`
`123
`
`Fig. 2 X-ray structure of apixaban bound to factor Xa
`
`

`

`482
`
`P. C. Wong et al.
`
`Table 2 In vitro potency (Ki) of apixaban against human, rabbit, rat
`and dog factor Xa (FXa) and the concentrations required to double
`the prothrombin time (PT), modified prothrombin time
`(EC29)
`PT EC29 (lM)
`mPT EC29 (lM)
`
`Species
`
`FXa Ki (nM)
`
`(mPT), activated partial thromboplastin time (aPTT) and HepTest
`in human, rabbit, rat or dog plasma [15]
`
`aPTT EC29 (lM)
`
`HepTest EC29 (lM)
`
`Human
`
`Rabbit
`
`Rat
`
`Dog
`
`0.081 ± 0.002
`0.16 ± 0.01
`1.3 ± 0.1
`1.7 ± 0.2
`
`3.6
`
`2.3
`
`7.9
`
`6.7
`
`n.d. not determined
`
`0.37
`
`0.6
`
`n.d.
`
`n.d.
`
`7.4
`
`4.8
`
`20
`[20
`
`0.4
`
`1.8
`
`n.d.
`
`n.d.
`
`of 0.62 nM [35]. It should be noted that when apixaban was
`evaluated as an inhibitor of FXa versus the physiological
`substrate prothrombin in its prothrombinase state, non-
`competitive inhibition was observed. This finding is con-
`sistent with prothrombin binding being dictated primarily
`by interactions at exosites of FXa [36]. Apixaban also
`inhibits thrombus-associated FXa activity with a concen-
`tration causing 50% inhibition (IC50) of 1.3 nM [37]. In
`summary, apixaban is capable of inhibiting the activity of
`free FXa, thrombus-associated FXa and FXa within the
`prothrombinase complex. Apixaban is a direct inhibitor of
`FXa from rats, rabbits and dogs, with Ki values of 1.3, 0.16
`and 1.7 nM, respectively (Table 2 [15]). Previous studies
`involving other small molecule, direct FXa inhibitors have
`also reported a species difference in FXa inhibition among
`humans, rabbits, rats and dogs [29, 38, 39].
`
`In vitro pharmacodynamic studies
`
`To evaluate the in vitro pharmacodynamic activity of
`apixaban in human plasma, studies were undertaken to
`examine [1] thrombin generation, [2] anticoagulant activity
`and [3] platelet aggregation. By inhibiting FXa, apixaban
`prevents the conversion of prothrombin to thrombin,
`resulting in decreased generation of thrombin. Using the
`thrombogram method, apixaban was shown to inhibit tissue
`factor-initiated thrombin generation in human platelet-poor
`plasma in vitro. The IC50 of the rate of thrombin generation
`was 50 nM, and the IC50 for attenuation of the peak
`thrombin concentration was 100 nM [40].
`In human
`platelet-rich plasma, apixaban inhibited tissue factor-
`induced thrombin generation, as measured by the release of
`prothrombin fragment 1 ? 2, with an IC50 of 37 nM [41].
`As expected for an inhibitor of FXa, addition of apix-
`aban to normal human plasma prolonged clotting times,
`including activated partial
`thromboplastin time (aPTT),
`prothrombin time (PT), modified PT (mPT, using diluted
`PT reagent) and HepTest. Among the three clotting time
`assays, it appears that the mPT and HepTest are 10–20
`times more sensitive than aPTT and PT in monitoring the
`in vitro anticoagulant effect of apixaban in human plasma
`(Table 2 [15]). In both the PT and aPTT assays, apixaban
`
`123
`
`had the highest potency in human and rabbit plasma, but
`was less potent in rat and dog plasma, which parallels its
`inhibitory potencies (Ki) against human, rabbit, rat and dog
`FXa (Table 2 [15]).
`In the human platelet aggregation assay, apixaban had
`no direct effects on platelet aggregation response to ADP,
`collagen, c-thrombin, a-thrombin and TRAP [15, 41].
`However,
`it
`indirectly inhibited platelet aggregation
`induced by thrombin derived from tissue factor-mediated
`coagulation pathway, with an IC50 of 4 nM [41]. The potent
`indirect antiplatelet effect of apixaban, together with its
`direct antithrombotic and anticoagulant activity, suggests
`that apixaban may possess dual mechanisms to prevent and
`treat both venous (platelet-poor and fibrin-rich) and arterial
`(platelet-rich and fibrin-poor) thrombosis. It should be
`noted that
`the in vitro tissue factor model of platelet
`aggregation is a useful tool for evaluation of the anti-
`platelet mechanisms of action of anticoagulants. However,
`caution should be exercised as in vitro antiplatelet poten-
`cies of compounds obtained in this model may not directly
`translate into antithrombotic potencies in patients in whom
`multiple prothrombotic mechanisms, complications of
`cardiovascular disease and polypharmacy are common.
`
`In vivo pharmacology
`
`The non-clinical pharmacology of apixaban has been
`studied in vivo in rats and rabbits. Its in vivo effects were
`assessed over a comprehensive dose range in various
`well-established non-clinical models of thrombosis and
`hemostasis. These non-clinical models have been well
`characterized with standard antiplatelet agents and anti-
`coagulants, making them appropriate for evaluating the
`antithrombotic potential and bleeding liability of apixaban.
`
`Antithrombotic and bleeding time effects in rats
`
`Dose-dependent effects of apixaban were examined in a
`broad range of experimental models of thrombosis and
`hemostasis in rats [42]. Efficacy was evaluated using
`established models of thrombosis, including arterial-venous
`
`

