ArticlePDF AvailableLiterature Review

Abstract and Figures

Blood loss and subsequent transfusions are associated with major morbidity and mortality. The use of antifibrinolytics can reduce blood loss in cardiac surgery, trauma, orthopedic surgery, liver surgery and solid organ transplantation, obstetrics and gynecology, neurosurgery and non-surgical diseases. The evidence of their efficacy has been mounting for years. Tranexamic Acid (TXA), a synthetic lysine-analogue antifibrinolytic, was first patented in 1957 and its use has been increasing in contrast to aprotinin, a serine protease inhibitor antifibrinolytic. This review aims to help acute care physicians navigate through the clinical evidence available for TXA therapy, develop appropriate dose regimens whilst minimizing harm, as well as understand its broadening scope of applications. Many questions remain unanswered regarding other clinical effects of TXA such as anti-inflammatory response to cardiopulmonary bypass, the risk of thromboembolic events, adverse neurological effects such as seizures, and its morbidity and mortality, all of which necessitate further clinical trials on its usage and safety in various clinical settings.
Content may be subject to copyright.
ONLINE FIRST
This is a provisional PDF only. Copyedited and fully formatted version will be made available soon.
ISSN: 1642-5758
e-ISSN: 1731-2515
Tranexamic acid: a clinical review
Authors: William Chuk Kit Ng, Angela Jerath, Marcin Wasowicz
DOI: 10.5603/AIT.a2015.0011
Article type: Review articles
Submitted: 2015-02-10
Accepted: 2015-03-06
Published online: 2015-03-17
This article has been peer reviewed and published immediately upon acceptance.
It is an open access article, which means that it can be downloaded, printed, and distributed freely,
provided the work is properly cited.
Articles in "Anaesthesiology Intensive Therapy" are listed in PubMed.
The final version may contain major or minor changes.
Powered by TCPDF (www.tcpdf.org)
Tranexamic acid: a clinical review
William Ng, Angela Jerath, Marcin Wasowicz
University of Toronto, UHN Department of Anesthesia, Toronto General Hospital, Canada
Abstract
Blood loss and subsequent transfusions are associated with major morbidity and mortality. The
use of antifibrinolytics can reduce blood loss in cardiac surgery, trauma, orthopedic surgery, liver
surgery and solid organ transplantation, obstetrics and gynecology, neurosurgery and non-surgical
diseases. The evidence of their efficacy has been mounting for years. Tranexamic Acid (TXA), a
synthetic lysine-analogue antifibrinolytic, was first patented in 1957 and its use has been
increasing in contrast to aprotinin, a serine protease inhibitor antifibrinolytic. This review aims to
help acute care physicians navigate through the clinical evidence available for TXA therapy,
develop appropriate dose regimens whilst minimizing harm, as well as understand its broadening
scope of applications. Many questions remain unanswered regarding other clinical effects of TXA
such as anti-inflammatory response to cardiopulmonary bypass, the risk of thromboembolic
events, adverse neurological effects such as seizures, and its morbidity and mortality, all of which
necessitate further clinical trials on its usage and safety in various clinical settings.
Key words: anesthesia, tranexamic acid, antifibrinolytic, blood conservation
Blood loss and subsequent transfusions are associated with major morbidity and mortality
[1, 2]. The use of antifibrinolytics can reduce blood loss in cardiac surgery, trauma, liver surgery
and solid organ transplantation and non-surgical diseases. The evidence of their efficacy has been
mounting for years [3, 4]. Synthetic lysine-analogue Tranexamic Acid (TXA, trans-4-
aminomethylcyclohexane-1-carboxylic acid), along with ε-aminocaproic acid (ɛ-ACA), were first
patented by S. Okamoto in 1957 [5]. Many questions remain unanswered regarding other clinical
effects of TXA such as anti-inflammatory response to cardiopulmonary bypass (CPB), the risk of
thromboembolic events and adverse neurologic effects (seizures), as well as the morbidity and
mortality of TXA, all of which necessitate further clinical trials on its usage and safety in various
clinical settings. Therefore, this review aims to help acute care physicians navigate through the
clinical evidence available for TXA treatment, develop appropriate dose regimens whilst
minimizing harm, as well as understand its broadening scope of applications [5].
1
Mechanisms of action
TXA is a synthetic lysine-analogue antifibrinolytic [6] that competitively inhibits the
activation of plasminogen to plasmin; at high concentrations it non-competitively blocks plasmin,
thus TXA inhibits the dissolution and degradation of fibrin clots by plasmin. The binding of TXA
to plasminogen is 6 to 10 times more potent that of ɛ-ACA [7]. TXA has been shown to increase
thrombus formation in a dose-dependent fashion in animal models, in contrast to aprotinin, which
inhibits thrombus formation [8].
Evidence from numerous studies show that TXA inhibits plasmin-induced platelet
activation during extracorporeal circulation, such as cardiopulmonary bypass (CPB) used in
cardiac surgery [9−12]. There are multiple factors that lead to bleeding following CPB, and
fibrinolysis is one of the few that can be mitigated by pharmacological intervention. TXA also
reduces excessive bleeding after CPB by several other mechanisms. Firstly plasmin-platelet
interaction leads to the selective release of ADP-granules from platelets, which is triggered by
platelet surface contact with the extracorporeal circuit. Soslau et al. [13] found that platelet dense-
granule ADP content in patients loaded with TXA pre-CPB were higher compared to those loaded
with TXA post-CPB, with a strong inverse relationship to blood loss. The same investigators
estimated the EC50 (half maximal effective concentration) of TXA required for the inactivation
of plasmin-induced platelet aggregation during CPB to be ≤ 15 µg mL-1 in vitro; thrombin
activation of platelets was also inhibited by plasmin-TXA binding to platelet receptors. Thus, one
may conclude that there are several pathways explaining the preservation of platelet function by
TXA during CPB.
Thirdly, TXA possibly attenuates the inflammatory response and related hemodynamic
instability in patients undergoing CPB. Hyperfibrinolysis probably plays a significant role in this
inflammatory response. In a randomized controlled trial (RCT) of 50 patients undergoing CPB,
TXA reduced significantly several of the biochemical markers of inflammatory response [14]: IL-
6, fibrin separation products, creatine-kinase (CK) and plasminogen activator inhibitor. Patients
receiving TXA had reduced incidences of inflammatory response and vasoplegic shock, fewer
mean hours of norepinephrine use (1.2 vs 25.4 h) and fewer hours of mechanical ventilation (6.5
vs 12 h) in intensive care after CPB. In a larger RCT, IL-6 had a direct relationship with
temperature, D-dimer, troponin I, CK, and lactic acid after CPB [15]. Furthermore, giving
additional post-CPB TXA significantly reduced the relative risk (RR 2.5) of inflammatory
response compared to pre-CPB TXA dosing alone.
Fourthly, hyperfibrinolysis contributes to coagulopathy in trauma and has an estimated
incidence of 15% [16]. In trauma, tissue damage causes the release of tissue plasminogen
2
activator induced by tissue ischemia and endothelial injury [17]. Point-of-care testing by
rotational thromboelastometry allows rapid identification of patients with hyperfibrinolysis in
trauma — a state associated with greater INR derangements, lower fibrinogen and higher
mortality rates [18, 19] when compared to physiologic fibrinolysis. TXA use in trauma thus has
physiologic justification, but the diagnosis of hyperfibrinolysis is crucial before initiating
treatment.
Lastly, there is a beneficial interaction of TXA with desmopressin: if the fibrinolytic
activity of desmopressin via the transient release of tissue plasminogen activators [11, 20] is
abolished by TXA preparation, desmopressin exerts salutary effect on platelet activation,
significantly reducing postoperative blood loss and transfusion [21].
Clinical usage and efficacy
The main purpose of TXA is the reduction of perioperative bleeding and transfusion
requirements in both cardiac and non-cardiac surgery. There are clear benefits both from the
mortality-morbidity and economic-cost perspectives. In a recent meta-analysis of over 100 RCTs
that compared TXA vs no TXA or a placebo in more than 10,000 patients undergoing surgery
[22], there was overwhelming evidence that TXA reduces the probability of transfusion by 38%.
Moreover, a cumulative meta-analysis suggests that this evidence has been available for more
than 10 years. Although the same study showed that fewer deaths occurred in the TXA group (RR
0.61, 95% CI 0.38 to 0.98), this became uncertain when analysis was restricted to the trials with
adequate concealment. Similarly, the Cochrane Review of the effect of antifibrinolytics on blood-
loss and transfusion of allogeneic blood [3] found that TXA significantly reduced blood
transfusion by 39%, representing an absolute risk reduction of 18%. However, TXA was not
associated with decreased mortality in all surgeries.
Cardiac surgery
Since the publication of findings by Mangano et al. [23] and Karkouti et al.
antifibrinolytic choice in cardiac surgery has shifted from aprotinin to TXA and ɛ-ACA. This was
due to the concern that aprotinin may be associated with an increased risk of cardiovascular or
cerebral vascular events, as well as renal dysfunction or failure. In a propensity-score matched
analysis (n = 10870) of patients at high risk of blood loss in cardiac surgery, Karkouti et al. [2]
reported an increased risk of renal toxicity of aprotinin when compared with TXA with a
potential increase in mortality, whilst both antifibrinolytics had similar hemostatic effectiveness.
Subsequently, in a 5-year follow-up of (n = 4374) patients having CABG surgery [24], Mangano
3
et al. found aprotinin to be associated with increased mortality compared with a control, TXA and
ɛ-ACA.
The use of TXA was further propelled by the BART [25] trial (Blood Conservation Using
Antifibrinolytics in a Randomized Trial) published by Fergusson et al. [25], who compared the
use of aprotinin, TXA and ɛ-ACA in high-risk cardiac surgery patients. Alarmingly, the 30-day
mortality rate, was 6% for the aprotinin group vs 3.9% for TXA (RR 1.55) and 4.0% for ɛ-ACA
(RR 1.52); however, there was a modest reduction in the risk of massive bleeding in the aprotinin
group compared with the two lysine-analogues. This led to the withdrawal of FDA and Health
Canada approvals for aprotinin [26]. On the other hand, critics of BART have noted that in the
high-risk patient subset, aprotinin may possibly have a better benefit/risk profile [27]. Indeed, this
is supported by the propensity-score (n = 1544) matched study by Karkouti et al. [28]
Knowledge of the efficacy of TXA vs a control in reducing blood loss and transfusion in
cardiac surgery has been available for decades [29, 30]. Recent meta-analyses confirm this
notion. A report by Henry et al. [31] stated a relative risk of blood transfusion with TXA of 0.68
with ~300 mL of blood saved while Ngaage et al. [32] reported an odds ratio of 0.53 with ~298
mL of blood saved. Although a comparison of TXA with a placebo showed a reduction in the
number of reoperations caused by blood loss [3, 32], once again the benefit of reduced
reoperation numbers due to blood loss is even more convincing with high-dose aprotinin [3, 31].
Even though TXA is approximately 7-times more potent than ɛ-ACA, they were comparable in
relative risk and actual volume of blood loss in cardiac surgery [3]. Moreover, TXA did not show
a decreased risk of mortality in cardiac surgery in the aforementioned meta-analyses.
While the BART protocol TXA regimen — 30 mg kg-1 loading dose followed by 16 mg
kg-1 h-1 infusion during surgery with an added 2 mg kg-1 in the circuit is in the high-dose range
[33, 34], it has been nevertheless widely adopted by many cardiac surgical centers since the
publication of BART. Early dose-response studies in cardiac surgery by Horrow et al. [35] found
that a prophylactic loading dose of 10 mg kg-1 with infusion at 1 mg kg-1 h-1 was optimal, when
compared to six incremental loading doses from 2.5 to 40 mg kg-1 followed by 0.25 mg to 4 mg
kg-1 h-1 infusion. Later, several authors found inconsistent plasma concentrations [36, 37] with the
Horrow regimen when the pharmacokinetic influence of the circuit and patient renal function
were included in analysis. A recent RCT pitted the Horrow regimen against the higher BART
regimen in cardiac surgery patients [38], and it found that although a high dose of TXA does not
reduce the incidence of blood product transfusion up to day 7 (63% low dose vs 60% high dose),
it is more effective than a low dose of TXA in decreasing transfusion (2.5 vs 4.1 U), blood loss
(590 vs 820 mL), and repeat surgery (2.5% vs 6%). A subgroup analysis of high-risk patients with
4
dual antiplatelet therapy or having complex surgery showed a further reduced incidence of
transfusion and suggests that a high dose of TXA may be better in that group.
Many patients undergoing cardiac surgery receive aspirin and/or clopidogrel
preoperatively. There is evidence that TXA partially corrects arachidonic acid-induced (aspirin)
and ADP-induced (clopidogrel) platelet aggregation defects [39] detected by multiple electrode
aggregometry, in patients on dual-antiplatelet therapy, concurring with the plasmin-induced
platelet inhibition [10] by the redistribution and degradation of glycoprotein Ib and IIb/IIIa
receptors.
