Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

A new oral anticoagulant: the 50-year challenge

Key Points

  • During the twentieth century, it was gradually recognized that thrombosis is a major cause of morbidity and mortality in the Western world. There is therefore a great need for effective prophylaxis and treatment of thromobsis, and the use of oral anticoagulants is extensive.

  • However, traditional oral anticoagulants have several drawbacks. Warfarin, the most commonly used oral anticoagulant, has a narrow therapeutic window, and suffers from frequent drug–drug and food–drug interactions, and so requires careful monitoring of its anticoagulant effects.

  • So, there is a strong medical need for a well-tolerated, novel oral anticoagulant with a wide therapeutic window, plus the combination of favourable absorption, distribution and metabolism and elimination properties to enable predictable protection without coagulation monitoring.

  • In response to this need, our team embarked on a project in the mid-1980s with the aim of devising an improved oral anticoagulant. Thrombin, a protease that has a key role in blood coagulation, was chosen as the target.

  • This article concentrates on the preclinical development of the first oral direct thrombin inhibitor ximelagatran (Exanta; AstraZeneca).

Abstract

It is rare for any drug introduced more than 50 years ago to remain unsurpassed today; yet the oral prevention and treatment of thrombosis are still achieved by the use of the vitamin K antagonists (coumarins), such as warfarin, which were introduced in the 1940s and 1950s. For these anticoagulants, careful monitoring of the effect is needed to avoid bleeding or loss of efficacy. On the basis of the need for improved oral anticoagulants, a goal was set in 1985 to develop a new oral anticoagulant that could replace the vitamin K antagonists. After providing some medical and historical context, this article discusses the challenges facing the multidisciplinary team of scientists who were involved in the project leading to the discovery of the anticoagulant ximelagatran (Exanta; AstraZeneca), the first oral direct thrombin inhibitor.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Blood coagulation.
Figure 2: The target and the chemical starting point.
Figure 3: A schematic representation of the development of chemistry of ximelagatran.
Figure 4: Binding of melagatran and fibrinopeptide A to thrombin.
Figure 5: Antithrombotic effects and bleeding in the rat.
Figure 6: A comparison of properties of melagatran and ximelagatran.
Figure 7: Pharmacokinetic profile of ximelagatran.

Similar content being viewed by others

References

  1. White, R. H. The epidemiology of venous thromboembolism. Circulation 107 (Suppl. 1), I4–I8 (2003).

    PubMed  Google Scholar 

  2. Feigin, V. L., Lawes, C. M., Bennett, D. A. & Anderson, C. S. Stroke epidemiology: a review of population-based studies of incidence, prevalence, and case-fatality in the late 20th century. Lancet Neurol. 2, 43–53 (2003).

    Article  Google Scholar 

  3. Fang, J. & Alderman, M. H. Dissociation of hospitalization and mortality trends for myocardial infarction in the United States from 1988 to 1997. Am. J. Med. 113, 208–214 (2002).

    Article  Google Scholar 

  4. Campbell, H. A. & Link, K. P. Studies on the hemorrhagic sweet clover disease. IV. The isolation and crystallization of the hemorrhagic agent. J. Biol. Chem. 21, 138 (1941).

    Google Scholar 

  5. Allen, E. V., Baker, N. W. & Waugh, J. M. A preparation from spoiled sweet clover [3,3'-methylene-bis-(4-hydroxycoumarin)] which prolongs coagulation and prothrombin time of the blood: A clinical study. JAMA 120, 1009–1015 (1942).

    Article  CAS  Google Scholar 

  6. Lehmann, J. On experimental hypo-prothrombinemia produced by methylene-bis-(hydroxycoumarin) and its use in the treatment of thrombosis. Lancet I, 318 (1942).

    Article  CAS  Google Scholar 

  7. Link, K. P. The discovery of dicumarol and its sequels. Circulation 19, 97–101 (1958).

    Article  Google Scholar 

  8. Jorpes, E. On heparin, its chemical nature and properties. Acta Med. Scand. 88, 427–433 (1936).

    Article  CAS  Google Scholar 

  9. Murray, D. W. G., Jaques, L. B., Perret, T. S. & Best, G. G. Heparin and the thrombosis of veins following injury. Surgery 2, 163–187 (1937).

