Semin Thromb Hemost 2017; 43(03): 300-310
DOI: 10.1055/s-0036-1584130
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Assessing Safety of Thrombolytic Therapy

Cornelis Kluft
1   Good Biomarker Sciences, Leiden, The Netherlands
2   Unit for Thrombosis Research, Department of Public Health, University of Southern Denmark, Esbjerg, Denmark
,
Johannes J. Sidelmann
2   Unit for Thrombosis Research, Department of Public Health, University of Southern Denmark, Esbjerg, Denmark
,
Jørgen B. Gram
2   Unit for Thrombosis Research, Department of Public Health, University of Southern Denmark, Esbjerg, Denmark
3   Department of Clinical Biochemistry, Hospital of South West Denmark, Esbjerg, Denmark
› Author Affiliations
Further Information

Publication History

Publication Date:
06 June 2016 (online)

Abstract

Thrombolytic therapy involves thrombolytic agents administered to patients suffering from venous or arterial thrombosis. The therapy induces systemic effects interrelated with the thrombolytic agent used. Bleeding is a prominent complication of thrombolytic therapy. Exhaustion of coagulation factors, generation of excessive amounts of fibrin degradation products (FDPs), therapy-induced activation of coagulation, therapy-induced anticoagulation, and formation of new fibrin all illustrate the complexity of effects of the treatment and challenges the hemostatic balance in the patients. The therapy-induced effects can be modulated by parallel administration of anticoagulants. Risk assessment is mandatory prior to thrombolytic therapy. Anticoagulated and unconscious patients represent particular safety concerns, and should be fully evaluated. Several guidelines describe the choice of tests and their safety limits in relation to pretreatment evaluation of anticoagulated patients. Fibrinogen depletion and FDPs during treatment may be promising markers for the evaluation of bleeding risk posttreatment. Future risk assessment measures should focus on the dynamics of the hemostatic balance. Here, thromboelastography may be considered a tool addressing clot formation, fibrin structure, and fibrinolytic resistance in parallel. Suitable laboratory analysis performed shortly after treatment may help to recognize severe treatment-induced systemic effects that can be counteracted by rational treatment, thereby reducing bleeding risk.

 
  • References

  • 1 Kramer MC, van der Wal AC, Koch KT , et al. Histopathological features of aspirated thrombi after primary percutaneous coronary intervention in patients with ST-elevation myocardial infarction. PLoS ONE 2009; 4 (6) e5817
  • 2 Rittersma SZ, van der Wal AC, Koch KT , et al. Plaque instability frequently occurs days or weeks before occlusive coronary thrombosis: a pathological thrombectomy study in primary percutaneous coronary intervention. Circulation 2005; 111 (9) 1160-1165
  • 3 Longstaff C, Varjú I, Sótonyi P , et al. Mechanical stability and fibrinolytic resistance of clots containing fibrin, DNA, and histones. J Biol Chem 2013; 288 (10) 6946-6956
  • 4 Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood 2014; 123 (18) 2768-2776
  • 5 Oklu R, Albadawi H, Watkins MT, Monestier M, Sillesen M, Wicky S. Detection of extracellular genomic DNA scaffold in human thrombus: implications for the use of deoxyribonuclease enzymes in thrombolysis. J Vasc Interv Radiol 2012; 23 (5) 712-718
