10 1 1 463 3921
10 1 1 463 3921
10 1 1 463 3921
Thrombocytopenia
Third Edition, Revised and Expanded
edited by
Theodore E. Warkentin
McMaster University
Hamilton Regional Laboratory Medicine Program, and
Hamilton Health Sciences, General Site
Hamilton, Ontario, Canada
Andreas Greinacher
Ernst-Moritz-Arndt University Greifswald
Greifswald, Germany
MARCEL
MARCEL
DEKKER,
INC. NEWYORK BASEL
DEKKER
Although great care has been taken to provide accurate and current information,
neither the author(s) nor the publisher, nor anyone else associated with this publica-
tion, shall be liable for any loss, damage, or liability directly or indirectly caused or
alleged to be caused by this book. The material contained herein is not intended to
provide specific advice or recommendations for any specific situation.
ISBN: 0-8247-5625-8
Headquarters
Marcel Dekker, Inc., 270 Madison Avenue, New York, NY 10016, U.S.A.
tel: 212-696-9000; fax: 212-685-4540
The publisher offers discounts on this book when ordered in bulk quantities. For more
information, write to Special Sales/Professional Marketing at the headquarters
address above.
Neither this book nor any part may be reproduced or transmitted in any form or by
any means, electronic or mechanical, including photocopying, microfilming, and re-
cording, or by any information storage and retrieval system, without permission in
writing from the publisher.
10 9 8 7 6 5 4 3 2 1
Editor-in-Chief
Samuel Z. Goldhaber, M.D.
Harvard Medical School
and Brigham and Women's Hospital
Boston, Massachusetts
T.E.W.
A.G.
Series Introduction
Samuel Z. Goldhaber
v
Preface to the Third Edition
Theodore E. Warkentin
Andreas Greinacher
Preface to the Second Edition
thrombin generation, and new animal models for studying this fascinating
syndrome. Even the clinical syndrome itself is now better understood: The
influence of previous heparin exposure on the timing of onset of HIT has been
clarified, and the peculiar transience of HIT antibodies has been shown. These
clinical and laboratory insights provide a firmer basis for estimating pretest
probabilities of HIT in various clinical settings, and also give a scientific
rationale for considering re-exposure to heparin in a patient with previous
HIT but whose antibodies are no longer detectable.
Ironically, improvements in a laboratory testing also mean that not all
detectable HIT antibodies are truly pathogenic. Thus, physicians need to
interpret results of diagnostic assays in the clinical context, so as to estimate
accurately the posttest probability of HIT. A major aim of this book is to
provide the relevant information for understanding such a ‘‘clinipathologic’’
framework of HIT.
More and more, it seems, HIT is an issue in cases of alleged medical
malpractice. This is because HIT often occurs in patients who received heparin
for prophylaxis of thrombosis. So it can seem evident even to a nonmedical
person that something fundamentically went wrong if the patient ended up
with severe pulmonary embolism or even limb loss.
To address these new developments, and others, all chapters of the first
edition have been updated. In addition, two new chapters have been added,
one discussing the use of argatroban for management of HIT and the other
dealing with U.S. perspectives on medicolegal aspects of HIT.
Theodore E. Warkentin
Andreas Greinacher
Preface to the First Edition
represents perhaps the most striking of all the HIT treatment paradoxes (see
Chap. 13): two antithrombotic agents with distinct adverse event profiles that
interact to produce a profound disturbance in procoagulant–anticoagulant
balance, i.e., increased thrombin generation (secondary to HIT) together with
acquired, severe protein C deficiency (secondary to coumarin treatment).
Finally, the concept of HIT as a hypercoagulable state with in vivo thrombin
generation provides a rationale for understanding the efficacy of new thera-
pies that either reduce thrombin generation by inhibition of factor Xa (e.g.,
danaparoid) or directly inactive thrombin (e.g., lepirudin).
A third theme of this book is the peculiarly inconstant nature of HIT, in
particular, its variable frequency and clinical presentation among different
patient populations treated with heparin. Figure 1 depicts HIT as an iceberg
within which a variety of clinical and laboratory factors interact to influence
antibody formation, development of thrombocytopenia, and, finally, result-
ing clinical complications, such as thrombosis. A recent, novel concept is that
the size and buoyancy of the HIT icebergs can vary among different patient
populations who receive heparin. This concept of multiple icebergs of HIT is
shown in Chapter 4, Fig. 3. Unraveling the clinical and laboratory determi-
nants for these differences in the icebergs among patient populations is a
major challenge of current and future investigation.
Why should HIT be the subject of a book? First and foremost, HIT is
common, and nonimmune HAT is very common. According to the Council
for International Organization of Medical Sciences (CIOMS III), adverse
drug reactions can be classified as common if they occur in 1–10% of patients,
and very common if they occur in 10% or more of patients. There is convincing
evidence that HIT occurs in as many as 5% of certain patient populations,
such as postoperative orthopedic patients receiving unfractionated heparin.
The clinical influence of HIT is substantial: about half of these patients
develop HIT-associated thrombosis. Nonimmune HAT occurs in as many as
30% of certain patient populations. Thus, physicians need to be able to re-
liably distinguish among the various thrombocytopenic disorders that occur
during heparin treatment. This will minimize the risk of inappropriate treat-
ments, such as failing to stop heparin administration in a patient with prob-
able HIT, or deciding to stop heparin in a patient with nonimmune HAT or
pseudo-HIT. Because HIT is a life- and limb-threatening iatrogenic illness
with many diagnostic and treatment pitfalls, medicolegal consequences of
caregiver’s action or inaction can be significant (see Chap. 18).
A second reason for the compilation of this book is that most of the
pieces of the HIT puzzle are now firmly in place. Consensus has emerged on
several key aspects of the syndrome, including the nature of its target antigen,
the participation of platelet and endothelial cell activation in the pathogenesis
of thrombosis, the frequency of HIT, and optimal laboratory testing. The
publication of this book reflects this coherence in our understanding of the
HIT syndrome. Yet there remain important unresolved issues: for example,
what is the fundamental nature of the ‘‘autoimune’’ response to the PF4–
heparin neoepitope? Why is the immune response to the HIT antigen so tran-
sient? Why do only a subset of patients who form HIT antibodies develop
clinical HIT?
Heparin has been, and will continue to be, one of the most important
agents for the prophylaxis and treatment of venous and arterial thromboem-
bolism. Consequently, HIT will continue to be an important management
problem for some time to come. Both of us have spent a decade of our sci-
entific careers providing, in the context of other investigators’ work, a rational
management approach aimed at minimizing morbidity and mortality among
the many patients who develop the most common immune-mediated adverse
drug reaction in clinical medicine. The importance of controlling thrombin
generation in HIT is now widely appreciated. The book should help guide
clinicians through the often paradoxical clinical and management problems
posed by patients with HIT (see Chaps. 13–17).
The book is a tribute to our scientific mentors, John G. Kelton and
Christian Mueller-Eckhardt, and the close cooperation of many of our scien-
tific colleagues and personal friends whose efforts made this project possible.
Preface to the First Edition xv
Theodore E. Warkentin
Andreas Greinacher
REFERENCES
Appendixes
1 Ten Clinical ‘‘Rules’’ for Diagnosing HIT 597
2 Estimating the Pretest Probability of HIT: The Four T’s 599
3 Platelet Count Monitoring for HIT 600
4 Treatment Recommendations 602
5 Danaparoid Dosing Schedules in HIT Patients 606
6 Dosing Schedules for Lepirudin Treatment of Patients with
HIT 608
7 Dosing Schedule for Lepirudin in Patients with HIT and
Renal Impairment 610
8 Dosing Schedules for Argatroban Treatment of Patients
with HIT (Approved Indications) 611
9 Timelines of an Episode of HIT 612
Index 613
Contributors
Harry N. Magnani, M.D., B.S., M.Sc., Ph.D. Medical Consultant, Oss, The
Netherlands
animal hepatic tissues (Gr. DN kaU [h epar], liver) (Howell and Holt, 1918).
Despite its in vitro anticoagulant action, the inability of heparin to prevent
platelet-mediated thrombosis (Shionoya, 1927) made it uncertain whether it
had antithrombotic potential. However, animal (Mason, 1924) and human
studies (Crafoord, 1937) showed that heparin could prevent thrombosis. By
the 1950s, heparin was established as an important therapeutic agent in the
treatment of venous and arterial thrombosis.
gested that heparin might cause arterial embolism in some patients. Rodger E.
Weismann, a 43-year-old Assistant Professor of Clinical Surgery at the
Dartmouth Medical School (Fig. 1), and his Resident in Surgery, Dr. Richard
W. Tobin, presented their 3-year experience with 10 patients who developed
unexpected peripheral arterial embolism during systemic heparin therapy at
the Mary Hitchcock Memorial Hospital, in Hanover, New Hampshire. Their
first patient with this complication was reported in detail, and to this day
represents a classic description of the syndrome:
This 62-year-old white woman was admitted to the Hitchcock Hospital
Feb 8, 1955, with left retinal detachment, complicating longstanding
myopia. . . . Left scleral buckling was carried out on Feb. 10, and strict
bed rest was required during the ensuing three weeks. On her beginning
ambulation, on March 6, signs and symptoms of left iliofemoral throm-
bophlebitis were noted, for which systemic heparinization was begun
(. . . heparin sodium in divided subcutaneous doses, totaling 150–300 mg
per day . . .). On March 16 . . . . , after 10 days of anticoagulation therapy,
sudden signs of right common femoral arterial occlusion led to the diag-
nosis of common femoral arterial embolism. Successful femoral em-
bolectomy was carried out. She was kept on adequate heparinization
and made a satisfactory initial recovery until March 19, . . . when signs
of sudden occlusion of the distal aorta appeared.
History of HIT 3
also observed similar events, none were raised (Weismann, personal commu-
nication, July 1998). However, a few years later, Brooke Roberts and col-
leagues from the University of Pennsylvania in Philadelphia described a series
of patients who were remarkably similar to those reported by Weismann and
Tobin (Roberts et al., 1964; Barker et al., 1966; Kaupp and Roberts, 1972).
The key features were summarized as follows:
To witness a series of apparently paradoxical events is disconcerting as
well as challenging. When such paradoxes involve totally unexpected
results following the use of a major therapeutic agent, it is at first
difficult to know whether the relationship is causal or merely
coincidental. When, however, the same series of events has been seen
repeatedly it is difficult to escape the conclusion that there is some causal
relationship, even though the mechanism by which it is accomplished
may be unknown. . . . During the last 9 years at the Hospital of the
University of Pennsylvania we have seen a group of 11 patients who
suffered unexplained arterial embolization for the first time while being
treated with heparin for some condition that could not of itself
reasonably be expected to cause arterial emboli. . . . All patients had
been receiving heparin for 10 days or more when the initial embolus
occurred. . . . All emboli removed were of a light color, seemingly made
up primarily of fibrin and platelets, and microscopically appeared to be
relatively free of red cells. . . . All patients in this group had multiple
emboli. . . . Of the 4 deaths, 3 were attributed to cerebral vascular
accidents presumably embolic in origin and 1 was thought to have
resulted from a perforation of the small bowel 2 weeks after the removal
of a mesenteric embolus (Roberts et al., 1964).
Roberts’ group also viewed the likely pathogenesis as that of emboli-
zation of platelet-fibrin–rich material originating within the aorta, rather than
the heart. Furthermore, they believed that the thrombi were initially formed
on aortic ulcerations that acted as a nidus for thrombus formation. This
pathogenesis was suggested by the observation that such adherent thrombi
could be removed from the proximal aorta in a few of the patients (Roberts et
al., 1964; Kaupp and Roberts, 1972).
D. The ‘‘
‘‘White Clot Syndrome’’
’’
Jonathan Towne, a vascular surgeon in Milwaukee, reported with his
colleagues (1979) that the pale thrombi characteristic of this syndrome
consisted of fibrin–platelet aggregates (electron microscropy). These workers
coined the term ‘‘white clot syndrome’’ to describe the characteristic appear-
ance of these arterial thromboemboli. Ironically, their report is also the first to
note the occurrence of phlegmasia cerulea dolens (severe venous limb is-
chemia) that progressed to venous limb gangrene in two of their patients (i.e.,
a syndrome of limb loss due to extensive venous thrombosis without arterial
white clots). Nonetheless, the designation of white clot syndrome has become
virtually synonymous with HIT in both North America and Europe (Benha-
mou et al., 1985; Stanton et al., 1988), despite the lack of specificity of these
thrombi for HIT (see Chap. 12).
ral et al., 1992). This important discovery led to an explosion of basic studies
in numerous laboratories that led to further characterization of the basic
pathogenesis of HIT (see Chaps. 6–8). Amiral’s discovery also led to the de-
velopment of new assays for HIT antibodies based on enzyme immunoassay
techniques (see Chap. 11).
The antigen site(s) recognized by HIT antibodies were identified as
being on PF4, rather than on heparin itself or a compound antigen (Li et al.,
2002) (see Chaps. 6–8). This observation highlights intriguing parallels
between HIT and the antiphospholipid syndrome. This latter disorder is also
characterized by pathogenic antibodies directed against one or more proteins
that express neoepitopes when bound to certain negatively charged phospho-
lipid surfaces (see Chap. 12). The presence of neoepitopes on the ‘‘self ’’
protein, PF4, suggests the HIT can be conceptualized as a transient, drug-
induced, platelet- and coagulation-activating autoimmune disorder. Indeed,
high-titer HIT antibodies that are able to activate platelets in vitro even in the
absence of pharmacologic heparin have been associated with the onset of
thrombocytopenia and thrombosis beginning several days after heparin has
been discontinued, so-called delayed-onset HIT (Warkentin and Kelton,
2001) (see Chap. 3).
A. Danaparoid Sodium
In 1982, a 48-year-old vacationing American developed deep venous throm-
bosis and pulmonary embolism following a transatlantic flight to Germany.
Heparin treatment was complicated by thrombocytopenia and progression of
venous thrombosis. Professor Job Harenberg of Heidelberg University, who
had performed phase I evaluations of the experimental glycosaminoglycan
anticoagulant danaparoid, requested this agent from the manufacturer (NV
Organon, The Netherlands). The platelet count recovered and the venous
thrombosis resolved (Harenberg et al., 1983, 1997). Over the next 6 years, this
patient developed recurrent thromboembolic events, each time successfully
treated with danaparoid. This favorable experience led to a named-patient,
compassionate-release program ending in March 1997, during which time
somewhat over 750 patients were treated with this agent. Additionally, Chong
and colleagues (2001) performed the first randomized, controlled clinical trial
evaluating danaparoid (see Chap. 14).
History of HIT 13
necrosis. The author questioned whether the warfarin had also contributed to
the pathogenesis of the venous limb gangrene. This hypothesis was directly
tested just 2 months later when a second young woman developed severe
phlegmasia cerulea dolens of an upper limb during treatment of deep vein
thrombosis complicating HIT with ancrod and warfarin. Treatment with
vitamin K and plasma given by pheresis reversed the phlegmasia. Further
laboratory studies supported this hypothesis of a disturbance in procoagu-
lant-anticoagulant balance during treatment of HIT with warfarin (Warken-
tin et al., 1997) (see Chaps. 3, 12, and 13).
Increasingly, HIT became viewed as a syndrome characterized by
multiple prothrombotic events, including not only platelet and endothelial
cell activation, but also profound activation of coagulation pathways. This
conceptual framework provides a rationale for antithrombotic therapy that
reduces thrombin generation in patients with HIT (Warkentin et al., 1998).
B. Argatroban
A synthetic small-molecule thrombin inhibitor derived from L-arginine, now
known as argatroban, was used in Japan during the 1980s as a treatment for
chronic arterial occlusion (Tanabe, 1986). During this time, argatroban also
underwent investigation as treatment for HIT in Japan, particularly in the
setting of hemodialysis (Matsuo et al., 1988). In 1993, exclusive rights to the
compound for the United States and Canada were acquired from Mitsubishi-
Tokyo Pharmaceuticals, Inc. by Texas Biotechnology Corporation (TBC) of
Houston. In 1995, clinical evaluation of this agent for HIT began in the
United States, using a prospective, multicenter, open-label design with
historical controls, the ARG-911 study (Lewis et al., 2001) (see Chap. 16).
Two groups of patients were studied: HIT without thrombosis (i.e., isolated
HIT) and HIT complicated by thrombosis (heparin-induced thrombocyto-
penia/thrombosis syndrome, or HITTS). Eligibility was based on clinical
suspicion of HIT, and serological confirmation of the diagnosis, therefore,
was not required. Both patient groups received the identical therapeutic-dose
regimen of argatroban (initially, 2 Ag/kg/min, then adjusted by activated
partial thromboplastin time [aPTT]). The favorable results of the ARG-911
16 Warkentin
B. Fondaparinux
Fondaparinux (Arixtra) is a synthetic pentasaccharide modeled after the
antithrombin-binding site of heparin. It selectively binds to antithrombin,
causing rapid and specific inhibition of factor Xa. Fondaparinux does not
bind to PF4, and as a corollary, HIT antibodies fail to recognize PF4 mixed
with fondaparinux, both in platelet activation and PF4-dependent antigen
assays.
Preliminary evidence suggests that although HIT antibody formation
occasionally occurs in association with fondaparinux use, such antibodies fail
to react in HIT assays in which fondaparinux replaces UFH or LMWH in
vitro (Warkentin et al., 2003b). Thus, this pentasaccharide anticoagulant
seems unlikely to cause an adverse effect resembling HIT. Although no pa-
tients developed HIT with either LMWH (enoxaparin) or fondaparinux in the
two orthopedic surgery trials reported, the duration of anticoagulant therapy
may have been too brief to reveal a true difference in risk of immune throm-
bocytopenia between LMWH (frequency 0.1–1.0%) and fondaparinux (an-
ticipated negligible frequency). Fondaparinux is approved in the United
States, Canada, and the European Union for antithrombotic prophylaxis in
orthopedic surgery situations (Table 1).
18 Warkentin
C. Bivalirudin
The 20-amino-acid hirudin analogue bivalirudin (Angiomax, formerly, Hir-
ulog) was used 10 years ago in the United States on a compassionate use basis
for the treatment of four patients with HIT (Nand, 1993; Reid and Alving,
1994; Chamberlin et al., 1995). Currently, this agent is approved in the United
States and Canada for anticoagulation of patients with unstable angina
undergoing percutaneous transluminal coronary angioplasty (Table 1) (see
Chap. 16). It is now seeing some off-label use for the treatment of HIT
(Francis et al., 2003). This agent is also being evaluated as an anticoagulant to
permit ‘‘on-pump’’ and ‘‘off-pump’’ cardiac surgery, in patients with or
without HIT (Warkentin and Greinacher, 2003). It is possible that increasing
use of bivalirudin or other nonheparin anticoagulants, e.g., argatroban
(Table 1) or ximelagatran (oral thrombin inhibitor currently under investi-
gation), in diverse clinical situations will result in reduced numbers of patients
developing HIT.
REFERENCES
Alving BM, Shulman NR, Bell WR, Evatt BL, Tack KM. In vitro studies of heparin-
associated thrombocytopenia. Thromb Res 1977; 11:827–834.
Amiral J, Bridey F, Dreyfus M, Vissac AM, Fressinaud E, Wolf M, Meyer D. Platelet
factor 4 complexed to heparin is the target for antibodies generated in heparin-
induced thrombocytopenia [letter]. Thromb Haemost 1992; 68:95–96.
Babcock RB, Dumper CW, Scharfman WB. Heparin-induced thrombocytopenia. N
Engl J Med 1976; 295:237–241.
Barker CF, Rosato FE, Roberts B. Peripheral arterial embolism. Surg Gynecol Obstet
1966; 123:22–26.
Bell WR. Heparin-associated thrombocytopenia and thrombosis. J Lab Clin Med
1988; 111:600–605.
Bell WR, Royall RM. Heparin-associated thrombocytopenia: a comparison of three
heparin preparations. N Engl J Med 1980; 303:902–907.
Bell WR, Tomasulo PA, Alving BM, Duffy TP. Thrombocytopenia occurring during
the administration of heparin. A prospective study in 52 patients. Ann Intern
Med 1976; 85:155–160.
Benhamou AC, Gruel Y, Barsotti J, Castellani L, Marchand M, Guerois C, Leclerc
MH, Delahousse B, Griguer P, Leroy J. The white clot syndrome or heparin
associated thrombocytopenia and thrombosis (WCS or HATT). Int Angiol
1985; 4:303–310.
Best CH. Preparation of heparin, and its use in the first clinical cases. Circulation 1959;
19:79–86.
Chamberlin JR, Lewis B, Wallis D, Messmore H, Hoppensteadt D, Walenga JM,
Moran S, Fareed J, McKiernan T. Successful treatment of heparin-associated
20 Warkentin
Warkentin TE, Kelton JG. Heparin and platelets. Hematol Oncol Clin North Am
1990; 4:243–264.
Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia.
Am J Med 1996; 101:502–507.
Warkentin TE, Kelton JG. Delayed-onset heparin-induced thrombocytopenia and
thrombosis. Ann Intern Med 2001; 135:502–506.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular-
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Warkentin TE, Elavathil LJ, Hayward CPM, Johnston MA, Russett JI, Kelton JG.
The pathogenesis of venous limb gangrene associated with heparin-induced
thrombocytopenia. Ann Intern Med 1997; 127:804–812.
Warkentin TE, Chong BH, Greinacher A. Heparin-induced thrombocytopenia: to-
wards consensus. Thromb Haemost 1998; 79:1–7.
Warkentin TE, Roberts RS, Hirsh J, Kelton JG. An improved definition of immune
heparin-induced thrombocytopenia in postoperative orthopedic patients. Arch
Intern Med 2003a. In press.
Warkentin TE, Cook RJ, Marder VJ, Kelton JG. Comparison of heparin-induced
thrombocytopenia antibody (HIT-Ab) generation and in vitro cross-reactivity
after elective hip or knee replacement surgery in patients receiving antithrom-
botic prophylaxis with fondaparinux or enoxaparin [abstr]. Blood 2003b. In
press.
Weismann RE, Tobin RW. Arterial embolism occurring during systemic heparin
therapy. Arch Surg 1958; 76:219–227.
2
Differential Diagnosis of Acute
Thrombocytopenia
Volker Kiefel
University of Rostock, Rostock, Germany
I. INTRODUCTION
Platelet count is affected by the rate of platelet production, the platelet life
span, and the distribution of platelets among different compartments (Wint-
robe et al., 1981). These three aspects of platelet kinetics have been extensively
studied with platelets radiolabeled with 51Cr (Aster and Jandl, 1964) or 111In
(Heaton et al., 1979).
Platelet production is equivalent to platelet turnover in a ‘‘steady state’’
and can be estimated by determining platelet mean life span and count: ap-
proximately 44 109/L per day of platelets are produced by normal persons
(Branehög et al., 1974). Platelet turnover is decreased in certain marrow dis-
orders (e.g., aplastic anemia and hereditary thrombocytopenia) and often as
a result of cytotoxic drugs used for the therapy of malignant disease. Plate-
let distribution is mainly influenced by spleen size. Normally, approximately
30% of platelets are sequestered in the spleen. In patients with splenomegaly,
this fraction can increase to 90%, and mild to moderate thrombocytopenia
can result. Conversely, in splenectomized subjects, more than 90% of the total
platelet mass is circulating.
This chapter will focus on the differential diagnosis of thrombocy-
topenic states that are characterized by a shortened platelet survival that
is mediated by immune mechanisms. Pathological conditions of enhanced
platelet destruction caused by nonimmune mechanisms will also be briefly dis-
cussed. Autoantibodies responsible for autoimmune thrombocytopenic pur-
pura (AITP), acquired platelet dysfunction, cyclic thrombocytopenia, and
25
26 Kiefel
Antigenic
Number of determinants
Glycoproteins subunits Molecular weight fora
coworkers (1982a) were the first to identify the glycoprotein (GP) IIb/IIIa
complex as the major target antigen recognized by platelet autoantibodies in
AITP, a finding confirmed by others. Epitopes of some antibodies have been
assigned to GP IIIa (Beardsley et al., 1984) or GP IIb (Tomiyama et al., 1987).
The other major target antigen is the platelet GP Ib/IX complex (Woods et al.,
1984; Kiefel et al., 1991). Other ‘‘rare’’ autoantibodies have been shown to
react with GP Ia/IIa (Castaldi et al., 1989), GP V (Mayer and Beardsley,
1996), GMP-140 (CD62), and the thrombopoietin receptor (Malloy et al.,
1995).
B. Clinical Manifestations
In its various manifestations AITP is a relatively common disorder. Different
forms are usually distinguished by clinical criteria.
Chronic AITP
Chronic AITP is the most common form of immune thrombocytopenia in
adults, with women more frequently affected (3:1). The annual incidence has
been estimated at approximately 1:31,000 in adults (Frederiksen and
Schmidt, 1999). It may occur as ‘‘idiopathic’’ immune thrombocytopenia or
as ‘‘secondary AITP’’: that is, observed together with other immunological
diseases, such as systemic lupus erythematosus (Waters, 1992), rheumatoid
arthritis (Hegde et al., 1983), Crohn’s disease (Kosmo et al., 1986), primary
biliary cirrhosis (Panzer et al., 1990); malignant diseases, such as lymphopro-
liferative disorders (Hegde et al., 1983) and solid tumors (Mueller-Eckhardt et
al., 1983a); infectious diseases including viral hepatitis (Pawlotsky et al., 1995;
Ibarra et al., 1986), human immunodeficiency virus (HIV) infection (Van der
Lelie et al., 1987; Walsh et al., 1985); after bone marrow transplantation
(Benda et al., 1989); and in diseases of unknown origin, such as sarcoidosis
28 Kiefel
C. Diagnosis
Clinical diagnosis of idiopathic AITP is based on its typical clinical picture
and the exclusion of other causes for thrombocytopenia: isolated thrombo-
cytopenia without evidence of impaired thrombocytopoiesis (normal num-
bers of megakaryocytes in the bone marrow). Moreover, spleen size is normal
and no other conditions known to enhance platelet clearance are found, such
as disseminated intravascular coagulation (DIC), thrombotic thrombocyto-
penia purpura (TTP), or large hemangiomas characteristic of the Kasa-
bach-Merritt syndrome. Familial thrombocytopenia implies a nonimmune
pathogenesis (Greinacher and Mueller-Eckhardt, 1994), for most patients
with hereditary thrombocytopenia do not have enhanced platelet destruction
(Najean and Lecompte, 1990).
It may be difficult to diagnose AITP on clinical grounds in patients with
malignancy, because immune thrombocytopenia may coexist with spleno-
megaly or neoplastic marrow infiltration. Moreover, not all ‘‘platelet anti-
body tests’’ are diagnostically helpful (George et al., 1996). In particular,
‘‘platelet-associated immunoglobulin G’’ (PAIgG) assays developed as
‘‘platelet Coombs tests’’ focused on technical considerations, rather than
on clinical usefulness (Dixon et al., 1975; Mueller-Eckhardt et al., 1978;
Hegde et al., 1985; Follea et al., 1982; Morse et al., 1981; Kunicki et al., 1982;
Leporrier et al., 1979; McMillan et al., 1979; Court and LoBuglio, 1986;
Kiefel et al., 1987a). However, it appears that platelet-bound IgG in these
assays bears little or no direct relation to immune-mediated platelet destruc-
tion (Mueller-Eckhardt et al., 1982; Kiefel et al., 1986). Rather, PAIgG re-
flects IgG stored in the platelet a-granules (George, 1990). However, with the
advent of platelet glycoprotein-specific assays (McMillan et al., 1987; Kiefel et
Differential Diagnosis of Acute Thrombocytopenia 29
al., 1987b; Kiefel, 1992), it is evident that IgG bound to GPs IIb/IIIa or Ib/IX
is specific for AITP.
D. Therapeutic Considerations
Therapy should be based on the degree of hemorrhagic diathesis observed.
Thus, patients with ‘‘wet purpura’’ are generally treated more aggressively
because they are considered to be at greatest risk for bleeding. In children with
severe acute AITP, it is important to reduce physical activity. Drugs that
inhibit platelet function, such as acetylsalicylic acid, should be avoided. If
therapy is necessary, prednisone at an initial dose of 1 mg/kg body weight
should be given for a limited period. The ivIgG preparations are very effective
in childhood AITP (Imbach et al., 1981, 1985), with doses of 2 g/kg body
weight (0.4 g/kg daily for 5 days or 1 g/kg for 2 days) usually effective for a
limited time in adult patients. Alternatively, blockade of immune phagocy-
tosis with sensitized autologous red blood cells has been proposed (Salama et
al., 1983; Becker et al., 1986): IgG anti-D (Rh0) may be given intravenously to
rhesus (D)-positive patients with AITP. Two doses of approximately 20 Ag/kg
body weight result in an increase in platelets in most patients. Although the
therapeutic effect of anti-D appears less rapidly than with high-dose ivIgG, it
is often more sustained. Immunosuppressive therapy with azathioprine
(Quiquandon et al., 1990) alone or together with corticosteroids may be at-
tempted in patients refractory to other forms of treatment. Refractory pa-
tients with dangerous bleeding complications have been successfully treated
with cyclophosphamide (Reiner et al., 1995). One of the most effective thera-
peutic measures in AITP is splenectomy. It should not be performed in
children younger than 6 years of age and not in the first 6 months of initially
diagnosed acute AITP. It results in partial or complete remissions in 50–80%
of patients (Shulman and Jordan, 1987). Possibly, patients with predomi-
nantly splenic sequestration of platelets have a higher chance of remission
after splenectomy (Najean et al., 1997). A good response to ivIgG may indi-
cate a higher remission rate after splenectomy (Law et al., 1997). Therapy of
AITP has been reviewed (Berchtold and McMillan, 1989; Waters, 1992; Eden
and Lilleyman, 1992; George et al., 1996).
Cyclic Thrombocytopenia
Cyclic thrombocytopenia, which predominantly occurs in women, is charac-
terized by rhythmic fluctuations of platelet counts. These fluctuations are in
phase with the menstrual cycle, lowest platelets counts being observed during
menses. Normal to high platelet counts are observed at midcycle (Tomer et
al., 1989). In many patients with this condition, thrombocytopenia is the re-
sult of accelerated platelet clearance, as determined with 111In-labeled plate-
lets. In two of the three patients described by Tomer, autoantibodies with GP
Ib/IX specificity were identified during both the thrombocytopenic period
and the period with normal platelet counts. These authors correlated platelets
counts with changing densities of the Fcg-receptor on the patients’ autolo-
gous monocytes. In one patient studied by Menitove, IgG anti-GP IIb/IIIa
was found (Menitove et al., 1989). In another case, an IgM anti-GP IIb/IIIa
has been reported (Kosugi et al., 1994). Data from another group suggest that
the pathophysiology underlying the clinical picture of cyclic thrombocyto-
penia may be heterogeneous: an autoimmune form with cyclic changes in
platelet destruction and a distinct condition with cyclic changes in thrombo-
cytopoiesis (Nagasawa et al., 1995).
Onyalai
An exceptionally severe variant of AITP, onyalai, is observed in some black
populations in southern Africa. Whites living in the same regions do not
appear to suffer from this disease. In a series of 103 patients (Hesseling, 1987),
all patients presented with hemorrhagic bullae of the mucous membranes of
the oropharynx. Six died, four of cerebral hemorrhage and two of hemor-
rhagic shock. Clinical diagnosis is based on the criteria of AITP and, in
addition, to the presence of hemorrhagic bullae (Hesseling, 1992). Antibodies
against GP IIb/IIIa—often of the IgM class—have been implicated. The bone
marrow contains normal counts of megakaryocytes, but patients may be
anemic at presentation owing to blood loss. Therapeutic options are discussed
elsewhere (Hesseling, 1992).
Differential Diagnosis of Acute Thrombocytopenia 31
Original Percent
Alloantigen designation positivea Localization Refs.
B. Clinical Picture
The clinical course of PTP has been summarized by the European PTP study
group (Mueller-Eckhardt et al., 1991). Women were predominantly affected
(99 out of 104). Mean age was 58.4 years. In 28 of 51 patients, marked febrile
transfusion reactions were observed following the transfusion that precipi-
tated PTP. The interval between transfusion and onset of severe thrombocy-
topenia was generally 6–10 days. In 68 out of 84 patients, the initial platelet
count was fewer than 10 109/L. Bleeding persisted for 3–37 days (mean, 10.2
days). Most patients required treatment. Fatal bleeding was not rare, occur-
ring in 7 of 75 patients (Shulman and Jordan, 1987) and in 2 of 38 patients
(Kroll et al., 1993) in two studies.
C. Diagnosis
The diagnosis of PTP should be considered in all patients with a sudden drop
in platelet count to fewer than 10 109/L. Thus, PTP and HIT may occur in
similar clinical situations, (i.e., about 1 week after surgery). In PTP, how-
ever, thrombocytopenia is more pronounced and associated with bleeding
(Lubenow et al., 2000), in contrast to the absence of petechiae and presence of
thromboembolic complications characteristic of HIT (see Chap. 3).
Moreover, PTP only occurs following recent transfusion. PTP has
occurred in association with delayed hemolytic transfusion reaction (Chap-
man et al., 1987; Maslanka and Zupanska, 1993). In a single case, PTP was
observed concurrently with drug-dependent immune hemolytic anemia
(Mueller-Eckhardt et al., 1987). Diagnosis is confirmed by detection of a
platelet-specific alloantibody against an epitope on platelet GP IIb/IIIa,
usually anti-HPA-1a. Platelets of the patient are always negative for the
corresponding antigen. Eluates prepared from patients’ autologous platelets
usually contain anti-HPA-1a (Kroll et al., 1993).
D. Therapeutic Considerations
The efficacy of corticosteroids is uncertain. In contrast, high-dose ivIgG
(Mueller-Eckhardt et al., 1983b) is clearly effective in most (Berney et al.,
Differential Diagnosis of Acute Thrombocytopenia 33
1985; Chong et al., 1986; Mueller-Eckhardt and Kiefel, 1988) but not all cases
(Kroll et al., 1993). Thus, ivIgG is the treatment of choice for PTP. Platelet
transfusions are ineffective, even if platelets from HPA-1a-negative donors
are given (Gerstner et al., 1979). As the bleeding tendency is often pro-
nounced, immediate therapy following clinical diagnosis is mandatory.
Drug n Comments
Quinidine 18
Quinine 3
Quinidine + quinine 5
Trimethoprim–sulfamethoxazole 6 1 metabolite—specific
ddAb (sulfamethoxazole)
Rifampicin (rifampin) 4
Nomifensine 2
Paracetamol (acetaminophen) 1 1 metabolite—specific ddAb
Carbamazepine 4
Diclofenac 5
Ibuprofen 3 1 metabolite—specific ddAb
Ranitidine 1
Vancomycin 1
Source: Institute for Clinical Immunology and Transfusion Medicine, University of Giessen.
Differential Diagnosis of Acute Thrombocytopenia 35
B. Clinical Disorders
Septicemia
Disseminated intravascular coagulation can complicate infections, especially
with gram-negative bacteria (Marder et al., 1994; Mammen, 1998). It has been
suggested that thrombocytopenia in septic patients is the consequence of
immune-mediated platelet damage, based on the observation of elevated
PAIgG levels (Kelton et al., 1979). However, this does not prove an autoim-
mune basis for the thrombocytopenia (Shulman and Reis, 1994), for PAIgG is
often also elevated in thrombocytopenia of nonimmune origin.
The pathogenesis of DIC in sepsis is multifactorial and includes direct
endothelial damage and platelet activation by endotoxins, resulting in expo-
sure of procoagulant material. In addition, the cytokines interleukin-1 and
tumor necrosis factor increase tissue factor activity, thereby shifting the bal-
Differential Diagnosis of Acute Thrombocytopenia 37
Malignant Disease
About 9–15% of patients with cancer have DIC at some point during their
disease (Pasquini et al., 1995). Overt bleeding is uncommon; rather, recur-
rent thromboembolism is characteristic, an entity known as Trousseau’s syn-
drome. An exception: acute promyelocytic leukemia is often accompanied
by a severe DIC and bleeding, often induced or worsened by chemotherapy
(Marder et al., 1994).
Other Conditions
Disseminated intravascular coagulation occurs in obstetric situations char-
acterized by release of thrombogenic material [e.g., the retained dead fetus
syndrome (Marder et al., 1994; Baglin, 1996), amniotic fluid embolism, or
placental separation]. Bites of certain snakes may cause hypofibrinogenemia
induced by enzymes that clot fibrinogen or directly activate platelets. Severe
hemolytic transfusion reactions can cause DIC, especially in association with
red cell antibodies, causing intravascular complement-mediated hemolysis
(e.g., ABO-incompatible transfusion). DIC seems to be aggravated by
complement-mediated damage of endothelial cells. Whether red cell lysis
alone (not mediated by complement) is able to induce DIC in humans remains
unclear (Mollison et al., 1993; Baglin, 1996). Other conditions associated with
DIC are trauma and localized processes in which activation of coagulation
occurs within giant hemangiomas (Kasabach-Merritt syndrome) or aortic
aneurysms.
C. Diagnosis
‘‘Global’’ coagulation tests, such as prothrombin and activated partial
thromboplastin times, are usually prolonged; fibrinogen concentrations are
often reduced. However, these parameters can be normal in DIC. Fibrin
degradation products mirror the action of plasmin on fibrin clots, and there-
fore are elevated in most patients with DIC. The D-dimer test readily detects
cross-linked fibrin degradation products. Elevated prothrombin fragment
F1+2 levels reflect thrombin activation as one of the central mechanisms
underlying DIC. Examination of a blood smear sometimes will show red cell
fragmentation in DIC. On the other hand, a high percentage of red cell frag-
38 Kiefel
Drug or
Platelets metabolite Serum Reaction Interpretation
a
+ + Patient + Drug-dependent antibody
+ Patient b
+ + Normal donor serum b
+ Normal donor serum b
+ + Patient + Autoantibody
+ Patient + (alloantibody)
+ + Normal donor serum
+ Normal donor serum
REFERENCES
Allford SL, Hunt BJ, Rose P, Machin SJ. Guidelines on the diagnosis and man-
agement of the thrombotic microangiopathic haemolytic anaemias. Br J Hae-
matol 2003; 120:556–573.
Differential Diagnosis of Acute Thrombocytopenia 43
tients treated with the oral glycoprotein IIb/IIIa inhibitors xemilofiban and
orbofiban: evidence for an immune etiology. Thromb Haemost 2002; 88:892–
897.
Brunner-Bollinger S, Kiefel V, Horber FF, Nydegger UE, Berchtold P. Antibody
studies in a patient with acute thrombocytopenia following infusion of plasma
containing anti-P1Al. Am J Hematol 1997; 56:119–121.
Castaldi PA, Mehrabani PA, Fournier DJ, Berndt MC. Autoimmune thrombocyto-
penia associated with B-CLL and an IgG autoantibody directed against the
human platelet GP Ia-IIa complex (VLA-2) [abstr]. Thromb Haemost 1989;
62:151.
Chapman JF, Murphy MF, Berney SI, Ord J, Metcalfe P, Amess JAL, Waters AH.
Post-transfusion purpura associated with anti-Bak1 and anti-PlA2 platelet anti-
bodies and delayed hemolytic transfusion reaction. Vox Sang 1987; 52:313–317.
Chong BH, Cade J, Smith JA, Tatoulis J. An unusual case of post-transfusion pur-
pura: good transient response to high-dose immunoglobulin. Vox Sang 1986;
51:182–184.
Christie DJ, Mullen PC, Aster RH. Fab-mediated binding of drug-dependent
antibodies to platelets in quinidine- and quinine-induced thrombocytopenia. J
Clin Invest 1985; 75:310–314.
Christie DJ, Mullen PC, Aster RH. Quinine- and quinidine platelet antibodies can
react with GP IIb/IIIa. Br J Haematol 1987; 67:213–219.
Christie DJ, Pulkrabek S, Putnam JL, Slatkoff ML, Pischel KD. Posttransfusion
purpura due to an alloantibody reactive with glycoprotein Ia/IIa (anti-HPA-
5b). Blood 1991; 77:2785–2789.
Christie DJ, Sauro SC, Cavanaugh AL, Kaplan ME. Severe thrombocytopenia in an
acquired immunodeficiency syndrome patient associated with pentamidine-de-
pendent antibodies specific for glycoprotein IIb/IIIa. Blood 1993; 82:3075–3080.
Court WS, LoBuglio AF. Measurement of platelet surface-bound IgG by a mono-
clonal 125I-anti-IgG assay. Vox Sang 1986; 50:154–159.
Crosby WH. Wet purpura, dry purpura. JAMA 1975; 232:744–745.
Curtis BR, Swyers J, Divgi A, McFarland JG, Aster RH. Thrombocytopenia after
second exposure to abciximab is caused by antibodies that recognize abciximab-
coated platelets. Blood 2002; 99:2054–2059.
Dixon R, Rosse WF, Ebbert L. Quantitative determination of antibody in idiopathic
thrombocytopenic purpura. Correlation of serum and platelet-bound antibody
with clinical response. N Engl J Med 1975; 292:230–236.
Eden OB, Lilleyman JS. Guidelines for management of idiopathic thrombocytopenic
purpura. Arch Dis Child 1992; 67:1056–1058.
Eisner EV, Kasper K. Immune thrombocytopenia due to a metabolite of para-
aminosalicylic acid. Am J Med 1972; 53:790–796.
Eisner EV, Shahidi NT. Immune thrombocytopenia due to a drug metabolite. N Engl J
Med 1972; 287:376–381.
Follea G, Mandrand B, Dechavanne M. Simultaneous enzymo-immunologic assays of
platelet associated IgG, IgM, and C3. A useful tool in assessment of immune
thrombocytopenias. Thromb Res 1982; 26:249–258.
Differential Diagnosis of Acute Thrombocytopenia 45
Herman JH, Kickler TS, Ness PM. The resolution of platelet serologic problems using
Western blotting. Tissue Antigens 1986; 28:257–268.
Hesseling PB. Onyalai in Namibia. Clinical manifestations, haematological findings,
course and management of 103 patients in the Kavango territory. Trans R Soc
Trop Med Hyg 1987; 81:193–196.
Hesseling PB. Onyalai. Baillieres Clin Haematol 1992; 5:457–473.
Huisman JG. Immunoblotting: an emerging technique in immunohematology. Vox
Sang 1986; 50:129–136.
Humphries JE. Transfusion therapy in acquired coagulopathies. Hematol Oncol Clin
North Am 1994; 8:1181–1201.
Ibarra H, Zapata C, Inostroza J, Mezzano S, Riedemann S. Immune thrombocyto-
penic purpura associated with hepatitis A. Blut 1986; 52:371–375.
Imbach P, Barandun S, d’Apuzzo V, Baumgartner C, Hirt A, Morell A, Rossi E,
Schöni M, Vest M, Wagner HP. High-dose intravenous gammaglobulin for
idiopathic thrombocytopenic purpura in childhood. Lancet 1981; 1:1228–1231.
Imbach P, Berchtold W, Hirt A, Mueller-Eckhardt C, Rossi E, Wagner HP, Gaedicke
G, Joller P, Müller B, Barandun S. Intravenous immunoglobulin versus oral
corticosteroids in acute thrombocytopenic purpura in childhood. Lancet 1985;
2:464–468.
Jubelirer SJ, Koenig BA, Bates MC. Acute profound thrombocytopenia following
C7E3 Fab (abciximab) therapy: case reports, review of the literature and impli-
cations for therapy. Am J Hematol 1999; 61:205–208.
Keimowitz RM, Collins J, Davis K, Aster RH. Post-transfusion purpura associated
with alloimmunization against the platelet-specific antigen, Baka. Am J Hematol
1986; 21:79–88.
Kelton JG, Neame PB, Gauldie J, Hirsh J. Elevated platelet-associated IgG in the
thrombocytopenia of septicemia. N Engl J Med 1979; 300:760–764.
Kelton JG, Smith JW, Horsewood P, Humbert JR, Hayward CPM, Warkentin TE.
Gova/Govb alloantigen system on human platelets. Blood 1990; 75:2172–2176.
Kereiakes DJ, Essell JH, Abbottsmith CW, Broderick TM, Runyon JP. Abciximab-
associated profound thrombocytopenia: therapy with immunoglobulin and
platelet transfusion. Am J Cardiol 1996; 78:1161–1163.
Kickler TS, Herman JH, Furihata K, Kunicki TJ, Aster RH. Identification of Bakb, a
new platelet-specific antigen associated with posttransfusion purpura. Blood
1988; 71:894–898.
Kiefel V. The MAIPA assay and its applications in immunohematology. Transfusion
Med 1992; 2:181–188.
Kiefel V, Spaeth P, Mueller-Eckhardt C. Immune thrombocytopenic purpura: auto-
immune or immune complex disease? Br J Haematol 1986; 64:57–68.
Kiefel V, Jäger S, Mueller-Eckhardt C. Competitive enzyme-linked immunoassay for
the quantitation of platelet-associated immunoglobulins (IgG, IgM, IgA) and
complement (C3c, C3d). Vox Sang 1987a; 53:151–156.
Kiefel V, Santoso S, Weisheit M, Mueller-Eckhardt C. Monoclonal antibody-specific
immobilization of platelet antigens (MAIPA): a new tool for the identification
of platelet reactive antibodies. Blood 1987b; 70:1722–1726.
Differential Diagnosis of Acute Thrombocytopenia 47
Theodore E. Warkentin
McMaster University and Hamilton Regional Laboratory Medicine Program,
Hamilton, Ontario, Canada
I. INTRODUCTION
53
54 Warkentin
Estimated frequencies of the various complications of HIT are taken from reports with serological con-
firmation of the diagnosis (Warkentin et al., 1995; Warkentin and Kelton, 1996; Warkentin et al., 1997).
‘‘Rare’’ indicates an estimated frequency <3% of HIT patients.
II. THROMBOCYTOPENIA
A. Timing
The characteristic delay of 5 or more days between initiation of heparin and
onset of thrombocytopenia was the major clue that led early investigators to
Clinical Picture of HIT 55
Figure 1 Continued.
58 Warkentin
HIT during low molecular weight heparin (LMWH) therapy. More time may
be required to generate clinically important levels of HIT-IgG so as to activate
platelets in the presence of PF4/LMWH, rather than PF4/H, complexes.
Figure 3 A 49-year-old patient exhibiting both typical- and rapid-onset HIT: The
platelet count began to fall on day 6 of subcutaneous (sc) UFH injections given for
antithrombotic prophylaxis following neurosurgery (typical HIT). An abrupt fall in
platelet count occurred twice on day 18, each after a 5000 U intravenous (iv) UFH
bolus (rapid HIT). Symptoms and signs of acute systemic reaction occurred 10 minutes
after each bolus (dyspnea, tachypnea, hypertension, chest tightness, restlessness). Note
that the patient’s platelet count never fell below 150 109/L, even though her serum
tested strongly positive for HIT antibodies by serotonin release assay. She developed
proximal deep venous thrombosis (DVT) shortly after developing HIT.
bodies again; those who do appear to form antibodies after day 5 (Gruel et al.,
1990; Warkentin and Kelton, 2001a). Indeed, several patients with well-
documented previous HIT have received full treatment courses of heparin
several months or years later without incident (Warkentin and Kelton, 2001a;
Lindhoff-Last et al., 2002).
Figure 4 Proportion of patients with HIT antibodies after an episode of HIT. The
time (in days) to a negative test by the activation assay (n = 144) or the antigen assay
(n = 93) is shown. The antigen test tended to become negative more slowly than did the
activation assay ( p = 0.007). (From Warkentin and Kelton, 2001a.)
62 Warkentin
whose clinical course suggests HIT, especially if the patient was recently hos-
pitalized or has undergone procedures in which heparin exposure may have
occurred.
Figure 5 Delayed onset of HIT: a 68-year-old woman who received UFH for heart
surgery was noted to have a platelet count of 40 109/L on postoperative day 19 and a
‘‘rash’’ of her lower extremities. She presented on day 38 with symptomatic DVT and
developed rapid-onset recurrent thrombocytopenia after receiving iv UFH. The pa-
tient was successfully treated with danaparoid sodium (D.S.) and warfarin. In retro-
spect, the thrombocytopenia first observed on postoperative day 19 almost certainly
was caused by delayed onset of HIT.
Clinical Picture of HIT 65
platelet count fell over the next 4 days to 14 109/L, along with laboratory
evidence for DIC (low fibrinogen and elevated fibrin D-dimer levels). This
patient’s platelet counts gradually recovered to normal over several months,
during which time recurrent thrombotic events were managed successfully
with an alternative anticoagulant.
The unusual clinical course of these patients could be related to very
high titers of platelet-activating IgG antibodies (Warkentin and Kelton,
2001b). Moreover, substantial platelet activation in vitro can be caused by
some of these patients’ sera even in the absence of added heparin. This find-
ing of substantial heparin-independent platelet activation resembles that
described in other patients with drug-induced immune thrombocytopenia,
in which prolonged thrombocytopenia has been reported in association with
drug-independent binding of IgG to platelets (Kelton et al., 1981). Given the
apparent rarity of these cases, it is perhaps surprising that this syndrome does
not occur more frequently, given that HIT—once initiated—resembles some-
what an autoimmune disorder, with IgG recognizing an autologous protein,
PF4. On the other hand, earlier discharge from the hospital and a higher
index of suspicion for this syndrome might mean that delayed onset of HIT
will become a relatively more common presentation of HIT in the future.
Delayed onset of HIT, however, should not be confused with delayed
clinical manifestation of HIT-associated thrombosis. For example, Fig. 3
shows a patient who developed typical onset of HIT while receiving postop-
erative heparin prophylaxis. However, isolated HIT was not clinically
recognized, and the patient presented subsequently with a DVT and a normal
platelet count; when heparin boluses were given, rapid onset of thrombocy-
topenia occurred. Presumably, subclinical HIT-associated DVT that began
during the episode of isolated HIT progressed to symptomatic thrombosis in
the absence of anticoagulation. In contrast, patients with delayed onset of
HIT develop thrombocytopenia several days after the use of heparin and
are thrombocytopenic when they present with thrombosis. Exacerbation of
thrombocytopenia occurs if further heparin is given.
The existence of delayed onset of HIT presents a diagnostic dilemma in
patients who are no longer receiving heparin but who develop thrombocyto-
penia 5 or more days after placement of a heparin-coated device, e.g., certain
intravascular grafts or stents (Cruz et al., 2003). Such a puzzling situation of
delayed onset of thrombocytopenia post–vascular surgery prompted inves-
tigators to postulate heparin contamination of a graft (the manufacturer
insisted otherwise) (Bürger et al., 2001). In my view, either delayed onset or a
protracted course of thrombocytopenia could reflect the generation and
persistence of unusual ‘‘autoimmune’’ HIT antibodies without the need to
invoke continuing exposure to heparin.
66 Warkentin
B. Severity of Thrombocytopenia
Figure 6 shows the platelet count nadirs of 142 patients with laboratory-
proved HIT in one medical community: the median platelet count nadir was
approximately 60 109/L (Warkentin, 1998a). This contrasts with ‘‘typical’’
drug-induced immune thrombocytopenic purpura (DITP; e.g., caused by
quinine/quinidine, sulfa antibiotics, or rifampin [see Chap. 2]), for which the
median platelet count nadir is 15 109/L or less, and patients usually develop
bleeding (Pedersen-Bjergaard et al., 1997). The platelet count is 15 109/L or
Figure 6 Platelet count nadirs in 142 patients with serologically confirmed HIT: the
data are taken from a study of 127 patients with serologically confirmed HIT that used
a definition of <150 109/L (Warkentin and Kelton, 1996), together with a group of
15 patients diagnosed with serologically confirmed HIT over the same time period
whose platelet count nadir was >150 109/L. There is a lognormal distribution of the
platelet count nadirs, with a median platelet count of 59 109/L. HIT is only occa-
sionally complicated by very severe thrombocytopenia. HIT-associated thrombosis
occurred in most patients irrespective of the severity of the platelet count nadir. For
comparison, schematic platelet count nadir distributions are shown for typical drug-
induced immune thrombocytopenic purpura (DITP) and atypical DITP caused by
abciximab. (Modified from Warkentin, 1998a.)
Clinical Picture of HIT 67
fewer in only about 5% of patients with HIT (Warkentin, 2003). But even in
this minority of HIT patients with very severe thrombocytopenia, thrombo-
sis, rather than bleeding, predominates. Patients with atypical drug-induced
thrombocytopenic purpura caused by anti-GPIIb/IIIa-blocking drug (e.g.,
abciximab [ReoPro]) appear to develop severity of thrombocytopenia inter-
mediate between that of typical DITP and HIT (Fig. 6).
Definition of Thrombocytopenia
Figure 6 illustrates that HIT is associated with thrombosis even when the
platelet count nadir is more than 150 109/L. This suggests that the standard
definition of thrombocytopenia (<150 109/L) may be inadequate for many
patients with HIT. Particularly in postoperative patients, a major fall in the
platelet count can occur without the nadir falling to less than 150 109/L (see
Figs. 1b and 3). Indeed, studies indicate that a 50% or greater fall in the plate-
let count from the postoperative peak is strongly associated with HIT anti-
bodies, even when the platelet count nadir remains higher than 150 109/L
(Ganzer et al., 1997; Warkentin et al., 2003). Moreover, this patient subgroup
is at increased risk for thrombosis.
Rule 3
A platelet count fall of more than 50% from the postoperative peak
between days 5 and 14 after surgery associated with heparin treatment
can indicate HIT even if the platelet count remains higher than 150
109/L.
It is possible that a greater than 50% platelet count fall definition is also
appropriate for medical patients (Girolami et al., 2003). Regardless of the
patient population, a clinician should have a high index of suspicion when
unexpected large-percentage declines in the platelet count occur during hepa-
rin treatment, irrespective of whether an arbitrary absolute threshold for
‘‘thrombocytopenia’’ is crossed.
Heparin UFH Line Nil Nil UFH 5000 UFH 5000 UFH 5000 UFH 5000 D.S D.S D.S D.S
used during flushes b.i.d. sc b.i.d. sc b.i.d. sc b.i.d. sc
CPB
Platelet 227 98 137 209 255 300 374 378 310 224 (PEa) 166 171 161 (nadir)
count
Percent Platelet fall during day 0–4 is unlikely to be HIT unless there was Rising Peak 18% 41% 56% No 57%
platelet recent heparin use (past 100 days) and the magnitude of the platelet platelet (378 ! (378 ! (378 ! further (378 !
count platelet fall is greater than expected. count count 310) 224) 166) fall 161)
fall
a
Pulmonary embolism (PE) occurred on postoperative day 8, in association with a platelet count fall of 41%, from 378 (postoperative peak) to 224 109/L.
The platelet count began to fall on day 7. The case illustrates why it is wrong to use the preoperative platelet count value as the ‘‘baseline,’’ as the fall in platelet
count from 227 (preoperative) to 224 (day 7) would be considered trivial, even though HIT-associated pulmonary embolism occurred. The preoperative and
first three postoperative days are in shaded boxes to indicate that these data should be censored in the interpretation of platelet counts in HIT. In this patient,
the abrupt fall in platelet count from 227 to 98 (day 0) is expected (heart surgery). This patient was treated successfully with danaparoid sodium (D.S.), with
longer-term anticoagulation with warfarin. The patient’s clinical course is also shown in Fig. 4B in Chap. 12.
Figure 1 Pathogenesis of HIT: a central role for thrombin generation. (See Fig.
13.1, p. 340 for full legend.)
Figure 2 Model of the interaction between argatroban and thrombin. (See Fig.
16.2, p. 440.)
Figure 3 (a) Model of the human platelet factor 4 tetramer. (b) AC dimer view of
the amino acids (‘‘ring of charge’’) crucial for heparin binding. [C-terminal a-helix
residues encompassing lysines 61–62 and 65–66 (cyan), arginines 20, 22, and 49
(green), and histidine 23, threonine 25, lysine 46 (yellow).] (See Fig. 7.2, p. 185, for
full legend. From a, Zhang et al., 1994; b, Loscalzo et al., 1985; Mayo et al., 1995a.)
III. THROMBOSIS
A. The HIT Paradox: Thrombosis but Not Hemorrhage
Table 1 summarizes the clinical spectrum and approximate frequency of
clinical sequelae associated with HIT. Spontaneous hemorrhage is not
characteristic of HIT, and petechiae are not typically observed, even in those
occasional patients whose platelet count is less than 10 109/L. Bleeding
complications were not increased over controls in two prospective studies of
HIT (Cipolle et al., 1983; Warkentin et al., 1995).
Rule 4
Petechiae and other signs of spontaneous bleeding are not clinical fea-
tures of HIT, even in patients with very severe thrombocytopenia.
The explanation for this clinical feature is unknown, but could be
related to unique pathophysiological aspects of HIT, such as in vivo platelet
activation, generation of procoagulant, platelet-derived microparticles,
and procoagulant alterations of endothelium and monocytes (see Chaps. 9
and 10).
Odds ratio
Hypercoagulable state for thrombosis
Heparin-induced thrombocytopenia:
Platelet count <150 109/L 36.9
Platelet fall >50% beginning z5 days of heparin 12.4
Platelet fall >50%, but platelet count remains >150 109/L 6.0
Factor V Leiden 6.6
Congenital protein C deficiency 14.4
Congenital protein S deficiency 10.9
Congenital antithrombin deficiency 24.1
Dysfibrinogenemia 11.3
Lupus anticoagulant 5.4
Source: Warkentin, 1995; Warkentin et al. 1995, 2003.
About 5% of patients (3 of 62) in the largest study died suddenly, two with
proved or probable pulmonary embolism (Warkentin and Kelton, 1996).
This experience supports the recommendation that further anticoagulation
be considered for patients in whom isolated HIT has been diagnosed (Hirsh
et al., 1998, 2001; Warkentin et al., 1998) (see Chaps. 1 and 13–16).
E. Venous Thrombosis
Large case series suggest that venous thrombotic complications predominate
in HIT (Warkentin and Kelton, 1996; Nand et al., 1997) (see Table 4 in Chap.
4). Indeed, pulmonary embolism occurs more often than all arterial throm-
botic events combined. Furthermore, the strength of association between
HIT and venous thromboembolism increases in relation to the severity of
thrombosis (Table 4). Other unusual venous thrombotic events complicating
HIT include cerebral vein (dural sinus) thrombosis (v.i.), hepatic vein throm-
bosis (Theuerkauf et al., 2000), and perhaps retinal vein thrombosis (Nguyen
et al., 2003). Thus:
Rule 7
In patients receiving heparin, the more unusual or severe a subsequent
thrombotic event, the more likely the thrombosis is caused by HIT.
Regardless of the severity of thrombosis, in any patient who develops a
symptomatic venous or arterial thrombosis while receiving heparin, the plate-
let count should be measured to evaluate whether HIT could be present.
