Sie sind auf Seite 1von 29

Laboratory Evaluation

of Hemostasis
Roger S. Riley, M.D., Ph.D., Ann R. Tidwell, MT(ASCP) SH, David
Williams, M.D., Ph.D., Arthur P. Bode, Ph.D., Marcus E. Carr, M.D., Ph.D.
Table of Contents
CBC/Platelet Count/Blood Smear Examination ________________________
In Vivo Evaluation of Primary Hemostasis _____________________________
Platelet Aggregometry _____________________________________________
Automated Platelet Function Analysis ________________________________
Platelet Aggregation with Impedance Platelet Counting _________
Platelet Aggregation Under Flow Condition ____________________
Acceleration of Kaolin Activated Clotting Time by
Platelet-Activating Factor ________________________________
Automated Optical Platelet Aggregometry
Whole Blood Hemostatometry ______________________________________
Thromboelastography _____________________________________
Clot Retraction ___________________________________________
Clot-Based Assays _______________________________________________
Activated Clotting Time (ACT) ______________________________
Prothrombin Time (PT) _____________________________________
Activated Partial Thromboplastin Time _______________________
Thrombin Time ___________________________________________
Clotting Factor Assays ____________________________________
Fibrinogen Analysis _______________________________________
Plasma Mixing Studies ___________________________________
Reptilase Time __________________________________________
Dilute Russell Viper Venom Assay __________________________
Activated Protein C Resistance ____________________________
Chromogenic Analysis ___________________________________________
Latex Agglutination/Turbidimetry __________________________________
Enzyme Immunoassay ___________________________________________
Flow Cytometry _________________________________________________
Electrophoresis _________________________________________________
Genetic and Molecular Assays ____________________________________
Electron Microscopy _____________________________________________
Radioimmunoassay ______________________________________________
References ______________________________________________________
Table of Contents
4
4
6
8
8
8
9
9
10
10
11
11
12
13
13
14
14
15
16
16
17
17
18
18
19
20
20
22
22
22
3
Introduction
H
e
m
o
s
t
a
s
i
s
Medical evaluation of the hemostasis sys-
tem began with visual observation of the
clotting process. During the time of medi-
cal blood letting, observation of the size of
the clot in a basin (clot retraction) was
used to determine when blood letting had
to be decreased. In the early 20
th
century,
manual timing of whole blood clotting
(i.e., Lee-White Whole Blood Clotting
Time), and later plasma, in glass tubes
permitted a more accurate measurement
of blood clotting. Further discoveries
about hemostasis in the 1930s and 1940s
led to more sophisticated laboratory tests,
including the prothrombin time, activated
partial thromboplastin time, and specific
assays of platelet function and fibrinolysis.
The advent of the monoclonal antibody,
molecular analysis, and the microcom-
puter in the 1980s led to an explosion of
knowledge about hemostasis and hemo-
stasis testing that is still growing. In the
hemostasis laboratory, automated assays
have replaced many of the manual proce-
dure of the past, and there is increasing in-
terest in rapid, point of care hemostasis as-
says for perioperative and critical care, as
well as self-testing to support the millions
of patients now receiving oral anticoagula-
tion for hypercoagulable diseases. Interest-
ingly, measurement of clot retraction is
still the focus of a variety of these tech-
niques, a fact that would no doubt be ap-
preciated by the early physicians. This pa-
per presents a global overview of the tech-
niques presently used in the hemostasis
laboratory, with the realization that many
of these may be quickly surpassed by new
information, developments, and applica-
tions in the near future.
Laboratory Evaluation of Hemostasis
may reveal evidence of liver, renal, or other causes of
acquired platelet dysfunction. A predominance of
large platelets may be the initial clue to the diagnosis
of the Bernard-Soulier syndrome. The May-Hegglin
anomaly, Chediak-Higashi syndrome, and other dis-
eases affecting platelets may be discovered by periph-
eral smear examination.(7)
Platelet Count
Modern hematology analyzers perform a platelet
count by electrical impedance or light scattering
techniques that are accurate to 5% in the range of
1000 - 3,000,000 platelets/!L. A measurement of plate-
let volume (mean platelet volume, MPV) is provided
at the same time, as well as a platelet size distribution
curve. Automated platelet counts can be affected by
platelet aggregates due to spontaneous aggregation,
cold agglutinins, EDTA anticoagulants ("spurious
thrombocytopenia, pseudothrombocytopenia") or
particulate debris, such as red or white cell fragments
("spurious thrombocytosis").(2-4) In addition, hema-
tology analyzers may overestimate the platelet count
in severe thrombocytopenia.(5) Therefore, confirma-
tion of atypical platelet counts by manual inspection
of a peripheral smear is essential. If necessary, plate-
let counts can be performed in a hemocytometer by
phase contrast microscopy to an accuracy of 10-
20%.
In Vivo Evaluation of
Primary Hemostasis
The Ivy skin bleeding time is an imprecise manual
screening assay of primary hemostasis that was
widely utilized in the past as a diagnostic assay for
patients with suspected bruising and bleeding disor-
ders, as a therapeutic guide in actively bleeding pa-
tients, and as a predictor of hemorrhage in the gen-
CBC/Platelet Count/Peripheral
Blood Smear Examination
The complete blood count (CBC), platelet count, and
peripheral blood smear examination are the most
fundamental assays of hemostasis and must be per-
formed in all patients with suspected hemostatic ab-
normalities.
Peripheral Blood Smear Examination
Peripheral smear examination is the critical first step
in the investigation of any suspected hematologic
disease.(6) Peripheral smear examination reveals in-
formation about platelet size, gross morphology, and
granularity, as well as associated abnormalities in red
and white blood cells. It is also helpful for confirma-
tion of the automated platelet count. An estimate of
the platelet count can be obtained by routine light
microscopy of a Wright's-stained peripheral smear by
multiplying the number of platelets per 1000x oil
magnification oil immersion field by 10,000, or more
accurately, by multiplying the sum of the number of
platelets counted in 8-10 fields under 1000 x oil mag-
nification by 2000.(7) A visual platelet counting tech-
nique based on the white blood cell count (PCW,
platelet count based on WBC) has also been devel-
oped for thrombocytopenic samples.(8) Every pe-
ripheral blood smear should be carefully evaluated
for the presence of platelet clumps that may falsely
lower the platelet count. Platelet aggregates usually
indicate a poorly collected or anticoagulated blood
specimen of the presence of EDTA-induced
autoantibodies.(7)
Acquired thrombocytopenia secondary to leukemia,
myeloproliferative disorders, or other hematologic
diseases is more common than congenital platelet
disorders. In addition, peripheral smear examination
eral population of patients undergoing surgery or
invasive procedures.(9)

Bleeding times are performed directly on the patient
by phlebotomists or technologists who are trained
and experienced in this assay. A blood pressure cuff is
placed on the upper arm and inflated to 40 mm Hg to
provide uniform capillary pressure, and a standard-
ized incision is made on the volar surface of the fore-
arm with a standard cutting device, such as the Sur-
4
Fig. 1. Photomicrograph of a normal peripheral
blood smear showing several platelets with normal
morphology (Arrows).
Platelet Count, Bleeding Time
Laboratory Evaluation of Hemostasis
from the incision with filter paper at 30-second inter-
vals until bleeding ceases. The result is reported in
seconds as the bleeding time.(10; 11)
The bleeding time is determined by many physiologic
factors, including skin resistance, vascular tone and
integrity, and platelet adhesion and aggregation.
Thus, a prolonged bleeding time may reflect an in-
trinsic platelet function defect, von Willebrand dis-
ease, vascular anomaly, or medications that affects
platelet function, such as aspirin. If the actual bleed-
ing time exceeds the expected bleeding time by five
minutes, a platelet function defect may be suspected.
Unfortunately, the precision, accuracy, and repro-
ducibility of the bleeding time are severely impaired
by factors such as the thickness and vascularity of the
skin, the location of the incision, skin temperature,
wound depth, and patient anxiety. Because of its im-
precision, the bleeding time must be used with ex-
treme caution in a patient care setting. The US Food
& Drug Administration no longer accepts bleeding
time data in patients as a surrogate marker for the
evaluation of new hemostatic drugs, and it is no
longer indicated for the preoperative screening for
hemostatic defects.(12-15) The routine utilization of
the bleeding time for the diagnostic evaluation of
patients with von Willebrand disease, storage pool
disorder, and other hereditary mucocutaneous hem-
orrhagic diseases has been questioned.(16) The
Fig 2. Example of optical and impedance platelet
counts with an automated hematology analyzer (Cell-
Dyne 4000). In the optical technique (upper histo-
gram), platelets (arrow) are discriminated from other
cells by light scatter at 7
o
and 90
o
. An upper volume
threshold is used to separate platelets from micro-
cytic red blood cells. In the impedance platelet count
(bottom histogram), platelets are differentiated from
other cells by electrical resistance. The mean platelet
volume (MPV) is determined from the platelet vol-
ume data provided by impedance measurements.
gicut

(International Technidyne Corp, Edison, NJ) and
the Triplett and Tip Tripper Bleeding Time Devices
(Helena Laboratories, Beaumont, TX). Blood is wicked
bleeding time has been entirely discontinued
at some medical institutions without a
measurable adverse affect on patient
care.(13)
5
Fig 3. Performing the bleeding time. Upper photo-
graph: A bleed pressure cuff was placed over the up-
per arm and the skin of the forearm cleaned with
alcohol. Middle photograph: Picture of skin incision
marks left after a template was applied. Blood is
starting to ooze from the wound. Bottom photo-
graph: Wicking the wound with filter paper to de-
termine the bleeding time.
Bleeding Time
Laboratory Evaluation of Hemostasis
costly assay restricted to specific clinical circum-
stances. A variety of commercial instruments and
reagents for platelet aggregometry are available from
Chrono-Log Corporation (Havertown, PA), Bio/Data
Corporation (Horsham, PA), and Helena Laboratories
(Beaumont, TX).
Glanzmann thrombasthenia and the Bernard-Soulier
syndrome are the best known inherited anomalies of
platelet surface receptors, although both diseases are
very rare. Glanzmann thrombasthenia arises from an
aberration in the most prevalent platelet surface re-
ceptor, GPIIbIIIa (specific binding site for fibrino-
gen), leading to moderate to severe bleeding prob-
Platelet Aggregometry
Conventional platelet aggregometry (light transmis-
sion aggregometry, turbidimetric aggregometry)
measures the in vitro response of platelets to various
chemical agents (i.e., aggregating agents, platelet ago-
nists) that induce platelet functional responses.(17)
In the clinical laboratory, platelet aggregometry is
utilized for the diagnosis of inherited and acquired
platelet disorders, the assay of von Willebrand factor
activity (ristocetin cofactor assay) and for the diag-
nosis of heparin-induced thrombocytopenia.(18)
Conventional optical platelet aggregometers are
modified spectrophotometers that measure light
transmission through platelet-rich plasma (PRP).
Although the turbidity of fresh PRP limits light
transmission, transmission progressively increases as
platelet aggregation causes the formation of larger
and larger particles.(17) More recent innovations
include whole blood aggregometers and lumi-
aggregometers. Whole blood aggregometers require
less patient blood and provide faster turn-around
time than optical aggregometers. Lumi-
aggregometers simultaneously measure platelet ag-
gregation and ATP secretion to provide a more accu-
rate diagnosis of platelet function defects. The plate-
let agonists routinely used in the clinical laboratory
to differentiate various platelet function defects in-
clude adenosine diphosphate (ADP), epinephrine,
collagen, ristocetin, and arachidonic acid. Other ago-
nists, such as thrombin, vasopressin, serotonin,
thromboxane A2 (TXA2), platelet activating factor,
and other agents are used by research and specialized
clinical laboratories.
Conventional platelet aggregation is a complex labo-
ratory assay that is particularly sensitive to the assay
conditions, as well as drugs and other substances in
the blood.(19) Because of these influences, platelet
aggregometry is an advanced, manually intense,
lems in affected individuals. Platelet aggregometry
reveals a lack of response to agonists requiring fi-
brinogen binding, including adenosine diphosphate
(ADP), epinephrine, arachidonic acid, and collagen. In
contrast, the aggregation response to ristocetin is
within normal limits. The Bernard-Soulier syndrome
is clinically similar, but arises from the absence of
another functionally important platelet surface recep-
tor, GPIb-V-IX. However, platelets from patients with
the Bernard-Soulier syndrome show normal aggrega-
tion to agonists requiring fibrinogen binding, but
show a lack of response to agents requiring GPIb (i.e.,
thrombin, ristocetin plus von Willebrand factor). The
6
Platelet Aggregometry
Primary
Aggregation
Maximal
Aggregation
Shape
Change
Dilution
Secondary
Aggregation
Time
L
i
g
h
t

T
r
a
n
s
m
i
s
s
i
o
n
Fig 4. Platelet aggregometry. The
curve shows the five stages of an
ideal response of platelets to the
addition of a platelet agonist. Fol-
lowing addition of the agonist, the
platelets undergo a shape change
after a short delay. This is fol-
lowed by the release of stored
agents, resulting in primary ag-
gregation. The synthesis and re-
lease of new agonists occurs after
another short delay, producing a
second wave of aggregation.
Eventually, maximal aggregation
has occurred and light transmis-
sion is at is lowest. In practice,
aggregation studies are per-
formed with platelet-rich plasma
and a variety of agonists (i.e.,
ADP, epinephrine, arachidonic
acid, collagen, ristocetin, throm-
bin, etc.). A conventional com-
mercial platelet aggregometer
(PACKS-4, Platelet Aggregation
Chromogenic Kinetics System-4)
is shown in the upper right.
Laboratory Evaluation of Hemostasis
Heparin-induced, immune-mediated thrombocy-
topenia (HIT type II) is an unfortunate, but relatively
common complication of heparin therapy arising
fromautoantibodies specific for a complex of heparin
and platelet factor 4 (PF4). The IgG/heparin/PF4 im-
mune complexes bind to the FcyRIIA (CD32) receptor
on the platelet membrane, resulting in platelet activa-
tion, the release of additional PF4, new immune com-
plexes, and rapid platelet consumption. The excess
PF4 also binds to glycosaminoglycans on endothelial
cells, leading to antibody-mediated endothelial dam-
age, thrombosis, and disseminated intravascular co-
agulation. Since serum from patients with HIT can
aggregate normal platelets in the presence of heparin,
platelet aggregometry with heparin is often used to
confirm the clinical suspicion of HIT.(20; 21) How-
ever, due to the operational complexity of this assay
and its relatively low sensitivity, this assay has been
Bernard-Soulier syndrome is also characterized by
thrombocytopenia and large platelets, while the plate-
let count and morphology are normal in Glanzmann
thrombasthenia but clot retraction is absent. These
two separate but specific defects in essential platelet
surface components have provided valuable informa-
tion on the role(s) of platelets in formation of the
initial hemostatic plug.
largely replaced by enzyme immunoassay and flow
cytometry. As a combinatorial strategy, the immuno-
assay can be used as a screening tool, with the aggre-
gometry test for confirmation in patients that are
antibody-positive.
The ability of vWF to aggregate platelets in the pres-
ence of the antibiotic ristocetin is the basis for the
ristocetin cofactor assay, the most common labora-
tory method to measure vWF activity for the diagno-
sis and monitoring of von Willebrand disease.(22)
This assay is performed by incubating formalin-fixed
platelets with test plasma, adding ristocetin, and then
performing platelet aggregation. The results are in-
terpolated from a standard curve prepared from ag-
gregation slopes obtained with testing of dilutions of
normal pooled plasma. Due to the time consuming
manual nature of the classic ristocetin cofactor assay,
7
Disorder Collagen
Epinephrine
ADP Arachidonic
Acid
Ristocetin
Bernard-Soulier
Disease
Normal Normal Normal Absent
Glanzmann
thromboasthenia
Absent Absent Absent Normal
Aspirin, many
drugs
Reduced or absent Variable Reduced or absent Normal
Storage pool
disease
Reduced or absent Variable Variable Normal
vWD, Type I Normal Normal Normal Reduced or absent
vWD, Type IIb Normal Normal Normal Increased
Fig. 5. Effect of aspirin on platelet function. Diagram
shows aggregation tracings (% aggregation vs. time)
for platelet-rich plasma from a donor who had recently
ingested aspirin. The aggregation response to aspirin
is markedly decreased to arachidonic acid (10% final
aggregation). Epinephrine (76%), ADP (79%), and col-
lagen (103%) show essentially normal responses.
0 1 2 3 4 5
Minutes
-20
0
20
100
40
60
80
%

A
g
g
r
e
g
a
t
i
o
n
Arachidonic acid
E
p
in
e
p
h
r
in
e
ADP
C
o
lla
g
e
n
Table I
Platelet Aggregometry - Characteristic Findings in Different Diseases
Laboratory Evaluation of Hemostasis
tion with clopidogrel or NSAISs in elective cardiac
surgery patients., monitoring the efficacy of therapy
with platelet GpIIb-IIIa antagonists in patients un-
dergoing percutaneous coronary intervention or re-
ceiving medical therapy for non-ST elevation acute
coronary syndromes., and predicting post-operative
blooding and blood product utilization in patients
undergoing cardiac surgery with cardiopulmonary
bypass.
Platelet Aggregation
under Flow Conditions
The PFA-100 (DADE-Behring, Miami FL, USA) is a
rapid, automated laboratory instrument that is sensi-
tive to quantitative and qualitative abnormalities of
platelets and von Willebrand factor (vWF). In the
PFA-100, citrated whole blood is aspirated from a res-
ervoir under constant vacuum conditions through a
microscopic 150 um aperture.(31-36) This aperture is
cut into a biologically active nitrocellulose membrane
in a disposable cartridge device coated with a combi-
nation of platelet agonists. These agonists are either
collagen (fibrillar Type I equine tendon) and epi-
nephrine (C/Epi) or collagen and adenosine-5-
diphosphate (C/ADP). The blood is forced through
the aperture at a high shear rate (5000-6000 seconds
-
1
) that roughly corresponds to the flow conditions
present in small arteries.(32; 33) As the blood is
forced through the aperture, platelets undergo adher-
ence, activation and aggregation on the membrane
surrounding the aperture and progressively form a
plug that finally occludes the aperture. The closure
time (CT) is the time required for the complete occlu-
sion to occur.
The PFA-100
TM
is more rapid and less expensive than
the bleeding time for the evaluation of platelet
function.(35; 37) Since there is a good correlation
between the bleeding time and the PFA-100 in certain
patient populations, there, there is a trend to replace
automated agglutination techniques are under evalu-
ation(23; 24), as well as techniques using enzyme
immunoassay.(25-28) The aggregation test as cur-
rently performed has a large standard deviation,
which is unfortunate considering that von Willebrand
disease is the most common hemostatic disorder en-
countered in the hematology clinic.
Automated Platelet
Function Analysis
The manual, laborious nature of conventional platelet
aggregometry is unsuitable for many applications
where point of care and/or rapid testing is indicated.
Therefore, there is increasing interest in non-
complex, automated techniques of platelet function
analysis particularly suitable for the cardiovascular
suite, cardiovascular laboratory, dialysis, or intensive
care unit.(29) A number of innovative techniques are
presently available, and more are likely forthcoming
in the near future.
Platelet Aggregation
with Impedance Platelet Counting
Plateletworks (Helena Laboratories, Beaumont, Texas)
is a rapid in vitro point of care platelet aggregation
screening technique based on impedance platelet
counting and specifically developed for cardiopul-
monary bypass and cardiac catheterization
settings.(30) The technique uses anticoagulated blood
to measure the change in platelet count due to plate-
let aggregation. Two separate samples of blood are
taken, including one containing ADP and collagen
platelet agonists. The platelet count is measured in
each tube using a small impedance hematology ana-
lyzer, and the percent aggregation is calculated. An
eight-profile hematology profile is provided at the
same time.(30) The Plateletworks assay has been re-
cently used to monitor the reversal of platelet inhibi-
the bleeding time with the PFA-100
TM
for a first-line
screening test for platelet dysfunction in patients
undergoing preoperative evaluation. Other clinical
applications of the PFA-100 include the following:
The non-specific identification of patients with
inherited platelet dysfunction, including
Bernard-Soulier syndrome, Glanzmanns
thrombasthenia, and other diseases.(38)
The evaluation of women with menorrhagia to
exclude platelet dysfunction.
The determination of aspirin resistance, aspirin
hyperresponsiveness, and the assessment of
8
Automated Platelet Function Analysis
Fig. 6. Schematic diagram of PFA-100 instrument.
Citrated blood is forced through a small mem-
brane at high shear rate meant to simulate physi-
ologic conditions. Platelet agonists on the mem-
brane initiate platelet adhesion and aggregation
that eventually occlude the membrane and stop
the flow of blood (Closure time). Diagram from
DADE-Behring.
Laboratory Evaluation of Hemostasis
the end point (collagen-induced thrombosis forma-
tion). At the time of this writing, the CSA is no longer
being commercially developed but has important
features which are not found on other available in-
struments.
The Platelet-Stat (Precision Haemostatics, Inc., Clovis,
CA) is a physiologic in vitro simulation of the tem-
plate bleeding time, using blood anticoagulated with
acid-citrate-dextrose (ACD). The device consists of a
membrane with a slit, similar to the template-induced
injury. Blood is forced at constant pressure from a
syringe through the slit, resulting in occlusion of the
slit as a platelet plug is formed. The time from the
start of blood flow through the slit until blood clot-
ting at the slit is termed the bleeding time. Phase I
studies show that the in vitro bleeding time (Platelet-
Stat) is successful in predicting dysfunctional plate-
lets. The Platelet-Stat has been successfully used to
diagnose TTP and monitor therapy with plasma
exchange.(48)
Acceleration of Kaolin Activated
Clotting Time by Platelet-Activating Factor
The hemoSTATUS (Medtronic, Minneapolis, MN) is
an automated system designed for whole blood
point-of-care platelet function testing, especially in
cardiovascular surgery. The assay principle is a com-
parison of the activated clotting time quantitated in
cartridges containing different concentrations of kao-
lin or kaolin and platelet-activating factor.
The system also provides quantitative analysis of
heparin concentration by heparin/protamine titra-
tion, as well as a base-line clotting time (platelet-
activated clotting time). Clinical evaluation of the
instrument has been controversial, with several stud-
ies failing to demonstrate a correlation of results with
perioperative blood loss or an adequate sensitivity to
drugs affecting platelet function.(49-52)
patient compliance with aspirin and other anti-
platelet receptor agents during therapy.(39-41)
Monitoring deamino-D-arginine (DDAVP) ther-
apy in vWD patients belonging to subsets of
vWD that are responsive to DDAVP including
most type 1 and some type 2 patients.
There are several cavets in the clinical utilization of
the PFA-100. Strict adherence to specimen require-
ments, specimen transportation, and specimen proc-
essing is required, since the PFA-100 is affected by
critical pre-analytical variables such as hematocrit or
platelet count, blood collection technique, and trans-
portation through pneumatic tube systems.(42) Since
the PFA-10 has been reported as insensitive to some
patients with platelet function defects, clinical corre-
lation is critical, with follow-up with a different
screening technique in cases of high clinical
suspicion.(16; 38) The PFA-100
TM
is insensitive to al-
terations in the quantity or quality of fibrinogen and
therefore has not been shown to be useful in evaluat-
ing patients for the presence of dysfibrinogenemia or
hypofibrinogenemia. It is not sensitive to defects or
deficiencies in the classic coagulation factors and
appears to have little if any significant utility in as-
sessing Hemophilia A and B.
The Clot Signature Analyzer (CSA, Xylum Corpora-
tion, Scarsdale, NY) is an automated in vitro instru-
ment designed to simulate in vivo clotting and plate-
let function under physiological conditions using
unanticoagulated whole blood.(43-47) In the CSA,
blood flow is passed through two channels. In the
punch channel, shear-induced platelet activation is
simulated by two small (0.015 cm) holes punched in a
blood conduit, causing a pressure drop in the lumen
until closure of the punch holes occurs (platelet he-
mostasis time). The collagen channel incorporates a
small aperture with a collagen fiber immobilized at
the center of the aperture. Platelets adhere to the col-
lagen and eventually close the aperture, representing
Automated Optical Platelet
Aggregometry
A recent innovation is the development of optical
platelet aggregometry for point of care analysis using
microbead agglutination technology. The VerifyNow
System (Accumetrics, San Diego, CA) consists of a
small optical analyzer and disposable, single-use as-
say cartridges that contain all necessary reagents,
including fibrinogen-coated microbeads. The patient
sample of 3.2% citrated whole blood is automatically
dispensed from the blood collection tube into the
assay cartridge without operator intervention. Assay
devices for the monitoring of aspirin and anti-GP Iib/
IIIa receptor antagonists (i.e., abciximab and eptifiba-
tide) are commercially available, and an assay to
monitor Clopidogrel (Plavix) therapy is under devel-
opment. To date, the VerifyNow assay has been pri-
marily used to measure aspirin resistance in patients
with coronary artery disease.(53; 54)
One instrument is especially marketed for the detec-
tion of GPIIb/IIIa receptor blockade in patients
treated with the platelet antagonist abciximab. The
Ultegra Accumetrics RPFA uses a turbidimetric opti-
cal detection system to measure the agglutination of
fibrinogen-coated microparticles in in anticoagulated
whole blood. In the assay, platelets with unblocked
GPIIbIIIa receptors are activated and cause micropar-
ticle agglutination with a change in optical light
transmission.(55; 56) However, a recent study did not
confirm the sensitivity of the Accumetrics RPFA in
comparison to conventional platelet aggregometry of
the Platelets assay.(57)
Whole Blood Hemostatometry
Thromboelastography, measurement of platelet con-
tractile force, and related procedures are analytical
techniques to measure the global process of coagula-
9
Automated Platelet Function Analysis
Laboratory Evaluation of Hemostasis
sample is placed in a shallow cup and is trapped be-
tween parallel surfaces when an upper plate is low-
ered onto the upper surface of the forming clot (Fig.
1). The upper surface is attached to a strain gauge
transducer. As the clot forms and the platelets pull
within the network, a downward force is transmitted
to the upper plate and transducer. The downward
force stresses the transducer and a voltage propor-
tional to the distance moved is generated. Since the
transducer actually measures distance moved, a cali-
bration constant relating distance moved to force is
used to convert distance to force. Early work with this
device confirmed that the forces produced by plate-
lets (platelet contractile force, PCF) in platelet rich
plasma or whole blood clots were significant (several
kilodynes in magnitude) and easily measured.(68)
The onset of force development occurred as soon as
the fibrin network was in place. Utilizing this new
technique, PCF was found to be directly dependent on
platelet count, to be sensitive to temperature and cal-
cium concentration, but to be relatively independent
of fibrinogen concentration over the normal fibrino-
gen range of 100 to 400 gm/dL.(69) PCF is also a very
stable parameter, that persists in whole blood stored
at room temperature for as long as ten days. In con-
trast, platelet function by conventional aggregometry
must be performed within four to six hours. The ro-
bust nature of the parameter and its absolute de-
pendence on platelet viability have led some groups
to examine the use of the PCF parameter as a marker
of platelet survival in stored and modified platelet
preparations.(70)
The thrombin generation time is another parameter
measurable by the Hemodyne. This is performed by
the use of Batroxobis, a snake venom proteolytic en-
zyme from the fer-de-lance that directly clots fibrino-
gen via cleavage of fibrinopeptide A. The addition of
batroxobin to citrated whole blood results in rapid
clot formation, but no initial PCF development. Al-
though batroxobin does not activate platelets, after a
tion (i.e., primary hemostasis to fibrinolysis) using
whole blood. Although this technology was originally
developed decades ago, there has been a recent resur-
gence of interest due to the increasing need for im-
mediate information in critically ill patients and
those undergoing liver transplantation, cardiovascu-
lar surgery, and other procedures where rapid hemo-
static changes occur.(58-63)
Thromboelastography
The conventional (rotational) thromboelastograph
uses a sample cuvette cup filled with native (unanti-
coagulated) whole blood to measure clot formation/
dissolution kinetics and the tensile strength of the
clot. A pin suspended from a torsion wire is lowered
into the cuvette and the cup is rotated through a 45
o

angle over a period of time. Torque from the rotating
cup is transmitted from the pin and suspending rod
to a recorder. There is no initial torque, but this in-
creases as the clot forms and decreases as fibrinolysis
occurs. More recent thromboelastographs use optical
detection systems to measure the movement of the
rotating pin, as well as computer hardware and soft-
ware for data collection and analysis.(64) Commercial
thromboelastographs include the TEG system Hae-
moscope Corporation (Niles, IL), and the ROTEG
(Pentapharm GmbH, Munich, Germany). Thromboe-
lastography has been extensively used for interopera-
tive cardiopulmonary and near-patient coagulation
monitoring to guide blood product utilization.(64)
Although thromboelastography can be measured in
citrated blood, the results are not compariable to
whole blood.(65)
Clot Retraction
A technology recently developed by Hemodyne, Inc.
(Richmond, VA) the Hemostasis Analysis System
permits direct measurement of the forces produced
in the sample during clot formation.(66; 67) The
variable lag phase PCF development is noted. During
the lag phase, thrombin is generated as a conse-
quence of sample re-calcification. Since the fibrin
network is in place prior to the generation of throm-
bin, PCF becomes apparent as soon as a small
amount of thrombin is generated. Thus, the inflection
or take off point in the PCF curve serves as a marker
of thrombin generation in the batroxobin mediated
assay. Assays of prothrombin fragment 1+2, reveal a
concurrent burst of activation fragment generation at
the moment of PCF upswing.(71) The lag phase is
thus the thrombin generation time (TGT). In normal
individuals, PCF developed by the addition of ba-
troxobin differs only in the time of onset. However, if
thrombin generation is inhibited by the addition of
anticoagulants or by the presence of clotting factor
10
Whole Blood Hemostatometry
Fig. 7. A schematic illustration of the Hemodyne he-
mostasis analyzer used to measure platelet contrac-
tile force and clot elastic modulus. The test specimen
is placed in a sample space between a thermostated
cup and a parallel upper surface. During blood clot-
ting, platelets pull fibrin strands inward, generating a
force that is detected by a displacement transducer
and converted to a voltage proportional to the
amount of force generated. Diagram used with per-
mission of Hemodyne, Inc.
Laboratory Evaluation of Hemostasis
pathways of the coagulation system (Fig. 8). Similar
functional assays have been developed to measure
fibrinolysis and other coagulation pathways.
The clinical coagulation laboratory uses clotting as-
says (prothrombin time, activated partial thrombo-
plastin time) in which tissue phospholipids are added
to platelet-poor plasma as full or partial thrombo-
plastins to to initiate clotting for screening of hemo-
philiac defects or for specific factor assays (Fig. 9).
Instruments for automated performance of clot-
based assays are available from several manufactur-
ers, including Beckman Coulter, Inc. (Fullerton, CA),
Dade Behring (Deerfield, IL), Diagnostica Stago, Inc.
(Parsippany NJ), Global Medical Instrumentation, Inc.
(St. Paul, Minnesota), and Sysmex Corporation (Kobe,
Japan). Several similar assays using whole blood are
available for near-patient testing. The most widely
used of these assays is the activated clotting time
used to monitor clotting during cardiopulmonary
bypass.
Activated Clotting Time (ACT)
The ACT was developed in 1966 as a modification of
the Lee-White whole blood clotting time to monitor
coagulation status and heparinization in immediate
need situations.(52) The ACT uses tubes containing a
negatively-charged particulate activator of coagula-
tion, such as kaolin, celite of diatomaceous earth.
When whole blood is drawn into the tube, the contact
system is activated and clotting occurs. The assay is
useful at high levels of heparin such as used in open-
heart surgery, but is also affected by platelets.(85-88)
The manual ACT has been replaced in recent years by
an increasingly sophisticated variety of
microprocessor-controlled instruments, exemplified
by those manufactured by Helena Laboratories Corp.
(Beaumont, Texas), ITC (Edison, NJ), Medtronics
(Minneapolis, MN), and Roche Diagnostics Corpora-
deficiencies, PCF in the batroxobin clots is dramati-
cally delayed and deficient. TGT is sensitive to the
effects of heparin(72; 73), low molecular weight
heparins(74), dermatan sulfate(75), non-heparin anti-
thrombins(76), inherited clotting factor deficienci-
es(77) and clotting factor deficiencies induced by
warfarin. In vitro studies indicate the potential for
documentation of the correction of deficient throm-
bin generation by hemostatic agents such as recom-
binant FVIIa.(78)
The Sonoclot Coagulation and Platelet Function Ana-
lyzer (Sienco Inc., Wheat Ridge, Colorado) is a versa-
tile, whole blood point of care system that uses a vis-
coelastic clot detection mechanism to analyze the
global process of hemostasis, including coagulation,
fibrin gel formation, clot retraction (platelet func-
tion) and fibrinolysis.(79) The Sonoclot uses the os-
cillation of a tubular probe within a blood sample to
generate an analog electronic signal that reflects re-
sistance to motion during clot formation and fibri-
nolysis. Data processing by a microcomputer gener-
ates a qualitative graph (Sonoclot Signature) as well
as quantitative results on clot formation kinetics and
the rate of fibrin polymerization. A variety of differ-
ent reagent kits are available for general coagulation
monitoring, as well as more specific purposes, includ-
ing heparin monitoring, hyperfibrinolysis screening,
hypercoagulable screening and platelet function
assessment.(80-84)
Each of these instruments has its own distinct fea-
tures and advantages for the diagnostic laboratory,
but a full specific assessment of global hemostasis
defects requires multiple approaches.
Clot-Based Assays
Functional assays based on clot formation as the
endpoint are widely used in the clinical laboratory to
determine the integrity of the intrinsic or extrinsic
tion (Indianapolis, IN). Many of these instruments
perform the PT, aPTT, thrombin time, fibrinogen
level, and other hemostatic assays in addition to the
activated clotting time. Some manufacturers also
provide ACT reagents containing heparinase so that a
patients baseline value can be established in the
presence of heparin. These instruments are increas-
ingly being applied to the near-patient monitoring of
direct thrombin inhibitors and low molecular weight
heparins in critical situations.(89-91)
11
Clot-Based Assays
PLT
PLT
Prothrombin
Fibrinogen
XIIa
XIa
X
Thrombin
Fibrin
XII
XI
IX
VIII
VIIIa
V
Va
VII/VIIa
TF
XIII
XIII
X-Linked
Fibrin
Xa
IXa
Contact
System
Fig. 8. A color-coded schematic illustration of the
coagulation system. The diagram shows components
of the contact system (orange), extrinsic pathway
(blue), intrinsic pathway (magenta), and common
pathway (green). In vivo, platelets (yellow) are essen-
tial for contributing phospholipid and providing a
surface for the tenase and prothrombinase reactions
to occur.
Laboratory Evaluation of Hemostasis
one of a variety of techniques (photo-optical, elec-
tromechanical, etc.)(Fig. 8). The result is reported in
seconds (prothrombin time), or as a ratio compared
to the laboratory mean normal control (prothrombin
ratio, PTR). The PT is critically dependent on the
characteristics of the thromboplastin used in the as-
say, as well as manner of blood coagulation, the type
of container, the type of anticoagulant, specimen
transport and storage conditions, incubation time
and temperature, assay reagents, and the method of
end point detection. This means that patients on
coumadin will have different clotting times when
tested in different laboratories, so a means of stan-
dardization of results must be employed.
The International Normalized Ratio (INR) was intro-
duced by the World Health Organization (WHO) in
the early 1980s as a means of standardizing PT
results.(94) For this purpose, a very responsive batch
of human brain extract was designated as the first
International Reference Preparation (IRP), and a cor-
Prothrombin Time (PT, Protime,
Quicks time, Partial Prothrombin Time)
The PT provides a functional determination of the
integrity of the extrinsic (tissue factor) pathway of
coagulation and is sensitive to the vitamin-K depend-
ent clotting factors (factors II, VII, IX, and X) as well
as to factors of the common pathway (fibrinogen,
prothrombin, factor V, factor X). The PT is a widely
used laboratory assay for the detection of inherited
or acquired coagulation defects related to the extrin-
sic pathway of coagulation, and is the standard test
for monitoring oral anticoagulation therapy
(coumadin).(92; 93)
In the PT an aliquot of test platelet-poor plasma is
incubated at 37
o
C with a reagent containing a tissue
factor, phospholipid (thromboplastin), and CaCl2. The
time required for clot formation is then measured by
rection factor (International Sensitivity Index, ISI)
was developed to correlate the sensitivity of commer-
cial thromboplastin preparations to the IRP. By defi-
nition, the ISI of the first IRP was 1.0. An additional
term, the INR, was introduced to compare a given
prothrombin ratio measurement to the IRP. Commer-
cial vendors of thromboplastin preparations supply
the ISI with each reagent lot. If the ISI is known, the
INR for each clotting time is easily calculated. How-
ever, the ISI can be affected by instrumentation and
other laboratory factors and thus must be verified by
each testing site according to standards of the College
of American Pathologists. Unfortunately, even with
the INR, current prothrombin reagent/instrument
calibration techniques are insufficient to provide
good intralaboratory agreement.(95; 96)
There is great interest in point of care and patient
self-testing of oral anticoagulation status are popular
for patient convenience and to improve the efficiency
of medical care. Considering the 600,000 to 900,000
patients in the United States with heart valves, and
the millions requiring oral anticoagulation for hyper-
coagulability states, it is not surprising that several
small, user-friendly instruments are presently avail-
able for home testing by prescription from blood ob-
tained by fingerstick. These instruments include the
Avocet PT-Pro (Avocet Medical, Inc. San-Jose, CA), the
CoaguChek (Roche Diagnostics, Basal, Switzerland),
the Harmony INR Monitoring System (LifeScan,
Inc., Milpitas, CA) , the INRatio Meter. (HemoSense,
Inc. San Jose, CA),and the HemosProTime Microco-
agulation System (ITC, Edison, NJ), Presently, these
assays are CLIA waived and have been covered by
Medicare since late 2001. Point of care monitoring of
the PT and INR has been the subject of several recent
reviews.(97-103)
12
Clot-Based Assays
Platelet-poor plasma
Incubation
Fibrin clot
Clotting agent, Ca
++
Fig. 8. Basic principle of clot-based assays
of coagulation. A clotting activator, cal-
cium, and a source of phospholipids is
incubated with platelet-poor plasma, re-
sulting in activation of the extrinsic clot-
ting system. The endpoint of the reaction
is the formation of a fibrin clot that can be
measured by visual, photo-optical, elec-
tromechanical means. The result is usually
reported as the time required for clot for-
mation. Common clot-based assays used in
the clinical hemostasis laboratory include
the PT, aPTT, thrombin time, reptilase
time, dilute Russell Viper venom time, and
activated protein C resistance assay. Clot-
based assays are also used for factor
analysis and to determine the presence of
factor deficiencies and anti-factor inhibi-
tors.
Laboratory Evaluation of Hemostasis
failure to promptly mix the blood with the citrate
anticoagulant, improper transport or storage, or a
prolonged interval between specimen collection and
analysis. The sensitivity of the assay to factor defi-
ciencies, inhibitors, and heparin also varies with the
reagents used in the assay. Because of these variables,
a normal aPTT result does not exclude a mild coagu-
lation factor deficiency or the presence of a low-titer
or slow-reacting inhibitor. However, a significant pro-
longation of the aPTT indicates the presence of a fac-
tor deficiency (VIII, IX, XI, XII, prekallikrein, HMWK),
while prolongation of both the PT and aPTT suggests
a deficiency of factor I, II, V, or X. The aPTT is not
affected by deficiencies of factor VII or XIII.
Numerous modifications of the aPTT have been de-
scribed for the functional analysis of specific
coagulation-related substances. Those routinely util-
ized in the coagulation laboratory at the present time
include the reptilase time, the Bethesda assay, protein
C and protein S activity, and several assays for lupus
anticoagulants (dilute Russell viper venom time
[dRVVT], platelet neutralization test, and hexagonal
phospholipid assay.
Specific anti-factor VIII antibodies (inhibitors) are a
serious medical problem for patients with hemo-
philia. Mixing studies can detect the presence of in-
hibitors, but other assays are required for the precise
measurement of antibody activity necessary for pa-
tient care.(121) The Bethesda assay is a modified
aPTT based on the ability of factor VIII inhibitors to
neutralize factor VIII activity in normal plasma. A
series of dilutions of patient plasma are added to a
standard amount of normal plasma and assayed for
factor VIII levels after two hours incubation at 37C:
the titer at which half of the FVIII activity remains is
used to calculate the Betheda units of inhibition.
Several modifications of the Bethesda assay have
been developed to improve its sensitivity.(122-124)
The new Oxford assay is similar, but uses factor VIII
Activated Partial Thromboplastin Time
(aPTT, Activated Prothrombin Time)
The partial thromboplastin time (PTT) is the clotting
time obtained when partial thromboplastin is
added to plasma. Partial thromboplastin is the phos-
pholipid fraction of a tissue extract, and differs from
a complete tissue extract (i.e., thromboplastin) by
the lack of tissue factor. The PTT is sensitive to the
intrinsic pathway of coagulation, but is most sensitive
to the contact factors (i.e., factor XII, prekallikrein,
high molecular weight kininogen) when a particulate
activating agent (i.e., silica, celite, kaolin, mi-
cronized silica, ellagic acid) is added to the reaction
(activated PTT, aPTT). Many different phosophlipid
reagents animal and plant origin, such as cephalin,
have been used as partial thromboplastins, and a va-
riety of activating substances are in use.(104-110)
In the aPTT an aliquot of undiluted, platelet-poor
plasma is incubated at 37
o
C with an activator and
phospholipid (partial thromboplastin). CaCl2. is then
added, and the time required for clot formation is
measured by one of a variety of techniques (photo-
optical, electromechanical, etc.). The aPTT result is
reported as the time required for clot formation after
the addition of CaCl2. The aPTT is functional deter-
mination of the intrinsic (factors XII, XI, IX, VIII, V, II,
I,) and common pathways of coagulation.(111; 112)
The aPTT is utilized to detect congenital and ac-
quired abnormalities of the intrinsic coagulation
pathway, monitor patients receiving heparin or co-
agulation factor replacement therapy, and to detect
inhibitors of the intrinsic and common
pathways.(113-120)
The aPTT clotting time may be influenced by many
pre-analytical and analytical variables and caution
must be used in the interpretation of the result. Pre-
analytical variables include slow or difficult specimen
collection, an improper blood:anticoagulant ratio,
concentrate as the source of factor VIII. Enzyme im-
munoassay, gel techniques, and other methods have
been also used to detect inhibitors.
The direct thrombin inhibitors are among the latest
form of anticoagulant drugs developed with the goal
of eliminating the side effects and improving the
therapeutic efficacy of anticoagulants which exert an
indirect antithrombin effect, including warfarin,
heparin, and low molecular weight heparin.(125) The
present generation of direct thrombin inhibitors in-
cludes recombinant hirudin (lepirudin), bivalirudin,
argatroban, and melagatran. Unfortunately, the direct
thrombin inhibitors present a problem for the hemo-
stasis laboratory, since conventional coagulation as-
says such as the aPTT, thrombin time, and activated
clotting time show poor reproducibility and linearity
in the presence of these drugs.(126) Two modifica-
tions of the aPTT, the ecarin clotting time (ECT) and
prothrombinase-induced clotting time (PiCT) have
been developed for monitoring the direct thrombin
inhibitors, as well as chromogenic and enzyme
immunoassays.(127-129) There is presently no clear
concensus on the most optimal laboratory method
for direct thrombin inhibitor monitoring, although
the automated chromogenic assays and chromogenic-
based point of care assays appear to offer adequate
sensitivity and precision and avoid interference prob-
lems by heparin and other substances.(126; 130-132)
Thrombin Time (Thrombin
Clotting Time, TCT, TT)
The thrombin time measures the thrombin-induced
conversion of fibrinogen to fibrin directly in patient
plasma, bypassing all other clotting factors. The
thrombin time is performed by the addition of a low
concentration of thrombin (usually bovine thrombin)
directly to the citrated plasma and measuring the
time required for the formation of fibrin monomers
by visual, mechanical, or opto-electronic
13
Clot-Based Assays
Laboratory Evaluation of Hemostasis
human plasma deficient (<1%) in the coagulation
factor under study. When the factor-deficient plasma
is mixed with patient plasma in a 1:1 ratio, the PT or
aPTT of the mixture is dependent on the amount of
factor present in the patient plasma. The factor activ-
ity of the patient plasma is determined from a stan-
dard curve, prepared from the PT or aPTT values of
1:1 mixtures of factor deficient substrate and a
serially-diluted reference plasma with known factor
activity. Factors II, V, VII, and X are assayed with the
PT, while the assays for factors VIII, IX, XI, XII, and
the contact factors (i.e., prekappikrein, high molecu-
lar weight kininogen, Passovoy) use the aPTT. The
accuracy of a clotting factor determination is im-
proved by using serial dilutions of patient plasma
and averaging the results. Factor inhibitors may inter-
fere with assay results until sufficiently diluted out.
The measurement of clotting factor activity is essen-
tial to determine the cause of an elevated PT or aPTT,
and to monitor the treatment of patients with known
factor deficiencies or inhibitors. In some patients, the
presence of a weak clotting factor inhibitor is some-
times initially suspected from non-linearity in the
dilution curves.
Fibrinogen Assay
Fibrinogen is the most abundant clotting protein in
the plasma, with a normal plasma level ranging rom
200-400 mg/dL. The quantitative determination of
plasma fibrinogen is essential in the diagnosis and
management of many coagulopathies. In addition,
since plasma fibrinogen levels are increased in some
patients who develop myocardial infarction and
stroke, there is interest in the measurement of fi-
brinogen for thrombotic risk assessment.(141) The
washed clot method (total clottable fibrinogen assay,
World Health Organization method) is the reference
technique for fibrinogen determination. In this tech-
nique, citrated plasma is incubated for an extended
techniques.(133; 134) The thrombin time is prolonged
by thrombin inhibitors and inhibitors of fibrin for-
mation and polymerization, but it is not affected by
problems with thrombin generation. Clinically, the
thrombin time is often used to monitor heparin ther-
apy, and to differentiate heparin effect, hypofibrino-
genemia, dysfibrinogenemia, elevated levels of fibrin
degradation products, and some paraproteins from
other coagulopathies as the cause of a prolonged PT
or aPTT.(135-138) It is also used to monitor heparin
reversal in following cardiothoracic surgery, and to
monitor thrombolytic therapy. Increased plasma fi-
brinogen may also prolong the thrombin time, possi-
bly by interfering with fibrin assembly.(139) The ref-
erence range for the thrombin time is affected by the
source and concentration of thrombin and other
factors.(140)
Heparin is the most common cause of a prolonged
thrombin time. This is confirmed by normalization of
the thrombin time or aPTT following in vitro heparin
neutralization with Heparinase, protamine sulfate,
Heptasorb, or other heparin-neutralizing agents, or
by the performance of the reptilase time. A fibrino-
gen assay, an inhibitor screen, or the dRVVT may be
indicated if heparin effect is not present.
Clotting Factor Assays (Factors II XII;
Contact factors)
The activity of individual coagulation factors are
usually determined in plasma using a one-stage clot-
ting assay. Two-stage and amidolytic (chromogenic
substrate) methods for the determination of factor
activity exist but are rarely used in the United States.
In the past, aliquots of plasma obtained from patients
with hereditary deficiencies of clotting factors were
used for factor analysis, but the supply of some factor
deficient plasmas was very limited, and some con-
tained HIV and/or hepatitis virus. Therefore, the one-
stage assays now use lyophilized, immunoadsorbed,
period of time with thrombin in the presence of ep-
silon aminocaproic acid (EACA) to prevent digestion
of the fibrin clot by plasmin. Other serum proteins
are removed by washing, the clot is dissolved in con-
centrated urea, and the fibrinogen concentration is
measured colormetrically.(142) This technique is un-
suitable for the determination of the large number of
specimens encountered in the clinical laboratory, but,
unfortunately, the accurate and precise measurement
of fibrinogen with the automated coagulometer has
proven difficult. Immunoassays (RID, ELISA, immu-
nonephlometric) for fibrinogen quantitation are also
available but are rarely used.
In spite of their flaws, the von Clauss technique and
the Clotting Rate Assay (Kinetic Fibrinogen Assay,
Prothrombin Time Derived Method) are most widely
used in the clinical laboratories. The von Clauss tech-
nique is based upon the principle that when a high
concentration of thrombin is added to plasma diluted
in buffer (1:5 or 1:10), the effects of clotting inhibitors
are diminished and the clotting time is directly pro-
portional to the level of clottable fibrinogen.(143)
Clotting times of patient plasma are read on a stan-
dard curve made with purified fibrinogen of known
concentration to interpolate a fibrinogen level in the
patient. The assay is accurate in the range of ap-
proximately 50 800 mg/dL. Since the von Clauss
technique requires a high level of technical skill, a
more recent prothrombin time-based kinetic assay is
preferred by many laboratories. In this assay, the rate
of increase in plasma turbidity is measured at 450 nm
during the thrombin-catalyzed conversion of fibrino-
gen to fibrin.(144; 145) This kinetic assay is rapid,
economical, and can be fully automated.(146) Gener-
ally, high levels of heparin or hirudin, but not thera-
peutic levels, can interfere with the clotting assays for
fibrinogen, and patients with known hyperfibrino-
lytic activity will continue to degrade the fibrinogen
in the collected blood sample before testing is com-
pleted unless a special tube is used containing apro-
14
Clot-Based Assays
Laboratory Evaluation of Hemostasis
monoclonal paraproteins, and drugs such as heparin.
Clinical and other laboratory clues are necessary to
identify the inhibitor. For example, lupus anticoagu-
lants are usually not associated with clinical bleeding,
while specific factor inhibitors frequently cause
bleeding. Generally, factor deficiencies produce a
complete correction of the prolonged clotting time
(i.e., corrected to within the normal range), specific
antibodies show very little, if any correction, and
non-specific may show a partial correction, (i.e.,
shortened clotting time but not to within the normal
range). The presence of heparin and other nonspe-
cific inhibitors can be confirmed by other coagula-
tion tests such as the thrombin clotting time and rep-
tilase time, while lupus anticoagulants are identified
by a phospholipid-sensitive test such as the dilute
Russell Venom time (dRVVT). The last clue is pro-
vided by the effect of incubation on the activity of the
inhibitor.

An immediate mixing study is performed by mixing
equal amounts of the "test" plasma with NPP (1:1
mix) and immediately performing a clotting time
(i.e., PT, aPTT, or TT) on the mixed plasma
specimen.(147-149) Most factor inhibitors (except
factor VIII) and a most lupus anticoagulants (fast
reacting inhibitors) produce an immediate clotting
time inhibition and do not require incubation. In
contrast, most factor VIII inhibitors and some lupus
anticoagulants (15%) are weak and/or time depend-
ent (slow reacting inhibitors), and require incuba-
tion of the 1:1 plasma mixture at room temperature
or 37
o
C for one or two hours (incubated mix) to
cause prolongation of the clotting time.(150-152) A
false diagnosis of a factor deficiency can result with-
out incubation, since slow-reacting inhibitors may
correct the immediate mix. Some laboratories also
include a 4:1 aPTT mix (i.e., 4 parts patient plasma, 1
part NPP) to improve the detection of weak inhibitors
that minimally prolong the aPTT (usually 3-5 seconds
above baseline). The markedly prolonged aPTT of
tinin or other plasmin inhibitor. Many studies have
shown that fibrin degradation products cause an
overestimation of the fibrinogen level by the washed
clot and immunologic assays, and an underestima-
tion by the clot-based techniques.(141) The kinetic
assay has also been reported to yield higher fibrino-
gen levels in patients receiving oral anticoagulation
than the von Clauss technique.
Plasma Mixing Studies (Clotting Factor
Inhibitor Screen, Circulating Anticoagulant
Screen)
A prolonged clotting test (i.e., PT, aPTT, and/or
thrombin time) indicates the presence of a factor de-
ficiency or inhibitor of coagulation. The plasma mix-
ing study is the initial step in the evaluation of a pro-
longed clotting time. The goal of a mixing study is to
determine if the prolonged clotting time is shortened
or corrected by mixing the test plasma with equal
volume of normal pooled plasma (NPP; also called
citrated normal plasma, CNP). Even a profound defi-
ciency of a clotting factor, such as the 1% factor VIII
level encountered in severe hemophilia, will be cor-
rected to the normal range by mixing with NPP, since
a 50% level of any factor will still yield a normal clot-
ting time. Factor assays are then performed to iden-
tify the deficient clotting factor.
The failure of a prolonged clotting test to correct in
the mixing study indicates the presence of a inhibi-
tory substance that is preventing clotting from oc-
curring. Unfortunately, this is somewhat difficult to
accomplish since there are several different types of
inhibitors (also called circulating anticoagulants).
Specific inhibitors are immunoglobulins with speci-
ficity for phospholipid ("lupus anticoagulants") or a
specific clotting factor ("factor inhibitors"). Global
or non-specific inhibitors affect more than part of
the clotting process and include fibrin(ogen) degra-
dation products, some pathologic antibodies such as
plasma from a patient with hereditary prekallikrein
deficiency is normalized by prolonged preincubation
(i.e., 10 minutes) of the plasma with aPTT reagent
before the assay is performed. This unique feature of
prekallikrein deficiency is reportedly due to the
aut oac t i vat i on of f ac t or XI I dur i ng
preincubation.(153)
Mixing studies are simple in principle, but can be
difficult to interpret. For example, if the laboratory
range for the aPTT is 24-35 seconds, and the patient
aPTT is 70 seconds, a 1:1 mixing study result of 34
seconds would clearly indicate a factor deficiency,
while a value of 69 seconds would indicate an inhibi-
tor. However, what if the mixing study produced val-
ues of 39 seconds, 51 seconds, or 63 seconds? The
situation is made even more difficult because there
are no official criteria for determining if a correc-
tion has occurred. Furthermore, a number of patient-
specific and laboratory-specific variable can affect
the result and are difficult to compensate for. These
include the biological heterogeneity of anti-factor
antibodies, the presence of drugs and other sub-
stances in the patient specimen, reagent and instru-
ment sensitivity, the source of NPP, the validity of the
laboratory reference range, pre-analytical variables,
and other factors. Therefore, each laboratory pres-
ently establishes their own criteria for interpreting
mixing studies. As summarized by Ledford-Kraemer
(2004), these criteria generally fall into three catego-
ries:
The use of the upper limit of the laboratory ref-
erence range as the correction target. A value,
such as 2SD, 3SD, or within 5 seconds of the
upper limit of the reference range is chosen as
the criteria for correction. A failure of correc-
tion is assumed if this value is not reached.
The use of NPP tested in conjunction with the
patient 1:1 mix. This is particularly valuable to
correct for the decreased activity of the labile
15
Clot-Based Assays
Laboratory Evaluation of Hemostasis
gen molecule, reptilase cleaves only fibrinopeptides A
and AP. The resulting fibrin monomers polymerize
end-to-end to form a fibrin clot. Reptilase has no fi-
brinolytic activity, does not activate plasminogen,
and is not inhibited by antifibrinolytics, thrombin
inhibitors (heparin, hirudin, anti-thrombin antibod-
ies) or antithrombin III.
The reptilase time is used in the evaluation of a pro-
longed aPTT, specifically to exclude the presence of
dysfibrinogenemia. Hypofibrinogenemia and dysfi-
brinogenemia are the usual causes of a prolonged
reptilase time. Prolongation of both the thrombin
time and reptilase time suggests hyopfibrinogenemia
or dysfibrinogenemia. A prolonged aPTT and normal
reptilase time indicates that heparin or other anti-
thrombins is the cause of the prolonged aPTT. Mye-
loma proteins reactive with thrombin may prolong
the reptilase time. Fibrin degradation products
(FDPs) may slightly prolong the reptilase time.
Dilute Russell Viper Venom Assay (dRVVT)
The dRVVT is used to detect lupus anticoagulants
(LA), one type of autoantibody characteristic of pa-
tients with the antiphospholipid antibody
syndrome.(155-157) LA are autoantibodies of the IgG
coagulation factors, V and VIII, during incu-
bated studies. Common criteria for correction
of the patient sample include to within 5 sec-
onds, or to within 10% or 15% of the NPP value.
The Rosner index for the aPTT mixing study
quantitates the amount of correction to the pa-
tient plasma aPTT. A correction is assumed if
the Rosner index is "15.
The Chang percentage, a formula that incorpo-
rates the degree of correction in relation to the
initial aPTT prolongation.
Chang and co-workers performed a detailed evalua-
tion of the sensitivity and specificity of different
methods to define correction of the 1:1 mix.(148; 149)
They found that the three most widely used criteria
for a correction of the aPTT 1:1 mix (upper limit of
normal, NPP aPTT + 5 seconds, Rosner index "15) all
had high sensitivity (88-100%) but low specificity (7-
13%) for detecting a factor deficiency, and low sensi-
tivity (7-15%) and high specificity (88-100%) for de-
tecting an anticoagulant. Using their correction for-
mula and a % correction cutoff at 50%, the immediate
aPTT 4:1 mix had a 75% sensitivity for a factor defi-
ciency and a 91% sensitivity for an anticoagulant. The
corresponding specificies were 91% and 75%. Using
an incubated aPTT 4:1 mix with a cutoff value of >
10% gave sensitivities and specificities of 100% for
both factor deficiencies and anticoagulants. There-
fore, the authors recommend performing immediate
and incubated 1:1 aPTT mixes, with the interpretation
as follows:
Reptilase Time
The reptilase time measures the conversion of fi-
brinogen to fibrin clot by reptilase (Batroxobin,
Atroxin), a thrombin-like enzyme derived from the
venom of the fer-de-lance (barba amarilla, Bothrops
atrox).(135; 136; 154) In contrast to thrombin, which
cleaves fibrinopeptides A, AP, and B from the fibrino-
and IgM classes that interfere with the function of
anionic phospholipids and prolong phospholipid-
dependent clotting tests such as the aPTT and
dRVVT.(158-162) The dRVVT is more specific for LA
than the aPTT since it is not influenced by deficien-
cies of the contact or intrinsic pathway factors or an-
tibodies to factors VIII, IX, or XI.(159; 163; 164) The
coagulant protein in Russells viper venom (RVV) is a
serine protease that directly activates factor X in the
presence of Ca
++
, bypassing the intrinsic and extrin-
sic pathways. The activated factor X then activates
prothrombin (factor II) in the presence of factor V
and phospholipid. In the dilute Russells viper venom
time (dRVVT), phospholipid is diluted to the point
that the clotting time becomes very sensitive to the
presence of substances blocking availability of the
phospholipid surface. The DVVtest is a commercial
reagent (American Diagnostica, Inc., Greenwich, CT)
developed to standardize the dRVVT. Similar reagents
are available from Precision Biologic (Dartmouth,
Nova Scotia) and other vendors. The DVVtest reagent
combines RVV, plant phospholipid, and calcium into
a single reagent. A second reagent, DVVconfirm, con-
tains RVV, extra plant phospholipid, and calcium. The
extra phospholipid in the DVVconfirm reagent is
provided to see if it corrects a prolonged DVVtest
time (by overwhelming the LA). The finding of a pro-
16
Clot-Based Assays
% Correction =
PP PT (or aPTT) - 1:1 (or 4:1) Mix PT (or aPTT)
PP PT (or aPTT) - CNP PT (or aPTT)
X 100
Index =
1:1 Mix aPTT - CNP aPTT
PP aPTT
X 100
Chang Percentage
Rosner Index
Fig. 9. Formu-
las for calcula-
tion of Chang
Percentage and
Rosner Index.
Laboratory Evaluation of Hemostasis
Chromogenic Analysis
Chromogenic analysis is a technique of enzyme
analysis developed in the early 1970s. Chromogenic
assays utilize synthetic substrates comprised of a
colored chemical substance (chromphore, chroma-
gen) linked to a short amino acid residue specific for
the enzyme of interest.(172-174) Enzymatic action
releases the chromophore, which is quantitated by
spectrophotometry. The selectivity of chromogenic
substrates is similar to the native enzyme substrate,
but they are often more sensitive. Other advantages of
chromogenic assays include reagent stability and the
adaptability to a wide range of automated laboratory
instruments, including those used in the chemistry
and immunology laboratories. The selectivity of a
chromogenic substrate to the desired enzyme is af-
fected by the relative concentrations of sample and
reagents, reaction conditions (i.e. pH, temperature,
buffer type and concentration, ionic strength, etc.),
the presence of inhibitors, substrate solubility and
stability, and other factors.(175) The best substrates
have high affinity for the enzyme and a high turnover
rate. The most common substrate is para-nitroaniline
(pNA), which has a maximumabsorption spectrumat
405 nm.
longed dRVVT with patient plasma is presumptive
evidence for the presence of a lupus anticoagulant.
This presumption is confirmed if the dRVVT is not
corrected with a mixture of normal and platelet
plasma, but is corrected by the substitution of plate-
lets for phospholipid. With the DVVtest and

DVVcon-
firm reagents, a DVVtest/DVVconfirm ratio >1.2 is
confirmatory for the presence of LA.
Activated Protein C Resistance
Assay (APCR)
The rapid screening assay for activated protein C re-
sistance for (APCR) is another widely used modifica-
tion of the aPTT. In 1993, Dahlback and coworkers
discovered a mutant clotting Factor V (Factor V Lei-
den) which results in the failure of Activated Protein
C to inactivate Va.(165-168) This defect in the protein
C pathway is associated with a significantly increased
risk of thromboembolic disease. The laboratory di-
agnosis of APCR begins with the rapid screening test,
followed by confirmation with a molecular assay if
the screening assay is positive. In the presently used
modification of Dahlbacks original aPTT-based
screening assay, the test plasma is first diluted with
factor Vdeficient plasma to inactivate therapeutic
concentrations of heparin, correct for coagulation
factor deficiencies, and counteract the effect of some
lupus inhibitors. aPTT assays are then performed
with and without the addition of exogenous activated
protein C (APC).(169-171) The added APC signifi-
cantly prolongs the aPTT in normal individuals, while
patients with APCR show less of an increase. The re-
sults are usually expressed as the ratio of the aPTT
with and without added APC. The modified APCR
screening assay is highly sensitive to factor V Leiden
and most other less common mutations of factor V,
can differentiate heterozygotes from homozygotes,
and is not influenced by heparin or warfarin at
therapeutic concentrations.(170)
The analysis of many coagulation factors utilize
chromogenic substrates for factor X.(176; 177) For
example, factor VIII is an enzymatic cofactor for fac-
tor IX. Activated factor IX causes the activation of
factor X, which then hydrolyzes the chromogenic sub-
strate and releases the pNA chromophore that is read
spectrophotometrically at 405 nm (Fig. 10). If the as-
say conditions are properly controlled, the color in-
tensity reflects the amount of factor VIII. In one com-
parative study of chromogenic analysis, an antigenic
assay, and the one-stage clotting assay for factor VIII,
the chromogenic factor VIII technique was the opti-
mal method, with good precision and freedom from
interference by lupus inhibitors, heparin, or other
anticoagulant drugs.(178)
Chromogenic substrates for thrombin, tissue-type
plasminogen activator, urokinase, coagulation factors
IX, X, and XII, and other substances are commercially
available from Chromogenix (Orangeburg, NY),
(Trinity Biotech Plc, Wicklow, Ireland) and other
companies.
17
Chromogenic Analysis
Factor X
Factor IXa, Ca
++
, Phospholipid
Factor VIII
Chromogenic
substrate
Peptide + pNA
Factor Xa
Fig. 10. A chromogenic method for
the determination of factor VIII
activity. Test plasma is incubated
with calcium, phospholipid, and
excess amounts of purified factors
IX and X. The activated factor X
generated by the reaction hydro-
lyzes a chromogenic substrate,
generating a colored reaction
product that is measured by a
spectrophotometer. The amount of
generated factor Xa is directly
proportional to the concentration
of factor VIII activity.
Laboratory Evaluation of Hemostasis
Turbidimetry and the related technique of nephelo-
metry are extensively utilized in the clinical immu-
nology for the quantitation of a large number of
medically important substances since they are pre-
cise, rapid, and automated. In the coagulation labora-
tory the use of these techniques is more limited. Light
scattering by particles in solution is the basic princi-
ple of turbidimetry and nephelometry. When an im-
mune complex is formed under carefully controlled
conditions, measurement of light scatter can provide
information regarding the quantity of analyte
present.(181) Turbidimetric techniques determine the
reduction in the intensity of incident light from all
interactions of an immune complex with a light
beam, while nephelometric techniques measure light
scattered at a specific angle to the incident beam.
Nephelometry is more sensitive to small particles
than turbidimetry, while turbidimetry more accu-
rately measures large complexes.(182)
Turbidimetry has been used since the early 1950's as
a method of quantitative analysis, particularly for
large immune complexes. In this technique, the
change in light intensity caused by interaction of a
light beam with a suspension of particles is deter-
mined spectrophotometrically. The major use of latex
particle agglutination and turbidimetry in the clinical
coagulation laboratory is the detection and semi-
quantitation of fibrin degradation products (FDPs)
and D-Dimers. Fibrin degradation products (FDPs)
are the result of plasmin degradation of fibrinogen,
fibrin monomers, fibrin polymers or cross-linked
fibrin, while D-dimers are degradation products that
arise specifically from the plasmin degradation of
fibrin crosslinked by Factor XIIIa activity.(183) Thus,
the measurement of cross-linked degradation prod-
ucts (XDPs), unlike total FDPs, is a specific measure
of fibrinolysis. Most turbidimetric assays for D-
dimers utilize latex beads or other microparticles
coated with monoclonal antibody specific for fibrin
D-dimer or the fragment D of fibrin but not with in-
Latex Agglutination
and Turbidimetry
Agglutination is a secondary immune phenomenon
that occurs when insoluble or particulate antigens
(cells or other particles) are cross-linked by an im-
mune reaction. Agglutination occurs because anti-
bodies have two or more antigen recognition sites
(bi- or multivalency). If multiple antigenic recogni-
tion sites are present on a particle, lattices can be
formed that grow in size and eventually become a
mass that is macroscopically visible. The major fac-
tors affecting the agglutination reaction include the
class, affinity and avidity of the antibody, the proxim-
ity and number of binding sites on the particle, the
relative concentrations of antibody and particles,
electrostatic interactions ("zeta potential"), and the
viscosity of the medium. Antibodies of the IgM class,
with ten antigen combining sites, are usually the best
agglutinins, and are more efficient than IgG in ag-
glutination.
Agglutination assays are classified as direct or indi-
rect, depending on whether the analyte is present in
its native state, or linked to a particle (carrier) to al-
low detection of the antigen-antibody reaction. Carri-
ers vary in size from about 0.05 micron to 7 micron,
and may be red blood cells, latex particles, liposomes,
microcapsules, or other particles.(179)
The use of latex particles for immunoassay was first
reported by Singer and Plotz in 1956.(180) Latex par-
ticles are usually coated by passive means, with the
quantity of the adsorbed protein adjusted to provide
agglutination of the analyte in its biological range. In
addition, the use of latex particles avoids much of the
variability encountered with red blood cells. Even so,
the prozone phenomenon can still be significant, and
careful adherence to the manufacturer's instructions
is necessary during the performance of clinical assays
utilizing coated microspheres.
tact fibrinogen, permitting the analysis of whole hu-
man plasma. Elevated D dimers are seen in DIC, pul-
monary embolism, arterial and venous thrombosis,
septicemia, cirrhosis, carcinoma, sickle cell crisis, and
following operative procedures. However, D-dimer
analysis is principally used in the evaluation of pa-
tients with suspected thromboembolic disease, espe-
cially pulmonary embolism and deep vein
thrombosis.(184-188) Both FDPs and XDPs are pre-
sent during late pregnancy and for approximately 48
hours post-surgery. During fibrinolytic therapy the
FDP test is positive, while the D-dimer test is negative
in the absence of thrombolysis. Enzyme immunoas-
say has also been utilized for the detection of
D-dimers.(184; 189-192) Other turbidimetric reagents
are available for the analysis of von Willebrand factor,
free protein S, and other substances.
Nephelometric techniques have been applied very
successfully to the immunochemical measurement of
specific proteins, drugs and other substances.(182;
193) In nephelometry, a known amount of specific
antibody is added to a solution containing the anti-
gen being measured. The intensity of light scattered
from the large antigen-antibody complexes formed
during the reaction is measured, and the rate signal is
transmitted to a microcomputer, where concentration
units are determined. Nephelometry is used by some
clinical laboratories for the quantitation of fibrino-
gen or factor VIII-related antigen.
Enzyme Immunoassay
The enzyme immunoassay (EIA) is a type of non-
isotopic immunoassay in which enzymes, coenzymes,
fluorigenic substrates, or enzyme inhibitors are used
as labels.(194; 195) The major prerequisite for an EIA
is that an antigen or antibody must be linked to an
enzyme without destroying the immunologic or en-
zymatic activity of the antigen-antibody complex. In
solid-phase EIA techniques, the antigen or antibody
18
Latex Agglutination
Laboratory Evaluation of Hemostasis
1970s and rapidly became an essential instrument for
the biologic sciences. Spurred by the HIV pandemic
and a plethora of discoveries in hematology, special-
ized flow cytometers for use in the clinical laboratory
were developed by several manufacturers. The major
clinical application of flow cytometry is diagnosis of
hematologic malignancy, but a wide variety of other
applications exist, such as reticulocyte enumeration
and cell function analysis. Presently, more than
40,000 journal articles referencing flow cytometry
have been published. This brief review of the princi-
ples and major clinical applications of flow cytome-
try may be supplemented by several recent review
articles and books. (199-203)
Prepared single cell or particle suspensions are nec-
essary for flow cytometric analysis. Various immuno-
flurescent dyes or antibodies can be attached to the
antigen or protein of interest. The suspension of cells
or particles is aspirated into a flow cell where, sur-
rounded by a narrow fluid stream, they pass one at a
time through a light beam. Light and/or fluorescence
scatter signals are detected and amplified. The result-
ing electrical pulses are digitized, and the data is
stored, analyzed, and displayed through a computer
system.(203; 204) The end result is quantitative in-
formation about every cell analyzed. Since large
numbers of cells are analyzed in a short period of
time (>1,000/sec), statistically valid information
about cell populations is quickly obtained.
The flow cytometer has been essential for the analy-
sis of platelet structure and function in the research
laboratory. Although the small physical size and bio-
variability of the platelet creates inherent difficulties
for flow cytometric analysis, several clinical assays
are performed by specialized flow cytometry labora-
tories. These assays will achieve more widespread
practice in the near future as standardized techniques
and controls become available. These assays have
been classified by Bode and Hickerson to include
must be bound to a polystyrene test tubes or micro-
titer tray, a particle of polystyrene, latex, or agarose, a
magnetized bead, or another physical support.
Enzymes utilized in immunoassay systems must also
be stable, available in a highly purified state, have a
high turnover rate, and undergo minimal interference
by substances likely to be in the test solution, and be
specific for the substrate. The final reaction product
should be detected by a convenient means with a low
detection limit. The most widely utilized enzyme in
enzyme immunoassay is horseradish peroxidase
(HRP). The substrate of HRP is hydrogen peroxide
(H202) and the product is oxygen. This oxygen pro-
duced during the reaction is used to oxidize a re-
duced, colorless chromagen (usually reduced ortho-
phenylenediamine, OPD). The final product, oxidized
OPD, has a brown color. Most EIA's utilize horserad-
ish peroxidase or alkaline phosphatase as labels, al-
though glucose oxidase, beta-D-galactosidase, and a
wide variety of other enzymes have also been used.
Utilizing fluorimetric techniques, the respective de-
tection limits for HRP, beta-galactosidase, and alka-
line phosphatase are 5, 0.2, and 10 attomol.(196) The
practical detection limit of the EIA is approximately
0.01 to 0.02 attomol of ligand.(197; 198) The enzyme-
Linked Immunosorbent Assay (ELISA) is the most
widely utilized type of enzyme immunoassay.
Enzyme immunoassay is a critical technique in the
clinical laboratory for a wide variety of analytes, in-
cluding both antigens and antibodies. In the hemo-
stasis laboratory, enzyme immunoassay is used for
the quantitation of antigen levels of most clotting
factors, fibrinolytic components, and regulatory sub-
stances.

Flow Cytometry
Flow cytometry is a technique of quantitative single
cell analysis. The flow cytometer was developed in the
platelet surface receptor quantitation and distribu-
tion for the diagnosis of congenital platelet function
disorders, platelet-associated IgG quantitation for the
diagnosis of immune thrombocytopenias and for
platelet cross-matching in transfusion, reticulated
platelet assay to detect stress platelets, fibrinogen
receptor occupancy studies for monitoring the clini-
cal efficacy of platelet-directed anticoagulation in
thrombosis, and the detection of activated platelet
surface markers, cytoplasmic calcium ion measure-
ments, and platelet microparticles for the assessment
of hypercoagulable states.(205)
Flow cytometry is a critical research technique for
the study of diseases of platelet surface receptors, and
has been applied to clinical diagnosis by larger labo-
ratories. The identification of Glanzmann thrombas-
thenia, the Bernard-Soulier syndrome, and even rarer
platelet receptor disorders is performed with panels
of monoclonal antibodies specific for the receptor
antigens under consideration. The use of monoclonal
antibody panels specific for different epitopes is par-
ticularly information in defining the heterogeneity
and extent of disease expression.
Flow cytometry has been utilized to detect both
platelet-associated immunoglobulins of autoimmune
and alloimmune origin. In general, this is performed
by incubating washed platelets with fluorochrome-
labeled antihuman immunoglobulin and quantitating
platelet surface fluorescence. Gating procedures are
used to exclude irrelevant cells and particles. The
differentiation of positive and negative results is de-
pendent upon an adequate negative control, usually
platelets from normal individuals. Flow cytometry is
ideal for the study of low numbers of patients in
thrombocytopenic patients, since it is sensitive and
avoids the platelet activation, with the release of en-
dogenous immunoglobulin molecules.(206) Although
procedural standardization and non-specificity have
limited the use of flow cytometric analysis for platelet
19
ELISA, Flow Cytometry
Laboratory Evaluation of Hemostasis
The recent wealth of new discoveries in the field of
hemostasis and thrombosis has included a number of
antiplatelet agents of value in the therapy of patients
with inherited or acquired hypercoagulability and/or
those undergoing vascular interventional procedures.
As the most prevalent and functionally important
platelet surface receptor, the GPIIbIIIa complex is the
target of several of these agents. Abciximab is a
murine monoclonal antibody that blocks the GPII-
bIIIa receptor to prevent platelet activation. Others,
such as Integrilin and Aggrastat are small peptides
that saturate the receptor. A variety of receptor oc-
cupancy assays have been developed to monitor the
clinical efficacy of these agents. Flow cytometry is the
most sensitive and versatile of these techniques, al-
though it is not a point of care assay at the present
time.(219)
Electrophoresis
Historically, the Laurell rocket assay, radial immuno-
diffusion, and other gel-based procedures were
widely used in the coagulation laboratory for quanti-
tative analysis of certain clotting factor protein levels,
especially the FVIII/vWF complex.. Today, these pro-
cedures have been largely replaced by enzyme immu-
noassay, and other more accurate and efficient meth-
ods, with the exception of von Willebrand factor mul-
timer analysis. This is performed by electrophoresis
on agarose gel containing some acrylamide. The pro-
tein bands are then transferred to nitrocellulose for
Western blot with polyclonal anti-vWF antibody and
finally visualized by radiolabeling, enzymatic detec-
tion, or chemiluminescence.(220; 221) The pattern of
multimeric units and satellite bands differentiates
von Willebrand disease into type I, type IIA, B, M, and
N, and type III. Multimer analysis is a specialized
procedure only performed by a few coagulation refer-
ence laboratories, but it is sometimes critically im-
portant to subtype von Willebrand disease to avoid
associated immunoglobulin in idiopathic thrombo-
cytopenia purpura (ITP), the technique appears more
clinically promising for the detection of alloimmune
antibodies arising from the transfusion of non-
autologous platelets (Post-transfusion Purpura) or
the entrance of fetal cells into the maternal circula-
tion (Neonatal Alloimmune Thrombocytopenia Pur-
pura). The antibodies are typically formed against
alleles of the human platelet alloantigen system
(HPA), notably HPA-1a (Pl-A1) and HPA1b
(Pl-A2).(207-209) These antibodies can be specifically
detected by flow cytometry, and flow cytometric
crossmatching can be performed to reveal HLA in-
compatibilities between donor platelets and sensi-
tized recipients.(210-212)
Reticulated platelets are recently released into the
circulation, larger than average, and contain small
amounts of RNA remaining from the megakarocytic
process.(213) The quantitation of reticulated platelets
is valuable for the differentiation of accelerated de-
struction from impaired production in patients with
thrombocytopenia of unknown etiology(214-216)
Reticulated platelets can be detected by the utiliza-
tion of thiazole orange, a brightly fluorescent dye that
binds to nucleic acid.(217; 218) The assay is some-
what difficult to perform, since the platelets must be
permeabilized and positive controls are difficult to
obtain.
Hypercoagulability is one of the most common medi-
cal problems, and it is not surprising that a wealth of
new discoveries have arisen from the application of
modern technology. Flow cytometry has been an es-
sential technique for the elaboration of platelet func-
tion and understanding the contribution of platelet
activation to hypercoagulability. To date, many flow
cytometric studies have involved the detection of
platelet surface markers, the study of cytosolic cal-
ciumion levels, and the detection of circulating plate-
let microparticles.(205)
inappropriate treatment (i.e., DDAVP is contraindi-
cated in vWD Type IIb).
Genetic and Molecular Assays
The advances of genetic and molecular methods of
study during the past two decades have had a pro-
found impact upon our understanding of hemostasis,
as well as other fields of medicine. The molecular
origin and function of many substances involved in
hemostasis are now understood, and the genetics and
molecular of the hemophilias and many diseases is
much clearer.(222-225) A diversity of new diagnostic
assays resulting from these discoveries is expected in
the near future, but at present the major clinical role
for molecular analysis is in the diagnosis of inherited
thrombophilia.
The majority of patients who develop recurrent ve-
nous thromboemboli (inherited thrombophilia) have
discernable abnormalities of the coagulation system,
including factor V Leiden, deficiencies of protein C,
protein S, antithrombin III, the prothrombin G20210A
gene mutation, homocysteinemia, elevated factor lev-
els, dysfibrinogenemia, or abnormalities of the fibri-
nolytic system.(226) Most of these abnormalities
cause deficiencies of the regulatory substances of
clotting. Genetic abnormalities are especially com-
mon in individuals who develop thrombi at an early
age (< 40 years) and in those with a family history of
thrombosis. Although no genetic abnormality is de-
tectable in about 15 percent to 20 percent of indi-
viduals with recurrent thromboembolic disease, re-
search in this area is rapidly proceeding and new ge-
netic abnormalities may be described in the near fu-
ture.
Factor V Leiden, the major cause of APCR, first iden-
tified in February 1993, is the most common inher-
ited cause of thrombosis known at this time.(167) It is
found in about 5 percent of the general population
20
Electrophoresis, Genetic & Molecular Assays
Laboratory Evaluation of Hemostasis
the tR2 haplotype and factor V Cambridge
(Arg306Thr).
A mutation in the prothrombin gene that produces
elevated levels of prothrombin was discovered in
1996.(227-231) The mutation involves a single amino
acid substitution (20210G>A) in the 3-UTR untrans-
lated region of the prothrombin gene on chromo-
some 11, leading to more effective mRNA translation
and elevated plasma prothrombin. There is increas-
ing evidence that the G20210A mutation is an impor-
tant risk factor for deep venous thrombosis, myocar-
dial infarction and stroke. The use of estrogen or oral
contraceptives increases the risk of thrombosis even
further in patients with the prothrombin 20210 muta-
tion.
and is responsible for 20 percent to 50 percent cases
of inherited thrombosis. Approximately 50,000 indi-
viduals die yearly in the United States from complica-
tions caused by this abnormality.(154; 168; 170) Het-
erozygous individuals are at five to 10 times greater
risk of thrombosis than the general population, while
homozygotes are at 50-100 times greater risk. The
use of estrogen or oral contraceptives increases the
risk of thrombosis even further. In 90 percent to 95
percent of cases, APCR is a result of a single point
mutation (Arg506Gln) in the gene for factor V on
chromosome 1q23, inherited as an autosomal domi-
nant trait. This mutation renders activated factor V
(Va) more resistant to inactivation by APC. The re-
maining five to 10 percent of APCR is due to other
genetic abnormalities in the factor V gene, including
Hyperhomocysteinemia and homocysteinemia are
inherited abnormalities of homocysteine metabo-
lism. Homocysteine is a naturally occurring sub-
stance involved in the metabolism of certain amino
acids, including cysteine and methionine. Abnormali-
ties in at least three enzymes, methylenetetrahydrofo-
late reductase (MTHFR), cystathionine beta-synthase
(CBS) and methionine synthase (MS) associated with
homocysteine metabolism in the body can lead to
increased homocysteine levels in the body (hyper-
homocysteinemia). Genetic abnormalities in these
enzymes, particularly homozygous defects in
MTHFR, are the common risk factors for thrombotic
disease, including heart disease and stroke.(232; 233)
Hyperhomocysteinemia also may be associated with
vitamin deficiency, advanced age, hypothyroidism,
impaired kidney function, systemic lupus erythema-
tosus and the use of certain medications, including
nicotinic acid, theophylline, methotrexate and L-dopa.
Inherited abnormalities in antithrombin, protein C,
protein S, other regulatory of the coagulation system
are less common and more complex genetically. In-
herited abnormalities in antithrombin, protein C,
protein S occur in two forms, leading to either low
plasma concentrations (Type I deficiency) or fun-
tionally abnormal but quantitatively normal (Type II
deficiency) of the involved proteins. To date, more
than 250 mutations have been described in the anti-
thrombin gene, together with more than 100 each in
the protein C and protein S genes. The likelihood of
clinically significant thrombotic disease or crises in
any one patient is greatly elevated when more than
one of these traits is present.
The molecular evaluation of the hypercoagulable
states was the subject of a recent review by Nagy,
Schrijver, and Zehnder (2004).(234) A variety of mo-
lecular techniques have been employed, but the field
is in a state of rapid development. The molecular
techniques presently employed in the evaluation of
21
Genetic & Molecular Assays
Assay Accuracy Throughput Present Clinical Applications
PCR/RFLP Good Limited Factor V Leiden (1691G>A),
prothrombin 20210AG>A, MTHFR
677C>T
PCR/ARMS Excellent Intermediate
Light Cycler Excellent Intermediate Factor V Leiden (1691G>A),
prothrombin 20210AG>A, MTHFR
677C>T
Array technology Excellent Very high Under development
Invader assays Excellent Limited Under development
Ligand-based
technologies
Excellent Very high Under development
Table II
Molecular Techniques for the Evaluation of Hypercoaguable States*
*Modified from Nagy PL, Schrijver I, Zehnder JL. Molecular diagnosis of hypercoagulable states. Lab Med.
2004;35:214-221.
Laboratory Evaluation of Hemostasis
platelets labelled with
14
C-serotonin are incubated
with patient's serum in the presence and absence of
therapeutic and high concentrations of heparin. If >
20% of the
14
C is released at a heparin concentration
of 0.1 U/ml heparin and < 20% is released by 100 U/
mL heparin, the test is positive for heparin antibod-
ies. This was the original assay used by Sheridan et
al. to establish laboratory diagnosis of the HIT syn-
drome after observing spontaneous aggregation of
platelets incubated in HIT patient plasma with hea-
prin, and has remained the gold standard for valida-
tion of other diagnostic techniques.(240) Now, how-
ever, even more sensitive assays based on flow cy-
tometry detection of circulating platelet microparti-
cles are under development.(241)
References
1. Bates SM, Weitz JI. 2005. Coagulation assays. Cir-
culation 112(4):e53-60.
2. Bain BJ, Arnold JA, Jowzi Z. 2004. Spurious auto-
mated platelet count. Am J Clin Pathol 122(2):316;
author reply 316.
3. Kakkar N. 2004. Spurious rise in the automated
platelet count because of bacteria. J Clin Pathol
57(10):1096-1097.
4. Kakkar N, Garg G. 2004. Spuriously elevated auto-
mated platelet count in severe burns--a report of
two cases. Indian J Pathol Microbiol 47(3):408-410.
5. Segal HC, Briggs C, Kunka S, Casbard A, Harrison
P, Machin SJ, Murphy MF. 2005. Accuracy of plate-
let counting haematology analysers in severe
thrombocytopenia and potential impact on plate-
let transfusion. Br J Haematol 128(4):520-525.
6. Gulati GL, Hyun BH. 1994. Blood smear examina-
tion. Hematol Oncol Clin North Am 8(4):631-650.
7. Moreno A, Menke D. 2002. Assessment of platelet
numbers and morphology in the peripheral blood
smear. Clin Lab Med 22(1):193-213, vii.
8. Sutor AH, Grohmann A, Kaufmehl K, Wundisch T.
2001. Problems with platelet counting in throm-
bocytopenia. A rapid manual method to measure
the inherited thrombophilia disorders are summa-
rized in Table II.
Electron Microscopy
Ultrastructural examination of the platelet (platelet
electron microscopy) is performed in research stud-
ies of platelets, and to confirm the diagnosis of sus-
pected storage pool disease or other diseases result-
ing from structural anomalies. Dr. James G. White of
the Department of Laboratory Medicine and Pathol-
ogy, University of Minnesota has reported the major-
ity of studies of platelet ultrastructure.(235-238)
Platelet ultrastructural examination is classically per-
formed from thin sections taken from PRP fixed in
glutaraldehyde/osmium solutions and dehydrated,
plastic-embedded, sectioned, and stained by conven-
tional techniques. However, Dr. White found that
dense bodies are better visualized by a simple whole-
mount procedure that involves the examination of
unfixed, unstained PRP fixed and dried on Formvar-
coated, carbon-stabilized grids.(239) The procedure is
rapid and easy to perform, and the presence of dense
bodies excludes the diagnosis of the Herman-
skyPudlak syndrome and storage pool disease.
Other techniques, such as scanning electron micros-
copy, the freeze-fracture technique, histochemical
staining for platelet peroxidase, and staining with
labelled gold particles have been extensively utilized
in research studies of the platelet.
Radioimmunoassay
Most clinical laboratories would wish to avoid the
regulatory burden and safety hazards of use of radio-
nuclides whenever possible. However, at this time,
the most sensitive and well-accepted laboratory assay
for heparin-induced thrombocytopenia is the
14
C-
serotonin release assay. In this assay, normal donor
low platelet counts. Semin Thromb Hemost
27(3):237-243.
9. Rodgers RP, Levin J. 1990. Bleeding time: a guide
to its diagnostic and clinical utility. Arch Pathol
Lab Med 114(12):1187-1188.
10. Bowie EJ, Owen CA, Jr. 1974. The bleeding time.
Prog Hemost Thromb 2(0):249-271.
11. Bell A. 1958. The Ivy bleeding time. Am J Med
Technol 24(4):264-270.
12. Rodgers RP, Levin J. 1990. A critical reappraisal of
the bleeding time. Semin Thromb Hemost
16(1):1-20.
13. Lehman CM, Blaylock RC, Alexander DP, Rodgers
GM. 2001. Discontinuation of the bleeding time
test without detectable adverse clinical impact.
Clin Chem 47(7):1204-1211.
14. Gewirtz AS, Miller ML, Keys TF. 1996. The clinical
usefulness of the preoperative bleeding time. Arch
Pathol Lab Med 120(4):353-356.
15. Peterson P, Hayes TE, Arkin CF, Bovill EG, Fair-
weather RB, Rock WA, Jr., Triplett DA, Brandt JT.
1998. The preoperative bleeding time test lacks
clinical benefit: College of American Pathologists'
and American Society of Clinical Pathologists'
position article. Arch Surg 133(2):134-139.
16. Quiroga T, Goycoolea M, Munoz B, Morales M,
Aranda E, Panes O, Pereira J, Mezzano D. 2004.
Template bleeding time and PFA-100 have low
sensitivity to screen patients with hereditary mu-
cocutaneous hemorrhages: comparative study in
148 patients. J Thromb Haemost 2(6):892-898.
17. Jarvis GE. 2004. Platelet aggregation: turbidimetric
measurements. Methods Mol Biol 272:65-76.
18. Hardeman MR, Vreeken J. 1990. The clinical sig-
nificance of in vitro platelet aggregometry.
Thromb Res 59(4):807-808.
19. Hutton RA, Ludlam CA. 1989. ACP Broadsheet 122:
August 1989. Platelet function testing. J Clin Pa-
thol 42(8):858-864.
20. Gibson J, Uhr E, Motum P, Rickard KA, Kronen-
berg H. 1987. Platelet aggregometry for the diag-
nosis of heparin induced thrombocytopenia--
thrombosis syndrome (HITTS). Pathology
19(1):105-106.
22
Electron Microscopy, RIA, References
Laboratory Evaluation of Hemostasis
cardiac surgical patients. J Cardiothorac Vasc An-
esth 18(2):136-140.
31. Ortel TL, James AH, Thames EH, Moore KD,
Greenberg CS. 2000. Assessment of primary hemo-
stasis by PFA-100 analysis in a tertiary care center.
Thromb Haemost 84(1):93-97.
32. Mammen EF, Comp PC, Gosselin R, Greenberg C,
Hoots WK, Kessler CM, Larkin EC, Liles D, Nugent
DJ. 1998. PFA-100 system: a new method for as-
sessment of platelet dysfunction. Semin Thromb
Hemost 24(2):195-202.
33. Kundu SK, Heilmann EJ, Sio R, Garcia C, Davidson
RM, Ostgaard RA. 1995. Description of an in vitro
platelet function analyzer--PFA-100. Semin
Thromb Hemost 21 Suppl 2:106-112.
34. Jilma B. 2001. Platelet function analyzer (PFA-100):
a tool to quantify congenital or acquired platelet
dysfunction. J Lab Clin Med 138(3):152-163.
35. Favaloro EJ. 2002. Clinical application of the PFA-
100. Curr Opin Hematol 9(5):407-415.
36. Carcao MD, Blanchette VS, Dean JA, He L, Kern
MA, Stain AM, Sparling CR, Stephens D, Ryan G,
Freedman J, Rand ML. 1998. The Platelet Function
Analyzer (PFA-100): a novel in-vitro system for
evaluation of primary haemostasis in children. Br
J Haematol 101(1):70-73.
37. Franchini M. 2005. The platelet function analyzer
(PFA-100): an update on its clinical use. Clin Lab
51(7-8):367-372.
38. Harrison P. 2005. The role of PFA-100 testing in
the investigation and management of haemostatic
defects in children and adults. Br J Haematol
130(1):3-10.
39. Sambola A, Heras M, Escolar G, Lozano M, Pino M,
Martorell T, Torra M, Sanz G. 2004. The PFA-100
detects sub-optimal antiplatelet responses in pa-
tients on aspirin. Platelets 15(7):439-446.
40. Coakley M, Self R, Marchant W, Mackie I, Mallett
SV, Mythen M. 2005. Use of the platelet function
analyser (PFA-100) to quantify the effect of low
dose aspirin in patients with ischaemic heart dis-
ease. Anaesthesia 60(12):1173-1178.
41. Feuring M, Schultz A, Losel R, Wehling M. 2005.
Monitoring acetylsalicylic acid effects with the
21. Brace LD. 1992. Testing for heparin-induced
thrombocytopenia by platelet aggregometry. Clin
Lab Sci 5(2):80-81.
22. Ewenstein BM. 2001. Use of ristocetin cofactor
activity in the management of von Willebrand
disease. Haemophilia 7 Suppl 1:10-15.
23. Miller CH, Platt SJ, Daniele C, Kaczor D. 2002.
Evaluation of two automated methods for meas-
urement of the ristocetin cofactor activity of von
Willebrand factor. Thromb Haemost 88(1):56-59.
24. Ermens AA, de Wild PJ, Vader HL, van der Graaf F.
1995. Four agglutination assays evaluated for
measurement of von Willebrand factor (ristocetin
cofactor activity). Clin Chem 41(4):510-514.
25. Vanhoorelbeke K, Cauwenberghs N, Vauterin S,
Schlammadinger A, Mazurier C, Deckmyn H. 2000.
A reliable and reproducible ELISA method to
measure ristocetin cofactor activity of von Wille-
brand factor. Thromb Haemost 83(1):107-113.
26. Vanhoorelbeke K, Cauwenberghs N, Vandecasteele
G, Vauterin S, Deckmyn H. 2002. A Reliable von
Willebrand factor: ristocetin cofactor enzyme-
linked immunosorbent assay to differentiate be-
tween type 1 and type 2 von Willebrand disease.
Semin Thromb Hemost 28(2):161-166.
27. Riddell AF, Jenkins PV, Nitu-Whalley IC, McCraw
AH, Lee CA, Brown SA. 2002. Use of the collagen-
binding assay for von Willebrand factor in the
analysis of type 2M von Willebrand disease: a
comparison with the ristocetin cofactor assay. Br J
Haematol 116(1):187-192.
28. Murdock PJ, Woodhams BJ, Matthews KB, Pasi KJ,
Goodall AH. 1997. von Willebrand factor activity
detected in a monoclonal antibody-based ELISA:
an alternative to the ristocetin cofactor platelet
agglutination assay for diagnostic use. Thromb
Haemost 78(4):1272-1277.
29. Nicholson NS, Panzer-Knodle SG, Haas NF, Taite
BB, Szalony JA, Page JD, Feigen LP, Lansky DM,
Salyers AK. 1998. Assessment of platelet function
assays. Am Heart J 135(5 Pt 2 Su):S170-178.
30. Lennon MJ, Gibbs NM, Weightman WM, McGuire
D, Michalopoulos N. 2004. A comparison of Plate-
letworks and platelet aggregometry for the as-
sessment of aspirin-related platelet dysfunction in
platelet function analyzer PFA-100. Semin Thromb
Hemost 31(4):411-415.
42. Dyszkiewicz-Korpanty A, Quinton R, Yassine J,
Sarode R. 2004. The effect of a pneumatic tube
transport system on PFA-100 trade mark closure
time and whole blood platelet aggregation. J
Thromb Haemost 2(2):354-356.
43. Fricke W, Kouides P, Kessler C, Schmaier AH, Kri-
janovski Y, Jagadeesan K, Joist J. 2004. A multicen-
ter clinical evaluation of the Clot Signature Ana-
lyzer. J Thromb Haemost 2(5):763-768.
44. Igawa T, Kornhauser R, Cilla DD, King JO, Kam-
bayashi J. 1999. Evaluation of the Clot Signature
Analyzer as a hemostasis test in healthy volun-
teers exposed to low doses of aspirin. Clin Appl
Thromb Hemost 5(2):117-121.
45. Li CK, Hoffmann TJ, Hsieh PY, Malik S, Watson
WC. 1998. The Xylum Clot Signature Analyzer: a
dynamic flow system that simulates vascular in-
jury. Thromb Res 92(6 Suppl 2):S67-77.
46. Faraday N, Guallar E, Sera VA, Bolton ED, Scharpf
RB, Cartarius AM, Emery K, Concord J, Kickler TS.
2002. Utility of whole blood hemostatometry us-
ing the clot signature analyzer for assessment of
hemostasis in cardiac surgery. Anesthesiology
96(5):1115-1122.
47. Haddadin AS, Ayoub CM, Sevarino FB, Rinder CS.
1999. Evaluation of hemostasis by the Clot Signa-
ture Analyzer: a potentially valuable device for the
anesthesiologist. J Clin Monit Comput
15(2):125-129.
48. Brubaker DB. 1989. An in vitro bleeding time test.
Am J Clin Pathol 91(4):422-429.
49. Coiffic A, Cazes E, Janvier G, Forestier F, Lanza F,
Nurden A, Nurden P. 1999. Inhibition of platelet
aggregation by abciximab but not by aspirin can
be detected by a new point-of-care test, the hemo-
status. Thromb Res 95(2):83-91.
50. Isgro F, Rehn E, Kiessling AH, Kretz KU, Kilian W,
Saggau W. 2002. Platelet function test HemoSTA-
TUS 2: tool or toy for an optimized management
of hemostasis? Perfusion 17(1):27-31.
51. Ereth MH, Nuttall GA, Klindworth JT, MacVeigh I,
Santrach PJ, Orszulak TA, Harmsen WS, Oliver WC,
Jr. 1997. Does the platelet-activated clotting test
23
References
Laboratory Evaluation of Hemostasis
routine cardiac surgery. Anesth Analg 96(1):51-57,
table of contents.
60. Hendriks HG, Meijer K, de Wolf JT, Porte RJ,
Klompmaker IJ, Lip H, Slooff MJ, van der Meer J.
2002. Effects of recombinant activated factor VII
on coagulation measured by thromboelastogra-
phy in liver transplantation. Blood Coagul Fibri-
nolysis 13(4):309-313.
61. Huang GS, Chang JH, Lee MS, Wu CC, Lin SP, Lin
SL, Wong CS. 2002. The effect of anesthetic tech-
niques on hemostatic function in arthroscopic
surgery: evaluation by thromboelastography. Acta
Anaesthesiol Sin 40(3):121-126.
62. Samama CM. 2001. Thromboelastography: the
next step. Anesth Analg 92(3):563-564.
63. Ti LK, Cheong KF, Chen FG. 2002. Prediction of
excessive bleeding after coronary artery bypass
graft surgery: the influence of timing and hepari-
nase on thromboelastography. J Cardiothorac Vasc
Anesth 16(5):545-550.
64. Luddington RJ. 2005. Thrombelastography/
thromboelastometry. Clin Lab Haematol
27(2):81-90.
65. Zambruni A, Thalheimer U, Leandro G, Perry D,
Burroughs AK. 2004. Thromboelastography with
citrated blood: comparability with native blood,
stability of citrate storage and effect of repeated
sampl i ng. Bl ood Coagul Fi br i nol ysi s
15(1):103-107.
66. Carr ME, Jr. 1995. Measurement of platelet force:
the Hemodyne hemostasis analyzer. Clin Lab
Manage Rev 9(4):312-314, 316-318, 320.
67. Carr ME, Jr. 1997. In vitro assessment of platelet
function. Transfus Med Rev 11(2):106-115.
68. Harker LA, Malpass TW, Branson HE, Hessel EA,
2nd, Slichter SJ. 1980. Mechanism of abnormal
bleeding in patients undergoing cardiopulmonary
bypass: acquired transient platelet dysfunction
associated with selective alpha-granule release.
Blood 56(5):824-834.
69. Carr ME, Jr., Carr SL, Tildon T, Fisher LM, Martin
EJ. 2003. Batroxobin-induced clots exhibit delayed
and reduced platelet contractile force in some
patients with clotting factor deficiencies. J
Thromb Haemost 1(2):243-249.
(HemoSTATUS) predict blood loss and platelet
dysfunction associated with cardiopulmonary
bypass? Anesth Analg 85(2):259-264.
52. Ereth MH, Nuttall GA, Santrach PJ, Klindworth JT,
Oliver WC, Jr., Schaff HV. 1998. The relation be-
tween the platelet-activated clotting test (Hemo-
STATUS) and blood loss after cardiopulmonary
bypass. Anesthesiology 88(4):962-969.
53. Chen WH, Lee PY, Ng W, Kwok JY, Cheng X, Lee
SW, Tse HF, Lau CP. 2005. Relation of aspirin resis-
tance to coronary flow reserve in patients under-
going elective percutaneous coronary interven-
tion. Am J Cardiol 96(6):760-763.
54. Lee PY, Chen WH, Ng W, Cheng X, Kwok JY, Tse HF,
Lau CP. 2005. Low-dose aspirin increases aspirin
resistance in patients with coronary artery dis-
ease. Am J Med 118(7):723-727.
55. Wheeler GL, Braden GA, Steinhubl SR, Kereiakes
DJ, Kottke-Marchant K, Michelson AD, Furman MI,
Mueller MN, Moliterno DJ, Sane DC. 2002. The
Ultegra rapid platelet-function assay: comparison
to standard platelet function assays in patients
undergoing percutaneous coronary intervention
with abciximab therapy. Am Heart J
143(4):602-611.
56. Wang JC, Aucoin-Barry D, Manuelian D, Monbou-
quette R, Reisman M, Gray W, Block PC, Block EH,
Ladenheim M, Simon DI. 2003. Incidence of aspi-
rin nonresponsiveness using the Ultegra Rapid
Platelet Function Assay-ASA. Am J Cardiol
92(12):1492-1494.
57. White MM, Krishnan R, Kueter TJ, Jacoski MV,
Jennings LK. 2004. The use of the point of care
Helena ICHOR/Plateletworks and the Accumetrics
Ultegra RPFA for assessment of platelet function
with GPIIB-IIIa antagonists. J Thromb Throm-
bolysis 18(3):163-169.
58. Bowbrick VA, Mikhailidis DP, Stansby G. 2003.
Value of thromboelastography in the assessment
of platelet function. Clin Appl Thromb Hemost
9(2):137-142.
59. Cammerer U, Dietrich W, Rampf T, Braun SL, Rich-
ter JA. 2003. The predictive value of modified
computerized thromboelastography and platelet
function analysis for postoperative blood loss in
70. Greilich PE, Carr ME, Zekert SL, Dent RM. 1994.
Quantitative assessment of platelet function and
clot structure in patients with severe coronary
artery disease. Am J Med Sci 307(1):15-20.
71. Krishnaswami A, Carr ME, Jr., Jesse RL, Kontos
MC, Minisi AJ, Ornato JP, Vetrovec GW, Martin EJ.
2002. Patients with coronary artery disease who
present with chest pain have significantly elevated
platelet contractile force and clot elastic modulus.
Thromb Haemost 88(5):739-744.
72. Carr ME, Jr., Hackney MH, Hines SJ, Heddinger SP,
Carr SL, Martin EJ. 2002. Enhanced platelet force
development despite drug-induced inhibition of
platelet aggregation in patients with thrombo-
angiitis obliterans--two case reports. Vasc Endo-
vascular Surg 36(6):473-480.
73. Carr ME, Jr., Carr SL, Greilich PE. 1996. Heparin
ablates force development during platelet medi-
ated clot retraction. Thromb Haemost
75(4):674-678.
74. Carr ME, Jr., Carr SL. 1994. At high heparin con-
centrations, protamine concentrations which re-
verse heparin anticoagulant effects are insuffi-
cient to reverse heparin anti-platelet effects.
Thromb Res 75(6):617-630.
75. Carr ME, Monge-Meberg P, McCardell K, Carr SL.
1996. Dermatan sulfate suppresses platelet force as
it prolongs the APTT. Blood 88(Suppl 1):79b.
76. Carr ME, Zekert S. 1993. Effect of non-heparin
thrombin antagonists on platelet force develop-
ment during clot retraction. Thromb Haemost
69:1241.
77. Reverter JC, Beguin S, Kessels H, Kumar R, Hem-
ker HC, Coller BS. 1996. Inhibition of platelet-
mediated, tissue factor-induced thrombin genera-
tion by the mouse/human chimeric 7E3 antibody.
Potential implications for the effect of c7E3 Fab
treatment on acute thrombosis and "clinical res-
tenosis". J Clin Invest 98(3):863-874.
78. Carr ME, Jr., Martin EJ, Kuhn JG, Ambrose H, Fern
S, Bryant PC. 2004. Monitoring of hemostatic
status in four patients being treated with recom-
binant factor VIIa. Clin Lab 50(9-10):529-538.
79. Liszka-Hackzell JJ, Ekback G. 2002. Analysis of the
information content in Sonoclot data and recon-
24
References
Laboratory Evaluation of Hemostasis
89. Cavusoglu E, Lakhani M, Marmur JD. 2005. The
activated clotting time (ACT) can be used to
monitor enoxaparin and dalteparin after intrave-
nous administration. J Invasive Cardiol
17(8):416-421.
90. Marmur JD, Anand SX, Bagga RS, Fareed J, Pan
CM, Sharma SK, Richard MF. 2003. The activated
clotting time can be used to monitor the low mo-
lecular weight heparin dalteparin after intrave-
nous administration. J Am Coll Cardiol
41(3):394-402.
91. Schussler JM, Lander SR, Wissinger LA, Anwar A,
Donsky MS, Johnson KB, Vallabhan RC,
Wischmeyer JB. 2004. Validation of the i-STAT
handheld activated clotting time for use with
bivalirudin. Am J Cardiol 93(10):1318-1319.
92. Jackson CM, Esnouf MP, Lindahl TL. 2003. A criti-
cal evaluation of the prothrombin time for moni-
toring oral anticoagulant therapy. Pathophysiol
Haemost Thromb 33(1):43-51.
93. Kitchen S, Preston FE. 1999. Standardization of
prothrombin time for laboratory control of oral
anticoagulant therapy. Semin Thromb Hemost
25(1):17-25.
94. Riley RS, Rowe D, Fisher LM. 2000. Clinical utiliza-
tion of the international normalized ratio (INR). J
Clin Lab Anal 14(3):101-114.
95. Horsti J, Uppa H, Vilpo JA. 2005. Poor agreement
among prothrombin time international normal-
ized ratio methods: comparison of seven com-
mercial reagents. Clin Chem 51(3):553-560.
96. Jackson CM, Esnouf MP. 2005. Has the time ar-
rived to replace the quick prothrombin time test
for monitoring oral anticoagulant therapy? Clin
Chem 51(3):483-485.
97. Choi TS, Greilich PE, Shi C, Wilson JS, Keller A,
Kroll MH. 2002. Point-of-care testing for
prothrombin time, but not activated partial
thromboplastin time, correlates with laboratory
methods in patients receiving aprotinin or
epsilon-aminocaproic acid while undergoing car-
diac surgery. Am J Clin Pathol 117(1):74-78.
98. Searles B, Nasrallah F, Graham S, Lajara RB. 2002.
Comparison of five point-of-care prothrombin
and activated partial thromboplastin time devices
struction of coagulation test variables. J Med Syst
26(1):1-8.
80. Furuhashi M, Ura N, Hasegawa K, Yoshida H,
Tsuchihashi K, Miura T, Shimamoto K. 2002. Sono-
clot coagulation analysis: new bedside monitoring
for determination of the appropriate heparin dose
during haemodialysis. Nephrol Dial Transplant
17(8):1457-1462.
81. Shibata T, Sasaki Y, Hattori K, Hirai H, Hosono M,
Fujii H, Suehiro S. 2004. Sonoclot analysis in car-
diac surgery in dialysis-dependent patients. Ann
Thorac Surg 77(1):220-225.
82. Miyashita T, Kuro M. 1998. Evaluation of platelet
function by Sonoclot analysis compared with
other hemostatic variables in cardiac surgery.
Anesth Analg 87(6):1228-1233.
83. LaForce WR, Brudno DS, Kanto WP, Karp WB.
1992. Evaluation of the SonoClot Analyzer for the
measurement of platelet function in whole blood.
Ann Clin Lab Sci 22(1):30-33.
84. Francis JL, Francis DA, Gunathilagan GJ. 1994.
Assessment of hypercoagulability in patients with
cancer using the Sonoclot Analyzer and throm-
boelastography. Thromb Res 74(4):335-346.
85. Simko RJ, Tsung FF, Stanek EJ. 1995. Activated
clotting time versus activated partial thrombo-
plastin time for therapeutic monitoring of hepa-
rin. Ann Pharmacother 29(10):1015-1021; quiz
1061.
86. Pesola DA, Pesola HR, Pesola GR. 1995. Advantages
for the use of the activated clotting time (ACT).
Am J Crit Care 4(5):414-415.
87. De Waele JJ, Van Cauwenberghe S, Hoste E, Benoit
D, Colardyn F. 2003. The use of the activated clot-
ting time for monitoring heparin therapy in criti-
cally ill patients. Intensive Care Med 29(2):325-328.
88. Despotis GJ, Joist JH, Hogue CW, Jr., Alsoufiev A,
Kater K, Goodnough LT, Santoro SA, Spitznagel E,
Rosenblum M, Lappas DG. 1995. The impact of
heparin concentration and activated clotting time
monitoring on blood conservation. A prospective,
randomized evaluation in patients undergoing
cardiac operation. J Thorac Cardiovasc Surg
110(1):46-54.
based on age of blood sample. J Extra Corpor
Technol 34(3):178-181.
99. Shiach CR, Campbell B, Poller L, Keown M, Chau-
han N. 2002. Reliability of point-of-care
prothrombin time testing in a community clinic: a
randomized crossover comparison with hospital
laboratory testing. Br J Haematol 119(2):370-375.
100. Tripodi A, Bressi C, Carpenedo M, Chantarangkul
V, Clerici M, Mannucci PM. 2004. Quality assur-
ance program for whole blood prothrombin time-
international normalized ratio point-of-care
monitors used for patient self-testing to control
oral anticoagulation. Thromb Res 113(1):35-40.
101. Tseng LW, Hughes D, Giger U. 2001. Evaluation of a
point-of-care coagulation analyzer for measure-
ment of prothrombin time, activated partial
thromboplastin time, and activated clotting time
in dogs. Am J Vet Res 62(9):1455-1460.
102. Yuoh C, Tarek Elghetany M, Petersen JR, Moham-
mad A, Okorodudu AO. 2001. Accuracy and preci-
sion of point-of-care testing for glucose and
prothrombin time at the critical care units. Clin
Chim Acta 307(1-2):119-123.
103. Poller L, Keown M, Chauhan N, van Den Besselaar
AM, Tripodi A, Jespersen J, Shiach C, Horellou MH,
Dias D, Egberg N, Iriarte JA, Kontopoulou-Griva I,
Otridge B. 2002. European Concerted Action on
Anticoagulation (ECAA): multicentre interna-
tional sensitivity index calibration of two types of
point-of-care prothrombin time monitor systems.
Br J Haematol 116(4):844-850.
104. Brandt JT, Arkin CF, Bovill EG, Rock WA, Triplett
DA. 1990. Evaluation of APTT reagent sensitivity
to factor IX and factor IX assay performance. Re-
sults from the College of American Pathologists
Survey Program. Arch Pathol Lab Med
114(2):135-141.
105. Kitchen S, Jennings I, Woods TA, Preston FE. 1996.
Wide variability in the sensitivity of APTT rea-
gents for monitoring of heparin dosage. J Clin
Pathol 49(1):10-14.
106. Manzato F, Mengoni A, Grilenzoni A, Lippi G. 1998.
Evaluation of the activated partial thromboplastin
time (APTT) sensitivity to heparin using five
25
References
Laboratory Evaluation of Hemostasis
multiple faces of the partial thromboplastin time
APTT. J Thromb Haemost 2(12):2250-2259.
119. Acosta M, Edwards R, Jaffe EI, Yee DL, Mahoney
DH, Teruya J. 2005. A practical approach to pediat-
ric patients referred with an abnormal coagula-
t ion profil e. Arch Pathol Lab Med
129(8):1011-1016.
120. Toh CH. 2005. A further face of the partial throm-
boplastin time APTT. J Thromb Haemost
3(5):1107-1108.
121. Sahud MA. 2000. Factor VIII inhibitors. Laboratory
diagnosis of inhibitors. Semin Thromb Hemost
26(2):195-203.
122. Austen DE, Lechner K, Rizza CR, Rhymes IL. 1982.
A comparison of the Bethesda and New Oxford
methods of factor VIII antibody assay. Thromb
Haemost 47(1):72-75.
123. Verbruggen B, Novakova I, Wessels H, Boezeman J,
van den Berg M, Mauser-Bunschoten E. 1995. The
Nijmegen modification of the Bethesda assay for
factor VIII:C inhibitors: improved specificity and
reliability. Thromb Haemost 73(2):247-251.
124. Klinge J, Auerswald G, Budde U, Klose H, Kreuz W,
Lenk H, Scandella D. 2001. Detection of all anti-
factor VIII antibodies in haemophilia A patients
by the Bethesda assay and a more sensitive im-
munoprecipitation assay. Haemophilia 7(1):26-32.
125. Hauptmann J. 2002. Pharmacokinetics of an
emerging new class of anticoagulant/
antithrombotic drugs. A review of small-molecule
thrombin inhibitors. Eur J Clin Pharmacol
57(11):751-758.
126. Hafner G, Roser M, Nauck M. 2002. Methods for
the monitoring of direct thrombin inhibitors.
Semin Thromb Hemost 28(5):425-430.
127. Potzsch B, Hund S, Madlener K, Unkrig C, Muller-
Berghaus G. 1997. Monitoring of recombinant
hirudin: assessment of a plasma-based ecarin
clotting time assay. Thromb Res 86(5):373-383.
128. Fenyvesi T, Jorg I, Harenberg J. 2002. Monitoring of
anticoagulant effects of direct thrombin inhibi-
tors. Semin Thromb Hemost 28(4):361-368.
129. Nowak G. 2003. The ecarin clotting time, a univer-
sal method to quantify direct thrombin inhibitors.
Pathophysiol Haemost Thromb 33(4):173-183.
commercial reagents: implications for therapeutic
monitoring. Clin Chem Lab Med 36(12):975-980.
107. Wojtkowski TA, Rutledge JC, Matthews DC. 1999.
The clinical impact of increased sensitivity PT
and APTT coagulation assays. Am J Clin Pathol
112(2):225-232.
108. Brigden ML, Johnston M. 2000. A survey of aPTT
reporting in Canadian medical laboratories. The
need for increased standardization. Thrombosis
Interest Group of Canada. Am J Clin Pathol
114(2):276-282.
109. Teruya J, Oropeza M, Ramsey G. 2001. A normal
aPTT does not guarantee adequate coagulation
factor levels. Anesthesiology 94(3):542; author
reply 543.
110. Kitchens CS. 2005. To bleed or not to bleed? Is that
the question for the PTT? J Thromb Haemost
3(12):2607-2611.
111. Bajaj SP, Joist JH. 1999. New insights into how
blood clots: implications for the use of APTT and
PT as coagulation screening tests and in monitor-
ing of anticoagulant therapy. Semin Thromb He-
most 25(4):407-418.
112. White GC, 2nd. 2003. The partial thromboplastin
time: defining an era in coagulation. J Thromb
Haemost 1(11):2267-2270.
113. Olson JD. 1999. Addressing clinical etiologies of a
prolonged aPTT. CAP Today 13(9):28, 30, 32 pas-
sim.
114. Chen CC, You JY, Ho CH. 2003. The aPTT assay as a
monitor of heparin anticoagulation efficacy in
clinical settings. Adv Ther 20(5):231-236.
115. Gibbar-Clements T, Shirrell D, Free G. 1997. PT and
APTT--seeing beyond the numbers. Nursing
27(7):49-51.
116. McConnell EA. 1986. APTT and PT--the tests of
time. Nursing 16(5):47.
117. Chng WJ, Sum C, Kuperan P. 2004. Causes of iso-
lated prolonged activated partial thromboplastin
time (APTT) in an acute general hospital: a guide
to fresh frozen plasma (FFP) usage. Ann Acad Med
Singapore 33(5 Suppl):S72-73.
118. Rapaport SI, Vermylen J, Hoylaerts M, Saito H,
Hirsh J, Bates S, Dahlback B, Poller L. 2004. The
130. Lange U, Nowak G, Bucha E. 2003. Ecarin chro-
mogenic assay--a new method for quantitative
determination of direct thrombin inhibitors like
hirudin. Pathophysiol Haemost Thromb
33(4):184-191.
131. Gosselin RC, King JH, Janatpour KA, Dager WE,
Larkin EC, Owings JT. 2004. Comparing Direct
Thrombin Inhibitors Using aPTT, Ecarin Clotting
Times, and Thrombin Inhibitor Management Test-
ing (September). Ann Pharmacother.
132. Choi TS, Khan AI, Greilich PE, Kroll MH. 2006.
Modified plasma-based ecarin clotting time assay
for monitoring of recombinant hirudin during
cardiac surgery. Am J Clin Pathol 125(2):290-295.
133. Eika C, Godal HC, Kierulf P. 1972. Detection of
small amounts of heparin by the thrombin
clotting-time. Lancet 2(7773):376.
134. Jespersen J, Sidelmann J. 1982. A study of the con-
ditions and accuracy of the thrombin time assay
of plasma fibrinogen. Acta Haematol 67(1):2-7.
135. Arnesen H. 1973. Studies on the thrombin clotting
time. II. The influence of fibrin degradation prod-
ucts. Scand J Haematol 10(4):291-297.
136. Arnesen H, Godal HC. 1973. Studies on the throm-
bin clotting time. I. The influence of fibrinogen
degradation products. Scand J Haematol
10(3):232-240.
137. Alonso KB. 1978. The seial thrombin time in the
diagnosis of consumptive coagulopathy. Ann Clin
Lab Sci 8(3):228-233.
138. Pizzuto J, Garcia-Mendez S, de-la-Paz Reyna M,
Morales MR, Aviles A, Zavala B, Gaos C. 1979.
Thrombin time dilution test: a simple method for
the control of heparin therapy. Thromb Haemost
42(4):1276-1285.
139. Carr ME, Jr., Gabriel DA. 1986. Hyperfibrinogene-
mia as a cause of prolonged thrombin clotting
time. South Med J 79(5):563-570.
140. Flanders MM, Crist R, Rodgers GM. 2003. Com-
parison of five thrombin time reagents. Clin Chem
49(1):169-172.
141. Lowe GD, Rumley A, Mackie IJ. 2004. Plasma fi-
brinogen. Ann Clin Biochem 41(Pt 6):430-440.
142. Marbet GA, Duckert F. 1992. Fibrinogen. In:
Jespersen J, Bertina RM, Haverkate F, editors.
26
References
Laboratory Evaluation of Hemostasis
mutation, and a laboratory testing algorithm.
Arch Pathol Lab Med 126(5):577-582.
155. Bertolaccini ML, Hughes GR, Khamashta MA.
2004. Revisiting antiphospholipid antibodies:
from targeting phospholipids to phospholipid
binding proteins. Clin Lab 50(11-12):653-665.
156. Mwanda OW. 2003. Lupus anticoagulants: patho-
physiology, clinical and laboratory associations: a
review. East Afr Med J 80(11):564-568.
157. de Groot PG, Derksen RH. 2005. Pathophysiology
of the antiphospholipid syndrome. J Thromb
Haemost 3(8):1854-1860.
158. Riley RS, Friedline J, Rogers JS, 2nd. 1997. Anti-
phospholipid antibodies: standardization and
testing. Clin Lab Med 17(3):395-430.
159. Triplett DA. 2000. Use of the dilute Russell viper
venom time (dRVVT): its importance and pitfalls.
J Autoimmun 15(2):173-178.
160. Derksen RH, de Groot PG. 2004. Tests for lupus
ant i coagul ant revi si ted. Thromb Res
114(5-6):521-526.
161. Passam F, Krilis S. 2004. Laboratory tests for the
antiphospholipid syndrome: current concepts.
Pathology 36(2):129-138.
162. Pierangeli SS, Harris EN. 2005. Clinical laboratory
testing for the antiphospholipid syndrome. Clin
Chim Acta 357(1):17-33.
163. Brandt JT, Triplett DA. 1989. The effect of phos-
pholipid on the detection of lupus anticoagulants
by the dilute Russell viper venom time. Arch Pa-
thol Lab Med 113(12):1376-1378.
164. Exner T, Papadopoulos G, Koutts J. 1990. Use of a
simplified dilute Russell's viper venom time
(DRVVT) confirms heterogeneity among 'lupus
anticoagulants'. Blood Coagul Fibrinolysis
1(3):259-266.
165. Dahlback B. 1995. New molecular insights into the
genetics of thrombophilia. Resistance to activated
protein C caused by Arg506 to Gln mutation in
factor V as a pathogenic risk factor for venous
thrombosis. Thromb Haemost 74(1):139-148.
166. Dahlback B. 1995. Thrombophilia: the discovery of
activated protein C resistance. Adv Genet
33:135-175.
ECAT Assay Procedures: a Manual of Laboratory
Techniques. Dordrecht: Kluwer. p 47-56.
143. Von Clauss A. 1957. Gerinnunsphysiologische
Schnellmethode zur Bestimmung des Fibrinogens.
Acta Haematol 17:237-246.
144. Donati MB, Vermylen J, Verstraete M. 1971. Fi-
brinogen degradation products and a fibrinogen
assay based on clotting kinetics. Scand J Haematol
Suppl 13:255-256.
145. Natelson EA, Dooley DF. 1974. Rapid determina-
tion of fibrinogen by thrombokinetics. Am J Clin
Pathol 61(6):828-833.
146. Tan V, Doyle CJ, Budzynski AZ. 1995. Comparison
of the kinetic fibrinogen assay with the von Clauss
method and the clot recovery method in plasma
of patients with conditions affecting fibrinogen
coagulability. Am J Clin Pathol 104:455-462.
147. Thom J, Ivey L, Eikelboom J. 2003. Normal plasma
mixing studies in the laboratory diagnosis of lu-
pus anticoagulant. J Thromb Haemostasis
1:2689-2691.
148. Chang SH, Tillema V, Scherr D. 2002. A "percent
correction" formula for evaluation of mixing
studies. Am J Clin Pathol 117(1):62-73.
149. Chang S. 2004. More on: normal plasma mixing
studies in the laboratory diagnosis of lupus anti-
coagulant. J Thromb Haemost 2(8):1480-1481.
150. Lossing TS, Kasper CK, Feinstein DI. 1977. Detec-
tion of factor VIII inhibitors with the partial
thromboplastin time. Blood 49:793-797.
151. Clyne LP, White PF. 1988. Time dependency of
lupuslike anticoagulants. Arch Intern Med
148:1060-1063.
152. Kaczor DA, Bickford NN, Triplett DA. 1991. Evalua-
tion of different mixing study reagents and dilu-
tion effect in lupus anticoagulant testing. Am J
Clin Pathol 95(3):408-411.
153. Asmis LM, Sulzer I, Furlan M, Lammle B. 2002.
Prekallikrein deficiency: the characteristic nor-
malization of the severely prolonged aPTT follow-
ing increased preincubation time is due to autoac-
tivation of factor XII. Thromb Res 105(6):463-470.
154. Van Cott EM, Soderberg BL, Laposata M. 2002.
Activated protein C resistance, the factor V Leiden
167. Dahlback B. 2003. The discovery of activated pro-
tein C resistance. J Thromb Haemost 1(1):3-9.
168. Nicolaes GA, Dahlback B. 2003. Activated protein C
resistance (FV(Leiden)) and thrombosis: factor V
mutations causing hypercoagulable states. Hema-
tol Oncol Clin North Am 17(1):37-61, vi.
169. Strobl FJ, Hoffman S, Huber S, Williams EC, Voelk-
erding KV. 1998. Activated protein C resistance
assay performance: improvement by sample dilu-
tion with factor V-deficient plasma. Arch Pathol
Lab Med 122(5):430-433.
170. Press RD, Bauer KA, Kujovich JL, Heit JA. 2002.
Clinical utility of factor V leiden (R506Q) testing
for the diagnosis and management of throm-
boembolic disorders. Arch Pathol Lab Med
126(11):1304-1318.
171. Sayinalp N, Haznedaroglu IC, Aksu S, Buyukasik Y,
Goker H, Parlak H, Ozcebe OI, Kirazli S, Dundar
SV, Gurgey A. 2004. The predictability of factor V
Leiden (FV:Q(506)) gene mutation via clotting-
based diagnosis of activated protein C resistance.
Clin Appl Thromb Hemost 10(3):265-270.
172. Aiyappa PA. 1981. Chromogenic substrate spec-
trophotometric assays for the measurement of
clotting function. Ann N Y Acad Sci 370:812-821.
173. Hutton RA. 1987. Chromogenic substrates in hae-
mostasis. Blood Rev 1(3):201-206.
174. Retzios AD, Rosenfeld R, Schiffman S. 1988. En-
zymes of the contact phase of blood coagulation:
kinetics with various chromogenic substrates and
a two-substrate assay for the joint estimation of
plasma prekallikrein and factor XI. J Lab Clin Med
112(5):560-566.
175. Christensen U. 1980. Requirements for valid as-
says of clotting enzymes using chromogenic sub-
strates. Thromb Haemost 43(2):169-174.
176. Madaras F, Chew MY, Parkin JD. 1981. Automated
estimation of factor Xa using the chromogenic
substrate S-2222. Haemostasis 10(5):271-275.
177. Brooks MB. 2004. Evaluation of a chromogenic
assay to measure the factor Xa inhibitory activity
of unfractionated heparin in canine plasma. Vet
Clin Pathol 33(4):208-214.
178. Chandler WL, Ferrell C, Lee J, Tun T, Kha H. 2003.
Comparison of three methods for measuring fac-
27
References
Laboratory Evaluation of Hemostasis
190. Fr V, Hainaut P, Fr T, Elamly A, Dessomme B,
Lavenne E, Reynaert MS. 2003. ELISA D-dimer
measurement for the clinical suspicion of pulmo-
nary embolism in the emergency department:
one-year observational study of the safety profile
and physician's prescription. Acta Clin Belg
58(4):233-240.
191. Michiels JJ, Schroyens W, De Backer W, van der
Planken M, Hoogsteden H, Pattynama PM. 2003.
Non-invasive exclusion and diagnosis of pulmo-
nary embolism by sequential use of the rapid
ELISA D-dimer assay, clinical score and spiral CT.
Int Angiol 22(1):1-14.
192. Ellis DR, Eaton AS, Plank MC, Butman BT, Ebert
RF. 1993. A comparative evaluation of ELISAs for
D-dimer and related fibrin(ogen) degradation
products. Blood Coagul Fibrinolysis 4(4):537-549.
193. Normansell DE. 1982. Quantitation of serum im-
munoglobulins. Crit Rev Clin Lab Sci
17(2):103-170.
194. Carpenter AB. 1997. Enzyme-linked Immunoas-
says. In: Rose NR, de Macario EC, Folds JD, Lane
HC, Nakamura RM, editors. Manual of Clinical
Laboratory Immunology. Washington, D.C.: ASM
Press. p 20-29.
195. Bock JL. 2000. The new era of automated immu-
noassay. Am J Clin Pathol 113(5):628-646.
196. Gosling JP. 1990. A decade of development in im-
munoassay methodology. Clin Chem 36:1408-1427.
197. Johannsson A, Ellis DH, Bates DL, Plumb AM,
Stanley CJ. 1986. Enzyme amplification for immu-
noassays. Detection limit of one hundredth of an
attomole. J Immunol Methods 87:7-11.
198. Ishikawa E, Hashida S, Tanaka K, Kohno T. 1989.
Development and applications of ultrasensitive
enzyme immunoassays for antigens and antibod-
ies. Clin Chim Acta 185(3):223-230.
199. Scheffold A, Kern F. 2000. Recent developments in
flow cytometry. J Clin Immunol 20(6):400-407.
200. Stewart CC. 2000. Multiparameter flow cytometry.
J Immunoassay 21(2-3):255-272.
201. Bakke AC. 2001. The principles of flow cytometry.
Lab Med 32(4):207-211.
202. Givan AL. 2001. Principles of flow cytometry: an
overview. Methods Cell Biol 63:19-50.
tor VIII levels in plasma. Am J Clin Pathol
120(1):34-39.
179. Kasahara Y. 1997. Agglutination immunoassays. In:
Rose NR, de Macario EC, Folds JD, Lane HC,
Nakamura RM, editors. Manual of Clinical Labora-
tory Immunology. Washington, D.C.: ASM Press. p
7-12.
180. Singer JM, Plotz CM. 1956. The latex fixation test. I.
Application to the serological diagnosis of rheu-
matoid arthritis. Amer J Med 21:888-891.
181. van Lente F. 1997. Light-scattering immunoassays.
In: Rose NR, de Macario EC, Folds JD, Lane HC,
Nakamura RM, editors. Manual of Clinical Labora-
tory Immunology. Fifth ed. Washington, D.C.: ASM
Press. p 13-19.
182. Whicher JT, Price CP, Spencer K. 1983. Immu-
nonephelometric and immunoturbidimetric as-
says for proteins. Crit Rev Clin Lab Sci
18(3):213-260.
183. Lugovskoy EV, Gritsenko PG, Kolesnikova IN,
Zolotarova EN, Chernishov VI, Nieuwenhuizen W,
Komisarenko SV. 2004. Two monoclonal antibod-
ies to D-dimer-specific inhibitors of fibrin polym-
erization. Thromb Res 113(3-4):251-259.
184. Bounameaux H. 2003. Review: ELISA D-dimer is
sensitive but not specific in diagnosing pulmo-
nary embolism in an ambulatory clinical setting.
ACP J Club 138(1):24.
185. Heim SW, Schectman JM, Siadaty MS, Philbrick JT.
2004. D-dimer testing for deep venous thrombo-
sis: a metaanalysis. Clin Chem 50(7):1136-1147.
186. Mavromatis BH, Kessler CM. 2001. D-dimer test-
ing: the role of the clinical laboratory in the diag-
nosis of pulmonary embolism. J Clin Pathol
54(9):664-668.
187. Sadosty AT, Goyal DG, Boie ET, Chiu CK. 2001.
Emergency department D-dimer testing. J Emerg
Med 21(4):423-429.
188. Wakai A, Gleeson A, Winter D. 2003. Role of fibrin
D-dimer testing in emergency medicine. Emerg
Med J 20(4):319-325.
189. Adcock D, Joiner-Maier D. 1999. An automated,
rapid ELISA for the determination of D-dimer in
the evaluation of venous thrombosis. Am Clin Lab
18(10):6.
203. McCoy P. 2002. Flow cytometry. In: McClatchey
KD, editor. Clinical Laboratory Medicine. 2nd ed.
Philadelphia: Lippincott, Williams, and Wilkins. p
1401-1425.
204. Chapman GV. 2000. Instrumentation for flow cy-
tometry. J Immunol Methods 243(1-2):3-12.
205. Bode AP, Hickerson DHM. 2002. Flow cytometry of
platelets for clinical analysis. Hematol Oncol Clin
North Amer In Press.
206. George JN. 1990. Platelet immunoglobulin G: its
significance for the evaluation of thrombocy-
topenia and for understanding the origin of
alpha-granule proteins. Blood 76(5):859-870.
207. Deckmyn H, Ulrichts H, Van De Walle G, Van-
hoorelbeke K. 2004. Platelet antigens and their
function. Vox Sang 87 Suppl 2:105-111.
208. Metcalfe P, Watkins NA, Ouwehand WH, Kaplan C,
Newman P, Kekomaki R, De Haas M, Aster R, Shi-
bata Y, Smith J, Kiefel V, Santoso S. 2003. Nomen-
clature of human platelet antigens. Vox Sang
85(3):240-245.
209. Rozman P. 2002. Platelet antigens. The role of hu-
man platelet alloantigens (HPA) in blood transfu-
sion and transplantation. Transpl Immunol
10(2-3):165-181.
210. Marti GE, Magruder L, Schuette WE, Gralnick HR.
1988. Flow cytometric analysis of platelet surface
antigens. Cytometry 9(5):448-455.
211. Neumller J, Tohidast-Akrad M, Jilch R, Schwartz
DW, Mayr WR. 1995. Standardization of the flow
cytometric determination of HLA class I antigens,
'platelet-specific' glycoproteins and activation
markers. Vox Sang 68(2):109-120.
212. Loudova M, Vokurkova D, Hoskova J, Krejsek J.
2002. [Immunofluorescence determination using
flow cytometry of platelet surface antigens in
peripheral blood of healthy individuals (pilot
study)]. Vnitr Lek 48(7):632-637.
213. Ault KA, Rinder HM, Mitchell J, Carmody MB, Vary
CP, Hillman RS. 1992. The significance of platelets
with increased RNA content (reticulated plate-
lets). A measure of the rate of thrombopoiesis. Am
J Clin Pathol 98(6):637-646.
214. Takubo T, Yamane T, Hino M, Ohta K, Koh KR,
Tatsumi N. 2000. Clinical significance of simulta-
28
References
Laboratory Evaluation of Hemostasis
neous measurement of reticulated platelets and
large platelets in idiopathic thrombocytopenic
purpura. Haematologia (Budap) 30(3):183-192.
215. Takubo T, Yamane T, Hino M, Tsuda I, Tatsumi N.
1998. Usefulness of determining reticulated and
large platelets in idiopathic thrombocytopenic
purpura. Acta Haematol 99(2):109-110.
216. Kurata Y, Hayashi S, Kiyoi T, Kosugi S, Kashiwagi
H, Honda S, Tomiyama Y. 2001. Diagnostic value of
tests for reticulated platelets, plasma glycocalicin,
and thrombopoietin levels for discriminating
between hyperdestructive and hypoplastic
thrombocytopenia. Am J Clin Pathol
115(5):656-664.
217. Joutsi-Korhonen L, Sainio S, Riikonen S, Javela K,
Teramo K, Kekomaki R. 2000. Detection of reticu-
lated platelets: estimating the degree of fluores-
cence of platelets stained with thiazole orange.
Eur J Haematol 65(1):66-71.
218. Russell KE, Perkins PC, Grindem CB, Walker KM,
Sellon DC. 1997. Flow cytometric method for de-
tecting thiazole orange-positive (reticulated)
platelets in thrombocytopenic horses. Am J Vet
Res 58(10):1092-1096.
219. Tsao PW, Bozarth JM, Jackson SA, Forsythe MS,
Flint SK, Mousa SA. 1995. Platelet GPIIb/IIIa recep-
tor occupancy studies using a novel fluorescein-
ated cyclic Arg-Gly-Asp peptide. Thromb Res
77(6):543-556.
220. Ryu T. 1987. Multimer analysis of von Willebrand
factor using monoclonal antibody. Nippon Ket-
sueki Gakkai Zasshi 50(8):1689-1696.
221. Tomita Y, Harrison J, Abildgaard CF. 1989. von
Willebrand factor multimer analysis using a sen-
sitive peroxidase staining method. Thromb Hae-
most 62(2):781-783.
222. Spronk HM, Govers-Riemslag JW, ten Cate H. 2003.
The blood coagulation system as a molecular
machine. Bioessays 25(12):1220-1228.
223. Oldenburg J, Schwaab R. 2001. Molecular biology
of blood coagulation. Semin Thromb Hemost
27(4):313-324.
224. Antonarakis SE. 1998. Molecular genetics of co-
agulation factor VIII gene and haemophilia A.
Haemophilia 4 Suppl 2:1-11.
225. Carmeliet P, Collen D. 1997. Molecular genetics of
the fibrinolytic and coagulation systems in hae-
mostasis, thrombogenesis, restenosis and athero-
sclerosis. Curr Opin Lipidol 8(2):118-125.
226. Crowther MA, Kelton JG. 2003. Congenital throm-
bophilic states associated with venous thrombo-
sis: a qualitative overview and proposed classifi-
cation system. Ann Intern Med 138(2):128-134.
227. Leroyer C, Mercier B, Oger E, Chenu E, Abgrall JF,
Ferec C, Mottier D. 1998. Prevalence of 20210 A
allele of the prothrombin gene in venous throm-
boembolism patients. Thromb Haemost
80(1):49-51.
228. Vicente V, Gonzalez-Conejero R, Rivera J, Corral J.
1999. The prothrombin gene variant 20210A in
venous and arterial thromboembolism. Haemato-
logica 84(4):356-362.
229. Tosetto A, Missiaglia E, Frezzato M, Rodeghiero F.
1999. The VITA project: prothrombin G20210A
mutation and venous thromboembolism in the
general population. Thromb Haemost
82(5):1395-1398.
230. Bick RL. 2003. Prothrombin G20210A mutation,
antithrombin, heparin cofactor II, protein C, and
protein S defects. Hematol Oncol Clin North Am
17(1):9-36.
231. McGlennen RC, Key NS. 2002. Clinical and labora-
tory management of the prothrombin G20210A
mutation. Arch Pathol Lab Med 126(11):1319-1325.
232. Eichinger S, Stumpflen A, Hirschl M, Bialonczyk C,
Herkner K, Stain M, Schneider B, Pabinger I,
Lechner K, Kyrle PA. 1998. Hyperhomocysteinemia
is a risk factor of recurrent venous thromboem-
bolism. Thromb Haemost 80(4):566-569.
233. Keijzer MB, den Heijer M, Blom HJ, Bos GM, Wil-
lems HP, Gerrits WB, Rosendaal FR. 2002. Interac-
tion between hyperhomocysteinemia, mutated
methylenetetrahydrofolatereductase (MTHFR)
and inherited thrombophilic factors in recurrent
venous thrombosis. Thromb Haemost
88(5):723-728.
234. Nagy PL, Schrijver I, Zehnder JL. 2004. Molecular
diagnosis of hypercoagulable states. Lab Med
35(4):214-221.
235. White JG. 2004. Electron microscopy methods for
studying platelet structure and function. Methods
Mol Biol 272:47-64.
236. White JG. 1998. Use of the electron microscope for
diagnosis of platelet disorders. Semin Thromb
Hemost 24(2):163-168.
237. White JG. 1981. Morphological studies of platelets
and platelet reactions. Vox Sang 40 Suppl 1:8-17.
238. White JG, Gerrard JM. 1978. The ultrastructure of
defective human platelets. Mol Cell Biochem
21(2):109-128.
239. White JG. 1969. The dense bodies of human plate-
lets: inherent electron opacity of the serotonin
storage particles. Blood 33(4):598-606.
240. Sheridan D, Carter C, Kelton JG. 1986. A diagnostic
test for Heparin-Induced Thrombocytopenia.
Blood 67:27-30.
241. Lee D, Warkentin T, Denomme G, Hayward CP,
Kelton JG. 1996. A diagnostic test for heparin-
induced thrombocytopenia: detection of platelet
microparticles using flow cytometry. Br J Haema-
tol 95:724-731.
29
References
Laboratory Evaluation of Hemostasis

Das könnte Ihnen auch gefallen