Skip to main content

Advertisement

ADVERTISEMENT

Direct Thrombin Inhibitors (Part 1 of 2)

Umesh K. Arora, and Meeney Dhir
January 2005
Thrombin plays a central role in clot formation through platelet activation and fibrin generation at the sites of arterial disruption, such as that caused by angioplasty or plaque rupture.1–3 Thrombin also amplifies its own generation by activating factors VIII and V, key cofactors in the activation of factor X and pro-thrombin, respectively, as well as Factor XI. Anticoagulant strategies to inhibit arterial thrombogenesis have therefore focused on inhibiting thrombin or preventing thrombin generation.4 Heparin in combination with aspirin has been a cornerstone of treatment in conditions characterized by arterial plaque disruption, acute coronary syndromes and percutaneous coronary intervention (PCI). Heparin acts as an anticoagulant by activating anti-thrombin, which in turn inhibits thrombin and activated Factor X (Factor Xa). However, heparin has a number of limitations that are not observed with direct thrombin inhibitors. This article will review the role of thrombin in arterial thrombogenesis, discuss the mechanism of action and the limitations of heparin, an indirect thrombin inhibitor, provide an overview of the direct thrombin inhibitors and review the current literature on the use of direct thrombin antagonists in acute coronary syndromes and in the setting of percutaneous coronary intervention. Role of thrombin in arterial thrombogenesis. Disruption of atherosclerotic plaque exposes blood to thrombogenic substances in the lipid-rich core (Figure 1). Thereafter, platelets adhere to subendothelial matrix components, such as collagen and von Willebrand factor, become activated, and contribute to thrombus formation.5 Following activation, platelets express increased amounts of anionic phospholipids on their surface which promote coagulation by supporting the assembly of coagulation factor complexes (the so-called prothrombinase and tenase complexes). Upon activation, platelets, release agonists, such as adenosine diphosphate and thromboxane A2, which allow adherent platelets to recruit additional platelets to sites of arterial injury. Coincident with platelet adhesion and activation, contact of blood with tissue factor expressed by activated macrophages and smooth muscle cells found in the core of the disrupted plaque results in activation of the coagulation system.1-3 Exposed tissue factor binds to activated Factor VII, and this complex in turn activates Factors IX and X.5 In the presence of calcium, Factors IXa and Xa bind to their cofactors on the surface of activated platelets (Factors VIIIa and Va, respectively) to form intrinsic tenase and prothrombinase complexes, respectively. Activation of Factor X by intrinsic tenase, and subsequent conversion of prothrombin to thrombin by prothrombinase, results in a burst of thrombin generation. Thrombin converts fibrinogen to fibrin, which forms a matrix for thrombus. During this process, thrombin binds to fibrin where it remains enzymatically active and protected from inactivation by fluid-phase inhibitors.6 By serving as a reservoir of active thrombin, the clot is intensely thrombogenic because thrombin bound to fibrin locally activates platelets,7 amplifies coagulation by activating Factors V and VIII,8 and converts fibrinogen to fibrin.6 In addition, fibrin-bound thrombin activates Factor XIII, which renders the thrombus more resistant to lysis both by cross-linking fibrin and by cross-linking 2-antiplasmin, the major plasmin inhibitor, onto fibrin. Fibrin-bound thrombin may also activate a latent carboxypeptidase B, known as thrombin activatable fibrinolysis inhibitor (TAFI).9 Once activated, this enzyme attenuates fibrinolysis, presumably by removing carboxy-terminal lysine residues from fibrin, thereby preventing the binding of plasminogen and plasmin.10 Additional sources of thrombin are induced during coronary thrombolysis. Plasmin is generated during pharmacologic thrombolysis and it promotes clotting by activating contact factors,11 transiently activating Factor V,12 and directly convert prothrombin to thrombin.13,14 Furthermore, lysis of coronary thrombi exposes additional fibrin-bound thrombin,15 and thrombin remains bound to soluble fibrin degradation products where it also is protected from inactivation by fluid-phase inhibitors.16 Limitations of Unfractionated Heparin and LMWH A. Heterogenous mixture of active and inactive factors. Heparin inhibits arterial thrombosis by activating antithrombin, which then inhibits thrombin and Factor Xa and IIa.17 Binding of heparin to antithrombin produces conformational changes in antithrombin that accelerates the rate at which it inhibits Factor Xa. Heparin also enhances thrombin inhibition by antithrombin through an additional mechanism: heparin binds simultaneously to both thrombin and antithrombin, thereby maintaining physical proximity of the two molecules. Heparin induces the Xa inducibility property of antithrombin through a pentasaccaride sequence known as the “essential pentasaccaride.” However, heparin induced inactivation of thrombin requires the presence of an 18 saccharide sequence. Since almost all the heparin chains are at least 18 saccharides in length, heparin has equivalent inhibitory activity against thrombin and Factor Xa.17 In contrast, with a mean molecular mass of about 5,000 Da, fewer than half the low-molecular-weight heparin chains are long enough to bridge thrombin to antithrombin.18 Because heparin catalysis of Factor Xa does not require bridging between Factor Xa and antithrombin, these smaller chains retain their ability to promote Factor Xa inhibition. Consequently, low-molecular-weight heparins have greater inhibitory activity against Factor Xa than thrombin.18B. Heparin is unable to inactivate fibrin-bound thrombin and platelet-bound Factor Xa. Although the heparins are capable of inactivating fluid-phase thrombin, they do not inhibit thrombin bound to fibrin5,19 or fibrin degradation products.15 In addition, Factor Xa bound to the surface of activated platelets is also resistant to inactivation by the heparin/antithrombin complex,20,21 as fibrin bound Xa remains active and continues to participate in thrombin generation, thereby increasing the amount of thrombin available to bind to fibrin. As a result, heparin