throbber
Journal of Thrombosis and Haemostasis, 6: 82(F829
`
`DOI: 10.1111/j.1538-7836.2008.02939.x
`
`ORIGINAL ARTICLE
`
`Apixaban, an oral, direct and highly selective factor Xa
`inhibitor: in vitro, antithrombotic and antihemostatic studies
`
`P. C. WONG, E. J. CRAIN, B. XIN, R. R. WEXLER, P. Y. S. LAM, D. J. PINTO, J. M. LUETTGEN and
`R. M. KNABB
`
`Thrombosis Research, Bristol—Myers Squibb Company, Pennington, NJ, USA
`
`To cite this article: Wong PC, Crain EJ, Xin B, Wexler RR, Larn PYS, Pinto DJ, Luettgen JM, Knabb RM. Apixaban, an oral, direct and highly
`selective factor Xa inhibitor: in vitro, antithrombotic and antihemostatic studies. J Thromb Haemost 2008; 6: 820-9.
`
`Summary. Background: Apixaban is an oral, direct and highly
`selective factor Xa (FXa) inhibitor in late-stage clinical devel-
`opment for the prevention and treatment of thromboembolic
`diseases. Objective: We evaluated the in vitro properties of
`apixaban and its in vivo activities in rabbit models of thrombosis
`and hemostasis. Methods: Studies were conducted in arteriove-
`
`thrombosis (AVST), venous thrombosis (VT),
`nous-shunt
`electrically mediated carotid arterial thrombosis (ECAT) and
`cuticle bleeding time (BT) models. Results: In vitro, apixaban is
`potent and selective, with a Ki of 0.08 nM for human FXa. It
`exhibited species diflerence in FXa inhibition [FXa K, (nM):
`0.16, rabbit; 1.3, rat; 1.7, dog] and anticoagulation [ECZX (uM,
`concentration required to double the prothrombin time): 3.6,
`hmnan; 2.3, rabbit; 7.9, rat; 6.7, dog]. Apixaban at 10 uM did
`not alter human and rabbit platelet aggregation to ADP, 'y-
`thrombin, and collagen. In vivo, the values for antithrombotic
`ED50 (dose that reduced thrombus weight or increased blood
`flow by 50% of the control) in AVST, VT and ECAT and the
`values for BT ED3x (dose that increased BT by 3-fold) were
`0.27 as 0.03, 0.11 J: 0.03, 0.07 3: 0.02 and > 3 mg kg‘1 h"
`i.v. for apixaban, 0.05 3: 0.01, 0.05 3: 0.01, 0.27 3: 0.08 and
`> 3 mg kg_1 h"l i.v. for the indirect FXa inhibitor fondapar-
`inux,
`and 0.53 d: 0.04,
`0.27 i 0.01,
`0.08 3: 0.01
`and
`0.70 3: 0.07 mg kg_1 day_1 p.o. for the oral anticoagulant
`warfarin, respectively. Conclusions: In summary, apixaban was
`effective in the prevention of experimental thrombosis at doses
`that preserve hemostasis in rabbits.
`
`Correspondence: Pancras C. Wong, Bristol-Myers Squibb Company,
`311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA.
`Tel.: +1 609 818 5572; fax: +1 609 818 7877.
`E-mail: pancras.wong@bms.com
`
`Presented in part at the 48th Annual Meeting of the American Society of
`Hematology, %12 December, 2006, Orlando, Florida (abstract 917) and at
`the XXI International Society on Thrombosis and Haemostasis Congress,
`&12 July 2007, Geneva, Switzerland (abstract no. P-W-656).
`
`Received 31 October 2007, accepted 15 February 2008
`
`factor Xa
`Keywords: apixaban, blood coagulation, direct
`inhibitor, hemostasis, oral anticoagulant, thrombosis.
`
`I ntroduction
`
`Thrombosis is a major cause of morbidity and mortality in the
`Western world and contributes to cardiovascular disorders.
`
`Despite recent advances in interventional and medical therapy
`for
`the prevention and treatment of a wide variety of
`thromboembolic events,
`the morbidity and mortality rates
`are still high [1]. Thus, there is a real unmet medical need for
`safer and more effective antithrombotic therapies. As thrombin
`plays a key role in the generation of an occlusive thrombus,
`inhibiting thrombin activity and/or its generation becomes a
`major drug target in the search of novel antithrombotic agents.
`Because thrombin is the last serine protease in the blood
`coagulation cascade that causes fibrin clot formation, the early
`research approach has been focused on the inhibition of
`thrombin activity by direct thrombin inhibitors [2]. Another
`approach is the inhibition of thrombin production by blocking
`the upstream proteases in the blood coagulation cascade, such
`as factor Xa (FXa). Experimental evidence suggests that FXa
`inhibitors have antithrombotic efficacy with lower bleeding risk
`in animals when compared with thrombin inhibitors [3,4].
`Thus, inhibition of FXa appears to be a promising mechanism
`for anticoagulant therapy, which prompted us to initiate a drug
`discovery program on small-molecule, direct FXa inhibitors
`[5,6].
