J. Gram, J. Jespersen (Auth.), J. Jespersen, R. M. Bertina, F. Haverkate (Eds.) - Laboratory Techniques in Thrombosis - A Manual-Springer Netherlands (1999)
J. Gram, J. Jespersen (Auth.), J. Jespersen, R. M. Bertina, F. Haverkate (Eds.) - Laboratory Techniques in Thrombosis - A Manual-Springer Netherlands (1999)
Thrombosis - a Manual
Laboratory Techniques in
Thrombosis - a Manual
Second revised edition of the ECAT
Assay Procedures
Edited by
J. Jespersen
Department of Clinical Biochemistry,
South Jutland University,
Ribe County Hospital, Esbjerg, Denmark
R. M. Bertina
Haemostasis and Thrombosis Research Centre,
Leiden University Medical Centre,
Leiden, The Netherlands
F. Haverkate
ECAT Foundation, Oegstgeest,
The Netherlands
                                    v
      LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
 8   Fibrinogen                                             79
       M P. M. de Maat, G. D. 0. Lowe and F. Haverkate
                                      vi
                               CONTENTS
24   Tissue-type plasminogen activator (t-PA) activity         223
       C. Kluft, P. Meijer, E. Ersdal and S. Rosen
                                    vii
Preface to the Second Edition
The first edition of this manual appeared in 1992 and was entitled ECAT Assay
Procedures. It was the result of a unique cooperation between experts brought
together by the European Concerted Action on Thrombosis and Disabilities
(ECAT). The Concerted Action was at that time under the auspices of the
Commission of the European Union. The second edition, like the first edition,
deals with diagnostic tests within the field of thrombosis. However, the second
edition has a broader scope because it is no longer limited by the frontiers of
ECAT. Experts allover the world, in and outside ECAT, have contributed to
this edition. The editors are very grateful for their contributions.
   The need for a new edition is obvious. Since 1992 new assays have been
introduced for research, diagnosis, and therapy of thrombosis; for other assays
improvements have been suggested, while a few others became redundant. The
editors waived the radioimmunoassays of ~-thrombog1obulin and platelet factor
4 due to the fact that the kits required for these assays are rarely, or no longer,
available. Also the PAI-1 activity assay was waived as it is liable to many
inconsistencies and to large variations.
   A list of names and addresses of manufacturers marketing the kits and
reagents has been compiled, together with a list of the recommended
nomenclature of quantities in thrombosis and haemostasis, in order to facilitate
the use of the updated version. These lists have been carefully compiled by
Johannes J. Sidelmann, PhD, Department of Clinical Biochemistry in Esbjerg,
Denmark.
   The editors hope that the updated second edition will add to the greatly
needed harmonization and standardization. Both are mandatory for diagnosis,
prognosis, monitoring of disease, and for the improvement of the accuracy of
tests within the field of thrombosis.
                                        ix
Preface to the First Edition
                                      xii
List of Contributors
                                       xiii
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
P. DECLERCK                                       Schwarzspanierstrasse 17
Laboratory for Pharmaceutical Biology and         A-1090 Vienna
Phytopharmacology                                 Austria
E. van Evenstraat 4
B-30oo Leuven                                     M.HAUMER
Belgium                                           Department of Vascular Biology
                                                  and Thrombosis Research
E.ERSDAL                                          University of Vienna
Chromogenix AB                                    Vienna, Austria
Taljegardsgatan 3
Molndahl, S-431 53 Sweden                         F. HAVERKATE
                                                  ECAT Foundation
E. M. FAIONI                                      Geversstraat 19 B
Hemophilia and Thrombosis Centre                  2341 GA Oegstgeest
Angelo Bianchi Bonomi                             The Netherlands
Via Pace 9
1-20122 Milan, Italy                              H.C.HEMKER
                                                  Department of Biochemistry
P. J. GAFFNEY                                     Cardiovascular Research Institute
Division of Haematology                           University of Maastricht
National Institute for Biological                 P.O. Box 616
Standards and Control                             6200 MD Maastricht
Blanche Lane, South Mimms                         The Netherlands
Potters Bar, Hertfordshire EN6 30G
UK                                                J. JESPERSEN
                                                  Department of Clinical Biochemistry
J. GRAM                                           Ribe County Hospital in Esbjerg
Department of Clinical Chemistry                  0stergade 80
Ribe County Hospital                              DK-6700 Esbjerg
0stergade 80                                      Denmark
DK-6700 Esbjerg
Denmark                                           I. JUHAN-VAGUE
                                                  Haematology Laboratory
M. R. GRIFFITHS                                   Faculty of Medicine Timone
Department of Vascular Biology                    27, Boulevard Jean Moulin
and Thrombosis Research                           F-13385 Marseille Cedex 5
University of Vienna                              France
Vienna, Austria
                                                  S. KITCHEN
A. HAEBERLI                                       Department of Haematology
Department of Internal Medicine                   Royal Hallamshire Hospital
University Hospital                               Glossop Road
Freiburgerstrasse                                 Sheffield S10 2JF
CH-3010 Bern                                      UK
Switzerland
                                                  C.KLUFT
J.HARENBERG                                       Gaubius Laboratory, TNO-PG
I. Medical Clinic                                 P.O. Box 2215
Faculty of Clinical Medicine                      2301 CE Leiden
University Hospital                               The Netherlands
Theodor-Kutzer-Ufer
D-68167 Mannheim                                  J.-C. LlBEER
Germany                                           Department of Clinical Biology
                                                  Scientific Institution of Public Health
E.HATTEY                                          Louis Pasteur
Department of Vascular Biology                    Juliette Wytsmanstraat 14
and Thrombosis Research                           B-1 050 Brussels
University of Vienna Medical Faculty              Belgium
                                            xiv
                             LIST OF CONTRIBUTORS
                                       xv
Nomenclature of haemostasis factors
7 Thrombin generation
                                             xvii
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                                             xviii
1
Introduction to laboratory assays in
haemostasis and thrombosis
J. GRAM and J. JESPERSEN
INTRODUCTION
The aim of this Assay Manual is to provide the routine laboratory with
information on the quantitation of a number of key variables within the
haemostatic system. Only via harmonization, or if possible standardization,
establishment of quality assessment schemes, and performing 'good laboratory
practice' can the routine laboratory provide 'good medical service'. Besides the
specific description of the methods of assay, these important aspects are dealt
with in depth in the chapters at the beginning of the book, including an extensive
description of sampling procedure.
   Here we shall concentrate on a short description of the haemostatic system,
the bridging between research and service, refinement of methodology, new
demands of the clinician and the laboratory, accuracy of measurement, etc.
IX
                      VIII-+ Villa
                            ~   ...            "-
       I
           /,//, ~ !~ ~":::r":nmb;O                            ~o
       \ Protein CIS                          ------                                     TAT
        \.,.                                     V -:::;tIVa
           ',\                            /    ..... -~                    .......
            "\                        I   Prothrombin ~ Thrombin
                                                                    -                    + Fragment1+2
                                 -
             ...... ~                 \
                        ,- ,_.....-
                                                                                     1
                        ~                                     ;'
                                                          /
                      .,."'0'ftlb.o. - - ---:,..1
                                          - I
                                 ftlO<1/JI;f/        I        Fibrinogen                 Fibrin monomer
                                                                                 ~
                                                          ,
                                                                                 FpA                     1
                                                                                                  Fibrin polymer
                                                              "
                 PAI-1                    t-PA                                           --- 1
                                ~~                                                                     Fibrin
                                                                               r
                Plasminogen                         ----+. Plasmin
                            T                                                            PAP             1
                                                                                                     Fibrin
                                                                        Plasmin
                        HRG                                                                        degradation
                                                                        inhibitor
                                                                                                    products
Figure 1.1 Some of the key components involved in activation of coagulation and fibrinolysis.
For details see relevant chapter
NEW METHODOLOGY
Considerable progress over the past decade, due to the development and
introduction of new assays, has provided new insight into the pathogenesis of
thrombosis and bleeding, but has also resulted in the recognition of a number
of risk indicators of cardiovascular diseases. The refinement of methodology
and increased understanding of pathophysiology has caused a shift from the
use of global clotting assays to the use of synthetic substrates, measurements
of single proenzymes and enzymes, and the use of monoclonal antibodies.
   One serious problem has always been the difficulty in converting a given
prolongation in clotting time to percentage or unit activity per mi. Although
some of the problems with the clotting assays have been reduced, new problems
have arisen.
   The use of monoclonal antibodies against many of the factors depicted in
Figure 1.1 has its own inborn problems due to formation of enzyme inhibitor
complexes, heterogeneity of proteins, formation of several split products with
the same epitope, also present in the original product, etc.
Pre-analytical factors
The pre-analytical factors which may affect measurement results of haemostatic
variables can be related to the preparation of the patient and to the blood
sampling procedure, and are described in Chapter 3.
                                        4
       LABORATORY ASSAYS IN HAEMOSTASIS AND THROMBOSES
Analytical factors
The analytical method is of great importance for the production of accurate
results in the research as well as in the clinicallaboratory7,9-11. Therefore a
strict and consistent description of a number of analytical factors is neces-
sary12,13 in order to secure transferability of accurate data between laboratories,
and in order to improve transferability from scientific publications to daily
laboratory work (Table 1.1). The principle of the method should be described,
and it should be detailed whether the measurement procedure makes use of
biological or chemical substrates for measurement of the haemostatic process.
It should be noted whether the principle causes a conflict with the application
of the method on some biological fluids. The analytical procedure should be
described in a logical sequence and preferentially in series of small steps. The
procedure of constructing a calibration curve should be detailed separately. It
should be clearly described how the measurement signal is converted to
concentration. The results should, when possible, adhere to international
WHOINIBSC standards and they should be presented in units officially accepted
by ISTH 14. Particular attention should be paid to reliability characteristics of
the method (Table 1.2). Furthermore, it should be elucidated whether the use
of the method in different laboratories gives transferable results. The reference
ranges of the method should be reported, and possible sex and age differences
of the reference ranges should be described. The intra-laboratory traceability
of data should be secured with the use of a well-described quality assurance
system. The specific daily quality assurance procedures should be described.
Finally, the characteristics of the quality control material and the frequency
of analysis should be reported.
CONCLUSION
As reviewed by others, the ideal total diagnostic test evaluation consists of five
phases: phase 1: analytical performance, phase 2: classification performance,
phase 3: clinical performance, phase 4: outcome performance, phase 5: utility
performance 15.
  This chapter exclusively deals with a proposal for minimal requirements for
analytical tests, which include the preparation of the subject under study, the
preparation of the samples and the analytical method. A proper description
                 Measurement principle
                 Analytical steps/instrumentation
                 Data reduction
                 Reliability characteristics
                 Transferability characteristics
                 Reference ranges
                 Quality assurance
                                           5
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
         Table 1.2 Reliability characteristics which may be described for an analytical
         method dealing with measurement of haemostatic quantities
References
 I. Thompson SG, Martin JC, Meade Tw. Sources of variability in coagulation factor assays.
    Thromb Haemostas 1987; 58: 1073-7.
 2. Thompson SG, Duckert F, Haverkate F, Thomson JM. The measurement of haemostatic
    factors in 16 European laboratories: quality assessment for the multicentre ECAT Angina
    Pectoris Study. Report from the European Concerted Action on Thrombosis and Disabilities
    (ECAT). Thromb Haemostas 1989; 61: 301--6.
 3. Thompson SG, Calori G, Thomson JM, Haverkate F, Duckert F. The impact of sequential
    quality assessment exercises on laboratory performance: the multicentre ECAT Angina Pectoris
    Study. Report from the European Concerted Action on Thrombosis and Disabilities (ECAT).
    Thromb Haemostas 1991; 65: 149-52.
 4. Gram J, Declerck PJ, Side1mann J, Jespersen J, Kluft C. Multicentre evaluation of commercial
    kit methods: plasminogen activator inhibitor activity. Thromb Haemostas 1993; 70: 852-7.
 5. Declerck PJ, Moreau H, Jespersen J, Gram J, Kluft C. Multicenter evaluation of commercially
    available methods for the immunological determination of plasminogen activator inhibitor-l
    (PAl-I). Thromb Haemostas 1993; 70: 858--63.
 6. Pyke SDM, Thompson SG, Buchwalsky R, Kienast J, on behalf of the ECAT Angina Pectoris
    Study Group. Variability over time of haemostatic and other vascular risk factors in patients
    suffering from angina pectoris. Thromb Haemostas 1993; 70: 743--6.
 7. Buttner J. The need for accuracy in laboratory medicine. Eur J Clin Chem Clin Biochem 1995;
    33: 981-8.
 8. International Standards Organisation (ISO) . Accuracy (trueness and precision) of measurement
    methods and results. Part I: General principles and definitions (ISOIDIS 5725-1). Geneva:
    ISO, 1990.
 9. Tietz Nw. A model for a comprehensive measurement system in clinical chemistry. CIin Chem
    1979; 25: 833-9.
10. Dybkaer R. Reference materials - a main element in a coherent reference measurement system.
    Eur J Clin Chem Clin Biochem 1991; 29: 241--6.
11. Bowers GN. Clinical chemistry analyte reference systems based on true value. Clin Chem
    1991; 37: 1665--6.
12. Fraser CG, Geary TO, Worth HGJ. Guidelines (1988) for preparation of laboratory procedure
    manuals for clinical chemistry. J Clin Chem Clin Biochem 1988; 26: 45-9.
                                               6
        LABORATORY ASSAYS IN HAEMOSTASIS AND THROMBOSES
13. Stamm D. Recommendation for description of a selected method in clinical chemistry. J Clin
    Chern Clin Biochern 1979; 17: 280--2.
14. Committee of the International Society on Thrombosis and Haemostasis (ISTH/SSC) and
    the Commission/Committee on Qualities and Units in Clinical Chemistry of the International
    Union of Pure and Applied Chemistry - International Federation of Clinical Chemistry
    (IUPAC-IFCClCQU (CC)). Nomenclature and units in thrombosis and haemostasis. Thromb
    Haemostas 1994; 71: 375-94.
IS. Magid E. Minimal requirements for test evaluation. Scand J Clin Lab Invest 1997; 57(Suppl.
    227): 90--4.
16. Bruns DE, ed. Reporting diagnostic accuracy. Clin Chem 1997; 43: 2211.
                                              7
2
Good medical laboratory services
guidelines
J.-C. LIBEER
INTRODUCTION
                                        9
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
All these points have contributed to specific proposals for quality systems for
medicallaboratories l1 . The proposals are intended to cover the pre- and
post-analytical phase, and also include national requirements which are not
always relevant for a quality system. In the meantime international organiza-
tions have tried to propose some more harmonized guidelines. ECLM (European
Confederation of Laboratory Medicine) is in favour of accreditation rather
than of certification of medical laboratories. Medical laboratories must follow
at least appropriate standards and guidelines developed for laboratories. ECLM
has reviewed the ISO/IEC Guide 25 from the point of view of medical
laboratories in an explanatory document12. The EC4 group (European Com-
munity Confederation of Clinical Chemistry) produced a consensus document
with essential criteria for quality systems in clinical biochemistry laboratories
in the EU 13. It comes as no surprise that all these different approaches embarrass
medical laboratory workers who are not specialized in this matter. The medical
laboratory profession has often tried to demonstrate the particular features of
their profession with regard to other laboratory fields, and even to other
countries. However, the same can also be demonstrated for every laboratory
field. If we look more at the common elements than at the differences, it is
possible to develop a harmonized approach on quality systems in accordance
with other laboratory fields. Each national proposal, and even the international
proposals, for a quality system in medical laboratories can be split into four
levels, as presented in Figure 2.1. Medical laboratories can use for their quality
management system, and for the technical and analytical competence, the same
approaches as other laboratory fields. The new ISO 25 guide especially covers
all quality system elements from the ISO 9000 series, and also covers many
aspects for which medical laboratories had been criticized. With the exception
of medical competence for staff members, clinical audit and more security
aspects, the new ISO 25 also covers the requirements from the EC4 document.
   In order to maintain a harmonized approach these additional aspects could
be combined in a third level. As social security systems, even in the EU, are
not yet harmonized, we will find some specific national (or regional) require-
ments in a fourth level. Usually, proposals for quality systems in medical
laboratories use an approach based on pre-analytical, analytical and post-
analytical procedures, and require a description of all phases of laboratory
analysis from the moment that a sample enters the laboratory until the sending
of the report to the requesting physician.
                                        11
             LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                                     MEDICAL COMPETENCE
                                     (EN?/ISO?)
  Concepts of TQM in medical laboratories will not only consider the quality
of service provided, but will also refer to other linked aspects such as safety
requirements, environmental impact and ethical considerations.
GMLS require that tests requested must be appropriate to the clinical problem,
tests must be analytically correctly performed and the results interpreted
correctly. Requests for tests and interpretation of results belong to medical
quality management. Correct analytical results are based on reagents, equipment,
sample quality and the whole process within the laboratory, including human
resources. Tools for quality management within the laboratory are system
quality and analytical quality management. A part of the quality is defined
outside the laboratory. Medical laboratories frequently use industrially prepared
kits and systems; their quality will be defined by the manufacturer and not by
the user. Sampling is often performed outside the laboratory. All elements
influencing the quality before samples reach the laboratory must be managed
in the pre-laboratory phase. After the release of the protocol with the laboratory
results, the requesting clinician must adequately use the data provided. This
phase of GMLS can be considered as a post-laboratory management layer.
                                                 13
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Choice of tests
Appropriateness of tests can be obtained only by dialogue between clinician
and clinical pathologist. A better understanding and collaboration between
the two will cut costs and prove more effective. The ultimate question should
be: 'will the patient find a benefit after undergoing or not undergoing this
laboratory test'. Strategies for diagnosis, prognosis or monitoring of diseases,
and whether an appropriate effect on the health care outcome for an individual
patient can be achieved, are extremely helpful in the choice of tests. In France
biologists have developed a system for helping requesting physicians in the
choice of test selections (references medicales opposables RMO and references
medicales positives)19. This example illustrates the role that can be played by
clinical pathologists in appropriate laboratory tests prescription behaviour.
LABORATORY KNOW-HOW
GMLS also requires fundamental and practical analytical and medical know-
ledge. Today this know-how is not always available in some laboratories. As an
example we mention here therapeutic drug monitoring. New technology allows
every laboratory to measure drug levels. A quality system can guarantee a
correct analytical result. However, the interpretation of these results demands
a fundamental and practical knowledge of pharmacokinetics, therapeutic range
and metabolism. Results without clinical validation are worthless to the clini-
cians; but currently many mistakes are observed in the information given to
clinicians, due to lack of knowledge by laboratory staff.
    The very rapidly changing techniques, and the introduction of new tests,
will require continuous training of laboratory staff. Although continuous
training is necessary in each profession, a mandatory system has been installed
in several countries for medical staff. These systems require the collection of
a number of CFU (continuous formation units) during a year. However, this
system does not guarantee that medical laboratory staff have real competence
in all those analyses performed in their laboratory. Without this competence
it is impossible to validate both analytical and clinical results and to guarantee
GMLS.
                                       16
                  GOOD MEDICAL LABORATORY SERVICES
CONTROL OF GMLS
CONCLUSIONS
                                                pre-laboratory
                                                analytical quality
                                                layer
          : ~~t~ ~~
          I
,I
                                          •••
                                         ••
        laboratory
         know-how                                           Post-laboratory
                                                            quality layer
                                        t
                              GMLS
Figure 2.2 The good medical laboratory services model
References
 1. OECD. Principles of good laboratory praxis. Monographs no. 45. Paris, 1992.
 2. Haeckel R. Definition of good laboratory services, TQM and (overall) laboratory management,
    norms and concepts for the accreditation of laboratories. Abstracts, Medlab 12th IFCC
    European Congress of Clinical Chemistry, 17-22 August; Basel, 1997; 15, abstract Ll5.
 3. Levey S. Jennings ER. The use of control charts in the clinical laboratory. Am J Clin Pathol
    1950;20: 1059-66.
 4. Shewhart WA. Economic control of quality of the manufactured product. New York: Van
    Nostrand, 1931.
 5. Westgard JO, Barry PL, Hunt MR, Groth T. A multi-rule Shewhart chart for quality control
    in clinical chemistry. Clin Chem 1981; 27: 493-501.
 6. Westgard JO, Barry PL. Cost-effective quality control: managing the quality and productivity
    of analytical processes. Washington, DC. AACC Press, 1986.
 7. Belk WP, Sunderman Fw. A survey of the accuracy of chemical analyses in clinical laboratories.
    Am J Clin Patholl947; 17: 853--61.
 8. Deming WE. Out of the crisis. Cambridge, MA: MIT Center for Advanced Engineering
    Study, 1982.
 9. European standard EN 45001, CEN/CENELEC. The joint European Standards Institutions,
    BINIlBN, Brussels, 1989.
10. ISO/IEC guide 25. General requirements for the competence of calibration and testing
    laboratories. Geneva: draft 5, 1997.
11. Libeer JC. Total quality management for clinical laboratories: a need or a new fashion? K.lin
    Lab Diag 1997; 5: 46--7.
12. EALlECLM. Accreditation for medical laboratories. Guidance on the interpretation of
    ISO/lEe. Guide 25. EAL-G25, ECLM-l, 1995.
13. Jansen RTP, Blaton V, Burnett D et af. Essential criteria for quality systems in medical
    laboratories. Eur J Clin Chem Clin Biochem 1997; 35: 121-2.
14. ISO 9001. Quality systems - model for quality assurance in design development, production,
    installation and servicing. Geneva: ISO, 1993.
15. ISO 9002. Quality systems - model for quality assurance in production, and servicing. Geneva:
    ISO, 1993.
16. EN 46001: Quality systems - Medical devices - particular requirements for the application
    of EN 29001. CEN/CENELEC, 1993.
17. EN 46002: Quality systems - medical devices - particular requirements for the application of
    EN 29002. CEN/CENELEC, 1993.
18. Proposal for a European Parliament and Council directive on in vitro diagnostic medical
    devices. Brussels: European Commission, April 1995.
19. Su1klaper, I. References medicales opposables (RMO): les 248 projets pour 1997. OptionlBio
    1997; 187: 13-14.
20. Buttner J. Diagnostic validity as a theoretical concept and as a measurable quantity. Eur J
    Clin Chem Clin Biochem 1995; 33: AI04-5.
21. Burnett D. Understanding Accreditation in Laboratory Medicine. London: ACB Ventura
    Publications, 1996.
                                              19
3
Blood collection and sample
preparation: pre-analytical variation
I. D. WALKER
INTRODUCTION
PATIENT REQUIREMENTS
The haematocrit varies according to posture, and differences are noted even
between sitting and lying - the haematocrit increasing significantly as subjects
assume a more erect position 1. The effect which changes in haematocrit make
to the plasma concentrations of haemostasis components are further accentuated
by the effects of dilution with liquid anticoagulant in the sample tube. The
change in haematocrit which follows alteration of position takes around 20-30
min to complete. Most hospital inpatients are venipunctured as they lie in or
on a bed, but most outpatients will have their blood sampled whilst they are
sitting, and often within less than 20 min of having walked into the consulting
room. Whilst it is impractical to suggest that, to gain uniformity, all patients
should be sampled after lying down for a period of at least 30 min - for some
tests this may be preferable or even mandatory, and in some studies it may be
essential to make a definite choice to sample participants in a particular posture
- either sitting or lying.
    Some haemostasis components show distinct circadian variation and
although, for most routine diagnostic purposes, blood specimens are collected
at random times of day, for those variables which show a marked circadian
variation, standardizing the timing of blood collection may be necessary. In
some instances it may also be important to standardize blood sampling to a
particular season or a particular point in the menstrual cycle.
    Frequently where laboratories standardize the timing of specimen collection
they seek to standardize other factors which may also influence haemostasis
variables - for example food and alcohol intake, caffeine consumption, cigarette
smoking and stress - all of which have documented effects on haemostasis,
particularly on factor VIlle and von Willebrand factor and on fibrinolytic
system components.
   Patients are sometimes given detailed instructions in an effort to avoid the
effects of smoking, food intake, caffeine or alcohol on test results. However,
unless the intent is to study the effect of these 'lifestyle'variables, over-zealous
preparation of patients may result in considerable deviation from the individual's
normal habits and risk producing unrepresentative results or, worse, results
which have been influenced by the effects of abstention. It is important in
preparing patients for venipuncture that care is taken to ensure that the sampling
conditions meet the requirements of the diagnostic or research questions to be
answered. Once it has been decided which clinical variables must either
be eliminated or accounted for, and how, most simply, this objective is to be
achieved, then it is a relatively simple step to define the patient preparation
                                       22
             BLOOD COLLECTION AND SAMPLE PREPARATION
VENIPUNCTURE TECHNIQUE
                                        24
             BLOOD COLLECTION AND SAMPLE PREPARATION
                                       25
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
SPECIMEN PROCESSING
All specimens for haemostasis tests must be labelled with the patient's full name
and identification details. The labels should be placed on the sample tubes at
the time of sample collection and in the patient's presence.
   Even small fibrin clots in the specimen render it unacceptable for investigation.
Specimens which have less than 90% expected fill of the collection tubes must
be rejected and another specimen requested. Haemolysis may affect the APTT
result but apparently not the PT result4 . After centrifugation the plasma should
be inspected for haemolysis, and haemolysed plasma should not be used for
APTT tests. Coagulometers using an optical detector may have problems with
end-point determinations on samples which are icteric or lipaemic, and alter-
native methods should be used.
Specimen transport
As soon as the specimen has been withdrawn from the patient's vein it is
beginning to deteriorate. It must therefore be transported as quickly as possible
to the laboratory. During transport specimens should be handled gently to
minimize trauma - in particular vibrational trauma - which may cause hae-
molysis and activation of coagulation. Sample tubes must remain tightly capped
to prevent loss of carbon dioxide and a subsequent rise in pH. Ideally tubes
should be held upright during transport.
   There is conflicting advice about the temperature at which specimens should
be held prior to centrifugation. Previously it was recommended that blood
specimens for haemostasis tests should be immediately placed on melting ice
or the equivalent ll . Whilst this is certainly possible in some circumstances it
is not always practical, and indeed in view of the well-documented effect of
cold activation of factor VIIC it may be preferable to maintain blood specimens
at room temperature (15-20 0c) if factor VIIC activity assay (Chapter 10) is
required or for a PT I2 (Chapter 6).
   Koepke et al. 13 demonstrated that there is no significant change in either PT
or APTT in specimens stored at room temperature for 6 h. However, the APTT
has been shown to shorten during storage at room temperature in specimens
taken from patients receiving heparin 14. This effect can be minimized by collecting
blood into CTAD to inhibit the release of platelet factor 4, a heparin ant-
agonist l5 .
   The rate of factor VIIIC deterioration at room temperature is similar to
that at 4 °C I6 . Specimens for factor VIIIC assay (Chapter 11) therefore do not
need to be maintained at 4°C, but it is im~ortant that specimens for VIIIC
assay are analysed within 2 h of collection 1 •
platelets in the plasma sample following centrifugation may interfere with the
planned test or assay, and to control and standardize the speed and duration
of centrifugation according to the particular requirements of each test to be
performed.
   For most tests and assays it is suggested2 that the minimum centrifugation
criteria should be 2500g for 15 min. For some tests it is essential to ensure that
as far as possible the plasma sample is rlatelet free. For this purpose a double
centrifugation may be recommended 1 • The centrifuge brake should not be
used. After centrifugation plasma must be drawn off carefully using a non-
contact pipette. Great care must be taken to avoid disturbing the platelet layer.
Deterioration of clotting factors can be minimized by rapid processing and
double centrifugation to eliminate most cells 17 . Samples which will not be
tested immediately but which are to be stored frozen may be improved by double
centrifugation.
Sample storage
The maximum acceptable time interval between venipuncture and testing of
the sample will depend on the temperature maintained during transport and
storage. NeeLS recommend the following: 2 h for samples stored at 22-24 DC,
4 h for samples stored at 2-4 DC, 2 weeks for samples stored at -20 DC and 6
months for samples rapidly frozen and stored at -70 De 2• In general clotting
factors in frozen plasma will be more stable at lower temperatures. Frozen
samples should be stored in small aliquots in well-identified containers which
are tightly capped. Ideally samples which will not be tested fresh should be
snap-frozen. If liquid nitrogen is available this should be the method of choice
for rapid freezing, but the process can also be achieved by using a refrigerated
bath at -50 DC or a mixture of dry ice and acetone (provided the containers
are resistant to acetone). If none of these methods is available plasma aliquots
may be placed in the deep-freeze at the lowest possible temperature available
to ensure rapid cooling and freezing.
   The temperature during frozen storage must be carefully controlled. Frozen
plasma samples should be thawed rapidly at 37 DC according to a standardized
protocol and tested immediately after thawing before the sample warms. If
testing cannot be performed immediately the sample may be held for a maximum
of 2 h at 4 DC until tested2 •
References
 1. Leppanen EA, Grasbeck R. Experimental basis of standardised specimen collection effect
    of posture on blood picture. Eur J Haemato11988; 40: 222-6.
 2. NCCLS. Collection, Transport, and Processing of Blood Specimens for Coagulation Testing
    and Performance of Coagulation Assays. Villanova, PA: NCCLS Document H21-A2, Vol.
    11, No. 23, 1991; 1.
 3. KIuft C, Meijer P. Update 1996: Blood collection and handling procedures for assessment of
    plasminogen activators and inhibitors. Fibrinolysis 1996; 10 (SuppL 2): 171-9.
 4. Gilmer PRo Preanalytical variables in blood coagulation testing. In Triplett DA, ed.
    Standardisation of coagulation Assays: an overview. Skoki Illinois: College of American
    Pathologists, 1982; 1-8.
 5. van Putten JJ, van de Ruit M, Beunis M, Hemker He. Heparin neutralisation during collection
    and processing of blood inhibited by pyridoxal5'-phosphate. Haemostasis 1984; 14: 253-61.
 6. ECCLS. Standard for Specimen Collection Part 1: Blood Containers, Vol. 3, 1983: 1.
 7. van den Besselaar AMHP, van Hallem-Vissei LP, Loeliger EA. The use of evacuated tubes
    for blood collection in oral anticoagulant control. Thromb Haemostas 1983; 50: 676-7.
 8. Ingram GIC, Brozovic M, Slater NGP. Bleeding Disorders Investigation and Management,
    2nd edn. Oxford: Blackwell Scientific Publications 1982; 244-5.
 9. Meijer P, van der Ham F, KIuft C. The use of Stabilyte® plasma may cause changes in pH in
    the assay of some fibrinolysis analytes and might affect results. Fibrinolysis 1996; 10 (SuppL
    2): 155-7.
10. Polak B, Barro C, Mossuz P, Pernod G. Inadequate quality of a blood collection tube containing
    an anticoagulant/platelet inhibitor mix. Thromb Haemostas 1997; 77: 1035-6.
11. NCCLS. Tentative guidelines for the standardised collection, transport and preparation of
    blood specimens for coagulation testing and performance of coagulation assays. Villanova,
    PA: NCCLS Publication, Vol. 2, No.4, 1982, 103-28.
12. Thomson 1M. Specimen collection for blood coagulation testing. In Koepke JA, ed. Laboratory
    Haematology. New York: Churchill Livingstone, 1984; 833-63.
13. Koepke JE, Rodgers JL, Ollivier MJ. Preinstrumental variables in coagulation testing. Am J
    Clin Patho11975; 64: 591-6.
14. van den Besselaar AMHP, Meeuwisse-Braun J, Jansen-Gruter R, Bertina RM. Monitoring
    heparin therapy by activated partial thromboplastin time. The effect of preanalytical condi-
    tions. Thromb Haemostas 1987; 57: 226-31.
15. van den Besselaar AMHP, Bertina RM. Standardisation and quality control in blood
    coagulation assays. In: Lewis SR, Verwilghen RL, eds. Quality assurance in haematology.
    London: Bailliere Tindall, 1988; 119-50.
16. Rotbalt F, Tuddenham EGD. Immunologic studies of factor VIII coagulant activity (VIIIC)
    assays based on a haemophilic and an acquired antibody to VIlle. Thromb Res 1981; 21:
    431-45.
17. DIN. Einstufenmethode zur Bestinunung der Faktor VIII-gerinnungsaktivitiit (FVIIIC).
    Deutsches Institut fUr Normung 58909, Teill, 2. Berlin: Beuth Verlag, 1987.
                                              28
4
Quality assessment of haemostatic
assays and external quality
assessment schemes
T. A. L. WOODS, S. KITCHEN and F. E. PRESTON
INTRODUCTION
The coagulation laboratory has a vital role in the diagnosis and management
of patients with familial and acquired haemorrhagic and thrombotic disorders.
Wherever possible the laboratory methods employed must reflect the state of
the art, and the results generated, including locally derived reference values,
should be reliable, reproducible and unambiguous. These issues are of particular
importance when investigations are undertaken for the diagnosis of a possible
familial disorder. When an error occurs, there is possibility of misdiagnosis
and, irrespective of whether this causes an individual to be misdiagnosed as
either having or not having a familial defect, the clinical consequences of such
a mistake are likely to be serious. Within recent years the workload and scope
of the routine laboratory has increased substantially, and this has been accomp-
anied by the introduction of automated equipment employing a variety of
different technologies. The potential for laboratory error is therefore considerable,
and monitoring of laboratory performance through a programme of quality
assurance is an essential laboratory requirement.
Quality assurance (QA) is an overall term that may be used to describe all
measures that are taken to ensure the reliability of laboratory testing and
reporting. This will range from the choice of test, the collection of a valid
sample from the patient, analysis of the specimen and the recording of results
                                        29
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
in a timely and accurate manner, through to interpretation of the results, where
appropriate, and communication of these results to the referring clinicians.
   Internal quality control (IQc) and external quality assessment (EQA) are
two distinct, yet complementary, components of a laboratory QA programme.
IQC is used to establish whether a series of techniques and procedures are
performing consistently over a period of time. It is therefore deployed to ensure
day-to-day laboratory consistency. EQA is used to identify the degree of agree-
ment between one laboratory's results and those obtained by other centres. In
large EQA schemes retrospective analysis of results obtained by participating
laboratories permits the identification not only of poor individual laboratory
performance, but also those reagents and methods that produce unreliable or
misleading results.
90~-------------------------------------,
80 I- ..•...............................•.•.......
..
 ~
 -s:
 ~
 ctI   70 - .
 ()
:>
 L-
 o
 ~
LL
                                            Date of test
Figure 4.1 Results of factor VIII:C assays on an internal quality control sample assayed on
different days_ Each point is a different assay on the same material. The solid lines represent the
mean and 2 standard deviations of 20 assays on this material, considered to represent the limits
of acceptable results
                                               33
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   Increasingly effective EQA scheme designs have evolved through the combined
effects of experience gained over many years, and the continuing and necessary
interactions that take place between participants and the scheme organizer. A
number of criteria have emerged as the fundamental building blocks of a success-
ful scheme. These may be summarized as:
1. Adequate topical data; for example, sufficient data returns for statistical
   analyses. This will be influenced by: (a) adequate number of participants;
   (b) prompt return of results from participants; (c) sufficient number of
   analytes in each distribution; (d) frequency of distribution.
2. Effective communication of performance data, through: (a) well-composed,
   informative and easily understood reports; (b) sequential historical grades.
3. Relevant structure for performance assessment: (a) samples that are stable
   and consistent; (b) matrix that is compatible with clinical samples.
International Normalized Ratio (INR) based on: (a) Quick's one-stage method,
(b) capillary reagent method.
Prothrombin time (diagnosis).
Activated partial thromboplastin time (APTT).
Thrombin time.
                                        35
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
References
 1. Thompson SG. Martin JC, Meade Tw. Sources of variability in coagulation factor assays.
    Thromb Haemostas 1987; 58: 1073-77.
 2. Dot D, Miro J, Fuentes-Arderiu X. Within-subject and between-subject biological variation
    of the prothrombin time and activated partial thromboplastin time. Ann Clin Biochem 1992;
    29: 422-5.
 3. Costongs GMPJ, Bas BM, Janson PCW Short-term and long-term intra-individual varia-
    tions and critical differences of coagulation parameters. 1. Clin Chern Biochem 1985; 23:
    405-10.
                                            36
5
Activated partial thromboplastin time
(APTI)
L.POLLER
INTRODUCTION
The term partial thromboplastin is used to distinguish the reagent from that
used in the prothrombin time, since the APTT reagent lacks the apoprotein
component of the complete tissue thromboplastin. The APTT is the main test
for screening for intrinsic clotting defects including haemophilia. It is also used
for detection of lupus anticoagulant and for laboratory monitoring of heparin
administration. The presence of an activator in the test system, to accelerate
the PIT test by effecting maximum activation, also increases its precision and
reproducibility by eliminating the variable effects of contact with glass surfaces.
APTT PHOSPHOLIPIDS
Thepartialthromboplastinconsistsofthephospholipidcomponentofthrombo-
plastin and is prepared from animal tissue or from vegetable sources. The
phospholipid acts as a platelet substitute in the intrinsic system. The lipid
composition of different APIT reagents, however, varies considerably. The
total concentrations of phospholipid and fatty acid in some widely used APTT
reagents have been shown to differ by as much as 300 times!. These discrepancies
markedly affect responses to coagulation defects and inhibitors of coagulation.
The requirements for the phospholipids in the test system may also vary
according to the nature of the clotting defect being measured. For example,
the concentration of negatively charged phospholipids, e.g. phosphatidyl serine,
has been shown to be criticae.
                                       37
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
OTHER COMPONENTS OF THE APTT TEST
Factors which affect the clotting response include the type of activator, length
of incubation with the plasma and the presence of buffers 3 •4 • Particulate
activators include kaolin, celite and micronized silica, whereas a commonly
used activator, ellagic acid, is non-particulate. The amount of activator present
in the various commercial techniques, and the length of incubation time
employed with the plasma, show considerable variation. The effectiveness of
an activator in the APTT is governed by many considerations, e.g. its con-
centration, the incubation time and the composition of the phospholipid. It is
the combination of the activator with the other components which appears to
determine the reliability of the tese. The trend is to use less opaque activation
to avoid interference with newer types of coagulometers. The use of different
types of coagulometer can also have a considerable effect on the clotting time6 .
CLINICAL USE
The main use of the APTT is for the screening of coagulation defects and the
presence of inhibitors. The test is prolonged by deficiencies of factors VIII,
IX, X, XI and XII and defects of the contact phase, e.g. prekallikrein, high
molecular weight kininogen. It also may be prolonged by gross defects of
factors II, V and fibrinogen. With reliable APTT systems, specific and
non-specific inhibitors of intrinsic clotting factors are detected. The degree of
abnormality depends upon the responsiveness of a particular APTT method
to a specific defect. When used as a screening test for lupus anticoagulant (LA)
                                       38
                   ACTIVATED PARTIAL THROMBOPLASTIN TIME
(Chapter 19) the detection rate is greatly influenced by the concentration and
type of phospholipid content of the reagent. Reagents containing the highest
concentration of phospholipid tend to be less responsive to LA. The APTT is
also the most widely used method for the laboratory monitoring of heparin
administration I 0, II. Although other more specific techniques have been advo-
cated, e.g. anti-Xa assays, the APTT has been almost universally preferred as
it is regarded as a global test of coagulation which assesses the overall effect
of heparin on clotting. The APTT may also be of value in case of unexpected
bleeding during anticoagulant therapy as there may be disproportionate
depression of factor IX. The APTT is also used as in the ECAT study as an
overall assessment of the intrinsic clotting system. Accelerated APTT have
been reported after operations, oral contraceptive administration and withdrawal
of oral anticoagulation.
The need for standardization of the APTT has been demonstrated in many
reports, partic~arly with regar~ to th.e ~de~pread a.ppli~a~ion of the test in
laboratory momtonng of hepann admimstratlOn 3,12-1 • Ongmally Basu et al. 18
recommended that the therapeutic range for heparin should be given by an
APTT ratio between 1.5 and 2.5. The intensity of treatment corresponding to
this range varies, however, with the local APTT test system. The variable
responses reagents in the US and UK demonstrated by national external quality
assessment surveys are illustrated in Figure 5.1. The different responses on
fresh samples from heparin-treated patients resulting from the use of different
coagulometers is shown in Figure 5.2. A calibrations constant was devised for
3.0
                  o
                  ~
                  ....
                  ~2.                                                D·
                 c..
                 «                                                   o·
                               .-""'=----+-+-r ----j-+---,----------,
                                           0.2            0.4               0.8
                                       M GOO
                                                 iu/ml
Figure 5.1 AP1T ratios from surveys from the College of American Pathologists aod UK external
quality assessment showing responses to in-vitro heparinized samples. The dotted line indicates
the Manchester AP1T response on flesh plasma from heparinized patients. The concentration of
heparin giving a 1.5 APTT ratio with the individual reagents is indicated by vertical arrows: M =
Manchester; G = GD Automated; D = Dade; 0 = Ortho
                                                 39
              LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
2.01····· 1: Electra
      ~ 1.5+ . .
                                                                                                                                                 2: KC
                                                                                                                                                 3: ACL
      m            I                                                                                                                       •     4: Coag·a·mate
      1ii
      c:           I
                   I
                                                                                                                                           •     5: Schnitger & Gross
                                                   !- ...........................................
      ~ 1.0 ............, ........... : ............                                                    ......................................   6:· (manual)
      o .                                                                      I             :                                             •     7: Cobas·Fibro
      .~                                                                                                                                         8: Fibrintimer
                                                                                                                                                 10: coagulometers in
      ~     0.5                                                                                                                                      use in 1 Laboratory
      (,)
                  o                      2             3         4             5             6      7                    9                10
                                                           Type of coagulometer
the ISTHlICSH study based on the orthogonal regression slope (as in proth-
rombin time standardization) of the plots of the log APTf results of the same
two reagents at different centres using different coagulometers8 . The marked
effect of the coagulometers is seen. Hirsh et al. 19 gave the equivalents for an
APTT ratio of 1.5 for the protamine titration as 0.2 units/ml and for the
anti-factor X assay as 0.3 units/mL The difficulty of employing these alterna-
tives for standardization is that the vast majority of centres prefer to use the
APTf for heparin control as it is regarded as a measure of the overall biological
response to heparin. As far as the heparin response is concerned anti-Xa levels
of 0.3 to 0.7 ulml have been recommended to demarcate the APTT ratio
prolongation 19. A problem with attempts to standardize heparin monitoring
by APTT is that APTT test systems differ in response to components of the
clotting mechanism other than heparin, e.g. anti-heparin activities. The
difficulties of standardization of APTT heparin monitorinl were highlighted
by the recent ISTHIICSH international collaborative study. APTT responses
of patients with recent thrombosis were much less to the same concentrations
of heparin. A calibration constant was determined for each local APTf system
from the orthogonal regression slope of plasmas from patients and healthy
subjects. It was found that the use of a single slope value for a brand of reagent
for all laboratories was not advisable, and each laboratory should perform its
own local calibration. The study recommended that the adoption of a reference
APTT reagent would be a useful step to initiate heparin monitoring standardi-
zation and to develop safe and effective local therapeutic ranges.
Stress, exercise, pregnancy, the post-partum state and surgical operations result
in acceleration of the test. Pro thrombotic changes associated with recent
deep-vein thrombosis, thromboembolic disorders and oestrogen administration
may result in accelerated clotting times4 • Acquired pathological states such as
liver disease, disseminated intravascular coagulation (DIC), and drug toxicity
cause prolongation. Certain drugs, including oral anticoagulants, heparin and
thrombolytic agents, prolong the APTT. The test is also prolonged in the new-
born20 •
                                                                                             40
               ACTIVATED PARTIAL THROMBOPLASTIN TIME
A single APTT reagent and technique was selected by the executive committee
to be employed at all participant centres in the BCAT studies because of the
variability of performance of the different APTT methods summarized earlier
in this chapter, due to lack of standardization. The Manchester APTT was
chosen as it has been shown in a number of published studies to give good
sensitivity to a wide range of coagulation disorders and inhibitors 17 • The
manual technique was selected for the BCAT studies because automated
techniques may have marked and variable coagulometer effects on APTT
results. To gain familiarity with the method, and to reduce the influence of
inter-laboratory variation in technique, representatives from all participant
centres attended training courses organized by BCAT. On-going programmes
of external quality control were conducted subsequently to monitor their
performance. As stated earlier, a variety of activators and incubation times are
employed in current conventional preparations. The trend is to use non-opaque
activators and shorter incubation times (3-4 min). In the absence of a recognized
standard or reference preparation for the APTT the Manchester APTT may
be regarded as a reference method to assess the responsiveness of other APTT
procedures to concentrations of heparin and specific clotting factors (particularly
VIII and IX). The responsiveness to lupus anticoagulant is an additional
criterion2 . Some APTT reagents are relatively insensitive to the lupus anti-
coagulant.
Details of technique
1. Lyophilized APTT reagent (store at -20°C). Reconstitute by adding exactly
   10 ml of cold Owren's buffer (see item 2, below). The stopper is replaced
   and is mixed gently to resuspend. The diluted suspension is dispensed into
   small volumes, e.g. 1.0 ml amounts, using non-wettable containers which
   are tightly capped and immediately frozen (-20 to -40 0C). This procedure
   should be accomplished with minimum delay, preferably within 15 min from
   reconstitution. The frozen aliquots of the reagent are stable for at least 3
   months.
      An aliquot of the diluted suspension should be thawed out rapidly when
   required by placing in a water bath at 37°C for 1-2 min immediately before
   use. This is maintained in crushed ice prior to use. The reagent is stable for
   at least 2 h under these conditions. Any remaining reagent is discarded after
   use and is not refrozen.
                                       41
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
2. Owren's buffer (sodium diethylbarbiturate (11.75 g) and sodium chloride
   (14.67 g) dissolved in 1570 m1 distilled water and 430 m1 0.1 N hydrochloric
   acid (PH 7.35) for reconstitution of APTT reagent (store at + 2 to 8°C).
3. Light kaolin (0.25 g/100 ml) is suspended in Owren's buffer (store at + 2 to
   8°C). It is mixed well before use. An aliquot is decanted when required into
   a separate container and any remainder is discarded.
Method
Testing should be performed on fresh plasma, collected and stored as indicated
in Chapter 2, as soon as possible after collection and no longer than 1 h after
venipuncture. The test plasma is kept in a stoppered polystyrene container at
room temperature.
   The kaolin suspension and calcium chloride are warmed in separate test
tubes in a water bath at 37°C. The following reagents are added in the order
indicated without delay into a glass tube, pre-warmed in a water bath:
   0.1 m1 test plasma
   0.1 m1 APTT reagent
   0.1 m1 warmed kaolin suspension (after resuspension) and start stopwatch.
The test tube is tilted three times immediately, and subsequently at approximately
1 min intervals in order to resuspend the kaolin.
   At exactly 10 min, 0.1 m1 warmed calcium chloride (0.025 moUL) is added.
The tube is tilted gently three times and left undisturbed in a water bath for
exactly 20 s from the addition of the calcium chloride. It is then tilted gently
until a solid clot forms. The clotting time (seconds) is recorded and the test is
performed in duplicate.
   The normal range for the Manchester APTT is based on results from a large
normal population of both sexes, spanning the adult age range. These extend
from 36.0 to 48.0 S, based on two standard errors from the mean with correction
for non-Gaussian distribution.
SOURCES OF ERROR
COAGULOMETER TESTING
Some of the newer types of coagulometer require a much less opaque reagent
than the above procedure with kaolin. Non-opaque liquid activation (sulphatides,
ellagic acid, etc.) and other activators of low opacity, e.g. micronized silica, are
incorporated into commercial products and are more compatible with some
coagulometers than is kaolin. The variable effects of different coagulometers
on the APTT response to heparin with a modification of the Manchester APTT
method employing sulphatides as the activator have been reported8 •
References
 1. Stevenson KJ, Easton AC, Curry A, Thomson JM, Poller L. The reliability of activated partial
    thromboplastin time methods and the relationship to lipid composition and ultrastructure.
    Thromb Haemostas 1986; 55: 250-8.
 2. Kelsey PR, Stevenson KJ, Poller L. The diagnosis of lupus anticoagulants by the activated
    partial thromboplastin time - the central role of phosphatidyl serine. Thromb Haemostas
    1984; 52: 172-5.
 3. Barrowcliffe TW, Gray E. Studies of phospholipid reagents used in coagulation II: factors
    influencing their sensitivity to heparin. Thromb Haemostas 1981; 46: 634-7.
 4. Thomson JM, Poller L. The activated partial thromboplastin time. In: Thomson JM, ed.
    Blood coagulation and haemostasis: a practical guide. Edinburgh: Churchill Livingstone,
    1985; 301-9.
 5. Triplett DA, Smith C. Sensitivity of the activated partial thromboplastin time: results of the
    CAP survey and a series of mild and moderate factor deficiencies. In: Triplett DA, ed.
    Standardisation of coagulation assays: an overview. Skokie, IL: College of American Patholo-
    gists, 1982; 137--63.
 6. Koepke JA. The use of proficiency testing results in the coagulation laboratory. In: Triplett
    DA, ed. Laboratory evaluation of coagulation. Chicago, IL: American Society of Clinical
    Pathologists, 1982; 368-87.
 7. Proctor RR, Rapaport SL. The partial thromboplastin time with kaolin. A simple screening
    test for first stage plasma clotting deficiencies. Am J Clin PatholI961; 36: 212-19.
 8. van der Velde EA, Poller L. The APTT monitoring of heparin. The ISTHlICSH collaborative
    study. Thromb Haemostas 1995; 73: 73-81.
 9. Jennings I, Kitchen S, Woods TA, Preston FE. Monitoring heparin therapy. Instrument effects
    on reagent sensitivity. Thromb Haemostas 1997 (Suppl.): 279, abstract 1141.
10. Thomson JM. The control of heparin therapy by the activated partial thromboplastin time:
    sensitivity of various thromboplastins to heparin. In: Triplett DA, ed. Standardisation of
    coagulation assays: an overview. Skokie, IL: College of American Pathologists, 1982; 195-206.
11. Triplett DA. Heparin: Clinical use and monitoring. In: Triplett DA, ed. Laboratory evalu-
    ation of coagulation. Chicago, IL: American Society of Clinical Pathologists, 1982; 271-313.
12. Poller L, Thomson JM. The partial thromboplastin (cephalin) time test. J Clin PathoI1972;
    25: 1038-44.
                                              43
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
13. Koepke 1. The partial thromboplastin time in the CAP survey programme. Am J Clin Pathol
    1974; 63: 990--4.
14. Shapiro GA, Huntzinger SW, Wilson JE. Variations among commercial activated partial
    thromboplastin time reagents in response to heparin. Am J Clin Patho11977; 67: 477-80.
15. CISMEL (Italian) Study Group. Activated partial thromboplastin time - a multicentre evalu-
    ation of commercial reagents in the diagnosis of haemophilia. Scand J Haematol1980; 25:
    308.
16. Brandt IT, Triplett DA. Laboratory monitoring of heparin. Effects of reagents and instru-
    ments on the activated partial thromboplastin time. Am J Clin Patho11981; 76: 530-7.
17. Mannucci PM. A multicentre evaluation of partial thromboplastin time reagents in the
    detection of mild haemophiliacs. In: Triplett DA, ed. Standardisation of coagulation assays:
    an overview. Skokie, IL: College of American Pathologists, 1982; 165-77.
18. Basu D, Gallus A, Hirsh JA. A prospective study of the value of monitoring heparin treatment
    with the activated partial thromboplastin time. N Engl J Med 1972; 287: 324-7.
19. Hirsh J, Raschke R, Warkentin TE, Dalen JE, Deykin D, Poller L. Heparin: mechanism of
    action, pharmacokinetics, dosing considerations, monitoring, efficacy and safety. Chest (Suppl.)
    1995; 108: 258-75S.
20. Hathaway WE, Bonnar J. Physiology of coagulation in the fetus and newborn infant. In:
    Perinatal coagulation. New York: Grune & Stratton, 1978; 69.
21. Refsurn N, Abildgaard U. 'Cephotest' in monitoring heparin therapy. Evaluation of a
    standardised activated partial thromboplastin time (AP1T) test. Farmakoterapi 1976; 1-2:
    27-35.
22. Shapiro GA, Huntzinger SW, Wilson JE. Variations among commercial activated partial
    thromboplastin time reagents in response to heparin. Am J Clin PathoI1977; 67: 477-80.
23. Van den Besselaar AMHP, Meeuwisse-Braun J, Jansen-Gruter R, Bertina RM. Monitoring
    heparin therapy by the activated partial thromboplastin time - the effect of pre-analytical
    conditions. Thromb Haemostas 1987; 57: 226-31.
                                               44
6
Prothrombin time (PT)
L.POLLER
INTRODUCTION
The prothrombin time (P1) is the screening test for the extrinsic (tissue) clotting
system. The PT was originally introduced by Quick l as a measure of a
coagulation defect in the newborn and in jaundiced patients, and subsequently
adapted for its present principal uses in the screening of extrinsic clotting and
in monitoring of oral anticoagulant dosage. Although specific methods were
subsequently developed designed for anticoagulant control, e.g. the P&P test2
and Thrombotese the Quick test continued in most countries to be the exclusive
control method. The PT is also the screening test for defects of extrinsic (tissue)
clotting. Its responsiveness to depression of the extrinsic clotting factors depends
on the source and type of tissue factor (TF) thromboplastin extract. TF is a
combination of an apoprotein and phospholipid. It has been purified and its
chemical structure has been defined4 ,5. Human recombinant TF relifdated
with natural or synthetic phospholipids is now being widely employed 8. The
EeAT study has been concerned principally with its application of the PT in
assessing extrinsic clotting function. The PT reflects changes in three of the
vitamin K-dependent clotting factors (factors II, VII and X), and the non-vitamin
K-dependent factor V. In the PT the recalcification time of citrated plasma is
accelerated by a TF extract. The test depends on activation by TF of factor X
in the presence of factor VII and the bypassing of the intrinsic pathway. The
speed of this reaction and the responsiveness of the PT to deficiencies of
clotting factors depends upon the properties and concentration of the TF, as
well as to the clotting factor concentration.
                                       45
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
SOURCES OF TISSUE FACTOR (TF)
Many modifications of the test have been introduced since Quick's original
rabbit brain extract. Other tissues in addition to rabbit brain have been used
as a source of TF in routine work, e.g. human brain, ox brain, monkey brain,
rabbit lung and human placenta. TF is a combination of an apoprotein and
phospholipid and is a complete thromboplastin as opposed to the partial
thromboplastin of the activated partial thromboplastin time (APTT) which
lacks the apoprotein. It has been purified and its chemical structure has been
defined4 ,5. Human recombinant TF is now being produced which, when re-
lipidated, can be employed as the TF in the PT test6 ,7. The dangers of virus
transmission from human tissue led to the most widely used preparations being
derived from rabbit brain, but there is now increasing demand for the human
recombinant preparations because of the avoidance of virus transmission
combined with high responsiveness. TF extracts vary in their procoagulant
properties and their ability to detect alterations in individual clotting factors
or systems. The reagent used in the ECAT study was a responsive rabbit brain
extract with a similar low International Sensitivity Index (lSI) to the new human
recombinant reagents.
2.0
1.8
         t="
         e..
          Ol
         g
          c      1.6
         ~
         ..!l!
          Co
         .8                                                0
          E
          e
         J::
                 1.4                                                  o Patients (n=60j
         f-                                                           + Normals (n=20j
         e..
         9;
1.2
                 1.0   -1----..-------.------.------.------.
                       1.0    1.2          1.4           1.6          1.8           2.0
Figure 6.1 Example of orthogonal regression slopes in lSI calibration showing good coincidence
of patient/normal and patient-only lines
terms of the first primary WHO IRP for thromboplastin (human combined
67/40), and as this reagent is no longer available against a secondary reference
preparation calibrated in terms of the first primary WHO, the responsiveness
of which was by definition 1.0. The slope of the orthogonal regression line
from which lSI are derived is obtained from plotting the log PT of 20 healthy
subjects and of 60 long-term-stabilized coumarin-treated patients' samples with
the local thromboplastin against the log PT with the relevant IRP (see Figure
6.1) as experience has shown that these numbers give reliable results within the
3% limits of precision of the CV of the slope of the calibration line. Responsive
thromboplastins have an lSI close to 1.0, whereas less responsive reagents have
higher values.
   To enable manufacturers of thromboplastin to relate their materials to the
WHO reference thromboplastin standard, three further reference thromboplastins
were later designated IRP. These were BCf/099 (human, plain), OBT179 (bovine,
combined) and RBT179 (rabbit, plain) to be used with the relevant species of
routine thromboplastin.
   Manufacturers of thromboplastin are expected to provide the lSI of their
reagent. Each package should contain a table or graph to convert into INR
results usually expressed as prothrombin ratios obtained with the local
thromboplastin.
   The WHO calibration scheme is hierarchical (see Figure 6.2). Starting with
                                             49
             LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
the first WHO primary IRP, the calibration model proceeds through second-
and third-generation IRP down to house standards and manufacturers'
production lots. Manufacturers of thromboplastin are expected to produce
their own house standard, which should be a lyophilized batch of thromboplastin
set aside for the calibration of production batches. The WHO IRP* are in very
limited supply, and are intended only for the calibration of national reference
preparations or working reference preparations, at 'national control laboratories' ,
not manufacturers' reagents.
   The European Commission Bureau Communautaire de Reference (BCR)
currently provides, at not-insignificant cost, certified reference thromboplastins
CRM/149S (rabbit plain)'9 and OBT179 (bovine combined)20. The human
plain reagent (BCT/099) has been discontinued and not yet replaced. The CRM
are intended to be available to manufacturers of commercial thromboplastins.
The International Council for Standardization in Haematology (ICSH) made
available an additional human plain IRP (BCT/441i'. A WHO human plain
IRP (recombinant human) replacement for BCT/441 human IRP is now
approved (RTFI95).
   The precision of a calibration is greater when similar reagents and techniques
are compared. The WHO scheme states that each step of the calibration should
be made, whenever possible, against a reference preparation which is most
similar in terms of the species of tissue extract. It is also important to compare
'plain' reagents, i.e. used with a PT technique with reference reagents that are
also plain. Similarly combined (with added factor V and fibrinogen) reagents
(e.g. Thrombotest) should be calibrated with a combined reference prepara-
tion22 . Calibration of successive batches of thromboplastin should be carried
*WHO Custodian Laboratory is the Central Laboratory for Blood Transfusion, Plesmanlaan 125,
1066 ex, Amsterdam, The Netherlands.
                                                 50
                              PROTHROMBIN TIME
INR
9.0
8.0
7.0
            -cQ)
             C)
            as     5.0
                                                                       8.0
                                                                       5.0
            Q)
            a:::   4.5
            ~0 4.0                                                     4.0
                                                                             ~
               3.8
            -I 3.6                                                           E
            0 3.4                                                            :::i
            ~ 3.2                                                            0
            a:::
               3.0                                                     3.0   ~
                                                                              :::I
            C 2.8                                                             Q)
            :c 2.6
                                                                              ~
                                                                             I!!
             E
             e     2.4                                                       Q)
                                                                             r=.
            e
            .r:.   2.2
                   2.0                                                 2.0--
            D..    1.9
                   1.8
                   1.7
                   1.8
                   1.5
                   1.4
                   1.3
                   1.2
                   1.1
                   1.0 I---~--~--l---~-~::__--+ 1.0
                      1.0  1.1 1.2 1.3
                          Intemational Sensitivity Index (lSI)
Figure 6.3 Nomogram for INR derivation. Example: observed ratio 2.65, with thromboplastin
lSI 1.3; INR = 4
is also used in calibration programmes takes into account also the imprecision
of the earlier calibration(s) of the IRP and in-house reference preparations
against which a new reagent is being calibrated. The CV of the lSI is therefore
greater than the CV of the slope.
Coagulometer effects
In view of the coagulometer effect on the ISe5-28 some manufacturers provide
a 'system lSI' for a particular thromboplastin!coagulometer combination. The
limitations of the procedure have been demonstrated by the variability of the
system lSI with the same reagent and coagulometer at different centres found
in collaborative studies29,3o.
   Coagulometers have now largely replaced the manual PT technique in most
countries. They may cause a disproportionate shortening of PT results from
healthy subjects compared to the shortening effects on patients' plasmas, thus
artificially increasing the prothrombin ratio and reducing lSI. In view of this
some manufacturers provide a 'system lSI' for a particular thromboplastin!
coagulometer combination. This may assist, but the limitations of the procedure
have been demonstrated by the variability of system lSI with the same reagent
and coagulometer at different centres found in collaborative studies29,3o. The
need for local system lSI at individual laboratories has therefore been demon-
strated, as well as the need for multiple calibrations where more than the
coagulometer is in use in a single laboratory. As conventional local lSI calibration
necessitates the performance of the generally discarded manual PT technique
with the thromboplastin IRP it is therefore not now usually feasible. The use
of certified lyophilized plasma calibrants has been recommended as an alternative
for local lSI determination29- 32 • The reliability of the procedure and the
minimum requirement of numbers of certified lyophilized plasmas for a
calibration is currently being studied. A European Concerted Action on
Anticoagulation (ECAA) exercise33 confirmed that lyophilized plasmas give a
reasonable approximation to fresh plasmas in a full calibration of 60 lyophilized
versus 60 fresh plasmas from stabilized treated patients. In an international
collaborative calibration at 15 centres there was a demonstrable but small
difference (mean 4.5%) from a fresh plasma calibration of the lSI of low lSI
ECAA human recombinant reference reagent when such plasmas were used,
presumably due to a freezing or lyophilization effect. Furthermore lyophilized
artificially depleted plasma could be reliably substituted for lyophilized coumarin
plasmas from coumarin-treated patients, which makes lyophilized plasma
calibration a practical procedure. A further ECAA report34 indicated that a
minimum of 20 lyophilized depleted plasmas, with a proportion of samples
from healthy subjects comparable to a full calibration (i.e. seven normals to 20
abnormal plasma samples), is required to give a similar lSI to a full calibration
based on 60 lyophilized plasmas and 20 fresh plasma samples from healthy
individuals.
                                        53
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                              Ratios equivalent to INR range 2.0 - 3.0
       3.0
        1.5                           III I I I I I I I I                                   I I
        1.0   I   I   I   I   I   I    I    I    I    I    I   I    I    I    I    I    I   -,---,
              1.0 1.1 1.2 1.3 1.4 1.5 1.6 1.7 1.8 1.9 2.0 2.1 2.2 2.3 2.4 2.5 2.6 2.7 2.8
                                                     lSI
Figure 6.4 Effect of lSI on therapeutic window for warfarin dosage
The mathematics of the lSI calculation described in the Appendix have proved
too complex for general laboratories. The calculation has been computerized in
several centres and specialist laboratories have developed their own programs.
'National Control Laboratories' should be able to provide a formatted disk with
a recommended calibration program and its methods of use. *
"'Such a program, a modification of which is being prepared for WHO Biologicals (Geneva), is
available at nominal charge on application to the ECAA Central Facility: Professor L. Poller,
University of Manchester, Oxford Road, Manchester M13 9PT, UK or from Health Technology,
WHO, Geneva.
                                                54
                              PROTHROMBIN TIME
SOURCES OF ERROR
The test is relatively simple but subject to a number of pre-test and test variables
which affect the result. Faulty blood collection and haemolysis affect the test.
The use of siliconized or plastic test tubes instead of the recommended
borosilicate glass tubes for the test, length and bore of glass tubes, angle and
speed of the tilting are amongst the variables which affect the PT when
performed manually.
ECATSTUDY
The universal use of all centres of the same reagent in the ECAT study overcame
some of the problems of PT standardization. A single recommended reagent
and technique was therefore selected by the Executive Committee to be employed
at all centres with a manual tilt-tube technique. The manual method was recom-
mended because coagulometers accelerate the test to varying degrees and may
also affect the lSI.