  • 6 Karnabatidis D, Spiliopoulos S, Tsetis D, Siablis D. Quality improvement guidelines for percutaneous catheter-directed intra-arterial thrombolysis and mechanical thrombectomy for acute lower-limb ischemia. Cardiovasc Intervent Radiol 2011; 34 (6) 1123-1136
  • 7 Brommer EJ. The level of extrinsic plasminogen activator (t-PA) during clotting as a determinant of the rate of fibrinolysis; inefficiency of activators added afterwards. Thromb Res 1984; 34 (2) 109-115
  • 8 Zamarron C, Lijnen HR, Collen D. Influence of exogenous and endogenous tissue-type plasminogen activator on the lysability of clots in a plasma milieu in vitro. Thromb Res 1984; 35 (3) 335-345
  • 9 Kluft CMP, Ersdal E, Rosen S. Tissue-Type Plasminogen Activator (t-PA) Activity. Dordrecht: Kluwer Academic Publisher; 1999
  • 10 Hilberg T, Prasa D, Stürzebecher J, Gläser D, Schneider K, Gabriel HH. Blood coagulation and fibrinolysis after extreme short-term exercise. Thromb Res 2003; 109 (5–6) 271-277
  • 11 Hrafnkelsdóttir T, Ottosson P, Gudnason T, Samuelsson O, Jern S. Impaired endothelial release of tissue-type plasminogen activator in patients with chronic kidney disease and hypertension. Hypertension 2004; 44 (3) 300-304
  • 12 Hoffmeister HM, Szabo S, Helber U, Seipel L. The thrombolytic paradox. Thromb Res 2001; 103 (Suppl. 01) S51-S55
  • 13 Collen D, Bounameaux H, De Cock F, Lijnen HR, Verstraete M. Analysis of coagulation and fibrinolysis during intravenous infusion of recombinant human tissue-type plasminogen activator in patients with acute myocardial infarction. Circulation 1986; 73 (3) 511-517
  • 14 Michels R, Hoffmann H, Windeler J, Barth H, Hopkins G. A double-blind multicenter comparison of the efficacy and safety of saruplase and urokinase in the treatment of acute myocardial infarction: Report of the sutami study group. J Thromb Thrombolysis 1995; 2 (2) 117-124
  • 15 Goto S, Kawai Y, Abe S , et al. Serial changes in coagulant activities after thrombolytic therapy for acute myocardial infarction. Angiology 1994; 45 (4) 273-281
  • 16 Gardell SJ, Ramjit DR, Stabilito II , et al. Effective thrombolysis without marked plasminemia after bolus intravenous administration of vampire bat salivary plasminogen activator in rabbits. Circulation 1991; 84 (1) 244-253
  • 17 Collen D, Zamarron C, Lijnen HR, Hoylaerts M. Activation of plasminogen by pro-urokinase. II. Kinetics. J Biol Chem 1986; 261 (3) 1259-1266
  • 18 Zamarron C, Lijnen HR, Collen D. Kinetics of the activation of plasminogen by natural and recombinant tissue-type plasminogen activator. J Biol Chem 1984; 259 (4) 2080-2083
  • 19 Gurewich V. Why so little progress in therapeutic thrombolysis? The current state of the art and prospects for improvement. J Thromb Thrombolysis 2015; 40 (4) 480-487
  • 20 Weitz JI. Limited fibrin specificity of tissue-type plasminogen activator and its potential link to bleeding. J Vasc Interv Radiol 1995; 6 (6, Pt 2, Suppl): 19S-23S
  • 21 Wiman B, Rånby M. Determination of soluble fibrin in plasma by a rapid and quantitative spectrophotometric assay. Thromb Haemost 1986; 55 (2) 189-193
  • 22 Francis CW, Kornberg A. Fibrinogen- and fibrin-degradation products during fibrinolytic therapy. Ann N Y Acad Sci 1992; 667: 310-323
  • 23 Weitz JI, Leslie B, Ginsberg J. Soluble fibrin degradation products potentiate tissue plasminogen activator-induced fibrinogen proteolysis. J Clin Invest 1991; 87 (3) 1082-1090
  • 24 Koppert PW, Kuipers W, Hoegee-de Nobel B, Brommer EJ, Koopman J, Nieuwenhuizen W. A quantitative enzyme immunoassay for primary fibrinogenolysis products in plasma. Thromb Haemost 1987; 57 (1) 25-28
