Thromb Haemost 1999; 82(02): 974-982
DOI: 10.1055/s-0037-1615941
Research Article
Schattauer GmbH

Mechanism of Action of Plasminogen Activators

Jeffrey I. Weitz
1   From the Department of Medicine, McMaster University and Hamilton Civic Hospitals Research Centre Hamilton, Ontario, CANADA
,
Ronald J. Stewart
1   From the Department of Medicine, McMaster University and Hamilton Civic Hospitals Research Centre Hamilton, Ontario, CANADA
,
James C. Fredenburgh
1   From the Department of Medicine, McMaster University and Hamilton Civic Hospitals Research Centre Hamilton, Ontario, CANADA
› Author Affiliations
Further Information

Publication History

Publication Date:
09 December 2017 (online)

Introduction

Acute coronary ischemic syndromes and stroke are usually caused by thrombosis in arteries where obstruction leads to ischemia of the heart or brain, respectively. Likewise, venous thrombosis predisposes to pulmonary emboli that cause infarction of lung tissue by blocking pulmonary arteries. Although antithrombotic drugs form the cornerstone of treatment of established thrombosis, pharmacologic lysis of fibrin thrombi, using plasminogen activators, is a widely used approach for treatment of acute myocardial infarction and selected cases of stroke or venous thromboembolism.

Plasminogen activators cause thrombus dissolution by initiating fibrinolysis (Fig. 1). The fibrinolytic system is comprised of inactive plasminogen, which is converted to plasmin by plasminogen activators.1 Plasmin, a trypsin-like serine protease, degrades fibrin into soluble fibrin degradation products. The fibrinolytic system is regulated to provide efficient localized activation of plasminogen on the fibrin surface, yet prevent systemic plasminogen activation. To localize plasminogen activation to the fibrin surface, both plasminogen and tissue-type plasminogen activator (t-PA), the major initiator of intravascular fibrinolysis, bind to fibrin. Plasminogen activator inhibitors,2 the most important of which is type-1 plasminogen activator inhibitor (PAI-1), prevent excessive plasminogen activation by t-PA and urokinase-type plasminogen activator (u-PA). Systemic plasmin is rapidly inhibited by α2-antiplasmin, whereas plasmin generated on the fibrin surface is relatively protected from inactivation by α2-antiplasmin.3

The beneficial effect of thrombolytic therapy reflects dissolution of fibrin within occlusive thrombi and subsequent restoration of antegrade blood flow. Bleeding, the major side effect of thrombolytic therapy, occurs because plasmin is a relatively nonspecific enzyme that does not distinguish between fibrin in occlusive thrombi and fibrin in hemostatic plugs. In addition, circulating plasmin also degrades fibrinogen and other clotting factors, a phenomenon known as the systemic lytic state. Although the contribution of the systemic lytic state to bleeding remains controversial, much attention has focussed on the development of plasminogen activators that produce thrombolysis without depleting circulating fibrinogen in the hope that agents with greater fibrin-specificity will produce less bleeding.

In addition to causing bleeding, currently available plasminogen activators have other limitations. Despite aggressive dosing regimens and adjunctive antithrombotic drugs, up to 25% of coronary thrombi are resistant to thrombolysis at 60 to 90 minutes. Early thrombotic reocclusion of previously opened coronary arteries further reduces the benefits of thrombolytic therapy.4-6 These problems have triggered the quest for more potent thrombolytic agents that have the potential to overcome factors that render some thrombi resistant to lysis. Furthermore, to simplify administration, plasminogen activators with longer half-lives have been developed so that bolus dosing is possible.

This chapter reviews the mechanism of action of currently available plasminogen activators, including agents with greater fibrin-specificity, longer half-lives, and a potential for increased thrombolytic potency.

