Open access peer-reviewed chapter

Not Only Heparin but Also Antibody Induces Thrombocytopenia

Written By

Thi-Huong Nguyen

Submitted: 15 July 2017 Reviewed: 08 January 2018 Published: 07 March 2018

DOI: 10.5772/intechopen.73576

From the Edited Volume

Thrombocytopenia

Edited by Pankaj Abrol

Chapter metrics overview

1,335 Chapter Downloads

View Full Metrics

Abstract

In the last two decades, heparin was widely used as an anticoagulant. Besides numerous advantages of heparin, some patients with heparin administration suffer from a side effect, the so-called heparin-induced thrombocytopenia (HIT), which can result in thromboses such as deep vein thrombosis, pulmonary embolism, occlusion of a limb artery, acute myocardial infarct, stroke, and a systemic reaction or skin necrosis. The basic on HIT complication have been investigated and led to clinical insights. Recent studies provided detail mechanisms among binding partners in HIT; especially, it has been shown that not only heparin but also a subset of antibody induce thrombocytopenia. In this chapter, insights into both heparin- and antibody-induced thrombocytopenia will be discussed and the novel mechanism of the autoimmune HIT caused by a subset of antibodies will be introduced.

Keywords

  • heparin-induced thrombocytopenia
  • HIT
  • mechanism
  • binding force
  • PF4
  • antibody

1. Introduction

Heparin-induced thrombocytopenia (HIT) as a severe adverse drug effect occurs when patients receive heparin anticoagulant to prevent and treat thromboembolic diseases. Depending on the length of heparin, HIT occurs in ≤5% of patients receiving high molecular weight unfractionated heparin, whereas ≤1% of patients receiving low molecular weight heparin. In HIT, the immune system considers the platelet factor 4 (PF4), which is altered in its conformation after binding to heparin (H), to be “foreign” and the formation of anti-PF4/H antibodies (aPF4/P Abs) occurs. Upon binding to the PF4/H complex, these antibodies activate circulating platelets and other cells. Typically, 5–14 days after heparin exposure, platelet count reduces to <15–20 × 109 cells/L (or a > 50% decrease in platelet count). HIT can result in thromboses such as deep vein thrombosis (DVT), pulmonary embolism (PE), occlusion of a limb artery, acute myocardial infarct, stroke, and a systemic reaction or skin necrosis. Importantly, there is also a subset of anti-PF4/Heparin antibodies (aPF4/H Abs) which, in the absence of heparin, can lead to symptomatic thrombocytopenia and excessive vascular thrombosis. The extreme sequela of the aPF4/H Abs is autoimmune HIT, in which individuals develop multiple vessel occlusions without drug exposure.

Advertisement

2. Heparin-induced thrombocytopenia

Heparin-induced thrombocytopenia (HIT) is a distinct clinicopathologic syndrome caused by platelet-activating antibodies that bridge between complexes of platelet factor 4-Heparin (PF4/H) and platelets [1, 2] or endothelial cells [3]. Human platelets are anuclear cell fragments with discoidal shapes of 1–2 μm, originating from the cytoplasm of bone marrow megakaryocytes [4]. Platelets store PF4 (a positively charged tetramer belonging to CXC chemokine family) in their alpha granules. Non-activated platelets release some PF4s (Figure 1A) [5]. When patients take anticoagulant polyanions like heparin, some of these heparins bind to PF4s forming ultra large PF4/H complexes (Figure 1B). Binding of heparin to PF4 induces a conformational change in PF4s [6, 7, 8] which results in an expression of new epitopes. Some patients develop antibodies against these neoepitopes (Figure 1B). These human-derived antibodies are defined as anti-PF4/H antibodies (aPF4/P Abs). Each resulting multimolecular complex of an aPF4/P Ab to a PF4/H complex contains two platelet binding sites, that is, one is on the PF4/H complexes, and another one is on the Fc part of the IgG which binds to FcγRIIa receptors [9, 10] on platelet membranes (Figure 1C). Cross-linking of the platelet Fc receptor results in platelet activation that releases more PF4s and facilitates formation additional ultra large immune complexes. These complexes rapidly recruit other platelets into the prothrombotic process (Figure 1D). Activation of platelets leads to the loss of platelets, massive platelet activation and even triggers clotting cascade that results in thrombin generation and increases the risk for vessel occlusions such as venous thrombosis, myocardial infarction or stroke [7, 11, 12]. The binding strength of a blood thrombus has major biological importance. A recent study could determine directly the binding strength between two platelets at single platelet level [13]. The binding force increases proportionally to the degree of platelet activation but reduces with blockade of specific platelet receptors. The method provides major perspectives for testing and improving the biocompatibility of new materials, quantifying the effect of drugs on platelet function, and assessing the mechanical characteristics of acquired/inherited platelet defects.

Figure 1.

Cartoon illustrates the formation of heparin-induced thrombocytopenia (HIT). (A) Non-activated platelets secrete several PF4s. (B) with heparin exposure, PF4s form ultra large complexes with long heparins that induce conformational changes in PF4s. Some patients develop aPF4/H Abs against PF4 neoepitopes. (C) Human-derived aPF4/H Abs bound PF4/H complexes can adhere to platelet membrane. (D) Fc parts of the antibodies link fcγRIIa receptors on platelet membranes that leads to platelet aggregation/activation. Adapted from [5].

Heparins are the glycosaminoglycans (GAGs) containing glucosamine residues with a high degree of sulfation that dictates their biological activities [6, 14, 15]. GAGs play an important role in the sequestration of plasmodium falciparum-infected red blood cells in the microvascular endothelium of different tissues [16, 17]. Their pharmacologic activity is mediated by a chemically unique pentasaccharide sequence present in about 30% of all heparin molecules. Heparin behaves like simple entropic spring forces, which is produced by sugar rings of heparin flipping to more energetic and more extended conformations [18, 19]. Both low and high molecular weight heparins are available. The source of high molecular weight unfractionated heparin (UFH) influences the risk of HIT, i.e. bovine UFH is more likely to cause HIT than porcine UFH [20, 21, 22]. Besides UFH, the low molecular weight heparins (LMWH) produced from UFH by chemical fractionation, are widely used in clinical practice [23, 24, 25, 26, 27]. Due to their shorter chain length, LMWHs show less strong interaction with PF4. UFH and PF4 form ultra large complexes (ULCs) when both are present approximately at an optimal 1:1 ratio. Comparing with UFH, LMWHs form smaller complexes with PF4. ULCs showed a greater capacity to promote platelet activation than small complexes [28]. These differences in complex formation between UFH and LMWHs translate into their risk for inducing HIT in patients. LMWHs induce HIT about 10 times less frequent than UFH, but HIT still randomly occurs during treatment with LMWHs [29, 30, 31, 32].

2.1. Boundary between antigenic and non-antigenic heparin

PF4/heparin (or polyanions) complexes can become antigenic or not depend on heparin (or polyanion) characteristics. To expose neoepitopes on PF4s relevant for HIT, at least three bonds between the polyanion and PF4 in the PF4/polyanion complex should be formed [33]. These neoepitopes on the PF4/polyanion complexes then allow binding of the aPF4/H Abs. The binding strength of the single sulfate groups on the polyanion with the PF4 does not differ among polyanions with a different degree of sulfation [33]. The quantity and resulting density of sulfate groups on the polyanion chain determine their molecular effects on PF4 [33]. In particular, the polyanions which bind to PF4 tetramer with less than three sulfate bonds are unable to expose the neoepitope [6, 34]. The results suggest an existence of a boundary between antigenic (risk for HIT) and non-antigenic heparins (non-risk for HIT). This boundary has been determined by applying multiple techniques such as atomic force microscopy-based atomic force microscopy (AFS) [35], isothermal titration calorimetry (ITC) [6], or circular dichroism (CD) spectroscopy in combination with enzyme-linked immunosorbent assay (ELISA) [7] (Figure 2).

Figure 2.

Determination of the boundary between antigenic and non-antigenic heparins. (A) Rupture force histograms fitted by Gaussian distributions show narrow widths (green arrows) for heparins ≤6-mer (HO05, HO06) and wider widths for longer heparins ≥8-mer (HO08, HO012, HO016). (B) ITC demonstrates lower enthalpy for short heparins (black dotted box) and higher enthalpy for long heparins (red-dotted box), while a saturation is found at ~11-mer. (C) Combination of CD spectroscopy and EIA shows that a boundary between short and long heparins is at ~30% ß-sheet contents and OD ~0.5. Overall, the boundary is determined between 8- and 11-mer. Adapted from [8, 35, 48].

AFS shows that both numbers of specific rupture events and magnitude of rupture forces rise with an increase of heparin length, suggesting that long heparins form with PF4 more bonds than short ones [35]. A larger variation of the rupture forces for long heparins ≥8-mer compared with short heparins ≤6-mer was observed (Figure 3A). The enthalpy obtained by ITC rises with the increase of heparin length and reaches maximal values at ~11-mer (Figure 3B) [36]. Combining the results obtained by AFS and ITC, the boundary between non-antigenic and antigenic heparin is determined between 8- to 11-mer. This boundary is further clarified by CD spectroscopy which is sensitive to the secondary structure and folding properties of proteins [37]. For PF4/H interactions, the change in β-sheet content was found to be ≤30% for short heparin and >30% for long heparins (Figure 3C). By ELISA, optical density (OD) was ≤0.5 for short heparin, while OD was >0.5 for longer heparins (>8-mer) (Figure 3C). The OD of 0.5 is the threshold to determine whether a heparin used in the ELISA was able to support binding of aPF4/H Abs. The combination of β-sheet content and OD values show clearly a dissimilar behavior between short and long heparins (Figure 3C).

Figure 3.

Model describing different binding pathways between short and long heparins when interacting with PF4 tetramers. (A) Depending on heparin length, short heparin can bind to one PF4 tetramer, (B) whereas long heparin bridges two PF4s and forces them closer to each other at a distance l < L, merging two hydrophobic surfaces of PF4s (green shaded area). Adapted from [35].

Linking together all the results from AFS, ITC, CD spectroscopy and ELISA, the boundary between antigenic and non-antigenic heparin has been proved between 8- and 11-mer. These findings are particularly important to understand PF4-Heparin binding processes and to develop new heparin-derived drugs with reduced risk for adverse immune reactions. Combination of these techniques allows better characterizing heparin boundary.

2.2. Kinetic properties and binding model of PF4/H complexes

Thermodynamic and kinetic parameters of the ligand-receptor interactions can be obtained by applying the Bell-Evans [38] or the Friddle [39, 40] models. The models show that the faster the molecule is pulled, the higher the rupture force will be measured. For simple ligand-receptor interaction in which multiple interactions are not involved, the rupture force (F) increases proportionally to the logarithmic loading rate. Even though there is some variation in the parameters obtained by these two models, Bell-Evans model is still a powerful tool to determine the kinetics of ligand-receptor interactions [41, 42]. For the PF4/H system, the PF4 tetramer is considered as one antigen or the interaction between heparin and PF4 is formed by a single bond, and therefore, applicable to the Bell-Evans model [35]. Short heparins show higher koff values than long heparins, indicating that PF4/long heparin complexes are more stable than PF4/short heparin complexes (Table 1). With binding affinity (KA) measured by ITC [6], the thermal on-rate (kon = koff. KA) of PF4/H complexes is calculated. The short heparins bind to PF4s with ~10–20 times faster than long heparins [35].

Parameter HO06 HO12 HO16
koff (s−1) 1.64 1.40 × 10−2 1.10 × 10−4
kon(M−1 s−1) 0.41 × 105 0.32 × 104 0.55 × 103
ΔE (kBT) −0.49 4.27 9.12

Table 1.

Thermodynamic and kinetic parameters of PF4/heparin interactions [35].

PF4-Heparins interaction is more complex than general ligand-receptor interactions which are attributed to the electrostatic attraction. Based on special features in force-distance curves and the magnitude of PF4/H binding forces, it has been proved that long heparin bound PF4s creating additional PF4-PF4 bonds [35]. Long heparins form two types of bonds with PFs, i.e. (i) PF4-Heparin and (ii) PF4-PF4 bond, whereas short heparins form only one PF4-Heparin bond. Even though the concept of the PF4-PF4 bond, in general, cannot be accepted because PF4s are highly positive proteins, and therefore, strongly repel each other. However, when forming a complex with a highly negative charged heparin, the positive-charged PF4 is probably neutralized that results in a mergence of two hydrophobic PF4 surfaces [34]. Based on these findings, a model for PF4-heparin interaction has been proposed (Figure 3). Due to their sizes, the short heparins simply bind to a single PF4 tetramer (Figure 3A), whereas the long heparins neutralize positive charges on PF4 tetramers and switch the charges between two PF4 tetramers from a repulsion to an attraction. Heparin reacts as a catalyst that forces two PF4 molecules close to each other within a distance l (l < L), resulting in two merged hydrophobic PF4 surfaces (Figure 3B). This way of interacting results in the extremely stable PF4/H complexes, especially for long heparins.

A sequence in the formation of PF4/heparin complexes has been identified. When a long heparin comes closely to PF4s, heparin forms first bonds with positively charged clusters on PF4s and then it pulls closely PF4s together to form PF4-PF4 bonds [35].

Based on bond energy (ΔE), quantitative information of bond transitions can be calculated following the study of Wang et al. [43]. The bond transitions of short heparin from the weak positive-charged area on PF4 release energy, whereasPF4-PF4 bonds consume energy [35]. In contrast to short heparin, the bond transitions of long heparins in both cases release energy, while their interactions with the positively charged clusters consume energy (Table 1). Based on energy level, PF4-PF4 interaction is attributed to be stronger than the bonds between heparin and non-clusters of positive-charged areas on PF4. However, PF4-PF4 interaction is weaker than the interaction between heparin and clusters of positive charges on PF4.

Advertisement

3. Antibody-induced thrombocytopenia

Immunocomplexes composed of aPF4/P Abs and PF4/polyanion (PF4/P) complexes on the platelet surface induce platelet aggregation via cross-linking FcγRIIA receptors [9, 10]. They also bind to the surface of endothelial cells and monocytes [44, 45, 46], inducing procoagulant activity [44, 47]. Heparin-induced thrombocytopenia has been well understood. Recent studies reported that a subset of human-derived autoantibodies in some patients also can induce thrombocytopenia in a heparin-similar manner.

3.1. Human-derived HIT antibodies

All aPF4/P Abs bind to immobilized PF4/P complexes in ELISA [48], but only some of them activate platelets in functional assays, e.g. the heparin-induced platelet activation assay (HIPA) [48] or the serotonin release assay (SRA) [49, 50]. Human-derived aPF4/P Abs compose of three groups, i.e. the antibodies do not activate platelets in HIPA test (group-1 Abs); the antibodies activate platelets in HIPA but require heparin (group-2 Abs); the antibodies activate platelets even without heparin (group-3 Abs) (Figure 4). Group-3 Abs developed from patients who had clinical autoimmune HIT, and therefore, they are defined as ‘autoantibodies’ [51].

Figure 4.

Different reaction patterns of aPF4/H antibodies. (Right) pyramid shows antibodies of three groups, all positive in EIA. Group-1 (blue) do not activate platelets (HIPA -); many Abs belonging to group-2 do not induce HIT (yellow), some induce HIT (gold) and others induce HIT with thrombosis (dark red). Recent studies found an additional small subset of patient’s content autoimmune group-3 HIT Abs (red). (Left) visualization of platelet aggregates-induced by different antibody groups imaged by scanning electron microscopy in the presence (+) or absence (−) of heparin: Group-1 abs induce (bottom left) only small aggregates reflecting the background platelet activation; group-2 Abs (middle, left) cause large aggregates only in the presence of heparin; group-3 Abs induce large aggregates even in the absence of heparin. Same scale bar for all images. Adapted from [55].

3.1.1. Characteristics of human-derived HIT antibodies

In contrast to the detailed characterization of the PF4/polyanion complexes, little is known about the features of aPF4/H Abs in the pathogenesis of HIT. Exploring the characteristics of HIT antibodies bears a potential to better understand general mechanisms of antibody-mediated autoimmunity HIT. However, there is a difficulty in subtracting the pathogenic HIT antibody directly from human sera because both pathogenic and non-pathogenic antibodies bind to the PF4/H antigen.

Newman et al. reported that aPF4/P Abs can be purified by PF4-agarose beads [3]. Later in 2000, Amiral et al. described that affinity purification of aPF4/P Abs resulted in a mixture of IgA, IgM, and IgG [52]. In this mixture, only a subset of IgG antibodies activates platelets [49]. Contamination of IgA, IgM, and IgG antibodies will increase the difficulty in characterizing aPF4/P Abs. To overcome this limitation, two-step affinity chromatography has currently established to separate aPF4/H Abs from HIT patients sera. By this method, aPF4/P Abs from sera of patients were successfully isolated for three antibody groups. The purified Abs showed similar characteristics as the original serum in EIA and HIPA. Titrating the antibodies in ELISA, all antibody groups show an increase of OD with increasing antibody concentration (Figure 5A). OD values are highest for group-3, followed by group-2 and then group-1 Abs. In the HIPA test, group-1 Abs did not cause platelet aggregation up to a concentration of 89.7 μg/mL; group-2 Abs induced platelet aggregation from concentrations ≥43.5 μg/mL, but only in the presence of heparin; while group-3 Abs induced platelet aggregation from concentrations ≥5.2 μg/mL independently of heparin (Figure 5B). This is consistent with previous findings that chondroitin sulfate plays an important role in platelet activation by PF4/P Abs, even in the absence of heparin [53, 54].

Figure 5.

Dose-dependent binding of aPF4/P Abs to PF4/H complexes in EIA and HIPA. (A) EIA shows the lowest OD of control IgG (black) as the background reaction, follow by group-1 (dark cyan), higher for group-2 (blue) and highest for group-3 (red) Abs. (B) HIPA tests show a dependence of platelet aggregation on antibody concentration: Group-1 Abs do not activate platelets, neither in the absence (−), nor in the presence (+) of reviparin up to a concentration of 89.7 μg/mL (dark cyan); group-2 Abs (blue) induced platelet activation (red part) at concentrations ≥44 μg/mL but only in the presence of reviparin; group-3 Abs (red) activated platelets at much lower concentrations (≥5 μg/mL) either in the presence or absence of reviparin. n = 5 sera per group. Adapted from [55].

3.1.2. Binding strength of human-derived HIT antibodies

The binding strength between the antibody and PF4/H complexes is determined by AFS. A single aPF4/H Abs is immobilized on the cantilever and then approach to the PF4/H complexes coated on a solid phase for interacting and measuring of their binding strength. Weakest binding forces were measured for monoclonal antibody KKO mimicking human HIT antibodies (43.6 ± 8.8 pN, gray) and group-1 Abs (44.0 ± 8.1 pN, green), higher for group-2 Abs (60.6 ± 15.4 pN, blue) and highest for group-3 Abs (72.4 ± 26.2 pN, red). Statistics showed no significant difference between KKO and group-1 Abs (p = 0.877), significant difference between group-1 and group-2 Abs (p < 0.001), or between group-2 and group-3 Abs (p = 0.006)) (Figure 6) [55].

Figure 6.

Binding characteristics of aPF4/H Abs. Each dot shows the mean and standard error of the rupture force for each respective antibody from five sera per group. (A) KKO and (B) group-1 Abs bind to PF4/H complexes with a binding strength mostly ≤60 pN (black dotted line), while (C) group-2 and (C) group-3 Abs consist of Abs with different binding forces. (D) a subset of group-3 Abs binds to PF4/H complexes with rupture forces higher than 100 pN (red-dotted line). Adapted from [55].

Group-3 Abs bound to PF4/H complexes with much higher binding energy (ΔH = −2.87 ± 2.06 × 108 cal/mol) than group-2 Abs (ΔH = −2.90 ± 0.4 × 104 cal/mol), and their dissociation constant (KD) (~5.3 nM) was about two orders of magnitude lower than that of group-2 Abs (~1.7 × 102 nM). The binding strength of PF4 to heparin ~150 pN [35] is higher than that between group-3 Abs and PF4/H complexes (mostly lower than 150 pN) [55]. Besides that, the group-3 Abs have a highest binding affinity (koff = 0.12 s−1) as compared with group-1 Abs y (koff = 15.6 s−1), group-2 Abs (koff = 2.0 s−1), or KKO (koff = 2.2 s−1). The lowest thermal off-rate specify that multiplexes induced by PF4/H complexes with group-3 Abs are more stable than those formed with other antibody groups. Furthermore, KKO and group-1 Abs contain antibodies with similar characteristics, and therefore, they interacted rather uniformly with PF4/H complexes. This has been clarified by obtaining the relatively small differences among the rupture forces (<60 pN, Figure 6A-B) measured from different cantilevers. However, group-2 Abs contain different types of antibodies as observed by a large variation of all binding forces (~40% exceeded 60 pN). For group-3 Abs, the variation of binding force is even higher than that of group-2 Abs as shown by ~44% of all binding forces ≥60 pN and ~15% even exceeded 100 pN. The low variability in binding forces of KKO and group-1 Abs has been attributed to the fact that they contain homogeneous antibodies, whereas the patient’s sera such as group-2 and group-3 Abs contained polyclonal mixtures of aPF4/P Abs differently reactive. Among these human-derived Abs, it has been proved that the group-2 contains also antibodies reacting like group-1 Abs, while group-3 is highly complicated as it composes of not only antibodies reacting like group-1 and group-2 Abs but also some additional super strong reactive antibodies. The aPF4/H Abs show different reactivity patterns under various pH and ionic strength conditions [56].

3.1.3. Autoimmune antibodies cluster PF4

The autoimmune group-3 Abs activate platelets in the absence of polyanions because they can self-cluster PF4 to form PF4/group-3 antibody complexes without the need of heparin [55]. This characteristic of autoimmune group-3 Abs has been proved by various methodologies:

First, the autoimmune group-3 Abs could be purified from the patient’s sera using a PF4-column (instead of the PF4/H column). Hardly any PF4/P Abs were obtained from control and group-1 sera; group-2 sera showed a minimally increased IgG yield. When these antibodies are concentrated to 50 μg/ml, only antibodies purified from group-3 sera activated platelets in the HIPA. The results indicate that group-3 sera contain antibodies with PF4 specificity, which activate platelets.

Next, only autoantibodies (group-3) show strong interaction with PF4 alone by ITC. When the antibodies were tested at the same concentration of 62.5 nM, KKO and group-2 Abs did not interact with PF4, while group-3 Abs interacted strongly. As the interaction between group-3 Abs and PF4 alone showed two binding sites (stoichiometry n = CABS/CPF4 = 0.53 ± 0.003), these Abs can cluster two PF4 molecules. Increasing antibody concentration did not improve binding of KKO to PF4 whereas group-2 Abs weakly interacted with PF4. However, the binding energy released by group-2 Abs is only 0.1% compared to that of group-3 Abs.

Consistently, PF4 or PF4/H EIA show that group-3 Abs bound quite strong to PF4 while other antibodies did not even though all Abs bound much stronger to PF4/H complexes than to PF4 alone. By AFS, group-1 and group-2 Abs showed much less binding events to PF4 than to PF4/H complexes, while the super-reactive group-3 Abs showed similar bindings. In addition, the interaction forces of group-3 Abs purified via a PF4-column with PF4/H complexes showed the highest range of binding forces (~100 pN). These results again indicate that group-3 Abs bind strongly to PF4 alone independently from heparin, while bindings of group-1 and group-2 Abs are heparin-dependent.

By dynamic light scattering (DLS), group-3 Abs formed the largest complexes with PF4 as compared to other antibody groups with even larger size than PF4/H complexes further indicate that group-3 Abs can cluster PF4.

The binding energy generated by the interaction of group-3 Abs with PF4 in the ITC experiments (ΔH = −3.5 ± 0.86 × 107cal/mol) is much higher than the energy released when a 16-mer heparin interacts with PF4 (ΔH = −7.26 ± 1.36 × 103 cal/mol) [6]. As 16-mer heparin can force two PF4 molecules together, based on their high energy release, group-3 Abs most probably also can force two PF4 tetramers together. In addition, the negative entropy of the reaction (ΔS = −11.7 ± 2.8 × 104 cal/mol. K) is attributed to PF4 conformational change when forming complexes with the group-3 Abs. By DLS, the size complexes formed by PF4 and group-3 Abs increases significantly when the group-2 Abs are added, indicating that group-3 Abs, induce a conformational change in PF4 and the resulting PF4/group-3 antibody complexes allow binding of group-2 Abs in the same way as polyanions do.

Altogether, PF4 form large complexes with heparin and allow group-2 Abs bind and induce platelet aggregation/activation (Figure 7A-C). Importantly, a subset of group-3 Abs cluster PF4 and the resulting PF4/Group-3 antibody complexes also allow binding of group-2 Abs and enhance platelet aggregation/activation even stronger than heparins do as shown by tighter and denser aggregates (Figure 7D-F).

Figure 7.

Group-3 Abs cluster PF4 and enhance platelet activation. (A) PF4 form large complexes with heparin and the resulting PF4/H complexes allow (B) group-2 Abs bind and (C) induce platelet aggregation/activation. (D) a subset of group-3 Abs cluster PF4 forming PF4/Group-3 antibody complexes which also (E) allow binding of group-2 Abs and (F) enhance platelet aggregation/activation evidenced by tighter and denser aggregates compared to (C). Adapted from [55].

3.2. HIT-like antibodies

Many studies in HIT have been performed with human aPF4/P Abs isolated from patient plasma because only one monoclonal antibody (KKO) mimicking human HIT antibodies did exist until recently [57]. KKO activates platelets [58] and monocytes [59] in vitro and in vivo by cross-linking FcγRIIa. KKO has been used to unravel the pathogenesis of HIT and is the basis for a recently FDA approved plasma-based antigen assay (HIT-HemosIL) for detection of PF4/P antibodies [60, 61]. KKO mimics the biological activity of human aPF4/P Abs [62] and has been used to understand the binding characteristics of an antibody recognizing PF4/P complexes and activating platelets [62, 63]. Binding of a non-HIT antibody RTO to PF4 monomers prevents PF4 tetramerization and inhibits KKO and human HIT IgG-induced platelet activation/aggregation in vitro, and thrombus progression in vivo. The probability and the interaction force of KKO binding to PF4 are much greater than those of RTO, while KKO/PF4 dissociation rate was approximately 10-fold slower than RTO/PF4 [62, 63], indicating that KKO binds stronger than RTO and KKO/PF4 complexes are more stable than RTO/PF4.

KKO interacts with PF4/H complexes coated platelets with ~4-fold higher forces than with PF4/H complexes coated on a solid phase, while RTO shows only a minor change [64]. The different binding forces strongly indicate that PF4 and PF4/H complexes either expose different epitopes or allow better access of platelet-activating Abs to their epitope when PF4 bound to the platelet surface compared to the presentation of PF4/H complexes on a solid phase. Most probably, PF4/H complexes exhibited the antigenic site differently depending on the bound substrates [53]. The findings provide an explanation for the surprising observation that KKO interact relatively weak when PF4/H complexes are immobilized on a solid phase [55], while it strongly activates platelets in functional assays. It is unresolved, which additional binding partners on the platelet surface interfere with the conformational change or different presentations of PF4/H complexes. Nevertheless, chondroitin sulfate [53] and polyphosphates [65] are potential candidates, as they interact with PF4.

However, KKO is a mouse IgG2b antibody (an absent subclass in humans) [66], while the platelet-activating aPF4/P Abs present in HIT plasma samples are predominantly IgG1. KKO behaves differently from human aPF4/P Abs, i.e. it binds only weakly to PF4/H complexes coated on a solid phase [64]. Recently, a chimeric monoclonal aPF4/H Abs with a human Fc fragment (5B9) has been developed [67]. The 5B9 antibody has been demonstrated to fully mimic the cellular effects of human HIT Abs [10, 68].

Advertisement

4. Diagnosis of HIT

Immunologic assays, such as polytypic ELISA, IgG-specific ELISA, and particle gel immunoassay (PGI) have a sensitivity, are widely used to detect aPF4/H Abs in the diluted human sera because of their high sensitivity (≥95%) and the fast turn-around. However, only ~50% of aPF4/H Abs detected by antigen tests are clinically irrelevant. The results from positive immunologic assays may lead to an overtreatment for HIT that can result in serious consequences, such as venous limb gangrene or fatal hemorrhage [69]. However, immunologic assays are still powerful tools to rule-out patients with HIT. The cut-off optical intensity (OD) in ELISA was defined at 0.5. An ELISA test showing OD > 0.5 is normally suspected to contain aPF4/H Abs. To increase the specificity of clinically relevant antibodies, a higher OD cut-off for the antigen tests (e.g. OD > 1.0) had been suggested [70].

Even though functional assays such as by serotonin release assay (SRA) [71] or HIPA [72] have a sensitivity of ~90% which is slightly lower than the immunologic assays, these tests show a much better specificity of over 90%. For the better identifying HIT, it is recommended that a positive PF4/H ELISA should prompt confirmatory testing by functional assays [73]. However, the functional assays are only available in specialized laboratories and not available in many countries. Therefore, many physicians rely on the results of antigen tests, especially for the first days after clinical suspicion of HIT has been raised until the results of the functional assay is reported.

Besides immunologic assays and functional assays, the chemiluminescent immunoassays such as HemosIL AcuStar HIT-IgG and HemosIL AcuStar HIT-Ab have been recently introduced. These methods are relatively faster (~30 minutes) than the immunologic assays (hours) and showed extremely high sensitivity (~100%) [74]. The assays seem to be ideal for ruling out HIT. Another study used a colorimetric test to detect HIT based on the interaction between platelets and tetrazolium-based indicator dye [75]. The authors reported the quality of detecting HIT is from 96 to 100% agreement with the functional assay C-SRA.

Advertisement

5. Conclusion

Not only heparin but also autoimmune antibodies induce thrombocytopenia. Large antigenic complexes formed between PF4 and either heparin or antibody activate platelets, cause a prothrombotic and result in a variety of thromboembolic and systemic consequences. In autoimmune HIT, aPF4/P Abs activate platelets in the absence of heparin. These antibodies are highly reactive. They can self-cluster PF4-molecules forming antigenic complexes and allow binding of otherwise aPF4/P Abs. The resulting immunocomplexes induce massive platelet activation in the absence of heparin. The source and length of heparins play an important role in inducing thrombocytopenia. Improvement of heparin quality together with discovering new non-heparin drugs should be highly desirable. Patients who are suspected of HIT need to be immediately stopped heparin exposure and switched to an alternative anticoagulant. Regarding patients with antibody-induced thrombocytopenia, the level of complication is much higher than the general heparin-induced thrombocytopenia. To date, these human-derived antibodies are hardly controlled, and therefore, efforts in the field would be appreciated. Clinical tests for detecting HIT antibodies as well as autoimmune HIT antibodies must be improved to achieve an appropriate identification of clinical HIT patients.

Advertisement

Acknowledgments

This work was supported by the Deutsche Forschungsgemeinschaft (DFG, Germany) (NG 133/1-1).

Advertisement

Conflict of interest

The authors declare no competing financial interests.

References

  1. 1. Greinacher A. Heparin-induced thrombocytopenia. The New England Journal of Medicine. 2015;373(19):1883-1884
  2. 2. Warkentin TE, Greinacher A. Heparin-Induced Thrombocytopenia. 4th ed. CRC Press; 2013
  3. 3. Blank M, Shoenfeld Y, Tavor S, Praprotnik S, Boffa MC, Weksler B, Walenga MJ, Amiral J, Eldor A. Anti-platelet factor 4/heparin antibodies from patients with heparin-induced thrombocytopenia provoke direct activation of microvascular endothelial cells. International Immunology. 2002;14(2):121-129
  4. 4. Michelson AD. Platelets. 3rd ed. Academic Press/Elsevier; 2013
  5. 5. Nguyen TH. Single-molecule force spectroscopy applied to heparin-induced thrombocytopenia. Journal of Molecular Recognition. 2016
  6. 6. Petitou M, van Boeckel CA. A synthetic antithrombin iii binding pentasaccharide is now a drug! What comes next?, Angewandte Chemie. 2004;43(24):3118-3133
  7. 7. Jackson SP. The growing complexity of platelet aggregation. Blood. 2007;109(12):5087-5095
  8. 8. Brandt S, Krauel K, Gottschalk KE, Renne T, Helm CA, Greinacher A, Block S. Characterisation of the conformational changes in platelet factor 4 induced by polyanions: Towards in vitro prediction of antigenicity. Thrombosis and Haemostasis. 2014;112(1):53-64
  9. 9. Kelton JG, Sheridan D, Santos A, Smith J, Steeves K, Smith C, Brown C, Murphy WG. Heparin-induced thrombocytopenia - laboratory studies. Blood. 1988;72(3):925-930
  10. 10. Rollin J, Pouplard C, Gruel Y. Risk factors for heparin-induced thrombocytopenia: Focus on fc gamma receptors. Thrombosis and Haemostasis. 2016;116(5):799-805
  11. 11. Whiteheart SW. Platelet granules: Surprise packages. Blood. 2011;118(5):1190-1191
  12. 12. Sixma JJ, Wester J. The hemostatic plug. Seminars in Hematology. 1977;14(3):265-299
  13. 13. Nguyen TH, Palankar R, Bui VC, Medvedev N, Greinacher A, Delcea M. Rupture forces among human blood platelets at different degrees of activation. Scientific Reports-UK. 2016;6:1-12. Article ID: 25402
  14. 14. Warkentin TE, Kelton JG. Heparin-induced thrombocytopenia. Annual Review of Medicine. 1989;40:31-44
  15. 15. Powell AK, Yates EA, Fernig DG, Turnbull JE. Interactions of heparin/heparan sulfate with proteins: Appraisal of structural factors and experimental approaches. Glycobiology. 2004;14(4):17R-30R
  16. 16. Valle-Delgado JJ, Urban P, Fernandez-Busquets X. Demonstration of specific binding of heparin to plasmodium falciparum-infected vs. non-infected red blood cells by single-molecule force spectroscopy. Nanoscale. 2013;5(9):3673-3680
  17. 17. Laremore TN, Zhang F, Dordick JS, Liu J, Linhardt RJ. Recent progress and applications in glycosaminoglycan and heparin research. Current Opinion in Chemical Biology. 2009;13(5-6):633-640
  18. 18. Marszalek PE, Oberhauser AF, Li H, Fernandez JM. The force-driven conformations of heparin studied with single molecule force microscopy. Biophysical Journal. 2003;85(4):2696-2704
  19. 19. Lee G, Nowak W, Jaroniec J, Zhang QM, Marszalek PE. Molecular dynamics simulations of forced conformational transitions in 1,6-linked polysaccharides. Biophysical Journal. 2004;87(3):1456-1465
  20. 20. Bailey RT Jr, Ursick JA, Heim KL, Hilleman DE, Reich JW. Heparin-associated thrombocytopenia: A prospective comparison of bovine lung heparin, manufactured by a new process, and porcine intestinal heparin. Drug Intelligence & Clinical Pharmacy. 1986;20(5):374-378
  21. 21. Francis JL, Palmer GJ, 3rd RM, Drexler A. Comparison of bovine and porcine heparin in heparin antibody formation after cardiac surgery. The Annals of Thoracic Surgery. 2003;75(1):17-22
  22. 22. Green D, Martin GJ, Shoichet SH, Debacker N, Bomalaski JS, Lind RN. Thrombocytopenia in a prospective, randomized, double-blind trial of bovine and porcine heparin. The American Journal of the Medical Sciences. 1984;288(2):60-64
  23. 23. Lee JC, Lu XA, Kulkarni SS, Wen YS, Hung SC. Synthesis of heparin oligosaccharides. Journal of the American Chemical Society. 2004;126(2):476-477
  24. 24. Codee JDC, Stubba B, Schiattarella M, Overkleeft HS, van Boeckel CAA, van Boom JH, van der Marel GA. A modular strategy toward the synthesis of heparin-like oligosaccharides using monomeric building blocks in a sequential glycosylation strategy. Journal of the American Chemical Society. 2005;127(11):3767-3773
  25. 25. de Paz JL, Noti C, Seeberger PH. Microarrays of synthetic heparin oligosaccharides, Journal of the American Chemical Society. 2006;128(9):2766-2767
  26. 26. Polat T, Wong CH. Anomeric reactivity-based one-pot synthesis of heparin-like oligosaccharides. Journal of the American Chemical Society. 2007;129(42):12795-12800
  27. 27. Xu YM, Cai C, Chandarajoti K, Hsieh PH, Li LY, Pham TQ, Sparkenbaugh EM, Sheng JZ, Key NS, Pawlinski R, Harris EN, Linhardt RJ, Liu J. Homogeneous low-molecular-weight heparins with reversible anticoagulant activity. Nature Chemical Biology. 2014;10(4):248-250
  28. 28. Rauova L, Poncz M, McKenzie SE, Reilly MP, Arepally G, Weisel JW, Nagaswami C, Cines DB, Sachais BS. Ultralarge complexes of pf4 and heparin are central to the pathogenesis of heparin-induced thrombocytopenia. Blood. 2005;105(1):131-138
  29. 29. Warkentin TE, Levine MN, Roberts RS, Gent M, Horsewood P, Kelton JG. Heparin-induced thrombocytopenia is more common with unfractionated heparin than with low-molecular-weight heparin. Thrombosis and Haemostasis. 1993;69(6):911-911
  30. 30. Linhardt RJ, Liu J. Synthetic heparin. Current Opinion in Pharmacology. 2012;12(2):217-219
  31. 31. Warkentin TE, Maurer BT, Aster RH. Heparin-induced thrombocytopenia associated with fondaparinux. New England Journal of Medicine. 2007;356(25):2653-2654
  32. 32. Martel N, Lee J, Wells PS. Risk for heparin-induced thrombocytopenia. With unfractionated and low-molecular-weight heparin thromboprophylaxis: A meta-analysis. Blood. 2005;106(8):2710-2715
  33. 33. Block S, Greinacher A, Helm CA, Delcea M. Characterization of bonds formed between platelet factor 4 and negatively charged drugs using single molecule force spectroscopy. Soft Matter. 2014;10(16):2775-2784
  34. 34. Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin V, Yaici A, Weitzenblum E, Cordier JF, Chabot F, Dromer C, Pison C, Reynaud-Gaubert M, Haloun A, Laurent M, Hachulla E, Cottin V, Degano B, Jais X, Montani D, Souza R, Simonneau G. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation. 2010;122(2):156-163
  35. 35. Nguyen TH, Greinacher A, Delcea M. Quantitative description of thermodynamic and kinetic properties of the platelet factor 4/heparin bonds. Nanoscale. 2015;7(22):10130-10139
  36. 36. Saboury AA. A review on the ligand binding studies by isothermal titration calorimetry. Journal of the Iranian Chemical Society. 2006;3(1):1-21
  37. 37. Greenfield NJ. Using circular dichroism spectra to estimate protein secondary structure. Nature Protocols. 2006;1(6):2876-2890
  38. 38. Evans E, Ritchie K. Dynamic strength of molecular adhesion bonds. Biophysical Journal. 1997;72(4):1541-1555
  39. 39. Friddle RW, Noy A, De Yoreo JJ. Interpreting the widespread nonlinear force spectra of intermolecular bonds. Proceedings of National Academy of Sciences USA. 2012;109(34):13573-13578
  40. 40. Noy A, Friddle RW. Practical single molecule force spectroscopy: How to determine fundamental thermodynamic parameters of intermolecular bonds with an atomic force microscope. Methods. 2013;60(2):142-150
  41. 41. P. Tripathi, A. Beaussart, D. Alsteens, V. Dupres, I. Claes, I. von Ossowski, W. M. de Vos, A. Palva, S. Lebeer, J. Vanderleyden and Y. F. Dufrene, Adhesion and nanomechanics of pili from the probiotic lactobacillus rhamnosus gg, ACS Nano 7 (2013), no. 4, 3685-3697
  42. 42. Hinterdorfer P, Dufrene YF. Detection and localization of single molecular recognition events using atomic force microscopy. Nature Methods. 2006;3(5):347-355
  43. 43. Wang B, Xu B. Transition model for ricin-aptamer interactions with multiple pathways and energy barriers. Physical Review. E, Statistical, Nonlinear, and Soft Matter Physics. 2014;89(2):022720
  44. 44. Cines DB, Tomaski A, Tannenbaum S. Immune endothelial-cell injury in heparin-associated thrombocytopenia. New England Journal of Medicine. 1987;316(10):581-589
  45. 45. Arepally GM, Mayer IM. Antibodies from patients with heparin-induced thrombocytopenia stimulate monocytic cells to express tissue factor and secrete interleukin-8. Blood. 2001;98(4):1252-1254
  46. 46. Tutwiler V, Madeeva D, Ahn HS, Andrianova I, Hayes V, Zheng XL, Cines DB, McKenzie SE, Poncz M, Rauova L. Platelet transactivation by monocytes promotes thrombosis in heparin-induced thrombocytopenia. Blood. 2016;127(4):464-472
  47. 47. Greinacher A, Eichler P, Lubenow N, Kwasny H, Luz M. 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;96(3):846-851
  48. 48. Kreimann M, Brandt S, Krauel K, Block S, Helm CA, Weitschies W, Greinacher A, Delcea M. Binding of anti-platelet factor 4/heparin antibodies depends on the thermodynamics of conformational changes in platelet factor 4. Blood. 2014;124(15):2442-2449
  49. 49. Warkentin TE. Heparin-induced thrombocytopenia. Current Opinion in Critical Care. 2015;21(6):576-585
  50. 50. Warkentin TE, Greinacher A, Gruel Y, Aster RH, Chong BH, scientific, t. standardization committee of the international society on and haemostasis. Laboratory testing for heparin-induced thrombocytopenia: A conceptual framework and implications for diagnosis. Journal of Thrombosis and Haemostasis. 2011;9(12):2498-2500
  51. 51. Warkentin TE, Basciano PA, Knopman J, Bernstein RA. Spontaneous heparin-induced thrombocytopenia syndrome: 2 new cases and a proposal for defining this disorder. Blood. 2014;123(23):3651-3654
  52. 52. Amiral J, Pouplard C, Vissac AM, Walenga JM, Jeske W, Gruel Y. Affinity purification of heparin-dependent antibodies to platelet factor 4 developed in heparin-induced thrombocytopenia: Biological characteristics and effects on platelet activation. British Journal of Haematology. 2000;109(2):336-341
  53. 53. Padmanabhan A, Jones CG, Bougie DW, Curtis BR, McFarland JG, Wang DM, Aster RH. Heparin-independent, pf4-dependent binding of hit antibodies to platelets: Implications for hit pathogenesis. Blood. 2015;125(1):155-161
  54. 54. Cines DB, Rauova L, Arepally G, Reilly MP, McKenzie SE, Sachais BS, M P. Heparin-induced thrombocytopenia: An autoimmune disorder regulated through dynamic autoantigen assembly/disassembly. Journal of Clinical Apheresis. 2007;22(1):31-36
  55. 55. Nguyen TH, Medvedev N, Delcea M, Greinacher A. Anti-platelet factor 4/polyanion antibodies mediate a new mechanism of autoimmunity. Nature Communications. 2017;8:14945
  56. 56. Nguyen TH, Greinacher A. Effect of ph and ionic strength on the binding strength of anti-pf4/polyanion antibodies. European Biophysics Journal (EBJ). 2017;46:795-801
  57. 57. Reilly MP, Taylor SM, Hartman NK, Arepally GM, Sachais BS, Cines DB, Poncz M, McKenzie SE. Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through fc gamma riia. Blood. 2001;98(8):2442-2447
  58. 58. 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;107(6):2346-2353
  59. 59. Kasthuri RS, Glover SL, Jonas W, McEachron T, Pawlinski R, Arepally GM, Key NS, Mackman N. Pf4/heparin-antibody complex induces monocyte tissue factor expression and release of tissue factor positive microparticles by activation of fc gamma ri. Blood. 2012;119(22):5285-5293
  60. 60. Jourdy Y, Nougier C, Rugeri L, Bordet JC, Sobas F, Negrier C. Prospective evaluation of automatized pf4/heparin immunoassays hemosil hit-ab (pf4-h) for the diagnosis of heparin-induced thrombocytopenia. International Journal of Laboratory Hematology. 2015;37(2):244-252
  61. 61. Althaus K, Hron G, Strobel U, Abbate R, Rogolino A, Davidson S, Greinacher A, Bakchoul T. Evaluation of automated immunoassays in the diagnosis of heparin induced thrombocytopenia. Thrombosis Research. 2013;131(3):E85-E90
  62. 62. Sachais BS, Litvinov RI, Yarovoi SV, Rauova L, Hinds JL, Rux AH, Arepally GM, Poncz M, Cuker A, Weisel JW, Cines DB. Dynamic antibody-binding properties in the pathogenesis of hit. Blood. 2012;120(5):1137-1142
  63. 63. Litvinov RI, Yarovoi SV, Rauova L, Barsegov V, Sachais BS, Rux AH, Hinds JL, Arepally GM, Cines DB, Weisel JW. Distinct specificity and single-molecule kinetics characterize the interaction of pathogenic and non-pathogenic antibodies against platelet factor 4-heparin complexes with platelet factor 4. The Journal of Biological Chemistry. 2013;288(46):33060-33070
  64. 64. Nguyen TH, Greinacher A. Platelet factor 4/heparin complexes present their epitopes differently on a solid phase system than on the platelet surface. Blood. 2017
  65. 65. Yarovoi SV, Cines DB, Zaitsev SV, Lebedeva T, Rauova L, Poncz M, Arepally GM, Khandelwal S, et al. Polyphosphate/platelet factor 4 complexes can mediate heparin-independent platelet activation in heparin-induced thrombocytopenia. Blood Advances. 2016;1:62-72
  66. 66. Thauvin-Robinet C, Munck A, Huet F, de Becdelievre A, Jimenez C, Lalau G, Gautier E, Rollet J, Flori J, Nove-Josserand R, Soufir JC, Haloun A, Hubert D, Houssin E, Bellis G, Rault G, David A, Janny L, Chiron R, Rives N, Hairion D, Collignon P, Valeri A, Karsenty G, Rossi A, Audrezet MP, Ferec C, Leclerc J, Georges M, Claustres M, Bienvenu T, Gerard B, Boisseau P, Cabet-Bey F, Cheillan D, Feldmann D, Clavel C, Bieth E, Iron A, Simon-Bouy B, Izard V, Steffann J, Viville S, Costa C, Drouineaud V, Fauque P, Binquet C, Bonithon-Kopp C, Morris MA, Faivre L, Goossens M, Roussey M, Girodon E. p. A. H. collaborating working group on, Cftr p. Arg 117 his associated with cbavd and other cftr-related disorders. Journal of Medical Genetics. 2013;50(4):220-227
  67. 67. Morio F, Aubin GG, Danner-Boucher I, Haloun A, Sacchetto E, Garcia-Hermoso D, Bretagne S, Miegeville M, Le Pape P. High prevalence of triazole resistance in aspergillus fumigatus, especially mediated by tr/l98h, in a french cohort of patients with cystic fibrosis. The Journal of Antimicrobial Chemotherapy. 2012;67(8):1870-1873
  68. 68. Grossi O, Horeau-Langlard D, Agard C, Haloun A, Lefebvre M, Neel A, Hamidou MA. Low-dose methotrexate in pah related to t-cell large granular lymphocyte leukaemia. The European Respiratory Journal. 2012;39(2):493-494
  69. 69. Smythe MA, Warkentin TE, Woodhouse AL, Zakalik D. Venous limb gangrene and fatal hemorrhage: Adverse consequences of hit “overdiagnosis” in a patient with antiphospholipid syndrome. American Journal of Hematology. 2011;86(2):188-191
  70. 70. Nagler M, Bachmann LM. H. ten Cate and A. ten Cate-Hoek, Diagnostic value of immunoassays for heparin-induced thrombocytopenia: A systematic review and meta-analysis, Blood. 2016;127(5):546-557
  71. 71. Warkentin TE. Laboratory testing for heparin-induced thrombocytopenia. Journal of Thrombosis and Thrombolysis. 2000;10:S35-S45
  72. 72. Greinacher A. Heparin-induced thrombocytopenia. New England Journal of Medicine. 2015;373(3):252-261
  73. 73. Warkentin TE. n. Vol.1, (September 2002), pp.63-72. Heparin-induced thrombocytopenia. Current Hematology Reports. 2002
  74. 74. Legnani C, Cini M, Pili C, Boggian O, Frascaro M, Palareti G. Evaluation of a new automated panel of assays for the detection of anti-pf4/heparin antibodies in patients suspected of having heparin-induced thrombocytopenia. Thrombosis and Haemostasis. 2010;104(2):402-409
  75. 75. Prechel MM, Escalante V, Drenth AF, Walenga JM. A colorimetric, metabolic dye reduction assay detects highly activated platelets: Application in the diagnosis of heparin-induced thrombocytopenia. Platelets. 2012;23(1):69-80

Written By

Thi-Huong Nguyen

Submitted: 15 July 2017 Reviewed: 08 January 2018 Published: 07 March 2018