throbber
New Anticoagulants for Treatment of
`Venous Thromboembolism
`Jeffrey I. Weitz, MD, FRCP(C), FACP, FCCP
`
`Abstract—Treatment of venous thromboembolism (VTE) usually starts with concomitant administration of heparin or
`low-molecular-weight heparin (LMWH) and a vitamin K antagonist. The parenteral anticoagulant, which is given for
`at least 5 days, is stopped once the vitamin K antagonist produces a therapeutic level of anticoagulation. Although the
`introduction of LMWH has simplified the initial treatment of VTE, problems remain. LMWH must be given by daily
`subcutaneous (SC) injection and vitamin K antagonists require routine coagulation monitoring, which is inconvenient
`for patients and physicians. Recently, 3 new anticoagulants have been introduced in an attempt to overcome these
`limitations. These include fondaparinux and idraparinux, synthetic analogs of the pentasaccharide sequence that
`mediates the interaction of heparin and LMWH with antithrombin, and ximelagatran, an orally active inhibitor of
`thrombin. These agents produce a predictable anticoagulant response;
`thus, routine coagulation monitoring is
`unnecessary. Because they do not bind to platelets or platelet factor 4, fondaparinux and idraparinux do not cause
`heparin-induced thrombocytopenia (HIT). Unlike vitamin K antagonists, ximelagatran has a rapid onset of action,
`thereby obviating the need for concomitant administration of a parenteral anticoagulant when starting treatment. The
`lack of an antidote for these new agents is a drawback, particularly for idraparinux, which has a long half-life.
`(Circulation. 2004;110[suppl I]:I-19–I-26.)
`
`Key Words: venous thromboembolism 䡲 pulmonary embolism 䡲 deep venous thrombosis 䡲 anticoagulants
`䡲 coagulation 䡲 pharmacology 䡲 thrombosis
`
`Venous thromboembolism (VTE), which includes deep
`
`vein thrombosis (DVT) and pulmonary embolism (PE),
`occurs in ⬇1 in 1000 white individuals per year.1 The
`primary goal of initial treatment of VTE is to limit thrombus
`extension.2 This not only reduces the risk of PE, a complica-
`tion that can be fatal, but also may minimize postphlebitic
`syndrome, a potentially debilitating long-term sequelae of
`extensive DVT.3
`Anticoagulants remain the cornerstone of treatment of
`VTE. The landmark study by Barritt and Jordan4 established
`the role of anticoagulants in this setting. These investigators
`randomized 35 patients with clinically diagnosed PE to
`treatment with heparin or to no treatment. There were no
`fatalities in the heparin-treated group. In contrast, 25% of
`those untreated died of autopsy-proven PE.
`Rapid anticoagulation is necessary to minimize the risk of
`thrombus extension and PE in patients with VTE. This
`concept
`is supported by the placebo-controlled study of
`Brandjes et al,5 which randomized 120 patients with proximal
`DVT to treatment with heparin plus a vitamin K antagonist or
`to a vitamin K antagonist alone. The study was stopped
`prematurely because the rate of symptomatic recurrent VTE
`was lower in those given heparin plus a vitamin K antagonist
`than in those treated only with a vitamin K antagonist (6.7%
`
`and 20%, respectively; P⫽0.058), as was the rate of VTE
`extension (8.2% and 39.6%, respectively; P⬍0.001).
`With currently available drugs, rapid anticoagulation can
`only be effected with parenteral anticoagulants, such as
`heparin or LMWH. The introduction of LMWH has simpli-
`fied the initial treatment of VTE. Compared with heparin,
`LMWH exhibits greater bioavailability after SC injection, has
`a longer half-life, and produces a more predictable anticoag-
`ulant response.6 Consequently, LMWH can be given subcu-
`taneously once or twice daily without coagulation monitor-
`ing. Meta-analyses of trials comparing SC LMWH with
`continuous intravenous (IV) heparin for initial treatment of
`VTE demonstrate that LMWH is at least as effective and safe
`as heparin.7,8 Because LMWH is more convenient to admin-
`ister, however, it is ideally suited for outpatient treatment of
`VTE,9,10 an approach that reduces health care costs9 –12 and
`improves patient satisfaction.13
`After initial treatment with heparin or LMWH, ongoing
`anticoagulant therapy is needed to prevent recurrent VTE.14,15
`Extended therapy usually involves administration of a vita-
`min K antagonist,2 although long-term LMWH may be a
`better choice in cancer patients with VTE.16 A 3-month
`course of anticoagulant treatment is adequate for patients
`with VTE complicating a transient risk factor, such as
`surgery. In contrast, more extended therapy is needed in
`
`From McMaster University and Henderson Research Centre, Hamilton, Ontario, Canada.
