You are on page 1of 15

review

Pathogenesis and management of antiphospholipid syndrome

Deepa R.J. Arachchillage and Mike Laffan

Department of Haematology, Imperial College Healthcare NHS Trust and Imperial College London, Hammersmith Hospital, London,
UK

antiphospholipid antibodies (aPL) (Miyakis et al, 2006)


Summary
(Table I). Vascular thrombosis can be arterial, venous, or
Antiphospholipid antibodies are a heterogeneous group of small vessel, in any tissue or organ and must be objectively
autoantibodies that have clear associations with thrombosis confirmed (i.e. unequivocal findings of appropriate imaging
and pregnancy morbidity, and which together constitute the studies or histopathology). Thrombosis in APS should show
‘antiphospholipid syndrome’ (APS). However, the patho- no signs of inflammation in the vessel wall (Miyakis et al,
physiology of these complications is not well understood and 2006). Although many patients with APS have an associated
their heterogeneity suggests that more than one pathogenic autoimmune disorder, thrombotic events in APS are not
process may be involved. Diagnosis remains a combination accompanied by histological evidence of vessel wall inflam-
of laboratory analysis and clinical observation but there have mation. Nonetheless, inflammatory mediators and inflamma-
been significant advances in identifying specific pathogenic tory responses in endothelial cells, monocytes and
features, such as domain I-specific anti-b2-glycoprotein-I neutrophils are implicated in APS pathogenesis. The Interna-
antibodies. This in turn has pointed to endothelial and com- tional Consensus Criteria (ICC: Sydney; updated Sapporo)
plement activation as important factors in the pathogenesis for APS (Miyakis et al, 2006) were designed for scientific
of APS. Consequently, although anticoagulation remains the clinical studies but have been adapted for the diagnosis of
standard treatment for thrombotic APS and during preg- APS in routine clinical practice. The British Committee for
nancy, the realisation that these additional pathways are Standards in Haematology (BCSH) guidelines (Keeling et al,
involved in the pathogenesis of APS has significant implica- 2012) adopt similar criteria for the diagnosis of both throm-
tions for treatment: agents acting outside the coagulation botic and obstetric APS. Non-criteria features of APS, such
system, such as hydroxychloroquine for pregnancy complica- as heart valve disease, livedo reticularis, thrombocytopenia
tions and sirolimus as an inhibitor of the mammalian target and nephropathy, may present in association with thrombo-
of rapamycin (mTOR) pathway, are now under evaluation sis and/or pregnancy morbidity or as isolated features. The
and represent a radical change in thinking for haematolo- APS Clinical Features Task Force of the 14th International
gists. Conventional anticoagulation is also under challenge Congress on aPL analysed the relevance of the most frequent
from new, direct acting anticoagulants. This review will pro- non-criteria manifestations and concluded that available data
vide a comprehensive overview of the evolving understanding support the inclusion of thrombocytopenia, heart valve dis-
of APS pathogenesis and how this and novel therapeutics will ease, renal microangiopathy (aPL nephropathy), chorea, and
alter diagnosis and management. longitudinal myelitis as part of the APS Classification Criteria
(Abreu et al, 2015). However, these features are not yet
Keywords: antiphospholipid antibodies, thrombosis, war- included in the ICC. APS can occur in isolation or in associ-
farin, pregnancy complications, complement, immunosup- ation with other autoimmune disorders, such as systemic
pression. lupus erythematous (SLE) or rheumatoid arthritis (Miyakis
et al, 2006).
Antiphospholipid syndrome (APS) is characterized by vascu- Antiphospholipid antibodies are a heterogeneous group of
lar thrombosis and/or pregnancy loss or morbidity in associ- autoantibodies, which include lupus anticoagulant (LA),
ation with persistent positivity of autoantibodies known as immunoglobulin (Ig)G and IgM anticardiolipin antibodies
(aCL) and anti-b2-glycoprotein-I (anti-b2GPI) antibodies.
Their primary targets are phospholipid-binding proteins,
although antibodies directed against phospholipids and other
Correspondence: Dr Deepa RJ Arachchillage, Department of proteins also occur. In the early 1950’s Conley and Hart-
haematology, Imperial College Healthcare NHS Trust and Imperial mann (1952) wrote a brief report describing two patients
College London,, Hammersmith Hospital, 4th Floor, Commonwealth with SLE and a ‘peculiar haemorrhagic disorder’ with pro-
Building, Du Cane Road, London W12 0NN, UK. longed blood clotting times and clear evidence of an
E-mail: d.arachchillage@imperial.ac.uk

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology doi: 10.1111/bjh.14632
Review

Table I. The international consensus (revised Sapporo) criteria for diagnosis of obstetric antiphospholipid syndrome.

Clinical criteria Laboratory criteria

1 One or more unexplained deaths of a morphologically normal 1 LA present in plasma, on two or more occasions at least 12 weeks
fetus at or beyond the 10th week of gestation apart
2 One or more pre-term births of a morphologically normal neonate 2 aCL of immunoglobulin (Ig)G and/or IgM isotype in serum or
before the 34th week of gestation because of: plasma, present in medium or high titre (i.e. >40 GPL units or
MPL units, or > the 99th centile), on two or more occasions,
(i) eclampsia or severe pre-eclampsia or
at least 12 weeks apart
(ii) recognized features of placental insufficiency
3 anti-b2GPI of IgG and/or IgM isotype in serum or plasma
3 Three or more unexplained consecutive spontaneous miscarriages (in titre >the 99th centile), present on two or more occasions at
before the 10th week of gestation, with maternal anatomic or least 12 weeks apart
hormonal abnormalities and paternal and maternal
chromosomal causes excluded

OAPS is diagnosed if at least one of the clinical criteria and one of the laboratory criteria are met

aCL, anticardiolipin antibodies; anti-b2GPI, anti-b2glycoprotein-I antibodies; GPL, IgG antiphospholipid units; LA, lupus anticoagulants; MPL,
IgM antiphospholipid units; OAPS, Obstetric antiphospholipid syndrome.

anticoagulant in plasma mixing studies. As this inhibitor was key events in APS are summarised in Fig 1. The aim of this
predominantly found in patients with SLE, the in vitro anti- review is to examine the evolving understanding of APS
coagulant phenomenon was called LA. The paradoxical cor- pathogenesis and how this and other therapeutic options will
relation between vascular thrombosis and LA was first change the way we manage this potentially devastating disor-
described by Bowie et al (1963) and was followed by many der.
others. Originally, it was thought that these autoantibodies
bound to phospholipid; hence ‘antiphospholipid antibodies’
Pathogenesis of Antiphospholipid syndrome
but in the 1990s it was shown that aPL recognized phospho-
lipids indirectly via phospholipid-binding plasma proteins Despite clear associations between aPL and thrombosis and
(Galli et al, 1990). Although many plasma proteins have been with pregnancy morbidity, the pathophysiology of these
found to be antibody ligands in APS, antibodies against complications is not well understood, with their heterogene-
b2GPI have the most significant association with pathogenic- ity suggesting that more than one pathogenic process is
ity (Bas de Laat et al, 2004). Some authors have challenged involved. Moreover, even though aPL are persistently present
the primacy of anti-b2GPI antibodies in the pathogenesis of in the systemic circulation, thrombotic events occur only
APS (Lackner & M€ uller-Calleja, 2016) and shown that some occasionally, suggesting that the development of aPL is a
co-factor independent antibodies can induce thrombus for- necessary, but not sufficient step in the development of APS
mation in a mouse model (Manukyan et al, 2016). and that other factors play a role. Such ‘second hits’ or ‘trig-
The laboratory diagnosis of APS requires the presence of gers’ probably push the thrombotic/haemostatic balance in
LA in plasma or aCL of IgG and/or IgM isotype in serum or favour of thrombosis and may include infection, endothelial
plasma, present in medium or high titre (i.e. >40 IgG injury, inflammation, immunological and other non-immu-
antiphospholipid units [GPL] or IgM antiphospholipid units nological procoagulant factors, such as oestrogen-containing
[MPL], or >the 99th centile) or anti-b2GPI of IgG and/or contraceptive pills, surgery and immobility (Pengo et al,
IgM isotype in serum or plasma (in titre >the 99th centile). 2011). The patient’s genetic constitution, in relation to genes
The positive results must be obtained on two or more occa- for inflammatory mediators, may also be a critical variable in
sions, at least 12 weeks apart (Miyakis et al, 2006). Inter- the development of clinical APS manifestations.
laboratory variability remains a problem in the performance
of the aCL and anti-b2GPI enzyme-linked immunosorbent
Antibody specificities and evidence for pathogenicity of
assays (ELISAs) due to the lack of uniformity in reference
anti-b2-GPI
materials for calibration.
Nonetheless, identification of these antibodies has allowed Mcneil et al (1990) found that the binding of aPL to cardi-
development of diagnostic testing and investigation of patho- olipin requires the presence of b2GPI as a cofactor. b2GPI is
genic mechanisms. These in turn offer additional therapeutic an apolipoprotein, a member of the complement control
options, which are currently under investigation. Even con- family and is considered to be a natural inhibitor of coagula-
ventional anticoagulant therapy is under review while the tion. In addition, in vitro studies have shown that it has anti-
role of direct acting oral anticoagulants is investigated. The angiogenic (Yu et al, 2008) and anti-apoptotic (Maiti et al,

