Skip to main content

Tranexamic acid for haemostasis and beyond: does dose matter?

Abstract

Tranexamic acid (TXA) is a widely used antifibrinolytic agent that has been used since the 1960’s to reduce blood loss in various conditions. TXA is a lysine analogue that competes for the lysine binding sites in plasminogen and tissue-type plasminogen activator impairing its interaction with the exposed lysine residues on the fibrin surface. The presence of TXA therefore, impairs the plasminogen and tPA engagement and subsequent plasmin generation on the fibrin surface, protecting fibrin clot from proteolytic degradation. However, critical lysine binding sites for plasmin(ogen) also exist on other proteins and on various cell-surface receptors allowing plasmin to exert potent effects on other targets that are unrelated to classical fibrinolysis, notably in relation to immunity and inflammation. Indeed, TXA was reported to significantly reduce post-surgical infection rates in patients after cardiac surgery unrelated to its haemostatic effects. This has provided an impetus to consider TXA in other indications beyond inhibition of fibrinolysis. While there is extensive literature on the optimal dosage of TXA to reduce bleeding rates and transfusion needs, it remains to be determined if these dosages also apply to blocking the non-canonical effects of plasmin.

Background

Fibrinolysis, plasminogen activation and the harnessing for therapeutic use

Fibrinolysis is the enzymatic process underlying the removal of fibrin deposits and blood clots [1]. Fibrin comprises cross-linked fibrin monomers that provides structural integrity to blood clots. However, fibrin also harbours a means of self-destruction via the exposure of lysine residues on its surface which provides docking sites for the key fibrinolytic components plasminogen and tissue-type plasminogen activator (tPA) [2]. This then allows for the localised formation of plasmin, that executes the fibrinolytic event (Fig. 1). This fibrin-targeted feature of tPA also serves to limit systemic plasminogen activation. Plasminogen, the zymogenic precursor to active plasmin, circulates in plasma in abundance and can also be converted to plasmin by a second plasminogen activator, urokinase-type plasminogen activator (uPA) [3]. While both uPA and tPA cleave plasminogen at the same site to generate active plasmin, there are important features that separate tPA and uPA. For example, tPA is a relatively poor plasminogen activator in solution, but becomes far more efficient at activating plasminogen on the clot surface. On the other hand, uPA-mediated plasminogen activation is not at all reliant on fibrin and it could be argued that its role in the removal of fibrin deposits and blood clots is secondary to tPA [2]. The plasminogen activators are also tightly modulated to limit indiscriminate plasmin generation. Both tPA and uPA are inhibited by plasminogen activator inhibitors (PAI)-1 and PAI-2, while plasmin itself is regulated by ɑ2-antiplasmin and secondarily by C1-inhibitor (C1-INH) [4] and ɑ2-macroglobulin [5, 6].

Fig. 1
figure 1

Schematic diagram of the main mechanisms of the fibrinolytic system and the target site of tranexamic acid (TXA). Lysine binding sites located within the second kringle domain of tPA, and in 4 of the 5 kringle domains of plasminogen can bind to exposed lysine residues on the fibrin surface. This interaction can be inhibited physiologically by Thrombin activatable fibrinolysis inhibitor (TAFI) or pharmacologically by competitive inhibition by the lysine analogue, TXA. The juxtaposition of tPA and plasminogen on the clot surface permits plasmin generation and subsequent fibrinolysis and the formation of fibrin degradation products. Key modulators of this process include plasminogen activator inhibitor-1, PAI-1 (and to some extent, PAI-2) that inhibit both tPA and uPA, and antiplasmin that specifically inhibits plasmin activity. Plasmin can also be inhibited by other molecules including C1-inhibitor (C1-INH) or by alpha2 macroglobulin (A2M)

tPA and uPA are also expressed outside the circulation and in different locations. For example, tPA is widely expressed in neurons [7] and can activate microglia [8, 9] and modulate the activity of N-methyl-D-aspartate (NMDA) receptors in the central nervous system (CNS) [10, 11]. uPA can also be found in the CNS [12, 13], but is also quite prominently expressed in extravascular compartments and is prominently expressed in the kidney [14]. This expression pattern also points to other important regulatory roles that are clearly distinct from blood clot removal. Cell-associated activities are linked to 12 known plasminogen receptors detected on most, if not all, immune cells [15]. The association of plasminogen with these cell receptors trigger downstream signalling pathways linked to immune cell recruitment, modulation of inflammation and improved wound healing [15,16,17].

Plasmin is also known to cleave substrates other than fibrin, such as coagulation factors (i.e. Factor V, VIII, XI, XII) and growth factors (i.e. pro-BDNF) [18, 19]. Similarly, plasmin also has the capacity to initiate non-haemostatic processes by activating the complement system, notably by cleavage of complement proteins C5, C3 and activating various metalloproteinases [20,21,22]. Plasmin can also target misfolded proteins for clearance including amyloid-beta (Aβ) [23, 24]. Within the CNS, tPA has been linked to various aspects of neuronal function, including the promotion of synaptic plasticity and increasing blood–brain barrier (BBB) permeability [25,26,27,28]. Some of these effects of tPA also involve plasmin generation within the CNS [29]. With such a broad array of various biological processes, plasmin and components of the fibrinolytic system appear to be an attractive target for treatment of a range of conditions.

Targeting fibrinolysis: the critical role of lysine residues

Indeed, the interaction of exposed lysine residues on the fibrin surface with lysine binding sites within both tPA and plasminogen, is key to the localized conversion of plasminogen into plasmin [2]. Up until now, the only clinical justification to modulate the plasminogen activating system has been for the purpose of either enhancing fibrin removal (i.e. thrombolysis), or to limit bleeding. Promoting fibrinolysis for myocardial infarction, pulmonary embolism or ischaemic stroke, has been attempted by the administration of high doses of exogenous tPA or uPA (or variants) to increase plasmin generation within the circulation to remove the offending blood clot. On the other side of the coin, blocking the fibrinolytic system has been achieved by interfering with the very process that facilitates the targeted binding of plasminogen and tPA to the fibrin surface, namely lysine residues. This can occur physiologically by the enzymatic removal of lysine residues from the fibrin surface by the carboxypeptidase, thrombin activatable fibrinolysis inhibitor (TAFI [30]), also known as carboxypeptidase U (CPU [31]). However, this has also been achieved pharmacologically with the use of lysine analogues that act as competitive inhibitors. These drugs were developed in Japan in the 1960s by Okamoto et al. [32, 33]. The most commonly used lysine analogues, tranexamic acid (TXA) or epsilon-aminocaproic acid (EACA), bind to the lysine binding sites in plasminogen and tPA, thereby disabling their ability to dock on the lysine residues on the fibrin surface, hence sparing fibrin from fibrinolysis. These lysine analogues (mostly TXA) have been widely used in routine haematology as effective antifibrinolytic agents in a variety of conditions associated with bleeding [34,35,36,37] (Fig. 2).

Fig. 2
figure 2

Schematic representation of the known effects of plasmin(ogen) beyond fibrinolysis and how tranexamic acid (TXA) can potentially modulate these processes

Tranexamic acid: current usage and understanding

Haemostatic indications

In different countries or jurisdictions, the approved indications for TXA use varies greatly. As indicated by the Therapeutic Goods Administration (TGA) Australia, the current approved indications for TXA (intravenous) include the reduction of peri- and post-operative blood loss in cardiac surgery and total hip or knee arthroplasty [38]. Oral tablets are recommended for short-term use to treat menorrhagia, hyphaema and clotting disorders [38]. In the USA, oral TXA is only indicated for patients with haemophilia or those undergoing dental extractions [38]. Pharmacokinetic data suggests that the elimination half-life of TXA is approximately 2 h when administered either orally or intravenously [39]. Bioavailability data appears to vary amongst administration routes and other factors, ranging from 33% to about 53% [38,39,40].

The effectiveness of TXA in reducing surgical bleeding and transfusion requirements is well established. TXA has been shown to reduce blood transfusion requirements by 25%-60% in orthopaedic surgeries, including primary knee and hip arthroplasty [41]. These beneficial outcomes are accompanied by no evidence for an increased risk of thromboembolic complications [42]. In cardiothoracic surgery, post-operative bleeding is a common complication as a result of cardiopulmonary bypass (CPB), circulatory dilution, aggressive anticoagulation, platelet dysfunction and a systemic inflammatory response to CPB. TXA has been shown to reduce bleeding post CPB as well as transfusion requirements without any evidence of increased death or thromboembolic complications [36, 43]. A seminal trial of TXA versus placebo in coronary artery surgery was the ATACAS trial, which found that patients randomised to TXA received 46% fewer blood products than those in the placebo group. The investigators found that the use of TXA would save approximately 57 units of blood products for every 100 patients treated, which are effects similar to those reported in other meta-analyses [44]. The trial also found that TXA treatment resulted in significantly lower rates of reoperation and a general improvement in blood-sparing effects [36]. TXA was however associated with a higher risk of seizures in this patient group, possibly due to the significantly higher dose of TXA used. TXA is also used aggressively to reduce obstetric bleeding, which was in fact the original purpose and goal for the development of TXA by Dr. Okamoto. The success of TXA in reducing bleeding was demonstrated in the WOMAN trial where early administration of TXA significantly reduced post-partum haemorrhage which could have otherwise resulted in death [45].

In trauma and traumatic brain injury (TBI), it was shown that TXA safely reduces the risk of mortality from injury-related bleeding when administered within the first 3 h [46, 47]. Interestingly, when TXA was administered after 3 h, it caused an increase in bleeding and mortality. The molecular underpinnings of this has eluded the field although it is speculated to be related to the ability of TXA to paradoxically increase fibrinolytic activity via activation of uPA [48]. The recently completed PATCH-Trauma trial, where patients with severe trauma and at risk of coagulopathy were randomised to TXA by paramedics (i.e. prehospital administration) also reported reduced mortality rates, but an improved functional outcome at 6 months was not shown in the TXA group [49].

In contrast, TXA has not been shown to be effective at improving outcomes in patients with gastrointestinal bleeding in the HALT-IT trial [50, 51]. Furthermore, extended use of a high dose TXA regimen did not reduce death from gastrointestinal bleeding compared to placebo. Concerningly, there were even more venous thromboembolic events in the TXA group, with the researchers recommending that TXA should not be used for the treatment of gastrointestinal bleeding outside the context of a randomised trial [50].

Similarly, TXA was not shown to be beneficial for intracerebral haemorrhage (ICH) in improving clinical outcomes as demonstrated by the TICH-2 trial and STOP-AUST trial [51, 52]. In particular, treatment for ICH requires early administration, as haematoma expansions occur shortly after ICH, and is often a determinant of favourable outcomes [53, 54]. Although the TICH-2 trial showed fewer deaths by day 7 in the TXA treatment arm, there was no difference in mortality at 90 days between the two groups [51]. In the STOP-AUST trial, patients who suffered from acute ICH were administered TXA within a 4-5 h time window [52]. Likewise, there was no effect on the growth of ICH. Several systematic reviews however appear to suggest otherwise, indicating that TXA had successfully reduced the risk of ICH growth, although longer term mortality outcomes, rebleeding risk and length of hospital stay remained unchanged [55, 56].

Given the variable effects of TXA on bleeding and functional outcomes, this may suggest that TXA is augmenting other processes unrelated to its blood-sparing effects which should be taken into consideration.

Haemostatic indications for TXA linked to amyloid-mediated plasmin generation

Cerebral amyloid angiopathy

Cerebral amyloid angiopathy (CAA) is a serious and increasingly recognised neurological condition largely diagnosed in those over the age of 50 years. CAA is caused by pathologic deposition of amyloid-beta (Aβ40) within the vessels of cortical and leptomeningeal arterioles [57, 58]. CAA results in intracranial bleeding in the form of lobar ICH, microbleeds, convexity subarachnoid haemorrhage (cSAH) and superficial siderosis [59, 60]. Several mechanisms have been proposed of how this occurs but there is evidence for BBB leakiness and inflammation due to cerebrovascular deposits of Aβ40. There are no current treatment options available for recurrent or incident stroke prevention in CAA other than generic stroke prevention lifestyle advice, lowering blood pressure and avoiding concurrent antithrombotic treatments. Existing studies of drugs that primarily target Aβ40 have not been successful [61, 62]. More recently however, TXA has been proposed as a novel means to reduce the risk of ICH in CAA. The rationale for this stems from the observation that amyloid deposits can initiate plasminogen activation in the same lysine-dependent manner as fibrin, and hence can be inhibited by TXA [63]. Indeed, the plasminogen activating system is one of the proteolytic systems involved in the clearance of amyloid [64]. Hence in CAA, amyloid deposits within cerebral vessels have the potential to generate plasmin. In this scenario, it is amyloid deposition, rather than fibrin formation that is driving plasmin generation. As plasmin has also been reported to increase BBB, TXA could provide a potential means to prevent spontaneous ICH in CAA patients [65]. While this is yet to be studied in humans, intravenous TXA was shown to be able to reduce acute hematoma expansion in a post-hoc analysis of CAA patients in the TICH-2 trial, although there was no favourable effect on clinical outcome [66].

A potential concern of using long term oral TXA as secondary stroke prevention in CAA, is that plasmin is involved in the clearance of amyloid and other misfolded proteins and that TXA may inhibit this process, causing increased Aβ40 deposition. However, a recent study evaluating the effect of long term oral TXA administration to mice predisposed to Alzheimer’s disease, did not reveal any change in Aβ40 levels in the brain [67] although more research is needed to investigate the safety and efficacy of TXA in CAA patients. Hence the stage is set to evaluate TXA as a novel means to reduce ICH in patients with CAA although the dosage and duration of TXA treatment needs to considered.

Non-haemostatic indications for TXA

Given plasmin(ogen)’s many non-canonical properties mentioned earlier, and the fact that many of these processes are lysine-dependent, it is not surprising that TXA may block plasmin’s ability to modulate these processes as well (Fig. 2). In some circumstances, these non-canonical effects of plasmin can be potentially deleterious [68, 69]. In the case of TBI, plasmin promotes the breakdown of laminin in the hippocampus, which subsequently triggers neuronal death [70]. At the same time, plasmin-mediated activation of the complement cascade can drive neuroinflammation and secondary injury in TBI [71]. A similar effect has also been observed in the ischemic brain [72]. As a result, there has been growing interest in the therapeutic use of TXA for purposes beyond its antifibrinolytic effects.

Inflammation and immunity

Plasminogen is well known to have an important role in inflammation and immunity. In the mid 1990’s, studies using plasminogen deficient mice revealed a critical role for plasminogen in immune cell (macrophage and lymphocyte) recruitment during the pro-inflammatory phase of the immune response [73]. Immune cells have a very high capacity to engage plasminogen and this is mediated by its ability to bind to plasminogen receptors on the surface of immune cells. At least 12 distinct cell surface plasminogen receptors have been described [74] and most of these contain a C-terminal lysine residue that is recognised by the lysine binding sites within the plasminogen kringle domains. Hence, TXA will block the binding of plasminogen to these receptors and interfere with intracellular signal transduction pathways initiated by direct receptor binding or via cell surface plasmin generation. This has already been demonstrated for the plasminogen receptor, Plg-RKT [17] and using Plg-RKT deficient mice in models of wound healing.

Growing evidence has now accumulated indicating the effect of TXA on inflammation and immunity in vivo. TXA has been shown to have anti-inflammatory effects attributed to plasmin inhibition [75]. More recently, Draxler and colleagues showed a reduction in postsurgical infection rates among patients undergoing cardiac surgery who were randomised to TXA compared to placebo despite all patients receiving prophylactic antibiotics [36, 76]. This effect was independent of transfusion requirements and TXA’s effect on bleeding reduction. TXA-mediated plasmin blockade was found to enhance the expression of immune-activating markers (CD83 on monocytes and CCR7 on dendritic cells) while reducing the expression of immunosuppressive markers (i.e. Programmed Cell Death Ligand-1; PDL-1) leading to improved immune competence on multiple myeloid and lymphoid populations in peripheral blood. Other studies have also shown similar results with TXA reducing levels of IL-6, IL-1β and TNF-α in patients undergoing CPB [77]. Draxler and colleagues have also shown that TXA reduces baseline IL-6, TNF-α, INF-γ and IL-10 levels in the blood of healthy subjects administered TXA [76]. A recent systematic review and meta-analysis further highlights the anti-inflammatory effects of TXA in cardiac surgery patients [78].

In orthopaedic surgery, TXA has also been shown to reduce plasma levels of CRP and IL-6 after total knee and hip arthroplasty. This may be related to both the direct and indirect effects of plasmin’s ability to activate several inflammatory pathways initiated by cell surface plasminogen receptors (above), including the complement system [79, 80].

While the capacity of TXA to influence these inflammatory processes has been associated with plasmin inhibition, TXA has also been reported to exert direct (i.e. plasmin-independent) anti-microbial effects in vitro [81, 82]. Furthermore, a more recent study reported that TXA could modulate levels of TNF and IL-1 in plasminogen deficient mice [83], again supporting the view that TXA could influence inflammation via other means.

While the above commentary relates to beneficial effects of TXA by blocking the immunosuppressive (and possibly pro-inflammatory) actions of plasmin, it also needs to be considered that elevated levels of plasmin can also exert beneficial effects, and in such a scenario, TXA treatment may counter this to the detriment of the host (see below).

Plasmin formation has also been reported to have both beneficial and detrimental effects during infection and sepsis, respectively, in infection models using plasminogen deficient mice [84]. It is unclear the mechanistic basis for the deleterious effect of plasmin in the sepsis model, but the authors of this study suggested that this might be related to a marked plasmin-mediated pro-inflammatory state, that included complement activation and subsequent STAT3 signalling, but this remains speculative. More recently, Vago et al. reported that plasmin could reduce sepsis severity by reducing inflammation and the formation of neutrophil extracellular traps in a process that was blocked by the addition of TXA [85].

Wound healing

As mentioned earlier, plasminogen is a key component in the wound healing process. Indeed, plasminogen-deficient mice display markedly delayed wound healing [86, 87] and has been reported to act as a master regulator and a potential drug candidate for the healing of radiation wounds [88]. A key question is whether the use of TXA might interfere with any lysine-dependent process associated with wound healing. Even if wound healing was delayed by TXA, this would only be evident for the duration of the TXA treatment period (usually less than 8 h) and therefore may not be clinically noticeable. On the other hand, a different scenario might occur in situations where TXA treatment is prolonged. On this note, recent case reports have provided evidence that topical TXA over 72 h slowed down wound healing during facial plastic surgery [89], however larger studies are needed to confirm this observation.

Melasma

For over 40 years, TXA has been used for cosmetic skin-whitening purposes [90] and more recently as a means to reduce UV-induced hyperpigmentation in melasma by inhibiting the ability of plasmin to increase melanin production in keratinocytes [91,92,93]. TXA seems to be able to block plasmin-mediated induction of arachidonic acid-induced pigmentation [93] while other in vitro research suggests that stimulation of autophagy by TXA as well as the suppression of VEGF-stimulated melanogenesis could also underlie the mechanism for TXA’s anti-melanogenic property [94, 95].

Hereditary Angioedema

Hereditary angioedema (HAE) is a potentially life-threatening condition characterised by recurrent attacks of cutaneous and submucosal angioedema as a result of mutations in the genes encoding C1-INH. Cross communication between coagulation and complement serine protease systems means that C1-INH also inhibits proteases of the coagulation, fibrinolytic and kinin pathways, being a critical physiological inhibitor of plasma kallikrein, FXIa and FXIIa [20, 96]. Edema therefore occurs as a result of excessive bradykinin production from the unregulated activation of the prekallikrein-kallikrein-HMWK-bradykinin system due to C1-INH deficiency or dysfunction [97]. TXA has been used successfully for prophylaxis in patients with HAE with the thought that it reduces the sensitivity to plasminogen cleavage, thus reducing the excess bradykinin production [98, 99] – a mechanism and clinical use completely unrelated to its haemostatic properties.

In close relation to this, TXA appears to play an important role in maintaining vascular integrity in other pathological contexts. Attenuation of the loss of endothelial cell adhesion junctions as well as maintaining the vascular-protective role of the endothelial glycocalyx are important pathways by which TXA achieves this [100].

TXA at the cellular level

At a more cellular level, recent studies have shown that TXA decreased the release of mitochondrial DNA (mtDNA) into plasma following burn injury in mice as well as inhibiting the spontaneous release of mtDNA from human endothelial cells and neutrophils [101]. TXA was also shown to modulate various intracellular signalling pathways in these studies, although the mechanistic underpinnings remain to be determined, and also if this indeed was a consequence of inhibition of the plasminogen activating system. The same group of researchers have shown that TXA enhanced mitochondrial respiration and ATP production; important processes that could work to protect these vital cellular systems and hence the host, in the setting of marked metabolic stress and derangement seen in trauma, surgery and sepsis [102, 103].

Perhaps linked to its effects on mitochondria are the anti-aging properties of TXA published over recent years. Hiramoto et al. have demonstrated that long-term oral TXA administration suppressed age-dependent wrinkle formation in mice with hereditary skin dryness [104]. This was accompanied by increased production of hyaluronic acid, increases in epidermal cell number and reduced proteolytic degradation of the extracellular matrix. Furthermore, there was reduced mast cell proliferation, increased β-endorphin levels and increased expression of μ-opioid receptors on the surface of fibroblasts. Curiously, continuous administration of TXA significantly increased the lifespan of mice and reduced blood levels of IL-6, TNF-α, reactive oxygen species and MMP-9 as well as suppressing age-related osteoporosis and reducing apoptosis in the gonadal organs of aging mice [105, 106].

With growing evidence on the non-haemostatic effects of TXA, it is surprising that most clinical studies described in the previous section have only been directed towards the antifibrinolytic actions of TXA.

Current trials evaluating the potential non-haemostatic benefits of TXA

The promising immune-modulatory potential of TXA is currently being investigated in the treatment of necrotizing soft-tissue infections (TRITON trial: A feasibility trial of tranexamic acid for necrotising soft-tissue infections) as well as its capacity to reduce the incidence of surgical site infection rates in patients undergoing gastrointestinal surgery (TRIGS trial: Tranexamic acid to reduce infection after gastrointestinal surgery) [107, 108]. In addition to its immune-modulating effects, plasmin also has BBB modulating capacities which can contribute to postoperative delirium. As such, a sub study of the TRIGS trial (TRIGS-D) is evaluating whether TXA might reduce neuroinflammation and subsequent postoperative delirium [108].

Dose considerations—how much TXA is enough?

Most studies involving the use of TXA are for haemostatic indications and trials often use bleeding or mortality as the primary outcome measure. A systematic review by Picetti et al. summarized various in vitro studies of fibrinolytic inhibition by TXA [109]. It has been suggested that TXA concentrations of 10–15 mg/L were sufficient to achieve a substantial level of fibrinolysis inhibition in vitro [109]. However, it is of note that the simulated conditions in vitro are vastly different from the clinical context. Nevertheless, these in vitro studies may still provide some guidance and insight into dosing regimens for fibrinolytic inhibition.

In patients, dosage of TXA appears to vary largely between procedures and/or clinical conditions. In particular, administration of TXA to patients undergoing cardiac surgery generally uses uniquely high doses of the drug at up to 100 mg/kg [110]. The ATACAS trial demonstrated reduced bleeding-related complications in cardiac surgery using a dose of 50–100 mg/kg, and post hoc analyses demonstrated likely immune-enhancing properties at this higher dose [110]. Although this provides additional benefit in reducing blood loss and infections, some observational studies have reported that higher doses can also increase the risk of postoperative seizures [36, 110]. The recent OPTIMAL randomised clinical trial compared the efficacy and adverse events of high-dose (30 mg/kg bolus, a 16 mg/kg/h maintenance dose, and a 2 mg/kg prime) versus low-dose tranexamic acid (10 mg/kg bolus, a 2 mg/kg/h maintenance dose, and a 1 mg/kg prime) in patients undergoing cardiac surgery with CPB [43]. This study found that patients receiving the high-dose TXA infusion had a statistically significant reduction in the proportion of patients requiring a blood transfusion [43]. Furthermore, the study results met criteria for noninferiority with respect to composite primary safety endpoints of 30-day mortality, seizure, kidney dysfunction, and thrombotic events [43].

In the context of trauma, patients who received repeated dosing of TXA (3 g in total) had a significantly lower mortality rate compared to placebo (STAAMP trial) [111]. Patients in the CRASH-2 and 3 trials who received a total of 2 g of TXA within 3 h from injury had a significantly reduced risk of bleeding-related deaths [46, 47].

However, as seen in the previously described studies, the potential for TXA to block the non-haemostatic effects of plasmin have not been accounted for and may have provided further benefit but had gone unnoticed. However, there is still a small pool of completed and ongoing studies that utilize TXA for non-haemostatic indications – what remains largely unknown is whether the dosages used are appropriate to block these non-haemostatic pathways of plasmin(ogen).

In patients with melasma, a recent network meta-analysis has suggested that the optimal treatment would be a total daily oral dosage of 500–750 mg for 12 weeks. In this analysis, it was also suggested that treatment duration could influence treatment outcome. However, it is not known if treatment durations beyond 12 weeks would provide extra benefit [112].

In the ongoing trial for necrotizing fasciitis (TRITON), intravenous TXA is administered for a total of 2 g daily for 4 days [107]. The primary outcome of this trial would be a reduction of at least 25% of the initial extent of infection.

In orthopaedic surgery, TXA-associated immune modulation was described in patients undergoing total knee arthroplasty (TKA) [113]. In this trial, only patients receiving a total of more than 6 g of TXA displayed anti-inflammatory effects until 72 h post-treatment [113]. In another trial, patients who underwent total hip arthroplasty and received a total of 3 g of TXA revealed a diminished inflammatory response at 3 days post-surgery [114]. Notably, a regime of four oral doses of TXA (2 g initial + 1 g subsequently) was found to promote both optimal blood loss reduction and a diminished inflammatory response post-operatively following TKA [115]. In another study, patients receiving a high initial dose of TXA (60 mg/kg) with 5 subsequent doses of 1 g over 24 h demonstrated a reduction in inflammatory markers as well as D-dimers [116]. It appears that repeated dosing regimens have been commonly used for patients undergoing orthopaedic surgeries.

Conversely, in trauma patients with severe injury it was found that the reduction in HLA-DR expression on monocytes was not associated with TXA administration up until 72 h post-treatment (2 g and 4 g intravenous bolus) (TAMPITI trial) [117]. This study ultimately found that TXA at this dose had minimal immune modulatory effects with respect to leukocyte and circulating cytokine levels. However, it is important to note that trauma causes a sharp decline in HLA-DR expression even prior to treatment, which can potentially mask the effects of TXA, if any.

This is in contrast to patients who underwent cardiac surgery where TXA administration was found to be immune-activating instead [76]. In the same study, it was also demonstrated in healthy volunteers that a 1 g TXA dose was able to promote immune activating effects at 24 h after administration [76]. These varied findings confirm TXA’s fibrin-independent actions.

Collectively, there appears to be a variable effect of TXA when it comes to its non-haemostatic effects in different clinical settings. We postulate that perhaps the dose and timing of TXA plays an important role in preventing certain lysine-dependent interactions over others. Whether higher doses of TXA are required to observe the immune-modulatory properties in addition to the antifibrinolytic effects warrants further investigation. A summary of the doses of TXA used in haemostatic and non-haemostatic indications is provided in Table 1.

Table 1 Dose of TXA used for haemostatic and non-haemostatic indications

This concept will be an important consideration in the interpretation of results arising from the TRIGS trial, once it is completed. What makes this study unique is that in contrast to other TXA studies, the primary outcome of the TRIGS trial is surgical site infection with transfusion requirements being a secondary outcome measure. The dose of TXA used in the TRIGS trial is a bolus of 15 mg/kg prior to surgical incision followed by an infusion of 5 mg/kg/h until the end of surgery. These doses are lower than the doses used in cardiac surgery and the ATACAS study (50 mg/kg) where changes to immune profiling and surgical site infections were seen. It will be interesting to see whether the lower dose in TRIGS translates to any changes in the non-haemostatic outcomes being measured.

Conclusion

The current knowledge of treatment regimens and dosage suggestions for TXA are based on achieving optimal antifibrinolytic effects. What remains unknown is the immune- and other impacts of TXA at the indicated dosages commonly used to exert its haemostatic effects. The currently available literature is scarce and often involves small, single-centre clinical trials. In future, it is essential for the non-haemostatic effects of TXA to be recognised and considered alongside its antifibrinolytic properties in prospective clinical studies. Similarly, it is important to determine the extent of whether the beneficial effect of TXA at reducing mortality in trauma and in other conditions may not only be due to its effect at reducing bleeding, but actually a consequence of blocking one of the many non-haemostatic effects of plasmin.

This knowledge may ultimately help determine the most appropriate dose and timing of TXA to achieve optimal haemostatic and non-haemostatic outcomes in different clinical settings.

Availability of data and materials

NA.

References

  1. Collen D, Lijnen HR. Basic and clinical aspects of fibrinolysis and thrombolysis. Blood. 1991;78(12):3114–24.

    Article  CAS  PubMed  Google Scholar 

  2. Cesarman-Maus G, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol. 2005;129(3):307–21.

    Article  CAS  PubMed  Google Scholar 

  3. Ploug J, Kjeldgaard NO. Urokinase an activator of plasminogen from human urine .I. Isolation and properties. Biochim Biophys Acta. 1957;24(2):278–82.

    Article  CAS  PubMed  Google Scholar 

  4. Brown EW, Ravindran S, Patston PA. The reaction between plasmin and C1-inhibitor results in plasmin inhibition by the serpin mechanism. Blood Coagul Fibrinolysis. 2002;13(8):711–4.

    Article  CAS  PubMed  Google Scholar 

  5. Ganrot PO. Inhibition of plasmin activity by alpha-2-macroglobulin. Clin Chim Acta. 1967;16(2):328–9.

    Article  CAS  PubMed  Google Scholar 

  6. Kolodziej SJ, Klueppelberg HU, Nolasco N, Ehses W, Strickland DK, Stoops JK. Three-dimensional structure of the human plasmin alpha2-macroglobulin complex. J Struct Biol. 1998;123(2):124–33.

    Article  CAS  PubMed  Google Scholar 

  7. Melchor JP, Strickland S. Tissue plasminogen activator in central nervous system physiology and pathology. Thromb Haemost. 2005;93(4):655–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rogove AD, Siao C, Keyt B, Strickland S, Tsirka SE. Activation of microglia reveals a non-proteolytic cytokine function for tissue plasminogen activator in the central nervous system. J Cell Sci. 1999;112(Pt 22):4007–16.

    Article  CAS  PubMed  Google Scholar 

  9. Siao CJ, Tsirka SE. Tissue plasminogen activator mediates microglial activation via its finger domain through annexin II. J Neurosci. 2002;22(9):3352–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Samson AL, Medcalf RL. Tissue-type plasminogen activator: a multifaceted modulator of neurotransmission and synaptic plasticity. Neuron. 2006;50(5):673–8.

    Article  CAS  PubMed  Google Scholar 

  11. Nicole O, Docagne F, Ali C, Margaill I, Carmeliet P, MacKenzie ET, et al. The proteolytic activity of tissue-plasminogen activator enhances NMDA receptor-mediated signaling. Nat Med. 2001;7(1):59–64.

    Article  CAS  PubMed  Google Scholar 

  12. Bahi A, Kusnecov AW, Dreyer JL. Effects of urokinase-type plasminogen activator in the acquisition, expression and reinstatement of cocaine-induced conditioned-place preference. Behav Brain Res. 2008;191(1):17–25.

    Article  CAS  PubMed  Google Scholar 

  13. Yepes M. Urokinase-type plasminogen activator is a modulator of synaptic plasticity in the central nervous system: implications for neurorepair in the ischemic brain. Neural Regen Res. 2020;15(4):620–4.

    Article  PubMed  Google Scholar 

  14. Zhang G, Eddy AA. Urokinase and its receptors in chronic kidney disease. Front Biosci. 2008;13:5462–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Miles LA, Plow EF, Waisman DM, Parmer RJ. Plasminogen receptors. J Biomed Biotechnol. 2012;2012:130735.

    PubMed  PubMed Central  Google Scholar 

  16. Godier A, Hunt BJ. Plasminogen receptors and their role in the pathogenesis of inflammatory, autoimmune and malignant disease. J Thromb Haemost. 2013;11(1):26–34.

    Article  CAS  PubMed  Google Scholar 

  17. Ny L, Parmer RJ, Shen Y, Holmberg S, Baik N, Backman A, Broden J, Wilczynska M, Ny T, Miles LA. The Plasminogen Receptor, Plg-RKT, plays a role in Inflammation and fibrinolysis during cutaneous wound healing in mice. Cell Death Dis. 2020;11:1054.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pang PT, Teng HK, Zaitsev E, Woo NT, Sakata K, Zhen S, et al. Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity. Sci. 2004;306(5695):487–91.

    Article  CAS  Google Scholar 

  19. van der Vorm LN, Remijn JA, de Laat B, Huskens D. Effects of Plasmin on von Willebrand Factor and Platelets: A Narrative Review. TH Open. 2018;2(2):e218–28.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Foley JH. Plasmin(ogen) at the Nexus of Fibrinolysis, Inflammation, and Complement. Semin Thromb Hemost. 2017;43(2):135–42.

    Article  CAS  PubMed  Google Scholar 

  21. Davis GE, Pintar Allen KA, Salazar R, Maxwell SA. Matrix metalloproteinase-1 and -9 activation by plasmin regulates a novel endothelial cell-mediated mechanism of collagen gel contraction and capillary tube regression in three-dimensional collagen matrices. J Cell Sci. 2001;114(Pt 5):917–30.

    Article  CAS  PubMed  Google Scholar 

  22. Keragala CB, Draxler DF, McQuilten ZK, Medcalf RL. Haemostasis and innate immunity - a complementary relationship: A review of the intricate relationship between coagulation and complement pathways. Br J Haematol. 2018;180(6):782–98.

    Article  PubMed  Google Scholar 

  23. Kingston IB, Castro MJ, Anderson S. In vitro stimulation of tissue-type plasminogen activator by Alzheimer amyloid beta-peptide analogues. Nat Med. 1995;1(2):138–42.

    Article  CAS  PubMed  Google Scholar 

  24. Gebbink MF, Bouma B, Maas C, Bouma BN. Physiological responses to protein aggregates: fibrinolysis, coagulation and inflammation (new roles for old factors). FEBS Lett. 2009;583(16):2691–9.

    Article  CAS  PubMed  Google Scholar 

  25. Yepes M, Sandkvist M, Moore EG, Bugge TH, Strickland DK, Lawrence DA. Tissue-type plasminogen activator induces opening of the blood-brain barrier via the LDL receptor-related protein. J Clin Invest. 2003;112(10):1533–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, et al. Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med. 2008;14(7):731–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Be Niego. Freeman R, Puschmann TB, Turnley AM, Medcalf RL. t-PA–specific modulation of a human blood-brain barrier model involves plasmin-mediated activation of the Rho kinase pathway in astrocytes. Blood. 2012;119(20):4752–61.

    Article  Google Scholar 

  28. Calabresi P, Napolitano M, Centonze D, Marfia GA, Gubellini P, Teule MA, et al. Tissue plasminogen activator controls multiple forms of synaptic plasticity and memory. Eur J Neurosci. 2000;12(3):1002–12.

    Article  CAS  PubMed  Google Scholar 

  29. Niego B, Medcalf RL. Plasmin-dependent modulation of the blood-brain barrier: a major consideration during tPA-induced thrombolysis? J Cereb Blood Flow Metab. 2014;34(8):1283–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bajzar L, Morser J, Nesheim M. TAFI, or plasma procarboxypeptidase B, couples the coagulation and fibrinolytic cascades through the thrombin-thrombomodulin complex. J Biol Chem. 1996;271(28):16603–8.

    Article  CAS  PubMed  Google Scholar 

  31. Willemse JL, Heylen E, Nesheim ME, Hendriks DF. Carboxypeptidase U (TAFIa): a new drug target for fibrinolytic therapy? J Thromb Haemost. 2009;7(12):1962–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Okamoto S, Sato S, Takada Y, Okamoto U. An Active Stereo-Isomer (Trans-Form) of Amcha and Its Antifibrinolytic (Antiplasminic) Action in Vitro and in Vivo. Keio J Med. 1964;13:177–85.

    Article  CAS  PubMed  Google Scholar 

  33. Okamoto SO, Okamoto U. A new potent antifibrinolytic substance and its effects on blood of animals. Keio J Med. 1962;11:105–15.

    Article  CAS  Google Scholar 

  34. Collaborators C-t, Shakur H, Roberts I, Bautista R, Caballero J, Coats T, 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. 2010;376(9734):23–32.

    Article  Google Scholar 

  35. CRASH-3 TC. Effects of tranexamic acid on death, disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): a randomised, placebo-controlled trial. Lancet. 2019;394(10210):1713–23.

    Article  Google Scholar 

  36. Myles PS, Smith JA, Forbes A, Silbert B, Jayarajah M, Painter T, et al. Tranexamic Acid in Patients Undergoing Coronary-Artery Surgery. N Engl J Med. 2017;376(2):136–48.

    Article  CAS  PubMed  Google Scholar 

  37. Collaborators WT. Effect of early tranexamic acid administration on mortality, hysterectomy, and other morbidities in women with post-partum haemorrhage (WOMAN): an international, randomised, double-blind, placebo-controlled trial. Lancet. 2017;389(10084):2105–16.

    Article  Google Scholar 

  38. AusPAR: Tranexamic Acid. In: Care DoHaA, editor.: Therapeutic Goods Administration Australia; 2010.

  39. Pilbrant A, Schannong M, Vessman J. Pharmacokinetics and bioavailability of tranexamic acid. Eur J Clin Pharmacol. 1981;20(1):65–72.

    Article  CAS  PubMed  Google Scholar 

  40. Grassin-Delyle S, Semeraro M, Lamy E, Urien S, Runge I, Foissac F, et al. Pharmacokinetics of tranexamic acid after intravenous, intramuscular, and oral routes: a prospective, randomised, crossover trial in healthy volunteers. Br J Anaesth. 2022;128(3):465–72.

    Article  CAS  PubMed  Google Scholar 

  41. Fillingham YA, Ramkumar DB, Jevsevar DS, Yates AJ, Bini SA, Clarke HD, et al. Tranexamic Acid Use in Total Joint Arthroplasty: The clinical practice guidelines endorsed by the american association of hip and knee surgeons, American society of regional anesthesia and pain medicine, American academy of orthopaedic surgeons, hip society, and knee society. J Arthroplasty. 2018;33(10):3065–9.

    Article  PubMed  Google Scholar 

  42. Zak SG, Tang A, Sharan M, Waren D, Rozell JC, Schwarzkopf R. Tranexamic acid is safe in patients with a history of coronary artery disease undergoing total joint arthroplasty. J Bone Joint Surg Am. 2021;103(10):900–4.

    Article  PubMed  Google Scholar 

  43. Shi J, Zhou C, Pan W, Sun H, Liu S, Feng W, et al. Effect of High- vs Low-Dose Tranexamic acid infusion on need for red blood cell transfusion and adverse events in patients undergoing cardiac surgery: the optimal randomized clinical trial. JAMA. 2022;328(4):336–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Shakur H, Elbourne D, Gülmezoglu M, Alfirevic Z, Ronsmans C, Allen E, et al. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Effects of tranexamic acid on death. disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): a randomised, placebo-controlled trial. The Lancet. 2019;394(10210):1713–23.

    Article  Google Scholar 

  47. Roberts I, Shakur H, Coats T, Hunt B, Balogun E, Barnetson L, et al. The CRASH-2 trial: a randomised controlled trial and economic evaluation of the effects of tranexamic acid on death, vascular occlusive events and transfusion requirement in bleeding trauma patients. Health Technol Assess. 2013;17(10):1–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Longstaff C, Locke M. Increased urokinase and consumption of α2-antiplasmin as an explanation for the loss of benefit of tranexamic acid after treatment delay. J Thromb Haemost. 2019;17(1):195–205.

    Article  CAS  PubMed  Google Scholar 

  49. Investigators PA-T, the ACTG, Gruen RL, Mitra B, Bernard SA, McArthur CJ, et al. Prehospital Tranexamic Acid for Severe Trauma. N Engl J Med. 2023;389(2):127–36.

    Article  Google Scholar 

  50. Collaborators H.-I.T. Effects of a high-dose 24-h infusion of tranexamic acid on death and thromboembolic events in patients with acute gastrointestinal bleeding (HALT-IT): an international randomised, double-blind, placebo-controlled trial. Lancet. 2020;395(10241):1927–36.

  51. Sprigg N, Flaherty K, Appleton JP, Salman RAS, Bereczki D, Beridze M, et al. Tranexamic acid for hyperacute primary IntraCerebral Haemorrhage (TICH-2): an international randomised, placebo-controlled, phase 3 superiority trial. The Lancet. 2018;391(10135):2107–15.

    Article  CAS  Google Scholar 

  52. Meretoja A, Yassi N, Wu TY, Churilov L, Sibolt G, Jeng JS, et al. Tranexamic acid in patients with intracerebral haemorrhage (STOP-AUST): a multicentre, randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2020;19(12):980–7.

    Article  CAS  PubMed  Google Scholar 

  53. Kazui S, Naritomi H, Yamamoto H, Sawada T, Yamaguchi T. Enlargement of spontaneous intracerebral hemorrhage. Incidence and time course Stroke. 1996;27(10):1783–7.

    CAS  PubMed  Google Scholar 

  54. Davis SM, Broderick J, Hennerici M, Brun NC, Diringer MN, Mayer SA, et al. Hematoma growth is a determinant of mortality and poor outcome after intracerebral hemorrhage. Neurology. 2006;66(8):1175–81.

    Article  CAS  PubMed  Google Scholar 

  55. Jiao X, Li M, Li L, Hu X, Guo X, Lu Y. Early Tranexamic Acid in Intracerebral Hemorrhage: A Meta-Analysis of Randomized Controlled Trials. Front Neurol. 2021;12:721125.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wang X, Ma L, Song J, You C. Tranexamic acid for adult patients with spontaneous intracerebral hemorrhage: a systematic review with meta-analysis. CNS Drugs. 2021;35(11):1163–72.

    Article  CAS  PubMed  Google Scholar 

  57. Revesz T, Holton JL, Lashley T, Plant G, Frangione B, Rostagno A, et al. Genetics and molecular pathogenesis of sporadic and hereditary cerebral amyloid angiopathies. Acta Neuropathol. 2009;118(1):115–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wermer MJH, Greenberg SM. The growing clinical spectrum of cerebral amyloid angiopathy. Curr Opin Neurol. 2018;31(1):28–35.

    Article  PubMed  Google Scholar 

  59. Linn J, Halpin A, Demaerel P, Ruhland J, Giese AD, Dichgans M, et al. Prevalence of superficial siderosis in patients with cerebral amyloid angiopathy. Neurology. 2010;74(17):1346–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Charidimou A, Boulouis G, Frosch MP, Baron JC, Pasi M, Albucher JF, et al. The Boston criteria version 2.0 for cerebral amyloid angiopathy: a multicentre, retrospective. MRI-neuropathol diagn accuracy study Lancet Neurol. 2022;21(8):714–25.

    Google Scholar 

  61. Bales KR, O’Neill SM, Pozdnyakov N, Pan F, Caouette D, Pi Y, et al. Passive immunotherapy targeting amyloid-β reduces cerebral amyloid angiopathy and improves vascular reactivity. Brain. 2016;139(Pt 2):563–77.

    Article  PubMed  Google Scholar 

  62. Gervais F, Chalifour R, Garceau D, Kong X, Laurin J, McLaughlin R, et al. Glycosaminoglycan mimetics: a therapeutic approach to cerebral amyloid angiopathy. Amyloid. 2001;8(Suppl 1):28–35.

    CAS  PubMed  Google Scholar 

  63. Wnendt S, Wetzels I, Gunzler WA. Amyloid beta peptides stimulate tissue-type plasminogen activator but not recombinant prourokinase. Thromb Res. 1997;85(3):217–24.

    Article  CAS  PubMed  Google Scholar 

  64. Tucker HM, Kihiko M, Caldwell JN, Wright S, Kawarabayashi T, Price D, et al. The plasmin system is induced by and degrades amyloid-beta aggregates. J Neurosci. 2000;20(11):3937–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mutimer CA, Keragala CB, Markus HS, Werring DJ, Cloud GC, Medcalf RL. Cerebral amyloid angiopathy and the fibrinolytic system: is plasmin a therapeutic target? Stroke. 2021;52(8):2707–14.

    Article  PubMed  Google Scholar 

  66. Seiffge DJ, Polymeris AA, Law ZK, Krishnan K, Zietz A, Thilemann S, et al. Cerebral amyloid angiopathy and the risk of hematoma expansion. Ann Neurol. 2022;92(6):921–30.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Liu Z, McCutcheon FM, Ho H, Chai J, Xiao Y, Tippett I, Keragala CB, Cloud GC, Medcalf RL. Tranexamic acid in a mouse model of cerebral amyloid angiopathy: setting the stage for a novel stroke treatment approach. Research and Practice in Thrombosis and Haemostasis. 2023;9:102166.

    Article  Google Scholar 

  68. Medcalf RL, Keragala CB. Fibrinolysis: a primordial system linked to the immune response. Int J Mol Sci. 2021;22(7):3406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Medcalf RL, Keragala CB. The fibrinolytic system: mysteries and opportunities. Hemasphere. 2021;5(6):e570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chen ZL, Strickland S. Neuronal death in the hippocampus is promoted by plasmin-catalyzed degradation of laminin. Cell. 1997;91(7):917–25.

    Article  CAS  PubMed  Google Scholar 

  71. Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation. 2018;15(1):24.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Zhao XJ, Larkin TM, Lauver MA, Ahmad S, Ducruet AF. Tissue plasminogen activator mediates deleterious complement cascade activation in stroke. PLoS ONE. 2017;12(7):e0180822.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Ploplis VA, French EL, Carmeliet P, Collen D, Plow EF. Plasminogen deficiency differentially affects recruitment of inflammatory cell populations in mice. Blood. 1998;91(6):2005–9.

    Article  CAS  PubMed  Google Scholar 

  74. Miles LA, Ny L, Wilczynska M, Shen Y, Ny T, Parmer RJ. Plasminogen Receptors and Fibrinolysis. Int J Mol Sci. 2021;22(4):1712.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Later AF, Sitniakowsky LS, van Hilten JA, van de Watering L, Brand A, Smit NP, et al. Antifibrinolytics attenuate inflammatory gene expression after cardiac surgery. J Thorac Cardiovasc Surg. 2013;145(6):1611.

    Article  CAS  PubMed  Google Scholar 

  76. Draxler DF, Yep K, Hanafi G, Winton A, Daglas M, Ho H, et al. Tranexamic acid modulates the immune response and reduces postsurgical infection rates. Blood Adv. 2019;3(10):1598–609.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Jimenez JJ, Iribarren JL, Lorente L, Rodriguez JM, Hernandez D, Nassar I, 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(6):R117.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Xie CM, Yao YT, HE LX, Yang K, Evidence In Cardiovascular Anesthesia G. Anti-inflammatory effect of tranexamic acid on adult cardiac surgical patients: A PRISMA-compliant systematic review and meta-analysis. Front Surg. 2022;9:951835.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Lei Y, Liu J, Liang X, Hu N, Pei F, Huang W. Effectiveness of preemptive antifibrinolysis with tranexamic acid in rheumatoid arthritis patients undergoing total knee arthroplasty: a study protocol for a randomized controlled trial. BMC Musculoskelet Disord. 2020;21(1):465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Barrett CD, Vigneshwar N, Moore HB, Ghasabyan A, Chandler J, Moore EE, et al. Tranexamic acid is associated with reduced complement activation in trauma patients with hemorrhagic shock and hyperfibrinolysis on thromboelastography. Blood Coagul Fibrinolysis. 2020;31(8):578–82.

    Article  CAS  PubMed  Google Scholar 

  81. Benjumea A, Diaz-Navarro M, Hafian R, Sanchez-Somolinos M, Vaquero J, Chana F, et al. Effect of Tranexamic Acid against Staphylococcus spp. and Cutibacterium acnes Associated with Peri-Implant Infection: Results from an In Vitro Study. Microbiol Spectr. 2022;10(1):e0161221.

    Article  PubMed  Google Scholar 

  82. Benjumea A, Diaz-Navarro M, Hafian R, Cercenado E, Sanchez-Somolinos M, Vaquero J, et al. Tranexamic acid in combination with vancomycin or gentamicin has a synergistic effect against staphylococci. Front Microbiol. 2022;13:935646.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Kacer D, Machnitzky E, Fung A, Greene A, Carter D, Rappold J, et al. Anti-fibrinolytic agent tranexamic acid suppresses the endotoxin-induced expression of Tnfalpha and Il1alpha genes in a plasmin-independent manner. Transfusion (Paris). 2023;63(Suppl 3):S168–76.

    CAS  Google Scholar 

  84. Guo Y, Li J, Hagstrom E, Ny T. Beneficial and detrimental effects of plasmin(ogen) during infection and sepsis in mice. PLoS ONE. 2011;6(9):e24774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Vago JP, Zaidan I, Perucci LO, Brito LF, Teixeira LC, Silva CMS, et al. Plasmin and plasminogen prevent sepsis severity by reducing neutrophil extracellular traps and systemic inflammation. JCI Insight. 2023;8(8):e166044.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Romer J, Bugge TH, Pyke C, Lund LR, Flick MJ, Degen JL, et al. Impaired wound healing in mice with a disrupted plasminogen gene. Nat Med. 1996;2(3):287–92.

    Article  CAS  PubMed  Google Scholar 

  87. Sulniute R, Shen Y, Guo YZ, Fallah M, Ahlskog N, Ny L, et al. Plasminogen is a critical regulator of cutaneous wound healing. Thromb Haemost. 2016;115(5):1001–9.

    Article  PubMed  Google Scholar 

  88. Fallah M, Viklund E, Backman A, Broden J, Lundskog B, Johansson M, et al. Plasminogen is a master regulator and a potential drug candidate for the healing of radiation wounds. Cell Death Dis. 2020;11(3):201.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Yalamanchili S, Frankel AS, Talei B, Azizzadeh B, Auersvald A. Wound healing complications with tranexamic acid: not the silver bullet after all. Aesthet Surg J. 2023.

  90. Tse TW, Hui E. Tranexamic acid: an important adjuvant in the treatment of melasma. J Cosmet Dermatol. 2013;12(1):57–66.

    Article  PubMed  Google Scholar 

  91. Forbat E, Al-Niaimi F, Ali FR. The emerging importance of tranexamic acid in dermatology. Clin Exp Dermatol. 2020;45(4):445–9.

    Article  CAS  PubMed  Google Scholar 

  92. Kim HJ, Moon SH, Cho SH, Lee JD, Kim HS. Efficacy and safety of tranexamic acid in melasma: a meta-analysis and systematic review. Acta Derm Venereol. 2017;97(7):776–81.

    Article  CAS  PubMed  Google Scholar 

  93. Maeda K, Naganuma M. Topical trans-4-aminomethylcyclohexanecarboxylic acid prevents ultraviolet radiation-induced pigmentation. J Photochem Photobiol B. 1998;47(2–3):136–41.

    Article  CAS  PubMed  Google Scholar 

  94. Cho YH, Park JE, Lim DS, Lee JS. Tranexamic acid inhibits melanogenesis by activating the autophagy system in cultured melanoma cells. J Dermatol Sci. 2017;88(1):96–102.

    Article  CAS  PubMed  Google Scholar 

  95. Zhu J-W, Ni Y-J, Tong X-Y, Guo X, Wu X-P, Lu Z-F. Tranexamic Acid Inhibits Angiogenesis and Melanogenesis <i>in Vitro</i> by Targeting VEGF Receptors. Int J Med Sci. 2020;17(7):903–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Maas C. Plasminflammation-an emerging pathway to bradykinin production. Front Immunol. 2019;10:2046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Busse PJ, Christiansen SC. Hereditary Angioedema. N Engl J Med. 2020;382(12):1136–48.

    Article  CAS  PubMed  Google Scholar 

  98. Bork K, Wulff K, Witzke G, Hardt J. Treatment for hereditary angioedema with normal C1-INH and specific mutations in the F12 gene (HAE-FXII). Allergy. 2017;72(2):320–4.

    Article  CAS  PubMed  Google Scholar 

  99. Bork K, Wulff K, Witzke G, Hardt J. Hereditary angioedema with normal C1-INH with versus without specific F12 gene mutations. Allergy. 2015;70(8):1004–12.

    Article  CAS  PubMed  Google Scholar 

  100. Diebel ME, Martin JV, Liberati DM, Diebel LN. The temporal response and mechanism of action of tranexamic acid in endothelial glycocalyx degradation. J Trauma Acute Care Surg. 2018;84(1):75–80.

    Article  CAS  PubMed  Google Scholar 

  101. Prudovsky I, Carter D, Kacer D, Palmeri M, Soul T, Kumpel C, et al. Tranexamic acid suppresses the release of mitochondrial DNA, protects the endothelial monolayer and enhances oxidative phosphorylation. J Cell Physiol. 2019;234(11):19121–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Eltania F, Lesmana R, Sudigdoadi S, Sudigdoadi S, Khairani AF, Goenawan H, et al. Tranexamic Acid Cream Protects Ultraviolet B-induced Photoaging in Balb/c Mice Skin by Increasing Mitochondrial Markers: Changes Lead to Improvement of Histological Appearance. Photochem Photobiol. 2020;96(4):863–9.

    Article  CAS  PubMed  Google Scholar 

  103. Sandler N, Ho H, Draxler DF, Bain CR, Smith JA, Hauser CJ, et al. Characterisation of Plasma Mitochondrial DNA, MMP-9 and Neutrophil Elastase in Patients Undergoing Coronary Artery Bypass Grafting: Effects of Tranexamic Acid and Postoperative Pneumonia. Heart Lung Circ. 2022;31(3):439–46.

    Article  PubMed  Google Scholar 

  104. Hiramoto K, Sugiyama D, Takahashi Y, Mafune E. The amelioration effect of tranexamic acid in wrinkles induced by skin dryness. Biomed Pharmacother. 2016;80:16–22.

    Article  CAS  PubMed  Google Scholar 

  105. Hiramoto K, Yamate Y, Sugiyama D, Matsuda K, Iizuka Y, Yamaguchi T. Effect of tranexamic acid in improving the lifespan of naturally aging mice. Inflammopharmacology. 2019;27(6):1319–23.

    Article  CAS  PubMed  Google Scholar 

  106. Hiramoto K, Oikawa H, Yamate Y, Sato EF. Tranexamic Acid Protects Ovary and Testis Functions and Ameliorates Osteoporosis in Mice. Pharmacology. 2020;105(11–12):652–61.

    Article  CAS  PubMed  Google Scholar 

  107. A Feasibility Trial of Tranexamic Acid for Necrotising Soft-tissue Infections. Available from: https://anzctr.org.au/Trial/Registration/TrialReview.aspx?id=382739&isReview=true.

  108. TRIGS trial: Tranexamic acid to Reduce Infection after Gastrointestinal Surgery. Available from: https://www.anzca.edu.au/profiles/ctn-trials/underway/trigs-trial.

  109. Picetti R, Shakur-Still H, Medcalf RL, Standing JF, Roberts I. What concentration of tranexamic acid is needed to inhibit fibrinolysis?. A systematic review of pharmacodynamics studies. Blood Coagul Fibrinolysis. 2019;30(1):1–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hodgson S, Larvin JT, Dearman C. What dose of tranexamic acid is most effective and safe for adult patients undergoing cardiac surgery? Interact Cardiovasc Thorac Surg. 2015;21(3):384–8.

    Article  PubMed  Google Scholar 

  111. Guyette FX, Brown JB, Zenati MS, Early-Young BJ, Adams PW, Eastridge BJ, et al. Tranexamic Acid During Prehospital Transport in Patients at Risk for Hemorrhage After Injury: A Double-blind, Placebo-Controlled. Randomized Clinical Trial JAMA Surgery. 2021;156(1):11–20.

    Google Scholar 

  112. Wang WJ, Wu TY, Tu YK, Kuo KL, Tsai CY, Chie WC. The optimal dose of oral tranexamic acid in melasma: A network meta-analysis. Indian J Dermatol Venereol Leprol. 2023;89(2):189–94.

    Article  PubMed  Google Scholar 

  113. Zhang S, Xu H, Xie J, Cao G, Lei Y, Pei F. Tranexamic acid attenuates inflammatory effect and modulates immune response in primary total knee arthroplasty: a randomized, placebo-controlled, pilot trial. Inflammopharmacology. 2020;28(4):839–49.

    Article  CAS  PubMed  Google Scholar 

  114. Changjun C, Xin Z, Yue L, Chengcheng Z, Qiuru W, Qianhao L, et al. Tranexamic acid attenuates early post-operative systemic inflammatory response and nutritional loss and avoids reduction of fibrinogen in total hip arthroplasty within an enhanced recovery after surgery pathway. Int Orthop. 2021;45(11):2811–8.

    Article  PubMed  Google Scholar 

  115. Wang D, Luo ZY, Yu ZP, Liu LX, Chen C, Meng WK, et al. The antifibrinolytic and anti-inflammatory effects of multiple doses of oral tranexamic acid in total knee arthroplasty patients: a randomized controlled trial. J Thromb Haemost. 2018;16(12):2442–53.

    Article  CAS  PubMed  Google Scholar 

  116. Lei Y-T, Xie J-W, Huang Q, Huang W, Pei F-X. The antifibrinolytic and anti-inflammatory effects of a high initial-dose tranexamic acid in total knee arthroplasty: a randomized controlled trial. Int Orthop. 2020;44(3):477–86.

    Article  PubMed  Google Scholar 

  117. Spinella PC, Thomas KA, Turnbull IR, Fuchs A, Bochicchio K, Schuerer D, et al. The immunologic effect of early intravenous two and four gram bolus dosing of tranexamic acid compared to placebo in patients with severe traumatic bleeding (TAMPITI): a randomized, double-blind, placebo-controlled. Single-Center Trial Front Immunol. 2020;11:2085.

    Article  CAS  PubMed  Google Scholar 

  118. Fillingham YA, Ramkumar DB, Jevsevar DS, Yates AJ, Shores P, Mullen K, et al. The efficacy of tranexamic acid in total hip arthroplasty: a network meta-analysis. J Arthroplasty. 2018;33(10):3083-9 e4.

    Article  PubMed  Google Scholar 

  119. Fillingham YA, Ramkumar DB, Jevsevar DS, Yates AJ, Shores P, Mullen K, et al. The efficacy of tranexamic acid in total knee arthroplasty: a network meta-analysis. J Arthroplasty. 2018;33(10):3090-8 e1.

    Article  PubMed  Google Scholar 

  120. Collaborators C-t. Effects of tranexamic acid on death, disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): a randomised, placebo-controlled trial. Lancet. 2019;394(10210):1713–23.

    Article  Google Scholar 

  121. Zaib A, Shaheryar M, Shakil M, Sarfraz A, Sarfraz Z, Cherrez-Ojeda I. Local Tranexamic acid for preventing hemorrhage in anticoagulated patients undergoing dental and minor oral procedures: a systematic review and meta-analysis. Healthcare (Basel). 2022;10(12):2523.

    Article  PubMed  Google Scholar 

  122. van Galen KP, Engelen ET, Mauser-Bunschoten EP, van Es RJ, Schutgens RE. Antifibrinolytic therapy for preventing oral bleeding in patients with haemophilia or Von Willebrand disease undergoing minor oral surgery or dental extractions. Cochrane Database Syst Rev. 2019;4(4):CD011385.

    PubMed  Google Scholar 

  123. Gaillard S, Dupuis-Girod S, Boutitie F, Riviere S, Moriniere S, Hatron PY, et al. Tranexamic acid for epistaxis in hereditary hemorrhagic telangiectasia patients: a European cross-over controlled trial in a rare disease. J Thromb Haemost. 2014;12(9):1494–502.

    Article  CAS  PubMed  Google Scholar 

  124. Collaborators H-IT. Effects of a high-dose 24-h infusion of tranexamic acid on death and thromboembolic events in patients with acute gastrointestinal bleeding (HALT-IT): an international randomised, double-blind, placebo-controlled trial. Lancet. 2020;395(10241):1927–36.

    Article  Google Scholar 

  125. Liu J, Nie X, Gu H, Zhou Q, Sun H, Tan Y, et al. Tranexamic acid for acute intracerebral haemorrhage growth based on imaging assessment (TRAIGE): a multicentre, randomised, placebo-controlled trial. Stroke Vasc Neurol. 2021;6(2):160–9.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Sprigg N, Flaherty K, Appleton JP, Al-Shahi Salman R, Bereczki D, Beridze M, et al. Tranexamic acid for hyperacute primary IntraCerebral Haemorrhage (TICH-2): an international randomised, placebo-controlled, phase 3 superiority trial. Lancet. 2018;391(10135):2107–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Gernsheimer TB, Brown SP, Triulzi DJ, Key NS, El Kassar N, Herren H, et al. Prophylactic tranexamic acid in patients with hematologic malignancy: a placebo-controlled, randomized clinical trial. Blood. 2022;140(11):1254–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fay A, Abinun M. Current management of hereditary angio-oedema (C’1 esterase inhibitor deficiency). J Clin Pathol. 2002;55(4):266–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

NA.

Funding

R.L.M is supported by a research grant awarded by the National Health and Medical Research Council (NHMRC) of Australia, grant ID 1156506. C.B.K was supported by an Australian Post-graduate research training scholarship. 

Author information

Authors and Affiliations

Authors

Contributions

T.L. contributed to manuscript conceptualisation, writing and editing. T.L also constructed the figure. R.M. contributed to manuscript conceptualisation, writing and editing. G.C. contributed to manuscript review and editing. P.M. contributed to manuscript review and editing. C.K. contributed to manuscript conceptualisation, figure development, writing and editing. C.K. also constructed the table.

Corresponding author

Correspondence to Charithani B. Keragala.

Ethics declarations

Ethics approval and consent to participate

NA.

Consent for publication

NA.

Competing interests

The corresponding author, Dr. Charithani Keragala is an associate editor of Thrombosis Journal. Prof. Robert Medcalf is co-Editor-in-Chief of Thrombosis Journal.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lam, T., Medcalf, R.L., Cloud, G.C. et al. Tranexamic acid for haemostasis and beyond: does dose matter?. Thrombosis J 21, 94 (2023). https://doi.org/10.1186/s12959-023-00540-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12959-023-00540-0

Keywords