`Based on years of research and development, we recently
`identified a direct FXa inhibitor, apixaban (BMS-562247),
`which is highly potent and selective with a K, of 0.08 nM for
`human FXa and with greater than 30 000-fold selectivity over
`other coagulation proteases [7]. Preliminary studies show that
`apixaban has high oral bioavailability in rats, dogs, and
`humans [8,9]. It prevented venous thromboembolic events with
`a favorable efficacy and safety profile in patients after knee
`replacement surgery [10]. Apixaban was also shown to be
`efficacious and safe in the treatment of patients with acute
`symptomatic deep vein thrombosis [11]. Currently, apixaban is
`in late-stage clinical development for the prevention and
`treatment of thromboembolic diseases.
`
`© 2008 International Society on Thrombosis and Haemostasis BMS 2002
`CFAD
`V. BMS
`IPR2015-01723
`
`

`
`In this study, we characterized the in vitro properties of
`apixaban and its antithrombotic profile for the prevention of
`thrombosis in rabbit models of arteriovenous—shunt thrombo-
`
`(AVST), venous thrombosis (VT) and electrolytically
`sis
`mediated carotid arterial thrombosis (ECAT), in comparison
`with the indirect FXa inhibitor fondaparinux and the oral
`anticoagulant warfarin. Antihemostatic effects of these agents
`were also studied in a well-characterized rabbit cuticle bleeding
`time (BT) model.
`
`Materials and methods
`
`Reagents
`
`The following drugs and chemicals were used in this study:
`chromogenic substrates S-2222, S-2238 and S-2765 from
`Chromogenix AB (distributed by DiaPhar1na Group, Inc.,
`West Chester, OH, USA); activated partial thromboplastin
`time (APTT) reagent Alexin from Trinity Biotech (St Louis,
`MO, USA); prothrombin time G’T) reagents Thromboplastin
`C Plus from Dade-Behring (Deerfield, IL, USA) or Thromb-
`max® with calcium from Sigma Chemical Co (St Louis, MO,
`USA); HepTest reagent from American Diagnostica (Stam-
`ford, CT, USA); human FXa from Haematologic Technolo-
`gies (Essex Junction, VT, USA); sodium warfarin, ADP and
`collagen from Sigma Chemical Co.; human 'y-thrombin from
`ICN Biomedicals, Inc. (Costa Mesa, CA, USA); fondaparinux
`(Arixtra®) from GlaxoSrr1ithKline (Research Triangle Park,
`NC, USA). Apixaban and DMP802 were synthesized at
`Bristol-Myers Squibb Company.
`
`Animals
`
`Studies were conducted in male New Zealand White rabbits
`
`weighing about 2-4 kg obtained from Covance (Denver, PA,
`USA). Experiments were conducted in accordance with the
`NIH Guide for the Care and Use of Laboratory Animals, and
`the regulations of the Animal Care and Use Committee of the
`Bristol-Myers Squibb Company.
`
`In vitro and ex vivo studies
`
`assays Using established protein purification
`Enzyme
`procedures, FX was isolated from citrated plasma obtained
`from healthy dogs, rats, and rabbits [12]. Purified FXa was
`obtained after activation with Russell’s viper venom followed
`by affinity chromatography. The resulting FXa was > 95%
`pure as judged by sodirun dodecylsulfate polyacrylamide gel
`electrophoresis. The
`substrate affinity values
`for FXa,
`expressed as the Michaelis—Menten—Henri constant (Km), for
`human, rabbit, rat and dog FXa were determined using the
`chromogenic substrate S-2765, and were 36, 60, 240 and 70 ]JM,
`respectively. The substrate hydrolysis was monitored by
`measuring absorbance at 405 nm at 25 °C for up to 30 min
`using a SpectraMax 384 Plus plate reader and SoftMax
`(Molecular Devices, Sunnyvale, CA, USA). FXa activity for
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`Apixaban in vitro/in vivo studies 821
`
`each substrate and inhibitor concentration pair was determined
`in duplicate. The Ki values were calculated by non-linear least-
`squares fitting of the steady-state substrate hydrolysis rates to
`the equation for competitive inhibition (Equation 1) using
`GRAFIT (Erithacus Software Ltd., Surrey, UK), where v
`equals reactions velocity in OD min_1, Vmax equals maxiumum
`reaction velocity, S equals substrate concentration, and I equals
`inhibitor concentration.