2 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

Fig 1. Key events in the history of Antiphospholipid syndrome (Adapted from figure originally drawn by Prof. Mike Greaves, with permission).
APS, antiphosphlipid syndrome; DOACs, direct acting oral anticoagulants; SLE, systemic lupus erythematosus.

Fig 2. Schematic representation of folded (cir-


cular) and unfolded conformation of beta2 gly-
coprotein I (b2GPI) and subsequent binding to
anti-b2GPI antibodies (Adapted from van Os
et al, 2011). In the presence of anionic phos-
pholipids, the circular conformation of the
protein unfolds, exposing antigenic determi-
nants that are normally shielded from the cir-
culation (A–C) which facilitate the binding to
antibodies (D).

2008) activities. It consists of 326 amino acids, arranged in 5 Ioannou et al, 2007); which will not bind to the closed circu-
highly homologous complement-control protein domains lating form. Unfolding of b2GPI on the endothelial surface
that are designated I to V from the N- to the C-terminus. may be facilitated by release of molecules, such as thiore-
b2GPI can adopt two different conformations: a circular con- doxin, in response to oxidative stress, which can reduce sur-
formation in plasma, maintained by interaction between the face thiols on b2GPI. Binding of b2GPI on endothelial cells
first and fifth domain and an ‘activated’ open conformation (EC) normally has a protective function against oxidative
(Agar et al, 2010). In the presence of anionic phospholipids, stress-induced cell injury (Ioannou et al, 2010, 2011) but
the circular conformation of the protein unfolds, exposing antibody binding fixes b2GPI in this open conformation on
antigenic determinants that are normally shielded from the the phospholipid surface and the antibody-b2GPI complexes
circulation (van Os et al, 2011) [Fig 2]. One of these ‘cryptic’ bind to a variety of receptors (e.g., Toll-like receptors 2 and
antigenic determinants, within domain I (DI), is the epitope 4, annexin A2 and GP1ba) on different cell types, including
for the pathological autoantibodies (de Laat et al, 2006; ECs, monocytes, trophoblasts and platelets (de Groot &

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 3


Review

Meijers, 2011). This binding may trigger intracellular sig- highest risk of thrombosis (de Laat et al, 2009), frequently
nalling and inflammatory responses. present in triple aPL-positive patients, are closely associated
Direct evidence for the pathogenicity of these antibodies with both thrombosis and pregnancy loss (de Laat et al,
comes from infusion of autoantibodies from APS patients to 2009) and increase thrombus size in mouse models (Peri-
mice with injured blood vessels showing that they potentiate cleous et al, 2015). Of note, about 30% of patients with anti-
thrombus formation (Arad et al, 2011). Specifically, purified b2GPI antibodies are negative for anti-DI–b2GPI antibodies
anti-b2GP1 IgG autoantibodies, but not anti-b2GPI depleted (Pengo et al, 2015) and the clinical significance of autoanti-
IgG or normal human IgG, potentiated thrombus size in a bodies reacting with epitopes other than DI was investigated
dose-dependent manner. Although several clinical studies in a multicentre study (Artenjak et al, 2015). This study
and systematic reviews suggested that LA is a stronger risk analysed serum samples from 201 autoimmune patients with
factor for the development of thrombosis (Galli et al, 2003; IgG anti- b2GPI activity (87 APS, 67 APS plus SLE and 47
de Groot et al, 2005), other studies have shown that the with SLE only) for their binding to six b2GPI peptides corre-
presence of LA alone is not associated with thromboembolic sponding to amino acid clusters on domains I-II, II, III and
events (Pengo et al, 2005; Pengo et al, 2007). Similar results III-IV. The results showed a diverse clinical association with
were obtained in the Leiden Thrombophila case–control reactivity to different epitopes on b2GPI, suggesting all
study (de Groot et al, 2005) where positive LA with negative domains were relevant. Therefore, more detailed profiling of
anti-b2GPI or antiprothrombin ELISAs was not a risk for domain specificity and avidity of anti-b2GPI antibodies may
deep vein thrombosis (DVT [odds ratio (OR) 13, 95% con- be useful as risk stratification for clinical events.
fidence interval (CI): 03–60]. However, a recent systematic Studies using cell cultures demonstrated that monocytes,
review and meta-analysis found that LA and aCL antibodies ECs and platelets can also be activated by aPL with anti-
were associated with an increased risk of venous thromboem- b2GPI activity inducing the expression of intercellular cell
bolism (VTE) [OR = 614 (CI 274; 138) and OR = 146 (CI adhesion molecule-1 (ICAM-1), vascular cell adhesion mole-
106; 203) respectively] but there was a non-significant trend cule-1 (VCAM-1) and E-selectin on EC, and of tissue factor
for anti-b2GPI (OR = 161 (CI 076; 343)] (Reynaud et al, (TF) on both EC and monocytes (Pierangeli et al, 2008).
2014). For arterial thrombosis, all three antibodies showed a Activated platelets increase expression of GPIIbIIIa and syn-
significant association: ORs for LA, aCL and anti-b2GPI were thesis of thromboxane A2 (Pierangeli et al, 2008). However,
358 (CI 129–992), 265 (CI 175–400) and 312 (CI 151– it is possible that cells in culture may not behave as they do
644) respectively (Reynaud et al, 2014). de Laat et al (2004) in vivo.
found that anti-b2GPI antibodies with LA activity are
responsible for the thromboembolic complications in APS
Non- criteria antibodies
(de Laat et al, 2004). Moreover, they showed that a subgroup
of anti-b2GPI recognizing epitope Gly40-Arg43 (G40-R43) in The role of so-called non-criteria antibodies, such as anti-
DI of the molecule cause LA and that their presence corre- phosphatidylserine/prothrombin, anti-phosphatidic acid,
lates strongly with thrombosis (de Laat et al, 2005). This has anti-vimentin/cardiolipin complex anti-protein C/S and a
been confirmed by various other studies (de Laat et al, 2009; variety of other autoantibodies with specificities, such as fac-
Pengo et al, 2015). tor XII, factor X, Annexin A5 and Annexin A2, in the patho-
Consequently, these pathogenic antibodies will only bind genesis and diagnosis of APS remains to be established. The
purified b2GPI when coated onto an appropriate negatively- role of anti-DI-b2GPI antibodies in pathogenesis of APS is
charged surface and this has important implications for the discussed above. A small cross-sectional study demonstrated
design, selection and significance of diagnostic assays (de that anti-protein C antibodies are associated with resistance
Laat et al, 2005, 2006). It has been shown that exposure of to endogenous protein C activation and a severe thrombotic
the critical DI epitope G40-R43 on b2GPI is highly variable phenotype in APS (Arachchillage et al, 2014). Several studies
between commercial anti-b2GPI assays. As a consequence, have analysed the role of the IgA isotype of aPL. In particu-
false negatives can arise in assays characterized by a reduced lar, the pathogenicity of IgA aPL was established by demon-
exposure of G40-R43 (Pelkmans et al, 2013). strating that mice, injected with IgA aPL from patients with
The presence of triple aPL positivity, as defined by the APS, developed thrombosis (Pierangeli et al, 1995). In a
detection of LA and high titres of aCL and anti-b2GPI anti- recent systematic review to establish the prevalence of non-
bodies, correlates better with both thrombosis and pregnancy criteria antibodies and of resistance to Annexin A5 anticoag-
morbidity than any other aPL profile (Pengo et al, 2010): the ulant activity (AnxA5R) in APS and control populations, 16
risk of recurrent thrombosis in triple positive patients was retrospective studies of 1404 APS patients, (1839 disease con-
around 30% over a 6-year follow-up period (Pengo et al, trols and 797 healthy controls) were examined (Rodriguez-
2010). Triple-positive patients also have high titres of Garcia et al, 2015). It was found that IgA anti-b2GPI anti-
autoantibodies that bind the major B-cell epitope on DI of bodies (129/229, 563%) were most prevalent, followed by
the b2GPI molecule (Banzato et al, 2011). Thus anti-DI- AnxA5R (87/163, 534%) and IgG anti-Domain I (241/548,
b2GPI autoantibodies confer LA activity associated with the 440%) (Rodriguez-Garcia et al, 2015). However, in addition