`Correspondence to Dr Jeffrey I. Weitz, Henderson Research Centre, 711 Concession Street, Hamilton, Ontario, Canada, L8V 1C3. E-mail
`jweitz@thrombosis.hhscr.org
`© 2004 American Heart Association, Inc.
`Circulation is available at http://www.circulationaha.org
`
`DOI: 10.1161/01.CIR.0000140901.04538.ae
`
`I-19
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on January 15, 2018
`
`MYLAN - EXHIBIT 1043
`
`

`

`I-20
`
`Circulation
`
`August 31, 2004
`
`patients with unprovoked VTE.17–24 Long-term anticoagula-
`tion therapy is problematic because vitamin K antagonists are
`not easy to administer.25 Routine coagulation monitoring is
`essential to ensure that a therapeutic response is obtained
`because vitamin K antagonists have a narrow therapeutic
`window. Thus, a subtherapeutic response does not effectively
`reduce the risk of thrombosis, whereas excessive anticoagu-
`lation increases the risk of bleeding.25 Furthermore, interac-
`tions with a range of drugs can reduce or enhance the
`anticoagulant effects of vitamin K antagonists, as can variable
`intake of foods containing vitamin K. In addition, genetically
`determined polymorphisms in the cytochrome P4502C9 en-
`zyme influence the metabolism of vitamin K antagonists.26,27
`These phenomena contribute to the need for routine coagu-
`lation monitoring. Even when coagulation monitoring is
`performed, patients using vitamin K antagonists have a
`therapeutic anticoagulant response less than half the time,25 a
`situation that places them at risk for complications.
`Recently, 2 new parenteral and 1 novel oral anticoagulant
`have been evaluated in patients with VTE. Parenteral agents
`with longer half-lives than heparin or LMWH have the
`potential to simplify initial or extended treatment of VTE,
`whereas rapidly acting oral anticoagulants provide an oppor-
`tunity for streamlined therapy by eliminating the need for
`parenteral agents for initial VTE treatment and by obviating
`the requirement for coagulation monitoring and dose adjust-
`ment. Focusing on new drugs that have been evaluated in
`patients with VTE, this article (1) reviews their pharmacol-
`ogy, (2) outlines the potential advantages of these agents over
`existing anticoagulants, (3) describes the results of clinical
`trials evaluating new anticoagulants for treatment of VTE, (4)
`provides perspective as to strengths and potential drawbacks
`of these new agents, and (5) identifies their evolving role in
`VTE management.
`
`Pharmacology of New Anticoagulants
`The new anticoagulants that have been evaluated for the
`treatment of VTE include 2 parenteral antithrombin-
`dependent
`inhibitors of activated factor X (factor Xa),
`fondaparinux and idraparinux, and ximelagatran, the first oral
`direct thrombin inhibitor. By targeting factor Xa, fondapa-
`rinux and idraparinux block thrombin generation. In contrast,
`ximelagatran inhibits thrombin, the enzyme that catalyzes the
`conversion of fibrinogen to fibrin (Figure 1). Thrombin also
`activates platelets and amplifies its own generation by feed-
`back activation of factors VIII and V, key cofactors in factor
`Xa and thrombin generation, respectively. The pharmacology
`of fondaparinux,
`idraparinux, and ximelagatran is briefly
`discussed.