 
  • References

  • 1 Bachmann F. The plasminogen-plasmin enzyme system. In: Colman RW, Hirsh J, Marder VJ, Salzman EW. eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 3rd ed.. Philadelphia: JB Lipincott Co; 1994: 1592-1622.
  • 2 Kruithof EKO, Baker MS, Bunn CL. Biological and clinical aspects of plasminogen activator inhibitor type. Blood 1995; 86: 4007-4024.
  • 3 Lijnen HR, Collen D. Alpha 2-antiplasmin. In: Barrett AJ, Salvesen G. eds. Proteinase Inhibitors. New York: Elsevier Science Publishers BV; 1986: 457-476.
  • 4 Lincoff AM, Topol EJ. Illusion of reperfusion. Does anyone achieve optimal reperfusion during acute myocardial infarction?. Circulation 1993; 88: 1361-1374.
  • 5 GUSTO Investigators. The effects of tissue plasminogen activator, streptokinase, or both on coronary-artery patency, ventricular function, and survival after acute myocardial infarction. N Engl J Med 1993; 329: 1615-1622.
  • 6 Verstraete M, Arnold AE, Brower RW, Collen D, De Bono DP, de Zwaan C, Erbel R, Hillis WS, Lennane RJ, Lubsen J. Acute coronary thrombolysis with recombinant human tissue-type plasminogen activator: initial patency and influence of maintained infusion on reocclusion rate. Am J Cardiol 1987; 60: 231-237.
  • 7 Smalling RW. Molecular biology of plasminogen activators: what are the clinical implications of drug design?. Am J Cardiol 1996; 78: 2-7.
  • 8 Hoylaerts M, Rijken DC, Lijnen HR, Collen D. Kinetics of the activation of plasminogen by human tissue plasminogen activator. Role of fibrin. J Biol Chem 1982; 257: 2912-2919.
  • 9 Horrevoets AJ, Pannekoek H, Nesheim ME. A steady-state template model that describes the kinetics of fibrin-stimulated [Glu1]- and [Lys78]-plasminogen activation by native tissue-type plasminogen activator and variants that lack either the finger or kringle-2 domain. J Biol Chem 1997; 272: 2183-2191.
  • 10 Wiman B, Lijnen HR, Collen D. On the specific interaction between the lysine-binding sites in plasmin and complementary sites in alpha2-antiplasmin and in fibrinogen. Biochim Biophys Acta 1979; 579: 142-154.
  • 11 Ranby M. Studies on the kinetics of plasminogen activation by tissue plasminogen activator. Biochim Biophys Acta 1982; 704: 461-469.
  • 12 Collen D, Gold HK. Fibrin-specific thrombolytic agents and new approaches to coronary arterial thrombolysis. In: Julian D, Kubler W, Norris RM, Swan HJC, Collen D, Verstraete M. eds. Thrombolysis in Cardiovascular Disease. New York: Marcel Decker Inc; 1990: 45-67.
  • 13 Stewart RJ, Fredenburgh JC, Weitz JI. 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 1998; 273: 18292-18299.
  • 14 Weitz JI, Leslie B, Ginsberg J. Soluble fibrin degradation products potentiate tissue plasminogen activator-induced fibrinogen proteolysis. J Clin Invest 1991; 87: 1082-1090.
  • 15 Olexa SA, Budzynski AZ. Binding phenomena of isolated unique plasmic degradation products of human cross-linked fibrin. J Biol Chem 1979; 254: 4925-4932.
  • 16 Love TW, Quertermous T, Runge MS, Michelson KD, Matsueda GR, Haber E. Attachment of an antifibrin antibody to the amino terminus of tissue-type plasminogen activator impairs stimulation by fibrin. Fibrinolysis 1994; 8: 326-332.
  • 17 Holvoet P, Laroche Y, Lijnen HR, Cauwenbergte RV, Demarsin E, Brouwers E, Matthyssens G, Collen D. Characterization of a chimeric plasminogen activator consisting of a single-chain Fv fragment derived from a fibrin fragment D-dimer-specific antibody and a truncated single-chain urokinase. J Biol Chem 1991; 266: 19717-19724.
  • 18 Dewerchin M, Lijnen HR, Stassen JM, De Cock F, Quertermous T, Ginsberg MH, Plow EF, Collen D. Effect of chemical conjugation of recombinant single-chain urokinase-type plasminogen activator with monoclonal antiplatelet antibodies on platelet aggregation and on plasma clot lysis in vitro and in vivo. Blood 1991; 78: 1005-1018.