   The Manchester Reagent thromboplastin was selected for the ECAT study
because it has a low lSI (1.1 approximately), shows good responsiveness to the
individual extrinsic clotting factors and little inter-batch variation. It has been
shown to detect the acceleration of extrinsic clotting which occurs in the days
following major sUf§ery36. It shows good responsiveness to depression of factor
VII in liver disease 7. The precision of testing with Manchester Reagent in
national external quality assessment has been shown to be greater than with
some other reagents38 .
TECHNIQUE
A standardized manual technique was provided therefore for the ECAT study
as follows:
APPENDIX
Recommended protocol for thromboplastin calibration
1. Blood samples from healthy individuals and patients stabilized on coumarin
   treatment are required for the conventional fresh plasma thromboplastin
   calibration. The total number of plasmas recommended is 20 normals and
   60 samples from coumarin treated patients. Testing is performed on separate
   days but these need not be consecutive.
   A recommended plan for a working day is as follows:
   PT tests on:
      Eight fresh plasmas from long-term-stabilized coumarin patients (single
      tests).
      Two fresh plasmas from healthy subjects (duplicate tests).
      One lyophilized plasma from a healthy subject (duplicate test, tested on
      one day only).
   The following reagents are to be tested: (a) IRP, (b) local thromboplastin.
   Number of tests per day: 50 (approx).
   Number of test days: eight (approx).
   As a guide, the time schedule of a single exercise could be as follows:
   1.1. Collection of blood samples. Blood drawn from healthy subjects and
        from patients stabilized on oral anticoagulants should be centrifuged
        immediately after collection and the plasma transferred to a non-wettable,
        stoppered container. Maintain at room temperature until tested.
   1.2. Testing of the plasma samples with the thromboplastins is described in
        section 6.
                                        56
                              PROTHROMBIN TIME
  Notes:
  (a) Single tests only should be performed on the samples from coumarin
      patients.
  (b) Tests on each plasma should be performed with each thromboplastin
      in turn before proceeding to the next plasma.
2. Selection of healthy subjects and patients
   2.1. Normals. These samples should be obtained from healthy adults of
        both sexes with a reasonable age spread. Select different normal subjects
        on each day of testing. Test each sample in duplicate.
   2.2. Plasmas from coumarin-treated patients. The patients should have been
        on oral anticoagulants for at least 6 weeks and have a result within the
        therapeutic range. Participant centres should include patient samples
        displaying a variety of levels of anticoagulation within the therapeutic
        range. Perform single tests only.
3. Sample collection
   3.1. The blood should be collected by clean venipuncture. One part of sterile
        trisodium citrate 0.109 moVL for nine parts of sample is recommended.
        If an evacuated tube is employed it should be siliconized (see section 5.2).
   3.2. The blood shall be centrifuged immediately after collection (approx 800g)
        for 5 min, at room temperature.
   3.3. The plasma shall be transferred by siliconized or plastic pipette to a
        stoppered, non-wettable container.
   3.4. The stoppered plasma container is to be kept at room temperature until
        testing.
4. Reconstitution of thromboplastins
   International Reforence Preparation for Thromboplastin (IRP). Extreme care
   should be taken to avoid wastage of IRP. Reconstitute each ampoule of
   IRP with requisite volume of reconstitution fluid. Leave thromboplastin in
   ampoule (or vial). Keep at room temperature. Ensure that the thromboplastin
   is completely resuspended before use. Gentle tapping on the side of the
   ampoule with a finger will facilitate resuspension. It is not necessary or
   desirable to shake the suspension vigorously.
5. Other reagents and equipment
   5.1. Calcium chloride 0.025 moVL; for recalcification of plasma/thrombo-
        plastin mixture. Store at 4 0c. Use fresh aliquots each day.
   5.2. Equipment. Syringes and evacuated tubes must be silicone-coated or
        made of good-quality plastic material to prevent contact activation of
        coagulation factors.
          Plastic or siliconized pipettes for transfer of plasma into glass test
        tubes at the time of testing.
          Use non-siliconized disposable glass test tubes for the actual testing.
          Water bath at 37 °C (tolerance limits: 37.0 °C ± 0.2 0C). Use calibrated
        thermometer.
          New pipetting tips must be used for each test.
          Accurate stopwatch.
                                       57
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
6. Testing procedure
   6.1. The WHO recommended procedure for calibration of thromboplastin
        is based on the manual tilt-tube (or hook) technique. This method must
        be used for all calibrations.
   6.2. The sequence of thromboplastin testing must be alternated on each day
        of testing. This sequence should be retained for the first day of testing
        for all patients' and normal plasmas. On the second day this sequence
        of testing should be changed.
           The exercise should take no more than 10 days to complete with, e.g.,
        two normals and six patients on each day. If it can be completed in a
        shorter period this is acceptable as long as the order of testing is changed.
        A specimen work plan is given below.
             I Fresh normal 1 I                                        I
                Coumarin 1
                Coumarin 2
                Coumarin 3
                Coumarin 4
                Coumarin 5
                Coumarin 6
             I Fresh normal 2 I
       Coumarin    =plasma from oral anticoagulated treated patient)
        Note: Test all 'normal' plasmas in duplicate but perform single tests
                only on coumarin plasma-treated samples. The sequence should
                be alternated on each day of testing.
   6.3. The coagulation end-point should be determined by the recommended
        tilt-tube technique. Test tubes should be kept under water at 37 °C
        as much as possible in order to maintain optimal temperature. Proceed
        as follows: pre-warm glass test tubes in the water bath prior to testing.
                                        58
                                PROTHROMBIN TIME
       The testing procedure for rabbit and human plain IRP is as follows:
       (a) transfer 0.1 ml thromboplastin to a test tube;
       (b) incubate for 2 min;
       (c) add 0.1 ml plasma;
       (d) mix gently with the thromboplastin;
       (e) wait 1 min for incubation to reach the optimal reaction tem-
            perature;
       (f) recalcify with 0.1 ml pre-warmed calcium chloride 0.025 moUL;
       (g) tilt test-tube manually three times in 5 seconds through an angle
            of 90°; record clotting time (seconds).
       For bovine IRP the volumes are different.
7. Prothrombin times must be recorded to one decimal place and must not be
   rounded to the nearest second. For example if a PT of 34.1 s is observed on
   the stopwatch this time must be recorded; the time should not be rounded
   to 34 s.
lSI calculation
The prothrombin times of all plasma specimens (20 healthy subjects and 60
patients) are converted to the corresponding logarithms. Let z be the logarithm
of a prothrombin timels determined with the IRP and x be the logarithm of
prothrombin timels determined with the local reagent to be calibrated. The
relationship z = al + blx is calculated by the following formulae, in which al
and b l are the orthogonal regression line parameters representing the intercept
and the slope respectively.
with
and al   =z-blx;
x is the arithmetic mean of x and z the mean of z; Sx and Sz are the standard
deviations of the x and z values and r the correlation coefficient.
   This orthogonal regression line estimates the relation between log prothrombin
timels of IRP and local thromboplastin. With the certified parameters a and
b of the calibration line of the IRP against WHO preparation 67/40, the intercept
and slope of the local reagent in relation to WHO first primary IRP 67/40 are
as follows:
                   intercept:    anew =a - b . al
                   slope:        blocal = lSI =b . bl
where a and b are the certified parameters and al and bl are the newly determined
local parameters.
                                        59
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
  To transform the logarithmic value into the value that would have been
obtained with the first WHO IRP human combined (67/40) the following
equation is used:
                                PT67140   =antilog(a + b . x)
where a and b are the certified parameters and x is the log PT.
References
 1. Quick 1. The prothrombin time in haemophilia and in obstructive jaundice. J Bioi Chern 1935;
    109: 73-4.
 2. Owren PA, Aas K. The control of dicoumarol therapy and the quantitative determination of
    prothrombin and proconvertin. Scand J Clin Lab Invest 1951; 3: 201-8.
 3. Owren PA. Thrombotest. A new method for controlling anticoagulant therapy. Lancet 1959;
    2: 754-8.
 4. Broze GJ, Leykam JE, Schwartz BD, Miletich JP. Purification of human brain tissue. J Bioi
    Chern 1985; 260: 10917-20.
 5. Guha A, Bach R, Konigsberg W, Nemerson Y. Affinity purification of human tissue factor:
    interaction of factor VII and tissue factor in detergent micelles. Proc Nat! Acad Sci USA
    1986;83:299-302.
 6. Morrissey JH, Fair DS, Edgington TS. Structure and properties of the human tissue factor
    apoprotein. Thromb Haemostas 1987; 58: 257.
 7. Fujikawa K, Suzuki K. Cellular coagulant and anticoagulant proteins. In: Poller L, ed. Recent
    advances in blood coagulation 5. Edinburgh: Churchill Livingstone, 1991; 35-51.
 8. Tripodi A, Arbini, A, Chantarangkul V, Mannucci PM. Recombinant tissue factor as substitute
    for conventional thromboplastin in the prothrombin time test. Thromb Haemostas 1992; 67:
    42-5.
 9. European Concerted Action on Anticoagulation (ECAA). Poller L, van den Besselaar AMHP,
    Jespersen J, Tripodi A, Houghton D. The ECAA field studies of coagulometer effects on the
    lSI of ECAA thromboplastins. Thromb Haemostas. 1998; in press.
10. Kirkwood TBL. Calibration of reference thromboplastins and standardisation of the
    prothrombin time ratio. Thromb Haemostas 1983; 49: 238-44.
11. Hermans J, van den Besselaar AMHP, Loeliger EA, van der Velde EA. A collaborative
    calibration study of reference materials for thromboplastins. Thromb Haemostas 1983; 50:
    712-17.
12. Thomson, JM, Tomenson JA, Poller L. The calibration of the second primary international
    reference preparation for thromboplastin (thromboplastin, human, plain, coded BCT/253).
    Thromb Haemostas 1984; 52: 336-42.
13. Poller, L, Taberner D. Dosage and control of oral anticoagulants: an international collaborative
    survey. Br J Haematol1982; 51: 479-85.
14. WHO Expert Committee on Biological Standardisation, 28th Report. WHO Technical Report
    Series. 1977; 610: 14-15,45-51.
15. WHO Expert Committee on Biological Standardisation, 33rd Report. WHO Technical Report
    Series. 1983; 687: 81-105.
16. ICTHlICSH. Prothrombin time standardisation: report of the expert panel on oral anticoagulant
    control. Thromb Haemostas 1979; 42: 1073-114.
17. Kendall MG, Stuart A. The advanced theory of statistics, 2. London: Charles Griffin, 1961.
18. Loeliger EA, Lewis SM. Progress in laboratory control of anticoagulants. Lancet 1982; 2:
    318-20.
19. van den Besselaar AMHP. Multicentre replacement of the International Reference Preparation
    rabbit plain. Thromb Haemostas 1993; 70: 794-9.
20. van den Besselaar AMHP, van der Velde EA. The manufacturer's calibration study. In: van
    den Besselaar AMHP, Gralnick HR, Lewis SM, eds. Thromboplastin calibration and oral
    anticoagulant control. The Hague: Martinus Nijhoff, 1984; 127-49.
21. Thomson JM, Darby KV, Poller L. Calibration of BCT/441, the ICSH Reference Preparation
    for Thromboplastin. Thromb Haemostas 1986; 55: 379-82.
                                               60
                                    PROTHROMBIN TIME
22. Tomenson IA. A statistician's independent evaluation. In: van den Besselaar AMHP, Gra1nick
    HR, Lewis SM, eds. Thromboplastin calibration and oral anticoagulant control. The Hague:
    Martinus Nijhoff, 1984; 87-108.
23. Hirsh I, Dalen IE, Deykin D, Poller L. Oral anticoagulants: mechanism of action, clinical
    effectiveness and optimal therapeutic range. Chest 1992; 102: 312-26S.
24. Altman DG, Bland 1M. Measurement in medicine: the analysis of method comparison studies.
    Statistician 1983; 32: 307-17.
25. D'Angelo A, Seveso MP, D'Angelo SV, Gilardoni F, Macagni A, Manotti C. Comparison of
    two automated coagulometers and the manual tilt tube method for the determination of
    prothrombin time. Am I Clin Patho11989; 92: 321--8.
26. Poggio M, van den Besselaar AMHP, van der Velde EA, Bertina RM. The effect of some
    instruments for prothrombin time testing on the international sensitivity index (lSI) of two
    rabbit tissue thromboplastin reagents. Thromb Haemostas 1989; 62: 868-74.
27. Ray MJ, Smith IR. The dependence of the International Sensitivity Index on the caogulometer
    used to perform the prothrombin time. Thromb Haemostas 1990; 63: 424-9.
28. Chantarangkul V, Tripodi A, Mannucci PM. The effect of instrumentation on thromboplastin
    calibration. Thromb Haemostas 1992; 67: 588-9.
29. Clarke K, Tabemer DA, Thomson 1M, Morris lA, Poller L. Assessment of value of calibrated
    lyophilized plasmas to determine International Sensitivity Index for coagulometers. I Clin
    Patho11992; 45: 58--60.
30. Poller L, Triplett DA, Hirsh I, Carroll I, Clarke K. The value of plasma calibrants in correcting
    coagulometer effects on International Normalized Ratios. Am I Clin Patho11995; 103: 358--65.
31. Houboyan IL, Goguel AF. Procedure of reference calibrated plasmas for prothrombin time
    standardisation. (Abstract, XIVth Congress ISTH, New York) Thromb Haemostas 1993; 69:
    663.
32. van den Besselaar AMHP. The use of lyophilized calibration plasmas and control blood for
    INR calculation on external quality control of the prothrombin time. Am I Clin Pathol 1994;
    102: 123-7.
33. European Concerted Action on Anticoagulation. Poller L, van den Besse1aar AMHP, Iespersen
    J, Tripodi A, Houghton D. Comparison of artificially deleted, lyophilized coumarin and fresh
    coumarin plasmas in thromboplastin calibration. Br I Haematol. 1998; 101: 462-7.
34. European Concerted Action on Anticoagulation. Poller L, Barrowcliffe Tw, van den Besselaar
    AMHP, Jespersen I, Tripodi A, Houghton D. Minimum lyophilized plasma requirement for
    lSI calibration. Am J Clin Pathol, (In press).
35. Dalen JE, Hirsh 1. ACCP 4th Consensus Conference Introduction. Chest 1995; 108: 225-65.
36. Poller L, McKernan A, Thomson 1M, Elstein M, Hirsch PI, Iones ffi. Fixed minidose warfarin:
    a new approach to prophylaxis against venous thrombosis after major surgery, Br Med J 1987;
    295: 1309-12.
37. Kitchen S, Malia RG, Greaves M, Preston FE. Comparison of human and rabbit brain
    thromboplastin in evaluation of haemostatic defect of liver disease. Lancet 1986; 2: 1463.
38. Taberner DA, Poller L, Thomson JM, Darby KY. Effect of international sensitivity index
    (lSI) of thromboplastins on precision of international normalised ratios (INR). J Clin Pathol
    1989; 42: 92-100.
                                                61
7
Endogenous thrombin potential
H. C. HEMKER and S. BEGUIN
INTRODUCTION
The one and only function of the clotting system is to generate thrombin in
the amounts and at the site required for haemostasis. The endogenous thrombin
potential (ETP), is a parameter that reflects this function quantitativelyl,2. The
combined action of the procoagulant and anticoagulant systems in the :'lood
is reflected in the course of thrombin concentration in a sample of blood after
the clotting system is triggered, i.e. in the thrombin-generation curve (TGC;
Figure 7.1). Such curves are characterized by an optional lag phase, a rise to
a peak value and a decline to zero in the course of approximately 10 min.
    It is the function of the enzyme thrombin to convert substrates. If a thrombin
substrate is present at a constant concentration during the whole of the TGC,
then its velocity of conversion will be proportional to the concentration of
thrombin. The course of the resulting product will therefore be represented by
a curve, the slope of which represents the reaction velocity and is proportional
to the level of thrombin (Figure 7.2, thin dotted line). No more product will
100
                       l~~. ".1
                      i'
                       .s
                                  o    5          10        15         20
                                              Time (min)
Figure 7.1   Thrombin generation curves (TGC) as obtained by the subsampling method
From left to right: PPP-Extrinsic system; PPP-Intrinsic system; PRP. The area under the curve is
defined as the endogenous thrombin potential (ETP)
                                             63
             LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                             150,---------------,
o 5 10 15 20 25
Time (min)
SUBSAMPLING METHODS
Materials
Buffers
Buffer A: 20 mmollL Tris-HCI, 100 mmollL NaC!, 0.5 giL bovine serum
albumin pH 7.35.
Buffer B: same as buffer A with 20 mmollL EDTA, pH 7.9.
Stopping fluids
For plasma: I mollL citric acid or acetic acid.
For blood: 10 J.1IllollL PPACK in physiological salt solution.
                                       65
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Triggers
Any tissue factor preparation can be used as the trigger for the extrinsic system.
A dilution that in normal plasma gives a clotting time of around 36 s will yield
a suitable Tac. As long as the thromboplastin clots normal plasma in < 120 S,
the ETP is independent of the tissue factor concentration. This corresponds
to IlJlIloVL of procoagulant phospholipid (pSIPC 1/4 weight %) and> 5 pmoVL
of tissue factor. Most commercial tissue factor preparations contain Ca2 +,
which should be taken into account when calculating the final Ca2+ concentration.
Usually polybrene is also added by the manufacturer, so effects of heparin may
go unobserved when using commercial thromboplastins. As a rule we use relipidated
recombinant human tissue factor (Innovin, Dade).
   Optically clear APTT reagents, i.e. contact activating solutions containing
phospholipids, can be used as a trigger for the intrinsic system. We used Actin
FS® (Dade), a suspension of ellagic acid and soybean phosphatides.
   As procoagulant phospholipid we use a suspension of phosphatidylserine
(PS) and phosphatidylcholine (PC) in 1:4 proportion at a concentration of
20 IlmoVL. The final concentration in the reaction mixture is 1 IlmoVL.
Substrates
The chromogenic substrate used for measuring thrombin in the subsampling
assayisS2238:H-D-Phe-Pip-Arg-pNA.2HCl.Theleaving-groupisparanitro-
aniline (pNA), the specific absorbance of which is 9.93 mOD per IlmollL
per cm light-path at the measuring wavelength of 405 nm. Because of the
high concentration of thrombin relative to the other proteases of the clotting
system (> 20:1), the specificity of the substrate for thrombin is a minor
concern, and cheaper and less specific substrates such as S2165 may be used
with good results.
Plasma preparation
Blood
Blood is collected on 0.13 moVL trisodium citrate; nine parts of blood to one
part of citrate solution. An open (non-vacuum) venipuncture is preferable, in
order to avoid platelet activation. For experiments in PRP an open system is
obligatory. The first millilitre of blood is to be discarded. For the continuous
method, blood is preferentially taken from fasting subjects; a recent meal may
cause marked turbidity impairing the spectrophotometric measurement.
PRP
Samples are centrifuged at 250g, at a temperature not lower than 15°C for 10
min. The supernatant PRP is pipetted off and the platelets are counted. In the
                                      66
                    ENDOGENOUS THROMBIN POTENTIAL
ppp
PPP is obtained as the supernatant from blood centrifuged at 1000g, at a
temperature not lower than 15°C for 15 min. This plasma is centrifuged a
second time at the same speed and the pellet is discarded. If a pool of normal
plasmas is made (at least 10 donors), the pooled plasma is centrifuged at 4 °C
for 1 hat 23 OOOg. All plasma, if not used immediately, is stored at -80°C.
An aliquot of 240 III of PPP is incubated with 40 III of buffer A (that may
contain substances to be tested) and 20 III of 20 IlmollL procoagulant
phospholipid, until temperature equilibration. Coagulation is initiated at t     =
oby addition of 60 III of CaCl2 0.1 mollL (16.7 mmollL final concentration)
containing tissue factor. Samples (10 Ill) of the reaction mixture are taken at
suitable intervals and added to prewarmed (37°C) cuvettes containing 490 III
of 200 IlmollL S2238 in buffer B. The reaction is stopped by the addition of
300 III of 1 mollL citric acid (or acetic acid) after 2 min and the optical density
(OD) is measured at 405 nm.
   The sampling interval is dependent upon the velocity of thrombin generation.
In human material the usual sampling frequency is two per minute. Sampling
at 5-10 s intervals is readily possible, so up to five experiments can be carried
out in parallel. In laboratory animals (mouse, rat, rabbit, pig) thrombin
generation may be much faster than in humans and the sampling frequency
must be increased in accordance. An evolving clot is wound out on a plastic
spatula and removed.
   The method can be adapted to microtitre plates if the volumes are propor-
tionally reduced. It is essential, however, that the dilution upon subsampling
is > 20 times, otherwise the interaction between thrombin and antithrombin
will continue at a rate high enough to influence the readings.
PRP
An aliquot of 240 III of PRP is incubated with 60 III of buffer A (that may
contain substances to be tested) until temperature equilibration. Coagulation
                =
is initiated at t 0 by adding 60 III 0.1 mmoVL CaCI2 • A low concentration of
recombinant tissue factor (1 % of the concentration used in the extrinsic system)
may be added to the CaCl2 solution in order to rule out variability caused by
the traces of thromboplastin that may be present in the PRP. This concentration
of tissue factor should give clotting times of around 10 min when added together
with Ca2 + to non-defibrinated PPP. In PRP, all experiments need to be carried
out within 60 min of venipuncture and only experiments carried out in parallel
can be directly compared, as thrombin generation is influenced by platelet
ageing.
Blood
It is possible to execute a thrombin-generation test on non-anticoagulated
blood. If the blood is obtained from an ideal venipuncture and collected in a
plastic tube a time lapse of minimally 7 min will be available before thrombin-
generation starts. Normally, however, 480 III of citrated blood is incubated with
                                                 =
120 III of buffer A. Coagulation is initiated at t 0 by adding 120 III 0.05 moVL
CaCl2 with or without recombinant tissue factor as in PRP. With blood,
subsamples of 20 III are usually taken. The stopping fluid is 10 llffioVL PPACK
in an isotonic salt solution. After stopping the tubes are centrifuged (5 min at
1000g) and pNA is determined photometrically in the clear supernatant. In
the course of the clotting process a 10% change in the haematocrit occurs. If
optimal precision is required this can be compensated for by hydrolysing the
sedimented erythrocytes in each tube and calculating the change in erythrocyte
content from the haemoglobin measured spectrophotometrically.
Equipment
A water bath at 37°C is used to contain both the plasma and the tubes with
chromogenic substrate. If experiments with whole blood are carried out it is
preferable to fill the bath with isotonic salt solution, so as to prevent haemolysis
if a drop of bath fluid should accidentally fall into an erythrocyte-containing
tube. In order to avoid the need for accurate manual timed sampling, and the
errors that it may cause, we equipped both the sampling and the stopping
pipette with a pushbutton that is connected to a personal computer, and that
records sampling and stopping times. We established a program in which the
spectrophotometer is also connected to this computer, so that the increase of
OD over time in the cuvette is calculated from the OD of each tube and the
incubation time. Amidolytic activities, expressed as the equivalent amount of
a-thrombin, are calculated via a calibration factor obtained with active site
titrated human a-thrombin.
                                       68
                   ENDOGENOUS THROMBIN POTENTIAL
A B C D E F
The sampling times are entered in column B and the corresponding amidolytic activities are
entered in column C. In column E the amount of u2M-thrombin is calculated and in column D
the amount of free thrombin, using an assumed value of k (in cell $F$l). In cell F2 the free
thrombin concentrations, calculated in column D, are summed over a range of sampling points
(here 25:30) where free thrombin can be assumed to have reached the zero level. The correct
value of k (Fl) is found by changing it (Le. $F$l) until F2 is as close as possible to zero. The
ETP is obtained as the sum (D3 ... D32) multiplied by the sampling interval in min ( here 0.5 )
150 y-----------------,
                      ~ 100
                      .s
                      c:
                      :c
                      E
                       2
                      ('3.   50
                                          5            10          15           20
                                                  Time (min)
Figure 7.3       Dissection of a thrombin generation curve as obtained in the subsampling method
Circles: Experimental amidolytic activity; Bold line: Course of free thrombin; Thin line: Course
of (12 -macroglobulin-thrombin complex
                                                  70
                   ENDOGENOUS THROMBIN POTENTIAL
CONTINUOUS, SPECTROPHOTOMETRIC METHOD
Principle
The concept behind ETP determination is simple. Thrombin being an enzyme,
its function is quantified by the amount of substrate that it converts. To a
volume of defibrinated PPP we add an artificial substrate of thrombin. Then
the clotting system is triggered and the amount of substrate that is converted
by thrombin during its transient presence in the sample is continuously moni-
tored. The concentration and the kinetic properties of the substrate are chosen
such that during the rise and fall of the thrombin concentration the velocity
of product (= colour) formation is always practically proportional to the
concentration of thrombin. In this way the amount of product formed is
proportional to the surface under the TGC, i.e. to the ETJi4, which is a direct
indicator of the amount of natural substrate that thrombin can convert when
it is generated in a volume of clotting blood in vivo.
Substrates
For the continuous method substrates are required that are converted by
thrombin at a suitable rate and that are not readily converted by abundant
proteases such as kallikrein. Those that are split by factor Xa with reaction
velocities similar to those of thrombin (Table 7.3) can be used because the peak
concentration of factor Xa in clotting plasma is < 5% of that of thrombin. Yet
these substrates should not bind with high affinity to factor Xa because that
would make them efficient factor Xa inhibitors. We used commercially available
Malonyl-a-aminoisobutyryl-arginine para-nitroanilide methyl ester hydro-
chloride (CH 3 0-CO-CH z-CO-Aib-Arg-pNA . HCI; SQ 68) from Serbio
Laboratories, France (European Patent 88400304.7). Other suitable substrates
were synthesized in our laboratory (Table 7.3), using a procedure described in
detail elsewhere10-1Z. The chromogenic leaving-group is always para-nitroaniline,
the specific absorbance of which is 9.93 mOD per lJllloVL per cm light-path
at the measuring wavelength of 405 nm. We determined conversion factors
between the 00 end-level and the ETP in nmoVL x min with each of the
substrates 8 (Table 7.4).
   Until 10% of the substrate is converted by free thrombin a linear relation
between ETP and 00 end-level can be assumed. The permitted upper limit of
product is therefore 50 lJllloVL, which sets an upper limit of 500 mOD to the
00 end-level. Usually an increase of 10 mOD is the lowest that can be reliably
measured against the yellow background of plasma. Depending upon the
kinetic properties of a substrate the range of ETP values that can be measured
                                      71
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
in the OD window of 10-500 mOD is different (Table 7.4). Substrates 1-5 are
suitable for measurements in a wide range around the normal level, whereas
substrates 6 and 7 can be used to assess with high precision the effect of
inhibitors (e.g. activated Protein C (APC), heparin) or of anticoagulant therapy,
but ETP that are higher than normal will be underestimated.
Caution! Considerable errors can be introduced if substrates are used that have
not been selected and characterized according to the principles outlined here.
Defibrination
Defibrination is required to avoid disturbance of the OD recording by the
turbidity of emerging fibrin. If fibrin polymerization can be avoided in a way
that does not interfere with thrombin generation, defibrination is no longer
required. For defibrination we use snake venom enzymes (the only action of
which, in the coagulation system, is on fibrinogen), such as Reptilase and
Ancrod. A Reptilase solution (Boehringer Mannheim, Germany) is prepared
so that 20 ~, added to 1 m1 of plasma, causes a clot in 4 min. Higher concentra-
tions should be avoided because they may activate factors V and/or VIII and
thereby increase the thrombin potential.
    Ancrod, the fibrinogen clotting enzyme of the Malayan pit viper, was obtained
as the commercial preparation Arvin® (Knoll AG, Ludwigshafen, Germany).
It is used at a final concentration of 1 Ulml, causing a clotting time of around
3 min in normal plasma. It does not activate clotting factors or platelets. It
also does not cause haemolysis and therefore can be added directly to the tube
in w4ich blood is taken. In this way defibrinated plasma can be obtained directly
upon centrifugation of the blood.
    Defibrination is carried out as follows: plasma samples are mixed with 1:50
volume of the Reptilase or Ancrod solution and incubated for 10 min at 37°C,
then kept on ice for 10 min. The fibrin is wound out on a small plastic spatula.
Unlike Reptilase, Ancrod does not cause haemolysis and can be added to whole
blood, rendering defibrinated plasma directly upon centrifugation. In large
series of samples, defibrination is the rate-limiting step of the procedure.
Measurement
The reactions can be carried out in any laboratory automaton capable of
measuring with sufficient precision the course of the optical density at 405 nm
at 30 s intervals during 12-15 min. We used the Cobas Bio and Cobas Fara
centrifugal analysers (Hoffmann-La Roche; a listing of the required settings
of the instruments is available from the authors).
   The reaction mixture consists of four parts of defibrinated plasma, that are
mixed with one part of (partial) thromboplastin solution and one part of start
solution. In the Cobas machines the volumes are 80 ~ of defibrinated plasma
(or 75 ~ defibrinated plasma to which may be added 5,.11 of a substance to be
tested) and 20 ~ of the thromboplastin, without Ca2+. Mter 30 s of incubation,
                                      72
                       ENDOGENOUS THROMBIN POTENTIAL
Data handling
The centrifugal analyser is connected to a personal computer via an installed
option that makes the results available on a communication line following the
RS-232 standard. The data are in the form of a succession of ASCII characters
representing the series of OD readings, the time of the reading is given by the
position in the list, knowing the sampling interval. The data are used as input
for a program (Pascal or Basic), using a function for character extraction from
the RS-232 interface buffer of the personal computer and transformed to a
standard format file which gives the OD as a function of time and from which
the ETP can be calculated off-line. Programs for reception-recording on
IBM-compatible PCs are available from the authors.
                                          5                  10            15
                                                Time (min)
                                              73
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
be applied to such progress curves (Table 7.2). Only the criterion for a zero
end-level of thrombin differs. If thrombin attains the zero level, the amount
of product formed by thrombin will be constant. To test this, column F
(Table 7.2) is added and Fl is changed until cell F2 is as close as possible to
zero. This can again be done by trial and error or via a spreadsheet tool. The
fixed end-level of product formed by free thrombin is proportional to the ETP.
This calculation renders the amount of substrate converted by free thrombin.
   Theoretically the relation between the area under the TGC (i.e. the ETP)
and the total amount of substrate converted is given by ETP A . Kmlkcat •    =
S5. Application of this formula requires exact knowledge of Km and kcat of
the substrate in plasma. These are difficult to obtain due to the instability of
thrombin in the presence of natural inhibitors. Data such as those in Table 7.3
are tentative values, only suited to obtain a global impression of the reaction
behaviour that is to be expected. We therefore determined the proportionality
between the 00 end-level and the ETP by assessing the ETP independently
from a TGC obtained by subsampling. This procedure was used to obtain the
conversion factors in Table 7.4. In actual practice it is not necessary to obtain
the absolute value of the ETP as long as it is possible to compare the results
of the test plasma with those of a standard normal (pool) plasma tested in
parallel, and expressed as a percentage of normal. A home-made normal
plasma pool should be used for this purpose as long as we do not have proof
that commercial control plasmas can be used.
Reference values
Under our experimental conditions, plasmas from normal donors are found
                                                          =
to give an 00 end-level of 54.8 ± 8.1 mOD (n 118) from SQ68 and 108.1 ±
Table 7.2 Calculation of the ETP from an optical density curve (continuous method).
A B C D E F
The sampling times are entered in column B and the corresponding optical densities are entered
in column C. In column E the amount of product due to Ct2M-thrombin activity is calculated
and in column D the amount of product due to the activity of free thrombin, using an assumed
value of k (in cell $F$I). In column F, from line 10 on, the differences between cells in column
E are entered (i.e. the first derivative of the function in column E). In cell F2 the derivative
calculated in column F are summed over a range of sampling points (here 25:30) where free
thrombin can be assumed to have reached the zero level. The correct value of k (Fl) is found by
changing it (i.e. $F$I) until F2 is as close as possible to zero. The ETP is proportional to the
end level in column D. The proportionality constant is dependent upon the substrate used
                                             74
                        ENDOGENOUS THROMBIN POTENTIAL
Table 7.3 Kinetic constants of chromogenic substrates used for measuring the ETP.
RV: Reaction velocity at 500 jllIloVL substrate concentration, relative to thrombin action on
MZ-Aib-Arg-pNA (8Q68); MZ: malonic acid monomethyl ester; DEMZ: diethyl malonic acid
monomethyl ester; Msc: 2-(methylsulphonyl)ethyloxycarbonyl; Boc: tert-butyl oxycarbonyl; *)
substrate 2 is not hydrolysed by factor Xa, the K.n mentioned is the binding constant measured
from competitive inhibition of 8Q 68. Note that some of these values differ from those
reported earlier because they represent results obtained with more advanced methods (R.
Wagenvoord, work in progress)
                    =
16.8 mOD (n 84) from Msc-Val-Arg-pNA. With the conversion constants
of Table 7.4, this represents an endogenous thrombin potential of
384.8 ± 51.7nmollL x min and 394.3 ± 62.3 nmoVL x min, respectively. The
normal value of the ETP in the intrinsic system appears not significantly higher:
414 ± 41 nmoVL x min.
Table 7.4 Relation between OD signal and ETP with different chromogenic substrates.
The third column gives the end-level of the thrombin dependent OD-signal with a normal pool
plasma (mean ± 8EM; n = 24 or more); the fourth column gives the % of the initial amount of
substrate (500 jllIlollL) that is converted in this process; the conversion factor is the multiplier
to calculate the ETP from the final OD level; the range gives the ETP values (% of normal) that
can be reliably measured with that substrate.
For substrates 7 and 8, the figures for the normal values have been calculated via the catalytic
efficiencies relative to 8Q688
                                                 75
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
    The within-run imprecision of the determination, i.e. the standard error of
a single value calculated from 20-24 identical samples determined in the same
run on a Cobas instrument, is about 4% (extrinsic 3.5%, intrinsic 4.4% (n =
22». The between-run imprecisions, obtained when the same normal plasma
is tested in different runs on different days, is on the mean 1% higher (extrinsic
                               =
system 4.7% intrinsic 5.6% (n 58».
    Hepes buffer and Tris buffer give the same normal ETP value in the extrinsic
system (403.8 nmoVL x min ± 12.4 versus 398.4 nmoVL x min ± 14.1; n 17),   =
in the intrinsic system Hepes buffer may give slightly lower values than Tris
buffer (399.0 ± 10.5 nmoVL x min versus 435.2 nmoVL x min ± 16.6; n 17).   =
    In normal PRP, measured under our conditions, the normal ETP is 411 ±
53 nmoVL x min (mean ± SEM, n 28).   =
Execution
The execution of the experiment is exceedingly simple. The thrombin-like
activity is determined, as in the samples from a manual thrombin-generation
experiment (see above) but only in one sample, after 15-20 min of incubation.
                                      76
                      ENDOGENOUS THROMBIN POTENTIAL
The ratio is determined between the activity in the sample after addition or
administration of the modifier and the blank or zero time plasma.
Pathophysiological aspects
Hyper- or hypocoagulability can be brought about by a large number of different
pathological conditions, and there exists a large spectrum of laboratory tests
to locate the underlying cause. To our knowledge, however, there is not yet a
single laboratory parameter that increases in all forms of hypercoagulability
and similarly decreases in all forms of hypocoagulability. Such an assay would
be useful for the detection of plasma-based thrombotic tendency and for the
surveillance of drug-induced hypocoagulability.
   We think that the ETP is a good candidate as a general indicator of
coagulability l-6. The ETP is decreased to 15% and 35% of normal by oral
anticoagulation (international normalized ratio 2.5--4.0) as well as by heparin
administration (APTT 1.5-2.5 x control)s. Contrary to the classical tests, the
ETP also renders the effect of mixed oral anticoagulant and heparin treat-
ments. Preliminary results indicate that the effect of dermatan sulphate and
direct irreversible thrombin inhibitors (hirudin) is also truthfully rendered by
the ETP.
   The ETP is increased in untreated subjects with congenital antithrombin
deficiency8, in persons with the prothrombin Leiden mutation (p. Kyrie, personal
communication), in persons with the factor V Leiden mutation 16 , and in women
using oral contraceptives 14 • In deep-vein thrombosis (phlebographically con-
firmed), it is increased by 29.4% (extrinsic) and 53% (intrinsic)s. In (angiographi-
cally assessed) coronary artery disease the increase is 10% and 17% respectivelys.
   The APe system is only partly active in isolated plasma because
thrombomodulin (TM) is linked to the vessel wall. TM or APe can be added
to the ETP assay to reveal APe resistance 13- 16 . The ratio of the ETP plus and
minus TM or APe is a sensitive indicator for disturbances of the APe system.
References
 1. Hemker HC. Thrombin generation, an essential step in haemostasis and thrombosis. In:
    Bloom AL, Forbes CD, Thomas DP, Tuddenham EGD, eds. Haemostasis and thrombosis.
    Edinburgh: Churchill Livingstone, 1993; 477-90.
 2. Hemker HC, Beguin S. Thrombin generation in plasma: its assessment via the endogenous
    thrombin potential. Thromb Haemostas 1995; 74: 134--8.
 3. Biggs R, Macfarane RG. Human blood coagulation and its disorders. Oxford: Blackwell
    Scientific Publications, 1967; 67.
 4. Hemker HC, Wielders SJH, Kessels H, Beguin S. Continuous registration of thrombin
    generation in plasma, its use for the determination of the thrombin potential. Thromb
    Haemostas 1993; 70: 617-24.
 5. Hemker HC, Willems GM, Beguin S. A computer assisted method to obtain the prothrombin
    activation velocity in whole plasma independent of thrombin decay processes. Thromb
    Haemostas 1986; 56: 9-17.
 6. Beguin S, Lindhout T, Hemker HC. The effect of trace amounts of tissue factor on thrombin
    generation in platelet rich plasma, its inhibition by heparin. Thromb Haemostas 1989; 61:
    25-9.
                                           77
           LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
 7. Kessels H, Beguin S, Andree H, Hemker HC. Measurement of thrombin generation in whole
    blood - the effect of heparin and aspirin. Thromb Haemostas 1994; 72: 78-83.
 8. Wielders S, MukheIjee M, Michiels J, Rijkers DT, Cambus JP, Knebel RW, Kakkar V, Hemker
    HC, Beguin S. The routine determination of the endogenous thrombin potential, first results
    in different forms of hyper and hypocoagulability. Thromb Haemostas 1997; 77: 629-36.
 9. Kessels H, Willems G, Hemker HC. Analysis of thrombin generation in plasma. Comput
    BioI Med 1994; 24: 277-88.
10. Rijkers DTS, Adams HPHM, Hemker HC, Tesser G. A convenient synthesis of amino acid
    p-nitroanilides, synthons in the synthesis of protease substrates. Tetrahedron 1995; 51: 11235--50.
11. Rijkers DTS, Wielders SJH, Tesser GJ, Hemker He. Design and synthesis of thrombin
    substrates with modified kinetic parameters. Thromb Res 1995; 79: 491-9.
12. Rijkers DT, Hemker HC, Tesser GJ. Synthesis of peptide p-nitroanilides mimicking fibrinogen
    and hirudin binding to thrombin. Design of slow reacting thrombin substrates. Int J Pept
    Protein Res 1996; 48: 182-93.
13. Duchemin J, Pittet JL, Tartary M, Beguin S, Gaussem P, Aihenc Gelas M, Aiach M. A new
    assay based on thrombin generation inhibition to detect both protein C and protein S deficiencies
    in plasma. Thromb Haemostas 1994; 71: 331-8.
14. Rosing, Tans G, Nicolaes GA, Thomassen MC, van OerIe R, van der Ploeg PM, Heijnen P,
    Hamulyak K, Hemker He. Oral contraceptives and venous thrombosis: different sensitivities
    to activated protein C in women using second and third generation oral contraceptives. Br J
    Haematol. 1997; 97: 2338-44.
15. Nicolaes GA, Thomassen MC, Tans G, Rosing J, Hemker HC. Effect of activated protein C
    on thrombin generation and on the thrombin potential in plasma of normal and APe resistant
    individuals. Blood Coag Fibrinolysis 1997; 8: 28--38.
16. Rotteveel RC, Roozendaal KJ, Eijsman L, Hemker He. The influence of oral contraceptives
    on the time integral of thrombin generation (thrombin potential). Thromb Haemostas 1993;
    70: 959-62.
                                                 78
8
Fibrinogen
M. P. M. DE MAAT, G. D. O. LOWE and F. HAVERKATE
INTRODUCTION
PATHOPHYSIOLOGY
Less specific cleavages of fibrinogen may occur in vivo 3 . For example, during
treatment with streptokinase or urokinase there is extensive fibrinogenolysis.
But even the plasma of healthy individuals shows some evidence of fibrinogen
cleavage4,5. It is assumed that fibrinogen is synthesized as high molecular weight
(HMW) fibrinogen with two intact carboxyl ends of the An-chains and becomes
degraded in the circulation. From the 340 kD HMW fibrinogen, the low
molecular weight (LMW) fibrinogens of about 305 kD and low' molecular
weight (LMW') fibrinogen of about 270 kD result after the removal of a 35
kD carboxyterminal polypeptide from one An-chain and from both An-chains,
respectively6,7. The identity of the proteolytic enzymes responsible for the
in-vivo generation of LMW fibrinogen under physiological conditions is at
present uncertain, but analysis of the carboxyl terminus of the An-chain has
excluded plasmin, gelatinase and trypsin 8,9. According to Holm and Godal6
the physiological distribution of heterogeneous fibrinogens in normal human
                                      79
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
plasma is 69.7%, 26.5% and 3.8% for HMW, LMW and LMW' fibrinogen
respectively6. This distribution has been confirmed by Furlan et al.1O. The
clotting times of LMW and LMW fibrinoren are prolonged compared with
the clotting time of HMW fibrinogen 7,l . The role of HMW and LMW
fibrinogen in the pathophysiology is unknown.
    Fibrinogen is necessary for clot formation and appropriate platelet function
in vivo. When measured by clotting-rate assays, its normal plasma level ranges
between 1.7 and 4.0 gIL, corresponding to 5--12 JIDloVL. The Michaelis-Menten
constant Km for the cleavage of the Aa_chain I2- 15 by thrombin is about 7
J.Ull0VL. Therefore, the velocity of clot formation is significantly affected by
different fibrinogen concentrations even within the 'physiological' range, and
contributes to the increased thrombotic risk associated with higher plasma
fibrinogen concentration.
    Low fibrinogen is an important diagnostic and pathophysiological variable
in various clinicopathological conditions l - 3, e.g. disseminated intravascular
coagulation (DIC), liver disease, thrombolytic or defibrination therapy. Therefore,
its measurement is often required by the clinician as an emergency test.
    In addition, epidemiological studies have been accumulating convincing
evidence l 6--19 that fibrinogen is an independent risk factor for cardiovascular
disease. An increase of the plasma fibrinogen of 1 standard deviation (15%)
approximately doubles the cardiovascular risk. Fibrinogen levels are associated
with many environmental, lifestyle and physiological risk factors, such as
smoking, body mass index, exercise, age, gender, race20--22, stress23 . It has been
suggested that the association of some of these factors, such as smoking, with
coronary artery disease may to some extent be mediated by increased fibrinogen
levels. The genetic contribution to the plasma level of fibrinogen is calculated
to be between 20% and 50%24,25 and several polymorphisms in the fibrinogen
genes have been identified that are related to the plasma fibrinogen level and the
regulation thereof4,26--28. Several drugs have been reported to reduce plasma
fibrinogen levels, such as fibric acid derivatives and ticlopidine29--32. The mechanism
by which these drugs lower fibrinogen has not yet been elucidated, but lowering
the fibrinogen levels by drugs is an interesting concept in risk reduction.
FIBRINOGEN ASSAYS
Reference values
The normal range of plasma fibrinogen by clotting assays is 1.7 to 4.0 giL.
The distribution of these values is slightly skewed to the right. Immunological
and heat precipitation methods give a higher normal range of 2.5-6.0 gIL.
Materials
Isotonic barbital-acetate buffer pH 7.35 (BAB)
1. Stock solution: 9.71 g sodium acetate. 3H20 and 14.71 g sodium 5,5-diethyl-
   barbiturate in 500 ml distilled water.
2. Working solution (barbital-acetate buffer BAB): 250 ml stock solution, 200
   ml NaCI4.25% (w/v), 217 ml HCI 0.1 mollL and 683 ml distilled water. The
   pH may be corrected with very small amounts of HCI.
Thrombin reagents are now widely available.
                                      81
             LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Procedure
Citrated plasma (fresh or frozen) is used for the assay. The plasma to be tested
is diluted 1 + 9 with BAB immediately before the assay.
   0.1 mIl + 9 diluted citrated plasma
   incubate 30 s at 37°C
   add 0.1 mI thrombin solution 33 IUlmI
   record the clotting time, using a coagulometer.
Calculation of results
The calibration curve of the Clauss assay is obtained from the clotting times
of serial dilutions (1 + 5, 1 + 9, 1 + 19, 1 + 29, 1 + 49) of a standard plasma
with known clottable fibrinogen concentration. Double logarithmic paper is
used to plot times on the ordinate against the concentrations (giL) on the
abscissa. The clotting times of the diluted test plasma samples are then converted
into fibrinogen concentrations. The calibration curve will be approximately
linear in the log-log plot (Figure 8.1).
50r-----------------~
                            '6'
                            !CD    10
                            ..,E
                            go
                            'i3
                            .g
1~~~----~~~~~
                                    0.5                               10
                                              flbrlnogen (g/l)
Figure 8.1 Typical example of a reference curve of the clotting rate assay by Clauss
                                               82
                                  FIBRINOGEN
Thrombin solution
500 Iu/ml solution of bovine or human thrombin in physiological saline (1 IV
- 1 NIH unit).
Clot solvent
400 g urea were solubilized in distilled water. Following the addition of 200 m1
of 1.0 moVL NaOH the solution was made up to 1000 ml with distilled water.
Procedure
  1 ml citrated plasma
  2 ml working buffer
  50 J.1l thrombin solution
are rapidly and thoroughly mixed and left at room temperature for 2 h. The
tube is placed upside-down on a lint-free cloth placed on absorbent material,
such as filter paper. The clot is allowed to synerize spontaneously and then
blotted to dryness using appropriate absorbent material. The clot is then
harvested and washed for 30 min in 50 ml 0.15 moVL NaCI solution. This
washing procedure is repeated twice, blotting dry between each washing. Do
not lose small pieces of the clot. Washing is the most difficult step of the assay.
Mter washing, transfer the clot to a tube containing 7.5 ml of the clot solvent
until complete solubility is achieved. The resultant solution is mixed and its
optical density (00) is measured at 280 nm and 315 nm in a quartz cuvette, 1
em lightpath, using clot solvent as a blank.
Calculation of results
The fibrinogen concentration (fbg) is calculated according to the following
formula:
           (00 of 280 nm - 00 of 315 nm) x 7.5/15.87         =fbg (gIL)
where 7.5 is the dilution factor and 15.87 is the extinction coefficient (E1'i:8) for
fibrin at alkaline pH. The relationship between 00 and fbg is linear up to an
                                        84
                                        FIBRINOGEN
References
 1. Doolittle RF. Fibrinogen and fibrin. In: Bloom AL, Thomas DP, eds. Haernostasis and
    thrombosis. Edinburgh: Churchill Livingstone, 1987; 192-215.
 2. Hantgan RR, Francis DW, Scheraga HA, Marder VI Fibrinogen structure and physiology.
    In: Colman RW, Hirsh J, Marder VJ, Salzman EW, eds. Hemostasis and thrombosis, 2nd edn.
    Philadelphia: Lippincott, 1987; 269-88.
 3. Francis CWO Marder VI Physiologic regulation and pathologic disorders of fibrinolysis. In:
    Colman RW, Hirsh J, Marder VJ, Salzman EW, eds. Hemostasis and thrombosis, 2nd edn.
    Philadelphia: Lippincott, 1987; 358-79.
 4. Mosesson MW, Finlayson JS, Umfleet RA, Galanakis D. Human fibrinogen heterogeneities.
    I. Structural and related studies of plasma fibrinogens which are high solubility catabolic
    intermediates. J Bioi Chern 1972; 247: 5210-19.
 5. Lipinska I, Lipinski B, Gurewich V. Fibrinogen heterogeneity in human plasma. Electrophoretic
    demonstration and characterization of two major fibrinogen components. J Lab Clin Med
    1974; 84: 509-16.
                                             85
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
 6. Holm B, Godal He. Quantitation of three normally occurring plasma fibrinogens in health
    and during so-called 'acute phase' by SDS e1ectrophoresis of fibrin obtained from EDTA-plasma.
    Thromb Res 1984; 35:279-90.
 7. Holm B, Nilsen DWT, Kieru1f P, Godal He. Purification and characterization of 3 fibrinogens
    with different molecular weights obtained from normal human plasma. Thromb Res 1985;
    37: 165-76.
 8. Nakashima A, Sasaki S, Miyazaki K, Miyata T, Iwanaga S. Human fibrinogen heterogeneity:
    the COOH-terminal residues of defective Aa chains of fibrinogen II. Blood Coag Fibrinol
    1992; 3: 361-70.
 9. Miiller E, Henschen A. Isolation and characterization of human plasma fibrinogen molecular
    size-variants by high-performance liquid chromatography and amino acid sequence analysis.
    In: Mosesson MW, Amrani DL, Siebenlist KR, DiOrio JP, eds. Fibrinogen 3. Biochemistry,
    biological functions, gene regulation and expression. Amsterdam: Elsevier, 1988; 279-82.
10. Furlan M, Felix R, Escher N, Liimm1e B. How high is the true fibrinogen content of fibrinogen
    standards? Thromb Res 1989; 56: 583-92.
II. Holm B, Brosstad F, Kierulf P, Godal HC. Polymerization properties of two normally
    circulating fibrinogens, HMW and LMW. Evidence that the COOH-terminal end of the
    a-chain is of importance for fibrin polymerization. Thromb Res 1985; 39: 595-606.
12. Bando M, Matsushima A, Hirano J, Imadu Y. Thrombin catalyzed conversion of fibrinogen
    to fibrin. J Biochem 1972; 71: 897-9.
13. Martinelli RA, Scheraga HA. Steady-state kinetic study of the bovine thrombin-fibrinogen
    interactions. Biochemistry 1980; 19: 2343-50.
14. Higgins DL, Lewis SD, Shafer JA. Steady-state kinetic parameters for the thrombin catalyzed
    conversion of human fibrinogen to fibrin. J Bioi Chern 1983; 258: 927fr82.
IS. Lewis SD, Shafer JA. A thrombin assay based upon the release of fibrinopeptide A from
    fibrinogen: definition of a new thrombin assay based upon the release of fibrinopeptide A
    from fibrinogen; definition of a new thrombin unit. Thromb Res 1984; 35: 111-20.
16. Ernst E, Resch K.L. Fibrinogen as a cardiovascular risk factor: a meta-analysis and review
    of the literature. Ann Intern Med 1993; 118: 956--63.
17. Lowe GD. Fibrinogen and cardiovascular disease: historical introduction. Eur Heart J 1995;16
    (Supp.l A): 2-5.
18. Thompson SG, Kienast J, Pyke SD, Haverkate F, van de Loo Je. Hemostatic factors and the
    risk of myocardial infarction or sudden death in patients with angina pectoris. European
    Concerted Action on Thrombosis and Disabilities Angina Pectoris Study Group. N Eng! J
    Med 1995; 332: 635-41.
19. Heinrich J, Balleisen L, Schulte H, Assmann G, van de Loo J. Fibrinogen and factor VII in
    the prediction of coronary risk: results from the PROCAM study in healthy men. Arterioscler
    Thromb 1994; 14: 54-9.
20. Balleisen L, Bailey J, Epping PH, Schulte H, van de Loo J. Epidemiological study on factor
    VII, factor VIII and fibrinogen in an industrial population: I. Baseline data on the relation
    to age, gender, body weight, smoking, alcohol, pill-using, and menopause. Thromb Haemostas
    1985; 54: 475--9.
21. Krobot K, Hense Hw, Cremer P, Eberle E, Keil U. Determinants of plasma fibrinogen: relation
    to body weight, waist-to-hip ratio, smoking, alcohol, age, and sex. Results from the second
    MONICA Augsburg survey 1989-1990. Arterioscler Thromb 1992; 12: 780-8.
22. Folsom AR, Wu KK, Davis CE, Conlan MG, Sorlie PD, Szklo M, for the Atherosclerosis
    Risk in Communities (ARIC) Study Investigators. Population correlates of plasma fibrinogen
    and factor VII, putative cardiovascular risk factors. Atherosclerosis 1991; 91: 191-205.
23. Mattiasson I, Lindgarde F. The effect of psychosocial stress and risk factors for ischaemic
    heart disease on the plasma fibrinogen concentration. J Intern Med 1993; 234: 45--51.
24. Humphries SE, Cook M, Dubowitz M, Stirling Y, Meade Tw. Role of genetic variation at
    the fibrinogen locus in determination of plasma fibrinogen concentrations. Lancet 1987; I:
    1452-5.
25. Hamsten A, Iseluis L, de Faire U, Blombiick M. Genetic and cultural inheritance of plasma
    fibrinogen concentration. Lancet 1987; 2: 988-92.
26. Green F, Hamsten A, Blombiick M, Humphries S. The role of beta-fibrinogen genotype in
    determining plasma fibrinogen levels in young survivors of myocardial infarction and healthy
    controls from Sweden. Thromb Haemostas 1993; 70: 915-20.
                                              86
                                         FIBRINOGEN
27. Ferrer Antunes CA, de Maat MPM, Palmeiro A, Pimentel J, Fernandes V. Association between
    polymorphisms in the fibrinogen a- and f}-genes on the post-trauma fibrinogen increase.
    Throm. Res. 1998: in press.
28. Montgomery HE, Clarkson P, Nwose OM, Mikailidis DP, Jagroop lA, Dollery G, Moult J,
    Benhizia F, Deanfield J, Jubb M, World M, McEwan JR, Winder A, Humphries S. The acute
    rise in plasma fibrinogen concentration with exercise is influenced by the G-(453)-A
    polymorphism of the beta-fibrinogen gene. Arterioscler Thromb Vasc Bioi 1996; 16: 386--91.
29. de Maat MPM, Arnold AER, van Buuren S, Wilson JHP, KIuft C. Modulation of plasma
    fibrinogen levels by ticlopidine in healthy volunteers and patients with stable angina pectoris.
    Thromb Haemostas 1996; 76: 166--70.
30. de Maat MPM, Knipscheer HC, Kastelein JJP, KIuft C. Modulation of plasma fibrinogen
    levels by ciprofibrate and gemfibrozil in primary hyperiipidaemia. Thromb Haemostas 1997;
    77: 75-9.
31. Mazoyer E, Ripoll L, Boisseau MR, Drouet L. How does ticlopidine treatment lower plasma
    fibrinogen? Thromb Res 1994; 75: 361-70.
32. Branchi A, Rovellini A, Sommariva D, Gugliandolo AG, Fasoli A. Effect of three fibrate
    derivatives and of two HMG.coA reductase inhibitors on plasma fibrinogen level in patients
    with primary hypercholesterolemia. Thromb Haemostas 1993; 70: 241-3.
33. De Maat MPM, Haverkate F. Critical evaluation of fibrinogen assays. In: Seghatchian MJ,
    Samama MM, Hecker SP, eds. Hypercoagulable states: fundamental aspects, acquired disorders,
    and congenital thrombophilia. Boca Raton: CRC Press, 1996; 105-16.
34. Von Clauss A. Gerinnungsphysiologische Schnellmethode zur Bestimmung des Fibrinogens.
    Acta Haematol1957; 17: 237-46.
35. Blombiick B, Blomback M. Preparation of human fibrinogen fraction 1-2. Arkiv Kemi 1956;
    10: 415-43.
36. Ratnoff OD, Menzie C. A new method for the determination of fibrinogen in small samples
    of plasma. J Lab Clin Med 1951; 37: 316--20.
37. Swain W, Feders MB. Fibrinogen assay. Clin Chern 1967; 13: 1026--8.
38. Vermylen C, de Vreker RA, Verstraete M. A rapid enzymatic method for assay of fibrinogen
    fibrin polymerization time (FPT test). Clin Chim Acta 1963; 8: 418-24.
39. Becker U, Bartl K, Wahlefeld AW. A functional photometric assay for plasma fibrinogen.
    Thromb Res 1984; 35: 475-84.
40. De Metz M, van Wersch JWI Use of a centrifugal analyzer for a chromogenic prothrombin
    time, a chromogenic activated partial thromboplastin time and a kinetic fibrinogen assay in
    a routine hospital laboratory. Haemostasis 1987; 17: 254-9.
41. Cambas JP, Bierme R. Martinon JC, Dousset B. Evaluation des performances d'un automate
    en coagulation: l'Electra 700 Nouv Rev Fr Hemoto11985; 25: 313-20.
42. Bick RZ, Wheeler A, Camosano NA. Comparative study of the DuPont antithrombin III
    and fibrinogen assay systems. Am J Clin Patho11985; 83: 541-6.
43. Hoffmann JJML, Verhappen MAL. Automated nephelometry of fibrinogen: analytical
    performance and observations during thrombolytic therapy. Clin Chem 1988: 34: 2135-40.
44. Exner T, Burridge J, Power P, Richard KA. An evaluation of currently available methods for
    plasma fibrinogen. Am J Clin Patho11979; 71: 521-7.
45. Schulz FH. Eine einfache Bewertungsmethode von Leberparenchymschiiden (volumetrische
    Fibrinbestimmung). Acta Hepatol1955; 3: 306--10.
46. Hoffman M, Greenberg CS. The effects of fibrin polymerization inhibitors on quantitative
    measurements of plasma fibrinogen. Am J Clin Patho11987; 88: 490-3.
47. Hoegee-de Nobel E, Voskuilen M, Briet E, Brommer EJP, Nieuwenhuizen W. A monoclonal
    antibody-based quantitative enzyme immunoassay for the determination of plasma fibrinogen
    concentrations. Thromb Haemostas 1988; 60: 415-18.
48. Sweetham PM, Thomas HF, Yarnell JWG, Beswich A, Baker lA, Elwood PC. Fibrinogen,
    viscosity and the 10-year incidence of ischaemic heart disease. The Caerphilly and Speedwell
    studies. Eur Heart J 1996; 17: 1814-20.
49. Cremer P, Nagel D, Labrot B, Mann H, Muche R, Elster H, Seidel D. Lipoprotein Lp(a) as
    predictor of myocardial infarction in comparison to fibrinogen, LDL cholesterol and other
    risk factors: results from the prospective Gottingen Risk Incidence and Prevalence Study
    (GRIPS). Eur J Clin Invest 1994; 24: 444-53.
50. Sweetnam PM, Yarnell JWG, Lowe GDO, Baker lA, O'Brien JR, Rumley A, Etherington
                                               87
            LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
      MD, Whitehead PJ, Elwood PC. The relative predictive power of heat-precipitation
      nephelometric and clottable (Clauss) fibrinogen in the prediction of ischaemic heart disease:
      the Caerphilly and Speedwell studies. Br J Haematol1998; 100:582-8.
51.   Jacobson K. Studies in the determination of fibrinogen in human blood plasma. Scand J Clin
      Lab Invest 1955 (Suppl. 14): 1-54.
52.   Gaffney PJ, Wong MY. Collaborative study of a proposed international standard for plasma
      fibrinogen measurement. Thromb Haemostas 1992; 68: 428-32.
53.   Thompson SG, Duckert F, Haverkate F, Thomson 1M. The measurement of haemostatic
      factors in 15 European laboratories: quality assessment for the multicentre ECAT Angina
      pectoris study. Thromb Haemostas 1989; 61: 301--6.
54.   Siefring GE, Riabov DK, Wehrly JA. Development and analytical performance of a functional
      assay for fibrinogen on the Du Pont aca™ analyzer. Clin Chern 1983; 29: 614-17.
55.   McDonagh J, Carrell N. Disorders of fibrinogen structure and function. In: Colman RW,
      Hirsch J, Marder VJ, Salzman EW, eds. Hemostasis and thrombosis, 2nd edn. Philadelphia:
      Lippincott, 1987; 301-17.
56.   Jespersen J, Sidelmann JA. A study of the conditions and accuracy of the thrombin time assay
      of plasma fibrinogen. Acta Haematol1982; 67: 2-7.
57.   Palareti G, Maccaferri M, Manotati C, Tripodi A, Chantaranghul V, Rodeghiero F,
      Ruggeri M, Mannucci PM. Fibrinogen assays: a collaborative study of six different methods.
      Clin Chem 1991; 37: 714-19.
58.   Chitolie A, Mackie 11, Grant D, Hamilton JL, Machin SM. Inaccuracy of the 'derived'
      fibrinogen measurement. Blood Coag Fibrin 1994; 5: 955-7.
59.   Rossi E, Mondonico P, Lombardi A, Preda L. Method for the determination of functional
      (clottable) fibrinogen by the new family of ACL coagulometers. Thromb Res 1988; 52: 453--68.
60.   Marder VJ, Shulman NR. High molecular weight derivatives of human fibrinogen produced
      by plasmin II. Mechanism of their anticoagulant activity. J Bioi Chern 1969; 244: 2120-4.
61.   Saleem A, Fretz K. An improved method for plasma fibrinogen based on thrombin time
      measurement. Clin Chim Acta 1975; 62: 131--6.
                                               88
9
Activated factor VII
J. H. MORRISSEY
INTRODUCTION
Factor VIla, a vitamin K -dependent plasma serine protease, is the first enzyme
in the coagulation cascade. The majority of this glycoprotein circulates as the
inert zymogen (factor VII) with a concentration in pooled normal plasma of
about 470 nglml (9.4 nmollL)l. However, low levels of activated factor VII
(factor VIla) are also present in the circulation. Typically, factor VIla makes
up less than I % of the total factor VII in plasma2•
   Factor VII zymogen consists of a single polypeptide chain (MW 50 000)
with the following domain structure (N-terminal to C-terminal): a y-carboxy-
glutamate-rich domain (Gla domain), two epidermal growth factor-like domains
(EGF domains), and a serine protease domain homologous to trypsin and
chymotrypsin3 • Factor VII has no measurable enzymatic activity until it is
converted to the active form (factor VIla) by hydrolysis of a single peptide
bond located between the second EGF domain and the protease domain. The
result is a two-chain protein of the same MW, whose heavy and light chains
are held together by a disulphide bond.
   Currently, two methods exist for measuring the levels of factor VIla in
plasma: an activity-based method employing a modified form of tissue facto~,
and an ELISA-based method using an antibody that binds to the C-terminus
of the factor VIla light chain4 • These methods yield markedly different values
for the plasma content of factor VIla.
PHYSIOLOGICAL ROLE
Factor VIla is a relatively weak serine protease until it binds to its essential
cofactor, tissue factor (also known as tissue thromboplastin). Tissue factor has
no enzymatic activity itself Instead, it binds to factor VIla and enhances its
                                     89
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
enzymatic activity in an allosteric manner. Thus, tissue factor can be considered
to be the regulatory subunit, and factor VIla the catalytic subunit, of the first
enzyme in the so-called extrinsic pathway of blood coagulation. The factor
VIla/tissue factor complex is considered to be the physiological initiator of
coagulation in normal haemostasis and many, if not all, thrombotic diseases5 .
    Tissue factor is an integral membrane glycoprotein (MW 45 000) present on
the surface of a variety of different cell types located outside the vasculature.
Tissue factor is also an inducible protein in monocytes and vascular endothelial
cells5 .
