Semin intervent Radiol 2005; 22(2): 121-129
DOI: 10.1055/s-2005-871867
Copyright © 2005 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA.

Differentiating Pharmacologic Agents Used In Catheter-Directed Thrombolysis

James L. Swischuk1 , H. Bob Smouse1
  • 1Department of Radiology, University of Illinois College of Medicine at Peoria, Peoria, Illinois
Further Information

Publication History

Publication Date:
28 June 2005 (online)

ABSTRACT

The use of catheter-directed thrombolysis is a proven treatment for arterial ischemia, deep vein thrombosis, and severe pulmonary embolism. For arterial ischemia, thrombolysis has resulted in improved amputation-free survival and fewer subsequent surgeries to reestablish blood flow to the ischemic limb. The management of patients with thromboembolic diseases is complex, and the multiple thrombolytic drugs available to choose from compound this complexity. Although some believe the available thrombolytic agents are interchangeable, real biochemical differences exist that may prove otherwise. This article describes these pharmacologic differences and how they may affect the clinical practice of catheter-directed thrombolysis.

REFERENCES

  • 1 Ouriel K, Shortell C K, DeWeese J A et al.. A comparison of thrombolytic therapy with operative revascularization in the initial treatment of acute peripheral arterial ischemia.  J Vasc Surg. 1994;  19 1021-1030
  • 2 Weaver F A, Comerota A J, Youngblood M, Froehlich J, Hosking J D, Papanicolaou G. Surgical revascularization versus thrombolysis for nonembolic lower extremity native artery occlusions: results of a prospective randomized trial. The STILE Investigators. Surgery versus Thrombolysis for Ischemia of the Lower Extremity.  J Vasc Surg. 1996;  24 513-521 discussion 521-523
  • 3 Ouriel K, Veith F J, Sasahara A A. Thrombolysis or peripheral arterial surgery: phase I results. TOPAS Investigators.  J Vasc Surg. 1996;  23 64-73 discussion 74-75
  • 4 Ouriel K, Veith F J, Sasahara A A. A comparison of recombinant urokinase with vascular surgery as initial treatment for acute arterial occlusion of the legs. Thrombolysis or Peripheral Arterial Surgery (TOPAS) Investigators.  N Engl J Med. 1998;  338 1105-1111
  • 5 Tillet W S, Garner R L. The fibrinolytic activity of hemolytic streptococci.  J Exp Med. 1933;  58 485-502
  • 6 Christensen L R, MacLeod C M. A proteolytic enzyme of serum: characterization, activation and reaction with inhibitors.  J Gen Physiol. 1945;  28 559-583
  • 7 Bell W R. Therapeutic agents: pharmacokinetics and pharmacodynamics.  Rev Cardiovasc Med. 2002;  3(suppl 2) s34-s44
  • 8 van Breda A, Katzen B T, Deutsch A S. Urokinase versus streptokinase in local thrombolysis.  Radiology. 1987;  165 109-111
  • 9 Graor R A, Young J R, Risius B, Ruschhaupt W F. Comparison of cost effectiveness of streptokinase and urokinase in the treatment of deep vein thrombosis.  Ann Vasc Surg. 1987;  1 524-528
  • 10 Tennant S N, Dixon J, Venable T C et al.. Intracoronary thrombolysis in patients with acute myocardial infarction: comparison of the efficacy of urokinase with streptokinase.  Circulation. 1984;  69 756-760
  • 11 Williams J RB. The fibrinolytic activity of the urine.  Br J Exp Pathol. 1951;  32 530-537
  • 12 Sobel G W, Mohler S R, Jones N W et al.. Urokinase: an activator of plasma profibrinolysin extracted from urine.  Am J Physiol. 1952;  171 768-769
  • 13 Barlow G H, Lazer L. Characterization of the plasminogen activator isolated from human embryo kidney cells: comparison with urokinase.  Thromb Res. 1972;  1 201-208
  • 14 Bernik M B, White W F, Oller E P, Kwaan H C. Immunologic identity of plasminogen activator in human urine, heart, blood vessels, and tissue culture.  J Lab Clin Med. 1974;  84 546-558