Post–orthopedic Proximal DVT 8/18 (44.4%) 26/647 (4.0%) 19.1 (5.9–58.3) <0.001
surgerya (Warkentin Bilateral proximal DVT 2/18 (11.1%) 4/647 (0.6%) 20.1 (1.7–150) 0.01
et al., 1995, 2003) Pulmonary embolism 2/18 (11.1%) 2/647 (0.3%) 40.3 (2.7–572) 0.004
Any thrombosis 13/18 (72.2%) 112/647 (17.3%) 12.4 (4.0–45.2) <0.001
Patients with central lineb Upper-limb DVT 14/145 (9.7%) 3/484 (0.6%) 17.1 (4.9–60.5) <0.001
(Hong et al., 2003)
Medicala (Girolami et al., Any thrombosis 3/5 (60%) 21/593 (3.5%) 40.8 (5.2–163) <0.001
2003)
a
HIT defined as >50% platelet count fall.
b
HIT defined as any abnormal platelet count fall with positive HIT serology (platelet fall was >50% in 93% of study patients).
Warkentin
Clinical Picture of HIT 75
Figure 8 Coumarin-induced skin necrosis (CISN): HIT is associated with two forms
of CISN: (1) venous limb gangrene, affecting extremities with active deep vein throm-
bosis, and (2) ‘‘classic’’ CISN, which involves central (nonacral) tissues, such as breast,
abdomen, thigh, flank, and leg, among other tissue sites. CISN complicating HIT
typically manifests as venous limb gangrene (f90%) (Warkentin et al., 1997, 1999),
whereas CISN in other settings most commonly affects central tissues (f90%) (Cole
et al., 1988). (From Warkentin, 1996b.)
rhagic infarction typically presents with ipsilateral flank pain without signs of
adrenal failure (Warkentin, 1996a). HIT explained at least 5% of patients
with adrenal hemorrhage at one institution (Vella et al., 2001).
This hemorrhagic manifestation of HIT is caused by thrombosis of
adrenal veins leading to hemorrhagic necrosis of the glands. Other hy-
percoagulable states associated with adrenal necrosis include dissem-
inated intravascular coagulation (DIC) complicating meningococcemia
(Waterhouse-Friderichsen syndrome) and the antiphospholipid antibody
syndrome (McKay, 1965; Carette and Jobin, 1989).
Figure 12 Clinical manifestations of DIC. (a) Livedo reticularis. (b) Patchy ischemic
necrosis of right foot. This 70-year-old woman developed HIT-associated DIC with
hypofibrinogenemia, elevated INR, and reduced antithrombin and protein C activity
levels 9 days after emergency cardiac surgery for cardiac catheterization-associated
dissection of the left main coronary artery (see text for additional clinical information).
(See color insert, Figs. 6c and 6d.)
Clinical Picture of HIT 83
F. Arterial Thrombosis
Lower limb artery thrombosis was the first recognized complication of HIT
(Weismann and Tobin, 1958; Roberts et al., 1964; Rhodes et al., 1973, 1977).
84 Warkentin
Arterial thrombosis most commonly involves the distal aorta (e.g., saddle
embolism) or the large arteries of the lower limbs, leading to acute limb
ischemia with absent pulses. Sometimes, platelet-rich thromboemboli from
the left heart or proximal aorta explain acute lower limb arterial ischemia
(Vignon et al., 1996). Other arterial thrombotic complications that are
relatively common in HIT include acute thrombotic stroke and myocardial
infarction. The relative frequency of arterial thrombosis in HIT by location,
namely, lower limb artery occlusion >> stroke syndrome > myocardial in-
farction (Benhamou et al., 1985; Kappa et al., 1987; Warkentin and Kelton,
1996; Nand et al., 1997), is reversed from that observed in the non-HIT
population (myocardial infarction > stroke syndrome >> lower limb artery
occlusion).
Uncommon but well-described arterial thrombotic events in HIT in-
clude mesenteric artery thrombosis (bowel infarction), brachial artery throm-
bosis (upper limb gangrene), and renal artery thrombosis (renal infarction).
Multiple arterial thrombotic events are quite common, as are recurrences
following surgical thromboembolectomy, especially if further heparin is given
during or after surgery. Occasionally, microembolization of thrombus orig-
inating from the heart or aorta causes foot or toe necrosis with palpable arte-
rial pulses.
Angiographic Appearance
Lindsey and colleagues (1979) reported a distinct angiographic appearance of
heparin-induced thromboembolic lesions, described as ‘‘broad-based, isolat-
ed, gently lobulated excrescences which produced 30–95% narrowing of the
arterial lumen. The abrupt appearance of such prominent luminal contour
deformities in arterial segments that were otherwise smooth and undistorted
was unexpected and impressive. . . . In each case, the lesions were located
proximal to sites of arterial occlusion.’’ The radiologic and surgical experi-
ence described suggests that distal embolization of ‘‘white’’ clots composed of
‘‘platelet-fibrin aggregates’’ accounted for the limb ischemia.
should check for unexpected platelet count declines, and test for HIT anti-
bodies, in patients who develop thrombosis of grafts, prostheses, or other
devices during heparin treatment.
Histopathology
Lymphocyte infiltration of the upper and middermis that can extend into the
epidermis characterizes the erythematous plaque (Bircher et al., 1990).
Dermal and epidermal edema (spongiosis) is observed in lesions that appear
eczematous. The T lymphocytes of helper–suppressor (CD4+) phenotype
86 Warkentin
Management
Heparin-induced skin lesions should be considered a marker for the HIT
syndrome. Platelet count monitoring, if not already being performed, should
be initiated and continued for several days, even after stopping heparin
administration. The reason is that some patients develop a fall in platelet
count, together with thrombosis (often affecting limb arteries), that begins
several days after stopping the heparin (Warkentin, 1996a, 1997). An alter-
native anticoagulant, such as danaparoid, lepirudin, or argatroban, should be
given, particularly in patients whose original indication for anticoagulation
still exists or who develop progressive thrombocytopenia. The skin lesions
themselves should be managed conservatively whenever possible, although
some patients require debridement of necrotic tissues followed by skin graft-
ing (Hall et al., 1980).
Rule 9
Erythematous or necrotizing skin lesions at heparin injection sites should
be considered dermal manifestations of the HIT syndrome, irrespective
of the platelet count, unless proved otherwise. Patients who develop
thrombocytopenia in association with heparin-induced skin lesions are
at increased risk for venous and, especially, arterial thrombosis.
The clinical features of postheparin bolus ASR are not typical of IgE-
mediated anaphylaxis (i.e., urticaria, angioedema, and hypotension are not
seen). Rather, the syndrome resembles febrile transfusion reactions com-
monly observed after platelet transfusions, suggesting a common pathogen-
esis of proinflammatory cytokines associated with cellular activation (Heddle
et al., 1994). Moreover, there are similarities between ASR and the admin-
istration of ADP in humans, including acute dyspnea, tachycardia, and tran-
sient thrombocytopenia (Davey and Lander, 1964).
A few patients have developed acute, transient impairment of antero-
grade memory (i.e., the ability to form new memories) following an intrave-
nous heparin bolus in association with acute HIT (Warkentin et al., 1994;
Pohl et al., 2000). This syndrome resembles that of transient global amnesia,
a well-characterized neurological syndrome of uncertain pathogenesis.
E. Heparin Resistance
Difficulty in maintaining therapeutic anticoagulation despite increasing hep-
arin dosage, or heparin resistance, is a common finding in patients with HIT-
associated thrombosis (Rhodes et al., 1977; Silver et al., 1983). Possible
explanations include neutralization of heparin by PF4 released from activated
platelets (Padilla et al., 1992) or pathophysiological consequences of platelet-
derived microparticles (Bode et al., 1991). Heparin resistance is not specific
for HIT, however, and occurs in many patients with extensive thrombosis of
various etiologies (e.g., cancer).
Clinical Picture of HIT 91
B. HIT in Pregnancy
Heparin-induced thrombocytopenia has complicated UFH treatment given
for venous thromboembolism complicating pregnancy (Van der Weyden
et al., 1983; Meytes et al., 1986; Copplestone and Oscier, 1987; Greinacher
et al., 1992) or the postpartum period (Calhoun and Hesser, 1987). HIT seems
to be rare in this patient population; no pregnant patients have been
diagnosed with HIT over a 20-year period in Hamilton. Plasma glycosami-
noglycans are increased during pregnancy (Andrew et al., 1992), which could
contribute to lower frequency or pathogenicity of HIT antibodies. HIT
antibodies cross the placenta (Greinacher et al., 1993), so it is at least
theoretically possible that a heparin-treated newborn delivered from a mother
with acute HIT could develop this drug reaction.
Pregnant patients with HIT have developed unusual events, such as
cerebral dural sinus thrombosis (Van der Weyden et al., 1983; Calhoun and
Hesser, 1987). Treatment options for pregnant patients with life-threatening
thrombosis include danaparoid or fondaparinux as these drugs do not cross
the placenta (see Chaps. 13 and 14). The more benign syndrome of heparin-
induced skin lesions without thrombocytopenia has also been reported in
pregnant patients (Drouet et al., 1992). Danaparoid was reported to be
effective in a patient who developed LMWH-induced skin lesions (de Saint-
Blanquat et al., 2000).
Cardiological complications
Myocardial infarction (Rhodes et al., 1973; Van der Weyden et al., 1983)
Occlusion of saphenous vein grafts post–coronary artery bypass surgerya
Intra-atrial thrombus (left and rightb heart chambers) (Scheffold et al., 1995;
Olbrich et al., 1998)
Intraventricular thrombus (left and rightb heart chambers) (Commeau et al., 1986;
Dion et al., 1989; Vignon et al., 1996)
Prosthetic valve thrombosis (Bernasconi et al., 1988; Vazquez-Jimenez et al., 1999)
Right heart failure secondary to massive pulmonary embolism
Cardiac arrest postintravenous heparin bolus (Ansell et al., 1986; Platell and Tan,
1986; Hewitt et al., 1998)
Neurological complications
Stroke syndrome
In situ thrombosis
Progressive stroke in patients receiving heparin for treatment of stroke
(Ramirez-Lassepas et al., 1984)
Cardiac embolization (Scheffold et al., 1995)
Cerebral vein (dural venous sinus) thrombosis (Van der Weyden et al., 1983;
Kyritsis et al., 1990; Meyer-Lindenberg et al., 1997; Warkentin and Bernstein,
2003); complicating pregnancy (Calhoun and Hesser, 1987)
Amaurosis fugax (Theuerkauf et al., 2000)
Ischemic lumbosacral plexopathy (Jain, 1986)
Paraplegia, transient (Maurin et al., 1991) or permanent (Feng et al., 1993),
associated with distal aortic thrombosis
Transient global amnesia (Warkentin et al., 1994)
Headachec
a
Thrombosis preferentially affects saphenous vein grafts rather than internal mammary artery
grafts (Liu et al., 2002; Ayala et al., 2002).
b
Although adherent thrombi that likely developed in situ have been reported (Dion et al.,
1989), emboli originating from limb veins can explain right-sided intra-atrial or intra-
ventricular clots.
c
Headache as a feature of HIT is suggested by (1) its occurrence in patients with acute systemic
reactions post–heparin bolus (see Fig. 1a, Chap. 4) and (2) its concurrence with onset of
thrombocytopenia in several patients who developed HIT in a clinical trial (unpublished ob-
servations of the author).
recent study of 108 neonates who received UFH flushes found no HIT anti-
bodies using a sensitive antigen assay (Klenner et al., 2003).
B. The ‘‘
‘‘Four Ts’’
’’
A new scoring system, the ‘‘4 Ts,’’ has been developed that takes advantage of
new information regarding the clinical features of HIT (Warkentin, 2003;
Warkentin and Heddle, 2003). Platelet count recovery is not a criterion,
because this information often is not available at initial evaluation, or heparin
may not have been stopped. For simplicity, four clinical features are assessed,
given scores of 0, 1, or 2 (Table 7). Thus, the maximal total score is 8.
94
2 1 0
9
Thrombocytopenia (acute) Nadir, 20–100 (at least Nadir, 10–19 10 /L or Nadir, <10 109/L
30% fall); or any >50% any 30–50% fall (or or any <30% fall
fall (nadir z20) >50% fall associated
with heart surgery)
Timingb of platelet count Clear onset between days Consistent with day 5–10 Platelet count fall V4
fall, thrombosis, or other 5–10 or V1 day (if fall, but not clear (e.g., days without recent
sequelae (first day of heparin heparin exposure within missing platelet counts) or heparin exposure
course = day zero) past 30 days) V1 day (heparin exposure
within past 31–100 days)
or platelet fall after day 10
Thrombosis or other sequelae New thrombosis; skin Progressive or recurrent None
(e.g., skin lesions, ASR) necrosis; ASR after iv thrombosis; erythematous
heparin bolus skin lesions; suspected
thrombosis (not yet proven);
asymptomatic upper-limb
DVT
Other cause of No explanation (besides HIT) Possible other cause Definite other cause is
thrombocytopenia for platelet count fall is evident is evident present
not evident
Abbreviations: ASR, acute systemic reaction (see Table 5); DVT, deep venous thrombosis.
a
Pretest probability score: 6–8 = high; 4–5 = intermediate; 0–3 = low.
b
First day of immunizing heparin exposure considered day zero; the day the platelet count begins to fall is considered the day of onset of
thrombocytopenia (it generally takes 1–3 more days until an arbitrary threshold that defines thrombocytopenia is passed). The scoring system
shown here has undergone minor modifications from previously published scoring systems (Warkentin, 2003; Warkentin and Heddle, 2003).
Warkentin
Clinical Picture of HIT 95
Estimated pretest probabilities of HIT thereby range from low (0–3) to high
(6–8), with an intermediate score (4–5) indicating moderate risk.
Maximal scores for each category are given when the clinical features
are highly consistent with HIT. Thus, a patient will score 8 if there is a
substantial fall in the platelet count that begins 5–10 days after commencing
heparin, together with thrombosis, and where no other plausible cause is
apparent during clinical assessment. Even a patient with no clinical evidence
of thrombosis can be assigned a high pretest probability (score 6 of 8) if the
clinical features are otherwise consistent with HIT. Another feature of this
system is that very low platelet count values (i.e., 10–19 and<10 109/L)
score only 1 and 0 points, respectively, thus reducing the chance that a patient
with posttransfusion purpura (PTP) or DITP will be misclassified as HIT and
inappropriately given anticoagulant therapy.
REFERENCES
Ansell JE, Clark WP Jr, Compton CC. Fatal reactions associated with intravenous
heparin [letter]. Drug Intell Clin Pharm 1986; 20:74–75.
Arthur CK, Grant SJB, Murray WK, Isbister JP, Stiel JN, Lauer CS. Heparin-
associated acute adrenal insufficiency. Aust NZ J Med 1985; 15:454–455.
Ayala E, McDonough RF, Morgensztern D, Kharfan-Dabaja MA, Byrnes JJ. Heparin-
induced thrombocytopenia presenting with thrombosis of multiple saphenous
vein grafts (abstr). Blood 2002; (suppl):3894.
Balestra B, Quadri P, Demarmels Biasiutti F, Furlan M, Lämmle B. Low molecular
weight heparin-induced thrombocytopenia and skin necrosis distant from in-
jection sites. Eur J Haematol 1994; 53:61–63.
Beland B, Busse H, Loick HM, Ostermann H, van Aken H. Phlegmasia cerulea dolens,
cerebral venous thrombosis, and fatal pulmonary embolism due to heparin-
induced thrombocytopenic thrombosis syndrome. Anesth Analg 1997; 85:1272–
1274.
Benhamou AC, Gruel Y, Barsotti J, Castellani L, Marchand M, Guerois C, Leclerc
MH, Delahousse B, Griguer P, Leroy J. The white clot syndrome or heparin-
associated thrombocytopenia and thrombosis (WCS or HATT). Int Angiol
1985; 4:303–310.
Bernasconi F, Metivet F, Estrade G, Garnier D, Donatien Y. Thrombose d’une pros-
thèse valvulaire mitrale au cours d’une thrombopénie induite par l’héparine.
Traitement fibrinolytique Presse Méd 1988; 17:1366.
Betrosian AP, Theodossiades G, Lambroulis G, Kostantonis D, Balla M, Papani-
kolaou M, Georgiades G. Heparin-induced thrombocytopenia with pulmonary
embolism and disseminated intravascular coagulation associated with low-
molecular-weight heparin. Am J Med Sci 2003; 325:45–47.
Bircher AJ, Flückiger R, Buchner SA. Eczematous infiltrated plaques to subcutaneous
heparin: a type IV allergic reaction. Br J Dermatol 1990; 123:507–514.
Bircher AJ, Itin PH, Büchner SA. Skin lesions, hypereosinophilia, and subcutaneous
heparin [letter]. Lancet 1994; 343:861.
Bleasel JF, Rasko JEJ, Rickard KA, Richards G. Acute adrenal insufficiency sec-
ondary to heparin-induced thrombocytopenia–thrombosis syndrome. Med J
Aust 1992; 157:192–193.
Bode AP, Castellani WJ, Hodges ED, Yelverton S. The effect of lysed platelets on
neutralization of heparin in vitro with protamine as measured by the activated
coagulation time (ACT). Thromb Haemost 1991; 66:213–217.
Boshkov LK, Warkentin TE, Hayward CPM, Andrew M, Kelton JG. Heparin-
induced thrombocytopenia and thrombosis: clinical and laboratory studies. Br J
Haematol 1993; 84:322–328.
Bowers MJ, Jones FGC. Thrombus in harvested marrow from a patient with recent
heparin-induced thrombocytopenia. Br J Haematol 2002; 119:294.
Broekmans AW, Bertina RM, Loeliger EA, Hofmann V, Klingemann HG. Protein C
and the development of skin necrosis during anticoagulant therapy [letter].
Thromb Haemost 1983; 49:251.
Brushwood DB. Hospital liable for allergic reaction to heparin used in injection flush.
Am J Hosp Pharm 1992; 49:1491–1492.
Bürger T, Tautenhahn J, Böck M, Fahlke J, Halloul Z, Lippert H. Can a coated
Clinical Picture of HIT 97
Kovacs KA, Lam YM, Pater JL. Bilateral massive adrenal hemorrhage. Assessment
of putative risk factors by the case-control method. Medicine (Balt) 2001;
80:45–53.
Kyritsis AP, Williams EC, Schutta HS. Cerebral venous thrombosis due to heparin-
induced thrombocytopenia. Stroke 1990; 21:1503–1505.
Laster J, Cikrit D, Waler N, Silver D. The heparin-induced thrombocytopenia syn-
drome: an update. Surgery 1987; 102:763–770.
Lee DH, Warkentin TE, Denomme GA, Lagrotteria DD, Kelton JG. Factor V Leiden
and thrombotic complications in heparin-induced thrombocytopenia. Thromb
Haemost 1998; 79:50–53.
Lindhoff-LastE,WenningB,SteinM, GerdsenF,BauersachsR,WagnerR.Riskfactors
and long-term follow-up of patients with the immune type of heparin-induced
thrombocytopenia. Clin Appl Thrombosis/Hemostasis 2002; 8:347–352.
Lindsey SM, Maddison FE, Towne JB. Heparin-induced thromboembolism: angio-
graphic features. Radiology 1979; 131:771–774.
Ling E, Warkentin TE. Intraoperative heparin flushes and acute heparin-induced
thrombocytopenia. Anesthesiology 1998; 89:1567–1569.
Lipton ME, Gould D. Case report: heparin-induced thrombocytopenia—a compli-
cation presenting to the vascular radiologist. Clin Radiol 1992; 45:137–138.
Liu JC, Lewis BE, Steen LH, Grassman ED, Bakhos M, Blakeman B, Wrona L, Leya
F. Patency of coronary artery bypass grafts in patients with heparin-induced
thrombocytopenia. Am J Cardiol 2002; 89:979–981.
Lubenow N, Kempf R, Eichner A, Eichler P, Carlsson LE, Greinacher A. Heparin-
induced thrombocytopenia: temporal pattern of thrombocytopenia in relation
to initial use or reexposure to heparin. Chest 2002; 122:37–42.
MacLean JA, Moscicki R, Bloch KJ. Adverse reactions to heparin. Ann Allerg 1990;
65:254–259.
Makhoul RG, Greenberg CS, McCann RL. Heparin-associated thrombocytopenia
and thrombosis: a serious clinical problem and potential solution. J Vasc Surg
1986; 4:522–528.
Markel A, Manzo RA, Bergelin RO, Strandness DE Jr. Pattern and distribution of
thrombi in acute venous thrombosis. Arch Surg 1992; 127:305–309.
Maurin N, Biniek R, Heintz B, Kierdorf H. Heparin-induced thrombocytopenia and
thrombosis with spinal ischaemia—recovery of platelet count following a
change to a low molecular weight heparin [letter]. Intensive Care Med 1991;
17:185–186.
McKay DG. Late manifestations of intravascular coagulation—tissue necrosis. In:
McKay DG, ed. Disseminated Intravascular Coagulation. An Intermediary
Mechanism of Disease. New York: Harper & Row, 1965:392–471.
Meyer-Lindenberg A, Quenzel E-M, Bierhoff E, Wolff H, Schindler E, Biniek R. Fata
cerebral venous sinus thrombosis in heparin-induced thrombotic thrombocy-
topenia. Eur Neurol 1997; 37:191–192.
Meytes D, Ayalon H, Virag I, Weisbort Y, Zakut H. Heparin-induced thrombocy-
topenia and recurrent thrombosis in pregnancy. A case report. J Reprod Med
1986; 31:993–996.
102 Warkentin
David H. Lee
Queen’s University, Kingston, Ontario, Canada
Theodore E. Warkentin
McMaster University and Hamilton Regional Laboratory Medicine Program,
Hamilton, Ontario, Canada
I. INTRODUCTION
Biological explanations
1. Patient population studied (frequency of HIT antibody formation differs
among patient populations, possibly because of differences in platelet
activation and PF4 release)
2. Type of heparin used (UFH more immunogenic than LMWH; bovine lung
heparin more immunogenic than porcine mucosal heparin; possibly, lot-to-lot
variability in immunogenicity of heparin)
3. Variable duration of heparin treatment (HIT typically begins between days 5
and 10)
4. Dose of heparin used (dose-dependent thrombocytopenia)
Technical explanations
1. Variable definition of thrombocytopenia used
2. Differing baseline platelet counts permitted for study entry
3. Requirement to repeat platelet count testing to confirm thrombocytopenia
4. Variable intensity of platelet count surveillance
5. Variable intensity of surveillance for thrombotic events
6. Failure to exclude nonimmune heparin-associated thrombocytopenia
a. Lack of use of in vitro test for HIT antibodies
b. Use of insensitive or nonspecific HIT antibody assays
c. Inclusion of patients with ‘‘early’’ thrombocytopenia
d. Failure to exclude patients whose platelet count recovered during continued
heparin treatment
e. Failure to exclude patients with other explanations for thrombocytopenia
Table 2 The Frequency of HIT: Prospective Studies of HIT in Medical Patients Using In Vitro Testing of Patient Serum/Plasma
for HIT Antibodies or Indicating a High Likelihood of HIT Based on Timing of Platelet Count Fall
Frequency of (immune) HIT (%)
Major Timing of Definition of
indication In vitro Route, Bovine Porcine platelet fall thrombocytopenia
Study for heparin test dose UFH UFH LMWH reported? (109/L)
Comparisons between bovine UFH and porcine UFH [studies and data in bold are randomized, controlled trials (RCTs)]
Ansell et al., 1980 VTE PRP(SR) iv ther 4/21 (19.0) 0/22 (0) Yes <150
(RCT)
Green et al., 1984 VTE HIPA iv ther 2/45 (4.4) 0/44 (0) Yes <150
(RCT)
Powers et al., 1984 VTE SRAa iv ther 2/65a (3.1) 0/66 (0) Yes <150
(RCT)
Bailey et al., 1986 VTE, ATE No test iv ther 1/21 (4.8) 0/22 (0) Yes <100
(RCT)
Cipolle et al., 1983; VTE, ATE PRP iv ther 6/100b (6.0) 1/111 (0.9) Yes <100
Ramirez-Lassepas [stroke] [3/54] (5.6) (1/83) (1.2)
et al., 1984 [stroke]
Predominant treatment for venous (VTE) or arterial (ATE) thromboembolism
Bell et al., 1976
Alving et al., 1977 VTE, ATE PRP/SRAc iv ther 3/52c (5.8) Yes <100
Powers et al., 1979 VTE No test iv ther 2/120d (1.7) Yesd <150
Gallus et al., 1980 VTE PRP iv ther 3/166e (1.8) Yes <100
sc proph 0/5 (0)
Holm et al., 1980 VTE No test iv ther 0/90f (0) Yes <100
Monreal et al., 1989 VTE No test iv ther 2/89 (2.2) Low <100
sc proph 0/49 (0) 0/43 (0) platelets
Lee and Warkentin
day 8
Kakkasseril et al., VTE, ATE PRP iv ther 4/142g (2.8) No <100
1985
Malcolm et al., 1979 Multiple PRP iv ther 1/66h (1.5) Yes <100
sc proph 0/38 (0)
Rao et al., 1989 Multiple PRP(SR) iv ther 0/94 (0) NA <100
sc proph 0/99 (0)
Girolami et al., 2003 VTE, ATE HIPA, EIA sc proph 5/360 (1.4) Yes >50% fall
Frequency of HIT
Schwartz et al., 1985 — No test Bolus iv 3/20 (15.0) 0/10 (0) Yes <150
ther
HIT was excluded if the platelet count rose during continued heparin after an early fall (e.g., Johnson et al., 1984). Also, where uncertainty existed
as to the number of patients with probable HIT, the lower number was indicated in the table, to avoid overestimating the number of patients with
HIT (contrast the analysis shown in Table 4). Some data relating to Cipolle et al. (1983) were obtained by personal communication, as reported
(Warkentin and Kelton, 1991). The study by Gallus et al. (1980) was excluded because the source of heparin was not specified. Some reports (e.g.,
Nelson et al., 1978) were excluded because timing of thrombocytopenia was not reported.
Abbreviations: ATE, arterial thromboembolism; EIA, PF4-heparin enzyme-immunoassay; HIPA, heparin-induced platelet activation test; iv ther,
intravenous therapeutic-dose heparin; LMWH, low molecular weight heparin; MI/ACS, myocardial infarction/acute coronary syndromes; PRP,
HIT assay using citrated platelet-rich plasma (PRP/SR, with serotonin release); sc proph, subcutaneous prophylactic-dose heparin; sc ther,
subcutaneous therapeutic; RCT, randomized controlled trial; SRA, serotonin release assay using washed platelets; UFH, unfractionated heparin;
VTE, venous thromboembolism.
a
Powers et al. (1984) described five patients who developed thrombocytopenia during bovine UFH use (none during porcine UFH use); two
patients whose platelet counts fell beginning on day 7 (to nadir of 41 109/L) and on day 8 (to nadir of 53 109/L) had positive testing for HIT
antibodies by SRA; test results are not available for one patient whose platelet count fell on day 5 (excluded from Table 2, but included in Table 4);
one patient with proved HIT developed progression of deep venous thrombosis, as indicated in Table 4 (laboratory records of Dr. J. G. Kelton).
b
Cipolle et al. (1983) described ten patients who received bovine UFH who may have had HIT; only six are included here, all of whom developed a
platelet count fall between days 7 and 10. Ramirez-Lassepas et al. (1984) examined the subgroup with underlying cerebrovascular disease and
reported six patients who developed thrombocytopenia after receiving bovine heparin. Only the three who developed late thrombocytopenia are
reported here.
c
Only 8 of 16 thrombocytopenia patients underwent PRP testing (all negative) and only 5 underwent SRA testing. The 3 who developed late
thrombocytopenia (HIT) had negative PRP testing, but did not have SRA testing.
d
Information on timing of platelet count fall to determine the likelihood of HIT was obtained by personal communication, as described (Warkentin
and Kelton, 1991).
e
Origin of heparin is uncertain and may have used mucosal heparin from sheep; at least 3, and as many as 5, patients appeared to have HIT based
on in vitro testing and timing of platelet count fall.
f
One case of early thrombocytopenia due to DIC reported in original paper was excluded.
Lee and Warkentin
g
At least four, and as many as nine, patients appeared to have HIT; the four that tested positive for HIT antibodies are included here.
h
One patient appeared to have HIT based on positive heparin rechallenge, despite negative in vitro HIT test.
i
Thrombocytopenic patients with negative EIA testing were excluded.
Table 3 The Frequency of HIT: Prospective Studies of Surgical Patients Using Confirmatory In Vitro Laboratory Testing
of Patient Serum or Plasma or Indicating a High Likelihood of HIT Based on Timing of Platelet Count Fall
b
Of seven patients with thrombocytopenia reported, two were excluded because of early onset of thrombocytopenia.
116 Lee and Warkentin
influencing the frequency of HIT include the type of heparin used and the
patient population.
also been observed in several other clinical trials of patients receiving UFH
following orthopedic surgery (see Warkentin et al., 1995, for discussion), but
only one study (Leyvraz et al., 1991) reported confirmatory in vitro testing.
There is little prospective information of the frequency of HIT in other
postoperative surgical populations treated with UFH (see Table 3). Three
studies have been performed on postoperative cardiac surgical patients who
also received postoperative UFH in addition to high doses of heparin during
preceding cardiopulmonary bypass (Trossaert et al., 1998; Warkentin et al.,
2000; Pouplard et al., 1999) (see Table 3). Pooling the three studies, about
2.4% of the patients developed serologically confirmed HIT. Interestingly the
frequency of HIT in this population appears to be lower than in orthopedic
patients receiving UFH, even though the cardiac surgical patients appear to
have a higher frequency of formation of HIT antibodies (Warkentin et al.,
2000).
Isolated limb perfusion (ILP) with melphalan employs extracorporeal
circulation (and thus high-dose UFH) to treat melanoma or unresectable
sarcoma limited to an extremity. In one study, HIT occurred in 3 of 108
patients (2.8%), who also received subcutaneous UFH prophylaxis following
ILP (Masucci et al., 1999). The occurrence of arterial thrombosis and partial
limb amputation in two of these patients with HIT led the investigators to
discontinue routine UFH prophylaxis post-ILP. The hypothesis that ILP is a
high-risk situation for HIT was supported by a prospective study showing
HIT antibody seroconversion in nine of nine patients who underwent this
procedure (despite not receiving postoperative UFH prophylaxis), with eight
patients having HIT antibodies ‘‘strong’’ enough to cause serotonin release.
(2001) found no cases of HIT in 624 pregnancies among 604 women treated
with LMWH.
Table 4 Proportion of Patients with HIT Developing HIT-Associated Thrombosis in Prospective Studies Employing In Vitro
Laboratory Testing or in Which Data on Timing of Platelet Count Fall Were Reported
Patients with
HIT (using Thrombotic
Major Definition of more sensitive complication of HIT
indication In vitro Number thrombocytopenia definition
Study for heparin test treated (109/L) of HIT) Venous Arterial
>50% fall)
Yamamoto et al., 1996 Hemodialysis PRP, EIA 154 Clotting, 5 0 1
platelet fall
Total medical: Venous/arterial thrombosis ratio = 9/7 = 1.3 1472 41 9 7
Orthopedic surgical patients (total joint arthroplasty)
Warkentin et al., 1995, Hip SRA 332 <150 9 7 1
2003a
(>50% fall) (18) (12) (1)
Frequency of HIT
Where there was uncertainty over the numbers of patients with HIT, the higher estimated value was indicated in the table, to minimize the bias
toward a high frequency of HIT-associated thrombosis (contrast analysis shown in Table 2).
Abbreviations: ATE, arterial thromboembolism; EIA, PF4-heparin enzyme-linked immunosorbent assay; HIPA, heparin-induced platelet ac-
tivation test (aggregation of washed platelets); LMWH, low molecular weight heparin; MI/ACS, myocardial infarction or acute coronary
syndromes; PRP, HIT assay using citrated platelet-rich plasma (PRP/SR, with serotonin release); SRA, serotonin release assay using washed
platelets; UFH, unfractionated heparin; VTE, venous thromboembolism.
a
Detailed clinical data on thrombosis were available only on the subset of patients with cerebrovascular disease (n = 137).
b
Ineffective testing was used (platelet aggregation without heparin).
c
Another five patients developed venous thrombosis in association with a positive HIPA assay, but the platelet count did not fall by >50%.
125
126 Lee and Warkentin
UFH used unless HIT assay Number of with HIT Patients with
Study Trial design otherwise indicated) used patients antibodies (%) HIT (%)
Medical patients
Amiral et al., 1996 Retrospective iv ther UFH EIA-IgM/A/G 19 (17.4)
109 1 (0.9)a
EIA-Ig 3 (2.8)
Kappers-Klunne Prospective iv ther UFH EIA-IgG 9 (2.5) 2 (0.6)
358
et al., 1997 HIPA 30 (8.4) 0 (0)
Hemodialysis patients
Greinacher et al., 1996 Prevalence study iv ther UFH HIPA 165 7 (4.2%) 0 (0)
de Sancho et al., 1996 Prevalence study iv ther UFH EIA-IgM/A/G 45 0 (0) 0 (0)
EIA-IgM/G 4 (3.1)
iv ther UFH 128 0 (0)b
f EIA-IgG 3 (2.3)
Boon et al., 1996 Prevalence study EIA-IgM/G 1 (0.8)
LMWH 133 0 (0)b
f EIA-IgG 1 (0.8)
Luzzatto et al., 1998 Prevalence study iv ther UFH EIA-IgG 50 6 (12.0) 0 (0)
Orthopedic postoperative surgical patients
Warkentin et al.,
sc proph UFH EIA-IgG 29 (14.1)
205 10 (4.9)
1995, 2000 Substudy of RCT SRA 19 (9.3)
sc proph LMWH EIA-IgG 11 (6.0)
182 2 (1.1)
SRA 5 (2.7)c
Warkentin et al., 2000 Prospective sc proph LMWH EIA-IgG 22 (8.6)
257 2 (0.8)
SRA 9 (3.5)c
Amiral et al., 1996 Retrospective sc proph LMWH EIA-IgM/A/G 8 (8.0)
100 0 (0)
EIA-IgG 2 (2.0)
Cardiac postoperative surgical patients (all received porcine UFH at cardiopulmonary bypass except where otherwise stated)
Visentin et al., 1996 Retrospective CPB: UFH EIA-IgM/G 44 27 (61.4)d
0 (0)
(NPH) EIA-IgG 23 (52.3)
Bauer et al., 1997 Prospective CPB: bovine UFH EIA-IgM/A/G 111 57 (51.4)e
0 (0)
(NPH) SRA 23 (52.3)
Trossaert et al., 1998 Retrospective CPB: UFH EIA-IgM/A/G 51 14 (27.5)f
Lee and Warkentin
0 (0)
sc proph UFH EIA-IgG 9 (17.6)
PRP 2 (3.9)
EIA-IgM/A/G 46 (29.3)
Pouplard et al., 1999 Prospective sc proph UFH EIA-IgG 157 24 (15.3) 6 (3.8)
6 (3.8)
SRA
EIA-IgM/A/G 37 (21.6)
sc proph LMWH EIA-IgG 171 24 (14.0) 0 (0)
SRA 2 (1.2)
Warkentin et al., 2000 Prospective CPB: UFH EIA-IgG 50 (50.0)
100 1 (1.0)
Frequency of HIT
Abbreviations: CPB, cardiopulmonary bypass; EIA, PF4-heparin enzyme-linked immunosorbent assay (-IgM/A/G, one or more of IgM, IgA,
and IgG antibodies present; -IgG, IgG antibodies only present); HIPA, heparin-induced platelet activation assay; iv ther, intravenous therapeutic-
dose heparin; LMWH, low molecular weight heparin; MI, myocardial infarction; RCT, randomized controlled trial; sc proph, subcutaneous
prophylactic dose heparin: SRA, serotonin release assay using washed platelets; UFH, unfractionated heparin; VTE, venous thromboembolism,
NPH, no postoperative heparin.
a
Thrombocytopenia defined as platelet count fall >50% from baseline.
b
Two patients with HIT antibodies had mild thrombocytopenia, but a causal relation to heparin was not stated.
c
20% serotonin release used in Warkentin et al. (2000) rather than 50% serotonin release cutoff used in Warkentin et al. (1995).
d
Ten patients had HIT-IgG preoperatively: incidence of new seroconversion was 17/44 (38.6%).
e
Incidence of new seroconversion was 43% for EIA and 9% for SRA.
f
Two patients had HIT-IgG preoperatively: incidence of new seroconversion was 12/51 (23.5%) for EIA-IgM/A/G and 7/51 (13.7%) for EIA-IgG.
None had a positive aggregation assay preoperatively.
g
80.2% underwent CPB, remainder ‘‘off-pump’’ surgery; 18.8% received postoperative UFH, LMWH, or both.
h
Excludes patients testing positive for HIT antibodies at baseline.
i
Includes only HIPA patients who also tested positive in EIA.
j
131
Two patients had HIT-IgG preoperatively: incidence of new seroconversion was 1/54 (1.9%).
132 Lee and Warkentin
Figure 3 Variable frequency of HIT antibody formation and clinical HIT among
different patient populations treated with UFH or LMWH. A schematic ‘‘iceberg,’’
shown on lower line, illustrates the relation among HIT-associated thrombosis,
thrombocytopenia, HIT antibodies detected by serotonin release assay (SRA), and
HIT antibodies detected by enzyme-immunoassay (EIA). The size of the iceberg re-
flects the relative frequency of HIT antibody formation by EIA (i.e., the cardiac-UFH
iceberg is about six times larger than the orthopedic-LMWH iceberg [50 vs. 8%
frequency of HIT antibody formation]). Noteworthy aspects include the observation
that HIT-associated thrombosis is most common in orthopedic-UFH patients, even
though HIT antibody formation is most common in cardiac-UFH patients, and the
observation that orthopedic-LMWH has a higher frequency of thrombosis than does
medical-UFH.
B. Anti-Xa–Inhibiting Pentasaccharide
Fondaparinux (Arixtra) is a novel antithrombin-binding pentasaccharide
that inhibits factor Xa without inhibiting thrombin (factor IIa). It has been
shown to be safe and effective for antithrombotic prophylaxis following
136 Lee and Warkentin
orthopedic surgery. Fondaparinux does not bind to PF4, and therefore its use
might avoid HIT. In systematic studies of HIT antibody formation associated
with fondaparinux or enoxaparin prophylaxis after elective hip or knee re-
placement therapy, low frequencies of HIT antibody formation were seen
with both anticoagulants (Warkentin et al., 2003b). However, whereas HIT
antibodies invariably ‘‘cross-reacted’’ with enoxaparin, they failed to recog-
nize PF4 mixed with fondaparinux (by fluid-phase EIA) (see Chap. 11).
Thus, fondaparinux offers the possibility of absent or negligible risk of
causing HIT.
regular platelet count monitoring for HIT. One explanation is the almost
ubiquitous use of heparin in hospitalized patients. Thus, a requirement that
regular, perhaps even daily, platelet count monitoring be performed seems
excessive. Additionally, there is no convincing evidence that regular platelet
count monitoring can prevent the thrombotic complications of HIT if the
physician response is merely to stop the heparin (Wallis et al., 1999).
However, a worthy consideration is that instituting alternative, parenteral
anticoagulation may prevent thrombosis in patients recognized as having
isolated HIT.
These comments notwithstanding, marked differences in risk for HIT
are apparent among different patient populations. Thus, it seems prudent to
recommend that patients at the highest risk of HIT, and for HIT-associated
thrombosis (e.g., postoperative patients receiving UFH, or any patient
receiving bovine lung UFH) should have platelet counts monitored regularly,
perhaps at least every other day. For patients whose risk for HIT appears to
be 0.1–1% (e.g., medical patients receiving UFH, surgical patients receiving
LMWH), less frequent monitoring may be appropriate. Since HIT is unlikely
to occur before day 5, or after day 14, the monitoring could be performed 2 or
3 times per week from days 4 to 14. Most patients have frequent complete
blood counts performed during the first few days of hospitalization, so
comparative platelet count results for days 0–3 are usually available.
Two recent consensus conferences have examined the issue of platelet
count monitoring for HIT (Warkentin, 2002; Warkentin and Greinacher,
2004). Although the recommendations were not identical, they had in common
the concept of stratifying the intensity of platelet count monitoring based upon
the risk of developing HIT and focusing the monitoring during the time when
HIT usually occurs. Table 8 summarizes draft recommendations (Warkentin
and Greinacher, 2004).
Regardless of the intensity of surveillance, all physicians who monitor
platelet counts need to understand how to distinguish HIT from nonimmune
HAT, because diagnostic confusion may lead to inappropriate decisions to
discontinue heparin therapy in patients with nonimmune HAT who otherwise
require anticoagulation because of high risk for thrombosis. Irrespective of
whether platelet count monitoring is being performed, HIT should be
considered promptly in the differential diagnosis of any patient who develops
symptoms or signs of new, progressive, or recurrent thrombosis during or
within a few days of discontinuing heparin treatment.
ACKNOWLEDGMENTS
Studies described in this chapter were supported by grants from the Heart
and Stroke Foundation of Ontario. Dr. Warkentin was a Research Scholar of
Frequency of HIT 139
the Heart and Stroke Foundation of Canada. Dr. Lee was supported by a
Research Fellowship from the Heart and Stroke Foundation of Canada.
REFERENCES
tionated heparin during open heart surgery are at high risk to form antibodies
reactive with heparin: platelet factor 4 complexes. J Lab Clin Med 1996;
128:376–383.
Vitoux JF, Roncato M, Hourdbhaigt P, Aiach M, Fiessinger J-N. Heparin-induced
thrombocytopenia and pentosan polysulfate: treatment with a low molecular
weight heparin despite in vitro platelet aggregation [letter]. Thromb Hemost
1985; 55:294–295.
Wallis DE, Workman DL, Lewis BE, Steen L, Pifarre R, Moran JF. Failure of early
heparin cessation as treatment for heparin-induced thrombocytopenia. Am J
Med 1999; 106:629–635.
Walls JT, Curtis JJ, Silver D, Boley TM, Schmaltz RA, Nawarawong W. Heparin-
induced thrombocytopenia in open heart surgical patients: sequelae of late
recognition. Ann Thorac Surg 1992a; 53:787–791.
Walls JT, Boley TM, Curtis JJ, Silver D. Heparin-induced thrombocytopenia in
patients undergoing intra-aortic balloon pumping after open heart surgery.
ASAIO J 1992b; 38:M574–M576.
Warkentin TE. Limitations of conventional treatment options for heparin-induced
thrombocytopenia. Semin Hematol 1998; 35(suppl 4):17–25.
Warkentin TE. Platelet count monitoring and laboratory testing for heparin-induced
thrombocytopenia: recommendations of the College of American Patholo-
gists. Arch Pathol Lab Med 2002; 126:1415–1423.
Warkentin TE. Pork or beef ? Ann Thorac Surg 2003; 75:15–16.
Warkentin TE, Greinacher A. Heparin-induced thrombocytopenia: recognition,
treatment, and prevention. Chest 2004. In press.
Warkentin TE, Kelton JG. Heparin-induced thrombocytopenia. Prog Hemost
Thromb 1991; 10:1–34.
Warkentin TE, Kelton JG. Interaction of heparin with platelets, including heparin-
induced thrombocytopenia. In: Bounameaux H, ed. Low-Molecular-Weight
Heparins in Prophylaxis and Therapy of Thromboembolic Diseases. New
York: Marcel Dekker, 1994:75–127.
Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia.
Am J Med 1996; 101:502–507.
Warkentin TE, Kelton JG. Temporal aspects of heparin-induced thrombocytopenia.
N Engl J Med 2001; 344:1286–1292.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Warkentin TE, Elavathil LJ, Hayward CPM, Johnston MA, Russett JI, Kelton JG.
The pathogenesis of venous limb gangrene associated with heparin-induced
thrombocytopenia. Ann Intern Med 1997; 127:804–812.
Warkentin TE, Chong BH, Greinacher A. Heparin-induced thrombocytopenia:
towards consensus. Thromb Haemost 1998a; 79:1–7.
Warkentin TE, Ling E, Ho A, Sheppard JI. ‘‘Incidental’’ unfractionated heparin
(UFH) vs normal saline (NS) flushes for intraoperative invasive catheters
and the frequency of formation of heparin-induced thrombocytopenia IgG
148 Lee and Warkentin
I. INTRODUCTION
Almost as soon as heparin was introduced into clinical medicine, the new drug
was reported to cause immediate small, but consistent, reductions in platelet
count (Sappington, 1939). Later it was also found to produce platelet dys-
function (Heiden et al., 1977), accounting for at least some of its hemorrhagic
risk (Hirsh, 1984; John et al., 1993). These effects, which most likely result
from direct contact between the sulfated glycosaminoglycans and platelets,
are distinct from the role heparin plays in immune-mediated heparin-induced
thrombocytopenia (HIT). However, direct heparin–platelet binding is critical
in the pathogenesis of HIT as well (Horne and Hutchison, 1998). Therefore,
the various ‘‘nonimmune’’ heparin–platelet interactions will be reviewed.
Table 1 Platelet Binding Parameters for Heparin Fractions of Different Molecular Mass
Because of its high charge density, as well as the high linear flexibility
confered by its constituent L-iduronic acid residues, heparin binds readily
to a variety of basic plasma proteins, which theoretically could compete with
platelets for heparin (Casu et al., 1988; Young et al., 1994). However, heparin
binding to only two plasma proteins, antithrombin and fibronectin, interferes
with heparin-induced platelet activation (Salzman et al., 1980; Chong and
Ismail, 1989) or with binding of heparin to platelets (Horne and Chao, 1990).
V. NONIMMUNE HEPARIN-ASSOCIATED
THROMBOCYTOPENIA (NONIMMUNE HAT)
Heparin also binds to several proteins that are secreted from stimulated
platelets. One of these, platelet factor 4 (PF4), binds heparin with unusually
high affinity (Capitanio et al., 1985; Loscalzo et al., 1985; Horne, 1993).
Unlike antithrombin or fibronectin, however, PF4 does not prevent heparin
Nonimmune Heparin–Platelet Interactions 155
from binding to platelets. In fact, it appears that heparin can bind to platelets
in complexes with PF4 (Horne and Hutchison, 1998).
Heparin-bound PF4 has been identified as the primary target of the
antibodies characteristic of HIT (Amiral et al., 1992; Visentin et al., 1994;
Kelton et al., 1994). Because platelets have specific-binding sites for PF4, this
protein was originally assumed to mediate the attachment of HIT immune
complexes to the platelet surface (Kelton et al., 1994). However, it was
subsequently demonstrated that heparin, rather than PF4, serves this pur-
pose; that is, the binding to platelets of heparin-PF4 complexes occurs at
heparin-binding sites, rather than PF4-binding sites (Greinacher et al., 1993;
Horne and Hutchison, 1998).
The sequence of interactions leading to platelet activation has recently
been described (Newman and Chong, 2000). It begins with the nonimmune
binding of heparin to the cells, which stimulates them to secrete a relatively
small amount of PF4. The released PF4 binds to heparin in solution to reveal
the target for HIT IgG antibodies (HIT-IgG). Immune complexes of heparin,
PF4, and HIT-IgG then attach to the platelets at heparin-binding sites. This
facilitates contact between the Fc termini of the IgG molecules and the
platelet Fc receptors, thereby activating the cells further and causing more
PF4 secretion. As this sequence accelerates, platelet aggregation occurs.
Because immune complexes comprised of heparin, PF4, and IgG from
HIT patients bind to platelets at heparin-binding sites, they must compete
with free heparin for binding to the platelet surface (Horne and Alkins, 1996;
Horne and Hutchison, 1998). When free heparin is in molar excess, heparin–
PF4–IgG complexes are displaced from the platelet surface (Fig. 2). When
PF4 is in excess, there is no free heparin, and binding of the complexes is the
only option.
Variable heparin–PF4 stoichiometry may explain why some patients
develop HIT-IgG without developing thrombocytopenia, and why HIT is
more common in certain clinical settings than in others, such as following
surgery (Boshkov et al., 1993; Amiral et al., 1995, 1996; Warkentin et al.,
1995, 2000; Visentin et al., 1996; Kappers-Klunne et al., 1997; Bauer et al.,
1997). When a patient is given heparin, the plasma concentration of PF4 rises
because PF4 is displaced from the endothelial surface, where it is normally
bound to heparan sulfate (Dawes et al., 1982; Rao et al., 1983; O’Brien et al.,
1985). PF4 neoantigens (or cryptic antigens) are formed, leading to the HIT
immune response (Chong and Newman, 1997). However, complexes of
heparin, PF4, and IgG are harmless unless they become bound to platelets,
and this cannot happen as long as there is sufficient free heparin to compete
effectively for the limited number of platelet-binding sites (Horne and
Hutchison, 1998). Therefore, as long as heparin remains in molar excess over
156 Horne
Figure 2 Drawing illustrating the importance of the molar ratio of heparin to PF4 in
determining binding of HIT immune complexes to platelets. Maximal binding of HIT-
IgG occurs when heparin and PF4 are present in equimolar concentrations. Note that
IgG Fc interactions with platelet Fc receptors are not shown in this figure. (From
Horne and Hutchison, 1998.)
REFERENCES
Aggarwal A, Sobel BE, Schneider DJ. Decreased platelet reactivity in blood anti-
coagulated with bivalirudin or enoxaparin compared with unfractionated hepa-
rin: implications for coronary intervention. J Thromb Thrombolysis 2002; 13:
161–165.
Ahmad S, Jeske WP, Walenga JM, Hoppensteadt DA, Wood JJ, Herbert JM, Mess-
more HL, Fareed J. Synthetic pentasaccharides do not cause platelet activation
by anti-heparin-platelet factor 4 antibodies. Clin Appl Thromb Hemost 1999;
5:259–266.
Amiral J, Bridey F, Dreyfus M, Vissac AM, Fressinaud E, Wolf M, Meyer D. Platelet
factor 4 complexed to heparin is the target for antibodies generated in heparin-
induced thrombocytopenia [letter]. Thromb Haemost 1992; 68:95–96.
Amiral J, Bridey F, Wolf M, Boyer-Neumann C, Fressinaud E, Vissac AM, Peynaud-
Debayle E, Dreyfus M, Meyer D. Antibodies to macromolecular platelet factor
4–heparin complexes in heparin-induced thrombocytopenia: a study of 44 cases.
Thromb Haemost 1995; 73:21–28.
Amiral J, Peynaud-Debayle E, Wolf M, Bridey F, Vissac AM, Meyer D. Generation of
antibodies to heparin–PF4 complexes without thrombocytopenia in patients
treated with unfractionated or low-molecular-weight heparin. Am J Hematol
1996; 52:90–95.
Balduini CL, Noris P, Bertolino G, Previtali M. Heparin modifies platelet count and
function in patients who have undergone thrombolytic therapy for acute
myocardial infarction [letter]. Thromb Haemost 1993; 69:522–523.
Barber AF, Kaser-Glanzmann R, Jakabova M, Luscher EF. Characterization of a
chondroitin 4-sulfate proteoglycan carrier for heparin neutralizing activity
(platelet factor 4) released from human blood platelets. Biochim Biophys Acta
1972; 286:312–329.
Bauer TL, Arepally G, Konkle BA, Mestichelli B, Shapiro SS, Cines DB, Poncz M,
McNulty S, Amiral J, Hauck WW, Edie RN, Mannion JD. Prevalence of
heparin-associated antibodies without thrombosis in patients undergoing car-
diopulmonary bypass surgery. Circulation 1997; 95:1242–1246.
Berglund U, Wallentin L. Influence on platelet function by heparin in men with
unstable coronary artery disease. Thromb Haemost 1991; 66:648–651.
Bock PE, Luscombe M, Marshall SE, Pepper DS, Holbrook JJ. The multiple
complexes formed by the interaction of platelet factor 4 with heparin. Biochem J
1980; 191:769–776.
Nonimmune Heparin–Platelet Interactions 159
Horne MK III, Chao ES. The effect of molecular weight on heparin binding to
platelets. Br J Haematol 1990; 74:306–312.
Horne MK III, Hutchison KJ. Simultaneous binding of heparin and platelet factor-4
to platelets: further insights into the mechanism of heparin-induced thrombo-
cytopenia. Am J Hematol 1998; 58:24–30.
John LCH, Rees GM, Kovacs IB. Inhibition of platelet function by heparin. An
etiologic factor in postbypass hemorrhage. Thorac Cardiovasc Surg 1993; 105:
816–822.
Johnson RA, Lazarus KH, Henry DH. Heparin-induced thrombocytopenia: a pro-
spective study. Am J Hematol 1984; 17:349–353.
Kappers-Klunne MC, Boon DMS, Hop WCJ, Michiels JJ, Stibbe J, van der Zwaan C,
Koudstaal PJ, van Vliet HHDM. Heparin-induced thrombocytopenia and
thrombosis: a prospective analysis of the incidence in patients with heart and
cerebrovascular diseases. Br J Haematol 1997; 96:442–446.
Kelton JG. Heparin-induced thrombocytopenia. Haemostasis 1986; 16:173–186.
Kelton JG, Smith JW, Warkentin TE, Hayward CPM, Denomme GA, Horsewood
P. Immunoglobin G from patients with heparin-induced thrombocytopenia
binds to a complex of heparin and platelet factor 4. Blood 1994; 83:3232–
3239.
Kestin AS, Valeri CR, Khuri SF, Loscalzo J, Ellis PA, MacGregor H, Birjiniuk V,
Ouimet H, Pasche B, Nelson MJ, Benoit SE, Rodino LJ, Barnard MR, Mi-
chelson AD. The platelet function defect of cardiopulmonary bypass. Blood
1993; 82:107–117.
Klein B, Faridi A, von Tempelhoff GF, Heilmann L, Mittermayer C, Rath W. A whole
blood flow cytometric determination of platelet activation by unfractionated
and low molecular weight heparin in vitro. Thromb Res 2002; 108:291–296.
Lane DA, Denton J, Flynn AM, Thunberg L, Lindahl U. Anticoagulant activities of
heparin oligosaccharides and their neutralization by platelet factor 4. Biochem J
1984; 218:725–732.
Lindahl U, Kjellén L. Heparin or heparan sulfate—what is the difference? Thromb
Haemost 1991; 66:44–48.
Lindhoff-Last E, Nakov R, Misselwitz F, Breddin HK, Bauersachs R. Incidence and
clinical relevance of heparin-induced antibodies in patients with deep vein
thrombosis treated with unfractionated or low-molecular-weight heparin. Br J
Haematol 2002; 118:1137–1142.
Ljungberg B, Beving H, Egberg N, Johnsson H, Vesterqvist O. Immediate effects of
heparin and LMW heparin on some platelet and endothelial derived factors.
Thromb Res 1988; 51:209–217.
Loscalzo J, Melnick B, Handin RI. The interaction of platelet factor four and
glycosaminoglycans. Arch Biochem Biophys 1985; 240:446–455.
Magnani HN. Heparin-induced thrombocytopenia (HIT): an overview of 230 patients
treated with Orgaran (Org 10172). Thromb Haemost 1993; 70:554–561.
Marshall SE, Luscombe M, Pepper DS, Holbrook JJ. The interaction of platelet factor
4 with heparins of different chain length. Biochim Biophys Acta 1984; 797:34–
39.
Mikhailidis DP, Barradas MA, Jeremy JY, Gracey L, Wakeling A, Dandona P.
162 Horne
Visentin GP, Malik M, Cyganiak KA, Aster RH. Patients treated with unfractionated
heparin during open heart surgery are at high risk to form antibodies reactive
with heparin:platelet factor 4 complexes. J Lab Clin Med 1996; 128:376–383.
Walenga JM, Jeske WP, Bara L, Samama MM, Fareed J. Biochemical and phar-
macologic rationale for the development of a synthetic heparin pentasaccharide.
Thromb Res 1997; 86:1–36.
Ware AJ, Coller BS. Platelet morphology, biochemistry, and function. In: Beutler E,
Lichtman MA, Coller BS, Kipps TJ, eds. Williams Hematology. 5th ed. New
York: McGraw-Hill, 1995:1161–1201.
Warkentin TE, Kelton JG. Interaction of heparin with platelets, including heparin-
induced thrombocytopenia. In: Bounameaux H, ed. Low-Molecular-Weight
Heparins in Prophylaxis and Therapy of Thromboembolic Diseases. New York:
Marcel Dekker, 1994:75–127.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular-
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Warkentin TE, Chong BH, Greinacher A. Heparin-induced thrombocytopenia: to-
wards consensus. Thromb Haemost 1998; 79:1–7.
Warkentin TE, Sheppard JA, Horsewood P, Simpson PJ, Moore JC, Kelton JG.
Impact of the patient population on the risk for heparin-induced thrombocy-
topenia. Blood 2000; 96:1703–1708.
Westwick J, Scully MF, Poll C, Kakkar VV. Comparison of the effects of low
molecular weight heparin and unfractionated heparin on activation of human
platelets in vitro. Thromb Res 1986; 42:435–447.
Xiao Z, Théroux P. Platelet activation with unfractionated heparin at therapeutic
concentrations and comparisons with a low-molecular-weight heparin and with
a direct thrombin inhibitor. Circulation 1998; 97:251–256.
Young E, Wells P, Holloway S, Weitz J, Hirsh J. Ex-vivo and in-vitro evidence that low
molecular weight heparins exhibit less binding to plasma proteins than un-
fractionated heparin. Thromb Haemost 1994; 71:300–304.
6
Heparin-Dependent Antigens in
Heparin-Induced Thrombocytopenia
Jean Amiral
HYPHEN BioMed, Neuville-sur-Oise, France
Dominique Meyer
INSERM U-143, Hôpital de Bicêtre, Bicêtre, France
I. INTRODUCTION
HIT is lower with the use of low molecular weight heparin (LMWH)
(Warkentin et al., 1995). However, recent studies suggest that this complica-
tion can also develop in the absence of IgG isotypes (Amiral et al., 1996c). In
some patients with apparent HIT, only IgA or IgM isotypes are present,
usually in high concentrations.
In this chapter, the current understanding of PF4–H antibody genera-
tion and its contribution to the complications of HIT will be discussed.
Formation of the PF4–H antigen complexes and their binding to blood and
endothelial cells, thus targeting the immune response onto these cells (Cines
et al., 1987; Visentin et al., 1994; Visentin and Aster, 1995; Horne and Hut-
chison, 1998), will be analyzed. Finally, the possibility that HIT can be caused
in the absence of detectable antibodies to PF4–H will be discussed, including
the hypothesis that preexisting antibodies to other chemokines could become
pathogenic during heparin treatment.
complex has a molecular weight (MW) of 350 kDa. The PF4 complexes can
also bind to endothelial cell proteoglycans (heparan sulfate). Heparin, when
present, having a greater affinity for PF4, displaces PF4 from the endothelial
cell glycosaminoglycans, thereby forming PF4–H complexes that are released
into the circulation.
