Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

CHAPTER

40 COAGULATION AND FIBRINOLYSIS


Ravi Sarode, Craig M. Kessler

OVERVIEW OF HEMOSTASIS, 828 Secondary Hemostasis, 833 ACQUIRED COAGULATION


Laboratory Evaluation of ­DISORDERS, 843
PHYSIOLOGIC HEMOSTASIS, 828
Hemostasis, 833 Disseminated Intravascular
Endothelium and Platelets, 828
Approach To Patients With Coagulation (DIC), 843
Coagulation Protein System, 829
Coagulation Disorders, 836 Liver Disease, 844
The Formation of Fibrin Clot and
HEREDITARY COAGULATION Vitamin K Deficiency, 844
the Fibrinolytic System, 830
­PROTEIN DEFECTS, 836 Massive Transfusion, 845
The Anticoagulation Protein
Systems, 831 Deficiency of Factor VIII (Hemophilia A) Acquired Coagulation Factor
Or Factor IX (Hemophilia B), 837 Deficiencies, 845
Current Hypothesis For Initiation of
The Hemostatic System, 832 Hereditary Deficiencies of Other SELECTED REFERENCES, 846
Coagulation Factors, 840
CLINICAL HEMOSTASIS, 833
Primary Hemostasis, 833

KEY POINTS PHYSIOLOGIC HEMOSTASIS


The physiologic hemostatic system, a tightly regulated balance between
•  hysiologic hemostasis consists of endothelium, platelets, plasma co-
P
formation and dissolution of hemostatic plugs modulated by a series of
agulation proteins, natural anticoagulants, and the fibrinolytic system.
enzymes and scaffolding proteins, has two major parts. The first part is
•  rimary hemostasis involves platelets and von Willebrand factor.
P its cellular component, which consists mostly of platelets and endothe-
Secondary hemostasis is initiated by the tissue factor pathway and lial cells but also includes neutrophils and monocytes. The second part
then amplified and propagated by the intrinsic pathway to generate is a large group of plasma proteins, which participate in clot formation
thrombin that converts fibrinogen to fibrin. (coagulation), dissolution of clots (fibrinolysis), and the action of natu-
•  outine coagulation tests (prothrombin time and activated partial
R rally occurring serine protease inhibitors (anticoagulation) that termi-
thromboplastin time) may help identify a hemostatic defect in a bleed- nate the activity of several enzymes of the coagulation and fibrinolytic
ing patient, but mild to moderate abnormalities of these tests do not systems.
predict a bleeding tendency in a nonbleeding patient.
•  cquired coagulopathy often reflects multiple coagulation defects
A ENDOTHELIUM AND PLATELETS
rather than specific protein defects.
Normal hemostasis involves interplay between the cellular components
and proteins involved in clot formation and lysis. Intact endothelium
that lines the vascular wall in a physiologic state exerts a hemostatic bal-
ance. When vascular integrity is breached, blood coagulation is initiated.
Intact endothelial cells secrete an ectonucleotidase, CD39, that degrades
OVERVIEW OF HEMOSTASIS adenosine diphosphate (ADP) (Pinsky et al., 2002). Endothelial cells also
Hemostasis is a physiologic response to a vascular injury to limit blood secrete prostacyclin and nitric oxide (NO), which prevent platelet activa-
loss. Therefore, it is initiated rapidly, is localized, and is well regulated. tion and aggregation, thereby reducing the risk of arterial thrombosis
Recently, there has been an evolution in our understanding of the physi- (Hong, 1980; Palmer et al., 1987; Shariat-­Madar et al., 2006). Endo-
ologic hemostatic system (Fig. 40.1). Initially, the hemostatic system was thelial cells additionally bind plasminogen, tissue plasminogen activa-
referred to as the coagulation cascade based on the waterfall model of Ratnoff tor, and single-­chain urokinase, all of which contribute to fibrinolysis
and Davies; MacFarland published simultaneously a sequence of proteo- and maintenance of hemostatic balance (Barnathan et al., 1988; Cesar-
lytic reactions beginning with factor XII (FXII; Hageman factor) activation man et al., 1994). The glycosaminoglycans present on endothelium bind
and leading to thrombin formation that proteolyzes fibrinogen to form antithrombin (AT), which then neutralizes thrombin and FXa (Marcum
a clot (MacFarland, 1964; Ratnoff & Davies, 1964). However, the physi- et al., 1984). Thrombomodulin on endothelium binds to thrombin; that
ologic basis of this pathway was questioned because patients with FXII complex then converts protein C to activated protein C (APC) (Esmon
deficiency did not bleed. Furthermore, in the mid-­1970s, the cofactors for & Owen, 1981).
FXII activation—prekallikrein and high-­ molecular-­weight kininogen— When a vessel wall is injured, subendothelial collagen is exposed and plate-
were identified and their deficiencies also did not cause bleeding (Weup- lets adhere to the site of injury (Van der Meijden et al., 2009). Von Willebrand
pers & Cochrane, 1972; Colman et al., 1975; Saito et al., 1975). Later, the factor helps platelets adhere to the injured vessel wall by binding to exposed
physiologic role of tissue factor (TF) and FVIIa was recognized along with collagen at the A3 domain and to the platelet receptor GP Ib/IX/V complex
its regulator tissue factor pathway inhibitor (TFPI) (Osterud & Rappaport, (He et al., 2003). This adhesion event activates platelets, initiating a signaling
1977). Thus, the physiologic hemostatic system is a cell-­based model in cascade within them (Zaffran et al., 2000). The stimulated platelets release the
which cellular elements such as endothelium, platelets, neutrophils, mono- contents of their granules, recruit additional platelets to aggregate to those
cytes, and macrophages express TF and provide phospholipid (PL) surface already adherent and provide a phospholipid surface for the activation of mul-
for various coagulation proteins for attachment and sequential activation tiple coagulation factors. Furthermore, in flowing blood, FXII autoactivates if
of coagulation factors to generate thrombin at the site of injury. Throm- exposed to collagen or in the milieu of activated platelets, leading to the cas-
bin converts fibrinogen to a fibrin clot. Natural anticoagulants regulate cade of proteolytic events producing thrombin (Van der Meijden et al., 2009).
thrombin generation, and the fibrinolytic system dissolves the clot once it Once prothrombin has been activated to thrombin in the intravas-
has served its purpose. cular compartment, the resulting thrombin stimulates endothelium to

828
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
TABLE 40.1
XII VII
Tissue factor Proteins of the Plasma Coagulation System
Surface
HK Surface-­Bound Vitamin K– Cofactors/
PK XI Zymogens Dependent Zymogens Substrates
XIIa VIIa
Factor XII Factor VII High-­molecular-­

PART 5
weight kininogen
Tissue factor/VIIa
Prekallikrein Factor IX Factor VIII
IX
XIa Factor XI Factor X Factor V
PL, Ca++ Factor II Fibrinogen
Ca++ Protein C Protein S

IXa X

PL, Ca++ important source of negatively charged phospholipids (mainly, phospha-


Ca++
VIII VIIIa tidylserine) and procoagulant membranes (Furie & Furie, 2005) needed
for coagulation factor action in the propagation phase. This pathway for
Xa
thrombin formation conjoins with FXII autoactivation on exposed ves-
sel collagen and downstream elaboration of thrombin as well (Van der
Prothrombin
Meijden et al., 2009). Critical reactions to form FXa and thrombin are
PL, Ca++ accelerated 300,000-­fold on the platelet surface. Platelets control three
important phases of coagulation: thrombin generation, fibrin formation,
Va V and clot retraction (Heemskerk et al., 2013).

THROMBIN COAGULATION PROTEIN SYSTEM


Fibrinogen XIII Characterization of Coagulation Proteins
The proteins that constitute the coagulation system consist of zymo-
gens and cofactors (Table 40.1). The zymogens (proenzymes) of the
Soluble fibrin monomers Ca++
coagulation system can be grouped into the phospholipid-­bound and
the surface-­bound categories. Phospholipid-­bound zymogens make up
XIIIa the physiologically important hemostatic system. These proteins are vita-
Polymerized fibrin clot min K dependent. Vitamin K is required for an essential γ-­carboxylation
reaction that takes place on the glutamic acid residue of each of these pro-
teins located in their amino-­terminal regions (see Chapter 43, Fig. 43.1).
This carboxylation reaction allows these proteins to bind to phospholipid
Covalently crosslinked fibrin clot and cell membranes, where they are activated. Without this carboxyl-
ation reaction, these proteins do not function normally in the hemostatic
Figure 40.1 Schematic diagram of physiologic blood coagulation. Note that labels system. Proenzymes include FX, FIX (Christmas factor), FVII, and FII
for the “contact phase” factors constituting factor XII (FXII), high-­molecular-­weight (prothrombin). Protein C is a vitamin K–dependent phospholipid-­bound
kininogen (HK), and prekallikrein (PK) are in gray. Although FXI is activated by FXIIa
on a surface under artificial in vitro conditions, such as in activated partial thrombo- zymogen; however, when activated (APC), it functions as an antico-
plastin time (APTT), this pathway is not believed to contribute to normal physiologic agulant by inactivating FVa and FVIIIa (see Chapter 42). Protein S is
hemostasis. Similarly, whereas tissue factor (TF)/FVIIa can directly activate X to Xa in another natural anticoagulant that is also vitamin K dependent and acts
the in vitro prothrombin time (PT) test under conditions in which supraphysiologic as a cofactor for APC.
concentrations of TF are employed, this reaction is shown as grayed because it does The surface-­bound proenzymes are proteins of the plasma kallikrein/
not contribute significantly to clot formation under normal in vivo physiologic con-
ditions. Normal clotting in vivo is initiated when sufficient TF/FVIIa becomes avail-
kinin system (see Table 40.1). Surface-­bound proenzymes include FXII
able to activate FIX to FIXa. Subsequently, FIXa in the presence of FVIIIa activates FX (Hageman factor), prekallikrein (Fletcher factor), and FXI. These protein
to FXa, which, in turn, activates prothrombin to thrombin in the presence of FVa. zymogens are also known as the contact system because FXII autoactivates
Thrombin not only then proceeds to clot fibrinogen and to activate platelets but ad- when associated with a negatively charged surface, such as a glass tube and
ditionally exerts critically important positive feedback by activating both factor VIII several physiologic substances (Wiggins & Cochrane, 1979; Miller et al.,
and factor V. Physiologic amplification of thrombin formation is believed to result
from thrombin activating FXI to FXIa, thereby providing an additional pathway for
1980). The autoactivation phenomenon of FXII allows for a common labo-
the activation of FIX. Phospholipid (PL) protein complexes present on the surface ratory test, activated partial thromboplastin time (APTT), which is used to
membranes of platelets in vivo. Ca++, Calcium ions. assess the integrity of the coagulation system. Although absolutely essen-
tial for a normal APTT, the proteins of the plasma kallikrein/kinin system
do not have a role in hemostasis. These proteins (FXII, prekallikrein, and
upregulate TF, and TF forms a complex with FVIIa. The complex of high-­molecular-­weight kininogen) may participate in blood pressure regu-
FVIIa–TF activates FIX to FIXa, which converts zymogen FX to enzy- lation, fibrinolysis, inflammation, angiogenesis, and thrombosis (Schmaier,
matically active FXa (see Fig. 40.1). In the presence of FVa from liver 2002, 2003, 2004; Shariat-­Madar et al., 2006; Maas et al., 2008; LaRusch
or injured endothelium, FXa activates prothrombin (FII) to thrombin et al., 2010).
(FIIa). Under normal circumstances, the rate-­limiting component of pro- Although deficiencies of FXII, prekallikrein, and high-­ molecular-­
thrombinase complex formation and the ultimate generation of throm- weight kininogen are not associated with bleeding, investigations indicate
bin activity is the concentration of FXa (Rand et al., 1996). Thrombin that they may have a role in arterial and venous thrombosis. Recently, acti-
then proteolyzes fibrinogen to form fibrin. In static in vitro systems, as vation of the contact factor coagulation pathway has been shown to occur
little as 5 to 10 pM TF is sufficient to induce clot formation, leading to in vivo from negatively charged surfaces such as polyphosphate, extracellu-
a 1000-­to 2000-­fold amplification of the process, which increases the lar RNA, and collagen. In detail, polyphosphate (PolyP) in humans consists
concentration of thrombin to 10 to 20 nM, which is sufficient to initiate of 60 to 100 phosphate units, and it is released from platelet dense granules
clot formation (Mann, 2003). In a static model of blood coagulation, the upon platelet activation. PolyP has prohemostatic, prothrombotic, and
addition of 5 pM TF results in an average clot time of approximately 5 proinflammatory properties. It has been identified as an activator of the
minutes—sufficiently fast for physiologic hemostasis. Thrombin also is contact phase (Wang et al., 2019). It is a potent activator of thrombin and
a major physiologic activator of platelets together with other agonists, coagulation FXI, and it accelerates FV activation and abolishes the inhibi-
such as collagen, ADP, platelet-­activating factor, and epinephrine. Inacti- tory effect of TFPI.
vated platelets probably circulate without promoting coagulation. When TF (47-­kDa protein) is an essential cofactor for activated FVIIa. It is
platelets are activated (see Chapter 41), their membranes become a site found in most tissues and cells. Upregulation of TF results in the forma-
for additional thrombin formation. According to a cell-­based model of tion of complexes with FVII that produce the initiation of hemostatic reac-
coagulation (Hoffman & Monroe, 2001), activated platelets offer the most tions. FVIII (antihemophilic factor) is a 330-­kDa protein. When activated

829
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
E
CHAPTER 40 COAGULATION AND FIBRINOLYSIS

D D

A Thrombin
Fibrinopeptide A
E Alpha knob
D D
Fibrinopeptide B
B Beta knob
D E D
E

C D D

Cross-link
D D E D D E D D E
E E E

D D D D D D
D
Figure 40.2 Formation of a fibrin clot. A, Sche-
matic of fibrinogen. B, Thrombin proteolyzes fi-
brinopeptides A and B from fibrinogen to leave
soluble fibrin monomer. Soluble fibrin monomer
then associates side to side (C) and end to end
(not shown, for clarity) to form fibrin polymers.
D, Thrombin-­activated factor XIII (factor XIIIa)
covalently cross-­links the fibrin polymers into an
increasingly complex structure and an ultimate-
ly insoluble clot (E). Note that “E” corresponds
to the central domain of the original fibrinogen
molecule and “D” to the peripheral domains.
(Modified with permission from Doolittle RF: Fi-
brinogen and fibrin, Sci Am 245:126–135, 1981.) E

to FVIIIa, it is a cofactor for FIXa in the activation of FX. Its absence FVa on phospholipid membranes or cell membranes in an ordered struc-
is associated with the most severe clinically recognized bleeding disor- ture with FII (prothrombin) to accelerate its activation to FIIa (thrombin).
der, hemophilia A. FV is also a 330-­kDa protein with homology to FVIII. When all of these components are present, the rate of FII activation by
When activated to FVa, it serves as a cofactor for FXa in the activation of FXa is increased 1.7 × 108-­fold over the rate of FII activation by FXa alone.
FII (prothrombin) to thrombin. Fibrinogen is a 330-­kDa protein that not Because these coagulation reactions occupy critical regulatory points
only is the main substrate of thrombin (FIIa) but is also the principal adhe- within the physiologic hemostatic system, they are also the target of a num-
sive molecule for platelet aggregation. When fibrinogen is proteolyzed by ber of anticoagulant agents for venous thrombosis that are currently in use,
thrombin, a fibrin monomer is formed. These monomers associate end to that is, direct FXa and thrombin inhibitors.
end and side to side to form a polymerized fibrin clot. The clot is stabilized
by activated FXIII, a tissue transglutaminase that cross-­links the strands of
associating fibrin (Fig. 40.2). THE FORMATION OF FIBRIN CLOT AND THE
Physiologic Protein Assemblies FIBRINOLYTIC SYSTEM
Certain critical protein assemblies in hemostatic reactions accelerate The six peptide chains of the fibrinogen molecule are organized into
these proteolytic events. The proteins of the coagulation system that a structure described as having a central E domain and two terminal
are essential for hemostasis or control of bleeding were originally iden- D domains. When thrombin is formed, it cleaves fibrinopeptide A
tified by observation of affected patients and, more recently, through from the Aα chain and fibrinopeptide B from the Bβ chain of fibrino-
mouse knockout studies. Deficiencies in coagulation factors VIII and gen in the E domain region (Doolittle, 1981). The remainder of this
IX are the most prominent bleeding disorders that occur in patients thrombin-­proteolyzed fibrinogen is called a soluble fibrin monomer. Sol-
who survive gestation and birth. Rare patients who have congenital uble fibrin monomers then assemble with an end-­to-­end and side-­to-­
deficiencies of coagulation factors VII, X, V, and II usually do not have side association to form a noncovalent fibrin polymer (see Fig. 40.2).
severe bleeding states. In contrast, murine models have demonstrated Activated FXIII, a transglutaminase, cross-­ links fibrin monomeric
that mouse embryos containing complete genetic knockouts of factor subunits into an insoluble, cross-­linked fibrin clot. When insoluble,
VII, X, V, or II die of massive hemorrhage during gestation or at birth cross-­linked fibrin is made, a new linkage between the D domains of
(Cui et al., 1996; Rosen et al., 1997; Sun et al., 1998; Dewerchin et al., two adjacent fibrin monomers occurs. In the process, a neo-­epitope of
2000). This information suggests that the human patient who has a interaction is formed (see Fig. 40.2).
deficiency of factor VII, X, V, or II may have some factor, albeit less The fibrinolytic system regulates fibrin clot degradation and clot dis-
than 1%, to allow for a milder clinical phenotype than that seen in solution. In fibrinolysis, the circulating zymogen plasminogen is converted
mouse models. Alternatively, other compensatory mechanisms may be to its active form plasmin mainly by endogenous tissue plasminogen acti-
operative in the human patient. All of these proteins participate in two vator (tPA) (Draxler & Medcalf, 2015). Two other plasminogen activators
critically important assemblies that are essential for normal hemostasis: are single-­chain urokinase plasminogen activator (ScuPA) and two-­chain
tenase and prothrombinase complexes. urokinase plasminogen activator (TcuPA). These activators are found in
Extrinsic tenase involves TF and FVIIa complex on the PL surface, the endothelium as well as in neutrophils and monocytes. The natural
converting FX to FXa. The intrinsic tenase complex comprises the assem- plasminogen activators tPA, ScuPA, and TcuPA convert zymogen plas-
bly of FIXa and FVIIIa on phospholipid surfaces to convert FX to FXa. minogen to the active enzyme plasmin (Fig. 40.3). tPA is produced con-
When all of these components are present, the rate of FX activation by stitutively, and ScuPA is increased in inflammatory states. Plasminogen
FIXa is increased 1.4 × 108-­fold over the rate of FX activation by FIXa activator inhibitor-­1 (PAI-­1) is the major inhibitor of tPA and TcuPA. α2-­
alone. The prothrombinase complex comprises the assembly of FXa and Antiplasmin, a serine protease inhibitor (serpin), is the most potent and

830
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
highly selective inhibitor of plasmin. However, the plasma concentration molecular mass by eliminating portions of the α-­chain (Marder & Budzyn-
of α2-­antiplasmin, even if high (approximately 1 mM), is only about half ski, 1974). Plasmin then cleaves the X fragment asymmetrically between
the plasma concentration of plasminogen. The fibrinolytic system is also the D and E domains to produce fragment Y. Plasmin further degrades
regulated by thrombin-­activatable fibrinolysis inhibitor (TAFI) (Vercau- the Y fragment to produce soluble D and E domains that are called soluble
teren et al., 2013). TAFI is a carboxypeptidase able to remove lysine resi- fibrinogen degradation products or, in the case of fibrin digestion by plasmin,
dues from fibrin, such that it indirectly controls plasmin by interfering with fibrin degradation products. Their presence indicates that plasmin has been

PART 5
the binding of plasminogen and tPA to fibrin. formed. Plasmin will also degrade insoluble, crossed-­linked fibrin (Fig.
The binding of both plasminogen and tPA to fibrin can increase plas- 40.4B). When it does, it liberates a D-­D-­dimer domain formed as a result
min generation by two orders of magnitude. The active plasmin molecule of the neo-­epitope between these two domains (see Fig. 40.4B) (Marder
is a potent protease, and it recognizes multiple substrates. Plasmin will et al., 1976). The presence of soluble D-­dimer indicates that, first, throm-
degrade soluble fibrinogen to produce fibrinogen degradation products bin has been formed, followed by clotting, then the clot has been cross-­
(Fig. 40.4A). Plasmin cleaves fibrinogen into an X fragment of similar linked by FXIIIa, and, finally, plasmin has been formed and has cleaved the
insoluble, cross-­linked fibrin clot.

THE ANTICOAGULATION PROTEIN SYSTEMS


Plasminogen Coagulation is tightly regulated by natural anticoagulants and the fibri-
nolytic system to avoid thrombus formation and extension. Three major
tPA Fibrin(ogen) anticoagulant systems regulate the enzymes of the coagulation protein
ScuPA TcuPA Degradation Products (FDP) system to help to inhibit clot formation. These systems are the protein C/
protein S system, the plasma serine protease inhibitor system, and TFPI,
a Kunitz-­type serine protease inhibitor. AT is the main serine protease
Plasmin inhibitor of coagulation enzymes of the plasma serine protease inhibitor
Fibrin, fibrinogen system. Each of these systems plays a critical role in the proper regulation
of the coagulation system. Moreover, in murine deletion models, com-
Figure 40.3 Fibrinolysis. Zymogen plasminogen is converted to plasmin by tissue
plete deficiencies of protein C, protein S, AT, and TFPI are incompat-
plasminogen activator (tPA), single-­chain urokinase plasminogen activator (ScuPA), ible with the survival of mammalian gestation, delivery, or life ex utero
and two-­chain urokinase plasminogen activator (TcuPA). Formed plasmin degrades (Huang et al., 1997; Jalbert et al., 1998; Hayashi et al., 2006; Burstyn-­
fibrinogen or fibrin to form fibrinogen or fibrin degradation products, respectively. Cohen et al., 2009).