  • 15 Mewissen M W, Seabrook G R, Meissner M H, Cynamon J, Labropoulos N, Haughton S H. Catheter-directed thrombolysis for lower extremity deep venous thrombosis: report of a national multicenter registry.  Radiology. 1999;  211 39-49
  • 16 Cragg A H, Smith T P, Corson J D et al.. Two urokinase dose regimens in native arterial and graft occlusions: initial results of a prospective, randomized clinical trial.  Radiology. 1991;  178 681-686
  • 17 Lijnen H R, Van Hoef B, De Cock F, Collen D. The mechanism of plasminogen activation and fibrin dissolution by single chain urokinase-type plasminogen activator in a plasma milieu in vitro.  Blood. 1989;  73 1864-1872
  • 18 Ouriel K, Kandarpa K, Schuerr D M, Hultquist M, Hodkinson G, Wallin B. Prourokinase versus urokinase for recanalization of peripheral occlusions, safety and efficacy: the PURPOSE trial.  J Vasc Interv Radiol. 1999;  10 1083-1091
  • 19 Swischuk J L, Fox P F, Young K et al.. Transcatheter intraarterial infusion of rt-PA for acute lower limb ischemia: results and complications.  J Vasc Interv Radiol. 2001;  12 423-430
  • 20 Wooster M B, Luzier A B. Reteplase: a new thrombolytic for the treatment of acute myocardial infarction.  Ann Pharmacother. 1999;  33 318-324
  • 21 Martin U, Sponer G, Strein K. Evaluation of thrombolytic and systemic effects of the novel recombinant plasminogen activator BM 06.022 compared with alteplase, anistreplase, streptokinase and urokinase in a canine model of coronary artery thrombosis.  J Am Coll Cardiol. 1992;  19 433-440
  • 22 Bode C, Smalling R W, Berg G et al.. Randomized comparison of coronary thrombolysis achieved with double-bolus reteplase (recombinant plasminogen activator) and front-loaded, accelerated alteplase (recombinant tissue plasminogen activator) in patients with acute myocardial infarction. The RAPID II Investigators.  Circulation. 1996;  94 891-898
  • 23 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
  • 24 Stewart R J, Fredenburgh J C, Leslie B A, Keyt B A, Rischke J A, Weitz J I. Identification of the mechanism responsible for the increased fibrin specificity of TNK-tissue plasminogen activator relative to tissue plasminogen activator.  J Biol Chem. 2000;  275 10112-10120
  • 25 Burkart D J, Borsa J J, Anthony J P, Thurlo S R. Thrombolysis of occluded peripheral arteries and veins with tenecteplase: a pilot study.  J Vasc Interv Radiol. 2002;  13 1099-1102
  • 26 Razavi M K, Wong H, Kee S T, Sze D Y, Semba C P, Dake M D. Initial clinical results of tenecteplase (TNK) in catheter-directed thrombolytic therapy.  J Endovasc Ther. 2002;  9 593-598
  • 27 Castaneda F, Swischuk J L, Li R, Young K, Smouse B, Brady T. Declining-dose study of reteplase treatment for lower extremity arterial occlusions.  J Vasc Interv Radiol. 2002;  13 1093-1098
  • 28 Chesebro J H, Knatterud G, Roberts R et al.. Thrombolysis in Myocardial Infarction (TIMI) Trial, Phase I: a comparison between intravenous tissue plasminogen activator and intravenous streptokinase. Clinical findings through hospital discharge.  Circulation. 1987;  76 142-154
  • 29 McLeod D C, Coln W G, Thayer C F, Perfetto E M, Hartzema A G. Pharmacoepidemiology of bleeding events after use of r-alteplase or streptokinase in acute myocardial infarction.  Ann Pharmacother. 1993;  27 956-962
  • 30 The GUSTO Investigators . An international randomized trial comparing four thrombolytic strategies for acute myocardial infarction.  N Engl J Med. 1993;  329 673-682
  • 31 Collaborative Group . ISIS-3: a randomised comparison of streptokinase vs tissue plasminogen activator vs anistreplase and of aspirin plus heparin vs aspirin alone among 41,299 cases of suspected acute myocardial infarction. ISIS-3 (Third International Study of Infarct Survival).  Lancet. 1992;  339 753-770
  • 32 The International Study Group . In-hospital mortality and clinical course of 20,891 patients with suspected acute myocardial infarction randomised between alteplase and streptokinase with or without heparin.  Lancet. 1990;  336 71-75
  • 33 Collaborative Group . Randomized trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. ISIS-2 (Second International Study of Infarct Survival).  J Am Coll Cardiol. 1988;  12(suppl A) 3A-13A
  • 34 Berkowitz S D, Granger C B, Pieper K S et al.. Incidence and predictors of bleeding after contemporary thrombolytic therapy for myocardial infarction. The Global Utilization of Streptokinase and Tissue Plasminogen activator for Occluded coronary arteries (GUSTO) I Investigators.  Circulation. 1997;  95 2508-2516
  • 35 Ouriel K, Kandarpa K. Safety of thrombolytic therapy with urokinase or recombinant tissue plasminogen activator for peripheral arterial occlusion: a comprehensive compilation of published work.  J Endovasc Ther. 2004;  11 436-446
  • 36 Rao A K, Pratt C, Berke A et al.. 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
  • 37 Bovill E G, Terrin M L, Stump D C et al.. Hemorrhagic events during therapy with recombinant tissue-type plasminogen activator, heparin, and aspirin for acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI), Phase II Trial.  Ann Intern Med. 1991;  115 256-265
  • 38 Weitz J I, Leslie B, Ginsberg J. Soluble fibrin degradation products potentiate tissue plasminogen activator-induced fibrinogen proteolysis.  J Clin Invest. 1991;  87 1082-1090
  • 39 Weitz J I, Rischke J. The role of (DD)E in the kinetics of plasminogen activation by tissue plasminogen activator.  Thromb Haemost. 1993;  69 544 , Abstract
  • 40 Pizzo S V, Schwartz M L, Hill R L, McKee P A. The effect of plasmin on the subunit structure of human fibrinogen.  J Biol Chem. 1972;  247 636-645
  • 41 Weitz J I, Koehn J A, Canfield R E, Landman S L, Friedman R. Development of a radioimmunoassay for the fibrinogen-derived peptide B beta 1-42.  Blood. 1986;  67 1014-1022
  • 42 Shen L L, McDonagh R P, McDonagh J, Hermans J. Early events in the plasmin digestion of fibrinogen and fibrin. Effects of plasmin on fibrin polymerization.  J Biol Chem. 1977;  252 6184-6189
  • 43 Sato H, Weisel J W. Polymerization of fibrinogen-derived fragment X and subsequent rearrangement of fibers.  Thromb Res. 1990;  58 205-212
  • 44 Weitz J I, Leslie B, Hirsh J, Klement P. Alpha 2-antiplasmin supplementation inhibits tissue plasminogen activator-induced fibrinogenolysis and bleeding with little effect on thrombolysis.  J Clin Invest. 1993;  91 1343-1350
  • 45 Owen J, Friedman K D, Grossman B A, Wilkins C, Berke A D, Powers E R. Quantitation of fragment X formation during thrombolytic therapy with streptokinase and tissue plasminogen activator.  J Clin Invest. 1987;  79 1642-1647
  • 46 Sugimoto K, Hofmann L V, Razavi M K et al.. The safety, efficacy, and pharmacoeconomics of low-dose alteplase compared with urokinase for catheter-directed thrombolysis of arterial and venous occlusions.  J Vasc Surg. 2003;  37 512-517
  • 47 Grunwald M R, Hofmann L V. Comparison of urokinase, alteplase, and reteplase for catheter-directed thrombolysis of deep venous thrombosis.  J Vasc Interv Radiol. 2004;  15 347-352
  • 48 Ouriel K, Gray B, Clair D G, Olin J. Complications associated with the use of urokinase and recombinant tissue plasminogen activator for catheter-directed peripheral arterial and venous thrombolysis.  J Vasc Interv Radiol. 2000;  11 295-298
  • 49 Cina C S, Goh R H, Chan J et al.. Intraarterial catheter-directed thrombolysis: urokinase versus tissue plasminogen activator.  Ann Vasc Surg. 1999;  13 571-575
  • 50 Semba C P, Bakal C W, Calis K A et al.. Alteplase as an alternative to urokinase. Advisory Panel on Catheter-Directed Thrombolytic Therapy.  J Vasc Interv Radiol. 2000;  11 279-287

James L SwischukM.D. 

Department of Radiology, University of Illinois College of Medicine at Peoria

1 Illini Drive, Peoria, IL 61605

    >