`

`Preclinical discovery of apixaban
`
`483
`
`Table 3 Potency of apixaban in multiple thrombosis models
`IC50 (lM)b
`
`ID50 (mg/kg/h)b
`
`Species
`
`Modela
`
`Ratc
`
`Rabbitd
`
`AV-ST
`
`TF-VT
`
`FeCl2-VT
`FeCl2-AT
`AV-ST
`
`pDVT
`
`ECAT
`
`1.20
`
`1.55
`
`0.39
`
`0.72
`
`0.27
`
`0.11
`
`0.07
`
`5.71
`
`7.57
`
`1.84
`
`3.23
`
`0.36
`
`0.065
`
`0.11
`
`a Experimental models included arterial-venous shunt
`thrombosis
`tissue factor-stasis venous thrombosis (TF-VT), FeCl2-
`(AV-ST),
`induced vena cava thrombosis (FeCl2-VT), carotid artery thrombosis
`(FeCl2-AT), prevention model of deep vein thrombosis (pDVT) and
`electrically induced carotid arterial thrombosis (ECAT)
`b Potency for 50% decrease in thrombus weight was determined for
`concentration (IC50) and dose (ID50)
`c Data from Schumacher et al. [42]
`d Data from Wong et al. [7, 15]
`
`tissue factor-stasis venous
`thrombosis (AV-ST),
`shunt
`thrombosis, and FeCl2-induced vena cava thrombosis and
`carotid artery thrombosis. Hemostasis was assessed in
`models of cuticle bleeding time, renal cortex bleeding time
`and mesenteric bleeding time. Apixaban was given by a
`continuous intravenous (IV) infusion 1 h prior to the
`induction of thrombosis or bleeding.
`Apixaban at 0.1, 0.3, 1 and 3 mg/kg/h IV produced
`dose-dependent increases in ex vivo PT (1.24, 1.93, 2.75
`and 3.98 times control, respectively). In the various models
`of thrombosis, doses and plasma concentrations of apix-
`aban for 50% thrombus reduction ranged from 0.39 to
`1.55 mg/kg/h and 1.84 to 7.57 lM, respectively (Table 3)
`[42]. The 3 mg/kg/h dose of apixaban increased cuticle,
`renal and mesenteric bleeding times to 1.92, 2.13 and 2.98
`times control,
`respectively. Bleeding time was not
`increased by apixaban at 0.1 and 0.3 mg/kg/h in any model.
`The 1 mg/kg/h dose produced an increase in mesenteric
`bleeding time, but showed no effect on renal or cuticle
`bleeding time. In comparison, heparin increased renal and
`cuticle bleeding times to two times those of apixaban when
`given at a dose (300 U/kg plus 10 U/kg/min IV) that
`matched the efficacy of apixaban (3 mg/kg/h IV) in arterial
`thrombosis. These studies demonstrate that in rats, apix-
`aban has broad-spectrum antithrombotic efficacy and that
`these beneficial effects can be obtained at doses that show
`limited activity in multiple models of provoked bleeding.
`
`Antithrombotic and bleeding time effects in rabbits
`
`The antithrombotic efficacy of apixaban was evaluated in
`anesthetized rabbits using established models of thrombo-
`sis, including AV-ST, electrically induced carotid arterial
`
`thrombosis (ECAT) and DVT (a thread-induced vena cava