Ppediatric cardiac and non-cardiac surgery
The efficacy of TXA in major pediatric surgery reproduces what was found in the adult
population. In a meta-analysis of over 2,000 pediatric patients undergoing cardiac or scoliosis
surgery the authors found no evidence that TXA was inferior in the reduction of blood loss vs
aprotinin at 24 h. Indeed, TXA reduced blood loss by 11 mL kg-1 (95% CI 9 to 13 mL kg-1), and
reduced packed red cell transfusion by 4 mL kg-1 (95% CI 2 to 7 mL kg-1). In scoliosis surgery,
TXA significantly reduced blood loss by 682 mL (95% CI 214 to 1149 mL). A recent RCT
comparing TXA with a control (n = 150) in pediatric patients undergoing cardiac surgery [41]
demonstrated a reduction of blood loss but not the units of blood transfused at 24 h. Similarly, a
retrospective study (n = 231) of pediatric patients having cardiac surgery [42] found that TXA
significantly reduced blood loss and reduced the amount of blood transfused intraoperatively, as
well as at 48 h. Moreover, the authors found a reduction in the number of patients requiring blood
transfusion (45/103 vs 77/127, P = 0.012) at 48 hours. Interestingly, both studies did not find
differences between cyanotic and acyanotic subgroups. However, according to Faraoni et al. [43]
in their meta-analysis of pediatric cardiac surgery and TXA, there is much heterogeneity in the
data from RCTs: transfusion policies were ill-defined, with variability in regimens and data on
TXA effect on morbidity and mortality. However, these authors found that in the 848 patients
included in the analysis, the amount of red cells, platelet and fresh frozen plasma transfused
showed decreasing trends between TXA and the control. Despite recent pharmacokinetic studies
on pediatric populations [44, 45] the ideal dose regimen of TXA in pediatric cardiac surgery is
still unknown [46]. In vitro studies in neonates have shown a significantly lower plasma
concentration (~6.5 µg mL-1 vs ~17 µg mL-1) required to prevent hyperfibrinolysis when
compared to adults [45]; this would set the basis of future clinical trials on dosing regimens and
risk-benefit balance.
Faraoni and Goobie also performed a systematic review [47] on the use of
antifibrinolytics in pediatric non-cardiac surgery and concluded that in pediatric spine surgery
5
(mainly scoliosis correction) and craniosynostosis surgery, TXA did decrease blood loss and
transfusion requirements. An older Cochrane meta-analysis [48] found antifibrinolytics as a class
in scoliosis surgery reduced blood loss by 426 mL and the amount of blood transfused by 327
mL; no subgroup analysis on the effect of TXA alone was carried out. There are few RCTs
[49−51] on TXA use in pediatric scoliosis surgery. Thus, the safety profile of TXA use in
pediatric spinal surgery remains unresolved.
Basta et al. [52] conducted a separate systematic review on major pediatric surgery
(cardiac, spinal and craniofacial) and found that antifibrinolytics reduced blood loss and
transfusion volumes, particularly in craniofacial surgery. Craniosynostosis is not an uncommon
pediatric disease requiring early surgical intervention and is associated with considerable blood
loss [53]. There are two RCTs of TXA vs control in craniosynostosis surgery [54, 55]: Goobie et
al. showed significant decrease in blood loss of 54 mL kg-1 and decreased volume of blood
transfused by 23 mL kg-1; Dadure et al. [55] found a decrease in transfusion requirement by 85%
(11 to 1.6 mL kg-1) intraoperatively and by 57% (16.6 to 7.2 mL kg-1) postoperatively. Moreover,
Goobie et al. [44] describes a dose regimen for craniosynostosis using a two-compartment model,
suggesting a 10 mg kg-1 loading TXA followed by a 5 mg kg-1 h-1 infusion to produce a threshold
plasma concentration of 16 µg mL-1.
Orthopedic surgery
The reduction of blood loss in orthopedic surgery is of great importance, especially in hip
or knee arthoplasty and spinal surgery. Pharmacological treatment with TXA is making a
resurgence in orthopedic surgery. Indeed, antifibrinolytic use in orthopedic surgery is supported
by a meta-analysis by Kagoma et al. [56], which found a reduction in blood loss, relative risk of
transfusion (RR 0.52) and no increased risk of thromboembolism; the dose of TXA administered
ranged between 10−15 mg kg-1. A large retrospective analysis by Poeran et al. [57] studied the
perioperative use of TXA in knee or hip arthroplasty (n = 872416). Patients who received TXA
had lower rates of blood transfusion (7.7 vs 20.1%), fewer thromboembolic events (0.6 vs 0.8%),
and reduced incidence of acute renal failure (1.6 vs 1.8%) as well as combined complications (1.9
vs 2.6%). With an increasing dose of TXA (none, < 1 g, ~ 2 g and > 3 g), there were decreasing
odds (OR 0.31 to 0.38) of blood transfusion, and no significant increased risk of complications.
Moreover, its efficacy and safety profile in orthopedic surgery is further supported by two
meta-analyses of TXA use in primary hip [58] and knee [59] arthoplasty. In hip surgery, Sukeik et
al. [58] found that TXA reduced intraoperative blood loss by 104 mL and postoperative blood
loss by 172 mL (n = 350). There was also a proportional reduction of patients requiring blood
transfusion (risk difference −0.20). In total knee replacement, Alshryda et al. [59] found
6
significant reduction in blood loss by 591 mL (n = 763). It should be noted that there was
significant heterogeneity in the trials. Subgroup analysis of high dose TXA (> 4 g) showed a
reduction in transfusion requirements with homogeneity. In both meta-analyses there was no
evidence of increased risk of thromboembolic events due to TXA. In addition, the use of the
tourniquet in knee arthroplasty can activate local fibrinolysis apart from standard tissue trauma
[60] and adds justification for TXA use. A meta-analysis of intravenous TXA use (n = 581) in
spinal surgery by Yang et al. [61] had comparable findings to pediatric scoliosis surgery: there
was reduction in postoperative blood loss by 389.21 mL and the amount of blood transfused by
134.55 mL with TXA. A RCT of intravenous TXA use in cervical laminoplasty also found a
decrease in blood loss (264 mL) but not intraoperative blood loss [62]; again there was no
increase in complications.
Topical use
The topical use of TXA has been examined in a Cochrane review [63] by Ker et al.
Although the authors found reliable evidence that topical TXA reduces bleeding and blood
transfusion in surgical patients, the risk of thromboembolism is unclear, as many studies do not
report this complication or are underpowered. Topical administration results in a ten-fold less
plasma concentration of TXA when compared to intravenous administration [8, 64], with a
potential reduction in adverse effects. Although topical TXA has been studied in cardiothoracic
[65−68], orthopedic [64, 69−72], otorhinolaryngologic [73, 74] and orthognathic [75−77]
surgeries, high-quality trials are lacking.
Topical TXA in knee arthoplasty has been reviewed in a meta-analysis [78] while Panteli
et al. showed that topical TXA reduced postoperative drain output (−268 mL), total blood loss
(−220 mL), hemoglobin drop (−0.94 g dL-1) and transfusion risk (RR 0.47, 95% CI 0.26 to 0.84);
there was no increase in thromboembolisms. The authors examined the subgroup using > 2 g of
topical TXA and found that these patients had a significantly less transfusion requirement (RR
0.41, P = 0.05). Similarly in a separate review of RCTs [72], Zhang et al. [72] also found the
intra-articular injection of TXA in knee arthroplasty found a reduction of blood loss (396 mL),
relative transfusion risk (RR 0.22), drainage output and hemoglobin drop; there was no increased
risk of thromboembolism. Once again there was significant heterogeneity in these trials. Two
RCTs [69, 70] by Alshryda et al. found that intra-articular injection of TXA in primary total hip (n
= 161) and knee (n = 157) arthroplasty reduced the absolute risk of blood transfusion by 19.6%
and 15.4%, respectively, and reduced blood loss, hemoglobin drop, as well as decreased the cost
per episode by £ 305 and £ 333 respectively. Moreover, there was decreased length of stay in
7
knee surgery by 1.2 days without increased in thromboembolic events. All these findings
cumulatively support the topical use of TXA in orthopedic surgery.
In a meta-analysis of topical antifibrinolytic use in cardiac surgery (n = 622), Abrishami et
al. [79] found reduced postoperative blood loss and transfusion requirements in patients
undergoing on-pump cardiac surgery. Mahaffey et al. [80] (n = 160) found that combined
intravenous and topical TXA was associated with decreased chest drain output at 3, 6 and 12 h
postoperatively. Even though the total amount of TXA was higher in the combined group, less
TXA (4.1 g vs 5.1 g) was given intravenously compared with the control. In addition, there was
no increase in adverse events. Spegar et al. [81] studied the augmentation of systemic TXA by
topical application (2.5 g in 250 mL saline into pericardial cavity) in valvular surgery (n = 100)
and found intergroup variance on blood loss and fresh frozen plasma but a non-significant
decrease in the volume of blood loss in the augmented group. In contrast, Fawzy et al. [66] in
their RCT (n = 38) found a decrease in postoperative blood loss (−626 mL vs −1040 mL) and
platelet transfusion (median units 0 vs 2) using 1 g TXA in 100 mL saline into the pericardial
cavity. Similar regimens used in two other RCTs found that topical TXA did reduce postoperative
blood loss in cardiac surgery without increased risk of adverse events [67, 68].
Trauma
TXA application in trauma is supported by firm clinical evidence. The most convincing
multicenter RCT in trauma [82] to date is the comparison of TXA vs placebo in over 20,000
patients by the CRASH-2 collaborators. Patients were assigned to a placebo or IV loading of 1 g
TXA within 8 h of trauma then followed by IV infusion of 1 g TXA over 8 h. It showed that all-
cause mortality was reduced in the TXA group (RR 0.91), and death due to bleeding was
significantly reduced (RR 0.85). Subsequent analysis showed that early treatment (≤ 1 h from
injury) reduced the risk of death from bleeding (RR 0.68), while treatment given after 3h of
injury seemed to increase the risk of death due to bleeding [83].
It is important to remember that physiologic fibrinolysis and even fibrinolytic shutdown
occurs in trauma, and not just hyperfibrinolysis. In a recent study (n = 180) of patients with an
Injury Severity Score of ≥ 15, there was a sizeable portion (64%) of patients with fibrinolysis
shutdown per thromboelastometry at 30 min [84]. The distribution of mortality was U-shaped
relative to the fibrinolysis system, the physiologic group had lowest mortality (5%), and the
hyperfibrinolysis (44%) and shutdown (26%) groups had higher mortality. This supports the
employment of careful patient selection when using exogenous inhibition of fibrinolytic system.
The use of thromboelastometry will help prevent indiscriminate TXA therapy.
8
Neurosurgery
The use of antifibrinolytics in intracranial hemorrhage (ICH) and particularly aneurysmal
subarachnoid hemorrhage (SAH) has been investigated for decades, with earlier findings of
decreased re-bleeding rates but increased risk of stroke. New strategies were introduced for the
prevention of cerebral vasospasm with shorter antifibrinolytic intervention periods [85]. There is
renewed interest in TXA and ɛ-ACA for these patients. The earlier position is outlined in the
review of antifibrinolytics vs a control in ICH by the Cochrane Stroke Group [86]: the treatment
did not benefit patient outcome and death was not reduced. Treatment did reduce risk of re-
bleeding (OR 0.55) with some heterogeneity across the trials. However, there was an increased
risk of ischemic stroke (OR 1.39), again with heterogeneity in the trials. In drawing conclusions
from these results, the authors did not support the routine use of antifibrinolytics in aneurysmal
SAH. In contrast, a study [87] by Roos et al. [87] did not show rates of increased ischemic stroke,
namely delayed cerebral ischemia from vasospasm, probably because patients in this trial were
given calcium channel antagonist nimodipine and hypervolemic therapy concurrently.
Since this meta-analysis, new strategies using antifibrinolytics in short duration have
shown promise of reduced re-bleeding with fewer adverse events [88−91]. Although the
mechanism for re-bleeding is multifactorial, increased fibrinolysis and decreased platelet-plug
stability are implicated [92]. The risk of rebleeding is highest in the first 6 h after aneurysmal
SAH, with a poor prognosis as assessed by a reduction of the Glasgow Outcome Scale from 40%
to 80% and a mortality rate of 20% to 60% [92]. Hillman et al. [91] in their RCT (n = 505)
compared early intravenous TXA with a control in patients with SAH for short duration (up to 72
h) and found a significant reduction of early re-bleeding from 10.8% to 2.4% and an 80%
reduction in the mortality rate in the early rebleeders. Using evidence from transcranial Doppler
and clinical measurements, recent literature shows a resurgence of TXA as a part of protocol
therapy alongside other preventative strategies for vasospasm in the acute phase of aneurysm
SAH, prior to aneurysm closure [88, 93].
As traumatic ICH includes epidural, subdural and subarachnoid hemorrhage, the use of
TXA has been gaining increasing interest since CRASH-2 [82]. A nested RCT within CRASH-2
by Perel et al. [94] reviewed the rate of ICH growth in 270 patients, and they found no moderate
benefits (total hemorrhage growth and/or new ischemic lesions) nor harmful effects with certainty
in traumatic brain injury. More recently, Sprigg et al. [95] performed a pilot RCT comparing TXA
vs a control in spontaneous intracerebral hemorrhage — the first trial studying this application —
and found it feasible to use intravenous TXA early with good tolerability. As a result, two large
multicenter trials — namely TICH-2 (International) [95] and STOP-2 (Australia) [97] — are in
9
progress to evaluate the efficacy and safety of TXA in the setting of spontaneous intracerebral
hemorrhage.
Hepatic surgery
Orthotopic liver transplantation (OLT) is associated with significant blood loss and the
need for transfusion of blood products, with fibrinolysis being a major player in this [98, 99].
There has been clinical evidence for the use of antifibrinolytics in OLT for over three decades;
previously aprotinin was commonly used in OLT and trials demonstrated potential advantages
(antioxidant, anti-inflammatory) of aprotinin over TXA [100]. There is one meta-analysis which
has studied the use of antifibrinolytics including TXA and aprotinin in OLT (n = 1407), while
Molenaar et al. [101] found that both drugs reduced intraoperative blood and fresh frozen plasma
requirements.