    CAS  Google Scholar 

  10. Kakkar, V. V. et al. Low molecular weight heparin and prevention of postoperative deep vein thrombosis. BMJ 284, 375 (1982).

    Article  CAS  Google Scholar 

  11. Turpie, A. G. G., Gallus, A. S. & Hoek, J. A. A synthetic pentasaccharide for the prevention of deep-vein thrombosis after total hip replacement. N. Engl. J. Med. 344, 619–625 (2001).

    Article  CAS  Google Scholar 

  12. Hirsh, J. et al. Oral anticoagulants: mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 119 (Suppl. 1), S8–S21 (2001).

    Article  Google Scholar 

  13. Hirsh, J. et al. Heparin and low-molecular-weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 119 (Suppl. 1), S64–S94 (2001).

    Article  Google Scholar 

  14. Mann, K. G. Thrombin formation. Chest 124 (Suppl. 3), S4–S10 (2003).

    Article  Google Scholar 

  15. Di Cera, E. Thrombin interactions. Chest 124 (Suppl. 3), S11–S7 (2003).

    Article  Google Scholar 

  16. Bettelheim, F. R. Clotting of fibrinogen. II. Fractionation of peptide material liberated. Biochem. Biophys. Acta 19, 121–130 (1956).

    Article  CAS  Google Scholar 

  17. Blombäck, B., Blombäck, M., Gröndahl, N. J. & Holmberg, E. Structure of fibrinopeptides, its relation to enzyme specificity and phylogeny and classification of species. Ark. Kemi 25, 411–416 (1966).

    Google Scholar 

  18. Blombäck, B., Blombäck, M., Olsson, P., Svendsen L. & Åberg, G. Synthetic peptides with anticoagulant and vasodilating activity. J. Clin. Lab. Invest. 24 (Suppl. 107), 59–64 (1969).

    Google Scholar 

  19. Claeson, G., Aurell, L., Karlson, G. & Friberger, P. Substrate structure and activity relationships. Topics Hematol: 16th Internatl Cong. Hematol. (Excerpta Medica) 415, 381–384 (1976).

    Google Scholar 

  20. Kettner, C. & Shaw, E. D-Phe–Pro–ArgCH2Cl — a selective affinity label for thrombin. Thromb. Res. 14, 969 (1979).

    Article  CAS  Google Scholar 

  21. Bajusz, S., Barabas, E., Tolnay, P., Szell, E. & Bagdy, D. Inhibition of thrombin and trypsin by tripeptide aldehydes. Int. J. Pept. Protein Res. 12, 217–221 (1978).

    Article  CAS  Google Scholar 

  22. Okamoto, S. et al. A synthetic thrombin inhibitor taking extremely active stereostructure. Thromb. Haemost. 42, A205 (1979).

    Google Scholar 

  23. Okamoto, S. et al. Potent inhibition of thrombin by the newly synthesized arginine derivative No. 805. The importance of stereo-structure of its hydrophobic carboxamide portion. Biochem. Biophys. Res. Commun. 101, 440–446 (1981).

    Article  CAS  Google Scholar 

  24. Walenga, J. M. An overview of the direct thrombin inhibitor argatroban. Pathophysiol. Haemost. Thromb. 32 (Suppl. 3), 9–14 (2002).

    Article  CAS  Google Scholar 

  25. Steinmetzer, T., Hauptmann, J. & Stürzebecher, J. Advances in the development of thrombin inhibitors. Expert Opin. Investig. Drugs 10, 845–864 (2001).

    Article  CAS  Google Scholar 

  26. Markwardt, F. The development of hirudin as an antithrombotic drug. Thromb. Res. 74, 1–23 (1994).

    Article  CAS  Google Scholar 

  27. White, H. D. & Chew, D. P. Bivalirudin: an anticoagulant for acute coronary syndromes and coronary interventions. Expert Opin. Pharmacother. 3, 777–788 (2002).