is relatively an inefficient inhibitor of thrombus propagation. Thrombin binds to fibrin via exosite 1, a substrate-binding site that is distinct from its active site.22–24 Current evidence suggests that fibrin-bound thrombin is protected from inactivation by the heparin/antithrombin complex because heparin binds simultaneously to fibrin and to the heparin-binding domain on thrombin, so-called exosite 2, thereby approximating the two molecules and promoting thrombin’s activity within a clot.25 The formation of a ternary heparin/thrombin/fibrin complex increases the affinity of thrombin for fibrin and induces conformational changes in the active site of the enzyme.26,27 The resistance of thrombin within the ternary complex to inactivation by fluid-phase inhibitors may reflect allosteric modulation of its active site or spatial constraints that impair the reactivity of inhibitors with thrombin. Considerably less is known about the molecular basis for the resistance of platelet-bound Factor Xa to inactivation by the heparin/ antithrombin complex, but a similar mechanism is likely. C. Heparin binds to endothelium and to plasma proteins. The binding of heparin to cells and plasma proteins such as vitronectin, fibronectin, histidine-rich glycoprotein, platelet Factor IV, and high-molecular-weight multimers of von Willebrand factor, in a pentasaccharide-independent fashion28,29 decreases its anticoagulant effect by limiting its availability to interact with antithrombin.30 This effect differs between patients because some heparin-binding proteins, such as vitronectin and fibronectin, are acute-phase reactants and can increase to a variable extent in acute illness.31,32 In addition, platelet Factor 4, a highly cationic protein that binds heparin with high affinity,28 is released from platelets during the clotting process. High-molecular-weight multimers of von Willebrand factor also are released from both activated platelets and endothelial cells during clotting. Because of its nonspecific binding to endothelium and plasma proteins, the half-life of heparin is dose dependent and its anticoagulant effect varies from patient to patient. Consequently, careful laboratory monitoring is necessary to ensure that an adequate anticoagulant effect is obtained. D. Heparin causes thrombocytopenia and paradoxical thrombosis. Thrombocytopenia induced by heparin typically appears five or more days after the start of heparin therapy. The thrombocytopenia is caused by heparin-dependent IgG antibodies that activate platelets through their Fc receptors. Recently, several laboratories have shown that these antibodies recognize a complex of heparin with platelet Factor 4. Paradoxically, thrombotic complications develop in many patients with heparin-induced thrombocytopenia, because of both in vivo platelet activation and the generation of procoagulant rich platelet derived microparticles. When heparin-induced thrombocytopenia is defined as a decrease in the platelet count to below 150,000 per cubic millimeter, previous studies have noted heparin induced thrombocytopenia in as many as 2–7%33,34 of patients receiving unfractionated heparin with most of the patients having one or more thrombotic events (venous more arterial). Most patients with heparin-induced thrombocytopenia (HIT) require alternative anticoagulation. This is because HIT is highly prothrombotic and is characterized by markedly increased thrombin generation. Unfractionated heparins seem to induce HIT more often than low molecular weight heparins. There are three anticoagulants for which there is an emerging consensus for their efficacy in management of HIT, and which are currently approved for treatment of HIT in several countries: the recombinant hirudin, lepirudin, a direct thrombin inhibitor; the synthetic direct thrombin inhibitor, argatroban; and the heparinoid, danaparoid sodium, mainly exhibiting antifactor-Xa activity. Hirudin, the drug for which most data from prospective trials exists, can be safely and effectively used in patients with HIT, its dramatically increased elimination half-life in patients with renal failure being the most important drawback. Argatroban, which is mainly eliminated by the liver, could be used preferentially in such patients with renal impairment. Interference with the international normalized ratio makes oral anticoagulation, which is necessary in many patients with HIT, problematic. Activated partial thromboplastin time is sufficient to monitor lepirudin and argatroban treatment in most cases. Danaparoid sodium, with an antifactor-X activity half-life of about 24 hours seems to be best suited for thrombosis prophylaxis in patients with HIT. In some patients monitoring by determining antifactor-Xa activity is necessary. No antidote is available for any of the drugs discussed, and bleeding complications are the most important adverse effects. In situations such as hemodialysis or cardiopulmonary bypass, not only the characteristics of the drug in use itself, but also availability of monitoring methods play an important role. Adjunctive treatments have not been systematically evaluated and should be used cautiously. Recent data suggest that re-exposure of patients with a history of HIT with heparin, for example during cardiopulmonary bypass, can be well tolerated provided no circulating HIT antibodies are detectable at the time of re-exposure, and heparin is strictly avoided pre- and post-operatively. These limitations of heparin in the treatment of arterial thrombosis have prompted the development of new antithrombotic agents that are capable of inactivating fibrin-bound thrombin. Of these new drugs, the most extensively studied to date are direct thrombin inhibitors. The Different Direct Thrombin Antagonists Direct thrombin inhibitors include hirudin, semisynthetic hirudin fragments (bivalirudin), small molecules that react with the active-site of thrombin (including PPACK and its derivatives, argatroban and others) and thrombin-binding DNA aptamers (Figure 2). Although all of these inhibitors bind directly to thrombin, their sites and modes of interaction are different (Figure 2). Two different sites on thrombin mediate substrate interactions: the active site and the exosite 1.35 Cleavage of the sessile bond in substrates occurs at the active site, whereas exosite 1 serves to dock substrates in the proper