αC αC
Plasmin
E
D D Fibrinogen

Plasmin
E
D D Fragment X

Plasmin
E
D D Fragment Y

E
D D Fragments
A
D and E

D D D D
E E E
E
D D D Figure 40.4 A, Plasmin-­ cleaved soluble fibrinogen or fibrin.
D When plasmin cleaves fibrinogen, initially small portions from
the α-­chain (αC) are removed to make Fragment X. Fragment X
is then asymmetrically cleaved into Fragment D and Fragment Y.
Fragment Y is further cleaved by plasmin into Fragments D and E.
(Modified with permission from Greenberg CS, Lai T-­S: Fibrin for-
mation and stabilization. In Loscalzo J, Schafer AI, editors: Throm-
bosis and hemorrhage, ed 3, Philadelphia, 2003, Lippincott Wil-
liams & Wilkins, p 83, Fig. 5.3B.) B, Plasmin-­cleaved insoluble,
D D cross-­linked fibrin. When insoluble, cross-­linked fibrin is prote-
olyzed by plasmin, the neo-­epitope between the D-­domains is
E preserved, and the liberated fragment consists of the D-­dimer to-
gether with an E domain. (Modified with permission from Doolit-
B tle RF: Fibrinogen and fibrin, Sci Am 245:126–135, 1981.)

831
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
Protein C/Protein S System
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
When activated, protein C, a 62-­kDa vitamin K–dependent protein, is
an enzyme that functions as an inhibitor. When free thrombin binds to VII
thrombomodulin on endothelium, this complex then converts protein C
to APC. Its activation is increased when protein C binds to the endothe- XI XIa Tissue factor
lial protein C receptor (EPCR). The complex formed by APC+EPCR
needs to dissociate in order to allow binding of APC with protein S to VIIa
inactivate FVa and FVIIIa (Esmon, 2006). Protein S, a 69-­kDa vitamin IX
K–dependent protein, is a cofactor, or receptor, for APC on cell mem- VIIIi Tissue factor/VIIa
branes. It allows APC to bind to cell surfaces in such a manner as to PL, Ca++
Ca++ Tissue
orient itself to inactivate FVa and FVIIIa. The enzyme uses this cofactor
IXa X factor
as a receptor to localize its activity to perform its inhibitory function.
pathway
Plasma protein S is in equilibrium between the free form (40%) and PL, Ca++ inhibitor
a bound form (60%) that is complexed to C4b-­binding protein; only (TFPI)
the free form functions as a cofactor for APC. APC also binds to three VIII VIIIa
Xa
receptors on endothelial cells: (1) endothelial cell protein C receptor Vi
to activate (2) protease-­activated receptor (PAR) 1 and (3) apolipo- Prothrombin
protein E receptor 2 (ApoER2) (Esmon, 1999; Coughlin, 2000; Yang Antithrombin PL, Ca++
et al., 2009). Activation of PAR1 on endothelial cells may contribute
to the anticoagulant function of activated protein C by liberating tPA. Va V
APC on endothelial cells also has an inflammatory effect via activation
of sphingosine-­1-­phosphate receptor transactivation and activation of Thrombin
ApoER2 (Finigan et al., 2005; Yang et al., 2009). Thus, APC reduces
thrombin formation, stimulates fibrinolysis, and initiates inflammation
to reduce thrombosis risk. Thrombin Thrombomodulin Cell surface

Antithrombin Protein C
This serpin is a 58-­kDa protein that inhibits each of the following hemo- Protein C
static enzymes: IIa, Xa, VIIa, IXa, XIa, kallikrein, and XIIa. It exerts its
anticoagulant effect primarily by inhibiting FIIa and FXa (Fig. 40.5). Anti-
thrombin has two functional sites: the heparin-­binding site (at the amino Activated
terminus of the protein) and the reactive site (Arg329-­Ser394). The ability protein C
of antithrombin to function as an inhibitor of coagulation protein enzymes
is potentiated by endogenous and exogenous heparins. In fact, it is the Protein S
presence of antithrombin that gives heparin its anticoagulant properties.
In the presence of heparins, antithrombin undergoes a conformational
change that increases from 1000-­fold to 4000-­fold its inhibitor effect on
Protein S
FIIa (Perry, 1994). Trace amounts of circulating TF or TF transiently
exposed by damaged vessels contribute to FVIIa inactivation by antithrom-
bin (Vatsyayan et al., 2014). C4b-binding protein
In addition to antithrombin, other serpins regulate the enzymes
of the hemostatic and inflammatory systems. Heparin cofactor II is a Figure 40.5 Natural inhibitors of coagulation: antithrombin (AT); components of
the protein C pathway (thrombomodulin, protein C, protein S); and tissue factor
serpin that specifically inhibits thrombin in the presence of dermatan pathway inhibitor (TFPI). Three major anticoagulant systems are recognized. An-
sulfate. Protein Z inhibitor is a serpin that specifically inhibits factor tithrombin inhibits factor Xa and thrombin to prevent thrombosis. It is important to
Xa in the presence of its cofactor protein Z—a vitamin K–dependent note that antithrombin inhibits every enzymatic form of blood coagulation serine
protein. C1 inhibitor (C1 esterase inhibitor) is the most potent inhibitor proteases (factor XIIa, plasma kallikrein, factor XIa, factor IXa, and factor VIIa), even
of FXIIa, kallikrein, and FXIa in plasma. Its main function is to regu- though this is not indicated in the figure for readability purposes. The protein C
system requires that this zymogen is activated to activated protein C by thrombin
late the amount of free bradykinin in the intravascular compartment and
when bound to the endothelial cell membrane protein thrombomodulin. Protein S
reduce inflammatory events (Han et al., 2002; Schmaier, 2002; Maas is a cofactor for activated protein C to inactivate factors Va and VIIIa. C4b-­binding
et al., 2008). The absence of C1 inhibitor is the causative factor for types protein regulates protein S activity. TFPI makes a quaternary complex with tissue
I and II hereditary angioedema, a disorder associated with excessive bra- factor/factor VIIa and factor Xa to inhibit both enzymes.
dykinin delivered to tissues.

Tissue Factor Pathway Inhibitor the cyclooxygenase-­2 enzyme that produces prostacyclins (PGI2) from
In addition to the serpins, there is the Kunitz-­type serine protease inhibi- arachidonic acid (Mitchell et al., 2008). PGI2 is a vasodilator and inhib-
tor, TFPI (Crawley & Lane, 2008). TFPI is the most potent inhibitor of the its platelet aggregation. The actions of prostacyclins are mediated by
FVIIa–tissue factor complex. Under physiologic conditions, TFPI exerts cell membrane prostacyclin (IP) receptors and intracellular peroxisome
its inhibitory effects by forming a quaternary complex with FVIIa, TF, proliferator-­activated receptors (PPARβ and PPARγ). Endothelial cells
and FXa (see Fig. 40.5). It is noteworthy that the murine TFPI knockout also express the NO synthase enzyme (eNOS, NOS3), and they release
is embryonically lethal (Jalbert et al., 1998). TFPI is produced by micro- NO derived from l-­arginine. NO is a potent vasodilator that also inhib-
vascular endothelium, and it seems mainly associated with glycosaminogly- its platelet adhesion and aggregation. NO activates guanylate cyclase and
cans on the endothelial wall. Only approximately 20% of TFPI circulates cGMP. Activated platelets adhering to collagen seem also able to produce
in plasma associated with lipoproteins (Caplice et al., 2001). Another fam- NO under shear flow (Cozzi et al., 2015), thus, controlling platelet adhe-
ily of Kunitz-­type serine protease inhibitors, the amyloid β-­protein pre- sion and vasoconstriction directly at the sites of vessel damage.
cursor (AβPP) and related members, is present in platelets and brain and Thrombospondin-­ 5, also known as cartilage oligomeric matrix protein
regulates factors XIa, IXa, Xa, VIIa/TF, and plasmin (Van Nostrand et al., (COMP), is an extracellular protein able to control vascular tone. COMP is
1990; Schmaier et al., 1993, 1995; Mahdi et al., 1995, 2000). This inhibitor released by a variety of cartilage and muscle tissues and activated platelets.
does not inhibit thrombin. The exact function of this inhibitor is not com- COMP inhibits thrombin and acts as a natural anticoagulant in mice (Liang
pletely known, but it is believed to be a cerebral anticoagulant (Del Zoppo, et al., 2015). It determines a dose-­dependent prolongation of thrombin time.
2009). AβPP and amyloid precursor-­like/protein 2 gene–deleted mice sur-
vive gestation but have an accelerated rate of thrombosis in a carotid artery
thrombosis model (Xu et al., 2009). CURRENT HYPOTHESIS FOR INITIATION OF THE
Prostacyclins, Nitric Oxide, and Thrombospondin-­5 HEMOSTATIC SYSTEM
In physiologic conditions, endothelial cells produce vasoactive hormones More proteins participate in coagulation reactions in vitro than are critical
able to control the primary phase of hemostasis. Endothelial cells express for hemostatic reactions in vivo. The original coagulation cascade waterfall

832
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
hypothesis initiated by FXII has been replaced by one whereby TF and forms the prothrombinase complex converting prothrombin to throm-
FVIIa are initiators (Gailani & Broze, 1993). TF is ubiquitous throughout bin. This intrinsic pathway is the primary pathway of thrombin forma-
the body, although it is rich in the brain, lung, and placenta. Its expression tion. Thus, the TF pathway is the initiator and the intrinsic pathway is
is upregulated after injury. Regulation of the expression of TF is a major the amplifier and propagator of thrombin, and these two pathways are
control mechanism for the initiation of hemostasis. TF is not fully active interrelated. The previous concept of independent extrinsic and intrin-
on intact cells; it becomes able to activate coagulation when cell membrane sic pathways is not physiologic because if these two were independent

PART 5
properties are modified. This process is called decryption of encrypted TF and pathways producing thrombin, then patients with hemophilia A and B
is still not completely understood. Different mechanisms of TF decryption would not have bleeding tendencies. The independence of extrinsic and
have been proposed: an exposition of phosphatidylserine, dimerization of intrinsic pathways to produce thrombin is “true only in the test tubes”
TF, and oxidation of a specific disulfide bond in TF (Smith et al., 2015). where prothrombin time (PT) and activated partial thromboplastin time
In conditions in which a vessel wall injury is limited to endothelial activa- (APTT), respectively, assess them. Thrombin is the main enzyme in the
tion, blood-­borne TF has been characterized on microparticles released coagulation system because it activates FV and FVIII to their activated
by leukocytes and involved in the initiation of coagulation (Furie & Furie, forms to amplify further thrombin generation and converts fibrinogen
2005). Activated macrophages have been shown to express TF on their to fibrin monomers. It also activates FXIII to FXIIIa to cross-­link these
cell membrane and microparticles involved in physiologic hemostasis and fibrin monomers to stabilize the clot.
thrombosis (Rak, 2014). There is a developing notion that endothelial cell
TF is the initial nidus that leads to hemostasis and thrombosis (Furie, 2009;
Ivanciu et al., 2014).
LABORATORY EVALUATION OF HEMOSTASIS
The FVIIa–TF complex activates FIX, leading, in turn, to FX acti- Laboratory evaluation of primary hemostatic defects presenting as
vation. It must be noted that at the concentrations of factors ordinar- mucocutaneous type bleeding due to platelets and vWF is described in
ily present in the body, FVIIa/TF does not directly activate appreciable Chapter 41. Patients with bleeding tendency due to secondary hemosta-
amounts of FX because of the presence of TFPI—a point emphasized sis defects often present with ecchymoses, soft tissue, and joint bleeds.
diagrammatically in Figure 40.1 by the graying of this reaction. Once FX Bleeding tendencies could be congenital or acquired. Therefore, an
has been activated to FXa, this FXa, in turn, contributes to the activation elaborate personal and family history of bleeding is the prerequisite for
of thrombin from prothrombin. Thrombin then proteolyzes fibrinogen appropriate laboratory testing. Most congenital bleeding disorders often
to form fibrin. Although TF-­dependent coagulation reactions are rapidly are diagnosed early in childhood; however, mild bleeding disorders may
inhibited by TFPI, if the stimulus for thrombin formation is sufficiently not be revealed until later in life when there is a robust hemostatic
strong, coagulation is maintained through the activation of FXI by throm- challenge.
bin (Meijers et al., 2000). Activation of FXI to FXIa results in increased Routine coagulation screening tests are useful to identify a hemo-
activation of FIX and eventually increases the formation of thrombin. static defect in a bleeding phenotype. However, performing these tests
Further modulation of hemostatic balance in the direction of clot forma- to assess hemostasis in a nonbleeding patient is not clinically useful—it
tion is exerted by TAFI (also known as carboxypeptidase U). Among its often may lead to unnecessary expensive further testing and could delay
actions, TAFI cleaves C-­terminal lysine residues from fibrin, thereby surgeries.
decreasing the binding of plasminogen to the clot and diminishing clot Prothrombin time (PT), APTT, fibrinogen, thrombin time and
lysis (Leurs et al., 2005). D-­dimers are considered screening coagulation tests along with platelet
count for initial evaluation of a bleeding patient. Whole blood is col-
lected in 3.2% sodium citrate (blue top) in a 9:1 ratio (blood:citrate) by
CLINICAL HEMOSTASIS a single needlestick without trauma or prolonged stasis and mixed well
by gently inverting 3 to 5 times (avoid vigorous mixing that may activate
PRIMARY HEMOSTASIS platelets) (Adcock, 1998). The specimen should be transported to the
At the site of endothelial cell injury, subendothelial collagen and von Wil- laboratory at room temperature, and the testing should be completed
lebrand factor (vWF) are exposed. The disrupted endothelium alters lami- within 4 hours of collection. However, if the testing cannot be per-
nar blood flow at the site of injury; thereby, platelets undergo shape change formed within 4 hours of collection, then ideally the specimen should
from discoid to spherical and produce pseudopodia that will breach the be centrifuged immediately to separate plasma from cellular compo-
site of endothelial damage by binding to vWF via GPIb/IX/V receptors. nent and frozen at −20°C and then transported on dry ice for testing
This step is called platelet adhesion (attachment of platelets to a nonplatelet to the laboratory. The separated plasma ideally should be platelet free
surface), which is followed by secretion. During secretion, contents of alpha (<10 K) before freezing because frozen plasma that is rich in platelets
and dense granules from platelets are released along with the generation upon thawing will cause disruption of platelets releasing platelet factor
of thromboxane A2 (TXA2) via the cyclooxygenase-­1 (COX-­1) pathway. 4, which binds to heparin and may give false low anti-­FXa activity, or
TXA2 causes vasoconstriction and activates platelets via the TXA receptor. phospholipids released from platelet membranes bind to lupus antico-
During the next step of platelet activation, the signal transduction within agulant and may give false-­negative results. The specimen should not
platelets leads to a conformational change in GPIIb/IIIa, allowing fibrino- be held at room temperature for more than 4 hours to avoid loss of
gen (acts as a ligand) to bring adjacent platelets together, causing aggrega- FVIII (and, to some extent FV, both being heat-­labile factors) to avoid
tion (attachment of one platelet to another). Activation of platelets leads getting spuriously long APTT and low FVIII (and vWF) that might
to exposure of phosphatidylserine (PS) similar to altered endothelium that result in a further unnecessary investigation. Improper sample handling
also exposes PS for secondary hemostasis that begins simultaneously to is often encountered in office clinics that send out their specimens to
generate thrombin. remote reference laboratories for coagulation testing. Thus, the pre-
analytic part of coagulation testing is crucial for accurate diagnosis and
management.
SECONDARY HEMOSTASIS
Secondary hemostasis can be divided into two different phases: (1) the Prothrombin Time
TF pathway (previously called the extrinsic pathway), which is the ini- PT is a simple test in which the patient plasma is mixed with a PT reagent
tiator of coagulation; and (2) the intrinsic pathway, which amplifies and that contains tissue thromboplastin and CaCl2. Tissue thromboplastin
propagates thrombin generation. The TF pathway begins with endo- consists of tissue factor and phospholipid. The source of tissue throm-
thelial injury and expression of TF on the cell surface, which binds the boplastin is important because nonhuman sources (e.g., rabbit brains)
small amount of FVIIa present in circulation to form a TF+FVIIa com- may have lower sensitivity to various vitamin K–dependent coagulation
plex. This complex is known as extrinsic tenase because this will activate factors as compared with human sources (e.g., placenta) or recombinant
FX to FXa on the cell surface in the presence of Ca++. This FXa along tissue thromboplastin. The international normalized ratio (INR) was
with FVa on the cell surface (PL) and Ca++ forms a prothrombinase com- developed to harmonize PT results for monitoring vitamin K antagonist
plex, converting prothrombin to thrombin. As soon as a small amount of (VKA) therapy. The INR is a calculated value obtained by first calcu-
thrombin is generated by the TF pathway, this thrombin activates the lating the prothrombin time ratio (PTR) by dividing the patient PT by
TFPI, which shuts down the TF pathway by inhibiting the TF+FVIIa the control PT, and then raising this PTR by an international sensitiv-
and FXa complex. The same thrombin also initiates the intrinsic path- ity index (ISI) [INR = (PTR) ISI]. Ideally, the INR should be used only
way by activating FXI to FXIa and FIX to FIXa on the cell surface (PL) for monitoring VKA therapy. However, due to the inclusion of the INR
with Ca++. FIXa along with FVIIIa on the cell surface (PL) and Ca++ in the model of end-­stage liver disease (MELD) score calculation, it is
forms the intrinsic tenase and converts FX to FXa, which, along with FVa, often also used to assess hemostasis in patients not on VKA for no good

