You are on page 1of 67

Copyright © 2007 by F. A. Davis.

15 Overview of Hemostasis
and Platelet Physiology

DONNA CASTELLONE

History of Blood Coagulation Objectives


Overview of Coagulation After completing this CHAPTER, the student will be ABLE to:
Vascular System 1. Describe the systems involved in hemostasis.
Overview 2. Describe the interaction of the vascular system
Mechanism of Vasoconstriction and platelets with regard to activation, adhe-
The Endothelium sion, and vasoconstriction.
Events Following Vascular Injury 3. Identify the process involved in the coagulation
cascade from activation to stable clot formation.
Primary Hemostasis
Platelets: An Introduction 4. Describe the role of platelets in hemostasis with
respect to platelet glycoproteins, platelet bio-
Platelet Development chemistry, and platelet function.
Platelet Structure and Biochemistry
5. Define the difference between primary and sec-
Platelet Function and Kinetics ondary hemostasis.
Platelet Aggregation Principle
6. Outline the intrinsic and extrinsic pathways,
Secondary Hemostasis the factors involved in each, and their role in
Classification of Coagulation Factors the coagulation system.
Physiological Coagulation (In Vivo) 7. List the coagulation factors, their common
Laboratory Model of Coagulation names, and function.
Extrinsic Pathway 8. Explain the interaction between the prothrom-
Intrinsic System bin time, activated partial thromboplastin time,
Activated Partial Thromboplastin Time and factor assays.
Common Pathway 9. Identify the relationship of the kinin and com-
Formation of Thrombin plement systems to coagulation.
Feedback Inhibition 10. Identify the inhibitors and their role in hemo-
Fibrinolysis stasis.
Coagulation Inhibitors
Kinin System
Complement System

229
Copyright © 2007 by F. A. Davis.

230 PART IV • Hemostasis and Disorders of Coagulation

HISTORY OF BLOOD COAGULATION Testing of blood plasma factors and platelets


depended on seeing the clotting process directly or
The study of blood coagulation can be traced back to microscopically. The first whole blood clotting time was
about 400 BC and the father of medicine, Hippocrates. done in 1780 by William Hewson, who noted that
He observed that the blood of a wounded soldier con- blood taken from healthy people clotted in 7 minutes
gealed as it cooled. Additionally, he noticed that bleed- while in some disease states, blood took from 15 to 20
ing from a small wound stopped as skin covered the minutes up to 11/2 hours to form a clot.
blood. If the skin was removed, bleeding started again. In 1897, Brodie and Russel begin observing the
Aristotle noted that blood cooled when removed from process on a glass slide. A drop of blood was placed on a
the body and that cooled blood initiated decay resulting glass cone, in a temperature-controlled glass chamber
in the congealing of the blood. If fibers were removed, agitated by an air jet. Blood no longer moved micro-
there was no clotting. This was known as the cooling scopically but clotted in 3 minutes and was completed
theory or blood coagulation. It was not until 1627 that at 8 minutes. In 1905, Golhorm used a wire loop
Mercurialis observed clots in veins that were at body attached to a glass tube. In 1910, Kottman observed an
temperature. In 1770, William Hewson challenged the increased viscosity in clotting blood in a Koagulo-
cooling theory and believed that air and lack of motion viskosimeter. Blood was rotated at 20 degrees 12 to 15
were important in the initiation of clotting. Hewson times per minute. In 1936, Baldes and Nygaard added
described the clotting process, demonstrating that the photoelectric tracings called a coagelogram, depicting
clot comes from the liquid portion of blood, the coagu- shape change by light transmittance.
lable lymph, and not from the cells, disproving the cool- In the 1960s, BBL introduced the Fibrometer. This
ing theory. It was Paul Morawitz in 1905 who instrument provided mechanical registration of clots
assembled coagulation factors into the scheme of coag- that allowed more reproducible timing and an expres-
ulation and demonstrated that in the presence of cal- sion of the clotting process.2
cium and thromboplastin, prothrombin (II) was
converted to thrombin, which in turn converted fib-
OVERVIEW OF COAGULATION
rinogen (I) into a fibrin clot. This theory persisted for 40
years until Paul Owren, in 1944, discovered a bleeding Coagulation is a complex network of interactions
patient who defied the four-factor concept of clotting. involving vessels, platelets, and factors. The ability to
Thus factor V was discovered. Owren also observed a form and to remove a clot is truly a system dependent
cofactor that was involved in the conversion of pro- on many synergistic forces. Hemostasis depends on a
thrombin to thrombin. In 1952, Loeliger named this system of checks and balances between thrombosis and
factor VII. Factor VIII was identified as classic hemo- hemorrhage that includes both procoagulants and anti-
philia prior to the identification of VII in 1936/1937. In coagulants. This scale needs to be kept in balance.
1947, Pavlovsky reported that the blood from some Thrombosis is an activation of the hemostatic system at
hemophiliac patients corrected the abnormal clotting an inappropriate time in a vessel. Thrombi formed in
time in others. In 1952, this was called Christmas dis- this fashion are pathologic and beyond the normal
ease, after the family in which it was discovered, or fac- hemostatic mechanism. If physiological anticoagulants
tor IX. Factor X deficiency was described in 1957 in a are decreased in the circulation there will be a clot. If
woman named Prower and a man named Stuart. Factor procoagulants or clotting factors are decreased, the
XI was described in 1953 as a milder bleeding tendency. scale will tip toward bleeding. Hemorrhage or excessive
In 1955, Ratnoff and Colopy identified a patient, John bleeding may be due to blood vessel disease, rupture,
Hageman, with a factor XII deficiency who died from a platelet abnormalities, and acquired or congenital
stroke—a thrombotic episode, not a bleeding disease. abnormalities. Hemostasis is comprised of the vascular
In 1960, Duckert described patients who had a bleed- system, platelets, and a series of enzymatic reactions of
ing disorder and characteristic delayed wound healing. the coagulation factors. The role of hemostasis is to
This fibrin stabilizing factor was called factor XIII. arrest bleeding from a vessel wall defect, while at the
Prekallikrein (1965) discovered from four siblings in same time maintaining fluidity within circulation.
the Fletcher family demonstrated no bleeding tenden- Under physiological conditions, fluidity is maintained
cies, as well as high-molecular-weight kininogen by the anticoagulant, profibrinolytic, and antiplatelet
(1975). These were both identified as contact activation properties of the normal endothelium.3
cofactors that participated in the activation of factor XI Coagulation is divided into two major systems: the
by factor XII.1 primary and secondary systems of hemostasis. The pri-
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 231

mary system comprises platelet function and vasocon- lumen of the blood vessel are the principal elements
striction. The secondary system involves coagulation regulating vascular functions. Physiologically, the sur-
proteins and a series of enzymatic reactions. Once the face of endothelial cells is negatively charged and repels
coagulation proteins become involved, fibrin is formed circulating proteins and platelets, which are negatively
and this reinforces platelet plug formation until healing charged.6 Vasoconstriction occurs very quickly and is
is complete. The product of the coagulation cascade is effective in stopping bleeding in small blood vessels but
the conversion of soluble fibrinogen into an insoluble cannot prevent bleeding in larger vessels. Other systems
fibrin clot. This is accomplished by the action of a pow- are required for this task.
erful coagulant, thrombin. Thrombin is formed by a pre-
cursor circulating protein, prothrombin. Dissolution of The Endothelium
the platelet plug is achieved by the fibrinolytic process.
The endothelium contains connective tissue such as
collagen and elastin. This matrix regulates the perme-
VASCULAR SYSTEM ability of the inner vessel wall and provides the princi-
Overview pal stimuli to thrombosis following injury to a blood
vessel. Circulating platelets recognize and bind to insol-
The vascular system prevents bleeding through vessel uble subendothelial connective tissue molecules. This
contraction, diversion of blood flow from damaged ves- process is dependent on molecules that are in plasma
sels, initiation of contact activation of platelets with and on platelets. Two factors, von Willebrand (vWF)
aggregation, and contact activation of the coagulation and fibrinogen, participate in the formation of the
system.4 The vessel wall contains varying amounts of platelet plug and the insoluble protein clot, resulting in
fibrous tissue such as collagen and elastin, as well as the activation of the coagulation proteins. Receptor
smooth muscle cells and fibroblasts. Arteries are the molecules not only adhere to platelets and damaged
vessels that take blood away from the heart and have the vessel components but also allow platelets to use vWF
thickest walls of the vascular system. Veins return blood and fibrinogen to bind platelets and form a plug. Blood
to the heart, and are larger with a more irregular lumen flows out through the wall and comes in contact with
than the arteries. Veins, however, are thin walled, with collagen. Collagen is an insoluble fibrous protein that
elastic fibers found only in larger veins. Arterioles are a accounts for much of the body’s connective tissue. Ves-
smaller subdivision of arteries, and venules are smaller sel injury leads to the stimulation of platelets. Platelets
subdivisions of veins. Capillaries are the thinnest walled contain more of the contractile protein actomyosin than
and most numerous of the blood vessels. They are com- any cells, other than muscle cells, giving them the abil-
posed of only one cell layer of endothelium that permits ity to contract. Basically platelets adhere to collagen and
a rapid rate of transport materials between blood and other platelets adhere to them. A plug is built and the
tissue.5 platelets’ ability to further contract compacts the mass.7
In forming the initial plug, platelets have now built
Mechanism of Vasoconstriction a template on a lipoprotein surface, which in turn acti-
vates tissue factor. The balance between coagulation
The process in which coagulation occurs begins with
proteins and anticoagulants now leans toward coagula-
injury to a vessel. The first response of a cut vessel is
tion. This process will accelerate vasoconstriction,
vasoconstriction or narrowing of the lumen of the arte-
platelet plug development, and the formation of cross-
rioles to minimize the flow of blood from the wound
linked fibrin clot (Fig. 15.1).
site. The blood is ordinarily exposed to only the
endothelial cell lining of the vasculature. When this is
Events Following Vascular Injury
invaded, the exposed deeper layers of the blood vessel
become targets for cellular and plasma components. 1. Thromboresistant properties of a blood vessel
Vasoconstriction occurs immediately and lasts a short maintain blood in a fluid state.
period of time. It allows for increased contact between 2. After vascular injury, subendothelial compo-
the damaged vessel wall, blood platelets, and coagula- nents of collagen induce platelet aggregation,
tion factors. Vasoconstriction is caused by several regu- which is mediated by vWF and platelet recep-
latory molecules including serotonin and thromboxane tor glycoprotein Ib.
A2, which interacts with receptors on the surface of cells 3. Further platelet recruitment occurs as a result
of the blood vessel wall. These are products of platelet from fibrinogen binding to its platelet recep-
activation and endothelium. Endothelial cells lining the tor, glycoprotein IIb/IIIa.
Copyright © 2007 by F. A. Davis.

232 PART IV • Hemostasis and Disorders of Coagulation

INJURY TO VESSEL are called megakaryocytes (Fig. 15.2). These large cells
(80 to 150 μm) are found in the bone marrow.
Megakaryocytes do not undergo complete cellular divi-
Activated platelets sion but undergo a process called endomitosis or
Exposed collagen
endoreduplication creating a cell with a multilobed
nucleus. Each megakaryocyte produces about 2000
platelets. Thrombopoietin is responsible for stimulating
maturation and platelet release. This hormone is gener-
Endothelium
Injury to vessel
ated primarily by the kidney and partly by the spleen
and liver.10 There is no reserve of platelets in the bone
marrow: 80% are in circulation and 20% are in the red
PLATELET RESPONSE pulp of the spleen. Platelets have no nucleus but do
have granules: alpha granules, and dense granules.
These granules are secreted during the platelet release
Platelet reaction and contain many biochemically active com-
ponents such as serotonin, ADP, and ATP. They are
destroyed by the reticuloendothelial system (RE).
Platelet development occurs in the following
GPIb
VWF
sequence:
1. MEGAKARYOBLASTS are the most immature cell
Collagen (10 to 15 μm) with a high nuclear to cytoplas-
Figure 15.1 Platelet response to vascular injury. mic ratio and two to six nucleoli.
2. PrOMEGAKARYOCYTE is a large cell of 80 μm
with dense alpha and lysosomal granules.
4. Tissue factor generates thrombin, which 3. BASOPHILIC MEGAKARYOCYTE shows evidence
results in cross-linked fibrin strands that rein- of cytoplasmic fragments containing mem-
force the platelet plug. branes, cytotubules, and several glycoprotein
5. Platelet actomyosin mediates clot retraction to receptors.
compact the platelet mass.8 4. The MEGAKARYOCYTE is composed of cytoplasmic
fragments that are released by a process called
PRIMARY HEMOSTASIS the budding of platelets.
Platelets: An Introduction
Platelets were recognized in 1882 by Bizzozero as a cell Platelet Structure and Biochemistry
structure different from red and white cells. However, it Platelets have a complex structure comprised of
was not until 1970 that platelets’ relationship to hemo- four zones: the peripheral zone, the sol gel zone, the
stasis and thrombosis became so important.9 Every
cubic millimeter of blood contains one-fourth of 1 bil-
lion platelets, resulting in approximately a trillion
platelets in the blood of an average woman. Each
From The College of American Pathologists, with permission.

platelet makes 14,000 trips through the bloodstream in


its life span of 7 to 10 days.7

Platelet Development
Platelets, or thrombocytes, are small discoid cells (0.5 to
3.0 μm) that are synthesized in the bone marrow and
stimulated by the hormone thrombopoietin. They are
developed through a pluripotent stem cell that has been
influenced by colony-stimulating factors (CSF) pro-
duced by macrophages, fibroblasts, T-lymphocytes, and
stimulated endothelial cells. The parent cells of platelets Figure 15.2 Megakaryocyte, the platelet parent cell.
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 233

GpIb

Mitochondria
GpIIb/IIIa
ADP
Microtubules
EPI
Thrombin

PAF Dense granules


Collagen
Thromboxane

Open canalicular
IIa system
Dense tubular
system VIII
IXa Ca Va
Figure 15.3 Schematic diagram of platelet
Xa Ca Vacuoles
morphology.

organelle zone, and the membrane system (Table 15.1). disc to spiny spheres. Glycoprotein (GP) Ib
Figure 15.3 is a diagram of platelet morphology. and vWF aid in adhesion. This is primary
aggregation and is reversible. This reaction is
Platelet Function and Kinetics mediated by the release of platelet granules.
• REACTION 2 (AGGREGATION): In response
Platelets play an important role in both the formation of
to chemical changes, these events lead to
a primary plug as well as the coagulation cascade. The
platelet aggregation in which platelets adhere
formation of a plug at the site of a cut vessel serves as the
to other platelets. Platelet shape change occurs.
initial mechanical barrier. The lumen of the vessel is
• REACTION 3 (RELEASE): Platelets release
lined with endothelial cells; a break in this will initiate a
the contents of their dense granules. The
series of reactions.
release of these granules constitutes a sec-
There are four phases to platelet function:
ondary aggregation that is irreversible. Throm-
• REACTION 1 (ADHESION): Platelets adhere boxane A2 is released by platelets, which
to collagen and undergo shape change from promotes vasoconstriction. ADP amplifies
the process.
• REACTION 4 (STABILIZATION OF THE
Table 15.1  The Four Functional CLOT): This reaction is responsible for throm-
Platelet Zones bus formation. The adherent and aggregated
platelets release factor V and expose platelet
1. The peripheral zone is associated with platelet adhe- factor 3 to accelerate the coagulation cascade
sion and aggregation. and promote activation of clotting factors and
2. The sol gel zone provides a cytoskeletal system for ultimately stabilize the platelet plug with a fib-
platelets and contact when the platelets are stimu- rin clot.
lated.
3. The organelle zone contains three types of granules:
The platelet membrane contains important recep-
alpha, dense bodies, and lysosomes. tors called GPs on the platelet surface. Further interac-
4. The membrane system contains a dense tubular sys- tions are mediated by both plasma protein receptors of
tem in which the enzymatic system for the produc- vWF and fibrinogen. Other activators of platelets are
tion of prostaglandin synthesis is found. thrombin, ADP, thromboxane A2, serotonin, epineph-
rine, and arachidonic acid.
Copyright © 2007 by F. A. Davis.

234 PART IV • Hemostasis and Disorders of Coagulation

The receptor for vWF is GPIb-IX. GPIIb/IIIa are of aggregation and is preceded by a shape change except
receptors for fibronectin, vWF, fibrinogen, and factors when platelets are stimulated with epinephrine (Fig.
V and VIII. This interaction recruits more platelets 15.4). Primary aggregation is a reversible process. The
to interact with each other.11 Adhesion of platelets second phase of platelet aggregation occurs when
to collagen and each other can occur without contrac- platelet granule contents are secreted. Secondary aggre-
tion or shape change. Contraction causes shape change gation is irreversible. Epinephrine, collagen, ADP, and
into a spiny sphere. Exposure of a negatively charged arachidonic acid are the aggregating agents most fre-
membrane leads to secretion of granular contents. quently used in clinical platelet aggregation.
These activated platelets release ADP and synthesized 1. Epinephrine (EPI): When added to platelet
thromboxane A2, which mediate activation of addi- rich plasma (PRP), it will stimulate platelets
tional platelets, resulting in the formation of a platelet to aggregate. Normal platelets will respond
plug.12 by releasing endogenous ADP from their gran-
ules. Both primary and secondary aggregation
is seen. An abnormal response is due to an
Platelet Aggregation Principle absent or decreased release of nucleotides
Aggregation defines the ability of platelets to stick to from dense granules.
one another. The formation of aggregates is observed 2. Adenosine DIPHOSPHATE (ADP): When added
with a platelet aggregometer. This is a photo-optical to PRP, it will stimulate platelets to change
instrument connected to a chart recorder. Light trans- their shape and aggregate. Aggregation is
mittance through the sample is increased and converted induced by exogenous ADP at a high dose of
into electronic signals, which are amplified and 20 μmol/L. The primary and secondary wave
recorded. A characteristic curve is formed with each aggregations are indistinguishable. Reversible
aggregating agent. Primary aggregation is the first wave aggregation may occur due to an inadequate

Platelet Aggregation Tracings


Normal Aggregation
Shape change

Primary aggregation: adhere and aggregate

Secondary aggregation: release of granules

Formation of plug

Dissaggregation

Shape change
Primary aggregation

Reversible aggregation

Abnormal: only primary aggregation Abnormal

Figure 15.4 Platelet aggregation. Note the


stages of aggregation, which include primary
No release of granules No response and secondary aggregation as well as shape
change and plug formation.
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 235

release of nucleotides. Lack of a secondary There are three groups in which coagulation fac-
wave is indicative of defective thromboxane tors can be classified:
production and/or a defective granule pool. 1. The fibrinogen group consists of factors I, V,
3. COLLAGEN: When added to PRP, the platelets VIII, and XIII. They are consumed during
adhere to the collagen, followed by shape coagulation. Factors V and VIII are labile and
change, release of endogenous ADP, and then will increase during pregnancy and inflamma-
aggregation. An abnormal response to collagen tion.
may be seen if thromboxane production is 2. The prothrombin group: Factors II, VII, IX, and
deficient. Aggregation is slower and less com- X all are dependent on vitamin K during their
plex, resulting in a decreased response. synthesis. This group is stable and remains
4. ARACHIDONIC ACID (AA): This is a fatty preserved in stored plasma.
acid present in membranes of human 3. The CONTACT group: Factor XI, factor XII,
platelets and liberated from phospholipids. prekallikrein, and high-molecular-weight
In the presence of the enzyme kininogen (HMWK) are involved in the intrin-
cyclooxygenase, oxy- gen is incorporated to sic pathway, moderately stable, and not con-
form the endoperoxide prostaglandin G2 sumed during coagulation.5
(PGG2). PGG2 is then con-
verted to thromboxane A2, a potent inducer The coagulation factors and their actions are listed
of platelet aggregation. in (Table 15.2).

Factor I, Fibrinogen
SECONDARY HEMOSTASIS
Substrate for thrombin and precursor of fibrin, it is a
Secondary hemostasis involves a series of blood protein
large globulin protein. Its function is to be converted
reactions through a cascade-like process that concludes
into an insoluble protein and then back to soluble com-
with the formation of an insoluble fibrin clot. This sys-
ponents. When exposed to thrombin, two peptides split
tem involves multiple enzymes and several cofactors as
from the fibrinogen molecule, leaving a fibrin monomer
well as inhibitors to keep the system in balance. Coagu-
to form a polymerized clot.
lation factors are produced in the liver, except for factor
VIII, which is believed to be produced in the endothe-
lial cells. When the factors are in a precursor form, the Factor II, Prothrombin
enzyme or zymogen is converted to an active enzyme or Precursor to thrombin, in the presence of Ca2+, it is
a protease. converted to thrombin (IIa), which in turn stimu- lates
The initiation of clotting begins with the activation platelet aggregation and activates cofactors pro- tein
of two enzymatic pathways that will ultimately lead to C and factor XIII. This is a vitamin K–dependent
fibrin formation: the intrinsic and extrinsic pathways. factor.
Both pathways are necessary for fibrin formation, but
their activating factors are different. Intrinsic activation Factor III, Thromboplastin
occurs by trauma within the vascular system, such as
Tissue factor activates factor VII when blood is exposed
exposed endothelium. This system is slower and yet
to tissue fluids.
more important versus the extrinsic pathway, which is
initiated by an external trauma, such as a clot and
occurs quickly. Factor IV, Ionized Calcium
This active form of calcium is needed for the activation
of thromboplastin and for conversion of prothrombin
Classification of Coagulation Factors
to thrombin.
Coagulation factors may be categorized into substrates,
cofactors, and enzymes. Substrates are the substance Factor V, Proaccelerin or Labile Factor
upon which enzymes act. Fibrinogen is the main sub-
strate. Cofactors accelerate the activities of the enzymes This is consumed during clotting and accelerates the
that are involved in the cascade. Cofactors include tis- transformation of prothrombin to thrombin. A vitamin
sue factor, factor V, factor VIII, and Fitzgerald factor. All K–dependent factor, 20% of factor V is found on
of the enzymes are serine proteases except factor XIII, platelets.
which is a transaminase.13
Copyright © 2007 by F. A. Davis.

236 PART IV • Hemostasis and Disorders of Coagulation

Table 15.2  Factor Facts

Half-life Clinical Picture If Factor for Screening


Factor Inheritance (hr) Deficient Hemostasis Tests
I Autosomal domi- 64 to 96 Bleed with trauma, stress, mucosal, 40 to 50  PT and aPTT
nant umbilical stump, intracranial, gas- mg/dL
trointestinal
II Autosomal recessive 48 Severe bleed, mucous membrane, spon- 20% to 30%  PT and aPTT
taneous
V Autosomal recessive 12 Moderate-severe bleed, mucosal, large 10% to 15%  PT and aPTT
ecchymoses
VII Autosomal recessive 4 to 6 Intra-articular bleed, severe mucosal, 10% to 15%  PT
epistaxis, hemarthrosis, genitourinary,
gastrointestinal, and intrapulmonary
VIII Sex-linked recessive 15 to 20 Severity based on levels, hematuria,
hemarthrosis, intra-articular, >10%  aPTT
intracranial
IX Sex-linked recessive 24 Severe mucous membrane, deep tissue, >10%  aPTT
intra-muscular
X Autosomal recessive 32 Mucous membrane, skin hemorrhages 10% to 15%  PT and aPTT
XI Autosomal recessive 60 to 80 Severity of bleeds vary, not proportional 30%  aPTT
to factor level
XII Autosomal recessive 50 to 70 Hemorrhage is rare, risk for thrombosis ?  aPTT
and dominant
XIII Autosomal recessive 40 to 50 Only homozygotes bleed, deep tissue 10% Normal PT
muscle, intracranial bleed and aPTT

Factor VI, Nonexistent Factor X, Stuart-Prower


Factor VII, Proconvertin or Stable Factor Final common pathway merges to form conversion
This is activated by tissue thromboplastin, which in of prothrombin to thrombin, activity also related to
turn activates factor X. It is a vitamin K–dependent factors VII and IX. It is vitamin K–dependent and can
factor. be independently activated by Russell’s viper venom.

Factor XI, Plasma Thromboplastin Antecedent


Factor VIII, Antihemophilic
Essential to intrinsic thromboplastin generating of the
This cofactor is used for the cleavage of factor X-Xa by cascade, it has increased frequency in the Jewish popu-
IXa. Factor VIII is described as VIII/vWF:VIII:C active lation. Bleeding tendencies vary, but there is the risk of
portion, measured by clotting, VIII:Ag is the antigenic postoperative hemorrhage.
portion, vWF:Ag measures antigen that binds to
endothelium for platelet function; it is deficient in Factor XII, Hageman factor
hemophilia A.
This surface contact factor is activated by collagen.
Patients do not bleed but have a tendency to thrombosis.
Factor IX, Plasma Thromboplastin Component
A component of the thromboplastin generating system, Factor XIII, Fibrin Stabilizing Factor
it influences amount as opposed to rate. It is deficient in In the presence of calcium, this transaminase stabilizes
hemophilia B, also known as Christmas disease. It is sex polymerized fibrin monomers in the initial clot. This is
linked and vitamin K–dependent. the only factor that is not found in circulating plasma.
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 237

High-Molecular-Weight Kininogen Laboratory Model of Coagulation


This surface contact factor is activated by kallikrein. Laboratory testing looks at the in vitro effect of the coag-
ulation process which is measured by the prothrombin
Prekallikrein, Fletcher Factor time (PT), activated partial thromboplastin time
(aPTT), thrombin time (TT), fibrin degradation prod-
This is a surface contact activator, in which 75% is
ucts (FDPs), and D-dimer. This section will focus on PT
bound to HMWK.
and a PTT, while Chapter 20 will concentrate on the
other routine tests mentioned. While the coagulation
Physiological Coagulation (In Vivo) cascade does not reflect what goes on in vivo, it provides
The original theory of coagulation used a cascade or a model in which the laboratory relates to for testing.
waterfall theory. This description depicted the genera- However, the coagulation cascade reflects the mecha-
tion of thrombin by the soluble coagulation factors and nisms that the laboratory uses for results. The screening
the initiation of coagulation. This theory identified two tests provide a tremendous amount of information to
starting points for the generation of thrombin: the the physician. They can be performed both quickly and
initiation of the Intrinsic pathway with factor XII accurately (Fig. 15.6).
and surface contact, and the extrinsic pathway with fac-
tor VIIa and tissue factor. These two pathways meet Extrinsic Pathway
at the common pathway, where they both generate
The extrinsic pathway is initiated by the release of tissue
factor Xa from X, leading to a common pathway
thromboplastin that has been expressed after damage to
of thrombin from prothrombin and the conversion of
fibrinogen to fibrin. This process holds true under labo-
ratory conditions (Fig. 15.5). The discovery of a natu-
rally occurring inhibitor of hemostasis, tissue factor aPTT (Heparin therapy) PT (Coumadin therapy)

pathway inhibitor (TFPI), is able to block the activity of INTRINSIC SYSTEM EXTRINSIC SYSTEM

the tissue factor VIIa complex, soon after it becomes Contact activation pathway Tissue factor pathway

active.14 XII VII


HMWK
Prekallikrein

XIIa Ca + Tissue factor


XI
Revised Coagulation Cascade: In Vivo
XIa
VIIIa
Vessel injury IX

XI XIa IXa
TF–VIIa IX IXa Ca

Ca Pl VIIIa VIII + Ca + PF

X
X Xa

Ca Pl Va Xa

II IIa V + Ca + PF

Fibrinogen Fibrin Prothrombin II Thrombin IIa Factor XIII

XIIIa
Fibrinogen I Fibrin XIIIa

Insoluble monomer
cross-linked fibrin Fibrin clot

Figure 15.5 In vivo coagulation cascade. Figure 15.6 In vitro coagulation cascade.
Copyright © 2007 by F. A. Davis.

