Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Thomas F.

Slaughter

56 Coagulation

Key Points
1. Under normal physiologic conditions, clot formation proteins; and (3) major structural components of the
requires participation of vascular endothelium, platelets, clot.
and plasma-mediated hemostasis. 5. A carefully performed history of bleeding remains the
2. Tissue factor (extrinsic pathway) initiates plasma-mediated most effective means for identifying bleeding and
hemostasis, whereas factor XI (intrinsic pathway) amplifies thrombotic tendencies.
the response. 6. Thrombosis is potentiated by stasis, vascular endothelial
3. Thrombin generation proves the key regulatory enzymatic injury, and underlying hypercoagulable conditions.
step in hemostasis. 7. Heparin-induced thrombocytopenia (HIT) comprises a
4. Platelets participate in clot formation as (1) anchoring heparin-mediated autoimmune response potentiating
sites for coagulation factor activation complexes; (2) platelet activation as well as venous and arterial
delivery “vehicles” releasing hemostatically active thromboses.

Normal Hemostasis Vascular Endothelial Role in Hemostasis

Hemostasis comprises cellular and biochemical processes that Under normal conditions, vascular endothelium provides a non-
limit blood loss resulting from injury, maintain intravascular thrombogenic surface to promote blood fluidity. Healthy endothe-
blood fluidity, and promote revascularization of thrombosed lial cells possess antiplatelet, anticoagulant, and profibrinolytic
vessels after injury. Normal physiologic hemostasis necessitates a effects to inhibit clot formation.1 Negatively charged vascular
delicate balance between procoagulant pathways responsible for endothelium repels platelets and produces prostacyclin (PGI2)
generation of a stable localized hemostatic “plug” and counter- and nitric oxide (NO), which are potent platelet inhibitors.2 Ade-
regulatory mechanisms inhibiting thrombus formation beyond nosine diphosphatase (ADPase) synthesized by vascular endothe-
the injury site. Vascular endothelium, platelets, and plasma coagu- lial cells degrades adenosine diphosphate (ADP), another potent
lation proteins play equally important roles in this process. Failure platelet activator. Given these endogenous antiplatelet effects,
to maintain balance commonly leads to excessive bleeding or nonactivated platelets do not adhere to healthy vascular endothe-
pathologic thrombus formation. lial cells. Vascular endothelium further expresses several inhibi-
Vascular endothelial injury, mechanical or biochemical, tors of plasma-mediated hemostasis, including thrombomodulin
leads to platelet deposition at the injury site, a process often (an indirect thrombin inhibitor), heparin-like glycosaminogly-
referred to as primary hemostasis. Although primary hemostasis cans, and tissue factor pathway inhibitor (TFPI).3 Finally, vascular
may prove adequate for a minor injury, control of more significant endothelium synthesizes tissue plasminogen activator (t-PA),
bleeding necessitates stable clot formation incorporating cross- which is responsible for activating fibrinolysis—a primary coun-
linked fibrin—a process mediated by activation of plasma clotting terregulatory mechanism limiting clot propagation.
factors and often referred to as secondary hemostasis. Although Despite these natural defense mechanisms to inhibit
the terms primary and secondary hemostasis remain relevant for thrombus generation, a variety of mechanical and chemical
descriptive and diagnostic purposes, advances in understanding stimuli may shift the balance such that the endothelium promotes
cellular and molecular processes underlying hemostasis suggest a clot formation. Damage to vascular endothelial cells exposes the
far more complex interplay between vascular endothelium, plate- underlying extracellular matrix (ECM) including collagen, von
lets, and plasma-mediated hemostasis than is reflected in this Willebrand factor (vWF), and other platelet-adhesive glycopro-
model. teins.4,5 Platelets bind to and are activated by exposure to ECM

1767
IV 1768 Anesthesia Management

components. Exposure of tissue factor, constitutively expressed by platelets to the site of injury. Newly active glycoprotein IIb/IIIa
fibroblasts in the ECM, activates plasma-mediated coagulation receptors on the platelet surface bind fibrinogen to provide
pathways to generate thrombin and, ultimately, fibrin clot. Certain for cross-linking with adjacent platelets (platelet aggregation).15
cytokines (i.e., interleukin-1, tumor necrosis factor, and γ- The importance of these receptors is reflected by the bleeding
interferon) and hormones (i.e., desmopressin acetate or endo- disorder associated with their hereditary deficiency: Glanzmann
toxin) induce prothrombotic changes in vascular endothelial thrombasthenia.
cells, including synthesis and expression of vWF, tissue factor,
plasminogen activator inhibitor (PAI-1, an inhibitor of fibrinoly-
sis), as well as downregulate normal antithrombotic cellular and Plasma-Mediated Hemostasis
biochemical pathways.6,7 Thrombin, hypoxia, and high fluid shear
stress induce prothrombotic vascular endothelial changes. Plasma-mediated hemostasis, the coagulation cascade, might best
Increased vascular endothelial synthesis of PAI-1 and associated be summarized as an amplification system to accelerate thrombin
inhibition of fibrinolysis have been implicated in the prothrom- generation from an inactive precursor (i.e., prothrombin). Trace
botic state and high incidence of venous thrombosis after plasma proteins, activated by exposure to tissue factor or expo-
surgery.8,9 sure to foreign surfaces, initiate a cascading series of reactions
culminating in conversion of soluble fibrinogen to insoluble
fibrin clot.16 Thrombin generation, the “thrombin burst,” repre-
Platelets and Hemostasis sents the key regulatory step in this hemostatic process. Thrombin
not only generates fibrin but also activates platelets and mediates
Platelets contribute a critical role in hemostasis. Derived from a host of additional processes affecting inflammation, mitogene-
bone marrow megakaryocytes, nonactivated platelets circulate as sis, and even downregulation of hemostasis.17
discoid anuclear cells.10 The platelet membrane is characterized Traditionally, the “coagulation cascade” describing plasma-
by numerous receptors and a surface-connected open canalicular mediated hemostasis has been depicted as intrinsic and extrinsic
system serving to increase platelet membrane surface area as well pathways, both of which culminate in a common pathway where
as to provide rapid communication between the platelet interior fibrin generation occurs.18 Although this cascade model has
and external environment.11 Under normal circumstances, plate- proved an oversimplification, it remains a useful descriptive tool
lets do not bind vascular endothelium; however, when injury for organizing discussions of plasma-mediated hemostasis (Fig.
exposes ECM, platelets undergo a series of biochemical and phys- 56-1). Coagulation factors are, for the most part, synthesized
ical alterations characterized by three major phases: adhesion, hepatically and circulate as inactive proteins termed zymogens.
activation, and aggregation. The somewhat confusing nomenclature of the classic coagulation
Exposure of subendothelial matrix proteins (i.e., collagen, cascade derives from the fact that inactive zymogens were identi-
vWF, fibronectin) allows for platelet adhesion to the vascular wall. fied using Roman numerals assigned in order of discovery. As the
vWF proves particularly important as a bridging molecule zymogen is converted to an active enzyme, a lower-case letter “a”
between ECM and platelet glycoprotein Ib/factor IX/factor V is added to the Roman numeral identifier. For example, inactive
receptor complexes.12 Absence of either vWF (von Willebrand prothrombin is referred to as factor II, whereas active thrombin
disease) or glycoprotein Ib/factor IX/factor V receptors (Bernard- is identified as factor IIa. Some numerals were subsequently with-
Soulier syndrome) results in a clinically significant bleeding drawn or renamed as our understanding of the biochemistry
disorder. underlying hemostasis evolved.
As platelets adhere along the ECM, a series of physical and The coagulation cascade characterizes a series of enzymatic
biochemical changes occur termed platelet activation. Platelets reactions in which inactive precursors—zymogens—undergo
contain two specific types of storage granules: alpha granules activation to amplify the overall reaction. Each stage of the
and dense bodies.11 Alpha granules contain numerous proteins cascade requires assembly of membrane-bound activation com-
essential to hemostasis and wound repair, including fibrinogen, plexes, each composed of an enzyme (activated coagulation
coagulation factors V and VIII, vWF, platelet-derived growth factor), substrate (inactive precursor zymogen), cofactor (accel-
factor, and others. Dense bodies contain the adenine nucleotides erator/ catalyst), and calcium.19 Assembly of these activation com-
ADP and adenosine triphosphate (ATP) as well as calcium, sero- plexes occurs on phospholipid membranes (most often platelet or
tonin, histamine, and epinephrine. During the activation phase, microparticle membranes) that serve to localize and concentrate
platelets release granular contents, resulting in recruitment reactants. In the absence of phospholipid membrane anchoring
and activation of additional platelets as well as propagation of sites, activation of coagulation factors slows dramatically, further
plasma-mediated coagulation.13 During activation, platelets serving to localize clot generation to injury sites.
undergo structural changes to develop pseudopod-like mem-
brane extensions and to release physiologically active micropar-
ticles, with both mechanisms serving to increase dramatically Extrinsic Pathway of Coagulation
platelet membrane surface area. Redistribution of platelet
membrane phospholipids during activation exposes newly acti- The extrinsic pathway of coagulation, widely recognized as the
vated glycoprotein platelet surface receptors and phospholipid initiating step in plasma-mediated hemostasis, begins with expo-
binding sites for calcium and coagulation factor activation com- sure of blood plasma to tissue factor.20 Tissue factor is prevalent
plexes, which is critical to propagation of plasma-mediated in subendothelial tissues surrounding vasculature; however, under
hemostasis.14 normal conditions the vascular endothelium minimizes contact
During the final phase of platelet aggregation, activators between tissue factor and plasma coagulation factors. After vas-
released during the activation phase serve to recruit additional cular injury, small concentrations of factor VIIa circulating in
Coagulation 1769 56