There are several studies which have compared the efficacy of TXA with aprotinin in
OLT. In one RCT (n = 127), prophylactic TXA had similar efficacy to aprotinin [102] in terms of
blood and component transfusions both intraoperatively and at 24 h. There were neither
differences in mortality and complications nor in coagulation laboratory data collected
intraoperatively, except aPTT. Similar results were found by Ickx et al. [103] (n = 51), with the
additional finding of inhibition of fibrinolysis by both TXA and aprotinin vs a control. Gurusamy
et al. [104] addressed different strategies of decreasing blood loss in OLT in their Cochrane
review. Although with respect to aprotinin and TXA, they concluded that the clinical trials had
been biased, there were no differences in 60-day mortality rates, re-transplantation risk or
thromboembolic events in the TXA group vs the control and no difference between aprotinin and
TXA in mortality or thromboembolism risk. Massicotte et al. [105] analyzed 400 patients
undergoing OLT who had received antifibrinolytics and found no difference between TXA and
aprotinin in blood loss (1082 vs 1007 mL), blood transfused per patient (0.5 vs 0.5 U), final
hemoglobin (93 vs 95 g L-1), percentage of transfusion-free cases (80 vs 82%) or the 1-year
survival rate (85.1 vs 87.4%). Interestingly, preoperative hemoglobin correlated with 1-year
survival and transfusion requirements.
Görlinger [106] analyzed ROTEM® (rotational thromboelastometry) in 642 OLTs and
suggests using prophylactic administration only in fulminant liver failure or reduced maximal clot
firmness, which indicates a high-risk for hyperfibrinolysis. Although 60% of patients displayed
hyperfibrinolysis during OLT, only 40% who showed early hyperfibrinolysis during prehepatic
and anhepatic phases, required antifibrinolytics. In the neohepatic phase, only patients with
increased fibrinolysis and clinical bleeding were treated. This selection by point-of-care testing is
aimed at reducing iatrogenic prothrombotic risk.
10
TXA use has also been investigated for blood conservation in hepatectomy performed for
tumor resection. These studies showed promising efficacy, according to a RCT by Wu et al. [107]
(n = 214). Although a Cochrane review addressed use of pharmacological intervention for blood
conservation in liver resection, and found that aprotinin and TXA significantly reduced the risk of
allogeneic blood transfusion compared with a control [108], this review included a few small
RCTs. A survey of Canadian hepatobiliary surgeons showed that even though low central venous
pressure strategy was used commonly during liver resection, other conservation strategies
including TXA were rarely employed [109]. High quality RCTs on perioperative morbidity and
mortality are needed to assess pharmacological intervention for blood conservation in
hepatectomy.
Obstetric and gynecology
As Menorrhagia is a common illness affecting women’s health and quality of life, TXA
has been used as a form of treatment for over four decades. A recent review [110] placed its
efficacy in the reduction of menstrual blood loss by 34 to 59%. An earlier Cochrane review [111]
on the use of antifibrinolytics (TXA and its precursor) in heavy menstrual bleeding found that
TXA vs a placebo significantly reduced mean blood loss (mean difference −94 mL). TXA also
significantly reduced blood loss when compared to mefenamic acid, norethisterone and
etamsylate; there was no difference in adverse effects between TXA and the other agents. A
recent RCT also found reductions in menstrual blood loss by a new oral formulation of TXA that
was both statistically significant (> 50 mL) and clinically meaningful to patients, at doses > 3.9 g
day-1 for up to 5 days of the cycle [112]. This reinforced the findings of a greater quality of life
with TXA use in women with menorrhagia in an early uncontrolled study [113] (n = 849), who
were assessed by a questionnaire based on one designed by Edlund et al. [114].
A Cochrane review [115] analyzed two RCTs (n = 453) comparing TXA vs a control in
women having a caesarean section or vaginal delivery [116, 117], and the authors found that
blood loss of > 400 mL was less common and mean blood loss was lower in the TXA group vs
the control (mean difference −75 mL). A recent meta-analysis on TXA in pregnancy and
postpartum [118] by Peisidis et al. [118] included several quasi-blinded trials [119−121] excluded
from the above Cochrane review, and found a combined estimated decrease of blood loss by 32.5
mL in TXA pre-caesarean section vs a control. A similar effect of TXA was found in a separate
meta-analysis by Ferrer et al. [122] in a reduction of postpartum blood loss by 92 mL compared
with the control.
Ducloy-Bouthers et al. [123] (n = 144) demonstrated that a high-dose TXA (4 g infusion
over 1 h followed by 1 g h-1 for 6 h) vs a control — in women with greater than 800 mL
11
postpartum hemorrhage — was effective in reducing blood loss (173 vs 221 mL). Moreover, the
TXA group had a shorter duration of hemorrhage, less progression to severe postpartum
hemorrhage and less incidence of transfusion. This trial was underpowered to detect rare adverse
effects. A RCT (n = 439) by Gungorduk et al. [124] showed that TXA — 1 g given intravenously
over 5 min at the delivery of the anterior shoulder — reduced blood loss during the 3rd and 4th
stage of labor compared to the control (261.5 vs 349.98 mL), significantly higher hematocrit and
hemoglobin levels on day one, with no major complications at a three week follow-up.
Gungorduk et al. [125] also performed a RCT (n = 660) of pre-caesarean intravenous TXA vs
control, and found a significantly lower mean blood loss, a lower proportion of women with
severe postpartum hemorrhage (> 1000 mL blood loss) and a lower risk of additional uterotonics
used. Finally, three new RCTs [126−128] comparing pre-caesarean section intravenous TXA vs a
control have shown a reduction of intraoperative and post-caesarean blood loss without increased
adverse events such as thromboembolism. Determining the different obstetric and gynecological
settings in which TXA may be beneficial remains a critical question for future research.
Other usages
A common minor surgery where post-operative bleeding remains a big issue is
tonsillectomy. A meta-analysis of the use of TXA in tonsillectomy [129] (n = 180), showed a
reduced volume of blood loss but not the number of patients with post-tonsillectomy hemorrhage.
Albirmawy et al. [73] found that post-resection topical TXA in pediatric adenoidectomy led to a
reduction of blood loss during surgery, decreased postoperative bleeding and transfusion.
Lastly, several old trials support the use of TXA in hereditary angioneurotic edema [130,
131]. The biological mechanism involves the inhibition of the complement system by TXA in the
presence C1 esterase deficiency and partial normalization of plasma kinin activation [130]. In
Japan, TXA is approved for conditions such as urticarial swelling, itch, eczema, drug eruptions or
toxicoderma [8], in which local hyperfibrinolysis and inflammation are involved [131].
The use of TXA in upper gastrointestinal bleeding was reviewed in a Cochrane meta-
analysis, and although TXA use vs a control reduced mortality risk, the effect was lost in a
subgroup analysis stratified for bias control and in an sequential analysis [132]. No such benefit
was demonstrated in TXA vs other anti-ulcer therapy. Although five serious cases of
thromboembolic events occurred in the TXA group, this was not statistically significant. No
RCTs were found assessing TXA use in upper gastrointestinal bleeding in liver disease [133]. The
use of TXA in hemoptysis from any cause was reviewed by the Cochrane Collaboration [134],
and TXA vs a control significantly reduced bleeding time (mean difference −19.47 h), without
any difference in side-effects.
12
Adverse effects and dosing
Topical administration of TXA to the central nervous system of animals has been shown
to cause seizures in a dose-related fashion [135, 136], this correlates with human reports of
seizures induced by accidental intrathecal injections of TXA [136−138]. Recently a dose-
response relationship of TXA has been proposed as a modifiable risk factor for seizures for
patients undergoing cardiac surgery [139]. TXA crosses the blood-brain barrier and penetrates the
eye, and produces cerebrospinal fluid concentration levels around 10% of the plasma
concentration [8]. Likewise, it diffuses into and out of synovial membranes and joint fluid. It is
now clear from current literature that moderate to high doses of TXA in cardiac surgery are
associated with an increased risk of seizures [139, 140]. In a multivariate analysis of over 11,000
patients after cardiac surgery, Sharma et al. [142] found TXA to be a strong independent predictor
for the development of postoperative generalized seizures [141] (OR 14.3); in addition, patients
with seizures had a 2.5 times higher mortality rate. Similarly, in a propensity-score adjusted
analysis (n = 4883) Koster et al. [143] showed that moderate dosing of TXA in cardiac surgery
doubled the rate of post-CPB seizure and in-hospital mortality. Similar concerns over the
increasing trend of post-CPB seizure in pediatric patients have led to the substitution of TXA by
ɛ-ACA in a major European center for pediatric cardiac surgery.
The postulated mechanism was TXA binding to GABAA receptors, subsequently blocking
GABAA-mediated inhibition in the CNS [144, 145]. Recently, Lecker et al. [145] have
demonstrated that TXA is structurally similar to glycine, and competitively inhibits glycine
inhibitory receptors in the cortical and spinal cord neurons in rats; TXA also inhibited the
GABAA receptors in cortical and spinal cord neurons. Both TXA dis-inhibitory pathways cause
increased excitatory synaptic drive evidenced by seizure-like events in cortical slices induced at
TXA concentrations of 31 μg mL-1 (200 μM), similar to that measured in CSF of patients
undergoing CPB. Finally, peak CSF TXA concentrations occurred when the infusions were
stopped after CPB, later than peak serum levels. When taken together, this explains the late-onset
of unexpected seizures in patients emerging from anesthesia after CPB. Moreover, ɛ-ACA had
10-fold less potency in glycine receptor inhibition, and aprotinin had no inhibitory potency.
There are various dose regimens for different indications cited by clinical trials; initially
an effective plasma concentration of TXA for antifibrinolysis was reported to be 5−10 μg mL-1
[147] or 10−16 μg mL-1 [13, 147, 148]. Subsequently, Dowd et al. [37] proposed the dosing
scheme later adopted in the BART study, in order to ensure plasma levels which would achieve
complete inhibition of fibrinolysis for cardiac patients undergoing CPB, namely a loading dose
30 mg kg-1, maintenance infusion at 16 mg kg-1 h-1with an additional 2 mg kg-1 in the circuit [25,
13
37]. Sharma et al. [150] conducted a pharmacokinetic study of the BART regimen in cardiac
surgery and demonstrated that plasma TXA concentrations were consistently higher than
suggested levels aiming to achieve 100% (>100 μg/mL) and 80% inhibition (> 10 μg mL-1) of
tissue plasminogen activator and these levels remained high for up to 6 hours postoperatively.
Approximately 95% of TXA is excreted via the urine unchanged, and excretion decreases with
increasing plasma creatinine levels. The dosage adjustment for renal-impaired patients remains an
unknown; Jerath et al. [151]used the 2 compartment model to guide a simulated reduction of the
maintenance infusion rate according to the GFR of patients during CPB to achieve > 100 μg mL-1
threshold plasma concentrations.
Despite the fact that TXA is minimally metabolized in the body, precautions should be
taken with prothrombotic medications [152] used concomitantly. According to the Cyklokapron®
product information [6], these medications include a combination hormone contraceptives, factor
IX, Xa and VIIa complex concentrates, anti-inhibitor coagulant concentrates, thrombin,
batroxobin or hemocoagulase.
There is currently no clinical evidence that the use of TXA increases the risk of
thromboembolic events, namely myocardial infarction, stroke, deep vein thrombosis or
pulmonary embolism according to meta-analyses and clinical trials cited in the general [22, 63],
trauma [82], orthopedic [48, 56, 58, 59, 78], cardiac [25, 32, 153] or obstetric & gynecological
[111, 112, 119, 123] settings. However, there are reports of catastrophic intracardiac or
intrapulmonary thromboses [154], whose putative cause was antifibrinolytic use, albeit none
involved TXA. The meta-analysis of antifibrinolytics in OLT (n = 1407) by Molenaar et al. [101]
found no increased risk of hepatic artery thromboses or thromboembolism. Ngaage et al. [32] in
their meta-analysis of TXA in cardiac surgery noted that thromboembolic events (myocardial
infarction and neurologic complication) and mortality observed were few but not increased
compared to non-treatment groups; the authors still warn against the indiscriminate use of TXA.
In patients with menorrhagia, TXA was not associated with an increased thromboembolism risk
in a nested case-control study [155] (n = 686), whereas other therapy groups had a significantly
increased risk, suggesting that menorrhagia is prothrombotic. Finally, Perel et al. [63] were
uncertain about the increased risk of thromboembolism and stroke in a Cochrane review of TXA
in emergency surgery; only three trials met the criteria for inclusion and the number of events
observed were small.
Overall, the cumulative evidence shows that TXA is a well-tolerated drug when delivered
orally, intravenously and/or topically. Gastrointestinal disturbance, allergic skin reaction, visual
disturbance occur more commonly [8] and seizures less commonly at high concentrations.
14
Unanswered questions
There are still important questions of mortality and morbidity of TXA use in surgery. One
may expect that a reduction of transfusion would translate into reduction in mortality and
morbidity. As mentioned above, Ker et al. [22] found fewer deaths (RR 0.61) occurred in the
TXA group in their meta-analysis, albeit with uncertainty of adequate concealment. The overall
potential for increased thromboembolism risk with TXA remains uncertain. The safety profile and
dose regimens of TXA in cardiac and non-cardiac surgeries in the pediatric population requires
further investigation, as previous studies are underpowered to detect differences in adverse effects
[47].
The adjustment of TXA in renal impairment warrants further investigation, especially
given the at-risk patient group undergoing cardiac surgery. There is no universally accepted dose
regimen despite definite concerns of seizure risk of high-dose TXA.
