Tranexamic Acid in Neurosurgery

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Neurosurgical Review

https://doi.org/10.1007/s10143-020-01324-0

REVIEW

Tranexamic acid in Neurosurgery: a controversy indication—review


José Luiz de Faria 1 & Josué da Silva Brito 1 & Louise Teixeira Costa e Silva 1 &
Christiano Tadeu Sanches Mattos Kilesse 1 & Nicolli Bellotti de Souza 1 & Carlos Umberto Pereira 2 &
Eberval Gadelha Figueiredo 3 & Nícollas Nunes Rabelo 1,3

Received: 13 January 2020 / Revised: 1 May 2020 / Accepted: 20 May 2020


# Springer-Verlag GmbH Germany, part of Springer Nature 2020

Abstract
Tranexamic acid (TXA) is one of the measures indicated to reduce bleeding and the need for volume replacement. However, data
on risks and benefits are controversial. This study analyzes the effectivity and risks of using tranexamic acid in neurosurgery. We
selected articles, published from 1976 to 2019, on the PubMed, EMBASE, Science Direct, and The Cochrane Database using the
descriptors: “tranexamic acid,” “neurosurgery,” “traumatic brain injury,” “subdural hemorrhage,” “brain aneurysm,” and “sub-
arachnoid hemorrhage.” TXA can reduce blood loss and the need for blood transfusion in trauma and spinal surgery. Despite the
benefits of TXA, moderate-to-high doses are potentially associated with neurological complications (seizures, transient ischemic
attack, delirium) in adults and children. In a ruptured intracranial aneurysm, the use of TXA can considerably reduce the risk of
rebleeding, but there is weak evidence regarding its influence on mortality reduction. The TXA use in brain surgery does not
present benefit. However, this conclusion is limited because there are few studies. TXA in neurosurgeries is a promising method
for the maintenance of hemostasis in affected patients, mainly in traumatic brain injury and spinal surgery; nevertheless, there is
lack of evidence in brain and vascular surgeries. Many questions remain unanswered, such as how to determine the dosage that
triggers the onset of associated complications, or how to adjust the dose for chronic kidney disease patients.

Keywords Tranexamic acid . Antifibrinolytic . Neurosurgery . Blood loss surgical . Brain injuries

Introduction patient’s head. Throughout the twentieth century, Cushing’s lat-


eral ventricular puncture, bandages, clips, hydrogen peroxide,
Neurosurgical procedures cause significant blood loss; thus, caustic agents, adrenaline, thrombin, and fibrin were used [6].
demanding measures to restore volumes, such as crystalloids Tranexamic acid (TXA), developed in the second half of
and blood products, are necessary. However, the use of these the twentieth century, is one of the alternatives for reducing
substances can result in hypokalaemia, anaphylactic reactions, blood loss and the need for volume replacement. However,
pulmonary trauma, infection, hemolytic reactions, cardiovas- data are controversial and there is uncertainty regarding the
cular overload, and sepsis postoperative [1–5]. recommended dosage, and associated risks [6–8].
Modern neurosurgery has already deployed different methods This study seeks to review the literature by analyzing the
to control bleeding. Chemical, thermal, and mechanical methods, effectivity and risks of deploying TXA in neurosurgery and trau-
and clinical measures were used, such as the elevation of the matic brain injury.

* Nícollas Nunes Rabelo


nicollasrabelo@hotmail.com
Methodology
1
Department of Neurosurgery, University Center UNiAtenas, We selected articles on the PubMed platform, EMBASE,
Paracatu, Minas Gerais, Brazil Science Direct, and The Cochrane Database using the terms:
2
Department of Neurosurgery of FBHC and Neurosurgery Service, “tranexamic acid,” “neurosurgery,” “traumatic brain injury,”
Aracaju, Sergipe, Brazil “subdural hemorrhage,” “brain aneurysm,” and “subarach-
3
Department of Neurosurgery, Hospital das Clinicas da Faculdade de noid hemorrhage” using the Boolean operator “OR” and
Medicina, University of Sao Paulo, Sao Paulo, Brazil “AND.” The search carried out by all the authors considered
Neurosurg Rev

articles published from 1976 to 2019 and resulted in 487 arti- GABA receptors in cortical and medullary neurons. Both inhib-
cles, 245 of which had their abstracts read. Meta-analysis, itory pathways of TXA cause an increased excitatory synaptic
randomized controlled trials, and cohort studies were includ- stimulus, as evidenced by similar to convulsion-like events in-
ed. We excluded papers that were not in English, Spanish, or duced at TXA concentrations of 31 μg mL−1 (200 μM). These
Portuguese, as well as brief notes and case reports, articles not events are similar to those measured in the cerebrospinal fluid
related to the topic, or not available in full. After applying the (CSF) of patients undergoing extracorporeal circulation [2, 9, 10]
exclusion criteria, there were 57 articles left (Fig. 1). TXA crosses the blood-brain barrier and enters the eye,
reaching around 10% of the plasma concentration in the
CSF. This, in turn, can lead to changes in color vision, espe-
Results and discussion cially in patients who use the drug chronically. In conscious
patients, gastrointestinal side effects (nausea, vomiting, and
Patented by S. Okamoto in 1957 [9], TXA (trans-4- diarrhea) are more commonly reported [7, 9, 11]. The half-
aminomethyl-cyclohexane-1-carboxylic acid) is a synthetic life of TXA is approximately 80 min in patients with pre-
analog of lysine, which, by competition, inhibits the activation served renal function and can be administered orally, intrave-
of plasminogen into plasmin. At high concentrations, TXA nously, intramuscularly, or topically [4, 7, 12].
blocks non-competitive plasmin, inhibiting the dissolution Despite the substantial discussion, there is yet no consensus
and degradation of fibrin clots. on either the minimum dose at which the drug generates ben-
Approximately 95% of TXA is excreted unchanged in the efits or the maximum concentration at which complications
urine, and excretion decreases with increasing plasma creatinine occur. TXA has been successfully administered in a number
levels. Dose-adjustment in patients with renal impairment re- of surgical procedures, such as orthopedic and cardiac surger-
mains an unknown factor. Recently, TXA was shown to be ies, solid organ transplants, and gynecological and obstetric
structurally similar to glycine, inhibiting competitively glycine interventions [3, 7, 13, 14]. TXA has also been successfully
receptors in cortical and spinal cord neurons in rats, as well as used in non-surgical procedures with a high risk of bleeding in