    Article  CAS  Google Scholar 

  28. Jones, D. M. et al. Design and synthesis of thrombin inhibitors. Lett. Peptide Sci. 2, 147–154 (1995).

    Article  CAS  Google Scholar 

  29. Lipinski, C. A., Lombardo, F., Dominy, B. W. & Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 23, 3–25 (1997).

    Article  CAS  Google Scholar 

  30. Bajusz, S., Széll nee Hasenöhrl, E., Barabás E., Bagdy D. & Mohai nee Nagy, Z. Novel anticoagulant agmatine derivatives and process for the preparation thereof. US Patent 4,346,078 (1982).

  31. Teger-Nilsson, A. C., Bylund, R., Gustafsson, D., Gyzander, E. & Eriksson U. In vitro effects of inogatran, a selective low molecular weight thrombin inhibitor. Thromb. Res. 85, 133–145 (1997).

    Article  CAS  Google Scholar 

  32. Eriksson, U. G., Renberg, L., Bredberg, U., Teger-Nilsson A. C. & Regårdh, C. G. Animal pharmacokinetics of inogatran, a low-molecular-weight thrombin inhibitor with potential use as an antithrombotic drug. Biopharm. Drug Dispos. 19, 55–64 (1998).

    Article  CAS  Google Scholar 

  33. Bode, W. et al. The refined 1.9 Å crystal structure of human α-thrombin: interaction with D-Phe–Pro–Arg chloromethylketone and significance of the Tyr–Pro–Pro–Trp insertion segment. EMBO J. 8, 3467–3475 (1989).

    Article  CAS  Google Scholar 

  34. Gustafsson, D. et al. Effects of melagatran, a new low-molecular-weight thrombin inhibitor, on thrombin and fibrinolytic enzymes. Thromb. Haemost. 79, 110–118 (1998).

    Article  CAS  Google Scholar 

  35. Elg, M., Gustafsson, D. & Deinum, J. The importance of enzyme inhibition kinetics for the effect of thrombin inhibitors in a rat model of arterial thrombosis. Thromb. Haemost. 78, 1286–1292 (1997).

    Article  CAS  Google Scholar 

  36. Elg, M., Gustafsson, D. & Carlsson, S. Antithrombotic effects and bleeding time of thrombin inhibitors and warfarin in the rat. Thromb. Res. 94, 187–197 (1999). A comparison of melagatran and warfarin on the therapeutic interval between antithrombotic effect and bleeding in the anaesthetized rat.

    Article  CAS  Google Scholar 

  37. Eriksson, U. G. et al. Absorption, distribution, metabolism, and excretion of ximelagatran, an oral direct thrombin inhibitor, in rats, dogs, and humans. Drug Metab. Dispos. 31, 294–305 (2003). A comprehensive description of the pharmacokinetics of ximelagatran and melagatran in animals and man.

    Article  CAS  Google Scholar 

  38. Gustafsson, D. et al. The direct thrombin inhibitor melagatran and its oral prodrug H 376/95: Intestinal absorption properties, biochemical and pharmacodynamic effects. Thromb. Res. 101, 171–181 (2001). The first full-paper description of ximelagatran.

    Article  CAS  Google Scholar 

  39. The TRIIM study group. A low molecular weight, selective thrombin inhibitor, inogatran, vs heparin for unstable coronary artery disease. Eur. Heart J. 18, 1416–1425 (1997).

  40. Artursson, P., Ungell, A. l. & Löfroth, J. E. Selective paracellular permeability in two models of intestinal absorption: cultured monolyers of human intestinal epithelial cells and rat intestinal segments. Pharm. Res. 10, 1123–1129 (1993).

    Article  CAS  Google Scholar 

  41. Carlsson, S., Elg, M. & Mattsson, C. Effects of ximelagatran, the oral form of melagatran, in the treatment of caval vein thrombosis in conscious rats. Thromb. Res. 107, 163–168 (2002).

    Article  CAS  Google Scholar 

  42. Eriksson, U. G. et al. Pharmacokinetics and pharmacodynamics of ximelagatran, a novel oral direct thrombin inhibitor, in young, healthy male subjects. Eur. J. Clin. Pharmacol. 59, 35–43 (2003).

    Article  CAS  Google Scholar 

  43. Bredberg, E. et al. Ximelagatran, an oral direct thrombin inhibitor, has a low potential for cytochrome P450-mediated drug-drug interactions. Clin. Pharmacokinet. 42, 765–777 (2003).