833
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
reason other than the convenience. When the INR was used for MELD A very high amount of the direct thrombin inhibitor will give false low
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
score calculation, the PT reagents most likely had a higher ISI with less fibrinogen levels.
sensitivity for vitamin K–dependent factor deficiencies than most current If there is clinical suspicion for hypofibrinogenemia, the direct fibrino-
PT reagents, especially recombinant tissue thromboplastin. With differ- gen assay should be performed rather than relying on PT and APTT to
ent ISI, the INR can be different; hence, the MELD score can also be be prolonged because, in the majority of patients with mild to moderate
significantly different. hypofibrinogenemia, PT and APTT are likely to be normal. When PT and
Isolated prolonged PT is usually due to an FVII deficiency and may also APTT were performed manually in the past, the endpoint was a solid, firm
be prolonged due to mild deficiencies of common pathway FII, FV, and FX clot formation on the side of the test tube. However, with automation, the
depending on the reagent’s sensitivity. Recombinant tissue thromboplastin endpoint of PT/APTT is the change in optical density or turbidity due to
is very sensitive to even mild reduction of FVII (40%–45% activity), which formation of fibrin strands. Therefore, the endpoint is often sooner than
is not clinically relevant since most patients with mild to moderate FVII the manual method; hence, automated PT and APTT are usually normal,
deficiency are clinically asymptomatic and are often diagnosed when there with mild to moderate hypofibrinogenemia.
is an incidental prolonged PT. Hemostatic levels of FVII for major sur-
gery are 10% to 20%; however, even mild prolongation of PT/INR often D-­Dimers
results in unnecessary plasma transfusion to correct a laboratory value that D-­Dimers are generated by plasmin that cleaves the cross-­linked fibrin
is not clinically relevant. Because of rebalanced hemostasis in patients with clot. Increased D-­dimers suggest that a clot is undergoing lysis in the cir-
cirrhosis (discussed later), several societies have recommended ignoring culation. In the D-­dimer assay, a monoclonal antibody against the D-­dimer
PT/INR values obtained preprocedurally in cirrhotics. molecule is attached to latex particles that agglutinate in the presence of
D-­dimers, causing a change in optical density. The optical density is then
Activated Partial Thromboplastin Time read off a standard curve to obtain a D-­dimer value. In the appropriate
APTT is another routinely performed test to assess clinical hemosta- clinical setting, it is useful for a diagnosis of disseminated intravascular
sis with poor sensitivity and specificity in a nonbleeding patient. APTT coagulation (DIC), where it is elevated, and to rule out deep vein throm-
generally assesses coagulation factors in the intrinsic pathway as well as bosis (DVT) and pulmonary embolism (PE) in patients with low clinical
the common pathway. In APTT, a contact activator (e.g., ellagic acid, probability, where the D-­dimer is not elevated. A large hematoma will also
silica, kaolin) is added to the patient’s plasma to initiate the activation increase D-­dimers while undergoing resolution.
of contact factors (prekallikrein, HMWK, and FXII) in the presence of
PLs. After incubation for 3 to 5 minutes, the plasma is recalcified and the APTT Mixing Study
clotting time is recorded (usual range, 23–35 sec). Unfortunately, APTT In the past, when there were no specific tests to detect lupus anticoagulant
reagents are not well standardized; hence, there is significant variation (such as dilute Russell viper venom time, PTT-­LA with hexagonal PL neu-
from one reagent to another and when we include coagulation instru- tralization, and silica clotting time), a mixing study involving equal parts of
ments from different companies, the variability in APTT is even more patient plasma and normal pooled plasma (NPP) has been often helpful. A
significant. The APTT reagents come with three different quantities of circulating inhibitor was suspected when there was incomplete correction,
PL with different sensitivities for various needs. APTT-­factor sensitive and a factor deficiency was suspected when there was complete correction of
(APTT-­FS) has the highest amount of PL, making this reagent very sen- the prolonged APTT. However, it must be emphasized that the routine use
sitive to detect even slightly lower levels of coagulation factors in the of such a mixing study for mildly prolonged APTT can lead to a missed or
intrinsic pathway. APTT-­LA (lupus anticoagulant sensitive) reagent has wrong diagnosis (Yates et al., 2019). This is due to several reasons, includ-
the smallest amount of PL, making it very sensitive to detect even a weak ing poorly standardized mixing study protocols. Often, these studies are
LA. APTT-­FSL (factor and lupus sensitive) has an intermediate amount performed without proper controls and there is often a lack of understand-
of PL; hence, it should be used in daily practice because this will detect ing of how many normal plasmas should be used to prepare NPP. Also,
clinically relevant decreased amounts of coagulation factors and lupus performing a mixing study only on immediate mixing and without 2-­hour
anticoagulant. incubation at 37°C leads to missing an autoantibody against FVIII, the
Isolated prolonged APTT is a common clinical finding. Further inves- cause of acquired hemophilia. Investigation of isolated prolonged APTT
tigation should be dictated by a personal and family history of bleeding should be based on a thorough clinical history of bleeding or no bleeding/
if that is the presenting symptom, whereas in a nonbleeding patient it is clotting. Mildly prolonged APTT due to lupus anticoagulant can be cor-
essential to check previous APTTs. If the previous APTTs were normal, rected with NPP, resulting in misinterpretation of a factor deficiency and,
then the current prolongation is due to an acquired defect, most likely hence, unnecessary measuring intrinsic pathway factors. The presence of a
lupus anticoagulant. However, if the previous APTTs are also prolonged, lupus anticoagulant may influence clot-­based assays for intrinsic pathway
then the contact factor deficiency should be considered and investigated factor levels, especially FVIII. However, if one looks at factor assay curves,
because FXII-­, HMWK-­, and PK-­deficient patients do not bleed. especially FVIII assay, there is an inhibitory pattern observed indicating
In a bleeding patient, if APTT is prolonged, unfractionated heparin that there is interference by the lupus anticoagulant. The lupus anticoagu-
should be excluded. If the previous APTTs were also prolonged, then a lant effect is revealed by the observation that with each subsequent dilution
congenital bleeding disorder of intrinsic pathway should be considered of the patient’s plasma during FVIII assay, the FVIII activity increases by
and investigated by measuring FVIII first (common thing first!) followed >20%. Such patients generally do not have a bleeding tendency and have
by FIX or, rarely, FXI. If the previous APTTs were normal and current had normal APTTs in the past.
presentation includes soft tissue hematomas, easy bruising, and other con- Acquired hemophilia is not an uncommon clinical condition and gener-
ditions, with a prolonged APTT, then acquired hemophilia should be con- ally presents with a prolonged APTT with significant clinical bleeding in
sidered due to autoantibody against FVIII. the soft tissues. Such a patient with normal APTTs in the past and in the
right clinical setting should be tested first for FVIII level, which is often
Thrombin Time (TT) diagnostic of acquired hemophilia. Later, a proper two-­step mixing study
Thrombin time (TT) is usually not a commonly ordered test because of its can be done to identify a time and temperature-­dependent inhibitor. In a
limited utility in laboratory assessment of most bleeding disorders. In the two-­step mixing study, the patient’s plasma is first mixed with the NPP
TT test, bovine thrombin (3–5 U/mL) is added to the patient’s plasma and and immediately tested for APTT (which often corrects, unless the patient
the clotting time recorded. The upper limit of the normal range should be has concomitant LA). The second tube with the patient’s plasma and NPP
adjusted close to 25 sec to make TT more sensitive to detect dysfibrino- is incubated at 37°C for 2 hours and then tested for APTT, which shows
genemia. The most frequent cause of prolonged TT is heparin followed by further prolongation as compared with the immediate mix.
hypofibrinogenemia and then dysfibrinogenemia. Direct thrombin inhibi- A routine mixing study for PT is not warranted because isolated pro-
tors (oral and parenteral) significantly prolong TT. longed PT due to autoantibody against FVII is extremely rare. Most of
these patients with isolated prolonged PT/INR generally have vitamin K
Fibrinogen Assay deficiency. Therefore, these patients should be given intravenous vitamin
Fibrinogen assay should be measured routinely in a bleeding patient K (10 mg), which is both diagnostic and therapeutic, confirming vitamin
and patients with cirrhosis. The fibrinogen assay is based on TT (Clauss K deficiency, especially in sick patients due to poor diet and antibiotic
method) in which a standard curve is generated by using known calibrators use.
and bovine thrombin at a very high concentration of 50 U/mL (for TT 3–5
U/mL) so that the clotting time is independent of thrombin concentration. Heparin Monitoring
Bovine thrombin is added to the patient’s plasma and the clotting time Therapeutic heparin monitoring is required to avoid over-­and under-­
obtained is used to extrapolate fibrinogen value from the standard curve. anticoagulation because of unpredictable pharmacokinetics (PK) and

834
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
pharmacodynamics (PD) of unfractionated heparin (UFH). The unpre- Surface Tissue
dictable PK and PD are due to the presence of many larger polysaccharide Activation Thromboplastin
molecules that get attached to the endothelium, platelets, and acute-­phase
reactant proteins, such as vWF and fibrinogen, causing reduced availability
for their action on antithrombin to act as an anticoagulant in the first 24 to Intrinsic Extrinsic
72 hours of therapy in patients with DVT/PE. Therefore, UFH is given XII, PK

PART 5
as a bolus to saturate these cells and proteins. The continuous intravenous
infusion is then monitored every 6 hours until the APTT is within the thera- HK
XI VII
peutic range. The APTT therapeutic range has to be established for each
new lot number of APTT reagent in each laboratory, that is, every 6 to 12 IX
months. This therapeutic range is established by measuring APTT and anti- VIII
­Xa activity simultaneously on 50 patients on stable UFH (but not on VKA)
and extrapolating corresponding APTT for anti-­Xa activity of 0.3 to 0.7 U/
mL from a linear regression curve. However, this therapeutic range is not Common
adjusted for different lots of UFH obtained by the hospital pharmacy!
Heparin resistance is often encountered when APTT is used to monitor X
UFH therapy in patients with very high levels of FVIII due to an acute-­phase Partial Prothrombin
effect or those with low AT levels (often acquired and rarely due to con- V
genital deficiency). The very high levels of FVIII can be suspected when the Thromboplastin Time
baseline APTT is either at the lower end of the normal range or even shorter Time II
than the lower limit of normal. Most clinicians often do not pay attention Fibrinogen
to this phenomenon, which results in patients getting higher amounts of
UFH. This increases anti-­Xa activity, which may lead to severe bleeding
despite APTT showing resistance. In the case of heparin resistance due to
high FVIII, an anti-­Xa assay should be used to monitor UFH. However, if Thrombin Time
anti-­Xa activity is also low despite higher UFH, then AT deficiency should
be suspected. If the AT activity is <50%, then AT concentrate may be given;
that will prolong APTT appropriately as well as improve ant-­Xa activity.

Anti-­Xa Activity Assay for Heparin Monitoring Fibrin


This assay provides an accurate measurement of both UFH and low-­ Figure 40.6 Organization of the coagulation system based on current assays. The
molecular-­weight heparin (LMWH) levels for therapeutic monitoring. intrinsic coagulation system consists of the protein factors XII, XI, IX, and VIII and
prekallikrein (PK) and high-­molecular-­weight kininogen (HK). The extrinsic coagu-
There are two types of anti-­Xa assays, one with and the other without lation system consists of tissue factor (tissue thromboplastin) and factor VII. The
exogenously added AT to the reagent. The assay without added AT common pathway of the coagulation system consists of factors X, V, and II, and
that relies on AT in the patient’s plasma is a better assay because it will fibrinogen (I).
detect heparin resistance due to AT deficiency by showing lower anti-
­Xa activity despite adequate UFH dose. UFH/LMWH present in the
patient’s plasma binds to the patient’s own AT and neutralizes the known lower than expected recovery in patient plasma. This lower recovery of
amount of FXa present in the reagent. The remaining free FXa acts on EHL-­FVIII is usually attributed to variable amounts of phospholipid pres-
a chromogenic substrate to generate a color. The higher the amount of ent in various APTT reagents and variable sensitivity of modified FVIII
heparin in plasma the lower the amount of free FXa and, hence, lighter to phospholipids in APTT reagents. Similarly, hemophilia A patients on
the color. This intensity of the color is read as an optical density, and the emicizumab have normal APTT; hence, FVIII levels or FVIII inhibitor
amount of anti-­Xa activity is read off of a standard curve generated with titer based on the one-­stage APTT cannot be measured. Therefore, the
commercial calibrators. This assay is quite robust and, currently, liquid chromogenic FVIII assay is used in such situations. In the chromogenic
reagents with 7 days’ stability on board the coagulation instrument have assay, activated FIX is added to the patient plasma, which then binds to
resulted in replacing APTT in monitoring UFH routinely. The heparin FVIIIa in patient plasma (the bovine thrombin in the reagent converts
assay is known to reduce the number of times that heparin dose has to be FVIII to FVIIIa). The FVIIIa and FIXa complex then acts on FX to give
adjusted as compared with APTT, thus saving both nursing and pharmacy FXa, which, in turn, cleaves a chromogenic substrate to generate a color.
time. Therefore, it is very cost-­effective for the hospitals. The amount of color is proportional to FVIII levels and reads off a stan-
dard curve. The other advantage of chromogenic FVIII assay is that it is
Coagulation Factor Assays unaffected by the presence of LA.
One-­stage clot-­based assays are the most commonly used methods for
coagulation factors. The basic principle is similar for all coagulation Viscoelastic Measurement of Real-­Time Clot
factor assays. A standard curve is prepared by making serial dilutions of PT and APTT are often useful screening tests to assess hemostasis affect-
NPP with the factor-­deficient plasma that is to be measured (e.g., FVIII-­ ing coagulation factors in a bleeding patient. However, they have many
deficient plasma for FVIII assay) to obtain 100%, 50%, 25%, and 12.5% limitations in assessing global hemostasis in real time in a bleeding patient
factor activity. Then, either PT (for FVII, II, V, and X assays) or APTT who needs blood component therapy. Also, the turnaround time for PT
(for FXII, XI, IX, and VIII assays) are performed and plotted against and APTT is not fast enough for surgeons/anesthesiologists to make a
appropriate dilutions (Figs. 40.6 and 40.7). The patient’s plasma is then timely decision in the operating room (OR).
similarly diluted with that particular factor-­deficient plasma, and corre- Thromboelastography (TEG) and rotational thromboelastometry
sponding PTs or APTTs are measured. The value of factor activity in the (ROTEM) are two global hemostasis assays with similar principles for
patient’s plasma is then extrapolated from the standard curve using PT or viscoelastic measurement of a clot in real time (Agren, 2013). Both use
APTT. The rationale for making serial dilution of patient plasma is mostly whole blood and should be used to differentiate between coagulopathic
to dilute out the effect of lupus anticoagulant interference in the APTT-­ and surgical bleeding. They cannot predict a bleeding tendency in a non-
based assay; with each subsequent dilution of patient plasma, the effect of bleeding patient. There is a pin in the cup that records the torsion during
LA is diluted whereas the factor activity increases. This curve obtained in the rotational movement while a clot is formed and various parameters
the presence of LA is known as a nonparallel curve. This pattern alerts the are reflected on a tracing. In TEG, the pin is fixed and the cup rotates; in
laboratory technician to make a further dilution of the patient plasma until ROTEM, the cup is fixed and the pin rotates. The tracing is displayed on
the LA effect has disappeared and the correct factor level is obtained. If this a monitor in the OR thus being a point-­of-­display rather than a point-­of-­
step is not performed, then patients with isolated prolonged APTT may care test because current machines are kept outside the ORs in the United
have spuriously low FVIII activity, which may lead to misdiagnosis and States. The clotting time (CT) in ROTEM or reaction time (R) in TEG
mismanagement of that patient. reflects the amount of thrombin generated. If CT or R is prolonged, this
suggests clotting factor(s) deficiency and plasma transfusion is indicated
Chromogenic FVIII Assay in a bleeding patient. The maximum clot firmness (MCF) in ROTEM or
There are caveats with the one-­stage FVIII assay at present, with many maximum amplitude (MA) in TEG reflects the strength of the clot pro-
extended half-­life (EHL) FVIII concentrates on the market that often show vided by both fibrinogen and platelets. The deficiency of either will reduce

835
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
Add surface XII Add tissue VII
activator, Surface Tissue factor
thromboplastin
exogenous HK
(tissue factor
phospholipid PK XI and exogenous
VIIa
and Ca++ XIIa phospholipid)
and Ca++ Tissue factor/VIIa
IX
XIa
PL, Ca++

Partial Prothrombin Time X


Thromboplastin IXa X (PT) Ca++
Time (PTT) PL, Ca++ Ca++
VIII VIIIa Xa
Xa Prothrombin
Prothrombin PL, Ca++
PL, Ca++
Va V
Va V
Thrombin
Thrombin Fibrinogen
Fibrinogen

Soluble fibrin monomers Soluble fibrin monomers

A Polymerized fibrin clot Polymerized fibrin clot B

Add Thrombin
Fibrinogen
exogenous
thrombin
and Ca++
Soluble fibrin monomers

Thrombin Time
Polymerized fibrin clot
C
Figure 40.7 Coagulation screening tests. A, Activated partial thromboplastin time (APTT, “PTT” on the figure) requires the presence of every protein except tissue factor and
factor VII. B, Prothrombin time (PT) requires tissue factor and factors VII, X, V, and II and fibrinogen. C, In thrombin time (or thrombin clotting time), exogenous thrombin
is added to plasma to proteolyze (clot) fibrinogen. The endpoint in each of these tests is the number of seconds until detection of a clot, following addition of the indicated
reagents to citrated platelet-­free patient plasma. Because virtually all measuring systems employed detect the formation of a polymerized fibrin clot, whether or not any
cross-­linking of fibrin occurs, none of these tests will provide information with respect to factor XIIIa activity.

these values. In ROTEM, FIBTEM is another test run simultaneously cost-­effective (Table 40.2). As mentioned before, preanalytic variables
that contains cytochalasin B to block platelet function. MCF in FIBTEM should be kept in mind when interpreting unexpected test results and
reflects the contribution of only fibrinogen to the clot firmness; if MCF is repeating tests to show that reproducibility is a better practice to avoid
low, that indicates the need for cryoprecipitate transfusion. patient harm. Routine coagulation tests often guide subsequent spe-
On the other hand, if the MCF is normal in FIBTEM but low in cific testing in a bleeding patient or patients with a positive history
EXTEM, it suggests a platelet defect (either number or function), which of bleeding. Investigation of incidentally encountered abnormal coagu-
indicates the need for platelet transfusion in a bleeding patient. In TEG, lation tests should be taken up judiciously to both avoid unnecessary
if MA is low, then a platelet mapping test can be performed to differenti- testing and health care cost, and patient harm by using blood compo-
ate between fibrinogen and platelet defect. In ROTEM, if INTEM CT nent (often plasma) to correct an abnormal test result. Access to prior
is prolonged (EXTEM CT being normal), it suggests the presence of coagulation test results is often useful to differentiate congenital from
heparin, which is then confirmed by HEPTEM that contains hepzyme to acquired bleeding disorders because prior tests are likely to be normal
neutralize heparin and correct prolonged INTEM CT. When EXTEM in acquired disorder versus mostly abnormal in congenital disorders
shows increased clot lysis (>15%), APTEM is performed. APTEM con- (Table 40.3).
tains aprotinin, which inhibits plasmin and shows correction of lysis
parameters. This finding indicates the use of a fibrinolytic agent such as
tranexamic acid or Amicar. FXIII deficiency can also affect clot stability HEREDITARY COAGULATION PROTEIN
and show increased clot lysis.
DEFECTS
Protein or factor deficiencies can be quantitative or qualitative. In quan-
APPROACH TO PATIENTS WITH COAGULATION titative disorders, the factor level determined by routine clot-­based meth-
DISORDERS ods (functional activity assays) is similar to that obtained by immunologic
(antigen) assays. In qualitative disorders, the functional assay result is
A thorough clinical personal and family history is absolutely essential decreased, but the antigen level is significantly higher or normal, indicat-
for appropriate testing and management of patients with a bleeding ing the presence of a dysfunctional protein or an inhibitor to the function
disorder. A systematic step-­by-­step approach is often rewarding and of that protein.

836
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
TABLE 40.2
Patterns of Clinical Bleeding in Disorders of Hemostasis
Characteristics Primary Hemostasis (Platelet/Vascular Problem) Secondary Hemostasis (Coagulation Factor Problem)

Onset Spontaneous, immediate after trauma Delayed after trauma

PART 5
Sites Skin, mucous membranes Deep tissues
Form Petechiae, ecchymosis Hematomas
Mucous membrane Common (nasal, oral, gastrointestinal, genitourinary) Less common
Other sites Rare Joint, muscle, central nervous system, retroperitoneal space
Clinical examples Thrombocytopenia, platelet defects, von Willebrand disease, scurvy Factor deficiency, liver disease, acquired inhibitors

TABLE 40.3 TABLE 40.4


Differential Diagnosis of Abnormal Coagulation Screening Tests Classification of Hemophilia A and B
Abnormal Activated Partial Thromboplastin Time (APTT) Alone CLASSIFICATION

Associated with bleeding: VIII, IX, and XI defects Severe Moderate Mild
Not associated with bleeding: XII, prekallikrein (PK), high-­molecular-­ Percentage of 50–70 10 30–40
weight kininogen, lupus anticoagulants patients
Abnormal Prothrombin Time (PT) Alone Factor VIII or factor <1 1–5 6–30
Factor VII defects IX activity, %
Combined Abnormal APTT and PT Pattern of bleeding ∼2–4 per ∼4–6 per year Uncommon
Medical conditions: Anticoagulants, disseminated intravascular coagula- episodes month
tion (DIC), liver disease, vitamin K deficiency, massive transfusion Cause(s) of bleeding Spontaneous Minor trauma Major trauma
Rarely, dysfibrinogenemia; factor X, V, and II defects Surgery

DEFICIENCY OF FACTOR VIII (HEMOPHILIA A) OR Table 40.2). The treatment of severe hemophilia (FVIII or FIX activ-
ity <1% of normal) has traditionally required frequent administration of
FACTOR IX (HEMOPHILIA B) plasma-­derived or genetically engineered products containing the specific
Hemophilia A (FVIII deficiency) is the most common severe congenital missing clotting factor, using strategies that reverse active bleeding or pre-
bleeding disorder, affecting 1 in 5000 to 10,000 males without ethnic vent spontaneous and minor traumatic hemorrhage (primary or secondary
predilection. Hemophilia B (FIX deficiency, Christmas disease) is also prophylaxis). Newer approaches utilize “disruptive” therapies, which pro-
a severe congenital bleeding disorder, affecting 1 in 25,000 to 30,000 mote thrombin generation and hemostasis by circumventing the need for
males. The FVIII and FIX genes are both located on the X chromosome, the deficient clotting factor (Weyland and Pipe, 2019).
with the result that hemophilia A and B are X-­linked recessive disorders Mild hemophilia (FVIII or FIX activity >5% of normal) is rarely symp-
primarily affecting males. Females carrying a hemophilia mutation on tomatic for spontaneous bleeding. However, it manifests with prolonged
one of their two X chromosomes are carriers. Hemophilia A can affect bleeding after trauma or major surgery, and patients rarely need factor
females when carriers have imbalanced lyonization of the normal X chro- replacement. Moderate severity hemophilia (FVIII or FIX activity 1% to
mosome or Turner syndrome (XO), or when they are daughters of an 5% of normal) is characterized by an intermediate bleeding phenotype.
affected male and a carrier female. Hemophilia is suspected based on bleeding symptoms or a family his-
Hemophilia is defined as mild, moderate, or severe depending on the tory of hemophilia. About one-­third of hemophilia A cases arise from
baseline factor activity level (Blanchette et al., 2014) (Table 40.4). Factor spontaneous mutations. Laboratory evaluation of patients with such a
levels correlate with the degree of bleeding symptoms (Pavlova & Olden- bleeding history should include APTT, PT, and platelet count. Diagnosis
burg, 2013). However, among patients, there is considerable variability in is confirmed by an FVIII or FIX assay. Hemophilia can be diagnosed as
bleeding pattern. early as the birth of a male neonate with a positive family history because
Severe hemophilia A and B usually manifest very early in life, often as FVIII and FIX levels can be directly measured in cord blood.
cephalohematomas at birth (whether a vaginal or cesarean section delivery), It is important to appreciate that in the coagulation laboratory, mea-
postintramuscular vitamin K hematomas, or prolonged bleeding with new- surement of at least two dilutions of patient-­citrated plasma should be
born heel sticks. The classic presentation of the hemophilias was provided assayed in duplicate against standard curves for FVIII or FIX coagulant
in the Talmud, which emphasized the risk of bleeding associated with cir- activity in order to fully characterize the severity of disease in those with
cumcision. Surprisingly, it is uncommon to observe bleeding at the umbili- severe FVIII or FIX deficiency because the dilution curves are not linear
cal stump in those affected by hemophilia A or B. As the child progresses at lower values. FVIII levels above 10% are easier to evaluate because the
into toddler age, bleeding predominantly manifests as acute and recurrent standard curve spans between 10% and 150% FVIII/FIX activity. Varia-
hemarthroses, muscle and soft-­tissue hematomas, and then gum and den- tions of the one-­stage assay method are the most commonly employed
tal bleeding associated with loss of deciduous teeth. In childhood through techniques to measure FVIII or FIX activities in plasma.
adulthood, the bleeding frequency increases in proportion to activity The type of clotting factor concentrate administered can affect assay of
intensity and trauma. However, even the frequency of spontaneous bleed- the factor—for example, assay discrepancies of up to 40% are possible dur-
ing increases—for example, hemorrhage unassociated with known trauma. ing clinical monitoring of recombinant FIX and plasma-­derived FIX con-
This suggests that even the stress of weight-­bearing, normal torque on the centrates (EMA, 2014) and between modified recombinant FVIII (rFVIII)
joints and supporting structures can precipitate bleeding events. The hinge extended half-­life products, such as pegylated FVIII and Fc fusion, versus
joints remain the primary sites of hemophilia-­related bleeding complica- standard (or B-­domain deleted) half-­life recombinant FVIII concentrates
tions throughout life, such as ankles, knees, and elbows, although even the (Nagoya et al., 2019).
ball-­and-­socket joints can bleed occasionally. Repeat hemorrhage into the Several additional comments regarding hemophilia care are pertinent:
same joint often leads to restricted range of motion, support muscle atro- 1. Only 25% of the world’s hemophilia population receives adequate care,
phy with disability, acute and chronic pain, and structural deterioration. and opportunities are still available to provide enough clotting factor
Iron uptake into the joint synovium triggers the local release of proinflam- concentrate to prevent joint damage, long-­term morbidity from bleed-
matory and proteolytic cytokines, particularly IL-­1 beta and IL-­6, which, ing complications, and death from bleeding.
in turn, induce chondrocyte apoptosis and cartilage damage (Van Vulpen 2. The standard of care for severe hemophilia A and B in the resource-­
et al., 2015) via impaired scavenging, degradation, and transport of iron rich world now includes the initiation of primary prophylaxis with the
from the joint synovium. Life-­threatening hemorrhage usually occurs in administration of clotting factor replacement therapy either before or
the brain and paratracheal regions and is typically related to trauma (see shortly after the first bleeding event in childhood. Prophylaxis regi-

