Tranexamic acid-associated seizures: Causes and treatment

Irene Lecker, Dian-Shi Wang, Paul D Whissell, Sinziana Avramescu, C David Mazer, Beverley A Orser, Irene Lecker, Dian-Shi Wang, Paul D Whissell, Sinziana Avramescu, C David Mazer, Beverley A Orser

Abstract

Antifibrinolytic drugs are routinely used worldwide to reduce the bleeding that results from a wide range of hemorrhagic conditions. The most commonly used antifibrinolytic drug, tranexamic acid, is associated with an increased incidence of postoperative seizures. The reported increase in the frequency of seizures is alarming, as these events are associated with adverse neurological outcomes, longer hospital stays, and increased in-hospital mortality. However, many clinicians are unaware that tranexamic acid causes seizures. The goal of this review is to summarize the incidence, risk factors, and clinical features of these seizures. This review also highlights several clinical and preclinical studies that offer mechanistic insights into the potential causes of and treatments for tranexamic acid-associated seizures. This review will aid the medical community by increasing awareness about tranexamic acid-associated seizures and by translating scientific findings into therapeutic interventions for patients.

© 2015 The Authors Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Tranexamic acid (TXA) concentrations measured in the cerebral spinal fluid (CSF) and serum of patients cause hyperexcitability in vitro. (A) The time course of TXA levels in the CSF and serum of 1 patient who experienced a seizure is shown on the left. The decline of TXA levels in the brain lags behind that in the blood. The timeline at the bottom of each figure indicates key surgical events during cardiopulmonary bypass (CPB). The red arrow highlights the concentrations when TXA administration was terminated. On the right are the summarized data of TXA concentrations in the CSF and serum during key surgical events (n = 4). TXA levels in the serum (2mM) are 10‐fold higher than those in the CSF (200 µM). (B) Clinically relevant concentration of TXA (200 µM) causes hyperexcitability by increasing the frequency of seizure‐like events in neocortical slices. *P < 0.05.
Figure 2
Figure 2
Tranexamic acid (TXA) is a competitive antagonist of glycine (Gly) receptors. (A) Glycine and TXA are structural analogues, suggesting that TXA competes with glycine at the agonist binding site of glycine receptors. (B) TXA (1mM) inhibits glycine (100 µM)‐activated currents in cortical neurons. The concentration–response plots for glycine current recorded in the absence and presence of TXA are shown. The results indicate that TXA is a competitive antagonist of glycine receptors.
Figure 3
Figure 3
Tonic glycine current is highly sensitive to tranexamic acid (TXA) inhibition. (A) Inhibitory receptors are expressed in synaptic and extrasynaptic regions of the neuron. These receptors are composed of different subunits and have distinct pharmacological properties. Extrasynaptic receptors mediate a tonic inhibitory conductance. (B) Summary table of the half‐maximal inhibitory concentration (IC50) values for TXA inhibition of synaptic and tonic currents mediated by glycine and γ‐aminobutyric acid type A (GABA) receptors. (C) TXA (1mM) inhibits synaptic and tonic glycine currents in a similar manner as the competitive glycine antagonist, strychnine. Synaptic currents were studied by recording miniature inhibitory postsynaptic currents. Tonic currents were evoked by applying a low concentration of glycine (10 µM), similar to the ambient concentration present in the extracellular fluid, to the bath solution. SEM = standard error of the mean.
Figure 4
Figure 4
The molecular mechanism underlying tranexamic acid (TXA)‐associated seizures and the reversal of TXA‐mediated inhibition by anesthetics. TXA binds to the glycine receptors, resulting in a decrease in inhibitory current. This reduction in anion conduction increases excitability, which gives rise to seizures. Anesthetics reverse the effect of TXA by increasing glycine receptor function and thereby prevent or reverse TXA‐induced seizures.