238 PART IV • Hemostasis and Disorders of Coagulation

a vessel. Factor VII forms a complex with tissue throm- Calcium is required for the activation of X to proceed
boplastin and calcium. This complex converts factors X rapidly. The reaction then enters the common pathway
and Xa, which in turn converts prothrombin to throm- where both systems involve factors I, II, V, and X. This
bin. Thrombin then converts fibrinogen to fibrin. This results in a fibrin monomer polymerizing into a fibrin
process takes between 10 and 15 seconds. clot. Factor XIII, or fibrin stabilizing factor, follows acti-
Prothrombin time (PT) developed by Armond vation by thrombin. This will convert initial weak
Quick in 1935 measures the extrinsic system of coagu- hydrogen bonds, cross-linking fibrin polymers to a
lation. It is dependent upon the addition of calcium more stable covalent bond.
chloride and tissue factor. It uses a lipoprotein extract
from rabbit brain and lung.1 Activated Partial Thromboplastin Time
PT uses citrate anticoagulated plasma. After the
addition of an optimum concentration of calcium and aPTT measures the intrinsic pathway. The test consists
an excess of thromboplastin, clot detection is measured of recalcifying plasma in the presence of a standardized
by an automated device for fibrin clot detection. The amount of platelet-like phosphatides and an activator of
result is reported in seconds. PT is exclusive for factor the contact factors. It will detect abnormalities to factors
VII, but this test is also sensitive to decreases in the com- VIII, IX, XI, and XII. The aPTT is also used to monitor
mon pathway factors. Therefore, if a patient presents heparin therapy. Heparin is an anticoagulant used to
with a prolonged PT and there is no other clinical treat and or prevent acute thrombotic events such as
abnormality or medication, the patient is most likely deep vein thrombosis (DVT), pulmonary embolism
factor VII deficient. The PT is also used to monitor oral (PE), or acute coronary syndromes. The action of
anticoagulation or warfarin therapy used to treat and heparin is to inactivate factors XII, XI, and IX in the
prevent blood clots. In many instances, patients are presence of antithrombin. Laboratory monitoring of
placed on life-long therapy and the dosage is monitored heparin therapy will be discussed in Chapter 19.
by the PT test. The attempt in anticoagulant therapy is
to impede thrombus formation without the threat of Common Pathway
morbidity or mortality from hemorrhage. The common pathway is the point at which the intrinsic
Warfarin is an oral anticoagulant, which means it and extrinsic pathways come together and factors I, II,
must be ingested. It was discovered in 1939 at the Uni- V, and X are measured. It is important to note that the
versity of Wisconsin quite by accident. It seems that a PT and the aPTT will not detect qualitative or quantita-
farmer found that his cattle were hemorrhaging to tive platelet disorders, or a factor XIII deficiency. Factor
death, for what appeared to be no reason. The cattle XIII is fibrin stabilizing factor and is responsible for sta-
were grazing in a field eating sweet clover. This contains bilizing a soluble fibrin monomer into an insoluble fib-
dicumarol, actually bis-hydroxy coumadin, which rin clot. If a patient is factor XIII deficient, the patient
caused the cattle to bleed.6 will form a clot but will not be able to stabilize the clot
There are several compounds of coumadin: and bleeding will occur later. Factor XIII is measured by
dicumarol, indanedione, and warfarin. Dicumarol a 5 mol/L urea test that looks at not only the formation
works too slowly, and indanedione has too many side of the clot but also if the clot lyzes after 24 hours.
effects. Warfarin or 4-oxycoumarin is the most com-
monly used oral anticoagulant. Coumadin works by
Formation of Thrombin
inhibiting the y-carboxylation step of clotting and the
vitamin K–dependent factors.15 Laboratory monitoring When plasma fibrinogen is activated by thrombin, this
of oral anticoagulation therapy will be discussed in conversion results in a stable fibrin clot. This clot is a
Chapter 19. visible result that the action of the protease enzyme
thrombin has achieved fibrin formation. Thrombin is
also involved in the XIII-XIIIa activation due to the
Intrinsic System
reaction of thrombin cleaving a peptide bond from each
Contact activation is initiated by changes induced by of two alpha chains. Inactive XIII along with Ca2+ ions
vascular trauma. Prekallikrein is required as a cofactor enables XIII to dissociate to XIIIa. If thrombin were
for the autoactivation of factor XII by factor XIIa. XI is allowed to circulate in its active form (Ia), uncontrol-
activated and requires a cofactor of HMWK. XIa acti- lable clotting would occur. As a result thrombin circu-
vates IX to IXa, which in the presence of VIIIa converts lates in its inactive form prothrombin (II).Thrombin, a
X to Xa. Also present are platelet phospholipids PF3. protease enzyme, cleaves fibrinogen (factor I) which
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 239

results in a fibrin monomer and fibrinogen peptides A patients with contact factor abnormalities (factors XI
and B. These initial monomers polymerize end to end and XII) do not bleed.8 See Figure 15.7 for a diagram of
due to hydrogen bonding. feedback inhibition.
Formation of fibrin occurs in three phases:
Fibrinolysis
1. Proteolysis: Protease enzyme thrombin cleaves
fibrinogen resulting in a fibrin monomer, A The fibrinolytic system is responsible for the dissolu-
and B fibrinopeptide. tion of a clot. Fibrin clots are not intended to be perma-
2. POLYMERIZATION: This occurs spontaneously due nent. The purpose of the clot is to stop the flow of blood
to fibrin monomer that line up end-to-end due until the damaged vessel can be repaired. The presence
to hydrogen bonding. or absence of hemorrhage or thrombosis depends on a
3. STABILIZATION: This occurs when the balance between the procoagulant and the fibrinolytic
fibrin monomers are linked covalently by system. The key components of the system are plas-
XIIIa into fibrin polymers forming an minogen, plasminogen activators, plasmin, fibrin, fib-
insoluble fibrin clot. rin/FDP, and inhibitors of plasminogen activators and
plasmin.6 Fibrinolysis is the process by which the
hydrolytic enzyme plasmin digests fibrin and fibrino-
Feedback Inhibition gen, resulting in progressively reduced clots. This sys-
Some activated factors have the ability to destroy other tem is activated in response to the initiation of the
factors in the cascade. Thrombin has the ability to tem- activation of the contact factors. Plasmin is capable of
porarily activate V and VIII, but as thrombin increases it digesting either fibrin or fibrinogen as well as other fac-
destroys V and VIII by proteolysis. Likewise, factor Xa tors in the cascade (V, VIII, IX, and XI). Normal plasma
enhances factor VII, but through a reaction with tissue contains the inactive form of plasmin in a precursor
factor pathway inhibitor (TFPI), it will prevent further called plasminogen. This precursor remains dormant
activation of X by VIIa and tissue factor. Therefore, until it is activated by proteolytic enzymes, the kinases,
these enzymes limit their own ability to activate the or plasminogen activators. Fibrinolysis is controlled by
coagulation cascade at different intervals. the plasminogen activator system. The components of
Thrombin feedback activation of factor IX can pos- this system are found in tissues, urine, plasma, lysoso-
sibly explain how intrinsic coagulation might occur in mal granules, and vascular endothelium.
the absence of contact factors. Tissue factor is expressed An activator, tissue-plasminogen activator (tPA)
following an injury forming a complex with VIIa, then results in the activation of plasminogen to plasmin
activating X and IX. TFPI prevents further activation of resulting in the degradation of fibrin. The fibrinolytic
X. Thrombin formation is further amplified by factors V, system includes several inhibitors. Alpha-2-antiplasmin
VIII, and XI, which leads to activation of the intrinsic is a rapid inhibitor of plasmin activity and alpha- 2-
pathway. This feedback theory helps to enforce why macroglobulin is an effective slow inhibitor of plas-

Feedback Inhibition:
Factor XI Factor XIa Factor IX Tissue factor + Factor VIIa Factor VII

Factor IXa Factor X


Factor VIII Factor VIIIa

Factor Va + Factor Xa

Factor V Factor II

Figure 15.7 Feedback inhi-


bition. Note the role of throm- Thrombin IIa
bin in the activation and
deactivation of coagulation
factors. Fibrinogen Fibrin
Copyright © 2007 by F. A. Davis.

240 PART IV • Hemostasis and Disorders of Coagulation

min activity. This system is in turn controlled by Kinin System


inhibitors to tPA and plasmin-plasminogen activator Another plasma protein system in coagulation is the
inhibitor 1 (PAI-1) and alpha-2-antiplasmin. Reduced kinin system. This system is capable of vascular dilata-
fibrinolytic activity may result in increased risk for car- tion leading to hypotension, shock, and end-organ
diovascular events and thrombosis. damage by its capability to increase vascular perma-
Pharmacologic activators are currently used for
bility.16 The kinins are peptides of 9 to 11 amino acids.
therapeutic thrombolysis, including streptokinase, The kinin system is activated by factor XII. Hageman
urokinase, and tPA. factor XIIa converts prekallikrein (Fletcher factor) into
Urokinase directly activates plasminogen into
kallikrein, and kallikrein converts kininogens into
plasmin, and streptokinase forms a streptokinase plas- kinins. The most important is bradykinin (BK). This is
minogen complex, which then converts plasminogen an important factor in vascular permeability as well as
into plasmin.16
a chemical mediator of pain. BK is capable of repro-
ducing many characteristics of an inflammatory state
Coagulation Inhibitors such as changes in blood pressure, edema, and pain,
Inhibitors are soluble plasma proteins that are natural resulting in vasodilation and increased microvessel
anticoagulants. They prevent the initiation of the clot- permeability.13
ting cascade. There are two major inhibitors in plasma
that keep the activation of coagulation under control.
Complement System
These inhibitors are:
1. Protease inhibitors: inhibitors of coagulation This system has a role in inflammation and the immune
factors, which include system as well as important thrombohemorrhagic
• Antithrombin disorders such as disseminated intravascular coagula-
• Heparin cofactor II tion (DIC). Activated complement fragments have the
• Tissue factor pathway inhibitor capacity to bind and damage self tissues. Regulators
• Alpha-2-antiplasmin of complement activation are expressed on cell surfaces.
• C1 These protect the cell from the effects of cell-bound
2. The protein C pathway: inactivation of acti- complement fragments. If this regulation process is
vated cofactors, which includes abnormal, it may participate in the pathogenesis of
• Protein C and protein S autoimmune disease as well as inflammatory disorders.
This system includes 22 serum proteins that play a
Each will be discussed in detail in Chapter 19. role in mediating immune and allergic reactions and
Table 15.3 has a listing on inhibitor and target reaction the lysis of cells due to a production of membrane
sites. attack complex (MAC). The lysis and disruption of red
blood cells and platelets lead to the release of procoagu-
lant material. This system is a sequential activation
Table 15.3  Serine Protease pathway. Complement is activated by plasmin through
Inhibitors the cleavage of C3 into C3a and C3b. C3 causes
increased vascular permeability, and because of the
Inhibitor Specificity degranulation or lysis of mast cells, which in turn
results in the release of histamine, C3b causes immune
Antithrombin (AT) IIa, Xa, IXa
adherence.13
Alpha-2-macroglobulin Nonspecific
The interrelationship between the complement,
Tissue factor pathway Xa, VIIa/TF complex kinin, and the coagulation system is complex and
inhibitor revealing. Coagulation and the elements that contribute
Heparin cofactor II IIa to the success of the hemostatic system are multifactor-
Alpha-2 protease inhibitor XIa, elastase ial, and with each decade, more knowledge about this
CI inhibitor XIIa, kallikrein, XIa, CI versatile system is learned. Figure 15.8 illustrates the
(complement system) important interrelationships between the coagulation,
fibrinolytic, complement, and kinin systems.
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 241

Kininogen Kallikrein Prekallikrein

Collagen
Phospholipids
Kinins Coagulation

Factor XII Factor XIIa Factor XIIa fragments

Plasminogen Plasmin

Fibrinolysis

Figure 15.8 Interrelationships


between the coagulation, fibri- C8C9 C3 activation C1 activation
nolytic, complement, and kinin (Lysis)
Complement activation
systems.

COND ENSED CASE


A 35-year-old woman needs to have an ovarian cyst removed. She had one delivery that was uneventful. Her mother
has a history of bleeding after tooth extraction. The physician needs to determine if there is a bleeding disorder. The
coagulation test results are as follows:
PT 12.5 seconds (Reference range, 10.5 to 13.3)
aPTT 32.1 seconds (Reference range, 28.7 to 35.5)
Platelets 320,000/mm3 (Reference range, 150,000 to 400,000/mm3)
Bleeding time 11 minutes (Reference, 8 minutes)
WHAT is the most SIGNIfiCANT ABNORMAL result in the COAGULATION PANEL?
Answer
The bleeding time is the only abnormal test, since it is greater than 8 minutes. This suggests a disorder within primary
hemostasis. This can be caused by any disorder of platelets, such as von Willebrand disease, or a problem due to
platelet secretion. Or it can be caused by several medications. Tests to rule out von Willebrand disease include factor
VIII assay, a vWF antigen and activity, as well as platelet aggregation testing. Upon performing a platelet aggregation,
there was only a primary wave for epinephrine, and no response for arachidonic acid. The patient was taking 81 μg of
aspirin a day as a preventive measure. This resulted in a prolonged bleeding time. The patient was removed from
aspirin and the bleeding returned to normal.

SUMMARy Points • Secondary hemostasis is composed of fibrin clot for-


mation and fibrin clot lysis.
• Hemostasis depends on a system of checks and bal-
ances between thrombosis and hemorrhage that • Platelet aggregation is mediated by von Willebrand’s
involve procoagulants and anticoagulants. factor (vWF) and platelet glycoprotein Ib (GP1b).
• The systems involved in hemostasis are the vascular • Platelets are small discoid cell fragments that are
system, platelets, coagulation system, and fibri- synthesized in the bone marrow and stimulated by
nolytic system. the hormone thrombopoietin.
• Primary hemostasis is composed of platelet function • There are four phases to platelet function at the site
and vasoconstriction. of injury: platelet adherence to collagen, platelet
Copyright © 2007 by F. A. Davis.

242 PART IV • Hemostasis and Disorders of Coagulation

aggregation, platelet granule release, and stabiliza- • The key components of the fibrinolytic system are
tion of the clot. plasminogen, plasminogen activators, plasmin, fib-
• Coagulation factors are produced in the liver with rin, fibrin degradation products, and inhibitors of
the exception of a portion of factor VIII, produced in plasminogen and plasmin.
the endothelial cells. • Streptokinase, urokinase, and tissue plasminogen
• The traditional coagulation pathway is divided into activator are activators of the plasmin-plasminogen
the intrinsic, extrinsic, and common pathways. system.
• The extrinsic pathway is monitored by the pro- • Tissue plasminogen activator is available as a phar-
thrombin time, while the intrinsic pathway is moni- macological product to break up pathologically
tored by the partial thromboplastin time. formed clots.
• The intrinsic pathway is initiated by factor XII and • Serine protease inhibitors and the protein C path-
surface contact with the endothelial cells. way are the major physiologic inhibitors of coagula-
• Tissue factor pathway inhibitor is able to block the tion.
activity of the tissue factor: factor VII complex soon • The kinin system is activated by factor XII and
after it becomes active. contributes to vascular permeability.
• Plasma fibrinogen activated by thrombin results in a • The complement system once activated may con-
stable fibrin clot. tribute to the release of procoagulant material.

CASE STUDY
A 15-year-old boy with chronic strep throat has presented with excessive bruising. His coagulation results were as fol-
lows:
PT 15.5 seconds (Reference range, 10.8 to 13.5)
aPTT 42.1 seconds (Reference range, 28.5 to 35.5)
Platelets 325,000 (Reference range, 150,000 to 400,000)
Bleeding 5 minutes (Reference, 8 minutes)
Which COAGULATION tests ARe ABNORMAL, AND how should this PHYSICIAN proceed in his trEATMENT of this PATIENT?
Insights to the Case Study
In this case, two parameters, the PT and aPTT, are elevated. The patient is not bleeding, but he shows a history of recent
bruising. Since both the PT and the aPTT are affected, one can assume the problem is in the common pathway, specifi-
cally factors I, II, V, and X. Factor assays could be performed to assess the level of activity of each of these clotting factors;
however, a closer examination into the patient’s history might reveal an additional feature. Since this patient has had
chronic strep throat, it is logical to assume that he has been on long-term antibiotics. Antibiotics may deplete the normal
flora, a source of vitamin K synthesis. Factors II, VII, IX, and X are vitamin K–dependent factors. Vitamin K is the essen-
tial cofactor for the gamma carboxyglutamic acid residues necessary to activate these factors. When vitamin K is in short
supply or depleted, these factors fail to function properly. In our patient, vitamin K can be given by mouth to resume
normal coagulation and correct bruising.

Review Questions
1. The factor with the longest half-life is: a. VIII.
a. II. b. II.
b. V. c. VII.
c. VII. d. X.
d. X.
3. Receptors that are found on the platelets are called:
2. If a patient has a prolonged PT, the patient is most a. glycoproteins.
likely deficient in factor: b. vWF.
Copyright © 2007 by F. A. Davis.

CHAPTER 15 • Overview of Hemostasis and Platelet Physiology 243

c. fibrinogen. 8. If a patient has just a prolonged aPTT, the patient


b. beta-thromboglobulin. may be deficient in the following factors:
a. VIII, X, II, and I
4. Vasoconstriction is caused by several regulatory
b. VIII, IX, XI, and XII
molecules, which include:
c. VIII, X, XI, and XII
a. fibrinogen and vWF.
d. VIII, XI, II, and XII
b. ADP and EPI.
c. Thromboxane A2 and serotonin. 9. The factor that is responsible for stabilizing a
d. Collagen and actomyosin. soluble fibrin monomer into an insoluble
clot is:
5. The vitamin K–dependent factors are: a. II.
a. I, II, V, and X. b. X.
b. II, VII, IX, and X. c. XII.
c. I, VII, V,and VIII. d. XIII.
d. II, IX, XI, and X.
10. An inhibitor of plasmin activity is:
6. The life span of a platelet is: a. tPA.
a. 5 to 8 days. b. PAI-1.
b. 7 to 10 days. c. alpha-2-antiplasmin.
c. 6 to 9 days. d. plasminogen.
d. 9 to 12 days.
11. Protein C and its cofactor protein S proteolytically
7. Alpha granules are found in: inactivate factors:
a. the peripheral zone. a. VIIa and Xa.
b. the sol gel zone. b. Va and VIIIa.
c. organelles. c. IXa and VIIa.
d. membranes. d. VIIIa and XIIa.

 TROUBLESHOOTING
WHAT Do I Do When the COAGULATION tubes must be filled to 90% capacity to preserve a 1:9
SAMPLE Is DRAWN Incorrectly? anticoagulant-to-blood ratio.
Preanalytic variables represent important sources of
error in patient testing and accuracy of results. In TRANSPORT AND HANDLING of Specimens
hemostasis testing, sample integrity is paramount. There are several coagulation proteins that are labile,
Areas in which sample integrity are most affected are in namely factors V and VIII. The activity of these factors
phlebotomy practices, transport and handling of speci- will be lost if the sample is not tested in an appropriate
mens, choice of coagulation tubes, and patient vari- time span. For maximum activity, testing should be per-
ables. formed within 4 hours for aPTT and up to 24 hours for
Phlebotomy PRACTICES PT. Plasma can be removed from the sample and stored
The sample must be provided from an atraumatic draw, at –20°C for up to 2 weeks. Additionally, samples must
on a properly identified patient, and the tube must be be centrifuged for a period of time that enables them to
inverted three to four times for proper mixing of anti- become platelet poor plasma. Platelet poor plasma is
coagulant. The order of draw in coagulation testing is defined as having a platelet count of less than 10,000,
important to avoid contamination of the sample with which depends upon proper centrifugation. If samples
tissue thromboplastin. Therefore, if multiple tubes are are not platelet poor, falsely shortened results may
drawn, the coagulation tube should be last. If only a occur as a result of activation of platelet factor 4. Activa-
coagulation sample is requested and the sample is tion of platelet factor 4 may also occur in heparinized
drawn through a butterfly, then a discard tube should samples that are allowed to sit on red cells for longer
be drawn first. If a syringe is needed for phlebotomy, a than 4 hours. In this case the platelet factor 4 may inac-
needle gauge of 12 to 19 is optimal. Additionally, the tivate the heparin giving a falsely shortened PTT result.

(continued on following PAGE)


Copyright © 2007 by F. A. Davis.

244 PART IV • Hemostasis and Disorders of Coagulation

 TROUBLESHOOTING (continued)
Which Tubes to Use? coagulation sample. For patients who have hematocrits
Most facilities are using blue top tubes, which contain that are >60% (neonated, polycythemia), falsely pro-
3.2% sodium citrate. The reasons for this are many and longed results will occur if the anticoagulant is not
include the fact that this concentration provides a adjusted, since there is too much anticoagulant for
closer osmolality to plasma, has less binding of cal- plasma. For patients who have hematocrits of less than
cium, and provides a more favorable environment for 22%, results will be falsely decreased as a result of too
heparinized samples. little anticoagulant because of increasing plasma vol-
ume. The standard formula for adjusting the volume of
PATIENT VARIABLES
anticoagulant is:
Many variables affect coagulation results such as med-
ication, physical and emotional stress, and patient age
and personal habits. These factors cannot be controlled New volume of sodium citrate =
by the laboratory. A patient’s hematocrit, however, is (1.85 × 10)—3 × (100 — Hct) ×
something that can be adjusted for when drawing a volume of sample

WORD KEY 3. Schetz M. Coagulation disorder in acute renal failure.


Kidney Int S53(suppl 66):96–101, 1998.
Anticoagulant • Delaying or preventing blood 4. Harmening D. Clinical Hematology and Fundamentals
coagulation of Hemostasis, 3rd ed. Philadelphia: FA Davis, 1997.
5. Turgeon ML. Clinical Hematology, Theory and Proce-
Fibronectin • Protein involved in wound healing and cell dures, 2nd ed. Boston: Little, Brown and Co, 1993.
adhesion
6. McKenzie SB. Textbook of Hematology. Baltimore:
Lumen • Space within an artery, vein, or intestine or tube Williams and Wilkins, 1996.
Morbidity • State of being diseased
7. Zucker M. The functioning of blood platelets. Sci Am
242:86–89, 1980.
Mortality • Death 8. Kjeldsberg C. Practical Diagnosis of Hematologic
Polymerize • Process by which a simple chemical sub- Disorders, 2nd ed. Chicago: ASCP Press, 1995.
9. Plaut D. Platelet Function Tests. ADVANCE for the
stance or substances are changed into a substance of a much
Administrators of the Laboratory, July 2003.
higher molecular weight but with the same proportions
10. Southern D, Leclair S. Platelet Maturation and Func-
Thrombolysis • Breaking up of a clot tion. In: Rodak B, ed. Diagnostic Hematology. Philadel-
Transaminase • Aminotransferase (an enzyme)
phia, WB Saunders, 1995.
11. Kolde H.-J. Haemostasis pentapharm. Basel: 2001.
Vasoconstriction • Decrease in the diameter of the blood 12. Rodgers G. Hemostasis Case Studies. Chicago: ASCP
vessels that decreases the blood flow Press, 2000.
13. Ogedegbe HO. An overview of hemostasis. Lab Med 33:
Viscosity • State of being sticky or gummy December 2002.
14. Fass D. Overview of Hemostasis, Mayo Clinic Coagula-
References tion Conference, Mayo Clinic, August 2004.
1. Owen CA. A History of Blood Coagulation. Rochester, 15. Kandrotis RJ. Pharmacology and pharmacokinetics of
MN; Mayo Foundation for Medical Education and antithrombotic agents. Clin Appl Thromb/Hemost
Research, 2001. 3:157–163, 1997.
2. Hougie C. Fundamentals of Blood Coagulation in Clinical 16. Bick R. Disorders of Thrombosis and Hemostasis.
Medicine. New York: McGraw-Hill Book Company, 1963. Chicago: ASCP Press, 1991.
Copyright © 2007 by F. A. Davis.

Quantitative and Qualitative


16 Platelet Disorders

Betty CIESLA

Quantitative Disorders of Platelets Objectives


Thrombocytopenia Related to Sample Integrity/ After completing this CHAPTER, the student will be ABLE to:
Preanalytic Variables
Thrombocytopenia Related to Decreased Production 1. Define the QUANTITATIVE PLATELET disorders.
Thrombocytopenia Related to Altered Distribution 2. Identify the types of bleeding that are seen in
of Platelets platelet disorders.
Thrombocytopenia Related to the Immune Effect 3. List four laboratory tests that are helpful in
of Specific Drugs or Antibody Formation evaluating platelet disorders.
Thrombocytopenia Related to Consumption of 4. State how preanalytic variables may affect the
Platelets platelet count.
Thrombocytosis
5. Describe three characteristics of the qualitative
Inherited Qualitative Disorders of Platelets platelet disorders von Willebrand disease,
Disorders of Adhesion Bernard Soulier, and Glanzmann’s thrombas-
thenia.
Platelet Release Defects
6. Identify drugs that are implicated in immune
Acquired Defects of Platelet Function thrombocytopenia.
Vascular Disorders Leading to 7. Evaluate conditions that may cause
Platelet Dysfunction thrombocytosis.
8. Compare and contrast acute versus chronic
idiopathic thrombocytopenic purpura.
9. Describe the effect of ristocetin on platelet
aggregation.
10. Define hemolytic uremic syndrome in terms of
incidence, key clinical features, and patient
management.
11. Define thrombotic thrombocytopenic PURPURA in
terms of incidence, key clinical features, and
severity.
12. Describe platelet abnormalities due to
acquired defects: drug induced, nonimmune,
or vascular.

245
Copyright © 2007 by F. A. Davis.

246 PART IV • Hemostasis and Disorders of Coagulation

QUANTITATIVE DISORDERS Thrombocytopenia Related


OF PLATELETS to Decreased Production
A normal platelet count is 150 to 450 × 109/L. In this Any condition that leads to bone marrow aplasia or a
range, an individual will have properly functioning lack of megakaryocytes, the platelet forming cell, will
platelets that assist in the coagulation process by lead to a thrombocytopenia. Most patients with
creating a platelet plug and stimulating the formation leukemia will exhibit a thrombocytopenia as a result of
of a solid fibrin clot. A decrease in platelet count will infiltration of the bone marrow with blast cells. Blasts of
cause bleeding from the mucous membranes such as any cellular origin crowd out normal bone marrow ele-
gum bleeding (gingival bleeding), nose bleeding (epis- ments leading to thrombocytopenia. Defects in platelet
taxis), extensive bruising (ecchymoses), or petechiae synthesis can occur in the megaloblastic anemias that
(pinpoint hemorrhages). A patient with a platelet count show a pancytopenia, a decrease in all cell lines. Cyto-
of 60,000 will bleed in surgery and a patient with toxic agents or chemotherapy usually interferes with
a platelet count of 30,000 may have petechial bleeding the cell cycle, thereby reducing the number of active
At less than 5000 platelets, there is a risk of bleeding platelets. Patients undergoing chemotherapy are care-
into the central nervous system. Laboratory tests fully monitored for platelet count and may need to be
that are helpful in the evaluation of platelet function given platelet support if the count drops too far below
are the evaluation of the peripheral smear for platelet 20.0 ×109/L.
number and morphology, the bleeding time test (or Megakaryocytic function is impaired during the
similar platelet function tests), platelet aggregation by infectious process. Infections with several viral agents
one of several methods, or other methods that assess such as cytomegalovirus, Epstein-Barr virus, varicella,
platelet function and aggregation. Thrombocytopenia and rubella and certain bacterial infections may cause a
or a decreased platelet count is caused by a number thrombocytopenia. The mechanism at work in viral
of factors. Decreased production of platelets or infections is thought to be megakaryocytic suppression;
increased destruction of platelets usually accounts for in bacteria, the mechanism is direct toxicity of circulat-
the pathophysiology of most QUANTITATIVE defects ing platelets.2
in platelets. Additionally, sample related conditions or
preanalytic variables may lead to falsely decreased
Thrombocytopenia Related to
platelet counts. Altered Distribution of Platelets
Thrombocytopenia Related to Sample The normal spleen holds one third of the platelet
Integrity/Preanalytic Variables volume. Several hematological conditions may lead to
an enlarged spleen as part of their pathological process:
Coagulation samples are drawn into blue top tubes con-
the myeloproliferative disorders, extramedullary hema-
taining sodium citrate. Sodium citrate anticoagulates a
topoiesis, and hemolytic anemias. As the spleen
specimen by binding calcium in a 1:9 anticoagulant-to-
enlarges, blood pools in this organ withholding
blood ratio. Sample tubes must be at least 90% full and
platelets from the peripheral circulation. If the organ is
the phlebotomy must be nontraumatic. The blue top
removed, then large numbers of platelets may spill into
tube must be inverted at least three or four times for
the circulation, causing possible thrombotic complica-
proper mixing of the anticoagulant. If this does not hap-
tions.3 An additional scenario in which platelet distri-
pen, there is a possibility of small clots being formed on
bution is altered is in massive transfusion. Once the
the top of the tube. Platelet satellitism is another condi-
total blood volume (10 units) has been replaced with
tion related to samples that may give a falsely decreased
two or three volume exchanges, the platelet and the
platelet count. First reported in 1963,1 this condition is
coagulation factors become diluted leading to a tran-
an in vitro phenomenon in which the patient’s platelets
sient thrombocytopenia.4
rosette around segmented neutrophils, monocytes, and
bands. This phenomenon occurs only in EDTA (ethyl-
enediaminetetraacetic acid) samples and produces a Thrombocytopenia Related to the
pseudo-thrombocytopenia unrelated to medication or Immune Effect of Specific Drugs
any other disease state (see Fig. 10.21). If platelet satel- or Antibody Formation
litism is observed on the peripheral smear, the sample Drug-induced immune thrombocytopenia produces a
should be redrawn in sodium citrate and cycled reduced platelet count that can be severe and danger-
through the automated hematology counter for a more ous. Several drug classifications are particularly relevant
accurate platelet count. and include quinines, NSAIDs (nonsteroidal anti-
Copyright © 2007 by F. A. Davis.

CHAPTER 16 • Quantitative and Qualitative Platelet Disorders 247

inflammatory drugs), sulfonamides, and diuretics.5 The may be increased in the marrow; however, they
mechanism for thrombocytopenia is 2-fold. On the one are poorly functioning.2 There are two types of ITP:
hand, ingestion of the drug will cause an antidrug anti- chronic and acute. Patients with acute ITP are usually
body formation that will bind to a glycoprotein on the children between the ages of 2 and 6 who have just
platelet surface and be removed by the reticuloendothe- recovered from a viral illness.2 The platelet counts may
lial system (RES). The second mechanism involves the drop precipitously, some as low as 20 ×109/L. In this
drug combining with a larger carrier protein to form an range, the child will usually show bruising, nose
antigen that triggers an antibody response and subse- bleeding, or petechiae but will not usually show life-
quent platelet destruction, potentially in the spleen. threatening hemorrhage. Fortunately, this low platelet
The incidence of drug-induced thrombocytopenia is 10 count usually resolves in less than 6 weeks as the child
cases per 1 million.5 fully recovers from the viral illness. Treatment, if neces-
Additionally, there are two rare conditions in which sary, may consist of intravenous immunoglobulin
thrombocytopenia may be quite dramatic. Fortunately, (IvIg or WinRho, anti–D immune globulin), splenec-
these are rare. The first, posttransfusion purpura (PTP), tomy, or platelet transfusion.2 Chronic ITP, on the other
occurs after transfusion of platelet-containing products hand, shows a platelet count between 30 and 60 ×
in which the recipient has developed an antibody. The 109/L in a much older age range of between 20 and 50
antibody is directed against an antigen on the platelet years of age. For these patients, an IgG antibody is
Pl1A, a primary platelet antigen, and therefore when produced that coats the platelets, causing them to be
donor platelets are transfused containing this antigen, sequestered and subsequently destroyed in the spleen.
the platelets are coated and removed by the spleen. The Splenomegaly is a frequent physical symptom. Most
resultant thrombocytopenia is quite long lasting, and patients are treated with prednisone, which suppresses
treatment is directed toward delaying antibody produc- the antibody response, increases the platelet count,
tion. The second condition, neonatal isoimmune throm- and decreases the hemorrhagic episodes. For those
bocytopenia, occurs as a result of maternal antibody who are nonresponsive, anti-CD20, Rituximab, has
made against a previous exposure to platelet antigens been shown to provide a sustained platelet response.6
from an earlier pregnancy. The antibody is usually Splenectomy is a therapeutic option, but it must be
directed against the Pl1A. Since this antibody can cross carefully considered. Recently, immune thrombocy-
the placenta, it can coat the baby’s platelets in utero. topenia related to infections has been investigated.
Infants born to mothers carrying these antibodies will Patients infected with HIV, hepatitis C, and HELICOBACTER
often show a normal platelet count initially but within pylori show thrombocytopenia at some point during
days they will develop petechiae and skin hemorrhages their disease. The precise mechanism, thought to be
with decreasingly low platelet counts. Infants are care- immune derived, is under study.7 Table 16.1 compares
fully observed and treatment is only begun when there is acute and chronic ITP.
a risk of central nervous system hemorrhage.2

Thrombocytopenia Related
to Consumption of Platelets Table 16.1  Chronic and Acute
Hematological conditions studied under this category Idiopathic Thrombocy-
usually include idiopathic thrombocytopenia purpura, topenic Purpura
thrombotic thrombocytopenic purpura, and hemolytic
uremic syndrome. In these conditions, excessive clots Acute Idiopathic Chronic Idiopathic
are formed throughout the body, which consume Thrombocytopenic Thrombocytopenic
platelets. Each of these conditions is serious and can Purpura Purpura
produce significant life-altering complications. Age Young children Adults
Prior History of rubella, No prior history
Idiopathic (Immune) Thrombocytopenic Purpura infection rubeola, or
Patients with idiopathic (immune) thrombocytopenic chickenpox
purpura (ITP) show a decreased platelet count that Platelet <20,000 30,000 to 80,000
is thought to be a result of immune destruction of count
platelets. In 66% of cases, the antibody is an auto- Duration 2 to 6 weeks Months to years
antibody directed against specific sites on glycoprotein Therapy None Steroids, splenectomy
(GP) IIb-IIIa or GP Ib-IX. Additionally, megakaryocytes
Copyright © 2007 by F. A. Davis.

248 PART IV • Hemostasis and Disorders of Coagulation

Thrombotic Thrombocytopenic Purpura


This devastating platelet disorder described by
Moschowitz in 1925 is acute and nonpredictable. More
prevalent in women than in men, thrombotic thrombo-
cytopenic purpura (TTP) can occur in women postpar-
tum or near delivery8 in those who have suffered from
other immune disorders like SLE (systemic lupus ery-
thematosus), and in those with previous viral infections
or gastric carcinomas. Platelet counts are less than 20 ×
109/L but other coagulation testing such as PT and PTT
are within reference range. Platelet thrombi are dis-
persed throughout the arterioles and capillaries subse-
quent to the accumulation of large von Willebrand
Figure 16.1 Schistocyte.
multimers made by the endothelial cells and platelets.
The etiology for this pathological accumulation of mul-
timers and subsequent thrombocytopenia is thought to to 82% presently.11 Few hospital facilities provide
be related to a deficiency of ADAMTS-13, a large metal- plasma exchange capabilities. Specialty teams of med-
loprotease.9,10 This protein is responsible for cleaving ical professionals using equipment designed for plasma-
large von Willebrand factor (vWF) multimers into pheresis are usually called upon. Timing is essential to
smaller proteins. Large vWF multimers have increased the patient’s welfare and long-term recovery. Recovery
binding sites for platelets as compared to smaller vWF for TTP patients has improved in the past decade, with
portions. If large vWF are not cleaved and allowed to more than 80% surviving.11
circulate, then excessive platelet clots may be formed.
Schistocytes are seen in the peripheral smear and are
Hemolytic Uremic Syndrome
directly related to shear stress as fragments of red cells
are removed once the cells try to sweep past the thrombi. Hemolytic uremic syndrome (HUS) frequently occurs in
Patients experience a severe anemia with a high level children between the ages of 6 months and 4 years. This
of hemolysis, with increased lactate dehydrogenase clinical condition mimics TTP, with the exception that
(LDH). Some of the hemolysis may be intravascular with the renal damage is more severe. The kidney is the pri-
hemoglobinuria. Decreased haptoglobin may be seen. mary site of damage by the toxin ESCHERICHIA coli
Microangiopathic hemolytic anemia (MHA) is the term O157:H7 or the SHIGELLA toxin.11 The endotoxin pro-
used to describe this process of severe anemia combined duced by this particular strain of E. coli inevitably leads
with schistocytes (Fig. 16.1). Oftentimes patients will to cell death, particularly in the renal environment,
present with neurological complications. These compli- where platelet thrombi predominate.12 A child may ini-
cations may include mild presentations of visual impair- tially present with bloody diarrhea and vomiting; how-
ment and intense headache ranging to more severe ever, hemolytic anemia, thrombocytopenia, and renal
presentations such as coma and paresthesias. Renal failure soon follow. Patients may also experience fever
dysfunction may occur, and patients with renal impair- and abdominal pain, and the hemolytic anemia is
ment experience an increased protein and possibly some microangiopathic with schistocytes present. The illness
blood in the urine. Treatment for TTP patients presents a in children is usually self-limiting once the toxin is elim-
dilemma for most physicians, as they watch their inated from the body; however, there have been reports
patients spiral rapidly downhill. Diagnosis is often diffi- of patients relapsing. Renal dialysis may be needed for
cult and often represents a diagnosis of exclusion. Once those children suffering from acute renal failure. Most
made, the patient’s condition has usually dramatically children make a complete recovery, but some have
worsened. Corticosteroids are often used in conjunction residual kidney problems into adulthood. HUS may
with plasma exchange, a dramatic procedure performed occur in adults, but it is more similar to TTP in course of
over a 3- to 5-day period in which the patient’s plasma is disease (Table 16.2).
removed and replaced by ABO matched fresh frozen Disorders such as heparin-induced thrombocy-
plasma that is cryoprecipitate poor (lacking fibrinogen topenia and disseminated intravascular coagulation
and vWF). The use of plasma exchange has dramatically also lead to thrombocytopenia. These will be covered in
improved the survival rate from a low of 3% before 1960 subsequent chapters.
Copyright © 2007 by F. A. Davis.

CHAPTER 16 • Quantitative and Qualitative Platelet Disorders 249

intrinsic defect of platelets. Many of these disorders


Table 16.2  Hemolytic Uremic (with the exception of vWD) are rare, and in most cases
Syndrome Versus the bleeding time is prolonged. Often, the qualitative
Thrombotic Thrombocy- defects are separated into disorders of adhesion and
topenic Purpura platelet release or storage pool defects.

Hemolytic Thrombotic
Uremic Thrombocytopenic Disorders of Adhesion
Syndrome Purpura von Willebrand’s Disease
Platelet count <20,000 <20,000 The most important disease of platelet adhesion is von
Organ(s) Kidney Neurological mani- Willebrand disease (vWD). Discovered in 1926 by Dr.
affected festations Eric von Willebrand, vWD is the most prevalent inher-
Kidney ited bleeding disorder worldwide, affecting 1% to 3% of
Age group Children Adults (more females the world population by conservative estimates. In ran-
than males) dom studies of children investigated for epistaxis and
Symptoms Fever, bloody Fever, headaches women investigated for menorrhagia, vWD was the
diarrhea Visual impairment, most frequent cause of bleeding.14,15 von Willebrand
MHA with schis- coma initially described a family of 12 children of which 10
tocytes MHA with schisto- had excessive nosebleeds, gum bleeds, and menorrha-
cytes gia. One of the youngest girls died at age 13 during her
Treatment Renal dialysis, Plasmapheresis fourth menstrual cycle of uncontrollable bleeding. vWD
blood transfu- is an autosomal dominant disorder marked by easy
sions bruising, nosebleeds, heavy menses, and excessive
bleeding after tooth extraction or dental procedures.
MHA, microangiopathic hemolytic anemia.
Type O individuals have a lower plasma concentration
of vWF than other blood types. For many patients, the
variabilities in clinical symptoms and laboratory presen-
Thrombocytosis tations have contributed to the underdiagnosis of this
Thrombocytosis is defined as a platelet count greater disorder. Women may represent a significant yet under-
than 450 × 109/L. The cause for an increased platelet served subset of individuals affected by vWD, since
count may be primary or secondary. A primary throm- menorrhagia is a frequent presenting feature of this dis-
bocytosis is seen in the myeloproliferative disorders (see ease. According to Luscher, vWD may be the underlying
Chapter 12), in which case platelets are high in number cause in 9% to 11% of cases of menorrhagia,16 yet it is
but have an impaired function. Of all of the myelopro- often not considered as a possible diagnosis by obstetri-
liferative disorders, essential thrombocythemia has the cians and gynecologists.
highest platelet value, at times exceeding 1 million As a disease entity, vWD is fairly complex with few
platelets. Secondary causes of thrombocytosis include clear-cut and consistent diagnostic clues. The basic
acute and chronic blood loss, chronic inflammatory dis- pathophysiology in vWD is a qualitative or quantitative
eases, postsplenectomy, and iron deficiency anemia. In defect in vWF. vWF is a large multimeric glycoprotein
these cases, the platelet function is normal, although the derived from two sources: endothelial cells and
increase in platelet numbers may last days to weeks. In megakaryocytes (Table 16.3). This protein is coded for by
severe iron deficiency anemia, the platelet count may chromosome 12 and is carried into plasma circulation
increase to 2 million, as a result of marrow stimula- by factor 8, one of the clotting factors. vWF serves as an
tion.13 Once iron therapy is initiated, the platelet count intermediary for platelet adhesion, providing a receptor
usually returns to normal. molecule for GP Ib of the platelets and the subendothe-
lium. With this platform in place, platelets, once acti-
vated by injury, adhere to the subendothelium forming a
INHERITED QUALITATIVE
platelet plug, recruiting more platelets to the site of
DISORDERS OF PLATELETS
injury and eventually leading to platelet aggregation and
Inherited qualitative platelet disorders are those in the formation of an insoluble fibrin clot. Without a fully
which platelet function is impaired usually due to an functioning vWF, platelet adhesion is impaired. Addi-
Copyright © 2007 by F. A. Davis.

250 PART IV • Hemostasis and Disorders of Coagulation

tionally, vWF binds GP IIb/IIIa. There are three PRIMARY


levels of vWD: type 1, type 2, and type 3. Seventy per- Table 16.4  Primary von Willebrand’s
cent of all individuals with vWD have the type 1 disor- Disease Derivatives*
ders characterized by an abnormal bleeding time and an
increased PTT in most patients. Type 2 vWD is the result Type 1 Type 2 Type 3
of a qualitative defect of wVF, and type 3, the rarest type,
is characterized by a total absence of vWF multimers Frequency 70% to 80% 15% to 20% Rare
and is autosomal recessive in its presentation. Type 2 Genetics Autosomal Autosomal Autosomal
vWD has many subtypes: type 2A, type 2B, type 2M, dominant dominant recessive
and type 2N. See Table 16-4 for a description of vWD Bleeding time  or N  
and its variants. The vWF protein can be measured by PTT  or N  or N 
several methods; those that assess its role in adhesion, RIPA  or N  
its secondary role in aggregation, and its role in clotting vWF agn.   Absent
factor activity. Ristocetin co-factor activity is the single
best predictive assay17 and relies on the use of reagent *Secondary vWD variants include types 2A, 2B, 2M, and 2N: these
platelets rather than patient’s platelets during ristocetin- are not discussed.
induced aggregation studies. Table 16.3 gives a descrip-
tion of a typical testing profile for vWD.
in Chapter 15, platelet glycoproteins play a significant
Treatment is usually tailored to the particular type
role in hemostasis. The receptor for vWf is GP Ib-IX.
or subtype of vWD. Some of the products that may be
This complex, Ib-IX, serves as a site for thrombin bind-
considered are desmopressin acetate (DDAVP), which
ing as well as regulating platelet shape and reactivity.18
causes the release of endothelial vWF. DDAVP may be
GP IIb and IIIa are receptors for fibronectin (an adhe-
given as an injectable agent or as a nasal spray, which
sive protein for platelets), vWF, fibrinogen, and factors
makes it portable and convenient. For patients who
V and VIII. BSS is inherited as an autosomal recessive
are nonresponsive, vWF can be raised by giving high
disorder, with near normal amounts of GPIb in het-
purity factor 8 products that contain a sufficient amount
erozygotes. If the disorder is inherited homozygously,
of vWF.
however, moderate or severe bleeding may occur. Epi-
staxis, gingival bleeding, menorrhagia, and purpura are
Bernard Soulier Syndrome the usual bleeding manifestation. Additionally, there is a
Bernard Soulier syndrome (BSS) is a rare adhesion thrombocytopenia with giant platelets observed on the
defect of platelets that involves the GP Ib/IX complex. peripheral smear. Ristocetin-induced platelet aggrega-
Once an injury has occurred, vWF acts as a medium tion is absent in BSS patients since there are no recep-
through which the platelet membrane GP Ib has a tors to bind to vWF, a key ingredient in ristocetin
receptor that allows its binding to collagen. As indicated induced platelet aggregation. Platelet aggregation
with other agents such as epinephrine, thrombin, and
collagen appears normal. The bleeding time test is
prolonged.
Table 16.3  Basic Test Profile Platelet transfusions are the treatment of choice for
active bleeding but they should be used prudently to
for vWD
prevent the stimulation of platelet antibodies. To date,
• Platelet count—measured by automated methods over 30 mutations of the glycoproteins involved in the
• PTT—measures anticoagulant portion of the factor GP Ib-IX complex have been described.19
VIII molecule
• Bleeding time—measures adhesion of platelets to site Glanzmann’s Thrombasthenia
of injury Glanzmann’s thrombasthenia (GT) is an autosomal
• vWF activity—measured by ristocetin-induced
recessive disorder, first described in 1918, most often
platelet aggregation (RIPA)
associated with consanguinity. Homozygous individu-
• vWF antigen—measured by immunoassay
als may experience variable bleeding patterns. When
bleeding does occur, it is usually from birth as umbilical
Most patients will have variable test results. It is recommended
that this test profile be performed multiple times within a time cord or circumcisional bleeding and may proceed to gin-
period to aid in diagnosis. gival bleeding, purpura, or prolonged bleeding from
Copyright © 2007 by F. A. Davis.

CHAPTER 16 • Quantitative and Qualitative Platelet Disorders 251

minor cuts or childhood events. The defect in GT is a have no radial bones and other skeletal and car-
deficiency or abnormality of GP IIb and IIIa. These gly- diac abnormalities. Thrombocytopenia is seen
coproteins serve as the intermediary for fibrinogen bind- in most patients.
ing to platelets, a necessary step in platelet aggregation. GRAY PLATELET syndrome: Platelets show a lack of
Aggregation cannot occur if GP IIb/IIIa is absent or if alpha granules and are noted in the peripheral
there is an absence of fibrinogen or calcium.20 Patients smear as appearing larger, having a gray or blue-
with GT will have a prolonged bleeding time, normal gray color. Patients may show thrombocytope-
platelet count and morphology, and abnormal aggrega- nia, bleeding tendencies, and bruisability.
tion with all aggregating agents except ristocetin.
Ristocetin-induced aggregation depends upon the inter-
action of vWF and platelet GP Ib. The GP IIb/IIIa com- ACQUIRED DEFECTS
plex does not play a role in this type of aggregation. OF PLATELET FUNCTION
Treatment in GT depends upon the severity of the bleed- Included in this category of platelet defects are those fac-
ing episode. Platelet transfusions may be considered but tors that are external to the platelet and that are nonim-
HLA-matched or ABO-matched transfusion may reduce mune, such as drug-related platelet abnormalities,
the possibilities of platelet alloimmunization. Oral con- extrinsic platelet abnormalities, or as a sequel to an
traceptives may be used to control menorrhagia, and underlying disorder. Of all the drugs that affect platelet
agents such as ethyleneaminocaproic acid (EACA) are function, aspirin is the most popular. Ingestion of
effective topical thrombin-inducing agents for proce- aspirin irreversibly inhibits cyclooxygenase (COX-1
dures such as tooth extractions.21 inhibitors) by inhibiting the formation of prostaglandin
synthesis. Both of these chemicals are necessary for the
Platelet Release Defects production of thromboxane A2, a potent platelet aggre-
gator. Without the production of proper amount of
Once platelets adhere to an injured surface, the con- thromboxane A2, platelet aggregation is impaired. This
tents of the platelets are released. Platelets contain alpha effect lasts for the entire life span of the platelet, 7 to 10
and dense granules, which are highly metabolic sub- days, and patients on aspirin will show a prolonged
stances containing procoagulant materials, vasocon- bleeding time. Patients should be queried about their
strictors ATP and ADP. The disorders that are described aspirin use or use of aspirin-containing products prior to
are inherited, usually have abnormal secondary phases any surgical event, elective or nonelective, to avoid any
of platelet aggregation, and show postoperative bleed- unexpected bleeding complications. The effect of
ing combined with menorrhagia and easy bruisability. aspirin on platelets is fairly rapid, occurring 45 minutes
In most of these disorders, the bleeding time is abnor- after ingestion.22 Additionally, aspirin as an antiplatelet
mal, but the platelet count may be normal. agent is used as a preventive for patients susceptible to
HerMANSKY-PUDLAK syndrome: An autosomal reces- strokes, heart attacks, or other cardiovascular events.
sive disorder characterized by a severe defi- Other drugs such as NSAIDs and the class of COX-2
ciency of dense granules. Patients show inhibitors such as naproxen and ibuprofen may affect
albinism and may have hemorrhagic events. platelet function. Certain antiplatelet agents such as
CHEDIAK-HIGASHI syndrome: An autosomal recessive ticlopidine and clopidogrel inhibit fibrinogen binding to
disorder, in which patients show albinism and GP IIb and IIIa. The plasma expander dextran also alters
giant lysosomal granules in neutrophils. Not platelet function. The coating of platelets with dextran
only are the white cells in these patients qualita- gives an antiplatelet effect by inhibiting the action of the
tively flawed, but platelet release is impaired. platelet membrane and its surface receptors.
Patients show frequent infections because of Platelet function may also be impaired by plasma
impaired phagocytic ability and death usually conditions that are less than favorable to the platelet. In
occurs in childhood. Patients manifest throm- most cases, disorders in platelets are secondary to the
bocytopenia and increased liver and spleen. main disorder but may not be present in the initial pres-
Wiskott-Aldrich syndrome: This is an X-linked entation. Conditions that may lead to disturbed platelet
recessive disorder in which patients show severe function include uremia due to renal disease and the
eczema, recurrent infections, immune defects, paraproteinemias such as multiple myeloma and
and thrombocytopenia. ..
Waldenstrom’s macroglobulinemia. The pathophysiol-
ThrOMBOCYTOPENIA with ABSENT RADII (TAR): A ogy involved in the platelet defect in these acquired dis-
rare disorder of the skeletal system in which orders is not clear-cut. Patients with renal disease are
patients
Copyright © 2007 by F. A. Davis.

252 PART IV • Hemostasis and Disorders of Coagulation

VASCULAR DISORDERS LEADING


TO PLATELET DYSFUNCTION
Skin, collagen, and blood vessels are essential elements
in the hemostatic system. Any abnormality, inherited or
acquired, in any one of these components of the vascu-
lar system will lead to mucosal bleeding such as: pur-
pura, petechia, ecchymoses, or telangiectasia (Fig.
16.3). Tests of platelet function and numbers in these
individuals will be normal. Senile purpura is a condi-
tion of aging in which skin loses its elasticity. Often-
times, older individuals will bruise more easily and
more prominently. Allergic purpura is seen in rare
..
childhood disorders such as Henoch-Schonlein pur-
pura, an immune complex disease that involves the
Figure 16.2 Purpura. skin, gastrointestinal tract, heart, and central nervous
system. The purpura is often seen in the lower extremi-
ties. Purpura may occur due to infectious agents such as
known to exhibit purpura (Fig. 16.2), epistaxis, and meningococcemia, Rocky Mountain spotted fever,
hemorrhage at times. A few of the factors involved in staphylococci, or streptococcal infections.23 Conditions
platelet dysfunction in uremia include decreased such as amyloidosis, vitamin C deficiency (scurvy), or
thromboxane synthesis, decreased adhesion, decreased Cushing syndrome may result in purpura.
platelet release, and decreased aggregation. Most of Inherited collagen disorders provoking the forma-
these patients will undergo peritoneal or hemodialysis, tion of purpuric lesions or telangiectasia are hereditary
which usually improves platelet function. hemorrhagic telangiectasia (Osler-Weber-Rendu dis-
.. ease), an autosomal dominant disorder of the blood ves-
Multiple myeloma and Waldenstrom’s macroglob-
ulinemia represent a group of plasma cell disorders in sels. In this condition, small pinpoint hemorrhagic
which a normal immunoglobulin is produced in excess lesions are seen on the tongue, roof of the mouth,
leading to hyperviscosity syndrome and a parapro- palate, face, and hands.23 In addition to these lesions,
teinemia. Platelets circulating in abnormal amounts of nosebleeds are prominent and the lesions in general
protein are unable to fully participate in the activation become more fragile with age. Kasabach-Merritt syn-
of coagulation factors and in the fibrin formation. drome is a rare congenital disorder featuring giant
Patients will show a prolonged bleeding time and may hemangiomas,23 bleeding, and thrombocytopenia.
Hemangiomas may be found on the liver, skin, or
show postoperative bleeding and ecchymoses. Table
16.5 displays a list of drugs that affect platelet function. spleen, and they are deep and bleed easily and pro-
fusely. DIC may develop if thromboplastic substances
are released when the blood vessels burst.

Table 16.5  Modified List of


Drugs that Affect
Platelet Function
• Penicillin
• Ampicillin
• Carbenicillin
• Cephalosporin
• Ticlopidine
• Clopidogrel
• Ibuprofen
• Aspirin
• Nitroglycerin
• Propranolol
• Nitroprusside
Figure 16.3 Telangiectasia.
Copyright © 2007 by F. A. Davis.

CHAPTER 16 • Quantitative and Qualitative Platelet Disorders 253

COND ENSED CASE


A 14-year-old girl had a tooth extracted and was noted to have unexpected bleeding following extraction. She bled for
24 hours before the bleeding could be stopped. The dentist recommended that she have a hematology evaluation for
the unexpected bleeding. WHAT questions concerning FAMILY history should be ASKED, AND WHAT BASELINE COAGULATION
tests should be considered?
Answer
This patient is exhibiting signs of mucosal bleeding, the type of bleeding seen in platelet adhesion defects such as vWD
and BSS. The family of the patient should be asked about the bleeding history of family members, such as umbilical
cord bleeding, circumcision bleeding, bleeding from minor cuts and abrasions, or gum or nose bleeding. The patient’s
mother revealed that her sibling had serious bleeding after a tonsillectomy procedure. This fact points to an autosomal
defect. Routine studies that should be ordered are bleeding time (platelet function assay), PT, and aPTT. Factor assay
should be considered if the PT or PTT are prolonged.

SUMMARy Points • vWD is the most common inherited qualitative


• A normal platelet count is 150 to 450 ×109/L. platelet disorder, affecting 1% to 3% of the world’s
population.
• Decreased platelet counts will lead to mucosal mem-
brane bleeding such as gingival bleeding, epistaxis, • There are three primary types of vWD: type 1, type
purpura, and petechiae. 2A, and type 3.
• Bernard Soulier syndrome is a platelet adhesion
• Preanalytic variables that may lead to thrombocy-
defect in which glycoprotein Ib is decreased or
topenia include improper mixing of tubes, improper
absent.
anticoagulant used, and improper amount of sample
collected. • Glanzmann’s thrombasthenia is a defect of platelet
aggregation that shows an absence of glycoprotein
• Acute idiopathic thrombocytopenia purpura is
IIb/IIIa.
often a condition of children recovering from
viral illness who show a dramatic drop in platelet • Platelets from patients with vWD disease and
count. Bernard Soulier syndrome will NOT aggregate with
ristocetin.
• Chronic idiopathic thrombocytopenia purpura
• Aspirin impairs platelet function by interfering with
occurs in adults as a result of an IgG antibody pro-
the synthesis of thromboxane A2, a potent platelet-
duced against platelets.
aggregating agent.
• Thrombotic thrombocytopenic purpura (TTP) and
• The platelet release function is impaired in the
hemolytic uremic syndrome (HUS) are consumptive
inherited disorders: Chediak-Higashi, Hermansky-
disorders of platelets.
Pudlak, Wiskott-Aldrich, gray platelet syndrome,
• Individuals with TTP present with fever, a microan- and thrombocytopenia with absent radii syndrome.
giopathic hemolytic anemia, neurological complica- • External conditions that alter platelet function
tions, thrombocytopenia, and renal failure. include drugs, paraproteinemias, uremia, and the
• Individuals with HUS are predominantly children, use of plasma expanders, like dextran.
with fever, bloody diarrhea, microangiopathic • Skin, collagen, and blood vessels are essential ele-
hemolytic anemia, thrombocytopenia, and renal ments in the hemostatic system.
failure.
• Any abnormality, inherited or acquired, in any one
• von Willebrand’s disease (vWD) is a disorder of of these components of the vascular system will lead
platelet adhesion in which von Willebrand factor is to mucosal bleeding such as purpura, petechiae,
decreased or absent. ecchymosis, or telangiectasia.
Copyright © 2007 by F. A. Davis.

254 PART IV • Hemostasis and Disorders of Coagulation

CASE STUDY
A 24-year-old woman was being evaluated by her gynecol- received a diagnosis of a bleeding disorder, it seems likely
ogist for menorrhagia. She gave a history of excessive that she and some of them may have von Willebrand’s
menses since the age of 12. A CBC revealed a microcytic disease, an autosomal dominant disorder. The patient’s
anemia and she began a course of ferrous sulfate therapy. CBC and platelet count is normal; however, the PTT is
Three months later, she had a follow-up visit with her slightly prolonged at 42 seconds. Factor assays for
gynecologist, and although her anemia was being cor- factor VIII and factor IX should be considered. Aggrega-
rected, she still complained of excessive menses. Her tion studies with collagen, ADP, and epinephrine were
physician recommended her for a hematology consult. normal. Ristocetin aggregation was absent and the
When asked about her family history, she revealed that her bleeding time test was abnormal with a result of
brother and mother had recurrent epistaxis and that her 12 minutes (reference range, 3 to 9 minutes). A
first cousin had a postpartum hemorrhage. The consulting preliminary diagnosis of type 1 von Willebrand’s
physician ordered a CBC, PT, PTT, platelet aggregation disease was made pending the result of the vWF:
studies, and bleeding time. BASED upon this PATIENT’s his- AG by immunoassay. The hematologist recommended
tory, WHAT is the most likely outcome of this testing AND contraceptives as a way to control the patient’s menstrual
WHAT ADDITIONAL tests ARe to be considered? bleeding, and the patient was counseled on therapy alter-
natives such as DDAVP should she need dental extrac-
Insights to the Case Study
tions or minor surgery.
This patient gives a strong family history of mucosal
bleeding. Although no member of her family has

Review Questions
1. Which of the following are defects of platelet a. Clotting factor disorder
adhesion? b. Platelet defect
a. Hermansky-Pudlak syndrome c. Thrombosis
b. Glanzmann’s thrombasthenia d. Vascular disorder
c. Bernard Soulier syndrome
5. The presence of thrombocytopenia and giant
d. Wiskott-Aldrich
platelets best describes:
2. Which one of the conditions will produce a a. classic von Willebrand’s disease.
thrombocytopenia due to an altered distribution b. Wiskott-Aldrich
of platelets? c. Glanzmann’s thrombasthenia.
a. Platelet satellitism d. Bernard Soulier syndrome.
b. Iron deficiency anemia 6. Chronic idiopathic thrombocytopenia purpura
c. Splenomegaly (ITP):
d. Chemotherapy a. is found in children.
b. usually spontaneously remits within several
3. One of the main differences between TTP and weeks.
HUS is: c. affects males more commonly than females.
a. neurological involvement. d. involves the immune destruction of platelets.
b. kidney failure.
c. thrombocytopenia. 7. Aspirin prevents platelet aggregation by inhibiting
d. microangiopathic hemolytic anemia. the action of:
a. PF 3.
4. Nose bleeding, deep bruising, and gum bleeding b. GP II.
are usually manifestations of which type of c. TXA2.
coagulation disorder? d. GP Ib.
Copyright © 2007 by F. A. Davis.

CHAPTER 16 • Quantitative and Qualitative Platelet Disorders 255

Consanguinity • Relationships among close blood


relatives
 TROUBLESHOOTING Cryoprecipitate • Product derived from fresh frozen
WHAT Do I Do When PrEOPERATIVE COAGULATION plasma that is rich in factor VIII, von Willebrand factor, and
Studies Are ABNORMAL? fibrinogen
Preoperative testing was ordered on a 43-year-old Cytotoxic • Antibody or toxin that attacks the cells of
woman scheduled for an elective hysterectomy. She has particular organs
suffered with dysfunctional uterine bleeding for 6
months. Rather than go to the hospital setting, she
Hemangiomas • Benign tumor of dilated blood vessels

went to a physician office laboratory that accepted her HLA • Human leukocyte antigens, which are found in
insurance. Her surgeon ordered a CBC with platelet white blood cells and are part of the major histocompatibil-
count and a PT and PTT. Her CBC was within reference ity complex
range but the results of her PT and aPTT were: Hyperviscosity • Excessive resistance to the flow of liquids
PT 10.6 seconds (Reference range, 10 to 14) Menorrhagia • Excessive menstrual bleeding
aPTT 53 seconds (Reference range, 28 to 38) Microangiopathic • Related to pathology of small blood
The elevated PTT was an unexpected result. Possi- vessels
bilities for an elevated PTT include a factor deficiency, Paresthesias • Abnormal sensation that results from an
the presence of a circulating anticoagulant, or a patient injury to one or more nerves, described as numbness or
on heparin. Heparin was eliminated as a possible con- prickly or tingling feeling
tributor to the prolonged PTT since there was no
patient history of anticoagulation therapy. Mixing stud-
Telangiectasia • Vascular lesion formed by dilation of a
group of small blood vessels, most frequently seen on face
ies are familiar screening tests in the clinical laboratory
and thighs
to determine whether there is a factor deficiency or a
circulating anticoagulant. The technologist decided to
perform mixing studies on this patient and proceeded References
with the laboratory protocol. In mixing studies, the 1. Glassy E, ed. Color Atlas of Hematology: An Illustrated
patient’s plasma is mixed with pooled normal plasma, Field Guide based on Proficiency Testing. Illinois:
in a 1:1 ratio and the elevated test is repeated. Pooled Chicogo College of American Pathologists, 1998: 206.
normal plasma contains all clotting factors and tech- 2. Bruce L. Quantitative disorders of platelets. In: Rodak B,
nologists use normal quality control material as the ed. Hematology: Clinical Principles and Applications,
source of pooled plasma. Once the test is repeated, if 2nd ed. Philadelphia: WB Saunders, 2002: 686.
3. Wolf BC, Neiman RS. Disorders of the Spleen. Philadel-
the result returns to the normal range, then it is
phia: WB Saunders, 1989: 22.
assumed that the source of aPTT elevation was a clot- 4. Blaney KD, Howard PR. Basic and Applied Concepts of
ting factor deficiency and factor assay tests on the Immunohematology. Boston: Mosby, 2000: 304.
plasma should be ordered. If the repeated test does not 5. vanden Bent PM, Meyboom PH, Egberts AC. Drug
return to the reference range, then it is assumed that the induced thrombocytopenia. Drug Saf 27:1243–1252,
patient plasma contains a circulating anticoagulant. As 2004.
an additional screening procedure, the aPTT test was 6. Bengston K, Skinner M, Ware R. Successful use of anti-
incubated for 1 to 2 hours. The rationale behind this CD20 (Rituximab) in severe life threatening childhood
additional step is to determine if there is a weak or ITP. J Pediatr 143:670–673, 2003.
time-dependent circulating inhibitor. Certain inhibi- 7. Michel M, et al. Does HELICOBACTER pylori initiate or per-
tors such as factor VIII inhibitor have a stronger petuate immune thrombocytopenia purpura? Blood
inhibitory effect with prolonged incubation. These 103:890, 2004.
8. Ezra Y, Rose M, Eldor H. Therapy and prevention of
pathological circulating inhibitors will be thoroughly
thrombotic thrombocytopenia purpura during preg-
discussed in Chapter 19. nancy: A clinical study of 16 pregnancies. Am J Hematol
51:1–6, 1996.
9. Zheng X, Chung D, Tekayama TK, et al. Structure of von
Willebrand factor cleaving protease (ADAMS-13), a
metalloprotease involved in thrombotic thrombocy-
topenic purpura. J Biol Chem 270:41059–41063, 2001.
WORD KEY 10. Levy GC, Nicolas WC, Lian EC, et al. Mutations in a
member of the ADAMTS gene family causing throm-
Alloimmunization • Antibodies that occur as a result of botic thrombocytopenic purpura. Nature 413:488–494,
antigens introduced to the body through blood and tissue 2001.
Copyright © 2007 by F. A. Davis.

256 PART IV • Hemostasis and Disorders of Coagulation

11. Kwaan HC, Soff GA. Management of thrombotic throm- 17. Philips MD, Santhouse A. von Willebrand disease:
bocytopenic purpura and hemolytic uremic syndrome. Recent advances in pathophysiology and treatment.
Semin Hematol 34:159–166, 1997. Am J Med Sci August:77–86, 1998.
12. Bell A. Extracorpuscular defects leading to increased 18. Liles DK, Knupp CL. Quantitative and qualitative
erythrocyte destruction: Nonimmune causes. In: platelet disorders and vascular disorders. In: Harmening
Rodak B, ed. Hematology: Clinical Principles and D, ed. Clinical Hematology and Fundamentals of
Applications, 2nd ed Philadelphia: WB Saunders, Hemostasis. Philadelphia: FA Davis, 2002: 481.
2002: 67. 19. Kunishima S, Kamiya T, Saito H. Genetic abnormalities
13. Bruce L. Quantitative disorders of platelets. In Rodak B, of Bernard-Soulier syndrome. Int J Hematol 76: 319–
ed. Hematology: Clinical Principles and Applications, 327, 2002.
2nd ed. Philadelphia: WB Saunders, 2002: 697. 20. Rogers RL, Lazarchick J. Identifying Glanzmann’s
14. Sondoval C, Dong S, Visintainer P. Clinical and labora- thrombasthenia. Lab Med 27:579–581, 1996.
tory features of 178 children with recurrent epistaxis. J 21. Paper R, Kelley LA. A Guide to Living With von Wille-
Pediatr Hematol 24:47–49, 2002. brand Disease. Pennsylvania: Aventis Bering, 2002: 53.
15. Saxena R, Gupta M, Gupta PC. Inherited bleeding disor- 22. Castellone D. Down and Dirty Coagulation: Practical
ders in Indian women with menorrhagia. Hemophilia Solutions and Answers. ASCP Workshop, May 7, 2004,
9:193–196, 2003. Abstract 5799, Baltimore, MD.
16. Lusher J. An underlying cause of menorrhagia. Mod 23. Bick RL, Scates SM. Qualitative platelet defects. Lab
Med 63:30–31, 1995. Med 23:95–103, 1992.
Copyright © 2007 by F. A. Davis.

17 Defects of Plasma
Clotting Factors

Betty CIESLA

Evaluation of a Bleeding Disorder Objectives


and Types of Bleeding
After completing this CHAPTER, the student will be ABLE to:
The Classic Hemophilias 1. Describe the variable types of bleeding found in
The Factor VIII Molecule patients with clotting factor deficiencies versus
Symptoms in the Hemophilia A patient platelet disorders.
Laboratory Diagnosis of Hemophilia Patients 2. Define the factor VIII molecule.
Treatment for Hemophilia A Patients 3. Outline the genetics of the hemophilia disorders.
Quality of Life Issues for Hemophilia A Patients
4. Describe the symptoms of an individual with
Hemophilia B or Christmas Disease hemophilia A and B.
Congenital Factor Deficiencies With Bleeding
Manifestations 5. Define the laboratory results in an individual
with hemophilia A and B.
Congenital Factor Deficiencies Where Bleeding
Is Mild or Absent 6. Describe the management and treatment of an
Factor XIII Deficiency individual with hemophilia A and B.
Bleeding Secondary to a Chronic Disease Process 7. Distinguish the clotting factor disorders with
The Role of Vitamin K in Hemostasis little or no bleeding.
Vitamin K Deficiency and Subsequent Treatment 8. Distinguish the acquired factor disorders with
regard to symptomatology and treatment.

257
Copyright © 2007 by F. A. Davis.

258 PART IV • Hemostasis and Disorders of Coagulation

EVALUATION OF A BLEEDING mal gene and passes the gene to her sons. Not every
DISORDER AND TYPES OF BLEEDING male child will be affected, only those who inherit the
Patients who experience recurrent bleeding episodes abnormal gene. Likewise, if daughters inherit the
are a select group of individuals that need to be evalu- abnormal gene, they are obligatory carriers. History is
ated for the source of their bleeding disorder. Bleeding rich with accounts of hemophilia from the Talmud to
may occur due to an inherited clotting factor defect or British monarchy. Queen Victoria carried the abnormal
an acquired deficiency secondary to some other cause. gene and passed it through her offspring (nine births,
Factors that should be considered in evaluating a bleed- five living children) into the Russian royal family, the
ing disorder are the patient history, physical examina- Spanish dynasty, and the German royal family (Fig.
17.1). Victoria herself had no family history of hemo-
tion, laboratory testing, and family bleeding history.
Often, the abnormal bleeding that they experience is philia so her abnormal gene was acquired as a result of
not perceived as abnormal because that is all that they spontaneous mutation, which occurs in 30% of cases.
have ever known. Therefore, the questions that are
asked relative to the types of and frequency of their The Factor VIII Molecule
bleeding need to be extremely specific and nonthreat- Factor VIII is the only one of the clotting factors that is
ening. Bleeding comes under two main categories: open not synthesized exclusively by the liver. It is unique
bleeds and closed bleeds. among clotting factors for two reasons. Factor VIII is
Open bleeds are those types of bleeding such as genetically controlled by the X chromosome (it is sex-
tongue bleeding, tonsil bleeding, gum bleeding, epi- linked), and it forms a complex with von Willebrand
staxis, menorrhagia, umbilical cord bleeding, and cir- factor (vWF), which transports the factor into the circu-
cumcisional bleeding. Closed bleeds are soft tissue lation and is synthesized by an autosomal chromosome
bleeds, genitourinary bleeding, gastrointestinal bleed- (Fig. 17.2). This clotting factor is also labile and unstable
ing, and bleeding into the muscle, joints, skin, bone, or in stored plasma. In individuals with hemophilia A, the
skull. Not every patient experiences all types of bleed- vWF level will be normal so that bleeding time will be
ing; some patients with clotting factor deficiencies normal; however, the aPTT will be abnormal because of
never experience a bleeding episode. Yet, it is prudent the reduced level of factor VIII.
to gather as much information as can be obtained to
assess an individual with a history of bleeding. Symptoms in the Hemophilia A Patient
Plasma clotting factors are inactive enzymes that
circulate in plasma awaiting activation when injury Clotting factors are measured in terms of their percent
occurs. They represent a significant ingredient to the activity as well as their function in coagulation tests.
proper clotting mechanism. Clotting factors that are Most clotting factors need to be available in the body at
poorly synthesized, inactivated by inhibitors, con- a minimum of 30% to achieve hemostasis. Bleeding
sumed by a rogue clotting process or functionally manifestation in hemophilia A individuals are related to
impaired will lead to faulty hemostasis. the level of factor VIII. There are three levels of clotting
factor activity in hemophilia:
• Severe, <1%
THE CLASSIC HEMOPHILIAS • Moderate, 1% to 5%
For most individuals the word HEMOPHILIA is at least a • Mild, 6% to 24%
recognizable term. Many negative perceptions arise Patients with severe hemophilia A will manifest
with this bleeding disorder including deep dark family early bleeding manifestations such as circumcisional
secrets, profuse bleeding from small wounds, excruciat- bleeds or umbilical cord bleeding. As they become more
ing pain, and early death. By definition, HEMOPHILIAS rep- mobile, ordinary activities such as crawling, walking, or
resent ANY of a group of disorders in which a particular running may present challenges. It is not uncommon to
clotting factor is decreased. With 13 clotting factors nec- see the severe hemophiliac child in protective gear
essary for clot formation, there should be a wide range (knee pads, ankle pads, helmet) for outside play. Bleed-
of hemophilias. Classically, however, only two disorders ing may occur in other areas such as the gastrointestinal
are referred to by the name HEMOPHILIAS: hemophilia A, tract, the kidneys (hematuria), or gums or in
factor VIII deficiency and hemophilia B, factor IX defi- hematomas. It is not accurate to say that individuals
ciency. Both of these disorders are sex-linked recessive with hemophilia bleed more profusely. Rather, bleeding
disorders, meaning that the mother carries the abnor- continues for a longer period of time due to the
2007 by F. A. Davis.

Edward Victoria
Duke of Kent Princess of Saxe-Coburg

Victoria
Albert Queen of England

Victoria Fredrick Ed VII Alexandra Alice Louis of Hesse Alfred Helena Louise Arthur Leopold Helen Beatrice Henry
of E ngland

Wilhelm II Sophie George V Irene Henry Fred Alix Nicholas II Alice of Alfonso XIII Eugenie Leopold Maurice
of Greece of Russia Athlone of Spain

George VI Waldemar Prince Henry Olga Tatiana Marie Anastasia Alexis Lady Rupert Alfonso Gonzalo
Sigmund May Abel
of Russia Smith

Princess Queen Prince Juan Carlos


Margaret Elizabeth Phillip of Spain

Normal male

Princess Prince Prince Prince Normal female


Anne Charles Andrew Edward

Hemophilic male

Carrier female

Male died in infancy.


probably hemophilc

Figure 17.1 Queen Victoria carried the abnormal gene for thalassemia and passed it through her offspring into the Russian royal family,
the Spanish dynasty, and the German royal family.
259
Copyright © 2007 by F. A. Davis.

260 PART IV • Hemostasis and Disorders of Coagulation

X-Chromosome AHF

AHF

Factor VIII Figure 17.2 Factor VIII com-


Complex plex is controlled by the X
Autosome #5 chromosome and an autosomal
chromosome. This complex
transports factor VIII into the cir-
culation. vWF, von Willebrand
vWF monomers vWF polymer factor; AHF, antihemophilic factor.

decreased level of clotting factor. Platelet counts are abnormal when mixed with a specific factor-deficient
normal and blood vessel function is adequate. Perhaps plasma suggests that the patient is missing the same
the most debilitating bleeds are muscle bleeds or joint clotting factor as that specific factor-deficient plasma. If
bleeds, which have the potential for causing long-term the patient and deficient plasma give a normal result,
disability, reduced range of motion, and intense pain. then obviously the patient supplied the factor missing
Joints become painful, swollen, and engorged with in the factor-deficient plasma. The aPTT result is plotted
blood. Hemarthrosis occurs in the joints as pooled on the factor-activity curve, and the level of factor activ-
blood damages the surrounding tissue while a clot ity is derived from the standard curve.
eventually forms. The joint become less and less
mobile, limiting physical activity (Fig. 17.3). Internal
hemorrhages into the muscles and deep soft tissues may
compress and damage nerves. Intracranial bleeding is a
leading cause of death in hemophilia A individuals, and
other complications like paralysis, coma, memory loss,
or stroke may precede an eventual fatality. Female carri-
ers for the hemophilia gene rarely have symptoms, yet
there are occasions when carrier females may become
symptomatic. The union of a hemophilia patient and a
female carrier would likely produce a symptomatic
female.

Laboratory Diagnosis
of Hemophilia Patients
Laboratory diagnosis of hemophilia patients is fairly
uncomplicated. Laboratory tests which are ordered
include bleeding time, PT, aPTT, and factor assays. In
hemophilia, the bleeding time test is normal, the PT is
normal, and aPTT is elevated, due to the reduced factor
VIII. Single factor assays provide a means of assessing
the percent activity of a clotting factor. These assays are
performed using the aPTT test. A standard curve is cre-
ated using serial dilutions of normal plasma of known
factor levels and assigning a 1:10 dilution of normal
plasma as 100% activity. Commercially prepared factor
deficient plasma is then mixed with a 1:10 dilution of Figure 17.3 Hemarthrosis occurs in the joints as pooled
patient plasma and aPTT is performed. An aPTT that is blood damages the surrounding tissues.
Copyright © 2007 by F. A. Davis.

CHAPTER 17 • Defects of Plasma Clotting Factors 261

Treatment for Hemophilia A Patients expenses for this product are unfortunately the most
Treatment options for hemophilia patients span decades costly, and these costs are passed on to potential users.
and present one of the saddest treatment histories of any
patient group with an inherited disorder. Factor Quality of Life Issues for
Hemophilia A Patients
VIII was discovered in 1937 and was termed anti-
hemophilic globulin.1 In the early days, treatment of Having a child with severe hemophilia A or B presents
hemophilia A patients consisted of giving whole blood special challenges to the parents and the family unit.
units to relieve symptoms. Not until 1957 was it real- The threat of hospitalizations, limited mobility, main-
ized that the deficient coagulation protein was a compo- streaming in schools, and the child’s drive for independ-
nent of the plasma portion of blood. Cryoprecipitate, a ence present potentially stressful environments. Added
plasma derivative, was discovered in 1964. This prod- to this is the cost of infusible factor, either recombinant
uct is produced as an insoluble precipitate that results or high purity products that could go as high as
when a unit of fresh frozen plasma is thawed in a stan- $50,000 if a patient has several bleeding episodes for
dard blood bank refrigerator. Cryoprecipitate contains which he needs to be hospitalized. Individuals with a
fibrinogen, factor VIII, and vWF. This product is chronic condition face many anxieties and may struggle
extracted from plasma and usually pooled before it is with feelings of isolation, anger, and disappointment
given to the patient according to weight and level of fac- (Table 17.1). Fortunately, in the United States, there are
tor VIII. This product presented a major breakthrough hemophilia treatment centers that offer a network of
for the hemophilia population because it was an easily needed services, and many states have local chapters of
transfusable product affording the maximum level of the National Hemophilia Foundation. 2 Prophylaxis
factor to the individual. Next in the chronology of treat- with factor concentrates limits bleeding episodes, and
ment products for hemophilia was clotting factor prod- the use of magnetic resonance imaging offers the physi-
ucts. These freeze-dried products were developed in the cian a more effective means of evaluating joint damage.3
early 1970s. The products were lyophilized and freeze Issues concerning medical insurance coverage continue
dried and could be reconstituted and infused at home. to plague the hemophilia community.
This treatment offered the hemophilia population an The development of factor VIII inhibitors occurs
independence that they had never previously experi- in 15% to 20% of all hemophilia A individuals.4 These
enced. Finally they were in control because they could inhibitors are autoantibodies against factor VIII that are
self-infuse when necessary and provide themselves with time and temperature dependent and capable of neu-
prompt care when a bleeding episode developed. But a tralizing the coagulant portion of factor VIII. Treatment
dark cloud loomed over the bleeding community. for patients who develop inhibitors is difficult and treat-
Approximately 80% to 90% of hemophilia A patients ment protocols follow various paths. When the
treated with factor concentrates became infected with inhibitor is low titer or the individual is a low respon-
the HIV virus. Factor concentrates were made from der, physicians may infuse an appropriate level of factor
pooled plasma from a donor pool that was less than ade- VIII in an attempt to neutralize the inhibitor.4 If this is
quately screened. Additionally, manufacturing compa- not effective, patients must be treated with a factor sub-
nies were less than stringent with sterilization methods
and screening for HIV virus did not occur in blood
banks until 1985. When each of these factors is brought
to bear, the tragedy to the bleeding community is easily Table 17.1  Quality of Life Issues
understood. According to the National Hemophilia for Hemophilia A and
Foundation,2 there are 17,000 to 18,000 hemophilia B Patients
patients (hemophilia A and B) in the United States. Of
• Joint damage
those, 4200 are infected with HIV/AIDS. There are no
• Reduced mobility
numbers available for wives or children who could have • Hemorrhage
been secondarily infected. Recombinant products • Fear
became available in 1989 and represent the highest • Physical restrictions
purity product because they are not human derived. • HIV/AIDS
Recombinant technology uses genetic engineering to • Hepatitis C
insert a clone of the factor VIII gene into mammalian • Future insurability
cells, which express the gene characteristic. Production
Copyright © 2007 by F. A. Davis.

262 PART IV • Hemostasis and Disorders of Coagulation

stitute, usually porcine factor VIII or alternative thera- A prothrombin, factor II deficiency may occur as a
pies such as anti-inhibitor coagulant complex.5 Gene result of a dysfunctional protein or as a result of dimin-
therapy, as a treatment alternative, continues to provide ished production of factor II. A structural defect in the
hope for those suffering from hemophilia. The idea here protein is termed dysproteinemia and individuals with
is to insert a copy of the factor VIII or factor IX gene into this particular deficiency may bleed. Additionally, a spe-
a virus vector that will then lodge in the body and start cific mutation in the prothrombin gene has been recog-
producing normal amounts of circulating factor. Com- nized since 1996. Located on chromosome 11, a single
plications from rejection of the virus vector in humans substitution of guanine to adenine at position 20210
have proved to be a delicate issue, yet there is optimism of the prothrombin gene produces prothrombin
that gene therapy for hemophilia patients could eventu- G20210A. This mutation increases the prothrombin
ally succeed. level and predisposes an individual to venous thrombo-
sis.7 Individuals should be screened for this mutation if
Hemophilia B or Christmas Disease any of the following are part of their patient history: a
history of venous thrombosis at any age, venous throm-
Individuals with hemophilia B lack factor IX clotting bosis in unusual sites, a history of venous thrombosis
factor. All of the conditions concerning inheritance, during pregnancy, and a first episode of thrombosis
clinical symptoms, laboratory diagnosis, and complica- before age 50.8
tions are the same for severe hemophilia B individuals Another mutation recently discovered (1993) is
as for severe hemophilia A individuals. Hemophilia B factor V Leiden. This mutation is produced by substi-
accounts for only 10% of those with hemophilia. tuting arginine with glutamine at position 506 of the
Patients with hemophilia B will have a prolonged aPTT factor V gene. The new gene product is factor V Leiden.
and will have decreased factor assay activity. Treatment In the normal coagulation scheme, once protein C is
of hemophilia B consists of factor IX concentrates or activated, it works to inactivate factors V and VIII, to
prothrombin complex that is a mixture of factors II, VII, inhibit the clotting mechanism. The mutated gene,
IX, and X. factor V Leiden, impedes the degradation of factor V
by protein C, causing activated protein C resistance.
Congenital Factor Deficiencies This condition accounts for increased clot forma-
With Bleeding Manifestations tion with the subsequent development of deep vein
Patients having deficiencies of factors II, V, VII, and X thrombosis or other hypercoagulability conditions
are rare and are usually the result of consanguinity. Most (see Chapter 19).
of these disorders are autosomal recessive, affecting
both males and females. Types of bleeding that may be Congenital Factor Deficiencies
Where Bleeding Is Mild or Absent
observed are skin and mucous membrane bleeding.
Joint and knee bleeding is unusual except for factor VII In this group of factor deficiencies are those concerned
deficient patients. These patients may show joint hem- with contact activation and clot stabilization. Factors
orrhages and epistaxis. In a recent survey of the 225 XI, XII, Fletcher, and Fitzgerald are each synthesized by
hemophilia treatment centers in the United States, 7% the liver and are involved early in the coagulation cas-
of patients were identified with having a rare bleeding cade, in vitro. They become responsive when they con-
disorder.6 Of these, factor VII was the most common. tact surfaces such as glass in test tubes or ellagic acid in
Abnormal preoperative screenings led to the diagnosis testing reagents. Factor XII deficiency is an autosomal
of most of these patients. When bleeding occurred in recessive trait where there is a prolonged PTT in labora-
one half of these patients, no therapy was necessary.6 tory testing. Individuals with this deficiency do not
Those individuals inheriting these deficiencies het- bleed, however, and are more prone to pathologic clot
erozygously tend to have few bleeding manifestations, formation. Factor XI deficiency or hemophilia C is an
since they will have one half of factor activity. Treatment autosomal recessive trait with a high predominance in
of patients with inherited deficiencies of factors II, VII, the Ashkenazi Jewish and Basque population in South-
and X consists of prothrombin complex concentrates. ern France. The heterozygous frequency of this gene in
Factor VII clears rapidly from the plasma, and therefore this population group is 1:8.9 Bleeding is unlikely,
booster doses are usually necessary to maintain clotting. unless trauma or surgery occurs. There is little correla-
Two new gene mutations, recently discovered, are espe- tion between the level of factor XI activity and the sever-
cially pertinent to this discussion. ity of bleeding episodes. Fletcher factor or prekallikrein
Copyright © 2007 by F. A. Davis.

CHAPTER 17 • Defects of Plasma Clotting Factors 263

deficiency manifests itself as an autosomal dominant each negatively affect clotting factor production and
and recessive trait. Again patients experience throm- clotting factor function. Factors that have a short half-
botic events such as myocardial infarction or pul- life such as factor VII and the vitamin K–dependent fac-
monary embolism. An interesting feature of this tors (II, VII, IX, and X) are particularly vulnerable. Liver
deficiency, in vitro, is that the initially prolonged aPTT disease brings a myriad of potential problems to coagu-
will shorten upon prolonged incubation with kaolin lation capability. In addition to poor production and
reagents. Fitzgerald factor deficiency, also called high- function of clotting factors, there is weak clearance of
molecular-weight kininogen deficiency, is a rare autoso- activated clotting factors and the accumulation of plas-
mal recessive trait. Deep vein thrombosis and minogen activators. If plasmin is activated to a high
pulmonary embolism are features of this disorder.10 degree, excessive clot lysis will be stimulated and DIC
and hemorrhaging may result. Unexpectedly elevated
Factor XIII Deficiency prothrombin times in a previously well patient may sig-
nal the advent of liver disease and the patient should be
Factor XIII is unique in that it is a transglutaminase
carefully monitored. Patients with liver disease who are
rather than a protease as are most of the other coagula-
bleeding are treated with fresh frozen plasma, a source
tion factors. The role of this factor in coagulation is to
of all clotting factors and natural inhibitors. As little as
provide stabilization to the fibrin clot through cross-
15 mL of plasma can increase the clotting factor activity
linkage of fibrin polymers. Proper levels of factor XIII
by 15% to 25%.12
are essential for proper wound healing, hemostasis, and
Renal disease, especially nephrotic syndrome,
the maintenance of pregnancy. This factor is not tested
usually leads to poor renal filtration and the presence of
for in the traditional coagulation tests such as PT, aPTT,
low-molecular-weight coagulation proteins in the urine
thrombin time, or bleeding time. Therefore, in a patient
of about 25% of patients with these disorders. Impaired
with factor XIII disorder, the traditional coagulation
platelet function is a feature of renal disease, and
screening test will be normal. Screening for factor XIII
patients with renal disorders are cautioned against tak-
deficiency is accomplished through the 5 mol/L urea
ing aspirin or other platelet inhibitors.
test, a primitive test which measures the stability or
firmness of the clot after 24 hours in a 5 mol/L urea
solution. If factor XIII is decreased, then the clot that is The Role of Vitamin K in Hemostasis
formed is stringy and loose, rather than the firm clot of
stable hemostasis. Additionally, quantitative assays for Vitamin K is a fat-soluble vitamin necessary for the
factor XIII are available. Congenital deficiencies of fac- activation of factors II, VII, IX, and X. This vitamin
tor XIII are rare autosomal recessive disorders. Deficien- is taken in through the diet in the form of green leafy
cies have been linked to poor wound healing, keloid vegetables, fish, and liver. It is also synthesized in small
formation, spontaneous abortion, and recurrent amounts by the intestinal bacteria BACTERoides FRAGILIS
hematomas. Approximately, one half of patients have a and some strains of ESCHERICHIA coli. Newborns are
family bleeding history, and large keloid scar formation usually vitamin K deficient because of the sterile
appears to be a consistent finding in these patients.11 environment of the small intestine, and therefore their
Treatment of inherited disorders is through fresh frozen levels of factors II, VII, IX, and X are low. Premature
plasma or cryoprecipitate, a source of factor XIII. infants have levels of vitamin K–dependent factors as
Acquired deficiencies of this factor may be associated low as 20% to 30%.13 As of the 1960s, all newborns are
with Crohn’s disease, leukemias, DIC, and ulcerative given vitamin K to avoid hemorrhagic disease of the
colitis. newborn.
The vitamin K–dependent factors are low-
molecular-weight proteins, with gamma-carboxyl
Bleeding Secondary to a residues at their terminal ends. To become activated and
Chronic Disease Process
fully participate in the coagulation scheme, they must
Liver disease, renal disease, and autoimmune processes take on a second carboxyl group through the action of
may lead to deficiencies in clotting factors that can the enzyme gamma glutamyl carboxylase (Fig. 17.4).
cause bleeding. Because almost all of the procoagulants This reaction requires vitamin K. Once this reaction is
and inhibitors are synthesized by the liver, conditions accomplished, these factors can then bind to calcium
such as alcoholic cirrhosis, biliary cancer, congenital and then to phospholipids for full participation in coag-
liver defects, obstructive liver disease, and hepatitis can ulation pathways.
Copyright © 2007 by F. A. Davis.

264 PART IV • Hemostasis and Disorders of Coagulation

COO– –OOC COO–


Table 17.2  Drugs That Cause a
CH2 CH Deficiency of Vitamin
K Clotting Factors
CH2 Vitamin K CH2 • Carbenicillin
• Moxalactam
CH COOH CH COOH • Cephamandole
• Cefoxitin
• Cefoperazone
NH2 NH2 • Tetracyclines
• Sulfonamides
Glutamic acid Gamma-carboxy glutamic acid • Aspirin
Figure 17.4 The carboxylation of the enzyme glutamic
acid. This reaction requires vitamin K.
itored by anticoagulant clinics and diets need to be
modified to compensate for the loss of vitamin K activ-
Vitamin K Deficiency and ity. Additionally, there is a long list of drugs that may
Subsequent Treatment interfere with vitamin K activity and subsequent hemo-
Vitamin K can be depleted through several mecha- stasis (Table 17.2).
nisms. Because body stores of vitamin K are extremely If a patient is vitamin K depleted, the PT and aPTT
limited, dietary sources are important. In patients who will most likely be elevated but able to be corrected by
have prolonged hospitalizations with only parenteral normal plasma. Factor assays of the specific vitamin K
nutrition, dietary deficiency will likely develop and the factors will reveal a depressed activity. Factor VII with
patient may need to be supplemented. Long-term the shortest half-life will be depleted first within 2 days;
antibiotic therapy that disrupts normal flora, a source of the other factors will take between 3 and 10 days to
vitamin K synthesis, may lead to vitamin K deficiency reach low hemostatic levels. With mild bleeding, oral
and subsequent bleeding. This is the case only if normal administration of vitamin K provides hemostatic recov-
nutrition is also disrupted. Chronic diarrhea, biliary ery within a couple of hours. More emergent bleeding
atresia, or other severe liver problems may lead to vita- situations may result in parenteral administration of
min K synthesis, because bile salts are needed for vitamin K, blood products, or infusion of prothrombin
proper absorption of vitamin K. Coumadin or warfarin concentrate complex. An interesting side note is reports
oral anticoagulant therapy reacts because this substance of patients who have used coumadin as an agent of
is a vitamin K antagonist and therefore gamma carboxy- suicide.14
lation of factors II, VII, IX, and X is prevented. Patients Acquired inhibitors of coagulation will be dis-
on oral anticoagulant therapy need to be carefully mon- cussed in Chapter 19.

COND ENSED CASE


A 7-year-old child had a fall from a piece of playground equipment. After 24 hours, he developed a deep hematoma in
his right thigh and his parents brought him to the emergency department to be evaluated. His family history did not
give any indication of any previous bleeding from birth or otherwise. WHAT tests should be ordered to rule out A COAG-
ULATION defect?

Answer
Although his family history does not indicate a clotting factor abnormality, preliminary clotting tests should include a
bleeding time, PT, and aPTT. This patient has a normal PT but an aPTT of 50 seconds (reference range, 20 to 38 sec-
onds). A factor assay was performed and indicated a mild factor VIII activity of 40% with a reference range of 50% to
150% activity. The patient was diagnosed with mild hemophilia A. This accident brought a previously undiagnosed
condition to light. This is important information in this patient’s personal and medical history. Future surgeries or
traumas will need to be carefully monitored.
Copyright © 2007 by F. A. Davis.

CHAPTER 17 • Defects of Plasma Clotting Factors 265

SUMMARy Points • From 15% to 20% of all hemophilia A individuals


develop factor VIII inhibitors.
• Patients with recurrent bleeding episodes need to be
evaluated for an inherited bleeding disorder. • Individuals with factor II, V, VII, and X deficiencies
may have minimal bleeding.
• Bleeding comes under two main categories: open
bleeds or closed bleeds. • Prothrombin complex concentrate is used to correct
deficiencies of factors II, VII, IX, and X.
• Plasma clotting factors need to maintain approxi-
mately 30% activity to achieve adequate clotting. • Prothrombin G20210A is a mutation of the pro-
• The factor VIII molecule is carried into plasma by thrombin molecule.
vWF. • Factor V Leiden is a genetic mutation of the factor V
• Hemophilias A and B are sex-linked recessive disor- molecule that predisposes to clotting episodes.
ders. • Deficiencies of factors XI, XII, Fletcher, and Fitzger-
• In hemophilia A, factor VIII is deficient; in hemo- ald usually lead to increased thrombotic events.
philia B, factor IX is deficient. • Factor XIII is unique among clotting factors because
• Women are carriers of the defective hemophilia it is a transglutaminase; the other clotting factors are
gene. proteases.
• Individuals with hemophilia experience prolonged • An inherited deficiency of factor XIII may lead to
bleeding from minor wounds. poor wound healing and spontaneous abortions.
• Individuals with hemophilia may experience many • Liver disease, renal disease, and autoimmune
types of bleeding including joint bleeding leading to processes may lead to deficiencies in clotting factors
hemarthrosis, hematomas, umbilical cord bleeding, that cause bleeding.
or mucosal bleeds. • Vitamin K is a fat-soluble vitamin necessary for the
• The bleeding time is normal in hemophilia A and B activation of factors II, VII, IX, and X.
patients; the aPTT is elevated. • Vitamin K is available through the diet; small
• Current treatment for hemophilia individuals con- amounts are synthesized by normal intestinal flora.
sists of recombinant factor products. • Newborns are vitamin K deficient and are given vita-
• Most individuals with hemophilia in the United min K at birth to avoid hemorrhagic disease of the
States use factor concentrates prophylactically. newborn.
• Prophylactic infusion of factor concentrates • If vitamin K is depleted, the PT and PTT will be pro-
has minimized the physical disabilities that longed.
may have occurred from unexpected bleeding • Coumadin, a therapeutic anticoagulant, is a vitamin
episodes. K antagonist.

CASE STUDY
A 54-year-old woman was admitted to the hospital with hematuria, anemia, easy bruising, and progressive weakness.
She gave no previous bleeding history or family history of bleeding even though she had multiple surgeries in the past.
Her surgeries included knee replacement. During this admission, she is complaining of a deep bruise in her right upper
thigh and hematuria. Her admitting laboratory data included the following:
WBC 6.0 × 109/L
Hgb 6.8 g/dL
Hct 20.2%
Platelets 321 × 109/L
PT 12.5 seconds (reference range, 10.5 to 12.4)
aPTT 67.6 seconds (reference range, <40)
Mixing studies: Immediate mixing and repeat PTT 39.6 seconds
aPTT after 1 hour 54.2 seconds
Factor VIII 4% (reference range, 50% to 150%)
WHAT is your INITIAL impression?
(continued on following PAGE)
Copyright © 2007 by F. A. Davis.

266 PART IV • Hemostasis and Disorders of Coagulation

(Continued)
Insights to the Case Study
This patient’s family history is helpful in eliminating a congenital hemostatic defect as a source of her hematuria. She has
had successful surgery events in the past but now suffers with hematuria and deep bruising. An elevated aPTT value can
be seen in anticoagulant therapy, particularly heparin, in clotting factor defects, and if a circulating inhibitor is present.
Mixing studies in this patient show variable results with initial correction of the patient’s aPTT and then subsequent pro-
longation upon incubation. A factor VIII inhibitor was considered as a likely explanation for the laboratory results and
the low factor VIII assay value. Inhibitors or autoantibodies against factor VIII may develop in populations other than
the hemophilia A population, where 10% to 30% develop these type of inhibitors. These inhibitors are directed against
a portion of the factor VIII molecule and are time and temperature dependent. Once identified, the inhibitor should be
quantitated using the Bethesda titer. In this procedure, equal volumes of pooled normal plasma that is platelet poor are
mixed with patient platelet poor plasma at pH 7.4. The mixture is incubated for 2 hours and the PTT is repeated. If the
patient plasma has anti–factor VIII activity, then some of the active factor VIII in the normal plasma will be affected. The
level of inhibitor is seen as a percentage of the normal activity of the factor when compared to the control plasma. One
Bethesda unit is equivalent to the inhibitor in which 50% factor activity will remain.

Review Questions
1. Which of the clotting factors is not a a. cryoprecipitate.
protease? b. fresh frozen plasma.
a. Factor II c. prothrombin complex concentrate.
b. Factor VII d. recombinant factor VIII.
c. Factor XIII
4. One of the more fatal bleeds in a hemophilia
d. Factor IX
patient involves:
2. Why is the bleeding time normal in hemo- a. intracranial bleeding.
philia A? b. mucosal bleeding.
a. Because of an increase in factor XIII c. joint bleeding.
b. Because the clotting problem is a factor VIII d. epistaxis.
problem
5. Which clotting factor deficiency is associated with
c. Because vWF is normal
poor wound healing?
d. Because the clotting problem is a factor IX prob-
a. Factor II
lem
b. Factor X
3. The purest treatment product for hemophilia A c. Factor XII
patients is: d. Factor XIII
Copyright © 2007 by F. A. Davis.

CHAPTER 17 • Defects of Plasma Clotting Factors 267

Hemarthrosis • Bloody effusion inside the joint


Hematoma • Swelling composed of a mass of clotted
 TROUBLESHOOTING blood confined to an organ, tissue, or space or caused by a
WHAT Do I Do When LABORATORy Results break in the blood vessel
Are Not Consistent With the PATIENT’s Keloid • Scar that forms at the site of injury that appears to
PHYSICAL PrESENTATION? have a rubbery consistency and shiny surface
A 74-year-old woman arrived in the emergency depart- Porcine • Of or relating to swine (pigs)
ment with bruising over most of her extremities.
She gave no family or personal history of bleeding
but did indicate that she had delivered eight children. References
Her bleeding time was slightly abnormal at 9 minutes 1. Corriveau DM. Major elements of hemostasis. In: Cor-
riveau DM, Fritsma GA, eds. Hemostasis and Thrombo-
(reference < 8 minutes), but her PT and aPTT were
sis in the Clinical Laboratory. Philadelphia: JB
within normal range. Factor assays of factors VIII and Lippincott, 1988: 6.
IX were normal, and platelet aggregation studies were 2. www.hemophilia.org. Accessed June 14, 2005.
normal. What are the possibilities for the incongruities 3. Berntrop E, Michiels JJ. A healthy hemophilic patient
in this patient workup? without arthropathy: From concept to clinical reality.
This patient presented a diagnostic dilemma. Semin Thromb Hemost 29:5–10, 2003.
Quality control was verified at all levels on all pieces of 4. Fritsma G. Hemorrhagic coagulation disorders. In
equipment used. A repeat bleeding time, PT, and aPTT Rodak B, ed. Hematology: Clinical Principles and
were performed and fell within ranges similar to the Applications, 2nd ed. Philadelphia: WB Saunders,
original. Factor assays were not repeated. These results 2002: 639–640.
stumped the coagulation staff. After careful considera- 5. Kleinman MB. Anti-inhibitor coagulant complex for the
tion of exactly what was being tested for, the possibility rescue therapy of acquired inhibitors to factor VIII: Case
report and review of literature. Hemophilia 8:694–697,
of a factor XIII deficiency was considered. Factor XIII is
2002.
necessary for clot stabilization and would healing. A 5 6. Acharya SS, Coughlin A, Dimichele DM, et al. Rare
mol/L urea test was performed, and the results were Bleeding Disorder Registry: Deficiencies of II, V,X, fib-
abnormal. An inherited deficiency of factor XIII is the rinogen and dysfibrinogenemia. J Thromb Hasemost
rarest of all of the bleeding disorders, presenting as 2:248–256, 2004.
autosomal recessive. Our patient has a history of multi- 7. Jensen R. Screening and molecular diagnosis in hemo-
ple pregnancies and successful deliveries; therefore an stasis. Clin Hemost Rev 13:12, 1999.
inherited coagulation deficiency was not considered. A 8. McGlennen RC, Key NS. Clinical laboratory manage-
thorough medication check revealed that the patient ment of the prothrombin G20210A mutation. Arch
was on cardiac medication, which potentially could Pathol Lab Med 126:1319–1325, 2002.
have caused an inhibitory effect on factor XIII, because 9. Seligsohn U. High frequency of factor XI (PTA)
all other factor-related assays were normal. Cryopre- deficiency in Ashkenazi Jews. Blood 51:1223,
1978.
cipitate was infused to prevent any future bleeding
10. Cheung PP, et al. Total kininogen deficiency (Williams
complication. The patient’s cardiac medication was dis- trait) is due to an argstop mutation in exon 5 of the
continued, and the patient was given an appropriate human kininogen gene. Blood 78:3919, 1991.
alternative medication for her cardiac condition. 11. Al-Sharif FZ, Aljurf MD, Al-Momen AM, et al. Clinical
(Many thanks to D. Castellone for the resource and laboratory features of congenital F XIII deficiency.
material for this case.) Saudi Med J 23:552–554, 2002.
12. Weiss AE. Acquired coagulation disorders. In: Cor-
riveau DM, Fritsma GA, eds. Hemostasis and Thrombo-
sis in the Clinical Laboratory. Philadelphia: JB
Lippincott, 1988: 176.
13. Zipursky A, Desa D, Hsu E, et al. Clinical and labora-
WORD KEY tory diagnosis of hemostatic disorders in newborn
infants. Am J Pediatr Hematol Oncol 1:217–226,
Atresia• As in biliary atresia, congenital closure, or 1979.
absence of some or all of the major bile ducts
14. Fritsma GA. Hemorrhagic coagulation disorders. In:
Crohn’s disease • Inflammatory bowel disease marked by Rodak B, ed. Hematology: Clinical Principles and Appli-
patchy areas of inflammation from the mouth to the anus cations. Philadelphia: WB Saunders, 2002: 173.
Copyright © 2007 by F. A. Davis.

This page has been left intentionally blank.


Copyright © 2007 by F. A. Davis.

Fibrinogen, Thrombin,
18 and the Fibrinolytic System

Betty CIESLA

The Role of Fibrinogen in Hemostasis Objectives


Disorders of Fibrinogen After completing this CHAPTER, the student will be ABLE to:
Afibrinogenemia 1. Identify the components of the fibrinolytic sys-
Hypofibrinogenemia tem.
Dysfibrinogenemia 2. Recall the role of fibrinogen in the coagulation
The Unique Role of Thrombin in Hemostasis and the fibrinolytic system.
Physiological Activators of Fibrinolysis 3. Describe plasmin in terms of activation and inhi-
Naturally Occurring Inhibitors of Fibrinolysis bition.
Measurable Products of the Fibrinolytic System 4. Differentiate the role of thrombin in both the
coagulation and fibrinolytic system.
Disseminated Intravascular Coagulation
The Mechanism of Acute Disseminated 5. Outline the inherited disorders of fibrinogen.
Intravascular Coagulation 6. Describe the laboratory testing for fibrinolytic
Clinical Symptoms and Laboratory Results in disorders.
Acute Disseminated Intravascular Coagulation 7. Define conditions that may precipitate dissemi-
Treatment in Acute Disseminated nated intravascular coagulation states.
Intravascular Coagulation
8. Describe the laboratory testing and management
of patients with disseminated intravascular coag-
ulation event.

269
Copyright © 2007 by F. A. Davis.

270 PART IV • Hemostasis and Disorders of Coagulation

THE ROLE OF FIBRINOGEN increased levels of lipoprotein will lead to less clot dis-
IN HEMOSTASIS solution, leaving clots available for a pathological
outcome.2
Fibrinogen is the principal substrate of the coagulation
and fibrinolytic system. This clotting factor has the
highest molecular weight of all of the clotting factors, DISORDERS OF FIBRINOGEN
and it is the substrate upon which the coagulation sys- Appropriate levels of fibrinogen are necessary to main-
tem is centered. This factor is heat labile but stable in tain hemostasis and to cause platelets to aggregate. The
storage. When fibrinogen is transformed to fibrin under reference range for fibrinogen is 200 to 400 mg/dL. Fib-
the influence of thrombin, it is the onset of solid clot for- rinogen is an acute-phase reactant, meaning that there
mation. The formation of fibrin occurs within minutes will be a transient increase in fibrinogen during inflam-
due in part to a positive feedback mechanism within the mation, pregnancy, stress, and diabetes and when tak-
hemostasis system. Once clotting factors are activated, ing oral contraceptives. Therefore, a careful patient
they accelerate the activity of the next factor, pushing history is necessary when evaluating a problem involv-
the reaction to conclusion. Negative feedback occurs ing fibrinogen. For the most part, decreases in fibrino-
when the activity of the reaction is delayed. This is the gen result from acquired disorders such as acute liver
role played by naturally occurring inhibitors within the disease, acute renal disease, or disseminated intravascu-
hemostatic system. With the assistance of factor XIII lar coagulation. Acquired increases in fibrinogen may be
and thrombin, the fibrinogen molecule is stabilized by demonstrated in hepatitis patients, pregnant patients,
cross-linked fibrin. Within hours, the fibrinolytic sys- or those with atherosclerosis.3 The inherited disorders
tem swoops in to dissolve the clots that have formed and of fibrinogen are afibrinogenemia, hypofibrinogenemia,
to restore blood flow. The creation of cross-linked fibrin and dysfibrinogenemia. These conditions are rare and
is an orderly process by which fibrinogen is cleaved into are marked by hematomas, hemorrhage, and ecchy-
fibrinopeptides A and B by thrombin. Fibrinogen is moses depending upon severity.
composed of three pairs of polypeptide chains: alpha,
beta, and gamma. When thrombin is generated, it
cleaves small portions of the alpha and beta chains, cre- Afibrinogenemia
ating fibrinopeptides A and B. The remaining portions The homozygous disorder, afibrinogenemia, is an auto-
of the alpha and beta chains stay attached to the fibrino- somal recessive disorder that shows less than 10 mg/dL
gen molecule. With fibrinopeptides A and B cleaved, the fibrinogen in the plasma. This small amount of fibrino-
fibrin monomer is created. These monomers sponta- gen is usually not demonstrable by traditional methods.
neously polymerize by hydrogen bonding to form a Infants with afibrinogenemia will show bleeding from
loose fibrin network, which is soluble. Trapped within the umbilical stump; poor wound healing and spon-
the soluble clot are thrombin, antiplasmins, plasmino- taneous abortion are also features of this disorder. Labo-
gen, and tissue plasminogen activator (tPA). Because ratory results will show elevated PT, aPTT, thrombin
thrombin is now protected from its inhibitors, it acti- time (TT), reptilase time, and abnormal platelet
vates factor XIII and calcium and then catalyzes the for- aggregation with most aggregating agents and elongated
mation of peptide bonds between monomers, forming bleeding time. Cryoprecipitate and fresh frozen plasma
fibrin polymers that lead to an insoluble and resistant are the replacement products used for medical manage-
clot1 (Fig. 18.1). Balance between the coagulation and ment of bleeds for these patients.
fibrinolytic systems is critical for maintenance of circu-
lation and injury repair. An imbalance in the coagula-
Hypofibrinogenemia
tion system could cause excess clotting; an imbalance of
the fibrinolytic system could cause hemorrhaging. Sev- Hypofibrinogenemia is the heterozygous form of afib-
eral other components may play a role in hemostatic rinogenemia. This disorder is autosomal recessive and
balance. In early studies, it has been suggested that indi- patients show between 20 and 100 mg/dL fibrinogen in
viduals with a high concentration of lipoprotein A may their plasma. Patients with this disorder may show mild
have reduced fibrinolytic activity due to decreased plas- spontaneous bleeding and severe postoperative bleed-
min generation. Cholesterol and triglycerides are all ing. Results of laboratory testing, whether prolonged
fatty components of lipoproteins. It is conceivable that or normal, will depend on the amount of fibrinogen
reduced plasmin generating activity in individuals with present.
Copyright © 2007 by F. A. Davis.

CHAPTER 18 • Fibrinogen, Thrombin, and the Fibrinolytic System 271

Thrombin's Action on Fibrinogen


Thrombin

Alpha chains A A
Fibrinogen
Beta chains B B
Gamma chains

Thrombin

A A
+ B B Fibrin peptides

Fibrin monomer

Spontaneous
polymerization

Hydrogen bonds Weak fibrin


polymer

FXIIIa
Ca+++

Covalent
bonds

Figure 18.1 Thrombin’s activity on


fibrinogen, from fibrin monomer to Cross-linked fibrin polymer
fibrin polymer. (stable fibrin clot)

Dysfibrinogenemia level of fibrinogen is normal. The clottable assay for


These fibrinogen disorders are autosomal dominant and quantitative fibrinogen is abnormal as this assay is
are inherited homozygously and heterozygously. They dependent on the proper amount and proper function-
ing fibrinogen.
produce a qualitative disorder of fibrinogen in which an
amino acid substitution produces a functionally abnor-
mal fibrinogen molecule. Although these disorders are THE UNIQUE ROLE OF
an academic curiosity, named for the city in which the THROMBIN IN HEMOSTASIS
patient was discovered, they are infrequently associated Thrombin holds a respected place in the coagulation
with a bleeding tendency. A few are associated with mechanism for its multiplicity of function and the
thrombosis.4 Approximately 40 abnormal fibrinogens numerous reactions it mediates. The impact of throm-
have been discovered. Because fibrin formation is bin is far reaching from the initial activation of the
affected by the abnormal fibrinogen molecule in dys- platelet system to the initiation of the fibrinolytic sys-
fibrinogenemia, most of the normal laboratory assess- tem and subsequent tissue repair. Prothrombin is the
ments for fibrinogen will be abnormal. The PT, aPTT, precursor to thrombin and can only be converted by the
TT, and reptilase time will be increased. An immuno- action of factor X, factor V, platelet factor 3, and cal-
logic assay of fibrinogen that measures the antigenic cium. Thrombin is generated in small concentrations
Copyright © 2007 by F. A. Davis.

272 PART IV • Hemostasis and Disorders of Coagulation

through injury to the endothelial cells and proceeds to XIIa, kallikrein, and high-molecular-weight kininogen.
initiate a more enhanced coagulation mechanism. Once Once produced, plasmin, a potent enzyme, does not
generated, thrombin is involved in the platelet release distinguish between fibrin and fibrinogen and works to
reaction as well as platelet aggregation. Secondarily, digest both. Additionally, plasmin also hydrolyzes fac-
thrombin stimulates platelets to produce the platelet tors V and VIII, and if circulating in the plasma as patho-
inhibitor, prostacyclin, or PGI2. With the coagulation logical free plasmin, the damage to the coagulation
system alerted, thrombin activates factors V and VIII, system is significant, as clots are dissolved indiscrimi-
key cofactors in thrombus formation. Protein C, a natu- nately. Of interest is the fact that tPA has been synthe-
rally occurring inhibitor to coagulation, is also activated sized by recombinant technology and is presently used
by thrombin. An additional product thrombomodulin as a pharmaceutical product during stroke episodes for
which is secreted by endothelial cells amplifies protein fibrinolytic therapy. As a “clot-busting” drug, it has been
C activity when complexed with thrombin. 5 With effective in thrombotic strokes and if injected within a
respect to the fibrinogen degradation, thrombin plays a small time-frame can spare the patient serious stroke
key role in negative feedback by converting plasmino- side effects. Another plasminogen activator is uroki-
gen to plasmin to digest the soluble fibrin clot. This nase, a protease present in the urine and produced by
interplay of thrombin disposition and thrombin initia- the kidney. The physiological effect of urokinase is min-
tion of clot disposal is part of the biologic control of imal in clot dissolution; however, like tPA it is a valuable
hemostasis. Once the clot is dissolved, thrombin plays a commercial product used in thrombolytic therapy, for
role in repairing tissue and wounds (Fig. 18.2). patients with heart attacks, strokes, and other throm-
botic episodes.6 Streptokinase is an exogenous fibri-
Physiological Activators of Fibrinolysis nolytic agent, produced when a bacterial cell product
forms a complex with plasminogen, a pairing that con-
A critical link in the chain of hemostasis is the dissolu- verts plasminogen to plasmin. This toxic product
tion of fibrin clots, which usually occurs several hours results from infection with beta-hemolytic streptococci
after the stable clot is formed. In this way, blood flow is and is a dangerous byproduct if this bacterial strain
restored at the local levels and tissue healing is precipi- develops into a systemic infection. It has the most activ-
tated. The body provides naturally occurring or physio- ity on fibrinogen.
logical activators that initiate this process. The key
component in this reaction is plasminogen, a plasma
enzyme synthesized in the liver with a half-life of 48 Naturally Occurring Inhibitors
of Fibrinolysis
hours. Plasminogen is converted to plasmin, chiefly
through the action of tissue plasminogen activator The balance of hemostasis is aided by those products
(tPA), a substance released through the activity of that restrain fibrinolytic activity. These products,
endothelial cell damage and the production of throm- plasminogen activator inhibitor 1 (PAI-1) and alpha-
bin. Additional plasminogen activators include factor 2-antiplasmin, act upon different substrates in the fibri-

Platelets Platelet Aggregation


T
Stimulates
H
H
R
R
O
O Fibrinogen Fibrin

Converts Plasminogen Plasmin


M
M
Protein C Activated protein C (APC)
B
B
I Promotes
Wound healing

N
N Tissue repair Figure 18.2 The multiple roles of
thrombin in hemostasis.
Copyright © 2007 by F. A. Davis.

