Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component...

11
REVIEW ARTICLES Paul G. Barash, MD Giovanni Landoni, MD Section Editors Rotational Thromboelastometry (ROTEM)-Based Coagulation Management in Cardiac Surgery and Major Trauma Kenichi A. Tanaka, MD, MSc,* Daniel Bolliger, MD,† Ratna Vadlamudi, MD,‡ and Alastair Nimmo, MB§ F OR MAJOR BLEEDING related to severe trauma, major surgery, or chronic anticoagulation, a rapid assessment of hemostatic function is crucial so that optimal fluid replace- ments and blood transfusion can be administered without de- lays. 1-6 Although the safety of blood products with regard to viral transmission risks has improved in recent years, 7,8 trans- fusions of allogeneic erythrocyte and plasma products have been implicated in serious adverse events, including nosoco- mial infections, acute lung injury, and organ dysfunction. 9-12 Obtaining conventional laboratory tests, such as the prothrom- bin time (PT), activated partial thromboplastin time (aPTT), and fibrinogen level, during acute bleeding is difficult because of a long turn-around time (30 min). 13,14 Furthermore, labo- ratory PT/international normalized ratio and aPTT may not be particularly useful in predicting bleeding after trauma or inva- sive procedures. 15,16 The prime example of bleeding management is preemptive transfusions of fresh-frozen plasma (FFP) and platelet concen- trates according to the erythrocyte requirement in major trauma cases. 17,18 This so-called “damage control resuscitation” (DCR; Table 1) originally was advocated for battlefield resuscitation in which laboratory testing and transfusion resources were limited. However, plasma product transfusion according to DCR became increasingly popular in US civilian trauma cen- ters and operating rooms. 17,19 The prevention of trauma-in- duced coagulopathy and subsequent nonsurgical bleeding is a major advantage of DCR, 20 but the DCR approach lacks a specific target for replacement and a consideration for interin- dividual variability in coagulation factor levels and vascular (endothelial) responses. Implementing transfusion algorithms based on point-of-care (POC) coagulation testing can be effec- tive in decreasing transfusion requirements in elective or urgent cardiac surgical settings. 2,5,21-23 In this review, the practical use of thromboelastometry is discussed relating to the diagnosis of coagulopathy and optimizing hemostatic interventions. POC TESTING AND TIMING OF HEMOSTATIC INTERVENTION PT (or international normalized ratio), aPTT, fibrinogen level (Clauss method and its modifications), and platelet count are the tests performed most commonly in managing perioper- ative bleeding. Except for the platelet count, these laboratory tests require a separation of plasma from whole blood before testing, and, thus, a typical turn-around time is in the range of 30-90 minutes. 13,24 Abnormalities detected in these tests are followed by requests for specific blood components. The choice of hemostatic therapies also affects the lag time before inter- vention. The time required for thawing FFP and cryoprecipitate is typically 30-60 minutes, but less time is needed for platelet concentrates and thawed plasma. Factor concentrates, such as fibrinogen concentrate, and recombinant activated factor VII (rFVIIa) can be administered rapidly (10 min) because they are reconstituted in small volumes and infused irrespective of blood type. One of the key facts behind DCR is to prevent the delay of transfusion therapy for patients in whom the risk of hemor- rhagic death is considerably greater than transfusion-associated complications. 25 However, a substantial number of patients may receive allogeneic plasma products inappropriately or in excess, which collectively increase the risk for transfusion- related adverse events. 26 By implementing rapid POC coagu- lation testing, hemostatic interventions can be more individu- alized and goal directed (targeted) instead of indiscriminate applications of DCR. Currently available POC coagulation tests are classified into 3 categories. The first category includes POC whole-blood PT and aPTT for a rapid evaluation (5-10 min) of plasmatic coagulation. POC PT has been shown to have a reasonable agreement with plasma-based PT. 13,27,28 However, PT and aPTT are sensitive only to severe hypofibrinogenemia (60-70 mg/dL) and are insensitive to FXIII deficiency or From the *Department of Anesthesiology, University of Pittsburgh Medical Center, Pittsburgh, PA; †Department of Anesthesia and Inten- sive Care Medicine, University of Basel Hospital, Basel, Switzerland; ‡Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA; and §Royal Infirmary of Edinburgh, Edinburgh, UK. Dr Bolliger was supported by a Myron B. Laver Grant, Department of Anaesthesia, University of Basel, Switzerland. Drs Tanaka, Bolliger, and Nimmo have received honoraria for consultation and/or lectures from TEM International (Munich, Germany). Address reprint requests to Kenichi A. Tanaka, MD, MSc, Visiting Professor of Anesthesiology, Department of Anesthesiology, UPMC Presbyterian C-215, 200 Lothrop Street, Pittsburgh, PA 15213. E-mail: [email protected] © 2012 Elsevier Inc. All rights reserved. 1053-0770/2606-0022$36.00/0 http://dx.doi.org/10.1053/j.jvca.2012.06.015 Key words: rotational thromboelastometry, coagulation monitoring, blood component transfusion, cardiac surgery, trauma 1083 Journal of Cardiothoracic and Vascular Anesthesia, Vol 26, No 6 (December), 2012: pp 1083-1093

Transcript of Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component...

Page 1: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

rps

TilDt

J

REVIEW ARTICLESPaul G. Barash, MD

Giovanni Landoni, MDSection Editors

Rotational Thromboelastometry (ROTEM)-Based Coagulation Management inCardiac Surgery and Major Trauma

Kenichi A. Tanaka, MD, MSc,* Daniel Bolliger, MD,† Ratna Vadlamudi, MD,‡ and Alastair Nimmo, MB§

3

merlaaawpr

FOR MAJOR BLEEDING related to severe trauma, majorsurgery, or chronic anticoagulation, a rapid assessment of

hemostatic function is crucial so that optimal fluid replace-ments and blood transfusion can be administered without de-lays.1-6 Although the safety of blood products with regard toviral transmission risks has improved in recent years,7,8 trans-fusions of allogeneic erythrocyte and plasma products havebeen implicated in serious adverse events, including nosoco-mial infections, acute lung injury, and organ dysfunction.9-12

Obtaining conventional laboratory tests, such as the prothrom-bin time (PT), activated partial thromboplastin time (aPTT),and fibrinogen level, during acute bleeding is difficult becauseof a long turn-around time (�30 min).13,14 Furthermore, labo-atory PT/international normalized ratio and aPTT may not bearticularly useful in predicting bleeding after trauma or inva-ive procedures.15,16

The prime example of bleeding management is preemptivetransfusions of fresh-frozen plasma (FFP) and platelet concen-trates according to the erythrocyte requirement in major traumacases.17,18 This so-called “damage control resuscitation” (DCR;

able 1) originally was advocated for battlefield resuscitationn which laboratory testing and transfusion resources wereimited. However, plasma product transfusion according toCR became increasingly popular in US civilian trauma cen-

ers and operating rooms.17,19 The prevention of trauma-in-duced coagulopathy and subsequent nonsurgical bleeding is amajor advantage of DCR,20 but the DCR approach lacks aspecific target for replacement and a consideration for interin-dividual variability in coagulation factor levels and vascular(endothelial) responses. Implementing transfusion algorithmsbased on point-of-care (POC) coagulation testing can be effec-tive in decreasing transfusion requirements in elective or urgentcardiac surgical settings.2,5,21-23 In this review, the practical useof thromboelastometry is discussed relating to the diagnosis ofcoagulopathy and optimizing hemostatic interventions.

POC TESTING AND TIMING OFHEMOSTATIC INTERVENTION

PT (or international normalized ratio), aPTT, fibrinogenlevel (Clauss method and its modifications), and platelet countare the tests performed most commonly in managing perioper-ative bleeding. Except for the platelet count, these laboratorytests require a separation of plasma from whole blood before

testing, and, thus, a typical turn-around time is in the range of

ournal of Cardiothoracic and Vascular Anesthesia, Vol 26, No 6 (Decemb

0-90 minutes.13,24 Abnormalities detected in these tests arefollowed by requests for specific blood components. The choiceof hemostatic therapies also affects the lag time before inter-vention. The time required for thawing FFP and cryoprecipitateis typically 30-60 minutes, but less time is needed for plateletconcentrates and thawed plasma. Factor concentrates, such asfibrinogen concentrate, and recombinant activated factor VII(rFVIIa) can be administered rapidly (�10 min) because theyare reconstituted in small volumes and infused irrespective ofblood type.

One of the key facts behind DCR is to prevent the delay oftransfusion therapy for patients in whom the risk of hemor-rhagic death is considerably greater than transfusion-associatedcomplications.25 However, a substantial number of patients

ay receive allogeneic plasma products inappropriately or inxcess, which collectively increase the risk for transfusion-elated adverse events.26 By implementing rapid POC coagu-ation testing, hemostatic interventions can be more individu-lized and goal directed (targeted) instead of indiscriminatepplications of DCR. Currently available POC coagulation testsre classified into 3 categories. The first category includes POChole-blood PT and aPTT for a rapid evaluation (5-10 min) oflasmatic coagulation. POC PT has been shown to have aeasonable agreement with plasma-based PT.13,27,28 However,

PT and aPTT are sensitive only to severe hypofibrinogenemia(�60-70 mg/dL) and are insensitive to FXIII deficiency or

From the *Department of Anesthesiology, University of PittsburghMedical Center, Pittsburgh, PA; †Department of Anesthesia and Inten-sive Care Medicine, University of Basel Hospital, Basel, Switzerland;‡Department of Anesthesiology, Emory University School of Medicine,Atlanta, GA; and §Royal Infirmary of Edinburgh, Edinburgh, UK.

Dr Bolliger was supported by a Myron B. Laver Grant, Departmentof Anaesthesia, University of Basel, Switzerland. Drs Tanaka, Bolliger,and Nimmo have received honoraria for consultation and/or lecturesfrom TEM International (Munich, Germany).

Address reprint requests to Kenichi A. Tanaka, MD, MSc, VisitingProfessor of Anesthesiology, Department of Anesthesiology, UPMCPresbyterian C-215, 200 Lothrop Street, Pittsburgh, PA 15213.E-mail: [email protected]

© 2012 Elsevier Inc. All rights reserved.1053-0770/2606-0022$36.00/0http://dx.doi.org/10.1053/j.jvca.2012.06.015Key words: rotational thromboelastometry, coagulation monitoring,

blood component transfusion, cardiac surgery, trauma

1083er), 2012: pp 1083-1093

Page 2: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

tet

abafPoN

na

p

m

c

s

(

1084 TANAKA ET AL

fibrinolysis.29 Further, PT and aPTT have different sensitivitieso hemodilution-induced decreases in procoagulant factor lev-ls,15,16 and neither test reflects the total amount of thrombinhat can be generated in plasma.24 The second category includes

whole-blood platelet function tests, which are used increasinglyto monitor therapeutic responses to aspirin, clopidogrel, andother platelet adenosine-5=-diphosphate antagonists.30,31 Evalu-ting the extent of platelet inhibition by antiplatelet agents maye useful in optimizing the perioperative risks of thrombosisnd hemorrhage.32,33 Although a detailed discussion of plateletunction monitoring is beyond the scope of this review,lateletMapping will be discussed later as a modified techniquef thrombelastography (TEG; Haemonetics-Haemoscope,iles, IL).34 The third category includes TEG and rotational

thromboelastometry (ROTEM; TEM Systems, Raleigh, NC).These 2 systems are suitable for timely decision making (�20min) in hemostatic interventions.2,4-6 Indeed, the decrease ofpostoperative blood loss without increasing the blood compo-

Table 1. Damage Control Resuscitation

Early surgical control of bleeding sitesEarly transfusion of plasma, platelets, and erythrocytes; minimized

crystalloid usagePermissive hypotension (mean arterial pressure �60 mmHg)Correction of hypothermia and acidosisTimely use of CaCl2, THAM, and rFVIIa

Abbreviations: rFVIIa, recombinant factor VIIa; THAM, tris-hydroxy-ethyl aminomethane (alkalizer).

Pin

Blood

Light detector

Diode

Dataprocessor

Clo

t fir

mne

ss (m

m)

20

60

100

10

Alpha

CFT

CT

Fig 1. Basic principles of rotational thromboelastometry. For a r

placed with CaCl2 and a coagulation activator in the stationary cup. T

hanges the torque between the pin and the cup. The measured rot

econds), clot formation time (CFT; seconds), �-angle (degrees), amp

MCF; millimeters), and maximum lysis (ML; percent decrease in amplitud

ent usage or mortality has been shown in a recent meta-nalysis, including TEG and ROTEM.35 Rapid detection of

coagulopathy by TEG or ROTEM allows a timely preparation(thawing) of blood products or a prompt intervention usingplasma-derived or recombinant factor concentrate.4-6,36,37

COAGULATION TESTING ON ROTEM

The basic principles and technical aspects of TEG andROTEM have been reviewed elsewhere.14,38-40 In this article,the practical applications of ROTEM are described because thissystem currently offers comprehensive tests of coagulation. Forstandard ROTEM measurements, a citrated whole-blood sam-ple (300 �L) is placed in a plastic cup using an automatedipette (Fig 1). The sample is recalcified with CaCl2, 0.2

mmol/L (StarTEM; 20 �L) and activated with 20 �L of anEXTEM (tissue factor [TF]) or INTEM (ellagic acid) reagent.Subsequently, the plastic pin is immersed in the blood. Oncethrombin is generated in the blood, platelets are activated toexpress glycoprotein (GP) IIb/IIIa receptors, and fibrin isformed and polymerized. The interactions of GP IIb/IIIa recep-tors and polymerized fibrin increase the torque (viscoelasticity)between the cup and the rotating pin (at a 4.75° angle). Thebreakdown of fibrin strands by fibrinolysis decreases thetorque. The change in torque is detected optically and is pro-cessed by the microprocessor to trace the clot formation andbreakdown.

The commonly used ROTEM variables include coagulationtime (CT; seconds), clot formation time (seconds), �-angle(degrees), amplitude at 10 minutes after CT (A10; millimeters),

20 30 40 50 60

MCF ML

CT Clotting timeCFT Clot formation timeAlpha Alpha angleA10 Amplitude 10 min after CTMCF Maximum clot firmnessML Maximum lysis

Time (min)

nal thromboelastometric measurement, a citrated blood sample is

tating pin is lowered into the blood, and subsequent clot formation

al thromboelastometric variables include the coagulation time (CT;

e at 10 minutes after CT (A10; millimeters), maximum clot firmness

A10

otatio

he ro

ation

litud

e 60 min after MCF). (Color version of figure is available online.)

Page 3: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

kav

a

b

atm

1085ROTEM IN COAGULATION MANAGEMENT

maximum clot firmness (MCF; millimeters), and maximumlysis (ML; percent decrease in amplitude 60 min after MCF;Fig 1). CT represents the onset of coagulation, whereas the clotformation time and �-angle represent the initial rate of fibrinpolymerization. MCF is a measurement of the maximal vis-coelastic strength of the clot. An ML �15% is used for thediagnosis of a premature breakdown of clot (hyperfibrinolysis).Normal ranges are summarized in Table 2.41 The referenceranges of TEG differ from those of ROTEM because of differ-ent sample types (citrated v noncitrated) and coagulation acti-vators (kaolin v INTEM or EXTEM).39

In addition to EXTEM and INTEM, several other tests canbe used in conjunction to diagnose specific coagulation prob-lems. FIBTEM is a modified EXTEM test (Fig 2A) withcytochalasin D, which inhibits platelet cytoskeletal reorganiza-tion and, thus, fibrin(ogen) binding to platelet GP IIb/IIIa.42 Bycombining EXTEM and FIBTEM, the differential diagnosis ofthrombocytopenia and/or hypofibrinogenemia is feasible within20 minutes (Fig 2B, C). APTEM is also a modified EXTEM, inwhich aprotinin inhibits plasmin in vitro if systemic fibrinolysiswas present (Fig 2D).37

HEPTEM contains heparinase in addition to the INTEMreagent. It is used as a pair with INTEM for the diagnosis ofsystemic heparin activity (Fig 2E).43 Although INTEM andaolin-activated TEG are intrinsic pathway tests, the sensitivitynd specificity are considerably different. Therefore, the cutoffalues for ROTEM cannot be applied simply to TEG.39

HEMOSTATIC MECHANISMS IN VIVO

In the event of a vascular injury (Fig 3A), a localizedhemostatic response is triggered by subendothelial collagen andTF, which are exposed to the circulating blood. Circulatingplatelets play a particularly important role in arterial hemosta-sis. The initial tethering of platelets to collagen is mediated byplatelet GP Ib/IX and the von Willebrand factor. A transientbinding of platelets to the von Willebrand factor becomesstabilized by collagen-induced platelet activation (via �2�1nd GP VI receptors).44 In parallel, a trace amount of thrombin

is generated by TF-FVIIa/FXa (extrinsic pathway). Thus, aden-osine-5=-diphosphate and thromboxane are released by colla-gen- and thrombin-activated platelets, forming the primaryhemostatic plug (Fig 3B). Subsequently, platelet aggregatesserve as catalytic surfaces and binding sites for coagulationresponses. Substrates (fibrinogen), proenzymatic factors (FII,

Table 2. Indications and Reference Rang

Tests Indicated to Evaluate

EXTEM, APTEM 2platelets, 2plasma factors, 1fibrinolysisFIBTEM 2fibrinogenINTEM, HEPTEM 2platelets, 2plasma factors, 1heparin

NOTE. Normal ranges (medians) are shown for EXTEM, FIBTEM, anreference range is used for HEPTEM. Data from Lang et al.41

Abbreviations: A10, amplitude at 10 minutes after coagulation timEXTEM, tissue factor reagent; FIBTEM, modified EXTEM test with cyreagent; INTEM, ellagic acid reagent; MCF, maximum clot firmness; Mprothrombin complex concentrate; TXA, tranexamic acid.

FIX, FX, FXI, FXIII), and accelerators (FV, FVIII) are con-

gregated (depicted as S-E-A in Fig 3C) on the activated plateletsurface to augment the local generation of thrombin and po-lymerized fibrin. After the initial activation of thrombin by theextrinsic pathway, the propagation of thrombin formationmainly involves the “intrinsic pathway.” Thrombin can activateFXI, which efficiently converts FIX to FIXa. On the activatedplatelet surface, FIXa in combination with thrombin-activatedFVIIIa becomes the major activator (intrinsic tenase) of FX.Subsequently, FXa and thrombin-activated FVa form a com-plex (prothrombinase), which exponentially increases the con-version of prothrombin (FII) to thrombin. Once activated byadenosine-5=-diphosphate-stimulated, each platelet expresses alarge number of GP IIb/IIIa receptors (�12,000) for fibrinogenbinding.45 Platelet-bound fibrinogen is converted to a fibrinmonomer by thrombin. Fibrin monomers are polymerized byplasma and platelet-derived FXIIIa, a transglutaminase, whichrequires thrombin-mediated activation.46 Polymerization of fi-rin on platelets stabilizes the primary hemostatic plug.47 On

ROTEM and TEG, major hemostatic responses involvingthrombin-activated platelets (GP IIb/IIIa), fibrin, and FXIIIaare reflected, although the contributions of the von Willebrandfactor, platelet GP Ib/IX, and other platelet agonists are mini-mal.

HEMOSTATIC INTERVENTIONS

It is well known that blood transfusion practice varies amongindividuals and institutions.48 In case of bleeding, FFP, plateletconcentrates, and cryoprecipitate often are transfused empiri-cally without laboratory testing. In addition to these allogeneicproducts, plasma-derived or recombinant factor concentrates,such as fibrinogen concentrates, prothrombin complex concen-trates, and rFVIIa, have been used for perioperative hemosta-sis.6,49 Recent clinical data have shown that ROTEM testing is

practical method to standardize the local transfusion prac-ice.5,50,51 Normal ranges of ROTEM testing are based on a

ulticenter study in healthy adult volunteers (Table 2).41

Hemostatic intervention(s) should be considered for nonsur-gical bleeding in the presence of abnormal ROTEM results.Threshold ROTEM values for interventions may vary amongdifferent types of vascular injury. In this section, several he-mostatic products are discussed in relation to ROTEM param-eters, but the availability of products differs among institutionsand countries. Therefore, it is prudent to validate or modify the

Rotational Thromboelastometric Tests

CT (s) CFT (s) A10 (mm) MCF (mm) ML (%)

2-74 (55) 46-148 (95) 43-65 (53) 49-71 (60) 0-18 (4)— — 9-24 (14) 9-25 (16) —

-246 (184) 40-100 (63) 44-68 (55) 52-72 (61) 0-12 (3)

EM. The EXTEM reference range is used for APTEM and the INTEM

PTEM, modified EXTEM test with aprotinin; CT, coagulation time;lasin D; FFP, fresh-frozen plasma; HEPTEM, heparinase plus INTEMximum lysis (percent decrease in amplitude 60 min after MCF); PCC,

es of

4

137

d INT

e; AtochaL, ma

herein-described algorithm for suitability at each institution.

Page 4: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

1086 TANAKA ET AL

EA.

CT

B.

C.

MCF 9 mm

MCF 13 mm

MCF 38 mm

MCF 58 mm

MCF 4 mm

FIBTEM

10 30 min 10 30 min

10 30 min

10 30 min10 30 min

D.

10 30 min

10 30 min

ML 100%

CT 1500 s

E.

CT 205 s

ML 0%

EXTEM

FIBTEM EXTEM MCF 40 mm

min30 10

MCF 38 mmFIBTEM EXTEM

APTEM

HEPTEM INTEM

EXTEM Fig 2. Examples of rotational throm-

boelastometric tracings. (A) Normal

(EXTEM, FIBTEM): EXTEM-MCF (normal,

49-71 mm) and FIBTEM (normal, 9-25

mm). (B) Thrombocytopenia (EXTEM,

FIBTEM): platelet count 74 � 109/L and

fibrinogen 170 mg/dL. (C) Thrombocyto-

penia and hypofibrinogenemia (EXTEM,

FIBTEM): platelet count 57 � 109/L and

fibrinogen 78 mg/dL. (D) Systemic fibri-

nolysis (EXTEM, APTEM): normal ML is

<15%. (E) Heparin effect (INTEM,

HEPTEM): prolonged CT (1,500 s) at

INTEM (normal range, 137-246 s) is cor-

rected at HEPTEM (ie, CTINTEM/CTHEPTEM �

1.0 in the absence of heparin). APTEM,

modified EXTEM test with aprotinin; CT,

coagulation time; EXTEM, tissue factor

reagent; FIBTEM, modified EXTEM test

with cytochalasin D; HEPTEM, heparinase

plus INTEM reagent; INTEM, ellagic acid

reagent; MCF, maximum clot firmness;

ML, maximum lysis (percent decrease in

10 30 min 10 30 minamplitude 60 min after MCF). (Color ver-

sion of figure is available online.)

Page 5: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

m

Dtmn

1087ROTEM IN COAGULATION MANAGEMENT

Platelets

The clot firmness of EXTEM shows the tensile strength ofthe whole-blood clot, which reflects the interaction betweenthrombin-activated platelets and polymerized fibrin via plateletGP IIb/IIIa receptors. For microvascular bleeding, EXTEM-and FIBTEM-A10 values can be used differentially to diagnosethe need for platelet transfusion or fibrinogen replacement.Thrombocytopenia (�50-100 � 109/L) is suspected when theEXTEM-MCF is �45 mm and the FIBTEM-MCF is �8-10mm (Table 3).5,52 When the FIBTEM-MCF is �10 mm, cryo-precipitate or fibrinogen is withheld unless bleeding is likelyto continue and lower fibrinogen levels. Platelet transfusionand fibrinogen replacement usually are indicated when theEXTEM-MCF is �35 mm.5,53 When the EXTEM-MCF is �45

m, severe thrombocytopenia (�50 � 109/L) is unlikely, buthereditary platelet dysfunction or antiplatelet effects of aspirinand P2Y12 antagonists (ticlopidine, clopidogrel, prasugrel, etc)may not be excluded. Platelet function tests can be used pre-operatively to screen high-risk patients. PFA-100 (Siemens,Tarrytown, NY) can be useful in screening patients with vonWillebrand disease or a platelet GP Ib defect (Bernard-Soulier

Vascular injuryBleeding

Event Primary hemostasisLocalization of factors

Thrombin generationFibrin polymerization

Elements CollagenTissue factor

Platelets ADP, ThromboxanevWF, FibrinogenFVII, FX, FII

Interventions FFP, PCCCryoprecipitateFibrinogenFXIII concentraterFVIIa

PlateletsCryoprecipitatevWF concentrateDesmopressin

(a) Vascular injury (b) Primary hemostasis (c) Coagulation

S-E-A S-E-A

Platelets (S) Fibrinogen(E) FII, FIX, FX, FXI, FXIII(A) FV, FVIII

Erythrocytes

Fig 3. Hemostatic processes and phase-specific interventions. (A)

Hemorrhage occurs after vascular injury. Extravascular (subendothe-

lial) collagen and tissue factor are exposed to the flowing blood.

Transfusion of erythrocytes is the initial intervention. The intact

vascular wall (blue), platelets (white ovals), erythrocytes (red circles),

and fibrin (green) are depicted. (B) Platelets adhere to the vascular

injury site by interacting with von Willebrand factor (vWF) by glyco-

protein Ib/IX receptors. Mural platelets are activated by collagen and

trace thrombin (by the extrinsic pathway involving factors VII [FVII],

X [FX], and II [FII]). They release adenosine-5=-diphosphate (ADP) and

thromboxane, stabilizing platelet-platelet interactions with fibrino-

gen. Thus, the primary (hemostatic) plug is established. Platelet

transfusion and measures to increase von Willebrand factor can

augment this process. (C) Activated platelet aggregates serve as a

catalytic surface and binding sites for coagulation responses. Sub-

strates (S; fibrinogen), proenzymatic factors (E; factors II, IX [FIX], X,

XI [FXI], and XIII [FXIII]), and accelerators (A; factors V [FV] and VIII

[FVIII]) are congregated. These factors can be replaced using fresh-

frozen plasma (FFP) or specific factor concentrates (see text for

details). PCC, prothrombin complex concentrate; rFVIIa, recombinant

factor VIIa.

syndrome).54 The VerifyNow system (Accumetrics, Inc., San s

iego, CA) and whole-blood impedance aggregometry (Mul-iplate, DynaBite, Munich, Germany) are used increasingly toonitor therapeutic responses to aspirin and P2Y12 antago-

ists.32,55 For TEG, PlateletMapping is available for the eval-uation of platelet inhibition by aspirin or clopidogrel.34,56-58 Forthis assay, a heparin-anticoagulated blood sample is used spe-cifically to inhibit thrombin, which masks the antiplatelet ef-fects of aspirin and clopidogrel. For PlateletMapping, fibrinpolymerization is achieved by the mixture of reptilase andactivated FXIII. Platelets are activated separately by the spe-cific platelet activator (arachidonic acid for aspirin and adeno-sine-5=-diphosphate for clopidogrel). Decreased maximum am-plitudes on PlateletMapping compared with kaolin-activatedTEG have been observed in perioperative patients with grossplatelet inhibition by aspirin or clopidogrel.59

Plasma and Prothrombin Complex Concentrate

The CT of EXTEM or INTEM can be used in determiningthe need for administering FFP or prothrombin complex con-centration (PCC). The latter refers to plasma-derived concen-trates of vitamin K-dependent factors (FVII, FIX, FX, pro-thrombin, protein C, and protein S).60 Coagulation factorreplacements are considered when CT values are prolonged(EXTEM-CT �100 s or INTEM-CT �240 s) and when resid-ual heparin effects, thrombocytopenia, and hypofibrinogenemiahave been addressed appropriately (Table 3). In patients whohave received intravenous heparin, a proper neutralization ofheparin can be confirmed by equal CT values between INTEMand HEPTEM (Fig 2E).43 Plasma or PCC can be used to correcta factor deficiency for a prolonged CT on HEPTEM.5

Similar to PT and aPTT, EXTEM-CT and INTEM-CT aredefined as the onset of blood coagulation after activation withTF and ellagic acid, respectively. However, ROTEM-CT val-ues are not equivalent to PT and aPTT. In trauma-inducedcoagulopathy, the correlation between the CT value and PT/aPTT was found to be rather poor (r � 0.47-0.53).61 Thereaction time (equivalent of CT) of kaolin-activated TEG hasbeen reported to correlate poorly with PT or aPTT.62,63 Abnor-mal PT/aPTT values (�1.5 times normal) are found frequentlywhen ROTEM parameters related to fibrin polymerization(EXTEM-A15 or clot formation time) are abnormal.61 Severehypofibrinogenemia (�60-70 mg/dL) can be the cause of pro-longed CT and PT/aPTT.

Fibrinogen-Rich Components

Cryoprecipitate is used commonly in North America forfibrinogen replacement. In many European countries, this prod-uct is no longer available, and plasma-derived fibrinogen con-centrate is used as a substitute.5,6,64 The minimal level offibrinogen previously was thought to be 80-100 mg/dL.65,66

However, higher fibrinogen levels (150-200 mg/dL) have beenrecommended in recent guidelines.67-69 It is plausible that aminimal fibrinogen level of 80-100 mg/dL is adequate forcongenital afibrinogenemia (ie, normal factor levels other thanfibrinogen), but higher fibrinogen levels (150-200 mg/dL) arenecessary for a multifactorial deficiency associated with peri-operative coagulopathy.70 There are several published data that

upport the efficacy of fibrinogen-rich components. In pediatric
Page 6: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

2rummtm

tF(Tlabobp

Fc

(dfi

gcft

1088 TANAKA ET AL

cardiac surgical patients (body weight �8 kg), Miller et al71

reported that 24-hour chest tube drainage was greater afterplatelet and FFP transfusions compared with platelet and cryo-precipitate (39.8 v 20.2 mL/kg). These patients received FFP orcryoprecipitate when the platelet transfusion was not effectiveto correct microvascular bleeding. The post-treatment fibrino-gen level was 158 � 27 mg/dL in the platelet-FFP group versus37 � 67 mg/dL in the platelet-cryoprecipitate group. Moreecently, Rahe-Meyer et al72 reported that perioperative bloodsage (erythrocytes, FFP, and platelets) was decreased byaintaining a higher plasma fibrinogen level (mean, 360 � 60g/dL) using a purified fibrinogen concentrate compared with

he conventional management (mean fibrinogen, 210 � 30g/dL) for replacement of the ascending aorta.The FIBTEM test has been used commonly in ROTEM for

he clinical assessment of fibrin polymerization in whole blood.IBTEM-MCF is well correlated with plasma fibrinogen levelsr � 0.85-0.87).24,61 In trauma-induced coagulopathy, a FIB-EM-A10 of �5 mm was reported to be a good predictor of

ow plasma fibrinogen (�100 mg/dL), with a sensitivity of 91%nd a specificity of 85%.61 In patients after cardiopulmonaryypass (CPB), a FIBTEM-MCF of 10 mm was a good estimatef plasma fibrinogen at 200 mg/dL.24,61 For microvascularleeding, fluid resuscitation and blood loss continuously lower

Table 3. Hemostatic Interventions Based

EXTEM-M

Clot Firmness �35 mm

FIBTEM-MCF*�10 mm Cryo/fibrinogen �

Platelet 1 UCryo/

Pla�10 mm Platelets 1-2 U Platel

Diagnosis

Prolonged CT ValuesINTEM-CT/HEPTEM-CT ratio �1.0 Residual heparin ProtaEXTEM-CT �100 s orINTEM-CT �240 s Low coagulation factors FFP 1FIBTEM-A10 �5 mm Very low fibrinogen

(�100 mg/dL)Cryo/

Fibrinolysis Patterns†Fibrinolysis �20 min Fulminant fibrinolysis TXA 1Fibrinolysis 20-40 min Early fibrinolysis TXA 1Fibrinolysis �40 min Clot retraction or late

fibrinolysisUsua

Abbreviations: A10, amplitude at 10 minutes after coagulation timcoagulation time; EACA, �-aminocaproic acid; EXTEM, tissue factor replasma; HEPTEM, heparinase plus INTEM reagent; INTEM, ellagicconcentrate; TXA, tranexamic acid.

*FIBTEM-A10 at 8 mm may be used as a cutoff instead of FIBTEM-Mfibrinogen concentrate, 2 g, is administered. If FIBTEM-A10 is �5 mm

†EXTEM and APTEM are repeated after each therapeutic intervent‡Antifibrinolytic agents are used only if the risk of bleeding is great

coagulation.

lasma fibrinogen. Therefore, it is reasonable to maintain a

IBTEM-MCF at 10 mm using a cryoprecipitate or fibrinogenoncentrate (Table 3).

FIBTEM testing can be performed in heparinized sampleseg, during CPB), and, therefore, hypofibrinogenemia can beetected early so that the cryoprecipitate can be thawed orbrinogen concentrates can be prepared.5,50,51

Recombinant Activated Factor VIIa

The use of rFVIIa is indicated in hemophiliac patients whohave developed neutralizing antibodies against FVIII or FIX.73

Its use also is common in severe perioperative bleeding aftercardiac surgery and major trauma.74 The efficacy of rFVIIa toenerate thrombin depends on the available TF.75 High TFoncentrations in PT and EXTEM make these tests insensitiveor delineating the in vivo efficacy of rFVIIa in surgical pa-ients76 and in hemophiliac patients.77 Alternatively, diluted TF

(eg, innovin, 1:17,000) has been tried, with mixed results, inmonitoring the therapeutic response to rFVIIa in hemophilia.78-80

Kaolin-activated TEG has been used to evaluate the TF-indepen-dent hemostatic activity of rFVIIa in hemophilia.81,82 In nonhe-mophiliac surgical patients, rFVIIa continues to be used as asecond-line intervention after the failure of platelet and plasmatransfusions.76,83 In this regard, TEG and ROTEM can be used to

otational Thromboelastometric Results

rameters

EXTEM-MCF

35-45 mm �45 mm

ogen �

1 UCryo/fibrinogen

If bleeding is uncontrolled, consider FFP orPCC based on EXTEM-CT as below orconsider platelet transfusion in patientson P2Y12 inhibitors

Management

25-50 mg

L/kg or PCC 20 IU/kgogen as above

or EACA 5-10 g‡EACA 5 gtreatment is required

EM, modified EXTEM test with aprotinin; Cryo, cryoprecipitate; CT,; FIBTEM, modified EXTEM test with cytochalasin D; FFP, fresh-frozeneagent; MCF, maximum clot firmness; PCC, prothrombin complex

t 10 mm. For fibrinogen replacement, Cryo, 10 U, or plasma-deriveddose of Cryo or fibrinogen concentrate is doubled.

n the risk of thrombosis or worsening of disseminated intravascular

on R

CF Pa

fibrinteletet 1 U

mine

0-15 mfibrin

-2 gg or

lly no

e; APTagentacid r

CF a, the

ion.er tha

diagnose major causes of bleeding, such as thrombocytopenia,

Page 7: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

s

tvccpf�ptn

oihC

ma

mmg8pfml

n

n

c

1089ROTEM IN COAGULATION MANAGEMENT

hypofibrinogenemia, and fibrinolysis, which preclude the opti-mal hemostatic effect of rFVIIa.76,84-86

Antifibrinolytic Therapy

Systemic fibrinolysis can be caused by an increased endo-thelial release of tissue plasminogen activator87 or by a de-creased protease inhibition of tissue plasminogen activator andplasmin.85 In ROTEM, hyperfibrinolysis is suspected when thedecrease of the amplitude over 1 hour is �15% of MCF (Table3, Fig 2D). The APTEM test is a modified EXTEM test withadded aprotinin (plasmin inhibitor) at ROTEM. The resolutionof fibrinolysis on APTEM compared with EXTEM confirmsongoing systemic fibrinolysis.14 In 15%-20% of patients withmajor trauma, overt hyperfibrinolysis is observed at ROTEMand TEG.36,37,88 Even in the absence of systemic fibrinolysis, afibrin clot tends to be more susceptible to a plasmin-mediatedbreakdown after hemodilution owing to a progressive loss ofendogenous fibrinolysis inhibitors.85 Using ROTEM, it may befeasible to use antifibrinolytic therapy selectively in patients atrisk for systemic fibrinolysis,89 which can be associated withevere injuries and increased mortality.37,88

ROTEM-BASED TRANSFUSION ALGORITHMS INCARDIAC SURGERY

Cardiovascular surgical patients are at increased risk forvascular thrombosis associated with atheromatous vascular dis-ease, atrial fibrillation, implanted coronary stents, and mechan-ical heart valves.90 Antiplatelet and antithrombotic therapiesoften are prescribed for preoperative patients. Balancing therisk of thrombosis against hemorrhage is one of the mostdifficult tasks for perioperative physicians.90,91 Intraoperatively,hese patients are anticoagulated with heparin for CPB orascular anastomosis. Hemorrhage and hemodilution decreaseirculating levels of coagulation factors and inhibitors.85 At theonclusion of surgery, heparin anticoagulation requires arompt reversal using protamine to establish hemostasis (clotormation). Antifibrinolytic therapy with tranexamic acid or-aminocaproic acid commonly is used during CPB as a pro-hylactic measure to decrease bleeding.92,93 However, theransfusion of allogeneic plasma and platelet products often isecessary to achieve hemostasis in complex CPB cases.83,93

Transfusion algorithms using TEG or ROTEM have beenshown previously to decrease postoperative blood loss andtransfusion requirements in cardiac surgery.2,22,23 An examplef ROTEM-based coagulation management in cardiac surgerys presented (Fig 4). EXTEM and FIBTEM are insensitive toeparin (�6 U/mL), and they can be tested toward the end ofPB (eg, rewarming).50,51 Based on EXTEM/FIBTEM results

(Table 3), hemostatic therapy after CPB can be planned inadvance, which may decrease the long interval from protamineadministration to hemostatic intervention(s). After the correc-tion of surgical bleeding and metabolic parameters (eg, pHstatus, body temperature), a ROTEM-based protocol allowspatient-specific hemostatic therapy targeted to replace deficientcoagulation element(s) rather than indiscriminately transfusingplatelets and plasma products.94

The initial approach to hemostasis generally involves the

restoration of plasma fibrinogen to the range of 150-200

g/dL (FIBTEM-MCF 8-10 mm; Table 3) with continuousntifibrinolytic therapy.5,6 Platelet transfusion is used in

patients with thrombocytopenia (EXTEM-MCF �45 mmand FIBTEM �8-10 mm) and those with platelet dysfunc-tion from antiplatelet therapy. Preoperative platelet functiontests may be helpful to diagnose and manage bleeding re-lated to platelet dysfunction.30,32 In patients who continuepreoperative vitamin K antagonist therapy and in those whounderwent extensive hemodilution or cell salvage, plasma orPCC may be necessary to restore procoagulant zymogens.EXTEM-CT values �100 seconds may indicate a procoagu-lant factor deficiency, particularly FVII, FIX, FX, and pro-thrombin (Table 3). INTEM-CT values �240 seconds alsomay be used to diagnose procoagulant factor deficiency, butexcess heparin or protamine can prolong INTEM-CT.43 As

uch as 20-30 mL/kg of plasma may be required to correctoderate-to-severe factor deficiency,95 whereas PCC can be

iven in smaller volumes (80 mL per 25-IU/kg dose for an0-kg person) to supplement key hemostatic factors.96,97 Inatients who required acute vitamin K antagonist reversalor cardiac surgery, PCC was shown to be hemostaticallyore effective by increasing plasma FX and prothrombin

evels compared with FFP.91,98

Additional clinical studies are necessary to establish thesafety and efficacy of plasma-derived and recombinant factorconcentrates in combination with conventional plasma andplatelet transfusions. The transfusion protocol based onROTEM and TEG should be useful in the evaluation of coagu-lopathy and in the patient-specific allocation of transfusion

Rewarming on CPB

Protamineadministration

EXTEM/FIBTEM

EXTEM/FIBTEM INTEM/HEPTEM

Order PLT, FFP or cryoprecipiate, if indicated (Table 3)

Hemostatic therapy oradditional protamine

Confirm coagulopathy and heparin neutralization

EXTEM/FIBTEMEvaluate therapeutic response, if indicated, INTEM/HEPTEM/APTEMor PLT function tests

Fig 4. Rotational thromboelastometry-based coagulation man-

agement in cardiac surgery. Early detection of coagulopathy and

preparations for hemostatic therapies are feasible using EXTEM and

FIBTEM in the late phase of cardiopulmonary bypass (CPB; at re-

warming). Clinical bleeding consistent with coagulopathy can be

confirmed after repeating EXTEM and FIBTEM and optimizing hepa-

rin neutralization (CTINTEM/CTHEPTEM � 1.0). If antifibrinolytic therapy is

ot used routinely, APTEM can be useful to exclude systemic fibri-

olysis as a cause of bleeding. Platelet (PLT) function tests should be

onsidered in patients who recently used aspirin and P2Y12 antago-

nists. APTEM, modified EXTEM test with aprotinin; CT, coagulation

time; EXTEM, tissue factor reagent; FFP, fresh-frozen plasma; FIB-

TEM, modified EXTEM test with cytochalasin D; HEPTEM, heparinase

plus INTEM reagent; INTEM, ellagic acid reagent.

products in cardiac surgical patients.2,22,23,48,50,51

Page 8: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

rslaovbdhtaRaa

t(i

ppbcprpcswntlac

h(rfi

ag

b

1090 TANAKA ET AL

ROTEM-BASED TRANSFUSION ALGORITHMS INMAJOR TRAUMA

Most patients with major traumatic injuries are admitted withvarious degrees of cardiorespiratory and metabolic distur-bances. With an ongoing need for �4 U of erythrocyte con-centrates or a blood loss of �150 mL/min, DCR should betriggered to coordinate timely and sufficient provision of bloodproducts (Fig 5). Although damage-control surgery is pivotal tothe survival of patients with multiple injuries,99 massive fluidesuscitation often is required to counter systemic hypoperfu-ion and worsening acidosis.20,25 The rapid infusion of crystal-oid, albumin, or hydroxyethyl starch can lead to hypothermiand extensive hemodilution of erythrocytes, fibrinogen, andther coagulation factors and inhibitors.100 It is crucial to pre-ent the lethal triad of coagulopathy, hypothermia, and acidosisy early resuscitative and hemostatic therapies. In view ofynamic changes in the coagulation system and the paucity ofematologic information (eg, chronic antithrombotic therapy),he use of ROTEM is most practical for the comprehensivessessment of hemostatic function in trauma patients. InitialOTEM testing using EXTEM, FIBTEM, and APTEM allowsrapid evaluation of thrombocytopenia, hypofibrinogenemia,

nd the profibrinolytic state (Fig 2).The initial approach based on ROTEM involves the restora-

ion of plasma fibrinogen to the range of 150-200 mg/dLFIBTEM-MCF 8-10 mm) using plasma (when a large volume

Initiation of DCR

EXTEM/FIBTEM APTEM

Order PLT, FFP or cryoprecipiate, if indicated (Table 3)

Surgical intervention, if necessary

Evaluate therapeutic response, if indicated, add antifibrinolytic therapy

Continue monitoring until bleeding stopsif indicated, add INTEM or PLT function test

DCR with ROTEM-modifiedhemostatic therapy

EXTEM/FIBTEM APTEM

EXTEM/FIBTEM APTEM

Fig 5. Rotational thromboelastometry-based coagulation man-

agement in major trauma. At the initiation of a massive transfusion

protocol in major trauma patients, the use of EXTEM/FIBTEM/

APTEM allows a rapid evaluation of thrombocytopenia, hypofibrino-

genemia, and the profibrinolytic state. Specific component replace-

ment and antifibrinolytic therapy can be included in the damage

control resuscitation (DCR). Rotational thromboelastometric

(ROTEM) assessment can be repeated during and after surgical in-

tervention, if indicated. For persistent microvascular bleeding, plate-

let (PLT) function testing should be considered to evaluate the defect

in the primary hemostasis (eg, aspirin and P2Y12 antagonists). AP-

TEM, modified EXTEM test with aprotinin; EXTEM, tissue factor

reagent; FFP, fresh-frozen plasma; FIBTEM, modified EXTEM test

with cytochalasin D; INTEM, ellagic acid reagent.

s permitted), cryoprecipitate, or fibrinogen concentrates (Table

REN

oelastography-guided transfusion algorithm reduces trans-

f1

a2

3).4 Platelet transfusion is used in patients with thrombocyto-enia (EXTEM-MCF �45 mm) and those with suspectedlatelet dysfunction. Once residual heparin effects and hypofi-rinogenemia are excluded, EXTEM-CT values �100 secondsan be addressed with coagulation factor replacements usinglasma or PCC (Table 3). EXTEM-CT seems to be moreesponsive to hemodilution-induced factor deficiency com-ared with INTEM-CT or kaolin TEG R-time because in-reased FVIII in stress shortens contact-activated tests.16 Inevere injury, low EXTEM-MCF values (�35 mm consistentith thrombocytopenia and hypofibrinogenemia) are accompa-ied by a profibrinolytic state (ML �15%).36,37,88 The resolu-ion of clot breakdown on APTEM confirms systemic fibrino-ysis (Fig 2), and intravenous administration of tranexamiccid, 1-2 g, should be considered unless ongoing intravascularoagulation is suspected clinically (Table 3).89 A coexisting

hypocoagulable and profibrinolytic state in major trauma seemsto indicate the severity of illness, which demonstrates a corre-lation with mortality.37,57,88,101

Hemostatic interventions used in DCR are different amongtrauma centers based on the institutional blood usage policyand the availability of specific components and concen-trates.67,69,102,103 It is acceptable to initiate DCR early usingallogeneic plasma and platelet products if the risk of hemor-rhagic death is considered high.25 Once surgical controls ofemorrhage are attained, more individualized, goal-directedtargeted) transfusion is preferred because of the cumulativeisks of transfusion, including acute lung injury, multiple organailure, immunomodulation, thromboembolic complications,nfection, and death.10,12,26,104,105

CONCLUSIONS

Timely hemostatic interventions are pivotal in controlling co-agulopathy and bleeding after major surgery and trauma. How-ever, the risks of hemorrhage and transfusion-related complica-tions are to be weighed constantly against each other.12,25,26,104

ROTEM has become increasingly popular in perioperative coag-ulation management that involves the replacement of multiplecoagulation factors. Unlike hereditary hemorrhagic disorders,which usually involve a single factor replacement, perioperativehemorrhage in major surgery and trauma often demands sequen-tial treatments using multiple allogeneic components or factorconcentrates.4-6,83,100 Using the goal-oriented transfusion algo-rithm, clinicians appropriately may select necessary transfusioncomponent(s) instead of empirically administering all compo-nents, with potential hazardous effects.94 Recent clinical data havesupported the use of ROTEM or TEG in evaluating the clinicalefficacies of various hemostatic therapies, which had been seldomstudied.64,71,72 Further clinical investigations of hemostatic thera-pies under the guidance of ROTEM and TEG are warranted toestablish the safety, efficacy, and economic impact of various

hemostatic components.

REFE

1. Despotis GJ, Santoro SA, Spitznagel E, et al: Prospective evaluationnd clinical utility of on-site monitoring of coagulation in patients under-oing cardiac operation. J Thorac Cardiovasc Surg 107:271-279, 1994

2. Shore-Lesserson L, Manspeizer HE, DePerio M, et al: Throm-

CES

usions in complex cardiac surgery. Anesth Analg 88:312-319,9993. Mayer SA, Rincon F: Ultra-early hemostatic therapy for

cute intracerebral hemorrhage. Semin Hematol 43:S70-S76,

006
Page 9: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

1091ROTEM IN COAGULATION MANAGEMENT

4. Schöchl H, Nienaber U, Hofer G, et al: Goal-directed coagulationmanagement of major trauma patients using thromboelastometry(ROTEM)-guided administration of fibrinogen concentrate and pro-thrombin complex concentrate. Crit Care 14:R55, 2010

5. Girdauskas E, Kempfert J, Kuntze T, et al: Thromboelastometri-cally guided transfusion protocol during aortic surgery with circulatoryarrest: A prospective, randomized trial. J Thorac Cardiovasc Surg140:1117-1124, 2010

6. Görlinger K, Dirkmann D, Hanke AA, et al: First-line therapywith coagulation factor concentrates combined with point-of-care co-agulation testing is associated with decreased allogeneic blood trans-fusion in cardiovascular surgery: A retrospective, single-center cohortstudy. Anesthesiology 115:1179-1191, 2011

7. Burnouf T, Radosevich M: Nanofiltration of plasma-derived bio-pharmaceutical products. Haemophilia 9:24-37, 2003

8. Gröner A: Reply. Pereira A. Cryoprecipitate versus commercialfibrinogen concentrate in patients who occasionally require a therapeu-tic supply of fibrinogen: Risk comparison in the case of an emergingtransfusion-transmitted infection. Haematologica 2007;92:846-9.Haematologica 93:e24-e26, 2008

9. Engoren MC, Habib RH, Zacharias A, et al: Effect of bloodtransfusion on long-term survival after cardiac operation. Ann ThoracSurg 74:1180-1186, 2002

10. Khan H, Belsher J, Yilmaz M, et al: Fresh-frozen plasma andplatelet transfusions are associated with development of acute lunginjury in critically ill medical patients. Chest 131:1308-1314, 2007

11. Koch CG, Li L, Sessler DI, et al: Duration of red-cell storageand complications after cardiac surgery. N Engl J Med 358:1229-1239,2008

12. Watson GA, Sperry JL, Rosengart MR, et al: Fresh frozenplasma is independently associated with a higher risk of multiple organfailure and acute respiratory distress syndrome. J Trauma 67:221-227,2009

13. Toulon P, Ozier Y, Ankri A, et al: Point-of-care versus centrallaboratory coagulation testing during haemorrhagic surgery. A multi-center study. Thromb Haemost 101:394-401, 2009

14. Ganter MT, Hofer CK: Coagulation monitoring: Current tech-niques and clinical use of viscoelastic point-of-care coagulation de-vices. Anesth Analg 106:1366-1375, 2008

15. Segal JB, Dzik WH; Transfusion Medicine/Hemostasis ClinicalTrials Network: Paucity of studies to support that abnormal coagulationtest results predict bleeding in the setting of invasive procedures: Anevidence-based review. Transfusion 45:1413-1425, 2005

16. Yuan S, Ferrell C, Chandler WL: Comparing the prothrombintime INR versus the APTT to evaluate the coagulopathy of acutetrauma. Thromb Res 120:29-37, 2007

17. Borgman MA, Spinella PC, Perkins JG, et al: The ratio of bloodproducts transfused affects mortality in patients receiving massivetransfusions at a combat support hospital. J Trauma 63:805-813, 2007

18. Gonzalez EA, Moore FA, Holcomb JB, et al: Fresh frozenplasma should be given earlier to patients requiring massive transfu-sion. J Trauma 62:112-119, 2007

19. Holcomb JB, Wade CE, Michalek JE, et al: Increased plasmaand platelet to red blood cell ratios improves outcome in 466 massivelytransfused civilian trauma patients. Ann Surg 248:447-458, 2008

20. Brohi K, Cohen MJ, Ganter MT, et al: Acute coagulopathy oftrauma: Hypoperfusion induces systemic anticoagulation and hyperfi-brinolysis. J Trauma 64:1211-1217, 2008

21. Despotis GJ, Levine V, Saleem R, et al: Use of point-of-care testin identification of patients who can benefit from desmopressin duringcardiac surgery: A randomised controlled trial. Lancet 354:106-110,1999

22. Nuttall GA, Oliver WC, Santrach PJ, et al: Efficacy of a simple

intraoperative transfusion algorithm for nonerythrocyte component uti-

lization after cardiopulmonary bypass. Anesthesiology 94:773-781,2001

23. Avidan MS, Alcock EL, Da Fonseca J, et al: Comparison ofstructured use of routine laboratory tests or near-patient assessmentwith clinical judgement in the management of bleeding after cardiacsurgery. Br J Anaesth 92:178-186, 2004

24. Ogawa S, Szlam F, Chen EP, et al: A comparative evaluation ofrotation thromboelastometry and standard coagulation tests in hemodi-lution-induced coagulation changes after cardiac surgery. Transfusion52:14-22, 2012

25. Holcomb JB: Optimal use of blood products in severely injuredtrauma patients. Hematology Am Soc Hematol Educ Program 1:465-469, 2010

26. Hannon T: Trauma blood management: Avoiding the collateraldamage of trauma resuscitation protocols. Hematology Am Soc Hema-tol Educ Program 1:463-464, 2010

27. Ho AM, Lee A, Ling E, et al: Agreements between the pro-thrombin times of blood treated in vitro with heparinase during car-diopulmonary bypass (CPB) and blood sampled after CPB and sys-temic protamine. Anesth Analg 96:15-20, 2003

28. Urwyler N, Staub LP, Beran D, et al: Is perioperative point-of-care prothrombin time testing accurate compared to the standard la-boratory test? Thromb Haemost 102:779-786, 2009

29. Tanaka KA, Key NS, Levy JH: Blood coagulation: Hemostasisand thrombin regulation. Anesth Analg 108:1433-1446, 2009

30. Breet NJ, van Werkum JW, Bouman HJ, et al: Comparison ofplatelet function tests in predicting clinical outcome in patients under-going coronary stent implantation. JAMA 303:754-762, 2010

31. Breet NJ, van Werkum JW, Bouman HJ, et al: High on-aspirinplatelet reactivity as measured with aggregation-based, cyclooxygen-ase-1 inhibition sensitive platelet function tests is associated with theoccurrence of atherothrombotic events. J Thromb Haemost 8:2140-2148, 2010

32. Ferreiro JL, Sibbing D, Angiolillo DJ: Platelet function testingand risk of bleeding complications. Thromb Haemost 103:1128-1135,2010

33. Tanaka KA, Dietrich W: Is it time to implement preoperativeplatelet function testing before invasive procedures? Br J Anaesth107:842-843, 2011

34. Craft RM, Chavez JJ, Bresee SJ, et al: A novel modification ofthe Thromboelastograph assay, isolating platelet function, correlateswith optical platelet aggregation. J Lab Clin Med 143:301-309, 2004

35. Afshari A, Wikkelsø A, Brok J, et al: Thrombelastography(TEG) or thromboelastometry (ROTEM) to monitor haemotherapyversus usual care in patients with massive transfusion. Cochrane Da-tabase Syst Rev 3:CD007871, 2011

36. Levrat A, Gros A, Rugeri L, et al: Evaluation of rotation throm-belastography for the diagnosis of hyperfibrinolysis in trauma patients.Br J Anaesth 100:792-797, 2008

37. Schöchl H, Frietsch T, Pavelka M, et al: Hyperfibrinolysis aftermajor trauma: Differential diagnosis of lysis patterns and prognosticvalue of thrombelastometry. J Trauma 67:125-131, 2009

38. Luddington RJ: Thrombelastography/thromboelastometry. ClinLab Haematol 27:81-90, 2005

39. Venema LF, Post WJ, Hendriks HG, et al: An assessment ofclinical interchangeability of TEG and RoTEM thromboelastographicvariables in cardiac surgical patients. Anesth Analg 111:339-344, 2010

40. Bolliger D, Seeberger MD, Tanaka KA: Principles and practiceof thromboelastography in clinical coagulation management and trans-fusion practice. Transfus Med Rev 26:1-13, 2012

41. Lang T, Bauters A, Braun SL, et al: Multi-centre investigationon reference ranges for ROTEM thromboelastometry. Blood Coagul

Fibrinolysis 16:301-310, 2005
Page 10: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

1092 TANAKA ET AL

42. Lang T, Toller W, Gütl M, et al: Different effects of abciximaband cytochalasin D on clot strength in thrombelastography. J ThrombHaemost 2:147-153, 2004

43. Mittermayr M, Velik-Salchner C, Stalzer B, et al: Detection ofprotamine and heparin after termination of cardiopulmonary bypass bythrombelastometry (ROTEM): Results of a pilot study. Anesth Analg108:743-750, 2009

44. Ruggeri ZM: Von Willebrand factor. J Clin Invest 100:S41-S46,1997

45. Peerschke EI, Zucker MB, Grant RA, et al: Correlation betweenfibrinogen binding to human platelets and platelet aggregability. Blood55:841-847, 1980

46. Hornyak TJ, Shafer JA: Interactions of factor XIII with fibrin assubstrate and cofactor. Biochemistry 31:423-429, 1992

47. Ni H, Denis CV, Subbarao S, et al: Persistence of plateletthrombus formation in arterioles of mice lacking both von Willebrandfactor and fibrinogen. J Clin Invest 106:385-392, 2000

48. Stover EP, Siegel LC, Parks R, et al: Variability in transfusionpractice for coronary artery bypass surgery persists despite nationalconsensus guidelines: A 24-institution study. Institutions of the Multi-center Study of Perioperative Ischemia Research Group. Anesthesiol-ogy 88:327-333, 1998

49. Karkouti K, Beattie WS, Wijeysundera DN, et al: Recombinantfactor VIIa for intractable blood loss after cardiac surgery: A propen-sity score-matched case-control analysis. Transfusion 45:26-34, 2005

50. Spalding GJ, Hartrumpf M, Sierig T, et al: Cost reduction ofperioperative coagulation management in cardiac surgery: Value of“bedside” thrombelastography (ROTEM). Eur J Cardiothorac Surg31:1052-1057, 2007

51. Lee SH, Lee SM, Kim CS, et al: Use of fibrin-based throm-boelastometry for cryoprecipitate transfusion in cardiac surgery involv-ing deep hypothermic circulatory arrest during cardiopulmonary by-pass. Blood Coagul Fibrinolysis 21:687-691, 2010

52. Larsen OH, Fenger-Eriksen C, Christiansen K, et al: Diagnosticperformance and therapeutic consequence of thromboelastometry acti-vated by kaolin versus a panel of specific reagents. Anesthesiology115:294-302, 2011

53. Lang T, Johanning K, Metzler H, et al: The effects of fibrinogenlevels on thromboelastometric variables in the presence of thrombocy-topenia. Anesth Analg 108:751-758, 2009

54. Harrison P, Robinson MS, Mackie IJ, et al: Performance of theplatelet function analyser PFA-100 in testing abnormalities of primaryhaemostasis. Blood Coagul Fibrinolysis 10:25-31, 1999

55. Paniccia R, Antonucci E, Maggini N, et al: Assessment ofplatelet function on whole blood by multiple electrode aggregometry inhigh-risk patients with coronary artery disease receiving antiplatelettherapy. Am J Clin Pathol 131:834-842, 2009

56. Tantry US, Bliden KP, Gurbel PA: Overestimation of plateletaspirin resistance detection by thrombelastograph PlateletMapping andvalidation by conventional aggregometry using arachidonic acid stim-ulation. J Am Coll Cardiol 46:1705-1709, 2005

57. Carroll RC, Craft RM, Langdon RJ, et al: Early evaluation ofacute traumatic coagulopathy by thrombelastography. Transl Res 154:34-39, 2009

58. Carroll RC, Worthington RE, Craft RM, et al: Post interven-tional cardiology urinary thromboxane correlates with PlateletMappingdetected aspirin resistance. Thromb Res 125:e118-e122, 2010

59. Collyer TC, Gray DJ, Sandhu R, et al: Assessment of plateletinhibition secondary to clopidogrel and aspirin therapy in preoperativeacute surgical patients measured by thrombelastography PlateletMapping.Br J Anaesth 102:492-498, 2009

60. Levy JH, Tanaka KA, Dietrich W: Perioperative hemostaticmanagement of patients treated with vitamin K antagonists. Anesthe-

siology 109:918-926, 2008

61. Rugeri L, Levrat A, David JS, et al: Diagnosis of early coagu-lation abnormalities in trauma patients by rotation thrombelastography.J Thromb Haemost 5:289-295, 2007

62. Kashuk JL, Moore EE, Le T, et al: Noncitrated whole blood isoptimal for evaluation of postinjury coagulopathy with point-of-carerapid thrombelastography. J Surg Res 156:133-138, 2009

63. Alexander DC, Butt WW, Best JD, et al: Correlation of throm-boelastography with standard tests of anticoagulation in paediatricpatients receiving extracorporeal life support. Thromb Res 125:387-392, 2010

64. Fenger-Eriksen C, Jensen TM, Kristensen BS, et al: Fibrinogensubstitution improves whole blood clot firmness after dilution withhydroxyethyl starch in bleeding patients undergoing radical cystec-tomy: A randomized, placebo-controlled clinical trial. J Thromb Hae-most 7:795-802, 2009

65. O’Shaughnessy DF, Atterbury C, Bolton Maggs P, et al: Guide-lines for the use of fresh-frozen plasma, cryoprecipitate and cryosuper-natant. Br J Haematol 126:11-28, 2004

66. American Society of Anesthesiologists Task Force on Perioper-ative Blood Transfusion and Adjuvant Therapies. Practice guidelinesfor perioperative blood transfusion and adjuvant therapies: An updatedreport by the American Society of Anesthesiologists Task Force onPerioperative Blood Transfusion and Adjuvant Therapies. Anesthesi-ology 105:198-208, 2006

67. German Medical Association: Cross-sectional guidelines fortherapy with blood components and plasma derivatives. 4th Revisededition. Transfus Med Hemother 36:419-436, 2009

68. Fries D, Innerhofer P, Perger P, et al: Coagulation managementin trauma-related massive bleeding—Recommendations of the TaskForce for Coagulation (AGPG) of the Austrian Society of Anesthesi-ology, Resuscitation and Intensive Care Medicine (OGARI). Anaes-thesiol Intensivmed Notfallmed Schmerzther 45:552-561, 2010

69. Rossaint R, Bouillon B, Cerny V, et al: Management of bleedingfollowing major trauma: An updated European guideline. Crit Care14:R52, 2010

70. Levy JH, Szlam F, Tanaka KA, et al: Fibrinogen and hemosta-sis: A primary hemostatic target for the management of acquiredbleeding. Anesth Analg 114:261-274, 2012

71. Miller BE, Mochizuki T, Levy JH, et al: Predicting and treatingcoagulopathies after cardiopulmonary bypass in children. Anesth Analg85:1196-1202, 1997

72. Rahe-Meyer N, Pichlmaier M, Haverich A, et al: Bleedingmanagement with fibrinogen concentrate targeting a high-normalplasma fibrinogen level: A pilot study. Br J Anaesth 102:785-792, 2009

73. Abshire T, Kenet G: Recombinant factor VIIa: Review of effi-cacy, dosing regimens and safety in patients with congenital andacquired factor VIII or IX inhibitors. J Thromb Haemost 2:899-909,2004

74. Levi M, Levy JH, Andersen HF, et al: Safety of recombinantactivated factor VII in randomized clinical trials. N Engl J Med363:1791-1800, 2010

75. Butenas S, Brummel KE, Branda RF, et al: Mechanism of factorVIIa-dependent coagulation in hemophilia blood. Blood 99:923-930,2002

76. Wasowicz M, Meineri M, McCluskey SM, et al: The utility ofthromboelastography for guiding recombinant activated factor VII ther-apy for refractory hemorrhage after cardiac surgery. J CardiothoracVasc Anesth 23:828-834, 2009

77. Lak M, Scharling B, Blemings A, et al: Evaluation of rFVIIa(NovoSeven) in Glanzmann patients with thromboelastogram. Haemo-philia 14:103-110, 2008

78. Sørensen B, Johansen P, Christiansen K, et al: Whole bloodcoagulation thrombelastographic profiles employing minimal tissue

factor activation. J Thromb Haemost 1:551-558, 2003
Page 11: Rotational Thromboelastometry (ROTEM)-Based Coagulation ... - ROTEM Article.pdf · blood component transfusion, cardiac surgery, trauma Journal of Cardiothoracic and Vascular Anesthesia,

1093ROTEM IN COAGULATION MANAGEMENT

79. Young G, Ebbesen LS, Viuff D, et al: Evaluation of throm-boelastography for monitoring recombinant activated factor VII exvivo in haemophilia A and B patients with inhibitors: A multicentretrial. Blood Coagul Fibrinolysis 19:276-282, 2008

80. Viuff D, Andersen S, Sørensen BB, et al: Optimizing thrombe-lastography (TEG) assay conditions to monitor rFVIIa (NovoSeven)therapy in haemophilia A patients. Thromb Res 126:144-149, 2010

81. Schneiderman J, Rubin E, Nugent DJ, et al: Sequential therapywith activated prothrombin complex concentrates and recombinantFVIIa in patients with severe haemophilia and inhibitors: Update of ourprevious experience. Haemophilia 13:244-248, 2007

82. Tanaka KA, Szlam F, Rusconi CP, et al: In-vitro evaluation ofanti-factor IXa aptamer on thrombin generation, clotting time, andviscoelastometry. Thromb Haemost 101:827-833, 2009

83. Sniecinski RM, Chen EP, Makadia SS, et al: Changing fromaprotinin to tranexamic acid results in increased use of blood productsand recombinant factor VIIa for aortic surgery requiring hypothermicarrest. J Cardiothorac Vasc Anesth 24:959-963, 2010

84. Lewis NR, Brunker P, Lemire SJ, et al: Failure of recombinantfactor VIIa to correct the coagulopathy in a case of severe postpartumhemorrhage. Transfusion 49:689-695, 2009

85. Bolliger D, Szlam F, Levy JH, et al: Haemodilution-inducedprofibrinolytic state is mitigated by fresh-frozen plasma: Implicationsfor early haemostatic intervention in massive haemorrhage. Br J An-aesth 104:318-325, 2010

86. Bolliger D, Szlam F, Molinaro RJ, et al: Thrombin generationand fibrinolysis in anti-factor IX treated blood and plasma spiked withfactor VIII inhibitor bypassing activity or recombinant factor VIIa.Haemophilia 16:510-517, 2010

87. Emeis JJ: Regulation of the acute release of tissue-type plas-minogen activator from the endothelium by coagulation activationproducts. Ann N Y Acad Sci 667:249-258, 1992

88. Kashuk JL, Moore EE, Sawyer M, et al: Primary fibrinolysis isintegral in the pathogenesis of the acute coagulopathy of trauma. AnnSurg 252:434-442, 2010

89. Shakur H, Roberts I, Bautista R, et al: Effects of tranexamic acidon death, vascular occlusive events, and blood transfusion in traumapatients with significant haemorrhage (CRASH-2): A randomised, pla-cebo-controlled trial. Lancet 376:23-32, 2010

90. Douketis JD, Spyropoulos AC, Spencer FA, et al: Perioperativemanagement of antithrombotic therapy: Antithrombotic therapy andprevention of thrombosis, 9th ed: American College of Chest Physi-cians evidence-based clinical practice guidelines. Chest 141:e326S-e350S, 2012

91. Demeyere R, Gillardin S, Arnout J, et al: Comparison of fresh

frozen plasma and prothrombin complex concentrate for the reversal of

oral anticoagulants in patients undergoing cardiopulmonary bypasssurgery: A randomized study. Vox Sang 99:251-260, 2010

92. Kikura M, Levy JH, Tanaka KA, et al: A double-blind, placebo-controlled trial of epsilon-aminocaproic acid for reducing blood loss incoronary artery bypass grafting surgery. J Am Coll Surg 202:216-222,2006

93. Fergusson DA, Hébert PC, Mazer CD, et al: A comparison ofaprotinin and lysine analogues in high-risk cardiac surgery. N EnglJ Med 358:2319-2331, 2008

94. DeLoughery TG: Logistics of massive transfusions. HematologyAm Soc Hematol Educ Program 1:470-473, 2010

95. Chowdary P, Chowdhury P, Saayman AG: Efficacy of standarddose and 30 ml/kg fresh frozen plasma in correcting laboratory param-eters of haemostasis in critically ill patients. Br J Haematol 125:69-73,2004

96. Riess HB, Meier-Hellmann A, Motsch J, et al: Prothrombincomplex concentrate (Octaplex) in patients requiring immediate rever-sal of oral anticoagulation. Thromb Res 121:9-16, 2007

97. Pabinger I, Brenner B, Kalina U, et al: Prothrombin complexconcentrate (Beriplex P/N) for emergency anticoagulation reversal: Aprospective multinational clinical trial. J Thromb Haemost 6:622-631,2008

98. Ogawa S, Szlam F, Ohnishi T, et al: A comparative study ofprothrombin complex concentrates and fresh-frozen plasma for warfa-rin reversal under static and flow conditions. Thromb Haemost 106:1215-1223, 2011

99. Johnson JW, Gracias VH, Schwab CW, et al: Evolution indamage control for exsanguinating penetrating abdominal injury.J Trauma 51:261-269, 2001

100. Bolliger D, Görlinger K, Tanaka KA: Pathophysiology andtreatment of coagulopathy in massive hemorrhage and hemodilution.Anesthesiology 113:1205-1219, 2010

101. Kaufmann CR, Dwyer KM, Crews JD, et al: Usefulness ofthrombelastography in assessment of trauma patient coagulation.J Trauma 42:716-720, 1997

102. Roback JD, Caldwell S, Carson J, et al: Evidence-based prac-tice guidelines for plasma transfusion. Transfusion 50:1227-1239, 2010

103. Ferraris VA, Brown JR, Despotis GJ, et al: Update to theSociety of Thoracic Surgeons and the Society of Cardiovascular An-esthesiologists blood conservation clinical practice guidelines. AnnThorac Surg 91:944-982, 2011

104. Stanworth SJ, Brunskill SJ, Hyde CJ, et al: Appraisal of theevidence for the clinical use of FFP and plasma fractions. Best PractRes Clin Haematol 19:67-82, 2006

105. Abdel-Wahab OI, Healy B, Dzik WH: Effect of fresh-frozenplasma transfusion on prothrombin time and bleeding in patients with

mild coagulation abnormalities. Transfusion 46:1279-1285, 2006