Intrinsic Pathway Extrinsic Pathway


Vascular endothelial
XII XIIa injury

XI XIa Tissue factor/


VIIIa VIIa complex
IX IXa

Section IV Anesthesia Management


X Xa “Prothrombinase complex”
Va

II IIa
XIII
Va/VIIIa XIIIa

Fibrinogen Fibrin monomer Cross-linked fibrin

Figure 56-1 Depiction of the classic coagulation cascade incorporating extrinsic and intrinsic pathways of coagulation. (From Slaughter TF: The coagulation
system and cardiac surgery. In Estafanous FG, Barash PG, Reves JG [eds]: Cardiac Anesthesia: Principles and Clinical Practice, 2nd ed. Philadelphia, Lippincott
Williams & Wilkins, 2001, p 320, with permission.)

plasma form phospholipid-bound activation complexes with inhibitor, tissue factor pathway inhibitor (TFPI).23 However, small
tissue factor, factor X, and calcium to promote conversion of quantities of thrombin generated before neutralization of the
factor X to Xa.21 Recently, the tissue factor/factor VIIa complex extrinsic pathway activate factor XI and the intrinsic pathway.
has been demonstrated to activate factor IX of the intrinsic The intrinsic pathway subsequently amplifies and propagates the
pathway, further demonstrating the key role of tissue factor in hemostatic response to maximize thrombin generation (Fig.
initiating hemostasis.22 56-2). Although factor XII may be activated by foreign surfaces
(i.e., cardiopulmonary bypass circuits or glass vials), the intrinsic
pathway appears to play a minor role in initiation of hemostasis.
Intrinsic Pathway of Coagulation Proteins of the intrinsic pathway may, however, contribute to
inflammatory processes, complement activation, fibrinolysis,
Classically, the intrinsic or contact activation system was described kinin generation, and angiogenesis.22,24
as a parallel pathway for thrombin generation by way of factor
XII. However, the rarity of bleeding disorders resulting from Common Pathway of Coagulation
contact activation factor deficiencies led to our current under- The final pathway, common to both extrinsic and intrinsic coagu-
standing of the intrinsic pathway as an amplification system to lation cascades, depicts thrombin generation and subsequent
propagate thrombin generation initiated by the extrinsic pathway.22 fibrin formation. Signal amplification through both extrinsic and
Recent cell-based models of coagulation suggest that thrombin intrinsic pathways culminates in formation of prothrombinase
generation by way of the extrinsic pathway is limited by a natural complexes (phospholipid membrane bound activation complexes)

Blood
Microparticles

Thrombin Fibrin
Platelets

Endothelium
TF TF TF TF TF TF TF TF

Tissue Initiation Propagation Stabilization


Binding of platelets Recruitment of platelets
to collagen to growing thrombus Platelet-platelet
TF-dependent Amplification of interaction
initiation of coagulation coagulation cascade Fibrin deposition

Figure 56-2 Clot formation at vascular injury site. Vascular injury exposes subendothelial tissue factor initiating plasma-mediated hemostasis via the extrinsic
pathway. The intrinsic pathway further amplifies thrombin and fibrin generation. Platelets adhere to exposed collagen to undergo activation, resulting in
recruitment and aggregation of additional platelets. (From Mackman N, Tilley RE, Key NS: Role of extrinsic pathway of blood coagulation in hemostasis and
thrombosis. Arterioscleros Thromb Vasc Biol 27:1688, 2007, with permission.)
IV 1770 Anesthesia Management