`
`Fondaparinux
`A synthetic analog of the antithrombin-binding pentasaccha-
`ride sequence found on heparin or LMWH (Figure 2),
`fondaparinux binds antithrombin with high affinity. Once
`bound, fondaparinux evokes conformational changes in the
`reactive center loop of antithrombin that enhance its reactiv-
`ity with factor Xa.28,29 Fondaparinux is a catalytic inhibitor;
`thus, after promoting the formation of the factor Xa/anti-
`thrombin complex, fondaparinux dissociates from antithrom-
`
`Figure 1. Sites of action of fondaparinux, idraparinux, and
`ximelagatran. Coagulation is initiated by the tissue factor/factor
`VIIa (TF/VIIa) complex, which activates factors IX and X. Acti-
`vated factor X (Xa) converts small amounts of prothrombin (II) to
`thrombin (IIa), which then feeds back to activate factors VIII and
`V, key cofactors in coagulation. The process is propagated by
`activated factor IX (IXa), which together with activated factor VIII
`(VIIIa) assembles on the surface of activated platelets to form
`intrinsic tenase, a complex that efficiently activates factor X. Xa,
`together with activated factor V (Va), assembles on the surface
`of activated platelets to form prothrombinase. This complex
`generates a burst of thrombin that, in the final stage of the
`coagulation process, converts fibrinogen to fibrin. By inhibiting
`factor Xa in an antithrombin-dependent fashion, fondaparinux
`and idraparinux block thrombin generation. In contrast, ximel-
`agatran undergoes biotransformation to melagatran, which then
`blocks thrombin activity by binding directly to the active site of
`the enzyme.
`
`bin and is available to activate additional antithrombin
`molecules.
`With excellent bioavailability after SC administration and
`a plasma half-life of 17 hours,30 fondaparinux is administered
`subcutaneously once daily. Systemic fondaparinux is ex-
`creted unchanged via the kidneys. Therefore, fondaparinux
`must be used with caution in patients with renal
`insufficiency.28,29
`When given SC in fixed doses, fondaparinux produces a
`predictable anticoagulant response. Consequently, routine
`coagulation monitoring is unnecessary. If necessary, fondapa-
`rinux can be monitored with antifactor Xa levels using a
`chromogenic assay.31 Fondaparinux has no effect on routine
`tests of coagulation, such as the activated partial thrombo-
`plastin time or activated clotting time.32
`
`Figure 2. Structures of fondaparinux and idraparinux. Fondapa-
`rinux is a synthetic analog of the natural pentasaccharide found
`in heparin and LMWH heparin. To enhance its affinity for anti-
`thrombin, idraparinux is 0-methylated and 0-sulfated.
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on January 15, 2018
`
`

`

`Weitz
`
`New Anticoagulants for VTE
`
`I-21
`
`Figure 3. Structures of ximelagatran and
`melagatran. Ximelagatran has ester and
`hydroxyl groups added to the carboxyl and
`amidine groups of melagatran. Bioconver-
`sion of ximelagatran to melagatran involves
`hydrolysis of the ester by esterases and
`reduction of the hydroxyl group. Intermedi-
`ates with one or the other of these protect-
`ing groups removed can be found tran-
`siently in the plasma.
`
`ximelagatran ingestion; melagatran circulates with a half-life
`of 3 hours in healthy volunteers and 4 to 5 hours in patients.
`Because melagatran has a relatively short half-life, ximelagat-
`ran is administered orally twice daily.34
`Ximelagatran produces a predictable anticoagulant re-
`sponse after fixed oral dosing. The half-life of melagatran
`does not vary among ethnic groups35 and is not influenced by
`obesity36 or mild to moderate hepatic insufficiency.37 Ximel-
`agatran does not interact with drugs that are metabolized by
`cytochrome P450 isozymes CYP2C19, CYP2C9, or
`CYP3A4.34
`At least 80% of systemic melagatran is eliminated un-
`changed via the kidneys. The half-life of melagatran is
`slightly prolonged in the elderly,
`likely reflecting their
`reduced creatinine clearance.38
`Coagulation monitoring is unnecessary for ximelagatran
`because it produces a predictable anticoagulant response.34
`Like all direct thrombin inhibitors, ximelagatran prolongs the
`activated partial thromboplastin time and international nor-
`malized ratio (INR). However, its effect on these tests is not
`dose-dependent and the extent of prolongation depends on the
`reagent used for testing. If monitoring is required, the ecarin
`clotting time, a test that has yet to be standardized, provides
`the best estimate of drug concentration.