`
`v—#maxS
`Km (1 + Kii) + S
`
`(1)
`
`Blood samples were collected in tubes
`Clotting assays
`containing 1/10 volrune of 3.2% sodirun citrate, and platelet-
`poor plasma was obtained after centrifuging at > 2000 X g for
`10 1r1in. Clotting times were measured with an automated
`coagulation analyzer (Sysmex®, Dade Behring Inc., Deerfield,
`IL, USA). PT, APTT, and HepTest reagents were reconstituted
`and assays were performed according to the manufacturer’s
`instructions. The modified PT (mPT) assay was performed by
`diluting 1mL of thromboplastin C Plus with 1.25 mL of
`100 mM calcirun chloride and using this diluted reagent in place
`of the normal PT reagent. For PT and mPT, plasma (50 uL)
`was warmed to 37 °C for 3 min before adding PT reagent
`(100 uL). For APTT, plasma (50 uL) was warmed to 37 °C for
`1 min before adding APTT reagent (50 uL). After two more
`minutes, 25 mM calcirun chloride (50 uL) was added. For
`HepTest, plasma (50 IL) was warmed to 37 °C for 2 min
`before bovine FXa (50 uL) was added. After two more
`minutes, HepTest ReCal mix
`(50 uL) was
`added.
`Determinations were performed in duplicate and expressed as
`a mean ratio of treated vs. baseline control. The concentrations
`
`required to prolong clotting time by 2-fold (ECZX) were
`expressed as total plasma concentrations, not final assay
`concentrations after addition of clotting assay reagents. For
`in vitro studies, apixaban was serially diluted into citrated
`plasma obtained from healthy dogs,
`rats, and rabbits,
`beginning with a 10 mM dimethylsulfoxide stock solution.
`
`aggregation was
`Platelet
`aggregation assays
`Platelet
`measured in citrated hmnan and rabbit platelet-rich plasma
`G’RP) in vitro with a platelet aggregometer (Model PAP-4D,
`BioData, Horsham, PA, USA). PRP was obtained from
`citrated blood after centrifuging at 250 X g for 6 min. Citrated
`PRP (250 IL) was mixed with 20 uL of vehicle, DMP802 at
`3 ]J.M or apixaban at 1—10 [JM, and incubated for 3 min at
`37 °C. DMP802,
`a glycoprotein (GP)IIb/IIIa
`receptor
`antagonist, was included as a positive control (IC50 = 29 nM
`against human platelet aggregation response to 10 [JM ADP)
`[13]. Peak platelet aggregation was determined after
`the
`addition of 20 uL of the agonist (ADP at 10 ]JM, 'y-thrombin
`at 35 nM, and collagen at 10 pg mL", final concentration).
`
`and
`assays Anti-FXa
`antithrombin
`and
`Anti-FXa
`the
`determined
`using
`antithrombin
`activities
`were
`chromogenic substrates S-2222 and S-2238,
`respectively,
`based on the method of Sato et al.
`[14] with modifications
`
`

`
`822 P. C. Wong et al
`
`[15]. The hydrolysis of the chromogenic substrates was assayed
`by measuring absorbance at 405 nm at 37 °C as described
`above. Anti-FXa or antithrombin activity was calculated by
`comparing the values of optical density from samples taken in
`the post-treatment period to those taken in the pretreatment
`period.
`
`In vivo studies
`
`The rabbit AVST model, described by Wong
`A VST model
`et al. [16], was used in this study. Briefly, male New Zealand
`White rabbits were anesthetized with ketamine (50 mg kg"l
`i.m.) and xylazine (10 mg kg"1 i.m.), and their femoral artery,
`jugular vein and femoral vein were catheterized. These
`anesthetics were supplemented as needed. Thrombosis was
`induced by an arteriovenous (AW-shunt device containing a
`silk thread. Blood flowed from the femoral artery via the AV
`shunt into the opposite femoral vein for 40 min. The shunt was
`then disconnected and the silk thread covered with thrombus
`
`was weighed.
`As apixaban has an oral bioavailability of < 5% in rabbits
`(unpublished result), it was administered intravenously for
`in vivo studies. To achieve a stable plasma level with
`minimum experimental variability, apixaban, fondaparinux
`or vehicle was given by a continuous intravenous infusion 1 h
`prior
`to shunt placement. The infusion was continued
`throughout the experiment. Warfarin or vehicle was dosed
`orally once daily for 4 days. On the fourth day after the last
`oral dose of warfarin or vehicle, rabbits were anesthetized
`1.5 h later, and the treatment effect was evaluated about 2 h
`postdose. Arterial blood samples for the determination of
`clotting times or plasma levels were collected 20 min after
`shunt placement. Plasma levels of apixaban were measured by
`a specific and sensitive liquid chromatographic mass spec-
`trometry method (LC/MS/MS).
`In rabbits treated with
`apixaban,
`fondaparinux or warfarin,
`the antithrombotic
`effects of these agents were expressed as percentage inhibition
`of
`thrombus
`formation based on the treated vs.
`the
`
`corresponding mean vehicle. The ED50 value (dose that
`produced 50% inhibition of
`thrombus
`formation) was
`determined as described below.
`
`The apixaban group treatment consisted of vehicle (10%
`N,N-dimethylacetamide; 30% 1,2-propanediol; 60% water)
`(11 2 4), and apixaban (mg kg“ h“) at 0.03 (n 2 7), 0.1
`(n = 7), 0.3 (n = 7),
`1 (n = 7), and 3 (n = 3). The fonda-
`parinux group treatment consisted of vehicle (saline) (:1 = 6),
`and fondaparinux (mg kg—‘ h'1)at0.01(n 2 5),0.03(n 2 5),
`0.1 (n = 5), 0.3 (n = 5), and 1 (n = 5). The warfarin group
`treatment consisted of vehicle (water) (11 = 6), and warfarin
`(mg kg" day”) at 0.1 (n 2 6), 0.3 (n 2 6), 1 (n 2 6), and 3
`(n = 6).
