Perioperative Hemostasis
Perioperative Hemostasis
Perioperative
Hemostasis
Coagulation for
Anesthesiologists
123
Perioperative Hemostasis
Carlo Enrique Marcucci • Patrick Schoettker
Editors
Perioperative Hemostasis
Coagulation for Anesthesiologists
Editors
Carlo Enrique Marcucci Patrick Schoettker
Department of Anesthesiology Department of Anesthesiology
University Hospital of Lausanne University Hospital of Lausanne
Lausanne Lausanne
Switzerland Switzerland
vii
SIZE OF TREATMENT EFFECT
Procedure/Treatment Additional studies with Additional studies with broad Procedure/Treatment should
SHOULD be performed/ focused objectives needed objectives needed; additional NOT be performed/adminis-
administered registry data would be helpful tered SINCE IT IS NOT HELP-
IT IS REASONABLE to per- FUL AND MAY BE HARMFUL
form procedure/administer Procedure/Tretment
treatment MAY BE CONSIDERED
Reproduced, with permission, from: Jacobs AK, Kushner FG, Ettinger SM et al. (2013) ACCF/AHA Clinical practice guideline methodology summit report: a
report of the american college of cardiology foundation/american heart association task force on practice guidelines. Circulation 127:268–310
Abbreviations
xi
xii Abbreviations
FI factor I
FNHTR febrile nonhemolytic transfusion reactions
FVIII factor VIII
FX factor X
FXa activated factor X
FXIII factor XIII
GP glycoprotein
GPIb glycoprotein Ib
HA human albumin
HAES/HES hydroxyethyl starch
HAV hepatitis A virus
HCR hemostatic control resuscitation
HEA Hypotensive epidural anesthesia
HELLP syndrome hemolysis, elevated liver enzymes, and
low platelets syndrome
HEV hepatitis E virus
HF hyperfibrinolysis
HHS hypertonic–hyperoncotic solutions
HIT heparin-induced thrombocytopenia
HIV human immunodeficiency virus
HLA human leukocyte antigen
HMWK high-molecular-weight kininogen
HNA human neutrophil antigen
HR hemostatic resuscitation
HUS hemolytic uremic syndrome
IBD inflammatory bowel disease
i-CFC isolated coagulation factor concentrate
ICH intracerebral hemorrhage
ICU intensive care unit
IL-6 interleukin 6
INR international normalized ratio
IPC intermittent pneumatic compression
ISS injury severity score
ISTH International Society on Thrombosis and Haemostasis
ITP immune thrombocytopenia
ITP idiopathic thombocytopenic purpura
JW Jehovah’s Witness
LMWH low-molecular-weight heparin
LPR low on-treatment platelet reactivity
MA maximum amplitude
MB methylene blue
MCF maximum clot firmness
MI myocardial infarction
MPS myeloproliferative syndrome
MTP massive transfusion protocol
xiv Abbreviations
xvii
xviii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
Part I
Pre-operative Hemostasis
The Cell-Based Coagulation Model
1
Christoph Sucker and Rainer B. Zotz
1.1 Introduction
C. Sucker
LaboMed Gerinnungszentrum Berlin, Tauentzienstrasse 7 b/c,
Berlin 10789, Germany
e-mail: [email protected]
R.B. Zotz (*)
Centrum für Blutgerinnungsstörungen und Transfusionsmedizin (CBT),
Immermannstrasse 65 a, Düsseldorf 40210, Germany
e-mail: [email protected]
(Arthus and Pagès 1890). Platelets were discovered in 1865 and described further in
the following years (Brewer 2006).
Paul Morawitz (1879–1936) published his classic theory of plasmatic hemostasis
just over 100 years ago. According to his model, prothrombin is converted to throm-
bin by tissue-derived thrombokinase; thrombin then converts fibrinogen to fibrin –
the most important step of coagulation (Morawitz 1905). This first concept of
hemostasis was modified and extended over the following years. In particular, more
coagulation factors were discovered, allowing for the development of more detailed
concepts of the hemostatic process (Wright 1962; Douglas 1999). This resulted in
the publication of the “Cascade Model” by MacFarlane and the “Waterfall Sequence
Model” of plasmatic hemostasis by Davie and Ratnoff (MacFarlane 1964; Davie
and Ratnoff 1964).
In the following sections, we illustrate the process of primary hemostasis, defined
as all aspects of platelet adhesion and platelet aggregation, and secondary hemosta-
sis, defined as the process of fibrin formation and fibrin stabilization. In the final
section, we briefly review the most important antithrombotic mechanisms: the fibri-
nolytic system, protein C/S system, and antithrombin.
The term “primary hemostasis” encompasses all aspects of platelet adhesion and
aggregation. Apart from platelets, components of the vessel wall – subendothelial
matrix components in particular – and von Willebrand factor (vWF) are involved in
this process (Riddel et al. 2007). The whole process is depicted in Fig. 1.1.
A healthy endothelium provides a physiological barrier, preventing initiation of
hemostasis by subendothelial matrix components, and has specific antithrombotic
properties, e.g., the secretion of antithrombotic agents such as nitric oxide and pros-
taglandins. In the case of vascular injury, a transient local vasoconstriction slows
down the blood flow and reduces extravascular blood loss. Hemostasis is initiated
when the rupture of this physiological barrier exposes the subendothelium to the
blood. VWF is a large multimeric glycoprotein which plays a crucial role in the
initial adhesion of platelets, particularly in vascular regions with high shear rates.
When subendothelial collagen is exposed to blood due to a vascular injury, vWF
quickly binds to it, leading to conformational changes of its binding areas for plate-
lets. This allows platelets to bind to vWF, mediated by their vWF receptors, the
glycoprotein (GP) Ib/V/IX complex. This process leads to an approximation of
platelets to the endothelial lesion and an initial adhesion. The adhesion is then fur-
ther stabilized by direct interaction of platelet receptors with subendothelial matrix
components, such as contact of platelet collagen receptors GP Ia/IIa and VI with
subendothelial collagen. Platelet adhesion to the injured vessel wall and the release
of a variety of inductors lead to platelet activation, and through several processes a
shape change occurs. The shape change increases the surfaces of the reactive plate-
lets by conversion from a discoid to a globular shape. The excretion of the content of
the platelets’ α- and δ-granules leads to the release of a variety of components (e.g.,
1 The Cell-Based Coagulation Model 5
Platelet
GP IIb-IIIa
GP IIb-IIIa
Platelet
GP Ib-V-IX
GP VI GP Ia-IIa
vWF
Endothelial cell Endothelial cell
Fig. 1.1 Schematic view of platelet adhesion and aggregation. Following endothelial injury,
platelets adhere to collagen by interaction of the receptor glycoprotein (GP) Ib/V/IX with von
Willebrand factor which is bound to collagen. This adhesion is stabilized by direct interaction of
platelet collagen receptors GP Ia/IIa and GP VI with collagen. Following activation of the aggrega-
tion receptors GP IIb/IIIa, platelets aggregate, mediated by von Willebrand factor or fibrinogen
Table 1.1 Characteristics of coagulation factors (factor XII is not regarded as an essential coagulation
factor according to the current hemostasis model and is, therefore, not shown in the table)
Molecular Plasma
Coagulation factor Function weight (mg/l) half-life (h)
I Fibrinogen Substrate 3,000 90
II Prothrombin Serine protease 100 65
V Proaccelerin Cofactor 10 15
VII Proconvertin Serine protease 0.5 5
VIII Antihemophilic factor Cofactor 0.1 10
IX Christmas factor Serine protease 5 25
X Stuart-Prower factor Serine protease 10 40
XI Plasma thromboplastin Serine protease 5 45
antecedent
XIII Fibrin-stabilizing factor Transglutaminase 30 200
and stabilization are summarized in Table 1.1. These “coagulation factors” include
enzymes of the serine protease family and cofactors that have no enzymatic activity
of their own but enhance the reactions of the coagulation enzymes. Some factors,
such as tissue factor (formerly factor III) and calcium (formerly factor IV) are no
longer regarded as coagulation factors today. Unless activated coagulation factors
have an individual name, such as thrombin (factor IIa), they are labeled with the
number of the factor and the suffix “a.”
In Morawitz’s first model of plasmatic hemostasis, prothrombin converted to
thrombin in the presence of tissue-derived thromboplastin and calcium, and throm-
bin then converted fibrinogen to fibrin (Morawitz 1905). In subsequent years, more
coagulation factors were detected and characterized, which led to an extension and
adaptation of the Morawitz model. However, despite the description of ever more
components of hemostasis, the complete interaction of these coagulation factors
and the way in which they convert prothrombin to thrombin remained unclear for
decades (Riddel et al. 2007). In 1964, two groups of researchers independently
reported a model of hemostasis in which the activation of one coagulation factor
resulted in the activation of another in a cascade or waterfall sequence, finally
resulting in the generation of thrombin. The “Cascade Model” was published by
MacFarlane in Nature (MacFarlane 1964), shortly followed by an independent
publication of the “Waterfall Model,” reported in Science by Davie and Ratnoff
(Davie and Ratnoff 1964). In these closely related models, each coagulation factor
was activated by another in cascade sequences, resulting in thrombin generation
from prothrombin and conversion of fibrinogen to fibrin. Both models described
two different ways in which thrombin formation was induced: firstly there was the
intrinsic pathway, for which all the required components were regarded as being
present in the blood, and secondly there was the extrinsic pathway, which required
an initiation by extravascular, subendothelially localized, and membrane-bound
tissue factor (TF) which is identical to the “thromboplastin” reported by Morawitz
in 1905. Both models concluded with a final common pathway in which factor X
was activated by either the intrinsic or extrinsic pathway, leading to the conversion
1 The Cell-Based Coagulation Model 7
Fibrinogen Fibrin
The cascade-type model of hemostasis remained accepted for a long time. However,
certain observations led to criticism of this model as it became clear that it was not
able to describe the real hemostatic process in vivo. Critical failings in the earlier
coagulation models led to further research and the development of a cell-based
model of hemostasis by Monroe and Hoffman in 2001. Today this model is widely
accepted and regarded as the best description of the process of hemostasis (Hoffman
and Monroe 2001; Roberts et al. 2006; Monroe and Hofman 2006).
The cell-based coagulation model distinguishes three distinct overlapping
phases of the hemostatic process. According to this concept, initiation of coagula-
tion occurs when blood is exposed to TF, expressed on TF-bearing cells. TF then
binds to factor VII which leads to activation of factor VII to factor VIIa. If the stimu-
lus is strong enough, further coagulation factors are activated, which results in the
formation of small amounts of thrombin generated by the enzymatic cleavage of
prothrombin (Fig. 1.3a). The small amounts of thrombin formed in this step, how-
ever, are not sufficient for the conversion of fibrinogen to fibrin and for the stabiliza-
tion of the fibrin clot by means of coagulation factor XIII. In the second phase of the
coagulation process, the amplification, thrombin activates platelets which then pres-
ent their thrombogenic surface of negatively charged phospholipids. Here, the coag-
ulation process is catalyzed and thus intensified (Fig. 1.3b). Thrombin activates
further coagulation factors, which bind to the activated platelet surfaces and mas-
sively increase the thrombin generation in the propagation phase of the coagulation
process (Fig. 1.3c). The “thrombin burst” generated by these mechanisms leads to
8 C. Sucker and R.B. Zotz
IX
a
VII X
IXa
II
Xa
VII TF IIa
VIIa TF
vWF/VIII VIIIa
b
Platelet activation
II
Xa
IIa IIa
Va Platelet
Tissue-factor (TF)
bearing cell V Va
XI XIa
c
X II
Platelet
IIa
Fibrinogen Fibrin
1 The Cell-Based Coagulation Model 9
The fibrinolytic system allows for the lysis of a fibrin clot (fibrinolysis). The main
fibrinolytic enzyme, plasmin, is derived from its precursor plasminogen upon acti-
vation by tissue-type plasminogen activator (t-PA) or urokinase-type plasminogen
activator (u-PA), which is mainly present in the urine. Plasmin itself cleaves fibrin,
producing fibrin degradation products. The fibrinolytic system is strictly regulated.
The most important inhibitors are plasminogen activator inhibitor (PAI), which
inactivates the plasminogen activators t-PA and u-PA; plasmin inhibitor (previously
termed α2-antiplasmin), which inactivates plasmin; and thrombin-activatable fibri-
nolysis inhibitor (TAFI), which inhibits the action of plasmin on the fibrin clot. The
process of fibrinolysis is depicted in Fig. 1.4. Notably, fibrinolytic capacity differs
significantly among different tissues, with high activities in the genitourinary tract
and the oral mucosa. This tissue-dependent variability of fibrinolytic capacity pro-
vides an explanation of the bleeding pattern in patients with pathologically enhanced
fibrinolysis (hyperfibrinolysis).
Fig. 1.3 (a) Initiation phase of the cell-based model of hemostasis: the complex of tissue factor
(TF) and factor VII activates factor VII to factor VIIa. The complex of TF and factor VIIa activates
factors IX and X, resulting in the generation of thrombin (IIa) by means of the complex of factor
Xa and its cofactor, factor Va (activated by factor Xa). (b) Amplification phase of the cell-based
model of hemostasis: thrombin activates platelets, which then present a thrombogenic surface. On
this catalytic surface, other coagulation factors such as V and XI are activated, and factor VIII is
released by its carrier, von Willebrand factor. Factor XIa activates factor IX. Activated platelets bind
factors Va, VIIIa, and IXa on their surface. (c) Propagation phase of the cell-based model of hemo-
stasis: factor VIIIa/IXa complex activates factor X on the catalytic platelet surfaces, and factors Xa/
Va activate prothrombin (factor II), resulting in a massive generation of thrombin (factor IIa) – a
“thrombin burst.” This burst is able to convert fibrinogen to fibrin and, in addition, to activate factor
XIII to XIIIa, which cross-links the fibrin fibers and stabilizes the fibrin clot
10 C. Sucker and R.B. Zotz
PAI
Plasminogen
Plasminogen activators
(t-PA; u-PA)
Antiplasmin Plasmin
TAFI
Fibrin Fibrin
degradation products
The protein C/S system is the main system to terminate an activated clotting process
(Esmon 2006). When large quantities of thrombin are generated in the process of
plasmatic hemostasis, binding of thrombin to its endothelial receptor thrombomodulin
(TM), supported by additional binding of the endothelial protein C receptor
(EPCR), initiates the breakdown of plasmatic hemostasis. The complex of throm-
bin and TM activates protein C. Together with its cofactor, protein S, activated
protein C (aPC) cleaves the activated coagulation factors Va and VIIIa, which leads
to a breakdown of thrombin formation and, thus, to a cessation of fibrin formation.
Deficiencies of protein C and protein S as well as a frequent mutation in the factor
V gene that prevents its cleavage by activated protein C, called factor V Leiden, are
important inherited risk factors predisposing to thrombotic events. These throm-
botic risk factors impair the physiological inactivation of the hemostatic process by
aPC, leading to a prolonged and increased thrombin formation and thus an elevated
thrombotic risk.
1.4.3 Antithrombin
References
Arthus M, Pagès C (1890) Nouvelle theorie chimique de la coagulation du sang. Arch Physiol
Norm Pathol 5:739–746
Brewer DB (2006) Max Schultze (1865), G. Bizzozero (1882) and the discovery of the platelet. Br
J Haematol 133:251–258
Davie EW, Ratnoff OD (1964) Waterfall sequence of intrinsic blood clotting. Science
145:1310–1312
Douglas AS (1999) Historical review: coagulation history, Oxford 1951–1953. Br J Haematol
107:22–32
Esmon CT (2006) Inflammation and the activated protein C anticoagulant pathway. Semin Thromb
Haemost 32:49–60
Hoffman M, Monroe DM 3rd (2001) A cell-based model of hemostasis. Thromb Haemost
85:958–965
MacFarlane RG (1964) An enzyme cascade in the blood clotting mechanism, and its function as a
biologic amplifier. Nature 202:498–499
Monroe DM, Hofman M (2006) What does it take to make the perfect clot? Arterioscler Thromb
Vasc Biol 26:41–48
Morawitz P (1905) Die Chemie der Blutgerinnung. Ergebn Physiol 4:307–422
Riddel JP, Bradley EA, Miaskowski C, Lillicrap DP (2007) Theories of blood coagulation. J Pediatr
Oncol Nurs 24:123–131
Roberts HR, Hoffman M, Monroe DM (2006) A cell-based model of thrombin generation. Semin
Thromb Hemost 32(Suppl 1):32–38
Schmidt A (1872) Neue Untersuchungen über die Faserstoffesgerinnung. Pflügers Arch für die
gesamte Physiol 6:413–538
Schmidt A (1892) Zur Blutlehre. Vogel, Leipzig
Wright IS (1962) The nomenclature of blood clotting factors. Can Med Assoc J 86:373–374
Laboratory Testing of Hemostasis
2
Fanny Bonhomme and Pierre Fontana
2.1 Introduction
Hemostasis laboratories can carry out large numbers of assays either to obtain accu-
rate and comprehensive diagnoses of hemostatic abnormalities or to monitor anti-
thrombotic treatment. Activated partial thromboplastin time (aPTT) and prothrombin
time (PT) are the most prescribed routine tests.
The evaluation of a patient’s history, symptoms and clinical signs are essential to
assess their bleeding tendency and to determine which laboratory test to use.
Moreover, in order to interpret laboratory test results, it is important to understand
how the assays are performed and to be aware of their limitations.
A normal range (reference range) is defined as the interval into which 95 % of
the values of a reference population fall; thus, 2.5 % of values are inferior to the
lower limit, and 2.5 % are superior to the upper limit (Fig. 2.1). Applied to hemo-
static testing, this means, for example, that 2.5 % of healthy individuals have a
prolonged aPTT (longer than the upper limit).
F. Bonhomme (*)
Division of Anesthesiology, Geneva University Hospitals,
Geneva, Switzerland
e-mail: [email protected]
P. Fontana
Division of Angiology and Hemostasis, Geneva University Hospital
and Faculty of Medicine, Geneva, Switzerland
95 %
2.5 % 2.5 %
Activated partial thromboplastin time is a clotting assay that measures the intrin-
sic pathway and is dependent on the concentrations of contact-phase factors
(high-molecular-weight-kininogen (HMWK), prekallikrein (PK), factor XII),
intrinsic factors VIII, IX, and XI, and on the common pathway factors (II, V, X,
and fibrinogen).
2 Laboratory Testing of Hemostasis 15
XII XIIa
Trauma
XI XIa
aPTT VIIIa TF PT
X Xa X
Va
II IIa
Fibrinogen Fibrin
Common pathway
aPPT PT
Activator Thromboplastin
phospholipids calcium
calcium
Time : s Time : s
PT converted to % (calibration with thivolle straight line)
INR = (PT patient / PT control)ISI
ISI = international sensitivity index
Fig. 2.3 aPTT and PT principles. aPTT is the time it takes to form a clot after adjunction of calcium
and a surface-activating agent into plasma. PT is the time it takes to form a clot after adjunction of
calcium and thromboplastin into plasma
Prothrombin time, or Quick time, measures the extrinsic pathway. It assesses the
function of factor VII and of the common factors II, V, and X, and fibrinogen. PT is
the time it takes for a clot to form after having added calcium and thromboplastin to
the plasma. Expressed in seconds, PT is very short in normal individuals (12–13 s).
In some countries, PT is expressed as a percentage of the PT of control plasma. For
monitoring the treatment of vitamin K antagonist (VKA), PT is standardized accord-
ing to the characteristics of the thromboplastin and calibrators used and is expressed
as an international normalized ratio (INR).
An isolated prolonged PT may rarely reflect an inherited factor VII deficiency (1 in
500,000 of the general population). More commonly it reflects a moderate deficiency
in vitamin K-dependent factors.
Prolonged PT, associated with prolonged aPTT, can be due to (Kamal et al.
2007):
• Deficiencies in factors II, V, and X or fibrinogen
• Presence of factor inhibitors
2 Laboratory Testing of Hemostasis 17
• VKA treatment
• Direct thrombin inhibitor
• Vitamin K deficiency, malabsorption
• Liver disease
• Disseminated intravascular coagulation
• Dilutional coagulopathy
• Hemorrhage
2.3.4 Fibrinogen
Fibrinogen is the most abundant clotting protein in plasma (2–4 g/l). Fibrinogen
abnormalities can be either qualitative (dysfibrinogenemia) or quantitative (total
lack, afibrinogenemia, or partial deficiency, hypofibrinogenemia).
Fibrinogen measurement is usually performed using a functional qualitative
method (von Clauss chronometric assay); when a high concentration of thrombin is
added to diluted plasma, the clotting time is proportional to the level of clottable
fibrinogen. Fibrinogen activity levels can also be estimated using a prothrombin
time-based kinetic assay, which is a rapid, inexpensive, automated assay, although
less specific of fibrinogen activity.
Immunoassays for fibrinogen antigen quantification are also available, but are
not used for screening tests. These immunological assays measure the protein con-
centration rather than the functional activity of fibrinogen.
Fibrinogen measurement is indicated in cases of apparent bleeding symptoms or
in cases of suspicion of disseminated intravascular coagulation, hepatic insuffi-
ciency, or hyperfibrinolysis (Table 2.2).
A combination of tests for PT and aPTT is usually the first step of a coagulation
assessment. Then, according to the results, further assays may be performed (Sié
and Steib 2006).
Fig. 2.4 Factor assays Deficient plasma in factor Deficient plasma in factor
principles. Specific factor VIII, or IX, or XI or XII II, or V, or VII, or X
assays are based on the
ability of the patient’s diluted
plasma to change the clotting
times of specific factor- Activator Thromboplastin
deficient plasmas, measured phospholipids calcium
by aPTT or PT calcium
Patient plasma
aPPT PT
Time : s
inhibitor. The control plasma contains the coagulation factors that could be deficient
in patient plasma. When the mixture corrects the clotting time, it is indicative of one
or more factor deficiencies. When the mixture fails to correct the clotting time, it is
indicative of an inhibitor (specific factor inhibitor or lupus anticoagulant).
Primary hemostasis involves the vessel wall, endothelial cells, platelets, and some
serine proteins (von Willebrand factor (vWF), thrombin, and fibrinogen).
Many assays are available for platelet function testing; however, no laboratory
test can explore vessel walls or endothelial cells. Platelet function assays are time-
consuming, difficult, and very sensitive to pre-analytical variables (drugs, food,
20 F. Bonhomme and P. Fontana
These tests are useful in patients suspected with disease of primary hemostasis
(purpura, cutaneo-mucous bleeding) with a normal platelet count. After centrifuga-
tion, the platelet-rich plasma is incubated with various aggregation activators.
Activators commonly used for exploring the different activation pathways of plate-
lets are arachidonic acid, ADP, collagen, TRAP (thrombin receptor agonist pep-
tide), epinephrine, and ristocetin. The modification of light transmission due to
platelet aggregation induced by the agonist is then studied. Platelet secretion may
also be studied after platelet activation with several agonists. Light transmission
platelet aggregometry is usually performed in qualified laboratories.
vWD is the most frequent inherited bleeding disorder. The diagnosis is based on
bleeding symptoms associated with qualitative or quantitative defects of vWF.
The patient’s bleeding history, in particular, the family history, is of most importance
for the diagnosis of vWD. A panel of assays are available (Favaloro 2009):
• vWF antigen measures the amount of vWF; plasma vWF levels vary with blood
group; O blood group patients usually have lower vWF levels (up to 40 %) than
individuals of other blood groups.
• Factor VIII coagulant activity measures the functional activity of factor VIII.
• vWF ristocetin cofactor activity and vWF collagen-binding activity measure the
functional activity of vWF.
Further assays are necessary for the diagnosis of subtypes:
• Factor VIII binding assay measures the affinity of vWF for factor VIII (useful in
type 2 N vW disease).
• vWF multimer analysis shows how the vWF monomer is multimerized (joined
into chains).
• Ristocetin-induced platelet agglutination measures the sensitivity of vWF to ris-
tocetin (useful in type 2B vW disease).
Several tests are routinely available for assessing the action of anticoagulants such
as heparins or vitamin K antagonist. New direct oral anticoagulants (specific inhibi-
tors of thrombin or factor Xa) cannot be monitored using standard coagulation
assays and require specific tests. If necessary with regard to antiplatelet therapy, a
number of specialized assays are available to assess platelet inhibition, but their util-
ity in clinical and routine practice remains to be determined.
profile of low molecular weight heparins (LMWH) is more predictable, and moni-
toring should be considered only in selected cases (e.g., mild renal insufficiency or
extremely high or low body weight).
References
Chee YL, Crawford JC, Watson HG et al (2008) Guidelines on the assessment of bleeding risk
prior to surgery or invasive procedures. British Committee for Standards in Haematology. Br J
Haematol 140:496–504
Dawood BB, Lowe GC, Lordkipanidze M et al (2012) Evaluation of participants with suspected
heritable platelet function disorders including recommendation and validation of a streamlined
agonist panel. Blood. doi:10.1182/blood-2012-07-444281
24 F. Bonhomme and P. Fontana
Dieri Al R, de Laat B, Hemker HC (2012) Thrombin generation: what have we learned? Blood Rev
26:197–203
Favaloro EJ (2009) Toward a new paradigm for the identification and functional characterization
of von Willebrand disease. Semin Thromb Hemost 35:60–75
Harrison P (2005) The role of PFA-100 testing in the investigation and management of haemostatic
defects in children and adults. Br J Haematol 130:3–10
Hemker HC, Dieri Al R, De Smedt E, Béguin S (2006) Thrombin generation, a function test of the
haemostatic-thrombotic system. Thromb Haemost 96:553–561
Hoffman MM, Monroe DM (2005) Rethinking the coagulation cascade. Curr Hematol Rep
4:391–396
Kamal AH, Tefferi A, Pruthi RK (2007) How to interpret and pursue an abnormal prothrombin
time, activated partial thromboplastin time, and bleeding time in adults. Mayo Clin Proc
82:864–873
Kitchens CS (2005) To bleed or not to bleed? Is that the question for the PTT? J Thromb Haemost
3:2607–2611
Lippi G, Salvagno GL, Ippolito L et al (2010) Shortened activated partial thromboplastin time:
causes and management. Blood Coagul Fibrinolysis 21:459–463
Lippi G, Salvagno GL, Montagnana M et al (2012) Quality standards for sample collection in
coagulation testing. Semin Thromb Hemost 38:565–575
Olson JD (1999) Addressing clinical etiologies of a prolonged aPTT. CAP Today 13:28, 30, 32
passim
Pernod G, Albaladejo P, Godier A et al (2013) Management of major bleeding complications and
emergency surgery in patients on long-term treatment with direct oral anticoagulants, thrombin
or factor-Xa inhibitors: proposals of the working group on perioperative haemostasis (GIHP) –
March 2013. Arch Cardiovasc Dis 106:382–393
Segal JB, Dzik WH, Transfusion Medicine/Hemostasis Clinical Trials Network (2005) Paucity of
studies to support that abnormal coagulation test results predict bleeding in the setting of inva-
sive procedures: an evidence-based review. Transfusion 45:1413–1425
Sié P, Steib A (2006) Central laboratory and point of care assessment of perioperative hemostasis.
Can J Anaesth 53:S12–S20
Sié P, Samama CM, Godier A et al (2011) Surgery and invasive procedures in patients on long-
term treatment with direct oral anticoagulants: thrombin or factor-Xa inhibitors.
Recommendations of the Working Group on perioperative haemostasis and the French Study
Group on thrombosis and haemostasis. Arch Cardiovasc Dis 104:669–676
Viscoelastic Tests of Hemostasis
3
Catherine Heim and Patrick Schoettker
3.1 Introduction
Traditional plasma-based coagulation tests, such as prothrombin time (PT) and acti-
vated partial thromboplastin time (aPTT), have proved their utility in detecting abnor-
malities in the clotting factor cascade. However, these tests evaluate only the initiation
of the clotting process and correspond to artificially created segments of hemostasis.
They are therefore of limited value in the assessment of the in vivo clotting process.
Further, long turnaround times make them poorly suited to the management of acute
perioperative bleeding.
In a setting of massive bleeding, the absence of real-time assessment of a patients’
capacity for coagulation and his evolving requirements for blood products can be a
major issue, leading to empirical treatment and the potential for inappropriate
administration of blood products.
The need for a rapid, comprehensive, physiological assessment of the entire
process of coagulation, as well as the patient’s overall hemostatic capacity, has led to
the development of ‘global hemostasis assays’; these include viscoelastic tests which
allow for a rapid bedside analysis of the patient’s in vivo hemostatic condition.
Viscoelastic tests measure the continuous process of clotting in whole blood and its
effects on changes in viscosity, elasticity and stability, thus providing global infor-
mation on the dynamics of a clot’s development, its stabilization and ultimately its
dissolution (Nair et al. 2010). To date, these tests are the only ones available allow-
ing for a rapid identification of hyperfibrinolysis, a common pattern in massive
3.3 Technology
platelets are activated, expressing glycoprotein (GP) IIb/IIIa receptors, and fibrin is
formed and subsequently polymerized. The interactions between the GP IIb/IIIa
receptors and the polymerized fibrin increase the sample’s viscoelasticity which in
turn increases the torque between the sample cup and the pin. The suspended pin is
connected to a detector system, either a torsion wire (TEG®) or an optical detector
via the reflection of light onto a small mirror (ROTEM®). As the sample clotting
process progresses, fibrin strands form between the cup and the pin, further imped-
ing rotational movements. These changes are also detected, either mechanically or
28 C. Heim and P. Schoettker
4°45
b 1
3
2 5
10 11
7 6
9 8
optically, then transmitted electronically and transformed into numerical and graph-
ical read-outs (Figs. 3.1a, b). When no clotting takes place in a ROTEM® assay, for
example, the pin’s movement is not obstructed, whereas if a clot does form, it
attaches to the pin and cup surfaces, impairing pin movement. An amplitude of
0 mm means unobstructed oscillation (no clot), whilst an amplitude of 100 mm can
be regarded as total firmness—the pin is completely blocked by the clot.
Whilst TEG® originally used non-anticoagulated whole blood, today this method is
essentially only used in research settings. For clinical use, both systems now employ
citrated whole blood which is recalcified to initiate coagulation. This allows longer
sample storage: in the case of ROTEM®, up to 120 min (Theusinger et al. 2010b).
It must be noted that blood sampling using sodium citrate tubes dilutes the blood
3 Viscoelastic Tests of Hemostasis 29
A30 A60
TEG analysis
K
R MA
CL
Clot firmness
α
CT MCF LY
CFT
ROTEM analysis
A5 A10 A20 A30 A60
Time
Fig. 3.2 TEG® and ROTEM ® output demonstrating clot initiation, propagation, stabilization and
lysis. TEG® parameters: R reaction time, K kinetics, α alpha angle, MA maximum amplitude, CL
clot lysis, A amplitude at set time in min (30 and 60). ROTEM® parameters: CT clotting time, CFT
clot formation time, α alpha angle, MCF maximum clot firmness, A amplitude at set time in min
(5, 10, 20 and 30), LY clot lysis
3 Viscoelastic Tests of Hemostasis 31
concentrations. Clinical reference values differ between the two systems and must
be interpreted accordingly. Reference values for TEG® are based on unspecified
surgical patient samples of limited size (Haemoscope Corporation 2007), whilst
those established for ROTEM® were determined in a multicenter study of patients
and healthy volunteers (Lang et al. 2005).
There are also differences in nomenclature between the devices and other minor
details exist (Table 3.3 and Fig. 3.2).
R ‘Reaction time’ (seconds). Time from the start of the test to initial fibrin
formation; corresponding to a clot amplitude of 2 mm. This represents the
enzymatic portion of the coagulation process.
Simplified: R represents coagulation factors.
K ‘Kinetics’ (seconds). Time necessary to achieve a given clot strength of an
amplitude of 20 mm.
Simplified: K represents thrombin.
α Alpha angle. The slope between R and K. Corresponds to the speed of fibrin
build-up and cross-linking/clot strengthening. Dependent on fibrinogen levels.
Simplified: α represents the speed of clot formation.
TEG® and ROTEM® basically provide similar informations on the kinetics and
strengths of clot formation and have been shown to produce comparable results
when samples were activated with the same exogenous activator (Zambruni et al.
2004). However, differences in operating characteristics when different activators
are used render their results non-interchangeable (Nielsen et al. 2005; Nielsen 2007;
Venema et al. 2010). The use of different nomenclature for identical parameters also
makes comparison difficult.
Depending on the activator used, clotting speed can vary significantly. INTEM-
activated plasma (ROTEM®), for example, generates a time to clot initiation three
times faster than kaolin-activated plasma, as used in TEG® (Nielsen 2007).
3 Viscoelastic Tests of Hemostasis 33
Further, viscoelastic test results are also dependent on the concentration of activa-
tor in the sample as this particularly affects clot initiation and propagation by influenc-
ing thrombin generation (Nielsen et al. 2005). Differences in the composition of the
plastic polymer sample cups, which could generate greater surface charges, could
potentially contribute an additional source of differences in results (Roche et al. 2006).
It has been demonstrated that the plastic tube in which citrated blood is collected may
provide additional inference in the activation of coagulation (Roche et al. 2006).
However, it seems unlikely that these would affect clinical decision making (Nielsen
2007), and relative trends within one method are still expected to be comparable.
These specificities explain part of the differences in the reference ranges shown
in Table 3.4. The manufacturer of TEG® recommends that each institution should
determine its own normal values.
Another difference is the speed of result acquisition, with ROTEM® having a
faster turnaround time (5–10 min) than TEG® (15–20 min). The main reason for this
is that ellagic acid (ROTEM®) activates coagulation faster than kaolin (TEG®).
The two methods have been the subjects of numerous claims about their equiva-
lency or superiority, but neither has actually been proven superior. One study compar-
ing the diagnostic performances of TEG® and ROTEM® in the detection of dilutional
coagulopathy, thrombocytopenia, hyperfibrinolysis and the presence of heparin sug-
gested that ROTEM® not only readily distinguishes all the types of coagulopathy cited
but also provides faster diagnosis; TEG®, however, failed to distinguish dilutional
coagulopathy from thrombocytopenia (Larsen et al. 2011). On the other hand,
Table 3.4 Reference ranges for TEG® (from ‘User manual TEG® 5000 Thromboelastograph
Hemostasis System’) and EXTEM (ROTEM®) (Lang et al. 2005)
Angle MA Sample
Sample type R (min) K (min) (degree) (mm) G (kd/sc) sizea
Celite/kaolin 4–8 0–4 47–74 54–72 6.0–13.2 132
Sodium citrate celite/kaolin 2–8 1–3 55–78 51–69 4.6–10.9 98
Native 12–26 3–13 14–46 42–63 3.2–7.1 132
Sodium citrate native 9–27 2–9 22–58 44–64 3.6–8.5 132
Tissue factor 1–3 1–3 57–78 55–75 6.0–13.0 178
Sodium citrate plus TF 0–2 0–5 52–82 46–72 2.7–12.5 41
Tissue factor kaolin 17–38 30–118 66–82 54–72 5.3–12.4 86
Citrated tissue factor kaolin 22–44 34–138 64–80 52–71 5.0–11.6 89
Tissue factor plus functional – – – 9–29 0–2.0 72
fibrinogen
Citrated tissue factor plus – – – 10–25 0.5–1.7 72
functional fibrinogen
Test Normal range (median)
CT (s) 42–74 (55)
CFT (s) 46–148 (95)
A10 (mm) 43–65 (53)
MCF (mm) 49–71 (60)
ML (%) 0–18 (4)
a
Sample sizes range from 41 to 78 depending on the participating hospitals
34 C. Heim and P. Schoettker
ROTEM® has a lower sensitivity to the effect of aspirin on platelets and is, unlike
TEG®, unable to detect the effect of low molecular weight heparin (LMWH). TEG®
has the advantage of offering a specific test of platelet function, the TEG® Platelet
Mapping™ assay. This test analyses platelet function by measuring clot strength and
maximum amplitude, reflecting maximum platelet function and detecting reduction
in platelet function, represented as percentage of inhibition (see Chap. 4).
Whilst the ROTEM® FIBTEM assay has been validated as an accurate estimate
of the fibrinogen contribution to clot strength independent of platelets, the TEG®
Functional Fibrinogen assay requires further validation (Solomon et al. 2012).
Neither test is a suitable substitute for the traditional plasma-based determination of
fibrinogen levels using the Clauss method. Furthermore, the correlation between
FIBTEM MCF and the plasmatic concentration of fibrinogen worsens after exoge-
nous fibrinogen administration (Solomon et al. 2011).
Based on these differences, caution is warranted when interpreting comparative
data generated by TEG® and ROTEM®. TEG®-based treatment algorithms should
not be used thoughtlessly for ROTEM®-based sample analysis and vice versa.
Therefore, whilst these two devices can, under similar circumstances, generate sim-
ilar data, their processes, involving different activator agents, are not equivalent and
must be well understood.
The assessments made by TEG® and ROTEM® are illustrated graphically along
the horizontal time axis (left to right). The shapes of these representations are
often as useful as the results of individual values. The types of activators used need
to be taken into account when interpreting reference ranges (Figs. 3.3 and 3.4).
Examples from ROTEM traces with permission from (www.rotem.de)
Normal
R; K; MA; Angle = Normal
Anticoagulants/hemophilia
Factor Deficiency
R; K = Prolonged;
MA; Angle = Decreased
Platelet Blockers
Thrombocytopenia/
Thrombocytopathy
R ~ Normal; K = Prolonged;
MA = Decreased
Hypercoagulation
R; K = Decreased;
MA; Angle = Increased
D.I.C
Stage 1
Hypercoagulable state with
secondary fibrinolisis
Stage 2
Hypocoagulable state
Fig. 3.3 TEG output in various clinical situations (From ‘User manual TEG® 5000
Thromboelastograph Hemostasis System’)
36 C. Heim and P. Schoettker
a Normal patient:
EXTEM INTEM
CT: 67s CFT: 87s a: 73° CT: 200s CFT: 67s a: 77°
CFR: 54mm MCF: 57mm ML: −% CFR: 54mm MCF: 61mm ML: −%
FIBTEM APTEM
CT: 66s CFT: −s a: 57° CT: 74s CFT: 89s a: 72°
CFR: 9mm MCF: 10mm ML: −% CFR: 53mm MCF: 61mm ML: −%
b Platelet deficiency:
EXTEM INTEM
CT: 57s CFT: 444s a: 80° CT: 200s CFT: 449s a: 72°
A10: 23mm MCF: 35mm ML: −% A10: 23mm MCF: 32mm ML: −%
FIBTEM APTEM
CT: 67s CFT: −s a: −° CT: 52s CFT: 398s a: 80°
A10: 15mm MCF: 16mm ML: −% A10: 25mm MCF: 35mm ML: −%
c Fibrinogen deficiency:
EXTEM INTEM
CT: 109s CFT: 263s a: 48° CT: 236s CFT: 220s a: 55°
A10: 31mm MCF: 38mm ML: −% A10: 33mm MCF: 42mm ML: −%
FIBTEM APTEM
CT: 185s CFT: −s a: −° CT: 98s CFT: 276s a: 46°
A10: 3mm MCF: 3mm ML: −% A10: 31mm MCF: 40mm ML: −%
d Hyperfibrinolysis:
EXTEM INTEM
CT: 59s CFT: 130s a: 65° CT: 200s CFT: 88s a: 74°
A10: 44mm MCF: 48mm ML: 100% A10: 46mm MCF: 48mm ML: 100%
FIBTEM APTEM
CT: 51s CFT: −s a: −° CT: 62s CFT: 132s a: 64°
A10: 7mm MCF: 7mm ML: 94% A10: 44mm MCF: 55mm ML: 0%
e Heparin influence:
EXTEM INTEM
CT: 67s CFT: 104s a: 68° CT: 852s CFT: 198s a: 51°
A10: 50mm MCF: 57mm ML: 0% A10: 41mm MCF: 48mm ML: 0%
FIBTEM HEPTEM
CT: 63s CFT: –s a: -° CT: 202s CFT: 75s a: 76°
A10: 6mm MCF: 8mm ML: 0% A10: 52mm MCF: 58mm ML: 0%
Several issues are as yet unresolved with regard to the standardization of methodol-
ogy and the interpretation of currently available viscoelastic tests. To date, neither
TEG® nor ROTEM® has been validated for clinical use. Standardization of inter-
pretation and evidence-based treatment algorithms are lacking and subject to
intense research. Further, significant inter-laboratory variability has been revealed
(with coefficients of variation exceeding 10 %) and needs to be addressed (Kitchen
et al. 2010; Chitlur et al. 2011). Regular external quality controls and proficiency
testing should be mandatory. Personnel using these devices need to be adequately
trained to avoid manipulation errors (Ganter and Hofer 2008).
Due to the fact that it is a mechanical device, with its pin in free suspension,
TEG® is easily affected by external vibrations or shocks and thus needs to be per-
formed in a vibration-proof environment. This may indeed be a difficult require-
ment for a point-of-care device in an emergency setting. The automated pipette,
touch screen and intuitive software of ROTEM®, on the other hand, are seen by
many as particularly user friendly.
TEG® and ROTEM® analyses are commonly used as point-of-care coagulation
tests in emergency and operation rooms as well as in intensive care settings.
3 Viscoelastic Tests of Hemostasis 41
Conclusion
TEG® and ROTEM® technology allows a real-time assessment of the viscoelas-
tic properties of clot formation and lysis in whole blood. Running several sam-
ples in parallel, using a variety of activators and inhibitors, allows detailed
separate analysis of clot initiation, propagation, stabilization and dissolution.
The influence of various components can be distinguished. Many newer diagno-
sis and treatment algorithms for bleeding patients include the use of TEG® or
ROTEM®. However, despite evidence that such algorithms reduce blood product
use and improve clinical outcomes, further studies are warranted in order to
determine a clinical correlation with their results and to allow for the develop-
ment of evidence-based treatment guidelines.
References
Afshari A, Wikkelso A et al (2011) Thromboelastography (TEG) or thromboelastometry (ROTEM)
to monitor haemotherapy versus usual care in patients with massive transfusion. Cochrane
Database Syst Rev (3):CD007871
Ak K, Isbir CS et al (2009) Thromboelastography-based transfusion algorithm reduces blood
product use after elective CABG: a prospective randomized study. J Card Surg 24(4):404–410
Brohi K (2009) Diagnosis and management of coagulopathy after major trauma. Br J Surg
96(9):963–964
Camenzind V, Bombeli T et al (2000) Citrate storage affects thrombelastograph analysis.
Anesthesiology 92(5):1242–1249
Chitlur M, Sorensen B et al (2011) Standardization of thromboelastography: a report from the
TEG-ROTEM working group. Haemophilia 17(3):532–537
Davenport R, Manson J et al (2011) Functional definition and characterization of acute traumatic
coagulopathy. Crit Care Med 39(12):2652–2658
Ganter MT, Hofer CK (2008) Coagulation monitoring: current techniques and clinical use of vis-
coelastic point-of-care coagulation devices. Anesth Analg 106(5):1366–1375
Gorlinger K, Dirkmann D et al (2011) First-line therapy with coagulation factor concentrates com-
bined with point-of-care coagulation testing is associated with decreased allogeneic blood
transfusion in cardiovascular surgery: a retrospective, single-center cohort study. Anesthesiology
115(6):1179–1191
Haemoscope Corporation (2007) Specifications and performance characteristics. In: TEG5000
thromboelastograph hemostasis system user manual. Haemoscope Corporation: Niles, USA
p 177
Hartert H (1948) Blutgerinningsstudien mit der thrombelastographie, einem neuen untersuchings-
verfahren. Klin Wochenschr 26(37–38):577–583
Holcomb JB, Minei KM et al (2012) Admission rapid thrombelastography can replace conven-
tional coagulation tests in the emergency department: experience with 1974 consecutive trauma
patients. Ann Surg 256(3):476–486
42 C. Heim and P. Schoettker
Jambor C, Reul V et al (2009) In vitro inhibition of factor XIII retards clot formation, reduces
clot firmness, and increases fibrinolytic effects in whole blood. Anesth Analg 109(4):
1023–1028
Johansson PI (2009) Hemostatic strategies for minimizing mortality in surgery with major blood
loss. Curr Opin Hematol 16(6):509–514
Johansson PI, Sorensen AM et al (2013) Low hemorrhage-related mortality in trauma patients in a
level I trauma center employing transfusion packages and early thromboelastography-directed
hemostatic resuscitation with plasma and platelets. Transfusion 53(12):3088–3099
Kashuk JL, Moore EE et al (2012) Initial experiences with point-of-care rapid thrombelastography
for management of life-threatening postinjury coagulopathy. Transfusion 52(1):23–33
Kitchen DP, Kitchen S et al (2010) Quality assurance and quality control of thrombelastography
and rotational thromboelastometry: the UK NEQAS for blood coagulation experience. Semin
Thromb Hemost 36(7):757–763
Lang T, Bauters A et al (2005) Multi-centre investigation on reference ranges for ROTEM throm-
boelastometry. Blood Coagul Fibrinolysis 16(4):301–310
Larsen OH, Fenger-Eriksen C et al (2011) Diagnostic performance and therapeutic consequence of
thromboelastometry activated by kaolin versus a panel of specific reagents. Anesthesiology
115(2):294–302
Levrat A, Gros A et al (2008) Evaluation of rotation thrombelastography for the diagnosis of
hyperfibrinolysis in trauma patients. Br J Anaesth 100(6):792–797
McCrath DJ, Cerboni E et al (2005) Thromboelastography maximum amplitude predicts postop-
erative thrombotic complications including myocardial infarction. Anesth Analg 100(6):
1576–1583
Nair SC, Dargaud Y et al (2010) Tests of global haemostasis and their applications in bleeding
disorders. Haemophilia 16(Suppl 5):85–92
Nielsen VG (2007) A comparison of the thrombelastograph and the ROTEM. Blood Coagul
Fibrinolysis 18(3):247–252
Nielsen VG, Cohen BM et al (2005) Effects of coagulation factor deficiency on plasma coagula-
tion kinetics determined via thrombelastography: critical roles of fibrinogen and factors II, VII,
X and XII. Acta Anaesthesiol Scand 49(2):222–231
Rahe-Meyer N, Solomon C et al (2009) Multicentric comparison of single portion reagents and
liquid reagents for thromboelastometry. Blood Coagul Fibrinolysis 20(3):218–222
Roche AM, James MF et al (2006) Just scratching the surface: varied coagulation effects of poly-
mer containers on TEG variables. Eur J Anaesthesiol 23(1):45–49
Romlin BS, Wahlander H et al (2013) Earlier detection of coagulopathy with thromboelastometry
during pediatric cardiac surgery: a prospective observational study. Paediatr Anaesth 23(3):
222–227
Solomon C, Cadamuro J et al (2011) A comparison of fibrinogen measurement methods with fibrin
clot elasticity assessed by thromboelastometry, before and after administration of fibrinogen
concentrate in cardiac surgery patients. Transfusion 51(8):1695–1706
Solomon C, Sorensen B et al (2012) Comparison of whole blood fibrin-based clot tests in thromb-
elastography and thromboelastometry. Anesth Analg 114(4):721–730
Sorensen B, Ingerslev J (2005) Tailoring haemostatic treatment to patient requirements - an update
on monitoring haemostatic response using thrombelastography. Haemophilia 11(Suppl 1):1–6
Spalding GJ, Hartrumpf M et al (2007) Cost reduction of perioperative coagulation management
in cardiac surgery: value of “bedside” thrombelastography (ROTEM). Eur J Cardiothorac Surg
31(6):1052–1057
Tapia NM, Chang A et al (2013) TEG-guided resuscitation is superior to standardized MTP resus-
citation in massively transfused penetrating trauma patients. J Trauma Acute Care Surg
74(2):378–385, discussion 385–376
Theusinger OM, Baulig W et al (2010a) In vitro factor XIII supplementation increases clot firm-
ness in rotation thromboelastometry (ROTEM). Thromb Haemost 104(2):385–391
Theusinger OM, Nurnberg J et al (2010b) Rotation thromboelastometry (ROTEM) stability and
reproducibility over time. Eur J Cardiothorac Surg 37(3):677–683
3 Viscoelastic Tests of Hemostasis 43
Venema LF, Post WJ et al (2010) An assessment of clinical interchangeability of TEG and RoTEM
thromboelastographic variables in cardiac surgical patients. Anesth Analg 111(2):339–344
Wallin O, Soderberg J et al (2008) Preanalytical effects of pneumatic tube transport on routine
haematology, coagulation parameters, platelet function and global coagulation. Clin Chem Lab
Med 46(10):1443–1449
Westbrook AJ, Olsen J et al (2009) Protocol based on thromboelastograph (TEG) out-performs
physician preference using laboratory coagulation tests to guide blood replacement during and
after cardiac surgery: a pilot study. Heart Lung Circ 18(4):277–288
Wilson D, Cooke EA et al (2001) Changes in coagulability as measured by thrombelastography
following surgery for proximal femoral fracture. Injury 32(10):765–770
Zambruni A, Thalheimer U et al (2004) Thromboelastography with citrated blood: comparability
with native blood, stability of citrate storage and effect of repeated sampling. Blood Coagul
Fibrinolysis 15(1):103–107
Point-of-Care Platelet Function Tests
4
Gabriele Casso, Fabio Lanzi, and Carlo E. Marcucci
4.1 Introduction
Platelets are small, discoid, anucleate blood cells produced by cytoplasmic frag-
mentation of megakaryocytes (George 2000). In the adult, 1 × 1012 inactive blood
platelets are continuously flowing over 1,000 m2 of vascular surface with minimal
adhesion or aggregation (Versteeg et al. 2013). Platelets play a key role in primary
hemostasis. Their main function, when activated, is to stop hemorrhage after tissue
trauma and vascular injury. However, platelets are also important contributors to
pathological thrombotic disorders (e.g., acute coronary syndromes, ischemic stroke,
peripheral occlusive arterial disease) (Davi and Patrono 2007; Jennings 2009) and
an increasing number of patients require long-term antiplatelet therapy.
Platelet activation by the principal agonists during primary hemostasis and the
site of action of antiplatelet drugs are described in Fig. 4.1.
Even though inherited or acquired platelet dysfunction can influence bleeding
during surgery, evidence for the usefulness of perioperative platelet function analy-
sis is still controversial (Kehrel and Brodde 2013).
The European Society of Anaesthesiology (ESA) recently published guidelines
on the management of severe perioperative bleeding (Kozek-Langenecker et al.
2013). Recommendations about perioperative platelet function analysis are weak.
Preoperative platelet function testing is only suggested in relation to a positive
bleeding anamnesis. In cases with a suspicion of decreased platelet function caused
by medical conditions or by antiplatelet therapy, platelet function assessment can be
used preoperatively.
Inhibitor drugs
Platelet agonists
Thrombin Vorapaxar
Aggregation Thromboxane A2 COX-1 inhibitor
NSAID
Platelet PAR1/4 receptor
Fibrinogen
Thromboxane receptor
Ticlopidine
ADP Clopidogrel
Abcximab GPIIb/IIIa Prasugrel
Activation P2Y12 Ticagrelor
Eptifibatide
receptor Cangrelor
Tirofiban
GP Ib/IX/V Adhesion
GP VI von Willebrand factor
Collagen
Endothelial cell
Collagen fibers
Fig. 4.1 Platelet agonists involved in platelet activation and site of action of antiplatelet drugs
Various methods for platelet function analysis exist today (Harrison 2005; Michelson
2009; Pakala and Waksman 2011; Kehrel and Brodde 2013). Most of them must be
performed in a laboratory setting on centrifuged platelet-rich plasma: light transmis-
sion aggregometry, for example, originally described by Born in 1962 (Born 1962),
is still considered the “gold standard.” These laboratory tests are time-consuming
and require training and laboratory skills.
Recently, several whole blood point-of-care tests have been developed which
can be performed in a near-patient setting, for example, in operating theaters or
intensive care units (Enriquez and Shore-Lesserson 2009). The potential advan-
tages of these tests are simplified workflows (no transport of samples to labora-
tory), rapid turnaround times, and targeted management of coagulation disorders.
4 Point-of-Care Platelet Function Tests 47
Platelet plug
No blood flow
Blood flow
Closure time
Capillary tubes
membranes are coated with collagen and either epinephrine (COL/EPI) or adenos-
ine diphosphate (COL/ADP). A new cartridge (Innovance® PFA P2Y) using a mem-
brane coated with ADP, prostaglandin E1, and calcium has recently become
available; it is supposed to be more sensitive to P2Y12 inhibitors. The activators, in
association with the high shear rates, induce platelet activation and aggregation
leading to a gradual occlusion of the small aperture by a growing platelet plug. Time
to complete occlusion of the membrane orifice, inducing cessation of blood flow, is
termed closure time (CT) and expressed in seconds (s). Platelet function is inversely
correlated to CT. The test is completed in 5–8 min (maximum CT 300 s) and must
be performed within 4 h of blood sample collection.
4.2.2 Plateletworks®
Table 4.2 Drug sensitivity, advantages, and disadvantages of the (PFA)-100/200® system
Assessment of drug
effect
+ −
Aspirin Rapid, automated, and easy Requires pipetting
P2Y12 inhibitors test
Dependent on hematocrit
Can be used by non-skilled and platelet count
personnel
sample (1 ml). The second step is to repeat the platelet count on another citrated
sample that has been exposed to a known platelet agonist (collagen, ADP, or AA).
The agonist will stimulate functional platelets to aggregate into clumps. These
aggregates exceed the threshold limitations for platelet size and the cell counter no
longer counts them as platelets. The difference in the platelet count between the
baseline and agonist platelet count (APC) in the stimulated samples provides a
direct measurement of platelet aggregation.
AA ADP Collagen
(aggregation) (aggregation) (aggregation) Interpretation
4.2.3 VerifyNow®
Table 4.5 Drug sensitivity, advantages, and disadvantages of the Plateletworks® system
Assessment of drug
effect
+ −
Aspirin Rapid and easy test Analysis time dependent
P2Y12 inhibitors (sample must be examined
GP IIb/IIIa inhibitors Whole blood, no requirements within 10 min of
for sample preparation collection)
Provides CBC and platelet
count Few clinical studies
Can be used in
thrombocytopenic patients
Disposable cartridge
Mixing chambers
contain fibrinogen-coated
Agglutinated bead-platelet beads and platelet activators
complexes precipitate
GP IIb/IIIa Fibrinogen
blood sample is directly inserted onto a special disposable cartridge. The cartridges’
mixing chambers contain fibrinogen-coated polystyrene beads and platelet activa-
tors: 4 μM of TRAP in the VerifyNow IIb/IIIa test®, 1 mM of AA in the VerifyNow
Aspirin test®, and 20 μM ADP + 22 nM PGE1 in the VerifyNow PRU test®.
4 Point-of-Care Platelet Function Tests 53
Table 4.6 Baseline values and expected ranges after abciximab and eptifibatide treatment for the
VerifyNow IIb/IIIa ® assay
Drug Baseline prior to drug administration ≥80 % inhibition ≥95 % inhibition
abciximab 125–330 PAU 0–44 PAU 0–13 PAU
eptifibatide 136–288 PAU 0–31 PAU 0–10 PAU
PAU platelet aggregation units
Table 4.7 Drug sensitivity, advantages, and disadvantages of the VerifyNow® system
Assessment of drug
effect
+ −
Aspirin Rapid and simple test (real Long incubation time for
P2Y12 inhibitors point-of-care) VerifyNow Aspirin test®
GP IIb/IIIa inhibitors
Can be used by non-skilled Cannot be used to detect
personnel
congenital platelet disorders
Does not require pipetting
Influence of thrombocytopenia
No sample preparation is unknown
required
2 sensor units
Hirudin blood
+ agonist
200 AU 200 AU
Aggregation (AU)
Aggregation (AU)
0 AU 0 AU
Time Time
thus allowing them to adhere to vascular injuries and artificial surfaces. The
Multiplate® analyzer provides a disposable test cell containing two independent
sensor units, each consisting of two, fine, highly conductive electrodes. A small
quantity (300 μl) of hirudin-anticoagulated whole blood is pipetted in the test cell,
mixed with 300 μl of saline using a stirring magnetic bar, and incubated at 37 °C for
3 min. A specific platelet activator is then added to the solution. Activated platelets
56 G. Casso et al.
Table 4.8 Reference ranges for the Assay AUC reference ranges
Multiplate® assay
ASPI test 71–115 U
ADP test 57–113 U
TRAP test 94–156 U
COL test 46–116 U
RISTO test 90–201 U
AUC area under the curve, U aggregation unit
adhere to and aggregate on the sensor electrodes; this leads to an increase in the
impedance measured between them. The impedance is recorded over a 6-min period
and displayed graphically. Both sensor units in each test cell measure the rise in
impedance independently. As an internal quality assessment, the two measurements
must coincide for the test to be valid.
The Multiplate® analyzer is equipped with five channels for the simultaneous
measurement of different samples and/or agonists. A wide array of different platelet
agonists is available to permit a differentiated diagnosis of acquired and inherited
platelet dysfunction:
ASPI test: cyclooxygenase-dependent aggregation (stimulation by 0.5 mM of AA)
sensitive to ASA, NSAIDs, and other inhibitors of platelet COX-1.
ADP test: ADP-induced platelet activation (stimulation by 6.5 μM ADP) sensitive
to clopidogrel, prasugrel, ticagrelor, and other P2Y12 receptor antagonists.
TRAP test: platelet stimulation via the thrombin receptor (using 32 μM of thrombin
receptor-activating peptide), sensitive to GP IIb/IIIa receptor antagonists (e.g.,
abciximab, eptifibatide, and tirofiban).
COL test: collagen-induced aggregation (using 3.2 μg of collagen).
RISTO test: vWF- and GP Ib-dependent aggregation (using 0.77 mg/ml ristocetin).
Table 4.9 Drug sensitivity, advantages, and disadvantages of the Multiplate® system
Assessment of drug
effect
+ −
MAFibrin
MAADP or AA – MAFibrin
% Inhibition = 100 – × 100
MAThrombin – MAFibrin
Time (min)
Fig. 4.5 Calculation of TEG Platelet Mapping® results. MA maximum amplitude of the TEG
assay expressed in millimeter (mm)
Table 4.11 Drug sensitivity, advantages, and disadvantages of the TEG Platelet Mapping® system
Assessment of drug
effect
+ −
Aspirin Whole blood Use of 3 different channels for
P2Y12 inhibitors measurements (2 TEG
GP IIb/IIIa inhibitors Low sample volume devices)
period in comparison to applying arbitrary delays and results in blood loss compa-
rable to clopidogrel-naive patients undergoing the same type of surgery (Mahla
et al. 2012).
References
Al-Azzam SI, Alzoubi KH et al (2012) The prevalence and factors associated with aspirin resis-
tance in patients premedicated with aspirin. Acta Cardiol 67(4):445–448
Alstrom U, Granath F et al (2009) Platelet inhibition assessed with VerifyNow, flow cytometry and
PlateletMapping in patients undergoing heart surgery. Thromb Res 124(5):572–577
Bliden KP, DiChiara J et al (2007) Increased risk in patients with high platelet aggregation receiv-
ing chronic clopidogrel therapy undergoing percutaneous coronary intervention: is the current
antiplatelet therapy adequate? J Am Coll Cardiol 49(6):657–666
Bochsen L, Wiinberg B et al (2007) Evaluation of the TEG platelet mapping assay in blood donors.
Thromb J 5:3
Born GV (1962) Aggregation of blood platelets by adenosine diphosphate and its reversal. Nature
194:927–929
Cammerer U, Dietrich W et al (2003) The predictive value of modified computerized thromboelas-
tography and platelet function analysis for postoperative blood loss in routine cardiac surgery.
Anesth Analg 96(1):51–57, table of contents
Cardinal DC, Flower RJ (1979a) The ‘electronic platelet aggregometer’ [proceedings]. Br J
Pharmacol 66(1):138P
4 Point-of-Care Platelet Function Tests 61
Cardinal DC, Flower RJ (1979b) The study of platelet aggregation in whole blood [proceedings].
Br J Pharmacol 66(1):94P–95P
Collet JP, Cuisset T et al (2012) Bedside monitoring to adjust antiplatelet therapy for coronary
stenting. N Engl J Med 367(22):2100–2109
Collyer TC, Gray DJ et al (2009) Assessment of platelet inhibition secondary to clopidogrel and
aspirin therapy in preoperative acute surgical patients measured by Thrombelastography
Platelet Mapping. Br J Anaesth 102(4):492–498
Craft RM, Chavez JJ et al (2004) A novel modification of the Thrombelastograph assay, isolating
platelet function, correlates with optical platelet aggregation. J Lab Clin Med 143(5):
301–309
Craft RM, Chavez JJ et al (2005) Comparison of modified Thrombelastograph and Plateletworks
whole blood assays to optical platelet aggregation for monitoring reversal of clopidogrel inhi-
bition in elective surgery patients. J Lab Clin Med 145(6):309–315
Dalen M, van der Linden J et al (2012) Correlation between point-of-care platelet function testing
and bleeding after coronary artery surgery. Scand Cardiovasc J 46(1):32–38
Davi G, Patrono C (2007) Platelet activation and atherothrombosis. N Engl J Med 357(24):
2482–2494
Edwards A, Jakubowski JA et al (2012) Evaluation of the INNOVANCE PFA P2Y test cartridge:
sensitivity to P2Y(12) blockade and influence of anticoagulant. Platelets 23(2):106–115
Enriquez LJ, Shore-Lesserson L (2009) Point-of-care coagulation testing and transfusion algo-
rithms. Br J Anaesth 103(Suppl 1):i14–i22
Fattorutto M, Pradier O et al (2003) Does the platelet function analyser (PFA-100) predict blood
loss after cardiopulmonary bypass? Br J Anaesth 90(5):692–693
Favaloro EJ (2001) Utility of the PFA-100 for assessing bleeding disorders and monitoring ther-
apy: a review of analytical variables, benefits and limitations. Haemophilia 7(2):170–179
Ferraris VA, Saha SP et al (2012) 2012 update to the Society of Thoracic Surgeons guideline on
use of antiplatelet drugs in patients having cardiac and noncardiac operations. Ann Thorac Surg
94(5):1761–1781
Forestier F, Coiffic A et al (2002) Platelet function point-of-care tests in post-bypass cardiac sur-
gery: are they relevant? Br J Anaesth 89(5):715–721
George JN (2000) Platelets. Lancet 355(9214):1531–1539
Gurbel PA, Bliden KP et al (2010) Adenosine diphosphate-induced platelet-fibrin clot strength: a
new thrombelastographic indicator of long-term poststenting ischemic events. Am Heart J
160(2):346–354
Harrison P (2005) Platelet function analysis. Blood Rev 19(2):111–123
Hobson AR, Petley GW et al (2007) A novel fifteen minute test for assessment of individual time-
dependent clotting responses to aspirin and clopidogrel using modified thrombelastography.
Platelets 18(7):497–505
Jennings LK (2009) Mechanisms of platelet activation: need for new strategies to protect against
platelet-mediated atherothrombosis. Thromb Haemost 102(2):248–257
Karger R, Donner-Banzhoff N et al (2007) Diagnostic performance of the platelet function ana-
lyzer (PFA-100) for the detection of disorders of primary haemostasis in patients with a bleed-
ing history-a systematic review and meta-analysis. Platelets 18(4):249–260
Karger R, Reuter K et al (2012) The Platelet Function Analyzer (PFA-100) as a screening tool in
neurosurgery. ISRN Hematol 2012:839242
Kehrel BE, Brodde MF (2013) State of the art in platelet function testing. Transfus Med Hemother
40(2):73–86
Koscielny J, Ziemer S et al (2004) A practical concept for preoperative identification of patients
with impaired primary hemostasis. Clin Appl Thromb Hemost 10(3):195–204
Kozek-Langenecker SA, Afshari A et al (2013) Management of severe perioperative bleeding:
guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 30(6):270–382
Kwak YL, Kim JC et al (2010) Clopidogrel responsiveness regardless of the discontinuation date
predicts increased blood loss and transfusion requirement after off-pump coronary artery
bypass graft surgery. J Am Coll Cardiol 56(24):1994–2002
62 G. Casso et al.
Tantry US, Bliden KP et al (2005) Overestimation of platelet aspirin resistance detection by throm-
belastograph platelet mapping and validation by conventional aggregometry using arachidonic
acid stimulation. J Am Coll Cardiol 46(9):1705–1709
Tantry US, Bonello L et al (2013) Consensus and update on the definition of on-treatment platelet
reactivity to adenosine diphosphate associated with ischemia and bleeding. J Am Coll Cardiol
62(24):2261–2273
Toth O, Calatzis A et al (2006) Multiple electrode aggregometry: a new device to measure platelet
aggregation in whole blood. Thromb Haemost 96(6):781–788
Tsantes A, Ikonomidis I et al (2012) Evaluation of the role of the new INNOVANCE PFA P2Y test
cartridge in detection of clopidogrel resistance. Platelets 23(6):481–489
van Werkum JW, Kleibeuker M et al (2010) A comparison between the Plateletworks-assay and
light transmittance aggregometry for monitoring the inhibitory effects of clopidogrel. Int J
Cardiol 140(1):123–126
Velik-Salchner C, Maier S et al (2009) An assessment of cardiopulmonary bypass-induced changes
in platelet function using whole blood and classical light transmission aggregometry: the
results of a pilot study. Anesth Analg 108(6):1747–1754
Versteeg HH, Heemskerk JW et al (2013) New fundamentals in hemostasis. Physiol Rev 93(1):
327–358
Voisin S, Bongard V et al (2011) Are P2Y12 reaction unit (PRU) and % inhibition index equivalent
for the expression of P2Y12 inhibition by the VerifyNow assay? Role of haematocrit and hae-
moglobin levels. Thromb Haemost 106(2):227–229
von Pape KW, Dzijan-Horn M et al (2007) Control of aspirin effect in chronic cardiovascular
patients using two whole blood platelet function assays. PFA-100 and Multiplate.
Hamostaseologie 27(3):155–160; quiz 161–162
Wahba A, Sander S et al (1998) Are in-vitro platelet function tests useful in predicting blood loss
following open heart surgery? Thorac Cardiovasc Surg 46(4):228–231
Weber CF, Dietrich W et al (2010) A point-of-care assessment of the effects of desmopressin on
impaired platelet function using multiple electrode whole-blood aggregometry in patients after
cardiac surgery. Anesth Analg 110(3):702–707
Weber CF, Gorlinger K et al (2012) Point-of-care testing: a prospective, randomized clinical trial
of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 117(3):531–547
Hemostasis Assessment
and Evaluation 5
Christoph Sucker and Rainer B. Zotz
Prothrombin time (PT) and activated partial thromboplastin time (aPTT) are the
most frequently used assays for the routine assessment of plasmatic hemostasis.
These widely available assays are employed to detect coagulation factor deficien-
cies, particularly those predisposing to perioperative bleeding. With regard to the
classic cascade or waterfall model of plasmatic hemostasis, PT detects factor defi-
ciencies localized in the extrinsic coagulation pathway and the final common path-
way, whereas aPTT is sensitive to defects in the intrinsic coagulation pathway and
the common pathway. Another assay, thrombin time (TT), detects an insufficient
conversion of fibrinogen to fibrin upon stimulation by thrombin. This assay is used
to diagnose an absence, deficiency, or dysfunction of fibrinogen but also factors
inhibiting or disturbing the action of thrombin on fibrinogen, such as heparin or
thrombin inhibitors. The effect of different coagulation defects on these screening
assays is reflected by the cascade or waterfall model of hemostasis (see Sect. 1.3).
The causes of prolonged PT and aPTT, whether or not they predispose to bleed-
ing, are summarized in Table 5.1. Notably, combinations of different defects of
plasmatic hemostasis may not only yield abnormal results in one test but may influ-
ence all screening assays. Since all the assays mentioned above are based on the
measurement of plasma turbidity for the detection of fibrin production, turbid
patient plasma, such as in severe hyperlipidemia, may interfere with them
significantly.
C. Sucker
LaboMed Gerinnungszentrum Berlin, Tauentzienstrasse 7 b/c, Berlin 10789, Germany
e-mail: [email protected]
R.B. Zotz (*)
Centrum für Blutgerinnungsstörungen und Transfusionsmedizin (CBT),
Immermannstrasse 65 a, Düsseldorf 40210, Germany
e-mail: [email protected]
Table 5.1 Routine coagulation assays distinguishing defects related or not related to bleeding
Defect related Defect not related
PT aPTT TT to bleeding to bleeding
Normal Normal Normal Factor XIII –
deficiency
Von Willebrand
disease (mild)
Prolonged Normal Normal Factor VII –
deficiency
Vitamin K
antagonist/
deficiency
Liver disease
Normal Prolonged Normal Factor VIII Lupus
deficiency anticoagulant
(hemophilia A) Factor XII
deficiency
Factor IX deficiency HMWK deficiency
(hemophilia B)
Factor XI deficiency (Pre)kallikrein
Von Willebrand deficiency
disease
Acquired
hemophilia
Prolonged Prolonged Normal Factor II deficiency ?
Factor V deficiency
Factor X deficiency
Combined
deficiencies
Normal/ Prolonged Prolonged Treatment with –
prolonged heparin, hirudin,
argatroban
Normal/ Normal Prolonged Afibrinogenemia Thrombotic
prolonged prolonged Hypofibrinogenemia dysfibrinogenemia
Hemorrhagic
dysfibrinogenemia
Heparin-like defects
Inhibitors of fibrin
polymerization
Treatment with
heparin, hirudin,
argatroban
Bleeding with normal screening assay and platelet count results can be caused by mild von
Willebrand disease, factor XIII deficiency, platelet defects or medication, low molecular weight
heparin (LMWH), hypothermia, acidosis, and hypocalcemia. Low platelet counts with normal
screening assays are found in cases of pseudothrombocytopenia, idiopathic thrombocytopenic pur-
pura (ITP), and hereditary platelet disorders (e.g., Bernard–Soulier syndrome, gray platelet syn-
drome). Low platelet counts with prolonged aPTT/PT are found in cases of disseminated
intravascular coagulation (DIC), hemodilution, and liver disease
5 Hemostasis Assessment and Evaluation 67
Table 5.2 Patient history for the preoperative exclusion of hemostatic defects associated with
bleeding
Patient history Important aspects
Bleeding symptoms Provoked bleeding: previous bleeding related to
surgery, dentistry, or trauma
Spontaneous bleeding: epistaxis, hematoma,
gingival bleeding, heavy menstrual bleeding, joint
and muscle bleeding
Diseases/conditions potentially Hepatic disease
associated with bleeding disorders Renal disease
Hematological disorders
Myelodysplastic disorder
Myeloproliferative disorder
Paraproteinemia
Cardiopulmonary bypass
Extracorporeal membrane oxygenation
Medication associated with Platelet inhibitors
bleeding disorders Aspirin, clopidogrel, prasugrel, ticagrelor,
dipyridamole
GpIIb/IIIa receptor inhibitors
Analgesics/antiphlogistics (ibuprofen,
diclofenac, etc.)
Cilostazol
Selective serotonin reuptake inhibitors (SSRI)
Beta-lactam antibiotics
Herbal medicine (e.g., ginkgo, garlic, ginger,
ginseng)
Oral anticoagulants
Cumarines, dabigatran, rivaroxaban, apixaban
Parenteral anticoagulants
Heparins, fondaparinux, hirudin, argatroban
Family history Increased bleeding tendency or defined
hemostatic defects in relatives
70 C. Sucker and R.B. Zotz
Patients, history
Suspicious Normal
Normal
References
Hayward CP, Harrison P, Cattaneo M et al (2006) Platelet function analyzer (PFA)-100 closure
time in the evaluation of platelet disorders and platelet function. J Thromb Haemost
4:312–319
Koscielny J, Ziemer S, Radtke H et al (2004) A practical concept for preoperative identification of
patients with impaired primary hemostasis. Clin Appl Thromb Hemost 10:195–204
Peterson P, Hayes TE, Arkin CF et al (1998) The preoperative bleeding time test lacks clinical
benefit: College of American Pathologists’ and American Society of Clinical Pathologists’
position article. Arch Surg 133:134–139
Podda G, Femia EA, Pugliano M et al (2012) Congenital defects of platelet function. Platelets
23:552–563
Shah U, Ma AD (2007) Tests of platelet function. Curr Opin Hematol 14:432–437
Congenital Bleeding Disorders
6
Maria Martinez, Lukas Graf, and Dimitrios A. Tsakiris
6.1 Introduction
Congenital bleeding disorders are found in all the world’s racial groups. They can
affect the plasmatic coagulation cascade, as well as platelet function, with different
patterns of inheritance (sex-linked recessive, autosomal recessive, and autosomal
dominant). Taken all together, they affect roughly 1 in 200 people. Hemophilia
A and B are inherited in a sex-linked recessive way, and together with the more
frequent von Willebrand disease, they constitute the largest group of inherited plas-
matic bleeding disorders (Table 6.1).
In order to avoid catastrophic outcomes, the identification of a congenital bleeding
disorder is crucial for the optimization of hemostasis in a perioperative setting.
Useful diagnostic tools include the global tests of coagulation – as first-choice labo-
ratory tests – prothrombin time (PT/INR) and activated partial thromboplastin time
(aPTT). There are also platelet function screening tests, such as the thrombocyte
global test PFA-100 for initial hemostasis or whole blood impedance aggregometry.
Figure 6.1 gives an overview of the diagnostic efficacy of these tests.
In general, treatment of hereditary deficiencies of coagulation proteins consists
of an appropriate replacement therapy under the supervision of an experienced
hemostaseologist. Dosing and frequency of substitution are dependent on the clini-
cal problem, risk of bleeding, and pharmacokinetics of the factor concentrates used.
Elimination half-lives and target levels required for an adequate hemostatic effect
under physiological conditions are shown in Table 6.2.
D-Dimers
FIBRIN
Table 6.2 Elimination half-life (t ½) of various coagulation factors and their minimum concentra-
tions required in plasma for an adequate hemostatic effect under physiological conditions
Factor Name t ½ (h) Hemostatic level Preoperative target level
I Fibrinogen 90 0.5–1 g/l >1 g/l
II Prothrombin 70 20–40 % >50 %
V 24 15–20 % >20 %
VII 6 15 % >50 %
VIII 12 5–10 % >50–100 %
IX 24 5–10 % >50–100 %
X 40 15–20 % >50 %
XI 60 15–20 % >20 %
XII 52 <1 %
XIII 72 2–5 % >50–100 %
vWF 12 40 % >50–100
6 Congenital Bleeding Disorders 73
range, without clinical bleeding. Therefore, an adequate personal and family bleed-
ing history and an adequate laboratory workup, including platelet function testing,
are required in such cases.
Laboratory Findings
Whereas PT/INR is normal, aPTT can be slightly increased in more severe forms of
vWD type 1, with low FVIII levels. VWF antigen and vWF activity are decreased
in parallel (Table 6.4).
Laboratory Findings
PT, aPTT, and FVIII levels show the same patterns as in vWD type 1. VWF:Ag is
typically normal or slightly low, while vWF activity is clearly lower, which leads to
a ratio vWF:RCo/vWF:Ag below 0.7. Analysis of multimers reveals a lack of large
vWF multimers (Table 6.4).
Laboratory Findings
Laboratory assessment of PT/INR, aPTT, FVIII, vWF activity, and antigen is simi-
lar to vWD type 2A. Additionally, there is a typical multimeric pattern with a lack
of large- and middle-sized multimers. Typically, using ristocetin in lower than nor-
mal concentrations causes these patients’ platelets to aggregate. This finding distin-
guishes type 2B from 2A (Table 6.4).
Laboratory Findings
VWF:Ag is normal, FVIII is normal or only slightly low, vWF activity is low, and
large vWF multimers are normal (Table 6.4).
Laboratory Findings
Findings are normal or prolonged aPTT, reduced FVIII activity, and normal or only
slightly decreased vWF antigen and activity (Table 6.4). Differentiating type 2N
from hemophilia A is only possible by measuring the FVIII binding capacity of
vWF (low in vWD 2N, normal in hemophilia A).
Laboratory Findings
Findings are normal PT/INR, prolonged aPTT due to the low FVIII levels, very low
or undetectable levels of vWF antigen and activity, and absence of the whole spec-
trum of vWF multimers in the multimeric analysis (Table 6.4).
76 M. Martinez et al.
Treatment of vWD
The mainstay for the treatment of vWD in situations of acute bleeding is substitu-
tion with a concentrate containing vWF (Haemate®, Wilate®, Wilfact®). Dosing is
usually weight adjusted. Elimination half-life of substituted vWF is 14–17 h; thus,
in most cases once or twice daily substitution is appropriate. Fresh frozen plasma
(FFP) is an insufficient source of vWF and should not be used for this purpose.
Cryoprecipitate preparations contain high concentrations of vWF, but are no longer
used because they are not pathogen inactivated.
Depending on the severity of the deficiency and the extent of the trauma, desmo-
pressin (DDAVP) (Octostim®, Minirin®) can be used as an alternative means to
increase endogenous vWF and FVIII levels by stimulating the release of vWF from
the endothelium. Desmopressin is given intravenously as a short infusion over 30 min
(0.3 μcg/kg body weight). Alternatively, it can be given subcutaneously, repeating
once or twice every 24 h. If active mucocutaneous bleeding is present, treatment
should include an antifibrinolytic agent (e.g., tranexamic acid 10 mg/kg bw IV)
because desmopressin also stimulates endogenous fibrinolysis by release of tissue
plasminogen activator. Intranasal application of desmopressin might be an option for
self-treatment in case of bleeding, but due to its variable bioavailability, it is not pre-
ferred in the perioperative setting. Desmopressin should be used cautiously in vWD
type 2B because it shows low efficacy and might aggravate thrombocytopenia: infu-
sion time can be prolonged to 60 min. Desmopressin has no effect in type 3 patients.
VWD type 3 is treated by substitution with a vWF/FVIII concentrate. The FVIII
component is essential in achieving adequate hemostasis at the beginning of the
treatment. High purity vWF concentrates with very low levels of FVIII, such as
Wilfact®, must initially be given together with an FVIII concentrate because it takes
hours for the endogenous FVIII level to rise after substitution with pure vWF.
Secondary prophylaxis with a vWF concentrate (10–20 IU/kg bw, two to three
times a week) is standard care in type 3 patients with recurrent bleeding
complications.
Laboratory Findings
Afibrinogenemia is characterized by prolonged aPTT, PT, and thrombin clotting
times. No detectable fibrinogen can be found. Hypofibrinogenemia has mostly nor-
mal global tests and variably decreased levels of functional fibrinogen.
Dysfibrinogenemia has decreased functional fibrinogen, but normal protein values
in the immunological assays.
varies in different countries: it ranges from 0.7 to 1.3 in 10,000 people. FIX defi-
ciencies, or hemophilia B, are rarer, with a prevalence of 1 in 50,000 people.
Factor VIII or IX levels in hemophilia are found to be below 50 % and are clas-
sified in three severity groups (severe, moderate, and mild). Classification is based
on the factor concentration, reflecting the clinical phenotype (Table 6.5). Mild
hemophilia often only first presents as a bleeding episode after trauma or surgery.
Severe hemophilia often presents as a bleeding tendency when a child starts to walk
or sometimes days after birth. The severity of bleeding correlates very well with the
factor level measured. Spontaneous muscle and joint bleeding (specially the ankles,
knees, and elbows) is typical for the severe form of hemophilia. A tentative explana-
tion for joint bleeding in hemophilia is the fact that soft tissues in the joints express
very low levels of tissue factor, a protein which is crucial in the initiation of the
coagulation cascade.
Laboratory Findings
Hemophilia A has normal PT/INR, prolonged aPTT, reduced FVIII activity in the
specific assay, and normal bleeding time.
Treatment of Hemophilia A
Substitution with the missing protein is the modern mainstay of hemophilia A treat-
ment. Primary or secondary prophylaxis with regular substitution, two to four times
a week, is a common mode of application. Prophylaxis begins in the first year of life
and continues throughout adulthood. Regular prophylaxis has been shown to protect
from chronic joint arthropathy and acute bleeding. In case of invasive procedures,
surgery, or trauma, a more intensive substitution scheme is chosen. Table 6.6 gives
dosing and target levels of FVIII. About 15–30 % of severe hemophiliacs develop
acquired alloantibodies against FVIII after repeated substitution with FVIII concen-
trates. Special treatment protocols based on immunotolerance induction or immu-
nosuppression are then needed for eradication of the inhibitor.
Hemophilia B, the hereditary FIX deficiency, is the second most common form of
hemophilia. FIX is a vitamin K-dependent serine protease synthesized in the liver.
It is activated by FVIIa or FXIa and in its activated form catalyzes the conversion of
factor X to Xa.
6 Congenital Bleeding Disorders 79
Laboratory Findings
Hemophilia B has normal PT/INR, prolonged aPTT, reduced FIX activity in the
specific assay, and normal bleeding time.
Treatment of Hemophilia B
Substitution with FIX concentrates in episodes of bleeding or suspected bleeding is
the mainstay of treatment for hemophilia B (Table 6.7). In severe forms, primary or
secondary prophylaxis with substitution is recommended two to three times a week.
Because FIX has a longer half-life than FVIII, it can be given at longer intervals.
After repeated substitution with concentrates, about 3–5 % of severe hemophilia B
patients develop acquired alloantibodies against FIX. Special treatment protocols
based on immunotolerance induction or immunosuppression are then needed to
eradicate the inhibitor.
80 M. Martinez et al.
Laboratory Findings
Factor II deficiency exhibits prolonged aPTT and PT/INR, normal bleeding time,
and reduced FII levels in the specific assay.
Treatment
Treatment consists of on demand substitution with FII concentrates. These are
available in the form of four-factor prothrombin complex concentrates (4fPCC,
Prothromplex®, Beriplex®, Octaplex®), containing FIX, FX, and FVII in addition to
FII. If these are not available, FFP can also be used as a source of FII, if appropri-
ately dosed (10 ml/kg BW).