`
`Potential Advantages of New Anticoagulants
`Fondaparinux was developed to replace heparin or LMWH
`for initial treatment of VTE, whereas idraparinux and ximel-
`agatran were designed to compete with vitamin K antago-
`nists. Because of their rapid onset of action, however,
`idraparinux and ximelagatran also may be useful for initial
`treatment of VTE, as well as for extended therapy.
`
`TABLE 1. Comparison of Fondaparinux and Idraparinux
`with LMWH
`
`Feature
`
`Target
`
`Fondaparinux
`
`Idraparinux
`
`LMWH
`
`Factor Xa
`
`Factor Xa
`
`Route of administration
`Half-life (h)
`Endothelial cell binding
`Protein binding
`Clearance
`HIT
`Antidote
`
`SC
`17
`None
`None
`Renal
`No
`None
`
`SC
`80
`None
`None
`Renal
`No
`None
`
`LMWH indicates low-molecular-weight heparin; SC, subcutaneous; HIT,
`heparin-induced thrombocytopenia.
`
`Factor Xa and
`thrombin
`SC
`4
`Some
`Some
`Renal
`Rare
`Partial neutralization
`with protamine sulfate
`
`Idraparinux
`A second-generation synthetic pentasaccharide, idraparinux
`is more negatively charged than fondaparinux (Figure 2).
`Consequently,
`idraparinux binds to antithrombin with an
`affinity higher than that of fondaparinux.33 Because it binds
`antithrombin so tightly, idraparinux has a plasma half-life
`similar to that of antithrombin, ⬇80 hours. With this long
`half-life, idraparinux is administered SC on a once-weekly
`basis. Like fondaparinux, idraparinux exhibits excellent bio-
`availability after SC injection and produces a predictable
`anticoagulant response, thereby obviating the need for routine
`coagulation monitoring.33
`
`Ximelagatran
`The first oral, direct thrombin inhibitor, ximelagatran is a
`prodrug of melagatran (Figure 3), a 429-dalton dipeptide that
`fits within the active-site cleft of thrombin and blocks the
`enzyme’s interactions with its substrates.34 Thus, melagatran
`is a stoichiometric inhibitor of thrombin that forms a revers-
`ible 1:1 complex with the enzyme. Melagatran exhibits poor
`bioavailability after oral administration. Ximelagatran, which
`has a molecular mass of 474, was developed to overcome this
`problem. With the addition of an ester and a hydroxyl group
`to the carboxyl and amidine groups of melagatran, respec-
`tively, ximelagatran is more lipophilic than melagatran.
`Consequently, ximelagatran is readily absorbed in the small
`intestine.34 Plasma levels of ximelagatran peak in the blood
`30 minutes after drug ingestion (Figure 4). Although ximel-
`agatran has no intrinsic anticoagulant activity, it is rapidly
`converted to melagatran via 2 intermediates. The bioavail-
`ability of melagatran after administration of oral ximelagatran
`is ⬇20%. Plasma levels of melagatran peak 2 hours after
`
`Figure 4. Plasma levels of ximelagatran and melagatran after
`administration of oral ximelagatran to healthy volunteers. Ximel-
`agatran levels in the blood peak 30 minutes after drug adminis-
`tration, whereas the levels of melagatran peak at 2 hours. Mel-
`agatran has a half-life of 3 hours in young healthy volunteers
`and 4 to 5 hours in patients.