CHAPTER 18 • Fibrinogen, Thrombin, and the Fibrinolytic System 273

P P P P
D D E D D E D D E

D D E D D E D D
P P P

D D D D D D E

E E D D

DD DD/E DY/YD

Figure 18.3 The formation of D-


dimer and fibrin degradation prod- P= Plasmin
ucts. P, plasmin; D, D domain; E, E D= D domain
domain. E= E domain

nolytic system. PAI-1 is secreted by endothelial cells less than 40 μg/mL. Individuals with an intact and oper-
during injury and suppresses the function of tPA in the ational hemostatic system have normal FDPs. These
plasminogen-plasmin complex. Plasmin as a substrate products are measured semiquantitatively through
is directly inhibited by alpha-2-antiplasmin in a 1:1 direct latex agglutination of a thrombin clotted sample.
ratio at the target area. This inhibitor prevents plasmin Latex particles are coated with monoclonal antibodies
binding to fibrin in an orderly fashion and claims the to the human fibrinogen fragments D and E. The test is
role as the most important inhibitor of the fibrinolytic performed on serum using two dilutions, 1:15 and 1:20.
system. Inherited deficiencies of this inhibitor invari- It does not distinguish between fibrinogen and fibrin.
ably lead to hemorrhagic episodes. Secondary agents Pathological levels of FDPs interfere with thrombin for-
that can inhibit fibrinolysis are alpha-2-macroglobulin, mation and platelet aggregation. Elevated levels may be
C1 inactivator, and alpha-1-antitrypsin. These sub- seen in DIC, pulmonary embolism, obstetrical compli-
stances, as protease inhibitors, act upon thrombin for- cations, and other conditions7 (Table 18.1).
mation. Because thrombin is one of the initiators of the Once fibrin has been cross-linked and stabilized
generation of plasmin, the secondary effect on the fibri- by factor XIII, a stable clot has been formed. When this
nolytic system is unavoidable. clot is dissolved by plasmin, D-dimers are released.
Therefore, D-dimers suggest a breakdown of fibrin clot

Measurable Products
of the Fibrinolytic System
Physiological fibrinolysis occurs in an orderly fashion, Table 18.1  Conditions That May
producing measurable products that can be captured by Elevate Fibrin
laboratory assays. Specifically, the byproducts of an Degradation Products
orderly fibrinolytic system are fibrin split/degrada-
tion (FSP/FDP) products composed of fibrin fragments • Disseminated intravascular coagulation
labeled as X, Y, D, and E and the D-dimers, D-D • Pulmonary embolism
(Fig. 18.3). • Abruptio placentae
The accurate and precise measurement of these • Preeclampsia
• Eclampsia
products is the basis for therapeutic decisions once
• Fetal death in utero
pathological clot forming and lysing has been initiated.
• Postpartum hemorrhage
FSPs/FDPs are formed from plasmin action on fibrin and • Polycystic disease
fibrinogen. As plasmin degrades the fibrinogen mole- • Malignancies
cule, different fragments are split leading to early and • Lupus nephritis
late degradation products. Normal levels of FDPs are • Thrombolytic therapy
eliminated through the RES system and usually measure
Copyright © 2007 by F. A. Davis.

274 PART IV • Hemostasis and Disorders of Coagulation

and indirectly are an indication that clots have been anced, hyperactivating the coagulation and/or the fibri-
formed at the site of injury, at the local level. Excess D- nolytic system. This process is systemic, leading to
dimers are indicative of breakdown of fibrin products excessive disposition of thrombi or excessive hemor-
within the circulating blood. D-dimers can be assayed rhage. Additionally, the process is consumptive, con-
semiquantitatively and quantitatively. The semiquanti- suming clotting factors and platelets as soon as they are
tative assay uses monoclonal antibodies specific for this activated for coagulation. Usually the decrease in clot-
domain. A simple agglutination test, undiluted patient ting factors is more overpowering than the increase in
plasma is mixed with latex solution. Noticeable aggluti- lysis. In broad terms, DIC is associated with obstetrical
nation is a positive test and indicative of deep vein complications, malignancy, massive trauma, bacterial
thrombosis (DVT), pulmonary embolism (PE), or sepsis, asplenia, or necrotic tissue. Under each of these
disseminated intravascular coagulation (DIC). Quanti- major headings are many other pathological possibilities
tative D-dimer tests are automated and use an enzyme- for the initiation of a DIC event (see Table 18.2). Although
linked immunosorbent assay (ELISA) procedure. The most DIC occurs as acute, explosive episodes, there are
advantage of this procedure is its ability to detect low conditions that may lead to a chronic compensated DIC
levels of D-dimer and to provide specific information as state. These are much more difficult to diagnose because
to whether pathological clotting as in DVT or PE has the bone marrow and liver perform an excellent job of
occurred. D-dimers assays have great utility in monitor- maintaining equilibrium between the coagulation and
ing thrombolytic therapy.8 the fibrinolytic system. Laboratory results may be mini-
mally abnormal; yet once the underlying pathology
DISSEMINATED INTRAVASCULAR intensifies, an acute DIC episode is likely.9
COAGULATION
The mere mention of the words “the patient has DIC” The Mechanism of Acute Disseminated
usually strikes fear into the hearts of attending physi- Intravascular Coagulation
cians, laboratorians, and nursing staff. The acute DIC As is customary in normal hemostasis, both the coagu-
event is almost always unanticipated and dramatic. Fatal lation and the fibrinolytic system are activated in paral-
outcomes do occur. DIC is triggered by an underlying lel. What is missing in DIC is the negative feedback
pathological circumstance occurring in the body (Fig. mechanism that holds the systems in balance. Table
18.4). As a result, the hemostatic system becomes unbal- 18.2 is a composite of events in the DIC cycle:

Trauma

Toxin
Sepsis

Vascular occlusion Thrombosis


Platelets aggregate
Thrombocytopenia Bleeding

Coagulation Factors depleted Bleeding

Platelet function depleted Bleeding


Fibrin deposited
-Plasmin initiated Decrease in fibrinogen Bleeding Figure 18.4 Conditions that may
-FSPs increased precipitate disseminated intravascular
Fibrin does not polymerize Bleeding coagulation (DIC). Note the multiple
pathways. FSPs, fibrin split products.
Copyright © 2007 by F. A. Davis.

CHAPTER 18 • Fibrinogen, Thrombin, and the Fibrinolytic System 275

thrombin is bypassed, the platelet count is normal


Table 18.2  Events Triggering Dis- unlike the platelet count in DIC. Yet all other parts of
seminated Intravascular the DIC coagulation profile are abnormal. There is con-
Coagulation troversy as to whether primary fibrinolysis is a disease
entity unto itself or rather just a continuum in the
Infections GASTROINTESTINAL disorders vicious DIC cycle.
• Gram-negative bacteria • Acute hepatitis
• Gram-positive bacteria OBSTETRICAL COMPLICATIONS Clinical Symptoms and Laboratory
• Malaria • Maternal toxemia Results in Acute Disseminated
Tissue Injury • Abruptio placentae Intravascular Coagulation
• Crush injury • Hemolytic disease of the
In hemorrhagic episodes, most patients have extensive
• Burns newborn
• Massive head injury • Group B streptococcal
skin and mucous membrane bleeding, including ecchy-
MALIGNANCY infection mosis, epistaxis, and petechiae. Areas of entry such as
• Acute promyelocytic • Retained dead fetus surgical incisions, catheters, or venipuncture sites may
leukemia Other also ooze and must be carefully observed. In thrombotic
• Acute monoblastic or • Snake bites episodes, patients may exhibit acrocyanosis, hypoten-
myeloblastic leukemia • Heparin-induced sion, or shock. Microthrombi may occur in the nose,
• Microangiopathic thrombosis genitalia or digits, or major organs such as kidney, liver,
disorders • Septic shock or brain.
• Thrombotic thrombo- • Hemolytic transfusion Acute DIC is a medical emergency. The entire basic
cytopenia purpura reaction DIC coagulation profile is abnormal (Table 18.3). PT and
• Heat stroke • Graft versus host disease PTT are prolonged, fibrinogen and platelets are
decreased, and FDPs and D-dimers are dramatically
increased. D-dimer results are an essential piece of data
in emerging DIC patients; however, other pathologies
• Excessive generation of thrombin is triggered
such an inflammation, renal disease, or local clot may
by thromboplastin release (endothelial cells,
elevate the result.10 For this reason, all laboratory data
placenta, leukemic cells [promyelocytes or
must be carefully reviewed in concert with clinical
monoblasts], tumors).
symptoms before reaching the diagnosis. It has been
• Simultaneous enzymatic conversion of fibrino-
shown that not all patients in DIC have decreased fib-
gen to fibrin occurs.
rinogen levels. Indeed, recent studies have shown that
• Plasmin generation simultaneously degrades
for those patients exhibiting DIC but near normal fib-
fibrinogen/fibrin into FDPs; excess FDPs are
rinogen, clinical outcomes were much poorer, resulting
formed.
in severe organ failure.11 Recovering patients should be
• FDPs have affinity for fibrin monomers but fail
monitored over time using the same initial profile for
to polymerize properly; excess FDPs have an
comparison and evaluation of their coagulation status.
anticoagulant effect.
The patient will develop a microangiopathic hemolytic
• Plasmin causes inactivation of factors V,VIII,
anemia due to microthrombi disposition in the small
XI, and XII.
vessels. Schistocytes will be observed as a morphologi-
• Hemorrhage occurs as soluble fibrin monomers
cal marker for this process.
are formed; platelets are inactivated and clot-
ting factors are inactivated as both are coated
by the soluble monomers.
• Clots that are formed are not stable. Table 18.3  Disseminated Intravas-
cular Coagulation
Although the body attempts to minimize damage
once a DIC event occurs, the physiological inhibitors
Laboratory Profile
protein C, protein S, and thrombomodulin are each • PT 
inactivated. A disorder related to DIC is primary fibri- • aPTT 
nolysis: activation of plasmin within the circulation by • Platelets 
sources other than thrombin activation. In this rare con- • Fibrinogen 
dition, plasmin acts on fibrinogen and fibrin indiscrim- • D-dimer 
inately, therefore hemorrhage is inevitable. Since
Copyright © 2007 by F. A. Davis.

276 PART IV • Hemostasis and Disorders of Coagulation

COND ENSED CASE


A 20-year-old woman came through the emergency Answer
department with unspecified complaints. A CBC The first step that comes to mind is to check the speci-
was ordered and her platelet count was recorded men for clots. Improperly mixed specimens are notori-
as 17.0 × 109/L. A repeat sample was ordered ous for containing small clots. Emergency department
from the emergency department, and with this run, personnel may not be aware that blue-top tubes need to
the platelet count was recorded as 6.0 × 109/L. The be inverted at least five times for proper mixing. This was
patient failed to delta check with her CBC history, done and no clots were observed. Next the technologist
revealing an admission 3 weeks prior with a platelet queried the physician as to whether or not this was an
count of 250 × 109/L. The technologist called the expected result. Although the physician was less than
physician immediately with the report of the cooperative, he did reveal that the patient has undergone
thrombocytopenia and inquired as to the patient a cardiac procedure and that the initial consensus was
history. that the thrombocytopenia was medication induced. The
WHAT ADDITIONAL steps should the technologist patient was admitted and transfused with platelet con-
TAKE to ensure the ACCURACY of this result? centrates, and the platelet count rose to 56 × 109/L. No
additional history is known at this time.

Treatment in Acute Disseminated • Afibrinogenemia, hypofibrinogenemia, and dysfib-


Intravascular Coagulation rinogenemia are all inherited disorders of fibrino-
If the precipitating event leading to the DIC is discov- gen. Each of these may also be acquired disorders.
ered, then successful treatment will involve resolution • Streptokinase is an exogenous fibrinolytic agent,
of this pathology. Surgery in the case of obstetrical com- produced when a bacterial cell product forms a
plications or widespread use of antibiotics in the case of complex with plasminogen.
septicemia may stem the bleeding episode. However, • Naturally occurring inhibitors of fibrinolysis are
because many clinicians are perplexed as to the root plasminogen activator inhibitor 1 and alpha-2-
cause of the precipitating events, judicious use of blood antiplasmin.
products will stop the bleeding. Fresh frozen plasma is a
• The byproducts of fibrinolysis are fibrin degradation
source of all of the clotting factors; packed red cells will
products and D-dimers.
restore oxygen-carrying capacity; and platelet concen-
trates will enable clot formation. Heparin has been used • Excess fibrin degradation products provide antico-
in DIC cases when combined with antithrombin. agulant activity.
Although controversial, this agent may provide needed • D-dimers are produced from a cross-linked and sta-
antithrombotic activity to delay excessive coagulation. bilized fibrin clot.
• Excess D-dimers are an indication that clots have
SUMMARy Points been formed and are being excessively lysed.
• Disseminated intravascular coagulation (DIC) is
• Fibrinogen is the key substrate of the coagulation usually triggered by an underlying pathological
and the fibrinolytic system. event.
• Fibrinogen has the highest molecular weight of all of • In DIC patients will excessively clot or excessively
the clotting factors. bleed, or both.
• Thrombin acts upon fibrinogen to convert it to • Laboratory results for a patient with acute DIC will
fibrin. show a prolonged PT and PTT, decreased fibrinogen
• Fibrin is stabilized by factor XIII and calcium to and platelets, and increased fibrin degradation prod-
become an insoluble clot. ucts and D-dimers.
• Plasminogen is converted to plasmin primarily • Treatment for DIC includes investigating and resolv-
through tissue plasminogen activator and then pro- ing the cause of the disorder and providing blood
ceeds to destroy the fibrin clot. bank products as needed.
Copyright © 2007 by F. A. Davis.

CHAPTER 18 • Fibrinogen, Thrombin, and the Fibrinolytic System 277

Review Questions
1. Which of the following is one of the key roles of c. To restore blood flow at the local level
thrombin with respect to fibrinogen? d. To inhibit coagulation
a. Changes fibrinogen into plasmin
4. Which bacterial cell product will precipitate a DIC
b. Releases fibrin split products
event?
c. Converts fibrinogen into fibrin
a. Neuraminidase
d. Activates factors V and VIII
b. Streptokinase
2. Which of the following laboratory assays will be c. Urokinase
norMAL in a patient with dysfibrinogenemia? d. tPA
a. Immunologic assay for fibrinogen
5. Which is the best possible treatment for a patient
b. Reptilase time
with DIC?
c. Thrombin time
a. Provide supporting blood products
d. PT and PTT
b. Give the patient tPA if there is excessive
3. What is the primary purpose of the fibrinolytic clotting
system? c. Resolve the underlying cause of the DIC
a. To form a stable fibrin clot event
b. To activate the complement system d. Give the patient heparin therapy

CASE STUDY
A 27-year-old man was brought to the emergency department in serious condition. Earlier in the day, he was hiking and
had been bitten on his leg by what he thought was probably a black snake. The leg was swollen, and the hiker was
extremely lethargic and barely conscious. Additionally, he was bleeding from the site where he was bitten. When blood
was drawn, the venipuncture site bled profusely. His lab results follow:
Platelets 27.0 × 109/L (Reference range, 150 to 450 × 109/L)
PFA Not performed
PT 21.2 seconds (Reference range, 11.8 to 14.5)
PTT 53.7 seconds (Reference range, 23.0 to 35.0)
Fibrinogen 110 mg/dL (Reference range, 200 to 400)
D-dimer 3170 ng/mL D-Dimer units (Reference range, 0 to 200)
Given these LABORATORy results WHAT is the most likely DIAGNOSIS? How CAN you ACCOUNT for his LABORATORy results?
Insights to the Case Study
Notice that the patient’s basic coagulation profile was abnormal. His PT and PTT were markedly abnormal, his platelet
count was markedly decreased, his fibrinogen was decreased, and his D-dimer was markedly prolonged. DIC was trig-
gered by the snake bite. The venom of poisonous snakes will directly activate factor X or factor II. When this happens,
clotting occurs within the vessels at an accelerated rate, consuming all of the clotting factors. Notice that the D-dimer
result is extremely elevated. D-dimer is the smallest breakdown product of fibrin. When elevated, it is indicative of cross-
linked fibrin within the circulating blood, rather than locally at the site of injury. The patient was given antivenin and
supported by blood products until his condition stabilized.
[Case submitted by Wendy Sutula, MS, MT(ASCP), SH, Washington Hospital Center.]
Copyright © 2007 by F. A. Davis.

278 PART IV • Hemostasis and Disorders of Coagulation

 TROUBLESHOOTING
WHAT Do I Do When The PATIENT Is Scheduled Based on the mixing study results, one could con-
for Surgery AND the PTT Is ABNORMAL, But He clude that the patient has a factor deficiency. Addition-
Denies Any Bleeding Episodes? ally, the incubated mixing study demonstrated that no
A 24-year-old man had routine preoperative blood slow-acting inhibitor is present. Because only the PTT
work done. Because of the results, he was referred to is affected, the most likely factor would be one or more
the hematology service. The young man denied any from the intrinsic pathway (factors XII, XI, IX, or VIII;
bleeding problems throughout his life and was taking HMWK; or prekallikrein). The hematologist then
no medications. None of his family members had any ordered factor assays, with the following results:
bleeding problems. A second sample reproduced the Factor VIII 109% activity (Reference range,
results of the first, which were as follows: 55% to 145%)
PT 13.9 seconds (Reference range, 11.8 to Factor IX 121% activity (Reference range,
14.5) 61% to 140%)
PTT 168.6 seconds (Reference range, 23.0 to Factor XI 86% activity (Reference range,
35.0) 65% to 135%)
Factor XII 33% activity (Reference range,
The patient’s PTT is extremely elevated. Three 50% to 150%)
questions come to mind. Is the patient on heparin? Is As can be seen from the laboratory data, this
there a circulating anticoagulant present? Does the patient was factor XII deficient. Unlike for factors VIII,
patient have a congenital acquired factor deficiency? A IX, and XI, patients with a factor XII deficiency do not
thrombin time was performed in the unlikely event have bleeding problems. Factor XII–deficient patients
that the patient was somehow receiving heparin (most tend to have very long PTTs, however, because the clot-
likely, low-molecular-weight heparin, which can be ting time of a PTT is dependent on the in vitro activa-
administered on an outpatient basis). The thrombin tion of factor XII. Similar to HMWK and prekallikrein
time was normal, so the hematologist then ordered a deficiency, factor XII–deficient patients may even have
PTT mixing study. a tendency toward thrombosis. This young man had
Mixing study: Immediate PTT = 32.9 his surgery with no complications.
50:50 mix: [Case submitted by Wendy Sutula, MS,
1-Hour incubated 50:50 mix: PTT = 34.3 MT(ASCP), SH, Washington Hospital Center.]

WORD KEY References


1. Loscalzo AD, Schafer AI. Thrombosis and Hemorrhage,
Acrocyanosis • Blue or purple mottled discoloration of the 2nd ed. Baltimore: Williams and Wilkins, 1998:
extremities, especially the fingers, toes, and nose 1027–1063.
Immunologic assay •
Measuring the protein and protein- 2. Falco C, Estelles A, Dalmau J, et al. Influence of lipopro-
bound molecules that are concerned with the reaction of the tein A levels and isoforms on fibrinolytic activity: Study
antigen with its specific antibody in families with high lipoprotein A levels. Thromb
Haemost 79:818–823, 1998.
Monoclonal• Arising from a single cell 3. Liles DK, Knup CL. Disorders of plasma clotting factors.
Recombinant • In genetic and molecular biology, pertain- In: Harmening D, ed. Clinical Hematology and Funda-
ing to genetic material combined from different sources mentals of Hemostasis. Philadelphia: FA Davis, 2002:
497.
Reptilase time • Coagulation procedure similar to throm- 4. Francis CW, Marder VJ. Physiologic regulation and
bin time except that the clotting is initiated by reptilase, a pathologic disorders of fibrinolysis. In: Colman RW, et al,
snake venom; using reptilase, heparin will not affect the assay eds. Hemostasis and Thrombosis: Basic Principles and
Thrombin time • Using thrombin as a substrate, this assay Clinical Practice, 3rd ed. Philadelphia: JB Lippincott,
1994: 1089.
measures the time it takes for fibrinogen to be converted to
fibrin 5. Hosaka Y, Takahashi Y, Ishii H. Thrombomodulin in
human plasma contributes to inhibit fibrinolysis through
Thrombolytic therapy • Using an agent that causes the acceleration of thrombin dependent activation of plasma
breakup of clots carboxypeptidase. Thromb Haemost 79:371, 1998.
Copyright © 2007 by F. A. Davis.

CHAPTER 18 • Fibrinogen, Thrombin, and the Fibrinolytic System 279

6. Fritsma G. Normal hemostasis and coagulation. In: 9. Cunningham VL. A review of disseminated intravascu-
Rodak B, ed. Hematology: Clinical Principles and Appli- lar coagulation: Presentation, laboratory diagnosis and
cations, 2nd ed. Philadelphia, WB Saunders, 2002: 625. treatment. M L O July:48, 1999.
7. Jensen R. The diagnostic use of fibrin breakdown prod- 10. Bick RL, Baker WF. Diagnostic efficacy of the D-dimer
ucts. Clin Hemost Rev 12:1–2, 1998. assay in disseminated intravascular coagulation (DIC).
8. Janssen MC, Sollershein H, Verbruggen B, et al. Rapid Thromb Res 65:785–790, 1992.
D-dimer assay to exclude deep vein thrombosis and 11. Wada H, Mori Y, Okabayashi K, et al. High plasma fib-
pulmonary embolism: Current status and new develop- rinogen levels is associated with poor clinical outcome
ments. Semin Thromb Hemost 24:393–400, 1998. in DIC patients. Am J Hematol 72:1–7, 2003.
Copyright © 2007 by F. A. Davis.

This page has been left intentionally blank.


Hak Cipta © 2007 by F. A. Davis.

Pengantar Thrombosis dan


19 Terapi Antikoagulan

MITRA TAGHIZADEH

Tujuan
Trombosis fisiologis dan patologis Setelah menyelesaikan bab ini, siswa akan mempelajari :
Patogenesis Trombosis
Cedera Vaskular 1. Definisikan trombofilia dan trombosis.
Kelainan Trombosit 2. Tunjukkan faktor risiko yang terkait dengan warisan
Kelainan Koagulasi dan trombosis didapat.
Kelainan Fibrinolitik 3. Buat daftar perubahan hemostatik yang bertanggung jawab
Faktor Antitrombotik (Penghambat Koagulasi) untuk patotrombosis logis.
4. Jelaskan antitrombin, protein C, dan protein S. berkenaan
Gangguan Trombotik
dengan properti, mode tindakan, faktor yang terpengaruh, dan
Gangguan Trombotik yang komplikasi yang terkait dengan defisiensi mereka.
Diwarisi Gangguan Trombotik 5. Buat daftar faktor risiko yang diwariskan untuk trombosis dan
yang Didapat frekuensi kejadiannya.
Diagnosis Laboratorium untuk 6. Buat daftar faktor risiko didapat yang paling umum
Gangguan Trombotik terkait dengan trombosis.
7. Jelaskan resistensi protein C yang diaktifkan dengan berkaitan
Terapi Antikoagulan dengan patofisiologi, cara kerja dan komplikasi terkait.
Obat Antiplatelet Obat 8. Jelaskan trombositopenia yang diinduksi heparin dalam
Antikoagulan Obat berkaitan dengan penyebab, manifestasi klinis pasien, dan
Trombolitik patofisiologi penyakit.
9. Sebutkan tes laboratorium yang digunakan untuk diagnosis
faktor V Leiden dan trombositopenia yang diinduksi heparin.
10. Sebutkan jenis obat antikoagulan yang digunakan
pengobatan gangguan trombotik.
11. Jelaskan mekanisme kerja dari masing-masing anti-obat koagulan
biasa digunakan untuk pengobatan gangguan trombotik.
12. Sebutkan tes laboratorium yang paling umum digunakan
untuk memantau terapi heparinterapi.
13. Sebutkan tes laboratorium yang paling umum digunakan
untuk memantau terapi bersama.

281
Hak Cipta © 2007 by F. A.
Davis.
282 Bagian IV • Hemostasis dan Gangguan Koagulasi

Hiperkoagulan mengacu pada kondisi lingkungan, "Gumpalan putih." Komplikasi yang terkait dengan trombosis
warisan, dan didapat yang mempengaruhi individu untuk arteri adalah penyumbatan sistem vaskular yang menyebabkan
trombosis. Trombosis adalah pembentukan bekuan darah infark jaringan. 1 Faktor penyebab trombosis arteri adalah
di pembuluh darah. Ada dua jenis trombosis yang hipertensi, hiper viskositas, kelainan trombosit kualitatif, dan
diketahui: trombosis arteri dan vena. Trombosis arteri aterosklerosis.
terutama terdiri dari trombosit dengan sejumlah kecil sel Trombosis vena terdiri dari fibrin dan sel darah merah
darah merah dan sel darah putih sedangkan trombosis vena dalam jumlah besar yang menyerupai bekuan darah yang
terdiri dari bekuan fibrin dan sel darah merah. Trombosis dapat terbentuk di dalam tabung reaksi. Trombosis vena
terjadi akibat cedera vaskular, aktivasi trombosit, aktivasi berhubungan dengan aliran darah yang lambat, aktivasi
koagulasi, defek pada sistem fibrinolitik, dan defek pada koagulasi, gangguan sistem fibrinolitik, dan defisiensi
inhibitor fisiologis. Trombosis arteri dan vena bersama inhibitor fisiologis. Komplikasi paling serius yang terkait
dengan komplikasi tromboemboli adalah penyebab kematian dengan trombosis vena adalah pelepasan bekuan darah. Hal
paling penting di negara maju. Lebih dari 800.000 orang ini terjadi ketika bekuan keluar dari tempat asalnya dan
meninggal setiap tahun akibat infark miokard (MI) dan stroke disaring di sirkulasi paru.
trombotik di Amerika Serikat. 1 Juga telah dilaporkan
bahwa penyakit tromboemboli vena adalah penyakit vaskular PATOGENESIS TOMBOSIS
yang paling umum setelah penyakit jantung aterosklerotik
dan stroke. 1 Perubahan hemostatik yang penting dalam patogenesis
trombosis adalah cedera vaskular karena efek toksik
Bab ini akan berfokus pada fisiologi dan patologi
kemoterapi; kelainan trombosit (lebih penting pada trombosis
trombosis, gangguan trombotik, diagnosis laboratorium, dan
arteri); kelainan koagulasi, defek fibrinolitik, dan defisiensi
antikoagulan terapi
faktorantitrombotik.
FISIOLOGI DAN
Cedera Vaskular
THROMBOSIS PATOLOGI
Cedera vaskular memainkan peran penting dalam trombosis
Hemostasis normal mengacu pada respons fisiologis tubuh
terhadap cedera vaskular. Pembentukan bekuan normal dan arteri. Cedera vaskular memulai adhesi platelet ke
pelarutan bekuan dilakukan dengan interaksi antara lima subendotel yang terpapar. Trombosit yang melekat
komponen utama: sistem vaskular, trombosit, sistem melepaskan isi alfa dan butiran padat seperti ADP, kalsium, dan
koagulasi, sistem fibrinolitik, dan inhibitor. Komponen ini serotonin, menyebabkan agregasi trombosit dan pembentukan
harus dalam keadaan fungsional agar hemostasis normal sumbat trombosit. Selain itu, pembekuan darah diawali oleh
terjadi. Ketidakseimbangan pada salah satu komponen di atas faktor jaringan yang dilepaskan dari sel endotel yang rusak.
akan memiringkan skala hemostatik untuk perdarahan atau
Bekuan fibrin yang terbentuk kemudian akan menstabilkan
trombosis. Ada dua sistem hemostasis : sistem
hemostasis primer dan sekunder. Hemostasis primer sumbat trombosit. Cedera endotel vaskular dapat terjadi
mengacu pada proses di mana sumbat trombosit terbentuk di karena cedera sel endotel, aterosklerosis,
lokasi cedera, sedangkan hemostasis sekunder hiperhomosisteinemia, atau gangguan lain yang dapat
didefinisikan sebagai interaksi faktor koagulasi untuk mengganggu aliran darah arteri. Pada pasien kanker, cedera sel
menghasilkan bekuan fibrin terkait silang untuk endotel vaskular dapat terjadi sebagai akibat dari efek toksik obat
menstabilkan sumbat trombosit untuk membentuk
kemoterapi.
trombosis fisiologis.