`
`The rabbit VT model, described by Hollenbach
`VT model
`et al. [17], was used in this study with modifications. Briefly,
`rabbits were anesthetized as above. The left femoral vein was
`
`catheterized, using 11-crn IntraMedic polyethylene tubing
`
`(PE-200; Becton Dickinson, Sparks, MD, USA). A prosthetic
`device was passed through the PE-200 tubing into the
`abdominal vena cava. The prosthetic device consisted of a
`single
`strand of #10 awg braided copper wire (14 cm)
`terminated with eight pieces of 4-0 silk threads 3 cm in
`length. The silk threads were positioned in the abdominal
`vena cava by advancing the copper guide wire. Thrombi were
`formed on the silk threads in a time-dependent fashion.
`Apixaban and fondaparinux were given intravenously as
`described above 1 h prior to the placement of the prosthetic VT
`device. Warfarin or its vehicle was dosed orally once daily for
`4 days in rabbits as described above, and the prosthetic VT
`device was placed 2 h after the last oral dose. Ninety minutes
`after the placement of the prosthetic device, the abdominal
`vena cava was isolated through a midline abdominal incision.
`The vena cava was ligated just above and below the prosthetic
`device with 2-0 silk. The vena cava segment between the
`ligations was excised, and the threads with associated thrombus
`were removed, blotted twice on paper, and weighed. The
`weight of thrombus formed on the threads was calculated by
`subtracting the average weight of eight pieces of 4-0 silk threads
`3 cm in length. Clotting times of apixaban, fondaparinux and
`warfarin in plasma samples collected during VT were measured
`as above.
`
`In the VT study, the apixaban group treatment consisted of
`vehicle (10% N,N-dimethylacetamide; 90% of 5% dextrose)
`(n 2 6), and apixaban (mg kg" h“) at 0.03 (n 2 6), 0.1
`(n = 6), 0.3 (n = 6), and 1 (n = 6). The fondaparinux group
`treatment consisted of vehicle (saline) (:1 = 6), and fondapar-
`inux (mg kg‘1 h") at 0.01 (n = 6), 0.03 (n = 6), 0.1 (n = 6),
`and 0.3 (n = 6). The warfarin group treatment consisted of
`vehicle (water) (11 = 6), and warfarin (mg kg_1 day_1) at 0.1
`(n = 5), 0.3 (n = 5),
`1 (n I 5), and 3 (n = 6). The ED50
`value (dose that produced 50% inhibition of mean vehicle
`thrombus weight) was determined as described below.
`
`rabbit ECAT model,
`The
`thrombosis model
`Arterial
`[15], was used in this
`study.
`described by Wong et al.
`Briefly, male New Zealand White rabbits were anesthetized
`as above. An electromagnetic flow probe was placed on a
`segment of an isolated carotid artery to monitor blood flow.
`Thrombosis was induced by electrical stimulation of the
`carotid artery for 3 min at 4 mA, using an external stainless-
`steel bipolar electrode. Carotid blood flow was measured
`continuously over a 90-min period to monitor thrombosis-
`induced occlusion.
`Integrated carotid blood flow over
`90 min was measured by the area under the flow—ti1ne
`curve, calculated using the trapezoidal rule, and expressed as
`percentage of total control carotid blood flow, which would
`result
`if
`control
`blood
`flow had
`been maintained
`
`continuously for 90 min. The administration of apixaban
`and fondaparinux was initiated intravenously as described
`above 1 h prior to the artery injury. Warfarin or its vehicle
`was dosed orally once daily for 4 days in rabbits, and
`thrombosis was initiated 2 h after the last oral dose. Clotting
`times of
`apixaban,
`fondaparinux
`and warfarin,
`and
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`

`
`concentrations of apixaban in plasma samples, taken during
`electrically induced arterial
`thrombosis, were measured as
`above. In addition, we also measured ex vivo anti-FXa and
`antithrombin activities.
`
`Results
`
`In vitro studies
`
`Apixaban in vitro/in vivo studies 823
`
`The Lineweaver—Burk plot of inhibition of
`Enzyme assays
`human FXa by apixaban indicates
`that apixaban is a
`competitive inhibitor vs. the chromogenic peptide substrate
`S-2765, with a K of 0.08 nM (Fig. 1). Apixaban also inhibited
`FXa from rats, dogs, and rabbits (Table 1). In terms of FXa Ki
`at 25 °C, apixaban has similar potency in inhibiting hmnan and
`rabbit FXa, but is 10-20 times less potent against rat and dog
`FXa (Table 1).