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on January 15, 2018
`
`

`

`I-22
`
`Circulation
`
`August 31, 2004
`
`TABLE 2. Comparison of Ximelagatran with Vitamin K Antagonists
`
`Feature
`
`Target
`
`Route of administration
`Onset of action (h)
`Food interactions
`Drug interactions
`
`Therapeutic window
`Coagulation monitoring required
`Liver function monitoring required
`Antidote
`
`Ximelagatran
`
`Thrombin
`
`Oral
`2
`None
`None
`
`Wide
`No
`Yes
`None
`
`Vitamin K Antagonists
`
`Vitamin K-dependent clotting factors
`(factor VII, IX, X, and prothrombin)
`Oral
`72 to 96
`Affected by vitamin K content of diet
`Multiple drugs affect pharmacokinetics
`or pharmacodynamics
`Narrow
`Yes
`No
`Vitamin K produces slow reversal
`
`The new anticoagulants have potential advantages over
`existing agents. Focusing first on the parenteral agents, both
`fondaparinux and idraparinux have properties that distinguish
`them from LMWH (Table 1), the anticoagulant that is rapidly
`replacing heparin for initial VTE treatment. Whereas the
`bioavailability of LMWH after SC injection is 80% to 90%,6
`fondaparinux and idraparinux exhibit almost complete bio-
`availability.28,29,33 In addition, there is less between-patient
`variability in the anticoagulant response with fondaparinux
`and idraparinux than with LMWH. These differences reflect
`the fact that fondaparinux and idraparinux are homogeneous
`preparations of synthetic pentasaccharide units that bind only
`to antithrombin.39 In contrast, LMWH preparations are com-
`posed of heparin fragments that range in molecular weight
`from 1000 to 10 000 daltons. Only 15% to 20% of these
`fragments possess a pentasaccharide sequence.6 Longer
`chains can bind nonspecifically to endothelial cells and
`various plasma proteins.40,41 Pentasaccharide-independent
`binding of heparin chains to endothelial cells reduces bio-
`availability because these cellular binding sites must be
`saturated before the LMWH chains can enter the circulation
`and interact with antithrombin. Binding to plasma proteins,
`the levels of which differ between patients, explains the
`between-patient variability in the anticoagulant response to
`LMWH. Although fondaparinux and idraparinux have greater
`bioavailability than LMWH and produce a more predictable
`anticoagulant response, it is unclear whether these properties
`endow the new anticoagulants with clinical advantages over
`LMWH.
`Fondaparinux and idraparinux have half-lives of 17 and 80
`hours, respectively. In contrast, LMWH has a half-life of only
`4 to 5 hours. Despite its relatively short half-life, LMWH is
`effective when given once daily for VTE treatment, although
`uncertainty remains as to whether once-daily LMWH is as
`effective as twice-daily dosing.42 A potential explanation for
`the extended antithrombotic activity of LMWH is its capacity
`to induce the release of tissue factor pathway inhibitor (TFPI)
`from the vasculature.43,44 An endogenous anticoagulant, TFPI
`limits the initiation of coagulation by inhibiting tissue factor-
`bound factor VIIa in a factor Xa-dependent fashion.45 In
`contrast to LMWH, neither fondaparinux nor idraparinux
`induces TFPI
`release. However,
`the long half-lives of
`
`fondaparinux and idraparinux may preclude the need for this
`additional anticoagulant mechanism.
`Although LMWH can induce HIT, the risk of HIT is lower
`with LMWH than with heparin.46 In contrast, fondaparinux
`and idraparinux do not cause HIT because they do not bind to
`platelets or platelet factor 4 (PF4). Thus, HIT is triggered by
`antibodies directed against
`the heparin/PF4 complex.47
`Fondaparinux and idraparinux do not bind to platelets.
`Therefore, they do not cause platelet activation and subse-
`quent PF4 release. Likewise, because these agents do not bind
`to PF4, they do not induce the conformational changes in PF4
`that render it antigenic. These properties endow fondaparinux
`and idraparinux with a safety advantage over heparin and
`LMWH and may render these new agents useful for HIT
`treatment, a possibility that requires evaluation in clinical
`trials.
`Osteoporosis can occur after long-term treatment with
`heparin or LMWH.48,49 The risk of this complication should
`be lower with fondaparinux and idraparinux because shorter
`heparin chains cause less bone loss than longer chains in cell
`culture systems and in laboratory animal models.50,51 For
`extended treatment, therefore, fondaparinux and idraparinux
`may be safer than heparin or LMWH.
`As an orally active anticoagulant, ximelagatran has poten-
`tial advantages over vitamin K antagonists (Table 2). Vitamin
`K antagonists have a delayed onset of action. Thus, these
`agents act as anticoagulants by interfering with vitamin
`K-dependent, post-translational modification of the vitamin
`K-dependent clotting factors, an essential step in the synthesis
`of functional coagulation proteins.25 The antithrombotic ef-
`fect of vitamin K antagonists most probably reflects reduc-
`tions in the functional levels of prothrombin and factor X, a
`process that requires 4 to 5 days of treatment. Because of their
`slow onset of action, initial therapy with vitamin K antago-
`nists is usually supported by overlapping treatment with a
`parenteral anticoagulant in patients with established throm-
`bosis, or in those at high risk for thrombosis.2 In contrast,
`ximelagatran has a rapid onset of action, thereby obviating
`the need for concomitant administration of a parenteral
`anticoagulant when starting treatment.
`As mentioned earlier, vitamin K antagonists have a narrow
`therapeutic window. Furthermore, genetic differences in me-
`tabolism and multiple drug and food interactions affect the
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on January 15, 2018
`
`

`

`anticoagulant response to vitamin K antagonists.35 These
`factors contribute to the need for routine coagulation moni-
`toring, which is inconvenient for patients and physicians. In
`contrast, ximelagatran, with a wider therapeutic window than
`vitamin K antagonists and no food or drug interactions, can
`be given without coagulation monitoring.34 Building on their
`unique properties, fondaparinux, idraparinux, and ximelagat-
`ran have been compared with conventional anticoagulant
`regimens in VTE patients.
`
`Clinical Trials With New Anticoagulants
`Fondaparinux and ximelagatran have completed phase III
`clinical trials for treatment of VTE. Idraparinux has com-
`pleted phase II evaluation, and phase III clinical trials are
`underway. The results of these trials are discussed.
`
`Fondaparinux
`Fondaparinux has been evaluated for treatment of VTE in 2
`phase III clinical trials. In the MATISSE-DVT study, 2205
`patients with DVT were randomized,
`in a double-blind
`fashion, to receive either fondaparinux (in dosages of 5, 7.5,
`or 10 mg SC once daily depending on body weight) or
`enoxaparin (1 mg/kg SC twice daily) for at least 5 days,
`followed by a minimum of a 3-month course of treatment
`with a vitamin K antagonist.52 At 3 months, rates of recurrent
`symptomatic VTE with fondaparinux or enoxaparin were
`3.9% and 4.1%, respectively, whereas rates of major bleeding
`were 1.1% and 1.2%, respectively.
`In the open-label MATISSE PE trial, 2213 patients with PE
`were randomized to receive either fondaparinux (in dosages
`of 5, 7.5, or 10 mg SC once daily depending on body weight)
`or heparin (by continuous IV infusion) for 5 days, followed
`by a minimum of a 3-month course of therapy with a vitamin
`K antagonist.53 At 3 months, rates of recurrent symptomatic
`VTE with fondaparinux or heparin were 3.8% and 5.0%,
`respectively, whereas rates of major bleeding were 1.3% and
`1.1%, respectively. The results of the MATISSE studies
`suggest
`that fondaparinux is as effective as LMWH or
`unfractionated heparin for initial VTE treatment. Further-
`more, fondaparinux is easier to administer than unfraction-
`ated heparin.
`
`Idraparinux
`In a phase II, dose-finding trial, idraparinux was compared
`with warfarin for treatment of patients with proximal DVT.54
`In this trial, 659 patients were treated for 5 to 7 days with
`enoxaparin and then randomized to receive once-weekly SC
`idraparinux (2.5, 5.0, 7.5, or 10 mg) or warfarin for 12 weeks.
`The primary efficacy end point, changes in compression
`ultrasound and perfusion lung scan findings, was similar in all
`idraparinux groups and did not differ from that in the warfarin
`group. There was a clear dose response for major bleeding in
`patients given idraparinux with an unacceptably high rate of
`major bleeding in those given the 10-mg dose. Two patients,
`both of whom were in the 5-mg idraparinux group, experi-
`enced fatal bleeds. Patients given the lowest dose of idrapa-
`rinux had less bleeding than those randomized to warfarin
`(P⫽0.029). Based on these data, ongoing phase III trials are
`comparing SC idraparinux monotherapy (at a dose of 2.5 mg
`
`Weitz
`
`New Anticoagulants for VTE
`
`I-23
`
`SC once weekly) with 5 to 7 days of enoxaparin, followed by
`at least 3 months of warfarin therapy for treatment of patients
`with DVT or PE.