The interaction between heparin and PF4 has been intensively studied
(Bock et al., 1980; Cowan et al., 1986; Stuckey et al., 1992; Maccarana and
Lindahl, 1993). In the presence of a stoichiometric concentration of heparin
and PF4 (which corresponds to 27 international units [IU] of heparin per
milligram of PF4), multimolecular PF4–H complexes (Greinacher et al.,
1994; Amiral et al., 1995) are generated. With stoichiometric concentrations,
heparin wraps around the PF4 molecule, altering its structure and rendering it
antigenic. Figure 1 shows the different complexes that can be formed between
heparin and PF4, depending on the respective concentrations of both sub-
stances. Only multimolecular complexes are believed to be antigenic in
heparin-treated patients. Thus, the immunogenicity of complexes is strictly
dependent on the respective concentrations of heparin and PF4. If we
consider the usual therapeutic range for heparin (0.1–1 IU/mL), the amount
of PF4 required for the generation of multimolecular PF4–H complexes is
from 3 to 40 Ag/mL. In patients undergoing cardiopulmonary bypass who
receive higher heparin concentrations (up to 3 IU/mL), the corresponding
(Capitanio et al., 1985; Rybak et al., 1989). Although the HIT antibodies
recognize PF4–H complexes in the fluid phase (Newman et al., 1998), it is
uncertain whether this typically occurs in vivo before interaction of PF4–H–
IgG complexes with the platelet surface, or whether HIT antibodies only bind
after PF4–H complexes are attached to the platelet surface.
Regardless, the clinical state of patients—determining the extent of
platelet and endothelial cell activation—seems to be a key factor for deter-
mining whether clinical HIT results (Boshkov et al., 1993; Reininger et al.,
1996). This contribution occurs in several ways: activated platelet generate
high PF4 concentrations that can complex with heparin, and activated cells
also expose a higher density of heparin-binding sites (Horne and Chao, 1989).
Furthermore, these platelets may be more readily activated by heparin-
dependent antibodies. This situation occurs in patients with acute or chronic
blood activation associated with cardiopulmonary bypass, atherosclerosis,
inflammation, infections, cancer, diabetes, orthopedic surgery, among others.
Another factor determining HIT antibody formation is the type of
heparin used for heparin binding to PF4, which depends on its oligosaccha-
ride composition, polysaccharide length, and grade of sulfation (Lindahl et
al., 1994; Greinacher et al., 1995). Formation of PF4–H complexes requires a
heparin molecule with at least 12–14 oligosaccharide units and a high
sulfation grade (more than three sulfate groups per disaccharide) (Amiral et
al., 1995). Furthermore, binding of heparin to blood and endothelial cells also
increases with heparin molecule length and sulfation grade (Sobel and Adel-
man, 1988; Horne and Chao, 1990; Harenberg et al., 1994). Heparin structure
thus has a dual effect in HIT: it is required to form PF4–H complexes and also
to target these complexes onto cells. These factors could explain the higher
frequency of PF4–H antibody development and of HIT in patients receiving
UFH, compared with LMWH. With UFH, PF4–H complexes are more easily
formed and require a lower heparin concentration than with LMWH. For
the latter drug, only the subset of molecules containing at least 12–14 oligo-
saccharide units (MW > 3600 Da) can generate immunoreactive PF4–H
complexes. Thus, because LMWH has a lower propensity to form PF4–H
complexes and binds less readily to platelets and endothelial cells, LMWH
therapy may be less likely to result in thrombocytopenia even in the presence
of pathogenic HIT antibodies.
PF4–H–reactive antibodies targeted to platelets induce platelet activa-
tion, resulting in thrombocytopenia and, often, thrombosis. Occasionally,
heparin-induced thrombosis occurs in the absence of thrombocytopenia
(Hach-Wunderle et al., 1994; Bux-Gewehr et al., 1996). Platelet activation
by the IgG isotype antibodies is mediated by interaction with the platelet
FcgRIIA receptors (Kelton et al., 1994; Denomme et al., 1997). Some studies
suggest an important role for FcgRIIA polymorphism (Brandt et al., 1995;
170 Amiral and Meyer
Burgess et al., 1995). However, the role of the FcgRIIA receptor polymor-
phism is controversial (Arepally et al., 1997; Denomme et al., 1997; Suh et al.,
1997; Bachelot-Loza et al., 1998) (Chap. 9).
Platelet activation might also occur through other mechanisms, such as
direct antibody binding to exposed cell antigens (Rubinstein et al., 1995), a
phenomenon that is dependent on the antigen electric charge (Schattner et al.,
1993). Heparin is highly electronegative. Evidence for direct activation
through antigen binding is supported by the positive platelet aggregation
produced by some patient plasma samples containing only antibodies of the
IgM or IgA isotypes. Furthermore, in vivo, platelets are in their blood and
endothelial environment. Formation of heparin-containing immune com-
plexes on cell surfaces can initiate blood and endothelial cell interactions, and
this can enhance the activating effect. Cell–cell interactions may occur and be
amplified through release products that chemoattract and activate cells, or
through transcellular metabolism (Nash, 1994; Marcus et al., 1995). Platelet
products (e.g., PF4) and platelet-derived microparticles (Warkentin et al.,
1994) can induce activation of leukocytes (Aziz et al., 1995; Jy et al., 1995;
Petersen et al., 1996). Leukocyte-release products, such as cathepsin G, can
directly activate platelets and cleave h-thromboglobulin to the active chemo-
kine NAP-2, thus establishing an amplification loop. Platelet–leukocyte
aggregates can form in vivo contributing to vascular occlusion, especially in
limb vessels (Fig. 2). In a recent study, antibodies to PF4–H from patients
with HIT were shown to induce synthesis of tissue factor by monocytes in the
presence of PF4 and heparin (Pouplard et al., 2001). This could be a com-
plementary pathway for inducing thrombosis.
Various characteristics of PF4–H antibodies are another key factor for
induction of HIT. Platelet activation induced by PF4–H antibodies is usually
weak and is only pathogenic when amplified. This is demonstrated by the
variable lag phase observed in platelet aggregation studies with different plas-
mas or sera from HIT patients. Antibody concentration is an important factor
for determining the extent of platelet activation. Antibody affinity is also very
important: the higher the affinity, the lower the concentration of antibodies
required for activating platelets. Recently, a subset of antibodies to PF4–H
complexes that had platelet-activating properties was isolated in three pa-
tients with HIT. These antibodies had the highest avidity for PF4–H. In con-
trast, the bulk of antibodies to PF4–H in these patients had no effect on plate-
let activation (Amiral et al., 2000). When IgM or IgA isotypes are present,
affinity for PF4–H complexes is usually lower than that of IgG isotypes and,
consequently, high concentrations are necessary for pathogenicity. Lastly,
HIT antibodies do not all bind to the same epitope on PF4–H complexes, and
this specificity could be an important factor in their action (Horsewood et al.,
1996; Pouplard et al., 1997; Suh et al., 1998). At least two neoepitopes have
Heparin-Dependent Antigens in HIT 171
been identified on PF4 that are distinct from the ‘‘region of positive charge’’ to
which heparin binds (Ziporen et al., 1998; Li et al., 2002) (see also color insert
and Chap. 7). Thus, anti-PF4–H antibodies are not equivalent, and those with
the strongest affinity are most pathogenic.
Recent data show that primary platelet activation in HIT involves ADP
receptors (Polgár et al., 1998), and that platelet aggregation involves GPIIb/
IIIa (Hérault et al., 1997; Jeske et al., 1997). These findings further emphasize
the importance of platelet activation amplification loops for producing the
clinical manifestations of HIT.
172 Amiral and Meyer
V. CONCLUSIONS
The conditions that permit formation of the molecular PF4–H target antigen
for HIT antibodies involve the properties of the heparin used, dose and
duration of therapy, and the clinical context of the treated patient. Immuno-
reactive complexes between PF4 and heparin are formed only under certain
conditions. Their formation in high concentrations is facilitated if underlying
disease favors platelet activation and release. Similar conditions enhance the
pathogenicity of the HIT-generated antibodies. These considerations help
unravel the apparent random generation of HIT antibodies in heparin-treated
patients, as well as the seemingly random occurrence of thrombotic events.
REFERENCES
Horsewood P, Warkentin TE, Hayward CPM, Kelton JG. The epitope specificity of
heparin-induced thrombocytopenia. Br J Haematol 1996; 95:161–167.
Jeske WP, Walenga JM, Szatkowski E, Ero M, Herbert JM, Haas S, Bakhos M. Effect
of glycoprotein IIb-IIIa antagonists on the HIT serum induced activation of
platelets. Thromb Res 1997; 88:271–281.
Jy W, Mao WW, Horstman LL, Tao J, Ahn YS. Platelet microparticles bind, activate
and aggregate neutrophils in vitro. Blood Cells Mol Dis 1995; 21:217–231.
Kelton JG, Smith JW, Warkentin TE, Hayward CPM, Denomme GA, Horsewood P.
Immunoglobulin G from patients with heparin-induced thrombocytopenia
binds to a complex of heparin and platelet factor 4. Blood 1994; 83:3232–3239.
Kelton J. Pathophysiology of heparin-induced thrombocytopenia [letter]. Br J
Haematol 1992; 82:778–784.
Li ZQ, Liu W, Park KS, Sachais BS, Arepally GM, Cines DB, Poncz M. Defining a
second epitope for heparin-induced thrombocytopenia/thrombosis antibodies
using KKO, a murine HIT-like monoclonal antibody. Blood 2002; 99:1230–
1236.
Lindahl U, Lidholt K, Spillmann D, Kjellen L. More to ‘‘heparin’’ than anticoagulant.
Throm Res 1994; 75:1–32.
Maccarana M, Lindahl U. Mode of interaction between platelet factor 4 and heparin.
Glycobiology 1993; 3:271–277.
Marcus AJ, Safier LB, Broekman MJ, Islam N, Fliessbach JH, Hajjar KA, Kaminski
WE, Jendraschak E, Silverstein RL, von Schacky C. Thrombosis and inflam-
mation as multicellular processes: significance of cell–cell interactions. Thromb
Haemost 1995; 74:213–217.
Nash GB. Adhesion between neutrophils and platelets: a modulator of thrombotic and
inflammatory events? Thromb Res 1994; 74:S3–S11.
Newman PM, Swanson RL, Chong BH. Heparin-induced thrombocytopenia: IgG
binding to PF4-heparin complexes in the fluid phase and cross-reactivity with
low molecular weight heparin and heparinoid. Thromb Haemost 1998; 80:292–
297.
Petersen F, Lidwig A, Flad HD, Brandt E. TNF-a renders human neutrophils
responsive to platelet factor 4. J Immunol 1996; 156:1954–1962.
Polgár J, Eichler P, Greinacher A, Clemetson KJ. Adenosine diphosphate (ADP) and
ADP receptor play a major role in platelet activation/aggregation induced by
sera from heparin-induced thrombocytopenia patients. Blood 1998; 91:549–554.
Pouplard C, Amiral J, Borg JY, Vissac AM, Delahousse B, Gruel Y. Differences in
specificity of heparin-dependent antibodies developed under low-molecular-
weight-heparin therapy and higher cross-reactivity with Orgaran. Br J
Haematol 1997; 99:273–280.
Pouplard C, Iochmann I, Renard O, Hérault O, Colombat P, Amiral J, Gruel Y.
Induction of monocyte tissue factor expression by antibodies to platelet factor 4
developed in heparin-induced thrombocytopenia. Blood 2001; 97:3300–3302.
Regnault V, de Maistre E, Carteaux JP, Gruel Y, Nguyen P, Tardy B, Lecompte T.
Platelet activation induced by human antibodies to interleukin-8. Blood 2003;
101:1419–1421.
176 Amiral and Meyer
Warkentin TE, Hayward CPM, Boshkov LK, Santos AV, Sheppard JI, Bode AP,
Kelton JG. Sera from patients with heparin-induced thrombocytopenia
generate platelet-derived microparticles with procoagulant activity: an expla-
nation for the thrombotic complications of heparin-induced thrombocytopenia.
Blood 1994; 84:3691–3699.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular-
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Ziporen L, Li ZQ, Park KS, Sabnekar P, Liu WY, Arepally G, Shoenfeld Y, Kieber-
Emmons T, Cines DB, Poncz M. Defining an antigenic epitope on platelet factor
4 associated with heparin-induced thrombocytopenia. Blood 1998; 92:3250–
3259.
7
Molecular Immunopathogenesis of
Heparin-Induced Thrombocytopenia
I. INTRODUCTION
Figure 1 Nonspecific role of heparin and other polyanions that lead to neoepitope
formation on PF4. (From Warkentin, 2003.)
Figure 3 Amino acid composition of human PF4 monomers: Residues crucial for
heparin binding [COOH-terminal a-helix residues encompassing lysines 61–62 and
65–66 (Loscalzo et al., 1985), arginines 20, 22, and 49, histidine 23, threonine 25, and
lysine 46 (Mayo, 1995b)] are boxed. Filled circles identify the six residues (37, 38, 39,
49, 55, and 57) in the 47 COOH-terminal region of PF4 at which human and rat PF4
differ.
Figure 5 Spectratype analysis of the hV 6.1 family; PBMC from a patient with
HIT were cultured in the presence or in the absence of the antigen (heparin–PF4).
‘‘Oligoclonal’’ expansion of T cells is observed only under stimulation with heparin–
PF4.
Molecular Immunopathogenesis of HIT 189
positions 49 and 55 only at a higher ratio of heparin to PF4 (0.8 U/mL vs. 0.5
U/mL). None of the 15 antibodies recognized peptides comprising the 26 or
15 COOH-terminal amino acid residues of the PF4 monomer, or reduced
alkylated human PF4 either in presence or absence of heparin.
These results, together with the observations by Ziporen and associates
(1998), point to the region of PF4 between the third and fourth cysteine
residues as the major antigenic site for HIT antibody binding (Fig. 4 [see color
insert, Fig. 5]). Li et al. (2002) using a series of mouse/human PF4 chimeras
identified another antigenic site, on PF4–heparin that requires both P34 and
an intact N-terminus (Fig. 4 [see color insert, Fig. 5]). The latter results,
together with our studies utilizing biotin-labeled affinity-purified HIT anti-
bodies in a competitive inhibition assay (Suh et al., 1998), indicate that at
least three dominant HIT antibody recognition sites can be distinguished
and further support the idea that HIT antibodies recognize conformation-
dependent ‘‘neoepitopes’’ formed on PF4 when it binds to heparin.
The finding that HIT antibodies can be of the IgM, IgG, or IgA isotype
(Visentin et al., 1994; Greinacher et al., 1994; Kelton et al., 1994; Amiral et al.,
1995, 1996b; Arepally et al., 1997; Suh et al., 1997) indicates that class switch-
ing, likely requiring helper T cells, takes place in patients mounting a humoral
immune response to heparin–PF4. Although HIT is a drug-induced disorder,
parallels for the role of T cells in HIT may be drawn from studies of auto-
immune conditions, such as systemic lupus erythematosus, systemic sclerosis,
and insulin autoimmune syndrome (Ito et al., 1993; Crow et al., 1994; Ku-
wana et al., 1995b). In both lupus and scleroderma, T-helper cells mediate
antigen-specific autoantibody production by B cells (Adams et al., 1991;
Mohan et al., 1993; Kuwana et al., 1995a).
We hypothesize that the heparin–PF4 complex not only is the target for
antibody, but also is the stimulus for T-cell activation, and have used T-cell
receptor (TCR) spectratyping (Maslanka et al., 1995), also called immuno-
scope (Cochet et al., 1992; Pannetier et al., 1993), and clonotyping (Maslanka
et al., 1996) to characterize the T-cell response to PF4–heparin complexes in
HIT. The TCRs of more than 95% of peripheral blood T cells are composed
of two highly variable a- and h-chain glycoproteins, which function together
in a complex with five other invariant molecules (CD3 complex) on the surface
of the cell. The genes encoding the TCR h-chain subunit undergo sequential
rearrangments analogous to that of the immunoglobulin superfamily of
genes, during which D and J segments first, and then V segments, are com-
bined to form various VDJ sequences (LaRoque and Robinson, 1996). Diver-
190 Visentin et al.
duced (cryptic peptides) are generated and presented to T cells in the context
of class II MHC molecules. Studies in murine systems provide examples of
‘‘autoimmune’’ states triggered by exogenous agents that perturb protein
processing (Hess et al., 1991; Griem et al., 1996), but this phenomenon is not
well characterized in the context of human disease. Our studies support a
model in which pharmacological doses of heparin cause aberrant processing
of PF4 by APCs, leading to the presentation of peptides not ordinarily seen by
the immune system. This hypothesis can be tested directly if T-cell clones can
be developed from mononuclear cells responding to heparin–PF4 cultures.
It appears that multiple factors influence the formation of antibodies
specific for heparin–PF4 complexes in patients receiving heparin. Currently,
there is no evidence to support genetic predisposition as a basis for antibody
formation in patients receiving heparin. Unlike the situation in alloimmune
thrombocytopenia (de Waal et al., 1986; Mueller-Eckhardt et al., 1989), no
connection between HIT and human leukocyte antigens (HLA) has been
found (Greinacher and Mueller-Eckhardt, 1993). IgM antibodies specific for
heparin–PF4 complexes are a common finding in HIT (Visentin et al., 1994,
1999), indicating a primary immune response, and it could be speculated that
patients who received UFH previously may be at greater risk to produce
heparin–PF4–specific antibodies and develop HIT, if rechallenged with hep-
arin. However, Cadroy et al. (1994) described a patient with a history of HIT
who mounted a brisk IgM response when challenged again with UFH 3 years
later. A report by Warkentin and Kelton (2001) suggests that there is no
anamnestic immune response in HIT (i.e., patients either have ‘‘typical’’ HIT
[onset at days 5–10] or ‘‘rapid’’ HIT, the latter apparently caused by residual
circulating HIT antibodies, rather than a secondary immune response). Fur-
thermore, HIT did not necessarily recur in patients who were exposed to
heparin a second time.
V. IMPLICATIONS
The identification of mutations of human PF4 that lead to loss of HIT anti-
body binding will not necessarily localize the epitopes at which antibodies
attach because the actual binding site(s) could be elsewhere in the PF4 tet-
ramer. Moreover, HIT antibodies appear to recognize multiple sites on PF4–
heparin (Suh et al., 1998). Because the PF4 molecule is a nearly symmetrical
tetramer (Ibel et al., 1986), the HIT epitope could be expressed four times
on each heparin–PF4 heterodimer, creating the potential for even a single
antibody clone to react with four sites on a PF4 tetramer complexed with hep-
arin. Studies from our group (Visentin et al., 1994, 1996) and others (Amiral
et al., 1995; Arepally et al., 1995) have shown that, although antibodies
192 Visentin et al.
ACKNOWLEDGMENTS
REFERENCES
Adams S, Leblanc P, Datta SK. Junctional region sequences of T-cell receptor beta-
chain genes expressed by pathogenic anti-DNA autoantibody-inducing helper T
cells from lupus mice: possible selection by cationic autoantigens. Proc Natl
Acad Sci USA 1991; 88:11271–11275.
Amiral J, Bridey F, Dreyfus M, Vissac AM, Fressinaud E, Wolf M, Meyer D. Platelet
factor 4 complexed to heparin is the target for antibodies generated in heparin-
induced thrombocytopenia [letter]. Thromb Haemost 1992; 68:95–96.
Amiral J, Bridey F, Wolf M. Antibodies to macromolecular platelet factor-4 heparin
complexes in heparin-induced thrombocytopenia: a study of 44 cases. Thromb
Haemost 1995; 73:21–28.
Amiral J, Marfaing-Koka A, Wolf M, Alessi MC, Tardy B, Boyer-Neumann C, Vissac
AM, Fressinaud E, Poncz M, Meyer D. Presence of autoantibodies to inter-
leukin-8 or neutrophil-activating peptide-2 in patients with heparin-associated
thrombocytopenia. Blood 1996a; 88:410–416.
Amiral J, Wolf M, Fischer AM, Boyer-Neumann C, Vissac AM, Meyer D. Patho-
genicity of IgA and/or IgM antibodies to heparin–PF4 complexes in patients
with heparin-induced thrombocytopenia. Br J Haematol 1996b; 92:954–959.
Arepally G, Reynolds C, Tomaski A, Amiral J, Jawad A, Poncz M, Cines DB.
Comparison of PF4–heparin ELISA assay with the 14C-serotonin release assay
Molecular Immunopathogenesis of HIT 193
function of the recombined germ-line segments. Proc Natl Acad Sci USA 1993;
90:4319–4323.
Poncz M, Surrey S, LaRocco P, Weiss MJ, Rappaport EF, Conway TM, Schwartz E.
Cloning and characterization of platelet factor 4 cDNA derived from a human
erythroleukemic cell line. Blood 1987; 69:219–223.
Rollins BJ. Chemokines [review]. Blood 1997; 90:909–928.
St. Charles R, Walz DA, Edwards BFP. The three-dimensional structure of bovine
platelet factor 4 at 3.0-A resolution. J Biol Chem 1989; 264:2092–2099.
Stringer SE, Gallagher JT. Specific binding of the chemokine platelet factor 4 to
heparan sulfate. J Biol Chem 1997; 272:20508–20514.
Stuckey JA, St. Charles R, Edwards DFP. A model of the platelet factor 4 complex
with heparin. Proteins 1992; 14:277–287.
Suh JS, Malik MI, Aster RH, Visentin GP. Characterization of the humoral immune
response in heparin-induced thrombocytopenia. Am J Hematol 1997; 54:196–
201.
Suh JS, Aster RH, Visentin GP. Antibodies from patients with heparin-induced
thrombocytopenia/thrombosis recognize different epitopes on heparin: platelet
factor 4 complexes. Blood 1998; 91:916–922.
Tardy B. Thrombocytopenia associated with low-molecular-weight heparin [letter].
Lancet 1991; 338:1217.
Visentin GP. Heparin-induced thrombocytopenia: molecular pathogenesis. Thromb
Haemost 1999; 82:448–456.
Visentin GP, Ford SE, Scott PJ, Aster RH. Antibodies from patients with heparin-
induced thrombocytopenia/thrombosis are specific for platelet factor 4 com-
plexed with heparin or bound to endothelial cells. J Clin Invest 1994; 93:81–88.
Visentin GP, Malik M, Cyganiak KA, Aster RH. Patients treated with unfractionated
heparin during open heart surgery are at high risk to form antibodies reactive
with heparin: platelet factor 4 complexes. J Lab Clin Med 1996; 128:376–383.
Visentin GP, Moghaddam M, Beery SE, McFarland JG, Aster RH. Heparin is not
required for detection of antibodies associated with heparin-induced thrombo-
cytopenia thrombosis. J Lab Clin Med 2001; 138:22–31.
Warkentin TE. Heparin-induced thrombocytopenia: pathogenesis and management
[review]. Br J Haematol 2003; 12:535–555.
Warkentin TE, Kelton JG. Heparin-induced thrombocytopenia. Prog Hemost
Thromb 1991; 10:1–34.
Warkentin TE, Kelton JG. Temporal aspects of heparin-induced-thrombocytopenia.
N Engl J Med 2001; 344:1286–1292.
Zhang X, Chen L, Bancroft DP, Lai CK, Maione TE. Crystal structure of recom-
binant human platelet factor 4. Biochemistry 1994; 33:8361–8366.
Ziporen L, Li ZQ, Park KS, Sabnekar P, Liu WY, Arepally G, Shoenfeld Y, Kieber-
Emmons T, Cines DB, Poncz M. Defining an antigenic epitope on platelet factor
4 associated with heparin-induced thrombocytopenia. Blood 1998; 92:3250–
3259.
8
Role of Sulfated Polysaccharides
in the Pathogenesis of Heparin-
Induced Thrombocytopenia
Susanne Alban
Christian-Albrechts University of Kiel, Kiel, Germany
Andreas Greinacher
Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
I. INTRODUCTION
B. Structure of Heparin
Heparin is a polydisperse mixture of GAGs with MWs ranging from 5 to 40
kDa, with an average MW of 13 kDa (Linhardt and Toida, 1997). It is com-
posed of alternating D-glucosamine residues linked 1!4 to either L-iduronic
acid or D-glucuronic acid (Casu, 1985). The principal repeating unit in
heparin is the trisulfated disaccharide [! 4)-O-a-L-iduronic acid-2-sulfate
(1 ! 4)-O-a-D-glucosamine-2,6-disulfate (1 !] (Fig. 1), which represents 75–
90% of the heparin chain (Linhardt et al., 1992). The remaining 10–25% of
disaccharide units differ in their degree and positions of sulfation (Linhardt
et al., 1988). Besides, there are disaccharides consisting of unsulfated glucu-
ronic acid and/or N-acetylglucosamine. With a SO
3 :COO ratio of 2.0–2.5,
heparin is the GAG with the highest charge density. By binding to domains
containing positively charged amino acids, especially arginine and lysine, it
Role of Sulfated Polysaccharides in HIT 199
Figure 2 Pentasaccharide sequence of the AT-binding site of heparin: sulfate groups essential for the AT-binding
are encircled.
Alban and Greinacher
Role of Sulfated Polysaccharides in HIT 201
(Dawes et al., 1982). Platelets do not release PF4 when incubated with heparin
in vitro (see Chap. 5). However, in vivo, heparin and some other GAGs are
able to increase plasma PF4 levels (Cella et al., 1986). Thus, endothelial-
bound, rather than platelet-stored, PF4 seems to be the predominant source
of the PF4 released by heparin. Most likely, heparin and other high-sulfated
polysaccharides are able to displace PF4 from endothelial heparan sulfate in
relation to their affinity for PF4 (O’Brien et al., 1985).
INN (Brand name) Degradation method Mean MW (kDa) Anti-Xa (U/mg) Anti-Xa: Anti-IIa ratioa
B. Danaparoid
Danaparoid sodium (Orgaran) is an alternative anticoagulant that is effective
for treating patients with HIT (see Chap. 14). This heparinoid consists of a
depolymerized mixture of GAGs extracted from porcine intestinal mucosa,
with a mean MW of 6 kDa. Its components are approximately 80% low
molecular weight heparan sulfate, 10% dermatan sulfate, 5% chondroitin
sulfate, and a small proportion of heparan sulfate (4%) with high affinity for
AT (Meuleman, 1992). Apart from the minor AT-binding heparan sulfate
component, the constituents of danaparoid have a DS per monosaccharide
between 0.5 and 0.7, as well as a low MW. Thus, the two important require-
ments to form multimolecular complexes with PF4 are not met. This is con-
210 Alban and Greinacher
sistent with the low cross-reactivity rate of danaparoid (about 10%) (Wilde
and Markham, 1997) (see Chaps. 11 and 14). As danaparoid inhibits platelet
activation by HIT antibodies even in the presence of heparin (Chong et al.,
1989b), it is possible that the GAG mixture binds to PF4 without producing
the antigen. Consequently, less PF4 is available for the small amount of
higher-sulfated heparan sulfate molecules responsible for AT binding and,
presumably, PF4 binding resulting in cross-reactivity with HIT antibodies
(Greinacher et al., 1992).
C. Pentasaccharides
Within the scope of developing new carbohydrate-based antithrombotics,
fondaparinux, a fully synthetic, chemically defined pentasaccharide (formerly
named Org31540/SR90107A, MW = 1728 Da; DS = 1.6; 700 anti-Xa U/
mg), has been developed, which corresponds to the AT-binding site of heparin
(Petitou et al., 1997) (Fig. 4). By its highly specific binding to AT, fondapa-
rinux selectively inhibits factor Xa and thus prevents thrombin generation
(Bauer et al., 2002). In four phase III clinical trials evaluating the prevention
of venous thromboembolism after major orthopedic surgery (>7300 patients),
fondaparinux showed superiority over the LMWH enoxaparin without
increasing clinically important bleeding (Turpie et al., 2001; Eriksson et al.,
2001; Bauer et al., 2001, Lassen et al., 2002; Turpie et al., 2002a,b) and has
recently been approved for this indication. At present, fondaparinux is under
further investigation for antithrombotic prophylaxis in other clinical settings,
as well as for treatment of deep vein thrombosis, pulmonary embolism, and
E. Conclusions
From experiments with well-defined GluS, the various structural require-
ments for a sulfated carbohydrate to form the HIT antigen have become clear.
Given this detailed knowledge, at least three carbohydrate-based anticoagu-
lant options can be proposed that should have a negligible risk for inducing
clinical HIT:
1. Mixtures of GAGs consisting predominantly of low-sulfated car-
bohydrates with correspondingly limited capacity to form anti-
genic complexes with PF4: A prototype of such an anticoagulant is
danaparoid.
2. Oligosaccharides with antithrombotic activity similar to the AT-
binding pentasaccharide: One such agent appears promising: fon-
daparinux did not cause HIT in more than 4000 patients treated
after orthopedic surgery.
3. GAGs with highly sulfated, but short, regions that are connected
by nonsulfated ‘‘spacers’’: Hereby, the thrombin-binding site and
the AT-specific pentasaccharide can be expressed in a single mole-
cule without reaching the critical length of a sulfated chain critical
for HIT antigen formation (Petitou et al., 1999).
Role of Sulfated Polysaccharides in HIT 213
The increasing use of LMWH already seems to have reduced the incidence of
HIT. We propose that the problem of HIT can be avoided further by using
anticoagulants meeting the foregoing outlined criteria in our treatment
arsenal.
ACKNOWLEDGMENTS
REFERENCES
Ahmad S, Jeske WP, Walenga JM, Hoppensteadt DA, Wood JJ, Herbert JM,
Messmore HL, Fareed J. Synthetic pentasaccharides do not cause platelet ac-
tivation by antiheparin-platelet factor 4 antibodies. Clin Appl Thromb Hemost
1999; 5:259–266.
Alban S. Synthese und physiologische Testung neuartiger Heparinoide. Ph.D dis-
sertation, University of Regensburg, Germany, 1993.
Alban S. Carbohydrates with anticoagulant and antithrombotic properties. In: Wit-
czak ZJ, Nieforth KA, eds. Carbohydrates in Drug Design. New York: Marcel
Dekker, 1997:209–276.
Alban S, Franz G. Anticoagulant activity of curdlan sulfates in dependence on their
molecular weight. Pure Appl Chem 1994a; 66:2403–2406.
Alban S, Franz G. Gas liquid chromatography–mass spectrometry analysis of anti-
coagulant active curdlan sulfates. Semin Thromb Hemost 1994b; 20:152–158.
Alban S, Jeske W, Welzel D, Franz G, Fareed J. Anticoagulant and antithrombotic
actions of a semisynthetic h-1,3-glucan sulfate. Thromb Res 1995; 78:201–210.
Amiral J. Le facteur 4 plaquettaire, cible des anticorps anti-héparine: application au
diagnostic biologique de la thrombopénie induite par l’éparine (TIH). Ann
Med Intern 1997; 148:142–149.
Amiral J, Bridey F, Dreyfus M, Vissac AM, Fressinaud E, Wolf M, Meyer D.
Platelet factor 4 complexed to heparin is the target for antibodies generated in
heparin-induced thrombocytopenia [letter]. Thromb Haemost 1992; 68:95–96.
Amiral J, Bridey F, Wolf M, Boyer-Neumann C, Fressinaud E, Vissac AM, Pey-
naud-Debayle E, Dreyfus M, Meyer D. Antibodies to macromolecular platelet
factor 4-heparin complexes in heparin-induced thrombocytopenia: a study of
44 cases. Thromb Haemost 1995; 73:21–28.
Amiral J, Peynaud-Debayle E, Wolf M, Bridey F, Vissac AM, Meyer D. Generation
of antibodies to heparin–PF4 complexes without thrombocytopenia in
patients treated with unfractionated or low-molecular-weight heparin. Am J
Hematol 1996a; 52:90–95.
Amiral J, Wolf M, Fischer A, Boyer-Neumann C, Vissac A, Meyer D. Pathogenicity
214 Alban and Greinacher
Casu B. Structure and biological activity of heparin. Adv Carbohydr Chem Biochem
1985; 43:51–134.
Casu B. Heparin structure. Haemostasis 1990; 20(suppl 1):62–73.
Cella G, Scattolo N, Luzzatto G, Stevanato F, Vio C, Girolami ASO. Effects on
platelets and on the clotting system of four glycosaminoglycans extracted from
hog mucosa and one extracted from aortic intima of the calf. J Med 1986;
17:331–346.
Choay J. Structure and activity of heparin and its fragments: an overview. Semin
Thromb Hemost 1989; 15:359–364.
Chong BH, Fawaz I, Chestermann CN, Berndt MC. Heparin-induced thrombocy-
topenia: mechanism of interaction of the heparin-dependent antibody with
platelets. Br J Haematol 1989a; 73:235–240.
Chong BH, Ismail F, Cade J, Gallus AS, Gordon S, Chesterman CN. Heparin-
induced thrombocytopenia: studies with a new molecular weight heparinoid,
Org 10172. Blood 1989b; 73:1592–1596.
Chong BH, Pilgrim RL, Cooley MA, Chesterman CN. Increased expression of plate-
let IgG Fc receptors in immune heparin-induced thrombocytopenia. Blood
1993; 81:988–993.
Cines DB, Tomaski A, Tannenbaum S. Immune endothelial-cell injury in heparin-
associated thrombocytopenia. N Engl J Med 1987; 316:581–589.
Coussement PK, Bassand JP, Convens C, Vrolix M, Boland J, Grollier G, Michels R,
Vahanian A, Vanderheyden M, Rupprecht HJ, Van de Werf F; PENTA-
LYSE investigators. A synthetic factor-Xa inhibitor (ORG31540/SR9017A) as
an adjunct to fibrinolysis in acute myocardial infarction. The PENTALYSE
study. Eur Heart J 2001; 22:1716–1724.
Dawes J, Pumphrey CW, McLaren KM, Prowse CV, Pepper DS. The in vivo release
of human platelet factor 4 by heparin. Thromb Res 1982; 27:65–76.
Denton J, Lane DA, Thunberg L, Slater AM, Lindahl U. Binding of platelet factor 4
to heparin oligosaccharides. Biochem J 1983; 209:455–460.
Elalamy I, Potevin F, Lecrubier C, Bara L, Marie JP, Samama MM. A fatal low-
molecular-weight heparin-associated thrombocytopenia after hip surgery: pos-
sible usefulness of PF4–heparin ELISA test. Blood Coagul Fibrinolysis 1996;
7:665–671.
Eriksson BI, Bauer KA, Lassen MR, Turpie AG. Fondaparinux compared with
enoxaparin for the prevention of venous thromboembolism after hip-fracture
surgery. N Engl J Med 2001; 345:1298–1304.
Eriksson BI, Lassen MR, PENTasaccharide in HIp-FRActure Surgery Plus In-
vestigators. Duration of prophylaxis against venous thromboembolism with
fondaparinux after hip fracture surgery: a multicenter, randomized, placebo-
controlled, double-blind study. Arch Intern Med 2003; 163:1337–1342.
Fareed J, Walenga JM, Hoppensteadt D, Haun X, Racanelli A. Comparative study
on the in vitro and in vivo activities of seven low-molecular-weight heparins.
Haemostasis 1988; 18(suppl 3):3–15.
Fondu P. Heparin-associated thrombocytopenia: an update. Acta Clin Belg 1995;
50:343–357.
216 Alban and Greinacher
I. INTRODUCTION
IIc
Molecular weight (kDa) 72 40 50–80
Extracellular Ig-like domains 3 2 2
Intracellular tyrosine motif d None ITAM(IIa, IIc) None
ITIM (IIb)
Noncovalent-associated subunits g-Chain None h-Chain, g-chain, ~-chain
Affinity constant 108M1 <107M1 IIIa: 3 107M1
IIIb: <107M1
IgG subclass aviditye 3=1 > 4{2 IIa-Arg131 3 > 1 { 2 > 4 IIIa-Val158 1 > 3 { 2,4
IIa-His131 3 > 1 = 2 > 4 IIIa-Val158 > IIIa-Phe158 for 1 and 3
IIb1 3 > 1 > 4 { 2 IIIb-NA1 > IIIb-NA2 for 1 and 3
Hematopoietic cell distribution CD34 progenitor cells, monocytes, IIa: platelets, endothelial cells, IIIa: monocytes, macrophages, NK
macrophages, dendritic cells monocytes, macrophages, cells, T cells
eosino-/baso-/neutrophils, IIIb: neutrophils
Langerhans/dendritic cells
IIb: B cells, monocytes
IIc: NK cells
a
Allelic polymorphisms that show differences in IgG binding; NA1/NA2 variants have multiple amino acid differences (Ory et al., 1989); SH+ individuals
(Bux et al., 1997) carry three copies of FcgRIIIB (Koene et al., 1998); lack of expression of IIc is due to a nonsense mutation (Metes et al., 1998).
b
Multiple mRNA transcripts from FcgRIB, IIA, and IIB, are the result of alternative splicing of primary transcripts.
c
Soluble forms of FcgRIIa and IIb (sIIa, sIIb) are devoid of the hydrophobic transmembrane exon; GPI, glycosylphosphatidylinositol.
d
ITAM, immunoreceptor tyrosine-based activation motif; ITIM, immunoreceptor tyrosine-based inhibition motif.
225
e
Numbers represent the relative order of IgG subclass binding to variants of FcgRIIa (Warmerdam et al., 1990), FcgRIIIa (Koene et al., 1997; Wu
et al., 1997), and FcgRIIIb (Salmon et al., 1990; Bredius et al., 1994b).
226 Denomme
males and female, among platelets from persons of different ages, or among
platelets representing the three possible genotypic classes of the FcgRIIa-Arg/
His131 allelic variants (Brandt et al., 1995).
Figure 1 Flow cytometric analysis of platelets activated with HIT sera, thrombin,
and calcium ionophore. Platelets (p) and microparticles (mp) were identified using
fluorescence (FL1, FITC anti-GPIba, y-axis) and size (forward scatter, x-axis). Nega-
tive controls included: (A) platelets incubated in buffer alone; (B) platelets incubated
with patients serum, which tested negative for HIT, in the presence of 0.1 U/mL
heparin; and (C) platelets incubated with HIT serum with no heparin added. Micro-
particles were generated by (D) platelets incubated with HIT serum in the presence of
0.1 U/mL heparin, as well as (E) thrombin-treated platelets (1 U/mL) and (F) calcium
ionophore-treated platelets (10 AM). The percentage of microparticles for each experi-
ment is shown in the lower left quadrant. (From Hughes et al., 2000.)
son and Anderson, 1990). Ligand binding leads to clustering of the FcgRIIa,
causing Src family protein tyrosine kinases to phosphorylate the ITAM re-
gion of FcgRIIa (Chacko et al., 1994; Huang et al., 1992). Following FcgRIIa
phosphorylation, additional tyrosine kinase activity (e.g., p72syk) increases
through the noncovalent interaction of their SH2 domains with the phos-
phorylated FcgRIIa ITAMs (Greinacher et al., 1994b; Yanaga et al., 1995;
Chacko et al., 1996). Subsequently, PLCg2 is phosphorylated by p72syk
(Blake et al., 1994); this phosphorylation is dependent on phosphatidylino-
sitol-trisphosphate [PtdIns(3,4,5)P3] (Gratacap et al., 1998). PLCg2 activa-
tion is crucial for the generation of DAG and IP3.
More recently, Gratacap and coworkers (2000) showed that FcgRIIa
activation alone does not produce sufficient levels of PtdIns(3,4,5)P3 to cause
PLCg2 activation, platelet release, and aggregation. Additionally, ADP re-
ceptor activation by Gi-protein signaling leads to the generation of PtdIns
(3,4,5)P3, which combined with activation of FcgRIIa generates optimal lev-
els of PtdIns(3,4,5)P3, leading to PLCg2 phosphorylation. Activated PLCg2
then generates DAG and IP3 from PtdIns(4,5)P2, mobilizing calcium, and
effecting platelet aggregation (Fig. 3). Moreover, lipid rafts appear to play an
gRIIa Numbers
B. Platelet Fcg
Variable expression of FcgRIIa numbers among individuals could affect
susceptibility to immune complex diseases (Rosenfeld et al., 1987), or even to
HIT. The number of platelet surface–expressed FcgRIIa molecules is in-
creased dramatically in patients with HIT (Chong et al., 1993b). However,
increased FcgRIIa expression is also seen after in vitro activation of platelets
by HIT antibodies. Thus, elevated FcgRIIa numbers may be a consequence of
platelet activation in HIT, rather than a proximate cause. This notion is sup-
ported by the fact that increased platelet FcgRIIa levels are seen in patients
with atherothrombosis and diabetes mellitus (Calverley et al., 2002). It re-
mains uncertain whether high baseline (pre-HIT) FcgRIIa numbers repre-
sents an important risk factor for HIT.
gRIIa
C. Plasma-Soluble Fcg
Soluble FcgRIIa, which is released from a-granules on platelet activation by
thrombin, has been demonstrated in plasma (Gachet et al., 1995). The soluble
form of the receptor lacks the amino acids for the transmembrane domain, a
result of alternative splicing that removes exon 5 from primary transcripts
(Rappaport et al., 1993). However, the relative amount of membrane versus
soluble FcgRIIa is fixed (Keller et al., 1993). Gachet and colleagues (1995)
reported that approximately 2 ng of soluble FcgRIIa is produced from 109
platelets. This value equals 2 ag, or 300 molecules, per platelet compared with
roughly 10 times as many molecules on the platelet surface. A much larger
amount of plasma-soluble FcgRIIa would be needed to inhibit significantly
PF4–H immune complexes from binding to platelet FcgRIIa. Therefore,
there is likely no effect of soluble FcgRIIa on membrane FcgRIIa-dependent
platelet activation, especially considering that immune complexes formed on
the platelet surface would sterically hinder soluble receptor interaction.
Moreover, plasma levels of soluble FcgRIIa are higher in patients with
HIT than in heparin-treated or other nonthrombocytopenic controls, pre-
sumably as a marker of in vivo platelet activation in HIT (Saffroy et al., 1997).
gRIIa-Arg/His131 Polymorphism
E. Fcg
There is an arginine-histidine (Arg/His) polymorphism at amino acid 131 of
the human FcgRIIa (Clark et al., 1989, Warmerdam et al., 1990). This allelic
variation affects the ability of human platelets to be activated by murine mono-
clonal IgG1 as well as by human IgG2 (Horsewood et al., 1991; Tomiyama et
al., 1992; Parren et al., 1992; Bachelot et al., 1995). This prompted Burgess et
al. (1995) to suggest that inherited FcgRIIa receptor variants could be a risk
factor for developing HIT. In a small cohort of patients, they found an over-
representation of the FcgRIIa–His131 variant. They hypothesized that IgG2
might be an important IgG subclass among HIT-IgG, as this could explain an
apparent association between HIT and the FcgRIIa–His131 variant.
However, subsequent reports argued against this hypothesis: IgG1
rather than IgG2 was the predominant subclass among HIT-IgG (Arepally
et al., 1997; Denomme et al., 1997; Suh et al., 1997). Nevertheless, in support
of a biological basis for a possible increased frequency of FcgRIIa–His131,
two groups found that HIT antibodies, including those that were predomi-
nantly IgG1, preferentially activated washed platelets of the His131 variant in
vitro (Denomme et al., 1997; Bachelot-Loza et al., 1998). However, Brandt et
al. (1995) found the opposite activation profile in platelet aggregation studies
using citrated platelet-rich plasma (i.e., the Arg131 variant was preferentially
activated by HIT plasmas). No consensus has emerged either among the six
studies that investigated whether one of the FcgRIIa–Arg/His131 phenotypes
predominated among patients with HIT: three studies show an overrepresen-
tation of FcgRIIa–His131 (Burgess et al., 1995; Brandt et al., 1995; Denomme
et al., 1997); two studies found no correlation with either variant (Arepally
Platelet Fc Receptor in HIT 235
et al., 1997; Bachelot-Loza et al., 1998); and one study (the largest) showed the
reverse correlation (Carlsson et al., 1998). This topic is considered in detail in
Section V.
preventing thrombosis. These data have implications for HIT, as there may be
a balance between platelet activation by HIT-IgG (predisposing to thrombo-
sis) and clearance of platelets by monocytes–macrophages (protecting some-
what against thrombosis).
Unlike mice, primate platelets possess FcgRIIa. Thus, a primate model
for HIT may be feasible, as suggested by a recent report (Ahmad et al., 2000).
The animals (Macaca mulatta) used do not express the human Arg–His poly-
morphism, perhaps explaining why less variability in platelet activation re-
sponse to HIT-IgG was observed in these in vitro studies. The primate model
may have value in evaluating therapeutic agents for HIT (Untch et al., 2002).
g Receptors in HIT
G. Monocyte Fcg
Monocytes and macrophages possess several different classes of FcgR (see
Table 1), and thus may play a part in influencing the frequency and severity of
both thrombocytopenia and thrombosis in HIT. One role, discussed in the
previous section, involves their potential to influence the balance between
platelet activation and reticuloendothelial-mediated platelet clearance in
HIT. Another function recently proposed for monocytes is that of contrib-
uting to the procoagulant state in HIT, (i.e., a role posited previously for
endothelial cells) (see Chap. 10). Pouplard and colleagues (2001) found that
by adding HIT-IgG and PF4 (or PF4–heparin) directly to isolated monocytes
or to whole blood, the monocytes produced tissue factor (TF), an effect that
could be inhibited by high concentrations of heparin. Arepally and Mayer
(2001) found that monocytes expressed surface TF when incubated with PF4
either in the presence of HIT-IgG or the HIT-mimicking murine monoclonal
antibody, KKO. Because monocytes express sulfated proteoglycans on their
surface, PF4 binding to monocytes can occur in the absence of added heparin.
These studies raise the possibility that monocytes play an important role in
the pathogenesis of the procoagulant state characteristic of HIT.
‘‘high responders’’ and the functional differences between the two polymor-
phic variants were later confirmed using other FcgRIIa-dependent assays,
such as erythrocyte antigen-rosetting, phagocytosis, and platelet activation
(Clark et al., 1989; Warmerdam et al., 1991; Parren et al., 1992; Salmon et al.,
1992). Murine monoclonal IgG1 antibodies activate platelets of all three Arg/
His131 phenotypes, but the homozygous FcgRIIa–Arg131 variant requires less
murine monoclonal antibody for platelet activation to occur.
The high-affinity binding of human IgG2 to FcgRIIa results when
histidine is substituted at amino acid 131 of the mature protein (Warmerdam
et al., 1991). FcgRIIa–His131 has a greater affinity for human IgG2, but a
lower affinity for murine IgG1. Therefore, the terms high and low responder,
used historically for the effects of murine monoclonal antibodies on Arg131
and His131 FcgRIIa phenotypes, respectively, is confusing, as the opposite
reaction profile is observed with human IgG2. The high/low responder ter-
minology has been largely replaced in favor of referring simply to the amino
acid polymorphism.
The FcgRIIa–Arg/His131 variant polymorphism can be determined in
three ways: (a) by functional assay, such as T-cell–dependent proliferation or
murine monoclonal antibody activation; (b) by specific binding using 41H16,
a monoclonal antibody the Fab of which binds exclusively to the FcgRIIa–
Arg131 variant; and (c) by molecular genotyping. Four DNA-based methods
have been developed to genotype for the FcgRIIa–Arg/His131 nucleotide
substitution (Clark et al., 1991; Osborne et al., 1994; Bachelot et al., 1995;
Jiang et al., 1996; Denomme et al., 1997). In one technique, the presence of the
FcgRIIa–Arg/His131 variant gene is determined using genomic DNA and a
sequence-specific primer–polymerase chain reaction (PCR) assay. Two PCR
reactions are necessary, each containing a common primer paired with a
unique primer having different 3V-ends to detect the presence of the G or A
variant nucleotide (Clark et al., 1991). This method has been modified using
different sequence-specific primers (Flesch et al., 1998) or using a nested
sequence-specific PCR (Carlsson et al., 1998). In a second technique, flank-
ing primers are used to amplify a region containing the nucleotide poly-
morphism, followed by dot-blotting and hybridization with allele-specific,
single-stranded oligonucleotide probes (Osborne et al., 1994; Burgess et al.,
1995; Denomme et al., 1997). In a third technique, Bachelot and coworkers
(1995) developed a denaturing gradient gel electrophoresis assay that distin-
guishes between the FcgRIIa–Arg/His131 variants also using flanking primers
that amplify a region containing the polymorphism. Last, restriction endo-
nuclease digestion of PCR-amplified genomic DNA has been developed using
one primer immediately proximal to the polymorphic site and containing a
mutation such that the polymorphism creates a restriction enzyme site for
only one of the alleles (Jiang et al., 1996).
238 Denomme
Predominant
FcgRIIa
Disease variant Comment
Figure 4 FcgRIIa–His131 gene frequencies in six studies of HIT are shown: The
first four studies were from North American centers, the last two from Europe.
Although the first three studies showed predominance of His131 in patients with HIT,
the last study showed predominance off Arg131 in patients with HIT complicated by
thrombosis. A complicating feature is the difference in gene frequencies between
certain control populations [e.g., between Denomme et al. (1997) and Carlsson et al.
(1998)]. Not shown in the figure is the significant difference between control patients
in the studies by Carlsson and Brandt ( p = 0.013).
patients). On the other hand, when Pouplard and colleagues (1999) examined
the Arg/His131 gene frequency among patients who formed antibodies against
PF4–heparin following cardiac surgery, they noted that platelet levels were
significantly lower only in the Arg/Arg131 group, when compared with
patients who did not form antibodies.
The explanation for the differences among these various studies is not
readily apparent. However, a complicating aspect is noted in Fig. 4: the fre-
Platelet Fc Receptor in HIT 241
ACKNOWLEDGMENTS
The author wishes to thank Dr. Lena E. Carlsson for her helpful review of
the manuscript. Some of the studies described were supported by a Career
Development Fellowship Award of the Canadian Blood Services. The
author was a Bayer/Canadian Blood Services/Medical Research Council
Scholar.
REFERENCES
Ahmad S, Jeske WP, Walenga JM, Aldabbagh A, Iqbal O, Fareed J. Human anti-
heparin-platelet factor 4 antibodies are capable of activating primate platelets:
towards the development of a HIT model in primates. Thromb Res 2000;
100:47–54.
Amiral J, Bridey F, Dreyfus M, Vissaco AM, Fressinaud E, Wolf M, Meyer D. Platelet
factor 4 complexed to heparin is the target for antibodies generated in heparin-
induced thrombocytopenia. Thromb Haemost 1992; 68:95–96.
Amiral J, Peynaud-Debayle E, Wolf M, Bridey F, Vissac A-M, Meyer D. Generation
of antibodies to heparin-PF4 complexes without thrombocytopenia in patients
treated with unfractionated or low-molecular-weight heparin. Am J Hematol
1996; 52:90–95.
Anderson GP, Anderson CL. Signal transduction by the platelet Fc receptor. Blood
1990; 76:1165–1172.
Anderson GP, van de Winkel JGJ, Anderson CL. Anti-GPIIb/IIIa (CD41)
monoclonal antibody-induced platelet activation requires Fc receptor-depend-
ent cell-cell interaction. Br J Haematol 1991; 79:75–83.
Arepally G, McKenzie SE, Jiang X-M, Poncz M, Cines DB. FcgRIIA H/R131
polymorphism, subclass-specific IgG anti-heparin/platelet factor 4 antibodies
and clinical course in patients with heparin-induced thrombocytopenia and
thrombosis. Blood 1997; 89:370–375.
Arepally GM, Mayer IM. Antibodies from patients with heparin-induced thrombo-
cytopenia stimulate monocytic cells to express tissue factor and secrete inter-
leukin-8. Blood 2001; 98:1252–1254.
Arepally GM, Kamei S, Park KS, Kamei K, Li ZQ, Liu W, Siegel DL, Kisiel W,
Cines DB, Poncz M. Characterization of a murine monoclonal antibody that
mimics heparin-induced thrombocytopenia antibodies. Blood 2000; 95:1533–
1540.
Bachelot C, Saffroy R, Gandrille S, Aiach M, Rendu F. Role of FcgRIIA gene
polymorphism in human platelet activation by monoclonal antibodies. Thromb
Haemost 1995; 74:1557–1563.
Bachelot-Loza C, Saffroy R, Lasne D, Chatellier G, Aiach M, Rendu F. Importance of
the FcgRIIa-Arg/His-131 polymorphism in heparin-induced thrombocytope-
nia diagnosis. Thromb Haemost 1998; 79:523–528.
Bevers EM, Comfurius P, Dekkers DW, Zwaal RF. Lipid translocation across the
plasma membrane of mammalian cells. Biochim Biophys Acta 1999; 1439:317–
330.
Blake RA, Asselin J, Walker T, Watson SP. Fc gamma receptor II stimulated for-
mation of inositol phosphates in human platelets is blocked by tyrosine kinase
inhibitors and associated with tyrosine phosphorylation of the receptor. FEBS
Lett 1994; 342:15–18.
Blank M, Cines DB, Arepally G, Eldor A, Afek A, Shoenfeld Y. Pathogenicity of
human antiplatelet factor 4/heparin in vivo: generation of mouse anti-PF4/
heparin and induction of thrombocytopenia by heparin. Clin Exp Immunol
1997; 108:333–339.
Blank M, Eldor A, Tavor S, Ziporen L, Cines DB, Arepally G, Afek A, Shoenfeld Y. A
Platelet Fc Receptor in HIT 243
Kelton JG, Smith JW, Santos AV, Murphy WG, Horsewood P. Platelet IgG Fc
receptor. Am J Hematol 1987; 25:299–310.
Kelton JG, Sheridan D, Santos A, Smith J, Steeves K, Smith C, Brown C, Murphy
WG. Heparin-induced thrombocytopenia: laboratory studies. Blood 1988; 72:
925–930.
Kelton JG, Smith JW, Warkentin TE, Hayward CPM, Denomme GA, Horsewood P.
Immunoglobulin G from patients with heparin-induced thrombocytopenia
binds to a complex of heparin and platelet factor 4. Blood 1994; 83:3232–3239.
Khairy M, Lasne D, Brohard-Bohn B, Aich M, Rendu F, Bachelot-Loza C. A
new approach in the study of the molecular and cellular events implicated
in heparin-induced thrombocytopenia. Formation of leukocyte-platelet aggre-
gates. Thromb Haemost 2001; 85:1090–1096.
King M, McDermott P, Schreiber AD. Characterization of the Fc-gamma receptor on
human platelets. Cell Immunol 1990; 128:462–479.
Koene HR, Kleijer M, Algra J, Roos D, von dem Borne AEGK, de Haas M.
FcgRIIIa-158V/F polymorphism influences the binding of IgG by natural killer
cell FcgRIIIa, independently of the FcgRIIIa-48L/R/H phenotype. Blood 1997;
90:1109–1114.
Koene HR, Kleijer M, Roos D, de Haas M, von dem Borne AEGK. FcgRIIIB gene
duplication: evidence for presence and expression of three distinct FcgRIIIB
genes in NA(1+,2+)SH(+) individuals. Blood 1998; 91:673–679.
Lebrazi J, Helft G, Abdelouahed M, Elalamy I, Mirshahi M, Samama MM, Lecompte
T. Human anti-streptokinase antibodies induce platelet aggregation in an Fc
receptor (CD32) dependent manner. Thromb Haemost 1995; 74:938–942.
Lee DP, Warkentin TE, Denomme GA, Hayward CPM, Kelton JG. A diagnostic test
for heparin-induced thrombocytopenia: detection of platelet microparticles
using flow cytometry. Br J Haematol 1996; 95:724–731.
Lehrnbecher T, Foster CH, Zhu S, Leitman SF, Goldin LR, Huppi K, Chanock SJ.
Variant genotypes of the low-affinity Fcgamma receptors in two control popu-
lations and a review of low-affinity Fcgamma receptor polymorphisms in con-
trol and disease populations. Blood 1999; 94:4220–4232.
McCrae KR, Shattil SJ, Cines DB. Platelet activation induces increased Fcg receptor
expression. J Immunol 1990; 144:3920–3927.
McKenzie SE, Taylor SM, Malladi P, Yuhan H, Cassel DL, Chien P, Schwartz E,
Schreiber AD, Surrey S, Reilly MP. The role of the human Fc receptor FcgRIIA
in the immune clearance of platelets: a transgenic mouse model. J Immunol
1999; 162:4311–4318.
Metes D, Ernst LK, Chambers WH, Sulica A, Herberman RB, Morel PA. Expression
of functional CD32 molecules on human NK cells is determined by an allelic
polymorphism of the FcgammaRIIC gene. Blood 1998; 91:2369–2380.
Newman PM, Chong BH. Heparin-induced thrombocytopenia: new evidence for the
dynamic binding of purified anti-PF4–heparin antibodies to platelets and the
resultant platelet activation. Blood 2000; 96:182–187.
Norris CF, Pricop L, Millard S, Taylor SM, Surrey S, Schwartz E, Salmon JE,
McKenzie SE. A naturally occurring mutation in FcgRIIA: a Q to K127 change
Platelet Fc Receptor in HIT 247
confers unique IgG binding properties to the R131 allelic form of the receptor.
Blood 1998; 91:656–662.
Ory PA, Clark MA, Kwoh EE, Clarkson SB, Goldstein IM. Sequences of comple-
mentary DNAs that encode the NA1 and NA2 forms of Fc receptor III on
human neutrophils. J Clin Invest 1989; 84:1688–1691.
Osborne JM, Chacko GW, Brandt JT, Anderson CL. Ethnic variation in frequency of
an allelic polymorphism of human FcgRIIa determined with allele specific
oligonucleotide probes. J Immunol Methods 1994; 173:207–217.
Parren PWHI, Warmerdam PAM, Boeije LCM, Arts J, Westerdaal NAC, Vlug A,
Capel PJA, Aarden LA, van de Winkel JGJ. On the interaction of IgG sub-
classes with the low affinity Fc gamma RIIa (CD32) on human monocytes,
neutrophils, and platelets. Analysis of a functional polymorphism to human
IgG2. J Clin Invest 1992; 90:1537–1546.
Pfueller SL, David R. Different platelet specificities of heparin-dependent plate-
let aggregating factors in heparin-associated immune thrombocytopenia. Br J
Haematol 1986; 64:149–159.
Polgár J, Eichler P, Greinacher A, Clemetson KJ. Adenosine diphosphate (ADP) and
ADP receptor play a major role in platelet activation/aggregation induced by
sera from heparin-induced thrombocytopenia patients. Blood 1998; 91:549–
554.
Pouplard C, May MA, Iochmann S, Amiral J, Marchand M, Gruel Y. Antibodies
to platelet factor 4–heparin after cardiopulmonary bypass in patients anti-
coagulated with unfractionated heparin or a low-molecular-weight heparin:
clinical implications for heparin-induced thrombocytopenia. Circulation 1999;
99:2530–2536.
Pouplard C, Iochmann S, Renard B, Herault O, Gruel Y. Induction of monocyte tissue
factor expression by antibodies to heparin-platelet factor 4 complexes developed
in heparin-induced thrombocytopenia. Blood 2001; 97:3300–3302.
Qiu WQ, de Bruin D, Brownstein BH, Pearse R, Ravetch JV. Organization of the
human and mouse low-affinity FcgR genes: duplication and recombination.
Science 1990; 248:732–735.
Rappaport EF, Cassel DL, Walterhouse DO, McKenzie SE, Surrey S, Keller MA,
Schreiber AD, Schwartz E. A soluble form of the human Fc receptor FcgRIIa:
cloning, transcript analysis and detection. Exp Hematol 1993; 21:689–696.
Rascu A, Repp R, Westerdaal NAC, Kalden JR, van de Winkel JGJ. Clinical rele-
vance of Fcg receptor polymorphisms. Ann NY Acad Sci 1997; 815:282–295.