837
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
mens should be maintained indefinitely, although some young adults Prenatal diagnosis of the hemophilias can be performed at a number of
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
can do well off prophylaxis. The target FVIII or FIX trough activity high-­risk obstetric centers by chorionic villous sampling at 9 to 14 weeks’
level to maintain should be individualized according to the patient’s gestation or by amniocentesis after the 16th week. If the genetic basis of
lifestyle. the disease is known within a family, then mutational analysis or exami-
3. Primary prophylaxis is not perfect in its ability to prevent joint deterio- nation for the intron 22 inversion can be performed on the cell samples
ration, even when the individual has not experienced any clinical hem- (Ljung, 1999; Arun & Kessler, 2001). Fetoscopic blood sampling can be
orrhage. In addition, there are those rare individuals who manifest no performed at 20 weeks’ gestation for coagulation factor analysis, although
joint deterioration despite recurrent bleeding events (Manco-­Johnson it carries a higher risk and is less precise. However, because fetal deoxy-
et al., 2007). ribonucleic acid is detectable in maternal circulation within 10 days of
4. All of the currently available clotting factor concentrates for FVIII or conception, collection of maternal plasma should be sufficient to make a
FIX replacement are viral safe, without any reported transmission of prenatal diagnosis of hemophilia presently, obviating the use of the inva-
human immunodeficiency virus, hepatitis C virus, hepatitis B virus, and sive diagnostic techniques listed earlier. In hemophilia B, 30% to 40% of
so on. The viral attenuation processes in the pooled human plasma-­ carriers will be missed by measurement of FIX activity, and mutation anal-
derived factor concentrates have rendered these products free from ysis is important for defining carrier status (Graham, 1979; Ljung, 1999).
transmitting such lipid-­ enveloped infectious agents as severe acute Similar to hemophilia A, hemophilia B arises spontaneously in one-­third
respiratory syndrome, West Nile virus, and so forth. However, these of affected individuals.
products still may potentially transmit non-­lipid-­coated viruses, such as
parvovirus B19, despite ultrafiltration. Treatment of Hemophilia
5. The most common cause of severe hemophilia A is a partial inversion The primary principle of hemophilia treatment is to prevent or reverse
of the FVIII gene up to and including intron 22, which accounts for bleeding episodes, spontaneously occurring, traumatic, or surgically
up to 40% to 45% of such patients (Naylor et al., 1992; Lakich et al., induced. The secondary goals of hemophilia treatment are to avoid the for-
1993). These inversions are due to homologous recombination between mation of alloantibody inhibitors, which neutralize the coagulation func-
the region that includes the F8A gene in intron 22 and one of the two tion of exogenously administered replacement products and to improve the
other homologous regions located more than 400 kb 5′ (telomeric) to convenience of treatment in order to improve adherence to any long-­term
the FVIII gene (Lakich et al., 1993; Kaufman et al., 2001). Several types treatment regimens. Well-­conducted prospective clinical trials have indi-
of these inversions have been described (Kaufman et al., 2001). If the cated that the development of FVIII inhibitors is the most serious compli-
intron 22 inversion is not detected, the definition of the responsible cation of FVIII replacement therapy and affects 29% to 45% of previously
mutation is difficult because of the large size of the FVIII gene (ap- untreated patients (PUPs) with severe hemophilia A. In contrast, there is a
proximately 186,000 bp long). In contrast, the FIX gene is one-­fifth the 1.5% to 3% incidence of neutralizing alloantibody inhibitor development
size of the FVIII gene, and the mutations responsible for hemophilia B in PUPs with severe hemophilia B. They predominate in those individuals
are easier to define by genetic analysis. with large FIX gene deletions.
More diagnostic testing may be required for patients with mild to mod- The landscape of hemophilia treatment has expanded rapidly over the
erate deficiency of FVIII for whom a diagnosis of hemophilia A may be past few years and bioengineering innovations have yielded modifications
less apparent (e.g., a female patient with low FVIII and apparent auto- of standard recombinant replacement products that extend their circu-
somal inheritance). In patients with von Willebrand disease, a secondary lating half-­lives. New technologies have also been applied to develop
deficiency of FVIII may occur (see Chapter 41) because FVIII is normally “disruptive” treatment strategies, which circumvent the need to replace
bound to vWF in the plasma (Weiss et al., 1977). Further, a unique vWF the specific deficient clotting protein by promoting thrombin generation
abnormality has been described in which a missense mutation in vWF using “mimetic” proteins or by “rebalancing” normal coagulation through
impairs its capacity to bind to and promote FVIII secretion into plasma. interfering with the natural modulators of the coagulation pathways, that
This abnormality, first described in a French patient from Normandy, has is, tissue factor pathway inhibitor, antithrombin, and so on. Lastly, gene
been named von Willebrand disease “Normandy,” or type 2N. It should therapy is emerging as a potential “cure” for the hemophilias.
be suspected in a patient with normal vWF antigen and functional studies All modern replacement products for hemophilia A and B are viral safe
(see Chapter 41) but reduced FVIII levels, whose inheritance is autosomal and equivalently efficacious in treating and preventing bleeding events. No
recessive rather than sex linked (Schneppenheim et al., 1996). transmission of blood-­borne pathogens (including prion-­related disease)
FIX activity levels during childhood remain at about 75% of adult lev- has been documented for any of the currently available products. Cryopre-
els. A 25% increment in FIX expression begins at puberty in both sexes cipitate for FVIII replacement or fresh frozen plasma for FIX replacement
(Andrew et al., 1992). It has been assumed that this is a result of steroid is no longer considered the standard of care in most of the world.
hormone action. It is interesting to note that a rare form of FIX deficiency, FVIII replacement concentrates are characterized as plasma-­derived
hemophilia B Leyden, undergoes postpubertal phenotypic resolution. (pd) from pooled normal plasma from tens of thousands of donors or as
Patients with this condition present with hemophilia B in early childhood, recombinant genetically engineered products. The plasma-­derived (pd)
with FIX activity ranging from less than 1% to 13% of normal. Plasma products are available as high specific activity, immunoaffinity processed,
levels rise to as high as 70% of normal after the onset of puberty with reso- highly purified FVIII without vWF protein or as low specific activity pd
lution of bleeding complications (Reitsma, 2001). Mutations in hemophilia FVIII concentrates containing substantial amounts of vWF protein. Stan-
B Leyden have been identified in the promoter region of the FIX gene, dard recombinant FVIII concentrates are all ultra-­purified and very high
within which a consensus sequence for steroid receptor binding is located specific activity products, further defined by the presence or absence of
(Crossley et al., 1992). extraneous human or animal proteins in the media bathing the murine-­
derived cell cultures synthesizing the FVIII or at the end commercial
Evaluation of Carriers product. They can be full-­length native FVIII protein or B-­domain deleted
Detection of carriers is useful for the prediction of symptoms and prena- truncated FVIII coagulant protein.
tal counseling. Carriers are expected to have approximately 50% of nor- The category of EHL rFVIII concentrates is further characterized
mal factor activity, a value generally able to prevent bleeding. However, according to how they are bioengineered to convey longer circulating activ-
some hemophilia carriers may experience menorrhagia and other bleeding ity levels. These techniques include pegylation of the B-­domain deleted
symptoms, phenotypically like males with mild hemophilia (Arun & Kes- FVIII construct; covalent bonding of the heavy and light chains of FVIII into
sler, 2001; Plug et al., 2006). In hemophilia A, if the mutation is known, the a single-­chain structure; or B-­domain deleted rFVIII fused to the IgG-­1 Fc-­
potential carrier may be tested for that mutation. Otherwise, the woman domain. EHL rFVIII products are also distinguished according to whether
should be tested for the common intron 22 inversion or get whole exo- they are synthesized by murine-­derived Chinese hamster ovary or baby ham-
some sequencing. A detailed genetic analysis of the woman, the affected ster kidney cell lines or synthesized in human embryonic kidney cell lines.
male, and intervening family members may define the mutation. These Replacement FIX concentrates are available as pd, high specific activity, or
techniques provide an extremely high likelihood of detection of carriers immunoaffinity or dual affinity chromatographically purified products. They
when all family members are available for study. A less optimal, but more are all nano-­filtered and solvent detergent treated to enhance viral safety.
readily available and cost-­effective approach is the calculation of the ratio Standard recombinant FIX concentrates are synthesized by Chinese
of FVIII activity to the level of vWF antigen to predict carrier status. The hamster ovary (CHO) cell lines. EHL rFIX concentrates are synthesized
ratio of FVIII/vWF had a median value of 0.71 (range, 0.18–2.2) in a group in CHO or human embryonic kidney cell lines and are further character-
of 147 carriers versus a median value of 1.39 (range, 0.58–2.48) in a group ized as pegylated, albumin-­fusion, or IgG-­1 Fc domain fusion proteins.
of 25 noncarriers. A value of FVIII/vWF:Ag ratio ≤0. 9 was found in only Although plasma-­derived FVIII and FIX replacement products remain
one noncarrier (4%) versus in 113 (76.9%) carriers (Labarque et al., 2017). commercially available and are very effective in promoting hemostasis in

838
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
individuals with hemophilia A and B, they are believed capable of transmit- (inhibitors) that interfere with the coagulation factor function. Inhibi-
ting nonenveloped viruses such as parvovirus B19 or prions, the agents tors occur in 20% to 30% of patients with severe hemophilia A and 3%
causing Creutzfeldt-­Jakob disease (CJD) and variant CJD (vCJD). These to 5% with severe hemophilia B. They are quite uncommon in patients
pathogens are not eliminated by current viral inactivation and product with moderate or mild hemophilia, probably because the infused factor is
purification technologies and are emblematic of the hypothetical poten- not recognized as a foreign antigen in these individuals (Eckhardt et al.,
tial of these products to transmit yet undiscovered or unrecognized blood-­ 2013). Inhibitors reduce or abolish/neutralize, depending on their titer or

PART 5
borne infectious agents in the future. Thus, in 2014, the Medical and the response to replacement therapy. Periodic inhibitor screening is very
Scientific Advisory Council of the National Hemophilia Foundation advo- important for effective and prompt management of this complication (Sou-
cated recombinant FVIII and FIX concentrates as the treatment of choice cie et al., 2014).
for individuals with hemophilia A and B. Characterization of a specific inhibitor requires careful quantitation. In
The important Survey of Inhibitors in Plasma-­Product Exposed Tod- general, lower-­titer FVIII inhibitors (<5 Bethesda units [BU]) can still be
dlers (SIPPET) study (Peyvandi et al., 2016) reported a cumulative allo- treated with replacement factor. In contrast, higher-­titer inhibitors (>10
antibody FVIII neutralizing inhibitor incidence of 44.5% (28.4% high BU) typically need the administration of the so-­called bypassing agents:
titer) in PUPs treated with recombinant FVIII concentrates produced in recombinant FVIIa, FEIBA (FVIII inhibitor bypassing agent that is an
hamster cell lines and 26.8% (18.6% high titer) with plasma-­derived FVIII activated prothrombin complex concentrate [PCC]), and recombinant por-
(pdFVIII) products containing vWF. As a result of these data, many clini- cine FVIII (Hay et al., 2006). A simple mixing test as described previously
cians believe strongly that PUPs with severe hemophilia A should receive for the APTT can underestimate the presence of an inhibitor. Further, as
plasma-­ derived products containing vWF to reduce the likelihood of indicated earlier, there is no unanimity on how such mixing assays should
inhibitor development. The caveat here is that the individual PUP’s FVIII be performed. In order to diagnose the presence of a FVIII or FIX inhibi-
gene mutation may be critical to determining susceptibility to inhibitor tor, a formal FVIII or FIX “Bethesda” inhibitor assay should be performed
formation (in the SIPPET study, no inhibitors occurred in PUPs with a (Kasper et al., 1975). In the classic Bethesda assay, the patient’s plasma is
low genetic risk [non-­null mutations] who received pdFVIII versus 31% serially diluted with imidazole buffered saline (IBS), then one part patient
inhibitor incidence in PUPs with high genetic risk [null mutations] treated plasma is mixed with one part normal pooled plasma and incubated for
with pdFVIII). In contrast, the risk of inhibitor formation in PUPs with 2 hours at 37°C. The control is a 1 : 1 mix of normal pooled plasma with
either low-­or high-­risk FVIII gene mutations did not differ when treated IBS. In the Nijmegen modification, the IBS is replaced with FVIII defi-
with rFVIII (43% and 47%, respectively). Furthermore, the fact that the cient plasma for serial dilution, which makes this method more sensitive
use of pdFVIII did not eliminate inhibitor formation in PUPs implies that to detect low-­titer inhibitors (Verbruggen et al., 1995). After incubation,
the replacement factor choice is only one of the multiple variables involved FVIII is measured in patient plasmas and the control plasma, and the per-
in inhibitor formation. A second caveat is that the results from the SIPPET centage of residual FVIII activity is determined. One BU is defined as the
study do not apply to inhibitor formation in previously treated hemophilia amount of inhibitor antibody in patient plasma that inhibits half the FVIII
A patients in whom rFVIII products have not been associated with increased activity in the control plasma (Fig. 40.8). Therefore, patient plasma pro-
inhibitor incidence. Lastly, SIPPET concerns cannot be extrapolated at ducing a residual FVIII activity of 50% is considered to contain 1 BU of
this time to the newer generations of EHL FVIII concentrates as these inhibitor per milliliter. If this same plasma had been diluted tenfold before
products have not been examined in similar prospective randomized trials. assaying for the inhibitor, then this result would indicate an inhibitor value
Replacement clotting factor concentrates are dosed on the basis of of 10 BU/mL.
weight and desired plasma activity. Plasma with 100% clotting factor activ- This assay is sensitive, but at times it is difficult to reproduce. A com-
ity has 1 U/mL of that clotting factor. By convention, one unit of a coagu- mon observation is that varying dilutions of plasma may yield different
lation factor is the amount of that factor present in 1 mL of normal pooled estimates of the inhibitor titer. For assigning the level of BUs, the dilution
human plasma. Each unit of intravenously administered pd or recombinant that comes closest to the 50% residual factor is chosen for the calcula-
FVIII/kg ideally raises the plasma FVIII activity by about 2%, and each tion of the FVIII inhibitor titer. Values on the standard curve should be
unit of recombinant FIX/kg raises the plasma FIX activity by about 1%. read-­only at between 25% and 75% residual FVIII activity. If below 25%,
To raise the FVIII plasma activity of a 70-­kg man by 100%, the calculated higher dilutions of the sample are needed. If residual activity using the 1:1
dose will be as follows: diluted patient specimen is above 75%, a demonstrable inhibitor may not
be present. It is, in fact, important for the laboratory to determine a lower
limit of sensitivity of this assay, typically about 0.5 BU. It is additionally of
note that a plot made of residual FVIII activity versus increasing volume of
original patient plasma in the Bethesda assay may distinguish between an
inhibitor capable of reducing FVIII activity into the unmeasurable range
The mean plasma half-­life of standard exogenously administered FVIII (type I inhibitor) and an inhibitor that preserves some degree of residual
to adults with severe hemophilia A is about 8 to 12 hours. EHL recombi- FVIII activity, even at high inhibitor titer (type II inhibitor), presumably
nant FVIII concentrates have mean circulating half-­lives of about 18 to 24 reflecting a target epitope at a less critical region of the FVIII molecule
hours (about 1.5 times normal). (Gawryl & Hoyer, 1982; Luna-­Zaizar et al., 2009).
To raise the FIX plasma activity of a 70-­kg man by 100%, the calcu-
lated dose will be as follows (Shapiro et al., 2005):
Treatment of Acute Bleeds Associated With Neutraliz-
ing Alloantibody Inhibitors in Hemophilia A and B
The approach to the reversal/treatment of acute hemorrhagic episodes is
predicated on circumventing the neutralizing effects of antibodies on exog-
enous replacement therapies. In the context of low responding inhibitors
About one-­third of FIX-­deficient patients have a lower recovery after (≤5 BU and no anamnestic response to reexposure to exogenous FVIII or
recombinant FIX infusion, requiring 20% higher dosing. The mean plasma FIX), high doses of any construct human FVIII or FIX concentrate (200
half-­life of exogenously administered pd or standard recombinant FIX to IU/kg) can be administered to saturate and “overwhelm” the respective
adults with severe hemophilia B is about 18 to 24 hours. EHL recombinant inhibitor. The caveat to this approach is that the incremental increase
FIX concentrates in adults have mean circulating half-­lives ranging between in clotting factor activity (recovery) and the circulating half-­life will be
82 to 110 hours (about 5 times normal). The mean circulating half-­lives of blunted by the effects of the inhibitor. Second, for hemophilia B patients,
FVIII or FIX concentrates in young children up to 6 years old are about 15% particularly those with large FIX gene deletions, there is a small but sig-
to 20% lower than in adults due to accelerated plasma clearances. Dosing nificant incidence of developing potentially serious and life-­threatening
strategies in young children should take these pharmacokinetic differences allergic or anaphylactic reactions to FIX and some also develop nephrotic
into consideration. The very active person with hemophilia may wish to syndrome. In these patients, as well as those with high responding allo-
maintain a trough clotting factor activity level of 15% to minimize bleeding antibody inhibitors to FVIII or FIX (>5 BU and anamnestic responses to
events during rigorous athletics or work-­related requirements. In contrast, reexposure to the clotting factor), the treatment of choice for acute bleeds
for the sedentary individual, a trough level of 1% may be sufficient. is a “bypassing agent,” such as FVIII inhibitor bypass activity (FEIBA) or
recombinant FVIIa.
Complications of Treatment On the other hand, recombinant porcine B-­domain deleted FVIII con-
An important complication of replacement therapy, occurring mainly centrate, synthesized in CHO cells to treat acute bleeding in acquired auto-
in patients with severe hemophilia, is the development of alloantibodies antibody FVIII inhibitor patients, has also been used in phase II clinical