Kelainan Trombosit
Trombosis fisiologis terjadi akibat respons alami tubuh
terhadap cedera vaskular. Itu terlokalisasi dan dibentuk untuk Trombosit adalah komponen utama trombosis arteri. Saat
mencegah kehilangan darah berlebih. Trombosis patologis trombosit berinteraksi dengan pembuluh darah yang
meliputi trombosis vena dalam, trombosis arteri, dan emboli cedera, terjadi adhesi dan agregasi trombosit. Pada
paru. Trombosis patologis dapat disebabkan oleh kondisi hemostasis normal, aktivasi platelet berlebih dicegah
yang didapat atau diturunkan. Trombosis arteri terutama dengan aktivitas antiplatelet sel endotel seperti pembentukan
terdiri dari trombosit dengan sedikit fibrin dan sel darah prostasiklin. Dalam keadaan penyakit, aktivasi trombosit
berlebih dapat mencerminkan penyakit tromboemboli atau
merah dan putih. Bekuan ini bisa juga disebut sebagai
eksaserbasi trombotik episodes. 1
Hak Cipta © 2007 by F. A.
Davis.
CHAPTER 19 • Pengantar Trombosis dan Terapi Antikoagulan 283

Fisik Tabel 19.2 • Kondisi Terkait


Dengan Warisan
Trombosis
Diwarisi Trombofilia Trombosis
• Defisiensi Protein
• Defisiensi Protein
Diakuisisi • Defisiensi antitrombin
• Protrombin G20210A
Gambar 19.1 Faktor risiko trombosis. • APCR
• Hiperhomosisteinemia
Kelainan Koagulasi
• FaktorVIII yang meningkat
Faktor risiko yang terkait dengan hiperkoagulabilitas dapat • Defisiensi faktor XII
dibagi menjadi faktor lingkungan, diperoleh, atau diwariskan.
torso 1,2 ( Gambar 19.1).
Faktor lingkungan terkait dengan kondisi Regulasi hemostatik yang mendukung peningkatan
sementara yang mungkin terjadi akibat pembedahan, risiko trombosis.
imobilisasi, dan kehamilan atau komplikasi terapeutik yang
terkait dengan kontrasepsi oral, terapi penggantian hormon, Kelainan Fibrinolitik
Fungsi sistem fibrinolitik adalah memecah bekuan
kemoterapi, dan pengobatan heparin.
serat. Seperti pada sistem koagulasi, sistem fibrolitik
Faktor risiko yang didapat berhubungan dengan kondisi
dikendalikan oleh sekelompok inhibitor tertentu.
yang menghalangi hemostasis normal seperti kanker,
Plasmin adalah bentuk aktif dari plasminogen dan
sindrom nefrotik, vasculitis, antiphopolippid antibody
memiliki peran utama dalam pemecahan serat. Plasmin
sminogen dan memiliki peran utama dalam pemecahan
dihambat oleh alfa-2-antiplasmin (penghambat utama
antibodi antifosfolipid, penyakit mieloproliferatif,
plasmin), alfa-2-makroglobulin, alfa-1-antitripsin, AT,
sindrom hiperviskositas, dan lain-lain 1 ( Tabel 19.1). Faktor
dan C1 esterase. Aktivasi plasminogen juga dihambat
risiko yang diwariskan dikaitkan dengan mutasi genetik yang
oleh protein seperti inhibitor aktivator plasminogen I,
mengakibatkan defisiensi inhibitor alami seperti protein C,
II, dan 3 (PAI-1, PAI-2, dan PAI-3) .3 Penurunan
protein S, atau antitrombin (AT); akumulasi factor
aktivitas fibrinolitik, khususnya penurunan kadar
prokoagulan seperti pada protrombin G20210A 1,3; atau
aktivator plasminogen jaringan (tPA ) dan peningkatan
plasminogen I, II, dan 3 (PAI-1, PAI-2, dan PAI-3). 3
kadar PAI-1, menyebabkan gangguan fibrinolisis in
Penurunan resistensi factor pembekuan terhadap aktivitas
vivo, yang mengakibatkan trombosis arteri dan vena.
antikoagulan inhibitor fisiologis seperti pada resistensi
protein C teraktivasi (APCR) (Tabel 19. 2). Kondisi tersebut
mengganggu Faktor Antitrombotik
(Penghambat Koagulasi)
Faktor antitrombotik adalah protein plasma yang
Tabel 19.1 • Kondisi Terkait mengganggu faktor pembekuan sehingga mencegah
Dengan Acquired pembentukan trombin dan trombosis. Tiga jenis
Trombosis inhibitor alami adalah AT, protrombin kofaktor II, dan
protein C.
• Kanker
• Pembedahan (terutama bedah ortopedi) Antitrombin
AT adalah protein plasma yang dibuat di hati. AT
• Penyakit hati
menetralkan aktivitas trombin (IIa), IXa, Xa, XIa, dan
• Imobilitas XIIa. Tindakan penghambatan AT terhadap faktor
• Sindrom nefritik pembekuan lambat; namun, aktivitasnya meningkat
• DIC tajam saat AT berikatan dengan heparin (Gbr. 19.2).
Defisiensi AT dikaitkan dengan trombosis.1,2,4
Hak Cipta © 2007 by F. A.
Davis.
284 Bagian IV • Hemostasis dan Gangguan Koagulasi

Protein S adalah protein yang bergantung pada vitamin


DI HEPARIN K yang dibuat di hati yang diperlukan untuk aktivasi
protein C. Setelah protein C diaktifkan, ia akan
menonaktifkan kofaktor Va dan VIIIa. Kekurangan
DI HEPARIN protein C dan S dikaitkan dengan trombosis.
Kompleks GANGGUAN TROMBOTIK
Serine
Protease
Gangguan Trombotik yang Diwarisi
Gangguan trombotik yang diturunkan dikaitkan dengan
mutasi genetik yang mengakibatkan defisiensi satu
Serine DI HEPARIN atau lebih inhibitor yang terjadi secara alami seperti
Protease
AT, kofaktor II heparin, protein C, dan protein S.
Gambar 19.2 Pengaruh Antitrombin pada protease Peningkatan faktor prokoagulan seperti mutasi
serine. protrombin G20210A dikaitkan dengan trombosis.
Penyebab lain untuk kelainan trombotik yang
Kofaktor Heparin II diturunkan adalah defisiensi AT, protein C dan S,
Heparin kofaktor II adalah penghambat koagulasi faktor V Leiden, atau bentuk hiperhomosisteinemia
lainnya. Ini bekerja melawan trombin, dan itu yang diturunkan yang disebabkan oleh defisiensi enzim
tergantung pada heparin. Defisiensi kofaktor heparin (lihat Tabel 19.2).
saja tidak terkait dengan thrombosis.1
Defisiensi Antitrombin
Protein C dan Protein S Defisiensi AT pertama kali ditemukan pada tahun
Protein C adalah protein yang bergantung pada 1965.1 Gangguan ini diturunkan sebagai kelainan
vitamin K yang dibuat di hati. Protein C bersirkulasi dominan autosomal. Telah dilaporkan bahwa 1 dari
600 orang mengalami defisiensi AT.1 Ada dua tipe
dalam bentuk zymogen. Protein C harus diaktivasi
defisiensi AT: tipe I dan tipe II. Tipe I adalah
menjadi serine protease (protein C teraktivasi) untuk gangguan kuantitatif di mana terjadi penurunan
mengerahkan efek penghambatannya terhadap faktor konsentrasi AT. Tipe II adalah kelainan kualitatif di
pembekuan. Protein C diaktivasi oleh aksi kompleks mana konsentrasi AT normal tetapi molekulnya tidak
trombin-trombomodulin dan protein S sebagai normal secara fungsional. Defisiensi AT dikaitkan
kofaktor1,2,4 (Gambar. 19.3). dengan trombosis vena rekuren, yang dapat mencakup
hampir setiap lokasi vena.1 Peristiwa trombotik
mungkin primer (dengan tidak adanya faktor pemicu)
atau dapat diikuti oleh faktor risiko lain seperti
Trombin
kehamilan, pembedahan, atau faktor risiko lainnya.
faktor yang diperoleh. Defisiensi AT yang didapat
mungkin terkait dengan DIC, penyakit hati, sindrom
Faktor VIII Faktor V
nefrotik, kontrasepsi oral, dan kehamilan.

Defisiensi Heaprin Kofaktor II


Faktor VIIIa Faktor Va Kofaktor II Heparin pertama kali ditemukan pada
tahun 1974. Kofaktor Heparin II diturunkan sebagai
APC APC
sifat dominan autosomal. Ini adalah faktor yang
Protein S Protein S
APC bergantung pada heparin yang efek penghambatannya
terutama melawan trombin. Banyak penelitian telah
menunjukkan bahwa defisiensi kofaktor heparin saja
tidak terkait dengan thrombosis .1
Protein C

Trombin Kekurangan Protein C


Defisiensi protein C diturunkan sebagai sifat dominan
Trombomodulin autosomal. Mirip dengan defisiensi AT, ada dua jenis
defisiensi protein C: tipe I (defisiensi kuantitatif
Sel Endotelium

Gambar 19.3 Jalur Protein C


Hak Cipta © 2007 by F. A.
Davis.
CHAPTER 19 • Pengantar Terapi Trombosis dan Antikoagulan 285

ciency) dan tipe II (de fi siensi kualitatif). Defisiensi faktor V, Arg506Gln, disebut sebagai faktor V
tipe I adalah bentuk yang paling umum dan dikaitkan Leiden.1 Faktor V Leiden adalah penyebab trombosis
dengan penurunan aktivitas imunologis dan fungsional yang diturunkan paling umum pada populasi kulit putih
protein C hingga 50% dari normal. Tipe II dicirikan di Eropa utara dan barat. Di Amerika Serikat, faktor V
oleh jumlah normal protein abnormal.4 Lebih dari 160 Leiden terlihat pada 6% kulit putih.1 Bentuk
mutasi protein C yang berbeda telah dilaporkan antara homozigot dari faktor V Leiden memiliki peningkatan
kedua tipe.1,4 Sebagian besar mutasi adalah mutasi risiko trombosis 80 kali lipat, sementara pembawa
yang salah atau tidak masuk akal. Komplikasi paling heterozigot memiliki peningkatan trombosis 2 hingga
umum yang terkait dengan defisiensi protein C adalah 10 kali lipat.1 Ingatlah bahwa faktor V dan VIII
tromboemboli vena pada orang dewasa heterozigot. diinaktivasi oleh kompleks protein C-protein S. Faktor
Komplikasi lain yang dilaporkan adalah trombosis V yang bermutasi, faktor V Leiden, tidak
arteri, purpura fulminan neonatal pada bayi baru lahir dinonaktifkan dan menyebabkan pembentukan
homozigot, dan nekrosis kulit yang diinduksi gumpalan yang berlebihan. Risiko trombotik semakin
warfarin. meningkat jika faktor risiko lain yang diturunkan atau
Banyak penelitian menunjukkan bahwa sebagian besar didapat hidup berdampingan. Komplikasi trombotik
pasien dengan defisiensi protein C saja tidak yang terkait dengan faktor V Leiden adalah
menunjukkan gejala.1 Temuan ini menunjukkan tromboemboli vena (VTE). Komplikasi lain yang
bahwa episode trombotik dapat dipicu oleh beberapa dilaporkan adalah keguguran berulang. Faktor V
faktor risiko tambahan yang diturunkan atau didapat Leiden juga telah dilaporkan sebagai faktor risiko
pada pasien ini. Defisiensi protein C yang didapat infark miokard. Merokok meningkatkan risiko
mungkin terkait dengan defisiensi vitamin K, penyakit trombosis hingga 30 kali lipat pada individu yang
hati, malnutrisi, DIC, dan terapi warfarin.1 memiliki faktor V Leiden.1 Penyebab lain dari protein
Kekurangan Protein S C yang diaktifkan (8%) terkait dengan kehamilan,
Kekurangan Protein S ditemukan pada tahun 1984.1 kontrasepsi oral, kanker, dan gangguan lain yang
Hal ini diturunkan secara dominan autosom. Protein S didapat (Gambar. 19.4) .
bersirkulasi dalam plasma dalam dua bentuk: bebas
(40%) dan terikat pada protein pengikat C4b (60%). Diagnosis Laboratorium APCR
Aktivitas kofaktor protein S dibawa terutama oleh APCR dapat dievaluasi dengan tes koagulasi, yang
protein bebas S. Seperti AT dan protein C, defisiensi mencakup tes aPTT dua bagian. Prinsip dari pengujian
protein S dibagi menjadi dua jenis. Tipe I adalah tersebut adalah penghambatan faktor Va oleh APC,
kelainan kuantitatif di mana protein total S (bebas dan yang akan menyebabkan perpanjangan aPTT. Oleh
terikat), protein S bebas, dan tingkat aktivitas protein karena itu, aPTT dilakukan pada plasma pasien dengan
S berkurang hingga sekitar 50% dari normal.1 Protein dan tanpa APC. Hasilnya dinyatakan dalam rasio. 1
S tipe II adalah defisiensi kelainan kualitatif dan
Pasien aPTT +APC
terbagi menjadi tipe IIa dan tipe IIb. Pada defisiensi ———
protein S tipe IIa, protein bebas S berkurang Pasien aPTT —APC
sedangkan protein total S normal. Pada tipe IIb, level
protein total dan bebas S adalah normal. 1 Defisiensi
protein S tipe IIb telah dilaporkan pada pasien dengan VII V
faktor V Leiden. Mirip dengan defisiensi protein C,
banyak pasien dengan trombosis memiliki faktor
risiko tambahan yang diturunkan atau didapat.1
Kebanyakan pasien dengan defisiensi protein S VIIa Va
mungkin mengalami trombosis vena. Namun,
trombosis arteri telah dilaporkan pada 25% pasien
dengan defisiensi protein S. Defisiensi protein S dapat APCR APCR
dikaitkan dengan defisiensi vitamin K, penyakit hati,
dan DIC. APC
Resistensi Protein C yang diaktifkan (Faktor V
Leiden)
APCR adalah gangguan dominan autosomal yang
ditemukan pada tahun 1993.1,4 APCR ditemukan
pada 20% sampai 60% pasien dengan trombosis Protein C
+
rekuren tanpa gangguan trombotik yang diwariskan Trombin/ KompleksTrombomodulin
sebelumnya. Mayoritas kasus (92%) diturunkan dan
disebabkan oleh mutasi Gambar 19.4 Jalur resistensi protein C yang diaktifkan
Hak Cipta © 2007 by F. A.
Davis.
286 Bagian IV • Hemostasis dan Gangguan Koagulasi

Rentang referensi bervariasi dari lab ke lab tetapi Hiperhomosisteinemia dapat diturunkan atau
secara umum rasio normal adalah 2 atau lebih besar. didapat. Homosistein adalah asam amino yang terbentuk
Kisaran <2 adalah diagnostik. selama konversi metionin menjadi sistein. Hasil
aPTT menurun bila APC ditambahkan ke plasma hipermosisteinemia baik dari defisiensi enzim yang
normal. Plasma dari pasien dengan APCR memiliki diperlukan untuk produksi homosistein (bentuk yang
rasio yang lebih rendah daripada rentang referensi diwariskan) atau defisiensi vitamin kofaktor (B6, B12,
yang ditetapkan untuk pasien normal. Tes DNA dan folat) dalam bentuk yang didapat. Peningkatan
tersedia untuk memastikan mutasi titik spesifik pada kadar homosistein dalam darah dilaporkan menjadi
pasien dengan faktor V Leiden. faktor risiko stroke, MI, dan gangguan trombotik.1,3
Gangguan sistem fibrinolitik seperti defisiensi
Mutasi Protrombin plasminogen, defisiensi tPA, dan peningkatan inhibitor
Mutasi protrombin (G20210A) adalah penyebab aktivator plasminogen berhubungan dengan penyakit
paling umum kedua dari bentuk hiperkoagulasi yang trombotik.
diturunkan. Ini disebabkan oleh mutasi titik tunggal.
Ini adalah kelainan autosom dominan yang Gangguan Trombotik yang Didapat
menyebabkan peningkatan konsentrasi protrombin Ada banyak situasi yang dapat menyebabkan gangguan
plasma. Risiko tromboemboli vena meningkat karena trombotik didapat. Mereka mungkin terkait dengan
kadar protrombin plasma meningkat ke tingkat yang penyakit yang mendasari seperti kanker, pembedahan,
lebih besar dari 115 IU / dL.1 Seperti pada faktor V penyakit hati, sindrom nefrotik, DIC, kehamilan, dan
Leiden, mutasi protrombin cenderung mengikuti defisiensi vitamin K. Obat-obatan seperti kontrasepsi
distribusi geografis dan etnis dengan prevalensi oral atau terapi penggantian hormon dapat
tertinggi pada kulit putih dari Eropa selatan. . Sekitar mempengaruhi trombosis.
setengah dari kasus dilaporkan di Eropa utara
Mirip dengan faktor V Leiden, episode trombotik Lupus Antikoagulan /Antiphospholipid Syndrome
berkembang lebih awal sebelum usia 40,1 Sindrom antifosfolipid (aPL) adalah kelainan yang
didapat di mana pasien menghasilkan antibodi terhadap
Gangguan Trombotik Turunan Lainnya protein pengikat fosfolipid yang dikenal sebagai beta-2-
Tingkat aktivitas yang meningkat dari faktor VIII glikoprotein I (þ2GPI) atau apolipoprotein (aPL) .5
berhubungan dengan VTE. Telah dilaporkan bahwa Manifestasi klinis dari antibodi aPL berhubungan
jika aktivitas faktor VIII lebih besar dari 150%, risiko dengan trombosis dan janin. kerugian. Subtipe IgG2
VTE meningkat menjadi 3 kali lipat, dan jika aktivitas aPL biasanya dikaitkan dengan trombosis. Episode
lebih besar dari 200%, risiko trombotik meningkat trombotik termasuk trombosis vena dan arteri serta
menjadi 11 kali lipat.1 Defisiensi faktor XII juga tromboemboli. Usia yang biasa pada saat trombosis
terkait dengan trombosis. umumnya sekitar 35 sampai 45 tahun. Pria dan wanita
Faktor XII adalah faktor kontak yang memulai sama-sama terpengaruh.5 Trombosis dapat terjadi
aktivasi jalur intrinsik. Pasien dengan defisiensi faktor secara spontan atau mungkin terkait dengan faktor
XII akan mengalami aPTT yang berkepanjangan predisposisi lain seperti terapi penggantian hormon,
tetapi tidak ada masalah perdarahan. Faktor XII kontrasepsi oral, pembedahan, atau trauma. Sejumlah
memainkan peran utama dalam sistem fibolitik dan kecil pasien dengan antibodi aPL dapat
aktivasi plasminogen ke plasma. Oleh karena itu, memanifestasikan perdarahan jika bersamaan dengan
pasien dengan defisiensi faktor XII akan mengalami trombositopenia atau koagulopati seperti
gangguan fibrinolisis dan rentan terhadap trombosis.3 hipoprotrombinemia.
Disfrinogenemia adalah kelainan bawaan dari Bentuk antibodi aPL yang paling umum adalah
molekul fibrinogen dengan gambaran klinis yang antikoagulan lupus (LA) dan antikardiolipin (ACA).
bervariasi. Dua puluh persen kasus mungkin Manifestasi trombotik mungkin primer (gangguan
menunjukkan trombosis arteri atau vena. Perdarahan autoimun independen) atau sekunder (terkait dengan
telah dilaporkan pada 20% kasus, dan 60% pasien gangguan autoimun lain seperti lupus eritematosus
mungkin asimtomatik sistemik [SLE]). In vitro, LA bekerja melawan uji
Defisiensi inhibitor jalur faktor jaringan (TFPI) koagulasi yang bergantung pada fosfolipid seperti
adalah penanda lain untuk trombosis. TFPI berperan aPTT, yang tidak dikoreksi dengan campuran 1: 1
penting dalam pencegahan pembentukan bekuan dengan plasma normal.4,5 Ini akan dijelaskan di bagian
darah. Ini menghambat kompleks faktor Xa dan faktor selanjutnya. Pasien dengan antibodi aPL mungkin
VIIa-TF.3. Defisiensi inhibitor ini berhubungan datang dengan
dengan gangguan tromboemboli.
Hak Cipta © 2007 by F. A.
Davis.
CHAPTER 19 • Pengantar Terapi Trombosis dan Antikoagulan 287

trombosis dan keguguran. Perdarahan jarang terjadi,


kecuali pasien mengalami trombositopenia atau Gambar 19.5 Patofisiologi HIT
penurunan protrombin juga.
apy. Sekitar 36% hingga 50% pasien dengan HIT
Uji Laboratorium untuk mengembangkan trombosis yang mengancam jiwa.
Antibodi Antifosfolipid Kecenderungan trombotik dapat berlangsung setidaknya
Tes umum yang digunakan untuk mendeteksi selama 30 hari. Trombosis vena (trombosis vena
antikoagulan lupus adalah aPTT, Kaolin clotting time ekstremitas) lebih sering terjadi daripada trombosis
(KCCT), encerkan Russell viper venom test arteri. Komplikasi lain dari HIT termasuk
(DRVVT), dan encerkan PT. Untuk aPTT dan trombositopenia, lesi kulit yang diinduksi heparin (10%
DRVVT jangka panjang, studi pencampuran harus sampai 20% pasien), dan resistensi heparin.1
dilakukan. Dalam studi pencampuran, plasma pasien Patogenesis HIT adalah bahwa antibodi diproduksi
dicampur dengan plasma normal dan tes diulang. untuk melawan kompleks faktor 4 heparin-platelet.
Dengan adanya antikoagulan lupus, studi Kompleks imun ini mengikat reseptor FC trombosit,
pencampuran tidak benar menjadi normal. menyebabkan aktivasi trombosit, pembentukan
Antikoagulan lupus diperkuat dengan penambahan mikropartikel trombosit, trombositopenia, dan keadaan
trombosit berlebih (uji netralisasi trombosit atau hiperkoagulasi (Gbr. 19.5).
fosfolipid fase heksagonal [Konfirmasi DVV]). 4,6. HIT tidak tergantung pada dosis atau rute
International Society of Hemostasis and Thrombosis pemberian heparin. Kondisi ini harus dicurigai pada
telah merekomendasikan empat kriteria untuk setiap pasien yang jumlah trombositnya turun di bawah
diagnosis antikoagulan lupus: (1) prolonga- tes yang 50% dari nilai dasar setelah 5 hari pengobatan heparin1
bergantung pada fosfolipid, (2) bukti adanya inhibitor dan pada pasien yang mengalami trombosis dengan atau
(studi pencampuran), (3) ) bukti bahwa inhibitor tanpa trombositopenia selama terapi heparin.
ditujukan terhadap fosfolipid (uji konfirmasi), dan (4)
kurangnya inhibitor spesifik lainnya (Tabel 19.3). Diagnosis Laboratorium HIT
Faktor lain yang membantu dalam diagnosis Diagnosis laboratorium HIT mencakup uji fungsional
antikoagulan lupus adalah presentasi klinis karena atau immunoassay. Tes fungsional mengukur aktivasi
pasien ini tidak mengalami perdarahan. Antikoagulan atau agregasi platelet dengan adanya serum HIT dan
lupus dapat hidup berdampingan dengan antibodi heparin. Tes fungsional termasuk agregasi platelet
antikardiolipin pada pasien yang mengalami trombosis yang diinduksi heparin, pelepasan platelet adenosine
didapat dan kematian janin. Oleh karena itu, tes triphosphate (ATP) yang diinduksi heparin oleh
ACLA juga direkomendasikan. ACLA dideteksi lumiaggre- gometry, pelepasan 14C-serotonin assay
dengan metode ELISA.6 Antibodi terdeteksi lainnya (14C-SRA) oleh ELISA, dan pembentukan
adalah anti-þ2GPI.6 mikropartikel platelet dengan aliran sitometri. Antibodi
faktor 4 heparin-platelet dideteksi dengan ELISA. Jika
Trombositopenia yang diinduksi Heparin dicurigai HIT, heparin harus dihentikan segera dan
HIT adalah komplikasi yang dimediasi kekebalan diganti dengan obat antikoagulan alternatif
yang terkait dengan terapi heparin. HIT dapat (danaparoid, arga- troban). Warfarin harus dihindari
berkembang pada 3% sampai 5% pasien yang pada fase akut trombosis karena dapat menyebabkan
menerima heparin tak terpecah. HIT biasanya organ vena ekstremitas.1 Pasien yang menerima
berkembang antara 5 dan 14 hari setelah terapi heparin harus memiliki basis-
heparin. Antibodies

Gambar 19.3  Kriteria Untuk Diagnosis


PF4 + Heparin
Antikoagulan Lupus

• Perpanjangan setidaknya satu uji ketergantungan Platelet activation


fosfolipid
• Kurangnya koreksi studi pencampuran
• Koreksi hasil abnormal dengan penambahan
fosfolipid berlebih
• Kekurangan inhibitor spesifik lainnya PF4 release
Hak Cipta © 2007 by F. A.
Davis.
288 Bagian IV • Hemostasis dan Gangguan Koagulasi obat-obatan, obat antikoagulan, dan obat trombolitik.
Obat antiplatelet mencegah aktivasi dan penggabungan
jumlah baris trombosit dan pemantauan trombosit platelet dan paling efektif dalam pengobatan penyakit
setiap hari ketiga antara 5 dan 14 hari arteri. Obat antikoagulan menghambat pembentukan
trombin dan fibrin dan biasanya digunakan untuk
DIAGNOSIS LABORATORIUM UNTUK pengobatan trombosis vena. Obat trombolitik digunakan
GANGGUAN TROMBOTIK untuk memecah gumpalan fibrin, memulihkan fungsi
Ketersediaan berbagai tes untuk mengevaluasi vaskular, dan mencegah hilangnya jaringan dan organ.
keadaan hiperkoagulasi telah meningkatkan diagnosis
kejadian trombotik yang diturunkan dan didapat.
Namun, pengujiannya mahal dan memakan waktu. Antiplatelet Drugs
Tes laboratorium ini harus dipertimbangkan untuk Ada banyak agen yang digunakan untuk melawan
pasien yang hasil tesnya akan mempengaruhi pilihan, trombosit. Aspirin (asam asetilsalisilat) adalah obat
intensitas, dan durasi terapi antikoagulan, keluarga antiplatelet yang mempengaruhi fungsi platelet secara
berencana, dan prognosis. permanen dengan menghambat enzim siklooksigenase
Peristiwa klinis yang membenarkan evaluasi (COX) dan dengan demikian pembentukan tromboksan
laboratorium dari keadaan hiperkoagulasi tercantum A2 (TXA2). TXA2 adalah zat pengaktif trombosit kuat
dalam Tabel 19.4. Pasien yang tidak memiliki riwayat yang dilepaskan dari trombosit yang teraktivasi.
keluarga yang positif harus dievaluasi untuk Aspirin diserap dengan cepat dari saluran
mengetahui bentuk trombosis yang didapat seperti gastrointestinal dan konsentrasi plasma mencapai
keganasan, gangguan mieloproliferatif, dan antibodi puncaknya 1 jam setelah konsumsi aspirin.1 Efek
aPL.1,7 Pemeriksaan laboratorium tidak boleh aspirin pada trombosit dimulai 1 jam setelah konsumsi
dilakukan pada saat trombosis akut atau saat pasien dan berlangsung selama seluruh masa hidup trombosit
menerima - menggunakan terapi antikoagulan karena (sekitar 1 minggu) .1 Aspirin efektif dalam pengobatan
dapat mempengaruhi hasil pemeriksaan.1 Kadar angina, MI akut, serangan iskemik transien, stroke,
produk degradasi serat dan (D-dimer) meningkat pada fibrilasi arteri, dan katup jantung prostat. Dosis efektif
pasien dengan tromboemboli vena akut. Kurangnya minimum untuk kondisi ini adalah 75 hingga 325 mg /
peningkatan D-dimer pada pasien yang dievaluasi hari. 1 Toksisitas aspirin termasuk ketidaknyamanan
untuk DVT akut atau PE memiliki nilai prediksi gastrointestinal, kehilangan darah, dan risiko
negatif yang sangat baik untuk trombosis. perdarahan sistemik. Aspirin dosis rendah (30 sampai
Tes fungsional lebih disukai daripada tes 75 mg / hari) terbukti memiliki efek antitrombotik.1
imunologi. Tes laboratorium skrining yang digunakan Beberapa pasien mungkin mengembangkan resistansi
untuk mengevaluasi pasien yang dicurigai memiliki terhadap aspirin. Pasien yang menjadi resisten terhadap
keadaan hiperkoagulasi dirangkum dalam Tabel 19.5. aspirin memiliki tingkat serangan jantung dan stroke
yang lebih tinggi. Resistensi aspirin dapat dievaluasi
dengan tes agregasi platelet.
ANTICOAGULANT THERAPY
Obat antiplatelet lainnya termasuk dipyridamole,
thienopyridines, ticlopidine, dan clopidogrel.1
Penyakit tromboemboli diobati dengan obat
antitrombotik. Agen antitrombotik termasuk
antiplatelet

Gambar 19.4  Kondisi itu memerlukan Gambar19.5  Laboratorium Skrining


Evaluasi Untuk Status Hiperkoagulasi Tes Untuk Hyper-Keadaan Koagulasi

• Trombosis berulang pada pasien berusia <45 • Resistensi protein C teraktivasi


tahun • Tes fungsional untuk antitrombin, protein C, dan
• Pasien dengan riwayat keluarga yang positif protein S.
• Trombosis spontan berulang • Protrombin G20210A dengan reaksi berantai
• Trombosis di tempat yang tidak biasa polimerase
• Resistensi heparin • APTT, DRVVT, studi pencampuran, dan tes
• Defisiensi protein C dan S. konfirmasi untuk antikoagulan lupus
• Trombosis yang berhubungan dengan kehamilan • Tes imunosorben terkait enzim untuk antibodi
dan terapi estrogen antikardiolipin
• Kehilangan kehamilan berulang tanpa sebab yang • Aktivitas Faktor VIII
jelas
Hak Cipta © 2007 by F. A.
Davis.
CHAPTER 19 • Pengantar Terapi Trombosis dan Antikoagulan 289

Obat Antikoagulan

Obat antikoagulan digunakan untuk pencegahan dan Lepirudin (rekombinan hirudin), Fon
pengobatan gangguan tromboemboli. Obat daparinux (pentasaccharides), Arga- troban,
antikoagulan jangka pendek seperti heparin diberikan dan Bivalirudin.
melalui infus intravena atau injeksi subkutan. Obat Coumadin
antikoagulan jangka panjang seperti Coumadin Coumadin adalah obat antagonis vitamin K yang
diberikan secara oral. menghambat faktor koagulasi yang bergantung pada
vitamin K. Warfarin adalah turunan Coumadin yang
Heparin
banyak digunakan di Amerika Serikat sebagai obat
Heparin hadir dalam jaringan manusia sebagai
antikoagulan oral. Warfarin menghambat karboksilasi
glikosaminoglikan tersulfasi tinggi yang terjadi secara
faktor yang bergantung pada vitamin K (II, VII, IX, X)
alami. Heparin yang tidak terpecah secara komersial
serta protein antikoagulan yang bergantung pada
(Commercially Unfractionated Heparin / UFH)
vitamin K seperti protein C dan protein S. Waktu paruh
diisolasi dari paru-paru sapi atau usus babi. Ini berisi
warfarin sekitar 36 jam .1 Warfarin diberikan secara
campuran rantai polisakarida dengan berat molekul
oral sebagai antikoagulan jangka panjang. Dosis
4000 sampai 30.000 dalton.1 Heparin sulfat adalah zat
warfarin bervariasi dari pasien ke pasien dan
seperti heparin yang dibuat oleh endotel vaskular.
tergantung pada simpanan vitamin K dalam makanan,
Aktivitas antikoagulan heparin ditingkatkan dengan
fungsi hati, kondisi medis yang sudah ada sebelumnya,
mengikat AT. Kompleks Heparin-AT menonaktifkan
dan obat-obatan yang bersamaan. Terapi warfarin
trombin dan faktor Xa (lihat Gambar 19.2).
dipantau oleh PT / rasio normalisasi internasional
Waktu paruh heparin bergantung pada dosis.
(INR). INR adalah metode untuk menstandarisasi
Heparin dibersihkan dari sirkulasi oleh sistem
pengujian PT terhadap perbedaan dalam reagen
retikloendotelial dan dimetabolisme oleh hati.1
tromboplastin komersial.10 INR ditetapkan oleh
Heparin diberikan dalam dosis yang disesuaikan
Organisasi Kesehatan Dunia (WHO) .1 Setiap reagen
dengan berat badan dengan bolus awal (dosis tinggi)
tromboplastin dikalibrasi terhadap sediaan referensi
diikuti dengan infus kontinyu (dosis rendah).
WHO.INR dihitung menggunakan rumus berikut: INR
Dosis heparin dipantau oleh nilai aPTT berkisar
= (Rasio PT) ISI, di mana ISI mengacu pada indeks
dari 1,5 sampai 2,5 kali rata-rata kisaran normal
sensitivitas internasional, yang dihitung untuk setiap
laboratorium. Kadar aPTT ini setara dengan kadar
reagen tromboplastin terhadap reagen tromboplastin
heparin 0,3 hingga 0,7 U / mL yang dapat diukur
referensi.1
dengan uji aktivitas faktor Xa.
Menurut panel konsensus American College of
Efek merugikan dari heparin termasuk
Chest Physicians, kisaran terapeutik dari INR adalah
perdarahan, HIT, dan resistensi heparin. Resistensi
2.0 hingga 3.0 untuk pengobatan tromboembolisme
heparin dapat terjadi sebagai akibat dari pengikatan
vena. Untuk katup jantung mekanis prostetik dan
heparin yang tidak spesifik dengan protein plasma,
pencegahan MI rekuren, dosis warfarin yang lebih
trombosit, dan sel endotel atau sebagai akibat dari
tinggi diperlukan untuk mencapai INR 2,5 hingga 3,5.1
defisiensi AT.
Efek samping yang paling umum dari terapi Coumadin
Heparin dengan Berat Molekul Rendah adalah perdarahan, yang berhubungan langsung dengan
Heparin dengan berat molekul rendah (LMWH) dosis.10
diturunkan dari UFH melalui pencernaan enzimatik Pasien dengan INR lebih dari 3,0 berada pada
untuk menghasilkan molekul glikosaminoglikan risiko perdarahan yang lebih tinggi. Warfarin melintasi
dengan berat molekul yang lebih kecil dan rendah. plasenta dan oleh karena itu harus dihindari selama
Berat rata-rata LMWH adalah sekitar 5000 dalton.1 kehamilan. Komplikasi lain yang jarang tetapi
LMWH memiliki waktu paruh yang lebih tinggi dan menghancurkan dari terapi warfarin adalah nekrosis
memiliki afinitas yang rendah untuk mengikat protein kulit. Fenomena ini sebagian besar terjadi pada pasien
plasma dan sel endotel.1,9 Waktu paruh obat tidak yang menerima warfarin dosis tinggi dan mungkin
tergantung pada dosis. LMWH diberikan secara mengalami defisiensi protein C heterozigot.1 Nekrosis
subkutan, sekali atau dua kali sehari berdasarkan berat kulit disebabkan oleh penurunan cepat protein C pada
badan, dan tidak memerlukan pemantauan.1 LMWH pasien yang telah mengalami defisiensi protein C yang
memiliki efek penghambatan yang lebih tinggi pada mengakibatkan keadaan trombotik.1
faktor Xa daripada pada faktor IIa.9 LMWH Thrombolytic Drugs
dibersihkan oleh ginjal. Reaksi merugikan dari Obat trombolitik umumnya digunakan pada trombosis
LMWH termasuk perdarahan, HIT, atau kepekaan arteri akut untuk trombolisis segera, pemulihan
terhadap LMWH. Obat LMWH tersedia di Amerika integritas vaskular, dan pencegahan jaringan dan
Serikat, yang disetujui oleh FDA, adalah heparinoid, organ.
Hak Cipta © 2007 by F. A.
Davis.
290 Bagian IV • Hemostasis dan Gangguan Koagulan
kerusakan. Kebanyakan obat fibrinolitik dibuat nase tidak spesifik serat, dan karena antigenik,
dengan teknik rekombinan dan dibuat setelah tPA dan dapat menyebabkan reaksi alergi.
urokinase. Urokinase tidak spesifik untuk fibrin dan Perdarahan adalah komplikasi paling umum
menyebabkan hipofbrinogenemia oleh pemecahan yang terkait dengan obat trombolitik. Terapi
fibrinogen. Urokinase dapat digunakan untuk trombolitik tidak memerlukan pemantauan,
pengobatan tromboemboli vena, MI, dan trombolisis namun, tes skrining sebelumnya seperti PT,
kateter yang membeku.1 Streptokinase adalah agen aPTT, TT, fibrinogen, dan jumlah trombosit
trombolitik yang diperoleh dari streptokokus beta- dapat membantu untuk memprediksi pasien
hemolitik. Streptoki- yang berisiko tinggi mengalami perdarahan.

KASUS KONDENSI

Perwakilan teknis untuk laboratorium rujukan mengalami nyeri hebat di belakang lutut kirinya 1 hari
setelah kunjungan ke salah satu akun laboratoriumnya. Dia mencoba untuk menganggapnya sebagai nyeri
otot karena pertandingan bola basket baru-baru ini, tetapi berjalan menjadi sulit baginya. Selama 24 jam
berikutnya, dia memperhatikan bahwa area nyeri menjadi bengkak, merah, dan bahkan lebih sensitif. APA
kesan KLINIS Anda?
Jawaban
Pasien ini mungkin mengalami trombosis vena dalam. Setelah ditanyai lebih lanjut, ditemukan bahwa
pasien telah melakukan jalan raya yang signifikan selama seminggu; sebagian besar waktu menjaga lutut
kirinya dalam posisi tertekuk. Dia akhirnya pergi ke unit gawat darurat, di mana trombosis itu didiagnosis.
Hasil PT dan aPTT-nya normal, tetapi hasil D-dimernya lebih tinggi dari kisaran normal. Venogram
menunjukkan adanya gumpalan di belakang lutut kiri. Pasien dirawat dengan tepat dan mulai dengan
rejimen Coumadin dengan pemantauan pasien rawat jalan yang cermat

dan protein S.
RINGKASAN POINT • Antitrombin dibuat di hati. Ini menghambat
• Hiperkoagulabilitas mengacu pada kondisi yang faktor IIa, IXa, Xa, XIa, dan XIIa. Heparin
menjadi predisposisi seseorang terhadap trombosis. meningkatkan aksi penghambatan antitrombin.
• Faktor risiko yang terkait dengan hiperkoagulabilitas • Protein C adalah protein yang bergantung
dapat dibagi menjadi faktor lingkungan, didapat, atau pada vitamin K yang dibuat di hati. Protein C
diturunkan. diaktivasi oleh kompleks trombin-
• Trombosis adalah pembentukan gumpalan darah di trombomodulin. Protein S adalah kofaktor
pembuluh darah. Trombosis bisa arteri atau vena. untuk aktivasi protein C. Protein C yang
• Trombosis arteri terutama terdiri dari trombosit diaktifkan menonaktifkan faktor Va dan VIIIa.
dengan sedikit sel darah merah dan sel darah putih, • Faktor risiko yang diturunkan terkait dengan
sedangkan trombosis vena terdiri dari bekuan fibrin mutasi genetik yang mengakibatkan defisiensi
dan sel darah merah inhibitor alami seperti protein C, protein S,
• Trombosis bisa terjadi akibat cedera vaskular, atau antitrombin; akumulasi faktor
aktivasi trombosit, aktivasi koagulasi, defek sistem prokoagulan seperti pada protrombin
fibroid, dan defek pada inhibitor fisiologis. G20210A; atau resistensi faktor pembekuan
• Trombosis fisiologis terjadi akibat respons alami terhadap aktivitas antikoagulan inhibitor
tubuh terhadap cedera vaskular. Itu terlokalisasi dan fisiologis seperti pada resistensi protein C yang
dibentuk untuk mencegah kehilangan darah berlebih. diaktifkan.
• Trombosis patologis meliputi trombosis vena dalam, • Mayoritas (92%) kasus resistensi protein C
trombosis arteri, dan emboli paru. Trombosis teraktivasi diturunkan dan disebabkan oleh
patologis dapat disebabkan oleh kondisi yang didapat mutasi faktor V Arg506Gln, yang disebut
atau diturunkan. sebagai faktor V Leiden.
• Tromboemboli terbentuk saat bekuan keluar dari
tempat asal dan disaring di sirkulasi paru.
• Antikoagulan fisiologis adalah protein plasma dan
termasuk antitrombin, kofaktor II heparin, protein C,
Hak Cipta © 2007 by F. A.
Davis.
Chapter 19 • Pengantar Terapi Trombosis dan Antikoagulan 291

• Gangguan trombotik yang didapat berhubungan • Trombositopenia yang diinduksi heparin


dengan penyakit atau obat yang mendasari. (HIT) adalah komplikasi trombotik yang
• Sindrom antifosfolipid disebabkan oleh antibodi dimediasi oleh imun yang berhubungan dengan
terhadap tes koagulasi yang bergantung pada terapi heparin. Antibodi tersebut diproduksi
fosfolipid seperti aPTT, yang tidak dikoreksi dengan untuk melawan kompleks heparin-platelet
campuran 1: 1 dengan plasma normal. Bentuk faktor 4.
antibodi aPL yang paling umum adalah antikoagulan • Tes diagnostik untuk HIT termasuk tes
lupus (LA) dan anticardiolipin (ACA). aktivasi platelet yang bergantung pada heparin
• Tes laboratorium untuk LA termasuk aPTT atau dan deteksi antibodi dengan ELISA.
dRRVT; studi pencampuran, dan studi konfirmasi.
Antibodi antikardiolipin diuji oleh ELISA.

STUDI KASUS
Seorang wanita berusia 30 tahun dirujuk ke rumah sakit untuk evaluasi. Dia disajikan dengan riwayat beberapa aborsi
spontan. Dia saat ini mengeluh sakit dan bengkak di paha kirinya. Riwayat keluarganya dan riwayat medis masa
lalunya biasa-biasa saja. Pasien saat ini menggunakan kontrasepsi oral. Hasil laboratorium pasien adalah sebagai
berikut:

WBC 8.0 × 10 9 / L (Rentang referensi, 4.4 hingga 11.0)


RBC 4.7 × 10 12 / L (Rentang referensi, 4.1 hingga 5.1)
Hgb 14,0 g / dL (Rentang referensi, 12,3 hingga 15,3)
Hct 43% (Rentang referensi, 36 hingga 450) 250 × 10 9 / L
Trombosit (Rentang referensi, 150 hingga 4000)
PT 13,5 detik (Rentang referensi, 10,9 hingga
aPTT 12,0) 52 detik (Rentang referensi, 34 hingga
dRVVT 38) Lama
aPTT 1: studi pencampuran 1 Tidak segera diperbaiki dan setelah inkubasi 2 jam.
dRVVconfirm Dikoreksi
ACA Menyajikan
Wawasan untuk Studi Kasus
Diagnosis antikoagulan lupus dibuat berdasarkan temuan fisik, riwayat pasien, dan hasil laboratorium.
Penemuan fisik menunjukkan bahwa pasien mengalami beberapa kali kehilangan janin dan mengalami nyeri
serta bengkak di pahanya pada saat evaluasi medis. Kurangnya riwayat keluarga yang positif dengan trombosis
mengesampingkan gangguan trombotik bawaan. Jumlah trombosit normal, menunjukkan bahwa episode
trombotik tidak terkait dengan penyebab aktivasi trombosit.
aPTT dan dRVVT keduanya memanjang; namun, pasien tidak mengalami masalah perdarahan. APTT dan dRVVT
yang berkepanjangan tanpa adanya perdarahan menyingkirkan adanya defisiensi faktor pembekuan. Studi
pencampuran dengan plasma normal membedakan defisiensi faktor dari inhibitor. Kurangnya perdarahan
menyingkirkan penghambat faktorVIII. Antikoagulan lupus adalah

(dilanjutkan mengikuti HALAMAN)


Hak Cipta © 2007 by F. A.
Davis.
292 Bagian IV • Hemostasis dan Gangguan Koagulan

(LANJUTAN)
terhadap tes yang bergantung pada fosfolipid in vitro. Dalam tes konfirmasi dRVVT, kelebihan fosfolipid ditambahkan ke
sistem tes untuk menetralkan antibodi lupus dan oleh karena itu memperbaiki dRVVT berkepanjangan yang awalnya
dilakukan. Tes netralisasi platelet adalah tes konfirmasi lain yang digunakan untuk konfirmasi antikoagulan lupus. Tes ini
digunakan untuk koreksi aPTT yang berkepanjangan pada pasien dengan antikoagulan lupus. Antikoagulan lupus termasuk
dalam kelompok antibodi yang disebut antibodi antifosfolipid (ACA), yang meliputi antikoagulan lupus dan antibodi
antikardiolipin. Antikoagulan lupus dapat hidup berdampingan dengan ACA pada beberapa pasien. Oleh karena itu, penting
untuk menguji kedua antibodi tersebut jika antikoagulan lupus dicurigai. Antibodi ACA dapat dideteksi dengan ELISA dan
positif pada pasien ini.

Tinjau Pertanyaan
1. Penghambat utama sistem fibrinolitik adalah:
a. Sebuah. antiplasmin. c. Sebuah. mutasi faktor V.
b. protein S. d. . penghapusan faktor VIII.
c. antitrombin.
d. protein C. 7. Kompleks trombin-trombomodulin diperlukan untuk:
a. Sebuah. aktivasi protein C.
2. Uji Venom Viper Russell's encer (dRVVT) sangat b. aktivasi antitrombin.
membantu dalam diagnosis: c. aktivasi protein S.
a. HIT d. aktivasi faktor V dan VIII.
b. penghambat faktor VIII.
c. antikoagulan lupus. 8. Trombositopenia yang diinduksi heparin disebabkan
d. ACA oleh:
a. Sebuah. antibodi terhadap faktor trombosit 4.
3. Antikoagulan lupus ditujukan untuk : b. antibodi terhadap kompleks faktor heparin-platelet 4.
a. Sebuah uji koagulasi yang bergantung pada c. antikoagulan lupus.
fosfolipid. d. antibodi terhadap heparin.
b. faktor VIII.
c. fibrinogen. 9. Manakah dari obat berikut yang akan membuat seseorang
d. faktor pembekuan yang bergantung pada vitamin berisiko mengalami trombosis?
K. a. Sebuah. Aspirin
b. Dipiridamol
4. Pernyataan mana yang benar tentang Coumadin? c. Streptokinase
a. Sebuah. Ini digunakan untuk pengobatan gangguan d. Kontrasepsi oral
perdarahan.
b. Ini bekerja pada faktor XII, XI, IX, dan X. 10. Manakah dari hasil berikut ini yang benar tentang
c. Ini digunakan untuk terapi jangka pendek. penghambat lupus?
d. Ia bekerja pada faktor pembekuan yang bergantung pada a. Sebuah. APTT berkepanjangan pada plasma yang
vitamin K. tidak diencerkan dan campuran 1: 1 plasma pasien
dengan plasma normal
5 . Pernyataan mana yang benar tentang protein C? b. Koreksi aPTT pada campuran plasma pasien 1: 1
a. Sebuah. Ini adalah kofaktor untuk protein S. dengan plasma normal setelah inkubasi 2 jam
b. Aktivitasnya dihambat oleh heparin. c. APTT murni yang tidak diencerkan dan aPTT yang
c. Ini membentuk kompleks dengan antitrombin. berkepanjangan pada campuran plasma pasien 1: 1
d. Ini adalah penghambat fisiologis koagulasi. dengan plasma normal
d. APTT normal yang tidak diencerkan dan campuran
6. Resistensi protein C yang teraktivasi dikaitkan 1: 1 dari plasma pasien dengan plasma normal
dengan:
a. Sebuah. mutasi faktor VIII.
b. penghapusan faktor VI.
Hak Cipta © 2007 by F. A.
Davis.
Chapter 19 • Pengantar Terapi Trombosis dan Antikoagulan 293

 PENYELESAIAN MASALAH
kisaran 1,5 sampai 2,5 kali rata-rata
Apa Yang Saya Lakukan Saat Hasil Lab kisaran normal yang ditetapkan oleh
Menunjukkan bahwa pasien Tidak Mengalami institusi. Dalam studi kasus, PTT
Heparin? pasien hampir tidak berubah bahkan
setelah 48 jam terapi heparin. Ada
Sampel koagulasi dari unit perawatan intensif beberapa kemungkinan untuk skenario
diberikan ke laboratorium pada shift malam. ini. Kemungkinan pertama yang
Pasien mengalami banyak trauma akibat terlintas dalam pikiran adalah
kecelakaan mobil. Dia mengalami banyak memeriksa sampel untuk gumpalan
patah tulang dan luka dalam. Kondisinya sangat kecil; meskipun sebagian besar
parah. Terapi heparin dimulai sebagai instrumen koagulasi otomatis
akibat dari beberapa trauma. PT dan aPTT memiliki perangkat penginderaan
yang masuk pasien dalam kisaran normal : gumpalan. Tidak ada gumpalan dalam
PT = 12,0 detik (11 hingga 14 detik) dan PTT sampel ini. Kemungkinan tambahan
= 26 detik (24 hingga 36 detik). Sampel adalah bahwa pasien mengalami
koagulasi terbaru, 2 hari sejak pasien masuk, defisiensi antitrombin sehingga heparin
menunjukkan PTT 32 detik. Unit sebagai antikoagulan tidak akan efektif.
perawatan intensif meminta sampel diulang Namun, pasien dengan defisiensi
karena pasien telah menggunakan heparin antitrombin biasanya rentan terhadap
selama 48 jam. pembentukan gumpalan, dan tidak ada
indikasi hal ini dalam riwayat pasien.
Kasus ini menggambarkan beberapa Berikutnya adalah kemungkinan
kesulitan dengan terapi heparin. Heparin trombositopenia yang diinduksi
ditemukan pada tahun 1916 sebagai heparin, suatu kondisi di mana heparin
polisakarida yang ditemukan di hati. yang tidak terpecah membentuk
Ini mengikat antitrombin membentuk kompleks dengan faktor trombosit IV,
kompleks yang menghambat aktivitas menyebabkan trombositopenia,
faktor pembekuan II, IX, X, XI dan XII. trombosis, dan resistensi heparin. Ini
adalah komplikasi signifikan dari terapi
Antikoagulan terapeutik biasanya heparin yang dapat menyebabkan
diberikan secara intravena, tetapi dapat kematian. Ahli teknologi dalam kasus
diberikan secara subkutan. Pasien ini menanyakan tentang jumlah trombosit
membersihkan heparin secara individual yang diterima pasien dan merujuk
dengan kecepatannya sendiri, dan tidak ada informasi tersebut.
hubungan yang bergantung pada dosis. tion ke ahli patologi.
Waktu paruh heparin adalah 90 menit, dan Dalam tindak lanjut, ditemukan bahwa
sebagian besar waktu heparin diberikan jumlah trombosit pasien telah anjlok
dalam dosis bolus 5000 hingga 10.000 unit, dari jumlah penerimaan 160.000
tergantung pada berat pasien. Heparin dapat
menjadi 60.000 pada pasien. 3 hari.
dipantau oleh PTT dan kurva faktor Xa-
aktivitas. Jika dipantau dengan PTT, Semua heparin yang tidak terpecah
terapeutik umum dihentikan termasuk pembilasan heparin
dari situs intravena. Pasien dimulai
dengan terapi alternatif heparin
dan terus mengalami kemajuan
yang lambat sampai akhirnya sembuh.
Hak Cipta © 2007 by F. A.
Davis.

KATA KUNCI Referensi


1. Deitcher SR, RodgersGM. Trombosis
Angina • Nyeri atau tekanan yang menindas di dada
dan terapi antitrombotik. Dalam: Greer
yang disebabkan oleh aliran darah yang tidak memadai dan JP, dkk, eds.Wintrobe'sClinical
oksigenasi ke otot jantung Hematology, edisi ke-11, Vol 2.
Aterosklerosis • Timbunan kolesterol-lipid-kalsium di Philadelphia: Lippincott Williams dan
dinding arteri Wilkins, 2004: 1714-1750.
2. Selamat malam SH, Griffin JH. Trombofilia
Fibrilasi • Biasanya mengacu pada ungkapan jantung herediter. Masuk: Williams WJ, dkk, eds.
karena salah suplai listrik ke jantung Hematology, edisi ke-6. New York: Mc
Ganggren • Kematian jaringan biasanya akibat suplai Graw-Hill, 2001: 1697–1706.
darah yang tidak mencukupi atau tidak ada 3. EhsanA, Plumley JA. Pengantar trombosis
dan terapi antikoagulan. Dalam:
Nekrosis • Kematian sel, jaringan, atau organ Harmening D, ed. Hematologi Klinis
Purpura fulminans • Bentuk purpura yang berkembang dan Dasar-dasar Hemostasis, edisi
pesat terjadi terutama pada anak-anak; berdurasi pendek dan ke-4. Philadelphia: FADavis, 2002:
535–558.
seringkali berakibat fatal
4. Bauer KA. Gangguan bawaan dari trombosis
Vaskulitis • Radang pembuluh darah. dan fibrinolisis. Dalam: Nathan DG, dkk,
Venogram • Radiografi vena eds. Hematology of Infancy and
Childhood, edisi ke-6, Vol 2. Philadelphia:
WB Saunders, 2003: 1583–1659.
5. Rand JH. Antikoagulan lupus dan gangguan
terkait. Masuk: WilliamsWJ, dkk, eds.
Hematology, edisi ke-6. NewYork:
McGraw-Hill, 2001: 1715–1727.
6. Konkle BA, Palermo C. Evaluasi
laboratorium negara bagian hypercoagulabl.
Masuk: Spandofer J, KonkleBA, Merli
Hak Cipta © 2007 by F. A.
Davis.
294 Bagian IV • Hemostasis dan Gangguan Koagulasi

GJ, eds. Manajemen dan Pencegahan Trombosis di


PrimaryCare.NewYork: OxfordUniversityPress, 2001:
16–25. 9. Merli G. Profilaksis untuk trombosis vena dalam dan
7. BauerKA.Approachtothrombosis. Masuk: LoscalzoJ, emboli paru pada pasien bedah dan medis. Masuk:
Schafer AI, eds. Thrombosis and Hemostasis, edisi ke- Spandofer J, Konkle BA, Merli GJ, eds. Manajemen dan
3. Philadelphia: Lippincott Williams dan Wilkins, Pencegahan Trombosis di Perawatan Primer. New
2003: 330–340. York: Oxford University Press, 2001: 135–147.
8. Rambut WD. Trombosis vena dalam dan emboli paru.
Masuk: KitchensCS, AlvingBM, KesslerCM, eds. 10. Schulman. Antikoagulasi oral. Masuk: Williams WJ,
Konsultatif Hemostasis dan Trombosis. Philadelphia: dkk, eds. Hematology, edisi ke-6. NewYork:
WB Saunders, 2002: 197–221. McGrawHill, 2001: 1777–1786.

You might also like