  • 19 Lijnen HR, Collen D. Molecular and cellular basis of fibrinolysis. In: Hoffman R, Benz EJ, Shattil SJ, Furie B, Cohen HJ, Silberstein LE. eds. Hematology. Basic principles and practice. 2nd ed.. New York: Churchill Livingstone Inc; 1995: 1588-1596.
  • 20 Madison EL, Sambrook JF. Probing structure-function relationships of tissue-type plasminogen activator by oligonucleotide-mediated site-specific mutagenesis. Methods Enzymol 1993; 223: 249-271.
  • 21 Lijnen HR, Collen D. Strategies for the improvement of thrombolytic agents. Thromb Haemost 1991; 66: 88-110.
  • 22 Tate KM, Higgins DL, Holmes WE, Winkler ME, Heyneker HL, Vehar GA. Functional role of proteolytic cleavage at arginine-275 of human tissue plasminogen activator as assessed by site-directed mutagenesis. Biochemistry 1987; 26: 338-343.
  • 23 Verheijen JH, Caspers MPM, Chang GTG, de Munk GAW, Pouwels PH, Enger-Valk BE. Involvement of finger domain and kringle 2 domain of tissue-type plasminogen activator in fibrin binding and stimulation of activity by fibrin. EMBO J 1986; 5: 3525-3530.
  • 24 Urano S, Metzger AR, Castellino FJ. Plasmin-mediated fibrinolysis by variant recombinant tissue plasminogen activators. Proc Natl Acad Sci USA 1989; 86: 2568-2571.
  • 25 Bringmann P, Gruber D, Liese A, Toschi L, Krätzschmar J, Schleuning WD, Donner P. Structural features mediating fibrin selectivity of vampire bat plasminogen activators. J Biol Chem 1995; 270: 25596-25603.
  • 26 Rijken DC, Otter M, Kuiper J, van Berkel TJ. Receptor-mediated endocytosis of tissue-type plasminogen activator (t-PA) by liver cells. Thromb Res 1990; (suppl X) 63-71.
  • 27 Camani C, Kruithof EKO. The role of the finger and growth factor domains in the clearance of tissue-type plasminogen activator by hepatocytes. J Biol Chem 1995; 270: 26053-26056.
  • 28 Larsen GR, Metzger M, Henson K, Blue Y, Horgan P. Pharmacokinetic and distribution analysis of variant forms of tissue-type plasminogen activator with prolonged clearance in rat. Blood 1989; 73: 1842-1850.
  • 29 Stockinger H, Kubbies M, Rudolph R, Stern A, Kohnert U, Fischer S. Binding of recombinant variants of human tissue-type plasminogen activator (t-PA) to human umbilical vein endothelial cells. Thromb Res 1992; 67: 589-599.
  • 30 Beebe DP, Miles LA, Plow EF. A linear amino acid sequence involved in the interaction of t-PA with its endothelial cell receptor. Blood 1989; 74: 2034-2037.
  • 31 Hajjar KA. The endothelial cell tissue plasminogen activator receptor specific interaction with plasminogen. J Biol Chem 1991; 266: 21962-21970.
  • 32 Kohnert U, Rudolph R, Verheijen JH, Weening-Verhoeff EJD, Stern A, Optiz U, Martin U, Lill H, Prinz H, Lechner M, Kresse GB, Buckel P, Fischer S. Biochemical properties of the kringle 2 and protease domains are maintained in the refolded t-PA deletion variant BM 06.022. Protein Eng 1992; 5: 93-100.
  • 33 Smalling RW. Pharmacological and clinical impact of the unique molecular structure of a new plasminogen activator. Eur Heart J 1997; 18 (suppl F) 11-16.
  • 34 Smalling RW, Bode C, Kalbfleisch J, Sen S, Limbourg P, Forycki F, Habib G, Feldman R, Hohnloser S, Seals A. More rapid, complete, and stable coronary thrombolysis with bolus administration of reteplase compared with alteplase infusion in acute myocardial infarction. RAPID Investigators. Circulation 1995; 91: 2725-2732.
  • 35 GUSTO Investigators. A comparison of reteplase with alteplase for acute myocardial infarction. The global use of strategies to open occluded coronary arteries (GUSTO III) investigators. N Engl J Med 1997; 337: 1118-1123.
