Skip to main content

Advertisement

Log in

Ancrod revisited: viscoelastic analyses of the effects of Calloselasma rhodostoma venom on plasma coagulation and fibrinolysis

  • Published:
Journal of Thrombosis and Thrombolysis Aims and scope Submit manuscript

Abstract

Fibrinogen depletion via catalysis by snake venom enzymes as a therapeutic strategy to prevent or treat thrombotic disorders was utilized for over four decades, with ancrod being the quintessential agent. However, ancrod eventually was found to not be of clinical utility in large scale stroke trial, resulting in the eventual discontinuation of the administration of the drug for any indication. It was hypothesized that ancrod, possessing thrombin-like activity, may have unappreciated robust coagulation kinetics. Using thrombelastographic methods, a comparison of equivalent tissue factor initiated thrombin generation and Calloselasma rhodostoma venom (rich in ancrod activity) on plasmatic coagulation kinetics was performed. The venom resulted in thrombi that formed nearly twice as fast compared to thrombin formed clots, and there was no difference in fibrinolytic kinetics initiated by tissue-type plasminogen activator. In plasma containing iron and carbon monoxide modified fibrinogen, which may be found in patients at risk of stroke, the coagulation kinetic differences observed with venom was still more vigorous than that seen with thrombin. These phenomena may provide insight into the clinical failure of ancrod, and may serve as an impetus to revisit the concept of fibrinogen depletion via fibrinogenolytic enzymes, not those with thrombin-like activity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Bell WR Jr (1997) Defibrinogenating enzymes. Drugs 54(Suppl 3):18–30

    Article  CAS  PubMed  Google Scholar 

  2. Soutar RL, Ginsberg JS (1993) Anticoagulant therapy with ancrod. Crit Rev Oncol Hematol 15:23–33

    Article  CAS  PubMed  Google Scholar 

  3. O-Yurvati AH, Laub GW, Southgate TJ, McGrath TJ (1994) Heparinless cardiopulmonary bypass with ancrod. Ann Thorac Surg 57:1656–1658

    Article  CAS  PubMed  Google Scholar 

  4. Brennan MB, MacKean GL (1994) Heparin-induced thrombosis treated with ancrod. Can J Surg 37:161–164

    CAS  PubMed  Google Scholar 

  5. Lewis BE, Leya FS, Wallis D, Grassman E (1994) Failure of ancrod in the treatment of heparin-induced arterial thrombosis. Can J Cardiol 10:559–561

    CAS  PubMed  Google Scholar 

  6. Sherman DG, Atkinson RP, Chippendale T, Levin KA, Ng K, Futrell N, Hsu CY, Levy DE (2000) Intravenous ancrod for treatment of acute ischemic stroke: the STAT study: a randomized controlled trial. Stroke Treatment with Ancrod Trial. JAMA 283:2395–2403

    Article  CAS  PubMed  Google Scholar 

  7. Hennerici MG, Kay R, Bogousslavsky J, Lenzi GL, Verstraete M, Orgogozo JM, ESTAT investigators (2006) Intravenous ancrod for acute ischaemic stroke in the European Stroke Treatment with Ancrod Trial: a randomised controlled trial. Lancet 368:1871–1878

    Article  CAS  PubMed  Google Scholar 

  8. Lubenow N, Warkentin TE, Greinacher A, Wessel A, Sloane DA, Krahn EL, Magnani HN (2006) Results of a systematic evaluation of treatment outcomes for heparin-induced thrombocytopenia in patients receiving danaparoid, ancrod, and/or coumarin explain the rapid shift in clinical practice during the 1990s. Thromb Res 117:507–515

    Article  CAS  PubMed  Google Scholar 

  9. Murphy GS, Marymont JH (2007) Alternative anticoagulation management strategies for the patient with heparin-induced thrombocytopenia undergoing cardiac surgery. J Cardiothorac Vasc Anesth 21:113–126

    Article  CAS  PubMed  Google Scholar 

  10. Levy DE, del Zoppo GJ, Demaerschalk BM, Demchuk AM, Diener HC, Howard G, Kaste M, Pancioli AM, Ringelstein EB, Spatareanu C, Wasiewski WW (2009) Ancrod in acute ischemic stroke: results of 500 subjects beginning treatment within 6 hours of stroke onset in the ancrod stroke program. Stroke 40:3796–3803

    Article  PubMed  Google Scholar 

  11. Hao Z, Liu M, Counsell C, Wardlaw JM, Lin S, Zhao X (2012) Fibrinogen depleting agents for acute ischaemic stroke. Cochrane Database Syst Rev 14(3):CD000091

    Google Scholar 

  12. Bell WR, Shapiro SS, Martinez J, Nossel HL (1978) The effects of ancrod, the coagulating enzyme from the venom of Malayan pit viper (A. rhodostoma) on prothrombin and fibrinogen metabolism and fibrinopeptide A release in man. J Lab Clin Med 91:592–604