Fig. 1 Representation of identification, screening, eligibility, and inclusion and exclusion criteria in the review
Neurosurg Rev

order to reduce massive intraoperative blood loss and thus also 2.9 in the TXA group and 15.5 ± 3.0 in the control group).
decrease the need for blood transfusion [1, 4, 9, 13, 15]. They used a loading dose of 1 g, followed by an immediate
maintenance dose of 100 mg/h until the closure of the surgical
Spine surgery incision. There was a significant reduction in intraoperative
blood loss in the TXA-treated group (613 ± 195 mL) in com-
Neurosurgery, especially spinal surgery, often involves signifi- parison with the control group (1079 ± 421 mL; P < 0.001),
cant blood loss, thereby requiring blood transfusions. lower postoperative blood loss (155 ± 86 mL versus 263 ± 105
Transfusion by-products can be detrimental to the patient due mL, respectively; P < 0.001), and decreased need for blood
to the risk of hemolytic and non-hemolytic reactions; infection; transfusion during the procedure (258 ± 246 mL versus 377 ±
and the dilution of plasma components, such as coagulation fac- 200 mL, respectively; P < 0.001). No major complications
tors, by the replacement of red blood cells and crystalloids [1–3, were observed during the study [21].
16]. In this regard, the use of antifibrinolytics may represent a In a meta-analysis [8] comprising 11 randomized controlled
valuable strategy to control perioperative hemorrhage. trials (RCT) with a total of 644 patients, TXA successfully re-
Studies (Table 1) on the application of TXA in spinal sur- duced intraoperative, postoperative, and total blood loss in spinal
gery point the decision on the moment of application and the surgeries. The major reduction was intraoperative blood loss by
dose that should be used as a first challenge. In most of the an average of 219 mL, ranging from 116 to 322 mL compared
analyzed, authors preferred to administer a dose ranging from with placebo (P < 0.05). The meta-analysis also concluded that
10 mg/kg (preferably in most studies) to 30 mg/kg immedi- the use of TXA leads to a reduction in the volume of blood
ately upon performing an incision, a maintenance dose of 0.5 transfusions (risk ratio—RR—0.67 [0.54, 0.83], P < 0.05).
to 2 mg/kg/h, followed by a preferable 1-mg/kg/h dose until There was no association between thromboembolic events and
the end of the surgical procedure, obtaining satisfactory re- TXA. Zhang et al. [16] drew similar conclusions in another meta-
sults. High TXA doses did not necessarily increase rates of analysis, in addition to emphasizing the need for more accurate
thromboembolism or convulsions in the case of ASA I and studies to evaluate the risk of intravenous use of TXA. The TXA
ASA II patients, with no risk factors for thromboembolism or use resulted lower risk of blood transfusion (odds ratio—OR—
significant renal changes [4, 8, 9, 13, 15–19]. 0.56 [0.36, 0.87], P = 0.01) and was not associated with throm-
Most studies have pointed out the beneficial potential of botic complications (OR 0.99 [0.06, 16.07], P = 0.99)
TXA, including reduction in bleeding in the intraoperative and In the studies, there was no significant difference between
postoperative periods, as well as the need for blood transfusion the TXA-treated group and the control group regarding com-
[4, 8, 9, 13, 15–19]. There is increasing evidence showing TXA plications, thereby demonstrating the safety of TXA at differ-
can reduce intraoperative bleeding in spinal column surgery by ent dosages [4, 8, 9, 13, 15–19].
up to 49% (P < 0.007) [2, 7, 17]. Patients receiving intraoperative The studies were used for the adoption or use of tranexamic
TXA treatment also showed a decreased tendency to remain acid in spinal surgery. Its blood loss reduction benefit is evi-
hospitalized after surgery [7, 15, 17], although only a few dent, although it is not possible to determine whether it is
showed significant results [4, 15, 20]. In general, the studies were greater before, during, or after the surgical procedure. Its po-
limited due to the small sample number. Therefore, interpretation tential for mortality and hospital stay is not clear. However,
for the general population must be done carefully. care should be taken with regard to the dosage adopted, with
Elwatidy et al. [7] obtained the most positive results. In low doses being preferable, since they have shown good re-
their study, high doses of TXA reduced total blood transfu- sults in recent studies. There is still no consensus on the pa-
sions by 80% and total blood loss by 49% (P < 0.007) in tient who can use it. Therefore, general care should be taken
laminectomy surgeries with posterior spinal fixation, as well with elderly patients and patients with chronic renal failure or
as laminectomy and the excision of spinal tumors. The blood decompensation. For them, the real impacts are not very clear
loss during surgery in the TXA group was 311.25 ± 412.49 due to their non-inclusion in the studies or the small presence.
mL versus 584.64 ± 797.30 mL in the placebo group (P = Therefore, an assessment of risks and benefits is necessary,
0.03). Nevertheless, the study included only 64 patients. according to the practice of each neurosurgeon.
Tsutsumimoto et al [18]. also reported that postoperative
blood loss in the first 16 h following surgery was significantly Brain surgery
lower in the TXA group than in the control group (P = 0.001).
The postoperative blood loss was 37% lower in the TXA The use of TXA in brain surgeries was analyzed in some RCT
group in comparison with control group. and retrospective cohort studies (Table 2). The studies
A retrospective study performed in 2012 included 106 pa- adopted a variable dose of TXA. The initial dose ranged from
tients (43 in the TXA group and 63 the in control group) and 10 to 25 mg/kg and the maintenance from 1 to 10 mg/kg/h.
evaluated the use of TXA in posterior spinal fusion for treat- Despite the different doses, the results were similar and were
ment of adolescent idiopathic scoliosis (age in years, 15.2 ± not associated with complications [1, 2, 24].
Neurosurg Rev

Table 1 Key results of studies on TXA effects in spinal surgery

Study Type Number of patients Type of surgery Key results


of (TXA/P) and
study intervention

Seddighi [4] RCT 20/20 Major spine surgery The intraoperative blood loss (P = 0.139) and blood loss of the
L, 10 mg/kg 1st postoperative day (P = 0.067) did not show significant
M, 0.5 mg/kg/h differences, but a lower mean in the TXA group.
P: saline The mean of blood transfusion did not show a statistically
significant difference (P = 0.86).
The total blood loss in the TXA group was 669.5 mL versus
912.5 mL in the control group.
There were no complications.
Elwatidy [7] RCT 32/32 Laminectomy with posterior spinal Use of tranexamic acid in spine surgery is safe and effective.
Adults: fixation, and laminectomy and It reduced blood loss by 49% (P < 0.007) and need for blood
I, 2 g excision of spinal tumor transfusion by 80% (P < 0.008).
M, 10 0 mg/h There were no complications.
P: saline
Children:
I, 30 mg/kg
M,1 mg/kg/h
P: saline
Cheriyan [8] MA 325/319 All spinal surgery TXA reduced intraoperative, postoperative, and total blood loss.
I, 10 mg/kg to 2 g There was one case of myocardial infarction in the TXA group
M, 1–2 mg/kg/h not associated to TXA treatment.
P: saline
Multiple
interventions
Carabini [13] RCT 31/30 Multilevel spine fusion surgery Reduced total volume of RBC transfusion (1140 mL in the TXA
I, 10 mg/kg group versus 1460 mL in the control group, P = 0.034)
M, 1 mg/kg/h Reduced the cell saver transfusion (P = 0.042)
P: placebo There were no complications.
Shi [15] RCT 50/46 Lumbar spinal stenosis and lumbar Reduced perioperative blood loss (P < 0.005).
I, 30 mg/kg spondylolisthesis Total blood loss was reduced by 41% (P < 0.001).
M, 2 mg/kg/h The mean of blood transfusion did not show a statistically
P: saline significant difference (P = 0.191).
There were no complications in the TXA group.
Zhang [16] MA 208/203 Spinal surgery Reduced the amount of blood loss, the volume of blood
I, 10–100 mg/kg/h transfusion, and the transfusion rate
M, 1–10 mg/kg/h The TXA use resulted in 44% decline of risk of blood
P: saline transfusion compared with the placebo group with an OR of
Multiple 0.56 (95% confidence interval—CI—0.36 to 0.87)
interventions Further studies are needed.
Shakeri [17] RCT 25/25 Laminectomy and posterior spinal Reduced intraoperative blood loss (P = 0.0001)
I, 15 mg/kg fusion Total blood loss in the TXA group was 632.2 ± 193.1 versus
M: none 1037 ± 242.6 in the control group.
P: saline Reduced hospitalization time (P = 0.001)
Reduced need for transfusion (P = 0.001)
There were no complications.
Tsutsumimoto RCT 20/20 “French-door” cervical laminoplasty Perioperative blood loss, especially postoperative blood loss
[18] I, 15 mg/kg from C3 to C6 during the first 16 h, in the TXA group was significantly
M: none lower than that in the control group (P < 0.01).
P: saline The postoperative blood loss was reduced by 37% (132.0 ± 45.3
in the TXA group versus 211.0 ± 41.5 mL in the control
group).
There were no complications.
Raksakietisak RCT 39/39 Complex spine surgery Blood loss in the placebo group was higher than that in the TXA
[19] I, 15 mg/kg group (P = 0.014)
After 3 h, 15 mg/kg Postoperative period had lower blood loss (P = 0.014)
P: saline Reduced blood transfusion by 64.6%
Colomina [20] RCT 44/51 Major spine surgery Significant reduction in intraoperative blood loss (P = 0.01)
I, 10 mg/kg The mean of blood transfusion did not show statistically
M, 2 mg/kg/h significant difference (P = 0.06).
Neurosurg Rev

Table 1 (continued)

Study Type Number of patients Type of surgery Key results


of (TXA/P) and
study intervention

P: saline In the TXA group: one case of deep vein thrombosis and
pulmonary embolism, with no significant differences
between the TXA group and the control group.
Yagi [21] RS 43/63 Posterior spinal fusion for treatment of Intraoperative blood loss, mean of blood transfusion, and
I, 1 g adolescent idiopathic scoliosis postoperative blood loss were significantly reduced (P <
M, 100 mg/h 0.01) in the TXA-treated group.
P: saline TXA was not associated with major complications.
Xue [22] RS 20/22 Posterior laminectomy and Reduced the need and amount of blood transfusion
I, 15 mg/kg posterolateral bone graft fusion Can shorten the extubating time and shorten the length of
M, 1 mg/kg/h postoperative hospital stay without increasing the incidence
P: no intervention of postoperative coagulation dysfunction or postoperative
deep venous thrombosis
Lower intraoperative blood transfusion in the TXA group
963.64 ± 341.63 mL than 1680 ± 112.14 mL in the control
group
Lower postoperative hospital stay: 4.23 ± 1.02 days in the TXA
group versus 5.55 ± 1.28 in the control group
Damade [23] RS 36/47 Decompressive surgery associated No significant difference was found in intraoperative and
I, 15 mg/kg with bone fixation postoperative blood loss.
M, variable Significant reduction in postoperative bleeding was found in the
P: no intervention TXA group.
Multiple The volume of transfused blood was significantly reduced (P <
0.05) in the TXA-treated group.

RCT, randomized controlled trial; MA, meta-analysis; RS, retrospective study; I, infusion dose; M, maintenance dose; P, placebo; RBC, red blood cells

Hooda et al. [2] used TXA in excision of intracranial me- These studies are insufficient for conclusions, because
ningioma, with N = 60 (30 in each group) with the following these are different procedures with a limited sample. Further
results: significant reduction in heart rate, increase in mean studies are needed. The TXA use may be negligible in brain
arterial pressure and plasminogen levels, intraoperative bleed- surgery.
ing reduction by 27% (RR 1.3 [1.1, 1.8], P = 0.03), and thus
improvement of hemodynamic maintenance accompanied by Ruptured intracranial aneurysms
a tendency to reduce the number of blood transfusions, but
without significant alteration (P = 0.89). Risk of rebleeding is a major complication in rupture of intra-
In a trial evaluating the use of TXA in elective craniotomy for cranial aneurysms occurring in 10 to 22% of affected patients.
the excision of supratentorial tumors, Vel et al. [1] obtained Its highest incidence occurs in the first 24 h, peaking in the
similar results as those described above, with a significant reduc- first 6 h after the event [29, 30]. The prognosis for patients
tion in intraoperative bleeding (P = 0.012) yet without a corre- experiencing rebleeding after the rupture of intracranial aneu-
sponding decrease in the need for blood transfusions (P = 0.109). rysms is quite poor, with 60% of these patients dying and
Mebel et al. [14] concluded that the use of TXA is safe, another 30% turning unable to perform daily activities, there-
with no significant differences in the rate of thromboembolic by requiring care [29, 31–33].
events as compared with the control group. A retrospective The use of TXA can considerably reduce the risk of
cohort study performed with 519 patients (245 of whom re- rebleeding, but there is weak evidence for a decrease in the
ceived TXA) who had undergone complex neurosurgical pro- overall number of deaths [31, 34–37]. In a Cochrane review
cedures showed significantly lower transfusion rates (P = published in 2013 comprising 10 randomized controlled trials
0.04) in the TXA-treated group (13% versus 7% in the control regarding the effects of antifibrinolytics agents on hemorrhage
group). (RR 0.65, 95% CI 0.44 to 0.97, 78 per 1000 participants), the
Thromboelastography is one option for optimizing the se- overall mortality rate was unaffected (RR 1.00, 95% CI 0.85
lection of patients who will benefit from TXA, since this to 1.18) [31].
method allows for the rapid evaluation of hypofibrinogenemia In a meta-analysis performed with 2872 individuals, 1380
[26–28]. patients received antifibrinolytics agents for the management
Neurosurg Rev

Table 2 Key points of studies on brain surgery

Study Type Number of patients Type of surgery Key points


of (TXA/P) and
study Intervention

Vel [1] RCT 50/50 Elective craniotomy for Saline group at 4, 6, and 8 h (P < 0.001)
I, 10 mg/kg excision of the Significant reduction in blood loss in the TXA group compared with the
M, 1 mg/kg/h supratentorial tumor saline group at 2, 4, and 6 h of surgery (P < 0.05)
P: saline There was no reduction in intraoperative blood transfusion requirements.
The mean total blood loss in the TXA group was 817.00 ± 423.3 mL and
1084.00 ± 604.8 mL in the saline group (P = 0.012)
There were no complications.
Hooda RCT 30/30 Excision of intracranial The amount of blood loss was significantly less in the tranexamic acid
[2] I, 20 mg/kg/ meningioma group compared with the placebo group (P = 0.03).
M,1 mg/kg/h The trend was towards lesser transfusion requirement in the tranexamic acid
P: saline group, even though it was not statistically significant.
There was no significant difference between the TXA group and the control
group, regarding complications.
Mebel RC 245/275 Complex skull base The rate of perioperative transfusion in patients who received tranexamic
[14] I, 10–25 mg/kg neurosurgical procedures acid was lower (7% versus 13%, P = 0.04).
M, 5–10 mg/kg/h There was no significant difference between the TXA group and the control
P: saline group, regarding complications.
Dadure RCT 19/21 Craniosynostosis surgery Volume of packed erythrocytes transfused was significantly reduced by
2011 I, 15 mg/kg 85% intraoperatively and by 57% throughout the study period (P < 0.05).
[24] M, 10 mg/kg/h Requiring blood transfusion was lower in the TXA group during surgery
45% versus 11% (P < 0.05) and during the whole study period 70%
versus 37% (P < 0.05).
Kurnik RC 23/30 Craniosynostosis surgery A 4-h postoperative infusion of TXA after open calvarial vault remodeling
[25] I, 10 mg/kg for craniosynostosis was as effective in reducing blood loss and trans-
M, 5 mg/mg/h per 24 fusion requirements as the 24-h infusion.
or 4 h

RCT, randomized controlled trial; RC, retrospective cohort; I, infusion dose; M, maintenance dose; P, placebo

of subarachnoid hemorrhage (SAH). It showed that TXA until aneurysm initiation or, at most, for 24 h after the initia-
treatment may be beneficial for diminishing rebleeding rate tion of medication. This paper will be important for assessing
in the short term, i.e., less than 3 days, which can be associated the efficacy and safety of early TXA use in SAH patients.
with calcium channel blockers and hypervolemic therapies. The use of TXA in a ruptured intracranial aneurysm was
Furthermore, TXA treatment did not increase the risk of cere- shown to be safe since it was not associated with worse out-
bral ischemic events, thereby constituting a potential strategy comes or thromboembolic events, according to more recent
to prevent poor functional outcomes [32]. studies on the topic [30, 32, 37]. However, the combination
Hillman et al. [37] analyzed the use of TXA in patients with chlorpromazine can lead to worse outcomes [34]. Studies
immediately after the diagnosis of SAH within 48 h before performed before 2001 using high TXA doses often reported a
first hospitalization. A 1-g dose of TXA was administered higher risk of thromboembolic complications [38, 39].
intravenously as soon as SAH was diagnosed at local hospitals Despite the studies presented conflicting conclusions and
(before patient transport), followed by 1 g every 6 h until the different protocols, the potential of TXA in reducing mortality
aneurysm was occluded, not exceeding 72 h of treatment. The and rebleeding justifies its adoption as a protocol in hemor-
results showed a significant reduction in rebleeding rate, rang- rhage stroke. Risks can be minimized by adopting short-term
ing from 10.8 to 2.4%, and reduced mortality rate due to intervention. More studies on the subject are needed in order
rebleeding by 80% upon TXA treatment, with no increase in to establish unified guidelines (Table 3).
drug-related ischemic and vasospasm events.
ULTRA [29] is a prospective, randomized, open-label,
multicenter study, which included 950 patients that evaluated Traumatic brain injury
the effect of TXA on non-traumatic SAH adults 24 h after the
onset of headache. Patients in the TXA-treated group received Traumatic brain injury (TBI) in Western countries is the lead-
a TXA bolus dose (1 g intravenously) immediately after ran- ing cause of death among 15- to 29-year olds and is the third
domization, followed by a continuous 1-g infusion for 8 h leading cause of death among all age groups [42].
Neurosurg Rev

Table 3 Key points of studies on ruptured intracranial aneurysms

Study Type of Number of patients (TXA/P) and Key points


study intervention

Chandra RCT 20/19 Rebleeding and mortality were reduced by one-fourth and one-fifth, respectively (P
[40] 6 g per day, given 1 g every 4 h < 0.01).
intravenously for 3 weeks No side effects of tranexamic acid therapy were seen, such as pulmonary embolus
or renal artery thrombosis.
Fodstad RCT 30/29 TXA protects patients with ruptured aneurysms from rebleeding for 1 or 2 weeks.
[34] 6 g per day in the first week; Increased incidence of severe vasospasm, fatal delayed cerebral ischemia,
4 g per day in the second week; pulmonary embolism, and myocardial infarction among the tranexamic
6 g per day, per orally, in the third to sixth acid–treated patients in combination with chlorpromazine.
week
Fodstad RCT 21/20 TXA reduces rebleeding rate. TXA can increase the occurrence of cerebral
[38] 6 g per day in the first week; ischemia.
4 g per day in the second to fifth week
Vermeulen RCT 241/238 Reduces rebleeding from 24 to 9% in the TXA group (P < 0.001)
[39] 6 g per day in the first week Reduces mortality in 2.5%
4 g/day following 3 weeks Increases incidence of ischemic complications, 15% in the control group and 24%
Centers with oral administration used 6 in the TXA group (P < 0.001)
g/day on the third and fourth weeks
Wijdicks NRCT 119/238 Rate of rebleeding (24 of 119, 20%) was close with that in the placebo (56 of 238,
[35] 6 g per day for 4 weeks 24%).
Rate of cerebral infarction (33 of 119, 28%) was almost identical to that after
long-term tranexamic acid (59 of 241, 24%)
Treatment with tranexamic acid for 4 days fails to reduce the incidence of
rebleeding but still increases the rate of cerebral infarction.
Roos [41] RCT 229/233 Reduced the occurrence of rebleeding (RR, 0.58; 95% CL, 0.42–0.80)
6 g/day in the first week The occurrence of ischemic complications was the same in the two groups
6 g/day per orally in the second and third TXA combined with treatment to prevent cerebral ischemia does not improve
weeks outcome.
Hillman RCT 254/251 Reduction in the rebleeding rate from 10.8 to 2.4% and an 80%
[37] 1 g immediately on diagnosis Reduction in the mortality rate from early rebleeding
1 g every 6 h until the aneurysm was There was no predisposition to thromboembolic events.
occluded, not exceeding 72 h
Gaberel MA 1380/1492 Short-term use diminishes rebleeding rate.
[32] Multiple There was no predisposition to thromboembolic events.
Post [30] RCT 119/390 Did not reduce rebleeding rate in the TXA group
1 g immediately on diagnosis Mortality was significantly lower in the TXA group than that in the standard care
1 g every 8 h until the aneurysm was group.
occluded, not exceeding 36 h There was no predisposition to thromboembolic events.

RCT, randomized controlled trial; MA, meta-analysis; NRCT, non-randomized, placebo-controlled clinical trial

The CRASH-2 trial [43] was the first multicenter random- TXA reduced overall mortality and bleeding compared with
ized controlled study to show efficacy and safety of TXA on patients treated after 3 h of trauma, which led to increased
TBI patients. This study had N = 20,211 (10,096 in the TXA mortality due to bleeding. This suggests the use of
group and 10,115 in the placebo group) and demonstrated thromboelastography to discriminate those who need treat-
lower relative (RR = 0.85 [0.76, 0.96]) and all-cause mortality ment from those who do not.
(RR = 0.91 [0.85, 0.97]) after a loading dose of 1 g for 10 min, In 2019, RCT analyzed the early use of TXA in patients
followed by a maintenance dose of 1 g for 8 h in adult trauma with subdural and epidural hemorrhage after TBI. Mean intra-
patients with or at risk of significant bleeding within 8 h of operative bleeding during surgery was significantly reduced in
injury. However, the author concluded that treatment beyond the TXA group (P = 0.012) when compared with the placebo
3 h of injury is unlikely to be effective. group; however, no statistical significance was found in the
Analyzing the data obtained from the CRASH-2 study, control of subdural and epidural hemorrhage [45].
Roberts et al. [44] concluded that TXA reduced mortality Recent results from a meta-analysis showed that early treat-
from bleeding in polytrauma patients, although this effect ment with TXA is more effective in reducing bruising. TBI
seemed to be related to the time at which drug infusion was patients treated with TXA exhibited reduced risk of hematoma
initiated. In the group of patients treated within 3 h of trauma, expansion, as well as favorable neurological outcomes due to
Neurosurg Rev

lower mortality rate, without affecting the rate of thrombosis The CRASH-3 trial [48] showed that treatment within 3 h
events [46]. of the mild-to-moderate head injurie leads to a substantial
The 2016 European guidelines (4th edition) on the reduction in mortality in patients with TBI (RR 0.78 [0.64–
management of major bleeding and coagulopathy fol- 0.95]). It included adults presenting a score of 12 or lower on
lowing trauma [47] strongly recommends (grade 1A) the Glasgow Coma Scale, any intracranial bleeding on com-
the early use of TXA in trauma patients who are bleed- puted tomography, and no major bleeding receiving a loading
ing or at risk of significant bleeding. The guidelines dose of 1 g for 10 min, followed by a maintenance dose of 1 g
further recommend the use of TXA within the first for 8 h. Thus, early treatment was shown to be more effective
3 h after the trauma has occurred (grade 1B), with ad- than later treatment. In addition, there were no benefits of
ministration of the first dose preferably in route to the TXA in terms of severe head injury (RR 0.99 [0.91–1.07]; P
hospital (grade 2C). value for heterogeneity 0.030).

Table 4 Key points of studies on traumatic brain injury

Study Type Participants Number of patients Key points


of (TXA/P) and
study intervention

CRASH-2 [43] RCT Adult trauma patients with, or at risk of, 10,096/10,115 TXA safely reduced the risk of death in bleeding trauma
significant bleeding who were within I, 1 mg patients.
8 h of injury M,1 mg over 8 h Treatment beyond 3 h of injury is unlikely to be effective.
P: saline Treatment after 3 h may increase the risk of death from
bleeding.
Roberts [44] RCT Crash-2 Trial similar patients 10,096/10,115 TXA treatment within the first 3 h of trauma reduced
I, 1 mg mortality.
M,1 mg over 8 h TXA treatment after 3 h of trauma increased bleeding
P: saline mortality.
Ebrahimi [45] RCT Patients with traumatic brain injury with 40/40 The TXA may reduce bleeding.
subdural and epidural hemorrhage I, 1 mg There was no statistical significance for subdural and
M,1 mg over 8 h subarachnoid hematoma reduction.
P: saline No complications were observed in any of the intervention
and control groups during the study as well.
Weng [46] MT Multiple patients with TBI 463/454 Significant effect on reducing the risk of hematoma
Multiple expansion.
Decreasing mortality rate
Improved favorable neurological outcomes
There was no increase in the rate of thrombotic events.
Chan [49] PS Patients with cerebral contusions or 81/570 Tranexamic acid was identified by univariate analysis to be
traumatic subarachnoid hemorrhage Multiple an independent factor associated with lower mortality.
Thromboembolic events were comparable in patients with
or without tranexamic acid.
El-Menyar [50] RS 102/102 Prehospital TXA administration is associated with less
Multiple in-hospital blood transfusion and massive transfusion
protocol.
There is no significant increase in the thromboembolic
events and mortality.
Chakroun-Walha PS All patients, aged at 18 years or older 96/84 The benefits of TXA in TBI without significant extracranial
[51] with TBI Multiple bleeding are to be considered.
The timing and indications of its use are still not defined
and the assessment of the outcome and the adverse
effects are still controversial.
Pulmonary embolism was statistically more frequent in
TXA group (11.5 versus 2.4%, P = 0.02).
CRASH-3 [48] RCT Adult trauma patients with, or at risk of, 6406/6331 Tranexamic acid is safe in patients with TBI treatment
significant bleeding who were within I, 1 mg within 3 h of injury reduces head injury-related death.
3 h of injury M,1 mg over 8 h Patients should be treated as soon as possible after injury.
P: saline Thromboembolic events were comparable in patients with
or without tranexamic acid.

RCT, randomized controlled trial; MA, meta-analysis; PS, prospective study; RS, retrospective study; I, infusion dose; M, maintenance dose; P, placebo
Neurosurg Rev

This study demonstrated that the effect of TXA ap- Risks and limitations of TXA use
pears to depend on the severity of TBI and the time
between injury and the initiation of treatment. Patients Moderate-to-high doses (100 mg/kg/bolus and 10 mg/kg/h for
with mild TBI (GCS score of 13–15 with intracranial example) of TXA is potentially associated with neurological
bleeding baseline on a CT scan) and patients with mod- complications (seizures, transient ischemic attack, delirium) in
erate TBI benefited from TXA use, especially in the first adults and children [11, 52, 53].
3 h after trauma with a considerable decrease in mortal- A meta-analysis comprising 11,388 individuals showed no
ity. Patients with a GCS score of three with bilateral and association with increased risk of myocardial disease and mor-
unilaterally unreacted pupils exhibited rapid expansion of tality, as well as weak association to neurological alterations.
intracranial hemorrhage and did not have bleeding reduc- However, there was a tendency to increase the risk of these
tion, which did not improve outcome. There was no in- complications, suggesting the need for a deeper analysis in-
creased risk of adverse events. In particular, the risk of cluding a larger number of patients [54]. Similarly, another
deep vein thrombosis, pulmonary embolism, stroke, and meta-analysis based on 69 papers showed no significant asso-
myocardial infarction in the TXA-treated group was low- ciation of TXA with an increased risk of thromboembolic
er when compared with that in the placebo group, dem- events [55].
onstrating the safety of TXA among these patients [48]. In cardiac surgeries, the use of TXA was correlated with
The TBI with its devastating results associated with the use higher mortality rates due to increased postoperative seizures.
by large RCTs justifies the use of TXA as a basic trauma The mechanism postulated for this effect was the binding of
protocol. The use, however, must follow the guidelines al- TXA to GABA receptors, which abolished GABA-mediated
ready defined and be early. The mortality-reduced potential inhibition in the central nervous system [9, 56, 57].
cannot be ignored. In TBI, TXA use presents its best potential There is currently no clinical evidence that the use of TXA
(Table 4). increases the risk of thromboembolic events, such as acute

Table 5 Benefits, risks, and


limitations of TXA use in Benefits and recommended Risks and limitations of
neurosurgery studies

Spine surgery Intraoperative bleeding reduction Different study protocols


Need for blood transfusion reduction Limited samples
Total blood loss reduction Larger doses do not enhance
TXA use is not linked to complications results
Low doses should be used Large doses may increase risk
of complications
Chronic patients need careful evaluation
Thromboembolic events are
rare
Brain surgery TXA use is not linked to complications Few studies on the topic
Use should be cautious Different study protocols
Use cannot be indiscriminate Limited samples
Chronic patients need careful evaluation New studies on the topic are
necessary
Ruptured Bleeding rate reduction Increase risk of cerebral
intracranial Mortality reduction infarction
aneurism Different study protocols
Low doses should be used
Chronic patients need careful evaluation Limited samples
Late use has no benefit
New studies on the topic are
necessary
Traumatic brain Mortality reduction Use after 3 h has no effect
injury TXA use is not linked to complications Larger doses do not
Treatment must be early recommended
Recommended use for severe and moderate TBI and
mild TBI in case of bleeding risk
Low doses are efficient
Neurosurg Rev

myocardial infarction, encephalic vascular accidents, deep 7. Elwatidy S, Jamjoom Z, Elgamal E, Zakaria A, Turkistani A, El-
Dawlatly A (2008) Efficacy and safety of prophylactic large dose of
vein thrombosis, or pulmonary embolism [9, 52, 54]
tranexamic acid in spine surgery. Spine (Phila Pa 1976) 33:2577–
(Table 5). 2580. https://doi.org/10.1097/brs.0b013e318188b9c5
8. Cheriyan T, Maier SP, Bianco K, Slobodyanyuk K, Rattenni RN,
Lafage V, Schwab FJ, Lonner BS, Errico TJ (2015) Efficacy of
tranexamic acid on surgical bleeding in spine surgery: a meta-anal-
Conclusion ysis. Spine J 15:752–761. https://doi.org/10.1016/j.spinee.2015.01.
013
TXA in neurosurgeries is a promising method for the mainte- 9. Ng WCK, Jerath A, Wasowicz M (2015) Tranexamic acid: a clin-
nance of hemostasis in affected patients, as demonstrated by ical review. Anestezjol Intens Ter 47:339–350. https://doi.org/10.
5603/ait.a2015.0011
its use in TBI and spine surgery; nevertheless, there is lack of
10. Lecker I, Orser BA, Mazer CD (2012) Saisir la chance de
evidence in brain and vascular surgeries, and several points comprendre les antifibrinolytiques. Can J Anesth 59:1–5. https://
remain unclear. There is still no consensus on the specific dose doi.org/10.1007/s12630-011-9621-4
at which TXA is beneficial or harmful, with conclusions re- 11. Goobie S (2013) The case for the use of tranexamic acid. Paediatr
Anaesth 23:281–284. https://doi.org/10.1111/pan.12093
stricted for each study.
12. Pisklakov S, Ibrahim H, Huang L (2017) Tranexamic acid and
New studies are necessary to provide more accurate anal- major spine surgery: trends and controversies. J Surg Anesth 1:1–8
ysis of the possible benefits of TXA for chronic kidney dis- 13. Carabini LM, Moreland NC, Vealey RJ, Bebawy JF, Koski TR,
ease patients. Larger sample studies are needed to investigate Koht A, Gupta DK, Avram MJ, Zeeni C, Gould RW, Hemmer
the impact on brain surgery and intracranial aneurysm LB, Sugrue PA, McClendon J (2018) A randomized controlled trial
of low-dose tranexamic acid versus placebo to reduce red blood cell
surgery. transfusion during complex multilevel spine fusion surgery. World
Neurosurg 110:e572–e579. https://doi.org/10.1016/j.wneu.2017.
Compliance with ethical standards 11.070
14. Mebel D, Akagami R, Flexman AM (2016) Use of tranexamic acid
is associated with reduced blood product transfusion in complex
Conflict of interest The authors declare that they have no conflict of
skull base neurosurgical procedures: a retrospective cohort study.
interest.
Anesth Analg 122:503–508. https://doi.org/10.1213/ANE.
0000000000001065
Ethical approval For this type of study, formal consent is not required. 15. Shi H, Ou Y, Jiang D, Quan Z, Zhao Z, Zhu Y (2017) Tranexamic
acid reduces perioperative blood loss of posterior lumbar surgery
Animal experiments This article does not contain any studies with hu- for stenosis or spondylolisthesis a randomized trial. Med (US) 96:
man participants or animals performed by any of the authors. 1–8. https://doi.org/10.1097/MD.0000000000005718
16. Zhang F, Wang K, Li FN, Huang X, Li Q, Chen Z, Tang YB, Shen
HX, Song QX (2014) Effectiveness of tranexamic acid in reducing
blood loss in spinal surgery: a meta-analysis. BMC Musculoskelet
References Disord 15:1–9. https://doi.org/10.1186/1471-2474-15-448
17. Shakeri M, Salehpour F, Shokouhi G, Aeinfar K, Aghazadeh J,
1. Udupi B, Siva Satya Prakash M, Adinarayanan S, Mishra S, Babu Mirzaei F, Alavi SAN (2018) Minimal dose of tranexamic acid is
L, Vel R (2014) Effect of low dose tranexamic acid on intra- effective in reducing blood loss in complex spine surgeries: a ran-
operative blood loss in neurosurgical patients. Saudi J Anaesth 9: domized double-blind placebo controlled study. Asian Spine J 12:
42. https://doi.org/10.4103/1658-354x.146304 484–489. https://doi.org/10.4184/asj.2018.12.3.484
2. Hooda B, Chouhan RS, Rath GP, Bithal PK, Suri A, Lamsal R 18. Tsutsumimoto T, Shimogata M, Ohta H, Yui M, Yoda I, Misawa H
(2017) Effect of tranexamic acid on intraoperative blood loss and (2011) Tranexamic acid reduces perioperative blood loss in cervical
transfusion requirements in patients undergoing excision of intra- laminoplasty. Spine (Phila Pa 1976) 36:1913–1918. https://doi.org/
cranial meningioma. J Clin Neurosci 41:132–138. https://doi.org/ 10.1097/brs.0b013e3181fb3a42
10.1016/j.jocn.2017.02.053 19. Raksakietisak M, Sathitkarnmanee B, Srisaen P, Duangrat T,
3. Choi HY, Hyun SJ, Kim KJ, Jahng TA, Kim HJ (2017) Chinachoti T, Rushatamukayanunt P, Sakulpacharoen N (2015)
Effectiveness and safety of tranexamic acid in spinal deformity Two doses of tranexamic acid reduce blood transfusion in complex
surgery. J Korean Neurosurg Soc 60:75–81. https://doi.org/10. spine surgery. Spine (Phila Pa 1976) 40:E1257–E1263. https://doi.
3340/jkns.2016.0505.004 org/10.1097/BRS.0000000000001063
4. Seddighi A, Nikouei A, Seddighi A, Zali A, Tabatabaei S, 20. Colomina MJ, Koo M, Basora M, Pizones J, Mora L, Bagó J (2016)
Yourdkhani F, Naimian S, Razavian I (2015) The role of Intraoperative tranexamic acid use in major spine surgery in adults:
tranexamic acid in prevention of hemorrhage in major spinal sur- a multicentre, randomized, placebo-controlled trial † †This Article
geries. Asian J Neurosurg 12:501–505. https://doi.org/10.4103/ is accompanied by Editorial Aew470. Br J Anaesth 118:380–390.
1793-5482.165791 https://doi.org/10.1093/bja/aew434
5. Meneghini L, Zadra N, Aneloni V, Metrangolo S, Faggin R, Giusti 21. Yagi M, Hasegawa J, Nagoshi N, Iizuka S, Kaneko S, Fukuda K,
F (2003) Erythropoietin therapy and acute preoperative Takemitsu M, Shioda M, MacHida M (2012) Does the intraopera-
normovolaemic haemodilution in infants undergoing craniosynos- tive tranexamic acid decrease operative blood loss during posterior
tosis surgery. Paediatr Anaesth 13:392–396 spinal fusion for treatment of adolescent idiopathic scoliosis? Spine
6. Paulo D, Semonche A, Choudhry O, Al-Mufti F, Prestigiacomo CJ, (Phila Pa 1976) 37:16–20. https://doi.org/10.1097/BRS.
Roychowdhury S, Nanda A, Gupta G (2019) History of hemostasis 0b013e318266b6e5
in neurosurgery. World Neurosurg 124:237–250. https://doi.org/ 22. Xue P, Yang J, Xu X, Liu T, Huang Y, Qiao F, Huang X (2018)
10.1016/j.wneu.2018.12.015 The efficacy and safety of tranexamic acid in reducing perioperative
Neurosurg Rev

blood loss in patients with multilevel thoracic spinal stenosis: a hemorrhage: a prospective randomized study. J Neurosurg 97:771–
retrospective observational study. Med (US) 97. https://doi.org/10. 778
1097/MD.0000000000013643 38. Fodstad H, Nilsson IM (1981) Coagulation and fibrinolysis in
23. Camille D, Gaëtan T, Vianney G, Solène V, Emmanuel F, Rémi G, blood and cerebrospinal fluid after aneurysmal subarachnoid haem-
Mourad O-S (2019) Blood loss and perioperative transfusions re- orrhage: effect of tranexamic acid (AMCA). Acta Neurochir (Wien)
lated to surgery for spinal tumors. Relevance of tranexamic acid. 56:25–38. https://doi.org/10.1007/BF01400969
Neurochirurgie. 65:377–381. https://doi.org/10.1016/j.neuchi. 39. Vermeulen M, Lindsay KW, Murray GD, Cheah F, Hijdra A,
2019.05.003 Muizelaar JP, Schannong M, Teasdale GM, Van Crevel H, Van
24. Dadure C, Sauter M, Bringuier S, Bigorre M, Raux O, Rochette A, Gijn J (1984) Antifibrinolytic treatment in subarachnoid hemor-
Canaud N, Capdevila X (2011) Intraoperative tranexamic acid re- rhage. N Engl J Med 311:432–437. https://doi.org/10.1056/
duces blood transfusion in children undergoing craniosynostosis NEJM198408163110703
surgery: a randomized double-blind study. Anesthesiology 114: 40. Chandra B (1978) Treatment of subarachnoid hemorrhage with
856–861. https://doi.org/10.1097/ALN.0b013e318210f9e3 tranexamic acid (a double-blind clinical trial). Stroke 9:105
25. Kurnik NM, Pflibsen LR, Do A, Bristol R, Singh DJ (2018) 41. Roos Y (2000) Antifibrinolytic treatment in subarachnoid hemor-
Craniosynostosis surgery and the impact of tranexamic acid dosing. rhage: a randomized placebo-controlled trial. Neurology 54:77–82.
J Craniofac Surg 29:96–98. https://doi.org/10.1097/SCS. https://doi.org/10.1212/wnl.54.1.77
0000000000004196 42. Moreira MM, Adry RAR d C, Pereira CU (2019) Características do
26. Huebner BR, Dorlac WC, Cribari C (2017) Tranexamic acid use in Hematoma Epidural Contralateral após Craniectomia
prehospital uncontrolled hemorrhage. Wilderness Environ Med 28: Descompressiva para Tratamento de Trauma Cranioencefálico
S50–S60. https://doi.org/10.1016/j.wem.2016.12.006 Grave. Revisão Sistemática Jbnc - J Bras Neurocir 28:101–110.
27. Na HS, Shin HJ, Lee YJ, Kim JH, Koo KH, Do SH (2016) The https://doi.org/10.22290/jbnc.v28i2.1681
effect of tranexamic acid on blood coagulation in total hip replace- 43. Roberts I, Shakur H, Coats T, Hunt B, Balogun E, Barnetson L,
ment arthroplasty: rotational thromboelastographic (ROTEM®) Cook L, Kawahara T, Perel P, Prieto-Merino D, Ramos M, Cairns
analysis. Anaesthesia 71:67–75. https://doi.org/10.1111/anae. J, Guerriero C (2013) The CRASH-2 trial: A randomised controlled
13270 trial and economic evaluation of the effects of tranexamic acid on
28. Napolitano LM, Cohen MJ, Cotton BA, Schreiber MA, Moore EE death, vascular occlusive events and transfusion requirement in
(2013) Tranexamic acid in trauma. J Trauma Acute Care Surg 74: bleeding trauma patients. Health Technol Assess (Rockv) 17:1–
1575–1586. https://doi.org/10.1097/TA.0b013e318292cc54 80. https://doi.org/10.3310/hta17100
29. Germans MR, Post R, Coert BA, Rinkel GJE, Vandertop WP, 44. Roberts I, Edwards P, Prieto D, Joshi M, Mahmood A, Ker K,
Verbaan D (2013) Ultra-early tranexamic acid after subarachnoid Shakur H (2017) Tranexamic acid in bleeding trauma patients: an
hemorrhage (ULTRA): study protocol for a randomized controlled exploration of benefits and harms. Trials 18:1–6. https://doi.org/10.
trial. Trials 14:1–6. https://doi.org/10.1186/1745-6215-14-143 1186/s13063-016-1750-1
30. Post R, Germans MR, Boogaarts HD, Ferreira Dias Xavier B, Van 45. Ebrahimi P, Mozaffari J, Bahrami R, Hanafi MG, Mousavinejad M
den Berg R, Coert BA, Vandertop WP, Verbaan D (2019) Short- (2019) Intravenous tranexamic acid for subdural and epidural intra-
term tranexamic acid treatment reduces in-hospital mortality in an- cranial hemorrhage: randomized, double-blind, placebo-controlled
eurysmal sub-arachnoid hemorrhage: a multicenter comparison trial. Rev Recent Clin Trials 14. https://doi.org/10.2174/
study. PLoS One 14:1–9. https://doi.org/10.1371/journal.pone. 1574887114666190620112829
0211868 46. Weng S, Wang W, Wei Q, Lan H, Su J, Xu Y (2019) Effect of
31. Baharoglu MI, Germans MR, Rinkel GJE, Algra A, Vermeulen M, tranexamic acid in patients with traumatic brain injury: a systematic
van Gijn J, Roos YBWEM (2013) Antifibrinolytic therapy for an- review and meta-analysis. World Neurosurg 123:128–135. https://
eurysmal subarachnoid haemorrhage. Cochrane Database Syst Rev doi.org/10.1016/j.wneu.2018.11.214
2013. https://doi.org/10.1002/14651858.CD001245.pub2 47. Rossaint R, Bouillon B, Cerny V, Coats TJ, Duranteau J,
32. Gaberel T, Magheru C, Emery E, Derlon JM (2012) Fernández-Mondéjar E, Filipescu D, Hunt BJ, Komadina R,
Antifibrinolytic therapy in the management of aneurismal sub- Nardi G, Neugebauer EAM, Ozier Y, Riddez L, Schultz A,
arachnoid hemorrhage revisited. A meta-analysis. Acta Neurochir Vincent JL, Spahn DR (2016) The European guideline on manage-
(Wien) 154:1–9. https://doi.org/10.1007/s00701-011-1179-y ment of major bleeding and coagulopathy following trauma: Fourth
33. Roos YBWE, Beenen LFM, Groen RJM, Albrecht KW, edition. Crit Care 20:1–55. https://doi.org/10.1186/s13054-016-
Vermeulen M (1997) Timing of surgery in patients with aneurys- 1265-x
mal subarachnoid haemorrhage: Rebleeding is still the major cause 48. Crash T (2019) Articles Effects of tranexamic acid on death, dis-
of poor outcome in neurosurgical units that aim at early surgery. J ability, vascular occlusive events and other morbidities in patients
Neurol Neurosurg Psychiatry 63:490–493 with acute traumatic brain injury ( CRASH-3 ): a randomised. 6736
34. Fodstad H, Forssell A, Liliequist B, Schannong M (1981) 49. Chan DYC, Tsang ACO, Li LF, Cheng KKF, Tsang FCP, Taw
Antifibrinolysis with tranexamic acid in aneurysmal subarachnoid BBT, Pu JKS, Ho WWS, Lui WM, Leung GKK (2019)
hemorrhage: a consecutive controlled clinical trial. Neurosurgery 8: Improving survival with tranexamic acid in cerebral contusions or
158–165. https://doi.org/10.1227/00006123-198102000-00004 traumatic subarachnoid hemorrhage: univariate and multivariate
35. Wijdicks EFM, Hasan D, Lindsay KW, Brouwers PJAM, Hatfield analysis of independent factors associated with lower mortality.
R, Murray GD, van Gijn J, Vermeulen M (1989) Short-term World Neurosurg 125:e665–e670. https://doi.org/10.1016/j.wneu.
tranexamic acid treatment in aneurysmal subarachnoid hemorrhage. 2019.01.145
Stroke 20:1674–1679. https://doi.org/10.1161/01.STR.20.12.1674 50. El-Menyar A, Sathian B, Wahlen BM, Abdelrahman H, Peralta R,
36. Eastin TR, Snipes CD, Seupaul RA (2014) Are antifibrinolytic Al-Thani H, Rizoli S (2019) Prehospital administration of
agents effective in the treatment of aneurysmal subarachnoid hem- tranexamic acid in trauma patients: a 1:1 matched comparative
orrhage? Ann Emerg Med 64:658–659. https://doi.org/10.1016/j. study from a level 1 trauma center. Am J Emerg Med. 38:266–
annemergmed.2014.06.004 271. https://doi.org/10.1016/j.ajem.2019.04.051
37. Hillman J, Fridriksson S, Nilsson O, Yu Z, Säveland H, Jakobsson 51. Chakroun-Walha O, Samet A, Jerbi M, Nasri A, Talbi A, Kanoun
K-E (2002) Immediate administration of tranexamic acid and re- H, Souissi B, Chtara K, Bouaziz M, Ksibi H, Rekik N (2018)
duced incidence of early rebleeding after aneurysmal subarachnoid Benefits of the tranexamic acid in head trauma with no extracranial
Neurosurg Rev