    Article  CAS  Google Scholar 

  44. Johansson, L. C. et al. Influence of age on the pharmacokinetics and pharmacodynamics of ximelagatran, an oral direct thrombin inhibitor. Clin. Pharmacokinet. 42, 381–392 (2003).

    Article  CAS  Google Scholar 

  45. Eriksson, B. I. 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. The METHRO III study. Thromb. Haemost. 89, 288–296 (2003).

    Article  CAS  Google Scholar 

  46. Eriksson, B. I. et al. The direct thrombin inhibitor melagatran followed by oral ximelagatran compared with enoxaparin for the prevention of venous thromboembolism after total hip or knee replacement: the EXPRESS study. J. Thromb. Haemost. 1, 2490–2496 (2003).

    Article  CAS  Google Scholar 

  47. Francis, C. W. et al. Comparison of ximelagatran with warfarin for the prevention of venous thromboembolism after total knee replacement. N. Engl. J. Med. 349, 1703–1712 (2003). A Phase III comparison of ximelagatran with warfarin of VTE prophylaxis in patients following knee-replacement surgery.

    Article  CAS  Google Scholar 

  48. Colwell, C. W. 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): EXULT B. Blood 102, A39 (2003).

    Google Scholar 

  49. Francis, C. W. et al. Efficacy and safety of the oral direct thrombin inhibitor ximelagatran compared with current standard therapy for acute, symptomatic deep vein thrombosis, with or without pulmonary embolism: The THRIVE Treatment Study. Blood 102, A7 (2003).

    Article  Google Scholar 

  50. Schulman, S., Wåhlander, K., Lundström, T., Clason, S. B. & Eriksson, H. Secondary prevention of venous thromboembolism with the oral direct thrombin inhibitor ximelagatran. N. Engl. J. Med. 349, 1713–1721 (2003). A Phase III comparison of ximelagatran with placebo of long-term secondary prevention of VTE in patients with a previous VTE.

    Article  CAS  Google Scholar 

  51. Executive Steering Committee on behalf of the SPORTIF III Investigators. Stroke prevention with the oral direct thrombin inhibitor ximelagatran compared with warfarin in patients with non-valvular atrial fibrillation (SPORTIF III): randomised controlled trial. Lancet 362, 1691–1698 (2003). A Phase III comparison of ximelagatran with warfarin of stroke prevention in patients with non-valvular atrial fibrillation.

  52. The SPORTIF Executive Steering Committee on behalf of the SPORTIF V Investigators. Stroke prevention using the oral direct thrombin inhibitor ximelagatran in patients with nonvalvular atrial fibrillation (SPORTIF V). Circulation 108, 2723 (2003).

  53. Wallentin, L. et al. Oral ximelagatran for secondary prophylaxis after myocardial infarction: the ESTEEM randomised controlled trial. Lancet 362, 789–797 (2003).

    Article  CAS  Google Scholar 

  54. Diener, H. C. Stroke prevention with the oral direct thrombin inhibitor ximelagatran in patients with nonvalvular atrial fibrillation: pooled analysis of the SPORTIF III and V trials. Cerebrovasc. Dis. 17 (Suppl. 5), 16 (2004).

    Google Scholar 

  55. Gustafsson, D. Oral direct thrombin inhibitors in clinical development. J. Intern. Med. 254, 322–334 (2003).

    Article  CAS  Google Scholar 

  56. Lassen, M. R. et al. A phase II randomized, double-blind, five-arm, parallel-group, dose–response study of a new oral directly-acting factor Xa inhibitor, razaxaban, for the prevention of deep vein thrombosis in knee replacement surgery. Blood 102, A41 (2003).

    Google Scholar 

  57. Ni, F., Meinwald, Y. C., Vasquez, M. & Scheraga, H. A. High-resolution NMR studies of fibrinogen-like peptides in solution: Structure of a thrombin-bound peptide corresponding to residues 7–16 of the Aα chain of human fibrinogen. Biochemistry 28, 3094–3105 (1989).

    Article  CAS  Google Scholar 

  58. Stubbs, M. T. et al. The interaction of thrombin with fibrinogen. A structural basis for its specificity. Eur. J. Biochem. 206, 187–195 (1992).