Plasminogen The presence of fibrin accelerates plasmin generation.30 Rapid


- PAI-1
inhibition of free plasmin also limits spread of fibrinolytic activ-
+
Antifibrinolytic - t-PA ity. In addition to enzymatic degradation of fibrin and fibrinogen,
Urokinase plasmin inhibits hemostasis by degrading essential cofactors V
agents
and VIII as well as reducing platelet glycoprotein surface recep-
Plasmin tors essential to adhesion and aggregation.31 Fibrin degradation
- products possess mild anticoagulant properties as well.
a2-Antiplasmin TFPI inhibits the tissue factor/factor VIIa complex and
thereby the extrinsic coagulation pathway, which is responsible
Fibrin(ogen) for initiation of hemostasis. TFPI and factor Xa form phospho­
Fibrin(ogen) degradation lipid membrane-bound complexes that incorporate and inhibit
products tissue factor/factor VIIa complexes.3 Most TFPI is bound to vas-
cular endothelium but may be released into circulation by heparin
Figure 56-3 Principal mediators of fibrinolysis. Dashed lines depict sites of administration. Heparin further catalyzes TFPI inhibitory activ-
action for promoters and inhibitors of fibrinolysis. (From Slaughter TF: The
ity.32 As TFPI rapidly extinguishes tissue factor/VIIa activity, the
coagulation system and cardiac surgery. In Estafanous FG, Barash PG, Reves
JG [eds]: Cardiac Anesthesia: Principles and Clinical Practice, 2nd ed. critical role of the intrinsic pathway to continued thrombin and
Philadelphia, Lippincott Williams & Wilkins, 2001, p 320, with permission.) fibrin generation becomes apparent.22
The protein C system proves particularly important in
comprised of factor Xa, factor II (prothrombin), factor Va (cofac- downregulating hemostasis because it inhibits thrombin as well
tor), and calcium ions.25 Prothrombinase complexes mediate the as the essential cofactors Va and VIIIa. Thrombin initiates this
thrombin burst—a surge in thrombin generation from the inac- inhibitory pathway by binding a membrane-associated protein,
tive precursor prothrombin. Thrombin proteolytically cleaves thrombomodulin, to activate protein C.33 Protein C, complexed
fibrinopeptides A and B from fibrinogen molecules to generate with the cofactor protein S degrades both cofactors Va and VIIIa.
fibrin monomers, which polymerize into fibrin strands (i.e., fibrin Loss of these critical cofactors limits formation of “tenase” and
clot).25 Finally, factor XIIIa, a transglutaminase activated by “prothrombinase” activation complexes essential to formation of
thrombin, covalently crosslinks fibrin strands to produce an insol- factor X and thrombin, respectively. Thrombin, once bound to
uble fibrin clot resistant to fibrinolytic degradation.26 Again, thrombomodulin, is inactivated and removed from circulation,
thrombin generation is the key enzymatic step regulating hemo­ providing another mechanism by which protein C downregulates
stasis.27 Not only does thrombin activity mediate conversion of hemostasis.33
fibrinogen to fibrin, but it also activates platelets, converts inactive The most significant serine protease inhibitors regulating
cofactors V and VIII to active conformations, activates factor XI hemostasis include antithrombin (AT) and heparin cofactor II.
and the intrinsic pathway of coagulation, upregulates cellular Antithrombin binds to and inhibits thrombin as well as factors
expression of tissue factor, and stimulates vascular endothelial IXa, Xa, XIa, and XIIa. Heparin cofactor II inhibits thrombin
expression of PAI-1 to downregulate fibrinolytic activity.17,27 alone. Although the precise physiologic role for heparin cofactor
II remains uncertain, antithrombin plays a major role in down-
Intrinsic Anticoagulant Mechanisms regulating hemostasis.34 Heparin, a catalyst accelerator, binds
Once activated, regulation of hemostasis proves essential to limit antithrombin to promote inhibition of targeted enzymes.35
clot propagation beyond the injury site. One simple, yet impor- Heparin-like glycosaminoglycans, located on vascular endothelial
tant, anticoagulant mechanism derives from flowing blood and cells, provide inhibitory sites for thrombin and factor Xa in
hemodilution. The early platelet/fibrin clot proves highly suscep- vivo.
tible to disruption by shear forces within flowing blood. Blood
flow further limits localization and concentration of both plate-
lets and coagulation factors such that a critical mass of hemostatic
components may fail to coalesce.25,28 However, later in the clotting Disorders of Hemostasis
process more robust counterregulatory mechanisms are neces-
sary to limit clot propagation. Four major counterregulatory Evaluation of Bleeding Disorders
pathways have been identified that appear particularly crucial for
downregulating hemostasis: fibrinolysis, TFPI, the protein C Few would argue the importance of assessing bleeding risk preop-
system, and serine protease inhibitors. eratively; however, appropriate methods for ascertaining this risk
The fibrinolytic system comprises a cascade of amplifying remain subject to debate. Although routine coagulation testing of
reactions culminating in plasmin generation and proteolytic deg- all surgical patients preoperatively intuitively may be appealing,
radation of fibrin and fibrinogen. As with the plasma-mediated this approach lacks predictive value for bleeding disorders and
coagulation cascade, inactive precursor proteins are converted to certainly lacks cost effectiveness.36 A carefully performed history
active enzymes, necessitating a balanced system of regulatory of bleeding remains the single most effective predictor for peri-
controls to prevent excessive bleeding or thrombosis (Fig. 56-3). operative bleeding.
The principal enzymatic mediator of fibrinolysis, plasmin, is gen- A well-conducted history should focus on prior bleeding
erated from an inactive precursor, plasminogen.29 In vivo, plasmin episodes experienced by the patient.37 Does the patient have a
generation most often is initiated by release of t-PA or urokinase history of “excessive” bleeding in association with trauma or prior
from vascular endothelium. Thrombin provides a potent stimulus surgery? Were blood transfusions or reoperation required to
for t-PA synthesis.27 Factor XIIa and kallikrein of the intrinsic control the bleeding? A history suggestive of a bleeding disorder
pathway activate fibrinolysis after exposure to foreign surfaces. might include frequent epistaxis of a severity necessitating
Coagulation 1771 56
“packing the nasal passage” or surgical intervention. Oral surgery (i.e., Humate-P: Armour Pharmaceuticals, Kankakee, IL) may be
and dental extractions prove a particularly good test of hemosta- indicated.43,44
sis due to high concentrations of fibrinolytic activity in the oral Although relatively uncommon, the hemophilias merit
cavity. Von Willebrand’s disease not infrequently manifests as consideration given their diverse clinical presentation. Hemo-
menorrhagia, and postpartum hemorrhage commonly occurs in philia A, characterized by variable degrees of factor VIII defi-
women with underlying disorders of hemostasis.38 A history of ciency, is an X-linked inherited bleeding disorder most frequently
spontaneous hemorrhage (nontraumatic) proves particularly presenting in childhood as spontaneous hemorrhage involving
concerning when associated with joints (hemarthroses) or deep joints or deep muscles or both. Hemophilia A occurs with an

Section IV Anesthesia Management


muscles. Identification of a bleeding disorder at an early age or in incidence of 1 : 5000 males; however, nearly one third of cases
family members suggests an inherited, as opposed to acquired, represent new mutations with no family history.45 In mild cases,
condition.39 A careful medication history including direct ques- patients with hemophilia may not be identified until later in life,
tions relating to consumption of aspirin-containing nonprescrip- often after unexplained bleeding with surgery or trauma. Classi-
tion drugs, herbs, and fish oil may prove noteworthy. Finally, cally, laboratory testing in patients with hemophilia reveals pro-
inquiries regarding coexisting diseases should be included longation of the aPTT, whereas the prothrombin time (PT) and
(i.e., renal, hepatic, thyroid, bone marrow disorders, and bleeding time remain within normal limits. Specific measurement
malignancy). of factor VIII:C is required to confirm the diagnosis and to clarify
For most patients, a thoughtfully conducted bleeding the severity of factor VIII deficiency. Mild cases of hemophilia A
history will eliminate need for preoperative laboratory-based may be treated with desmopressin; however, in most cases peri-
coagulation testing. Regardless, there remain several valid reasons operative management of these patients necessitates consultation
for preoperative coagulation testing. Should the preoperative with a hematologist and administration of recombinant or puri-
history or physical examination reveal signs or symptoms sugges- fied factor VIII concentrates.43,46 An increasingly common com-
tive of a bleeding disorder further laboratory-based assessment plication of hemophilia, particularly in the case of hemophilia A,
of coagulation would be indicated. Preoperative screening tests of has been development of alloantibodies directed against the
coagulation may be indicated, despite a negative history, in cases factor VIII protein. In cases of high titer antibodies, administra-
in which major surgery commonly associated with significant tion of factor VIII concentrates may fail to control bleeding.
bleeding is planned (i.e., cardiopulmonary bypass). Finally, preop- Several approaches have reduced bleeding in these patients,
erative testing may prove justified in settings in which the patient including substitution of porcine factor VIII, administration of
is unable to provide a bleeding history preoperatively. Should “activated” or “non­activated” prothrombin complex concentrates,
evidence of a bleeding disorder be detected preoperatively, under- or treatment with recombinant factor VIIa (NovoSeven, Novo
lying mechanisms must be ascertained if possible before proceed- Nordisk, Copenhagen, Denmark) (see Chapter 57).44,47 Also
ing with surgery. inherited as an X-linked disorder, hemophilia B (i.e., Christmas
disease) occurs in approximately 1 : 40,000 and necessitates blood
component replacement with factor IX concentrates.
Inherited Bleeding Disorders
Although inherited disorders of hemostasis may involve platelet Acquired Bleeding Disorders
function, plasma-mediated hemostasis, or fibrinolytic pathways,
von Willebrand’s disease characterized by quantitative or qualita- A detailed account of acquired hemostatic disorders is beyond
tive deficiencies of vWF proves the most common of inherited the scope of this discussion; however, given that a limited number
bleeding disorders.38 Variants include types I and III with varying of drugs and coexisting medical conditions account for the major-
quantitative vWF deficiencies and type II comprising a collection ity of acquired bleeding disorders, these conditions merit consid-
of qualitative defects affecting vWF function. Under normal con- eration. Heparin, warfarin, and fibrinolytic drugs historically
ditions, vWF plays a critical role in platelet adhesion to ECM. accounted for most serious drug-induced bleeding complications
vWF further acts as a carrier molecule preventing proteolytic (Table 56-1).48,49 More recently, antiplatelet drug therapy further
degradation of factor VIII in free plasma.40 Classically, patients has complicated perioperative management (Table 56-2).49-52
with von Willebrand’s disease describe a history of easy bruising, Unfractionated heparin comprises a heterogeneous mixture of
recurrent epistaxis, and menorrhagia, all characteristic of defects membrane-associated glycosaminoglycans derived from either
in primary (i.e., platelet mediated) hemostasis. In more severe bovine or porcine mucosal tissues. Specificity and potency of
cases (i.e., type III vWD), concomitant reductions in factor unfractionated heparin varies by molecular weight, which ranges
VIII may lead to serious spontaneous hemorrhage, including between 5,000 and 30,000 daltons.35 Heparin derives its antico-
hemarthroses, which is common in hemophilia. Laboratory agulant effect by interacting with plasma antithrombin, which in
testing often demonstrates mild to moderate prolongation of the turn inhibits serine proteases participating in plasma-mediated
activated partial thromboplastin time (aPTT), prolonged bleed- hemostasis.34 The half-life of heparin is 1 to 2 hours and varies
ing time, decreased immunoreactive vWF concentrations, and directly with total dose. Heparin is cleared from the circulation
reduced platelet aggregation in response to ristocetin.38,41 Increas- both renally and hepatically. Most often, heparin’s anticoagulant
ingly, the PFA-100 and similar ex-vivo platelet function tests have effect is monitored using the aPTT with a target prolongation
replaced bleeding times in assessing for vWD.42 Measurable of one and one-half to two times control, commonly used for
reductions in factor VIII activity may occur in severe cases. Mild treatment of venous thrombosis.53 At heparin concentrations
cases of vWD often respond to desmopressin acetate (DDAVP); exceeding measurement limits of the aPTT, such as during
however, given a significant bleeding history, specific replacement cardiopulmonary bypass or interventional cardiovascular proce-
of vWF and factor VIII with select factor VIII concentrates dures, the activated clotting time (ACT) provides an alternative,
IV 1772 Anesthesia Management

Table 56-1 Anticoagulant Agents

Plasma Stop before Prolongation


Drug Site of Action Route Half-life Excretion Antidote Procedure of PT/aPTT

Unfractionated IIa/Xa IV/SC 1.5 hr Hepatic Protamine 6 hr No/Yes


heparin

LMWH Xa SC 4.5 hr Renal Protamine (partial 12-24 hr No/No


reversal)

Streptokinase Plg IV 23 min Hepatic Antifibrinolytics 3 hr Yes/Yes

t-PA Plg IV <5 min Hepatic Antifibrinolytics 1 hr Yes/Yes

Warfarin Vitamin K–dependent Oral 2-4 days Hepatic Vitamin K 2-4 days Yes/No
factors rfVIIa
PCCs
Plasma

Pentasaccharide Xa SC 14-17 hr Renal rfVIIa 4 days No/No

Bivalirudin IIa IV 25 min Hepatic None 2-3 hr Yes/Yes

Argatroban IIa IV 45 min Hepatic None 4-6 hr *Yes/Yes

Hirudin IIa IV 1.5 hr Renal PMMA, dialysis 8 hr *Yes/Yes

APC Va/VIIIa IV 2 hr Hepatic None 12 hr No/Yes

Ximelagatran IIa Oral 3 hr Renal None 24 hr Yes/Yes

*Argatroban and lepirudin may increase the normal PT 4 to 5 seconds.


APC, activated protein C; HIT, heparin-induced thrombocytopenia; IV, intravenous; LMWH, low molecular weight heparin; PCCs, prothrombin complex concentrates; Plg,
plasminogen; PMMA, polymethyl methylacrylate; rFVlla, recombinant factor VIIa; SC, subcutaneous; IIa, thrombin; t-PA, tissue plasminogen activator.
From Roberts HR, Monroe DM, Escobar MA: Current concepts of hemostasis: Implications for therapy. Anesthesiology 100:722-730, 2004, with permission.

albeit less sensitive, measure of heparin anticoagulation.54 ment of anti–factor Xa activity. Furthermore, should rapid reversal
Heparin’s anticoagulant effect is rapidly reversible by protamine of LMWH prove necessary, protamine proves only partially
administration.55 effective.57
More recently, low-molecular-weight heparins (LMWHs) Oral anticoagulant therapy in the form of coumarin deriva-
have gained favor as a result of reduced dosing frequency and the tives, interferes with hepatic synthesis of vitamin K–dependent
lack of need for monitoring. Owing to shorter saccharide chain coagulation factors: factors II, VII, IX, X, protein C, and protein
lengths, LMWHs exhibit reduced inhibitory activity toward S.58 Specifically, coumarin inhibits carboxylation of glutamic acid
thrombin while retaining factor Xa inhibitory activity.56 Theoreti- residues essential for anchoring coagulation factor activation
cally, LMWHs should be associated with reduced bleeding complexes to phospholipid membranes. Coumarin therapy is
tendencies. Regardless, LMWHs have a more predictable phar- monitored using the INR (International Normalized Ratio)
macokinetic response, fewer effects on platelet function, and a system derived from the PT. Generally, an INR of 2 to 3 is con-
reduced risk for heparin-induced thrombocytopenia (HIT). sidered reflective of adequate anticoagulation.53 In most cases,
Although monitoring of LMWHs is not performed routinely, the excessive coumarin anticoagulation is managed by withholding
PT and aPTT most often are unaffected, necessitating measure- drug administration. However, given a half-life of 2 to 4 days,

Table 56-2 Antiplatelet Agents

Drug Site of Action Route Plasma Half-life Metabolism Antidote Stop before Procedure Prolongation of PT/aPTT

Aspirin COX 1-2 Oral 20 min Hepatic None 7 days No/No

Dipyridamole Adenosine Oral 40 min Hepatic None 24 hr No/No

Clopidogrel ADP Oral 7 hr Hepatic None 5 days No/No

Ticlopidine ADP Oral 4 days Hepatic None 10 days No/No

Abciximab GPIIb-IIIa IV 30 min Renal None 72 hr No/No

Eptifibatide GPIIb-IIIa IV 2.5 hr Renal None 24 hr No/No

Tirofiban GPIIb-IIIa IV 2 hr Renal Hemodialysis 24 hr No/No

ADP, adenosine diphosphate; COX, cyclooxygenase diphosphate; IV, intravenous; GP, glycoprotein.
From Roberts HR, Monroe DM, Escobar MA: Current concepts of hemostasis: Implications for therapy. Anesthesiology 100:722-730, 2004, with permission.
Coagulation 1773 56
more rapid reversal in perioperative settings may be needed. coagulation. Numerous underlying disorders may precipitate
Vitamin K administered orally or parentally in doses of 1 to 2 mg DIC, including trauma, amniotic fluid embolus, malignancy,
reverses coumarin anticoagulation.58 More rapid reversal may be sepsis, or incompatible blood transfusions.62 Most often, DIC
achieved by administration of plasma or prothrombin complex presents clinically as a diffuse bleeding disorder associated with
concentrates.59 Recombinant factor VIIa also has proved effective consumption of coagulation factors and platelets during wide-
at reversing coumarin-associated bleeding. spread microvascular thrombotic activity. Underlying fibrinolysis,
Despite relatively short half-lives and increasing fibrin spe- providing a counterregulatory mechanism to maintain vascular
cificity, fibrinolytic drug administration continues to be associ- integrity, proves common as well. Laboratory findings typical of

Section IV Anesthesia Management


ated with significant bleeding potential. Bleeding after fibrinolytic DIC include reductions in platelet count, prolongation of the PT,
drugs proves multifactorial in origin, resulting from proteolytic aPTT, and thrombin time (TT), and elevated concentrations of
degradation of fibrin and fibrinogen, proteolysis of cofactors V soluble fibrin and fibrin(ogen) degradation products. Chronic
and VIII, and digestion of platelet glycoprotein surface recep- DIC states have been identified with relatively normal screening
tors.29 In addition, both fibrin and fibrinogen degradation frag- tests of coagulation accompanied by elevated concentrations of
ments impair platelet function and inhibit assembly of fibrin soluble fibrin and fibrin(ogen) degradation products.63 Manage-
monomers into strands. Once the fibrinolytic agent is cleared ment of DIC requires alleviating the underlying condition pre-
from plasma, antifibrinolytic drugs prove of limited benefit. cipitating hemostatic activation. Otherwise, treatment includes
Therapy must rely on selective repletion of platelet and plasma selective blood component transfusions to replete coagulation
protein constituents. In many cases, cryoprecipitate will be admin- factors and platelets consumed in the process. Antifibrinolytic
istered to replace fibrinogen and platelet infusions will be needed therapy generally is contraindicated in DIC owing to potential for
to compensate for qualitative platelet dysfunction. Plasma infu- catastrophic thrombotic complications.
sions may be required to replete plasma coagulation factors, par-
ticularly cofactors V and VIII.
Prothrombotic States
Liver Disease
Hemostatic defects associated with hepatic failure prove complex Thrombophilia, a propensity for thrombotic events, most often
and multifactorial. Severe liver disease impairs synthesis of coag- manifests clinically in the form of venous thrombosis (frequently
ulation factors, impedes clearance of activated clotting and fibri- deep venous thrombosis of the lower extremity).64 As with bleed-
nolytic proteins, and produces quantitative and qualitative platelet ing disorders, thrombophilia may result from inherited or
dysfunction. Laboratory findings commonly associated with liver acquired conditions (Table 56-3). In the majority of cases, a pre-
disease include a prolonged PT, possible prolongation of the cipitating event may be identified.65 For example, thrombotic
aPTT, and thrombocytopenia.60 Prolongation of bleeding time complications often occur after surgery, during pregnancy, and in
reflects qualitative defects in platelet adhesion and aggregation. association with obesity or underlying malignancy.66 Random
Treatment of bleeding in the setting of liver disease most often is screening of asymptomatic patients for thrombotic risk has not
based on laboratory abnormalities. Plasma and platelets fre- proved cost effective or clinically efficacious.67,68 As with bleeding
quently are administered for acute bleeding. Low fibrinogen con- disorders, a careful history focusing on prior thrombotic events,
centrations may necessitate cryoprecipitate administration. In family history of thrombosis, and concurrent drug therapy offers
addition, low-grade fibrinolysis may benefit from antifibrinolytic greater predictive value than random screening.
drug administration in select settings.

Renal Disease Common Inherited Thrombotic Disorders


Platelet dysfunction commonly occurs in association with chronic
renal failure, as reflected by a prolonged bleeding time and Although understanding of thrombotic disorders from a
propensity for bleeding associated with surgery or trauma. biochemical and molecular level remains limited at present,
Underlying mechanisms appear multifactorial and may include careful testing may identify an underlying heritable thrombotic
accumulation of guanidinosuccinic acid and nitric oxide in
uremic plasma.61 In addition, red blood cell concentration has
Table 56-3 Hypercoagulable States and Risk for Perioperative Thrombosis
been speculated to play a role as correction of anemia results in
shortened bleeding times, presumably related to the role of red High Risk
blood cells in causing platelet margination along the vessel wall Heparin-induced thrombocytopenia (HIT)
Antithrombin deficiency
under laminar flow conditions.28 Both dialysis and correction of
Protein C deficiency
anemia have been reported to shorten bleeding times in patients
Protein S deficiency
with chronic renal failure. Traditionally, cryoprecipitate adminis- Antiphospholipid antibody syndrome
tration occurs in this setting; however, desmopressin (0.3 µg/kg)
and conjugated estrogens similarly may shorten bleeding times.43 Moderate Risk
Risks associated with these therapies, and the predictive value of Factor V Leiden genetic polymorphism
bleeding time for subsequent hemorrhage, remain unclear. Prothrombin G20210A genetic polymorphism
Hyperhomocysteinemia
Disseminated Intravascular Coagulation Dysfibrinogenemia
Disseminated intravascular coagulation (DIC) comprises a patho- Postoperative prothrombotic state
Malignancy
physiologic hemostatic response to tissue factor/factor VIIa
Immobilization
complex exposure and activation of the extrinsic pathway of
IV 1774 Anesthesia Management

predisposition in as many as 50% of patients with venous throm- agulant or anticardiolipin antibodies.72 Despite the prolonged
boembolism.69 Relatively common inherited conditions underly- aPTT, antiphospholipid syndrome poses no increased bleeding
ing prothrombotic tendencies include single point mutations in risk but rather increases the potential for thrombosis. Antibodies
genes for factor V (factor V Leiden) or prothrombin (prothrombin associated with antiphospholipid syndrome interfere with phos-
G20210A). In the case of the factor V Leiden mutation, the essen- pholipids common to many laboratory-based tests of coagulation.
tial cofactor Va acquires resistance to degradation by protein C.70 Isolated prolongation of an aPTT in a preoperative patient merits
This simple alteration in balance between hemostasis and the consideration of the diagnosis of antiphospholipid syndrome.
protein C counterregulatory system induces a prothrombotic ten- Patients with this syndrome who have experienced a thrombotic
dency present in approximately 5% of the population. In the case complication are at increased risk for recurrent thrombosis and
of the prothrombin gene mutation, increased prothrombin con- most often are managed by life-long anticoagulation.73
centrations in plasma again generate a hypercoagulable state. Less
common inherited forms of thrombophilia include deficiencies Heparin-Induced Thrombocytopenia
of antithrombin, protein C, and protein S.34 Inherited forms of Heparin-induced thrombocytopenia describes an autoimmune-
thrombophilia are characterized by highly variable penetrance mediated drug reaction occurring in as many as 5% of patients
affected by blood type, sex, and other confounding variables. In receiving heparin therapy. As opposed to other drug-induced
the absence of coexisting precipitating conditions, absolute thrombocytopenias, HIT results in platelet activation and poten-
thrombotic risk secondary to a heritable thrombophilia proves tial for venous and arterial thromboses.74,75 Evidence suggests that
limited.71 In the presence of a family history or test abnormality HIT is mediated by immune complexes (composed of IgG anti-
suggesting thrombophilia with no history of thrombosis, risks body, platelet factor 4 [PF4], and heparin) that bind platelet Fcγ
associated with long-term preventive anticoagulation may out- receptors to activate platelets. Anti-PF4/heparin antibodies may
weigh potential benefits. After a thrombotic complication, “activate” vascular endothelium, monocytes, and macrophages
however, these patients most often are managed with life-long by upregulating tissue factor expression (Fig. 56-4). Patients
anticoagulation. developing HIT during heparin therapy experience substantially
increased risk for thrombosis (odds ratio 20-40, absolute risk
30%-75%).74,75
Common Acquired Thrombotic Disorders In most cases, HIT manifests clinically as thrombocytope-
nia occurring 5 to 14 days after initiating heparin therapy.
Antiphospholipid Syndrome However, with prior heparin exposure, thrombocytopenia or
Antiphospholipid syndrome describes an acquired autoimmune thrombosis may occur within 1 day. In some reported cases,
disorder characterized by venous and/or arterial thromboses as thrombosis occurred simultaneously with thrombocytopenia,
well as recurrent pregnancy loss. This syndrome may occur sec- providing no warning. Although HIT remains a clinical diagnosis,
ondarily to autoimmune disorders such as systemic lupus ery- laboratory testing provides confirmatory evidence. Both func-
thematosus or rheumatoid arthritis, or it may occur in isolation. tional (i.e., serotonin platelet release assays or heparin-induced
Characteristically, antiphospholipid syndrome results in mild platelet aggregation) and quantitative (i.e., solid-phase enzyme-
prolongation of the aPTT and positive testing for lupus antico- linked immunosorbent assays [ELISA]) tests for PF4/heparin

Heparin 4 IgG
PF

PF4

Activated
platelet
PF

F4
4

P
Procoagulant
microparticles
PF4
IgG

Vascular Heparan sulfate


endothelium Clot PF4

Figure 56-4 Mechanisms underlying thrombosis in heparin-induced thrombocytopenia (HIT). Immune complexes composed of heparin, platelet factor 4 (PF4),
and antibodies bind to platelet surface Fcγ receptors to activate platelets. PF4/heparin immune complexes further activate vascular endothelium, monocytes,
and macrophages to increase tissue factor expression. (From Slaughter TF, Greenberg CS: Heparin-associated thrombocytopenia and thrombosis: Implications
for perioperative management. Anesthesiology 87:669, 1997, with permission.)
Coagulation 1775 56
immune complexes prove beneficial.75 Recent evidence suggests sample of patient plasma with phospholipid, calcium, and an
that PF4/heparin antibodies alone, in the absence of thrombocy- activator of the intrinsic pathway of coagulation (i.e., celite, kaolin,
topenia or thrombosis, may predict adverse outcomes.76,77 silica, or ellagic acid). In most cases, coagulation factor deficien-
A diagnosis of HIT should be entertained for any patient cies are detectable at factor concentrations below 30% to 40% of
experiencing thrombosis or thrombocytopenia (absolute or rela- normal; however, aPTT reagents vary in sensitivity to factor con-
tive [≥ 50% reduction in platelet count]) during or after heparin centrations, necessitating institutional specific determination of
administration. In cases in which HIT is suspected, heparin must “normal” ranges.81 Prolongation of the aPTT is evaluated further
be discontinued immediately (i.e., including unfractionated with mixing studies to determine whether delayed clot formation

Section IV Anesthesia Management


heparin, heparin-bonded catheters, heparin flushes, LMWH). is attributable to a coagulation factor deficiency or an inhibitor
Alternative non-heparin anticoagulation must be administered (i.e., heparin, antiphospholipid antibody, fibrin split products).
concurrently. In most cases, a direct thrombin inhibitor (i.e., bival­ The mixing study is performed by mixing the patient’s plasma
irudin, lepirudin, argatroban) is substituted for heparin until sample with “normal” donor plasma. In the case of a coagulation
adequate prolongation of the INR may be achieved with warfa- factor deficiency, time to clot formation will correct and sequen-
rin.78,79 Typically, PF4/heparin immune complexes clear from the tial assays for specific coagulation factor concentrations allow for
circulation within 3 months. If possible, patients experiencing identification of the deficiency. A common means of confirming
HIT should avoid future exposure to unfractionated heparin; heparin prolongation of the aPTT is to perform a thrombin clot-
however, several reports describe subsequent limited periopera- ting time (TCT). In this test, the patient’s citrate-containing
tive reexposure to unfractionated heparin after laboratory testing plasma sample is mixed with thrombin and time to clot formation
to ensure absence of PF4/heparin immune complexes. is measured in seconds.39 The most common mechanism under-
lying prolongation of the TCT is presence of heparin or a direct
thrombin inhibitor anticoagulant; however, dysfibrinogenemia or
fibrin(ogen) degradation products also may impair clot formation
Monitoring Coagulation in this setting.