orientation. Direct thrombin inhibitors interact with one or both of these sites and inactivate thrombin by preventing it from interacting with its substrates. A. Hirudin and its derivatives. Hirudin is the prototype of direct thrombin antagonists. Hirudin is a 65 amino acid polypeptide originally isolated from the salivary glands of the medicinal leech, Hirudo medicinalis, and is now produced through recombinant DNA technology.36 Unlike native hirudin, the recombinant forms are not sulfated at the Tyr 63 site and exhibit at least a 10-fold reduced affinity for thrombin. Hirudin is a potent and specific inhibitor of thrombin which forms a 1:1 stoichiometric, slowly reversible complex with the enzyme. Analysis of the crystal structure of the thrombin-hirudin complex demonstrates the extensive contact that hirudin makes with thrombin with its globular amino-terminal “head” region interacting with the active site of thrombin and the carboxy-terminal “tail” binding to exosite 1 on the enzyme.37 Hirugen is a synthetic dodecapeptide comprising residues 53 to 64 of the carboxy-terminal region of hirudin.38 Sulfation of the tyrosine residue at position 63 increases the affinity of hirugen for thrombin. By binding to thrombin on exosite 1, hirugen blocks the enzyme’s interaction with its substrates, including fibrinogen and the thrombin receptor on platelets. Because it does not interact with the active site of thrombin, however, hirugen does not block hydrolysis of low-molecular-weight thrombin-directed substrates. Bivalirudin was formed by adding D-Phe-Pro-Arg-Pro-(Gly)4 to the amino-terminal of hirugen. This converted a weak competitive inhibitor of thrombin (hirugen) to a potent bivalent inhibitor, bivalirudin. Like hirugen, bivalirudin interacts with both the active site and exosite 1 on thrombin.39 However, bivalirudin differs from hirudin in that it produces only transient inhibition of the active site of thrombin. Once complexed with thrombin, the Arg-Pro bond on the amino-terminal extension of bivalirudin is cleaved, thereby converting bivalirudin into a lower affinity, hirugen-like inhibitor.40,41 This lower affinity binding may confer a better safety profile upon bivalirudin. B. Covalent inhibitors (D-Phe-Pro-ArgCH2Cl (PPACK) and its derivatives).42–44 PPACK is the prototype of a class of inhibitors that form covalent complexes with thrombin. PPACK interacts with the active site of thrombin and inhibits the enzyme by alkylating the active center histidine. Recent crystallographic studies confirm the tight interaction of this molecule with thrombin and its covalent derivatization of His-57 and Ser-195 within the catalytic triad. Boroarginine derivative of PPACK, D-Phe-Pro-Arg-borate, has been developed and appears to be a slightly more specific inhibitor of thrombin than the parent molecule. Although this agent has antithrombotic activity in laboratory animal models and has the potential for oral bioavailability, crossreactivity with other serine proteases has stopped further development. C. Non-covalent inhibitors or active site inhibitors.42-44 A number of low-molecular-weight active site inhibitors of thrombin have been developed. The prototype of these noncovalent inhibitors is argatroban.45 Others include napsagatran,46 inogatran,47 melagatran,48 and L372,460. Argatroban is a small (527 Da) DTI that binds reversibly to the active site of the thrombin molecule. It is selective for thrombin (inhibition constant 0.04 µmol/L), and has little effect on related serine proteases. Argatroban is intravenously administered and has an elimination half-life of 40 to 50 minutes. Monitoring of the aPTT is required to assess its anticoagulant activity. Dose adjustments may be required to attain a steady-state aPTT of 1.5 to 3 times the mean normal value. Argatroban is hepatically metabolized via hydroxylation and aromatization reactions, so dosing reductions and careful monitoring are recommended in patients with hepatic dysfunction. The metabolized products are removed via biliary excretion. Renal impairment has no influence on the elimination half-life of argatroban and dose adjustments therefore are unnecessary in patients with renal dysfunction. Argatroban has no known antidote. Although it is a potent inhibitor of thrombin in vitro, results with argatroban in laboratory animals and in humans with acute coronary syndromes have been mixed. Argatroban has been used successfully in patients with heparin-induced thrombocytopenia. Napsagatran, inogatran, melagatran, and Ximelagatran are other examples of low-molecular-weight noncovalent thrombin inhibitors. All are potent and selective inhibitors of thrombin. There are at least 10 oral direct thrombin inhibitors (DTIs) in clinical development, out of them Ximelagatran (Exanta) and BIBR 1048 are most promising. Both are prodrugs with two protecting groups that are eliminated after absorption from the gastrointestinal tract. Their main active substances, melagatran and BIBR 953, are both potent and selective DTIs. Following oral administration, Ximelagatran is rapidly absorbed and converted to melagatran, the active form, achieving peak plasma concentrations in 1.6 to 1.9 hours. Melagatran is a potent, reversibly binding, active-site inhibitor of thrombin. After ximelagatran administration, melagatran has been shown to inhibit the generation of thrombin, probably because of reduced thrombin-mediated positive feedback to the coagulation system. In experimental models of thrombosis, melagatran has been shown to have a shallower dose-response curve than warfarin and, therefore, a better separation between efficacy and bleeding. Oral bioavailability, measured as the plasma concentration of the active metabolite, seems to be higher for ximelagatran (20%) than for BIBR 1048 (estimated to 5%). BIBR 953 has a longer half-life (about 12 hours) than does melagatran (3 to 5 hours) after oral administration of BIBR 1048 and ximelagatran, respectively. Both melagatran and BIBR 953 are mainly eliminated via the renal route. The variability of the plasma concentration of melagatran after oral administration of ximelagatran is low. There are no clinically relevant interactions with food or cytochrome P450 metabolized drugs and ximelagatran. In clinical studies, ximelagatran has been administered in a twice daily fixed-dose regimen without coagulation monitoring. Results