bleeding: a prospective follow-up of 180 patients. Eur J Trauma A, Aglio LS, Smith TR, Mekary DR, Zaidi H (2019) The safety
Emerg Surg 45:1–8. https://doi.org/10.1007/s00068-018-0974-z and efficacy of tranexamic acid in adult spinal deformity surgery: a
52. Sponseller PD, Johnson CC, Nami N, Wetzler JA, Frank SM, meta-analysis. Value Health 22:S176. https://doi.org/10.1016/j.
Goobie SM, Johnson DJ (2016) High-dose versus low-dose jval.2019.04.757
tranexamic acid to reduce transfusion requirements in pediatric sco- 56. Sharma V, Katznelson R, Jerath A, Garrido-Olivares L, Carroll J,
liosis surgery. J Pediatr Orthop 37:e552–e557. https://doi.org/10. Rao V, Wasowicz M, Djaiani G (2014) The association between
1097/bpo.0000000000000820 tranexamic acid and convulsive seizures after cardiac surgery: a
53. Martin K, Breuer T, Gertler R, Hapfelmeier A, Schreiber C, Lange multivariate analysis in 11 529 patients. Anaesthesia 69:124–130.
R, Hess J, Wiesner G (2011) Tranexamic acid versus ε- https://doi.org/10.1111/anae.12516
aminocaproic acid: efficacy and safety in paediatric cardiac surgery. 57. Murkin JM, Falter F, Granton J, Young B, Burt C, Chu M (2010)
Eur J Cardio-thoracic Surg 39:892–897. https://doi.org/10.1016/j. High-dose tranexamic acid is associated with nonischemic clinical
ejcts.2010.09.041 seizures in cardiac surgical patients. Anesth Analg 110:350–353.
54. Ngaage DL, Bland JM (2010) Lessons from aprotinin: is the routine https://doi.org/10.1213/ANE.0b013e3181c92b23
use and inconsistent dosing of tranexamic acid prudent? Meta-
analysis of randomised and large matched observational studies.
Publisher’s note Springer Nature remains neutral with regard to jurisdic-
Eur J Cardio-thoracic Surg 37:1375–1383. https://doi.org/10.
tional claims in published maps and institutional affiliations.
1016/j.ejcts.2009.11.055
55. Hariharan D, Mammi M, Daniels K, Petrucci K, Lamba N,
Cerecedo-Lopez CD, Doucette J, Papatheodorou S, Hulsbergen

You might also like