`
`Clotting assays As expected for an inhibitor of FXa,
`addition of apixaban to normal hmnan plasma prolonged
`clotting times,
`including APTT, PT, mPT, 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 1). In both the PT and APTT assays,
`apixaban had the highest potency in hmnan and rabbit plasma,
`but was less potent in rat and dog plasma (Table 1).
`
`In this study, the apixaban group treatment consisted of
`vehicle (10% N,N-dimethylacetamide; 90% of 5% dextrose),
`and apixaban (mg kg_1h"l) at 0.01, 0.03, 0.1, 0.3, and 1
`(n = 6 per group). The fondaparinux group treatment
`consisted of vehicle (saline), and fondaparinux (mg kg‘1 h")
`at 0.1, 0.3, 1, and 3 (n = 6 per group). The warfarin group
`treatment consisted of vehicle (water) (n = 6), and warfarin
`(mg kg_1 day_1) at 0.03, 0.1, 0.3, 1, and 3 (n = 6 per group).
`The ED50 (dose that increased carotid blood flow to 50% of the
`control) of compounds and the EC50 (plasma concentration
`that increased carotid blood flow to 50% of the control) of
`apixaban were estimated as described below.
`
`The rabbit cuticle BT model [4] was
`Cuticle bleeding model
`used in this study. Briefly,
`rabbits were anesthetized as
`described above. A standard cut was made at the apex of the
`cuticle with a razor blade. Blood was allowed to flow freely by
`keeping the bleeding site in contact with 37 °C lactated Ringer’s
`solution. BT was defined as the time after transection when
`
`bleeding ceased. It was measured by averaging the bleeding
`time of three nail cuticles. The maximum bleeding recorded was
`20 n1in. Apixaban,
`fondaparinux
`and warfarin were
`administered as described above.
`In rabbits treated with
`
`the BT effect was expressed as a ratio of
`anticoagulants,
`treated vs.
`the mean vehicle value. The ED3X (dose that
`increased BT 3-fold) values of compounds were estimated as
`described below.
`
`5
`E
`V
`
`43
`0'1
`
`The apixaban group treatment consisted of vehicle (10%
`N,N-dimethylacetamide; 30% 1,2-propanediol; 60% water)
`(n = 6), and apixaban (mg kg‘1h") at
`1
`(n = 6) and 3
`(n = 6). The fondaparinux group treatment consisted of
`vehicle (saline) (n = 6), and fondaparinux (mg kg] h_1) at
`0.3 (n = 6),
`1 (n = 6), and 3 (n = 6). The warfarin group
`treatment consisted of vehicle (water) (n = 6), and warfarin
`(mg kg") at 0.1 (n : 5), 0.3 (n : 5),
`1
`(n : 5), and 3
`(n = 5).
`
`Statistical analysis
`
`The statistical analyses used were analysis of variance and the
`Student—Newman—Keuls test using the SAS system (SAS for
`Windows release 8.02A; Cary, NC, USA). ED50 doses were
`determined using the
`four-parameter
`logistic
`equation,
`y: A + [(3 — A)/(1 + [(C/x)D])], where A : minimiun
`y value, B = maximmn, C = log ED50 and D = slope fac-
`tor, and the logistic fit was analyzed by XLfit© (IDBS,
`Bridgewater, NJ, USA). Antithrombotic ED50 values were
`determined using a maximmn value of 100 and a minimiun
`value of zero, whereas BT ED3X values were determined using a
`maximmn value (BT treated/BT control) of seven and a
`minimiun value of one. A value of P < 0.05 was considered
`
`statistically significant. All data are means 3: SE.
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`0 no inhibitor
`o 0.25 nM
`A 0.5 nM
`A 1 nM
`
`n 2 nM
`:1 4 nM
`
`Ono inhibitor
`00.25 nM
`A0.5 nM
`A1 nM
`-2 nM
`D 4 nM
`
`
`
`100
`
`200
`[$2765] (|JM)
`
`300
`
`400
`
`5 ,
`
`/-\ 4 —
`
`8 _
`g
`E 3
`V
`93
`2 2
`
`_
`
`1 _
`
`,
`
`+
`
`*
`
`0
`
`0.02
`
`0.04
`
`0.06
`
`0.08
`
`1/[S2765] (pm-‘)
`
`Fig. 1. Plot of apixaban inhibition of human FXa activity at different
`concentrations of the chromogenic peptide substrate S-2765. Top panel:
`Untransformed data. The solid lines are from non-linear fits of the data to
`
`the equation for competitive inhibition with K; = 0.075 :: 0.0031 nM,
`S-2765 Km = 31 :: 1.4 uM, and maximum rate = 5.6 :: 0.062
`mOD min'1 (moD = OD X 1000 where OD is optical density).
`Bottom panel: Lineweaver—Burk plot of the data as in the top panel.