Laboratory Findings
Factor V deficiency exhibits prolonged PT/INR and aPTT, normal bleeding time,
and decreased FV activity in the specific assay.
Treatment
Treatment consists in substituting FV on demand. As there is no FV concentrate avail-
able, FFP is the only source. FV is an unstable protein even under proper storage condi-
tions. Retention values of FFP after thawing give no more than 60 % of FV activity
with respect to the original material before deep freezing. Alternatively, recombinant
activated factor VII (rFVIIa, NovoSeven®) could be used off-label for hemostasis.
The incidence of this defect is about one in two million people. The genetic defect
which causes this combined factor deficiency affects the transport system between
82 M. Martinez et al.
Laboratory Findings
PT and aPTT are prolonged. Isolated reduction in factor V and VIII activities is
found in the specific assay.
Treatment
Depending on the severity of the deficit, FVIII concentrates and FFP can be used. In
mild bleeding, antifibrinolytics may also be an alternative.
The congenital FVII deficiency is the most frequent of the rare bleeding disorders
(30 %). The prevalence of its severe form (FVII <2 %) is estimated to be about 1 in
500,000 people. The clinical phenotype presents a wide spectrum of bleeding from
mucosal to intracranial hemorrhages. The minimum FVII level required for an
adequate hemostatic effect is 10–15 %.
Laboratory Findings
Factor VII deficiency exhibits prolonged PT/INR, normal aPTT, and low FVII
activity levels in the specific assays.
Treatment
Factor VII concentrates can be used for substitution in cases of bleeding (high purity
FVII, recombinant FVIIa, or 4fPCC). Preferably nonactivated forms of concentrates
should be used. In heterozygous deficiencies, no substitution is normally required
even in case of surgery, since levels of FVII in these cases range from 20 to 50 %.
Caution is required in cases of potential vitamin K deficiency, where FVII can soon
reach inappropriately low levels. Although its elimination half-life of 5 h is short, a
less frequent substitution scheme turns out to be adequate even in case of surgery.
In severe cases, especially in children, prophylactic substitution can be given
three times a week.
In this combined deficiency, the defect is situated in the enzyme responsible for the
gamma-glutamyl carboxylation of the coagulation factors. Vitamin K acts as cofac-
tor for gamma-glutamyl carboxylase. Carboxylation takes place in the liver and
enables specific proteins to bind calcium. This step is essential for the coagulation
enzymes to anchor on the coagulation-active phospholipids of platelets and cell
membranes. Affected patients not only have low levels of procoagulant factors II,
6 Congenital Bleeding Disorders 83
VII, IX, and X but also of the natural coagulation inhibitors, proteins C and S.
Clinically, this bleeding disorder manifests itself early in life, with umbilical cord
bleeding and central nervous system hemorrhages.
Laboratory Findings
This combined deficiency exhibits prolonged PT/INR and aPTT and low levels of
FII, FVII, FIX, FX, and proteins C and S.
Treatment
Treatment consists in on demand substitution of the missing coagulation proteins
using a concentrate. Four-factor PCC products are used for this purpose
(Prothromplex®, Beriplex®, Octaplex®). High doses of vitamin K in adults (1 × 10–15
mg daily) orally or intravenously might also lead to a partial increase of the factor
levels and prevent major bleeding. If not otherwise possible, fresh frozen plasma
(FFP) can be used as a source of FII, FVII, FIX, and FX. Depending on the severity
of the clinical phenotype, a secondary chronic prophylaxis with high-dose vitamin
K or four-factor PCC might be considered.
Laboratory Findings
Prolonged PT/INR and aPTT and low factor X activity.
Treatment
Treatment consists in FX substitution using a concentrate. There are no high-purity
FX concentrates available. Four-factor PCC products are used for this purpose
(Prothromplex®, Beriplex®, Octaplex®). Alternatively, a combined FIX/FX product
can be used (Factor IX/X Behring®). If these are not available, FFP can be used as a
source of FX. Treatment can be combined with antifibrinolytics.
circulation. Thrombin activates FXI. The activated FXIa further catalyzes the acti-
vation of FIX.
The majority of people affected by FXI deficiency have a low but detectable FXI
level. The severity of bleeding, however, does not correlate with the factor levels
measured. Bleeding manifests itself as mucosal or surgical bleeding. Joint or mus-
cle bleeding is not common at all. FXI deficiency is the second most common bleed-
ing disorder in women after vWD.
Laboratory Findings
FXI deficiency exhibits prolonged aPTT, normal PT/INR, and low FXI levels in the
specific assays.
Treatment
Treatment consists of substitution with FXI concentrate (Hemoleven®). Dosing
should be calculated cautiously, because overdosing or even correction to high nor-
mal levels can cause thrombotic complications. FFP may be used as an alternative
source for FXI. Treatment may be combined with antifibrinolytics.
Congenital deficiency of FXII is not associated with bleeding, so that it is not con-
sidered a hemorrhagic disorder. On the contrary, there is suggestive evidence that it
might cause a tendency to thrombosis, since FXII is involved in the activation of the
complement pathway and of fibrinolysis.
Laboratory Findings
Factor XII deficiency exhibits normal PT/INR, prolonged aPTT, and reduced levels
of FXII activity in the specific assays.
Treatment
There is currently no recognized indication for FXII substitution.
Laboratory Findings
FXIII deficiency exhibits normal PT/INR and activated aPTT and low levels of
FXIII activity and antigen in the specific assays.
Treatment
Treatment consists of FXIII substitution on demand using a FXIII concentrate
(Fibrogammin®). A single dose of 1,250 IE is usually sufficient for a long-lasting
effect since the protein’s elimination half-life is long (72 h). In severe cases, regular
secondary prophylaxis may be required every 4–6 weeks.
Laboratory Findings
Glanzmann’s thrombasthenia exhibits normal PT/INR and aPTT, but prolonged BT.
In platelet aggregation assays, it shows a specific pattern in the responsiveness of
platelets to various agonists. Using flow cytometry, it shows a loss or severe reduc-
tion of GP IIb/IIIa density on platelets.
Treatment
The most efficient treatment for this disorder is transfusion of normal platelets.
However, this may cause rare transfusion-associated adverse events, such as the gen-
eration of iso- or alloantibodies against GP IIb/IIIa. Although the risk of alloimmuniza-
tion has clearly been reduced since the introduction of leukocyte filtration and
single-donor thrombocytapheresis in transfusion medicine, platelet transfusions should
be avoided if possible. Very often, surgery cannot be performed on these patients with-
out platelet transfusion. In such cases, human leukocyte antigen-compatible products
should be chosen, and the number of transfusions should be kept to a minimum periop-
eratively. Antifibrinolytics combined with rFVIIa (NovoSeven®) are successfully used
in mild or moderate bleeding. Desmopressin might also help in case of mild bleeding.
86 M. Martinez et al.
Laboratory Findings
BSD exhibits normal PT/INR, normal aPTT, prolonged BT, and thrombocytopenia
with large platelet volume (MPV >10 fl). Platelet aggregation assays show a specific
pattern in the responsiveness of platelets to various agonists. Using flow cytometry,
it shows loss or severe reduction of GP Ib/IX density on platelets.
Treatment
The most efficient treatment of BSD is transfusion of normal platelets. However, this
might cause rare transfusion-associated adverse events, such as iso- or alloantibodies
against GPIb/IX. Although the risk of alloimmunization has clearly been reduced
since the introduction of leukocyte filtration and single-donor thrombocytapheresis
in transfusion medicine, platelet transfusions should be avoided if possible. Very
often surgery cannot be performed on these patients without platelet transfusion. In
such cases, human leukocyte antigen-compatible products should be chosen, and the
number of transfusions should be kept to a minimum perioperatively. Antifibrinolytics,
along with rFVIIa (NovoSeven®), are successfully used in mild or moderate bleed-
ing. Desmopressin might also help in case of mild bleeding.
Bibliography
Bornikova L, Peyvandi F, Allen G, Bernstein J, Manco-Johnson MJ (2011) Fibrinogen replace-
ment therapy for congenital fibrinogen deficiency. J Thromb Haemost 9(9):1687–1704
Gomez K, Bolton-Maggs P (2008) Factor XI deficiency. Haemophilia 14(6):1183–1189
Hsieh L, Nugent D (2008) Factor XIII deficiency. Haemophilia 14(6):1190–1200
Huang JN, Koerper MA (2008) Factor V deficiency: a concise review. Haemophilia 14(6):
1164–1169
Lee CA, Berntorp EE, Hoots WK (eds) (2005) Textbook of haemophilia. Blackwell Publishing
Ltd, Oxford
Mannucci PM, Duga S, Peyvandi F (2004) Recessively inherited coagulation disorders. Blood
104(5):1243–1252
Mariani G, Bernardi F (2009) Factor VII deficiency. Semin Thromb Hemost 35(4):400–406
6 Congenital Bleeding Disorders 87
7.1 Introduction
7.2 Thrombocytopenia
Thrombotic microangiopathy
Early stage liver disease
Dilution Drugs
Antiphospholipid antibodies
DIC HIT
Vitamin K defect
Hemorrhage ITP
Anticoagulation
Cirrhosis Sepsis
Inflammation
Hypersplenism
Coagulopathy Thrombocytopenia
Fig. 7.1 The critically ill patient. DIC disseminated intravascular coagulation, HIT heparin-
induced thrombocytopenia, ITP immune thrombocytopenia
Acquired multifactorial deficiencies are common and may result from vitamin K
deficiency, liver disease, or DIC.
Qualitative
defects of
fibrinogen
and
platelets
Parallel decrease
Endothelium
of
Platelets Elevated vWF
procoagulant
Coagulation Elevated FVIII
and
Fibrinolysis
anticoagulant
factors
Classic
comorbidities
and
complications
of CLD
Fig. 7.2 Hemostasis in chronic liver disease (CLD). vWF von Willebrand factor, FVIII factor VIII
7.4.2.1 Management
The correction of a coagulopathy in a non-bleeding patient is not required (Roberts
et al. 2010; Gines et al. 2012). Conservative measures such as vitamin K substitu-
tion are recommended for the optimization of hemostasis in CLD. Predicting bleed-
ing complications in CLD patients is challenging. The clinician should base her/his
evaluation on three indicators:
1. Patient
The majority of hemorrhages are due to the rupture of portosystemic varices
(Lisman and Leebeek 2007). Bacterial infections, vitamin K deficiency (low
dietary intake, loss of intestinal bacterial flora after antibiotherapy, intra-/extra-
hepatic cholestasis), and renal failure increase the tendency to bleed.
96 S. Barelli et al.
2. Laboratory values
Current routine coagulation tests offer only limited help in the proper evalua-
tion of the state of hemostatic balance in CLD (Tripodi and Mannucci 2011). For
example, a prolonged prothrombin time (PT) does not necessarily indicate that
the CLD patient is naturally anticoagulated, and a borderline PT could underes-
timate the risk of bleeding. Emerging global coagulation tests better reflect the
interplay of all the factors involved in hemostasis (thrombin generation tests,
thromboelastography) (Roberts et al. 2010).
3. Type of bleeding or surgery
Acute variceal bleeding is mainly the consequence of anatomical changes
and depends on the Child-Pugh score and endoscopic features. It is best treated
with fluid resuscitation, pharmacological treatment, and endoscopy. For elec-
tive surgical procedures (paracentesis, central venous catheter insertion, or
liver biopsy), the potential need for transfusion must anticipated. Transfusions
should target a hemoglobin level of >70–80 g/l and platelet counts >50 G/L.
Prophylactic transfusion of fresh frozen plasma (FFP) is associated with a risk
of volume overload, infection, and transfusion-related acute lung injury
(TRALI). This makes the use of prothrombin complex concentrates (PCC)
more appropriate. There is no advantage to using recombinant FVIIa (Bosch
et al. 2008).
7.4.3.1 Diagnosis
DIC is not a disease in itself, but a syndrome (Levi et al. 2002). Several clinical
conditions mimic the hemostatic pattern of DIC. Overall, DIC should be suspected
when a patient presents with:
• An underlying medical or surgical condition compatible with DIC
• Multiple organ failure
• Bleeding and/or thrombotic diathesis
• Thrombocytopenia associated with multiple coagulation factor deficiencies and
hyperfibrinolysis (low fibrinogen, high D-Dimers, fibrin degradation products,
and fibrin monomer levels).
We recommend using the scoring system of the International Society on
Thrombosis and Haemostasis (ISTH) to easily identify overt DIC (Taylor et al.
2001) (Table 7.3).
7.4.3.2 Management
Treatment options should target the cause of DIC and aim to improve the patient’s
condition rather than the laboratory values (Levi et al. 2009; Thachil and Toh 2012;
Wada et al. 2013).
Transfusion of blood components is indicated ONLY during active bleeding or in
periprocedural situations. Target blood component levels are:
• Platelets >50 G/L in case of active bleeding and >20 G/L in patients with a high
risk of bleeding. Platelet concentrates can be administered.
• PT or aPTT <1.5 times the upper limit of the normal range. FFP should be privi-
leged. PCC may be considered in actively bleeding patients if FFP transfusion is
impossible.
• Fibrinogen >1.5 g/l. The administration of fibrinogen concentrate or cryoprecipi-
tate may be recommended in actively bleeding patients with fibrinogen levels
persistently <1.5 g/l despite FFP replacement.
7.4.3.3 Conclusion
Critically ill patients are at risk of developing DIC, and thus this diagnosis should
be assessed as early as possible.
Platelet count and simple coagulation tests (PT, fibrinogen level, or D-dimers)
are of great value in identifying overt DIC and following its process.
Inhibitors are autologous antibodies to coagulation factors that can develop in dif-
ferent clinical circumstances, such as acquired hemophilia A in the peripartum or in
the context of lymphoproliferative neoplasms (alloantibodies only develop in the
context of factor substitution for hemophilia A or B). In theory, it is possible to
develop an inhibitor to any given coagulation factor; however, inhibitors to FVIII
and vWF (see Sect. 7.6) are the most frequent. Patients with acquired FVIII defi-
ciency (also called “acquired hemophilia A”) present a broad spectrum of clinical
signs ranging from superficial bruising to life-threatening bleeding. The incidence
of acquired FVIII deficiency is 1.4/million/year (Collins 2012). The main treatment
consists in treating the underlying clinical problem, if it can be clearly identified, for
example, as a lymphoproliferative syndrome. If this is not possible, in most of the
cases, the hematologist will initiate an immunosuppressive treatment with cortico-
steroids, monoclonal anti-CD20 antibodies (rituximab), or cyclophosphamide, or a
combination of those therapies. However, no treatment will correct the targeted
coagulation factor immediately. Should a surgical procedure be necessary, adequate
hemostasis must be reached using a treatment that bypasses the targeted coagulation
factor. Invasive procedures should thus be avoided when possible. One drug used to
overcome the inhibitors of coagulation factors is rFVIIa (NovoSeven®). It bypasses
factors VIII and IX and directly activates factor X, which then activates prothrom-
bin to thrombin. Another possible bypassing agent is factor VIII inhibitor bypassing
activity (FEIBA®). Both agents seem to have about the same effect on hemostasis
(Baudo et al. 2012). Standardized laboratory assays cannot monitor responses to
either of these agents. The performances of thrombelastography and thrombin gen-
eration, as well as the interpretation of aPTT graphs, are promising indicators, but
are not yet standardized (Pivalizza and Escobar 2008). If neither hemostatic agent
succeeds in stopping the hemorrhagic process, one option for rapid inhibitor
7 Acquired Hemostatic Disorders 99
Acquired von Willebrand syndrome (AvWS) is a rare disorder, but probably more
frequent than we might expect. It is normally associated to an underlying disorder,
such as lymphoproliferative or myeloproliferative neoplasms, other malignancies,
100 S. Barelli et al.
aortic stenosis (also when part of the Heyde syndrome), or, more rarely, autoim-
mune diseases. In these conditions vWF is eliminated more rapidly, either due to
autoantibodies or to absorption on the surface of platelets or malignant cells. Infused
vWF and FVIII concentrates will also rapidly be eliminated, if the underlying con-
dition has not been treated. The same holds true when desmopressin is administered
to increase endogenous vWF and FVIII. Table 7.4 summarizes treatment option for
AvWS. Anecdotal reports mention the efficacy of plasma exchanges with 3–4 l/day
of FFP for several days (Bovill et al. 1986; Silberstein et al. 1987; Federici et al.
2000; Maddox et al. 2005). This method can be useful, but it is not feasible for
emergency procedures. It is recommended that emergency patients receive immu-
noglobulins intravenously (2 g/kg, if possible divided into 2 doses, infused 24 h
apart). However, this treatment is ineffective in patients with AvWS in a monoclonal
IgM setting (Federici et al. 2000). Factor improvement occurs 12–72 h after treat-
ment and can last from days to weeks (Tiede et al. 2011). In cases showing no factor
correction or cases of emergency surgery, 30–100 IU/kg of Haemate P® can be
infused before surgery and 20–30 IU/kg two hours into surgery. In the postoperative
setting, 20–30 IU/kg of Haemate P® 3 times daily for 5 days seems to be an ade-
quate treatment (Maddox et al. 2005; Tiede et al. 2011). If the response to Haemate
P® is diminishing, a further dose of 1 g/kg of immunoglobulins can be given intra-
venously. rFVIIa is another possibility to control bleeding, administered at the usual
dosage of 90 μg/kg. However, there is currently only a limited experience of this
treatment. Antifibrinolytic drugs, such as tranexamic acid, can be used in surgery on
sites with a high fibrinolytic activity, such as the gastrointestinal tract or the oral
cavity, but they are contraindicated in patients with macrohematuria.
During surgery, normal factor levels should be aimed for; in the postoperative
setting, plasma levels of at least 50 % seem to be sufficient (Frank et al. 2002).
See Chap. 8
7.8 Malignancies
splanchnic and cerebral vein thrombosis. Some patients may also present superficial
vein thrombosis. Venous malignancy per se is a risk factor for DVT (Prandoni et al.
2005). About 10 % of patients with idiopathic DVT have an underlying malignancy.
Arterial events comprise stroke, myocardial infarction, and arterial embolism.
The activation of coagulation in patients with a malignancy is caused by the
acute-phase reaction, anticancer treatments, and prothrombotic properties of tumor
cells. The latter induce tissue factor, protease-activating factor X, plasminogen acti-
vator inhibitors which lead to impaired fibrinolysis and cytokines targeting the
endothelium, leukocytes, and platelets. The whole process is influenced by the stage
of the disease, invasive procedures (e.g., central venous catheter), and the patient’s
profile (inherent thrombophilia).
7.8.1 Diagnosis
7.8.2 Management
For patients with a high risk of VTE who are undergoing abdominal or pelvic
surgery for cancer, but who do not otherwise exhibit a high risk for major bleeding
complications, extended pharmacological prophylaxis with LMWH is recom-
mended (4 weeks).
In patients with DVT of the leg and/or a PE and active cancer, treatment with
LMWH is recommended over VKA therapy. In patients with DVT and cancer who
are not treated with LMWH, VKA is suggested over dabigatran or rivaroxaban as a
long-term therapy.
In patients with acute DVT and/or PE and a contraindication to anticoagulation
(hemorrhage, surgery, or thrombocytopenia), the use of an inferior vena cava filter
is recommended (see also Chap. 8). A conventional course of anticoagulant therapy
should be considered as soon as the risk of bleeding resolves.
In cancer patients who have upper extremity DVT that is associated with a cen-
tral venous catheter, 3 months of anticoagulation treatment is recommended if the
catheter is removed. If the catheter is not removed, anticoagulation treatment should
be continued as long as the central venous catheter remains.
7.9.1 Mechanism
7.9.2 Diagnosis
• Essential thrombocytosis
• Severe aortic stenosis (Heyde syndrome triad)
Polycythemia vera (PV) and essential thrombocythemia (ET) are two types of MPS
that are associated with up to 50 % of thrombotic events (Elliott and Tefferi 2005),
with arterial thrombotic events being more common than venous events (Fenaux et al.
1990; De Stefano et al. 2008). The most common events among patients with PV and
ET are ischemic strokes (De Stefano et al. 2008). The prevalence of splanchnic and
cerebral vein thrombosis is unusually high in patients with MPS, and such events
might be the presenting feature of the disease, even before diagnosis (Reikvam and
Tiu 2012). Thrombotic risk is higher due to elevated hematocrit with hyperviscosity,
thrombocytosis, and leukocytosis (Wolanskyj et al. 2006). Janus kinase 2 (JAK2)
mutations can also add to the thrombotic risk through increased red cell adhesiveness
(Buss et al. 1985) and altered platelet signaling (Falanga et al. 2007). Patients with
MPS are treated with aspirin and/or cytoreduction, according to their risk level.
7.10.3 Treatment
References
Abdelmalik N, Ruhe HG et al (2008) Effect of the selective serotonin reuptake inhibitor paroxetine
on platelet function is modified by a SLC6A4 serotonin transporter polymorphism. J Thromb
Haemost 6(12):2168–2174
Ahmad F, Solymoss S et al (1996) Characterization of an acquired IgG inhibitor of coagulation
factor XIII in a patient with systemic lupus erythematosus. Br J Haematol 93(3):700–703
Aird WC, Mark EJ (2002) Case records of the Massachusetts General Hospital. Weekly clinico-
pathological exercises. Case 15–200. A 53-year-old man with a myocardial infarct and throm-
boses after coronary-artery bypass grafting. N Engl J Med 346(20):1562–1570
Ang AL, Kuperan P et al (2009) Acquired factor V inhibitor. A problem-based systematic review.
Thromb Haemost 101(5):852–859
Ataoglu A, Canan F (2009) Mean platelet volume in patients with major depression: effect of
escitalopram treatment. J Clin Psychopharmacol 29(4):368–371
Baudo F, Caimi T et al (2010) Diagnosis and treatment of acquired haemophilia. Haemophilia
16(102):102–106
Baudo F, Collins P et al (2012) Management of bleeding in acquired hemophilia A: results from
the European Acquired Haemophilia (EACH2) Registry. Blood 120(1):39–46
106 S. Barelli et al.
Boehlen F, Casini A et al (2013) Acquired factor XIII deficiency: a therapeutic challenge. Thromb
Haemost 109(3):479–487
Bosch J, Thabut D et al (2008) Recombinant factor VIIa for variceal bleeding in patients with
advanced cirrhosis: a randomized, controlled trial. Hepatology 47(5):1604–1614
Bovill EG, Ershler WB et al (1986) A human myeloma-produced monoclonal protein directed
against the active subpopulation of von Willebrand factor. Am J Clin Pathol 85(1):115–123
Buclin T, Schmidt PM et al (1992) Acquired factor V inhibitor treated with intravenous immuno-
globulins. Schweiz Med Wochenschr 122(51–52):1968–1970
Buss DH, Stuart JJ et al (1985) The Incidence of thrombotic and hemorrhagic disorders in associa-
tion with extreme thrombocytosis: An analysis of 129 cases. Am J Hemato 20:365–372
Cochran KA, Cavallari LH et al (2011) Bleeding incidence with concomitant use of antidepres-
sants and warfarin. Ther Drug Monit 33(4):433–438
Collins PW (2012) Therapeutic challenges in acquired factor VIII deficiency. Hematology Am Soc
Hematol Educ Program 2012:369–374
Dalton SO, Johansen C et al (2003) Use of selective serotonin reuptake inhibitors and risk of upper
gastrointestinal tract bleeding: a population-based cohort study. Arch Intern Med 163(1):59–64
De Stefano V, Za T et al (2008) Recurrent thrombosis in patients with polycythemia vera and essen-
tial thrombocythemia: Incidence, risk factors, and effect of treatments. Haematologica
93:372–380
de Raucourt E, Barbier C et al (2003) High-dose intravenous immunoglobulin treatment in two
patients with acquired factor V inhibitors. Am J Hematol 74(3):187–190
Elliott MA, Tefferi A (2005) Thrombosis and haemorrhage in polycythaemia vera and essential
thrombocythaemia. Br J Haematol128:275–290
Falanga A, Marchetti M et al (2007) V617f Jak-2 Mutation in patients with essential thrombocy-
themia: relation to platelet, granulocyte, and plasma hemostatic and inflammatory molecules.
Exp Hematol 35:702–711
Federici AB, Rand JH et al (2000) Acquired von Willebrand syndrome: data from an international
registry. Thromb Haemost 84(2):345–349
Fenaux P, Simon M et al (1990) Clinical course of essential thrombocythemia in 147 cases. Cancer
66:549–556
Franchini M, Lippi G (2011) Acquired factor V inhibitors: a systematic review. J Thromb
Thrombolysis 31(4):449–457
Franchini M, Lippi G et al (2008) The use of recombinant activated factor vii in platelet-associated
bleeding. Hematology 13:41–45
Frank RD, Kunz D et al (2002) Acquired von Willebrand disease–hemostatic management of
major orthopedic surgery with high-dose immunoglobulin, desmopressin, and continuous fac-
tor concentrate infusion. Am J Hematol 70(1):64–71
Fu YX, Kaufman R et al (1996) Multimodality therapy of an acquired factor V inhibitor. Am J
Hematol 51(4):315–318
Gines P, Fernandez J et al (2012) Management of critically-ill cirrhotic patients. J Hepatol 56(Suppl 1):
S13–S24
Huth-Kuhne A, Baudo F et al (2009) International recommendations on the diagnosis and treat-
ment of patients with acquired hemophilia A. Haematologica 94(4):566–575
Jeimy SB, Krakow EF et al (2008) An acquired factor V inhibitor associated with defective factor
V function, storage and binding to multimerin 1. J Thromb Haemost 6(2):395–397
Jervis K, Senthilnathan V et al (2009) Management of a patient with lupus anticoagulant and
antiphospholipid syndrome for off-pump coronary artery bypass grafting using the Hepcon
system. Anesth Analg 108(4):1116–1119
Landolfi R, Rocca B et al (1995) Bleeding and thrombosis in myeloproliferative disorders:
Mechanisms and treatment. Crit Rev Oncol Hematol 20:203–222
Lebrun A, Leroy-Matheron C et al (2008) Successful treatment with rituximab in a patient with an
acquired factor V inhibitor. Am J Hematol 83(2):163–164
Levi M, Ten Cate H (1999) Disseminated intravascular coagulation. N Engl J Med
341(8):586–592
7 Acquired Hemostatic Disorders 107
Tjonnfjord GE (2004) Activated prothrombin complex concentrate (FEIBA) treatment during sur-
gery in patients with inhibitors to FVIII/IX: the updated Norwegian experience. Haemophilia
10(Suppl 2):41–45
Tribl B, Knobl P et al (1995) Rapid elimination of a high-titer spontaneous factor V antibody by
extracorporeal antibody-based immunoadsorption and immunosuppression. Ann Hematol
71(4):199–203
Tripodi A, Mannucci PM (2011) The coagulopathy of chronic liver disease. N Engl J Med
365(2):147–156
Trouillas P, von Kummer R (2006) Classification and pathogenesis of cerebral hemorrhages after
thrombolysis in ischemic stroke. Stroke 37:556–561
Vanderschueren S, De Weerdt A et al (2000) Thrombocytopenia and prognosis in intensive care.
Crit Care Med 28(6):1871–1876
Wada H, Thachil J et al (2013) Guidance for diagnosis and treatment of DIC from harmonization
of the recommendations from three guidelines. J Thromb Haemost 11(11):2078–2079
Warkentin TE (2011) How I diagnose and manage HIT. Hematology Am Soc Hematol Educ
Program 2011:143–149
William BM (2008) Adjunctive role for recombinant activated factor VII in the treatment of bleed-
ing secondary to a factor V inhibitor. Blood Coagul Fibrinolysis 19(4):327–328
Wolanskyj AP, Schwager SM et al (2006) Essential thrombocythemia beyond the first decade: life
expectancy, long-term complication rates, and prognostic factors. Mayo Clin Proc
81:159–166
Zvetkova E, Antonova N et al (2010) Platelet morphological, functional and rheological properties
attributable to addictions. Clin Hemorheol Microcirc 45(2–4):245–251
Antiplatelet Therapy
and Anticoagulation 8
Pierre-Guy Chassot, Stefano Barelli, Sabine Blum,
Anne Angelillo-Scherrer, and Carlo E. Marcucci
8.1.1 Introduction
TXA2 ADP
Reversible
ADP inhib.:
ticagrelor
cangrelor
α-granule elinogrel
GP IIb/IIIa
GP Ib/V/IX
TXA2
ADP
Reversible
GP IIb/IIIa inhib.:
vWF Fib abciximab
eptifibatide
tirofiban
Fig. 8.1 Classification of the different AP agents. Blockers of von Willebrand factor/adhesion
molecules and thrombin receptor blockers are under investigation (Adapted with permission from
www.pac4.ch). ADP adenosine diphosphate, cAMP cyclic adenosine monophosphate, COX-1
cyclooxygenase-1, Fib fibrinogen, inhib inhibitor, NSAID nonsteroidal anti-inflammatory drug,
TXA2 thromboxane A2, vWF von Willebrand factor
The different AP agents are classified according to the type of receptor they inhibit
on the platelet (Fig. 8.1). Their pharmacology is described in Tables 8.1 and 8.2. At
doses of 50–160 mg/day, aspirin completely inhibits the cyclooxygenase-1 (COX-1)
enzyme which converts arachidonic acid to thromboxane A2 (TXA2), the ligand for
the homonymous platelet receptor. However, 6–10 % of the population shows a low
response to aspirin treatment, resulting in insufficient platelet inhibition. In some
patients, this is due to insufficient inhibition of the COX-1 enzyme by aspirin. Yet
in others, due to a predominance of alternative activation pathways (e.g., ADP,
thrombin), the platelet function remains normal in spite of sufficient COX-1 inhibi-
tion. Competitive interaction with nonsteroidal anti-inflammatory drugs (NSAIDs)
may also reduce aspirin efficiency (Patrono and Rocca 2010).
Clopidogrel (Plavix™, Iscover™) is a prodrug which is oxidized into an active
metabolite in a two-step process by hepatic cytochromes. This metabolite irrevers-
ibly blocks the adenosine diphosphate (ADP) receptor (P2Y12) and reduces platelet
activity by 50–60 % (Hall and Mazer 2011). Clopidogrel’s efficiency may be
8 Antiplatelet Therapy and Anticoagulation 111
20
Haemorrhagic Optimal
risk window
15 Thrombotic
risk
10
Q1 Q2 Q3 Q4
Residual platelet activity (quartiles)
Fig. 8.2 Rate of major adverse cardiac events (MACE, in blue) after percutaneous coronary inter-
vention and stenting according to the residual platelet activity after a loading dose of aspirin and
clopidogrel. Since adverse events cluster in the highest quartile (Q4), there is a larger benefit to
decrease platelet activity from Q4 to 50 % than from 50 % to Q1. The curve for hemorrhagic risk
(in red) is mirrorlike and varies in the opposite direction. These two curves determine an optimal
window with the best combination of minimal risk of bleeding and maximal platelet inhibition
(Adapted with permission from Price (2009))
thrombosis. However, it does increase the risk of spontaneous hemorrhage 1.5 times
and of surgical bleeding up to four times (Wiviott et al. 2007). Considering its
potency, prasugrel cessation 7 days before surgery is recommended.
Ticagrelor (Brilinta™, Brilique™, Possia™) is a powerful and reversible ADP
receptor blocker. One hour after a loading dose, 80 % of platelet activity is inhibited
and after cessation, it takes 3 days for platelet function to recover (Gurbel et al.
2009). Ticagrelor is more efficient than clopidogrel in preventing stent thrombosis,
yet does not increase the hemorrhagic risk (Wallentin et al. 2009). Because some
patients produce a long-acting metabolite, it is recommended to stop ticagrelor 5
days before an operation.
Cangrelor is an intravenous fast onset and offset drug, which will be useful for
preoperative substitution of long-acting agents. It abolishes platelet aggregation but
allows a complete recovery of platelet activity within 1–3 h of stopping the perfu-
sion (Angiolillo et al. 2012).
Dual AP therapy is essential after ACS or stent implantation because vascular
lesions and stents behave like unstable plaques if they are not fully covered by a
cellular layer. It takes 6 weeks for the frame of a BMS to become covered by smooth
muscle cells and 3 months to be protected by a normal endothelium. DES have a
slower endothelialization rate: 20 % at 3 months and 60 % at 1 year (Joner et al.
2006). Thus, the minimal duration of dual AP therapy following implantation is 6
8 Antiplatelet Therapy and Anticoagulation 113
weeks for BMS and 12 months for DES (Table 8.3) (Task Force on Myocardial
Revascularization of the European Society of, the European Association for Cardio-
Thoracic et al. 2010). These periods can be prolonged beyond 1 year for high-risk
stents (DES implanted in dominant, ostial, or bifurcated positions) and high-risk
patients (previous ST, diabetes, cardiac or renal failure). Late DES thrombosis is a
rare (0.6 %/year) but catastrophic event with a mortality of 9–45 % since it leads to
the acute interruption of flow in a previously normal vessel (Dangas et al. 2011).
New-generation DES have a faster rate of endothelialization and a lower incidence
of ST; depending on the type of stent, the duration of dual AP therapy is 6–12
months.
Mortality (%)
50
BMS, CABG
40 DES
30
20
10
1 3 6 12
Delay (months)
Fig. 8.3 Schematic illustration of the relationship between mortality for noncardiac surgery and
time since revascularization in case of perioperative interruption of dual antiplatelet therapy.
During the first 6 weeks after coronary artery bypass graft surgery (CABG), bare-metal stents
(BMS), or drug-eluting stents (DES), mortality is around 30 %. After BMS and CABG, it takes
3 months for postoperative mortality to reach the level of patients with no active coronary artery
disease, whereas after DES the plateau of the curve is reached only after 12 months
The body of evidence shows that aspirin or clopidogrel taken alone increase average
blood loss by 20 % during noncardiac surgery (Chassot et al. 2007). Some opera-
tions can show a significant increase in postoperative hemorrhage, such as
8 Antiplatelet Therapy and Anticoagulation 115
Current recommendations are based on the safest possible management of the dan-
gers of discontinuing AP agents prematurely (Douketis et al. 2008; American
Society of Anesthesiologists Task Force on Neuraxial et al. 2009; Task Force on
Myocardial Revascularization of the European Society of, the European Association
for Cardio-Thoracic et al. 2010; Korte et al. 2011). They are illustrated as an algo-
rithm in Fig. 8.4 and are summarized in Table 8.4.
• Aspirin for primary prevention can be interrupted 5 days before surgery, except
in high-risk cases, such as diabetics.
• Aspirin or clopidogrel monotherapy for secondary prevention after a stroke,
ACS, MI, or coronary revascularization should be maintained throughout the
perioperative period, whatever the duration of treatment.
• Aspirin plus dipyridamole dual therapy after stroke should be maintained
throughout the perioperative period.
• Aspirin plus clopidogrel/prasugrel/ticagrelor dual therapy in patients with a low
cardiovascular risk: maintain continuous treatment with aspirin; stop clopidogrel
5 days, prasugrel 7 days, and ticagrelor 5 days before surgery; restart
116 P.-G. Chassot et al.
High-risk
surgery
Elective Vital/urgent
High-risk All surgery surgery
intracranial surgery
Stop clopidogrel3
neurosurgery
Maintain aspirin
Delay Excessive
surgery bleeding risk2
Fig. 8.4 Algorithm for the preoperative management of patients under antiplatelet therapy. Low-
risk conditions are depicted in yellow, high-risk conditions in red, and decisions in magenta. 1
High-risk stents: multiple stents, long stent, proximal location (left main), and bifurcation lesions;
patients with previous stent thrombosis; stent in unique patent vessel. 2 Excessive risk of bleeding:
invasive surgery associated with severe bleeding and difficult hemostasis, or bleeding in closed
spaces (intracranial neurosurgery, intramedullary canal surgery, posterior eye chamber ophthalmic
surgery). 3 The same recommendations apply to prasugrel and ticagrelor. In all cases, restart AP
within 24 h postoperative. BMS bare-metal stent, DES drug-eluting stent, ACS acute coronary syn-
drome, MI myocardial infarction, PCI percutaneous coronary intervention, CABG coronary artery
bypass graft surgery (Adapted with permission from www.pac4.ch and form Eberli et al. (2010))
Table 8.4 Recommended preoperative management of patients under antiplatelet therapy accord-
ing to the hemorrhagic risk of surgery
Low-risk patient Intermediate risk High-risk patient
>3 months after 6–12 weeks after PCI, <6 weeks after PCI,
PCI, BMS, BMS, CABG, or CVA BMS, CABG, ACS, CVA
CABG or CVA 3–6 months after MI or <3 months MI or ACS
>6 months after ACS <6 months after MI or
ACS or MI >12 months after ACS with complications
>12 months high-risk DES <12 months after DES
after regular
DES
Low hemorrhagic risk Maintain aspirin Proceed with elective Postpone elective surgery
Transfusion not or clopidogrel surgery: maintain Proceed with vital/urgent
required aspirin surgery: maintain aspirin
Peripheral and wall Maintain clopidogrel, and clopidogrel,
surgery, minor ENT and prasugrel, or ticagrelor prasugrel, or ticagrelor
orthopedics, endoscopy if prescribed without interruption
without biopsy/
resection, eye anterior
chamber, dentistry
Intermediate risk Maintain aspirin Postpone elective Postpone elective surgery
Transfusion may be or clopidogrel surgery according to Proceed with vital
required risk balance surgery: maintain aspirin
Visceral and vascular Proceed with vital and clopidogrel,
surgery, major ENT and surgery: maintain prasugrel, or ticagrelor
orthopedics, urology, aspirin, maintain without interruption
endoscopy with biopsy/ clopidogrel, prasugrel,
resection or ticagrelor if
prescribed
High hemorrhagic risk Stop aspirin or Postpone elective Postpone elective surgery
Transfusion required clopidogrel if surgery Proceed with vital/urgent
Cardiac surgery, surgery necessary Proceed with vital/ surgery: maintain aspirin
with massive bleeding (5 days urgent surgery: Stop clopidogrel (5 days),
Surgery in closed space preoperatively) maintain aspirin prasugrel (7 days),
(intracranial, Restart <24 h Stop clopidogrel ticagrelor (3–5 days).
intramedullary canal, postoperatively (5 days), prasugrel Substitution with
posterior eye chamber) (7 days), or ticagrelor tirofiban or eptifibatide
(3–5 days) if (3–4 days perfusion)
prescribed, restart
<24 h postoperatively
Adapted with permission from Chassot et al. (2010) and from www.pac4.ch
ACS acute coronary syndrome, BMS bare-metal stents, CABG coronary artery bypass graft, CVA
cerebrovascular accident, DES drug-eluting stent, ENT ear, nose and throat surgery, MI myocardial
infarction, PCI percutaneous coronary intervention
prasugrel, when aspirin is maintained (Savonitto et al. 2010). The perfusion can be
stopped 6–8 h before surgery. After the operation, AP therapy should be resumed
within the first 24 h.
8.2 Anticoagulants
8.2.1 Introduction
8.2.2.1 Introduction
Unfractionated heparin (UFH) and low-molecular-weight heparins (LMWH) are
polysaccharides that bind to antithrombin (AT) and potentiate its inhibitory effect
on thrombin (FIIa) and activated factor X (FXa) (Fig. 8.5). This effect varies accord-
ing to the type of heparin molecule used:
• UFH predominantly potentiates anti-IIa activity.
• LMWHs predominantly potentiate anti-Xa activity.
Fondaparinux is a synthetic pentasaccharide that selectively binds and activates
AT. It is too short a molecule to enable bridging between AT and thrombin; thus, it
selectively potentiates the anti-Xa activation of AT without effecting thrombin; see
Fig. 8.5 (Garcia et al. 2012).
8.2.2.2 Monitoring
The anticoagulant response to UFH varies between patients; it is therefore standard
practice to monitor UFH and adjust the dose based on the results of coagulation
tests. When administered at therapeutic doses, the anticoagulant effect of UFH is
usually monitored using the activated partial thromboplastin time (aPTT). Activated
clotting time (ACT) allows monitoring of the higher UFH doses given in the context
of PCI or cardiac surgery. A therapeutic aPTT range of 1.5–2.5 times the control
time is widely accepted (Basu et al. 1972; Garcia et al. 2012). UFH levels can also
be monitored using anti-Xa assays. “Heparin resistance” is a term used when
patients require unusually high doses of heparin to achieve a therapeutic aPTT
(Green et al. 1994; Levine et al. 1994; Anand et al. 1997; Garcia et al. 2012). Several
mechanisms explain heparin resistance: AT deficiency (Olson et al. 1998), increased
UFH clearance (Hirsh et al. 1976; Green et al. 1994), high levels of heparin-binding
proteins (Whitfield et al. 1983; Brey 1992), and elevated levels of FVIII (Edson
et al. 1967; Levine et al. 1994) and/or fibrinogen (Edson et al. 1967).
Monitoring LMWH and fondaparinux is unnecessary, except in the contexts
of obesity, renal failure, or pregnancy (Garcia et al. 2012). LMWH and
120 P.-G. Chassot et al.
aPTT
PT/INR
Rivaroxaban
LMWH Apixaban
UFH
Dabigatran
TT
AT
Xa Xa Rivaroxaban
Apixaban
AT AT
AT
IIa IIa Dabigatran
Fig. 8.5 Anticoagulants and their targets (anticoagulation in the context of heparin-induced
thrombocytopenia is presented in Chap. 7). Vitamin K antagonists produce their anticoagulant
effect by interfering with the γ-carboxylation of the vitamin K-dependent factors II, VII, IX, and
X (in black). Unfractionated heparin (UFH) is an indirect anticoagulant that binds to antithrombin
(AT), enhancing its ability to inhibit activated factor X (FXa), thrombin (FIIa), and other coagula-
tion factors. Low-molecular-weight heparins (LMWH) derived from UFH by chemical or enzy-
matic depolymerization and fondaparinux is a synthetic analog of the AT-binding pentasaccharide
found in UFH and LMWH. Fondaparinux, too short to enable bridging between AT and thrombin,
selectively potentiates the anti-FXa activity of AT. Similarly LMWHs only have a marginal impact
on thrombin. Dabigatran is a direct, selective inhibitor of thrombin, hence independent of AT activ-
ity. Rivaroxaban and apixaban are direct, highly selective, factor Xa inhibitors. Clot-based assays
comprise the prothrombin time (PT), the activated partial thromboplastin time (aPTT), and the
thrombin time (TT). PT is used to assess the extrinsic and common pathways of coagulation.
Clotting is initiated by recalcifying citrated plasma in the presence of thromboplastin (a mixture of
tissue factor and phospholipids). In order to promote standardization of the PT, the World Health
Organization (WHO) developed an international reference thromboplastin and recommends that
the PT ratio be expressed as the International Normalized Ratio or INR to evaluate the effect of
anti-vitamin K anticoagulants. aPTT is used to assess the integrity of the intrinsic coagulation
pathway (prekallikrein, high-molecular-weight kininogen, factors XII, XI, IX, VIII) and final com-
mon pathway (factors II, V, X, and fibrinogen). It is performed by recalcifying citrated plasma in
the presence of a thromboplastic material that does not have tissue factor activity (hence the term
partial thromboplastin) and a negatively charged substance (i.e., celite, kaolin, or silica). TT mea-
sures the final step of the clotting pathway, the conversion of fibrinogen to fibrin. The test is per-
formed by recalcifying citrated plasma in the presence of dilute bovine or human thrombin
(Adapted with permission from EHM Swiss Medical Publishers Ltd. Gavillet and Angelillo-
Scherrer (2012))
8.2.2.3 Reversal
Protamine sulfate is a basic protein that was originally extracted from salmon testi-
cles. It displaces AT and neutralizes heparin by forming a complex with it, and it has
a partial antagonist effect on LMWH. In the absence of heparin, protamine sulfate
shows an anticoagulant effect. It is routinely used after cardiopulmonary bypass,
but rarely for bleeding resulting from heparin administration. The administration of
protamine sulfate can be associated with hemodynamic changes ranging from mild
systemic hypotension to severe pulmonary hypertension with hemodynamic col-
lapse. Several mechanisms can contribute to these effects: direct protamine-induced
histamine release, anaphylactic reactions (IgE mediated), and anaphylactoid reac-
tions (IgG mediated). The immune-mediated reactions can be based on anti-prot-
amine antibodies or on anti-heparin-protamine complex antibodies. It is generally
recommended to slowly administer protamine through a peripheral venous line,
since central venous administration could exacerbate the adverse reactions. Three
elements need to be specified in order to calculate a correct dosage:
1. Route of heparin administration (subcutaneous half-life > intravenous half-life)
2. Type of heparin (half-life of LMWH > half-life of UFH)
3. Delay between heparin administration and protamine sulfate administration
Dosage is also based on the fact that 1 mg of protamine sulfate inactivates 100 IU
of heparin or 100 IU anti-Xa of LMWH.
When the clinical setting requires the neutralization of LMWH’s anticoagulant
effect, the following approach is proposed (Garcia et al. 2012):
• If LMWH was administered within 8 h, protamine sulfate must be given at a dose
of 1 mg per 100 IU of anti-Xa activity up to a maximum single dose of 50 mg
(1 mg enoxaparin equals approximately 100 IU anti-Xa).
• A second dose of 0.5 mg protamine sulfate per 100 IU anti-Xa should be pro-
vided if bleeding persists.
• Smaller doses of protamine sulfate can be administered if the time since LMWH
administration is longer than 8 h.
It is important to accurately calculate the necessary protamine dose since it has
an intrinsic anticoagulant effect, which may lead to increased bleeding in case of an
overdose. Fondaparinux does not bind to protamine sulfate. If uncontrollable bleed-
ing occurs with fondaparinux, recombinant activated FVII may be effective
(Bijsterveld et al. 2002).
8 Antiplatelet Therapy and Anticoagulation 123
Vitamin K antagonists (VKA) are classic oral anticoagulation drugs that generate the
same effect as a vitamin K deficiency. They include phenprocoumon, warfarin, and
acenocoumarol. In elective perioperative settings, these drugs are replaced by UFH,
LMWH, or fondaparinux, as these periods are associated with a thromboembolic risk
of 0–2 % if VKA are interrupted (Mourelo et al. 2008) and a bleeding risk of 2–25 %
if VKA are continued during surgery (Jaffer et al. 2010). There are several ways to
reverse the anti-vitamin K effect. For elective surgery, the patient can simply stop
taking VKA with overlapping treatment of LMWH. If surgery is more urgent, but not
immediate, the patient can receive a vitamin K supplement, for example, 10 mg/day
of intravenous vitamin K1. Finally, for same-day urgent surgery, prothrombin com-
plex concentrates (PCC) can be administered. For a 70 kg patient with an estimated
plasma volume of 2,500 ml, the substitution dose is calculated as follows:
If the patient’s weight is significantly different, the plasma volume can be esti-
mated according to the following formula:
For a durable reversible effect, vitamin K1 is associated with PCC. The duration
of vitamin K1 administration depends on the VKA half-life.
8.2.4.1 Introduction
Novel oral anticoagulants (NOACs) specifically target either thrombin or FXa
(Fig. 8.5). They have a rapid onset of action, few drug interactions, and predictable
pharmacokinetics and pharmacodynamics, making routine coagulation monitoring
unnecessary. However, there are situations in which assessment of the anticoagulant
effect of OACs is important: these include hemorrhage or thrombosis occurring
under anticoagulation, emergency surgery, polypharmacy, overdose, renal or liver
failure, compliance monitoring, and extreme bodyweights. Moreover, NOACs
affect routine coagulation tests (Table 8.5).
Management protocols of NOACs prior to elective surgery exist, but clinical
experience is currently insufficient to provide solid guidelines on the management
of emergencies including major bleeding in patients receiving NOACs. No specific
antidotes are available at present.
checking renal function and slightly modify the reversal protocol in case of renal
failure (Table 8.6). Reversal can be monitored by measuring anti-FXa activity (spe-
cific assays) (Barrett et al. 2010; Samama et al. 2012).
situation and not on laboratory tests. It is important to realize that these products
are highly prothrombotic and that their administration might be complicated by
thrombotic events. Their use should therefore be limited to life-threatening situa-
tions. For dabigatran, reversal can be monitored by measuring the thrombin time
(see Sect. 8.2.4.3). However, because this test is highly sensitive to dabigatran, an
assessment of its concentration by specific tests would be more accurate (Stangier
et al. 2007). For anti-Xa drugs, reversal can be monitored by measuring anti-Xa
activity (Barrett et al. 2010; Samama et al. 2012). Hemodialysis could complete the
reversal strategy for dabigatran (Warkentin et al. 2012).
The use of inferior vena cava filter is recommended in patients with acute proximal
deep vein thrombosis and/or pulmonary embolism who have a contraindication to
anticoagulants, i.e., an unacceptable risk of bleeding (Garcia et al. 2012). If the
contraindication to anticoagulation is temporary (e.g., during active bleeding), it is
possible to insert a temporary retrievable filter and remove it when anticoagulation
treatment can be safely restarted. However, it is worth noting that most retrievable
filters are not removed (Mismetti et al. 2007; Dabbagh et al. 2010; Jaff et al. 2011;
Garcia et al. 2012). Furthermore, retrievable filters that do not get removed might
display a higher complication rate than permanent filters (Mismetti et al. 2007,
p 223; Dabbagh et al. 2010, p 493; Nicholson et al. 2010, p 1827).
8 Antiplatelet Therapy and Anticoagulation 127
Insertion of an inferior vena cava filter does not eliminate the risk of pulmonary
embolism and does increase the risk of deep vein thrombosis. Consequently, it is
suggested that patients who have an inferior vena cava filter inserted should receive
a conventional course of anticoagulants when the contraindication to anticoagula-
tion is withdrawn (Garcia et al. 2012). Venous thrombosis at the site of filter inser-
tion occurs in about 10 % of patients (Streiff 2000).
References
Ageno W, Gallus AS et al (2012) Oral anticoagulant therapy: Antithrombotic Therapy and
Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines. Chest 141(2 Suppl):e44S–e88S
Albaladejo P, Marret E et al (2011) Non-cardiac surgery in patients with coronary stents: the
RECO study. Heart 97(19):1566–1572
American Society of Anesthesiologists Task Force on Neuraxial Opioids, Horlocker TT et al
(2009) Practice guidelines for the prevention, detection, and management of respiratory depres-
sion associated with neuraxial opioid administration. Anesthesiology 110(2):218–230
Anand SS, Brimble S et al (1997) Management of iliofemoral thrombosis in a pregnant patient
with heparin resistance. Arch Intern Med 157(7):815–816
Angiolillo DJ, Firstenberg MS et al (2012) Bridging antiplatelet therapy with cangrelor in patients
undergoing cardiac surgery: a randomized controlled trial. JAMA 307(3):265–274
Aradi D, Komocsi A et al (2010) Prognostic significance of high on-clopidogrel platelet reactivity
after percutaneous coronary intervention: systematic review and meta-analysis. Am Heart J
160(3):543–551
Artang R, Dieter RS (2007) Analysis of 36 reported cases of late thrombosis in drug-eluting stents
placed in coronary arteries. Am J Cardiol 99(8):1039–1043
Asmis LM, Alberio L et al (2012) Rivaroxaban: Quantification by anti-FXa assay and influence on
coagulation tests: a study in 9 Swiss laboratories. Thromb Res 129(4):492–498
Barash P, Akhtar S (2010) Coronary stents: factors contributing to perioperative major adverse
cardiovascular events. Br J Anaesth 105(Suppl 1):i3–i15
Barrett YC, Wang Z et al (2010) Clinical laboratory measurement of direct factor Xa inhibitors:
anti-Xa assay is preferable to prothrombin time assay. Thromb Haemost 104(6):1263–1271
Basu D, Gallus A et al (1972) A prospective study of the value of monitoring heparin treatment
with the activated partial thromboplastin time. N Engl J Med 287(7):324–327
Bates ER, Lau WC et al (2011) Clopidogrel-drug interactions. J Am Coll Cardiol 57(11):
1251–1263
Berger PB, Bellot V et al (2001) An immediate invasive strategy for the treatment of acute myo-
cardial infarction early after noncardiac surgery. Am J Cardiol 87(9):1100–1102, A1106,
A1109
Bhatt DL, Fox KA et al (2006) Clopidogrel and aspirin versus aspirin alone for the prevention of
atherothrombotic events. N Engl J Med 354(16):1706–1717
Bijsterveld NR, Moons AH et al (2002) Ability of recombinant factor VIIa to reverse the antico-
agulant effect of the pentasaccharide fondaparinux in healthy volunteers. Circulation
106(20):2550–2554
Biondi-Zoccai GG, Lotrionte M et al (2006) A systematic review and meta-analysis on the hazards
of discontinuing or not adhering to aspirin among 50,279 patients at risk for coronary artery
disease. Eur Heart J 27(22):2667–2674
Brey RL (1992) Antiphospholipid antibodies and ischemic stroke. Heart Dis Stroke 1(6):379–382
Burger W, Chemnitius JM et al (2005) Low-dose aspirin for secondary cardiovascular prevention –
cardiovascular risks after its perioperative withdrawal versus bleeding risks with its
continuation – review and meta-analysis. J Intern Med 257(5):399–414
128 P.-G. Chassot et al.
Chassot PG, Delabays A et al (2007) Perioperative antiplatelet therapy: the case for continuing
therapy in patients at risk of myocardial infarction. Br J Anaesth 99(3):316–328
Chassot PG, Marcucci C et al (2010) Perioperative antiplatelet therapy. Am Fam Physician
82(12):1484–1489
Chernoguz A, Telem DA et al (2011) Cessation of clopidogrel before major abdominal procedures.
Arch Surg 146(3):334–339
Connolly SJ, Ezekowitz MD et al (2009) Dabigatran versus warfarin in patients with atrial fibril-
lation. N Engl J Med 361(12):1139–1151
Dabbagh O, Nagam N et al (2010) Retrievable inferior vena cava filters are not getting retrieved:
where is the gap? Thromb Res 126(6):493–497
Dangas GD, Caixeta A et al (2011) Frequency and predictors of stent thrombosis after percutane-
ous coronary intervention in acute myocardial infarction. Circulation 123(16):1745–1756
Douketis JD, Berger PB et al (2008) The perioperative management of antithrombotic therapy:
American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th
Edition). Chest 133(6 Suppl):299S–339S
Eberli D, Chassot PG et al (2010) Urological surgery and antiplatelet drugs after cardiac and cere-
brovascular accidents. J Urol 183(6):2128–2136
Edson JR, Krivit W et al (1967) Kaolin partial thromboplastin time: high levels of procoagulants
producing short clotting times or masking deficiencies of other procoagulants or low concen-
trations of anticoagulants. J Lab Clin Med 70(3):463–470
Eerenberg ES, Kamphuisen PW et al (2011) Reversal of rivaroxaban and dabigatran by prothrom-
bin complex concentrate: a randomized, placebo-controlled, crossover study in healthy sub-
jects. Circulation 124(14):1573–1579
Eisenberg MJ, Richard PR et al (2009) Safety of short-term discontinuation of antiplatelet therapy
in patients with drug-eluting stents. Circulation 119(12):1634–1642
Fujimoto H, Ishimura R et al (2009) Two cases of acute coronary syndrome that occurred by pre-
operative discontinuation of antiplatelet therapy in the chronic phase after stent implantation.
J Cardiol 54(3):470–474
Gaglia MA Jr, Waksman R (2011) Systematic review of thienopyridine discontinuation and its
impact upon clinical outcomes. Eur Heart J 32(19):2358–2364
Garcia DA, Baglin TP et al (2012) Parenteral anticoagulants: Antithrombotic Therapy and
Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines. Chest 141(2 Suppl):e24S–e43S
Gavillet M, Angelillo-Scherrer A (2012) Quantification of the anticoagulatory effect of novel anti-
coagulants and management of emergencies. Cardiovasc Med 15(5):170–179
Gogarten W, Vandermeulen E et al (2010) Regional anaesthesia and antithrombotic agents: recom-
mendations of the European Society of Anaesthesiology. Eur J Anaesthesiol 27(12):999–1015
Green D, Hirsh J et al (1994) Low molecular weight heparin: a critical analysis of clinical trials.
Pharmacol Rev 46(1):89–109
Gurbel PA, Bliden KP et al (2009) Randomized double-blind assessment of the ONSET and
OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coro-
nary artery disease: the ONSET/OFFSET study. Circulation 120(25):2577–2585
Hall R, Mazer CD (2011) Antiplatelet drugs: a review of their pharmacology and management in
the perioperative period. Anesth Analg 112(2):292–318
Hirsh J, van Aken WG et al (1976) Heparin kinetics in venous thrombosis and pulmonary
embolism. Circulation 53(4):691–695
Jaff MR, McMurtry MS et al (2011) Management of massive and submassive pulmonary embo-
lism, iliofemoral deep vein thrombosis, and chronic thromboembolic pulmonary hypertension:
a scientific statement from the American Heart Association. Circulation 123(16):1788–1830
Jaffer AK, Brotman DJ et al (2010) Variations in perioperative warfarin management: outcomes
and practice patterns at nine hospitals. Am J Med 123(2):141–150
Joner M, Finn AV et al (2006) Pathology of drug-eluting stents in humans: delayed healing and late
thrombotic risk. J Am Coll Cardiol 48(1):193–202
8 Antiplatelet Therapy and Anticoagulation 129
Kearon C, Akl EA et al (2012) Antithrombotic therapy for VTE disease: Antithrombotic Therapy
and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines. Chest 141(2 Suppl):e419S–e494S
Korte W, Cattaneo M et al (2011) Peri-operative management of antiplatelet therapy in patients
with coronary artery disease: joint position paper by members of the working group on
Perioperative Haemostasis of the Society on Thrombosis and Haemostasis Research (GTH),
the working group on Perioperative Coagulation of the Austrian Society for Anesthesiology,
Resuscitation and Intensive Care (OGARI) and the Working Group Thrombosis of the
European Society for Cardiology (ESC). Thromb Haemost 105(5):743–749
Kubitza D, Becka M et al (2005) Safety, pharmacodynamics, and pharmacokinetics of single doses
of BAY 59-7939, an oral, direct factor Xa inhibitor. Clin Pharmacol Ther 78(4):412–421
Levine MN, Hirsh J et al (1994) A randomized trial comparing activated thromboplastin time with
heparin assay in patients with acute venous thromboembolism requiring large daily doses of
heparin. Arch Intern Med 154(1):49–56
Mega JL, Simon T et al (2010) Reduced-function CYP2C19 genotype and risk of adverse clinical
outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis.
JAMA 304(16):1821–1830
Mismetti P, Rivron-Guillot K et al (2007) A prospective long-term study of 220 patients with a
retrievable vena cava filter for secondary prevention of venous thromboembolism. Chest
131(1):223–229
Moore M, Power M (2004) Perioperative hemorrhage and combined clopidogrel and aspirin ther-
apy. Anesthesiology 101(3):792–794
Mourelo R, Kaidar-Person O et al (2008) Hemorrhagic and thromboembolic complications after
bariatric surgery in patients receiving chronic anticoagulation therapy. Obes Surg 18(2):
167–170
Nicholson W, Nicholson WJ et al (2010) Prevalence of fracture and fragment embolization of Bard
retrievable vena cava filters and clinical implications including cardiac perforation and tampon-
ade. Arch Intern Med 170(20):1827–1831
Nuttall GA, Brown MJ et al (2008) Time and cardiac risk of surgery after bare-metal stent percu-
taneous coronary intervention. Anesthesiology 109(4):588–595
Olson JD, Arkin CF et al (1998) College of American Pathologists Conference XXXI on labora-
tory monitoring of anticoagulant therapy: laboratory monitoring of unfractionated heparin
therapy. Arch Pathol Lab Med 122(9):782–798
Patel MR, Mahaffey KW et al (2011) Rivaroxaban versus warfarin in nonvalvular atrial fibrillation.
N Engl J Med 365(10):883–891
Patrono C, Rocca B (2010) The future of antiplatelet therapy in cardiovascular disease. Annu Rev
Med 61:49–61
Pengo V, Crippa L et al (2011) Questions and answers on the use of dabigatran and perspectives
on the use of other new oral anticoagulants in patients with atrial fibrillation. A consensus
document of the Italian Federation of Thrombosis Centers (FCSA). Thromb Haemost
106(5):868–876
Pharma BS (2009) Xarelto. Summary of product characteristics. www.xarelto.com/html/
downloads/Xarelto_Summary_of_Product_Characteristics_May2009.pdf. Accessed Mar 2011
Price MJ (2009) Bedside evaluation of thienopyridine antiplatelet therapy. Circulation 119(19):
2625–2632
Rabbitts JA, Nuttall GA et al (2008) Cardiac risk of noncardiac surgery after percutaneous coro-
nary intervention with drug-eluting stents. Anesthesiology 109(4):596–604
Samama MM, Contant G et al (2012) Evaluation of the anti-factor Xa chromogenic assay for the
measurement of rivaroxaban plasma concentrations using calibrators and controls. Thromb
Haemost 107(2):379–387
Savonitto S, D’Urbano M et al (2010) Urgent surgery in patients with a recently implanted coro-
nary drug-eluting stent: a phase II study of ‘bridging’ antiplatelet therapy with tirofiban during
temporary withdrawal of clopidogrel. Br J Anaesth 104(3):285–291
130 P.-G. Chassot et al.
Schouten O, van Domburg RT et al (2007) Noncardiac surgery after coronary stenting: early sur-
gery and interruption of antiplatelet therapy are associated with an increase in major adverse
cardiac events. J Am Coll Cardiol 49(1):122–124
Schulz S, Schuster T et al (2009) Stent thrombosis after drug-eluting stent implantation: incidence,
timing, and relation to discontinuation of clopidogrel therapy over a 4-year period. Eur Heart J
30(22):2714–2721
Sharma AK, Ajani AE et al (2004) Major noncardiac surgery following coronary stenting: when is
it safe to operate? Catheter Cardiovasc Interv 63(2):141–145
Stangier J, Rathgen K et al (2007) The pharmacokinetics, pharmacodynamics and tolerability of
dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br J Clin
Pharmacol 64(3):292–303
Streiff MB (2000) Vena caval filters: a comprehensive review. Blood 95(12):3669–3677
Task Force on Myocardial Revascularization of the European Society of, Cardiology; The
European Association for Cardio-Thoracic Surgery; et al (2010) Guidelines on myocardial
revascularization. Eur Heart J 31(20):2501–2555
Taylor K, Filgate R et al (2011) A retrospective study to assess the morbidity associated with
transurethral prostatectomy in patients on antiplatelet or anticoagulant drugs. BJU Int
108(Suppl 2):45–50
Valgimigli M, Campo G et al (2012) Short- versus long-term duration of dual-antiplatelet therapy
after coronary stenting: a randomized multicenter trial. Circulation 125(16):2015–2026
van Ryn J, Stangier J et al (2010) Dabigatran etexilate–a novel, reversible, oral direct thrombin
inhibitor: interpretation of coagulation assays and reversal of anticoagulant activity. Thromb
Haemost 103(6):1116–1127
Wallentin L, Becker RC et al (2009) Ticagrelor versus clopidogrel in patients with acute coronary
syndromes. N Engl J Med 361(11):1045–1057
Warkentin TE, Margetts P et al (2012) Recombinant factor VIIa (rFVIIa) and hemodialysis to man-
age massive dabigatran-associated postcardiac surgery bleeding. Blood 119(9):2172–2174
Weitz JI (2010) New oral anticoagulants in development. Thromb Haemost 103(1):62–70
Whitfield LR, Lele AS et al (1983) Effect of pregnancy on the relationship between concentration
and anticoagulant action of heparin. Clin Pharmacol Ther 34(1):23–28
Wilson SH, Fasseas P et al (2003) Clinical outcome of patients undergoing non-cardiac surgery in
the two months following coronary stenting. J Am Coll Cardiol 42(2):234–240
Wiviott SD, Braunwald E et al (2007) Prasugrel versus clopidogrel in patients with acute coronary
syndromes. N Engl J Med 357(20):2001–2015
Zhang D, He K et al (2009) Comparative metabolism of 14C-labeled apixaban in mice, rats,
rabbits, dogs, and humans. Drug Metab Dispos 37(8):1738–1748
Intravenous Fluids and Coagulation
9
Herbert Schöchl, Christoph Schlimp,
and Wolfgang Voelckel
9.1 Introduction
H. Schöchl (*)
AUVA Trauma Center, Salzburg, Austria
Ludwig Boltzmann Institute for experimental and clinical Traumatology, Viennna, Austria
e-mail: [email protected]
C. Schlimp
Ludwig Boltzmann Institute for experimental and clinical Traumatology, Viennna, Austria
W. Voelckel
AUVA Trauma Center, Salzburg, Austria
Fig. 9.1 ROTEM® results from an in vitro model of 33 % hemodilution. Two ROTEM® tests were
performed: an extrinsically activated test using tissue factor (EXTEM) and an extrinsically acti-
vated test without platelet component (FIBTEM). The most pronounced reduction in maximum
clot firmness (MCF) was observed with 6 % HES 130/0.4. Dilution of whole blood with 5 %
human albumin and 4 % gelatin produced similar reductions in MCF. Normal saline induced only
moderate changes in MCF
(Schierhout and Roberts 1998). There is a paucity of evidence that one colloid is
superior to another with regard to survival (Bunn and Trivedi 2012).
Infusion of large amounts of fluid results in nonspecific dilution of coagulation
factors, coagulation inhibitors, and platelets (Table 9.1). In addition, the administra-
tion of artificial colloids leads to specific alterations of coagulation factors. Acquired
von Willebrand syndrome and low FVIII activity can be observed, impairing both
9 Intravenous Fluids and Coagulation 133
Crystalloid fluids are widely used for volume resuscitation. These solutions are safe
and well tolerated at low volumes, but their intravascular volume expansion effect
is low. Within minutes, 80 % or more of the infused volume can cross the capillary
a b
c d
Voluven® Gelofusion®
Fig. 9.2 In vitro effects on clotting of 50 % dilution of whole blood with different fluids: (a) no
dilution (whole blood), (b) Ringer’s lactate, (c) 6 % HES 130/0.4, and (d) 4 % gelatin. 6 % HES
130/0.4 disturbed the fibrin meshwork more than Ringer’s lactate or gelatin (With permission from
Sorensen and Fries (2012))
134 H. Schöchl et al.
membrane from the intravascular compartment into the interstitial space (McIlroy
and Kharasch 2003). Therefore, large amounts of crystalloids are often necessary to
provide a sufficient increase in intravascular volume, introducing a significant risk
of soft tissue edema.
Normal saline (NS) is a solution with 0.9 % of sodium chloride, with an osmo-
lality of 308 mOsm/l. It contains 154 mmol/l of sodium and 154 mmol/l of chlo-
ride. It can therefore be considered neither physiological nor balanced, yet NS is
frequently used as a plasma substitute. In contrast, balanced electrolyte solutions
are isotonic and have electrolyte compositions close to that of plasma (Guidet et al.
2010).
In vivo and in vitro studies have suggested a hypercoagulable state following mod-
erate hemodilution with crystalloids (Ruttmann 2002; Ruttmann et al. 1996, 2001,
2006; Petroianu et al. 2000; Ng et al. 2002). Other investigators refute such a pro-
hemostatic effect and suggest that these findings are an in vitro phenomenon only
(Innerhofer et al. 2002).
High-volume crystalloid replacement therapy results in dilution not only of pro-
hemostatic coagulation factors but also of coagulation system inhibitors, such as
antithrombin III (AT III) and proteins S and C (Ruttmann et al. 1996). A variety of
positive feedback loops in the clotting mechanism are regulated by AT III; there-
fore, a small change in its concentration may have a profound effect on the initiation
and amplification of the clotting process – potentially much greater than that sug-
gested by the change in absolute concentration (Jesty 1986). Indeed, Dunbar and
Chandler measured thrombin generation in plasma diluted to 40 % of normal,
which resulted in an increase in thrombin generation. Dilution in excess of 40 %,
however, caused a decline in thrombin generation. Thus, when all plasma proteins
decrease at the same time, including inhibitory factors such as AT III, thrombin
generation initially increases (Dunbar and Chandler 2009).
In vivo studies comparing both fluids reported only minor differences between bal-
anced and non-balanced solutions. Waters et al. compared RL with NS in major
aortic surgery. Only minor, nonsignificant differences in blood loss, in favor of RL,
were observed. There were no differences in transfusion of red blood cells (RBCs)
or fresh frozen plasma (FFP) (Waters et al. 2001).
Studies comparing colloids in NS with balanced electrolyte solutions have not
reported relevant differences in blood loss. For example, Casutt et al. revealed that
HES 130/0.4 in a balanced solvent had similar effects on hemostasis (Casutt et al.
2010). Kulla et al. investigated patients undergoing abdominal surgery. No significant
differences in coagulation parameters and blood loss were observed between bal-
anced and unbalanced HES solutions (Kulla et al. 2008). Schaden et al. recently com-
pared two different HES preparations containing tetrastarch in balanced solution, or
NS, in healthy volunteers. Blood was subjected to a blood gas analysis, an assessment
of platelet function by multiple electrode aggregometry, and thromboelastometry.
After infusion of either 20 ml/kg HES in balanced solution or in NS, there was no
significant change in calcium concentration, and only a minor impact on platelet
aggregation and clot formation as assessed by ROTEM® (Schaden et al. 2012).
136 H. Schöchl et al.
9.4.1 Albumin
effects on dynamic clot formation, followed by HA. The latter reduced activated
clotting time by 2 % and exhibited a 47 % reduction in clot rate (rate of fibrin forma-
tion) (Coats et al. 2006).
Our own group recently compared NS with 5 % HA, 4 % gelatin, and 6 % HES
130/0.4 in an in vitro dilution model investigating ROTEM® variables. Compared to
undiluted whole blood, 33 % in vitro hemodilution with 5 % HA reduced maximum
clot firmness in the extrinsically activated EXTEM test, and a more pronounced
reduction was observed in the fibrin polymerization test (FIBTEM). Our in vitro
study confirmed the observation that 5 % HA has less effect on ROTEM® test results
than other colloids (Schlimp et al. 2013).
9.4.2 Dextran
due to the fact that it is eliminated by the kidneys more quickly than other colloids
(de Jonge and Levi 2001).
In most European countries, dextrans have disappeared from the market due to
potentially life-threatening side effects such as severe anaphylactic reactions, acute
renal failure, and bleeding diatheses.
including platelets. Prolonged bleeding times, observed after dextran infusion, are
in part related to diminished platelet function (Alexander et al. 1975).
9.4.3 Gelatin
incorporated into the clot as it forms, interfering with fibrin architecture and impair-
ing the clot’s strength and stability (Mardel et al. 1998; Kheirabadi et al. 2008).
9.4.4 Starches
HES are derived from amylopectin and consist of highly branched polysaccharides,
similar to glycogen. HES solutions are available either as iso-oncotic 6 % or hyper-
oncotic 10 % fluids. The volume effect of a 10 % solution exceeds the infused vol-
ume, and it is therefore considered a plasma expander (Westphal et al. 2009).
Compared to gelatin and crystalloids, the initial volume expansion of HES is greater
and lasts longer (Van der Linden and Ickx 2006).
HES are polydisperse fluids with a wide range of MW. HES solutions can be
categorized as high-MW (>400 kDa), medium-MW (200–400 kDa), and low-MW
fluids (<200 kDa) (Jungheinrich and Neff 2005).
The most important physicochemical aspect of HES is the degree of substitution,
defined as the proportion of hydroxyl groups per glucose molecule replaced by
hydroxyethyl subunits. Increased substitution increases the solubility of starch in
water and, more importantly, reduces the rate of destruction by serum amylase
(Kozek-Langenecker 2005). Highly substituted (hetastarch 0.62–0.75), medium-
substituted (pentastrach 0.5), and low-substituted (tetrastarch 0.4) HES preparations
are available.
9 Intravenous Fluids and Coagulation 141
Several different HS preparations, from 1.6 to 29.9 %, with and without the addition
of artificial colloids, have been used in experimental and clinical studies (Johnson
144 H. Schöchl et al.
and Criddle 2004). However, sound conclusions regarding the influence of HS/HHS
are impossible due to the heterogeneity of the fluids.
Coats et al. investigated the in vitro effect of HHS (including dextran) on coagu-
lation. A mild pro-hemostatic effect was observed up to a dilution of 11 %, but
anticoagulant properties were evident above this level. At 15 % dilution, coagula-
tion was grossly deranged (Coats and Heron 2004).
Wilder et al. reported that HS has strong anticoagulant and antiplatelet effects.
Dilution with HS to a level of only 5 % caused a significant prolongation of PT. In
contrast, there was no significant change in PT following 5 % dilution with hyper-
tonic sorbitol or mixtures based on glycine (Wilder et al. 2002).
A 20 % dilution with 3 % saline resulted in a 52 % reduction of thrombin genera-
tion and 11 % reduction of fibrin formation. Increasing the dilution to 30 %
decreased fibrin formation by 89 % (Brummel-Ziedins et al. 2006).
In vitro, 10 % dilution of blood with HHS has been shown to cause significant
impairment of fibrin polymerization (Hanke et al. 2011). Haas et al. studied the
effect of HHS in vivo in an uncontrolled bleeding model in pigs. Median fibrinogen
polymerization was significantly higher among animals receiving HHS, compared
with those receiving 4 % gelatin or 6 % HES 130/0.4. Furthermore, median blood
loss after liver incision was significantly lower in the HHS group (Haas et al. 2008a).
All artificial colloids interfere with the process of fibrinogen polymerization, reduc-
ing maximum clot firmness in viscoelastic coagulation tests. To overcome this prob-
lem, fibrinogen supplementation has proved effective both in vitro and in vivo (Fries
et al. 2005; Schramko et al. 2009; Grottke et al. 2010; Schlimp et al. 2013). Using a
9 Intravenous Fluids and Coagulation 145
a b c
Fig. 9.3 Electron microscopy scans of blood clots formed from (a) undiluted whole blood,
(b) blood diluted 65 % with gelatin, and (c) blood diluted 65 % with gelatin and the addition of
fibrinogen concentrate (With permission from Fries and Martini (2010))
33 % in vitro dilution model, Schlimp et al. recently showed that fibrinogen concen-
trate was effective in increasing clot firmness in blood samples diluted with either
albumin or gelatin. In samples diluted with HES, however, the effect was poor
(Schlimp et al. 2013). Fries et al. reported improved clot firmness following fibrino-
gen administration in a porcine model of uncontrolled bleeding (Fig. 9.3) (Fries
et al. 2005). Grottke et al. replaced 80 % of blood volume with HES 130/0.4, RL,
and retransfused erythrocytes. The animals randomly received 70 mg/kg fibrinogen
concentrate, 200 mg/kg fibrinogen concentrate, or a placebo before blunt liver
injury was induced. Total blood loss was significantly lower, and survival was sig-
nificantly higher, in both fibrinogen groups as compared to controls. Microscopy
revealed no thrombosis in either group (Grottke et al. 2010).
Factor XIII supplementation, together with fibrinogen, partially restores clot forma-
tion in blood samples diluted in vitro with different colloids (Niemi et al. 2005). Again,
the effect is significantly lower in the HES group compared to albumin and gelatin.
The combination of factor XIII and fibrinogen effectively normalized maximum clot
firmness in blood samples diluted with either albumin or gelatin. Haas and coworkers
further investigated the effect of fibrinogen, factor XIII, and FFP in a 60 % hemodilu-
tion model. Fibrinogen together with factor XIII restored crystalloid-induced impair-
ment of clot strength, while FFP only shortened coagulation times (Haas et al. 2008b).
As shown in elective surgery patients, desmopressin has the potential to reverse
the colloid-induced decreases in levels of factor VIII and vWF (Conroy et al. 1996).
Another potential option for such reversal is the administration of factor VIII/vWF
concentrate, but there are no clinical data to support this strategy.
Conclusion
Crystalloids exert only minor effects on coagulation parameters, apart from the
effects of hemodilution. Artificial colloids impair coagulation and platelet func-
tion to a greater extent than natural colloid albumin. Gelatin and rapidly degrad-
able, short-acting HES seem to have similar effects on blood loss. There is no
conclusive evidence that balanced solutions are superior to non-balanced fluids.
146 H. Schöchl et al.
References
Alexander B, Odake K et al (1975) Coagulation, hemostasis, and plasma expanders:a quarter cen-
tury enigma. Fed Proc 34(6):1429–1440
Angle N, Hoyt DB et al (1998) Hypertonic saline resuscitation diminishes lung injury by suppress-
ing neutrophil activation after hemorrhagic shock. Shock 9(3):164–170
Batlle J, del Rio F et al (1985) Effect of dextran on factor VIII/von Willebrand factor structure and
function. Thromb Haemost 54(3):697–699
Brodin B, Hesselvik F et al (1984) Decrease of plasma fibronectin concentration following infu-
sion of a gelatin-based plasma substitute in man. Scand J Clin Lab Invest 44(6):529–533
Brummel-Ziedins K, Whelihan MF et al (2006) The resuscitative fluid you choose may potentiate
bleeding. J Trauma 61(6):1350–1358
Bulger EM, May S et al (2010) Out-of-hospital hypertonic resuscitation following severe trau-
matic brain injury: a randomized controlled trial. JAMA 304(13):1455–1464
Bulger EM, May S et al (2011) Out-of-hospital hypertonic resuscitation after traumatic hypovole-
mic shock: a randomized, placebo controlled trial. Ann Surg 253(3):431–441
Bunn F, Trivedi D (2012) Colloid solutions for fluid resuscitation. Cochrane Database Syst Rev
7:CD001319
Casutt M, Kristoffy A et al (2010) Effects on coagulation of balanced (130/0.42) and non-balanced
(130/0.4) hydroxyethyl starch or gelatin compared with balanced Ringer's solution: an in vitro
study using two different viscoelastic coagulation tests ROTEMTM and SONOCLOTTM. Br
J Anaesth 105(3):273–281
Choi PT, Yip G et al (1999) Crystalloids vs. colloids in fluid resuscitation: a systematic review.
Crit Care Med 27(1):200–210
Clagett GP, Anderson FA Jr et al (1995) Prevention of venous thromboembolism. Chest 108
(4 Suppl):312S–334S
Coats TJ, Heron M (2004) The effect of hypertonic saline dextran on whole blood coagulation.
Resuscitation 60(1):101–104
Coats TJ, Brazil E et al (2006) The effects of commonly used resuscitation fluids on whole blood
coagulation. Emerg Med J 23(7):546–549
Conroy JM, Fishman RL et al (1996) The effects of desmopressin and 6% hydroxyethyl starch on
factor VIII:C. Anesth Analg 83(4):804–807
de Jonge E, Levi M (2001) Effects of different plasma substitutes on blood coagulation: a com-
parative review. Crit Care Med 29(6):1261–1267
de Jonge E, Levi M et al (1998) Impaired haemostasis by intravenous administration of a gelatin-
based plasma expander in human subjects. Thromb Haemost 79(2):286–290
Denis R, Smith RW et al (1987) Relocation of nonalbumin proteins after albumin resuscitation.
J Surg Res 43(5):413–419
Deusch E, Gamsjager T et al (2003) Binding of hydroxyethyl starch molecules to the platelet sur-
face. Anesth Analg 97(3):680–683
Deusch E, Thaler U et al (2004) The effects of high molecular weight hydroxyethyl starch solu-
tions on platelets. Anesth Analg 99(3):665–668
Dunbar NM, Chandler WL (2009) Thrombin generation in trauma patients. Transfusion
49(12):2652–2660
Egli GA, Zollinger A et al (1997) Effect of progressive haemodilution with hydroxyethyl starch,
gelatin and albumin on blood coagulation. Br J Anaesth 78(6):684–689
Engvall E, Ruoslahti E et al (1978) Affinity of fibronectin to collagens of different genetic types
and to fibrinogen. J Exp Med 147(6):1584–1595
Entholzner EK, Mielke LL et al (2000) Coagulation effects of a recently developed hydroxyethyl
starch (HES 130/0.4) compared to hydroxyethyl starches with higher molecular weight. Acta
Anaesthesiol Scand 44(9):1116–1121
Eriksson M, Saldeen T (1995) Effect of dextran on plasma tissue plasminogen activator (t-PA)
and plasminogen activator inhibitor-1 (PAI-1) during surgery. Acta Anaesthesiol Scand
39(2):163–166
9 Intravenous Fluids and Coagulation 147
Evans PA, Glenn JR et al (1998) Effects of gelatin-based resuscitation fluids on platelet aggrega-
tion. Br J Anaesth 81(2):198–202
Evans PA, Heptinstall S et al (2003) Prospective double-blind randomized study of the effects of
four intravenous fluids on platelet function and hemostasis in elective hip surgery. J Thromb
Haemost 1(10):2140–2148
Fenger-Eriksen C, Tonnesen E et al (2009) Mechanisms of hydroxyethyl starch-induced dilutional
coagulopathy. J Thromb Haemost 7(7):1099–1105
Finfer S, Bellomo R et al (2004) A comparison of albumin and saline for fluid resuscitation in the
intensive care unit. N Engl J Med 350(22):2247–2256
Franz A, Braunlich P et al (2001) The effects of hydroxyethyl starches of varying molecular
weights on platelet function. Anesth Analg 92(6):1402–1407
Fries D, Martini WZ (2010) Role of fibrinogen in trauma-induced coagulopathy. Br J Anaesth
105(2):116–121
Fries D, Innerhofer P et al (2002) The effect of the combined administration of colloids and lac-
tated Ringer's solution on the coagulation system: an in vitro study using thrombelastograph
coagulation analysis (ROTEG. Anesth Analg 94(5):1280–1287
Fries D, Krismer A et al (2005) Effect of fibrinogen on reversal of dilutional coagulopathy: a por-
cine model. Br J Anaesth 95(2):172–177
Gamsjager T, Gustorff B et al (2002) The effects of hydroxyethyl starches on intracellular calcium
in platelets. Anesth Analg 95(4):866–869
Grottke O, Braunschweig T et al (2010) Effects of different fibrinogen concentrations on blood
loss and coagulation parameters in a pig model of coagulopathy with blunt liver injury. Crit
Care 14(2):R62
Guidet B, Soni N et al (2010) A balanced view of balanced solutions. Crit Care 14(5):325
Haas T, Fries D et al (2008a) Less impairment of hemostasis and reduced blood loss in pigs after
resuscitation from hemorrhagic shock using the small-volume concept with hypertonic saline/
hydroxyethyl starch as compared to administration of 4% gelatin or 6% hydroxyethyl starch
solution. Anesth Analg 106(4):1078–1086
Haas T, Fries D et al (2008b) The in vitro effects of fibrinogen concentrate, factor XIII and fresh
frozen plasma on impaired clot formation after 60% dilution. Anesth Analg 106(5):1360–1365
Hanke AA, Maschler S et al (2011) In vitro impairment of whole blood coagulation and platelet
function by hypertonic saline hydroxyethyl starch. Scand J Trauma Resusc Emerg Med 19:12
Hauser CJ, Shoemaker WC et al (1980) Oxygen transport responses to colloids and crystalloids in
critically ill surgical patients. Surg Gynecol Obstet 150(6):811–816
Innerhofer P, Fries D et al (2002) In vivo effect of haemodilution with saline on coagulation. Br J
Anaesth 89(6):934
Jacob M, Chappell D et al (2012) The intravascular volume effect of Ringer's lactate is below 20%:
a prospective study in humans. Crit Care 16(3):R86
James MF, Michell WL et al (2011) Resuscitation with hydroxyethyl starch improves renal func-
tion and lactate clearance in penetrating trauma in a randomized controlled study: the FIRST
trial (Fluids in Resuscitation of Severe Trauma). Br J Anaesth 107(5):693–702
Jamnicki M, Zollinger A et al (1998) Compromised blood coagulation: an in vitro comparison
of hydroxyethyl starch 130/0.4 and hydroxyethyl starch 200/0.5 using thrombelastography.