  • 36 Keyt BA, Paoni NF, Refino CJ, Berleau L, Nguyen H, Chow A, Lai J, Pena L, Pater C, Ogez J. A faster-acting and more potent form of tissue plasminogen activator. Proc Natl Acad Sci USA 1994; 91: 3670-3674.
  • 37 Paoni NF, Keyt BA, Refino CJ, Chow AM, Nguyen HV, Berleau LT, Badillo J, Peña LC, Brady K, Wurm FM, Ogez J, Bennett WF. A slow clearing, fibrin-specific, PAI-1 resistant variant of t-PA (T103N, KHRR 296-299 AAAA). Thromb Haemost 1993; 70: 307-312.
  • 38 Refino CJ, Paoni NF, Keyt BA, Pater CS, Badillo JM, Wurm FM, Ogez J, Bennett WF. A variant of t-PA (T103N, KHRR 296-299 AAAA) that, by bolus, has increased potency and decreased systemic activation of plasminogen. Thromb Haemost 1993; 70: 313-319.
  • 39 Stewart RJ, Fredenburgh JC, Keyt BA, Weitz JI. The fibrin-specificities of tissue-type plasminogen activator and, to a lesser extent, the TNK variant are compromised by kringle-dependent interactions with (DD)E. Blood 1997; 90: 144. Abstract.
  • 40 Stewart RJ, Fredenburgh JC, Leslie BA, Keyt BA, Weitz JI. The fibrin-specificity of plasminogen activators is compromised by their interactions with (DD)E. Thromb Haemost 1997; (Suppl) 193.
  • 41 Cannon CP, McCabe CH, Gibson CM, Ghali M, Sequeira RF, McKendall GR, Breed J, Modi NB, Fox NL, Tracy RP, Love TW, Braunwald E. TNK-tissue plasminogen activator in acute myocardial infarction. Results of the thrombolysis in myocardial infarction (TIMI) 10A dose-ranging trial. Circulation 1997; 95: 351-356.
  • 42 Cannon CP, Gibson M, McCabe CH, Adgey AA, Schweiger MJ, Sequeira RF, Grollier G, Giugliano RP, Frey M, Mueller HS, Steingart RM, Weaver WD, Van de Werf F, Braunwald E. TNK-Tissue plasminogen activator compared with front loaded Alteplase in acute myocardial infarction. Results of the TIMI 10B trial. Circulation 1998; 98: 2805-2814.
  • 43 Larsen GR, Timony GA, Horgan PG, Barone KM, Henson KS, Angus LB, Stoudemire JB. Protein engineering of novel plasminogen activators with increased thrombolytic potency in rabbits relative to activase. J Biol Chem 1991; 266: 8156-8161.
  • 44 Rijken DC, Emeis JJ, Gerwig GJ. On the composition and function of the carbohydrate moiety of tissue-type plasminogen activator from human melanoma cells. Thromb Haemost 1985; 54: 788-791.
  • 45 Nesheim ME, Fredenburgh JC, Larsen GR. The dissociation constants and stoichiometries of the interactions of Lys-plasminogen and chloromethyl ketone derivatives of tissue plasminogen activator and the variant dFEIX with intact fibrin. J Biol Chem 1990; 265: 21541-21548.
  • 46 den Heijer P, Vermeer F, Ambrosioni E, Sadowski Z, Lopez-Sendon JL, von Essen R, Beaufils P, Thadani U, Adgey J, Pierard L, Brinker J, Davies RF, Smalling RW, Wallentin L, Caspi A, Pangerl A, Trickett L, Hauck C, Henry D, Chew P. Evaluation of a weight-adjusted single-bolus plasminogen activator in patients with myocardial infarction: a double-blind, randomized angiographic trial of lanoteplase versus alteplase. Circulation 1997; 98: 2117-2125.
  • 47 Gardell SJ, Duong LT, Diehl RE, York JD, Hare TR, Register RB, Jacobs JW, Dixon RAF, Friedman PA. Isolation, characterization, and cDNA cloning of a vampire bat salivary plasminogen activator. J Biol Chem 1989; 264: 17947-17952.
  • 48 Gardell SJ, Hare TR, Bergum PW, Cuca GC, O’Neill-Palladino L, Zavodny SM. Vampire bat salivary plasminogen activator is quiescent in human plasma in the absence of fibrin unlike human tissue plasminogen activator. Blood 1990; 76: 2560-2564.
  • 49 Gardell SJ, Ramjit DR, Stabilito II, Fujita T, Lynch JJ, Cuca GC, Jain D, Wang SP, Tung JS, Mark GE. Effective thrombolysis without marked plasminemia after bolus intravenous administration of vampire bat salivary plasminogen activator in rabbits. Circulation 1991; 84: 244-253.