Given the link between inflammatory response and coagulation-fibrinolysis systems and
the likely attenuation of inflammatory response by TXA in CPB [14, 15], clinicians ought to
identify at-risk patients who may benefit from its treatment.
There are several case reports of young, healthy individuals developing ischemic cerebral
events after TXA use, especially those with heterozygous MTFR C677T genes (methylene
tetrafolate transferase) [156]. In the meta-analyses and large clinical trials, the risk of stroke,
pulmonary thromboembolism and deep venous thrombosis with TXA use remains uncertain. The
interaction of pharmacology with genetic factors is an exciting field for research.
Several ongoing multicenter RCTs on TXA are worth following. The STOP-AUST trial
[98] is comparing early (≤ 4.5 h of stroke onset) intravenous TXA use with a placebo in patients
with confirmed intracerebral hemorrhage by CT angiography contrast extravasation — a
biomarker of likely hematoma growth. The hypothesis is that TXA will reduce intracerebral
hematoma growth at 24 h. CRASH-3 is an international pragmatic trial quantifying the effect of
early TXA (the same regimen as CRASH-2) on mortality and morbidity in 10,000 patients with
traumatic brain injury [157]. Lastly, Shakur et al. [158] is leading an international pragmatic trial,
a.k.a. the WOMAN trial, on the use of TXA in 15,000 women with a clinical diagnosis of
postpartum hemorrhage, with the authors hypothesizing a reduction of mortality and/or
hysterectomy. This trial has a large third world representation with an obvious contextual
relevance. In view of the thrombotic and bleeding complications of cardiac surgery [5], Myles et
al. is leading a multicenter RCT (ATACUS, n = 4600) investigating aspirin and TXA in CABG
surgery [159]. It is a 2 × 2 factorial trial assessing whether aspirin, TXA, or both can reduce
15
mortality and/or morbidity after elective CABG. Ischemic (renal, cerebral, bowel) complication is
a secondary endpoint. This will yield important data on TXA in cardiac surgery.
Summary
TXA as an antifibrinolytic treatment applied in a perioperative setting has strong
pharmacological and clinical grounds. Although there are other situations in which the use of
TXA is desirable, these require definitive trials on morbidity and mortality. TXA administration
should be based on clinical judgment, guided by patient history, thromboelastometry, laboratory
and radiologic investigation, and tailored to the treatment location and capacity for intervention
and transfusion. Future reviews should include guidelines on TXA dose regimens minimizing
seizure risk, and conclusions regarding the thromboembolic risk. The ongoing research outlined
will help answer these questions.
Ackowledgements
1. The authors declare no financial disclosure.
2. 2. The authors declare no conflict of interest.
References
References
1. Karkouti K, Wijeysundera DN, Yau TM et al.: The independent association of massive
blood loss with mortality in cardiac surgery. Transfusion 2004; 44: 1453–1462.
2. Karkouti K, Beattie WS, Dattilo KM et al.: A propensity score case-control comparison of
aprotinin and tranexamic acid in high-transfusion-risk cardiac surgery. Transfusion 2006; 46:
327–338.
3. Henry DA, Carless PA, Moxey AJ et al.: Anti-fibrinolytic use for minimising perioperative
allogeneic blood Transfusion Cochrane database Syst Rev. 2011; 3: CD001886.
4. Ortmann E, Besser MW, Klein AA: Antifibrinolytic agents in current anaesthetic practice.
Br J Anaesth. 2013; 111: 549–63.
5. Ker K, Roberts I: Tranexamic acid for surgical bleeding. BMJ 2014; 349: g4934.
6. Pfizer Canada Inc. Cyklokapron® - Product monograph [Internet]. Kirkland: Pfizer
Canada Inc; 2013 [cited 2014 Dec 11]. p. 1–22. Available from: http:
//www.pfizer.ca/en/our_products/products/monograph/195
7. Dunn CJ, Goa KL: Tranexamic acid. Drugs 1999; 57: 1005–1032.
8. McCormack PL: Tranexamic acid: a review of its use in the treatment of
hyperfibrinolysis. Drugs 2012; 72: 585–617.
16
9. Harker LA, Malpass TW, Branson HE, Hessel II EA, Slichter SJ: Mechanism of abnormal
bleeding in patients undergoing cardiopulmonary bypass: acquired transient platelet dysfunction
associated with selective alpha-granule release. Blood 1980; 56: 824–834.
10. De Haan J, Van Oeveren W: Platelets and soluble fibrin promote plasminogen activation
causing downregulation of platelet glycoprotein Ib/IX complexes: protection by aprotinin.
Thromb Res 1998; 92: 171–179.
11. Kucuk O, Kwaan HC, Frederickson J, Wade L, Green D: Increased fibrinolytic activity in
patients undergoing cardiopulmonary bypass operation. Am J Hematol 1986; 23: 223–229.
12. Khuri SF, Valeri CR, Loscalzo J et al.: Heparin causes platelet dysfunction and induces
fibrinolysis before cardiopulmonary bypass. Ann Thorac Surg 1995; 60: 1008–1014.
13. Soslau G, Horrow J, Brodsky I: Effect of tranexamic acid on platelet ADP during
extracorporeal circulation. Am J Hematol 1991; 38: 113–119.
14. Jimenez JJ, Iribarren JL, Lorente L et al.: Tranexamic acid attenuates inflammatory
response in cardiopulmonary bypass surgery through blockade of fibrinolysis: a case control
study followed by a randomized double-blind controlled trial. Crit Care 2007; 11: R117.
15. Jiménez 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 BioMed Central Ltd 2011; 6: 138.
16. Schöchl H, Frietsch T, Pavelka M, Jámbor C: Hyperfibrinolysis after major trauma:
differential diagnosis of lysis patterns and prognostic value of thrombelastometry. J Trauma 2009;
67: 125–131.
17. Hess JR, Brohi K, Dutton RP et al.: The coagulopathy of trauma: a review of mechanisms.
J Trauma 2008; 65: 748–754.
18. Levrat a, Gros A, Rugeri L et al.: Evaluation of rotation thrombelastography for the
diagnosis of hyperfibrinolysis in trauma patients. Br J Anaesth 2008; 100: 792–797.
19. Kutcher ME, Cripps MW, McCreery RC et al.: Criteria for empiric treatment of
hyperfibrinolysis after trauma. J Trauma Acute Care Surg 2012; 73: 87–93.
20. Melissari E, Scully MF, Paes T, Kakkar VV: The influence of LMW heparin on the
coagulation and fibrinolytic response to surgery. Thromb Res 1985; 37: 115–126.
21. Özal EE, Kuralay E, Bingöl H, Cingöz F, Ceylan S, Tatar H: Does tranexamic acid reduce
desmopressin-induced hyperfibrinolysis? J Thorac Cardiovasc Surg 2002; 123: 539–543.
22. Ker K, Edwards P, Perel P, Shakur H, Roberts I: Effect of tranexamic acid on surgical
bleeding: systematic review and cumulative meta-analysis. BMJ 2012; 344: e3054.
17
23. Mangano DT, Tudor IC, Dietzel C: The risk associated with aprotinin in cardiac surgery.
N Engl J Med 2006; 354: 353–365.
24. Mangano DT, Miao Y, Vuylsteke A et al.: Mortality associated with aprotinin during 5
years following coronary artery bypass graft surgery. JAMA 2007; 297: 471–479.
25. Fergusson DA, Hébert 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.
26. Ferraris VA, Brown JR, Despotis GJ et al.: 2011 update to the Society of Thoracic
Surgeons and the Society of Cardiovascular Anesthesiologists blood conservation clinical
practice guidelines. Ann Thorac Surg. Elsevier Inc.; 2011; 91: 944–982.
27. Beattie WS, Karkouti K: The post-BART anti-fibrinolytic dilemma? J Cardiothorac Vasc
Anesth Elsevier Inc.; 2011; 25: 3–5.
28. Karkouti K, Wijeysundera DN, Yau TM, McCluskey SA, Tait G, Beattie WS: The risk-
benefit profile of aprotinin versus tranexamic acid in cardiac surgery. Anesth Analg 2010; 110:
21–29.
29. Laupacis A, Fergusson D: Drugs to minimize perioperative blood loss in cardiac surgery.
Anesth Analg 1997; 85: 1258–1267.
30. Henry DA, Carless PA, Moxey AJ et al.: Anti-fibrinolytic use for minimising perioperative
allogeneic blood Transfusion Cochrane database Syst Rev 2007; (4): CD001886.
31. Henry D, Carless P: The safety of aprotinin and lysine-derived antifibrinolytic drugs in
cardiac surgery: a meta-analysis. Can Med Assoc J 2009; 180: 183–194.
32. Ngaage DL, Bland JM: Lessons from aprotinin: is the routine use and inconsistent dosing
of tranexamic acid prudent? Meta-analysis of randomised and large matched observational
studies. Eur J Cardio-thoracic Surg 2010; 37: 1375–1383.
33. Armellin G, Vinciguerra A, Bonato R, Pittarello D, Giron GP: Tranexamic acid in primary
CABG surgery: high vs low dose. Minerva Anestesiol 2004; 70: 97–107.
34. Dhir A: Antifibrinolytics in cardiac surgery. Ann Card Anaesth 2013; 16: 117–125.
35. Horrow JC, Van Riper DF, Strong MD, Grunewald KE, Parmet JL: Dose response of
tranexamic acid. Anesthesiology 1995; 82: 383–392.
36. Nuttall GA, Gutierrez MC, Dewey JD et al.: A preliminary study of a new tranexamic acid
dosing schedule for cardiac surgery. J Cardiothorac Vasc Anesth 2008; 22: 230–235.
37. Dowd NP, Karski JM, Cheng DC et al.: Pharmacokinetics of tranexamic acid during
cardiopulmonary bypass. Anesthesiology 2002; 97: 390–399.
38. Sigaut S, Tremey B, Ouattara A: Comparison of two doses of tranexamic acid in adults
undergoing cardiac surgery with cardiopulmonary bypass. Anesthesiology 2014; 2: 590–600.
18
39. Weber CF, Görlinger K, Byhahn C et al.: Tranexamic acid partially improves platelet
function in patients treated with dual antiplatelet therapy. Eur J Anaesthesiol 2011; 28: 57–62.
40. Schouten ES, van de Pol AC, Schouten ANJ, Turner NM, Jansen NJG, Bollen CW: The
effect of aprotinin, tranexamic acid, and aminocaproic acid on blood loss and use of blood
products in major pediatric surgery: a meta-analysis. Pediatr Crit Care Med 2009; 10: 182–190.
41. Shimizu K, Toda Y, Iwasaki T et al.: Effect of tranexamic acid on blood loss in pediatric
cardiac surgery: a randomized trial. J Anesth 2011; 25: 823–830.
42. Giordano R, Palma G, Poli V et al.: Tranexamic acid therapy in pediatric cardiac surgery:
a single-center study. Ann Thorac Surg 2012; 94: 1302–1306.
43. Faraoni D, Willems A, Melot C, De Hert S, Van der Linden P: Efficacy of tranexamic acid
in paediatric cardiac surgery: a systematic review and meta-analysis. Eur J Cardiothorac Surg
2012; 42: 781–786.
44. Goobie SM, Meier PM, Sethna NF et al.: Population pharmacokinetics of tranexamic acid
in paediatric patients undergoing craniosynostosis surgery. Clin Pharmacokinet 2013; 52: 267–
276.
45. Yee BE, Wissler RN, Zanghi CN, Feng C, Eaton MP: The effective concentration of
tranexamic acid for inhibition of fibrinolysis in neonatal plasma in vitro. Anesth Analg 2013; 117:
767–772.
46. Faraoni D, Goobie SM: New insights about the use of tranexamic acid in children
undergoing cardiac surgery: from pharmacokinetics to pharmacodynamics. Anesth Analg 2013;
117: 760–762.
47. Faraoni D, Goobie SM: The efficacy of antifibrinolytic drugs in children undergoing
noncardiac surgery: a systematic review of the literature. Anesth Analg 2014; 118: 628–636.
48. Tzortzopoulou A, Cepeda MS, Schumann R, Carr DB: Antifibrinolytic agents for reducing
blood loss in scoliosis surgery in children. Cochrane database Syst Rev 2008; (3): CD006883.
49. Neilipovitz DT, Murto K, Hall L, Barrowman NJ, Splinter WM: A randomized trial of
tranexamic acid to reduce blood transfusion for scoliosis surgery. Anesth Analg 2001: 82–87.
50. Sethna NF, Zurakowski D, Brustowicz RM, Bacsik J, Sullivan LJ, Shapiro F: Tranexamic
acid reduces intraoperative blood loss in pediatric patients undergoing scoliosis surgery.
Anesthesiology 2005; 102: 727–732.
51. Xu C, Wu A, Yue Y: Which is more effective in adolescent idiopathic scoliosis surgery:
batroxobin, tranexamic acid or a combination? Arch Orthop Trauma Surg 2012; 132: 25–31.
52. Basta MN, Stricker PA, Taylor JA: A systematic review of the use of antifibrinolytic
agents in pediatric surgery and implications for craniofacial use. Pediatr Surg Int 2012; 28: 1059–
1069.
19
53. Holcomb JB: Tranexamic acid in elective craniosynostosis surgery: it works, but how?
Anesthesiology 2011; 114: 737–738.
54. Goobie SM, Meier PM, Pereira LM et al.: Efficacy of tranexamic acid in pediatric
craniosynostosis surgery: a double-blind, placebo-controlled trial. Anesthesiology 2011; 114:
862–871.