`
`

`
`824 P. C. Wong et al
`
`Table 1 In vitro potency (K) of apixaban against human, rabbit, rat and dog FXa and the concentrations (ECZX) required to double the prothrombin time
`(PT), modified prothrombin time (mPT), activated partial thromboplastin time (APTT) and HepTest in human, rabbit, dog and/or rat plasma
`
`Species
`
`Human
`Rabbit
`Rat
`Dog
`
`FXa K, (nM)
`
`PT ECZX (uM)
`
`mPT ECZX (W)
`
`APTT ECZX (uM)
`
`HepTest ECZX (um)
`
`0.081
`0.16
`1.3
`1.7
`
`0.002*
`0.01 *
`0.1
`0.2
`
`36*
`2.3
`7.9
`6.7
`
`0.37
`0.6
`ND
`ND
`
`7 .4*
`4.8
`20
`> 20
`
`0.4
`1.8
`ND
`ND
`
`*Data from Pinto et al. [8]. ND, not determined.
`
`In vitro platelet aggregation responses
`Platelet aggregation
`to ADP,
`'y-thrombin and collagen averaged 47 J: 5%,
`53 J: 4% and 51 J: 5%, respectively in human PRP, and
`50 J: 5%, 56 J: 5% and 60 J: 1%, respectively, in rabbit
`PRP. These platelet responses were not significantly changed
`by apixaban at 1, 3 and 10 ]JM, but were almost completely
`inhibited by the GPIIb/IIIa antagonist DMP802 at 3 [JM (data
`not shown).
`
`In vivo studies
`
`AVST model Mean thrombus weights in the different
`vehicle-treated AVST rabbits were similar, and ranged from
`290 J: 11 to 327 J: 15 mg (n
`6 per group). As shown in
`Fig. 2, apixaban, fondaparinux and warfarin were efficacious
`in
`the AVST rabbits
`and produced
`dose-dependent
`antithrombotic effects;
`their ED50 values are reported in
`Table 2. At their top doses studied in this model, apixaban at
`3 mg kg] h_1 i.v., fondaparinux at
`1 mg kg] h_1 i.v. and
`warfarin at 3 mg kg] day_l p. 0. reduced thrombus weight by
`98%,
`86% and 77%,
`respectively,
`relative
`to
`their
`corresponding vehicle group. We observed that apixaban at
`0.03, 0.1, 0.3, 1 and 3 mg kg” h"' i.v. produced linear dose-
`proportional increases in plasma levels of 34 J: 2, 121 J: 9,
`490 J: 104, 1155 J: 153 and 3705 J: 525 nM,
`respectively
`(11
`3-7 per group). The EC50 for apixaban was estimated
`to be 357 J: 90 nM.
`
`VT model Mean thrombus weights in the different vehicle-
`treated VT rabbits were similar, and ranged from 64 J: 2 to
`79 J: 7 mg (n = 6 per group). In this model, apixaban,
`fondaparinux
`and warfarin
`produced
`dose-dependent
`antithrombotic effects (Fig. 2); their ED50 values are given in
`Table 2. At their top doses studied in this model, apixaban at
`1 mg kg] h_1, fondaparinux at 0.3 mg kg_1 h"l and warfarin
`at 3 mg kg_1day"1 reduced thrombus formation by 83%,
`74% and 84%, respectively, relative to their corresponding
`vehicle group.
`
`350
`
`300
`
`Rabbit AVST
`
`
`
`Thrombusweight(mg)
`
`250
`
`200
`
`50
`
`150
`
`100
`
`>(ov-_:v:_»~—(o
`0.00O
`
`>v—_Cq-CO
`O0
`
`Apixaban
`
`Fondaparinux
`
`Warfarin
`
`Dose (mg kg‘1 h"‘ IV or mg kg‘1 d“ P0)
`
`80
`
`O)O
`
`
`
`Thrombusweight(mg) 4>O
`
`20
`
`>
`
`0.03
`
`".03.
`O0
`
`Apixaban
`
`“.03.
`CO
`
`0.01
`
`0.03
`
`0.003
`Fondaparinux
`
`Warfarin
`
`Dose (mg kg‘1 h“ IV or mg kg‘1 d‘1 PO)
`
`Figure 3 (top panel) shows the
`Arterial thrombosis model
`effects of vehicle and apixaban on carotid blood flow after
`electrical stimulation. Basal carotid blood flow in the vehicle-
`
`the
`averaged 21 J: 4 mL min—1. After
`treated animals
`initiation of thrombosis, blood flow was gradually decreased,
`and the artery was totally occluded in about 35 min in vehicle-
`treated animals. Apixaban at 0.01—1 mg kg"1 h_1 i.v. produced
`
`Fig. 2. Antithrombotic effects in the arteriovenous-shunt thrombosis
`(AVST) and venous thrombosis (VT) rabbit models. Top panel: Effects of
`apixaban, fondaparinux and warfarin on thrombus formation in AVST.
`Means :: SE, and n = }7 per group; *P < 0.05 vs. the corresponding
`vehicle. Preliminary AVST data for apixaban at 0.0}1 mg kg'1 h‘1 were
`reported in Pinto et al. [7]. Bottom panel: Effects of apixaban, fonda-
`parinux and warfarin on thrombus formation in VT. Mean :: SE, and
`n = 5-6 per group; *P < 0.05 vs. the corresponding vehicle.