Anesth Analg 87(5):989–993
Jamnicki M, Bombeli T et al (2000) Low- and medium-molecular-weight hydroxyethyl starches:
comparison of their effect on blood coagulation. Anesthesiology 93(5):1231–1237
Jesty J (1986) The kinetics of inhibition of alpha-thrombin in human plasma. J Biol Chem
261(22):10313–10318
Jin SL, Yu BW (2010) Effects of acute hypervolemic fluid infusion of hydroxyethyl starch and
gelatin on hemostasis and possible mechanisms. Clin Appl Thromb Hemost 16(1):91–98
Johnson AL, Criddle LM (2004) Pass the salt: indications for and implications of using hypertonic
saline. Crit Care Nurse 24(5):36–38
Johnson SD, Lucas CE et al (1979) Altered coagulation after albumin supplements for treatment
of oligemic shock. Arch Surg 114(4):379–383
148 H. Schöchl et al.
Jones SB, Whitten CW et al (2003) The influence of crystalloid and colloid replacement solutions
in acute normovolemic hemodilution: a preliminary survey of hemostatic markers. Anesth
Analg 96(2):363–368
Jungheinrich C, Neff TA (2005) Pharmacokinetics of hydroxyethyl starch. Clin Pharmacokinet
44(7):681–699
Jungheinrich C, Sauermann W et al (2004) Volume efficacy and reduced influence on measures
of coagulation using hydroxyethyl starch 130/0.4 (6%) with an optimised in vivo molecular
weight in orthopaedic surgery : a randomised, double-blind study. Drugs R D 5(1):1–9
Kapiotis S, Quehenberger P et al (1994) Effect of hydroxyethyl starch on the activity of blood
coagulation and fibrinolysis in healthy volunteers: comparison with albumin. Crit Care Med
22(4):606–612
Karoutsos S, Nathan N et al (1999) Thrombelastogram reveals hypercoagulability after adminis-
tration of gelatin solution. Br J Anaesth 82(2):175–177
Kasper SM, Meinert P et al (2003) Large-dose hydroxyethyl starch 130/0.4 does not increase
blood loss and transfusion requirements in coronary artery bypass surgery compared with
hydroxyethyl starch 200/0.5 at recommended doses. Anesthesiology 99(1):42–47
Kermode JC, Zheng Q et al (1999) Marked temperature dependence of the platelet calcium signal
induced by human von Willebrand factor. Blood 94(1):199–207
Kheirabadi BS, Crissey JM et al (2008) Effects of synthetic versus natural colloid resuscita-
tion on inducing dilutional coagulopathy and increasing hemorrhage in rabbits. J Trauma
64(5):1218–1228
Kozek-Langenecker SA (2005) Effects of hydroxyethyl starch solutions on hemostasis.
Anesthesiology 103(3):654–660
Kozek-Langenecker SA (2009) Influence of fluid therapy on the haemostatic system of intensive
care patients. Best Pract Res Clin Anaesthesiol 23(2):225–236
Kulla M, Lampl L et al (2008) Hydroxyethyl starch 6% 130/0.42 in acetate-buffered Ringer's solution
as a part of a balanced-volume resuscitation in abdominal surgery. Anästhesiol Intensivmed 1:7–18
Langeron O, Doelberg M et al (2001) Voluven, a lower substituted novel hydroxyethyl starch (HES
130/0.4), causes fewer effects on coagulation in major orthopedic surgery than HES 200/0.5.
Anesth Analg 92(4):855–862
Laxenaire MC, Mertes PM (2001) Anaphylaxis during anaesthesia. Results of a two-year survey
in France. Br J Anaesth 87(4):549–558
Lucas CE, Ledgerwood AM et al (1982) Altered coagulation protein content after albumin resus-
citation. Ann Surg 196(2):198–202
Mardel SN, Saunders FM et al (1998) Reduced quality of clot formation with gelatin-based plasma
substitutes. Br J Anaesth 80(2):204–207
McIlroy DR, Kharasch ED (2003) Acute intravascular volume expansion with rapidly administered
crystalloid or colloid in the setting of moderate hypovolemia. Anesth Analg 96(6):1572–1577
Meng ZH, Wolberg AS et al (2003) The effect of temperature and pH on the activity of factor VIIa:
implications for the efficacy of high-dose factor VIIa in hypothermic and acidotic patients.
J Trauma 55(5):886–891
Mittermayr M, Streif W et al (2007) Hemostatic changes after crystalloid or colloid fluid admin-
istration during major orthopedic surgery: the role of fibrinogen administration. Anesth Analg
105(4):905–917
Mittermayr M, Streif W et al (2008) Effects of colloid and crystalloid solutions on endogenous
activation of fibrinolysis and resistance of polymerized fibrin to recombinant tissue plasmino-
gen activator added ex vivo. Br J Anaesth 100(3):307–314
Mortier E, Ongenae M et al (1997) In vitro evaluation of the effect of profound haemodilution with
hydroxyethyl starch 6%, modified fluid gelatin 4% and dextran 40 10% on coagulation profile
measured by thromboelastography. Anaesthesia 52(11):1061–1064
Myburgh J, Cooper DJ et al (2007) Saline or albumin for fluid resuscitation in patients with trau-
matic brain injury. N Engl J Med 357(9):874–884
Neff TA, Doelberg M et al (2003) Repetitive large-dose infusion of the novel hydroxyethyl starch
130/0.4 in patients with severe head injury. Anesth Analg 96(5):1453–1459
9 Intravenous Fluids and Coagulation 149
Ng KF, Lam CC et al (2002) In vivo effect of haemodilution with saline on coagulation: a random-
ized controlled trial. Br J Anaesth 88(4):475–480
Niemi TT, Silvanto M et al (2005) Albumin induced hypercoagulability does not reduce blood loss
in patients undergoing total hip arthroplasty. Scand J Surg 94(3):227–232
Niemi TT, Suojaranta-Ylinen RT et al (2006) Gelatin and hydroxyethyl starch, but not albumin,
impair hemostasis after cardiac surgery. Anesth Analg 102(4):998–1006
Perel P, Roberts I (2012) Colloids versus crystalloids for fluid resuscitation in critically ill patients.
Cochrane Database Syst Rev 6:CD000567
Perner A, Haase N et al (2012) Hydroxyethyl starch 130/0.42 versus Ringer's acetate in severe
sepsis. N Engl J Med 367(2):124–134
Petroianu GA, Liu J et al (2000) The effect of In vitro hemodilution with gelatin, dextran, hydroxy-
ethyl starch, or Ringer's solution on Thrombelastograph. Anesth Analg 90(4):795–800
Rehm M, Finsterer U (2003) Treating intraoperative hyperchloremic acidosis with sodium bicar-
bonate or tris-hydroxymethyl aminomethane: a randomized prospective study. Anesth Analg
96(4):1201–1208, table of contents
Roberts I, Blackhall K et al (2011) Human albumin solution for resuscitation and volume expan-
sion in critically ill patients. Cochrane Database Syst Rev (11):CD001208
Ruttmann TG (2002) Haemodilution enhances coagulation. Br J Anaesth 88(4):470–472
Ruttmann TG, James MF et al (1996) Haemodilution induces a hypercoagulable state. Br J
Anaesth 76(3):412–414
Ruttmann TG, Jamest MF et al (2001) Haemodilution-induced enhancement of coagulation is
attenuated in vitro by restoring antithrombin III to pre-dilution concentrations. Anaesth
Intensive Care 29(5):489–493
Ruttmann TG, Lemmens HJ et al (2006) The haemodilution enhanced onset of coagulation as
measured by the thrombelastogram is transient. Eur J Anaesthesiol 23(7):574–579
Scatchard G, Batchelder AC et al (1944) Chemical, Clinical, and Immunological Studies on the
Products of Human Plasma Fractionation. Vi. The Osmotic Pressure of Plasma and of Serum
Albumin. J Clin Invest 23(4):458–464
Schaden E, Wetzel L et al (2012) Effect of the carrier solution for hydroxyethyl starch on platelet
aggregation and clot formation. Br J Anaesth 109(4):572–577
Schierhout G, Roberts I (1998) Fluid resuscitation with colloid or crystalloid solutions in critically
ill patients: a systematic review of randomised trials. BMJ 316(7136):961–964
Schlimp CJ, Cadmoro J et al (2013) The effect of fibrinogen concentrate and factor XIII on throm-
boelastometry in 33 % diluted blood with albumin, gelatin, hydroxyethyl starch or saline
in vitro. Blood Transfus 11(4):510–517
Schramko AA, Kuitunen AH et al (2009) Role of fibrinogen-, factor VIII- and XIII-mediated clot
propagation in gelatin haemodilution. Acta Anaesthesiol Scand 53(6):731–735
Schramko A, Suojaranta-Ylinen R et al (2010) Hydroxyethylstarch and gelatin solutions impair
blood coagulation after cardiac surgery: a prospective randomized trial. Br J Anaesth
104(6):691–697
Smith GJ, Kramer GC et al (1985) A comparison of several hypertonic solutions for resuscitation
of bled sheep. J Surg Res 39(6):517–528
Sorensen B, Fries D (2012) Emerging treatment strategies for trauma-induced coagulopathy. Br J
Surg 99(Suppl 1):40–50
Sossdorf M, Marx S et al (2009) HES 130/0.4 impairs haemostasis and stimulates pro-inflammatory
blood platelet function. Crit Care 13(6):R208
Stogermuller B, Stark J et al (2000) The effect of hydroxyethyl starch 200 kD on platelet function.
Anesth Analg 91(4):823–827
Strauss RG, Pennell BJ et al (2002) A randomized, blinded trial comparing the hemostatic effects
of pentastarch versus hetastarch. Transfusion 42(1):27–36
Tabuchi N, de Haan J et al (1995) Gelatin use impairs platelet adhesion during cardiac surgery.
Thromb Haemost 74(6):1447–1451
Thaler U, Deusch E et al (2005) In vitro effects of gelatin solutions on platelet function: a compari-
son with hydroxyethyl starch solutions. Anaesthesia 60(6):554–559
150 H. Schöchl et al.
10.1 Plasma
Plasma is the acellular fraction of blood, separated from the cellular blood compo-
nents either by centrifugation of citrated whole blood or donor apheresis, with typi-
cal unit volumes averaging from 200 to 300 mL. There are several types of plasma
products used for supplementation or replacement of soluble coagulation factors.
The most widely recognized plasma component is fresh frozen plasma (FFP), which
requires freezing at −18 °C within 6–8 h of collection (AABB 2013a). Plasma fro-
zen within 24 h after phlebotomy (FP24) is similar to FFP in its preparation but
differs in that it may be frozen at or below −18 C within 24 h after collection. Frozen
plasma products prepared for transfusion requires thawing and warming to between
30 and 37 C. This takes 20–30 min, depending upon the equipment used for thawing
and the unit volume. If the prepared plasma is not transfused within the initial 24-h
post-thaw period, it can be relabeled as “thawed plasma” for use within 5 days after
T.M. Boyd, MD
American University of the Caribbean School of Medicine,
Medical Sciences Campus, #1 University Drive at Jordan Road, Cupecoy, Dutch Lowlands,
St. Maarten
e-mail: [email protected]
E. Lockhart, MD
Department of Pathology, Clinical Director of Transfusion Medicine, Duke University Health
System, Rm 1720, Duke North, DUMC 2928, Durham, NC 27710, USA
e-mail: [email protected]
I. Welsby, MBBS, FRCA (*)
Duke University, 2301 Erwin Rd, 3094, Durham, NC 27710, USA
e-mail: [email protected]
the initial thaw (AABB 2013a; Benjamin and McLaughlin 2012; Eder and Sebok
2007). The utility of thawed plasma is twofold: (1) it provides rapidly available
plasma for the management of massive hemorrhage, and (2) it extends available
plasma inventory by avoiding wastage of plasma suitable for transfusion (Downes
et al. 2001). One rarely used plasma product, available in the USA, is liquid plasma.
Unlike the previously described plasma products, liquid plasma has never been fro-
zen; it is separated from a whole blood unit no later than 5 days before its expiration
date and has a shelf life of 26 days from the date of the source blood collection
(40 days if CPDA-1 is used as the anticoagulant for the parent component) (AABB
2013a; Benjamin and McLaughlin 2012).
Forms of pathogen-reduced plasma are widely available in Europe but until
recently have not been available in the USA. Solvent/detergent-treated plasma (S/D
plasma) is one of the most frequently used pathogen-reduced plasma products.
Pooled plasma composed of a given ABO type undergoes viral inactivation with
solvents (i.e., 1 % tri(n-butyl) phosphate) and detergents (i.e., 1 % Triton-X 100),
which are subsequently extracted by oil and affinity ligand chromatography for
selective binding of prion proteins (PrPSc). This treatment significantly inactivates
lipid-enveloped viruses such as HIV and Hepatitis C as well as cellular pathogens
such as bacteria and protozoa. While the pathogen inactivation process is ineffective
against non-lipid-enveloped viruses such as hepatitis A (HAV) and parvovirus B19,
product specifications identify minimum levels of B19 and hepatitis E virus (HEV)
genetic material permissible, as well as minimum levels of neutralizing antibodies
directed against HAV and B19 (Benjamin and McLaughlin 2012). This product had
not been approved for use in the USA until January 2013, when the FDA approved
Octaplas® (Octapharma, Austria) for transfusion (FDA 2013; Riedler et al. 2003;
Ozier et al. 2011; Sachs et al. 2005; Sinnott et al. 2004).
Methylene blue (MB)-treated plasma is another pathogen-reduced product
available in Europe. Methylene blue is an aniline dye which, upon light activation,
generates reactive oxygen species that inactivate enveloped and some nonenvel-
oped viruses, with lesser activity directed against protozoa and bacteria (Benjamin
and McLaughlin 2012). Unlike S/D plasma, MB plasma is a single donor
component.
Plasma contains ABO isoagglutinins, the naturally occurring antibodies directed
against ABO antigens, and therefore must be ABO compatible with the recipient’s
red cells (see Table 10.1); however, cross-matching is not required. Group AB
plasma, which lacks antibodies directed against A and B antigens, is compatible
with all blood types and is used as emergency release plasma when there is insuffi-
cient time for determining the recipient’s blood type (Wehrli et al. 2009). The Rh
(D) type is not always matched because immunization to Rh (D) antigen has rarely
been reported as a result of transfusion of Rh (D)-positive plasma to Rh (D)-negative
individuals. Hemolytic reactions as a consequence of infusion of an undetected anti-
body directed toward recipient RBC antigens are rarely seen, as is alloimmunization
to RBC antigens (Ching et al. 1991).
10 Split Blood Products 153
10.1.2 Indications
Notable variation exists in published guidelines for plasma transfusion (ASA 2006;
Ferraris et al. 2011; Iorio et al. 2008; O’Shaughnessy et al. 2004; Fuller and Bucklin
2010; Roback et al. 2010), although there is agreement on its indication for replace-
ment of coagulation factors in bleeding or surgical patients, particularly those suf-
fering from disseminated intravascular coagulation (DIC) or undergoing massive
transfusion. Many guidelines recommend using the international normalized ratio
(INR) at or greater than 1.5 as an indication for plasma transfusion (O’Shaughnessy
et al. 2004). However, a consensus on laboratory value “triggers” for plasma trans-
fusion has not been reached. For massive hemorrhage, empiric transfusion of plasma
in set ratios to RBC units is widely practiced in the USA (Holcomb et al. 2013; Dzik
et al. 2011). Retrospective and observational studies on massive transfusion in mili-
tary and civilian trauma have reported associations between higher plasma/RBC
transfusion ratios and improved survival (Holcomb et al. 2013; Borgman et al. 2007;
Shaz and Hillyer 2010), although definitive data on optimal use of plasma in this
setting has yet to emerge (Holcomb et al. 2013).
Plasma is also indicated as a replacement fluid in therapeutic apheresis for throm-
botic thrombocytopenia purpura (TTP) (Szczepiorkowski et al. 2010) and for coagu-
lation factor replacement in patients with congenital deficiencies of single factors
(such as FV and FXI), but should not be used for factor replacement in congenital
factor deficiencies if factor concentrates are readily available (i.e., FVIII concen-
trates in hemophilia A patients). Additional indications include rapid warfarin rever-
sal in an actively bleeding patient, although guidelines are emerging preferentially
recommending prothrombin complex concentrates as a first-line agent (Ageno et al.
2012; Keeling et al. 2011; Morgenstern et al. 2010; Ansell et al. 2008). Plasma is not
appropriate to use as a nutritional supplement or as a source of immunoglobulin and
154 T.M. Boyd et al.
should not be used as a volume expander when blood volume can safely and ade-
quately be replaced with other agents such as colloids (AABB 2013).
Table 10.2 Average values of coagulation factors found in different plasma and cryoprecipitate
preparations
Thawed Thawed
plasma from plasma from Cryo from FP24
FFP (Downes FP24 (Yazer (cryo24) (Yazer Liquid plasma
Factors et al. 2001) et al. 2008) et al. 2010) (Gosselin et al. 2013)
Ref. range
(U/mL) Day 1 Day 5 Day 1 Day 5 Standard Cryo24 Day 1 Day 15 Day 30
Factor V 0.70 0.66 1.40 0.87 1.10 0.77 0.50
(0.70–1.50)
Factor VII 0.90 0.72 1.09 0.96 0.97 0.78 1.08
(0.60–1.60)
VWF:Ag 448.1 505.9 0.73 0.50 0.40
(340–820)
Factor VIII 1.07 0.63 0.60 0.69 2.16 2.52 0.72 0.56 0.50
(0.60–1.50)
FX 0.85 0.80 1.10 1.11 1.12
FIX (0.60–1.50) 1.20 1.26 0.86 0.84 0.76
Antithrombin III 0.89 0.92
(0.80–1.20)
Protein C 1.05 0.96 0.88 0.89 0.86
(0.70–1.40)
Protein S 0.70 0.52 0.90 0.91
(0.58–1.28)
Protein S activity 0.90 0.48 0.22
(0.76–1.35 IU/
mL)
Fibrinogen 225 225 320 318 455.8 575.8 2.92 2.76 2.75
(150–350 mg/dL)
Further details including the variability of these levels can be found in the individual references
(Downes et al. 2001; Yazer et al. 2008; Gosselin et al. 2013; Yazer et al. 2010)
as compared with FFP; as such, the use of S/D plasma is not recommended for use
in patients with severe protein S deficiency (Theusinger et al. 2011).
MB-treated plasma, however, may have markedly reduced hemostatic (Pock
et al. 2007) and antithrombotic (Alvarez-Larran et al. 2004) efficacy; other photo-
chemical pathogen reduction treatments of plasma, such as amotosalen, are also
being evaluated and appear more equivalent to FFP (de Alarcon et al. 2005; de
Valensart et al. 2009; Mintz et al. 2006a, b).
Platelets are small (2–3 μm in diameter) anucleate cell fragments which bind to sites
of injury, providing the phospholipid surface for coagulation enzymes to assemble
and generate thrombin (Hoffman and Monroe 2001). In addition, they contribute
key protein and molecular elements for fibrin clot formation as well as exerting a
contractile force that draws together the margins of injury. Platelet concentrates are
obtained either from whole blood or by collection from donors via apheresis.
Platelet concentrates derived from a single 450–500 mL whole blood collection
contain greater than 5.5 × 1010 platelets per unit, with a typical adult dose formed by
pooling 4–6 concentrates (AABB 2013a). Apheresis platelets contain over 3 × 1011
platelets per unit, and contrary to other platelet concentrates, they have the advan-
tage that they represent only a single donor exposure per transfusion, reducing the
risk of transfusion-transmitted infections. While some in vitro differences have been
observed between platelets derived from the different collection methods
(Vasconcelos et al. 2003), these have little clinical consequence, and these products
are interchangeable (Slichter 2007; Chambers and Herman 1999).
One notable characteristic of platelet components is their cold intolerance. The
exposure of platelet concentrates to temperatures of 4 °C or less results in platelet
shape change, functional defects, and increased circulatory clearance rates
(Hoffmeister et al. 2003; Rao and Murphy 1982). Platelets also have the shortest
shelf life of any transfused product: the time from collection to expiration is 5 days;
attempts to extend the approved storage time have failed (Dumont et al. 2010). This
is due in part to the relatively short functional life of platelets (7–10 days in the
circulation) but also to the risks of bacterial proliferation due to storage at room
temperature (Palavecino et al. 2010).
158 T.M. Boyd et al.
bleeding time was shown to steadily increase below this level (Slichter and
Harker 1978).
Pathogen-inactivated, photochemically treated (PCT) platelets offer the promise
to minimize transfusion-related infection with a broad range recognized and emerg-
ing bacteria, viruses, and protozoa (McCullough et al. 2004). Synthetic psoralens
intercalate with microbial DNA or RNA and, upon exposure to ultraviolet light,
cross-link pyrimidine bases to prevent microbial replication. While equivalent
hemostatic efficacy was seen in clinical trials of standard versus PCT platelets
(McCullough et al. 2004; Cid et al. 2012), the in vivo recovery of PCT platelets was
lower and others dispute their efficacy (Kerkhoffs et al. 2010). PCT platelets are
approved for use in Europe, but not in the USA.
Platelets require a supportive milieu derived from the plasma they are stored in;
most apheresis units are stored in 100 % plasma with ACD-A anticoagulant.
Reduction of plasma volume allows diversion of the plasma for other uses and may
reduce the risk of plasma-associated TRALI occurring on transfusion of plasma-
containing platelets. The use of platelet additive solutions (PAS) such as InterSol®
(Fenwal Inc., Zurich, IL) allows plasma reduction to be tolerated by supplementing
electrolytes and buffers. PAS platelets demonstrate in vivo recovery and survival
that exceed FDA requirements (van der Meer et al. 2010; Dumont et al. 2013).
Future developments in available platelet products include reconstituted, cryo-
preserved platelets. The in vivo survival of transfused thawed, resuspended, cryo-
preserved platelets met FDA criteria (Dumont et al. 2013), and in vivo hemostatic
activity appears adequate (Khuri et al. 1999). However, while the relevance to
hemostasis is unclear, adequate 24-h in vivo recovery remains a regulatory hurdle.
While fresh whole blood may be viewed as ideal treatment for trauma resuscitation,
there are several logistical and functional limitations. First, it would limit availabil-
ity of other blood components, and second, if not used when fresh, how long could
it be stored and how rapidly do storage lesions develop? The longer whole blood
remains in storage, the more platelets and white cells aggregate in the product,
increasing the risks of adverse reactions following transfusion. Erythrocytes pro-
mote aggregation and activation of platelets within the product, thereby defeating
one of the therapeutic benefits of using whole blood. Although some in vitro mea-
sures support the hemostatic function for whole blood refrigerated for up to 21 days
(Pidcoke et al. 2013). Many issues would have to be addressed before whole blood
can be licensed by the FDA and readily available for transfusion.
scope of this chapter, particularly the infectious complications, but will be reviewed
briefly.
All blood components bear the risk of transmitting infectious diseases, despite care-
ful screening of blood donors and (in many countries) universal testing of the blood
supply for infectious disease markers. Pooled products such as pooled platelets or
cryoprecipitate, which have not undergone pathogen inactivation, present increased
risks due to the multiple donor exposures. Existing and emerging infectious agents
are of greatest local or regional importance in endemic areas, but international travel
increases exposure to pathogens. If all pathogens are not included in existing screen-
ing mechanisms in a traveler’s native country, travel may increasingly limit suitable
blood donors. Rigorous donor screening, serologic testing, and nucleic acid ampli-
fication testing have proven extremely effective in reducing the risk of transfusion-
transmitted infections in developed nations (Bowden and Sayers 1990), but
transfusion-transmitted infection remains a serious concern throughout the develop-
ing world.
In 2009, the American Association of Blood Banks Transfusion-Transmitted
Diseases Committee made up of volunteer members with expertise in infectious
disease convened to publish a supplement to the journal Transfusion on the threat to
the North American blood supply from emerging infectious diseases (Stramer et al.
2009). They prioritized specific agents into categories: red agents had the highest
priority, followed by orange, yellow, and white. For further understanding of the
classification system, readers are directed to the reference (Stramer et al. 2009).
Red agents include human variant Creutzfeldt-Jakob disease, dengue viruses,
and Babesia species. Orange agents include Chikungunya virus, St. Louis encepha-
litis virus, Leishmania species, Plasmodium species, and T. cruzi. Yellow agents
include chronic wasting disease prions, human herpes virus 8, HIV variants, human
parvovirus B19, influenza A virus subtype H5N1, simian foamy virus, Borrelia
burgdorferi, and hepatitis A virus. White agents include hepatitis E and Anaplasma
phagocytophilum (Stramer et al. 2009).
Acute hemolytic transfusion reactions are one of the most serious complications of
transfusion and remain as one of the leading causes of transfusion-related mortality
worldwide (Eder and Chambers 2007; Vamvakas and Blajchman 2010). These
reactions result from RBC lysis or accelerated clearance by the reticuloendothelial
system resulting from RBC transfusion into a recipient with preformed antibodies
directed against donor erythrocytes. Rarely, plasma-rich blood products have been
implicated in hemolytic reactions by passive transfer of antibodies directed against
recipient erythrocytes (Fung et al. 2007). Antibodies directed against ABO antigens
are the most frequent source of incompatibility (Ching et al. 1991; Josephson et al.
2010), for example, transfusion of O platelets containing anti-A and anti-B. Pre-
transfusion plasma reduction and ABO matching easily avoid this complication.
transfusion but may present toward the end of the transfusion process or even within
1–2 h afterward due to the increasing level of cytokine exposure. The diagnosis of
FNHTR is one of exclusion, having ruled out other causes of febrile reactions such
as hemolytic transfusion reactions, septic reactions, or contributions from comor-
bidities or medications. Treatment is supportive, including antipyretics such as acet-
aminophen (Heddle 2007).
Allergic transfusion reactions are one of the most common adverse transfusion reac-
tions, with incidence rates estimated between 1 and 3 % (Vamvakas 2007; Domen
and Hoeltge 2003). Clinical presentation varies, but most manifests solely with
cutaneous symptoms such as urticaria, pruritus, erythema, and angioedema (Domen
and Hoeltge 2003). These minor allergic reactions are thought to be most often
mediated by preexisting recipient IgE or IgG targeting plasma protein antigens.
Passive transfer of IgE directed against recipient plasma allergens or other environ-
mental allergens has also been observed. Accordingly, allergic reactions occur most
frequently with plasma-rich products (including platelets) but may also occur in
plasma-deplete products such as red cell units. Antihistamines such as diphenhydr-
amine or famotidine form the cornerstone for treatment of minor allergic transfu-
sion reactions with corticosteroids reserved for more severe reactions.
Rarely, severe allergic reactions may rapidly progress to anaphylactic shock within
minutes after symptom onset. These reactions are characterized by bronchospasm,
hypotension, nausea and vomiting, chest pain, and tachycardia. IgA-deficient patients
who have developed class-specific anti-IgA are at risk; however, this group only
represents a fraction of anaphylactic transfusion reactions (AABB 2013a). Causative
agents vary widely from anti-haptoglobin antibodies to passive transfer of allergens to
which a patient is already sensitized, such as recently ingested foods (i.e., peanuts)
(Jacobs et al. 2011). Ultimately, anaphylaxis is an unpredictable and potentially fatal
transfusion outcome requiring swift action to prevent an adverse outcome. Severe
allergic reactions or anaphylaxis should be managed in a similar fashion to anaphy-
laxis from other causes, with administration of epinephrine (with or without other
vasopressors as needed) and corticosteroids, maintenance of a patent airway, and vol-
ume infusion to maintain hemodynamic stability. Testing for associated DIC and post-
poning procedures requiring further transfusion should also be considered.
10.4.6.1 Pathophysiology
Transfusion of plasma-containing blood products—which include all blood products
other than washed cellular blood products—may result in a syndrome of non-
cardiogenic pulmonary edema and acute respiratory distress. Clinical findings defining
TRALI include (1) onset during or within 6 hours of transfusion; (2) severe hypoxemia,
10 Split Blood Products 163
such as less than 90 % oxygen saturation on room air; (3) diffuse bilateral pulmonary
infiltrates on chest x-ray; (4) absence of volume overload; and (5) no preexisting acute
lung injury (Kleinman et al. 2004). TRALI may also be associated with fever, chills,
hypotension, and transient leukopenia. The primary pathophysiologic mechanism is
believed to be a reaction between donor anti-leukocyte antibodies and recipient leuko-
cytes, which results in leukocyte activation (Marques et al. 2005), sequestration, and
infiltration into the pulmonary capillary bed (Fung and Silliman 2009). Leukocyte acti-
vation results in pulmonary microvascular injury and capillary leakage with an influx
of proteinaceous fluid into the alveolar space (Bux and Sachs 2007). A “two-hit”
hypothesis for the pathogenesis of TRALI holds that the first hit is due to recipient
neutrophils primed for activation by virtue of the patient’s underlying clinical condi-
tion. The second hit involves activation of these neutrophils by anti-leukocyte antibod-
ies or biological response modifiers contained in the transfused product (Silliman
2006). In rare cases, the transfused product may provide both hits (Kelher et al. 2009).
Female donors sensitized to human leukocyte antigens (HLA) by pregnancy are
most frequently implicated as the source of blood products which have been linked to
TRALI cases (Densmore et al. 1999; Powers et al. 2008; Triulzi et al. 2009). The
frequency of anti-HLA antibodies ranges from 1.7 % for never pregnant females to
32.2 % for four or more pregnancies, whereas males and previously transfused donors
all showed very low frequency of anti-HLA antibodies (in the range of 1–2 %) (Triulzi
et al. 2009; Kakaiya et al. 2010). As a result, many blood collection agencies in the
USA and Europe limit or prohibit collection of plasma-rich blood products from
female donors (Eder et al. 2010; Funk et al. 2012; Jutzi et al. 2008; Keller-Stanislawski
et al. 2010; Lucas et al. 2012; Middelburg et al. 2010; van Stein et al. 2010).
The best available, current TRALI incidence estimate comes from a prospective
study surveilling hypoxemia after transfusion of over 450,000 units between 2006
and 2009. Ninety-one TRALI cases were identified with an incidence of 1 in
3,141 in 2006 compared with 1 in 12,346 in 2009 after the introduction of gender-
based mitigation strategies (Toy et al. 2012).
10.4.6.3 Prevention
It has been well established that donors implicated in TRALI cases are more likely
to be female and multiparous (Toy et al. 2012; Gajic et al. 2007b). While not all
studies support the benefit of avoiding female plasma (Welsby et al. 2010), the over-
whelming evidence supports the implementation of gender-based policies for reduc-
ing TRALI incidence from plasma transfusion. These factors resulted in the
implementation of a new TRALI risk mitigation policy during the mid- to late 2000s
throughout most of Europe and the USA, in which plasma units were predominantly
obtained from male donors, thereby avoiding the transfusion of plasma units from
female donors. This policy is feasible for plasma transfusion because the number of
plasma units collected from whole blood or apheresis collections meets, or is in
excess of, demand. In contrast, this policy is challenging for group AB platelet and
plasma components (Reesink et al. 2012), where restriction of units to male donors
jeopardizes supply (see Table 10.3). Despite that, in 2013 the AABB announced
new TRALI risk mitigation standards requiring high-plasma volume components
come from males, females who have not been pregnant, or females who have been
tested since their most recent pregnancy to rule out the presence of anti-HLA anti-
bodies, regardless of ABO group. These new standards are to go into effect in 2014
(AABB 2013b).
Some blood centers have implemented a policy of testing selected populations of
platelet apheresis donors for anti-HLA antibodies or resuspending apheresis plate-
lets in platelet additive solution (PAS) (Lucas et al. 2012; Reesink et al. 2012;
Kleinman et al. 2010) although there are inadequate data to evaluate its effect on the
incidence of TRALI following platelet transfusion (Kakaiya et al. 2010; Reesink
et al. 2012).
10 Split Blood Products 165
Table 10.3 Following implementa- Component Cases (n) TRALI rates per 106
tion of the American Red Cross
A, B, O plasma 9 1.9
TRALI mitigation strategies, the
AB plasma 12 24.9
incidence of TRALI cases 2008–2010
fell for all but group AB plasma RBC 39 2.2
where female donors still comprise Apheresis platelets 19 8
40 % of the group AB donor pool Adapted from Reesink et al. (2012)
10.4.7.1 Pathophysiology
Transfusion-associated circulatory overload (TACO) will cause transfusion-related
respiratory insufficiency and is thought to occur when the rate of transfusion exceeds
the recipient’s cardiovascular adaption to the additional workload. The rapid infu-
sion of excessive volume can result in dyspnea, hypoxemia, elevated central venous
pressure, and pulmonary edema (Eder and Chambers 2007).
TACO is typically reported in elderly patients and small children, due to their
relatively small circulating volume but can occur in all age ranges. Compromised
cardiac function, positive fluid balance, and rapid blood product administration are
additional risk factors for TACO, which appears to occur more frequently in opera-
tive or intensive care settings, where large fluid volumes and blood are administered
(Li et al. 2011).
While the primary mechanism of TACO centered around fluid overload
(Popovsky 2004; Li et al. 2010), this has recently been questioned as the median
transfusion volume in patients who develop TACO is only 3 (2–7) units (Li et al.
2011) and a large proportion of reported TACO cases occur after a single blood unit
exposure (Popovsky et al. 1996). Similarly, Roubinian et al. reported no statistically
significant differences in hourly fluid balance or the number of blood component
units transfused in the 24-h interval preceding the TACO or TRALI episode
(Roubinian et al. 2012).
TACO is also typically associated with increased systemic blood pressure
(Popovsky 2010; Klein and Anstee 2006), which exceeds that expected from the
166 T.M. Boyd et al.
As all blood products are collected and stored in citrate-based anticoagulants, large
volume transfusions may be complicated by hypocalcemia (Eder and Chambers
2007). Citrate binds divalent cations such as calcium and magnesium and is rapidly
metabolized by the liver. Whereas citrate is easily cleared during nonurgent transfu-
sions, citrate load during massive transfusion may overwhelm this clearance mecha-
nism (Sihler and Napolitano 2010). In the awake patient, hypocalcemia presents
initially with chills, tingling, dizziness, and tetany; continued progression of citrate
toxicity can lead to prolonged QT interval, decreased left ventricular function, and
cardiac arrhythmias. While hypocalcemia can be managed by slowing the rate of
transfusion, in ongoing massive transfusion, and in patients with liver dysfunction
or under general anesthesia, calcium replacement therapy should be guided by the
patient’s ionized calcium concentration.
10 Split Blood Products 167
References
AABB (2013) Circular of information for the use of human blood and blood components. In:
Cross AR (ed) Bethesda. http://www.aabb.org/resources/bct/Documents/coi_ct1013.pdf. Last
accessed 19 May 2014
AABB (2013) AABB Revises and Clarifies TRALI Risk Reduction Requirements. http://www.aabb.
org/sa/standards/Pages/revised-trali-risk-reduction-requirements.aspx. Accessed 1 Oct 2013
Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G (2012) Oral anticoagu-
lant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College
of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 141:e44S–e88S
Alvarez-Larran A, Del Rio J, Ramirez C, Albo C, Pena F, Campos A, Cid J, Muncunill J, Sastre
JL, Sanz C, Pereira A (2004) Methylene blue-photoinactivated plasma vs. fresh-frozen plasma
as replacement fluid for plasma exchange in thrombotic thrombocytopenic purpura. Vox Sang
86:246–251
Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G (2008) Pharmacology and man-
agement of the vitamin K antagonists: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines (8th Edition). Chest 133:160S–198S
ASA (2006) Practice guidelines for perioperative blood transfusion and adjuvant therapies: an
updated report by the American Society of Anesthesiologists Task Force on Perioperative
Blood Transfusion and Adjuvant Therapies. Anesthesiology 105:198–208
ASA. American Society of Anesthesiologists practice guidelines for perioperative blood transfu-
sion and adjuvant therapies. http://www.asahq.org/publicationsAndServices/practiceparam.
htm#blood. Accessed 1 Oct 2013
Benjamin RJ, McLaughlin LS (2012) Plasma components: properties, differences, and uses.
Transfusion 52(Suppl 1):9S–19S
Blajchman MA, Goldman M (2001) Bacterial contamination of platelet concentrates: incidence,
significance, and prevention. Semin Hematol 38:20–26
Borgman MA, Spinella PC, Perkins JG, Grathwohl KW, Repine T, Beekley AC, Sebesta J, Jenkins
D, Wade CE, Holcomb JB (2007) The ratio of blood products transfused affects mortality in
patients receiving massive transfusions at a combat support hospital. J Trauma 63:805–813
Bowden R, Sayers M (1990) The risk of transmitting cytomegalovirus infection by fresh frozen
plasma. Transfusion 30:762–763
Brecher ME, Hay SN (2005) Bacterial contamination of blood components. Clin Microbiol Rev
18:195–204
British Committee for Standards in Haematology. Guidelines for the use of prophylactic anti-D
immunoglobulin. www.bcshguidelines.com/documents/Anti-D_bcsh_07062006.pdf. Accessed
9 Feb 2012
168 T.M. Boyd et al.
Eder AF, Sebok MA (2007) Plasma components: FFP, FP24, and thawed plasma. Immunohematology
23:150–157
Eder AF, Herron RM Jr, Strupp A, Dy B, White J, Notari EP, Dodd RY, Benjamin RJ (2010)
Effective reduction of transfusion-related acute lung injury risk with male-predominant plasma
strategy in the American Red Cross (2006–2008). Transfusion 50:1732–1742
Ely EW, Smith AC, Chiles C, Aquino SL, Harle TS, Evans GW, Haponik EF (2001) Radiologic
determination of intravascular volume status using portable, digital chest radiography: a pro-
spective investigation in 100 patients. Crit Care Med 29:1502–1512
FDA. FDA approves octaplas to treat patients with blood clotting disorders. http://www.fda.gov/
NewsEvents/Newsroom/PressAnnouncements/ucm336009.htm. Accessed 17 Jan 2013
Ferraris VA, Brown JR, Despotis GJ, Hammon JW, Reece TB, Saha SP, Song HK, Clough ER,
Shore-Lesserson LJ, Goodnough LT, Mazer CD, Shander A, Stafford-Smith M, Waters J,
Baker RA, Dickinson TA, FitzGerald DJ, Likosky DS, Shann KG (2011) 2011 update to the
Society of Thoracic Surgeons and the Society of Cardiovascular Anesthesiologists blood con-
servation clinical practice guidelines. Ann Thorac Surg 91:944–982
Flamholz R, Jeon HR, Baron JM, Baron BW (2000) Study of three patients with thrombotic
thrombocytopenic purpura exchanged with solvent/detergent-treated plasma: is its decreased
protein S activity clinically related to their development of deep venous thromboses? J Clin
Apher 15:169–172
Franchini M, Lippi G (2012) Fibrinogen replacement therapy: a critical review of the literature.
Blood Transfus 10:23–27
Fuller AJ, Bucklin BA (2010) Blood product replacement for postpartum hemorrhage. Clin Obstet
Gynecol 53:196–208
Fung YL, Silliman CC (2009) The role of neutrophils in the pathogenesis of transfusion-related
acute lung injury. Transfus Med Rev 23:266–283
Fung MK, Downes KA, Shulman IA (2007) Transfusion of platelets containing ABO-incompatible
plasma: a survey of 3156 North American laboratories. Arch Pathol Lab Med 131:909–916
Funk MB, Guenay S, Lohmann A, Henseler O, Heiden M, Hanschmann KM, Keller-Stanislawski
B (2012) Benefit of transfusion-related acute lung injury risk-minimization measures–German
haemovigilance data (2006–2010). Vox Sang 102:317–323
Gajic O, Gropper MA, Hubmayr RD (2006) Pulmonary edema after transfusion: how to differenti-
ate transfusion-associated circulatory overload from transfusion-related acute lung injury. Crit
Care Med 34:S109–S113
Gajic O, Rana R, Winters JL, Yilmaz M, Mendez JL, Rickman OB, O’Byrne MM, Evenson LK,
Malinchoc M, DeGoey SR, Afessa B, Hubmayr RD, Moore SB (2007a) Transfusion-related
acute lung injury in the critically ill: prospective nested case-control study. Am J Respir Crit
Care Med 176:886–891
Gajic O, Yilmaz M, Iscimen R, Kor DJ, Winters JL, Moore SB, Afessa B (2007b) Transfusion
from male-only versus female donors in critically ill recipients of high plasma volume compo-
nents. Crit Care Med 35:1645–1648
Goldberg AD, Kor DJ (2012) State of the art management of transfusion-related acute lung injury
(TRALI). Curr Pharm Des 18:3273–3284
Gosselin RC, Marshall C, Dwyre DM, Gresens C, Davis D, Scherer L, Taylor D (2013) Coagulation
profile of liquid-state plasma. Transfusion 53:579–590
Heddle N (2007) Febrile nonhemolytic transfusion reactions. In: Transfusion Reactions, 3rd edn.
AABB Press, Bethesda
Hedges SJ, Dehoney SB, Hooper JS, Amanzadeh J, Busti AJ (2007) Evidence-based treatment
recommendations for uremic bleeding. Nat Clin Pract Nephrol 3:138–153
Heger A, Romisch J, Svae TE (2006) A biochemical comparison of a pharmaceutically licensed
coagulation active plasma (Octaplas) with a universally applicable development product
(Uniplas) and single-donor FFPs subjected to methylene-blue dye and white-light treatment.
Transfus Apher Sci 35:223–233
Hillyer CD, Josephson CD, Blajchman MA, Vostal JG, Epstein JS, Goodman JL (2003) Bacterial
contamination of blood components: risks, strategies, and regulation: joint ASH and AABB
170 T.M. Boyd et al.
recipient safety: an illustration of the need for proper risk assessments. Transfusion
49:903–912
Kleinman S, Grossman B, Kopko P (2010) A national survey of transfusion-related acute lung
injury risk reduction policies for platelets and plasma in the United States. Transfusion
50:1312–1321
Kopko PM, Popovsky MA, MacKenzie MR, Paglieroni TG, Muto KN, Holland PV (2001) HLA
class II antibodies in transfusion-related acute lung injury. Transfusion 41:1244–1248
Kopko PM, Paglieroni TG, Popovsky MA, Muto KN, MacKenzie MR, Holland PV (2003) TRALI:
correlation of antigen-antibody and monocyte activation in donor-recipient pairs. Transfusion
43:177–184
Kozek-Langenecker SA, Afshari A, Albaladejo P, Santullano CA, De Robertis E, Filipescu DC,
Fries D, Gorlinger K, Haas T, Imberger G, Jacob M, Lance M, Llau J, Mallett S, Meier J, Rahe-
Meyer N, Samama CM, Smith A, Solomon C, Van der Linden P, Wikkelso AJ, Wouters P,
Wyffels P (2013) Management of severe perioperative bleeding: guidelines from the European
Society of Anaesthesiology. Eur J Anaesthesiol 30:270–382
Li G, Daniels CE, Kojicic M, Krpata T, Wilson GA, Winters JL, Moore SB, Gajic O (2009) The
accuracy of natriuretic peptides (brain natriuretic peptide and N-terminal pro-brain natriuretic)
in the differentiation between transfusion-related acute lung injury and transfusion-related cir-
culatory overload in the critically ill. Transfusion 49:13–20
Li G, Kojicic M, Reriani MK, Fernandez Perez ER, Thakur L, Kashyap R, Van Buskirk CM, Gajic
O (2010) Long-term survival and quality of life after transfusion-associated pulmonary edema
in critically ill medical patients. Chest 137:783–789
Li G, Rachmale S, Kojicic M, Shahjehan K, Malinchoc M, Kor DJ, Gajic O (2011) Incidence and
transfusion risk factors for transfusion-associated circulatory overload among medical inten-
sive care unit patients. Transfusion 51:338–343
Lucas G, Win N, Calvert A, Green A, Griffin E, Bendukidze N, Hopkins M, Browne T, Poles A,
Chapman C, Massey E (2012) Reducing the incidence of TRALI in the UK: the results of screening
for donor leucocyte antibodies and the development of national guidelines. Vox Sang 103:10–17
Marques MB, Tuncer HH, Divers SG, Baker AC, Harrison DK (2005) Acute transient leukopenia
as a sign of TRALI. Am J Hematol 80:90–91
McCullough J, Steeper TA, Connelly DP, Jackson B, Huntington S, Scott EP (1988) Platelet utili-
zation in a university hospital. JAMA 259:2414–2418
McCullough J, Vesole DH, Benjamin RJ, Slichter SJ, Pineda A, Snyder E, Stadtmauer EA, Lopez-
Plaza I, Coutre S, Strauss RG, Goodnough LT, Fridey JL, Raife T, Cable R, Murphy S, Howard
FT, Davis K, Lin JS, Metzel P, Corash L, Koutsoukos A, Lin L, Buchholz DH, Conlan MG
(2004) Therapeutic efficacy and safety of platelets treated with a photochemical process for
pathogen inactivation: the SPRINT Trial. Blood 104:1534–1541
Meng ZH, Wolberg AS, Monroe DM 3rd, Hoffman M (2003) The effect of temperature and pH on
the activity of factor VIIa: implications for the efficacy of high-dose factor VIIa in hypothermic
and acidotic patients. J Trauma 55:886–891
Middelburg RA, Van Stein D, Zupanska B, Uhrynowska M, Gajic O, Muniz-Diaz E, Galvez NN,
Silliman CC, Krusius T, Wallis JP, Vandenbroucke JP, Briet E, Van Der Bom JG (2010) Female
donors and transfusion-related acute lung injury: a case-referent study from the International
TRALI Unisex Research Group. Transfusion 50:2447–2454
Mintz PD, Bass NM, Petz LD, Steadman R, Streiff M, McCullough J, Burks S, Wages D, Van
Doren S, Corash L (2006a) Photochemically treated fresh frozen plasma for transfusion of
patients with acquired coagulopathy of liver disease. Blood 107:3753–3760
Mintz PD, Neff A, MacKenzie M, Goodnough LT, Hillyer C, Kessler C, McCrae K, Menitove JE,
Skikne BS, Damon L, Lopez-Plaza I, Rouault C, Crookston KP, Benjamin RJ, George J, Lin
JS, Corash L, Conlan MG (2006b) A randomized, controlled Phase III trial of therapeutic
plasma exchange with fresh-frozen plasma (FFP) prepared with amotosalen and ultraviolet A
light compared to untreated FFP in thrombotic thrombocytopenic purpura. Transfusion
46:1693–1704
Moffatt SE (2013) Hypothermia in trauma. Emerg Med J 30:989–996
172 T.M. Boyd et al.
Morgenstern LB, Hemphill JC 3rd, Anderson C, Becker K, Broderick JP, Connolly ES Jr,
Greenberg SM, Huang JN, MacDonald RL, Messe SR, Mitchell PH, Selim M, Tamargo RJ
(2010) Guidelines for the management of spontaneous intracerebral hemorrhage: a guideline
for healthcare professionals from the American Heart Association/American Stroke
Association. Stroke 41:2108–2129
Murad MH, Stubbs JR, Gandhi MJ, Wang AT, Paul A, Erwin PJ, Montori VM, Roback JD (2010)
The effect of plasma transfusion on morbidity and mortality: a systematic review and meta-
analysis. Transfusion 50:1370–1383
Nascimento B, Rizoli S, Rubenfeld G, Fukushima R, Ahmed N, Nathens A, Lin Y, Callum J
(2011) Cryoprecipitate transfusion: assessing appropriateness and dosing in trauma. Transfus
Med 21:394–401
O’Shaughnessy DF, Atterbury C, Bolton Maggs P, Murphy M, Thomas D, Yates S, Williamson
LM (2004) Guidelines for the use of fresh-frozen plasma, cryoprecipitate and cryosupernatant.
Br J Haematol 126:11–28
Ozier Y, Muller JY, Mertes PM, Renaudier P, Aguilon P, Canivet N, Fabrigli P, Rebibo D, Tazerout
M, Trophilme C, Willaert B, Caldani C (2011) Transfusion-related acute lung injury: reports to
the French Hemovigilance Network 2007 through 2008. Transfusion 51:2102–2110
Palavecino EL, Yomtovian RA, Jacobs MR (2010) Bacterial contamination of platelets. Transfus
Apher Sci 42:71–82
Peter JV, John P, Graham PL, Moran JL, George IA, Bersten A (2008) Corticosteroids in the pre-
vention and treatment of acute respiratory distress syndrome (ARDS) in adults: meta-analysis.
BMJ 336:1006–1009
Pidcoke HF, McFaul SJ, Ramasubramanian AK, Parida BK, Mora AG, Fedyk CG, Valdez-Delgado
KK, Montgomery RK, Reddoch KM, Rodriguez AC, Aden JK, Jones JA, Bryant RS, Scherer
MR, Reddy HL, Goodrich RP, Cap AP (2013) Primary hemostatic capacity of whole blood: a
comprehensive analysis of pathogen reduction and refrigeration effects over time. Transfusion
53(Suppl 1):137S–149S
Platelet transfusion therapy. National Institutes of Health Consensus Conference (1987) Transfus
Med Rev 1:195–200.
Pock K, Heger A, Janisch S, Svae TE, Romisch J (2007) Thrombin generation capacity is impaired
in methylene-blue treated plasma compared to normal levels in single-donor fresh-frozen
plasma, a licensed solvent/detergent-treated plasma (Octaplas) and a development product
(Uniplas). Transfus Apher Sci 37:223–231
Popovsky MA (2004) Transfusion and the lung: circulatory overload and acute lung injury. Vox
Sang 87(Suppl 2):62–65
Popovsky MA (2009) Transfusion-associated circulatory overload: the plot thickens. Transfusion
49:2–4
Popovsky MA (2010) The Emily Cooley Lecture 2009 To breathe or not to breathe-that is the ques-
tion. Transfusion 50:2057–2062
Popovsky MA, Moore SB (1985) Diagnostic and pathogenetic considerations in transfusion-
related acute lung injury. Transfusion 25:573–577
Popovsky MA, Audet AM, Andrzejewski C Jr (1996) Transfusion-associated circulatory overload
in orthopedic surgery patients: a multi-institutional study. Immunohematology 12:87–89
Powers A, Stowell CP, Dzik WH, Saidman SL, Lee H, Makar RS (2008) Testing only donors with
a prior history of pregnancy or transfusion is a logical and cost-effective transfusion-related
acute lung injury prevention strategy. Transfusion 48:2549–2558
Rao AK, Murphy S (1982) Secretion defect in platelets stored at 4 degrees C. Thromb Haemost
47:221–225
Rebulla P, Finazzi G, Marangoni F, Avvisati G, Gugliotta L, Tognoni G, Barbui T, Mandelli F,
Sirchia G (1997) The threshold for prophylactic platelet transfusions in adults with acute
myeloid leukemia. Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto. N Engl J Med
337:1870–1875
Reesink HW, Lee J, Keller A, Dennington P, Pink J, Holdsworth R, Schennach H, Goldman M,
Petraszko T, Sun J, Meng Y, Qian K, Rehacek V, Turek P, Krusius T, Juvonen E, Tiberghien P,
Legrand D, Semana G, Muller JY, Bux J, Reil A, Lin CK, Daly H, McSweeney E, Porretti L,
10 Split Blood Products 173
Stramer SL, Hollinger FB, Katz LM, Kleinman S, Metzel PS, Gregory KR, Dodd RY (2009)
Emerging infectious disease agents and their potential threat to transfusion safety. Transfusion
49(Suppl 2):1S–29S
Su L, Kamel H (2007) How do we investigate and manage donors associated with a suspected case
of transfusion-related acute lung injury. Transfusion 47:1118–1124
Szczepiorkowski ZM, Winters JL, Bandarenko N, Kim HC, Linenberger ML, Marques MB,
Sarode R, Schwartz J, Weinstein R, Shaz BH (2010) Guidelines on the use of therapeutic
apheresis in clinical practice–evidence-based approach from the Apheresis Applications
Committee of the American Society for Apheresis. J Clin Apher 25:83–177
Theusinger OM, Baulig W, Seifert B, Emmert MY, Spahn DR, Asmis LM (2011) Relative concen-
trations of haemostatic factors and cytokines in solvent/detergent-treated and fresh-frozen
plasma. Br J Anaesth 106:505–511
Tobian AA, Sokoll LJ, Tisch DJ, Ness PM, Shan H (2008) N-terminal pro-brain natriuretic peptide
is a useful diagnostic marker for transfusion-associated circulatory overload. Transfusion
48:1143–1150
Toy P, Gajic O, Bacchetti P, Looney MR, Gropper MA, Hubmayr R, Lowell CA, Norris PJ,
Murphy EL, Weiskopf RB, Wilson G, Koenigsberg M, Lee D, Schuller R, Wu P, Grimes B,
Gandhi MJ, Winters JL, Mair D, Hirschler N, Sanchez Rosen R, Matthay MA (2012)
Transfusion-related acute lung injury: incidence and risk factors. Blood 119:1757–1767
Triulzi DJ, Kleinman S, Kakaiya RM, Busch MP, Norris PJ, Steele WR, Glynn SA, Hillyer CD,
Carey P, Gottschall JL, Murphy EL, Rios JA, Ness PM, Wright DJ, Carrick D, Schreiber GB
(2009) The effect of previous pregnancy and transfusion on HLA alloimmunization in blood
donors: implications for a transfusion-related acute lung injury risk reduction strategy.
Transfusion 49:1825–1835
Vamvakas E (2007) Allergic and anaphylactic reactions. In: Transfusion reactions, 3rd edn. AABB
Press, Bethesda
Vamvakas EC, Blajchman MA (2010) Blood still kills: six strategies to further reduce allogeneic
blood transfusion-related mortality. Transfus Med Rev 24:77–124
van der Meer PF, Kerkhoffs JL, Curvers J, Scharenberg J, de Korte D, Brand A, de Wildt-Eggen J
(2010) In vitro comparison of platelet storage in plasma and in four platelet additive solutions,
and the effect of pathogen reduction: a proposal for an in vitro rating system. Vox Sang
98:517–524
van Stein D, Beckers EA, Sintnicolaas K, Porcelijn L, Danovic F, Wollersheim JA, Brand A, van
Rhenen DJ (2010) Transfusion-related acute lung injury reports in the Netherlands: an obser-
vational study. Transfusion 50:213–220
Vasconcelos E, Figueiredo AC, Seghatchian J (2003) Quality of platelet concentrates derived by
platelet rich plasma, buffy coat and Apheresis. Transfus Apher Sci 29:13–16
Vilahur G, Choi BG, Zafar MU, Viles-Gonzalez JF, Vorchheimer DA, Fuster V, Badimon JJ (2007)
Normalization of platelet reactivity in clopidogrel-treated subjects. J Thromb Haemost
5:82–90
Viuff D, Lauritzen B, Pusateri AE, Andersen S, Rojkjaer R, Johansson PI (2008) Effect of haemo-
dilution, acidosis, and hypothermia on the activity of recombinant factor VIIa (NovoSeven). Br
J Anaesth 101:324–331
Vlaar AP, Binnekade JM, Prins D, van Stein D, Hofstra JJ, Schultz MJ, Juffermans NP (2010) Risk
factors and outcome of transfusion-related acute lung injury in the critically ill: a nested case-
control study. Crit Care Med 38:771–778
Walker RH (1987) Special report: transfusion risks. Am J Clin Pathol 88:374–378
Wallis JP (2003) Transfusion-related acute lung injury (TRALI)–under-diagnosed and under-
reported. Br J Anaesth 90:573–576
Wallis JP (2007) Transfusion-related acute lung injury (TRALI): presentation, epidemiology and
treatment. Intensive Care Med 33(Suppl 1):S12–S16
Wehrli G, Taylor NE, Haines AL, Brady TW, Mintz PD (2009) Instituting a thawed plasma proce-
dure: it just makes sense and saves cents. Transfusion 49:2625–2630
10 Split Blood Products 175
Welsby IJ, Troughton M, Phillips-Bute B, Ramsey R, Campbell ML, Bandarenko N, Mathew JP,
Stafford-Smith M (2010) The relationship of plasma transfusion from female and male donors
with outcome after cardiac surgery. J Thorac Cardiovasc Surg 140:1353–1360
Wheeler AP, Bernard GR, Thompson BT, Schoenfeld D, Wiedemann HP, de Boisblanc B, Connors
AF Jr, Hite RD, Harabin AL (2006) Pulmonary-artery versus central venous catheter to guide
treatment of acute lung injury. N Engl J Med 354:2213–2224
Wikkelso A, Lunde J, Johansen M, Stensballe J, Wetterslev J, Moller AM, Afshari A (2013)
Fibrinogen concentrate in bleeding patients. Cochrane Database Syst Rev 8, CD008864
Yazer MH, Cortese-Hassett A, Triulzi DJ (2008) Coagulation factor levels in plasma frozen within
24 hours of phlebotomy over 5 days of storage at 1 to 6 degrees C. Transfusion
48:2525–2530
Yazer MH, Triulzi DJ, Hassett AC, Kiss JE (2010) Cryoprecipitate prepared from plasma frozen
within 24 hours after phlebotomy contains acceptable levels of fibrinogen and VIIIC.
Transfusion 50:1014–1018
Zhou L, Giacherio D, Cooling L, Davenport RD (2005) Use of B-natriuretic peptide as a diagnos-
tic marker in the differential diagnosis of transfusion-associated circulatory overload.
Transfusion 45:1056–1063
Coagulation Factor Concentrates
11
Lars M. Asmis
11.1 Introduction
L.M. Asmis
Unilabs Coagulation Laboratory, Centre for Perioperative Thrombosis and Hemostasis,
Hufgasse 17, Zurich CH 8008, Switzerland
e-mail: [email protected]
since been shown that FFP may be contaminated with fragments of cellular origin,
termed as microparticles, as well as soluble mediators including antibodies and
interleukins (George et al. 1986; Theusinger et al. 2011). These contaminants can
potentially lead to adverse effects. A rare but dangerous complication of FFP trans-
fusion is transfusion-related acute lung injury (TRALI) that is associated with
increased mortality (Inaba et al. 2010).
The ongoing quest for efficient and safe transfusion products has resulted in the
development of isolated coagulation factor concentrates (i-CFC) and recombinant
coagulation factor concentrates (r-CFC). This chapter focuses on isolated (single)
and combined coagulation factor concentrates. Based on differences in regulation
and approval procedures in various countries, there are relevant geographical differ-
ences in the availability and use of coagulation factor products. Indications for the
various products vary significantly from country to country. Substitution of i-CFC
in acquired coagulation factor deficiencies represents on-label use in some coun-
tries, while it is off label in others. This chapter discusses the most frequently used
coagulation factor concentrates.
11.2.1.1 Background
Fibrinogen was the first coagulation factor to be discovered. In an open wound, the
white aggregates of fibrin strands can be seen with the naked eye. In the nineteenth
century, Home coined the term “fibrin,” and Virchow postulated that there was a
precursor molecule, which he called “fibrinogen.” In 1905, fibrinogen was already
defined as a coagulation factor I (FI) in a review by Morawitz (1905). Hiippala and
colleagues established fibrinogen’s key role in perioperative hemostasis and demon-
strated that it was the first coagulation factor to reach critical levels in massive
bleeding (see “Indications”, below) (Hiippala et al. 1995).
11.2.1.2 Description
Fibrinogen, synthesized in the liver, is an abundant plasma protein with concentrations
typically ranging between 1.5–4.0 g/l and 6–13 µmol/l. Fibrinogen’s molecular
weight is approximately 340 kDa. It is a heterodimer with two sets of α, β, and γ
chains. The six chains are N covalently linked in the central E domain. Two sets of α,
β, and γ chains extend from the E domain to form 2 so-called D domains at their
distal ends. Thrombin (FIIa) cleaves activation peptides from the central (E domain)
end of the α and β chains, releasing two sets of fibrinopeptides, A and B, thereby
exposing polymerization sites. After the release of these fibrinopeptides, the mole-
cules are called fibrin or fibrin monomers. The fibrin monomers aggregate to form
soluble fibrin strands that form by the interaction of the D and E domains of neigh-
boring molecules. Initially, the interaction between fibrin monomers is reversible; the
11 Coagulation Factor Concentrates 179
BA
AB
D E D
BA
AB
D E D D E D D E D
D E D
D E D D E D D E D
BA
AB
D E D FIIa FIIIa
Soluble fibrinogen Soluble fibrin monomers Insoluble fibrin strand
Fig. 11.1 Model of fibrinogen polymerization. Thrombin (FIIa)-induced cleavage of the fibrino-
peptides A and B from soluble fibrinogen molecules leads to the generation of soluble fibrin mono-
mers that reversibly aggregate into a soluble fibrin monomer gel. Activated factor XIII (FXIIIa)
can irreversibly stabilize the monomers into an insoluble and stable fibrin strand through covalent
cross-linking
resulting fibrin strand can potentially disintegrate again. Only once factor XIII
(FXIII) has covalently cross-linked the individual fibrin monomers does the fibrin
strand become irreversibly interconnected (Nieuwenhuizen 1995; de Maat and
Verschuur 2005; Sorensen et al. 2011b; Levy et al. 2012) (see Fig. 11.1).
The three polypeptide chains (Aα, Bβ, and γ) are encoded by three distinct genes,
all residing on chromosome 4. The three genes contain interleukin 6 (IL-6) response
elements, which explains why fibrinogen is an acute phase reactant.
Fibrinogen is a glycoprotein that has a plasma half-life of approximately 100 h
(Blomback et al. 1966). Liver disease and sepsis increase turnover. In liver cirrhosis,
glycosylation may be altered, leading to acquired dysfibrinogenemia, characterized
by circulating fibrinogen molecules with reduced biological activity. Degradation of
soluble fibrinogen is mediated by “normal protein degradation, the coagulation
process, and unknown pathways.” Stabilized fibrin (and soluble fibrinogen) degra-
dation occurs by plasmin-induced proteolysis (de Maat and Verschuur 2005).
Fibrinogen has at least four major biological functions: it is a substrate of coagu-
lation; it is a cross-linker in platelet aggregation; it is a coagulation inhibitor as
“antithrombin I”; and it is a substrate for interaction with other proteins (Weisel and
Litvinov 2013). Most other coagulation factors are serine proteases capable of spe-
cifically recognizing target molecules and activating many of them. For example,
one activated molecule of factor X (FXa) can activate more than 1,000 prothrombin
molecules (FII), while one thrombin molecule can activate more than 1,000 mole-
cules of fibrinogen. Enzymatically active coagulation factors can continue to inter-
act with many other target molecules while fibrinogen, in its role as substrate, will
be consumed. One activated molecule of fibrinogen will give rise to one molecule
of fibrin in the resulting clot. Fibrinogen’s second role is that of the cross-linker in
platelet aggregation. Once platelets have been activated “outside in,” an “inside-
out” activation will occur that leads to an altered structure of the glycoprotein IIb/
IIIa (GP IIb/IIIa) receptor on the platelet surface. This structural change permits the
interaction of one fibrinogen molecule with several GP IIb/IIIa receptors on several
180 L.M. Asmis
platelets, leading to the formation of platelet aggregates. The third role involves
fibrinogen’s ability to bind thrombin (Sorensen et al. 2011b). Finally, there is FXIII,
which is reported to covalently link α2-plasmin inhibitor (α2-PI) on to fibrinogen
(Richardson et al. 2013). This reaction is important for the stability of the resulting
fibrin clot (see below).
11.2.1.3 Indications
11.2.1.3.1 Acquired Fibrinogen Disorders
Hiippala’s pioneering work identified the clinical relevance of fibrinogen in massively
bleeding patients (Hiippala et al. 1995). He studied 60 massively bleeding patients
and measured platelet counts and coagulation factor levels. Based on “critical
levels,” or thresholds, which he took from a prominent coagulation textbook of the
time, he observed that the fibrinogen threshold of 1.0 g/l was reached at 142 % of
blood loss. Other parameters he monitored only fell below their respective thresh-
olds considerably later: platelet count at 230 %, FII at 201 %, FV at 229 %, and
FVII at 236 %.
Others corroborated the finding that fibrinogen will be the first factor to reach
critical levels in massive bleeding (Martini et al. 2005; Fenger-Eriksen et al. 2009c).
Acquired hypofibrinogenemia is thus a frequently observed problem in massively
bleeding patients. Suggested treatment thresholds lie ≥1.5 g/l (Fenger-Eriksen et al.
2009a). A less frequent problem is acquired dysfibrinogenemia. This may occur in
end-stage liver cirrhosis as a consequence of an altered glycosylation of fibrinogen
molecules that interferes with the plasma assembly of fibrin monomers. The laboratory
hallmark of this disorder is longer thrombin time and a reduced ratio of functional
to antigenic fibrinogen tests (<0.7).
11.2.1.4 Monitoring
Tests for fibrinogen were proposed in the middle of the last century: the Schulz
method in 1955 and the Clauss method in 1957 (Schulz 1955; Clauss 1957;
Nieuwenhuizen 1995; Lowe et al. 2004). Interestingly, Hartert had published his
work on thromboelastography several years before that, in 1948, but functional
fibrinogen tests for viscoelastic point-of-care (POC) devices (including functional
“fibrinogen” tests on ROTEM, TEG, and related devices) were only standardized
and validated much later (Hartert 1948).
11 Coagulation Factor Concentrates 181
11.2.1.6 Dosing
The following dosing regimen has been suggested in the context of classic coagula-
tion testing: dose (g) = desired increase in g/l × plasma volume (plasma vol-
ume = 0.07 × (1-hematocrit) × body weight (kg)) (Acharya and Dimichele 2008). In
the context of thromboelastographic/metric testing, 2 g of fibrinogen in a 70-kg
patient generally increase the FIBTEM by 4 mm (Rahe-Meyer et al. 2009). The
manufacturer of fibrinogen concentrate recommends 30–60 mg/kg or 2–4 g in a
70-kg patient (Fenger-Eriksen et al. 2009a). This dose will increase plasma fibrino-
gen by approximately 1 g/l (Solomon et al. 2010). Threshold and target values for
fibrinogen have changed considerably in the recent years (see Sect. 11.3) (Sorensen
et al. 2011b). In the recent Rahe-Meyer RCT, the target post-substitution fibrinogen
level was 3.6 g/l or a maximum cloth firmness (MCF) of 22 mm in ROTEM in
patients undergoing major aortic replacement surgery.
11.2.2.1 Background
Factor XIII (FXIII) was the last coagulation factor to be discovered. First indica-
tions of its existence date back to the 1920s. Fibrin clots that formed in the pres-
ence of calcium became insoluble in the presence of weak bases (Barkan 1923). A
similar experiment was performed in 1944 with purified fibrinogen. The authors
concluded that an additional “stabilizing factor” to calcium was necessary to
explain their findings (Robbins 1944). Duckert first described human FXIII defi-
ciency in 1960 (Duckert et al. 1960). The protein was attributed with its current
name in 1963 by the International Committee on Blood Clotting Factors (Muszbek
et al. 2011).
11 Coagulation Factor Concentrates 183
11.2.2.2 Description
FXIII is a transglutaminase, an enzyme that cross-links proteins. This clearly distin-
guishes it from the classic coagulation factors that are serine proteases – enzymes
that cleave proteins. In plasma, FXIII bound to fibrinogen forms a tetramer with two
catalytic A domains and two inhibitory B domains. Cellular FXIII, present in plate-
lets, monocytes, and bone marrow-derived cells, has two catalytic A domains
(Muszbek et al. 2011; Levy and Greenberg 2012).
The catalytic A domain is an 83 kDa protein that can be activated by proteolytic
activation (by thrombin) or non-proteolytic activation (in the presence of high calcium
concentrations).
The FXIIIA encoding genes reside on chromosome 6 (6p24-25). It is expressed
in bone marrow-derived cells including megakaryocytes, macrophages/monocytes,
and in hepatocytes. The B domain, which lacks enzymatic activity, is an 80 kDa
protein. The FXIIIB gene is located on chromosome 1 (1q31-32.1) and is expressed
primarily in hepatocytes (Hsieh and Nugent 2008).
In the context of the fibrin clot, FXIII has two functions with fibrin and fibrino-
gen as its main substrates. FXIII-mediated cross-linking of soluble fibrin monomers
results in the generation of an insoluble or stabilized fibrin strand that is essential for
in vivo hemostasis. Furthermore, FXIII mediates the cross-linking of fibrinolysis
inhibitors to fibrinogen, e.g., the α2-plasmin inhibitor (α2-PI, also called
α2-antiplasmin) (Mosesson et al. 2008; Fraser et al. 2011). Fibrinogen circulating in
a context of sufficient FXIII will be rich in α2-PI cross-linked to it. This type of
fibrinogen will eventually give rise to fibrin strands rich in α2-PI. The presence of
the fibrinolysis inhibitor in the fibrin strands hypothetically assures an increased
stability of the hemostatic plug. Fibrinogen with low α2-PI supposedly gives rise to
fibrin strands that are more prone to plasmin’s fibrinolytic activity and may thus be
associated with an increased risk of bleeding.
11.2.2.3 Indications
11.2.2.3.1 Acquired FXIII Deficiency
Acquired FXIII deficiency has been described in a variety of clinical settings due to
consumption of the coagulation factor and due to inhibition by acquired inhibitors
(Levy and Greenberg 2012). Consumption of FXIII can occur in massive bleeding,
but its incidence is not very well defined in the literature. A study in 1,004 adult
patients in a university hospital setting showed a prevalence of 21 % of patients with
values below the reference range. A 62 % incidence of acquired FXIII deficiency was
found in a population of patients with acute leukemia. In view of this high frequency,
some published algorithms for bleeding management incorporate FXIII. Conditions
associated with large wound areas, such as chronic inflammatory bowel disease
(IBD: ulcerative colitis and Crohn’s disease), massive burns, extensive orthopedic
surgery of the spine, etc., are known to be associated with low FXIII levels. However,
whether low levels of FXIII are associated with a clinical bleeding diathesis in the
context of IBD, for example, is contested (Van Bodegraven et al. 1995; Chamouard
et al. 1998). Acquired deficiency due to autoantibody formation is an extremely rare
event, with less than 100 reported cases to date (Boehlen et al. 2013).
184 L.M. Asmis
11.2.2.4 Monitoring
FXIII monitoring can be done using functional and antigenic tests. The usual test is
clot solubility in monochloroacetic acid solution, but this functional test is neither
standardized nor quantitative. Quantitative FXIII activity tests use activated FXIII
to assay its transglutaminase activity. This can be done in two main ways: (1) mea-
surement of ammonia released as an early byproduct of the transglutaminase reac-
tion (ammonia release assays) and (2) measurement of amine substrates that are
covalently linked to another substrate (amine-incorporation assays). More commer-
cial kits use the first method than the second. As there are non-FXIII-dependent
ammonia-releasing reactions that can account for up to 15 % of the measured activ-
ity, FXIII measurements <20 % should be interpreted with caution and confirmed in
an experienced laboratory. It should be noted that high ammonia concentrations in
severe hepatic dysfunction can interfere with ammonia release assays. Antigenic
tests can be directed against subunit A, subunit B, or the combination of both (Hsieh
and Nugent 2008; Karimi et al. 2009). In the context of perioperative FXIII moni-
toring, any automated, rapid, reproducible, and precise test is suitable. Knowing the
lower detection limit and test range used in one’s institution is important. Peyvandi’s
review shows that FXIII correlates badly with clinical bleeding (Peyvandi et al.
2012b).
Some literature suggests that, due to absent or inadequate testing strategies,
acquired FXIII deficiency goes largely undetected (Lawrie et al. 2010). Classic
global coagulation tests, including prothrombin time (PT) and activated partial
thromboplastin time (aPTT), are “blind” to FXIII (see Fig. 11.2). Specialized tests,
such as thrombin time and fibrinogen according to Clauss, do not incorporate FXIII
activity. The common stop signal for all the coagulation time-based tests is the
appearance of soluble fibrin monomers in the test vial. Fibrin monomers can be
detected optically or mechanically. Whatever the method, coagulation time mea-
surements stop prior to the onset of FXIII’s action.
PK
HK
FXII
aPTT FVII PT/Quick
FIX Ca++
Ca++
FVIIIa PL
FX Ca++
FVa PL
FXIII
Fig. 11.2 The cascade model of coagulation and coagulation tests. Coagulation factors are
depicted as circles. Yellow circles indicate enzymatically active coagulation factors (exception:
fibrinogen, which is not enzymatically active, but a substrate). Yellow circles with a pink insert
represent activated coagulation factors. Orange circles indicate cofactors. Tissue factor is depicted
in blue, as it normally does not circulate in plasma. The factors involved in the activated partial
thromboplastin time (aPTT) and prothrombin time according to Quick (PT) are circled by light
blue and purple polygons, respectively. It is important that both FXIII and von Willebrand factor
(not depicted) are not assayed in the so-called standard coagulation tests. Prothrombin complex
concentrates contain FIX, FX, and FII and FVII to a variable degree. Substitution with PCC thus
influences both PT and aPTT. Abbreviations: Ca 2+ calcium, Fibrins soluble fibrin, Fibrini insoluble
fibrin, HK high molecular weight kininogen, PL phospholipids, PK prekallikrein
11.2.2.6 Dosing
No prospectively evaluated treatment thresholds or target values have been estab-
lished for the frequently acquired FXIII deficiencies.
In cases of congenital deficiency and in patients at a high risk of bleeding, 10 IU/kg
of FXIII are given at 4–6 week intervals (Bolton-Maggs et al. 2004) for prophylaxis.
This translates into 500–1,000 IU per application. In settings of acute bleeding,
10–20 IU/kg are recommended. This corresponds to single doses of 750–1,500 IU/kg
for a patient of approximately 75 kg.
11.2.3.1 Background
Von Willebrand factor (vWF) is named after the Finnish internist Erik A von
Willebrand, who first described a family with the disease in 1926 (von Willebrand
1926; Federici et al. 2006). The family came from the Åland Islands in the Baltic
Sea. In contrast to the hemophilias, both sexes were affected. The disease was char-
acterized by a prolonged bleeding time, despite a normal platelet count, normal PT,
(described by Quick in 1935), and normal-to-increased aPTT (described by Langdell
in 1953). The disease’s description was published under the designation “hereditary
pseudohemophilia.” In the 1950s, it became possible to measure FVIII and the first
cases of “pseudohemophilia” were described in patients of both sexes who had
reduced FVIII levels. In 1963, reduced platelet adhesiveness in affected patients
became detectable (Salzman 1963). In 1971, using a specific antibody against FVIII,
Zimmermann was able to show that there was another protein responsible for the
phenotype (Zimmerman et al. 1971), initially called FVIII-related protein. In 1971,
Howard and Firkin showed that an antituberculosis agent, ristocetin, could trigger
platelet agglutination in normal subjects, but not in patients with von Willebrand’s
disease (Howard et al. 1973). Von Willebrand factor was finally isolated in 1972
(Bouma et al. 1972), and as there are several distinct underlying pathophysiological
mechanisms, von Willebrand syndrome (vWS) is the recommended designation.
11.2.3.2 Description
VWF is a multimeric glycoprotein with monomers approximately 2,050 amino
acids long. These monomers are assembled into dimers, which are subsequently
polymerized into multimers of 2–20 units with a molecular weight of up to
20 × 106 D. VWF is encoded on the short arm of chromosome 12. It is primarily
expressed and stored in endothelial cells (Weibel–Palade bodies) and megakaryo-
cytes (α granules). Endothelial cells express DDAVP receptors that can induce vWF
secretion. DDAVP stimulation can lead to three- to five-fold increase in plasma
levels of vWF. Platelet activation and degranulation will also lead to a release of
stored vWF (Laffan et al. 2004).
11 Coagulation Factor Concentrates 187
The vWF monomer has 4 N terminal D domains (D1, D2, D’, D3), then 3 A
domains (A1, A2, A3) followed by another D domain (D4), a B domain, and three
terminal C domains (C1, C2, CK). VWF has three main functions, the first of which
is binding and stabilizing FVIII. FVIII binds to the vWF N terminal D′-D3 domains.
Its second function is interaction with platelets through glycoprotein GP Ibα expressed
on the platelet surface. Platelet GP Ibα interacts with the vWF A1 domain. Third,
vWF can bind to collagen in the subendothelium. Collagen-binding sites on vWF are
in the A3 or A1 domains (Schneppenheim and Budde 2011). The vWF-cleaving
protease, ADAMTS13, cleaves vWF in the A2 domain (Crawley et al. 2011).
VWF is a key molecule in hemostasis as it interacts with all three of its subsys-
tems. It interacts with vascular wall collagen in cases of trauma or other causes of
denudation of the vascular lining; it provides the landing strips for platelets by bind-
ing to their GP 1bα, thereby immobilizing them in the subendothelial matrix; and it
stabilizes plasmatic FVIII. Similarly to fibrinogen, vWF plays a role in both pri-
mary and secondary hemostasis.
Endothelial cells release vWF as long chains of covalently linked dimers. These
multimers are degraded by ADAMTS13 as they circulate. As the molecules decrease
in size, they become less “sticky” (Schneppenheim and Budde 2011).
11.2.3.3 Indications
11.2.3.3.1 Acquired Deficiency
Unlike other coagulopathies, such as fibrinogen or FXIII deficiencies, where
acquired deficiencies are more frequent than inherited forms, acquired vWS is less
frequent than its congenital variant. By 2000, fewer than 200 cases had been reported
(Sucker et al. 2009). Based on those figures and the author’s personal experience,
underreporting is very likely. Acquired vWS has been described in association with
underlying disorders including myeloproliferative disorders (where affected cells
may bind and consume vWF), lymphoproliferative disorders (autoantibodies against
vWF), other neoplasms, autoimmune disorders, cardiovascular disorders (vWF
consumption due to aortic stenosis, i.e., Heyde syndrome), and drug effects
(hydroxyl-ethyl starch is reported to reduce vWF antigen (vWF:AG), but interfer-
ence with vWF function is also possible) (Mohri 2006; Federici et al. 2013). A high
frequency of a new form of acquired vWS has been associated with patients using
ventricular assist devices. The supposed mechanism is the mechanical destruction
(versus the action of ADAMTS13) of the molecule (Dassanayaka et al. 2013).
Interestingly, in the context of acute bleeding, consumption of vWF without any of
the underlying disorders cited above is a rarely reported problem. This clearly
distinguishes vWF from fibrinogen, which is the first molecule to reach critical
levels in acute bleeding. This is possibly due to vWF’s multiple production sites
(megakaryocytes in bone marrow, endothelial cells) and storage pools (platelets,
endothelial cells).
11.2.3.4 Monitoring
Perioperatively, vWF testing should be based on pretest probability. If a validated
bleeding questionnaire is positive, then preoperative hemostasis testing is indicated
(Tosetto et al. 2006). VWF:AG, vWF ristocetin cofactor (vWF:RCo), and their ratio
are sufficient to detect most of the clinically relevant manifestations of vWS.
Historically, vWF:AG was the first to be measured. This can be done manually
using ELISA technology, but also using modern tests. The discovery that ristoce-
tin could agglutinate platelets was used to design the first functional vWF:RCo
test. Large vWF molecules tend to have vWF:RCo to vWF:Ag ratios of close to 1.
When vWF consumption increases, due to a stenotic heart valve, for example,
there is an increased proportion of smaller, less “sticky” vWF molecules, and the
vWF:RCo to vWF:Ag ratio will drop. Ratios of less than 0.70 are considered
indicative of acquired vWS or forms of congenital type II vWS, characterized by
qualitative vWF defects.
Exact subtyping of vWS can be done by specialized laboratories capable of mea-
suring vWF collagen-binding capacity (vWF:CB), vWF FVIII-binding capacity
(vWF:FVIII), and performing vWF multimer (vWF:MM) analysis (Laffan et al.
2004; Sucker et al. 2009).
11.2.3.6 Dosing
Treatment of vWS aims to correct the deficient adhesion of platelets to the subendo-
thelium and to redress FVIII deficiency if necessary. Dosing recommendations are
mostly based on similarities to congenital vWF deficiency contexts and some lim-
ited experience with acquired vWS (AvWS) (Federici 2005; Franchini 2008; Tiede
et al. 2011). As a rule of thumb, substitution of 1 IU of vWF (RCo) per kg body-
weight will raise vWF activity by approximately 2 %. VWS acquired due to severe
aortic stenosis (Heyde syndrome) or other valvular defects is probably one of the
most frequent forms of AvWS. Either no substitution or a single preoperative dose
of Haemate P® 500–1,000 IU IV can be considered (not evidence based, in line with
(Mohri 2006; Federici et al. 2013) and personal experience). Once a defective heart
valve has been replaced, vWF levels return to normal and within hours the bleeding
diathesis will disappear. As in other indications, the severity of the AvWS, the clini-
cal context (active bleeding or not, previous bleeding complications), and potential
treatment complications, particularly thromboembolism, must be taken into consid-
eration. Treatment thresholds and targets are discussed later in Sect. 11.3. There are
differing vWF concentrates on the market. They can be categorized according to
purification method, viral inactivation, vWF:RCO to vWF:AG ratio, vWF:RCO to
FVIII ratio, and vWF MM content (Franchini 2008). The one for which most expe-
rience exists is Haemate P®. The pharmacokinetics of concentrates may also show
interindividual variability (Kessler et al. 2011).
11.2.4.1 Background
Prothrombin complex concentrates (PCC) were historically also referred to as PPSB.
This latter abbreviation summarized the four coagulation factors: prothrombin (FII),
proconvertin (FVII), Stuart–Prower factor (FX), and antihemophilic factor B (FIX).
The first description of PCC dates back to more than 50 years ago when Didisheim
described the preparation of this human plasma fraction and its potential application
in humans (Didisheim et al. 1959). Surgenor described the original isolation proce-
dure – barium sulfate elution – in 1959, and methodology improved over the
190 L.M. Asmis
following years. In 1965, Tullis reported on the clinical use of prothrombin com-
plexes in the New England Journal of Medicine (Tullis et al. 1965). First reports on
the adverse thromboembolic effects of PCC led to the addition of heparin (Kasper
1975; Menache 1975). When cases of PCC-associated transmission of hepatitis B
and further thromboembolism-associated deaths were described, an international
task force was convened. This led to products being retracted from the market, stron-
ger regulation regarding isolation procedures, product surveillance regarding the con-
tamination by activated coagulation factors, the addition of inhibitors (antithrombin,
protein C, and protein S) to the products, viral inactivation procedures, and a formal
recommendation by the European Medicines Agency (EMEA) regarding minimal
and desired potencies for the various factors (Hellstern 1999; Hellstern et al. 1999).
11.2.4.2 Description
PCC are subdivided into two major categories based on their composition: three-
factor concentrates (containing FII, FIX, and FX – some contain traces of FVII) and
four-factor concentrates (containing all four factors, FII, FVII, FIX, and FX). PCC
were originally intended for the treatment of hemophilia B or hereditary deficiency
of FIX. In view of this, PCC were “labeled” according to their FIX content. Most
current PCC contain 500 or 600 IU of FIX per vial and thus have this figure in their
brand name. Procedures aimed at increasing product safety are frequently carried
out (and an appropriate suffix added to the product name), such as nanofiltration
(N or NF or F), pasteurization (P), solvent detergent treatment (SD or D), and vapor
and/or heat (VH or T). It is important to know that PCC from different producers
differ significantly in their relative and absolute content of the various coagulation
factors (Samama 2008; Sorensen et al. 2011a). Based on the variable yield achieved
during production, different lots from the same producer also vary in composition,
which is the reason why their factor content is generally indicated as a range and not
a single figure.
A further categorization of PCC is possible based on whether or not activated
coagulation factors are included in the formulation. Current PCC, registered for
most of the indications noted below, are nonactivated PCC. Activated PCC (aPCC)
represent a formulation that was designed and intended for use with hemophilia
patients who had acquired antibodies directed against the coagulation factor that
they had a deficiency for – so-called inhibitor patients. Factor eight inhibitor bypass-
ing agent (FEIBA) is the only registered plasma-based aPCC. It contains FII, FIX,
and FX, primarily but not solely in their nonactivated forms, and FVII largely in its
activated form (FVIIa) (Cromwell and Aledort 2012).
11.2.4.3 Indications
11.2.4.3.1 Acquired Disorders
Most coagulation factors are synthesized in the liver, including FII, FVII, FIX, and
FX. After synthesis in the hepatocyte, they are modified prior to secretion into
the blood stream. One of the modification steps is mediated by γ carboxylase, the
vitamin K-dependent enzyme that is the site of action of vitamin K antagonists.
11 Coagulation Factor Concentrates 191
Acquired deficiencies of FII, FVII, FIX, and FX can occur for several reasons,
including (1) coagulation inhibition due to vitamin K antagonists (VKA), (2)
coagulation factor consumption in the context of coagulopathy or uncontrolled
bleeding, (3) inhibition of coagulation by direct anticoagulants, and (4) other rare
causes.
The most frequent cause of an acquired prothrombin complex deficiency is the
use of VKA. Approximately 1 % of the population is estimated to be anticoagulated
at any given time. In randomized trials for atrial fibrillation, the bleeding risk of
patients treated by VKA ranged from 1.3 to 4.2 % per year (Wiedermann and
Stockner 2008).
Coagulation factor consumption in uncontrolled bleeding will lead to a critical
prothrombin complex factor deficiency after loss of approximately 200–240 % of
the calculated blood volume (Hiippala et al. 1995). Coagulation factor consumption
can also occur in the absence of active blood loss, e.g., in disseminated intravascular
coagulopathy.
While the reversal of VKA effects on coagulation is a well-established indication
for PCC, the data on PCC use to reverse the effects of novel oral anticoagulants
(NOAC), including direct FIIa inhibitors (dabigatran) and direct FXa inhibitors
(including apixaban, edoxaban, rivaroxaban, and others), is only now emerging
(Eerenberg et al. 2011). The FEIBA aPCC has recently been included in European
guidelines for the treatment of NOAC-related bleeding (Kozek-Langenecker et al.
2013). However, to date, no RCTs have been published on PCC or aPCC use in
actively bleeding patients (Siegal and Cuker 2013).
Rare causes of acquired deficiencies involving one or more factors of the pro-
thrombin complex include severe nutritional vitamin K deficiency, an acquired
inhibitor (antibody) directed against FIX in hemophilia B patients, a propeptide
mutation of the FIX gene leading to pseudohemophilia B in patients treated with
VKA (Ulrich et al. 2008), absorption of FX to amyloid protein in systemic AL amy-
loidosis, and inhibitors directed against thrombin (FIIa) in patients with the same
disease (Thompson et al. 2010).
11.2.4.4 Monitoring
Factors II, VII, and X are key factors in determining PT; thus PT will detect deficien-
cies of these factors and is suitable for PCC monitoring (see Fig. 11.3). The activity
and antigen levels of these three factors can also be used for this purpose.
Characteristics including plasma half-lives are given in Table 11.1. FIX is a vitamin
K-dependent (VKD) factor, as are the three mentioned above. However, it is not a
key determinant of PT. For FIX, aPTT and FIX determinations are the sensitive tests
necessary to monitor substitution.
Point-of-care (POC) technologies including rotational thromboelastometry
(ROTEM) and thromboelastography (TEG), as well as other systems, can assay
a
6
[Fibrinogen] (g/l)
5
4
3
2
1
1 2 3 4 5 6 7
Time (days)
b
6
[Fibrinogen] (g/l)
5
4
3
2
1
1 2 3 4 5 6 7
Time (days)
Fig. 11.3 The threshold and target question. (a) Depicts the time course of plasma fibrinogen
concentration in a fictive patient, e.g., with hereditary afibrinogenemia (yellow curve). After (one
big) substitution, the concentration increases into the normal range (depicted by dark blue area)
and drops off with a half-life of approximately 72–100 h over the following days. (b) Illustrates the
time course of a fictive perioperatively bleeding patient (red curve). The transfusion threshold in
this example is set at 1.0 g/l (lower border of dark blue box) and the transfusion target at 2.0 g/l
(upper limit of light blue box). Initially multiple (small) substitutions are needed to reach the trans-
fusion target value. After two more instances of substitution, the fibrinogen levels stabilize as the
fictive patient ceases to bleed. Note that in the acutely bleeding patient, the observed half-life of
fibrinogen is initially much shorter than 72 h and only later approaches the value described under
physiological conditions
11 Coagulation Factor Concentrates 193
FII-, FVII-, FIX-, and FX-dependent pathways. Tests targeting the “extrinsic” system
(using tissue factor as the starting reagent) are available for thromboelastometric/
graphic test systems. PCC influence the clotting times of such tests, including
EXTEM and RapidTEG. However, as anticoagulated patients may present normal
194 L.M. Asmis
clotting or R times in these whole blood tests, sensitivity is an issue. POC testing
systems have the advantage of shorter turnaround times than routine coagulation
tests. However, the fact that they generally use whole blood can have a negative
impact on their sensitivity toward VKA, other anticoagulants, and possibly PCC.
11.2.4.6 Dosing
Clinical trials have not been able to resolve questions about the optimal dose of
PCC. Doses reported in the studies cited above range from 7 to more than 80 IU/kg
bodyweight. As RCT data is lacking, guidelines and other official dosing informa-
tion are helpful (Makris et al. 1997; Mannucci and Douketis 2006; Weber et al.
2013). Algorithms dependent on the international normalized ratio (INR) exist.
In perioperative bleeding, 20–30 IU/kg bodyweight has been suggested by a group
of Austrian experts (OEGARI). The numerous confounding factors include differ-
ing target values for the INR; the fact that dosing algorithms are often based on
pharmacological models which may or may not be applicable to a given patient; the
question of whether the product is applied to an actively bleeding or non-bleeding
patient; and the clinical context in which PCC are prescribed. Whenever possible,
dosing should be standardized within institutions or clinics and should follow an
evidence-based algorithm appropriate to the patient’s clinical context (van Aart
et al. 2006).
Factors reported to be associated with adverse outcome include repeated dosing
(with potential accumulation of coagulation factors with a long half-lives, such as
FII), coadministration with other coagulation products, and nonobservance of
recommended infusion speeds (Pabinger et al. 2010).
11 Coagulation Factor Concentrates 195
11.2.5.1 Background
Recombinant human activated factor seven (rFVIIa) was developed more than 20
years ago for hemophilia patients suffering from inhibitor formation. Antibodies
directed against FVIII, or inhibitors, may complicate the treatment of hemophilia
patients who are treated by FVIII products. The inhibitors may then neutralize the
patient’s own and the exogenous FVIII, leading to severe bleeding complications.
Bypassing agents that circumvent the FIX/FVIII complex can stop bleeding in these
patients. Pure plasma-derived FVIIa showed clinical efficacy in proof-of-principle
experiments. Subsequently, recombinant FVIIa was developed and tested in clinical
trials (Hedner 2007, 2012).
Distinguishing the clinical setting in which patients are treated with FVIIa appears
relevant. On-label indications include only hypocoagulable patients. In off-label
indications patients may be normo- or even hypercoagulable.
11.2.5.2 Description
FVIIa circulates in plasma and comprises approximately 1 % of the circulating
plasma FVII pool. The precursor single-chain FVII molecule is synthesized in the
liver and is one of the vitamin K-dependent coagulation factors. FVII is a 50 kD pro-
tein that has the shortest plasma half-life among coagulation proteins (see Table 11.1).
The activation of FVII to two-chain FVIIa involves cleavage at a single peptide bond.
This activation can be mediated by FXa, FVIIa itself, and other coagulation factors.
By binding to surface-bound tissue factor, the enzymatic activity of FVIIa is dramati-
cally increased. The cell surface-bound complex of calcium/tissue factor/FVIIa is
capable of activating FX and FIX (Hedner and Brun 2007; Vadivel and Bajaj 2012).
Two mechanisms of action have been postulated for rFVIIa. The first is the
tissue factor-mediated process described above on the surface of cells expressing
tissue factor. The second is tissue factor independent and believed to occur by
direct binding of FVIIa to the surface of activated platelets (Hoffman 2003;
Logan and Goodnough 2010).
11.2.5.3 Indications
On-label indications include hemophilia A or B with inhibitors, congenital FVII
deficiency, and acquired hemophilia. The treatment of these rare disorders goes
beyond the scope of this chapter and will not be discussed in detail. Off-label indi-
cations for which clinical trial evidence exists, include body trauma, brain trauma,
cardiovascular surgery, intracerebral hemorrhage, upper GI bleeding in the context
of cirrhosis, liver transplantation, and hematopoietic stem cell transplantation
(Logan and Goodnough 2010).
196 L.M. Asmis
11.2.5.4 Monitoring
Monitoring of FVIIa treatment requires specialized coagulation tests. Classic coag-
ulation tests including prothrombin time, activated partial thromboplastin time, and
FVIII or FIX activity are unsuitable for this purpose. Some specialized tests, includ-
ing thrombin generation tests, thromboelastography/rotational thromboelastometry,
and aPTT waveform analysis, have been used (Hoffman and Dargaud 2012).
11.2.5.6 Dosing
Off-label use of FVIIa requires an individualized risk–benefit assessment, with a
particular focus on thromboembolic complications.
Reported dosing schemes in the on-label context range from 15 to 30 μg/kg every
4–6 h to 90 μg/kg every 2 h IV. In the off-label context, doses ranging 5–200 μg/kg
were studied (Warren et al. 2007; Logan and Goodnough 2010). In cases where an
off-label use appears unavoidable, it is the author’s opinion that the lowest possible
dose should be used (van de Garde et al. 2006; Narayan et al. 2008).
the population covers both sexes and all relevant age groups. The reference range
reflects the distribution of normal values for a given parameter and typically includes
the central 95 or 99 % of individuals. There is no evidence to suggest that values
below the lower limit of this range are associated with bleeding or that they might
represent a useful threshold for transfusion in the perioperative setting.
If the reference range is not the appropriate tool to define the parameter’s thresh-
old, another procedure must be agreed upon. To address this issue, there is a need
for large RCTs that show reduced mortality in transfused or substituted patients and
that define thresholds. Currently, such studies are not available. In their absence,
threshold levels are best defined locally. What are the important factors to consider
when defining a threshold?
The threshold question is test dependent on at least three levels. Classic coagula-
tion tests measure coagulation factor antigen or activity levels. Antigen assessments
are defined by weight per volume, while activity assays test a biological function in
relation to the amount of coagulation factor contained in 1 ml of plasma. It is unclear
which value is the more reliable for use in a transfusion algorithm. Moreover, for a
given test type, e.g., functional fibrinogen, test results can vary depending on which
test kit is used. Furthermore, fibrinogen measured using the same test can vary
between individual laboratory platforms.
The clinical context is another relevant factor to be considered when setting a
coagulation factor threshold; threshold levels for different indications may vary. For
hereditary disorders, such as afibrinogenemia or FXIII deficiency, guidelines and
experts propose 0.5 g/l and 5–10 % as levels generally sufficient for hemostasis
(Ciavarella et al. 1987; Anwar et al. 2002; Mannucci 2010). Recent data show that
these numbers are not appropriate in states of acquired deficiency. Instead, in the
setting of hypofibrinogenemia induced by acute bleeding, fibrinogen levels from 0.8
to 2.0 g/l have been proposed (Spahn et al. 2007; Rossaint et al. 2010).
Another relevant clinical factor is the hemostatic state. An actively bleeding patient
with acquired fibrinogen deficiency below a given cutoff may require treatment. In the
absence of bleeding, the treatment is likely to be different. Transfusion thresholds for
bleeding (therapeutic intention) and non-bleeding patient populations (prophylactic
intention) need to be established. If the volume of blood loss is used as the transfusion
trigger, then target concentrations should be defined. In fact, Rahe-Meyer et al. pub-
lished a randomized trial in major aortic surgery where the threshold for fibrinogen
transfusion was defined by the volume of blood loss; the target concentration was
defined for a FIBTEM MCF of 22 mm, corresponding approximately to a fibrinogen
level (Clauss) of 3.6 g/l (Rahe-Meyer et al. 2013b).
The timing of testing is another complicating factor. Classic coagulation tests are
performed in citrate plasma. The centrifugation of whole blood and the production
of cell-free citrate plasma necessitate time. Between drawing the blood, centrifuga-
tion, and producing the test result, a turnaround time of 30–40 min is common.
However, within this time span, an acutely bleeding patient’s coagulation status can
change dramatically, undergoing multiple transfusions and substitutions. To mini-
mize turnaround time in acutely bleeding patients, whole blood tests have been
designed utilizing POC testing devices. Fibrinogen tests exist for such POC test
198 L.M. Asmis
devices (FIBTEM for ROTEM and functional fibrinogen for TEG). These fibrino-
gen tests are performed on whole blood and measure other aspects of fibrinogen
than the classic coagulation tests. Because of their design as whole blood tests,
POC-based tests have a higher degree of outcome variability (Okuda et al. 2003;
Theusinger et al. 2010).
Large outcome-based studies are necessary to properly answer the transfusion
threshold question. These studies are scarce or nonexistent for isolated coagulation
factor products. In the absence of appropriate evidence, institutions should stan-
dardize their approach and define test procedures in order to optimize treatment
efficacy and patient safety. Locally defined thresholds should be reevaluated regu-
larly and changed in response to new evidence.
Isolated coagulation factor concentrates including fibrinogen, FXIII, vWF, and PCC
represent some of the therapeutic options for patient management in perioperative
settings. Evidence based on RCT is only beginning to emerge. Safety signals include
thromboembolic rates in the range of 1–3 % for PCC. These risks appear to be less
important for other isolated coagulation factor concentrates. Besides safety, health-
care cost needs to be integrated into the final risk–benefit evaluation of the use of
coagulation products. RCTs designed for perioperative settings and investigating
predefined treatment algorithms will help define the evidence-based strategies of
the future.
Key Points
With current knowledge and in the absence of RCT-based strategies, an individual-
ized risk–benefit evaluation should be performed for every patient prior to the
use of isolated coagulation factor concentrates.
Transfusion thresholds and targets depend on the clinical context (congenital versus
acquired hemostatic disorders, active bleeding versus no bleeding).
Transfusion thresholds and targets need to be validated locally.
Transfusion thresholds and targets need to be established in an evidence-based
context.
References
Acharya SS, Dimichele DM (2008) Rare inherited disorders of fibrinogen. Haemophilia
14(6):1151–1158
Aledort LM (2008) Factor VIII inhibitor bypassing activity (FEIBA) – addressing safety issues.
Haemophilia 14(1):39–43
Alter HJ, Klein HG (2008) The hazards of blood transfusion in historical perspective. Blood
112(7):2617–2626
Alter HJ, Holland PV et al (1972) Posttransfusion hepatitis after exclusion of commercial and
hepatitis-B antigen-positive donors. Ann Intern Med 77(5):691–699
11 Coagulation Factor Concentrates 199
Anwar R, Minford A et al (2002) Delayed umbilical bleeding–a presenting feature for factor XIII
deficiency: clinical features, genetics, and management. Pediatrics 109(2):E32
Barkan G (1923) To the question of reversibility of fibrin generation II. Biochem Ztschr
139:291–301
Berseus O, Boman K et al (2013) Risks of hemolysis due to anti-A and anti-B caused by the trans-
fusion of blood or blood components containing ABO-incompatible plasma. Transfusion
53(Suppl 1):114S–123S
Beste C, Ness V et al (2012) Mechanisms mediating parallel action monitoring in fronto-striatal
circuits. Neuroimage 62(1):137–146
Blomback B, Carlson LA et al (1966) Turnover of 131-I-labelled fibrinogen in man. Studies in
normal subjects, in congenital coagulation factor deficiency states, in liver cirrhosis, in polycy-
themia vera and in epidermolysis bullosa. Acta Med Scand 179(5):557–574
Boehlen F, Casini A et al (2013) Acquired factor XIII deficiency: a therapeutic challenge. Thromb
Haemost 109(3):479–487
Bolton-Maggs PH, Perry DJ et al (2004) The rare coagulation disorders–review with guidelines for
management from the United Kingdom Haemophilia Centre Doctors’ Organisation.
Haemophilia 10(5):593–628
Bosch J, Thabut D et al (2004) Recombinant factor VIIa for upper gastrointestinal bleeding in
patients with cirrhosis: a randomized, double-blind trial. Gastroenterology 127(4):
1123–1130
Bosch J, Thabut D et al (2008) Recombinant factor VIIa for variceal bleeding in patients with
advanced cirrhosis: a randomized, controlled trial. Hepatology 47(5):1604–1614
Bouma BN, Wiegerinck Y et al (1972) Immunological characterization of purified anti-haemophilic
factor A (factor VIII) which corrects abnormal platelet retention in Von Willebrand’s disease.
Nat New Biol 236(65):104–106
Chamouard P, Grunebaum L et al (1998) Significance of diminished factor XIII in Crohn’s disease.
Am J Gastroenterol 93(4):610–614
Ciavarella D, Reed RL et al (1987) Clotting factor levels and the risk of diffuse microvascular
bleeding in the massively transfused patient. Br J Haematol 67(3):365–368
Clauss A (1957) Rapid new physiological method for measurement of fibrinogen. Acta Haematol
17:237–246
Crawley JT, de Groot R et al (2011) Unraveling the scissile bond: how ADAMTS13 recognizes
and cleaves von Willebrand factor. Blood 118(12):3212–3221
Cromwell C, Aledort LM (2012) FEIBA: a prohemostatic agent. Semin Thromb Hemost
38(3):265–267
Dassanayaka S, Slaughter MS et al (2013) Mechanistic pathway(s) of acquired von willebrand
syndrome with a continuous-flow ventricular assist device: in vitro findings. ASAIO J
59(2):123–129
de Maat MP, Verschuur M (2005) Fibrinogen heterogeneity: inherited and noninherited. Curr Opin
Hematol 12(5):377–383
Dentali F, Marchesi C et al (2011) Safety of prothrombin complex concentrates for rapid antico-
agulation reversal of vitamin K antagonists. A meta-analysis. Thromb Haemost 106(3):
429–438
Didisheim P, Loeb J et al (1959) Preparation of a human plasma fraction rich in prothrombin,
proconvertin, Stuart factor, and PTC and a study of its activity and toxicity in rabbits and man.
J Lab Clin Med 53(2):322–330
Diprose P, Herbertson MJ et al (2005) Activated recombinant factor VII after cardiopulmonary
bypass reduces allogeneic transfusion in complex non-coronary cardiac surgery: randomized
double-blind placebo-controlled pilot study. Br J Anaesth 95(5):596–602
Duckert F, Jung E et al (1960) A hitherto undescribed congenital haemorrhagic diathesis probably
due to fibrin stabilizing factor deficiency. Thromb Diath Haemorrh 5:179–186
Dunbar NM, Ornstein DL et al (2012) ABO incompatible platelets: risks versus benefit. Curr Opin
Hematol 19(6):475–479
200 L.M. Asmis
Dzik WH, Blajchman MA et al (2011) Clinical review: Canadian National Advisory Committee
on Blood and Blood Products–Massive transfusion consensus conference 2011: report of the
panel. Crit Care (London, England) 15(6):242
Eerenberg ES, Kamphuisen PW et al (2011) Reversal of rivaroxaban and dabigatran by prothrombin
complex concentrate: a randomized, placebo-controlled, crossover study in healthy subjects.
Circulation 124(14):1573–1579
Evatt BL, Gomperts ED et al (1985) Coincidental appearance of LAV/HTLV-III antibodies in
hemophiliacs and the onset of the AIDS epidemic. N Engl J Med 312(8):483–486
Favaloro EJ (2011) Rethinking the diagnosis of von Willebrand disease. Thromb Res 127(Suppl 2):
S17–S21
Federici AB (2005) Management of von Willebrand disease with factor VIII/von Willebrand factor
concentrates: results from current studies and surveys. Blood Coagul Fibrinolysis 16(Suppl 1):
S17–S21
Federici AB (2009a) Classification of inherited von Willebrand disease and implications in clinical
practice. Thromb Res 124(Suppl 1):S2–S6
Federici AB (2009b) The safety of plasma-derived von Willebrand/factor VIII concentrates in the
management of inherited von Willebrand disease. Expert Opin Drug Saf 8(2):203–210
Federici AB, Berntorp E et al (2006) The 80th anniversary of von Willebrand’s disease: history,
management and research. Haemophilia 12(6):563–572
Federici AB, Budde U et al (2013) Current diagnostic and therapeutic approaches to patients
with acquired von Willebrand syndrome: a 2013 update. Semin Thromb Hemost
39(2):191–201
Fenger-Eriksen C, Ingerslev J et al (2009a) Fibrinogen concentrate–a potential universal hemo-
static agent. Expert Opin Biol Ther 9(10):1325–1333
Fenger-Eriksen C, Jensen TM et al (2009b) Fibrinogen substitution improves whole blood
clot firmness after dilution with hydroxyethyl starch in bleeding patients undergoing radical
cystectomy: a randomized, placebo-controlled clinical trial. J Thromb Haemost 7(5):
795–802
Fenger-Eriksen C, Tonnesen E et al (2009c) Mechanisms of hydroxyethyl starch-induced dilu-
tional coagulopathy. J Thromb Haemost 7(7):1099–1105
Franchini M (2008) Surgical prophylaxis in von Willebrand's disease: a difficult balance to manage.
Blood Transfus 6(Suppl 2):s33–s38
Fraser SR, Booth NA et al (2011) The antifibrinolytic function of factor XIII is exclusively
expressed through alpha(2)-antiplasmin cross-linking. Blood 117(23):6371–6374
George JN, Pickett EB et al (1986) Platelet membrane microparticles in blood bank fresh frozen
plasma and cryoprecipitate. Blood 68(1):307–309
Gill R, Herbertson M et al (2009) Safety and efficacy of recombinant activated factor VII: a ran-
domized placebo-controlled trial in the setting of bleeding after cardiac surgery. Circulation
120(1):21–27
Hardy JF (2012) Erythrocyte transfusions: an evidence-based approach. Ann Fr Anesth Reanim
31(7–8):617–625
Hartert H (1948) Studies on blood coagulation using thromboelastography: a novel diagnostic test.
Klin Wochenschr 26:577–585
Hedner U (2007) Recombinant factor VIIa: its background, development and clinical use. Curr
Opin Hematol 14(3):225–229
Hedner U (2012) Activated factor VII: my story. Haemophilia 18(2):147–151
Hedner U, Brun NC (2007) Recombinant factor VIIa (rFVIIa): its potential role as a hemostatic
agent. Neuroradiology 49(10):789–793
Hellstern P (1999) Production and composition of prothrombin complex concentrates: correlation
between composition and therapeutic efficiency. Thromb Res 95(4 Suppl 1):S7–S12
Hellstern P, Halbmayer WM et al (1999) Prothrombin complex concentrates: indications, contra-
indications, and risks: a task force summary. Thromb Res 95(4 Suppl 1):S3–S6
Hiippala ST, Myllyla GJ et al (1995) Hemostatic factors and replacement of major blood loss with
plasma-poor red cell concentrates. Anesth Analg 81(2):360–365
11 Coagulation Factor Concentrates 201
Hoffman M (2003) A cell-based model of coagulation and the role of factor VIIa. Blood Rev
17(Suppl 1):S1–S5
Hoffman M, Dargaud Y (2012) Mechanisms and monitoring of bypassing agent therapy. J Thromb
Haemost 10(8):1478–1485
Howard MA, Sawers RJ et al (1973) Ristocetin: a means of differentiating von Willebrand’s
disease into two groups. Blood 41(5):687–690
Hsieh L, Nugent D (2008) Factor XIII deficiency. Haemophilia 14(6):1190–1200
Inaba K, Branco BC et al (2010) Impact of plasma transfusion in trauma patients who do not
require massive transfusion. J Am Coll Surg 210(6):957–965
Karimi M, Bereczky Z et al (2009) Factor XIII deficiency. Semin Thromb Hemost 35(4):426–438
Karlsson M, Ternström L et al (2009) Prophylactic fibrinogen infusion reduces bleeding after coro-
nary artery bypass surgery. A prospective randomised pilot study. Thromb Haemost
102(1):137–144
Kasper CK (1975) Thromboembolic complications. Thromb Diath Haemorrh 33(3):640–644
Kerebel D, Joly LM et al (2013) A French multicenter randomised trial comparing two dose-
regimens of prothrombin complex concentrates in urgent anticoagulation reversal. Crit Care
17(1):R4
Kessler CM, Friedman K et al (2011) The pharmacokinetic diversity of two von Willebrand factor
(VWF)/factor VIII (FVIII) concentrates in subjects with congenital von Willebrand disease.
Results from a prospective, randomised crossover study. Thromb Haemost 106(2):279–288
Korte WC, Szadkowski C et al (2009) Factor XIII substitution in surgical cancer patients at high
risk for intraoperative bleeding. Anesthesiology 110(2):239–245
Kozek-Langenecker SA, Afshari A et al (2013) Management of severe perioperative bleeding:
guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 30(6):270–382
Kuo G, Choo Q et al (1989) An assay for circulating antibodies to a major etiologic virus of human
non-A, non-B hepatitis. Science 244(4902):362–364
Laffan M, Brown SA et al (2004) The diagnosis of von Willebrand disease: a guideline from the
UK Haemophilia Centre Doctors’ Organization. Haemophilia 10(3):199–217
Lawrie AS, Green L et al (2010) Factor XIII–an under diagnosed deficiency–are we using the right
assays? J Thromb Haemost 8(11):2478–2482
Legler TJ, Riggert J et al (2000) Testing of individual blood donations for HCV RNA reduces the
residual risk of transfusion-transmitted HCV infection. Transfusion 40(10):1192–1197
Leissinger CA, Blatt PM et al (2008) Role of prothrombin complex concentrates in reversing
warfarin anticoagulation: a review of the literature. Am J Hematol 83(2):137–143
Levi M, Levy JH et al (2010) Safety of recombinant activated factor VII in randomized clinical
trials. N Engl J Med 363(19):1791–1800
Levy JH, Greenberg C (2013) Biology of Factor XIII and clinical manifestations of Factor XIII
deficiency. Transfusion 53(5):1120–1131
Levy JH, Szlam F et al (2012) Fibrinogen and hemostasis: a primary hemostatic target for the
management of acquired bleeding. Anesth Analg 114(2):261–274
Lodge JP, Jonas S et al (2005) Efficacy and safety of repeated perioperative doses of recombinant
factor VIIa in liver transplantation. Liver Transpl 11(8):973–979
Logan AC, Goodnough LT (2010) Recombinant factor VIIa: an assessment of evidence regarding
its efficacy and safety in the off-label setting. Hematology Am Soc Hematol Educ Program
2010:153–159
Lowe GD, Rumley A et al (2004) Plasma fibrinogen. Ann Clin Biochem 41(Pt 6):430–440
Lubetsky A, Hoffman R et al (2004) Efficacy and safety of a prothrombin complex concentrate
(Octaplex) for rapid reversal of oral anticoagulation. Thromb Res 113(6):371–378
Majeed A, Eelde A et al (2012) Thromboembolic safety and efficacy of prothrombin complex
concentrates in the emergency reversal of warfarin coagulopathy. Thromb Res 129(2):
146–151
Makris M, Greaves M et al (1997) Emergency oral anticoagulant reversal: the relative efficacy of
infusions of fresh frozen plasma and clotting factor concentrate on correction of the coagulopathy.
Thromb Haemost 77(3):477–480
202 L.M. Asmis
Rasche H, Haghou F et al (1982) Blood clotting factor XIII substitution in acute leukaemia: result
of a randomized and controlled study. Dtsch Med Wochenschr 107(49):1882–1886
Richardson VR, Cordell P et al (2013) Substrates of Factor XIII-A: roles in thrombosis and wound
healing. Clin Sci (Lond) 124(3):123–137
Riess HB, Meier-Hellmann A et al (2007) Prothrombin complex concentrate (Octaplex) in patients
requiring immediate reversal of oral anticoagulation. Thromb Res 121(1):9–16
Robbins KC (1944) A study on the conversion of fibrinogen to fibrin. Am J Physiol 142:581–588
Rossaint R, Bouillon B et al (2010) Management of bleeding following major trauma: an updated
European guideline. Crit Care (London, England) 14(2):R52
Salzman EW (1963) Measurement of platelet adhesiveness. A simple in vitro technique demon-
strating an abnormality in Von Willebrand’s disease. J Lab Clin Med 62:724–735
Samama CM (2008) Prothrombin complex concentrates: a brief review. Eur J Anaesthesiol
25(10):784–789
Schlimp CJ, Cadamuro J et al (2013) The effect of fibrinogen concentrate and factor XIII on
thromboelastometry in 33% diluted blood with albumin, gelatine, hydroxyethyl starch or saline
in vitro. Blood Transfus 11:510–517
Schneppenheim R, Budde U (2011) von Willebrand factor: the complex molecular genetics of a
multidomain and multifunctional protein. J Thromb Haemost 9(Suppl 1):209–215
Schulz FH (1955) A simple volumetric determination of fibrinogen. Acta Hepatol 3:306–310
Shaw RE, Johnson CK et al (2013) Balancing the benefits and risks of blood transfusions in
patients undergoing cardiac surgery: a propensity-matched analysis. Interact Cardiovasc
Thorac Surg 17(1):96–102
Siegal DM, Cuker A (2013) Reversal of novel oral anticoagulants in patients with major bleeding.
J Thromb Thrombolysis 35(3):391–398
Sniecinski RM, Karkouti K et al (2012) Managing clotting: a North American perspective. Curr
Opin Anaesthesiol 25(1):74–79
Solomon C, Pichlmaier U et al (2010) Recovery of fibrinogen after administration of fibrinogen
concentrate to patients with severe bleeding after cardiopulmonary bypass surgery. Br J
Anaesth 104(5):555–562
Solomon C, Cadamuro J et al (2011) A comparison of fibrinogen measurement methods with fibrin
clot elasticity assessed by thromboelastometry, before and after administration of fibrinogen
concentrate in cardiac surgery patients. Transfusion 51(8):1695–1706
Sorensen B, Spahn DR et al (2011a) Clinical review: prothrombin complex concentrates–
evaluation of safety and thrombogenicity. Crit Care 15(1):201
Sorensen B, Tang M et al (2011b) The role of fibrinogen: a new paradigm in the treatment of
coagulopathic bleeding. Thromb Res 128(Suppl 1):S13–S16
Spahn DR, Cerny V et al (2007) Management of bleeding following major trauma: a European
guideline. Crit Care (London, England) 11(1):R17
Stroncek DF, Rebulla P (2007) Platelet transfusions. Lancet 370(9585):427–438
Sucker C, Michiels JJ et al (2009) Causes, etiology and diagnosis of acquired von Willebrand
disease: a prospective diagnostic workup to establish the most effective therapeutic strategies.
Acta Haematol 121(2–3):177–182
Theusinger OM, Nurnberg J et al (2010) Rotation thromboelastometry (ROTEM) stability and
reproducibility over time. Eur J Cardiothorac Surg 37(3):677–683
Theusinger OM, Baulig W et al (2011) Relative concentrations of haemostatic factors and cyto-
kines in solvent/detergent-treated and fresh-frozen plasma. Br J Anaesth 106(4):505–511
Thompson CA, Kyle R et al (2010) Systemic AL amyloidosis with acquired factor X deficiency:
a study of perioperative bleeding risk and treatment outcomes in 60 patients. Am J Hematol
85(3):171–173
Tiede A, Rand JH et al (2011) How I treat the acquired von Willebrand syndrome. Blood
117(25):6777–6785
Tosetto A, Rodeghiero F et al (2006) A quantitative analysis of bleeding symptoms in type 1 von
Willebrand disease: results from a multicenter European study (MCMDM-1 VWD). J Thromb
Haemost 4(4):766–773
204 L.M. Asmis
Tullis JL, Melin M et al (1965) Clinical use of human prothrombin complexes. N Engl J Med
273(13):667–674
Ulrich S, Brand B et al (2008) Congenital hypersensitivity to vitamin K antagonists due to FIX
propeptide mutation at locus −10: a (not so) rare cause of bleeding under oral anticoagulant
therapy in Switzerland. Swiss Med Wkly 138(7–8):100–107
Vadivel KB, Bajaj SP (2012) Structural biology of factor VIIa/tissue factor initiated coagulation.
Front Biosci 17:2476–2494
Vamvakas EC, Blajchman MA (2009) Transfusion-related mortality: the ongoing risks of allogeneic
blood transfusion and the available strategies for their prevention. Blood 113(15):3406–3417
van Aart L, Eijkhout HW et al (2006) Individualized dosing regimen for prothrombin complex
concentrate more effective than standard treatment in the reversal of oral anticoagulant therapy:
an open, prospective randomized controlled trial. Thromb Res 118(3):313–320
Van Bodegraven AA, Tuynman HA et al (1995) Fibrinolytic split products, fibrinolysis, and factor
XIII activity in inflammatory bowel disease. Scand J Gastroenterol 30(6):580–585
van de Garde EM, Bras LJ et al (2006) Low-dose recombinant factor VIIa in the management of
uncontrolled postoperative hemorrhage in cardiac surgery patients. J Cardiothorac Vasc Anesth
20(4):573–575
von Willebrand EA (1926) Hereditar pseudohemophili. Fin Lakaresallsk Handl 68:87–112
Warmuth M, Mad P et al (2012) Systematic review of the efficacy and safety of fibrinogen concen-
trate substitution in adults. Acta Anaesthesiol Scand 56(5):539–548
Warren O, Mandal K et al (2007) Recombinant activated factor VII in cardiac surgery: a systematic
review. Ann Thorac Surg 83(2):707–714
Weber CF, Gorlinger K et al (2012) Point-of-care testing: a prospective, randomized clinical trial
of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 117(3):531–547
Weber CF, Klages M et al (2013) Perioperative coagulation management during cardiac surgery.
Curr Opin Anaesthesiol 26(1):60–64
Weisel JW, Litvinov RI (2013) Mechanisms of fibrin polymerization and clinical implications.
Blood 121(10):1712–1719
Wiedermann CJ, Stockner I (2008) Warfarin-induced bleeding complications – clinical presentation
and therapeutic options. Thromb Res 122(Suppl 2):S13–S18
Zimmerman TS, Ratnoff OD et al (1971) Immunologic differentiation of classic hemophilia (factor 8
deficiency) and von Willebrand’s disease, with observations on combined deficiencies of anti-
hemophilic factor and proaccelerin (factor V) and on an acquired circulating anticoagulant
against antihemophilic factor. J Clin Invest 50(1):244–254
Procoagulant Drugs
12
Rainer B. Zotz, Nikola Zotz, and Christoph Sucker
Procoagulant drugs are non-transfusional agents that are primarily used when
bleeding is the consequence of a specific defect of hemostasis. In this chapter, the
pharmacological properties and uses of desmopressin, antifibrinolytics, and vitamin K
will be reviewed. Desmopressin, with its various procoagulant pharmacological
effects, is used in the prevention and treatment of bleeding related to congenital
and acquired coagulation factor deficiencies or platelet function disorders.
Antifibrinolytics (inhibitors in particular steps of fibrinolysis) are the treatment of
choice in bleeding caused by hyperfibrinolysis. Because of their low cost and their
mild side effects, desmopressin and antifibrinolytics are also used as blood-saving
agents in surgery. Vitamin K is given in states of vitamin K deficiency which lead to
vitamin K deficiency bleedings. This is relevant to patients taking vitamin K antago-
nists in cases of urgent invasive procedures, asymptomatic and excessively elevated
INR values, and bleeding.
12.2 Desmopressin
12.2.1 Description
Mannucci et al. 1986; Agnelli et al. 1995), but could not be confirmed in later trials
(Wong et al. 2003; Pivalizza et al. 2003).
Finally, drug-induced bleeding disorders, especially if caused by antiplatelet
agents, can be treated successfully with desmopressin (Levi et al. 2011).
Theoretically, desmopressin could also be used in an attempt to control acute bleed-
ing caused by novel anticoagulants (Brem et al. 2013).
Repeated doses of desmopressin over short intervals of time are associated with
a phenomenon known as tachyphylaxis – the progressively lower rises in the factor
VIII and vWF levels. This must be taken into account when planning treatment for
a prolonged period of time, e.g., in surgery with treatment lasting 7 days or longer.
The factor VIII level should be checked daily, and blood samples should be taken
1 h after the completion of the infusion. In such cases it is particularly important to
monitor the plasma sodium level (Vicente et al. 1993).
Desmopressin can cause side effects in about 30 % of patients, but in the vast major-
ity of cases, these are transient and mild. Administration is frequently accompanied
by headache, facial flushing, and a mild decrease in blood pressure and heart rate
(Mannucci 1998). More severe, but rarer, are episodes of fluid overload, severe
hyponatremia, and seizures due to the modest antidiuretic effect of the hemostatic
agent. This affects mostly very young patients after the administration of several
doses or patients receiving hypotonic fluids. Thus, desmopressin should be used
with caution in small children and patients with congestive heart failure or renal
insufficiency. Fluid intake should also be regulated.
There are reports of the occurrence of arterial thrombotic episodes associated
with the use of desmopressin, but no significant difference in the frequency of
venous and arterial thrombosis could be shown in cardiac, orthopedic, or other
major surgeries. Nevertheless, in patients with a history of cardiovascular events or
diffuse atherosclerosis, caution should be exercised when considering desmopressin
treatment (Castaman 2008).
Finally, desmopressin is not contraindicated in an uncomplicated pregnancy.
A recent review determined it had a good safety record and was effective in selected
cases in reducing bleeding complications associated with pregnancy and childbirth
(Trigg et al. 2012). However, as with all drugs, in this indication desmopressin
should be used with caution.
12.3 Antifibrinolytics
12.3.1 Description
and it has been used for years in cases of most types of bleeding or surgery in
patients with congenital or acquired bleeding disorders (Schulman 2012).
The antifibrinolytic amino acids tranexamic acid (trans-4-(aminomethyl)cyclo-
hexane carboxylic acid) and aminocaproic acid (6-aminohexanoic acid) both oper-
ate by blocking the lysine binding sites on plasminogen molecules, inhibiting the
formation of plasmin and therefore inhibiting fibrinolysis. Tranexamic acid is about
ten times more potent than aminocaproic acid and binds to both the strong and weak
sites of the plasminogen molecule to a higher extent (Mannucci 1998). The mechanism
also has a protective effect on thrombocytes, because the inhibited conversion of
plasminogen to plasmin prevents the plasmin-induced cleavage of several receptors
on thrombocytes (Quinton et al. 2004).
Because of their mode of action, antifibrinolytic amino acids are used in disease
patterns with an expected local or generalized hyperfibrinolysis. This is relevant for
nonsurgical and surgical bleeding. Recent studies and reviews have shown evidence
that tranexamic acid reduces blood transfusion particularly in patients undergoing
nonelective surgery.
Patients with congenital or acquired bleeding disorders may also benefit from the
use of antifibrinolytic amino acids in cases of epistaxis, gingival bleeding, or men-
orrhagia. Furthermore, these agents are useful for the prevention of bleeding follow-
ing minor surgical procedures or dental extractions (Seligsohn 2012).
The half-lives of tranexamic acid and aminocaproic acid are 2.3 and 2 h, respec-
tively. They can be administered orally (in the form of tablets or as an oral solution)
or intravenously.
For the treatment of acute bleeding syndromes, due to elevated fibrinolytic activity,
it is suggested that 5 g of aminocaproic acid be administered during the first hour of
treatment, followed by a continuous rate of 1 g/h. This method of treatment should
be continued for about 8 h or until the bleeding situation is under control. In trials
regarding the reduction of perioperative blood loss, dose regimens for aminocaproic
acid varied significantly. Loading or bolus doses ranged from 75 to 150 mg/kg;
maintenance doses ranged from 1 to 2 g/h or 12.5 to 30 mg/kg/h infused over vary-
ing time periods (Henry et al. 2011).
The dosage recommendations for tranexamic acid are as follows: as intravenous
injection in local fibrinolysis, 0.5–1 g two to three times daily, and in generalized
hyperfibrinolysis, 1 g (15 mg/kg) every 6–8 h. The general recommendations for oral
application are 3–4 g daily. In trials regarding the reduction of perioperative blood
loss, dose regimens for tranexamic acid differed significantly with varying doses and
time frames for drug administration. In trials involving cardiac surgery, the loading
or bolus doses ranged from 2.5 to 100 mg/kg. The maintenance doses for these car-
diac trials ranged from 0.25 to 4.0 mg/kg/h delivered over 1–12 h. A similar variation
was observed in trials not involving cardiac surgery (Henry et al. 2011). Dosing of
tranexamic acid must be reduced in patients with renal dysfunction.
The side effects of tranexamic acid and aminocaproic acid are dose dependent. They
usually involve the gastrointestinal tract (nausea, vomiting, abdominal pain, and
diarrhea). Headache and dizziness can also be observed. Sometimes allergic reac-
tions may occur. No striking increase in the risk of thrombosis was observed when
the drugs were used during operations (Mannucci 1998; Henry et al. 2011). The use
of tranexamic acid in moderate (24 mg/kg) to high doses (≥100 mg/kg) is associ-
ated with convulsive seizures after cardiopulmonary bypass (Kalavrouziotis et al.
2012; Koster et al. 2013).
214 R.B. Zotz et al.
12.4 Vitamin K
12.4.1 Description
Vitamin K was discovered by chance in 1929 and was immediately associated with
blood coagulation. Vitamin K is a group of structurally similar, fat-soluble vitamins
that cannot be synthesized by the human body, but are provided via nutrition and
gastrointestinal bacterial flora. This group of vitamins includes two natural vita-
mers: vitamin K1 and vitamin K2. Vitamin K1 – also known as phylloquinone, phy-
tomenadione, or phytonadione – is synthesized by plants, and the highest
concentration is found in leafy green vegetables. Vitamin K2 – also known as mena-
quinone – constitutes the main storage form in animals and has several subtypes
which differ in isoprenoid chain length. Vitamin K enables the posttranslational
γ-carboxylation of coagulation factors II (prothrombin), VII, IX, and X and pro-
teins C, S, and Z. Prothrombin and factors VII, IX, and X represent the classic
vitamin K-dependent plasma clotting factors and participate in the formation of a
fibrin clot. In contrast, proteins C, S, and Z are inhibitors of the procoagulant sys-
tem. Protein C exerts its inhibitory activity by inactivating factors Va and VIIIa and
enhances fibrinolysis with protein S as a cofactor. Protein Z serves as a cofactor for
the inhibition of factor Xa by protein Z-dependent protease inhibitor (Ferland
2012).
The only verified area of application of vitamin K is the therapy and prevention of
states of vitamin K deficiency which lead to vitamin K deficiency bleedings that
cannot be cured by nutrition. This includes vitamin K prophylaxis in neonates
immediately after delivery. The prophylactic administration of vitamin K to preg-
nant women treated with anticonvulsives, antituberculosis drugs, or coumarin deri-
vates does not seem to prevent deficiency in the newborn infants (Puckett and
Offringa 2009). Vitamin K deficiency bleeding has a low incidence in neonates and
infants as well as in adults. Reasons for vitamin K deficiency in adults include
chronic liver disease, short bowel syndrome, chronic inflammatory bowel disease,
and biliary tract obstruction leading to poor nutrition and malabsorption of fat-
soluble vitamins (De Simone and Sarode 2013). Until now, no randomized clinical
trials have been conducted to assess the benefits and harms of vitamin K use for
upper gastrointestinal bleeding in patients with acute or chronic liver disease. Its use
in this treatment can neither be recommended nor advised against (Marti-Carvajal
and Sola 2012). Impaired vitamin K synthesis can also be caused by a disturbance
of gut flora by a broad-spectrum antibiotic therapy.
The most frequent reason for severe vitamin K deficiency bleeding in adults is
the intake of vitamin K antagonists (coumarin derivates). Intracranial hemorrhage,
for instance, is seen in up to 2 % of patients receiving these drugs and has a mortal-
ity rate as high as 79 %. Patients with superwarfarin poisoning represent special
12 Procoagulant Drugs 215
The side effects and contraindications of vitamin K depend on the route of admin-
istration. The main disadvantages of oral administration are that complete absorp-
tion is not certain and can be adversely affected by vomiting or regurgitation. With
regard to intramuscular vitamin K administration, hematomas are possible.
Intravenous vitamin K administration carries the risk of allergic and anaphylactic
reactions, which might be lower when slow administration in saline solution is
used, e.g., over 30 min. Subcutaneous administration exhibits a lower risk of hema-
toma or anaphylaxis; however, drug absorption has been shown to be inconsistent
(Burke 2013).
216 R.B. Zotz et al.
References
Ageno W, Gallus AS, Wittkowsky A et al (2012) Oral anticoagulant therapy: antithrombotic
therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-
Based Clinical Practice Guidelines. Chest 141:e44S–e88S
Agnelli G, Parise P, Levi M et al (1995) Effects of desmopressin on hemostasis in patients with
liver cirrhosis. Haemostasis 25:241–247
Balduini CL, Noris P, Belletti S et al (1999) In vitro and in vivo effects of desmopressin on platelet
function. Haematologica 84:891–896
Baudo F, Collins P, Huth-Kühne A et al (2012) Management of bleeding in acquired hemophilia
A: results from the European Acquired Haemophilia (EACH2) Registry. Blood 120:39–46
Bonnar J, Sheppard BL (1996) Treatment of menorrhagia during menstruation: randomised con-
trolled trial of ethamsylate, mefenamic acid, and tranexamic acid. Br Med J 313:579–582
Brem E, Koyfman A, Foran M (2013) Review of recently approved alternatives to anticoagulation
with warfarin for emergency clinicians. J Emerg Med 45:143–149
Burke CW (2013) Vitamin k deficiency bleeding: overview and considerations. J Pediatr Health
Care 27:215–221
Burroughs AK, Matthews K, Qadiri M et al (1985) Desmopressin and bleeding time in patients
with cirrhosis. Br Med J 291:1377–1381
Carless PA, Henry DA, Moxey AJ et al (2004) Desmopressin for minimising perioperative allogeneic
blood transfusion. Cochrane Database Syst Rev (1):CD001884
Cash JD, Gader AM, da Costa J (1974) Proceedings: the release of plasminogen activator and factor
VIII to lysine vasopressin, arginine vasopressin, I-desamino-8-d-arginine vasopressin, angio-
tensin and oxytocin in man. Br J Haematol 27:363–364
Casonato A, Pontara E, Dannhaeuser D et al (1994) Re-evaluation of the therapeutic efficacy of
DDAVP in type IIB von Willebrand’s disease. Blood Coagul Fibrinolysis 5:959–964
Castaman G (2008) Desmopressin for the treatment of haemophilia. Haemophilia 14:15–20
Cattaneo M (2002) Desmopressin in the treatment of patients with defects of platelet function.
Haematologica 87:1122–1124
Coppola A, Di Minno G (2008) Desmopressin in inherited disorders of platelet function.
Haemophilia 14:31–39
CRASH-2 Collaborators (Intracranial Bleeding Study) (2011) Effect of tranexamic acid in trau-
matic brain injury: a nested randomised, placebo controlled trial (CRASH-2 Intracranial
Bleeding Study). BMJ 343:d3795
Crescenzi G, Landoni G, Biondi-Zoccai G et al (2008) Desmopressin reduces transfusion needs
after surgery: a meta-analysis of randomized clinical trials. Anesthesiology 109:1063–1076
De Simone N, Sarode R (2013) Diagnosis and management of common acquired bleeding disorders.
Semin Thromb Hemost 39:172–181
Favaloro EJ (2011) Diagnosis and classification of von Willebrand disease: a review of the
differential utility of various functional von Willebrand factor assays. Blood Coagul Fibrinolysis
22:553–564
Federici AB, Budde U, Castaman G et al (2013) Current diagnostic and therapeutic approaches to
patients with acquired von Willebrand syndrome: a 2013 update. Semin Thromb Hemost
39:191–201
Fergusson DA, Hebert PC, Mazer CD et al (2008) A comparison of aprotinin and lysine analogues
in high-risk cardiac surgery. N Engl J Med 358:2319–2331
Ferland G (2012) The discovery of vitamin K and its clinical applications. Ann Nutr Metab
61:213–218
Franchini M (2007) The use of desmopressin as a hemostatic agent: a concise review. Am J
Hematol 82:731–735
Franchini M, Lippi G (2011) The use of desmopressin in acquired haemophilia A: a systematic
review. Blood Transfus 9:377–382
Franchini M, Manzato F, Salvagno GL et al (2009) The use of desmopressin in congenital factor
XI deficiency: a systematic review. Ann Hematol 88:931–935
12 Procoagulant Drugs 217
Morrison JJ, Dubose JJ, Rasmussen TE et al (2012) Military Application of Tranexamic Acid in
Trauma Emergency Resuscitation (MATTERs) Study. Arch Surg 147:113–119
Murphy GJ, Reeves BC, Rogers CA et al (2007) Increased mortality, postoperative morbidity, and
cost after red blood cell transfusion in patients having cardiac surgery. Circulation
116:2544–2552
Nurden AT, Freson K, Seligsohn U (2012) Inherited platelet disorders. Haemophilia 18:154–160
Pereira A, del Valle OM, Sanz C (2003) DDAVP enhances the ability of blood monocytes to form
rosettes with activated platelets by increasing the expression of P-selectin sialylated ligands on
the monocyte surface. Br J Haematol 120:814–820
Perel P, Ker K, Morales Uribe CH et al (2013) Tranexamic acid for reducing mortality in emer-
gency and urgent surgery. Cochrane Database Syst Rev (1):CD010245
Pivalizza EG, Warters RD, Gebhard R (2003) Desmopressin before liver transplantation. Can J
Anaesth 50:748–749
Podda G, Femia EA, Pugliano M et al (2012) Congenital defects of platelet function. Platelets
23:552–563
Puckett RM, Offringa M (2009) Prophylactic vitamin K for vitamin K deficiency bleeding in
neonates. Cochrane Database Syst Rev (4):CD002776
Quinton TM, Kim S, Derian CK et al (2004) Plasmin-mediated activation of platelets occurs by
cleavage of protease-activated receptor 4. J Biol Chem 279:18434–18439
Ragni MV, Novelli EM, Murshed A et al (2013) Phase II prospective open-label trial of recombinant
interleukin-11 in desmopressin-unresponsive von Willebrand disease and mild or moderate
haemophilia A. Thromb Haemost 109:248–254
Rappold JF, Pusateri AE (2013) Tranexamic acid in remote damage control resuscitation.
Transfusion 53:96S–99S
Roberts I, Shakur H, Ker K et al (2011) Antifibrinolytic drugs for acute traumatic injury. Cochrane
Database Syst Rev (1):CD004896
Sabate A, Dalmau A, Koo M et al (2012) Coagulopathy management in liver transplantation.
Transplant Proc 44:1523–1525
Sadler JE (1998) Biochemistry and genetics of von Willebrand factor. Annu Rev Biochem
67:395–424
Sakariassen KS, Cattaneo M, vd Berg A et al (1984) DDAVP enhances platelet adherence and
platelet aggregate growth on human artery subendothelium. Blood 64:229–236
Salzman EW, Weinstein MJ, Weintraub RM et al (1986) Treatment with desmopressin acetate to
reduce blood-loss after cardiac-surgery - a double-blind randomized trial. N Engl J Med
314:1402–1406
Schulman S (2012) Pharmacologic tools to reduce bleeding in surgery. Hematology Am Soc
Hematol Educ Program 2012:517–521
Seligsohn U (2012) Treatment of inherited platelet disorders. Haemophilia 18:156
Shakur H, Roberts I, Bautista R et al (2010) Effects of tranexamic acid on death, vascular occlusive
events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a
randomised, placebo-controlled trial. Lancet 376:23–32
Sucker C, Michiels JJ, Zotz RB (2009) Causes, etiology and diagnosis of acquired von Willebrand
disease: a prospective diagnostic workup to establish the most effective therapeutic strategies.
Acta Haematol 121:177–182
Tiede A, Rand JH, Budde U et al (2011) How I treat the acquired von Willebrand syndrome. Blood
117:6777–6785
Trigg DE, Stergiotou I, Peitsidis P et al (2012) A systematic review: the use of desmopressin for
treatment and prophylaxis of bleeding disorders in pregnancy. Haemophilia 18:25–33
Vavra I, Machova A, Holeced V et al (1968) Effect of a synthetic analogue of vasopressin in ani-
mals and in patients with diabetes insipidus. Lancet 1:948–952
Vicente V, Estelles A, Laso J et al (1993) Repeated infusions of Ddavp induce low response of
Fviii and Vwf but not of plasminogen activators. Thromb Res 70:117–122
Villar A, Jimenez-Yuste V, Quintana M et al (2002) The use of haemostatic drugs in haemophilia:
desmopressin and antifibrinolytic agents. Haemophilia 8:189–193
12 Procoagulant Drugs 219
Watts CD, Pagnano MW (2012) Minimising blood loss and transfusion in contemporary hip and
knee arthroplasty. J Bone Joint Surg Br 94B:8–10
Weiss HJ, Sussman II, Hoyer LW (1977) Stabilization of factor VIII in plasma by the von
Willebrand factor. Studies on posttransfusion and dissociated factor VIII and in patients with
von Willebrand’s disease. J Clin Invest 60:390–404
Wise RJ, Dorner AJ, Krane M et al (1991) The role of von Willebrand factor multimers and propeptide
cleavage in binding and stabilization of factor VIII. J Biol Chem 266:21948–21955
Wong AYC, Irwin MG, Hui TWC et al (2003) Desmopressin does not decrease blood loss and
transfusion requirements in patients undergoing hepatectomy. Can J Anaesth 50:14–20
Zotz RB (2009) A Stratified Metaanalysis of Desmopressin (DDAVP) for Minimising Perioperative
Allogeneic Blood Transfusion. Blood (ASH Annual Meeting Abstracts), 2009;114:1306
Patient Blood Management
13
Oliver M. Theusinger, Stephanie L. Kind,
and Donat R. Spahn
13.1 Introduction
budget of developed countries (Morton et al. 2010). The known and hidden costs,
including the treatment of side effects, have been estimated close to USD 2,000 per
red blood cell (RBC) unit in the USA (Shander et al. 2010; Ferraris et al. 2012). As
blood donations are generally decreasing and previously regular blood donors grow
older, a shortage of blood products is predicted (Borkent-Raven et al. 2010).
The Society for the Advancement of Blood Management defines patient blood
management as “the appropriate use of blood and blood components, with a goal of
minimizing their use”.
The aim of patient blood management is to preoperatively identify patients at
risk and offer individualized therapy to decrease the likelihood of transfusion
(Williams and McCarthy 2003; Goodnough and Shander 2007; Spahn et al.
2008; Gombotz et al. 2011b), thereby improving outcome, and reducing side
effects and overall costs. Avoiding RBC transfusion completely by introducing
a patient blood management program is not possible, nevertheless a decrease in
provided and used blood products has been shown after implementation (Wells
et al. 2002; Cobain et al. 2007; Gombotz et al. 2007; Moskowitz et al. 2010;
Kotze et al. 2012).
Patient blood management is a multi-disciplinary approach built upon three
pillars: treatment of preoperative anemia, minimizing intraoperative blood loss,
and increasing tolerance of anemia (Gombotz et al. 2011a; Spahn et al. 2012)
(Table 13.1). Ideally, improvements in all three are strived for, but optimizing even
one pillar reduces transfusion needs drastically. In 94 % of elective interventions,
intraoperative RBC transfusion needs can be predicted from the preoperative hemo-
globin level, possible blood loss, and individual transfusion triggers (Gombotz et al.
2007; Gombotz 2011). In May 2010, the WHO recognized patient blood manage-
ment as a significant means to improve transfusion safety, and since then it has
promoted its implementation in order to improve patient care.
13 Patient Blood Management 223
The following chapter provides an overview of all the aspects that have to be
taken into consideration and suggests what such a program could, or should,
look like.
One unit of RBCs transfused to adults increases the hemoglobin level by approxi-
mately 1 g/l and the hematocrit by about 3 % (Liumbruno et al. 2009, 2011).
Blood products are frequently administered to older and sicker patients, there-
fore causing more serious complications and various side effects. Current esti-
mates of viral transmission via blood products are 1:280,000–1:357,000 for
hepatitis B, 1:1,149,000 for hepatitis C, and 1:1,467,000 for human immunodefi-
ciency virus (HIV) (Epstein and Holmberg 2010). Although viral and bacterial
transmission via RBC units is considered to be under control in developed coun-
tries (Pomper et al. 2003; Goodnough 2005), RBC transfusion increases morbid-
ity and mortality several fold (Klein et al. 2007; Murphy et al. 2007; Spahn et al.
2008; Jakobsen et al. 2012). Effects on mortality are most commonly seen close
to the time of transfusion, but effects have been shown up to 5 years after the
initial transfusion (Reeves and Murphy 2008a, b; Jakobsen et al. 2012). Because
of transfusion-related immunomodulation (TRIM), a higher incidence of postop-
erative infection and cancer recurrence is observed (Amato and Pescatori 2006 ).
Immunosuppression is caused by a decreased activity of T-cells, macrophages,
and other natural killer cells. Blood transfusion is also associated with an increased
risk of developing non-Hodgkin lymphoma (NHL), chronic lymphocytic leuke-
mia, and small lymphocytic lymphoma, up to 15 years after transfusion (Castillo
et al. 2010). Furthermore, it is assumed that protein misfolding diseases such as
Alzheimer’s disease might be transmitted through blood transfusions, however
further studies need to be performed (Morales et al. 2011; de Calignon et al.
2012).
Besides an increased frequency of MOF, thromboembolic events such as myo-
cardial infarction and stroke are correlated to blood transfusion (Moore et al. 1997).
Overall, around 4 % of RBC transfusions are associated with transfusion-related
complications.
It is therefore fundamental to accept intraoperative hemoglobin levels of between
6 and 7 g/dl in order to reduce possible side effects (Spahn and Madjdpour 2006;
Spahn et al. 2008). Perioperative anemia does not increase postoperative mortality
or morbidity in cardiac surgery, whereas restrictive transfusion recommendations
decrease complications (Moskowitz et al. 2010).
Adverse effects are directly dependent on the number of units transfused and the
mean storage time (Offner 2004; Shorr et al. 2004; Koch et al. 2008; Spinella et al.
2009; Ness 2011; Tung et al. 2012). Even transfusion of a single unit has been asso-
ciated with increased mortality, renal dysfunction, and sepsis (Rogers et al. 2006;
Ferraris et al. 2012). Longer stored erythrocytes have significantly more harmful
effects, such as increased in-hospital mortality, increased 1 year mortality, renal
failure, and sepsis (Spahn et al. 2008; Belizaire et al. 2012). Ventilator associated
pneumonia is increased by 1 % per day of mean storage time (Shorr et al. 2004;
Bernard et al. 2009). This is due to the time-dependent metabolic, biochemical, and
molecular changes undergone by stored RBCs. These include: adenosine triphos-
phate (ATP) depletion; bioactive generation of histamine, cytokines, and lipids;
reduction of nitric oxide; and a decrease of 2.3-diphosphoglycerate leading to a
left-shift of the oxyhemoglobin dissociation curve (Chin-Yee et al. 1997; Offner
2004; Raat et al. 2005). Further, stored RBCs have a higher inflammatory potential,
due to increased vascular endothelial growth factor levels; they are also stiffer.
Increased hemolysis of aged RBCs after massive transfusion has recently been
shown in a guinea pig model. Cell-free, plasma hemoglobin led to vascular injury
and renal insufficiency (Upile et al. 2009; Urner et al. 2012). Hemoglobin toxicity
was prevented by co-infusion of haptoglobin, showing possible protective effects
(Urner et al. 2012).
Unfortunately it is not yet clear exactly when negative effects occur, though a
storage time over 14 days has been associated with negative outcomes such as
increased in-hospital mortality, renal failure, sepsis, and intubation prolonged
beyond 72 h (Koch et al. 2008; Blasi et al. 2012).
13 Patient Blood Management 225
Preoperative
- Timing of surgical intervention depending on - Careful hemostasis and surgical technique - Optimize cardiac output
the concentration of preoperative erythrocytes - (minimal invasive) - Optimize mechanical ventilation
- Use of measures to avoid allogeneic blood - Strict indication for blood transfusion
- Hemostasis and coagulation monitoring
- Use of hemostats
Intraoperative
- Stimulation of erythropoiesis with iron and/or - Monitor bleeding and prevent post-bleeding - Optimize reserve of anemia
EPO - Hemostasis and coagulation monitoring - Optimize O2-supply
- Be aware of drug interaction that can increase - Minimize diagnostic and interventional loss of - Reduction of O2-consumption
anemia blood - Avoidance and prompt treatment of infections
- Prophylaxis of upper gastrointestinal bleeding - Strict indication for blood transfusion
- Avoidance and prompt treatment of infections
Postoperative
- Autologous blood conservation methods
Re-evaluation
Fig. 13.1 Strategies for Pre-, per- and postoperative Patient Blood Management
O.M. Theusinger et al.
13 Patient Blood Management 227
mortality and a 30 % increase of morbidity. This risk is doubled when blood products
are transfused (Musallam et al. 2011).
Postoperative anemia is common. Moderate postoperative anemia does, however,
not influence rehabilitation (Carson et al. 2011; Vuille-Lessard et al. 2012). In
order to minimize the degree of postoperative anemia or to reduce its incidence,
patient blood management focuses on the detection and treatment of preoperative
anemia and the reduction of RBC loss during surgery (Westenbrink et al. 2011).
anemia influences the course of diseases negatively (Endres et al. 2009). Preoperative
iron treatment, as well as the use of EPO in orthopedic surgery, reduces the rate of
allogeneic blood transfusions and infections (Spahn 2010). Today, 90 % of existing
preoperative anemia is not treated prior to surgery with expected significant blood
loss, resulting in a three to fourfold increase in the transfusion rate (2nd Austrian
Benchmark study; http://www.bmg.gv.at).
13.3.2.2 Pharmacotherapy
Calcium is crucial in all phases of coagulation and plasma concentration of ionized
calcium should be kept between 1.15 and 1.29 mmol/l in bleeding patients (Lier
et al. 2008). In lactic acidosis (blood loss, vasoconstrictions, hypoxia), plasma lac-
tate and free ionized calcium levels are inversely related. It has been shown that
acidosis affects clot firmness and is not restored by sodium bicarbonate infusion
(Martini et al. 2006). Calcium is further depleted by the transfusion of citrate con-
taining allogeneic blood products (Vivien et al. 2005). Hence, acidosis, hypother-
mia, and hypocalcaemia must be avoided and treated at all times.
To reduce intraoperative blood loss, the Western Australian Blood Authority rec-
ommends the use of perioperative vasoconstrictors such as diluted epinephrine in
13 Patient Blood Management 229
combination with local anesthesia. Topical agents, like collagen or thrombin, are
additional means of reducing intraoperative blood loss (Achneck et al. 2010).
Other blood saving techniques include catecholamine administration and the
application of systemic antifibrinolytics, such as aminocaproic acid or tranexamic
acid (Gombotz 2011). Both compounds are synthetic lysine analogues that inhibit
fibrinolysis by blocking the lysine-binding site on plasminogen, thus preventing
fibrin from binding to plasminogen (Holcomb 2004; Zufferey et al. 2006).
13.3.2.4 Monitoring
Point of care (POC) monitoring devices, such as TEG® (Haemonetics® Corporation,
Braintree, MA, USA) or ROTEM® (TEM® International GmbH, Munich, Germany),
are increasingly used to monitor coagulation in vivo. Hemostatic tests are performed
on whole blood, evaluating platelet interaction with coagulation factors in all three
phases of coagulation: initiation, propagation, and lysis of the clot (Ganter and
Hofer 2008; Theusinger et al. 2010). Because the results are shown in real time,
clinicians can distinguish between surgical bleeding, hemostasis disorders, and
hyperfibrinolysis, and assess the extent of dilutional coagulopathy.
230 O.M. Theusinger et al.
The results of POC tests can guide the transfusion of blood products or the
administration of coagulation factors. The use of a POC, goal directed transfusion
algorithm improves outcomes in cases of massive hemorrhage (Spahn et al. 2008,
2012; Ganter and Spahn 2010; Gorlinger et al. 2011; Liumbruno et al. 2011; Young
et al. 2011; Theusinger et al. 2014).
Ideally, coagulation should be monitored using viscoelastic POC tests before and
after therapy, and 30 min after every administration of Factor XIII (Fibrogammin®).
Figure 13.2 illustrates just one possible means of handling massive bleeding in a
clinical setting and needs to be adjusted to local recommendations. The early use of
coagulation factors combined with POC testing leads to a reduction of RBC transfu-
sions, decreased incidence of thrombotic events, and better outcomes (Gorlinger
et al. 2011; Weber et al. 2012). Finally, the intraoperative collaboration between
anesthesiologists and surgeons can reduce intraoperative blood loss up to 50 %
(Filicori et al. 2010; Zwart et al. 2010).
Due to a correlation between physical symptoms and the amount of blood loss,
the American Society of Anesthesiologists (ASA) recommends using physiologi-
cal transfusions triggers to identify transfusion needs (Ferraris et al. 2011;
Carson et al. 2012a). Examples are tachycardia, hypotension, mixed venous oxy-
gen pressure of less than 32 mmHg, electrocardiogram (ECG) changes, oxygen
extraction greater than 50 %, or an increase in lactate (Madjdpour and Spahn
2005; Spahn and Madjdpour 2006; Ferraris et al. 2007) (Table 13.3). With hyper-
oxic ventilation in patients with critical hemoglobin values, tissue oxygenation is
not impaired (Meier et al. 2004; Weiskopf 2010). Thus a high fraction of inspired
oxygen (FiO2) is a possible bridge until blood products are available (Weiskopf
2010).
Low hemoglobin transfusion triggers should be applied. While the use of
restrictive transfusion triggers has not been shown to increase adverse outcomes,
a reduction in the RBC transfusion and infection rate can be found, leading to
decreased mortality and overall cost (Carless et al. 2010; Carson et al. 2012a). If
cardiovascular comorbidities are treated preoperatively, postoperative hemoglo-
bin values of 8 g/dl in orthopedic patients are well tolerated (Grover and
McManus 2006).
The individual minimal hematocrit depends on the patient and his/her comor-
bidities. In general surgical patients, hemoglobin levels of <7 g/dl are tolerated
(Carless et al. 2010), and <8 g/dl is accepted in high risk patients (Carson et al.
2011, 2012b). Elderly patients without cardiovascular disease tolerate hemoglobin
values of 9 g/dl (Spahn et al. 1996).
13 Patient Blood Management 231
Diagnostic Intervention
®
Preoperative history ROTEM after anesthesia induction
1. Drugs affecting coagulation - Transplant surgery
- Antiplatelet drugs - Cardiac and vascular surgery
- Heparin - Difficult cancer surgery
- Oral anticoagulation (Vit. K antagonists, - Liver insufficiency
Xa antagonists, IIa antagonists) - Intra-abdominal sepsis
2. Coagulation status? - Emergency room entry
3. HIT II?
INTEM (CT and CFT prolonged) and HEPTEM normal Protamine sulfate 1:1 to heparin
OR crystalloid and colloid volume substitution
ACT pathological and heparinase ACT normal
EXTEM/INTEM:
Decrease of MCF after maximum was reached Tranexamic acid
APTEM: normal - 15 mg/kg BW as bolus iv
HYPERFIBRINOLYSIS - 1–2 mg/kg/h during surgery iv as perfusion
Quick’s value < 30% and 4 factor prothrombin complex concentrate 1000–2000 IU
Factor V > 20%
- Factor II, VII, IX and X
OR
EXTEM/INTEM: CT, CFT prolonged Depending on the patients’ body weight
Fig. 13.2 Second version of the transfusion algorithm of the University Hospital of Zürich 2012©,
Switzerland with permission from Theusinger et al. (2014)
232 O.M. Theusinger et al.
In recent years, the doctrine for resuscitation of patients suffering from massive bleed-
ing has changed from a supportive treatment with crystalloids/colloids and RBC units,
to a standardized massive transfusion protocol (Young et al. 2011). In 2002, the
Province of Ontario, Canada, introduced the pioneering Ontario Nurse Transfusion
Coordinators Provincial Blood Conservation Program (ONTraC). A decrease of RBC
transfusion of 14–24 % was noticed 1 year after implementation (Freedman et al.
2005). To date, based on decreased mortality and morbidity, patient blood manage-
ment programs have shown themselves to be efficient in all medical fields, in vari-
ous countries (Moskowitz et al. 2010; Spahn 2010; Goodnough et al. 2011; Kotze
et al. 2012).
Western Australia’s patient blood management program is probably the best
known: it is very well organized and administered by the state government and all
major hospitals are included.
By carrying out RBC transfusion only when there is no other alternative and
thus reducing the use of blood products, health care costs are reduced. Due to
improved collection, testing and processing, the direct costs of blood products
have progressively increased (Kamper-Jorgensen et al. 2010; Abraham and Sun
2012). As stated in the introduction, the known and hidden costs have been esti-
mated to be USD 2,000 (Ferraris et al. 2012; Shander et al. 2010) per RBC unit.
USD 1.62 to USD 6.03 million per year and hospital are spent on transfusion
13 Patient Blood Management 233
related activities in surgical specialties only. Only 30 % of patients who have their
blood-typed and screened eventually receive peri- or postoperative transfusions.
These non-transfused patients contribute substantially to overall costs (Shander
et al. 2010). The introduction of a patient blood management program signifi-
cantly reduces the prevalence of anemia, transfusion rates, the length of hospital
stays, and re-admission rates, while also reducing costs (Kotze et al. 2012; Spahn
et al. 2012).
The patient blood management paradigm shift in transfusion medicine (Farrugia
2011) not only saves lives, but reduces health care costs (Spahn et al. 2012).
It should be therefore part of good clinical practice.
References
Abraham I, Sun D (2012) The cost of blood transfusion in Western Europe as estimated from six
studies. Transfusion 52:1983–1988
Achneck HE, Sileshi B et al (2010) A comprehensive review of topical hemostatic agents: efficacy
and recommendations for use. Ann Surg 251(2):217–228
Addeo R, Caraglia M et al (2009) Two faces for Janus: recombinant human erythropoiesis-
stimulating agents and cancer mortality. Expert Rev Hematol 2(5):513–515
Amato A, Pescatori M (2006) Perioperative blood transfusions for the recurrence of colorectal
cancer. Cochrane Database Syst Rev 25;(1):CD005033.
Auerbach M, Rodgers GM (2007) Intravenous iron. N Engl J Med 357(1):93–94
Australian Patient Blood Management Guidelines. http://www.nba.gov.au/guidelines/module2/po-
mod2.pdf
Belizaire RM, Prakash PS et al (2012) Microparticles from stored red blood cells activate neutrophils
and cause lung injury after hemorrhage and resuscitation. J Am Coll Surg 214(4):648–655
Bernard AC, Davenport DL et al (2009) Intraoperative transfusion of 1 U to 2 U packed red blood
cells is associated with increased 30-day mortality, surgical-site infection, pneumonia, and sepsis
in general surgery patients. J Am Coll Surg 208(5):931–937, 937 e931–932; discussion
938–939
Bierbaum BE, Callaghan JJ et al (1999) An analysis of blood management in patients having a
total hip or knee arthroplasty. J Bone Joint Surg Am 81(1):2–10
Blasi B, D’Alessandro A et al (2012) Red blood cell storage and cell morphology. Transfus Med
22(2):90–96
Borkent-Raven BA, Janssen MP et al (2010) The PROTON study: profiles of blood product trans-
fusion recipients in the Netherlands. Vox Sang 99(1):54–64
Brookhart MA, Schneeweiss S et al (2010) Comparative mortality risk of anemia management
practices in incident hemodialysis patients. JAMA 303(9):857–864
Buddeberg F, Schimmer BB et al (2008) Transfusion-transmissible infections and transfusion-
related immunomodulation. Best Pract Res Clin Anaesthesiol 22(3):503–517
Carless PA, Henry DA et al (2010) Transfusion thresholds and other strategies for guiding alloge-
neic red blood cell transfusion. Cochrane Database Syst Rev (10):CD002042
Carson JL, Terrin ML et al (2011) Liberal or restrictive transfusion in high-risk patients after hip
surgery. N Engl J Med 365(26):2453–2462
Carson JL, Carless PA et al (2012a) Transfusion thresholds and other strategies for guiding alloge-
neic red blood cell transfusion. Cochrane Database Syst Rev 4, CD002042
Carson JL, Grossman BJ et al (2012b) Red blood cell transfusion: a clinical practice guideline
from the AABB. Ann Intern Med 157:49–58
Castillo JJ, Dalia S et al (2010) Association between red blood cell transfusions and development of
non-Hodgkin lymphoma: a meta-analysis of observational studies. Blood 116(16):2897–2907
234 O.M. Theusinger et al.
Chaiwat O, Lang JD et al (2009) Early packed red blood cell transfusion and acute respiratory
distress syndrome after trauma. Anesthesiology 110(2):351–360
Chang CH, Chang CC et al (2002) Reduction in erythropoietin doses by the use of chronic intrave-
nous iron supplementation in iron-replete hemodialysis patients. Clin Nephrol 57(2):136–141
Chin-Yee I, Arya N et al (1997) The red cell storage lesion and its implication for transfusion.
Transfus Sci 18(3):447–458
Claridge JA, Sawyer RG et al (2002) Blood transfusions correlate with infections in trauma
patients in a dose-dependent manner. Am Surg 68(7):566–572
Cobain TJ, Vamvakas EC et al (2007) A survey of the demographics of blood use. Transfus Med
17(1):1–15
Corwin HL, Gettinger A et al (2004) The CRIT study: anemia and blood transfusion in the criti-
cally ill–current clinical practice in the United States. Crit Care Med 32(1):39–52
Cosgrove DM, Loop FD et al (1985) Determinants of blood utilization during myocardial revascu-
larization. Ann Thorac Surg 40(4):380–384
Cuenca J, Garcia-Erce JA et al (2004) Patients with pertrochanteric hip fracture may benefit from
preoperative intravenous iron therapy: a pilot study. Transfusion 44(10):1447–1452
Dara SI, Rana R et al (2005) Fresh frozen plasma transfusion in critically ill medical patients with
coagulopathy. Crit Care Med 33(11):2667–2671
de Calignon A, Polydoro M et al (2012) Propagation of tau pathology in a model of early
Alzheimer’s disease. Neuron 73(4):685–697
Emmert MY, Salzberg SP et al (2011) How good patient blood management leads to excellent
outcomes in Jehovah’s witness patients undergoing cardiac surgery. Interact Cardiovasc Thorac
Surg 12(2):183–188
Endres HG, Wedding U et al (2009) Prevalence of anemia in elderly patients in primary care:
impact on 5-year mortality risk and differences between men and women. Curr Med Res Opin
25(5):1143–1158
Epstein JS, Holmberg JA (2010) Progress in monitoring blood safety. Transfusion
50(7):1408–1412
Etchason J, Petz L et al (1995) The cost effectiveness of preoperative autologous blood donations.
N Engl J Med 332(11):719–724
Farrugia A (2011) Falsification or paradigm shift? Toward a revision of the common sense of
transfusion. Transfusion 51(1):216–224
Ferraris VA, Ferraris SP et al (2007) Perioperative blood transfusion and blood conservation in
cardiac surgery: the Society of Thoracic Surgeons and The Society of Cardiovascular
Anesthesiologists clinical practice guideline. Ann Thorac Surg 83(5 Suppl):S27–S86
Ferraris VA, Brown JR et al (2011) 2011 update to the Society of Thoracic Surgeons and the
Society of Cardiovascular Anesthesiologists blood conservation clinical practice guidelines.
Ann Thorac Surg 91(3):944–982
Ferraris VA, Davenport DL et al (2012) Surgical outcomes and transfusion of minimal amounts of
blood in the operating room. Arch Surg 147(1):49–55
Filicori F, Di Saverio S et al (2010) Packing for damage control of nontraumatic intra-abdominal
massive hemorrhages. World J Surg 34(9):2064–2068
Freedman J, Luke K et al (2005) A provincial program of blood conservation: the Ontario
Transfusion Coordinators (ONTraC). Transfus Apher Sci 33(3):343–349
Freischlag JA (2004) Intraoperative blood salvage in vascular surgery – worth the effort? Crit Care
8(Suppl 2):S53–S56
Fukusaki M, Sumikawa K (2000) The combination of hemodilution and controlled hypotension:
physiology and clinical application. J Anesth 14(4):194–203
Fukusaki M, Maekawa T et al (1997) Acute haemodilution and prostaglandin E1-induced hypoten-
sion: effects on the coagulation-fibrinolysis system. Eur J Anaesthesiol 14(4):443–449
Gajic O, Dzik WH et al (2006) Fresh frozen plasma and platelet transfusion for nonbleeding
patients in the intensive care unit: benefit or harm? Crit Care Med 34(5 Suppl):S170–S173
Ganter MT, Hofer CK (2008) Coagulation monitoring: current techniques and clinical use of
viscoelastic point-of-care coagulation devices. Anesth Analg 106(5):1366–1375
13 Patient Blood Management 235
Ganter MT, Spahn DR (2010) Active, personalized, and balanced coagulation management saves
lives in patients with massive bleeding. Anesthesiology 113(5):1016–1018
Glance LG, Dick AW et al (2011) Association between intraoperative blood transfusion and
mortality and morbidity in patients undergoing noncardiac surgery. Anesthesiology
114(2):283–292
Gombotz H (2011) Patient blood management is key before elective surgery. Lancet
378(9800):1362–1363
Gombotz H, Rehak PH et al (2007) Blood use in elective surgery: the Austrian benchmark study.
Transfusion 47(8):1468–1480
Gombotz H, Hofman A et al (2011a) Patient blood management (part 2). Practice: the 3 pillars.