`thrombosis model). Hemostasis was assessed in a rabbit
`model of cuticle bleeding time. Apixaban was given by a
`continuous IV infusion 1 h prior to the induction of
`thrombosis or cuticle incision.
`
`Antithrombotic studies
`
`Apixaban exhibited strong antithrombotic activity in the
`rabbit models of AV-ST, ECAT and DVT, which com-
`pared well with standard antithrombotic agents (Figs. 3, 4)
`[7, 15, 43]. For instance, apixaban, the direct FXa inhibitor
`rivaroxaban, the direct thrombin inhibitor dabigatran and
`the oral anticoagulant warfarin showed similar efficacy in
`the prevention model of DVT (Fig. 3) [7, 15]. In the pre-
`vention model of ECAT, apixaban was as efficacious as the
`antiplatelet agent clopidogrel and warfarin (Fig. 4) [7, 15,
`43]. Doses and plasma concentrations of apixaban for 50%
`thrombus reduction ranged from 0.07 to 0.27 mg/kg/h and
`0.065 to 0.36 lM, respectively (Table 3) [15]. The 1 mg/
`kg/h dose was associated with approximately 80% anti-
`thrombotic efficacy in these models. Interestingly,
`the
`potency of apixaban in arterial and venous thrombosis
`prevention models was broadly equivalent. Apixaban also
`effectively inhibited the growth of a pre-formed intravas-
`cular thrombus in a treatment model of DVT, suggesting
`that apixaban shows potential for the treatment of estab-
`lished thrombosis [7].
`
`Bleeding time studies
`
`The bleeding potential of apixaban was compared with
`those of rivaroxaban, dabigatran and warfarin in the rabbit
`cuticle bleeding time model [7, 15]. At the highest effec-
`tive doses studied (each of which caused *80% inhibition
`of thrombus formation), warfarin increased bleeding time
`almost six-fold, whereas apixaban, rivaroxaban and da-
`bigatran prolonged bleeding time 1.13-, 1.9 and 4.4-fold,
`respectively (Fig. 3) [7, 15]. As shown in Fig. 3, the anti-
`thrombotic efficacy and bleeding profiles of warfarin and
`dabigatran were less favorable than those of apixaban and
`rivaroxaban. It should be noted; however, that extrapola-
`tion of pre-clinical bleeding time data to humans requires
`caution. Provoked bleeding measured in anaesthetized
`healthy animals may not directly translate into spontaneous
`bleeding observed in the clinical setting, where complica-
`tions of cardiovascular disease and polypharmacy are often
`present. Nevertheless, pre-clinical bleeding time studies are
`still useful for generating hypotheses for clinical investi-
`gation, for example by allowing the anti-haemostatic pro-
`files of experimental agents to be ranked and compared
`with those of established agents such as warfarin. The pre-
`clinical comparison of these agents’ therapeutic windows,
`
`123
`
`