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`

`
`Apixaban in vitro/in vivo studies 825
`
`Table 2 In vivo potency of apixaban, fondaparinux and warfarin in rabbit models of arteriovenous-shunt thrombosis (AVST), venous thrombosis (VT),
`electrically mediated carotid arterial thrombosis (ECAT) and bleeding time (BT)
`
`Compound
`
`Apixaban
`Fondaparinux
`Warfarin
`
`AVST* EDSJ
`
`0.27 : 0.03
`0.05 : 0.01
`0.53 : 0.04
`
`VT EDSJ
`
`: 0.03
`0.11
`0.05 : 0.01
`0.27 : 0.01
`
`ECAT EDSJ
`
`0.02
`0.07
`0.27 :: 0.08
`0.08
`0.01
`
`BT ED3XT
`
`> 3
`> 3
`0.70
`
`0.07
`
`Results are expressed as mean :: SE. *Preliminary AVST data for apixaban were reported in Pinto et al. [7]. ‘Expressed in mg kg” h" i.v. for
`apixaban and fondaparinux and in mg kg"1 day'1 p.o. for warfarin.
`
`100
`
`80
`
`60
`
`’‘§
`'5
`3
`2,
`
`E,
`

`3 40
`3-3
`
`Apixaban
`(mg kg"‘ h“ IV)
`
`-9- Vehicle
`
`-0- 0.01
`
`—E|- 0.03
`
`—I— 0.1
`
`fi 0.3
`
`—A— 1
`NO
`
`20
`
`Time after injury (min)
`
`9 8
`
`100
`
`CO0
`
`O)O
`
`-P0
`
`
`
`
`
`Integratedbloodflow(%control)
`
`V 0.010.030.1 0.3
`
`1
`
`V 0.1 0.3 1
`
`3
`
`V 0.03 0.1 0.3
`
`1
`
`3
`
`Apixaban
`
`Fondaparinux
`
`Warfarin
`
`Dose (mg kg“ h"‘ IV or mg kg“ d“ PO)
`
`Fig. 3. Antithrombotic effects in the rabbit model of electrically mediated carotid arterial thrombosis. Top panel: Effects of vehicle and apixaban
`on carotid blood flow (expressed as % of control carotid blood flow) after artery injury. Means :: SE, and n = 6 per group. Bottom panel: Dose-
`dependent effects of apixaban, fondaparinux and warfarin on integrated blood flow. Mean :: SE, and n = 6 per group. *P < 0.05 vs. the corresponding
`vehicle.
`
`a dose-dependent increase in duration of the patency of the
`injured artery. At 0.03—l mg kg_1 h"l
`i.v.,
`there was no
`occlusion in any of the animals up to 90 min.
`
`Figure 3 (bottom panel) also shows the effects of apixaban,
`fondaparinux and warfarin on integrated blood flow (ED50
`values are given in Table 2). We observed that apixaban at
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`

`
`826 P. C. Wong et al
`
`607 E] aP'|‘|'
`50*’ I PT
`
`*
`
`aPTTorPT(3)
`
`(A!C.’
`
`20‘
`
`10‘
`
`O
`
`>
`
`001
`
`003
`
`0.1
`
`03
`
`Apixaban
`
`Fondapari nux
`
`Dose (mg kg“ h“ IV or mg kg“ d" PO)
`
`Fig. 4. Ex vivo activated partial thromboplastin time (APTT) and prothrombin time (PT) effects of apixaban, fondaparinux and warfarin in rabbits.
`Means :: SE, and n = %19 per group. *P < 0.05 vs. the oorresponding vehicle.
`
`0.01, 0.03, 0.1, 0.3 and 1 mg kg_1 h_l i.v. produced linear dose-
`proportional increases in plasma levels of 13 J: 1, 45 :: 4,
`146 :: 8, 435 J: 25 and 1495 J: 50 nM, respectively (n = 6
`per group). The integrated blood flow EC50 for apixaban was
`estimated to be 106 J: 31 nM.
`
`Figure 4 shows the summary
`Ex vivo coagulation markers
`of the ex vivo APTT and PT responses
`to apixaban,
`fondaparinux and warfarin obtained from the AVST, VT
`and ECAT studies. Apixaban significantly prolonged ex vivo
`APTT at 3 mg kg] h_1, and PT at doses of 0.3 mg kg_1 h"l
`and higher (Fig. 4). Fondaparinux at the doses studied did not
`have significant effects on ex vivo APTT and PT. Warfarin
`significantly
`prolonged
`ex vivo APTT at
`doses
`of
`0.3 mg kg"1 day_1
`and higher,
`and PT at
`doses of
`0.1 mg kg"1 day_1 and higher (Fig. 4).
`Figure 5 (top panel) shows the ex vivo effect of apixaban on
`FXa and thrombin activity. Apixaban produced a dose-
`dependent inhibition of ex vivo FXa activity and did not
`change ex vivo thrombin activity. A good correlation was
`observed between the antithrombotic effect of apixaban and its
`ex vivo anti-FXa activity (r2 = 0.94), and also between the
`plasma
`concentrations
`and
`ex vivo
`anti-FXa
`activity
`(r2 = 0.90).