Anasthesiol Intensivmed Notfallmed Schmerzther 46(7–8):466–474
Gombotz H, Hofmann A et al (2011b) Patient blood management (part 1) – patient-specific con-
cept to reduce and avoid anemia, blood loss and transfusion. Anasthesiol Intensivmed
Notfallmed Schmerzther 46(6):396–401
Goodnough LT (2005) Risks of blood transfusion. Anesthesiol Clin North America 23(2):
241–252, v
Goodnough LT (2007) Erythropoietin and iron-restricted erythropoiesis. Exp Hematol 35(4 Suppl
1):167–172
Goodnough LT, Shander A (2007) Blood management. Arch Pathol Lab Med 131(5):695–701
Goodnough LT, Nemeth E et al (2010) Detection, evaluation, and management of iron-restricted
erythropoiesis. Blood 116(23):4754–4761
Goodnough LT, Maniatis A et al (2011) Detection, evaluation, and management of preoperative
anaemia in the elective orthopaedic surgical patient: NATA guidelines. Br J Anaesth
106(1):13–22
Gorlinger K, Dirkmann D et al (2011) First-line therapy with coagulation factor concentrates com-
bined with point-of-care coagulation testing is associated with decreased allogeneic blood
transfusion in cardiovascular surgery: a retrospective, single-center cohort study. Anesthesiology
115(6):1179–1191
Grover K, McManus P (2006) Randomized clinical trial of the effect of quilting latissimus dorsi
flap donor site on seroma formation (Br J Surg) 2006; 93; 825–830. Br J Surg 93(12):1563;
author reply 1563–1564
Gude D (2012) TRALI: a volley of caution. Ann Card Anaesth 15(1):86–87
Hess JR, Brohi K et al (2008) The coagulopathy of trauma: a review of mechanisms. J Trauma
65(4):748–754
Hill GE, Frawley WH et al (2003) Allogeneic blood transfusion increases the risk of postoperative
bacterial infection: a meta-analysis. J Trauma 54(5):908–914
Ho AM, Karmakar MK et al (2005) Are we giving enough coagulation factors during major trauma
resuscitation? Am J Surg 190(3):479–484
Holcomb JB (2004) Methods for improved hemorrhage control. Crit Care 8(Suppl 2):S57–S60
Isbister JP, Shander A et al (2011) Adverse blood transfusion outcomes: establishing causation.
Transfus Med Rev 25(2):89–101
Jakobsen CJ, Ryhammer PK et al (2012) Transfusion of blood during cardiac surgery is associated
with higher long-term mortality in low-risk patients. Eur J Cardiothorac Surg 42:114–120
Jans O, Kehlet H et al (2012) Transfusion-related mortality after primary hip arthroplasty – an
analysis of mechanisms and confounders. Vox Sang 103:301–308
Kamper-Jorgensen M, Hjalgrim H et al (2010) Expensive blood safety initiatives may offer less
benefit than we think. Transfusion 50(1):240–242
Khan H, Belsher J et al (2007) Fresh-frozen plasma and platelet transfusions are associated with
development of acute lung injury in critically ill medical patients. Chest 131(5):1308–1314
Khanna MP, Hebert PC et al (2003) Review of the clinical practice literature on patient character-
istics associated with perioperative allogeneic red blood cell transfusion. Transfus Med Rev
17(2):110–119
Klein HG, Spahn DR et al (2007) Red blood cell transfusion in clinical practice. Lancet
370(9585):415–426
236 O.M. Theusinger et al.
Koch CG, Li L et al (2006) Morbidity and mortality risk associated with red blood cell and blood-
component transfusion in isolated coronary artery bypass grafting. Crit Care Med
34(6):1608–1616
Koch CG, Li L et al (2008) Duration of red-cell storage and complications after cardiac surgery.
N Engl J Med 358(12):1229–1239
Kotze A, Carter LA et al (2012) Effect of a patient blood management programme on preoperative
anaemia, transfusion rate, and outcome after primary hip or knee arthroplasty: a quality
improvement cycle. Br J Anaesth 108(6):943–952
Lauscher P, Mirakaj V, Rosenberger P, Meier J (2012) Practical guidelines for blood transfusions
in Germany. Anasthesiol Intensivmed Notfallmed Schmerzther 47(6):410–417
Leichtle SW, Mouawad NJ et al (2011) Does preoperative anemia adversely affect colon and rectal
surgery outcomes? J Am Coll Surg 212(2):187–194
Lier H, Krep H et al (2008) Preconditions of hemostasis in trauma: a review. The influence of
acidosis, hypocalcemia, anemia, and hypothermia on functional hemostasis in trauma. J Trauma
65(4):951–960
Liumbruno G, Bennardello F et al (2009) Recommendations for the transfusion of red blood cells.
Blood Transfus 7(1):49–64
Liumbruno GM, Bennardello F et al (2011) Recommendations for the transfusion management
of patients in the peri-operative period. III. The post-operative period. Blood Transfus
9(3):320–335
Madjdpour C, Spahn DR (2005) Allogeneic red blood cell transfusions: efficacy, risks, alternatives
and indications. Br J Anaesth 95(1):33–42
Malone DL, Dunne J et al (2003) Blood transfusion, independent of shock severity, is associated
with worse outcome in trauma. J Trauma 54(5):898–905
Martini WZ, Dubick MA et al (2006) Does bicarbonate correct coagulation function impaired by
acidosis in swine? J Trauma 61(1):99–106
Meier J, Kemming GI et al (2004) Hyperoxic ventilation reduces six-hour mortality after partial
fluid resuscitation from hemorrhagic shock. Shock 22(3):240–247
Mircescu G, Garneata L et al (2006) Intravenous iron supplementation for the treatment of anaemia
in pre-dialyzed chronic renal failure patients. Nephrol Dial Transplant 21(1):120–124
Moore FA, Moore EE et al (1997) Blood transfusion. An independent risk factor for postinjury
multiple organ failure. Arch Surg 132(6):620–624, discussion 624–625
Morales R, Duran-Aniotz C et al (2011) De novo induction of amyloid-beta deposition in vivo.
Mol Psychiatry 17:1347–1353
Morton J, Anastassopoulos KP et al (2010) Frequency and outcomes of blood products transfusion
across procedures and clinical conditions warranting inpatient care: an analysis of the 2004
healthcare cost and utilization project nationwide inpatient sample database. Am J Med Qual
25(4):289–296
Moskowitz DM, McCullough JN et al (2010) The impact of blood conservation on outcomes in
cardiac surgery: is it safe and effective? Ann Thorac Surg 90(2):451–458
Munoz M, Bisbe E et al (2005) Allogeneic blood transfusion and wound healing disturbance after
orthopaedic surgery. Anesth Analg 101(6):1889–1890, author reply 1890
Murphy GJ, Reeves BC et al (2007) Increased mortality, postoperative morbidity, and cost after red
blood cell transfusion in patients having cardiac surgery. Circulation 116(22):2544–2552
Musallam KM, Tamim HM et al (2011) Preoperative anaemia and postoperative outcomes in
non-cardiac surgery: a retrospective cohort study. Lancet 378(9800):1396–1407
Na HS, Shin SY et al (2011) Effects of intravenous iron combined with low-dose recombinant
human erythropoietin on transfusion requirements in iron-deficient patients undergoing
bilateral total knee replacement arthroplasty. Transfusion 51(1):118–124
Ness PM (2011) Does transfusion of stored red blood cells cause clinically important adverse
effects? A critical question in search of an answer and a plan. Transfusion 51(4):666–667
Nguyen XD, Dengler T et al (2011) A novel tool for high-throughput screening of granulocyte-
specific antibodies using the automated flow cytometric granulocyte immunofluorescence test
(Flow-GIFT). ScientificWorldJournal 11:302–309
13 Patient Blood Management 237
Norda R, Tynell E et al (2006) Cumulative risks of early fresh frozen plasma, cryoprecipitate and
platelet transfusion in Europe. J Trauma 60(6 Suppl):S41–S45
Nunez TC, Cotton BA (2009) Transfusion therapy in hemorrhagic shock. Curr Opin Crit Care
15(6):536–541
Offner PJ (2004) Age of blood: does it make a difference? Crit Care 8(Suppl 2):S24–S26
Patel MS, Carson JL (2009) Anemia in the preoperative patient. Med Clin North Am
93(5):1095–1104
Pati S, Matijevic N et al (2010) Protective effects of fresh frozen plasma on vascular endothelial
permeability, coagulation, and resuscitation after hemorrhagic shock are time dependent and
diminish between days 0 and 5 after thaw. J Trauma 69(Suppl 1):S55–S63
Pfanner G, Koscielny J et al (2007) Preoperative evaluation of the bleeding history.
Recommendations of the working group on perioperative coagulation of the Austrian Society
for Anaesthesia, Resuscitation and Intensive Care. Anaesthesist 56(6):604–611
Pomper GJ, Wu Y et al (2003) Risks of transfusion-transmitted infections: 2003. Curr Opin
Hematol 10(6):412–418
Popovsky MA, Chaplin HC Jr et al (1992) Transfusion-related acute lung injury: a neglected, seri-
ous complication of hemotherapy. Transfusion 32(6):589–592
Raat NJ, Berends F et al (2005) The age of stored red blood cell concentrates at the time of transfu-
sion. Transfus Med 15(5):419–423
Rachoin JS, Daher R et al (2009) Microbiology, time course and clinical characteristics of infec-
tion in critically ill patients receiving packed red blood cell transfusion. Vox Sang
97(4):294–302
Rana R, Fernandez-Perez ER et al (2006) Transfusion-related acute lung injury and pulmonary
edema in critically ill patients: a retrospective study. Transfusion 46(9):1478–1483
Reeves BC, Murphy GJ (2008a) Increased mortality, morbidity, and cost associated with red blood
cell transfusion after cardiac surgery. Curr Opin Anaesthesiol 21(5):669–673
Reeves BC, Murphy GJ (2008b) Increased mortality, morbidity, and cost associated with red blood
cell transfusion after cardiac surgery. Curr Opin Cardiol 23(6):607–612
Rogers MA, Blumberg N et al (2006) Allogeneic blood transfusions explain increased mortality in
women after coronary artery bypass graft surgery. Am Heart J 152(6):1028–1034
Rossaint R, Cerny V et al (2006) Key issues in advanced bleeding care in trauma. Shock
26(4):322–331
Rossaint R, Bouillon B et al (2010) Management of bleeding following major trauma: an updated
European guideline. Crit Care 14(2):R52
Shander A, Knight K et al (2004) Prevalence and outcomes of anemia in surgery: a systematic
review of the literature. Am J Med 116(Suppl 7A):58S–69S
Shander A, Hofmann A et al (2007) Estimating the cost of blood: past, present, and future directions.
Best Pract Res Clin Anaesthesiol 21(2):271–289
Shander A, Hofmann A et al (2010) Activity-based costs of blood transfusions in surgical patients
at four hospitals. Transfusion 50(4):753–765
Shorr AF, Duh MS et al (2004) Red blood cell transfusion and ventilator-associated pneumonia:
a potential link? Crit Care Med 32(3):666–674
Spahn DR (2010) Anemia and patient blood management in hip and knee surgery: a systematic
review of the literature. Anesthesiology 113(2):482–495
Spahn DR, Madjdpour C (2006) Physiologic transfusion triggers: do we have to use (our) brain?
Anesthesiology 104(5):905–906
Spahn DR, Rossaint R (2005) Coagulopathy and blood component transfusion in trauma. Br J
Anaesth 95(2):130–139
Spahn DR, Zollinger A et al (1996) Hemodilution tolerance in elderly patients without known
cardiac disease. Anesth Analg 82(4):681–686
Spahn DR, Cerny V et al (2007) Management of bleeding following major trauma: a European
guideline. Crit Care 11(1):R17
Spahn DR, Moch H et al (2008) Patient blood management: the pragmatic solution for the prob-
lems with blood transfusions. Anesthesiology 109(6):951–953
238 O.M. Theusinger et al.
Spahn DR, Theusinger OM et al (2012) Patient blood management is a win-win: a wake-up call.
Br J Anaesth 108(6):889–892
Spinella PC, Carroll CL et al (2009) Duration of red blood cell storage is associated with increased
incidence of deep vein thrombosis and in hospital mortality in patients with traumatic injuries.
Crit Care 13(5):R151
Stanworth SJ, Brunskill SJ et al (2004) Is fresh frozen plasma clinically effective? A systematic
review of randomized controlled trials. Br J Haematol 126(1):139–152
Surgenor SD, DeFoe GR et al (2006) Intraoperative red blood cell transfusion during coronary
artery bypass graft surgery increases the risk of postoperative low-output heart failure.
Circulation 114(1 Suppl):I43–I48
Surgenor SD, Kramer RS et al (2009) The association of perioperative red blood cell transfusions
and decreased long-term survival after cardiac surgery. Anesth Analg 108(6):1741–1746
Taylor RW, O’Brien J et al (2006) Red blood cell transfusions and nosocomial infections in critically
ill patients. Crit Care Med 34(9):2302–2308, quiz 2309
Theusinger OM, Leyvraz PF et al (2007) Treatment of iron deficiency anemia in orthopedic surgery
with intravenous iron: efficacy and limits: a prospective study. Anesthesiology 107(6):
923–927
Theusinger OM, Spahn DR et al (2009) Transfusion in trauma: why and how should we change our
current practice? Curr Opin Anaesthesiol 22(2):305–312
Theusinger OM, Nurnberg J et al (2010) Rotation thromboelastometry (ROTEM) stability and
reproducibility over time. Eur J Cardiothorac Surg 37(3):677–683
Theusinger OM, Felix C et al (2012) Strategies to reduce the use of blood products: a European
perspective. Curr Opin Anaesthesiol 25(1):59–65
Theusinger OM, Stein P et al (2014) Applying ‘Patient Blood Management’ in the trauma center.
Curr Opin Anaesthesiol 27(2):225–232
Tokuno O, Hayakawa I et al (2012) Evaluation with the BacT/ALERT microbial detection system
of bacterial contamination in autologous blood donation and transfusion. Transfus Med
22(1):73–74
Toy P, Popovsky MA et al (2005) Transfusion-related acute lung injury: definition and review. Crit
Care Med 33(4):721–726
Tung JP, Fraser JF et al (2012) Age of blood and recipient factors determine the severity of
transfusion-related acute lung injury (TRALI). Crit Care 16(1):R19
Upile T, Jerjes W et al (2009) Blood product transfusion and cancer prognosis. Clin Adv Hematol
Oncol 7(10):656–661
Urner M, Herrmann IK et al (2012) Effects of blood products on inflammatory response in endo-
thelial cells in vitro. PLoS One 7(3):e33403
Vamvakas EC, Blajchman MA (2010) Blood still kills: six strategies to further reduce allogeneic
blood transfusion-related mortality. Transfus Med Rev 24(2):77–124
Vincent JL, Baron JF et al (2002) Anemia and blood transfusion in critically ill patients. JAMA
288(12):1499–1507
Vivien B, Langeron O et al (2005) Early hypocalcemia in severe trauma. Crit Care Med
33(9):1946–1952
Vuille-Lessard E, Boudreault D et al (2012) Postoperative anemia does not impede functional
outcome and quality of life early after hip and knee arthroplasties. Transfusion 52(2):
261–270
Weber CF, Gorlinger K et al (2012) Point-of-care testing: a prospective, randomized clinical trial
of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 117(3):531–547
Weiskopf RB (2010) Emergency transfusion for acute severe anemia: a calculated risk. Anesth
Analg 111(5):1088–1092
Wells AW, Mounter PJ et al (2002) Where does blood go? Prospective observational study of red
cell transfusion in north England. BMJ 325(7368):803
Weltert L, D’Alessandro S et al (2010) Preoperative very short-term, high-dose erythropoietin
administration diminishes blood transfusion rate in off-pump coronary artery bypass: a random-
ized blind controlled study. J Thorac Cardiovasc Surg 139(3):621–626, discussion 626–627
13 Patient Blood Management 239
14.1 Introduction
F. Gronchi (*)
Department of Anesthesiology, Lausanne University Hospital (CHUV),
Lausanne CH-1011, Switzerland
e-mail: [email protected]
M. Ranucci
Department of Anesthesia and Intensive Care, IRCCS Policlinico San Donato,
20097 San Donato Milanese, Milan, Italy
e-mail: [email protected]
Due to the nature of their disease, cardiac surgery patients are usually treated with
anticoagulants and/or antiaggregant agents. The optimal timing for surgery on these
patients is still debated, and decisions should balance the risk of thrombosis and
postoperative bleeding.
A guideline to perioperative antithrombotic treatment management is summarized
in Table 14.1.
thrombin. For these reasons they usually require lower doses of UFH to reach and
maintain adequate anticoagulation during CPB.
In recent years, VKAs have been partially replaced by novel oral anticoagulants
that probably will gain even wider popularity in years to come. The most com-
monly used are the direct thrombin inhibitor dabigatran and the direct factor Xa
inhibitors apixaban and rivaroxaban. Patients treated with these drugs cannot easily
be treated with PCC, and the timing of surgery should be based on their half-life
(Table 14.1).
246 F. Gronchi and M. Ranucci
14.2.3.1 Aspirin
Antiplatelet therapy has the most important role in the prevention of acute coronary
syndromes. Aspirin treatment may increase perioperative bleeding and transfusion
requirements; however, these side effects seem limited, and discontinuation of the
treatment is not recommended. In addition, withdrawing the treatment would unrea-
sonably increase the risk of ischemic events (Class IIa, level of evidence A).
Continuing the treatment with aspirin and stopping clopidogrel prior to surgery
are currently the best compromise between hemorrhagic and thrombotic risks (Class
I, level of evidence B).
Activation of the extrinsic pathway starts at the operative site, and in spite of the
administration of heparin (aiming an ACT >400 s), the initiation of CPB induces a
surge in thrombin and fibrin generation (Chandler and Velan 2003; Hunt et al. 1998)
even though the level of hemostatic proteins in the plasma is decreased by 30 % at
248 F. Gronchi and M. Ranucci
BK FXIIa TF
Sheed
Contact Leukocyte
blood
activation activation
(open system)
Bypass circuit
that time. Hemodilution, blood loss, and volume replacement are responsible for
this decrease, whereas the role of consumption is variable and strongly dependent
on the duration of the CPB.
During cardiac operations both the extrinsic and the intrinsic coagulation path-
ways are activated, leading to thrombin generation (Fig. 14.1). However, thrombin
generation is largely due to the release of tissue factor (TF) (De Somer et al. 2002)
with the consequent activation of the extrinsic pathway. TF is expressed at the site
of surgical trauma but is also expressed by platelets due to an interaction with circu-
lating leucocytes (Chung et al. 2007). And, since inflammatory pathways, such as
the complement pathway, are intrinsically linked to the hemostatic system, the CPB
circuit’s surface can increase the release of TF by monocytes and neutrophils.
(Tabuchi et al. 2003; el Habbal et al. 1995).
After the onset of CPB, the intrinsic pathway (composed of factors XII and XI,
kininogen, and prekallikrein) is activated by interaction with the artificial surfaces
(Campbell et al. 2001). Factor XII is activated into factor XIIa, which converts
prekallikrein into kallikrein. Kallikrein then cleaves kininogen into kinin, which has
two effects: a potent hypotensive effect and a pathway amplifying effect, with a
positive feedback on bradykinin production and FXII activation. Factor XII also
cleaves inactive plasminogen into its active form plasmin, triggers the complement
pathway, and activates FXI into FXIa, initiating the intrinsic pathway.
Prekallikrein and kininogen levels decrease during CPB not only because of
hemodilution but also because of consumption and binding to the CPB circuit. On
the contrary, bradykinin levels are increased tenfold, through activation of the con-
tact system on the one hand and decreased pulmonary blood flow on the other; the
lung and the kidney are responsible for bradykinin metabolism (Campbell et al.
2001).
14 Perioperative Coagulation in Cardiovascular Surgery 249
14.3.3 Fibrinolysis
During CPB, 30 % of antithrombin III (AT III) is consumed because its natural
anticoagulant activity is markedly increased by UFH. Indeed, UFH increases the
catalytic activity of AT III by a factor of 1,000. Inactive thrombin-antithrombin III
complexes are formed and then eliminated, allowing for an effective anticoagula-
tion during CPB. If levels of AT III are too low, heparin resistance will be observed.
The clinical consequence of this will be the impossibility to reach an ACT >480 s
after UFH doses of 300–400 UI/kg. Hence, it is imperative to keep AT III levels
high enough to prevent excessive coagulation and inflammation activation, leading
to increased bleeding and transfusions (Ranucci et al. 2005a; Paparella et al. 2009).
AT III is a circulating plasma protein and can be administered through FFP; alterna-
tively, concentrated recombinant AT III is available and reduces volume load.
Although administration of recombinant AT III has proven to lower coagulation
and inflammation activation, no study has been able to demonstrate any impact on
postoperative bleeding and transfusion rates (Levy et al. 2002; Koster et al. 2003;
Avidan et al. 2005).
While thrombin production increases during CPB, protein C activity and endo-
thelial expression of its receptor decrease (Weiler 2010; Danese et al. 2010).
Hemodilution, negative protein C feedback on its own receptor, and negative
thrombomodulin feedback on the endothelium all participate in the increase of
thrombin formation.
There also is marked activation of coagulation in shed blood, where large amounts
of TF generate thrombin, activating platelets and triggering fibrinolysis (de Haan
et al. 1993). Hence, the reinfusion of saved mediastinal blood can potentially
250 F. Gronchi and M. Ranucci
The nadir hematocrit in CPB has been associated with postoperative morbidity
and mortality (Ranucci et al. 2005b; Karkouti et al. 2005; Fang et al. 1997; DeFoe
et al. 2001). Hemodilution during CPB is dependent on the priming volume, car-
dioplegia volume, and any additional fluid. Any strategy aimed at limiting infused
volumes will allow for a higher hematocrit during CPB and its postoperative
phase and, consequently, a lower transfusion rate. Many strategies are available in
order to diminish the priming volume, such as reducing the length and size of the
CPB circuits (Class I, level of evidence A), using vacuum-assisted venous drain-
age (Class IIb, level of evidence C), and applying retrograde autologous priming
(Class IIb, level of evidence B). Reducing priming volume from 1,400 to 800 ml
has been proven to significatively reduce transfusion rates. Another technique
available for minimizing hemodilution is modified ultrafiltration (MUF): after
CPB weaning, blood is ultrafiltrated through a hemofilter connected to the venous
and aortic cannulae (Class I, level of evidence A). MUF ensures not only the
removal of excess water, minimizing hemodilution, but also the removal of
inflammatory cytokines, resulting in a reduction of blood loss and transfusion
requirements.
14.4.4 Anticoagulation
14.4.5 Fibrinolysis
Bleeding patient ?
-Post CPB diffuse bleeding
-Post operative blood >250 ml/h or >50 ml/10 min
Maintain T° >36 °C, pH >7.3 Active rewarming, correct acidosis with NaHCO3, treat
Ca2+ >1.0 mmol/l, Hb >7 g/dl hypocalcemia with Ca2+, correct anemia with packed RBC
PCC 30 UI/kg or
Check coagulation factors TEG or EX TEM/INTEM
FFP 15 ml/kg
Tranexamic acid
Check for hyperfibrinolysis TEG or APTEM
15 mg/kg
Fig. 14.2 Hemostatic therapy algorithm. CPB cardiopulmonary bypass, RBC red blood cells,
TEG thromboelastography, TEG FF TEG-based functional fibrinogen assay, EXTEM tissue factor-
activated ROTEM assay, INTEM ellagic acid-activated ROTEM assay, HEPTEM heparinized
ROTEM assay, APTEM aprotinin-based ROTEM assay, PCC prothrombin complex concentrate,
FFP fresh frozen plasma
group received less rFVIIa. The conventional group, on the other hand, showed
more blood loss, a lower PaO2/FiO2 index, prolonged postoperative ventilation, an
increased rate of undesirable events, and a higher 6-month mortality rate. These
results confirm that a POC-guided approach is superior to an approach based on
standard laboratory tests. Whereas standard tests provide only quantitative information
on hemostasis, POCs rapidly provide qualitative information, allowing the clinician
to act faster and target a specific problem.
It should be noted that, even if controlling a bleeding defect is advisable, pick-
ing out the main culprit is difficult. This is due to the multitude of potential mech-
anisms causing the coagulopathy. Luckily, there is rarely a need to target a single
factor or specific clotting or fibrinolysis pathway. Indeed, acting on one part of the
clotting pathway is often sufficient to compensate for the disorder present else-
where in it.
Our pharmacological array allows us to promote hemostasis and fibrin for-
mation (and to slow down fibrinolysis) by interfering in the delicate balance
between coagulation activation and physiological anticoagulation (Mannucci
and Levi 2007). Nevertheless, the use of pharmacological agents carries the
potential risk of thrombotic complications. A transfusion algorithm is shown in
Fig. 14.2.
256 F. Gronchi and M. Ranucci
The use of fibrinogen, factor XIII, PCC, desmopressin, recombinant factor VII,
and antifibrinolytics is discussed in Chaps. 11 and 12.
Unfractionated heparin remains the gold standard for anticoagulation in cardiac sur-
gery, with or without CPB, given its easy titration, safety margin, reversibility, and
low cost. However, its use carries the risk of developing heparin-induced thrombo-
cytopenia (HIT). Heparin binds to PF4 and 50 % of cardiac surgery patients develop
antibodies (Ab) to these heparin-PF4 complexes (anti-heparin-PF4 Ab). In 1–5 % of
these patients, the anti-heparin-PF4 Ab will activate platelets by binding its Fc frag-
ment to the platelet FcRII receptor (Warkentin et al. 2000).
The anti-heparin-PF4 Ab-mediated platelet activation and the resulting release
of proclotting factors increase the production of thrombin. The continuing expo-
sure, or reexposure, to heparin can lead to venous thrombosis (17–55 % of cases)
(Warkentin and Kelton 1996; Warkentin et al. 2000) or arterial thrombosis (3–10 %
of cases) (Warkentin and Kelton 1996). Although rare, it can also cause anaphylac-
toid reactions after intravenous injection, cutaneous necrosis at the site of injection,
adrenal hemorrhage, or disseminated intravascular coagulation (DIC).
In cardiac surgery, the most common complication of untreated or unrecognized
HIT is arterial thrombosis, which carries an associated mortality of 5–10 %.
The diagnosis of HIT is based on a typical clinical presentation and the presence of
anti-PF4 Ab in patients treated with UFH or LMWH (although it is ten times less
frequent with the latter). HIT typically appears between the fifth and tenth day of
heparin treatment or within 24 h if anti-heparin-PF4 Ab are still circulating after
prior sensitization (<100 days). Less frequently, HIT can occur up to 3 weeks after
the cessation of heparin. In cardiac surgery, the pattern of HIT differs from the
thrombocytopenia generally seen after CPB: a patient who undergoes CPB gener-
ally experiences a platelet drop immediately on arrival in the intensive care unit;
subsequently, the platelet count starts recovering, reaching preoperative values
258 F. Gronchi and M. Ranucci
around 5–6 days after the operation (Pouplard et al. 2005). In this setting, HIT is
more often represented by a continuous drop in platelet count or a very late recovery
(Pouplard et al. 2005).
An easy tool for the clinical diagnosis of HIT is the 4Ts score (Table 14.2).
A low 4Ts score implies a very low probability of the patient actually having a HIT
(0–0.3 %) (Lo et al. 2006; Pouplard et al. 2007); however, some patients with a high
4Ts score (24–61 %) do not have HIT at all (Lo et al. 2006; Pouplard et al. 2007).
In cardiac surgery, the usefulness of the 4Ts score is limited because 2 of the 4 Ts
are always present after the procedure anyway (Thrombocytopenia and other
causes) and a third (Time) is unreliable.
HIT is associated with a daily thrombosis rate of 5 % (Lubenow et al. 2005). Thus,
considering the long laboratory turnaround times for HIT tests and the nondiagnostic
presence of isolated anti-PF4 Ab, a fast clinical diagnosis of HIT is imperative.
After cardiac surgery, patients with strongly suspected or confirmed HIT, whether
or not complicated by thrombosis, should be treated with an alternative, nonheparin
anticoagulant such as danaparoid, lepirudin, argatroban, fondaparinux, or bivaliru-
din (Linkins et al. 2012). Any treatment with VKA must be interrupted until the
platelet count has substantially recovered (usually, to at least 150 × 109/l). VKA
Table 14.2 Estimating the pretest probability of HIT with the 4Ts score: low probability, 0–3
points; moderate probability, 4–5 points; and high probability, 6–8 points
Score = 2 Score = 1 Score = 0
Thrombocytopenia >50 % fall 30–50 % fall <30 % fall
or or or
>20 × 109/l 10–20 × 109/l <10 × 109/l
Timing of platelet Platelet fall days 5–10 Platelet fall > day 10 Platelet fall ≤ day
count fall or after start of heparin 4 without
thrombosis (day Platelet fall within Platelet fall within 1 day and exposure to
0 = first day of 1 day and exposure to exposure to heparin within heparin in past
most recent heparin within past past 31–100 days 100 days
heparin exposure) 5–30 days Platelet fall days 5–10 but not
clear (e.g., missing counts)
Thrombosis (or Confirmed new Recurrent venous thrombosis Thrombosis
other clinical thrombosis in patient receiving suspected
sequelae) therapeutic anticoagulants
Skin necrosis at Suspected thrombosis
injection site (awaiting confirmation)
Anaphylactoid reaction Erythematous skin lesions at
to IV heparin bolus heparin injection sites
Adrenal hemorrhage
OTher causes of No alternative Possible other cause is Probable other
Thrombocytopenia explanation for platelet evident cause is evident
fall is evident
14 Perioperative Coagulation in Cardiovascular Surgery 259
therapy can only be resumed with low maintenance doses (maximum 5 mg of war-
farin or 6 mg of phenprocoumon), and the nonheparin anticoagulant has to be con-
tinued until the platelet count has reached a stable plateau and the international
normalized ratio (INR) has reached the intended target range. There must be a mini-
mum overlap of 5 days between nonheparin anticoagulation and VKA therapy
before the nonheparin anticoagulant is withdrawn (Linkins et al. 2012).
Patients with a history, or a severe suspicion, of previous HIT should be tested for
anti-heparin-PF4 Ab. These usually disappear about 100 days after the last exposure
to heparin. If anti-heparin-PF4 Ab are no longer present, cardiac surgery can be per-
formed using standard UFH (Warkentin et al. 2008), but after receiving protamine,
no further doses of heparin should be administered, and the prophylaxis of throm-
botic events should be based on alternative anticoagulants. Conversely, if active anti-
bodies are still present, the operation should be postponed (if feasible) until they
disappear. An algorithm for the management of patients with HIT who need cardiac
surgery is presented in Fig. 14.3.
Different strategies have been proposed for patients with active antibodies whose
cardiac surgery cannot be postponed. A first possibility is to replace heparin with
another anticoagulant, such as argatroban (Edwards et al. 2003; Furukawa et al. 2001),
lepirudin (Koster et al. 2000a; Riess et al. 2007), or bivalirudin (Koster et al. 2000a,
2007; Dyke et al. 2007). However, caution should be applied in patients with impaired
renal function, especially when bivalirudin or, in particular, lepirudin is used.
In patients with acute HIT, there is no direct evidence supporting the use of one
alternative nonheparin anticoagulant over another. Although off-label, bivalirudin is
Table 14.3 Alternative anticoagulation with bivalirudin for urgent cardiac surgery in patients
diagnosed with HIT
Drug Start Bolus Infusion Monitoring Additional doses Stop
Bivalirudin Before 1 mg/kg 2.5 mg/kg/min ACT >400 s During CPB: 10–15
(Koster cannulation IV and or 2.5 times 0.1–0.5 mg/kg IV min
et al. 2007) 50 mg of baseline After weaning: before
in CPB value 50 mg in CPB weaning
prime followed by
50 mg/h infusion
For details, see text
the only one that is supported by prospective multicenter cohort studies of patients
with HIT, who require urgent cardiac surgery, and indirectly by small randomized
heparin-controlled trials in patients without HIT (Dyke et al. 2007; Koster et al.
2007, 2009). This hirudin-derived peptide does not cross-react with anti-PF4 Ab.
Bivalirudin is a reversible thrombin inhibitor with a half-life of 25 min; it is elimi-
nated by plasmatic proteolysis and renal excretion (20 %) and can be ultrafiltrated.
Bivalirudin is administered with an initial bolus of 1 mg/kg IV, followed by an
infusion of 2.5 mg/kg/h (Koster et al. 2007) (Table 14.3). Targeting an ACT >300 s
(or 2.5 times the baseline), additional 0.1–0.5 mg/kg boluses can be given. The infu-
sion should be stopped 10–15 min before weaning. In the case of CPB, 50 mg has
to be added to the priming volume, and due to bivalirudin’s metabolic properties, its
use demands certain changes. First, surgery has to be normothermic. Second, since
stasis of blood can enhance the enzymatic breakdown of bivalirudin, the following
modifications of the CBP circuit are recommended:
• The use of a closed-circuit CPB when possible or replacing cardiotomy suction
by a citrate anticoagulated cell saver.
• Avoiding hemofiltration during CPB.
• Inserting shunt lines from arterial filter to the cardiotomy reservoir.
• Intermittent flushing of soft venous reservoirs.
• After weaning from CPB, add a bolus of 50 mg followed by a continued infusion
of 50 mg/h in the circuit.
• After weaning, once return on bypass is excluded, process the blood in the circuit
with a citrate anticoagulated cell saver for reinfusion.
In the case of CABG surgery, assessments of graft patency or leakage must be
performed with unheparinized normal saline. When grafting an internal mammary
artery, the vessel should be transected as late as possible before grafting.
Another strategy involves combining heparin with a short-acting potent anti-
platelet agent, such as a prostacyclin analog (e.g., epoprostenol, iloprost) (Antoniou
et al. 2002) or a glycoprotein (GP) IIb/IIIa inhibitor (e.g., tirofiban) (Koster et al.
2000b) to attenuate platelet activation (Table 14.4). Prostacyclin analogs inhibit
platelet activation by increasing adenylate cyclase activity and have very short half-
lives (6 min for epoprostenol and 15–30 min for iloprost). The most important side
effect reported is profound hypotension. Tirofiban has a half-life of about 2 h and is
eliminated by renal and biliary excretion.
Table 14.4 Anticoagulation with platelet inhibitors and heparin for urgent cardiac surgery in patient diagnosed with HIT
Drug Start Bolus Infusion Heparin Monitoring Additional doses Stop
Prostacyclin After induction Avoid 6–12 ng/kg/min 100–300 UI/kg when ACT Increase perfusion by 6 ng/kg/min 20 min after
(Antoniou et al. of anesthesia HIPA negative HIPA test until HIPA negative protamine
2002)
Tirofiban (Koster Before 10 mcg/kg 0.15 mcg/min 300 UI/kg after ACT None 1 h before end
et al. 2001) cannulation tirofiban bolus >480 s of CPB
14 Perioperative Coagulation in Cardiovascular Surgery
References
Alghamdi AA, Davis A et al (2006) Development and validation of Transfusion Risk Understanding
Scoring Tool (TRUST) to stratify cardiac surgery patients according to their blood transfusion
needs. Transfusion 46(7):1120–1129
Antoniou T, Kapetanakis EI et al (2002) Cardiac surgery in patients with heparin-induced throm-
bocytopenia using preoperatively determined dosages of iloprost. Heart Surg Forum
5(4):354–357
Arthurs Z, Cuadrado D et al (2006) The impact of hypothermia on trauma care at the 31st combat
support hospital. Am J Surg 191(5):610–614
Avidan MS, Levy JH et al (2005) Recombinant human antithrombin III restores heparin responsive-
ness and decreases activation of coagulation in heparin-resistant patients during cardiopulmonary
bypass. J Thorac Cardiovasc Surg 130(1):107–113
Berger JS, Frye CB et al (2008) Impact of clopidogrel in patients with acute coronary syndromes
requiring coronary artery bypass surgery: a multicenter analysis. J Am Coll Cardiol
52(21):1693–1701
Butler K, Teng R (2010) Pharmacokinetics, pharmacodynamics, safety and tolerability of multiple
ascending doses of ticagrelor in healthy volunteers. Br J Clin Pharmacol 70(1):65–77
Cammerer U, Dietrich W et al (2003) The predictive value of modified computerized thromboelas-
tography and platelet function analysis for postoperative blood loss in routine cardiac surgery.
Anesth Analg 96(1):51–57, table of contents
Campbell FW, Goldstein MF et al (1984) Management of the patient with protamine hypersensi-
tivity for cardiac surgery. Anesthesiology 61(6):761–764
Campbell DJ, Dixon B et al (2001) Activation of the kallikrein-kinin system by cardiopulmonary
bypass in humans. Am J Physiol Regul Integr Comp Physiol 281(4):R1059–R1070
Chandler WL, Velan T (2003) Estimating the rate of thrombin and fibrin generation in vivo during
cardiopulmonary bypass. Blood 101(11):4355–4362
Chandler WL, Velan T (2004) Plasmin generation and D-dimer formation during cardiopulmonary
bypass. Blood Coagul Fibrinolysis 15(7):583–591
Chandler WL, Alessi MC et al (2000) Formation, inhibition and clearance of plasmin in vivo.
Haemostasis 30(4):204–218
Chung J, Suzuki H et al (2007) Identification of tissue factor and platelet-derived particles on
leukocytes during cardiopulmonary bypass by flow cytometry and immunoelectron micros-
copy. Thromb Haemost 98(2):368–374
Danese S, Vetrano S et al (2010) The protein C pathway in tissue inflammation and injury: patho-
genic role and therapeutic implications. Blood 115(6):1121–1130
Davidson SJ, McGrowder D et al (2008) Can ROTEM thromboelastometry predict postoperative
bleeding after cardiac surgery? J Cardiothorac Vasc Anesth 22(5):655–661
de Haan J, van Oeveren W (1998) Platelets and soluble fibrin promote plasminogen activation
causing downregulation of platelet glycoprotein Ib/IX complexes: protection by aprotinin.
Thromb Res 92(4):171–179
de Haan J, Schonberger J et al (1993) Tissue-type plasminogen activator and fibrin monomers
synergistically cause platelet dysfunction during retransfusion of shed blood after cardiopul-
monary bypass. J Thorac Cardiovasc Surg 106(6):1017–1023
De Somer F, Van Belleghem Y et al (2002) Tissue factor as the main activator of the coagulation
system during cardiopulmonary bypass. J Thorac Cardiovasc Surg 123(5):951–958
DeFoe GR, Ross CS et al (2001) Lowest hematocrit on bypass and adverse outcomes associated
with coronary artery bypass grafting. Northern New England Cardiovascular Disease Study
Group. Ann Thorac Surg 71(3):769–776
Dehmer GJ, Fisher M et al (1995) Reversal of heparin anticoagulation by recombinant platelet
factor 4 in humans. Circulation 91(8):2188–2194
Despotis GJ, Joist JH et al (1995) The impact of heparin concentration and activated clotting time
monitoring on blood conservation. A prospective, randomized evaluation in patients undergoing
cardiac operation. J Thorac Cardiovasc Surg 110(1):46–54
14 Perioperative Coagulation in Cardiovascular Surgery 263
Diago MC, Garcia-Unzueta MT et al (1997) Serum soluble selectins in patients undergoing cardio-
pulmonary bypass. Relationship with circulating blood cells and inflammation-related cytokines.
Acta Anaesthesiol Scand 41(6):725–730
Dyke CM, Aldea G et al (2007) Off-pump coronary artery bypass with bivalirudin for patients with
heparin-induced thrombocytopenia or antiplatelet factor four/heparin antibodies. Ann Thorac
Surg 84(3):836–839
Edwards JT, Hamby JK et al (2003) Successful use of Argatroban as a heparin substitute during
cardiopulmonary bypass: heparin-induced thrombocytopenia in a high-risk cardiac surgical
patient. Ann Thorac Surg 75(5):1622–1624
el Habbal MH, Carter H et al (1995) Neutrophil activation in paediatric extracorporeal circuits:
effect of circulation and temperature variation. Cardiovasc Res 29(1):102–107
Engstrom M, Schott U et al (2006) Acidosis impairs the coagulation: a thromboelastographic
study. J Trauma 61(3):624–628
Fang WC, Helm RE et al (1997) Impact of minimum hematocrit during cardiopulmonary bypass on
mortality in patients undergoing coronary artery surgery. Circulation 96(9 Suppl):II-194–II-199
Finley A, Greenberg C (2013) Review article: heparin sensitivity and resistance: management
during cardiopulmonary bypass. Anesth Analg 116(6):1210–1222
Fitchett D, Eikelboom J et al (2009) Dual antiplatelet therapy in patients requiring urgent coronary
artery bypass grafting surgery: a position statement of the Canadian Cardiovascular Society.
Can J Cardiol 25(12):683–689
Franz A, Braunlich P et al (2001) The effects of hydroxyethyl starches of varying molecular
weights on platelet function. Anesth Analg 92(6):1402–1407
Freyburger G, Janvier G et al (1993) Fibrinolytic and hemorheologic alterations during and after
elective aortic graft surgery: implications for postoperative management. Anesth Analg
76(3):504–512
Frick PG, Brogli H (1966) The mechanism of heparin rebound after extracorporeal circulation for
open cardiac surgery. Surgery 59(5):721–726
Furukawa K, Ohteki H et al (2001) The use of argatroban as an anticoagulant for cardiopulmonary
bypass in cardiac operations. J Thorac Cardiovasc Surg 122(6):1255–1256
Gallandat Huet RC, Siemons AW et al (2000) A novel hydroxyethyl starch (Voluven) for effective
perioperative plasma volume substitution in cardiac surgery. Can J Anaesth 47(12):
1207–1215
Hajjar LA, Vincent JL et al (2010) Transfusion requirements after cardiac surgery: the TRACS
randomized controlled trial. JAMA 304(14):1559–1567
Hongo RH, Ley J et al (2002) The effect of clopidogrel in combination with aspirin when given
before coronary artery bypass grafting. J Am Coll Cardiol 40(2):231–237
Hunt BJ, Parratt RN et al (1998) Activation of coagulation and fibrinolysis during cardiothoracic
operations. Ann Thorac Surg 65(3):712–718
Jobes DR, Aitken GL et al (1995) Increased accuracy and precision of heparin and protamine
dosing reduces blood loss and transfusion in patients undergoing primary cardiac operations.
J Thorac Cardiovasc Surg 110(1):36–45
Karkouti K, Beattie WS et al (2005) Hemodilution during cardiopulmonary bypass is an indepen-
dent risk factor for acute renal failure in adult cardiac surgery. J Thorac Cardiovasc Surg
129(2):391–400
Karkouti K, O’Farrell R et al (2006) Prediction of massive blood transfusion in cardiac surgery.
Can J Anaesth 53(8):781–794
Koch CG, Li L et al (2006) Transfusion in coronary artery bypass grafting is associated with
reduced long-term survival. Ann Thorac Surg 81(5):1650–1657
Koster A, Hansen R et al (2000a) Recombinant hirudin as an alternative for anticoagulation during
cardiopulmonary bypass in patients with heparin-induced thrombocytopenia type II: a 1-year
experience in 57 patients. J Cardiothorac Vasc Anesth 14(3):243–248
Koster A, Loebe M et al (2000b) Cardiopulmonary bypass in a patient with heparin-induced
thrombocytopenia II and impaired renal function using heparin and the platelet GP IIb/IIIa
inhibitor tirofiban as anticoagulant. Ann Thorac Surg 70(6):2160–2161
264 F. Gronchi and M. Ranucci
Pouplard C, Gueret P et al (2007) Prospective evaluation of the ‘4Ts’ score and particle gel
immunoassay specific to heparin/PF4 for the diagnosis of heparin-induced thrombocytopenia.
J Thromb Haemost 5(7):1373–1379
Purkayastha S, Athanasiou T et al (2006) Does clopidogrel affect outcome after coronary artery
bypass grafting? A meta-analysis. Heart 92(4):531–532
Quinton TM, Kim S et al (2004) Plasmin-mediated activation of platelets occurs by cleavage of
protease-activated receptor 4. J Biol Chem 279(18):18434–18439
Ranucci M, Castelvecchio S (2009) Management of mini-cardiopulmonary bypass devices: is it
worth the energy? Curr Opin Anaesthesiol 22(1):78–83
Ranucci M, Frigiola A et al (2005a) Postoperative antithrombin levels and outcome in cardiac
operations. Crit Care Med 33(2):355–360
Ranucci M, Romitti F et al (2005b) Oxygen delivery during cardiopulmonary bypass and acute
renal failure after coronary operations. Ann Thorac Surg 80(6):2213–2220
Ranucci M, Balduini A et al (2009a) A systematic review of biocompatible cardiopulmonary
bypass circuits and clinical outcome. Ann Thorac Surg 87(4):1311–1319
Ranucci M, Castelvecchio S et al (2009b) Predicting transfusions in cardiac surgery: the easier, the
better: the Transfusion Risk and Clinical Knowledge score. Vox Sang 96(4):324–332
Ranucci M, Baryshnikova E et al (2011) Multiple electrode whole-blood aggregometry and bleeding
in cardiac surgery patients receiving thienopyridines. Ann Thorac Surg 91(1):123–129
Riess FC, Poetzsch B et al (2007) Recombinant hirudin for cardiopulmonary bypass anticoagula-
tion: a randomized, prospective, and heparin-controlled pilot study. Thorac Cardiovasc Surg
55(4):233–238
Rivard GE, Brummel-Ziedins KE et al (2005) Evaluation of the profile of thrombin generation
during the process of whole blood clotting as assessed by thromboelastography. J Thromb
Haemost 3(9):2039–2043
Shore-Lesserson L, Manspeizer HE et al (1999) Thromboelastography-guided transfusion algo-
rithm reduces transfusions in complex cardiac surgery. Anesth Analg 88(2):312–319
Shuhaibar MN, Hargrove M et al (2004) How much heparin do we really need to go on pump?
A rethink of current practices. Eur J Cardiothorac Surg 26(5):947–950
Slaughter TF, Sreeram G et al (2001) Reversible shear-mediated platelet dysfunction during car-
diac surgery as assessed by the PFA-100 platelet function analyzer. Blood Coagul Fibrinolysis
12(2):85–93
Sniecinski RM, Chandler WL (2011) Activation of the hemostatic system during cardiopulmonary
bypass. Anesth Analg 113(6):1319–1333
Snyder-Ramos SA, Mohnle P et al (2008) The ongoing variability in blood transfusion practices in
cardiac surgery. Transfusion 48(7):1284–1299
Society of Thoracic Surgeons Blood Conservation Guideline Task Force, Ferraris VA et al (2007)
Perioperative blood transfusion and blood conservation in cardiac surgery: the Society of
Thoracic Surgeons and The Society of Cardiovascular Anesthesiologists clinical practice
guideline. Ann Thorac Surg 83(5 Suppl):S27–S86
Society of Thoracic Surgeons Blood Conservation Guideline Task Force, Ferraris VA et al (2011) 2011
update to the Society of Thoracic Surgeons and the Society of Cardiovascular Anesthesiologists
blood conservation clinical practice guidelines. Ann Thorac Surg 91(3):944–982
Spahn DR, Dettori N et al (2004) Transfusion in the cardiac patient. Crit Care Clin
20(2):269–279
Stover EP, Siegel LC et al (1998) Variability in transfusion practice for coronary artery bypass
surgery persists despite national consensus guidelines: a 24-institution study. Institutions of the
Multicenter Study of Perioperative Ischemia Research Group. Anesthesiology 88(2):327–333
Suttner S, Piper SN et al (2004) The influence of allogeneic red blood cell transfusion compared
with 100 % oxygen ventilation on systemic oxygen transport and skeletal muscle oxygen tension
after cardiac surgery. Anesth Analg 99(1):2–11
Szalados JE, Ouriel K, Shapiro JR (1994) Use of the activated coagulation time and heparin dose-
response curve for the determination of protamine dosage in vascular surgery. J Cardiothorac
Vasc Anesth. 8(5):515–518
266 F. Gronchi and M. Ranucci
K. Görlinger (*)
Department of Anesthesiology and Intensive Care Medicine,
University Hospital Essen, University Duisburg-Essen, Essen, Germany
e-mail: [email protected], [email protected]
E. Schaden
Department of Anesthesiology, General Intensive Care and Pain Control,
Medical University of Vienna, Vienna, Austria
F.H. Saner
Department of General, Visceral and Transplant Surgery,
University Hospital Essen, University Duisburg-Essen, Essen, Germany
in the presence of elevated levels of large vWF multimers, increases platelet micro-
thrombi formation and can therefore result in sinusoidal microcirculatory distur-
bances and subsequent progression of liver injury. This can eventually lead to
multiple organ failure (Lisman et al. 2006; Pereboom et al. 2009a; Uemura et al.
2011). A marked imbalance between decreased ADAMTS13 activity and increased
production of large vWF multimers has been shown to be closely related to func-
tional liver capacity, hepatic encephalopathy, hepatorenal syndrome, and intractable
ascites in advanced liver cirrhosis; it may also be useful in predicting long-term
survival of cirrhotic patients (Uemura et al. 2011; Takaya et al. 2012). Therefore,
some end-stage liver cirrhotic patients show conditions similar to thrombotic throm-
bocytopenic purpura (TTP). Besides sequestration of platelets in the spleen due to
portal hypertension and subsequent hypersplenism (Al-Busafi et al. 2012; Bhavsar
et al. 2012; Kedia et al. 2012), this mechanism may substantially contribute to
thrombocytopenia in liver cirrhotic patients. This thrombocytopenia seems to rebal-
ance the increased platelet adhesion and aggregation resulting from increased levels
of large vWF multimers in plasma and decreased ADAMTS13 activity. Therefore,
platelet transfusion should be restricted to bleeding complications since it may
result in further liver damage and exacerbated portal and portopulmonary hyperten-
sion (Elias et al. 2013). Notably, platelet dysfunction and acquired dysfibrinogen-
emia may also occur in liver cirrhosis (Caldwell and Sanyal 2009; Math et al. 2010;
Tripodi and Mannucci 2011). Furthermore, changes in pro- and antifibrinolytic
drivers have been reported. Here, plasminogen and alpha2-antiplasmin levels
decrease while tissue-plasminogen activator and plasminogen activator inhibitor-1
levels simultaneously increase (Schaden et al. 2013; Tripodi and Mannucci 2011).
Endotoxemia and subsequent tissue factor expression on monocytes are common in
patients with liver cirrhosis or following liver transplantation (Esch et al. 2010).
Therefore, infections can quickly result in alterations in hemostasis in cirrhotic
patients by inducing disseminated intravascular coagulation (DIC) (Smalberg and
Leebeek 2009; Chavez-Tapia et al. 2011b). Similar, but more pronounced changes
of pro- and anticoagulant factors are observed in acute liver injury/failure (Agarwal
et al. 2012), but data regarding fibrinolysis in acute liver dysfunction are inconclu-
sive (Agarwal et al. 2012; Lisman et al. 2012a, b). Recently published data showed
evidence of reduced fibrinolytic activity in acute liver failure, similar to that shown
in early phase sepsis (Adamzik et al. 2010; Brenner et al. 2012; Lisman et al. 2012a, b).
Taken together, blood coagulation in chronic liver dysfunction is rebalanced, even
though at an earlier stage it is prone to tipping toward thrombosis or hemorrhage,
depending on concomitant risk factors (Fig. 15.1) (Schaden et al. 2013; Tripodi and
Mannucci 2011). Recent studies have nevertheless shown that patients with liver
cirrhosis are at greater risk of thrombosis than bleeding, even if routine plasmatic
coagulation tests suggest hypocoagulability (Lisman et al. 2010; Ditisheim et al.
2012; Tripodi and Mannucci 2011; Tripodi et al. 2011). Therefore, prophylactic
correction of laboratory values by transfusion of blood products may have a delete-
rious effect on liver cirrhotic patients (Ditisheim et al. 2012; Schaden et al. 2013;
Tripodi and Mannucci 2011).
15 Perioperative Hemostasis in Hepatic Surgery 269
The prothrombin time (PT) test, first described in 1935, was developed and imple-
mented to monitor anticoagulation with vitamin K antagonists (VKA) (Owren and
Aas 1951). Thromboplastins of different origin are added to recalcified citrated
plasma, and the time until coagulation starts is measured. This test only reflects the
activity of vitamin K-dependent procoagulant factors in plasma; it is neither capable
of measuring the activity of the vitamin K-dependent anticoagulants proteins C and
S, nor the complex interaction of cells and coagulation factors in whole blood
(Schaden et al. 2013; Tripodi and Mannucci 2011). Due to the use of different throm-
boplastins, results from different laboratories are not comparable. The INR was
established – and is indeed useful – to monitor anticoagulation in patients on VKA.
Later, the INR was used to detect and quantify coagulopathy in many other clinical
settings without ever having been validated for them, e.g., to predict bleeding in elec-
tive surgery, to guide hemostatic therapy in massive bleeding after trauma or surgery,
and also to define coagulopathy in liver disease. Meanwhile, it has been shown that
the correlation between INR and bleeding in patients scheduled for surgery is poor
(Koscielny et al. 2004). This has been demonstrated in patients with liver cirrhosis as
well (Stravitz et al. 2012; Tripodi and Mannucci 2011). In particular, no correlation
could be observed between PT and the bleeding time observed directly on the liver
surface during laparoscopic liver biopsy (Ewe 1981). However, the validity of the
INR as a prognostic parameter in liver dysfunction is not affected by this finding
(Stravitz et al. 2012).
270 K. Görlinger et al.
surgery and in intensive care units (Haas et al. 2012a, b). Furthermore, the diagnos-
tic performance of a panel of specific reagents and additives used in thromboelas-
tometry has been shown to be superior to mono-analysis using kaolin-based tests
(Larsen et al. 2011). On the one hand, algorithms based on the use of kaolin-acti-
vated tests alone, usually lead to platelet transfusion in cases of reduced clot firm-
ness (Larsen et al. 2011; Sakai et al. 2012). On the other hand, algorithms based on
a panel of ROTEM® reagents may avoid platelet transfusion when goal-directed
fibrinogen substitution is more appropriate (Figs. 15.2 and 15.3a–h) (Larsen et al.
2011; Görlinger et al. 2010, 2011a, b). This is of special importance in liver trans-
plantation since platelet transfusion is associated with a significant reduction in
1-year survival (74 % vs. 92 %; P < 0.001) in this clinical setting (Pereboom et al.
2009b). Notably, viscoelastic tests showed normo- (Stravitz et al. 2012) or even
hypercoagulability (Agarwal et al. 2012) in patients with acute liver failure, further
challenging the bleeding tendency concept in liver dysfunction. Here, hypercoagu-
lability seems to be better detected by whole blood thromboelastometry than by
TG tests using platelet-poor plasma (Fig. 15.3a) (Tripodi et al. 2009a). Furthermore,
tissue factor expression on monocytes, detected by thromboelastometry in septic
patients as well as in patients undergoing liver transplantation or extracorporeal
organ support, may play an important role in hypercoagulability and thrombosis in
liver cirrhotic patients (Fig. 15.3b) (Adamzik et al. 2010; Esch et al. 2010; Görlinger
et al. 2012a).
CAVE:
Patients with chronic inflammatory
LTX diseases of the bile system (i.e.
PBC) or malignant tumors have a
high risk of thrombotic events or
vascular occlusion!
YES Thrombotic
Fulminant
events in patients NO
liver failure history
? ?
YES
NO
A10EX≤ 25 mm
YES Prophylactic administration
or CTEX> 80 s
of tranexamic acid
at beginning of (25 mg/kg bw)
surgery
NO
NO
NO NO
NO Aggravation of YES
fibrinolysis in close-
meshed controls
?
Check and optimise
No NO Diffuse YES basic conditions:
therapy clinical bleeding
! ? Tc> 35 °C pH > 7,2
Cai> 1 mmol/l Hb > 8 g/dl
NO
Administration of Consider AT substitution,
CTEX > 80 s YES prothrombin complex if AT << Quick (PT in %)
(Check: PT !) concentrate (PCC) or in patients with high risk
(25(-40) IU/kg bw) of thrombotic events
NO Administration of protamine,
only if heparin effect
YES HEPTEM-Test: YES is undesirable
CTIN > 240 s CTHEP << CTIN
? NO
Transfusion of FFP
NO (6-12 U)
Ongoing
NO
diffuse bleeding
?
YES
MULTIPLATE: Transfusion of
YES platelet concentrates
platelet dysfunction
? (1-2 U pooled or apharesis)
NO 10-15 min after each
pH > 7,2 specific intervention
and a reexamination of
Back to CTEX < 80 s
ROTEM NO
and
YES Consider ROTEM® - analysis
administration of
analysis A10FIB > 15 mm F XIII or rFVIIa should be done as a
(control) and control of sucess
A10EX > 45 mm
15 Perioperative Hemostasis in Hepatic Surgery 273
immunomodulation, increased nosocomial infection rates, and, last but not least,
therapy delay due to the thawing process all have to be considered when using
FFP (Pandit and Sarode 2012). Data from patients with liver cirrhosis are not
available yet (Levy et al. 2012; Sørensen et al. 2011). Results obtained in cardiac
surgery and liver transplantation, however, prove that when guided by POC coag-
ulation monitoring with thromboelastometry, the administration of specific coag-
ulation factor concentrates, like fibrinogen and 4-factor prothrombin complex
concentrates, corrects coagulopathy without increasing the thrombotic risk
(Fig. 15.2) (Görlinger et al. 2010; Görlinger et al. 2011a, 2012b; Kirchner et al.
2012; Weber et al. 2012). Furthermore, several other authors reported on the
advantages of coagulation management guided by thromboelastometry in liver
transplantation (Blasi et al. 2012; Minov et al. 2012; Noval-Padillo et al. 2010;
Roullet et al. 2010; Stancheva et al. 2011; Tripodi et al. 2009b; Trzebicki et al.
2010). Platelet transfusion can also be guided by POC monitoring (Figs. 15.2 and
15.3c–d) (Larsen et al. 2011; Görlinger et al. 2010, 2011a, b, 2012a, b). However,
platelet transfusion has been shown to be associated with a significant reduction
in 1-year survival (74 % vs. 92 %; P < 0.001) in liver transplantation, independent
of whether the platelet count was below or above 50/nL before platelet transfusion
(Pereboom et al. 2009b). Therefore, the indication to transfuse platelets should be
considered carefully. Cryoprecipitate, containing fibrinogen and factor XIII, but
also vWF and factor VIII, would further increase the already high levels of the
latter two, possibly contributing to a procoagulant switch with subsequent throm-
bosis (see below) (Dasher and Trotter 2012). Notably, the factor XIII Val34Leu
mutation, either alone or in combination with the PAI-1 4G/5G mutation, has been
shown to be a risk factor for an increased rate of liver fibrosis development in
patients with chronic hepatitis B or C (Dik et al. 2012).
a b
0’ 1 EXTEM 0’ 1 EXTEM
St. : 08h55 St. : 11h42
CT : 67s CT : 33s
0’ 3 FIBTEM 0’ 3 FIBTEM
St. : 08h56 St. : 08h56
Fig. 15.3 Interpretation of ROTEM® analyses in patients undergoing liver transplantation. (a)
Hypercoagulability in an infant with Budd-Chiari syndrome (hepatic vein thrombosis). Platelet
count 796/nL; plasma fibrinogen concentration >10 g/L; d-dimer 228 μg/dL; Quick 68 %; aPTT
33.8 s; AT 111 %. (b) Hypercoagulability due to infection and tissue factor expression on mono-
cytes. Quick 18 %; INR 3.5; aPTT 62.8; plasma fibrinogen concentration 6.56 g/L. The mismatch
between an increased INR (PT) in routine plasmatic coagulation tests and a reduced CT in whole
blood viscoelastic tests (ROTEM®) is typical for tissue factor expression on circulation cells (mono-
cytes or malignant cells). (c) Fibrinogen deficiency. Administration of fibrinogen concentrate (cryo-
precipitate) is indicated according to the POC algorithm in Fig. 15.2 in case of bleeding and A10EX
≤35 mm and A10FIB ≤8 mm (corresponding to MCFEX ≤45 mm and MCFFIB ≤10 mm). Fibrinogen
dosage (mg) = targeted increase in A10FIB (mm) × 6.25 mg/kg fibrinogen × kg bw. (d)
Thrombocytopenia compensated by a high plasma fibrinogen level. Platelet count 22/nL; plasma
fibrinogen concentration 4.2 g/L. Transfusion of platelet concentrates is indicated according to the
POC algorithm in Fig. 15.2 in case of bleeding and A10EX ≤35 mm and A10FIB >8 mm (correspond-
ing to MCFEX ≤45 mm and MCFFIB >10 mm). (e) Fulminant fibrinolysis in the anhepatic phase of
liver transplantation. Clot firmness in EXTEM® is reduced to zero within 15 min; flat line in
FIBTEM®. Recommended therapy according to the POC algorithm in Fig. 15.2: 25 mg/kg
tranexamic acid and 50 mg/kg fibrinogen concentrate (cryoprecipitate if fibrinogen concentrate is
not available). (f) Self-limiting fibrinolysis after reperfusion in a liver transplantation with a good
graft function. In the absence of bleeding, there is no need for therapeutic intervention. (g) Liberation
of heparinoids from the liver graft after reperfusion. Marked prolongation of CT and CFT in
INTEM® and almost normal CT in HEPTEM® (reference range for CT in INTEM® and HEPTEM®.
100–240 s). The effect of heparinoids usually is short acting and does not require any therapy in the
absence of bleeding. In principle, administration of FFP or PCC is not indicated here. (h) Deficiency
of vitamin K-dependent coagulation factors (II, VII, IX, X). Administration of PCC (or FFP if PCC
is not available) is indicated according to the POC algorithm in Fig. 15.2 in case of bleeding and
normalization of clot firmness (A10 or MCF) in EXTEM® and FIBTEM®, and CTEX >80 s (refer-
ence range for CT in EXTEM®. 40–80 s). Dosage of PCC = 25 (−40) IU/kg bw; dosage of FFP = 15
(−30) mL/kg bw. alp alpha angle, AT antithrombin, bw body weight, CFT clot formation time, CT
clotting time, EX EXTEM®, FFP fresh frozen plasma, FIB FIBTEM®, HEP HEPTEM®, IN
INTEM®, MCF maximum clot firmness, PCC 4-factor prothrombin complex concentrate, Quick
activity as % of normal based on PT, PT prothrombin time, aPTT activated partial thromboplastin
time, Run run time of the test, St. start time of the test
15 Perioperative Hemostasis in Hepatic Surgery 275
c d
0’ 1 EXTEM 0’ 1 EXTEM
St. : 18h27 St. : 09h02
CT : 73s CT : 57s
0’ 3 FIBTEM 0’ 3 FIBTEM
St. : 18h29 St. : 09h04
e f
0’ 1 EXTEM 0’ 1 EXTEM
St. : 17h06 St. : 08h27
Run : 65.1’
CT : 325s CT : 32s
CFT : 103s
MCF : 9mm MCF : 51mm
alp : 72°
0’ 3 FIBTEM 0’ 1 EXTEM
St. : 17h08 St. : 10h24
Run : 108.2
CT : 47s
CFT : 226s
MCF : 44mm
alp : 63°
g h
0’ 2 INTEM 0’ 1 EXTEM
St. : 15h04 St. : 23h38
CT : 621s CT : 119s
0’ 4 HEPTEM 0’ 3 FIBTEM
St. : 16h26 St. : 23h40
CT : 260s
(Görlinger 2006; Görlinger et al. 2011b). This algorithm has been shown to reduce
transfusion requirements in patients undergoing liver transplantation, without
increasing the incidence of thrombotic/thromboembolic events (Görlinger et al.
2010, 2012b; Kirchner et al. 2012). Use of ROTEM®/TEG® for perioperative coagu-
lation monitoring and targeted therapy of coagulopathy in patients undergoing vis-
ceral and transplant surgery is highly recommended in the European Society of
Anesthesiology’s guidelines for the management of severe perioperative bleeding
(Kozek-Langenecker et al. 2013). However, all therapeutic interventions which
reduce the need for blood transfusion and help avoid thrombotic/thromboembolic
events should be investigated further since the strongest predictor of survival in
patients undergoing liver transplantation is the number of blood transfusions (Esmat
Gamil et al. 2012).
It is of note that rFVIIa (NovoSeven®, Novo Nordisk A/S, Bagsværd, Denmark) is not
labeled for use in liver dysfunction and liver transplantation, and studies have failed to
demonstrate a significant benefit in bleeding of the upper gastrointestinal tract or in
liver transplantation (Chavez-Tapia et al. 2011a; Dasher and Trotter 2012; Pandit and
Sarode 2012; Simpson et al. 2012). Keeping a potential increased risk of thrombosis in
mind, the off-label use of rVIIa in patients with severe bleeding that is unresponsive to
other hemostatic interventions can be considered (Kozek-Langenecker et al. 2013).
According to our ROTEM®-guided algorithm, the administration of rFVIIa can be con-
sidered as a rescue therapy in case of ongoing diffuse bleeding despite optimization of
hemostasis, surgical hemostasis, and ROTEM®-guided hemostatic therapy (Fig. 15.2).
However, this has not been necessary in any cases since the implementation of our
ROTEM®-guided algorithm (Görlinger et al. 2010).
In line with the observations described above, patients with liver dysfunction are not
“auto-anticoagulated” (Pincus et al. 2012; Schaden et al. 2013; Tripodi et al. 2011),
but according to the findings of more global coagulation tests (thromboelastometry
and thrombin generation assays), they tend rather to hypercoagulability with the
inherent risk of thrombosis (Agarwal et al. 2012). Besides deep vein thrombosis,
portal vein thrombosis and pulmonary embolism thrombosis can also affect the arte-
rial system (hepatic artery thrombosis, myocardial infarction, or stroke). Even the
progression of liver fibrosis in chronic liver disease might be a consequence of pro-
coagulant imbalance (Tripodi et al. 2011). Hence, venous thromboembolism (VTE)
prophylaxis is required during the hospitalization of patients with liver dysfunction
(Koliscak and Maynor 2012). Despite this, 75 % of these patients do not receive
VTE prophylaxis (Dabbagh et al. 2010; Aldawood et al. 2011).
References
Adamzik M, Eggmann M, Frey UH et al (2010) Comparison of thromboelastometry with
procalcitonin, interleukin 6, and C-reactive protein as diagnostic tests for severe sepsis in
critically ill adults. Crit Care 14:R178
Agarwal B, Wright G, Gatt A et al (2012) Evaluation of coagulation abnormalities in acute liver
failure. J Hepatol 57:780–786
Al-Busafi SA, McNabb-Baltar J, Farag A, Hilzenrat N (2012) Clinical manifestations of portal
hypertension. Int J Hepatol 2012:203794
Aldawood A, Arabi Y, Aljumah A et al (2011) The incidence of venous thromboembolism and
practice of deep venous thrombosis prophylaxis in hospitalized cirrhotic patients. Thromb J 9:1
Bechmann LP, Sichau M, Wichert M et al (2011) Low-molecular-weight heparin in patients with
advanced cirrhosis. Liver Int 31:75–82
Bhavsar MS, Vora HB, Khiria LS, Giriyappa VH (2012) Portal hypertension: Effect of early
splenic artery ligation on platelets count during splenectomy. Saudi J Gastroenterol
18:380–383
Blasi A, Beltran J, Pereira A et al (2012) An assessment of thromboelastometry to monitor blood
coagulation and guide transfusion support in liver transplantation. Transfusion 52:1989–1998
Brenner T, Schmidt K, Delang M et al (2012) Viscoelastic and aggregometric point-of-care testing
in patients with septic shock – cross-links between inflammation and haemostasis. Acta
Anaesthesiol Scand 56:1277–1290
Caldwell SH, Sanyal AJ (2009) Coagulation disorders and bleeding in liver disease: future direc-
tions. Clin Liver Dis 13:155–157
Chavez-Tapia NC, Alfaro-Lara R, Tellez-Avila F, Barrientos-Gutiérrez T et al (2011a) Prophylactic
activated recombinant factor VII in liver resection and liver transplantation: systematic review
and meta-analysis. PLoS One 6:e22581
Chavez-Tapia NC, Barrientos-Gutierrez T, Tellez-Avila F et al (2011b) Meta-analysis: antibiotic
prophylaxis for cirrhotic patients with upper gastrointestinal bleeding – an updated Cochrane
review. Aliment Pharmacol Ther 34:509–518
Dabbagh O, Oza A, Prakash S (2010) Coagulopathy does not protect against venous thromboem-
bolism in hospitalized patients with chronic liver disease. Chest 137:1145–1149
Dara SI, Rana R, Afessa B et al (2005) Fresh frozen plasma transfusion in critically ill medical
patients with coagulopathy. Crit Care Med 33:2667–2671
280 K. Görlinger et al.
Dasher K, Trotter JF (2012) Intensive care unit management of liver-related coagulation disorders.
Crit Care Clin 28:389–398
Dik K, de Bruijne J, Takkenberg RB et al (2012) Factor XIII Val34Leu mutation accelerates the
development of fibrosis in patients with chronic hepatitis B and C. Hepatol Res 42:668–676
Ditisheim S, Goossens N, Spahr L, Hadengue A (2012) Coagulation and cirrhosis: new insight.
Rev Med Suisse 8:1652–1656
Doepker B, Mount KL, Ryder LJ et al (2012) Bleeding risk factors associated with argatroban
therapy in the critically ill. J Thromb Thrombolysis 34:491–498
Elias JE, Mackie I, Eapen CE et al (2013) Porto-pulmonary hypertension exacerbated by platelet
transfusion in a patient with ADAMTS13 deficiency. J Hepatol 58:827–830. doi:10.1016/j.
jhep.2012.11.003, pii: S0168-8278(12)00835-5
Esch JS, Jurk K, Knoefel WT et al (2010) Platelet activation and increased tissue factor expression
on monocytes in reperfusion injury following orthotopic liver transplantation. Platelets
21:348–359
Esmat Gamil M, Pirenne J, Van Malenstein H et al (2012) Risk factors for bleeding and clinical
implications in patients undergoing liver transplantation. Transplant Proc 44:2857–2860
Ewe K (1981) Bleeding after liver biopsy does not correlate with indices of peripheral coagulation.
Dig Dis Sci 26:388–393
Garcia DA, Baglin TP, Weitz JI et al (2012) American College of Chest Physicians. Parenteral
anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College
of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 141:24S–43S
Gatt A, Riddell A, Calvaruso V et al (2010) Enhanced thrombin generation in patients with
cirrhosis-induced coagulopathy. J Thromb Haemost 8:1994–2000
Görlinger K (2006) Coagulation management during liver transplantation. Hamostaseologie 26(3
Suppl 1):S64–S76
Görlinger K, Dirkmann D, Müller-Beißenhirtz H et al (2010) Thromboelastometry-based periop-
erative coagulation management in visceral surgery and liver transplantation: experience of 10
years and 1105 LTX. Liver Transplant 16(6 Suppl1):S86
Görlinger K, Dirkmann D, Hanke AA et al (2011a) First-line therapy with coagulation factor con-
centrates combined with point-of-care coagulation testing is associated with decreased alloge-
neic blood transfusion in cardiovascular surgery: a retrospective, single-center cohort study.
Anesthesiology 115:1179–1191
Görlinger K, Dirkmann D, Weber CF et al (2011b) Algorithms for transfusion and coagulation
management in massive haemorrhage. Anästh Intensivmed 52:145–159
Görlinger K, Bergmann L, Dirkmann D (2012a) Coagulation management in patients undergoing
mechanical circulatory support. Best Pract Res Clin Anaesthesiol 26:179–198
Görlinger K, Fries D, Dirkmann D et al (2012b) Reduction of fresh frozen plasma requirements
by perioperative point-of-care coagulation management with early calculated goal-directed
therapy. Transfus Med Hemother 39:104–113
Görlinger K, Dirkmann D, Solomon C, Hanke AA (2013) Fast interpretation of thromboelastome-
try in non-cardiac surgery: reliability in patients with hypo-, normo-, and hypercoagulability.
Br J Anaesth 110:222–230
Green L, Safa O, Machin SJ et al (2012) Development and application of an automated chromo-
genic thrombin generation assay that is sensitive to defects in the protein C pathway. Thromb
Res 130:780–784
Guyatt GH, Akl EA, Crowther M et al (2012) Executive summary: antithrombotic therapy and
prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based
Clinical Practice Guidelines. Chest 141:7S–47S
Haas T, Spielmann N, Mauch J et al (2012a) Comparison of thromboelastometry (ROTEM®) with
standard plasmatic coagulation testing in paediatric surgery. Br J Anaesth 108:36–41
Haas T, Spielmann N, Mauch J et al (2012b) Reproducibility of thrombelastometry (ROTEM®):
point-of-care versus hospital laboratory performance. Scand J Clin Lab Invest 72:313–317
Hall TC, Garcea G, Metcalfe M et al (2011) Management of acute non-cirrhotic and non-malignant
portal vein thrombosis: a systematic review. World J Surg 35:2510–2520
15 Perioperative Hemostasis in Hepatic Surgery 281
Hall DP, Lone NI, Watson DM et al (2012) Factors associated with prophylactic plasma transfu-
sion before vascular catheterization in non-bleeding critically ill adults with prolonged pro-
thrombin time: a case-control study. Br J Anaesth 109:919–927
Hanke AA, Joch C, Görlinger K (2013) Long-term safety and efficacy of a pasteurized nanofil-
trated prothrombin complex concentrate (Beriplex P/N): a pharmacovigilance study. Br J
Anaesth 110:764–772
Hatano E, Terajima H, Yabe S et al (1997) Hepatic artery thrombosis in living related liver trans-
plantation. Transplantation 64:1443–1446
Holland L, Warkentin TE, Refaai M et al (2009) Suboptimal effect of a three-factor prothrombin
complex concentrate (Profilnine-SD) in correcting supratherapeutic international normalized
ratio due to warfarin overdose. Transfusion 49:1171–1177
Kalina U, Bickhard H, Schulte S (2008) Biochemical comparison of seven commercially available
prothrombin complex concentrates. Int J Clin Pract 62:1614–1622
Kedia S, Goyal R, Mangla V et al (2012) Splenectomy in cirrhosis with hypersplenism: improvement
in cytopenias, child’s status and institution of specific treatment for hepatitis C with success.
Ann Hepatol 11:921–929
Khan H, Belsher J, Yilmaz M et al (2007) Fresh-frozen plasma and platelet transfusions are associated
with development of acute lung injury in critically ill medical patients. Chest 131:1308–1314
Kirchner C, Goerlinger K, Dirkmann D et al (2012) Safety and efficacy of prothrombin complex
and fibrinogen concentrates in liver transplantation. Liver Transplant 18(Suppl 1):S189
Koliscak L, Maynor L (2012) Pharmacologic prophylaxis against venous thromboembolism in
hospitalized patients with cirrhosis and associated coagulopathies. Am J Health Syst Pharm
69:658–663
Koscielny J, Ziemer S, Radtke H et al (2004) A practical concept for preoperative identification of
patients with impaired primary hemostasis. Clin Appl Thromb Hemost 10:195–204
Kozek-Langenecker S, Sørensen B, Hess JR, Spahn DR (2011) Clinical effectiveness of fresh
frozen plasma compared with fibrinogen concentrate: a systematic review. Crit Care
15:R239
Kozek-Langenecker SA, Afshari A, Albaladejo P et al (2013) Management of severe perioperative
bleeding. Guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 30:
1–112 [in press]
Larsen OH, Fenger-Eriksen C, Christiansen K et al (2011) Diagnostic performance and therapeutic
consequence of thromboelastometry activated by kaolin versus a panel of specific reagents.
Anesthesiology 115:294–302
Levy JH, Szlam F, Tanaka KA, Sniecienski RM (2012) Fibrinogen and hemostasis: a primary
hemostatic target for the management of acquired bleeding. Anesth Analg 114:261–274
Lier H, Vorweg M, Hanke A, Görlinger K (2013) Thromboelastometry guided therapy of severe
bleeding. Essener Runde algorithm. Hamostaseologie 33:51–61
Lisman T, Bongers TN, Adelmeijer J et al (2006) Elevated levels of von Willebrand Factor in cir-
rhosis support platelet adhesion despite reduced functional capacity. Hepatology 44:53–61
Lisman T, Bakhtiari K, Pereboom IT et al (2010) Normal to increased thrombin generation in
patients undergoing liver transplantation despite prolonged conventional coagulation tests.
J Hepatol 52:355–361
Lisman T, Bakhtiari K, Adelmeijer J et al (2012a) Intact thrombin generation and decreased fibri-
nolytic capacity in patients with acute liver injury or acute liver failure. J Thromb Haemost
10:1312–1319
Lisman T, Bakthiari K, Adelmeijer J et al (2012b) Intact thrombin generation and decreased fibri-
nolytic capacity in patients with acute liver failure argue against routine prophylactic correction
of coagulation prior to liver transplantation. Liver Transplant 18(Suppl 1):S187
Maruyama H, Takahashi M, Shimada T, Yokosuka O (2012) Emergency anticoagulation treat-
ment for cirrhosis patients with portal vein thrombosis and acute variceal bleeding. Scand J
Gastroenterol 47:686–691
Math SK, Sanders MA, Hollensead SC (2010) Unexpected laboratory diagnosis: acquired dysfi-
brinogenemia in a bleeding patient with liver disease. MLO Med Lab Obs 42:30–34
282 K. Görlinger et al.
Minov AF, Dziadz’ko AM, Rummo OO (2012) The thromboelastometric criteria of hemostasis
disorders correction during liver transplantation. Anesteziol Reanimatol 2:35–41
Mosnier LO (2011) Platelet factor 4 inhibits thrombomodulin-dependent activation of thrombin-
activatable fibrinolysis inhibitor (TAFI) by thrombin. J Biol Chem 286:502–510
Noval-Padillo JA, León-Justel A, Mellado-Miras P et al (2010) Introduction of fibrinogen in the
treatment of hemostatic disorders during orthotopic liver transplantation: implications in the
use of allogenic blood. Transplant Proc 42:2973–2974
Owren PA, Aas K (1951) The control of dicumarol therapy and the quantitative determination of
prothrombin and proconvertin. Scand J Clin Lab Invest 3:201–208
Pandit TN, Sarode R (2012) Blood component support in acquired coagulopathic conditions: is
there a method to the madness? Am J Hematol 8(Suppl 1):S56–S62
Pereboom IT, Adelmeijer J, van Leeuwen Y et al (2009a) Development of a severe von Willebrand
factor/ADAMTS13 dysbalance during orthotopic liver transplantation. Am J Transplant
9:1189–1196
Pereboom IT, de Boer MT, Haagsma EB et al (2009b) Platelet transfusion during liver transplanta-
tion is associated with increased postoperative mortality due to acute lung injury. Anesth Analg
108:1083–1091
Pincus KJ, Tata AL, Watson K (2012) Risk of venous thromboembolism in patients with chronic
liver disease and the utility of venous thromboembolism prophylaxis. Ann Pharmacother
46:873–878
Roullet S, Pillot J, Freyburger G et al (2010) Rotation thromboelastometry detects thrombocytopenia
and hypofibrinogenaemia during orthotopic liver transplantation. Br J Anaesth 104:422–428
Sakai T, Matsusaki T, Dai F et al (2012) Pulmonary thromboembolism during adult liver transplan-
tation: incidence, clinical presentation, outcome, risk factors, and diagnostic predictors. Br J
Anaesth 108:469–477
Sarani B, Dunkman WJ, Dean L et al (2008) Transfusion of fresh frozen plasma in critically ill
surgical patients is associated with an increased risk of infection. Crit Care Med
36:1114–1118
Schaden E, Metnitz PG, Pfanner G et al (2012) Coagulation Day 2010: an Austrian survey on the
routine of thromboprophylaxis in intensive care. Intensive Care Med 38:984–990
Schaden E, Saner FH, Goerlinger K (2013) Coagulation pattern in critical liver dysfunction. Curr
Opin Crit Care 19:142–148
Shah NL, Northup PG, Caldwell SH (2012) A clinical survey of bleeding, thrombosis, and blood
product use in decompensated cirrhosis patients. Ann Hepatol 11:686–690
Simpson E, Lin Y, Stanworth S et al (2012) Recombinant factor VIIa for the prevention and treatment
of bleeding in patients without haemophilia. Cochrane Database Syst Rev 3:CD005011
Smalberg JH, Leebeek FW (2009) Superimposed coagulopathic conditions in cirrhosis: infection and
endogenous heparinoids, renal failure, and endothelial dysfunction. Clin Liver Dis 13:33–42
Sørensen B, Spahn DR, Innerhofer P et al (2011) Clinical review: prothrombin complex concen-
trates -evaluation of safety and thrombogenicity. Crit Care 15:201
Stancheva A, Spassov L, Tzatchev K (2011) Correlation between rotation thrombelastometry
ROTEM analysis and standard haemostatic parameters during liver transplantation. Clin Lab
57:407–413
Stanworth SJ, Grant-Casey J, Lowe D et al (2011a) The use of fresh-frozen plasma in England:
high levels of inappropriate use in adults and children. Transfusion 51:62–70
Stanworth SJ, Walsh TS, Prescott RJ et al (2011b) A national study of plasma use in critical care:
clinical indications, dose and effect on prothrombin time. Crit Care 15:R108
Stellingwerff M, Brandsma A, Lisman T, Porte RJ (2012) Prohemostatic interventions in liver
surgery. Semin Thromb Hemost 38:244–249
Stravitz RT, Lisman T, Luketic VA et al (2012) Minimal effects of acute liver injury/acute liver
failure on hemostasis as assessed by thromboelastography. J Hepatol 56:129–136
Takaya H, Uemura M, Fujimura Y et al (2012) ADAMTS13 activity may predict the cumulative
survival of patients with liver cirrhosis in comparison with the Child-Turcotte-Pugh score and
the Model for End-Stage Liver Disease score. Hepatol Res 42:459–472
15 Perioperative Hemostasis in Hepatic Surgery 283
Tripodi A, Mannucci PM (2011) The coagulopathy of chronic liver disease. NEJM 365:147–155
Tripodi A, Primignani M, Chantarangkul V (2006) Thrombin generation in patients with cirrhosis:
the role of platelets. Hepatology 44:440–445
Tripodi A, Cappellini MD, Chantarangkul V et al (2009a) Hypercoagulability in splenectomized
thalassemic patients detected by whole-blood thromboelastometry, but not by thrombin genera-
tion in platelet-poor plasma. Haematologica 94:1520–1527
Tripodi A, Primignani M, Chantarangkul V et al (2009b) The coagulopathy of cirrhosis assessed
by thromboelastometry and its correlation with conventional coagulation parameters. Thromb
Res 124:132–136
Tripodi A, Legnani C, Lemma L (2010) Abnormal Protac-induced coagulation inhibition chromo-
genic assay results are associated with an increased risk of recurrent venous thromboembolism.