Traditionally, perioperative coagulation monitoring has focused Prothrombin Time


on (1) preoperative testing to identify patients at increased risk The PT assesses integrity of the extrinsic and common pathways
for perioperative bleeding and (2) intraoperative monitoring of of plasma-mediated hemostasis. It measures time required in
heparin therapy during cardiac and vascular surgery. As discussed seconds for clot formation to occur after mixing a sample of
previously, routine preoperative coagulation testing lacks either patient plasma with tissue factor (thromboplastin) and calcium.
predictive value or cost efficacy.80 However, in patients with a As with the aPTT, thromboplastin test reagents vary in sensitivity,
history suggestive of a bleeding disorder or whose surgery may limiting the ability to compare results between laboratories. Given
be associated with a high incidence of bleeding or coagulopathy, the importance of monitoring PT results for patients on long-
baseline preoperative coagulation testing may be advisable. term warfarin therapy, the INR was introduced as a means of
Given the widely acknowledged pharmacokinetic and normalizing PT results among different laboratories.81 Thrombo-
pharmacodynamic response to heparin, monitoring anticoagula- plastin reagents are tested against an international recombinant
tion during cardiac and vascular surgery remains common. standard and assigned an international sensitivity index (ISI)
Patient-specific factors affecting response to heparin include age, based on results. The INR subsequently is calculated as INR =
weight, intravascular volume, and plasma/membrane concentra- (patient PT/standard PT)ISI in which the “standard PT” represents
tions of antithrombin, heparin cofactor II, platelet factor 4, and the geometric mean of multiple normal samples from the testing
other heparin binding proteins. Therefore, patients experience laboratory. Institution of the INR substantially reduced interlabo-
widely divergent anticoagulant responses to identical weight- ratory variations in the PT. As with the aPTT, prolongation of the
based doses of heparin. Regardless, monitoring of heparin anti- PT is assessed further with mixing studies.
coagulation must be performed if only to prevent the rare, but
potentially lethal, complication resulting from inadvertent failure Platelet Count and Bleeding Time
to administer heparin before cardiopulmonary bypass or vascular The platelet count remains a standard component in screening
surgery. for coagulation abnormalities. Automated platelet counts are per-
The ideal test of perioperative coagulation would prove formed in bulk using either optical- or impedance-based meas-
simple to perform, accurate, reproducible, diagnostically specific, urements. Recommendations regarding optimal platelet counts
and cost effective. No current coagulation monitor meets these prove somewhat arbitrary; however, platelet counts exceeding
expectations; however, integrating results from multiple forms of 100,000/µL usually are associated with normal hemostasis.
monitoring may provide valuable diagnostic insight into periop- Abnormally low platelet counts merit further assessment, includ-
erative coagulopathies. ing a visual platelet count using a blood smear. Sample hemodilu-
tion and platelet clumping are common causes for false low
platelet counts. EDTA-induced platelet clumping, a common arti-
fact explaining thrombocytopenia, may be identified by manually
Common Laboratory-Based Measures counting platelets or by substituting a citrate anticoagulant, as
of Coagulation opposed to EDTA common to platelet collection vacutainers.
Although the bleeding time, as a test of platelet dysfunc-
Activated Partial Thromboplastin Time tion, has lost much of its popularity in the past 10 years, many
The aPTT assesses integrity of the intrinsic and common path- clinicians continue to rely on bleeding time assessment as a
ways of plasma-mediated hemostasis. It measures the time measure of platelet function and primary hemostasis. With the
required in seconds for clot formation to occur after mixing a use of a commercial template cutting device and a sphygmoma-
IV 1776 Anesthesia Management

nometer adjusted to 40 mm Hg venous pressure, bleeding times anticoagulants prolong time to clot formation; however, the
of 2 to 10 minutes on the anterior forearm are considered normal. ACT proves somewhat resistant to platelet dysfunction and
Limitations of the bleeding time include poor reproducibility, thrombocytopenia.
time needed to perform the test, and potential for scarring. Fur- ACT testing remains a popular perioperative coagulation
thermore, the bleeding time is affected by numerous confounding monitor because of its simplicity, low cost, and linear operating
variables, including skin temperature, skin thickness, age, ethnic- response at high heparin concentrations. Limitations of ACT
ity, anatomic test location, and a host of other factors.82 In general, monitoring include lack of sensitivity at low heparin concentra-
the bleeding time has not proved predictive of bleeding and for tions and poor reproducibility.83 ACT tubes containing hepari-
that reason its use as a preoperative screening test to assess bleed- nase improve sensitivity for detecting low heparin concentrations.
ing risk is not recommended. Further limitations of the ACT include artifactual prolongation
of results with hemodilution or hypothermia and the fact that
ACT values beyond 600 seconds exceed the linear response range
for the assay. Duplicate measurements improve results; however,
Common Point-of-Care Measures newer electrochemically based ACT analyzers (ISTAT; Abbott
of Coagulation Laboratories, Chicago, IL) improve reproducibility such that
single ACT determinations may prove adequate.
Although laboratory-based measures of coagulation remain the The high-dose thrombin time (HiTT) (International Tech-
mainstay of preoperative coagulation testing, increasing availabil- nidyne, Edison, NJ) provides an alternative functional measure
ity of sensitive and specific point-of-care coagulation monitoring of heparin anticoagulation. Primarily employed in cardiac surgi-
may soon offer opportunities to direct blood component and cal settings, the HiTT assay contains high thrombin concentra-
hemostatic drug therapy more specifically without delays inher- tions to cleave fibrinogen directly and generate fibrin clot.84 Given
ent to standard laboratory testing. Commercially available point- excess thrombin concentrations, clot formation occurs independ-
of-care monitors applicable in the perioperative setting may be ently of plasma coagulation factors other than fibrinogen. As a
considered in four broad categories: (1) functional measures of result, HiTT is prolonged by heparin (or other thrombin inhibi-
coagulation or assays that measure the intrinsic ability of blood tors), extreme degrees of hypofibrinogenemia or dysfibrinogene-
to generate clot; (2) heparin concentration monitors; (3) visco­ mia, and high concentrations of fibrin split products (as occur
elastic measures of coagulation; and (4) platelet function with fibrinolysis). During most surgical procedures requiring
monitors. heparin administration, HiTT prolongation will correlate with
heparin anticoagulant effect.
Functional Measures of Coagulation
The activated coagulation time (ACT), described by Hattersley in Heparin Concentration Measurement
1966 as a variation of the Lee-White whole blood clotting time, Protamine titration remains the most popular point-of-care
employs a contact activation initiator, typically celite (diatoma- method for determining heparin concentration in perioperative
ceous earth) or kaolin, to accelerate clot formation and reduce settings. Protamine, a strongly basic polycationic protein, directly
time for assay completion. Kaolin is recommended for use in inhibits heparin in a stoichiometric manner. In other words, 1 mg
patients receiving the antifibrinolytic drug aprotinin because of protamine will inhibit 1 mg (approximately 100 U) of heparin,
celite ACTs in this setting are subject to artifactual prolongation.83 thereby forming the basis for protamine titration as a measure of
Failure to appreciate this drug-mediated effect may result in inad- heparin concentration. As increasing concentrations of protamine
equate heparin administration if using celite ACT measurements are added to a sample of heparin-containing blood, time to clot
with aprotinin. Whether high plasma concentrations of aprotinin formation decreases until the point at which the protamine con-
may affect the kaolin ACT remains unclear. centration exceeds heparin concentration to delay clot formation.
Current commercial ACT monitors automate clot detec- If a series of blood samples with incremental doses of protamine
tion. One of the more widely available ACT monitors uses a glass are analyzed, the sample in which the protamine and heparin
test tube containing a small magnet (Hemochron; International concentrations are most closely matched will clot first. In this
Technidyne, Inc., Edison, NJ). After adding sample blood, the manner, protamine titration methodology allows for heparin con-
tube is placed into the analyzer and the tube is rotated slowly at centration estimation. Assuming that the heparin-protamine
37°C, allowing the magnet to maintain contact with a proximity titration curve for an individual patient remains constant through-
detection switch. As fibrin clot forms, the magnet becomes out the operative period, protamine titration methods may esti-
entrapped and dislodged from the detection switch, thereby trig- mate heparin doses required to achieve a desired plasma heparin
gering an alarm to signal completion of the ACT. Another ACT concentration or the protamine dose needed to reverse a given
device uses a “plumb bob” flag assembly that is raised and released heparin concentration in blood.54 Current point-of-care heparin
repeatedly to settle in the sample vial containing blood and concentration monitors employ automated measurement tech-
contact activator (Hepcon and ACT II; Medtronic Blood Manage- niques (Hepcon HMS; Medtronic Blood Management, Parker,
ment, Parker, CO). With clot formation, the descent rate of the CO). Advantages of heparin concentration measurement include
flag through the blood sample slows, triggers an optical detector, sensitivity for low heparin concentrations as well as relative
and sets off an alarm to signal completion of the ACT. insensitivity to hemodilution and hypothermia. In addition,
The ACT in normal individuals is 107 ± 13 seconds (mean heparin concentration measures are unaffected by aprotinin.
± SD). However, the time at which a patient’s “baseline” ACT is A major limitation of heparin concentration monitoring is
measured influences the result.83 After surgical incision, baseline failure to assess directly for an anticoagulant effect. To use an
ACT may decrease. Because the ACT measures clot formation extreme example, consider a patient with a homozygous defi-
by way of intrinsic and common pathways, heparin and other ciency of antithrombin; in this setting, heparin concentration
Coagulation 1777 56
determination alone would fail to identify the lack of anticoagu- proved costly, time consuming, and technically demanding.
lant effect after heparin administration. Platelet dysfunction may occur as a result of diverse inherited or
acquired disorders affecting surface receptors involved in adhe-
Viscoelastic Measures of Coagulation sion or aggregation, storage granules, internal activation path-
Initially developed in the 1940s, viscoelastic measures of coagula- ways, phospholipid membranes, or other mechanisms.51 Lack of
tion have undergone a resurgence in popularity. The unique standardized quality controls necessitates use of local donors’
aspect of viscoelastic monitors lies in their ability to measure the blood to establish normal control ranges. Furthermore, even
entire spectrum of clot formation from early fibrin strand genera- established tests such as platelet aggregation lack performance

