Skip to main content

Bat Plasminogen Activator: Desmoteplase – From Bat to Bench to Bedside of Stroke Victims

  • Chapter
  • First Online:
Toxins and Hemostasis

Abstract

Desmoteplase (DSPA) was identified in the salivary venom of Desmodus rotundus (a blood-feeding vampire bat common in Latin America) triggered by observations as early as 1964. The initial interest in DSPA as a therapeutic modality was raised by the success of recombinant tissue plasminogen activator (rt-PA) in acute myocardial infarction (AMI) and the evolving paradigm of fibrin-specificity as the key to safe and effective thrombolysis. The early research on DSPA confirmed an extremely high fibrin specificity and a potential for a lower bleeding propensity, demonstrated in a variety of preclinical studies. Obviously, the unique task in the Vampire Bat – curbing clot formation without disintegrating the other salivary proteins – has led to a protease which serves no other known function than activating plasminogen in the presence of fibrin. This fibrin and substrate specificity distinguishes DSPA from rt-PA, which, apart from clot lysis, has a number of additional physiological roles.

The first clinical trial in AMI confirmed the absence of fibrinogen depletion even at doses of 750 μg/kg (90 μg/kg is effective in acute ischaemic stroke, AIS). DSPA was abandoned by Schering AG for strategic reasons and, in 2001, its further development was redirected by PAION to acute ischemic stroke (AIS), based on the assumption that a more fibrin-specific thrombolytic should pose a lower bleeding risk and allow a longer post-stroke treatment window. Also in 2001, the discovery of the ability of rt-PA to enhance NMDA-induced neurotoxicity (which models the glutamate neurotoxicity known in vivo) gave rise to the speculation that DSPA, by virtue of its high specialization, might be different. The subsequent multi-level research indeed confirmed that DSPA is devoid of any neurotoxic properties. The main reason seems to lie in a structural difference: The deleterious augmentation of NMDA neurotoxicity by rt-PA (and its mutants, shown in vitro) requires the kringle 2 (K2) domain, a moiety lacking in DSPA. This distinction may prove advantageous also in other areas, as, more generally, the kringle 2 seems to capacitate (r)t-PA for numerous mechanisms it is known to operate, sometimes to a harmful end. An important example is its K2-dependent ability to activate platelet-derived growth factor (PDGF), which is instrumental in (r)t-PA induced weakening of the blood brain barrier.

DSPA is now in a global phase III program in AIS, sponsored by PAION’s partner Lundbeck, treating patients up to 9 h after stroke onset, a wide extension of the current window of 3–4.5 h.

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

Access this chapter

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Chapter
USD 29.95
Price excludes VAT (USA)
eBook
USD 169.00
Price excludes VAT (USA)
Softcover Book
USD 219.99
Price excludes VAT (USA)
Hardcover Book
USD 219.99
Price excludes VAT (USA)

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  • Belwood, J.J., Morton, P.A., 1991. Vampires: the real story. The truth about the bats people love to hate is even more fascinating than the myths. Bats 9, 11–16.

    Google Scholar 

  • Benchenane, K., López-Atalaya, J.P., Fernandez-Monreal, M., Touzani, O., Vivien, D., 2004. Equivocal roles of tissue-type plasminogen activator in stroke-induced injury. Trends Neurosci. 27, 155–160.

    Article  PubMed  CAS  Google Scholar 

  • Benchenane, K., Castel, H., Boulouard, M., Bluthé, R., Fernandez-Monreal, M., Roussel, B.D., López-Atalaya, J.P., Butt-Gueulle, S., Agin, V., Maubert, E., Dantzer, R., Touzani, O., Dauphin, F., Vivien, D., Ali, C., 2007. Anti-NR1 N-terminal-domain vaccination unmasks the crucial action of tPA on NMDA-receptor-mediated toxicity and spatial memory. J. Cell. Sci. 120, 578–585.

    Article  PubMed  CAS  Google Scholar 

  • Bergum, P.W., Gardell, S.J., 1992. Vampire bat salivary plasminogen activator exhibits a strict and fastidious requirement for polymeric fibrin as its cofactor, unlike human tissue-type plasminogen activator. A kinetic analysis. J. Biol. Chem. 267, 17726–17731.

    CAS  Google Scholar 

  • Bringmann, P., Gruber, D., Liese, A., Toschi, L., Krätzchmar, J., Schleuning, W.D., Donner, P., 1995. Structural features mediating fibrin selectivity of vampire bat plasminogen activators. J. Biol. Chem. 270, 25596–25603.

    Article  PubMed  CAS  Google Scholar 

  • Busch, E., Krüger, K., Fritze, K., Allegrini, P.R., Hoehn-Berlage, M., Hossmann, K.A., 1997. Blood-brain barrier disturbances after rt-PA treatment of thromboembolic stroke in the rat. Acta Neurochir. Suppl. 70, 206–208.

    PubMed  CAS  Google Scholar 

  • Collen, D., 1996. Fibrin-selective thrombolytic therapy for acute myocardial infarction. Circulation 93, 857–865.

    Article  PubMed  CAS  Google Scholar 

  • Epple, G., Schleuning, W.D., Kettelgerdes, G., Kottgen, E., Gessner, R., Praus, M., 2004. Prion protein stimulates tissue-type plasminogen activator-mediated plasmin generation via a lysine-binding site on kringle 2. J. Thromb. Haemost. 2, 962–968.

    Article  PubMed  CAS  Google Scholar 

  • Fanne, R.A., Nassar, T., Yarovoi, S., Rayan, A., Lamensdorf, I., Karakoveski, M., Vadim, P., Fanne, R.A., Jamal, M., Cines, D.B., Al-Roof Higazi, A., 2010. Blood-brain barrier permeability and tPA-mediated neurotoxicity. Neuropharmacology 58, 972–980.

    Google Scholar 

  • Fernández-Monreal, M., López-Atalaya, J.P., Benchenane, K., Cacquevel, M., Dulin, F., Le Caer, J.P., Rossier, J., Jarrige, A.C., Mackenzie, E.T., Colloc’h, N., Ali, C., Vivien, D., 2004. Arginine 260 of the amino-terminal domain of NR1 subunit is critical for tissue-type plasminogen activator-mediated enhancement of N-methyl-D-aspartate receptor signaling. J. Biol. Chem. 279, 50850–508506.

    Article  PubMed  Google Scholar 

  • Fredriksson, L., Ehnman, M., Fieber, C., Eriksson, U., 2005. Structural requirements for activation of latent platelet-derived growth factor CC by tissue plasminogen activator. J. Biol. Chem. 280, 26856–26862.

    Article  PubMed  CAS  Google Scholar 

  • Furlan, A.J., Eyding, D., Albers, G.W., Al-Rawi, Y., Lees, K.R., Rowley, H.A., Sachara, C., Soehngen, M., Warach, S., Hacke, W., DEDAS Investigators, 2006. Dose escalation of desmoteplase for acute ischemic stroke (DEDAS): evidence of safety and efficacy 3 to 9 hours after stroke onset. Stroke 37, 1227–1231.

    Article  PubMed  CAS  Google Scholar 

  • Gulba, D.C., Praus, M., Witt, W., 1995. DSPA alpha – properties of the plasminogen activators of the vampire bat Desmodus rotundus. Fibrinolysis 9(Suppl.), 91–96.

    CAS  Google Scholar 

  • Hacke, W., Kaste, M., Fieschi, C., von Kummer, R., Davalos, A., Meier, D,. Larrue, V., Bluhmki, E., Davis, S., Donnan, G., Schneider, D., Diez-Tejedor, E., Trouillas, P., 1998. Randomised double-blind placebo-controlled trial of thrombolytic therapy with intravenous alteplase in acute ischaemic stroke (ECASS II). Second European-Australasian Acute Stroke Study Investigators. Lancet, 352: 1245–1251.

    Article  CAS  Google Scholar 

  • Hacke, W., Albers, G., Al-Rawi, Y., Bogousslavsky, J., Davalos, A., Eliasziw, M., Fischer, M., Furlan, A., Kaste, M., Lees, K.R., Soehngen, M., Warach, S., for The DIAS Study Group, 2005. The desmoteplase in acute ischemic stroke trial (DIAS). A phase II MRI-based 9-hour window acute stroke thrombolysis trial with intravenous desmoteplase. Stroke 36, 66–73

    Article  PubMed  CAS  Google Scholar 

  • Hacke, W., Furlan, A.J., Al-Rawi, Y., Davalos, A., Fiebach, J.B., Gruber, F., Kaste, M., Lipka, L.J., Pedraza, S., Ringleb, P.A., Rowley, H.A., Schneider, D., Schwamm, L.H., Leal, J.S., Söhngen, M., Teal, P.A., Wilhelm-Ogunbiyi, K., Wintermark, M., Warach, S., 2009. Intravenous desmoteplase in patients with acute ischaemic stroke selected by MRI perfusion-diffusion weighted imaging or perfusion CT (DIAS-2): a prospective, randomised, double-blind, placebo-controlled study. Lancet Neurol. 8, 141–50

    Article  PubMed  CAS  Google Scholar 

  • Kassner, A., Roberts, T.P., Moran, B., Silver, F.L., Mikulis, D.J., 2009. Recombinant tissue plasminogen activator increases blood-brain barrier disruption in acute ischemic stroke: an MR imaging permeability study. Am. J. Neuroradiol. 30, 1864–1869.

    Article  PubMed  CAS  Google Scholar 

  • Keyt, B.A., Paoni, N.F., Refino, C.J., Berleau, L., Nguyen, H., Chow, A., Lai, J., Peña, L., Pater, C., Ogez, J., Etcheverry, T., Botstzein, D., Bennett, W.F., 1994. A faster-acting and more potent form of tissue plasminogen activator. Proc. Natl. Acad. Sci. U.S.A. 91, 3670–3674.

    Article  PubMed  CAS  Google Scholar 

  • Kidwell, C.S., Latour, L., Saver, J.L., Alger, J.R., Starkman, S., Duckwiler, G., Jahan, R., Vinuela, F., UCLA Thrombolysis Investigators, Kang, D.W., Warach, S., 2008. Thrombolytic toxicity: blood brain barrier disruption in human ischemic stroke. Cerebrovasc. Dis. 25, 338–343.

    Article  PubMed  CAS  Google Scholar 

  • Krätzschmar, J., Haendler, B., Langer, G., Boidol, W., Bringmann, P., Alagon, A., Donner, P., Schleuning, W.D., 1991. The plasminogen activator family from the salivary gland of the vampire bat Desmodus rotundus: cloning and expression. Gene 105, 229–237.

    Article  PubMed  Google Scholar 

  • Kruithof, E.K., Schleuning, W.D., 2004. A comparative study of amyloid-beta (1–42) as a cofactor for plasminogen activation by vampire bat plasminogen activator and recombinant human tissue-type plasminogen activator. Thromb. Haemost. 92, 559–567.

    PubMed  CAS  Google Scholar 

  • Liberatore, G.T., Samson, A., Bladin, C., Schleuning, W.D., Medcalf, R.L., 2003. Vampire bat salivary plasminogen activator (desmoteplase): a unique fibrinolytic enzyme that does not promote neurodegeneration. Stroke 34, 537–543.

    Article  PubMed  CAS  Google Scholar 

  • López-Atalaya, J.P., Roussel, B.D., Ali, C., Maubert, E., Petersen, K.-U., Berezowski, V., Cecchelli, R., Orset, C., Vivien, D., 2007. Recombinant Desmodus rotundus salivary plasminogen activator (desmoteplase) crosses the blood-brain barrier through a LDL receptor-related protein (LRP)-dependent mechanism without exerting neurotoxic effects. Stroke 38, 1036–1043.

    Article  PubMed  Google Scholar 

  • López-Atalaya, J.P., Roussel, B.D., Levrat, D., Parcq, J., Nicole, O., Hommet, Y., Benchenane, K., Castel, H., Leprince, J., van To, D., Bureau, R., Rault, S., Vaudry, H., Petersen, K.-U., Santos, J.S., Ali, C., Vivien, D., 2008. Toward safer thrombolytic agents in stroke: molecular requirements for NMDA receptor-mediated neurotoxicity. J. Cereb. Blood Flow Metab. 28, 1212–1221.

    Article  PubMed  Google Scholar 

  • Multicenter Acute Stroke Trial-Europe Study Group, 1996. Thrombolytic therapy with streptokinase in acute ischemic stroke. N. Engl. J. Med. 335, 145–150.

    Article  Google Scholar 

  • Muschick, P., Zeggert, D., Donner, P., Witt, W., 1993. Thrombolytic properties of Desmodus (vampire bat) salivary plasminogen activator DSPA alpha1, alteplase and streptokinase following intravenous bolus injection in a rabbit model of carotid artery thrombosis. Fibrinolysis 7, 284–290

    CAS  Google Scholar 

  • NINDS. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group, 1995. Tissue plasminogen activator for acute ischemic stroke. N. Engl. J. Med. 33, 1581–1587.

    Google Scholar 

  • Petersen, K.-U., 2007. Thrombolytics – a field in development. Riv. It. Neurobiologia 53, 7–14.

    Google Scholar 

  • Reddrop, C., Moldrich, R.X., Beart, P.M., Farso, M., Liberatore, G.T., Howells, D.W., Petersen, K.-U., Schleuning, W.D., Medcalf, R.L., 2005. Vampire bat salivary plasminogen activator (desmoteplase) inhibits tissue-type plasminogen activator-induced potentiation of excitotoxic injury. Stroke 36, 1241–1246.

    Article  PubMed  CAS  Google Scholar 

  • Renatus, M., Stubbs, M.T., Huber, R., Bringmann, P., Donner, P., Schleuning, W.D., Bode, W., 1997. Catalytic domain structure of vampire bat plasminogen activator: a molecular paradigm for proteolysis without activation cleavage. Biochemistry 36, 3483–3493.

    Article  Google Scholar 

  • Research Report A315, 1992. Lysis of human plasma and whole blood clots in vitro by ZK 152 387 (DSPA alpha1), ZK 153 786 (DSPA alpha2), ZK 155 278 (DSPA beta), ZK 153 858 (alteplase), ZK 130 932 (scu-PA), ZK 113 905 (urokinase), and ZK 156 552 (anistreplase). (PAION, data on file)

    Google Scholar 

  • Research Report PN01 PCD 00002/01, 2001. Comparing the therapeutic benefit of desmoteplase, reteplase, and alteplase in a rat model of embolic middle cerebral artery (MCA) occlusion. (PAION, data on file)

    Google Scholar 

  • Schering Report, Gulba, D., 2002. rDSPA alpha1 in the establishment of early patency in myocardial infarction. (PAION, data on file)

    Google Scholar 

  • Schleuning, W.D., Alagon, A., Boidol, W., Bringmann, P., Petri, T., Krätzschmar, J., Haendler, B., Langer, G., Baldus, B., Witt, W., Donner, P., 1992. Plasminogen activators from the saliva of Desmodus rotundus (common vampire bat): unique fibrin specificity. Ann. N. Y. Acad. Sci. 667, 395–403.

    Article  PubMed  CAS  Google Scholar 

  • Schleuning, W.D., 2001. Vampire bat plasminogen activator DSPA-alpha-1 (desmoteplase): a thrombolytic drug optimized by natural selection. Haemostasis 31, 118–122.

    PubMed  CAS  Google Scholar 

  • Stewart, R.J., Fredenburgh, J.C., Weitz, J.I., 1998. Characterization of the interactions of plasminogen and tissue and vampire bat plasminogen activators with fibrinogen, fibrin, and the complex of D-dimer noncovalently linked to fragment E. J. Biol. Chem. 273, 18292–18299.

    Article  PubMed  CAS  Google Scholar 

  • Sobel, B.E., Geltman, E.M., Tiefenbrunn, A.J., Jaffe, A.S., Spadaro, J.J. Jr., Ter-Pogossian, M.M., Collen, D., Ludbrook, P.A., 1984. Improvement of regional myocardial metabolism after coronary thrombolysis induced with tissue-type plasminogen activator or streptokinase. Circulation 69, 983–690.

    Article  PubMed  CAS  Google Scholar 

  • Su, E.J., Fredriksson, L., Geyer, M., Folestad, E., Cale, J., Andrae, J., Gao, Y., Pietras, K., Mann, K., Yepes, M., Strickland, D.K., Betsholtz, C., Eriksson, U., Lawrence, D.A., 2008. Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat. Med. 14, 731–737.

    Article  PubMed  CAS  Google Scholar 

  • Su, E.J., Fredriksson, L., Schielke, G.P., Eriksson, U., Lawrence, D.A., 2009. Tissue plasminogen activator-mediated PDGF signaling and neurovascular coupling in stroke. J. Thromb. Haemost. 7(Suppl. 1), 155–158.

    Article  PubMed  CAS  Google Scholar 

  • Tebbe, U., Bramlage, P., Graf, A., Lechleitner, P., Bode, C., Riess, F.C., Clemens, N., Al-Rawi, Y., Konstantinides, S., Goldhaber, S.Z., 2009. Desmoteplase in acute massive pulmonary thromboembolism. Thromb. Haemost. 101, 557–562.

    PubMed  CAS  Google Scholar 

  • Toschi, L., Bringmann, P., Petri, T., Donner, P., Schleuning, W.D., 1998. Fibrin selectivity of the isolated protease domains of tissue-type and vampire bat salivary gland plasminogen activators. Eur. J. Biochem. 252, 108–112.

    Article  PubMed  CAS  Google Scholar 

  • Tsirka, S.E., 2002. Tissue plasminogen activator as a modulator of neuronal survival and function. Biochem. Soc. Trans. 30, 222–225.

    Article  PubMed  CAS  Google Scholar 

  • Yepes, M., Sandkvist, M., Moore, E.G., Bugge, T.H., Strickland, D.K., Lawrence, D.A., 2003. Tissue-type plasminogen activator induces opening of the blood-brain barrier via the LDL receptor-related protein. J. Clin. Invest. 112, 1533–1540.

    PubMed  CAS  Google Scholar 

  • Yepes, M., Roussel, B.D., Ali, C., Vivien, D., 2009. Tissue-type plasminogen activator in the ischemic brain: more than a thrombolytic. Trends Neurosci. 32, 48–55.

    Article  PubMed  CAS  Google Scholar 

  • Witt, W., Balds, B., Bringmann, P., Cashon, L., Donner, P., Schleuning, W.D., 1992. Thrombolytic properties of Desmodus rotundus (vampire bat) salivary plasminogen activator in experimental pulmonary embolism in rats. Blood 79, 1213–1217.

    PubMed  CAS  Google Scholar 

  • Witt, W., Maass, B., Baldus, B., Hildebrand, M., Donner, P., Schleuning, W.D., 1994. Coronary thrombolysis with Desmodus salivary plasminogen activator in dogs. Fast and persistent recanalization by intravenous bolus administration. Circulation 90, 421–426.

    CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wolfgang Söhngen .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Springer Science+Business Media B.V.

About this chapter

Cite this chapter

Söhngen, W., Petersen, KU., Söhngen, M. (2010). Bat Plasminogen Activator: Desmoteplase – From Bat to Bench to Bedside of Stroke Victims. In: Kini, R., Clemetson, K., Markland, F., McLane, M., Morita, T. (eds) Toxins and Hemostasis. Springer, Dordrecht. https://doi.org/10.1007/978-90-481-9295-3_23

Download citation

Publish with us

Policies and ethics