`
`Ximelagatran
`The double-blind, phase III THRIVE treatment study ran-
`domized 2491 patients with acute DVT to receive either oral
`ximelagatran (36 mg twice daily) alone for 6 months or
`enoxaparin (1 mg/kg SC twice daily for a minimum of 5
`days), followed by warfarin (target INR of 2.0 to 3.0) for 6
`months.55 The primary end point, objectively documented
`recurrent VTE, occurred in 2.1% and 2.0% of patients given
`ximelagatran and enoxaparin/warfarin, respectively. Rates of
`major bleeding were similar in the ximelagatran and enox-
`aparin/warfarin groups (1.3% and 2.2%, respectively), as
`were the all-cause mortality rates (2.3% and 3.4%, respec-
`tively; P⫽NS). These results suggest that oral ximelagatran is
`as effective as conventional anticoagulant
`treatment with
`enoxaparin followed by warfarin in preventing recurrent VTE
`in patients with acute DVT. However, unlike enoxaparin or
`dalteparin, ximelagatran can be given orally, and in contrast
`to warfarin, ximelagatran does not require coagulation mon-
`itoring or dose adjustment. Consequently, ximelagatran has
`the potential to streamline treatment.
`Ximelagatran also has been evaluated for the long-term
`secondary prevention of recurrent thrombosis in patients with
`VTE. An open-label, phase III study (THRIVE III) random-
`ized 1233 patients who had completed a 6-month course of
`anticoagulant therapy for treatment of VTE to oral ximelagat-
`ran (24 mg twice daily) or placebo for an additional 18
`months.56 The primary end point, objectively documented
`recurrent VTE, occurred in 2.8% of patients given ximelagat-
`ran and in 12.6% of those randomized to placebo (hazard
`ratio 0.16; P⬍0.001). Major bleeding rates were similar
`between the ximelagatran and placebo groups (1.1% versus
`1.3%, respectively; hazard ratio 1.16), and there were no fatal
`or intracranial bleeds. Hence, data from this study indicate
`that
`lower-dosage ximelagatran (24 mg twice daily) can
`effectively prevent recurrent VTE.
`
`Potential Role of New Anticoagulants in
`VTE Treatment
`The introduction of LMWH provided a major advance in
`initial VTE treatment. With once- or twice-daily SC admin-
`istration and no need for coagulation monitoring, LMWH
`paved the way for out-of-hospital treatment. New anticoagu-
`lants have the potential to further streamline VTE treatment
`and to offer potential safety advantages over existing agents.
`Based on the results of
`the MATISSE-DVT and
`MATISSE-PE trials,52,53 fondaparinux can be used in place of
`LMWH or heparin for initial treatment of VTE. Although
`more convenient to administer than heparin, like LMWH,
`fondaparinux must be given daily by SC injection. However,
`fondaparinux may be safer than heparin or LMWH because it
`does not cause HIT.
`Idraparinux and ximelagatran have the potential to be used
`as sole therapy for VTE treatment. In the case of idraparinux,
`a single SC injection at the time of VTE diagnosis circum-
`vents the need for daily SC injections of LMWH for the initial
`
`Downloaded from
`
`http://circ.ahajournals.org/
`
` by guest on January 15, 2018
`
`

`

`I-24
`
`Circulation
`
`August 31, 2004
`
`5 to 7 days of treatment. Thereafter, once-weekly SC idrapa-
`rinux can be used in place of a vitamin K antagonist. With no
`need for routine coagulation monitoring and dose adjustment,
`idraparinux is more convenient to administer than vitamin K
`antagonists. Although promising, the efficacy and safety of
`idraparinux for VTE treatment remains to be established in
`the ongoing phase III trials.
`Like idraparinux, ximelagatran also can be used for both
`initial and extended VTE treatment. Ximelagatran has the
`advantage of oral bioavailability, thereby obviating the need
`for SC injections, and in contrast to vitamin K antagonists,
`ximelagatran does not require coagulation monitoring or
`dosage adjustment. Based on the results of the THRIVE
`treatment trial,55 ximelagatran alone (at a dosage of 36 mg
`twice daily) appears to be as effective and safe as conven-
`tional treatment with LMWH followed by warfarin for DVT
`treatment.