  • 25 Soria J, Soria C, Mirshahi M , et al. A specific marker of thrombolysis: DDE complex [in French]. C R Acad Sci III 1987; 304 (11) 307-311
  • 26 Kluft C, Los P, Jie AF , et al. The mutual relationship between the two molecular forms of the major fibrinolysis inhibitor alpha-2-antiplasmin in blood. Blood 1986; 67 (3) 616-622
  • 27 Wiman B, Collen D. On the kinetics of the reaction between human antiplasmin and plasmin. Eur J Biochem 1978; 84 (2) 573-578
  • 28 Hamilton KK, Fretto LJ, Grierson DS, McKee PA. Effects of plasmin on von Willebrand factor multimers. Degradation in vitro and stimulation of release in vivo. J Clin Invest 1985; 76 (1) 261-270
  • 29 Holmberg L, Ljung R, Nilsson IM. The effects of plasmin and protein Ca on factor VIII:C and VIII:CAg. Thromb Res 1983; 31 (1) 41-50
  • 30 Nogami K, Shima M, Matsumoto T, Nishiya K, Tanaka I, Yoshioka A. Mechanisms of plasmin-catalyzed inactivation of factor VIII: a crucial role for proteolytic cleavage at Arg336 responsible for plasmin-catalyzed factor VIII inactivation. J Biol Chem 2007; 282 (8) 5287-5295
  • 31 Tracy RP, Rubin DZ, Mann KG , et al. Thrombolytic therapy and proteolysis of factor V. J Am Coll Cardiol 1997; 30 (3) 716-724
  • 32 Omar MN, Mann KG. Inactivation of factor Va by plasmin. J Biol Chem 1987; 262 (20) 9750-9755
  • 33 Kalafatis M, Mann KG. The role of the membrane in the inactivation of factor va by plasmin. Amino acid region 307-348 of factor V plays a critical role in factor Va cofactor function. J Biol Chem 2001; 276 (21) 18614-18623
  • 34 Samis JA, Ramsey GD, Walker JB, Nesheim ME, Giles AR. Proteolytic processing of human coagulation factor IX by plasmin. Blood 2000; 95 (3) 943-951
  • 35 Pryzdial EL, Lavigne N, Dupuis N, Kessler GE. Plasmin converts factor X from coagulation zymogen to fibrinolysis cofactor. J Biol Chem 1999; 274 (13) 8500-8505
  • 36 Cocho D, Borrell M, Martí-Fàbregas J , et al. Pretreatment hemostatic markers of symptomatic intracerebral hemorrhage in patients treated with tissue plasminogen activator. Stroke 2006; 37 (4) 996-999
  • 37 Sun X, Berthiller J, Derex L, Trouillas P, Diallo L, Hanss M. Post-thrombolysis haemostasis changes after rt-PA treatment in acute cerebral infarct. Correlations with cardioembolic aetiology and outcome. J Neurol Sci 2015; 349 (1–2) 77-83
  • 38 Martí-Fàbregas J, Borrell M, Cocho D , et al. Hemostatic markers of recanalization in patients with ischemic stroke treated with rt-PA. Neurology 2005; 65 (3) 366-370
  • 39 Weitz JI, 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 (4) 1343-1350
  • 40 Gurewich V, Pannell R. Recombinant human C1-inhibitor prevents non-specific proteolysis by mutant pro-urokinase during optimal fibrinolysis. Thromb Haemost 2009; 102 (2) 279-286
  • 41 Korninger C, Collen D. Studies on the specific fibrinolytic effect of human extrinsic (tissue-type) plasminogen activator in human blood and in various animal species in vitro. Thromb Haemost 1981; 46 (2) 561-565
  • 42 Mutch NJ, Moore NR, Mattsson C, Jonasson H, Green AR, Booth NA. The use of the Chandler loop to examine the interaction potential of NXY-059 on the thrombolytic properties of rtPA on human thrombi in vitro. Br J Pharmacol 2008; 153 (1) 124-131
  • 43 Stewart D, Kong M, Novokhatny V, Jesmok G, Marder VJ. Distinct dose-dependent effects of plasmin and TPA on coagulation and hemorrhage. Blood 2003; 101 (8) 3002-3007
  • 44 Pettigrew LC, Dobbs MR. Stroke: thrombolysis and antithrombotic therapy. In: D. J. Moliterno, S. D. Kristensen, R. De Caterina eds. Therapeutic Advances in Thrombosis, 2nd ed. Oxford, UK: Blackwell Publishing Ltd; 2012
  • 45 Andreotti F, Kluft C, Hackett DR, Davies GJ, Maseri A. Thrombin generation after fast or prolonged regimens of tissue-type plasminogen activator. Lancet 1993; 342 (8876): 937-938
  • 46 Andreotti F, Kluft C, Davies GJ, Hackett DR, Prevost R, Maseri A. Prolonged coagulation instability is associated with a higher-dose regimen of tissue-type plasminogen activator in patients with acute myocardial infarction. Ann N Y Acad Sci 1992; 667: 450-453
  • 47 Sánchez PL, Fernández-Avilés F. An integrated approach to the management of patients after the early phase of ST segment elevation myocardial infarction. In: S Yusuf, JA Cairns, AJ Camm, EL Fallen, BJ Gersh. Evidence-Based Cardiology, 3rd ed. Oxford, UK: Wiley-Blackwell; 2009
  • 48 Adeoye O, Sucharew H, Khoury J , et al. Combined approach to lysis utilizing eptifibatide and recombinant tissue-type plasminogen activator in acute ischemic stroke-full dose regimen stroke trial. Stroke 2015; 46 (9) 2529-2533
  • 49 Pancioli AM, Adeoye O, Schmit PA , et al; CLEAR-ER Investigators. Combined approach to lysis utilizing eptifibatide and recombinant tissue plasminogen activator in acute ischemic stroke-enhanced regimen stroke trial. Stroke 2013; 44 (9) 2381-2387
  • 50 Bouma BN, Meijers JC. Thrombin-activatable fibrinolysis inhibitor (TAFI, plasma procarboxypeptidase B, procarboxypeptidase R, procarboxypeptidase u). J Thrombosis Haemostasis 2003; 1 (7) 1566-1574
  • 51 Minnema MC, Peters RJ, de Winter R , et al. Activation of clotting factors XI and IX in patients with acute myocardial infarction. Arterioscler Thromb Vasc Biol 2000; 20 (11) 2489-2493
  • 52 Hagedorn I, Schmidbauer S, Pleines I , et al. Factor XIIa inhibitor recombinant human albumin Infestin-4 abolishes occlusive arterial thrombus formation without affecting bleeding. Circulation 2010; 121 (13) 1510-1517
  • 53 Leung PY, Hurst S, Berny-Lang MA , et al. Inhibition of factor xii-mediated activation of factor xi provides protection against experimental acute ischemic stroke in mice. Transl Stroke Res 2012; 3 (3) 381-389
  • 54 Schmaier AH. Extracorporeal circulation without bleeding. Sci Transl Med 2014; 6 (222) 222fs7
  • 55 Tucker EI, Marzec UM, White TC , et al. Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI. Blood 2009; 113 (4) 936-944
  • 56 Fernandez-Cadenas I, Alvarez-Sabin J, Ribo M , et al. Influence of thrombin-activatable fibrinolysis inhibitor and plasminogen activator inhibitor-1 gene polymorphisms on tissue-type plasminogen activator-induced recanalization in ischemic stroke patients. J Thromb Haemost 2007; 5 (9) 1862-1868
  • 57 Sasaki T, Yoshimoto N, Sugimoto K , et al. Intravenous and oral administrations of DD2 [7-Amino-2-(sulfanylmethyl)heptanoic acid] produce thrombolysis through inhibition of plasma TAFIa in rats with tissue factor-induced microthrombosis. Thromb Res 2012; 130 (4) e222-e228
  • 58 Hendrickx ML, Zatloukalova M, Hassanzadeh-Ghassabeh G, Muyldermans S, Gils A, Declerck PJ. In vitro and in vivo characterisation of the profibrinolytic effect of an inhibitory anti-rat TAFI nanobody. Thromb Haemost 2014; 111 (5) 824-832
  • 59 Nagashima M, Werner M, Wang M , et al. An inhibitor of activated thrombin-activatable fibrinolysis inhibitor potentiates tissue-type plasminogen activator-induced thrombolysis in a rabbit jugular vein thrombolysis model. Thromb Res 2000; 98 (4) 333-342
  • 60 Merlini PA, Cugno M, Rossi ML , et al. Activation of the contact system and inflammation after thrombolytic therapy in patients with acute myocardial infarction. Am J Cardiol 2004; 93 (7) 822-825
  • 61 Pönitz V, Pritchard D, Grundt H, Nilsen DW. Specific types of activated factor XII increase following thrombolytic therapy with tenecteplase. J Thromb Thrombolysis 2006; 22 (3) 199-203
  • 62 Agostoni A, Gardinali M, Frangi D , et al. Activation of complement and kinin systems after thrombolytic therapy in patients with acute myocardial infarction. A comparison between streptokinase and recombinant tissue-type plasminogen activator. Circulation 1994; 90 (6) 2666-2670
  • 63 Ewald GA, Eisenberg PR. Plasmin-mediated activation of contact system in response to pharmacological thrombolysis. Circulation 1995; 91 (1) 28-36
  • 64 Munkvad S, Jespersen J, Gram J, Kluft C. Depression of factor XII-dependent fibrinolytic activity characterizes patients with early myocardial reinfarction after recombinant tissue-type plasminogen activator therapy. J Am Coll Cardiol 1991; 18 (2) 454-458
  • 65 Müller F, Gailani D, Renné T. Factor XI and XII as antithrombotic targets. Curr Opin Hematol 2011; 18 (5) 349-355
  • 66 Klement P, Liao P, Bajzar L. A novel approach to arterial thrombolysis. Blood 1999; 94 (8) 2735-2743
  • 67 Vercauteren E, Gils A, Declerck PJ. Thrombin activatable fibrinolysis inhibitor: a putative target to enhance fibrinolysis. Semin Thromb Hemost 2013; 39 (4) 365-372
  • 68 Hashimoto M, Yamashita T, Oiwa K, Watanabe S, Giddings JC, Yamamoto J. Enhancement of endogenous plasminogen activator-induced thrombolysis by argatroban and APC and its control by TAFI, measured in an arterial thrombolysis model in vivo using rat mesenteric arterioles. Thromb Haemost 2002; 87 (1) 110-113
  • 69 Williams JE, Hantgan RR, Hermans J, McDonagh J. Characterization of the inhibition of fibrin assembly by fibrinogen fragment D. Biochem J 1981; 197 (3) 661-668
  • 70 Huynh T, Cox JL, Massel D , et al; FASTRAK II Network. Predictors of intracranial hemorrhage with fibrinolytic therapy in unselected community patients: a report from the FASTRAK II project. Am Heart J 2004; 148 (1) 86-91
  • 71 Fiumara K, Kucher N, Fanikos J, Goldhaber SZ. Predictors of major hemorrhage following fibrinolysis for acute pulmonary embolism. Am J Cardiol 2006; 97 (1) 127-129
  • 72 Emberson J, Lees KR, Lyden P , et al; Stroke Thrombolysis Trialists' Collaborative Group. Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: a meta-analysis of individual patient data from randomised trials. Lancet 2014; 384 (9958): 1929-1935
  • 73 Jauch EC, Saver JL, Adams Jr HP , et al; American Heart Association Stroke Council; Council on Cardiovascular Nursing; Council on Peripheral Vascular Disease; Council on Clinical Cardiology. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2013; 44 (3) 870-947
  • 74 Minematsu K, Toyoda K, Hirano T , et al; Japan Stroke Society. Guidelines for the intravenous application of recombinant tissue-type plasminogen activator (alteplase), the second edition, October 2012: a guideline from the Japan Stroke Society. J Stroke Cerebrovasc Dis 2013; 22 (5) 571-600