J Thromb Thrombolysis 30:215–219
Tripodi A, Anstee QM, Sogaard KK et al (2011) Hypercoagulability in cirrhosis: causes and con-
sequences. J Thromb Haemost 9:1713–1723
Tripodi A, Chantarangkul V, Primignani M et al (2012) Thrombin generation in plasma from
patients with cirrhosis supplemented with normal plasma: considerations on the efficacy of
treatment with fresh-frozen plasma. Intern Emerg Med 7:139–144
Trzebicki J, Flakiewicz E, Kosieradzki M et al (2010) The use of thromboelastometry in the
assessment of hemostasis during orthotopic liver transplantation reduces the demand for
blood products. Ann Transplant 15:19–24
Uemura M, Fujimura Y, Ko S et al (2011) Determination of ADAMTS13 and its clinical signifi-
cance for ADAMTS13 supplementation therapy to improve the survival of patients with
decompensated liver cirrhosis. Int J Hepatol 2011:759047
Villa E, Cammà C, Marietta M et al (2012) Enoxaparin prevents portal vein thrombosis and liver
decompensation in patients with advanced cirrhosis. Gastroenterology 143:1253–1260
Violi F, Basili S, Raparelli V et al (2011) Patients with liver cirrhosis suffer from primary haemo-
static defects? Fact or fiction? J Hepatol 55:1415–1427
Watson GA, Sperry JL, Rosengart MR et al (2009) Fresh frozen plasma is independently associated
with a higher risk of multiple organ failure and acute respiratory distress syndrome. J Trauma
67:221–227
Weber CF, Görlinger K, Meininger D et al (2012) Point-of-care testing: a prospective, randomized
clinical trial of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 117:531–547
Perioperative Hemostasis in
Pediatric Surgery 16
Thorsten Haas
T. Haas, MD
Department of Anesthesia, Zurich University Children’s Hospital,
Steinwiesstrasse 75, Zurich CH-8032, Switzerland
e-mail: [email protected]
if their bleeding history shows negative results (Dzik 2004; Koscielny et al. 2004;
Chee et al. 2008; Kozek-Langenecker 2010; Samkova et al. 2012). This approach is
based on the fact that the prediction of perioperative bleeding could not be reliably
determined by routine coagulation screening tests, while the combination of a clini-
cal examination, together with a detailed (family) history, showed superior results
in ensuring the detection of impaired hemostasis. For example, von Willebrand syn-
drome type I is the most common type of a clinically relevant congenital bleeding
disorder, with an incidence of 1 in 500; because it cannot be detected by a prolonged
activated partial thromboplastin time (aPTT), it will most likely come to light via a
detailed bleeding history. Templates of standardized questionnaires can be down-
loaded from the Austrian Society of Anesthesiology (ÖGARI) (http://www.oegari.
at/web_files/dateiarchiv/116/Recommendation%20questions%20bleeding%20
symptoms%202009.pdf), the Canadian Pediatric Bleeding Questionnaire (http://
www.ahcdc.ca/inheritedbleeds.html), and the International Society on Thrombosis
and Haemostasis bleeding assessment tool (http://www.isth.org/default/assets/file/
bleeding_type1_vwd.pdf).
If this preoperative approach reveals evidence of impaired hemostasis, or a child
suffers from a congenital or known acquired coagulation disorder, an interdisciplin-
ary work-up with a hematologist or another dedicated physician specialized in pedi-
atric bleeding disorders is indicated.
16.2.2.2 Thromboelastometry
Although data on the use of rotational thromboelastometry for intraoperative coagula-
tion management in children were scarce (Haas et al. 2008, 2012c; Hayashi et al.
2011; Romlin et al. 2011), a recently published meta-analysis showed significant
reduction in requirements of allogeneic blood products if thrombelastography/throm-
boelastometry was used to guide transfusion strategy (Afshari et al. 2011). In addition,
the use of thromboelastometry for pediatric care is recommended by the European
guidelines for perioperative bleeding management (Kozek-Langenecker et al. 2013).
Thus it seems reasonable to also use this advanced coagulation test for coagulation
management in children once it has been implemented as routine for adults.
16 Perioperative Hemostasis in Pediatric Surgery 287
While keeping in mind the functional maturity of the coagulation system, age-
dependent reference ranges for the ROTEM® (Oswald et al. 2010) or TEG® (Miller
et al. 1997) should be taken into account. The most striking finding from the evalu-
ation of these reference ranges is that children aged 0–3 months exhibited acceler-
ated coagulation and strong clot firmness, despite showing prolonged standard
plasma coagulation test results. Similarly to adults, fibrinogen concentrations, plate-
let count, and FXIII in children also contribute to clot firmness as measured using
ROTEM® assays. Furthermore, it was demonstrated that children aged 4–24 months
showed the lowest 2.5 % percentiles for clot strength, indicating low reserve when
exposed to hemodilution and blood loss. However, as a rule of thumb, from the 6th
month of life, adult reference ranges from ROTEM® can safely be used to guide
intraoperative coagulation management.
acidosis, inevitably leads to impairment of the coagulation process and may worsen
bleeding (Dirkmann et al. 2008). Thus, forced-air warming should be rigorously
performed, and temperature, as well as pH values, continuously measured during
pediatric surgery.
Meta-analyses of major pediatric surgery (Sethna et al. 2005; Tzortzopoulou et
al. 2008; Schouten et al. 2009; Goobie et al. 2011) and scoliosis surgery in children
(Tzortzopoulou et al. 2008) have nicely demonstrated that the prophylactic admin-
istration of antifibrinolytic agents (e.g. tranexamic acid or TXA) may decrease
blood loss and reduce allogeneic blood transfusion significantly. Therefore, the
prophylactic use of TXA can generally be recommended for major pediatric sur-
gery with estimated high blood loss or need for transfusion. Optimum doses are
still debated; reported dosing ranges from 10 to 100 mg/kg bolus, while high doses
might provoke seizures. Recent studies favored an initial bolus followed by con-
tinuous infusion due to TXA’s relatively short half-live of about 120 min. Based on
recently published data it seems reasonable to use an initial dose of 10–15 mg/kg
infused over 15 min, followed by continuous infusion of up to 5 mg/kg/h through-
out the entire surgical phase and upon transfer to the postoperative ward (Goobie
et al. 2013).
Alternative means to minimize blood transfusion in major pediatric surgery were
recently reviewed (Lavoie 2011). Results showed that acute normovolemic hemo-
dilution (mostly in adolescents) showed modest benefits. Other strategies such as
preoperative autologous donations (partly in combination with administration of
erythropoietin), intraoperative cell salvage (may be feasible if child’s body weight
is >10 kg), or deliberate hypotension (in the absence of a hypovolemic state), or
a combination of them, might be helpful in reducing allogeneic blood transfu-
sion. However, approaches must largely depend on individual expertise and local
facilities.
20
40
60
10 20 30 40 50 min
assay remains relatively low despite adequate substitution with fibrinogen. Although
it was shown that the FXIII-dependent increase in clot firmness can be displayed
using the ROTEM®/TEG® (Nielsen et al. 2004; Theusinger et al. 2010), to date no
commercially available point-of-care test for measuring FXIII activity is available.
Recommended means of substitution for FXIII include administration of a purified
factor concentrate, or FFP.
mm mm
60 60
40 40
20 20
20 20
40 40
60 60
10 20 30 40 10 20 30
Fig. 16.2 ROTEM® trace of low platelet count. INTEM MCF decreased (40 mm); FIBTEM MCF
normal (10 mm)
292 T. Haas
16.3.3.2.2 Rationale
Adequate thrombin generation is inevitably needed to induce the building of a sta-
ble clot. Notably, a perioperative decrease in thrombin generation, to levels that are
unlikely to initiate clot building, is typically found late on in severe bleeding, even
in neonates with lower vitamin K dependent coagulation factor levels. Unfortunately,
neither aPTT nor PT correlates with clinically relevant coagulopathies or bleeding
conditions (Johansson et al. 2010). Little evidence supports the use of standard
coagulation tests for the guidance of perioperative coagulation therapy (Spahn et al.
2013). As viscoelastic tests are not used everywhere, empirical thresholds still exist
to serve as rough estimates of disturbed hemostasis. In contrast, the prolonged coag-
ulation times of thromboelastometry, in conjunction with increased bleeding ten-
dency, might be interpreted as a relevant deficiency in coagulation factors and can
16.3.3.3 Hyperfibrinolysis
Besides the recommendation to use antifibrinolytic agents as prophylactic treat-
ment, there is no evidence based data published that hyperfibrinolysis is a common
problem during major surgery in children. However, due to the fact that hyperfibri-
nolysis is significantly associated with higher morbidity and mortality (Schochl et
al. 2009), and in light of current recommendations for adults, it seems adequate to
treat signs of hyperfibrinolysis (ROTEM® maximum lysis >15 % and maintenance
of MCF using the APTEM test) in children with 10–20 mg/kg tranexamic acid, if
relevant bleeding occurs (Fig. 16.4).
mm
60
40
20
20
40
60
Fig. 16.4 ROTEM® trace of
hyperfibrinolysis. Moderate
lysis. Severe lysis 10 20 30 40 50 min
294 T. Haas
Willebrand factor (vWF) released from their storage sites within endothelial cells
occur 30–60 min after desmopressin administration. If bleeding persists, adminis-
tration of either vWF/FVIII concentrate (50 IU vWF:c/kg) or a platelet transfusion
(20 ml/kg in children <15 kg; 1 apheresis unit platelets for children >15 kg) may be
initiated, depending on the assumed underlying coagulation disorder.
References
Afshari A, Wikkelso A, Brok J, Moller AM, Wetterslev J (2011) Thrombelastography (TEG) or
thromboelastometry (ROTEM) to monitor haemotherapy versus usual care in patients with
massive transfusion. Cochrane Database Syst Rev (3):CD007871
Andrew M, Paes B, Milner R, Johnston M, Mitchell L, Tollefsen DM, Powers P (1987) Development
of the human coagulation system in the full-term infant. Blood 70(1):165–172
Andrew M, Paes B, Milner R, Johnston M, Mitchell L, Tollefsen DM, Castle V, Powers P
(1988) Development of the human coagulation system in the healthy premature infant. Blood
72(5):1651–1657
Andrew M, Vegh P, Johnston M, Bowker J, Ofosu F, Mitchell L (1992) Maturation of the hemo-
static system during childhood. Blood 80(8):1998–2005
Chee YL, Crawford JC, Watson HG, Greaves M (2008) Guidelines on the assessment of bleeding
risk prior to surgery or invasive procedures. British Committee for Standards in Haematology.
Br J Haematol 140(5):496–504
16 Perioperative Hemostasis in Pediatric Surgery 295
Danes AF, Cuenca LG, Bueno SR, Mendarte Barrenechea L, Ronsano JB (2008) Efficacy and
tolerability of human fibrinogen concentrate administration to patients with acquired fibrino-
gen deficiency and active or in high-risk severe bleeding. Vox Sang 94(3):221–226
Dickneite G, Pragst I, Joch C, Bergman GE (2009) Animal model and clinical evidence indicating
low thrombogenic potential of fibrinogen concentrate (Haemocomplettan P). Blood Coagul
Fibrinolysis 20(7):535–540
Dirkmann D, Hanke AA, Gorlinger K, Peters J (2008) Hypothermia and acidosis synergistically
impair coagulation in human whole blood. Anesth Analg 106(6):1627–1632
Dzik WH (2004) Predicting hemorrhage using preoperative coagulation screening assays. Curr
Hematol Rep 3(5):324–330
Ekert H, Brizard C, Eyers R, Cochrane A, Henning R (2006) Elective administration in infants of
low-dose recombinant activated factor VII (rFVIIa) in cardiopulmonary bypass surgery for
congenital heart disease does not shorten time to chest closure or reduce blood loss and need
for transfusions: a randomized, double-blind, parallel group, placebo-controlled study of
rFVIIa and standard haemostatic replacement therapy versus standard haemostatic replacement
therapy. Blood Coagul Fibrinolysis 17(5):389–395
Fenger-Eriksen C, Lindberg-Larsen M, Christensen AQ, Ingerslev J, Sorensen B (2008) Fibrinogen
concentrate substitution therapy in patients with massive haemorrhage and low plasma fibrino-
gen concentrations. Br J Anaesth 101(6):769–773
Fenger-Eriksen C, Jensen TM, Kristensen BS, Jensen KM, Tonnesen E, Ingerslev J, Sorensen B
(2009a) Fibrinogen substitution improves whole blood clot firmness after dilution with
hydroxyethyl starch in bleeding patients undergoing radical cystectomy: a randomized, pla-
cebo-controlled clinical trial. J Thromb Haemost 7(5):795–802
Fenger-Eriksen C, Tonnesen E, Ingerslev J, Sorensen B (2009b) Mechanisms of hydroxyethyl
starch-induced dilutional coagulopathy. J Thromb Haemost 7(7):1099–1105
Fenger-Eriksen C, Moore GW, Rangarajan S, Ingerslev J, Sorensen B (2010) Fibrinogen estimates
are influenced by methods of measurement and hemodilution with colloid plasma expanders.
Transfusion 50(12):2571–2576
Fries D (2006) Dilutional coagulopathy: development, diagnostic options and management.
Hamostaseologie 26(3 Suppl 1):S15–S19
Fries D, Innerhofer P, Perger P, Gutl M, Heil S, Hofmann N, Kneifel W, Neuner L, Pernerstorfer T,
Pfanner G, Schochl H, Ziegler B, Kolblinger C, Kozek-Langenecker S (2010) Coagulation
management in trauma-related massive bleeding.- Recommendations of the Task Force for
Coagulation (AGPG) of the Austrian Society of Anesthesiology, Resuscitation and Intensive
Care Medicine (OGARI). Anasthesiol Intensivmed Notfallmed Schmerzther 45(9):552–561
Friesen RH, Perryman KM, Weigers KR, Mitchell MB, Friesen RM (2006) A trial of fresh autolo-
gous whole blood to treat dilutional coagulopathy following cardiopulmonary bypass in
infants. Paediatr Anaesth 16(4):429–435
Gerlach R, Tolle F, Raabe A, Zimmermann M, Siegemund A, Seifert V (2002) Increased risk for
postoperative hemorrhage after intracranial surgery in patients with decreased factor XIII activ-
ity: implications of a prospective study. Stroke 33(6):1618–1623
Goobie SM, Meier PM, Pereira LM, McGowan FX, Prescilla RP, Scharp LA, Rogers GF, Proctor
MR, Meara JG, Soriano SG, Zurakowski D, Sethna NF (2011) Efficacy of tranexamic acid in
pediatric craniosynostosis surgery: a double-blind, placebo-controlled trial. Anesthesiology
114(4):862–871
Goobie SM, Meier PM, Sethna NF, Soriano SG, Zurakowski D, Samant S, Pereira LM (2013)
Population pharmacokinetics of tranexamic acid in paediatric patients undergoing craniosynos-
tosis surgery. Clin Pharmacokinet 52(4):267–276
Guzzetta NA, Miller BE (2011) Principles of hemostasis in children: models and maturation.
Paediatr Anaesth 21(1):3–9
Haas T, Fries D, Velik-Salchner C, Oswald E, Innerhofer P (2008) Fibrinogen in craniosynostosis
surgery. Anesth Analg 106(3):725–731
Haas T, Korte W, Spielmann N, Mauch J, Madjdpour C, Schmugge M, Weiss M (2012a)
Perioperative course of FXIII in children undergoing major surgery. Paediatr Anaesth
22(7):641–646
296 T. Haas
Tzortzopoulou A, Cepeda MS, Schumann R, Carr DB (2008) Antifibrinolytic agents for reducing
blood loss in scoliosis surgery in children. Cochrane Database Syst Rev (3):CD006883
Uhrig L, Blanot S, Baugnon T, Orliaguet G, Carli PA, Meyer PG (2007) Use of recombinant acti-
vated factor VII in intractable bleeding during pediatric neurosurgical procedures. Pediatr Crit
Care Med 8(6):576–579
Weinkove R, Rangarajan S (2008) Fibrinogen concentrate for acquired hypofibrinogenaemic
states. Transfus Med 18(3):151–157
Perioperative Hemostasis in Obstetrics
17
Albrice Levrat, Christian Kern, and Cyril Huissoud
17.1 Introduction
A. Levrat
Department of Critical Care, Annecy Regional Hospital, Annecy, France
C. Kern
Department of Anesthesia, University Hospital Vaud CHUV, Lausanne, Switzerland
C. Huissoud (*)
Department of Obstetrics and Gynecology, Croix Rousse University Hospital,
Hospices Civils de Lyon, Lyon, France
e-mail: [email protected]
PPH is defined as blood loss >500 mL within 24 h of vaginal delivery (Subtil et al.
2004). Apart from certain exceptional cases, the occurrence of bleeding is unpre-
dictable. There are multiple hemorrhage mechanisms (e.g., wounds to the birth
canal, post-cesarean hemoperitoneum), and not all of them correspond to placental
expulsion pathologies. Primary uterine atony is the most frequent cause of PPH,
while uterine inertia may be secondary and can complicate any type of PPH.
PPH can be aggravated by uncontrollable uterine bleeding, caused by two
main mechanisms: tissue hypoxia associated with acute anemia or reduced circu-
lation or the occurrence of a coagulopathy leading to abnormal bleeding of the
placental bed.
actively assisted via manual removal of the placenta from the uterus. Placental expul-
sion is systematically followed by a uterine examination, mainly in order to check
that the uterine cavity is empty.
Four phases can be distinguished in a normal placental expulsion:
• Phase one: uterine contraction immediately after the fetus is pushed out. Because
the uterus is less distended, it undergoes a simple phenomenon of passive elastic
retraction. This phase is usually followed by a few minutes of rest during which
the contractions are less intense.
• Phase two: characterized by renewed, more intense uterine contractions. These
contractions will lead to a reduction of the placental insertion site and will put a
strain on the chorionic villi in the compact layer of the basal plate. This creates a
gap between the compact outer layer and the spongy intermediate layer of the
decidua, and the lesion of these uteroplacental blood vessels leads to the forma-
tion of a hematoma. The hematoma widens the gap and completes the detach-
ment of the placenta.
• Phase three: processus of hemostasis. This crucial phase is carried out by:
• The formation of clots in the affected uteroplacental blood vessels. Hemostasis
is set off by an abundant liberation of tissue factor as the gap between the layers
grows. It is reinforced by the modifications to coagulation which develop during
pregnancy.
• Uterine contractions that will seal off the open blood vessels embedded in the
uterine wall.
• Phase four: the migration of the freed placenta down the birth canal and its expulsion
from the vagina.
Relaxation of the uterus, or an intrauterine retention of placental tissue, may
lead to the development of a hemorrhage. It is imperative to carry out a uterine
examination and to keep the patient under strict observation.
In case of a cesarean section, placental expulsion is actively managed or it is manu-
ally extracted; uterine examination is systematic. Cesarean birth is associated with an
increased risk of hemorrhage: intrinsically (sectioning of veins during hysterotomy,
hemoperitoneum, and hematoma), due to its causes (e.g., placenta previa, HELLP
syndrome), and due to more frequent uterine atony.
Hemostasis of the uterine placental bed plays an important role in reducing the risks
of hemorrhage. The hemostatic system enters into action as soon as the placenta
detaches itself, allowing the formation of clots in the blood vessels. The modifications
in coagulation activity linked to pregnancy are designed to increase the effectiveness
of hemostasis.
Fibrinogen is the blood factor which undergoes the greatest variations in concentra-
tion during pregnancy, but coagulation modifications are by no means limited to
fibrinogen. Fibrin formation depends directly on the capacity to generate thrombin.
Understanding all these modifications is therefore fundamental to understanding its
impact on the blood clot.
1981), which plays an important role in the generation of thrombin. The reduction
of this factor occurs in order to counterbalance increases in the other factors
(Holmes and Wallace 2005). Tissue factor plays a key role in triggering coagula-
tion in vivo. The absence of an increase in soluble tissue factor (Bellart et al.
1998b), the reduction in monocyte tissue factor (Holmes et al. 2002), and the
increase in tissue factor pathway inhibitor (TFPI) (Sandset et al. 1989) may con-
tribute to limiting the risks of activating coagulation during pregnancy.
• The factors inhibiting coagulation pathways are also affected by pregnancy.
The level of antithrombin remains stable (Hellgren and Blomback 1981; Stirling
et al. 1984) or increases slightly (Dargaud et al. 2010). The level of protein C
hardly changes or also increases slightly, perhaps due to the influence of increased
levels of thrombomodulin; concentrations of its inhibitor do increase (Hellgren
2003). The levels of the free form of protein S and the total level of protein S both
diminish (Clark et al. 1998; Kjellberg et al. 1999). Holmes and Wallace sug-
gested that these modifications were aimed at a reducing coagulation inhibition
(Holmes and Wallace 2005).
• The fibrinolytic system is also subject to changes. Overall fibrinolytic activity is
reduced despite increases in plasminogen and its activators, tissue plasminogen
activator (tPA) and urokinase (Nakashima et al. 1996). This is because of parallel
increases of plasminogen activator inhibitor 1 (PAI-1) and especially of plas-
minogen activator inhibitor 2 (PAI-2) secreted by the placenta. The level of PAI-
2, which is very low outside of pregnancy, rises considerably and may lead to
strong inhibition of the conversion of plasminogen to plasmin (Nakashima et al.
1996; Bellart et al. 1997). However, the concentration of thrombin-activatable
fibrinolysis inhibitor (TAFI) is not affected by pregnancy (Chetaille et al. 2000),
and the level of α2-antiplasmin remains stable or increases slightly (Hellgren and
Blomback 1981).
Pregnancy has little effect on standard plasmatic tests such as prothrombin time
(PT, or Quick’s time) or activated coagulation time (ACT), and they are poor at
detecting hypercoagulable states (Holmes and Wallace 2005; Huissoud et al. 2009).
During pregnancy, there is an increase in activation markers for coagulation,
such as the thrombin-antithrombin complex (TAT) and fragments Fi and F2 released
by the degradation of prothrombin (Clark et al. 1998; Holmes and Wallace 2005;
Uchikova and Ledjev 2005). Dargaud et al. (2010) suggested that the increase in F1
and F2 is linked to the significant increase of FVII. Likewise, during pregnancy,
there is an increase in the levels of fibrinopeptide A (which is a product of the con-
version of fibrinogen to fibrin) and of d-dimers (Bellart et al. 1998a, b; Uchikova
and Ledjev 2005; Dargaud et al. 2010). The best way to evaluate coagulability is to
calculate the capacity to generate thrombin or the endogenous thrombin potential
(ETP). Recent work by Dargaud et al. demonstrated that ETP levels increased from
the first to the third trimester (Dargaud et al. 2010). These results support data from
thromboelastometry which show an increase in the α angle and a reduction in clot
formation time (CFT) as gestation progresses. All these results reflect an increase in
the “capacity to coagulate” which occurs from the first through to the third trimester
(Huissoud et al. 2009; Dargaud et al. 2010).
304 A. Levrat et al.
PPH is a true race against time. Gaining hemostatic control of macroscopic bleeding
remains an absolute priority, whether this must be done using surgery or via embo-
lization using interventional radiology.
At the same time, resuscitation management must be aggressive: the early use of
hemostatic products associated with constant monitoring of coagulation efficacy is
mandatory. As with any serious situation, the existence of written protocols is essen-
tial. Adequate management of these patients will require nonstop collaboration
between specialist obstetricians, anesthetists, and radiologists.
side effects. PCC therefore seem to be a potential solution for the management of
such hemorrhages despite the fact that there are currently no specific guidelines.
Several retrospective studies have emphasized the value of PCC during periopera-
tive bleeding, especially in combination with the administration of fibrinogen. The
advantages of a strategy combining the targeted transfusion of coagulation factors
with thromboelastographic monitoring seem to be an interesting alternative to the
conventional transfusion strategies which are still often quite empirical. However,
the thromboembolic risks linked to using PCC have yet to be properly evaluated:
vigilance is recommended when using these products during PPH.
As with severe trauma, platelet levels should be maintained at over 100,000/mm3
during active bleeding: Simon et al.’s retrospective analysis identified a lower
threshold to be a risk factor independent of PPH. Thus, the more the platelet level
decreases, the greater the number of pRBC that should be administered.
The administration of fibrinogen is recommended if the perfusion of FFP is
unable to maintain fibrinogen levels above 2 g/L. Indeed, in the study by Charbit
et al., fibrinogen concentration was the only independent parameter to be associated
with PPH developing a severe course (a fibrinogen level of less than 2 g/L gave a
100 % positive predictive value of serious bleeding).
In addition to fibrinogen’s role in clot formation, it has the other advantages of
only requiring a low infusion volume and very little preparation time. Thus, fibrino-
gen use allows a reduction of transfusion volume requirements during massive bleed-
ing. These studies also underlined the importance of regularly measuring fibrinogen
levels during PPH, while keeping in mind that normal fibrinogen levels during late
pregnancy are around 4 g/L.
United Kingdom recommendations emphasize that a level of at least 1.5 g/L is
needed to improve hemostasis.
Thromboelastometry allows for close monitoring of fibrinogen levels in plasma:
in patients suffering PPH, this level is highly correlated to the amplitude of the fibrin
clot measured after 5 min, using rotational thromboelastometry (ROTEM®). When
using an algorithm with appropriate transfusion thresholds, ROTEM® can thus help
save on transfusion requirements, as in cardiac surgery. A FIBTEM® or TEG ampli-
tude measured below 12 mm (about 2 g/L fibrinogen) is a sign that justifies early
administration of fibrinogen. Finally, monitoring fibrinogen levels using ROTEM’s
FIBTEM® test highlights the potential effects that fluids can have on coagulopathy:
in vitro clot firmness decreases significantly after hemodilution using colloids rather
than crystalloids.
Several case studies, case series, and registry studies have been published on the
use of recombinant activated factor VII (rFVIIa). Results suggest that rFVIIa provides
a reduction in transfusion requirements and facilitates surgical hemostasis in 60–80 %
of obstetric cases. The dose is variable: 90–120 μg/kg. rFVIIa can be used after the
failure of other medical and surgical treatments but also prior to hysterectomy. In order
to optimize rFVIIa’s efficacy, the patient’s blood should have a pH >7.20, a tempera-
ture >34 °C, a fibrinogen level >1 g/L, and platelets above 50,000/mm3.
Ferrer et al.’s meta-analysis showed that tranexamic acid reduces obstetric bleed-
ing. Since then, a randomized multicenter study evaluating high doses of tranexamic
308 A. Levrat et al.
acid for use in PPH (4 g in the first hour, then 1 g/h for 6 h) versus placebo showed
significant reductions in blood loss and transfusion requirements in the group
receiving tranexamic acid. Side effects (nausea, vomiting, or problems of vision)
are more common however, and appear to be linked to the high doses; an interna-
tional randomized trial (WOMAN Trial) is underway using lower doses. There has
been a great deal of recent interest in antifibrinolytics in the context of traumatic
hemorrhage: they are inexpensive products, reduce blood product use, and do not
appear to increase the risk of thromboembolism. In short, the significant efficacy of
antifibrinolytic agents demonstrates the importance of fibrinolysis in the pathophys-
iology of PPH.
In practice, the evaluation of hyperfibrinolysis using conventional biological
tests (d-dimers, fibrin degradation products, and euglobulin lysis time) is not easily
applicable in emergency situations. On the other hand, thromboelastometry permits
a rapid diagnosis of acute hyperfibrinolysis, such as coagulopathies associated with
catastrophic amniotic fluid embolism.
Overall, optimal management of PPH necessitates:
• The existence of protocols adapted to the institution’s organization of critical
obstetric care and transfusion services, in order to optimize timely coordination
• Bearing in mind the limits of standard hemostasis tests in this type of situation
and acknowledging the increasing preference for using thromboelastometric
techniques to guide the transfusion process
• Several weeks of monitoring for patients who have suffered a severe hemor-
rhagic event (this appears indispensable; indeed, in a series of 317 patients with
severe PPH, 22 % of them, in fact, had a constitutional defect of hemostasis:
hypofibrinogenemia, drops in von Willebrand factor or factor XI)
References
Bellart J, Gilabert R et al (1997) Fibrinolysis changes in normal pregnancy. J Perinat Med
25(4):368–372
Bellart J, Gilabert R et al (1998a) Coagulation and fibrinolytic parameters in normal pregnancy and
in pregnancy complicated by intrauterine growth retardation. Am J Perinatol 15(2):81–85
Bellart J, Gilabert R et al (1998b) Endothelial cell markers and fibrinopeptide A to D-dimer ratio
as a measure of coagulation and fibrinolysis balance in normal pregnancy. Gynecol Obstet
Invest 46(1):17–21
Bouvier-Colle MH, Deneux C et al (2004) Estimation de la mortalité marternelle en France: une
nouvelle méthode. J Gynecol Obstet Biol Reprod (Paris) 33(5):421–429
Cerneca F, Ricci G et al (1997) Coagulation and fibrinolysis changes in normal pregnancy.
Increased levels of procoagulants and reduced levels of inhibitors during pregnancy induce a
hypercoagulable state, combined with a reactive fibrinolysis. Eur J Obstet Gynecol Reprod
Biol 73(1):31–36
Chetaille P, Alessi MC et al (2000) Plasma TAFI antigen variations in healthy subjects. Thromb
Haemost 83(6):902–905
Clark P, Brennand J et al (1998) Activated protein C sensitivity, protein C, protein S and coagula-
tion in normal pregnancy. Thromb Haemost 79(6):1166–1170
Dargaud Y, Hierso S et al (2010) Endogenous thrombin potential, prothrombin fragment 1 + 2 and
D-dimers during pregnancy. Thromb Haemost 103(2):469–471
17 Perioperative Hemostasis in Obstetrics 309
18.1 Introduction
Injury is a major public health problem: it is the leading cause of death in the first
four decades of life (Krug et al. 2000; Eastridge et al. 2006). Exsanguination
accounts for up to 50 % of trauma deaths in the first 24 h and for up to 80 % in the
operating theater (Association of Anaesthetists of Great et al. 2010). Despite signifi-
cant advances in trauma care, exsanguination remains the leading cause of prevent-
able death (Sauaia et al. 1995; Demetriades et al. 2004; Kauvar and Wade 2005;
Gruen et al. 2006; Kauvar et al. 2006; Cothren et al. 2007). Caring for a severely
injured and bleeding patient is to face a challenge of timely and effective hemor-
rhage control and rapid restoration of physiology.
Coagulopathy is common after trauma and directly related to poor outcome (Brohi
et al. 2003; MacLeod et al. 2003; Maegele et al. 2007). Over the last years, transfusion
strategies for patients with severe trauma-related bleeding have undergone important
changes due to a better understanding of epidemiology, the mechanisms and conse-
quences of bleeding and coagulopathy (Spinella and Holcomb 2009). Impaired hemo-
stasis following trauma was thought to be essentially due to a loss or inhibition of
coagulation proteases, similar as in surgical haemorrhage. In 2003, two large observa-
tional studies found that 1 in 4 trauma patients presents impaired coagulation on hos-
pital arrival, independent of exogenous factors (Brohi et al. 2003; MacLeod et al.
2003). This identification of early coagulopathy has progressed to the recognition of
an endogenous component of trauma-related hemostatic impairment, and the term
“acute traumatic coagulopathy” (ATC) has been adopted (Brohi et al. 2003). ATC is
C. Heim (*)
Department of Anaesthesia, Centre Hospitalier Universitaire Vaudois CHUV,
Lausanne, Switzerland
e-mail: [email protected]
K. Brohi
Trauma Service, Department of Surgery,
Royal London Hospital, London, UK
+ + +
VIIa IXa Xa
Platelet Thrombin Fibrin FDPs
VIII V +
− −
Plasmin
+
−
PAI- tPA
−1
aPC
Thrombin-TM
PC TM TM TM TM Endothelium
Fig. 18.1 The ATC pathway (From Brohi et al. (2007a) with permission)
Trauma-induced
coagulopathy
platelet count and clot integrity, as analyzed by viscoelastic tests, has been demon-
strated. However, studies examining functional characterization of platelets after
trauma are sparse, and the pathophysiological role of platelets in this condition has
yet to be defined (Rugeri et al. 2007; Plotkin et al. 2008).
Trauma-related impairment of hemostasis has been described with multiple terms,
the most frequently used being acute traumatic coagulopathy (ATC), acute coagu-
lopathy of trauma shock (ACoTS), endogenous acute coagulopathy (EAC), and
trauma-induced coagulopathy (TIC). In this chapter, we use ATC as the term describ-
ing the endogenous component of trauma-related coagulopathy, while TIC describes
the concomitant factors worsening coagulopathy further (Frith et al. 2010). While
ATC occurs immediately after the injury, other mediators of TIC develop over time
and in conjunction with measures against shock and resuscitation. TIC is multifacto-
rial and involves all the components of the hemostatic system, notably dysfunctional
platelets and activation of the endothelium (Fig. 18.2).
Six key initiators of TIC have been identified (Hess et al. 2008):
• Tissue trauma
• Shock
• Hemodilution
• Hypothermia
• Acidemia
• Inflammation
injuries destroy more cells than an isolated penetrating injury. As mentioned above,
tissue injury is a prerequisite, but in isolation is not sufficient to have a full picture
of ATC (Frith et al. 2010).
18.2.2 Shock
Shock – next to tissue injury – seems to be the other key driver of ATC. Several
studies have shown a linear relationship between the extent of systemic hypoperfu-
sion, measured by base deficit (BD), and the degree of coagulopathy (Brohi et al.
2007a, b; Niles et al. 2008; Frith et al. 2010; Jansen et al. 2011). Systemic hypoper-
fusion seems to activate protein C via increased generation of the thrombin-
thrombomodulin complex. aPC exerts an anticoagulant effect by irreversibly
deactivating factors Va and VIIIa (Brohi et al. 2007a, b). Further, in shock, fibrino-
lysis is exacerbated through the combined effects of endothelial tPA release and the
shock-mediated inhibition of PAI-1(Brohi et al. 2008). Cellular hypoperfusion and
activation of inflammation are closely related. Hemorrhagic shock and resuscitation
lead to alterations in microcirculation via the release of a multitude of inflammatory
mediators, cytokines, and oxidants. The activation of inflammation and the induc-
tion of apoptotic cell death as a consequence of hypoxic cellular damage and
ischemia-reperfusion phenomena are the presumed causes of secondary organ
injury leading to organ failure and death (Duchesne et al. 2010). At the coagulation
level, cellular hypoxia, thrombin, and various cytokines will together promote endo-
thelial cell activation (ECA) (Faller 1999). ECA will induce a downregulation of
thrombomodulin (TM) and enhance fibrinolysis via increased levels of PAI-1.
Destruction of the endothelial glycocalyx limits activation of antithrombin and
upregulates platelet-activating factor and the expression of TF (Johansson et al.
2011) (Fig. 18.3).
18.2.3 Hemodilution
During fluid resuscitation, the dilution of coagulation factors can be a major con-
tributor to clinical coagulopathy (Armand and Hess 2003; Maegele et al. 2007).
High volumes (>2,000 ml) of crystalloids or colloids during prehospital resuscita-
tion are associated with a worse coagulation profile, an increased need for blood
transfusion, and a higher incidence of organ failure (Wafaisade et al. 2010;
Hubetamann et al. 2011; Hussmann et al. 2011). Further, administration of blood-
products such as packed red blood cells (PRBC), fresh frozen plasma (FFP), and
platelets (PLT) may cause significant dilution due to the anticoagulant properties of
their storage solutions. Coagulopathy is directly proportional to the volume of clear
fluid infused, independent of their type (Haas et al. 2008a, b; Bolliger et al. 2009).
Even in the absence of exogenous resuscitation, endogenous hemodilution occurs as
a physiological response to hypovolemia. The shift of hypocoagulable fluid from
the cellular and interstitial space into plasma will lead to minor dilution of
316 C. Heim and K. Brohi
Prothrombin ratio
grouped according to injury ∗ ∗
1.7
severity score (ISS) and base ∗
1.6
deficit (BD). (b) Mortality of
patients grouped according to
1.5 ∗ ∗
1.4 ∗
ISS and BD (From Frith et al. ∗
(2010) with permission) 1.3
1.2 >12
1.1 6.1−12
1 0.6−6
<16 ≤0
16−24 Base deficit
25−35
ISS >35 (mmol L−1)
∗
b
∗
70
60 ∗ ∗
Mortality (%)
50
∗ ∗
40
∗ ∗
30
∗ ∗
20 ∗ ∗
∗
>12
10 ∗ 6.1−12
0 0.6−6
<16 ≤0
16−24 Base deficit
25−35
ISS >35 (mmol L−1)
18.2.4 Hypothermia
Trauma patients are prone to hypothermia. This is due to either environmental fac-
tors or administration of cold resuscitation fluids combined with the reduced heat
production of maximally vasoconstricted musculature. Hypothermia has important,
reversible effects on the functionality of coagulation factors, platelet protease activ-
ity, and fibrinolysis (Wolberg et al. 2004; Thorsen et al. 2011). The activity of TF or
FVIIa complex decreases in a linear fashion with reduced body temperature.
Hypothermia reduces platelet adhesion to vWF on endothelial surfaces, limiting the
signal transduction from initial adhesion to activation. Whereas platelet function
18 Perioperative Hemostasis in Trauma 317
18.2.5 Acidemia
18.2.6 Inflammation
Coagulation and inflammation are closely linked (Esmon 2005). The combination
of tissue injury and shock is an important inducer of a systemic inflammatory
response syndrome. Inflammation initiates clotting, decreases the activity of physi-
ological anticoagulatory mechanisms, and impairs the fibrinolytic system (Gruen
et al. 2012). The innate immune system is activated early after traumatic injury
through the release of intracellular debris, exposure of the subendothelium, endo-
thelial activation, and the hypoxic environment of the microcirculation. Further,
resuscitation fluids have been shown to have immunomodulating properties.
Crystalloids and colloids cause neutrophil activation in patients in hemorrhagic
shock (Rhee et al. 1998, 2000), and crystalloids lead to degradation of glycocalyx,
contributing to further activation of inflammation (Van der Linden and Ickx 2006).
Immunomodulatory treatment strategies, limiting the activation of the inflammatory
response, might constitute promising novel approaches in the future.
(INR) (Brohi et al. 2003). Laboratory-based clotting tests have logistic issues, limit-
ing their utility in acute trauma care, and there is an emerging consensus that such
tests are inappropriate for monitoring coagulopathy and guiding therapy in trauma
hemorrhage (Segal et al. 2005; Yuan et al. 2007; Toulon et al. 2009; Kashuk et al.
2010a, b). The absence of rapid diagnostic tools has been shown to lead to delayed
correction of ATC and suboptimal use of blood products (Hess and Hiippala 2005;
Gonzalez et al. 2007; Davenport et al. 2011). There is renewed interest in viscoelas-
tic hemostatic assays (VHA), such as thromboelastography (TEG®) and rotational
thromboelastometry (ROTEM®). Several studies support the superiority of VHA in
terms of guiding adequate blood product provision in comparison with traditional
plasma-based coagulation tests (Afshari et al. 2011), and the use of TEG/ROTEM
in massively bleeding trauma patients is recommended in current guidelines
(Rossaint et al. 2010). Davenport et al. (2011) identified the typical thromboelasto-
graphic profile of ATC as slow clot formation and persistently reduced clot strength
of around 40 %. This prospective study (the largest to date) on the diagnostic modal-
ities of ATC identified a clot amplitude at 5 min (CA5) ≤35 mm as diagnostic
marker of ATC; it had a greater sensitivity than PT in predicting the need for mas-
sive transfusion. Furthermore, all ROTEM parameters showed a negative predictive
value of close to 100 % for massive transfusion, indicating that a normal ROTEM
trace can be used for timely termination of massive hemorrhage protocols (Davenport
et al. 2011). Based on current knowledge, it would seem reasonable to implement
VHA-based algorithms allowing for a goal-directed resuscitation strategy, although
evidence from clinical trials is still lacking (Dzik et al. 2011).
Tissue oxygenation remains the principle goal of resuscitation. Due to the relationship
between tissue oxygenation, impaired hemostasis, and mortality, the most important
320 C. Heim and K. Brohi
blood component for the prevention and treatment of ATC are packed red blood cells
(Hess et al. 2008; Dzik et al. 2011). Erythrocytes contribute to hemostasis by promot-
ing marginalization of platelets and allowing their interaction with the endothelium
(Valeri et al. 2007). An inverse correlation has been demonstrated between the hema-
tocrit and bleeding time in vitro (Eugster and Reinhart 2005). Furthermore, erythro-
cytes support thrombin generation through the exposure of procoagulant phospholipids
(Peyrou et al. 1999) and activate platelets via liberation of adenosine diphosphate
(ADP) (Joist et al. 1998). Clearly defined targets for hemoglobin or hematocrit during
active hemorrhage are lacking, but the most recent European guidelines recommend a
hemoglobin target of 7–9 g/l (Spahn 2013).
Even in severely injured patients, PLT counts on admission are rarely below critical
levels (Murthi et al. 2008; Stansbury et al. 2013). The fall in PLT count seen later in
the resuscitation time course is probably due to dilution. Low PLT count at admis-
sion, however, is strongly related to mortality and a need for PRBC (Brown et al.
2011; Stansbury et al. 2013). High PLT to PRBC transfusion ratios have reportedly
increased survival in massively transfused patients (Inaba et al. 2010; Holcomb
et al. 2011; Johansson et al. 2012). However, evidence is even scarcer than for FFP
ratios. Current recommendations indicate maintaining a PLT count above 100,000/
μl with an initial dose of 4–8 platelet concentrates (Spahn 2013). However, there is
18 Perioperative Hemostasis in Trauma 321
Fibrinolytic processes are an essential part of ATC and HF has a mortality rate of
close to 80 % (Brohi et al. 2008; Kashuk et al. 2010a, b; Schochl et al. 2011; Tauber
et al. 2011). Antifibrinolytic agents reduce blood loss in patients (with both normal
and exaggerated fibrinolytic response to surgery) by inhibiting plasminogen activa-
tion and therefore activation of fibrinolysis (Henry et al. 2011). The recent placebo-
controlled randomized CRASH-2 trial including over 20,000 trauma patients has
shown a significant reduction in all-cause mortality of approximately 10 % for
trauma patients receiving TXA versus placebo and to an approximately 30 % reduc-
tion in transfusion requirements. Benefit was maximal when administration was
early – within 3 h of injury (CRASH-2 trail Collaborators et al. 2010). This might
be due to high levels of free tPA detected early after a traumatic event, resulting in
enhanced plasmin generation and therefore increased fibrin degradation products
(Brohi et al. 2007a, b, 2008; Rugeri et al. 2007; Levrat et al. 2008). However, admin-
istration later than 3 h after injury was associated with worsened outcome (CRASH-2
trail Collaborators et al. 2010). The risk for thromboembolic complications appeared
to be relatively low. The use of antifibrinolytic drugs, such as TXA, is therefore
recommended in patients suffering from, or at risk of, hemorrhagic shock
(Association of Anaesthetists of Great et al. 2010).
18.5.5 Fibrinogen
Fibrinogen is an endogenous acute phase protein and an essential substrate for clot
formation. Activated by thrombin, fibrinogen forms insoluble fibrin strands and
functions as a ligand for platelet aggregation. After major trauma, fibrinogen is one
of the first coagulant proteins to reach critically low concentrations, and hypofibrino-
genemia has been associated with poor outcome (Harrigan et al. 1989; Hiippala et al.
1995; Rourke et al. 2012). Administration of crystalloids and colloids leads to dilu-
tion, functional deficiency, and abnormal fibrinogen polymerization. Furthermore,
hypothermia increases the breakdown of fibrinogen as its production is impaired in
acidemia. The use of fibrinogen concentrate results in reduced perioperative bleeding
and transfusion requirements (Rahe-Meyer et al. 2009), improves clot strength in
dilutional coagulopathy (Haas et al. 2008a, b), and shows that higher fibrinogen to
PRBC ratios have been associated with improved survival (Stinger et al. 2008).
Recent data indicate that coagulopathy induced by synthetic colloids may be reversed
by fibrinogen administration (Fenger-Eriksen et al. 2009). Despite the lack of high
quality RCT data supporting the benefit of fibrinogen supplementation, European
322 C. Heim and K. Brohi
18.5.6 Calcium
Factor XIII (FXIII) is fundamental to clot firmness because it binds to platelets via
their GP IIb/IIIa receptor. Furthermore, FXIII increases clot resistance against fibri-
nolysis by cross-linking to fibrin. In combination with functional platelets, it appears
to have inhibitory effects on plasmin-mediated HF (Dirkmann et al. 2012). Trauma
and major hemorrhage are known causes of acquired FXIII deficiency (Egbring
et al. 1996). Its substitution has been shown to improve clot firmness and might
therefore contribute to reduced bleeding (Nielsen et al. 2004). It has also been
shown to have some compensatory effects in settings with low fibrinogen levels
(Theusinger et al. 2010). A clinical combination suggesting the need for substitu-
tion of FXIII might be weak clot strength despite adequate fibrinogen levels. A clear
role for FXIII administration to bleeding trauma patients, however, has yet to be
defined.
18 Perioperative Hemostasis in Trauma 323
Clinical pathways in healthcare tend to improve the quality of the delivered care
(Rotter et al. 2010). Timely provision of blood products to a massively bleeding
patient is a challenge for any institution. The concept of hemostatic resuscitation
calls for the immediate readiness of components so as to enable their delivery in an
empirically agreed ratio on site. The implementation of massive transfusion proto-
cols (MTP) has been shown to facilitate this process (Cotton et al. 2009; Young
et al. 2011). Based on a mathematical model suggesting that a 1:1:1 ratio of PRBC
to FFP to PLT will come close to the composition of whole blood, formula-driven
resuscitation strategies have been advocated as the structural base for MTP (Hess
et al. 2008). Retrospective studies suggesting that early transfusion of FFP at a
fixed, high ratio will improve survival rates support the formula-driven approach
(Borgman et al. 2007; Cotton et al. 2008; Gunter et al. 2008; Maegele et al. 2008;
Spinella et al. 2008; Zink et al. 2009). However, in a study correcting for survivor
bias, no similar reduction in mortality was seen (Snyder et al. 2009). Regarding the
incidence of morbidity, there seems to be a controversy in the literature as to whether
formula-driven care increases or decreases the risk of multiple organ failure and
other complications in the trauma population (Maegele et al. 2008; Cotton et al.
2009). Furthermore, empirical administration of blood products can lead to over-
transfusion and has been shown to result in delayed correction of ATC and subopti-
mal use of blood products (Hess and Hiippala 2005; Gonzalez et al. 2007). Lastly,
identification of patients in need of this formula-based approach may be difficult as
there is no evidence-based strategy allowing for adequate triggers of a MTP. To date,
the vast majority of studies on formula-driven resuscitation focus essentially on
finding the optimal ratio of empirical blood-product use. However, whether the
concept of formula-driven resuscitation improves coagulation parameters and
patient outcomes has not been reliably addressed yet.
324 C. Heim and K. Brohi
Box 18.1
Three-strategy approach to transfusion in trauma patients at risk/with verified
massive hemorrhage (Adapted from Dzik et al. (2011))
1. Early administration of tranexamic acid (1 g over 10 min followed by an
infusion of 1 g over 8 h)
2. If critical bleeding, immediate administration of blood components (RBC
and FFP) in a fixed ratio
3. Adjustments of the fixed ratio of transfusion support based on clinical
course and results of goal-directed blood therapy
Conclusion
Coagulopathy after traumatic injury is incompletely understood and has a poor
prognosis. Early identification and proactive management is crucial for optimal
patient outcome. The concept of hemostatic resuscitation has become a pivotal
part of major trauma management. However, solid evidence for optimal transfu-
sion strategies is lacking. In the massively bleeding patient, a readily available
up-front hemostatic support with a predefined foundation ratio of blood compo-
nents seems adequate as first-line approach. This standardized initial step will
ideally be followed by an individualized goal-directed therapy, guided by the
clinical course and point-of-care testing.
Whichever strategy is chosen – the formula-driven or the individually tailored
approach – the 4 pillars of damage control resuscitation remain the major deter-
minants in achieving the best possible outcome:
• Early hemorrhage control
• Permissive hypotension
• Limited fluids
• Rapid reversal of coagulopathy
18 Perioperative Hemostasis in Trauma 325
References
Afshari A, Wikkelso A et al (2011) Thrombelastography (TEG) or thromboelastometry (ROTEM)
to monitor haemotherapy versus usual care in patients with massive transfusion. Cochrane
Database Syst Rev (3):CD007871
Armand R, Hess JR (2003) Treating coagulopathy in trauma patients. Transfus Med Rev
17(3):223–231
Association of Anaesthetists of Great, Ireland B et al (2010) Blood transfusion and the anaesthe-
tist: management of massive haemorrhage. Anaesthesia 65(11):1153–1161
Bickell WH, Wall MJ Jr et al (1994) Immediate versus delayed fluid resuscitation for hypotensive
patients with penetrating torso injuries. N Engl J Med 331(17):1105–1109
Boffard KD, Riou B et al (2005) Recombinant factor VIIa as adjunctive therapy for bleeding
control in severely injured trauma patients: two parallel randomized, placebo-controlled,
double-blind clinical trials. J Trauma 59(1):8–15; discussion 15–18
Bolliger D, Szlam F et al (2009) Finding the optimal concentration range for fibrinogen replace-
ment after severe haemodilution: an in vitro model. Br J Anaesth 102(6):793–799
Borgman MA, Spinella PC et al (2007) The ratio of blood products transfused affects mortality in
patients receiving massive transfusions at a combat support hospital. J Trauma 63(4):805–813
Brohi K, Singh J et al (2003) Acute traumatic coagulopathy. J Trauma 54(6):1127–1130
Brohi K, Cohen MJ et al (2007a) Acute coagulopathy of trauma: mechanism, identification and
effect. Curr Opin Crit Care 13(6):680–685
Brohi K, Cohen MJ et al (2007b) Acute traumatic coagulopathy: initiated by hypoperfusion: mod-
ulated through the protein C pathway? Ann Surg 245(5):812–818
Brohi K, Cohen MJ et al (2008) Acute coagulopathy of trauma: hypoperfusion induces systemic
anticoagulation and hyperfibrinolysis. J Trauma 64(5):1211–1217; discussion 1217
Brown LM, Call MS et al (2011) A normal platelet count may not be enough: the impact of admis-
sion platelet count on mortality and transfusion in severely injured trauma patients. J Trauma
71(2 Suppl 3):S337–S342
Burris D, Rhee P et al (1999) Controlled resuscitation for uncontrolled hemorrhagic shock.
J Trauma 46(2):216–223
Callum JL, Nascimento B et al (2009) Editorial: “formula-driven” versus “lab-driven” massive
transfusion protocols: at a state of clinical equipoise. Transfus Med Rev 23(4):247–254
Chakroun T, Gerotziafas GT et al (2006) The influence of fibrin polymerization and platelet-
mediated contractile forces on citrated whole blood thromboelastography profile. Thromb
Haemost 95(5):822–828
Cohen MJ, Call M et al (2012) Critical role of activated protein C in early coagulopathy and later
organ failure, infection and death in trauma patients. Ann Surg 255(2):379–385
CRASH-2 trail Collaborators, Shakur H et al (2010) Effects of tranexamic acid on death, vascular
occlusive events, and blood transfusion in trauma patients with significant haemorrhage
(CRASH-2): a randomised, placebo-controlled trial. Lancet 376(9734):23–32
Cothren CC, Moore EE et al (2007) Epidemiology of urban trauma deaths: a comprehensive
reassessment 10 years later. World J Surg 31(7):1507–1511
Cotton BA, Gunter OL et al (2008) Damage control hematology: the impact of a trauma exsan-
guination protocol on survival and blood product utilization. J Trauma 64(5):1177–1182;
discussion 1182–1183
Cotton BA, Au BK et al (2009) Predefined massive transfusion protocols are associated with a reduc-
tion in organ failure and postinjury complications. J Trauma 66(1):41–48; discussion 48–49
Cotton BA, Reddy N et al (2011) Damage control resuscitation is associated with a reduction in
resuscitation volumes and improvement in survival in 390 damage control laparotomy patients.
Ann Surg 254(4):598–605
Cotton BA, Harvin JA et al (2012) Hyperfibrinolysis at admission is an uncommon but highly
lethal event associated with shock and prehospital fluid administration. J Trauma Acute Care
Surg 73(2):365–370; discussion 370
326 C. Heim and K. Brohi
Curry NS, Davenport RA et al (2012) Transfusion strategies for traumatic coagulopathy. Blood
Rev 26(5):223–232
Darlington DN, Kheirabadi BS et al (2011) Coagulation changes to systemic acidosis and bicar-
bonate correction in swine. J Trauma 71(5):1271–1277
Davenport R, Khan S (2011) Management of major trauma haemorrhage: treatment priorities and
controversies. Br J Haematol 155(5):537–548
Davenport R, Manson J et al (2011) Functional definition and characterization of acute traumatic
coagulopathy. Crit Care Med 39(12):2652–2658
Demetriades D, Murray J et al (2004) Trauma fatalities: time and location of hospital deaths. J Am
Coll Surg 198(1):20–26
Dirkmann D, Gorlinger K et al (2012) Factor XIII and tranexamic acid but not recombinant factor
VIIa attenuate tissue plasminogen activator-induced hyperfibrinolysis in human whole blood.
Anesth Analg 114(6):1182–1188
Djaldetti M, Fishman P et al (1979) pH-induced platelet ultrastructural alterations. A possible
mechanism for impaired platelet aggregation. Arch Surg 114(6):707–710
Duchesne JC, McSwain NE Jr et al (2010) Damage control resuscitation: the new face of damage
control. J Trauma 69(4):976–990
Dutton RP (2012) Resuscitative strategies to maintain homeostasis during damage control surgery.
Br J Surg 99(Suppl 1):21–28
Dutton RP, McCunn M et al (2004) Factor VIIa for correction of traumatic coagulopathy. J Trauma
57(4):709–718; discussion 718–719
Dzik WH, Blajchman MA et al (2011) Clinical review: Canadian National Advisory Committee
on Blood and Blood Products–Massive transfusion consensus conference 2011: report of the
panel. Crit Care 15(6):242
Eastridge BJ, Malone D et al (2006) Early predictors of transfusion and mortality after injury:
a review of the data-based literature. J Trauma 60(6 Suppl):S20–S25
Egbring R, Kroniger A et al (1996) Factor XIII deficiency: pathogenic mechanisms and clinical
significance. Semin Thromb Hemost 22(5):419–425
Esmon CT (2005) The interactions between inflammation and coagulation. Br J Haematol
131(4):417–430
Eugster M, Reinhart WH (2005) The influence of the haematocrit on primary haemostasis in vitro.
Thromb Haemost 94(6):1213–1218
Faller DV (1999) Endothelial cell responses to hypoxic stress. Clin Exp Pharmacol Physiol
26(1):74–84
Fenger-Eriksen C, Jensen TM et al (2009) Fibrinogen substitution improves whole blood clot firm-
ness after dilution with hydroxyethyl starch in bleeding patients undergoing radical cystectomy:
a randomized, placebo-controlled clinical trial. J Thromb Haemost 7(5):795–802
Floccard B, Rugeri L et al (2012) Early coagulopathy in trauma patients: an on-scene and hospital
admission study. Injury 43(1):26–32
Flower O, Smith M (2011) The acute management of intracerebral hemorrhage. Curr Opin Crit
Care 17(2):106–114
Frith D, Goslings JC et al (2010) Definition and drivers of acute traumatic coagulopathy: clinical
and experimental investigations. J Thromb Haemost 8(9):1919–1925
Furie B, Furie BC (2008) Mechanisms of thrombus formation. N Engl J Med 359(9):938–949
Godier A, Samama CM et al (2012) Plasma/platelets/red blood cell ratio in the management of the
bleeding traumatized patient: does it matter? Curr Opin Anaesthesiol 25(2):242–247
Gonzalez EA, Moore FA et al (2007) Fresh frozen plasma should be given earlier to patients
requiring massive transfusion. J Trauma 62(1):112–119
Gruen RL, Jurkovich GJ et al (2006) Patterns of errors contributing to trauma mortality: lessons
learned from 2,594 deaths. Ann Surg 244(3):371–380
Gruen RL, Brohi K et al (2012) Haemorrhage control in severely injured patients. Lancet
380(9847):1099–1108
Gunter OL Jr, Au BK et al (2008) Optimizing outcomes in damage control resuscitation: identify-
ing blood product ratios associated with improved survival. J Trauma 65(3):527–534
18 Perioperative Hemostasis in Trauma 327
Kashuk JL, Moore EE et al (2010b) Primary fibrinolysis is integral in the pathogenesis of the acute
coagulopathy of trauma. Ann Surg 252(3):434–442; discussion 443–444
Kauvar DS, Wade CE (2005) The epidemiology and modern management of traumatic hemor-
rhage: US and international perspectives. Crit Care 9(Suppl 5):S1–S9
Kauvar DS, Lefering R et al (2006) Impact of hemorrhage on trauma outcome: an overview of
epidemiology, clinical presentations, and therapeutic considerations. J Trauma 60(6
Suppl):S3–S11
Kermode JC, Zheng Q et al (1999) Marked temperature dependence of the platelet calcium signal
induced by human von Willebrand factor. Blood 94(1):199–207
Krug EG, Sharma GK et al (2000) The global burden of injuries. Am J Public Health
90(4):523–526
Levrat A, Gros A et al (2008) Evaluation of rotation thrombelastography for the diagnosis of
hyperfibrinolysis in trauma patients. Br J Anaesth 100(6):792–797
Li T, Zhu Y et al (2011) Ideal permissive hypotension to resuscitate uncontrolled hemorrhagic
shock and the tolerance time in rats. Anesthesiology 114(1):111–119
Lier H, Krep H et al (2008) Preconditions of hemostasis in trauma: a review. The influence of
acidosis, hypocalcemia, anemia, and hypothermia on functional hemostasis in trauma. J
Trauma 65(4):951–960
MacLeod JB, Lynn M et al (2003) Early coagulopathy predicts mortality in trauma. J Trauma
55(1):39–44
Maegele M, Lefering R et al (2007) Early coagulopathy in multiple injury: an analysis from the
German Trauma Registry on 8724 patients. Injury 38(3):298–304
Maegele M, Lefering R et al (2008) Red-blood-cell to plasma ratios transfused during massive trans-
fusion are associated with mortality in severe multiple injury: a retrospective analysis from the
Trauma Registry of the Deutsche Gesellschaft fur Unfallchirurgie. Vox Sang 95(2):112–119
Mann KG (1999) Biochemistry and physiology of blood coagulation. Thromb Haemost
82(2):165–174
Martini WZ (2009) Coagulopathy by hypothermia and acidosis: mechanisms of thrombin genera-
tion and fibrinogen availability. J Trauma 67(1):202–208; discussion 208–209
Martini WZ (2011) Fibrinogen availability and coagulation function after hemorrhage and resus-
citation in pigs. Mol Med 17(7–8):757–761
Martini WZ, Holcomb JB (2007) Acidosis and coagulopathy: the differential effects on fibrinogen
synthesis and breakdown in pigs. Ann Surg 246(5):831–835
Martini WZ, Dubick MA et al (2006) Does bicarbonate correct coagulation function impaired by
acidosis in swine? J Trauma 61(1):99–106
Martinowitz U, Kenet G et al (2001) Recombinant activated factor VII for adjunctive hemorrhage
control in trauma. J Trauma 51(3):431–438; discussion 438–439
McSwain N Jr, Barbeau J (2011) Potential use of prothrombin complex concentrate in trauma
resuscitation. J Trauma 70(5 Suppl):S53–S56
Meng ZH, Wolberg AS et al (2003) The effect of temperature and pH on the activity of factor
VIIa: implications for the efficacy of high-dose factor VIIa in hypothermic and acidotic
patients. J Trauma 55(5):886–891
Morrison CA, Carrick MM et al (2011) Hypotensive resuscitation strategy reduces transfusion
requirements and severe postoperative coagulopathy in trauma patients with hemorrhagic
shock: preliminary results of a randomized controlled trial. J Trauma 70(3):652–663
Murad MH, Stubbs JR et al (2010) The effect of plasma transfusion on morbidity and mortality: a
systematic review and meta-analysis. Transfusion 50(6):1370–1383
Murthi SB, Dutton RP et al (2008) Transfusion medicine in trauma patients. Expert Rev Hematol
1(1):99–109
Nielsen VG, Gurley WQ Jr et al (2004) The impact of factor XIII on coagulation kinetics and clot
strength determined by thrombelastography. Anesth Analg 99(1):120–123
Niles SE, McLaughlin DF et al (2008) Increased mortality associated with the early coagulopathy
of trauma in combat casualties. J Trauma 64(6):1459–1463; discussion 1463–1465
Patanwala AE, Acquisto NM et al (2011) Prothrombin complex concentrate for critical bleeding.
Ann Pharmacother 45(7–8):990–999
18 Perioperative Hemostasis in Trauma 329
Stern SA, Dronen SC et al (1995) Multiple resuscitation regimens in a near-fatal porcine aortic
injury hemorrhage model. Acad Emerg Med 2(2):89–97
Stinger HK, Spinella PC et al (2008) The ratio of fibrinogen to red cells transfused affects survival
in casualties receiving massive transfusions at an army combat support hospital. J Trauma 64(2
Suppl):S79–S85; discussion S85
Tauber H, Innerhofer P et al (2011) Prevalence and impact of abnormal ROTEM(R) assays in
severe blunt trauma: results of the ‘Diagnosis and Treatment of Trauma-Induced Coagulopathy
(DIA-TRE-TIC) study’. Br J Anaesth 107(3):378–387
Theusinger OM, Baulig W et al (2010) In vitro factor XIII supplementation increases clot firmness
in Rotation Thromboelastometry (ROTEM). Thromb Haemost 104(2):385–391
Thorsen K, Ringdal KG et al (2011) Clinical and cellular effects of hypothermia, acidosis and
coagulopathy in major injury. Br J Surg 98(7):894–907
Toulon P, Ozier Y et al (2009) Point-of-care versus central laboratory coagulation testing during
haemorrhagic surgery. A multicenter study. Thromb Haemost 101(2):394–401
Valeri CR, Khuri S et al (2007) Nonsurgical bleeding diathesis in anemic thrombocytopenic
patients: role of temperature, red blood cells, platelets, and plasma-clotting proteins.
Transfusion 47(4 Suppl):206S–248S
Van der Linden P, Ickx BE (2006) The effects of colloid solutions on hemostasis. Can J Anaesth
53(6 Suppl):S30–S39
Varga-Szabo D, Pleines I et al (2008) Cell adhesion mechanisms in platelets. Arterioscler Thromb
Vasc Biol 28(3):403–412
Vivien B, Langeron O et al (2005) Early hypocalcemia in severe trauma. Crit Care Med
33(9):1946–1952
Wafaisade A, Wutzler S et al (2010) Drivers of acute coagulopathy after severe trauma: a multi-
variate analysis of 1987 patients. Emerg Med J 27(12):934–939
Wohlauer MV, Moore EE et al (2012) Early platelet dysfunction: an unrecognized role in the acute
coagulopathy of trauma. J Am Coll Surg 214(5):739–746
Wolberg AS, Meng ZH et al (2004) A systematic evaluation of the effect of temperature on coagu-
lation enzyme activity and platelet function. J Trauma 56(6):1221–1228
Young PP, Cotton BA et al (2011) Massive transfusion protocols for patients with substantial hem-
orrhage. Transfus Med Rev 25(4):293–303
Yuan S, Ferrell C et al (2007) Comparing the prothrombin time INR versus the APTT to evaluate
the coagulopathy of acute trauma. Thromb Res 120(1):29–37
Zink KA, Sambasivan CN et al (2009) A high ratio of plasma and platelets to packed red blood
cells in the first 6 hours of massive transfusion improves outcomes in a large multicenter study.
Am J Surg 197(5):565–570; discussion 570
Perioperative Hemostasis
in Neurosurgery 19
Julien Picard, Pierre Bouzat, Gilles Francony,
Jean-François Payen, and Patrick Schoettker
surface of normal brain tissue cells, and their thromboplastin content increases after
brain trauma (Pathak et al. 2005; Goh et al. 2005). Thromboplastins can be expressed
on the surface of injured blood vessels in brain lesions. This expression can also
result from inflammatory or tumoral processes. Increased expression of tissue factor
has been reported in astrocytic tumors and is correlated to their degree of malig-
nancy (Guan et al. 2002).
Injured brains and certain tumors in particular have a direct effect on fibrinolysis
(Goh et al. 1997). Tissue plasminogen activator (tPA) is an initiator of fibrinolysis,
and increased levels of tPA are reported in brain tumors. Hyperfibrinolysis is also
seen in trauma-related bleeding. This cascade of mechanisms can result in an exag-
gerated response called disseminated intravascular coagulation (DIC).
Hyperfibrinolysis can sometimes result from factor XIII deficiency, as this factor
will catalyze fibrin cross-linking into stable polymers (Gerlach et al. 2002). Cases
of postoperative bleeding have been reported in such situations (Vrettou et al. 2010),
and some authors recommend screening factor XIII levels before neurosurgery
(Gerlach et al. 2002).
Brain injuries and brain tumors, as well as other neurosurgical procedures in general,
can have an impact on overall coagulation, leading to thrombotic or hemorrhagic
disorders (Seifman et al. 2011). This can be explained by an imbalance between
19 Perioperative Hemostasis in Neurosurgery 333
procoagulant and anticoagulant systems, both locally and systemically, and by the
expression of several substances in physiological or pathological cerebral tissues.
These substances include tissue plasminogen activator and tissue factor, the latter
known as the main initiator of coagulation processes in the cell-based model of coag-
ulation activation (Gerlach et al. 2009). Hemorrhagic coagulopathy from cerebral or
systemic causes raises a significant risk of secondary central nervous system injury
by the hemorrhagic progression of lesions. Thrombotic events, however, will only
affect global outcome.
19.1.2.1 Thrombosis
Hypercoagulability can be detected during neurosurgical procedures as soon as the
intraoperative period (Abrahams et al. 2002). Deep venous thrombosis and pulmo-
nary emboli are frequent complications in the neurosurgical setting. Risk factors
include intracranial surgery, age, active malignancy, and hemi/para-paresia or para-
plegia. Surgery for brain tumors and vertebro-medullary trauma is considered to be at
a high risk of thromboembolic complications. Symptomatic deep venous thrombosis
can affect up to 32 % of patients undergoing brain tumor neurosurgery (Agnelli
1999). The incidence of symptomatic and asymptomatic deep venous thrombosis
ranges from 4 to 34 % for all causes of intracranial surgery (Hamilton et al. 2011) and
from 1.5 to 18 % for subarachnoid hemorrhage (Vespa et al. 2011). The risk of throm-
bosis is heterogenous during spinal surgery: vertebro-medullary traumas show a high
incidence of symptomatic deep venous thrombosis (12–23 %) and asymptomatic
deep venous thrombosis (81 % diagnosed by phlebography), whereas the risks for
other surgical procedures are mild (osteosynthesis and extended laminectomy, 0.3–
2.7 %) or low (herniated disc or less than two levels of laminectomy, <1 %) (Audibert
et al. 2005). We have seen that thromboplastin release increases in traumatic brain
injury (TBI) and tumoral lesions, but procoagulant mediators are also promoted in
such situations. Significantly higher concentrations of plasma tissue factor pathway
inhibitor have been reported in patients with brain tumors (Gerlach et al. 2003).
19.1.2.2 Bleeding
Coagulopathy resulting from brain injury has been extensively studied. It includes
multiple mechanisms: release of cerebral tissue factor and activation/amplification
of coagulation, disseminated intravascular coagulation, hyperfibrinolysis, platelet
dysfunction, brain and global hypoperfusion, and activation of protein C.
These mechanisms are well documented in trauma settings (Laroche et al. 2012).
They can occur in isolation or in association with acute traumatic coagulopathy,
acidosis, and hypothermia. Coagulopathy due to TBI is statistically associated with
the severity of injury, progression of hemorrhagic lesions, and increased morbidity
and mortality. Mortality rises from 8 to 32 % in cases of hemorrhagic progression
lesions (Allard et al. 2009). It is, however, difficult to establish a strong unilateral
causal relationship between coagulopathy and the severity of brain injury: hemor-
rhagic progression of lesions seems to be partly predetermined by the primary injury
whatever the state of coagulation (Kurland et al. 2012), and the severity of the pri-
mary injury determines the severity of coagulopathy. This is probably a phenome-
non of autoamplification: the more severe the primary injury, the more severe the
334 J. Picard et al.
consecutive brain hypoperfusion and the worser the coagulopathy, affecting the evo-
lution of lesions and outcome. This could explain why it may not be possible to
completely reverse outcome even while normalizing coagulation. However, acquired
iatrogenic coagulopathy is a risk factor for bad outcomes. The incidence of TBI-
related coagulopathy ranges from 10 to 97 % depending on severity and definition.
Its mean incidence is 33 % (Harhangi et al. 2008). The time course of TBI coagu-
lopathy has been studied. It occurs in the first 5 days, mostly in the first 24 h (mean
23 ± 2 h post admission). Early onset (<12 h) has been associated with greater sever-
ity and poorer outcome (Lustenberger et al. 2010). It is important to note that thera-
peutic hypothermia does not seem to cause hemorrhagic progression of brain
contusions (Resnick et al. 1994). What is observed in brain trauma is also relevant
in neurosurgery, and some of the mechanisms described in TBI coagulopathy have
been documented in the perioperative period (Goh et al. 1997).
Planned surgery
Aspirin Clopidogrel
GP IIb/IIIa-antagonists
STOP medication ?
STOP medication ?
Yes No No Yes
Surgery
Delay surgery Delay surgery
Fig. 19.1 Management of antiplatelet agents in planned neurosurgery (Gerlach et al. 2009;
Beynon et al. 2012; Okano et al. 2014)
Emergency surgery
Aspirin Clopidogrel
GP IIb/IIIa-antagonists
Consider platelet
transfusion
Point-of-care whole-blood
platelet function analysis
Platelet dysfunction ?
Yes No
Consider platelet
Surgery
transfusion
Fig. 19.2 Management of antiplatelet agents in emergency neurosurgery (Beshay et al. 2010;
Batchelor and Grayson 2013; Beynon et al. 2013; Li et al. 2013)
19.1.3.8 Thrombolytics
Intravenous thrombolysis for myocardial infarction or pulmonary embolism shows
a quite low incidence of spontaneous ICH (less than 2 %) (Patel and Mody 1999).
However, the incidence is higher following thrombolysis for ischemic stroke: rates
between 2.4 and 10.7 % have been described following intravenous thrombolysis,
7.8–15.4 % following intra-arterial mechanical plus chemical thrombolysis, and
6.3–9.9 % following intra-arterial procedures following intravenous thrombolysis
(Mokin et al. 2012). This can be explained by the increased fragility of ischemic
brain tissue, particularly due to the early disruption of the blood-brain barrier.
In contrast, modern thrombolytic agents (alteplase) have very short half-lives, and
there is usually no use in providing antagonization. Association with other medica-
tions which interfere with hemostasis and coagulation seems, of course, to increase
the risk of bleeding.
Spinal complications are rare after thrombolytic therapy and usually present as
epidural hematoma (Clark and Paradis 2002).
The anatomical specificities of the central nervous system can lead to diagnostic and
therapeutic difficulties, as even a small volume of blood can have dramatic conse-
quences, and the diagnosis of bleeding can be delayed because of a lack of obvious
hemorrhagic exteriorization. Sometimes, the first sign of a bleeding disorder will be
a neurological deterioration in the patient. When available, the patient’s medical
history will be invaluable. Preexisting hemostatic defects can be detected at a pre-
anesthetic consultation, and some authors have proposed standardized question-
naires to evaluate coagulation on admission (Koscielny et al. 2004). In some
situations, hemostatic perturbations can be discovered during surgery. This is par-
ticularly true in emergency situations, with comatose or sedated patients, or with
unknown coagulation diseases. Thus, the surgeon’s appreciation of potential bleed-
ing and hemostasis constitutes precious information that should not be neglected.
340 J. Picard et al.
Not enough attention is paid to the complex pathophysiology and kinetics of acute
coagulopathy: this is partly due to the limitations with usual hemostasis assays. In
addition to the routine coagulation parameters, viscoelastic assays (ROTEM®/
TEG®) can give an overall understanding of the coagulation status.
Thromboelastometry/thromboelastography appears to be useful in the treatment of
bleeding trauma patients and provides information about the speed and quality of
clot formation (Spahn et al. 2013). They are performed in whole blood and thus
provide clinically relevant data. Thromboelastometry assays are increasingly used
to guide transfusion strategy; however, data on patients with isolated brain injury
are lacking. A recent publication detailed thromboelastometric patterns of isolated
brain-injured patients, but there are no prospective data on the subject (Schöchl
et al. 2011).
19 Perioperative Hemostasis in Neurosurgery 341
D1 = admission
Severe TBI
FIBTEM S 2010-06-19 01:29 2: APRES FIBRI EXTEM S 2010-06-19 01:25 2: APRES FIBRI
Fig. 19.3 Thromboelastometric (ROTEM) profile of a patient with severely traumatic brain injury
on day 1 (hemorrhagic complication: extensive hemorrhagic brain contusions)
Day 4
Pulmonary
embolism
FIBTEM S 2010-06-22 14:32 2: EMBOLIE PULM EXTEM S 2010-06-22 14:29 2: EMBOLIE PULM
Fig. 19.4 Thromboelastometric (ROTEM) profile of a patient with severely traumatic brain injury
on day 4 (thromboembolic evolution: bilateral pulmonary emboli)
19.3.1.1 Platelets
Thrombocytopenia is directly associated to mortality and adverse outcome in traumatic
intracranial hemorrhage patients (Allard et al. 2009). General recommendations are to
maintain a platelet level greater than 100,000/μL, but a platelet count below 175,000/
19 Perioperative Hemostasis in Neurosurgery 343
19.3.1.2 Fibrinogen
European guidelines on the management of bleeding following trauma recom-
mend treatment with fibrinogen concentrate or cryoprecipitate if significant bleed-
ing is accompanied by a plasma fibrinogen level below 1.5–2.0 g/L. The
development of thromboelastometric assays has focused attention on the role of
fibrinogen. It plays a central role in the coagulation process and the literature sug-
gests that its administration could reduce blood loss and requirements for blood-
derived products in both cardiovascular surgery and trauma (Karlsson et al. 2008;
Fenger-Eriksen et al. 2008).
Nevertheless, good-quality multicentered randomized trials are lacking, and it is
currently not known whether the administration of high doses of fibrinogen is asso-
ciated with an increased thromboembolic risk.
19.3.2.3 Deferoxamine
Iron can induce brain damage. Deferoxamine – an iron chelator – has been shown
to reduce hemoglobin-induced edema in a rat model (Nakamura et al. 2004). It has
also been shown to reduce brain atrophy and improve neurological function after
intracranial hemorrhage in the rat (Okauchi et al. 2010). There are nevertheless no
clinical studies published.
Bleeding remains the most serious threat to neurosurgical patients, but the problem
is certainly more complex when the early and massive activation of coagulation
processes leads rapidly to a hypercoagulable state. Abrahams et al. (2002) studied
the evolution of coagulability during neurosurgical procedures and found increased
coagulability from the induction of anesthesia and skin incision through to intraop-
erative and postoperative periods (Abrahams et al. 2002). Interestingly, these
changes were more pronounced in patients undergoing craniotomy than in patients
undergoing spinal procedures.
Thromboembolic complications are known to be more frequent in neurosurgi-
cal patients. Classic risk factors including immobility, malignancy, trauma, and a
delayed introduction of thromboprophylaxis certainly do play a role in the increased
number of thromboembolic events observed in these patients, but they are not the
only mechanisms. There is growing evidence that a hypercoagulable state can
develop perioperatively in neurosurgical pathologies and interventions. Goobie
et al. (2001) evaluated coagulation by using thromboelastography (TEG) data from
30 pediatric patients undergoing craniotomy; they found that peri- and postoperative
hypercoagulable TEG profiles showed shortened coagulation times and increased
maximal amplitude of clot strength, compared to preoperative profiles.
Experimental investigations have shown that thrombin is responsible for
increased brain edema and neuronal death, and the intracerebral injection of throm-
bin inhibitors, such as argatroban, has been shown to significantly reduce edema in
a rat model of ICH (Kitaoka et al. 2002); thus, supporting the concept that clotting
and bleeding states are closely linked.
References
Aberg M, Nilsson IM, Vilhardt H (1979) The release of fibrinolytic activator and factor VIII after
injection of DDAVP. In: Davidsson JF (ed) Progress in chemical fibrinolysis and thrombolysis.
Churchill Livingstone, Edinburgh, pp 92–97
Abrahams JM, Torchia MB, McGarvey M, Putt M, Baranov D, Sinson GP (2002) Perioperative
assessment of coagulability in neurosurgical patients using thromboelastography. Surg Neurol
58:5–11
Agnelli G (1999) Prevention of venous thromboembolism after neurosurgery. Thromb Haemost
82:925–930
Allard CB, Scarpelini S, Rhind SG, Baker AJ, Shek PN, Tien H, Fernando M, Tremblay L,
Morrison LJ, Pinto R, Rizoli SB (2009) Abnormal coagulation tests are associated with pro-
gression of traumatic intracranial hemorrhage. J Trauma 67:959–967
Audibert G, Faillot T, Vergnes MC, Bosson JL, Bernard C, Payen JF, Lestienne B, Bruder N (2005)
Thromboprophylaxis in elective spinal surgery and spinal cord injury. Ann Fr Anesth Reanim
24:928–934
Batchelor JS, Grayson A (2013) A meta-analysis to determine the effect of preinjury antiplatelet
agents on mortality in patients with blunt head trauma. Br J Neurosurg 27:12–18
Beshay JE, Morgan H, Madden C, Yu W, Sarode R (2010) Emergency reversal of anticoagulation
and antiplatelet therapies in neurosurgical patients. J Neurosurg 112:307–318
Beynon C, Hertle DN, Unterberg AW, Sakowitz OW (2012) Clinical review: traumatic brain injury
in patients receiving antiplatelet medication. Crit Care 16:228
Beynon C, Scherer M, Jakobs M, Jung C, Sakowitz OW, Unterberg AW (2013) Initial experiences
with Multiplate® for rapid assessment of antiplatelet agent activity in neurosurgical emergen-
cies. Clin Neurol Neurosurg 115:2003–2008
Brummel-Ziedins K, Whelihan MF, Ziedins EG, Mann KG (2006) The resuscitative fluid you
choose may potentiate bleeding. J Trauma 61:1350–1358
Chan KH, Mann KS, Chan TK (1989) The significance of thrombocytopenia in the development
of postoperative intracranial hematoma. J Neurosurg 71:38–41
Clark MA, Paradis NA (2002) Spinal epidural hematoma complicating thrombolytic therapy with
tissue plasminogen activator–a case report. J Emerg Med 23:247–251
Cravens GT, Brown MJ, Brown DR, Wass CT (2006) Antifibrinolytic therapy use to mitigate blood
loss during staged complex major spine surgery: postoperative visual color changes after
tranexamic acid administration. Anesthesiology 105:1274–1276
Cucchiara B, Messe S, Sansing L, Kasner S, Lyden P, CHANT Investigators (2008) Hematoma
growth in oral anticoagulant related intracerebral hemorrhage. Stroke 39:2993–2996
Dai Y, Lee A, Critchley LA, White PF (2009) Does thromboelastography predict postoperative
thromboembolic events? A systematic review of the literature. Anesth Analg 108:734–742
Fenger-Eriksen C, Lindberg-Larsen M, Christensen AQ, Ingerslev J, Sørensen B (2008) Fibrinogen
concentrate substitution therapy in patients with massive haemorrhage and low plasma fibrino-
gen concentrations. Br J Anaesth 101:769–773
Finsterer J, Pelzl G, Hess B (2001) Severe, isolated thrombocytopenia under polytherapy with
carbamazepine and valproate. Psychiatry Clin Neurosci 55:423–426
Gattas DJ, Dan A, Myburgh J, Billot L, Lo S, Finfer S, CHEST Management Committee (2012)
Fluid resuscitation with 6 % hydroxyethyl starch (130/0.4) in acutely ill patients: an updated
systematic review and meta-analysis. Anesth Analg 114:159–169
Gerlach R, Tölle F, Raabe A, Zimmermann M, Siegemund A, Seifert V (2002) Increased risk for
postoperative hemorrhage after intracranial surgery in patients with decreased factor XIII activity:
implications of a prospective study. Stroke 33:1618–1623
Gerlach R, Scheuer T, Böhm M, Beck J, Woszczyk A, Raabe A, Scharrer I, Seifert V (2003)
Increased levels of plasma tissue factor pathway inhibitor in patients with glioblastoma and
intracerebral metastases. Neurol Res 25:335–338
Gerlach R, Krause M, Seifert V, Goerlinger K (2009) Hemostatic and hemorrhagic problems in
neurosurgical patients. Acta Neurochir 151:873–900
19 Perioperative Hemostasis in Neurosurgery 347
Gerstner T, Teich M, Bell N, Longin E, Dempfle CE, Brand J, König S (2006) Valproate-associated
coagulopathies are frequent and variable in children. Epilepsia 47:1136–1143
Goh KY, Tsoi WC, Feng CS, Wickham N, Poon WS (1997) Haemostatic changes during surgery
for primary brain tumours. J Neurol Neurosurg Psychiatry 63:334–338
Goh KY, Poon WS, Chan DT, Ip CP (2005) Tissue plasminogen activator expression in meningio-
mas and glioblastomas. Clin Neurol Neurosurg 107:296–300
Goobie SM, Soriano SG, Zurakowski D, McGowan FX, Rockoff MA (2001) Hemostatic changes in
pediatric neurosurgical patients as evaluated by thrombelastograph. Anesth Analg 93:887–892
Gorelick PB, Weisman SM (2005) Risk of hemorrhagic stroke with aspirin use: an update. Stroke
36:1801–1807
Görlinger K (2006) Coagulation management during liver transplantation. Hamostaseologie
26:S64–S76
Grounds M (2003) Recombinant factor VIIa (rFVIIa) and its use in severe bleeding in surgery and
trauma: a review. Blood Rev 17(Suppl 1):S11–S21
Guan M, Su B, Lu Y (2002) Quantitative reverse transcription-PCR measurement of tissue factor
mRNA in glioma. Mol Biotechnol 20:123–129
Hamilton MG, Yee WH, Hull RD, Ghali WA (2011) Venous thromboembolism prophylaxis in
patients undergoing cranial neurosurgery: a systematic review and meta-analysis. Neurosurgery
68:571–581
Harhangi BS, Kompanje EJ, Leebeek FW, Maas AI (2008) Coagulation disorders after traumatic
brain injury. Acta Neurochir (Wien) 150:165–175
Henry DA, Moxey AJ, Carless PA, O’Connell D, McClelland B, Henderson KM, Sly K, Laupacis
A, Fergusson D (2007) Anti-fibrinolytic use for minimising perioperative allogeneic blood
transfusion. Cochrane Database Syst Rev (1):CD001886
Holtzer CD, Reisner-Keller LA (1997) Phenytoin-induced thrombocytopenia. Ann Pharmacother
31:435–437
Huang FP, Xi G, Keep RF, Hua Y, Nemoianu A, Hoff JT (2002) Brain edema after experimental
intracerebral hemorrhage: role of hemoglobin degradation products. J Neurosurg 96:287–293
Karlsson M, Ternström L, Hyllner M, Baghaei F, Nilsson S, Jeppsson A (2008) Plasma fibrinogen
level, bleeding, and transfusion after on-pump coronary artery bypass grafting surgery: a pro-
spective observational study. Transfusion 48:2152–2158
Kashuk JL, Moore EE, Sabel A, Barnett C, Haenel J, Le T, Pezold M, Lawrence J, Biffl WL,
Cothren CC, Johnson JL (2009) Rapid thrombelastography (r-TEG) identifies hypercoagula-
bility and predicts thromboembolic events in surgical patients. Surgery 146:764–772
Kitaoka T, Hua Y, Xi G, Hoff JT, Keep RF (2002) Delayed argatroban treatment reduces edema in
a rat model of intracerebral hemorrhage. Stroke 33:3012–3018
Koscielny J, Ziemer S, Radtke H, Schmutzler M, Pruss A, Sinha P, Salama A, Kiesewetter H, Latza
R (2004) A practical concept for preoperative identification of patients with impaired primary
hemostasis. Clin Appl Thromb Hemost 10:195–204
Kottke-Marchant K, Powers JB, Brooks L, Kundu S, Christie DJ (1999) The effect of antiplatelet
drugs, heparin, and preanalytical variables on platelet function detected by the platelet function
analyzer (PFA-100). Clin Appl Thromb Hemost 5:122–130
Kozek-Langenecker SA (2005) Effects of hydroxyethyl starch solutions on hemostasis.