55. Dadure C, Sauter M, Bringuier S et al.: Intraoperative tranexamic acid reduces blood
transfusion in children undergoing craniosynostosis surgery: a randomized double-blind study.
Anesthesiology 2011; 114: 856–861.
56. Kagoma YK, Crowther MA, Douketis J, Bhandari M, Eikelboom J, Lim W: Use of
antifibrinolytic therapy to reduce transfusion in patients undergoing orthopedic surgery: a
systematic review of randomized trials. Thromb Res. Elsevier B.V. 2009; 123: 687–696.
57. Poeran J, Rasul R, Suzuki S et al.: Tranexamic acid use and postoperative outcomes in
patients undergoing total hip or knee arthroplasty in the United States: retrospective analysis of
effectiveness and safety. BMJ 2014; 349: g4829.
58. Sukeik M, Alshryda S, Haddad FS, Mason JM: Systematic review and meta-analysis of
the use of tranexamic acid in total hip replacement. J Bone Joint Surg Br 2011; 93: 39–46.
59. Alshryda S, Sarda P, Sukeik M, Nargol a, Blenkinsopp J, Mason JM: Tranexamic acid in
total knee replacement: a systematic review and meta-analysis. J Bone Joint Surg Br 2011; 93:
1577–1585.
60. Klenerman L, Chakrabarti R, Mackie I: Changes in haemostatic system after application
of a tourniquet. Lancet 1977; 970–972.
61. Yang B, Li H, Wang D, He X, Zhang C, Yang P: Systematic review and meta-analysis of
perioperative intravenous tranexamic acid use in spinal surgery. PLoS One 2013; 8: e55436.
62. Tsutsumimoto T, Shimogata M, Ohta H, Yui M, Yoda I, Misawa H: Tranexamic acid
reduces perioperative blood loss in cervical laminoplasty: a prospective randomized study. Spine
(Phila Pa 1976). 2011; 36: 1913–1918.
63. Perel P, Ker K, Morales Uribe CH, Roberts I: Tranexamic acid for reducing mortality in
emergency and urgent surgery. Cochrane database Syst Rev 2013; 1: CD010245.
64. Wong J, Abrishami A, El Beheiry H et al.: Topical application of tranexamic acid reduces
postoperative blood loss in total knee arthroplasty: a randomized, controlled trial. J Bone Joint
Surg Am 2010; 92: 2503–2513.
65. Baric D, Biocina B, Unic D et al.: Topical use of antifibrinolytic agents reduces
postoperative bleeding: a double-blind, prospective, randomized study. Eur J Cardiothorac Surg
2007; 31: 366–371; discussion 371.
20
66. Fawzy H, Elmistekawy E, Bonneau D, Latter D, Errett L: Can local application of
Tranexamic acid reduce post-coronary bypass surgery blood loss? A randomized controlled trial.
J Cardiothorac Surg 2009; 4: 25.
67. Dell’Amore A, Caroli G, Nizar A et al.: Can topical application of tranexamic acid reduce
blood loss in thoracic surgery? A prospective randomised double blind investigation. Heart Lung
Circ 2012; 21: 706–710.
68. Nouraei M, Gholipour Baradari A, Ghafari R, Habibi MR, Emami Zeydi A, Sharifi N:
Decreasing blood loss and the need for transfusion after CABG surgery: a double-blind
randomized clinical trial of topical tranexamic acid. Turkish J Med Sci 2013; 43: 273–278.
69. Alshryda S, Mason J, Sarda P et al.: Topical (intra-articular) tranexamic acid reduces
blood loss and transfusion rates following total hip replacement: a randomized controlled trial
(TRANX-H). J Bone Joint Surg Am 2013; 95: 1969–1974.
70. Alshryda S, Mason J, Vaghela M et al.: Topical (intra-articular) tranexamic acid reduces
blood loss and transfusion rates following total knee replacement: a randomized controlled trial
(TRANX-K). J Bone Joint Surg Am 2013; 95: 1961–1968.
71. Chang C-H, Chang Y, Chen DW, Ueng SWN, Lee MS: Topical tranexamic acid reduces
blood loss and transfusion rates associated with primary total hip arthroplasty. Clin Orthop Relat
Res 2014; 472: 1552–1557.
72. Zhang Y, Fu X, Liu W-X, Li Y-M, Ma X-L, Li Z-J: Safety and efficacy of intra-articular
injection of tranexamic acid in total knee arthroplasty. Orthopedics 2014; 37: e775–e782.
73. Albirmawy OA, Saafan ME, Shehata EM, Basuni AS, Eldaba AA: Topical application of
tranexamic acid after adenoidectomy: a double-blind, prospective, randomized, controlled study.
Int J Pediatr Otorhinolaryngol. Elsevier Ireland Ltd; 2013; 77: 1139–1142.
74. Athanasiadis T, Beule AG, Wormald PJ: Effects of topical antifibrinolytics in endoscopic
sinus surgery: a pilot randomized controlled trial. Am J Rhinol 2007; 21: 737–742.
75. Sindet-Pedersen S, Ramström G, Bernvil S, Blombäck M: Hemostatic effect of tranexamic
acid mouthwash in anticoagulant-treated patients undergoing oral surgery. N Engl J Med 1989;
320: 840–843.
76. Lee APH, Boyle CA, Savidge GF, Fiske J: Effectiveness in controlling haemorrhage after
dental scaling in people with haemophilia by using tranexamic acid mouthwash. Br Dent J 2005;
198: 33–38; discussion 26.
77. Kaewpradub P, Apipan B, Rummasak D: Does tranexamic acid in an irrigating fluid
reduce intraoperative blood loss in orthognathic surgery? A double-blind, randomized clinical
trial. J Oral Maxillofac Surg 2011; 69: e186–e189.
21
78. Panteli M, Papakostidis C, Dahabreh Z, Giannoudis PV: Topical tranexamic acid in total
knee replacement: a systematic review and meta-analysis. Knee. Elsevier B.V. 2013; 20: 300–
309.
79. Abrishami A, Chung F, Wong J: Topical application of antifibrinolytic drugs for on-pump
cardiac surgery: a systematic review and meta-analysis. Can J Anaesth 2009; 56: 202–212.
80. Mahaffey R, Wang L, Hamilton A, Phelan R, Arellano R: A retrospective analysis of blood
loss with combined topical and intravenous tranexamic acid after coronary artery bypass graft
surgery. J Cardiothorac Vasc Anesth Elsevier Inc 2013; 27: 18–22.
81. Spegar J, Vanek T, Snircova J et al.: Local and systemic application of tranexamic acid in
heart valve surgery: a prospective, randomized, double blind LOST study. J Thromb
Thrombolysis 2011; 32: 303–310.
82. Shakur H, Roberts I, Bautista R et al.: Effects of tranexamic acid on death, vascular
occlusive events, and blood transfusion in trauma patients with significant haemorrhage
(CRASH-2): a randomised, placebo-controlled trial. Lancet. Elsevier Ltd 2010; 376: 23–32.
83. 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 Elsevier Ltd 2011; 377: 1096–101, 1101.e1–2.
84. Moore HB, Moore EE, Gonzalez E et al.: Hyperfibrinolysis, physiologic fibrinolysis, and
fibrinolysis shutdown: the spectrum of postinjury fibrinolysis and relevance to antifibrinolytic
therapy. J Trauma Acute Care Surg 2014; 00(00): 1–6.
85. Meier K, Hoesch R: Antifibrinolytic Therapy in Intracranial Hemorrhage. Drug Dev Res.
2013; 74: 478–484.
86. Roos YBWEM, Rinkel GJE, Vermeulen M, Algra A, van Gijn J: Antifibrinolytic therapy
for aneurysmal subarachnoid haemorrhage. Cochrane database Syst Rev 2003; (2): CD001245.
87. Roos Y: Antifibrinolytic treatment in subarachnoid hemorrhage: a randomized placebo-
controlled trial. STAR Study Group. Neurology 2000; 54: 77–82.
88. Albuquerque FC: Rebleeding and its prevention after subarachnoid hemorrhage. World
Neurosurg Elsevier Inc 2013; 79: 245–246.
89. Starke RM, Kim GH, Fernandez A et al.: Impact of a protocol for acute antifibrinolytic
therapy on aneurysm rebleeding after subarachnoid hemorrhage. Stroke 2008; 39: 2617–2621.
90. Harrigan MR, Rajneesh KF, Ardelt AA, Fisher WS: Short-term antifibrinolytic therapy
before early aneurysm treatment in subarachnoid hemorrhage: effects on rehemorrhage, cerebral
ischemia, and hydrocephalus. Neurosurgery 2010; 67: 935–939; discussion 939–940.
22
91. Hillman J, Fridriksson S, Nilsson O, Yu Z, Saveland H, Jakobsson K-E: Immediate
administration of tranexamic acid and reduced incidence of early rebleeding after aneurysmal
subarachnoid hemorrhage: a prospective randomized study. J Neurosurg 2002; 97: 771–778.
92. Larsen CC, Astrup J: Rebleeding after aneurysmal subarachnoid hemorrhage: A literature
review. World Neurosurgery Elsevier Inc 2013: 307–312.
93. Chwajol M, Starke RM, Kim GH, Mayer SA, Connolly ES: Antifibrinolytic therapy to
prevent early rebleeding after subarachnoid hemorrhage. Neurocrit Care 2008; 8: 418–426.
94. Perel P, Al-Shahi Salman R, Kawahara T et al.: Effect of tranexamic acid in traumatic
brain injury: a nested randomised, placebo controlled trial (CRASH-2 Intracranial Bleeding
Study). BMJ 2011; 343: d3795.
95. Sprigg N, Renton CJ, Dineen RA, Kwong Y, Bath PMW: Tranexamic acid for spontaneous
intracerebral hemorrhage: a randomized controlled pilot trial (ISRCTN50867461). J Stroke
Cerebrovasc Dis Elsevier Ltd 2014; 23: 1312–1318.
96. Sprigg N: ISRCTN93732214: Tranexamic acid for IntraCerebral Haemorrhage (TICH-2)
[Internet]. ISRCTN Registry. 2013 [cited 2014 Dec 15]. Available from: http:
//www.isrctn.com/ISRCTN93732214
97. Meretoja A, Churilov L, Campbell BCV et al.: The spot sign and tranexamic acid on
preventing ICH growth-AUStralasia Trial (STOP-AUST): protocol of a phase II randomized,
placebo-controlled, double-blind, multicenter trial. Int J Stroke 2014; 9: 519–524.
98. Steib A, Gengenwin N, Freys G, Boudjema K, Levy S, Otteni JC: Predictive factors of
hyperfibrinolytic activity during liver transplantation in cirrhotic patients. Br J Anaesth 1994; 73:
645–648.
99. Segal H, Cottam S, Potter D, Hunt BJ: Coagulation and fibrinolysis in primary biliary
cirrhosis compared with other liver disease and during orthotopic liver transplantation.
Hepatology 1997; 25: 683–688.
100. Xia VW, Steadman RH: Antifibrinolytics in orthotopic liver transplantation: current status
and controversies. Liver Transpl 2005; 11: 10–18.
101. Molenaar IQ, Warnaar N, Groen H, Tenvergert EM, Slooff MJH, Porte RJ: Efficacy and
safety of antifibrinolytic drugs in liver transplantation: a systematic review and meta-analysis.
Am J Transplant 2007; 7: 185–194.
102. Dalmau A, Sabaté A, Koo M et al.: The prophylactic use of tranexamic acid and aprotinin
in orthotopic liver transplantation: a comparative study. Liver Transpl 2004; 10: 279–284.
103. Ickx BE, van der Linden PJ, Melot C et al.: Comparison of the effects of aprotinin and
tranexamic acid on blood loss and red blood cell transfusion requirements during the late stages
of liver transplantation. Transfusion 2006; 46: 595–605.
23
104. Gurusamy KS, Pissanou T, Pikhart H, Vaughan J, Burroughs AK, Davidson BR: Methods
to decrease blood loss and transfusion requirements for liver transplantation. Cochrane database
Syst Rev 2011; (12): CD009052.
105. Massicotte L, Denault AY, Beaulieu D, Thibeault L, Hevesi Z, Roy A: Aprotinin versus
tranexamic acid during liver transplantation: impact on blood product requirements and survival.
Transplantation 2011; 91: 1273–1278.
106. Görlinger K. Coagulation management during liver transplantation. Hamostaseologie
2006; 26 (3 Suppl 1): S64–76.
107. Wu C-C, Ho W-M, Cheng S-B et al.: Perioperative parenteral tranexamic acid in liver
tumor resection: a prospective randomized trial toward a “blood transfusion” — free
hepatectomy. Ann Surg 2006; 243: 173–180.
108. Gurusamy KS, Li J, Sharma D, Davidson BR: Pharmacological interventions to decrease
blood loss and blood transfusion requirements for liver resection. Cochrane database Syst Rev
2009; (4): CD008085.
109. Truong JL, Cyr DP, Lam-McCulloch J, Cleary SP, Karanicolas PJ: Consensus and
controversy in hepatic surgery: a survey of Canadian surgeons. J Surg Oncol 2014; 110: 947–951.
110. Lumsden MA, Wedisinghe L: Tranexamic acid therapy for heavy menstrual bleeding.
Expert Opin Pharmacother 2011; 12: 2089–2095.
111. Lethaby A, Farquhar C, Cooke I: Antifibrinolytics for heavy menstrual bleeding.
Cochrane database Syst Rev 2000; (4): CD000249.
112. Lukes AS, Moore KA, Muse KN et al.: Tranexamic acid treatment for heavy menstrual
bleeding: a randomized controlled trial. Obstet Gynecol 2010; 116: 865–875.