4 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

to the retrospective data collection, the results were affected microvascular thrombotic lesions often recur. In cultured
by wide variation in the sample size, discrepancy in assay vascular ECs, IgG antibodies from patients with APS stimu-
methodology and the different cut-off levels used for assay lated the mammalian/mechanistic target of rapamycin
positivity. Prospective multicentre studies with adequate (mTOR) through the phosphatidylinositol 3-kinase (PI3K)–
sample size using more uniform methodology are essential to AKT pathway (Canaud et al, 2014) leading to cell prolifera-
determine the implications of these antibodies for the man- tion. It has been shown that mTOR pathway activation plays
agement of APS. a role in endothelial proliferation and intimal hyperplasia in
aPL positive patients, which leads to microthrombosis,
peripheral ischaemia, skin ulcers, diffuse alveolar haemor-
Pathogenic mechanisms involving other pathways
rhage or aPL nephropathy (Manning & Cantley, 2007). The
There is evidence from several animal models that comple- vascular endothelium of proliferating intrarenal vessels from
ment activation, with excess C3a and C5a generation, plays a patients with APS nephropathy showed evidence of mTOR
role in thrombotic manifestations in APS. Complement acti- complex (mTORC) pathway activation.
vation contributes to vascular inflammation and complement Another proposed mechanism of aPL-initiated thrombosis
inhibition may ameliorate aPL-induced thrombosis, offering is through interference with the anticoagulant activity of acti-
a potential new approach to therapy (Girardi et al, 2003). vated protein C (APC), resulting in acquired activated pro-
Mice treated with IgG from APS patients with high levels of tein C resistance (APCr) (Nojima et al, 2005): aPL inhibition
aPL and subjected to a femoral vein pinch model of throm- of the thrombomodulin mediated activation of protein C
bosis, developed larger thrombi and higher soluble TF (PC), as well as the anticoagulant activity of APC (Malia
activity than controls (Romay-Penabad et al, 2014). The co- et al, 1990), have been observed. Upregulation of the TF
administration of rEV576 (coversin), a recombinant protein pathway (Adams et al, 2001) by down regulation of its prin-
inhibitor of C5 activation, resulted in significantly smaller cipal inhibitor, TF pathway inhibitor (TFPI) (Liestol et al,
thrombi and reduced TF activity (Romay-Penabad et al, 2007), may also increase thrombotic risk and studies have
2014). Consistent with this hypothesis, low complement reported the presence of auto-antibodies against TFPI
levels (C3, C5) have been demonstrated in patients with APS (aTFPI) in APS patients (Adams et al, 2001). Many in vitro
(Oku et al, 2009). In a study of 186 patients with aPL, Breen studies have shown aPL-mediated upregulation of TF expres-
et al (2012) found significantly increased levels of fragment sion on monocytes (Lambrianides et al, 2010) and endothe-
Bb and C3a compared to normal controls. In the RAPS lial cells associated with inflammatory cytokines and
(Rivaroxaban in Antiphospholipid Syndrome) trial of 111 adhesion molecules (Willis et al, 2015); ab2GPI has also been
APS patients; it was demonstrated that APS patients with shown to suppress TFPI-dependent inhibition of the TF
previous VTE had significantly increased complement activa- pathway of coagulation (Boles & Mackman, 2010).
tion compared to normal controls, which was decreased by
rivaroxaban compared to warfarin therapy (Arachchillage
Pathogenesis of pregnancy complications in
et al, 2016a).
APS
Catastrophic APS (CAPS) is a rare and potentially fatal
variant of APS characterized by sudden onset of extensive The presence of aPL probably constitutes the single most
microvascular thrombosis at multiple sites leading to multi- recognisable risk factor in the majority of cases of recurrent
organ failure (Cervera et al, 2014). Widespread complement pregnancy loss and late placenta-mediated obstetric compli-
activation may contribute to CAPS and this is supported by cations. Animal studies have demonstrated that the passive
individual case reports of patients unresponsive to anticoagu- transfer of aPL promotes fetal loss and placental thrombosis
lation and immunosuppression, but successfully treated with and also inhibits trophoblast and decidual cell functions
C5 complement inhibitor eculizumab (Shapira et al, 2012). in vitro (Branch et al, 1990). Cross-species investigations
As discussed below, complement activation may also be a have shown that the exposure of pregnant rats to a purified
central mechanism in aPL-induced pregnancy loss and IgG fraction from women with APS directly inhibits embryo
intrauterine fetal growth restriction (Salmon et al, 2003). growth. Immunopathology of obstetric APS differs from that
Complement pathway activation offers several potential tar- of thrombotic APS, especially in the case of recurrent early
gets for therapeutic complement inhibition, as shown in miscarriages, where thrombosis is neither a universal nor a
Fig 3. specific feature (Out et al, 1991). Nonetheless, LA may be
Microvascular thrombosis is one of the major pathological associated with extensive placental necrosis, infarction and
manifestations of APS and may occur alone or in combina- thrombosis in women with recurrent pregnancy loss; exten-
tion with large-vessel thrombosis. Kidneys represent the most sive villous infarction following first trimester miscarriage in
important site of microvascular thrombotic/microangiopathic a patient with SLE and positive LA and aCL was first
APS, which is associated with renal failure, thrombotic described by Nayar and Lage (1996).
microangiopathy and hypertension (Griffiths et al, 2000). In Annexin V is a cationic protein that normally serves an
patients who undergo renal transplantation for APS, the anticoagulant function by aggregating on trophoblast

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 5


Review

Thrombosis
Classical Pathway Pregnancy morbidity

(Activated by antigen/antibody Potent anaphylatoxin


Anaphylatoxins (C3a, C5a) chemotaxis, cell activation
complexes)
aPL ALXN1007
(C5a inhibitor) C5a
C1q Activated C1

C4b2a3b
C4b2a3b Membrane
attack
Lectin pathway complex
C4+C2 C3a C3 C3b C5 C5b
Activated by Mannan C5b-9
Binding lectin (MBL) C3bBb
C6 C7 C8 C9

C3bBb3b
Cell activation
and lysis
Factor B+D
C3 C3b

rEV576(coversin):
Recombinant Eculizumab: Humanized
protein inhibitor of Monoclonal antibody, binds
Alternative pathway
C5 activation to C5, prevents its cleavage
Activated by microbiological membranes, to C5a and C5b
bacterial LPS, immune complexes,
mammalian cell membranes

Fig 3. Complement pathway activation. aPL induced activation of classical pathway leading to thrombosis and pregnancy complications and
potential targets for therapeutic complement inhibition (Adapted from Arachchillage & Hillmen, 2015). The complement cascade is activated by
one of the three pathways. Activation leads to the formation of C3 convertase, resulting in the formation of C5 convertase and membrane attack
complex (MAC). aPL recognize domain I of b2GPI and stabilizes its open configuration on the cell surface. This is followed by binding of com-
plement factor C1q resulting in the activation of the complement system. The formed anaphylatoxins cause cell damage and induce a prothrom-
botic phenotype, leading to both thrombosis and pregnancy failure. Potential therapeutic targets of complement inhibition are also shown. aPL,
antiphospholipid antibodies; LPS, lipopolysaccharide.