Anesthesiology 103:654–660
Kunio NR, Differding JA, Watson KM, Stucke RS, Schreiber MA (2012) Thrombelastography-
identified coagulopathy is associated with increased morbidity and mortality after traumatic
brain injury. Am J Surg 203:584–588
Kurland D, Hong C, Aarabi B, Gerzanich V, Simard JM (2012) Hemorrhagic progression of a
contusion after traumatic brain injury: a review. J Neurotrauma 29:19–31
Lackmann GM (2004) Valproic-acid-induced thrombocytopenia and hepatotoxicity: discontinua-
tion of treatment? Pharmacology 70:57–58
Laroche M, Kutcher ME, Huang MC, Cohen MJ, Manley GT (2012) Coagulopathy after traumatic
brain injury. Neurosurgery 70:1334–1345
Lavoie A, Ratte S, Clas D, Demers J, Moore L, Martin M, Bergeron E (2004) Preinjury warfarin
use among elderly patients with closed head injuries in a trauma center. J Trauma 56:802–807
348 J. Picard et al.
Lee KR, Kawai N, Kim S, Sagher O, Hoff JT (1997) Mechanisms of edema formation after intra-
cerebral hemorrhage: effects of thrombin on cerebral blood flow, blood-brain barrier permea-
bility, and cell survival in a rat model. J Neurosurg 86:272–278
Lethagen S (1994) Desmopressin (DDAVP) and hemostasis. Ann Hematol 69:173–180
Li X, Sun Z, Zhao W, Zhang J, Chen J, Li Y, Ye Y, Zhao J, Yang X, Xiang Y, Li G, Mao J, Zhang
W, Zhang M, Zhang W (2013) Effect of acetylsalicylic acid usage and platelet transfusion on
postoperative hemorrhage and activities of daily living in patients with acute intracerebral hem-
orrhage. J Neurosurg 118:94–103
Lin J, Hanigan WC, Tarantino M, Wang J (2003) The use of recombinant activated factor VII to
reverse warfarin-induced anticoagulation in patients with hemorrhages in the central nervous
system: preliminary findings. J Neurosurg 98:737–740
Luostarinen T, Niiya T, Shramko A, Rosenberg P, Niemi T (2011) Comparison of hypertonic saline
and mannitol on whole blood coagulation in vitro assessed by thromboelastometry. Neurocrit
Care 14:238–243
Luostarinen T, Silvasti-Lundell M, Medeiros T, Romani R, Hernesniemi J, Niemi T (2012)
Thromboelastometry during intraoperative transfusion of fresh frozen plasma in pediatric neu-
rosurgery. J Anesth(May 6)
Lustenberger T, Talving P, Kobayashi L, Inaba K, Lam L, Plurad D, Demetriades D (2010) Time
course of coagulopathy in isolated severe traumatic brain injury. Injury 41:924–928
Mannucci PM, Ruggeri ZM, Pareti FI, Capitanio A (1977) 1-Deamino-8-d-arginine vasopressin: a
new pharmacological approach to the management of haemophilia and von Willebrand’s dis-
eases. Lancet 23:869–872
Manohar C, Avitsian R, Lozano S, Gonzalez-Martinez J, Cata JP (2011) The effect of antiepileptic
drugs on coagulation and bleeding in the perioperative period of epilepsy surgery: the Cleveland
Clinic experience. J Clin Neurosci 18:1180–1184
Mayer SA, Brun NC, Begtrup K, Broderick J, Davis S, Diringer MN, Skolnick BE, Steiner T,
Recombinant Activated Factor VII Intracerebral Hemorrhage Trial Investigators (2005)
Recombinant activated factor VII for acute intracerebral hemorrhage. N Engl J Med 352:777–785
Mittal MK, Rabinstein AA (2012) Anticoagulation-related intracranial hemorrhages. Curr
Atheroscler Rep 14:351–359
Mokin M, Kan P, Kass-Hout T, Abla AA, Dumont TM, Snyder KV, Hopkins LN, Siddiqui AH,
Levy EI (2012) Intracerebral hemorrhage secondary to intravenous and endovascular intraarte-
rial revascularization therapies in acute ischemic stroke: an update on risk factors, predictors,
and management. Neurosurg Focus 32(4):E2
Nakamura T, Keep RF, Hua Y, Schallert T, Hoff JT, Xi G (2004) Deferoxamine-induced attenu-
ation of brain edema and neurological deficits in a rat model of intracerebral hemorrhage.
J Neurosurg 100:672–678
Neff TA, Doelberg M, Jungheinrich C, Sauerland A, Spahn DR, Stocker R (2003) Repetitive large-
dose infusion of the novel hydroxyethyl starch 130/0.4 in patients with severe head injury.
Anesth Analg 96:1453–1459
Okano A, Oya S, Fujisawa N, Tsuchiya T, Indo M, Nakamura T, Chang HS, Matsui T (2014)
Analysis of risk factors for chronic subdural haematoma recurrence after burr hole surgery:
Optimal management of patients on antiplatelet therapy. Br J Neurosurg 28:204–208
Okauchi M, Hua Y, Keep RF, Morgenstern LB, Schallert T, Xi G (2010) Deferoxamine treatment
for intracerebral hemorrhage in aged rats: therapeutic time window and optimal duration.
Stroke 41:375–382
Okur M, Kaya A, Çaksen H, Taşkın G (2012) Lamotrigine-associated thrombocytopenia and leu-
kopenia. J Emerg Med 42:584–585
Pan CF, Shen MY, Wu CJ, Hsiao G, Chou DS, Sheu JR (2007) Inhibitory mechanisms of gabapen-
tin, an antiseizure drug, on platelet aggregation. J Pharm Pharmacol 59:1255–1261
Patel SC, Mody A (1999) Cerebral hemorrhagic complications of thrombolytic therapy. Prog
Cardiovasc Dis 42:217–233
Pathak A, Dutta S, Marwaha N, Singh D, Varma N, Mathuriya SN (2005) Change in tissue throm-
boplastin content of brain following trauma. Neurol India 53:178–182
19 Perioperative Hemostasis in Neurosurgery 349
Treib J, Haass A, Pindur G, Grauer MT, Treib W, Wenzel E, Schimrigk K (1996a) Influence of low
and medium molecular weight hydroxyethyl starch on platelets during a long-term hemodilu-
tion in patients with cerebrovascular diseases. Arzneimittelforschung 46:1064–1066
Treib J, Haass A, Pindur G, Grauer MT, Wenzel E, Schimrigk K (1996b) Decrease of fibronectin
following repeated infusion of highly substituted hydroxyethyl starch. Infusionsther
Transfusionsmed 23:71–75
Treib J, Haass A, Pindur G, Grauer MT, Jung F, Wenzel E, Schimrigk K (1997) Increased haemor-
rhagic risk after repeated infusion of highly substituted medium molecular weight hydroxyethyl
starch. Arzneimittelforschung 47:18–22
Vespa P, Participants in the International Multi-Disciplinary Consensus Conference on the Critical
Care Management of Subarachnoid Hemorrhage (2011) Deep venous thrombosis prophylaxis.
Neurocrit Care 15:295–297
Vrettou CS, Stavrinou LC, Halikias S, Kyriakopoulou M, Kollias S, Stranjalis G, Koutsoukou A
(2010) Factor XIII deficiency as a potential cause of supratentorial haemorrhage after posterior
fossa surgery. Acta Neurochir 152:529–532
Weber AA, Adamzik M, Bachmann HS, Görlinger K, Grandoch M, Leineweber K, Müller-
Beissenhirtz H, Wenzel F, Naber C (2008) Methods to evaluate the pharmacology of oral
antiplatelet drugs. Herz 33:287–296
Wu G, Xi G, Huang F (2006) Spontaneous intracerebral hemorrhage in humans: hematoma
enlargement, clot lysis, and brain edema. Acta Neurochir 96:78–80
Xi G, Wagner KR, Keep RF, Hua Y, de Courten-Myers GM, Broderick JP, Brott TG, Hoff JT
(1998) Role of blood clot formation on early edema development after experimental intracere-
bral hemorrhage. Stroke 29:2580–2586
Xi G, Hua Y, Bhasin RR, Ennis SR, Keep RF, Hoff JT (2001) Mechanisms of edema formation
after intracerebral hemorrhage: effects of extravasated red blood cells on blood flow and blood-
brain barrier integrity. Stroke 32:2932–2938
Xi G, Reiser G, Keep RF (2003) The role of thrombin and thrombin receptors in ischemic, hemor-
rhagic and traumatic brain injury: deleterious or protective? J Neurochem 84:3–9
Xi G, Keep RF, Hoff JT (2006) Mechanisms of brain injury after intracerebral haemorrhage.
Lancet Neurol 5:53–63
Bleeding Management in Elective
Orthopedic Surgery 20
Oliver M. Theusinger
20.1 Introduction
O.M. Theusinger, MD
Institute of Anesthesiology, University of Zurich, University Hospital Zurich,
Rämistarsse 100, Zurich 8091, Switzerland
e-mail: [email protected]
transfusion decisions is evident: blood that is not lost does not have to be replaced.
Preoperative anemia, which varies from 20 to 51 %, has been identified as another
major issue. The World Health Organization (WHO) has defined this as a Hb level
≤12 g/dL in women and ≤13 g/dL in men, which corresponds to a hematocrit (Hct)
of ≤36 % in women and ≤39 % in men (Rosencher et al. 2003; Theusinger et al.
2007; Kendoff et al. 2011). This has been shown to be an independent risk factor for
increased 30-day mortality and major morbidity in all surgical patients (Musallam
et al. 2011). In up to 30 % of patients, iron deficiency is the cause of anemia and
should be corrected at least 4 weeks prior to surgery in order to achieve optimal
results (Theusinger et al. 2007). Optimizing Hb mass before orthopedic surgery, as
well as limiting intraoperative blood loss, is associated with improved outcomes
after 90 days (Kotze et al. 2012).
The indications for RBC transfusions are controversial, and their liberal use versus
their more restricted use is largely discussed. Current literature tends to support a
more restrictive use, with Hb concentrations of 6–8 g/dL or awaiting physiological
triggers (Klein et al. 2007).
This method was a standard for many years but is barely used nowadays, as there is
a poor cost-benefit relationship for the patient. Patients donate one or more units of
their own blood preoperatively (4–6 weeks) and are allowed a recovery period of
4 weeks prior to surgery. The units are stored in a blood bank and then retransfused
preoperatively. Patients have been shown to become anemic due to their donation
(Vamvakas and Pineda 2000). Several studies have shown that pre-donation is no
longer recommended in elective orthopedic surgery (Tartter et al. 1986; Theusinger
et al. 2007) because such risks as transfusion reactions, circulatory overload, bacterial
contamination, clerical errors, and negative outcome rates have all been demon-
strated (Goldman et al. 1997).
The cost-effectiveness has also been discussed as more than 45 % of donated
blood is discarded and patients become anemic due to this technique (Goldman
et al. 1997; Habler and Messmer 1997; Franchini et al. 2008).
Anemia related to iron deficiency is the most common type of anemia among
patients scheduled for elective orthopedic surgery (prevalence of up to 30 %)
(Theusinger et al. 2007). Preoperative iron supplements have been proposed in
order to elevate Hb levels (Weiss and Goodnough 2005) as they can be substituted
orally and parenterally.
After significant blood loss, there is a fivefold increase of the erythropoietic
response following intravenous (i.v.) iron administration (Weiss and Goodnough
2005). This, in combination with erythropoietin (EPO), has been shown to be asso-
ciated with a decrease in mortality due to reduced transfusion and infection rates
(Cuenca et al. 2004, 2005, 2006).
Iron therapy was found to be generally safe and effective, especially high molecu-
lar weight iron. Ferric carboxymaltose (Ferinject®, Vifor Int., St. Gallen, Switzerland)
has been shown to be well tolerated, with no safety concerns (Kulnigg et al. 2008) for
dosages up to 1 g of i.v. iron administered every 15 min. Life-threatening anaphylactic
reactions have been described for solutions containing dextran (Chertow et al. 2006),
354 O.M. Theusinger
Patients undergoing surgery are getting older and are on a variety of medications. While
it was usual to discontinue anticoagulants such as vitamin K antagonists or aspirin sev-
eral days before surgery, discontinuing clopidogrel and aspirin in patients with unstable
20 Bleeding Management in Elective Orthopedic Surgery 355
coronary perfusion and/or recently implanted stents before elective orthopedic surgery
is strongly contraindicated (Spahn et al. 2006, 2007; Chassot et al. 2007a, b).
Recommendations promote aspirin as a lifelong therapy, whereas clopidogrel should be
used only as long as the coronary stents are not fully endothelialized. This usually takes
6–24 weeks depending on the technique used (Chassot et al. 2007a, b). The hemorrhagic
risk due to antiplatelet therapy is modest: 2.5–20 % more blood loss with aspirin and
30–50 % with the combination of aspirin and clopidogrel (Chassot et al. 2007a, b).
Nonsteroidal painkillers (NSAID) are widely used for pre- and postoperative
analgesia and might adversely affect hemostasis after the surgery. They should be
stopped at least 24 h prior to surgery. NSAR inhibit prostaglandin synthesis and
particularly block cyclooxygenase (COX), which is the central enzyme in the pro-
cess of prostaglandin formation. COX-2-selective NSAR have no undesirable COX-
1-related side effects such as impaired platelet aggregation and prolonged bleeding
time or consecutive increased blood loss during surgery. If NSAR are considered for
postoperative pain relief, COX-2-selective ones should be used (Slappendel et al.
2002; Weber et al. 2003). The costs of optimizing perioperative medication are
negligible, but close attention should be paid to this issue during the preoperative
surgical and anesthesiology consultations.
Body temperature during surgery affects platelet aggregation and bleeding time.
The pathophysiological effect of hypothermia on platelet activation/adhesion is due
to the inhibition of the interaction between von Willebrand factor and the platelet
glycoprotein Ib-IX-V complex. It also slows down the metabolic rate of coagulation
factor enzymes. A decrease of 1.5 °C is associated with increased blood loss of
about 50 % during total hip replacement (Schmied et al. 1996; Winkler et al. 2000).
Maintenance of physiological body temperature is therefore an obligation, and all
efforts should be taken to achieve this goal.
Okamoto and coworkers began searching for substances that inhibit the action of
plasmin during the 1950s. One of 200 lysin derivates studied was tranexamic acid
(trans-4-aminomethylcyclohexane-1-carboxylic acid, or TXA) (Okamoto et al.
1997). Its use has been shown to have significant beneficial blood-sparing effects
in elective surgery (Henry et al. 2007), while a dose of 1 g reduced bleeding and
blood loss for elective knee and hip arthroplasty (Aguilera et al. 2013; Delanois
and Mont 2013). Costs and side effects are low at this dose, suggesting that a
generalization of the use of antifibrinolytic agents in orthopedic surgery might be
possible.
The technique of HEA is as yet not widely carried out for perioperative blood man-
agement in orthopedics. Nevertheless, it has been shown to be an effective method
for reducing perioperative blood loss (Niemi et al. 2000). HEA aims to achieve an
epidural dermatome block (at least as far as the T2 level) and to establish a suffi-
ciently extensive and dense block of the cardioacceleratory fibers of the thoracic
sympathetic chain. Bradycardia is usually prevented by using a continuous i.v. infu-
sion of a low-dose epinephrine solution. This leads to reduced arterial pressure but
maintains heart rate, central venous pressure, stroke volume, and cardiac output
within normal ranges. Mean arterial blood pressure can be lowered to 50 mmHg
resulting in a reduction of intraoperative (Nelson and Bowen 1986) and postopera-
tive blood loss, as seen in several orthopedic trials (Juelsgaard et al. 2001; Tenholder
and Cushner 2004; Eroglu et al. 2005). HEA seems, therefore, to be an attractive
alternative for certain specific patients and situations.
some studies have still reported a positive effect (Juelsgaard et al. 2001; Henry et al.
2002; Goodnough et al. 2003).
Conclusions
Concerns regarding the safety and high costs of allogenic blood products have
led to increased research and efforts to implement patient-blood management
programs in orthopedic surgery. A multidisciplinary approach to optimizing the
care of patients who might need transfusions should be an obligation for patients
undergoing orthopedic procedures. Both an individualized evaluation of the
patient and clinical management of the transfusion decision-making process are
necessary (including the application of appropriate indications), as are the mini-
mization of blood loss and optimization of the patient’s red cell mass. The need
to reduce allogenic blood transfusions and healthcare costs while ensuring that
blood components are available for the patients who need them is a cornerstone
of appropriate team work.
Anemic patients (women, Hb <12 g/dL; men, Hb <13 g/dL: WHO) should be
treated preoperatively with i.v. iron and EPO as this is the optimal method for
preparing a patient for elective surgery (Goodnough et al. 2000; Coyne et al.
2007; Dahl et al. 2008; So-Osman et al. 2014a, b).
The use of intraoperative platelet gel and fibrin sealant may be an adequate
option as they improve hemostatic effects, lower the incidence of postoperative
wound healing disturbances, present fewer infections, and lead to shorter hospi-
tal stays (Cuenca et al. 2004).
Intraoperative salvage is effective in patients where excessive blood loss
>1.5 L is expected (Goodnough et al. 1999). Due to the high costs associated
with these systems, their utility should be critically assessed before surgery.
Postoperative salvage is still debated, with some controversy regarding possible
side effects. So far, most of the concerns have been proven wrong and the method
itself is inexpensive and easy to use (Healy et al. 1994; Goodnough et al. 2000, 2003;
Juelsgaard et al. 2001; Henry et al. 2002; Cuenca et al. 2004, 2005; Jelkmann 2008).
Finally, it is not only important for surgeons and anesthesiologists to have
early knowledge of the patient’s preoperative Hb level and the expected blood
loss during surgery, but they should also have detailed knowledge about the dif-
ferent alternatives available for reducing the need for allogenic blood products.
All hospitals should adapt their methods with regard to the type of surgery to be
performed and to their financial possibilities.
References
Aguilera X, Martinez-Zapata MJ et al (2013) Efficacy and safety of fibrin glue and tranexamic acid
to prevent postoperative blood loss in total knee arthroplasty: a randomized controlled clinical
trial. J Bone Joint Surg Am 95(22):2001–2007
Alfrey CP, Fishbane S (2007) Implications of neocytolysis for optimal management of anaemia in
chronic kidney disease. Nephron Clin Pract 106(4):c149–c156
360 O.M. Theusinger
Kulnigg S, Stoinov S et al (2008) A novel intravenous iron formulation for treatment of anemia in
inflammatory bowel disease: the ferric carboxymaltose (FERINJECT) randomized controlled
trial. Am J Gastroenterol 103(5):1182–1192
Levy O, Martinowitz U et al (1999) The use of fibrin tissue adhesive to reduce blood loss and the
need for blood transfusion after total knee arthroplasty. A prospective, randomized, multicenter
study. J Bone Joint Surg Am 81(11):1580–1588
Lincoln JA, Lefkowitz DL et al (1995) Exogenous myeloperoxidase enhances bacterial phagocy-
tosis and intracellular killing by macrophages. Infect Immun 63(8):3042–3047
Macdougall IC, Strauss WE et al (2014) A randomized comparison of ferumoxytol and iron
sucrose for treating iron deficiency anemia in patients with CKD. Clin J Am Soc Nephrol
9(4):705–712
Madjdpour C, Spahn DR (2005) Allogeneic red blood cell transfusions: efficacy, risks, alternatives
and indications. Br J Anaesth 95(1):33–42
Mankad PS, Codispoti M (2001) The role of fibrin sealants in hemostasis. Am J Surg 182(2
Suppl):21S–28S
Marik PE, Corwin HL (2008) Efficacy of red blood cell transfusion in the critically ill: a systematic
review of the literature. Crit Care Med 36(9):2667–2674
Matot I, Scheinin O et al (2002) Effectiveness of acute normovolemic hemodilution to minimize
allogeneic blood transfusion in major liver resections. Anesthesiology 97(4):794–800
McShane AJ, Martin JL (1987) Preoxygenation and pulse oximetry may delay detection of esoph-
ageal intubation. J Natl Med Assoc 79(9):987, 991–992
Monk TG, Goodnough LT et al (1999) A prospective randomized comparison of three blood con-
servation strategies for radical prostatectomy. Anesthesiology 91(1):24–33
Moonen AF, Neal TD et al (2006) Peri-operative blood management in elective orthopaedic sur-
gery. A critical review of the literature. Injury 37(Suppl 5):S11–S16
Morgan GE Jr, Mikhail MS et al (2002) Geriatric anesthesia. In: Morgan GE Jr, Mikhail MS,
Murray MJ (eds) Clinical anesthesiology. McGraw-Hill, New York, pp 875–881
Musallam KM, Tamim HM et al (2011) Preoperative anaemia and postoperative outcomes in non-
cardiac surgery: a retrospective cohort study. Lancet 378(9800):1396–1407
Mustoe TA, Pierce GF et al (1987) Accelerated healing of incisional wounds in rats induced by
transforming growth factor-beta. Science 237(4820):1333–1336
Nelson CL, Bowen WS (1986) Total hip arthroplasty in Jehovah’s Witnesses without blood
transfusion. J Bone Joint Surg Am 68(3):350–353
Niemi TT, Pitkanen M et al (2000) Comparison of hypotensive epidural anaesthesia and spinal
anaesthesia on blood loss and coagulation during and after total hip arthroplasty. Acta
Anaesthesiol Scand 44(4):457–464
Okamoto S, Hijikata-Okunomiya A et al (1997) Enzyme-controlling medicines: introduction.
Semin Thromb Hemost 23(6):493–501
Pierce GF, Mustoe TA et al (1989) Platelet-derived growth factor and transforming growth factor-
beta enhance tissue repair activities by unique mechanisms. J Cell Biol 109(1):429–440
Reize P, Wulker N (2007) Foreign blood saving measures in orthopedic surgery. Orthopade
36(6):537–543
Rice L, Alfrey CP (2005) The negative regulation of red cell mass by neocytolysis: physiologic
and pathophysiologic manifestations. Cell Physiol Biochem 15(6):245–250
Rohling RG, Zimmermann AP et al (2000) Intravenous versus oral iron supplementation for
preoperative stimulation of hemoglobin synthesis using recombinant human erythropoietin.
J Hematother Stem Cell Res 9(4):497–500
Rosencher N, Kerkkamp HE et al (2003) Orthopedic Surgery Transfusion Hemoglobin European
Overview (OSTHEO) study: blood management in elective knee and hip arthroplasty in
Europe. Transfusion 43(4):459–469
Rosencher N, Poisson D et al (2005) Two injections of erythropoietin correct moderate anemia in
most patients awaiting orthopedic surgery. Can J Anaesth 52(2):160–165
Schmidt AH, Templeman DC et al (1998) Blood conservation in hip trauma. Clin Orthop Relat Res
(357):68–73
20 Bleeding Management in Elective Orthopedic Surgery 363
Schmied H, Kurz A et al (1996) Mild hypothermia increases blood loss and transfusion require-
ments during total hip arthroplasty. Lancet 347(8997):289–292
Shander A, Hofmann A et al (2007) Estimating the cost of blood: past, present, and future direc-
tions. Best Pract Res Clin Anaesthesiol 21(2):271–289
Shander A, Hofmann A et al (2010) Activity-based costs of blood transfusions in surgical patients
at four hospitals. Transfusion 50(4):753–765
Shore-Lesserson L, Manspeizer HE et al (1999) Thromboelastography-guided transfusion algo-
rithm reduces transfusions in complex cardiac surgery. Anesth Analg 88(2):312–319
Slappendel R, Weber EW et al (2002) Does ibuprofen increase perioperative blood loss during hip
arthroplasty? Eur J Anaesthesiol 19(11):829–831
So-Osman C, Nelissen RG et al (2014a) Patient blood management in elective total hip- and knee-
replacement surgery (part 1): a randomized controlled trial on erythropoietin and blood salvage
as transfusion alternatives using a restrictive transfusion policy in erythropoietin-eligible
patients. Anesthesiology 120(4):839–851
So-Osman C, Nelissen RG et al (2014b) Patient blood management in elective total hip- and knee-
replacement surgery (part 2): a randomized controlled trial on blood salvage as transfusion
alternative using a restrictive transfusion policy in patients with a preoperative hemoglobin
above 13 g/dl. Anesthesiology 120(4):852–860
Spahn DR, Casutt M (2000) Eliminating blood transfusions: new aspects and perspectives.
Anesthesiology 93(1):242–255
Spahn DR, Goodnough LT (2013) Alternatives to blood transfusion. Lancet 381(9880):
1855–1865
Spahn DR, Howell SJ et al (2006) Coronary stents and perioperative anti-platelet regimen:
dilemma of bleeding and stent thrombosis. Br J Anaesth 96(6):675–677
Spahn DR, Chassot PG et al (2007) Pragmatic treatment versus elaborative but incomplete testing:
a Hobson’s choice? Anesthesiology 107(4):526–529
Spalding GJ, Hartrumpf M et al (2007) Bedside thrombelastography. Cost reduction in cardiac
surgery. Anaesthesist 56(8):765–771
Spence RK (1997) Emerging trends in surgical blood transfusion. Semin Hematol 34(3 Suppl
2):48–53
Tartter PI, Quintero S et al (1986) Perioperative blood transfusion associated with infectious com-
plications after colorectal cancer operations. Am J Surg 152(5):479–482
Tenholder M, Cushner FD (2004) Intraoperative blood management in joint replacement surgery.
Orthopedics 27(6 Suppl):s663–s668
Terada N, Arai Y et al (2001) Acute normovolemic hemodilution for radical prostatectomy: can it
replace preoperative autologous blood transfusion? Int J Urol 8(4):149–152
Theusinger OM, Levy JH (2013) Point of care devices for assessing bleeding and coagulation in
the trauma patient. Anesthesiol Clin 31(1):55–65
Theusinger OM, Leyvraz PF et al (2007) Treatment of iron deficiency anemia in orthopedic
surgery with intravenous iron: efficacy and limits: a prospective study. Anesthesiology
107(6):923–927
Theusinger OM, Stein P et al (2014) Applying ‘Patient Blood Management’ in the trauma center.
Curr Opin Anaesthesiol 27(2):225–232
Vamvakas EC, Pineda AA (2000) Autologous transfusion and other approaches to reduce alloge-
neic blood exposure. Baillieres Best Pract Res Clin Haematol 13(4):533–547
Wang GJ, Hungerford DS et al (2001) Use of fibrin sealant to reduce bloody drainage and hemo-
globin loss after total knee arthroplasty: a brief note on a randomized prospective trial. J Bone
Joint Surg Am 83-A(10):1503–1505
Weber EW, Slappendel R et al (2003) COX 2 selectivity of non-steroidal anti-inflammatory drugs
and perioperative blood loss in hip surgery. A randomized comparison of indomethacin and
meloxicam. Eur J Anaesthesiol 20(12):963–966
Weiskopf RB, Aminoff MJ et al (2003) Acute isovolemic anemia does not impair peripheral or
central nerve conduction. Anesthesiology 99(3):546–551
Weiss G, Goodnough LT (2005) Anemia of chronic disease. N Engl J Med 352(10):1011–1023
364 O.M. Theusinger
Winkler M, Akca O et al (2000) Aggressive warming reduces blood loss during hip arthroplasty.
Anesth Analg 91(4):978–984
Wong JC, Torella F et al (2002) Autologous versus allogeneic transfusion in aortic surgery: a multi-
center randomized clinical trial. Ann Surg 235(1):145–151
Wrotniak M, Bielecki T et al (2007) Current opinion about using the platelet-rich gel in orthopaedics
and trauma surgery. Ortop Traumatol Rehabil 9(3):227–238
Perioperative Coagulation in Neuraxial
and Peripheral Nerve Blocks 21
Kyle Kirkham and Eric Albrecht
21.1 Introduction
Given that most of these factors are either unmodifiable or unpredictable at the
start of a procedure, most of the attention placed on the prevention of hemorrhagic
complications has been towards coagulopathies induced by anticoagulant or anti-
thrombotic medication. The 2010 American Society of Regional Anesthesia and
Pain Medicine (ASRA) guidelines on regional anesthesia and antithrombotic med-
ications summarized a total of 26 case reports of hemorrhagic complication: 13
were associated with peripheral or plexus nerve blocks and antithrombotic therapy,
while 13 others occurred without concurrent antithrombotic therapy (Horlocker
et al. 2010). The majority of serious outcomes in this series were related to major
hemorrhage, which is theoretically more easily managed than a neuraxial hema-
toma. However, this series demonstrated that hospital admission may be prolonged
by this outcome, discomfort may be significant, transient neurological deficits may
occur, and consequences may be severe enough to result in death (Maier et al.
2002).
When deciding whether to proceed with a regional technique, consideration must
be given to the site of needle placement and to the consequences of a hemorrhagic
complication specific to that site. Current guidelines therefore advocate caution when
considering peripheral or plexus regional techniques in cases involving anticoagulated
patients. ASRA’s formal recommendation is that these techniques be considered
equivalent to neuraxial procedures (Horlocker et al. 2010). The European Society of
Anaesthesiology (ESA) similarly suggests that deep blocks, including lumbar plexus
and paravertebral blocks as well as the removal of indwelling catheters, should be
considered under neuraxial guidelines (Gogarten et al. 2010) if there is a newer one.
In general, a cautious approach is justified for any patient on anticoagulant medica-
tion. Even superficial procedures like axillary or popliteal neural blockade, where
accompanying vessels are directly compressible, should be carefully weighed up from
a risk-benefit perspective and where reasonable medication should be withheld for
appropriate time periods before needle or catheter placement.
continued their dose until at least 1 day prior to delivery, resulting in only one occur-
rence of traumatic cannula placement in the intervention group vs. two in the control
group and no spinal hematoma. While 3 of the 61 cases described by Vandermeulen
et al. (1994) were using NSAID concurrently, prospective case series have failed to
demonstrate an increased risk associated with continuing aspirin or NSAID during
the performance of neuraxial procedures (Horlocker et al. 1995, 2002).
As a result, NSAID dosing alone does not require adjustment for the purpose of
regional anesthesia. However, the balance of evidence from case reports suggests that
combining nonselective NSAID with other forms of thromboprophylaxis increases
the risk of major hemorrhagic complications (Ruff and Dougherty 1981; Moen et al.
2004). The practical implication of this observation is that a cautious approach should
be taken, as reflected in current guidelines. These limit postoperative use of NSAID
therapy to COX-2 selective agents if the administration of thromboprophylaxis is
anticipated, and patients on pre-procedure combination therapy should have their non-
selective NSAID withheld for 12–24 h prior to the planned procedure.
Thienopyridine derivatives have been associated with increased bleeding in sur-
gical settings and are generally considered contraindications for regional anesthe-
sia. The pharmacokinetics of these agents determines the recommendations for safe
time periods before invasive procedures. There is consensus that clopidogrel should
be discontinued 7 days prior to a regional anesthetic procedure (see Table 21.1).
Ticlopidine has a variable half-life that depends on the duration of its administra-
tion, and there is disagreement between European and North American recommen-
dations. The ESA minimum recommended period is 10 days (Gogarten et al. 2010),
whereas ASRA guidelines recommend 14 days (Horlocker et al. 2010). These
agents have, for example, been associated with both neuraxial and retroperitoneal
hematoma after lumbar plexus blockade.
Antagonist agents of the platelet GP IIb/IIIa receptor include abciximab, eptifi-
batide, and tirofiban. They have considerably different durations of effect on platelet
function, with a return to normal function requiring only 4–8 h after eptifibatide or
368 K. Kirkham and E. Albrecht
tirofiban, but a minimum of 24–48 h after abciximab therapy. The antiplatelet effect
of these agents is profound: it is prudent to target the upper limits of these ranges
when planning the removal of an indwelling catheter and to carefully monitor neu-
rological function afterward. In addition to their intrinsic activity, these agents gen-
erate a risk of significant thrombocytopenia that may last beyond their
pharmacological duration (Berkowitz et al. 1997). As such, it is recommended that
a platelet count be performed for any patient who has received a GP IIb/IIIa
inhibitor.
The performance of regional anesthesia in cases using low molecular weight hepa-
rins (LMWH) deserves special consideration for three reasons. Firstly, in many cen-
ters, LMWH has replaced UFH or oral anticoagulants as the thromboprophylaxis of
choice. As a result, significant numbers of patients are exposed to this class of medi-
cation annually. Secondly, traditional coagulation tests such as activated partial
thromboplastin time (aPTT) and activated clotting time (ACT) are unaffected by
LMWH, making monitoring challenging. Thirdly, the introduction of LMWH, par-
ticularly in North America, had a demonstrable impact on the reported number of
hemorrhagic complications from neuraxial needle placement in a relatively short
period of time. Between 1993 and 1998, 60 cases of spinal hematoma were reported
after the introduction of LMHW to the United States (Horlocker et al. 2010). It has
been estimated that this represented a frequency of approximately 1 in 3,100 epi-
dural catheter procedures and 1 in 40,800 spinal procedures (Schroeder 1998).
These observations contrasted with prior European experience where, in one exam-
ple, Bergqvist et al. (1992) pooled 9,013 neuraxial patients receiving LMWH from
11 studies and found no incidence of hematoma. This geographical difference may
partially be accounted for by different dosing regimen, with North American
patients often receiving twice-daily dosing.
The increased risk of twice-daily dosing is reflected in both current North
American and European guidelines. Recommendations include delaying needle
placement or catheter removal for a period of 10–12 h after prophylactic once-daily
dosing and then restarting therapy for a minimum of 2–4 h after the procedure.
Initiation of postoperative therapy should begin no earlier than 6–8 h after needle
placement. Twice-daily or therapeutic dosing should be withheld for a minimum of
24 h before a regional procedure or the removal of an indwelling catheter. In this
setting, therapy may be restarted a similar 2–4 h after the procedure. However,
ASRA guidelines further recommend delaying the initiation of postoperative
370 K. Kirkham and E. Albrecht
This class of medication has recently expanded, with new agents in development
and approved for clinical use. Many of these drugs present unique challenges to the
regional anesthesiologist. The anti-Xa drug with the longest history, in both Europe
and North America, is fondaparinux, but the introduction of rivaroxaban, apixaban,
and others has led to an increasing number of patients presenting for chronic antico-
agulation. The half-lives of these agents are typically long (allowing for once-daily
dosing), but are varied: fondaparinux near 20 h and rivaroxaban closer to 6 h. The
literature supporting periprocedural guidelines is developing. Recommendations are
typically cautious and based directly on what is known of the agents’ pharmacology.
For example, the manufacturer of the univalent direct thrombin inhibitor dabigatran
372 K. Kirkham and E. Albrecht
21.2.6 Thrombolytics
References
Bergqvist D, Lindblad B, Matzsch T (1992) Low molecular weight heparin for thromboprophy-
laxis and epidural/spinal anaesthesia – is there a risk? Acta Anaesthesiol Scand 36:605–609
Berkowitz SD, Harrington RA, Rund MM et al (1997) Acute profound thrombocytopenia after
c7E3 Fab (Abciximab) therapy. Circulation 95:809–813
CLASP Collaborative Group (1994) CLASP: a randomized trial of low-dose aspirin for the pre-
vention and treatment of pre-eclampsia among 9364 pregnant women. Lancet 343:619–629
Gogarten W, Vandermeulen E, Van Aken H et al (2010) Regional anaesthesia and antithrombotic
agents: recommendations of the European Society of Anaesthesiology. Eur J Anaesthesiol
27:999–1015
Horlocker TT, Wedel DJ, Schroeder DR et al (1995) Preoperative antiplatelet therapy does not
increase the risk of spinal hematoma associated with regional anesthesia. Anesth Analg
80:303–309
Horlocker TT, Bajwa ZH, Ashraf Z et al (2002) Risk assessment of hemorrhagic complications
associated with nonsteroidal anti-inflammatory medications in ambulatory pain clinic patients
undergoing epidural steroid injection. Anesth Analg 95:1691–1697
Horlocker TT, Wedel DJ, Rowlingson JC et al (2010) Regional anesthesia in the patient receiving
antithrombotic or thrombolytic therapy, American Society of Regional Anesthesia and Pain
Medicine evidence-based guidelines (third edition). Reg Anesth Pain Med 35:64–101
Linkins L-A, Dans AL, Moores LK et al (2012) Treatment and prevention of heparin-induced
thrombocytopenia: antithrombotic therapy and prevention of thrombosis, 9th ed: American
College of Chest Physicians evidence-based clinical practice guidelines. Chest 141(2
Suppl):e495S–e530S
Liu SS, Mulroy MF (1998) Neuraxial anesthesia and analgesia in the presence of standard heparin.
Reg Anesth Pain Med 23(6 Suppl 2):157–163
Liu SS, Buvanendran A, Viscusi ER et al (2011) Uncomplicated removal of epidural catheters in
4365 patients with international normalized ratio greater than 1.4 during initiation of warfarin
therapy. Reg Anesth Pain Med 36:231–235
Maier C, Gleim M, Weiss T et al (2002) Severe bleeding following lumbar sympathetic blockade
in two patients under medication with irreversible platelet aggregation inhibitors. Anesthesiology
97:740–743
Moen V, Dahlgren N, Irestedt L (2004) Severe neurological complications after central neuraxial
blockades in Sweden 1990–1999. Anesthesiology 101:950–959
Parvizi J, Viscusi ER, Harrison GF et al (2006) Can epidural anesthesia and warfarin be coadmin-
istered? Clin Orthop Relat Res 456:133–137
Raj PP, Shah RV, Kaye AD et al (2004) Bleeding risk in interventional pain practice: assessment,
management, and review of the literature. Pain Physician 7:3–51
Ruff RL, Dougherty JH (1981) Complications of lumbar puncture followed by anticoagulation.
Stroke 12:879–881
Schroeder DR (1998) Statistics: detecting a rare adverse drug reaction using spontaneous reports.
Reg Anesth Pain Med 23(Suppl 2):183–189
Singelyn FJ, Verheyen C, Piovella F et al (2007) The safety and efficacy of extended thrombopro-
phylaxis with fondaparinux after major orthopedic surgery of the lower limb with or without a
neuraxial or deep peripheral nerve catheter: the EXPERT study. Anesth Analg
105:1540–1547
Tryba M (1993) Epidural regional anesthesia and low molecular heparin: Pro. [German].
Anasthesiol Intensivmed Notfallmed Schmerzther 28:179–181
Vandermeulen EP, Van Aken H, Vermylen J (1994) Anticoagulants and spinal-epidural anesthesia.
Anesth Analg 79:1165–1177
Warkentin TE, Levine MN, Hirsh J et al (1995) Heparin-induced thrombocytopenia in patients
treated with low-molecular-weight heparin or unfractionated heparin. N Engl J Med
332:1330–1335
Part III
Post-operative Hemostasis
Coagulation Disorders in Intensive
Care Patients 22
Marcel Levi
22.1 Introduction
Critically ill patients commonly suffer from coagulation abnormalities (Levi and
Opal 2006). A myriad of altered coagulation parameters is often detectable, such as
thrombocytopenia, longer global coagulation times, reduced levels of coagulation
inhibitors, or high levels of fibrin split products. Each of these clotting derange-
ments may derive from a variety of different pathophysiological mechanisms. Some
patients may have a marked coagulopathy, yet it can go largely undetected when
measured using routine coagulation assays. Proper identification of the underlying
cause for these coagulation abnormalities is required since different coagulation
disorders may necessitate different diagnostic and therapeutic management strate-
gies. This chapter reviews the most frequently occurring coagulation abnormalities
in patients in intensive care units (ICUs). Emphasis is put on the differential diagno-
sis, the underlying molecular and pathogenetic pathways, and the appropriate diag-
nostic and therapeutic interventions.
22.2 Incidence
M. Levi, MD
Department of Vascular Medicine and Internal Medicine, Academic Medical Center,
University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
e-mail: [email protected]
2000). The risk of intracerebral bleeding in critically ill patients during ICU admis-
sion is relatively low (0.3–0.5 %), but 88 % of patients with this complication have
platelet counts <100 × 109/l. Moreover, a decrease in platelet count may indicate
ongoing coagulation activation, which contributes to microvascular failure and
organ dysfunction. Early identification of these patients is crucial to the provision
of adequate supportive care (Ahmed et al. 2009; Schultz 2009). Regardless of the
cause, thrombocytopenia is an independent predictor of ICU mortality in multi-
variate analyses, with various studies showing a relative risk of 1.9–4.2 (Fig. 22.1)
(Strauss et al. 2002; Vanderschueren et al. 2000). Other coagulation test abnormali-
ties frequently observed in ICU patients include elevated fibrin split products and
reduced levels of coagulation inhibitors. Fibrin split products, such as D-dimer,
were detectable in 42 % of a consecutive case series of ICU patients, in 80 % of
trauma patients, and in 99 % of patients with sepsis (Bernard et al. 2001; Shorr
et al. 2002). Low levels of coagulation inhibitors, such as antithrombin and protein
C, are found in 40–60 % of trauma patients and 90 % of sepsis patients (Bernard
et al. 2001).
1,400 50
1,200
40
Number of patients
1,000
Mortality
800 30
600 20
400
10
200
0 0
<50 50-100 100-150 >150
Platelet count (×109/I)
Fig. 22.1 Distribution of nadir platelet count (black bars) and survival (striped bars) in a pooled
analysis of four clinical studies of consecutive groups of patients admitted to an ICU (From Levy
and Opal (2010) with permission)
22 Coagulation Disorders in Intensive Care Patients 379
There are many causes of thrombocytopenia in critically ill patients. Table 22.1
summarizes the most frequently occurring diagnoses.
Sepsis is a clear risk factor for thrombocytopenia in critically ill patients, and the
severity of sepsis correlates with the decrease in platelet count (Mavrommatis et al.
2000). The main factors contributing to thrombocytopenia in patients with sepsis
380 M. Levi
occur. A consecutive series of critically ill ICU patients who received heparin
revealed an incidence of 1 % in this setting (Verma et al. 2003). Unfractionated
heparin carries a higher risk of HIT than low molecular weight heparin (LMWH).
Thrombosis may occur in 25–50 % of patients with HIT (with fatal thrombosis in
4–5 %) (Warkentin 2003). The diagnosis of HIT is based on the detection of HIT
antibodies in combination with the occurrence of thrombocytopenia in a patient
receiving heparin, with or without concomitant arterial or venous thrombosis. It
should be mentioned that the commonly used ELISA (enzyme-linked immunosor-
bent assay) for HIT antibodies has a high negative predictive value (100 %), but a
very low positive predictive value (10 %) (Verma et al. 2003). The gold standard for
the diagnosis of HIT is a sensitive platelet activation assay; however, this test is not
routinely available. Normalization of the number of platelets 1–3 days after discon-
tinuation of heparin may further support the diagnosis of HIT.
The category of thrombotic microangiopathies encompasses syndromes such as
thrombotic thrombocytopenic purpura, hemolytic–uremic syndrome, severe malig-
nant hypertension, chemotherapy-induced microangiopathic hemolytic anemia, and
the HELLP syndrome (hemolysis, elevated liver enzymes, low platelet count)
(Moake 2002). A common pathogenetic feature of these clinical entities appears to
be endothelial damage, causing platelet adhesion and aggregation, thrombin forma-
tion, and impaired fibrinolysis. The multiple clinical consequences of this extensive
endothelial dysfunction include thrombocytopenia, mechanical fragmentation of
red blood cells with hemolytic anemia, and obstruction of the microvasculature of
various organs such as the kidneys and the brain (leading to renal failure and neuro-
logic dysfunction, respectively). Despite this common final pathway, the various
thrombotic microangiopathies have different underlying etiologies. Thrombotic
thrombocytopenic purpura is caused by congenital or acquired (autoimmune) defi-
ciency of von Willebrand factor-cleaving protease (ADAMTS13); this results in
endothelial cell-attached ultra-large von Willebrand multimers that readily bind to
platelet surface glycoprotein Ib and cause platelet adhesion and aggregation (Tsai
2003). In hemolytic–uremic syndrome, a cytotoxin released upon infection with a
specific serogroup of gram-negative microorganisms (usually E. coli serotype
O157:H7) is responsible for endothelial cell and platelet activation. In case of
malignant hypertension or chemotherapy-induced thrombotic microangiopathy,
direct mechanical and chemical damages to the endothelium are, respectively, pre-
sumed responsible for the enhanced endothelial cell–platelet interaction. A diagnosis
of thrombotic microangiopathy relies upon the combination of thrombocytopenia,
the Coombs-negative hemolytic anemia, and the presence of schistocytes in the
blood smear. Additional information can be achieved by measurement of
ADAMTS13; however, low levels of ADAMTS13 may occur in all forms of throm-
botic microangiopathy (van den Born et al. 2008).
Drug-induced thrombocytopenia is another frequent cause of thrombocytopenia
in an ICU setting (Stephan et al. 1999). It may be caused by drug-induced myelo-
suppression, such as that caused by cytostatic agents or by immune-mediated mech-
anisms. Drug-induced thrombocytopenia is a difficult diagnosis in an ICU setting as
patients are often exposed to multiple agents and have numerous other potential
382 M. Levi
It is important to emphasize that global coagulation tests, such as PT and aPTT, are
a poor reflection of in vivo hemostasis. However, these tests are a convenient method
of quickly estimating the concentration of one or at times multiple coagulation
factors for which each test is sensitive (Table 22.2) (Greaves and Preston 2001).
In general, coagulation tests will take longer if coagulation factor levels are below
50 %. This is relevant since the coagulation factor levels needed for adequate hemo-
stasis are somewhere between 25 and 50 % (Edmunds 2001).
Longer global coagulation tests may be due to a deficiency of one or more coagu-
lation factors. In addition, but more rarely, the presence of an inhibiting antibody
with a major in vivo relevance (such as in acquired hemophilia), but a clinically
insignificant laboratory phenomenon, should be considered. The presence of lupus
anticoagulant may cause a longer aPTT and can be associated with thrombocytope-
nia as well. Paradoxically, lupus anticoagulant may dramatically increase the risk of
thrombosis. The presence of an inhibiting antibody can be confirmed using a simple
mixing experiment. As a general rule, if a longer global coagulation test cannot be
corrected by mixing 50 % of patient plasma with 50 % of normal plasma, then an
inhibiting antibody is likely to be present.
in a phase II/III clinical study involving 750 patients with sepsis and disseminated
intravascular coagulation. Twenty-eight-day mortality was 17.8 % in the thrombo-
modulin group and 21.6 % in the placebo group. Markers of coagulation activation
were lower in the thrombomodulin group than in the placebo group. There were no
differences between the groups in bleeding or thrombotic events.
Conclusions
Coagulation abnormalities occur frequently in critically ill patients and may have
a significant impact on the outcome. Treatment of ICU patients should be directed
primarily at their underlying condition, but an adequate explanation for the coag-
ulation abnormalities of these critically ill patients, together with supportive
therapy, may also be required. Deficiencies in platelets and coagulation factors in
bleeding patients or patients at risk of bleeding can be made up by transfusion of
platelet concentrate or plasma products, respectively. In addition, pro-hemostatic
treatment may be beneficial in cases of severe bleeding, whereas restoring physi-
ological anticoagulant pathways may be helpful in patients with sepsis and DIC.
References
Ahmed A, Kojicic M, Herasevich V et al (2009) Early identification of patients with or at risk of
acute lung injury. Neth J Med 67:268–271
Bailey B, Amre DK, Gaudreault P (2003) Fulminant hepatic failure secondary to acetaminophen
poisoning: a systematic review and meta-analysis of prognostic criteria determining the need
for liver transplantation. Crit Care Med 31:299–305
Bernard GR, Vincent JL, Laterre PF et al (2001) Efficacy and safety of recombinant human activated
protein C for severe sepsis. N Engl J Med 344:699–709
Dhainaut JF, Yan SB, Joyce DE et al (2004) Treatment effects of drotrecogin alfa (activated)
in patients with severe sepsis with or without overt disseminated intravascular coagulation.
J Thromb Haemost 2:1924–1933
Eckle I, Seitz R, Egbring R et al (1991) Protein C degradation in vitro by neutrophil elastase. Biol
Chem Hoppe Seyler 372:1007–1013
Edmunds LH (2001) Hemostatic problems in surgical patients. In: Colman RW, Hirsh J, Marder
VJ, Clowes AW, George JN (eds) Hemostasis and thrombosis. Basic principles and clinical
practice. Lippincott William & Wilkins, Philadelphia, pp 1031–1043
Esmon CT (1987) The regulation of natural anticoagulant pathways. Science 235:1348–1352
Esmon CT (2000) The endothelial cell protein C receptor. Thromb Haemost 83:639–643
Esmon CT (2001) Role of coagulation inhibitors in inflammation. Thromb Haemost
86:51–56
Faust SN, Levin M, Harrison OB et al (2001) Dysfunction of endothelial protein C activation in
severe meningococcal sepsis. N Engl J Med 345:408–416
Folman CC, Linthorst GE, van Mourik J et al (2000) Platelets release thrombopoietin (Tpo) upon
activation: another regulatory loop in thrombocytopoiesis? Thromb Haemost 83:923–930
Francois B, Trimoreau F, Vignon P et al (1997) Thrombocytopenia in the sepsis syndrome: role of
hemophagocytosis and macrophage colony-stimulating factor. Am J Med 103:114–120
Greaves M, Preston FE (2001) Approach to the bleeding patient. In: Colman RW, Hirsh J, Marder
VJ, Clowes AW, George JN (eds) Hemostasis and thrombosis. Basic principles and clinical
practice. Lippingcott William&Wilkins, Philadelphia, pp 1031–1043
Griffin JH, Fernandez JA, Gale AJ et al (2007) Activated protein C. J Thromb Haemost 5(Suppl 1):
73–80: 73–80
388 M. Levi
Hellstern P, Muntean W, Schramm W et al (2002) Practical guidelines for the clinical use of
plasma. Thromb Res 107(Suppl 1):S53
Hermanides J, Huijgen R, Henny CP et al (2009) Hip surgery sequentially induces stress hypergly-
caemia and activates coagulation. Neth J Med 67:226–229
Hirsh J, Heddle N, Kelton JG (2004) Treatment of heparin-induced thrombocytopenia: a critical
review. Arch Intern Med 164:361–369
Levi M (2012) Safety of prohemostatic interventions. Semin Thromb Hemost 38:282–288
Levi M, Lowenberg EC (2008) Thrombocytopenia in critically ill patients. Semin Thromb Hemost
34:417–424
Levi M, Opal SM (2006) Coagulation abnormalities in critically ill patients. Crit Care 10:222
Levi M, ten Cate H (1999) Disseminated intravascular coagulation. N Engl J Med 341:586–592
Levi M, van der Poll T (2008) The role of natural anticoagulants in the pathogenesis and manage-
ment of systemic activation of coagulation and inflammation in critically ill patients. Semin
Thromb Hemost 34:459–468
Levi M, Cromheecke ME, de Jonge E et al (1999) Pharmacological strategies to decrease
excessive blood loss in cardiac surgery: a meta-analysis of clinically relevant endpoints.
Lancet 354:1940–1947
Levi M, de Jonge E, van der Poll T (2001) Rationale for restoration of physiological anticoagulant
pathways in patients with sepsis and disseminated intravascular coagulation. Crit Care Med
29:S90–S94
Levi M, van der Poll T, Buller HR (2004) Bidirectional relation between inflammation and coagu-
lation. Circulation 109:2698–2704
Levi M, Nieuwdorp M, van der Poll T et al (2008) Metabolic modulation of inflammation-induced
activation of coagulation. Semin Thromb Hemost 34:26–32
Levi M, Toh CH, Thachil J et al (2009) Guidelines for the diagnosis and management of dissemi-
nated intravascular coagulation. Br J Haematol 145:24–33
Levi M, Levy JH, Andersen HF et al (2010) Safety of recombinant activated factor VII in random-
ized clinical trials. N Engl J Med 363:1791–1800
Levi M, Opal S (2010) Coagulation abnormalities in the critically ill. Surgical Intensive Care
Medicine. O’Donnell J, Nacul F, Springer US: 371–378
Levi M, Schouten M, van der Poll T (2008) Sepsis, coagulation, and antithrombin: old lessons and
new insights. Semin Thromb Hemost 34:742–746
Mannucci PM (1997) Desmopressin (DDAVP) in the treatment of bleeding disorders: the first 20
years. Blood 90:2515–2521
Mannucci PM, Levi M (2007) Prevention and treatment of major blood loss. N Engl J Med
356:2301–2311
Mavrommatis AC, Theodoridis T, Orfanidou A et al (2000) Coagulation system and platelets are
fully activated in uncomplicated sepsis. Crit Care Med 28:451–457
Mesters RM, Helterbrand J, Utterback BG et al (2000) Prognostic value of protein C concentrations
in neutropenic patients at high risk of severe septic complications. Crit Care Med 28:2209–2216
Moake JL (2002) Thrombotic microangiopathies. N Engl J Med 347:589–600
Nawroth PP, Stern DM (1986) Modulation of endothelial cell hemostatic properties by tumor
necrosis factor. J Exp Med 163:740–745
Porte RJ, Molenaar IQ, Begliomini B et al (2000) Aprotinin and transfusion requirements in
orthotopic liver transplantation: a multicentre randomised double-blind study. EMSALT Study
Group. Lancet 355:1303–1309
Ranieri VM, Thompson BT, Barie PS et al (2012) Drotrecogin alfa (activated) in adults with septic
shock. N Engl J Med 366:2055–2064
Saito H, Maruyama I, Shimazaki S et al (2007) Efficacy and safety of recombinant human soluble
thrombomodulin (ART-123) in disseminated intravascular coagulation: results of a phase III,
randomized, double-blind clinical trial. J Thromb Haemost 5:31–41
Schultz MJ (2009) Early recognition of critically ill patients. Neth J Med 67:266–267
22 Coagulation Disorders in Intensive Care Patients 389
Shakur H, Roberts I, Bautista R et al (2010) Effects of tranexamic acid on death, vascular occlusive
events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a
randomised, placebo-controlled trial. Lancet 376:23–32
Shorr AF, Thomas SJ, Alkins SA et al (2002) D-dimer correlates with proinflammatory cytokine
levels and outcomes in critically ill patients. Chest 121:1262–1268
Stephan F, Hollande J, Richard O et al (1999) Thrombocytopenia in a surgical ICU. Chest
115:1363–1370
Strauss R, Wehler M, Mehler K et al (2002) Thrombocytopenia in patients in the medical intensive
care unit: bleeding prevalence, transfusion requirements, and outcome. Crit Care Med
30:1765–1771
Taylor FBJ, Stearns-Kurosawa DJ, Kurosawa S et al (2000) The endothelial cell protein C receptor
aids in host defense against Escherichia coli sepsis. Blood 95:1680–1686
Taylor FBJ, Toh CH, Hoots WK et al (2001) Towards definition, clinical and laboratory criteria, and
a scoring system for disseminated intravascular coagulation. Thromb Haemost 86:1327–1330
Thachil J, Toh CH, Levi M et al (2012) The withdrawal of activated protein C from the use in
patients with severe sepsis and DIC. Br J Haematol 37:10–2141
Tsai HM (2003) Platelet activation and the formation of the platelet plug: deficiency of ADAMTS13
causes thrombotic thrombocytopenic purpura. Arterioscler Thromb Vasc Biol 23:388–396
van den Born BJ, van der Hoeven NV, Groot E et al (2008) Association between thrombotic micro-
angiopathy and reduced ADAMTS13 activity in malignant hypertension. Hypertension
51:862–866
Vanderschueren S, De Weerdt A, Malbrain M et al (2000) Thrombocytopenia and prognosis in
intensive care. Crit Care Med 28:1871–1876
Vary TC, Kimball SR (1992) Regulation of hepatic protein synthesis in chronic inflammation and
sepsis. Am J Physiol 262:C445–C452
Verma AK, Levine M, Shalansky SJ et al (2003) Frequency of heparin-induced thrombocytopenia
in critical care patients. Pharmacotherapy 23:745–753
Vlaar AP, Schultz MJ, Juffermans NP (2009) Transfusion-related acute lung injury: a change of
perspective. Neth J Med 67:320–326
Warkentin TE (2003) Heparin-induced thrombocytopenia: pathogenesis and management. Br J
Haematol 121:535–555
Warkentin TE, Aird WC, Rand JH (2003) Platelet-endothelial interactions: sepsis, HIT, and
antiphospholipid syndrome. Hematology Am Soc Hematol Educ Program 70:497–519
Wheeler AP, Bernard GR (1999) Treating patients with severe sepsis. N Engl J Med 340:207–214
Perioperative Thromboprophylaxis
23
Marc Aldenkortt and Marc Licker
23.1 Introduction
TF VIIa
X IX
VIIIa
IXa
Va
Xa
Activated
Platelets
Prothrombinase
Complex
IIa
Fibrinogen Fibrin
Fig. 23.1 Cell-based activation of the coagulation cascade (primary and secondary hemostasis)
upon endothelial injury (TF tissue factor; coagulation factors and their activated forms IIa, Va, X/
Xa, VIIa, and IX/IXa)
23.2.1 Definition
Deep vein thrombosis (DVT) and pulmonary embolism (PE) are considered two
manifestations of the same anatomo-clinical entity, namely, venous thromboembo-
lism (VTE). Venous thrombi are predominately composed of red blood cells, plate-
lets, and leukocytes, all bound together by fibrin. PE is the third most common
cause of hospital-related death, and it is the most common preventable cause of
hospital-related death.
23.2.2 Prevalence
The highest risk of DVT occurs in the first postoperative week, but it remains important
up to 3–6 weeks following major thoracoabdominal or orthopedic procedures (White
et al. 2003). In the absence of antithrombotic prophylaxis and if screened by contrast
venography, duplex ultrasound, or fibrinogen labeling, the prevalence of asymptomatic
23 Perioperative Thromboprophylaxis 393
DVT can be as high as 40–80 % following total knee arthroplasty (TKA) or total hip
arthroplasty (THA) and 15–30 % following major abdominal surgery (Stringer et al.
1989; White et al. 2003). Pain or tenderness at palpation is a characteristic symptom of
DVT that may be accompanied by local swelling, erythema, and increased warmth, as
well as the presence of dilated veins (collaterals) on the chest wall or legs. The preva-
lence of symptomatic VTE has largely decreased over the last four decades because of
important changes in perioperative surgical care, including less invasive surgery and
earlier ambulation. Following major joint surgery, the rate of symptomatic VTE events
has declined from 15 to 30 % prior to 1980 (without prophylaxis) to less than 2 % after
2000 (with prophylaxis) (Januel et al. 2012). Ethnicity, geographical location, and life-
style may also influence the prevalence of VTE. In major orthopedic surgery, the rate
of proximal DVT diagnosed by venography exceeds 20 % in Western countries but is
less than 10 % in Asian countries (Kanchanabat et al. 2011).
Most thrombi start in the calf and have few clinical consequences if they remain
confined below the knee. The probability that calf DVT will extend to involve the
proximal iliofemoral and/or visceral veins, and subsequently cause PE, increases
with the severity and persistence of the initiating prothrombotic stimulus.
Alternatively, the fresh thrombus may dissolve by activation of the fibrinolytic
394 M. Aldenkortt and M. Licker
system, or the granulation tissue may invade the occlusive blood clot which can
later be recanalized. Although the patency of the vessel can be restored, destruction
of the venous valves results in chronic venous hypertension and secondary varices.
Studies investigating the spontaneous evolution of untreated proximal DVT are
scarce, but they do suggest a risk of post-thrombotic syndrome (leg swelling, ten-
derness, and ulcers) in 25–50 % and of clinical PE in 10–40 % of cases (Huber et al.
1992). Importantly, about 10 % of PE are rapidly fatal as a result of acute heart
failure – the right ventricle being unable to face the sudden elevation of afterload if
thrombi occlude more than two thirds of the pulmonary vasculature. In patients
surviving an acute PE episode, pulmonary perfusion normalizes in about two thirds
of patients, while in 4–6 %, relapsing embolization of small clots from persisting
DVT may lead to chronic pulmonary hypertension (Pengo et al. 2004).
From an economic standpoint, VTE increase the global burden on both the
healthcare system and individuals by up to USD 1.5 billion a year in the USA and
EUR 3.07 billion in the European Union (Oger 2000; Dobesh 2009). In Europe, the
cost of VTE in orthopedic surgery has been estimated at EUR 8,265 per patient,
thereby doubling the average cost of orthopedic inpatient care (Ollendorf et al.
2002). More recently, the risk-adjusted total healthcare cost, including the benefi-
ciary’s cost share over 1 year, was found to be USD 13,500 higher in patients with
a VTE (versus without a VTE) following TKA (Baser et al. 2011).
The concept of primum non nocere applies to VTE prophylaxis as it does to all areas
of medicine. Each patient should be assessed and treated on an individual basis and
balance maximal antithrombotic efficacy against minimal bleeding, while taking
into account patient convenience.
Several models of VTE risk assessment have been published, namely, those
developed by Caprini, Rogers, Cohen, and Kucher (Caprini et al. 2001; Cohen et al.
2005; Kucher et al. 2005; Rogers et al. 2007). Each of these models encompasses a
list of exposure risk factors (presenting illness or procedure) and predisposing risk
factors (genetic and clinical characteristics). The Caprini score was validated in a
large retrospective study on a sample of general, vascular, plastic, and urological
surgery patients (Table 23.2) (Bahl et al. 2010). Patient and surgical risk factors are
summed to produce a cumulative score which falls into one of four levels of risk,
with a growing incidence of VTE: low risk (VTE ~2 %) if the total score is 0–1,
moderate risk (VTE ~5–10 %) if the score equals 2, high risk (VTE 20–40 %) if the
score is 3–4, and very high risk (VTE ≥40 %) if the score is ≥5.
Alternatively, the American College of Chest Physicians’ (ACCP) guidelines
stratify the risks of VTE according to the type of procedure (orthopedic, pelvic/
abdominal, cardiac, thoracic, or intracranial/spinal) and to some disease character-
istics (trauma, cancer) (Falck-Ytter et al. 2012; Gould et al. 2012a). Though simple,
this approach fails to provide an individualized approach to risk assessment and
thromboprophylaxis.
Table 23.2 Assessment of the risk of venous thromboembolism
Risk factor 1 pt 2 pts 3 pts 5 pts
Age 41–60 years old 61–74 years old ≥75 years old
Procedure/trauma Minor surgery Arthroscopy, laparoscopy Elective major lower limb
(>45 min) arthroplasty (hip, knee)
Major surgery (>45 min) Multiple trauma (<1 month)
Hip, pelvis, or leg fracture
Central venous access Acute spinal cord injury (<1 month)
Functional status Bed rest (medical patient) Immobilizing plaster cast
(<1 month)
Confined to bed (>72 h)
Physical status COPD Cancer
CHF or AMI
23 Perioperative Thromboprophylaxis
The risk of bleeding should be carefully balanced against the need for pharmaco-
logical VTE prophylaxis, given the dire consequences of blood loss (hemorrhagic
shock) and compressive hematoma requiring homologous transfusion and/or reop-
eration (Chee et al. 2008). In addition to surgical characteristics (site, extent, and
duration), assessing the risk of bleeding should also consider the following: the
patient and family history (excessive bleeding after tooth extraction, surgery/trauma,
or abortion/delivery); the presence of renal failure, liver disease, or peptic ulcer; and
the concomitant use of anticoagulant/antiplatelet drugs. The average cost incurred
by major bleeding averages USD 113 per patient receiving anticoagulant medica-
tion (Muntz et al. 2004).
1
www.nice.org.uk/guidance/index.
398 M. Aldenkortt and M. Licker
2011; Urbankova et al. 2005). A recent systematic review indicated that IPC pro-
vided sufficient prophylaxis for the majority of gynecology patients undergoing
benign surgery; in comparison with no prophylaxis, IPC has been shown to reduce
DVT by 60 % (relative risk 0.40, 95 % CI 0.29–0.56; p < 0.05) (Rahn et al. 2011).
Direct comparison between ECS and IPC has failed to demonstrate a significant
superiority of either method, although IPC was associated with lower DVT rates in
three out of ten studies (Morris and Woodcock 2010). Since IPC and ECS are not as
effective as antithrombotic drugs, these devices should be used as support to phar-
macological prophylaxis, as a sole therapy in low-risk patients, or if drugs are con-
traindicated (e.g., early postoperative period, increased risk of bleeding) (Kakkos
et al. 2008). The ACCP guidelines assign a 2A recommendation to the use of com-
bination prophylaxis in the highest-risk group since the rate of DVT can be reduced
from 19 % with pharmacological prophylaxis alone to less than 4 % with combined
modalities (Kakkos et al. 2012). Unfortunately, compliance with IPC is question-
able and studies on the optimal device are still lacking.
Following the success of aspirin, vitamin K antagonists (VKA), and heparin in pre-
venting thrombotic events, several parenterally administered anticoagulants (hirudin
analogues, argatroban) have been developed for patients with heparin-induced
thrombocytopenia (HIT) and antithrombin deficiency (Fig. 23.2). More recently, oral
TF VIIa
1940s
ORAL PARENTERAL
Oral Vitamine K Antagonists
X IX
Vit K Antagonist
Factors Fondaparinus
VIIIa
II, VII, IX, X IXa AT
1980s
SC Low Molecular Weight Heparin Va Danaparoid
Xa
1990s Activated AT
IV Direct Thrombin inhibitors Rivaroxaban Platelets
(Desirudin, Bivalirubin) Apixaban Prothrombinase AT
2002 Complex LMWH
IV Indirect inhibitors of factors Xa Aspirin, clopidrogel UFH
(Fondaparinus, Danaparoid) prasugrel
2004
IIa Desirudin
Oral direct inhibitors of Thrombin Dabigatran Bivalirubin
(Dabigatran)
2008
Oral direct inhibitors of factor Xa
(Rivaroxaban)
Fibrinogen Fibrin
Fig. 23.2 Common antithrombotic drugs: historical development and site of action in the coagu-
lation cascade
23 Perioperative Thromboprophylaxis 399
23.4.4.1 Aspirin
Aspirin causes an irreversible inhibition of cyclooxygenase, an essential enzyme for the
production of thromboxane A2, a powerful stimulant of platelet aggregation. Thus, aspi-
rin produces an effective anti-aggregant action for the platelet’s entire lifespan. Although
platelets play a role in the initiation and propagation of VTE, aspirin (even at a high
dose) is less effective than heparin derivatives in preventing VTE. Current guidelines,
therefore, do not recommend thromboprophylaxis by aspirin alone, except in patients at
risk of bleeding. Patients receiving aspirin to prevent cardiovascular thrombotic events
(atrial fibrillation, prior myocardial infarct or stroke, or vascular stent implantation), but
requiring noncardiac surgery, may continue aspirin around the time of surgery, instead
of stopping it 7–10 days preoperatively (Grade 2C) (Douketis et al. 2012).
23.4.4.2.2 Pharmacokinetics
Rapidly absorbed from the gastrointestinal tract after oral intake, VKA have high
bioavailability and reach maximal blood concentrations about 90 min after admin-
istration. Warfarin has a longer half-life (36–42 h), circulates highly bound to
plasma proteins, and accumulates in the liver where the two isomers are metaboli-
cally transformed. Acenocoumarol has a lower degree of digestive absorption and a
shorter half-life (8–12 h), with up to 36 % of the drug being retrieved inert in the
stool. Phenprocoumon has 100 % bioavailability and a very long half-life (5–6 days).
The dose–response of VKA may vary up to tenfold between individuals and two-
to threefold within an individual. The cytochrome P450 complex (e.g., CYP2C9,
CYP3A4) has become clearly established as the predominant catalyst responsible
for the metabolism of its more potent S-enantiomer and its activity may be
Table 23.4 Common antithrombotic drugs
400
23.4.4.2.3 Monitoring
Because of the large variability of plasma concentration and the unpredictability of
VKA, regular monitoring of their effect on coagulation is of utmost importance. The
international normalized ratio (INR) is the gold standard test for VKA monitoring,
and the targeted range should be within 2.0–3.0 (or a closer range, 2.0–2.5, in patients
at risk of bleeding). The INR is a mathematically transformed or calculated value
converting PT in seconds to a standard ratio value. Theoretically, the INR eliminates
the differences in sensitivity of various PT reagents. However, the INR may not elim-
inate all of the PT assay’s variables, requiring adjustment to make the reagent’s INR
more accurate with regard to a specific local instrument in a local laboratory.
23.4.4.2.4 Reversal
In anticipation of elective surgery and to reverse a mildly elevated INR, withholding
VKA for 3–4 days is indicated, perhaps complemented with the administration of
oral vitamin K (Keeling et al. 2011). In patients with life-threatening bleeding,
administration of vitamin K, fresh frozen plasma (FFP), prothrombin complex con-
centrates (PCC, including the 4 coagulation factors: II, VII, IX, and X), or even
recombinant activated factor VII (rfVIIa) may be justified (Wozniak et al. 2012).
Mechanism of Action
UFH produces its major anticoagulant effect by irreversibly inactivating thrombin
(IIa) and activated factor X (factor Xa) through an antithrombin-dependent mecha-
nism with an anti-factor Xa to anti-factor IIa ratio of 1:1. Antithrombin (AT) is a
native anticoagulant protein that inactivates several clotting factors (IXa, Xa, XIIa,
and thrombin). Only one third of an administered dose of UFH binds to AT, while
the remaining fraction has a minimal anticoagulant effect. The UFH–AT complex is
100–1,000 times more potent as an anticoagulant than AT alone (Hirsh et al. 2001).
The UFH–AT complex, through its action on thrombin, not only prevents fibrin
formation but also inhibits thrombin-induced activation of factors V, VIII, and IX as
well as platelets. Thereby, it prevents further growth and propagation of the throm-
bus, but it is unable to inactivate thrombin or factor Xa within a formed clot. In
vitro, the high-molecular-weight heparin fraction also binds to platelets and von
Willebrand factor; depending on the experimental settings, it can either induce or
inhibit platelet aggregation.
Pharmacokinetics
After subcutaneous (s.c.) injection, bioavailability is dose dependent, ranging
from 30 % at lower doses to as much as 70 % at higher doses, and its anticoagu-
lant effect lasts around 1–2 h. Heparin is mainly cleared via internalization in
endothelial cells and macrophages (heparinase and sulfatase), a small part being
cleared by the kidney.
Pharmacokinetic limitations of heparin are related to its propensity to bind to
positively charged surfaces (plastic tubing), macrophages, endothelial cells, osteo-
clasts, and circulating proteins; this results in a poorly predictable response.
Heparin’s half-life is prolonged with increasing dosages and in patients with renal
failure, whereas it can be decreased or increased in patients with liver impairment.
Importantly, acutely ill patients may require higher doses of UFH to achieve an
antithrombotic effect, given increased binding to inflammatory cells and proteins as
well as to deficient levels of AT (heparin resistance).
Monitoring
Anticoagulation with heparin can easily be monitored by measuring activated par-
tial thromboplastin time (aPTT) or anti-Xa activity and, in a cardiac surgery setting
or in a catheterization laboratory, by measuring activated clotting time (ACT).
Monitoring is not required for the purpose of thromboprophylaxis.
Reversal
Protamine sulfate is considered the treatment of choice for patients who develop
significant bleeding complications while on UFH. This basic protein 5-kDa cationic
polypeptide binds to negatively charged UFH, thereby neutralizing its antithrombin
effect while incompletely reversing factor Xa inhibition.
One UI of protamine neutralizes one UI of heparin. Potentially life-threatening
adverse effects such as hemodynamic collapse, bronchospasm, and pulmonary
hypertension may exceptionally occur in susceptible patients (prior exposure to
protamine, fish allergies, and vasectomy). With high doses of protamine, increased
23 Perioperative Thromboprophylaxis 403
Mechanism of Action
Compared with UFH, LMWH has a reduced ability to deactivate thrombin as it can-
not bind to AT and thrombin simultaneously. The anti-factor Xa to anti-factor IIa
ratio is between 2:1 and 4:1 (Hirsh et al. 2001). Compared with UFH, LMWH has
a smaller effect on platelet function and platelet adhesion, therefore interfering less
with primary hemostasis.
Pharmacokinetics
Because LMWH binds less to plasma proteins and cells, they exhibit a more predict-
able dose–response relationship. Following a single s.c. injection of LMWH, absorp-
tion is almost complete, plasma activity peaks after 3 h, the elimination half-life
approximates 4 h, whereas the mean residence time of anti-Xa activity is about 6 h,
404 M. Aldenkortt and M. Licker
independent of the dose (20–80 mg) (Turpie 1998). Following repeated s.c. doses,
there is no evidence of accumulation or alterations in distribution and clearance.
In pregnant women, LMWH does not cross the placental barrier, as detected by
anti-factor Xa activity. LMWH is weakly metabolized in the liver, and relatively small
amounts are eliminated by the kidneys. The dosage of LMWH should be adjusted to
body weight rather than on a fixed regimen, particularly in obese subjects.
Monitoring
Measurement of aPTT can only serve as an indicator of overdosage. Monitoring is
not necessary for the prevention of VTE nor is it generally in therapeutic anticoagu-
lation, except in three situations: renal failure, obesity, and pregnancy. In these
patients, the anti-Xa level should be monitored, with a target range of 0.5–1.1 U/ml.
Reversal
Protamine sulfate can be used to reverse the effects of LMWH (1:1 ratio) although
this antagonizing effect is neither complete nor predictable.
Pharmacokinetics
After s.c. application, fondaparinux exhibits almost 100 % bioavailability. The drug
is eliminated via renal filtration, and its half-life is about 17–21 h (Nagler et al.
2012). In vitro studies show no cross-reactivity with HIT antibodies. Given its pre-
dictable antithrombotic effects and prolonged half-life, fondaparinux can be admin-
istered safely once a day (2.5 mg).
Monitoring
Monitoring fondaparinux is not routinely necessary. In certain situations, such as
acute renal failure, a chromogenic anti-Xa heparin assay or a specific drug assay can
be performed (pentasaccharide target 0.14–0.19 mg/l).
23 Perioperative Thromboprophylaxis 405
Reversal
Protamine sulfate and FFP fail to antagonize fondaparinux-induced antithrombotic
effects, and there is limited evidence that recombinant factor VII could be used to
stop fondaparinux-related bleeding, although it was found to normalize lengthened
coagulation assays (Dzik 2012).
Pharmacokinetics
After s.c. administration, the absolute bioavailability of danaparoid sodium
approaches 100 %, and the time to reach peak plasma anti-Xa activity levels is
approximately 4–5 h (Wilde and Markham 1997). Elimination is mainly via renal
filtration and biological half-life activity approximates 25 h.
Monitoring
No routine monitoring is requested. Under special circumstances (i.e., renal failure),
the anti-Xa level should be determined.
Reversal
No agent can reverse the effect of danaparoid. In acute bleeding situations, plasma-
pheresis has been shown to effectively reduce the plasma anti-Xa levels (Dzik).
Pharmacokinetics
After s.c. administration, desirudin reaches maximum plasma concentrations after
1–3 h, has a terminal half-life of 2 h, and is primarily excreted by the kidneys (80–
90 %) (Graetz et al. 2011). After intravenous (i.v.) injection, bivalirudin has an
immediate onset of action with therapeutic ACT achieved within 5 min and a half-
life of 25 min. It is mainly cleared by proteolytic cleavage and hepatic metabolism;
20 % of the dose is eliminated by the kidneys.
Monitoring
For perioperative thromboprophylaxis, monitoring is not deemed necessary even in
patients with moderate renal impairment (ClCr of 30–60 ml/min). An aPTT assay
should be performed on patients with severe renal dysfunction and for therapeutic
anticoagulation (target aPTT ratio between 1.5 and 2.5). An alternative to aPTT is
the ecarin clotting time assay (ECT), which reflects anti-factor II activity.
Reversal
There is no specific antidote available. Hirudin-induced bleeding can be partially
reversed by PCC and hemofiltration, hollow-fiber filters, and high- and low-flux
polysulfone dialysers (Dzik).
these patients, after 8–12 days of treatment, desirudin (15 mg/day, started preopera-
tively) was found superior to UFH (5,000 UI s.c. three times daily, started preopera-
tively) or LMWH (40 mg enoxaparin s.c. once daily), while showing a similar
safety profile (Salazar et al. 2010; Eriksson et al. 1997a, b).
In 2005, the US Food and Drug Administration approved bivalirudin as an alter-
native anticoagulant to heparin in patients undergoing percutaneous coronary inter-
ventions. Dose adjustments of bivalirudin are necessary in patients with moderate
renal insufficiency, and it is contraindicated in severe renal impairment. Because of
its narrow therapeutic window and its potential for increased bleeding events, lepi-
rudin’s use as an anticoagulant is limited.
Pharmacokinetics
After oral administration, dabigatran has a low bioavailability (<10 %), independent
of the dose of the prodrug. The time to maximum plasma concentration is 1.25–
2.5 h, and its half-life is about 12–14 h (Schulman and Majeed 2012). The drug is
neither metabolized, induced, nor inhibited by cytochrome P450 drug-metabolizing
enzymes. Renal excretion is the primary elimination pathway. The remainder under-
goes conjugation with glucuronic acid to form acyl glucuronides, which are excreted
via the bile.
Monitoring
Given its predictable and consistent anticoagulant effects, treatment with dabigatran
does not require routine coagulation monitoring or dose titration. Thrombin time
(TT) is the assay most responsive to dabigatran in the clinically relevant plasma
concentration range, whereas the aPTT and PT are the least so. Longer blood coagu-
lation parameters (aPTT, PT,) occur in parallel with increasing concentrations of
dabigatran.
Reversal
No specific antidote exists. Blood products (FFP, PCC, platelets) remain the main-
stay of treatment in case of overdose or bleeding (Dzik). Maintaining enforced
diuresis and dialysis may be attempted as the drug binds weakly to plasma proteins
(Kaatz et al. 2012).
(150 and 220 mg once daily) initiated postoperatively was shown to be non-inferior
to s.c. enoxaparin sodium (40 mg once daily, initiated prior to surgery) with regard
to the incidence of total VTE events and all-cause mortality in patients undergoing
TKR or THR surgery (Burness and McKeage 2012). A meta-analysis of 14 studies
indicated that DTI (ximelagatran, dabigatran, and desirudin) were equally effective
to VKA and LMWH in the prevention of major VTE following major joint surgery.
However, they were associated with a higher mortality and more frequent bleeding
(except desirudin) (Salazar et al. 2010).
In North America and Europe, dabigatran is licensed for thromboprophylaxis in
total knee and hip arthroplasty (TKA and THA), whereas in Switzerland, it is exclu-
sively licensed for the prevention of stroke and systemic embolism in patients with
atrial fibrillation.
Pharmacokinetics
After oral ingestion (10 mg), rivaroxaban has an 80–100 % bioavailability, with
peak plasma concentrations reached in 2–4 h, and an elimination half-life of 5–9 h
or 9–13 h in the elderly (Weinz et al. 2009). Rivaroxaban is extensively bound to
plasma proteins, it undergoes metabolic degradation in the liver, and it is mainly
eliminated through renal filtration (Eriksson et al. 2009).
Monitoring
Monitoring the antithrombotic effect of rivaroxaban is unnecessary, and no assay
has yet been validated. It is of note that increasing plasma–drug concentrations cor-
relates with longer PT and the inhibition of factor Xa activity.
Reversal
PCC were found to neutralize the anticoagulant effect of rivaroxaban in healthy
volunteers. Likewise, partial reversal of longer PT and bleeding time was achieved
after the administration of rfVIIa (Eerenberg et al. 2011).
Pharmacokinetics
After oral administration, apixaban has 50 % bioavailability, with peak plasma con-
centration reached after 0.5–3 h, and an elimination half-life of 12–15 h if it is given
once daily or 8–11 h if given twice daily (Eriksson et al. 2009). The drug mainly
undergoes hepatic oxidation into a phenol compound and is excreted in the bile;
25 % is eliminated via the kidneys. It has a low potential for drug–drug
interaction.
The recommended dose is 2.5 mg orally, twice daily.
Monitoring
Apixaban does not interfere with platelet aggregation, while it shows a slight dose-
dependent lengthening of INR and aPTT. No routine monitoring is required for
thromboprophylactic purposes.
Reversal
There are no specific reversal agents. Guidelines merely recommend supportive
treatment in cases of drug-associated bleeding, including the administration of
blood products (FFP, PCC) and activated charcoal if drug ingestion was within the
last couple of hours (Dzik) (Kaatz et al. 2012).
The US Agency for Healthcare Research and Policy has ranked the prevention of
VTE as the most valuable patient safety practice, given its cost-effectiveness and
benefit–risk ratio. In many countries, thromboprophylaxis serves as a quality indi-
cator of hospital health care. However, there is still a large gap between official
recommendations and medical practice since compliance with guidelines varies
from 13.3 to 94 % (Gomez-Outes et al. 2012; Cohen et al. 2008). In addition,
although many at-risk patients do not receive adequate prophylaxis, there is also
evidence to suggest that low-risk patients being prescribed thromboprophylaxis
comprises another important problem (Bikdeli and Sharif-Kashani 2012).
Dissemination of information through professional and social networks will
enhance global awareness of the importance of VTE. Methods such as computer-
based decision systems and preprinted orders have been shown to be most effective in
optimizing compliance with thromboprophylactic guidelines. Finally, periodic audits
focusing on outcome data and treatment adhesion, with feedback to nursing and medi-
cal teams, may reinforce consistent use of VTE prophylaxis (Galante et al. 2012).
Since 2010, guidance on VTE prophylaxis in surgical patients has been issued by
a number of medical organizations: the ACCP (February 2012), the American
College of Physicians (ACP, November 2011), the Scottish Intercollegiate Guideline
Network (SIGN, December 2010), and the UK National Institute for Health and
Clinical Excellence (NICE, January 2010). Although these recommendations are
based upon data derived from large RCT, several issues deserve further clarification,
including the role of screening for asymptomatic DVT, the best timing for the initia-
tion of pharmacological prophylaxis, the optimal duration of prophylaxis in high-
risk patients, and the indications for newer anticoagulant agents.
The plethora of official recommendations can be confusing, if not overwhelm-
ing. For the sake of clarity, we summarize the available effective methods of throm-
boprophylaxis for specific surgical categories taking into account the risk of
bleeding and VTE (Table 23.5) (Darvall and Bradbury 2012; Falck-Ytter et al.
2012; Gould et al. 2012a; Muntz and Michota 2010; Deitelzweig et al. 2008).
In very-low-risk patients (expected incidence of VTE <0.5 %), early ambula-
tion following the intervention will suffice, with no need for any specific pharma-
cological or mechanical prophylaxis. In low-risk patients (VTE ~1.5 %), ECS or
IPC devices should be applied during the period of patient immobilization.
Moderate-risk patients (VTE ~3 %) should benefit from LMWH, UFH, or
mechanical prophylaxis. In high-risk patients (VTE ~6 %), dual prophylaxis
should be instituted with GES or IPC combined with LMWH or UFH. If concerns
are raised about the potentially devastating consequences of bleeding (e.g., hem-
orrhagic shock, intracranial or ocular surgery), then mechanical prophylaxis
should be used alone. For instance, for every 1,000 neurosurgical patients who
receive prophylactic doses of UFH or LMWH, 91 VTE events can be prevented
but at the expense of 7 intracranial hemorrhagic events and 28 more cases of
minor bleeding (Hamilton et al. 2011).
Table 23.5 Evidence-based guidelines for perioperative prophylaxis of venous thromboembolism
Grade of
Type of surgery Thromboprophylactic method evidence Duration of TP
Very-low-risk VTE <0.5 %
Caprini score 0 No prophylaxis, early ambulation 1B –
Same day surgery (hernia repair)
Orthopedic procedure <30 min
Repair of small fractures
Isolated lower leg injuries + immobilization Mechanical prophylaxis 2C
Low-risk VTE ~1.5 %
Caprini score 1–2 Mechanical prophylaxis 2C Hospital stay or until free ambulation
General surgery, gynecological surgery,
laparoscopic surgery
23 Perioperative Thromboprophylaxis
Grade of
Type of surgery Thromboprophylactic method evidence Duration of TP
High risk VTE ~6 %
General or abdominal, pelvic surgery UFH or LMWH 1B Start (2–12 h before or) 12–24 h postop,
Thoracic surgery + VTE risk factors UFH or LMWH + mechanical prophylaxis 2C ≥7–10 days, 30 days if cancer (1B)
(low bleeding risk) 2C
Neurosurgery + VTE risk factors Mechanical prophylaxis + LMWH or UFH (if low 2C Start 24–48 h postop, if low risk of
Spinal surgery + VTE risk factors bleeding risk) intracranial/spinal bleeding
Major trauma + VTE risk factors (e.g., Mechanical prophylaxis if no lower leg injury 2C Start 24–48 h postop, if low risk of
spinal/brain injury) Mechanical prophylaxis + UFH or LMWH (if low 2C intracranial/spinal bleeding
bleeding risk)
If UFH and LMWH are contraindicated, choose one of
following
Aspirin 2C
Fondaparinux 2C
Total hip or knee arthroplasty (THA, TKA) Consider one of the following options Start ≥12 h preop or ≥12 h postop
Hip fracture surgery (HFS) LMWH (UFH, adjusted-dose VKA or aspirin) 1B Extend prophylaxis to 10–35 days
(2B/2C) postop or until end of rehabilitation
Fondaparinux 1B, 2B during the hospital stay
Apixaban, rivaroxaban, or dabigatran 1B, 2B
Mechanical prophylaxis 1C
LMWH is preferred over other drugs 2C
Dual prophylaxis, mechanical + pharmacological
Major orthopedic surgery (e.g., HA, TKA, Consider one of the following options
HFS, spinal surgery)
+ patient uncooperative with injection or Apixaban or dabigatran 1B
mechanical devices Rivaroxaban or adjusted dose of VKA 1B
Major orthopedic surgery + risk of bleeding Consider one of the following options
Mechanical prophylaxis 2C
No prophylaxis
M. Aldenkortt and M. Licker
Mechanical prophylaxis: intermittent pneumatic compression device or elasticated compressive stockings (>18 h/day)
LMWH low-molecular-weight heparin, UFH unfractionated heparin, VKA vitamin K antagonist, TP thromboprophylaxis
23 Perioperative Thromboprophylaxis 413
In bariatric surgery, PE has been reported as the most common cause of postop-
erative death, accounting for 30 % of all deaths in the International Bariatric Surgery
Registry. Hence, the ACCP task force recommends the routine administration of
UFH, LMH, or fondaparinux, in combination with optimally used IPC. Weight-
adjusted doses of antithrombotic drugs should be prescribed in order to achieve a
consistent VTE risk reduction, although the incidence of major bleeding increases
in parallel (Becattini et al. 2012).
In major orthopedic, abdominal, or pelvic surgery, prophylaxis with UFH,
LMWH, VKA, or fondaparinux should be continued beyond the hospital stay
(4 weeks instead of 5–7 days) or until appropriate recovery of function (Kakkar
et al. 2008; Rasmussen et al. 2009). Special attention must be given to elderly
patients due to their higher prevalence of comorbidities, renal dysfunction, and
impaired functional capacity. Peptic ulcer, hemophilia, or von Willebrand disease,
as well as the use of antiplatelet drugs, makes thromboprophylaxis a particular chal-
lenge. Moreover, central neuraxial blockade should be performed to a dedicated
time frame: a spinal or epidural needle should only be inserted 8–12 h after the last
s.c. dose of UFH or 18 h after a once daily dose of LMWH (Gogarten et al. 2010;
Horlocker et al. 2010a).
Conclusions
Arterial and venous thromboembolic events commonly occur after surgery and
cause potentially preventable morbidity and mortality. Current evidence-based
management strategies have significantly reduced the incidence of VTE, although
there is room for further improvements with the administration of safer anti-
thrombotic agents and the implementation of specific guidelines coupled with
rigorous clinical audits.
References
Akl EA, Terrenato I et al (2008) Low-molecular-weight heparin vs unfractionated heparin for
perioperative thromboprophylaxis in patients with cancer: a systematic review and meta-
analysis. Arch Intern Med 168(12):1261–1269
Astrup T (1958) The haemostatic balance. Thromb Diath Haemorrh 2(3–4):347–357
Bahl V, Hu M, Henke PK (2010) A validation study of a retrospective venous thromboembolism
risk scoring method. Ann Surg 251(2):344–350
Baser O, Supina D et al (2011) Clinical and cost outcomes of venous thromboembolism in
Medicare patients undergoing total hip replacement or total knee replacement surgery. Curr
Med Res Opin 27(2):423–429
Bauer KA et al (2011) Recent progress in anticoagulant therapy: oral direct inhibitors of thrombin
and factor Xa. J Thromb Haemost 9(Suppl 1):12–19
Becattini C, Agnelli G et al (2012) Venous thromboembolism after laparoscopic bariatric surgery
for morbid obesity: clinical burden and prevention. Surg Obes Relat Dis 8(1):108–115
Bikdeli B, Sharif-Kashani B (2012) Prophylaxis for venous thromboembolism: a great global
divide between expert guidelines and clinical practice? Semin Thromb Hemost 38(2):
144–155
Budnitz DS, Pollock DA et al (2006) National surveillance of emergency department visits for
outpatient adverse drug events. JAMA 296(15):1858–1866
414 M. Aldenkortt and M. Licker
Burness CB, McKeage K (2012) Dabigatran etexilate: a review of its use for the prevention of
venous thromboembolism after total hip or knee replacement surgery. Drugs 72(7):963–986
Caprini JA, Arcelus JI et al (2001) Effective risk stratification of surgical and nonsurgical patients
for venous thromboembolic disease. Semin Hematol 38(2 Suppl 5):12–19
Carlquist JF, Anderson JL (2011) Using pharmacogenetics in real time to guide warfarin initiation:
a clinician update. Circulation 124(23):2554–2559
Chee YL, Crawford JC et al (2008) Guidelines on the assessment of bleeding risk prior to surgery
or invasive procedures. British Committee for Standards in Haematology. Br J Haematol
140(5):496–504
Cohen AT, Alikhan R et al (2005) Assessment of venous thromboembolism risk and the benefits
of thromboprophylaxis in medical patients. Thromb Haemost 94(4):750–759
Cohen AT, Tapson VF et al (2008) Venous thromboembolism risk and prophylaxis in the acute
hospital care setting (ENDORSE study): a multinational cross-sectional study. Lancet
371(9610):387–394
Darvall K, Bradbury A (2012) Pathways for venous thromboembolic prophylaxis in medical and
surgical patients. Phlebology 27(Suppl 2):33–42
Deitelzweig SB, McKean SC et al (2008) Prevention of venous thromboembolism in the orthope-
dic surgery patient. Cleve Clin J Med 75(Suppl 3):S27–S36
Delis KT, Knaggs AL et al (2004) Effects of epidural-and-general anesthesia combined versus
general anesthesia alone on the venous hemodynamics of the lower limb. A randomized study.
Thromb Haemost 92(5):1003–1011
Dobesh PP (2009) Economic burden of venous thromboembolism in hospitalized patients.
Pharmacotherapy 29(8):943–953
Douketis JD, Spyropoulos AC et al (2012) Perioperative management of antithrombotic therapy:
antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest
Physicians Evidence-Based Clinical Practice Guidelines. Chest 141(2 Suppl):e326S–e350S
Duran A, Sengupta A et al (2012) Cost effectiveness of rivaroxaban versus enoxaparin for preven-
tion of post-surgical venous thromboembolism from a U.S. payer’s perspective.
Pharmacoeconomics 30(2):87–101
Dzik WS (2012) Reversal of drug-induced anticoagulation: old solutions and new problems.
Transfusion 52(Suppl 1):45S–55S
Eerenberg ES, Kamphuisen PW et al (2011) Reversal of rivaroxaban and dabigatran by prothrom-
bin complex concentrate: a randomized, placebo-controlled, crossover study in healthy
subjects. Circulation 124(14):1573–1579
Eriksson BI, Ekman S et al (1997a) Prevention of thromboembolism with use of recombinant
hirudin. Results of a double-blind, multicenter trial comparing the efficacy of desirudin
(Revasc) with that of unfractionated heparin in patients having a total hip replacement. J Bone
Joint Surg Am 79(3):326–333
Eriksson BI, Wille-Jorgensen P et al (1997b) A comparison of recombinant hirudin with a low-
molecular-weight heparin to prevent thromboembolic complications after total hip replace-
ment. N Engl J Med 337(19):1329–1335
Eriksson BI, Quinlan DJ et al (2009) Comparative pharmacodynamics and pharmacokinetics of
oral direct thrombin and factor Xa inhibitors in development. Clin Pharmacokinet 48(1):
1–22
Falck-Ytter Y, Francis CW et al (2012) Prevention of VTE in orthopedic surgery patients: anti-
thrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines. Chest 141(2 Suppl):e278S–e325S
Fareed J, Thethi I et al (2012) Old versus new oral anticoagulants: focus on pharmacology. Annu
Rev Pharmacol Toxicol 52:79–99
Galante M, Languasco A et al (2012) Venous thromboprophylaxis in general surgery ward admis-
sions: strategies for improvement. Int J Qual Health Care 24(6):649–656
Geerts WH, Bergqvist D et al (2008) Prevention of venous thromboembolism: American College
of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 133(6
Suppl):381S–453S
23 Perioperative Thromboprophylaxis 415
Geraghty AJ, Welch K (2011) Antithrombotic agents for preventing thrombosis after infrainguinal
arterial bypass surgery. Cochrane Database Syst Rev (6):CD000536
Gogarten W, Vandermeulen E et al (2010) Regional anaesthesia and antithrombotic agents: recom-
mendations of the European Society of Anaesthesiology. Eur J Anaesthesiol 27(12):
999–1015
Gomez-Outes A, Terleira-Fernandez AI et al (2012) Dabigatran, rivaroxaban, or apixaban versus
enoxaparin for thromboprophylaxis after total hip or knee replacement: systematic review,
meta-analysis, and indirect treatment comparisons. BMJ 344:e3675
Gould MK, Garcia DA et al (2012a) Prevention of VTE in nonorthopedic surgical patients: anti-
thrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines. Chest 141(2 Suppl):e227S–e277S
Gould MK, Garcia DA et al (2012b) Prevention of VTE in nonorthopedic surgical patients: anti-
thrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines. Chest 141(2 Suppl):e227S–e277S
Graetz TJ, Tellor BR et al (2011) Desirudin: a review of the pharmacology and clinical application
for the prevention of deep vein thrombosis. Expert Rev Cardiovasc Ther 9(9):1101–1109
Greinacher A, Warkentin TE (2008) The direct thrombin inhibitor hirudin. Thromb Haemost
99(5):819–829
Hahnenkamp K, Theilmeier G et al (2002) The effects of local anesthetics on perioperative coagu-
lation, inflammation, and microcirculation. Anesth Analg 94(6):1441–1447
Hamilton MG, Yee WH et al (2011) Venous thromboembolism prophylaxis in patients undergoing
cranial neurosurgery: a systematic review and meta-analysis. Neurosurgery 68(3):571–581
Heron R, Davie A et al (2001) Interrater reliability of the Glasgow Coma Scale scoring among
nurses in sub-specialties of critical care. Aust Crit Care 14(3):100–105
Hirsh J, Warkentin TE et al (2001) Heparin and low-molecular-weight heparin: mechanisms of
action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 119(1 Suppl):
64S–94S
Horlocker TT, Wedel DJ et al (2010) Regional anesthesia in the patient receiving antithrombotic or
thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine Evidence-
Based Guidelines (Third Edition). Reg Anesth Pain Med 35(1):64–101
Huber O, Bounameaux H et al (1992) Postoperative pulmonary embolism after hospital discharge.
An underestimated risk. Arch Surg 127(3):310–313
Januel JM, Chen G et al (2012) Symptomatic in-hospital deep vein thrombosis and pulmonary
embolism following hip and knee arthroplasty among patients receiving recommended prophy-
laxis: a systematic review. JAMA 307(3):294–303
Junqueira DR, Perini E et al (2012) Unfractionated heparin versus low molecular weight heparin
for avoiding heparin-induced thrombocytopenia in postoperative patients. Cochrane Database
Syst Rev (9):CD007557
Kaatz S, Kouides PA et al (2012) Guidance on the emergent reversal of oral thrombin and factor
Xa inhibitors. Am J Hematol 87(Suppl 1):S141–S145
Kakkar VV, Spindler J et al (1972) Efficacy of low doses of heparin in prevention of deep-vein
thrombosis after major surgery: a double-blind, randomised trial. Lancet 300(7768):101–106
Kakkar AK, Brenner B et al (2008) Extended duration rivaroxaban versus short-term enoxaparin
for the prevention of venous thromboembolism after total hip arthroplasty: a double-blind,
randomised controlled trial. Lancet 372(9632):31–39
Kakkos SK, Caprini JA et al (2008) Combined intermittent pneumatic leg compression and phar-
macological prophylaxis for prevention of venous thromboembolism in high-risk patients.
Cochrane Database Syst Rev (4):CD005258
Kakkos SK, Warwick D et al (2012) Combined (mechanical and pharmacological) modalities for
the prevention of venous thromboembolism in joint replacement surgery. J Bone Joint Surg Br
94(6):729–734
Kanchanabat B, Stapanavatr W et al (2011) Systematic review and meta-analysis on the rate of
postoperative venous thromboembolism in orthopaedic surgery in Asian patients without
thromboprophylaxis. Br J Surg 98(10):1356–1364
416 M. Aldenkortt and M. Licker
Keeling D, Baglin T et al (2011) Guidelines on oral anticoagulation with warfarin – fourth edition.
Br J Haematol 154(3):311–324
Khaw FM, Moran CG et al (1993) The incidence of fatal pulmonary embolism after knee replace-
ment with no prophylactic anticoagulation. J Bone Joint Surg Br 75(6):940–941
Kucher N, Koo S et al (2005) Electronic alerts to prevent venous thromboembolism among hospi-
talized patients. N Engl J Med 352(10):969–977
Mauermann WJ, Shilling AM et al (2006) A comparison of neuraxial block versus general anes-
thesia for elective total hip replacement: a meta-analysis. Anesth Analg 103(4):1018–1025
Mismetti P, Laporte S et al (2004) Prevention of venous thromboembolism in orthopedic surgery
with vitamin K antagonists: a meta-analysis. J Thromb Haemost 2(7):1058–1070
Morris RJ, Woodcock JP (2010) Intermittent pneumatic compression or graduated compression
stockings for deep vein thrombosis prophylaxis? A systematic review of direct clinical com-
parisons. Ann Surg 251(3):393–396
Muntz JE, Michota FA (2010) Prevention and management of venous thromboembolism in the
surgical patient: options by surgery type and individual patient risk factors. Am J Surg 199(1
Suppl):S11–S20
Muntz J, Scott DA et al (2004) Major bleeding rates after prophylaxis against venous thromboem-
bolism: systematic review, meta-analysis, and cost implications. Int J Technol Assess Health
Care 20(4):405–414
Nagler M, Haslauer M et al (2012) Fondaparinux – data on efficacy and safety in special situations.
Thromb Res 129(4):407–417
Neumann I, Rada G et al (2012) Oral direct Factor Xa inhibitors versus low-molecular-weight
heparin to prevent venous thromboembolism in patients undergoing total hip or knee replace-
ment: a systematic review and meta-analysis. Ann Intern Med 156(10):710–719
Oger E (2000) Incidence of venous thromboembolism: a community-based study in Western
France. EPI-GETBP Study Group. Groupe d’Etude de la Thrombose de Bretagne Occidentale.
Thromb Haemost 83(5):657–660
Ollendorf DA, Vera-Llonch M et al (2002) Cost of venous thromboembolism following major
orthopedic surgery in hospitalized patients. Am J Health Syst Pharm 59(18):1750–1754
Pengo V, Lensing AW et al (2004) Incidence of chronic thromboembolic pulmonary hypertension
after pulmonary embolism. N Engl J Med 350(22):2257–2264
Petitou M, Nancy-Portebois V et al (2009) From heparin to EP217609: the long way to a new
pentasaccharide-based neutralisable anticoagulant with an unprecedented pharmacological
profile. Thromb Haemost 102(5):804–810
Phillips SM, Gallagher M et al (2008) Use graduated compression stockings postoperatively to
prevent deep vein thrombosis. BMJ 336(7650):943–944
Poultsides LA, GonzalezDellaValle A et al (2012) Meta-analysis of cause of death following total
joint replacement using different thromboprophylaxis regimens. J Bone Joint Surg Br
94(1):113–121
Rahn DD, Mamik MM et al (2011) Venous thromboembolism prophylaxis in gynecologic surgery:
a systematic review. Obstet Gynecol 118(5):1111–1125
Rasmussen MS, Jorgensen LN et al (2009) Prolonged thromboprophylaxis with low molecular
weight heparin for abdominal or pelvic surgery. Cochrane Database Syst Rev (1):CD004318
Roderick P, Ferris G et al (2005) Towards evidence-based guidelines for the prevention of venous
thromboembolism: systematic reviews of mechanical methods, oral anticoagulation, dextran
and regional anaesthesia as thromboprophylaxis. Health Technol Assess 9(49):iii–iv, ix–x,
1–78
Rogers SO Jr, Kilaru RK et al (2007) Multivariable predictors of postoperative venous thrombo-
embolic events after general and vascular surgery: results from the patient safety in surgery
study. J Am Coll Surg 204(6):1211–1221
Salazar CA, Malaga G et al (2010) Direct thrombin inhibitors versus vitamin K antagonists or low
molecular weight heparins for prevention of venous thromboembolism following total hip or
knee replacement. Cochrane Database Syst Rev (4):CD005981
23 Perioperative Thromboprophylaxis 417
Schenone M, Furie BC et al (2004) The blood coagulation cascade. Curr Opin Hematol
11(4):272–277
Schulman S, Majeed A (2012) The oral thrombin inhibitor dabigatran: strengths and weaknesses.
Semin Thromb Hemost 38(1):7–15
Shantsila E, Lip GY et al (2009) Heparin-induced thrombocytopenia. A contemporary clinical
approach to diagnosis and management. Chest 135(6):1651–1664
Stringer MD, Steadman CA et al (1989) Deep vein thrombosis after elective knee surgery. An
incidence study in 312 patients. J Bone Joint Surg Br 71(3):492–497
Tanaka KA, Key NS et al (2009) Blood coagulation: hemostasis and thrombin regulation. Anesth
Analg 108(5):1433–1446
Turpie AG (1998) Pharmacology of the low-molecular-weight heparins. Am Heart J 135(6 Pt 3
Su):S329–S335
Turpie AGG, Bauer KA et al (2002) Fondaparinux vs enoxaparin for the prevention of venous
thromboembolism in major orthopedic surgery: a meta-analysis of 4 randomized double-blind
studies. Arch Intern Med 162(16):1833–1840
Urbankova J, Quiroz R et al (2005) Intermittent pneumatic compression and deep vein thrombosis
prevention. A meta-analysis in postoperative patients. Thromb Haemost 94(6):1181–1185
Van De Car DA, Rao SV et al (2010) Bivalirudin: a review of the pharmacology and clinical appli-
cation. Expert Rev Cardiovasc Ther 8(12):1673–1681
Weinz C, Schwarz T et al (2009) Metabolism and excretion of rivaroxaban, an oral, direct factor
Xa inhibitor, in rats, dogs, and humans. Drug Metab Dispos 37(5):1056–1064
White RH, Zhou H et al (2003) Incidence of symptomatic venous thromboembolism after different
elective or urgent surgical procedures. Thromb Haemost 90(3):446–455
White RH, Chew H et al (2007) Targeting patients for anticoagulant prophylaxis trials in patients
with cancer: who is at highest risk? Thromb Res 120(Suppl 2):S29–S40
Wilde MI, Markham A (1997) Danaparoid. A review of its pharmacology and clinical use in the
management of heparin-induced thrombocytopenia. Drugs 54(6):903–924
Windisch C, Kolb W et al (2011) Pneumatic compression with foot pumps facilitates early postop-
erative mobilisation in total knee arthroplasty. Int Orthop 35(7):995–1000
Wolberg AS, Aleman MM et al (2012) Procoagulant activity in hemostasis and thrombosis:
Virchow’s triad revisited. Anesth Analg 114(2):275–285
Wozniak M, Kruit A et al (2012) Prothrombin complex concentrate for the urgent reversal of
warfarin. Assessment of a standard dosing protocol. Transfus Apher Sci 46(3):309–314
Part IV
Economic Aspects and Organization
Economic Aspects and Organization
24
Klaus Görlinger and Sibylle A. Kozek-Langenecker
K. Görlinger (*)
Department of Anaesthesiology and Intensive Care Medicine, University Hospital Essen,
University of Duisburg-Essen, Duisburg-Essen, Germany
e-mail: [email protected], [email protected]
S.A. Kozek-Langenecker
Department of Anaesthesia and Intensive Care, Evangelical Hospital Vienna, Vienna, Austria
Author (year) Country Cost calculation PRBC FFP Platelets Cryoprecipitate Fibrinogen PCC rFVIIa
Varney and Guest UK Mean activity- £635 (€1,031) £378 (€614) £347 (€564) £834 (€1,354) – – –
(2003) based costs (NHS
costs)
Glenngård et al. Sweden Societal costs €702 for 2 filtered allogeneic PRBC units – – – –
(2005) (Transfusion reactions accounted for 35 % of
costs)
€598 for 2 autologous PRBC units – – – –
€285 for intraoperative PRBC salvage (>4 – – – –
units)
Agrawal et al. UK Mean activity- £546.12 (€804.86) for transfusion of 2 PRBC units:
(2006) based costs Mean staff costs £37.24 (blood bank £9.68 + ward £27.56); mean costs of disposables £13.25; mean
Economic Aspects and Organization
costs for blood products £287.56; mean costs of wastage £11.86; other derived cost (e.g., hospital stay)
£196.21
Murphy et al. UK Acquisition costs £135.50 £31.50 (€46) £214 (€315) – – – –
(2007) (€200)
Shander et al. USA and Mean acquisition $248.18 in Englewood Hospital Medical Center, Englewood, NJ, USA
(2010) Europe costs $203.47 in Rhode Island Hospital, Providence, RI, USA
$193.70 in Centre Hospitalier Univers. Vaudois, Lausanne, Switzerland
$153.72 in General Hospital Linz, Austria
USA and Mean activity- $1,183.32 in Englewood Hospital Medical Center, Englewood, NJ, USA
Europe based costs $726.05 in Rhode Island Hospital, Providence, RI, USA
$611.44 in Centre Hospitalier Univers. Vaudois, Lausanne, Switzerland
$522.45 in General Hospital Linz, Austria
(continued)
423
Table 24.1 (continued)
424
Author (year) Country Cost calculation PRBC FFP Platelets Cryoprecipitate Fibrinogen PCC rFVIIa
Toner et al. (2011) US Mean acquisition $210.74 ± 37.9 $60.70 ± 20 $533.90 ± 69 – – – –
costs (apheresis)
Median costs for $50.00 ± 120 – – – – – –
mandated onsite per unit
screening
Median storage $68.00± 81 – – – – – –
and retrieval costs per unit
Mean charge to $343.63 ± 135 – – – – – –
patient per unit
Görlinger et al. Germany Virtual internal €85 €65 €250 (pooled – €288/g €126 per €3,203 per 4.8
(2011a, b) and transfer prices platelet conc.) 500 IU mg (240 KIU)
Hanke et al. (2012)
Weber et al. (2012) Germany Acquisition costs €72 0.162 €/g or €231 (pooled – €233 per g €114 per €2,784 per 4.8
€40.50 per platelet conc.) 600 IU mg (240 KIU)
250 ml unit
Abraham and Sun Western Mean activity- €877.69 for 2 – – – – – –
(2012) Europe based costs units
K. Görlinger and S.A. Kozek-Langenecker
Table 24.2 Additional length of stay (LOS) at intensive care unit (ICU), high-dependency unit (HDU), and at hospital (HP), and incremental hospital costs
24
due to bleeding complications in major surgery and acute coronary syndromes (ACS)
Number of Patients with Additional length of stay at ICU/ Incremental costs per
Author (year) Clinical setting, country patients (n) bleeding (%) hospital (HP) (days) hospitalization ($)
Murphy et al. (2007) Cardiac surgery, UK 8,598 57.1 % transfused Hazard ratio for discharge from ICU/ RBC units – relative increase
patients HDU, 0.69 (95 % CI, 0.65–0.72) and in costs; mean (95 % CI)
Any: 57.1 % from HP, 0.63 (95 % CI, 0.60–0.67) at Any: 1.42 (1.37–1.46)
1 unit: 13.6 % any postoperative time 1: 1.11 (1.08–1.14)
2 units: 14.5 % 2: 1.21 (1.18–1.25)
3–4 units:15.2 % 3–4: 1.41 (1.36–1.46)
5–9 units:10.0 % 5–9: 1.81 (1.71–1.90)
>9 units: 3.8 % >9: 3.35 (3.03–3.70)
Rao et al. (2008) Non-ST-segment elevation 1,235 36.8 % Unadjusted
Economic Aspects and Organization
Both bleeding and allogeneic blood transfusion have been shown to be associated
with an increased incidence of ischemic and thromboembolic events, as well as
in-hospital and posthospital costs (Dobesh 2009; Ruppert et al. 2011; Sharples
et al. 2006; Spiess et al. 2004; Vekeman et al. 2011). Therefore, hemostatic inter-
ventions that can effectively stop microvascular bleeding without increasing the
incidence of ischemic and thromboembolic events are very likely to substantially
improve cost-effectiveness.
In recent decades, several attempts have been made to find a universal hemo-
static agent capable of ensuring hemostasis during and after major surgery and
trauma, independent of the individual cause of the bleeding. Almost all the drugs
studied in this context either failed to reduce bleeding and transfusion require-
ments if given as prophylaxis or were associated with severe adverse events,
such as acute renal failure or thrombotic/thromboembolic events and even
increased mortality.
24 Economic Aspects and Organization 427
24.4.1 Antifibrinolytics
Aprotinin was withdrawn from the market in November 2007 due to the reported
increased incidence of renal dysfunction and mortality in cardiac surgery patients,
compared to treatment with lysine analogues (Brown et al. 2007; Fergusson et al.
2008; Mangano et al. 2006). Lysine analogues (tranexamic acid or TXA, and
ε-aminocaproic acid) reduce perioperative blood loss and transfusion requirements
and can be highly cost-effective in major orthopedic surgery, cardiac surgery, post-
partum hemorrhage, liver resection and transplantation, and trauma (CRASH-2
Trial C2010; CRASH-2 Trial Collaborators 2011; Ducloy-Bouthors et al. 2011;
Fergusson et al. 2008; Ferrer et al. 2009; Goobie et al. 2011; Greiff et al. 2012;
Gurusamy et al. 2009; Henry et al. 2011; Ickx et al. 2006; Martin et al. 2011;
Molenaar et al. 2007; Novikova and Hofmeyer 2010; Sander et al. 2010; Sukeik
et al. 2011; Rajesparan et al. 2009; Tzortzopoulou et al. 2008). Notably, in the
CRASH-2 study, a reduction in mortality was only observed if TXA was adminis-
tered within 3 h of a trauma. In contrast, prophylactic administration of TXA more
than 3 h after trauma was associated with increased mortality (CRASH-2 Trial
Collaborators 2011). The timing of TXA administration therefore seems to be cru-
cial. During liver transplantation, a targeted therapy of hyperfibrinolysis also pro-
duced a similar reduction of transfusion requirements, which was detected using
viscoelastic tests (thromboelastometry/thromboelastography) (Ickx et al. 2006;
Görlinger et al. 2010). During liver transplantation, therefore, the efficacy and
safety of a prophylactic administration of lysine analogues, compared to a targeted
therapeutic intervention, is still being debated (Schofield et al. 2012). A large, inter-
national, prospective, randomized clinical trial looking at the outcome effects of
TXA in 15,000 women with postpartum hemorrhage is ongoing (Shakur et al.
2010).
However, administration of TXA in patients undergoing major surgery has been
shown to reduce transfusion requirements cost-effectively without significantly
increasing the incidence of deep vein thrombosis (Rajesparan et al. 2009).
Particularly in countries with limited financial resources, lysine analogues have
been shown to significantly save costs and lives (Guerriero et al. 2010). Indeed,
cost-effectiveness analysis based on the CRASH-2 trial data showed that an early
administration of TXA to bleeding trauma patients is likely to be highly cost-
effective in low, middle, and high income settings (Guerriero et al. 2011).
According to current literature, the use of recombinant activated factor VII (rFVIIa)
should be restricted to its licensed indications since outside these indications its
effectiveness in reducing transfusion requirements and mortality remains unproven,
but the risks of arterial thromboembolic events and costs are high. All prospective
randomized trials dealing with the prophylactic administration of rFVIIa have failed
to show any effect on mortality (Chavez-Tapia et al. 2011; Dutton et al. 2009;
428 K. Görlinger and S.A. Kozek-Langenecker
Hauser et al. 2010; Levi 2010; Kozek-Langenecker et al. 2013; Simpson et al.
2012). A reduction in the number of transfused packed red blood cells (PRBCs) by
2.6 U and a reduction in the need for massive transfusion (14 % versus 33 %) could
only be demonstrated in severe blunt trauma (Boffard et al. 2005). However, a sec-
ond trauma study, powered for a reduction in mortality, was terminated early since
statistical significance could not be achieved (Dutton et al. 2009; Hauser et al.
2010). From a pharmacoeconomic point of view, the cost of 400 μg/kg rFVIIa
(about EUR 20,000) is extremely high compared to a reduction in transfusion
requirements of 2.6 U of PRBC. Indeed, a cost-effectiveness analysis of using
rFVIIa as an off-label rescue treatment for critical bleeding requiring massive trans-
fusion was recently published. The total cost per life-year gained through massive
transfusion was USD 1,148,000 (95 % CI: USD 825,000–1,471,000), and the incre-
mental cost of rFVIIa as part of that life-saving treatment was USD 736,000 (95 %
CI: USD 527,000–945,000) (Ho and Litton 2012). The incremental costs of rFVIIa
were much greater than the usual acceptable cost-effective limit (< USD 100,000
per life-year) for most patients with critical bleeding. Furthermore, two prospective
randomized trials in patients with intracerebral hemorrhage observed a significantly
increased incidence of arterial thromboembolic complications, including myocar-
dial and cerebral infarction (7 % versus 2 % (p = 0.12) and 10 % versus 1 % (p = 0.01),
respectively) (Mayer et al. 2005; Sugg et al. 2006). A distinct trend to more criti-
cally serious (thromboembolic) adverse events, including stroke, has also been
observed in a prospective randomized study of liver transplantation (placebo 10 %;
60 μg/kg rFVIIa 19 %; 120 μg/kg rFVIIa 12 %; p > 0.05) and cardiac surgery (pla-
cebo 7 %; 40 μg/kg rFVIIa 14 % (p = 0.25); 80 μg/kg rFVIIa 12 % (p = 0.43)) (Gill
et al. 2009; Lodge et al. 2005). Furthermore, the risk of arterial thrombotic compli-
cations (in particular stroke and myocardial infarction) after the “off-label use” of
rFVIIa has been highlighted in several other publications (Howes et al. 2009; Levi
et al. 2010; O’Connell et al. 2006; Simpson et al. 2012). Finally, two recently pub-
lished studies showed that rFVIIa had minimal clinical impact on outcomes for
patients requiring less than 30 units of PRBCs. For patients transfused more than 30
units of PRBCs, differences in 24-h and 30-day mortality suggest that rFVIIa con-
verted early deaths from exsanguination to late deaths from multiple organ failure
(Morse et al. 2011; Nascimento et al. 2011). Therefore, in accordance with the
manufacturer’s recommendations, most recent guidelines recommend not to use
rFVIIa in non-licensed indications (AAGBI 2010; BGMA 2009a; Kozek-
Langenecker et al. 2013; Lin et al. 2012; Rossaint et al. 2010; Stürmer et al. 2011).
salvage to be EUR 285 per transfusion (>4 units). This was comparatively much
lower than the cost of a 2-unit transfusion of filtered allogeneic PRBCs, which was
EUR 598. The administrative costs of preoperative autologous blood donation
were found to be much higher due to the higher logistic and personnel expenses
(Glenngård et al. 2005).
for allogeneic blood products and coagulation factor concentrates. Balanced against
this were additional costs for thromboelastometry (depreciation costs of three
ROTEM® devices, disposables, and reagents for 2,400 ROTEM® tests per year)
amounting to EUR 16,500 (Görlinger et al. 2010). Over an observation period of ten
years (covering 21,814 visceral surgeries including 1,105 liver transplantations), the
total cost saving amounted to EUR 1.6 million (EUR 1,765,280 of savings on allo-
geneic blood products and coagulation factor concentrates and EUR 165,000 of
additional costs for thromboelastometry) (Goerlinger et al. 2010). Activity-based
costs and secondary costs induced by transfusion-associated adverse events were
not considered in these costs analyses. Similar results have been published by other
groups (Noval-Padillo et al. 2010; Trzebicki et al. 2010; Wang et al. 2010).
Thromboelastometry-based, goal-directed therapy with fibrinogen concentrate and
PCC was not associated with an increased incidence of thromboembolic events
(Kirchner et al. 2012). This is in line with other publications reporting on the low
thrombotic risk of fibrinogen concentrate and PCC, so long as an overdose is
avoided (Grottke et al. 2011; Hanke et al. 2013; Kozek-Langenecker et al. 2011;
Majeed et al. 2012; Manco-Johnson et al. 2009; Sørensen et al. 2011; Warmuth
et al. 2012). Therefore, increased thrombosis-related costs are not to be expected.
2011a; Hanke et al. 2012; Weber et al. 2012), and hospital costs (Görlinger et al.
2011a; Hanke et al. 2012; Spalding et al. 2007; Spiess et al. 1995; Weber et al.
2012). Not one study demonstrated increased hospital costs. Off-label use of
rFVIIa – integrated into some algorithms as a rescue therapy if the algorithm’s
hemostatic therapy has failed – was almost completely eliminated in five studies
(Girdauskas et al. 2010; Görlinger et al. 2011a; Hanke et al. 2012; Spalding et al.
2007; Weber et al. 2012). This was certainly important to the cost savings in these
studies and potentially also for a reduction in the incidence of thrombotic/throm-
boembolic events. Only the four most recent studies performed, between 2010 and
2012, demonstrated an actual improvement in patient outcomes in the POC group
(Girdauskas et al. 2010; Görlinger et al. 2011a; Hanke et al. 2012; Weber et al.
2012). These studies used a similar POC coagulation and transfusion management
algorithm, first published in 2007, based on first-line, goal-directed therapy with
fibrinogen concentrate and four-factor PCCs guided by thromboelastometry
(Görlinger et al. 2007). Two studies additionally used whole blood impedance
aggregometry (Görlinger et al. 2011a; Weber et al. 2012). Three of them demon-
strated a reduction in thrombotic/thromboembolic events (Görlinger et al. 2011a;
Hanke et al. 2012; Weber et al. 2012). Furthermore, the randomized clinical trial in
coagulopathic patients undergoing complex cardiac surgery recently published by
Weber et al. (2012) showed significant reductions in postoperative pulmonary dys-
function, postoperative ventilation time, stay in ICU, composite adverse events
(acute renal failure, sepsis, thrombotic complications, and allergic reactions) (8 %
versus 38 %; p < 0.001), and six-month mortality (4 % versus 20 %; p = 0.013).
Since this single-center study was not set up to look at mortality and safety, the
results will have to be confirmed by adequately powered multicenter RCTs and
huge prospective observational studies.
Clinical setting, Study type, number of Costs for POC ICU LOS Mean reason for cost
Author (year) country patients POC device Cost saving diagnostics (calculated costs) saving
Spiess et al. Cardiac surgery, Before-and-after; 1,079 TEG Cost saving not NA NA Decreased transfusion
(1995) USA patients specified requirement
Spalding et al. Cardiac surgery, Before-and-after; 1,422 ROTEM €51,000 per €1,580 per Decreased PRBC,
(2007) Germany patients month (−44 %) month (3.1 % of platelets, PCC, FXIII,
cost saving and rFVIIa
Görlinger et al. University Before-and-after (2007 ROTEM €1,064,073 per €24,000 (2.3 % NA Decrease FFP and
(2008) Hospital, versus 2004); about year of cost saving) PRBC transfusion
Germany 50,000
Goerlinger Visceral surgery Before-and-after (2009 ROTEM €270,167 per €16,500 (6.1 % NA Decrease FFP, PRBC,
et al. (2010) and LTX, versus 1999); about year (−36 %) of cost saving) and platelet
Germany 4,800 patients incl. 240 transfusion
LTX
Görlinger et al. Visceral surgery Before-and-after ROTEM €1,765,280 in €165,000 (9.3 % NA Decrease FFP, PRBC,
(2010) and LTX, (1999–2009); 21,814 10 years of cost saving) and platelet
Germany patients incl. 1,105 LTX transfusion
Görlinger et al. Cardiac surgery, Before-and-after (2009 ROTEM and €50,000 per NA NA Decrease FFP and
(2011a, b) Germany versus 2005); 3,865 Multiplate year (−6.5 %) PRBC transfusion
patients
Hanke et al. Type A aortic Before-and-after; 10 ROTEM €2,757 per case NA −5.8 days Decreased FFP and
(2012) dissection, patients (×2,980 PRBC transfusion
Germany €a = €17,284)
Weber et al. Complex cardiac RCT; 100 patients ROTEM and €79,034 (€1,580 €6,520 (€130.40 −3 h (median) Decreased FFP,
(2012) surgery, Germany multiplate per patient) per patient) PRBC, platelets, and
rFVIIa
Esler et al. Cardiac suregry, Before-and-after; 2,176 ROTEM and AU$ 928,998 NA NA Decreased FFP,
(2013) Australia patients Multiplate (€608,385) per PRBC and platelet
year (−48.3%) transfusion (−39.2%)
See Dasta et al. (2005)
K. Görlinger and S.A. Kozek-Langenecker
NA not analyzed
a
Costs per ICU day
24 Economic Aspects and Organization 435
All hospitals dealing with bleeding patients should have a task force on managing
severe bleeding, with the mission of continually developing and implementing stan-
dard operating procedures (SOPs) or algorithms adapted to their local patient popu-
lation’s needs and the diagnostic and therapeutic options available. The successful
implementation of such SOPs can only be accomplished in a multi-specialty setting.
Input and representation from departments such as anesthesiology, intensive care
medicine, hemostaseology and hematology, transfusion medicine, laboratory medi-
cine, trauma and emergency medicine, surgery, and obstetrics are necessary to suc-
cessfully formulate and implement such protocols. Once a severe bleeding
management protocol has been agreed upon, education of the entire nursing and
physician staff is equally essential to its success. Once implemented, this process
may lead to improved clinical outcomes and decreased overall blood use, with
extremely little waste of vital blood products (Görlinger and Schlenke 2012; Levy
et al. 2010; Markova et al. 2012; Milligan et al. 2011; Nunez et al. 2010).
Perioperative bleeding management can be understood as a part of “patient blood
management,” focusing on the stabilization of perioperative hemostasis and a reduc-
tion of perioperative blood loss and transfusion requirements (Gombotz 2012;
Goodnough and Shander 2012; Shander et al. 2012). Leadership in perioperative
bleeding management depends on the kind of strategy hospitals use. In hospitals
using formula-driven transfusion protocols with transfusion packages, clinical
scores to predict the need for massive transfusion are essential to activate massive
transfusion protocols (Maegele et al. 2012). A transfusion medicine department
would have a central position here (Johansson 2007). In contrast, hospitals using
laboratory- or POC-driven protocols focus on early detection and differentiation
between coagulopathies and subsequent goal-directed therapy. Here, POC testing
not only facilitates timely detection of hemostatic disorders and goal-directed ther-
apy, but it can also be used to improve staff education and interdisciplinary com-
munication by visualization of hemostasis as a didactic tool. In hospitals using this
concept, anesthetists, intensivists, hematologists, and laboratory scientists are in
central positions for the management of severe perioperative bleeding (Görlinger
2012; Kozek-Langenecker 2010; Lier et al. 2013). The optimal location for the POC
device – either in the emergency room, operating room or ICU, or in the central
laboratory – is dependent on the local situation, regional structure, staff education,
and the hospital’s requirements. If the POC device is located in the central labora-
tory, then a quick, effective system to transport blood samples and an electronic
connection to get results back to the operating room are crucial. If the POC device
is used in a mobile way, moving between the operating room and the ICU, then
extensive staff education and training is necessary, as well as strict quality control
management (Perry et al. 2010; Spannagl et al. 2010). If this can be done, turn-
around times for POC testing and the “time-to-treat” for a bleeding patient can be
minimized (Haas et al. 2012b). However, any local concept should be based on an
interdisciplinary consensus and should aim for the optimal treatment of the bleeding
patient concerned. In order to support physicians in hospitals in their
436 K. Görlinger and S.A. Kozek-Langenecker
References
Abraham I, Sun D (2012) The cost of blood transfusion in Western Europe as estimated from six
studies. Transfusion 52(9):1983–1988
Agrawal S, Davidson N, Walker M, Gibson S, Morgan CL, Cowell W (2006) Assessing the total
costs of blood delivery to hospital oncology and haematology patients. Curr Med Res Opin
22(10):1903–1909
Association of Anaesthetists of Great Britain and Ireland, Thomas D, Wee M, Clyburn P, Walker
I, Brohi K, Collins P, Doughty H, Isaac J, Mahoney PM, Shewry L (2010) Blood transfusion
and the anaesthetist: management of massive haemorrhage. Anaesthesia 65(11):
1153–1161
Avidan MS, Alcock EL, Da Fonseca J, Ponte J, Desai JB, Despotis GJ, Hunt BJ (2004) Comparison
of structured use of routine laboratory tests or near-patient assessment with clinical judgement
in the management of bleeding after cardiac surgery. Br J Anaesth 92(2):178–186
Berenson K, Casciano R, Makenbaeva D, Mozaffari E, Lamerato L, Corbelli J (2010) Economic
consequences of severe bleeding in patients with acute coronary syndrome in the USA. Adv
Ther 27(8):564–579
Boffard KD, Riou B, Warren B, Choong PI, Rizoli S, Rossaint R, Axelsen M, Kluger Y, NovoSeven
Trauma Study Group (2005) Recombinant factor VIIa as adjunctive therapy for bleeding con-
trol in severely injured trauma patients: two parallel randomized, placebo-controlled, double-
blind clinical trials. J Trauma 59(1):8–15
Borgman MA, Spinella PC, Holcomb JB, Blackbourne LH, Wade CE, Lefering R, Bouillon B,
Maegele M (2011) The effect of FFP:RBC ratio on morbidity and mortality in trauma patients
based on transfusion prediction score. Vox Sang 101:44–54
Brown JR, Birkmeyer NJ, O'Connor GT (2007) Meta-analysis comparing the effectiveness and
adverse outcomes of antifibrinolytic agents in cardiac surgery. Circulation 115(22):2801–2813
Bufe A, Frey S, Briswalter S (2009) Costs caused by bleeds within the therapy of acute coronary
syndromes in Germany. Herz 34(6):479–484. [Article in German]
Callum JL, Rizoli S (2012a) Assessment and management of massive bleeding: coagulation
assessment, pharmacologic strategies, and transfusion management. Hematol Am Soc Hematol
Educ Progr 2012:522–528
Callum JL, Rizoli S (2012b) Plasma transfusion for patients with severe hemorrhage: what is the
evidence? Transfusion 52(Suppl 1):30S–37S
Carless PA, Henry DA, Moxey AJ, O'Connell D, Brown T, Fergusson DA (2010) Cell salvage for
minimising perioperative allogeneic blood transfusion. Cochrane Database Syst Rev
(4):CD001888
Chavez-Tapia NC, Alfaro-Lara R, Tellez-Avila F, Barrientos-Gutiérrez T, González-Chon O,
Mendez-Sanchez N, Uribe M (2011) Prophylactic activated recombinant factor VII in liver
resection and liver transplantation: systematic review and meta-analysis. PLoS One
6(7):e22581
Christensen MC, Krapf S, Kempel A, von Heymann C (2009) Costs of excessive postoperative
hemorrhage in cardiac surgery. J Thorac Cardiovasc Surg 138(3):687–693
Cotton BA, Gunter OL, Isbell J, Au BK, Robertson AM, Morris JA Jr, St Jacques P, Young PP
(2008) Damage control hematology: the impact of a trauma exsanguination protocol on
survival and blood product utilization. J Trauma 64(5):1177–1182
24 Economic Aspects and Organization 437
Cotton BA, Au BK, Nunez TC, Gunter OL, Robertson AM, Young PP (2009) Predefined massive
transfusion protocols are associated with a reduction in organ failure and postinjury complica-
tions. J Trauma 66(1):41–48
CRASH-2 Collaborators, Roberts I, Shakur H, Afolabi A, Brohi K, Coats T, Dewan Y, Gando S,
Guyatt G, Hunt BJ, Morales C, Perel P, Prieto-Merino D, Woolley T (2011) The importance of
early treatment with tranexamic acid in bleeding trauma patients: an exploratory analysis of the
CRASH-2 randomised controlled trial. Lancet 377(9771):1096–1101, 1101.e1–1102
CRASH-2 Trial Collaborators, Shakur H, Roberts I, Bautista R, Caballero J, Coats T, Dewan Y,
El-Sayed H, Gogichaishvili T, Gupta S, Herrera J, Hunt B, Iribhogbe P, Izurieta M, Khamis H,
Komolafe E, Marrero MA, Mejía-Mantilla J, Miranda J, Morales C, Olaomi O, Olldashi F,
Perel P, Peto R, Ramana PV, Ravi RR, Yutthakasemsunt S (2010) Effects of tranexamic acid on
death, vascular occlusive events, and blood transfusion in trauma patients with significant
haemorrhage (CRASH-2): a randomised, placebo-controlled trial. Lancet 376(9734):23–32
Cushing M, Shaz BH (2011) Blood transfusion in trauma patients: unresolved questions. Minerva
Anestesiol 77(3):349–359
Dasta JF, McLaughlin TP, Mody SH, Piech CT (2005) Daily cost of an intensive care unit day: the
contribution of mechanical ventilation. Crit Care Med 33(6):1266–1271
Davenport R, Manson J, De'Ath H, Platton S, Coates A, Allard S, Hart D, Pearse R, Pasi KJ,
MacCallum P, Stanworth S, Brohi K (2011) Functional definition and characterization of acute
traumatic coagulopathy. Crit Care Med 39(12):2652–2658
Davies L, Brown TJ, Haynes S, Payne K, Elliott RA, McCollum C (2006) Cost-effectiveness of
cell salvage and alternative methods of minimising perioperative allogeneic blood transfusion:
a systematic review and economic model. Health Technol Assess 10(44):iii–iv, ix–x, 1–210
Dente CJ, Shaz BH, Nicholas JM, Harris RS, Wyrzykowski AD, Patel S, Shah A, Vercruysse GA,
Feliciano DV, Rozycki GS, Salomone JP, Ingram WL (2009) Improvements in early mortality
and coagulopathy are sustained better in patients with blunt trauma after institution of a massive
transfusion protocol in a civilian level I trauma center. J Trauma 66(6):1616–1624
Dobesh PP (2009) Economic burden of venous thromboembolism in hospitalized patients.
Pharmacotherapy 29(8):943–953
Ducloy-Bouthors AS, Jude B, Duhamel A, Broisin F, Huissoud C, Keita-Meyer H, Mandelbrot L,
Tillouche N, Fontaine S, Le Goueff F, Depret-Mosser S, Vallet B, EXADELI Study Group,
Susen S (2011) High-dose tranexamic acid reduces blood loss in postpartum haemorrhage. Crit
Care 15(2):R117
Dutton R, Hauser C, Boffard K, Dimsitts J, Bernard G, Holcomb J, Leppäniemi A, Tortella B,
Bouillon B, CONTROL Steering Committee (2009) Scientific and logistical challenges in
designing the CONTROL trial: recombinant factor VIIa in severe trauma patients with refractory
bleeding. Clin Trials 6(5):467–479
Dzik WH, Blajchman MA, Fergusson D, Hameed M, Henry B, Kirkpatrick AW, Korogyi T,
Logsetty S, Skeate RC, Stanworth S, MacAdams C, Muirhead B (2011) Clinical review:
Canadian National Advisory Committee on Blood and Blood Products – massive transfusion
consensus conference 2011: report of the panel. Crit Care 15(6):242
Esler C, Ghag S, Kirstenfeld C, Fung YL, Pearse B (2013) Health Services Support Agency
(HSSA): Implementation of ROTEM® (Rotational thromboelastometry) in cardiac theatre
reduces use of blood products. Poster, HAA 20–23. Queensland, Australia
Fergusson DA, Hébert PC, Mazer CD, Fremes S, MacAdams C, Murkin JM, Teoh K, Duke PC,
Arellano R, Blajchman MA, Bussières JS, Côté D, Karski J, Martineau R, Robblee JA, Rodger
M, Wells G, Clinch J, Pretorius R, BART Investigators (2008) A comparison of aprotinin and
lysine analogues in high-risk cardiac surgery. N Engl J Med 358(22):2319–2331
Ferrer P, Roberts I, Sydenham E, Blackhall K, Shakur H (2009) Anti-fibrinolytic agents in postpartum
haemorrhage: a systematic review. BMC Pregnancy Childbirth 9:29
Gill R, Herbertson M, Vuylsteke A, Olsen PS, von Heymann C, Mythen M, Sellke F, Booth F,
Schmidt TA (2009) Safety and efficacy of recombinant activated factor VII: a randomized
placebo-controlled trial in the setting of bleeding after cardiac surgery. Circulation
120(1):21–27
438 K. Görlinger and S.A. Kozek-Langenecker
Girdauskas E, Kempfert J, Kuntze T, Borger MA, Enders J, Fassl J, Falk V, Mohr FW (2010)
Thromboelastometrically guided transfusion protocol during aortic surgery with circulatory
arrest: a prospective, randomized trial. J Thorac Cardiovasc Surg 140(5):1117–1124.e2
Glance LG, Dick AW, Mukamel DB, Fleming FJ, Zollo RA, Wissler R, Salloum R, Meredith UW,
Osler TM (2011) Association between intraoperative blood transfusion and mortality and mor-
bidity in patients undergoing noncardiac surgery. Anesthesiology 114(2):283–292
Glenngård AH, Persson U, Söderman C (2005) Costs associated with blood transfusions in
Sweden – the societal cost of autologous, allogeneic and perioperative RBC transfusion.
Transfus Med 15(4):295–306
Goerlinger K, Dirkmann D, Müller-Beißenhirtz H, Paul A, Hartmann M, Saner F (2010)
Coagulation management during liver transplantation. Inflamm Res 59(Suppl 1):S147–S148
Gombotz H (2012) Patient blood management: a patient-orientated approach to blood replacement
with the goal of reducing anemia, blood loss and the need for blood transfusion in elective
surgery. Transfus Med Hemother 39(2):67–72
Goobie SM, Meier PM, Pereira LM, McGowan FX, Prescilla RP, Scharp LA, Rogers GF, Proctor
MR, Meara JG, Soriano SG, Zurakowski D, Sethna NF (2011) Efficacy of tranexamic acid in
pediatric craniosynostosis surgery: a double-blind, placebo-controlled trial. Anesthesiology
114(4):862–871
Goodnough LT, Shander A (2012) Patient blood management. Anesthesiology 116(6):1367–1376
Görlinger K (2012) Panta rhei: blood, professional career and anesthesiological self-conception.
Anaesthesist 61(6):481–482
Görlinger K, Schlenke P (2012) Patient blood management: clinical hemotherapy and hemostasis
management in perioperative settings. Transfus Med Hemother 39(2):57–58
Görlinger K, Jambor C, Hanke AA et al (2007) Perioperative coagulation management and control
of platelet transfusion by point-of-care platelet function analysis. Transfus Med Hemother
34:396–411
Görlinger K, Moog R, Saner F, Müller-Beißenhirtz H (2008) Einfluss eines Point-of-Care- und
Faktorenkonzentrate-basierten Gerinnungsmanagements auf den Transfusionsbedarf und die
Kostenentwicklung am Universitätsklinikum Essen. Anästh Intensivmed 49(Suppl 7):
S226
Görlinger K, Dirkmann D, Müller-Beißenhirtz H, Paul A, Hartmann M, Saner F (2010)
Thromboelastometry-based perioperative coagulation management in visceral surgery and
liver transplantation: experience of 10 years and 1105 LTX. Liver Transplant 16(Suppl 1):S86
Görlinger K, Dirkmann D, Hanke AA, Kamler M, Kottenberg E, Thielmann M, Jakob H, Peters J
(2011a) First-line therapy with coagulation factor concentrates combined with point-of-care
coagulation testing is associated with decreased allogeneic blood transfusion in cardiovascular
surgery: a retrospective, single-center cohort study. Anesthesiology 115(6):1179–1191
Görlinger K, Dirkmann D, Weber CF et al (2011b) Algorithms for transfusion and coagulation
management in massive haemorrhage. Anästh Intensivmed 52:145–159
Görlinger K, Fries D, Dirkmann D, Weber CF, Hanke AA, Schöchl H (2012) Reduction of fresh
frozen plasma requirements by perioperative point-of-care coagulation management with early
calculated goal-directed therapy. Transfus Med Hemother 39(2):104–113
Görlinger K, Dirkmann D, Hanke AA (2013) Potential value of transfusion protocols in cardiac
surgery. Curr Opin Anaesthesiol 26(2):230–243
Greiff G, Stenseth R, Wahba A, Videm V, Lydersen S, Irgens W, Bjella L, Pleym H (2012)
Tranexamic acid reduces blood transfusions in elderly patients undergoing combined aortic
valve and coronary artery bypass graft surgery: a randomized controlled trial. J Cardiothorac
Vasc Anesth 26(2):232–238
Griffee MJ, Deloughery TG, Thorborg PA (2010) Coagulation management in massive bleeding.
Curr Opin Anaesthesiol 23(2):263–268
Grottke O, Braunschweig T, Spronk HM, Esch S, Rieg AD, van Oerle R, ten Cate H, Fitzner C,
Tolba R, Rossaint R (2011) Increasing concentrations of prothrombin complex concentrate
induce disseminated intravascular coagulation in a pig model of coagulopathy with blunt liver
injury. Blood 118(7):1943–1951
24 Economic Aspects and Organization 439
Guerriero C, Cairns J, Jayaraman S, Roberts I, Perel P, Shakur H (2010) Giving tranexamic acid to
reduce surgical bleeding in sub-Saharan Africa: an economic evaluation. Cost Eff Resour Alloc
8(1):1
Guerriero C, Cairns J, Perel P, Shakur H, Roberts I (2011) CRASH 2 trial collaborators. Cost-
effectiveness analysis of administering tranexamic acid to bleeding trauma patients using evi-
dence from the CRASH-2 trial. PLoS One 6(5):e18987
Gurusamy KS, Li J, Sharma D, Davidson BR (2009) Pharmacological interventions to decrease
blood loss and blood transfusion requirements for liver resection. Cochrane Database Syst Rev
(4):CD008085
Haas T, Spielmann N, Mauch J, Madjdpour C, Speer O, Schmugge M, Weiss M (2012a)
Comparison of thromboelastometry (ROTEM®) with standard plasmatic coagulation testing in
paediatric surgery. Br J Anaesth 108(1):36–41
Haas T, Spielmann N, Mauch J, Speer O, Schmugge M, Weiss M (2012b) Reproducibility of
thrombelastometry (ROTEM®): point-of-care versus hospital laboratory performance. Scand J
Clin Lab Invest 72(4):313–317
Hanke AA, Herold U, Dirkmann D, Tsagakis K, Jakob H, Görlinger K (2012) Thromboelastometry
based early goal-directed coagulation management reduces blood transfusion requirements,
adverse events, and costs in acute type A aortic dissection: a pilot study. Transfus Med
Hemother 39(2):121–128
Hanke AA, Joch C, Görlinger K (2013) Long-term safety and efficacy of a pasteurized nanofil-
trated prothrombin complex concentrate (Beriplex P/N): a pharmacovigilance study. Br J
Anaesth 110:1–9. doi:10.1093/bja/aes501. [Published online 2013 Jan 18]
Hauser CJ, Boffard K, Dutton R, Bernard GR, Croce MA, Holcomb JB, Leppaniemi A, Parr M,
Vincent JL, Tortella BJ, Dimsits J, Bouillon B, CONTROL Study Group (2010) Results of the
CONTROL trial: efficacy and safety of recombinant activated Factor VII in the management of
refractory traumatic hemorrhage. J Trauma 69(3):489–500
Henry DA, Carless PA, Moxey AJ, O’Connell D, Stokes BJ, Fergusson DA, Ker K (2011) Anti-
fibrinolytic use for minimising perioperative allogeneic blood transfusion. Cochrane Database
Syst Rev (3):CD001886
Ho KM, Litton E (2012) Cost-effectiveness of using recombinant activated factor VII as an
off-label rescue treatment for critical bleeding requiring massive transfusion. Transfusion
52(8):1696–1702
Ho AM, Dion PW, Yeung JH, Holcomb JB, Critchley LA, Ng CS, Karmakar MK, Cheung CW,
Rainer TH (2012) Prevalence of survivor bias in observational studies on fresh frozen
plasma: erythrocyte ratios in trauma requiring massive transfusion. Anesthesiology
116(3):716–728
Holcomb JB, Wade CE, Michalek JE, Chisholm GB, Zarzabal LA, Schreiber MA, Gonzalez EA,
Pomper GJ, Perkins JG, Spinella PC, Williams KL, Park MS (2008) Increased plasma and
platelet to red blood cell ratios improves outcome in 466 massively transfused civilian trauma
patients. Ann Surg 248(3):447–458
Howes JL, Smith RS, Helmer SD, Helmer SD, Taylor SM (2009) Complications of recombinant
activated human coagulation factor VII. Am J Surg 198(6):895–899
Ickx BE, van der Linden PJ, Melot C, Wijns W, de Pauw L, Vandestadt J, Hut F, Pradier O (2006)
Comparison of the effects of aprotinin and tranexamic acid on blood loss and red blood cell
transfusion requirements during the late stages of liver transplantation. Transfusion
46(4):595–605
Inaba K, Branco BC, Rhee P, Blackbourne LH, Holcomb JB, Teixeira PG, Shulman I, Nelson J,
Demetriades D (2010) Impact of plasma transfusion in trauma patients who do not require mas-
sive transfusion. J Am Coll Surg 210(6):957–965
Innerhofer P, Westermann I, Tauber H, Breitkopf R, Fries D, Kastenberger T, El Attal R, Strasak
A, Mittermayr M (2013) The exclusive use of coagulation factor concentrates enables reversal
of coagulopathy and decreases transfusion rates in patients with major blunt trauma. Injury
44(2):209–216
James AH, McLintock C, Lockhart E (2012) Postpartum hemorrhage: when uterotonics and
sutures fail. Am J Hematol 87(Suppl 1):S16–S22
440 K. Görlinger and S.A. Kozek-Langenecker
Johansson PI (2007) The blood bank: from provider to partner in treatment of massively bleeding
patients. Transfusion 47(2 Suppl):176S–181S
Johansson PI (2010) Goal-directed hemostatic resuscitation for massively bleeding patients: the
Copenhagen concept. Transfus Apher Sci 43(3):401–405
Johansson PI (2012) Coagulation monitoring of the bleeding traumatized patient. Curr Opin
Anaesthesiol 25(2):235–241
Johansson PI, Stensballe J (2009) Effect of Haemostatic Control Resuscitation on mortality in
massively bleeding patients: a before and after study. Vox Sang 96(2):111–118
Johansson PI, Stensballe J (2010) Hemostatic resuscitation for massive bleeding: the paradigm of
plasma and platelets – a review of the current literature. Transfusion 50(3):701–710
Johansson PI, Stensballe J, Ostrowski SR (2012) Current management of massive hemorrhage in
trauma. Scand J Trauma Resusc Emerg Med 20:47
Johnson JL, Moore EE, Kashuk JL, Banerjee A, Cothren CC, Biffl WL, Sauaia A (2010) Effect of
blood products transfusion on the development of postinjury multiple organ failure. Arch Surg
145(10):973–977
Kaushal R, Bates DW, Franz C, Soukup JR, Rothschild JM (2007) Costs of adverse events in
intensive care units. Crit Care Med 35(11):2479–2483
Khan H, Belsher J, Yilmaz M, Afessa B, Winters JL, Moore SB, Hubmayr RD, Gajic O (2007)
Fresh-frozen plasma and platelet transfusions are associated with development of acute lung
injury in critically ill medical patients. Chest 131(5):1308–1314
Kirchner C, Goerlinger K, Dirkmann D, Schumann R, Treckmann J, Paul A, Saner FH (2012)
Safety and efficacy of prothrombin complex and fibrinogen concentrates in liver transplanta-
tion. Liver Transplant 18(Suppl 1):S189
Kozek-Langenecker SA (2010) Perioperative coagulation monitoring. Best Pract Res Clin
Anaesthesiol 24(1):27–40
Kozek-Langenecker S, Sørensen B, Hess JR, Spahn DR (2011) Clinical effectiveness of fresh
frozen plasma compared with fibrinogen concentrate: a systematic review. Crit Care
15(5):R239
Kozek-Langenecker SA, Afshari A, Albaladejo P et al (2013) Guidelines on the management of
severe perioperative bleeding. Eur J Anaesthesiol 30(6):270–382
Leahy MF, Mukhtar SA (2012) From blood transfusion to patient blood management: a new para-
digm for patient care and cost assessment of blood transfusion practice. Intern Med J
42(3):332–338
Levi M, Levy JH, Andersen HF, Truloff D (2010) Safety of recombinant activated factor VII in
randomized clinical trials. N Engl J Med 363(19):1791–1800
Levy JH, Dutton RP, Hemphill JC 3rd, Shander A, Cooper D, Paidas MJ, Kessler CM, Holcomb
JB, ; Hemostasis Summit Participants JH (2010) Multidisciplinary approach to the challenge of
hemostasis. Anesth Analg 110(2):354–364
Lier H, Vorweg M, Hanke A, Görlinger K (2013) Thromboelastometry guided therapy of severe
bleeding. Essener Runde algorithm. Hamostaseologie 33(1):51–61
Lin Y, Moltzan CJ, Anderson DR, National Advisory Committee on Blood and Blood Products
(2012) The evidence for the use of recombinant factor VIIa in massive bleeding: revision of the
transfusion policy framework. Transfus Med 22(6):383–394
Liumbruno G, Bennardello F, Lattanzio A, Piccoli P, Rossetti G, Italian Society of Transfusion
Medicine and Immunohaematology (SIMTI) Work Group (2009) Recommendations for the
transfusion of plasma and platelets. Blood Transfus 7(2):132–150
Lodge JP, Jonas S, Jones RM, Olausson M, Mir-Pallardo J, Soefelt S, Garcia-Valdecasas JC,
McAlister V, Mirza DF, rFVIIa OLT Study Group (2005) Efficacy and safety of repeated periop-
erative doses of recombinant factor VIIa in liver transplantation. Liver Transpl 11(8):
973–979
Maegele M, Lefering R, Paffrath T, Tjardes T, Simanski C, Bouillon B, Working Group on
Polytrauma of the German Society of Trauma Surgery (DGU) (2008) Red-blood-cell to plasma
ratios transfused during massive transfusion are associated with mortality in severe multiple
injury: a retrospective analysis from the Trauma Registry of the Deutsche Gesellschaft für
Unfallchirurgie. Vox Sang 95(2):112–119
24 Economic Aspects and Organization 441
Stewart RM, Cohn SM, Bulger EM, Cotton BA, Nunez TC, Ivatury R, Meredith JW, Miller P,
Pomper GJ, Marin B (2011) High ratios of plasma and platelets to packed red blood cells do not
affect mortality in nonmassively transfused patients. J Trauma 71(2 Suppl 3):S329–S336
Sander M, Spies CD, Martiny V, Rosenthal C, Wernecke KD, von Heymann C (2010) Mortality
associated with administration of high-dose tranexamic acid and aprotinin in primary open-
heart procedures: a retrospective analysis. Crit Care 14(4):R148
Sarani B, Dunkman WJ, Dean L, Sonnad S, Rohrbach JI, Gracias VH (2008) Transfusion of fresh
frozen plasma in critically ill surgical patients is associated with an increased risk of infection.
Crit Care Med 36(4):1114–1118
Sarode R, Refaai MA, Matevosyan K, Burner JD, Hampton S, Rutherford C (2010) Prospective
monitoring of plasma and platelet transfusions in a large teaching hospital results in significant
cost reduction. Transfusion 50(2):487–492
Scalea TM, Bochicchio KM, Lumpkins K, Hess JR, Dutton R, Pyle A, Bochicchio GV (2008)
Early aggressive use of fresh frozen plasma does not improve outcome in critically injured
trauma patients. Ann Surg 248(4):578–584
Schaden E, Saner FH, Goerlinger K (2013) Coagulation pattern in critical liver dysfunction. Curr
Opin Crit Care 19(2):142–148
Schöchl H, Nienaber U, Hofer G, Voelckel W, Jambor C, Scharbert G, Kozek-Langenecker S,
Solomon C (2010) Goal-directed coagulation management of major trauma patients using
thromboelastometry (ROTEM)-guided administration of fibrinogen concentrate and prothrom-
bin complex concentrate. Crit Care 14(2):R55
Schöchl H, Nienaber U, Maegele M, Hochleitner G, Primavesi F, Steitz B, Arndt C, Hanke A,
Voelckel W, Solomon C (2011) Transfusion in trauma: thromboelastometry-guided coagula-
tion factor concentrate-based therapy versus standard fresh frozen plasma-based therapy. Crit
Care 15(2):R83
Schöchl H, Maegele M, Solomon C, Görlinger K, Voelckel W (2012) Early and individualized goal-
directed therapy for trauma-induced coagulopathy. Scand J Trauma Resusc Emerg Med 20:15
Schöchl H, Schlimp CJ, Voelckel W (2013) Potential value of pharmacological protocols in trauma. Curr
Opin Anaesthesiol 26. doi:10.1097/ACO.0b013e32835cca92. [Epub ahead of print 2013 Jan 14]
Schofield N, Sugavanam A, Mallett S (2012) Blood product usage and fibrinolysis in liver trans-
plantation before and after cessation of aprotinin. Liver Transplant 18(Suppl 1):S113
Schuster KM, Davis KA, Lui FY, Maerz LL, Kaplan LJ (2010) The status of massive transfusion
protocols in United States trauma centers: massive transfusion or massive confusion?
Transfusion 50(7):1545–1551
Shakur H, Elbourne D, Gülmezoglu M, Alfirevic Z, Ronsmans C, Allen E, Roberts I (2010)
The WOMAN Trial (World Maternal Antifibrinolytic Trial): tranexamic acid for the treatment
of postpartum haemorrhage: an international randomised, double blind placebo controlled trial.
Trials 11:40
Shander A (2007) Financial and clinical outcomes associated with surgical bleeding complications.
Surgery 142(4 Suppl):S20–S25
Shander A, Hofmann A, Gombotz H, Theusinger OM, Spahn DR (2007) Estimating the cost of
blood: past, present, and future directions. Best Pract Res Clin Anaesthesiol 21(2):271–289
Shander A, Hofmann A, Ozawa S, Theusinger OM, Gombotz H, Spahn DR (2010) Activity-based
costs of blood transfusions in surgical patients at four hospitals. Transfusion 50(4):753–765
Shander A, Javidroozi M, Ozawa S, Hare GM (2011) What is really dangerous: anaemia or trans-
fusion? Br J Anaesth 107(Suppl 1):i41–i59
Shander A, Van Aken H, Colomina MJ, Gombotz H, Hofmann A, Krauspe R, Lasocki S, Richards T,
Slappendel R, Spahn DR (2012) Patient blood management in Europe. Br J Anaesth 109(1):55–68
Sharples L, Buxton M, Caine N, Cafferty F, Demiris N, Dyer M, Freeman C (2006) Evaluation of
the ventricular assist device programme in the UK. Chapter 6: Analysis of patient-specific data
on resource use and costs. Health Technol Assess 10(48):67–72
Shore-Lesserson L, Manspeizer HE, DePerio M, Francis S, Vela-Cantos F, Ergin MA (1999)
Thromboelastography-guided transfusion algorithm reduces transfusions in complex cardiac
surgery. Anesth Analg 88(2):213–319
444 K. Görlinger and S.A. Kozek-Langenecker
Simpson E, Lin Y, Stanworth S, Birchall J, Doree C, Hyde C (2012) Recombinant factor VIIa for
the prevention and treatment of bleeding in patients without haemophilia. Cochrane Database
Syst Rev (3):CD005011
Snyder CW, Weinberg JA, McGwin G Jr, Melton SM, George RL, Reiff DA, Cross JM, Hubbard-
Brown J, Rue LW 3rd, Kerby JD (2009) The relationship of blood product ratio to mortality:
survival benefit or survival bias? J Trauma 66(2):358–362
Solomon C, Collis RE, Collins PW (2012) Haemostatic monitoring during postpartum haemor-
rhage and implications for management. Br J Anaesth 109:851–863
Sørensen B, Spahn DR, Innerhofer P, Spannagl M, Rossaint R (2011) Clinical review: prothrombin
complex concentrates – evaluation of safety and thrombogenicity. Crit Care 15(1):201
Spalding GJ, Hartrumpf M, Sierig T, Oesberg N, Kirschke CG, Albes JM (2007) Cost reduction of
perioperative coagulation management in cardiac surgery: value of “bedside” thrombelastogra-
phy (ROTEM). Eur J Cardiothorac Surg 31(6):1052–1057
Spannagl M, Dick A, Junker R (2010) POCT in coagulation. Quality assurance. Hamostaseologie
30(2):82–90
Spiess BD, Gillies BS, Chandler W et al (1995) Changes in transfusion therapy and reexploration
rate after institution of a blood management program in cardiac surgical patients. J Cardiothorac
Vasc Anesth 9:168–173
Spiess BD, Royston D, Levy JH, Fitch J, Dietrich W, Body S, Murkin J, Nadel A (2004) Platelet
transfusions during coronary artery bypass graft surgery are associated with serious adverse
outcomes. Transfusion 44(8):1143–1148
Spinella PC, Holcomb JB (2009) Resuscitation and transfusion principles for traumatic hemor-
rhagic shock. Blood Rev 23(6):231–240
Stanworth SJ, Grant-Casey J, Lowe D, Laffan M, New H, Murphy MF, Allard S (2011a) The use
of fresh-frozen plasma in England: high levels of inappropriate use in adults and children.
Transfusion 51(1):62–70
Stanworth SJ, Walsh TS, Prescott RJ, Lee RJ, Watson DM, Wyncoll D, Intensive Care Study of
Coagulopathy (ISOC) investigators (2011b) A national study of plasma use in critical care:
clinical indications, dose and effect on prothrombin time. Crit Care 15(2):R108
Stokes ME, Ye X, Shah M, Mercaldi K, Reynolds MW, Rupnow MF, Hammond J (2011) Impact
of bleeding-related complications and/or blood product transfusions on hospital costs in inpa-
tient surgical patients. BMC Health Serv Res 11:135
Stürmer KM, Neugebauer E, Deutsche Gesellschaft für Unfallchirurgie (federführend). S3-Leitlinie
Polytrauma/Schwerverletzten-Behandlung. AWMF online 07/2011. http://www.awmf.org/uploads/
tx_szleitlinien/012-019l_S3_Polytrauma_Schwerverletzten-Behandlung_2011-07_01.pdf
Sugg RM, Gonzales NR, Matherne DE, Ribo M, Shaltoni HM, Baraniuk S, Noser EA, Grotta JC
(2006) Myocardial injury in patients with intracerebral hemorrhage treated with recombinant
factor VIIa. Neurology 67(6):1053–1055
Sukeik M, Alshryda S, Haddad FS, Mason JM (2011) Systematic review and meta-analysis of the
use of tranexamic acid in total hip replacement. J Bone Joint Surg Br 93(1):39–46
Tavares M, DiQuattro P, Nolette N, Conti G, Sweeney J (2011) Reduction in plasma transfusion
after enforcement of transfusion guidelines. Transfusion 51(4):754–761
The Board of the German Medical Association on the Recommendation of the Scientific Advisory
Board (Bundesärztekammer) (2009) Chapter 7 – Procoagulators. Cross-sectional guidelines
for therapy with blood components and plasma derivatives, 4th edn. Transfus Med Hemother.
36(6):419–436
The Board of the German Medical Association on the Recommendation of the Scientific Advisory
Board (Bundesärztekammer) (2009) Chapter 4 – Plasma for therapeutic use. Cross-sectional
guidelines for therapy with blood components and plasma derivatives, 4th edn. Transfus Med
Hemother 36(6):388–397
Theusinger OM, Felix C, Spahn DR (2012) Strategies to reduce the use of blood products: a
European perspective. Curr Opin Anaesthesiol 25(1):59–65
Toner RW, Pizzi L, Leas B, Ballas SK, Quigley A, Goldfarb NI (2011) Costs to hospitals of acquir-
ing and processing blood in the US: a survey of hospital-based blood banks and transfusion
services. Appl Health Econ Health Policy 9(1):29–37
24 Economic Aspects and Organization 445
Toulon P, Ozier Y, Ankri A, Fléron MH, Leroux G, Samama CM (2009) Point-of-care versus
central laboratory coagulation testing during haemorrhagic surgery. A multicenter study.
Thromb Haemost 101(2):394–401
Tripodi A, Mannucci PM (2011) The coagulopathy of chronic liver disease. N Engl J Med
365(2):147–156
Trzebicki J, Flakiewicz E, Kosieradzki M, Blaszczyk B, Kołacz M, Jureczko L, Pacholczyk M,
Chmura A, Lagiewska B, Lisik W, Wasiak D, Kosson D, Kwiatkowski A, Lazowski T (2010)
The use of thromboelastometry in the assessment of hemostasis during orthotopic liver trans-
plantation reduces the demand for blood products. Ann Transplant 15(3):19–24
Tzortzopoulou A, Cepeda MS, Schumann R, Carr DB (2008) Antifibrinolytic agents for reducing
blood loss in scoliosis surgery in children. Cochrane Database Syst Rev (3):CD006883
Varney SJ, Guest JF (2003) The annual cost of blood transfusions in the UK. Transfus Med
13(4):205–218
Vekeman F, LaMori JC, Laliberte F et al (2011) Risks and cost burden of venous thromboembo-
lism and bleeding for patients undergoing total hip or knee replacement in a managed-care
population. J Med Econ 14(3):324–334
Vogt KN, Van Koughnett JA, Dubois L, Gray DK, Parry NG (2012) The use of trauma transfusion
pathways for blood component transfusion in the civilian population: a systematic review and
meta-analysis. Transfus Med 22(3):156–166
Wang G, Bainbridge D, Martin J, Cheng D (2009) The efficacy of an intraoperative cell saver
during cardiac surgery: a meta-analysis of randomized trials. Anesth Analg 109(2):320–330
Wang SC, Shieh JF, Chang KY, Chu YC, Liu CS, Loong CC, Chan KH, Mandell S, Tsou MY (2010)
Thromboelastography-guided transfusion decreases intraoperative blood transfusion during ortho-
topic liver transplantation: randomized clinical trial. Transplant Proc 42(7):2590–2593
Warmuth M, Mad P, Wild C (2012) Systematic review of the efficacy and safety of fibrinogen
concentrate substitution in adults. Acta Anaesthesiol Scand 56(5):539–548
Watson GA, Sperry JL, Rosengart MR, Minei JP, Harbrecht BG, Moore EE, Cuschieri J, Maier RV,
Billiar TR, Peitzman AB, Inflammation and Host Response to Injury Investigators (2009)
Fresh frozen plasma is independently associated with a higher risk of multiple organ failure and
acute respiratory distress syndrome. J Trauma 67(2):221–227
Waydhas C, Görlinger K (2009) Coagulation management in multiple trauma. Unfallchirurg
112(11):942–950
Weber CF, Görlinger K, Meininger D, Herrmann E, Bingold T, Moritz A, Cohn LH, Zacharowski K
(2012) Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulo-
pathic cardiac surgery patients. Anesthesiology 117(3):531–547
Zehtabchi S, Nishijima DK (2009) Impact of transfusion of fresh-frozen plasma and packed red
blood cells in a 1:1 ratio on survival of emergency department patients with severe trauma.
Acad Emerg Med 16(5):371–378
Index