113. Winkler UH: The effect of tranexamic acid on the quality of life of women with heavy
menstrual bleeding. Eur J Obstet Gynecol Reprod Biol 2001; 99: 238–243.
114. Edlund M, Andresson K, Rybo G, Lindoff C, Astedt B, Schoultz B: Reduction of menstrual
blood loss in women suffering from idiopathic menorrhagia with a novel antifibrinolytic drug
(Kabi 2161). BJOG An Int J Obstet Gynaecol 1995; 102: 913–917.
115. Novikova N, Hofmeyr GJ: Tranexamic acid for preventing postpartum haemorrhage.
Cochrane database Syst Rev. Chichester, UK: John Wiley & Sons, Ltd 2010; (7): CD007872.
116. Clinical study on the efficacy of tranexamic acid in reducing postpartum blood lose: a
randomized, comparative, multicenter trial. Zhonghua Fu Chan Ke Za Zhi. 2001; 36: 590–592.
117. Gai M, Wu L, Su Q, Tatsumoto K: Clinical observation of blood loss reduced by
tranexamic acid during and after caesarian section: a multi-center, randomized trial. Eur J Obstet
Gynecol Reprod Biol 2004; 112: 154–157.
24
118. Peitsidis P, Kadir RA: Antifibrinolytic therapy with tranexamic acid in pregnancy and
postpartum. Expert Opin Pharmacother 2011; 12: 503–516.
119. Gohel M, Patel P, Gupta A, Desai P: Efficacy of tranexamic acid in decreasing blood loss
during and after cesarean section: A randomized case controlled prospective study. J Obstet
Gynecol India 2007; 57: 227–230.
120. Sekhavat L, Tabatabaii A, Dalili M, Farajkhoda T, Tafti AD: Efficacy of tranexamic acid
in reducing blood loss after cesarean section. J Matern Fetal Neonatal Med 2009; 22: 72–75.
121. Gobbur V, Shiragur S, Jhanwar U, Tehalia M: Efficacy of tranexamic acid in reducing
blood loss during lower segment caesarean section. Int J Reprod Contraception, Obstet Gynecol
2014; 3: 414.
122. Ferrer P, Roberts I, Sydenham E, Blackhall K, Shakur H: Anti-fibrinolytic agents in post
partum haemorrhage: a systematic review. BMC Pregnancy Childbirth 2009; 9: 29.
123. Ducloy-Bouthors A-S, Jude B, Duhamel A et al.: High-dose tranexamic acid reduces blood
loss in postpartum haemorrhage. Crit Care 2011; 15: R117.
124. Gungorduk K, Ascoğlu O, Yldrm G, Ark C, Tekirdağ Aİ, Besmoglu B: Can intravenous
injection of tranexamic acid be used in routine practice with active management of the third stage
of labor in vaginal delivery? A Randomized Controlled Study. Obstet Gynecol Surv 2013; 68:
673–675.
125. Gungorduk K, Yildirim G, Asicioglu O, Gungorduk Ozgu C, Sudolmus S, Ark C: Efficacy
of intravenous tranexamic acid in reducing blood loss after elective cesarean section: a
prospective, randomized, double-blind, placebo-controlled study. Am J Perinatol 2011; 28: 233–
239.
126. Xu J, Gao W, Ju Y: Tranexamic acid for the prevention of postpartum hemorrhage after
cesarean section: a double-blind randomization trial. Arch Gynecol Obstet 2013; 287: 463–468.
127. Movafegh A, Eslamian L, Dorabadi A: Effect of intravenous tranexamic acid
administration on blood loss during and after cesarean delivery. Int J Gynaecol Obstet
International Federation of Gynecology and Obstetrics 2011; 115: 224–226.
128. Sentürk MB, Cakmak Y, Yildiz G, Yildiz P: Tranexamic acid for cesarean section: a double-
blind, placebo-controlled, randomized clinical trial. Arch Gynecol Obstet 2013; 287: 641–645.
129. Chan CC, Chan YY, Tanweer F: Systematic review and meta-analysis of the use of
tranexamic acid in tonsillectomy. Eur Arch Otorhinolaryngol 2013; 270: 735–748.
130. Sheffer AL, Fearon DT, Austen KF, Rosen FS: Tranexamic acid: preoperative prophylactic
therapy for patients with hereditary angioneurotic edema. J Allergy Clin Immunol 1977; 60: 38–
40.
25
131. Laurberg G: Plasma kinin activation in tranexamic acid treated patients with hereditary
angioneurotic edema. Arch Dermatol Res 1978; 262: 153–156.
132. Gluud LL, Klingenberg SL, Langholz E: Tranexamic acid for upper gastrointestinal
bleeding. Cochrane database Syst Rev 2012; 1(1): CD006640.
133. Martí-Carvajal AJ, Solà I, Martí-Carvajal PI: Antifibrinolytic amino acids for upper
gastrointestinal bleeding in patients with acute or chronic liver disease. Cochrane database Syst
Rev 2012; 9: CD006007.
134. Prutsky G, Domecq JP, Salazar CA, Accinelli R: Antifibrinolytic therapy to reduce
haemoptysis from any cause. Cochrane database Syst Rev 2012; 4(4): CD008711.
135. Schlag M, Hopf R, Zifko U, Redl H: Epileptic seizures following cortical application of
fibrin sealants containing tranexamic acid in rats. Acta Neurochir (Wien) 2002; 144: 63–69.
136. Pellegrini A, Giaretta D, Chemello R, Zanotto L, Testa G: Feline Generalized Epilepsy
Induced by Tranexamic Acid (AMCA). Epilepsia 1982; 23: 35–45.
137. Mohseni K, Jafari A, Nobahar MR, Arami A: Polymyoclonus seizure resulting from
accidental injection of tranexamic acid in spinal anesthesia. Anesth Analg 2009; 108: 1984–1986.
138. Sabzi F, Teimouri H, Zokai A: Myoclonus, seizure, and ventricular fibrillation after
intrathecal injection of tranexamic acid. J Tehran Univ Hear Cent 2009; 4: 253–255.
139. Kaabachi O, Eddhif M, Rais K, Zaabar MA: Inadvertent intrathecal injection of
tranexamic acid. Saudi J Anaesth 2011; 5: 90–92.
140. 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.
141. Murkin JM, Falter F, Granton J, Young B, Burt C, Chu M: High-dose tranexamic Acid is
associated with nonischemic clinical seizures in cardiac surgical patients. Anesth Analg 2010;
110: 350–353.
142. 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.
143. Koster A, Börgermann J, Zittermann A, Lueth JU, Gillis-Januszewski T, Schirmer U:
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.
144. Martin K, Breuer T, Gertler R et al.: Tranexamic acid versus ɛ-aminocaproic acid:
efficacy and safety in paediatric cardiac surgery. Eur J Cardiothorac Surg 2011; 39: 892–897.
145. Lecker I, Orser B a, Mazer CD: “Seizing” the opportunity to understand antifibrinolytic
drugs. Can J Anaesth 2012; 59: 1–5.
26
146. Andersson L, Nilsoon IM, Colleen S, Granstrand JB, Melander B: Role of urokinase and
tissue activator in sustaining bleeding and the management thereof with EACA and AMCA. Ann
NY Acad Sci 1968; 146: 642–656.
147. Benoni G, Björkman S, Fredin H: Application of pharmacokinetic data from healthy
volunteers for the prediction of plasma concentrations of tranexamic acid in surgical patients.
Clin Drug Investig 1995; 10: 280–287.
148. Fiechtner BK, Nuttall GA, Johnson ME et al.: Plasma tranexamic acid concentrations
during cardiopulmonary bypass. Anesth Analg 2001; 92: 1131–1136.
149. Andersson L, Eriksson O, Hedlund P-O, Kjellman H, Lindqvist B: Special considerations
with regard to the dosage of tranexamic acid in patients with chronic renal diseases. Urol Res
1978; 6: 83–88.
150. 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.
151. Jerath A, Wasowicz M, Ph D, Yang Q, Sc B, Pang KS: Safer tranexamic acid dosing for
patients with renal dysfunction undergoing cardiac surgery [Internet]. ASA Abstract. ASA; 2013
[cited 2015 Jan 14]. p. 1. Available from: http:
//www.asaabstracts.com/strands/asaabstracts/abstract.htm;
jsessionid=6B61B2C90FD0F04CA9ECC17977AC6B4C?year=2013&index=7&absnum=3955
152. Ferring Pharmaceuticals. LYSTEDA TM (tranexamci acid) Tablets - PI [Internet].
Parsippany: Ferring Pharmaceuticals; 2013 [cited 2014 Dec 7]. p. 1–20. Available from: http:
//lysteda.com/assets/pi_ferring2013.pdf
153. Ma SCA, Brindle W, Burton G et al.: Tranexamic acid is associated with less blood
transfusion in off-pump coronary artery bypass graft surgery: a systematic review and meta-
analysis. J Cardiothorac Vasc Anesth 2011; 25: 26–35.
154. Ogawa S, Richardson JE, Sakai T, Ide M, Tanaka KA: High mortality associated with
intracardiac and intrapulmonary thromboses after cardiopulmonary bypass. J Anesth 2012; 26: 9–
19.
155. Sundström A, Seaman H, Kieler H, Alfredsson L: The risk of venous thromboembolism
associated with the use of tranexamic acid and other drugs used to treat menorrhagia: a case-
control study using the General Practice Research Database. BJOG 2009; 116: 91–97.
156. Nardi K, Pelone G, Bartolo M et al.: Ischaemic stroke following tranexamic acid in young
patients carrying heterozygosity of MTHFR C677T. Ann Clin Biochem 2011; 48: 575–578.
27
157. Dewan Y, Komolafe EO, Mejía-Mantilla JH, Perel P, Roberts I, Shakur H: CRASH-3 —
tranexamic acid for the treatment of significant traumatic brain injury: study protocol for an
international randomized, double-blind, placebo-controlled trial. Trials 2012; 13: 87.
158. Shakur H, Elbourne D, Gülmezoglu M et al.: Study protocol The WOMAN Trial (World
Maternal Antifibrinolytic Trial): tranexamic acid for the treatment of postpartum haemorrhage :
an international randomised, double blind placebo controlled trial. Trials 2010; 11: 40–53.
159. Myles PS, Smith J, Knight J et al.: Aspirin and Tranexamic Acid for Coronary Artery
Surgery (ATACAS) Trial: rationale and design. Am Heart J 2008; 155: 224–230.
Corresponding author:
Marcin Wasowicz, MD
Department of Anesthesia EN3-438,
Toronto General Hospital, 200 Elizabeth St,
Toronto, ON M5G2C4, Canada
Tel.: 1 (416) 340-3567, fax: 1 (416) 340-3698
e-mail: Marcin.Wasowicz@uhn.ca
Received: 10.02.2015
Accepted: 22.02.2015
28
Table 1 − Major studies of tranexamic acid in cardiac surgery surgery
Study LOE n & population Intervention & Comparator Relevant Outcome
Mangano et al.
[23] 2b 4374 adults for
CABG AP, TXA & ACA vs control
All antifibrinolytics reduced blood loss. AP doubled
risk of renal failure, increased risk of stroke or
myocardial infarction
Karkouti et al.
[2] 2b
898 propensity
matched adults for
high − risk CS
AP vs TXA
TXA and AP have similar hemostatic effectiveness;
AP group had higher rates of renal dysfunction (24%
vs 17%, P = 0.01)
Mangano et al.
[24] 2b 4374 adults after
CABG
AP vs control, TXA vs
control, ACA vs control
AP increased mortality risk 20.8% vs 12.7% at 5
years. TXA/ACA did not show increased risk
BART trial,
Fergusson et
al. [25]
1b 2331 adults for
high − risk CS AP vs TXA vs ACA
AP group had higher mortality 6% vs TXA 3.9% and
ACA 4.0%; AP showed modest decrease in massive
bleeding vs TXA/ACA
Henry et al.
[31, 30] 1a
25000+ adults for
major surgery
including 3006 CS
subgroup
TXA vs control, AP vs
control, ACA vs control
In CS subgroup: TXA vs had RR 0.68 for BTx,
perioperative BL − 300 mL
Ngaage et al.
[32]
1a,
2a
10000+ adults for
CS
TXA vs control, AP vs
control, TXA vs AP
RCTs alone: TXA had OR 0.53 for BTx, BL 298 mL
less. Combined RCT & OS: TXA BL − 283 mL
Schouten et al.
[40] 1a 1893 children for
CS
TXA vs control, AP vs
control, ACA vs control
TXA group had BL 11 mL/kg less and reduced BTx
by 4 mL/kg. TXA not inferior to AP
n − number of patients, LOE − Level of evidence, TXA − tranexamic acid, AP − aprotinin, ACA − aminocaproic acid, CS −
cardiac surgery CABG − coronary arterial bypass grafts, HR − hazard ratio, OR − odds ratio, RR − relative risk, BTx −
blood transfusion, BL − blood loss, RCT − randomized controlled trials, OS − observational studies
Table 2 − Major studies of tranexamic acid in orthopedic surgery
Study LOE population & n Intervention & Comparator Outcome
Kagoma et al.
[56] 1a
Patients for THR or
TKA. 1432 for BL,
1557 for BTx, 1637
for VTE meta −
analysis
ATF vs control
ATF group had BTx RR 0.52 (p <0.00001) and less
BL 1.1 SMD vs control. No increased risk of VTE.
TXA group had less BL 393 mL vs control.
Poeran et al.