    Article  CAS  Google Scholar 

  59. Gustafsson, D. & Elg, M. The pharmacodynamics and pharmacokinetics of the oral direct thrombin inhibitor ximelagatran and its active metabolite melagatran: a mini-review. Thromb. Res. 109, S9–S15 (2003).

    Article  CAS  Google Scholar 

  60. Johansson, L. C., Andersson, M., Fager, G., Gustafsson, D. & Eriksson, U. G. No influence of ethnic origin on the pharmacokinetics and pharmacodynamics of melagatran, following oral administration of ximelagatran, a novel oral direct thrombin inhibitor, to healthy male volunteers. Clin. Pharmacokinet. 42, 475–484 (2003).

    Article  CAS  Google Scholar 

  61. Sarich, T. C. et al. No influence of obesity on the pharmacokinetics and pharmacodynamics of melagatran, the active form of the oral direct thrombin inhibitor, ximelagatran. Clin. Pharmacokinet. 42, 485–492 (2003).

    Article  CAS  Google Scholar 

  62. Wåhlander, K., Eriksson-Lepkowska, M., Frison, L., Fager, G. & Eriksson, U. G. No influence of mild-to-moderate hepatic impairment on the pharmacokinetics and pharmacodynamics of ximelagatran, an oral direct thrombin inhibitor. Clin. Pharmacokinet. 42, 755–764 (2003).

    Article  Google Scholar 

  63. Eriksson, B. I. 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. Thromb. Haemost. 87, 231–237 (2002).

    Article  CAS  Google Scholar 

  64. Wolzt, M. et al. Consistent pharmacokinetics of the oral direct thrombin inhibitor ximelagatran in patients with nonvalvular atrial fibrillation and in healthy subjects. Eur. J. Clin. Pharmacol. 59, 537–543 (2003). Reports the pharmacokinetics of ximelagatran and melagatran in patients and elderly healthy volunteers.

    Article  CAS  Google Scholar 

  65. Wåhlander, K. 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. 107, 93–99 (2002).

    Article  Google Scholar 

  66. Eriksson, U. G. et al. Influence of severe renal impairment on the pharmacokinetics and pharmacodynamics of oral ximelagatran and subcutaneous melagatran. Clin. Pharmacokinet. 42, 743–753 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The discoveries of ximelagatran and melagatran and their development and documentation for clinical use are the result of hard work in dedicated, multidisciplinary teams within AstraZeneca. Also important have been the skillful advice and participation of many physicians in the large clinical study programme. Consequently, many share the honour of this long-term project that led to Exanta.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David Gustafsson.

Ethics declarations

Competing interests

D. G., R. B., T. A., I. N., J.-E. N., U. E. and U. B. are all employees of AstraZeneca. A.-C. T.-N. was an employee of AstraZeneca but retired some years ago.

Related links

Related links

DATABASES

Entrez Gene

Fibrinogen

FXa

thrombin

FURTHER INFORMATION

Encyclopedia of Life Sciences

Blood coagulation

venous thrombosis

Glossary

THROMBUS

An aggregation of platelets and fibrin with entrapped cells, mainly erythrocytes, in a blood vessel or a cardiac cavity. In a vessel, the thrombus frequently causes occlusion, thereby hindering blood flow.

MYOCARDIAL INFARCTION

Commonly known as a heart attack, this is death of part of the heart muscle due to sudden loss of blood supply. Typically, the loss of supply is caused by complete blockage of a coronary artery by a blood clot.

RULE OF FIVE

Identifies several key properties that should be considered for compounds with oral delivery in mind. These properties are molecular mass <500 Da, cLogP <5, number of hydrogen-bond donors <5 and number of hydrogen-bond acceptors <10.

ACTIVATED PARTIAL THROMBOPLASTIN TIME

Plasma coagulation assay, originally developed for monitoring the effects of unfractionated heparin, in which plasma coagulation is activated in two steps.

pKa

The pKa for any group is the pH at which it would be protonated in 50% of molecules. More molecules will become protonated with decreasing pH, and vice versa.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Gustafsson, D., Bylund, R., Antonsson, T. et al. A new oral anticoagulant: the 50-year challenge. Nat Rev Drug Discov 3, 649–659 (2004). https://doi.org/10.1038/nrd1466

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd1466

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing