You are on page 1of 8

Clinica Chimica Acta 504 (2020) 73–80

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/cca

Review

Pathogenesis and management of heparin-induced thrombocytopenia and T


thrombosis
Pan Zhoua,1, Jia-Xin Yinb,1, Hua-Lin Taoa, , Hong-wei Zhangc,
⁎ ⁎

a
Department of Clinical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
b
Class 1, Degree 2016, Department of Medical Laboratory, Southwest Medical University, Luzhou, Sichuan, China
c
Department of Blood Transfusion, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China

ARTICLE INFO ABSTRACT

Keywords: Heparin-induced thrombocytopenia and thrombosis is a severe immune-mediated adverse drug effect caused by
Heparin the IgG antibodies to platelet factor4/heparin complexes. Activated platelets, vascular endothelium, and
Heparin-induced thrombocytopenia and monocytes generate the life-threatening thrombocytopenia and thrombosis. In this review, we will update the
thrombosis reader on recent findings on the pathogenesis and clinical management of heparin-induced thrombocytopenia
Platelet factor 4
and thrombosis. Firstly, PF4/heparin complexes make IgM mediate complement activation by classical pathway.
Pathogenesis
Secondly, Marginal zoneB cells play a crucial role in producing anti-PF4/heparin complex IgG antibody. Thirdly,
Management
two activation signals of platelets (protease-activated receptor 1/Fc gamma IIA receptor) were confirmed. Based
on these findings, we present a potential laboratory test of HITT (receptor glycoprotein Ⅳ) and two possible
treatments by using receptor inhibitors (vorapaxar/atopaxar) and IgG-degrading enzyme (streptococcus pyo-
genes/glutamyl endopeptidase V8/matrix metalloproteinases).

1. Introduction large complexes (ULCs), triggering platelet activation and low platelet
counts [12]. Another mechanism is that high doses of heparin cause
Despite the clinical introduction of a few new drugs, heparin has platelets to bind to fibrinogen, resulting in a mild form of thrombocy-
remained to be a widely used anticoagulant in clinical practice since topenia [13]. When HIT type 1 occurs, the platelet counts normalize
1970s because of its low cost, satisfactory efficacy, and accessibility. about four days after or even without any treatments [12,14,15]. Thus,
However, heparin can also cause some adverse reactions including skin HIT type 1 isn’t significant in clinical practice.
lesions, thrombocytopenia, massive hemorrhage, and osteoporosis, The second type of HIT, HITT or HIT type 2 is more severe caused by
among which thrombocytopenia is the most severe one [1–4]. the ULCs-targeted IgG antibody in vivo, and patients with this type will
Studies on mechanism have revealed that HITT is caused by im- progress to life-threatening thrombosis and thrombocytopenia, ampu-
mune antibodies to complexes between unfractionated heparin (UFH) tation or even death. HITT usually occurs 5–10 days after treatment
and platelet factor 4 (PF4). This iatrogenic disease usually occurs after with heparin [16]. The platelet counts could not recover unless UFH is
surgeries like knee joint replacement, hip joint replacement, cardiac discontinued [15,17]. This paper will update the reader on recent de-
surgery and extracorporeal circulation [5–8] and there is a 50% chance velopments on the pathogenesis and clinical management of HITT and
to lead to thrombosis complications of microarteries, microveins and present a potential laboratory test and some possible treatments.
even systemic circulation [9–11]. Therefore, there is a need to increase
research on the pathogenesis and management of HITT. 2. Heparin
There are two types of heparin-induced thrombocytopenia (HIT).
The first type, nonimmune heparin-associated thrombocytopenia or A common anticoagulant drug in clinical treatment [18,19] fol-
HIT type 1, usually occurs when patients are exposed to UFH or high lowing its discovery in 1916 and application in 1940s, heparin is a
doses of heparin for the first time. The major mechanism is that heparin linear polysaccharide highly sulfated, and most heparin contains acet-
binds to surface-bound PF4 of platelets to form the PF4/heparin ultra- ylation groups and sulfate groups often found in organs rich in mast

Corresponding authors at: Department of Clinical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China (H.-L. Tao).

Department of Blood Transfusion, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China (H.-W. Zhang).
E-mail addresses: lzyxyjyx@163.com (H.-L. Tao), wangchong8210@163.com (H.-w. Zhang).
1
The first two authors contributed equally to this paper.

https://doi.org/10.1016/j.cca.2020.02.002
Received 16 August 2019; Received in revised form 30 January 2020; Accepted 3 February 2020
Available online 04 February 2020
0009-8981/ © 2020 Elsevier B.V. All rights reserved.
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

cells (i.e., intestine, liver, and lung cells) [20,21]. Heparin has been antigenicity of C3a up to 10,000-folds [52]. In addition, the CD21 on B
extracted from pigs, cows, or other animals by various chemical or cells can bind to T cells to differentiate B cells into subtypes and gen-
physical means, and commercial heparin has many types such as he- erate ULCs-targeted IgG antibody [51] (Fig. 2d). Recent studies have
parin, low molecular weight heparin (LMWH), and ultra low molecular demonstrated ULCs-targeted IgG antibody is mainly composed of type 1
weight heparin (ULMWH). The resources of heparin are related to the IgG (IgG1) and a small amount of type 2 IgG (IgG2) [45,51]. These
risk of HITT. Some studies have shown that bovine heparin may be antibodies are mainly produced by MZ B cells [53–55]. Additionally,
more likely to cause HITT than porcine heparin [22,23]. MZ B cells can also produce IgM antibodies in T cell-independent im-
The anticoagulant effect of heparin depends on the antithrombin in mune responses [53]. The mechanism of antibody production is illu-
the plasma and the structure of heparin would allow the heparin mo- strated in Fig. 2.
lecule to act as an intermediate binding thrombin and antithrombin at
two ends respectively [24]. Once banded to heparin molecules, the 3.3. Binding of antibodies and participation of cells
structure of antithrombin would be changed so that it moves to the
other side of the heparin molecule to conjugate with thrombin to de- 3.3.1. FcγRIIa receptor and ULCs-targeted IgG antibody
activate the thrombin [25,26]. Subsequent reactions will inactivate MZ B cells, which are differentiated from B cells, can release ULCs-
more thrombin and serine proteases [27,28]. Moreover, heparin can targeted IgG antibody into the systemic circulation. When the complex
bind to heparin cofactor Ⅱ to further inhibit plasma coagulation ULCs are formed from heparin and PF4, PF4 is changed to exhibit a new
[20,29–31]. binding motif. Therefore, the free form of ULCs-targeted IgG antibody
Various types of heparin can induce thrombocytopenia. can bind to the new motif to form PF4-H-IgG complex that can bind
Experiments have shown that UFH is more likely to interact with the with the FcγRIIa receptor to transmit the signals and promotes cell
platelets to induce thrombocytopenia for its higher molecular weight, activation.
and it is more likely to bind to endothelial cells and macrophages than FcγRIIa receptor (CD32a), a type I transmembrane glycoprotein of
fractionated heparin is [21,32]. This corresponds to the fact that pa- 40KDa, is composed of two extracellular Ig domains, namely a trans-
tients treated with LMWH suffer lower incidence of HITT [33,34]. membrane domain and an intracellular domain (ITAM) [37,56,57], and
it exists not only on the membrane surface of platelets, but also on the
3. Pathogenesis of HITT surface of monocytes, endothelial cells and other types of cell mem-
branes [56,58,59]. Moreover, FcγRIIa receptor is a low-affinity receptor
Recent studies have shown that the key step of HITT is B cells ac- of IgG. When this receptor binds to the ULCs-targeted IgG antibody, the
tivation by the ULCs. The activated B cells generate IgG1-based immune ITAM in the cytoplasm of FcγRIIa receptor will be activated, and then
antibody (ULCs-targeted IgG antibody) which will conjugate with ULCs the ITAM-dependent signaling pathway, such as phosphatidylinositol-3
to form PF4-H-IgG complex [35,36]. kinase (PI3- kinase) and spleen tyrosine kinase (syk) pathway, will be
PF4-H-IgG complex would mainly bind to the Fc gamma IIA re- consequentially activated to transmit signals. All these are related to
ceptors (FcγRIIa receptors) on the platelets to activate the platelets and cell activation [37,60–62].
release PF4 [2,37,38]. Moreover, the complex would bind to the It is reported that FcγRIIa receptor is encoded by FCGR2 gene and
FcγRIIa receptors on monocytes to produce tissue factor (TF) and in- polymorphism of FCGR2 [37,63]. One genotype is that the arginine (R)
flammatory mediators [39,40]. The activated platelets and monocytes on the position 131 is replaced by histidine (H) in the second Ig domain,
are involved in promoting thrombosis. The TF and inflammatory leading to different binding affinity of FcγRIIa receptor to IgG. Studies
mediators may be associated with inflammation [39]. have shown that the FcγRIIa-131H homozygous cells are easier to bind
to IgG 1[64,65].
3.1. PF4 and formation of ULCs The receptor glycoprotein IV (GPIV) is related to FcγRIIa receptor.
GPIV doesn’t contain ITAM but contains two Ig domains outside the
PF4, a tetramer molecule with a large amount of positive charge, is membrane. When FcγRIIa receptor binds to the PF4-H-IgG complex, it
a member of CXC catabolic factor and is also known as CXCL4. It is detaches GPIV on the platelet membrane by mediating metalloprotei-
produced by megakaryocytes and stored in the platelet alpha-granules. nase and releases into plasma the fragment of 55 kDa which is also
When platelets are stimulated and activated, a large quantity of PF4 known as soluble GPIV(sGPIV) [66]. GPIV is a specific receptor of
will be released into the peripheral blood [41–43] to bind to the ne- platelet, and Qiao J’s study shows that HITT patients’ plasma level of
gatively charged glycosaminoglycan (GAG) chains in the platelets [44] sGPIV is higher than that of the healthy ones [37]. Since FcγRIIa re-
(Fig. 1a). Then, negatively charged heparin binds to the free positive ceptor can activate the platelets to release GPIV into the plasma [37], it
charge PF4, resulting in the changes of the configuration of the tetramer is possible to develop plasma G PIV as a biomarker of platelets acti-
PF4 [45]. As a result, more ULCs will be formed and immune reactions vation and use this biomarker as a potential laboratory test of HITT.
will be triggered eventually [46]. More investigations are needed to test this hypothesis.
Studies have demonstrated that the path of ULCs formed by PF4 and It is indicated that ULCs-targeted IgG antibody can be cleaved by
heparin is a bell curve [44,47]. The formation of ULCs reaches its peak Streptococcus pyogenes (IdeS) [145]. The fragments of ULCs-targeted
when the molar ratio of heparin and PF4 is 1:1. Otherwise, the for- IgG antibodies still have the ability to bind to ULCs. When the ULCs-
mation of ULCs will be inhibited [2,44,47,48]. The schematic diagram targeted IgG antibodies are cleaved, the binding affinity of PF4-H-IgG
of the formation of ULCs is shown in Fig. 1. complexes to FcγRIIa receptors is vastly reduced [145,146]. Corre-
spondingly, the ability of platelets to obtain the first activation signal
3.2. Production of ULCs-targeted IgG antibody from FcγRIIa receptor is also greatly reduced, and the risk of thrombosis
in HITT is reduced in turn. Therefore, injection of IdeS could be a po-
More pathogenetic studies have shown that free form of ULCs can tential management of severe HITT [145]. There are many enzymes
bind to free form of IgM or to the IgM on the membranes of B cells sharing the similar function to cleave the IgG antibody, such as human
[9,49] (Fig. 2a). The IgM activates the classical pathway to produce proteases (matrix metalloproteinases) and glutamyl endopeptidase V8
complement fragments C3d that binds to the complexes to form com- (GluV8) [146–148]. More studies are needed to demonstrate the effi-
plement complexes [50,51] (Fig. 2b). Furthermore, the IgM makes the cacy of these enzymes in treating HITT.
complex bind to the complement receptor (CD21), and the combination
will activate the downstream singling pathways [49]. As research in- 3.3.2. Monocyte
dicates, the combination of C3d and CD21 can increase the immune In the pathogenesis of HITT, monocytes, platelets and endothelial

74
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

Fig. 1. (a) Heparin binds to platelet-bound PF4, exposes PF4 new antigen, and forms platelet-bound ULCs. (b) Heparin binds to free PF4, exposing PF4 new antigen,
forming free ULCs.

cells may also be activated and could interact with one other to pro- PAR-1, on the cell surface transmitting a second activation signal to
mote HITT [44,67,68]. It has been shown that monocytes have three platelets, is a G protein coupling protein receptor, which is mainly
functions in the pathogenesis of the disease [5,45]. Firstly, monocytes expressed on the platelet membrane. Apart from PAR-1, there is also a
can phagocytize and clearly activate platelets with antibody complexes PAR-4 expression on the platelet membrane. These two receptors are
[5], reducing the risk of thrombosis and decreasing the amount of related to platelet activation [70–72], of which PAR-1 is the main
platelets. Secondly, after the activation of monocytes, a large amount of binding receptor of thrombin [73].
TF and mononuclear particles are released, which promotes the acti- Studies have shown that there is a thrombin cleavage site at the N-
vation of other cells and is associated with inflammation. It has also terminus in the structure of PAR-1. Thrombin binds to the N-terminus
been shown that activation would promote the expression of MHC class of PAR-1 via the hirudin sequence [74–76], and the N-terminus is
II molecules and CD83 in monocytes [53,57,69] (Fig. 3a). Thirdly, cleaved to form a new N-terminus [71].The new N-terminus is activated
monocytes provide a second activation signal to platelets. When by being bound with other parts of extracellular PAR-1, which acts as a
monocytes are activated by PF4-H-IgG complexes, the cells can release guanine nucleotide exchange factor, allowing GTP to react with other G
thrombin. Monocytes with bounding thrombin on the cell surface pass a protein subunits in the cell [77–79]. Then a series of related signaling
second activation signal to the platelets through protease-activated generate IP3 (inositol triphosphate). Then calcium, ADP, adrenaline,
receptor 1(PAR-1) (Fig. 3b), causing platelets to express phosphati- serotonin will be released into the cytosol to activate other signal
dylserine and form prethrombus platelets [55,68]. pathways in the platelet [77,80,81], inducing platelets activation.

Fig. 2. (a) Free IgM binds to ULCs to form a


complex. (b) IgM activates complement by
the classical pathway, binding complement
fragment C3d. (c) A complex that binds to B
cells transmits an activation signal to B cells.
(d) T cells bind to the complex bind to CD21
on B cells, and transmit an activation signal.
(e) B cells differentiate into MZB cells, pro-
ducing a complex antibody mainly of IgG1.

75
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

Fig. 3. (a) Monocyte phagocytose the anti-


body complex and be activated, and release
thrombin, and express MHC class II mole-
cules and CD83 on membrane. (b) Surface
thrombin on activated-monocytes binds to
PAR-1, transmitting the second signal of
activation. (c) FcγRIIa bind to PF4-H-IgG
complex on platelet, transmitting the first
signal of activation. (d) Activated-platelets
release a large amount of PF4, and PF4 have
positive feedback on the HIT. (e)
Endothelial cells are injured after bind to
the antibody complexes, and combine with
activated-platelets to form thrombus.

PAR-1 transmits the second activation signal to the platelets in the thrombocytopenia [11,89]. Each parameter has three grades 0, 1 and 2
pathogenesis of HITT, and it can be developed as a target for the and the total score is used to determine the risk, with the score 0 to 3
treatment of HITT. Currently a few PAR-1 inhibitors have been ap- corresponding to a low risk, 4 to 5 moderate risk, and 6 to 8 high risk
proved by the FDA. For example, vorapaxar (SCH-530348), atopaxar [87,90,91].
(E5555) [82–84], and volapasha have been approved in 2014 to pre- The diagnosis of HITT requires stronger evidence for laboratory
vent and treat thrombosis. The mechanism of these drugs is that the tests (platelet activation assays/immunoassays). Available tests vary
PAR-1 inhibitors can competitively bind to PAR-1 to inhibit platelet considerably in their specificity and sensitivity. The platelet activation
activation [73]. Although PAR-1 inhibitors are used for treating assays including 14C-serotonin release assay (SRA), platelet aggrega-
thrombosis, it is not clear whether such drugs can be used for treating tion assay (PAA), and flow cytometric assay (FCA) have high specificity
HITT. This will probably become a new direction in HITT research. (> 95%) but lower sensitivity (56–100%) [48,92], while immunoassays
including enzyme linked immunosorbent (ELISA), immunoturbidi-
3.3.3. Platelet metric and the like, have high sensitivity (> 99%) but lower specificity
In the pathogenesis of HITT, the platelet-membrane receptors either (30–70%) [48]. Given the deficiencies of current tests, multiple diag-
bind to the PF4-H-IgG complex or the PF4-H-IgG complex on the GAG nosis methods should be used to improve the specificity and accuracy. It
side chain of other platelets (Fig. 3c) to obtain the first activation signal. has been indicated by some studies that the use of 14c-5-hydro-
When the platelets receive the first activation signal, the P-selectins xtryptamine release test and heparin-induced platelet activation test
[85] will be up-regulated in the platelets, contributing to the formation (HIPA) could contribute to the diagnosis of HITT [93]. In addition,
of thrombus and prethrombus state. improved scoring methods, such as Hep scoring system [88] suggest the
It has been reported that the second activation signal (transmitted possibility to standardize the diagnosis and detection of HITT in the
by PAR-1) will enhance the effect of the first activation signal [57]. future studies.
Platelet agglutination occurs when the platelets are activated. Mean- On the other hand, as FcγRIIa receptor will produce sGPVI and re-
while, the microparticles secreted by the platelets through micro- lease it to the plasma when the receptor is activated in HITT patient,
vesicles will further the formation of thrombus. The released PF4 by there might be possibility that the plasma level of GPVI can be seen as a
activated platelets will counteractively accelerate the development of biomarker of potential laboratory test of HITT and more studies are
HITT [5,86] (Fig. 3d). Platelet activation, thrombus formation, and needed to test this hypothesis.
clearance of PF4-H-IgG complex lead to the decrease of platelet counts. Once HITT is confirmed, heparin should be stopped immediately.
Moreover, the heparin coated catheter should be replaced, the extra-
3.3.4. Endothelial cells corporeal circulation should be discontinued, and other types of an-
PF4-H-IgG complex binds to FcγRIIa receptor on vascular en- ticoagulants should be used [61,93,94]. There are a few novel drugs to
dothelial cells to stimulate injury to vascular endothelial cells [5]. The treat HITT, but they have not been widely used in clinical practice.
combination of platelet activation, thrombin tissue factors, mono- Overall, three types of drugs, including commercial heparin, thrombin
nuclear particles, and other factors will cause thrombocytopenia and inhibitors, and oral anticoagulants, are used for treating HITT.
vascular thrombosis (Fig. 3e).
Fig. 3 4.1. Commercial heparin

4. Management of HIT 4.1.1. LMWH


LMWH is an anticoagulant extracted from UFH [21,95,96] by way
In clinical practice, the 4Ts scoring system proposed by Warkentin is of oxidation, beta elimination, and deamination. The risk of HITT with
often used to assess the risk of HITT in patients [87–89].The scoring LMWH is between 0.2% and 0.6% [97,98], therefore, LMWH has been a
system includes four parameters: thrombocytopenia, timing of platelet replacement to UFH for several decades [21,99].
count fall, thrombosis or other clinical sequelae, and other causes of The structure of LMWH contains pentaccharide binding with

76
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

thrombin and antithrombin, thus inactivating thrombin and Xa factor immediately and replacement drugs should be used. After the platelets
and leading to anticoagulation. It was also reported that LMWH has a counts increase to the normal level, vitamin K antagonist (e.g., war-
low affinity with macrophages, endothelial cells, monocytes, and PF4, farin) can be used temporarily as an anticoagulant [135–137]. How-
therefore it contributes to a lower risk of HITT compared with UFH ever, this process may increase the risk of thrombosis [135].
[24,100,101]. It has been shown that DOACs can be used as the primary or sec-
The length of LMWH is between UFH and ultra-low molecular ondary drugs for treating HITT [138,139]. Compared with VKA, DOACs
weight heparin (ULMWH), which enables it to be injected sub- act on a single coagulation factor, with similar efficacy and a lower risk
cutaneously rather than intravenously like heparin [93] so it is a of thrombosis. DOACs do not react to PF4, PF4 complex, or HIT anti-
commonly used replacement for UFH [21]. bodies [140,141], and have the advantages of rapid onset, easy use, and
short half-life. Since DOCAs are usually eliminated through kidney, the
4.1.2. ULMWH deficiency in renal function or hypofunction can affect the efficacy of
ULMWH is an analogue of heparin synthesized by chemical means DOACs [142–144].
[102] and it has the unique pentaccharide structure of heparin
[21,103,104] to ensure its same function as heparin. The molecular 5. Conclusion
weight of ULMWH is usually about 2,000 Da, which is lower than those
of LMWH (5,000 Da) and UFH (13,000 Da) [102]. HITT is a severe side effect in patients treated with heparin, the
Pharmacological studies have demonstrated that ULMWH has a primary anticoagulant in medical practice, therefore the prevention,
longer half-life in the plasma compared with LMWH and UFH. In ad- diagnosis, and management of HITT are of great significance. In the
dition, the efficacy of ULMWH is usually better than those of UFH and future, more attention should be paid to the understanding of HITT
LMWH and the HITT risk is lower [103,105,106]. ULMWH has many pathogenesis to better diagnose and manage HITT. Future studies could
types such as sodium sulfodar-heparin, AVE5026 (semuloparin), and ro- test the role of the plasma level of GPVI as a biomarker in the laboratory
14 (a derivative of bemiparin) [107,108]. ULMWH can also be injected test of HITT. Also, ULCs-targeted IgG antibody, FcγRIIa, and PAR-1
subcutaneously and can be used to replace UFH [21]. could be the three major targets for HITT management, the efficacy of
the first of which has been shown above, and more efforts should be
4.2. Direct thrombin inhibitors made on those of FcγRIIa and PAR-1 receptors. Currently, FcγRIIa and
PAR-1 receptor inhibitors like vorapaxar (sch-530348) and atopaxar
4.2.1. Bivalirudin (E5555) are available, but they are used for treating other diseases
Bivalirudin, a synthetic peptide containing 20 amino acids residues, (cardiovascular death/coronary artery disease) [149,150] and it re-
is a thrombin inhibitor [94,109,110]. It is often used as an antic- mains unclear whether these receptor inhibitors and other IgG antibody
oagulant in cardiovascular surgery, extracorporeal membrane oxyge- enzymes (GluV8, matrix metalloproteinases) can be used as a potential
nation (ECMO) and an alternative for treating HITT [109,111]. Dif- alternative treatment of HITT. Future studies are needed to shed light
ferent from UFH, bivalirudin can directly bind to thrombin to prevent on it.
coagulation without any necessary help from any components in the
plasma. Bivalirudin’s mechanism of action is that it directly inhibits the Acknowledgments
catalytic activity site of thrombin and disturbs the binding site between
thrombin and fibrin [109,112].The half-life of bivalirudin is only This study was supported by Medical Science Innovation Project for
25 min, shorter than that of UFH. Most bivalirudin can be hydrolyzed Youth of Sichuan Province (Grant Number: Q14031).
by protease, and only 20% is excreted through the kidney [113–115].
As some studies demonstrate, bivalirudin can replace UFH to treat References
or prevent anticoagulant, by virtue of its less risk of HITT. However, it
could increase the incidence of cardiovascular stent thrombosis [1] N. van Rein, J.S. Biedermann, F. van der Meer, et al., Major bleeding risks of
[116,117]. In addition, the cost of bivalirudin is significantly higher different low-molecular-weight heparin agents: a cohort study in 12 934 patients
treated for acute venous thrombosis, J. Thromb. Haemost. 15 (2017) 1386–1391.
than that of UFH [118,119], so it can not replace heparin to teat HITT. [2] L. Rauova, M. Poncz, S.E. McKenzie, et al., Ultralarge complexes of PF4 and he-
parin are central to the pathogenesis of heparin-induced thrombocytopenia, Blood
4.2.2. Agatreban 105 (2005) 131–138.
[3] M. Schindewolf, M. Paulik, H. Kroll, et al., Low incidence of heparin-induced skin
Agatreban, a synthetic arginine derivative, is a thrombin inhibitor lesions in orthopedic surgery patients with low-molecular-weight heparins, Clin.
[94,120,121]. Pharmacological studies show that agatreban can rapidly Exp. Allergy 48 (2018) 1016–1024.
bind to the active center of thrombin to prevent the formation of fibrin, [4] G. Le Templier, M.A. Rodger, Heparin-induced osteoporosis and pregnancy, Curr.
Opin. Pulm. Med. 14 (2008) 403–407.
which would achieve anticoagulant effect [122,123].
[5] L. Rauova, G. Arepally, S.E. McKenzie, B.A. Konkle, D.B. Cines, M. Poncz, Platelet
Studies have indicated that agatreban has low antigenicity and a and monocyte antigenic complexes in the pathogenesis of heparin-induced
low risk of producing antibodies, that the efficacy of agatreban does not thrombocytopenia (HIT), J. Thromb. Haemost. 7 (2009) 249–252.
[6] J.I. Weitz, J. Harenberg, New developments in anticoagulants: past, present and
rely on other components in the plasma [112,124] and that it can be
future, Thromb. Haemost. 117 (2017) 1283–1288.
used in the treamtment of HITT, or as an anticoagulant in cardiac [7] A. Greinacher, P. Eichler, T. Lietz, T.E. Warkentin, Replacement of unfractionated
surgery and hemodialysis [108,125,126]. Agatreban has a half-life of heparin by low-molecular-weight heparin for postorthopedic surgery antith-
40–50 min, which is mainly metabolized by the liver and finally ex- rombotic prophylaxis lowers the overall risk of symptomatic thrombosis because
of a lower frequency of heparin-induced thrombocytopenia, Blood 106 (2005)
creted with feces [127] and it is not a burden to the kidney, so it can be 2921–2922.
used in patients with renal insufficiency [112,128]. [8] S. Motohashi, T. Matsuo, H. Inoue, M. Kaneko, S. Shindo, Clinical significance of
the serotonin release assay and platelet count monitoring after cardiac surgery,
Clin. Appl. Thromb. Hemost. 24 (2018) 944–949.
4.3. Direct oral anticoagulants (DOACs) [9] L. Rauova, G. Arepally, M. Poncz, D.B. Cines, Molecular and cellular pathogenesis
of heparin-induced thrombocytopenia (HIT), Autoimmun. Rev. 17 (2018)
Direct oral anticoagulant (DOAC) is a group of anticoagulants that 1046–1052.
[10] R.A. DeSimone, M.M. Cushing, Heparin-induced thrombocytopenia suggested by
exert anticoagulant effects through intestinal absorption [129,130]. microscopic examination of a below-knee amputation, Transfusion 58 (2018)
There are five types of FDA-approved DOACs: apixaban, rivaroxaban, 2784–2785.
edoxaban, betrixaban, and dabigatran, which are direct inhibitors of [11] T.E. Warkentin, Clinical picture of heparin-induced thrombocytopenia (HIT) and
its differentiation from non-HIT thrombocytopenia, Thromb. Haemost. 116 (2016)
factor Xa with the exception of dabigatran as an enzyme inhibitor 813–822.
[131–134]. Usually, when HITT occurs, heparin should be stopped

77
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

[12] T. Bakchoul, I. Marini, Drug-associated thrombocytopenia, Hematol. Am. Soc. [42] P.M. Sandset, CXCL4-platelet factor 4, heparin-induced thrombocytopenia and
Hematol. Educ. Program 2018 (2018) 576–583. cancer, Thromb. Res. 129 (Suppl 1) (2012) S97–S100.
[13] B.H. Chong, P.A. Castaldi, Platelet proaggregating effect of heparin: possible [43] M.B. Zucker, I.R. Katz, Platelet factor 4: production, structure, and physiologic and
mechanism for non-immune heparin-associated thrombocytopenia, Aust. NZ J. immunologic action, Proc. Soc. Exp. Biol. Med. 198 (1991) 693–702.
Med. 16 (1986) 715–716. [44] L. Rauova, J.D. Hirsch, T.K. Greene, et al., Monocyte-bound PF4 in the patho-
[14] C. Gao, B. Boylan, J. Fang, D.A. Wilcox, D.K. Newman, P.J. Newman, Heparin genesis of heparin-induced thrombocytopenia, Blood 116 (2010) 5021–5031.
promotes platelet responsiveness by potentiating alphaIIbbeta3-mediated outside- [45] S. Khandelwal, G.M. Arepally, Immune pathogenesis of heparin-induced throm-
in signaling, Blood 117 (2011) 4946–4952. bocytopenia, Thromb. Haemost. 116 (2016) 792–798.
[15] A.W. Shih, J.I. Sheppard, T.E. Warkentin, Platelet count recovery and serorever- [46] Z. Cai, S.V. Yarovoi, Z. Zhu, et al., Atomic description of the immune complex
sion in immune HIT despite continuation of heparin: further observations and involved in heparin-induced thrombocytopenia, Nat. Commun. 6 (2015) 8277.
literature review, Thromb. Haemost. 117 (2017) 1868–1874. [47] L. Rauova, L. Zhai, M.A. Kowalska, G.M. Arepally, D.B. Cines, M. Poncz, Role of
[16] T.E. Warkentin, A. Greinacher, Management of heparin-induced thrombocyto- platelet surface PF4 antigenic complexes in heparin-induced thrombocytopenia
penia, Curr. Opin. Hematol. 23 (2016) 462–470. pathogenesis: diagnostic and therapeutic implications, Blood 107 (2006)
[17] A.T. Gurbuz, W.G. Elliott, A.A. Zia, Heparin-induced thrombocytopenia in the 2346–2353.
cardiovascular patient: diagnostic and treatment guidelines, Eur. J. Cardiothorac. [48] G.M. Arepally, Heparin-induced thrombocytopenia, Blood 129 (2017) 2864–2872.
Surg. 27 (2005) 138–149. [49] S. Khandelwal, J. Ravi, L. Rauova, et al., Polyreactive IgM initiates complement
[18] G. Murray, R. Mackenzie, The effect of heparin on portal thrombosis: its use in activation by PF4/heparin complexes through the classical pathway, Blood 132
mesenteric thrombosis and following splenectomy, Can. Med. Assoc. J. 41 (1939) (2018) 2431–2440.
38–40. [50] M.P. Mullen, D.L. Wessel, K.C. Thomas, et al., The incidence and implications of
[19] A.E. Macdonald, Heparin in thrombosis of the central vein, Trans. Am. anti-heparin-platelet factor 4 antibody formation in a pediatric cardiac surgical
Ophthalmol. Soc. 38 (1940) 313–326. population, Anesth. Analg. 107 (2008) 371–378.
[20] H.C. Hemker, A century of heparin: past, present and future, J. Thromb. Haemost. [51] S. Khandelwal, G.M. Lee, C.G. Hester, et al., The antigenic complex in HIT binds to
14 (2016) 2329–2338. B cells via complement and complement receptor 2 (CD21), Blood 128 (2016)
[21] A. Onishi, A.K. St, J.S. Dordick, R.J. Linhardt, Heparin and anticoagulation, Front. 1789–1799.
Biosci. (Landmark Ed) 21 (2016) 1372–1392. [52] P.W. Dempsey, M.E. Allison, S. Akkaraju, C.C. Goodnow, D.T. Fearon, C3d of
[22] J.L. Francis, G.R. Palmer, R. Moroose, A. Drexler, Comparison of bovine and complement as a molecular adjuvant: bridging innate and acquired immunity,
porcine heparin in heparin antibody formation after cardiac surgery, Ann. Thorac. Science 271 (1996) 348–350.
Surg. 75 (2003) 17–22. [53] Y. Zheng, M. Yu, A. Podd, et al., Critical role for mouse marginal zone B cells in
[23] S. Ahmad, Heparin-induced thrombocytopenia: impact of bovine versus porcine PF4/heparin antibody production, Blood 121 (2013) 3484–3492.
heparin in HIT pathogenesis, Front. Biosci. 12 (2007) 3312–3320. [54] K. Krauel, A. Schulze, R. Jouni, et al., Further insights into the anti-PF4/heparin
[24] R.J. Linhardt, S. Claude, Hudson award address in carbohydrate chemistry. IgM immune response, Thromb. Haemost. 115 (2016) 752–761.
Heparin: structure and activity, J. Med. Chem. 46 (2003) 2551–2564. [55] G.A. Denomme, T.E. Warkentin, P. Horsewood, J.A. Sheppard, M.N. Warner,
[25] S. Jonas, D. Quartermain, Low molecular weight heparin and the treatment of J.G. Kelton, Activation of platelets by sera containing IgG1 heparin-dependent
ischemic stroke. Animal results, the reasons for failure in human stroke trials, antibodies: an explanation for the predominance of the Fc gammaRIIa “low re-
mechanisms of action, and the possibilities for future use in stroke, Ann. N.Y. sponder” (his131) gene in patients with heparin-induced thrombocytopenia, J.
Acad. Sci. 939 (2001) 268–270. Lab. Clin. Med. 130 (1997) 278–284.
[26] S.T. Olson, I. Bjork, R. Sheffer, P.A. Craig, J.D. Shore, J. Choay, Role of the an- [56] S.I. Rosenfeld, R.J. Looney, J.P. Leddy, D.C. Phipps, G.N. Abraham, C.L. Anderson,
tithrombin-binding pentasaccharide in heparin acceleration of antithrombin-pro- Human platelet Fc receptor for immunoglobulin G. Identification as a 40,000-
teinase reactions. Resolution of the antithrombin conformational change con- molecular-weight membrane protein shared by monocytes, J. Clin. Invest. 76
tribution to heparin rate enhancement, J. Biol. Chem. 267 (1992) 12528–12538. (1985) 2317–2322.
[27] D.R. Al, R. Wagenvoord, G.W. van Dedem, S. Beguin, H.C. Hemker, The inhibition [57] V. Tutwiler, D. Madeeva, H.S. Ahn, et al., Platelet transactivation by monocytes
of blood coagulation by heparins of different molecular weight is caused by a promotes thrombosis in heparin-induced thrombocytopenia, Blood 127 (2016)
common functional motif–the C-domain, J. Thromb. Haemost. 1 (2003) 907–914. 464–472.
[28] H.C. Hemker, D.R. Al, R. Wagenvoord, S. Beguin, The Choay domain – the [58] S.P. Karas, W.F. Rosse, R.J. Kurlander, Characterization of the IgG-Fc receptor on
structure responsible for the anticoagulant action of heparins, Bull. Acad. Natl. human platelets, Blood 60 (1982) 1277–1282.
Med. 187 (59–66) (2003) 66–67. [59] D.B. Cines, A. Tomaski, S. Tannenbaum, Immune endothelial-cell injury in he-
[29] S.T. Olson, I. Bjork, Role of protein conformational changes, surface approxima- parin-associated thrombocytopenia, N. Engl. J. Med. 316 (1987) 581–589.
tion and protein cofactors in heparin-accelerated antithrombin-proteinase reac- [60] M. Arman, K. Krauel, Human platelet IgG Fc receptor FcgammaRIIA in immunity
tions, Adv. Exp. Med. Biol. 313 (1992) 155–165. and thrombosis, J. Thromb. Haemost. 13 (2015) 893–908.
[30] G.F. Briginshaw, J.N. Shanberge, Identification of two distinct heparin cofactors in [61] L. Alberio, Heparin-induced thrombocytopenia: some working hypotheses on pa-
human plasma. II. Inhibition of thrombin and activated factor X, Thromb. Res. 4 thogenesis, diagnostic strategies and treatment, Curr. Opin. Hematol. 15 (2008)
(1974) 463–477. 456–464.
[31] B. Mulloy, Structure and physicochemical characterisation of heparin, Handb. Exp. [62] A.J. Moroi, S.P. Watson, Impact of the PI3-kinase/Akt pathway on ITAM and
Pharmacol. (2012) 77–98. hemITAM receptors: haemostasis, platelet activation and antithrombotic therapy,
[32] J. Leentjens, M. Peters, A.C. Esselink, Y. Smulders, C. Kramers, Initial antic- Biochem. Pharmacol. 94 (2015) 186–194.
oagulation in patients with pulmonary embolism: thrombolysis, unfractionated [63] C.E. Hargreaves, M.J. Rose-Zerilli, L.R. Machado, et al., Fcgamma receptors: ge-
heparin, LMWH, fondaparinux, or DOACs? Br. J. Clin. Pharmacol. 83 (2017) netic variation, function, and disease, Immunol. Rev. 268 (2015) 6–24.
2356–2366. [64] P.A. Warmerdam, J.G. van de Winkel, A. Vlug, N.A. Westerdaal, P.J. Capel, A
[33] S. Beitland, I. Sandven, L.K. Kjaervik, P.M. Sandset, K. Sunde, T. Eken, single amino acid in the second Ig-like domain of the human Fc gamma receptor II
Thromboprophylaxis with low molecular weight heparin versus unfractionated is critical for human IgG2 binding, J. Immunol. 147 (1991) 1338–1343.
heparin in intensive care patients: a systematic review with meta-analysis and trial [65] M.B. Urowitz, D. Gladman, D. Ibanez, et al., Clinical manifestations and coronary
sequential analysis, Intensive Care Med. 41 (2015) 1209–1219. artery disease risk factors at diagnosis of systemic lupus erythematosus: data from
[34] D.R. Junqueira, L.M. Zorzela, E. Perini, Unfractionated heparin versus low mole- an international inception cohort, Lupus 16 (2007) 731–735.
cular weight heparins for avoiding heparin-induced thrombocytopenia in post- [66] E.E. Gardiner, M. Al-Tamimi, F.T. Mu, et al., Compromised ITAM-based platelet
operative patients, Cochrane Database Syst. Rev. 4 (2017) D7557. receptor function in a patient with immune thrombocytopenic purpura, J.
[35] T.E. Warkentin, J.A. Sheppard, J.C. Moore, R.J. Cook, J.G. Kelton, Studies of the Thromb. Haemost. 6 (2008) 1175–1182.
immune response in heparin-induced thrombocytopenia, Blood 113 (2009) [67] G.M. Arepally, I.M. Mayer, Antibodies from patients with heparin-induced
4963–4969. thrombocytopenia stimulate monocytic cells to express tissue factor and secrete
[36] A. Greinacher, D. Juhl, U. Strobel, et al., Heparin-induced thrombocytopenia: a interleukin-8, Blood 98 (2001) 1252–1254.
prospective study on the incidence, platelet-activating capacity and clinical sig- [68] Z. Xiao, G.P. Visentin, K.M. Dayananda, S. Neelamegham, Immune complexes
nificance of antiplatelet factor 4/heparin antibodies of the IgG, IgM, and IgA formed following the binding of anti-platelet factor 4 (CXCL4) antibodies to
classes, J. Thromb. Haemost. 5 (2007) 1666–1673. CXCL4 stimulate human neutrophil activation and cell adhesion, Blood 112 (2008)
[37] J. Qiao, M. Al-Tamimi, R.I. Baker, R.K. Andrews, E.E. Gardiner, The platelet Fc 1091–1100.
receptor, FcgammaRIIa, Immunol. Rev. 268 (2015) 241–252. [69] M. Joglekar, S. Khandelwal, D.B. Cines, M. Poncz, L. Rauova, G.M. Arepally,
[38] I. Palomo, J. Pereira, M. Alarcon, et al., Prevalence of heparin-induced antibodies Heparin enhances uptake of platelet factor 4/heparin complexes by monocytes
in patients with chronic renal failure undergoing hemodialysis, J. Clin. Lab. Anal. and macrophages, J. Thromb. Haemost. 13 (2015) 1416–1427.
19 (2005) 189–195. [70] K. De Ceunynck, C.G. Peters, A. Jain, et al., PAR1 agonists stimulate APC-like
[39] L.A. Haile, R. Rao, S.K. Polumuri, et al., PF4-HIT antibody (KKO) complexes ac- endothelial cytoprotection and confer resistance to thromboinflammatory injury,
tivate broad innate immune and inflammatory responses, Thromb. Res. 159 Proc. Natl. Acad. Sci. USA 115 (2018) E982–E991.
(2017) 39–47. [71] D.A. Morrow, E. Braunwald, M.P. Bonaca, et al., Vorapaxar in the secondary
[40] C. Pouplard, S. Iochmann, B. Renard, et al., Induction of monocyte tissue factor prevention of atherothrombotic events, N. Engl. J. Med. 366 (2012) 1404–1413.
expression by antibodies to heparin-platelet factor 4 complexes developed in he- [72] M.L. Kahn, Y.W. Zheng, W. Huang, et al., A dual thrombin receptor system for
parin-induced thrombocytopenia, Blood 97 (2001) 3300–3302. platelet activation, Nature 394 (1998) 690–694.
[41] J. Jian, Y. Pang, H.H. Yan, et al., Platelet factor 4 is produced by subsets of [73] A. Tello-Montoliu, S.D. Tomasello, M. Ueno, D.J. Angiolillo, Antiplatelet therapy:
myeloid cells in premetastatic lung and inhibits tumor metastasis, Oncotarget 8 thrombin receptor antagonists, Br. J. Clin. Pharmacol. 72 (2011) 658–671.
(2017) 27725–27739. [74] S.L. Jacques, M. LeMasurier, P.J. Sheridan, S.K. Seeley, A. Kuliopulos, Substrate-

78
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

assisted catalysis of the PAR1 thrombin receptor. Enhancement of macromolecular Haemost. 109 (2013) 471–478.
association and cleavage, J. Biol. Chem. 275 (2000) 40671–40678. [105] L. Hao, Q. Zhang, T. Yu, L. Yu, Y. Cheng, Modulation of ultra-low-molecular-
[75] A.J. Leger, S.L. Jacques, J. Badar, et al., Blocking the protease-activated receptor weight heparin on [Ca(2)(+)]i in nervous cells, Brain Res. Bull. 86 (2011)
1–4 heterodimer in platelet-mediated thrombosis, Circulation 113 (2006) 355–359.
1244–1254. [106] N.O. Badger, Fondaparinux (Arixtra(R)), a safe alternative for the treatment of
[76] T.K. Vu, D.T. Hung, V.I. Wheaton, S.R. Coughlin, Molecular cloning of a functional patients with heparin-induced thrombocytopenia? J. Pharm. Pract. 23 (2010)
thrombin receptor reveals a novel proteolytic mechanism of receptor activation, 235–238.
Cell 64 (1991) 1057–1068. [107] M.R. Lassen, O.E. Dahl, P. Mismetti, D. Destree, A.G. Turpie, AVE5026, a new
[77] R. Flaumenhaft, K. De Ceunynck, Targeting PAR1: now What? Trends Pharmacol. hemisynthetic ultra-low-molecular-weight heparin for the prevention of venous
Sci. 38 (2017) 701–716. thromboembolism in patients after total knee replacement surgery–TREK: a dose-
[78] K. Al-Khafaji, M. Mutyala, N. Al-Khafaji, et al., Protease-activated receptor 1 in- ranging study, J. Thromb. Haemost. 7 (2009) 566–572.
hibitors: novel antiplatelet drugs in prevention of atherothrombosis, Am. J. Ther. [108] M. Klingele, H. Bomberg, A. Lerner-Graber, et al., Use of argatroban: experiences
24 (2017) e730–e736. in continuous renal replacement therapy in critically ill patients after cardiac
[79] T. Pouget, X. Pillois, M. Fiore, Adenylate cyclase inhibition is required for normal surgery, J. Thorac. Cardiovasc. Surg. 147 (2014) 1918–1924.
redistribution of platelet surface GPIb in response to PAR1 activation, Thromb. [109] L. Koltowski, J. Legutko, K.J. Filipiak, et al., Bivalirudin use in acute coronary
Res. 173 (2019) 151–154. syndrome patients undergoing percutaneous coronary interventions in Poland:
[80] L. Covic, C. Singh, H. Smith, A. Kuliopulos, Role of the PAR4 thrombin receptor in clinical update from expert group of the Association on Cardiovascular
stabilizing platelet-platelet aggregates as revealed by a patient with Hermansky- Interventions of the Polish Cardiac Society, Cardiol. J. 26 (2019) 1–7.
Pudlak syndrome, Thromb. Haemost. 87 (2002) 722–727. [110] T.E. Warkentin, A. Koster, Bivalirudin: a review, Expert Opin. Pharmacother. 6
[81] P. Lova, F. Campus, R. Lombardi, et al., Contribution of protease-activated re- (2005) 1349–1371.
ceptors 1 and 4 and glycoprotein Ib-IX-V in the G(i)-independent activation of [111] F. Sanfilippo, S. Asmussen, D.M. Maybauer, et al., Bivalirudin for alternative an-
platelet Rap1B by thrombin, J. Biol. Chem. 279 (2004) 25299–25306. ticoagulation in extracorporeal membrane oxygenation: a systematic review, J.
[82] T. Kosoglou, L. Reyderman, R.G. Tiessen, et al., Pharmacodynamics and phar- Intensive Care Med 32 (2017) 312–319.
macokinetics of the novel PAR-1 antagonist vorapaxar (formerly SCH 530348) in [112] A. Koster, D. Faraoni, J.H. Levy, Argatroban and bivalirudin for perioperative
healthy subjects, Eur. J. Clin. Pharmacol. 68 (2012) 249–258. anticoagulation in cardiac surgery, Anesthesiology 128 (2018) 390–400.
[83] M.L. O'Donoghue, D.L. Bhatt, S.D. Wiviott, et al., Safety and tolerability of ato- [113] T.E. Warkentin, A. Greinacher, A. Koster, Bivalirudin, Thromb. Haemost. 99
paxar in the treatment of patients with acute coronary syndromes: the lessons from (2008) 830–839.
antagonizing the cellular effects of Thrombin-Acute Coronary Syndromes Trial, [114] E. Ljajikj, A. Zittermann, M. Morshuis, et al., Bivalirudin anticoagulation for left
Circulation 123 (2011) 1843–1853. ventricular assist device implantation on an extracorporeal life support system in
[84] V.L. Serebruany, M.H. Kim, D.F. Hanley, Vorapaxar monotherapy for secondary patients with heparin-induced thrombocytopenia antibodies, Interact Cardiovasc.
stroke prevention: a call for randomized trial, Int. J. Stroke 11 (2016) 614–617. Thorac. Surg. 25 (2017) 898–904.
[85] J.F. Arthur, J. Qiao, Y. Shen, et al., ITAM receptor-mediated generation of reactive [115] R. Robson, The use of bivalirudin in patients with renal impairment, J. Invasive
oxygen species in human platelets occurs via Syk-dependent and Syk-independent Cardiol. 12 (Suppl F) (2000) 33F–36F.
pathways, J. Thromb. Haemost. 10 (2012) 1133–1141. [116] S. Kianoush, B. Bikdeli, M.M. Desai, J.W. Eikelboom, Risk of stent thrombosis and
[86] D.B. Cines, S.V. Yarovoi, S.V. Zaitsev, et al., Polyphosphate/platelet factor 4 major bleeding with bivalirudin compared with active control: a systematic review
complexes can mediate heparin-independent platelet activation in heparin-in- and meta-analysis of randomized trials, Thromb. Res. 136 (2015) 1087–1098.
duced thrombocytopenia, Blood Adv. 1 (2016) 62–74. [117] H. Tamez, D.S. Pinto, A.J. Kirtane, et al., Effect of short procedural duration with
[87] A. Cuker, P.A. Gimotty, M.A. Crowther, T.E. Warkentin, Predictive value of the 4Ts bivalirudin on increased risk of acute stent thrombosis in patients with STEMI,
scoring system for heparin-induced thrombocytopenia: a systematic review and Jama Cardiol. 2 (2017) 673.
meta-analysis, Blood 120 (2012) 4160–4167. [118] S.P. Hoole, N.E. West, Bivalirudin in the treatment of acute coronary syndrome,
[88] T. Thomas, A. Fuentes, Q. Xu, K. Donahue, Evaluation of heparin induced BMJ 352 (2016) i86.
thrombocytopenia using probability scores in a mechanical circulatory support [119] S.P. Marso, A.P. Amin, J.A. House, et al., Association between use of bleeding
population, J. Thromb. Thrombol. (2019). avoidance strategies and risk of periprocedural bleeding among patients under-
[89] E.A. Obeng, K.M. Harney, T. Moniz, A. Arnold, E.J. Neufeld, C.R. Trenor, Pediatric going percutaneous coronary intervention, JAMA 303 (2010) 2156–2164.
heparin-induced thrombocytopenia: prevalence, thrombotic risk, and application [120] S.K. Swan, M.J. Hursting, The pharmacokinetics and pharmacodynamics of ar-
of the 4Ts scoring system, J. Pediatr. 166 (2015) 144–150. gatroban: effects of age, gender, and hepatic or renal dysfunction,
[90] M. Crowther, D. Cook, G. Guyatt, et al., Heparin-induced thrombocytopenia in the Pharmacotherapy 20 (2000) 318–329.
critically ill: interpreting the 4Ts test in a randomized trial, J. Crit. Care 29 (2014) [121] K. McKeage, G.L. Plosker, Argatroban, Drugs 61 (515–522) (2001) 523–524.
470–477. [122] T. Richey, H. Iwata, H. Oowaki, E. Uchida, S. Matsuda, Y. Ikada, Surface mod-
[91] T.E. Warkentin, N.M. Heddle, Laboratory diagnosis of immune heparin-induced ification of polyethylene balloon catheters for local drug delivery, Biomaterials 21
thrombocytopenia, Curr. Hematol. Rep. 2 (2003) 148–157. (2000) 1057–1065.
[92] T.E. Warkentin, Anti-platelet factor 4/heparin antibodies in orthopedic surgery [123] R.W. Yeh, I.K. Jang, Argatroban: update, Am. Heart J. 151 (2006) 1131–1138.
patients receiving antithrombotic prophylaxis with fondaparinux or enoxaparin, [124] J.M. Walenga, S. Ahmad, D. Hoppensteadt, O. Iqbal, M.J. Hursting, B.E. Lewis,
Blood 106 (2005) 3791–3796. Argatroban therapy does not generate antibodies that alter its anticoagulant ac-
[93] T.E. Warkentin, A. Greinacher, A. Koster, A.M. Lincoff, Treatment and prevention tivity in patients with heparin-induced thrombocytopenia, Thromb. Res. 105
of heparin-induced thrombocytopenia: American College of Chest Physicians (2002) 401–405.
Evidence-Based Clinical Practice Guidelines (8th Edition), Chest 133 (2008) [125] E. Mohanty, S. Nazir, J.I. Sheppard, D.A. Forman, T.E. Warkentin, High-dose in-
340S–380S. travenous immunoglobulin to treat spontaneous heparin-induced thrombocyto-
[94] A. Davenport, Antibodies to heparin-platelet factor 4 complex: pathogenesis, penia syndrome, J. Thromb. Haemost. (2019).
epidemiology, and management of heparin-induced thrombocytopenia in hemo- [126] M. Giorgi-Pierfranceschi, N. Mumoli, R. Scarpioni, E. Croci, F. Dentali, Argatroban
dialysis, Am. J. Kidney Dis. 54 (2009) 361–374. for treatment of heparin-induced thrombocytopenia and thrombosis in a patient
[95] J. Hirsh, M.N. Levine, Low molecular weight heparin, Blood 79 (1992) 1–17. with multiple myeloma undergoing hemodialysis, J. Am. Geriatr. Soc. 64 (2016)
[96] G.R. Hetzel, C. Sucker, The heparins: all a nephrologist should know, Nephrol. 1748–1750.
Dial. Transplant. 20 (2005) 2036–2042. [127] A. Alatri, A.E. Armstrong, A. Greinacher, et al., Results of a consensus meeting on
[97] N. Martel, J. Lee, P.S. Wells, Risk for heparin-induced thrombocytopenia with the use of argatroban in patients with heparin-induced thrombocytopenia re-
unfractionated and low-molecular-weight heparin thromboprophylaxis: a meta- quiring antithrombotic therapy - a European Perspective, Thromb. Res. 129
analysis, Blood 106 (2005) 2710–2715. (2012) 426–433.
[98] M.N. Levine, J. Hirsh, M. Gent, et al., Prevention of deep vein thrombosis after [128] B.M. Everett, R. Yeh, S.Y. Foo, et al., Prevalence of heparin/platelet factor 4 an-
elective hip surgery. A randomized trial comparing low molecular weight heparin tibodies before and after cardiac surgery, Ann. Thorac. Surg. 83 (2007) 592–597.
with standard unfractionated heparin, Ann. Intern. Med. 114 (1991) 545–551. [129] B.T. Samuelson, A. Cuker, Measurement and reversal of the direct oral antic-
[99] F. Atiq, P.M. van den Bemt, F.W. Leebeek, T. van Gelder, J. Versmissen, A sys- oagulants, Blood Rev. 31 (2017) 77–84.
tematic review on the accumulation of prophylactic dosages of low-molecular- [130] T.E. Warkentin, M. Pai, L.A. Linkins, Direct oral anticoagulants for treatment of
weight heparins (LMWHs) in patients with renal insufficiency, Eur. J. Clin. HIT: update of Hamilton experience and literature review, Blood 130 (2017)
Pharmacol. 71 (2015) 921–929. 1104–1113.
[100] J. Hirsh, S.S. Anand, J.L. Halperin, V. Fuster, Guide to anticoagulant therapy: [131] J.W. Skelley, J.A. Kyle, R.A. Roberts, Novel oral anticoagulants for heparin-in-
Heparin: a statement for healthcare professionals from the American Heart duced thrombocytopenia, J. Thromb. Thrombol. 42 (2016) 172–178.
Association, Circulation 103 (2001) 2994–3018. [132] M.A. Miyares, K.A. Davis, Direct-acting oral anticoagulants as emerging treatment
[101] J. Hirsh, T.E. Warkentin, S.G. Shaughnessy, et al., Heparin and low-molecular- options for heparin-induced thrombocytopenia, Ann. Pharmacother. 49 (2015)
weight heparin: mechanisms of action, pharmacokinetics, dosing, monitoring, 735–739.
efficacy, and safety, Chest 119 (2001) 64S–94S. [133] A.E. Burnett, C.E. Mahan, S.R. Vazquez, L.B. Oertel, D.A. Garcia, J. Ansell,
[102] Y. Xu, S. Masuko, M. Takieddin, et al., Chemoenzymatic synthesis of homogeneous Guidance for the practical management of the direct oral anticoagulants (DOACs)
ultralow molecular weight heparins, Science 334 (2011) 498–501. in VTE treatment, J. Thromb. Thrombol. 41 (2016) 206–232.
[103] Z. Liu, S. Ji, J. Sheng, F. Wang, Pharmacological effects and clinical applications of [134] J. Lekura, J.S. Kalus, Overview of betrixaban and its role in clinical practice, Am.
ultra low molecular weight heparins, Drug Discov. Ther. 8 (2014) 1–10. J. Health Syst. Pharm. 75 (2018) 1095–1102.
[104] M.A. Lima, C. Viskov, F. Herman, et al., Ultra-low-molecular-weight heparins: [135] L.A. Linkins, A.L. Dans, L.K. Moores, et al., Treatment and prevention of heparin-
precise structural features impacting specific anticoagulant activities, Thromb. induced thrombocytopenia: antithrombotic therapy and prevention of thrombosis,

79
P. Zhou, et al. Clinica Chimica Acta 504 (2020) 73–80

9th ed: American College of Chest Physicians Evidence-Based Clinical Practice [144] S. Yanagisawa, Y. Inden, A. Fujii, et al., Renal function and risk of stroke and
Guidelines, Chest 141 (2012) e495S–e530. bleeding in patients undergoing catheter ablation for atrial fibrillation:
[136] L. Joseph, A.I. Casanegra, M. Dhariwal, et al., Bivalirudin for the treatment of Comparison between uninterrupted direct oral anticoagulants and warfarin ad-
patients with confirmed or suspected heparin-induced thrombocytopenia, J. ministration, Heart Rhythm 15 (2018) 348–354.
Thromb. Haemost. 12 (2014) 1044–1053. [145] C. Kizlik-Masson, Q. Deveuve, Y. Zhou, et al., Cleavage of anti-PF4/Heparin IgG by
[137] M. Schindewolf, J. Steindl, J. Beyer-Westendorf, et al., Frequent off-label use of a bacterial protease and potential benefit in heparin-induced thrombocytopenia,
fondaparinux in patients with suspected acute heparin-induced thrombocytopenia Blood (2019).
(HIT)–findings from the GerHIT multi-centre registry study, Thromb. Res. 134 [146] R.J. Brezski, R.E. Jordan, Cleavage of IgGs by proteases associated with invasive
(2014) 29–35. diseases: an evasion tactic against host immunity? MABS-AUSTIN 2 (2010)
[138] P.N. Tran, M.H. Tran, Emerging role of direct oral anticoagulants in the man- 212–220.
agement of heparin-induced thrombocytopenia, Clin. Appl. Thromb. Hemost. 24 [147] A.J. Gearing, S.J. Thorpe, K. Miller, et al., Selective cleavage of human IgG by the
(2018) 201–209. matrix metalloproteinases, matrilysin and stromelysin, Immunol. Lett. 81 (2002)
[139] L.A. Linkins, T.E. Warkentin, Rivaroxaban for treatment of HIT: a riveting first 41–48.
experience, Thromb. Res. 135 (2015) 1–2. [148] M.H. Ryan, D. Petrone, J.F. Nemeth, E. Barnathan, L. Bjorck, R.E. Jordan,
[140] J.M. Walenga, M. Prechel, D. Hoppensteadt, et al., Apixaban as an alternate oral Proteolysis of purified IgGs by human and bacterial enzymes in vitro and the
anticoagulant for the management of patients with heparin-induced thrombocy- detection of specific proteolytic fragments of endogenous IgG in rheumatoid sy-
topenia, Clin. Appl. Thromb. Hemost. 19 (2013) 482–487. novial fluid, Mol. Immunol. 45 (2008) 1837–1846.
[141] J.M. Walenga, M. Prechel, W.P. Jeske, et al., Rivaroxaban–an oral, direct Factor Xa [149] S.A. Arif, J. D'Souza, M. Gil, S. Gim, Vorapaxar for reduction of thrombotic car-
inhibitor–has potential for the management of patients with heparin-induced diovascular events in myocardial infarction and peripheral artery disease, Am. J.
thrombocytopenia, Br. J. Haematol. 143 (2008) 92–99. Health Syst. Pharm. 72 (2015) 1615–1622.
[142] A. Kantorovich, Renal function and direct oral anticoagulant dose reduction, Am. [150] S.D. Wiviott, M.D. Flather, M.L. O'Donoghue, et al., Randomized trial of atopaxar
J. Med. 130 (2017) e363. in the treatment of patients with coronary artery disease: the lessons from an-
[143] J. Fanikos, A.E. Burnett, C.E. Mahan, P.P. Dobesh, Renal function and direct oral tagonizing the cellular effect of Thrombin-Coronary Artery Disease Trial,
anticoagulant treatment for venous thromboembolism, Am. J. Med. 130 (2017) Circulation 123 (2011) 1854–1863.
1137–1143.

80

You might also like