[57] 2b 872416 patients for
THR or TKA
TXA ≤ 1g, 2g or ≥ 3g vs
None
TXA group had lower rates BTx 7.7 vs 20.1%, RD
1.2 vs 1.6%. Decreasing OR 0.31 − 0.38 (p<0.001)
as TXA dose increased. No increase in VTE or other
complications.
Sukeik et al.
[58] 1a 350 patients for
THR TXA vs control
Less intraoperative BL by 104 mL, postoperative BL
by 172 mL, decrease RD for BTx − 0.20
(p<0.0001).
Alshryda et al.
[59] 1a
Patients for TKA,
763 for BL, 824 for
BTx analysis
TXA vs control BL reduced by 591 mL, RR of BTx 2.56 (95% CI
2.1 − 3.1) in favour of TXA.
Yang et al.
[60] 1a 581 patients for
spinal surgery TXA vs control BL reduced by 389 mL. No increase in VTE.
Schouten et al.
[40] 1a 207 children for
scoliosis surgery
TXA vs control, AP vs
control BL reduced by 682 mL. Not inferior to AP.
n − number of patients, LOE − Level of evidence, THR − total hip replacement, TKA − total knee arthroplasty, ATF −
antifibrinolytic, TXA − tranexamic acid, AP − aprotinin, ACA − aminocaproic acid, BTx − blood transfusion, BL − blood
loss, SMD − standardized mean difference, VTE − venous thromboembolism, RD − renal dysfunction, RR − relative risk,
RD − risk difference
Table 3 − Major studies of topical tranexamic acid use in surgery
Study LOE population & n Intervention Outcome
Panteli et al.
[78] 1a
Patients for TKA.
512 in BTx, 257 in
BL, 178 in DO meta
− analyses
Topical TXA in NS vs
control
RR of BTx 0.41 (P = 0.05), reduced postoperative
BL by 220 mL, and DO by 268 mL. Higher dose > 2
g TXA further significantly reduced BL. No increase
VTE
Alshryda et al.
[69] 1b 161 patients for
THR
Intra − articular injection
TXA vs control
Reduction AR by 19.6%, CPE by £305, reduced BL
by 129 mL, Hb drop by 0.84g dl-1
Alshryda et al.
[70] 1b 157 patients for
TKA
Intra − articular injection
TXA vs control
Reduction AR by 15.4%, CPE by £333, LOS by
1.2d, reduced BL by 168 mL, Hb drop by 0.83g dl-1
Abrishami et
al. [79] 1a 622 patients for CS Topical TXA/AP vs control
Reduced chest DO BL at 24h by 220 mL, overal
reduction of 1 unit PC per patient (95% CI − 1.54 to
− 0.53, P < 0.0001)
n − number of patients, LOE − Level of evidence, TXA − tranexamic acid, AP − aprotinin, ACA − aminocaproic acid, TKA
− total knee arthroplasty, THR − total hip replacement, CS − cardiac surgery, BTx − blood transfusion, BL − blood loss, DO
− drain output, VTE − venous thromboembolism, RR − relative risk, AR − absolute risk, CPE − cost per episode, Hb −
hemoglobin, LOS − length of stay in days (d), PC − packed red blood cells
Table 4 − Major studies of tranexamic acid in obstetrics & gynecology
Study LOE population & n Intervention Outcome
Lethaby et al.
[111]
1a,
2a*
Women of
reproductive age
with MN: 84 in
TXA vs placebo
analysis, 171 in
TXA vs alternative
analysis.
TXA or precursor vs
placebo, TXA vs alternative
therapy NSAID, ETM, PRO
WMD in BL of 94 mL in favour of TXA. TXA
reduced BL compared to alternatives
Novikova et
al. [115] 1a 453 women for CS
or VD TXA vs control BL of > 400 mL less common in TXA group. WMD
− 75 mL
Gungorduk et
al. [125] 1b 660 women for CS Pre − CS intravenous TXA
vs control
Lower mean BL (500 vs 600 mL). RR of PPH 2.7
(95% CI 1.1−6.3), and RR of additional uterotonics
1.7 (95% CI 1.1−2.6) in favour of TXA
n − number of patients, LOE − Level of evidence, TXA − tranexamic acid, AP − aprotinin, ACA − aminocaproic acid, MN
− menorrhagia or heavy menstrual bleeding, CS − cesarean section, VD − vaginal delivery, PPH − post − partum
hemorrhage, VTE − venous thromboembolism, BL − blood loss, WMD − weighted mean difference, NSAID − non −
steroidal anti − inflammatory mefenamic acid, ETM − ethamsylate, PRO − oral luteal phase progestagen norethisterone, RR −
relative risk, RCT − randomized controlled trial
*3 single RCT analyses of TXA vs NSAID, TXA vs ETM,and TXA vs
PRO therapies
Table 5 − Major studies on tranexamic acid in trauma and neurosurgery
Study LOE population & n Intervention & Comparator Outcome
CRASH − 2
by Shakur et
al. [82]
1b 20211 adult trauma
patients
1 g TXA on admission then
1g over 8h vs placebo
All cause mortality reduced, RR 0.91 (95% CI
0.85−0.97). Death from bleeding reduced, RR 0.85
(95% CI 0.76−0.96).
Roberts et al.
[83] 2b 20211 adult trauma
patients
Same as CRASH − 2.
Exploratory analysis of
treatment ≤ 1h, 1−3h, ≥ 3h
from injury
Early (< 1h) TXA had RR of death 0.68 (95% CI
0.57−0.82) and intermediate (1−3h) TXA had RR of
death 0.79 (95% CI 0.64−0.97). Late TXA > 3h had
increased RR of death 1.44 (95% CI 1.12−1.84)
Hillman et al.
[91] 1b 505 patients with
aneurysmal SAH
TXA 1g on diagnosis, then
1g every 6h until aneurysm
clipped, (max 72h) vs
control
Reduction in bleeding rate 10.8% to 2.4%, 80%
mortality reduction. No increased risk of ischemic
events by TCD
n − number of patients, LOE − Level of evidence, TXA − tranexamic acid, AP − aprotinin, ACA − aminocaproic acid, SAH
− subarachnoid hemorrhage, RR − relative risk, TCD − transcranial doppler studies
... This inhibition prevents the conversion of plasminogen to plasmin, which is responsible for degrading fibrin clots [1]. As a result, TXA can effectively stabilize formed clots and impede the breakdown of fibrin [2]. ...
Article
Full-text available
Background Intra-articular tranexamic acid (TXA) has been proven effective in reducing postoperative bleeding in anterior cruciate ligament reconstruction (ACLR). We aimed to evaluate the effect of intra-articular injection of TXA with different acting times after an ACLR procedure. Patients and methods Patients receiving ACLR and intra-articular injection of TXA between September 2023 and January 2024 were randomly divided into two groups, with drainage clamped for 4 h (TXA 4 h group) or 8 h (TXA 8 h group). Postoperative drainage output was the primary outcome. The secondary outcomes included the visual analog scale (VAS), grade of hemarthrosis, and International Knee Documentation Committee (IKDC) functional score. The data of another two groups of patients (TXA 2 h group and placebo group) were retrieved from a previous study as historical control groups for subsequent analysis. Results 121 patients were included. There were no significant differences in drainage output between TXA 4 h and TXA 8 h groups. On postoperative day 3, significantly decreased grades of hemarthrosis were noted in the TXA 8 h group ( P = 0.030). There were no significant differences in the VAS at different postoperative time points or in the IKDC scores. Comparison with the placebo and TXA 2 h groups revealed significant reduction in postoperative drainage among the TXA 4 h and 8 h groups. The IKDC scores were significantly worse in the TXA 8 h group compared with the TXA 2 h ( P < 0.001) and placebo ( P = 0.009) groups. Conclusions A 4 h clamping time for intra-articular TXA administration after ACLR may be considered in current practice, as it effectively reduces drainage and pain without negatively impacting functional outcomes. Level of evidence Level III, cohort study.
... [4,9,10] Tranexamic acid (TXA) is a synthetic derivative of lysine that reversibly blocks the lysine binding sites on plasminogen, and thus, blocks the action of plasmin that plays a role in the breakdown of fibrin and maintains and stabilizes the blood clot. [11] Studies have found reduced recurrence rate and reoperation rate with use of TXA in cSDH; however, the exact mechanism is not fully known, and it is believed to be owing to the stabilizing effect of the TXA on the blood clots that reduces the active bleeding and expansion of the hematoma cavity. [8,12,13] However, it is an active area of research, and the mechanism, efficacy, and optimal dose and timing of administration are yet to be fully elucidated. ...
Article
Full-text available
Chronic subdural hematoma (cSDH) is a prevalent neurosurgical disorder with increasing incidence, especially among the geriatric population. Although surgical management has been the conventional approach, the optimal strategy remains debated. Tranexamic acid (TXA), an antifibrinolytic agent, has emerged as a potential alternative, but its efficacy and safety require thorough investigation. This systematic review and meta-analysis aimed to evaluate the role of TXA in the management of cSDH, focusing on its impact on recurrence rates, clinical outcomes, and adverse events. Randomized controlled trials (RCTs) reporting on the safety and efficacy of TXA in adult patients with cSDH were systematically searched in multiple databases. Primary outcomes included clinical and radiological recurrence requiring re-surgery, while secondary outcomes encompassed adverse events and mortality associated with TXA treatment. A comprehensive search yielded 35 articles, with four RCTs meeting the inclusion criteria. The meta-analysis, involving 148 patients, demonstrated no significant reduction in mean postoperative volume at follow-up or recurrence rates with TXA use. Heterogeneity was low to moderate, and no significant adverse events were observed. Although there is a trend toward early and more complete resolution of cSDH with TXA acid after burr hole drainage, the results lack statistical significance to advocate its routine use in post-surgical management. TXA appears safe in cSDH, emphasizing the need for further research to establish its definitive role in reducing recurrence rates.
... The CRASH-3 trial also demonstrated its efficacy in trauma with severe traumatic brain injury [25]. TXA inhibits fibrinolysis by binding to lysine sites on plasmin and plasminogen [26], suggesting that it may be particularly effective in patients with traumainduced coagulopathy and DIC with fibrinolytic phenotype. ...
Article
Disseminated intravascular coagulation (DIC) is a complex condition with diverse etiologies. While its association with sepsis has been widely studied, less focus has been given to DIC arising from other critical conditions, such as trauma, burns, acute pancreatitis, and obstetric complications. The 2024 Clinical Practice Guidelines, developed by the Japanese Society on Thrombosis and Hemostasis (JSTH), aim to fill this gap and offer comprehensive recommendations for managing DIC across various conditions. This study, Part 4 of the guideline series, addresses DIC management in trauma, burns, obstetric complications, acute pancreatitis/liver failure, viral infections, and autoimmune diseases. For trauma-associated DIC, early administration of fresh-frozen plasma (FFP), coagulation factor concentrates such as fibrinogen and prothrombin complex concentrates, and tranexamic acid is recommended. The guidelines also highlight DIC in obstetrics, which is associated with massive bleeding, and recommend the administration of fibrinogen concentrate, antithrombin concentrate, and tranexamic acid. Through a systematic review of the current evidence, the guidelines provide stratified recommendations aimed at improving clinical outcomes in DIC management beyond sepsis, thereby serving as a valuable resource for healthcare providers globally.
... In the majority of studies, TXA has been validated to be effective in hemostasis without increasing the risk of thrombotic events (10). However, TXA may not only be contraindicated in patients with TXA allergy, a history of thromboembolism, but also is associated with epilepsy, nausea, vomiting, and diarrhea (11)(12)(13)(14)(15). Therefore, it is essential to find a safe and effective alternative hemostatic regimen for avoiding adverse drug reactions to TXA. ...
Article
Full-text available
Aims This study compared the hemostatic effects and complications of oxidized regenerated cellulose (ORC) and topical TXA in total knee arthroplasty (TKA), thus providing a reference for the use of ORC as an alternative hemostatic agent to TXA in TKA. Methods A total of 105 patients were included in this study and randomized into blank control, ORC, and TXA groups. The primary outcomes were total blood loss, hemoglobin drop (Hb drop), transfusion rates, and incidence of thrombosis. The secondary outcomes included operation time, tourniquet duration, coagulation parameters, inflammation markers, and complication rates. Results Total blood loss was 1,002.47 ± 308.58 ml and 964.68 ± 273.00 ml in the ORC and TXA groups, respectively, both significantly lower than that in the blank control group (1,168.94 ± 405.04 ml) (P1 = 0.043 and P2 = 0.014, respectively). Hb Drop was statistically insignificantly different between the ORC (36.03 ± 12.17 g/L) and TXA (34.32 ± 10.19 g/L) groups (P = 0.555). There was no statistically significant difference in transfusion rate, operation time, tourniquet duration, coagulation parameters, inflammation markers, and complication rates among the three groups. Conclusion In conclusion, our prospective randomized controlled trial (RCT) highlights that, oxidized regenerated cellulose (ORC) can reduce postoperative invisible blood loss in total knee arthroplasty and achieve a hemostatic effect similar to topical tranexamic acid (TXA). This provides a safe and effective hemostatic option for patients with severe underlying diseases or contraindications to tranexamic acid. Clinical Trial registration https://www.chictr.org.cn/bin/project/edit?pid=186370, identifier (ChiCTR2200066633).
... Fibrin clots formed during the surgical procedure are thereby preserved, reducing bleeding and hematoma formation. 10 The drug has been used for hip and knee arthroplasty and knee arthroscopic surgeries to reduce bleeding and transfusion requirements. It has been hypothesized that its use in arthroscopic RCR would also improve outcomes, thereby enhancing the ease of the procedure. ...