of published clinical studies are encouraging, both with regard to efficacy and bleeding. Major indications in Phase III studies with ximelagatran are the prevention of venous thromboembolism (VTE) in hip and knee replacement surgery, treatment and long-term secondary prevention of VTE and prevention of stroke in patients with nonvalvular atrial fibrillation. It is anticipated that with a favorable outcome of the Phase III clinical studies new oral DTIs, with the oral fixed-dose regimen without routine coagulation monitoring, will ease the use of today’s anticoagulant therapy. D. DNA aptamers. A single-stranded, 15-nucleotide DNA aptamer49 has been developed that binds to exosite 1 on thrombin with high affinity. Like hirugen, the aptamer blocks thrombin’s interaction with its substrates but does not inhibit thrombin-mediated hydrolysis of low-molecular-weight synthetic substrates. The DNA aptamer is a potent anticoagulant in vitro and is an effective antithrombotic in laboratory animals. However, with a half-life of minutes, its clinical utility is limited. Recently, DNA aptamers that bind to exosite 2 on thrombin also have been identified. Because there is allosteric linkage between the two exosites on thrombin, DNA aptamer binding to exosite 2 can influence ligand binding to exosite 1. Exosite 2 aptamers have yet to be tested in laboratory animals, but are likely to have short half-lives in vivo. Despite their different mechanisms of action, all direct thrombin inhibitors have potential advantages over heparin. Consequently, researchers have assumed that results with one agent will be obtained with any drug in this class. This concept is problematic because even agents that interact with the same sites on thrombin may have different benefit-to-risk profiles. For example, closer inspection of clinical trials results suggests that bivalirudin is safer than hirudin, a phenomenon that may be explained by differences in the pharmacokinetic profiles of the 2 drugs. Advantages of Direct Thrombin Inhibitors Over Heparin Although unfractionated heparin is widely used for thrombin inhibition in the management of unstable coronary artery disease, clinical and experimental evidence suggests that it is suboptimal (Table 1). Recent pharmaceutical strategies to improve upon unfractionated heparin’s efficacy profile have centered on the development of 2 major classifications of thrombin inhibition medications: the naturally occurring leech protein hirudin (and synthetic analogs) and low-molecular-weight (LMW) heparins. Direct thrombin inhibitors have both biologic and pharmacokinetic advantages over heparin. The biologic advantage of direct thrombin inhibitors reflects their capacity to inactivate fibrin-bound thrombin. Because thrombin binds to fibrin via exosite 1, the activity of agents that interact with the active site is not compromised when the enzyme binds to fibrin. Hirudin and bivalirudin bind to exosite 1, as well as the active site of thrombin, and consequently compete with fibrin for access to exosite 1. However, in the absence of heparin, thrombin has only weak affinity for fibrin25 so that these agents readily interact with fibrin-bound thrombin. Unlike heparin, direct thrombin inhibitors do not bind to plasma proteins or endothelial cells. Hirudin forms a slowly reversible complex with thrombin and has a plasma half-life of 40 minutes after intravenous administration, and approximately 120 minutes after subcutaneous injection. Bivalirudin has a plasma half-life of 24 minutes after intravenous infusion. Unlike hirudin, bivalirudin produces only transient inhibition of the active site of thrombin and may, therefore, be safer. Furthermore, they produce a more predictable anticoagulant response than unfractionated heparin.50,51 Theoretically, this pharmacokinetic advantage of direct thrombin inhibitors could obviate the need for laboratory monitoring. The narrow therapeutic window for hirudin, however, makes monitoring necessary.52–54 Comparison of Direct Thrombin Inhibitors (Bivalirudin Versus Hirudin) Bivalirudin has both safety and potential efficacy advantages over hirudin. Bivalirudin appears to have a wider therapeutic window than hirudin, possibly reflecting the fact that bivalirudin produces only transient inhibition of the active site of thrombin. The wider safety margin of bivalirudin relative to hirudin permits administration of higher doses. This may provide bivalirudin with an efficacy advantage over hirudin because, as stoichiometric inhibitors of thrombin, high doses of these drugs must be given to inactivate the large amounts of thrombin generated in the vicinity of an arterial thrombus. Bivalirudin complexes and inhibits thrombin generated at sites of arterial injury. By the time these enzyme/inhibitor complexes reach the microcirculation, where thrombomodulin is more abundant,58 release of the amino-terminal domain of bivalirudin leaves only the carboxy-terminal segment bound to exosite 1 on thrombin. In contrast to bivalirudin, hirudin forms a very slowly reversible complex with thrombin and prevents the enzyme from binding to thrombomodulin. Despite the promise of direct thrombin inhibitors, clinical trials with hirudin as an adjunct to thrombolytic therapy61,62 or in patients undergoing coronary angioplasty63 have failed to show durable advantages over heparin. This likely reflects the fact that the high doses of hirudin needed for the treatment of arterial thrombosis cannot be given safely, particularly when hirudin is used in conjunction with thrombolytic agents. This concept is supported by the fact that hirudin is better than low-molecular-weight heparin for the prevention of venous thrombosis in common high-risk orthopedic patients,64 a setting where lower, and safer, doses of hirudin are effective. With its wider therapeutic window, bivalirudin may be better than hirudin because it can be given in the doses necessary to block arterial thrombogenesis. This possibility deserves testing in well-designed phase III trials, as does bivalirudin’s potential for promoting protein C activation. Differences between bivalirudin and hirudin, as well as other direct thrombin inhibitors, highlight the pitfalls of considering all direct thrombin inhibitors to have equivalent risk-benefit profiles. Part 2 of this article will be continued in the February 2005 issue.
1. Fuster V, Badimon L, Badimon JJ, et al. The pathogenesis of coronary artery disease and the acute coronary syndromes. N Engl J Med 1992;326:242–250. 2. Fuster V, Badimon L, Badimon JJ, et al. The pathogenesis of coronary artery disease and the acute coronary syndromes. II. N Engl J Med 1991;326:310–318. 3. Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation 1995;92: 657–671. 4. Fuster V, BadimonJJ, Chesebro JH, et al. Plaque rupture, thrombosis, and therapeutic implications. Hemostasis 1996;26(Suppl 4):269–284. 5. Furie B and Furie BC. Molecular and cellular biology of blood coagulation. N Engl J Med 1992;326:800–806. 6. Weitz JI, Hudoba M, Massel D, et al. Clot-bound thrombin is protected from inhibition by heparin-antithrombin III but is susceptible to inactivation by antithrombin III-independent inhibitors. J Clin Invest 1990;86:385–391. 7. Kumar R, Béguin S, Hemker C. The effect of fibrin clots and clot-bound thrombin on the development of platelet procoagulant activity. Thromb Haemost 1995;74:962–968. 8. Kumar R, Béguin S, Hemker HC, The influence of fibrinogen and fibrin on thrombin generation evidence for feedback activation of the clotting system by clot bound thrombin. Thromb Haemost 1994;72:713–721. 9. Bajzar L, Manuel R, Nesheim ME. Purification and characterization of TAFI, a thrombin-activable fibrinolysis inhibitor. J Biol Chem 1995;270:14477–14784. 10. Sakharov DV, Plow EF, Rijken DC, On the mechanism of the antifibrinolytic activity of plasma carboxypeptidase B. J Biol Chem 1997;272:14477–14482. 11. Ewald GA, Eisenberg PR. Plasmin-mediated activation of contact system in response to pharmacological thrombolysis. Circulation 1995;91:28–36. 12. Lee CD, Mann KG. Activation/inactivation of human factor V by plasmin. Blood 1989;73:185–190. 13. Eisenberg PR, Miletich JP, Sobel BE, et al. Differential effects of activation of prothrombin by streptokinase compared with urokinase and tissue-type plasminogen activator (t-PA). Thromb Res 1988;50:707–717. 14. Seitz R, Pelzer H, Immel A, et al. Prothrombin activation by thrombolytic agents. Fibrinolysis 1993;7:109–115. 15. Mirshahi M, Soria J, Soria C, et al. Evaluation of the inhibition by heparin and hirudin of coagulation activation during rt-PA-induced thrombolysis. Blood 1989;74:1025–1030. 16. Weitz JI, Leslie B, Hudoba M. Thrombin binds to soluble fibrin degradation products where it is protected from inhibition by heparin-antithrombin but susceptible to inactivation by antithrombin-independent inhibitors. Circulation 1998;97:544–552. 17. Hirsh J. Heparin. N Engl J Med 1991;324:1565–1574. 18. Weitz JI. Low-molecular-weight heparins. N Engl J Med 1997;37:688–698. 19. Hogg PJ, Jackson CM. Fibrin monomer protects thrombin from inactivation by heparin-antithrombin III: Implications for heparin efficacy. Proc Natl Acad Sci USA 1989;86:3619–3623. 20. Teitel JM, Rosenberg RD. Protection of factor Xa from neutralization by the heparin-antithrombin complex. J Clin Invest 1983;71:1383–1391. 21. Pieters J, Willems G, Hemker HC, et al. Lindhout, Inhibition of factor IXa and factor Xa by antithrombin III/heparin during factor X activation. J Biol Chem 1988;263:15313–15318. 22. Wilner GD, Danitz MP, Mudd MS, et al. Selective immobilization of -thrombin by surface-bound fibrin. J Lab Clin Med 1981;97:403–411. 23. Berliner LJ, Sugawara Y, Fenton JW,II. Human -thrombin binding to nonpolymerized fibrin-Sepharose: Evidence for an anionic binding region. Biochemistry 1985;27:7005–7009. 24. Kaminski M, McDonagh J. Inhibited thrombins. Interactions with fibrinogen and fibrin. Biochem J 1987;242:881–887. 25. Hogg PJ, Bock PE. Modulation of thrombin and heparin activities by fibrin. Thromb Haemost 1997;77:424–433. 26. Hogg PJ, Jackson CM. Formation of a ternary complex between thrombin, fibrin monomer, and heparin influences the action of thrombin on its substrates. J Biol Chem 1990;265:248–255. 27. Hogg PJ, Jackson CM, Labanowski JK, et al. Binding of fibrin monomer and heparin to thrombin in a ternary complex alters the environment of the thrombin catalytic site, reduces affinity for hirudin, and inhibits cleavage of fibrinogen. J Biol Chem 1996;271:26,088–26,095. 28. Lane DA. Heparin binding and neutralizing proteins. In: D.A. Lane and U. Lindahl, editors. Heparin: Chemical and Biological Properties and Clinical Applications Boca Raton: CRC Press, 1989, pp.363–391. 30. de Romeuf C, Mazurier. Heparin binding assay of von Willebrand factor (vWF) in plasma milieu: Evidence of the importance of the multimerization degree of vWF. Thromb Haemost 1993;69:436–440. 30. Young E, Prins M, Levine MN, et al. Heparin binding to plasma proteins, an important mechanism for heparin resistance. Thromb Haemost 1992;67:639–643. 31. Young E, Wells P, Holloway S, et al. Ex-vivo and in-vitro evidence that low molecular weight heparins exhibit less binding to plasma proteins than unfractionated heparin. Thromb Haemost 1994;71:300–304. 32. Young E, Cosmi B, Weitz J, et al. Comparison of the non-specific binding of unfractionated heparin and low molecular weight heparin (enoxaparin) to plasma proteins. Thromb Haemost 1993;70:625–630. 33. Warkentin TE, Levine MN, Hirsh J, et al. Heparin-induced thrombocytopenia in patients treated with low-molecular-weight heparin or unfractionated heparin. N Engl J Med 1995;332:1330–1335. 34. Singer RL, Mannion JD, Bauer TL, et al. Complications from heparin-induced thrombocytopenia in patients undergoing cardiopulmonary bypass. Chest 1993;104:1436–1440. 35. Stubbs MT, Bode W. A player of many parts: The spotlight falls on thrombin’s structure. Thromb Res 1993;69:1–58. 36. Markwardt F. Past, present, and future of hirudin. Haemostasis 1991;21(Suppl 1): 11–26. 37. Rydel TJ, Ravichandran KG, Tulinsky A, et al. The structure of a complex of recombinant hirudin and human-thrombin. Science 1990;249:277–280. 38. Maraganore JM, Bourdon P, Jablonski J, et al. Design and characterization of hirulogs: A novel class of bivalent peptide inhibitors of thrombin. Biochemistry 1990;29:7095–7101. 39. Skrzypczak-Jankun E, Carperos VE, Ravichandran KG, et al. Structure of the hirugen and hirulog 1 complexes of -thrombin. J Mol Biol 1991;221:1379–1393. 40. Witting JI, Bourdon P, Brezniak DV, et al. Thrombin-specific inhibition by and slow cleavage of hirulog-1. Biochem J 1992;283:737–743. 41. Parry MA, Maraganore JM, Stone SR. Kinetic mechanism for the interaction of Hirulog with thrombin. Biochemistry 1994;33:14807–14814. 42. Lyle TA. Small-molecule inhibitors of thrombin. Perspect Drug Discov Design 1993;1:453–460. 43. Tapparelli G, Metternich R, Ehrhardt C, et al. Synthetic low-molecular weight thrombin inhibitors: Molecular design and pharmacological profile. Trends Pharmacol Sci 1993;14:366–376. 44. Das J, Kimball SD, Thrombin active site inhibitors. Bioorg Med Chem 1995;3:999–1007. 45. Kikumoto R, Tamao Y, Tezuka T, et al. Selective inhibition of thrombin by (2R,4R)-4-methyl-1-[N2-[(3-methyl-1,2,3,4-tetrahydro-8-quinolinyl)sulfonyl]-l-arginyl)]-2-piperidinecarboxylic acid. Biochemistry 1984:23:85–90. 46. Hilpert K, Ackermann J, Banner DW, et al. Design and synthesis of potent and highly selective thrombin inhibitors. J Med Chem 1994;37:3889–3901. 47. Teger Nilsson TC, Bylund R, Gutsafsson D, et al. In vitro effects of inogatran, a selective low molecular weight thrombin inhibitor. Thromb Res 1997;85:133–145. 48. Gustafsson D, Antonsson T, Bylund R, et al. Effect of melagatran, a new low-molecular-weight thrombin inhibitor, on thrombin and fibrinolytic enzymes. Thromb Haemost 1998;79:110–118. 49. Wang KY, Krawczyk SH, Bischofberger N, et al. The tertiary structure of a DNA aptamer which binds to and inhibits thrombin determines activity. Biochemistry 1993;26;32:11285–11292. 50. Shuman RT, Rothenberger RB, Campbell CS, et al. Highly selective tripeptide thrombin inhibitors. J Med Chem 1993;36:314–319. 51. Kettner C, Mersinger L, Knabb R, The selective inhibition of thrombin by peptides of boroarginine. J Biol Chem 1990;265:18289–18297. 52. Marbet GA, Verstraete M, Kienast J, et al. Clinical pharmacology of intravenously administered recombinant desulfatohirudin (CGP 39393) in healthy volunteers. J Cardiovasc Pharmacol 1993;22:364–372. 53. Fox I, Dawson A, Loynds P, et al. Anticoagulant activity of Hirulog, a direct thrombin inhibitor, in humans. Thromb Haemost 1993;69:157–163. 54. Antman EM, for the TIMI 9A Investigators. Hirudin in acute myocardial infarction. Safety report from the Thrombolysis and Thrombin Inhibition in Myocardial Infarction (TIMI) 9A Trial. Circulation 1994;90;1624–1630. 55. The Global Use of Strategies to Open Occluded Coronary Arteries (GUSTO) IIa Investigators. Randomized trial of intravenous heparin versus recombinant hirudin for acute coronary syndromes. Circulation 1994;90:1631–1637. 56. Neuhause KL, von Essen R, Tebbe U, et al. Safety observations from the pilot phase of the randomized r-Hirudin for Improvement of Thrombolysis (HIT-III) study. A study of the Arbeitsgemeinschaft Leitender Kardiologischer Krankenausärzte (ALKK). Circulation 1994;90:1638–1642. 57. Bittl JA, Strony J, Brinker JA, et al., for the Hirulog Angioplasty Study Investigators. Treatment with bivalirudin (Hirulog) as compared with heparin during coronary angioplasty for unstable or post-infarction angina. N Engl J Med 1995;333:764–769. 58. Bittl JA. Comparative safety profiles of hirulog and heparin in patients undergoing coronary angioplasty. The Hirulog Angioplasty Study Investigators. Am Heart J 1995;130:658–665. 59. White HD, Aylward PE, Frey MJ, et al., for the Hirulog Early Reperfusion/Occlusion (HERO) Trial Investigators. Randomized, double-blind comparison of hirulog versus heparin in patients receiving streptokinase and aspirin for acute myocardial infarction (HERO). Circulation 1997;96:2155–2161. 60. Esmon CT. Molecular events that control the protein C anticoagulant pathway. Thromb Haemost 1993;70:29–35. 61. Topol EJ. Toward a new frontier in myocardial reperfusion therapy. Emerging platelet preeminence. Circulation 1998;97:211–218. 62. The Platelet Receptor Inhibition in Ischemic Syndrome Management in Patients Limited by Unstable Signs and Symptoms (PRISM-PLUS) Study Investigators. Inhibition of the platelet glycoprotein IIb/IIIa receptor with tirofiban in unstable angina and non-Q-wave myocardial infarction. N Engl J Med 1998;338:1488–1497. 63. The Global Use of Strategies to Open Occluded Coronary Arteries (GUSTO) IIb Investigators. A comparison of recombinant hirudin with heparin for the treatment of acute coronary syndromes. N Engl J Med 1996;335:775–782. 64. Antman EM. Hirudin in acute myocardial infarction. Thrombolysis and Thrombin Inhibition in Myocardial Infarction (TIMI) 9B trial. Circulation 1996;4:911–921. 65. Serruys PW, Herrman JP, Simon P, et al., for the HELVETICA Investigators. A comparison of hirudin with heparin in the prevention of restenosis after coronary angioplasty. N Engl J Med 1995;333:757–763. 66. Eriksson BI, Ekman S, Kalebo P, et al. Prevention of deep-vein thrombosis after total hip replacement: Direct thrombin inhibition with recombinant hirudin, CGP 39393. Lancet 1996;347:635–639. 67. Eikelboom JW, Anand SS, Mehta SR, et al. Prognostic significance of thrombocytopenia during hirudin and heparin therapy in acute coronary syndrome without ST elevation: Organization to Assess Strategies for Ischemic Syndromes (OASIS-2)study. Circulation 2001;6;103:643–645. 68. OASIS Pilot Study Investigators. Organization to Assess Strategies for Ischemic++ Syndromes. Flather MD, Weitz JI, Yusuf S, et al A.Reactivation of coagulation after stopping infusions of recombinant hirudin and unfractionated heparin in unstable angina and myocardial infarction without ST elevation: Results of a randomized trial. Eur Heart J 2000;21:1473–1481. 69. White H. Hirulog and Early Reperfusion or Occlusion (HERO)-2 Trial Investigators. Thrombin-specific anticoagulation with bivalirudin versus heparin in patients receiving fibrinolytic therapy for acute myocardial infarction: The HERO-2 randomised trial. Lancet 2001;358:1855–1863. 70. Lidon RM, Theroux P, Juneau M, et al. Initial experience with a direct antithrombin, hirulog, in unstable angina. Anticoagulant, antithrombotic, and clinical effects. Circulation 1993;88(4 Pt 1):1495–1501. 71. Topol EJ, Bonan R, Jewitt D, et al. Use of a direct antithrombin, hirulog, in place of heparin during coronary angioplasty. Circulation 1993;87:1622–1629. 72. Topol EJ. Evolution of improved antithrombotic and antiplatelet agents: Genesis of the Comparison of Abciximab Complications with Hirulog [and back-Up Abciximab] Events Trial (CACHET). Am J Cardiol 1998;82(8B):63P–68P. 73. Lincoff AM, Bittl JA, Harrington RA, et al. Bivalirudin and provisional glycoprotein IIb/IIIa blockade compared with heparin and planned glycoprotein IIb/IIIa blockade during percutaneous coronary intervention: REPLACE-2 randomized trial. J Am Med Assoc 2003;289:853–863. 74. Kondo LM, Wittkowsky AK, Wiggins BS. Argatroban for prevention and treatment of thromboembolism in heparin-induced thrombocytopenia. Ann Pharmacother 2001;35:440–451. 75. Alderman EL. Results from late-breaking clinical trials sessions at ACC 1998. American College of Cardiology. J Am Coll Cardiol 1998;32:1–7. 76. Lewis BE, Wallis DE, Berkowitz SD, et al. Argatroban anticoagulant therapy in patients with heparin-induced thrombocytopenia. Circulation 2001;103:1838–1843. 77. Jang IK, Brown DF, Giugliano RP, et al. A multicenter, randomized study of argatroban versus heparin as adjunct to tissue plasminogen activator (TPA) in acute myocardial infarction: Myocardial Infarction with Novastan and TPA (MINT) study. J Am Coll Cardiol 1999;33:1879–1885. 78. Wahlander K, Lapidus L, Olsson CG, et al. Pharmacokinetics, pharmacodynamics and clinical effects of the oral direct thrombin inhibitor ximelagatran in acute treatment of patients with pulmonary embolism and deep vein thrombosis. Thromb Res 2002;107:93–99. 79. Elg M, Gustafsson D, Carlsson S. Antithrombotic effects and bleeding time of thrombin inhibitors and warfarin in the rat. Thromb Res 1999;94:187–197. 80. Eriksson H, Wåhlander K, Lundström T, et al., for the THRIVE III Investigators. Extended secondary prevention with the oral direct thrombin inhibitor ximelagatran for 18 months after 6 months of anticoagulation in patients with venous thromboembolism: a randomized, placebo-controlled trial. Blood 2002;100:81a. 81. Petersen P. A two-year follow-up of ximelagatran as an oral anticoagulant for the prevention of stroke and systemic embolism in patients with nonvalvular atrial fibrillation (abstract P06.131).Neurology 2002;58(Suppl 3):A477. 82. Francis CW, Berkowitz SD, Comp PC, et al. Randomized, double-blind comparison of ximelagatran, an oral direct thrombin inhibitor, and warfarin to prevent venous thromboembolism (VTE) after total knee replacement (TKR) (abstract 300). Blood 2002;100:82a. 83. Heit JA, Colwell CW, Francis CW, et al. Comparison of the oral direct thrombin inhibitor ximelagatran with enoxaparin as prophylaxis against venous thromboembolism after total knee replacement: A phase 2 dose-finding study. Arch Intern Med 2001;161:2215–2221. 84. Eriksson H, Wåhlander K, Gustafsson D, et al. A randomized, controlled, dose-guiding study of the oral direct thrombin inhibitor ximelagatran compared with standard therapy for the treatment of acute deep vein thrombosis: THRIVE I. J Thromb Haemost 2003;1:41–47. 85. Eriksson BI, Bergqvist D, Kalebo P, et al. Ximelagatran and melagatran compared with dalteparin for prevention of venous thromboembolism after total hip or knee replacement: The METHRO II randomised trial. Lancet 2002;360:1441–1447. 86. Eriksson BI, Agnelli G, Cohen AT, et al. The oral direct thrombin inhibitor ximelagatran, and its subcutaneous (sc) form melagatran, compared with enoxaparin for prophylaxis of venous thromboembolism in total hip and total knee replacement (THR and TKR): The EXPRESS Study (abstract 299). Blood 2002;100:82a. 87. Eriksson BI, Arfwidsson AC, Frison L, et al. A dose-ranging study of the oral direct thrombin inhibitor, ximelagatran, and its subcutaneous form, melagatran, compared with dalteparin in the prophylaxis of thromboembolism after hip or knee replacement: METHRO I. MElagatran for THRombin inhibition in Orthopaedic surgery. Thromb Haemost 2002; 87:231–237. 88. Eriksson BI, Agnelli G, Cohen AT, et al. Direct thrombin inhibitor melagatran followed by oral ximelagatran in comparison with enoxaparin for prevention of venous thromboembolism after total hip or knee replacement. Thromb Haemost 2003;89:288–296. 89. Francis CW, Davidson BL, Berkowitz SD, et al. Ximelagatran versus warfarin for the prevention of venous thromboembolism after total knee arthroplasty. A randomized, double-blind trial. Ann Intern Med 2002;137:648–655. 90. Colwell CW, Berkowitz SD, Davidson BL, et al. Randomized, double-blind comparison of ximelagatran, an oral direct thrombin inhibitor, and enoxaparin to prevent venous thromboembolism (VTE) after total hip arthroplasty (THA) (abstract 2952). Blood 2001;98:706a. 91. Olson SB, Peters P, on behalf of the SPORTIF II Investigators. Fixed-dose oral direct thrombin inhibitor ximelagatran as an alternative for dose-adjusted warfarin in patients with non-valvular atrial fibrillation (abstract). Eur Heart J 2001;22:330. 92. Fixed-dose ximelagatran at least as effective as adjusted-dose warfarin for stroke prevention in AF patients: Results of SPORTIF III trial. Medscape CRM News.

Advertisement

Advertisement

Advertisement