    CAS  PubMed  Google Scholar 

  13. Dambisya YM, Lee TL, Gopalakrishnakone P (1994) Action of Calloselasma rhodostoma (Malayan pit viper) venom on human blood coagulation and fibrinolysis using computerized thromboelastography (CTEG). Toxicon 32:1619–1626

    Article  CAS  PubMed  Google Scholar 

  14. Dempfle CE, Argiriou S, Kucher K, Müller-Peltzer H, Rübsamen K, Heene DL (2000) Analysis of fibrin formation and proteolysis during intravenous administration of ancrod. Blood 96:2793–2802

    CAS  PubMed  Google Scholar 

  15. Dempfle CE, Alesci S, Kucher K, Müller-Peltzer H, Rübsamen K, Borggrefe M (2001) Plasminogen activation without changes in tPA and PAI-1 in response to subcutaneous administration of ancrod. Thromb Res 104:433–438

    Article  CAS  PubMed  Google Scholar 

  16. Dempfle CE, Argiriou S, Alesci S, Kucher K, Müller-Peltzer H, Rübsamen K, Heene DL (2001) Fibrin formation and proteolysis during ancrod treatment. Evidence for des-A-profibrin formation and thrombin independent factor XIII activity. Ann N Y Acad Sci 936:210–214

    Article  CAS  PubMed  Google Scholar 

  17. Liu S, Marder VJ, Levy DE, Wang SJ, Yang F, Paganini-Hill A, Fisher MJ (2011) Ancrod and fibrin formation: perspectives on mechanisms of action. Stroke 42:3277–3280

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fu J, Huang J, Lei M, Luo Z, Zhong X, Huang Y, Zhang H, Liu R, Tong J, He F (2015) Prevalence and impact on stroke in patients receiving maintenance hemodialysis versus peritoneal dialysis: a prospective observational study. PLoS ONE 10:e0140887

    Article  PubMed  PubMed Central  Google Scholar 

  19. Li X, Song G, Jin Y, Liu H, Li C, Han C, Ren S (2014) Higher level of heme oxygenase-1 in patients with stroke than TIA. J Thorac Dis 6:772–777

    PubMed  PubMed Central  Google Scholar 

  20. Matika RW, Nielsen VG, Steinbrenner EB, Sussman AN, Madhrira M (2014) Hemodialysis patients have plasmatic hypercoagulability and decreased fibrinolytic vulnerability: role of carbon monoxide. ASAIO J 60:716–721

    Article  CAS  PubMed  Google Scholar 

  21. Nielsen VG, Kulin W, LaWall JS, MacFarland FN, Chen A, Hadley HA, DaDeppo AJ, Steinbrenner EB, Matika RW (2015) Chronic migraineurs form circulating carboxyhemefibrinogen and iron-bound fibrinogen. CNS Neurol Disord 14:1079–1085

    Article  CAS  Google Scholar 

  22. Shah N, Welsby IJ, Fielder MA, Jacobsen WK, Nielsen VG (2015) Sickle cell disease is associated with iron mediated hypercoagulability. J Thromb Thrombolysis 40:182–185

    Article  CAS  PubMed  Google Scholar 

  23. Nielsen VG, Redford DT, Boyle PK (2016) Effect of iron and carbon monoxide on fibrinogenase-like degradation of plasmatic coagulation by venoms of six Agkistrodon species. Basic Clin Pharmacol Toxicol. doi:10.1111/bcpt.12504

    Google Scholar 

  24. Nielsen VG, Cohen BM, Cohen E (2006) Elastic modulus-based thrombelastographic quantification of plasma clot fibrinolysis with progressive plasminogen activation. Blood Coagul Fibrinolysis 17:75–81

    Article  CAS  PubMed  Google Scholar 

  25. Ellis TC, Nielsen VG, Marques MB, Kirklin JK (2007) Thrombelastographic measures of clot propagation: a comparison of alpha to maximum rate of thrombus generation. Blood Coagul Fibrinolysis 18:45–48

    Article  PubMed  Google Scholar 

  26. Nielsen VG, Boyer LV (2016) Iron and carbon monoxide attenuate degradation of plasmatic coagulation by Crotalus atrox venom. Blood Coagul Fibrinolysis (in press)

  27. Nielsen VG, Redford DT, Boyle PK (2016) Effect of iron and carbon monoxide on fibrinogenase-like degradation of plasmatic coagulation by venoms of four Crotalus species. Blood Coagul Fibrinolysis (in press)

Download references

Acknowledgments

This investigation was supported by the Department of Anesthesiology.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vance G. Nielsen.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nielsen, V.G. Ancrod revisited: viscoelastic analyses of the effects of Calloselasma rhodostoma venom on plasma coagulation and fibrinolysis. J Thromb Thrombolysis 42, 288–293 (2016). https://doi.org/10.1007/s11239-016-1343-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11239-016-1343-6

Keywords

Navigation