Section IV Anesthesia Management


tion through clot retraction and eventual fibrinolysis. The throm- standardization resulting in widely varying results among labora-
boelastograph (TEG) (Haemoscope, Niles, IL), developed by tories. Complicating assessment further is the fact that platelets
Hartert in 1948, uses a small 0.35-mL blood sample placed into are highly susceptible to activation or desensitization during
a disposable cuvette within the instrument. The cuvette is main- sample collection, transport, storage, and processing.
tained at a temperature of 37°C and continuously rotates around Although viscoelastic measures of coagulation (i.e., TEG/
an axis of approximately 5 degrees. A metal piston attached by a Sonoclot) may detect severe platelet dysfunction, sensitivity and
torsion wire to an electronic recorder is lowered into the blood specificity are limited. The standard laboratory-based method for
within the cuvette. As clot formation occurs, the piston becomes identifying qualitative platelet dysfunction remains optical
enmeshed within the clot and rotation of the cuvette is trans- aggregometry performed using specific platelet agonists and
ferred to the piston, torsion wire, and electronic recorder.85 samples of platelet-rich plasma. Flow cytometry employing fluo-
Although variables derived from the TEG tracing do not rescent-labeled antibodies provides another sensitive method for
coincide directly with laboratory-based tests of coagulation, the quantitating platelet activation, responsiveness, and surface recep-
TEG depicts characteristic abnormalities in clot formation and tor availability.87 Despite representing standards of care, these
fibrinolysis. Various parameters describing clot formation and measures remain technically challenging, costly, and time-
lysis are identified and measured by the TEG. For example, the R consuming laboratory-based assays.
value (reaction time) measures time to initial clot formation Fortunately, an increasing array of platelet function assays
(normal, 7.5 to 15 minutes). Comparable to the whole blood clot- specifically designed as point-of-care instruments are available.88,89
ting time, addition of a contact activator such as celite or kaolin As a measure of primary hemostasis, the PFA-100 (Platelet Func-
to the sample cuvette reduces time to results. The R value may be tion Analyzer; Dade International Inc, Miami, FL) increasingly
prolonged by a deficiency of one or more plasma coagulation has replaced the bleeding time in assessment of primary hemo­
factors or inhibitors such as heparin. Maximum amplitude (MA) stasis. Unique among both laboratory-based and point-of-care
provides a measure of clot strength and may be decreased by platelet function monitors, the PFA-100 incorporates high-shear
either qualitative or quantitative platelet dysfunction or decreased conditions to simulate small vessel injury in the presence of either
fibrinogen concentration. Normal MA is 50 to 60 mm. The alpha ADP or epinephrine, both potent platelet activators.90 Time to
angle and K (BiKoatugulierung or coagulation) values measure clot–mediated aperture occlusion is reported as “closure time.”
rate of clot formation and may be prolonged by any variable The PFA-100 has proved effective in detecting von Willebrand’s
slowing clot generation such as a plasma coagulation factor defi- disease as well as aspirin-mediated platelet dysfunction. This
ciency or heparin anticoagulation.86 instrument, as a component of a standardized screening protocol,
The Sonoclot (Sienco, Inc., Wheat Ridge, CO) provides an reduces time to identify and classify platelet dysfunction. Limita-
alternative viscoelastic measure of coagulation. In contrast to the tions of the PFA-100 include interference by thrombocytopenia
TEG, Sonoclot immerses a rapidly vibrating probe into a 0.4-mL and hemodilution.
sample of blood. As clot formation occurs, impedance to probe The HemoSTATUS (Medtronic Blood Systems, Parker,
movement through the blood increases and generates an electri- CO) platelet function test exploits the ability of platelet activating
cal signal and characteristic clot “signature.” The Sonoclot may be factor (PAF) to accelerate clot formation of the kaolin-activated
used to derive the ACT, as well as to provide information regard- ACT. HemoSTATUS testing is performed on the Medtronic HMS
ing clot strength and fibrinolysis. coagulation analyzer employing a 6-channel kaolin ACT car-
Both the TEG and Sonoclot generate characteristic dia- tridge preloaded with serially increasing concentrations of PAF.
grams by translating mechanical resistance to sensor movement A clot ratio is determined for each individual patient assay based
in a blood sample to a waveform. One of the more common on the ratio of the PAF-accelerated ACT to the standard ACT. An
applications for the TEG has been real-time detection of excess individual patient’s clot ratio is compared with a maximal clot
fibrinolysis during liver transplantation. The TEG also may prove ratio derived from normal volunteers to provide a relative measure
beneficial in differentiating surgically related bleeding from that of platelet function. In the presence of platelet dysfunction, higher
of a coagulopathy. More widespread application of viscoelastic concentrations of PAF are required to achieve a comparable PAF-
coagulation monitoring has been hindered by lack of specificity activated ACT. In cardiac surgery, investigators demonstrated a
associated with abnormal findings and qualitative assay interpre- relationship between platelet dysfunction and postoperative
tation.86 Digital automation of these instruments has simplified bleeding.83 Furthermore, the maximal clot ratio as determined by
and improved reproducibility. HemoSTATUS improved after desmopressin or platelet adminis-
tration. Other investigators, however, failed to identify a relation-
ship between platelet dysfunction as measured by HemoSTATUS
Platelet Function Monitors and postoperative bleeding.
The Rapid Platelet Function Analyzer (RPFA; Accumetrix,
Assessment of platelet function has proved challenging for a San Diego, CA) is an automated turbidimetric whole blood plate-
number of reasons. Historically, tests of platelet function have let function assay that assesses ability of activated platelets to bind
IV 1778 Anesthesia Management