membranes and blocking FXa and prothrombin binding. regulatory protein that blocks C3 and C4 activation) leads to
Thrombosis during the development of the normal materno- progressive embryonic loss in mice (Xu et al, 2000). It has
placental circulation may arise via interference with this been hypothesised that aPL bound to trophoblasts activate
function (Rand et al, 1997, 2010a) and women with APS complement via the classical pathway, generating split prod-
have disrupted and reduced annexin V binding to the surface ucts that mediate placental injury causing fetal loss and
of trophoblastic cells of the intervillous space compared with growth restriction. Passive transfer of IgG from women with
controls (Rand et al, 1997). During differentiation to syn- recurrent miscarriage and aPL results in a significant increase
cytium, trophoblast membrane anionic phospholipids also in the frequency of fetal resorption and reduced average
bind b2GPI and hence b2GPI-dependent aPL, which may weight of the surviving fetuses, compared to mice treated
then lead to defective placentation via complement activa- with IgG from healthy individuals (Holers et al, 2002). These
tion, inflammatory damage and placental apoptosis (Girardi, effects can be blocked by inhibition of the complement cas-
2010). cade using a C3 convertase inhibitor or by antibodies or pep-
Studies of animal and human placenta have demonstrated tides that block C5a-C5a receptor interactions (Girardi et al,
that complement activation by aPL may play a major role in 2003). Furthermore, mice deficient in C3 are resistant to the
the pathogenesis of recurrent pregnancy loss (Salmon et al, fetal injury induced by aPL (Holers et al, 2002; Girardi et al,
2003). Appropriate complement inhibition is an essential 2003) and studies in factor B-deficient mice indicate that the
requirement for normal pregnancy, as evidenced by the find- alternative complement pathway is also required. Inflamma-
ing that deficiency of Crry (a membrane-bound complement tory tissue injury is probably mediated by tumour necrosis

6 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

factor (TNF) alpha (TNFa), which increases in murine


Management of thrombosis in antiphos-
decidua after exposure to aPL (Berman et al, 2005) and anti-
pholipid syndrome
body blocking of TNFa reduces fetal resorption (Blank et al,
2003). Finally, the therapeutic effect of heparin has been Despite the implication of other mechanisms as described
shown to operate via inhibition of complement rather than above, anticoagulation rather than immunosuppression
inhibition of coagulation (Girardi et al, 2004). The PRO- remains the mainstay of management in patients with
MISE study (Predictors of pregnancy outcome: biomarkers thrombotic APS. Two randomised controlled trials have
In APS and SLE) is a prospective multicentre observational demonstrated that high-intensity warfarin is not superior to
study which will further evaluate the role of complement in moderate-intensity warfarin (International Normalized Ratio
SLE and aPL-associated pregnancy complications [INR] 20-30) for the prevention of recurrent VTE
(NCT00198068). (Crowther et al, 2003; Finazzi et al, 2005). The current rec-
Animal and human cell culture studies have demonstrated ommendation is that patients with APS and otherwise
that binding of aPL (particularly b2GPI-dependent antibod- unprecipitated VTE should remain on anticoagulants indefi-
ies) to human trophoblasts affects other critical cell func- nitely with a target INR of 25, which reduces recurrent VTE
tions; inhibition of proliferation and syncitia formation, by 80–90% compared to no treatment (Ruiz-Irastorza et al,
decreased production of human chorionic gonadotrophin 2011; Danowski et al, 2013). Indefinite anticoagulation for
and defective secretion of growth factors, as well as inducing APS patients is supported by evidence that APS patients
apoptosis (Pierangeli et al, 2008). Moreover aPL may impair carry a higher risk of recurrent thrombosis compared to
the sequential expression of cell adhesion molecules, such as aPL-negative subjects and that the thrombotic risk may
integrins and cadherins, in trophoblastic and decidual cells increase with time (Ruiz-Irastorza et al, 2002). However, in a
essential for normal placentation (Di Simone et al, 2002). systematic review, the strength of this association was uncer-
Mechanisms for aPL-mediated fetal loss and complications tain because the available evidence was of very low quality
are summarised in Table II. (Garcia et al, 2013). Thrombosis after a clear precipitant

Table II. Mechanisms for aPL-mediated fetal loss or complications

Mechanism Supporting evidence from studies in humans or animals/comments

Intraplacental thrombosis Animal studies have demonstrated that the passive transfer of aPL promotes fetal loss and placental
thrombosis and also inhibits trophoblast and decidual cell functions in vitro (Branch et al, 1990).
Placental thrombosis and infarction shown in APS patients with intra-uterine fetal death (Out et al,
1991; Nayar & Lage, 1996)
aPL binding to monocytes, endothelial cells, platelets and plasma components of the coagulation
cascade (Zhang & Mccrae, 2005) may lead to placental thrombosis.
aPL might induce a procoagulant state at the placental level by disrupting the anticoagulant annexin
A5 shield on trophoblast and endothelial cell monolayers (Rand et al, 1997)
Inflammation b2GPI-dependent aPL are able to react with human stromal decidual cells in vitro, inducing a
proinflammatory phenotype(Zhang & Mccrae, 2005)
Interference with Annexin V function Thrombosis during the development of the normal materno-placental circulation may arise via
interference with function of Annexin V (Rand et al, 1997, 2010a) and women with APS have
diminished annexin V binding to the surface of trophoblastic cells of the intervillous space
compared with controls (Rand et al, 1997).
Inhibition of syncytium-trophoblast aPL (particularly b2GPI-dependent antibodies) bind to human trophoblasts and affect several cell
differentiation and defective functions (Girardi et al, 2003), inducing cell injury and apoptosis, inhibition of proliferation and
placentation/placental apoptosis syncitia formation, decreased production of human chorionic gonadotrophin, defective secretion of
growth factors and impaired invasiveness which lead to defective placentation.
Complement activation Passive transfer of IgG from women with recurrent miscarriage and aPL results in a significant
increase in the frequency of fetal resorption and IUGR, compared to mice treated with IgG from
healthy individuals [(Holers et al, 2002). These effects can be blocked by inhibition of the
complement cascade using a C3 convertase inhibitor or by antibodies or peptides that block C5a-
C5a receptor interactions (Girardi et al, 2003).
Mice deficient in complement C3 are resistant to fetal injury induced by aPL (Holers et al, 2002;
Girardi et al, 2003).
Heparin prevents obstetric complications caused by aPL, because it blocks complement activation
rather than through its antithrombotic properties (Girardi et al, 2004).

aPL, antiphospholipid antibodies; APS, antiphosphlipid syndrome; IUGR, intrauterine growth restriction; b2GPI, b2glycoprotein-I.