References

    1. Henry DA, Carless PA, Moxey AJ, et al. Anti‐fibrinolytic use for minimising perioperative allogeneic blood transfusion. Cochrane Database Syst Rev 2007;(3):CD001886.
    1. Dunn CJ, Goa KL. Tranexamic acid: a review of its use in surgery and other indications. Drugs 1999;57:1005–1032.
    1. Roberts I, Shakur H, Afolabi A, et al. The importance of early treatment with tranexamic acid in bleeding trauma patients: an exploratory analysis of the CRASH‐2 randomised controlled trial. Lancet 2011;377:1096–1101.
    1. Hoylaerts M, Lijnen HR, Collen D. Studies on the mechanism of the antifibrinolytic action of tranexamic acid. Biochim Biophys Acta 1981;673:75–85.
    1. Iwamoto M. Plasminogen‐plasmin system IX. Specific binding of tranexamic acid to plasmin. Thromb Diath Haemorrh 1975;33:573–585.
    1. Thorsen S. Differences in the binding to fibrin of native plasminogen and plasminogen modified by proteolytic degradation. Influence of omega‐aminocarboxylic acids. Biochim Biophys Acta 1975;393:55–65.
    1. McEvoy MD, Reeves ST, Reves JG, et al. Aprotinin in cardiac surgery: a review of conventional and novel mechanisms of action. Anesth Analg 2007;105:949–962.
    1. Berman M, Cardone D, Sharples L, et al. Safety and efficacy of aprotinin and tranexamic acid in pulmonary endarterectomy surgery with hypothermia: review of 200 patients. Ann Thorac Surg 2010;90:1432–1436.
    1. Bertholini DM, Butler CS. Severe hyponatraemia secondary to desmopressin therapy in von Willebrand's disease. Anaesth Intensive Care 2000;28:199–201.
    1. Bhat A, Bhowmik DM, Vibha D, et al. Tranexamic acid overdosage‐induced generalized seizure in renal failure. Saudi J Kidney Dis Transpl 2014;25:130–132.
    1. Butala BP, Shah VR, Bhosale GP, et al. Medication error: subarachnoid injection of tranexamic acid. Indian J Anaesth 2012;56:168–170.
    1. de Leede‐van der Maarl MG, Hilkens P, Bosch F. The epileptogenic effect of tranexamic acid. J Neurol 1999;246:843.
    1. Garcha PS, Mohan CV, Sharma RM. Death after an inadvertent intrathecal injection of tranexamic acid. Anesth Analg 2007;104:241–242.
    1. Ichikawa J, Kodaka M, Nishiyama K, et al. A case of postoperative convulsive seizure following tranexamic acid infusion during aortic valve replacement. Masui 2013;62:186–189.
    1. Kaabachi O, Eddhif M, Rais K, et al. Inadvertent intrathecal injection of tranexamic acid. Saudi J Anaesth 2011;5:90–92.
    1. Kalavrouziotis D, Voisine P, Mohammadi S, et al. High‐dose tranexamic acid is an independent predictor of early seizure after cardiopulmonary bypass. Ann Thorac Surg 2012;93:148–154.
    1. Keyl C, Uhl R, Beyersdorf F, et al. High‐dose tranexamic acid is related to increased risk of generalized seizures after aortic valve replacement. Eur J Cardiothorac Surg 2011;39:114–121.
    1. Koster A, Borgermann J, Zittermann A, et al. Moderate dosage of tranexamic acid during cardiac surgery with cardiopulmonary bypass and convulsive seizures: incidence and clinical outcome. Br J Anaesth 2013;110:34–40.
    1. Mahmoud K, Ammar A. Accidental intrathecal injection of tranexamic acid. Case Rep Anesthesiol 2012;2012:646028.
    1. Manji RA, Grocott HP, Leake J, et al. Seizures following cardiac surgery: the impact of tranexamic acid and other risk factors. Can J Anaesth 2012;59:6–13.
    1. Martin K, Wiesner G, Breuer T, et al. The risks of aprotinin and tranexamic acid in cardiac surgery: a one‐year follow‐up of 1188 consecutive patients. Anesth Analg 2008;107:1783–1790.
    1. Merriman B, Mayson K, Sawka A, et al. Postoperative seizure in a neurosurgical patient: should tranexamic acid be on the differential? Can J Anaesth 2013;60:506–507.
    1. Mohseni K, Jafari A, Nobahar MR, et al. Polymyoclonus seizure resulting from accidental injection of tranexamic acid in spinal anesthesia. Anesth Analg 2009;108:1984–1986.
    1. Murkin JM, Falter F, Granton J, et al. High‐dose tranexamic acid is associated with nonischemic clinical seizures in cardiac surgical patients. Anesth Analg 2010;110:350–353.
    1. Sander M, Spies CD, Martiny V, et al. Mortality associated with administration of high‐dose tranexamic acid and aprotinin in primary open‐heart procedures: a retrospective analysis. Crit Care 2010;14:R148.
    1. Sharma V, Katznelson R, Jerath A, et al. The association between tranexamic acid and convulsive seizures after cardiac surgery: a multivariate analysis in 11 529 patients. Anaesthesia 2014;69:124–130.
    1. Wang CS, Yang CJ, Chen SC, et al. Generalized convulsion resulted in hyperammonemia during treatment with tranexamic acid for hemoptysis. Ir J Med Sci 2011;180:761–763.
    1. Wong JO, Yang SF, Tsai MH. Accidental injection of tranexamic acid (Transamin) during spinal anesthesia. Ma Zui Xue Za Zhi 1988;26:249–252.
    1. Yeh HM, Lau HP, Lin PL, et al. Convulsions and refractory ventricular fibrillation after intrathecal injection of a massive dose of tranexamic acid. Anesthesiology 2003;98:270–272.
    1. Casati V, Romano A, Novelli E, et al. Tranexamic acid for trauma. Lancet 2010;376:1049–1050.
    1. Jimenez JJ, Iribarren JL, Brouard M, et al. Safety and effectiveness of two treatment regimes with tranexamic acid to minimize inflammatory response in elective cardiopulmonary bypass patients: a randomized double‐blind, dose‐dependent, phase IV clinical trial. J Cardiothorac Surg 2011;6:138.
    1. Makhija N, Sarupria A, Kumar Choudhary S, et al. Comparison of epsilon aminocaproic acid and tranexamic acid in thoracic aortic surgery: clinical efficacy and safety. J Cardiothorac Vasc Anesth 2013;27:1201–1207.
    1. Rabinovici R, Heyman A, Kluger Y, et al. Convulsions induced by aminocaproic acid infusion. DICP 1989;23:780–781.
    1. Gofton TE, Chu MW, Norton L, et al. A prospective observational study of seizures after cardiac surgery using continuous EEG monitoring. Neurocrit Care 2014;21:220–227.
    1. Hunter GR, Young GB. Seizures after cardiac surgery. J Cardiothorac Vasc Anesth 2011;25:299–305.
    1. Sindet‐Pedersen S, Stenbjerg S. Effect of local antifibrinolytic treatment with tranexamic acid in hemophiliacs undergoing oral surgery. J Oral Maxillofac Surg 1986;44:703–707.
    1. Wellington K, Wagstaff AJ. Tranexamic acid: a review of its use in the management of menorrhagia. Drugs 2003;63:1417–1433.
    1. Ducloy‐Bouthors AS, Jude B, Duhamel A, et al. High‐dose tranexamic acid reduces blood loss in postpartum haemorrhage. Crit Care 2011;15:R117.
    1. Rahman Z, Hoque R, Ali A, et al. Blood conservation strategies for reducing peri‐operative blood loss in open heart surgery. Mymensingh Med J 2011;20:45–53.
    1. Sabovic M, Lavre J, Vujkovac B. Tranexamic acid is beneficial as adjunctive therapy in treating major upper gastrointestinal bleeding in dialysis patients. Nephrol Dial Transplant 2003;18:1388–1391.
    1. Zufferey PJ, Miquet M, Quenet S, et al. Tranexamic acid in hip fracture surgery: a randomized controlled trial. Br J Anaesth 2010;104:23–30.
    1. Roberts I, Kawahara T. Proposal for the inclusion of tranexamic acid (anti‐fibrinolytic‐lysine analogue) in the WHO model list of essential medicines. Geneva, Switzerland: World Health Organization, 2010.
    1. Fergusson DA, Hebert PC, Mazer CD, et al. A comparison of aprotinin and lysine analogues in high‐risk cardiac surgery. N Engl J Med 2008;358:2319–2331.
    1. Pellegrini A, Giaretta D, Chemello R, et al. Feline generalized epilepsy induced by tranexamic acid (AMCA). Epilepsia 1982;23:35–45.
    1. Schlag MG, Hopf R, Zifko U, et al. Epileptic seizures following cortical application of fibrin sealants containing tranexamic acid in rats. Acta Neurochir (Wien) 2002;144:63–69.
    1. Yamaura A, Nakamura T, Makino H, et al. Cerebral complication of antifibrinolytic therapy in the treatment of ruptured intracranial aneurysm. Animal experiment and a review of literature. Eur Neurol 1980;19:77–84.
    1. Lecker I, Wang DS, Romaschin AD, et al. Tranexamic acid concentrations associated with human seizures inhibit glycine receptors. J Clin Invest 2012;122:4654–4666.
    1. Estrera AL, Sheinbaum R, Miller CC, et al. Cerebrospinal fluid drainage during thoracic aortic repair: safety and current management. Ann Thorac Surg 2009;88:9–15.
    1. Kratzer S, Irl H, Mattusch C, et al. Tranexamic acid impairs γ‐aminobutyric acid receptor type A‐mediated synaptic transmission in the murine amygdala: a potential mechanism for drug‐induced seizures? Anesthesiology 2014;120:639–649.
