You are on page 1of 24

Coagulopathy associated with trauma Page 1 of 24

Official reprint from UpToDate®


www.uptodate.com ©2013 UpToDate®

Coagulopathy associated with trauma

Authors Section Editors Deputy Editor


Mitchell Cohen, MD, FACS Heidi L Frankel, MD, FACS Kathryn A Collins, MD, PhD, FACS
Matthew E Kutcher, MD Lawrence LK Leung, MD

Disclosures

All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: Jul 2013. | This topic last updated: Dec 10, 2012.

INTRODUCTION — Trauma remains a leading cause of death and disability in adults in spite of advances in resuscitation, surgical
management, and critical care [1]. Between 25 to 35 percent of injured civilian trauma patients develop a biochemically evident coagulopathy
upon arrival in the emergency department, in spite of improved efficiency of trauma systems, military and civilian, in reducing the time interval
between acute injury and treatment [2-4]. Coagulopathy may be the result of physiologic derangements such as acidosis, hypothermia, or
hemodilution related to fluid or blood administration; however, an acute coagulopathy can also occur in severely injured patients independent
of, or in addition to, these factors [2,3]. Several terms are used in the literature to refer to this condition, including acute traumatic
coagulopathy (ATC), early coagulopathy of trauma (ECT), trauma-induced coagulopathy, and the acute coagulopathy of trauma-shock
(ACoTS) [2,3,5,6].

The etiology, diagnosis and treatment of coagulopathy associated with trauma will be reviewed here. The general principles of shock
management in the trauma patient and the treatment of excessive anticoagulation related to medical treatment are discussed elsewhere.
(See "Correcting excess anticoagulation after warfarin" and "Initial evaluation and management of shock in adult trauma".)

IMPACT — Coagulopathy in trauma patients, and specifically acute traumatic coagulopathy (ATC), is associated with higher transfusion
requirements, longer intensive care unit and hospital stays, more days requiring mechanical ventilation, and a greater incidence of multiorgan
dysfunction. Compared with patients who do not have coagulopathy, those with coagulopathy have a threefold to fourfold greater mortality,
and are up to eight times more likely to die within the first 24 hours following injury [2-4,7,8].

Injury to brain tissue may predispose to acute traumatic coagulopathy and about one-third of patients with traumatic brain injury (TBI) have a
coagulopathy.

ETIOLOGY — The etiology of coagulopathy in the injured patient is multifactorial with overlapping contributions depending upon the injury
and nature of resuscitation. Normal coagulation is a balance between hemostatic and fibrinolytic processes which permit control of bleeding
following mild injury while preventing inappropriate intravascular thrombosis. (See "Overview of hemostasis".)

Etiologies that upset this balance include the classic elements of the ‘vicious triad’ which includes acidosis related to tissue injury and shock,
hypothermia from exposure and fluid administration, and hemodilution due to fluid or component blood product administration. Systemic
consumption of clotting factors manifesting as disseminated intravascular coagulation (DIC) may occur early after injury due to inadequate
clotting factor repletion in the face of ongoing consumption, or later in the hospital course triggered by secondary insults (eg, sepsis).
Frequently complicating these etiologies but mechanistically different, acute traumatic coagulopathy (ATC) is a biochemical response to
injury and shock leading to hyperfibrinolysis and hypocoagulability.

Acidosis — Inadequate tissue perfusion in patients with hypovolemic shock due to bleeding leads to metabolic (lactic) acidosis, which can
be exacerbated by excessive chloride and component blood administration. (See "Shock in adults: Types, presentation, and diagnostic
approach", section on 'Hypovolemic shock'.)

Acidosis causes demonstrable clotting dysfunction in experimental models at pH<7.2 by interfering with the assembly of coagulation factor
complexes involving calcium and negatively-charged phospholipids [9-12]. As an example, the activity of the
factor Xa/Va/phospholipid/prothrombin (“prothrombinase”) complex is reduced by 50, 70, and 90 percent at a pH of 7.2, 7.0, and 6.8,
respectively. However, correction of acidosis alone does not always correct the associated coagulopathy, indicating that tissue injury causes
coagulopathy via additional mechanisms [13,14].

Hypothermia — Hypothermia in injured patients is graded into mild (36 to 34ºC), moderate (34 to 32ºC), and severe (<32ºC) [15]. Nearly two-
thirds of trauma patients have a temperature below 36ºC on presentation; 9 percent of trauma patients have a temperature at or below 33ºC
[11,16-19].

Hypothermia following injury is due to cold exposure at the time of injury, during transport, and during the trauma examination compounded
by the administration of cold intravenous fluids. Patients who require surgery are at a greater risk for hypothermia due to further physical
exposure in the operating room, additional fluid administration, and the effects of general anesthesia. Injured patients with hypothermia
generally have worse outcomes compared with non-injured patients with hypothermia; however, hypothermia alone is a weak independent
predictor of mortality [15,20]. Acidosis and hypothermia are synergistic with increased mortality when both are present, compared with one or
the other [21].

The effect of hypothermia on clotting includes platelet dysfunction and impaired enzymatic function. Overall thrombin generation in activated
in vitro clotting systems is generally preserved at a temperature of 33ºC; however, impairment of tissue factor activity, platelet aggregation,
and platelet adhesion are evident at temperatures between 33 to 37ºC [11,16]. It is important to note that no effects on standard coagulation
tests are seen in hypothermia-induced coagulopathy without special sample handling due to the standard practice of pre-warming blood
samples to 37ºC prior to analysis, which corrects the defect.

Specific measures to correct hypothermia include controlling physical exposure, the administration of warmed fluids, and passive rewarming
with blankets and forced-air devices. Rapid identification and control of bleeding is vital to preserve normal temperature. Continuous

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 2 of 24

temperature monitoring is essential to ensure that mild hypothermia does not worsen. In the case of moderate or severe hypothermia and
coagulopathy, central rewarming may be needed.

Resuscitation-associated (dilutional) coagulopathy — Resuscitation-associated coagulopathy (RAC) refers to alterations of the


coagulation system induced by large volumes of fluid or unbalanced component blood administration during the management of shock [22].
Trauma resuscitation historically focused on the treatment of hypotension and acidosis with aggressive crystalloid resuscitation followed by
packed red blood cells (PRBC). At that time, treatment of coagulopathy was initiated only in response to abnormal standard coagulation
tests. Similarly, platelet transfusion was generally not performed until laboratory evidence of thrombocytopenia was present.

Computer modeling [23], in vitro experiments [24], and clinical studies in healthy volunteers have found that large volume resuscitation with
crystalloid, colloid, and packed red blood cells leads to dilution of plasma clotting proteins [25]. A retrospective study of 8724 injured patients
found a positive correlation between prehospital fluid resuscitation volume and coagulopathy [4]. Coagulopathy was present on admission in
more than 50 percent of patients who received >3L of intravenous fluid prior to arrival, but coagulopathy was also present in 10 percent of
patients administered <500 mL and in 32 patients who had received no prehospital fluids, which appears to reflect the frequently overlapping
contribution of acute traumatic coagulopathy (ATC). (See 'Acute traumatic coagulopathy' below.)

Another factor contributing to resuscitation associated coagulopathy is the effect of storage time on packed red blood cells which undergo
progressive functional and structural changes over time. The ‘storage lesion’ includes decreased pH, chelation of calcium, low 2,3
diphosphoglycerate levels, and decreased clotting factor concentration. Transfusion of older blood can further impair microvascular
perfusion, and has inflammatory and immunomodulatory effects [26-28]. (See "Red blood cell transfusion in adults: Storage, specialized
modifications, and infusion parameters", section on 'Effect of storage conditions on transfusion outcome'.)

The median duration of storage of a unit of red blood cells in the United States is about 15 days, which is relatively old, with even older units
frequently allocated to high-use facilities such as trauma centers [29]. Thus, the storage lesion is particularly relevant for massively
transfused trauma patients. Several studies suggest that an older storage age of transfused PRBCs may be linked to higher morbidity and
mortality [30,31] but this remains an area of controversy and current investigation [32].

Disseminated intravascular coagulation — Disseminated intravascular coagulation (DIC) is a systemic process producing consumptive
coagulopathy in concert with diffuse microvascular thrombosis (table 1). In trauma patients, tissue-injury-induced exposure of tissue factor
and activation of the extrinsic coagulation cascade leads to thrombin generation proportional to injury severity. In addition, systemic
embolism of tissue-specific thromboplastins from sites of injury (including bone marrow lipid material, amniotic fluid, and brain phospholipids)
may predispose patients to DIC [33]. (See "Pathogenesis and etiology of disseminated intravascular coagulation".)

Acute traumatic coagulopathy — Acute traumatic coagulopathy (ATC) is an impairment of hemostasis and activation of fibrinolysis that
occurs early after injury and is biochemically evident prior to, and independent of, the development of significant acidosis, hypothermia, or
hemodilution. The risk of ATC increases with hypotension, higher injury severity scores, worsening base deficit, and head injury [3,7,8]. Once
established, ATC is often compounded by other etiologies. (See 'Acidosis' above and 'Hypothermia' above and 'Resuscitation-associated
(dilutional) coagulopathy' above.)

Patients with ATC frequently meet criteria for DIC and some authors have argued that acute traumatic coagulopathy (ATC) may represent an
early, partially-compensated stage of DIC [22,34,35]. However, the concept of DIC as a final common pathway for several different
phenomena is insufficient to explain the hematologic abnormalities post-injury [7,36,37]. Coagulopathy in the absence of thrombocytopenia
and hypofibrinogenemia, as seen in ATC, argues against consumption as a necessary underlying mechanism [13] Although D-dimer levels
are frequently elevated and fibrinogen levels depleted in acutely injured patients, indicating intravascular fibrin deposition and active
fibrinolysis [38], functional thrombin generation (assayed by the presence of prothrombin fragments and thrombin-antithrombin complexes)
remains intact [7,39-41]. Furthermore, ATC occurs only when tissue injury is combined with systemic hypoperfusion. Thus, it is most likely
that ATC is mechanistically distinct from DIC but that these frequently overlap. Exploring this distinction is an area of ongoing research.
(See 'Disseminated intravascular coagulation' above and 'Diagnosis' below.)

Mechanism — Initial observations in hypoperfused trauma patients found a correlation between acute traumatic coagulopathy (ATC) and
elevated levels of activated protein C (aPC), reduced levels of non-activated protein C, and elevated soluble thrombomodulin [7]. Activation
of the thrombomodulin-protein C system is a principle pathway mediating ATC, a mechanism that is distinct from clotting factor consumption
or dysfunction [7,42].

Under normal circumstances, tissue injury leads to thrombin generation, fibrin deposition, and clot formation via the extrinsic pathway (figure
1). The enzymatic pathways making up the coagulation cascade are discussed in detail elsewhere. (See "Overview of hemostasis".)

Initiation of the clotting process is localized to the site of tissue injury. Systemic coagulation due to the escape of thrombin from the injury site
is inhibited by circulating antithrombin III, or by the binding of thrombin to constitutively-expressed thrombomodulin on nearby undamaged
endothelial cells [43].

Protein C, a systemic anticoagulant, is proteolytically converted from an inactive zymogen to activated protein C (aPC) by the complex of
thrombin with thrombomodulin. Activated protein C is a serine protease that proteolytically inactivates factors Va and VIIIa and depletes
plasminogen inhibitors (figure 2 and table 2) [42,44]. In this manner, aPC can serve a protective function by inhibiting thrombosis during
periods of decreased flow.

Sustained hypoperfusion is associated with increased circulating soluble thrombomodulin levels, which can increase the availability of
thrombomodulin-bound thrombin [7]. Thus, thrombin can be diverted from a predominantly procoagulant role to a pathologic, anticoagulant
role via excess activation of protein C [7]. Although the precise biochemical mechanisms are still under investigation, the importance of
protein C in this process has been confirmed. Inhibition of protein C by an antibody-mediated mechanism in an animal model has been
shown to prevent the development of ATC in response to trauma and hemorrhagic shock [42].

With widespread protein C activation, thrombin generation is inhibited, impairing clot formation, and fibrinolysis is enhanced causing
degradation of existing clot. Consumption of endogenous plasminogen activator inhibitor-1 (PAI-1) by ATC-mediated aPC destabilizes the
fibrinolytic balance, leading to uninhibited tissue plasminogen activator (tPA)-mediated conversion of plasminogen to plasmin [45]. Diversion
of thrombin to protein C activation may also reduce activation of thrombin-activatable fibrinolysis inhibitor (TAFI), further enhancing fibrinolytic
activity [46]. These mechanisms lead to the hyperfibrinolytic state seen in trauma patients with ATC, which is reflected in increased levels of

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 3 of 24

tissue plasminogen activator (tPA), decreased plasminogen activator inhibitor (PAI-1), and increased D-dimer [7].

Activated protein C also has antiinflammatory and cytoprotective effects. Profound activation and consumption of protein C can deplete
protein C stores, potentially leading to infectious and later thrombotic sequelae. These effects are mediated via binding of aPC to a protease-
activated receptor-1 (PAR-1) and endothelial protein C receptor (EPCR) that are likely independent of the role of aPC as an anticoagulant
[47]. The importance of these mechanisms is suggested by separate murine models in trauma and sepsis. In these experiments, selective
antibody-mediated inhibition of the anticoagulant function of aPC reduced the rate of coagulopathy, but not mortality, while inhibition of
anticoagulant and cytoprotective functions increased mortality after a challenge with trauma/hemorrhagic shock [42], or injection of
lipopolysaccharide (LPS) [48].

These animal model results are supported by a recent study of 203 critically-injured trauma patients, for whom early coagulopathy was linked
to high levels of activated protein C, and later, protein C depletion as early as six hours after injury [49]. Patients who demonstrated protein C
depletion had a significantly increased risk of acute lung injury, ventilator-associated pneumonia, multiorgan failure, and death. A smaller
prospective study that also identified protein C depletion in trauma patients found elevated markers of endothelial injury and coagulopathy,
and noted a threefold higher risk of mortality [50].

Cytoprotective functions of aPC may also play a role in pulmonary capillary endothelial barrier function as suggested by in vitro studies
[51,52], and in human studies that found that persistently low protein C levels in critically-injured, mechanically-ventilated trauma patients are
associated with increased rates of pneumonia [53]. Ongoing studies are evaluating the interactions of the protein C system, innate [54,55]
and cellular immunity [56,57], and endothelial activation and barrier permeability [52,58-60].

The recently identified importance of ATC-associated depletion of protein C stores after traumatic injury suggests a potential mechanism for
later hypercoagulability and risk of thromboembolic complications after trauma [49,61,62]. Although this phenomenon has not been studied in
the trauma population, a prothrombotic state associated with protein C depletion has been reported in septic patients [44]. In one
observational study, injured patients with coagulopathy on admission had a significantly higher risk of thromboembolic complications, but a
link to protein C depletion was not specifically investigated [63]. However, the potential link between protein C depletion and late
hypercoagulability after trauma requires further investigation. Despite initial trials demonstrating the efficacy of recombinant aPC
supplementation in sepsis, the recent PROWESS-SHOCK trial failed to show a survival benefit in severe sepsis, and the drug was voluntarily
withdrawn from the market [64,65]; as such, there is no role for protein C replacement in trauma.

DIAGNOSIS — Severely injured trauma patients are routinely screened with standard laboratory evaluation, including complete blood count,
serum electrolytes, arterial blood gas analysis, and standard coagulation tests. These laboratory studies provide an assessment of acidosis
and hemodilution, indicate the severity of shock (as measured by base deficit and/or lactate), guide specific component blood product
administration, and serve as a baseline for the assessment of ongoing hemorrhage. (See "Initial evaluation and management of shock in
adult trauma", section on 'Evaluation and management'.)

Readily obtainable coagulation tests (prothrombin time, international normalized ratio, and activated partial thromboplastin time) are the
current standard for establishing a definitive diagnosis of coagulopathy. Fibrinogen and D-dimer levels are also available in most clinical
laboratories, and should be drawn in patients with evidence of hemorrhage or shock since these are surrogate markers of clotting factor
consumption and hyperfibrinolysis. (See "Clinical use of coagulation tests".)

Although these assays are commonly relied upon to evaluate bleeding risk in trauma patients, these tests were originally designed to screen
for heritable coagulopathies such as hemophilia, and subsequently used to monitor anticoagulant therapy [66]. The normal ranges are
derived from the general population and correlate poorly with bleeding risk in elective general and vascular surgical operations [67].
Additionally, the results from standard laboratory analysis can take up to 30 minutes and may not accurately reflect the patient’s evolving
coagulation status; the delay in the availability of results may make them irrelevant in the face of exsanguinating hemorrhage. As a result,
there has been a trend toward the use of point-of-care laboratory testing (thromboelastography) and clinical scoring systems to guide
management of the severely injured patient. (See 'Thromboelastography' below.)

Patients with prolonged clotting times due to known bleeding diatheses or pharmacological anticoagulation are not traditionally defined as
having ATC, although ATC physiology may exacerbate any preexisting coagulation disorders. (See "Approach to the adult patient with a
bleeding diathesis" and "Correcting excess anticoagulation after warfarin".)

Standard coagulation tests — Clinical studies have used several diagnostic criteria to identify clinically relevant coagulopathy following
injury. These include prothrombin time (PT)>18 seconds [39], international normalized ratio (INR)>1.5 [68], activated partial thromboplastin
time (PTT)>60 seconds [39], or any of these values at a threshold of 1.5 times the laboratory reference value [69]. The prevalence of
prolonged PT is higher, but prolongation of the PTT is more specific.

 In a trauma registry study involving 20,103 patients, the PT and PTT were prolonged in 28 and 8 percent of patients, respectively [3].
The adjusted odds ratios for mortality were 1.35 for PT and 4.26 for PTT prolongation.

 A five-center international retrospective study of 3646 trauma patients identified patients with significantly greater transfusion
requirements and increased mortality using a more liberal cutoff >1.2 of prothrombin time ratio (an inter-center standardized version of
the INR) [70]. This lower INR value may be a more appropriate cutoff for patients with more severe injury (injury severity scale [ISS]
>15) and shock.

Decreased platelet count (100,000/µL) and decreased platelet function also contribute to coagulopathy and poor outcome following trauma
[71]. Little information about platelet function is evident from the platelet count alone. In one study, serial functional analyses demonstrated
increased platelet activation and function in survivors following traumatic injury, while non-survivors had significant dysfunction [72].
Instrumentation available to assess platelet function includes the platelet function analyzer (PFA-100), and the electrical impedance whole
blood aggregometer (Multiplate®) [73,74]. However, these devices have not been clinically evaluated in trauma and resuscitation.
(See "Platelet function testing".)

It is unknown whether ATC might exist in patients with normal-range values of PT/INR and PTT. A careful study of markers of coagulopathy
and inflammation in 80 trauma patients identified that increasing injury severity correlated with elevated markers of endothelial cell and
glycocalyx damage, protein C activation, and clotting factor consumption even when INR and PTT values were in the normal range,
suggesting that the biochemical derangement of ATC lies on a continuum dependent upon injury and shock severity [41]. Similarly, there

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 4 of 24

have been anecdotal reports of injured patients with clinically relevant hyperfibrinolysis identified using thromboelastography, but without
prolonged PT or PTT. Thus, patients who present with the combination of significant injury (ISS>15) and a base deficit (<-6) should be
closely monitored and aggressively treated for clinical coagulopathy even in the face of normal standard coagulation tests, pending further
clinical studies.

Because ATC can occur in patients who have normal platelet and fibrinogen levels, specific platelet or fibrinogen levels are not included in
current diagnostic criteria; however, when abnormalities are present, thrombocytopenia and hypofibrinogenemia certainly contribute to
clinical hemorrhage and should be corrected [2,3,7]. (See 'Treatment' below.)

Thromboelastography — Thromboelastography assesses the viscoelastic properties of clot formation in fresh or citrated whole blood in real-
time. The test synthesizes information obtained from multiple coagulation tests (PT, PTT, thrombin time, fibrinogen level, and platelet count)
into a single read out (figure 3) providing information regarding clot initiation, clot strength, and fibrinolysis simultaneously. Until recently, the
bulk of its clinical use has been as a point-of-care adjunct during cardiopulmonary bypass [75] and liver transplantation [76]. As a functional
test of clot formation and lysis, it is conceptually well-suited to monitor the progress or resolution of coagulopathy after injury.

Thromboelastography parameters have been validated against standard laboratory tests [77,78], thrombin-antithrombin complex levels for
TEG® [79], and euglobin clot lysis times for RoTEM® [80]. Specific elements of the clotting cascade can be interrogated by performing tests
in the presence of specific clotting activators or inhibitors (table 3). (See "Platelet function testing", section on 'Thromboelastography'.)

Representative tracings compared with normal are given for each of the following abnormalities:

 Primary fibrinolysis – (figure 4A-B)


 Secondary hyperfibrinolysis – (figure 4A, 4C)
 Thrombocytopenia – (figure 4A, 4D)
 Clotting factor consumption – (figure 4A, 4E)
 Hypercoagulability – (figure 4A, 4F)

Multiple studies have used thromboelastography to diagnose immediate hypocoagulability and later hypercoagulability following moderate
injury despite normal-range standard coagulation tests [81,82]. Studies involving trauma patients have correlated thromboelastography
parameters with increased mortality [83-86]. In one of these studies, the mortality rate for a group of patients who demonstrated
hyperfibrinolysis was significantly greater (77 versus 41 percent) compared with the group that did not demonstrate hyperfibrinolysis [85]. Cut-
off values for RoTEM®-based parameters correlate with standard laboratory transfusion cut-off values [87]. The use of thromboelastography
in trauma as a guide to transfusion is discussed below. (See 'Thromboelastography-based transfusion' below.)

Factor levels — Although coagulation factors are not commonly assessed in injured patients, coagulation factor depletion due to
hemodilution and unbalanced component blood transfusion exacerbates coagulopathy associated with trauma. Fibrinogen (factor I) is the
first factor to become depleted [88]. Of the commonly numbered coagulation factors, V and VIII are the most labile and may become
selectively depleted during trauma resuscitation, particularly in the setting of low plasma to red blood cell unit transfusion ratios.

Clinical scoring systems — Rapid clinical assessment of the trauma patient provides information that helps predict the potential for
coagulopathy and empiric need for massive transfusion based upon the mechanism and severity of injuries, hemodynamic status of the
patient, and evidence of active hemorrhage (eg, positive focused assessment with sonography for trauma [FAST], or brisk bleeding from a
chest tube) [89-91].

Several clinical scoring systems have been evaluated for this purpose, but are not widely used. These include the Trauma-Associated
Severe Hemorrhage (TASH) score [91], McLaughlin score [92], and Assessment of Blood Consumption (ABC) score [89]. A retrospective
review compared these clinical scores in 596 patients. Significantly higher TASH (13 versus 6), McLaughlin (3.4 versus 2.4) and ABC (2
versus 1) scores were seen in patients who required massive transfusion compared with patients who did not, but no significant predictive
differences were identified between these scores [89]. None of these scoring systems include coagulation parameter measurements,
highlighting the fact that massive hemorrhage is generally due to active bleeding requiring surgical or interventional control, not as a result of
coagulopathy.

The utility of any clinical scoring system requires the demonstration that mortality is reduced as a result of a high score triggering a
transfusion protocol, and that earlier activation of these protocols is associated with a further decrease in mortality. There are no prospective
trials that demonstrate such a survival benefit; however, significant reductions in overall blood product use and hospital costs have been
reported [93,94].

Given that many massive transfusion protocols call for the early use of plasma and increased transfusion ratios (1:1:1), a sensitive scoring
system may allow the early identification of patients at risk for ATC and more precise transfusion triggers, but these scoring systems have not
been designed or validated as diagnostic tests for coagulopathy.

TREATMENT — A diagnosis of acute traumatic coagulopathy (ATC) predicts significantly higher transfusion requirements in the first 24
hours of hospitalization. In one study, patients with ATC on admission received an average of 10 units of blood, compared with the 2
received by those with normal clotting studies [7]. The need for blood transfusion and number of units transfused is a predictor of systemic
inflammation, acute respiratory distress, and mortality following traumatic injury [95-97].

Although red blood cell transfusion improves perfusion and oxygen carrying capacity, there is an increasing awareness that traditional
transfusion protocols produce or exacerbate resuscitation-associated coagulopathy. Resuscitation protocols continue to vary widely between
trauma centers and ratios of plasma:PRBC range from 1:1 to 1:10 [23,98-100]. With low plasma ratios, treatment of coagulopathy becomes
delayed, and ultimately a greater volume of blood is required. The recognition of this problem has led to a widespread re-evaluation of
transfusion protocols, particularly in patients who demonstrate acute traumatic coagulopathy.

Although no prospective, randomized trials are yet available to provide a definitive approach to transfusion for the trauma population, patients
with acute traumatic coagulopathy are at risk for massive transfusion and they appear to benefit from a resuscitation protocol using fresh
frozen plasma, packed red blood cells, and platelets (1:1:1 ratios) given early and aggressively, while limiting crystalloid [101,102]. However,
the value of this approach is controversial, and benefits remain to be proven in randomized trials. Not all investigators feel there is a clear
survival benefit to 1:1:1 resuscitation [68,103,104]. The retrospective nature of these studies introduces selection and survival biases, with
patients who have died early in the resuscitation effort often excluded [105]. Patients may have had longer survival not because they

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 5 of 24

received more plasma, but rather received more plasma because they survived long enough to do so [106]. The initial management of the
severely injured patient is discussed in detail elsewhere. (See "Initial evaluation and management of shock in adult trauma".)

Thromboelastography-based transfusion — Transfusion protocols based upon thromboelastography parameters including clotting time,
clot formation time, amplitude, and clot lysis index (table 4) have been suggested for standard and rotational thromboelastography based
upon single center experiences [83,87,90,107-109].

Although thromboelastography-guided transfusion triggers are preliminary, the authors feel that thromboelastography is a promising
technology. As institutional experience and clinical guidelines accrue, the rapid turnaround and multifaceted information provided by point-of-
care thromboelastography will likely provide improved criteria for transfusion in acutely injured patients. Thrombelastography-
guided “thrombostatic” resuscitation protocols will likely emerge as the new standard, but only single-center studies are currently available
[109,110].

An example protocol from Denver Health Medical Center, providing normal ranges and transfusion cutoffs, is given in the table (table 5)
[110,111]. Examples of the utility of thromboelastography in trauma are given below:

 The Denver group found that TEG® “G” values and thrombin generation parameters obtained six hours after arrival were significantly
associated with survival, while the INR was not [112].

 A retrospective evaluation of TEG® used in 44 combat patients found that maximal amplitude (MA) correlated more strongly with 24-
hour transfusion requirements than standard laboratory values [113].

 In another retrospective study of 832 patients requiring massive transfusion (21 percent trauma patients), patients whose transfusion
was guided by TEG® parameters received more plasma and platelets, and had significantly better 30-day survival (32 versus 20
percent) compared with retrospective controls transfused using standard laboratory measurements [114].

Pharmaceutical hemostatic agents — In addition to repletion of coagulation factors by transfusion, several pharmaceutical hemostatic
agents are available for the treatment of severe coagulopathy in the injured patient including recombinant factor VIIa, prothrombin complex
concentrate, antifibrinolytic agents (tranexamic acid, aminocaproic acid, aprotinin) and desmopressin.

 Recombinant factor VIIa – Recombinant factor VIIa was initially developed and approved for the treatment of hemophilia and
congenital factor deficiencies [115]. The recognition of injury-exposed tissue factor binding to activated factor VII as the principle
trigger of clot formation after trauma led to interest in using recombinant factor VIIa for the management of acute traumatic
coagulopathy. Recombinant human factor VIIa is an adjunctive treatment for coagulopathy associated with trauma but should be
reserved for salvage therapy. When used, it is important to correct acidosis, hypothermia, thrombocytopenia, and hypofibrinogenemia
prior to its use. The dosing of recombinant factor VIIa in trauma patients is discussed in detail elsewhere. (See "Therapeutic uses of
recombinant coagulation factor VIIa in non-hemophiliacs", section on 'Managing post-traumatic hemorrhage'.)

 Prothrombin complex concentrate – Prothrombin complex concentrate (PCC) is a factor concentrate enriched for factors II, VII, IX, and
X originally developed for hemorrhagic complications of hemophilia B [115]. The clinical use of PCC has been well studied in the
reversal of warfarin anticoagulation, since PCC is preferentially rich in the vitamin K-dependent clotting factors [116]. Some centers
have used PCC to correct coagulopathy after trauma and preliminary studies in animal models of hemorrhagic shock are promising,
but PCC has not been thoroughly evaluated in trauma patients [117]. (See "Plasma derivatives and recombinant DNA-produced
coagulation factors", section on 'Prothrombin complex concentrates'.)

 Antifibrinolytic therapy – An interest in antifibrinolytic therapy has been renewed because of the presence of enhanced fibrinolysis
following severe trauma, [7,118-122]. Antifibrinolytic therapy may be appropriate for patients with ongoing hemorrhagic shock who
have an elevated D-dimer and depleted fibrinogen.

 In the Clinical Randomization of an Antifibrinolytic in Significant Hemorrhage (CRASH-2) trial, an absolute mortality reduction of
1.5 percent was identified in patients receiving empiric tranexamic acid compared with placebo [119]. The coagulation status
(standard parameters or thromboelastography) of the patients enrolled was not specified. Enrollment criteria included adult trauma
patients within eight hours of injury with significant hemorrhage (systolic blood pressure <90 mmHg or heart rate >110 beats per
min, or both), or those who were considered to be at risk of significant hemorrhage.
 The subsequent Military Application of Tranexamic acid in Trauma Emergency Resuscitation (MATTERs) study evaluated
outcomes in 896 patients who were treated with tranexamic acid or not [123]. For the first half of the study, patients received
tranexamic acid at the discretion of the treating surgeon, and thereafter, the drug was administered as part of a major hemorrhage
protocol or clinical practice guideline to patients requiring emergency blood products or patients with evidence of hyperfibrinolysis.
Unadjusted mortality rates were significantly reduced for treated (17 versus 24 percent) versus nontreated casualties. In a
subgroup of patients requiring massive transfusion, greater reductions in mortality were seen for those who were treated (28
versus 14 percent).
 Further studies of tranexamic acid in patients with demonstrable ATC are needed to identify appropriate treatment criteria [119].
Other antifibrinolytic agents include aminocaproic acid and aprotinin, but these have not been evaluated in patients with traumatic
coagulopathy [120]. (See "Initial evaluation and management of shock in adult trauma", section on 'Antifibrinolytic agents'.)

 Desmopressin – Desmopressin was developed for the treatment of inherited bleeding diatheses [124], and to counteract uremic
bleeding [125]. There is insufficient clinical evidence to support the use of desmopressin in the trauma population except in those
patients with preexisting bleeding diatheses [126]. Preliminary animal studies show that desmopressin administration improves but
does not completely correct hypothermia or acidosis-induced platelet dysfunction, but clinical validation of these experimental data
needs to be performed [127,128]. (See "Platelet dysfunction in uremia", section on 'Desmopressin (dDAVP)'.)

Monitoring — Reliance upon standard serial laboratory measurements is not compatible with the timely correction of coagulopathy [129].
Thromboelastography provides more comprehensive information in real-time and readily identifies hyperfibrinolysis [84]. Serial
thromboelastography, when available, should be used to monitor the patient’s coagulation status, and to guide transfusion and the correction

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 6 of 24

of coagulopathy. (See 'Thromboelastography' above and 'Thromboelastography-based transfusion' above.)

Where thromboelastography is not available, serial PT/INR, PTT, hemoglobin/hematocrit, platelet count, and fibrinogen levels should be
obtained on arrival and following transfusion to verify an appropriate response to blood products and/or pharmaceutical hemostatic agents,
before and after operative interventions, and as dictated by the patient’s clinical course.

Serial measurements of arterial blood gas for pH and base deficit are indicated for monitoring the resolution of acidosis and tissue
hypoperfusion in response to resuscitation. During the course of operative intervention and ongoing shock resuscitation, central temperature
monitoring (eg, via Foley catheter or esophageal temperature probe) should be performed until normothermia is reestablished.

In the massively resuscitated patient, the early institution of intermittent intraabdominal pressure transducer is also appropriate to monitor for
the development of abdominal compartment syndrome and the need for abdominal decompression. (See "Abdominal compartment
syndrome", section on 'Measurement of intraabdominal pressure'.)

SUMMARY AND RECOMMENDATIONS

 Coagulopathy is associated with greater transfusion requirements, longer intensive care unit and hospital stays, more days of
mechanical ventilation, and a greater incidence of multiorgan failure and mortality. The identification and early correction of
coagulopathy is important to decrease fluid and transfusion requirements, decrease complications and improve survival. (See 'Impact'
above.)

 The etiology of coagulopathy in the injured patient is multifactorial with overlapping contributions from acidosis related to tissue injury
and shock, hypothermia related to exposure and fluid administration, hemodilution due to fluid or component blood product
administration. In severely injured patients, an additional biochemical process that remains incompletely characterized underlies ‘acute
traumatic coagulopathy’. Consumption of clotting factors manifesting as disseminated intravascular coagulation (DIC) may contribute
upon presentation or later in the hospital course. (See 'Etiology' above.)

 Acute traumatic coagulopathy (ATC) is an impairment of hemostasis and activation of fibrinolysis that occurs in response to severe
injury and is biochemically evident prior to, and independent of, the development of significant acidosis, hypothermia, or hemodilution.
Acute traumatic coagulopathy is mediated primarily by activation of the thrombomodulin-protein C system. (See 'Acute traumatic
coagulopathy' above.)

 Standard coagulation tests including prothrombin time/international normalized ratio (PT/INR), activated partial thromboplastin time
(PTT), fibrinogen level, and platelet count are part of the initial laboratory evaluation of trauma patients. In patients without pre-existing
coagulation defects, a prolonged prothrombin time (PT) and/or activated partial thromboplastin time (PTT) greater than 1.5 times
normal on admission defines the presence of acute traumatic coagulopathy. Clinically relevant ATC can occur in patients who have
normal platelet and fibrinogen levels. (See 'Diagnosis' above.)

 Thromboelastography is emerging as an important tool for identifying patients with acute traumatic coagulopathy and for real-time
monitoring of ongoing resuscitation efforts in injured patients. Thromboelastography measures the viscoelastic properties of clot
formation providing information on clot initiation, clot strength, and fibrinolysis. Additional clinical evaluation is ongoing in an effort to
establish guidelines for more widespread use. (See 'Thromboelastography' above.)

 For patients diagnosed with acute traumatic coagulopathy, we suggest plasma-based resuscitation, targeting ratios of packed red
blood cells, fresh frozen plasma, and platelets approaching 1:1:1 over protocols with lower ratios (Grade 2C). Injured patients with
acute traumatic coagulopathy are more likely to require massive transfusion and benefit from early matched transfusion. We prefer to
use thromboelastography to guide transfusion and monitor patients rather than treatment and follow-up guided by standard
coagulation tests. (See 'Treatment' above and "Initial evaluation and management of shock in adult trauma", section on 'Transfusion of
blood products'.)

 Pharmaceutical hemostatic agents available as adjuncts for the treatment of severe coagulopathy in the injured patient include
recombinant factor VIIa, prothrombin complex concentrate, antifibrinolytic agents (tranexamic acid, aminocaproic acid, aprotinin) and
desmopressin. (See 'Pharmaceutical hemostatic agents' above and "Initial evaluation and management of shock in adult trauma",
section on 'Developing treatments for hemorrhage'.)

Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES

1. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med 2006; 3:e442.
2. Brohi K, Singh J, Heron M, Coats T. Acute traumatic coagulopathy. J Trauma 2003; 54:1127.
3. MacLeod JB, Lynn M, McKenney MG, et al. Early coagulopathy predicts mortality in trauma. J Trauma 2003; 55:39.
4. Maegele M, Lefering R, Yucel N, et al. Early coagulopathy in multiple injury: an analysis from the German Trauma Registry on 8724
patients. Injury 2007; 38:298.
5. Spivey M, Parr MJ. Therapeutic approaches in trauma-induced coagulopathy. Minerva Anestesiol 2005; 71:281.
6. Hess JR, Brohi K, Dutton RP, et al. The coagulopathy of trauma: a review of mechanisms. J Trauma 2008; 65:748.
7. Brohi K, Cohen MJ, Ganter MT, et al. Acute traumatic coagulopathy: initiated by hypoperfusion: modulated through the protein C
pathway? Ann Surg 2007; 245:812.
8. Niles SE, McLaughlin DF, Perkins JG, et al. Increased mortality associated with the early coagulopathy of trauma in combat casualties.
J Trauma 2008; 64:1459.
9. Engström M, Schött U, Romner B, Reinstrup P. Acidosis impairs the coagulation: A thromboelastographic study. J Trauma 2006;
61:624.
10. Martini WZ. Coagulopathy by hypothermia and acidosis: mechanisms of thrombin generation and fibrinogen availability. J Trauma
2009; 67:202.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 7 of 24

11. Meng ZH, Wolberg AS, Monroe DM 3rd, Hoffman M. The effect of temperature and pH on the activity of factor VIIa: implications for the
efficacy of high-dose factor VIIa in hypothermic and acidotic patients. J Trauma 2003; 55:886.
12. Hess JR. Blood and coagulation support in trauma care. Hematology Am Soc Hematol Educ Program 2007; :187.
13. Martini WZ, Dubick MA, Pusateri AE, et al. Does bicarbonate correct coagulation function impaired by acidosis in swine? J Trauma
2006; 61:99.
14. Martini WZ, Dubick MA, Wade CE, Holcomb JB. Evaluation of tris-hydroxymethylaminomethane on reversing coagulation abnormalities
caused by acidosis in pigs. Crit Care Med 2007; 35:1568.
15. Tsuei BJ, Kearney PA. Hypothermia in the trauma patient. Injury 2004; 35:7.
16. Wolberg AS, Meng ZH, Monroe DM 3rd, Hoffman M. A systematic evaluation of the effect of temperature on coagulation enzyme
activity and platelet function. J Trauma 2004; 56:1221.
17. Farkash U, Lynn M, Scope A, et al. Does prehospital fluid administration impact core body temperature and coagulation functions in
combat casualties? Injury 2002; 33:103.
18. Kermode JC, Zheng Q, Milner EP. Marked temperature dependence of the platelet calcium signal induced by human von Willebrand
factor. Blood 1999; 94:199.
19. Lier H, Krep H, Schroeder S, Stuber F. Preconditions of hemostasis in trauma: a review. The influence of acidosis, hypocalcemia,
anemia, and hypothermia on functional hemostasis in trauma. J Trauma 2008; 65:951.
20. Shafi S, Elliott AC, Gentilello L. Is hypothermia simply a marker of shock and injury severity or an independent risk factor for mortality in
trauma patients? Analysis of a large national trauma registry. J Trauma 2005; 59:1081.
21. Dirkmann D, Hanke AA, Görlinger K, Peters J. Hypothermia and acidosis synergistically impair coagulation in human whole blood.
Anesth Analg 2008; 106:1627.
22. Schreiber MA. Coagulopathy in the trauma patient. Curr Opin Crit Care 2005; 11:590.
23. Hirshberg A, Dugas M, Banez EI, et al. Minimizing dilutional coagulopathy in exsanguinating hemorrhage: a computer simulation. J
Trauma 2003; 54:454.
24. Brazil EV, Coats TJ. Sonoclot coagulation analysis of in-vitro haemodilution with resuscitation solutions. J R Soc Med 2000; 93:507.
25. Coats TJ, Brazil E, Heron M. The effects of commonly used resuscitation fluids on whole blood coagulation. Emerg Med J 2006; 23:546.
26. Tinmouth A, Fergusson D, Yee IC, et al. Clinical consequences of red cell storage in the critically ill. Transfusion 2006; 46:2014.
27. Fukuda T, Nakashima Y, Harada M, et al. Effect of whole blood clotting time in rats with ionized hypocalcemia induced by rapid
intravenous citrate infusion. J Toxicol Sci 2006; 31:229.
28. Levy JH. Massive transfusion coagulopathy. Semin Hematol 2006; 43:S59.
29. Whitaker, B, Sullivan, M. The 2005 Nationwide Blood Collection and Utilization Survey Report 2005. AABB.
http://aabb.org/programs/biovigilance/nbcus/Documents/05nbcusrpt.pdf (Accessed on December 15, 2010).
30. Koch CG, Li L, Sessler DI, et al. Duration of red-cell storage and complications after cardiac surgery. N Engl J Med 2008; 358:1229.
31. Spinella PC, Carroll CL, Staff I, et al. Duration of red blood cell storage is associated with increased incidence of deep vein thrombosis
and in hospital mortality in patients with traumatic injuries. Crit Care 2009; 13:R151.
32. Dzik W. Fresh blood for everyone? Balancing availability and quality of stored RBCs. Transfus Med 2008; 18:260.
33. Hess JR, Lawson JH. The coagulopathy of trauma versus disseminated intravascular coagulation. J Trauma 2006; 60:S12.
34. Gando S. Acute coagulopathy of trauma shock and coagulopathy of trauma: a rebuttal. You are now going down the wrong path. J
Trauma 2009; 67:381.
35. Levi M. Disseminated intravascular coagulation. Crit Care Med 2007; 35:2191.
36. Gando S, Saitoh D, Ogura H, et al. Natural history of disseminated intravascular coagulation diagnosed based on the newly
established diagnostic criteria for critically ill patients: results of a multicenter, prospective survey. Crit Care Med 2008; 36:145.
37. Taylor FB Jr, Toh CH, Hoots WK, et al. Towards definition, clinical and laboratory criteria, and a scoring system for disseminated
intravascular coagulation. Thromb Haemost 2001; 86:1327.
38. Gando S, Tedo I, Kubota M. Posttrauma coagulation and fibrinolysis. Crit Care Med 1992; 20:594.
39. Brohi K, Cohen MJ, Davenport RA. Acute coagulopathy of trauma: mechanism, identification and effect. Curr Opin Crit Care 2007;
13:680.
40. Shaz BH, Winkler AM, James AB, et al. Pathophysiology of early trauma-induced coagulopathy: emerging evidence for hemodilution
and coagulation factor depletion. J Trauma 2011; 70:1401.
41. Johansson PI, Sørensen AM, Perner A, et al. Disseminated intravascular coagulation or acute coagulopathy of trauma shock early
after trauma? An observational study. Crit Care 2011; 15:R272.
42. Chesebro BB, Rahn P, Carles M, et al. Increase in activated protein C mediates acute traumatic coagulopathy in mice. Shock 2009;
32:659.
43. Cadroy Y, Diquélou A, Dupouy D, et al. The thrombomodulin/protein C/protein S anticoagulant pathway modulates the thrombogenic
properties of the normal resting and stimulated endothelium. Arterioscler Thromb Vasc Biol 1997; 17:520.
44. Esmon CT. Protein C pathway in sepsis. Ann Med 2002; 34:598.
45. Rezaie AR. Vitronectin functions as a cofactor for rapid inhibition of activated protein C by plasminogen activator inhibitor-1.
Implications for the mechanism of profibrinolytic action of activated protein C. J Biol Chem 2001; 276:15567.
46. Bajzar L, Jain N, Wang P, Walker JB. Thrombin activatable fibrinolysis inhibitor: not just an inhibitor of fibrinolysis. Crit Care Med 2004;
32:S320.
47. Mosnier LO, Zlokovic BV, Griffin JH. The cytoprotective protein C pathway. Blood 2007; 109:3161.
48. Xu J, Ji Y, Zhang X, et al. Endogenous activated protein C signaling is critical to protection of mice from lipopolysaccaride-induced
septic shock. J Thromb Haemost 2009; 7:851.
49. Cohen MJ, Call M, Nelson M, et al. Critical role of activated protein C in early coagulopathy and later organ failure, infection and death
in trauma patients. Ann Surg 2012; 255:379.
50. Johansson PI, Stensballe J, Rasmussen LS, Ostrowski SR. A high admission syndecan-1 level, a marker of endothelial glycocalyx
degradation, is associated with inflammation, protein C depletion, fibrinolysis, and increased mortality in trauma patients. Ann Surg
2011; 254:194.
51. Finigan JH, Boueiz A, Wilkinson E, et al. Activated protein C protects against ventilator-induced pulmonary capillary leak. Am J Physiol
Lung Cell Mol Physiol 2009; 296:L1002.
52. Finigan JH, Dudek SM, Singleton PA, et al. Activated protein C mediates novel lung endothelial barrier enhancement: role of
sphingosine 1-phosphate receptor transactivation. J Biol Chem 2005; 280:17286.
53. Cohen MJ, Bir N, Rahn P, et al. Protein C depletion early after trauma increases the risk of ventilator-associated pneumonia. J Trauma

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 8 of 24

2009; 67:1176.
54. Ganter MT, Brohi K, Cohen MJ, et al. Role of the alternative pathway in the early complement activation following major trauma. Shock
2007; 28:29.
55. Huber-Lang M, Sarma JV, Zetoune FS, et al. Generation of C5a in the absence of C3: a new complement activation pathway. Nat Med
2006; 12:682.
56. Esmon CT. The interactions between inflammation and coagulation. Br J Haematol 2005; 131:417.
57. Xu J, Zhang X, Pelayo R, et al. Extracellular histones are major mediators of death in sepsis. Nat Med 2009; 15:1318.
58. Bae JS, Yang L, Manithody C, Rezaie AR. The ligand occupancy of endothelial protein C receptor switches the protease-activated
receptor 1-dependent signaling specificity of thrombin from a permeability-enhancing to a barrier-protective response in endothelial
cells. Blood 2007; 110:3909.
59. Feistritzer C, Riewald M. Endothelial barrier protection by activated protein C through PAR1-dependent sphingosine 1-phosphate
receptor-1 crossactivation. Blood 2005; 105:3178.
60. Schuepbach RA, Feistritzer C, Fernández JA, et al. Protection of vascular barrier integrity by activated protein C in murine models
depends on protease-activated receptor-1. Thromb Haemost 2009; 101:724.
61. Meissner MH, Chandler WL, Elliott JS. Venous thromboembolism in trauma: a local manifestation of systemic hypercoagulability? J
Trauma 2003; 54:224.
62. Selby R, Geerts W, Ofosu FA, et al. Hypercoagulability after trauma: hemostatic changes and relationship to venous thromboembolism.
Thromb Res 2009; 124:281.
63. Knudson MM, Collins JA, Goodman SB, McCrory DW. Thromboembolism following multiple trauma. J Trauma 1992; 32:2.
64. Silva E, de Figueiredo LF, Colombari F. Prowess-shock trial: a protocol overview and perspectives. Shock 2010; 34 Suppl 1:48.
65. FDA Drug Safety Communication: Voluntary market withdrawal of Xigris [drotrecogin alfa (activated)] due to failure to show a survival
benefit. http://www.fda.gov/Drugs/DrugSafety/ucm277114.htm (Accessed on October 25, 2011).
66. Owen CA Jr. Historical account of tests of hemostasis. Am J Clin Pathol 1990; 93:S3.
67. Eckman MH, Erban JK, Singh SK, Kao GS. Screening for the risk for bleeding or thrombosis. Ann Intern Med 2003; 138:W15.
68. Kashuk JL, Moore EE, Johnson JL, et al. Postinjury life threatening coagulopathy: is 1:1 fresh frozen plasma:packed red blood cells the
answer? J Trauma 2008; 65:261.
69. Hoyt DB, Dutton RP, Hauser CJ, et al. Management of coagulopathy in the patients with multiple injuries: results from an international
survey of clinical practice. J Trauma 2008; 65:755.
70. Frith D, Goslings JC, Gaarder C, et al. Definition and drivers of acute traumatic coagulopathy: clinical and experimental investigations.
J Thromb Haemost 2010; 8:1919.
71. Hess JR, Lindell AL, Stansbury LG, et al. The prevalence of abnormal results of conventional coagulation tests on admission to a
trauma center. Transfusion 2009; 49:34.
72. Jacoby RC, Owings JT, Holmes J, et al. Platelet activation and function after trauma. J Trauma 2001; 51:639.
73. Favaloro EJ. Clinical application of the PFA-100. Curr Opin Hematol 2002; 9:407.
74. Velik-Salchner C, Maier S, Innerhofer P, et al. Point-of-care whole blood impedance aggregometry versus classical light transmission
aggregometry for detecting aspirin and clopidogrel: the results of a pilot study. Anesth Analg 2008; 107:1798.
75. Shore-Lesserson L, Manspeizer HE, DePerio M, et al. Thromboelastography-guided transfusion algorithm reduces transfusions in
complex cardiac surgery. Anesth Analg 1999; 88:312.
76. Kang Y. Thromboelastography in liver transplantation. Semin Thromb Hemost 1995; 21 Suppl 4:34.
77. Jeger V, Zimmermann H, Exadaktylos AK. Can RapidTEG accelerate the search for coagulopathies in the patient with multiple
injuries? J Trauma 2009; 66:1253.
78. Cotton BA, Faz G, Hatch QM, et al. Rapid thrombelastography delivers real-time results that predict transfusion within 1 hour of
admission. J Trauma 2011; 71:407.
79. Schreiber MA, Differding J, Thorborg P, et al. Hypercoagulability is most prevalent early after injury and in female patients. J Trauma
2005; 58:475.
80. Levrat A, Gros A, Rugeri L, et al. Evaluation of rotation thrombelastography for the diagnosis of hyperfibrinolysis in trauma patients. Br
J Anaesth 2008; 100:792.
81. Kaufmann CR, Dwyer KM, Crews JD, et al. Usefulness of thrombelastography in assessment of trauma patient coagulation. J Trauma
1997; 42:716.
82. Park MS, Martini WZ, Dubick MA, et al. Thromboelastography as a better indicator of hypercoagulable state after injury than
prothrombin time or activated partial thromboplastin time. J Trauma 2009; 67:266.
83. Carroll RC, Craft RM, Langdon RJ, et al. Early evaluation of acute traumatic coagulopathy by thrombelastography. Transl Res 2009;
154:34.
84. Schöchl H, Frietsch T, Pavelka M, Jámbor C. Hyperfibrinolysis after major trauma: differential diagnosis of lysis patterns and prognostic
value of thrombelastometry. J Trauma 2009; 67:125.
85. Theusinger OM, Wanner GA, Emmert MY, et al. Hyperfibrinolysis diagnosed by rotational thromboelastometry (ROTEM) is associated
with higher mortality in patients with severe trauma. Anesth Analg 2011; 113:1003.
86. Tauber H, Innerhofer P, Breitkopf R, et al. Prevalence and impact of abnormal ROTEM(R) assays in severe blunt trauma: results of
the 'Diagnosis and Treatment of Trauma-Induced Coagulopathy (DIA-TRE-TIC) study'. Br J Anaesth 2011; 107:378.
87. Rugeri L, Levrat A, David JS, et al. Diagnosis of early coagulation abnormalities in trauma patients by rotation thrombelastography. J
Thromb Haemost 2007; 5:289.
88. Hiippala ST, Myllylä GJ, Vahtera EM. Hemostatic factors and replacement of major blood loss with plasma-poor red cell concentrates.
Anesth Analg 1995; 81:360.
89. Nunez TC, Voskresensky IV, Dossett LA, et al. Early prediction of massive transfusion in trauma: simple as ABC (assessment of blood
consumption)? J Trauma 2009; 66:346.
90. Schreiber MA, Perkins J, Kiraly L, et al. Early predictors of massive transfusion in combat casualties. J Am Coll Surg 2007; 205:541.
91. Yücel N, Lefering R, Maegele M, et al. Trauma Associated Severe Hemorrhage (TASH)-Score: probability of mass transfusion as
surrogate for life threatening hemorrhage after multiple trauma. J Trauma 2006; 60:1228.
92. McLaughlin DF, Niles SE, Salinas J, et al. A predictive model for massive transfusion in combat casualty patients. J Trauma 2008;
64:S57.
93. Cotton BA, Au BK, Nunez TC, et al. Predefined massive transfusion protocols are associated with a reduction in organ failure and
postinjury complications. J Trauma 2009; 66:41.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 9 of 24

94. O'Keeffe T, Refaai M, Tchorz K, et al. A massive transfusion protocol to decrease blood component use and costs. Arch Surg 2008;
143:686.
95. Charles A, Shaikh AA, Walters M, et al. Blood transfusion is an independent predictor of mortality after blunt trauma. Am Surg 2007;
73:1.
96. Dunne JR, Malone DL, Tracy JK, Napolitano LM. Allogenic blood transfusion in the first 24 hours after trauma is associated with
increased systemic inflammatory response syndrome (SIRS) and death. Surg Infect (Larchmt) 2004; 5:395.
97. Silverboard H, Aisiku I, Martin GS, et al. The role of acute blood transfusion in the development of acute respiratory distress syndrome
in patients with severe trauma. J Trauma 2005; 59:717.
98. Cotton BA, Gunter OL, Isbell J, et al. Damage control hematology: the impact of a trauma exsanguination protocol on survival and
blood product utilization. J Trauma 2008; 64:1177.
99. Alam HB, Rhee P. New developments in fluid resuscitation. Surg Clin North Am 2007; 87:55.
100. Gonzalez EA, Moore FA, Holcomb JB, et al. Fresh frozen plasma should be given earlier to patients requiring massive transfusion. J
Trauma 2007; 62:112.
101. de Biasi AR, Stansbury LG, Dutton RP, et al. Blood product use in trauma resuscitation: plasma deficit versus plasma ratio as
predictors of mortality in trauma (CME). Transfusion 2011; 51:1925.
102. Brown LM, Aro SO, Cohen MJ, et al. A high fresh frozen plasma: packed red blood cell transfusion ratio decreases mortality in all
massively transfused trauma patients regardless of admission international normalized ratio. J Trauma 2011; 71:S358.
103. Sperry JL, Ochoa JB, Gunn SR, et al. An FFP:PRBC transfusion ratio >/=1:1.5 is associated with a lower risk of mortality after massive
transfusion. J Trauma 2008; 65:986.
104. Scalea TM, Bochicchio KM, Lumpkins K, et al. Early aggressive use of fresh frozen plasma does not improve outcome in critically
injured trauma patients. Ann Surg 2008; 248:578.
105. Rajasekhar A, Gowing R, Zarychanski R, et al. Survival of trauma patients after massive red blood cell transfusion using a high or low
red blood cell to plasma transfusion ratio. Crit Care Med 2011; 39:1507.
106. Snyder CW, Weinberg JA, McGwin G Jr, et al. The relationship of blood product ratio to mortality: survival benefit or survival bias? J
Trauma 2009; 66:358.
107. Kheirabadi BS, Crissey JM, Deguzman R, Holcomb JB. In vivo bleeding time and in vitro thrombelastography measurements are better
indicators of dilutional hypothermic coagulopathy than prothrombin time. J Trauma 2007; 62:1352.
108. Martini WZ, Cortez DS, Dubick MA, et al. Thrombelastography is better than PT, aPTT, and activated clotting time in detecting clinically
relevant clotting abnormalities after hypothermia, hemorrhagic shock and resuscitation in pigs. J Trauma 2008; 65:535.
109. Schöchl H, Nienaber U, Hofer G, et al. Goal-directed coagulation management of major trauma patients using thromboelastometry
(ROTEM)-guided administration of fibrinogen concentrate and prothrombin complex concentrate. Crit Care 2010; 14:R55.
110. Kashuk JL, Moore EE, Sawyer M, et al. Postinjury coagulopathy management: goal directed resuscitation via POC
thrombelastography. Ann Surg 2010; 251:604.
111. Stahel PF, Moore EE, Schreier SL, et al. Transfusion strategies in postinjury coagulopathy. Curr Opin Anaesthesiol 2009; 22:289.
112. Kashuk JL, Moore EE, Wohlauer M, et al. Initial experiences with point-of-care rapid thrombelastography for management of life-
threatening postinjury coagulopathy. Transfusion 2012; 52:23.
113. Plotkin AJ, Wade CE, Jenkins DH, et al. A reduction in clot formation rate and strength assessed by thrombelastography is indicative of
transfusion requirements in patients with penetrating injuries. J Trauma 2008; 64:S64.
114. Johansson PI, Stensballe J. Effect of Haemostatic Control Resuscitation on mortality in massively bleeding patients: a before and after
study. Vox Sang 2009; 96:111.
115. Limentani SA, Roth DA, Furie BC, Furie B. Recombinant blood clotting proteins for hemophilia therapy. Semin Thromb Hemost 1993;
19:62.
116. Ansell J, Hirsh J, Poller L, et al. The pharmacology and management of the vitamin K antagonists: the Seventh ACCP Conference on
Antithrombotic and Thrombolytic Therapy. Chest 2004; 126:204S.
117. Dickneite G, Dörr B, Kaspereit F, Tanaka KA. Prothrombin complex concentrate versus recombinant factor VIIa for reversal of
hemodilutional coagulopathy in a porcine trauma model. J Trauma 2010; 68:1151.
118. Kashuk JL, Moore EE, Sawyer M, et al. Primary fibrinolysis is integral in the pathogenesis of the acute coagulopathy of trauma. Ann
Surg 2010; 252:434.
119. CRASH-2 trial collaborators, Shakur H, Roberts I, 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; 376:23.
120. Henry DA, Moxey AJ, Carless PA, et al. Anti-fibrinolytic use for minimising perioperative allogeneic blood transfusion. Cochrane
Database Syst Rev 2001; :CD001886.
121. Mangano DT, Tudor IC, Dietzel C, et al. The risk associated with aprotinin in cardiac surgery. N Engl J Med 2006; 354:353.
122. McMichan JC, Rosengarten DS, Philipp E. Prophylaxis of post-traumatic pulmonary insufficiency by protease-inhibitor therapy with
aprotinin: a clinical study. Circ Shock 1982; 9:107.
123. Morrison JJ, Dubose JJ, Rasmussen TE, Midwinter MJ. Military Application of Tranexamic Acid in Trauma Emergency Resuscitation
(MATTERs) Study. Arch Surg 2012; 147:113.
124. Mannucci PM, Ruggeri ZM, Pareti FI, Capitanio A. 1-Deamino-8-d-arginine vasopressin: a new pharmacological approach to the
management of haemophilia and von Willebrands' diseases. Lancet 1977; 1:869.
125. Mannucci PM, Remuzzi G, Pusineri F, et al. Deamino-8-D-arginine vasopressin shortens the bleeding time in uremia. N Engl J Med
1983; 308:8.
126. Mannucci PM, Levi M. Prevention and treatment of major blood loss. N Engl J Med 2007; 356:2301.
127. Hanke AA, Dellweg C, Kienbaum P, et al. Effects of desmopressin on platelet function under conditions of hypothermia and acidosis:
an in vitro study using multiple electrode aggregometry*. Anaesthesia 2010; 65:688.
128. Ying CL, Tsang SF, Ng KF. The potential use of desmopressin to correct hypothermia-induced impairment of primary haemostasis--an
in vitro study using PFA-100. Resuscitation 2008; 76:129.
129. Ho AM, Karmakar MK, Dion PW. Are we giving enough coagulation factors during major trauma resuscitation? Am J Surg 2005;
190:479.

Topic 15147 Version 6.0

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 10 of 24

GRAPHICS

Coagulation parameters in acute and chronic disseminated intravascular coagulation

Parameter Acute (decompensated) DIC Chronic (compensated) DIC


Platelet count Reduced Variable

Prothrombin time Prolonged Normal

Activated partial thromboplastin time Prolonged Normal

Thrombin time Prolonged Normal

Plasma fibrinogen Reduced Normal-elevated

Plasma factor V Reduced Normal

Plasma factor VIII Reduced Normal

Fibrin degradation products Elevated Elevated

D-dimer Elevated Elevated

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 11 of 24

Coagulation cascade initiated by tissue factor release at the site


of tissue injury

Factors in squares are inactive forms, whereas factors in circles are active
enzymes.
PT: prothrombin; Th: thrombin.
Adapted from: Ferguson JJ, et al. Eur Heart J 1998; 19(Suppl D):D6.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 12 of 24

Regulation of fibrinolysis by plasminogen activator


inhibitor-1 (PAI-1), α2-antiplasmin, and thrombin-
activatable fibrinolysis inhibitor (TAFI)

PAI-1 inhibits plasmin formation by inhibiting tissue-type plasminogen


activator (t-PA). α2-antiplasmin inhibits the activity of plasmin, thereby
inhibiting fibrinolysis. TAFI circulates in plasma as a zymogen. It is
activated by thrombin when thrombin is bound on endothelial
thrombomodulin, and therefore represents a link between blood
coagulation and fibrinolysis. During fibrinolysis, plasmin cleaves intact
fibrin at lysine residues, initially yielding large, insoluble fibrin
fragments with lysine residues at their carboxyl termini. Plasminogen
binds avidly to C-terminal lysine residues within the partially degraded
fibrin clot and assumes a conformation that is susceptible to activation
by t-PA, thereby promoting plasmin formation, continued fibrinolysis
("rapid lysis by plasmin"), and generation of smaller, soluble fibrin
fragments that are dispersed by flowing blood. Activated TAFI (TAFIa) is
a carboxypeptidase that removes lysine residues from the carboxy (C)
termini of partially degraded fibrin fragments. By removing C-terminal
lysine residues from large fibrin fragments in the partially degraded
clot, TAFI inhibits recruitment of plasminogen to the clot, thereby
slowing fibrinolysis ("slow lysis by plasmin").
Diagram supplied by William P Fay, MD.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 13 of 24

Components of the plasma fibrinolytic system

Molecular weight (d) Activity


Plasminogen 88,000 (single chain) Proenzyme form of fibrinolytic enzyme

Plasmin 88,000 (two chain) Active fibrinolytic enzyme

TPA 70,000 (one/two chain) Enzyme present in tissues that coverts plasminogen to plasmin

UPA 54,000 (two chain) Plasminogen activator (different from tPA)

α2PI 70,000 (single chain) Specific fast-acting inhibitor in plasma

PAI-1 40,000 (single chain) Fast-acting inhibitor of tPA (and UPA) secreted by endothelial cells)

d: Daltons; TPA: tissue plasminogen activator; UPA: urokinase-like plasminogen activator; α2PI: alpha-2 plasmin inhibitor; PAI-1:
plasminogen activator inhibitor-1.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 14 of 24

TEG tracing interpretation

TEG: thromboelastograph
%: percent.
Image of the TEG® Thromboelastograph ® Hemostasis Analyzer Parameters LY30 Clot Breakdown is used
by permission of Haemonetics Corporation.
Image of the TEG® Thromboelastograph ® Hemostasis Analyzer Tracing is used by permission of
Haemonetics Corporation.
TEG® and Thromboelastograph ® are registers trademarks of Haemonetics Corporation in the US, other
countries, or both.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 15 of 24

Various types of thromboelastograms

Native TEG Measures clot formation in the absence of a specific activator in either untreated whole blood or in
citrated whole blood after recalcification.

Rapid-TEG Tissue factor is used as the activator inducing clot formation via the extrinsic pathway. Compared with
(rTEG) native TEG, rTEG gives a readout of clot characteristics within about 10 minutes. The corresponding
RoTEM® test is the EXTEM.

Kaolin-TEG Phospholipid and kaolin are used to induce activation via the intrinsic pathway. The corresponding
RoTEM® test is the INTEM, which uses phospholipid and ellagic acid as activators.

Heparinase Simultaneous measurements can be performed comparing heparinase-treated blood with untreated blood
treatment to identify the effects of endogenous heparan sulfates, exogenous heparin and heparinoids. The
corresponding RoTEM® assay is the HEPTEM.

Platelet Platelet mapping assesses platelet function via the cyclooxygenase and ADP/P2 signaling pathways. The
mapping decrement in MA can be measured in the presence of platelet antagonists such as acetylsalicylic acid,
dipyridamole, and clopidogrel and compared with the untreated MA.

Fibrin function The RoTEM® FIBTEM test activates the extrinsic pathway in the presence of cytochalasin D, which is a
testing cytoskeletal inhibitor of platelet activity. This test qualitatively assesses fibrinogen levels since the clot
formation in this test is attributable to fibrin polymerization alone.

TEG: thromboelastogram; ADP/P2: adenosine diphosphate/P2 receptor; MA: maximal amplitude.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 16 of 24

Normal thromboelastography and parameters

Normal citrated rapid TEG tracing with normal ACT, R time, K time, alpha angle, MA, G,
and LY30.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 17 of 24

Primary hyperfibrinolysis as assessed by thromboelastography

Citrated rapid thromboelastography study (rapid-TEG) showing primary


hyperfibrinolysis. The red tracing indicates a normal study.
This study has a normal ACT, normal R time, normal K time, and normal alpha angle;
low MA and G, reflecting significant fibrinolysis prior to achieving maximal clot
strength; high LY30 reflecting hyperfibrinolysis.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 18 of 24

Secondary hyperfibrinolysis

Citrated rapid-TEG showing secondary hyperfibrinolysis. The red tracing indicates a


normal study.
This study shows a normal ACT, R time, K time, and alpha angle; high LY30 reflecting
hyperfibrinolysis; normal MA and G, reflecting onset of fibrinolysis after achieving
maximal clot strength.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 19 of 24

Thrombocytopenia

Citrated rapid-TEG reflecting thrombocytopenia.The red line is a normal tracing.


This study shows a normal ACT, R time, K time, and LY30; low alpha angle, MA, and G,
reflecting platelet hypocoagulability.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 20 of 24

Clotting factor consumption and hypofibrinogenemia

Citrated rapid-TEG reflecting clotting factor consumption and hypofibrinogenemia. The


red line indicates a normal study.
This study shows a prolonged ACT, R time, and K time, reflecting enzymatic
hypocoagulability; markedly low alpha angle, MA, and G, reflecting platelet
hypocoagulability and poor fibrin deposition; normal LY30.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 21 of 24

Hypercoagulability

Citrated rapid-TEG reflecting hypercoagulability. The red tracing indicates a normal


study.
This study shows a shortened to normal ACT, R time, and K time, reflecting enzymatic
hypercoagulability; high alpha angle, MA, and G, reflecting platelet hypercoagulability
and/or excessive fibrin deposition; normal LY30. To distinguish the relative contribution
of platelet hypercoagulability and excessive fibrin deposition, one would need to
perform TEG platelet mapping or RoTEM® fibrin function testing.
R (reaction time): time from sample placement or activation until the tracing amplitude reaches 2 mm;
K (clot formation time): time elapsed between the R-time and the point where the tracing amplitude
reaches 20 mm; alpha angle: angle between the middle line of the tracing and a tangential line to the
developing "body" of the tracing; MA (maximal amplitude): the maximal amplitude reached after clot
initiation; G: a calculated measure of total clot strength based on amplitude; LY30: the amount of clot
lysis detected 30 minutes after the maximal amplitute (MA) is reached; SP (split point): time from
sample placement (or activation with an exogenous procoagulant) until the earliest detectable
resistance.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 22 of 24

Thromboelastography definitions

Clot
Parameter Measurement TEG® ROTEM® Enzymatic
Abnormalities
phase abbreviation abbreviation stage

Clot Clotting time Time from start of Reaction time Clot time (CT) Early Prolonged by
initiation sample to 2 mm (R) activation of clotting factor
clot amplitude clotting deficiencies,
cascade anticoagulants, and
resulting in hypofibrinogenemia.
initial Shortened in
thrombin hypercoagulable
burst states.

Clot Clot formation Time from 2 mm Clot formation Clot formation Clot Prolonged by
kinetics time to 20 mm clot time (K) time (CFT) potentiation clotting factor
amplitude by activation deficiencies,
of platelets hypofibrinogenemia,
and thrombocytopenia,
thrombin- and platelet
mediated dysfunction.
cleavage of
Angle Angle of tangent Alpha angle Alpha angle soluble Abnormally low in
line from 2 mm to fibrinogen clotting factor
20 mm clot deficiencies,
formation hypofibrinogenemia,
thrombocytopenia,
and platelet
dysfunction.

Clot Maximal clot Amplitude Maximal Maximal clot Maximal clot Abnormally low in
strength strength measured at peak amplitude (MA) firmness (MCF) strength hypofibrinogenemia,
clot strength achieved via thromboyctopenia,
GP IIb/IIIa- or platelet
mediated dysfunction.
platelet-fibrin
Clot Calculated from G Maximal clot interactions Abnormally high in
viscoelasticity maximal elasticity (MCE) platelet
amplitude hypercoagulability.

Clot Clot lysis Percentage of loss Lysis at 30 min Lysis index at 30 Activation of Abnormally high in
lysis of amplitude at (LY30), min (LI30), fibrinolytic enzymatic or
fixed time after estimated maximal lysis system mechanical
maximal percentage of (ML) hyperfibrinolysis.
amplitude lysis (EPL)

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 23 of 24

Thromboelastography-guided transfusion parameters

r-TEG parameter Normal range Transfusion trigger


TEG-ACT 78-110 seconds >120 seconds (FFP)

Alpha angle 66°-82° <66° (cryoprecipitate)

K value 30-120 seconds >120 seconds (cryoprecipitate)

MA (maximum amplitude) 54-72 mm <54 mm (platelets)

G value 5.3-12.4 dynes/second <5.3 dynes/second*

LY-30 (lysis at 30 min) 0-8 percent >8 percent•

Values in the table are for non-citrated whole blood samples.


r-TEG: rapid thromboelastography; ACT: activated clotting time; FFP: fresh frozen plasma.
* Reevaluate all parameters per massive transfusion algorithm-decision tree.
• Consider antifibrinolytic.
Reproduced with permission from: Stahel PF, Moore EE, Schreier SL, et al. Transfusion strategies in postinjury coagulopathy. Curr Opin
Anaesthesiol 2009; 22:289. Copyright © 2009 Lippincott Williams & Wilkins.

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02
Coagulopathy associated with trauma Page 24 of 24

http://0-www.uptodate.com.innopac.wits.ac.za/contents/coagulopathy-associated-with... 2013/09/02

You might also like