Article
Full-text available
Objective The aim of this study was to examine if tranexamic acid (TXA) can assist in improving outcomes of arthroscopic rotator cuff repair (RCR). Methods The databases of PubMed, Embase, Web of Science, CENTRAL, and Scopus were searched for all types of studies examining the efficacy of TXA for arthroscopic RCR. Twelve studies, 10 randomized controlled trials (RCTs), and 2 retrospective studies were considered eligible. Results Meta-analysis of only 2 studies using a visual clarity grading system showed better visualization with the use of TXA. A similar difference was noted for studies using the visual analog scale. Operating time was not significantly different between the groups, but subgroup analysis of RCTs demonstrated reduced operating time with TXA. Meta-analysis showed no difference in 24-hour pain scores between TXA and control groups. Qualitative assessment of studies for blood loss showed no significant effect of TXA. No major complications were reported in any of the studies. Conclusion This study has pooled evidence suggesting that TXA can improve visual clarity in arthroscopic RCR and may also result in a reduction in operating time. TXA does not seem to reduce blood loss or 24-hour postoperative pain scores. Level of Evidence Level II, Therapeutic Study.
Article
Background and Objectives Highly effective procedures for the preparation of allogeneic platelet gel (PG) use Ca‐gluconate and batroxobin, an expensive commercial reagent. In this preliminary study, we explored the use of the plasmin‐inhibitor, low‐cost drug tranexamic acid (TXA) in place of batroxobin, based on the literature supporting TXA ability to prevent fibrinolysis and stabilize the gel formed by fibrin polymerization prompted by Ca‐gluconate. Materials and Methods Eight blood centres determined PG weight and volume of non‐gelled, liquid portion in 116 PG prepared in 20‐mL commercial BioNest D bags. Ten‐millilitre platelet aliquots from platelets in 100% plasma or in 35% plasma/65% platelet additive solution (PAS) were aseptically transferred into the D bag, followed by the injection of 3.3 mL of 10% Ca‐gluconate and 0.4 mL of TXA. After 30‐min incubation, PG weight and non‐gelled liquid volume were determined. Results PG weight and liquid volume at 30 min were 6.5 ± 3.4 g and 7.4 ± 3.5 mL with platelets in 100% plasma, and 3.7 ± 3.0 g and 10.2 ± 3.3 mL with PAS platelets, respectively. Conclusion This study provides preliminary evidence supporting the use of TXA as a low‐cost reagent for PG manufacturing from platelets in 100% plasma.
Article
Introduction: Tranexamic acid (TA) is an anticoagulant drug that is used worldwide. However, the adverse effects of TA may insult the nervous system. This study aimed to investigate the dual effects of TA on SH-SY5Y cells, including its detrimental and neuroprotective effects. Methods: SH-SY5Y cells were treated with various concentrations of TA and exposed to H2O2 for 24 hours. The neuroprotective effects of TA were evaluated in H2O2-challenged cells. To assess the neuroprotective effects of TA, SH-SY5Y cells were pretreated with TA for 12 hours and then exposed to H2O2 for 24 hours. Cell viability was assessed using the MTT assay. Flow cytometry was used to evaluate cellular apoptosis. The expression of Bax, Bcl-2, and Caspase-3 genes was analyzed by real-time PCR. Additionally, Akt phosphorylation was evaluated using western blotting. Results: At high concentrations, TA reduced cell viability and induced apoptosis by up-regulating BAX and Caspase-3 gene expression and down-regulating BCL-2 transcript. Furthermore, Akt phosphorylation was reduced following TA treatment. TA exhibited protective effects against H2O2-induced cell stress by down-regulating Bax and Caspase-3 gene expression, up-regulating Bcl-2 expression, and increasing the p-AKT/AKT ratio. Conclusion: Our findings demonstrated that TA exerts its neuroprotective effect at lower concentrations, but induces apoptosis in SH-SY5Y cells at high concentrations.
Article
Full-text available
Emergency or urgent surgery, which can be defined as surgery which must be done promptly to save life, limb, or functional capacity, is associated with a high risk of bleeding and death. Antifibrinolytic agents, such as tranexamic acid, inhibit blood clot breakdown (fibrinolysis) and can reduce perioperative bleeding. Tranexamic acid has been shown to reduce the need for a blood transfusion in adult patients undergoing elective surgery but its effects in patients undergoing emergency or urgent surgery is unclear. OBJECTIVES To assess the effects of tranexamic acid on mortality, blood transfusion and thromboembolic events in adults undergoing emergency or urgent surgery. SEARCH METHODS We searched the following electronic databases: the Cochrane Injuries Group's Specialised Register (22 August 2012); Cochrane Central Register of Controlled Trials (2012, issue 8 of 12); MEDLINE (Ovid SP) 1950 to August Week 2, 2012; PubMed 1 June 2012 to 22 August 2012; EMBASE (Ovid SP) 1980 to 2012 Week 33; ISI Web of Science: Conference Proceedings Citation Index-Science (CPCI-S) 1990 to 22 August 2012; ISI Web of Science: Science Citation Index Expanded (SCI-EXPANDED) 1970 to 22 August 2012. We also searched online trial registers on 22 August 2012 to identify unpublished studies. SELECTION CRITERIA Randomised controlled trials comparing tranexamic acid with no tranexamic acid or placebo in adults undergoing emergency or urgent surgery. DATA COLLECTION AND ANALYSIS Two authors examined titles, abstracts and keywords of citations from the electronic databases for eligibility and extracted data for analysis and risk of bias assessment. Outcome measures of interest were mortality, receipt of a blood transfusion, units of blood transfused, reoperation, seizures and thromboembolic events (myocardial infarction, stroke, deep vein thrombosis and pulmonary embolism). MAIN RESULTS We identified five trials involving 372 people that met the inclusion criteria. Three trials (260 patients) contributed data to the analyses. The effect of tranexamic acid on mortality (RR 1.01; 95% CI 0.14 to 7.3) is uncertain. However, tranexamic acid reduces the probability of receiving a blood transfusion by 30% although the estimate is imprecise (RR 0.70; 95% CI 0.52 to 0.94). The effect on deep venous thrombosis (RR 2.29; 95% CI 0.68 to 7.66), and stroke (RR 2.79; 95% CI 0.12 to 67.10) is uncertain. There were no events of pulmonary embolism or myocardial infarction. None of the trials reported units of blood transfused, reoperation, or seizure outcomes. AUTHORS' CONCLUSIONS There is evidence that tranexamic acid reduces blood transfusion in patients undergoing emergency or urgent surgery. There is a need for a large pragmatic clinical trial to assess the effects of routine use of tranexamic acid on mortality in a heterogeneous group of urgent and emergency surgical patients.
Article
Obstetric hemorrhage accounts for ∼25% of maternal morbidity. Many methodologies are used to promote active management of the third stage of labor. Tranexamic acid successfully reduces blood loss and transfusion requirements in nonobstetric conditions. This double-blind, randomized, controlled trial was undertaken to estimate the effects of adding tranexamic acid to an active management protocol. The parturients were at 34 to 42 weeks’ gestation, with a live fetus in cephalic presentation, and expecting vaginal birth. The experimental group (n = 228) received 1 g/10 mL tranexamic acid diluted with 20 mL 5% glucose; the placebo group (n = 226) received 30mL5%glucose. Tranexamic acid or placebo was administered intravenously over 5 minutes at delivery of the fetus’ anterior shoulder. Standard management of the third and fourth stages of labor was applied. The primary outcome was the volume of blood loss during the third and fourth stages of labor. Other outcomes were the incidence of postpartum hemorrhage (PPH; >500 mL), severe PPH (>1000 mL), need for blood transfusion, need for additional uterotonic drugs, and adverse effects of tranexamic acid. Participants were contacted 3 weeks after delivery to assess the incidence of thromboembolic events. Data were analyzed on an intent-to-treat basis. The final analysis included 220 and 219 in the tranexamic acid and control groups, respectively. The groups were similar in patient characteristics and risk factors for PPH. At the third and fourth stages of labor, mean estimated blood losses were 261.5 ± 146.8 and 349.98 ± 188.85 mL in the tranexamic acid and control groups, respectively (P < 0.001). Four patients (1.8%) in the tranexamic acid group and 15 (6.8%) in the control group had PPH of greater than 500 mL (RR, 3.76; 95% CI, 1.27–11.15; P = 0.01). Six (2.7%) and 19 (8.7%) women in the tranexamic acid and control groups, respectively, required additional uterotonic agents (RR 3.18; 95% CI, 1.29–7.81; P = 0.007). The groups did not differ in need for blood transfusion. Predelivery hemoglobin and hematocrit levels were the same in the 2 groups, but at 1 day postpartum, hemoglobin levels were 9.9 T 1.4 and 9.3 T 0.9 g/dL (P G 0.001), and hematocrit levels were 30.2% ± 1.2% and 29.0% ± 1.3% (P < 0.001) in the tranexamic acid and placebo groups, respectively. No major complications and no significant changes in prothrombin time, active prothrombin time, or liver or renal function tests were noted in either group. Rates for nausea, vomiting, and diarrhea were 15.0%, 13.6%, and 7.3% in the tranexamic acid group and 5.5%, 6.4%, and 1.8% in the control group. At the clinical checkup 3 weeks postpartum, no episode of thrombosis was reported in patients given tranexamic acid. Tranexamic acid can reduce the average amount of blood loss during the third and fourth stages of labor. Determining the clinical settings in which this medication may be beneficial is critical for future research.
Article
Concerns regarding the safety of transfused blood have led to the development of a range of interventions to minimise blood loss during major surgery. Anti-fibrinolytic drugs are widely used, particularly in cardiac surgery, and previous reviews have found them to be effective in reducing blood loss, the need for transfusion, and the need for re-operation due to continued or recurrent bleeding. In the last few years questions have been raised regarding the comparative performance of the drugs. The safety of the most popular agent, aprotinin, has been challenged, and it was withdrawn from world markets in May 2008 because of concerns that it increased the risk of cardiovascular complications and death. OBJECTIVES: To assess the comparative effects of the anti-fibrinolytic drugs aprotinin, tranexamic acid (TXA), and epsilon aminocaproic acid (EACA) on blood loss during surgery, the need for red blood cell (RBC) transfusion, and adverse events, particularly vascular occlusion, renal dysfunction, and death. SEARCH STRATEGY: We searched: the Cochrane Injuries Group's Specialised Register (July 2010), Cochrane Central Register of Controlled Trials (The Cochrane Library 2010, Issue 3), MEDLINE (Ovid SP) 1950 to July 2010, EMBASE (Ovid SP) 1980 to July 2010. References in identified trials and review articles were checked and trial authors were contacted to identify any additional studies. The searches were last updated in July 2010. SELECTION CRITERIA: Randomised controlled trials (RCTs) of anti-fibrinolytic drugs in adults scheduled for non-urgent surgery. Eligible trials compared anti-fibrinolytic drugs with placebo (or no treatment), or with each other. DATA COLLECTION AND ANALYSIS: Two authors independently assessed trial quality and extracted data. MAIN RESULTS: This review summarises data from 252 RCTs that recruited over 25,000 participants. Data from the head-to-head trials suggest an advantage of aprotinin over the lysine analogues TXA and EACA in terms of reducing perioperative blood loss, but the differences were small. Compared to control, aprotinin reduced the probability of requiring RBC transfusion by a relative 34% (relative risk [RR] 0.66, 95% confidence interval [CI] 0.60 to 0.72). The RR for RBC transfusion with TXA was 0.61 (95% CI 0.53 to 0.70) and was 0.81 (95% CI 0.67 to 0.99) with EACA. When the pooled estimates from the head-to-head trials of the two lysine analogues were combined and compared to aprotinin alone, aprotinin appeared more effective in reducing the need for RBC transfusion (RR 0.90; 95% CI 0.81 to 0.99).Aprotinin reduced the need for re-operation due to bleeding by a relative 54% (RR 0.46, 95% CI 0.34 to 0.62). This translates into an absolute risk reduction of 2% and a number needed-to-treat (NNT) of 50 (95% CI 33 to 100). A similar trend was seen with EACA (RR 0.32, 95% CI 0.11 to 0.99) but not TXA (RR 0.80, 95% CI 0.55 to 1.17). The blood transfusion data were heterogeneous and funnel plots indicate that trials of aprotinin and the lysine analogues may be subject to publication bias.When compared with no treatment aprotinin did not increase the risk of myocardial infarction (RR 0.87, 95% CI 0.69 to 1.11), stroke (RR 0.82, 95% CI 0.44 to 1.52), renal dysfunction (RR 1.10, 95% CI 0.79 to 1.54) or overall mortality (RR 0.81, 95% CI 0.63 to 1.06). Similar trends were seen with the lysine analogues, but data were sparse. These data conflict with the results of recently published non-randomised studies, which found increased risk of cardiovascular complications and death with aprotinin. There are concerns about the adequacy of reporting of uncommon events in the small clinical trials included in this review.When aprotinin was compared directly with either, or both, of the two lysine analogues it resulted in a significant increase in the risk of death (RR 1.39, 95% CI 1.02, 1.89), and a non-significant increase in the risk of myocardial infarction (RR 1.11 95% CI 0.82, 1.50). Most of the data contributing to this added risk came from a single study - the BART trial (2008). AUTHORS' CONCLUSIONS: Anti-fibrinolytic drugs provide worthwhile reductions in blood loss and the receipt of allogeneic red cell transfusion. Aprotinin appears to be slightly more effective than the lysine analogues in reducing blood loss and the receipt of blood transfusion. However, head to head comparisons show a lower risk of death with lysine analogues when compared with aprotinin. The lysine analogues are effective in reducing blood loss during and after surgery, and appear to be free of serious adverse effects.