    1. Olsen RW, Tobin AJ. Molecular biology of GABAA receptors. FASEB J 1990;4:1469–1480.
    1. Sigel E, Steinmann ME. Structure, function, and modulation of GABAA receptors. J Biol Chem 2012;287:40224–40231.
    1. Greenfield LJ Jr. Molecular mechanisms of antiseizure drug activity at GABAA receptors. Seizure 2013;22:589–600.
    1. Henschel O, Gipson KE, Bordey A. GABAA receptors, anesthetics and anticonvulsants in brain development. CNS Neurol Disord Drug Targets 2008;7:211–224.
    1. Sillito AM. The effectiveness of bicuculline as an antagonist of GABA and visually evoked inhibition in the cat's striate cortex. J Physiol 1975;250:287–304.
    1. Karkar KM, Thio LL, Yamada KA. Effects of seven clinically important antiepileptic drugs on inhibitory glycine receptor currents in hippocampal neurons. Epilepsy Res 2004;58:27–35.
    1. Lynch JW. Molecular structure and function of the glycine receptor chloride channel. Physiol Rev 2004;84:1051–1095.
    1. Furtmuller R, Schlag MG, Berger M, et al. Tranexamic acid, a widely used antifibrinolytic agent, causes convulsions by a γ‐aminobutyric acid A receptor antagonistic effect. J Pharmacol Exp Ther 2002;301:168–173.
    1. Chebib M, Johnston GA. GABA‐activated ligand gated ion channels: medicinal chemistry and molecular biology. J Med Chem 2000;43:1427–1447.
    1. Macdonald RL, Olsen RW. GABAA receptor channels. Annu Rev Neurosci 1994;17:569–602.
    1. Mody I. Distinguishing between GABAA receptors responsible for tonic and phasic conductances. Neurochem Res 2001;26:907–913.
    1. Lynch JW. Native glycine receptor subtypes and their physiological roles. Neuropharmacology 2009;56:303–309.
    1. Farrant M, Nusser Z. Variations on an inhibitory theme: phasic and tonic activation of GABAA receptors. Nat Rev Neurosci 2005;6:215–229.
    1. Parker AJ, Lee JB, Redman J, et al. Strychnine poisoning: gone but not forgotten. Emerg Med J 2011;28:84.
    1. Young AB, Snyder SH. Strychnine binding associated with glycine receptors of the central nervous system. Proc Natl Acad Sci U S A 1973;70:2832–2836.
    1. Downie DL, Hall AC, Lieb WR, et al. Effects of inhalational general anaesthetics on native glycine receptors in rat medullary neurones and recombinant glycine receptors in Xenopus oocytes. Br J Pharmacol 1996;118:493–502.
    1. Hales TG, Lambert JJ. The actions of propofol on inhibitory amino acid receptors of bovine adrenomedullary chromaffin cells and rodent central neurones. Br J Pharmacol 1991;104:619–628.
    1. Belelli D, Callachan H, Hill‐Venning C, et al. Interaction of positive allosteric modulators with human and Drosophila recombinant GABA receptors expressed in Xenopus laevis oocytes. Br J Pharmacol 1996;118:563–576.
    1. Tan KR, Rudolph U, Luscher C. Hooked on benzodiazepines: GABAA receptor subtypes and addiction. Trends Neurosci 2011;34:188–197.
    1. Curtis DR, Game CJ, Lodge D. Benzodiazepines and central glycine receptors. Br J Pharmacol 1976;56:307–311.
    1. Hui AC, Wong TY, Chow KM, et al. Multifocal myoclonus secondary to tranexamic acid. J Neurol Neurosurg Psychiatry 2003;74:547.
    1. Bojko B, Vuckovic D, Cudjoe E, et al. Determination of tranexamic acid concentration by solid phase microextraction and liquid chromatography‐tandem mass spectrometry: first step to in vivo analysis. J Chromatogr B Analyt Technol Biomed Life Sci 2011;879:3781–3787.
    1. Sharma V, Fan J, Jerath A, et al. Pharmacokinetics of tranexamic acid in patients undergoing cardiac surgery with use of cardiopulmonary bypass. Anaesthesia 2012;67:1242–1250.
    1. Fox MA. Tranexamic acid: how much is enough? Anesth Analg 2010;111:580–581.
    1. Merino JG, Latour LL, Tso A, et al. Blood‐brain barrier disruption after cardiac surgery. Am J Neuroradiol 2013;34:518–523.
    1. Dubber AH, McNicol GP, Douglas AS. Amino methyl cyclohexane carboxylic acid (AMCHA), a new synthetic fibrinolytic inhibitor. Br J Haematol 1965;11:237–245.
    1. Dubber AH, McNicol GP, Douglas AS, et al. Some properties of the antifibrinolytically active isomer of amino‐methylcyclohexane carboxylic acid. Lancet 1964;2:1317–1319.

Source: PubMed

3
Abonnere