Regnault V, de Maistre E, Carteaux JP, Gruel Y, Nguyen P, Tardy B, Lecompte T.
Platelet activation induced by antibodies to interleukin-8. Blood 2003; 101:
1419–1421.
Reilly MP, Taylor SM, Hartman NK, Arepally GM, Cines DB, Poncz M, McKen-
zie SE. Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse
model requires human platelet factor 4 and platelet activation through Fcg-
RIIA. Blood 2001; 98:2442–2447.
Rhodes GR, Dixon RH, Silver D. Heparin induced thrombocytopenia with throm-
botic and hemorrhagic manifestations. Surg Gynecol Obstet 1973; 136:409–416.
248 Denomme
Roberts B, Rosato FE, Rosato EF. Heparin: a cause of arterial emboli? Surgery 1964;
55:803–808.
Roitt I, Brostoff J, Male D, Eds. Immunology. London: Gower Medical Publishing,
1985.
Rosenfeld SI, Looney RJ, Leddy JP, Phipps DC, Abraham GN, Anderson CL.
Human platelet Fc receptor for immunoglobulin G Identification as a 40,000-
molecular-weight membrane protein shared by monocytes. J Clin Invest 1985;
76:2317–2322.
Rosenfeld SI, Ryan DH, Looney RJ, Anderson CL, Abraham GN, Leddy JP.
Human Fcg receptors: stable inter-donor variation in quantitative expression
on platelets correlates with functional responses. J Immunol 1987; 144:3920–
3927.
Rubinstein E, Boucheix C, Worthington RE, Carroll RC. Anti-platelet antibody
interactions with Fcg receptor. Semin Thromb Hemost 1995; 21:10–22.
Saffroy R, Bachelot-Loza C, Fridman WH, Aiach M, Teullaud J-L, Rendu F. Plasma
levels of soluble Fcg receptors II (sCD32) and III (sCD16) in patients with
heparin-induced thrombocytopenia. Thromb Haemost 1997; 78:970–971.
Salem HH, Van der Weyden MB. Heparin-induced thrombocytopenia. Variable
platelet-rich plasma reactivity to heparin-dependent aggregating factor. Pathol-
ogy 1983; 15:297–299.
Salmon JE, Edberg JC, Kimberly RP. Fcg receptor III on human neutrophils. J Clin
Invest 1990; 85:1287–1295.
Salmon JE, Edberg JC, Brogle NL, Kimberly RP. Allelic polymorphism of human Fcg
receptor IIA and Fcg receptor IIIB. Independent mechanisms for differences in
human phagocyte function. J Clin Invest 1992; 89:1274–1281.
Sanders LAM, van de Winkel JGJ, Rijkers GT, Voorhorst-Ogink MM, de Haas M,
Capel PJA, Zegers BJM. Fc gamma receptor IIa (CD32) heterogeneity in
patients with recurrent bacterial respiratory tract infections. J Infect Dis 1994;
170:854–861.
Sedmak DD, Davis DH, Singh U, van de Winkel JG, Anderson CL. Expression of IgG
Fc receptor antigens in placenta and on endothelial cells in humans. An im-
munohistochemical study. Am J Pathol 1991; 138:175–181.
Sheridan D, Carter C, Kelton JG. A diagnostic test for heparin-induced thrombo-
cytopenia. Blood 1986; 67:27–30.
Sims PJ, Wiedmer T, Esmon CT, Weiss HJ, Shattil SJ. Assembly of the platelet
prothrombinase complex is linked to vesiculation of the platelet plasma mem-
brane. Studies in Scott syndrome: an isolated defect in platelet procoagulant
activity. J Biol Chem 1989; 264:17049–17057.
Su K, Wu J, Edberg JC, McKenzie SE, Kimberly RP. Genomic organization of
classical human low-affinity Fcg receptor genes. Genes Immun 2002; 3(suppl 1):
S51–S56.
Suh JS, Malik MI, Aster RH, Visentin GP. Characterization of the humoral response
in heparin-induced thrombocytopenia. Am J Hematol 1997; 54:196–201.
Taylor SM, Reilly MP, Schreiber AD, Chien P, Tuckosh JR, McKenzie SE. Throm-
bosis and shock induced by activating antiplatelet antibodies in human Fcg-
Platelet Fc Receptor in HIT 249
RIIA transgenic mice: the interplay among antibody, spleen, and Fc receptor.
Blood 2000; 96:4254–4260.
Tax WJM, Williams HW, Reckers PPM, Capel PJA, Keone RAP. Polymorphism in
mitogenic effect of IgG1 monoclonal antibodies against T3 antigen on human T
cells. Nature 1983; 304:445.
Tax WJM, Hermes FFM, Willems RW, Capel JA, Koene RAP. Fc receptors for
mouse IgG 1 on human monocytes: polymorphism and role in antibody-
induced T cell proliferation. J Immunol 1984; 133:1185–1189.
Tomer A. A sensitive and specific functional flow cytometric assay for the diagnosis of
heparin-induced thrombocytopenia. Br J Haematol 1997; 98:648–656.
Tomiyama Y, Kunicki TJ, Zipf TF, Ford SB, Aster RH. Response of human platelets
to activating monoclonal antibodies: importance of FcgRII (CD32) phenotype
and level of expression. Blood 1992; 80:2261–2268.
Untch B, Ahmad S, Messmore HL, Schultz CL, Ma Q, Hoppensteadt DA, Walenga
JM, Fareed J. Development of a non-human primate sub-clinical model of
heparin-induced thrombocytopenia: platelet response to human anti-heparin-
platelet factor 4 antibodies. Thromb Res 2002; 106:149–156.
van de Winkel JGJ, Capel PJA. Human IgG Fc receptor heterogeneity: molecular
aspects and clinical implications. Immunol Today 1993; 14:215–221.
Visentin GP, Ford SE, Scott JP, Aster RH. Antibodies from patients with heparin-
induced thrombocytopenia/thrombosis are specific for platelet factor 4 com-
plexed with heparin or bound to endothelial cells. J Clin Invest 1994; 93:81–
88.
Warkentin TE, Sheppard JI. Generation of platelet-derived microparticles and pro-
coagulant activity by heparin-induced thrombocytopenia IgG/serum and other
IgG platelet agonists: a comparison with standard platelet agonists. Platelets
1999; 10:319–326.
Warkentin TE, Hayward CPM, Smith CA, Kelly PM, Kelton JG. Determinants of
donor platelet variability when testing for heparin-induced thrombocytopenia. J
Lab Clin Med 1992; 120:371–379.
Warkentin TE, Hayward CPM, Boshkov LK, Santos AV, Sheppard JI, Bode AP,
Kelton JG. Sera from patients with heparin-induced thrombocytopenia gen-
erate platelet-derived microparticles with procoagulant activity: an explanation
for the thrombotic complications of heparin-induced thrombocytopenia. Blood
1994; 84:3691–3699.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Tech M, Gent M,
Kelton JG. Heparin-induced thrombocytopenia in patients treated with low-
molecular-weight heparin or unfractionated heparin. N Engl J Med 1995; 332:
1330–1335.
Warmerdam PAM, van de Winkel JGJ, Gosselin EJ, Capel PJA. Molecular basis for
a polymorphism of human Fcg receptor II (CD32w). J Exp Med 1990; 172:19–
25.
Warmerdam PAM, van de Winkel JGJ, Vlug A, Westerdaal NAC, Capel PJA. A
single amino acid in the second domain of the human Fcg receptor II is critical
for human IgG2 binding. J Immunol 1991; 147:1338–1343.
250 Denomme
Weismann RE, Tobin RW. Arterial embolism occurring during systemic heparin
therapy. Arch Surg 1958; 76:219–227.
Wu J, Edberg JC, Redecha PB, Bansal V, Guyre PM, Coleman K, Salmon JE,
Kimberly RP. A novel polymorphism of FcgRIIIa (CD16) alters receptor
function and predisposes to autoimmune disease. J Clin Invest 1997; 100:1059–
1070.
Yanaga F, Poole A, Asselin J, Blake R, Schieven GL, Clark EA, Law CL, Watson SP.
Syk interacts with tyrosine-phosphorylated proteins in human platelets acti-
vated by collagen and cross-linking of the Fc gamma-IIA receptor. Biochem J
1995; 313:471–478.
10
Immune Vascular Injury in Heparin-
Induced Thrombocytopenia
Gowthami M. Arepally
Duke University Medical Center, Durham, North Carolina, U.S.A.
Mortimer Poncz
University of Pennsylvania School of Medicine and Children’s Hospital
of Philadelphia, Philadelphia, Pennsylvania, U.S.A.
Douglas B. Cines
University of Pennsylvania School of Medicine, Philadelphia,
Pennsylvania, U.S.A.
I. INTRODUCTION
251
252 Arepally et al.
Figure 1 Model of HIT antibody interactions with endothelial cells: (1) HIT
antibodies bind to circulating antigen that becomes localized to platelets. (2) Platelet
activation occurs after Fc receptor binding, leading to platelet granule release. ADP
is released from platelet dense granules and PF4 is released from platelet a-granules.
(3) Released PF4 binds to platelets and endothelial cell heparan sulfate (HS), dis-
placing antithrombin (AT) from endothelial cells. (4) Antigen complexes on endo-
thelial cells bind HIT antibodies. (5) HIT antibody binding to endothelial cells leads
to endothelial cell activation, resulting in further platelet activation.
Immune Vascular Injury in HIT 255
1985). The affinity of PF4 binding to ECs is lower than to purified heparin (Kd
= 2 nM vs. 2–3 AM, respectively) (Rybak et al., 1989), consistent with the
biochemical hetrogeneity of vascular matrix. PF4 has a 10- to 100-fold greater
affinity for EC HSPG than does antithrombin (Jordan et al., 1982) and
markedly attenuates its antiprotease cofactor activity on intact vessels (Busch
et al., 1980; Stern et al., 1985).
PF4 also binds to the chemokine receptor, Duffy (Hadley et al., 1994),
which has been identified on ECs in postcapillary venules and in the
splanchnic bed, even in individuals who do not express the antigen on their
erythrocytes (Peiper et al., 1995). The distribution of Duffy on ECs in other
vascular beds is less well studied. The binding site on PF4 for Duffy has not
been deduced, and the capacity of Duffy-bound PF4 to bind heparin or HIT
antibodies has not been reported. PF4 does not appear to bind to any of the
other members of the chemokine receptor family (for review see Baggiolini et
al., 1997), including CXCR4, the only other related receptor yet identified on
ECs (Volin et al., 1998). However, PF4 exposed to leukocyte lysates is cleaved
between Thr-16 and Ser-17 to yield a peptide with 30-fold greater biological
activity than its parent molecule (Gupta et al., 1995). Such proteolysis may
enable modified PF4 to bind to CXCR1 or R2 (formerly called IL-8R-a and -
h, respectively) (for review see Baggiolini et al., 1997), but no evidence has
been presented that ECs express either receptor, nor has binding of modified
PF4 to CXCR4 been shown.
The CXC chemokines that contain a Glu-Leu-Arg (ELR) NH2-termi-
nal sequence induce angiogenesis, whereas those that do not, such as PF4 and
gIP-10, inhibit angiogenesis (Baggiolini and Moser, 1997). The molecular
basis by which PF4 inhibits angiogenesis is unclear. Considering that ECs lack
CXCR1 and CXCR2, the effect of PF4 may be indirect. PF4 interrupts the
binding of heparin-binding proteins, such as vascular endothelial cell growth
factor (VEGF) and basic fibroblast growth factor (hFGF) to their receptors
(Gengrinovitch et al., 1995; Peng et al., 1997). However, PF4 also inhibits
angiogenesis stimulated by a truncated version of VEGF that lacks the hepa-
rin-binding domain (Gengrinovitch et al., 1995). Whether PF4 influences the
behavior of ECs from disordered vessels in other ways, or whether the PF4-
mediated signal transduction through chemokine receptors is modulated by
HIT antibodies, deserves additional study.
PF4 binds preferentially to ECs at sites of angiogenesis (Hansell et al.,
1995). For example, in a hamster cheek pouch model, [125I]PF4 is taken up
preferentially by ECs within the neovasculature where its binding is inhibited
by the related CXC chemokine, gIP-10 (Luster et al., 1995). PF4 inhibits cell
proliferation in vitro (Sharpe et al., 1990; Maione et al., 1991; Gupta and
Singh, 1994) and tumor-induced neovascularization (Sharpe et al., 1990;
Maione et al., 1991) in vivo. Both the heparin-binding domain (Maione et al.,
258 Arepally et al.
1990; Gupta and Singh, 1994) and other portions of the molecule (Maione et
al., 1991; Gupta and Singh, 1994; Gupta et al., 1995; Lecomte-Raclet et al.,
1998) have been implicated.
PF4 also binds to thrombomodulin (TM), a 60.3 kDa protein consti-
tutively expressed on the surface of ECs. Binding of thrombin to TM alters its
substrate specificity, such that proteolytic cleavage of protein C is accelerated
20,000-fold (Esmon, 1989). TM is posttranslationally modified by association
with a chondroitin sulfate A–like GAG, which invests it with the capacity to
bind cationic peptides at physiological pH. The binding of eosinophilic
cationic protein, major basic protein, and histidine-rich glycoprotein to these
GAG residues inhibits the function of TM, whereas the binding of PF4 (but
not h-thromboglobulin or thrombospondin) increases protein C-cofactor
activity 25-fold in a cell-free system (Slungaard and Key, 1994; Dudek et al.,
1997). The results of one recent study suggest that PF4 may exert a
physiogically relevant anticoagulant effect (Slungaard et al., 2003). Addition
of PF4 to cultured endothelial cells accelerates APC generation approximate-
ly 5- to 10-fold depending on vascular origin. Injection of PF4 into primates
infused with thrombin increases APC generation 2- to 3-fold and prolongs the
baseline aPTT. Additional studies should clarify whether HIT antibodies
interfere with the anticoagulant function of PF4 and thereby may predispose
to warfarin-associated venous limb gangrene.
Recent in vitro and in vivo studies suggest potential mechanisms
whereby PF4 may play a vital role in promoting atherogenesis. Human ath-
erosclerotic lesions are invested with PF4 (Pitsilos et al., 2003). PF4 is found
not only along the overlying endothelium, but also in foam cells and acellular
portions of the plaque. In vitro, PF4 binds to the LDL receptor and to
proteoglycans, forming ternary complexes that show limited migration into
clathrin-coated pits, thereby retarding endocytosis and catabolism of LDL
(Sachais et al., 2002). PF4 also binds directly to oxidized LDL, promoting
foam cell formation (Nassar et al., 2003). In mice, activated platelets deposit
PF4 on endothelium and monocytes, potentiating effects of P-selectin on plate-
let-leukocyte aggregate formation and atherosclerotic development (Huo et
al., 2003). Antibodies to heparin–PF4 complexes have recently been identified
as an independent predictor of myocardial infarction at 30 days in patients
presenting with acute coronary ischemic syndromes (Williams et al., 2003).
Thus, HIT antibodies may modify the interactions of PF4 with diseased
endothelium by (1) binding to PF4/proteoglycan complexes in atherosclerotic
lesions, (2) inducing formation of platelet-leukocyte aggregates (Khairy et al.,
2001), or (3) binding to circulating monocytes (Pouplard et al., 2001; Arepally
and Mayer, 2001), thereby increasing local inflammation and stimulating
procoagulant processes.
Immune Vascular Injury in HIT 259
Figure 2 Binding of IgG (A–D) and IgM (E) from the plasma of a patient with
HIT to cultured human umbilical vein endothelial cells (HUVEC): (A) Plasma alone;
(B) plasma plus 0.05 U/mL heparin; (C) plasma plus 10 Ag/mL PF4; (D) plasma plus
0.05 U/mL heparin plus 10 Ag/mL PF4; (E) binding of IgM from plasma containing
10 Ag/mL PF4. Both IgG (C) and IgM (E) bound to HUVEC in the presence of PF4
alone. The binding of each was completely inhibited by heparin. (From Visentin et
al., 1994.)
platelet fibrin(ogen) and release of ADP in the process. HIT sera/IgG exerted
little effect on the behavior of HUVEC in the absence of prestimulation by
platelets or cytokines, even in the presence of exogenous PF4. The mecha-
nism(s) by which platelet or cytokine activation facilitate binding of HIT
antibodies to cell-associated heparin/PF4 requires further investigation.
The studies outlined above support the notion that platelet activation and/or
an inflammatory milieu contribute to endothelial cell dysfunction, predispos-
ing to HIT-associated thrombosis (HITT). This view of HIT as an inflam-
matory disorder has gained experimental support through recent findings of
monocyte activation. HIT plasma or IgG stimulates monocytes to elaborate
tissue factor–dependent procoagulant activity in monocytes (Pouplard et al.,
2001). This procoagulant effect required small amounts of heparin to acti-
vate the monocytes in whole blood, but appeared to be heparin-independent
when isolated mononuclear cells were studied, presumably by exposing cell-
associated proteoglycans (Pouplard et al., 2001). Heparin-independent upreg-
ulation of monocyte tissue factor activity by HITT antibodies was confirmed
in a second study using human and murine antibodies (Arepally and Mayer,
2001). In this latter study, maximal tissue factor expression was detected at 4–
6 hours, suggesting that new synthesis, rather than de-encryption, was
primarily responsible for increased procoagulant activity. Activation of
monocyes likely requires ligation of cellular FcgRII receptors, as signaling
intermediates downstream of this receptor become phosphorylated in the
presence of HITT antibody (Ma and Arepally, 2002). The development of
monoclonal antibodies against PF4 and PF4/heparin (Arepally et al., 2000)
and the availability of a murine model of HITT should help to delineate the
contribution of monocyte activation to the immune pathogenesis of throm-
bocytopenia and thrombosis.
REFERENCES
Aird WC. The role of endothelium in severe sepsis and multiple organ dysfunction
syndrome. Blood 2003; 101:3765–3777.
Aird WC, Edelberg JM, Weiler-Guettler H, Simmons WW, Smith TW, Rosenberg
RD. Vascular bed-specific expression of an endothelial cell gene is programmed
by the tissue microenvironment. J Cell Biol 1997; 138:1117–1124.
Amiral J, Wolf M, Fischer A-M, Boyer-Neumann C, Vissac A-M, Meyer D. Patho-
genicity of IgA and/or IgM antibodies to heparin–PF4 complexes in patients
with heparin-induced thrombocytopenia. Br J Haematol 1996; 92:954–959.
Arepally G, McKenzie SE, Jiang X-M, Poncz M, Cines DB. FcgRIIA H/R131
polymorphism, subclass-specific IgG anti-heparin/platelet factor 4 antibodies
and clinical course in patients with heparin-induced thrombocytopenia and
thrombosis. Blood 1997; 89:370–375.
Arepally GM, Kamei S, Park KS, Kamei K, Li ZQ, Liu W, Siegel DL, Kisiel W, Cines
DB, Poncz M. Characterization of a murine monoclonal antibody that mimics
heparin-induced thrombocytopenia antibodies. Blood 2000; 95:1533–1540.
Arepally GM, Mayer IM. Antibodies from patients with heparin-induced thrombo-
cytopenia stimulate monocytic cells to express tissue factor and secrete inter-
leukin-8. Blood 2001; 98:1252–1254.
Bachelot-Loza C, Saffroy R, Lasne D, Chatellier G, Aiach M, Rendu F. Importance of
the FcgRIIa-Arg/His-131 polymorphism in heparin-induced thrombocytope-
nia diagnosis. Thromb Haemost 1998; 79:523–528.
Baggiolini M, Moser B. Blocking chemokine receptors. J Exp Med 1997; 186:1189–
1191.
Baggiolini M, Dewald B, Moser B. Human chemokines: an update. Annu Rev
Immunol 1997; 15:675–705.
Barber AJ, Käser-Glanzmann R, Jakábová M, Lüscher EF. Characterization of
a chondroitin 4-sulfate proteoglycan carrier for heparin neutralizing activity
(platelet factor 4) released from human blood platelets. Biochim Biophys Acta
1972; 286:312–329.
Bassenge E. Endothelial function in different organs. Prog Cardiovasc Dis 1996; 39:
209–228.
Bauer TL, Arepally G, Konkle BA, Mestichelli B, Shapiro SS, Cines DB, Poncz M,
McNulty S, Amiral J, Hauck WW, Edie RN, Mannion JD. Prevalence of hepa-
264 Arepally et al.
Gupta SK, Hassel T, Singh JP. A potent inhibitor of endothelial cell proliferation is
generated by proteolytic cleavage of the chemokine platelet factor 4. Proc Natl
Acad Sci USA 1995; 92:7799–7803.
Hadley TJ, Lu Z-H, Wasniowska K, Martin AW, Peiper SC, Hesselgesser J, Horuk R.
Postcapillary venule endothelial cells in kidney express a multispecific chemo-
kine receptor that is structurally and functionally identical to the erythroid iso-
form, which is the Duffy blood group antigen. J Clin Invest 1994; 94:985–991.
HajMohammadi S, Enjyoji K, Princivalle M, Christi P, Lech M, Beeler D, Rayburn H,
Schwartz JJ, Barzegar S, de Agostini Al, Post MJ, Rosenberg RD, Shworak
NW. Normal levels of anticoagulant heparan sulfate are not essential for normal
hemostasis. J Clin Invest 2003; 111:989–999.
Hansell P, Maione TE, Borgström P. Selective binding of platelet factor 4 to regions of
active angiogenesis in vivo. Am J Physiol 1995; 269:H829–H836.
Hebert JM, Savi P, Jeske WP, Walenga JM. Effect of SR121566A, a potent GP IIb–
IIIa antagonist, on the HIT serum/heparin-induced platelet-mediated activa-
tion of human endothelial cells. Thromb Haemost 1998; 80:326–331.
Huo Y, Schober A, Forlow SB, Smith DF, Hyman MC, Jung S, Littman DR, Weber
C, Ley K. Circulating activated platelets exacerbate atherosclerosis in mice
deficient in apolipoprotein E. Nat Med 2003; 9:61–67.
Jordan RE, Favreau LV, Braswell EH, Rosenberg RD. Heparin with two binding sites
for antithrombin or platelet factor 4. J Biol Chem 1982; 257:400–406.
Khairy M, Lasne D, Brohard-Bohn B, Aiach M, Rendu F, Bachelot-Loza C. A new
approach in the study of the molecular and cellular events implicated in heparin-
induced thrombocytopenia. Formation of leukocyte-platelet aggregates.
Thromb Haemost 2001; 85:1090–1096.
Kinsella MG, Wight TN. Modulation of sulfated proteoglycan synthesis by bovine
aortic endothelial cells during migration. J Cell Biol 1986; 102:679–687.
Laszik Z, Mitro A, Taylor FB Jr, Ferrell G, Esmon CT. Human protein C receptor is
present primarily on endothelium of large blood vessels: implications for the
control of the protein C pathway. Circulation 1997; 96:3633–3640.
Lecomte-Raclet L, Alemany M, Sequeira-Le Grand A, Amiral J, Quentin G, Vissac
AM, Caen JP, Han ZC. New insights into the negative regulation of hema-
topoiesis by chemokine platelet factor 4 and related peptides. Blood 1998;
91:2772–2780.
Leung DYM, Moake JL, Havens PL, Kim M, Pober JS. Lytic anti-endothelial cell
antibodies in haemolytic-uraemic syndrome. Lancet 1988; 2:183–186.
Levin EG, Del Zoppo GJ. Localization of tissue plasminogen activator in endothelium
of a limited number of vessels. Am J Pathol 1994; 144:855–861.
Loscalzo J, Melnick B, Handin RI. The interaction of platelet factor four and
glycosaminoglycans. Arch Biochem Biophys 1985; 240:446–455.
Lowe-Krentz LJ, Joyce JG. Venous and aortic porcine endothelial cells cultured under
standard conditions synthesize heparan sulfate chains which differ in charge.
Anal Biochem 1991; 193:155–163.
Lüscher TF, Barton M. Biology of the endothelium. Clin Cardiol 1997; 20(suppl 2):II-
3–II-10.
Immune Vascular Injury in HIT 267
Luster AD, Greenberg SM, Leder P. The IP-10 chemokine binds to a specific cell
surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial
cell proliferation. J Exp Med 1995; 182:219–231.
Ma AD, Arepally G. HIT antibodies and monocyte signaling [abstr]. Blood 2002;
100:16a.
Maione TE, Gray GS, Petro J, Hunt AJ, Donner AL, Bauer SI, Carson HF, Sharpe
RJ. Inhibition of angiogenesis by recombinant human platelet factor-4 and
related peptides. Science 1990; 247:77–79.
Maione TE, Gray GS, Hunt AJ, Sharpe RJ. Inhibition of tumor growth in mice by an
analogue of platelet factor 4 that lacks affinity for heparin and retains potent
angiostatic activity. Cancer Res 1991; 51:2077–2083.
Marcum JA, Rosenberg RD. Anticoagulantly active heparin-like molecules from
vascular tissue. Biochemistry 1984; 23:1730–1737.
Marcum JA, Rosenberg RD. Anticoagulantly active heparan sulfate proteoglycan and
the vascular endothelium. Semin Thromb Hemost 1987; 13:464–474.
Marcum JA, McKenney JB, Rosenberg RD. The acceleration of thrombin–
antithrombin complex formation in rat hindquarters via heparinlike molecules
bound to the endothelium. J Clin Invest 1984; 74:341–350.
McCrae KR, DeMichele A, Samuels P, Roth D, Kuo A, Meng Q-H, Rauch J, Cines
DB. Detection of endothelial cell-reactive immunoglobulin in patients with anti-
phospholipid antibodies. Br J Haematol 1991; 79:595–605.
Meroni PL, Del Papa N, Borghi MO. Antiphospholipid and antiendothelial
antibodies. Int Arch Allergy Immunol 1996; 111:320–325.
Nader HB, Buonassisi V, Colburn P, Dietrich CP. Heparin stimulates the synthesis
and modifies the sulfation pattern of heparan sulfate proteoglycan from
endothelial cells. J Cell Physiol 1989; 140:305–310.
Nakamura N, Ban T, Yamaji K, Yoneda Y, Wada Y. Localization of the apoptosis-
inducing activity of lupus anticoagulant in an annexin V-binding subset. J Clin
Invest 1998; 101:1951–1959.
Nassar T, Sachais BS, Akkawi S, Kowalska MA, Bdeir K, Leitersdorf E, Hiss E,
Ziporen L, Aviram M, Cines D, Poncz M, Higazi AA. Platelet factor 4 enhances
the binding of oxidized low-density lipoprotein to vascular wall cells. J Biol
Chem 2003; 278:6187–6193.
Nishinaga M, Ozawa T, Shimada K. Homocysteine, a thrombogenic agent, suppresses
anticoagulant heparan sulfate expression in cultured porcine aortic endothelial
cells. J Clin Invest 1993; 92:1381–1386.
Patton WA II, Granzow CA, Getts LA, Thomas SC, Zotter LM, Gunzel KA, Lowe-
Krentz LJ. Identification of a heparin-binding protein using monoclonal anti-
bodies that block heparin binding to porcine aortic endothelial cells. Biochem J
1995; 311:461–469.
Peiper SC, Wang Z-X, Neote K, Martin AW, Showell HJ, Conklyn MJ, Ogborne K,
Hadley TJ, Lu Z-H, Hesselgesser J, Horuk R. The Duffy antigen/receptor for
chemokines (DARC) is expressed in endothelial cells of Duffy negative indi-
viduals who lack the erythrocyte receptor. J Exp Med 1995; 181:1311–1317.
Peng H, Wen TC, Igase K, Tanaka J, Matsuda S, Aburaya J, Sakanaka M. Sup-
268 Arepally et al.
Theodore E. Warkentin
McMaster University and Hamilton Regional Laboratory Medicine Program,
Hamilton, Ontario, Canada
Andreas Greinacher
Ernst-Moritz-Amdt University Greifswald, Greifswald, Germany
I. INTRODUCTION
Information on relative sensitivity and specificity of functional and antigens assays for HIT and
antiphospholipid antibody syndrome are provided elsewhere (Visentin et al., 1994; Ginsberg et al., 1995;
Berube et al., 1998; Warkentin et al., 2000).
Abbreviations: EIA, enzyme immunoassay; h2-GPI, beta2-glycoprotein I; LMWH, low molecular weight
heparin; PF4-H, platelet factor 4-heparin.
a
Cardiolipin is found primarily in the inner leaflet of the mitochondrial membrane.
b
Phosphatidylserine is located in the inner leaflet of platelet membranes; thus, antiphospholipid
antibodies with antiphosphatidylserine activity could be relatively more important in the pathogenesis of
thrombocytopenia.
al., 1983). If the serotonin release method is used, the PRP is incubated at
37jC for 30 min with [14C]serotonin (0.1 ACi/mL of PRP added from a stock
solution of 50 ACi/mL of [14C]serotonin) (Lee et al., 1996).
3. Wash the platelets by pelleting them from PRP, then gently resus-
pend the platelets in calcium- and magnesium-free Tyrode’s buffer,
pH 6.3, containing glucose (5.6 mmol/L) and apyrase (2.5 U/mL).
Comment. Tyrode’s buffer consists of physiological concentrations of
sodium chloride (NaCl, 137 mmol/L), potassium chloride (2.7 mmol/L),
calcium chloride (CaCl2, 2 mmol/L), magnesium chloride (MgCl2, 1.0
mmol/L), and sodium dihydrogen phosphate (NaH2PO4, 3.3 mmol/L);
however, calcium-free and magnesium-free Tyrode’s is used in this wash
step to avoid activating the coagulation factors and platelets. The low pH
prevents platelets from aggregating during pelleting. Apyrase is an enzyme
that degrades adenine nucleotides (i.e., accumulation of the ADP from the
platelets is prevented). Azide-free bovine serum albumin (3.5 mg/mL) and
hirudin (1 U/mL) are included in the wash buffer in Greifswald, but not
Hamilton, although HEPES (5 mmol/L) is added to this buffer in Hamilton.
Following resuspension, the platelets are incubated for 15 min at 37jC
(Greifswald).
4. Pellet the washed platelets as before, and then gently resuspend the
platelets into calcium- and magnesium-containing Tyrode’s buffer,
pH 7.4, without apyrase or hirudin.
Comment. Following resuspension, the platelets should ‘‘rest’’ for 45
min at 37jC (Greifswald). The final resuspension buffer (Tyrode’s buffer at
physiological pH) contains calcium (2 mmol/L) and magnesium (1 mmol/L
Hamilton; 2 mmol/L Greifswald). The platelet count is adjusted to a
minimum of 300 wells 109/L; thus, after addition of washed platelets (75
AL) to the microtiter wells containing test serum (20 AL) and heparin-buffer (5
AL in Hamilton, 10 AL in Greifswald), the final platelet concentration will be
at least 215 109/L. Apyrase must not be included in this buffer, as the ADP
released during assessment of HIT-induced platelet activation is an important
potentiator of the platelet Fc receptor–mediated platelet activation (Polgár et
al., 1998).
0.3 U/mL; (4) UFH, 100 U/mL; (5) low molecular weight heparin (LMWH),
enoxaparin, 0.1 U/mL; and (6) UFH, 0.3 U/mL plus a monoclonal antibody
(IV.3) that inhibits platelet Fc receptor–mediated platelet activation. In
Greifswald, routine testing is performed using (1) buffer; (2) LMWH
(reviparin), 0.2 U/mL; (3) UFH, 100 U/mL; and (4) danaparoid, 0.2 U/mL
(to assess cross-reactivity); (5) sometimes LMWH 0.2 U/mL plus IV.3 is
performed to resolve unclear results. In Greifswald, the LMWH preparation
reviparin (Clivarine) is used because of its narrow molecular weight (MW)
range (80% of its chains have molecular mass of 2.4–7.2 kDa, i.e., 4–12
disaccharide units) (Jeske et al., 1997); this results in more consistent
formation of PF4–heparin complexes, enhancing sensitivity of the assay
(Greinacher et al., 1994b). Platelets are incubated with various test and
positive and negative control sera under these various reaction conditions
for up to 45 min (Greifswald) or 60 min (Hamilton). The order of pipetting is
important in optimizing assay results (Eichler et al., 1999) (see Table 3).
The order of pipetting is important. After adding serum to the microtiter plate wells, high heparin concentrations are added to the appropriate
wells: this will disrupt PF4–heparin complexes that may be present in the serum. After adding washed platelets, buffer, LMWH (low
concentrations), and danaparoid (for cross-reactivity testing) are added. If inhibition by monoclonal antibody IV.3 is tested, this reagent is added
before addition of the washed platelet suspension. In Hamilton, the pipetting order for the SRA is (1) addition of buffer-heparin, (2) serum, and (3)
platelets.
277
278 Warkentin and Greinacher
Table 4 Comparison Between Citrated Platelet-Rich Plasma and Washed Platelet Assays
Technical aspects Washed platelet assay Platelet-rich plasma assay Comments
Platelet preparation High g centrifugation during Low g centrifugation: less Availability of PF4 may be higher using
washing: increased baseline baseline platelet activation washed platelets (greater formation of
platelet activation PF4–heparin antigen complexes)
Apyrase Apyrase added to wash solution, No apyrase used Apyrase degrades ADP, and prevents its
but not to the final (no wash steps) accumulation; thus, platelet refractoriness
resuspension (reaction) buffer to ADP-mediated potentiation of HIT
serum-induced platelet activation is
avoided by apyrase
Reaction milieu Physiological calcium Low (micromolar) calcium IgG-mediated platelet activation optimal
concentration (2 mmol/L) owing to citrate with physiological calcium concentrations
IgG levels Reduced IgG levels during final Normal plasma IgG levels Reduced inhibition of Fc receptor–mediated
reaction platelet activation by IgG in washed
platelet assays
Plasma protein levels Reduced plasma protein levels Normal plasma protein levels Reduced nonidiosyncratic platelet activation
by heparin using washed platelets (?)
Temperature Room temperature 37jC Unknown significance
Reaction assessment Microtiter plates Conventional aggregometer Many assays performed simultaneously
using microtiter plates
See text for further details on differences between washed platelet and citrated platelet-rich plasma assays (pp. 279–282).
Warkentin and Greinacher
Laboratory Testing for HIT 281
and test reaction conditions that can be analyzed, and the long in-
cubation period employed (up to 60 min). The incubation period in
HIT assays should be at least 20–30 min (Stewart et al., 1995).
S1 85.4 ++++ ++++ ++++ ++++ ++++ ++++ +++ ++++ ++ +++
S2 84.4 ++++ ++++ ++++ ++++ ++++ +++ +++ ++++ +++ +++
Laboratory Testing for HIT
bosis begins several days after the patient’s last exposure to heparin (War-
kentin and Kelton, 2001) (see Chap. 3). A second explanation is that some
HIT antibodies recognize platelet-bound PF4 in the absence of an exogenous
source of heparin, perhaps by PF4 bound to platelet glycosaminoglycans.
Alternatively, as HIT antibodies are heterogeneous, there may be pathogenic
antibody subpopulations that bind relatively well to PF4 even in the absence
of heparin or heparin-like molecules (Newman and Chong, 1999; Amiral et
al., 2000). This phenomenon has implications for the interpretation of tests of
cross-reactivity of LMWH and danaparoid, as discussed later.
reader. The upper limit of the normal range is usually set at the mean plus 3
SD obtained using normal sera. Some laboratories set an indeterminate range
for samples that are only minimally above the upper normal range.
C. Fluid-Phase EIA
The fluid-phase EIA for HIT antibodies (Newman et al., 1998) is an adaption
of a staphylococcal protein A antibody-capture EIA method (Nagi et al.,
1993). By permitting antibody–antigen interactions to occur in a fluid phase,
problems of protein (antigen) denaturation inherent in solid-phase assays are
avoided.
Platelet factor 4 (5% biotinylated) is mixed with an optimal concentra-
tion of heparin, and this antigen mixture is incubated with diluted patient
serum or plasma (Fig. 3). Subsequently, the antigen–antibody mixture is
incubated with protein G-Sepharose in a microcentrifuge tube. Biotinylated
antigen–antibody complexes become bound to the protein G–Sepharose by
antibody Fc, and the complexes, are separated from unbound-antigen by cen-
trifugation and washing. The amount of biotin–PF4–heparin–antibody com-
plexes immobilized to the beads is measured using peroxidase substrate after
initial incubation with streptavidin-conjugated peroxidase.
290 Warkentin and Greinacher
itate to the bottom (negative) (Meyer et al., 1999; Eichler et al., 2001). The
assay is technically easy, can be performed rapidly, and is readily automated.
Results are read visually.
Eichler and colleagues (2001) compared this new assay with two func-
tional assays (HIPA test; SRA) and both commercially available solid-phase
PF4-dependent EIAs. In preselected samples, the H/PF4–PaGIA had a sen-
sitivity intermediate between that of the functional and commercial antigen
assays. The specificity appeared to resemble that of the functional assays.
In contrast, Risch and coworkers (2003) found many more sera to test
positive using the H/PF4–PaGIA, compared to a commercial EIA (Asse-
rochromR), among 42 patients sampled 10–18 days following cardiac sur-
gery (69% vs. 26%). Since none of the patients had clinical evidence for
HIT, this suggested the diagnostic specificity of the H/PF4–PaGIA to be far
less than the solid-phase EIA. These authors did not test sera from patients
with HIT, and therefore were unable to assess test sensitivity (Warkentin,
2003b).
The manufacturer’s instructions indicate that the assay is to be read as
‘‘positive’’ (any agglutination within the gel), ‘‘negative’’ (no agglutination)
using neat (undiluted) serum, or ‘‘borderline.’’ However, when a positive or
borderline test result was obtained, Alberio et al. (2003) repeated the assay
with undiluted and serially diluted plasma (up to 1 in 1024) until the result was
negative. The reported titer was the last positive result followed by either
borderline or negative results. Patients judged clinically to have had ‘‘prob-
able’’ or ‘‘highly probable/definite’’ HIT had antibody titers of 4 or more in
39 of 54 (72%) cases, compared with only 2 of 85 (2%) judged ‘‘unlikely’’ to
have had HIT. Further, all 19 of the patient samples that tested positive in a
c-PRP aggregation assay tested positive in the PaGIA (generally, in a titer
of 8 or higher). Among all patients studied, the percentage with associated
thrombotic complications increased from 8% (negative or low titer) to 55%
(positive titer 4–16) to 74% (positive titer 32–256). This study suggests that
reporting quantitatively the results of the HPF4–PaGIA—with a titer of 4 or
more being clinically significant—may increase diagnostic usefulness.
EIA will also test positive using a washed platelet activation assay (Grei-
nacher et al., 1994a). The sensitivity of both EIA and SRA was even higher
(>90%) for detecting antibodies that caused HIT in prospectively studied
postoperative patients (Warkentin et al., 2000).
Although both antigen and activation assays have similarly high sen-
sitivity for clinical HIT, there is evidence that antigen assays have greater
sensitivity for detecting HIT antibodies not associated with thrombocytope-
nia or other clinical events (Amiral et al., 1995; Arepally et al., 1995; Bauer et
al., 1997; Warkentin et al., 2000) (Fig. 4). Stated another way, the SRA is
more specific for clinical HIT than the antigen assay. The biological expla-
nation for greater specificity of a sensitive activation assay for clinical HIT,
compared with an antigen assay, could relate to the functional heterogeneity
of HIT antibodies against antigenic determinants on PF4, only some of which
activate platelets strongly (Amiral et al., 2000). Data reported by Visentin and
colleagues (1994) also support a higher sensitivity of antigen assays for de-
tecting HIT antibodies. These workers studied 12 HIT plasmas that tested
positive in both SRA and PF4–heparin–EIA. However, at a 1:100 sample
dilution, only 2 of the 12 samples still tested positive in the activation assay. In
contrast, even at a 1:200 dilution, all 12 plasmas still tested positive in the EIA.
Bachelot and colleagues (1998) observed that HIT plasmas that tested only
weakly positive in the PF4–heparin–EIA tended to give negative washed
platelet SRA results when using platelets with the least reactive FcgIIA
receptor genotype, Arg131 (see Chap. 9).
The difference in sensitivity for HIT antibodies between the PF4–hep-
arin–EIA and aggregation studies using c-PRP is considerable. Only about
33–64% of samples that test positive in the PF4–heparin–EIA also test pos-
itive using c-PRP aggregation (Greinacher et al., 1994a; Nguyên et al., 1995;
Rugeri, et al., 1999). Although one laboratory reported a greater sensitivity
using c-PRP aggregation than the EIA (Look et al., 1997), these workers did
not employ a two-point method, and so may have observed false-positive
results using the aggregation assay.
Table 7 summarizes possible explanations for discrepancies in results of
activation and antigen assays for HIT.
Figure 5 Continued.
with a low pretest probability, should lead to repeating the test or perfor-
mance of the complementary activation or antigen assay. Additionally,
further clinical information should be sought (e.g., Has another explanation
for the thrombocytopenia become apparent? Could the patient have had an
unrecognized recent heparin exposure?).
In contrast, for patients with the typical temporal onset of thrombocy-
topenia (i.e., a platelet count fall that begins 5–10 days after beginning heparin
treatment), we believe that, in general, there are two different pretest
probabilities for HIT: moderate and high. Because HIT is a relatively
common explanation for thrombocytopenia that begins during this charac-
Laboratory Testing for HIT 299
Figure 6 Diagnosis of HIT in doubt: Although this 71-year-old patient with mod-
erate pretest probability of HIT had positive laboratory testing for HIT antibodies,
the clinical course casts doubt on the actual role HIT played in the thrombocyto-
penia and thrombotic events. Thrombocytopenia began on day 5 of heparin treat-
ment (nadir, 31 109/L; day 8), together with clinical and laboratory evidence for
septicemia. Laboratory testing for HIT antibodies was strongly positive by activa-
tion assay (SRA, 90% release at 0.1 U/mL heparin, <5% release at 100 U/mL
heparin) and weakly positive by PF4–heparin–EIA: O.D. = 0.709). Clinical evidence
for HIT includes the symptomatic DVT and (possible) pulmonary embolism; how-
ever, the dramatic increase in platelet count during therapeutic-dose heparin treat-
ment and the bacteremia are strong evidence for septicemia, rather than HIT, as an
explanation for the thrombocytopenia.
Thus, HIT antibody testing is among the most useful of platelet im-
munology assays. For comparison, Fig. 7 also shows the profile of a ‘‘nonin-
formative assay’’ (see line A). This is the profile for various tests of ‘‘platelet-
associated IgG’’ for the diagnosis of autoimmune thrombocytopenia. Certain
glycoprotein-specific platelet antibody tests have operating characteristics
intermediate between those for HIT and a noninformative assay. For
example, the MAIPA (monoclonal antibody immobilization of platelet
antigens) assay has only moderate sensitivity but high specificity for diagnosis
of autoimmune thrombocytopenia (see Chap. 2).
V. IN VITRO CROSS-REACTIVITY
A. Cross-Reactivity Using Activation Assays
Cross-reactivity studies have been performed most frequently using activa-
tion assays. However, there are no standard methods for, or even a standard
definition of, in vitro cross-reactivity. In one study of LMWH and danapa-
roid cross-reactivity, an increase in platelet activation in the presence of the
drug over baseline was used to determine cross-reactivity (Warkentin, 1996).
This definition was used to avoid falsely attributing cross-reactivity to drug-
independent platelet activation that is produced by some patients’ sera. The
reason for this definition was the common phenomenon that platelet activa-
tion can be caused by a patient’s serum even in the absence of added heparin.
In the HIPA test, comparison of the lag time to aggregation can be used to
judge cross-reactivity: if a sample shows platelet aggregation with heparinoid
or LMWH earlier than in the presence of buffer, then cross-reactivity is
present. In general, in vitro cross-reactivity with danaparoid is usually
clinically insignificant (Warkentin, 1996; Newman et al., 1998) (see Chap. 14).
assay (SPRCA), has been developed (Sinor et al., 1990; Sinor and Stone, 1994;
Leach et al., 1994, 1995, 1997). Platelets are coated onto U-shaped microtiter
platelet wells, and either heparin-serum (immune complex method), or
heparin-albumin with subsequent addition of serum after a wash step (hapten
method), is added. After washing, red cells coated with anti-IgG are added to
the wells. Following centrifugation, the appearance of the indicator red cells
on the well bottoms is scored on a 10-point scale, ranging from negative (tight
red cell button) to strongly positive (diffuse red cell binding on the well
bottom). According to Leach and colleagues (1997), both immune complex
and hapten reaction profiles are commonly seen with putative HIT sera
(immune complex > both > hapten). However, only limited comparisons
with conventional HIT assays have been performed, and the diagnostic use-
fulness of these assays is unknown. Disadvantages include the subjective
scoring system, as well as the need to ensure that HLA and platelet-specific
alloantibodies do not interfere with testing.
There have been attempts to identify specific binding of heparin-depen-
dent antibodies to platelet glycoproteins using immunoblotting and immuno-
precipitation (Lynch and Howe, 1985; Howe and Lynch, 1985; Greinacher
et al., 1994d). However, no study has identified a consistent reaction profile
diagnostic of HIT. There is thus no experimental evidence implicating the
involvement of platelet glycoproteins as an immune target in the pathogenesis
of HIT.
ACKNOWLEDGMENTS
Studies described in this chapter were supported by the Heart and Stroke
Foundation of Ontario (operating grants A2449, T2967, B3763, and T4502),
and by the Deutsche Forschungsgemeinschaft Gir 1096/2-1 and Gir 1096/2-
2. Dr. Warkentin was a Research Scholar of the Heart and Stroke Foun-
dation of Canada.
REFERENCES
Ardlie NG, Packham MA, Mustard JF. Adenosine diphosphate-induced platelet ag-
gregation in suspensions of washed rabbit platelets. Br J Haematol 1970; 19:7–
17.
Amiral J, Bridey F, Dreyfus M, Vissac AM, Fressinaud E, Wolf M, Meyer D. Platelet
factor 4 complexed to heparin is the target for antibodies generated in heparin-
induced thrombocytopenia. Thromb Haemost 1992; 68:95–96.
Amiral J, Bridey F, Wolf M, Boyer-Neumann C, Fressinaud E, Vissac AM, Peynaud-
Debayle E, Dreyfus M, Meyer D. Antibodies to macromolecular platelet factor
4–heparin complexes in heparin-induced thrombocytopenia: a study of 44 cases.
Thromb Haemost 1995; 73:21–28.
Amiral J, Pouplard C, Vissac AM, Walenga JM, Jeske W, Gruel Y. Affinity purifica-
tion of heparin-dependent antibodies to platelet factor 4 developed in heparin-
induced thrombocytopenia: biological characteristics and effects on platelet
activation. Br J Haematol 2000; 109:336–341.
Arepally G, Reynolds C, Tomaski A, Amiral J, Jawad A, Poncz M, Cines DB. Com-
parison of the PF4/heparin ELISA assay with the 14C-serotonin assay in the
diagnosis of heparin-induced thrombocytopenia. Am J Clin Pathol 1995; 104:
648–654.
Babcock RB, Dumper CW, Scharfman WB. Heparin-induced immune thrombocy-
topenia. N Engl J Med 1976; 295:237–241.
Bachelot-Loza C, Saffroy R, Lasne D, Chatellier G, Aiach M, Rendu F. Importance of
the FcgRIIa–Arg/His-131 polymorphism in heparin-induced thrombocytope-
nia diagnosis. Thromb Haemost 1998; 79:523–528.
Bauer TL, Arepally G, Konkle BA, Mestichelli B, Shapiro SS, Cines DB, Poncz M,
McNulty S, Amiral J, Hauck WW, Edie RN, Mannion JD. Prevalence of
heparin-associated antibodies without thrombosis in patients undergoing car-
diopulmonary bypass surgery. Circulation 1997; 95:1242–1246.
Berube C, Mitchell L, Silverman E, David M, Saint Cyr C, Laxer R, Adams M, Vegh
P, Andrew M. The relationship of antiphospholipid antibodies to thromboem-
bolic events in pediatric patients with systemic lupus erythematosus: a cross-
sectional study. Pediatr Res 1998; 44:351–366.
Chong BH, Ismail F, Cade J, Gallus AS, Gordon S, Chesterman CN. Heparin-induced
thrombocytopenia: studies with a new molecular weight heparinoid, Org 10172.
Blood 1989; 73:1592–1596.
Chong BH, Burgess J, Ismail F. The clinical usefulness of the platelet aggregation test
for the diagnosis of heparin-induced thrombocytopenia. Thromb Haemost
1993a; 69:344–350.
Chong BH, Pilgrim RL, Cooley MA, Chesterman CN. Increased expression of platelet
IgG Fc receptors in immune heparin-induced thrombocytopenia. Blood 1993b;
81:988–993.
Chong BH, Murray B, Berndt MC, Dunlop LC, Brighton T, Chesterman CN. Plasma
P-selectin is increased in thrombotic consumptive platelet disorders. Blood
1994; 83:1535–1541.
Collins JL, Aster RH, Moghaddam M, Piotrowski MA, Strauss TR, McFarland JG.
Diagnostic testing for heparin-induced thrombocytopenia (HIT): and enhanced
306 Warkentin and Greinacher
Leach MF, Cooper LK, AuBuchon JP. Detection of drug-dependent platelet anti-
bodies by use of solid-phase red cell adherence techniques. Immunohematology
1995; 11:143–149.
Leach MF, Cooper LK, AuBuchon JP. Detection of drug-dependent, platelet-reactive
antibodies by solid-phase red cell adherence assays. Br J Haematol 1997; 97:
755–761.
Lindhoff-Last E, Gerdsen F, Ackermann H, Bauersachs R. Determination of heparin–
platelet factor 4–IgG antibodies improves diagnosis of heparin–induced throm-
bocytopenia. Br J Haematol 2001; 113:886–890.
Look KA, Sahud M, Flaherty S, Zehnder JL. Heparin-induced platelet aggregation
vs. platelet factor 4 enzyme-linked immunosorbent assay in the diagnosis of
heparin-induced thrombocytopenia–thrombosis. Am J Clin Pathol 1997; 108:
78–82.
Lynch DM, Howe SE. Heparin-associated thrombocytopenia: antibody binding spe-
cificity to platelet antigens. Blood 1985; 66:1176–1181.
Makhoul RG, Greenberg CS, McCann RL. Heparin-induced thrombocytopenia and
thrombosis: a serious clinical problem and potential solution. J Vasc Surg 1986;
4:522–528.
Meyer O, Salama A, Pittet N, Schwind P. Rapid detection of heparin-induced platelet
antibodies with particle gel immunoassay (ID-HPF4). Lancet 1999; 354:1525–
1526.
Mustard JF, Perry DW, Ardlie NG, Packham MA. Preparation of suspensions of
washed platelets from humans. Br J Haematol 1972; 22:193–204.
Nagi PK, Ackermann F, Wendt H, Savoca R, Bosshard HR. Protein A antibody-
capture ELISA (PACE): an ELISA format to avoid denaturation of surface-
adsorbed antigens. J Immunol Methods 1993; 158:267–276.
Newman PM, Chong BH. Further characterization of antibody and antigen in
heparin-induced thrombocytopenia. Br J Haematol 1999; 107:303–309.
Newman PM, Swanson RL, Chong BH. Heparin-induced thrombocytopenia: IgG
binding to PF4–heparin complexes in the fluid phase and cross-reactivity with
low molecular weight heparin and heparinoid. Thromb Haemost 1998; 80:292–
297.
Nguyên P, Lecompte T, and Groupe d’Etude sur l’Hémostase et la Thromboses
(GEHT) de la Société Francßaise d’Hématologie. Nouv Rev Fr Hematol 1994;
36:353–357.
Nguyên P, Droullé C, Potron G. Comparison between platelet factor 4/heparin
complexes ELISA and platelet aggregation test in heparin-induced thrombo-
cytopenia [letter]. Thromb Haemost 1995; 74:793–810.
Packham MA, Guccione MA, Perry DW. ADP does not release platelet granule
contents in a plasma-free system [abstr]. Fed Proc 1971; 30:201.
Pengo V, Biasiolo A, Fior MG. Autoimmune antiphospholipid antibodies are directed
against a cryptic epitope expressed when h2-glycoprotein I is bound to a suitable
surface. Thromb Haemost 1995; 73:29–34.
Pfueller SL, David R. Different platelet specificities of heparin-dependent platelet
aggregating factors in heparin-induced thrombocytopenia. Br J Haematol 1986;
64:149–159.
Laboratory Testing for HIT 309
I. INTRODUCTION
A. The Concept of Pseudo-HIT
Heparin-induced thrombocytopenia (HIT) is strongly associated with life-
and limb-threatening venous and arterial thrombosis, including pulmonary
embolism, venous limb gangrene, and large vessel arterial occlusion. How-
ever, HIT is by no means a unique explanation for the combination of throm-
bocytopenia and thrombosis (Table 1).
In these pseudo-HIT disorders, thrombocytopenia usually occurs early
during the course of heparin treatment. This could reflect the prothrombotic
process associated with the patient’s primary diagnosis. Alternatively, hepa-
rin could exacerbate the platelet count fall by nonimmune proaggregatory
effects on platelets (see Chap. 5). If the patient previously received heparin,
physicians might consider HIT in the differential diagnosis of the platelet
count fall.
However, one pseudo-HIT syndrome in particular closely resembles
even the typical day 5–10 timing of thrombocytopenia characteristic of HIT:
adenocarcinoma-associated disseminated intravascular coagulation (DIC).
In these patients, the fall in platelet count begins soon after stopping heparin
treatment. Because the patients usually will have received heparin for 5–10
days to treat adenocarcinoma-associated thrombosis, the timing of the onset
of thrombocytopenia closely resembles immune HIT. Furthermore, the fre-
313
314 Warkentin
Pathogenesis of thrombocytopenia
Pseudo-HIT disorder and thrombosis Timing
lels with a similar syndrome in HIT patients, suggests that a common factor
(coumarin anticoagulation) may play a crucial pathogenic role in both
disorders (Warkentin, 2001). Likewise, similarities between HIT and the
lupus anticoagulant syndrome suggest that they could also share common
pathogenic mechanisms (Arnout, 1996, 2000; Gruel, 2000).
venous limb gangrene occurs in a limb with preceding active DVT, this
suggests that local factors, such as direct extension of thrombosis, as well as
exacerbation of distal thrombosis by venous stasis, contribute to large- and
small-vessel thrombosis characteristic of this syndrome.
Venous thrombosis complicating adenocarcinoma, especially when
complicated by DIC or severe venous ischemia or necrosis, should be treated
with heparin, rather than warfarin or other coumarin anticoagulants. Rever-
sal of warfarin anticoagulation (with vitamin K and plasma infusion, but not
with prothrombin complex concentrates, as these do not contain sufficient
protein C) and prompt control of DIC with heparin could salvage a limb with
severe phlegmasia, or limit damage in a patient with venous gangrene. An
effective agent often is LMWH (Prandoni, 1997; Lee et al., 2003). I recom-
318 Warkentin
B. Pulmonary Embolism
Mild thrombocytopenia is common in patients with pulmonary embolism.
Sometimes the thrombocytopenia is severe and associated with laboratory
markers of DIC (Stahl et al., 1984; Mustafa et al., 1989) (Fig. 3). The throm-
bocytopenia presumably results from thrombin-induced platelet activation.
Large thromboemboli within the high-flow pulmonary vessels may act as a
reservoir for clot-bound thrombin that is relatively protected from inhibition
by antithrombin-dependent inhibitors (Weitz et al., 1990). This view is
indirectly supported by the observation that thrombocytopenia commonly
occurs in patients with pulmonary embolism, but not in patients with DVT
alone (Monreal et al., 1991; Warkentin et al., 2003a). Further, increased
heparin clearance has been demonstrated in experimental pulmonary embo-
lism (Chiu et al., 1977).
Because HIT is also strongly associated with pulmonary embolism
(Warkentin et al., 1995, 2003a), a diagnostic and therapeutic dilemma results
when a patient presents with pulmonary embolism and thrombocytopenia 5
or more days after surgery managed with postoperative heparin prophylaxis
(Fig. 4). Initiating therapeutic heparin could have catastrophic consequences
for the patient who has circulating HIT antibodies, although in sufficient
doses it is effective for a patient with pulmonary embolism and DIC without
HIT. Because these two possibilities cannot be readily distinguished on
clinical grounds alone, one should manage such a patient with an alternative
anticoagulant until the results of HIT antibody testing become available
(Warkentin, 2000).
C. Diabetic Ketoacidosis
Diabetic ketoacidosis (DKA) can be associated with acute thromboembolic
complications. Evidence for in vivo platelet activation was observed in one
Pseudo–HIT 319
study of 10 patients who had elevated plasma levels of platelet factor 4 and h-
thromboglobulin during DKA that resolved following recovery (Campbell et
al., 1985) Evidence for activation of coagulation includes elevated fibrin
degradation products and reduced antithrombin (Paton, 1981). Figure 5
illustrates a patient with ‘‘white clots’’ in the femoral artery, leading to
amputation, who was initially thought to have HIT. However, HIT antibody
testing and subsequent clinical events proved that the patient did not have
HIT as the initial explanation for this dramatic clinical presentation of
thrombocytopenia and thrombosis complicating DKA (although HIT oc-
curred later in the clinical course). I am also aware of a patient with essential
thrombocythemia who developed postoperative DKA, thrombocytopenia,
and bilateral lower limb artery thrombosis that occurred too early during
UFH prophylaxis (days 2–3) to have been caused by immune HIT. A similar
example of early-onset severe thrombocytopenia and arterial thrombosis
resulting in amputation of an arm was reported in a patient with diabetic
ketoacidosis and adult respiratory distress syndrome (ARDS) (Phillips et al.,
1994). Although the authors suggested HIT secondary to heparin ‘‘flushes’’ as
the diagnosis, pseudo-HIT seems more likely based upon the temporal
features of the case, as well as the negative laboratory testing for HIT
antibodies.
Warkentin et al., 2000; Galli et al., 2003). The parallels between these
disorders led Arnout (1996) to hypothesize that IgG-mediated platelet
activation could explain thrombosis in APLAS. Supportive experimental
data include the observations that antiphospholipid antibodies enhance
platelet activation induced by other agonists (Martinuzzo et al., 1993).
Furthermore, Arvieux et al. (1993) observed that murine monoclonal anti-
bodies reactive against h2GP I induced platelet activation in the presence of
subthreshold concentrations of ADP and epinephrine, an effect dependent on
binding to platelet FcgIIa receptors. However, other workers were unable to
demonstrate enhanced platelet activation in the presence of IgG antiphos-
pholipid antibodies (Shi et al., 1993; Ford et al., 1998) or showed no role for
platelet FcgIIa receptors (Lutters et al., 2001; Jankowski et al., 2003).
E. Thrombolytic Therapy
Acute thrombocytopenia is common in patients treated with streptokinase,
especially when combined with heparin (Balduini et al., 1993) (Fig. 6). This
could represent a direct, activating stimulus of heparin on platelets that
perhaps is exacerbated by procoagulant effects of thrombolytic therapy. For
example, fibrin degradation products generated by thrombolytic agents bind
and protect thrombin from inhibition by heparin (Weitz et al., 1998). Such a
mechanism could explain thrombocytopenia after use of any thrombolytic
drug.
Pseudo–HIT 325
G. Infective Endocarditis
Infective endocarditis is frequently complicated by thrombocytopenia. These
patients are also at risk for septic emboli manifesting as thrombotic or
hemorrhagic stroke, myocardial infarction, renal infarction, or even acute
limb ischemia (de Gennes et al., 1990). Thus, the profile of macrovascular
thrombosis and thrombocytopenia characteristic of HIT can be mimicked,
especially as heparin is often used to anticoagulate patients with septic
endocarditis (Delahaye et al, 1990). Microembolization leading to multiple
small infarcts or microabscesses, in such organs as muscles, adrenal glands,
and spleen, is an additional feature of endocarditis (Ting et al., 1990) that is
not seen in HIT.
I. Posttransfusion Purpura
Posttransfusion purpura (PTP) is a rare syndrome characterized by severe
thrombocytopenia and mucocutaneous bleeding that begins 5–10 days after
blood transfusion, usually red cell concentrates. More than 95% of affected
patients are older women, in keeping with its pathogenesis of an anamnestic
recurrence of platelet-specific alloantibodies in women previously sensitized
328 Warkentin
importance of a high clinical suspicion for HIT even in complex situations for
which other explanations for thrombocytopenia are present. The wider
availability of assays for HIT antibodies should help clinicians better diag-
nose and manage patients who develop thrombocytopenia and thrombosis
during or shortly following heparin treatment.
REFERENCES
Adamson DJA, Currie JM. Occult malignancy is associated with venous thrombosis
unresponsive to adequate anticoagulation. Br J Clin Pract 1993; 47:190–191.
Aird WC. The role of the endothelium in severe sepsis and the multiple organ
dysfunction syndrome. Blood 2003a; 101:3765–3777.
Aird WC. The hematologic system as a marker of organ dysfunction in sepsis. Mayo
Clin Proc 2003b; 78:869–881.
Arnout J. The pathogenesis of the antiphospholipid antibody syndrome: a hypothesis
based on parallelisms with heparin-induced thrombocytopenia. Thromb Hae-
most 1996; 75:536–541.
Arnout J. The role of h2-glycoprotein I-dependent lupus anticoagulants in the patho-
genesis of the antiphospholipid syndrome. Verh K Acad Geneeskd Belg 2000;
62:353–372.
Arnout J, Vermylen J. Current status and implications of autoimmune antiphospho-
lipid antibodies in relation to thrombotic disease. J Thromb Haemost 2003;
1:931–942.
Arvieux J, Roussel B, Pouzol P, Colomb MG. Platelet activating properties of murine
monoclonal antibodies to beta2-glycoprotein I. Thromb Haemost 1993; 70:
336–341.
Asherson RA, Khamashta MA, Ordi-Ros J, Derksen RH, Machin SJ, Barquinero J,
Outt HH, Harris EN, Vilardell-Torres M, Hughes GR. The ‘‘primary’’ anti-
phospholipid syndrome: major clinical and serological features. Medicine 1989;
68:366–374.
Auger WR, Permpikul P, Moser KM. Lupus anticoagulant, heparin use, and throm-
bocytopenia in patients with chronic thromboembolic pulmonary hypertension:
a preliminary report. Am J Med 1995; 99:392–396.
Balduini CL, Noris P, Bertolino G, Previtali M. Heparin modifies platelet count and
function in patients who have undergone thrombolytic therapy for acute
myocardial infarction [letter]. Thromb Haemost 1993; 69:522–532.
Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A,
Steingrub JS, Garber GE, Helterbrand JD, Ely EW, Fisher JC Jr. Efficacy and
safety of recombinant human activated protein C for severe sepsis. N Engl J
Med 2001; 344:699–709.
Bone RC, Francis PB, Pierce AK. Intravascular coagulation associated with the adult
respiratory distress syndrome. Am J Med 1976; 61:585–589.
Pseudo–HIT 331
Mustafa MH, Mispireta LA, Pierce LE. Occult pulmonary embolism presenting with
thrombocytopenia and elevated fibrin split products. Am J Med 1989; 86:490–
491.
Owings JT, Bagley M, Gosselin R, Romac D, Disbrow E. Effect of critical injury on
plasma antithrombin activity: low antithrombin levels are associated with
thromboembolic complications. J Trauma 1996; 41:396–405.
Paton RC. Haemostatic changes in diabetic coma. Diabetologia 1981; 21:172–177.
Petri M. Pathogenesis and treatment of the antiphospholipid antibody syndrome. Adv
Rheumatol 1997; 81:151–177.
Phillips DE, Payne DK, Mills GM. Heparin-induced thrombotic thrombocytopenia.
Ann Pharmacother 1994; 28:43–46.
Prandoni P. Antithrombotic strategies in patients with cancer. Thromb Haemost 1997;
78:141–144.
Regnault V, Helft G, Wahl D, Czitrom D, Vuillemenot A, Papouin G, Roda L,
Danchin N, Lecompte T. Antistreptokinase platelet-activating antibodies are
common and heterogeneous. J Thromb Haemost 2003; 1:1055–1061.
Rinaldo JE, Perez H. Ischemic necrosis of both lower extremities as a result of the
microembolism syndrome complicating the adult respiratory distress syndrome
caused by Escherichia coli pneumonia and septicemia. Am Rev Respir Dis 1982;
126:932–935.
Rosse WF. Paroxysmal nocturnal hemoglobinuria as a molecular disease. Medicine
1997; 76:63–93.
Shi W, Chong BH, Chersterman CN. h2-Glycoprotein I is a requirement for anti-
cardiolipin antibodies binding to activated platelets: differences with lupus anti-
coagulants. Blood 1993; 81:1255–1262.
Smith OP, White B. Infectious purpura fulminans: diagnosis and treatment. Br J
Haematol 1999; 104:202–207.
Stahl RL, Javid JP, Lackner H. Unrecognized pulmonary embolism presenting as
disseminated intravascular coagulation. Am J Med 1984; 76:772–778.
Ting W, Silverman NA, Arzouman DA, Levitsky S. Splenic septic emboli in endo-
carditis. Circulation 1990; 82(suppl 5):IV105–IV109.
Triplett DA. Lupus anticoagulants/antiphospholipid-protein antibodies: the great
imposters. Lupus 1996; 5:431–435.
Vaughan DE, Kirshenbaum JM, Loscalzo J. Streptokinase-induced, antibody-me-
diated platelet aggregation: a potential cause of clot propagation in vivo. J Am
Coll Cardiol 1988; 11:1343–1348.
Warkentin TE. Heparin-induced thrombocytopenia: IgG-mediated platelet activation
platelet microparticle generation, and altered procoagulant/anticoagulant bal-
ance in the pathogenesis of thrombosis and venous limb gangrene complicating
heparin-induced thrombocytopenia. Transfusion Med Rev 1996; 10:249–258.
Warkentin TE. Venous thromboembolism in heparin-induced thrombocytopenia.
Curr Opin Pulm Med 2000; 6:343–351.
Warkentin TE. Venous limb gangrene during warfarin treatment of cancer-associated
deep venous thrombosis. Ann Intern Med 2001; 135:589–593.
Warkentin TE, Kelton JG. Interaction of heparin with platelets, including heparin-
induced thrombocytopenia. In: Bounameaux H, ed. Low-Molecular-Weight
334 Warkentin
I. INTRODUCTION
Paradoxical effect
Treatment for HIT of treatment Comments
A. Disclaimer
There are several challenging aspects to treating patients with HIT. In par-
ticular, these patients are not clinically homogeneous: they represent a com-
plex mix of varying initial indication for heparin, location and severity of
HIT-associated thrombosis, and, not infrequently, dysfunction of one or more
vital organs. This presents difficulties both for performing clinical studies
as well as in the application of treatment recommendations for individual
338 Greinacher and Warkentin
ranged from 1 month to 1 year, despite the absence of apparent HIT in any
patient. The implications of ‘‘subclinical’’ HIT antibody seroconversion on
influencing subsequent cardiovascular morbidity and mortality awaits pro-
spective study.
progressive, or recurrent thrombosis resulting from HIT itself; or (3) for both
reasons.
For a HIT patient with thrombosis in whom heparin administration has
been discontinued, there is, nevertheless, a very high risk for subsequent
thrombosis. This was shown in two historically controlled prospective treat-
ment cohort studies (Greinacher et al., 1999a,b), in which the incidence of
thrombotic events ranged from 5 to 10% per patient day (see Chap. 15). This
high event rate (6.1% per day in the meta-analysis) occurred after stopping
heparin therapy and after laboratory confirmation of HIT, but before insti-
tution of alternative anticoagulation with lepirudin (mean period of treat-
ment delay; 1.7 days) (Greinacher et al., 2000). This experience suggests that
alternative anticoagulant therapy should not be delayed for results of HIT
antibody testing in patients strongly suspected of having HIT.
Figure 2 Thrombin generation and fibrin formation in acute HIT. (A) Thrombin
generation, as assessed by thrombin–antithrombin (TAT) complexes, is markedly in-
creased in acute HIT (mean, 55 ng/mL; normal,<4.1 ng/mL). Whereas danaparoid
reduces thrombin generation in these patients, the defibrinogenating snake venom,
ancrod, does not. (B) Levels of cross-linked fibrin degradation products (D-dimer) are
increased in patients with acute HIT (mean, f4–5 Ag/mL; normal, <0.5 Ag/mL).
Whereas danaparoid reduces D-dimer levels, ancrod increases their levels. Baseline (B)
samples were obtained at diagnosis of HIT and before treatment with danaparoid or
ancrod; subsequent values are shown for day 1 (D1, 1–24 h postinitiation of treat-
ment), day 2 (25–48 h), and day 3 (49–72 h). *p < 0.001, **p < 0.002. (From War-
kentin, 1998.)
Mechanism of action,
pharmacokinetics Monitoring Undesirable effects Comments
Treatment of HIT
Catalyzes the inactivation Anti-Xa levels during Cross-reactivity (XR) with Anticoagulant effect
of factor Xa by AT, treatment by an HIT antibodies: in vitro depends on adequate
and of thrombin (IIa) amidolytic assay XR usually not associated AT levels
by AT and HCII using danaparoid with adverse effects; patients Does not significantly
Bioavailability after sc reference curve should be monitored for prolong the aPTT, ACT,
injection f100%; peak Monitoring recommended in in vivo XR (unexplained PT/INR (does not
anti-Xa levels, 4–5 h patients with: (1) significant platelet count fall, progressive interfere with monitoring
after injection renal impairment; (2) body new TECs); in vivo XR is of overlapping oral
(Danhof et al., 1992) weight <45 kg or >110 kg; estimated to occur in f3% anticoagulants)
Mean plasma distribution (3) life- or limb-threatening of patients (Magnani, 1993) Reduce dosage if serum
time following iv bolus, thrombosis; (4) unexpected Bleeding complications in creatinine >265 Amol/L
f2.3 h bleeding; (5) critically ill or compassionate-release No antidote: In case of
Plasma t1/2 of anti-Xa unstable patient study (Ortel and Chong, overdosage, stop the
activity, 17–28 h 1998): fatal (0.9%), major drug and treat bleeding
(mean, 25 h); t1/2 of nonfatal bleeding (6.5%); with blood products as
anti-IIa activity, 2–4 h no major bleeds in RCT indicated
(Danhof et al., 1992) (Chong, 1996)
Skin hypersensitivity: rare
Abbreviations: ACT, activated clotting time; aPTT, activated partial thromboplastin time; AT, antithrombin; HCII, heparin cofactor II; iv,
intravenous; PT/INR, prothrombin time/international normalized ratio; RCT, randomized controlled trial; sc, subcutaneous; t1/2, drug half-life.
345
346
Direct, noncovalent, aPTT during treatment; a Development of antihirudin f40% of patients develop antihirudin
irreversible inhibitor more precise monitoring antibodies in f40% of antibodies on day 5 or later of
of free and clot-bound is possible by the ECT patients. In about 3% of treatment; in only f5% of these
thrombin (see Chaps. 15, 17, 19) patients, these antibodies patients is a dose reduction or
Bioavailability after Daily aPTT monitoring enhance the anticoagulant increase needed; risk of anaphylactic
sc injection, f100%; is recommended in all effect of hirudin, and reactions post–iv bolus
peak effect, 2–3 h patients (see Comments require a substantial dose No major effect on PT/INR
Mean plasma distribution re: antihirudin antibodies) reduction. (Greinacher et al., 2000)
time after iv bolus, f2 h Monitoring by ECT Anaphylactic reactions: Reduce dosage if serum creatinine
Mean plasma t1/2, 1.3 h; recommended: f0.015% (first exposure) >120 Amol/L (see Table 3 in
t1/2 greatly prolonged 1. During cardiopulmo- f0.15% (reexposure) Chap. 15)
in renal failure (f200 h nary bypass surgery associated with iv bolus No antidote: In case of overdosage,
in nephrectomized 2. Unexpected bleeding injection (Greinacher stop the drug and treat bleeding
patients) Monitoring by quantitative et al., 2003) with blood products as indicated
hirudin EIA recommended: Allergic reactions: very rare (hemofiltration with a high-flux
1. Decreased prothrombin Skin hypersentivity: very rare membrane is a possible treatment
levels (Lindhoff-Last Bleeding complications in for life-threatening bleeding)
et al., 2000) HIT patients in prospective
studies: major bleeding in
two prospective studies, 13.4,
17% (see Chap. 15)
Abbreviations: aPTT, activated partial thromboplastin time; ECT, ecarin-clotting time; EIA, enzyme immunoassay; sc, subcutaneous; iv, intravenous;
t1/2 drug half-life.
Greinacher and Warkentin
Table 5 Main Characteristics of the Direct Thrombin Inhibitor, Argatroban
Treatment of HIT
Mechanism of action,
pharmacokinetics Monitoring Undesirable effects Comments
Direct, noncovalent, reversible aPTT during treatment; no data No reported side effects besides Only iv use of argatroban has been
inhibitor of free and clot- exist as to whether more pre- bleeding complications (num- tested in HIT
bound thrombin cise monitoring at higher doses ber of patients treated is Reduce dosage by 75% in case of
f50% of the drug is would be achieved using too low to rule out possibi- liver impairment
plasma protein bound; other methods, such as ECT lity of rare side effects) No dose reduction in renal failure
Steady state is reached Target INR is >4.0 when No antidote: in case of overdosage
1–3 h after starting warfarin is overlapped with or severe bleeding, stop the drug
iv infusion argatroban (however, following and treat bleeding with blood
Mean plasma t1/2 is discontinuation of argatroban, products as indicated
40–50 min; t1/2 is the usual target INR of Argatroban prolongs the INR and
prolonged 4- to 2.0–3.0 applies during further requires a strategy adopted to
5-fold in moderate warfarin treatment) the INR reagent used for
liver impairment overlapping treatment with
warfarin (see Chap. 16)
Abbreviations: aPTT, activated partial thromboplastin time; ECT, ecarin-clotting time; INR, international normalized ratio; iv, intravenous; t1/2, drug
half-life.
347
348 Greinacher and Warkentin
1995; Amiral et al., 1997; Turpie et al., 2001), but there is no reported
experience with this agent for HIT.
kentin and Greinacher, 2003). Three options for such patients are bivalirudin,
lepirudin, and danaparoid (listed in order of preference). Unfortunately, the
lack of a specific antidote, the need for special intraoperative monitoring, and
other considerations mean that none is ideal for managing CPB. Another
approach is to administer heparin together with a potent antiplatelet agent,
e.g., tirofiban (GPIIb/IIIa antagonist) or epoprostenol (prostacyclin ana-
logue). This special topic of managing cardiac surgery patients with acute or
previous HIT is discussed in detail in Chap. 19, as well as in relation to specific
anticoagulant agents in Chaps. 14–17.
Danaparoid and lepirudin have also been used to provide intraoperative
anticoagulation, as well as to ‘‘flush’’ blood vessels during vascular surgery in
patients with acute HIT.
Recommendation. Alternative anticoagulation should be used for heart
or vascular surgery in a patient with acute or recent HIT with detectable
HIT antibodies. Either bivalirudin, lepirudin, or danaparoid are ap-
propriate alternatives for intraoperative anticoagulation, provided that
appropriate, rapid-turnaround laboratory monitoring and blood prod-
uct support to manage potentially severe bleeding complications are
available. Another approach is to give heparin together with a potent
antiplatelet agent (grade 2C).
et al., 2002c). The patient should receive routine doses of UFH for the surgical
procedure itself. Preoperative anticoagulation (e.g., for heart catheterization)
and postoperative antithrombotic prophylaxis can be achieved with a non-
heparin agent such as danaparoid (750 U b.i.d.-t.i.d.) or r-hirudin (15 mg
b.i.d. s.c.) (Eriksson et al., 1997) (see Chap. 14).
Recommendation. In a patient with a previous history of HIT, heart or
vascular surgery can be performed using heparin, provided that HIT
antibodies are absent (by sensitive assay) and heparin use is restricted to
the surgical procedure itself (grade 1C).
V. ADJUNCTIVE THERAPIES
A. Medical Thrombolysis
Thrombocytopenia is not a contraindication to thrombolytic therapy in
patients with HIT. Streptokinase (Fiessinger et al., 1984; Cohen et al.,
1985; Bounameaux et al., 1986; Cummings et al., 1986; Mehta et al., 1991),
urokinase (Leroy et al., 1985; Krueger et al., 1985; Clifton and Smith, 1986),
and tissue plasminogen activator (t-PA) (Dieck et al., 1990; Schiffmann et al.,
1997) have been used both systemically and by local infusion (Quinones-
Baldrich et al., 1989). In patients at high bleeding risk, an ultra-low–dose t-PA
(2 mg/h over 12 h) was successfully applied without bleeding complications
(Olbrich et al., 1998). As thrombin generation is not inhibited by thrombol-
ysis, concomitant nonheparin anticoagulation should be given, in reduced
dose, until the fibrinolytic effects have waned.
Recommendation. Regional or systemic pharmacological thrombolysis
should be considered as a treatment adjunct in selected patients with
limb-threatening thrombosis or pulmonary embolism with severe
cardiovascular compromise (grade 2C).
B. Surgical Thromboembolectomy
Vascular surgery is often needed to salvage an ischemic limb threatened by
HIT-associated acute arterial thromboembolism involving large arteries
(Sobel et al., 1988). When performing vascular surgery during acute HIT, it
is appropriate to maintain anticoagulation at least in the lower therapeutic
range, if possible, before, during, and after surgery, until platelet count
recovery. In patients with latent HIT (i.e., no longer thrombocytopenic, but
with clinically significant levels of HIT antibodies still present), the intensity
of anticoagulation depends on the perceived risk of vessel (or graft) occlu-
sion. In patients at high risk of occlusion (e.g., surgery involving below-knee
vessels), the patient should be therapeutically anticoagulated before vessel
clamping (in addition to receiving intraoperative flushes with anticoagu-
lant), with therapeutic anticoagulation maintained for several days after
surgery. In surgery involving larger vessels, the use of intraoperative flushes
alone, followed by postoperative prophylactic-dose anticoagulation, might
be sufficient.
Treatment of HIT 355
C. Intravenous Gammaglobulin
In vitro, both intact IgG as well as its Fc fragments inhibit HIT antibody-
induced platelet activation, an effect that depends somewhat on the method of
immunoglobulin preparation (Greinacher et al., 1994a) (see Chap. 9). Case
reports describe rapid increase in the platelet counts after high-dose intrave-
nous (iv) IgG (Vender et al., 1986; Frame et al., 1989; Nurden et al., 1991;
Grau et al., 1992; Prull et al., 1992; Warkentin and Kelton, 1994). The
possibility that ivIgG treatment interrupts platelet activation by HIT anti-
bodies provides a rationale for its use as an adjunct to anticoagulant therapy
in certain life- or limb-threatening situations. The dose should be 1 g/kg body
weight per day for 2 consecutive days.
Recommendation. ivIgG is a possible adjunctive treatment in selected
patients requiring rapid blockade of the Fc receptor-dependent platelet-
activating effects of HIT antibodies (e.g., management of patients with
cerebral venous thrombosis, severe limb ischemia, or very severe throm-
bocytopenia) (grade 2C).
D. Plasmapheresis
Plasmapheresis has been associated with successful treatment outcomes in
uncontrolled studies of patients with severe HIT (Vender et al., 1986; Bouvier
et al., 1988; Nand and Robinson 1988; Thorp et al., 1990; Manzano et al.,
1990; Brady et al., 1991; Poullin et al., 1998). Whether this is due to removal
of HIT antibodies or pathogenic immune complexes, or even correction of
356 Greinacher and Warkentin
E. Antiplatelet Agents
Dextran
Dextran in high concentrations inhibits platelet function and fibrinogen
polymerization. It also inhibits HIT antibody-mediated platelet aggregation
(Sobel et al., 1986). However, a prospective randomized trial (Chong et al.,
2001) (see Chap. 14) showed that in patients with severe HIT-associated
thrombosis, dextran was less effective therapy than danaparoid. It is unknown
whether dextran would provide additional clinical benefit if combined with
another anticoagulant. Neither of us uses dextran for the management of HIT.
Recommendation. Dextran should not be used as primary therapy for
acute HIT complicated by thrombosis (grade 1B).
C. Ancrod
Ancrod is a defibrinogenating enzyme obtained from the venom of the
Malayan pit viper. The thrombin-like enzyme cleaves fibrinopeptide A from
fibrinogen but, in contrast with thrombin, does not proteolyze fibrinopeptide
B (Bell, 1997). On the basis of uncontrolled studies, ancrod has been used
successfully to treat several patients with HIT, primarily in Canada (Teasdale
et al., 1989; Cole et al., 1990; Demers et al., 1991).
However, ancrod does not inhibit thrombin generation, and in HIT
patients it even appears to increase thrombin generation initially (Fig. 2)
(Warkentin, 1998). Animal models indicate that under special clinical circum-
stances, such as septicemia, ancrod contributes to enhanced fibrin deposition
(Krishnamurti et al., 1993). These data could help explain why some patients
have developed venous limb gangrene during combined treatment with
ancrod and warfarin (Warkentin et al., 1997; Gupta et al., 1998) (i.e., in-
creased thrombin generation during ancrod treatment could contribute to
the disturbance in procoagulant–anticoagulant balance during warfarin ther-
apy that has been hypothesized to explain venous limb gangrene) (Warken-
tin et al., 1997). In a retrospective nonblinded comparison, ancrod appeared
to be less effective than danaparoid in one medical community (Warkentin,
1996).
The manufacturer discontinued ancrod in 2002.
D. Platelet Transfusions
Usually there is no need to treat thrombocytopenia with platelet transfusions,
as patients with HIT rarely bleed spontaneously. Indeed, platelet transfusions
should be avoided because the transfused platelets can be activated by the
same immune mechanisms as the patient’s own platelets. Anecdotal experi-
ence describes thrombotic events soon after platelet transfusions given to
patients with acute HIT (Babcock et al., 1976; Cimo et al., 1979). Several
consensus conferences (Contreras, 1998; Hirsh et al., 2001b; British Commit-
tee for Standards in Haematology, 2003) stated that thrombotic thrombo-
cytopenic purpura (TTP) and HIT are two disorders in which prophylactic
360 Greinacher and Warkentin
REFERENCES
Cohen JI, Cooper MR, Greenberg CS. Streptokinase therapy of pulmonary emboli
with heparin-associated thrombocytopenia. Arch Intern Med 1985; 145:1725–
1726.
Contreras M. The appropriate use of platelets: an update from the Edinburgh Con-
sensus Conference. Br J Haematol 1998; 101(suppl 1):10–12.
Copplestone A, Oscier DG. Heparin-induced thrombocytopenia in pregnancy. Br J
Haematol 1987; 65:248.
Cummings JM, Mason TJ, Chomka EV, Pouget JM. Fibrinolytic therapy of acute
myocardial infarction in the heparin thrombosis syndrome. Am Heart J 1986;
112:407–409.
Danhof M, de Boer A, Magnani HN, Stiekema JCJ. Pharmacokinetic considerations
on Orgaran (Org 10172) therapy. Hemostasis 1992; 22:73–84.
Demers C, Ginsberg JS, Brill-Edwards P, Panju A, Warkentin TE, Anderson DR,
Turner C, Kelton JG. Rapid anticoagulation using ancrod for heparin-induced
thrombocytopenia. Blood 1991; 78:2194–2197.
Dieck JA, Rizo-Patron C, Unisa A, Mathur V, Massumi GA. A new manifestation
and treatment alternative for heparin-induced thrombosis. Chest 1990; 98:
1524–1526.
Eichler P, Lubenow N, Greinacher A. Results of the third prospective study of treat-
ment with lepirudin in patients with heparin-induced thrombocytopenia (HAT)
[abstr]. Blood 2002; 100(suppl 1):704a.
Elalamy I, Lecrubier C, Potevin F, Abdelouahed M, Bara L, Marie JP, Samama M.
Absence of in vitro cross-reaction of pentasaccharide with the plasma heparin-
dependent factor of twenty-five patients with heparin-associated thrombocyto-
penia. Thromb Haemost 1995; 74:1384–1385.
Eriksson BI, Wille-Jorgensen P, Kalebo P, Mouret P, Rosencher N, Bosch P, Baur M,
Ekman S, Bach D, Lindbratt S, Close P. A comparison of recombinant hirudin
with a low-molecular-weight heparin to prevent thromboembolic complications
after total hip replacement. N Engl J Med 1997; 337:1329–1335.
Farner B, Eichler P, Kroll H, Greinacher A. A comparison of danaparoid and lepirudin
in heparin-induced thrombocytopenia. Thomb Haemost 2001; 85:950–957.
Fiessinger JN, Aiach M, Rocanto M, Debure C, Gaux JC. Critical ischemia during
heparin-induced thrombocytopenia. Treatment by intra-arterial streptokinase.
Thromb Res 1984; 33:235–238.
Frame JN, Mulvey KP, Phares JC, Anderson MJ. Correction of severe heparin-
associated thrombocytopenia with intravenous immunoglobulin. Ann Intern
Med 1989; 111:946–947.
Francis JL, Drexler JL, Gwyn G, Moroose R. Bivalirudin, a direct thrombin inhibitor,
in the treatment of heparin-induced thrombocytopenia [abstr]. J Thromb Hae-
most 2003; 1(suppl 1):P1909.
Girolami B, Prandoni P, Stefani PM, Tanduo C, Sabbion P, Eichler P, Ramon R,
Baggio G, Fabris F, Girolami A. The incidence of heparin-induced throm-
bocytopenia in hospitalized medical patients treated with subcutaneous un-
fractionated heparin: a prospective cohort study. Blood 2003; 101:2955–2959.
Gosselin RC, Dager WE, King JH, Janatpour KA, Mahackian KA, Larkin EC,
Treatment of HIT 363
tive trials to assess the value of parenteral treatment with lepirudin and its
therapeutic aPTT range. Blood 2000; 96:846–851.
Greinacher A, Lubenow N, Eichler P. Anaphylactic and anaphylactoid reactions
associated with lepirudin in patients with heparin-induced thrombocytopenia.
Circulation 2003; 108:2062–2065.
Gruel Y, Lang M, Darnige L, Pacouret G, Dreyfus X, Leroy J, Charbonnier B. Fatal
effect of re-exposure to heparin after previous heparin-associated thrombocy-
topenia and thrombosis. Lancet 1990; 336:1077–1078.
Gruel Y, Lermusiaux P, Lang M, Darnige L, Rupin A, Delahousse B, Guilmot JL,
Leroy J. Usefulness of antiplatelet drugs in the management of heparin-asso-
ciated thrombocytopenia and thrombosis. Ann Vasc Surg 1991; 5:552–555.
Gupta AK, Kovacs MJ, Sauder DN. Heparin-induced thrombocytopenia. Ann Phar-
macother 1998; 32:55–59.
Guyatt G, Schunëmann H, Cook D, Jaeschke R, Pauker S, Bucher H. Grades of
recommendation for antithrombotic agents. Chest 2001; 119(suppl):3S–7S.
Hall AV, Clark WF, Parbtani A. Heparin-induced thrombocytopenia in renal failure.
Clin Nephrol 1992; 38:86–89.
Harrison L, Johnston M, Massicotte MP, Crowther M, Moffat K, Hirsh J. Com-
parison of 5-mg and 10-mg loading doses in initiation of warfarin therapy. Ann
Intern Med 1997; 126:133–136.
Henny CHP, ten Cate H, ten Cate JW, Prummel MF, Peters M, Büller HR.
Thrombosis prophylaxis in an AT III deficient pregnant women: application of
a low molecular weight heparinoid. Thromb Haemost 1986; 55:301.
Hérault JP, Lalé A, Savi P, Pflieger AM, Herbert JM. In vitro inhibition of heparin-
induced platelet aggregation in plasma from patients with HIT by SR 121566, a
newly developed Gp IIb/IIIa antagonist. Blood Coagul Fibrinolysis 1997;
8:206–207.
Hérault JP, Peyrou V, Savi P, Bernat A, Herbert JM. Effect of SR121566A, a potent
GP IIb–IIIa antagonist on platelet-mediated thrombin generation in vitro and
in vivo. Thromb Haemost 1998; 79:383–388.
Herbert JM, Savi P, Jeske WP, Walenga JM. Effect of SR 121566A, a potent GP IIb–
IIIa antagonist, on the HIT serum/heparin-induced platelet mediated activation
of human endothelial cells. Thromb Haemost 1998; 80:326–331.
Hirsh J, Warkentin TE, Raschke R, Granger C, Ohman EM, Dalen JE. Heparin and
low-molecular-weight heparin. Mechanisms of action, pharmacokinetics, dosing
considerations, monitoring, efficacy, and safety. Chest 1998; 114:489S–510S.
Hirsh J, Dalen JE, Guyatt G. The sixth (2000) ACCP guidelines for antithrombotic
therapy for prevention and treatment of thrombosis. Chest 2001a; 119(suppl):
1S–2S.
Hirsh J, Warkentin TE, Shaughnessy SG, Anand SS, Halperin JL, Raschke R,
Granger C, Ohman EM, Dalen JE. Heparin and low-molecular-weight heparin.
Mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and
safety. Chest 2001b; 119(suppl):64S–94S.
Horellou MH, Conard J, Lecrubier C, Samama M, Roque-D’Orbcastel O, de Fenoyl
O, Di Maria G, Bernadou A. Persistent heparin-induced thrombocytopenia
Treatment of HIT 365
despite therapy with low molecular weight heparin. Thromb Haemost 1984;
51:134.
Huhle G, Geberth M, Hoffmann U, Heene DL, Harenberg J. Management of heparin-
associated thrombocytopenia in pregnancy with subcutaneous r-hirudin. Gy-
necol Obstet Invest 2000; 49:67–69.
Janson PA, Moake JL, Garpinito C. Aspirin prevents heparin-induced platelet aggre-
gation in vivo. Br J Haematol 1983; 53:166–168.
Jouanny P, Jeandel C, Laurain MC, Penin F, Cuny G. Thrombopénie a l’héparine
et filtre cave. Difficultés du traitement. J Mal Vas 1993; 18:320–322.
Kappa JR, Horn MK III, Fisher CA, Cottrell ED, Ellison A, Addonizio VP Jr.
Efficacy of iloprost (ZK36374) versus aspirin in preventing heparin-induced
platelet activation during cardiac operations. J Thorac Cardiovasc Surg 1987;
97:405–413.
Kappa JR, Fisher CA, Addonizio VP. Heparin-induced platelet activation: the role of
thromboxane A2 synthesis and the extent of granule release in two patients. J
Vasc Surg 1989; 9:574–579.
Kelton JG, Sheridan D, Santos A, Smith J, Steeves K, Smith C, Brown C, Murphy
WG. Heparin-induced thrombocytopenia: laboratory studies. Blood 1988; 72:
925–930.
Keularts IMLW, Béguin S, de Zwaan C, Hemker HC. Treatment with a GPIIb/III
antagonist inhibits thrombin generation in platelet rich plasma from patients.
Thromb Haemost 1998; 80:370–371.
Kirchmaier CM, Bender N. Heparin-induzierte Thrombozytopenie mit arterieller und
venöser Thrombose. Inn Med 1988; 15:174–178.
Kleinschmidt S, Ziegenfuss T, Seyfert UT, Greinacher A. Septisch toxisches Herz
Kreislauf Versagen als Folge einer Heparin-induzierten Thrombozytopenie mit
‘‘White Clot Syndrome’’. Anaesthesiol Intensivmed Notfallmed Schmerzther
1993; 28:58–60.
Klement D, Rammos S, von Kries R, Kirschke W, Kniemeyer HW, Greinacher A.
Heparin as a cause of thrombus progression. Heparin-associated thrombocy-
topenia is an important differential diagnosis in paediatric patients even with
normal platelet counts. Eur J Pediatr 1996; 155:11–14.
Krishnamurti C, Bolan C, Colleton CA, Reilly TM, Alving BM. Role of plasminogen
activator inhibitor-1 in promoting fibrin deposition in rabbits infused with an-
crod or thrombin. Blood 1993; 82:3631–3636.
Krueger SK, Andreas E, Weinand E. Thrombolysis in heparin-induced thrombocy-
topenia with thrombosis. Ann Intern Med 1985; 103:159.
Lagrange F, Vergnes C, Brun JL, Paolucci F, Nadal T, Leng JJ, Saux MC, Banwarth B.
Absence of placental transfer of pentasaccharide (fondaparinux, arixtra) in the
dually perfused human cotyledon in vitro. Thromb Haemost 2002; 87:831–835.
Laster JL, Elfrink R, Silver D. Reexposure to heparin of patients with heparin-asso-
ciated antibodies. J Vasc Surg 1989; 9:677–681.
Leroy J, Leclerc MH, Delahousse B, Guerois C, Foloppe P, Gruel Y, Toulemonde F.
Treatment of heparin-associated thrombocytopenia and thrombosis with low
molecular weight heparin (CY 216). Semin Thromb Hemost 1985; 11:326–329.
366 Greinacher and Warkentin
Lewis BE, Wallis DE, Berkowitz SD, Matthai WH, Fareed J, Walenga JM, Barth-
olomew J, Sham R, Lerner RG, Zeigler ZR, Rustagi PK, Jang IK, Rifkin SD,
Moran J, Hursting MJ, Kelton JG, for the ARG-911 Study Investigators.
Circulation 2001; 103:1838–1843.
Lewis BE, Wallis DE, Leya F, Hursting MJ, Kelton JG. Argatroban anticoagulation
in patients with heparin-induced thrombocytopenia. Arch Intern Med 2003;
163:1849–1856.
Lindhoff-Last E, Piechottka GP, Rabe F, Bauersachs R. Hirudin determination in
plasma can be strongly influenced by the prothrombin level. Thromb Res 2000;
100:55–60.
Lubenow N, Greinacher A. Heparin-induced thrombocytopenia. Recommendations
for optimal use of recombinant hirudin. BioDrugs 2000; 14:109–125.
Lubenow N, Eichler P, Leitz T, Greinacher A. Meta-analysis of three prospective
studies of lepirudin in the prevention of thrombosis in patients with heparin-
induced thrombocytopenia. Blood [abstr] 2002a; 100(suppl 1):501a–502a.
Lubenow N, Eichler P, Greinacher A. Results of a large drug monitoring program
confirms the safety and efficacy of Refludan (lepirudin) in patients with immune-
mediated heparin-induced thrombocytopenia [abstr]. Blood 2002b; 100(suppl
1):502a.
Lubenow N, Kempf R, Eichner A, Eichler P, Carlsson LE, Greinacher A. Heparin-
induced thrombocytopenia: temporal pattern of thrombocytopenia in relation
to initial use or reexposure to heparin. Chest 2002c; 122:37–42.
Magnani HN. Heparin-induced thrombocytopenia (HIT): an overview of 230 patients
treated with Orgaran (Org 10172). Thromb Haemost 1993; 70:554–561.
Magnani HN. Orgaran (danaparoid sodium) use in the syndrome of heparin-induced
thrombocytopenia. Platelets 1997; 8:74–81.
Mak KH, Kottke-Marchant K, Brooks LM, Topol EJ. In vitro efficacy of platelet
glycoprotein IIb/IIIa antagonist in blocking platelet function in plasma of pa-
tients with heparin-induced thrombocytopenia. Thromb Haemost 1998; 80:
989–993.
Makhoul RG, Greenberg CS, McCann RL. Heparin-associated thrombocytopenia
and thrombosis: a serious clinical problem and potential solution. J Vasc Surg
1986; 4:522–528.
Makhoul RG, McCann RL, Austin EH, Greenberg CS, Lowe JE. Management of
patients with heparin-associated thrombocytopenia and thrombosis requiring
cardiac surgery. Ann Thorac Surg 1987; 43:617–621.
Manzano L, Yebra M, Vargas JA, Barbolla L, Alvarez-Mon M. Plasmapheresis in
heparin-induced thrombocytopenia and thrombosis [letter]. Stroke 1990; 21:
1236.
Markwardt F, Fink G, Kaiser B, Klöcking HP, Nowak G, Richter M, Stürzebecher
J. Pharmacological survey of recombinant hirudin. Pharmazie 1988; 43:202–
207.
Mattioli AV, Bonetti L, Sternieri S, Mattioli G. Heparin-induced thrombocytopenia
in patients treated with unfractionated heparin: prevalence of thrombosis in a 1
year follow-up. Ital Heart J 2000; 1:39–42.
Treatment of HIT 367
Harry N. Magnani
Medical Consultant, Oss, The Netherlands
I. INTRODUCTION
371
372 Chong and Magnani
Pharmacology
Danaparoid exerts its antithrombotic effects predominantly by inhibition of
factor Xa; it has only minimal antifactor IIa (antithrombin) activity. Its ratio
of antifactor Xa (anti-Xa) to antithrombin (anti-IIa) is 22:1 or higher, which is
considerably greater than that of LMWH (2:1–4:1) and UFH (1:1) (Meule-
man et al., 1982; Gordon et al., 1990; Meuleman, 1992). Its inhibition of
factor Xa is mediated by AT, and its minor effect on thrombin by both AT and
heparin cofactor II. Its highly selective inhibition of factor Xa confers on this
drug the advantage of a linear inhibitory effect on thrombin generation and
fibrin formation. Another advantage is that danaparoid does not interfere
with normal platelet function (Meuleman et al., 1982; Mikhailidis et al., 1984,
1987; Meuleman, 1987). Thus, unlike UFH, danaparoid does not interfere
with platelet accretion to experimental thrombi, although thrombus growth is
markedly reduced by its prevention of fibrin accretion. Similarly, danaparoid
has minimal effects on formation of the platelet-dependent hemostatic plug
(Meuleman, 1992). These beneficial characteristics of danaparoid contribute
to the high therapeutic index (i.e., favorable benefit/risk ratio) of this drug.
situation (Warkentin et al., 1998). These agents have in common the capacity
to inhibit thrombin generation, either by inhibition of factor Xa (danaparoid)
or by direct inhibition of thrombin (r-hirudin, argatroban).
In the compassionate-use program, it was recommended that HIT
patients with acute thrombosis receive intravenous danaparoid administered
as a bolus of 2500 U, followed by an infusion of danaparoid at 400 U/h for 4 h,
followed by 300 U/h for 4 h, and then 150–200 U/h for at least 5 days, aiming
for a plasma anti-Xa level of 0.5–0.8 anti-Xa U/mL. Table 1 describes a
similar protocol that takes into account the amount of danaparoid per
marketed ampule (750 anti-Xa U/ampule), as well as certain initial bolus
dose adjustments based on body weight. Danaparoid is also effective when
administered subcutaneously (de Valk et al., 1995): in this situation, the
equivalent 24-h actual or estimated intravenous dose is given in two to three
divided doses by subcutaneous injection over a 24-h period. For example,
2250 U (3 ampules) every 12 h by subcutaneous injection is approximately
equal to 190 U/h by intravenous infusion given over 24 h. In the compas-
sionate-use program, 464 patients with acute thromboembolism were treated
with danaparoid, with efficacy judged to be over 90% (Ortel and Chong,
1998).
Danaparoid treatment for HIT patients also proved efficacious in a
prospective, randomized, controlled clinical study (Chong et al., 2001). In this
trial, HIT patients with an acute thrombosis (venous, arterial, or both) were
randomized to receive either danaparoid plus warfarin, or dextran 70 plus
warfarin. Dextran is a glucose polymer, with an average molecular mass of
70 kDa. It is a weak antithrombotic agent that has been used to prevent DVT
in postoperative patients (Aberg and Rausing, 1978; Bergqvist, 1980). It is
known to block HIT antibody-induced platelet aggregation in vitro (Sobel et
al., 1986), and it has been suggested as a potentially useful drug for the
treatment of HIT. The reason for its use in the control group was that dextran
70 was the only rapidly acting antithrombotic drug available for the treatment
of HIT-associated thrombosis in Australia at study commencement in 1988.
The danaparoid treatment regimen was slightly different from that of
the compassionate-use program. Danaparoid was given as a bolus of 2400 U,
followed by an infusion of 400 U/h for 2 h, 300 U/h for 2 h, and then 200 U/h
for 5 days. In the dextran 70 arm, patients received dextran, 1 L on day 1, and
then 500 mL/day from days 2 to 5. In both treatment arms, the patients also
received warfarin, with doses adjusted to an INR of 2–4; the warfarin was
continued for 3 months. Patients were also stratified at randomization,
depending on the severity of their thrombosis, using predefined criteria.
Resolution of thrombocytopenia showed a nonsignificant trend in fa-
vor of danaparoid over dextran 70. Among the patients stratified as having
‘‘mild’’ thrombosis, a slightly higher percentage of thromboembolic events
Danaparoid for Treatment of HIT 377
Venous thromboembolism
Prophylaxis (prior HIT) 750 U sc, b.i.d. or t.i.d.
Prophylaxis (acute HIT) Treatment doses (see below) may be appropriate for prophylaxis
of acute HIT (see pp. 16, 348–349, 379)
Venous or arterial thrombo- 2250 U iv bolusa followed by 400 U/h for 4 h, 300 U/h for 4 h,
embolism: Treatment then 150–200 U/h for z5 days, aiming for a plasma anti-Xa
(either prior or acute HIT) level of 0.5–0.8 U/mL
Subcutaneous administration schedule: 1500–2250 U sc b.i.d. (given
almost 100% bioavailability, 2250 U sc b.i.d. is approximately
equal to an iv infusion rate of 200 U/h)
Embolectomy or other Preoperative: 2250 U iv bolusa; intraoperative flushes: 750 U in
peripheral vascular surgery 250 mL saline, using up to 50 mL (see p. 355); postoperative:
750 U sc t.i.d. (low-risk patients) or 150–200 U/h (high-risk
patients) beginning at least 6 h after surgery
Hemodialysis (on alternate 3750 U iv before 1st and 2nd dialyses; 3000 U for 3rd dialysis;
days) then 2250 U for subsequent dialyses, aiming for plasma anti-Xa
level of <0.3* U/mL predialysis, and 0.5–0.8 U/mL during
dialysis (see also Chap. 18).
Hemofiltration 2250 U iv bolus, followed by 600 U/h for 4 h, then 400 U/h for
4 h, then 200–400 U/h aiming for a plasma anti-Xa level of
0.5–1.0 U/mL (see also Chap. 18).
Cardiopulmonary bypass 125 U/kg iv bolus after thoracotomy; 3 U/mL in priming fluid of
surgery (CPB) apparatus; 7 U/kg/h iv infusion commencing after CPB hookup,
and continued until 45 min before expectation of stopping CPB
(see also Chap. 19)
Cardiac catheterization Preprocedure: 2250 U iv bolus (3000 U if 75–90 kg and 3750 U
if >90 kg)
Percutaneous transluminal Preprocedure: bolus as per foregoing; postprocedure: 150–200 U/h
coronary angioplasty for 1–2 days after PTCA (or until removal of balloon pump)
(PTCA) or intra-aortic
balloon pump
Catheter patency 750 U in 50 mL saline, then 5–10 mL per port, or as required
Pediatric dosage considerations Prophylaxis: 10 U/kg sc b.i.d.
Treatment: 30 U/kg b.w., iv bolus, then 1.2–2.0 U/kg b.w./h
depending upon severity of thrombosis
Abbreviations: b.w., body weight; b.i.d., twice daily; iv, intravenous: t.i.d., three times daily.
Compatibility with intravenous solutions: Danaparoid is compatible for dilution with the following
solutions: saline, dextose, dextrose–saline, Ringer’s, lactated Ringer’s, 10% mannitol.
Preparation of solution for infusion: One option is to add four ampules containing 3000 U (i.e., 750 anti-
Xa U/0.6 mL ampule) of danaparoid to 300 mL of intravenous solution (i.e., a solution that comprises 10
U danaparoid per milliliter of intravenous solution: thus, an infusion rate of 40 mL/h corresponds to a
dose of 400 U/h: 20 mL/h to a dose of 200 U/h, and so on.
a
Adjust iv danaparoid bolus for body weight:<60 kg, 1500 U; 60–75 kg, 2250 U; 75–90 kg, 3000 U; >90
kg, 3750 U.
378 Chong and Magnani
treated with danaparoid (83%) improved compared with those who received
dextran 70 (73%). In contrast, a substantial and significant difference in
treatment outcome occurred in patients with ‘‘serious’’ thrombosis: 88% of
danaparoid-treated thromboembolic events recovered, compared with 44%
of those treated with dextran 70. These data suggest that the use of an effective
anticoagulant to treat thromboembolism associated with HIT is particularly
important in those with more severe disease.
Recently, treatment outcomes in patients with HIT who received either
danaparoid or lepirudin have been compared (Farner et al., 2001). Although
not a randomized trial, this study had important strengths. First, all patients
had serologically confirmed HIT, with about 70% having thrombosis at the
time of study entry. Second, all patients met the same inclusion and exclu-
sion criteria, had similar baseline characteristics, and were treated during the
same time period (25 months ending April 1996). Third, many patients were
studied (danaparoid, n = 126; lepirudin, n = 175). Furthermore, patients
were subdivided into those treated with prophylactic or therapeutic doses.
This study suggests that both danaparoid and lepirudin have similar
efficacy for treatment of HIT-associated thrombosis when given in therapeutic
doses: the day 42 success rate was about 80% for either agent, when failure
was defined as the cumulative event rate of a composite endpoint of new
thrombosis, death, or limb loss. When evaluating the single endpoint of new
thrombosis in those patients who received therapeutic doses of study drug,
danaparoid and lepirudin also showed similar efficacy (90.6 vs. 92.1%;
p = 0.74). Moreover, safety analysis of all patients (regardless of dose
received) showed significantly fewer major bleeds with danaparoid (2.5 vs.
10.4%; p = 0.009). These data suggest that the favorable therapeutic index of
danaparoid extends even to patients with a serious prothrombotic disorder
such as acute HIT complicated by thrombosis.
Figure 2 Comparison of the outcomes of HIT patients, with and without throm-
boembolic complications (TEC), before the start of alternative anticoagulation; dana-
paroid vs. lepirudin: Time-to-event analysis of the incidences of a combined endpoint
(new thromboembolic events, limb amputation, death; maximum, one endpoint per
patient) up to day 42. Among patients without TEC at baseline (most of whom were
treated with a prophylactic-dose regimen), there was a significantly higher incidence of
the combined endpoint among patients treated with danaparoid, compared with lepi-
rudin ( p = 0.02, long-rank test). This suggests that the approved, prophylactic-dose
regimen for danaparoid (750 U b.i.d. or t.i.d. by subcutaneous injection, without
anticoagulant monitoring) may be relatively less effective for managing patients with
isolated HIT, compared with the prophylactic-dose regimen for lepirudin (initial in-
travenous infusion rate, 0.10 mg/kg/h; subsequently, dose adjusted by aPTT). In
contrast, the combined endpoint did not differ significantly between danaparoid and
lepirudin for patients with TEC at baseline, suggesting that therapeutic (treatment)
doses of danaparoid (see Table 1) have similar efficacy as does therapeutic-dose
lepirudin (see Table 2 in Chap. 15). Also shown is the number of patients at risk on the
starting day and at subsequent 7-day intervals. (From Farner et al., 2001.)
Danaparoid for Treatment of HIT 381
Disappointingly, the new regimen does not appear to have reduced the
frequency of severe postoperative bleeding. This new dosing regimen was
used by Olin and coworkers (2000) to manage five patients with acute or prior
HIT for CPB: four of the patients experienced prolonged bleeding requiring
reexploration and extensive transfusions, and two patients had clots identi-
fied in the surgical field. Other investigators later amended the protocol to
continue danaparoid until completion of CPB (rather than stopping the
anticoagulant 45 min before the expected end of pump run) because of
problems with clotting in the CPB circuit or in the operative field (Fernandes
et al., 2000).
Advances in surgical method may permit other treatment approaches
in selected patients. For example, the off-pump (‘‘beating heart’’) technique
does not utilize CPB, and thus a far lower dose of danaparoid may be feasible
for intraoperative anticoagulation. This approach was used successfully to
perform multiple coronary artery bypass grafting in a patient with acute HIT
and unstable angina (Warkentin et al., 2001). A relatively low target plasma
antifactor Xa level (0.6 U/mL) was used, rather than the conventional level
(>1.5 U/mL) sought during CPB (see Chap. 19).
A randomized, double-blind comparison of danaparoid (n = 34) with
heparin (n = 37) for off-pump coronary artery bypass grafting in non-HIT
patients showed a nonsignificant trend to greater postoperative blood loss
(mean, 264 mL) but a significant increase in patients exposed to homologous
blood (53% vs. 27%) with danaparoid. However, clinical outcomes appeared
similar, and the authors concluded that danaparoid could be a valuable
option in patients undergoing off-pump surgery when heparin is contra-
indicated (Carrier et al., 2002).
Other approaches for managing CPB or off-pump surgery in patients
with acute or previous HIT are discussed further in Chaps. 13 and 19.
C. Laboratory Monitoring
Measurement of plasma anti-Xa levels using an amidolytic assay can be used
for the laboratory monitoring of danaparoid’s anticoagulant action. The
heparinoid does not significantly prolong the activated partial thromboplas-
tin time (APTT), prothrombin time (INR), or activated-clotting time (ACT),
except at very high doses. Hence, these assays cannot be used for laboratory
monitoring of danaparoid. However, monitoring is not required in many
clinical situations. The drug has a bioavailability of almost 100% after
subcutaneous administration, and because of its lack of plasma protein
interaction, predictable plasma levels are usually obtained with subcutaneous
or intravenous use. However, laboratory monitoring is recommended in the
following clinical settings: (1) patients with substantial renal impairment; (2)
patients with unusually low or high body weight; (3) patients with life- or
limb-threatening thrombosis; (4) patients with unexpected bleeding; and (5)
critically ill or unstable patients.
It must be emphasized that for any assay of danaparoid-associated
plasma anti-Xa activity, the standard calibration curve must be constructed
386 Chong and Magnani
using danaparoid, and not UFH or even LMWH (Laposata et al., 1998).
Because of its lack of interaction with plasma heparin-binding proteins,
danaparoid gives a dose-response relation different from these heparins,
and plasma anti-Xa levels during danaparoid treatment will be overestimated
if a LMWH standard curve is used. Indeed, there are differences in the stated
therapeutic range among these various glycosaminoglycan anticoagulants
(UFH, 0.2–0.4 U/mL by protamine titration; UFH, 0.3–0.7 anti-Xa U/mL;
LMWH, 0.6–1.0 U/mL; danaparoid, 0.5–0.8 anti-Xa U/mL) (Hirsh et al.,
1998; Laposata et al., 1998; Warkentin et al., 1998). In some clinical treatment
settings using danaparoid, it might be advisable to aim for a lower anti-Xa
level (e.g., about 0.3 U/mL for a patient judged to have a high risk of bleed-
ing); sometimes, a higher target anti-Xa level should be sought (e.g., about 1.0
U/mL for a patient with life- or limb-threatening venous or arterial throm-
bosis).
Anti-Xa levels are determined using a chromogenic assay (i.e., a method
similar to that performed for monitoring LMWH treatment). A standard
reference curve must be constructed using various dilutions of danaparoid
(e.g., 1.6, 1.0, 0.5, 0.3, and 0 U/mL danaparoid, diluted in pooled normal
platelet-poor plasma). Control plasma samples are prepared by adding
known quantities of danaparoid to normal pooled plasma aliquots (assuming
100% recovery of the known quantity of danaparoid added) in three different
concentrations approximating treatment situations (e.g., 0.2, 0.7, and 1.25
U/mL, corresponding to low-, mid-, and high-control danaparoid levels).
Aliquots stored at 70jC are stable indefinitely if used only once, without
refreezing and rethawing.
E. Adverse Effects
Severe bleeding, the most serious adverse effect of danaparoid, rarely occurs
except in patients who are treated with very high doses of the drug, or in those
who develop drug accumulation (renal failure), or who have additional hemo-
static or vascular defects. However, serious bleeding occurred in a significant
number of patients who had undergone CPB with danaparoid (Magnani et al.,
1997; Westphal et al., 1997, Fernandes et al., 2000; Olin et al., 2000). In con-
trast, bleeding was not seen in the randomized trial in which HIT patients
with venous or arterial thromboses received danaparoid (Chong, 1996). Com-
pared with CPB patients, these patients underwent less intense anticoagula-
tion and did not suffer from the additional insults of CPB and chest incision.
Skin hypersensitivity reactions have been reported with danaparoid, but
these are rare (Magnani, 1993). Osteoporosis (an important complication of
prolonged UFH treatment) was not detected in any danaparoid-treated pa-
tients in the compassionate-use program, even in those treated for more than
3 months. Despite the issue of in vitro cross-reactivity with danaparoid, it is
noteworthy that new-onset immune-mediated thrombocytopenia has never
been reported with this agent.
F. Availability of Danaparoid
Table 2 lists the countries in which danaparoid has been approved for the
treatment of HIT, either with or without associated thrombosis. In some
countries in which danaparoid is approved for DVT prophylaxis, physicians
have the legal option to prescribe danaparoid for HIT (i.e., for ‘‘off-label’’ use
in a nonapproved indication) (see Chaps. 19 and 20). Danaparoid is no longer
390 Chong and Magnani
Heparin-induced
DVT prophylaxis thrombocytopenia
North America
Canada X X X X
United States Xd
Europe
Austria X X X X
Belgium X X X
Denmark X X X
Finland X X
France Xe X X
Germany X X
Great Britain X X
Greece X
Ireland X X X
Italy X
Luxembourg X X X
Netherlands Xf Xf X X
Norway X X
Portugal X X X
Sweden X X X X
Switzerland X X X
Australasia and Africa
Australia X X
Japang
Korea X
New Zealand X X X X
South Africa X X
a
Danaparoid is no longer marketed in some of these countries, e.g., United States, Great
Britain, Norway.
b
Orthopedic and general surgery only (unless otherwise indicated); approval includes starting
danaparoid 1–4 h preoperatively (except for U.S.).
c
Approved dose of 750 U b.i.d.–t.i.d. may be too low for acute HIT (see pp. 16, 348–349, 379).
d
Elective hip surgery only.
e
Orthopedic and cancer surgery only.
f
Approval modified to facilitate approval for HIT in Finland and Germany.
g
Approved only for treatment of DIC.
Danaparoid for Treatment of HIT 391
marketed in some countries (e.g., United States [since April 2002], United
Kingdom, Norway).
III. CONCLUSION
ACKNOWLEDGMENTS
REFERENCES
Gitlin SD, Deeb GM, Yann C, Schmaier AH. Intraoperative monitoring of dana-
paroid sodium anticoagulation during cardiovascular operations. J Vasc Surg
1998; 27:568–575.
Gordon DL, Linhardt R, Adams HP. Low-molecular-weight heparins and heparin-
oids and their use in acute or progressing ischaemic stroke. Clin Neurophar-
macol 1990; 13:522–543.
Greinacher A, Michels I, Muller-Eckhardt C. Heparin-associated thrombocytopenia:
the antibody is not heparin specific. Thromb Haemost 1992; 67:545–549.
Harrison L, Johnston M, Massicotte MP, Crowther M, Moffat K, Hirsh J. Com-
parison of 5-mg and 10-mg loading doses in initiation of warfarin therapy. Ann
Intern Med 1997; 126:133–136.
Henny CP, ten Cate H, ten Cate JW, Surachno S, van Bronswijk H, Wilmink JM,
Ockelford PA. Use of a new heparinoid as anticoagulant during acute haemo-
dialysis of patients with bleeding complications. Lancet 1983; 1:890–893.
Henny CP, ten Cate H, ten Cate JW, Moulijn AC, Sie TH, Warren P, Buller HR. A
randomized blind study comparing standard heparin and a new low molecular
weight heparinoid in cardiopulmonary bypass surgery in dogs. J Lab Clin Med
1985a; 106:187–196.
Henny CP, ten Cate H, Surachno S, Stevens P, Buller HR, den Hartog M, ten Cate JW.
The effectiveness of a low molecular weight heparinoid in chronic intermittent
haemodialysis. Thromb Haemost 1985b; 54:460–462.
Hirsh J, Warkentin TE, Raschke R, Granger C, Ohman EM, Dalen JE. Heparin and
low-molecular-weight heparin. Mechanisms of action, pharmacokinetics, dosing
considerations, monitoring, efficacy, and safety. Chest 1998; 114:489S–510S.
Hirsh J, Warkentin TE, Shaughnessy SG, Anand SS, Halperin JL, Raschke R,
Granger C, Ohman EM, Dalen JE. Heparin and low-molecular-weight heparin:
mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and
safety. Chest 2001; 119(suppl):64S–94S.
Hoek JA, Nurmohamed MT, Hamelynck KJ, Marti RK, Knipscheer HC, ten Cate H,
Büller HR, Magnani HN, ten Cate JW. Prevention of deep vein thrombosis
following total hip replacement by low molecular weight heparinoid. Thromb
Haemost 1992; 67:28–32.
Insler SR, Kraenzler EJ, Bartholomew JR, Kottke-Marchant K, Lytle B, Starr NJ.
Thrombosis during the use of the heparinoid Organon 10172 in a patient with
heparin-induced thrombocytopenia. Anesthesiology 1997; 86:495–498.
Kikta MJ, Keller MP, Humphrey PW, Silver D. Can low molecular weight heparins
and heparinoids be safely given to patients with heparin-induced thrombocy-
topenia syndrome? Surgery 1993; 114:705–710.
Kodityal S, Manhas AH, Udden M, Rice L. Danaparoid for heparin-induced throm-
bocytopenia: an analysis of treatment failures. Eur J Haematol 2003; 71:1–5.
Koster A, Meyer O, Hausmann H, Kuppe H, Hetzer R, Mertzlufft F. In vitro cross-
reactivity of danaparoid sodium in patients with heparin-induced thrombocy-
topenia type II undergoing cardiovascular surgery. J Clin Anesth 2000; 12:324–
327.
Laposata M, Green D, Van Cott EM, Barrowcliffe TW, Goodnight SH, Sosolik RC.
College of American Pathologists Conference Therapy. The clinical use and
394 Chong and Magnani
Andreas Greinacher
Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
I. INTRODUCTION
central core (residues 3–30, 37–46, 56–57), a ‘‘finger’’ (residues 31–36), and a
loop (residues 47–55). Hirudin is very stable at extremes of pH (1.5–13.0) and
at high temperatures (>90jC). It is soluble in water, but insoluble in alcohol
or acetone. The isoelectric point of hirudin is approximately 4.
Hirudins for therapeutic use are now produced by recombinant bio-
technology, using the yeast Saccharomyces cerevisiae, yielding recombinant
hirudin (r-hirudin). Lepirudin, a desulfatohirudin, differs from natural hiru-
din by lacking the sulfate group at Tyr-63 and also has an NH2-terminal
leucine residue in place of the isoleucine. Although such structural differences
result in a 10-fold reduction in the dissociation constant of r-hirudin, as
compared with natural hirudin, r-hirudins remain highly selective inhibitors
of thrombin, with an inhibition constant for thrombin in the picomolar range
(Stone and Hofsteenge, 1986).
B. Pharmacology
Lepirudin acts independently of the cofactors antithrombin and heparin
cofactor II (Markwardt, 1992) and forms tight, noncovalent 1:1 complexes
with thrombin. Interacting with both binding sites, lepirudin is a bivalent
inhibitor of thrombin (cf. argatroban, a univalent DTI) (Fig. 1). Lepirudin
inhibits all the biological activities of thrombin.
Three amino acids (residues 46–48) near the NH2-terminus of hirudin
bind to the active site cleft on thrombin, while the core of the hirudin molecule
closes off the active site pocket of thrombin. The COOH-terminal tail of
hirudin interacts with the fibrinogen anion-binding site, helping to block
thrombin-catalyzed fibrinogen cleavage. Hirudin inhibits the feedback loop
whereby thrombin enhances its own generation via activation of factors Va
and VIIIa (Kaiser and Markwardt, 1986; Pieters et al., 1989). In addition to
inhibiting free thrombin, hirudin inhibits clot-bound thrombin (Hogg and
Jackson, 1989; Weitz et al., 1990) and thrombin bound to fibrin split products
(Weitz et al., 1998). In contrast, heparin-antithrombin complexes are unable
to access and inactivate clot-bound thrombin. This important difference
between hirudin and heparin might explain why hirudin is more effective
than heparin in dissolving mural thrombi in experimental models (Meyer
et al., 1998). Hirudin shows virtually no interaction with plasma proteins
(Glusa and Markwardt, 1990), and its activity is standardized in thrombin
inhibitory units (TIU): 1 TIU is the amount of hirudin inhibiting 1 U of
thrombin at 37jC. The specific activity of lepirudin is 16,000 TIU/mg.
C. Pharmacokinetics
Lepirudin is administered parenterally. Studies of plasma pharmacokinetics
in healthy subjects reveal a two-compartment model. The initial plasma half-
Lepirudin in HIT Treatment 399
Table 1 Maximum aPTT Ratios (Mean vs. Baseline Values) After Lepirudin
Administration
aPTT ratio
Dose group (mode of application),
(mg/kg body weight) iv bolus 6 h iv infusion sc
Single-dose studies
0.01 1.3 ND ND
0.02 1.5 ND ND
0.04 1.9 ND ND
0.05 ND ND 1.2
0.07 2.3 ND ND
0.10 2.3 1.4 1.4
0.15 ND 1.7 1.6
0.20 2.1 1.7 1.9
0.30 2.4 ND ND
0.35 ND ND 1.9
0.50 2.8 ND 2.0
Multiple-dose studies
0.1 every 24 h 2.4 ND ND
0.1 every 12 h 2.2 ND ND
0.5 every 24 h ND ND 2.3
0.5 every 12 h ND ND 2.1
The maximum ratios of mean aPTT values versus baseline are shown. For iv bolus application,
6-h continuous iv infusion, and sc application, maximum ratios were usually reached at 0–10
min, 3–6 h, and 2–3 h after first application, respectively. This is in good correlation with the
peak plasma lepirudin concentrations achieved with these different modes of application.
Generally, maximum aPTT ratios increased with higher lepirudin doses, with maximum values
of 2.1–2.4 prolongation of baseline for repeated doses above 0.1 mg/kg body weight.
Abbreviations: aPTT, activated partial thromboplastin time; iv, intravenous; ND, not done; sc,
subcutaneous.
The approved dose for lepirudin in patients with HIT and acute
thrombosis (with normal renal function) is an iv bolus of 0.40 mg/kg,
followed by an iv infusion of 0.15 mg/kg/h (Table 2). However, in patients
without massive, life-threatening thrombosis, and especially in elderly
patients, I recommend that the bolus be omitted and an initial infusion rate
of 0.10 mg/kg/h commenced so as to avoid overdosage in case of unrecog-
nized renal insufficiency. The infusion rate should then be adjusted according
to aPTT after 4 h.
Renal clearance (160–200 mL/min for an adult with normal body
surface area of 1.73 m2) and degradation account for approximately 90%
of the systemic clearance of lepirudin. The t1/2h of r-hirudin lengthens with
Lepirudin in HIT Treatment 401
Repeat aPTT determinations should be made 4–6 h after any dose adjustment. Abbreviations: aPTT, activated partial thromboplastin time; b.w.,
body weight; CPB, cardiopulmonary bypass; ECT, ecarin clotting tim; iv, intravenous; MI, myocardial infarction; PCI, percutaneous coronary
intervention; UA, unstable angina.
a
A maximum body weight of 100 kg should be used for dose calculations.
b
Adjust for renal insufficiency.
c
The ratio is based on comparison with the normal laboratory mean aPTT. If Actin FS or Neothromtin reagents are used, the aPTT target range is
usually 1.5–3.0.
d
Used in the HAT-1, HAT-2, and HAT-3 trials.
e
This is the author’s recommended starting dose in all HIT patients, unless life- or limb-threatening thrombosis is present.
f
Tested in a prospective, randomized trial after orthopedic surgery (Eriksson et al., 1996, 1997).
g
Stop 15 min before end of CPB; put 5 mg into CPB after disconnection to avoid clotting of pump.
h
The target lepirudin level pre-CPB (>2.5 Ag/mL) is lower than the level sought during CPB (3.5–4.5 Ag/mL) because of the addition of lepirudin to
the pump priming fluid (0.2 mg/kg body weight).
403
404 Greinacher
Lepirudin in HIT Treatment 405
E. Dose Adjustments
Generally, in lepirudin-treated patients, laboratory values to monitor the anti-
coagulant effect should be obtained prior to treatment, 4 h after the start of iv
infusion, and 4 h after every change in dose. For most patients, the primary
anticoagulation parameter used should be the aPTT, and testing should be
performed at least once daily during treatment with lepirudin. If the target
range is exceeded, the infusion should be stopped for 2 h and restarted at a
50% lower dose once the therapeutic range has been reached (Greinacher
et al., 1999a,b). When the dose is subtherapeutic, the infusion rate should be
increased by 20%.
Figure 2 Lepirudin standard curve. This curve was generated using seven normal
plasmas spiked with various concentrations of lepirudin (Ag/mL) using reagent Actin
FS and the BCS analyzer (Dade-Behring, Germany). Note that incremental changes in
activated partial thromboplastin time (aPTT) are much smaller as the dose-response
curve flattens at greater plasma lepirudin concentrations.
406 Greinacher
Renal Impairment
Lepirudin has been studied in patients with varying degrees of renal impair-
ment. It can be used safely and effectively in reduced dose (Table 3). In case of
transient renal failure close monitoring of aPTT is mandatory. To avoid
overdosing due to compensated renal insufficiency, I recommend avoiding the
initial lepirudin bolus (unless severe thrombosis is present) and starting the
lower infusion rate of 0.10 mg/kg/h iv, with subsequent adjustments accord-
ing to aPTT.
Transitioning to Warfarin
In patients with HIT, warfarin (or other coumarins) should be initiated only
after platelet levels have normalized. Further, no loading dose of warfarin
should be given. To cover the initial prothrombotic effects of warfarin,
therapeutic levels of lepirudin (aPTT ratio = 1.5–2.5) should be maintained
for at least 4–5 days after the initiation of oral anticoagulation. In case of a
rapid increase in the international normalized ratio (INR), prothrombin
levels should be used for dose finding.
F. Reversal/Removal of Lepirudin
Bleeding is an important and potentially severe consequence of hirudin
treatment (Antman, 1994; Neuhaus et al., 1994; Frank et al., 1999). As with
all DTIs, no specific antidote is available. In a patient with minor bleeding and
normal renal function, stopping the drug may suffice, since the drug concen-
tration drops quickly. However, when bleeding is life-threatening or the
patient has renal failure, cessation alone may not be adequate.
Table 3 Dosing Schedule for Lepirudin in Patients with HIT and Renal
Impairment
Objectives
The three HAT trials examined whether lepirudin administered iv to patients
with serologically confirmed HIT would safely reduce the risk of new arterial
or venous thrombosis, limb amputations, and death. The laboratory objective
was to determine whether the drug would allow an increase in the platelet
count in thrombocytopenic patients or maintain the baseline platelet val-
ues (in nonthrombocytopenic patients), while providing effective anticoagu-
lation. The latter was defined as a prolongation of the aPTT by 1.5- to 2.5-fold
over baseline values with no more than two dose increases. (Note: If Actin FS
or Neothromtin reagents were used, the aPTT target range was a 1.5- to 3.0-
fold prolongation.)
Patients
Patients were eligible for study if their platelet count fell by more than 50% or
to fewer than 100 109/L or if they exhibited new thrombosis while receiving
heparin. A strict criterion for study entry was laboratory confirmation of the
clinical diagnosis of HIT by the heparin-induced platelet activation (HIPA)
test (Greinacher et al., 1991; Eichler et al., 1999) (see Chap. 11).
Clinical outcomes included a composite endpoint (new thrombosis,
limb amputation, death) as well as each individual endpoint. Clinical events
that occurred between diagnosis and start of treatment with lepirudin were
included, as were all clinical events that occurred up to day 14 after stopping
lepirudin treatment. Clinical outcomes for lepirudin were compared with a
historical control group treated conventionally by Kaplan-Meier time-to-
event analysis, beginning at laboratory confirmation of HIT for lepirudin-
treated patients and one day after laboratory confirmation for controls.
Laboratory response was defined as (1) the maintenance of an on-
treatment aPTT ratio higher than 1.5 in at least 80% of measurements and
requiring no more than two dose increases and (2) an increase in the platelet
count to more than 30% from the nadir and to more than 100 109/L by day
10 of lepirudin treatment (thrombocytopenic patients), or maintenance of
normal platelet counts on days 3 and 10 (nonthrombocytopenic patients).
Lepirudin in HIT Treatment 409
B. HAT-1 Study
The HAT-1 study involved 82 patients with confirmed HIT: 51 patients were
assigned to dose regimen A1, 5 to regimen A2, 18 to regimen B, and 8 to
regimen C (Table 4) (Greinacher et al., 1999a). The median duration of treat-
ment was 10 days (range 3–47 days) for regimen A1, 9 days (7–29 days) for A2,
15 days (2–58 days) for B, and 9 days (3–25 days) for C.
Efficacy Outcomes
Compared with the control group, the lepirudin-treated group had signifi-
cantly lower rates of the combined endpoint of new thrombosis, limb am-
putation, and death at day 35 (25.4% vs. 52.1%; p = 0.014). This represented
a 51.2% reduction in risk for the combined endpoint (Table 5). Similarly, the
Lepirudin studies
Table 5 Incidences of Composite and Individual Clinical Endpoints in the Individual Clinical Studies and Their Meta-analyses
(see also Fig. 3)
Safety Outcomes
At 4 weeks the cumulative rate of bleeding events was not statistically dif-
ferent in the lepirudin group compared to control (39.6% vs. 35.3%; p =
0.60). During the study, 27 patients (32.9%) experienced one bleeding event,
with 11 patients (13.4%) suffering 15 major bleeding events. Of those, 8 were
at invasive sites and 7 were spontaneous. There were no significant differences
between groups in the frequency of bleeding events requiring transfusion
(9.9% vs. 9.1%; p = 0.59). There were no intracerebral or fatal hemorrhages
in the lepirudin group.
C. HAT-2 Study
The HAT-2 study involved 112 patients with confirmed HIT: 65 patients were
assigned to dose regimen A1, 4 to regimen A2, and 43 to regimen B (Table 4)
(Greinacher et al., 1999b). The overall median duration of treatment was 11
days (range 0–104 days); for regimen A1, it was 13 days (0–104 days); for A2,
10 days (1–58 days); and for B, 8 days (1–67 days).
Efficacy Outcomes
The average combined outcome rate (expressed per patient-day) markedly
decreased from the pretreatment period (5.1%) to the periods during (1.5%)
and after (0.6%) lepirudin treatment. Platelet count recovery was achieved in
87 of 94 (92.6%) evaluable patients, with median platelet counts increasing
about fourfold over the first 10 days.
Compared with the historical control group, the cumulative frequency
of the composite endpoint (new thrombosis, limb amputation, death) was
lower in the lepirudin group at all time points after laboratory confirmation
of HIT (Table 5). At 5 weeks, the frequencies were 30.9% (95% CI 21.0–40.7)
Lepirudin in HIT Treatment 413
Figure 3 Time-to-event analyses of efficacy and safety endpoints in the HAT-1 and
HAT-2 studies (combined) in comparison with the historical control group. (A)
Composite endpoint (new thromboembolic complication, limb amputation, or death);
(B) new thromboembolic complication; (C) limb amputation; (D) death; and (E) major
bleeding.
414 Greinacher
for the lepirudin group and 52.1% (95% CI 40.4–63.9) for the historical
control group ( p = 0.12). Lepirudin-treated patients fared somewhat better
than historical controls at 5 weeks for the individual outcomes of new
thromboses (17.4% vs. 32.1%; p = 0.26), and death (10.5% vs. 22.3%;
p = 0.21) but not for limb amputation (10.0% vs. 8.2%; p = 0.43). The
adjusted risk ratio for lepirudin-treated patients relative to historical controls
was 0.709 (95% CI 0.44–1.14; p = 0.15). Causes of death included multi-
organ failure (n = 3), sepsis (n = 2), heart failure (n = 2), pulmonary
embolism, ventricular fibrillation, shock, and apnea (n = 1 each). None of
the deaths were judged to be related to adverse effects of lepirudin.
Safety Outcomes
At 35 days, the cumulative frequency of bleeding events was 44.6% (95% CI
33.8–55.4) in the lepirudin group and 27.2% (95% CI 16.3–38.0) in the con-
trol group ( p = 0.0001 by log-rank test). There were no significant differences
between the lepirudin and control groups, however, in the frequency of
bleeding events requiring transfusion (12.9% vs. 9.1%; p = 0.23). Most se-
vere bleeding occurred at invasive sites. The frequency of serious spontaneous
bleeding, including gastrointestinal hemorrhages (2.1% for lepirudin vs.
5.0% for control) and pulmonary hemorrhages (2.1% for lepirudin vs.
1.7% for control), was low and not significantly different. No cerebral hem-
orrhages occurred in the lepirudin group.
D. HAT-3 Study
A third prospective trial, HAT-3, was the largest and involved 205 patients: 98
patients were assigned to dose regimen A1, 12 to regimen A2, and 84 to
regimen B (Eichler et al., 2002). Ten patients received lepirudin for CPB
(regimen C), and one received lepirudin by the sc route. Fifteen patients were
enrolled twice. For the efficacy parameters only the first treatment cycle was
calculated. For safety analysis, especially allergic reactions, all treatment cy-
cles were included.
Median treatment duration was 10 days over all treatment groups, 9
days (range 1–197 days) for regimen A1, 12 days (5–21 days) for regimen A2,
10 days (1–47 days) for regimen B, and 7 days (1–37 days) for regimen C.
Efficacy Outcomes
The results of HAT-3 confirmed the efficacy of lepirudin in HIT, as outcomes
were similar to HAT-1 and HAT-2. Of the 84 patients with HIT and isolated
thrombocytopenia, 9 (10.7%) experienced new thrombosis, 7 (8.3%) under-
Lepirudin in HIT Treatment 415
went limb amputation, and 11 (13.1%) died. Since patients were counted only
once if multiple events occurred, the incidence of the combined endpoint was
27.4% (23/84). Of the 110 patients with HIT and thrombosis (treatment reg-
imens A1 and A2), 19 (17.3%) experienced new thromboses, 6 (5.5%) un-
derwent limb amputation, and 16 (14.5%) died; in this group, the combined
endpoint rate was 31.8% (35/98).
The average combined event-rate per day, as observed before, during,
and after lepirudin treatment, supports the previous observation that heparin
cessation alone may not prevent serious complications in HIT. The mean
1.7-day delay in initiating therapy for clinically suspected HIT while await-
ing laboratory confirmation was associated with severe, even life-threaten-
ing clinical consequences, with an average event-rate per day of 5.6%. This
dropped to 0.87% during treatment and was 0.77% after cessation of lepir-
udin therapy.
Compared to the historical control, from laboratory confirmation until
end of the observation period, the combined endpoint was markedly reduced
(26.2% vs. 52.1%; p = 0.002) primarily due to a reduction in new thrombosis
(9.9% vs. 32.1%; p = 0.0002).
Safety Outcomes
The overall incidence of major bleeding was 19.5%, with 5 fatal outcomes
(2.4%). One patient who received concomitant thrombolysis experienced
intracranial hemorrhage. Of the 205 patients, 8 (3.9%)—7 in the HIT plus
thrombosis group and 1 in the HIT group—experienced 11 episodes of
allergic reactions to lepirudin, with 4 associated with an antibody to lepirudin.
There were no cases of anaphylaxis.
Efficacy Outcomes
When outcomes were assessed from the start of lepirudin treatment, the
combined endpoint for new thrombosis, limb amputation, and death was
416 Greinacher
Lepirudin in HIT Treatment 417
Safety Outcomes
There were no fatal or intracranial bleeds but, compared with the controls, the
cumulative incidence of bleeding was higher in the lepirudin group than in the
control group (42.0% vs. 23.6%; p = 0.001), and more lepirudin-treated
patients experienced bleeding requiring transfusion (18.8% vs. 7.1%; p =
0.02).
One of the more important points to emerge from the meta-analysis was
the relationship of aPTT ratios with lepirudin safety and efficacy. For low
aPTT ratios (<1.5), the incidence of the combined endpoint was not signif-
icantly reduced compared to the control (RR = 0.86; p = 0.72). In addition,
the risk of bleeding was not significantly greater in the lepirudin group than
in the control group (RR = 1.57; p = 0.42). At medium aPTT ratios (1.5–
2.5), efficacy was significantly greater for the lepirudin-treated patients than
for the controls (RR = 0.42; p = 0.009), but there was also an increased risk
of bleeding (RR = 3.21; p = 0.0003). At higher aPTT ratios (>2.5), the
efficacy of lepirudin was not enhanced, but there was an even greater risk of
bleeding.
Efficacy Outcomes
Of the 111 patients in this study during treatment with lepirudin, 3 (2.7%)
experienced new thromboses, 3 (2.7%) underwent limb amputation, and 5
(4.5%) deaths occurred. Most of the deaths were related to underlying
disease, not to HIT or treatment with lepirudin. Since patients were counted
only once if multiple events occurred, the incidence of the combined endpoint
was 10/111 (9.0%). The median platelet count rebounded to 150 109/L
within 4 days of beginning lepirudin treatment.
Safety Outcomes
Episodes of major bleeding occurred in 16/111 (14.4%) of patients in this
meta-analysis. aPTT ratios above 2.5 were associated with an increased risk of
bleeding, and bleeding rates were significantly lower in patients with aPTT
<60. Nearly all patients with bleeding complications had impaired renal
function. Antihirudin antibodies were detected in 36 of the 108 (33.3%) eval-
uable patients. There were no differences in adverse events or outcomes be-
tween patients with and without antihirudin antibodies. No anaphylaxis was
observed.
Efficacy Outcomes
In the routine clinical settings of the DMP, lepirudin-treated patients with
isolated HIT and HIT with thrombosis had the lowest incidence of all clinical
endpoints reported with any agent. The incidence of the combined clinical
endpoint in the 496 patients with HIT and thrombosis was 21.9%: 26 patients
(5.2%) experienced new thrombosis, 29 (5.8%) underwent limb amputation,
and 54 patients (10.9%) died. The largest cause of death was multiorgan
failure (23/54 patients [42.6%]), emphasizing the serious underlying medical
condition of these patients. The incidence of new thrombosis in this study
(5.2%) was lower than that observed in the HAT-1 and -2 meta-analysis
(10.1%). This may be due to physicians’ increased clinical experience with
lepirudin, as illustrated by the decision to begin lepirudin treatment immedi-
ately upon clinical diagnosis of HIT, thereby improving efficacy and safety
outcomes.
The combined endpoint of new thrombosis, limb amputation, and
death occurred in 96 (15.7%) of the 612 patients with isolated HIT; 13 pa-
tients (2.1%) experienced new thrombosis, 8 (1.3%) underwent limb ampu-
tation, and 75 patients (12.3%) died. These event rates are the lowest reported
for any agent used to treat HIT. As seen in the group of patients with HIT plus
thrombosis, the largest cause of death in this group was multiorgan failure
(39/75 patients, 52.0%).
The overall mortality rate due to new thrombosis in the group of 1108
patients treated with regimen A1 or B (thus, excluding patients receiving ‘‘mis-
cellaneous’’ treatments) (see Table 4) was low (15 patients, or 1.4%). Efficacy
variables in the DMP were even more favorable than those seen in the meta-
analyses of the HAT studies. This DMP thus confirms the efficacy of lepiru-
din in routine clinical practice for both the prophylaxis and the treatment of
thromboembolism in patients with HIT.
There were no differences in the mean infusion rates in patients with
HIT and thrombosis (0.12 mg/kg/h) and those with isolated HIT (0.11 mg/kg/
h) in the DMP. As lepirudin dose is adjusted based on aPTT, the major
difference between the two regimens is the initial bolus in HIT patients with
acute thrombosis. However, as discussed earlier, in my view the bolus should
be avoided in most situations to prevent overdosing.
Safety Outcomes
In the DMP incidence of bleeding was greatly decreased when compared to
the HAT clinical trials. In the group of 496 patients with HIT plus thrombo-
sis, there were 27 (5.4%) major bleeding episodes, and among the 612 patients
with isolated HIT, 36 (5.9%) had major bleeding. Allergic reactions were re-
420 Greinacher
ported in 4 (0.8%) patients in the HIT plus thrombosis group and in 1 (0.2%)
patient with isolated HIT. No anaphylaxis was reported.
The decreased incidence of bleeding events in the DMP most likely is
attributed to physicians’ greater experience with administering lepirudin and
monitoring its effects.
I. Antibody Formation
Because hirudin is a protein obtained from a nonhuman species, lepirudin can
induce antibody production in humans. Antibodies are induced by both iv
therapeutic-dose and sc prophylactic-dose use (Greinacher et al., 2003a).
Antihirudin antibodies have been detected in 44–74% of patients treated with
lepirudin (Huhle et al., 1998; Song et al., 1999; Eichler et al., 2000). Of 196
HIT patients treated with lepirudin for 5 or more days, 44% developed
antihirudin antibodies of the IgG class (Eichler et al., 2000). These antibodies
were not associated with an increase in thrombin-antithrombin (TAT)
complexes (Fig. 5). None of these patients developed allergic reactions to
lepirudin. Antibody formation occurred as early as day 4 and peaked at days
8–9 (Eichler et al., 2000).
Antilepirudin antibodies can extend the half-life of lepirudin (Liebe et
al., 2002), most likely by reduced renal filtration of lepirudin-antilepirudin
complexes (Fig. 6); in about 2–3% of patients with antilepirudin antibodies,
an inhibitory effect is seen (Huhle et al., 2001; Fischer et al., 2003). The
Lepirudin in HIT Treatment 423
J. Allergic Reactions
Lepirudin administration during prospective studies in patients with HIT was
associated with a low incidence of allergic events, as well as during the much
larger clinical trials in patients with acute coronary syndromes. Among the
adverse events reported were eczema, rash, pruritus, hot flushes, fever, chills,
urticaria, bronchospasm, cough, stridor, dyspnea, angioedema (variously of
the face, tongue, larynx), and injection-site reactions. Any causal relationship
of lepirudin to these adverse events is unclear.
To date, of 35,000–60,000 patients treated with lepirudin, nine patients
were judged to have had severe anaphylaxis in close temporal association with
424 Greinacher
Figure 6 This 53-year-old woman was admitted to the hospital because of an ankle
fracture. She received low molecular weight heparin for 10 days, but was switched to
unfractionated heparin because of a distal deep vein thrombosis (DVT). Ten days later
she presented with proximal DVT, pulmonary embolism, and a rapid fall in platelet
count from more than 200 to 12 109/L. She was switched to intravenous (iv) lep-
irudin (schedule A1). After normalization of platelet counts, she received overlapping
oral anticoagulation (phenprocoumon), with lepirudin stopped when the INR reached
2.0. Antihirudin antibodies were first detected on day 7; at the same time, the aPTT
increased despite a stable hirudin dosage of 0.05 mg/kg b.w. per hour.
lepirudin use (Greinacher et al., 2003b). All reactions occurred within minutes
of iv bolus lepirudin administration, with four fatal outcomes (three acute
cardiorespiratory arrests, one hypotension-induced MI). In these four cases, a
previous uneventful treatment course with lepirudin was identified (1–12
weeks earlier). In an additional patient with nonfatal anaphylaxis (who did
not receive a bolus), we found high-titer IgG antilepirudin antibodies. Since
lepirudin has been used in approximately 35,000 patients, the risk of ana-
phylaxis is estimated at 0.015% (5/32,500) in first-exposure and 0.16% (4/
2500) in reexposed patients (assuming 7.5% reexposure frequency). We and
others (Bircher et al., 1996) demonstrated high titer antihirudin antibodies of
the IgG class, but not of the IgE class in patients with hirudin-associated
anaphylaxis. IgG-dependent anaphylaxis likely is Fc receptor-mediated and
Lepirudin in HIT Treatment 425
C. Hemodialysis
Hirudin was the first anticoagulant to be used for hemodialysis, as performed
by Haas (1924) in Germany. Because native hirudin preparations were crude
and supply of leeches insufficient, hirudin was replaced by heparin to prevent
clotting during dialysis. Currently, more published reports describe lepirudin
for hemodialysis than the other DTIs (see Chap. 18).
Management of these patients requires careful dosing and frequent
monitoring. HIT patients with transient renal failure are difficult to manage
with lepirudin, because substantial dose adjustments are necessary, depend-
ing on the extent of renal failure. To reduce bleeding risk, we prefer admin-
istering a continuous iv infusion, starting at 0.005 mg/kg/h, with adjustments
Lepirudin in HIT Treatment 427
D. Lepirudin in Pregnancy
Data on the treatment of HIT during pregnancy are limited. In general, the
use of lepirudin during pregnancy is not recommended, as it crosses the
placenta. Zebrafish experiments indicate that thrombin has an important role
in early embryogenesis and that inhibition by lepirudin may cause cell regu-
lation defects (Jagadeeswaran et al., 1997). Experiments in rabbits showed
a fetal hirudin plasma concentration that was 1/60 that of the maternal
concentration (Markwardt et al., 1988), and embryotoxic effects were seen in
rabbits at high, but not low, doses (30 vs. 1–10 mg/kg/day, respectively) (Ber-
lex Laboratories, data on file).
Reports on the use of lepirudin in pregnancy are sparse (Lindhoff-Last
and Bauersachs, 2002). A pregnant woman with systemic lupus erythemato-
sus who was treated with dalteparin developed HIT at week 25. Her platelet
count dropped from 230 to 59 109/L, after which she was treated with
lepirudin (15 mg sc twice daily), with aPTT and ECT used to monitor her
dosage. Following delivery by cesarean section, she experienced no postpar-
tum bleeding complications, and treatment with lepirudin was continued for
several weeks thereafter (Huhle et al., 2000b). Another pregnant woman with
lupus anticoagulant and HIT was successfully treated for 36 weeks with
lepirudin.
A case report described a breastfeeding woman diagnosed with HIT
who was treated with sc lepirudin, 50 mg twice daily (Lindhoff-Last et al.,
2000a). No lepirudin was detected in her breast milk, although plasma levels
were within therapeutic range. Neither bleeding nor thrombosis occurred in
mother or infant.
Lepirudin and danaparoid are each classified by the FDA as pregnancy
category B, based on limited animal data. However, danaparoid does not
cross the placenta, and it has been used for prophylaxis and therapy of HIT
during pregnancy (Greinacher et al., 1993; Dager and White, 2002) (see
Chaps. 13 and 14).
E. Lepirudin in Children
Although rare in children, HIT is important in the differential diagnosis of
thrombocytopenia or unexplained thrombosis in the presence of heparin
administration (Ranze et al., 1999). Because of the rarity of HIT and its
clinical heterogeneity in pediatric patients, it is difficult to design a standard-
428 Greinacher
V. CONCLUSION
The r-hirudin lepirudin is a DTI that provides rapid and effective anti-
coagulation and significantly reduces the risk of thrombosis in patients with
HIT, including those with isolated thrombocytopenia. Fewer than 10% of all
patient groups with HIT developed a new thrombosis after start of active
treatment. The drug also reduced risk of limb amputation and death.
Published data on lepirudin include more than 8500 treated patients. Of
these, about 1500 patients were treated for HIT (the largest experience with a
DTI). An additional 7300 patients received lepirudin for ACS and PCI.
Lepirudin is given parenterally by iv infusion or sc injection. Recom-
mended lepirudin dosage schedules have been established (Table 2). Lepir-
udin has a short half-life, which presents an advantage if invasive surgical
procedures are indicated. However, its elimination strongly depends on renal
function. Bolus dosing should be avoided, especially in elderly patients, to
avoid overdosing. Lepirudin can be used safely and effectively in patients with
renal impairment by appropriate dosing according to serum creatinine and
regular monitoring. Lepirudin also allows for a safe and uncomplicated
transition to warfarin.
The most common adverse event in the prospective clinical trials was
bleeding. No antidote exists for the DTIs. Excess lepirudin can be removed by
hemofiltration, but clinical data are limited. Daily monitoring of aPTT is
recommended with dosage adjustments made as needed to maintain the target
aPTT value. Routine monitoring with ECT should be performed in high-dose
situations, such as those required during CPB.
Besides the 399 patients with HIT treated in prospective trials, an
additional 1329 patients received lepirudin for HIT in a postmarketing
surveillance study. Data on these patients, collected under routine clinical
conditions, showed the lowest incidence of the clinical endpoints of death,
new thrombosis, and amputations, with risk reductions exceeding those
reported in the prospective clinical trials. Even more importantly, the inci-
dence of major bleeding was low. These differences support the assumption
that outcomes in patients with HIT can be substantially improved by im-
mediately stopping heparin and starting lepirudin when HIT is strongly sus-
Lepirudin in HIT Treatment 429
ACKNOWLEDGMENTS
The HAT studies were performed jointly by the combined clinical research
team of Behring-Werke AG, Hoechst, and Aventis. The laboratory studies
were supported by Deutsche Forschungsgemeinschaft GR 1096-2/2 and 2/3,
and 2/4. Analysis of the HAT-3 data and the HAT 1-2-3 meta-analysis was
supported by a grant from Berlex Laboratories (Montville, NJ) and
Pharmion (Cambridge, UK). The study on a comparison of danaparoid and
lepirudin was supported by Organon NV (Oss, The Netherlands)
and Thiemann/Celltech (Essen, Germany). The assistance of Theresia Lietz
in data analysis is highly appreciated.
REFERENCES
Butler KD, Dolan SL, Talbot MD, Wallis RB. Factor VII and DDAVP reverse the
effect of recombinant desulphatohirudin (CGB 39393) on bleeding in the rat.
Blood Coagul Fibrinolysis 1993; 4:459–464.
Callas DD, Hoppensteadt DA, Iqbal O, Rubsamen K, Fareed J. Ecarin clotting time
(ECT) is a reliable method for the monitoring of hirudins, argatroban, efegatran
and related drugs in therapeutic and cardiovascular indications [abstr]. Blood
1995; 86(10 suppl 1):866a.
Clore GM, Sukumaran DK, Nilges M, Zarbock J, Gronenborn AM. The confor-
mation of hirudin in solution. A study using nuclear magnetic resonance, dis-
tance geometry and restrained molecular dynamics. EMBO J 1987; 6:529–553.
Dager WE, White RH. Use of lepirudin in patients with heparin-induced throm-
bocytopenia and renal failure requiring hemodialysis. Ann Pharmacother 2001;
35:885–890.
Dager WE, White RH. Treatment of heparin-induced thrombocytopenia. Ann Phar-
macother 2002; 36:489–503.
de Denus S, Spinler SA. Clinical monitoring of direct thrombin inhibitors using the
ecarin clotting time. Pharmacotherapy 2002; 22:433–435.
Deitcher SR, Topoulos AP, Bartholomew JR, Kichuk-Chrisant MR. Lepirudin anti-
coagulation for heparin-induced thrombocytopenia. J Pediatr 2002; 140:264–
266.
Dickneite G, Friesen HJ, Kumpe G, Reers M. Reduction of rH induced bleeding in
pigs by the administration of von Willebrand factor. Platelets 1996; 7:283–290.
Dickneite G, Nicolay U, Friesen HJ, Reers M. Development of an anti-bleeding agent
for recombinant hirudin induced skin bleeding in the pig. Thromb Haemost
1998; 80:192–198.
Diehl KH, Römisch J, Hein B, Jessel A, Ronneberger H, Paques EP. Investigation of
activated prothrombin complex concentrate as potential hirudin antidote in
animal models. Haemostasis 1995; 25:182–192.
Direct Thrombin Inhibitors Trialists’ Collaborative Group. Direct thrombin inhibi-
tors in acute coronary syndrome: principal results of a meta-analysis based on
individual patients’ data. Lancet 2002; 359:294–302.
Eichler P, Budde U, Haas S, Kroll H, Loreth RM, Meyer O, Pachmann U, Potzsch B,
Schabel A, Albrecht D, Greinacher A. First workshop for detection of heparin-
induced antibodies: validation of the heparin-induced platelet activation test
(HIPA) in comparison with a PF4/heparin ELISA. Thromb Haemost 1999;
81:625–629.
Eichler P, Friesen HJ, Lubenow N, Jaeger B, Greinacher A. Antihirudin antibodies
in patients with heparin-induced thrombocytopenia treated with lepirudin:
incidence, effects on aPTT, and clinical relevance. Blood 2000; 96:2373–2378.
Eichler P, Lubenow N, Greinacher A. Results of the third prospective study of treat-
ment with lepirudin in patients with heparin-induced thrombocytopenia (HIT)
[abstr]. Blood 2002; 100(suppl 1):704a.
Eriksson BI, Ekman S, Kälebo P, Zachrisson B, Bach D, Close P. Prevention of deep-
vein thrombosis after total hip replacement: direct thrombin inhibition with
recombinant hirudin, CGP 39393. Lancet 1996; 347:635–639.
Lepirudin in HIT Treatment 431
King DJ, Kelton JG. Heparin-associated thrombocytopenia. Ann Intern Med 1984;
100:535–540.
Kornalik F, Blombäck B. Prothrombin activation induced by ecarin—a prothrombin
converting enzyme from Echis carinatus venom. Thromb Res 1975; 6:57–63.
Koster A, Kuppe H, Hetzer R, Sodian R, Crystal GJ, Mertzlufft F. Emergent cardio-
pulmonary bypass in five patients with heparin-induced thrombocytopenia type
II employing recombinant hirudin. Anesthesiology 1998; 89:777–780.
Koster A, Hansen R, Grauhan O, Hausmann H, Bauer M, Hetzer R, Kuppe H,
Mertzlufft F. Hirudin monitoring using the TAS ecarin clotting time in patients
with heparin-induced thrombocytopenia type II. J Cardiothorac Vasc Anesth
2000a; 14:249–252.
Koster A, Hansen R, Kuppe H, Hetzer R, Crystal GJ, Mertzlufft F. Recombinant
hirudin as an alternative for anticoagulation during cardiopulmonary bypass in
patients with heparin-induced thrombocytopenia type II: a 1-year experience in
57 patients. J Cardiothorac Vasc Anesth 2000b; 14:243–248.
Latham P, Revelis AF, Joshi GP, DiMaio JM, Jessen ME. Use of recombinant hirudin
in patients with heparin-induced thrombocytopenia with thrombosis requiring
cardiopulmonary bypass. Anaesthesiology 2000; 92:263–266.
Liebe V, Brückmann M, Fischer KG, Haase KK, Borggrefe M, Huhle G. Biological
relevance of anti-recombinant hirudin antibodies–results from in vitro and in
vivo studies. Semin Thromb Hemost 2002; 28:483–489.
Lindhoff-Last E, Bauersachs R. Heparin-induced thrombocytopenia-alternative anti-
coagulation in pregnancy and lactation. Semin Thromb Haemost 2002; 28:439–
445.
Lindhoff-Last E, Willeke A, Thalhammer C, Nowak G, Bauersachs R. Hirudin treat-
ment in a breastfeeding woman [letter]. Lancet 2000a; 355:467–468.
Lindhoff-Last E, Piechottka GP, Rabe F, Bauersachs R. Hirudin determination in
plasma can be strongly influenced by the prothrombin level. Thromb Res 2000b;
100:55–60.
Liu H, Fleming NW, Moore PG. Anticoagulation for patients with heparin-induced
thrombocytopenia using recombinant hirudin during cardiopulmonary bypass.
J Clin Anesth 2002; 14:452–455.
Longrois D, de Maistre E, Bischoff N, Dopff C, Meistelman C, Angioi M, Lecompte T.
Recombinant hirudin anticoagulation for aortic valve replacement in heparin-
induced thrombocytopenia. Can J Anaesth 2000; 47:255–260.
Lubenow N, Greinacher A. Hirudin in heparin-induced thrombocytopenia. Semin
Thromb Hemost 2002; 28:431–438.
Lubenow N, Eichler P, Leitz T, Greinacher A. Meta-analysis of three prospective
studies of lepirudin in the prevention of thrombosis in patients with heparin-
induced thrombocytopenia [abstr]. Blood 2002a; 100(suppl 1):501a–502a.
Lubenow N, Eichler P, Greinacher A. Results of a large drug-monitoring program
confirms the safety and efficacy of Refludan (lepirudin) in patients with im-
mune-mediated heparin-induced thrombocytopenia (HIT) [abstr]. Blood 2000b;
100(suppl 1):502a.
Markwardt F. Development of hirudin as an antithrombotic agent. Semin Thromb
Hemost 1989; 15:269–282.
434 Greinacher
Markwardt F. Hirudin: the promising antithrombotic. Cardiovasc Drug Rev 1992; 10:
211–232.
Markwardt F, Fink G, Kaiser B, Klocking HP, Nowak G, Richter M, Sturzebecher J.
Pharmacological survey of recombinant hirudin. Pharmazie 1988; 43:202–207.
Mehta SR, Eikelboom JW, Rupprecht H-.J, Lewis BS, Natarajan MK, Yi C, Pogue J,
Yusuf S. Efficacy of hirudin in reducing cardiovascular events in patients with
acute coronary syndrome undergoing early percutaneous coronary interven-
tion. Eur Heart J 2002; 23:117–123.
Meyer BJ, Badimon JJ, Chesebro JH, Fallon JT, Fuster V, Badimon L. Dissolution of
mural thrombus by specific thrombin inhibition with r-hirudin: comparison
with heparin and aspirin. Circulation 1998; 97:681–685.
Nand S, Wong W, Yuen B, Yetter A, Schmulbach E, Gross Fisher S. Heparin-induced
thrombocytopenia with thrombosis: incidence, analysis of risk factors, and
clinical outcomes in 108 consecutive patients treated at a single institution. Am J
Hematol 1997; 56:12–16.
Neuhaus KL, von Essen R, Tebbe U, Jessel A, Heinrichs H, Maurer W, Doring W,
Harnjanz D, Kotter V, Kalhammer E, et al. Safety observations from the pilot
phase of the randomized r-Hirudin for Improvement of Thrombolysis (HIT-III)
study: a study of the Arbeitsgemeinschaft Leitender Kardiologischer Kranken-
hausärzte (ALKK). Circulation 1994; 90:1638–1642.
Neuhaus KL, Molhoek GP, Zeymer U, Tebbe U, Wegschieder K, Schroder R, Camez
A, Laarman GJ, Grollier GM, Lok DJ, Kuckuck H, Lazarus P. Recombinant
hirudin (lepirudin) for the improvement of thrombolysis with streptokinase in
patients with acute myocardial infarction: results of the HIT-4 trial. J Am Coll
Cardiol 1999; 34:966–973.
Nguyen TN, Gal P, Ransom JL, Carlos R. Lepirudin use in a neonate with heparin-
induced thrombocytopenia. Ann Pharmacother 2003; 37:229–233.
Nishida S, Fujita T, Kohno N, Atoda H, Morita T, Takeya H, Kido I, Paine MJ,
Kawabata S, Iwanaga S. cDNA cloning and deduced amino acid sequence of
prothrombin activator (ecarin) from Kenyan Echis carinatus venom. Biochem-
istry 1995; 34:1771–1778.
Novoa E, Seegers WH. Mechanisms of alpha-thrombin and beta-thrombin-E for-
mation: use of ecarin for isolation of meizothrombin 1. Thromb Res 1980; 18:
657–668.
Nowak G. Clinical monitoring of hirudin and direct thrombin inhibitors. Semin
Thromb Hemost 2001; 27:537–541.
Nowak G, Bucha E. Prothrombin conversion intermediate effectively neutralizes toxic
levels of hirudin. Thromb Res 1995; 80:317–325.
Nowak G, Bucha E. Quantitative determination of hirudin in blood and body fluids.
Semin Thromb Haemost 1996; 22:197–202.
Nowak G, Bucha E, Gööck T, Prasa D, Thieler H. Pharmakokinetik von Hirudin bei
gestörter Nierenfunktion. Haemostaseologie 1991; 11:152–157.
Nowak G, Bucha E, Gööck T, Thieler H, Markwardt F. Pharmacology of r-hirudin in
renal impairment. Thromb Res 1992; 66:707–715.
Nowak G, Bucha E, Brauns I, Czerwinski R. Anticoagulation with r-hirudin in regular
Lepirudin in HIT Treatment 435
Bruce E. Lewis
Loyola University Medical Center, Maywood, Illinois, and Catholic
Health Partners, Chicago, Illinois, U.S.A.
Marcie J. Hursting
Clinical Science Consulting, Potomac, Maryland, U.S.A.
I. INTRODUCTION
II. ARGATROBAN
A. Chemical Description
Argatroban is a synthetic direct thrombin inhibitor derived from L-arginine
(Okamoto and Hijikata, 1981; Kikumoto et al., 1984). Its chemical structure is
shown in Fig. 1. Argatroban (molecular weight, 526.66) consists of a mixture
of 21-(R) and 21-(S) stereoisomers in a ratio of approximately 65:35 (Rawson
et al., 1993). There is no interconversion between these stereoisomers.
B. Clinical Pharmacology
Mechanism of Action
Argatroban is a potent and selective inhibitor of thrombin (Okamoto and
Hijikata, 1981; Kikumoto et al., 1984). Argatroban was developed using the
approach of rational drug design through the mimicry of substrates of
thrombin. It displays an inhibitory constant (Ki) of 0.04 Amol/L for thrombin
and has little or no effect on related serine proteases (K1 values of 5 Amol/L for
Argatroban Therapy in Heparin-Induced Thrombocytopenia 439
trypsin, 210 Amol/L for factor Xa, and 800 Amol/L for plasmin) (Kikumoto
et al., 1984). Argatroban exerts its anticoagulant effects in the absence of
any cofactor by inhibiting thrombin-catalyzed or -induced reactions, such as
fibrin formation, the activation of factors V, VIII, and XIII, and platelet
aggregation (Okamoto and Hijikata-Okunomiya, 1993).
Argatroban effectively inhibits free and clot-bound thrombin (Berry
et al., 1994; Hantgan et al., 1998). Argatroban is over 500-fold more potent
than r-hirudin in its relative ability to inhibit clot-bound versus free thrombin
(Berry et al., 1994). The lower molecular weight of argatroban, compared
with hirudin, may allow it better accessibility to thrombin incorporated
within the clot. The ability to inhibit effectively clot-bound thrombin may
be of particular benefit in treating hypercoagulable states such as HIT and
also in reducing extension of existing thromboses.
Structural studies have shown that argatroban binds tightly to throm-
bin by inserting the dual hydrophobic moieties on its arginine backbone into
deep clefts near the thrombin active site (Banner and Hadvary, 1993). Thus,
physiological substrates of thrombin are sterically hindered from access to the
catalytic pocket of thrombin. Figure 2 (see color insert) shows a model of the
interaction between argatroban and thrombin. This interaction is reversible,
unlike the irreversible interaction between r-hirudin and thrombin (Chap. 15).
The combination of reversible binding and a short elimination half-life (see
next subsection) may improve the ability to control anticoagulation in the
intensive care setting.
440 Lewis and Hursting
Figure 2 Model of the interaction between argatroban and thrombin. (See color
insert.)
which is somewhat less than the 1–2 h half-life of r-hirudin (Vanholder et al.,
1997). Argatroban is excreted primarily in the feces, presumably by biliary
secretion.
Pharmacokinetic–Pharmacodynamic Relationship
The pharmacokinetic and pharmacodynamic profiles of intravenously ad-
ministered argatroban are consistent with an anticoagulant agent that is
predictable, has a fast onset of action, and is rapidly eliminated (Swan and
Hursting, 2000; Swan et al., 2000).
The anticoagulant effects of argatroban are routinely monitored using
the activated partial thromboplastin time (aPTT). Higher levels of antico-
agulation, such as that required during interventional procedures, are moni-
tored using the activated clotting time (ACT). Argatroban also increases in a
dose-dependent fashion the prothrombin time (PT)/ International Normal-
ized Ratio (INR), thrombin time (TT), and ecarin clotting time (ECT) (Naga-
sawa et al., 1981; Clark et al., 1991; Walenga et al., 1999a; Swan et al., 2000;
Sheth et al., 2001). High-performance liquid chromatography (Rawson et al.,
1993; Walenga et al., 1999a) and liquid chromatography/tandem mass spec-
trometry (Tran et al., 1999) methods for measuring plasma argatroban are
described but are not practical (or needed) for routine clinical monitoring.
Immediately upon starting argatroban infusion, anticoagulant effects
are produced as plasma argatroban concentrations begin to rise. Steady-
state levels of both drug and anticoagulant effect typically are attained within
1–3 h (faster when a loading bolus is administered) and maintained with low
intra- and intersubject variability until the infusion is discontinued or the
dosage adjusted. Plasma drug concentrations increase proportionally with
doses up to 40 Ag/kg/min and are well correlated with steady-state antico-
agulant effects. The relationship at steady state between argatroban dose up
to 10 Ag/kg/min, plasma argatroban concentration, and anticoagulant effect
(aPTT) is shown in Fig. 3. On stopping infusion, plasma argatroban concen-
trations decline rapidly (half-life of 39–51 min), and anticoagulant effects
return to pretreatment values with similar effect half-lives (Swan et al., 2000).
Special Populations
Age, gender, and renal function exert no clinically significant effects on the
pharmacokinetics or pharmacodynamics of argatroban. Patients with mod-
erate hepatic impairment (Child–Pugh score>6), compared with healthy
volunteers, have an approximate fourfold decrease in drug clearance (to 1.5
mL/min/kg) and an approximate threefold increase in elimination half-life (to
152 min) (Swan and Hursting, 2000). Owing to the decreased clearance, a
442 Lewis and Hursting
Drug-Drug Interactions
No pharmacokinetic or pharmacodynamic drug interactions have been
demonstrated between argatroban and aspirin (Clarke et al., 1991), erythro-
mycin (Tran et al., 1999), acetaminophen, digoxin, or lidocaine (Inglis et al.,
2002). In practice, argatroban coadministered with these frequently used
medications should require no dosage adjustments.
No pharmacokinetic interactions have been demonstrated between
argatroban and warfarin (Brown and Hursting, 2002). However, because
Argatroban Therapy in Heparin-Induced Thrombocytopenia 443
D. Reversal of Argatroban
Argatroban has a gentle dose-response relationship that offers a wide margin
of safety during dose titration (see Fig. 3). However, as with any anticoag-
ulant, bleeding is a major safety concern. Excessive anticoagulation, with or
without bleeding, may be controlled by discontinuing argatroban or decreas-
ing its infusion dose. Anticoagulant parameters generally return to baseline
within 2–4 h after discontinuation of argatroban (Swan et al., 2000; Swan and
Hursting, 2000). This reversal takes longer (at least 6 h and up to more than
20 h) in patients with hepatic impairment (Swan and Hursting, 2000).
There is no specific antidote to argatroban available. If life-threatening
bleeding occurs and excessive plasma levels of argatroban are suspected,
argatroban should be discontinued immediately, and the patient should be
provided symptomatic and supportive therapy. Argatroban can be cleared,
albeit slowly, using high flux dialysis membranes (Murray et al., 2003), which
suggests a possible means to facilitate its removal if needed urgently. Recom-
binant factor VIIa has been suggested as a possible pharmacologic agent for
treating severe bleeding in this setting (Alving, 2002), although this remains to
be evaluated.
Study Objectives
The objective of study ARG-911 was to evaluate the use of argatroban as
an anticoagulant for the prophylaxis of thrombosis in HIT patients and
the treatment of HIT patients with thrombosis. Similarly, the objective of
studies ARG-915 and ARG-915X was to evaluate the safety and efficacy of
argatroban in HIT patients, with or without thrombosis, requiring anti-
coagulation.
Study Population
Adult patients were eligible if they had a clinical diagnosis of HIT with or
without thrombosis. HIT was defined as a platelet count <100 109/L, or a
50% decrease in the platelet count after initiation of heparin therapy, with no
apparent explanation other than HIT. Patients with a documented history of
a positive HIT antibody test who needed anticoagulation were also eligible for
the HIT study arm in the absence of thrombocytopenia. Patients were
excluded if they had an unexplained aPTT greater than 2 times control at
baseline, documented coagulation disorder or bleeding diathesis unrelated to
HIT, a lumbar puncture within the prior 7 days, or a history of previous
Figure 4 Schematic of the study design for ARG-911, ARG-915, and ARG-915X.
Patients with a clinical diagnosis of HIT with or without thrombosis were eligible.
The starting dose of argatroban, 2 Ag/kg/min, was titrated to achieve an aPTT 1.5–
3.0 times the baseline aPTT (not to exceed 100 s). Outcomes over a 37-day period
were compared with those of a historical control group.
Argatroban Therapy in Heparin-Induced Thrombocytopenia 447
Treatment
The treatment group received an initial dose of argatroban 2 Ag/kg/min via
continuous intravenous infusion. The aPTT was measured at least 2 h later,
and dosage was adjusted (up to 10 Ag/kg/min, maximum) until the aPTT was
1.5–3 times the baseline aPTT value (not to exceed 100 s). The aPTT was
measured daily and 2 h after each dosage adjustment. Patients remained on
argatroban for up to 14 days, until the underlying condition resolved or
appropriate anticoagulation was provided with other agents.
Assessments
The primary efficacy assessment was a composite endpoint of all-cause death,
all-cause amputation, or new thrombosis within a 37-day study period.
Additional analyses included the evaluation of event rates for the components
of the composite endpoint and death due to thrombosis. Secondary efficacy
endpoints included the achievement of adequate anticoagulation (i.e., an
aPTT >1.5 times baseline) and resolution of thrombocytopenia (i.e., platelet
count >100 109/L or z1.5 times baseline by study day 3).
Major bleeding was defined as overt and associated with a hemoglobin
decrease z2 g/dL, that led to a transfusion of z2 units, or that was intra-
cranial, retroperitoneal, or into a major prosthetic joint. Other overt bleeding
was considered minor.
B. ARG-911
In study ARG-911, 304 patients having clinically diagnosed HIT (n = 160) or
HIT with thrombosis (n = 144) received argatroban at a mean dose of 2.0 Ag/
kg/min for an average of 6 days. This study also enrolled 193 historical
controls (HIT, n = 147; HIT with thrombosis, n = 46). Although not required
for enrollment, laboratory confirmation of HIT antibodies occurred in 57%
of the argatroban-treated patients and 77% of controls; the remaining in-
dividuals were either never tested or had a negative result (Lewis et al., 2001).
448 Lewis and Hursting
Efficacy
As seen in Table 1, the composite endpoint was reduced significantly in
argatroban-treated patients versus controls with HIT (25.6% vs. 38.8%, p =
0.014). In HIT with thrombosis, the composite endpoint occurred in 43.8% of
argatroban-treated patients compared with 56.5% of controls ( p = 0.13).
Significant between-group differences by time-to-event analysis of the com-
posite endpoint favored argatroban treatment in HIT ( p = 0.010, hazard
ratio = 0.60; 95% CI, 0.40–0.89) (Fig. 5a) and HIT with thrombosis ( p =
0.014, hazard ratio = 0.57; 95% CI, 0.36–0.90) (Fig. 5b).
Argatroban therapy, compared with controls, significantly reduced
death due to thrombosis in each study arm (HIT, p = 0.005; HIT with
thrombosis, p < 0.001). There were no between-group differences in all-cause
mortality. The incidence of amputation (as the most severe outcome) was
similar between groups. Argatroban therapy also significantly reduced the
percentage of patients experiencing new thrombosis in each study arm (HIT,
p < 0.001; HIT with thrombosis, p = 0.044).
Argatroban-treated patients achieved therapeutic aPTTs generally at
first measure (i.e., within 4–5 h of starting therapy) and maintained these
levels throughout infusion. Resolution of thrombocytopenia occurred by day
3 in 53% of argatroban-treated patients with HIT and 58% of patients having
HIT with thrombosis. Compared with controls, argatroban-treated patients
had a significantly more rapid rise in platelet counts.
Safety
Major bleeding occurred in 6.9% (21/304) of argatroban-treated patients,
compared with 6.7% (13/193) of historical controls. In each group, there were
two fatal bleeding events. One patient experienced a fatal intracranial
hemorrhage 4 days after discontinuation of argatroban and following uroki-
nase and warfarin therapy; one historical control also experienced a fatal
intracranial hemorrhage. Minor bleeding rates were similar between the
groups (41%). The most common adverse events among argatroban-treated
patients with HIT or HIT with thrombosis, respectively, were diarrhea (11%)
and pain (9%).
C. Argatroban-915
A total of 418 patients with acute HIT (n = 189) or HIT with thrombosis (n =
229) were prospectively treated with argatroban in study ARG-915 or its
extension (together referred to as ‘‘Argatroban-915’’) (Lewis et al., 2003). The
mean argatroban dose was 1.8 Ag/kg/min, and the mean duration of therapy
Table 1 Comparisons of Argatroban-Treated Patients with Historical Controls in ARG-911
Figure 5 Time to first event for the composite endpoint through day 37 in study
ARG-911. Significant differences in favor of argatroban therapy were detected in
(a) the HIT study arm (argatroban group, n = 160; historical controls, n = 147) and
(b) the HIT with thrombosis study arm (argatroban group, n = 144; historical con-
trols, n = 46). (Data from Lewis et al., 2001.)
Argatroban Therapy in Heparin-Induced Thrombocytopenia 451
was 6 days. Comparisons were made with 185 historical controls with acute
HIT with or without thrombosis (obtained from ARG-911).
Efficacy
Efficacy results (Table 2) were confirmatory and supportive of those from
ARG-911. There were significant improvements in the composite endpoint
for argatroban-treated patients versus controls among those with HIT
(28.0% vs. 38.8%, p = 0.04) or HIT with thrombosis (41.5% vs. 56.5%,
p = 0.07). Argatroban treatment was significantly favored, compared with
control, by time-to-event analysis of the composite endpoint in HIT ( p =
0.02, hazard ratio = 0.64, 95% CI, 0.43–0.93) or HIT with thrombosis ( p =
0.008, hazard ratio = 0.56, 95% CI, 0.36–0.87).
Consistent with ARG-911, the positive benefits on the composite
endpoint were driven in main part by significant reductions in new thrombosis
( p < 0.001 in each study arm) (Table 2). There were no significant between-
group differences in all-cause mortality or amputation. Argatroban therapy
significantly reduced the incidence of death due to thrombosis in patients
having HIT with thrombosis ( p = 0.008).
Similar, predictable aPTT responses occurred in patients with HIT or
HIT with thrombosis. The target aPTT was typically achieved by first
assessment, and mean aPTT values remained generally constant throughout
the infusion. Platelet counts recovered more rapidly in argatroban-treated
patients than controls ( p < 0.001 for each study arm).
Safety
Major bleeding rates were not different between argatroban-treated patients
and controls in either study arm (Table 2). Twenty-four (5.7%) argatroban-
treated patients experienced major bleeding, including a single fatal event in a
patient hospitalized for rectal bleeding and who received urokinase. No
patient experienced an intracranial hemorrhage. Minor bleeding rates were
not different between the groups and were similar to those in ARG-911.
Table 2 Comparisons of Argatroban-Treated Patients and Historical Controls with Acute HIT in Argatroban-915
patients. Adverse outcome rates were significantly less than those of patients
with active HIT (e.g., composite endpoint rates of 16.1% vs. 36.3%, p <
0.001) and were comparable with rates that have been reported for similar
patients without HIT receiving standard therapy. Among 42 treatment
courses in 40 patients with a history of HIT who had fully recovered from
their initial episode of HIT, had a normal platelet count, and had no exposure
to heparin or other parenteral anticoagulants during their hospitalization,
none had a bleeding or new thromboembolic event (Matthai et al., 2004).
Argatroban therefore provided effective anticoagulation, upon initial and
repeat exposure, in patients with a history of HIT requiring acute anti-
coagulation for a variety of indications.
E. Argatroban Reexposure
Across the prospective studies of HIT, 55 patients underwent therapy with
argatroban on more than one occasion. The argatroban dosing and duration
were similar between these patients (repeat group) and patients upon their
first exposure (initial group, n = 754). Event rates in the repeat group were
less than with those in the initial group for the composite endpoint (20% vs.
34%), new thrombosis (3.6% vs. 11.1%), and major bleeding (3.6% vs.
6.6%). The patients reexposed to argatroban had no allergic reactions or
apparent differences, relative to the initial group, in adverse experiences.
Hence, argatroban is well tolerated upon reexposure, while providing effec-
tive antithrombotic therapy for HIT (Lewis et al., 2000).
Table 3 Dosing Schedules for Argatroban Treatment of Patients with HIT (Approved
Indications)
Monitoring and
Clinical use Bolusa IV infusiona adjusting therapy
steadt et al. 1997; Sheth et al., 2001), may be useful for supplemental moni-
toring during argatroban and warfarin cotherapy, if desired.
Clinical Studies
Argatroban has been evaluated in three multicenter, open-label prospective
studies in patients with HIT undergoing PCI, including percutaneous trans-
luminal coronary angioplasty, stent implantation, or rotational atherectomy.
The studies (ARG-216, ARG-310, and ARG-311) were similar in design with
respect to eligibility criteria, argatroban dosing regimen, and main outcome
assessments, and their pooled analysis has been reported (Lewis et al., 2002).
Among these studies, 91 patients with HIT underwent 112 PCIs on
argatroban anticoagulation (Lewis et al., 2002). Patients received 325 mg oral
aspirin 2–24 h before PCI. In the catheterization laboratory, patients received
intravenous argatroban at 25 Ag/kg/min (initial bolus dose of 350 Ag/kg)
titrated to achieve an ACT of 300–450 s during PCI (mean infusion dose, 23
Ag/kg/min). Additional bolus doses of 150 Ag/kg to achieve or maintain the
target ACT were allowed, though usually not needed. Target ACT values
were achieved typically within 10 min of initiating argatroban and were
maintained throughout the infusion. When argatroban was discontinued
after the procedure, ACTs rapidly returned to baseline.
Primary efficacy endpoints were subjective assessment of the satisfac-
tory outcome of the procedure and adequate anticoagulation, which occurred
in 94.5% and 97.8%, respectively, of patients undergoing their initial PCI
Argatroban Therapy in Heparin-Induced Thrombocytopenia 461
with argatroban (n = 91) (Table 4). Death (no patients), myocardial in-
farction (4 patients), and revascularization at 24 h after PCI (4 patients)
occurred in 7 (7.7%) patients. Other efficacy endpoints were also consistent
with argatroban enabling a satisfactory outcome (Table 4). One patient (1%)
experienced major periprocedural bleeding (nonfatal retroperitoneal hemor-
rhage). No unsatisfactory outcomes occurred during repeat PCIs with arga-
troban (n = 21; mean separation of 150 days from the initial PCI). Overall,
the clinical outcomes compared favorably with those reported historically for
heparin anticoagulation during PCI.
In a separate multicenter prospective study of 101 patients (including 1
patient with HIT) undergoing PCI, reduced doses of argatroban were
evaluated in combination with the GPIIb/IIIa antagonists abciximab (n =
99) or eptifibatide (n = 2) (Jang et al., 2003). Patients received argatroban as
an initial bolus of 250 Ag/kg followed by an infusion of 15 Ag/kg/min, and
additional boluses of 150 Ag/kg were allowed, if necessary, to achieve a target
ACT of 275–325 s. This target ACT was reached in 94 patients. Death (no
patients), myocardial infarction (3 patients), and urgent revascularization
at 30 days (2 patients) occurred in 3 (3%) patients. Two additional patients
had cardiac symptoms and elevated troponin without significant creatine ki-
nase elevation. There were 2 major bleeding events (1 retroperitoneal, 1 groin
B. Peripheral Intervention
Case reports describe the successful use of argatroban anticoagulation in
patients with HIT during renal stent implant (Lewis et al., 1997) and carotid
stent implant (Lewis et al., 1998). The argatroban dose and target ACT values
were the same as those recommended for PCI in the absence of GPIIb/IIIa
inhibition.
C. Argatroban in Hemodialysis
Argatroban has been used successfully for anticoagulation during hemodial-
ysis in patients with HIT (Matsuo et al., 1988, 1990; Koide et al., 1995; Reddy
et al., 2002; Mihindu et al., 2002), including a patient with comorbid hepatic
failure (Dager and White, 2003) (see also Chap. 18). In the latter case,
argatroban effectively prevented clotting in the dialyzer circuit, and consistent
with reduced argatroban clearance in patients with hepatic impairment, the
aPTT and INR were slow to recover after stopping argatroban. The effective
use of argatroban for the maintenance of catheter or graft patency between
dialysis treatments has also been reported (Mihindu et al., 2002). Although
guidelines are available for argatroban use in dialysis-dependent patients with
HIT at a major medical center (O’Shea et al., 2003), the safety and efficacy of
argatroban in patients with HIT undergoing hemodialysis have not been fully
evaluated in a clinical trial.
Argatroban administration by bolus alone, infusion alone, or bolus plus
infusion has been evaluated in a prospective cross-over study of 12 patients
with end-stage renal disease undergoing chronic hemodialysis (Murray et al,
2003). Target ACTs during dialysis were 140–180% of the baseline value. The
most satisfactory intradialysis anticoagulation was achieved using a steady-
state infusion of argatroban (2 Ag/kg/min begun f4 h before dialysis), or a
250 Ag/kg bolus dose at the start of dialysis followed by a continuous 2 Ag/kg/
min infusion. In 38 separate hemodialysis sessions, no dialysis membrane
required changing, and one (2.6%) session was shortened owing to circuit
clotting that occurred after 3 h of hemodialysis. There were no bleeding
events. Although confirmatory studies are required, it is anticipated that sim-
ilar dosing regimens may be adequate for inpatients with HIT already at
steady-state argatroban levels or outpatients with a history of HIT who re-
quire hemodialysis.
In support of the general safety of argatroban as an anticoagulant
during hemodialysis in patients with HIT, 54 argatroban-treated patients
with HIT underwent hemodialysis in studies ARG-911, ARG-915, and ARG-
915X. These protocols made no recommendations regarding argatroban
dosing during hemodialysis. For the patients who did, versus did not, undergo
464 Lewis and Hursting
D. Stroke
The effect of argatroban anticoagulation on stroke in HIT has been retro-
spectively evaluated using case records from studies of argatroban for HIT
(LaMonte et al., 2003). Stroke occurred in 3.0% of 1005 individuals with HIT
and was a significant predictor of death in argatroban-treated patients and
historical controls (odds ratio z3 for each group). Almost all strokes (33/35,
94%) were ischemic, consistent with the prothrombotic nature of HIT. Stroke
occurred most often in females, in those with a traditional risk factor for
stroke, in patients with more severe thrombocytopenia, and within 2 weeks of
HIT presentation. Compared with controls, argatroban therapy for HIT was
associated with reduced frequency of new stroke (1.8% vs. 4.7%, p = 0.032)
and stroke-associated mortality (1.0% vs. 3.1%, p = 0.036). These benefits
were achieved without increased intracranial or major bleeding. These data
highlight the importance of considering HIT in the differential diagnosis when
stroke occurs, particularly in hospital in-patients.
In a randomized, double-blind clinical study of patients treated with
argatroban vs. placebo within 12 h of ischemic stroke onset, there were no
significant between-group differences in intracranial hemorrhage or major
bleeding rates (LaMonte, 2003). Although not conducted in patients with
HIT, the study further supports the safety of argatroban anticoagulation in
patients with stroke.
As mentioned previously, argatroban is approved in Japan for use in
nonlacunar stroke and in Korea for use in acute cerebral thrombosis.
2001; Kieta et al., 2003; Ohno et al., 2003), and during (Edwards et al., 2003)
or immediately before and after CPB (Lubenow et al., 2003). Levels of
anticoagulation used during off-pump coronary bypass surgery tend to par-
allel those used during angioplasty. However, in the experience to date with
argatroban anticoagulation in HIT patients in this setting, infusion doses have
been generally similar to those recommended for the prophylaxis or treatment
of thrombosis in HIT, with target ACTs >200 s (Ide et al., 2001; Ohno et al.,
2003) or twice the baseline value (Kieta et al., 2003). A consistently safe and
effective dose to support CPB surgery in humans has not been established.
In vitro studies indicate that argatroban is at least as effective as heparin
in preventing thrombin generation in extracorporeal membrane oxygenation
(ECMO) circuits (Yonekawa et al., 2002). The successful use of argatroban
anticoagulation in ECMO has been described, including an adult patient with
HIT (Johnston et al., 2003) and two neonates, including one in whom ECMO
was continued for 78 days (Kawada et al., 2000). In each case, ACTs were
typically maintained >200 s, although further study is required to establish
dosing recommendations.
VI. CONCLUSION
REFERENCES
Lewis BE, Wallis DE, Zehnder JL, Barton JC, for the ARG-911/915/915X inves-
tigators. Argatroban reexposure in patients with heparin-induced thrombocy-
topenia [abstr]. Blood 2000; 96(Part 1):52a.
Lewis BE, Wallis DE, Berkowitz SD, Matthai WH, Fareed J, Walenga JM, Bar-
tholomew J, Sham R, Lerner RG, Zeigler ZR, Rustagi PK, Jang I-K, Rifkin SD,
Moran J, Hursting MJ, Kelton JG, for the ARG-911 Study Investigators.
Argatroban anticoagulant therapy in patients with heparin-induced thrombo-
cytopenia. Circulation 2001; 103:1838–1843.
Lewis B, Matthai WH, Cohen M, Moses JW, Hursting MJ, Leya F, for the ARG-
216/310/311 investigators. Argatroban anticoagulation during percutaneous
coronary intervention in patients with heparin-induced thrombocytopenia.
Catheter Cardiovasc Interv 2002; 57:177–184.
Lewis BE, Wallis DE, Leya F, Hursting MJ, Kelton JG, for the ARG-915 inves-
tigators. Argatroban anticoagulation in patients with heparin-induced throm-
bocytopenia. Arch Intern Med 2003; 163:1849–1856.
Lubenow N, Selleng S, Wollert HG, Eichler P, Mullejans B, Greinacher A. Heparin-
induced thrombocytopenia and cardiopulmonary bypass: perioperative arga-
troban use. Ann Thorac Surg 2003; 75:577–579.
Matsuo T, Chikahira Y, Yamada T, Nakao K, Ueshima S, Matsuo O. Effect of
synthetic thrombin inhibitor (MD805) as an alernative drug on heparin induced
thrombocytopenia during hemodialysis. Thromb Res 1988; 52:165–171.
Matsuo T, Yamada T, Yamanshi T, Ryo R. Anticoagulant therapy with MD805 of a
hemodialysis patient with heparin-induced thrombocytopenia. Thromb Res
1990; 58:663–666.
Matsuo T, Kario K, Matsuda S, Yamaguchi N, Kakishita E. Effect of thrombin
inhibition on patients with peripheral arterial obstructive disease: a multi-
center clinical trial of argatroban. J Thromb Thrombolysis 1995; 2:131–136.
Matthai WH, Hursting MJ, Lewis BE. Argatroban use in patients with a history of
heparin-induced thrombocytopenia who require acute anticoagulation [abstr].
Blood 2001; 98(part 2):45a.
Matthai WH, Hursting MJ, Lewis BE, Kelton JG. Argatroban anticoagulation in
patients with a history of heparin-induced thrombocytopenia. Submitted for
publication, 2004.
Mihindu JCL, Weinhold JR, Huntington N. Use of argatroban anticoagulation in
extracorporeal circulation and central venous catheter during hemodialysis in
patients with suspected heparin induced thrombocytopenia [abstr]. J Am Soc
Nephrol 2002; 13:189.
Mukundan S, Zeigler ZR. Direct antithrombin agents ameliorate disseminated intra-
vascular coagulation in suspected heparin-induced thrombocytopenia throm-
bosis syndrome. Clin Appl Thromb Hemost 2002; 8:287–289.
Murray P, Reddy B, Grossman E, Hammes M, Trevino S, Ferrell J, Tang I, Khorana
A, Fosbinder T, Swan SK. Safety and tolerability of argatroban anticoagulation
in patients with end-stage renal disease undergoing hemodialysis: a prospective
study [abstr]. J Thromb Haemost 2003; 1(suppl 1):P1911.
Nagasawa H, Fukutake K, Hada M, Takahashi E, Natsubara Y, Samori T, Ike-
472 Lewis and Hursting
Walenga JM, Ahmad S, Hoppensteadt DA, Iqbal O, Hursting MJ, Lewis BE.
Argatroban therapy does not generate antibodies that alter its anticoagulant
activity in patients with heparin-induced thrombocytopenia. Thromb Res
2002; 105:401–405.
Wallis DE, Workman DL, Lewis BE, Steen L, Pifarré R, Moran JF. Failure of early
heparin cessation as treatment for heparin-induced thrombocytopenia. Am J
Med 1999; 106:629–635.
Warkentin TE. Heparin-induced thrombocytopenia: pathogenesis and management.
Br J Haematol 2003; 121:535–555.
Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia.
Am J Med 1996; 101:502–507.
Willerson JT, Casscells W. Thrombin inhibitors in unstable angina: rebound or
continuation of angina after argatroban withdrawal? J Am Coll Cardiol 1993;
21:1048–1051.
Yonekawa KE, Nagakawa PA, Nugent DJ, Young G. Argatroban is as effective as
heparin at preventing thrombin generation in extracorporeal membrane oxy-
genation circuits [abstr]. Blood 2002; 11(part 2):127b.
17
Bivalirudin for the Treatment of
Heparin-Induced Thrombocytopenia
John R. Bartholomew
Cleveland Clinic Foundation, Cleveland, Ohio
I. INTRODUCTION
II. BIVALIRUDIN
A. Chemistry
Bivalirudin is a small synthetic 20-amino-acid peptide that is a specific and
reversible inhibitor of thrombin (Parry et al., 1994) (Fig. 1). Although it is an
analogue of hirudin, its amino acid sequence is considerably shorter. Bivali-
rudin unites a carboxy-terminal segment of 12 amino acids (dodecapeptide)
derived from native hirudin (residues 53–64) to an active site-binding tetra-
peptide sequence (D-Phe-Pro-Arg-Pro) at its amino terminal (Maraganore et
al., 1990; Nawarskas and Anderson, 2001; White and Chew, 2002). Four gly-
cine residues bridge these two segments together. The amino-terminal segment
has a high affinity and specificity for binding to the active site of thrombin
(Fareed et al., 1999; Sciulli and Mauro, 2002), while the carboxy terminal
B. Pharmacology
Bivalirudin is a bivalent DTI, i.e., it binds two distinct regions of thrombin:
the active (catalytic) site and the fibrinogen-binding site. Moreover, like
lepirudin and argatroban, bivalirudin binds to both free (soluble) and clot-
bound (fibrin-bound) thrombin. It forms a 1:1 stoichiometric complex that
neutralizes thrombin during coagulation and thrombus formation (Maraga-
nore and Adelman, 1996). Thus, bivalirudin inhibits proteolytic cleavage of
fibrinogen, thrombin-mediated activation of factors V and VIII, and throm-
bin-induced platelet activation.
Bivalirudin (unlike lepirudin) is a reversible inhibitor of thrombin (Fig.
1). It acts initially as a noncompetitive inhibitor, rendering thrombin inactive.
Circulating proteases (including other thrombin molecules) slowly cleave
bivalirudin near the amino-terminal end (between arg3-pro4), thus eventually
releasing the amino-terminal segment from the active site region of throm-
bin (Bates and Weitz, 1998; Carswell and Plosker, 2002; Reed and Bell, 2002;
Sciulli and Mauro, 2002). This allows thrombin to resume catalytic function.
As mentioned, bivalirudin also inhibits thrombin by the binding of its
carboxy-terminal segment to the fibrinogen-binding site on thrombin. This
occurs at the same time that the amino-terminal segment attaches to the active
site, thus resulting in dual blockage with complete inhibition of thrombin’s
multiple activities (Sciulli and Mauro, 2002). Once the amino-terminal moiety
of bivalirudin is cleaved, however, the carboxy-terminal region acquires low-
affinity, weakly competitive binding properties. Fibrinogen can now displace
the bivalirudin remnant from thrombin and align itself over the active site to
be converted to fibrin (Parry et al., 1994).
Bivalirudin is not inactivated by platelet factor 4, nor does it require any
cofactor for its activity. It does not bind to red blood cells or proteins other
than thrombin.
C. Pharmacokinetics
Bivalirudin has predictable pharmacokinetics, and exhibits a linear dose-
response relationship when given by the intravenous (iv) route to healthy
Bivalirudin in HIT Treatment 479
D. Pharmacodynamics
Bivalirudin produces an immediate effect after iv administration. It causes
prolongation of the prothrombin time (PT)/international normalized ratio
(INR), activated clotting time (ACT), the activated partial thromboplastin
time (aPTT), and the thrombin time (TT) (Fox et al., 1993; Lidon et al., 1993;
480 Bartholomew
Bivalirudin in HIT Treatment 481
Sharma et al., 1993; Topol et al., 1993). Although there is some interindivid-
ual variability, a dose of bivalirudin given as an infusion of 0.2 mg/kg/h in-
creased the aPTT from 27 to 62 s in one study, while an infusion rate of 1.0
mg/kg/h resulted in an average aPTT of 98 s in another group of patients
(Lidon et al., 1993).
The INR is also prolonged somewhat during bivalirudin infusion. In 54
healthy volunteers, a dose of 0.05–0.6 mg/kg of bivalirudin given over 15 min
iv increased the INR to between 1.25 and 2.43 (Fox et al., 1993). In a study by
Lidon and coworkers (1993), the PT was prolonged to between 12 and 16 s
with a dose of 0.2 mg/kg/h, while Francis and colleagues (2003; and
unpublished data) recently reported the mean INR to be 1.51 (range 1.26–
2.08) in 40 patients with suspected HIT treated with bivalirudin. Two recent
abstracts also mention a slight prolongation in the INR (Bufton et al.,
2002a,b). Although the increase in the INR seems not to be as great as with
the DTI argatroban, physicians should be aware of DTI-coumarin interac-
tions during overlapping therapy (see Chap. 13).
Bivalirudin decreases fibrinopeptide A levels (a marker of fibrinogen
cleavage) in patients with coronary artery disease (Cannon et al., 1993; Ren et
al., 1997). It may also increase the bleeding time in some patients (Topol et al.,
1993).
Bivalirudin does not inhibit platelet activation or aggregation directly,
but it has been shown to inhibit thrombin-mediated platelet aggregation
without affecting adenosine 5V-diphosphate (ADP) or collagen-mediated
platelet activation (Weitz and Maraganore, 2001; Wiggins et al., 2002; Witt-
kowsky, 2002) (Fig. 3). This antiplatelet effect may make bivalirudin more
useful than heparin in the platelet-rich environment of acute coronary syn-
dromes, where both platelet activation and thrombin formation play signifi-
cant roles.
The anticoagulant effects of bivalirudin reverse rapidly, with coagula-
tion times returning to baseline within 1–2 h after stopping the infusion (Fox
et al., 1993).
E. Dosage
Bivalirudin is approved for iv administration only. It has not yet been ap-
proved for use in HIT, and therefore no dosing guidelines for this indication
are established (Dager and White, 2002). In trials to date, however, several
dosing regimens have been reported. In three patients with venous and arterial
thrombosis treated with bivalirudin for HIT, Chamberlin and associates
(1994) used doses ranging from 0.05 to 0.2 mg/kg/h. Their goal was to
maintain a therapeutic aPTT greater than 50 s. Bufton and coworkers
(2002a) used an average dose of 0.27 mg/kg/h in one patient who received
bivalirudin for over 2 months.
Francis and colleagues (2003; and unpublished data) have used bivali-
rudin in 40 patients with clinically suspected HIT. Only two patients
were given iv boluses. Initial infusion rates usually ranged from 0.15 to
0.20 mg/kg/h; the overall mean infusion rate was 0.165 mg/kg/h (maximum
0.38 mg/kg/h). The target aPTT was a 1.5- to 2.5-fold prolongation of the
F. Administration
Bivalirudin is administered iv and produces a rapid anticoagulant effect (Fig.
2). In several small trials, however, it has also been given by subcutaneous (sc)
injection. In contrast to its rapid clearance following iv injection, its antico-
agulant effects are sustained for several hours following sc administration
(Fox et al., 1993) (Fig. 4). The peak anticoagulant effect occurred between 1
and 2 h after sc administration in a study of human volunteers, with detectable
plasma levels measured up to 6 h postinjection. The aPTT was prolonged
from 150 F 19.4% to 176 F 19.4% of the baseline value, and the INR
increased from 1.18 F 0.05 to 1.48 F 0.17 (Fox et al., 1993). Urinary excretion
of the drug was complete by 8–12 h.
A number of drugs commonly used in patients undergoing PCI have
been tested for Y-site compatibility with bivalirudin. Testing was for short-
term mixing, rather than longer-term interactions (4 h). Drugs found to be
compatible with bivalirudin included abciximab, dexamethasone, digoxin,
diphenhydramine, dobutamine, dopamine, epinephrine, eptifibatide, esmo-
lol, furosemide, heparin, lidocaine, morphine, nitroglycerin, potassium chlo-
ride, sodium bicarbonate, tirofiban, and verapamil (The Medicines
Company, 2001; Reed and Bell, 2002). Table 2 lists nine drugs found to cause
haze formation or gross precipitation, which thus should not be administered
in the same line as bivalirudin.
Drug-drug interaction studies have been performed with the thienopy-
ridine derivative ticlopidine, the glycoprotein (GP) IIb/IIIa inhibitors abcix-
imab, eptifibatide and tirofiban, and low molecular weight heparin and
unfractionated heparin (Reed and Bell, 2002). No pharmacodynamic inter-
actions occurred between bivalirudin and these agents. In patients undergoing
PCI, use of bivalirudin in conjunction with heparin, warfarin, or thrombolytic
therapy has been associated with increased risk of bleeding (The Medicines
Company, 2001). Aspirin was associated with a mild increase in bleeding
times in patients receiving bivalirudin infusions when compared to placebo.
These changes were not felt to be clinically significant (Fox et al., 1993).
G. Monitoring
The PT/INR, ACT, aPTT, and TT all rise linearly with increases in the dose of
bivalirudin. The ACT is generally used to monitor bivalirudin in patients
undergoing PCI, while the aPTT has been used in patients treated for HIT and
other non-PCI indications. Currently, the ECT (see Chap. 19) is recom-
mended for monitoring during on-pump cardiac surgery (CPB). Dosing in
PCI generally aims to maintain the ACT above 300 or 350, while in patients
486 Bartholomew
Bivalirudin in HIT Treatment 487
Alteplase
Amiodarone hydrochloride
Amphotericin B
Chlorpromazine hydrochloride
Diazepam
Prochlorperazine edisylate
Reteplase
Streptokinase
Vancomycin hydrochloride
Source: Reed and Bell, 2002; The
Medicines Company, 2001.
H. Reversal
There is no specific antagonist to bivalirudin. If renal function is normal,
bivalirudin is eliminated rapidly, and its anticoagulant effect clears within a
few hours after discontinuing the infusion. Kaplan and Francis (2002) have
suggested that recombinant factor VIIa and desmopressin may be of benefit if
bleeding occurs. Bivalirudin can be removed by hemodialysis (Irvin et al.,
1999).
Koster and colleagues (2003b) recently demonstrated that large
amounts of bivalirudin can be removed by hemofiltration and plasmaphere-
sis. They utilized five different hemofilters in an in vitro study (conditions
mimicking CPB) and observed a correlation between pore size and elimina-
tion rate. In their study, 65% of bivalirudin was removed using a hemofilter
with a large pore size (65,000 Da) (Mintech Hemocor HPH 700, Minneapolis,
MN), an amount comparable to that eliminated with a plasmapheresis filter
system (69%). This represents a 50% improvement over the amount of
lepirudin that can be removed through filtration (moreover, lepirudin filtra-
tion correlates poorly with pore size). These authors suggest that hemofiltra-
tion using appropriate filters may be useful for routine management of
patients who receive bivalirudin for cardiac surgery.
I. Adverse Effects
Bleeding is the major adverse effect of bivalirudin and occurs more commonly
in patients with renal impairment. Injection site pain has been reported in
individuals given sc bivalirudin (Fox et al., 1993). Mild headache, diarrhea,
Bivalirudin in HIT Treatment 489
Bivalirudin Heparin
Event (n = 2161) (n = 2151)
Cardiovascular
Hypotension 262 (12%) 371 (17%)
Hypertension 135 (6%) 115 (5%)
Bradycardia 118 (5%) 164 (8%)
Gastrointestinal
Nausea 318 (15%) 347 (16%)
Vomiting 138 (6%) 169 (8%)
Dyspepsia 100 (5%) 111 (5%)
Genitourinary
Urinary retention 89 (4%) 98 (5%)
Miscellaneous
Back pain 916 (42%) 944 (44%)
Pain 330 (15%) 358 (17%)
Headache 264 (12%) 225 (10%)
Injection site pain 174 (8%) 274 (13%)
Insomnia 142 (7%) 139 (6%)
Pelvic pain 130 (6%) 169 (8%)
Anxiety 127 (6%) 140 (7%)
Abdominal pain 103 (5%) 104 (5%)
Fever 103 (5%) 108 (5%)
Nervousness 102 (5%) 87 (4%)
nausea, and abdominal cramps have also been reported (Fox et al., 1993). In
the Hirulog Angioplasty Study (HAS) (now known as the Bivalirudin
Angioplasty Trial [BAT]), the most frequent adverse effects included back
pain, nausea, hypotension, pain, and headache. Approximately 5–10% of
patients reported insomnia, hypertension, vomiting, anxiety, dyspepsia, bra-
dycardia, abdominal pain, fever, nervousness, pelvic pain, and pain at the in-
jection site (Bittl et al., 1995; Sciulli and Mauro, 2002) (Table 3).
bosis and in one study involving humans. In a rat model of venous thrombosis
using injections of tissue thromboplastin combined with stasis, the adminis-
tration of bivalirudin demonstrated a dose-dependent interruption of throm-
bus formation (Maraganore et al., 1991).
Ginsberg et al. (1994a) studied iv and sc injections of bivalirudin in 10
patients with calf-vein thrombosis to determine if single injections could
inhibit thrombin generation in a sustained fashion. Prothrombin fragment
(F1+2) levels were used as an index of thrombin generation. Significant
reductions in F1+2 levels were noted at 6 h postinjection, but by 24 h levels
had increased significantly. These workers speculated that higher doses, more
frequent sc injections, or prolonged infusion was required to achieve ongoing
inhibition.
n 17 54 40 20 46
Overall DVT rate 10 (59%) 23 (43%) 16 (40%) 7 (35%) 8 (17%)a
Proximal DVT rate 7 (41%) 9 (17%) 6 (15%) 4 (20%) 1 (2%)b
Pulmonary embolism 0 2 (4%) 0 0 0
Major bleeding 0 1 (2%) 1 (3%) 0 1 (2%)
Minor bleeding 0 2 (4%) 0 1 (5%) 0
finding trial of 258 patients. The encouraging results led to larger studies of
patients requiring urgent angioplasty because of unstable or postinfarction
angina, the Hirulog (bivalirudin) Angioplasty Study (HAS) (for review, see
Nawarskas and Anderson, 2001). The primary endpoint was in-hospital
death, myocardial infarction (MI), or abrupt vessel closure within 24 h of
initiating PCI, or rapid clinical deterioration of cardiac origin. In the original
publication, no statistically significant difference in the primary endpoint was
noted between bivalirudin and heparin (Bittl et al., 1995), causing the sponsor
(Biogen) to abandon further drug development.
Subsequently, The Medicines Company reanalyzed the trial data (in-
cluding an additional 214 patients analyzed by intention-to-treat principle
who were not included in the per-protocol analysis initially reported). In this
study, renamed as Bivalirudin Angioplasty Trial (BAT), the frequency of
secondary endpoints (including death or MI, and major hemorrhage) were
found to be significantly reduced with bivalirudin. Bivalirudin was at least as
effective as heparin in preventing ischemic complications in patients who
underwent angioplasty for unstable angina and included fewer episodes of
492 Bartholomew
Table 5 Major Clinical Studies Using Bivalirudin in Cardiac Patients (PCI and
Non-PCI Indications)
Study acronym
or description Trial (Ref.)
PCI indications
Dose-finding study Multicenter, open-label study (Topol et al., 1993)
HAS Hirulog (Bivalirudin) Angioplasty Study
(Bittl, 1995; Bittl et al., 1995)
BAT Bivalirudin Angioplasty Trial (Bittl et al., 2001)a
CACHET Comparison of Abciximab Complications with
Hirulog Ischemic Events Trial (Lincoff et al.,
2002b)
REPLACE-1 Randomized Evaluation in PCI Linking Angiomax
to Reduced Clinical Events (REPLACE)-1 Trial
(Lincoff et al., 2002a)
REPLACE-2 Randomized Evaluation in PCI Linking Angiomax
to Reduced Clinical Events (REPLACE)-2 Trial
(Lincoff et al., 2003)
Angiomax in Practice Cho et al. (2003b)
Registry
Non-PCI (unstable angina or acute MI)
TIMI-7 Thrombin Inhibition in Myocardial Ischemia-7
(Fuchs and Cannon, 1995)
HERO-1 Hirulog Early Reperfusion/Occlusion-1
(White et al., 1997)
HERO-2 Hirulog Early Reperfusion/Occlusion-2
(White, 2001)
a
Bittl et al. (1995) reported the first study (combining two randomized, controlled trials)
comparing bivalirudin against heparin for PCI; this study, subsequently called the Bivalirudin
Angioplasty Trial (BAT), was later reanalyzed (including data from an additional 214 pa-
tients) (Bittl et al., 2001).
botic efficacy was believed by the authors to be acceptable, but detailed results
are forthcoming.
There is one case report of long-term (2-month) use of bivalirudin to
treat serologically confirmed HIT complicated by recurrent left leg ischemia
and arterial thrombosis while on low molecular weight heparin (Bufton et al.,
2002a). The patient received a continuous infusion of bivalirudin (22 mg/h)
using a CADD pump.
Table 7 summarizes theoretical advantages of bivalirudin as a treat-
ment for HIT.
were achieved in 98% and 96% of the patients, respectively. There were no
abrupt closures, nor was thrombus formation reported during or after PCI.
One patient died of cardiac arrest about 46 h after successful PCI.
Short half-life (25–36 min) Avoids need for initial iv bolus; rapid reversal
of anticoagulation (useful if patient develops
bleeding or if used for intraoperative
anticoagulation)a
Predominant enzymic Minor renal excretion (20%) means that risk of
metabolism overdosing in renal failure less than with
lepirudin; less risk of postoperative bleeding
(compared with lepirudin) if used for in-
traoperative anticoagulation (in case of
postoperative renal insufficiency)b
Minimal effect on PT/INR Simplifies transition to oral anticoagulation
(compared with argatroban)
Low immunogenicity Reduced risk of allergy and anaphylaxis
(compared with lepirudin)
Abbreviation: iv, intravenous.
a
Possible disadvantages of a short half-life include need for frequent sc administration (e.g.,
three or four times daily) and rapid loss of anticoagulation (with risk of rebound thrombosis)
if prematurely discontinued in patient with acute HIT.
b
Possible disadvantage of enzymic metabolism includes loss of anticoagulant action in stagnant
blood (implications for cardiac anesthesiology) (see Chap. 19).
Bivalirudin in HIT Treatment 499
The patient required 2 units of packed red blood cells intraoperatively and
underwent surgical reexploration 8 h postoperatively because of increased
chest tube drainage. No bleeding was found, and the patient’s recovery was
otherwise uneventful.
V. ANTIBIVALIRUDIN ANTIBODIES
Bivalirudin is the only anticoagulant associated with lower rates of both is-
chemic and bleeding complications compared to heparin in studies of PCI.
These complications are associated with increased morbidity and mortality
and also higher costs (Lauer, 2000; Compton, 2002). Bivalirudin may also be
Bivalirudin in HIT Treatment 501
associated with shorter hospital stay, use of fewer closure devices, lower in-
cidence of hematoma formation, earlier sheath removal, and more selective
use of the GP IIb/IIIa inhibitors. Potential savings are also possible in pa-
tients treated for HIT by reducing its devastating and costly thrombotic
complications.
VII. CONCLUSION
REFERENCES
Allie DE, Lirtzman MD, Wyatt CH, Keller VA, Khan MH, Khan MA, Fail PS,
Hebert CJ, Ellis SD, Mitran E, Chaisson G, Stagg S Jr, Allie AA, Walker CM.
Bivalirudin as a foundation anticoagulant in peripheral vascular disease: a safe
and feasible alternative for renal and iliac interventions. J Invasive Cardiol 2003;
15:334–342.
Alving BM. How I treat heparin-induced thrombocytopenia and thrombosis. Blood
2003; 101:31–37.
Antman EM, Braunwald E. A second look at bivalirudin. Am Heart J 2001; 142:929–
931.
Bates SM, Weitz JI. Direct thrombin inhibitors for treatment of arterial thrombosis:
potential differences between bivalirudin and hirudin. Am J Cardiol 1998;
82:12–18P.
Bates SM, Weitz JI. The mechanism of action of thrombin inhibitors. J Invasive
Cardiol 2000; 12(suppl F):27F–32F.
Berkowitz SD. Bivalirudin in heparin-induced thrombocytopenia (HIT) or heparin-
502 Bartholomew
tory activation after vascular injury in the rabbit. Cardiovasc Drugs Ther 1999;
13:429–434.
Lauer MA. Cost analysis of bivalirudin in percutaneous coronary intervention. J
Invasive Cardiol 2000; 12(suppl F):37F–40F.
Lidon RM, Theroux P, Juneau M, Adelman B, Maraganore J. Initial experience with a
direct antithrombin, Hirulog, in unstable angina. Anticoagulant, antithrom-
botic, and clinical effects. Circulation 1993; 88:1495–1501.
Lidon RM, Theroux P, Lesperance J, Adelman B, Bonan R, Duval D, Levesque J. A
pilot, early angiographic patency study using a direct thrombin inhibitor as
adjunctive therapy to streptokinase in acute myocardial infarction. Circulation
1994; 89:1567–1572.
Lincoff AM, Bittl JA, Kleiman NS, Kereiakes DJ, Harrington RA, Sarembook IJ,
Jackman JD, Mehta S, Maierson EF, Chew DP, Topol EJ. The REPLACE 1
Trial: a pilot study of bivalirudin versus heparin during percutaneous coronary
intervention with stenting and GP IIb/IIIa blockade [abstr]. J Am Coll Cardiol
2002a; 39(suppl A):16A.
Lincoff AM, Kleiman NS, Kottke-Marchant K, Maierson ES, Maresh K, Wolski KE,
Topol EJ. Bivalirudin with planned or provisional abciximab versus low-dose
heparin and abciximab during percutaneous coronary revascularization: results
of the Comparison of Abciximab Complications with Hirulog for Ischemic
Events Trial (CACHET). Am Heart J 2002b; 143:847–853.
Lincoff AM, Bittl JA, Harrington RA, Feit F, Kleiman NS, Jackman JD, Sarembock
IJ, Cohen DJ, Spriggs D, Ebrahimi R, Keren G, Carr J, Cohen EA, Betriu A,
Desmet W, Kereiakes DJ, Rutsch W, Wilcox RG, deFeyter PJ, Vahanian A,
Topol EJ. Bivalirudin and provisional glycoprotein IIb/IIIa blockade compared
with heparin and planned glycoprotein IIb/IIIa blockade during percutaneous
coronary intervention: REPLACE-2 randomized trial. JAMA 2003; 289:853–
863.
Mahaffey KW. Anticoagulation for acute coronary syndromes and percutaneous
coronary intervention in patients with heparin-induced thrombocytopenia.
Curr Cardiol Rep 2001; 3:362–370.
Mahaffey KW, Lewis BE, Wildermann NM, Berkowitz SD, Oliverio RM, Turco MA,
Shalev Y, Lee PV, Traverse JH, Rodriguez AR, Ohman EM, Harrington RA,
ATBAT Investigators. J Invasive Cardiol 2003. In press.
Maraganore JM, Adelman BA. Hirulog: a direct thrombin inhibitor for management
of acute coronary syndromes. Coron Artery Dis 1996; 7:438–448.
Maraganore JM, Bourdon P, Jablonski J, Ramachandran KL, Fenton JW II. Design
and characterization of hirulogs: a novel class of bivalent peptide inhibitors of
thrombin. Biochemistry 1990; 29:7095–7101.
Maraganore T, Oshima FA, Sugitachi A. Comparison of anticoagulant and anti-
thrombotic activities of hirulog-1 and argatroban (MD-805) [abstr]. Thromb
Haemost 1991; 65:651.
Merry AF, Raudkivi P, White HD, Nand P, Middleton N, McDougall J, Mills BP,
Frampton CM. Bivalirudin versus heparin and protamine in off pump coronary
artery bypass surgery. Ann Thorac Surg 2004. In press.
506 Bartholomew
Karl-Georg Fischer
University Hospital Freiburg, Freiburg, Germany
I. HEPARIN-INDUCED THROMBOCYTOPENIA
IN HEMODIALYSIS PATIENTS
B. Unsuitable Approaches
Low Molecular Weight Heparin
LMWH is not recommended as an alternative anticoagulant. In vitro tests for
HIT antibodies show a high degree of cross-reactivity between UFH and
LMWH (Greinacher et al., 1992b; Vun et al., 1996). Furthermore, in vivo
cross-reactivity manifesting as persistent or recurrent thrombocytopenia or
thrombosis during LMWH treatment of HIT appears to be common (Grei-
nacher et al., 1992a; Horellou et al., 1984; Roussi et al., 1984). Because non-
heparin anticoagulants are available, LMWH should not be used even if in
vitro cross-reactivity is reported to be negative.
Regional Heparinization
Regional heparinization is defined as application of heparin at the inlet of
the extracorporeal circuit and its neutralization by protamine at the outlet
of the circuit. However, its use in HIT is problematic because of the potential
for heparin ‘‘contamination’’ of the patient, as well as for heparin ‘‘rebound
anticoagulation’’ (recurrence of heparin anticoagulation owing to shorter
half-life of protamine compared with heparin) (Blaufox et al., 1966). More-
over, direct injurious effects of protamine on the clotting cascade can occur.
Consequently, this regimen is not recommended for HD of patients with HIT.
Aspirin
Acetylsalicylic acid has been used as an antiplatelet agent together with con-
tinued anticoagulation with UFH for HD of patients with HIT (Hall et al.,
1992; Janson et al., 1983; Matsuo et al., 1989). This approach is not recom-
mended for several reasons: (1) protection against heparin-induced platelet
activation may be incomplete or absent, as aspirin’s effects on blocking the
thromboxane-dependent pathway of platelet activation does not reliably in-
hibit platelet activation by HIT antibodies (Kappa et al., 1987; Polgár et al.,
1998); (2) the bleeding risk of uremic patients is increased; and (3) theoret-
ically, it may lead to induction of persistently high levels of HIT antibodies.
Danaparoid Sodium
Danaparoid sodium (Orgaran, formerly known as Org 10172) is the alterna-
tive anticoagulant that has been most widely used for management of HD in
patients with HIT (Chong and Magnani, 1992; Greinacher et al., 1992a, 1993;
Henny et al., 1983; Magnani, 1993; Neuhaus et al., 2000; Ortel et al., 1992;
Roe et al., 1998; Tholl et al., 1997; Wilde and Markham, 1997). However,
danaparoid has been withdrawn from certain markets, such as the United
States and the United Kingdom (see Chap. 14). Some of its characteristics
require specific attention:
1. The anticoagulant activity of danaparoid can be monitored only by
measurement of antifactor Xa levels based on a danaparoid calibration curve;
however, many laboratories do not routinely perform these assays. Except for
an emergency situation, such as when HIT is strongly suspected and danapa-
roid is the only available alternative, HD should not be performed without
monitoring the antifactor Xa activity to evaluate the dose required for ade-
quate anticoagulation. Once the optimal dose is identified, it can often be used
without alteration for several subsequent HD sessions, provided no bleeding
or inappropriate clotting occurs and no surgical intervention is scheduled.
Periodic measurement of antifactor Xa activity to validate the appropriate
514 Fischer
Table 1 Alternative Anticoagulation for Hemodialysis and Hemofiltration of Patients with HIT
Continuous Monitoring Target
Agent Dialysis Procedure Bolus infusion parametera range
Danaparoid Intermittent HD Before first 2 HDs 3750 (2500)b,c — Anti-Xa activity 0.5–0.8d,e
sodium (Orgaran) (every 2nd day) Subsequent HD Predialytic anti-Xa
activity
(U/mL)d,f
<0.3 3000 (2000)b,c —
0.3–0.35 2500 (1500)b,c —
0.35–0.4 2000 (1500)b,c —
>0.4 0g —
Intermittent HD 1st HD 3750 (2500)b,c — Anti-Xa activity 0.5–0.8d,e
(daily) 2nd HD 2500 (2000)b,c —
Subsequent HD See above —
Continuous HD/HF Initial bolus 2500 (2000)b,c
First 4 h — 600 (600)c,h Anti-Xa activity 0.5–1.0d
Next 4 h — 400 (400)c,h
Subsequently — 200–600g,h,i
(150–400)c,g,h,i
Lepirudin Intermittent HD 0.08–0.15j,k,l — aPTT ratiom,n 2–2.5e,o
(Refludan) (every 2nd day) Hirudin conc.p 0.5–0.8e,q
Continuous HDr,s Initial bolus 0.01j,k,l,t — aPTT ratiom,n 1.5–2.0u,v
Subsequent boluses 0.005–0.01j,k,l,t —
Alternatively — 0.005–0.01t,w,x
Argatroban Intermittent HDr 0.1j 0.1–0.2x aPTT ratiom 1.5–3.0o
(Novastany) (every 2nd day)
Many of the approaches listed have not been formally studied, and none is approved. Treatment examples are given based on a limited number of cases successfully treated with
the respective regimen. The different anticoagulants thus cannot be uncritically applied in the dosages given here. The choice of anticoagulant should depend on the experience
Fischer
of the center and the anticoagulant monitoring available. Doses for danaparoid as given by the manufacturer.
Abbreviations: HD, hemodialysis; HF, hemofiltration; conc., concentration; Xa, factor Xa; aPTT, activated partial thromboplastin time.
a
Monitoring the condition of the dialyzer after a HD session as well as the time required for termination of bleeding of the fistula should also be included.
b
Dosage for bolus given in anti-Xa units (U).
c
Dosage in parentheses for patients with body weight <55 kg.
d
Target range given in anti-factor Xa U/mL.
e
Peak activity determined after about 30 min of HD; this level is not required throughout the whole HD session.
f
Determination 30–60 min before start of the respective HD session.
g
If fibrin deposition in the dialyzer or clots in the extracorporeal circuit occur, give bolus of 1500 anti-Xa U.
h
Hemodialysis in HIT
Argatroban
Argatroban (Novastan; MD-805) is a potent arginine-derived, synthetic, cat-
alytic site-directed thrombin inhibitor lacking antiplatelet and antifibrinolytic
activities (Koide et al., 1995; Matsuo et al., 1992). This agent is approved in
the United States and Canada as a treatment for HIT (see Chaps. 1, 13, and
Hemodialysis in HIT 521
16). It does not cross-react with HIT antibodies. Apart from an even better
relative ability to inhibit fibrin-bound versus soluble thrombin (Berry et al.,
1996; Lunven et al., 1996), the principal advantages of argatroban over
heparin are similar to r-hirudin (Markwardt, 1991; Matsuo et al., 1992).
However, argatroban is metabolized primarily by the liver, and its half-life is
only moderately extended in patients with renal insufficiency.
After argatroban proved to be a valuable anticoagulant in HD (Matsuo
et al., 1986), it was applied successfully to HIT patients undergoing this
procedure (Koide et al., 1995; Matsuo et al., 1992). In the studies ARG-911,
ARG-915, and ARG-915X, no differences in the primary efficacy endpoint or
in bleeding were observed in 54 HD patients compared to non-HD patients
being treated with comparable doses of argatroban. A recent prospective
crossover study of 12 maintenance HD patients showed three different arga-
troban dosing regimens (bolus alone, infusion alone, or bolus plus infusion) to
be safe and well tolerated (Murray et al., 2003) (see also Chap. 16). Arga-
troban also proved effective and safe in HD patients with antithrombin
deficiency (Ota et al., 2003). Whether anticoagulation with argatroban alone
is always sufficient to prevent clotting in the extracorporeal circuit is unclear:
in one HD patient treated with argatroban, marked spontaneous platelet
aggregation occurred, perhaps due to HIT together with additional platelet
activation known to occur in HD (Koide et al., 1995). Because platelet aggre-
gation could not be suppressed by argatroban alone in this patient, aspirin was
added to achieve patency of the extracorporeal circuit.
As nonspecific inactivation of argatroban may occur in blood, periodic
monitoring of its anticoagulant activity is recommended (Matsuo et al., 1992);
for example, by measuring the aPTT (Koide et al., 1995; Matsuo et al., 1992)
or the ECT (Berry et al., 1998).
Argatroban appears to be at least as well suited as r-hirudin for anti-
coagulation of HIT patients requiring HD. Its predominant hepatic elimina-
tion theoretically favors argatroban for alternative anticoagulation in renal
failure.
Oral Anticoagulation
For HIT patients requiring long-term anticoagulation, orally active agents
are usually given. Although coumarins interfere with the clotting cascade,
fibrin formation within the extracorporeal circuit is not always sufficiently
blocked. In these cases, additional low-dose intravenous anticoagulation with
UFH is usually given for regular maintenance HD. However, in HIT patients
requiring HD, alternative low-dose anticoagulation has not been formally
studied. The need for additional intravenous anticoagulation depends on the
increase of the INR, which should be checked regularly before HD. Priming
522 Fischer
D. Other Approaches
Dermatan Sulfate
Dermatan sulfate is a natural glycosaminoglycan that selectively inhibits both
soluble and fibrin-bound thrombin through potentiation of endogenous
heparin cofactor II. It does not interfere with platelet function. Dermatan
sulfate has been used successfully to anticoagulate patients with HIT (Agnelli
et al., 1994), and has also been applied successfully as an anticoagulant for
HD (Boccardo et al., 1997).
Nafamostat Mesilate
Nafamostat mesilate (FUT-175), a synthetic nonspecific serine protease
inhibitor with a short half-life, has been evaluated for regional hemodialysis
in patients at risk of bleeding (Akizawa et al., 1993). It has also been applied
occasionally to HIT patients on HD (Koide et al., 1995). However, owing to
significant clot formation at the dialyzer outlet, despite a twofold prolonga-
tion of aPTT, reported both in HIT and non-HIT patients (Koide et al., 1995;
Matsuo et al., 1993; Takahashi et al., 2003), this anticoagulant cannot cur-
rently be recommended for HD of HIT patients.
Prostacyclin
Prostacyclin (PGI2, epoprostenol), a potent antiplatelet agent with a short
half-life, has been evaluated both as a substitute for, and as an adjunct to
standard heparin for HD of patients with acute or chronic renal insufficiency
(Turney et al., 1980; Smith et al., 1982; Samuelsson et al., 1995). Adverse
effects, such as nausea, vomiting, and hypotension, can be avoided by dose
reduction, use of bicarbonate- instead of acetate-containing dialysate, or
infusion of the drug at the inlet of the extracorporeal circuit. Because of its
mode of action, prostacyclin cannot inhibit activation of coagulation during
HD (Rylance et al., 1985; Novacek et al., 1997). Moreover, in a HIT patient
receiving continuous venovenous HD, prostacyclin was unable to suppress
platelet consumption effectively after heparin had been reinstituted, owing to
a false-negative platelet aggregation assay (Samuelsson et al., 1995). Prosta-
cyclin does not seem to be a suitable antithrombotic agent for HD in HIT.
Whether it may be a useful adjunct in selected cases remains to be clarified.
Hemodialysis in HIT 523
IV. SUMMARY
REFERENCES
Beijering RJR, ten Cate H, Nurmohamed MT, ten Cate JW. Anticoagulants and
extracorporeal circuits. Semin Thromb Hemost 1997; 23:225–233.
Benz K, Nauck M, Beck KH, Böhler J, Fischer KG. Filtration characteristics of
recombinant hirudin by different hemodialyzers in vitro [abstr]. Ann Hematol
2000; 79(suppl 1):A26.
Berry CN, Girardot C, Lecoffre C, Lunven C. Effects of the synthetic thrombin
inhibitor argatroban on fibrin- or clot-incorporated thrombin: comparison with
heparin and recombinant hirudin. Thromb Haemost 1996; 72:381–386.
Berry CN, Lunven C, Girardot C, Lechaire I, Girard D, Charles MC, Ferrari P,
O’Brien DP. Ecarin clotting time: a predictive coagulation assay for the anti-
thrombotic activity of argatroban in the rat. Thromb Haemost 1998; 79:228–
233.
Bichler J, Gemmerli R, Fritz H. Studies for revealing a possible sensitization to hirudin
after repeated intravenous injections in baboons. Thromb Res 1991; 61:39–51.
Blaufox MD, Hampers CL, Merrill JP. Rebound anticoagulation occurring after
regional heparinization for hemodialysis. ASAIO Trans 1966; 12:207–209.
Boccardo P, Melacini D, Rota S, Mecca G, Boletta A, Casiraghi F, Gianese F. Indi-
vidualized anticoagulation with dermatan sulphate for haemodialysis in chronic
renal failure. Nephrol Dial Transplant 1997; 12:2349–2354.
Boon DMS, van Vliet HHDM, Zietse R, Kappers-Klunne MC. The presence of anti-
bodies against a PF4–heparin complex in patients on haemodialysis. Thromb
Haemost 1996; 76:480.
Bredlich RO, Stracke S, Gall H, Proebstle TM. Heparin-associated platelet aggrega-
tion syndrome with skin necrosis during haemodialysis. Dtsch Med Woch-
enschr 1997; 122:328–332.
Bucha E, Nowak G, Czerwinski R, Thieler H. r-Hirudin as anticoagulant in regular
hemodialysis therapy: Finding of therapeutic r-hirudin blood/plasma concen-
trations and respective dosages. Clin Appl Thromb Hemost 1999a; 5:164–170.
Bucha E, Kreml R, Nowak G. In vitro study of r-hirudin permeability through mem-
branes of different haemodialyzers. Nephrol Dial Transplant 1999b; 14:2922–
2926.
Chong BH, Magnani HN. Orgaran in heparin-induced thrombocytopenia. Haemo-
stasis 1992; 22:85–91.
Danhof M, De Boer A, Magnani HN, Stiekema JC. Pharmacokinetic considerations
of Orgaran (Org 10172) therapy. Haemostasis 1992; 22:73–84.
De Sancho M, Lema MG, Amiral J, Rand J. Frequency of antibodies directed against
heparin–platelet factor 4 in patients exposed to heparin through chronic hemo-
dialysis [letter]. Thromb Haemost 1996; 75:695–696.
Eichler P, Friesen HJ, Lubenow N, Jaeger B, Greinacher A. Antihirudin antibodies in
patients with heparin-induced thrombocytopenia treated with lepirudin: inci-
dence, effects on aPTT, and clinical relevance. Blood 2000; 96:2373–2378.
Finazzi G, Remuzzi G. Heparin-induced thrombocytopenia—background and impli-
cations for haemodialysis. Nephrol Dial Transplant 1996; 11:2120–2122.
Fischer KG. Hirudin in renal insufficiency. Semin Thromb Hemost 2002; 28:467–482.
Fischer KG, van de Loo A, Böhler J. Recombinant hirudin (lepirudin) as anticoagu-
Hemodialysis in HIT 525
lant in intensive care patients treated with continuous hemodialysis. Kidney Int
1999; 56(suppl 72):S46–S50.
Fischer KG, Weiner SM, Benz K, Nauck M, Böhler J. Treatment of hirudin overdose
with hemofiltration [abstr]. Blood Purif 2000; 18:80–81.
Fischer KG, Liebe V, Hudek R, Piazolo L, Haase KK, Borggrefe M, Huhle G. Anti-
hirudin antibodies alter pharmacokinetics and pharmacodynamics of recombi-
nant hirudin. Thromb Haemost 2003; 89:973–982.
Flanigan MJ, Pillsbury L, Sadewasser G, Lim VS. Regional hemodialysis anti-
coagulation: hypertonic tri-sodium citrate or anticoagulant citrate dextrose-A.
Am J Kidney Dis 1996; 27:519–524.
Frank RD, Farber H, Stefanidis I, Lanzmich R, Kierdorf HP. Hirudin elimination by
hemofiltration: a comparative in vitro study of different membranes. Kidney Int
1999; 56(suppl 72):S41–S45.
Frank RD, Farber H, Lazmich R, Floege J, Kierdorf HP. In vitro studies on hiru-
din elimination by haemofiltration: comparison of three high-flux membranes.
Nephrol Dial Transplant 2002; 17:1957–1963.
Greinacher A, Drost W, Michels I, Leitl J, Gottsmann M, Kohl HJ, Glaser M,
Mueller-Eckhardt C. Heparin-associated thrombocytopenia: successful therapy
with the heparinoid Org 10172 in a patient showing cross-reaction to LMW
heparins. Ann Hematol 1992a; 64:40–42.
Greinacher A, Michels I, Mueller-Eckhardt C. Heparin-associated thrombocytope-
nia: the antibody is not heparin specific. Thromb Haemost 1992b; 67:545–549.
Greinacher A, Philippen KH, Kemkes-Matthes B, Möckl M, Mueller-Eckhardt C,
Schaefer K. Heparin-associated thrombocytopenia type II in a patient with end-
stage renal disease: successful anticoagulation with the low-molecular-weight
heparinoid Org 10172 during haemodialysis. Nephrol Dial Transplant 1993;
8:1176–1177.
Greinacher A, Zinn S, Wizemann U, Birk W. Heparin-induced antibodies as a risk
factor for thromboembolism and haemorrhage in patients undergoing chronic
haemodialysis. Lancet 1996; 348:764.
Greinacher A, Eichler P, Lubenow N, Kwasny H, Luz M. Heparin-induced thrombo-
cytopenia with thromboembolic complications: meta-analysis of 2 prospective
trials to assess the value of parenteral treatment with lepirudin and its thera-
peutic aPTT range. Blood 2000; 96:846–851.
Griessbach U, Sturzebecher J, Markwardt F. Assay of hirudin in plasma using a
chromogenic thrombin substrate. Thromb Res 1985; 37:347–350.
Grötsch H, Hropot M. Degradation of rDNA hirudin and a-human thrombin hirudin
complex in liver and kidney homogenates from rat. Thromb Res 1991; 64:763–
767.
Gupta AK, Kovacs MJ, Sauder DN. Heparin-induced thrombocytopenia. Ann Phar-
macother 1998; 32:55–59.
Haas G. Versuche der Blutauswaschung am Lebenden mit Hilfe der Dialyse. Klin
Wochenschr 1925; 4:13–14.
Hafner G, Peetz D, Klingel R, Prellwitz W. Methods for hirudin determination in
plasma. J Lab Med 2000; 24:172–178.
526 Fischer
Lewis BE, Walenga JM, Wallis DE. Anticoagulation with Novastan (argatroban) in
patients with heparin-induced thrombocytopenia and heparin-induced throm-
bocytopenia and thrombosis syndrome. Semin Thromb Hemost 1997; 23:197–
202.
Liebe V, Piazolo L, Fischer KG, Hudek R, Heene DL, Huhle G. A monoclonal mouse
anti-r-hirudin antibody neutralizes r-hirudin in vivo—potential use as antidote
[abstr]. Ann Hematol 2001; 80(suppl 1):A42.
Lindhoff-Last E, Betz C, Bauersachs R. Use of a low-molecular-weight heparinoid
(danaparoid sodium) for continuous renal replacement therapy in intensive care
patients. Clin Appl Thromb Hemost 2001; 7:300–304.
Lubenow N, Greinacher A. Heparin-induced thrombocytopenia. Recommendations
for optimal use of recombinant hirudin. Biodrugs 2000; 14:109–125.
Lunven C, Gauffeny C, Lecoffre C, O’Brien DP, Roome NO, Berry CN. Inhibition by
argatroban, a specific thrombin inhibitor, of platelet activation by fibrin clot-
associated thrombin. Thromb Haemost 1996; 75:154–160.
Luzzatto G, Bertoli M, Cella G, Fabris F, Zaia B, Girolami A. Platelet count, anti-
heparin/platelet factor 4 antibodies and tissue factor pathway inhibitor plasma
antigen level in chronic dialysis. Thromb Res 1998; 89:115–122.
Magnani HN. Heparin-induced thrombocytopenia (HIT): an overview of 230 patients
treated with Orgaran (Org 10172). Thromb Haemost 1993; 70:554–561.
Markwardt F. Past, present and future of hirudin. Haemostasis 1991; 21:11–26.
Matsuo T, Nakao K, Yamada T, Matsuo O. Effect of a new anticoagulant (MD 805)
on platelet activation in the hemodialysis circuit. Thromb Res 1986; 41:33–41.
Matsuo T, Yamada T, Chikahira Y, Kadowaki S. Effect of aspirin on heparin-induced
thrombocytopenia (HIT) in a patient requiring hemodialysis. Blut 1989; 59:393–
395.
Matsuo T, Kario K, Kodama K, Okamoto S. Clinical application of the synthetic
thrombin inhibitor, argatroban (MD-805). Semin Thromb Hemost 1992; 18:
155–160.
Matsuo T, Kario K, Nakao K, Yamada T, Matsuo M. Anticoagulation with nafa-
mostat mesilate, a synthetic protease inhibitor, in hemodialysis patients with a
bleeding risk. Haemostasis 1993; 23:135–141.
Matsuo T, Koide M, Kario K. Application of argatroban, a direct thrombin inhibitor,
in heparin-intolerant patients requiring extracorporeal circulation. Artif Organs
1997; 21:1035–1038.
Moberg PQ, Geary VM, Sheikh FM. Heparin-induced thrombocytopenia: a possible
complication of heparin-coated pulmonary artery catheters. J Cardiothorac
Anesth 1990; 4:226–228.
Müller A, Huhle G, Nowack R, Birck R, Heene DL, van der Woude FJ. Serious
bleeding in a haemodialysis patient treated with recombinant hirudin. Nephrol
Dial Transplant 1999; 14:2482–2483.
Murray P, Reddy B, Grossman E, Hammes M, Trevino S, Ferrell J, Tang I, Khorana
A, Fosbinder T, Swan SK. Safety and tolerability of argatroban anticoagulation
in patients with end-stage renal disease undergoing hemodialysis: a prospective
study [abstr]. J Thromb Haemost 2003; 1(suppl 1):P1911.
528 Fischer
heparin sparing with prostacyclin during regular dialysis therapy. Lancet 1980;
2:219–222.
Unver B, Sunder-Plassmann G, Hörl WH, Apsner R. Long-term citrate anticoagu-
lation for high-flux haemodialysis in a patient with heparin-induced thrombo-
cytopenia type II. Acta Med Austriaca 2002; 29:146–148.
Vanholder RC, Camez AA, Veys NM, Soria J, Mirshahi M, Soria C, Ringoir S.
Recombinant hirudin: a specific thrombin inhibiting anticoagulant for hemo-
dialysis. Kidney Int 1994; 45:1754–1759.
Vanholder R, Camez A, Veys N, van Loo A, Dhondt AM, Ringoir S. Phar-
macokinetics of recombinant hirudin in hemodialyzed end-stage renal failure
patients. Thromb Haemost 1997; 77:650–655.
Van Wyk V, Badenhorst PN, Luus HG, Kotzé HF. A comparison between the use of
recombinant hirudin and heparin during hemodialysis. Kidney Int 1995;
48:1338–1343.
Vargas Hein O, von Heymann C, Lipps M, Ziemer S, Ronco C, Neumayer HH,
Morgera S, Welte M, Kox WJ, Spies C. Hirudin versus heparin for antico-
agulation in continuous renal replacement therapy. Intensive Care Med 2001;
27:673–679.
Vecino A, Navarro-Antolin J, Teruél J, Navarro J, Cesar J. Lipid composition of
platelets in patients with uremia. Nephron 1998; 78:271–273.
Vun CM, Evans S, Chong BH. Cross-reactivity study of low molecular weight hepa-
rins and heparinoid in heparin-induced thrombocytopenia. Thromb Res 1996;
81:525–532.