  • 50 de Munk GAW, Rijken DC. Fibrinolytic properties of single chain urokinase-type plasminogen activator (pro-urokinase). Fibrinolysis 1990; 4: 1-8.
  • 51 Stump DC, Thienpont M, Collen D. Urokinase-related proteins in human urine. Isolation and characterization of single-chain urokinase (pro-urokinase) and urokinase-inhibitor complex. J Biol Chem 1986; 261: 1267-1273.
  • 52 Rijken DC, Binnema DJ, Los P. Specific fibrinolytic properties of different molecular forms of pro- urokinase from a monkey kidney cell culture. Thromb Res 1986; 42: 761-768.
  • 53 Wun TC, Schleuning WD, Reich E. Isolation and characterization of urokinase from human plasma. J Biol Chem 1982; 257: 3276-3283.
  • 54 Winkler ME, Blaber M. Purification and characterization of recombinant single-chain urokinase produced in Escherichia coli . Biochemistry 1986; 25: 4041-4045.
  • 55 Nelles L, Lijnen R, Collen D, Holmes WE. Characterization of recombinant human single chain urokinase-type plasminogen activator mutants produced by site-specific muatagnesis of Lysine 158. J Biol Chem 1987; 262: 5682-5689.
  • 56 Longstaff C, Clough AM, Gaffney PJ. Kinetics of plasmin activation of single chain urinary-type plasminogen activator (scu-PA) and demonstration of a high affinity interaction between scu-PA and plasminogen. J Biol Chem 1992; 267: 173-179.
  • 57 Ichinose A, Fujikawa K, Suyama T. The activation of pro-urokinase by plasma kallikrein and its inactivation by thrombin. J Biol Chem 1986; 261: 3486-3489.
  • 58 Behrendt N, Ronne E, Dano K. The structure and function of the urokinase receptor, a membrane protein governing plasminogen activation on the cell surface. Biol Chem Hoppe Seyler 1995; 376: 269-279.
  • 59 Weaver WD, Hartmann JR, Anderson JL, Reddy PS, Sobolski JC, Sasahara AA. New recombinant glycosylated prourokinase for treatment of patients with acute myocardial infarction. Prourokinase study group. J Am Coll Cardiol 1994; 24: 1242-1248.
  • 60 Gurewich V, Pannell R, Louie S, Kelley P, Suddith RL, Greenlee R. Effective and fibrin-specific clot lysis by a zymogen precursor form of urokinase (pro-urokinase). A study in vitro and in two animal species. J Clin Invest 1984; 73: 1731-1739.
  • 61 de Boer A, Kluft C, Gerloff J, Dooijewaard G, Gunzler WA, Beier H, van der Meer FJ, Cohen AF. Pharmacokinetics of saruplase, a recombinant unglycosylated human single-chain urokinase-type plasminogen activator and its effects on fibrinolytic and haemostatic parameters in healthy male subjects. Thromb Haemost 1993; 70: 320-325.
  • 62 Stump DC, Kieckens L, De Cock F, Collen D. Pharmacokinetics of single chain forms of urokinase-type plasminogen activator. J Pharmacol Exp Ther 1987; 242: 245-250.
  • 63 Henkin J, Marcotte P, Yang H. The plasminogen-plasmin system. Prog Cardiovasc Dis 1991; 34: 135-164.
  • 64 Marcotte PA, Henkin J, Credo RB, Badylak SF. A-chain isozymes of recombinant and natural urokinases: Preparation, characterization, and their biochemical and fibrinolytic properties. Fibrinolysis 1992; 6: 69-78.
  • 65 Lijnen HR, Nelles L, Holmes WE, Collen D. Biochemical and thrombolytic properties of a low molecular weight form (comprising Leu 144 through Leu 411) of recombinant single-chain urokinase-type plasminogen activator. J Biol Chem 1988; 263: 5594-5598.
  • 66 Nelles L, Lijnen HR, Collen D, Holmes WE. Characterization of a fusion protein consisting of amino acids 1 to 263 of tissue-type plasminogen activator and amino acids 144 to 411 of urokinase-type plasminogen activator. J Biol Chem 1987; 262: 10855-10862.
  • 67 Pierard L, Jacobs P, Gheysen D, Hoylaerts M, Andre B, Topisirovic L, Cravador A, de Foresta F, Herzog A, Collen D, De Wilde M, Bollen A. Mutant and chimeric recombinant plasminogen activators. Production in eukaryotic cells and preliminary characterization. J Biol Chem 1987; 262: 11771-11778.
  • 68 Langer-Safer PR, Ahern TJ, Angus LB, Barone KM, Brenner MJ, Horgan PG, Morris GE, Stoudemire JB, Timony GA, Larsen GR. Replacement of finger and growth factor domains of tissue plasminogen activator with plasminogen kringle 1. Biochemical and pharmacological characterization of a novel chimera containing a high affinity fibrin-binding domain linked to a heterologous protein. J Biol Chem 1991; 266: 3715-3723.
  • 69 Haber E, Bode C, Matsueda GR, Reed GL, Runge MS. Antibody targeting as a thrombolytic strategy. Ann NY Acad Sci 1992; 667: 365-381.
  • 70 Castellino FJ. A unique enzyme-protein substrate modifier reaction: plasmin/streptokinase interaction. Trends Biochem Sci 1979; 1-5.
  • 71 Collen D. On the regulation and control of fibrinolysis. Thromb Haemost 1980; 43: 77-89.
  • 72 Rao AK, Pratt C, Berke A, Jaffe A, Ockene I, Schreiber TL, Bell WR, Knatterud G, Robertson TL, Terrin ML. Thrombolysis in myocardial infarction (TIMI) trial—phase I: hemorrhagic manifestations and changes in plasma fibrinogen and the fibrinolytic system in patients treated with recombinant tissue plasminogen activator and streptokinase. J Am Coll Cardiol 1988; 11: 1-11.
  • 73 Francis CW, Marder VJ. Physiologic regulation and pathologic disorders of fibrinolysis. In: Colman RW, Hirsh J, Marder VJ, Salzman EW. eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 3rd ed.. Philadelphia: JB Lipincott Co; 1994: 1076-1103.
  • 74 Smith RA, Dupe RJ, English PD, Green J. Fibrinolysis with acylenzymes: a new approach to thrombolytic therapy. Nature 1981; 290: 505-508.
  • 75 Marder VJ, Hirsh J, Bell WR. Rationale and practical basis of thrombolytic therapy. In: Colman RW, Hirsh J, Marder VJ, Salzman EW. eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 3rd ed.. Philadelphia: JB Lipincott Co; 1994: 1514-1540.
  • 76 Collen D. Thrombolytic therapy. Thromb Haemost 1997; 78: 742-746.
  • 77 Collen D, Schlott B, Engelborghs Y, Van Hoef B, Hartmann M, Lijnen HR, Behnke D. On the mechanism of the activation of human plasminogen by recombinant staphylokinase. J Biol Chem 1993; 268: 8284-8289.
  • 78 Silence K, Collen D, Lijnen HR. Regulation by α2-antiplasmin and fibrin of the activation of plasminogen with recombinant staphylokinase in plasma. Blood 1993; 82: 1175-1183.
  • 79 Lijnen HR, Van Hoef B, De Cock F, Okada K, Ueshima S, Matsuo O, Collen D. On the mechanism of fibrin-specific plasminogen activation by staphylokinase. J Biol Chem 1991; 266: 11826-11832.
  • 80 Vanderschueren S, Barrios L, Kerdsinchai P, Van den Heuvel P, Hermans L, Vrolix M, De Man F, Benit E, Muyldermans L, Collen D. A randomized trial of recombinant staphylokinase versus alteplase for coronary artery patency in acute myocardial infarction. The STAR trial group. Circulation 1995; 92: 2044-2049.
  • 81 Vanderschueren SM, Stassen JM, Collen D. On the immunogenicity of recombinant staphylokinase in patients and in animal models. Thromb Haemost 1994; 72: 297-301.
  • 82 Collen D, Moreau H, Stockx L, Vanderschueren S. Recombinant staphylokinase variants with altered immunoreactivity. II: Thrombolytic properties and antibody induction. Circulation 1996; 94: 207-216.
  • 83 GUSTO Investigators. An international randomized trial comparing four thrombolytic strategies for acute myocardial infarction. The GUSTO investigators. N Engl J Med 1993; 329: 673-682.
  • 84 Cairns JA, Kennedy JW, Fuster V. Coronary thrombolysis. Chest 1998; 114: 634S-657S.
  • 85 Nesheim M, Wang W, Boffa M, Nagashima M, Morser J, Bajzar L. Thrombin, thrombomodulin and TAFI in the molecular link between coagulation and fibrinolysis. Thromb Haemost 1997; 78: 386-391.