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 7


Review

should not prompt testing for APS and requires only arterial thrombosis had a severe thrombotic phenotype at the
3 months of anticoagulation (Keeling et al, 2012). time of starting a DOAC and standard risk APS patients who
With regards to the optimal intensity of anticoagulation were on warfarin with target INR of 20–30 had no recur-
following arterial thrombosis, an earlier prospective cohort rent events. Only two patients [2/96 (21%)] had bleeding
study, the Antiphospholipid Antibodies and Stroke Study events.
(APASS), found no benefit of low intensity warfarin anti- Rivaroxaban has been shown to effectively reduce throm-
coagulation (target INR 14–28) over aspirin (325 mg/day) bin generation in plasma from patients with thrombotic APS
in stroke prevention (Levine et al, 2004). However, the compared to warfarin in a recently published randomised
study had important limitations, such as low target INR clinical trial (Cohen et al, 2016), but there are no data from
and aPL testing being performed only on entry to the intervention trials designed to assess clinical endpoints. Sev-
study, raising the possibility that some participants may eral other clinical trials assessing the use of DOAC is throm-
have had transient antibody positivity. Other studies botic APS are still recruiting participants (NCT02157272,
demonstrated that moderate-intensity warfarin (INR 20– NCT02295475, NCT02116036). However, given their general
30) is effective for arterial thrombosis and APS, although efficacy, DOAC could be considered as an alternative antico-
patients with arterial thrombosis were poorly represented agulant in patients with APS and VTE that are usually trea-
in these trials (only, 24% and 32% respectively) (Crowther ted with standard-intensity warfarin when there is known
et al, 2003; Finazzi et al, 2005). Ruiz-Irastorza et al (2007) VKA allergy or poor anticoagulant control. DOACs have not
recommended, following a systematic review of cohort been studied in patients with arterial thrombosis even with-
studies, that APS with arterial thrombosis and/or recurrent out APS and the comparison has been always with standard
venous events should be treated with warfarin at an INR intensity warfarin anticoagulation. Therefore, DOACs are not
>30. Although the 13th International Congress on recommended in APS patients with arterial thrombosis or
Antiphospholipid Antibodies task force also recommended those with recurrent VTE whilst achieving therapeutic antico-
an INR >30 or combined platelet inhibitor-anticoagulant agulation with warfarin.
(INR 20–30) therapy for arterial thrombosis (Ruiz-Iras- For the small proportion of patients with APS who
torza et al, 2011), this was a non-graded recommendation develop recurrent thrombosis despite apparently adequate
due to lack of consensus within the panel members. Stan- anticoagulation, an increase in warfarin intensity is recom-
dard practice currently is for a target INR of 25, with mended (i.e: target INR of 35). There are limited therapeutic
escalation to a higher target INR should thrombosis recur options for patients who have recurrent thrombosis despite
(Keeling et al, 2012). high-intensity warfarin. These include addition of an antipla-
The narrow therapeutic range and multiple interactions of telet agent or low molecular weight heparin (LMWH),
warfarin are further complicated in APS by the variable including high-intensity LMWH (maintaining peak anti Xa
responsiveness of thromboplastins to LA, leading to mislead- levels 16–20 iu/ml for once daily dosing and peak 08–10
ing INR results. However, monitoring of INR is not a major iu/ml for twice daily dosing) or combined factor Xa and fac-
issue in the majority of patients with APS and the problem tor IIa inhibitors (Arachchillage et al, 2016b). When antico-
is limited to specific thromboplastins (Tripodi et al, 2001). agulation has failed, other available therapeutic options
include combining anticoagulation with immunosuppression
and/or immunomodulation with modalities including ritux-
Direct acting oral anticoagulants
imab, hydroxychloroquine, statins rituximab complement
Direct acting oral anticoagulants (DOACs) are fixed dose inhibitors and mTOR inhibitors, such as sirolimus.
oral anticoagulants with predictable anticoagulant effect and,
consequently, no need for routine monitoring. DOACs are
Hydroxychloroquine
established as therapeutic alternatives to vitamin K antago-
nists (VKAs) for treatment and secondary prevention of VTE Hydroxychloroquine (HCQ) is a well-established treatment
in patients without APS. Although aPL are reported in for patients with SLE and rheumatoid arthritis due to its
approximately 10% of patients with VTE (Andreoli et al, anti-inflammatory effects; inhibiting cytosolic phospholipase
2013), phase III clinical trials have not compared warfarin A2 and reducing the release of the cytokines interleukin (IL)
versus DOAC for this group and experience is limited to case 6 and TNF (Sperber et al, 1993). It is currently recom-
reports. A total of 96 APS patients are reported to be treated mended as a baseline therapy in all patients with SLE who
with DOACs [82/96 (855%) rivaroxaban, 13/96 (135%) have no contraindications (Ruiz-Irastorza et al, 2010). HCQ
dabigatran and 1/96 (1%) apixaban] (Bachmeyer & Elalamy, also inhibits platelet aggregation and arachidonic acid release,
2014; Schaefer et al, 2014; Win & Rodgers, 2014; Noel et al, reducing thrombus size in mice models of APS (Espinola
2015; Sciascia et al, 2015; Son et al, 2015; Betancur et al, et al, 2002; Rand et al, 2010b) and protects the Annexin V
2016; Signorelli et al, 2016). Only 8/96 (83%) of the patients anticoagulant shield from disruption by aPL (Rand et al,
received a DOAC as acute treatment for VTE. 17 patients 2010a). Its antithrombotic activity is demonstrated by its effi-
had recurrent thrombosis. However, those who had recurrent cacy as thromboprophylaxis after hip surgery (Johansson

8 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

et al, 1981) and in patients with SLE, without increased Complement inhibition
bleeding (Schmidt-Tanguy et al, 2013). Recent APS consensus
The experimental evidence discussed above has led to specu-
guidelines support its use as an adjuvant to anticoagulation
lation that inhibition of complement may be a useful thera-
in patients with recurrent thrombosis despite anticoagulation
peutic strategy in APS, although only preliminary and
(Ruiz-Irastorza et al, 2011). A phase III multicentre trial
anecdotal data are available to date. A phase II clinical trial
exploring the effect of HCQ as primary thrombosis prophy-
evaluating the use of eculizumab to enable renal transplanta-
laxis in individuals with persistently positive antiphospholipid
tion and prevent post-transplant thrombotic microangiopa-
antibodies (NCT01784523) has been now completed and
thy (TMA) in patients with a history of CAPS
findings are awaited.
(NCT01029587) is ongoing, but not recruiting participants.
Preliminary data of this study from 80 patients showed that
Statins eculizumab is effective in reducing the incidence of acute
antibody-mediated rejection in sensitized deceased donor
Statins inhibit the enzyme 3-hydroxy-3-methyl-glutaryl-
kidney transplant recipients (KTR). Patient and graft survival
coenzyme A (HMG-CoA) reductase (HMGCR), which has a
and kidney function at 1 year were similar to those expected
central role in hepatic cholesterol production, but also have
for non-sensitized KTR (Glotz et al, 2015). Another phase II
pleiotropic effects including anti-inflammatory and
multicentre clinical trial evaluating the safety and tolerability
antithrombotic actions on EC and monocytes (Ferrara et al,
of intravenous ALXN1007 (C5a inhibitor) in persistently
2003). Furthermore, there is evidence they increase fibrinol-
aPL-positive patients with at least one of the non-criteria
ysis and decrease tissue factor mRNA expression; opposite
manifestations of APS is underway NCT02128269.
effects to aPL (Ferrara et al, 2004). These observations and
the demonstrated VTE reduction in patients receiving sta-
tins (Ridker, 2009) suggest a possible benefit from their use Sirolimus
in APS. In a prospective open-label pilot study of fluvas-
Sirolimus is an mTOR inhibitor that is currently used to pre-
tatin in aPL positive patients, Erkan et al (2014a) demon-
vent reactive arterial stenosis after coronary stenting. A
strated that fluvastatin can reduce pro-inflammatory and
cohort of patients with APS nephropathy who required
pro-thrombotic biomarkers, such as IL6, IL1b, vascular
transplantation and were receiving sirolimus had no recur-
endothelial growth factor, TNFa and interferon-a. Based on
rence of vascular lesions and had decreased vascular prolifer-
available evidence, the 14th International Congress on
ation on biopsy as compared to APS patients not receiving
Antiphospholipid Antibodies Task Force Report on APS
sirolimus (Canaud et al, 2014). Graft survival at 144 months
Treatment Trends stated that statins cannot be recom-
was 7/10 (70%) versus 3/27 (11%) in the two groups. Fur-
mended in APS patients in the absence of hyperlipidaemia,
ther larger studies are required to establish the routine use of
but may be a useful treatment adjunct in APS patients with
mTOR inhibitors in APS patients.
recurrent thrombosis despite adequate anticoagulation
(Erkan et al, 2014b).
Potential future therapeutic options
Rituximab TIFI is a 20-amino acid peptide from human cytomegalo-
virus, which shares similarities with the PL-binding site in
Rituximab appears to be an effective treatment for thrombo-
the b2GPI molecule, DV. In vitro evidence suggests that TIFI
cytopenia and haemolytic anaemia associated with aPL (Erre
inhibits the binding of labelled b2GPI to human ECs and
et al, 2008) and may improve some other manifestations,
mouse monocytes (Ostertag et al, 2006). In a mouse model
such as skin ulcers (Erkan et al, 2013). Rituximab has been
of APS, infusion of TIFI reduced the binding of fluores-
used for the treatment of difficult cases, such as refractory
ceinated b2GPI to ECs and reduced thrombosis size after aPL
thrombocytopenia or other non-criteria aPL manifestations,
infusion (de la Torre et al, 2012). A dimeric peptide of b2-
CAPS, and recurrent VTE, in over 50 case reports. In a
glycoprotein-DI (DI dimer) has a similar effect (Agostinis
review by Kumar and Roubey (2010), it was reported that
et al, 2014). These peptides are not yet in clinical trials but
rituximab had a beneficial clinical effect in 19 of 21 pub-
may offer a new paradigm for APS treatment in the future.
lished cases and aPL levels were significantly decreased in ten
of 12 cases. Review of the CAPS registry of 20 patients trea-
ted with rituximab reported a recovery rate of 75% of acute Management of CAPS
episodes in combination with multiple treatment strategies,
Anticoagulation, intravenous immunoglobulin (IVIG),
including anticoagulation, steroids, plasma exchange and
plasma exchange, immunosuppressive therapy, prostacyclin,
cyclophosphamide (Berman et al, 2013); consequently, the
fibrinolytics and defibrotide have all been used in the man-
responses cannot be clearly attributed to rituximab alone.
agement of CAPS. Case reports described above suggest ecu-
Furthermore, there was no consistency in the reduction in
lizumab may be effective in CAPS unresponsive to other
aPL level in these patients.

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 9


Review

treatments (Shapira et al, 2012) and in preventing relapse independent of its anticoagulant activity. Heparin may also
after kidney transplantation. There are reported cases of act by inhibiting aPL binding to trophoblastic cell mem-
rituximab use in patients with CAPS with variable responses branes, modulating trophoblast apoptosis, promoting tro-
(Erre et al, 2008). Espinosa et al (2011) reviewed 9 such phoblast cell invasiveness, and reducing complement
patients, two of who died; but notably, in three of the sur- activation with the ensuing inflammatory response at the
viving patients’ tests for aPL became negative after rituximab decidual-placental interface (Franklin & Kutteh, 2003).
treatment. Women with obstetric APS but no prior history of VTE,
post-partum thromboprophylaxis should be considered/of-
fered for 6 weeks following delivery based on risk assessment.
Management of asymptomatic carriers of aPL
However, there is variation in guidelines as well as clinical
Another unanswered question is whether primary thrombo- practice regarding this. The Nimes Obstetricians and Haema-
prophylaxis is indicated in subjects with aPL and how risk of tologists Antiphospholipid Syndrome (NOH-APS) observa-
a first thrombosis can be stratified in order to personalise tional study reported that the annual rates of DVT (146%;
treatment. A prospective evaluation of the incidence and risk range 115–182%), pulmonary embolism (043%; range
factors for a first vascular event in 258 asymptomatic indi- 026–066%), superficial vein thrombosis (044%; range 028–
viduals with aPL determined that the annual incidence rate 068%), and cerebrovascular events (032%; range 018–
was 186% compared to 01% in the general population with 053%) were significantly higher in women with pure obstet-
a median follow-up of 35 months (range 1-48) (Ruffatti ric APS compared with aPL-negative women with obstetric
et al, 2011). Hypertension and the presence of LA were iden- morbidity, despite LDA primary prophylaxis (Gris et al,
tified as independent risk factors for development of throm- 2012). British Committee for Standards in Haematology
bosis (Ruffatti et al, 2011). In a study of 104 subjects with (Keeling et al, 2012) and Royal College of Obstetricians and
triple positivity, there were 25 first thrombotic events (53% Gynaecologists guidelines are in keeping with this observa-
per year) with a cumulative incidence of 371% (95% CI: tion. The RCOG Green-top Guideline (https://www.rcog.org.
199%-543%) after 10 years. Aspirin did not significantly uk/globalassets/documents/guidelines/gtg-37a.pdf), states
reduce the incidence of thromboembolic events (Pengo et al, ‘Persistent antiphospholipid antibodies (LA and/or anticardi-
2011). The Antiphospholipid Antibody Acetylsalicyclic Acid olipin and/or anti-b2-GPI 1 antibodies) in women without
(APLASA) study randomised individuals with asymptomatic, previous VTE should be considered as a risk factor for
persistently positive aPL to receive 81 mg of aspirin or pla- thrombosis such that if she has other risk factors she may be
cebo daily (Erkan et al, 2007). There was no significant dif- considered for antenatal or postnatal thromboprophylaxis’
ference in outcome between the two arms. On current (either 10 day or 6 weeks based on risk assessment). If VTE
evidence, thromboprophylaxis with aspirin and/or VKA is develops during pregnancy, treatment with therapeutic doses
not recommended for asymptomatic, aPL positive individu- of LMWH should be employed during the remainder of the
als. Whether subjects who are triple aPL-positive should be pregnancy and for at least 6 weeks postnatally and until at
treated differently remains to be established. least 3 months of treatment has been given in total (https://
www.rcog.org.uk/globalassets/documents/guidelines/gtg-37b.
pdf). However, women with thrombosis during pregnancy
Treatment of obstetric APS
and positive aPL should be reassessed after pregnancy in case
Heparin and low dose aspirin (LDA) is the treatment of long-term anticoagulation is indicated.
choice for recurrent pregnancy loss associated with APS. This With standard treatment, approximately 70% of pregnant
is based on several important studies (Kutteh, 1996; Rai et al, women with APS have successful pregnancy outcome (Bram-
1997). In a meta-analysis of data from five trials that ran- ham et al, 2010). There are limited therapeutic options and
domly assigned patients to either heparin and aspirin or no guidelines for those who do not respond to heparin and
aspirin alone for the management of APS-related pregnancies LDA. Addition of low-dose prednisolone to standard treat-
(Mak et al, 2010), the overall live birth rates in 334 patients ment in the first-trimester improved the live birth rate in
were 7427% and 5585% respectively (relative risk 1301; refractory aPL-related first trimester pregnancy loss (Bram-
95% CI 1040, 1629). The American College of Chest Physi- ham et al, 2011): nearly two-thirds of pregnancies (61%)
cians (ACCP) guidelines (Bates et al, 2012) recommend resulted in live births, of which 8 (57%) were uncomplicated
treating women with obstetric APS who meet the revised term pregnancies. However, the frequency of some complica-
Sapporo criteria with heparin and LDA in the antepartum tions remained elevated, mainly preterm delivery (21%).
period as soon as pregnancy is confirmed. LMWH is gener- Data on the usefulness of IVIG in obstetric APS are conflict-
ally favoured due to a lower incidence of heparin-induced ing (Parke et al, 1989; Branch et al, 2000).
thrombocytopenia and low risk of osteopenia compared to In a mouse model of obstetric APS HCQ prevented fetal
unfractionated heparin. The key observation made by Girardi death and the placental metabolic changes as measured by
et al (2004), that heparin can inhibit complement-mediated proton magnetic resonance spectroscopy (Bertolaccini et al,
apoptosis, provides a possible explanation for its efficacy 2016). The same study showed that HCQ prevented

10 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

complement activation in vivo and in vitro. Complement C5a complications of pregnancy. Warfarin, heparin and/or anti-
levels in serum samples from APS patients and APS-mice platelet drugs are still the standard of care for APS-thrombo-
were lower after treatment with HCQ while the antibodies sis and although attractive, DOAC can, at present, only be
titres remained unchanged. HCQ prevented not only placen- considered as an alternative in patients with APS and VTE
tal insufficiency but also abnormal fetal brain development that are usually treated with standard-intensity warfarin
in APS (Bertolaccini et al, 2016). A European multicentre when there is known VKA allergy or poor anticoagulant con-
retrospective study of 30 patients with APS and 35 pregnan- trol. The realisation that additional pathways are involved in
cies showed a better outcome of pregnancies that were trea- the pathogenesis of APS has significant implications for treat-
ted by the addition of HCQ when compared with previous ment. Agents acting outside the coagulation system, such as
pregnancies under the conventional treatment (Mekinian HCQ for pregnancy complications and sirolimus as an inhi-
et al, 2015). Sciascia et al (2016) reported HCQ-treatment bitor of the mTOR pathway are now under evaluation and
was associated with a higher rate of live births (67% vs. 57%; represent a potential radical change in thinking for haematol-
P = 005) and a lower prevalence of aPL–related pregnancy ogists.
morbidity (47% vs. 63%; P = 0004).

Author contribution
Conclusion
DRJ Arachchillage performed the literature search and wrote
Despite the clear association between aPL and the throm- the first draft. M Laffan critically reviewed the manuscript
botic and obstetric manifestations of APS, the exact mecha- and both authors approved the final manuscript.
nisms by which aPL cause these problems is yet to be fully
established. However, the activation of complement and
Conflict of interest
endothelial cells by DI-specific anti-b2GPI antibodies has
emerged as an important component. Incorporation of DI- The authors state that they have no relevant conflict of inter-
specific anti-b2GPI antibodies into diagnostic panels may est.
allow better identification of those at high risk for clinical
events. Related but distinct pathways also link aPL to the

References Estimated frequency of antiphospholipid anti- autoantibodies from patients with antiphospho-
bodies in patients with pregnancy morbidity, lipid syndrome are sufficient to potentiate arte-
Abreu, M.M., Danowski, A., Wahl, D.G., Amigo, stroke, myocardial infarction, and deep vein rial thrombus formation in a mouse model.
M.C., Tektonidou, M., Pacheco, M.S., Fleming, thrombosis: a critical review of the literature. Blood, 117, 3453–3459.
N., Domingues, V., Sciascia, S., Lyra, J.O., Petri, Arthritis Care & Research, 65, 1869–1873. Artenjak, A., Locatelli, I., Brelih, H., Simonic,
M., Khamashta, M. & Levy, R.A. (2015) The rel- Arachchillage, D.R.J. & Hillmen, P. (2015) Parox- D.M., Ulcova-Gallova, Z., Swadzba, J., Musial,
evance of “non-criteria” clinical manifestations ysmal nocturnal haemoglobinuria in pregnancy. J., Iwaniec, T., Stojanovich, L., Conti, F., Vale-
of antiphospholipid syndrome:14th International In: Disorders of Thrombosis and Haemostasis in sini, G., Avcin, T., Cohen Tervaert, J.W., Shoen-
Congress on Antiphospholipid Antibodies Tech- Pregnancy (ed. by Cohen, H & O’Brien, P). pp. feld, Y., Blank, M., Ambrozic, A., Sodin-Semrl,
nical Task Force Report on Antiphospholipid 327–342. 2nd edn. Springer, London. S., Bozic, B. & Cucnik, S. (2015) Immunoreac-
Syndrome Clinical Features. Autoimmunity Arachchillage, D.R., Efthymiou, M., Mackie, I.J., tivity and avidity of IgG anti-beta2-glycoprotein
Reviews, 14, 401–414. Lawrie, A.S., Machin, S.J. & Cohen, H. (2014) I antibodies from patients with autoimmune
Adams, M.J., Donohoe, S., Mackie, I.J. & Machin, Anti-protein C antibodies are associated with diseases to different peptide clusters of beta2-
S.J. (2001) Anti-tissue factor pathway inhibitor resistance to endogenous protein C activation glycoprotein I. Immunologic Research, 61, 35–44.
activity in patients with primary antiphospho- and a severe thrombotic phenotype in antiphos- Bachmeyer, C. & Elalamy, I. (2014) Rivaroxaban
lipid syndrome. British Journal of Haematology, pholipid syndrome. Journal of Thrombosis and as an effective treatment for recurrent superficial
114, 375–379. Haemostasis, 12, 1801–1809. thrombophlebitis related to primary antiphos-
Agar, C., van Os, G.M., Morgelin, M., Sprenger, Arachchillage, D.R., Mackie, I.J., Efthymiou, M., pholipid syndrome. Clinical and Experimental
R.R., Marquart, J.A., Urbanus, R.T., Derksen, Chitolie, A., Hunt, B.J., Isenberg, D.A., Dermatology, 39, 840–841.
R.H., Meijers, J.C. & de Groot, P.G (2010) Khamashta, M., Machin, S.J. & Cohen, H. Banzato, A., Pozzi, N., Frasson, R., De Filippis, V.,
Beta2-glycoprotein I can exist in 2 conforma- (2016a) Rivaroxaban limits complement activa- Ruffatti, A., Bison, E., Padayattil, S.J., Denas, G.
tions: implications for our understanding of the tion compared with warfarin in antiphospholipid & Pengo, V. (2011) Antibodies to Domain I of
antiphospholipid syndrome. Blood, 116, 1336– syndrome patients with venous thromboem- beta(2)Glycoprotein I are in close relation to
1343. bolism. Journal of Thrombosis and Haemostasis, patients risk categories in Antiphospholipid Syn-
Agostinis, C., Durigutto, P., Sblattero, D., Borghi, 14, 2177–2186. drome (APS). Thrombosis Research, 128, 583–
M.O., Grossi, C., Guida, F., Bulla, R., Macor, P., Arachchillage, D.R., Besser, M., Maclean, R., 586.
Pregnolato, F., Meroni, P.L. & Tedesco, F (2014) Baglin, T. & van Veen, J.J. (2016b) Combined Bas de Laat, H., Derksen, R.H. & de Groot, P.G.
A non-complement-fixing antibody to beta2 gly- factor IIa and Xa inhibitor therapy for thrombo- (2004) beta2-glycoprotein I, the playmaker of
coprotein I as a novel therapy for antiphospho- sis whilst on therapeutic anticoagulant. Throm- the antiphospholipid syndrome. Clinical
lipid syndrome. Blood, 123, 3478–3487. bosis Research, 143, 137–140. Immunology, 112, 161–168.
Andreoli, L., Chighizola, C.B., Banzato, A., Pons- Arad, A., Proulle, V., Furie, R.A., Furie, B.C. & Bates, S.M., Greer, I.A., Middeldorp, S., Veenstra,
Estel, G.J., Ramire de, J.G. & Erkan, D. (2013) Furie, B. (2011) beta(2)-Glycoprotein-1 D.L., Prabulos, A.M. & Vandvik, P.O. (2012)

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 11


Review

VTE, thrombophilia, antithrombotic therapy, syndrome during pregnancy. The Pregnancy asymptomatic antiphospholipid antibody-posi-
and pregnancy: Antithrombotic Therapy and Loss Study Group. American Journal of Obstet- tive individuals. Arthritis and Rheumatism, 56,
Prevention of Thrombosis, 9th ed: American rics and Gynecology, 182, 122–127. 2382–2391.
College of Chest Physicians Evidence-Based Breen, K.A., Seed, P., Parmar, K., Moore, G.W., Erkan, D., Vega, J., Ramon, G., Kozora, E. & Lock-
Clinical Practice Guidelines. Chest, 141, e691S– Stuart-Smith, S.E. & Hunt, B.J. (2012) Comple- shin, M.D. (2013) A pilot open-label phase II
e736S. ment activation in patients with isolated trial of rituximab for non-criteria manifestations
Berman, J., Girardi, G. & Salmon, J.E. (2005) antiphospholipid antibodies or primary of antiphospholipid syndrome. Arthritis and
TNF-alpha is a critical effector and a target for antiphospholipid syndrome. Thrombosis and Rheumatism, 65, 464–471.
therapy in antiphospholipid antibody-induced Haemostasis, 107, 423–429. Erkan, D., Willis, R., Murthy, V.L., Basra, G.,
pregnancy loss. Journal of Immunology, 174, Canaud, G., Bienaime, F., Tabarin, F., Bataillon, Vega, J., Ruiz-Limon, P., Carrera, A.L., Papa-
485–490. G., Seilhean, D., Noel, L.H., Dragon-Durey, lardo, E., Martinez-Martinez, L.A., Gonzalez,
Berman, H., Rodriguez-Pinto, I., Cervera, R., M.A., Snanoudj, R., Friedlander, G., Halbwachs- E.B. & Pierangeli, S.S. (2014a) A prospective
Morel, N., Costedoat-Chalumeau, N., Erkan, D., Mecarelli, L., Legendre, C. & Terzi, F. (2014) open-label pilot study of fluvastatin on proin-
Shoenfeld, Y. & Espinosa, G. (2013) Rituximab Inhibition of the mTORC pathway in the flammatory and prothrombotic biomarkers in
use in the catastrophic antiphospholipid syn- antiphospholipid syndrome. The New England antiphospholipid antibody positive patients.
drome: descriptive analysis of the CAPS registry Journal of Medicine, 371, 303–312. Annals of the Rheumatic Diseases, 73, 1176–1180.
patients receiving rituximab. Autoimmunity Cervera, R., Rodriguez-Pinto, I., Colafrancesco, S., Erkan, D., Aguiar, C.L., Andrade, D., Cohen, H.,
Reviews, 12, 1085–1090. Conti, F., Valesini, G., Rosario, C., Agmon- Cuadrado, M.J., Danowski, A., Levy, R.A., Ortel,
Bertolaccini, M.L., Contento, G., Lennen, R., Levin, N., Shoenfeld, Y., Ferrao, C., Faria, R., T.L., Rahman, A., Salmon, J.E., Tektonidou,
Sanna, G., Blower, P.J., Ma, M.T., Sunassee, K. Vasconcelos, C., Signorelli, F. & Espinosa, G. M.G., Willis, R. & Lockshin, M.D. (2014b) 14th
& Girardi, G. (2016) Complement inhibition by (2014) 14th International Congress on International Congress on Antiphospholipid
hydroxychloroquine prevents placental and fetal Antiphospholipid Antibodies Task Force Report Antibodies: task force report on antiphospho-
brain abnormalities in antiphospholipid syn- on Catastrophic Antiphospholipid Syndrome. lipid syndrome treatment trends. Autoimmunity
drome. Journal of Autoimmunity, 75, 30–38. Autoimmunity Reviews, 13, 699–707. Reviews, 13, 685–696.
Betancur, J.F., Bonilla-Abadia, F., Hormaza, A.A., Cohen, H., Hunt, B.J., Efthymiou, M., Arachchil- Erre, G.L., Pardini, S., Faedda, R. & Passiu, G.
Jaramillo, F.J., Canas, C.A. & Tobon, G.J. (2016) lage, D.R., Mackie, I.J., Clawson, S., Sylvestre, (2008) Effect of rituximab on clinical and labo-
Direct oral anticoagulants in antiphospholipid Y., Machin, S.J., Bertolaccini, M.L., Ruiz-Castel- ratory features of antiphospholipid syndrome: a
syndrome: a real life case series. Lupus, 25, 658– lano, M., Muirhead, N., Dore, C.J., Khamashta, case report and a review of literature. Lupus, 17,
662. M. & Isenberg, D.A. (2016) Rivaroxaban versus 50–55.
Blank, M., Krause, I., Wildbaum, G., Karin, N. & warfarin to treat patients with thrombotic Espinola, R.G., Pierangeli, S.S., Gharavi, A.E. &
Shoenfeld, Y. (2003) TNFalpha DNA vaccina- antiphospholipid syndrome, with or without Harris, E.N. (2002) Hydroxychloroquine
tion prevents clinical manifestations of experi- systemic lupus erythematosus (RAPS): a ran- reverses platelet activation induced by human
mental antiphospholipid syndrome. Lupus, 12, domised, controlled, open-label, phase 2/3, non- IgG antiphospholipid antibodies. Thrombosis
546–549. inferiority trial. Lancet Haematology, 3, e426– and Haemostasis, 87, 518–522.
Boles, J. & Mackman, N. (2010) Role of tissue fac- e436. Espinosa, G., Berman, H. & Cervera, R. (2011)
tor in thrombosis in antiphospholipid antibody Conley, C. & Hartmann, R.C. (1952) A hemor- Management of refractory cases of catastrophic
syndrome. Lupus, 19, 370–378. rhagic disorder caused by circulating anticoagu- antiphospholipid syndrome. Autoimmunity
Bowie, E.J., Thompson, J.H. Jr, Pascuzzi, C.A. & lant in patients with disseminated lupus Reviews, 10, 664–668.
Owen, C.A. Jr (1963) Thrombosis in systemic erythematosus. Journal of Clinical Investigation, Ferrara, D.E., Liu, X., Espinola, R.G., Meroni, P.L.,
lupus erythematosus despite circulating antico- 31, 2. Abukhalaf, I., Harris, E.N. & Pierangeli, S.S.
agulants. Journal of Laboratory and Clinical Med- Crowther, M.A., Ginsberg, J.S., Julian, J., Denburg, (2003) Inhibition of the thrombogenic and
icine, 62, 416–430. J., Hirsh, J., Douketis, J., Laskin, C., Fortin, P., inflammatory properties of antiphospholipid
Bramham, K., Hunt, B.J., Germain, S., Calatayud, Anderson, D., Kearon, C., Clarke, A., Geerts, antibodies by fluvastatin in an in vivo animal
I., Khamashta, M., Bewley, S. & Nelson-Piercy, W., Forgie, M., Green, D., Costantini, L., model. Arthritis and Rheumatism, 48, 3272–3279.
C. (2010) Pregnancy outcome in different clini- Yacura, W., Wilson, S., Gent, M. & Kovacs, M.J. Ferrara, D.E., Swerlick, R., Casper, K., Meroni,
cal phenotypes of antiphospholipid syndrome. (2003) A comparison of two intensities of war- P.L., Vega-Ostertag, M.E., Harris, E.N. & Pier-
Lupus, 19, 58–64. farin for the prevention of recurrent thrombosis angeli, S.S. (2004) Fluvastatin inhibits up-regula-
Bramham, K., Thomas, M., Nelson-Piercy, C., in patients with the antiphospholipid antibody tion of tissue factor expression by
Khamashta, M. & Hunt, B.J. (2011) First-trime- syndrome. The New England Journal of Medicine, antiphospholipid antibodies on endothelial cells.
ster low-dose prednisolone in refractory 349, 1133–1138. Journal of Thrombosis and Haemostasis, 2, 1558–
antiphospholipid antibody-related pregnancy Danowski, A., Rego, J., Kakehasi, A.M., Funke, A., 1563.
loss. Blood, 117, 6948–6951. Carvalho, J.F., Lima, I.V., Souza, A.W. & Levy, Finazzi, G., Marchioli, R., Brancaccio, V., Schinco,
Branch, D.W., Dudley, D.J., Mitchell, M.D., R.A. (2013) Guidelines for the treatment of P., Wisloff, F., Musial, J., Baudo, F., Berrettini,
Creighton, K.A., Abbott, T.M., Hammond, E.H. antiphospholipid syndrome. Revista Brasileira de M., Testa, S., D’Angelo, A., Tognoni, G. & Bar-
& Daynes, R.A. (1990) Immunoglobulin G frac- Reumatologia, 53, 184–192. bui, T. (2005) A randomized clinical trial of
tions from patients with antiphospholipid anti- Di Simone, N., Castellani, R., Caliandro, D. & Car- high-intensity warfarin vs. conventional
bodies cause fetal death in BALB/c mice: a uso, A. (2002) Antiphospholid antibodies regu- antithrombotic therapy for the prevention of
model for autoimmune fetal loss. American late the expression of trophoblast cell adhesion recurrent thrombosis in patients with the
Journal of Obstetrics and Gynecology, 163, 210– molecules. Fertility and Sterility, 77, 805–811. antiphospholipid syndrome (WAPS). Journal of
216. Erkan, D., Harrison, M.J., Levy, R., Peterson, M., Thrombosis and Haemostasis, 3, 848–853.
Branch, D.W., Peaceman, A.M., Druzin, M., Silver, Petri, M., Sammaritano, L., Unalp-Arida, A., Franklin, R.D. & Kutteh, W.H. (2003) Effects of
R.K., El-Sayed, Y., Silver, R.M., Esplin, M.S., Vilela, V., Yazici, Y. & Lockshin, M.D. (2007) unfractionated and low molecular weight hep-
Spinnato, J. & Harger, J. (2000) A multicenter, Aspirin for primary thrombosis prevention in arin on antiphospholipid antibody binding
placebo-controlled pilot study of intravenous the antiphospholipid syndrome: a randomized, in vitro. Obstetrics and Gynecology, 101, 455–
immune globulin treatment of antiphospholipid double-blind, placebo-controlled trial in 462.

12 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

Galli, M., Comfurius, P., Maassen, C., Hemker, J.E. (2002) Complement C3 activation is B., Kveder, T., de Moerloose, P., Boehlen, F.,
H.C., de Baets, M.H., van Breda-Vriesman, P.J., required for antiphospholipid antibody-induced Rand, J., Ulcova-Gallova, Z., Mertens, K. & de
Barbui, T., Zwaal, R.F. & Bevers, E.M. (1990) fetal loss. Journal of Experimental Medicine, 195, Groot, P.G. (2009) The association between cir-
Anticardiolipin antibodies (ACA) directed not 211–220. culating antibodies against domain I of beta2-
to cardiolipin but to a plasma protein cofactor. Ioannou, Y., Pericleous, C., Giles, I., Latchman, glycoprotein I and thrombosis: an international
Lancet, 335, 1544–1547. D.S., Isenberg, D.A. & Rahman, A. (2007) Bind- multicenter study. Journal of Thrombosis and
Galli, M., Luciani, D., Bertolini, G. & Barbui, T. ing of antiphospholipid antibodies to discontin- Haemostasis, 7, 1767–1773.
(2003) Lupus anticoagulants are stronger risk uous epitopes on domain I of human beta(2)- