Heparin was discovered 90 years ago,1  and within 2 decades it was being widely used as an anticoagulant.2  The numerous advantages of heparin lend to its broad use including its immediate onset of action, relatively short half-life (∼60 minutes), simple laboratory monitoring (aPTT), ability to be reversed (using protamine), and low cost. Despite a long experience of using heparin, the counterintuitive notion that an anticoagulant could be intensely prothrombotic took almost 4 decades to become recognized.

Our understanding of heparin-induced thrombocytopenia (HIT) has increased considerably and to a large measure exemplifies the best of translational research. Sometimes, basic studies investigating the pathophysiology of HIT lead to clinical insights. In other instances, clinical observations prompted basic studies. Together, these basic and applied investigations have dramatically enhanced our understanding of the pathogenesis and treatment of HIT.

On June 1, 1957, at the Fifth Scientific Meeting of the International Society of Angiology in New York, Rodger E. Weismann, an Assistant Professor of Clinical Surgery at the Dartmouth Medical School, and his Resident in Surgery, Dr Richard W. Tobin, described 10 patients who developed arterial embolism during systemic heparin therapy.3  The first reported embolic event was a femoral artery embolic occlusion, which occurred in a 62-year-old woman who was receiving heparin because of a deep-vein thrombosis (DVT). Three days after successful femoral embolectomy, and while continuing to receive heparin, she developed sudden occlusion of the distal aorta requiring distal aortic and bilateral iliac embolectomies.3 

Multiple thromboemboli were observed in 9 of the 10 patients reported. Six patients died as a result of the thromboembolism, and 2 survivors underwent above-knee amputation.3  The clots were described as “pale, soft, salmon-colored” and were composed mostly of fibrin, platelets, and leukocytes. The arterial emboli began on average 10 days after commencing heparin treatment.

An additional 11 patients were described 5 years later by Roberts and colleagues.4  These vascular surgeons noted the paradox of “unexplained arterial embolization [occurring] 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.” Thus, both groups3,4  noted the temporal hallmark of HIT, a delay of approximately 1 week from initiation of heparin to onset of its thrombotic manifestations.

Routine platelet count measurements were not routinely performed until the 1970s. This may explain why thrombocytopenia was not reported in the first 24 patients with heparin-induced arterial emboli.3-6  In 1969, the term “heparin-induced thrombocytopenia” was used by Natelson7  to describe a 78-year-old man with pulmonary embolism who developed severe thrombocytopenia after heparin. Over the ensuing days, 2 separate periods of heparin readministration were each characterized by abrupt platelet count declines and corresponding increases in fibrinogen. The addition of heparin to the patient's citrated platelet-rich plasma caused platelet aggregation. The striking dichotomy of a heparin-induced platelet count decrease, but rise in the level of fibrinogen, captures the essence of HIT: heparin simultaneously promotes and treats the thrombosis.

The first to identify the central features of the HIT syndrome—thrombocytopenia, thrombosis, and its immune pathogenesis—were Drs Silver, Rhodes, and Dixon.8  In their 1973 paper, they described 2 patients with severe thrombocytopenia, myocardial infarction, and heparin resistance, with platelet count recovery on discontinuing heparin treatment.8  Both patients developed rapid recurrence of thrombocytopenia when heparin rechallenges were given.

An immune basis for this syndrome was suggested by increased numbers of bone marrow megakaryocytes and a rapid recurrence of the thrombocytopenia upon heparin reexposure. A circulating heparin-dependent, platelet-activating substance (subsequently identified as IgG) was found in the patients' blood, which caused aggregation of donor platelets in the presence of heparin.

A subsequent report by Rhodes and coinvestigators9  helped establish HIT as a distinct syndrome. Eight patients were described with thrombocytopenia (platelet count nadir, 25 × 109/L) that occurred during heparin administration. Thrombotic, rather than hemorrhagic, complications predominated: 7 patients had new or recurrent thromboembolic events, and one patient had a hemorrhagic stroke. Complement-fixing, heparin-dependent antibodies were found in blood from 5 patients.

During the latter half of the 1970s, at least 7 groups of investigators10-16  reported patients who resembled those of Rhodes and colleagues. Together, these studies provided further evidence of an immunologic basis for HIT. A common feature of these reports was evidence of platelet activation induced by patient serum, plasma, or purified immunoglobulin.11-17 

Doubts persisted regarding the immune basis of HIT. After all, Rhodes and colleagues had also noted that when their patients were deliberately rechallenged with heparin several months after their episode of HIT, thrombocytopenia failed to recur.8,9  Moreover, HIT patients typically did not exhibit severe thrombocytopenia and purpura—features characteristic of established drug-induced immune-mediated thrombocytopenia reported with quinine, among other drugs. Also arguing against the model of typical immune-mediated drug-induced thrombocytopenia were our observations that thrombocytopenia could resolve while the patient continued on heparin therapy.18 

But other evidence supported an immune pathogenesis: for example, Cines and associates19  documented heparin-dependent binding of complement to platelets. Another observation that suggested a complex antigen was provided by Green and coworkers.12  These investigators demonstrated that the antibodies did not bind to heparin per se, but required heparin plus a cofactor found in platelet lysates.

The uncertainty regarding etiopathogenesis was also reflected in the name of the condition: HIT was often termed heparin-associated thrombocytopenia to reflect the assumption that the thrombocytopenia might not be directly related to the heparin.20  Some investigators21-23  raised the possibility that a procoagulant contaminant within the heparin preparation could be causing the reaction, perhaps by triggering disseminated intravascular coagulation. Further support for this hypothesis was provided by the demonstration that different heparin preparations carried different risks for thrombocytopenia.18,24  In addition, consistent with a “contaminant” possibility was the fact that heparin is unique among almost all medications: it consists of polydispersed and heterogeneous polysaccharide chains that have been extracted and partially purified from either beef lung or pork intestinal mucosa.

Heparin itself can directly interact with platelets and cause variable platelet aggregation when added to platelets in vitro. In addition, some investigators have reported that when heparin is injected into patients, there can be an early, transient drop in platelet count.25,26  At a platelet immunobiology workshop held in Milwaukee, it was proposed that the early, nonimmune disorder would be termed “HIT, Type 1” and the later onset immune disorder would be called “HIT, Type 2.27  Today these designations are not commonly used and most physicians designate the immune disorder simply as heparin-induced thrombocytopenia (HIT).

Our research group became interested in developing a laboratory test to diagnose HIT. Although platelet-associated IgG levels were elevated in HIT patients, we found that it was not a diagnostically useful assay. Subsequent investigations have documented that the various assays for PAIgG have minimal diagnostic usefulness in any thrombocytopenia.19,28,29  We next prospectively evaluated the widely used assay of heparin-dependent platelet aggregation.30  The results indicated that heparin-dependent platelet aggregation was only a moderately useful test for HIT and although it had a high specificity, its sensitivity was low. We also made an observation that subsequently proved informative, namely that platelet aggregation was maximal at pharmacologic concentrations of heparin (0.1 U/mL), but that higher heparin concentrations (above 5 U/mL final) often produced a negative result.

Our next aim was to improve the diagnostic usefulness of the functional (platelet activation) assay for HIT.31  The sensitivity was increased by optimizing the reactivity of the test platelets and by using the release of radiolabeled 14C-serotonin as the end point. The specificity was increased by observing that the platelet-activating effects of HIT antibodies were inhibited at high heparin concentrations (Figure 1). The serotonin release assay (SRA) was prospectively evaluated against a group of 600 patient and control samples, and proved both sensitive and specific. Subsequent modifications have further enhanced the assay's diagnostic utility.32 

Figure 1

The relationship between the percentage release of C-serotonin from platelets obtained from healthy individuals (ordinate) and the final concentration of heparin (abscissa). Each series of points is the result obtained with serum from a different patient with definite heparin-induced thrombocytopenia, The lines numbered 1 to 4 represent 4 different serum samples not from patients with heparin-induced thrombocytopenia. These samples induced release at 0.1 and 100 U/mL of heparin. Numbers 1 and 2 are sera from 2 patients with immune thrombocytopenic purpura. Number 3 is the serum from a hospitalized control, and number 4 is heat-aggregated IgG. Adapted from Sheridan et al31  with permission.

Figure 1

The relationship between the percentage release of C-serotonin from platelets obtained from healthy individuals (ordinate) and the final concentration of heparin (abscissa). Each series of points is the result obtained with serum from a different patient with definite heparin-induced thrombocytopenia, The lines numbered 1 to 4 represent 4 different serum samples not from patients with heparin-induced thrombocytopenia. These samples induced release at 0.1 and 100 U/mL of heparin. Numbers 1 and 2 are sera from 2 patients with immune thrombocytopenic purpura. Number 3 is the serum from a hospitalized control, and number 4 is heat-aggregated IgG. Adapted from Sheridan et al31  with permission.

Close modal

A sensitive and specific test for a disorder can also be used to characterize the component reactions. For example, we were able to confirm the work of previous investigators that the reaction was mediated by IgG. Furthermore, the unimodal pattern of antigen-dependent progressive platelet activation followed by reduced platelet activation (at higher heparin concentrations) suggested to us that this was an immune complex type of reaction.31 

In an attempt to identify the platelet target for the HIT antibodies, we tested platelets with congenital deficiencies of glycoprotein IIb/III (Glanzmann thrombasthenia) and platelets deficient in glycoprotein Ib/IX/V complex (Bernard-Soulier syndrome).33  The identical reaction to normal platelets indicated that these glycoproteins did not directly participate in the reaction. Next, IgG from patients with HIT was purified and digested. The requirement of F(ab′)2 indicated that HIT was an immunologic disorder recognizing a specific, but as yet unidentified, epitope. Inhibition by human or animal Fc indicated that HIT was an immune complex disorder. Finally, the complete inhibition of platelet reactivity via a monoclonal antibody against the platelet Fc receptor confirmed that platelet activation in HIT was mediated through the platelet Fc receptor.33 

We now know that HIT-mediated platelet activation is a dynamic process34  in which the Fab from the HIT-IgG binds to platelet-associated PF4. Subsequently the Fc region on the IgG molecule binds to the Fc receptor on the same35  or adjacent platelets, which in turn triggers platelet activation.33,36 

The platelet Fc receptor was identified by Karas et al37  with further characterization by Rosenfeld et al38  as well as by our group.39  Platelets carry the FcγIIa receptor, which is a low affinity receptor present at relatively low copy numbers (∼1000 to 2000 copies per platelet).37-39  IgG immune complexes, which form in patients with HIT, produce receptor cross-linking, which strongly activates platelets. A common polymorphism of FcγIIa (arg131/his131) have proven to be of minor relevance in HIT.

It is rare for a prothrombotic disorder to cause both arterial and venous thrombosis. HIT is an exception. In many HIT patients there was evidence of hypercoagulability, but the pathway that could initiate both arterial as well as venous thrombosis remained unexplained. Three explanations were proposed: (1) HIT antibodies could bind to and injure endothelial cells, thereby initiating coagulation40,42 ; (2) HIT antibodies could bind to monocytes and release tissue factor43,44 ; and (3) HIT antibodies induce a platelet procoagulant response.

Our group focused on the platelet procoagulant explanation. Using flow cytometry, we found that HIT sera caused platelets to form microparticles (Figure 2).45  In contrast, quinine/quinidine-induced thrombocytopenia sera did not cause microparticles, even though there was a dramatic increase in drug-dependent antibody binding to platelets. The procoagulant nature of the HIT microparticles was evidenced by shortening of the Russell viper venom time.45  We also showed that in at least some patients, there were detectable circulating platelet-derived microparticles during the acute episode of HIT.45  In subsequent comparative studies, we found that the magnitude of this platelet procoagulant response induced by HIT antibodies exceeded that of other physiologic agonists, including thrombin and collagen.46  Microparticle formation induced by HIT sera was sufficiently reliable to be used as a diagnostic test for HIT.47 

Figure 2

Platelet-derived microparticles generated by heparin-induced thrombocytopenia serum. Platelets and microparticles were identified using fluorescence (FL1, FITC-antiGPIb3) and size (FSC, forward scatter) characteristics. The platelets had been incubated with heparin-induced thrombocytopenia serum in the presence of 0, 0.1, 0.3, or 100 U/mL heparin. The microparticles (inset box) were generated in the presence of 0.1 and 0.3 U/mL heparin, but not at 0 or 100 U/mL heparin. Results of the representative experiment are shown. Adapted from Warkentin et al45  with permission.

Figure 2

Platelet-derived microparticles generated by heparin-induced thrombocytopenia serum. Platelets and microparticles were identified using fluorescence (FL1, FITC-antiGPIb3) and size (FSC, forward scatter) characteristics. The platelets had been incubated with heparin-induced thrombocytopenia serum in the presence of 0, 0.1, 0.3, or 100 U/mL heparin. The microparticles (inset box) were generated in the presence of 0.1 and 0.3 U/mL heparin, but not at 0 or 100 U/mL heparin. Results of the representative experiment are shown. Adapted from Warkentin et al45  with permission.

Close modal

To better understand these microparticles, we examined their morphology using a variety of microscopic techniques (Figure 3).48  Using confocal microscopy, HIT antibodies plus heparin could be shown to produce platelet particles. Scanning and transmission electron microscopy showed that these microparticles ranged in size from 0.1 to 1.0 μm in diameter. These studies demonstrated that platelet activation resulted in the formation of localized points of swelling on the platelet body with the formation of well-defined buds. These platelet buds are released from the platelets to form the microparticles.48 

Figure 3

Representative scanning electron micrographs of resting platelets and platelets activated with HIT serum in the presence of heparin. Resting platelets or platelets incubated with HIT serum in the presence of 0.1 U/mL heparin were fixed with 2% glutaraldehyde and processed for scanning electrom microscopy (A) The morphology of the representative normal resting platelet. Resting platelets were observed to generally maintain a discoid form. (B) The morphology of a representative platelet incubated with HIT serum. These platelets demonstrated several morphologic changes including absence of a discoid form, presence of pseudopodia and presence of microparticles near the end of the pseudopodia (indicated by arrows). Reprinted from Hughes et al48  with permission.

Figure 3

Representative scanning electron micrographs of resting platelets and platelets activated with HIT serum in the presence of heparin. Resting platelets or platelets incubated with HIT serum in the presence of 0.1 U/mL heparin were fixed with 2% glutaraldehyde and processed for scanning electrom microscopy (A) The morphology of the representative normal resting platelet. Resting platelets were observed to generally maintain a discoid form. (B) The morphology of a representative platelet incubated with HIT serum. These platelets demonstrated several morphologic changes including absence of a discoid form, presence of pseudopodia and presence of microparticles near the end of the pseudopodia (indicated by arrows). Reprinted from Hughes et al48  with permission.

Close modal

In 1992, in a significant advance for HIT-related research, Amiral et al49  identified the elusive platelet component of the HIT antigen when he demonstrated that platelet factor 4 (PF4), a tetrameric member of the C-X-C subfamily of chemokines, formed complexes with heparin that bound HIT antibodies. This seminal observation was confirmed and extended by other groups.42,50,51  The antigen was shown to be present on multimolecular complexes of PF4 and heparin that required an optimal stoichiometric ratio of PF4:heparin of 1:1 to 2:1. In addition, it was shown that endothelial cells could bind PF4 and also produce the antigen. The identification of PF4 as the target antigen allowed the development of enzyme-immunoassay (EIA) techniques.52 

The amino acid composition of PF4 had already been identified in 1987 by Poncz and colleagues.53  The small size of the PF4 monomer (70 amino acids) allowed Horsewood to synthesize a series of overlapping peptides that spanned the entire length of the PF4 molecule.54  We found that a minimal length of PF4 (19 amino acids encompassing the carboxy-terminal peptide including the lysine-rich moiety that binds heparin) was required for reactivity with certain HIT antibodies.54  But, it was not possible to identify a linear epitope on PF4 that served as a target antigen. These results suggested that heparin molecules bundle the PF4, resulting in conformational changes to the molecule, which in turn, become the binding sites for the HIT-IgG.54  Studies by the groups in Milwaukee55  and Philadelphia56,57  have identified several regions on PF4 that are binding sites for HIT antibodies. More recent studies suggest that the antigen sites are located at apposition points where PF4 tetramers are brought into proximity through charge neutralization by heparin.58 Figure 4 presents a schematic of the pathogenesis of HIT.

Figure 4

A schematic representation of the interaction HIT-IgG with platelets. In panel A platelet factor 4 (PF4) is released from platelet α-granules after platelet activation. The PF4 binds to the platelet surface through glycosaminoglycans (GAGs) that are associated with tetramers of PF4. Panel B shows heparin binding to the PF4. The size of the PF4 bundle in part is related to the chain length of the heparin. Panel C shows HIT-IgG binding to epitopes on the PF4/heparin complex. Panel D shows ultra-large complexes (ULC) of heparin/PF4 and IgG binding to platelets via the platelet FcRγIIa. The immune complexes are potent platelet activators leading to the release of platelet-derived procoagulant microparticles.

Figure 4

A schematic representation of the interaction HIT-IgG with platelets. In panel A platelet factor 4 (PF4) is released from platelet α-granules after platelet activation. The PF4 binds to the platelet surface through glycosaminoglycans (GAGs) that are associated with tetramers of PF4. Panel B shows heparin binding to the PF4. The size of the PF4 bundle in part is related to the chain length of the heparin. Panel C shows HIT-IgG binding to epitopes on the PF4/heparin complex. Panel D shows ultra-large complexes (ULC) of heparin/PF4 and IgG binding to platelets via the platelet FcRγIIa. The immune complexes are potent platelet activators leading to the release of platelet-derived procoagulant microparticles.

Close modal

Amiral et al59  have also documented several other potential heparin-dependent antigens, including interleukin-8 and neutrophil-activating peptide-2, but antibodies against these chemokines probably explain only a tiny minority of cases of HIT.60 

The demonstration that HIT was caused by PF4/heparin-containing immune complexes that activated platelets through their Fcγ receptors suggests a central role of the IgG immunoglobulin class in the pathophysiology of HIT. The HIT-IgG is polyclonal and IgG1 predominates with smaller amounts of IgG2.61  Although PF4/heparin-reactive IgM and IgA antibodies are also formed in heparin-treated patients, their role in HIT remains uncertain.60,62,63 

Certain aspects about the immunobiology of HIT remain unexplained. The high frequency of heparin-induced autoantibodies against the self protein PF4 is perplexing. In addition, the reason for the remarkably rapid production of HIT-IgG antibodies, occurring as early as 5 days after commencing heparin, is uncertain. As well, the brief persistence of this antibody in patients with HIT (Figure 5)64  also remains unexplained, as does the lack of an anamnestic immune response upon reexposure.64,65 

Figure 5

Kaplan-Meier analysis of the proportion of patients with heparin-dependent antibodies after an episode of heparin-induced thrombocytopenia. The time (in days) to a negative test by the activation assay (144 patients) or the antigen assay (93 of the 144 patients) is shown. All 144 patients initially had positive tests for heparin-dependent antibodies and underwent subsequent testing within 180 days. The date the test result became negative was estimated to be the date midway between the date of the last positive test and the date of the first negative test. The antigen assay tended to become negative more slowly than did the activation assay (P = .007 by the sign test). The bars indicate 95% confidence intervals. Reprinted from Warkentin and Kelton64  with permission.

Figure 5

Kaplan-Meier analysis of the proportion of patients with heparin-dependent antibodies after an episode of heparin-induced thrombocytopenia. The time (in days) to a negative test by the activation assay (144 patients) or the antigen assay (93 of the 144 patients) is shown. All 144 patients initially had positive tests for heparin-dependent antibodies and underwent subsequent testing within 180 days. The date the test result became negative was estimated to be the date midway between the date of the last positive test and the date of the first negative test. The antigen assay tended to become negative more slowly than did the activation assay (P = .007 by the sign test). The bars indicate 95% confidence intervals. Reprinted from Warkentin and Kelton64  with permission.

Close modal

The chemical and structural determinants allowing heparin to induce antigenic changes in the PF4 molecule have been studied by a number of investigators.50,66,67  The impetus was both scientific and commercial: one goal was the identification of a nonheparin polyanion that could be useful for in vitro testing. Visentin and coworkers identified such a substance (polyvinyl sulfonate) that is currently used in a commercial EIA.67  Another focus of research has been to identify a heparin species that would neither initiate nor propagate HIT. In vitro studies indicated that a variety of sulfated polysaccharides could substitute for heparin to inducing the antigenic changes in PF4. However, there were 2 key determinants that were required for antigenicity. First, a certain chain length (ie, approximately 1000 Da); and second, a minimal amount of sulfation per saccharide unit were required.50,66  These results suggested that progressively smaller heparin preparations carry a progressively lower risk of HIT. Clinical validation of this potential has been provided by the demonstration that low-molecular-weight heparin (enoxaparin) carries a lower risk of HIT of inducing antibodies and of causing clinical HIT, compared with UFH.68 

Recently, there has been interest in whether fondaparinux, a pentasaccharide modeled after the antithrombin-binding region of heparin, will be even less likely to cause or propagate HIT. Interestingly, despite a negligible potential to create epitopes recognized by HIT antibodies in vitro, studies suggest that anti-PF4/heparin antibodies identical to those formed by unfractionated heparin can be generated during treatment with fondaparinux.69  However, these antibodies do not usually bind to PF4 in the presence of fondaparinux,69  suggesting a very low potential to cause or to potentiate HIT. To date, only 2 patient cases of HIT possibly caused by fondaparinux have been reported.70,71  It is possible that in the future, fondaparinux will be used as a treatment for HIT.

Systematic serosurveillance studies of heparin use have shown that only a small proportion, at most 5% to 30% of patients who form HIT-IgG, will develop clinical HIT, depending upon the patient population in question.52,63,72  Among antibody-positive patients, only approximately half have detectable antibodies of IgG class; of these, approximately half again show biologic activity.63  Even among patients with platelet activating antibodies, the risk of HIT increases with the greater magnitude of HIT-IgG–induced platelet activation. However, even some patients with strong platelet-activating antibodies do not develop HIT, suggesting that patient-dependent risk factors, such as platelet Fcγ receptor numbers73  or increased platelet-associated PF4 levels74-76  may also play a role. The relationship among the clinical presentation of HIT, the presence of HIT antibodies, and the type of heparin used can be conceptualized as an iceberg (Figure 6).77 

Figure 6

The relationship among the clinical expression of HIT (thrombocytopenia with or without thrombosis), the type of heparin used, and the antibodies that cause HIT. This can be conceptualized as an “iceberg.” The visible component of the iceberg (the portion above the waterline) represents clinically evident features of HIT, such as thrombocytopenia and/or thrombosis. The mass of the iceberg corresponds to the entire spectrum of anti-PF4/heparin antibodies generated. Some of these antibodies will be biologically active (platelet-activating), and others will be nonplatelet-activating which may make them unlikely to cause clinical consequences. The type of heparin given to the patient determines the overall size of the iceberg, with unfractionated heparin (UFH) being the largest (most immunogenic), and low-molecular-weight heparin and fondaparinux have lesser immunogenicity. Also illustrated in this figure is the vivo cross-reactivity. UFH forms antigens which are readily recognized by HIT antibodies. In contrast, fondaparinux forms poorly recognized antigens. LMWH is intermediate. Illustration by A.Y. Chen.

Figure 6

The relationship among the clinical expression of HIT (thrombocytopenia with or without thrombosis), the type of heparin used, and the antibodies that cause HIT. This can be conceptualized as an “iceberg.” The visible component of the iceberg (the portion above the waterline) represents clinically evident features of HIT, such as thrombocytopenia and/or thrombosis. The mass of the iceberg corresponds to the entire spectrum of anti-PF4/heparin antibodies generated. Some of these antibodies will be biologically active (platelet-activating), and others will be nonplatelet-activating which may make them unlikely to cause clinical consequences. The type of heparin given to the patient determines the overall size of the iceberg, with unfractionated heparin (UFH) being the largest (most immunogenic), and low-molecular-weight heparin and fondaparinux have lesser immunogenicity. Also illustrated in this figure is the vivo cross-reactivity. UFH forms antigens which are readily recognized by HIT antibodies. In contrast, fondaparinux forms poorly recognized antigens. LMWH is intermediate. Illustration by A.Y. Chen.

Close modal

Whether the presence of anti-PF4/heparin antibodies confers adverse prognosis in certain clinical situations—even in the absence of clinical HIT—is an emerging topic of debate.78-81 

The development of an animal (murine) model of HIT has been difficult. The problem reflects the lack of FcγIIA receptors on murine platelets. Furthermore, murine platelets do not express a PF4 immunologically similar to human PF4. Nonetheless progress is being made in this area. Using a double-transgenic mouse expressing both FcγRIIA and human PF4, Reilly and coinvestigators82  injected the mice with KKO,83  a mouse monoclonal antibody raised against human PF4/heparin. The mice were then injected with heparin, developing thrombocytopenia, including some with thrombosis. This model was used to demonstrate the importance of platelet-bound PF4 in the development of thrombocytopenia. The investigators further evolved the murine model using the same double transgenic mice, but with a murine PF4 knockout. This model demonstrated the importance of host factors such as hypercholesterolemia in thrombocytopenia and thrombosis.84  A murine model was used to investigate the immunogenic potential of various ratios of PF4 (murine) to unfractionated heparin.85  These investigators determined that high ratios of murine PF4 to heparin were more immunogenic than the more equimolor ratio that serves as the optimum target for antibody binding.

The thrombocytopenia of HIT is typically moderate (median platelet count nadir, ∼60 × 109/L), which is different from classic drug-induced immune thrombocytopenic purpura (median nadir, 10 × 109/L).86  Some patients with HIT will have a drop in the platelet count that does not necessarily reach the conventional threshold that defines thrombocytopenia (platelet count < 150 × 109/L).87  This can pose diagnostic challenges, most often in the setting of postoperative thromboprophylaxis, where elevated platelet counts can be observed.

HIT was first described as an arterial prothrombotic disorder, perhaps because of its dramatic onset and its recognition by vascular surgeons.3-6  For a number of years physicians did not recognize venous thrombi as part of the syndrome. The problem with attributing venous thromboembolism (VTE) to the HIT syndrome was that patients requiring heparin were sometimes at the highest risk for developing VTE. Consequently, any association could be collateral rather than causal. Clarification of the importance of VTE as a manifestation of HIT was provided by Boshkov et al88  This retrospective study related the site of thrombosis to potential risk factors among patients with serologically confirmed HIT. These researchers found that the postoperative state was strongly associated with development of HIT-associated VTE. It is now appreciated that VTE predominates over arterial thrombosis in HIT in a ratio of approximately 4:1.89,90  It is also apparent that patient-dependent risk factors play a role in the site of the thrombosis. For example, recent arterial surgery or severe atherosclerosis is associated with arterial thrombosis in HIT, while a central venous catheter predisposes to development of upper limb DVT in patients with HIT.88,91  Inherited prothrombotic risk disorders such as factor V Leiden have not been shown to be play a major role in explaining HIT-associated thrombosis, although a contributory role in specific circumstances remains plausible.92 

Venous limb gangrene is another manifestation of HIT, which usually occurs as a result of warfarin-induced microthrombosis due to an altered procoagulant-anticoagulant balance. In this disorder, HIT-associated hypercoagulability interacts with warfarin induced protein C depletion.93-95  Other venous thrombotic events in HIT include unilateral or bilateral adrenal hemorrhagic necrosis (resulting from adrenal vein thrombosis) and cerebral venous (dural sinus) thrombosis.70,96 

These concepts help explain the oftentimes severe clinical course of patients with delayed-onset HIT.96-98  In this disorder, thrombocytopenia and thrombosis begin days to weeks after heparin has been stopped. Antibodies from these patients can cause platelet activation, in vitro, in the absence of heparin.97  It is possible that delayed-onset HIT is caused by binding of HIT-IgG to a complex of platelet-bound. PF4 and chondroitin sulfate.75,99 

By 1998, there was sufficient agreement regarding the pathophysiologic, clinical, and laboratory diagnostic features of HIT that a consensus statement was prepared by representatives of 3 research groups.100  HIT was conceptualized as a clinicopathologic syndrome with one or more clinical events (thrombocytopenia with or without thrombosis) temporally related to heparin administration and caused by HIT antibodies.

The treatment of HIT has paralleled an understanding of its pathophysiology. Initial strategies did not treat the hyperco-agulability of HIT, and consisted of heparin cessation alone,89,101  warfarin,89,102  ancrod,102,103  and antiplatelet agents such as aspirin.104  More recently, agents that inhibit the generation of thrombin, or that inhibit thrombin itself, such as danaparoid,102,105-107  lepirudin,108-111  and argatroban have been used.112-114 

An important first step after diagnosing HIT is to stop the heparin. Ironically, for some patients, continuation of heparin does not result in clinical worsening, and even resolution of thrombocytopenia has been observed. In other patients, the thrombotic events would begin or worsen when the heparin was discontinued. In 1996, we described a group of patients with HIT and isolated thrombocytopenia.89  When these patients were managed conservatively by cessation of heparin (with or without initiation or continuation of warfarin) approximately 50% developed a thrombosis within the next days to weeks (Figure 7).89  Similar observations were made by Wallis and colleagues who also reported that heparin cessation often failed to prevent thrombotic events.101  The precise risk of thrombosis after discontinuation of heparin in a patient with HIT and isolated thrombocytopenia remains uncertain with risk estimates range from 20% to 50%.115  The duration of anticoagulation for a patient with HIT and isolated thrombocytopenia is not known, but we typically continue anticoagulation for 6 to 8 weeks. This is anecdotal and in part reflecting the ongoing prothrombotic risk we observed in our retrospective study (Figure 7).

Figure 7

Cumulative frequency of thrombosis in HIT patients presenting with isolated thrombocytopenia. Approximately 50% of HIT patients initially recognized with isolated thrombocytopenia developed objective evidence for thrombosis during the subsequent 30-day period. Reprinted from Warkentin and Kelton89  with permission.

Figure 7

Cumulative frequency of thrombosis in HIT patients presenting with isolated thrombocytopenia. Approximately 50% of HIT patients initially recognized with isolated thrombocytopenia developed objective evidence for thrombosis during the subsequent 30-day period. Reprinted from Warkentin and Kelton89  with permission.

Close modal

In the 1990s, many physicians would diagnose HIT and immediately discontinue the heparin and anticoagulate with warfarin. But one patient who was managed this way, but dramatically worsened, led us to question this approach. In a case control study,93  we found that warfarin therapy in patients with HIT was often complicated by a dramatic progression of the DVT to distal extremity necrosis (venous gangrene), even when pulses were palpable. Some of these patients ultimately required limb amputation. Plasma samples from several patients demonstrated that warfarin therapy was associated with a severe reduction in the natural anticoagulant protein C, while at the same time the warfarin failed to inhibit the increased thrombin generation.93  One of the hallmarks of this syndrome was a dramatic increase in the INR to supratherapeutic levels (typically, greater than 4.0), which was explained by parallel reductions in factor VII. This syndrome, which has also been observed by other investigators,94,95  has led to the treatment principle that warfarin should be avoided during the acute (thrombocytopenic) phase of HIT.

Our current approach is to postpone warfarin therapy in a patient with acute HIT until there has been substantial resolution of the thrombocytopenia (platelet count rise to at least 150 × 109/L).65  Furthermore, careful overlap of the direct thrombin inhibitor (DTI) and warfarin, or of danaparoid/warfarin therapy, should occur, that is, starting with low (maintenance) doses of warfarin, ensuring at least a 5-day DTI/warfarin or danaparoid/warfarin overlap, and maintaining the DTI or danaparoid until the platelet count has reached a stable plateau within the normal range.65 

Two distinctly different direct thrombin inhibitors (DTIs) have been evaluated in prospective studies using historical controls.108-114  Lepirudin is a recombinant hirudin derivative and argatroban is a small, synthetic molecule. The strengths and weaknesses of these trials have been recently reviewed.115  The lepirudin studies108-111  had the diagnosis confirmed serologically in treated patients, whereas the argatroban studies112-114  relied on a clinical diagnosis of HIT (with approximately 65% showing positive antibodies). Both agents showed reductions in new thromboembolic events among patients with HIT-associated thrombosis (pooled analyses vs historical controls, relative risk [RR] of 0.28 and 0.45 for lepirudin and argatroban, respectively). Advantages of DTIs include their ability to inhibit thrombin rapidly, simple monitoring with the aPTT, and short half-life (in situations of bleeding or need for surgery). Potential drawbacks include difficulty judging adequacy of anticoagulation by aPTT in some circumstances, potential for rebound hypercoagulability if stopped prematurely, and drug accumulation in renal (lepirudin) or hepatobiliary (argatroban) dysfunction. Moreover, it is now recognized that for many patients, the manufacturer's recommended starting doses are too high.65  In addition, antihirudin antibodies have also been associated with acute anaphylaxis after lepirudin bolus.116 

In many countries (although not in the United States), danaparoid, a mixture of anticoagulant glycosaminoglycans, predominantly heparan, dermatan, and chondroitin sulfate, is available.65  Danaparoid has been shown to be more effect than dextran-70, ancrod, or coumadin102,106  Advantages of danaparoid include its long half-life, ability to monitor drug levels directly (via anti-Xa levels), and the theoretical disruption of PF4-containing immune complexes on platelet surfaces.105  Disadvantages include potential for cross-reactivity of HIT antibodies; however, the success rate is high even when the drug is given without prior testing for cross-reactivity.102,106  An additional challenge is the requirement for measuring anti-Xa levels to achieve optimal anticoagulent monitoring.

Anecdotal reports describe success with bivalirudin and fondaparinux as treatments for patients with HIT.117,118 

HIT will continue to be a significant problem for years to come. Although novel inhibitors to the coagulation cascade are currently being evaluated both in the laboratory and in clinical trials, it is likely that heparin will continue to be widely used because of its ease of use, short half-life, low cost, and ability to be neutralized, among other attributes. For certain surgical procedure such as cardiopulmonary bypass, there are no obvious alternative agents. But the understanding of HIT has led to improved treatment strategies, as well as the ability to prevent many cases. The increased use of progressively smaller heparin moieties will continue to reduce the risk of HIT. The recent introduction of the pentasaccharide fondaparinux offers the potential of an exceptionally low risk of HIT.

Many of the studies described in this report were supported by grants from the Heart and Stroke Foundation of Ontario.

Contribution: J.G.K. and T.E.W. cowrote the review.

Conflict-of-interest disclosure: T.E.W. is a consultant for GlaxoSmithKline, GTI Inc, and Organon Inc. (part of Schering-Plough), and has received support for research studies and speaker honoraria from GlaxoSmithKline and Organon, and speaker honoraria from Sanofi-Aventis. J.G.K. is a scientific advisor for GlaxoSmithKline.

Correspondence: John G. Kelton, MD, McMaster University, 1200 Main St West, Room 2E1, Hamilton, ON, L8N 3Z5, Canada; e-mail: keltonj@mcmaster.ca.

1
Howell
 
WH
Holt
 
E
Two new factors in blood coagulation: heparin and pro-antithrombin.
Am J Physiol
1918
, vol. 
47
 (pg. 
328
-
341
)
2
Crafoord
 
C
Preliminary report on post-operative treatment with heparin as a preventive of thrombosis.
Acta Chir Scand
1936
, vol. 
79
 (pg. 
407
-
426
)
3
Weismann
 
RE
Tobin
 
RW
Arterial embolism occurring during systemic heparin therapy.
Arch Surg
1958
, vol. 
76
 (pg. 
219
-
225
)
4
Roberts
 
B
Rosato
 
FE
Rosato
 
EF
Heparin: a cause of arterial emboli?
Surgery
1963
, vol. 
55
 (pg. 
803
-
808
)
5
Barker
 
CF
Rosato
 
FE
Roberts
 
B
Peripheral arterial embolism.
Surg Gynecol Obstet
1966
, vol. 
123
 (pg. 
22
-
26
)
6
Kaupp
 
HA
Roberts
 
B
Arterial embolization during subcutaneous heparin therapy: case report.
J Cardiovasc Surg
1982
, vol. 
13
 (pg. 
210
-
212
)
7
Natelson
 
EA
Lynch
 
EC
Alfrey
 
CP
Gross
 
JB
Heparin-induced thrombocytopenia. An unexpected response to treatment of consumption coagulopathy.
Ann Intern Med
1969
, vol. 
71
 (pg. 
1121
-
1125
)
8
Rhodes
 
GR
Dixon
 
RH
Silver
 
D
Heparin induced thrombocytopenia with thrombotic and hemorrhagic manifestations.
Surg Gynecol Obstet
1973
, vol. 
136
 (pg. 
409
-
416
)
9
Rhodes
 
GR
Dixon
 
RH
Silver
 
D
Heparin induced thrombocytopenia: eight cases with thrombotic-hemorrhagic complications.
Ann Surg
1977
, vol. 
186
 (pg. 
752
-
758
)
10
Fratantoni
 
JC
Pollet
 
R
Gralnick
 
HR
Heparin-induced thrombocytopenia: confirmation of diagnosis with in vitro methods.
Blood
1975
, vol. 
45
 (pg. 
395
-
401
)
11
Babcock
 
RB
Dumper
 
CW
Scharfman
 
WB
Heparin-induced thrombocytopenia.
N Engl J Med
1976
, vol. 
295
 (pg. 
237
-
241
)
12
Green
 
D
Harris
 
K
Reynolds
 
N
Roberts
 
M
Patterson
 
R
Heparin immune thrombocytopenia: evidence for a heparin-platelet complex as the antigenic determinant.
J Lab Clin Med
1978
, vol. 
91
 (pg. 
167
-
175
)
13
Nelson
 
JC
Lerner
 
RG
Goldstein
 
R
Cagin
 
NA
Heparin-induced thrombocytopenia.
Arch Intern Med
1978
, vol. 
138
 (pg. 
548
-
552
)
14
Trowbridge
 
AA
Caraveo
 
J
Green
 
JB
Amaral
 
B
Stone
 
MJ
Heparin-related immune thrombocytopenia: studies of antibody-heparin specificity.
Am J Med
1978
, vol. 
65
 (pg. 
277
-
283
)
15
Cimo
 
PL
Moake
 
JL
Weinger
 
RS
Ben-Menachem
 
Y
Khalil
 
KG
Heparin-induced thrombocytopenia: association with a platelet aggregating factor and arterial thrombosis.
Am J Hematol
1979
, vol. 
6
 (pg. 
125
-
133
)
16
Hussey
 
CV
Bernhard
 
VM
McLean
 
MR
Fobian
 
JE
Heparin induced platelet aggregation: In vitro confirmation of thrombotic complications associated with heparin therapy.
Ann Clin Lab Sci
1979
, vol. 
9
 (pg. 
487
-
493
)
17
Chong
 
BH
Grace
 
CS
Rozenberg
 
MC
Heparin-induced thrombocytopenia: effect of heparin platelet antibody on platelets.
Br J Haematol
1981
, vol. 
49
 (pg. 
531
-
540
)
18
Powers
 
PJ
Kelton
 
JG
Carter
 
CJ
Studies on the frequency of heparin-associated thrombocytopenia.
Thromb Res
1984
, vol. 
33
 (pg. 
439
-
443
)
19
Cines
 
DB
Kaywin
 
P
Bina
 
M
Tomaski
 
A
Schreiber
 
AD
Heparin-associated thrombocytopenia.
N Engl J Med
1980
, vol. 
303
 (pg. 
788
-
795
)
20
King
 
DJ
Kelton
 
JG
Heparin-associated thrombocytopenia.
Ann Intern Med
1984
, vol. 
100
 (pg. 
535
-
540
)
21
Bell
 
WR
Tomasulo
 
PA
Alving
 
BM
Duffy
 
TP
Thrombocytopenia occurring during the administration of heparin.
Ann Intern Med
1976
, vol. 
85
 (pg. 
155
-
160
)
22
Alving
 
BM
Shulman
 
NR
Bell
 
WR
Evatt
 
BL
Tack
 
KM
In vitro studies of heparin-associated thrombocytopenia.
Thromb Res
1977
, vol. 
11
 (pg. 
827
-
834
)
23
Bell
 
WR
Heparin-associated thrombocytopenia and thrombosis.
J Lab Clin Med
1988
, vol. 
111
 (pg. 
600
-
605
)
24
Bell
 
WR
Royall
 
RM
Heparin-associated thrombocytopenia: a comparison of three heparin preparations.
N Engl J Med
1980
, vol. 
303
 (pg. 
902
-
907
)
25
Gollub
 
S
Ulin
 
AW
Heparin-induced thrombocytopenia in man.
J Lab Clin Med
1962
, vol. 
59
 (pg. 
430
-
435
)
26
Chong
 
BH
Pitney
 
WR
Castaldi
 
PA
Heparin-induced thrombocytopenia: association of thrombotic complications with heparin-dependent IgG antibody that induces thromboxane synthesis and platelet aggregation.
Lancet
1982
, vol. 
2
 (pg. 
1246
-
1249
)
27
Chong
 
BH
Berndt
 
MC
Heparin-induced thrombocytopenia.
Blut
1989
, vol. 
58
 (pg. 
53
-
57
)
28
Kelton
 
JG
Meltzer
 
D
Moore
 
J
, et al. 
Drug-induced thrombocytopenia is associated with increased binding of IgG to platelets both in vivo and in vitro.
Blood
1981
, vol. 
58
 (pg. 
524
-
529
)
29
Kelton
 
JG
Powers
 
PJ
Carter
 
CJ
A prospective study of the usefulness of the measurement of platelet-associated IgG for the diagnosis of idiopathic thrombocytopenic purpura.
Blood
1982
, vol. 
60
 (pg. 
1050
-
1053
)
30
Kelton
 
JG
Sheridan
 
D
Brain
 
H
Powers
 
PJ
Turpie
 
AGG
Carter
 
CJ
Clinical usefulness of testing for a heparin-dependent platelet-aggregating factor in patients with suspected heparin-associated thrombocytopenia.
J Lab Clin Med
1984
, vol. 
103
 (pg. 
606
-
612
)
31
Sheridan
 
D
Carter
 
C
Kelton
 
JG
A diagnostic test for heparin-induced thrombocytopenia.
Blood
1986
, vol. 
67
 (pg. 
27
-
30
)
32
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
, vol. 
120
 (pg. 
371
-
379
)
33
Kelton
 
JG
Sheridan
 
D
Santos
 
A
, et al. 
Heparin-induced thrombocytopenia: laboratory studies.
Blood
1988
, vol. 
72
 (pg. 
925
-
930
)
34
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
, vol. 
96
 (pg. 
182
-
187
)
35
Horsewood
 
P
Hayward
 
CP
Warkentin
 
TE
, et al. 
Investigation of the mechanisms of monoclonal antibody-induced platelet activation.
Blood
1991
, vol. 
78
 (pg. 
1019
-
1026
)
36
Chong
 
BH
Fawaz
 
I
Chesterman
 
CN
Berndt
 
MC
Heparin-induced thrombocytopenia: mechanism of interaction of the heparin-dependent antibody with platelets.
Br J Haematol
1989
, vol. 
73
 (pg. 
235
-
240
)
37
Karas
 
SP
Rosse
 
WF
Kurlander
 
RJ
Characterization of the IgG-Fc receptor on human platelets.
Blood
1982
, vol. 
60
 (pg. 
1277
-
1282
)
38
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
, vol. 
76
 (pg. 
2317
-
2322
)
39
Kelton
 
JG
Smith
 
JW
Santos
 
AV
Murphy
 
WG
Horsewood
 
P
Platelet IgG Fc receptor.
Am J Hematol
1987
, vol. 
25
 (pg. 
299
-
310
)
40
Warkentin
 
TE
An overview of the heparin-induced thrombocytopenia syndrome.
Semin Thromb Hemost
2004
, vol. 
30
 (pg. 
273
-
283
)
41
Cines
 
DB
Tomaski
 
A
Tannenbaum
 
S
Immune endothelial-cell injury in heparin-associated thrombocytopenia.
N Engl J Med
1987
, vol. 
316
 (pg. 
581
-
589
)
42
Visentin
 
GP
Ford
 
SE
Scott
 
JP
Aster
 
RH
Antibodies from patients with heparin-induced thrombocytopenia/thrombosis are specific for platelet factor 4 complexed with heparin or bound to endothelial cells.
J Clin Invest
1994
, vol. 
93
 (pg. 
81
-
88
)
43
Pouplard
 
C
Iochmann
 
S
Renard
 
B
, et al. 
Induction of monocyte tissue factor expression by antibodies to heparin-platelet factor 4 complexes developed in heparin-induced thrombocytopenia.
Blood
2001
, vol. 
97
 (pg. 
3300
-
3302
)
44
Arepally
 
GM
Mayer
 
IM
Antibodies from patients with heparin-induced thrombocytopenia stimulate monocytic cells to express tissue factor and secrete interleukin-8.
Blood
2001
, vol. 
98
 (pg. 
1252
-
1254
)
45
Warkentin
 
TE
Hayward
 
CP
Boshkov
 
LK
, et al. 
Sera from patients with heparin-induced thrombocytopenia generate platelet-derived microparticles with procoagulant activity: an explanation for the thrombotic complications of heparin-induced thrombocytopenia.
Blood
1994
, vol. 
84
 (pg. 
3691
-
3699
)
46
Warkentin
 
TE
Sheppard
 
JI
Generation of platelet-derived microparticles and procoagulant activity by heparin-induced thrombocytopenia IgG/serum and other IgG platelet agonists: a comparison with standard platelet agonists.
Platelets
1999
, vol. 
10
 (pg. 
319
-
326
)
47
Lee
 
DH
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
, vol. 
95
 (pg. 
724
-
731
)
48
Hughes
 
M
Hayward
 
CP
Warkentin
 
TE
Horsewood
 
P
Chorneyko
 
KA
Kelton
 
JG
Morphological analysis of microparticle generation in heparin-induced thrombocytopenia.
Blood
2000
, vol. 
96
 (pg. 
188
-
194
)
49
Amiral
 
J
Bridey
 
F
Dreyfus
 
M
, et al. 
Platelet factor 4 complexed to heparin is the target for antibodies generated in heparin-induced thrombocytopenia.
Thromb Haemost
1992
, vol. 
68
 (pg. 
95
-
96
)
50
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
, vol. 
83
 (pg. 
3232
-
3239
)
51
Greinacher
 
A
Pötzsch
 
B
Amiral
 
J
Dummel
 
V
Eichner
 
A
Mueller-Eckhardt
 
C
Heparin-associated thrombocytopenia: isolation of the antibody and characterization of a multimolecular PF4-heparin complex as the major antigen.
Thromb Haemost
1994
, vol. 
71
 (pg. 
247
-
251
)
52
Amiral
 
J
Bridey
 
F
Wolf
 
M
, et al. 
Antibodies to macromolecular platelet factor 4-heparin complexes in heparin-induced thrombocytopenia: a study of 44 cases.
Thromb Haemost
1995
, vol. 
73
 (pg. 
21
-
28
)
53
Poncz
 
M
Surrey
 
S
LaRocco
 
P
, et al. 
Cloning and characterization of platelet factor 4 cDNA derived from a human erythroleukemic cell line.
Blood
1987
, vol. 
69
 (pg. 
219
-
223
)
54
Horsewood
 
P
Warkentin
 
TE
Hayward
 
CP
Kelton
 
JG
The epitope specificity of heparin-induced thrombocytopenia.
Br J Haematol
1996
, vol. 
95
 (pg. 
161
-
167
)
55
Suh
 
JS
Aster
 
RH
Visentin
 
GP
Antibodies from patients with heparin-induced thrombocytopenia/thrombosis recognize different epitopes on heparin: platelet factor 4.
Blood
1998
, vol. 
91
 (pg. 
916
-
922
)
56
Ziporen
 
L
Li
 
ZQ
Park
 
KS
, et al. 
Defining an antigenic epitope on platelet factor 4 associated with heparin-induced thrombocytopenia.
Blood
1998
, vol. 
92
 (pg. 
3250
-
3259
)
57
Li
 
ZQ
Liu
 
W
Park
 
KS
, et al. 
Defining a second epitope for heparin-induced thrombocytopenia/thrombosis antibodies using KKO, a murine HIT-like monoclonal antibody.
Blood
2002
, vol. 
99
 (pg. 
1230
-
1236
)
58
Greinacher
 
A
Gopinadhan
 
M
Guenther
 
JU
, et al. 
Close approximation of two platelet factor 4 tetramers by charge neutralization forms the antigens recognized by HIT antibodies.
Arterioscler Thromb Vasc Biol
2006
, vol. 
26
 (pg. 
2386
-
2393
)
59
Amiral
 
J
Marfaing-Koka
 
A
Wolf
 
M
, et al. 
Presence of autoantibodies to interleukin-8 or neutrophil-activating peptide-2 in patients with heparin-associated thrombocytopenia.
Blood
1996
, vol. 
88
 (pg. 
410
-
416
)
60
Greinacher
 
A
Juhl
 
D
Strobel
 
U
, et al. 
Heparin-induced thrombocytopenia: a prospective study on the incidence, platelet-activating capacity and clinical significance of anti-PF4/heparin antibodies of the IgG, IgM, and IgA classes.
J Thromb Haemost
2007
, vol. 
5
 (pg. 
1666
-
1673
)
61
Denomme
 
GA
Warkentin
 
TE
Horsewood
 
P
Sheppard
 
JI
Warner
 
MN
Kelton
 
JG
Activation of platelets by sera containing IgG1 heparin-dependent antibodies: an explanation for the predominance of the FcγRIIa “low responder” (his131) gene in patients with heparin-induced thrombocytopenia.
J Lab Clin Med
1997
, vol. 
130
 (pg. 
278
-
284
)
62
Lindhoff-Last
 
E
Gerdsen
 
F
Ackermann
 
H
Bauersachs
 
R
Determination of heparin-platelet factor 4-IgG antibodies improves diagnosis of heparin-induced thrombocytopenia.
Br J Haematol
2001
, vol. 
113
 (pg. 
886
-
890
)
63
Warkentin
 
TE
Sheppard
 
JI
Moore
 
JC
Moore
 
KM
Sigouin
 
CS
Kelton
 
JG
Laboratory testing for the antibodies that cause heparin-induced thrombocytopenia: how much class do we need?
J Lab Clin Med
2005
, vol. 
146
 (pg. 
341
-
346
)
64
Warkentin
 
TE
Kelton
 
JG
Temporal aspects of heparin-induced thrombocytopenia.
N Engl J Med
2001
, vol. 
344
 (pg. 
1286
-
1292
)
65
Warkentin
 
TE
Greinacher
 
A
Koster
 
A
Lincoff
 
AM
Treatment and prevention of heparin-induced thrombocytopenia: ACCP evidence-based clinical practice guidelines.
Chest
2008
, vol. 
133
 
suppl
(pg. 
340S
-
380S
)
66
Greinacher
 
A
Alban
 
S
Dummel
 
V
, et al. 
Characterization of the structural requirements for a carbohydrate based anticoagulant with a reduced risk of inducing the immunological type of heparin-associated thrombocytopenia.
Thromb Haemost
1995
, vol. 
74
 (pg. 
886
-
892
)
67
Visentin
 
GP
Moghaddam
 
M
Beery
 
SE
McFarland
 
JG
Aster
 
RH
Heparin is not required for detection of antibodies associated with heparin-induced thrombocytopenia thrombosis.
J Lab Clin Med
2001
, vol. 
138
 (pg. 
22
-
31
)
68
Warkentin
 
TE
Levine
 
MN
Hirsh
 
J
, et al. 
Heparin-induced thrombocytopenia in patients treated with low-molecular-weight heparin or unfractionated heparin.
N Engl J Med
1995
, vol. 
332
 (pg. 
1330
-
1335
)
69
Warkentin
 
TE
Cook
 
RJ
Marder
 
VJ
, et al. 
Anti-platelet factor 4/heparin antibodies in orthopedic surgery patients receiving antithrombotic prophylaxis with fondaparinux or enoxaparin.
Blood
2005
, vol. 
106
 (pg. 
3791
-
3796
)
70
Warkentin
 
TE
Maurer
 
BT
Aster
 
RH
Heparin-induced thrombocytopenia associated with fondaparinux.
N Engl J Med
2007
, vol. 
356
 (pg. 
2653
-
2654
)
71
Rota
 
E
Bazzan
 
M
Fantino
 
G
Fondaparinux-related thrombocytopenia in a previous low-molecular-weight heparin (LMWH)-induced thrombocytopenia (HIT).
Thromb Haemost
2008
, vol. 
99
 (pg. 
779
-
781
)
72
Warkentin
 
TE
Sheppard
 
JA
Horsewood
 
P
Simpson
 
PJ
Moore
 
JC
Kelton
 
JG
Impact of the patient population on the risk for heparin-induced thrombocytopenia.
Blood
2000
, vol. 
96
 (pg. 
1703
-
1708
)
73
Chong
 
BH
Pilgrim
 
RL
Cooley
 
MA
Chesterman
 
CN
Increased expression of platelet IgG Fc receptors in immune heparin-induced thrombocytopenia.
Blood
1993
, vol. 
81
 (pg. 
988
-
993
)
74
Rauova
 
L
Poncz
 
M
McKenzie
 
SE
, et al. 
Ultralarge complexes of PF4 and heparin are central to the pathogenesis of heparin-induced thrombocytopenia.
Blood
2005
, vol. 
105
 (pg. 
131
-
138
)
75
Rauova
 
L
Zhai
 
L
Kowalska
 
MA
Arepally
 
GM
Cines
 
DB
Poncz
 
M
Role of platelet surface PF4 antigenic complexes in heparin-induced thrombocytopenia pathogenesis: diagnostic and therapeutic implications.
Blood
2006
, vol. 
107
 (pg. 
2346
-
2353
)
76
Lambert
 
MP
Rauova
 
L
Bailey
 
M
Sola-Visner
 
MC
Kowalska
 
MA
Poncz
 
M
Platelet factor 4 is a negative autocrine in vivo regulator of megakaryocytopoiesis: clinical and therapeutic implications.
Blood
2007
, vol. 
110
 (pg. 
1153
-
1160
)
77
Warkentin
 
TE
HIT: lessons learned.
Pathophysiol Haemost Thromb
2006
, vol. 
35
 (pg. 
50
-
57
)
78
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
, vol. 
1
 (pg. 
39
-
42
)
79
Bennett-Guerrero
 
E
Slaughter
 
TF
White
 
WD
, et al. 
Preoperative anti-PF4/heparin antibody level predicts adverse outcome after cardiac surgery.
J Thorac Cardiovasc Surg
2005
, vol. 
130
 (pg. 
1567
-
1572
)
80
Carrier
 
M
Rodger
 
MA
Fergusson
 
D
, et al. 
Increased mortality in hemodialysis patients having specific antibodies to the platelet factor 4-heparin complex.
Kidney Int
2008
, vol. 
73
 (pg. 
213
-
219
)
81
Arnold
 
DM
Kelton
 
JK
Heparin-induced thrombocytopenia: an iceberg rising (editorial).
Mayo Clin Proc
2005
, vol. 
80
 (pg. 
988
-
990
)
82
Reilly
 
MP
Taylor
 
SM
Hartman
 
NK
, et al. 
Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through FcγRIIA.
Blood
2001
, vol. 
98
 (pg. 
2442
-
2447
)
83
Arepally
 
GM
Kamei
 
S
Park
 
KS
, et al. 
Characterization of a murine monoclonal antibody that mimics heparin-induced thrombocytopenia antibodies.
Blood
2000
, vol. 
95
 (pg. 
1533
-
1540
)
84
Reilly
 
MP
Taylor
 
SM
Franklin
 
C
, et al. 
Prothrombotic factors enhance heparin-induced thrombocytopenia and thrombosis in vivo in a mouse model.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
2687
-
2694
)
85
Suvarna
 
S
Espinasse
 
B
Qi
 
R
, et al. 
Determinants of PF4/heparin immunogenicity.
Blood
2007
, vol. 
110
 (pg. 
4253
-
4260
)
86
Warkentin
 
TE
Drug-induced immune-mediated thrombocytopenia—from purpura to thrombosis.
N Engl J Med
2007
, vol. 
356
 (pg. 
891
-
893
)
87
Warkentin
 
TE
Roberts
 
RS
Hirsh
 
J
Kelton
 
JG
An improved definition of immune heparin-induced thrombocytopenia in postoperative orthopedic patients.
Arch Intern Med
2003
, vol. 
163
 (pg. 
2518
-
2524
)
88
Boshkov
 
LK
Warkentin
 
TE
Hayward
 
CPM
Andrew
 
M
Kelton
 
JG
Heparin-induced thrombocytopenia and thrombosis: clinical and laboratory studies.
Br J Haematol
1993
, vol. 
84
 (pg. 
322
-
328
)
89
Warkentin
 
TE
Kelton
 
JG
A 14-year study of heparin-induced thrombocytopenia.
Am J Med
1996
, vol. 
101
 (pg. 
502
-
507
)
90
Greinacher
 
A
Farner
 
B
Kroll
 
H
Kohlmann
 
T
Warkentin
 
TE
Eichler
 
P
Clinical features of heparin-induced thrombocytopenia including risk factors for thrombosis. A retrospective analysis of 408 patients.
Thromb Haemost
2005
, vol. 
94
 (pg. 
132
-
135
)
91
Hong
 
AP
Cook
 
DJ
Sigouin
 
CS
Warkentin
 
TE
Central venous catheters and upper-extremity deep-vein thrombosis complicating immune heparin-induced thrombocytopenia.
Blood
2003
, vol. 
101
 (pg. 
3049
-
3051
)
92
Lee
 
DH
Warkentin
 
TE
Denomme
 
GA
Lagrotteria
 
DD
Kelton
 
JG
Factor V Leiden and thrombotic complications in heparin-induced thrombocytopenia.
Thromb Haemost
1998
, vol. 
79
 (pg. 
50
-
53
)
93
Warkentin
 
TE
Elavathil
 
LJ
Hayward
 
CP
Johnston
 
MA
Russett
 
JI
Kelton
 
JG
The pathogenesis of venous limb gangrene associated with heparin-induced thrombocytopenia.
Ann Intern Med
1997
, vol. 
127
 (pg. 
804
-
812
)
94
Srinivasan
 
AF
Rice
 
L
Bartholomew
 
JR
, et al. 
Warfarin-induced skin necrosis and venous limb gangrene in the setting of heparin-induced thrombocytopenia.
Arch Intern Med
2004
, vol. 
164
 (pg. 
66
-
70
)
95
Smythe
 
MA
Warkentin
 
TE
Stephens
 
JL
Zakalik
 
D
Mattson
 
JC
Venous limb gangrene during overlapping therapy with warfarin and a direct thrombin inhibitor for immune heparin-induced thrombocytopenia.
Am J Hematol
2002
, vol. 
71
 (pg. 
50
-
52
)
96
Warkentin
 
TE
Bernstein
 
RA
Delayed-onset heparin-induced thrombocytopenia and cerebral thrombosis after a single administration of unfractionated heparin.
N Engl J Med
2003
, vol. 
348
 (pg. 
1067
-
1069
)
97
Warkentin
 
TE
Kelton
 
JG
Delayed-onset heparin-induced thrombocytopenia and thrombosis.
Ann Intern Med
2001
, vol. 
135
 (pg. 
502
-
506
)
98
Rice
 
L
Attisha
 
WK
Drexler
 
A
Francis
 
JL
Delayed-onset heparin-induced thrombocytopenia.
Ann Intern Med
2002
, vol. 
136
 (pg. 
210
-
215
)
99
Prechel
 
MM
McDonald
 
MK
Jeske
 
WP
Messmore
 
HL
Walenga
 
JM
Activation of platelets by heparin-induced thrombocytopenia antibodies in the serotonin release assay is not dependent on the presence of heparin.
J Thromb Haemost
2005
, vol. 
3
 (pg. 
2168
-
2175
)
100
Warkentin
 
TE
Chong
 
BH
Greinacher
 
A
Heparin-induced thrombocytopenia: towards consensus.
Thromb Haemost
1998
, vol. 
79
 (pg. 
1
-
7
)
101
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
, vol. 
106
 (pg. 
629
-
635
)
102
Lubenow
 
N
Warkentin
 
TE
Greinacher
 
A
, et al. 
Results of a systematic evaluation of treatment outcomes for heparin-induced thrombocytopenia in patients receiving danaparoid, ancrod, and/or coumarin explain the rapid shift in clinical practice during the 1990s.
Thromb Res
2006
, vol. 
117
 (pg. 
507
-
515
)
103
Demers
 
C
Ginsberg
 
JS
Brill-Edwards
 
P
, et al. 
Rapid anticoagulation using ancrod for heparin-induced thrombocytopenia.
Blood
1991
, vol. 
78
 (pg. 
2194
-
2197
)
104
Selleng
 
K
Selleng
 
S
Raschke
 
R
, et al. 
Immune heparin-induced thrombocytopenia can occur in patients receiving clopidogrel and aspirin.
Am J Hematol
2005
, vol. 
78
 (pg. 
188
-
192
)
105
Chong
 
BH
Ismail
 
F
Cade
 
J
Gallus
 
AS
Gordon
 
S
Chesterman
 
CN
Heparin-induced thrombocytopenia: studies with a new low molecular weight heparinoid, Org 10172.
Blood
1989
, vol. 
73
 (pg. 
1592
-
1596
)
106
Chong
 
BH
Gallus
 
AS
Cade
 
JF
, et al. 
Prospective randomised open-label comparison of danaparoid and dextran 70 in the treatment of heparin-induced thrombocytopenia and thrombosis.
Thromb Haemost
2001
, vol. 
86
 (pg. 
1170
-
1175
)
107
Lo
 
GK
Sigouin
 
CS
Warkentin
 
TE
What is the potential for overdiagnosis of heparin-induced thrombocytopenia?
Am J Hematol
2007
, vol. 
82
 (pg. 
1037
-
1043
)
108
Greinacher
 
A
Völpel
 
H
Janssens
 
U
, et al. 
Recombinant hirudin (lepirudin) provides safe and effective anticoagulation in patients with heparin-induced thrombocytopenia: a prospective study.
Circulation
1999
, vol. 
99
 (pg. 
73
-
80
)
109
Greinacher
 
A
Janssens
 
U
Berg
 
G
, et al. 
Lepirudin (recombinant hirudin) for parenteral anticoagulation in patients with heparin-induced thrombocytopenia: Heparin-Associated Thrombocytopenia Study (HAT) investigators.
Circulation
1999
, vol. 
100
 (pg. 
587
-
593
)
110
Greinacher
 
A
Eichler
 
P
Lubenow
 
N
Kwasny
 
H
Luz
 
H
Heparin-induced thrombocytopenia with thromboembolic complications: meta-analysis of 2 prospective trials to assess the value of parenteral treatment with lepirudin and its therapeutic aPTT range.
Blood
2000
, vol. 
96
 (pg. 
846
-
851
)
111
Lubenow
 
N
Eichler
 
P
Lietz
 
T
Geinacher
 
A
and the HIT Investigators Group
Lepirudin in patients with heparin-induced thrombocytopenia—results of the third prospective study (HAT-3) and a combined analysis of HAT-1, HAT-2, and HAT-3.
J Thromb Haemost
2005
, vol. 
3
 (pg. 
2428
-
2436
)
112
Lewis
 
BE
Wallis
 
DE
Berkowitz
 
SD
, et al. 
Argatroban anticoagulant therapy in patients with heparin-induced thrombocytopenia.
Circulation
2001
, vol. 
103
 (pg. 
1838
-
1843
)
113
Lewis
 
BE
Wallis
 
DE
Leya
 
F
Hursting
 
MJ
Kelton
 
JG
Argatroban anticoagulation in patients with heparin-induced thrombocytopenia.
Arch Intern Med
2003
, vol. 
163
 (pg. 
1849
-
1856
)
114
Lewis
 
BE
Wallis
 
DE
Hursting
 
MJ
Levine
 
RL
Leya
 
F
Effects of argatroban therapy, demographic variables, and platelet count on thrombotic risks in heparin-induced thrombocytopenia.
Chest
2006
, vol. 
129
 (pg. 
1407
-
1416
)
115
Hirsh
 
J
Heddle
 
N
Kelton
 
JG
Treatment of heparin-induced thrombocytopenia: a critical review.
Arch Intern Med
2004
, vol. 
164
 (pg. 
361
-
369
)
116
Greinacher
 
A
Lubenow
 
N
Eichler
 
P
Anaphylactic and anaphylactoid reactions associated with lepirudin in patients with heparin-induced thrombocytopenia.
Circulation
2003
, vol. 
108
 (pg. 
2062
-
2065
)
117
Dang
 
CH
Durkalski
 
VL
Nappi
 
JM
Evaluation of treatment with direct thrombin inhibitors in patients with heparin-induced thrombocytopenia.
Pharmacotherapy
2006
, vol. 
26
 (pg. 
461
-
468
)
118
Lobo
 
B
Finch
 
C
Howard
 
A
Minhas
 
S
Fondaparinux for the treatment of patients with acute heparin-induced thrombocytopenia.
Thromb Haemost
2008
, vol. 
99
 (pg. 
208
-
214
)
Sign in via your Institution