  • 75 Kitchen S, Gray E, Mackie I, Baglin T, Makris M ; BCSH committee. Measurement of non-coumarin anticoagulants and their effects on tests of haemostasis: guidance from the British Committee for Standards in Haematology. Br J Haematol 2014; 166 (6) 830-841
  • 76 Mackie I, Cooper P, Lawrie A, Kitchen S, Gray E, Laffan M ; British Committee for Standards in Haematology. Guidelines on the laboratory aspects of assays used in haemostasis and thrombosis. Int J Lab Hematol 2013; 35 (1) 1-13
  • 77 CLSI. Collection, Transport, and Processing of Blood Specimens for Testing Plasma-Based Coagulation Assays and Molecular Hemostasis Assays; Approved Guideline. 5th ed. CLSI Document h21-a5. Wayne, PA: Clinical and Laboratory Standards Institute; 2008
  • 78 Drescher MJ, Spence A, Rockwell D, Staff I, Smally AJ. Point-of-care testing for coagulation studies in a stroke protocol: a time-saving innovation. Am J Emerg Med 2011; 29 (1) 82-85
  • 79 Poller L, Keown M, Chauhan N , et al; ECCA Steering Group Members. European Concerted Action on Anticoagulation. Correction of displayed international normalized ratio on two point-of-care test whole-blood prothrombin time monitors (CoaguChek Mini and TAS PT-NC) by independent international sensitivity index calibration. Br J Haematol 2003; 122 (6) 944-949
  • 80 Poller L. International Normalized Ratios (INR): the first 20 years. J Thromb Haemost 2004; 2 (6) 849-860
  • 81 Mazya MV, Lees KR, Markus R , et al; Safe Implementation of Thrombolysis in Stroke Investigators. Safety of intravenous thrombolysis for ischemic stroke in patients treated with warfarin. Ann Neurol 2013; 74 (2) 266-274
  • 82 Nilsson JB, Boman K, Jansson JH, Nilsson T, Näslund U. The influence of acute-phase levels of haemostatic factors on reperfusion and mortality in patients with acute myocardial infarction treated with streptokinase. J Thromb Thrombolysis 2008; 26 (3) 188-195
  • 83 Mani H, Herth N, Kasper A , et al. Point-of-care coagulation testing for assessment of the pharmacodynamic anticoagulant effect of direct oral anticoagulant. Ther Drug Monit 2014; 36 (5) 624-631
  • 84 Diener HC, Foerch C, Riess H, Röther J, Schroth G, Weber R. Treatment of acute ischaemic stroke with thrombolysis or thrombectomy in patients receiving anti-thrombotic treatment. Lancet Neurol 2013; 12 (7) 677-688
  • 85 Tan K, Booth D, Newell SJ, Dear PR, Hughes C, Richards M. Point-of-care testing of neonatal coagulation. Clin Lab Haematol 2006; 28 (2) 117-121
  • 86 Kate M, Szkotak A, Witt A, Shuaib A, Butcher K. Proposed approach to thrombolysis in dabigatran-treated patients presenting with ischemic stroke. J Stroke Cerebrovasc Dis 2014; 23 (6) 1351-1355
  • 87 De Luca R, Fontana P, Poncet A, de Moerloose P, Pfister RE. Evaluation of the GEM®PCL Plus point-of-care device for neonatal coagulation assessment: an observational study on cord blood. Thromb Res 2014; 134 (2) 474-478
  • 88 Du S, Harenberg J, Krämer S, Krämer R, Wehling M, Weiss C. Measurement of non-vitamin k antagonist oral anticoagulants in patient plasma using heptest-stat coagulation method. Ther Drug Monit 2015; 37 (3) 375-380
  • 89 Ebner M, Peter A, Spencer C , et al. Point-of-care testing of coagulation in patients treated with non-vitamin k antagonist oral anticoagulants. Stroke 2015; 46 (10) 2741-2747
  • 90 Harenberg J, Du S, Krämer S , et al. Novel methods for assessing oral direct factor Xa and thrombin inhibitors: use of point-of-care testing and urine samples. Semin Thromb Hemost 2013; 39 (1) 66-71
  • 91 Harenberg J, Du S, Wehling M , et al. Measurement of dabigatran, rivaroxaban and apixaban in samples of plasma, serum and urine, under real life conditions. An international study. Clin Chem Lab Med 2016; 54 (2) 275-283
  • 92 Shepherd MF, Jacobsen JM, Rosborough TK. Argatroban therapy using enzymatic anti-factor IIa monitoring. Ann Pharmacother 2011; 45 (3) 422-423
  • 93 Dias JD, Norem K, Doorneweerd DD, Thurer RL, Popovsky MA, Omert LA. Use of thromboelastography (teg) for detection of new oral anticoagulants. Arch Pathol Lab Med 2015; 139 (5) 665-673
  • 94 Bowry R, Fraser S, Archeval-Lao JM , et al. Thrombelastography detects the anticoagulant effect of rivaroxaban in patients with stroke. Stroke 2014; 45 (3) 880-883
  • 95 Adelmann D, Wiegele M, Wohlgemuth RK , et al. Measuring the activity of apixaban and rivaroxaban with rotational thrombelastometry. Thromb Res 2014; 134 (4) 918-923
  • 96 Kitchen DP, Kitchen S, Jennings I, Woods T, Walker I. Quality assurance and quality control of thrombelastography and rotational Thromboelastometry: the UK NEQAS for blood coagulation experience. Semin Thromb Hemost 2010; 36 (7) 757-763
  • 97 Matosevic B, Knoflach M, Werner P , et al. Fibrinogen degradation coagulopathy and bleeding complications after stroke thrombolysis. Neurology 2013; 80 (13) 1216-1224
  • 98 Vandelli L, Marietta M, Gambini M , et al. Fibrinogen decrease after intravenous thrombolysis in ischemic stroke patients is a risk factor for intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2015; 24 (2) 394-400
  • 99 Skeik N, Gits CC, Ehrenwald E, Cragg AH. Fibrinogen level as a surrogate for the outcome of thrombolytic therapy using tissue plasminogen activator for acute lower extremity intravascular thrombosis. Vasc Endovascular Surg 2013; 47 (7) 519-523
  • 100 Saito M, Nakabayashi T, Iuchi K , et al. Effects of direct percutaneous transluminal coronary angioplasty treatment of acute myocardial infarction on plasma levels of haemostatic and fibrinolytic factors. Blood Coagul Fibrinolysis 1993; 4 (5) 801-804
  • 101 Ho CH, Wang SP. Serial thrombolysis-related changes after thrombolytic therapy with TPA in patients with acute myocardial infarction. Thromb Res 1990; 58 (3) 331-341
  • 102 Trouillas P, Derex L, Philippeau F , et al. Early fibrinogen degradation coagulopathy is predictive of parenchymal hematomas in cerebral rt-PA thrombolysis: a study of 157 cases. Stroke 2004; 35 (6) 1323-1328
  • 103 Meng R, Ji X, Li B, Zhou J, Li W, Ding Y. Dynamical levels of plasma F(1+2) and D-dimer in patients with acute cerebral infarction during intravenous urokinase thrombolysis. Neurol Res 2009; 31 (4) 367-370
  • 104 Ueda T, Hatakeyama T, Sakaki S, Ohta S, Kumon Y, Uraoka T. Changes in coagulation and fibrinolytic system after local intra-arterial thrombolysis for acute ischemic stroke. Neurol Med Chir (Tokyo) 1995; 35 (3) 136-143
  • 105 Madsen DE, Ingerslev J, Sidelmann JJ, Thorn JJ, Gram J. Intraoperative blood loss during orthognathic surgery is predicted by thromboelastography. J Oral Maxillofac Surg 2012; 70 (10) e547-e552
  • 106 Kozek-Langenecker SA, Afshari A, Albaladejo P , et al. Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 2013; 30 (6) 270-382
  • 107 Wikkelsø A, Lunde J, Johansen M , et al. Fibrinogen concentrate in bleeding patients. Cochrane Database Syst Rev 2013; 8: CD008864
  • 108 Larsen OH, Fenger-Eriksen C, Ingerslev J, Sørensen B. Improved point-of-care identification of hyperfibrinolysis is needed. Thromb Res 2012; 130 (4) 690-691