839
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
100 Laboratory monitoring of emicizumab is not required while on routine
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
prophylactic dosing. In fact, the APTT will be artefactually decreased and
the one-­stage FVIII activity assay will be misleadingly elevated and will
75 not accurately correlate with the clinical hemostatic status of the patient.
Chromogenic FVIII activity assays that use bovine reagents do measure
FVIII activity in emicizumab patients who have received an exogenous
FVIII replacement. Similar limitations occur with the clot-­based Bethesda
assay; a bovine-­reagent chromogenic-­based Bethesda inhibitor assay is
50 available. Emicizumab can be measured in plasma, but this does not pro-
vide a reliable “FVIII-­equivalence” assay to predict in vivo hemostasis.
Another emerging disruptive therapy for hemophilia A or B with or
Residual factor VIII (%)

without inhibitors is fitusiran (Sanofi-­Genzyme), which interferes with


antithrombin synthesis in the liver and, thus, leads to increased thrombin
generation in vivo. Fitusiran binds to and degrades messenger ribonucleic
acid for antithrombin and thereby interferes with AT translation by the
hepatocyte. This synthetic, double-­stranded small interfering ribonucleic
25 acid oligonucleotide is also administered subcutaneously as a weekly or
monthly therapeutic; clinical trials have demonstrated reduced annualized
bleeding rates in patients with hemophilia A and B, with or without inhibi-
tors (Pasi et al., 2017). In these studies, two severe hemophilia A patients
have developed a fatal thrombosis after concurrent repeated administra-
tion of high-­dose factor VIII concentrate. Theoretical benefits have been
speculated for fitusiran to prevent bleeding complications in other rare
bleeding disorders, such as severe deficiency of factors V, VII, X, and XI.
No clinical trials in these latter coagulopathies have been conducted as yet.
Concizumab (Novo Nordisk) is the third disruptive therapy for hemo-
philia A and B with and without inhibitors. It is a daily subcutaneously
administered high-­affinity, anti-­TFPI monoclonal antibody, which results
10 in an increased generation of FXa and, ultimately, increased thrombin gen-
0 1.0 2.0 eration. Clinical trials have demonstrated decreased annualized bleeding
Bethesda units per mL rates with few side effects (Shapiro et al., 2019). That being said, the clini-
Figure 40.8 Expression of factor VIII inhibitor titer in Bethesda units. In this assay cal trials with a different anti-­TFPI construct (Bayer) were terminated due
for an inhibitor to factor VIII, the percent of residual factor VIII activity in normal to thrombotic complications.
plasma is determined after incubation with patient plasma. By convention, 1 in-
ternational Bethesda unit of inhibitor is the amount of antibody that destroys 0.5 Gene Therapy for the Hemophilias
U factor VIII activity after 2 hours of incubation at 37°C. As shown in the figure, the Congenital hemophilia A and B appear to be ideal disorders for gene ther-
percent of residual factor VIII activity on a log scale is plotted against Bethesda units/
mL in a linear scale. For a sample to be evaluable in this assay, the level of factor VIII apy for the following reasons:(1) FVIII and FIX genes were cloned in the
activity should fall between 25% and 75%. Patient samples that produce residual early 1980s, (2) the proteins expressed by the genes (FVIII/FIX) are easily
factor VIII activity below 25% need to be diluted further so that the 50% residual measured in small amounts of plasma, (3) incremental increases in pro-
factor VIII activity point can be found. (Redrawn with permission from Bockenstedt tein expression due to gene therapy could change the phenotypic bleeding
PL: Laboratory methods in hemostasis. In Loscalzo J, Schafer AI, editors: Thrombosis profile in a significant manner, (4) hemophilic animal models have been
and hemorrhage, ed 3, Philadelphia, 2003, Lippincott Williams & Wilkins, p 370,
Fig. 21.7.)
developed to provide proof of principle that gene therapy would be feasible
and useful, and (5) individuals with congenital severe hemophilia A and
B have been enthusiastic and willing to participate in gene therapy clini-
trials to treat bleeding in allo-­FVIII antibody inhibitor patients with con- cal trials with the potential that their bleeding rates and the consequences
genital hemophilia A. The product has demonstrated good efficacy and therefrom could be eliminated or at least ameliorated.
safety (Mannucci & Franchini, 2017) but has not been licensed by the FDA The most successful clinical gene therapy studies currently employ
for use in congenital hemophilia A with inhibitors. Recombinant porcine nonintegrating recombinant adenoassociated viral (AAV) vectors to carry
FVIII was developed on the premise that there is a lower cross-­reactivity of the genetic “payload” to the targeted hepatocytes for cellular transduction.
porcine FVIII with antihuman FVIII antibodies. Although no long-­term immunologic or oncologic toxicities have been
observed with either FVIII or FIX gene therapies to date, longitudinal
Disruptive Therapies for the Hemophilias vigilance is indicated. In addition, gene therapy trials have been hampered
The concept of “rebalancing” the coagulation cascade to generate throm- somewhat by the presence of preexisting neutralizing antibodies to the
bin and to establish hemostasis without specific replacement of the defi- AAV capsid in potential participants. Also, all of the gene therapy trials
cient coagulation factor protein has evolved rapidly and has yielded novel have observed some degree of lost transgene expression over time, attrib-
therapeutic strategies for hemophilia A and B with and without alloanti- uted to either episomal latency within the hepatocyte or by yet to be fully
body inhibitors. elucidated immunologic mechanisms.
Emicizumab-­kxwh (Hemlibra, Genentech/Roche) is the first commer- The most successful FIX gene therapy results have been accomplished
cially available agent in this category. It is indicated for the prevention of in trials that incorporate a FIX molecule with a gain-­of-­function FIX
bleeding in any individual (infants and adults, but there are no clinical efficacy mutation (FIX-­Padua) with AAV capsids (Batty & Lillicrap, 2019). Some
or safety data in patients less than 1 year of age) with severe hemophilia A patients have experienced sustained FIX activity levels in the 30% to 40%
with and without inhibitors (Barg et al., 2019). It has the advantage of being range (compared to baseline levels of ≤2%). Gene therapy trials in severe
subcutaneously administered in weekly, every other week, or every 4 weeks hemophilia A have also achieved clinically significant incremental increases
dosing regimens. Emicizumab is a genetically engineered bispecific IgG anti- in FVIII levels, frequently in the normal range (≥50% activity). The dura-
body that functions as an “FVIII mimetic” by substituting for the severely bility of the response has emerged as an issue to watch over time in these
deficient FVIII and serving as a cofactor to produce the tenase complex in patients (Pasi et al., 2020).
the coagulation cascade. As a substitute for FVIIIa, it bridges FIXa and FX to
facilitate hemostasis. This is critical to the ultimate thrombin generation and
multiple well-­conducted clinical trials have demonstrated impressive reduc- HEREDITARY DEFICIENCIES OF OTHER COAGULA-
tions in annualized bleeding rates in severe hemophilia A with or without
inhibitors (Mahlangu et al., 2018, Oldenburg et al., 2017). The most con-
TION FACTORS
cerning adverse events within these clinical trials, only in inhibitor patients The other hereditary deficiencies of coagulation factors have an autosomal
who received repeated and large amounts of FEIBA for breakthrough bleeds, inheritance (Table 40.5). With the exception of FXI deficiency, these dis-
was the development of thrombotic complications in 5 individuals. A unique orders are very rare. However, they have a higher prevalence in areas where
feature of these hypercoagulable events is the presence of thrombotic micro- consanguineous marriage is practiced (Mannucci et al., 2004). Deficiencies
angiopathic anemia and the unusual location of some of the thrombi. of factors XIII, X, VII, V, and II are defined rare inherited coagulation

840
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
TABLE 40.5
Characterization of Coagulation Factors and Their Deficiencies
Molecular Normal Circulating
Factor Weight, kDa Gene Location Half-­Life Incidence Inheritance Bleeding Severity

Fibrinogen 330 4q31.3-­q32.1 2–4 days 1 : 1 million Recessive Mild–severe*

PART 5
II 72 11p11.2 3–4 days Very rare Recessive Mild–moderate
V 330 1q24.2 36 hours 1 : 1 million Recessive Moderate
V and VIII com- — LMAN1:18q21.32 36 hours for FV; 10–14 1 : 2 million Recessive Mild–moderate
bined MCFD2:2p21 hours for FVIII
VII 50 13q34 3–6 hours 1 : 500,000 Recessive Mild–severe
VIII 330 Xq28 10–14 hours 1 : 10,000 Sex-­linked Mild–severe
IX 56 Xq27 18–24 hours 1 : 30,000 Sex-­linked Mild–severe
X 58 13q34 40–60 hours 1 : 500,000 Recessive Mild–severe
XI 160 4q35.2 40–70 hours Rare† Recessive Mild–moderate
XII 80 5q33-­qter 50–70 days Rare Recessive No bleeding
PK 88 4q33-­q35 Not known Very rare Recessive No bleeding
HK 120 3q27 9–10 hours Extremely rare Recessive No bleeding
XIII 320 A:6p25.1 11–14 days <1 : 1 million Recessive Moderate–severe
B:1q31.3

HK, High-­molecular-­weight kininogen; PK, prekallikrein.


*May be associated with thrombosis.
†Rare except in those of Ashkenazi Jewish descent

disorders (RICDs) that represent 2% to 5% of inherited coagulation fac- sample conditions appears to be able to identify the bleeding phenotype
tor defects in the general population. The most common RICDs include of patients with FXI deficiency (Pike et al., 2015). In all patients with
deficiencies of FXI and FVII, followed by FV, FX, fibrinogen, and FXIII FXI deficiency, preoperative management is critical to prevent bleeding
(Ruiz-­Saez, 2013). complications. Patients with a severe deficiency (FXI <10%–20%) should
In general, most patients with RICDs have partial defects because they receive treatment with fresh frozen plasma to raise their FXI levels. Usu-
have relatively mild bleeding disorders. Most coagulation-­based assays ally, replacement therapy of 20 mL/kg loading dose with a maintenance
have difficulty detecting these factors once their levels are below 5% to dose of 5 to 10 mL/kg every 24 hours is sufficient to cover patients with
10%. In vivo, however, the presence of even this small level of coagulation severe FXI deficiency through elective surgery (Kessler et al., 1996).
factor can greatly influence bleeding risk. In contrast, findings in knock- Patients with levels from 20% to 70% may or may not bleed.
out mice indicate that a true null of each of these proteins is associated The severity of the surgery, the patient’s past history of bleeding
with virtually 100% mortality from hemorrhage before or at the time of following prior surgeries, and perhaps the bleeding histories of fam-
birth. With hereditary factor deficiencies, heterozygous-­ deficient indi- ily members with the same level of FXI activity should be considered
viduals have approximately 50% (most commonly, 30%–60%) of the nor- when the strategy for bleeding prevention is being formulated. Antifi-
mal level of the affected factor. The symptoms of these disorders are quite brinolytic therapy with tranexamic acid or epsilon aminocaproic acid
variable. Coagulation factor deficiencies may be suspected on the basis of has been a very effective adjunctive therapy when used alone or in com-
symptoms, family history, or abnormal screening tests, and the laboratory bination with replacement therapies. This is particularly useful in the
diagnosis is confirmed by specific factor assays. Once a specific protein United States, where only fresh frozen plasma or solvent/detergent (S/D)
deficiency is recognized, clinical history usually distinguishes a congenital treated, pooled human plasma (Octaplas, Octapharma) is available for
from an acquired defect. FXI replacement therapy.
Pooled human plasma-­derived FXI concentrates are available in France
Disorders with Prolonged APTT and Normal PT and the United Kingdom (LFB Hemoleven and BPL FXI), but neither
has been approved by the United States FDA. Rare arterial and venous
Factor XI thrombotic episodes have been reported with both (or that can have a
FXI deficiency is common in Ashkenazi Jews, in whom heterozygote prothrombotic effect) (Bauduer et al., 2015; Ling et al., 2016). Off-­label
frequency is 8% (Asakai et al., 1991; Emsley et al., 2010). This ethnic use of lower-­ dose (15–30 μg/kg) recombinant factor VIIa concentrate
population constitutes about 50% of the FXI-­deficient patients seen in (NovoSeven) has also provided effective and safe hemostasis for surgery in
the United States. Most patients with FXI deficiency rarely have a spon- severe FXI deficiency. No large clinical trials have utilized this approach
taneous hemorrhage. This notion is further supported by the lack of any (O’Connell et al., 2008, Riddell et al. 2011).
spontaneous bleeding reported in a recent trial in which antisense treat- The development of neutralizing antibodies that function as inhibitors
ment was administered to decrease the activity of FXI to 25% normal in to exogenously administered sources of FXI in patients with severe FXI
knee replacement surgery with the aim to reduce the incidence of venous deficiency have been described, although these are rare (Salomon et al.,
thrombosis (Buller et al., 2015). Bleeding typically occurs after trauma 2003). These patients should be treated with rFVIIa concentrate (Kenet
or surgery, particularly involving areas of the body with high fibrino- et al., 2009).
lytic activity (mouth, nose, genitourinary tract). Women can experience
menorrhagia and postpartum hemorrhage. Bleeding can occur in het- FXII, Prekallikrein, and High-­Molecular-­Weight Kininogen
erozygotes and does not necessarily correlate with the residual FXI level FXII, prekallikrein, and HMWK deficiencies are associated with pro-
(Leiba et al., 1965; Bolton-­Maggs et al., 1988). However, the bleeding longed APTT but are not associated with any bleeding risk. FXII defi-
tendency may be modified by additional defects such as hemophilia, von ciency is most common, occurring in all racial and ethnic backgrounds. It
Willebrand disease, and platelet function defects (Brenner et al., 1997). is associated with a very long PTT. Prekallikrein deficiency is less common
Diagnosis depends on the determination of the FXI activity below the (Girolami et al., 2014) but is seen in the United States in all ethnic groups.
reference range. The most widely used APTT reagents will have pro- It is associated with a slightly prolonged APTT that corrects to normal
longed APTT results for patient samples with FXI activity below 20% when sitting on a bench for 1 hour at 37°C. HMWK deficiency is a rare
to 25% and mixed results when levels are 25% to 60%. The lower limit disorder that also is associated with a very long APTT. These protein defi-
of the normal range is probably between 60% and 70% (Bolton-­Maggs ciencies are not associated with bleeding—a point emphasized diagram-
et al., 1995). Therefore, a normal APTT does not rule out a mild FXI matically in Figure 40.1 by the graying of this portion of the coagulation
deficiency. In cases in which the marked elevation of FVIII may be pres- “cascade.” Because no replacement therapy for hemostasis is necessary for
ent, APTT can be normalized even when other factors are reduced FXII, PK, or HMWK deficiency, it is important to recognize these defects
(Lawrie et al., 1998). Thrombin generation assay performed in specific to prevent unnecessary treatment. Deficiencies of FXII, PK, and HMWK

841
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
have been associated with reduced thrombosis risk. Deletions of these pro- FII (Prothrombin) Deficiency
teins in mice are associated with delayed times to thrombosis in arterial Prothrombin deficiency may be the rarest inherited coagulation factor
thrombosis models (Renne et al., 2005; Stavrou et al., 2015; Merkoulov deficiency (1 : 2,000,000) (Bolton-­Maggs et al., 2004). A true prothrombin
et al., 2008). When a deficiency in the proteins of the kallikrein/kinin sys- deficiency in mice is incompatible with life after birth (Sun et al., 1998).
tem is suspected, a FXII assay should be performed first because it is the Hypoprothrombinemia (type I deficiency) manifests as a concomitant
most common. reduction in prothrombin activity and antigen levels. Bleeding symptoms
include mucosal bleeding, hematomas, and hemarthrosis. Dysprothrom-
Disorders with Prolonged APTT and PT binemia (type II deficiency) presents with reduced activity and normal
Prolonged PT and APTT most commonly are due to general medical antigen levels. The clinical presentation of dysprothrombinemia is less
conditions described later in the Acquired Coagulation Disorders section. predictable—patients may be asymptomatic or may have only mild bleed-
When hereditary disorders are considered, the following, although much ing manifestations (Bolton-­Maggs et al., 2004). Depending on the sensitiv-
less common, should be considered. ity of the reagents, both PT and APTT may be prolonged in prothrombin
deficiency. However, a specific FII assay is best if there is clinical suspi-
Disorders of Fibrinogen cion or positive family history in the presence of normal screening tests. A
Hereditary fibrinogen abnormalities include two categories of plasma PT reagent-­based prothrombin assay with factor-­deficient plasma is most
fibrinogen defects: type I, afibrinogenemia or hypofibrinogenemia, with convenient. Purified or recombinant factor II is currently not available.
absent or low plasma fibrinogen antigen levels (quantitative fibrinogen Prothrombin complex concentrates are the treatment of choice, although
deficiencies); and type II, dysfibrinogenemia or hypodysfibrinogenemia, fresh frozen plasma can serve as an alternative source of prothrombin. The
which determines normal or reduced antigen levels associated with the half-­life of the protein is long (2–4 days); thus, replacement therapy every
disproportionately low functional activity (qualitative fibrinogen deficien- 3 to 4 days is typically sufficient.
cies). In the type I disorder, null mutations occur in the FGA, FGB, and
FGG fibrinogen encoding genes (de Moerloose et al., 2013). FV Deficiency
Fibrinogen deficiencies may prolong the PT and APTT if the plasma Heterozygous FV deficiency is usually asymptomatic. Homozygous FV defi-
concentration of the protein is sufficiently low, usually less than 100 mg/ ciency is rare (1 : 1,000,000), presenting in children with a mild to severe
dL. Afibrinogenemia is a bleeding disorder of variable severity (Fried & bleeding phenotype (Girolami et al., 1998; Lak et al., 1999). Individuals with
Kaufman, 1980; Lak et al., 1999). Umbilical stump and mucosal bleeding severe factor V deficiency (FV:c <1%) may well have some functional FV
are the most common symptoms, as is an increased incidence of muscu- considering that 50% of true null mice die at the time of development of
loskeletal and central nervous system bleeding. Patients also exhibit poor the cardiovascular system (days 9 to 11), and the other half die at birth from
wound healing. Hypofibrinogenemia is a decreased level of normal fibrino- hemorrhage (Cui et al., 1996). A biological reason for a milder than expected
gen that has a similar but milder pattern of bleeding. Both afibrinogen- bleeding phenotype in FV-­deficient subjects may be related to the presence
emia and hypofibrinogenemia are associated with recurrent miscarriage, of residual factor V activity in platelets (Bouchard et al., 2015). The most
as well as with antepartum and postpartum hemorrhage (Goodwin, 1989; frequent symptoms are mucosal tract bleeding and bleeding after invasive
Kobayashi et al., 2000). Paradoxically, reports have described thrombotic procedures. FV deficiency is associated with prolongation of both the APTT
events in patients with afibrinogenemia (Chafa et al., 1995; Lak et al., 1999; and PT (but a normal thrombin time), and it is confirmed by a PT reagent-­
Dupuy et al., 2001). based, single-­stage FV assay. Patients with FV deficiency should also have
Dysfibrinogenemia is a qualitative fibrinogen deficiency characterized an FVIII assay performed to evaluate for combined deficiency of FV and
by the production of dysfunctional fibrinogen (Miesbach et al., 2010). FVIII (Nichols et al., 1998; Zhang & Ginsburg, 2004). The only suitable
Most patients with congenital dysfibrinogenemia are heterozygous replacement product available is fresh frozen plasma (solvent/detergent if
because of molecular defects in produced protein, although rare homozy- available). Human platelets contain 20% of total plasma FV; they can also
gous cases have been reported. Dysfibrinogenemias are most commonly be a source of this protein in acute bleeding (Tracy et al., 1982). The goal
acquired in association with liver disease. Acquired dysfibrinogenemias of replacement therapy should be to elevate the FV level to at least 10% to
are mainly due to posttranslational modifications of the fibrinogen pro- 15% (Peyvandi & Mannucci, 1999). The half-­life of FV is quite short (16–36
tein as a result of synthesis in an abnormal liver. These defects are com- hours); thus, to achieve the hemostatic level of 20%, daily infusions may be
mon in patients with hepatitis B and C. Patients with dysfibrinogenemia needed. During treatment, secondary volume overload needs to be taken
are usually asymptomatic or have mild bleeding. In some cases, however, into account and actively treated. Acquired inhibitors to FV are relatively
thrombosis has been reported with or without a bleeding history (Hanss common from the surgical use of topical bovine thrombin to aid incision
& Biot, 2001). The thrombin time and reptilase time, which measure hemostasis. The bovine thrombin is contaminated with bovine FV; when
clotting time during the conversion of fibrinogen into fibrin, are often the patient makes an antibody against bovine FV, that antibody then cross-­
prolonged in dysfibrinogenemia. When thrombin proteolyzes fibrino- reacts with human FV, causing the deficiency. Most cases are not severe, but
gen in the thrombin time, fibrinopeptides A and B are released from the their management can be challenging due to the lack of specific replacement
Aα and Bβ chains of fibrinogen. Reptilase clots fibrinogen by liberating therapy (Ardillon et al., 2014). Acquired FV deficiency is treated with plate-
only fibrinopeptide A (Funk et al., 1971). Proteolysis of fibrinopeptide A let transfusion because FV in platelets is protected from the antibody and
from fibrinogen is sufficient to induce clot formation. Fibrinopeptide B FV is delivered to the site of injury by platelets. Plasma infusion is ineffective
liberation increases the rate of association of the fibrin monomers, but because the antibody will immediately neutralize the transfused FV.
not the actual physiologic clot (Martinelli & Scheraga, 1980; Nawara-
wong et al., 1991). Bleeding risk is associated only with fibrinopeptide FX Deficiency
A release defects, not with fibrinopeptide B release defects. Therefore, Heterozygous FX deficiency is asymptomatic. Homozygous FX deficiency
a patient can have long PT and APTT from a fibrinopeptide B release is a severe bleeding disorder that presents in infancy. The bleeding phe-
defect and not have any bleeding risk—hence, the reason for using both notype correlates well with FX activity levels. Symptoms include umbilical
the thrombin time and reptilase time to assess dysfibrinogenemias. Assays cord bleeding, mucosal bleeding, severe soft-­tissue hematomas, and hem-
that measure clottable fibrinogen will show lower levels than assays that arthroses (Peyvandi et al., 1998a). One-­stage PT-­based FX assays are suf-
measure fibrinogen antigen. A normal fibrinogen has a ratio of clottable ficient for diagnosis, although additional assays are commercially available.
fibrinogen activity to fibrinogen antigen greater than 95%. Ratios less Before the diagnosis of an inherited deficiency is made, it is important to
than this raise the possibility of a dysfibrinogenemia. For many years, rule out a vitamin K deficiency or other acquired causes for the FX defi-
cryoprecipitate has served as a good source of fibrinogen when replace- ciency. For example, FX deficiency is the most common factor deficiency
ment is needed. A number of virally inactivated fibrinogen concentrates associated with primary amyloidosis, occurring in less than 10% of patients
are now available for therapeutic administration, both in Europe and and probably due to adsorption of FX onto amyloid fibrils (Uprichard &
North America. Human fibrinogen viral attenuated concentrates (Ria- Perry, 2002). Factor X concentrates are available in some countries in
STAP [CSL Behring] and Fibryga [Octapharma]) are available for the Europe. Bleeding can also be treated with prothrombin complex concen-
treatment and prevention of acute bleeding episodes in persons with con- trates, fresh frozen plasma, and recombinant FVIIa (Boggio & Green,
genital fibrinogen deficiency, including afibrinogenemia and hypofibri- 2001). The half-­life of factor X (40–60 hours) may require daily treatment
nogenemia. Primary prophylaxis is easily achieved with these products; or continuous infusion to keep FX levels above 20%.
however, thrombotic complications may arise.
These concentrates are not indicated for acquired noncongenital hypo- Combined Deficiency of FV and FVIII
fibrinogenemias, including those with dysfibrinogenemia, DIC, massive Combined FV and FVIII deficiency is a rare autosomal-­recessive disor-
postpartum hemorrhage, and others. der that results from a single gene defect rather than from coinheritance

842
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
of defects in both FV and FVIII genes. This defect of the endoplasmic assays such as PT, PTT, and TT are normal. A reduced plasma factor
reticulum results in impaired transport of both FV and FVIII from the XIII activity, determined with an immunoassay for factor XIII, leads to the
intracellular site of synthesis. Patients typically have FV and FVIII levels diagnosis. Clot dissolution in 5M urea or monochloroacetic acid is useful
between 5% and 30%, their bleeding phenotype is mild to moderate, and for the diagnosis (Lorand et al., 1980), but these assays are not standard-
it is weakly related to the factor level activity. In two-­thirds of patients, this ized and have low sensitivity. More robust quantitative activity assays are
results from the null expression of LMAN1 (previously known as ERGIC- based on cross-­linking of glycine–ethyl ester into a specific peptide (Fick-

PART 5
­53) (Nichols et al., 1998). LMAN1 shuttles between the endoplasmic retic- enscher et al., 1991), incorporation of an amine substrate into fibrinogen
ulum and the Golgi and is believed to facilitate protein trafficking through (Kohler et al., 1998), or incorporation of a biotinylated peptide to spermine
the secretion pathway (Zhang et al., 2003). Other patients have a mutation attached to microtiter plates (Hitomi et al., 2009).
of MCFD2, which directly interacts with LMAN1 (Zhang et al., 2003). Testing for FXIII deficiency is suggested for individuals with a posi-
Bleeding manifestations include epistaxis, easy bruising, menorrhagia, tive bleeding history, particularly with features such as delayed bleeding,
and postpartum hemorrhage, as well as bleeding following surgery, den- umbilical stump bleeding, or miscarriage, in which the PT and APTT are
tal extraction, and trauma (Seligsohn et al., 1982; Peyvandi et al., 1998b). normal (Anwar & Miloszewski, 1999). Of severely FXIII-­deficient patients,
Testing usually demonstrates disproportionate prolongation of the PTT 30% die in middle age from spontaneous intracerebral hemorrhage unless
compared with the PT. Replacement therapy is usually reserved for surgi- they are receiving prophylactic therapy (Lorand et al., 1980). Acquired
cal, traumatic, or gynecologic bleeding (Mumford et al., 2014). It includes deficiency of FXIII has been described in a variety of diseases, including
both FVIII concentrates and fresh frozen plasma (as a source of FV). Henoch-­Schönlein purpura, isoniazid treatment for tuberculosis, various
forms of colitis, erosive gastritis, and some forms of leukemia (Board et al.,
Combined Deficiency of Vitamin K–Dependent Clotting Factors 1993), although FXIII level determination and therapeutic management
Vitamin K–dependent clotting factor deficiency affecting multiple vita- are controversial. The half-­life of FXIII is very long; thus, replacement
min K–dependent coagulation (II, VII, IX, and X) and anticoagulant with cryoprecipitate is usually sufficient for most patients. Plasma-­derived
(protein S and protein C) factors occurs with vitamin K deficiency and and recombinant FXIII concentrates are currently commercially available
hepatic dysfunction. It also results from heritable dysfunction of hepatic (Dorey, 2014).
enzyme γ-­glutamyl carboxylase (type I) or the vitamin K epoxide reductase
enzyme complex (type II) (Brenner et al., 1998; Zhang & Ginsburg, 2004). Hereditary Hemorrhagic Disorders of Fibrinolysis
Severely affected individuals may present as neonates with fatal hemor- Hereditary bleeding disorders resulting from excessive fibrinolysis are
rhages, umbilical stump bleeding, or spontaneous intracranial hemorrhage; rare. α2-­Antiplasmin deficiency determines enhanced plasmin activity and
in infancy or early childhood with hemarthroses, soft-­tissue hematomas, fibrinolysis (Fay et al., 1992; Aoki et al., 1980). Patients typically present
or gastrointestinal hemorrhages; or as adults with easy bruising, mucosal with delayed bleeding. Measurement of α2-­antiplasmin with either immu-
bleeding, and bleeding following surgery. Diagnosis is established by the nologic or functional assays leads to the diagnosis. Plasma transfusion and
prolongation of both APTT and PT and associated reductions in levels of oral fibrinolysis inhibitors can control bleeding episodes and enable surgi-
vitamin K–dependent clotting factors. These patients may respond to vita- cal procedures.
min K (oral or parenteral) supplementation with normalization of APTT, Acquired α2-­antiplasmin deficiency has been described in amyloido-
PT, and factor levels, as well as the resolution of bleeding symptoms. This sis, DIC, acute promyelocytic leukemia malignancies, severe liver disease
makes the establishment of a diagnosis of an inherited abnormality dif- (impaired synthesis), nephrotic syndrome (increased renal excretion),
ficult in that other acquired causes of vitamin K deficiency (hemorrhagic abdominal aortic aneurysm, and following the administration of thrombo-
disease of the newborn, liver disease, and prolonged use of broad-­spectrum lytic agents (Okajima et al., 1994). Although not recommended for routine
antibiotics) or factitious warfarin administration could manifest similarly. use as a screening test, the whole-­blood clot lysis time can be helpful as
For patients who do not respond fully to vitamin K administration, fresh a first-­order test to detect deficiencies of α2-­antiplasmin. In brief, whole
frozen plasma can be used for acute bleeding or surgery. In addition, pro- blood is allowed to clot; then, the time to clot lysis is recorded. In normal
thrombin complex concentrates and combination therapy and vitamin K individuals, the presence of α2-­antiplasmin effectively prevents lysis from
supplementation may constitute alternative treatment options (Napolitano occurring in this in vitro setting, even 24 hours after initial clot forma-
et al., 2010). tion. In contrast, in the presence of a severe deficiency of α2-­antiplasmin
or in some fibrinolytic states, clot lysis can be observed after several hours.
In such instances, follow-­up testing specific for the α2-­antiplasmin activity
Disorders with Normal APTT and Prolonged PT should be employed. Also, a euglobulin lysis time (ELT) can evaluate the
FVII Deficiency global function of the fibrinolytic system. Plasma is admixed with dilute
FVII deficiency is the most common rare hereditary coagulation factor acid to precipitate a plasma fraction relatively rich in plasminogen activa-
deficiency (Mariani & Bernardi, 2009). The bleeding manifestations are tor, plasminogen (euglobulin fraction), and fibrinogen but relatively poor
variable, with epistaxis, mucosal bleeding, and menorrhagia commonly in antiplasmins. This euglobulin fraction is then redissolved in the buf-
reported (Peyvandi et al., 1997). Severe FVII deficiency is autosomal fer and clotted by recalcification; the time for clot lysis is then measured.
recessive, often presenting shortly after birth, and may have a dramatic Euglobulin clot lysis is normally complete in 2 to 5 hours, but the time may
presentation with intracranial hemorrhage in 15% to 60% of cases (Ragni be shortened with increased fibrinolysis associated with the increased plas-
et al., 1981). The diagnosis is suspected with the finding of isolated pro- minogen activator activity. A shortened ELT is an expression of a hyperfi-
longation of PT. However, FVII, a vitamin K–dependent clotting factor, is brinolytic state; also, it can be adopted to rule out acquired disorders such
low in the newborn period and will also be low in the presence of vitamin as DIC. Plasminogen activator, plasminogen, and plasminogen activator
K deficiency. Therefore, the reevaluation of infants with mild deficien- inhibitor may be assayed directly.
cies is required after they reach a few months of age or after vitamin K
replacement in older patients. Functional FVII activity is measured by a ACQUIRED COAGULATION DISORDERS
PT-­based, FVII-­deficient plasma coagulant assay. The use of recombi-
nant human thromboplastin will yield results that are more likely to reflect Acquired bleeding disorders are due to anticoagulation, DIC, liver disease,
in vivo FVII levels. Samples for FVII testing should not be stored at 4°C, as vitamin K deficiency, massive transfusion, and specific inhibitors of coagu-
this can lead to cold activation of FVII as a result of C1 inhibitor inactiva- lation proteins. The topic of anticoagulation is addressed extensively in
tion and FXIIa formation in the tube with FVII activation (Kitchen et al., Chapter 43. In general, prolongations of PT and APTT arising in a patient
1992). Cold activation of FVII results in an overestimation of the actual should raise the possibility, as discussed in the following sections, of anti-
plasma FVII level. Therapeutic options include recombinant FVIIa (15–20 coagulants, DIC, liver disease, vitamin K deficiency, and massive transfu-
μg/kg), fresh frozen plasma, or 4-­factor prothrombin complex concen- sion. Only after these clinical states have been excluded should attention be
trates. For major surgery, plasma FVII levels of at least 20% are sufficient directed to specific protein defects influencing PT and PTT.
(Bolton-­Maggs et al., 2004).

Disorders with Normal APTT and PT DISSEMINATED INTRAVASCULAR COAGULATION


FXIII Deficiency (DIC)
Factor XIII stabilizes and cross-­links fibrin (Hsieh & Nugent, 2008). Fac- DIC is a clinicopathologic syndrome in which activation of the coagulation
tor XIII deficiency can determine delayed bleeding, usually 24 to 36 hours and fibrinolysis systems results in the simultaneous formation of thrombin
after surgery or trauma; spontaneous bleeding also occurs. Coagulation and plasmin with the consumption of coagulation factors and inhibitors of

843
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
TABLE 40.6
LIVER DISEASE
ISTH Diagnostic Scoring System for DIC
Hepatocytes synthesize all coagulation factors except for FVIII and vWF.
Risk assessment: Does the patient have an underlying disorder known to Therefore, both acute and chronic liver diseases affect laboratory coagula-
be associated with overt DIC? tion parameters. Chronic liver disease (cirrhosis) is increasing worldwide
• If yes, proceed. due to alcohol, hepatitis C, and other causes. Acute fulminant liver disease
• If not, do not use this algorithm. is often associated with drugs, viral infection, sepsis, and more.
Order global coagulation tests (PT, platelet count, fibrinogen, Cirrhosis
D-­dimer)
Patients with cirrhosis often have prolonged PT and mild to moderate
Score the test results:
thrombocytopenia. Cirrhotic patients have been shown to have rebalanced
• Platelet count (>100 k/μL = 0, <100 k/μL = 1, <50 k/μL = 2) hemostasis, in which, despite abnormal coagulation parameters, they have
• Elevated D-­dimer (<0.4 μg/mL = 0, 0.4–4.0 μg/mL = 2, >4.0 μg/mL = 3) normal hemostasis with no excessive bleeding tendency (Tripodi & Man-
• Prolonged PT (<3 sec = 0, >3 sec but <6 sec = 1, >6 sec = 2) nucci, 2011). Large epidemiologic studies have demonstrated that cirrhot-
• Fibrinogen level (>100 mg/dL = 0, <100 mg/dL = 1)
ics have double the incidence of DVT/PE and portal vein thrombosis than
Calculate score: the general population (Ambrosino et al., 2017; Mancusio, 2016). Most
• If ≥5, compatible with overt DIC: Repeat score daily. bleeding in cirrhosis is associated with increased portal pressure caus-
• If <5, suggestive (not affirmative) for nonovert DIC: Repeat next 1–2 ing variceal bleeding—that is, surgical rather than coagulopathic bleed-
days. ing—that responds to surgical intervention and not to blood component
transfusion to correct laboratory values. Blood components increase portal
DIC, Disseminated intravascular coagulation; ISTH, International Society on Throm-
bosis and Haemostasis; PT, prothrombin time. pressure further, leading to worsening of variceal bleeding and harm to
Note that in this example of score implementation, a D-­dimer test having an upper the patients.
normal limit of 0.4 μg/mL was employed. More generally, for the particular analyte
employed as an “elevated fibrin marker,” no increase = 0, moderate increase = 2, Rebalanced Primary Hemostasis
and strong increase = 3. Cirrhotics have mild to moderate thrombocytopenia, which is multifac-
torial, including splenomegaly, decreased thrombopoietin (TPO), bone
marrow suppression due to nutritional deficiency, or infection. However,
the system. There are a few scoring systems for DIC diagnosis; however, there are extremely high levels of vWF with increasing severity of cirrhosis
they often are not used in clinical practice, probably due to poor sensitivity from Child-­Pugh stage A to C with mean vWF being two-­to fourfold
and specificity as well as reproducibility in daily practice (Iba et al., 2019, that of normal (300%–600%). This increased vWF improves the adhesive
Yamazaki et al., 2019) (Table 40.6). function of platelets manyfold and, thus, normalizes primary hemostasis.
These changes result in the clinical features of usually widespread A randomized clinical trial using eltrombopag (a TPO mimetic) was dis-
bleeding (especially from puncture sites, wounds, and surgical sites) continued due to higher portal vein thrombosis in the treatment arm com-
and laboratory findings of prolonged PT, APTT, decreased fibrinogen, pared with placebo, suggesting that cirrhotics do not need platelet counts
elevated D-­dimers, and thrombocytopenia. The peripheral blood smear >50 K for surgical hemostasis (Afdhal et al., 2012).
examination may show microangiopathic hemolytic anemia (MHA) with Also, platelet transfusions often fail to increase platelet count due to
schistocytes and helmet cells in both acute and chronic DIC. However, sequestration in the enlarged spleen within minutes of the transfusion.
the absence of MHA does not exclude DIC. DIC can be an acute, life-­ Multiple platelet transfusions are detrimental due to the presence of ∼300
threatening, or chronic clinical condition depending on the underlying cc of plasma in each dose of platelet that further increases portal pressure;
cause. Identifying DIC and the underlying condition responsible for it are the resulting bleeding is often attributed to thrombocytopenia instead.
critical to adequate management.
DIC can occur in patients with sepsis, malignancy, obstetric complica- Rebalanced Secondary Hemostasis
tions, or massive tissue injury (Schmaier, 1991). DIC can also arise during The liver makes both procoagulants and natural anticoagulants. However,
surgery as a result of the release of thromboplastin material. In circula- routine coagulation tests, PT/INR, and APTT reflect only decreased pro-
tory arrest operations on the arch of the aorta or the main pulmonary coagulants and not the decreased natural anticoagulants (AT, PC, and PS).
arteries, DIC is a frequent complication related to hypothermia induced The cirrhotic plasma in the presence of thrombomodulin generates the
in the patient and tissue destruction. Abruptio placentae, placenta previa, same amount of thrombin as normal plasma despite an INR >3, reflecting
amniotic fluid embolism, and HELLP syndrome (hemolysis, elevated liver rebalanced secondary hemostasis (Tripodi et al., 2005). The viscoelastic
enzymes, and low platelets) are associated with acute hemorrhagic DIC, measurements with TEG/ROTEM often show normal reaction time/clot-
whereas the retained dead fetus is associated with a DIC that is not hemor- ting time (R/CT) values despite very long PT/INR (De Pietri et al., 2016).
rhagic but rather prothrombotic. DIC with sepsis is most commonly seen TEG/ROTEM should not be used routinely other than for R/CT param-
with gram-­negative infection but can occur with gram-­positive infection eters because both MA and MCF are usually lower than normal due to
and in the immunosuppressed patient with fungemia. thrombocytopenia. Primary hemostasis is rebalanced in-­vivo and does not
This clinical laboratory phenotype of DIC most probably results from reflect in MA/MCF due to absence of endothelium in the system.
a hyperfibrinolytic state and is the form of DIC most commonly recog- Thus, it is futile to attempt correction of mild to moderately prolonged
nized. A prothrombotic state as the result of DIC is usually associated with PT/INR (1.5–2.5). If the PT/INR is prolonged >2.5, it could be due to
normal PT, normal to shortened APTT due to high FVIII with mildly vitamin K deficiency and/or hypo/dysfibrinogenemia, which is observed in
reduced platelet count and normal or elevated fibrinogen (acute-­phase up to 30% to 45% of cirrhotics. Many patients with cirrhosis have fibrino-
effect, like FVIII). The D-­dimers are elevated. gen <100 mg/dL; hence, fibrinogen should be routinely measured if the
patient is scheduled for a high-­risk procedure or has nonvariceal bleeding.
Management of DIC Hypo/dysfibrinogenemia should be treated with cryoprecipitate.
The treatment of the underlying cause often results in the resolution of
DIC. The practice of using plasma, platelets, or cryoprecipitate to cor- Rebalanced Fibrinolytic System
rect an abnormal coagulation test parameter in a nonbleeding patient Plasminogen and α2-­antiplasmin are reduced, but endothelial-­derived tPA
can result in worsening of the clinical condition due to exacerbation of and PAI-­I are increased; therefore, fibrinolysis is also rebalanced. Usually,
microthromboses as a result of the “adding fuel to the fire” effect. There- patients with end-­stage cirrhosis may exhibit hyperfibrinolysis with large
fore, in a nonbleeding patient with only laboratory evidence of DIC, a ecchymoses and severe hypofibrinogenemia. These patients may benefit
low-­dose heparin (3–5 IU/kg/hour) may help to neutralize thrombin from antifibrinolytics along with cryoprecipitate.
and, thus, downregulate the DIC process. This effect is often observed
as improved platelet count and fibrinogen value, and gradual improve- VITAMIN K DEFICIENCY
ment of PT and APTT—of course, treatment of the underlying cause
is imperative. In a patient with acute DIC and bleeding, again, control Vitamin K, a lipid-­ soluble vitamin, is provided by dietary intake of
of the underlying cause is essential, along with appropriate use of blood leafy green vegetables and by the synthesis of intestinal flora. In clinical
components, including plasma to correct coagulopathy, platelet transfu- practice, vitamin K deficiency is seen most often in acutely ill patients
sion to maintain platelet count >20 × 109/L and cryoprecipitate to main- on antibiotics who have subsisted on parenteral nutrition. Not infre-
tain fibrinogen >100 mg/dL. quently, intravenous fluids are not supplemented with vitamin K. After

844
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
a few weeks of parenteral nutrition and antibiotic treatment, the patient tissue), hematuria, and, rarely, joint bleed. These patients have a pro-
becomes vitamin K deficient, which is reflected in the gradual increase longed APTT with normal PT. In the right clinical setting when there
in PT/INR from the normal at baseline. Vitamin K deficiency can also is significant bleeding and the patient has previously normal APTT, a
be seen in patients who have an anatomic bypass of the small intestine, stat FVIII level, especially by the chromogenic method, would establish
malabsorption, biliary tract obstruction, and, rarely, reduced dietary the diagnosis. If there is no access to stat FVIII assay, then a two-­step
intake. For example, alcoholics are often vitamin K deficient. Warfarin mixing may be performed to show time-­and temperature-­dependent

PART 5
also inhibits the enzymatic pathway necessary for vitamin K utilization inhibitor. FVIII levels are often reduced to single digits or in the teens
(see Fig. 43.1). Vitamin K has a critical role in the γ-­carboxylation reac- despite a high-­titer inhibitor, suggesting that the assay is probably
tion of several glutamic acid residues within coagulation factors II, VII, detecting dissociated FVIII from its IgG inhibitor in vitro. Hemor-
IX, and X and proteins C, S, and Z. This γ-­carboxylation is critical for the rhage in these patients is treated with a bypassing agent such as FEIBA
proteins to bind to cells and phospholipids so that they can participate or rFVIIa. Recently, emicizumab and recombinant porcine FVIII have
in physiologic coagulation reactions. Thus, patients will have reduced been other options. AHA generally responds well to immunosuppres-
vitamin K–dependent factors II, VII, IX, and X. If antigen levels of these sion therapy with corticosteroids (1 mg/kg), cyclophosphamide, and
patients are measured, they will frequently be higher. All such clinical rituximab (both low, 100-­mg weekly × 4 fixed-­dose as well as lymphoma
scenarios should be treated with 10 mg intravenous vitamin K in 25 to 50 dose, 375 mg/m2 × 4 weeks) (Aggarwal, 2005). The response to therapy
mL of normal saline slowly infused over 15 to 30 minutes. This approach can be monitored with improvement in APTT initially and then FVIII
is both diagnostic and therapeutic, avoiding unnecessary investigations levels, and inhibitor titer twice a month.
of prolonged PT/INR. The intravenous route is preferred for its rapid FX deficiency is rarely encountered in patients with amyloidosis, in
onset of action, within 2 to 4 hours, as opposed to oral and subcutaneous whom the amyloid fibrils selectively adsorb FX from the plasma (Chan &
routes that have unpredictable absorption. Ogunsile, 2017). In such cases, patients present with bleeding tendencies
that are associated with prolonged PT and APTT with normal thrombin
time. FX is usually moderate to severely deficient (<5% activity). Plasma
MASSIVE TRANSFUSION infusion, plasma exchange, and nonactivated PCC have been often ineffec-
Massive transfusion is defined as the replacement of more than 1.0 blood tive because, within minutes of infusion, the amyloid fibrils rapidly adsorb
volume or at least 10 units of packed red blood cells in 24 hours, usually out FX. The best option is to use FEIBA to treat bleeding.
following major trauma. Trauma-­induced coagulopathy (TIC) is unique Recombinant human thrombin has mostly replaced bovine thrombin
and still not well understood (Cohen et al., 2017). TIC is not merely in clinical practice. However, rarely upon exposure to bovine thrombin,
either dilutional coagulopathy due to infusion of liters of colloids/crys- a patient may make an antibody against either bovine thrombin or bovine
talloids or red cell transfusions during the transfer from the trauma FV that is present as a contaminant in bovine thrombin (Gaava et al.,
site to the emergency department (ED) of the hospital nor just mas- 2016). These antibodies may cross-­react with human thrombin or human
sive DIC initiated by the tissue factor released after trauma. TIC exists FV, causing prolonged PT and APTT. Thrombin time is also prolonged
in addition to these and may involve endothelial perturbation affecting with an antibody against bovine thrombin but not with an antibody against
thrombomodulin and activated protein C pathway. The coagulopathy FV. A mixing study will support a diagnosis of inhibitor due to incomplete
is made worse by hypothermia and acidosis. Therefore, recently, the to no correction of prolonged PT/APTT/TT. Most of these patients do
use of liquid plasma and low titer O whole blood at the trauma site is not bleed; however, if there is bleeding, they should be treated with corti-
undergoing clinical trials to improve hemostasis and prevent the onset of costeroids that often eliminate the inhibitor(s). These inhibitors are often
coagulopathy. Once the coagulopathy sets in, then it is difficult to catch transient following exposure to some antibiotics. In severe bleeding cases,
up with when the patient arrives in the ED. Massive transfusion proto- FEIBA or rFVIIa can be used.
cols (MTPs) have been developed to manage such patients by transfus-
ing balanced amounts of various blood components. ROTEM is also Lupus Anticoagulant
used as an algorithmic approach to provide goal-­directed transfusion This circulating inhibitor was initially identified in patients with sys-
therapy because of the real-­time tracings displayed in the OR. Plasma temic lupus erythematosus (SLE); because it prolonged APTT similarly
transfusion is indicated when EXTEM CT is prolonged, or transfusion to heparin, it was called lupus anticoagulant (LA). It was initially thought
of cryoprecipitate if the FIBTEM shows a low value along with low to interact with phospholipids in the coagulation cascade during APTT
EXTEM MCF, or transfusion of platelets if low EXTEM MCF is asso- testing in the test tube. It is triply misnomered because most LAs are (1)
ciated with normal FIBTEM. In places where ROTEM is unavailable, found in non-­SLE patients, (2) are prothrombotic, and (3) interact with
MTPs consisting of a red cell:plasma:platelet ratio of 1:1:1 or 2:1:1 are PL bound to a protein moiety such as prothrombin or β2-­glycoprotein
used. Although surgical mishap resulting in massive transfusions during I. Between 1960 and 1980, there was no specific diagnostic test for LA.
nontrauma surgery may occur, coagulopathy in such a patient is more Hence, a mixing study was used to differentiate between factor deficiency
controlled than trauma. Often, goal-­directed transfusion therapy with and LA as a cause of prolonged APTT. Recently, more specific tests—
ROTEM guidance is useful in these circumstances. However, postpar- such as the dilute Russell viper venom test (DRVVT), silica clotting time,
tum hemorrhage seems to require a trauma level transfusion strategy, and PTT-­LA—are available to detect LA (see Chapter 42). Hence, they
especially regarding fibrinogen levels. should be used to identify the cause of prolonged APTT in a nonbleeding
or thrombotic patient because a weak LA with mildly prolonged APTT
will show complete correction on mixing study, leading to unnecessary
ACQUIRED COAGULATION FACTOR DEFICIENCIES investigation of factor deficiency in the intrinsic pathway (Pengo, 2009).
Rarely, spontaneous autoantibody against coagulation protein can be The sensitivity of each APTT reagent for LA depends on its PL con-
formed and may present with a bleeding tendency (Kruse-­Jarres et al., tent. Some patients with a strong LA may also have prolonged PT/INR
2017). The most common of these rare acquired bleeding disorders that should be recognized at the beginning of VKA therapy because such
is acquired hemophilia A (AHA) due to an autoantibody formation patients should be monitored with chromogenic FX rather than INR for
against FVIII. The inhibitor is an IgG that binds to FVIII and causes proper anticoagulation therapy.
its clearance or inhibits its function, resulting in a severe bleeding ten- Rarely, patients with a very strong LA may also have an autoantibody
dency that, if not diagnosed promptly and treated, can be fatal. AHA is against prothrombin, causing very prolonged PT and APTT. These
encountered in two peaks—one in young women during the postpartum patients with “lupus anticoagulant and hypoprothrombinemia syndrome”
period and the second in the elderly (6th–8th decades). Some patients generally have severe prothrombin deficiency, and present with significant
may have an underlying autoimmune disorder; otherwise, it is seen as a clinical bleeding rather than clotting despite the presence of LA. Most
primary disorder. Both males and females are affected and present with of these patients are children, who respond well to corticosteroids. The
significant soft-­tissue hemorrhage (usually in areas with much adipose bleeding should be managed with PCC.

845
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
SELECTED REFERENCES
CHAPTER 40 COAGULATION AND FIBRINOLYSIS
Adcock DM, Kressin DC, Marlar RA: Minimum speci- Marder VJ, Budzynski AZ, Barlow GH: Comparison of Shariat-­Madar Z, Mahdi F, Schmaier AH: Identification
men volume requirements for routine coagulation the physio-­chemical properties of fragment D deriva- and characterization of prolylcarboxypeptidase as an
testing: dependence on citrate concentration, Am J tives of fibrinogen and fragment D-­D of cross-­linked endothelial cell prekallikrein activator, J Biol Chem
Clin Pathol 109:595–599, 1998. fibrin, Biochim Biophys Acta 427:1–14, 1976. 277:17962–17969, 2002.
A seminal paper that describes the importance of an appropri- Classic paper describing the D-­dimer. Paper that identifies a physiologic activator of the plasma kal-
ate volume of blood to anticoagulant ratio to avoid fallacies Mahlangu J, Oldenburg J, Paz-­Priel I, et al.: Emicizumab likrein/kinin system, the so-­called contact activation system.
of testing. prophylaxis in patients who have hemophilia A with- Tripodi A, Mannucci PM: The coagulopathy of chronic
Cohen MJ, Christie SA: Coagulopathy of trauma, Crit out inhibitors, N Engl J Med 379(9):811–822, 2018. liver disease, N Engl J Med 365:147–156, 2011.
Care Clin 33(1):101–118, 2017. Article describing an important advancement in the manage- An excellent paper describing rebalanced hemostasis in cir-
An important paper that describes the unique coagulopathy ment of hemophila A. rhosis.
of trauma. Pengo V, Tripodi A, Reber G, et al.: Update of the Yates SG, Fitts E, De Simone N, Sarode R: Prolonged
Gailiani D, Broze G: Factor XI activation in a revised guidelines for lupus anticoagulant detection. Subcom- activated partial yhromboplastin time: to mix or not to
model of blood coagulation, Science 253:909–912, mittee on lupus anticoagulant/Antiphospholipid anti- mix—is that the question? Transfus Apher Sci 58(1):39–
1991. body of the Scientific and Standardisation Committee 42, 2019.
Classic paper that presents for the first time the modern view of the international society on thrombosis and haemo- An important paper describing fallacies of routine mixing
of assembly and interaction of proteins of the coagulation stasis, J Thromb Haemost 7:1737, 2009. studies for prolonged activated partial thromboplastin time.
system. An important paper on diagnostic criteria for lupus antico-
Access the complete reference list online at Elsevier
Iba T, Levy JH, Yamakawa K, et al.: Scientific and agulant.
eBooks for Practicing Clinicians.
Standardization Committee on DIC of the inter- Proctor RR, Rapaport SI: The partial thromboplastin
national society on thrombosis and haemostasis, J time with kaolin: a simple screening test for first stage
Thromb Haemost 17(8):1265–1268, 2019. plasma clotting factor deficiencies, Am J Clin Pathol
An important paper describing the ISTH recommendations 36:212–219, 1961.
on DIC. Description of activated partial thromboplastin time.

846
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
REFERENCES Cui J, O’Shea KS, Purkayastha A, et al.: Fatal haemorrhage and incomplete block to
Adcock DM, Kressin DC, Marlar RA: Minimum specimen volume requirements for embryogenesis in mice lacking coagulation factor V, Nature 384:66–68, 1996.
routine coagulation testing: dependence on citrate concentration, Am J Clin Pathol Del Zoppo GJ: Bleeding in the brain: amyloid-­β may keep clots away, Nat Med 15:1132–
109:595–599, 1998. 1133, 2009.
Afdhal NH, Giannini EG, Tayyab G, et al.: Eltrombopag before procedures in patients de Moerloose P, Casini A, Neerman-­Arbez M: Congenital fibrinogen disorders: an up-
with cirrhosis and thrombocytopenia, N Engl J Med 23;367(8):716–24, 2012. date, Semin Thromb Hemost 39(6):585–595, 2013.
Aggarwal A, Grewal R, Green RJ, et al.: Rituximab for autoimmune haemophilia: a De Pietri L, Bianchini M, Montalti R, et al.: Thrombelastography-­guided blood product
proposed treatment algorithm, Haemophilia 11:13–19, 2005. use before invasive procedures in cirrhosis with severe coagulopathy: a randomized,
Agren A, Wikman AT, Holmstrom M, et al.: Thromboelastography (TEG(R)) com- Controlled trial.Hepatology 63(2):566–73, 2016.
pared to conventional coagulation tests in surgical patients—a laboratory evaluation, Dewerchin M, Liang Z, Moons L, et al.: Blood coagulation factor X deficiency caus-
Scand J Clin Lab Invest 73:214–220, 2013. es partial embryonic lethality and fatal neonatal bleeding in mice, Thromb Haemost
Andrew M, Vegh P, Johnston M, et al.: Maturation of the hemostatic system during 83:185–190, 2000.
childhood, Blood 80:1998–2005, 1992. Doolittle RF: Fibrinogen and fibrin, Sci Am 245:126–135, 1981.
Ambrosino P, Tarantino L, Di Minno G, et al.: The risk of venous thromboembo- Dorey E: First recombinant factor XIII approved, Nat Biotechnol 32(3):210, 2014.
lism in patients with cirrhosis, A systematic review and meta-­analysis Thromb Haemost. Draxler DF, Medcalf RL: The fibrinolytic system—more than fibrinolysis? Transfus
5 117(1):139–148, 2017. Med Rev 29(2):102–109, 2015.
Anwar R, Miloszewski KJ: Factor XIII deficiency, Br J Haematol 107:468–484, 1999. Dupuy E, Soria C, Molho P, et al.: Embolized ischemic lesions of toes in an afibrino-
Aoki N, Sakata Y, Matsuda M, et al.: Fibrinolytic states in a patient with congenital genemic patient: possible relevance to in vivo circulating thrombin, Thromb Res
deficiency of alpha 2-­plasmin inhibitor, Blood 55:483–488, 1980. 102:211–219, 2001.
Ardillon L, Lefrançois A, Graveleau J, et al.: Management of bleeding in severe factor V Eckhardt CL, van Velzen AS, Peters M, et al.: Factor VIII gene (F8) mutation and risk of
deficiency with a factor V inhibitor, Vox Sang 107(1):97–99, 2014. inhibitor development in nonsevere hemophilia A, Blood 122:1954–1962, 2013.
Arun B, Kessler CM: Clinical manifestations and therapy of the hemophilias. In Colman Emsley J, Mcewan PA, Gailani D: Structure and function of factor XI, Blood 115:2569–
RW, Hirsh J, Marder VJ, et al.: Hemostasis and thrombosis: basic principles and clinical 2577, 2010.
practice, 4, Philadelphia, 2001, Lippincott-­Raven. Esmon CT: The endothelial protein C receptor, Curr Opin Hematol 13:382, 2006.
Asakai R, Chung DW, Davie EW, et al.: Factor XI deficiency in Ashkenazi Jews in Esmon CT: Endothelial protein C receptor, Thromb Haemost 82:251–258, 1999.
Israel, N Engl J Med 325:153–158, 1991. Esmon CT, Owen WG: Identification of an endothelial cell cofactor for thrombin—
Barg AA, Rosenberg N, Budnik I, et al.: Prospective controlled monitoring of Hemlibra Catalytic activation of protein C, Proc Natl Acad Sci U S A 78:2249–2252, 1981.
prophylaxis initiation in a large Cohort of hemophilia a patients:real-­world data, Blood European Medicines Agency (EMA): Workshop report: Characterisation of new clotting fac-
134, 2019. tor concentrates (FVIII, FIX) with respect to potency assays used for labelling and testing
Barnathan ES, Kuo A, van der Keyl H, et al.: Binding of tissue type plasminogen activa- of post infusion samples. Human Medicines Development and Evaluation, 26 June 2014.
tor to human endothelial cells: evidence for two distinct binding sites, J Biol Chem EMA/135928/2014.
263:7792–7799, 1988. Fay WP, Shapiro AD, Shih JL, et al.: Brief report: complete deficiency of plasminogen-­
Batty P, Lillicrap D: Advances and challenges for hemophilia gene therapy, Hum Mol activator inhibitor type 1 due to a frame-­shift mutation, N Engl J Med 327:1729–1733,
Genet 28(Issue R1):R95–R101, 2019. 1992.
Bauduer F, de Raucourt E, Boyer-­Neumann C, et al.: French Postmarketing Study Fickenscher K, Aab A, Stuber W: A photometric assay for blood coagulation factor XIII,
Group. Factor XI replacement for inherited factor XI deficiency in routine clinical Thromb Haemost 65:535–540, 1991.
practice: results of the HEMOLEVEN prospective 3-­year postmarketing study, Hae- Finigan JH, Dudek SM, Singleton PA, et al.: Activated protein C mediates novel lung
mophilia 21(4):481–489, 2015. endothelial barrier enhancement: role of sphingosine 1-­phosphate receptor transacti-
Blanchette VS, Key NS, Ljung LR, et al.: Definitions in hemophilia: Communication vation, J Biol Chem 280:17286–17293, 2005.
from the SSC of the ISTH, J Thromb Haemost 12(11):1935–1939, 2014. Fried K, Kaufman S: Congenital afibrinogenemia in 10 offspring of uncle–niece mar-
Board PG, Losowsky MS, Miloszewski KJ, et al.: Inherited and acquired deficiency, riages, Clin Genet 17:223–227, 1980.
Blood Rev 7:229–242, 1993. Funk C, Gmür J, Herold R, et al.: Reptilase-­R—a new reagent in blood coagulation, Br
Bockenstedt PL: Laboratory methods in hemostasis. In Loscalzo J, Schafer AI, editors: J Haematol 21:43–52, 1971.
Thrombosis and hemorrhage, 3, Philadelphia, 2003, Lippincott Williams & Wilkins. Furie B: Pathogenesis of thrombosis, Hematology Am Soc Hematol Educ Program255–258,
Boggio L, Green D: Recombinant human factor VIIa in the management of amyloid-­ 2009.
associated factor X deficiency, Br J Haematol 112:1074–1075, 2001. Furie B, Furie BC: Thrombus formation in vivo, J Clin Invest 115:3355–3362, 2005.
Bolton-­Maggs PH, Patterson DA, Wensley RT, et al.: Definition of the bleeding ten- Gailani D1, Broze Jr GJ: Factor XI activation in a revised model of blood, Coagulation
dency in factor XI–deficient kindreds—a clinical and laboratory study, Thromb Hae- 253(5022):909–912, 1991.
most 73:194–202, 1995. Girolami MT, Hofer CK: Coagulation monitoring: current techniques and clinical use
Bolton-­Maggs PH, Perry DJ, Chalmers EA, et al.: The rare coagulation disorders—re- of viscoelastic point-­of-­care coagulation devices, Anesth Analg 106:1366–1375, 2008.
view with guidelines for management from the United Kingdom Haemophilia Centre Gawryl MS, Hoyer LW: Inactivation of factor VIII coagulant activity by two different
Doctors’ Organisation, Haemophilia 10:593–628, 2004. types of human antibodies, Blood 60:1103–1109, 1982.
Bolton-­Maggs PH, Young Wan-­Yin B, McCraw AH, et al.: Inheritance and bleeding in Gavva C, Yates SG, Rambally S: Transfusion management of factor V deficiency: three
factor XI deficiency, Br J Haematol 69:521–528, 1988. case reports and review of the literature, Transfusion 56(7):1745–1749, 2016.
Bouchard BA, Chapin J, Brummel-­Ziedins KE, et al.: Platelets and platelet-­derived Girolami A, Simioni P, Scarano L, et al.: Hemorrhagic and thrombotic disorders due to
factor Va confer hemostatic competence in complete factor V deficiency, Blood factor V deficiencies and abnormalities: an updated classification, Blood Rev 12:5–51,
125(23):3647–3650, 2015. 1998.
Brenner B, Laor A, Lupo H, et al.: Bleeding predictors in factor-­XI–deficient patients, Girolami A, Vidal J, Salagh M, et al.: The old and the new in prekallikrein deficiency:
Blood Coagul Fibrinolysis 8:511–515, 1997. Historical context and a family from Argentina with PK deficiency due to a new muta-
Brenner B, Sanchez-­Vega B, Wu SM, et al.: A missense mutation in gamma-­glutamyl tion (Arg541Gln) in exon 14 associated with a common polymorphysm (Asn124Ser)
carboxylase gene causes combined deficiency of all vitamin K–dependent blood co- in exon 5, Semin Thromb Hemost 40(5):592–599, 2014.
agulation factors, Blood 92:4554–4559, 1998. Goodwin TM: Congenital hypofibrinogenemia in pregnancy, Obstet Gynecol Surv
Büller HR, Bethune C, Bhanot S, et al.: Factor XI antisense oligonucleotide for preven- 44:157–161, 1989.
tion of venous thrombosis, N Engl J Med 372:232–240, 2015. Graham JB: Genotype assignment (carrier detection) in the haemophilias, Clin Haematol
Burstyn-­Cohen T, Heeb MJ, Lemke G: Lack of protein S in mice causes embryonic 8:115–145, 1979.
lethal coagulopathy and vascular dysgenesis, J Clin Invest 119:2942–2953 Greenberg CS, Lai T-­S: Fibrin formation and stabilization. In Loscalzo J, Schafer AI,
Caplice NM, Panetta C, Peterson TE, et al.: Lipoprotein (a) binds and inactivates tis- editors: Thrombosis and hemorrhage, 3, Philadelphia, 2003, Lippincott Williams &
sue factor pathway inhibitor: a novel link between lipoproteins and thrombosis, Blood Wilkins.
98(10):2980–2987, 2001. Han ED, MacFarlane RC, Mulligan AN, et al.: Increased vascular permeability in C1
Cesarman GM, Guevara CA, Hajjar KA: An endothelial cell receptor for plasminogen/ inhibitor–deficient mice mediated by the bradykinin type 2 receptor, J Clin Invest
tissue plasminogen activator (t-­PA). II. Annexin II–mediated enhancement of t-­PA– 109:1057–1063, 2002.
dependent plasminogen activation, J Biol Chem 269:21198–21203, 1994. Hanss M, Biot F: A database for human fibrinogen variants, Ann N Y Acad Sci 936:89–90,
Chafa O, Chellali T, Sternberg C, et al.: Severe hypofibrinogenemia associated with bi- 2001.
lateral ischemic necrosis of toes and fingers, Blood Coagul Fibrinolysis 6:549–552, 1995. Hay CRM, Brown S, Collins PW, et al.: The diagnosis and management of factor VIII
Chan IS, Ogunsile FJ: An acquired factor X inhibitor: the importance of understanding and IX inhibitors: a guideline from the United Kingdom Haemophilia Center Doc-
coagulation, Am J Med 130(7):e307–e308, 2017. tors Organization, Br J Haematol 133:591–605, 2006.
Colman RW, Bagdasarian A, Talarnos RC, et al.: Williams trait: human kininogen de- Hayashi M, Matsushita T, Mackman N, et al.: Fatal thrombosis of antithrombin-­
ficiency with diminished levels of plasminogen proactivator and prekallikrein associ- deficient mice is rescued differently in the heart and liver by intercrossing with low
ated with abnormalities of the Hageman factor–dependent pathways, J Clin Invest tissue factor mice, J Thromb Haemost 4:177–185, 2006.
56:1650–1662, 1975. He L, Pappan LK, Grenache DG, et al.: The contributions of the alpha 2 beta 1 integrin
Cohen MJ, Christie SA: Coagulopathy of trauma, Crit Care Clin 33(1):101–118, 2017. to vascular thrombosis in vivo, Blood 102:3652–3657, 2003.
Coughlin SR: Thrombin signaling and protease-­activated receptors, Nature 407:258– Heemskerk JW, Mattheij NJ, Cosemans JM: Platelet-­based coagulation: different popu-
264, 2000. lations, different functions, J Thromb Haemost 11(1):2–16, 2013.
Cozzi MR, Guglielmini G, Battiston M, et al.: Visualization of nitric oxide production Hitomi K, Kitamura M, Perez Alea M, et al.: A specific colorimetric assay for measuring
by individual platelets during adhesion in flowing blood, Blood 125(4):697–705, 2015. transglutaminase 1 and factor XIII activities, Anal Biochem 394:281–283, 2009.
Crawley JTB, Lane DA: The haemostatic role of tissue factor pathway inhibitor, Arte- Hoffman M, Monroe DM: A cell-­ based model of hemostasis, Thromb Haemost
rioscler Thromb Vasc Biol 28:233–242, 2008. 85(6):958–965, 2001.
Crossley M, Ludwig M, Stowell KM, et al.: Recovery from hemophilia B Leyden: an Hong SL: Effect of bradykinin and thrombin on prostacyclin synthesis in endothelial
androgen-­responsive element in the factor IX promoter, Science 257:377–379, 1992. cells from calf and pig aorta and human umbilical cord vein, Thromb Res 18:787–795,
1980.

846.e1
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
Hsieh L, Nugent D: Factor XIII deficiency, Hemophilia 14:1190–1200, 2008. Marcum JA, McKenney JB, Rosenberg RD: Acceleration of thrombin-­antithrombin
REFERENCE
Huang Z-­F, Higuchi D, Lasky N, et al.: Tissue factor pathway inhibitor gene disruption complex formation in rat hindquarters via heparinlike molecules bound to endothe-
produces intrauterine lethality in mice, Blood 90:944–951, 1997. lium, J Clin Invest 74:341–350, 1984.
Iba T, Levy JH, Yamakawa K, et al.: Scientific and Standardization Committee on Marder VJ, Budzynski AZ: Degradation products of fibrinogen and cross-­linked fibrin-­
DIC of the international society on thrombosis and haemostasis, J Thromb Hae- projected clinical applications, Thromb Diath Haemorrh 32:49–56, 1974.
most(8)1265–1268, 2019. Mariani G, Bernardi F: Factor VII deficiency, Semin Thromb Hemost 35:400–406, 2009.
Ivanciu L, Krishnaswamy S, Camire RM: New insights into the spatiotemporal localiza- Martinelli RA, Scheraga HA: Steady state kinetic study of the bovine thrombin-­
tion of prothrombinase in vivo, Blood 124:1705–1714, 2014. fibrinogen interaction, Biochemistry 19:2343–2350, 1980.
Jalbert LR, Rosen ED, Moons L, et al.: Inactivation of the gene for anticoagulant Meijers JCM, Tekelenburg WL, Bouma BN, et al.: High levels of coagulation factor XI
protein C causes lethal perinatal consumptive coagulopathy in mice, J Clin Invest as a risk factor for venous thrombosis, N Engl J Med 342:696–701, 2000.
102:1481–1488, 1998. Merkoulov S, Komar AA, Schmaier AH, et al.: Deletion of the murine kininogen gene
Johansson PI, Stissing T, Bochsen L, et al.: Thrombelastography and tromboelastom- 1 (mKng1) causes loss of plasma kininogen and delays thrombosis, Blood 111:1274–
etry in assessing coagulopathy in trauma, Scand J Trauma Resusc Emerg Med 17:45, 1281, 2008.
2009. Miesbach W, Scharrer I, Henschen A, et al.: Inherited dysfibrinogenemia: clinical phe-
Kasper CK, Aledort L, Counts RB, et al.: A more uniform measurement of factor VIII notypes associated with five different fibrinogen structure defects, Blood Coagul Fi-
inhibitors, Thromb Diath Haemorrh 34:869–872, 1975. brinolysis 21:35–40, 2010.
Kaufman RJ, Antonarakis SE, Fay PJ, et al.: Factor VIII and hemophilia A. In Colman Miller G, Silberberg M, Kaplan AP: Autoactivability of human Hageman factor, Biochem
RW, Hirsh J, Marder VJ, et al.: Hemostasis and thrombosis, 4, Philadelphia, 2001, Lip- Biophys Res Commun 92:803–810, 1980.
pincott Williams & Wilkins, pp 135–156. Mitchell JA, Ali F, Bailey L, et al.: Role of nitric oxide and prostacyclin as vasoactive
Kenet G, Lubetsky A, Luboshitz J, et al.: Lower doses of rFVIIa therapy are safe and ef- hormones released by the endothelium, Exp Physiol 93(1):141–147, 2008.
fective for surgical interventions in patients with severe FXI deficiency and inhibitors, Mumford AD, Ackroyd S, Alikhan R, et al.: Guideline for the diagnosis and manage-
Haemophilia(5)1065–1073, 2009. ment of the rare coagulation disorders: a United Kingdom haemophilia centre Doc-
Kessler CM, Hoyer L, Feinstein DI: The hemophilias. In McArthur JR, Schechter GP, tors’ Organization guideline on behalf of the British Committee for standards in Hae-
editors: Hematology: the educational program of the American society of Hematology, Wash- matology, Br J Haematol 167(3):304–326, 2014.
ington, DC, 1996, American Society of Hematology, pp 95–105. Nagoya A, Kaneko M, Kazama, et al.: Discrepancies between the one-­stage clotting
Kitchen S, Malia RG, Preston FE: A comparison of methods for the measurement of assay and chromogenic assay in patients with hemophilia A receiving standard or
activated factor VII, Thromb Haemost 68:301–305, 1992. extended half-­life factor VIII products in clinical settings, Thromb Res 185:150–152,
Kobayashi T, Kanayama N, Tokunaga N, et al.: Prenatal and peripartum management 2019.
of congenital afibrinogenaemia, Br J Haematol 109:364–366, 2000. Napolitano M, Mariani G, Lapecorella M: Hereditary combined deficiency of the vita-
Kohler HP, Ariens RA, Whitaker P, et al.: A common coding polymorphism in the min K–dependent clotting factors, Orphanet J Rare Dis 5:21, 2010.
FXIII α-­subunit gene (FXIIIVal34Leu) affects cross-­linking activity, Thromb Haemost Nawarawong W, Wyshock E, Meloni FJ, et al.: The rate of fibrinopeptide B release
80:704, 1998. modulates the rate of clot formation: a study with an acquired inhibitor to fibrinopep-
Kruse-­Jarres R, Kempton CL, Baudo F, et al.: Acquired hemophilia A: updated review of tide B release, Br J Haematol 79:296–301, 1991.
evidence and treatment guidance, Am J Hematol 92(7):695–705, 2017. Naylor JA, Green PM, Rizza CR, et al.: Factor VIII gene explains all cases of haemo-
Labarque V, Perinparajah V, Bouskill V, et al.: Utility of factor VIII and factor VIII to philia A, Lancet 340:1066–1067, 1992.
von Willebrand factor ratio in identifying 277 unselected carriers of hemophilia A, Nichols WC, Seligsohn U, Zivelin A, et al.: Mutations in the ER-­Golgi intermediate
Am J Hematol 92:E94–E96, 2017. compartment protein ERGIC-­53 cause combined deficiency of coagulation factors V
Lak M, Keihani M, Elahi F, et al.: Bleeding and thrombosis in 55 patients with inherited and VIII, Cell 93:61–70, 1998.
afibrinogenaemia, Br J Haematol 107:204–206, 1999. O’Connell NM, Riddell AF, Pascoe G, et al.: Recombinant factor VIIa to prevent surgi-
Lakich D, Kazazian H, Antonarakis SE, et al.: Inversions disrupting the factor VIII gene cal bleeding in factor XI deficiency, Haemophilia 14(4):775–781, 2008.
are a common cause of severe hemophilia A, Nat Genet 5:236–241, 1993. Oldenburg J, Mahlangu JN, Kim B, et al.: Emicizumab prophylaxis in hemophilia A
LaRusch GA, Mahdi F, Shariat-­Madar Z, et al.: Factor XII stimulates ERK1/2 and Akt with inhibitors, N Engl J Med 377(9):809–818, 2017.
through uPAR, integrins, and the EGFR to initiate angiogenesis, Blood 115:5111– Okajima K, Kohno I, Soe G, et al.: Direct evidence for systemic fibrinogenolysis in patients
5120, 2010. with acquired alpha-­2-­plasmin inhibitor deficiency, Am J Hematol 45:16–24, 1994.
Lawrie AS, Kitchen S, Purdy G, et al.: Assessment of actin FS and actin FSL sensitivity Osterud B, Rappaport SI: Activation of factor IX by the reaction product of tissue factor
to specific clotting factor deficiencies, Clin Lab Haematol 20:179–186, 1998. and factor VII, Proc Natl Acad Sci U S A 74:5260–5264, 1977.
Leiba H, Ramot B, Many A: Heredity and coagulation studies in ten families with factor Palmer RMJ, Ferrige AG, Moncada S: Nitric oxide release accounts for the biologic
XI (plasma thromboplastin antecedent) deficiency, Br J Haematol 11:654–665, 1965. activity of endothelium-­derived relaxing factor, Nature 327:524–526, 1987.
Leurs J, Hendriks D, Carboxypeptidase U, TAFla): A metallocarboxypeptidase with a Pavlova A: Oldenburg J: defining severity of hemophilia: more than factor levels, Semin
distinct role in haemostasis and a possible risk factor for thrombotic disease, Thromb Thromb Hemost 39(7):702–710, 2013.
Haemost 94:471–487, 2005. Pasi KJ, Rangarajan S, Georgiev P, et al.: Targeting of antithrombin in hemophilia A or
Liang Y, Fu Y, Qi R, et al.: Cartilage oligomeric matrix protein is a natural inhibitor of B with RNAi therapy, N Engl J Med 377(9):819–828, 2017.
thrombin, Blood 126:905–914, 2015. Pasi KJ, Rangarajan S, Mitchell N, et al.: Multiyear follow-­up of AAV5-­hFVIII-­SQ
Ling G, Kagdi H, Subel B, et al.: Safety and efficacy of factor XI (FXI) concentrate use gene therapy for hemophilia A, N Engl J Med 2 382(1):29–40, 2020.
in patients with FXI deficiency: a single-­centre experience of 19 years, Haemophilia Pengo V, Tripodi A, Reber G, et al.: Update of the guidelines for lupus anticoagulant
22(3):411–418, 2016. detection. Subcommittee on lupus anticoagulant/Antiphospholipid antibody of the
Ljung RCR: Prenatal diagnosis of haemophilia, Haemophilia 5:84–87, 1999. Scientific and Standardisation Committee of the international society on thrombosis
Lorand L, Losowsky MS, Miloszewski KJM: Human factor XIII: fibrin-­stabilizing fac- and haemostasis, J Thromb Haemost 7:1737, 2009.
tor. In Spaet TH, editor: Progress in hemostasis and thrombosis, New York, 1980, Grune Perry DJ: Antithrombin and its inherited deficienciestripodi, Blood Rev 8(1):37–55, 1994.
and Stratton, pp 245–249. Peyvandi F, Mannucci PM: Rare coagulation disorders, Thromb Haemost 82:1207–1214,
Luna-­Zaizar H, Esparza-­Flores MA, Lopez-­Guido B, et al.: Kinetics of factor VIII: C 1999.
inhibitors and treatment response in severe hemophilia patients, Int J Lab Hematol Peyvandi F, Mannucci PM, Asti D, et al.: Clinical manifestations in 28 Italian and Ira-
31:673–682, 2009. nian patients with severe factor VII deficiency, Haemophilia 3:242–246, 1997.
Maas C, Govers-­Riemslag JW, Bouma B, et al.: Misfolded proteins activate factor XII Peyvandi F, Mannucci PM, Lak M, et al.: Congenital factor X deficiency: spectrum of
in humans, leading to kallikrein formation without initiating coagulation, J Clin Invest bleeding symptoms in 32 Iranian patients, Br J Haematol 102:626–628, 1998a.
118:3208–3218, 2008. Peyvandi F, Tuddenham EG, Akhtari AM, et al.: Bleeding symptoms in 27 Iranian
MacFarland D: An enzyme cascade in the blood clotting mechanism, and its functions as patients with the combined deficiency of factor V and factor VIII, Br J Haematol
a biochemical amplifier, Nature 202:498–499, 1964. 100:773–776, 1998b.
Mahlangu J, Oldenburg J, Paz-­Priel I, et al.: Emicizumab prophylaxis in patients who Peyvandi F, Mannucci PM, Garagiola I, et al.: A randomized trial of factor VIII and
have hemophilia A without inhibitors, N Engl J Med 379(9):811–822, 2018. neutralizing antibodies in hemophilia A, N Engl J Med 374:2054–2064, 2016.
Mahdi F, Rehemtulla A, Van Nostrand WE, et al.: Protease nexin-­2/amyloid β-­protein Pike GN, Cumming AM, Hay CR, et al.: Sample conditions determine the ability of
precursor regulates factor VIIa and the factor VIIa–tissue factor complex, Thromb Res thrombin generation parameters to identify bleeding phenotype in FXI deficiency,
99:267–276, 2000. Blood 126(3):397–405, 2015.
Mahdi F, Van Nostrand WE, Schmaier AH: Protease nexin-­2/amyloid β-­protein pre- Pinsky DJ, Broekman MJ, Peschon JJ, et al.: Elucidation of the thromboregulatory role
cursor inhibits factor Xa in the prothrombinase complex, J Biol Chem 270:23468– of CD39/ectoapyrase in the ischemic brain, J Clin Invest 109:1031–1040, 2002.
23474, 1995. Plug I, Mauser-­Bunschoten EP, Bröcker-­Vriends AH, et al.: Bleeding in carriers of he-
Manco-­Johnson MJ, Abshire TC, Shapiro AD, et al.: Prophylaxis versus episodic treat- mophilia, Blood 108:52–56, 2006.
ment to prevent joint disease in boys with severe hemophilia, N Engl J Med 357:535– Ragni MV, Lewis JH, Spero JA, et al.: Factor VII deficiency, Am J Hematol 10:79–88,
544, 2007. 1981.
Mann KG: Thrombin formation, Chest 124:4S–10S, 2003. Rak J: New checkpoint of the coagulant phenotype, Blood 124:3511–3513, 2014.
Mancuso A: Management of portal vein thrombosis in cirrhosis: an update, Eur J Gas- Rand MD, Lock JB, van’t Veer C, et al.: Blood clotting in minimally altered whole
troenterol Hepatol 28(7):739–743, 2016. blood, Blood 88:3432–3445, 1996.
Mannucci PM, Duga S, Peyvandi F: Recessively inherited coagulation disorders, Blood Ratnoff OD, Davies E: Waterfall sequence for intrinsic blood coagulation, Science
104:1243–1252, 2004. 145:1310–1312, 1964.
Mannucci PM, Franchini M: Porcine recombinant factor VIII: an additional weapon to Reitsma PH: Genetic principles underlying disorders of procoagulation and anticoagu-
handle anti-­factor VIII antibodies, Blood Transfus 15(4):365–368, 2017. lation proteins. In Colman RW, Hirsh J, Marder VJ, et al.: Hemostasis and thrombosis:
Marder VJ, Budzynski AZ, Barlow GH: Comparison of the physicochemical proper- basic principles and clinical practice, 4, Philadelphia, 2001, Lippincott-­Raven.
ties of fragment D derivatives of fibrinogen and fragment D-­D of cross-­linked fibrin, Renne T, Pozgajova M, Gruner S, et al.: Defective thrombus formation in mice lacking
Biochim Biophys Acta 18;427(1):1–14, 1976. coagulation factor XII, J Exp Med 202:271–281, 2005.

846.e2
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
Riddell A, Abdul-­Kadir R, Pollard D, et al.: Monitoring low dose recombinant factor Tracy PB, Eide LL, Bowie EJ, et al.: Radioimmunoassay of factor V in human plasma

REFERENCE
VIIa therapy in patients with severe factor XI deficiency undergoing surgery, Thromb and platelets, Blood 60:59–63, 1982.
Haemost 106(3):521–527, 2011. Tripodi A, Mannucci PM: The coagulopathy of chronic liver disease, N Engl J Med
Rosen ED, Chan JCY, Idusogie E, et al.: Mice lacking factor VII develop normally but 365:147–156, 2011.
suffer fatal perinatal bleeding, Nature 390:290–294, 1997. Tripodi A, Salerno F, Chantarangkul V, et al.: Evidence of normal thrombin generation
Ruiz-­Saez A: Occurrence of thrombosis in rare bleeding disorders, Semin Thromb He- in cirrhosis despite abnormal conventional coagulation tests, Hepatology 41(3):553–
most 39:684–692, 2013. 558, 2005.
Saito H, Ratnoff OD, Waldmann R, et al.: Fitzgerald trait: deficiency of a hitherto Uprichard J, Perry DJ: Factor X deficiency, Blood Rev 16:97–110, 2002.
unrecognized agent, Fitzgerald factor, participating in surface mediated reactions of Van der Meijden PEJ, Munnix ICA, Auger JM, et al.: Dual role of collagen in factor
clotting, fibrinolysis, generation of kinins, and the property of diluted plasma enhanc- XII–dependent thrombus formation, Blood 114:881–890, 2009.
ing vascular permeability, J Clin Invest 55:1082–1089, 1975. Van Nostrand WE, Schmaier AH, Farrow JS, et al.: Protease nexin-­II (amyloid β-­
Salomon O, Zivelin A, Livnat T, et al.: Prevalence, causes, and characterization of factor protein precursor): a platelet α-­granule protein, Science 248:745–748, 1990.
XI inhibitors in patients with inherited factor XI deficiency, Blood 101:4783–4788, Van Vulpen LF, Schutgens RE, Coeleveld K, et al.: IL-­1β, in contrast to TNFα, is
2003. pivotal in blood-­induced cartilage damage and is a potential target for therapy, Blood
Schmaier AH: Disseminated intravascular coagulation—Pathogenesis and management, 126:2239–2246, 2015.
J Intens Care Med 6:209–228, 1991. Vatsyayan R, Kothari H, Mackman N, et al.: Inactivation of factor VIIa by antithrombin
Schmaier AH: The plasma kallikrein/kinin system counterbalances the renin angioten- in vitro, ex vivo and in vivo: role of tissue factor and endothelial cell protein C recep-
sin system, J Clin Invest 109:1007–1009, 2002. tor, PloS One 9(8), 2014. e103505.
Schmaier AH: The kallikrein/kinin and the renin angiotensin systems have a multi-­ Verbruggen B, Novakova I, Wessels H, et al.: The Nijmegen modification of the
layered interaction, Am J Physiol Reg Integr Comp Physiol 285:R1–R13, 2003. Bethesda assay for FVIII: C inhibitors—improved specificity and reliability, Thromb
Schmaier AH: The physiologic basis of assembly and activation of the plasma kallikrein/ Haemost 73:247–251, 1995.
kinin system, Thromb Haemost 91:1–3, 2004. Vercauteren E, Gils A, Declerck PJ: Thrombin activatable fibrinolysis inhibitor: a puta-
Schmaier AH, Dahl LD, Hasan AAK, et al.: Factor IXa inhibition by protease nexin-­2/ tive target to enhance fibrinolysis, Semin Thromb Hemost 39(4):365–372, 2013.
amyloid β-­protein precursor on phospholipid vesicles and cell membranes, Biochem- Wang Y: Ivanov I, Smith SA,D, Morrissey JH. Polyphosphate, Zn2+ and high molecular
istry 34:1171–1178, 1995. weight kininogen modulate individual reactions of the contact pathway of blood clot-
Schmaier AH, Dahl LD, Rozemuller AJM, et al.: Protease nexin-­2/amyloid β-­protein ting, J Thromb Haemost 17(12):2131–2140, 2019.
precursor: a tight-­binding inhibitor of coagulation factor IXa, J Clin Invest 92:2540– Weiss HJ: Sussman II, Hoyer LW: stabilization of factor VIII in plasma by the von
2545, 1993. Willebrand factor: studies on posttransfusion and dissociated factor VIII and in pa-
Schneppenheim R, Budde U, Krey S, et al.: Results of a screening for von Willebrand tients with von Willebrand’s disease, J Clin Invest 60:390–404, 1977.
disease type 2N in patients with suspected haemophilia A or von Willebrand disease Weuppers KD, Cochrane CG: Plasma prekallikrein: Isolation, characterization, and
type 1, Thromb Haemost 76:598–602, 1996. mechanism of action, J Exp Med 135:1–20, 1972.
Seligsohn U, Zivelin A, Zwang E: Combined factor V and factor VIII deficiency among Weyand AC, Pipe SW: New therapies for hemophilia, Blood 133:389–398, 2019.
non-­Ashkenazi Jews, N Engl J Med 307:1191–1195, 1982. Wiggins RC, Cochrane CC: The autoactivability of human Hageman factor, J Exp Med
Shapiro AD, Paola JD, Cohen A, et al.: The safety and efficacy of recombinant human 150:1122–1133, 1979.
blood coagulation factor IX in previously untreated patients with severe or moder- Xu F, Previti M, Nieman M, et al.: A βPP/APLP2 family of Kunitz serine protease
ately severe hemophilia B, Blood 105:518–525, 2005. inhibitors regulate cerebral thrombosis, J Neurosci 29:5666–5670, 2009.
Shapiro AD, Angchaisuksiri P, Astermark J, et al.: Subcutaneous concizumab prophy- Yamazaki R, Nishiyama O, Saeki S, et al.: The utility of the Japanese Association for
laxis in hemophilia A and hemophilia A/B with inhibitors: phase 2 trial results, Blood Acute Medicine DIC scoring system for predicting survival in acute exacerbation of
28(22):134, 2019. fibrosing idiopathic interstitial pneumonia, PloS One 14(8):e0212810, 2019.
Shariat-­Madar Z, Mahdi F, Warnock M, et al.: Bradykinin B2 receptor knockout mice Yang XV, Banerjee Y, Fernandez JA, et al.: Activated protein C ligation of ApoER2
are protected from thrombosis by increased nitric oxide and prostacyclin, Blood (LRP8) causes Dab1-­dependent signaling in U937 cells, Proc Natl Acad Sci U S A
108:192–199, 2006. 106:274–279, 2009.
Smith SA, Travers RJ, Morrissey JH: How it all starts: initiation of the clotting cascade, Yates SG, Fitts E, De Simone N, Sarode R: Prolonged activated partial yhromboplastin
Crit Rev Biochem Mol Biol 1– 11, 2015. time: to mix or not to mix – is that the question? Transfus Apher Sci 58(1):39–42, 2019
Soucie JM, Miller CH, Kelly FM, et al.: Haemophilia Inhibitor Research Study Investi- Feb.
gators: a study of prospective surveillance for inhibitors among persons with haemo- Zaffran Y, Meyer SC, Negrescu E, et al.: Signaling across the platelet adhesion recep-
philia in the United States, Haemophilia 20:230–237, 2014. tor glycoprotein Ib-­IX induces αIIbβ3 activation in both platelets and a transfected
Stavrou EX, Fang C, Merkulova A, et al.: Reduced thrombosis on KLKB1-­/-­ mice is me- Chinese hamster ovary cell system, J Biol Chem 275:16779–16787, 2000.
diated by increased Mas receptor, prostacyclin, Sirt1 and KLF4 and decreased tissue Zhang B, Cunningham MA, Nichols WC, et al.: Bleeding due to disruption of a cargo-­
factor, Blood 125:710–719, 2015. specific ER-­to-­Golgi transport complex, Nat Genet 34:220–225, 2003.
Sun WY, Witte DP, Degen JL, et al.: Prothrombin deficiency results in embryonic and Zhang B, Ginsburg D: Familial multiple coagulation factor deficiencies: new biologic
neonatal lethality in mice, Proc Natl Acad Sci U S A 95:7597–7602, 1998. insight from rare genetic bleeding disorders, J Thromb Haemost 2:1564–1572, 2004.

846.e3
Downloaded for Gemma Fabiola Pérez Aguilar (gemma.perez@udem.edu) at University of Monterrey from ClinicalKey.com by Elsevier on
September 19, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.

You might also like