fibrinogen-coated polystyrene beads. On addition of the whole In preliminary investigations, platelet contractile force was
blood test sample, thrombin receptor activating peptide (TRAP) reduced after cardiopulmonary bypass and modestly correlated
directly activates platelets within the sample to stimulate glyco- with perioperative blood loss.
protein IIb/IIIa platelet surface receptor expression. As activated Advances in our understanding of hemostasis and throm-
platelets bind and aggregate fibrinogen-coated beads, light trans- bosis at the molecular level have contributed directly to recent
mission through the sample increases to generate a signal. biotechnologic innovations in point-of-care testing. Further
Although the RPFA is simple to operate and provides a rapid advances in point-of-care coagulation monitoring offer the
bedside measure of platelet function, a baseline reference meas- opportunity for clinicians to make informed decisions about
urement is required for each patient to calculate the extent of transfusion therapy and hemostatic drug administration to mini-
subsequent changes in platelet function.88 Potential applications mize perioperative bleeding. In considering any point-of-care
of this methodology in the perioperative setting remain unclear. coagulation testing, one must recognize that results will not nec-
Plateletworks (Helena Laboratories, Beaumont, TX) essarily mirror those reported from laboratory-based testing.
employs a hemocytometer to perform automated platelet counts Reagent sensitivities vary from manufacturer to manufacturer
on whole blood samples collected in the presence and absence of and even one lot of reagent to another. In addition, point-of-care
platelet-stimulating agonists such as collagen or ADP. The differ- testing typically relies on whole blood samples for testing as
ence in platelet counts before and after agonist addition provides opposed to laboratory-based testing using plasma or processed
a direct measure of platelet aggregation (i.e., platelet responsive- platelets. A final consideration for which point-of-care platelet
ness) and is reported as “% aggregation.” Preliminary investiga- function testing provides a particularly relevant example is that
tions demonstrated reasonable correlations between platelet monitors from different manufacturers measure differing aspects
counts performed with the Ichor hematology analyzer and of platelet- or plasma-mediated hemostasis. In the case of platelet
laboratory-based instruments.88 In addition, the Plateletworks function monitors, a dozen instruments purport to measure
assay proved effective in identifying platelet dysfunction in the “platelet function.” However, using different instruments one may
setting of glycoprotein IIb/IIIa antagonists and appears to obtain results varying from “severe” platelet dysfunction to “no
correlate relatively well with laboratory-based measures of plate- platelet dysfunction” in a single sample of blood. Before adopting
let aggregation. any point-of-care monitoring, an understanding of the quality
Employing a sample of whole blood, the Hemodyne assurance requirements, test methodology, and concomitant
Hemostasis Analyzer (Hemodyne, Richmond, VA) measures strengths and weaknesses will be essential to benefit patient
platelet contractile force, the force produced by platelets during care.
clot retraction, and clot elastic modulus, a measure of clot rigidity.

References
1. Sagripanti A, Carpi A: Antithrombotic and pro- 11. Andrews RK, Berndt MC: Platelet physiology and 23. Hoffman M: A cell-based model of coagulation and
thrombotic activities of the vascular endothelium. thrombosis. Thromb Res 114:447-453, 2004. the role of factor VIIa. Blood Rev 17(Suppl 1):S1-S5,
Biomed Pharmacother 54:107-111, 2000. 12. Chen J, Lopez JA: Interactions of platelets with 2003.
2. Freedman JE, Loscalzo J: Nitric oxide and its relation- subendothelium and endothelium. Microcirculation 24. Esmon CT: Crosstalk between inflammation and
ship to thrombotic disorders. J Thromb Haemost 12:235-246, 2005. thrombosis. Maturitas 47:305-314, 2004.
1:1183-1188, 2003. 13. Chaer RA, Graham JA, Mureebe L: Platelet function 25. Furie B, Furie BC: In vivo thrombus formation. J
3. Kato H: Regulation of functions of vascular wall cells and pharmacologic inhibition. Vasc Endovascular Thromb Haemost 5(Suppl 1):12-17, 2007.
by tissue factor pathway inhibitor: Basic and clinical Surg 40:261-267, 2006. 26. Mosesson MW: Fibrinogen and fibrin structure
aspects. Arterioscler Thromb Vasc Biol 22:539-548, 14. Furie B, Furie BC: Thrombus formation in vivo. J Clin and functions. J Thromb Haemost 3:1894-1904,
2002. Invest 115:3355-3362, 2005. 2005.
4. May AE, Neumann FJ, Preissner KT: The relevance 15. Fullard JF: The role of the platelet glycoprotein IIb/ 27. Crawley JT, Zanardelli S, Chion CK, Lane DA: The
of blood cell–vessel wall adhesive interactions for IIIa in thrombosis and haemostasis. Curr Pharm Des central role of thrombin in hemostasis. J Thromb
vascular thrombotic disease. Thromb Haemost 10:1567-1576, 2004. Haemost 5(Suppl 1):95-101, 2007.
82:962-970, 1999. 16. Hoffman M: Remodeling the blood coagulation 28. Barshtein G, Ben-Ami R, Yedgar S: Role of red blood
5. Wu KK, Thiagarajan P: Role of endothelium in cascade. J Thromb Thrombolysis 16:17-20, 2003. cell flow behavior in hemodynamics and hemostasis.
thrombosis and hemostasis. Annu Rev Med 47:315- 17. Coughlin SR: Protease-activated receptors in hemo­ Expert Rev Cardiovasc Ther 5:743-752, 2007.
331, 1996. stasis, thrombosis and vascular biology. J Thromb 29. Medcalf RL: Fibrinolysis, inflammation, and regula-
6. Grignani G, Maiolo A: Cytokines and hemostasis. Haemost 3:1800-1814, 2005. tion of the plasminogen activating system. J Thromb
Haematologica 85:967-972, 2000. 18. Schenone M, Furie BC, Furie B: The blood coagula- Haemost 5(Suppl 1):132-142, 2007.
7. Westrick RJ, Eitzman DT: Plasminogen activator tion cascade. Curr Opin Hematol 11:272-277, 30. Cesarman-Maus G, Hajjar KA: Molecular mecha-
inhibitor-1 in vascular thrombosis. Curr Drug Targets 2004. nisms of fibrinolysis. Br J Haematol 129:307-321,
8:966-1002, 2007. 19. Mann KG, Brummel-Ziedins K, Orfeo T, Butenas S: 2005.
8. Parolari A, Mussoni L, Frigerio M, et al: Increased Models of blood coagulation. Blood Cells Mol Dis 31. Andrews RK, Karunakaran D, Gardiner EE, Berndt
prothrombotic state lasting as long as one month after 36:108-117, 2006. MC: Platelet receptor proteolysis: A mechanism
on-pump and off-pump coronary surgery. J Thorac 20. Gomez K, McVey JH: Tissue factor initiated for downregulating platelet reactivity. Arterioscler
Cardiovasc Surg 130:303-308, 2005. blood coagulation. Front Biosci 11:1349-1359, Thromb Vasc Biol 27:1511-1520, 2007.
9. Kassis J, Hirsh J, Podor TJ: Evidence that postopera- 2006. 32. Shirk RA, Vlasuk GP: Inhibitors of factor VIIa/tissue
tive fibrinolytic shutdown is mediated by plasma 21. Eilertsen KE, Osterud B: Tissue factor: (patho)physi- factor. Arterioscler Thromb Vasc Biol 27:1895-1900,
factors that stimulate endothelial cell type I plas- ology and cellular biology. Blood Coagul Fibrinolysis 2007.
minogen activator inhibitor biosynthesis. Blood 15:521-538, 2004. 33. Wu KK, Matijevic-Aleksic N: Thrombomodulin: A
80:1758-1764, 1992. 22. Gailani D, Renne T: The intrinsic pathway of coagula- linker of coagulation and fibrinolysis and predictor
10. Hartwig JH: The platelet: form and function. Semin tion: A target for treating thromboembolic disease? J of risk of arterial thrombosis. Ann Med 32(Suppl
Hematol 43:S94-S100, 2006. Thromb Haemost 5:1106-1112, 2007. 1):73-77, 2000.

You might also like