`The data from the THRIVE III study56 indicate that a
`lower-dosage ximelagatran regimen (24 mg twice daily)
`prevents recurrent VTE. This study highlights the fact that
`patients with unprovoked VTE have a rate of recurrent VTE
`of 7% to 10% per year when anticoagulant
`therapy is
`stopped.17–24 When given in dosages sufficient to produce a
`target INR of 2.0 to 3.0, warfarin reduces this risk by ⬎90%,
`but is associated with an average rate of major bleeding of 2%
`per year, of which 10% are fatal.17–24 A lower-intensity
`warfarin regimen (target INR 1.5 to 1.9) is less effective at
`preventing recurrent VTE than usual-intensity warfarin (tar-
`get INR 2.0 to 3.0) in this setting and does not appear to
`reduce the risk of bleeding.57 Although effective, warfarin is
`inconvenient because of the need for routine coagulation
`monitoring and dosage adjustment. Ximelagatran has the
`potential to overcome this barrier.
`
`Potential Limitations of New Anticoagulants
`What are the drawbacks of these new anticoagulants? Like all
`anticoagulants, the major side effect of these new drugs is
`bleeding. To counteract this problem, a safe rapidly-acting
`antidote is desirable. Unfortunately, none of these new agents
`has an antidote. When rapid reversal is required because of
`major bleeding or the need for urgent surgery, the lack of an
`antidote is more problematic for agents with a long half-life,
`such as idraparinux, than for those with a short half-life, such
`as ximelagatran. Although procoagulants, including recombi-
`nant factor VIIa, may reverse the anticoagulant effects of
`fondaparinux,
`idraparinux, or ximelagatran in animals or
`healthy volunteers,58 – 61
`the effect of
`these agents on
`anticoagulant-induced bleeding has yet
`to be addressed.
`Furthermore, recombinant factor VIIa is not available in all
`hospitals and the drug is expensive, particularly if repeated
`doses are needed. Although not well studied, dialysis is likely
`to clear melagatran, but not fondaparinux or idraparinux.
`A side effect unique to ximelagatran is elevation of liver
`enzymes. Overall, ⬇6% (range, 5% to 10%) of patients
`treated with long-term ximelagatran have a transient increase
`in alanine aminotransferase levels to over 3 times the upper
`limit of normal. Typically, this occurs between 1 and 6
`months after the start of treatment. Concomitant elevation of
`bilirubin levels is found in only 0.4% of patients. The
`
`increase in serum alanine aminotransferase is generally
`asymptomatic and reversible, regardless of whether ximel-
`agatran treatment is continued or stopped. Although this
`phenomenon appears to be benign, more data on patients
`treated long-term are needed. Until this information is avail-
`able, ximelagatran will need to be restricted to patients with
`normal or near normal hepatic function at baseline. Further-
`more, it is likely that testing of liver function will need to be
`performed when initiating ximelagatran treatment and during
`the first 6 months of therapy. Although less problematic than
`the routine coagulation monitoring and dosage adjustments
`needed with vitamin K antagonists, the requirement for liver
`function test monitoring may limit
`the convenience of
`ximelagatran.
`Cost is a consideration with any new drug. Fondaparinux is
`more expensive than LMWH, and idraparinux and ximelagat-
`ran are likely to cost more than vitamin K antagonists, even
`with their attendant cost of coagulation monitoring. There-
`fore, cost-effectiveness analyses will be required to determine
`the extent to which these new treatments reduce health care
`costs. Patient convenience also deserves consideration. By
`obviating the need for routine coagulation monitoring, agents
`such as idraparinux or ximelagatran can improve the quality
`of life for individuals with limited access to anticoagulation
`clinics.
`Finally, compliance with these new drugs may be difficult
`to assess in the absence of coagulation monitoring. Attention
`to packaging and ongoing supervision of patients will help
`minimize this problem.
`
`Conclusions and Future Directions
`New anticoagulants have the potential to further refine VTE
`treatment. Still unknown, however, is the role of these agents
`in selected high-risk VTE patients, such as those with cancer,
`and their safety in pregnancy. With its short half-life and oral
`bioavailability, ximelagatran could prove more convenient
`than LMWH in cancer patients with VTE, provided that its
`safety can be established in those with hepatic metastases and
`abnormal liver function tests. Fondaparinux also may prove
`useful
`in this setting, although its longer half-life may
`complicate timing of invasive procedures.
`The safety of these n

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