`
`Cuticle bleeding time model Mean cuticle BT in the vehicle-
`treated groups of apixaban, fondaparinux and warfarin were
`172 J: 2, 181 J: 7 and 183 J: 7 s, respectively (n = 6 per
`group). Warfarin at 0.1, 0.3,
`1 and 3 mg kg‘1day"1 p.o.
`produced dose-dependent
`increases
`in BT (228 J: 14,
`371 i 24, 929 J: 70 and 1129 J: 43 s, respectively), with an
`ED3X of 0.70 mg kg_1 day_1 (Table 2). As compared to
`vehicle, warfarin at 1 and 3 mg kg” day"' p.o. increased BT
`significantly by 5.1-fold and 6.2-fold, respectively (P < 0.05).
`
`In contrast, fondaparinux and apixaban at antithrombotic
`doses increased BT slightly (fondaparinux, 166 J: 4, 210 J: 12
`and 213 J: 11 s at 0.3, 1 and 3 mg kg] h_1 i.v., respectively;
`apixaban, 191 J: 8 and 228 J: 14 s at 1 and 3 mg kg” h"1
`i.v., respectively). At 3 mg kg‘1h"'
`i.v., fondaparinux and
`apixaban increased BT by 1.3-fold and 1.2-fold relative to their
`vehicle, respectively with ED3x of > 3 mg kg] h_1 for both
`compounds (Table 2).
`
`Discussion
`
`This study shows that apixaban is a potent, highly selective and
`direct inhibitor of FXa. It is also very efficacious for the
`prevention of arterial and venous thrombosis at doses that
`preserve hemostasis in rabbits.
`Analysis of enzyme kinetics shows that apixaban is a direct
`and competitive inhibitor of free hmnan FXa vs. a synthetic
`tripeptide substrate and does not need antithrombin III for its
`activity. It should be noted that apixaban is likely to behave as
`a mixed-type inhibitor of FXa activation of prothrombin in
`blood [18]. In addition, apixaban inhibited the prothrombin-
`ase-bound FXa activity in vitro, which resulted in the reduction
`of the conversion of prothrombin to thrombin with potency in
`the nanomolar range [18,19]. Together, these in vitro studies
`suggest that apixaban is a potent, selective, direct and effective
`inhibitor of both free and prothrombinase-bound FXa.
`Previously, we, as well as others, reported a species difference
`in FXa inhibition in hmnans, rabbits, rats and dogs with small-
`molecule, direct FXa inhibitors [6,20,21]. This study extends
`these findings, and shows that apixaban was more potent at
`prolonging APTT and PT in hmnan and rabbit plasma than in
`rat and dog plasma, which parallels its inhibitory potencies
`against human,
`rabbit,
`rat and dog FXa.
`Interestingly,
`apixaban was more potent in the prolongation of PT than
`
`© 2008 International Society on Thrombosis and Haemostasis
`
`

`
`FXa
`
`Thrombin
`
`Apixaban
`(mg kg"‘ h“ IV)
`
`100-
`
`- *
`
`Apixaban in vitro/in vivo studies 827
`
`sol
`
`c
`.9
`5 60*
`c
`E
`
`o\° 40
`
`20
`
`O_
`
`100
`
`so
`
`-5O
`
`N‘?
`
`0
`
`O
`
`
`
`
`
`
`
`Integratedbloodflow(O/0control)
`
`
`
`
`
`
`Apixabanplasmaconcentration(nM)
`100 1 0'
`
`E
`E
`E
`E
`E
`‘%'
`
`E
`
`E
`
`m
`
`01OOO
`
`1 000
`
`6*
`
`20
`
`40
`
`60
`
`80
`
`100
`
`EX vivo anti-FXa activity (%)
`
`0
`
`I
`l
`80
`60
`40
`20
`EX vivo anti-FXa activity (%)
`
`100
`
`Fig. 5. Ex vivo anti-FXa and antithrombin activities. Top panel: ex vivo anti-FXa and antithrombin effects of apixaban in arterial thrombosis rabbits from
`Fig. 3. Bottom panel: Correlation of ex vivo anti-FXa with antithrombotic effects and plasma concentrations of apixaban in arterial thrombosis rabbits.
`*P < 0.05, compared with the vehicle. Mean :: SE, and n = 6 per group.
`
`APTT in vitro and ex vivo. However, this effect may not extend
`to other direct FXa inhibitors, which may show varying effects
`on APTT and PT [3,4,2l]. Among the clotting assays studied,
`the mPT and HepTest have the highest sensitivity to apixaban
`in human plasma. Although the HepTest and mPT appear to
`be promising clotting assays to monitor the anticoagulant
`activity of apixaban, further clinical validation is needed.
`Because apixaban exhibited similar in vitro anti-FXa po-
`tency in humans and rabbits, we investigated its in vivo activity
`in well-characterized rabbit models of thrombosis and hemo-
`
`stasis. Although apixaban has hig

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