`

`484
`
`P. C. Wong et al.
`
`Fig. 3 Plots of thrombus reduction and bleeding time versus dose in
`apixaban,
`rivaroxaban, dabigatran and warfarin-treated rabbits.
`Thrombus reduction, measured in the prevention model of venous
`thrombosis, was expressed as the percentage reduction in thrombus
`weight after treatment, relative to the mean vehicle thrombus weight.
`Bleeding time effect was expressed as a ratio of treated versus the
`mean vehicle value. Data are mean ± SE (n = 6 per group for the
`thrombosis and bleeding time studies and n = 12 per dose for plasma
`concentrations) (data from Wong et al. [7, 15]; reproduced with
`
`as summarized in Fig. 3, remains a hypothesis, and head-
`to-head clinical studies are required to validate these
`results.
`
`Combination therapy
`
`therapy with clopidogrel and aspirin
`Dual antiplatelet
`currently represents the standard of care for the reduction
`of atherothrombotic events in a broad range of patients. To
`understand the benefit-risk ratio of apixaban therapy in
`combination with standard antiplatelet therapy, apixaban
`was evaluated in combination with clinically relevant doses
`of aspirin and/or clopidogrel for the prevention of arterial
`thrombosis in rabbit models [44]. These evaluations
`showed that the triple combination of apixaban, aspirin and
`clopidogrel resulted in improved antithrombotic activity
`versus mono-therapies, without excessively increasing
`bleeding time in rabbits. Such data suggest that intensive
`
`permission). Reproduced from ‘‘Favorable therapeutic index of the
`direct factor Xa inhibitors, apixaban and rivaroxaban, compared with
`the thrombin inhibitor dabigatran in rabbits’’ published in ‘‘Journal of
`Thrombosis and Haemostasis’’ (2009), John Wiley and Sons; and
`from ‘‘Apixaban, an oral, direct and highly selective factor Xa
`inhibitor:
`in vitro, antithrombotic and antihemostatic studies’’
`published in ‘‘Journal of Thrombosis and Haemostasis’’ (2008), John
`Wiley and Sons
`
`antithrombotic therapy with apixaban, aspirin and clopi-
`dogrel may be a viable option for enhancing antithrombotic
`efficacy without unacceptable increases in bleeding.
`This hypothesis was tested in a large phase III study,
`APPRAISE-2, in high-risk patients with recent ACS trea-
`ted with apixaban or placebo in addition to mono (aspirin)
`or dual antiplatelet (aspirin plus clopidogrel) therapy. Very
`recently, the trial was discontinued based on ‘‘evidence of a
`clinically important increase in bleeding among patients
`randomized to apixaban, and this increase in bleeding was
`not offset by clinically meaningful reductions in ischemic
`events’’ [45]. The investigators of the APPRAISE-2 trial
`will continue to review the available data to better under-
`stand the effects of apixaban in this ACS patient population
`and will publish the results [45].
`As discussed above, the translatability of preclinical
`bleeding models to safety in clinical settings requires
`caution. It appears that the preclinical cuticle bleeding
`
`123
`
`

`

`Preclinical discovery of apixaban
`
`485
`
`Fig. 4 Dose-dependent effects of apixaban, clopidogrel and warfarin
`on integrated blood flow in the electrically induced carotid arterial
`thrombosis rabbit model. Data are mean ± SE (n = 6 per group,
`except n = 12 for clopidogrel). *P \ 0.05 versus the corresponding
`
`vehicle (V) (data from Wong et al. [15, 43]). Reproduced from
`‘‘Apixaban, an oral, direct and highly selective factor Xa inhibitor: in
`vitro, antithrombotic and antihemostatic studies’’ published in ‘‘Jour-
`nal of Thrombosis and Haemostasis’’ (2008), John Wiley and Sons.
`
`effect of apixaban in combination with dual antiplatelet
`therapy in rabbits does not translate directly into sponta-
`neous bleeding observed in the APPRAISE-2 trial. The
`underlying causes for this disconnect are not known, but
`may be related to species differences, bleeding time versus
`spontaneous bleeding, vascular bed differences, and the
`fact that unlike animal bleeding models, the APPRAISE-2
`patients had the highest tendency to bleed due to advanced
`age, diabetes, complications of cardiovascular disease,
`other comorbidities and the additive hazards of combina-
`tion antiplatelet treatment. Finally, the APPRAISE-2 find-
`ing does not mean that apixaban cannot benefit other
`patient populations, as recent phase III clinical trials of
`apixaban have demonstrated promising results in patients
`with venous thromboembolism (ADVANCE 1, 2, 3) and
`atrial fibrillation (AVERROES) [18, 21, 22, 46].
`
`Ex vivo coagulation markers
`
`The traditional clotting time tests for adjusting anticoagu-
`lant doses of heparin (aPTT) and warfarin (PT) are not
`sensitive for specific, single-target anticoagulants such as
`the FXa inhibitors. As shown in Fig. 5, apixaban only
`prolonged ex vivo aPTT and PT modestly, even at the
`highest dose that produced 80% antithrombotic efficacy in
`rabbits [7]. As expected from its mechanism of action,
`apixaban did not prolong thrombin time (TT). Among the
`clotting time tests, mPT was the most sensitive for apix-
`aban and tracked well with the antithrombotic activity of
`apixaban. Similar mPT results were also observed with
`
`Fig. 5 Plots of thrombus reduction (bar graph) and ex vivo clotting
`times (line graph) in apixaban-treated rabbits. Thrombus reduction,
`measured in the prevention model of venous thrombosis, was
`expressed as the percentage reduction in thrombus weight after
`treatment, relative to the mean vehicle thrombus weight (data from
`Fig. 3). For clarity, only mean data for thrombus reduction and the
`bolus dose (mg/kg) are shown. Activated partial thromboplastin time
`(aPTT), prothrombin time (PT), modified prothrombin time (mPT)
`and thrombin time (TT) were expressed as the treated/control ratio.
`Data are mean ± SE (n = 6 per group for thrombus reduction and
`n = 12 per group for clotting times) (data from Wong et al. [7];
`reproduced with permission). Reproduced from ‘‘Favorable thera-
`peutic index of
`the direct
`factor Xa inhibitors, apixaban and
`rivaroxaban, compared with the thrombin inhibitor dabigatran in
`rabbits’’ published in ‘‘Journal of Thrombosis and Haemostasis’’
`(2009), John Wiley and Sons
`
`123
`
`

`

`P. C. Wong et al.
`
`486
`
`Fig. 6 Ex vivo anti-FXa and
`anti-thrombin effects of
`apixaban in arterial thrombosis
`rabbits from Fig. 4 (top) and
`correlation of ex vivo anti-FXa
`with antithrombotic effects and
`plasma concentrations of
`apixaban in arterial thrombosis
`rabbits (bottom). *P \ 0.05,
`compared with the vehicle.
`Mean ± SE and n = 6 per
`group (data from Wong et al.
`[15]; reproduced with
`permission). Reproduced from
`‘‘Apixaban, an oral, direct and
`highly selective factor Xa
`inhibitor: in vitro,
`antithrombotic and
`antihemostatic studies’’
`published in ‘‘Journal of
`Thrombosis and Haemostasis’’
`(2008), John Wiley and Sons
`
`other FXa inhibitors such as rivaroxaban [7]. Data from a
`phase II study with apixaban show that the anti-FXa assay
`is more accurate and precise than the mPT test [47].
`Indeed, we also observed that the anti-FXa assay tracked
`well with antithrombotic activity in rabbits with arterial
`thrombosis [15]. As shown in Fig. 6, apixaban produced a
`dose-dependent
`inhibition of FXa and did not
`inhibit
`thrombin activity ex vivo [15]. The ex vivo anti-FXa
`activity of apixaban correlated well with both its anti-
`thrombotic activity and plasma concentration (Fig. 6).
`Thus,
`the anti-FXa activity assay may be suitable for
`monitoring the anticoagulant and plasma levels of apixaban
`if needed in certain situations such as an overdose, acute
`bleeding or urgent surgery.
`
`Drug metabolism and pharmacokinetics
`
`The metabolism and pharmacokinetics of apixaban have
`been studied extensively in animals and humans. In these
`studies, absorption of apixaban after oral administration
`was rapid, with a time to peak plasma concentration (Tmax)
`of 1–2 h. Absolute oral bioavailability of apixaban was
`good in rats, dogs and humans [48–50]. Following IV
`administration, apixaban was slowly eliminated in rats,
`
`

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