Ward DM, Mehta RL. Extracorporeal management of acute renal failure patients at
high risk of bleeding. Kidney Int 1993; 43:S-237–S-244.
Warkentin TE. Danaparoid (Orgaran) for the treatment of heparin-induced throm-
bocytopenia (HIT) and thrombosis: effects on in vivo thrombin and cross-linked
fibrin generation, and evaluation of the clinical significance of in vitro cross-
reactivity (XR) of danaparoid for HIT-IgG [abstr]. Blood 1996; 88:626a.
Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia.
Am J Med 1996; 101:502–507.
Warkentin TE, Chong BH, Greinacher A. Heparin-induced thrombocytopenia:
towards consensus. Thromb Haemost 1998; 79:1–7.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular-
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Wilde MI, Markham A. Danaparoid. A review of its pharmacology and clinical use in
the management of heparin-induced thrombocytopenia. Drugs 1997; 54:903–
924.
Willey ML, de Denus S, Spinler SA. Removal of lepirudin, a recombinant hirudin, by
hemodialysis, hemofiltration, or plasmapheresis. Pharmacotherapy 2002; 22:
492–499.
Yamamoto S, Koide M, Matsuo M, Suzuki S, Ohtaka M, Saika S, Matsuo T.
Heparin-induced thrombocytopenia in hemodialysis patients. Am J Kidney Dis
1996; 28:82–85.
19
Management of Cardiopulmonary
Bypass Anticoagulation in Patients
with Heparin-Induced
Thrombocytopenia
Bernd Poetzsch
Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
Katharina Madlener
Kerckhoff-Klinik, Bad Nauheim, Germany
I. INTRODUCTION
Immediate cessation of, and avoidance of reexposure to, heparin are impor-
tant principles underlying the management of patients with immune-mediated
heparin-induced thrombocytopenia (HIT) (Chong and Berndt, 1989; War-
kentin et al., 1998). Because further antithrombotic therapy is often necessary
for these patients, several alternative anticoagulant strategies have been
developed (see Chaps. 13–17). However, patients with HIT who require car-
diac surgery present special problems. Considerable activation of the hemo-
static system results when blood is exposed to the artificial surfaces of the
cardiopulmonary bypass (CPB) pump used for most heart surgery, making
high-dose anticoagulation mandatory (Edmunds, 1993; Slaughter et al.,
1994). Heparin is the current anticoagulant of choice for CPB, and there is
relatively little experience with other forms of anticoagulation in this patient
setting. Moreover, any alternative anticoagulant considered for HIT patients
should ideally meet certain requirements. First, the agent should be effective
in minimizing activation of coagulation during CPB. Second, a rapid and
simple method of monitoring its anticoagulating effects should be available to
531
532 Poetzsch and Madlener
A. Danaparoid Sodium
The efficacy of danaparoid sodium for CPB anticoagulation was first shown in
a dog model (Henny et al., 1985). Subsequently, this agent was used for pa-
tients with HIT who needed heart surgery (Doherty et al., 1990; Magnani,
1993; Wilhelm et al., 1996). In a retrospective analysis Magnani and co-
workers (1997) summarized the experience in 53 patients with HIT who
underwent CPB using danaparoid for anticoagulation. The patients included
in this study generally received an intravenous (iv) bolus of 8750 antifactor Xa
(anti-Xa) units (U) of danaparoid after thoracotomy. The CPB circuit was
primed with 7500 U. During CPB, booster iv injections (1500 U) were to be
administered up to once hourly if there was visually apparent clot or fibrin
formation. Plasma levels of anti-Xa activity generally were not monitored
during CPB.
With this fixed-dosing schedule, ‘‘clots’’ in the operative field, as an
indicator of inadequate anticoagulation, were observed in 18 (34%) patients.
One patient, reported elsewhere, developed near-fatal thrombosis of the CPB
circuit during weaning from bypass (Grocott et al., 1997). Severe postoper-
ative bleeding, defined as more than 20 U of blood transfused, was noted
in 11 (20%) patients. As a result of these data, the authors recommended
a modified treatment regimen that included a priming dose of 3 U/mL, a
weight-adjusted postthoracotomy iv bolus dose of 125 U/kg body weight
(b.w.), and a constant iv infusion of 7 U/kg b.w. per hour started immediately
CPB Anticoagulation in HIT 533
after institution of the CPB, and stopped 45 min before the expected end of
CPB. Thus, for a 70-kg person undergoing an operation with a CPB time of
2 h and a priming volume of 1500 mL, a total dose of approximately 13,860 U
danaparoid, or 198 U/kg, is recommended by the authors.
However, this revised protocol was developed empirically, with adjust-
ments made based on some of the complications observed using the fixed-dose
protocol (Grocott et al., 1997). Even though the revised protocol means that
many patients would receive a lower dose of danaparoid than with the earlier
fixed-dose regimen, this might not lead to reduced bleeding outcomes.
Paradoxically, less effective anticoagulation during CPB could lead to more
thrombin generation during the procedure potentially leading to even greater
postoperative bleeding because of secondary hyperfibrinolysis, even if the
postoperative danaparoid levels are not high. Indeed, Insler and colleagues
(1997) reported a patient receiving danaparoid for CPB who first developed
clots in the operative field and arterial filter of the CPB, followed by severe
postoperative bleeding requiring surgical reexploration. Regardless of the
explanation for excessive bleeding, even a weight-modified treatment regimen
bears the risk of under- or overanticoagulation, if the anticoagulant effect is
not monitored.
Therefore, we developed a danaparoid-dosing schedule for CPB with
dose adjustments made according to the results of anti-Xa measurements
(Table 1). Unfortunately, although both the activated clotting time (ACT)
and the activated partial thromboplastin time (aPTT) are prolonged by the
higher plasma levels of danaparoid used during CPB, there is no acceptable
linear correlation (Gitlin et al., 1998). Because only the plasma anti-Xa levels
correlate linearly with the plasma levels of danaparoid, we based our schedule
on anti-Xa levels. Similar to the protocol recommended by Magnani and
coworkers (1997), the iv bolus and priming dose are adjusted to body weight
and priming volume, respectively. After beginning CPB, plasma anti-Xa
levels should be maintained at 1.5 F 0.3 U/mL. The continuous infusion is
stopped 30 min before the expected end of bypass. However, in our experi-
ence, even if such an anti-Xa–adjusted danaparoid treatment regimen is used,
increased postoperative bleeding is a problem. Possible explanations for the
increased postoperative bleeding include an ongoing anticoagulant effect of
danaparoid (half-life, approximately 17 h) for which there is no pharmaco-
logical antagonist (Meuleman, 1992), as well as the incomplete inhibition of
thrombin generation during CPB, potentially leading to increased postoper-
ative hyperfibrinolysis.
In all, CPB anticoagulation with danaparoid can lead to successful
outcomes. About three quarters (36 of 47; 77%) of the patients reported by
Magnani and coworkers (1997) were alive 6 weeks after cardiac surgery
with danaparoid. Nevertheless, the disadvantages of danaparoid, including
534 Poetzsch and Madlener
B. Recombinant Hirudin
Recombinant hirudin (r-hirudin), an anticoagulant naturally produced by the
salivary gland of the leech (Hirudo medicinalis), is now approved in most
countries for clinical use by the iv route. Hirudin is a single-chain polypeptide
of 65 amino acids (7000 Da) that forms a tight 1:1 stoichiometric complex with
thrombin, thereby occupying the putative fibrinogen-binding site and block-
ing the catalytic site of thrombin. As a result, all of the thrombin-catalyzed
CPB Anticoagulation in HIT 535
Argatroban
Argatroban is a specific thrombin inhibitor derived from L-arginine. It is a
small molecule (532 Da) that binds reversibly to thrombin. It has a half-life of
about 40–50 min in normal humans. The potential of argatroban to be an
effective anticoagulant in patients with HIT has been documented by the
studies of Lewis and coworkers (1997a,b, 2001, 2003) (see Chap. 16).
Although argatroban has been a successful anticoagulant in a CPB model,
only limited information is available on its use in humans for this purpose.
Furukawa and coworkers report a patient who successfully underwent CPB
anticoagulation with argatroban (Furukawa et al., 2001). Argatroban was
administered as a bolus injection of 0.1 mg/kg followed by a continuous
infusion at 5–10 Ag/kg/min. The ACT was used for monitoring.
the fibrinogen level at the recommended target fibrinogen range (0.2–0.5 g/L).
Furthermore, it is uncertain what fibrinogen level is required, if any, to
prevent clinically important fibrin formation during CPB. Third, reversal of
‘‘anticoagulation’’ requires a blood product, fibrinogen concentrates
(Europe) or cryoprecipitate (North America), to replace fibrinogen. Finally,
ancrod does not inhibit thrombin generation and has even been associated
with increased thrombin generation in some clinical settings, such as acute
HIT (Warkentin, 1998). It is possible that this could lead to thrombotic or
post-CPB hemorrhagic complications when used for the management of
acute HIT. All of the considerations suggest that ancrod is not a suitable
alternative to heparin in the setting of CPB surgery.
An intriguing option for patients with a history of HIT, but in whom per-
sisting HIT antibodies can no longer be detected, is to consider reexposure to
heparin for CPB, and to avoid heparin completely both before surgery (e.g., at
heart catheterization) and in the postoperative period. This approach has
been used successfully by some physicians (Makhoul et al., 1987; Pötzsch et
al., 2000; Selleng et al., 2001; Warkentin and Kelton, 2001), and it is based on
the following rationale. First, HIT antibodies are transient, and they usually
are not detectable after 100 days following an episode of HIT (see Chap. 3).
Thus, no immediate problems would be expected in a patient without residual
HIT antibodies whose previous episode of HIT was ‘‘remote’’ (i.e., more than
several months before the need for heart surgery). Second, it appears that a
minimum of 5 days are required before clinically significant levels of HIT
antibodies are generated following any episode of heparin treatment (War-
kentin and Kelton, 2001). In the event that a recurrent immune response to
platelet factor 4–heparin is induced by reexposure to heparin during CPB, it is
unlikely that the newly generated HIT antibodies will contact exogenously
administered heparin. As a consequence, platelet activation by HIT anti-
bodies should not occur, and thus the thrombotic risk should not be increased.
We reported 10 patients with a documented history of HIT, but no detectable
HIT antibodies at the time of the proposed surgery, who thus underwent CPB
anticoagulation with heparin (Pötzsch et al., 2000). In none of the 10 patients
was a thromboembolic complication or prolonged thrombocytopenia ob-
served. Further, no increase in HIT antibody concentrations occurred during
a 10-day follow-up period. These data are in contrast to reports of a rapid
‘‘anamnestic’’ immune response in HIT. However, there is evidence that these
episodes represent acute-onset HIT in a patient who has residual circulating
544 Poetzsch and Madlener
HIT antibodies resulting from a recent episode of HIT, rather than a rapid
recurrence of HIT antibodies because of immune memory caused by a remote
exposure to heparin (Warkentin and Kelton, 2001).
As outlined in Fig. 2, we recommend that HIT antibody–negative
patients with a history of HIT who require CPB for heart surgery should be
treated according to established heparin protocols. The use of heparin should
be restricted to the operative period itself; if necessary, postoperative anti-
coagulation should be achieved with an alternative anticoagulant (see Chaps.
13–16).
Testing for HIT antibodies in patients with a history of HIT before
anticipated heparin reexposure at heart surgery should be performed using
one or more sensitive tests (see Chap. 11) if this approach is to be considered.
Particularly in cardiac surgical centers where there is limited experience with
nonheparin anticoagulation for CPB, risk-benefit considerations favor a brief
use of heparin for these patients. For example, a patient who developed near-
fatal CPB circuit thrombosis during danaparoid anticoagulation had had
HIT 11 years earlier and had no detectable HIT antibodies at the time
danaparoid was used (Grocott et al., 1997).
There are several reasons why a patient with HIT might require urgent heart
surgery, including the result of life-threatening thrombotic complications of
HIT affecting the heart (e.g., acute coronary insufficiency or myocardial
infarction; removal of intracardiac thrombus), or because HIT has compli-
cated the course of a critically ill patient receiving heparin before anticipated
heart surgery (e.g., while awaiting a heart for cardiac transplantation, or
during use of an intra-aortic balloon pump). The latter group of patients
appear to have a relatively high risk of developing HIT (Walls et al., 1992). A
suitable alternative for patients who require urgent coronary revasculariza-
tion during acute HIT might be the use of an off-pump (‘‘beating-heart’’)
strategy. This surgical technique requires lower levels of anticoagulation.
Therefore, the dose of the nonheparin anticoagulant could be markedly
reduced, which could reduce postoperative bleeding. Such an approach using
danaparoid as heparin substitute was recently reported by Warkentin and
colleagues (2001). The authors found a minimum plasma anti-Xa level of 0.6
U/mL sufficient to perform surgery off-pump.
Studies of the frequency of HIT antibody formation (Visentin et al.,
1996; Bauer et al., 1997; Warkentin et al., 2000) following heart surgery
suggest that as many as 15–50% of patients form HIT antibodies, using an
enzyme immunoassay that detects IgG antibodies that recognize platelet
factor 4–heparin complexes (see Chap. 4). With the washed platelet serotonin-
release assay, HIT antibodies are detected in 13–20% of patients (Bauer et al.,
1997; Warkentin et al., 2000). However, despite this high rate of serocon-
version, only about 1–3% of patients who receive further postoperative
anticoagulation with unfractionated heparin develop HIT. Currently, there
is no convincing evidence that patients who form HIT antibodies in the
absence of thrombocytopenia are at increased risk for thrombosis (Bauer
et al., 1997; Trossaërt et al., 1998; Warkentin et al., 2000). However,
postoperative cardiac surgical patients who develop clinical HIT appear to
be at increased risk for both venous and arterial thrombotic events (Walls
et al., 1990; van Dyck et al., 1996; Pouplard et al., 1999).
Given these data and clinical experience, an algorithm has been developed to
assist in determining the need for alternative anticoagulation for CPB in HIT
patients (Fig. 2). After the decision to avoid use of heparin in the CPB setting,
the important remaining question is, which strategy should be chosen? Be-
cause each of the different approaches described here provides specific
546 Poetzsch and Madlener
REFERENCES
Bauer TL, Arepally G, Konkle BA, Mestichelli B, Shapiro SS, Cines DB, Poncz M,
McNulty S, Amiral J, Hauck WW, Edie RN, Mannion JD. Prevalence of
heparin-associated antibodies without thrombosis in patients undergoing
cardiopulmonary bypass surgery. Circulation 1997; 95:1242–1246.
Chong BH, Berndt MC. Heparin-induced thrombocytopenia. Blut 1989; 58:53–57.
Cole CW, Bormanis J. Ancrod: a practical alternative to heparin. J Vasc Surg 1988;
8:59–63.
Davis Z, Anderson R, Short D, Garber D, Valgiusti A. Favorable outcome with
bivalirudin anticoagulation during cardiopulmonary bypass. Ann Thorac Surg
2003; 75:264–265.
Demers C, Ginsberg JS, Brill-Edwards P, Panju A, Warkentin TE, Anderson DR,
Turner C, Kelton JG. Rapid anticoagulation using ancrod for heparin-induced
thrombocytopenia. Blood 1991; 78:2194–2197.
Doherty DC, Ortel TL, De Bruijn N, Greenberg CS, Van Trigt P III. ‘‘Heparin-free’’
cardiopulmonary bypass: first reported use of heparinoid (Org 10172) to pro-
vide anticoagulation for cardiopulmonary bypass. Anesthesiology 1990; 73:
562–565.
Edmunds LH Jr. Blood-surface interactions during cardiopulmonary bypass. J Car-
diovasc Surg 1993; 8:404–410.
Furukawa K, Ohteki H, Hirahara K, Narita Y, Koga S. The use of argatroban as an
anticoagulant for cardiopulmonary bypass in cardiac operations. J Thorac
Cardiovasc Surg 2001; 122:1255–1257.
Gitlin SD, Deeb GM, Yann C, Schmaier AH. Intraoperative monitoring of dan-
aparoid sodium anticoagulation during cardiovascular operations. J Vasc Surg
1998; 27:568–575.
Grocott HP, Root J, Berkowitz SD, deBruijn N, Landolfo K. Coagulation compli-
cating cardiopulmonary bypass in a patient with heparin-induced thrombo-
cytopenia receiving the heparinoid, danaparoid sodium. J Cardiothorac Vasc
Anesth 1997; 11:875–877.
Henny CP, ten Cate H, ten Cate JW, Moulijn AC, Sie TH, Warren P, Büler HR. A
randomized blind study comparing standard heparin and a new low molecular
weight heparinoid in cardiopulmonary bypass in dogs. J Lab Clin Med 1985;
106:187–196.
Insler SR, Kraenzler EJ, Bartholomew JR, Kottke-Marchant K, Lytle B, Starr NJ.
Thrombosis during use of the heparinoid Organon 10172 in a patient with
heparin-induced thrombocytopenia. Anesthesiology 1997; 86:495–498.
Kappa JR, Horn D, McIntosh CL, Fisher CA, Ellison N, Addonizio VP. Iloprost
(ZK36374), a new prostacyclin analogue, permits open cardiac surgery in pa-
tients with heparin-induced thrombocytopenia. Surg Forum 1985; 36:285–286.
Kappa JR, Cottrell ED, Berkowitz HD, Fisher CA, Sobel M, Ellison N, Addonizio
VP Jr. Carotid endarterectomy in patients with heparin-induced platelet acti-
vation: comparative efficacy of aspirin and iloprost (ZK36374). J Vasc Surg
1987; 5:693–701.
Koster A, Hansen R, Grauhan O, Hausmann H, Bauer M, Hetzer R, Kuppe H,
Mertzlufft F. Hirudin monitoring using the TAS ecarin clotting time in patients
548 Poetzsch and Madlener
I. INTRODUCTION
II. PATHOPHYSIOLOGY
III. FREQUENCY
Only four studies have addressed the frequency of HIT in children. Spadone
and coworkers (1992) collected cases of suspected HIT in a neonatal intensive
care unit (ICU) between 1988 and 1990. Of 1329 newborns enrolled, about
70% received unfractionated heparin (UFH): either 0.5–1.0 IU/mL added to
553
554 Klenner and Greinacher
Venous thrombosis
Iliac vein 16.2 21, 24, 27, 32, 38, 39, 42,
43, 48, 53, 60
Femoral vein 13.2 21, 27, 34, 35, 36, 43, 48,
54, 58
Inferior vena cava 11.8 9, 24, 35, 38, 43, 48, 51, 58
Pulmonary embolism 10.3 6, 32, 37, 49, 51, 65, 66
Progression of deep-vein 5.9 34, 40, 42, 44
thrombosis
Subclavian vein 5.9 17, 36, 38, 55
Calf vein 5.9 32, 35, 36, 48
Superior vena cava 5.9 12, 13, 54, 57
Jugular vein 2.9 62, 64
Rare: renal vein, arm veins, <2 9, 10, 7, 45, 44
intracranial veins, pulmonary
vein, shunt
Arterial thrombosis
Femoral artery 4.4 11, 23, 59
Iliac artery 2.9 2, 16
Foot arteries 2.9 23, 30
Rare: renal artery <2 54
Others
Intracardiac thrombi 5.9 4, 19, 25, 37
Bleeding 5.9 21, 40, 47, 63
Clots in dialyzer 4.4 6, 65, 66
Neurological deficits 2.9 36, 41
Rare: subdural hematoma, <2 61
thrombosis of pulmonary valve
a
Patients may have had more than one complication. Case numbers refer to Table 2.
A: Confirmed HIT (clinical criteria present, laboratory test positive); all children received UFH except
case 45
1 M, nb Preterm, sepsis <100 TP
2 F, nb Heart surgery 191 Iliac artery
3 M, nb Norwood I, hypoplastic heart 36 TP
4 M, nb ECMO <1 TP, ventricular thrombus
5 F, nb Tetralogy of Fallot, heart 45 TP
surgery
6 F, nb Aortic stenosis, hypoplastic NA TP, clots in circuit, PE, lung
left ventricle, heart surgery hemorrhage, renal vein
7 F, nb CHD, heart surgery 48 TP, clot in shunt
8 F, 2m Tetralogy of Fallot, premature, TP
heart surgery, NEC
9 M, 3m Tricuspid valve atresia, 34 Vena cava, renal vein
Blalock-Taussig shunt
10 M, 5m Hypoplastic left heart, heart TP, left pulmonary vein
surgery, ECMO
11 F, 6m Heart surgery 46 Fem artery, DVT, TP
12 M, 8m Tetralogy of Fallot, heart surgery TP, clot in superior vena cava
13 M, 10m Heart surgery 46 TP, vena cava
14 M, 10m Preterm, VACTERL-syndrome 45 TP
15 M, 12m Renal failure, tetralogy of NA TP
Fallot
16 M, 13m CHD, renal agenesis, heart 46 TP, iliac artery
surgery
17 F, 15m Heart surgery 123 TP, SC
18 M, 15m CHD, heart surgery 10 TP
19 F, 17m Acute myocarditis 80 TP, intracardiac
20 M, 23m Hemofiltration NA TP
21 F, 2y Fontan operation 55 TP, DVT
22 M, 2y Fontan operation 73 TP, heparin resistance
23 M, 3y Tricuspid, pulmonary valve 40 Fem artery, foot gangrene
atresia, Fontan operation
24 M, 4y Double-inlet left ventricle, 25 TP, vena cava, DVT
Fontan operation
25 F, 4y Cardiomyopathy, heart 16 TP, intracardiac, bleeding
transplant
26 F, 4y Lung disease, mechanical 50 TP
ventilation
27 F, 7y Cardiomyopathy 71 TP, DVT
28 F, 8y Turner’s syndrome, sinus vein 190 TP, heparin resistance
thrombosis
29 F, 9y DVT, APS 82 TP
30 M, 10y Leg artery thrombosis, PS def 39 TP, foot arteries
31 M, 11y APS 52 TP
Heparin-Induced Thrombocytopenia in Children 557
Table 2 Continued
Gender, Platelet nadir Thrombocytopenia (TP) and
No. age Diagnosis, procedure ( 109/L) thrombotic complication(s)
Table 2 Continued
Gender, Platelet nadir Thrombocytopenia (TP) and
No. age Diagnosis, procedure ( 109/L) thrombotic complication(s)
Abbr: y, died; Amp, amputation; APS, antiphospholipid syndrome; Arg, argatroban; ASA, aspirin
(acetylsalicylic acid); CHD, congenital heart disease; DS, danaparoid sodium; DVT, deep-vein throm-
bosis; ECMO, extracorporeal membrane oxygenation; EIA, PF4-dependent enzyme immunoassay; F,
female; fem, femoral; HIPA, heparin-induced platelet activation test; Lep, lepirudin; +Lep, switch to
lepirudin after suspected or confirmed cross-reactivity to danaparoid; LMWH, low molecular weight hep-
arin; m, month; M, male; MTHFR, methylene tetrahydrofolate reductase; NA, not available; nb, new-
born; NEC, necrotizing enterocolitis; OAC, oral anticoagulants; PE, pulmonary embolism; PS def, protein
S deficiency; r-tPA, recombinant tissue-plasminogen activator; S, survived; SC, subclavian; TP, throm-
bocytopenia; UAC, umbilical artery catheter; UFH, unfractionated heparin; UK, urokinase; XR,
confirmed cross-reactivity with danaparoid; XR?, suspected cross-reactivity with danaparoid; y, year.
Four patients developed HIT during low-dose UFH given for catheter pat-
ency (cases 1, 14, 47, 52). Hemodialysis or hemofiltration accounted for
UFH use in four (5.9%) patients (cases 20, 33, 65, 66). In 17 of the 68 pa-
tients, the laboratory test for HIT was negative or not performed (cases 53–68,
inclusive).
The most frequent manifestation of HIT in the 68 children was a
decrease in platelet count (85.3%). HIT was associated with thromboembolic
complications in about two thirds of the patients, most commonly involving
iliac and femoral veins, the inferior vena cava, and pulmonary embolism
(Table 1). Less commonly, intracardiac thrombi or neurological events oc-
curred, or clotting of the dialyzer. Only about 10% (7/68) of patients devel-
oped arterial thrombosis. Thus, there is a strong preponderance of venous
thrombosis in pediatric HIT.
Eight (11.8%) of the 68 children died (cases 4, 6, 7, 24, 25, 52, 57, 61) and
three required amputations (cases 23, 30, 35). In four children, only partial
recanalization of thrombosed veins occurred (cases 29, 34, 36, 43).
This summary does not include the 14 newborns reported by Spadone
and colleagues (1992). These workers primarily observed arterial thrombosis,
with at least 11 (78.6%) developing aortic thrombosis (one infant died with-
out imaging studies). Two newborns with thrombosis had normal platelet
counts. Eleven (78.6%) survived, the remaining three developing mesenteric
ischemia. Arterial thrombosis likely was related to umbilical artery catheters
Heparin-Induced Thrombocytopenia in Children 561
Figure 1 Reasons for preceding heparin therapy in children with HIT. Among the
various age groups, the reasons for heparin therapy that led to HIT varied consid-
erably: whereas newborns and infants usually developed HIT after cardiac surgery,
among teenagers HIT more often complicated the use of heparin during treatment of
thrombosis.
562 Klenner and Greinacher
(used in all but one of the 14 neonates). In adults, intravascular catheters are
a risk factor for HIT-associated thrombosis (Hong et al., 2003), but whether
the arterial thrombi observed by Spadone et al. indeed were HIT-related is
unclear.
V. LABORATORY TESTING
2003b). None of the infants developed HIT antibodies using the adult cutoff
[UFH group: mean optical density (OD), 0.020; maximum, 0.328; saline
group: mean OD, 0.019, maximum, 0.239]. Minor changes in OD (increase >
0.100) occurred in six patients (three in each group) (Fig. 2). Therefore, these
minor increases in OD are unlikely to be related to UFH use and could rep-
resent a nonspecific increase in antibody levels in ill patients (acute phase reac-
tion). Among the subjects receiving placebo, all OD values were below 0.400
(the accepted adult cutoff value), suggesting that this level is also appropriate
for neonates.
The limitations of antigen assays observed in adults likely also apply to
children. Thus, in 5–10% of cases, the antigen assay could be false-negative
Figure 2 Six neonates with rising absorbance levels in platelet factor 4–dependent
enzyme immunoassay (EIA). Six of 213 neonates participating in a randomized, dou-
ble-blind trial comparing heparin with normal saline for maintenance of peripheral
venous catheter patency developed a rise in absorbance of more than 0.100 optical
density (OD) units by PF4/polyvinyl sulfonate EIA (GTI, Brookfield, WI). All OD
values were less than the positive adult cutoff (0.400 OD units). No differences were
observed between patients receiving heparin (solid lines) compared to patients re-
ceiving saline (dotted lines). As the maximum OD in saline controls was 0.239, the
0.400 cutoff seems appropriate also for pediatric patients. (Klenner et al., 2003b.)
564 Klenner and Greinacher
A. Danaparoid
Danaparoid (see Chap. 14) is a mixture of low molecular weight glycos-
aminoglycans that catalyze the inactivation of factor Xa (FXa) by anti-
thrombin. It has minimal anti-factor IIa activity. Dosing schedules for adults
(appropriately weight-adjusted for the child) can be used for guidance. For
antithrombotic prophylaxis, 10 IU/kg body weight given twice daily by sub-
cutaneous injection is recommended. For therapeutic anticoagulation in pe-
diatric HIT patients, an initial i.v. bolus of 30 IU/kg is followed by continuous
infusion of 1.2–2.0 IU/kg/h (Monagle et al., 2001). The anti-FXa level should
be measured during treatment for optimal dosing. Target levels of anti-FXa
activity are 0.4–0.6 IU/mL for standard and 0.5–0.8 IU/mL for higher dana-
paroid doses (Severin et al., 2002b).
Heparin-Induced Thrombocytopenia in Children 565
B. Lepirudin
Lepirudin (see Chap. 15) is a direct inhibitor of free and clot-bound thrombin
through noncovalent, irreversible binding. In adults with HIT complicated by
thrombosis, the approved dose is an initial bolus of 0.4 mg/kg followed by
continuous i.v. infusion (0.15 mg/kg/h) adjusted by activated partial throm-
boplastin time (aPTT). The usual target aPTT ratio should be 1.5–2.5 times
the normal laboratory mean aPTT. Dosing in children is based on anecdotal
experience. Schiffmann et al. (1997) gave a bolus of lepirudin (0.2 mg/kg)
and a continuous infusion (ranging between 0.1 and 0.7 mg/kg/h) adjusted by
aPTT. Severin et al. (2002b) achieved therapeutic anticoagulation with a
continuous infusion of 0.1 mg/kg/h in a 15-year-old boy, and with an infusion
rate of about 0.15 mg/kg/h in an 8-year-old girl. In an 11-year-old girl, 0.15–
0.22 mg/kg/h was given. In a premature infant, Nguyen and coworkers (2003)
gave a 0.2 mg/kg bolus followed initially by 0.1 mg/kg/h infusion rate; the
dose was adjusted daily based on the aPTT, and 0.03–0.05 mg/kg/hr provided
adequate anticoagulation. Since pharmacokinetics depend largely on renal
function, we recommend starting lepirudin with an i.v. infusion of 0.1 mg/
kg/h (if renal function is normal) and to adjust the dose according to aPTT
4 h later, without initial bolus. This minimizes both risk of overdosing and
anaphylaxis (see Chap. 15).
C. Argatroban
Argatroban (see Chap. 16) is a synthetic direct thrombin inhibitor that binds
reversibly to the active site of thrombin. In adults, the recommended initial
dose of argatroban is 2 Ag/kg/min given by continuous i.v. infusion and
adjusted by aPTT (target range, 1.5–3 times the baseline aPTT). Safety and
efficacy of argatroban in pediatric patients have not been established. Ar-
gatroban has been used in neonates (Boshkov et al., 2003a,b). In one new-
born, an initial bolus (200 Ag/kg) was followed by continuous i.v. infusion (7.5
Ag/kg/min). A 5-month-old infant was also treated with an initial bolus (250
Ag/kg) with subsequent infusion (10 Ag/kg/min).
D. Coumarin
Oral anticoagulants of the coumarin class (warfarin, phenprocoumon) are
not appropriate for therapy of acute HIT (see Chap. 13). HIT patients are at
relatively high risk of developing coumarin-induced skin necrosis and venous
limb gangrene syndromes (see Chap. 3). Therefore, coumarin should be
566 Klenner and Greinacher
Since the pivotal trial in adult orthopedic patients (Warkentin et al., 1995),
it has become clear that LMWH induces HIT less frequently than does UFH.
In children, HIT appears to occur most often among the very young follow-
ing cardiac surgery and among adolescents given UFH to treat spontaneous
thrombosis (Table 2). Data from Pouplard and colleagues (1999) suggest that
HIT also could occur less often if LMWH rather than UFH is used for anti-
thrombotic prophylaxis post–cardiac surgery. This approach should be inves-
tigated in children.
Similarly, in the second group of at-risk pediatric patients (adolescents
with thrombosis), it seems plausible that the frequency of HIT would be
reduced if LMWH is given instead of UFH. Pharmacokinetic studies of
Therapeutic doses
Massicotte et al., 1996 23 0 0 2 0
Dix et al., 2000 143 2 26 6 1
Dix et al., 1998 9 0 4 0 0
Nohe et al., 1999 38 0 No data 0 0
Massicotte et al., 2003a 36 2 32 2 0
Total 249 4 62 10 1
(1.6%) (24.9%) (4%) (0.4%)
Prophylactic doses
Dix et al., 2000 30 1 2 0 0
Dix et al., 1998 5 0 2 0 0
Broyer et al., 1991 46 1 5 7 0
Nohe et al., 1999 10 0 No data 0 0
Massicotte et al., 1999 47 No data 0 3 0
Andrew et al., 1992 78 11 0 0 0
Elhasid et al., 2001 41 0 0 0 0
Massicotte et al., 2003b 78 11 48 0 0
Total 335 24 57 10 0
(7.2%) (17%) (3%)
Heparin-Induced Thrombocytopenia in Children 567
LMWH in infants and children have been conducted for enoxaparin and
reviparin. With both drugs, therapeutic anti-FXa levels are achieved 3–4
hours following the first subcutaneous injection and are similar in both infants
and older children (Albisetti and Andrew, 2002; Massicotte et al., 2003c).
The safety and efficacy of prophylactic and therapeutic doses of LMWH
(enoxaparin, reviparin, dalteparin) in children have been evaluated in clinical
trials for a variety of conditions. LMWH is safe and effective for anticoagu-
lation of infants and children of varying age (Table 3) (Roschitz et al., 2002).
Based on adult experience, substituting UFH with LMWH whenever possi-
ble will likely reduce the risk of HIT in children. However, no data exist in
children to support this supposition.
VIII. SUMMARY
REFERENCES
Albisetti M, Andrew M. Low molecular weight heparin in children. Eur J Pediatr 2002;
161:71–77.
Andrew M, Vegh P, Johnston M, Bowker J, Ofosu F, Mitchell L. Maturation of the
hemostatic system during childhood. Blood 1992; 80:1998–2005.
Bocquet R, Blanot S, Dautzenberg MD, Pierre-Kahn A, Carli P. Antiphospholipid
antibody syndrome in pediatric neurosurgery: a hemostasis problem. Ann Fr
Anesth Reanim 1999; 18:991–995.
Boon DM, Michiels JJ, Stibbe J, van Vliet HH, Kappers-Klunne MC. Heparin-
induced thrombocytopenia and antithrombotic therapy. Lancet 1994; 344:1296.
568 Klenner and Greinacher
thrombosis in children after the Fontan operation: report of two cases. Tex
Heart Inst J 2003; 30:58–61.
Potter C, Gill JC, Scott JP, McFarland JG. Heparin-induced thrombocytopenia in a
child. J Pediatr 1992; 121:135–138.
Pouplard C, May MA, Iochmann S, Amiral J, Vissac AM, Marchand M, Gruel Y.
Antibodies to platelet factor 4-heparin after cardiopulmonary bypass in patients
anticoagulated with unfractionated heparin or a low-molecular-weight heparin:
clinical implications for heparin-induced thrombocytopenia. Circulation 1999;
99:2530–2536.
Ranze O, Ranze P, Magnani HN, Greinacher A. Heparin-induced thrombocytopenia
in paediatric patients—a review of the literature and a new case treated with
danaparoid sodium. Eur J Pediatr 1999; 158(suppl 3):S130–S133.
Ranze O, Rakow A, Ranze P, Eichler P, Greinacher A, Fusch C. Low-dose danaparoid
sodium catheter flushes in an intensive care infant suffering from heparin-
induced thrombocytopenia. Pediatr Crit Care Med 2001; 2:175–177.
Risch L, Fischer JE, Schmugge M, Huber AR. Association of anti-heparin platelet
factor 4 antibody levels and thrombosis in pediatric intensive care patients with-
out thrombocytopenia. Blood Coagul Fibrinolysis 2003; 14:113–116.
Roschitz B, Sudi K, Beitzke A, Gamillscheg A, Leschnik B, Muntean W. Sub-
cutaneous low molecular weight heparin (LMWH) versus intravenous unfrac-
tionated heparin (UFH) bolus in pediatric cardiac catheterization [abstr]. Ann
Hematol 2002; 81:A69.
Sauer M, Gruhn B, Fuchs D, Altermann W, Zintl F. Heparin-induced type II
thrombocytopenia within the scope of high dose chemotherapy with subsequent
stem cell rescue. Klin Padiatr 1998; 210:102–105.
Saxon BR, Black MD, Edgell D, Noel D, Leaker MT. Pediatric heparin-induced
thrombocytopenia: management with Danaparoid (orgaran). Ann Thorac Surg
1999; 68:1076–1078.
Schiffmann H, Unterhalt M, Harms K, Figulla HR, Völpel H, Greinacher A. Suc-
cessful treatment of heparin-induced thrombocytopenia type II in childhood
with recombinant hirudin. Monatssch Kinderheilk 1997; 145:606–612.
Schmugge M, Risch L, Huber AR, Benn A, Fischer JE. Heparin-induced throm-
bocytopenia-associated thrombosis in pediatric intensive care patients. Pedia-
trics 2002; 109:E10.
Severin T, Sutor AH. Heparin-induced thrombocytopenia in pediatrics. Semin
Thromb Hemost 2001; 27:293–299.
Severin T, Dittrich S, Zieger B, Kampermann J, Kececioglu D, Sutor AH. HIT II after
Fontan procedure—treatment with Lepirudin [abstr]. Ann Hematol 2002a; 81:
A77.
Severin T, Zieger B, Sutor AH. Anticoagulation with recombinant hirudin and
danaparoid sodium in pediatric patients. Semin Thromb Hemost 2002b; 28:447–
454.
Spadone D, Clark F, James E, Laster J, Hoch J, Silver D. Heparin-induced throm-
bocytopenia in the newborn. J Vasc Surg 1992; 15:306–311.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-Induced Thrombocytopenia in Children 571
Kevin M. McIntyre
Harvard Medical School, Boston VA Health Care System, and Brigham and
Women’s Hospital, Boston, Massachusetts, U.S.A.
Theodore E. Warkentin
McMaster University and Hamilton Regional Laboratory Medicine Program,
Hamilton, Ontario, Canada
I. INTRODUCTION
573
574 McIntyre and Warkentin
The key issue in a medicolegal case is whether the physician failed to uphold
the perceived ‘‘standard of care’’ in the context of the facts presented and
expert opinions expressed; and whether such failure, if established, was a
substantial contributing factor in bringing about the plaintiff’s injury. Unless
the legal action is dropped, or an out-of-court settlement is reached, the action
generally progresses to the courtroom (with or without prior judicially
overseen medical review, in some states), where it usually is heard and decided
by a jury.
The ‘‘standard of care,’’ as it applies to a malpractice case, generally
refers to how a qualified and reasonably competent practitioner of the same
generalist or specialist class would perform acting under the same or similar
circumstances. The standard of care is developed for the jury through the
interaction of expert witnesses and the lawyers for plaintiff and defendant
sides (McIntyre, 2001). Depending upon the state, this interaction can consist
of experts’ opinions expressed through written reports, and/or as testimony
expressed in response to questioning that primarily is conducted by the
opposing attorney (deposition).
Given the progressively accumulating scientific data base, there is
controversy as to whether a jury system is appropriate for ever more complex
medical issues (McIntyre, 2001; Mello et al., 2003). The jury system may
function reasonably well when the failure of the physician is clear (e.g.,
removal of the wrong leg). But as the requirement for judgment on the part of
the physician increases, especially in a disorder such as HIT with complex
diagnostic and treatment dilemmas, the jury may increasingly find the medical
issues in particular difficult to understand. Even for interested physicians and
scientists, it can be challenging to follow the exponential expansion of the
scientific literature on HIT, with its implications for an evolving standard of
care based upon this new information. The standard of care to be applied to
an individual case must consider the available information at the time the
treatment was applied, which can differ significantly from the standard at the
time of the subsequent medicolegal action itself. Further, diagnostic uncer-
tainty in specific cases of HIT, as discussed earlier, can be considerable, with
the implication that the standard of care to be applied to that case becomes
increasingly ill-defined and uncertain as well.
While the summary of the medicolegal process put forth above tends to
make the U.S. system appear quite fair and orderly, there are those who
believe the process has serious flaws. On the medical side, the increasing
emphasis on scientifically sound evidence as the basis for the development of a
‘‘standard of care’’ for medical practice may have improved the quality of the
medical care in the United States, as judged by such entities as Physician
576 McIntyre and Warkentin
factor’’ (or alternate terminology, depending upon the state), in causing the
damages or injuries alleged by the plaintiff. Depending upon the jurisdiction,
the experts need to express their opinions ‘‘based on a reasonable degree
of medical probability’’ or ‘‘based upon a reasonable degree of medical cer-
tainty’’ or similar such wording.
There are at least seven clinical practice guidelines that discuss one or more
aspects of platelet count surveillance, diagnosis, or management of HIT,
including the guidelines summarized in this book (Olson et al., 1998; Hirsh et
al., 1998, 2001; Greinacher and Warkentin, 2000, 2001; Warkentin, 2002;
Greinacher et al., 2003; see Chap. 13). Although the major purpose of clinical
practice guidelines is to enhance quality of care, it is also possible that their
existence could contribute to medicolegal risk if recommendations are not
followed (McIntyre, 2001). Of course, it is not possible for any consensus
conference or practice guideline to deal explicitly with all of the complex
issues that can arise in any individual case. Thus, there is a major role for
physician judgment in the context of the individual case itself (McIntyre,
2001). Additionally, as Olson (1995) states, ‘‘Readers themselves must assess
the quality and validity of consensus statements as they do all literature.’’
Finally, it remains unproven whether clinical practice guidelines have a
positive impact on patient care (McIntyre, 2001).
Clinical practice guidelines also can demonstrate that the scientific basis
for many recommendations is preliminary, incomplete, or even contradictory.
Increasingly, attempts are being made formally to grade the strength of the
recommendations. The increasing sophistication of the grading systems has
paralleled the growing scope of the recommendations themselves, as shown
by the evolution of the grading systems used by the successive Consensus
Conferences on Antithrombotic Therapy held under the aegis of the Amer-
ican College of Chest Physicians (Sackett, 1989; Cook et al., 1992, 1995;
Guyatt et al., 1998, 2001). The significance of the strength of a recommen-
dation is that a strong scientific basis theoretically would render more difficult
the defense of a deviation of practice from a guideline based upon such strong
evidence (McIntyre, 2001).
Regarding HIT, most of the recommendations are based upon obser-
vational data (level C evidence), rather than randomized clinical trials that
might lead to higher grade recommendations. Nevertheless, it is possible for a
level 1 recommendation to be derived from observational data, provided that
the evidence is convincing and widely accepted.
578 McIntyre and Warkentin
C. Diagnosis of HIT
Although many cases of HIT can be diagnosed with accuracy on clinical
grounds, there are others in which the diagnosis is not certain, and laboratory
confirmation or refutation is important. Moreover, availability of laboratory
testing is variable and may not have been obtained in a patient, or may have
given results that are at odds with the clinical diagnosis. For example, some
tests have limited sensitivity (e.g., platelet aggregation assays), so a negative
test does not necessarily exclude HIT. Further, the transience of HIT anti-
bodies means that the diagnosis cannot usually be established even when
sensitive tests are performed using blood samples obtained several months
580 McIntyre and Warkentin
after recovery from acute HIT (see Chap. 3). As discussed in Chap. 12, some
disorders so closely mimic HIT on clinical grounds as to merit the designa-
tion, ‘‘pseudo-HIT.’’ These disorders can cause ‘‘experts’’ on opposite sides
to disagree fundamentally on whether HIT was even present. Ironically,
certain of the pseudo-HIT disorders also have a high risk for thrombosis or
fatal outcomes. Issues of turnaround time and diagnostic usefulness of
particular assays can also be an issue in some medicolegal actions, given the
considerable heterogeneity in availability and type of diagnostic testing
among various medical centers, as well as the evolving medical literature on
the diagnostic implications of a positive or negative test result (see Chap. 11).
D. Treatment of HIT
Medicolegal issues involving the treatment of HIT are complicated by such
issues as treatment paradoxes, use of nonapproved medications, new infor-
mation about the natural history of isolated HIT, as well as the recent
availability of new drugs for prevention and treatment of thrombosis
complicating HIT, all of which have influenced the standard of care.
Treatment ‘‘Paradoxes’’
There are many seemingly counterintuitive treatment paradoxes in the
management of HIT (Warkentin, 2001; see Table 1 in Chap. 13). Many of
these paradoxes are known to specialists, although a generalist practitioner
may not be familiar with them. For example, prophylactic platelet trans-
fusions are considered to be relatively contraindicated in HIT, even when a
patient is severely thrombocytopenic. This is because bleeding is rare in HIT,
and because platelet transfusions at least theoretically might increase the risk
for thrombosis. Thus, a platelet transfusion ordered for a patient with HIT
who suffers a subsequent thrombosis might be regarded as prima facie (‘‘at
first view’’) evidence for malpractice. However, in a complex case of HIT,
mitigating factors could include uncertainty about the cause of thrombocy-
topenia at the time of transfusion, and other factors suggesting a high risk for
bleeding. Further, although published anecdotes suggest a link between
platelet transfusions and subsequent thrombosis, the growing awareness that
the natural history of HIT itself is for thrombosis to occur frequently even in
the absence of platelet transfusions suggests that any causal association
between a platelet transfusion and a subsequent adverse outcome is specula-
tive. Indeed, this view is reflected in the ‘‘weakest’’ grade (level 2C) assigned
this recommendation proscribing platelet transfusions.
Another example of a counterintuitive treatment paradox is the recom-
mendation that LMWH is contraindicated as treatment for HIT (Hirsh et al.,
U.S. Perspectives on Legal Aspects of HIT 581
2001; see Chap. 13). This recommendation reflects the observation that even
though using LMWH will prevent most cases of HIT (Warkentin et al., 1995,
2000, 2003), use of these preparations in a patient who already has established
HIT has a high probability of treatment failure (Ranze et al., 2000). Although
this recommendation against use of LMWH for treating HIT was given a rela-
tively ‘‘high-grade’’ recommendation by the ACCP Consensus Conference
(1C+), an alternate view holds that LMWH therapy might be acceptable
provided that in vitro cross-reactivity testing is negative using platelet aggre-
gation assays (Slocum et al., 1996). However, problems with such an approach
involve treatment delays pending laboratory testing, as well as the availability
of other agents with low (danaparoid) or absent cross-reactivity (argatroban,
lepirudin). Nevertheless, this contrary viewpoint does show that expert opin-
ion potentially can differ substantially regarding treatment recommendations.
Nonapproved Medications
Although three drugs are widely regarded as safe and effective for managing
HIT, differences exist in their approval status among different countries. For
example, although danaparoid is approved in the United States for preven-
tion of deep vein thrombosis following orthopedic surgery, it does not have a
specific indication in the United States for prevention or treatment of HIT-
associated thrombosis. Nevertheless, its former availability on the U.S.
market meant that physicians had the legal right to use the drug for ‘‘off-
label’’ treatment of HIT (Preuss and Conour, 1999): According to the U.S.
Food and Drug Administration (FDA, 1982), ‘‘accepted medical practice
often includes drug use that is not reflected in approved labeling.’’ Danapa-
roid was withdrawn from the U.S. market by the manufacturer in April 2002
(although it remains available in some other countries [see Chap. 14]).
Another example of a drug that has been used ‘‘off-label’’ to treat HIT is
bivalirudin, a hirudin derivative approved in the U.S. and Canada for anti-
coagulation during percutaneous coronary interventions (Francis et al., 2003;
see Chap. 17). For theoretical reasons, it seems likely that the pentasaccharide
anticoagulant, fondaparinux, will be effective for treating HIT, and it is pos-
sible that this agent (now approved in the U.S., Canada, European Union,
and elsewhere for antithrombotic prophylaxis in certain orthopedic settings)
will see increasing ‘‘off-label’’ use for treating or preventing thrombosis in
HIT in the coming years (see also Chaps. 4 and 8).
Argatroban is the only drug currently approved in the United States for
treatment and prevention of thrombosis in patients with HIT (lepirudin
having been approved in the United States only for treatment of thrombosis
complicating HIT). However, protocols do exist for ‘‘off-label’’ treatment of
HIT using either danaparoid or lepirudin for isolated HIT; in the case of a
582 McIntyre and Warkentin
V. SUMMARY
REFERENCES
Caplan RA, Posner KL, Cheney FW. Effect of outcome on physician judgments of
appropriateness of care. JAMA 1991; 265:1957–1960.
Cook DJ, Guyatt GH, Laupacis A, Sackett DL. Rules of evidence and clinical
recommendations on the use of antithrombotic agents. Chest 1992; 102(suppl):
305S–311S [published erratum appears in Chest 1994; 105:647].
Cook DJ, Guyatt GH, Laupacis A, Sackett DL, Goldberg RJ. Clinical recommenda-
tions using levels of evidence for antithrombotic agents. Chest 1995; 108(suppl):
227S–230S.
FDA Drug Bulletin 1982; 12:4–5.
Francis JL, Drexler A, Gwyn G, Moroose R. Bivalirudin, a direct thrombin inhibitor,
in the treatment of heparin-induced thrombocytopenia [abstr]. J Thromb Hae-
most 2003; 1(suppl 1):P1909.
Funk S, Eichler P, Albrecht D, Ganzer D, Strobel U, Lubenow N, Greinacher A.
Heparin-induced thrombocytopenia (HIT) in orthopedic patients: a prospective
cohort trial comparing UFH and LMWH [abstr]. Ann Hematol 2000; 79(suppl
I):A92.
Ganzer D, Gutezeit A, Mayer G, Greinacher A, Eichler P. Thromboembolieprophy-
laxe als auslöser thromboembolischer Komplicationen. Eine Untersuchung zur
inzidenz der Heparin-induzierten Thrombozytopenie (HIT) Typ II. Z Orthop
1997; 135:543–549.
Greinacher A, Warkentin TE. Treatment of heparin-induced thrombocytopenia: an
overview. In: Warkentin TE, Greinacher A, eds. Heparin-Induced Thrombo-
cytopenia. New York: Marcel Dekker, Inc., 2000:261–290.
Greinacher A, Warkentin TE. Treatment of heparin-induced thrombocytopenia: an
584 McIntyre and Warkentin
Warkentin TE. Platelet count monitoring and laboratory testing for heparin-induced
thrombocytopenia: recommendations of the College of American Pathologists.
Arch Pathol Lab Med 2002; 126:1415–1423.
Warkentin TE. Heparin-induced thrombocytopenia: pathogenesis and management.
Br J Haematol 2003; 121:535–555.
Warkentin TE, Greinacher A. Heparin-induced thrombocytopenia: recognition, treat-
ment, and prevention. Chest 2004. In press.
Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia.
Am J Med 1996; 101:502–507.
Warkentin TE, Kelton JG. Temporal aspects of heparin-induced thrombocytopenia.
N Engl J Med 2001; 344:1286–1292.
Warkentin TE, Levine MN, Hirsh J, Horsewood P, Roberts RS, Gent M, Kelton JG.
Heparin-induced thrombocytopenia in patients treated with low-molecular-
weight heparin or unfractionated heparin. N Engl J Med 1995; 332:1330–1335.
Warkentin TE, Elavathil LJ, Hayward CPM, Johnston MA, Russett JI, Kelton JG.
The pathogenesis of venous limb gangrene associated with heparin-induced
thrombocytopenia. Ann Intern Med 1997; 127:804–812.
Warkentin TE, Chong BH, Greinacher A. Heparin-induced thrombocytopenia: to-
wards consensus. Thromb Haemost 1998; 79:1–7.
Warkentin TE, Sikov WM, Lillicrap DP. Multicentic warfarin-induced skin necrosis
complicating heparin-induced thrombocytopenia. Am J Hematol 1999; 62:44–
48.
Warkentin TE, Sheppard JI, Horsewood P, Simpson PJ, Moore JC, Kelton JG. Im-
pact of the patient population on the risk for heparin-induced thrombocytope-
nia. Blood 2000; 96:1703–1708.
Warkentin TE, Roberts RS, Hirsh J, Kelton JG. An improved definition of immune
heparin-induced thrombocytopenia in postoperative orthopedic patients. Arch
Intern Med 2003. In press.
22
Legal Aspects of Heparin-Induced
Thrombocytopenia: European
Perspectives
Klaus Ulsenheimer
University of Munich, Munich, Germany
Another important legal aspect is the duty of the physician to inform the
patient about the actual risk of thrombosis, including the possible risks and
expected benefits of prophylaxis. This information is divided into two parts:
procedure-related and therapy-related. One example is the current discussion
on pharmacological prophylaxis of thrombosis in outpatients. The Bundes-
gerichtshof (federal court) (1996a,b,c) is of the opinion that the current dis-
cussion in medical science about the danger of thrombosis and the means of
pharmacological prophylaxis in outpatients justify a duty to provide infor-
mation. In these cases, ‘‘patient autonomy demands information about pos-
sible dangers of treatment and the means available to avoid or alleviate such
undesired effects.’’
Information about prophylaxis against thrombosis must include hem-
orrhage; allergic reactions, including HIT and its possible complications; the
need for platelet count monitoring; and, for long-term prophylaxis, osteopo-
rosis. Even if the risk of HIT is very low, information about it must be pro-
vided. Furthermore, information about certain life-threatening consequences
of HIT, such as permanent organ damage and even death, as well as possible
countermeasures against these outcomes, must be communicated. Statistical
probabilities in mathematical terms are not of major importance: they are,
according to the Bundesgerichtshof (1994a), of ‘‘minor importance only.’’
Critical, however, is whether the complications are relatively specific for the
treatment intervention in question. Thus, even extremely rare risks need to be
mentioned if they are known to be associated specifically with an intervention
590 Ulsenheimer
and if their occurrence would have noticeable influence on the patient’s life
and occupation (Bundesgerichtshof 1994b, 1996a; Oberlandesgericht Hamm,
1995). Consequently, there should be no doubt about the duty to inform
about the risks associated with HIT, as these complications are known to be
caused by heparin and, therefore, are specific for this intervention. Indeed, the
court (Oberlandesgericht Celle) ruled that the physician should have in-
formed the patient, at least briefly, about the risk of HIT (Oberlandesgericht
Celle, 2002).
If a doctor recommends using heparin in a low-risk patient outside an
approved indication, the patient must also be informed about this. This is
because in legal terms, approval of a drug is ‘‘like a seal of quality, that—
independent of the actual quality or safety—can be decisive for the patient’s
decision-making in the area where the pharmaceutical law is applicable, so he
has to be informed’’ (Bundesgerichtshof, 1996b).
Physicians are allowed to use drugs to treat diseases for which they have
not been approved, provided the medical necessity arises and there is a ratio-
nale or precedence for its benefit and reasonable safety in the clinical context
(Oberlandesgericht Köln, 1991; Bundessozialgericht, 2002). The regulatory
approval of a drug merely creates a state of confidence; that is, the doctor can
rely on the fact ‘‘that the risk versus benefit ratio is considered favorable in the
light of the evidence provided by the manufacturer and the examination of the
Bundesinstitut für Arzneimittel und Medizinprodukte’’ (Weißauer, 1994).
On the other hand, for a doctor using a nonapproved drug or an ap-
proved drug outside its approved indication, this state of confidence does not
cover him or her in the event of damaging side effects. The physician may then
be required to justify the treatment, for example, by showing that the drug has
been used with a favorable side effect profile, that reputable specialists
recommend its use, and that no alternative treatments are available. This is
not an uncommon situation in the management of complications of HIT
itself, where an alternative anticoagulant, danaparoid sodium, is often used
outside its approved indication (i.e., antithrombotic prophylaxis following
orthopedic surgery), or even without any approval in some countries, for the
treatment or prevention of thrombosis associated with HIT.
The physician must inform the patient about reasonable precautions in
optimizing the safety of a prescribed treatment. This includes educating
outpatients about typical signs and symptoms of therapeutic complications.
For example, informing patients about the possibility and significance of skin
reactions at heparin injection sites as an early manifestation of HIT is
appropriate. The main content of the information given to the patient should
be documented in writing by the physician, as legal protection in the event of a
subsequent adverse event occurring. It is especially prudent to document the
European Legal Aspects 591
Cost is not an argument against indicated prophylaxis. The less expensive but
equally effective drug should be used, but social law’s duty to work econom-
ically does not legitimize lowering the standard of medical care. Medical duty
for care and the duty to work economically are not mutually exclusive, as
social law acknowledges the need for approved treatment. However, eco-
nomic considerations and price have to give way to aspects of effectiveness of
a drug and its indication. So, if there is an appropriate indication for
prophylaxis for an individual patient’s health, insurance must bear the cost.
REFERENCES
2 1 0
We recommend:
1. Monitoring for typical-onset HIT: stratifying the intensity of
platelet count monitoring for HIT based upon its risk
A. Patients at highest risk for HIT (1–5%) (e.g., postoperative
patients receiving prophylactic-dose UFH after major sur-
gery): monitoring during heparin therapy, at least every second
day from day 4 to day 14.*
Patients receiving therapeutic-dose UFH: platelet count mon-
itoring once dailyy from day 4 to day 14.*
B. Patients at intermediate risk for HIT (0.1–1%) (e.g., medical/
obstetrical patients receiving prophylactic-dose UFH, or post-
operative patients receiving prophylactic-dose LMWH, or
postoperative patients receiving intravascular catheter
‘‘flushes’’ with UFH): monitoring during heparin therapy, at
least every 2 or 3 days from day 4 to day 14,* when practical.z
C. Patients at low risk for HIT (<0.1%) (e.g., medical/obstetrical
patients receiving prophylactic- or therapeutic-dose LMWH,
or medical patients receiving only intravascular catheter
‘‘flushes’’ with UFH): routine platelet count monitoring is
not recommended.§
These are draft recommendation (Seventh American College of Chest Physi-
cians Consensus Conference on Antithrombotic Therapy, September 2003).
Readers should consult the publication (Warkentin and Greinacher, 2004) to
obtain the final recommendations.
2. Monitoring for rapid-onset HIT: for a patient recently exposed to
heparin (within the past 100 days), a repeat platelet count within 24
hours following reinitiation of heparin.
* The crucial time period for monitoring ‘‘typical-onset’’ HIT is between days 4 and 14 (first
day of heparin = day zero), where the highest platelet count from day 4 (inclusive) onwards
represents the ‘‘baseline.’’ Platelet count monitoring can cease before day 14 when heparin is
stopped.
y
Once-daily platelet count monitoring recommended as daily blood draws required for aPTT
monitoring.
z
Frequent platelet count monitoring may not be practical when UFH or LMWH is given to
outpatients.
§
Monitoring as per ‘‘intermediate’’ risk is appropriate if UFH was given before initiating
LMWH.
Appendixes 601
* The grades of recommendation are from Guyatt et al. (2001) and Hirsh et al. (2001) (in Chap.
13) and are described in Chap. 13.
Appendixes 603
Abbreviations: aPTT, activated partial thromboplastin time; b.w., body weight; CPB, cardiopulmonary
bypass; ECT, ecarin clotting time; iv, intravenous; MI, myocardial infarction; PCI, percutaneous coronary
intervention; UA, unstable angina.
a
A maximum body weight of 100 kg should be used for dose calculations.
b
Adjust for renal insufficiency.
c
The ratio is based on comparison with the normal laboratory mean aPTT. If Actin FS or Neothromtin
reagents are used, the aPTT target range is usually 1.5–3.0.
d
Used in the HAT-1, HAT-2, and HAT-3 trials.
e
This is the author’s recommended starting dose in all HIT patients, unless life- or limb-threatening
thrombosis is present.
f
Tested in a prospective, randomized trial after orthopedic surgery (Eriksson et al., 1996, 1997).
g
Stop 15 min before end of CPB; put 5 mg into CPB after disconnection to avoid clotting of pump.
h
The target lepirudin level pre-CPB (>2.5 Ag/mL) is lower than the level sought during CPB (3.5–4.5 Ag/
mL) because of the addition of lepirudin to the pump priming fluid (0.2 mg/kg b.w.).
610 Appendixes
613
614 Index