You are on page 1of 11

|

Received: 12 November 2018    Accepted: 3 April 2019

DOI: 10.1111/jth.14450

REVIEW ARTICLE

Trauma‐induced coagulopathy: The past, present, and future

Lucy Z. Kornblith1  | Hunter B. Moore2 | Mitchell J. Cohen2

1
Department of Surgery, Zuckerberg San
Francisco General Hospital, University of Abstract
California, San Francisco, California Trauma remains a leading cause of death worldwide, and most early preventable
2
Department of Surgery, Denver Health
deaths in both the civilian and military settings are due to uncontrolled hemorrhage,
Medical Center, University of Colorado,
Denver, Colorado despite paradigm advances in modern trauma care. Combined tissue injury and shock
result in hemostatic failure, which has been identified as a multidimensional molec‐
Correspondence
Lucy Z. Kornblith, Department of Surgery, ular, physiologic and clinical disorder termed trauma‐induced coagulopathy (TIC).
Zuckerberg San Francisco General Hospital
Understanding the biology of TIC is of utmost importance, as it is often responsible
and the University of California, 1001
Potrero Avenue, Building 1, Suite 210, San for uncontrolled bleeding, organ failure, thromboembolic complications, and death.
Francisco, CA 94110.
Investigations have shown that TIC is characterized by multiple phenotypes of im‐
Email: lucy.kornblith@ucsf.edu
paired hemostasis due to altered biology in clot formation and breakdown. These
Funding information
coagulopathies are attributable to tissue injury and shock, and encompass underlying
National Institute of General Medical
Sciences, Grant/Award Number: endothelial, immune and inflammatory perturbations. Despite the recognition and
1K23GM130892‐01; National Institutes
identification of multiple mechanisms and mediators of TIC, and the development
of Health, Grant/Award Number:
UM1HL120877; US Department of Defense, of targeted treatments, the mortality rates and associated morbidities due to hem‐
Grant/Award Number: W911QY‐15‐C‐0044;
orrhage after injury remain high. The purpose of this review is to examine the past
Eastern Association for the Surgery of
Trauma, Trauma Research Scholarship, and present understanding of the multiple distinct but highly integrated pathways
Grant/Award Number: P0526402
implicated in TIC, in order to highlight the current knowledge gaps and future needs
in this evolving field, with the aim of reducing morbidity and mortality after injury.

KEYWORDS
blood coagulation disorders, exsanguination, hemorrhagic shock, hemostasis, trauma

1 | I NTRO D U C TI O N is a multiphenotypic disease state that encompasses disorders of


coagulation and inflammation characterized by impairments in clot
Trauma remains a leading cause of death worldwide,1 and it is ex‐ formation, breakdown, and overall vascular homeostasis. TIC is as‐
pected that rates of death due to injury will continue to rise as popu‐ sociated with increased early transfusion requirements, the devel‐
2
lations age. In the 1970s, Trunkey described a trimodal distribution opment of organ failure, and mortality.5,6 Death from hemorrhage is
of trauma deaths,3 with immediate deaths thought to be non‐pre‐ not the sole consequence of TIC. Owing to the innate crosstalk be‐
ventable and due to hemorrhage. However, since that time there tween coagulation and inflammation, there are widespread adverse
have been paradigm changes in our understanding of death due to downstream inflammatory and immune consequences associated
hemorrhage, including the recognition of trauma‐induced coagulop‐ with early trauma coagulopathies, including organ dysfunction and
athy (TIC), as well as advances in resuscitation techniques designed thromboembolic complications.6
4
to mitigate the effects of TIC. It is now clear that one‐quarter to Despite these focused investigations into the mechanisms and
one‐third of hemorrhaging trauma patients suffer from TIC, which mediators of TIC and improvements in care related to identification
and treatment, hemorrhage‐related mortality continues to be the
Manuscript handled by: Frits Rosendaal most common cause of preventable deaths after injury.7 Therefore,
Final decision: Frits Rosendaal, 3 April 2019 a targeted scientific focus on advancing our understanding of TIC

852  |  wileyonlinelibrary.com/journal/jth


© 2019 International Society on Thrombosis and J Thromb Haemost. 2019;17:852–862.
Haemostasis
KORNBLITH et al. |
      853

across molecular and clinical realms remains of the utmost impor‐ of prehospital crystalloid given to patients who developed TIC re‐
tance in order to save a large percentage of injured patients from quiring transfusion,17 there remains evidence that resuscitation and
death due to early hemorrhage and late organ failure. Although hypothermia continue to contribute to TIC,18 but are not necessary
substantial advances have been made, the links between the injury‐ prerequisites. However, patients who develop TIC are profoundly
induced impairments of each of the various mechanisms that con‐ more acidemic, supporting the pivotal importance of a shock state in
tribute to the failure of clot formation, lysis and vascular homeostasis the setting of tissue injury.17
remain areas of continuing investigation. In addition, the field is des‐ Finally, critical in the evolution of our understanding of TIC be‐
perate for clear causal and intervenable pathways between coagula‐ yond consumption of coagulation factors was the progress made in
tion, inflammation, and immune dysfunction. However, mechanistic defining coagulation outside of the classic enzymatic coagulation
studies related to TIC are fraught with the limitations of analyzing cascade. In 2001, Hoffman and Monroe proposed the ‘cell‐based
blood and vascular homeostasis in ex vivo environments that often model of hemostasis’ based on overlapping events of initiation (ex‐
lack endothelium, flow, and local tissue injury milieu, while measur‐ trinsic pathway on tissue factor‐bearing cells), amplification (posi‐
ing microvascular environments with macro‐level assays. The overall tive feedback of thrombin on platelets) and propagation of clotting
purpose of this review is to understand the past and present in order (intrinsic pathway on activated platelets) that were regulated by
to highlight the necessary future directions of this evolving field. cell surfaces rather than enzymatic protein and protease reactions
alone.19 This model of hemostasis has increased the amount of re‐
search focused on multiple pathways contributing to TIC, rather
2 | H I S TO RY than solely on a dilutional failure of the enzymatic processes of clot
formation, and a large body of investigation has contributed to the
Injury‐induced disordered clotting that is associated with bleeding current multidimensional understanding of the biological contribu‐
and death is not a modern concept. Historic battlefield descrip‐ tors to TIC, which are addressed specifically below and summarized
tions of this date back to the 18th century. During the Korean and in Table 1.
Vietnam wars, patients in hemorrhagic shock were identified as hav‐ However, despite years of basic and translational investiga‐
ing prolonged prothrombin and partial thromboplastin times early tions of TIC, in 2013 Gando and the Standardization Committee
after injury and prior to interventions, hemodilution, or hypother‐ on Disseminated Intravascular Coagulation of the International
mia, ultimately requiring more blood and having a higher mortality Society on Thrombosis and Haemostasis reported that what was
rate.8 What followed was the evolution of purported mechanisms of termed TIC was similar to consumptive states such as disseminated
this coagulopathy, including an initial focus on consumptive states intravascular coagulation (DIC). 20 However, the best evidence at
and the dilutional effects of resuscitation with fluids and blood this time suggests that, although TIC encompasses the criteria of
products. DIC, it remains distinct from and not sufficiently characterized
These observations of coagulopathy during major wars stimu‐ by the broad, less precise major (low platelet count, prolonged
lated significant military and civilian trauma research, and further prothrombin time, and increased levels of soluble fibrin or fibrin‐
critical concepts emerged, including the recognition of the clear degradation products) and specific (low antithrombin levels, low
transition from a clinically hypocoagulable to hypercoagulable state protein C levels, and increased thrombin‐antithrombin complex
and the central role that hypoperfusion due to shock played. Multiple levels) DIC criteria outlined by the Standardization Committee
groups simultaneously showed that hemorrhage and resuscitation on Disseminated Intravascular Coagulation of the International
induced depletion of coagulation factors, and that dilutional coagu‐ Society on Thrombosis and Haemostasis21 (Table 2). The definition
lopathy led to an irreversible physiologic collapse.9 These concepts of TIC has been evolving for decades, shifting from the perception
were formally combined into the ‘Bloody Vicious Cycle’ by Kashuk of a resuscitation‐induced dilutional coagulopathy, to a multifac‐
et al in the early 1980s.10 Following this, it was increasingly recog‐ torial and multimechanistic event. TIC is distinct from and more
nized that postinjury hemorrhage exacerbated by the ‘lethal triad’ of complex than DIC (Table 2), covering a wide range of impaired clot
hypothermia, acidosis and coagulopathy was a vicious cycle that led formation and lysis, in combination with failure of vascular homeo‐
to more coagulopathy and, ultimately, high mortality rates.11 This in‐ stasis and immunoactivation resulting in multiple clinical pheno‐
creasing recognition led to changes in clinical practice in the 1980s, typic states that can cause pathologic bleeding, clotting, organ
with the formalization of damage control surgical and resuscitative failure, and death.
techniques to halt the cycle of acidosis, hypothermia, and coagu‐
lopathy.12 In the early 2000s, Brohi, Cohen and colleagues formally
described the entity of acute traumatic coagulopathy in human and 3 | M EC H A N I S M S A N D M E D I ATO R S
animal models that reflected an endogenous biology independent
of resuscitation, required combined tissue injury and shock, and had Table 1 shows the estimated average quantitative values (mean or
6,13‒16
significant associations with poor outcomes and mortality. median) for each listed coagulation parameter in trauma patients
Although modern study of trauma patients has identified no sig‐ with TIC in the early and later periods after injury, and in those with‐
nificant difference in the presentation core temperature or amount out TIC, supported by existing literature, as cited.16,17,21‒31
|
854       KORNBLITH et al.

TA B L E 1   Estimated average coagulation parameters in trauma TA B L E 1   (Continued)


patients with trauma‐induced coagulopathy (TIC) early (0 h) and
later (beyond 0 h) after injury vs those without TIC TIC No TIC

TIC No TIC   Early Late Early


27,28
Endothelial markers
  Early Late Early
Soluble thrombomodu‐ 6.7 UND 4.7
Procoagulants16,17 lin (ng/mL)
Fibrinogen (ng/mL) 160 499 234 Von Willebrand factor 200 UND 175
Thrombin (%) 67 70 81 activity (%)
Factor V activity (%) 42 74 70 Syndecan‐1 (ng/mL) 108 UND 17
Factor VII activity (%) 75 76 95 Angiopoietin‐2 (pg/mL) 3000 UND 2000
Factor VIII activity (%) 200 180 275 Complement activation29,30
Factor IX activity (%) 104 185 129 C3a (ng/mg) 55 15 20
Factor X activity (%) 67 71 86 C5a 0.5 ng/mg 0.25 ng/ 0.20 ng/
mL mg
Plasminogen activator 25 19 38
inhibitor‐1 (ng/mL) sC5b‐9 0.18 ng/mg 0.025 μg/ 0.01 μg/
mg mg
Native thrombin generation22
s‐RAGE (pg/mL) 300 UND 275
Lag (min) 16 UND 15
Global hemostatic measures16,21,31
Peak (nmol/L) 163 UND 202
Rapid TEG ACT (s) 113 113 113
Area under the curve 3145 UND 3129
(nmol/L × min) Rapid TEG LY30 (physi‐ 1.6 1.6 1.6
16,17,23 ologic phenotype) (%)
Anticoagulants
Rapid TEG LY30 (hyper‐ 4.3 1.6 NA
Protein C activity (%) 81 74 95
fibrinolytic pheno‐
Activated protein C 12 1.2 2 type) (%)
(ng/mL)
Rapid TEG LY30 0.1 0.1 NA
Tissue plasminogen 28 4.5 15 (fibrinolytic shutdown
activator (ng/mL) phenotype) (%)
Thrombin‐antithrom‐ 190 75 37 Rapid TEG K time (min) 1.4 1.5 1.2
bin III complex (μg/L)
Rapid TEG angle (°) 73 72 75
Prothrombin frag‐ 25 15 5
Rapid TEG MA (mm) 61 62 65
ment 1 + 2 (nm/L)
Prothrombin time (s) 17 15 14
Antithrombin III (%) 78 74 89
Partial thromboplastin 38 39 28
D‐dimer (μg/L) 6.8 3.6 4.0
time (s)
Platelet biology24‒26
Abbreviations: AA, arachadonic acid; ACT, activated coagulation time;
Platelet count (×109/L) 224 95 227
GP, glycoprotein; HMGB‐1, high‐mobility group protein B1; LY30,
Soluble P‐selectin 13 7 2 percent lysis at 30 min; MA, maximum amplitude; NA, not applicable;
(CD62P+) (%) s‐RAGE, soluble receptor for advanced glycation endproducts; TEG,
GPIIb‐IIIa (PAC‐1) (%) 16 8 1 thromboelastography; TRAP, thrombin receptor‐activating peptide‐6;
UND, underdefined in previous studies.
Platelet ADP response 59 49 60 a
Estimated average quantitative values (mean or median) for each listed
(AU)
coagulation parameter in trauma patients with TIC at early and later
Platelet TRAP response 97 88 97 timepoints after injury vs those without TIC. Quantitative values are
(AU) supported by existing literature, as cited.
Platelet collagen re‐ 47 50 49
sponse (AU)
TEG‐platelet mapping 96 44 79
3.1 | Tissue injury and shock
ADP inhibition (%)
TEG‐platelet mapping 37 8 39 Impaired coagulation after injury was long thought to have only iat‐
AA inhibition (%) rogenic causes.9 For decades, trauma resuscitation practices guided
HMGB1 on plate‐ 13 17 7 by the luminaries of shock research were based on restoring flow
let surface and oxygen‐carrying capacity with large volumes of packed red
(CD42b+ HMGB1+) (%)
blood cells (oxygen) and crystalloid (flow). The gradual change in re‐
suscitation practice to larger and larger volumes being used for of
(Continues) cold anemic resuscitation resulted in an iatrogenic coagulopathy (IC)
KORNBLITH et al. |
      855

characterized by impaired thrombin production and platelet func‐ characterized by hypercoagulability and impaired cytoprotectivity.
9‒11
tion due to hypothermia, acidosis, and dilutional coagulopathy. On the basis of study of circulating aPC levels in trauma patients,16
Although these iatrogenic effects are still contributors to coagulopa‐ the cytoprotective level can be assumed to be somewhere in the
thy and can coexist with TIC, they are termed IC, are separate in range of 1.05‐6.00 ng/mL, as these levels correspond to preserva‐
biology from TIC, and are, fortunately, more infrequent with mod‐ tion of FVa, FVIIIa, and t‐PA levels, as well as lower D‐dimer lev‐
ern hemostatic and goal‐directed resuscitation techniques. An in‐ els. Conversely, the maladaptive level can be assumed to be in the
creasing body of work has now demonstrated that, to induce the >6.00 ng/mL range, as this level was shown to be associated with
mechanistic and clinical phenotypes of TIC, tissue injury (thought markedly decreased FVa and FVIIIa levels, as well as increased t‐PA
to activate the clotting cascade, produce thrombin, and stimulate and D‐dimer levels.16
resultant anticoagulant pathways) must be combined with tissue The transition from hypocoagulability to hypercoagulability oc‐
hypoperfusion (thought to release damage‐associated molecular curs very rapidly. Somewhere between 6 and 24 hours after injury,
patterns [DAMPs], activate the contact pathway, and induce throm‐ the majority of patients undergo this transition, with ≥90% being
bomodulin and endothelial protein C receptor [EPCR] expression at normal or hypercoagulable according to global hemostatic measures
the endothelial surface to activate protein C). It has become clear by 24 hours after injury, regardless of TIC.34 In addition, the coagu‐
that shock is essential for the development of the phenotypes of TIC lation factor activity in patients with and without TIC begins to re‐
via the multiple mediators described below. bound for most factors by 12 hours, with significantly higher activity
than baseline activity by 72 hours.17 The depletion of the non‐active
zymogen form of protein C is associated with both a transition to
3.2 | Activation and depletion of protein C
a thrombotic phenotype and, importantly, a loss of cytoprotective
One of the primary, and the first described, mechanisms of TIC is ac‐ signaling and resultant organ failure and infectious complications.16
13,14,16,32,33
tivation of protein C system. Protein C is a serine protease With newly engineered activated forms of protein  C that have
with dual anticoagulant functions: induction of proteolytic cleavage augmented cytoprotective function with little or no anticoagulant
of activated factor V (FVa) and activated FVIII (FVIIIa), and derepres‐ protease activity, as well as antibodies or aptamers against the an‐
sion of fibrinolysis through inhibition of plasminogen activator inhib‐ ticoagulant function of protein C, one can imagine that, in the near
itor‐1 (resulting in increased tissue‐type plasminogen activator [t‐PA] future, there may be resuscitation whereby we might inhibit the an‐
13,14,32
activity). Since the initial report that severe injury combined ticoagulant function and concurrently augment the cytoprotective
with shock resulted in increased levels of activated protein C (aPC), function of protein C immediately after trauma. Further precision
concomitant reductions in FV and FVIII, and increased fibrinolysis, a medicine modulation of the relative amounts of inhibition and aug‐
series of human studies and mechanistic animal and in vitro cell cul‐ mentation could be performed as patients move through their post‐
13‒16,32
ture models have confirmed these findings. The activation of injury coagulation and inflammation phases.
this pathway starts with tissue injury‐driven production of thrombin,
which combines with thrombomodulin, the EPCR. The non‐activated
3.3 | Factor depletion, impaired thrombin
zymogen form of protein C on the endothelial surface is then acti‐
generation, and fibrinogen deficiency
vated, resulting in cleavage of FVa and FVIIIa and derepression of
fibrinolysis. Multiple studies have corroborated that approximately Factor depletion does indeed exist after trauma.17 In addition, stud‐
25%‐33% of severely injured patients have elevated aPC levels, ies have demonstrated that clotting factor deficiencies occur without
which are associated with increased blood transfusion and mortal‐ significant fluid resuscitation, immediately after injury, and are asso‐
ity, and, in patients who survive, higher rates of infection and single ciated with worse outcomes.35 Although they are reproducible and
13,14,16,32
and multiple organ failure. correlate with the severity of injury and degree of shock, the factor
Interestingly, aPC also has cytoprotective functions, including level nadirs do not often reach the historically reported 20%‐30%
stabilization of endothelial and epithelial junctions, antiapoptosis, level at which coagulation should be impaired.17 Of course, the criti‐
and cleavage of extracellular histones. The enhanced activation cal values for factor depletion were derived from closed studies in
of the protein C system may therefore represent an evolutionary non‐activated (non‐injury) blood.36 Whether the reductions in factor
maladaptive response. As humans have not evolved to survive the levels do cause tissue‐specific impaired coagulation at the site of in‐
massive injury created by being shot, stabbed, and run over by auto‐ jury that requires levels higher than 20%‐30% in the setting of injury,
mobiles, it is hypothesized that the human physiologic response to and what circulating levels are ideal after significant injury for both
these events probably results in the activation of a large amount of tissue‐specific hemostasis and survival, remain open experimental
protein C in an attempt to activate inflammomodulatory pathways and clinical questions.
to enable survival after severe trauma. The unfortunate sequela of Clot formation requires thrombin generation on injured tissues.
this ‘too much of a good thing’ response is systemic and local anti‐ There are some suggestions that the measurement of thrombin gen‐
coagulation resulting in increased bleeding with the associated en‐ eration constitutes an optimal real‐time assessment of coagulation
hanced mortality and morbidity. For those who survive their initial capacity in the setting of injury, because thrombin generation does
injury, there is a rapid transition to a state of protein C depletion, not cease with fibrin deposition. However, both hypocoagulable and
|
856       KORNBLITH et al.

TA B L E 2   Qualitative changes in coagulation parameters in TA B L E 2   (Continued)


trauma‐induced coagulopathy (TIC) as compared with major and
specific criteria for disseminated intravascular coagulation (DIC)   TIC DIC
from the Scientific Subcommittee on Disseminated Intravascular sC5b‐9 ↑ NI
Coagulation of the International Society on Thrombosis and
s‐RAGE ↑ NI
Haemostasis21
Global hemostatic measures16,21,31
  TIC DIC
Rapid TEG ACT ↑ NI
16,17
Procoagulants Rapid TEG LY30 (physiologic → NI
Fibrinogen ↓ ↓ phenotype)

Thrombin ↓ NI Rapid TEG LY30 (hyperfibrinolytic ↑ NI


phenotype)
Factor V activity ↓ NI
Rapid TEG LY30 (fibrinolytic shut‐ ↓ NI
Factor VII activity ↓ NI
down phenotype)
Factor VIII activity ↓ NI
Rapid TEG K time ↑ NI
Factor IX ↓ NI
Rapid TEG angle ↓ NI
Factor X ↓ NI
Rapid TEG MA ↓ NI
Plasminogen activator inhibitor‐1 ↓ NI
Prothrombin time ↑ ↑
Native thrombin generation22
Partial thromboplastin time ↑ NI
Lag ↓ NI
Abbreviations: AA, arachadonic acid; ACT, activated coagulation time;
Peak ↑ NI
GP, glycoprotein; HMGB‐1, high‐mobility group protein B1; LY30, per‐
Area under the curve ↑ NI cent lysis at 30 min; MA, maximum amplitude; NI, not included; s‐RAGE,
Anticoagulants 16,17,23 soluble receptor for advanced glycation endproducts; TEG, thromboe‐
lastography; TRAP, thrombin receptor‐activating peptide‐6.
Protein C activity ↓ ↓ a
Qualitative changes for each listed coagulation parameter in trauma
Activated protein C ↑ NI patients with TIC vs major (bold) and specific (gray) criteria for DIC by
Tissue plasminogen activator ↑ NI the Scientific Subcommittee on Disseminated Intravascular Coagulation
of the International Society on Thrombosis and Haemostasis.
Thrombin‐antithrombin III complex ↑ ↑
Prothrombin fragment 1 + 2 ↑ NI
hypercoagulable thrombin generation profiles have been associated
Antithrombin III ↓ ↓
with trauma, with variable associations with bleeding and thrombo‐
D‐dimer ↑ ↑
embolic outcomes.37,38 Studies do suggest that there is sufficient
Platelet biology24‒26
thrombin generation in TIC, 22,38 but trauma patients with and with‐
Platelet count → ↓ out TIC show evidence of increased thrombin‐antithrombin complex
Soluble P‐selectin (CD62P+) ↑ NI and prothrombin fragment levels,39 and the biochemical milieu of
GPIIb‐IIIa (PAC‐1) ↑ NI injury and shock may increase thrombin generation because of de‐
Platelet ADP response ↓ NI creased antithrombin activity.40
Platelet TRAP response ↓ NI Fibrinogen is the terminal substrate for clot formation, and a low
Platelet collagen response ↓ NI quantity or low quality of fibrinogen in TIC is associated with bleed‐
TEG‐platelet mapping ADP ↑ NI ing and death.41,42 Animal models of TIC have confirmed low levels
inhibition of fibrinogen.43 Fibrinogen replacement in the form of cryoprecipi‐
TEG‐platelet mapping AA inhibition → NI tate or newer fibrinogen concentrates is used in many protocols for
HMGB1 on platelet surface ↑ NI the treatment of TIC,44 and often to maintain levels of fibrinogen
(CD42b+ HMGB1+) well above the standard triggers for fibrinogen replacement, de‐
Endothelial markers27,28 spite weak supporting evidence for this.45 Whether this technique
Soluble thrombomodulin ↑ NI improves outcomes continues to be a moving target, as goal‐di‐
Von Willebrand factor activity ↑ NI rected treatment of hypofibrinogenemia remains without clear ev‐

Syndecan‐1 ↑ NI
idence‐based treatment cutoffs, but with ever‐broadening therapies
beyond blood components.
Angiopoietin‐2 ↑ NI
Complement activation29,30
C3a ↑ NI 3.4 | Altered postinjury platelet biology
C5a ↑ NI
Although the pivotal importance of platelets in the overlapping
stages of clot formation is highlighted in the ‘cell‐based model of
(Continues) hemostasis’ of Hoffman and Monroe, the effect of local vs diffuse
KORNBLITH et al. |
      857

injury and shock states on platelet biology remains unclear in of postinjury hemorrhage. The standard of care in TIC is platelet
41,46‒48
TIC. However, platelets contribute significantly to the transfusion as part of a balanced resuscitation ratio (regardless
strength of clot formation in systemic blood during the postinjury of platelet count),7 but investigations have shown that not only
state. Kornblith et al demonstrated that the platelet contribution to does impaired postinjury platelet aggregation fail to predict the
clot strength is higher than that of fibrinogen at all time points after need for platelet transfusion, 58 but platelet transfusion does not
injury, by using functional assessment of whole blood in trauma pa‐ reverse postinjury impairment in platelet aggregation59 and does
41
tients. In addition, thrombocytopenia is a poor prognostic marker not improve outcomes for patients receiving antiplatelet medica‐
following injury, and is independently associated with transfusion, tions who have brain injury.60 Multiple studies have demonstrated
47
progression of brain injury, and death after injury. However, even that platelet transfusions may increase morbidity and mortality
with normal platelet counts, nearly half of injured patients show after injury. 59 The causative mechanisms for this are unknown, but,
impaired platelet aggregation in aggregometry assays immediately without doubt, further studies of TIC‐related postinjury platelet
after injury, 24,46 and this has been replicated in animal models of biology, platelet transfusions and alternative platelet therapies are
injury and hemorrhagic shock.49 Excessive platelet activation24 with required.61
subsequent exhaustion is one of the proposed mechanisms. This in‐
jury‐induced alteration in platelet biology has been found to have
3.5 | Dysregulated fibrinolysis
independent associations with brain injury, severe injury, and shock,
and all injury patterns consistent with significant endothelial dam‐ Local hypercoagulability promoting hemostasis at the site of injury
age. 24
Multiple studies have demonstrated increased morbidity and is proposed to activate systemic fibrinolysis to “guard against throm‐
mortality in patients who have impairments in platelet aggregation bosis” in remote non‐injured tissue.62 Elevated fibrinolysis meas‐
after injury. 24,46 ured with viscoelastic assays has repeatedly been demonstrated to
Numerous unresolved aspects of postinjury platelet biology be associated with a high mortality rate and massive transfusion in
remain to be addressed. For example, because of the nature of trauma, but is found in <20% of the most severely injured trauma
ex vivo aggregometry assays (lack of endothelium, lack of flow, patients.32,63 Although excessive fibrinolysis has pathologic conse‐
and exogenous agonist stimulation of platelets), it remains unclear quences, low fibrinolytic activity in animals has also been demon‐
where impaired postinjury platelet aggregation lies on the spec‐ strated to cause microvascular occlusion of vital organs, to cause
trum of adaptive to maladaptive responses to injury. It should not irreversible recovery from hemorrhagic shock,64 and to be reversible
be ignored that the majority of investigations in this area have re‐ by administration of a profibrinolytic agent.65
lied on point‐of‐care platelet function assays that were intended Moore et al showed that low fibrinolytic activity measured with
to assess the effects of antiplatelet medication on platelet inhi‐ viscoelastic assays, termed fibrinolysis shutdown, is also associated
bition, and it is of concern that viscoelastic assays do not always with increased mortality in severely injured patients.66 Low fibrino‐
correlate well with point‐of‐care assessment of platelet function lytic activity (defined as lysis at 30 minutes by thromboelastography
in trauma patients. 50 In fact, unpublished work by the authors of [LY30] of <0.9%) has been associated with increased mortality as
this review, using multiple electrode aggregometry, has shown compared with moderate levels of fibrinolysis (LY30 of 0.9%‐2.9%)
identified that platelets may be endogenously activated by injury, at multiple large‐volume trauma centers.66‒68 Fibrinolysis shutdown
diminishing their aggregation response to exogenous agonists. early after injury may be protective in some patients.68 This coagu‐
This raises concern that the identification of impaired platelet lation change could counterbalance the platelet inhibition and pro‐
aggregation after injury may detect multiple phenotypes of post‐ longed prothrombin time that has been described in patients with
injury platelet biology, including a physiologic response (platelets evidence of prior fibrinolytic activation.67,69 Historic and recent
activated by injury and unable to respond further to platelet‐ac‐ observations in trauma support the idea that patients can present
tivating agonists) and a maladaptive impairment in platelet aggre‐ to the hospital with a spectrum of fibrinolytic activity, whereby pa‐
gation. Therefore, future investigations need to use additional tients at the pathologic extremes are at risk of increased mortality.
methods to assess the health and function of the postinjury This concept has argued for the selective use of antifibrinolytics,
platelets by incorporating endothelium and flow (microfluidics), supported by a reduction in mortality by use in goal‐directed resus‐
assessment of structure (microscopy), mitochondrial health (mi‐ citation.70 Specifically, concerns have been raised that tranexamic
tochondrial respiration), and improved biomarkers of platelet and acid (TXA) may cause harm in certain trauma patients, in partic‐
endothelial function. 48,51‒53 ular patients with moderate (physiologic) levels of fibrinolysis.71
Additionally, given that platelet function extends beyond co‐ Patients who receive TXA are also at risk of prolonged fibrinolysis
agulation to bidirectional endothelial interaction and regulation, 54 inhibition. Fibrinolysis shutdown beyond 24 hours of injury is asso‐
control of local fibrinolysis at injury sites,19,55 and their role as ciated with increases in mortality and ventilator requirements,72,73
core effector cells in local and systemic inflammation, 56,57 a focus and Myers et al identified, in a propensity‐matched retrospective
on multiple areas of postinjury platelet biology should remain an review of a modern population of trauma patients, that TXA may be
active area of TIC research. Finally, an expanded understand‐ an independent risk factor for the development of venous throm‐
ing of postinjury platelet biology is critical to improving the care boembolism after injury.74 In aggregate, the evidence supports the
|
858       KORNBLITH et al.

need for improved understanding of complex biological and clinical


3.8 | DAMPs and others
interactions and outcomes related to TXA administration in trauma
patients. The spectrum of DAMPs following injury remains a confusing
mix of individual and combined mediators of biological interest,
including: cytokines, soluble receptor for advanced glycation end‐
3.6 | Inflammation and immune dysfunction:
products, high‐mobility group protein B1, tissue inhibitor metallo‐
endotheliopathy, DAMPs, and others
proteinase‐3, and numerous others. 26,30,88,89 These DAMPs have
Although the effects of trauma on the biochemistry of coagulation been measured and implicated in the sterile inflammation of trauma
constitute the subject of much important work, concurrent pro‐ and the crosstalk between coagulation and inflammation. 26,30,88,89
gress has been made in elucidating the effects of injury and shock Beyond this, many mechanisms abound: myosin may be implicated
on inflammatory and immune dysfunction. Emerging literature de‐ as a link between coagulation and inflammation biology and ef‐
scribes a conglomeration of sterile inflammatory and immune re‐ fects on TIC and fibrinolysis90 ; the contributions of metabolites to
sponses that center around the endothelium and that are related TIC and postinjury hemorrhage are probably underappreciated91;
but unique to biochemical coagulopathies of TIC. 52,75‒78 The injury and the relative levels of production of α‐thrombin and meizo‐
response establishes a cascade of inflammatory and immune dys‐ thrombin are being investigated as a switch between coagulation
function resulting in single and multiple organ failure (acute kidney and sterile inflammation after trauma.92 Indeed, whether each of
injury, acute respiratory distress syndrome, and hepatic dysfunc‐ these mediators has a true mechanistic effect or is just a correla‐
79,80
tion) and a higher susceptibility to infection. Much work has tive measure of injured patients remains an important question
been performed in sepsis models, but, more recently, bridging that will be answered only by a combination of both reductionist
work has been performed between coagulation and inflammation fundamental mechanistic biology and multivariate big‐data ap‐
in trauma. proaches to clinical data. As highlighted by Billiar in PLOS Medicine
in 201793 and at his 2019 Western Trauma Association Founders
Basic Science Lecture, both DAMP and pathogen‐associated mo‐
3.7 | Endotheliopathy
lecular pattern signaling are at the scientific frontier of TIC, and an
Kozar et al have identified degradation of the endothelial glycoca‐ expanded focus on trauma‐induced immune activation in response
lyx after trauma. Further work has suggested that a catecholamine to both sterile signals and microbial signals should guide the future
surge is associated with glycocalyx degradation and associated au‐ of this field.
toheparinization that can be identified in viscoelastic assays 81,82
In addition, clinical studies have shown increased levels of syn‐
decan‐1, a degradation product of the endothelial glycocalyx, to 4 | TH E FU T U R E
be associated with inflammation, coagulopathy, and mortality. 27
Importantly in relation to this, plasma has been shown to be re‐
4.1 | Improved assays
storative to the endothelial glycocalyx,79,83 but clinical studies
of early plasma administration have had mixed results regarding The evolution of the identification and measurement of TIC has fol‐
protection from morbidity and mortality. 84,85
Other pathways may lowed the evolution in mechanistic understanding. Originally, TIC
also contribute to the endotheliopathy of trauma. Multiple inves‐ was identified by focusing on single, static, ex vivo assessments of
tigators have reported cytoprotective functions of aPC, including the enzymatic coagulation cascade by measuring prolonged con‐
endothelial barrier protection, antiapoptosis, and cleavage of ex‐ ventional coagulation assays6,94; however, functional assessment of
tracellular histones. Kutcher et al showed that release of extracel‐ whole‐blood clot formation and degradation in real time via viscoe‐
lular histones in the setting of injury and shock is associated with lastic assays for both the identification and the management of TIC
organ failure and death, and the compensatory activation of pro‐ has become the standard of care.70,95 With the knowledge gained
tein C and subsequent clearance of histones was found to be pro‐ of the cellular and biochemical milieu created by combined tissue
tective. 86
This remains an active area of investigation, and there injury and shock, concerns have emerged regarding the ability to
are hypothesis‐generating investigations showing that endothe‐ use ex vivo conventional coagulation and viscoelastic assays for TIC
lial leakage primes neutrophil reactive oxygen species release interpretation, identification of “normal ranges” of assays, and guid‐
through the alternative pathway of complement, 87
which could be ance of therapy.
mediated by coagulation products. Given that cultured endothelial Furthermore, there exists little ability to characterize and dy‐
cells exposed to plasma from coagulopathic trauma patients have namically track the multiple competing and overlapping phenotypes
increased permeability consistent with endothelial barrier dys‐ that comprise TIC. Emerging evidence has highlighted that there are
function, suggesting that circulating mediators in trauma patients both clinical and biological phenotypes of TIC, and single assays are
can directly impair the endothelium, significant further work to therefore ineffective for the purposes of identification and treat‐
characterize, diagnose, and treat the endotheliopathy of trauma ment of TIC.96 In addition, the optimal combination of assays that
is needed. completely and effectively measures the complex and integrated
KORNBLITH et al. |
      859

pathways of all aspects of TIC‐related failure in coagulation, inflam‐ 3. Baker CC, Oppenheimer L, Stephens B, Lewis FR, Trunkey DD.
mation and vascular homeostasis are unknown and perhaps unde‐ Epidemiology of trauma deaths. Am J Surg. 1980;140:144–50.
4. Holcomb JB, Tilley BC, Baraniuk S, Fox EE, Wade CE, Podbielski
veloped. Current best evidence supports the idea that TIC can be
JM, et  al. Transfusion of plasma, platelets, and red blood cells
identified by abnormalities in either conventional coagulation assays in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with se‐
or viscoelastic assays, but not always both.96 Given this, combining vere trauma: the PROPPR randomized clinical trial. JAMA.
multiple assays for diagnosis and therapy in TIC is probably the opti‐ 2015;313:471–82.
5. Cohen MJ, Christie SA. New understandings of post injury coagu‐
mal strategy at present. Future development of improved methods,
lation and resuscitation. Int J Surg. 2016;33:242–5.
including in vivo biosensors of the biochemical milieu targeting as‐ 6. Brohi K, Singh J, Heron M, Coats T. Acute traumatic coagulopathy.
sessments of circulating cells in flow and shear environments with J Trauma. 2003;54:1127–30.
the contribution of hematocrit and endothelium,97 should constitute 7. Cannon JW. Hemorrhagic shock. N Engl J Med. 2018;378:370–9.
8. Simmons RL, Collins JA, Heisterkamp CA, Mills DE, Andren R,
a focus in the evolution of TIC science.
Phillips LL. Coagulation disorders in combat casualties. I. Acute
changes after wounding. II. Effects of massive transfusion. 3. Post‐
resuscitative changes. Ann Surg. 1969;169:455–82.
4.2 | In silico modeling
9. Dunn EL, Moore EE, Breslich DJ, Galloway WB. Acidosis‐induced
coagulopathy. Surg Forum. 1979;30:471–3.
Ultimately, however, we envision that biological assays will be rap‐
10. Kashuk JL, Moore EE, Millikan JS, Moore JB. Major abdominal vas‐
idly replaced by multivariate modeling of the complex coagulation cular trauma—a unified approach. J Trauma. 1982;22:672–9.
milieu after trauma. This will be augmented by computational mod‐ 11. Rossaint R, Cerny V, Coats TJ, Duranteau J, Fernandez‐Mondejar
els that can target reductionist precision medicine approaches to E, Gordini G, et al. Key issues in advanced bleeding care in trauma.
Shock. 2006;26:322–31.
resuscitation,98 while also predicting and characterizing dynamic
12. Stone HH, Strom PR, Mullins RJ. Management of the major coagu‐
phenotypes. This may allow for targeted prediction and optimal lopathy with onset during laparotomy. Ann Surg. 1983;197:532–5.
personalized treatment rules for clinicians to use on their individual 13. Brohi K, Cohen MJ, Ganter MT, Matthay MA, Mackersie RC,
patients.99,100 It is clear that we still have much to accomplish in the Pittet JF. Acute traumatic coagulopathy: initiated by hypoper‐
fusion: modulated through the protein  C pathway? Ann Surg.
open science of TIC to reduce morbidity and mortality for trauma
2007;245:812–18.
patients. 14. Cohen MJ, Brohi K, Ganter MT, Manley GT, Mackersie RC, Pittet
JF. Early coagulopathy after traumatic brain injury: the role of hy‐
poperfusion and the protein C pathway. J Trauma. 2007;63:1254–
AC K N OW L E D G M E N T S 61; discussion 61‐2.
15. Chesebro BB, Rahn P, Carles M, Esmon CT, Xu J, Brohi K, et  al.
This work was supported by grants from the Department of Increase in activated protein C mediates acute traumatic coagu‐
Defense (DoD W911QY‐15‐C‐0044) (M. J. Cohen) and the National lopathy in mice. Shock. 2009;32:659–65.
Institutes of Health (NIH UM1HL120877) (M. J. Cohen), the Eastern 16. Cohen MJ, Call M, Nelson M, Calfee CS, Esmon CT, Brohi K, et al.
Critical role of activated protein C in early coagulopathy and later
Association for the Surgery of Trauma, Trauma Research Scholarship
organ failure, infection and death in trauma patients. Ann Surg.
(P0526402) (L. Z. Kornblith), and the National Institutes of Health 2012;255:379–85.
(NIH 1K23GM130892‐01) (L. Z. Kornblith). 17. Kutcher ME, Kornblith LZ, Vilardi RF, Redick BJ, Nelson MF,
Cohen MJ. The natural history and effect of resuscitation ratio on
coagulation after trauma: a prospective cohort study. Ann Surg.
AU T H O R C O N T R I B U T I O N S 2014;260:1103–11.
18. Lester ELW, Fox EE, Holcomb JB, Brasel KJ, Bulger EM, Cohen MJ,
L. Z. Kornblith, H. B. Moore and M. J. Cohen contributed equally to et al.; PROPPR study group. The impact of hypothermia on out‐
this review. This manuscript is original work, has not been previously comes in massively transfused patients. J Trauma Acute Care Surg.
2019;86:458–63.
published, and is not under consideration for publication elsewhere.
19. Hoffman M, Monroe DM 3rd. A cell‐based model of hemostasis.
The manuscript has been read and approved for submission to JTH Thromb Haemost. 2001;85:958–65.
by all qualified authors. H. B. Moore is a Co‐Founder and Board 20. Gando S, Wada H, Thachil J; Scientific and Standardization
Member of Thrombo Therapeutics Incorporated, and has served on Committee on DIC of the International Society on Thrombosis
and Haemostasis (ISTH). Differentiating disseminated intravascu‐
the Advisory Committee for Instrument Laboratories. M. J. Cohen
lar coagulation (DIC) with the fibrinolytic phenotype from coagu‐
and L. Z. Kornblith have no actual or potential conflicts of interest lopathy of trauma and acute coagulopathy of trauma‐shock (COT/
capable of influencing their judgment. ACOTS). J Thromb Haemost. 2013;11:826–35.
21. Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M; Scientific
Subcommittee on Disseminated Intravascular Coagulation (DIC) of
REFERENCES the International Society on Thrombosis and Haemostasis (ISTH).
Towards definition, clinical and laboratory criteria, and a scor‐
1. Norton R, Kobusingye O. Injuries. N Engl J Med. 2013;368:1723–30. ing system for disseminated intravascular coagulation. Thromb
2. Rhee P, Joseph B, Pandit V, Aziz H, Vercruysse G, Kulvatunyou Haemost. 2001;86:1327–30.
N, et al. Increasing trauma deaths in the United States. Ann Surg. 22. Dunbar NM, Chandler WL. Thrombin generation in trauma pa‐
2014;260:13–21. tients. Transfusion. 2009;49:2652–60.
|
860       KORNBLITH et al.

23. Scherer RU, Spangenberg P. Procoagulant activity in patients with 41. Kornblith LZ, Kutcher ME, Redick BJ, Calfee CS, Vilardi RF, Cohen
isolated severe head trauma. Crit Care Med. 1998;26:149–56. MJ. Fibrinogen and platelet contributions to clot formation: im‐
24. Jacoby RC, Owings JT, Holmes J, Battistella FD, Gosselin RC, plications for trauma resuscitation and thromboprophylaxis. J
Paglieroni TG. Platelet activation and function after trauma. J Trauma Acute Care Surg. 2014;76:255–6; discussion 62‐3.
Trauma. 2001;51:639–47. 42. Rourke C, Curry N, Khan S, Taylor R, Raza I, Davenport R, et al.
25. Kornblith LZ, Robles AJ, Conroy AS, Hendrickson CM, Calfee CS, Fibrinogen levels during trauma hemorrhage, response to replace‐
Fields AT, et  al. Perhaps it's not the platelet: ristocetin uncovers ment therapy, and association with patient outcomes. J Thromb
the potential role of von Willebrand factor in impaired platelet ag‐ Haemost. 2012;10:1342–51.
gregation following traumatic brain injury. J Trauma Acute Care 43. Martini WZ, Holcomb JB. Acidosis and coagulopathy: the differ‐
Surg. 2018;85:873–80. ential effects on fibrinogen synthesis and breakdown in pigs. Ann
26. Vogel S, Bodenstein R, Chen Q, Feil S, Feil R, Rheinlaender J, et al. Surg. 2007;246:831–5.
Platelet‐derived HMGB1 is a critical mediator of thrombosis. J Clin 44. Schochl H, Nienaber U, Hofer G, Voelckel W, Jambor C, Scharbert
Invest. 2015;125:4638–54. G, et al. Goal‐directed coagulation management of major trauma
27. Gonzalez Rodriguez E, Ostrowski SR, Cardenas JC, Baer LA, patients using thromboelastometry (ROTEM)‐guided administra‐
Tomasek JS, Henriksen HH, et  al. Syndecan‐1: a quantitative tion of fibrinogen concentrate and prothrombin complex concen‐
marker for the endotheliopathy of trauma. J Am Coll Surg. trate. Crit Care. 2010;14:R55.
2017;225:419–27. 45. Rossaint R, Bouillon B, Cerny V, Coats TJ, Duranteau J, Fernandez‐
28. Ganter MT, Cohen MJ, Brohi K, Chesebro BB, Staudenmayer KL, Mondejar E, et  al.; Task Force for Advanced Bleeding Care in
Rahn P, et al. Angiopoietin‐2, marker and mediator of endothelial Trauma. Management of bleeding following major trauma: an up‐
activation with prognostic significance early after trauma? Ann dated European guideline. Crit Care. 2010;14:R52.
Surg. 2008;247:320–6. 46. Kutcher ME, Redick BJ, McCreery RC, Crane IM, Greenberg MD,
29. Burk AM, Martin M, Flierl MA, Rittirsch D, Helm M, Lampl L, et al. Cachola LM, et al. Characterization of platelet dysfunction after
Early complementopathy after multiple injuries in humans. Shock. trauma. J Trauma Acute Care Surg. 2012;73:13–19.
2012;37:348–54. 47. Brown LM, Call MS, Margaret Knudson M, Cohen MJ, Trauma
30. Cohen MJ, Carles M, Brohi K, Calfee CS, Rahn P, Call MS, et  al. Outcomes G, Holcomb JB, et al. A normal platelet count may not
Early release of soluble receptor for advanced glycation endprod‐ be enough: the impact of admission platelet count on mortality
ucts after severe trauma in humans. J Trauma. 2010;68:1273–8. and transfusion in severely injured trauma patients. J Trauma.
31. Roberts DJ, Kalkwarf KJ, Moore HB, Cohen MJ, Fox EE, Wade 2011;71:S337–42.
CE, et al. Time course and outcomes associated with transient 48. Li R, Elmongy H, Sims C, Diamond SL. Ex vivo recapitulation of
versus persistent fibrinolytic phenotypes after injury: a nested, trauma‐induced coagulopathy and preliminary assessment of
prospective, multicenter cohort study. J Trauma Acute Care Surg. trauma patient platelet function under flow using microfluidic
2019;86:206–13. technology. J Trauma Acute Care Surg. 2016;80:440–9.
32. Davenport RA, Guerreiro M, Frith D, Rourke C, Platton S, Cohen 49. Sillesen M, Johansson PI, Rasmussen LS, Jin G, Jepsen CH, Imam
M, et al. Activated protein C drives the hyperfibrinolysis of acute AM, et  al. Platelet activation and dysfunction in a large‐animal
traumatic coagulopathy. Anesthesiology. 2017;126:115–27. model of traumatic brain injury and hemorrhage. J Trauma Acute
33. Howard BM, Kornblith LZ, Cheung CK, Kutcher ME, Miyazawa BY, Care Surg. 2013;74:1252–9.
Vilardi RF, et al. Inducing acute traumatic coagulopathy in vitro: 50. Martin G, Shah D, Elson N, Boudreau R, Hanseman D, Pritts TA,
the effects of activated protein C on healthy human whole blood. et al. Relationship of coagulopathy and platelet dysfunction to
PLoS One. 2016;11:e0150930. transfusion needs after traumatic brain injury. Neurocrit Care.
34. Sumislawski JJ, Kornblith LZ, Conroy AS, Callcut RA, Cohen MJ. 2018;28:330–7.
Dynamic coagulability after injury: is delaying venous thrombo‐ 51. Baimukanova G, Miyazawa B, Potter DR, Muench MO, Bruhn R,
embolism chemoprophylaxis worth the wait? J Trauma Acute Care Gibb SL, et al. Platelets regulate vascular endothelial stability: as‐
Surg. 2018;85:907–14. sessing the storage lesion and donor variability of apheresis plate‐
35. Rizoli SB, Scarpelini S, Callum J, Nascimento B, Mann KG, Pinto R, lets. Transfusion. 2016;56(Suppl. 1):S65–75.
et al. Clotting factor deficiency in early trauma‐associated coagu‐ 52. Johansson PI, Henriksen HH, Stensballe J, Gybel‐Brask M,
lopathy. J Trauma. 2011;71:S427–34. Cardenas JC, Baer LA, et  al. Traumatic endotheliopathy: a pro‐
36. Dzik WH. The James Blundell Award Lecture 2006: transfusion spective observational study of 424 severely injured patients. Ann
and the treatment of haemorrhage: past, present and future. Surg. 2017;265:597–603.
Transfus Med. 2007;17:367–74. 53. Brouns SLN, van Geffen JP, Heemskerk JWM. High‐throughput
37. Park MS, Xue A, Spears GM, Halling TM, Ferrara MJ, Kuntz MM, measurement of human platelet aggregation under flow: applica‐
et al. Thrombin generation and procoagulant microparticle profiles tion in hemostasis and beyond. Platelets. 2018;29:662–9.
after acute trauma: a prospective cohort study. J Trauma Acute 54. Kornblith LZ, Robles AJ, Conroy AS, Hendrickson CM, Calfee CS,
Care Surg. 2015;79:726–31. Fields AT, et  al. Perhaps it's not the platelet: ristocetin uncovers
38. Cardenas JC, Rahbar E, Pommerening MJ, Baer LA, Matijevic N, the potential role of von Willebrand factor in impaired platelet ag‐
Cotton BA, et al. Measuring thrombin generation as a tool for pre‐ gregation following traumatic brain injury. J Trauma Acute Care
dicting hemostatic potential and transfusion requirements follow‐ Surg. 2018;85:873–80.
ing trauma. J Trauma Acute Care Surg. 2014;77:839–45. 55. Mazepa M, Hoffman M, Monroe D. Superactivated platelets:
39. Shaz BH, Winkler AM, James AB, Hillyer CD, MacLeod JB. thrombus regulators, thrombin generators, and potential clinical
Pathophysiology of early trauma‐induced coagulopathy: emerg‐ targets. Arterioscler Thromb Vasc Biol. 2013;33:1747–52.
ing evidence for hemodilution and coagulation factor depletion. J 56. Tweardy DJ, Khoshnevis MR, Yu B, Mastrangelo MA, Hardison EG,
Trauma. 2011;70:1401–7. Lopez JA. Essential role for platelets in organ injury and inflamma‐
40. Gissel M, Brummel‐Ziedins KE, Butenas S, Pusateri AE, Mann KG, tion in resuscitated hemorrhagic shock. Shock. 2006;26:386–90.
Orfeo T. Effects of an acidic environment on coagulation dynam‐ 57. Ding N, Chen G, Hoffman R, Loughran PA, Sodhi CP, Hackam DJ,
ics. J Thromb Haemost. 2016;14:2001–10. et al. Toll‐like receptor 4 regulates platelet function and contributes
KORNBLITH et al. |
      861

to coagulation abnormality and organ injury in hemorrhagic shock activator inhibitor‐1 and postinjury complications. Blood.
and resuscitation. Circ Cardiovasc Genet. 2014;7:615–24. 2016;128:206.
58. Stettler GR, Moore EE, Moore HB, Nunns GR, Huebner BR, 74. Myers SP, Kutcher ME, Rosengart MR, Sperry JL, Peitzman AB,
Einersen P, et al. Platelet adenosine diphosphate receptor inhibi‐ Brown JB, et al. Tranexamic acid administration is associated with
tion provides no advantage in predicting need for platelet transfu‐ an increased risk of posttraumatic venous thromboembolism. J
sion or massive transfusion. Surgery. 2017;162:1286–94. Trauma Acute Care Surg. 2019;86:20–7.
59. Henriksen HH, Grand AG, Viggers S, Baer LA, Solbeck S, Cotton 75. Naumann DN, Hazeldine J, Davies DJ, Bishop J, Midwinter MJ,
BA, et  al. Impact of blood products on platelet function in pa‐ Belli A, et al. Endotheliopathy of trauma is an on‐scene phenome‐
tients with traumatic injuries: a translational study. J Surg Res. non, and is associated with multiple organ dysfunction syndrome:
2017;214:154–61. a prospective observational study. Shock. 2017;49:420–428.
60. Holzmacher JL, Reynolds C, Patel M, Maluso P, Holland S, Gamsky 76. Ostrowski SR, Henriksen HH, Stensballe J, Gybel‐Brask M,
N, et al. Platelet transfusion does not improve outcomes in pa‐ Cardenas JC, Baer LA, et  al. Sympathoadrenal activation and
tients with brain injury on antiplatelet therapy. Brain Injury. endotheliopathy are drivers of hypocoagulability and hyperfi‐
2018;32:325–30. brinolysis in trauma: a prospective observational study of 404
61. Shukla M, Sekhon UD, Betapudi V, Li W, Hickman DA, Pawlowski severely injured patients. J Trauma Acute Care Surg. 2017;82:293
CL, et al. In vitro characterization of SynthoPlate (synthetic plate‐ –301.
let) technology and its in vivo evaluation in severely thrombocyto‐ 77. Pati S, Potter DR, Baimukanova G, Farrel DH, Holcomb JB,
penic mice. J Thromb Haemost. 2017;15:375–87. Schreiber MA. Modulating the endotheliopathy of trauma: factor
62. Innes D, Sevitt S. Coagulation and fibrinolysis in injured patients. J concentrate versus fresh frozen plasma. J Trauma Acute Care Surg.
Clin Pathol. 1964;17:1–13. 2016;80:576–84; discussion 84‐5.
63. Cotton BA, Harvin JA, Kostousouv V, Minei KM, Radwan ZA, 78. Rodriguez EG, Cardenas JC, Lopez E, Cotton BA, Tomasek JS,
Schochl H, et al. Hyperfibrinolysis at admission is an uncommon Ostrowski SR, et al. Early identification of the patient with
but highly lethal event associated with shock and prehospital fluid endotheliopathy of trauma by arrival serum albumin. Shock.
administration. J Trauma Acute Care Surg. 2012;73:365–70; dis‐ 2017;50:31–37.
cussion 70. 79. Kozar RA, Pati S. Syndecan‐1 restitution by plasma after hemor‐
64. Hardaway RM, Chun B, Rutherford RB. Histologic evidence of rhagic shock. J Trauma Acute Care Surg. 2015;78:S83–6.
disseminated intravascular coagulation in clinical shock. Vasc Dis. 80. Wu F, Peng Z, Park PW, Kozar RA. Loss of syndecan‐1 abrogates
1965;2:254–65. the pulmonary protective phenotype induced by plasma after
65. Hardaway RM, Drake DC. Prevention of “irreversible” hemor‐ hemorrhagic shock. Shock. 2017;48:340–5.
rhagic shock with fibrinolysin. Ann Surg. 1963;157:39–47. 81. Johansson PI, Stensballe J, Rasmussen LS, Ostrowski SR. High cir‐
66. Moore HB, Moore EE, Gonzalez E, Chapman MP, Chin TL, Silliman culating adrenaline levels at admission predict increased mortality
CC, et al. Hyperfibrinolysis, physiologic fibrinolysis, and fibri‐ after trauma. J Trauma Acute Care Surg. 2012;72:428–36.
nolysis shutdown: the spectrum of postinjury fibrinolysis and 82. Ostrowski SR, Johansson PI. Endothelial glycocalyx degradation
relevance to antifibrinolytic therapy. J Trauma Acute Care Surg. induces endogenous heparinization in patients with severe in‐
2014;77:811–17; discussion 7. jury and early traumatic coagulopathy. J Trauma Acute Care Surg.
67. Cardenas JC, Wade CE, Cotton BA, George MJ, Holcomb JB, 2012;73:60–6.
Schreiber MA, et  al.; PROPPR Study Group. TEG lysis shutdown 83. Pati S, Matijevic N, Doursout MF, Ko T, Cao Y, Deng X, et  al.
represents coagulopathy in bleeding trauma patients: analysis of Protective effects of fresh frozen plasma on vascular endothelial
the PROPPR cohort. Shock. 2019;51:273–83. permeability, coagulation, and resuscitation after hemorrhagic
68. Gomez‐Builes JC, Acuna SA, Nascimento B, Madotto F, Rizoli shock are time dependent and diminish between days 0 and 5 after
SB. Harmful or physiologic: diagnosing fibrinolysis shutdown in a thaw. J Trauma. 2010;69(Suppl. 1):S55–63.
trauma cohort with rotational thromboelastometry. Anesth Analg. 84. Moore HB, Moore EE, Chapman MP, McVaney K, Bryskiewicz G,
2018;127:840–9. Blechar R, et  al. Plasma‐first resuscitation to treat haemorrhagic
69. Moore HB, Moore EE, Huebner BR, Dzieciatkowska M, Stettler shock during emergency ground transportation in an urban area: a
GR, Nunns GR, et al. Fibrinolysis shutdown is associated with a randomised trial. Lancet. 2018;392:283–91.
fivefold increase in mortality in trauma patients lacking hypersen‐ 85. Sperry JL, Guyette FX, Brown JB, Yazer MH, Triulzi DJ, Early‐
sitivity to tissue plasminogen activator. J Trauma Acute Care Surg. Young BJ, et  al. Prehospital plasma during air medical transport
2017;83:1014–22. in trauma patients at risk for hemorrhagic shock. N Engl J Med.
70. Gonzalez E, Moore EE, Moore HB, Chapman MP, Chin TL, 2018;379:315–26.
Ghasabyan A, et  al. Goal‐directed hemostatic resuscitation of 86. Kutcher ME, Xu J, Vilardi RF, Ho C, Esmon CT, Cohen MJ.
trauma‐induced coagulopathy: a pragmatic randomized clinical Extracellular histone release in response to traumatic injury: im‐
trial comparing a viscoelastic assay to conventional coagulation plications for a compensatory role of activated protein C. J Trauma
assays. Ann Surg. 2016;263:1051–9. Acute Care Surg. 2012;73:1389–94.
71. Moore HB, Moore EE, Huebner BR, Stettler GR, Nunns GR, 87. Boueiz A, Hassoun PM. Regulation of endothelial barrier func‐
Einersen PM, et al. Tranexamic acid is associated with increased tion by reactive oxygen and nitrogen species. Microvasc Res.
mortality in patients with physiological fibrinolysis. J Surg Res. 2009;77:26–34.
2017;220:438–43. 88. Ganter MT, Brohi K, Cohen MJ, Shaffer LA, Walsh MC, Stahl GL,
72. Meizoso JP, Karcutskie CA, Ray JJ, Namias N, Schulman CI, Proctor et al. Role of the alternative pathway in the early complement ac‐
KG. Persistent fibrinolysis shutdown is associated with increased tivation following major trauma. Shock. 2007;28:29–34.
mortality in severely injured trauma patients. J Am Coll Surg. 89. Cohen MJ, Brohi K, Calfee CS, Rahn P, Chesebro BB, Christiaans
2017;224:575–82. SC, et al. Early release of high mobility group box nuclear pro‐
73. Moore HBME, Gonzalez E, Huebner BJ, Sheppard F, Banerjee A, tein 1 after severe trauma in humans: role of injury severity and
Sauaia A, et  al. Reperfusion shutdown: delayed onset of fibri‐ tissue hypoperfusion. Crit Care. 2009;13:R174.
nolysis resistance after resuscitation from hemorrhagic shock 90. Deguchi H, Sinha RK, Marchese P, Ruggeri ZM, Zilberman‐
is associated with increased circulating levels of plasminogen Rudenko J, McCarty OJT, et  al. Prothrombotic skeletal muscle
862      | KORNBLITH et al.

myosin directly enhances prothrombin activation by binding fac‐ 98. Menezes AA, Vilardi RF, Arkin AP, Cohen MJ. Targeted clinical
tors Xa and Va. Blood. 2016;128:1870–8. control of trauma patient coagulation through a thrombin dynam‐
91. Wiener G, Moore HB, Moore EE, Gonzalez E, Diamond S, Zhu S, ics model. Sci Transl Med. 2017;9:pii:eaaf5045.
et al. Shock releases bile acid inducing platelet inhibition and fibri‐ 99. Moore SE, Decker A, Hubbard A, Callcut RA, Fox EE, Del Junco
nolysis. J Surg Res. 2015;195:390–5. DJ, et al.; PROMMTT Study Group. Statistical machines for trauma
92. Whelihan MF, Mann KG. The role of the red cell membrane in hospital outcomes research: application to the PRospective,
thrombin generation. Thromb Res. 2013;131:377–82. Observational, Multi‐Center Major Trauma Transfusion (PROMMTT)
93. Billiar TR, Vodovotz Y. Time for trauma immunology. PLoS Med. Study. PLoS One. 2015;10:e0136438.
2017;14:e1002342. 100. Hubbard A, Munoz ID, Decker A, Holcomb JB, Schreiber MA,
94. MacLeod JB, Lynn M, McKenney MG, Cohn SM, Murtha M. Bulger EM, et al.; PROMMTT Study Group. Time‐dependent pre‐
Early coagulopathy predicts mortality in trauma. J Trauma. diction and evaluation of variable importance using superlearn‐
2003;55:39–44. ing in high‐dimensional clinical data. J Trauma Acute Care Surg.
95. Prat NJ, Meyer AD, Ingalls NK, Trichereau J, DuBose JJ, Cap AP. 2013;75:S53–60.
Rotational thromboelastometry significantly optimizes transfu‐
sion practices for damage control resuscitation in combat casual‐
ties. J Trauma Acute Care Surg. 2017;83:373–80.
96. Christie SA, Kornblith LZ, Howard BM, Conroy AS, Kunitake How to cite this article: Kornblith LZ, Moore HB, Cohen MJ.
RC, Nelson MF, et al. Characterization of distinct coagulopathic Trauma‐induced coagulopathy: The past, present, and future. J
phenotypes in injury: pathway‐specific drivers and implica‐ Thromb Haemost. 2019;17:852–862. https​://doi.org/10.1111/
tions for individualized treatment. J Trauma Acute Care Surg.
jth.14450​
2017;82:1055–62.
97. Rong G, Corrie SR, Clark HA. In  vivo biosensing: progress and
perspectives. ACS Sens. 2017;2:327–38.

You might also like