    In addition to its ability to bind to and enhance the activity of factor VIla,
tissue factor also binds zymogen factor VII with high affinity. Once bound to
tissue factor, factor VII is more easily activated to factor VIla. A variety of
serine proteases have been shown in vitro to catalyse the activation of factor VII,
including thrombin and factors IXa, Xa, and XIla. In addition, factor VIla can
catalyse the activation of factor VII in the presence of tissue factor ('factor
VII autoactivation,)6. It is presently unclear which proteases are the most
important for activation of factor VII to VIla during clotting in vivo.
    Recently it has been demonstrated that plasma contains trace levels of factor
VIIa2, which are proposed to be important in priming the clotting system
following the exposure of tissue factor to the blood7,8. Since plasma has a high
content of protease inhibitors, the active forms of most of the coagulation
serine proteases have extremely short biological half-lives (~l min) and con-
sequently cannot be directly measured in plasma samples. Factor VIla, however,
is extremely resistant to plasma protease inhibitors, allowing it to circulate with
a relatively long half-life (about 2.5 h, compared with a biological half-life of
about 5 h for factor VII)9.
PATHOPHYSIOLOGICAL ASPECTS
Substantial clinical interest in factor VII was generated by the results of the
first N orthwick Park Heart Study, a large prosrective study of the incidence
of heart disease in a cohort of middle-aged men! . This study implicated plasma
factor VII coagulant activity as a significant risk factor for ischaemic heart
disease. A similar, although less strong, correlation was also reported by the
PROCAM study!!. Interestingly, follow-up studies of both populations indicate
that elevated factor VII coagulant activity appears to be most significant as a
predictor of fatal heart attacks although, again, this was more significant in
the Northwick Park Heart Study than in the PROCAM study!2,13.
   Not every study has found a correlation between elevated factor VII coagulant
activity and risk of heart disease, leading to some controversy about the utility
of factor VII as a risk factor for thrombotic diseases. A major point of dis-
agreement between laboratories has centred on what is actually measured in
factor VII coagulant assays (Chapter 10). These assays employ tissue factor to
initiate coagulation, and tissue factor is well known to promote the conversion
of factor VII to VIla; thus, they measure an aggregate of both factor VII and
VIla. Furthermore, the sensitivity of the assay to factor VIla versus factor VII
                                       90
                            ACTIVATED FACTOR VII
appears to depend upon the assay configuration 7 ,14. For example, assays
employing bovine thromboplastin are more sensitive to the plasma content of
factor VIla than assays employing human or rabbit thromboplastin 15 . In
particular, it was proposed that the assay of factor VII coagulant activity used
by the Northwick Park Heart Study might be more sensitive to plasma levels
of factor VIla than assays used in other epidemiological studies. This was
demonstrated directly when the activity-based assay for plasma factor VIla
was developed 16 .
   Consistent with earlier predictions7, trace levels of factor VIla have been
found in the plasma of all normal volunteers tested to date2• The amount of
factor VIla in pooled normal plasma is about 3 ng/ml. Using the activity-
based assay, plasma factor VIla levels have been shown to be elevated during
pregnancy and in diseases associated with thrombotic risk, such as diabetes2,17-19.
Several prospective studies are currently addressing the role of factor VIla as
a risk predictor of thrombotic disease. Plasma factor VIla levels have been
shown to be especially sensitive to warfarin treatment, including low-dose
warfarin which is difficult to monitor otherwise2,20. Interestingly, haemophilia
B patients were found to have substantially lower factor VIla levels than normal,
while haemophilia A patients had normallevels21 .
   The recently reported ELISA for factor VIla yields values that are lOO-fold
lower than the activity-based assay4. Unlike the values obtained with the
activity-based assay, plasma factor VIla levels measured by the ELISA are
correlated with prothrombin fragment F I +2 levels (Chapter 23). Some possible
reasons for the differences in measured factor VIla levels using these two assays
are discussed below.
Principle
Recombinant tissue factor lacking the membrane anchor and cytoplasmic
domains (soluble tissue factor, or sTF) retains the ability to bind to factor VIla
and enhance its enzymatic activity. However, unlike wild-type tissue factor,
sTF has selectively lost the ability to promote the conversion of factor VII to
VIIa22 • Therefore, sTF shortens the clotting time of plasma in a manner that
is absolutely dependent upon its content of preformed factor VIla. This unique
property of sTF allowed the development of a clot-based assay for plasma
factor VIla that is free from interference from zymogen factor VIe. In order
to isolate the assay from variability in the content of other clotting factors in
the test plasma, the test plasma sample is typically diluted lO-fold and mixed
with an excess of factor VII-deficient plasma (which is correspondingly deficient
in factor VIla). The diluted plasma sample and factor VII-deficient plasma are
mixed with a reagent composed of sTF and phospholipid vesicles, and the time
to clot formation is measured following addition of a CaCl2 solution. A standard
curve is prepared in which varying amounts of purified factor VIla are added
to factor VII-deficient plasma. Factor VIla levels in the test plasma samples
are then determined by reference to the standard curve.
                                       91
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Materials
HEPES-buffered saline (HBS)
50 mmoVL HEPES-NaOH buffer, pH 7.5 (at 25°C), containing 100 mmoVL
NaCl and 0.02% w/v NaN 3 •
HEPES-BSA buffer
50 mmoVL HEPES-NaOH buffer, pH 7.5 (at 25°C), containing 0.02% w/v
NaN 3 and 1% w/v bovine serum albumin, fatty acid-free (Calbiochem). Store
at 4°C.
Phospholipid vesicles
In our laboratory we prepare phospholipid vesicles using a 20:40:40 mixture
of the following purified phospholipids: bovine brain phosphatidylserine (PS),
bovine liver phosphatidylethanolamine (PE), and egg phosphatidylcholine (PC)
(available from Avanti Polar Lipids). The phospholipids are supplied as 10
mg/ml solutions in chloroform. Dispense 80 J.11 PS (0.8 mg), 160 J.11 PE (1.6 mg)
and 160 J.1l PC (1.6 mg) into an Eppendorf tube and dry down under a stream
of nitrogen or argon. When dry, place under high vacuum for an additional
hour to remove any remaining traces of solvent. Add I ml HBS and vortex
well to resuspend the phospholipids. Disperse the aggregated phospholipids
either by passing the suspension 15 times through a vesicle extruder containing
a filter with 100 nm pores (Liposofast vesicle extruder from Avestin), or by
sonication. The result is a 5 mmollL solution of phospholipid vesicles.
Alternatively, a commercial cephalin preparation that is free from activators
of the contact system may be used.
Calibrator
Obtain citrated plasma samples from two donors known to have high and low levels
of plasma factor VIla. Freeze in small aliquots at -80°C. When ready to use, thaw
quickly at 37°C, mix well and hold at room temperature until needed. Immediately
before use, dilute the control plasmas lO-fold with HEPES-BSA buffer.
Samples
Use platelet-poor, citrated plasma. Avoid contact with unsiliconized glass, and do
not store at 4 °C or on ice. Samples can be stored frozen at -80°C. When ready
to use, thaw quickly at 37°C, mix well and hold at room temperature until needed.
Immediately before use, dilute the samples lO-fold with HEPES-BSA buffer.
Procedure
The clot-based activity assay can use any method for detection of clot formation
that is suitable for measuring prothrombin times, such as the manual tilt-tube
method or an automated coagulometer. In any case it is essential that the
clotting tests be performed using either plastic containers or glass containers
that have been siliconized. Plasma samples should never come into contact
with untreated glass surfaces.
   Prior to performing the assay, thaw plasma samples and sTF reagent rapidly
at 37°C, mix well and hold at room temperature until used. Pre-warm the
calcium chloride solution and the cuvettes or test tubes in which clotting times
will be measured to 37°C. Add, in the following order, to the pre-warmed
cuvette or test tube:
  50 III diluted plasma sample or diluted calibrator
  50 III factor VII-deficient plasma
  50 III sTF reagent.
                                         93
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Wait 180 s (to ensure the materials reach 37°C), then add:
  50 J.1l calcium chloride solution.
Mix well and measure the time to clot formation from the moment the calcium
chloride solution is added. Typically, the clotting times are determined in
duplicate.
Calibration
The clotting times for the diluted calibrators are measured in duplicate and a
standard curve is prepared by plotting the data on a double-logarithmic scale,
to which a line or second-order polynomial is fitted by the least-squares method
(occasionally a third-order polynomial must be fitted, especially if the standard
curve is extended to include very low concentrations of factor VIla). This
method allows the measurement of plasma factor VIla levels in ng/ml with
reference to the purified factor VIla used in preparing the calibrators. An
example of a typical standard curve obtained in this way is given in Figure
9.lA. However, as the potency of purified factor VIla may vary from laboratory
to laboratory, it has been proposed that the level of factor VIla in the calibrators
itself be calibrated a~ainst the First International Standard Factor VIla
concentrate (89/688)2 . Consistent with this suggestion, the commercially
available kit (Staclot VIla-rTF; Diagnostica Stago) is calibrated against this
standard and yields factor VIla values in mVlml. A typical calibration curve
obtained with the Staclot VIla-rTF assay kit is given in Figure 9.lB.
100 r - - - - - - - - - - ,
3 10
Figure 9.1 Typical calibration curves for the activity assay for plasma factor VIla (clotting time
versus factor VIla concentration). Panel A: calibration curve using the method described in this
chapter, expressed in nglmL purified factor VIla. If necessary (Le. for measuring very low levels
of factor VIla), the standard curve can readily be extended to 0.03 nglmL. Panel B: calibration
curve using the commercially available kit (Staclot VIla-rTF; Diagnostica Stago), expressed in
mU/ml factor VIla (relative to the First International Standard Factor VIla concentrate). Data
for both curves were collected on a model ST4 coagulometer (Diagnostica Stago). In both cases
the indicated factor VIla concentrations are those obtained after dilution with HBS-BSA buffer.
Plasma factor VIla levels determined for test samples must therefore be multiplied by the dilution
factor of the sample (typically, lO-fold).
                                               94
                           ACTIVATED FACTOR VII
Reference values
Using the sTF-based clotting assay, the levels of factor VIla in the plasma of
188 normal volunteers was 0.5-8.4 ng/ml, with a mean value of 3.6 ± 1.4 ng/ml.
Pitfalls
1. Potency of the factor VIla standard. As mentioned above, the potency of
   different preparations of purified factor VIla can vary, making it very
   difficult to compare the absolute levels of plasma factor VIla measured in
   different laboratories. Hubbard and Barrowcliffe26 recommended that the
   assay be calibrated against the First International Standard Factor VIla
   concentrate (89/688), allowing the results to be presented in mU/ml factor
   VIla rather than ng/ml. This is an excellent suggestion, and the process need
   only be performed once per batch of factor VIla calibrators. An alternative
   is to use (or calibrate against) the commercially available activity assay for
   factor VIla, which is itself already calibrated against this standard.
2. Cold activation of plasma samples. Artifactual generation of factor VIla in
   citrated plasma samples can occur if liquid plasma samples are held at
   reduced temperature (such as 4 0c) in the presence of untreated glass or
   any other activator of the contact system. Cold activation can be avoided
   by collecting, processing and storing plasma samples in containers made of
   plastic or siliconized glass. Furthermore, blood should be handled and
   processed at room temperature, and platelet-poor plasma should be frozen
   at -80°C. When needed, the plasma samples should be thawed rapidly at
   37°C, mixed thoroughly, and held at room temperature until assayed.
   Samples can be refrozen and rethawed, although the levels of factor VIla
   will be diminished somewhat with each freeze-thaw cycle.
3. Quality of factor VII-deficient plasma. Factor VII-deficient plasma must
   have very low levels of factor VIla, otherwise the standard curve will plateau
   at lower levels of added factor VIla. In this case, factor VII-deficient plasma
   with lower endogenous levels of factor VIla must be obtained.
Typically, antibodies raised against factor VII or VIla recognize both the
zymogen and activated forms equally well. Recently, however, a factor
                                      95
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
VIla-specific ELISA has been reported, the basis for which is a c~ture antibody
that binds to an activation-specific neoantigen on factor VIla . This capture
antibody is a polyclonal rabbit antibody raised against a synthetic peptide that
comprises the last 12 amino acids at the C-terminus of the factor VIla light
chain. This peptide region is highly likely to differ in structure in factor VII
versus factor VIla, because in factor VII the peptide sequence is embedded in
a continuous polypeptide chain, whereas in factor VIla it has a free C-terminus
and is likely to be highly solvent-exposed.
   In practice, ELISA plates are coated with the capture antibody and, after
suitable blocking and washing steps, the wells are incubated with plasma
samples to allow capture of factor VIla. After washing, the bound factor VIla
is detected with a biotinylated second antibody against factor VIINIla (either
a monoclonal or polyclonal antibody), which is subsequently detected using
streptavidin-alkaline phosphatase and a suitable alkaline phosphate substrate.
The assay is calibrated using purified factor VIla.
   The activity-based assay and the ELISA give excellent agreement when
measuring purified factor VIla added to plasma4 • However, results from the
two assays differ dramatically when used to measure the endogenous levels of
factor VIla in plasma. Thus, according to the factor VIla-specific ELISA,
normal plasma contains 0.025 ± 0.01 nglml factor VIla, which is some 100-fold
lower than that measured by the activity assays.
   The reason for the discrepancy between the factor VIla-specific ELISA and
the activity-based assays for plasma factor VIla is currently not known. One
potential explanation is that the C-terminus of the factor VIla light chain may
undergo proteolysis during its relatively long lifetime in the circulation. If so,
this would render factor VIla unable to bind to the anti-peptide antibody while
potentially having no effect on its activity. Indeed, plasma is known to contain
high levels of arginine-specific carboxypeptidase activity27, which could remove
the last amino acid of the factor VIla light chain. If the C-terminus of the
factor VIla light chain does undergo proteolysis during circulation, this may
explain why the two assays agree when measuring purified factor VIla that is
added to plasma, but do not agree when measuring the endogenous factor VIla
in plasma. Clearly, further research is needed to clarify these questions.
References
1. Fair DS. Quantitation of factor vn in the plasma of normal and warfarin-treated individuals
   by radioimmunoassay. Blood 1983; 62: 784--91.
2. Morrissey JH, Macik BG, Neuenschwander PF, Comp PC. Quantitation of activated factor
   VII levels in plasma using a tissue factor mutant selectively deficient in promoting factor VII
   activation. Blood 1993; 81: 734-44.
3. Hagen FS, Gray CL, O'Hara P, Grant FJ, Saari GC, Woodbury RG, Hart CE, Insley M,
   Kisiel W, Kurachi K et al. Characterization of a cDNA coding for human factor VII. Proc
   Nat! Acad Sci USA 1986; 83: 2412-16.
4. Philippou H, Adami A, Amersey RA, Stubbs PJ, Lane DA. A novel specific immunoassay
   for plasma two-chain factor VIla: Investigation of FVlIa levels in normal individuals and in
   patients with acute coronary syndromes. Blood 1997; 89: 767-75.
5. Carson SD, Brozna JP. The role of tissue factor in the production of thrombin. Blood Coag
   Fibrinol1993; 4: 281-92.
                                              96
                                  ACTIVATED FACTOR VII
 6. Nakagaki T, Foster DC, Berkner KL, Kisiel W. Initiation of the extrinsic pathway of blood
    coagulation: Evidence for the tissue factor dependent autoactivation of human coagulation
    factor VII. Biochemistry 1991; 30: 10819-24.
 7. Hultin MB, Jesty J. Factor VIla levels in patients with hemophilia. Blood 1992; 80: 3255-6.
 8. Rapaport SI. Regulation of the tissue factor pathway. Ann NY Acad Sci 1991; 614: 51-62.
 9. Seligsohn U, Kasper CK, Osterud B, Rapaport SI. Activated factor VII: presence in factor
    IX concentrate and persistence in the circulation after infusion. Blood 1978; 53: 828-37.
10. Meade TW, Mellows S, Brozovic M, Miller GJ, Chakrabarti RR, North WRS, Haines AP,
    Stirling y, Imeson JD, Thompson SG. Haemostatic function and ischaemic heart disease:
    principal results of the Northwick Park heart study. Lancet 1986; 2: 533-7.
11. Balleisen L, Schulte H, Assmann G, Epping P-H, Van de Loo J. Coagulation factors and the
    progress of coronary heart disease. Lancet 1987; 2: 46l.
12. Ruddock V, Meade Tw. Factor VII activity and ischaemic heart disease: fatal and non-fatal
    events. Q J Med 1994; 87: 403-6.
13. Heinrich J, Balleisen L, Schulte H, Assmann G, Van de Loo J. Fibrinogen and factor VII in
    the prediction of coronary risk: results from the PROCAM study in healthy men. Arterioscler
    Thromb 1994; 14: 54-9.
14. Mann KG. Factor VII assays, plasma triglyceride levels, and cardiovascular disease risk.
    Arteriosclerosis 1989; 9: 783-4.
15. Hemker HC, Muller AD, Gonggrup R. The estimation of activated human blood coagulation
    factor VII. J Mol Med 1976; 1: 127-34.
16. Miller GJ, Stirling Y, Esnouf MP, Heinrich J, Van de Loo J, Kienast J, Wu KK, Morrissey
    JH, Meade Tw, Martin JC et al. Factor VII-deficient substrate plasmas depleted of protein
    C raise the sensitivity of the factor VII bio-assay to activated factor VII: an international
    study. Thromb Haemostas 1994; 71: 38-48.
17. Kario K, Miyata T, Sakata T, Matsuo T, Kato H. Fluorogenic assay of activated factor VII:
    Plasma factor VIla levels in relation to arterial cardiovascular diseases in Japanese. Arterioscler
    Thromb 1994; 14: 265-74.
18. Kario K, Matsuo T, Matsuo M, Kolde M, Yamada T, Nakamura S, Sakata T, Kato H, Miyata
    T. Marked increase of activated factor VII in uremic patients. Thromb Haemostas 1995; 73:
    763-7.
19. Kario K, Matsuo T, Kobayashi H, Matsuo M, Sakata T, Miyata T. Activation of tissue factor-
    induced coagulation and endothelial cell dysfunction in non-insulin-dependent diabetic patients
    with microalbuminuria. Arterioscler Thromb Vase Bioi 1995; 15: 1114-20.
20. Raskob GE, Durica SS, Morrissey JH, Owen WL, Comp PC. Effect of treatment with low-dose
    warfarin-aspirin on activated factor VII. Blood 1995; 85: 3034-9.
2l. Wildgoose P, Nemerson Y, Hansen LL, Nielsen FE, Glazer S, Hedner U. Measurement of
    basal levels of factor VIla in hemophilia A and B patients. Blood 1992; 80: 25-8.
22. Neuenschwander PF, Morrissey JR. Deletion of the membrane anchoring region of tissue
    factor abolishes autoactivation of factor VII but not cofactor function. Analysis of a mutant
    with a selective deficiency in activity. J BioI Chem 1992; 267: 14477-82.
23. Rezaie AR, Fiore MM, Neuenschwander PF, Esmon CT, Morrissey JH. Expression and
    purification of a soluble tissue factor fusion protein with an epitope for an unusual calcium-
    dependent antibody. Protein Express Purif 1992; 3: 453-60.
24. Shigematsu Y, Miyata T, Higashi S, Miki T, Sadler JE, Iwanaga S. Expression of human
    soluble tissue factor in yeast and enzymatic properties of its complex with factor VIla. J BioI
    Chem 1992; 267: 21329-37.
25. Ruf W, Rehemtulla A, Morrissey JH, Edgington TS. Phospholipid-independent and -dependent
    interactions required for tissue factor receptor and cofactor function. J BioI Chem 1991; 266:
    2158-66.
26. Hubbard AR, Barrowcliffe Tw. Measurement of activated factor VII using soluble mutant
    tissue factor - Proposal for standardization. Thromb Haemostas 1994; 72: 649-50.
27. Plummer T.H, Husmann M. Human plasma carboxypeptidase N: isolation and characterization.
    J BioI Chern 1978; 253: 3907-12.
                                                97
10
Factor VII activity and antigen
G. MARIANI, G. LIBERTI, T. D'ANGELO and L. LO COCO
INTRODUCTION
Elevated FVII levels have been reported to be associated with an increased risk
of cardiovascular disease2, whilst FVII deficiencies are associated with a variable
bleeding tendency. As to the congenital deficiencies, there has been a considerable
degree of uncertainty on the relation between the clinical presentation of the
deficiency on one hand and FVII levels and the underlying genetic defect on
the other3 . Other elements may playa role in the interpretation of this
                                       99
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
relationship, such as FVII plasma assays and other variations (gene poly-
morphisms) within the FVII gene.
   An accurate laboratory evaluation of FVII levels thus appears the basic
requirement for the evaluation of any change in blood coagulation in which
FVII is involved, and the first assays to be carried out are FVII coagulant
activity (FVIIc) and FYII antigen (FVIIAg). FYIIAg expresses the total amount
of the protein in plasma; FVllc reflects the functional activity of the protein;
in fact, FVIIc is the result of the combined effects of FVIIa and FVII zymogen
in a one-stage clotting time. Unfortunately the results are dependent on the
type of thromboplastin selected4 .
FVllc activity is the basic assay in evaluating the level or function of the
extrinsic pathway of blood coagulation. FVllc has been developed to study
the congenital deficiencies of the factor and thromboplastin preparations from
different species were used to identify some deficiency variants 3,5. Bovine
thromboFlastin has also been used to evaluate the state of activation of the
molecule. Furthermore, FVIIc and/or FVIIAg assars are also currently used
in liver disease to evaluate the hepatocyte function 7 ••
   FVllc is a sensitive assay allowing the detection of a few nanograms of the
protein. The reproducibility of the assay is also good, with very low intra- and
inter-assay coefficients of variation 9 •
Method outlines
The specific assay of clotting factors of the extrinsic pathway (factors II, V,
VII, X) in general consists of performing the prothrombin time (PT) (Chapter
6) of a mixture made up of a dilution of normal or patient's plasma and a
plasma specifically lacking the factor to be assayed (substrate/plasma). The
substrate plasma can be obtained from patients with a congenital, severe
deficiency « 1%) or from normal plasma artificially depleted by specific
monoclonal or polyclonal antibodies. The percentage of factor activity present
in plasma can be determined by the degree of correction of the clotting time
when the plasma to be tested is added to the substrate plasma. The clotting
time (expressed in seconds) obtained for this mixture is compared to the clotting
time obtained when dilutions of normal plasma (reference plasma) are added
to the same substrate plasma. The reference plasma contains, by definition,
100% FVII activity.
Reagents
FVII-deficient substrate plasma
Given the rarity of patients with a congenital deficiency in the extrinsic pathway
factors, substrate plasmas obtained by immunodepletion are currently used to
                                       100
                     FACTOR VII ACTIVITY AND ANTIGEN
'Reference plasma'
A 'home-made' pooled normal plasma is preferable as a rule. This can be
prepared taking into account specific needs (only males or females, both,
specific age ranges, etc.), and can be stored for up to 6 months at -80 0c. By
definition it will contain 100% of the normal activity of 1 unitlml; alternatively
it can be calibrated against primary (WHO) and secondary standard (SSC)
(IU/ml).
Assay procedure
Before starting the assay, test plasma(s) and reference plasma must be thawed
at 37°C for no more than 5 min and vortexed. During the assay, plasmas must
                                       101
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
be kept at room temperature. Reconstitution of the thromboplastin reagent is
performed according to the manufacturer's instructions. Pre-warm a suitable
amount of thromboplastin at 37°C.
1. Make serial dilutions of the reference plasma (l: 10 to 1:320) and of the test
   plasma (1: 10 and 1:20) in dilution buffer. Keep these at room temperature
   and assay within 30 min.
2. Add 0.1 m1 of each dilution to 0.1 m1 of substrate plasma (undiluted), vortex
   the mixture and allow to incubate 1-2 min at 37°C.
3. Add 0.2 ml of the pre-warmed Ca-thromboplastin reagent (see reagent
   section: tissue factor reagent) and start the stopwatch. When a clearly visible
   clot forms, stop the watch. Carry out the assay duplicate (if duplicates agree
   within 5% calculate the mean). Start the run from the last dilution and, for
   replicates, from the first.
Results
Plot the results of the reference curve on two-cycle log-log graph paper with
the percentage of dilution on the abscissa and the time in seconds on the
ordinate (Figure 10.1). Connected points should yield a straight line. Devia-
tions from linearity may occur for the highest dilutions (1:160 or 1:320). If
deviation from linearity is relevant, repeat the assays. Assign 100% of FVII
activity to the 1: 10 dilution of the reference plasma. Read percentage factor
activity from the abscissa of the graph by finding the point where the clotting
100
"[
                                            '.
                                                 r~   .
                    ::!
                    3     50
                    CD
                   1ii' 40                                  0
                   1!                                              c
                          30
20
                                1.0                       10             100
                                                      % activity
Figure 10.1 FYII activity calibration curve. For example, if the clotting time of the plasma is
20 S, connect a point from the ordinate to the abscissa with a straight line and the factor VII will
correspond to 100% on the abscissa.
                                                      102
                     FACTOR VII ACTIVITY AND ANTIGEN
time obtained for the 1: 10 patient's plasma dilution intercepts the reference
curve. Evaluate only those values which fall within the straight part of the
reference curve. If the patient's clotting times do not fit with the straight part
of the reference curve, plot clotting times from other dilutions and take into
account the dilution factor in the calculation. If FVII levels are very Iowa 1:5
dilution can be tested.
Quality control
The use of control plasmas IS recommended. Control samples should be included
in each run and tested in the same manner as the test plasma samples. Each
laboratory should establish a 'normal reference range'. Compare the reference
curves obtained in different runs: if there are differences exceeding 10%, check
controls, reagents and instrument. New reference ranges should be established
for each new batch of substrate plasma, pooled normal plasma and instrument.
   Under optimal conditions intra-assay coefficient of variation (CV) is 2-4%;
inter-assay CV should also be < 5%. Normal subjects are expected to have
FVlIc levels ranging from 55% to 170%, the amplitude of this range being
related to well-known genetic and environmental determinants9 •10 .
Factor VII antigen (FYIIAg) assay was originally set up to discriminate between
the 'true' deficiencies that are due to the lack of synthesis of the factor (FYIIAg-
negative) and those variants (FYIIAg-positive ) in which the gene defect allowed
the synthesis of normal or reduced levels of a dysfunctional factor 3•5•9 • Indeed,
the assay of FVIIAg is important in characterizing FYII congenital deficiencies,
but this assay, compared to the FVlIc assay may also give us a rough idea of
the activation state of the molecule (higher levels of FVlIc) and of the 'total
FVII mass' as an index of an increased factor synthesis.
                                        103
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   One of the greatest limits of the available FVIIAg assays consists in its
sensitivity, for it is still difficult to discriminate between the absolute lack of
FVII protein and the presence of low levels of FVII protein « 4-5%).
   We will here describe the most frequentlx used method for FVIIAg assay,
which was originally described by Boyer et al. I and subsequently commercialized
as an ELISA kit by Diagnostica Stago (Asserachrome® VII:Ag).
Method outlines
The F(abh fragment of an antibody to FVII immobilized on a plastic support
captures the FVII from a plasma sample. Subsequently, another antibody
coupled with peroxidase binds to the free epitopes of FYII, forming a 'sandwich'.
The presence of the peroxidase is then revealed by the addition of a substrate,
orthophenylenediamine (OPD), in the presence of hydrogen peroxide. The
intensity of the colour (optical density, 00) appearing in the mixture has a
direct relation with the FVII concentration initially present in the plasma
sample. Results obtained with the plasma samples to be tested are compared
to the 00 values obtained with dilutions of normal plasma (reference plasma)
that were tested under the same conditions. An antigen concentration of 100%
is assigned to the reference plasma.
Reagents
1. Plastic plates coated with an antibody to FVII: are ready to use in a sealed
   envelope to be opened just before use.
2. Phosphate buffer: ready in a concentrated solution, to be diluted with distilled
   water before use with a dilution factor 1:10.
3. Washing solution: ready in a concentrated solution, to be diluted with distilled
   water before use 1:20.
4. Specific rabbit anti-FVII antiserum coupled with peroxidase: this is stored in
   a lyophilized state with stabilizers; the complex is reconstituted with phosphate
   buffer; in this state it is stable for up to 24 h at 2-8 cc.
5. Orthophenylenediamine substrate: present in tablets to be dissolved just before
   use; after dissolution H 20 2 must be added. The colourless OPD/H 2 0 2
   solution is stable for only I h at room temperature.
6. Acid solution: to stop the reaction between peroxidase and OPD/H 20 2 , 3
   moVL sulphuric acid or I moVL HCI are used.
7. Specimen collection, preparation and storage: as for FVlIc assay, citrated
   plasma has to be used; it can be assayed in fresh plasma or after storage at
   -80 cC.
8. 'Reference plasma': a 'home made' pooled normal plasma is preferable as a
   rule. It can be prepared taking into account the specific needs (males, females,
   specific age ranges, etc.), and can be stored for up to 6 months at - 80 cC.
   Alternatively it can be calibrated against WHO standard.
9. Needed equipment: multichannel pipettes, plate-washing equipment and
   plate 00 reader set at 492 nm are needed.
                                        104
                     FACTOR VII ACTIVITY AND ANTIGEN
Assay procedure
Before starting the assay, test plasma(s) and reference plasma, if frozen, must
be thawed at 37°C and vortexed.
   Patient's and reference plasma samples have to be pre-diluted 1:21 in
phosphate buffer. The diluted reference plasma is further diluted from 1:2 to
1:16 in the same buffer to prepare the reference curve.
1. FVII antigen immobilization: 200 III of patient's or reference sample are
   pipetted into the precoated wells. The strips are covered and the test mixtures
   left to incubate for 2 h at room temperature. The phosphate buffer is used
   as a blank for the assay.
2. The wells are washed five times with washing solution.
3. Immobilization of the immunoconjugate takes place by adding 200 III of
   the anti-FVII antibody linked to peroxidase. This step lasts 2 h.
4. The wells are washed five times with washing solution. This is followed
   immediately by step 5.
5. The enzymatic reaction resulting in colour development. This is started by
   the addition of the OPOIH2 0 2 substrate (200 Ill). This reaction must last
   exactly 3 min.
6. The stopping acid is added (50 III of 3 mollL H 2 S04 or 100 III of 1 mollL
   HCl) and the final well content is left to equilibrate for 10 min, then 00 is
   read at 492 nm against the blank.
Results
Plot results of the reference curve on two-cycle log-log graph paper with the
percentage of dilution of reference plasma on the abscissa and the 00 at 492
nm on the ordinate. Connected points should yield a straight line; 100% of the
antigen is assigned to the 1:21 dilution of the reference plasma. Read percentage
factor antigen from the abscissa of the graph by finding the point where the
00 obtained for the I :21 patient's plasma dilution intercepts the reference
curve. Evaluate only those values which fall within the straight part of the
reference curve.
Quality control
The use of control plasmas is recommended. They should be run periodically
and tested in the same manner as the test plasma samples. Each laboratory
should establish a 'normal reference range' for FVIIAg. Compare the slopes
of the standard curves obtained in previous runs: if there are differences
exceeding 10%, check controls, reagents and instruments. New normal ranges
should be established for each new batch of the kit and pooled normal plasma.
  Under optimal conditions the intra-assay CV is 5-7%; inter-assay CV should
be < 10%. Normal subjects are expected to have FVIIAg levels ranging from
50% to 150%, the amplitude of this range being related to well-known genetic
and environmental determinants9 ,1O.
                                       105
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Specificity and sensitivity
The specificity of the assay is good as it is insensitive to the severe deficiencies
of the other vitamin K-dependent clotting factors. Values below 2-3% have to
be considered with caution.
Acknowledgements
This work was carried out within the framework of the European Union
Concerted Action BMHI-CT94-1202 'The role of the FVII-tissue factor
pathway in ischaemic heart disease (Clotart),.
References
 I. Seligsohn U, Kasper CK, Osterud B, Rapaport SI. Activated factor VII: presence in factor
    IX concentrates and persistence in the circulation after infusion. Blood 1978; 58: 828-37.
 2. Meade TW, Mellows S, Brozovich M, Miller GJ, Chakrabarti RR, North RR, Haines AP,
    Stirling y, Imeson JD, Thompson So. Hemostatic function and ischemic heart disease:
    principal results of the Northwick Park Heart Study. Lancet 1986; 2: 533-7.
 3. Mariani G, LoCoco L, Bernardi F, Pinotti M. Molecular and clinical aspects of factor VII
    deficiency. Blood Coag Fibrinol1998; 9: S83-8.
 4. Poggio M, Tripodi A, Mariani G, Mannucci PM. Factor VII clotting assay: influence of
    different thromboplastins and factor VII-deficient plasmas. Thromb Haemostas 1991; 65:
    160-4.
 5. Mariani G, Ghirardini A, Iacopino G. Congenital deficiencies of the vitamin K dependent
    clotting factors. In: Shearer MJ, Seghatchian MJ, eds. Vitamin K and vitamin K -dependent
    proteins: analytical, physiological, and clinical aspects. Boca Raton: CRC Press; 1993; 163-93.
 6. Kario K, Matsuo T, Asada R, Sakata T, Kato H, Miyata T. The strong positive correlation
    between factor VII clotting activity using bovine thromboplastin and the activated factor VII
    level. Thromb Haemostas 1995; 73: 429-34.
 7. Pereira SP, Langley PG, Williams R. The management of abnormalities of hemostasis in
    acute liver failure. Semin Liver Dis 1996; 16: 403-14.
 8. Patrassi GM, Sartori MT, Viero M, Boeri G, Simioni P, Bassi N, Piccinni P, Girolami A.
    Protein C, factor VII and prothrombin time as early markers of liver function recovery or
    failure after liver transplantation. Blood Coag Fibrino11993; 4: 863-7.
 9. Bernardi F, Marchetti G, Pinotti M, Arcieri P, Baroncini C, Papacchini M, Zepponi E, Ursicino
    N, Chiarotti F, Mariani G. Factor VII gene polymorphisms contribute about one third of the
    FYII level variation in plasma. Arterieroscl Thromb Vasc Bioi 1996; 16: 72-f1.
10. Mariani G, Bernardi F, Bertina R, Vincente Garcia V, Pridz H, Samama M, Sandset PM, Di
    Nucci GD, Testa MG, Bendz B, Chiarotti F, Ciarla MY, Strom R. for the European Community
    Concerted Action 'Clotart'. Serum phospholipids are prominent determinants of Factor VII
    levels in the most common FVII genotype. Thromb Haemostas (In press).
II. Boyer C, Wolf M, Rothschild C, Migaud M, Amiral J, Mannucci PM, Meyer D, Larrieu MJ.
    An enzyme immunoassay (ELISA) for the quantitation of human factor VII. Thromb
    Haemostas 1986; 56: 250--5.
                                               106
11
Factor VIII clotting activity
P. M. MANNUCCI and A. TRIPODI
INTRODUCTION
Factor VIII activity has been historically measured with two types of methods:
the one-stage assay, based on the activated partial thromboplastin time (APTf)
(Chapter 5) and the two-stage assay, based on the thromboplastin generation
test. The relative merits of the two assays have been debated 1•2 . In general,
results obtained on the same samples are similar; however, discrepancies
can be seen in plasmas which contain activated factors of the coagulation
cascade. These factors make the clotting times obtained with the one-stage
assay shorter than expected on the basis of the factor VIII content, resulting
in a significant overestimation of the factor VIII activity. For the same reason,
the one-stage assay is often unsuitable to determine factor VIII in concentrates.
Even though the two-stage assay does not seem to suffer from the same
drawback, it is difficult to perform because the reagents employed need a high
degree of standardization difficult to achieve in less specialized clinical
laboratories.
   More recently, new two-stage methods which employ chromogenic substrate
have been proposed for the measurement of factor VIU 3•4 ; commercial kits are
available from various manufacturers (Chromogenix; Dade Behring; Diagnostica
Stago) and some of them have been evaluated and found suitable for clinical
application 5•6 • However, to our knowledge, their use is still limited and more
extensive clinical validation is probably needed. For this reason we have chosen
to discuss here the one-stage assay. More information on the other assays can
be found in the cited references. Before giving details of the method, we shall
provide a brief account of the general principles of bioassays on which factor
VIII and other clotting factor assays are based.
                                      107
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
ASSAY CHARACTERISTICS AND PRINCIPLES
Test system
The theory of the bioassay implies that when two samples at different factor
VIII activities (test and reference) are processed at different doses (dilutions)
with the specific assay system (APTT), this will give different responses (clotting
times). If those responses are plotted against the corresponding log-transformed
factor VIII activity one should obtain a pair of dose-response curves which
are parallel throughout their length. The horizontal distance between the two
curves is a measure of the potency ratio between the two samples (Figure 11.1).
It is important to emphasize that parallelism of the two curves is a prerequisite
for correct estimation of factor VIII activity. The occurrence of substantial
deviation from parallelism means that the two samples are not treated in
the same way by the system. In most cases deviation from parallelism is
likely to be due to the presence of an inhibitory substance in patient plasma
or to the deficiency in the substrate plasma of factor(s) other than that to be
measured.
    This situation may occur, for instance, when substrate plasma used for the
assay has lost the activity of the labile factor V during prolonged and/or poor
storage conditions. To establish parallelism, patient and reference plasma must
be run at several dilutions. Hence, the simplified and widely adopted procedure
of testing a complete dose-response curve for reference plasma and only one
dilution of the test plasma must be regarded as incorrect. As mentioned above,
the system must be deficient in the factor to be measured. The most obvious
and cheap solution to this problem is the plasma from a patient with severe
haemophilia A (factor VIII clotting activity < 1 U/dl) with no antibody to
factor VIII. This solution is, however, less and less practicable because the
increasing risk of transmission of virus-related infections makes the majority
of severe haemophilic patients unsuitable for the purpose. A valid alternative
is to rely on factor VIII-deficient plasmas obtained by artificial depletion of
normal plasma by means of monoclonal antibody against factor VIle.
Commercial immunodepleted plasmas are also available (e.g. Ortho, Diagnostica
Stago).
                                       108
                            FACTOR VIII CLOniNG ACTIVITY
80
          70
  u
  CD
  <II
  CD      60
   E
  :;:;
   C>
   c:
  :;:;    50
  ....0
  0
                                                                                             Reference
          40
                4                10           20               50          100
                      Log    factor VIII activity (U/dl)
Figure 11.1 Graphical calculation of factor VllI clotting activity. The dilutions for the reference
and patient plasmas were 1:10,1:50 and 1:250. According to the graphical calculation (see text),
the factor VIII activity for the patient plasma is 57 U/dl of the reference; if the potency of the
reference calibrated against the international standard is 0.85 then the final result will be 57 x 0.85
= 48 IU/di. Should the patient plasma be tested at higher or lower dilution than the reference, the
results must be multiplied or divided by the appropriate dilution factor.
Reference plasma
A convenient source of reference plasma is plasma pooled from a suitable
number of healthy donors. It is important to note that plasma pooled from an
insufficient number of donors might be not representative of the average value
in the normal population. Hence, the assignment of an arbitrary potency of
100 Uldl may lead to important under- or overestimations of factor VIII levels
in the test sample.
   Due to the large biological variability in a normal population (factor VIII
in normals varies from 50 to 150 U/dl), the adequacy of a pooled plasma in
reflecting the average normal content of factor VIII is largely dependent on
the number of healthy donors selected for the preparation. Fifty or more donors
seem to be an adequate number. However, collecting such a large number of
donors might be impracticable and not strictly necessary. At least in theory
plasma from a single donor is suitable as reference plasma, provided it is
calibrated against the international standard for factor VIII/von Willebrand
factor, which is available upon request from the National Institute for Biological
Standards and Control (NIBSC). The calibration is made by assaying the local
                                                   109
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
reference plasma against the international standard. To minimize error it is
important to carry out at least four independent assays which involve a complete
fresh set of the international standard and test dilutions each time. Since the
supply of standard is often limited, a suitable alternative might be to perform
two assays from the same vial of the reconstituted standard. The potency of
the local plasma is eventually obtained by multiplying the calculated potency
by that of the International Standard.
METHOD
Due to the large variation in sensitivity to factor VIII among commercial AP1T
reagents, it is impossible to give technical details which apply to all. As a general
rule each laboratory should establish the best conditions with the chosen re-
agent. For instance, it is advisable to make a complete dose-response curve by
testing several dilutions of reference plasma. The first dilution should be
sufficiently high to avoid interference with other plasma components; usually,
a 1:5 or 1: 10 dilution is suitable to start with and the other dilutions should be
spaced by the same factor (i.e. for a factor of five, 1:10, 1:50, 1:250, 1:1250,
etc.). It is advisable to use the same dilution interval for standard and patient
plasma alike. The last dilution to be tested depends on the reagent and substrate
plasma used; usually it will be the one whose clotting time is close to, but yet
shorter than, that of the 'blank' (Le. sample replaced by buffer). For choosing
the most suitable standard dilutions it is advisable to inspect the dose-response
curve and choose at least three dilutions which lie on the steeper and linear
part of the curve.
   The dilutions of patient plasma should be chosen in accordance with the
expected factor VIn activity. In general the patient clotting times should be as
close as possible to, but yet longer than, those of the reference plasma. Therefore
if the standard is run at 1:10, 1:50, and 1:250, the patient plasma will be tested
either at 1:5,1:25 and 1:125 or 1:20, 1:100 and 1:500 when lower or higher-
than-normal factor VII1 activity is expected. To avoid the time-trend deterioration
of samples and reagents, it is advisable to take replicate readings of each
dilution in a balanced order (i.e. starting from the first dilution of the standard
to the last of the patient samples and the replicate in the opposite order).
   The estimation of the potency of a patient sample relative to the standard
can be done by using a graphical or mathematical solution. The latter is
preferable because it avoids the bias in fitting the dose-response curves and
gives objective criteria to assess for linearity and parallelism. Simple calcula-
tions can be performed by pocket calculator as described by Ingram8 • Com-
prehensive statistical advice on writing computer programs for bioassay are
beyond the scope of this chapter, and can be found in the published literature9--11 .
For the graphical calculation the mean values of the replicate readings for each
dilution are plotted (vertical axis) against the log-transformed percentage
activity (horizontal axis), the first dilution of the reference and patient plasmas
being arbitrarily taken as 100 Uldl activity (Figure 11.1). After drawing the
best-fit lines through the data points and checking for parallelism, the activity
of factor VIII in the patient plasma is derived by drawing a horizontal line
                                        110
                      FACTOR VIII CLOTTING ACTIVITY
from the point where the patient curve intercepts the 100 U/dl activity to the
point where it crosses the reference curve. From that point a vertical line is
then dropped to intercept the horizontal axis. This value represents the
percentage activity of factor VIII in the patient plasma relative to the reference
(Figure 11.1). When patient plasma is tested at a higher or lower dilution than
the standard, the result must be multiplied or divided by the appropriate dilution
factor.
   The procedure outlined below is intended to fit the characteristics of the
commercial APTT reagent and coagulometer currently used in our laboratory.
Other preparations can be successfully used provided the optimal conditions
are chosen as recommended above.
Reagents
1. Automated APTT (Organon Teknika). This reagent contains micronized
   silica as activator and rabbit brain phospholipids as platelet substitute.
2. Imidazole buffer is prepared by dissolving 3.4 g of imidazole and 5.85 g of
   NaCI in approximately 70 ml distilled water; 18.6 ml of 1 moVL HCI is then
   added, the pH adjusted to 7.3-7.4 and the final volume adjusted to 100 ml.
   This stock solution is stored at 4 DC and used to prepare the working solution
   by diluting it 1:10 with distilled water.
3. CaCl2 0.025 moVL.
4. Substrate plasma with factor VIII content < 1 U/dl.
5. Reference plasma calibrated against the international standard for factor
   VIII/von Willebrand factor.
Equipment
1. Plastic tubes to make dilutions.
2. Automated micropipettes equipped with plastic tips.
3. Photo-optical or mechanical coagulometer (Coag A Mate X 2, Organon
   Teknika, or other commercial brand). The procedure described can also be
   carried out with the manual tilt-tube technique.
Procedure
1. Three dilutions (1:10, 1:50 and 1:250) of the reference and patient plasma
   should be prepared immediately before starting the assay. As mentioned
   above. the actual dilution of the patient plasma will depend on the expected
   factor VIII activity, a rough idea thereof being obtained by the APTT value
   of the test plasma.
2. 100 ~l of each dilution is pipetted into the wells of the coagulometer in
   balanced order (see above).
3. Add 100 ~ of substrate plasma.
4. Pipette (or program the coagulometer to do so) 100 ~l of APTT reagent.
                                       111
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
5. Incubate the mixture for exactly 5 min (if other reagent is used, follow the
   instructions of the manufacturer).
6. At the end of the incubation add 100 ~ of CaC12 0.025 moVL.
7. Record the clotting time which will be used for the calculation of factor
   VIII activity (see above).
NORMAL VALUES
Factor VIII clotting activity in the normal population varies between 50 and
150 U/dl, it is recommended that each laboratory establish its own normal
range.
References
 I. Kirkwood TBL, Rizza CR, Snape TJ, Rhymes IL, Austen DEG. Identification of sources of
    interlaboratory variation in factor VIII assay. Br J Haematol1981; 37: 559-68.
 2. Nilsson 1M, Kirkwood TBL, Barrowcliffe Tw. In vivo recovery of factor VIII: comparison
    of one-stage and two-stage assay methods. Thromb Haemostas 1979; 42: 1230--9.
 3. Segatchian MJ, Miller-Andersson M. A colorimetric evaluation of factor VIII: C. Med Lab
    Sci 1978; 35: 347-54.
 4. Rosen S, Andersson M, Mikaelsson M, Oswaldsson U. Determination of factor VIII activity
    with chromogenic substrate kit method. I. Basic performance. Thromb Haemostas 1984; 40:
    139-45.
 5. Rosen S, Andersson M, Blombiick M et al. Clinical application of a chromogenic substrate
    method for the determination of factor VIII activity. Thromb Haemostas 1985; 54: 818-23.
 6. Tripodi A, Mannucci PM. Factor VIII activity as measured by an amidolytic assay compared
    with a one-stage clotting assay. Am J Clin Patho11986; 86: 341-4.
 7. Takase T, Rotblat F, Goodall AH, KemotT PBA, Middleton S, Chand S, Denson KW, Austen
    DEG, Tuddenham EGD. Production of factor VIII deficient plasma by immunodepletion
    using three monoclonal antibodies. Br J Haematol1987; 66: 497-502.
                                            112
                          FACTOR VIII CLOniNG ACTIVITY
 8. Ingram GIC. Blood coagulation factor VIII: genetics, physiological control and bioassay. In:
    Sobotka H, Stewart CP, eds. Advances in clinical chemistry, vol 8. New York: Academic Press,
    1965; 189-236.
 9. Counts RB, Hayes JE. A computer program for analysis of clotting factor assays and other
    parallel-line bioassay. Am J Clin Patho11979; 71: 167-7l.
10. Williams KN, Davidson JMF, Ingram GIe. A computer program for the analysis of para11el-line
    bioassays of clotting factors. Br J Haematol1975; 31: 13-23.
11. Kirkwood TBL, Snape TJ. Biometric principles in clotting and clot lysis assays. Clin Lab
    Haemato11980; 2: 155-67.
12. Koster T, Blann AD, Briet E, Vandenbroucke JP, Rosendaal FR. Role of clotting factor VIII
    in effect of von Willebrand factor on occurrence of deep-vein thrombosis. Lancet 1995; 345:
    152-5.
                                              113
12
Von Willebrand factor
P. M. MANNUCCI and R. COPPOLA
INTRODUCTION
The method is used on the sandwich principle. In the first step, plastic wells of
rnicrotitre plates are coated with antibodies against vWF (capture antibody).
In the second step, suitable dilutions of test and standard plasmas are incubated
to allow vWF antigen to bind the capture antibody. After washing the wells
                                      115
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
to remove the unbound antigen, a second antibody to vWF labelled with
horseradish peroxidase (detecting antibody) is added. This results in the
formation of a sandwich complex in a quantity proportional to the vWF
content in patient and standard plasmas. After a second washing to remove
excess detecting antibody, the peroxidase activity is measured photometrically
by addition of hydrogen peroxide and o-phenylenediamine. The optical density
readings are then converted into U/dl of vWF antigen by comparison with a
calibration curve obtained by processing dilutions of a pooled normal plasma
or standard plasma.
METHOD
Reagents
1. Coating buffer: 50 mmoVL Tris-HC1, pH 9.00.
2. Over-coating buffer: as for coating buffer, added with 3% bovine serum
   albumin.
3. Washing buffer: 20 mmollL Tris-HC1, 100 mmollL NaCl, 0.1% Tween, 0.1%
   bovine serum albumin, pH 7.4.
4. Dilution buffer: as for washing buffer, with added 0.5% bovine serum
   albumin.
5. Substrate buffer: 22 mmollL trisodium citrate, 50 mmollL Na2HP04' Adjust
   pH to 5.5 with H 3P04.
6. 400 ~glml o-phenylenediamine (Sigma).
7.30%H20 2·
S. 3 moVL H 2S04,
9. Two different monoclonal antibodies were used as capture and detecting
   antibodies.
Immediately before use, prepare a solution of o-phenylenediamine 40 mg and
H 20 2 40 ~ and make the volume to 100 ml with substrate buffer.
Procedure
The detecting antibody was conjugated with horseradish peroxidase (Sigma~
(final concentration 250 ~glml) according to the method of Nakane and Kawaoi
                                     116
                              VON WILLEBRAND FACTOR
and stored in small aliquots at -20°C in the presence of glycerol (50% final
concentration). Polystyrene plates (Nunc) are coated overnight at 4 °C with
125 p,]Jwell of a 2.5 p,g1ml solution of capture antibody in coating buffer. At
the end of the incubation the solution is discarded and the wells are overcoated
with 200 p,l of overcoating buffer for 1 or 2 h. The wells are then washed three
times with washing buffer and added with 100 p.]Jwell of standard plasma (in
duplicate) diluted from 1:50 to 1:3200 in dilution buffer. Patient samples are
tested at two different dilutions chosen according to the expected concentration
of vWF in the sample (usually, 1:100 and 1:400). After incubation for 2 h (or
overnight) at room temperature, the wells are washed three times and added
with 100 ~]Jwell of detecting antibody (5 p,g1ml) in dilution buffer. After
incubation for 1 h at room temperature, the wells are washed three times and
added with 100 p,]Jwell of o-phenylenediamine and H 2 0 2 solution. After
incubation for 10 min at room temperature (dark), the reaction is stopped by
adding 50 p,]Jwell H 2S04 , The optical density is then measured at 486 nm with
a microplate photometer. vWF antigen in patient samples is derived by
extrapolation from the calibration curve (Figure 12.1).
   Results obtained with the above method in plasma samples from healthy
subjects and patients with von Willebrand disease compared favourably with
those obtained with the commercial kit Asserachrom vWF (Diagnostica Stago),
which can be used as a suitable alternative.
REFERENCE PLASMA
As mentioned for factor VIII clotting assay, plasmas from a large number of
healthy donors must be pooled in order to get a standard truly representative
                                                                                   ,.
                                                                                   III
                                                                                   en
                                                                                   o
                                                                             2.0 ::a
                                                                                   ,.z
                                                                                   III
                                                                                   ~
                                                                         1.5
                                                                                   ,.....
                                                                         1.0 :
                                                                                   ell
                                                                                   :::I
                                                                                   3
                                                                             0.5
Figure 12.1 Plasma dilution curve of vWF antigen in pooled normal plasma
                                            117
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
of the average normal plasma and hence suitable to construct the calibration
curve for vWF antigen assay. Fifty or more healthy donors, equally divided
between males and females and covering different age classes, are adequate. As
an alternative a pooled plasma from a limited number of donors (even from a
single donor) can be used, provided such plasma is first calibrated against the
International Standard for factor VIII and vWF (National Institute for Bio-
logical Standards and Control). For details on the calibration procedure, see
Chapter 11.
REFERENCE RANGE
vWF antigen in the normal population varies between 50 and 150 D/dI. However,
age group- and blood group-related differences have been reported within the
same population8 ,9. In general, vWF antigen increases with increasing age,
and blood group 0 values are on average lower than those of non-group O.
Accordingly, it is strongly recommended that each laboratory establish its own
normal range.
References
 1. MacFarlane DE, Stibbe J, Kirby EP, Zucker MB, Grant RA, McPherson 1. A method for
    assaying von Willebrand factor (ristocetin cofactor). Thromb Diath Haemorrh 1975; 34:
    306--8.
 2. Laurel CB. Quantitative estimation of proteins by electrophoresis in agarose gel containing
    antibodies. Anal Biochem 1966; 15: 45-52.
 3. Zimmerman TS, Hoyer LW, Dickson L, Edgington TS. Determination of the von Willebrand's
    disease antigen (factor VIII related antigen) in plasma by quantitative immunoelectrophoresis.
    J Lab Clin Med 1975; 86: 152-9.
 4. Bartlett A, Dormandy KM, Hawkey CM, Stableforth P, Voller A. Factor VIII-related antigen:
    measurement by enzyme immunoassay. Br Med J 1976; I: 994-6.
 5. Silveira AMV, Yamamoto T, Adamson L, Hessel B, Blomback B. Application of an enzyme-
    linked immunosorbent assay (ELISA) to von Willebrand factor (vWF) and its derivatives.
    Tbromb Res 1986; 43: 91-102.
                                              118
                              VON WILLEBRAND FACTOR
 6. Federici AB, Tombesi S, Coppola R, Colibretti ML, Gobbi A, Albertini A, Mannucci PM.
    Preliminary results on the characterization of monoclonal antibodies to von Willebrand factor
    (vWF). Abstract of the International Symposium on Biotechnology of plasma proteins:
    Haemostasis, thrombosis and iron proteins. Florence, 9-11 April 1990.
 7. Nakane PK, Kawaoi PA. Peroxidase-labeled antibody: a new method of conjugation. J
    Histochem Cytochem 1974; 22: 1084-91.
 8. Cox Gill J, Endres-Brooks J, Bauer PJ, Marks WJ, Montgomery RR. The effect of ABO blood
    group on the diagnosis of von Willebrand disease. Blood 1987; 69: 1691-5.
 9. Rodeghiero F, Castaman G, Dini E. Epidemiological investigation of the prevalence of von
    Willebrand's disease. Blood 1987; 69: 454-9.
10. Ruggeri ZM, Zimmerman TS. von Willebrand factor and von Willebrand disease. Blood 1987;
    74: 895-904.
                                              119
13
Antithrombin activity and antigen
J. CONARD
INTRODUCTION
Citrated blood is used: nine volumes of blood are collected into tubes containing
one volume of sodium citrate 0.11 or 0.129 moUL. After centrifugation at
about 2000g for 15-20 min, aliquots of 0.5 ml of plasma are transferred to
plastic tubes that are stoppered and frozen at -20°C or lower. They can be
stored up to 4 months before assay.
   At the time of the test, samples are rapidly thawed at 37°C. Lipaemic
plasmas may not be suitable for nephelometric assays.
ACTIVITY ASSAVS3 ,4
Antithrombin Inhibition
Principle
Plasma AT is determined as the capacity for inactivating thrombin in the
presence of heparin which has a catalytic effect on the reaction AT-thrombin.
An excess of purified thrombin is added to the plasma. The amount of thrombin
inhibited by the complex AT-heparin depends on the amount of AT in the
sample. Residual thrombin hydrolyses a thrombin-sensitive chromogenic
substrate and para-nitroaniline (PNA) released is measured at 405 nm. The
method can be summarized as follows:
1. AT + heparin in excess              ~    [AT-heparin]
2. [AT-heparin] + thrombin             ~    [AT-heparin-thrombin] + residual
   in excess                                thrombin
3. Substrate + residual thrombin       ~    pNA (405 nm).
Assay procedure
Bovine thrombin is more appropriate than human thrombin so that the test is
specific for AT, by avoiding the interference of heparin cofactor 115,6.
   An end-point assay has been used in manual methods but kinetic automated
assays are preferred for better reliability7,8. Many instruments are suitable and
most of the automates used in coagulation laboratories are adapted for this
test: Diagnostica Stago, Dade-Behring, Instrumentation Laboratory, Bio-
Merieux, for instance.
   Different substrates are used: S-2238 Chromogenix, CBS 3447 Stago.
   The AT determination can also be performed in some fully automated instru-
ments such as the ACA Du Pont. This is a 'closed' system requiring both the
                                      122
                   ANTITHROMBIN ACTIVITY AND ANTIGEN
instrument and special packs. The principle is the same but the substrate is different.
The instrument and reagents are remarkably stable, allowing the performance of
the calibration only when changing pack batches. Pathological and normal
lyophilized control plasmas are checked regularly, about once per week.
Anti-Xa inhibition
Factor Xa, like bovine (but not human) thrombin, has the advantage of being
more specific for AT, since it is not inhibited by heparin cofactor II. For this
reason an AT chromogenic assay based on factor Xa inhibition has been
developed9 .
   The principle of the test is similar to the antithrombin inhibition test but
the substrate is different: 8-2765 Chromogenix.
ANTIGEN ASSAYS
Principle
Immunoprecipitation is performed in a liquid phase by adsorption on latex
particles. A laser nephelometer measures light scattered in a forward direction
by immunocomplexes in an incident beam of monochromatic light. The diffused
light is measured in an angle between 13° and 24°. The reading is performed
at 840 nm.
Instrument
The Behring Nephelometer Analyser (BNA) is an automated instrument, con-
venient to perform large series of AT determinations (45 cuvettes divided into
nine groups of five cuvettes each).
Assay procedure
After the plasmas have been introduced into the cuvettes, all steps are automatic.
In lipaemic or lyophilized plasmas, AT determination may not be possible due
                                         123
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
to the turbidity; in this situation the result is marked with a cross. Haemolysis
does not interfere with the result.
REFERENCE VALUES
CONGENITAL DEFICIENCY IN AT
Type!
Type II
  RS                                             Normal         ~
  HBS                                            Normal         Normal
  PE                                             Limit normal   ~
ACQUIRED DEFICIENCY
Pathological conditions
  Liver cirrhosis
  Nephrotic syndrome
  Postoperative period
  Disseminated intravascular coagulation
Treatments
  Heparin
  Hormones: (a) oral contraceptives (contain ethinyloestradiol); (b) replacement therapy
    with conjugated equine oestrogens or oral oestradiol
  L-Asparaginase
   factors and inhibitors synthesized in the liver. This deficiency is not usually
   associated with thrombosis.
2. Nephrotic syndrome: when the urinary loss of AT exceeds its synthesis, a
   plasma deficiency is observed and, together with increased levels of co-
   agulation factors, it might be involved in the increased risk of thrombosis
   observed in this disease.
3. Postoperative period: a decrease has been reported after knee surgery, and
   since this type of surgery is highly thrombogenic, some authors have proposed
   AT concentrates in association with heparin24.
4. Disseminated intravascular coagulation (DIe): AT is inconstantly decreased
   and it cannot be used as a criterion for the diagnosis of DIe. The decrease
   is more frequent in patients with DIC related to cancer with liver metastases.
   In severe sepsis, studies have shown an improvement of the biological signs
   of DIC after administration of AT concentrates but the effect on mortality
   was very moderate or absent.
CONCLUSION
References
 1. Abildgaard U. Antithrombin and related inhibitors of coagulation. In: Poller L, ed. Recent
    advances in blood coagulation. Edinburgh: Churchill Livingstone, 1981; 151.
 2. Rosenberg RD, Bauer KA. The heparin-antithrombin system: a natural anticoagulant
    mechanism. In: Colman RW, Hirsch J, Marder VJ, Salzman EW, eds. Hemostasis and
    Thrombosis: basic principles and clinical practice. Philadelphia: Lippincott, 1994; 837--60.
 3. Handeland GF, Abildgaard U, Aasen AO. Simplified assay for antithrombin III activity using
    chromogenic peptide substrate. Manual and automated method. Scand J Haematol1983; 31:
    427-36.
 4. Odegard OR, Abildgaard U. Antithrombin III: critical review of assay methods. Signification
    of variations in health and disease. Haemostasis 1978; 7: 127-34.
 5. Conard J, Horellou MH, Bara L. Bovine or human thrombin in antithrombin III (AT III)
    amidolytic assays. Thromb Haemostas 1985; 54: 25.
 6. Friberger P, Egeberg N, Holmer E, Hellgren M, Blombiick M. Antithrombin assay. The use
    of human or bovine thrombin and the observation of a second heparin cofactor. Thromb Res
    1982; 25: 433--6.
 7. Conard J. Automation of antithrombin III. Methods on routinely available instruments. Sem
    Thromb Haemostas 1983; 9: 263-7.
 8. Odegard OR, Rosenlund B, Ervik E. Automated antithrombin III assay with a centrifugal
    analyzer. Haemostasis 1978; 7: 202-9.
 9. Demers D, Henderson P, Blajchman MA, Wells MJ, Mitchell L, Johnston M, Ofosu FA,
    Fernandez-Rachubinski F, Andrew M, Hirsch J, Ginsberg JS. An antithrombin III assay based
    on factor Xa inhibition provides a more reliable test to identify congenital antithrombin III
    deficiency than an assay based on thrombin inhibition. Thromb Haemostas 1993; 69: 231-5.
10. Parvez Z, Fareed J, Argawal P, Messmore HI, Moncada R. Laser nephelometric quantitation
    of antithrombin III (AT III). Thromb Res 1981; 24: 367-37712.
II. Peters M, Jansen E, Ten Cate.Jw, Kahle LH, Ockelford P, Breederveld C. Neonatal antithrombin
    III. Br J Haematol1984; 58: 579-87.
12. Teger-Nilsson AC. Antithrombin in infancy and childhood. Acta Paediatr Scand 1975; 64:
    624-8.
13. Weenink GH, Treffers PE, Kahle LH, Ten Cate JW. Antithrombin III in normal pregnancy.
    Thromb Res 1982; 26: 281-7.
14. Meade T, Dyer S, Howard DJ, Imeson JD, Stirling Y. Antithrombin III and procoagulant
    activity: sex differences and effects of the menopause. Br J Haematol1990; 74: 77-81.
15. Tait RC, Walker 10, Davidson JF, Islam SIA, Mitchell R. Antithrombin III activity in healthy
    blood donors: age and sex related changes and the prevalence of asymptomatic deficiency.
    Br J Haemato11990; 75: 141-2.
16. Thaler E, Lechner K. Antithrombin deficiency and thromboembolism. In: Prentice CRM, ed.
    Qinics in haematology. London: Saunders, 1981; 369-90.
17. Lane DA, Bayston T, 01ds RJ, Fitches AC, Cooper DN, Millar DS, Jochmans K, Perry DJ,
    Okajima K, Thein SL, Emmerich J. Antithrombin mutation database: 2nd (1997) update.
    Thromb Haemostas 1997; 77: 197-211.
                                              127
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
18. Finazzi G, Caccia R, Barbui T. Different prevalence of thromboembolism in the subtypes of
    congenital antithrombin III deficiency: review of 404 cases. Thromb Haemostas 1987; 58:
    1094.
19. Sas G, Pepper DS, Cash ID. Further investigations on antithrombin III in the plasma of
    patients with the abnormality antithrombin 'Budapest'. Thromb Diathes Haemorrh 1975; 33:
    564-72.
20. Krudler JW, Strebus AF, Meinders AB, Briet E. Anticoagulant effect of unfractioned heparin
    in antithrombin-depleted plasma in vitro. Haemostasis 1996; 26: 85-9.
21. Lechner K, Kyrle PA. Antithrombin III concentrates. Are they clinically useful? Thromb
    Haemostas 1995; 73: 340-8.
22. Conard J, Horellou MH, Van Dreden P, Lecompte T, Samama M. Thrombosis and pregnancy
    in congenital deficiencies in AT III, protein C or protein S: study of 78 women. Thromb
    Haemostas 1990; 63: 319-20.
23. Beresford CH. Antithrombin III deficiency. Blood 1998; 2: 239-50.
24. Francis ChW, Pellegrini VD Jr, Stulberg BN, Miller ML, Marder VJ. Prevention of venous
    thrombosis after total knee arthroplasty. J Bone Joint Surg 1990; 72A: 976-82.
25. Conard J, Samama M, Basdevant A, Guy-Grand B, de Lignieres B. Differential AT III
    response to oral and parenteral administration of 17~-estradiol. Thromb Haemostas 1983;
    49: 252.
26. Caine YG, Bauer KA, Barzegar S, Ten Cate H, Sacks FM, Waish BW, Schiff I, Rosenberg
    RD. Coagulation activation following estrogen administration to postmenopausal women.
    Thromb Haemostas 1992; 68: 392-5.
27. Scarabin PY, Alhenec-Gelas M, Plu-Bureau G, Taisne P, Agher R, Aiach A. Effects of oral
    and transdermal estrogen/progesterone regimens on blood coagulation and fibrinolysis in
    postmenopausal women. A randomized controlled trial. Arterioseler Thromb Vase Bioi 1997;
    17: 3071-8.
28. Bounameaux H, Duckert F, Walter M, Bounameaux Y. The determination of antithrombin
    III. Comparison of six methods. Effect of oral contraceptive therapy. Thromb Haemostas
    1978; 39: 607-15.
                                            128
14
Protein C activity and antigen
R. M. BERTINA
INTRODUCTION
Physiological role
 Activated protein C is the key enzyme of the protein C anticoagulant pathway.
 Its anticoagulant properties reside in its capacity to inactivate the cofactors Va
 and VIlla by proteolytic degradation4. Protein S - another vitamin K-dependent
 plasma protein, anionic phospholipid membranes and Ca2 + ions are essential
 cofactors for the inactivation reactions. By inactivating the cofactors Va and
 VllIa6 •7 , activated protein C will dramatically reduce the rate of thrombin
 formation, as can be visualized by the increase in the activated partial thrombo-
 plastin time (AnT) after addition of APC to pooled normal plasma.
    The activation of protein C occurs at the endothelial surface by the thrombin-
 thrombomodulin complex8 • Thrombomodulin is a membrane protein which
.serves as a receptor/cofactor for thrombin. Binding of thrombin to throm-
 bomodulin changes the enzymatic properties of thrombin from a procoagulant
 enzyme (activation of fibrinogen and platelets) to an anticoagulant enzyme
 (activation of protein C). During activation an Arg-Leu bond is cleaved, after
                                       129
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Pathophysiological aspects
Hereditary defects
The importance of the protein C anticoagulant system for the control of
thrombin formation in vivo is most dramatically demonstrated in neonates who
are homozygotes or double heterozygotes for hereditary protein C deficiencyJ3,14.
These patients develop massive and fatal (if untreated) thromboembolism
shortly after birth. The most prominent clinical manifestations are large purpuric
skin lesions (purpura fulminans syndrome), central nervous system thrombosis
and central ophthalmic thrombosis.
   Interestingly the clinical phenotype of heterozygotes for a hereditary protein
C deficiency is not uniform. Two different phenotypes can be recognized. In
the clinically dominant type of protein C deficien? heterozygotes have an
increased risk of developing venous thrombosisl5--1 . Families with this type
of protein C deficiency are identified by screening symptomatic patients with
unexplained familial thrombophilia: about 5% of these patients are protein
C_deficient I8 ,19. The prevalence of this phenotype in the general population
has been estimated to be 1116000 19 .
   In the clinically recessive type of protein C deficiency heterozygotes only
very rarely have thrombosis20 . Families with this type of protein C deficiency
are found among the parents of homo~gous protein C-deficient patients but
also among healthy blood donors 13,20-2 . The prevalence of this phenotype in
the general population has been estimated to be 1130021 ,22.
   This apparent discrepancy is explained by the fact that familial thrombophilia
is a multigenic disorder. This means that in the clinically dominant type of
protein C deficiency, apart from the protein C gene defect, other genetic defects
are segregating that contribute to the thrombotic risk. This was first demonstrated
by Koeleman et al. 23 for thrombophilic families in which both a protein C gene
mutation and the factor V Leiden mutation were segregating.
   The laboratory diagnosis of hereditary protein C deficiency focuses on the
identification of homozygotes/double heterozygotes and heterozygotes of the
clinically dominant form of the disorder. If not on oral anticoagulant treatment
protein C (activity or antigen) levels should be lower than the lower limit of
                                       130
                     PROTEIN C ACTIVITY AND ANTIGEN
the nonnal range, while the plasma concentration of other vitamin K-dependent
proteins is within the normal range 24 . The treatment of patients with oral
anticoagulants offers a special problem for laboratory diagnosis because the
treatment in itself causes a decrease in the plasma protein C level (dependent
on the intensity of the treatment). For typical therapeutic doses of coumarins
protein C antigen and activity decrease to about 50% and 25%, respectively24·25.
   Therefore, in these patients, protein C levels should be below the lower limit
of the range observed at the relevant intensity of treatment, while the ratios
PC/FII, PC/FX and/or PC/FYII should also be lower than normally observed
in these patients24.
Acquired abnormalities
Abnormal protein C levels have also been reported in a variety of clinical con-
ditions. Increased protein C levels have been reported for users of oral contracep-
tives26, for ~atients with (acute) angina pectoris27, and patients with a nephrotic
syndrome2 ,29. Reduced levels of protein C have been reported for patients in
the postoperative period30 and for patients with liver disease 31 , various forms
of disseminated intravascular coagulation (DIC)32, insulin-dependent
diabetes33,34, essential hypertension 35 and sickle-cell disease 36. Sometimes only
a decrease in specific activity or protein C activity/protein C antigen ratio is
observed30. In general it is not clear whether the reduced protein C levels trigger
or provoke the development of thrombosis in such patients.
   Further, it is important to realize that protein C levels increase with age
(about 4% per decade)37 and that treatment protocols might also influence
protein C levels. Danazol and stanozoloes,39, for instance, have been reported
to result in an increase in plasma protein C levels, while treatment with
L-asparaginase40 or oral anticoagulants41 will result in a decrease of plasma
protein C levels.
Protein C assays
Two different types of assay can be used to measure the concentration of
protein C in plasma: immunological assays and functional assays (for reviews
see ref. 421. The immunological assays include electroimmunoassays16,43,
ELISA44,4 and radioimmunoassays46,47. Functional assays include a variety
of methods differing in concept, specificity and complexity48. All these assays
differ in sensitivity, precision, accuracy and costs. The selection of a protein
C assay for use in the daily routine therefore strongly depends on the infra-
structure of the local clinical laboratory.
Protein C antigen
Initially the recommended method for measuring protein C antigen was the
electroimmunoassay (Laurell or rocket assay) using a locally prepared rabbit
                                       131
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
antiserum against human protein CiS. To date anti-protein C serum is com-
mercially available from many different manufacturers; some manufacturers
even provide ready-to-use Laurell plates for the measurement of protein C
antigen (Diagnostica Stago, American Diagnostica).
Principle of electroimmunoassay
A fixed amount of sample is allowed to migrate in an electric field in an agarose
gel containing specific anti-protein C antibodies at alkaline pH (8.8). Negatively
charged proteins such as protein C will migrate towards the anode. The protein
C will interact with the specific rabbit anti-protein C antibodies. These interac-
tions will result in the formation of insoluble immunoprecipitates (precipita-
tion peaks or rockets) after equilibrium has been established. The length of the
precipitation rocket is directly related to the concentration of protein C in the
plasma. The protein C antigen concentration in a test sample is determined by
measuring the length of the precipitation rocket and reading the antigen content
from a calibration curve which relates length of the rocket to the protein C
antigen content.
Calculations
The assay is calibrated with dilutions of a pooled normal plasma (1/1, 1/2, 1/4,
1/8) in electrophoresis buffer. Usually the potency of this standard is arbitrarily
set to 100% or 1 unitlml (1 unit being the amount of protein C present in 1 ml
of pooled normal plasma). Some of the commercial standards have been
calibrated against the first international standard for protein C in plasma which
contains 0.82 IU per ampoule49 .
   The length of the rockets obtained for the dilutions of the standard is plotted
against the concentration of protein C antigen (percentage, U/ml, IU/ml) on
log-log paper for the construction of the calibration curve (usually a straight
line). The lengths of the rockets for test samples are read on this calibration
curve by intrapolation.
Normal ranges
Among healthy volunteers protein C antigen varies between 0.65 and 1.45 U/ml
   =
(n 33; see ref. 16). In patients on oral anticoagulant treatment protein C
antigen is a function of the intensity of treatment (see Table 14.1)1 .
General comments
For the e1ectroimmunoassay of protein C antigen it is essential that ethylene-
diamine tetra-acetic acid (EDTA) is present in the gel and electrophoresis buffer.
Mean Range
                                              133
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Only under these conditions will the fully carboxylated and non-carboxylated
forms of protein C migrate at an identical rate. This is important because
otherwise the higher mobility of the non-carboxylated protein (as present in
plasmas of patients treated with oral anticoagulants) will result in relatively
over-long precipitation ~eaks and therefore in apparently higher protein C
antigen concentrations43 , 0. A disadvantage of the use of EDTA is that in most
cases twice as much antiserum is needed to obtain the same peak height for
undiluted pooled normal plasma. Good-quality rabbit and anti-protein C serum
can be used at concentrations between 0.5% and 2%.
   The staining and destaining solutions contain methanol. If the use of methanol
needs to be avoided, methanol can be replaced by ethanol (some loss in destaining
efficiency).
Second-generation tests
As mentioned above, the discovery of a specific protein C activator (Protac®)
in the venom of Agkistrodon contortrix contortrix 60-62 resulted in a second
generation of protein C activity assays. Because the activity of Protac® is not
dependent on ea2+ ions, and is insensitive to plasma protease inhibitors, Protac®
can be used to activate protein C directly in plasma; during a second incubation
the activity of the APC formed can be quantitated with a spectrophotometric
or clotting technique. Several manufacturers distribute these protein C activity
tests (e.g. Spectrolyse@ and Bioclot@, from Biopool, MDA@ Protein C from
Organon Teknika, Stachrom® Protein C and Staclot® Protein C from Diagnostica
Stago and Coamatic® Protein C from Chromogenix). Most of the chromogenic
tests use a different chromogenic substrate for the measurement of APe. Some
of these tests have been used and evaluated in comparative studies49,50,59,63,64.
Most of these tests have both end-point and kinetic modifications. For several
reasons the author prefers kinetic versions of the test. One of these reasons is
that it is sometimes impossible to have a satisfactory blank in the end-point
method (see, for further discussion, ref. 48). In the test of Biopool (Spectrolyse®)
such problems have been solved elegantly by including an inhibitory anti-protein
C antibody in the blank.
   A general problem with the protein C anticoagulant activity assays is the
stability of the standards with respect to the actual prolongation of clotting
time. This needs to be checked and confirmed very carefully. An obvious
advantage of these clotting tests is that they will also detect these protein C
variants that have defects interfering with the proper formation of a Ca2 +_
dependent conformation (these protein C variants have normal amidolytic
activity and reduced anticoagulant activity). On the other hand these variants
are rather rare6S . Moreover, many of the protein C anticoagulant activity tests
have been found to be sensitive to some extent to the presence of the factor V
Leiden mutation66,67.
References
 1. Esmon CT. The protein C anticoagulant pathway. Arterioscler Thromb 1992; 12: 135-45.
 2. Oahlbiick B, Stenfio J. A natural anticoagulant pathway: protein C, S, C4b-binding protein
    and thrombomodulin. In: Bloom AL, Forbes CO, Thomas OP, Tuddenham EGO, eds.
    Haemostasis and thrombosis. Edinburgh: Churchill Livingstone 1994; 671-98.
 3. Sills RH, Marlar RA, Montgomery RR, Oesphande GN, Humbert JR. Severe homozygous
    protein C deficiency. J Pediat 1984; 105: 409-13.
 4. Marlar RA, Kleiss AJ, Griffin JH. Mechanism of action of human activated protein C, a
    thrombin-dependent anticoagulant enzyme. Blood 1982; 59: 1067-72.
 5. Walker FJ. Protein S and the regulation of activated protein C. Sern Thromb Hemostas 1984;
    10: 131-8.
 6. Koedam JA, Meijers JCM, Sixma 11, Bouma BN. Inactivation of human factor VIII by
    activated protein C. J Clin Invest 1988; 82: 1236-43.
 7. Suzuki K, Stenflo J, Oahlbiick B, Teodorsson B. Inhibition of human coagulation factor V
    by activated protein C. J BioI Chern 1983; 258: 1914-20.
 8. Esmon CT. The roles of protein C and thrombomodulin in the regulation of blood coagulation.
    J Bioi Chern 1989; 264: 4743-6.
 9. Esmon NL, De Bault LE, Esmon CT. Proteolytic formation and properties of y-carboxyglutamic
    acid - domainless protein C. J Bioi Chern 1983; 258: 5548-53.
                                             136
                         PROTEIN C ACTIVITY AND ANTIGEN
10. Suzuki K, Nishioka I, Hashimoto S. Protein C inhibitor. Purification from human plasma
    and characterization. I Bioi Chern 1983; 258: 163-8.
II. Van der Meer FIM, van Tilburg NH, van Wijngaarden A, van der Linden IK, Briet E, Bertina
    RM. A second plasma inhibitor of activated protein C: ucantitrypsin. Thromb Haernostas
    1989; 62: 756-62.
12. Heeb MI, Gruber A, Griffin IH. Metal ion dependent inhibition of activated protein C by
    u2-macrog!obulin and u 2-antiplasmin in human blood. Arteriosclerosis 1990; 10: 893a.
13. Marlar RA, Montgomery RR, Broekmans AW. Diagnosis and treatment of homozygous
    protein C deficiency. I Pediat 1989; 114: 528-34.
14. Marlar RA, Neumann A. Neonatal purpura fulminans due to homozygous protein C or
    protein S deficiencies. Sem. Thromb Hemostas 1990; 16: 299-309.
15. Griffin IH, Evatt B, Zimmerman TS, K.1eiss AI, Wideman C. Deficiency of protein C in
    congenital thrombotic disease. I Clin Invest 1981; 68: 1370-3.
16. Broekmans AW, Veltkamp n, Bertina RM. Congenital protein C deficiency and venous
    thromboembolism. A study of three Dutch families. N Engl J Med 1983; 309: 340-4.
17. Allaart CF, Poort SR, Rosendaal FR, Reitsma PH, Bertina RM, Briet E. Increased risk of venous
    thrombosis in carriers of hereditary protein C deficiency defect. Lancet 1993; 341: 134-8.
18. Gladson CL, Scharrer I, Hack V, Beck KH, Griffin JH. The frequency of type I heterozygous
    protein S and protein C deficiency in 141 unrelated young patients with venous thrombosis.
    Thromb Haemostas 1988; 59: 18-22.
19. Bertina RM. Prevalence of hereditary thrombophilia and the identification of genetic risk
    factors. Fibrinolysis 1988; 2(S2): 7-13.
20. Seligsohn U, Berger A, Abend M, Rubin I, Attias D, Zivelin A, Rapaport SI. Homozygous
    protein C deficiency manifested by massive venous thrombosis in the newborn. N Eng! J Med
    1984; 310: 559-62.
21. Miletich I, Sherman L, Broze G Ir. Absence of thrombosis in subjects with heterozygous
    protein C deficiency. N Eng! I Med 1987; 317: 991-6.
22. Tait RC, Walker 10, Reitsma PH, Islam SIAM, McCan F, Poort SR, Conkie lA, Bertina
    RM. Prevalence of protein C deficiency in the healthy population. Thromb Haemostas 1995;
    73: 87-93.
23. Koeleman BPC, Reitsma PH, AHaart CF, Bertina RM. Activated protein C resistance as an
    additional risk factor for thrombosis in protein C deficient families. Blood 1994; 84: 1031-5.
24. Bertina RM, Broekmans AW, van der Linden IK, Mertens K. Protein C deficiency in a Dutch
    family with thrombotic disease. Thromb Haemostas 1982; 48: 1-5.
25. Pabinger I, Kyrle PA, Speiser W, Stoffels U, lung M, Lechner K. Diagnosis of protein C
    deficiency in patients on oral anticoagulant treatment: comparison of three different functional
    protein C assays. Thromb Haemostas 1990; 63: 407-12.
26. Cohen H, Mackie U, Walshe K, Gil1mer MD, Machin S1. A comparison of the effects of two
    triphasic oral contraceptives on haernostasis. Br J Haernatol1988; 69: 259-63.
27. Gensini GF, Rostagno C, Abbat R, Favilla S, Mannucci PM, Semeri GGN, Bonomi AB.
    Increased protein C and fibrinopeptide A concentration in patients with angina. Thromb Res
    1988; 30: 517-25.
28. Cosio FG, Harker C, Batard MA, Brandt JT, GrifTm IH. Plasma concentrations of the natural
    anticoagulants protein C and protein S in patients with proteinuria. J Lab Clin Med 1985;
    106: 218-22.
29. Pabinger-Fasching I, Lechner K, Niessner H, Schmidt P, Ba1zar E, Mannhalter CL. High
    levels of plasma protein C in nephrotic syndrome. Thromb Haemostas 1985: 53: 5-7.
30. Blarney SL, Lowe GDO, Bertina RM, K.1uft C, Sue Ling HM, Davies lA, Forbes CD. Protein
    C antigen levels in major abdornina1 surgery: relationships of deep vein thrombosis, malignancy
    and treatment with stanozolol. Thromb Haernostas 1985; 54: 622-5.
31. Vigano S, Mannueei PM, D'Angelo A, Gelfi C, Gensini GF, Rostagno C, Semeri GGN. The
    significance of protein C antigen in acute and chronic liver biliary disease. Am I Clin Pathol
    1984; 84: 454-8.
32. Marlar RA, Endres-Brooks I, Miller C. Serial studies of protein C and its plasma inhibitor
    in patients with disseminated intravascular coagulation. Blood 1985; 66: 59-63.
33. Ceriello A. Quatrano A, Dello Russo P, Marchi E, Barbanti M, Rita Milani M, Giugliano
    D. Protein C deficiency in insulin-dependent diabetes: a hyperg!ycemia-related phenomenon.
    Thromb Haernostas 1990; 64: 104-7.
                                               137
          u\BORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
34. Vukovich TC, Schernthaner G. Decreased protein C levels in patients with insulin-dependent
    type I diabetes mellitus. Diabetes 1986; 35: 617-19.
35. Kloczko J, Wojtukiewicz M, Bielawiec M, Borowska M. Reduced protein C levels in patients
    with essential hypertension. Thromb Haemostas 1987; 58: 793.
36. Green D, Scott JP. Is sickle cell crisis a thrombotic event? Am J Hematol1986; 23: 317-21.
37. Miletich JP. Laboratory diagnosis of protein C deficiency. Sem Thromb Haemostas 1990; 16:
    169-76.
38. Gonzalez R, Alberca I, Sala N, Vicente V. Protein C deficiency - response to danazol and
    DDAVP. Thromb Haemostas 1985; 53: 320--2.
39. Broekmans AW, Conard J, van Weijenberg RG, Horellou MH, Kluft C, Bertina RM. Treatment
    of hereditary protein C deficiency with stanozolol. Thromb Haemostas 1987; 57: 20--4.
40. Conard J, Horellou MH, Dreden P, Potevin F, Zittoun R, Samama M. Decrease in protein
    C in L-asparaginase-treated patients. Br J Haematol1985; 59: 725-41.
41. Vigano S, Mannucci PM, Solinas S, Botasso B, Mariani G. Decrease in protein C antigen and
    formation of an abnormal protein soon after starting oral anticoagulant therapy. Br J Haematol
    1984; 57: 213-20.
42. Marlar RA, Adcock DM. Clinical evaluation of protein C: a comparative review of antigenic
    and functional assays. Progr Patho11989; 20: 1040--7.
43. Mikami S, Tuddenham EGD. Studies on immunological assay of vitamin K-dependent factors.
    II. Comparison of four immunoassay methods with functional activity of protein C in human
    plasma. Br J Haematol1986; 62: 183-93.
44. Boyer C, Rothschild C, Wolf M, Amiral J, Meyer D, Larrieu M1. A new method for the
    estimation of protein C by ELISA. Thromb Res 1984; 36: 579-89.
45. Suzuki K, Moriguchi A, Nagayoshi A, Mutoh S, Katszuki S, Hashimoto S. Enzyme
    immunoassay of human protein C by using monoclonal antibodies. Thromb Res 1985; 38:
    611-21.
46. Ikeda K, Stenfio 1. A radioimmunoassay for protein C. Thromb Res 1985; 39: 297-306.
47. Epstein DJ, Bergum PW, Bajaj SP, Rapaport SI. Radioimmunoassay for protein C and factor
    X. Plasma antigen levels in abnormal haemostatic states. Am J Clin Patho11984; 82: 573--81.
48. Bertina RM. Specificity of protein C and protein S assays. Res Clin Lab 1990; 20: 127-38.
49. Hubbard AR. Standardization of protein C in plasma: establishment of an International
    Standard. Thromb Haemostas 1988; 59: 464-7.
50. Bertina RM. An international collaborative study on the performance of protein C antigen
    assays. Thromb Haemostas 1987; 57: 112-17.
51. Mannucci PM, Boyer C, Tripodi A, Vigano d' Angelo S, Wolf M, Valsecchi C, d'Angelo A,
    Meyer D and Larrieu MJ. Multicenter comparison of five functional and two immunological
    assays for protein C. Thromb Haemostas 1987; 57: 44-8.
52. Francis RB, Patch MJ. A functional assay for protein C in human plasma. Thromb Res 1983;
    32: 605-13.
53. Comp PC, Nixon RR, Esmon CT. Determination of functional levels of protein C, an
    antithrombotic protein, using thrombin-thrombomodulin complex. Blood 1984; 63: 15-21.
54. Sala N, Owen NG, Collen D. Functional assay of protein C in human plasma. Blood 1984;
    63: 671-5.
55. Bertina RM, Broekmans AW, Krommenhoek-van Es C, van Wijngaarden A. The use of a
    functional and immunologic assay for plasma protein C in the study of the heterogeneity of
    congenital protein C deficiency. Thromb Haemostas 1984; 51: 1-5.
56. Vigano-D'Angelo S, Comp PC, Esmon CT, D'Angelo A. Relationship between protein C
    antigen and anticoagulant activity during oral anti-coagulation and in selected disease states,
    J Clin Invest 1986; 77: 416-25.
57. Martinoli JL, Stocker K. Fast functional protein C assay using Protac, a novel protein C
    activator. Thromb Res 1986; 43: 253-64.
58. Francis RB, Seyfert U. Rapid amidolytic assay of protein C in whole plasma using an activator
    from the venom of Agkistrodon contortrix contortrix. Am J Clin Patho11987; 87: 619-25.
59. Vinazzer H, Pangraz U. Protein C: comparison of different assays in normal and abnormal
    plasma samples. Thromb Res 1987; 46: 1-8.
60. Stocker K, Fischer H, ¥eier J, Brogli M, Svendsen L. Protein C activators in snake venoms.
    Behring Institut Mitteilungen 1986; 79: 37-47.
                                               138
                        PROTEIN C ACTIVITY AND ANTIGEN
61. Exner T, Vaasjoki R. Characterization and some properties of the protein C activator from
    Agkistrodon contortrix contortrix venom. Thromb Haemostas 1988; 59: 40-4.
62. Orthner CL, Bhattacharya P, Strickland DK. Characterization of a protein C activator from
    the venom of Agkistrodon contortrix contortrix. Biochemistry 1988; 27: 2558-64.
63. Sturk A, Morrien-Salomons WM, Huisman MY, Borm JJJ, Biiller HR, ten Cate JW. Analytical
    and clinical evaluation of commercial protein C assays. Clin Chim Acta 1987; 165: 263-70.
64. Franchi F, Tripodi A, Valsecchi C, Mannucci PM. Functional assays of protein C: comparison
    of two snake-venom assays with two thrombin assays. Thromb Haemostas 1988; 60: 145--7.
65. Reitsma PH, Bernardi F, Doig RG, Gaudrille S, Greengard JS, Ireland H, Krawczak M, Lind
    B, Long GL, Poort SR, Saito H, Sala N, Witt I, Cooper D. Protein C deficiency: a database
    of mutations, 1995 update. Thromb Haemostas 1995; 73: 876-89.
66. Ireland H, Bayston T, Thompson E, Adami A, Gon~lves C, Lane DA, Finazzi G, Barbui T.
    Apparent heterozygous type II protein C deficiency caused by the factor V 506 Arg to Gin
    mutation. Thromb Haemostas 1995; 73: 731-2.
67. Girolami A, Simioni P, Tormene D, Vianello F. Pitfall of protein C resistance. Blood Coagul
    Fibrinol 1996; 7:712-13.
68. Mannucci PM, Boyer C, Tripodi A, Vigano-D'Angelo S, Wolf M, Valsecchi C, D'Angelo A,
    Meyer D, Larrieu MJ. Multicenter Comparison of five functional and two immunologic
    assays for protein C. Thromb Haemostas 1987; 57: 44-8.
                                             139
15
Protein S antigen
R. M. BERTINA
INTRODUCTION
Physiological role
PS is the protein cofactor of the anticoagulant enzyme activated protein C
(APC)9 and therefore is an essential component of the PC anticoagulant
pathway lO,ll. It stimulates the inactivation of factor Va and factor VIlla by
APC both in plasma and purified systems I2- 14 . It is also required for the
expression of the fibrinolytic effect of APe in a whole-blood clot lysis systeml5 .
PS forms a I: I complex with APC on phospholipid surfaces and is thus thought
to stimulate the phospholipid-dependent inactivation of factors Va and VIlla.
The APC cofactor activity of PS is regulated by two independent processes.
First, PS is cleaved by thrombin in the TSR region; this cleavage results in the
formation of a molecule consisting of two chains linked by one disulphide
bridge, which no longer has APC cofactor activity I6,17. However, under physi-
ological conditions (Le. in the presence of Ca2 +) this reaction is rather slow
and therefore will require high local thrombin concentrations.
                                       141
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   A second mechanism controlling the APC cofactor activity of PS is its ability
to form a 1:1 complex with the C4b binding protein (C4bBP), an important
regulatory component of the complement systemlO. The C4bBP PS complex
has no APC cofactor activityl8. However, this complex - which still can bind
to phospholipid membranes - may playa role in the regulation of complement
activation 19 . More recently evidence has been obtained that - at least in vitro
- both free and C4bBP-bound PS also have an anticoagulant activity which is
independent of APC20-22• The physiological relevance of this APC-independent
anticoagulant activity is still unknown.
   Binding of PS to C4bBP occurs via a separate unique subunit of C4bBP,
the /3-chain23 • The Kd for binding of PS to C4bBP is very low (5 x 10-10 mollL)
in the presence of Ca2+ (see ref. 23). This might explain why in plasma almost
all /3-chain containing C4bBP is found to be bound to PS8. It is only the excess
PS that circulates as free PS8,25. Interestingly, the genes for the C4bBP u- and
/3-chains are under different regulation25 , In citrated human plasma or in serum,
free and C4bBP PS occur in a ratio of 2:3 8,10,24.
Pathophysiological aspects
Hereditary defects
The importance of PS for the control of thrombin formation in vivo is most
dramatically demonstrated in neonates who are homozygotes or double
heterozygotes for hereditary PS deficiency26,27. Such patients develop, shortly
after birth, clinical symptoms (among others, purpura fulminans syndrom~
that are very similar to those reported for homozygous protein C deficiencr .
Heterozygotes for hereditary PS deficiency have an increased risk for the
development of venous thrombosis at relatively young age29-31. More recent
studies indicate that PS deficiency might also be a weak risk factor for arterial
thrombosis 32 •
   In plasma PS circulates both as free and C4bBP-complexed PS. This will
complicate the laboratory diagnosis of hereditary PS deficiencies. In fact three
types of deficiencies have been recognized33 • In type I PS deficiency there is a
reduction of total PS antigen till about 50% of normal23 ,30. Type I deficiencies
are caused by alterations in the PS gene that do not permit normal transcription,
translation and/or secretion of the protein34• In type II PS deficiency the total
PS antigen is normal while one of the PS genes makes a molecule with a
strongly reduced biological activity34. In the laboratory this deficiency can be
diagnosed by the reduced ratio PS activity/free PS antigen. Unfortunately most
of the reported cases of type II PS deficiency are patients with an apparent
reduced PS activity due to the presence of the factor V Leiden mutation (see
Chapter 16 on PS activity). The third type of hereditary PS deficiency is the
so-called type III deficiency. In patients with this type of deficiency the free PS
antigen is reduced while total PS is within the normal range. Recent studies
demonstrated that the phenotypes of both type I and ti~e III PS deficiency
can be found in carriers of the same PS gene defece ' 6. This was partly
explained by an age-dependent increase in total PS (mainly due to an increase
                                       142
                              PROTEIN S ANTIGEN
in C4bBP-bound PS). From these studies it is clear that free PS measurements
discriminate better between carriers and non-carriers of a PS gene defect than
total PS measurements.
Acquired abnormalities
Abnormal plasma levels of total and/or free PS have been reported for a number
of clinical conditions. Sometimes the interpretation of the reported data is
hampered by the lack of information on the specificity of the assay (especially
true for some of the total PS antigen assays).
   Reduced levels of total PS have been reported for patients usin~ oral
anticoagulants7,24 during pregnancy and use of oral contraceptives37- 9 and
in patients with liver disease24• Conflicting data (reduced/increased PS antigen)
have been reported for patients with diabetes mellitus40,41. Increased PS antigen
(total) has been found in patients with nephrotic syndrome42 . The simultaneous
increase in C4bBP, however, might result in an acquired deficiency of free PS43.
No change in total PS antigen has been observed during disseminated intra-
vascular coagulation7,24.
   Under conditions in which acute-phase reactions mal occur, elevated C4bBP
levels might cause a reduction in free PS concentration2 . This may occur during
an acute thrombotic episode, although elevated total and free PS antigen have
also been reported for acute deep vein thrombosis44 .
PROTEIN S ASSAYS
Materials
ELISA buffer
This buffer is used for making the dilutions of the samples, for washing the
micro titre plates, and as diluent for the anti-PS conjugate. It consists of:
67 mmollL Na2HP04' 2H20, 14 mmollL KH2P04, 0.1% (v/v) Tween 20 (PH
7.5). This solution is freshly prepared for each series of tests.
Coating buffer
This buffer is used for the coating of microtitre plates. It consists of 0.1 mollL
NaHC0 3, 0.5 mollL NaCI (PH 9.0).
Substrate buffer
This buffer is used to dissolve the HRP-substrate OPD (orthophenylene
diamine). It consists of 22.2 mmollL citric acid, 51.4 mmollL NaH2P04 . H 20
(pH 5.0) and can be stored for several weeks at 4 0c.
                                        144
                              PROTEIN S ANTIGEN
Substrate solution
20 mg orthophenylene diamine (dihydrochloride) (Sigma) is dissolved in 50 ml
substrate buffer (see above). About I min before addition to the microtitre
plates 20 ~ of a 30% H 20 2 solution is added. This volume of substrate solution
should be sufficient for four or five plates.
Stop reagent
4 N H 2S04 is used to stop the peroxidase-catalysed conversion of OPD.
Catching antibody
As catching antibody rabbit antibodies against human PS are used, routinely
these antibodies are obtained by isolating the non-Ca2+ -dependent fraction of
the anti-PS antibodies from the antiserum by immunoaffmity chromatography
on PS Sepharose (2 mg purified human PS/ml gel)24,46. Stock solutions (>
0.050 mg IgG/ml) are stored in I ml aliquots at -20°C.
Tagging antibody
The same non-Ca2 +-dependent anti-PS antibodies that are used as catching
antibodies are used as tagging antibodies. Horseradish peroxidase (HRP) is
coupled to the anti-PS IgG using the reagent N-succinimidyI3-(2-piridylthio)
propionate (SPDP) as described elsewhere52• The conjugate is stored in suitable
aliquots at -20°C in a buffer containing 0.3 mollL NaC!. Dilutions of the
conjugate (usually about 114000) are prepared about 30 min prior to use. It
should be noted that anti-PS HRP conjugate can be purchased from a number
of different manufacturers.
Other materials
For the transfer of samples, conjugate, or substrate solution to the micro titre
plate multichannel pipettes are used. Absorbances are read on a multichannel
analyser (Titertek Multiscan Plus) using a filter of 492 nm.
                                        145
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Test procedure
Test protocol
Suitable dilutions of test sample and standard (100 ~) are added to the wells
and incubated for 18 h (usually overnight) at room temperature to allow for
equilibrium distribution of PS over solid and fluid phase. Assay plates then are
washed five times with ELISA buffer before 100 ~lIwell of a dilution of
HRP-Iabelled anti-PS immunoglobulin is added (usually a 114000 dilution of
the stock in ELISA buffer). Plates are then incubated for about 45 min at room
temperature. Subsequently plates are emptied and washed five times with
ELISA buffer. Then 100 Ill/well of substrate solution (see above) is added.
Plates are incubated for about 30 min (until sufficient colour has been formed)
before the peroxidase activity is stopped by the addition of 100 ~l/well of 4 N
H 2 S04 , Finally the absorbance at 492 nm is read in a multichannel analyser
(see Other materials, above).
Calibration
The ELISA for total PS antigen is calibrated with dilutions of pooled normal
plasma (usually from 1/500 serially to 1132000). Routinely, samples from
patients not using oral anticoagulants are tested in 112000 and 1/4000 dilutions,
while plasmas of patients using oral anticoagulants are tested in 111000 and
112000 dilutions. Each dilution is tested in duplicate. All dilutions are made in
ELISA buffer. To calculate the amount of total PS antigen in plasmas, the
absorbance values were read from the standard curve which was obtained of
a least-square fit of third-order polynomial to the data plotted on a double-
logarithmic scale. Test results are valid when the SD is less than 10%.
   Each laboratory relies on its own pooled plasma. Care should be taken that
sufficient individual donations have been included in the pool (plasmas of
females using oral contraceptives should not be included). Total PS antigen is
expressed as a percentage or as units/ml, where 1 unit reflects the amount of
PS present in 1 ml pooled normal plasma.
                                      146
                              PROTEIN S ANTIGEN
Reference ranges
Vsing the aforementioned ELISA, total PS antigen was measured in 40 healthy
volunteers; mean PS antigen was: 1.12 ± 0.16 unit/ml, while individual values
ranged from 0.77 to 1.44 unit/ml.
   In 40 patients using oral anticoagulant treatment mean PS antigen was: 0.52
± 0.10 unit/ml, while individual values ranged from 0.34 to 0.69 units/ml. As
mentioned above, it is recommended that the laboratory diagnosis of PS is
based on the measurements of both total and free PS. It is recommended,
however, to use separate normal ranges for males and females 53 ,54; eventually
these can be extended with a normal range for oral contraceptive users. The
influence of age could also be taken into account35 . Calibration of local pooled
normal plasma can be made against the first international standard PS, plasma
(coded 93/590), which has a potency of 0.90 IV/ampoule for total PS, free PS
and PS activity 55.
References
 1. Di Scipio RG, Hermodson MA, Yates SG, Davie EW. A comparison of human prothrombin,
    factor IX, factor X and protein S. Biochemistry 1977; 16: 696-706.
                                         148
                                    PROTEIN S ANTIGEN
 2. Di Scipio RG, Davie EW Characterization of protein S, a y-carboxyglutarnic acid containing
    protein from bovine and human plasma. Biochemistry 1979; 18: 899-904.
 3. Lundwall A, Dackowski W, Cohen E, Schaffer M, Mahr A, Dahlbiick B, Stenfio 1. Isolation
    and sequence of the c-DNA for human protein S, a regulator of blood coagulation. Proc Natl
    Acad Sci USA 1987; 83: 6716-20.
 4. Fair DS, Marlar RA. Biosynthesis and secretion of factor VII, protein C, protein S and the
    protein C inhibitor from a human hepatoma cell-line. Blood 1986; 67: 64--70.
 5. Stern D, Brett J, Harris K, Nawroh P. Participation of endothelial cells in the protein C-protein
    S anticoagulant pathway: the synthesis and release of protein S. J Cell Bioi 1986; 102: 1971-8.
 6. Ogura M, Tanabe N, Nishioka J, Suzuki K, Saito H. Biosynthesis and secretion of functional
    protein S by a human megakaryoblastic ceU line (MEG-Ol). Blood 1987; 70: 301-6.
 7. D'Angelo A, Vigano-D'Angelo S, Esmon CT, Comp PC. Acquired deficiencies of protein S.
    J Clin Invest 1988; 81: 1445-54.
 8. Griffin JH, Gruber A, Fernandez JA. Reevaluation of total, free and bound protein Sand
    C4b-binding protein levels in plasma anticoagulated with citrate or hurudin. Blood 1992; 79:
    3203--11.
 9. Walker F1. The regulation of activated protein C by a new protein; a possible function for
    bovine protein S. J BioI Chern 1980; 255: 5521-4.
10. Dahlbiick B, Stenfto 1. A natural anticoagulant pathway: protein C, S, C4b-binding protein
    and thrombomodulin. In: Bloom AL, Forbes CD, Thomas DP, Tuddenham EGD, eds.
    Haemostasis and thrombosis. Edinburgh: ChurchiU Livingstone, 1994; 671-98.
11. Esmon CT, Schwarz HP. An update on clinical and basic aspects of the protein C anticoagulant
    pathway. Trends Cardiovasc Med 1995; 5: 141-8.
12. Walker F1. Regulation of activated protein C by protein S. The role of phospholipid in factor
    Va inactivation. J BioI Chem 1981; 256: 11128-31.
13. Walker FJ, Chavin SI, Fay PJ. Inactivation of factor VIII by activated protein C and protein
    S. Arch Biochem Biophys 1987; 252: 322-8.
14. Rosing J, Hoekema L, Nicolaes GAF, Christella M, Thomassen LGD, Hemker HC, Varadi
    K, Schwarz HP, Tans G. Effects of protein S and factor Xa on peptide bond cleavages during
    inactivation of factor Va and factor Va R506Q by activated protein C. J BioI Chem 1995; 270:
    27852-8.
15. De Fouw NJ, Haverkate F, Bertina RM, Koopman J, van Wijngaarden A, van Hinsbergh
    VWM. The cofactor role of protein S in the acceleration of whole blood clot lysis by activated
    protein C in vitro. Blood 1986; 67: 1189-92.
16. Walker FJ. Regulation of vitamin K dependent protein S. Inactivation by thrombin. J BioI
    Chem 1984; 259: 10335-9.
17. Sugo T, Dahlbiick B, Holmgren A, Stenfio 1. Calcium binding of bovine protein S. Effect of
    thrombin cleavage and removal of the y-carboxyglutarnic acid-containing region. J Bioi Chem
    1986;261:5116-20.
18. Dahlbiick B. Inhibition of protein Ca cofactor function of human and bovine protein. J Bioi
    Chem 1986; 261: 12022-7.
19. Schwalbe R, Dahlbiick B, Hillarp A, Nelsestuen G. Assembly of protein S and C4b-binding
    protein on membranes. J BioI Chem 1990; 265: 16074-81.
20. Heeb MJ, Mesters RM, Tans G, Rosing J, Griffin JH. Binding of protein S to factor Va
    associated with inhibition of prothrombinase that is independent of activated protein C. J
    Bioi Chern 1993; 268: 2872-7.
21. HackengTM, van't Veer C, Meijers JC, Bouma BN. Human protein S inhibits prothrombinase
    complex activity on endothelial ceUs and platelets via direct interactions with factors Va and
    Xa. J BioI Chem 1994; 269: 21051-8.
22. van Wijnen M, Stam JG, van't Veer C, Meijers JCM, Reitsma PH, Bertina RM, Bouma BN.
    The interaction of protein S with the phospholipid surface is essential for the activated protein
    C-independent activity of protein S. Thromb Haemostas 1996; 76: 397-403.
23. Hillarp A, Dahlbiick B. Novel subunit in C4b-binding protein required for protein S binding.
    J BioI Chem 1988; 263: 12759--64.
24. Bertina RM, van Wijngaarden A, Reinalda-Poot J, Poort SR, Born VJJ. Determination of
    plasma protein S - the protein cofactor of activated protein C. Thromb Haemostas 1985; 33:
    268-72.
                                                149
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
25. Garcia de Frutos P, AIim RIM, Hiirdig Y, ZOller B, Dahlbiick B. Differential regulation of
    a- and fl-chains of C4b-binding protein during acute-phase response, resulting in stable plasma
    levels of free anticoagulant protein S. Blood 1994; 84: 815-22.
26. Mahasandana C, Suvatte V, Chuansumrit A, Marlar RA, Manco Johnson MJ, Jacobson LJ,
    Hathaway WE. Homozygous protein S deficiency in an infant with purpura fulrninans. J
    Pediatr 1990; 117: 75{}-3.
27. Gomez E, Ledford MR, Pegelow CH, Reitsma PH, Bertina RM. Homozygous protein S
    deficiency due to a one base pair deletion that leads to a stop codon in exon III of the protein
    S gene. Thromb Haemostas 1994; 71: 72~.
28. Marlar RA, Neumann A. Neonatal purpura fulrninans due to homozygous protein C or
    protein S deficiencies. Sem Thromb 1990; 16: 299-309.
29. Comp PC, Esmon CT. Recurrent venous thrombo-embolism in patients with a partial deficiency
    of protein S. N Eng! J Med 1984; 311: 1525-8.
30. Schwarz HP, Fischer M, Hopmeyer P, Batard MA, Griffm JH. Plasma protein S deficiency
    in familial thrombotic disease. Blood 1984; 64: 1297-300.
31. Engesser L, Broekmans AW, Briet E, Brommer EJP, Bertina RM. Hereditary protein S
    deficiency: clinical manifestations. Ann Intern Med 1987; 106: 677-82.
32. Zoller B, Garcia de Frutos P, Dahlbiick B. Evaluation of the relationship between protein S
    and C4b binding protein isoforms in hereditary protein S deficiency demonstrating type I and
    type III deficiencies to be phenotypic variants of the same genetic disease. Blood 1995; 85:
    3524-31.
33. Bertina RM. Hereditary deficiencies of protein C and protein S. In: Aznar J, Espana F, eds.
    Protein C pathway. Iberica: Springer Verlag, 1991; 111-20.
34. Gandrille S, Borgel D, Ireland H, Lane DA, Simmonds R, Reitsma PH, Mannhalter C,
    Pabinger I, Saito H, Suzuki K, Formstone C, Cooper DN, Espinosa Y, Sala N, Bernardi F,
    AiachM. Protein S deficiency: a database of mutations. ThrombHaemostas 1997; 77: 1201-14.
35. Simmonds RE, Zoller B, Ireland H, Thompson E, Garcia de Frutos P, Dahlbiick B, Lane
    DA. Genetic and phenotypic analysis of a large (122-member) protein S deficient kindred
    provides an explanation for the familial coexistence of type I and type III plasma phenotypes.
    Blood 1997; 89: 4364-70.
36. Zoller B, Garcia de Frutos P, Dahlbiick B. Evaluation of the relationship between protein S
    and C4b-binding protein isoforms in hereditary protein S deficiency demonstrating type I and
    type III deficiencies to be phenotypic variants of the same genetic disease. Blood 1995; 85:
    3524-31.
37. Maim J, Laurell M, Dahlback B. Changes in the plasma levels of vitamin K-dependent proteins
    C and S and of C4b-binding protein during pregnancy and oral contraception. Br J Haematol
     1988; 68: 437--43.
38. Comp PC, Thurnau GR, Webb J, Esmon CT. Functional and immunologic protein S levels
    are decreased during pregnancy. Blood 1986; 68: 881-5.
39. Boerger 1M, Morris PC, Thurnau CT, Esmon CT, Comp PC. Oral contraceptives and gender
    affect protein S status. Blood 1987; 69: 692--4.
40. Takahashi H, Tatewaki H, Wada K, Shibatu A. Plasma protein S in disseminated intravascular
    coagulation, liver disease, collagen disease, diabetes mellitus, and under oral anticoagulant
    therapy. Clin Chim Acta 1989; 182: 195-208.
41. Schwarz HP, Schemthaner G, Griffm JH. Decreased plasma levels of protein S in well-controled
    type I diabetes mellitus. Thromb Haemostas 1987; 57: 240.
42. Rostoker G, Goualt-Heilmann M, Levent M, Robeva R, Lang P, Lagrue G. High level of
    protein C and protein S in nephrotic syndrome. Nephron 1987; 46: 22{}-1.
43. Vigano d' Angelo S, D'Angelo A, Kaufman CE Jr, Sholer C, Esmon CT, Comp PC. Protein
    S deficiency occurs in the nephrotic syndrome. Ann Intern Med 1987; 107: 42-7.
44. Toulon P, Gandrille S, Vitoux IF, Fiessinger IN, Sultan Y, Aiach M. High total and free
    protein S antigen in patients with acute deep vein thrombosis. Thromb Res 1990; 59: 213-17.
45. Poort SR, Deutz-Terlouw PP, van Wijngaarden A, Bertina RM. Immunoradiometric assay
    for the calcium-stabilized conformation of human protein S. Thromb Haemostas 1987; 58:
    998-1004.
46. Deutz-Terlouw PP, Ballering L, Van Wijngaarden A, Bertina RM. Two ELISA's for
    measurement of protein S, and their use in the laboratory diagnosis of protein S deficiency.
    Clin ChimActa 1989; 186: 321-34.
                                               150
                                   PROTEIN S ANTIGEN
47. Woodhams BJ. The simultaneous measurement of total and free protein S by ELISA. Thromb
    Res 1988;50:213-20.
48. Arniral J, Adam M, Plassant V, Minard F, Vissac AM. Immunoassays for the measurement
    of protein S. In: Gaffney PJ, ed. Fibrinolysis, current prospects. London: John Libbey, 1988;
    125-8.
49. Krachmalnicoff A, Tombesi S, Va!secchi C, Albertini A, Mannucci PM. A monoclonal
    antibody to human protein S used as the capture antibody for measuring total protein S by
    enzyme immunoassay. Clin Chem 1990; 36: 43--6.
50. Fair DS, Revak DJ. Quantitation of human protein S in the plasma of normal and warfarin
    treated individuals by radioimmunoassay. Thromb Res 1984; 36: 527-35.
51. Maim J, Bernhager R, Holmberg L, Dahlbiick B. Plasma concentrations of C4b-binding
    protein and vitamin K-dependent protein S in term and preterm infants: low levels of protein
    S-C4b-binding complexes. Br J Haematol1988; 68: 445-9.
52. Carlsson J, Orevin H, Axen R. Protein thiolation and reversible protein-protein conjugation.
    N-succinimidyI3-(2-pyridyldithio) propionate, a new hetero-bifunctional reagent. Biochem J
    1978; 173: 723-37.
53. Faioni EM, Valsecchi C, Palla A, Taioli E, Razzari C, Mannucci PM. Free protein S deficiency
    in a risk factor for venous thrombosis. Thromb Haemostas 1997; 78: 1343--6.
54. Gari M, Falkon L, UrrUtia T, Vallve C, Borrell M, Fontcuberta J. The influence of low protein
    S levels in young women on the definition of the normal range. Thromb Res 1994; 73: 149-52.
55. Hubbard AR. Standardisation of protein S in plasma: calibration of the lst international
    standard. Thromb Haemostas 1997; 78: 1237-41.
56. Edson JR, Vogt JM, Huesman DA. Laboratory diagnosis of inherited protein S deficiency.
    Am J Clin Pathol 1990; 94: 176-86.
57. Comp PC, Doray D, Patton D, Esmon CT. An abnormal plasma distribution of protein S
    occurs in functional protein S deficiency. Blood 1986; 67: 5~.
58. Amiral J, Grosley B, Boyer-Neumann C, Marfaing-Koka A, Peynaud-Debayle E, Wolf M,
    Meijer D. New direct assay of free protein S antigen using two distinct monoclonal antibodies
    specific for the free form. Blood Coag FibrinoI1994; 5: 179-86.
59. Wolf M, Boyer-Neumann C, Peynaud-Debayle E, Marfaing-Koka A, Amiral J, Meijer D.
    Clinical applications of a direct assay of free protein S antigen using monoclonal antibodies.
    A study of 59 cases. Blood Coag Fibrino11994; 5: 187-92.
60. Aillaud MF, Pouymayon K, Brunet D, Parrot G, Alessi MC, Arnira! J, Juhan-Vague I. New
    direct assay of free protein S antigen applied to diagnosis of protein S deficiency. Thromb
    Haemostas 1996; 75: 283-5.
61. Giri TK, Hillarp A, Hiirdig Y, Zoller B, Dahlbiick B. A new direct, fast and quantitative
    enzyme-linked ligand absorbent assay for measurement of free protein S antigen. Thromb
    Haemostas 1998; 79: 767-72.
                                              151
16
Protein S activity
E. M. FAIONI
INTRODUCTION
The function of protein S
Protein S (PS) activity assays are based on the property of PS to act as a co-
factor to activated protein C (APC) in the proteolytic inactivation of factors
Va and VIlla (reviewed in ref. I). Evidence from in-vitro studies suggests that
PS expresses its cofactor activity by binding and localizing APC on cellular
membranes (platelets, endothelial cells, leukocytes) where the clotting process
is ongoing (Figure 16.1). Thrombin-inactivated PS, PS bound to C4b-binding
protein (C4b-BP) and non-carboxylated PS cannot function as a cofactor to
APe. In the first case, proteolysis by thrombin of residues in the thrombin-
sensitive region of PS leads to formation of a two-chain disulphide-linked PS
which does not bind phospholipids as efficiently as native PS; in the second
case, possibly due to the presence of the bulky C4b-BP, masking of the
APC-binding region may occur. Finally, when the vitamin K-dependent
carboxylation of glutamic acid residues in the amino terminal region of PS
(Gla-domain) is impaired (as, for example, during oral anticoagulant therapy),
the resulting partially carboxylated molecule loses calcium-mediated Gla-
dependent binding capacity to negatively charged phospholipids2 •
   In healthy individuals approximately 40% of circulating PS is free, while the
remainder is bound to C4b-BPI. The stoichiometry of the C4b-BP-PS interaction
is 1:1 1• C4b-BP is a spider-like molecule containing two types of polypeptide
chains, and only the ~ chain, not the a chain, is capable of binding PS (Figure
16.1)1. For a long time the consensus was that increased concentrations of
C4b-BP, which may be found during acute-phase reactions, would shift the
equilibrium between the bound and the free form of PS towards the former,
thus decreasing the anticoagulant potential of the protein C pathway. Recently
it has been observed that differential regulation of (l and ~ chain synthesis
                                      153
           LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                                                    \ ~P."-PS
                                                        ~Gla-domajn
   8,
   ~
       ~
       ~
                     @.a
                     ~
                         Villa
                                  ~f:
                                 APC
                                             ca2+
                                                       I   - - - thrombin
                                       cel/ membrane
Figure 16.1 Schematic representation of APC cofactor function of protein S. Protein S (PS)
anchors activated protein C (APC) to negatively charged membrane surfaces, thus localizing its
proteolytic inhibitory activity to Va and VIlla. PS is in dynamic equilibrium with C4b-binding
protein (C4b-BP) which binds PS through its fI subunit. The C4b-BP-PS complex does not function
as APC cofactor. Thrombin can inactivate PS, and non-carboxylated forms of PS lose high-affinity
interaction with membranes. Though the binding regions of PS for C4b-BP have in fact been
attributed to the amino-terminal portion of PS (see ref. 1), the figure does not aim to represent
precise intermolecular connections.
Procedure
1.   Reconstitute Protac® (protein C activator) with 0.75 ml distilled water/vial.
2.   Reconstitute PS-deficient plasma with 1.0 ml distilled water/vial.
3.   Reconstitute PS control plasma with 1.0 ml distilled water/vial.
4.   Reconstitute bovine tissue thromboplastin factor with 3.0 ml distilled water/
     vial.
Note: all reagents can be aliquoted, frozen and stored at -20°C once they have
been reconstituted. See insert accompanying the kit for duration and condi-
tions of storage.
    Patient samples, control plasma and PS-deficient plasma must be diluted
1: lOin factor diluent (saline). Protein C in PS-deficient plasma must be activated
by mixing one part of Protac® and two parts of PS-deficient plasma. Incubation
is carried out at 37°C for 20 min. This is the substrate plasma that the ACL
adds to the diluted plasma samples together with bovine thromboplastin and
calcium to obtain clotting. The ACL generates a reference curve made up of
four points (0%, 50%, 75%, 100%) in the older models, and two points (0%,
100%) in the newer ACL 6000 model. In each run a maximum of 13 samples
(at one dilution), including the control plasma, can be analysed.
    For instructions on where to place the reagents and the plasma samples
correctly in the rotor and in the coagulometer, see accompanying insert in the
kit.
    In the new ACL 6000, a dedicated PS activity program exists which is
user-friendly, prepares the plasma dilutions, and gives percentage PS activity
per sample at the end of the assay. In older models, sample dilutions must be
made by the operator and the factor II program has to be used. Since this is a
procoagulant factor program it cannot correctly construct the reference curve
and calculate the sample values (prolongation of clotting time is obviously
viewed by this program as a 'lack' of factor). Hence, clotting times in seconds
                                        157
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
have to be obtained from the ACL video screen (they are not reported on the
printout) and activity values intrapolated from the calibration curve using the
following linear function:
          y   =fix); where y =activity (%) and x =clotting time (s).
Notes
We suggest that plasma samples of unknown PS activity be run at two dilutions
instead of one. The 1:20 dilution (and higher) should be made independently
(Le. not by further diluting 1:2 the original 1: 10 dilution, but by making the
1:20 dilution directly) in PS-deficient plasma diluted 1: 10 (experience has shown
this is the best way to achieve sample curves parallel to the reference curve).
The reason for adopting additional dilutions is that dilution mistakes are
avoided or reduced by this practice, and that suspicion of artifacts which alter
PS activity is generated by the finding of non-parallel curves (this practice, for
example, allowed us to conclude that resistance to APC interfered with the PS
activity assay). When this happens the sample should be tested again at three
or four dilutions.
   Though the manufacturer suggests repeating the assay when a correlation
coefficient < 0.98 for the reference curve is found, a better practice, especially
when setting up the assay ex novo, is to include a standard curve among the
samples. This is to say that the reference plasma should be diluted appropriately
and run as a sample, in order to verify two things:
1. that the clotting times corresponding to points in the curve generated by
   the machine are the same as those generated by the added reference plasma
   dilutions;
2. that the reference curve is calculated on a sufficient number of points (0%,
   50%,75%,100%) in the newer models.
If machine- and hand-generated curves are similar after a few trial runs, the
curve generated by the machine may be used. A control run as described should,
however, be performed on a regular basis.
men and women have been adopted both for antigen and activity of free PS.
At present in our laboratory the reference range for men is 76-166% and for
women is 58-143%. Obviously, carriers of factor V Leiden were excluded from
the calculation.
Pitfalls
Besides the already-mentioned interference of resistance to APC and anti-
phospholipid antibodies in the PS activity assay, elevated factor VIla levels
can also shorten the prothrombin-time-based clotting assay leading to
underestimation of PS activity 17. It is often suggested, in case of interference,
to perform the assay at higher dilutions and to consider as valid the result
obtained at the highest dilution (in the belief that the interfering factor is
'diluted out'). We find this is not good laboratory practice. It is difficult to
know a priori the potency of the interfering factor, and consequently if and
when it will be diluted out. In addition, non-parallel curves may theoretically
be found in the presence of interferences by factors as yet unknown (as was
the case for interference by resistance to APC before the latter was described).
In all these cases the best procedure is simply not to measure PS activity by
plasma-based methods.
   Low levels of PS activity may be an inherited or an acquired trait. Acquired
PS activity deficiency is usually associated with low free PS levels as in liver
disease, oral anticoagulant treatment, pregnancy, hormone replacement theragr'
the nephrotic syndrome, cancer and post-infectious anti-PS antibodies',24-= .
   In summary, a diagnosis of inherited PS deficiency should never be based
on an activity test performed only once at a single dilution. A pathological
result should be confirmed at multiple dilutions after ruling out known interfer-
ences and acquired deficiencies. If measurement of free PS is not performed
in parallel, or in any case when a dysfunctional defect is suspected, genetic
analysis is recommended.
Alternative methods
Several methods for measuring PS activity have been reported. All are based
on the measurement of the cofactor activity of PS to APC 28-35. What changes
from one method to the other is the type of clotting assay used. Two are
commercialized, by Behring (Protein S Reagent, Dade Behring) and by
Diagnostica Stago (Staclot protein S). The method described by D'Angelo et
al. 2 is the only one that requires extraction of PS from plasma by anti-PS
monoclonal antibodies linked to beads; after elution the PS activity is determined
in a Xa-based clotting assay and its concentration is measured by ELISA. This
method has several advantages over others: first, it is not sensitive to interference
by substances present in plasma; second, a real specific activity of PS can be
measured, allowing the correct detection of type II deficiencies. However, the
drawback of this method is the requirement of the monoclonal antibody and
the difficulty in automation.
                                        159
           LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
COMMENTS
References
 1. Dahlbiick B. The protein C anticoagulant system: inherited defects as basis for venous
    thrombosis. Thromb Res 1995; 77: 1-43.
 2. D'Angelo A, Vigano-D'Angelo S, Esmon CT, Comp Pc. Acquired deficiency of protein S.
    Protein S activity during oral anticoagulation, in liver disease, and in disseminated intravascular
    coagulation. J Clin Invest 1988; 81: 1445-54.
 3. Garcia de Frutos P, Alim RIM, Hiirdig Y, ZOller B, Dahlbiick B. Differential regulation of a
    and fl chains of C4b-binding protein during acute-phase response resulting in stable plasma
    levels of free anticoagulant protein S. Blood 1994; 84: 815-22.
 4. de Fouw NJ, Haverkate F, Bertina RM, Koopman J, van Wijngaarden A, van Hinsbergh
    VWM. The cofactor role of PS in the acceleration of whole blood clot lysis by activated
    protein C in vivo. Blood 1986; 67: 1189-92.
 5. Godowski PJ, Mark MR, Chen J, Sadick MD, Raab H, Hammonds RG. Reevaluation of the
    roles of protein S and Gas 6 as ligands for the receptor tyrosine kinase Rserryro 3. Cell 1995;
    82: 355-8.
 6. Zoller B, Garcia de Frutos P, Dahlbiick B. Evaluation of the relationship between protein S
    and C4b-binding protein isoforms in hereditary protein S deficiency demonstrating type I and
    type III deficiencies to be phenotypic variants of the same genetic disease. Blood 1995; 85:
    3524-31.
 7. Simmonds RE, Zoller B, Ireland H, Thompson E, de Frutos PG, Dahlbiick B, Lane DA.
    Genetic and phenotypic analysis of a large (122) member protein S-deficient kindred provides
    an explanation for the familial coexistence of type I and type III plasma phenotypes. Blood
    1997;89:4364-70.
 8. Rossi E, Gatti L, Guarneri D, Finotto E, Lombardi A, Preda L. Functional protein S in
    women with lupus anticoagulant inhibitor. Thromb Res 1992; 65: 253-62.
 9. Lawrie AS, Lloyd ME, Mohamed F, Irons S, Hughes GR, Savidge GF. Assay of protein S in
    systemic lupus erythematosus. Blood Coag Fibrinol1995; 6: 322-4.
10. Faioni EM, Franchi F, Asti D, Sacchi E, Bernardi F, Mannucci PM. Resistance to activated
    protein C in nine thrombophilic families: interference in a protein S functional assay. Thromb
    Haemostas 1993; 70: 1067-71.
II. Faioni EM, Boyer-Neumann C, Franchi F, Wolf M, Meyer D, Mannucci PM. Another protein
    S functional assay is sensitive to resistance to activated protein C. Thromb Haemostas 1994;
    72: 648.
12. Cooper PC, Hampton KK, Makris M, Abuzenadah A, Paul B, Preston FE. Further evidence
    that activated protein C resistance can be misdiagnosed as inherited functional protein S
    deficiency. Br J HaematoI1994; 88: 201-3.
13. Simioni P, Gavasso S, Luni S, Invidiato S, Girolami A. A protein S functional assay yields
    unsatisfactory results in patients with activated protein C resistance. Blood Coag Fibrinol
    1995; 6: 286-7.
14. Lane DA, Mannucci PM, Bauer KA, Bertina RM, Bochkov NP, Boulyjenkov V, Chandy M,
    Dahlbiick B, Ginter EK, Miletich JP, Rosendaal FR, Seligsohn U. Inherited thrombophilia:
    Part 1. Thromb Haemostas 1996; 76: 651-62.
IS. Gandrille S, Borgel D, Ireland H, Lane DA, Simmonds R, Reitsma PH, Mannhalter C,
    Pabinger I, Saito H, Suzuki K., Formstone C, Cooper DN, Espinosa Y, Sala N, Bernardi F,
                                                160
                               PROTEIN S ACTIVITY ASSAYS
      Aiach M. Protein S deficiency: a database of mutations. For the Plasma Coagulation
      Inhibitors Subcommittee of the Scientific and Standardization Committee of the Inter-
      national Society on Thrombosis and Haemostasis. Thromb Haemostas 1997; 77: 1201-14.
16.   Lane DA, Mannucci PM, Bauer KA, Bertina RM, Bochkov NP, Boulyjenkov V, Chandy M,
      Dahlbiick B, Ginter EK, Miletich JP, Rosendaal FR, Seligsohn U. Inherited thrombophilia:
      Part 2. Thromb Haemostas 1996; 76: 824-34.
17.   Preda L, Tripodi A, Valsecchi C, Lombardi A, Finotto E, Mannucci PM. A prothrombin
      time-based functional assay of protein S. Thromb Res 1990; 60: 19-32.
18.   Maccaferri M, Legnani C, Preda L, Palareti G. Protein S activity in patients with heredofamilial
      protein S deficiency and in patients with juvenile venous thrombosis. Results of a functional
      method. Thromb Res 1991; 64: 647-58.
19.   Boyer-Neumann C, Bertina RM, Tripodi A, D'Angelo A, Wolf M, Vigano D'Angelo S,
      Mannucci PM, Meyer D, Larrieu Ml Comparison of functional assays for protein S: European
      collaborative study of patients with congenital and acquired deficiency. Thromb Haemostas
      1993; 70: 946-50.
20.   Mannucci PM, Valsecchi C, Krachmalnicoff A, Faioni EM, Tripodi A. Familial dysfunction
      of protein S. Thromb Haemostas 1989; 62: 763-6.
21.   Hubbard AR. Standardisation of protein S in plasma: calibration of the 1st International
      Standard. Thromb Haemostas 1997; 78: 1237-41.
22.   Tait RC, Wright EM, Walker ID. Influence of age and sex on the diagnosis of protein S
      deficiency. Thromb Haemostas 1997; 78 (Suppl.): 161 (abstract no. PD-659).
23.   Faioni EM, Valsecchi C, Palla A, Taioli E, Razzari C, Mannucci PM. Free protein S deficiency
      is a risk factor for venous thrombosis. Thromb Haemostas 1997; 78: 1343-6.
24.   Comp PC, Thurnau GR, Welsh J, Esmon CT. Functional and immunologic protein S levels
      are decreased during pregnancy. Blood 1986; 68: 881-5.
25.   Vigano-D'Angelo S, D'Angelo A, Kaufman MJ, Sholer C, Esmon CT, Comp PC. Protein S
      deficiency occurs in the nephrotic syndrome. Ann Intern Med 1987; 107: 42-7.
26.   MaIm J, Laurell M, Dahlbiick B. Changes in the plasma levels of vitamin K-dependent proteins
      C and S and of C4b-binding protein during pregnancy and oral contraception. Br J Haematol
      1988; 68: 437-43.
27.   D'Angelo A, Mazzola G, Bergmann F, Safa 0, Della Valle P, Vigano D'Angelo S. Autoimmune
      protein S deficiency: a disorder predisposing to thrombosis. Haematologica 1995; 80 (Suppl.):
      114-21.
28.   Comp PC, Doray D, Patton D, Esmon CT. An abnormal plasma distribution of protein S
      occurs in functional protein S deficiency. Blood 1986; 67: 504-8.
29.   Kamiya T, Sugihara T, Ogata K, Saito H, Suzuki K, Nishioka J, Hashimoto S, Yamagata K.
      Inherited deficiency of protein S in a Japanese family with recurrent venous thrombosis: a
      study of three generations. Blood 1986; 67: 406-10.
30.   van de Waart P, Preissner KT, Bechtold JR, Miiller-Berghaus G. A functional test for protein
      S activity in plasma. Thromb Res 1987; 48: 427-37.
31.   Suzuki K, Nishioka 1 Plasma protein S activity measured using Protac, a snake venom derived
      activator of protein C. Thromb Res 1988; 48: 241-51.
32.   Schwarz HP, Muntean W, Watzke H, Richter B, Griffin JH. Low total protein S antigen but
      high protein S activity due to decreased C4b-binding protein in neonates. Blood 1988; 71:
      562-5.
33.   Wolf M, Boyer-Neumann C, Martinoli JL, Leroy-Matheron C, Amiral J, Meyer D, Larrieu
      MI. A new functional assay for human protein S activity using activated factor V as substrate.
      Thromb Haemostas 1989; 62: 1144-5.
34.   Kobayashi I, Anemiya N, Endo T, Okuyama K, Tamura K, Kume S. Functional activity of
      protein S determined with use of protein C activated by venom activator. Clin Chem 1989;
      35: 1644-8.
35.   Wiesel M-L, Charmantier J-L, Freyssinet J-M, Grunebaum L, Schuhler S, Cazenave J-P.
      Screening of protein S deficiency using a functional assay in patients with venous and arterial
      thrombosis. Thromb Res 1990; 58: 461-8.
36.   Heeb MJ, Rosing J, Bakker HM, Fernandez JA, Tans G, GrifTm JH. Protein S binds to and
      inhibits factor Xa. Proc Nat! Acad Sci USA 1994; 91: 2728-32.
                                                 161
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
37. Heeb MJ, Mesters RM, Tans G, Rosing J, Griffin JH. Binding of protein S to factor Va
    associated with inhibition of prothrombinase that is independent of activated protein C. J
    Bioi Chern 1993; 268: 2872-7.
38. Hackeng TM, van't Veer C, Meijers JCM, Bouma BN. Human protein S inhibits prothrombinase
    complex activity on endothelial cells via a direct interaction of protein S with factor Va and
    Xa. Evidence for an activated protein C independent anticoagulant function of protein S in
    plasma. J Bioi Chern 1994; 269: 21051-8.
39. Koppelman SJ, Hackeng TM, Sixma JJ, Bouma BN. Inhibition of the intrinsic factor X
    activating complex by protein S: evidence for a specific binding of protein S to factor VIII.
    Blood 1995; 86: 1062-71.
40. van Wijnen M, van't Veer C, Meijers JCM, Bertina RM, Bouma BN. A plasma coagulation
    assay to determine the activated protein C independent anticoagulant activity of protein S.
    Thromb Haemostas 1997; 78 (Suppl.): 187 (abstr. no. OC-756).
                                              162
17
Activated protein C (APC) resistance
A. TRIPODI
INTRODUCTION
antibodies still remains a problem, although some methods have been reported
to prevent interference ,13. An additional advantage of using the above
modification was shown in a recent large collaborative study designed to compare
the diagnostic efficacy of 13 plasma-based clotting methods for their abilit.(
to detect the FV:Q506 mutation in comparison with that of the DNA analysis I .
The results showed that predilution of test plasmas with FV-deficient plasma
considerably improved the ability of APTT-based methods to discriminate
carriers from non-carriers of the mutation l4 . However, to make a decision
about how to organize a diagnostic work-up in the thrombophilia laboratory,
one must also take into consideration that plasma-based methods (namely,
APTT-based methods) when performed on undiluted test plasmas are, at least
in principle, able to detect acquired APC resistance (Le. not due to the FV:Q506
mutation), whereas predilution of test plasma in FV-deficient plasma and DNA
analysis are not. Whether acquired APC resistance, which has been reported
to be fre~uent in pregnancyl5, oral contraceptive intakeS,I5--17, increased FVIII
activityl ,19 and cerebrovascular diseases20 is clinically important, and therefore
deserves to be detected, is still a matter of debate. Recently, van der Born et
al. 2o have shown in a population-based case--control study that a reduced
response to APC, independent of the FV:Q506 mutation, is associated with an
increased risk of cerebrovascular disease (the lower the APC response, the
higher the odds ratio for cerebrovascular disease). Should this finding be
confirmed in subsequent studies and/or extended to venous thromboembolism,
graded APC responses measured by APTT-based methods on undiluted plasma
could be useful to assess more accurately the thrombotic risk in thrombophilic
patients. On the basis of the above considerations, the following sections of
this chapter will be mainly devoted to the description of the APTT-based
method. Additional information for setting up other methods for APC resistance
and for DNA analysis for the FV:Q506 mutation can be found in the refer-
ences cited.
APTT·BASED METHOD
This method measures the in-vitro anticoagulant response of test plasma to APC.
Principle
Test plasmas are incubated with a suitable preparation of commercial or
home-made APTT reagent (activator plus phospholipids) for a standard period
of time (see below) to activate the contact phase. Coagulation is then initiated
by the addition of CaCl2 in the absence and in the presence of APC, and the
time for clot formation is recorded.
Reagents
Most commercially available reagents are suitable. However, reaction condi-
tions described below apply only to the brand of reagents mentioned here.
Optimal conditions should be established for each reagent.
                                       164
                               APC RESISTANCE
1. APT[ reagent (Automated APT[ reagent, Organon Teknika).
2. CaCl2 butTer (PH 7.5): 10 mmoVL Tris-HCl, 50 mmoVL NaCl, 30 mmoVL
   CaCI2 , 0.1% bovine serum albumin.
3. APC/CaClrbuffer. 15 nmollL APC preparation (Enzymes Research
   Laboratories) in CaCl2 butTer.
Procedure
This applies to the manual (tilt-tube) technique used in combination with the
above reagents. Most commercially available coagulometers can be used in the
APTTmode.
1. Place the necessary glass test tubes in a water bath at 37°C.
2. Pipette 0.1 m1 undiluted test plasma and 0.1 m1 APTT reagent. Incubate for
   3 min.
3. Pipette 0.1 m1 CaCl2 butTer and simultaneously start a stopwatch. Mix and
   tilt the tube back and forth regularly until clot forms, and record the clotting
   time.
4. Repeat the measurement for the same plasma with APC/CaCl2 butTer instead
   of CaCl2 butTer.
The above procedure can also be carried out on plasma prediluted 1:5 in
FV-deficient plasma (STA Factor V, Diagnostica Stago).
   Results are usually expressed as ratio of clotting time with APC to clotting
time without APC [APC sensitivity (APC-S) ratio]. The value of normalization
by a pooled normal plasma will be discussed in the next section. Many labora-
tories use a commercial APTT-based method (Chromogenix). This follows
from the method originally described by Dahlback and colleagues!
Interpretation of results
Lower than normal APTT APC-S ratios are to be expected in the following
situations: heterozygous and homozygous FV:Q506 mutation; FV haplotype
HR2; women on oral contraceptives; subjects with high FVIII activity and
patients with cerebrovascular disease. Very low APTT APC-S ratios mimicking
homozygosity for the FV:Q506 mutation can be found in heterozygous patients
who also carry a quantitative FV defect2 ! . Results are difficult to interpret for
patients on oral anticoagulants, on heparin, or with lupus anticoagulants and
clotting factor deficiencies.
Expression of results
                          APTT-based ~      No
                                     ~ APC-resistance
                             Borderline
                      /
                          FV-Def. Plasma+INormall--   No
                                  ~APc-resistance
                    IAbn~            ~
                             DNA analysis
Figure 17.1 Flow chart for APe resistance testing
                                            167
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
false-negative results for the FV:Q506 mutation. If this is not the case, the use
of the modified APTT-based method (i.e. predilution of test plasma in
FV-deficient plasma) is recommended.
References
 1. Dahlbiick B, Carlsson M, Svensson PJ. Familial thrombophilia due to a previously UIIIeCOgnized
    mechanism characterized by poor anticoagulant response to activated protein C: prediction
    of a cofactor to activated protein C. Proc Natl Acad Sci USA 1993; 90: 1004-8.
 2. Exner T, Murray B, Chong BH, Chesterman CN. Improved APC resistance method based
    on a Russell viper venom clotting test. Thromb Haemostas 1995; 73: 119.
 3. Le DT, Greengard JS, Mujumdar V, Rapaport SI. Use of a generally applicable tissue factor-
    dependent factor V assay to detect activated protein C-resistant factor Va in patients receiving
    warfarin and in patients with a lupus anticoagulant. Blood 1995; 85: 1704-11.
 4. Ahlenc-Gelas M, Aillaud MF, Bonvarlet MN, Dupuy G, Juhan-Vague I, Aiach M. Specificity
    of an assay based on a factor V-depleted plasma in patients carrying the Arg 506 Gn mutation.
    Thromb Haemostas 1996; 75: 971-7.
 5. Bertina RM, Koeleman BPC, Koster T, Rosendaal FR, Dirven RJ, de Ronde H, van der
    Veblen PA, Reitsma PH. Mutation in blood coagulation factor V associated with resistance
    to activated protein C. Nature 1994; 369: 64-7.
 6. Bernardi F, Faioni EM, Castoldi E, Lunghi B, Castaman G, Sacchi E, Mannucci PM. Factor
    V genetic component different from factor V R506Q contributes to the activated protein C
    resistance phenotype. Blood 1997; 90: 1552-7.
 7. Varadi K, Moritz B, Lang H, Bauer K, Preston E, Peake I, Rivard GE, Keil B, Schwarz HP.
    A chromogenic assay for activated protein C resistance. Br J Haematol1995; 90: 884-9.
 8. Rosing J, Tans G, Nicolaes GAF, Thomassen MCLGD, van Oerle R, van der Ploeg PMEN,
    HeYnen P, Hamulyak K, Hemker He. Oral contraceptives and venous thrombosis: different
    sensitivities to activated protein C in women using second- and third-generation oral contracep-
    tives. Br J Haemato11997; 97: 233-8.
 9. de Ronde H, Bertina RM. Laboratory diagnosis of APe resistance: a critical evaluation of
    the test and the development of diagnostic criteria. Thromb Haemostas 1994; 72: 880-6.
10. Halbmayer DM, Hanshofer A, Schon R, Fisher M. Influence of lupus anticoagulant on a
    commercially available kit for APC-resistance. Thromb Haemostas 1994; 72: 645-6.
II. Jorquera n, Montoro JM, Angeles Fernandez M, Aznar JA, Aznar J. Modified test for
    activated protein C resistance. Lancet 1994; 344: 1162-3.
12. Trossaert M, Conard J, Horellou MH, Samama MM, Ireland H, Bayston FA, Lane DA.
    Modified APC-resistance assay for patients on oral anticoagulants. Lancet 1994; 344: 1709.
13. Martorell JR, Munoz-Castillo A, Gil JL. False positive activated protein C resistance test due
    to antiphospholipid antibodies is corrected by platelet extract. Thromb Haemostas 1995; 74:
    796-7.
14. Tripodi A, Negri B, Bertina RM, Mannucci PM. Screening of the FV:Q506 mutation. Evalu-
    ation of thirteen plasma-based methods for their diagnostic efficacy in comparison with DNA
    analysis. Thromb Haemostas 1997; 77: 436-9.
IS. Hellgren M, Svensson PJ, Dahlbii.ck B. Resistance to activated protein C as a basis for venous
    thromboembolism associated with pregnancy and oral contraceptives. Am J Obstet Gynecol
    1995; 173:210-13.
16. Olivieri 0, Friso S, Manzato F, Guella A, Bernardi F, Lunghi B, Girelli D, Azzini M, Brocco
    G, Russo C et al. Resistance to activated protein C in healthy women taking oral contracep-
    tives. Br J Haematol1995; 91: 465-70.
17. Henkens CMA, Bom VJJ, Seinen AJ, van der Meer J. Sensitivity to activated protein C;
    influence of oral contraceptives and sex. Thromb Haemostas 1995; 73: 402-4.
18. Henkens CMA, Bom VJJ, van der Meer J. Lowered APe-sensitivity ratio related to increased
    factor VIII clotting activity. Thromb Haemostas 1995; 74: 1198-9.
19. Laffan MA, Manning R. The influence of factor VIII on measurement of activated protein
    C resistance. Blood Coagul Fibrinol1996; 7: 761-5.
                                               168
                                     APC RESISTANCE
20. van der Born JG, Bots ML, Haverkate F, Slagboom PE, Meijer P, de Jong PTVM, Hofman
    A. Grobbee PE, Kluff C. Reduced response to activated protein C is associated with increased
    risk for cerebrovascular disease. Ann Intern Med 1996; 125: 265-9.
21. Simioni P, Scudeller A, Radossi P, Gavasso S, Girolami B, Tormene D, Girolami A. 'Pseudo
    homozygous' activated protein C resistance due to double heterozygous factor V defects
    (factor V Leiden mutation and type I quantitative factor V defect) associated with thrombosis:
    report of two cases belonging to two unrelated kindreds. Thromb Haemostas 1996; 75: 422-6.
22. Sidelman J, Gram J, Pedersen OD, Jespersen 1. Influence of plasma platelets on activated
    protein C resistance assay. Thromb Haemostas 1995; 74: 993-4.
23. Shizuka R, Kauda T, Amagai H, Kobayashi I. False positive activated protein C sensitivity
    ratio (APe). Sensitivity ratio caused by freezing and by contamination of plasma with platelets.
    Thromb Res 1995; 78: 189-90.
24. Bertina RM. Laboratory diagnosis of resistance to activated protein C. Thromb Haemostas
    1997; 78: 478-82.
25. Tripodi A, Chantarangkul V, Negri B, Mannucci PM. Standardization of the APC-resistance
    test. Effects of norma1ization of results by means of pooled normal plasma. Thromb Haemostas
    1998; 79: 564-6.
                                               169
18
Tissue factor pathway inhibitor
(TFPI)
P. M. SANDSET
INTRODUCTION
Physiological roles
Inhibition of factor Xa and the tissue factor/factor Vila complex
The target proteases of TFPI activity are factor VIla and factor Xa. Inhibition
is mediated by interaction of the protease with the first and second Kunitz
domains, respectively6. TFPI inhibits factor Xa directly7,8, whereas inhibition
of the tissue factor (TF)NIIa complex requires the presence of factor Xa and
calcium ions6,8. Inhibition of the TFNIIa complex is commonly thought to
proceed in two steps, first by the formation of a TFPI/Xa complex and then
                                       171
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
the building of a quaternary TFPIlXafTFNIIa complex7,8. An alternative
hypothesis is that TFPI binds directly to the XafTFNIIa activator complex.
At supra-physiological concentrations, TFPI may also inhibit TFMIa complexes
in the absence of factor Xa9, which may be clinically relevant when recombinant
TFPI is considered for use as a therapeutic agent.
   The TFNIIa catalytic complex cleaves both factor IX and factor X10. Gener-
ation of factor Xa produces rapid feedback inhibition of the TFMIa complex,
but amplification of coagulation through the intrinsic cascade (factors VIII,
IX, and XI) normally provides sustained activation and efficient haemostasis.
In the absence of factor VIII and factor IX, TFPI effectively turns off activation
of coagulation, which explains why individuals with factor VIII or factor IX
deficiencies bleed II.
                                       172
                    TISSUE FACTOR PATHWAY INHIBITOR
Pathophysiological aspects
A number of studies have reported on the plasma levels of TFPI in various
clinical conditions. It does not behave as an acute-phase reactant26,27. Plasma
levels are significantly affected in patients with dyslipidaemia, with low levels
in patients with ~-li~oproteinaemia 13 and high levels in patients with
hypercholesterolaemia 8. Occasionally, low TFPI levels have been detected in
patients with severe disseminated intravascular coagulation (DIC), but it is
now evident that DIe may proceed despite normal or elevated plasma TFPI
levels13,27,29-31.
   Low levels of TFPI associated with a clear familial pattern have not been
detected, which may either indicate that inherited TFPI deficiency is rare or that
the approach to detect deficiency has not been appropriate 32 . Experimentally,
gene targeting techniques have been used to disrupt exon 4 of the TFPI gene in
mice33 . Homozygous animals suffered severe circulatory insufficiency, consumptive
coagulopathy, and died in utero. Heterozygous animals displayed normal phenotype
with plasma TFPI levels 50% of normal, but further studies are needed to rule
out the possibility that heterozygosity is associated with thrombosis.
   TFPI deficiency may be induced experimentally by the infusion of specific
antibodies or Fab fragments against TFPI34-36. Immunodepletion of TFPI in
the rabbit to values less than 20% of normal dramatically reduced the threshold
by which TF34,3S, but not factor Xa36, could produce severe DIe. These studies
indicate that there is a threshold above which TFPI can prevent the coagulation-
triggering effect of TF, and that TFPI is not a significant inhibitor of factor
Xa in vivo.
TFPIASSAYS
The development of assays for TFPI is severely complicated by the fact that
TFPI is contained in different intravascular pools and that TFPI is heterogeneous
both with regard to truncation and association with lipoproteins. The question
of assay is therefore not only a matter of assay methodology but also a matter
of what to assay. One may ask whether the assay of TFPI in a randomly
collected plasma sample, which contains mainly truncated forms of TFPI
associated with lipoproteins, can yield useful information on the functional
status of TFPI in that particular individual. The answer will require further
knowledge on the equilibrium between different pools of TFPI under various
physiological conditions and disease states, and knowledge of the biological
activity and role of the different forms of TFPI. At present a pragmatic approach
is to assay TFPI in plasma collected before, and 5-15 min after, a standardized
dose of unfractionated heparin, e.g. 5000 IV or 50-100 IV/kg, to acquire
information on resting plasma TFPI levels and on heparin-releasable TFPI.
The latter procedure should still be considered an experimental procedure as
part of a research protocol.
    Basically, four types of assays can be recognized:
1. TFPI end-point activity assay,
2. TFPI coagulant activity assay,
                                       173
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
3. immunological assay for total TFPI,
4. immunological assay for free TFPL
Activity assays can be constructed to assay the ability of TFPI to inhibit
TFNIIa catalytic activity or to inhibit factor Xa. Since the physiological role
of TFPI as a factor Xa inhibitor is questionable, this chapter will only describe
assays for TFNIIa inhibitory activity. Our in-house chromogenic substrate
assay37, adapted for use with commercial reagents, will be described in detail.
Assay of anticoagulant activity, and free and total antigen, will be described
only briefly.
Materials
Buffers
1. Stock buffer: Tris-saline-citrate (TSC) buffer: 50 mmollL Tris . HCl,
   100 mmollL NaCl, 10 mmollL Na 3 citrate (pH 7.5). Sodium azide may be
   added to 0.02% to keep the buffer stable at 4 °C for several months.
                                      174
                    TISSUE FACTOR PATHWAY INHIBITOR
2. Assay buffer (TSCIBSA buffer): TSC buffer containing bovine serum albumin
   (BSA) to final concentration 2 mg/ml. The assay butTer is stable for 7 days
   at 4 dc. It is used to dilute all reagents used in the assay.
3. Dilution buffer: TSC/BSA buffer containing polybrene (hexadimethrine
   bromide, Sigma), final concentration 2 Ilg/ml, to neutralize heparin present
   in the samples.
Reaction mixture
The following reagents, and procedures (1-4), are recommended for the
preparation of a combined reaction mixture (5). All reagent should be kept in
ice water during preparation:
1. Factor VIla: purified from human plasma (from Stago) or human,
   recombinant material (Novo-Nordisk). Lyophilized material is first diluted
   as described by the manufacturer, then further diluted in ice-cold assay
   butTer to final concentration 62.5 ng/ml (1.25 nmollL), and stored in suitable
   aliquots (e.g. 250 jJl) at -70°C. Before assay, aliquots are thawed and diluted
    lO-fold in assay butTer to final concentration 6.25 ng/ml (125 pmollL).
2. Tissue factor: human, recombinant, relipidated TF (Innovin, Ortho).
   Lyophilized material is first diluted as described by the manufacturer and
   is stable for 1-2 weeks at 4 °C (may not be frozen). The preparation is further
   diluted 20-fold in assay buffer immediately before preparation of the reaction
   mixture (see below).
3. Factor Xa: bovine material (from Chemogenix). Lyophilized material is first
   diluted as described by the manufacturer and further diluted in ice-cold assay
   buffer to final concentration 3.3 nkatlml (0.25 U/ml or 40 nmollL) and stored
   in suitable aliquots (e.g. 250 jJl) at -70°C. Before assay, aliquots are thawed
   and diluted lO-fold in assay butTer to final concentration 0.33 nkatlml
   (4 nmollL).
4. CaCl2 : 75 mmollL.
5. Combined reaction mixture: may be prepared immediately before assay by
   mixing equal volumes of factor VIla, TF, factor Xa, and CaCl2 prediluted
   as described above (1-4). A convenient volume sufficient for one microtitre
   plate is 2.5 ml of each, total 10 ml.
Substrates
The following procedure is recommended for preparation of substrate reagents:
1. Factor X: bovine material (Chromogenix). The reagent must not contain
   factor Xa activity. Lyophilized material is first diluted as described by the
   manufacturer and further diluted in ice-cold assay buffer to final concentration
   0.4 U/ml (64 mmollL) and stored in suitable aliquots at -70°C.
2. S-2222: chromogenic substrate (Chromogenix). Lyophilized material should
   be dissolved in distilled water to final concentration 2 mg/ml (2.7 mmollL).
   Provided it is protected from light, the solution is stable for 6 months at
   4°C.
                                       175
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Stop reagent
Acetic acid 50%.
Other materials
Flat-bottomed microtitre plates are purchased from Dynatech, but other plates
may also be used after analysis of their suitability for the assay. Absorbances
are read on a conventional multichannel analyser using a filter of 405 nm (e.g.
Thermomax, Molecular Devices, CA, USA).
Test procedure
Test protocol
Suitable dilutions of standards, controls, and test plasma (25 J.Ll) are added to
the wells. The combined reaction mixture (100 J.Ll) containing factors VIla and
Xa, TF and CaCl2 (see Reaction mixture, above) is then added to each well
                                       176
                    TISSUE FACTOR PATHWAY INHIBITOR
using a multichannel pipette. The micro titre plate is sealed using Parafilm and
placed in an incubator on gentle shaking at 37 °C for 30 min. Substrate reagent
mixture (50~) containing factor X and S-2222 (see Substrates, above) is then
added to each well using a multichannel pipette and the plate is further incubated
at 37 °C for approximately 30 min (until sufficient colour has developed, i.e.
OD405 > 1.2 in buffer control). The reaction is quenched by the addition of
50% acetic acid (50 ~). Finally, the absorbance at 405 nm is read in a multi-
channel photometer (see Other materials, above).
Calibration
The assay is calibrated with dilutions of PNP in assay buffer (see Buffers,
above). An eight-point calibration curve is made by pipetting 25 ~ in duplicate
wells of 4% (1125),3%,2% (1/50), 1.5%, 1% (11100), 0.5%, 0.25%, and 0%
plasma, which is diluted 5-fold by the addition of the reaction mixture to final
concentrations 0-0.8% plasma. Routinely, plasma samples from patients not
receiving heparin are tested in duplicates in 1150 and/or 11100 dilutions, while
plasmas of patients treated with heparin are tested in duplicates in 11100 and/or
11200 dilutions. Testing of the samples in two dilutions is recommended. Each
run should include one normal and one high-level control (see Calibrators,
controls and samples, above). The intra-assay CV should be kept below 4-6%
and inter-assay CV below 8-10%.
   TFPI activity is calculated from the standard curve which is obtained by a
quadratic fit of the data plotted on a linear scale, and is expressed as a percentage
or in units/mI, where I unit is defined as the TFPI activity present in I mI PNP.
Normal reference levels determined with this method include a wide range with
individual values from 0.45 to 1.75 Ulmi. The activity increases with age and
is positively correlated with total cholesterol37 •
TFPI ANTIGEN
References
 1. Wun TC, Kretzmer KK, Girard TJ, Miletich JP, Broze GJ, Jr. Cloning and characterization
    of a cDNA coding for the lipoprotein-associated coagulation inhibitor shows that it consists
    of three tandem Kunitz-type inhibitory domains. J Bioi Chem 1988; 263: 6001-4.
 2. Wesselschmidt R, Likert K, Girard T, Wun TC, Broze GJ, Jr. Tissue factor pathway inhibitor:
    the carboxy-terminus is required for optimal inhibition of factor Xa. Blood 1992; 79: 2004--10.
 3. Wesselschmidt R, Likert K, Huang Z, MacPhail L, Broze GJ, Jr. Structural requirements for
    tissue factor pathway inhibitor interactions with factor Xa and heparin. Blood Coagul Fibrinol
    1993; 4: 661-9.
 4. Bajaj MS, Kuppuswamy MN, Saito H, Spitzer SG, Bajaj SP. Cultured normal human
    hepatocytes do not synthesize lipoprotein-associated coagulation inhibitor: evidence that
    endothelium is the principal site of its synthesis. Proc Nat! Acad Sci USA 1990; 87: 8869-73.
 5. Warn-Cramer BJ, Almus FE, Rapaport SI. Studies of the factor Xa-dependent inhibitor of
    factor VILa/tissue factor (extrinsic pathway inhibitor) from cell supemates of cultured human
    umbilical vein endothelial cells. Thromb Haemostas 1989; 61: 101-5.
 6. Girard TJ, Warren LA, Novotny WF, Likert KM, Brown SG, Miletich JP, Broze GJ, Jr.
    Functional significance of the Kunitz-type inhibitory domains of lipoprotein-associated
    coagulation inhibitor. Nature 1989; 338: 518-20.
 7. Warn-Cramer BJ, Rao LV, Maid SL, Rapaport SI. Modifications of extrinsic pathway inhibitor
    (EPI) and factor Xa that affect their ability to interact and to inhibit factor VILa/tissue factor:
    evidence for a two-step model of inhibition. Thromb Haemostas 1988; 60: 453--6.
 8. Broze GJ, Jr, Warren LA, Novotny WF, Higuchi DA, Girard JJ, Miletich JP. The lipoprotein-
    associated coagulation inhibitor that inhibits the factor VII-tissue factor complex also inhibits
    factor Xa: insight into its possible mechanism of action. Blood 1988; 71: 335-43.
 9. Callander NS, Rao LV, Nordfang 0, Sandset PM, Warn-Cramer B, Rapaport SI. Mechanisms
    of binding of recombinant extrinsic pathway inhibitor (rEPI) to cultured cell surfaces. Evidence
    that rEPI can bind to and inhibit factor VIla-tissue factor complexes in the absence of factor
    Xa. J Bioi Chern 1992; 267: 876-82.
10. 0sterud B, Rapaport SI. Activation of factor IX by the reaction product of tissue factor and
    factor VII: additional pathway for initiating blood coagulation. Proc Nat! Acad Sci USA
    1977; 74: 5260-4.
11. Repke D, Gemmell CH, Guha A, Turitto VT, Broze GJ, Jr, Nemerson Y. Hemophilia as a
    defect of the tissue factor pathway of blood coagulation: effect of factors VIII and IX on
    factor X activation in a continuous-flow reactor. Proc Natl Acad Sci USA 1990; 87: 7623-7.
12. Sandset PM, Abildgaard U, Larsen ML. Heparin induces release of extrinsic coagulation
    pathway inhibitor (EPI). Thromb Res 1988; 50: 803-13.
13. Novotny WF, Brown SG, Miletich JP, Rader DJ, Broze GJ, Jr. Plasma antigen levels of the
    lipoprotein-associated coagulation inhibitor in patient samples. Blood 1991; 78: 387-93.
14. Hubbard AR, Jennings CA. Inhibition of the tissue factor-factor VII complex: involvement
    of factor Xa and lipoproteins. Thromb Res 1987; 46: 527-37.
                                                 179
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
15. Warn-Cramer BJ, Maid SL, Zivelin A, Rapaport SI. Partial purification and characterization
    of extrinsic pathway inhibitor (the factor Xa-dependent plasma inhibitor of factor VIla/tissue
    factor). Thromb Res 1987; 48: 11-22.
16. Novotny WF, Girard TJ, Miletich JP, Broze GJ, Jr. Purification and characterization of the
    lipoprotein-associated coagulation inhibitor from human plasma. J Bioi Chern 1989; 264:
    18832-7.
17. Hansen JB, Huseby NE, Sandset PM, Svensson B, Lyngmo V, Nordoy A. Tissue-factor
    pathway inhibitor and lipoproteins. Evidence for association with and regulation by LDL in
    human plasma. Arterioscler Thromb 1994; 14: 223-9.
18. Novotny WF, Girard TJ, Miletich JP, Broze GJ, Jr. Platelets secrete a coagulation inhibitor
    functionally and antigenically similar to the lipoprotein associated coagulation inhibitor.
    Blood 1988; 72: 2020-5.
19. Novotny WF, Palmier M, Wun TC, Broze GJ, Jr, Miletich JP. Purification and properties of
    heparin-releasable lipoprotein-associated coagulation inhibitor. Blood 1991; 78: 394-400.
20. Broze GJ, Jr, Lange GW, Duffin KL, MacPhail L. Heterogeneity of plasma tissue factor
    pathway inhibitor. Blood Coagul Fibrinol1994; 5: 551-9.
21. Hansen JB, Huseby KR, Huseby NE, Ezban M, Nordoy A. Tissue factor pathway inhibitor
    in complex with low density lipoprotein isolated from human plasma does not possess
    anticoagulant function in tissue factor-induced coagulation in vitro. Thromb Res 1997; 85:
    413-25.
22. Lindahl AK, Abildgaard U, Larsen ML, Staalesen R, Hammer AK, Sandset PM, Nordfang
    0, Beck TC. Extrinsic pathway inhibitor (EPI) released to the blood by heparin is a more
    powerful coagulation inhibitor than is recombinant EPI. Thromb Res 1991; 62: 607-14.
23. Lindahl AK, Jacobsen PB, Sandset PM, Abildgaard U. Tissue factor pathway inhibitor with
    high anticoagulant activity is increased in post-heparin plasma and in plasma from cancer
    patients. Blood Coagul Fibrinol1991; 2: 713-21.
24. Nordfang 0, Bjorn SE, Valentin S, Nielsen LS, Wildgoose P, Beck TC, Hedner U. The
    C-terminus of tissue factor pathway inhibitor is essential to its anticoagulant activity.
    Biochemistry 1991; 30: 10371-6.
25. Wun TC, Kretzmer KK, Palmier MO, Day KC, Huang MD, Welsch DJ, Lewis C, Wolfe RA,
    Zobel JF, Lange GW et af. Comparison of recombinant tissue factor pathway inhibitors
    expressed in human SK hepatoma, mouse C127, baby hamster kidney, and Chinese hamster
    ovary cells. Thromb Haemostas 1992; 68: 54-9.
26. Sandset PM, Hogevold HE, Lyberg T, Andersson TR, Abildgaard U. Extrinsic pathway
    inhibitor in elective surgery: a comparison with other coagulation inhibitors. Thromb Haemostas
     1989; 62: 856-60.
27. Warr TA, Rao LV, Rapaport SI. Human plasma extrinsic pathway inhibitor activity: II. Plasma
    levels in disseminated intravascular coagulation and hepatocellular disease. Blood 1989; 74:
    994-8.
28. Sandset PM, Lund H, Norseth J, Abildgaard U, Ose L. Treatment with hydroxyrnethylglutaryl-
    coenzyme A reductase inhibitors in hypercholesterolemia induces changes in the components
    of the extrinsic coagulation system. Arterioscler Thromb 1991; 11: 138-45.
29. Bajaj MS, Rana SV, Wysolmerski RB, Bajaj SP. Inhibitor of the factor VIla-tissue factor
    complex is reduced in patients with disseminated intravascular coagulation but not in patients
    with severe hepatocellular disease. J Clin Invest 1987; 79: 1874-8.
30. Brandtzaeg P, Sandset PM, Joe GB, 0vstebo R, Abildgaard U, Kierulf P. The quantitative
    association of plasma endotoxin, antithrombin, protein C, extrinsic pathway inhibitor and
    fibrinopeptide A in systemic meningococcal disease. Thromb Res 1989; 55: 459-70.
31. Sandset PM, Reise 0, Aasen AO, Abildgaard U. Extrinsic pathway inhibitor in postoperative!
    posttraumatic septicemia: increased levels in fatal cases. Haemostasis 1989; 19: 189-95.
32. Sandset PM, Bendz B. Tissue factor pathway inhibitor: clinical deficiency states. Thromb
    Haemostas 1997; 78: 467-70.
33. Huang ZF, Higuchi D, Lasky N, Broze GJ, Jr. Tissue factor pathway inhibitor gene disruption
    produces intrauterine lethality in mice. Blood 1997; 90: 944-51.
34. Sandset PM, Warn-Cramer BJ, Maki SL, Rapaport SI. Immunodepletion of extrinsic pathway
    inhibitor sensitizes rabbits to endotoxin-induced intravascular coagulation and the generalized
    Shwartzman reaction. Blood 1991; 78: 1496-502.
35. Sandset PM, Warn-Cramer BJ, Rao LV, Maki SL, Rapaport SI. Depletion of extrinsic pathway
                                               180
                          TISSUE FACTOR PATHWAY INHIBITOR
      inhibitor (EPI) sensitizes rabbits to disseminated intravascular coagulation induced with tissue
      factor: evidence supporting a physiologic role for EPI as a natural anticoagulant. Proc Natl
      Acad Sci USA 1991; 88: 708-12.
36.   Warn-Cramer BJ, Rapaport SI. Studies of factor XaJphospholipid-induced intravascular
      coagulation in rabbits. Effects of immunodepletion of tissue factor pathway inhibitor.
      ArteriosclerThromb 1993; 13: 1551-7.
37.   Sandset PM, Larsen ML, Abildgaard U, Lindahl AK, Odegaard OR. Chromogenic substrate
      assay of extrinsic pathway inhibitor (EPI): levels in the normal population and relation to
      cholesterol. Blood Coagul Fibrinol1991; 2: 425-33.
38.   Sandset PM, Abildgaard U, Pettersen M. A sensitive assay of extrinsic coagulation pathway
      inhibitor (EPI) in plasma and plasma fractions. Thromb Res 1987; 47: 389-400.
39.   Hubbard AR, Weller D, Gray E. Measurement of tissue factor pathway inhibitor in normal
      and post-heparin plasma. Blood Coagul Fibrinol1994; 5: 819-23.
40.   Berrettini M, Malaspina M, Parise P, Lucarelli G, Kisiel W, Nenci 00. A simple chromogenic
      substrate assay of tissue factor pathway inhibitor activity in plasma and serum. Am J Clin
      Patho11995; 103: 391-5.
41.   Boguacki J, Hammelburger J. Functional and immunologic methods for the measurement of
      human tissue factor pathway inhibitor. Blood Coagul Fibrinol1995; 6 (Suppl. 1): S65-72.
42.   Warr TA, Warn-Cramer BJ, Rao LV, Rapaport SI. Human plasma extrinsic pathway inhibitor
      activity: I. Standardization of assay and evaluation of physiologic variables. Blood 1989; 74:
      201-6.
43.   Nemerson Y. Tissue factor and hemostasis [published erratum appears in Blood 1988; 71:
      1178]. Blood 1988; 71: 1-8.
44.   Sandset PM, Andersson TR. Coagulation inhibitor levels in pneumonia and stroke: changes
      due to consumption and acute phase reaction. J Intern Med 1989; 225: 311-16.
45.   Abildgaard U, Lindahl AK, Sandset PM. Heparin requires both antithrombin and extrinsic
      pathway inhibitor for its anticoagulant effect in human blood. Haemostasis 1991; 21: 254-7.
46.   Nordfang 0, Valentin S, Beck TC, Hedner U. Inhibition of extrinsic pathway inhibitor shortens
      the coagulation time of normal plasma and of hemophilia plasma. Thromb Haemostas 1991;
      66: 464-7.
47.   Goodwin CA, Melissari E, Kakkar VV, Scully ME Plasma levels of tissue factor pathway
      inhibitor in thrombophilic patients. Thromb Res 1993; 72: 363-6.
48.   Hansen JB, Huseby KR, Huseby NE, Sandset PM, Hanssen TA, Nordoy A. Effect of
      cholesterol lowering on intravascular pools of TFPI and its anticoagulant potential in type
      II hyperlipoproteinemia. Arterioscler Thromb Vase Bioi 1995; 15: 879-85.
49.   Lindahl AK, Abildgaard U, Staalesen R. The anticoagulant effect in heparinized blood and
      plasma resulting from interactions with extrinsic pathway inhibitor. Thromb Res 1991; 64:
      155-68.
50.   Kokawa T, Enjyoji K, Kumeda K, Kamikubo Y, Harada-Shiba M, Koh H, Tsushima M,
      Yamamoto A, Kato H. Measurement of the free form of TFPI antigen in hyperlipidemia.
      Relationship between free and endothelial cell-associated forms of TFPI. Arterioscler Thromb
      Vasc Bioi 1996; 16: 802-8.
51.   0stergaard PB, Beck TC, 0rsted H, Svendsen A, Nordfang 0, Sandset PM, Hansen JB. An
      enzyme linked immunosorption assay for tissue factor pathway inhibitor. Thromb Res 1997;
      87:447-59.
52.   Hansen JB, Sandset PM, Huseby KR, Huseby NE, Nordoy A. Depletion of intravascular
      pools of tissue factor pathway inhibitor (TFPI) during repeated or continuous intravenous
      infusion of heparin in man. Thromb Haemostas 1996; 76: 703-9.
53.   Abumiya T, Enjyoji K, Kokawa T, Kamikubo Y, Kato H. An anti-tissue factor pathway
      inhibitor (TFPI) monoclonal antibody recognized the third Kunitz domain (K3) of free-form
      TFPI but not lipoprotein-associated forms in plasma. J Biochem (Tokyo) 1995; 118: 178-82.
                                                 181
19
Lupus anticoagulant
D. A. TRIPLETT
INTRODUCTION
                                        183
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                     Table 19.1 Antiphospholipid-protein antibodies
                     (APA)
patients less than 50 l.ears of age and 5-15% of women with recurrent
spontaneous abortions .7.
Abbreviations: APTf, activated partial thromboplastin time; KCT, kaolin clotting time; SCT,
silica clotting time; dPT dilute prothrombin time; dRVVT, dilute Russell viper venom time
                                           185
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
see if there is relative correction of the abnormal clotting times. A number of
sources of PL have been utilized including freeze-thawed platelets (platelet
neutralization procedure), excess PL, platelet vesicles, and hexagonal phase
PL8,17. When utilizing freeze-thawed platelets, or lyophilized platelets, heparin
may give a false-positive result (i.e. platelet factor 4 will neutralize heparin).
This may be a problem in the hospitalized patient in whom the presence of
heparin was not considered when an abnormal clotting study was obtained.
The hexagonal phase PL confirmatory study has proved to be quite sensitive
and specific for LA detection 17 . Hexagonal phase PL is part of an 'integrated
system' utilizing an APTT and incorporating in the sequential steps the addition
of a source of normal plasma as well as polybrene to neutralize heparin, if
present in the sample. Other examples of integrated tests include commercially
available dRVVT systems. These employ a sensitive dRVVT to screen and a
dRVVT reagent which contains high PL concentrations to confirm.
References
I. Triplett DA. Antiphospholipid-protein antibodies: laboratory detection and clinical relevance.
   Thromb Res 1995; 78: 1-31.
2. Love PE, Santoro SA. Antiphospholipid antibodies: anticardiolipin and the lupus anticoagulant
   in systemic lupus erythematosus (SLE) and in non-SLE disorders. Ann Intern Med 1990;
   112: 692-8.
3. Roubey RAS, Hoffman M. From antiphospholipid syndrome to antibody-mediated thrombosis.
   Lancet 1997; 350: 1491-3.
                                             186
                                 LUPUS ANTICOAGULANT
 4. Hughes GRY. The antiphospholipid syndrome: ten years on. Lancet 1993; 342: 341-4.
 5. Harris EN, Gharavi AE, Boey ML, Patel BM, MacWorth-Young CG, Loizou S, Hughes GRY.
    Anticardiolipin antibodies: detection by radioimmunoassay and association with thrombosis
    in systemic lupus erythematosus. Lancet 1983; 2: 1211-14.
 6. Ginsburg KS, Liang MH, Newcomer L, Goldhaber SZ, Schur PH, Hennekens CH, Stampfer
    MJ. Anticardiolipin antibodies and the risk for ischemic stroke and venous thrombosis. Ann
    Intern Med 1992; 117: 997-1002.
 7. Kittner SJ, Gorelick PB. Antiphospholipid antibodies and stroke: an epidemiological perspective.
    Stroke 1992; 23: 119-122.
 8. Brandt JT, Triplett DA, A1ving B, Scharrer I. Criteria for the diagnosis of lupus anticoagulants:
    an update. Thromb Haemostas 1996; 74: 1185-90.
 9. Triplett DA. Screening for the lupus anticoagulant. Res Clin Lab 1989; 19: 379-89.
10. Sletnes K, Graven K, WisloffF. Preparation of plasma for the detection of lupus anticoagulants
    and antiphospholipid antibodies. Thromb Res 1992; 66: 43-53.
II. Kaczor DA, Bickford NM, Triplett DA. Evaluation of different mixing study reagents and
    dilution effect in lupus anticoagulant testing. Am J Clin Patho11991; 95: 408-11.
12. Johns AS, Charnley L, Ockelford PA, Pattison NS, McKay EJ, Corkill M, Hart H. Comparison
    of tests for the lupus anticoagulant and antiphospholipid antibodies in systemic lupus
    erythematosus. Clin Exp Rheumatol1994; 12: 523-6.
13. Schjetlein R, Wisloff F. An evaluation of two commercial test procedures for the detection
    of lupus anticoagulant. Am J Clin Patho11995; 103: 108-11.
14. Triplett DA, Brandt JT, Maas RL. The laboratory heterogeneity of lupus anticoagulants.
    Arch Pathol Lab Med 1985; 109: 946-51.
15. Clyne LP, White PF. Time dependency of lupus-like anticoagulants. Arch Intern Med 1988;
    148: 1060-3.
16. Triplett DA. Simultaneous occurrence of lupus anticoagulant and factor VIII inhibitors. Am
    J Hematol1997; 56:195-6.
17. Triplett DA, Barna LK, Unger GA. A hexagonal (II) phase phospholipid neutralization assay
    for lupus anticoagulant identification. Thromb Haemostas 1993; 70: 787-93.
                                                187
20
Heparin cofactor II
S. J. BAUMAN and F. C. CHURCH
INTRODUCTION
STRUCTURAL ASPECTS
HCII is a 480 amino acid glycoprotein with a molecular weight of approximately
66000. HCII contains three possible N-glycosylation sites. At the amino terminal
end of the protein are two sulphated tyro sines. HCII contains three cysteines,
all of which are available for sulphydryl modification, indicating that none is
involved in disulphide bonds. The gene for HCII is located on chromosome
22. The protein is expressed in hepatocytes and secreted into the blood 7 ,8.
   The reactive site loop of the serpin is the structure recognized by the serine
                                        189
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
protease as a substrate. When the protease attempts to cleave this 'pseudo-
substrate' it becomes trapped in a stable 1:1 complex with the inhibitor. Hell
is unique among thrombin inhibitors because its reactive site bond is made of
the dipeptide Leu444-Ser445 as opposed to the usual Arg-Ser which are
designated as the PI-PI' residues9 . In particular, thrombin prefers Arg at the
PI position lO • Hell is a poor thrombin inhibitor in the absence of
glycosaminoglycan (2 x 104 M- I min-I). However, when glycosaminoglycans
such as heparin or dermatan sulphate are introduced to the inhibition reaction,
the rates increase dramatically to 3 x 108 and 6 x 108 M- I min-I, respectivelyli.
   The glycosaminoglycan binding region of Hell has been localized to the
D-helix of the molecule. The D-helix is made up of residues Lysl71 through
Phe195. There are six basic residues within this helix which form a positively
charged surface-exposed structure (Lys173, Arg184, Lys185, Arg189, Argl92,
and Arg193). Although not well defined, it seems as though the residues at the
beginning of the helix (Lys173, Arg184, and Lys185) are involved in binding
to heparin, while the latter amino acids (Lys185, Arg189, Arg192, and Arg193)
are involved in dermatan sulphate binding I2- 15 •
   Unlike other serpins, Hell contains a unique amino terminal structure. The
region between Gly54 and Asp75 contains two stretches of acidic amino acids.
This region, called the 'acidic domain', is similar to the carboxyl terminus of
the leech anticoagulant hirudin and the amino terminus of the tethered-ligand
thrombin receptor. Both of these similar structures are known to bind anion-
binding exosite-I of thrombin. An intramolecular interaction with the Rositively
charged D-helix and the acidic domain of Hell has been proposed 6,17.
   The acidic domain is an integral component of the proposed mechanism of
glycosaminoglycan accelerated thrombin inhibition by He1l 18 . When glyco-
saminoglycan is present in the inhibition reaction, it binds the D-helix of Hell
and displaces the acidic amino terminal domain. In addition to the interaction
between the reactive site loop of Hell and the active site region of thrombin,
the acidic domain is free to interact with anion-binding exosite-I of thrombin.
The multiple points of contact between Hell, thrombin and the
glycosaminoglycan are believed to be responsible for the increase in inhibition
rates.
PATHOPHYSIOLOGICAL ASPECTS
Although a definitive role for Hell has not been found in vivo, the involvement
of HCII, or lack thereof, has been studied in many different clinical scenarios.
Two genetic variations have been described. Hell Oslo was identified by Blinder
et al. 5. The Hell Oslo gene contained a point mutation at base 651 that
resulted in an Arg189- His mutation. This protein lost all ability to accelerate
thrombin inhibition in the presence of dermatan sulphate. This patient was
identified from a pool of blood donors as having decreased Hell activity in
the presence of dermatan sulphate but not heparin. Otherwise the patient was
asymptomatic. Hell Awaji was a result of a thymine insertion within the gene
that resulted in a frame-shift mutation l9 • The mutated gene produced a truncated
protein that was rapidly degraded. The patient with Hell Awaji presented with
                                       190
                             HEPARIN COFACTOR II
a thrombotic tendency, although his sister, who was positive for the same
mutation, was asymptomatic.
   In addition to genetic anomalies, changes in HCII levels have been
documented in many clinical conditions. Levels of HCII were reported to be
decreased in patients with disseminated intravascular coagulation, hepatic
failure, acute pancreatitis, human immunodeficiency virus, and diabetes type
I, as well as after elective surgeryl,20--23. Patients with chronic renal failure on
haemodialysis, and those with nephrotic syndrome, reportedly have increased
levels of HCII24,25. Females have higher levels of HCII than males. The levels
of HCII increase during pregnancy or oral contraceptive use26 . Preterm infants
have markedly decreased levels of HCII compared to term infants, who have
approximately 50% of adult HCII levels 27 .
Reference range
The normal concentration of heparin cofactor II for a healthy adult is 1.2 ±
0.4 (mean 2 SO) or approximately 80 IJg/mll. Healthy, full-term infants have
heparin cofactor II concentrations of approximately 50% of normal adult
values27 .
QUANTITATION
HCII can be assayed either when purified from plasma or directly in plasma
in a number of different ways. Antigen for HCII can be detected by elli.-ryme-
linked immunosorbent assay (ELISA), rocket immunoelectrophoresis (more
commonly referred to as Laurell rockets), and by Western immunoblotting
techniques. HCII activity can also be studied by looking at the ability to inhibit
thrombin in the presence of the glycosaminoglycan dermatan sulphate.
Methods of assay
ELISA for Hell
Principle
The principle of the HCII ELISA is to coat the microtitre plate with a polyclonal
antibody to HCII to capture the protein from the diluted sample. Next, an
HCII antibody that is conjugated to horseradish peroxidase is incubated in
each well. This antibody will react with the captured protein. A final reaction
is performed within each well to quantitate the amount of conjugate present
by its ability to develop a substrate. By comparing the amount of conjugate
in a series of standards with the test sample dilutions, an accurate amount of
HCII antigen can be quantitated. This assay is based on the method described
by Toulon et al. 28 .
Materials
1. Carbonate buffer: this buffer is used for making dilutions of samples. It is
   50 mmollL sodium carbonate, pH 9.6.
                                        191
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
2. Wash buffer: this buffer is used for all wash steps. It is made of the carbonate
   buffer with ISO mmoVL NaCI and 0.1% polyoxyethylene-sorbitan mono-
   laurate (Tween 20).
3. Blocking buffer: this buffer is used to block any available binding sites within
   each well that have not bound capture antibody. It consists of carbonate
   buffer with 10 mglml bovine serum albumin.
4. Dilution buffer: this buffer is used to dilute standard and test samples. It
   consists of SO mmoVL sodium phosphate, pH 7.S, ISO mmoVL NaCl, 0.1 %
   Tween 20, and 1 mglml bovine serum albumin.
S. Substrate reactants: the peroxidase reaction is done by adding a solution of
   tetramethylbenzidine and hydrogen peroxide prepared according to the
   instructions in the tetramethylbenzidine (TMB) Peroxidase EIA Substrate
   Kit from BioRad laboratories.
6. Stop solution: a 1 moVL solution of H2S04 will stop the peroxidase reaction.
7. Capture antibody: this is a polyc1onal antibody against HCII. It can be found
   commercially (American Diagnostica) or produced using methods described
   previousli9 .
8. Conjugated antibody: this antibody is the capture antibody conjugated to
   horseradish peroxidase using N-succinimidyl 3-(2-piridylthio)proprionate
   (SPDP), as described by Carlsson et al. 3o, or the periodate method of Nakane
   et al. 3l •
9. Assay plates: these plates must be of high quality to ensure homogeneity,
   coating performance and uniform background absorbance. Two possible
   sources are immunoassay plates from Titertek or polystyrene microtitre
   plates type I from Nunc.
Method
1. Coating: coating of microtitre plates is done with 200 J.ll/well of 10 J.lglL
   capture antibody to HCII diluted in carbonate buffer. Seal plates with
   parafilm and incubate for 12 h at room temperature. The coating solution
   is carefully removed from the wells and the plate is washed S x 3 min with
   200 J.lVwell Wash Buffer. The plates are then blocked with 200 J.lVwell of
   blocking buffer for 2 h at room temperature, after which the plates are
   washed S x 3 min with 200 J.LVwell wash buffer.
2. Test samples: using the dilution buffer test samples were diluted 1:2000 and
   1:4000; 200 J.Ll of diluted samples are added to the wells and incubated for
   2 h at room temperature in a moist chamber. Wells are emptied and washed
   5 x 3 min with 200 J.lVwell wash buffer. The conjugated antibody is then
   added 200 J.LVwell diluted to a concentration of S J.lglml with dilution buffer
   and incubated for 2 h at room temperature. Finally the plate is washed S x
   3 min with 200 J.LVwell wash buffer. The peroxidase reaction is performed
   using 200 J.LVwell of the tetramethylbenzidine and hydrogen peroxide solution
   made according to the instructions provided with the TMB Peroxidase EIA
   Substrate Kit. This solution incubates for 2 min at room temperature and
   is quenched using 100 J.LVwell of stop solution. The chromogenic activity in
   each well is measured at 4S0 nm on a microplate reader.
3. Calibration: calibrate this assay with pooled normal plasma samples diluted
   in dilution buffer. The standard curve is created with 1: 1000, 1:2000, 1:4000,
                                       192
                            HEPARIN COFACTOR"
Laurell immunoelectrodiffusion
Principle
An agarose gel is made of uniform thickness containing a polyclonal antibody
to HCII. Exact volumes of pooled normal plasma dilutions and test plasma
are applied to the gel and electrophoresed. As the antibodies and samples
migrate through the gel, a rocket-shaped precipitation zone of antigen-antibody
complexes forms. Unbound HCII within the tip of the rocket redissolves the
complexes at the leading front as they migrate, until all of the free antigen is
completely complexed. This assay is based on the methods described by Laure1l32
and Tran and Duckert for HC1I 33 .
Materials
1. Barbital buffer: this buffer is used to make the gel used in this procedure. It
   consists of sodium barbital (S,S'-diethyl-barbituric acid) buffer with an ionic
   strength of 0.07 and 2 mmoVL calcium lactate at pH 8.6.
2. 1% agarose gel: this is used for electrophoresis of the standards and the test
   samples; 1% (w/v) agarose is dissolved in the barbital buffer.
3. Hell antibody: this antibody is added to the agarose gel when the temperature
   is approximately 40-45 °C. HCII polyc1onal antibodies are available
   commercially, or can be produced using the method described in ref. 28.
   The lowest amount of antibody needed to give well-defined rockets should
   be used. This amount will usually be from 0.5% to 2% (v/v) of antibody in
   the gel. The amount will, however, differ depending on the specific antibody
   preparation used.
Method
1. Electrophoretic set-up: this method was first described by Laurell in 1966.
   It entails forming a gel mould with two rectangular glass plates separated
   by I.S mm spacer on each side and along the bottom. Mter securing the
   apparatus the mould is filled with the 1% agarose with HCII antibody liquid
   mixture (approximately 40 0q. Mter at least 30 min the top plate is removed
   and small uniform holes are cut in the gel on a parallel line approximately
   2 em from one of the long edges.
2. Electrophoresis of samples and standards: plasma samples are diluted 1:2
   and standard samples of pooled normal plasma are made undiluted and
   diluted 3:4, 2:4 and 1:4 in isotonic NaCl. The gel is arranged so the application
   holes are at the cathodic end of the gel. Apply each sample of standard
   dilution to a separate well in exactly the same volume (5 JJ.l). The gel is run
                                        193
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   at 10 V/cm for 2-10 h depending on the amount of antigen versus antibody
   applied to the gel. When the gel has completed running it can be stained
   with Coomassie blue. The ideal height of the rockets is 2-5 cm.
3. Measurement and evaluation: a standard curve is drawn of the various
   dilutions of pooled normal plasma based on their dilution factor versus the
   height in millimetres of the associated rocket (measured from the centre of
   the application hole to the tip of the rocket). The height of the test sample
   plasma rockets can be used to deduce a percentage of HClI compared to
   the pooled normal plasma standard curve.
Materials
1. Dilution buffer: this buffer is used for making dilutions of plasma samples;
   it comprises 20 mmollL Tris-HCI, pH 7.4, 150 mmollL NaCl, 0.1%
   polyethylene glycol (pEG).
2. Reaction buffer: this buffer is used to ensure that the dermatan sulphate-
   accelerated inhibitory activity of HClI is what is being measured. It consists
   of dilution buffer with 4 ~g/ml polybrene, 133 ~g/ml dermatan sulphate
   (nitrous acid-treated), 8 mmollL EDTA, and 1 mg/ml bovine serum albumin.
3. Thrombin solution: human a-thrombin is the enzyme used to measure HClI
   inhibitory activity. The solution to be used in the assay is made up to
   approximately 10 NIH Ulml in dilution buffer.
4. Chromogenic substrate solution: the chromogenic substrate solution is used
   to measure the amount of thrombin amidolytic activity in a given sample.
   The chromogenic substrate used in this assay is tosyl-Gly-Pro-Arg-p-
   nitroanilide (Chromozym TH-Boehringer Mannheim). It is made up to 750
   ~mollL in dH 20.
5. Stop solution: glacial acetic acid is used to stop the hydrolysis of the substrate.
Method
1. Inhibition reaction: all reactions are carried out in 1.7 ml polypropylene
   bullet tubes. Test plasma samples (1-4 ~l) are diluted to a 40 ~ volume with
                                         194
                            HEPARIN COFACTOR II
Immunoblotting of Hell
Principle
Samples are run using sodium dodecyl sulphate polyacrylamide gel electro-
phoresis (SDS-PAGE) methodology and qualitatively compared to a protein
standard or a plasma-purified standard HCII sample to confirm the correct
molecular weight. The proteins are electrophoretically transferred to a nitrocel-
lulose membrane (or polyvinylidine difluoride membrane). The membrane is
then probed with an anti-HCII polyclonal antibody. If the antibody is conjugated
to horseradish peroxidase the activity can be developed and used to localize
the protein. This assay is based on the methods described by Laemmle4 and
Toulon et a1. 35 • Note: This method can also be used to qualitate complexes
formed by HCIl with thrombin and glycosaminoglycans.
Materials:
1. TBS (Tris buffered saline) buffer: this is the base buffer for all steps in
   probing the transfer membrane. It is made of 20 mmoVL Tris-HCI, pH 7.5,
   and 50 mmoVL NaCl.
2. Blocking buffer: this buffer will block all binding sites on the transfer
   membrane where protein was not transferred. It is made up with TBS buffer
   containing 5% (w/v) instant non-fat dried milk.
3. Wash buffer: this buffer is used to wash the blot after each step. It is made
   of TBS buffer with 0.05% (v/v) Tween-20.
4. Primary (anti-Hell) antibody solution: a polyclonal antibody against HCII
                                      195
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
Method
1. Gel electrophoresis: the plasma samples and standards can be run either
   reduced or unreduced. Using SDS-PAGE methodolo@;; run protein out on
   a 7.5% polyacrylamide gel containing 0.1% (v/v) SDS 4.
2. Electrophoretic transfer: the protein from the SDS gel is electrophoretically
   transferred to the transfer membrane using the methods described by Towbin
   et al. 36 •
3. Immunoblotting procedure: the transfer membrane is blocked using blocking
   buffer for 2 h at room temperature. Next the blot is washed 3 x 10 min with
   wash buffer. The blot is incubated at room temperature in primary antibody
   solution for 1.5 h. The wash step is repeated. The blot is incubated in the
   secondary antibody solution for 1.5 h. The wash step is again repeated.
   Finally, the peroxidase activity is detected by applying the development
   solution and observing colour development.
Hell can be obtained commercially or purified from plasma using the method
of Griffith et al. 37 or Tollefsen et al. 3. Hell can also be purified by immuno-
isolation as described by Toulon et al.2O. Purified Hell can be used as a standard
in all quantitative assays to calculate exact molar amounts of protein present
in patient samples.
   Additional assays to evaluate HCII activity have been described using purified
Hell. These include assays for Hell inhibition of chymotrypsin, interaction
of Hell with neutrophil elastase and cathepsin G, leukocyte chemoattractant
activity, and inhibition of thrombin in the absence or presence of glyco-
saminoglycan (heparin or dermatan sulphate)1l,38-40.
                                       196
                                   HEPARIN COFACTOR II
References
 1. Tollefsen OM, Pestka CA. Heparin cofactor II activity in patients with disseminated
    intravascular coagulation and hepatic failure. Blood 1985; 66: 769-74.
 2. Briginshaw GF, Shanberge IN. Identification of two distinct heparin cofactors in human
    plasma: II. Inhibition of thrombin and activated factor X. Thromb Res 1974; 4: 463-77.
 3. Tollefsen OM, Majerus Ow, Blank MK. Heparin cofactor II: purification and properties of
    a heparin-dependent inhibitor of thrombin in human plasma. I Bioi Chem 1982; 257: 2162-9.
 4. Wunderwald P, Schrenk WI, Port H. Antithrombin BM from human plasma: an antithrombin
    binding moderately to heparin. Thromb Res 1982; 25: 177-91.
 5. Griffith MI, Carraway T, White GC, Oombrose FA. Heparin cofactor activities in a family
    with hereditary antithrombin III deficiency: evidence for a second heparin cofactor in human
    plasma. Blood 1983; 61: Ill-18.
 6. Ragg H. A new member of the plasma protease inhibitor gene family. Nucl Acids Res 1986;
      14: 1073-87.
 7. Church FC, Shirk RA, Philips IE. Heparin Cofactor II. In: High KA, Roberts HR, eds.
    Molecular basis of thrombosis and hemostasis. New York: MarcelOekker, 1995; 379-92.
 8. Tollefsen OM. Insight into the mechanism of action of heparin cofactor II. Thromb Haemostas
      1995;74: 1209-14.
 9. Schecter I, Berger A. On the size of the active site in proteases I. Papain. Biochem Biophys
    Res Commun 1967; 27: 157-62.
10. Griffith MI, Noyes CM, Tyndall lA, Church Fe. Structural evidence for leucine at the reactive
    site of heparin cofactor II. Biochemistry 1985; 24: 6777-82.
II. Rogers SI, Pratt CW, Whinna HC, Church Fe. Role of thrombin exosites in inhibition by
    heparin cofactor II. I Bioi Chern 1992; 267: 3613-17.
12. Whinna HC, Blinder MA, Szewczyk M, Tollefsen OM, Church FC. Role of lysine 173 in
    heparin binding to heparin cofactor II. I Bioi Chern 1991; 266: 8129-35.
13. Whinna HC, Church FC. Characterization of a synthetic peptide from the glycosarninoglycan
    binding site of heparin cofactor II. Letts Pept Sci 1994; I: 3-8.
14. Blinder MA, Tollefsen OM. Site-directed mutagenesis of arginine 103 and lysine 185 in the
    proposed glycosaminoglycan-binding site of heparin cofactor II. I Bioi Chern 1990; 265:
      286-91.
IS. Blinder MA, Andersson TR, Abildgaard U, Tollefsen OM. Heparin cofactor II Oslo. Mutation
    of Arg-189 to His decreases the alfInity for dermatan sulfate. I Bioi Chem 1989; 264: 5128--33.
16. Blinder MA, Marasa IC, Reynolds Ch, Oeaven LL, Tollefsen OM. Heparin cofactor II:
    cONA sequence, chromosome localization, restriction length polymorphism and expression
    in Escherichia coli. Biochemistry 1988; 27: 752-9.
17. van Oeerlin VMO, Tollefsen OM. The N-terminal acidic domain of heparin cofactor II
    mediates the inhibition of a-thrombin in the presence of glycosarninoglycans. I Bioi Chern
    1991;266:20223-31.
18. Sheehan JP, Tollefsen OM, Sadler IE. Heparin cofactor II is regulated allosterically and not
      primarily by template effects. Studies with mutant thrombins and glycosarninoglycans. I BioI
      Chern 1994; 269: 32747-51.
19.   Kondo S, Tokanaga F, Kario K, Matsuo T, Koide T. Molecular and cellular basis for type I
      heparin cofactor II deficiency (heparin cofactor II Awaji). Blood 1996; 87: 1006-12.
20.   Toulon P, Chadeuf G, Bouillot IL et al. Involvement of heparin cofactor II in chymotrypsin
      neutralization and in the pancreatic proteinase-antiproteinase interaction during acute
      pancreatitis in man. Eur I Clin Invest. 1991; 21: 303-9.
21.   Toulon P, Larnine M, Ledjev I et al. Heparin cofactor II deficiency in patients infected with
      the human immunodeficiency virus. Thromb Haemostas 1993; 70: 730-5.
22.   Ouboscq C, Quintana I, Barros I, Kordich L. Heparin cofactor II in diabetic patients. Blood
      Coag Fibronol. 1994; 5: 201-4.
23.   Andersson TR, Berner NS, Larsen ML, Odegaard OR, Abildgaard U. Plasma heparin cofactor
      II, protein C and antithrombin in elective surgery. Acta Chir Scand 1987; 153: 291-6.
24.   Toulon P, Iacquot C, Capron L, Frydman MO, Vignon 0, Aich M. Antithrombin III and
      heparin cofactor II in patients with chronic renal failure undergoing regular hemodialysis.
      Thromb Haemostas 1987; 57: 263-8.
                                                197
           LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
25. Toulon P, Gandrille S, Remy P, Chadeuf G, Jouvin MH, Aiach M. Significance of high levels
    of heparin cofactor II in the plasma and urine of adult patients with nephrotic syndrome.
    Nephron 1992; 60: 176-80.
26. Mackie IJ, Segal H, Burren T et af. Heparin cofactor II levels are increased by the use of
    combined oral contraceptives. Blood Coag Fibrinol1990; 1: 647-51.
27. Andersson TR, Bangstad H, Larsen ML. Heparin cofactor II, antithrombin and protein C
    in plasma from term and preterm infants. Acta Paediatr Scand 1988; 77: 485-8.
28. Toulon P, Costa JM, Amiral 1. An enzyme-linked immunosorbent assay for heparin cofactor
    II (HCII). Application to the measurement of HCII in clinical materials. Clin Chim Acta
    1992; 205: 65-73.
29. Harlow E, Lane D, eds. Antibodies: A laboratory manual. Cold Spring Harbor: Cold Spring
    Harbor Laboratory, 1988; 726.
30. Carlsson J, Orevin H, Axen R. Protein thiolation and reversible protein-protein conjugation.
    N- succinimidyI3-(2-pyridyldithio) proprionate, a new hetero-bifunctional reagent. Biochem
    J 1978; 173: 723-37.
31. Nakane PK, Kawaoi A. Peroxidase-labeled antibody: a new method of conjugation. J
    Histochem Cytochem 1974; 22: 1084-91.
32. Laurell C-B. Quantitative estimation of proteins by electrophoresis in agarose gel containing
    antibodies. Anal Biochem 1966; 15: 45-52.
33. Tran TH, Duckert F. Heparin cofactor II determination-levels in normals and patients with
    hereditary antithrombin III deficiency and disseminated intravascular coagulation. Thromb
    Haemostas 1984; 52: 112-16.
34. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage
    T4. Nature 1970; 227: 680--5.
35. Toulon P, Chadeuf G, Bouillot JL et af. Involvement of heparin cofactor II in chymotrypsin
    neutralization and in the pancreatic proteinase-antiproteinase interaction during acute pacreatitis
    in man. Eur J Clin Invest 1991; 21: 303-9.
36. Towbin HJ, Staehelin T, Gordon 1. Electrophoretic transfer of proteins from polyacrylamide
    gels to nitrocellulose sheets. Procedure and some applications. Proc Nat! Acad Sci 1979; 76:
    4350--4.
37. Griffith MJ, Noyes CM, Church Fe. Reactive site peptide structural similarity between heparin
    cofactor II and antithrombin III. J Bioi Chem 1985; 260: 2218-25.
38. Church FC, Noyes CM, Griffith MJ. Inhibition of chymotrypsin by heparin cofactor II. Proc
    Nat! Acad Sci USA 1985; 82: 6431-4.
39. Pratt CW, Tobin RL, Church Fe. Interaction of heparin cofactor II with neutrophil elastase
    and cathepsin G. J Bioi Chem 1990; 265: 6092-7.
40. Hoffman M, Pratt CW, Corbin LW, Church FC. Characteristics of the chemotactic activity
    of heparin cofactor II proteolysis products. J Leuk Bioi 1990; 48: 156-62.
                                                 198
21
Fibrinopeptide A (FPA)
A. HAEBERLI
INTRODUCTION
PATHOPHYSIOLOGY
METHODS OF ASSAY
Since normal concentrations are around or below 2 ng/ml « 1.3 nmoVL) and
pathological values very often in the range of 3-20 ng/ml (2-13 nmoVL), the
only sufficiently sensitive methods are radioimmunoassay (RIA) or enzyme-
linked radioimmunoassay (ELISA). Nossel and co-workers were the first to
develop a RIA for the determination of FPA2,3. The antibodies used by Nossel
became available to a broader community and therefore the number of publica-
tions studying thrombotic and prethrombotic activities in different diseases by
means of FPA measurements increased enormously. Since 1971 several modifica-
tions of the original RIA of Nosse1 have been published (e.g. refs 60 and 61).
Some of these modifications have been introduced into the RIA procedure
recommended below.
   Two commercial RIA kits have been available for many years. The RIA-mat
FPA, originally manufactured by Mallinckrodt, then prepared and sold by
Byk-Sangtec (Dietzenbach, Germany) was a ready to use kit with all reagents
including the 125 labelled Tyr-FPA; it has since been discontinued.
   The other RIA is prepared by Imco. Imco supplies only three vials containing
anti-FPA antiserum, FPA standard and Tyr-FPA for iodination with 1251. The
iodination of Tyr-FPA, all dilutions of the standards and the antiserum, as
well as all the solutions necessary for the performance of the RIA, have to be
prepared. It is at present not clear whether Imco will continue to supply the
reagents.
   In 1980 a solid-phase ELISA for the measurement of FPA was published4,5.
This ELISA test is commercially available as a complete kit, Asserachrom FPA,
supplied by Diagnostica Stago. It is this assay which is now most frequently
used for the determination of FPA. Unfortunately it has recently been dis-
continued.
   The FPA-ELISA distributed by Diagnostica Stago was rather unique in its
concept, and might not be easy to set up with reagents still available; it will
therefore be discussed only very briefly.
   Since the Mallinckrodt kit RIA kit is no longer commercially available, the
methodological section below will preferentially deal with the RIA method as
described by Imco. The reagents supplied by Imco can easily be replaced by
reagents distributed by other companies. A list of companies supplying
anti-FPA-antiserum, fibrinopeptide A as standard and Tyr-fibrinopeptide A
for iodination is given at the end of this chapter.
PRECAUTIONS
References
 I. Marder VJ, Francis cw, Doolittle RF. Fibrinogen, structure and physiology. In: Colman RW,
    Hirsh J, Marder VJ, Salzman EW, eds. Haemostasis and thrombosis. Philadelphia: Lippincott,
    1982; 145-63.
 2. Nossel HL, Younger LR, Wilner GD, Procupez D, Canfield RE, Butler VP. Radioimmunoassay
    of human fibrinopeptide A. Proc Natl Acad Sci USA 1971; 68: 2350-3.
 3. Nossel HL, Yudelman I, Canfield RE, Butler VP, Spanondis K, Wilner GD, Qureshi GD.
    Measurement of fibrinopeptide A in human blood. J Clin Invest 1974; 54: 43-53.
 4. Soria J, Soria C, Ryckewaert JJ. A solid phase immuno enzymologica1 assay for the measurement
    of human fibrinopeptide. A. Thromb Res 1980; 20: 425-35.
 5. Amiral J, Walenga JM, Fareed 1. Development and performance characteristics of a competitive
    enzyme immunoassay for fibrinopeptide A. Sem Thromb Haemostas 1984; 10; 228--42.
 6. Bauer KA, Rosenberg RD. The pathophysiology of the prethrombotic state in humans:
    Insights gained from studies using markers of hemostatic system activation. Blood 1987; 70:
    343-50.
                                              204
                                     FIBRINOPEPTIDE A
 7. Hirsh 1. Blood tests for the diagnosis of venous and arterial thrombosis. Blood 1981; 57: 1-8.
 8. Joist JH. Fibrinopeptide A in the diagnosis and treatment of deep venous thrombosis and
    pulmonary embolism. Clin Lab Med 1984; 4: 363-80.
 9. Douglas IT, Blamey SL, Lowe GDO, Carter DC, Forbes CD, Plasma beta-thromboglobu1in,
    fibrinopeptide A and Bfl15-42 antigen in relation to postoperative DVT, malignancy and
    stanozolol treatment. Thromb Haemostas 1985; 53: 235-8.
10. Leeksma OC, Meijer-Huizinga F, Stoepman-van-Dalen EA, van Ginkel CJW, van Aken WG,
    van Mourik JA. Fibrinopeptide A and the phosphate content of fibrinogen in venous
    thromboembolism and disseminated intravascular coagulation. Blood 1986; 67: 1460--7.
11. Nossel HL. Radioimmunoassay of fibrinopeptides in relation to intravascular coagulation
    and thrombosis. N Engl J Med 1976; 295: 428-32.
12. Yudelman 1M, Nossel HL, Kaplan KL, Hirsh J. Plasma fibrinopeptide A levels in symptomatic
    venous thromboembolism. Blood 1978; 51: 1189-95.
13. Sobel M, Sternbergh C, Marques D, Grimsdale AS. A comparative study of heparin responses
    in arterial and venous thromboembolism using molecular markers for thrombosis. Circulation
    1993; 88: 426--31.
14. Cronlund M, Hardin J, Burton J, Lee L, Haber E, Bloch KJ, Fibrinopeptide A in plasma of
    normal subjects and patients with disseminated intravascular coagulation and systemic lupus
    erythematosus. J Clin Invest 1976; 58: 142-51.
15. Mombelli G, Monotti R, Haeberli A, Straub pw. Relationship between fibrinopeptide A and
    fibrinogen/fibrin fragment E in thromboembolism, DIC and various non-thromboembolic
    diseases. Thromb Haemostas 1987; 58: 758-63.
16. Eisenberg PR, Lucore C, Kaufman L, Sobel BE, Jaffe AS, Rich S. Fibrinopeptide A levels
    indicative of pulmonary vascular thrombosis in patients with primary pulmonary hypertension.
    Circulation 1990; 82: 841-7.
17. Tulchinsky M, Zeller JA, Reba RC. Urinary fibrinopeptide A in evaluation of patients with
    suspected acute pulmonary embolism. A prospective pilot study. Chest 1991; 100: 394-8.
18. Eisenberg PR, Sherman LA, Schectman K, Perez J, Sobel BE, Jaffe AS. Fibrinopeptide A: a
    marker of acute coronary thrombosis. Circulation 1985; 71: 912-18.
19. Ga11ino A, Haeberli A, Baur HR, Straub pw. Fibrin formation and platelet aggregation in
    patients with severe coronary artery disease: relationship with the degree of myocardial
    ischemia. Circulation 1985; 72: 27-30.
20. Gallino A, Haeberli A, Hess T, Mombelli G, Straub pw. Fibrin formation and platelet
    aggregation in patients with acute myocardial iufarction: effects of intravenous and subcutaneous
    low-dose heparin. Am Heart J 1986; 112; 285-90.
21. Gensini GF, Rostagno C, Abbate R, Favilla S, Mannucci PM, Neri Semeri GG. Increased
    protein C and fibrinopeptide A concentration in patients with angina. Thromb Res 1988; 50:
    517-25.
22. Irie T, Imaaizumi T, Matuguchi T, Koyanagi S, Kanaide H, Takeshita A, Nakamura M.
    Increased fibrinopeptide A during anginal attacks in patients with variant angina. J Am Coli
    Cardio11989; 14: 589-94.
23. Mombelli G, 1m Hof V, Haeberli A, Straub pw. Effect of heparin on plasma fibrinopeptide
    A (fpA) in acute myocardial iufarction. Circulation 1984; 69: 684--9.
24. Neri Semeri G, Gensini GF, Camovali M, Prisco D, Rogasi PG, Casolo GC, Fazi A, Abbate
    R. Association between time of increased fibrinopeptide A levels in plasma and episodes of
    spontaneous angina: a controlled prospective study. Am Heart J 1987; 113: 672-8.
25. Nichols AB, Owen J, Kaplan KL, Sciacca RR, Cannon PJ, Nossel HL. Fibrinopeptide A,
    platelet factor 4, and fI-thromboglobulin levels in coronary heart disease. Blood 1982; 60:
    650-4.
26. Theroux P, Latour JG, Leger-Gauthier C, De Lara J, Fibrinopeptide A and platelet factor
    levels in unstable angina pectoris. Circulation 1987; 75: 156--62.
27. Van Hulsteijn H, Kolff J, Briet E, van de Laarse A, Bertina R. Fibrinopeptide A and
    beta-thromboglobulin in patients with angina pectoris and acute myocardial infarction. Am
    Heart J 1984; 107: 39-45.
28. Landi G, Barbarotto R, Morabito A, D'Angelo A, Mannucci PM. Prognostic significance of
    fibrinopeptide A in survivors of cerebral iufarction. Stroke 1990; 21: 424--7.
29. Feinberg WM, Erickson LP, Bruck D, Kittelson 1. Hemostatic markers in acute ischemic
    stroke: association with stroke type, severity, and outcome. Stroke 1996; 27: 1296--300.
                                                205
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
30. Conway EM, Bauer KA, Barzegar S, Rosenberg RD. Suppression of hemostatic system
    activation by oral anticoagulants in the blood of patients with thrombotic diseases. J Clin
    Invest 1987; 80: 1535--44.
31. Yudelman I, Greenberg J. Factors affecting fibrinopeptide A levels in patients with venous
    thromboembolism during anticoagulant therapy. Blood 1982; 59: 787-92.
32. Merlini PA, Ardissino D, Bauer KA et al. Persistent thrombin generation during heparin
    therapy in patients with acute coronary syndromes. Arterioscler Thromb Vasc Bioi 1997; 17:
    1325-30.
33. Melandri G, Semprini F, Cervi V et al. Benefit of adding low molecular weight heparin to the
    conventional treatment of stable angina pectoris. A double-blind, randomized, placebo-
    controlled trial. Circulation 1993; 88: 2517-23.
34. Rao AK, Sun L, Chesebro JH et al. Distinct effects of recombinant desulfatohirudin (Revasc)
    and heparin on plasma levels of fibrinopeptide A and prothrombin fragment FI.2 in unstable
    angina: a multicenter trial. Circulation 1996; 94: 2389-95.
35. Solymoss S, Bovill EG. Markers of in vivo activation of coagulation: interrelationships change
    with intensity of oral anticoagulation. Am J Clin Patho11996; 105: 293-7.
36. Merlini PA, Ardissino D, Bauer KA et al. Activation of the hemostatic mechanism during
    thrombolysis in patients with unstable angina pectoris. Blood 1995; 86: 3327-32.
37. Merlini PA, Bauer KA. Thrombin generation and activity during thrombolysis and concomitant
    heparin therapy in patients with acute myocardial infarction. J Am CoIl Cardiol1995; 25:
    203-9.
38. Rapold HJ, de Bono D, Arnold AER et al. Plasma fibrinopeptide A levels in patients with
    acute myocardial infarction treated with alteplase. Correlation with concomitant heparin,
    coronary artery patency, and recurrent ischemia. Circulation 1992; 85: 928-34.
39. Rapold HJ, Grimaudo V, Declerck PJ, Kruithof EKO, Bachmann F. Plasma levels of
    plasminogen activator inhibitor type I, ~-thromboglobulin, and fibrinopeptide A before,
    during, and after treatment of acute myocardial infarction with alteplase. Blood 1991; 78:
    1490--5.
40. Scharfstein JS, Abendschein DR, Eisenberg PR et al. Usefulness of fibrinogenolytic and
    procoagulant markers during thrombolytic therapy in predicting clinical outcomes in acute
    myocardial infarction. Am J Cardio11996; 78: 503-10.
41. Auger MJ, Galloway MJ, Leinster SJ, McVerry BA, Mackie MJ. Elevated fibrinopeptide A
    levels in patients with clinically localised breast carcinoma. Haemostasis 1987; 17: 336-9.
42. Edwards RL, Klaus M, Matthews E, McCullen C, Bona RD, Rickles FR. Heparin abolishes the
    chemotherapy-induced increase in plasma fibrinopeptide A levels. Am J Med 1990; 89: 25-8.
43. Gugliotta L, Vigano S, D'Angelo A, Guarini A, Tura S, Mannucci PM. High fibrinopeptide
    A (FPA) levels in acute non-lymphocytic leukemia are reduced by heparin administration.
    Thromb Haemostas 1984; 52: 301-4.
44. Mombelli G, Roux A, Haeberli A, Straub pw. Comparison of 12sI-fibrinogen kinetics and
    fibrinopeptide A in patients with disseminated neoplasias. Blood 1982; 60: 381-8.
45. Myers TJ, Rickels FR, Barb C, Cronlund M. Fibrinopeptide A in acute leukemia: relationship
    of activation of blood coagulation to disease activity. Blood 1981; 57: 518-25.
46. Peuscher FW, Cleton FJ, Armstrong L, Stoepman-van-Dalen EA, van Mourik JA, van Aken
    WG. Significance of plasma fibrinopeptide A (fpA) in patients with malignancy. J Lab Clin
    Med 1980; 96: 5-14.
47. Douglas IT, Shah M, Lowe GOO, Belah JJF, Forbes CD, Prentice CRM. Plasma fibrinopeptide
    A and ~-thromboglobulin in preeclampsia and pregnancy hypertension. Thromb Haemostas
    1982; 47: 54-8.
48. Jones RL. Fibrinopeptide A in diabetes mellitus. Relation to levels of blood glucose, fibrinogen
    disappearance, and hemodynamic changes. Diabetes 1985; 34: 836-43.
49. Rosove MH, Frank HJL, Harwig SSL. Plasma ~-thromboglobulin, platelet factor 4,
    fibrinopeptide A, and other hemostatic functions during improved, short-term glycemic control
    in diabetes mellitus. Diabetes Care 1984; 7: 174-9.
50. Ceriello A, Giacomello R, Stel G et al. Hyperglycemia-induced thrombin formation in diabetes:
    the possible role of oxidative stress. Diabetes 1995; 44: 924-8.
51. Coccherri S, Mannucci PM, Palaretti G, Gervasoui W, Poggi M, Vigano S. Significance of
    plasma fibrinopeptide A and high molecular weight fibrinogen in patients with liver cirrhosis.
    Br J Haematol1982; 52: 503-9.
                                               206
                                    FIBRINOPEPTIDE A
52. Mombelli G, Fiori G, Monotti R, Haeberli A, Straub PW: Fibrinopeptide A in liver cirrhosis.
    Evidence against a major contribution of disseminated intravascular coagulation to
    coagulopathy of chronic liver disease. J Lab Clin Med 1993; 121: 83-90.
53. Violi F, Ferro D, Basili S et al. Association between low-grade disseminated intravascular
    coagulation and endotoxemia in patients with liver cirrhosis. Gastroenterology 1995; 109:
    531-9.
54. Alkjaersig N, Fletcher AP, de Ziegler D, Steingold KA, Meldrum DR, Judd HL. Blood
    coagulation in postmenopausal women given estrogen treatment: comparison of transdermal
    and oral administration. J Lab C1in Med 1988; Ill: 224-8.
55. Inauen W, Baumgartner HR, Haeberli A, Straub PW: Excessive deposition of fibrin, platelets
    and platelet thrombi on vascular subendothelium during contraceptive drug treatment. Thromb
    Haemostas 1987; 57: 306-9.
56. Melis GB, Fruzzetti F, Paoletti AM, Carmassi F, Fioretti P. Fibrinopeptide A plasma levels
    during low-estrogen oral contraceptive treatment. Contraception 1984; 30: 575-83.
57. Melis GB, Fruzzetti F, Ricci C, Carmassi F, Fioretti P. Oral contraceptives and venous
    thromboembolic disease: the effect of the oestrogen dose. Maturitas, Suppl. 1988; 1: 131-9.
58. Herren T, Bartsch P, Haeberli A, Straub PW: Increased thrombin-antithrombin III complexes
    after 1 h of physical exercise. J Appl Physiol1992; 73: 2499-504.
59. Blirtsch P, Welsch B, Albert M, Friedmann B, Levi M, Kruithof EKO. Balanced activation
    of coagulation and fibrinolysis after a 2-h triathlon. Med Sci Sports Exerc 1995; 27: 1465-70.
60. Hoffman V, Straub PW: A radioimmunoassay technique for the rapid measurement of human
    fibrinopeptide A. Thromb Res 1977; 11: 171-81.
61. Kockum C, Frebelius S. Rapid radioimmunoassay of human fibrinopeptide A - removal of
    cross-reacting fibrinogen with bentonite. Thromb Res 1980; 19: 589-98.
62. Alkjaersig N, Fletcher AP. Catabolism and excretion of fibrinopeptide A. Blood 1982; 60:
    148-56.
63. Gallino A, Haeberli A, Straub PW: Fibrinopeptide A excretion in urine in patients with
    atherosclerotic artery disease. Thromb Res 1985; 38: 237--44.
64. Leeksma OC, Meijer-Huizinga F, Stoepman-van Dalen EA, van Aken WG, van Mourik JA.
    Fibrinopeptide A in urine from patients with venous thromboembolism, disseminated
    intravascular coagulation and rheumatoid arthritis - evidence for dephosphorylation and
    carboxyterminal degradation of the peptide by the kidney. Thromb Haemostas 1985; 54:
    792-8.
65. Wilensky RL, Zeller JA, Wish M, Tulchinsky M. Urinary fibrinopeptide A levels in ischemic
    heart disease. J Am Coli Cardio11989; 14: 597-603.
66. Ardissino 0, Gamba MG, Mer1ini PA et al. Fibrinopeptide A excretion in urine: a marker of
    the cumulative thrombin activity in stable versus unstable angina pectoris. Am J Cardio11991;
    68: 58-63B.
67. Miller GJ, Bauer KA, Bazegar S et al. The effect of quality and timing of venepuncture on
    markers of blood coagulation in healthy middle-aged men. Thromb Haemostas 1995; 73:
    82-6.
                                              207
22
Thrombin-antithrombin (TAT)
complexes
J.HARENBERG
INTRODUCTION
METHODS OF ASSAY
Principle and assay characteristics
The assay principle was described first by Neumann et al. 3 and Brower et al. 4
for the detection of elastase-ul-antiproteinase complexes, and is based on the
properties of the appropriate antibodies to bind selectively the corresponding
moieties of the complex. The assay for measuring the TAT complex consists
                                     209
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
of antibodies directed against thrombin as well as antithrombin. Nanogram
quantities of TAT can be detected by these assays. Prothrombin, as well as
antithrombin, are not detected by the antibodies.
  The TAT complex has a molecular we~t of 88,000 and elutes as a single
peak from a sodium dodecyl sulphate gel .
Materials
At present only one enzyme immunoassay test kit is commercially available.
This ELISA test system has been developed by Behringwerke (now Dade
Behring)6, and contains:
1. plastic tubes coated with antibodies to human thrombin, stability 4 weeks
   at +4 DC;
2. antibodies to human antithrombin conjugated to horseradish peroxidase
   (preservative phenol max I giL), stability 4 weeks at +4 DC or 3 months at
   -20 DC;
3. buffered solution containing rabbit y-globulin and 5 mglml bovine serum
   albumin (preservative phenol max 1 giL), stability 4 weeks at +4 DC or 3
   months at -20 DC;
4. butTered solution for dilution of plasma samples 0.1 mol Tris-HCl pH 7.4
   with 5 mglml bovine serum albumin (preservative sodium azide max I giL),
   stability 4 weeks at +4 DC or 3 months at -20 DC;
5. PBS phosphate buffer saline solution containing 0.01 % Tween, stability as
   given on the vial;
6. citrate buffer solution containing 0.1 moVL citrate and 0.2 moVL sodium
   phosphate, pH 7.0 (preservative sodium-p-ethyl-mercury-mercapto-benzene
   sulphonate max 0.1 giL);
7. 200 mglmll,2-phenylenediamine monohydrochloride, stability only in the
   dark and 3 h at + 4 DC;
8. 0.5 N sulphuric acid, stability as given on the vial.
Plasma samples from human as well as various animal species (mouse, rat,
rabbit, guinea pig, dog, pig, sheep, baboon) can be analysed by this assay, but
notthe hen7 •
Procedure
Venous blood is withdrawn from individuals/animal species, in plastic syringes
or siliconized glass tubes containing 3.8% sodium citrate (9: 1 v/v). Samples are
centrifuged for 15 min at 2000g and at +4 DC; the obtained plasma is immediately
quick-frozen and stored at -30 DC.
   The antibody specific to thrombin is bound to polystyrene plastic tubes.
Disposable tubes contain a volume of 2.0 ml or 0.35 ml on microtitre plates.
Plasma is incubated in the test tube. The thrombin of the TAT complex reacts
with the thrombin antibody on the surface of the plastic tube. The antithrombin
                                      210
                  THROMBIN-ANTITHROMBIN COMPLEXES
EVALUATION OF RESULTS
Thrombosis and pulmonary embolism
Patients with deep vein thrombosis (DVT) and/or pulmonary embolism have
been studied to demonstrate the validity of the TAT assay. Elevated plasma
concentrations have been found in all patients with very recent clinical symptoms.
   Normal TAT complexes in plasma exclude pulmonary embolism in patients
with clinically suspected pulmonary embolism8,9. In conclusion, many clinical
studies demonstrate the validity of the various activation variables of blood
coagulation. The sensitivity of all activation parameters ranges from 30% to
60% and the specificity from 55% to 85%. However, there is no conclusion, at
present, as to which activation variables possess the highest positive or negative
predictive value.
Postoperative care
TAT complexes have been measured in postoperative care medicine with the
aim of defining a predictive index for the development of DVT. TAT levels
were significantly higher in patients developing DVT when compared to patients
without DVT. Statistical analysis revealed no satisfactory discriminative power
for the diagnosis of developing DVT at any of the studied cut-off values for
TATIO. TAT levels were measured in plasma of patients who underwent total
knee replacement in order to determine the possible value of this assay in
screening for thromboembolic complications. No difference was observed in
patients who developed DVT and those who did notll.
   Preoperative plasma levels of TAT complexes have been shown to correlate
with the development of venous thromboembolism after major hip or knee
surgery. However, even if much progress of the validity of TAT levels has been
made in perioperative medicine, further studies are needed concerning whether
rational recommendations about prophylaxis and screening for venous
thromboembolism can be made based on the results of preoperative TAT level.
Diabetes mellitus
In patients with diabetes mellitus an activation of the coagulation system has
been described several times in the past, and has been attributed to the
morphological changes of the microvascular system. The levels of TAT were
significantly elevated in diabetes mellitus as compared to a healthy control
group23. However, as yet this has not been supported by others24 .
   Thus, a variety of clinical conditions has been reported to be associated with
an increase in TAT concentration (Table 22.1).
                                        212
                      THROMBIN-ANTITHROMBIN COMPLEXES
Table 22.1 Elevated concentrations of TAT in plasma in relation to pathological conditions
Reference
Acquired diseases
 acute deep vein thrombosis                                       9
  acute pulmonary embolism                                        8
  oral anticoagulation and heparinization                         8-16
  postoperative medicine                                          10,11
  acute myocardial infarction and unstable angina                 12-16
  disseminated intravascular coagulation                          17
  liver disease                                                   18,19
 malignancy, leukaemia and chemotherapy                           20,21
  diabetes mellitus                                               23,24
Hereditary diseases                                               25,26
The following points may influence the validity of the results: incorrect blood
sampling with coagulation of blood in vitro and incorrect storage of reagents.
Blood samples from patients with haemolysis, hypercholesterolaemia, hyper-
bilirubinaemia and high levels of rheumatic factors may influence the results.
   The concentrations of TAT are influenced by technical problems during
withdrawal of blood. Blood sampling must be performed very carefully by a
clean venipuncture. The tourniquet of the forearm must be released after
puncture of a large vein of the forearm, and free-flowing blood must be taken
into a plastic syringe or into a siliconized glass tube containing 3.8% sodium
citrate. Despite careful blood sampling TAT levels in plasma have been described
to decrease within 3 months at -80 °C27 .
                                            213
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   Recently the determination of TAT levels has been extended to various
animal species, to new mechanical devices (artificial heart valves, coated
extracorporeal circuits), cell culture systems and to determine the efficacy of
new anticoagulants. In most investigations other variables for the measurement
of activation of blood coagulation are included. At present it remains open
which variable determines the activation of blood coagulation with the highest
sensitivity, specificity, negative or positive predictive value.
REFERENCE VALUE
Standard curves are linear in a range 0.5--60 nglml. The recovery rate of purified
TAT added to normal plasma ranged from 90% to 110%. The intra-assay
coefficient of variation was 2.0--6.1 %. The inter-assay coefficient of variation
was between 2.4% and 4.7% when 2-60 nglml TAT were added to pooled
plasma. The linearity of plasma sample concentrations were determined after
various dilutions of the sample with TAT-poor pool plasma. The lower detection
limit of the assay is 0.5 nglml.
The ELISA for the determination of TAT can be performed with larger
(ordinary test tubes, 5ml) and smaller volumes (microtitre plates) of the reagents.
The latter method has been developed in our laboratory and is now available
from Dade-Behring. The correlation between these two techniques is r 0.97.            =
No other methods are available at the moment from other manufacturers.
References
 I. Teitel 1M, Bauer KA, Lau HK, Rosenberg RD. Studies of the prothrombin activation pathway
    utilizing radioimmunoassays for the F2FI +2 fragment and thrombin-antithrombin complex.
    Blood 1982; 59: 1086-91.
 2. Rezaie AR. Tryptophan 60-D in the B-insertion loop of thrombin modulates the thrombin-
    antithrombin reaction. Biochemistry 1996; 35: 1918-24.
 3. Neumann S, Gunzer G, Hennrich N, Lang H. PMN-elastase assay: enzyme immunoassay for
    human polymorphonuclear elastase complexed with proteinase inhibitor. J Clin Chem Clin
    Biochern 1984; 22: 693-7.
 4. Brower MS, Harpel Oc. Alpha I-antitrypsin human leukocyte elastase complexes in blood:
    quantification by an enzyme linked differential antibody immunosorbent assay and comparison
    with alpha-2-plasmin inhibitor-plasmin complexes. Blood 1983; 61: 842-9.
 5. Lau HK, Rosenberg RD. The isolation and characterization of a specific antibody population
    directed against the thrombin-antithrombin complex. J Bioi Chern 1980; 255: 5885-93.
                                             214
                     THROMBIN-ANTITHROMBIN COMPLEXES
 6. Enzygnost TAT ELISA test kit. Enzyme immunoassay for the determination of thrombin!
    antithrombin III complex. Behringwerke AG, Marburg, Germany, 1997.
 7. Ravanat C, Freund M, Dol F, Cadroy Y, Roussi J, Incardona F, Maffrand JP, Boneu B, Drouet
    L, Legrand C. Cross-reactivity of human molecular markers for detection of prethrombotic
    states in various animal species. Blood Coagul Fibrinol1995; 6: 446-55.
 8. Carmassi F, Morale M, de Negri F, Puccetti R, Pistelli F, Mariani G, Pazzagli M, Palla A,
    Giuntini e. Thrombin-antithrombin III complexes as an additional diagnostic aid in pulmonary
    embolism. Haemostasis 1996; 26: 16-22.
 9. Grosset AB, Spiro TE, Beynon J, Rodgers GM. Enoxaparin, a low-molecular-weight heparin
    suppresses prothrombin activation more effectively than unfractionated heparin in patients
    treated for venous thromboembolism. Thromb Res 1997; 86: 349-54.
10. Hoek JA, Nurmohamed MT, ten Cate Jw, Biiller HR, Knipscheer He. Thrombin-antithrombin
    III complexes in the prediction of deep vein thrombosis following total hip replacement.
    Thromb Haemostas 1989; 62: 1050-2.
11. Ginsberg JS, Brill-Eduards P, Panju A, Patel A, McGinnis J, Smith F, Dale I, Johnston M,
    Ofosu F. Pre-operative plasma levels of thrombin-antithrombin III complexes correlate with
    the development of venous thrombosis after major hip or knee surgery. Thromb Haemostas
    1995; 74: 602-5.
12. Carvalho de Slus J, Azevedo J, Soria C, Barros F, Ribeiro C, Parreira F, Caen JP. Factor VII
    hyperactivity in acute myocardial thrombosis - a relation to the coagulation activation. Thromb
    Res 1988; 51: 165-73.
13. Pseja P, Lewandowski K, Turowiecka Z, Zozu1inska M, Tokarz A, Zawilska K. Fluctuation
    of thrombin-antithrombin III complex in patients with acute myocardial infarction - influence
    of low-dose heparin administration. Folia Haematol Leipz 1990; 117: 219-23.
14. van den Burg PJ, Hospers JE, van Vliet M, Mosterd WL, Bouma BN, Huisveld IA. Changes
    in haemostatic factors and activation products after exercise in healthy subjects with different
    ages. Thromb Haemostas 1995; 74: 1457-64.
15. Munkvad S, Gram J, Jespersen I A depression of active tissue plasminogen activator in
    plasma characterizes patients with unstable angina pectoris who develop myocardial infarction.
    Eur Heart J 1990; ll: 525-8.
16. Scharfstein JS, Abendschein DR, Eisenberg PR, George D, Cannon CP, Becker RC, Sobel
    B, Cupples LA, Braunwald E, Loscalzo I Usefulness of fibrinogenolytic and procoagulant
    markers during thrombolytic therapy in predicting clinical outcomes in acute myocardial
    infarction. TIMI 5 Investigators. Thrombolysis in Myocardial Infarction. Am J Cardio11996;
    78: 503-10.
17. Okajima K, Uchiba M, Murakami K, Okabe H, Takatsuki K. Determination of plasma
    soluble fibrin using a new ELISA method in patients with disseminated intravascular
    coagulation. Am J Hemato11996; 51: 186-91.
18. He S, Bremme K, Blombiick M. Acquired deficiency of antithrombin in association with a
    hypercoagulable state and impaired function of liver and/or kidney in preeclampsia. Blood
    Coagul Fibrinol1997; 8: 232-8.
19. Takahashi H, Tatewaki W, Wada K, Yoshikawa A, Shibata A. Thrombin and plasmin
    generation in patients with liver disease. Am J Hemato11989; 32: 30-5.
20. Nakashima J, Tachibana M, Ueno M, Baba S, Tazaki H. Tumor necrosis and coagulopathy
    in patients with prostate cancer. Cancer Res 1995; 55: 4881-5.
21. Avvisati G, ten Cate JW, Sturk A, Lamping R, Petti MG, Mandelle F. Acquired alpha-2-
    antiplasmin deficiency in acute promyelocytic leukaemia. Br J Haematol1988; 70; 43-8.
22. Gugliotta L, D'Angelo A, Mattioli Belmonte M, Vigano-D' Angelo S, Colombo G, Datani
    L, Gianni L, Lauria F, Tura S. Hypercoagulability during L-asparaginase treatment: the effect
    of antithrombin III supplementation in vivo. Br J Haemato11990; 74: 465-70.
23. Takashahi H, Tsuda A, Tatewaki W, Wada K, Niwano H, Shibata A. Activation of blood
    coagulation and fibrinolysis in diabetes mellitus. Evaluation by plasma levels of thrombin-
    antithrombin III complex and plasmin-alpha-2-plasmin inhibitor complex. Thromb Res 1989;
    55: 727-35.
24. van Wersch JWJ, Westerhuis LW, Venekamp WI Coagulation activation in diabetes mellitus.
    Haemostasis 1990; 20: 263-9.
                                               215
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
25. Simioni P, Scarano L, Gavasso S, Sardella C, Girolami B, Scudeller A, Girolami A. Prothrombin
    fragment 1-2 and thrombin-antithrombin complex levels in patients with inherited APC
    resistance due to factor V Leiden mutation. Br J Haemato11996; 92: 43~1.
26. Martinelli I, Bottasso B, Duca F, Faioni E, Mannucci PM. Heightened thrombin generation
    in individuals with resistance to activated protein C. Thromb Haemostas 1996; 75: 703-5.
27. Iversen LH, Thorlacius-Ussing O. Short-time stability of markers of coagulation and fibrinolysis
    in frozen plasma. Thromb Res 1996; 81: 253-{) I.
28. Pelzer H, Schwarz A, Heimburger N. Determination of human thrombin-antithrombin III
    complex in plasma with an enzyme linked immunosorbent assay. Thromb Haemostas 1988;
    59: 101-6.
29. Hoek JA, Sturk A, ten Cate JW, Lamping RJ, Berens F, Borm J1. Laboratory and clinical
    evaluation of an assay of thrombin-antithrombin III complexes in plasma. Clin Chern 1988;
    34: 2058-62.
                                               216
23
Prothrombin fragment F1 +2
A.HAEBERLI
INTRODUCTION
PATHOPHYSIOLOGY
METHODS OF ASSAY
The first test was developed in 1982 by the group of Teitel et a/. l and is a
radioimmunoassay using polyclonal antibodies against prothrombin fragment
Fl +2 and F2. This radioimmunoassay has since been used almost exclusively
by this group, and has not become available to a larger community. In 1991
Pelzer et al. presented an ELISA2which later became commercially available
(Enzygnost Fl +2, Dade-Behring). Since this test has been widely used in the
past 6 years it will be discussed in some detail below. Shortly afterwards two
other ELISA were developed for the measurement of prothrombin fragment
Fl +2. One was the Thrombonostika FI.2 (Organon-Teknika) using monoclonal
antibodies specific for prothrombin fragment FI +23. This test has been used
by several groups for the evaluation of activation of blood coagulation in
different diseases. This test is no longer commercially available. The last ELISA
kit was developed and sold by Baxter. It was in use for rather a short time and
is no longer available. Therefore, in the discussion of the method, only the test
still available, by Dade-Behring (Enzygnost FI +2) will be considered further.
As for other highly sensitive tests for low-grade activation of blood coaglllation,
e.g. fibrinopeptide A, blood sampling and processing is of importance l7, 29-34.
Although Dade-Behring recommends using citrate for blood collection, it is
advisable to add at least heparin to the anticoagulant solution for blood
                                       218
                      PROTHROMBIN FRAGMENT F1 +2
collection, since Tripodi et al. have shown a poor comparability between two
commercial prothrombin fragment Fl +2 ELISA methods35 . This was due at
least in part to the difference in the anticoagulants recommended by the
manufacturers, citrate versus heparin. Bauer and co-workers even recommend
using PPACK (phenyl-arginyl-prolyl-chloromethylketone), from Calbiochem,
a specific thrombin inhibitor, in addition to ensure an optimum quality of
blood samples29 • From our experience this recommendation is supported. In
our laboratory PPACK is used for all immunological determinations of
coagulation activation markers such as fibrinopeptide A (Chapter 21),
prothrombin fragment Fl +2, thrombin-antithrombin TAT complexes (Chapter
22) or plasmin-antiplasmin (PAP) complexes (Chapter 29).
   In 1992 Soerensen and co-workers published an interesting paper on the
determination of prothrombin fragment Fl +2 in urine 36 • Unfortunately there
was no follow-up. Therefore it is not ensured that the measurement of the
fragment in urine is a worthwhile alternative.
The sandwich assay developed by Pelzer et al. 2 makes use of two polyclonal
antibodies against prothrombin and prothrombin fragment Fl + 2. The catching
antibodies, which are immobilized on micro titre plates, were raised in rabbits
against a 14 amino acid negatively charged synthetic peptide corresponding to
the C-terminal part of prothrombin fragment F1 +2. They do not cross-react
with intact prothrombin. The other antibody (polyclonal, rabbit) reacts with
prothrombin and prothrombin fragment Fl +2. It has been coupled to peroxidase.
The enzyme activity is determined with the substrate o-phenylenediamine (OPD)
in the presence of hydrogen peroxide. The test kit Enzygnost Fl +2 micro, supplied
by Dade-Behring, contains all the necessary reagents to perform the assay. One
minor disadvantage of the assay is that the incubation steps must be performed
at 37°C in a water bath or on a special heating plate. It is recommended to perform
the assay strictly according to the instructions given by the manufacturer. In
general the assay performs well with good reproducibility if no modifications are
introduced. The standard curve ranges from 0.04 to 10 nmollL. The optical density
read at 492 nm (490--500 nm) is plotted against the standard values on a double-
logarithmic graph. The limit of detection is close to 0.02 nmollL, the intra-assay
variations are 6.9-10.4% and the inter-assay variations are 6.7-11 %
The normal values are variable to some extent depending on which assay was
used. For the Enzygnost F1 +2 Pelzer et al. 2 give a mean (± SD) of 0.67 ± 0.19
(range 0.32-1.2 nmoIlL). In our laboratory the normal values are 0.62 ± 0.21
nmollL (range 0.41--0.87 nmollL). Values higher than 1 nmollL mostly indicate
activation of blood coagulation. Values higher than 6 nmollL are rarely seen.
For other assays used formerly the following values were given: Thrombonostika
                                       219
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                ,
                E
                c
                                                          /
                                                              /
                                                                  /
                I
                c
                !                           V
                                                /
                    0.1                 V
                                  v
                                0.1                                      10
                                                nMorelL
Figure 23.1 Calibration curve, prothrombin fragment FI +2
F1.2 (Organon Teknika): 0.51 nmoVL (range 0.21-2.78 nmoVL)3 and radioim-
munoassay according to Teitel et al.: 1.97 ± 0.97 nmoVL 1 or 1.5 ± 0.7 nmoVL
(depending on another set of antibodies used)4. A minor increase with age
above 44 years, but no difference in sex, has been shown37 . 38.
   The inter-assay variations should always be monitored, preferentially with
in-house control plasma. A reasonable control plasma is prepared by adding
to a pooled citrated plasma (10 donors or more) 100 Ilg PPACKlml, stored
frozen in aliquots until use.
References
 I. Teitel lM, Bauer KA, Lau HI{, Rosenberg RD. Studies of tbe protbrombin activation patbway
    utilizing radioimmunoassays for the F2IFI +2 fragment and tbrombin-antithrombin complex.
    Blood 1982; 59: 1086-97.
 2. Pelzer H, Schwarz A, Stiiber W Determination of human prothrombin activation fragment
    1+2 in plasma witb an antibody against a synthetic peptide. Thromb Haemostas 1991; 65:
    15J-9.
 3. Hursting MI, Butman BT, Steiner JP et al. Monoclonal antibodies specific for prothrombin
    fragment 1.2 and tbeir use in a quantitative enzyme-linked immunosorbent assay. Clin Chem
    1993; 39:583-91.
 4. Bauer KA, Rosenberg RD. The patbophysiology of tbe pretbrombotic state in humans: insights
    gained from studies using markers of hemostatic system activation. Blood 1987; 70: 343-50.
 5. Bouman CSC, Ypma ST, Sybesma IPHB. Comparison of the efficacy of D-dimer, fibrin
    degradation products and prothrombin fragment I +2 in clinically suspected deep venous
    thrombosis. Thromb Res 1995; 77: 225-34.
 6. Estivals M, Pelzer H, Pichon SI, Boccalon H, Boneu B. Protbrombin fragment 1+2, tbrombin-
    antithrombin III complexes and D-dimers in acute deep vein tbrombosis. Effects of heparin
    treatment. Br I Haematol1991; 78: 421-4.
 7. KyrJe PA, Eichinger S, Pabinger I et al. Prothrombin fragment Fl +2 is not predictive for
    recurrent venous tbromboembolism. Thromb Haemostas 1997; 77: 829-33.
 8. Yurdakok M, Yigit S, Gurakan B, Dundar S, Kirazli S. Thrombin-antithrombin III and
    prothrombin fragment 1.2 levels in early respiratory distress syndrome. Am I Hemato11996;
    51: 247-8.
                                            220
                           PROTHROMBIN FRAGMENT F1 +2
 9. Kienast J, Thompson SG, Raskino C et aZ. Prothrombin activation fragment 1 + 2 and
    thrombin-antithrombin III complexes in patients with angina pectoris: relation to the presence
    and severity of coronary atherosclerosis. Thromb Haemostas 1993; 70: 550-3.
10. Rao AK, Sun L, Chesebro JH et aZ. Distinct effects of recombinant desulfatohirudin (Revasc)
    and heparin on plasma levels of fibrinopeptide A and prothrombin fragment FI.2 in unstable
    angina: a multicenter trial. Circulation 1996; 94: 2389-95.
II. Biasucci LM, Liuzzo G, Caligiuri G et aZ. Temporal relation between ischemic episodes and
    activation of the coagulation system in unstable angina. Circulation 1996; 93: 2121-7.
12. Merlini PA, Bauer KA, Oltrona L et aZ. Persistent activation of coagulation mechanism in
    unstable angina and myocardial infarction. Circulation 1994; 90: 61-8.
13. Catto A, Carter A, Ireland H et aZ. Factor V Leiden gene mutation and thrombin generation
    in relation to the development of acute stroke. Arterioscler Thromb Vasc Bioi 1995; 15: 783-5.
14. Bruhn HD, Conard J, Mannucci M et al. Multicentric evaluation of a new assay for prothrombin
    fragment FI +2 determination. Thromb Haemostas 1992: 68: 413-17.
15. Elias A, Bonfils S, Daoud-Elias M et af. Influence of long term oral anticoagulants upon
    prothrombin fragment 1+2, thrombin-antithrombin III complex and D-dimer levels in patients
    affected by proximal deep vein thrombosis. Thromb Haemostas 1993; 69: 302-5.
16. Granger CB, Miller JM, Bovill EG et aZ. Rebound increase in thrombin generation and activity
    after cessation of intravenous heparin in patients with acute coronary syndromes. Circulation
    1995;91: 1929-35.
17. Merlini PA, Ardissino D, Bauer KA et af. Persistent thrombin generation during heparin
    therapy in patients with acute coronary syndromes. Arterioscler Thromb Vasc Bioi 1997; 17:
    1325-30.
18. Millenson MM, Bauer KA, Kistler JP, Barzegar S, Tulin L, Rosenberg RD. Monitoring 'mini-
    intensity' anticoagulation with warfarin: Comparison of the prothrombin time using a sensitive
    thromboplastin with prothrombin fragment FI +2 levels. Blood 1992; 79: 2034-8.
19. Sobel M, Stembergh C, Marques D, Grimsdale AS. A comparative study of heparin responses
    in arterial and venous thromboembolism using molecular markers for thrombosis. Circulation
    1993; 88: 426--31.
20. Solymoss S, Bovill EG. Markers of in vivo activation of coagulation: Interrelationships change
    with intensity of oral anticoagulation. Am J Clin Patho11996; 105: 293-7.
21. Baglin TP, Luddington R, Jennings I, Richards EM. Thrombin generation and myocardial
    infarction during infusion of tissue-plasminogen activator. Lancet 1993; 341:504-5.
22. Merlini PA, Ardissino D, Bauer KA et af. Activation of the hemostatic mechanism during
    thrombolysis in patients with unstable angina pectoris. Blood 1995; 86: 3327-32.
23. Scharfstein JS, Abendschein DR, Eisenberg PR et aZ. Usefulness of fibrinogenolytic and
    procoagulant markers during thrombolytic therapy in predicting clinical outcomes in acute
    myocardial infarction. Am J Cardiol1996; 78: 503-10.
24. Simioni P, Scarano L, Gavasso S et af. Prothrombin fragment 1+2 and thrombin-antithrombin
    complex levels in patients with inherited APC resistance due to factor V Leiden mutation. Br
    J Haematol1996; 92: 435-41.
25. Zoller B, Holm J, Svensson P, Dahlback B. Elevated levels of prothrombin activation fragment
    I +2 in plasma from patients with heterozygous Arg (506) to Gin mutation in the factor V
    gene (APC-resistance) and/or inherited protein S deficiency. Thromb Haemostas 1996; 75:
    270-4.
26. Ceriellow A, Giacomello R, Stel G et al. Hyperglycemia-induced thrombin formation in
    diabetes: the possible role of oxidative stress. Diabetes 1995; 44: 924-8.
27. Salah AA, Dorey LG, Dombrowski MP et aZ. Thrombosis and hormone replacement therapy
    in postmenopausal women. Am J Obstet Gyneco11993; 169: 1554-7.
28. Bartsch P, Welsch B, Albert M, Friedmann B, Levi M, Kruithof EKO. Balanced activation
    of coagulation and fibrinolysis after a 2-h triathlon. Med Sci Sports Exerc 1995; 27: 1465-70.
29. Bauer KA, Barzegar S, Rosenberg RD. Influence of anticoagulants used for blood collection
    on plasma prothrombin fragment Fl +2 measurements. Thromb Res 1991; 63: 617-28.
30. Greenberg CS, Hursting MJ, Macik BG, Ortel TL, Kane WH, Moore BM. Evaluation of
    preanalytical variables associated with measurement of prothrombin fragment 1.2. Clin Chern
    1994; 40: 1962-9.
31. Iversen LH, Thorlacius-Ussing O. Short-time stability of markers of coagulation and fibrinolysis
    in frozen plasma. Thromb Res 1996; 81: 253---{j I.
                                               221
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                                             222
24
Tissue-type plasminogen activator
(t-PA) activity
c. KLUFT, P. MEIJER, E. ERSDAL and S. ROSEN
INTRODUCTION
+ t·PA activity
Figure 24.1 Occurrence of t-PA and PAI-I in an average, healthy, young volunteer. The vertical
axis represents relative molar concentrations
PRINCIPLE
The assay of t-PA captured by the antibody in the micro titre plate involves
two parallel reactions resulting from the simultaneous addition of plasminogen
and chromogenic plasmin substrate:
1. Activation of plasminogen in solution by active t-PA immobilized by
   antibodies bound to the microtitre wall
     Alr-t-PA + plasminogen -+ plasmin
     (bound)         (free)
2. Generated plasmin acts on the chromogenic substrate and the release of
   p-nitroaniline (pNA) is detected at 405 nm
     Plasmin + chromogenic substrate -+ pNA
The pNA generated after a fixed time interval is compared to corresponding
data of a calibration curve obtained by using different dilutions of a purified
t-PA standard. This t-PA standard should be calibrated against the WHO t-PA
standard, presently lot 86/670. The concentration ranges of t-PA that can be
evaluated are flexible and dependent upon the incubation times chosen for
                                            224
                                  t-PAACTIVITY
binding t-PA and for plasmin generation. These times should in turn be selected
according to the petformance of the actual antibody used. With a high-petforming
antibody, 1 h-long incubation times are sufficient for the detection of as little as
0.02 IU/mL.
SOLUTIONS
1. Distilled water.
2. 0.1 mollL Tris-acetate washing buffer, pH 8.4, 0.1% (v/v) Tween 80.
3. Assay reagents with 0.1 mg/ml Glu-plasminogen and 1 mmollL S-2403 in
   washing buffer.
4. t-PA standard dilution range made in 0.1 mollL Tris-HCl pH 8.4, 0.15 mollL
   NaCl, 5 mmollL disodium-EDTA, 0.1% (v/v) Tween 80.
ASSAY PROCEDURE
Blood collection 10
Select for venipuncture the sitting or lying position for the whole study and
allow for sufficient time to adjust to changes. Sitting or lying makes a difference
to haematocrit. The effects introduced by dilution with the usual liquid anti-
coagulants in blood tubes accentuate the difference between the two situa-
tions. Short-term effects of smoking, stress, exercise, alcohol, or beverages
containing caffeine on t-PA blood levels are known to exist; to minimize these,
subjects should refrain from smoking and the beverages mentioned for at least
1 h before venipuncture, and refrain for 18-24 h from alcohol. To minimize
the effects of stress, samples are taken from subjects who have been resting for
20 min. Fasting is preferred, but a light breakfast without fat and no tea or
coffee is permissible for morning samples. For sampling at other periods the
lunch and dinner times should be documented as well as the composition of
the meals. In view of the diurnal variations in t-PA, the time, or times, of blood
collection should be carefully chosen, standardized in studies, and be registered.
Subjects should have a normal or stable day/night rhythm in their lifestyle and
not be in a process of adaptation (unless this is the subject of the study).
                                        225
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   Blood should be collected in pre-cooled plastic or siliconized tubes containing
a specific anticoagulant. To prevent t-PA from being inactivated, blood should
reach pH 6 as soon as possible, which can be achieved with special anticoagulants
such as the commercially available Stabilyte®(Biopool) tubes 3 or rapid acidifi-
cation.
   The acidified blood can be kept at room temperature, but preferably at 4°C,
and is centrifuged within 2-4 h after collection. The minimum condition is
centrifugation for 20 min at 1000g. The plasma collected can be stored frozen,
but the pH 6 condition requires freezing by a thorough and rapid method
(snap-freezing). For all storage the temperature should be constantly below
-30°C; storage at -70 °C or lower is strongly recommended for this type of
plasma.
Laboratory assay
1. 50,.u of undiluted plasma at pH - 6, and 100 ,.u distilled water are added
   to a microtitre plate well coated with a monoclonal t-PA antibody (thus
   maintaining a pH of 6 during the binding step).
2. Incubation for 1 h at room temperature.
3. Empty the well and wash three times with 300 III 0.1 mol/L Tris-acetate
   washing buffer, pH 8.4, 0.1% (v/v) Tween 80.
4. Add 150 ,.u assay reagent containing a mixture of 0.08 mglml Glu-plasminogen
   and 1 mmollL chromogenic substrate S-2403.
5. Read 00 at 405 nm directly after the addition in 4 and after 15, 30 or
   60 min, depending on the expected range of activities (e.g. pre- or post-
   occlusion; cf. Chapter 31).
6. Calculation of results: the t-PA activity of the test sample can be expressed
   in IVlml from evaluation using a standard curve obtained from parallel runs
   of t-PA standard dilutions.
(PATHO)PHYSIOLOGY
For most data reported before 1990, when adequate blood sampling for t-PA
activity was discovered, the variability in t-PA activity4 requires re-analysis
whether or not it concerned a true variation in circulating t-PA activity and
was not a consequence of variability of PAI-l expressed via effects in vitro.
Data on circulating t-PA activity are limited so far.
   With the method described, analysis of Stabilyte plasma from 67 healthy
volunteers (aged 20-65 years; 22 males, 45 females) sampled at rest in the
morning showed a median value of 0.46 IVlml and a wide range from 0.07 to
1.3 IU/ml9 • For 40 of these individuals blood was also collected in parallel in
normal citrate showing a median value of 0.03 IU/ml (range 0-0.5), illustrating
the importance of the proper collection of the blood9 • Comparable values were
observed in other studies on 94 young women not using oral contraceptives
(unpublished), in healthy males (mean age 43)11; and in two groups of 40
healthy males and females (age 20-55 years)12. The mean values reported in
the studies mentioned were 0.385 (n =94); 0.29 (n =8)11; 0.34 (n =24)11; 0.29
(n =40)12 and 0.27 IUlml (n =40)12.
                                       226
                                  t-PA ACTIVITY
PITFALLS
1. For each variant of the method a check should be included for a number
   of potential interfering and essential factors. These include:
                                        227
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
References
 1. KIuft C. Constitutive synthesis of tissue-type plasminogen activator (t-PA) and plasminogen
    activator inhibitor type I (PAl-I): conditions and therapeutic targets. Fibrinolysis 1994; 8
    (Suppl. 2): 1-7.
 2. Chmielewska J, Ranby M, Wiman B. Evidence for a rapid inhibitor to tissue plasminogen
    activator in plasma. Thromb Res 1983; 31: 427-36.
 3. Ranby M, Sundell IB, Nilsson TK. Blood collection in strong acidic citrate anticoagulant
    used in a study of dietary influence on basal tPA activity. Thromb Haernostas 1989; 62: 917-22.
 4. Verheijen JH. Tissue-type plasminogen activator activity assay. In: Jespersen J, Bertina RM,
    Haverkate F, eds. ECAT assay procedures. A manual of laboratory techniques. Dordrecht:
    KIuwer, 1992; 139--46.
 5. Maat de MPM, KIuft C, Boer de K, Knot EAR, Jie AFH. Acid treatment of plasma for the
    inactivation of plasminogen activator inhibitor-l (PAl-I). Thromb Res 1988; 52: 425-30.
 6. Wejkum L, Rosen S, S0rskog L, Brandt B, Chmielewska I Blood sampling and determination
    of tissue plasminogen activator activity with COA-SE~ t-PA. Fibrinolysis 1990; 4 (Suppl.
    2): 152-4.
 7. Mahmoud M, Gaffney PI Bioirnmunoassay (BIA) of tissue plasminogen activator (t-PA)
    and its specific inhibitor (t-PAJINH). Thromb Haernostas 1985; 53: 356-9.
                                               228
                                         t-PA ACTIVITY
 8. Meijer P, Boon R, Jie AFH, Rosen S, Kluft C. Bioimmunoassay for tissue-type plasminogen
    activator (t-PA) in human plasma: Evaluation of blood sampling and handling procedures
    and comparison with other t-PA activity methods. Fibrinolysis 1992; 6 (Suppl. 3): 97-9.
 9. Rosen S, Wejkum L, Billing-Claeson S, Ghosh R, Grdic K, Chmielewska J, Meijer P, Kluft
    C, Tengborn L, Larsson G, Conkie J, Walker 1. Evaluation of a bioimmunoassay for t-PA
    activity and its relation to PAl-I activity and antigen levels. 1998 (Submitted).
10. Kluft C, Meijer P. Update 1996: Blood collection and handling procedures for assessment of
    plasminogen activators and inhibitors (Leiden Fibrinolysis Workshop). Fibrinolysis 1996; 10
    (Suppl. 2): 171-9.
11. Velthuis-te Wierik EJM, Meijer P, Kluft C, Berg YD. Beneficial effect of a moderately energy-
    restricted diet on fibrinolytic factors in non-obese men. Metabolism 1995; 44: 1548-52.
12. Velthuis-te Wierik EJM, Kluft C, Berg VD, Weststrate JA. Consumption of reduced-fat
    products, haemostatic parameters and oral glucose tolerance test. Fibrinolysis 1996; 10: 159--66.
13. Marckmann P, Bladbjerg E, Jespersen J. Dietary fish oil (4 g daily) and cardiovascular risk
    markers in healthy men. Arterioseler Thromb Vase Bioi 1997; 17: 3384-91.
14. Myrup B, Rossing P, Jensen T, Gram J, Kluft C, Jespersen J. The effect of the relationship
    between tissue-type plasminogen activator and plasminogen activator inhibitor type I on
    tissue-type plasminogen activator activity in insulin-dependent diabetes mellitus. Fibrinolysis
    1994; 8 (Suppl. 2): 22-4.
15. Myrup B, Rossing P, Jensen T, Gram J, Kluft C, Jespersen J. Fibrinolysis in insulin-dependent
    diabetic patients with and without nephropathy. Fibrinolysis 1996; 10: 331-5.
16. Schuit AJ, Schouten EG, Kluft C, de Maat M, Menheere PP, Kok FJ. Effect of strenuous
    exercise on fibrinogen and fibrinolysis in healthy elderly men and women. Thromb Haemostas
    1997; 78: 845-51.
17. Wa11nofer AE, van Griensven JM, Schoemaker HC, Cohen AF, Lambert W, Kluft C, Meijer
    P, Kooistra T. Effect of isotretinoin on endogenous tissue-type plasminogen activator (t-PA)
    and plasminogen activator inhibitor I (PAl-I) in humans. Thromb Haemostas 1993; 70:
    1005-8.
18. Eliasson M, Evrin P-E, Lundblad D, Asplund K, Ranby M. Influence of gender, age and
    sampling time on plasma fibrinolytic variables and fibrinogen. Fibrinolysis 1993; 7: 316-23.
19. Chandler WL, Schwartz RS, Stratton JR, Vitiello MY. Effects of endurance training on the
    circadian rhythm of fibrinolysis in men and women. Med Sci Sports Exerc 1996; 28: 647-55.
20. van der Born JG, de KnijffP, Haverkate F, Bots ML, Meijer P, de Jong PT, Hofman A, Kluft
    C, Grobbee DE. Tissue plasminogen activator and risk of myocardial infarction. The Rotterdam
    Study. Circulation 1997; 95: 2623-7.
21. Jern C, Ladenva1l P, Wall U, Jern S. Association between a tissue-type plasminogen activator
    (t-PA) gene polymorphism and forearm vascular release rate of t-PA. In: In vivo studies on
    local release of tissue-type plasminogen activator from vascular endothelium. Wall University
    thesis, Gotenborg University, Sweden, 1997.
22. Stegnar M, Mavri A. Reproducibility of fibrinolytic response to venous occlusion in healthy
    subjects. Thromb Haemostas 1995; 73: 453-7.
23. Lindahl B, Asplund K, Eliasson M, Evrin P-E. Insulin resistance syndrome and fibrinolytic
    activity: the Northern Sweden Monica study. Int J Epidemiol1996; 25: 291-9.
24. Brussaard HE, Gevers Leuven JA, Krans HMJ, Meijer P, Buytenhek R, Kluft C. Non-esterified
    fatty acids are related with hypofibrinolysis in type 2 diabetes mellitus. Fibrinolysis 1994; 8
    (Supp!. 2): 25-7.
25. Szymanski LM, Durstine JL, Davis PG, Dowda M, Pate RR. Factors affecting fibrinolytic
    potential: cardiovascular fitness, body composition, and lipoprotein(a). Metabolism 1996; 45:
    1427-33.
26. Shaukat N, Douglas IT, Bennett JL, Bono de DP. Can physical activity explain the differ-
    ences in insulin levels and fibrinolytic activity between young Indo-origin and European
    relatives of patients with coronary artery disease? Fibrinolysis 1995; 9: 55-63.
27. Ishizaki M, Tsuritani I, Noborisaka Y, Yamada Y, Tabata M, Nakagawa H. Relationship
    between job stress and plasma fibrinolytic activity in male Japanese workers. Int Arch Occup
    Environ Health 1996; 68: 315-20.
28. Eritsland J, Seljeflot I, Abdelnoor M, Arnesen H. Long-term influence of omega-3 fatty acids
    on fibrinolysis, fibrinogen, and serum lipids. Fibrinolysis 1994; 8: 120-5.
                                                229
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
29. Eliasson M, Asplund K, Evrin P-E, Huhtasaari F, Johansson I. Plasma fibrinogen, fibrinolysis
    and (pro)vitamins; is there a connection? Fibrinolysis 1995; 9: 87-92.
30. Juhan-Vague I, Pyke SDM, Alessi MC, Jespersen J, Haverkate F, Thompson MA on behalf
    of the ECAT Study Group. Fibrinolytic factors and the risk of myocardial infarction or
    sudden death in patients with angina pectoris. Circulation 1996; 94: 2057-63.
31. Chandler WL, Stratton JR. Laboratory evaluation of fibrinolysis in patients with a history
    of myocardial infarction. Am J Clin Patho11994; 102: 248-52.
32. Brussaard HE, Gevers Leuven JA, Frolich M, KIuft C, Krans HM. Short-term oestrogen
    replacement therapy improves insulin resistance, lipids and fibrinolysis in postmenopausal
    women with NIDDM. Diabetologia 1997; 40: 843-9.
33. Ishii A, Yamada S, Yamada R, Hamada H. T-PA activity in peripheral blood obtained from
    pregnant women. J Perinat Med 1994; 22: 113-17.
34. Stegnar M, Meglic A, Meglic L, Novak-Antolic Z. Fibrinolysis after delivery: Caesarean
    section versus vaginal delivery. Fibrinolysis 1994; 8: 270--5.
35. Pernot C, Hartgens F, Keizer HA, Kuipers H, Hamulyak K. Effects of self-administration
    of high dose androgenic-anabolic steroids on fibrinolytic activity in non-elite bodybuilders.
    Fibrinol Proteo11996; 10 (Suppl. 2): 53-4.
36. Ferro D, Quintarelli C, Saliola M, Allessandri C, Basili S, Bonavita MS, Violi F. Prevalence
    of hyperfibrinolysis in patients with liver cirrhosis. Fibrinolysis 1993; 7: 59-62.
37. Nielsen JD, Gram J, Fabrin K, Holm-Nielsen A, Jespersen J. Lack of correlation between
    blood fibrinolysis and the immediate or post-operative blood loss in transurethral resection
    of the prostate. Br J Haematol1997; 80: 105-10.
38. Huisman LGM, Griensven van JMT, KIuft C. Usefulness and limitations of Actilyse for
    standardisation of tissue-type plasminogen activator (t-PA) assays in blood. Fibrinolysis 1996;
    10 (Suppl. 2): 125-7.
                                               230
25
Tissue-type plasminogen activator
antigen (t-PA Ag)
M. C. ALESSI and I. JUHAN-VAGUE
INTRODUCTION
Physiological role
Tissue-type plasminogen activator (t-PA) constitutes an important agent in the
fibrinolytic pathway. It is synthesized and released continuously into the blood
by endothelial cells and then rapidly cleared by the liver. The secreted form, a
single-chain glycoprotein composed of 530 acids (70 kD), can be converted by
plasmin or kallikrein into a double-chain form with one disulphide bond.
   Several domains have been distinguished on the molecule: a finger domain
homologous to the type I fingers of fibronectin, an epidermal-growth-factor-like
domain, two 'kringle' domains similar to the triple disulphide bound structures
found in many plasma proteins, a connecting peptide between the second
kringle and the plasmin cleavage site and the carboxyl terminal domain
constituting the chain bearing the active site.
   The physiological role of t-PA is to activate plasminogen to plasmin which
degrades fibrin to soluble degradation products. Fibrinolysis appears to be
regulated by specific molecular interactions between t-PA and fibrin, as well
as between plasmin and plasmin inhibitor. Because fibrin is required as a
cofactor, the activation of plasminogen can occur only on the fibrin surface
(for review see ref. 1).
Pathophysiological role
Circulating t-PA levels are thought to have a major effect on fibrinolytic capacity,
and proper regulation of the t-PA is essential. Increased plasma levels of t-PA
antigen have been found to be predictive of arterial ischaemic events 2 • It is
                                        231
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
noteworthy that this increase in circulating t-PA antigen does not reflect an
increase in fibrinolytic capacity but rather the opposite. This has been attributed
to a parallel evolution of PAI-I which results in t-PAlPAI-1 complexes rather
than a rise in active t-PA.
   Decreased release of t-PA has been associated with thrombosis. Various tests
have been designed to evaluate t-PA release. Different aspects of the response
are probably measured depending on the stimulus used, e.g. venostasis (Chap-
ter 31)3, desamino-8-o-arginine vasopressin (DDAVP) injection4 or physical
exerciseS. The capacity of the endothelium to release t-PA can be assessed by
comparing either t-PA activity or antigen level before and after stimulation.
   Circadian variations in t-PA antigen have been described in relation with
variations in plasminogen activator inhibitor 1 (PAl-I). Increase in t-PA antigen
concentration has been reported in the elderly and in inflammatory syndrome,
in association with several clinical conditions involving increased PAI-I
concentration. The association of hyperfibrinolysis with haemorrhage has been
attributed to a congenital lack of inhibitor, but a tendency to bleeding has also
been observed in patients displaying high t-PA release. Venostasis revealed
defects in t-PA release in patients with severe von Willebrand disease and
abnormally low release of t-PA has been reported in 10-20% of patients with
recurrent deep vein thrombosis (for review see ref. 6, and see Chapter 31).
METHODOLOGY
t-PA activity in whole blood or plasma varies due to reactions between t-PA
and inhibitors resulting in formation of t-PA inhibitor complexes. Immunological
determination of t-PA allows a total quantification of circulating t-PA.
   Efforts to develop assay procedures for t-PA antigen in plasma have been
under way for many years and several techniques have been described7- 17• The
earliest methods were radioimmunoassays but they were soon replaced by
enzyme-linked immunosorbent assay (ELISA) which have the advantage of
using a stable enzyme label. The switch to ELISA was concomitant with
improvements in calibrator preparation, in antibody sources and in the buffer
as well as the tracer. Current commercially available kits contain stable and
reproducible reagents (see Table 25.1).
Techniques
Radioimmunoassay
Radioimmunoassay based on 12sI-labelled antibodies against t-PA ('2sI_Ab)
was the first method to be used. Briefly samples were incubated in polystyrene
tubes of microtitre plates coated with IgG from goat or rabbit antiserum. After
washing, 125I_Ab was added. Finally, after another wash, radioactivity on the
walls of the recipient was counted in a y-scintillator. Results were evaluated by
comparison with a standard curve constructed with pure t-PA.
   In one variant of this technique, the sample, antiserum and l2SI-t-PA were
                                       232
                                 t-PA ANTIGEN
ELISA
ELISA techniques rapidly superseded radioimmunoassay because they are so
much easier to perform. Specific IgG or the F(ab')2 fragments were coupled
on polystyrene wells in which samples were incubated for a few hours. After
washing, peroxidase or [3-D-galactosidase-labelled antibodies were added.
Specific substrate (orthophenylenediamine methylumbelliferyl [3-D galactoside)
was then added for various periods of time depending on the assay. The
relationship between t-PA and fluorescence intensity or coloration was measured
using a spectrofluorometer (ex 360 nm, em 450 nm) or a spectrophotometer
(492 nm) respectively.
   Rijken et al. 9 described a three-step ELISA procedure with the last step
consisting of the use of a rabbit anti-(goat) IgG labelled with alkaline phos-
phatase. Holvoet et al. 14 proposed a two-site ELISA using a coating phase
comprising the IgG fractions of two monoclonal antibodies directed against
different exposed epitopes on the t-PA molecule. They also proposed an ELISA
for free t-PA using a murine monoclonal antibody directed against the active
site of t_PA I8 • Several groups have also developed ELISA techniques to quantify
t-PA complexed with PAI-I 19,20.
Materials
Calibrator t-PA
t-PA is synthesized and secreted at a much higher level by numerous cultured
human cell lines than by endothelial cells. t-PA purified from the culture medium
of Bowes melanoma cell line has served as a rich source of calibrator t-PA.
Several purification processes have been described. t-PA was first purified using
immunoaffmity chromatography with antibodies directed against porcine t-PA,
chromatography on arginine Sepharose and gel filtration. Then extraction of
t-PA was achieved by three chromatographies using zinc chelate Sepharose and
concanavalin A Sepharose, followed by filtration on Sephadex G50. More
recently t-PA has been purified by immunoaffinity chromatography using
monoclonal antibodies directed against human t-PA.
   Wun and Capuano 17 obtained calibrator t-PA from Hela cells stimulated
with 12-0-tetradecanoyl phorbol13 acetate by chromatography on Erythrina
latissima inhibitor Sepharose 4B. The resulting product was a mixture of the
single- and double-chain forms. Purification in the presence of aprotinine
allowed production of a 90% pure single-chain form (sct-PA). t-PA has been
produced by DNA recombinant technology. Concentrations in the purified
preparation were determined by protein quantification and amino acid analysis.
                                      233
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
  International standard NIBSC for t-PA was first prepared in 1983 from
melanoma cells (coded 83/517) and then in 1986 (coded 86/670). This latter
was prepared by high-performance liquid chromatography on a gel exclusion
column. Preparation contained 98% sct-PA2 !.
Antibodies
All the assays described up to now have made use of specific antibodies raised
in goats or rabbits. IgG were purified by ammonium sulphate precipitation or
from protein A Sepharose. Monoclonal antibodies have also been produced in
order to obtain stable and reproducible antibodies.
Sample collection
Blood samples should be taken after a 20-min rest period (see also Chapter 3).
Since t-PA levels are subject to circadian variations, timing should be carefully
standardized and subjects should have a normal day/night rhythm. A good
clear venipuncture should be made using minimal stasis. Both free flow and
vacuum techniques can be used. The first few millilitres of blood should be
discarded. Cooling does not seem to be necessary. Any anticoagulant is suitable.
Interestingly, t-PA antigen in plasma is not essentially influenced by the pH of
the anticoagulant or by the presence of platelet-stabilizing agent (CTAD).
Anticoagulant should be consistently applied when chosen. Preparation of
plasma is not very critical as long as haemolysis and clotting in the blood is
prevented, and cellular elements are largely removed. Blood can be kept at
temperatures between 25°C and 4 °C for at least 2 days; significant losses are
encountered after 24 h when 37°C is used. For prolonged storage, plasma
aliquots can be frozen and thawed for assay. Reported snap-freezing and rapid
thawing is allowed23 •
Problems
False-positives
The presence of an excess of non-immune IgG can reduce the risk of false-
positives resulting from the presence of antibodies against IgG or rheumatoid
factors in the plasma samples.
                                      234
                                  t-PA ANTIGEN
Calibrator
Crypticity
Two major problems with current immunoassays were poor recovery and lack
of parallelism between the dilution curves of plasma and buffer. However, t-PA
in serum free cell culture media requires no manipulation for full detection of
antigen and addition of EDTA, poly-L-Iysine to plasma improves results24 •
Wun and Capuano 17 have shown that the addition of lysine or arginine (final
concentration 0.5 moVL) and slight acidification of plasma are highly efficient
in revealing the plasma t-PA antigen. This portion has been termed cryptic.
   This phenomenon has been attributed to the efficiency of the reaction between
t-PA antigen in citrated plasma and immobilized t-PA-specific antibodies. The
reaction rate can be considerably enhanced by adding EDTA, lysine or acid.
It has been speculated that a large portion of the t-PA in citrated plasma is in
a complexed form. EDTA, lysine or acid would have the capacity to dissociate
these complexes, making t-PA accessible to the antibody.
A suitable assay for t-PA antigen is an ELISA using polyclonal antibodies and
peroxidase substrate (Biopool). This assay appears to be particularly reliable,
as background due to non-specific immunoglobulin, i.e. rheumatoid factors,
is eliminated by determining two optical densities: one is the presence of normal
goat IgG (N-well) and the other is the presence of goat anti t-PA IgG (A-well).
The t-PA signal corresponds to the difference between the N-well and the
A-well. In order to enhance recovery and suppress t-PA crypticity, a dilution
buffer composed of saline phosphate containing Ig/L Tween 20 and 5 mmoVL
EDTA is used. This composition is as effective as cidification or lysine. The
assay allows direct determination of the immunoreactivity of t-PA alone or
complexed with purified inhibitors, i.e. PAl-I, PAI-2, plasmin inhibitor, CI
inhibitor. Consistently precise results have been obtained by different laboratories,
the 'within' and 'between' assay coefficients being 5% and 8% respectively.
    Several other ELISA kits are now available for t-PA antigen quantification
in plasma. Some use F(ab')2 antibody fragments to prevent interference from
rheumatoid factors. Others use monoclonal or polyclonal antibodies. Various
types of calibrator t-PA are used; it may be obtained from melanoma cells or
recombinant technology. Since most of these assays use an adequate buffer,
they feature low detection limits and satisfactory reproducibility (Table 25.1).
                                        235
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                   Table 25.1    Leading manufacturers
CONCLUSION
Methods for measuring t-PA antigen in plasma have improved in the past few
years. Several techniques have been proposed but few comparative data are
available. A complete characterization of each is needed to determine reactivity
against all the forms of t-PA. This should lead to better inter-laboratory
correlation.
References
 I. Bachmann F. Fibrinolysis. In: Verstraete M, Vermylen J, Lijnen R, Arnout J, eds. Thrombosis
    and haemostasis. Leuven: Leuven University Press, 1987; 227--65.
 2. Juhan-Vague I, Pyke SDM, Alessi MC, Jespersen J, Haverkate F, Thompson SG. Fibrinolytic
    factors and the risk of myocardial infarction or sudden death in patients with angina pectoris.
    Circulation 1996; 94: 2057.
 3. Robertson B, Pandolfi M, Nilsson 1M. Fibrinolytic capacity in healthy volunteers at different
    ages as studied by standardized venous occlusion of arms and legs. Acta Med Scand 1972;
    191: 199.
 4. Brommer EJP, Barrett-Bergshoeff MM, Allen RA, Schicht L, Bertina RM, Shalekamp
    MADH. The use of desmopressin acetate (DDAVP) as a test of the fibrinolytic capacity of
    patients, analysis of responders and non-responders. Thromb Haemostas 1982; 48: 156.
 5. Cash JD. Control mechanism of activator release. In: Davidson JF, Rowan RM, Samama
    MM, Desnoyers PC, eds. Progress in chemical fibrinolysis and thrombolysis, vol. 3. New York:
    Raven Press, 1978; 65.
 6. Juhan-Vague I, Aillaud MF, Alessi Me. Biological variation in t-PA activity and antigen. In:
    KIuft C, ed. Tissue-type plasminogen activator (t-PA): physiological and clinical aspects, vol.
    2. Boca Raton, FL: CRC Press, 1988; 69.
 7. Holmberg L, Kristoffersson A, Lecander I, Wallen P, Astedt B. Immunoradiometric
    quantification of tissue plasminogen activator secreted by fetal organs. Comparison with
    urokinase. Scan J Clin Lab Invest 1982; 42: 347.
 8. Mac Gregor IR, Prowse Cv. Tissue plasminogen activator in human plasma measured by
    radioimmunoassay. Thromb Res 1983; 31: 461.
 9. Rijken DC, Juhan-Vague I, DeCock F, Collen D. Measurement of human tissue-type
    plasminogen activator by a two-site immunoradiometric assay. J Lab Clin Med 1983; 101:
    274.
10. Matsuo 0, Kato K, Matsuo C, Matsuo T. Determination of tissue plasminogen activator by
    an enzyme-immunoassay method. Analyt Biochem 1983; 135: 58.
II. Bergsdorf N, Nilsson T, Wallen P. An enzyme linked immunosorbent assay for determination
    of tissue plasminogen activator applied to patients with thromboembolic disease. Thromb
    Haemostas 1983; 50: 740.
                                               236
                                        t-PA ANTIGEN
12. Urden G, Blombiick M. Determination of tissue plasminogen activator in plasma samples
    by means of a radioimmunoassay. Scand J Clin Invest 1984; 44: 495.
13. Rijken DC, Van Hinsbergh VWM, Sens EHe. Quantitation of tissue-type plasminogen
    activator in human endothelial cell cultures by use of an enzyme immunoassay. Thromb Res
    1984; 33: 145.
14. Holvoet P, Cleemput H, Collen D. Assay of human tissue type plasminogen activator (t-PA)
    with an enzyme linked immunosorbent assay (ELISA) based on three murine monoclonal
    antibodies to t-PA. Thromb Haemostas 1985; 54: 684.
15. Kaizu T, Kojima K, Iwasaki K, Yamashita T. One-step sandwich enzyme-linked immunosorbent
    assay of human tissue plasminogen activator using monoclonal antibodies. Thromb Res 1985;
    40: 91.
16. Takada A, Shizume K, Ozawa T, Takahashi S, Takada Y. Characterization of various antibodies
    against tissue plasminogen activator using highly sensitive enzyme immunoassay. Thromb
    Res 1986; 42: 63.
17. Wun TC, Capuano A. Immunoradiometric quantitation of tissue plasminogen activator-
    related antigen in human plasma: crypticity phenomenon and relationship to plasma fibrinolysis.
    Blood 1987; 69: 1348.
18. Holvoet P, Boes J, Collen D. Measurement of free, one chain tissue-type plasminogen activator
    in human plasma with an enzyme-linked immunosorbent assay based on an active site specific
    murine monoclonal antibody. Blood 1987; 69: 284.
19. Amiral J, Plassart V, Grosley M, Mimilla F, Contant G, Guy-Ader AM. Measurement of
    t-PA and t-PAlPAI-1 complexes by ELISA, using monoclonal antibodies: clinical relevance.
    Thromb Res 1988 (Suppl. VII): 99.
20. Rijken DC, Juhan-Vague I, Collen D. Complexes between tissue-type plasminogen and
    proteinase inhibitors in human plasma identified with an immunoradiometric assay. J BioI
    Chem 1982; 101: 285.
21. Gaffney PJ, Curtis AD. A collaborative study to establish the 2nd international standard for
    tissue plasminogen activator (t-PA). Thromb Haemostas 1987; 58: 1085.
22. Gaffney PI Specific assays of plasminogen activators and inhibitors. European School of
    Hematology: Fibrinolysis, 2--6 October 1989.
23. KIuft C, Meijer P. Update 1996: Blood collection and handling procedures for assessment of
    plasminogen activators and inhibitors (Leiden Fibrinolysis Workshop). Fibrinolysis 1996; 10
    (Suppl. 2): 171.
24. Ranby M, Nguyen G, Scarabin PY, Samama M. Immunoreactivity of tissue plasminogen
    activator and of its inhibitor complexes. Thromb Haemostas 1989; 61: 409.
                                               237
26
Plasminogen activator inhibitor-1
(PAI-1) antigen
P. J. DECLERCK
INTRODUCTION
Calibration
To date, no official calibrator for PAI-l antigen is available. Two NIBSC prepara-
tions have been made, i.e 87/512 and 921654. NIBSC 87/512, an interim standard
prepared by adding melanoma PAI-l to human plasma, contains a PAI-l
antigen level of 175 nglampoule24,25. NIBSC 92/654 was prepared by adding
recombinant PAI-l to human plasma26 and contains approximately 200 ng
PAI-l antigen/ampoule. However, a recent multi-laboratory calibration study
did not result in the assignment of a consensus value for preparation NIBSC
92/654 (reported at the ISTH-SSC Subcommittee meeting on Fibrinolysis,
Florence 1997). Anyway, for the time being these values may serve as a guide.
Table 26.1 Relative amounts of PAI-l antigen and PAI-l activity in plasma and platelets
Expressed relative to total PAI-l antigen present in blood (arbitrarily set at '100')
                                               240
                                      PAI-1 ANTIGEN
Table 26.2 Available methods for the measurement of human PAI-l antigen in plasma
Assay dependency
Initial studies had shown that PAl-1 antigen values also strongly depend upon
the assay used. Therefore it is obvious, that when reporting PAI-l antigen
values in a study: (a) the type of the assay and its characteristics should be
indicated and (b) samples from a normal, age-matched population should be
included in the analysis.
    The reason for this assay dependency may be found in two parameters: (a)
the differential reactivity of the antibodies towards the different forms or (b)
the calibration of the standard. A multicentre study24 on the evaluation of
seven commercially available kits (Table 26.2) for the determination of PAI-I
antigen in plasma revealed differences ranging between 4- and 10-fold in various
normal samples. Importantly, it was demonstrated that, after normalization
of the data (relative to a well-defined, amino-acid-calibrated PAI-I sample),
for six out of seven kits no significant differences could be observed. One kit,
even after normalization of the values, yielded data that were significantly
different from the normalized data obtained with the other kits. This indicated
that the major cause of differences in 'absolute' antigen values obtained with
the different kits is mainly due to differences in calibration of the standard.
The results also suggested that, even though PAl-l can adopt various conforma-
tions, and that various monoclonal antibodies are used in the assay kits, the
potential differential reactivity of the different PAl-l forms may, in the majority
of cases, be considered to be of minor importance.
    It is important to realize that these observations were obtained with
'normal' plasma samples and may be extrapolated to several clinical situa-
tions. Usually, t-PA antigen and PAI-l antigen in plasma correlate quite
well, and it has been shown that levels of active PAI-l and t-PA/PAI-l
complexes are not independent 29 • However, due to differences in relative
sensitivities between active PAl-I, t-PAlPAI-l complexes and latent PAl-I,
it cannot be excluded that in particular disease states in which the coordination
between t-PA and PAI-l levels is disturbed 3o , or in which a specific increase
in platelet PAI-l is observed 31 , the different kits may respond differently
and correlations may be less pronounced. Therefore, in particular clinical
situations putative, small differences between the kits may become much
more pronounced. A similar remark is valid for analysis of PAI-I in samples
not from plasma origin (e.g. tissue extracts, cell culture supernatants) in
which PAI-I may occur predominantly in only one form (e.g endothelial cell
culture supernatants containing mainly latent PAl-I).
                                       242
                                       PAI-1 ANTIGEN
Evaluation of PAI·1 antigen from other species
A number of experimental animal models (mice, rats, rabbits, pigs) have been
developed to study thrombosis, atherosclerosis32, and are being used to evaluate
the role of PAl-I in various disease states. In general the immunological reagents
raised again human PAI-I are not suitable for the development of sensitive
assays for the specific immunological ~uantitation of PAI-I from other species.
Recently, PAI-I from other species 3-36has been expressed, purified and
characterized. Subsequently, monoclonal antibodies were raised against murine
PAl-I, rat PAI-I and porcine PAl-I, and are being applied for the construction
of monoclonal antibody-based ELISAs allowing the specific detection of PAI-I
antigen in the respective species37- 39•
References
 I. Declerck PI, Iuhan Vague I, Felez I, Wiman B. Pathophysiology of fibrinolysis. I Intern Med
    1994:236;425-32.
 2. Schleef RR, Higgins DL, Pi11emer E, Levitt LJ. Bleeding diathesis due to decreased functional
    activity of type I plasminogen activator inhibitor. I Clin Invest 1989: 83 : 1747-52.
 3. Dieval I, Nguyen G, Gross S, Delobel I, Kruithof EK. A lifelong bleeding disorder associated
    with deficiency of plasminogen activator inhibitor type I. Blood 1991; 77: 528-32.
 4. Lee MH, Vosburgh E, Anderson K, McDonagh J. Deficiency of plasma plasminogen activator
    inhibitor I results in hyperfibrinolytic bleeding. Bleed 1993; 81 : 2357-62.
 5. Fay WP, Shapiro AD, Shih IL, Schleef RR, Ginsburg D. Brief report: complete deficiency of
    plasminogen-activator inhibitor type 1 due to a frame-shift mutation. N Engl I Med 1992;
    327: 1729-33.
 6. Fay WP, Parker AC, Condrey LR, Shapiro AD. Human plasminogen activator inhibitor I
    (PAl I) deficiency: characterization of a large kindred with a null mutation in the PAl I gene.
    Blood 1997; 90: 204-8.
 7. Erickson LA, Fici GI, Lund IE, Boyle TP, Polites HG, Marotti KR. Development of venous
    occlusions in mice transgenic for the plasminogen activator inhibitor-I gene. Nature 1990;
    346:74-76.
 8. Carmeliet P, Stassen 1M, Schoonjans L, Ream B, van den Oord JJ, De Mol M, Mulligan RC,
    Collen D. Plasminogen activator inhibitor-I gene deficient mice. II. Effects on hemostasis,
    thrombosis and thrombolysis. I Clin Invest 1993; 92: 2756-60.
 9. Urden G, Chmielewska I, Carlsson T, Wiman B. Immunological relationship between
    plasminogen activator inhibitors from different sources. Thromb Haemostas 1987; 57: 29-34.
10. MacGregor IR, Booth NA. An enzyme-linked immunosorbent assay (ELISA) used to study
    the cellular secretion of endothelial plasminogen activator (PAl-I). Thromb Haemostas 1998;
    59: 68-72.
II. Rijken DC, Sprengers ED, van der Hoeven RJ, Kluft C, Engesser L. Plasminogen activator
    inhibitor 1 (PAl-I) antigen levels in human plasma samples. Fibrinolysis 1988;2 (Suppl. 2) :
    79-82.
12. Kruithof EK, Nicolosa G, Bachmann F. Plasminogen activator inhibitor 1: development of
    a radioimmunoassay and observations on its plasma concentration during venous occlusion
    and after platelet aggregation. Blood 1987; 70 : 1645-53.
13. Lund LR, Georg B, Nielsen LS, Mayer M, Dano K, Andreasen PA. Plasminogen activator
    inhibitor type I: cell-specific and differentiation-induced expression and regulation in human
    cell lines, as determined by enzyme-linked immunosorbent assay. Mol Cell Endocrinol 1988;
    60: 43-53.
14. Booth NA, Simpson AJ, Croll A, Bennett B, MacGregor IR. Plasminogen activator inhibitor
    (PAl-I) in plasma and platelets. Br J Haematol1988; 70: 327-33.
15. Declerck PI, Alessi MC, Verstreken M, Kruithof EK, Juhan Vague I, Collen D. Measurement
    of plasminogen activator inhibitor I in biologic fluids with a murine monoclonal antibody-
    based enzyme-linked immunosorbent assay. Blood 1988; 71: 220-5.
                                              243
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
16. Declerck PJ, De Mol M, Vaughan DE, Collen D. Identification of a conformationally distinct
    form of plasminogen activator inhibitor-I, acting as a noninhibitory substrate for tissue-type
    plasminogen activator. I Bioi Chern 1992; 267: 11693-6.
17. Urano T, Strandberg L, Johansson LB, Ny T. A substrate-like form of plasminogen-activator-
    inhibitor type I. Conversions between different forms by sodium dodecyl sulphate. Eur J
    Biochem 1992; 209: 985-2.
18. Munch M, Heegaard CW, Andreasen PA. Interconversions between active, inert and substrate
    forms of denatured/refolded type-l plasminogen activator inhibitor. Biochim Biophys Acta
    1993; 1202: 29-37.
19. Declerck PJ, De Mol M, Alessi MC, Baudner S, Paques EP, Preissner KT, Muller Berghaus
    G, Collen D. Purification and characterization of a plasminogen activator inhibitor I binding
    protein from human plasma. Identification as a multimeric form of S protein (vitronectin). J
    Bioi Chern 1988; 263: 15454-61.
20. Sigurdardottir 0, Wiman B. Complex formation between plasminogen activator inhibitor I
    and vitronectin in purified systems and in plasma. Biochim Biophys Acta 1990; 1035: 56-61.
21. Declerck PJ, Verstreken M, Collen D. Measurement of different forms of plasminogen
    activator inhibitor 1 (PAl-I) using various monoclonal antibody-based enzyme-linked
    immunosorbent assays. Fibrinolysis 1990; 4 (Suppl. 2): 132-3.
22. Kluft C, Jie AF. Comparison of specificities of antigen assays for plasminogen activator
    inhibitor I (PAl-I). Fibrinolysis 1990; 4 (Suppl. 2): 136-7.
23. Juhan Vague I, Alessi MC, Fossat C, Declerck PJ, Kruithof EK. Plasma determination of
    plasminogen activator inhibitor I antigen must be performed in blood collected on antiplateletl
    anticoagulant mixture. Thromb Haemostas 1987; 58: 1096.
24. Declerck PJ, Moreau H, Jesperson J, Gram J, Kluft C. Multicenter evaluation of commercially
    available methods for the immunological determination of plasminogen activator inhibitor-l
    (PAl-I). Thromb Haemostas 1993; 70: 858-63.
25. Lijnen HR. Report of the meeting of the subcommittee on Fibrinolysis, 6-9 July 1992,
    Miinchen, Germany, 1992.
26. Gaffney PJ, Edgell TA. The international standard for plasminogen activator inhibitor I
    (PAl-I) activity. Thromb Haemostas 1996; 76: 80-83.
27. Kluft C, Verheijen JH. Leiden fibrinolysis working party: blood collection and handling
    procedures for the assessment of tissue-type plasminogen activator and plasminogen activator
    inhibitor 1. Fibrinolysis 1990; 4 (Suppl. 2): 155-{i1.
28. Kruithof EK, Tran Thang C, Gudinchet A, Hauert J, Nicoloso G, Genton C, Welti H,
    Bachmann F. Fibrinolysis in pregnancy: a study of plasminogen activator inhibitors. Blood
    1987; 69: 460-{i6.
29. Alessi MC, Juhan-Vague I, Declerck PJ, Anfosso F, Gueunoun E, Collen D. Correlations
    between t-PA and PAI-I antigen and activity and t-PAlPAI-1 complexes in plasma of control
    subjects and of patients with increased t-PA of PAI-I levels. Thromb Res 1990; 60: 509-16.
30. Andreotti F, de Bart AC, Regan T, Maseri A, Kluft C. Factors affecting the relation between
    plasminogen activator inhibitor (PAl) activity and PAI-I antigen levels in normals and in
    patients with arterial disease. Fibrinolysis 1990; 4 (Suppl. 2): 34--5.
31. Alessi MC, Juhan-Vague I, Declerck PJ, Collen D. Molecular forms of plasminogen activator
    inhibitor-I (PAl-I) and tissue-type plasminogen activator (t-PA) in human plasma. Thromb
    Res 1991; 62: 275-85.
32. Badimon L. Models to study thrombotic disorders. Thromb Haemostas 1997; 78: 667-71.
33. Bijnens AP, Knockaert I, Cousin E, Kruithof EK, Declerck PI Expression and characterization
    of recombinant porcine plasminogen activator inhibitor-I. Thromb Haemostas 1997; 77:
    350-{i.
34. Ngo TH, Knockaert I, Declerck PI Expression, purification and characterization of
    recombinant rat plasminogen inhibitor 1. Fibrinol Proteol 1997; II: 37-43.
35. Hofmann KJ, Mayer EJ, Schultz LD, Socher SH, Reilly CF. Purification and characterisation
    of recombinant rabbit plasminogen activator inhibitor-I expressed in Saccharomyces cerevisiae,
    Fibrinolysis 1992; 6: 263-72.
36. Carmeliet P, Kieckens L, Schoonjans L, Ream B, Van Nuffelen A, Prendergast G, Cole M,
    Bronson R, Collen D, Mulligan RC. Plasminogen activator inhibitor-I gene-deficient mice.
    I. Generation by homologous recombination and characterization. J Clin Invest 1993; 92:
    2746-55.
                                               244
                                      PAI-1 ANTIGEN
37. Declerck PJ, Verstreken M, Collen D. Immunoassay of murine t-PA, u-PA and PAI-1 using
    monoclonal antibodies raised in gene-inactivated mice. Throm Haemostas 1995; 74: 1305-9.
38. Ngo TH, Verheyen S, Knockaert I, Declerck PI Monoclonal antibody-based immunoassays
    for the specific quantitation of rat PAI-1 antigen and activity in biological samples. Thromb
    Haemostas. 1998; 79: 808-12.
39. Leng H, Brouwers E, Knockaert I, Declerck PI Monoclonal antibody based immunoassays
    for the specific quantitation of porcine PAI-1 antigen and activity in biological fluids. (In
    preparation).
                                              245
27
Plasminogen activity
P. J. GAFFNEY
INTRODUCTION
INDIRECT METHODS
Direct
I.   Affinity chromatography                    Zoltan et al. 21
2.   Haemaggiutination inhibition               Ludlam and Das22
3.   I=unoelectrophoresis                       Ganrot and Nilehn l6
4.   Latex flocculation                         Wuetal.20
5.   Radial i=unodiffusion                      Storiko23
6.   Radioi=unoassay                            Rabiner et al. 18
7.   Amidolytic (SK-plgn complex)               Friberger et al.24
Indirect
1. Caseinolytic                                 Re=ert and Cohen6*
2.   Esterolytic                                Troll et al. 11
3.   Fibrinolytic (clot lysis)                  Berg et al. 25
4.   Fibrinolytic (plate assay)                 Wolf26
5.   Fluorescence                               Bell et al. 27 Pochron et al. 28
6.   Spectrophotometric                         Smith et al. 29
*Many workers have since extended and modified this method particularly, and others in this
table. The references are intended to provide the source of the original or definitive method
Direct assays
Table 27.1 lists some direct assays with appropriate references, should details
of these be required. Since the level of plasminogen in plasma is reasonably
constant (approximately 0.16 mglml plasma), and since the normal level is high
compared with other components of haemostasis, methods of great sensitivity
(e.g. radioimmunoassay, latex flocculation and haemagglutination inhibition)
seem unnecessary. Thus the most practical and quantitative methods are those
which can be applied to most components of blood which are present in reason-
ably large amounts; namely, radial immunodiffusion using some modification
of the procedure of Mancini et al. 35 and the immunoelectrophoretic or 'rocket'
procedure36 . Since these are described in many textbooks it is necessary here
only to emphasize their simplicity and reproducibility. Obviously the only
problem in these assays is that denatured plasminogen and degraded forms of
plasminogen will be measured as biologically intact molecules. The user must
assess the clinical situation of the patient whose plasma is being assayed before
a judgement can be made concerning the suitability of these assays.
                                             248
                                          PLASMINOGEN ACTIVITY
                                                                           PL [85]
                50                    ..!!...       PL                   60         t
 "
                                                                                               II
                           I                                                   Is
            ,
                                                -
                           I
                     ~ •..:r-                                                   I 25 t         L
           L              8
                                                                                    GJ
                I.AnIVE                                                  ACTIVE
Figure 27.1 Schematic representation of the two-step activation of plasminogen followed by the
autodigestive step from active to inactive plasmin. This scheme was compiled from data reported
elsewhere32- 34• PLG, PLG-i PL and UK denote plasminogen, plasminogen intermediate, plasmin
and urokinase. The numbers in brackets and numbers above the polypeptide chains (denoted by
straight lines) are molecular weights x 103• Small arrows denote the approximate locations at which
urokinase cleaves plasminogen to form an intermediate and finally plasmin. Hand L denote the
heavy and light chains of active plasmin, while H' and L' are their digested equivalents in the
auto-digested inactive form of plasmin. A token number of disulphide bonds (S) is shown to
explain the total loss of only 5000 MW peptides during the activation of plasminogen, while the
molecular weights of the subunits show more significant changes. Step I shows the loss of 5000
MW peptide material during the formation of plasminogen intermediate, step 2 shows the cleavage
of an arginylvaIyl bond to form the two disulphide-bonded subunits of active plasmin, and plasmin
converts active plasmin to an inactive form by the cleavage of the light and heavy chains, most
probably at their C-terminal ends. (Taken from ref. 31).
   Fortunately one method which can be described as direct and yet measures
functional activity has been developed 37 • This depends on the fact that a
streptokinase-plasminogen (SK-plgn) complex can hydrolyse the presumed
plasmin-specific chromogenic substrates S-2251 (Chromogenix) and
Chromozym-PL (Pentapharm). Since this complex is only poorly inhibited by
PI and other plasma inhibitors it is possible to convert all the plasminogen in
plasma to the SK-plgn complex following a IO-min incubation period. The
complex, as measured by the hydrolysis of chromogenic substrate, is a direct
reflection of the level of plasminogen in the test plasma. This assay obviates
the problems of plasminogen activation referred to above, and has allowed
plasminogen assays to be conducted in plasma with some degree of confidence.
   Althoup a slight inhibition of the SK-plgn complex by excess SK has been
reported 3 , this does not affect the validity of assays for plasminogen when
comparing one plasma with another. Purified plasminogen preparations are
affected more by this inhibition, and this is probably the cause of the non-
parallelism found when plasma plasminogen is compared in a dose-response
manner with a purified glutamic acid-plasminogen (glu-plgn) standard (78/646)
                                                    249
                LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
(Figure 27.2). It was found 3 ! essential to maintain a constant plgn/SK ratio
during the dose-response analysis of test and standard preparations of
plasminogen (Figure 27.3). Thus, while recommending this procedure as the
only reliable direct plasma plasminogen assay, certain precautions must be
taken when calibrating the assay using the glu-plgn standard (78/646). Problems
have been reported when this type of assay is applied to patient plasma samples,
notably patients with disseminated intravascular coagulation (DIC) and elevated
fibrin degradation products39,40, and patients with elevated levels of fibrinogen
associated with ischaemic heart disease4 !. The fact that fibrino~en has been
shown to enhance the enzymatic activity of the SK-plgn complex 9,40 has been
used to obviate these effects and avoid an overestimation of plasminogen levels
in the plasma of patients with DIe and elevated levels of fibrinogen. The assay
to be described below was initially designed for the assay of plasminogen in
plasma using a calibrated standard for glu-plgn (78/646). Due to the effect of
fibrinogen and fibrin-degradation products on the activity of the SK-plgn
complex on chromogenic substrates we recommend that the purified plasminogen
standard should be used in the presence of 1.0 mglml of plasminogen-free
fibrinogen which is incorporated into buffer A below to give buffer B.
Furthermore, assay of plasminogen in plasma from patients undergoing
thrombolytic therapy is not recommended by this procedure. Under these
                      PLG GLUVSP
            1.00
0.70
   CD       0.40
   "'"a.0
    '"
   CD
  II:
   0>
   0
  ...J
            0.10
-0.20
)(
            -0.50   +----.----r-----,,----r----,----.---,
                    1.20      1.30     1.40   1.50      1.60     1.70       1.80       1.90
                                                 Log Dose
                     )( STD   + UNKl
Figure 27.2    The log/log dose-response curves of the SK-plgn complexes formed in two plasmas
(+, n) compared with that formed from purified glu-plgn (x). The response variable is the rate
(x 10-3 S-I) of optical density change of S-2251 at 405 nm31
                                               250
                                          PLASMINOGEN ACTIVITY
0.8
   E
   c::
    It)
              0.6
    ....
    0
    iii
    "'"
    '"Q;
    0.
    'c::co"
    0>        0.4
    .!:
    "
    "'cco"
    .0
    0
    '"
   .0         0.2
   ~
    0>
    0
   ....J
0.0
              -0.2   +---r----,---,------r---..,---r----,r-----,
                 1.20         1.30       1.40       1.50     1.60        1.70    1.80        1.90   2.00
                                                      Log (Vol of   PLGN/~I)
Equipment
Spectrophotometer (405 nm wavelength setting); semi-microcuvettes (1 ml);
centrifuge; water bath (37°C); stopwatch; disposable plastic test tubes.
  Extra equipment for initial rate method: photometer cuvette housing at
37°C; recorder.
Reagents
1. Plasminogenfreefibrinogen: freeze-dried reagent from IMCO. Reconstitute
  with distilled water to give a solution (2%) in 0.3 moUL NaCl.
2. Buffer A: 50 mmoVL Tris HCl-12 mmoVL NaCl. Add 6.1 g Tris and 0.7 g
   NaCI to distilled water; adjust to pH 7.4 at room temperature with 1 moUL
   HCl and make up to 1000 ml with distilled water. Stable for 2 months at
   2-6°C.
3. Buffer B: buffer A with plasminogen-free human fibrinogen added at 10.0
   mg/ml. Prepare fresh on day of use.
4. Chromogenic substrate:
   Alternatives:
   (a) H-D-Val-Leu-Lys-pNA, 2 HCl (S-2251)
   (b) Tos-Gly-Pro-Lys-pNA, 2 HCl (Chromozym - PL)
   Dissolve contents of manufacturer-supplied ampoule in sufficient distilled
   water to make a 3.0 mmoVL solution. This is stable at 0-6 °C for 6 months.
   Add 1 part of this solution to 8 parts buffer, mix well; this solution is
   subsequently referred to as the substrate and is 333 ~oUL. The substrate
   should be prepared on the day of assay and incubated at 37°C.
5. Activator (SK): streptokinase (Chromogenix) or Streptase from
   Dade-Behring is reconstituted with distilled water to give a concentration
   of 10 000 iulml.
6. Plasminogen standard: glutamic acid plasminogen (glu-plgn) standard (code
   78/646) is available from the National Institute for Biological Standards and
   Control, Potters Bar, Hertfordshire EN6 3QG, UK. It contains 10 units per
                                      252
                            PLASMINOGEN ACTIVITY
   ampoule and these units are the same as those defined by the second
   international reference preparation for plasmin. Reconstitute ampoule with
   4 mI of buffer. This solution will be referred to as the standard, and contains
   2.5 units per mI.
7. Acetic acid (500/0): this is used only in the end-point method.
Assay procedure
Establishment of a calibration curve using the standard and the assay of the
test plasma involves the same procedure; thus the construction of an assay for
each will be described simultaneously.
1. To 100 III of the standard and 100 J.1l of a 1:1 diluted buffer A test plasma
   add 400 J.1l of the activator (SK) solution and incubate at 37°C for 10 min.
2. Dilute the SK test plasma and SK standard 111, 112, 113 and 114 with buffers
   A and B respectively.
3. To 100 J.1l of each dilution of the SK standard and SK test plasma add
   900 J.1l of the chromogenic substrate solution and do this in a manner which
   is dependent on whether (a) continuous OD recording and initial rate is
   available or (b) the end-point procedure is followed:
   (a) Initial rate OD: use disposable plastic cuvettes and stagger the addition
        of substrate so that there is time to follow the OD at 405 nm for about
        1 min in a thermostatically controlled (37°C) spectrophotometer. Each
        dilution of SK standard and SK test plasma will give a different OD/
        min, and these data give a linear plot over a range of 0.2-2.0 units of
        plasminogen. The blank used in each case is 900 III of chromogenic
        substrate with 100 III of distilled water.
   (b) End-point: add chromogenic substrate to each dilution of SK standard
        and SK test plasma in disposable plastic test tubes and incubate for
        exactly 3 min before stopping each reaction mixture by the addition of
        100 J.1l of acetic acid (50%). Read the colour generated in each reaction
        mixture in a 1 mI plastic cuvette (light path = 1 em). Use a blank made
        up of 900 J.1l of substrate, 100 J.1l of distilled water and 100 III 50% acetic
        acid.
Note: buffer B is recommended in DIe plasma samples. However, it is
worthwhile checking the assay using buffer A only, since the cost of the
buffer B reagent is quite considerable.
Calculations
1. Plot the log OD against the log of the amount of plasminogen in units
   contained in each dilution of the standard measured. The graph is linear
   over a wide range (0.2-2.0 units/mI) (see Figure 27.3).
2. Each dilution of the test plasma generated an OD/min reading. The four
   separate values can be read from the calibration curve of the plasminogen
   standard. The average of these dilution-adjusted values can be used to
   express the plasminogen concentration in plasma as units of plasminogen/mI.
                                         253
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
References
 1. Christensen LR, McLeod MC. A proteolytic enzyme of serum. Characterisation, activation
    with inhibitors. J Gen Physiol 1949; 28: 559-83.
 2. Nilsson 1M, Skanse B, Gydell K. Fibrinolysis in Boeck's sarcoid. Acta Med Scand 1958; 159:
    463-70.
 3. Lassen M. The estimation of fibrinolytic components by means of the lysis time method.
    Scand J Clin Lab Invest 1958; 10: 384-89.
 4. Kowalski E, Kopec M, Niewiarowski S. An evaluation of the euglobulin method for the
    determination of fibrinolysis. J Clin Patho11959; 12: 215-18.
 5. Vignal A, Blatrix C, Steinbuch M. Fibrinogen free of plasminogen as substrate for fibrinolytic
    assays. Nature, Lond 1962; 193: 693.
 6. Remmert LF, Cohen P. Partial purification and properties of a proteolytic enzyme of human
    serum. J Bioi Chem 1949; 181: 431-48.
 7. Alkjaersig N, Fletcher AP, Sherry S. The mechanism of clot dissolution by plasmin. J Clin
    Invest 1959; 38: 1086-95.
 8. Derechin M. The assay of human plasminogen with casein as substrate. Biochen J 1961; 78:
    443-8.
 9. Hedner U, Nilsson 1M. Determination of plasminogen in human plasma by a casein method.
    Thromb Diathes Haemorrh 1965; 14: 545-61.
10. Johnson AJ, Kline DL, Alkjaersig N. Assay methods and standard preparations for plasmin,
    plasminogen and urokinase in purified systems, 1967-1968. Thromb Diathes Haemorrh 1969;
    21: 259-72.
II. Troll W, Sherry S, Wachman 1. The action of plasmin on synthetic substrates. J Bioi Chern
    1984; 208: 85-93.
12. Kline DL, Fishman JB. Plasmin. The humoral protease. Ann NY Acad Sci 1957; 68 : 25-36.
13. Lassen M. The esterase activity of the fibrinolytic system. Biochem J 1958; 69: 360--6.
14. Roberts PS. Measurement of rate of plasmin action on synthetic substrates. J Bioi Chem 1958;
    232: 285-91.
15. Heimburger N. Plasminogen assay. A review and evaluation of various methods. In: Davidson
    JF, Samana MM, Desnoyers PC, eds. Vol. 1. New York: Raven Press, 173-90.
16. Ganrot PO, Nilehn JE. Immunochemical determination of human plasminogen. Clin Chim
    Acta 1968; 22: 335-40.
17. Hedner U, Nilsson 1M. Comparison between a direct and indirect method for determining
    plasminogen. Thromb Diathes Haemorrh 1971; 26: 289-94.
18. Rabiner SF, Goldfine ID, Hart A. Radioimmunoassay of human plasminogen and plasmin.
    J Lab Clin Med 1969; 74: 265-73.
19. Wu KK, Jacobsen CD, Hoak JC. Highly sensitive method for the assay of plasminogen. J.
    Lab Clin Med 1973; 81: 484-8.
20. Wu KK, Jacobsen CD, Hosk JC. Assay of plasminogen using latex flocculation. Proc Soc
    Exp Bioi Med 1973; 144:391-3.
21. Zoltan RP, Mertz ET, Russell HT. Assay of human plasminogen in plasma by affinity
    chromatography. Clin Chem 1972; 18: 654-7.
22. Ludlam CA, Das Pc. Plasminogen assay by haemagglutination inhibition technique. J Clin
    Patho11971; 24: 136-42.
23. Storiko K. Normal values for 23 different human plasma proteins determined by single radial
    immunodiffusion. Blut 1968; 6: 200-8.
24. Friberger P, Knos M, Gustavsson S, Aurell L, Claeson G. Methods for determination of
    plasmins, antiplasmin and plasminogen by means of substrate S-2251. Haemostasis 1978; 7:
    138--45.
25. Berg W, Korsan-Bengtsen K, Y gge 1. Determination of plasminogen in human plasma by the
    lysis time method. Thromb Diathes Haemorrh 1966; 16: 1-17.
26. Wolf P. Modification of the fibrin plate for measurement of the components of the fibrinolytic
    system. Thromb Diathes Haemorrh 1968; 20: 50--65.
27. Bell P J, Dziobowski CT, Englert ME. A sensitive fluorometric assay for plasminogen, plasmin
    and streptokinase. Anal Biochem 1974; 61: 200-8.
28. Pochron SP, Mitchell GA, Albareda I, Huseby RM, Gargiulo RJ. A fluorescent substrate
    assay for plasminogen. Thromb Res 1978; 13: 733-9.
                                              254
                                 PLASMINOGEN ACTIVITY
29. Smith RE, Bissell ER, Mitchell AR, Pearson KW. Direct photometric or fiuorometric assay
    of proteineases using substrates containing 7-amino-4-trifiuoromethylcoumarin. Thromb Res
    1980; 17:392-402.
30. Brakman P. Fibrinolysis. A standardized fibrin plate method and a fibrinolytic assay of
    plasminogen. Amsterdam: Scheltema Holkema, 1967.
31. Gaffney PJ. Standardisation of plasminogen assays. Haemostasis 1988; (Suppl. 1): 47-60.
32. Rickli EE, Otavsky UI. Release of an N-terminal peptide from human plasminogen during
    activation with urokinase. Biochim Biophys Acta 1973; 295: 381-4.
33. Walther PI, Steinman HM, Hill RL, McKee PA. Activation of human plasminogen by
    urokinase. Partial characterization of a preactivation peptide. I BioI. Chem 1974; 249: 1173-81.
34. WIman B, Wallen P. Activation of human plasminogen by an insoluble derivative or urokinase.
    Eur J Biochem 1973; 36: 25-31.
35. Mancini G, Carbonara AO, Heremans IF. Immunochemical quantitation of antigens by single
    radial immunodiffusion. Immunochemistry 1965; 7: 261-4.
36. Laurell CB. Quantitative estimation of proteins by electrophoresis in agarose gel containing
    antibodies. Anal Biochem 1966; IS: 45-9.
37. Friberger P, Knos M. Plasminogen determination in human plasma. In: Chromogenic peptide
    substrates: chemistry and clinical usage. Edinburgh: Churchill Livingstone, 1979; 128-40.
38. Gaffney PJ, Philo RD. Measurement of some haemostatic components using biological and
    amidolytic assays. In: Davidson JF, Nilsson 1M, Asted B, eds. Progress in fibrinolysis, Vol 5.
    Edinburgh: Churchill Livingstone, 1981; 196-202.
39. Soria C, Soria J, Bertrand 0, Dunn F, Samama M, Bachmann F. The amidolytic activity of
    the SK-plasminogen complex is enhanced by a potentiator which is generated in the presence
    of vascular plasminogen activator - role of fibrin degradation products. Thromb Haemostas
    1982;47: 193-6.
40. Gram J, Jespersen J. A functional plasminogen assay utilising the potentiating effect of
    fibrinogen to correct for the overestimation of plasminogen in pathological plasma samples.
    Thromb Haemostas 1985; 53: 255-9.
41. Gram J, Munkvad S, Jespersen J. Elevated plasma concentrations of fibrinogen assay cause
    an overestimation of functional plasminogen. Thromb Haemostas 1989; 61: 154.
42. Gaffney PJ, Philo RD. A commentary on new methodology in haemostasis using chromogenic
    substrates. In: Fareed J, Mesmore HL, Fenton JW, Brinkhous KM, eds. Perspectives in
    haemostasis. New York: Pergamon Press, 1981; 405-17.
                                               255
28
Plasmin inhibitor activity (previously
u2-antiplasmin)
C. KLUFT and P. MEIJER
INTRODUCTION
PRINCIPLE
SOLUTIONS
1. Tris buffer: 40 mmoVL Tris-HCI buffer, pH 7.4 (at 25°C) containing 86
   mmoVL NaCI, 120 mmoVL methylamine and 0.008% (v/v) Tween 80.
                                      258
                        PLASMIN INHIBITOR ACTIVITY
ASSAY PROCEDURE
(PATHO)PHYSIOLOGY
In normal individuals, aged between 20 and 50 years, and sex ratio approximately
1, plasmin inhibitor activity assayed with the IPIT showed, in our laboratory,
a narrow range: 104 ± 11% (SD), n     =  1, range 85-139% without differences
between males and females. This compared well with experience with the SPIT
of 95 ± 7% (SD), n = 25, range 83-108%, and another estimation of the range
             =
(80-136% n 90f5. Very similar methods consistently show a narrow normal
range for plasmin inhibitor activity with a standard deviation of 9% in a
                          =                                 =
meta-anal~sis in 1982 (n 265)16 and with SD of 17% (n 50)26 and 15-17%
(n = 100)2 in some other reports. Variations with season28 and age28,29 may
exist.
   The lower limit of the normal range allows a clear distinction of heterozy~otes
for plasmin inhibitor deficiency from normals and normal family members 0-32.
The upper limit resembles that used by others (l20-140%? (see above).
                                       259
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   Plasmin inhibitor is an acute-phase reactant1,34,35, and can be found to be
elevated to about 130-140% of normal. Elevated plasmin inhibitor has been
observed in some cases with thrombotic complications25 ,33,36,37 and in cases
with type II hyperlipoproteinaemia38 and progressive renal failure 39 . Reduced
plasma levels of plasmin inhibitor activity can occur due to congenital
deficiencies type I and II40,41 and is considered 'an overlooked cause of haemor-
rhage,42. Acquired deficiencies are known for thrombolytic therapy, liver diseases,
nephrotic syndrome, disseminated intravascular coagulation (in relation to
antithrombin changes43), amyloidosis, leukaemia (especially acute promyelocytic
leukaemia26,44), L-asparaginase therapy45, the postoperative period35 and
extracorporeal circulation40 . In some cases the reduction might involve
inactivation by elastase 26,44,46.
PITFALLS
1. Quality ofplasmin: the rapid interaction between plasmin and the PB-plasmin
   inhibitor involves a second site interaction between the two molecules, which
   requires in plasmin an intact function of the site. As reported for some types
   of plasmin preparations47, plasmin can lose the functional integrity of this
   site, and lose its suitability for the assay. At present most, if not all,
   commercially available plasmin preparations are suitable, but outdated
   material may contain larger proportions of degraded plasmin. The presence
   of substantial amounts of degraded plasmin is observed as a deviation of
   the calibration curve at higher inhibition levels. It is chosen to work only in
   an area of 0-50010 inhibition of plasmin, and accordingly to further dilute
   samples with high activity.
2. Viscosity of the plasmin solution: it should be recognized that the plasmin
   solution has increased viscosity and its handling should be carefully con-
   trolled, as prescribed.
3. Spontaneous activity of plasma on the chromogenic substrate: in clinical
   samples such as after recent activation of fibrinolysis, plasmin captured by
   u2-macroglobulin may occur in plasma. Enzymes captured br u2-macro-
   globulin retain significant activity on chromogenic substrates4 . The spon-
   taneous activity of plasma can in such special circumstances be elevated,
   and should be subtracted from the recorded activities in the assay to avoid
   underestimation of plasmin inhibitor activity. The spontaneous activity can
   be easily assayed itself in the procedure described, replacing plasmin by
   buffer. Alternatively, the spontaneous activity can be irreversibly inhibited
   by the addition of small synthetic inhibitors (see point 4t9.
4. Activation in vitro: during and shortly after thrombolytic therapy, high levels
   of thrombolytics occur in collected plasma samples. Activation of
   plasminogen can continue in vitro, also at 0 °C, and can artificially reduce
   plasmin inhibitor activity5o. Additions to blood and plasma of PPACK51
   or GGACK52 for inactivation of t-PA and u-PA, respectively, do not disturb
   the assay of plasmin inhibitor activity53.
                                       260
                             PLASMIN INHIBITOR ACTIVITY
OTHER METHODS
Technical variations of the activity assay
Several different chromogenic substrates are in use 54 • Commercially available
methods include Coamatic® Plasmin Inhibitor, Spectrolyse® a2-antiplasmin,
Actichrome® a2-antiplasmin, Berichrome® arantiplasmin, a2-antiplasmin,
Dade, arantiplasmin Instrumentation Laboratory, Chromostrate
~-antiplasmin, Orthokrome Antiplasmin, Stachrom® Antiplasmin, and Unitest
a2-antiplasmin. Protocols for the use of these methods on several instruments
are available. For each performance it should have been documented and
checked that the method is specific, according to the consensus procedures24 •
   Immunochemical methods (radial and electroimmunodiffusion, enzyme
immunoassays) may assess more or all molecular forms of plasmin inhibitor.
It should be recognized that these methods may be complicated by differences
in titre of antisera for the various molecular forms, as shown for the PB and
NPB form 55 • Correlation between activity and antigen methods can accord-
ingly be quite good 56,57, but may also be poor7, depending upon the specificity
of the antibodies used.
Binding to plasminogen
Binding to plasminogen can be evaluated with plasma samples using a modified
crossed immunoelectrophoresis method7 •
References
 1. Wiman B. Human urantiplasmin. Methods Enzymo11981; 80: 395-408.
 2. Kluft C. ~-Antiplasmin activity. In: Jespersen J, Bertina RM, Haverkate F, eds. ECAT assay
    procedures: a manual of laboratory techniques. Dordrecht: Kluwer, 1992; 165-71.
 3. Blombiick M, Abildgaard U, van den Besselaar AM, Clementson K1, Dahlbiick B, Exner T,
    Francis cw, Gaffney P, Gralnick H, Hoyer LW et al. Nomenclature of quantities and units
    in thrombosis and haemostasis (recommendation 1993). A collaborative project of the Scientific
    and Standardization Committee of the International Society on Thrombosis and Haemostasis
    (ISTH/SSC) and the Commission/Committee on Quantities and Units (in Clinical Chemistry)
    of the International Union of Pure and Applied Chemistry-International Federation of
    Clinical Chemistry (IUPAC-IFCC/CQU(Cc». Thromb Haemostas 1994; 71: 375-94.
                                              261
           LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
 4. Collen D, Wiman B. Turnover of antiplasmin, the fast-acting plasmin inhibitor of plasma.
    Blood 1979; 53:313-24.
 5. Knot EAR, Drijfhout HR, Ten Cate JW, De Jong E, Iburg AHC, Kahle LH, Grijm R.
    u2-Plasmin inhibitor metabolism in patients with liver cirrhosis. J Lab Clin Med 1985; 105:
    353--61.
 6. Oemmensen I. Different molecular forms u2-antiplasmin. In Collen D, Wiman B, Verstraete
    M, eds. The physiological inhibitors of coagulation and fibrinolysis. Amsterdam: Elsevier!
    North-Holland, 1979; 131--6.
 7. Kluft C, Los P, Jie AFH, Van Hinsbergh VWM, Vellenga E, Jespersen J, Henny e. The mutual
    relationship between the two molecular forms of the major fibrinolysis inhibitor u 2-antiplasmin
    in blood. Blood 1986; 67: 616-22.
 8. Koyama T, Koike Y, Toyota S, Miyagi F, Suzuki N, Aoki N. Different NH2-terminal form
    with 12 additional residues of uz-plasmin inhibitor from human plasma and culture media
    of Hep G2 cells. Biochem Biophys Res Commun 1994; 200: 417-22.
 9. Bangert K, Johnsen AH, Christensen U, Thorsen S. Different N-terminal forms of uz-piasmin
    inhibitor in human plasma. Biochem J 1993: 291: 623-5.
10. Sumi Y, Ichikawa Y, Nakamura Y, Miura 0, Aoki N. Expression and characterization of pro
    uz-plasmin inhibitor. J Biochem 1989: 106: 703-7.
II. Wiman B, Collen D. On the kinetics of the reaction between human antiplasmin and plasmin.
    Eur J Biochem 1978; 84: 573-8.
12. Wiman B, Boman L, Collen D. On the kinetics of the reaction between human antiplasmin
    and a low-molecular-weight form of plasmin. Eur J Biochem 1978; 87: 143--6.
13. Petersen LC, Clemmensen I. Kinetics of plasmin inhibition in the presence of synthetic
    tripeptide substrate. Biochem J 1981; 199: 121-7.
14. Christensen U, Bangert K, Thorsen S. Reaction of human az-antiplasmin and plasmin.
    Stopped-flow fluorescence kinetics. FEBS Lett 1996; 387: 58---{j2.
15. Teger-Nilsson AC, Friberger P, Gyzander E. Determination of a new rapid plasmin inhibitor
    in human body blood by means of a plasmin specific tripeptide substrate. Scand J Clin Lab
    Invest 1997; 37: 403-9.
16. Friberger P. Chromogenic peptide substrates. Their use for the assay of factors in the fibrinolytic
    and the plasma kallikrein-kinin systems. Scand J Clin Lab Invest 1982; 42 (Suppl. 162): 41-7.
17. Gallimore MJ, Amundsen E, Aasen AO, Larsbraaten M, Lyngaas K, Svendsen L. Studies on
    plasma antiplasmin activity using a new plasmin specific chromogenic tripeptide substrate.
    Thromb Res 1979; 14: 51-60.
18. Naito K, Aoki N. Assay of u 2-plasmin inhibitor activity by means of a plasmin specific
    tripeptide substrate. Thromb Res 1978; 12: 1147-56.
19. Matsuda T, Ogawara M, Miura R, Seki T, Matsumoto T, Teramura Y, Nakamara K. Selective
    determination uz-plasmin inhibitor activity in plasma using chromogenic substrate. Thromb
    Res 1984;33:379-88.
20. Meijer P. Westerveld W, Huisman LGM. An improved method for the detection of
    u2-antiplasmin on the automated coagulation analyzer MLA Electra 1000e. Fibrinolysis
    1994; 8 (SuppI.2): 160-2.
21. Woodham B (Letter to the editor) and Meijer P (Reply). Fibrinolysis 1995; 9: 129-31.
22. Meijer P, Kluft C. Criteria for an automated specific plasmin inhibitor test (SPIT) in human
    plasma. Fibrinolysis 1996; 10 (Suppl. 2): 121-3.
23. Billing Clason S, Meijer P, Kluft C, Ersdal E. A novel method, Coamatic Plasmin Inhibitor,
    for the specific determination of plasmin inhibitor activity in plasma. Fibrinolysis 1996; 10
    (SuppI2): 165-7.
24. Meijer P, Hanss M, Christensen U, Wiman B, Kluft e. Criteria for the specific measurement
    of plasmin inhibitor activity. (Submitted).
25. Engesser L. Thrombophilia. Disorders of blood coagulation and fibrinolysis. Thesis, Leiden
    University, 1988.
26. Aoki N. Hemostasis associated with abnormalities of fibrinolysis. Blood Rev 1989; 3: 11-17.
27. Dick W, Cullmann W. Automatisierung eines neuen amidolytischen Verfahrens zur
    u z-Antiplasmin-Bestimmung im Plasma. Lab Med 1983; 7: 51-4.
28. Stout RW, Crawford VLS, McDermott MI, Rocks MJ, Morris TCM. Seasonal changes in
    haemostatic factors in young and elderly subjects. Age Ageing 1996; 25: 256-8.
29. Nowak-GOttl, Funk M, Mosch G,Wegerich B, Kornhuber B, Breddin HK. Univariate tolerance
                                                 262
                               PLASMIN INHIBITOR ACTIVITY
      regions for fibrinogen, antithrombin III, protein C, protein S, plasminogen and uz-antipiasmin
      in children using the new automated coagulation laboratory (ACL) method. Klin Padiatr
      1994; 206: 437-9.
30.   Kluft C, Vellenga E, Brommer EJP, Wijngaards G. A familial hemorrhagic diathesis in a
      Dutch family: an inherited deficiency of uz-antiplasmin. Blood 1982; 59: 1169-80.
31.   Miles LA, Plow EF, Donnelly KJ, Hougie C, Griffm IH. A bleeding disorder due to deficiency
      of uz-antiplasmin. Blood 1982; 59: 1246-51.
32.   Kluft C, Nieuwenhuis HK, Rijken DC, Groeneveld E, Wijngaards G, Van Berkel W,
      Dooijewaard G, Sixma JJ. uz-Antiplasrnin Enschede: dysfunctional uz-antiplasmin molecule
      associated with an autosomal recessive hemorrhagic disorder. J Clin Invest 1987; 80: 1391-400.
33.   Kluft C, Leebeek FWG. az-Antiplasmin and thrombosis: results of a questionnaire. Fibrinolysis
      1988: 2 (Suppl. 2): 47-8.
34.   Matsuda M, Wakabayashi K, Aoki N, Morioka Y: uz-Antiplasmin inhibitor is among acute-
      phase reactants. Thromb Res 1980; 17: 527-32.
35.   Kluft e. Fibrinolytic shut-down after surgery. In: Sawaya R, ed. Fibrinolysis and the central
      nervous system. Philadelphia, PA: Hanley & Belfus, 1990; 127-40.
36.   Brommer EJP, Gevers Leuven JA, Kluft C, Wijngaards G. Fibrinolytic inhibitor in type II
      hyperlipoproteinaemia. Lancet 1982; 1:1066.
37.   RatnoffOD. The role of haemostatic mechanisms. Clin Haematol1981; 10: 261-81.
38.   Lowe GDO, Stromberg P, Forbes CD, McArdle BM,Lorirner AR, Prentice CRM. Increased
      blood viscosity and fibrinolytic inhibitor in type II hyperlipoproteinaemia. Lancet 1982; I:
      472.
39.   Gordge MP, Faint RW, Rylance PB, Kluft C, Nield GH. Abnormal fibrinolysis in progressive
      renal failure due to a reduction in circulating tissue plasminogen activator. Br J Haematol
       1988; 69: 133.
40.   Saito H. uz-Plasmin inhibitor and its deficiency states. J Lab Clin Med 1988; 112: 671-8.
41.   Ikernatsu S, Fukutake K, Aoki N. Heterozygote for plasmin inhibitor deficiency developing
      hemorrhagic tendency with advancing age. Thromb Res 1996; 82: 129-36.
42.   Griffin GC, Mammen EF, Sokol RJ, Perrotta AL, Stoyanovich A, Abildgaard CF.
      uz-Antiplasmin deficiency. An overlooked cause of hemorrhage. Am J Pediatr Hematol Oncol
       1993; 15: 328-30.
43.   Okajima K. Clinical relevance of determination of plasma ATIII and uz-PI activities in
      patients with DIC. Application of the molecular markers for the analysis of pathophysiology
      of DIe. Jpn J Clin Patho11994; 42: 45-55.
44.   Avvisati G, Ten Cate Jw, Sturk A, Lamping RJ, Petti MC, Mandelli F. Acquired az-antip1asrnin
      deficiency in acute promyeloytic leukaemia. Br J Haematol1988; 70: 43-8.
45.   Vellenga E, Kluft C, Mulder NH, Wijngaards G, Nieweg HO. The influence of L-asparaginase
      therapy on the fibrinolytic system. Br J Haematol1984; 57: 247-54.
46.   Brower MS, Harpel PC. Proteolytic cleavage and inactivation of uz-plasmin inhibitor and
      CI-inactivator by human polymorphonuclear leukocyte elastase. J Bioi Chern 1982; 257:
      9849-54.
47.   Kluft C, Traas DW, Jie AFH, Hoegee-de Nobel E. The suitability of various plasmin prepara-
      tions for the functional assay of uz-antiplasmin in plasma. Thromb Haemostas 1982; 48:
      320-4.
48.   Gyzander E, Teger-Nilsson AC. Activity of the uz-macroglobulin-plasrnin complex on the
      plasmin specific substrate H-o-Val-Leu-Lys-p-nitroanilide. Thromb Res 1980; 19: 165-75.
49.   Gram J, Jespersen 1. On the significance of antithrombin III, uz-rnacroglobulin, az-antiplasrnin,
      histidine-rich glycoprotein, and protein C in patients with acute myocardial infarction and
      deep vein thrombosis. Thromb Haemostas 1985; 54: 503-5.
50.   Rijken DC, Seifried E, Barrett-Bergshoeff MM, Dooijewaard G. Plasminogen activation at
      low temperatures in plasma samples containing therapeutic concentrations of tissue-type
      plasminogen activator or other thrombolytic agents. Thromb Haemostas 1990; 64: 47-52.
51.   Seifried E, Tanswell P. Comparison of specific antibody, D-Phe-Pro-Arg-CHz CI and aprotinin
      for prevention of in vitro effects of recombinant tissue-type plasminogen activator on
      haemostasis parameters. Thromb Haemostas 1987; 58: 921--{).
52.   Lijnen HR, Van Hoef B, Collen D. Differential reactivity of Glu-Gly-Arg-CHz CI, a synthetic
      urokinase inhibitor, with single-chain and two-chain forms of urokinase-type plasminogen
      activator. Eur J Biochem 1987; 162: 351--{).
                                                 263
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
53. Oethinger MD, Seifried E. In vitro effects of urokinase - prevention by different inhibitors.
    Thromb Haemostas 1990; 64: 402-6.
54. Kluft C, Jie AFH, Los P, Dooijewaard G, Traas DW. Expression of the two forms of
    urantiplasmin in functional and immunochemical assays. In: Davidson JF, Bachmann F,
    Bouvier CA, Kruithof EKO, eds. Progress in fibrinolysis, vol VI. Edinburgh: Churchill
    Livingstone, 1983: 386--7.
55. Svendsen LG, Fareed J, Walenga JM, Hoppensteadt D. Newer synthetic peptide substrates
    in coagulation testing: some practical considerations for automated methods. Semin Thromb
    Hemostas 1983; 9: 250-62.
56. Matsuda T, Ogawara M, Miura R, Seki T, Matsumoto T, Teramura Y, Nakamara K. Selective
    determination of ~-p1asmin inhibitor activity in plasma using chromogenic substrate. Thromb
    Res. 1984; 33:379-88.
57. Aoki N, Yamanaka T. The u2-plasmin inhibitor levels in liver diseases. Clin Chim Acta 1978;
    84: 99-105.
58. Sakata Y, Aoki N. Cross-linking of ~-plasmin inhibitor to fibrin by fibrin-stabilizing factor.
    J Clin Invest 1980; 65: 290-7.
59. Kluft C, Los P, Jie AF. The molecular form of u2-antiplasmin with affmity for plasminogen
    is selectively bound to fibrin by factor XIII. Thromb Res 1984; 33: 419-25.
                                              264
29
Plasmin-a2 -antiplasmin complexes
(plasmin-plasmin inhibitor
complexes)
E. HATTEY, M. HAUMER, M. R. GRIFFITHS, V. CARROLL and
B. R. BINDER
INTRODUCTION
METHODS OF ASSAY
For the detection of PPI complexes in plasma most investigators use a com-
bination of antibodies against the two complex-forming proteins in a sandwich
system, with either radioactive or enzymatic labelling. There are currently
several ELISA systems commercially available (Dade-Behring, Technoclone,
Teijin). In addition several investigators use their own in-house test systems,
e.g. RIA34 or ELlSA35 in the human system, and for studies in animal models36 .
Thereby the use of different antibodies, different standards, varying test condi-
tions (incubation temperature, addition of inhibitors) has led to discrepancies
in the literature with respect to the normal range values (Table 29.1). Despite
the great difference between some of the reported baseline values Meijer et al.
found a significant correlation when the same samples were tested with the
three commercially available assay systems37 . It is important to note that the
mean baseline values obtained with these assays (Technoclone: 142 ng/ml,
Dade-Behring: 368 ng/ml, Teijin: 758 ng/ml) correspond with the respective
normal values published for these assays, and reflect the same baseline fibrinolytic
activities. The difference in the standards used, and perhaps in some cases even
the small sample size, seem to account for the variation in the reported basal
values for healthy donors. However, a systematic difference might exist between
a number of publications reporting rather lower mean baseline levels and others
with rather higher baseline levels. In the latter cases higher sample dilutions
(1:50-1:800) were used, thereby shifting the equilibrium between plasminogen
activators and inhibitors in plasma towards plasminogen activation ('diluting
out inhibitors'). If such diluted samples are then incubated for a prolonged
time (1 h or more) at 37°C during the assay procedure increased PPI formation
might occur ex vivo. We therefore recommend addition of inhibitors to the
sample dilution buffer and incubation at lower temperature, or only for a short
time. The avoidance of PPI formation ex vivo is especially important when the
TATIPPI ratio is determined.
                                        266
      Table 29.1   Reference values reported for various PPI kits
      Company name        Reported values          Range            Sample     Fibrinolysis inhibitors   Incubation       Sample     Reforence
                                                                                                                                                          "tJ
                                                                    dilution                                              size (n)
                                                                                                                                                          ):
                                                                                                                                                          C/J
      Commercial Assays                                                                                                                                   :s:::
      Technoclone         l31 nglml (median)       10-514 nglml     1:10       Aprotinin 2000 KIU/mI,    4°C overnight     87        Carroll et al. 41    Z
                                                                               benzamidine 20 mmollL                                                      ~
                                                                                                                                                         II)
      Technoclone         142 nglml (mean)                          1:10       As above                  As above         113        Meijer et al. 37     ):.
                                                                                                                                                          Z
      Dade-Behring        210 ± 88 (mean)          80-470 nglml     1:2        Aprotinin 10 KIU/mi       15 min at 37°C   178        Pelzer et al. 42     --I
I\)
                                                                                                                                                          :;;
!?l   Dade-Behring        290 nglml (median)       120-700 nglml    1:2        None                      15 min at 37°C   466        Company
                                                                                                                                     instructions         ~:s:::
      Teijin              758 nglml (mean)                          1:800      None?                     1 h at 37°C      113        Meijer et al. 37     Z
                                                                                                                                                          ()
      Non-commercial assays                                                                                                                               0
                                                                                                                                                          :s:::
      RIA                 3.5 nmollL (mean)        2-8nmollL        1.5        SBTI 100 Ilglml,          4hatRT*           15        Levi et   al. 34     "tJ
                                                                                                                                                          I
                          (-490 nglml)             (280-1120 nglml)            benzamidine 10 mmoVL                                                       m
                                                                                                                                                          X
      ELISA               573 ± 131 nglml          350-790 nglml    1:50       EACAO.l mollL             1 h at 37°C       30        Montes et al. 35     m
                          (mean)                                                                                                                          C/J
   As mentioned above, there are several test kits commercially available for
the determination of PPI complexes in plasma. For those who wish to establish
their own test system the following procedure is described:
Test principle
The test described here is a solid-phase enzyme immunoassay in which MPW7AP,
a specific monoclonal antibody directed against the neoantigen of the PPI
complex, is adsorbed on plastic microtitre plates38,39. During incubation with
test samples PPI complexes are selectively bound, and after washing away
unbound material the complexes are detected by MPW2PG POX, a peroxidase-
labelled monoclonal antibody against the kringle 1-3 region of the plasmin(ogen)
part of the complex40 • Quantification of labelled antigen-antibody conjugates
is achieved by ABTS, a chromogenic substrate for peroxidase.
Solutions
 I. Coating buffer: 1.59 g Na2C03.1 OH20, 2.93 g NaHC03, 100 mg thimerosal
    with distilled water to I L, pH 9.6.
 2. Coating solution: 20 Ilg/ml MPW7AP in coating buffer; 100 j.tlIwell.
 3. Phosphate buffered saline (PBS): 8 g NaCI, 0.2 g KH 2P0 4 , 1.44 g
    Na2HP042H20 with distilled water to I L, pH 7.4.
 4. Washing buffer: PBS containing 0.5% Tween 20.
 5. Blocking solution: PBS containing 1% BSA.
 6. Dilution buffer I (DBI): PBS containing 1%(w/v) BSA, 2000 KIV/ml
    aprotinin and 20 mmol IL benzamidine chloride or a specific inhibitor for
    t-PA, e.g. PPACK.
                                      268
                   PLASMIN-a2-ANTIPLASMIN COMPLEXES
 7. Dilution buffer 2 (DB2): OBI containing 1% PPI depleted plasma.
 8. Dilution buffer 3 (DB3): OBI containing 10% PPI depleted plasma.
 9. Detecting solution: 10 Ilg/ml MPW2PG POX in OBI; 100 Ill/well.
10. Substrate buffer: 1.29 g citric acid monohydrate, 1.375 g Na2HP042H20
    with distilled water to 100 ml, pH 4.0.
11. Substrate solution: 1 mg ABTS/ml solution containing 1 III H 20 2 30%/ml
    solution in substrate buffer; 100 Ill/well.
12. Stop solution: 320 mg NaFIlOO ml distilled water; 100 Ill/well.
Procedure
Preparation of plates
The wells of an ELISA plate are filled with 100 Ill/well coating solution,
preferably by use of a multichannel micropipette. The plate should remain
covered with a self-adhesive plastic foil for at least 16 h at 4 °C but can also be
stored that way for a prolonged period. Before use, the plate is emptied and
refilled with 100 Jll/well of blocking solution and incubated for 1 hat 37°C to
block excessive reactive groups on the plate surface. For all incubation steps
the plate remains covered with the plastic foil.
Sample preparation
Standard preparation
Evaluation of results
For evaluation of the results the reading of the samples is compared with those
of a reference PPI complex preparation.
Standardization
Since PPI complexes are not easily prepared in a stable purified form - the
complex is susceptible to proteolytic degradation and different epitopes might
be generated whether the complex is formed in excess of plasmin or of inhibitor l
- we decided to prepare PPI complexes directly in plasma and to calibrate them
with PPI complexes generated from purified components. For this purpose
citrated plasma was incubated with more than saturating concentrations of
urokinase to produce maximum amount of PPI complexes. Thereafter the
reaction was terminated by addition of benzamidine and Trasylol. The complexes
so formed are stable either frozen at -70°C or lyophilized at 4 0C. To determine
the actual amount of complexes in the standard plasma the readings in the
ELISA assay were compared with those of PPI complexes generated from
purified components. For this purpose plasmin was prepared freshly from
purified gIu-plasminogen by incubation with urokinase bound to Sepharose
                                      270
                     PLASMIN-a2 -ANTIPLASMIN COMPLEXES
Specificity
The described test is specific for PPI complexes and allows full plasma recovery,
provided the standard was diluted in PPI-depleted plasma in the same con-
centration as the plasma samples to be tested.
2.0
1.8
1.6
1.4
1.2
                                                         Plasmin    Plasmin
                   0.6                                       +          +
                                                         Buffer Plasmin inhibitor
                   0.4
                                         ...   PPI-Complex prepared in Plasma
                   0.2                   •     PPI-Complexes from purified
                                               Componen1s
                                       0.1             0.2
                                       IAQhnl Complexes
Figure 29.1 Standard curves obtained with the described test. Comparison of PPI complexes
prepared in plasma with PPI complexes from purified components. In the insert the arnidolytic
determination of plasmin activity from freshly prepared plasmin incubated with or without excess
of u2"antiplasmin is shown. From the differences in plasmin activity (plasmin inhibited) the
concentration of formed complexes is calculated
                                               271
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
p<O.05
                             •
                             •                          •
                             •                          •
                                                                              ••
          ~
                             ••                         •                      •
                             I·•                                             ..
         is.
          E                                          • I                     ••
          8                                          •                        •
                         •                       ••• •                 -   • •
                                                                           •• •••
                                               -I!•••       I,
               0
                    -HIP.1
                       II
                        Mlmale
                                                 ••
                                                            Ii
                                                            :.
                                                  Normal male
                                                                             ••
                                                                            PTCA
Figure 29.2 PPI complex values in male survivors of myocardial infarction and in matched
controls. In comparison values from patients 12 months after successful PTCA are shown (median
values indicated)
                                               272
                     PLASMIN-a 2 -ANTIPLASMIN COMPLEXES
correlate in principle with each other. The differences in the levels of PPI
complexes might, however, be due to different standards used, as well as to
PPI complex formation ex vivo.
References
 1. Wiman B. Human alpha-2 antiplasmin. Methods Enzymo11981; 80: 395-408.
 2. Sakata Y, Aoki N. Significance of cross-linking of alpha 2-plasmin inhibitor to fibrin in
    inhibition of fibrinolysis and in hemostasis. J Clin Invest 1982; 69: 536-42.
 3. Aoki N, Harpel PC. Inhibitors of the fibrinolytic enzyme system. Semin Thromb Hemostas
    1984;10: 24--41.
 4. Hoylaerts M, Rijken DC, Lijnen HR, Collen D. Kinetics of the activation of plasminogen
    by human tissue plasminogen activator. Role of fibrin. J BioI Chern 1982; 257: 2912-19.
 5. Beckmann R, Geiger M, Binder BR. Plasminogen activation by tissue plasminogen activator
    in the presence of stimulating CNBr fragment FCB-2 of fibrinogen is a two-phase reaction.
    Kinetic analysis of the initial phase of slow plasmin formation. J BioI Chern 1988; 263:
    7176--80.
 6. Urano T, Kamiya T, Sakaguchi S, Takada Y, Takada A. Fibrinogenolysis and fibrinolysis in
    normal volunteers and patients with thrombosis after infusion of urokinase. Thromb Res
    1985; 39: 145-55.
 7. Levi M, Roem D, Kamp AM, de Boer JP, Hack CE, ten Cate JW. Assessment of the relative
    contribution of different protease inhibitors to the inhibition of plasmin in vivo. Thromb
    Haemostas 1993; 69: 141-6.
 8. Gabazza EC, Taguchi 0, Yamakami T, Machishi M, Ibata H, Suzuki S. Evaluating
    prethrombotic state in lung cancer using molecular markers. Chest 1993; 103: 196-200.
 9. Okajima K, Kohno I, Soe G, Okabe H, Takatsuki K, Binder BR. Direct evidence for systemic
    fibrinogenolysis in patients with acquired alpha 2-plasmin inhibitor deficiency. Am J Hematol
    1994; 45: 16-24.
10. Gabazza EC, Taguchi 0, Yamakami T et al. Alteration of coagulation and fibrinolysis systems
    after multidrug anticancer therapy for lung cancer. Eur J Cancer 1994; 30A: 1276-81.
11. Nanninga PB, van Teunenbroek A, Veenhof CH, Buller HR, ten Cate JW. Low prevalence
    of coagulation and fibrinolytic activation in patients with primary untreated cancer. Thromb
    Haemostas 1990; 64: 361-4.
12. Mari D, Mannucci PM, Coppola R, Bottasso B, Bauer KA, Rosenberg RD. Hypercoagulability
    in centenarians: the paradox of successful aging. Blood 1995; 85: 3144-9.
13. Uenomachi H, Sonoda M, Miyauchi T et al. Relationship between intracoronary thrombolysis
    and fibrino-coagulation - special reference to TATIPIC and FPAlPIC. Jpn Circ J 1996; 60:
    149-56.
14. Dietrich W Reducing thrombin formation during cardiopulmonary bypass: is there a benefit
    of the additional anticoagulant action of aprotinin? J Cardiovasc Pharmacol1996; 27 (Suppl.
    1): S50-7.
15. Kawasuji M, Ueyama K, Sakakibara N et al. Effect of low-dose aprotinin on coagulation
    and fibrinolysis in cardiopulmonary bypass. Ann Thorac Surg 1993; 55: 1205-9.
16. Malyszko J, Malyszko JS, Pawlak D, Pawlak K, Buczko W, Mysliwiec M. Hemostasis, platelet
    function and serotonin in acute and chronic renal failure. Thromb Res 1996; 83: 351-61.
17. Jansen PM, Boermeester MA, Fischer E et al. Contribution of interleukin-l to activation of
    coagulation and fibrinolysis, neutrophil degranulation, and the release of secretory-type
    phospholipase A2 in sepsis: studies in nonhuman primates after interleukin-l alpha
    administration and during lethal bacteremia. Blood 1995; 86: 1027-34.
18. Pedersen OD, Gram J, Jespersen 1. Plasminogen activator inhibitor type-I determines plasmin
    formation in patients with ischaemic heart disease. Thromb Haemostas 1995; 73: 835-40.
19. Agnelli G, Iorio A, Parise P, Goldhaber SZ, Levine MN. Fibrinogenolysis and thrombin
    generation after reduced dose bolus or conventional rtPA for pulmonary embolism. Blood
    Coagul Fibrinolysis 1997; 8: 216-22.
20. Nielsen EW, Johansen HT, Hogasen K, Wuillemin W, Hack CE, Mollnes TE. Activation of
    the complement, coagulation, fibrinolytic and kallikrein-kinin systems during attacks of
    hereditary angioedema. Scand J Immunol1996; 44: 185-92.
                                              273
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
21. Handa K, Takao M, Nomoto J et al. Evaluation of the coagulation and fibrinolytic systems
    in men with intermittent claudication. Angiology 1996; 47: 543-8.
22. Handa K, Kimoto K, Kawaguchi H et al. Plasmin and thrombin inhibitors in essentially
    untreated patients with coronary artery spasm. Int J Cardiol 1993; 42: 263-7.
23. Cugno M, Hack CE, de Boer JP, Eerenberg AJ, Agostoni A, Cicardi M. Generation of plasmin
    during acute attacks of hereditary angioedema. J Lab Clin Med 1993; 121: 38-43.
24. Quehenberger RP, Loner U, Kapiotis S et al. Effects of the third generation oral contracep-
    tives containing newly developed progestagens on fibrinolytic parameters. Fibrinol Proteol
    1997; 11: 97-101.
25. Quehenberger P, Kapiotis S, Partan C et al. Studies on oral contraceptive-induced changes
    in blood coagulation and fibrinolysis and the estrogen effect on endothelial cells. Ann Hematol
    1993; 67: 33-6.
26. Takahashi H, Tatewaki W, Wada K, Hanano M, Shibata A. Thrombin vs. plasmin generation
    in disseminated intravascular coagulation associated with various underlying disorders [see
    comments]. Am J Hemato11990; 33: 90-5.
27. Kario K, Matsuo T. Lipid-related hemostatic abnormalities in the elderly: imbalance between
    coagulation and fibrinolysis. Atherosclerosis 1993; 103: 131-8.
28. Kario K, Matsuo T, Kodama K, Matsuo M, Yamamoto K, Kobayashi H. Imbalance between
    thrombin and plasmin activity in disseminated intravascular coagulation. Assessment by the
    thrombin-antithrombin-III complexlplasmin-alpha-2-antiplasmin complex ratio. Haemostasis
    1992; 22: 179-86.
29. Merryman P, Tannenbaum SH, Gralnick HR et al. Fibrinolytic and coagulant responses to
    regional limb perfusions of tumor necrosis factor, interferon-gamma, andlor melphalan.
    Thromb Haemostas 1997; 77: 53-6.
30. Vanderpoll T, Coyle SM, Levi M et al. Effect of a recombinant dimeric tumor necrosis factor
    receptor on inflammatory responses to intravenous endotoxin in normal humans. Blood 1997;
    89: 3727-34.
31. Pajkrt D, Vanderpoll T, Levi M et al. Interleukin 10 inhibits activation of coagulation and
    fibrinolysis during endotoxinemia. Blood 1997; 89: 2701-5.
32. Corssmit EP, Levi M, Hack CE, ten Cate Jw, Sauerwein HP, Romijn JA. Fibrinolytic response
    to interferon-alpha in healthy human subjects. Thromb Haemostas 1996; 75: 113-17.
33. van Deventer SJ, Buller HR, ten Cate JW, Aarden LA, Hack CE, Sturk A. Experimental
    endotoxemia in humans: analysis of cytokine release and coagulation, fibrinolytic, and
    complement pathways. Blood 1990; 76: 2520-6.
34. Levi M, de Boer JP, Roem D, ten Cate Jw, Hack CEo Plasminogen activation in vivo upon
    intravenous infusion of DDAVP. Quantitative assessment of plasmin-alpha 2-antiplasmin
    complex with a novel monoclonal antibody based radioimmunoassay. Thromb Haemostas
    1992; 67: 111-16.
35. Montes R, Paramo JA, Angles Cano E, Rocha E. Development and clinical application of a
    new ELISA assay to determine plasmin-alpha 2-antiplasmin complexes in plasma. Br J
    Haematol1996; 92: 979-85.
36. de Boer JP, Creasy AA, Chang A et al. Activation patterns of coagulation and fibrinolysis in
    baboons following infusion with lethal or sublethal dose of Escherichia coli. Circ Shock 1993;
    39: 59-67.
37. Meijer P, Kamerling SWA, van de Ham FJ et al. Baseline levels of alpha-2-antiplasmin-
    plasmin complex in human plasma. Fibrinolysis 1994; 8 (Suppl. 2): 124-5.
38. Hattey E, Wojta J, Huber K, Binder BR. Development and evaluation of ELISA systems for
    determination of plasmin-alpha-2-antiplasmin (PIP) complexes in plasma. Fibrinolysis 1986;
    (Suppl.) 1: A152.
39. Hattey E, Beckmann R, Krutisch G, Huber K. Evaluation of limited activation of the plasma
    fibrinolytic system during thrombolysis by rt-PA. Fibrinolysis 1988; 2 (Suppl.l): 2.
40. Hattey E, Wojta J, Binder BR. Monoclonal antibodies against plasminogen and alpha-2-
    antiplasmin: binding to native and modified antigens. Thromb Res 1987; 45 : 485-95.
41. Carroll VA, Griffiths MR, Geiger Met al. Plasma protein C inhibitor is elevated in survivors
    of myocardial infarction. Arterioscler Thromb Vasc Bio11997; 17: 114-18.
42. Pelzer H, Pilgrim A, Schwarz A, Merte D, Keuper H, Hock H. Determination of alpha2
    antiplasmin complex in human plasma with an enzyme-linked immunosorbent assay.
    Fibrinolysis 1993; 7: 69-74.
                                               274
30
Soluble fibrin and degradation
products of fibrinogen (FgOP), fibrin
(FbOP; O-dimer) and total of FgOP
and FbOP (TOP)
W. NIEUWENHUIZEN and R. BOS
INTRODUCTION
SOLUBLE FIBRIN
D-dlmer
Eventually, these X-oligomers can be digested to fragments D-dimer, i.e. two
covalently bound D-domains 1,2, and fragments E. It should be emphasized
that so-called D-dimer assays detect not only D-dimer, but also all fibrin
                                       276
                     SOLUBLE FIBRIN AND FIBRINOGEN
degradation products comprising one or more D-dimer motifs (e.g. X-oligomers).
D-dimer as such rarely occurs in patient plasmas.
Type of assay
With the advent of monoclonal antibody (mAb) technology it became possible
to develop assays that can be performed with plasma samples, thus avoiding
the serum artifacts mentioned above.
   This chapter includes new assays based on mAb which determine in plasma
samples s~ecifically soluble fibrin21, degradation products of fibrinogen
(FgDP)I4-' 6, degradation products of fibrin I4,IS,17 (FbDP including D-dimer)
and the total of FgDP plus FbDP, designated as TDp 10,14,IS.
   For soluble fibrin and D-dimer an increasing number of test kits has become
commercially available, including qualitative and semi-quantitative latex slide
tests, and quantitative assays (EIA and latex immunoassays, LIA). In tables
                                        277
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
30.3 and 30.4 commercially available kits of soluble fibrin and D-dimer are
listed which have been frequently referred to in the literature up to now.
Note: The Chromogenix assay 'Coatest Soluble Fibrin' is based on the ability
of fibrin to potentiate the t-PA - catalysed conversion of plasminogen to
plasmin, which is analysed using a chromogenic substrate (S-2403). Other
assays detect antigen levels.
   The details of how to perform the tests mentioned above can be found in
the kits inserts. As an example we describe below in more detail the quantitative,
sandwich-type enzyme immuno-assays (EIA) manufactured by Organon Teknika
and sold under the names Fibrinostika Soluble Fibrin, Fibrinostika FgDP,
Fibrinostika FbDP and Fibrinostika TDP (Table 30.1).
Flbrlnostika FgDP
The capture antibody in this EIA is a mAb (FDP-14) specific for degraded
forms of both fibrinogen and fibrin, i.e. it does not react with intact fibrin and
Table 30.1 EIA for soluble fibrin and degradation products of fibrinogen and fibrin, and the
total of the latter two, manufactured by Organon Teknika as Fibrinostika Soluble Fibrin, FgDP,
FbDP and TDP, respectively
                                             278
                     SOLUBLE FIBRIN AND FIBRINOGEN
fibrinogen 14 • When the test sample is incubated in the well of a microtitre plate
coated with FDP-14 both FgDP and FbDP will be bound. The tagging antibody
(YI8) in this EIA is specific for fibrinopeptide A-containing material 15; it does
not react with free fibrinopeptide A.
   On the basis of the specificities of FDP-14 and Yl8 the assa~ is specific for
degradation products comprising fibrinopeptide A, i.e. FgDp 1 •
   Analysis of 42 healthy individuals in an outside laboratory showed an upper
                                        =
limit for FgDP of 250 ng FE/ml (FE fibrinogen equivalent unit). This is an
indication only, and each laboratory should determine its own reference range.
Fibrinostika FbOP
The capture of mAb of this EIA is FDP-14, i.e. the same as in Fibrinostika
FgDP. However, the tagging mAb is specific for FbDP. It is important to note
that it does not discriminate between crosslinked (including D-dimer) and
non-crosslinked FbDP. In that respect it is not fully comparable with the existing
EIA for D-dimer. The combination of the specificities of the capture and
tagging antibodies in Fibrinostika FbDP make this EIA specific for FbDp 17•
   Analysis of 42 healthy individuals in an outside laboratory showed an upper
limit for FbDP of 310 ng FE/mI. This is an indication only, and each laboratory
should determine its own reference range.
Fibrinostika TOP
The capture antibody, in this EIA also, is FDP-14. Thus, in the first step of
this EIA both FgDP and FbDP are bound. The tagging antibody consists of
a mixture of mAbs used in Fibrinostika FgDP and Fibrinostika FbDP. This
means that in Fibrinostika TDP both FgDP and FbDP (= TDP) are detected.
The EIA has been published in a version in which polyc1onal tagging antibodies
were used (10).
   The reproducibilities are given in Table 30.2. Analysis of 42 healthy individuals
in an outside laboratory showed an upper limit for TDP of 650 ng FE/mt. This
is an indication only, and each laboratory should determine its own reference
range.
PATHOPHYSIOLOGY
Soluble Fibrin
D-dlmer
D-dimer assays from different sources have been compared with respect to
efficiency, and clinical characteristics27- 33 • A wide range of sensitivities and
specificities has been reported because of inter- and intra-assay variations and
differences in patient populations32 • Latex agglutination assays seem to be
relatively insensitive as screening tests28•32 • ELISA methods have previously
been proven to be reliable27 in the exclusion of thrombosis as occurring in
pulmonary embolism. The latest rapid methods have also been claimed to
                                            280
                         SOLUBLE FIBRIN AND FIBRINOGEN
Table 30.4 Commercially available assays for D-dimer
BioMerieux        FDPSlidex                    x
                  Vidas D-dimer                                                    x
Biopool           TintElize D-dimer                         x
                  Minute D-dimer               x
                  Nephelotex D-dimer                                        x
Boehringer        Tinaquant D-Dimer"                                        x
Mannheim
Dade-Behring      Enzygnost D-dimer                         x
                  BCD-dimer                                                 x
                  Turbiquant D-dimerd                                                     x
                  Data-Fi Dimertest            x
Diagnostica-      D-di Test                    x
Stago
                  Asserachrom D-dimer                       x
                  STA Liatest D-di                                          x
                  Instant I.A.                        x
Hemoliance        Ortho Dimertest              x
Iatron            LPIA ACE D-dimer                                          x
                  IATRO DDIE Test              x
IL                IL D-dimer Test                                           x
Nycomed           Nycocard D-dimer                    x
Organon           Fibrinosticon                x
Teknika
                  Fibrinostika FbDP*                        x
Sigma             Accuclot D-dimer             x
• Products from Agen are distributed by a variety of companies including American Diagnostica,
  Ortho, Dade-Behring, Fujirebio and Chromogenix.
b SimpliRED from Agen is based on agglutination of the patients' own red cells.
C Tinaquant is used by Boehringer Mannheim for application on the Hitachi analysers; in addition
  Boehringer Mannheim is the distributor of Stago's Liatest, manual latex and ELISA.
d Turbiquant is an automatic turbidimetric test for Behring's Turbitimer system.
                  =         =                       =                              =
Abbreviations: LX late; IF immune filtration; EIA enzyme immunoassay; MLX microlatex.
• Assay also measures non-crosslinked FbDP
References
 1. Doolittle RF. Fibrinogen and fibrin. Sci Am 1981; 245: 92-101.
 2. GraeffH, Hafter R. Detection and relevance of cross-linked fibrin derivatives in blood. Semin
    Thromb Haemostas 1982; 8: 57-68.
                                              282
                         SOLUBLE FIBRIN AND FIBRINOGEN
 3. Gaffney PJ, Perry MJ. 'Giant' fibrin fragments and thrombosis. Thromb Haemost 1985; 54:
    931 (abstract).
 4. Gaffney PJ, Creighton LC, Harris R, Perry MJ. Monoclonal antibodies (MABS) to crosslinked
    fibrin fragments. Their characterization and potential clinical use. In: Miiller-Berghaus G. et
    ai., eds. Fibrinogen and its derivatives: biochemistry, physiology and pathophysiology.
    Amsterdam: Excerpta Medica, 1986; 273-84.
 5. Merskey C, Lalezazi P, Johnson Al A rapid simple sensitive method for measuring fibrinolytic
    split products in human serum. Proc Soc Exp BioI Med NY 1969; 131: 871-5.
 6. Connaghan DG, Francis DW, Ryan DH, Marder VI Prevalence and clinical implications of
    heparin-associated false positive tests for serum fibrin(ogen) degradation products. Am J Clin
    Patho11986; 86: 304-10.
 7. Haverkate F, Timan G, Nieuwenhuizen W Anticlotting properties of fragments D from human
    fibrinogen and fibrin. Eur J Clin Invest 1979; 9: 253-5.
 8. Nieuwenhuizen W, Gravesen M. Anticoagulant and calcium-binding properties of high
    molecular weight derivatives of human fibrinogen, produced by plasmin (fragments X).
    Biochim Biophys Acta 1981; 668: 81-8.
 9. Nieuwenhuizen W, Voskuilen M, Hermans 1. Anticoagulant and calcium-binding properties
    of high molecular weight derivatives of human fibrinogen (plasmin fragments Y). Biochim
    Biophys Acta 1982; 708: 313-16.
10. Koopman J, Haverkate F, Koppert PW, Nieuwenhuizen W, Brommer EJP, van der Werf WGC.
    New enzyme immunoassay of fibrin-fibrinogen degradation products in plasma using a
    monoclonal antibody. J Lab Clin Med 1987; 109: 75-84.
11. Marder VJ, Shulman NR. High molecular weight derivatives of human fibrinogen produced
    by plasmin II. Mechanism of their anticoagulant activity. J Bioi Chern 1969; 244: 2120--4.
12. Gaffney PJ, Perry MI Unreliability of current serum fibrin degradation products (FDP)
    assays. Thromb Haemost 1985; 53: 301-2.
13. Niewiarowski S, Stewart GJ, Marder VI Formation of highly ordered polymers fibrinogen
    and fibrin degradation products. Biochim Biophys Acta 1970; 221: 326--41.
14. Koppert PW, Koopman J, Haverkate F, Nieuwenhuizen W Production and characterization
    of a monoclonal antibody reactive with a specific neoantigenic determinant (comprising B~
    54-118) in degradation products of fibrin and of fibrinogen. Blood 1986; 68: 437-41.
15. Koppert PW, Huijsmans CMG, Nieuwenhuizen W A monoclonal antibody, specific for human
    fibrinogen, fibrinopeptide A-containing fragments and not reacting with free fibrinopeptide
    A. Blood 1985; 66: 503-7
16. Koppert PW, Kuipers W, Hoegee-de Nobel E, Brommer EJP, Koopman J, Nieuwenhuizen W
    A quantitative enzyme immunoassay for primary fibrinogeno1ysis products in plasma. Thromb
    Haemostas 1987; 57: 25-8.
17. Koppert Pw, Hoegee-de Nobel E, Nieuwenhuizen W A monoclonal antibody-based enzyme
    immunoassay for fibrin degradation products in plasma. Thromb Haemostas 1988; 59: 310-15.
18. Kroneman H, Nieuwenhuizen W, Knot EAR. Monoclonal antibody-based plasma assays for
    fibrin(ogen) and derivatives, and their clinical relevance. Blood Coagul Fibrinol1990; 1:
    91-111.
19. Scheefers-Borchel U, Miiller-Berghaus G, Fuhge P, Eberle R, Heimburger N. Discrimination
    between fibrin and fibrinogen by a monoclonal antibody against a synthetic peptide. Proc
    Nat! Acad Sci USA 1985; 82: 7091-5.
20. Nieuwenhuizen W, Hoegee-de Nobel E, Laterveer R. A rapid monoclonal antibody-based
    enzyme immunoassay (EIA) for the quantitative determination of Soluble Fibrin in plasma.
    Thromb Haemostas 1992; 68; 273-7.
21. Bos R, Laterveer GH, Lockwood D, Szewczyk K, Nieuwenhuizen W A new enzyme
    immunoassay for Soluble fibrin, with a high discriminating power for thrombotic disorders.
    Thromb Haemostas 1998, in press.
22. Nieuwenhuizen W Soluble fibrin as a molecular marker for a pre-thrombotic state; a mini
    review. Blood Coagul Fibrinol1993; 4: 93-6.
23. Schielen WJG, Adams HPHM, van Leuven K, Voskuilen M, Tesser GI, Nieuwenhuizen W
    The sequence y-[312-324] is a fibrin-specific epitope. Blood 1991; 77: 2169-73.
24. Dempfle CE, Pfitzner SA, DoHman M, Huch K, Stehle G, Heene DL. Comparison of
    immunological and functional assays for measurement of Soluble fibrin. Thromb Haemostas
    1995; 74: 673-9.
                                              283
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
25. Wiman B, RAnby M. Determination of Soluble fibrin in plasma by a rapid and quantitative
    spectrophotometric assay. Thromb Haemostas 1986; 55: 189-93.
26. Bredbacka, S, Blombiick M, Wiman B. Soluble Fibrin - a predictor for the development and
    outcome of multiple organ failure. Am J Hemato11994; 46: 289-94.
27. Bounameaux H, de Moerloose P, Perrier A, Reber G. Plasma measurement of D-dimer as
    diagnostic aid in suspected venous thromboembolism - an overview. Thromb Haemost 1994;
    71: 1--6.
28. Elias A, ApteI I, Huc B, Chale JJ, Nguyen, F, Cambus JP, Boccalon H, Boneu B. D-dimer
    test and diagnosis of deep vein thrombosis: a comparative study of 7 assays. Thromb Haemostas
    1996; 76: 518-22.
29. Veitl M, Hamwi A, Kurturan A, Virgolini I, Vukovich Th. Comparison of four rapid D-dimer
    tests for diagnosis of pulmonary embolism. Thromb Res 1996; 82: 3~07.
30. Janssen MCH, Heebels AE, de Metz M, Verbruggen H, Wollersheim H, Janssen S, Schuurmans
    MMJ, Novakova IRO. Reliability of five rapid D-dimer assays compared to ELISA in the
    exclusion of deep venous thrombosis. Thromb Haemostas 1997; 72: 262--6.
31. Legnani C, Pancani C, Palareti G, Guazzaloca G, Fortunato G, Grauso F, Golfieri R,
    Gianpalma E, Coccheri S. Comparison of new rapid methods for D-dimer measurement to
    exclude deep vein thrombosis in symptomatic outpatients. Blood Coagul Fibrinol1997; 8:
    296-302.
32. Lee AY, Ginsberg JS. The role of D-dimer in the diagnosis of venous thromboembolism. Curr
    Opin Pulmon Med 1997; 3: 275-9.
33. Freyburger G, Trillaud H, Labrouche S, Gauthier P, Javorschi S, Bernard P, Grenier N. D-dimer
    strategy in thrombosis exclusion. A gold standard study in 100 patients suspected of deep
    venous thrombosis on pulmonary embolism: 8 DD methods compared. Thromb Haemo-
    stasl998; 79: 32-7.
34. Nieuwenhuizen W. A reference material for harmonization of D-dimer assays; SSC
    Communication. Thromb Haemostas 1997; 77: 1031-3.
                                              284
31
Venous occlusion test in fibrinolysis
assays
J.JESPERSEN
INTRODUCTION
Since its introduction more than 30 years ago 1,2 the venous occlusion test (VO
test) has been widely used to assess systemic fibrinolytic capacity after venous
occlusion. In the 1980s the YO test was included in the EeAT procedures and
in the EeAT clinical follow-up studies in order to have a response after a
standardized stimulus (low basal systemic fibrinolytic activity).
   Despite its apparent imperfection (see section on imprecision and stability
over time) with a relatively low correlation between repeated measurements of
the YO test, as reported in a subset of the EeAT prospective Angina Pectoris
Study 3, the plasma plasminogen activator tissue-type (t-PA) activity after the
YO test was decreased in patients at risk of a coronary event. Furthermore, all
the results of the EeAT Angina Pectoris Study of approximately 3000
individuals from suspected angina pectoris, and 106 definite coronary events
over a 2-year follow-up, pointed to a lowered fibrinolytic capacity as a risk
marker of a cardiac event, but its predictive power was smaller than that of
plasma protein concentration (baseline value)4.
Pathophysiology
In a number of observational studies a poor fibrinolytic response after venous
occlusion has frequently been observed in patients with recurrent venous
thrombosis5-8. At least five families have been identified with recurrent venous
thrombosis and an impaired fibrinolytic response to venous occlusion9-13. The
incidence of abnormal fibrinolysis in patients with venous thrombosis is about
30%6,8,14, and the YO test result also appears to have a predictive value for the
recurrence of venous thrombosis 15.
                                      285
         LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   A reduced fibrinolytic response after a VO test characterizes patients with
ischaemic heart disease (lHD), as revealed from case-control studies l 6--l9.
Decreased fibrinolytic capacity after venous occlusion has been shown to have
a predictive value as a risk marker for reinfarction and cardiac death among
young survivors of acute myocardial infarction 20 . As in the ECAT Angina
Pectoris Study the latter study used venous occlusion for 10 min; this relatively
short occlusion time was chosen in order to achieve a high patient acceptance
rate, i.e. high compliance.
EQUIPMENT
1. Blood pressure cuff, balloon and sphygmomanometer.
2. At least two blood test tubes placed in an ice-water bath.
      Table 31.1 Estimates of components of variation in plasma t-PA before and after venous occlusion (VO), and PAI-I of healthy young men with steady   o~
      lifestyles followed over I year (percentage of total variance shown in parentheses) (modified from ref. 35)                                         ~
                                                                                                                                                          -I
                                                                                                             Variation
                                                                                                                                                          m
                                                                                                                                                          o
                                                                                                                                                          :::c
      Variable                            nind        nabs       Mean                Total    Inter-personal        Intra-personal"-   Seasonal           z
                                                                                                                                                          oc
      t-PA protein (ng/ml)                16          79         4.1           SD2   1.88     0.95 (51%)            0.93 (49%)                            m
I\)                                                                            SD    1.37     0.98                  0.96                                  en
<Xl
<Xl                                                                                                                 15.92 (51%)        1.47 (5%)          Z
      t-PA protein + VO (ng/ml)           16          79         11.9          SD2   31.14     13.75 (44%)
                                                                               SD    5.58      3.71                 3.99               1.21               ~
                                                                                                                                                          JJ
      log (PAI-I protein) (ng/ml)         16          79         0.73 b        S~    0.105    0.037 (35%)           0.068 (65%)                           o
                                                                               SD    0.32     0.19                  0.26                                  s:::
                                                                                                                                                          OJ
      alnclude intraserial analytical variation - t-PA, CV 8%; PAl-I, CV 7%.
                                                                                                                                                          oen
      bpAI_I protein 5.37 ng/ml.                                                                                                                           en
      nind, number of individuals; nob., number of observations                                                                                             I
                                                                                                                                                           »
                                                                                                                                                           ~
                                                                                                                                                           z
                                                                                                                                                          c
                                                                                                                                                          »r-
                             VENOUS OCCLUSION TEST
Table 31.2 Descriptive statistics of plasma t-PA, and PAl-I, measured in healthy young adults
(n = 74);+ VO, after venous occlusion (n = 44) (modified from ref. 35)
                                      Non-parametric                 Parametric
*Medians: 3.S nglml (males) vs 2.6 nglml (females),p= 0.01 (Mann-Whitney U-test)
COMMENTS
Recently the VO test has been criticized because of an excessively high variability
of the venous occlusion itseW 1• This is not a problem only related to the VO
test, but also to the measurement of a large number of haemostatic factors
without venous occlusion. Both analytical imprecision and within-person (intra-
individual). variability can be substantial compared with variability amo~
persons3,3 ,42,43 (see also the section on imprecision and stability over time)37, •
Despite such a variability, even baseline PAI-l protein level and activity were
positively associated with, for example, coronary risk when adjustments were
made for centre, age, and sex4 • This indicates that the underlying relationship
to thrombotic risk must be greater than anticipated, since even a single
measurement was found to be an important risk marker, despite only moderate
reproducibility. This point also seems to occur with the VO test, but underlines
the need for improved measurement procedure, harmonization, and
standardization of haemostatic assays, including fibrinolytic assays and the
VO test. Thus, until a consensus is established on how to perform the VO test,
type of assays, degree of deviation from established on reference range, and
whether correction for haemoconcentration is needed or not, its main application
is in well-defined clinical studies.
                                             289
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
   This laboratory manual can be an important platform in the needed
standardization process. In the meantime each laboratory must describe any
deviation from the suggested standardized procedure, and must establish its
own reference material. Examples of such procedures are available (e.g. refs.
6, 8, 14, 32,40, 45,and 46).
References
 1. Nilsson 1M, Robertson B. Effect of venous occlusion on coagulation and fibrinolytic
    components in normal subjects. Thromb Diathes Haemorrh 1968; 20: 397--408.
 2. Robertson BR, Pandolfi M, Nilsson 1M. 'Fibrinolytic capacity' in healthy volunteers as
    estimated from effect of venous occlusion of arms. Acta Chir Scand 1972; 138: 429-36.
 3. Pyke SDM, Thompson SG, Buchwalsky R, Kienast J, on behalf of the ECAT Angina Pectoris
    Study Group. Variability over time of haemostatic and other cardiovascular risk factors in
    patients suffering from angina pectoris. Thromb Haemostas 1993; 70: 743-6.
 4. Thompson SG, Kienast J, Pyke SDM, Haverkate F, van de Loo JCW, for the European
    Concerted Action on Thrombosis and Disabilities Angina Pectoris Study Group. Hemostatic
    factors and the risk of myocardial infarction or sudden death in patients with angina pectoris.
    N EnglJ Med 1995; 332: 635--41.
 5. Isacson S, Nilsson 1M. Defective fibrinolysis in blood and vein walls in recurrent 'idiopathic'
    venous thrombosis. Acta Chir Scand 1972; 138: 313-19.
 6. Nilsson 1M, Ljungner H, Tengborn L. Two different mechanisms in patients with venous
    thrombosis and defective fibrinolysis: low concentration of plasminogen activator or increased
    concentration of plasminogen activator inhibitor. Br Med J 1985; 290: 1453-6.
 7. Stalder M, Hauert J, Kruithof EKO, Bachmann F. Release of vascular plasminogen activator
    (v-PA) after venous stasis: electrophoretic-zymographic analysis of free and complexed v-PA.
    Br J Haemato11985; 61: 169-76.
 8. Juhan-Vague I, Valadier J, Alessi MC et al. Deficient t-PA release and elevated PA inhibitor
    levels in patients with spontaneous or recurrent deep venous thrombosis. Thromb Haemostas
    1987; 57: 62-72.
 9. Johansson L, Hedner U, Nilsson 1M. A family with thromboembolic disease associated with
    deficient fibrinolytic activity in vessel wall. Acta Med Scand 1978; 203: 477-80.
10. Alexandre P, Larcan A, Briquel ME. Recurring thrombo-embolic accidents caused by family-
    related deficiency of the fibrinolysis system. Blut 1980; 41: 437--44.
II. Jorgensen M, Mortensen JZ, Madsen AG, Thorsen S, Jacobsen B. A family with reduced
    plasminogen activator activity in blood associated with recurrent venous thrombosis. Scand
    J Haeinatol1982: 29: 217-23.
12. Stead Nw, Bauer KA, Kinney TR et al. Venous thrombosis in a family with defective release
    of vascular plasminogen activator and elevated plasma factor VIII/von Willebrand's factor.
    Am J Med 1983; 74: 33-9.
13. Boyko OB, Pizzo SY. Mesenteric vein thrombosis and vascular plasminogen activator. Arch
    Pathol Lab Med 1983; 107: 541-2.
14. Alessi MC, Juhan-Vague I, Valadier J, Philip-Joet C, Holvoet P, Collen D. Relevance of free
    tPA assay following venous occlusion in patients with venous thromboembolic disease. Thromb
    Haemostas 1988; 59: 346-7.
15. Korninger C, Lechner K, Niessner H, GOssinger H, Kundi M. Impaired fibrinolytic capacity
    predisposes for recurrence of venous thrombosis. Thromb Haemost 1984; 52: 127-30.
16. Walker ID, Davidson JF, Hutton I, Lawrie TOY. Disordered 'fibrinolytic potential' in coronary
    heart disease. Thromb Res 1977; 10: 509-20.
17. Gritsyuk AI, Schogelsky VI. Evaluation of blood coagulation and prethrombotic state in
    patients with coronary atherosclerosis by application of controlled local venous blockade.
    Circulation 1979; 60: 220A.
18. Rawles JM, Warlow C, Ogston D. Fibrinolytic capacity of arm and leg veins after femoral
    shaft fracture and acute myocardial infarction. Br Med J 1975; 2: 61-2.
19. Hamsten A, Blombiick M, Wiman B. Svensson J, Szamosi A, de Faire U, Mettinger L.
    Haemostatic function in myocardial infarction. Br Heart J 1986; 55: 58-66.
                                               290
                               VENOUS OCCLUSION TEST
20. Hamsten A, de Faire U, Walldius G et al. Plasminogen activator inhibitor in plasma: risk
    factor for recurrent myocardial infarction. Lancet 1987; 2: 3-9.
21. Jespersen 1. The diurnal increase in euglobulin fibrinolytic activity in women using oral
    contraceptives and in normal women, and the generation of intrinsic fibrinolytic activity.
    Thromb Haemostas 1986; 56: 183-8.
22. Rijken DC, Wijngaards G, Welbergen J. Relationship between tissue plasminogen activator
    and the activators in blood and vascular wall. Thromb Res 1980; 18: 815-30.
23. Kluft C. Wijngaards G, Jie AFH. The factor XII-independent plasminogen proactivator
    system of plasma includes urokinase-related activity. Thromb Haemostas 1981; 46: 343.
24. Kluft C, Jie AFH. Interaction between the extrinsic and intrinsic system of fibrinolysis. In:
    Davidson JF et al. eds. Progress in clinical fibrinolysis and thrombolysis. Edinburgh: Churchill
    Livingstone, 1979: 25-31.
25. Kluft C, Dooijewaard G, Emeis JJ. Role of the contact system in fibrinolysis. Semin Thromb
    Hemostas 1987; 13: 50-68.
26. Jespersen 1. Pathophysiology and clinical aspects of fibrinolysis and inhibition of coagulation.
    Experimental and clinical studies with special reference to women on oral contraceptives and
    selected groups of thrombosis prone patients. Thesis. Dan Med Bull 1988; 35: 1-33.
27. Rijken DC, Juhan-Vague I, De Cock F, Collen D. Measurement of human tissue-type
    plasminogen activator by two-site immuno radiometric assay. J Lab Clin Med 1983; 101:
    274-84.
28. Verheijen JH, Mullaart E, Chang GTG, Kluft C, Wijngaards G. A simple, sensitive
    spectrophotometric assay for extrinsic (tissue-type) plasminogen activator applicable to
    measurements in plasma. Thromb Haemostas 1982; 48: 266-9.
29. Chmielewska IN, Wiman B. Determination of tissue plasminogen activator and its 'fast'
    inhibitor in plasma. Clin Chem 1986; 32: 482-5.
30. Jespersen 1. How to detect defects of tissue plasminogen activator in thrombosis-prone patients.
    An introduction to discussion. Fibrinolysis 1988; 2 (Suppl. 2): 104-11.
31. Tengborn L. Laboratory investigation of fibrinolytic and other haemostatic risk factors in
    patients with venous thromboembolism. In: Nilsson TK, Boman K, Jansson JH, eds. Clinical
    aspects of fibrinolysis. Stockholm: Almqvist & Wiksell International, 1991; 67-84.
32. Nguyen G, Horellou MH, Kruithof EKO, Conard J, Samama MM. Residual plasminogen
    activator inhibitor activity after venous stasis as a criterion for hypofibrinolysis: a study in
    83 patients with confirmed deep vein thrombosis. Blood 1988; 72: 601-5.
33. SyrjiUii MT, Krusius T, Petiijii J, Vahtera E, Rasi V. Venous occlusion test: Assessment of
    fibrinolytic capacity by D-dimer latex agglutination test. Fibrinolysis 1993; 7: 41-5.
34. Stegnar M, Pentek M. Fibrinolytic response to venous occlusion in healthy subjects:
    Relationship to age, gender, body weight, blood lipids and insulin. Thromb Res 1993; 69:
    81-92.
35. Marckmann P, Sandstrom B, Jespersen 1. The variability of and associations between measures
    of blood coagulation, fibrinolysis and blood lipids. Atherosclerosis 1992; 96: 235-44.
36. Kluft C. Constitutive synthesis of tissue-type plasminogen activator (t-PA) and plasminogen
    activator inhibitor type 1 (PAl -1): conditions and therapeutic targets. Fibrinolysis 1994; 8
    (Suppl. 2): 1-7.
37. Thompson SG, Martin JC, Meade TW Sources of variability in coagulation factor assays.
    Thromb Haemostas 1987; 58: 1073-7.
38. Jennings I, Luddington RJ, Harper PL. Changes in endothelial-related coagulation proteins
    in response to venous occlusion. Thromb Haemostas 1991; 65: 374-6.
39. Keber D. Mechanism of tissue plasminogen activator release during venous occlusion.
    Fibrinolysis 1988; 2 (Suppl. 2): 96-103.
40. Petiijii 1. Fibrinolytic response to venous occlusion for 10 and 20 minutes in healthy subjects
    and in patients with deep vein thrombosis. Thromb Res 1989; 56: 251-63.
41. Stegnar M, Mavri A. Reproducibility of fibrinolytic response to venous occlusion in healthy
    subjects. Thromb Haemostas 1995; 73: 453-7.
42. Gram J, Declerck PJ, Sidelmann J, Jespersen J, Kluft C. Multicentre evaluation of commercial
    kit methods: plasminogen activator inhibitor activity. Thromb Haemostas 1993; 70: 852-7.
43. Declerck PJ, Moreau H, Jespersen J, Gram J, Kluft C. Multicenter evaluation of commercially
    available methods for the immunological determination of plasminogen activator inhibitor-l
    (PAl-I). Thromb Haemostas 1993; 70: 858-63.
                                               291
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
44. Jansson J-H, Norberg B, Nilsson TK. Impact of acute phase on concentrations of tissue
    plasminogen activator and plasminogen activator inhibitor in plasma after deep-vein thrombosis
    or open-heart surgery. Clin Chern 1989; 35: 1544-5.
45. Nicoloso G, Hauert J, Kruithof EKO, Van Melle G, Bachmann F. Fibrinolysis in normal
    subjects - comparison between plasminogen activator inhibitor and other components of the
    fibrinolytic system. Thromb Haemostas 1988; 59: 299-303.
46. Sultan Y, Harris A, Strauch G, Venot A, De Lauture D. A dynamic test to investigate potential
    tissue plasminogen activator activity. Comparison of deamino-8-D-argininevasopressin with
    venous occlusion in normal subjects and patients. J Lab Clin Med 1988; 111: 645-53.
                                              292
32
List of manufacturers
Materials and reagents for the procedures and the assays below correspond with
the order and the content of the assay chapters and are followed by a list of
addresses.
Chapter 8: Fibrinogen
  Standard 89/644                                   NIBSC
                                    294
                         LIST OF MANUFACTURERS
                                    295
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
  Commercially available kits
  IL Test Protein S assay kit                        Instrumentation
                                                     Laboratory
  Protein S Reagent                                  Dade-Behring
  Staclot protein S                                  Diagnostica Stago
  Standard 93/590                                    NIBSC
                                     296
                        LIST OF MANUFACTURERS
  Suppliers of reagents for the FPA
  radioimmunoassay                                 Bachem
                                                   Imco
                                                   Kordia
                                                   Wherl
                                     298
                        LIST OF MANUFACTURERS
  PPACK                                           Calbiochem
  Thimerosal                                      Merck
  Tween 20                                        Sigma
  Commercially available kits
  PAP kits (ELISA)                               Dade Behring
                                                 Technoc1one
                                                 Teijin
ADDRESSES
Agen Biomedical Ltd                   American Diagnostica Inc.
11 Durbell Street                     222 Railroad Avenue
PO Box 391                            PO Box 1165
Acacia Ridge 4110                     Greenwich CT 06836-1165
Brisbane                              USA
Australia                             Phone: + 1 2036610000
Phone: +61 733706300                  Fax: + 1 203661 7784
Fax: + 61 733706370                   E-mail: [email protected]
Internet: http://www.agen.com.au
E-mail: [email protected]              Avanti Polar Lipids Inc.
                                      700 Industrial Park Drive
                                      Alabaster
                                      AL 35007
American Biogenetic Sciences          USA
1375 Akron Street Copiague            Phone: + 1 80022706511 + 1205663
NY 11726                              2494
USA                                   Fax: + 1 800 229 10041 + 1 205 663
Phone: + 1 516 789 2600               0756
Fax: + 1 516789 1661                  Internet: http://www.avantilipids.com
Internet: http://www.mabxa.com        E-mail: [email protected]
                                   299
        LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
                                      303
Index
                                               305
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
 TAT complexes 212                                factor VIII inhibitors 185
CTAJ) 25,26,234                                   factor XII 3
                                                  familial thrombophilia 130
D-dimer 36, 275                                   fibrin monomer 276
DDAVP 232                                         fibrin(ogen) degradation products 36,276
deep vein thrombosis                              fibrinogen
   antithrombin activity and antigen 124             fibrinogen 79
   APTT 40                                           introduction 1
  endogenous thrombin potential 77                   quality assessment 31, 36
   FPA 199                                           soluble fibrin and degradation products of
   protein S antigen 143                                  fibrinogen 276
   prothrombin fragment Fl +2 218                 fibrinopeptide A (FPA) 199,217
   soluble fibrin and degradation products of
        fibrinogen 279,282                        good laboratory practice 3, 9
  TAT complexes 211,213                           good medical laboratory services (GMLC) 9
   t-PA antigen 232
diabetes mellitus                                 haematocrit 22
   FPA 200                                        haemophilia A
   heparin cofactor II 191                           activated factor VII 91
  protein C activity and antigen 131                factor VIII clotting activity 112
  protein S antigen 143                              von Willebrand factor 118
   prothrombin fragment Fl +2 218                 HDL cholesterol 227
   t-PA activity 227                              heart disease 90
  von Willebrand factor 118                       heparin cofactor
disseminated intravascular coagulation (DIC)        antithrombin activity and antigen 121
  antithrombin activity and antigen 126             APC resistance 165
  APTT 40                                           APTT 37,38,39,43
  factor VIII clotting activity 112                 blood collection 23
  fibrinogen 80, 85                                 endogenous thrombin potential 67
   FPA 199                                          fibrinogen 85
  heparin cofactor II 191                           heparin cofactor II 189
  plasmin inhibitor activity 260                    lupus anticoagulant 186
  plasmin---a-antiplasmin complexes 265,            PT 47
        266                                         quality assessment 36
  protein C activity and antigen 131                soluble fibrin and degradation products of
  prothrombin fragment Fl+2 218                           fibrinogen 277,282
  TAT complexes 213                                 TFPI 172
  TFPI 173                                        hepaticfailure 191
DNA analysis 27                                   hirudin 190
dysfibrinogenaenlia 80,277                        histidine-rich glycoprotein 247
                                                  hormone replacement 159
eclampsia 282                                     human immunodeficiency 191
elastase---al-antiproteinase complex 209          human leuserpin-2 189
elastase 196, 260                                 hypercholesteroiaemia 173, 213
EN 45001 10                                       hyperiipaemia 85
EN 46001 14                                       hyperlipoproteinaemia 260
EN 46002 14                                       hypertension 118, 131
evacuated tubes 24, 42, 226
                                                  infections 184
factor V Leiden 25,130,213                        inflammatory diseases 112, 118
factor VII 26, 31                                 INR 24, 35, 36, 133
factor VIII                                       insulin 286
  APC resistance 163,164                          insulin resistance 227
  APTT 43                                         ischaenlic heart disease 90
  blood col1ection 22, 23, 25, 26                 international sensitivity index (lSI)   54, 59
  factor VIII clotting activity 107               ISO 25 11,12
  quality assessment 32, 33, 36                   ISO 9001 14
  von Willebrand factor 117                       IS09002 14
                                                306
                                            INDEX
kallikrein 231                                        APTT 39
                                                      endogenous thrombin potential 77
L-asparaginase 126,131,260                            FPA 200
leukaemia 213, 260                                    heparin cofactor II 191
lifestyle variables 22                                protein C activity and antigen 131
lipids 286                                            protein S antigen 143
~-lipoproteinaemia 173                                prothrombin fragment FI +2 218
liver cirrhosis                                       prothrombin time 47
   antithrombin activity and antigen 125
   FPA 200                                        plasmin 276,286
   soluble fibrin and degradation products of     plasmin inhibitor 231,247,257
         fibrinogen 282                           plasmin~-antiplasmin complexes 265
   t-PA activity 227                              plasminogen 2,25,36,247
   von Willebrand factor 118                      plasminogen activator (t-PA)
liver disease                                       blood collection 25
   APTI 40                                          introduction 3
   protein C activity and antigen 131               PAl-l antigen 239
   protein S activity 159                           plasmin inhibitor activity 260
   protein S antigen 143                            plasmin-a-antiplasmin complexes 265
   prothrombin time 47,60                           t-PA activity 223
   soluble fibrin and degradation products of       t-PA antigen 231
         fibrinogen 282                             venous occlusion test 285, 288, 289
   TAT complexes 213                              plasminogen activator inhibitor-I (PAl-I)
liver function 46                                   blood collection 25
liver transplantation 282                           PAl-I antigen 239
lupus anticoagulant                                 t-PA activity 223
   APC resistance 165                               venous occlusion test 286, 288, 289
   APTT 37,38                                     platelet factor 3 26
   quality assessment 36                          platelet factor 4 26
                                                  polybrene 47
Ilrmacroglobulin 65, 130, 260                     post-infectious 159
malignancy 213,282                                post-surgical patients
menopausal status 124                               antithrombin activity and antigen 126,
mesenteric vein thrombosis 124                            131
myocardial infarction                               factor VIII clotting activity 112
   FPA 199                                          plasmin inhibitor activity 260
   plasmin-a-antiplasmin complexes 266,             TAT complexes 213
        272                                         t-PA activity 227
   prothrombin fragment Fl +2 218                   von Willebrand factor 118
   soluble fibrin and degradation products of     pre-analytical factors 4
       fibrinogen 282                             pre-analytical variation 21
   TAT complexes 213                              precision 4, 6
   t-PA activity 227                              pro-urokinase system 3
   venous occlusion test 286                      proficiency testing 32
                                                  protein C 32, 36, 129
nephropathy 227                                   protein S 36, 141, 153
nephrotic syndrome                                prothrombin fragment Fl +2 209,217
  antithrombin activity and antigen 126           prothrombin time (PT)
  heparin cofactor II 191                           APTT 37
  plasmin inhibitor activity 260                    factor VII activity and antigen 100, 103
  protein C activity and antigen 131                fibrinogen 83
  protein S activity 159                            good medical laboratory services 24
  protein S antigen 143                             introduction 2, 4
                                                    lupus anticoagulant 183
oral anticoagulants                                 prothrombin time 45
  antithrombin activity and antigen 124,            quality assessment 32, 35
       126                                          TFPI 172
  APC resistance 164                              PTCA 272
                                                307
          LABORATORY TECHNIQUES IN THROMBOSIS - A MANUAL
pulmonary embolism                                   AP1T 37
  antithrombin activity and antigen 124              blood collection 21
  soluble fibrin and degradation products of         introduction 2, 4
       fibrinogen 279,282                            protein S activity 157
  TAT complexes 211,213                              prothrombin time 45, 56
pulmonary thrombosis 199                             quality assessment 36
purpura fulminans 130, 142                        thrombosis
                                                     antithrombin activity and antigen 125
quality assessment 1, 10, 14, 18,29,30,32,           APC resistance 167
       39                                            soluble fibrin and degradation products of
quality management 11,12,14,17                            fibrinogen 280
                                                     TFPI 173
renal failure 191,260                                t-PA antigen 232
respiratory distress syndrome 218                 Thrombotest 51
ristocetin cofactor 115                           thrombotic diseases 90,239,276
                                                  tissue factor pathway inhibitor (TFPI) 65,
sample preparation 21                                     99,171
sepsis 212                                        total quality management (TQM) 10
Shewhart plots 10                                 triglycerides 227
sickle-cell disease 131
soluble fibrin 275                                urokinase-type PA 239,260
spontaneous abortions 184                         unstable angina
Stabilyte 25, 226                                   TAT complexes 213
streptokinase                                       TFPI 182
   plasmin-u-antiplasmin complexes 266            uraemia
   plasminogen activity 247                           plasmin-u-antiplasmin complexes 266,
   soluble fibrin and degradation products of              270
        fibrinogen 276,282                            plasminogen activity 247
stroke 183, 266                                       soluble fibrin and degradation products of
surgery                                                    fibrinogen 282
   antithrombin activity and antigen 124              venous occlusion test 286
   APTf 40                                            von Willebrand factor 118
   heparin cofactor II 191
   prothrombin fragment Fl +2 211                 venous occlusion test 285
   prothrombin time 47                            venous thromboembolism 164
systemic lupus erythematosus (SLE) 183            venous thrombosis
                                                     antithrombin activity and antigen 121
thrombin-antithrombin (TAT)                          lupus anticoagulant 183
        complex 209, 217                             protein S antigen 142
thrombin 1                                           venous occlusion test 285
thrombocytopenic purpura 118                         von Willebrand factor 130
thromboembolic disease 183                        vitronectin 239
thromboembolism 286                               von Willebrand disease 118,232
thrombomodulin 65, 129                            von Willebrand factor 22, 36, 115
thrombophilia 3
thromboplastin                                    warfarin 47,91
   activated factor VII 89                        Westgard rules 10
308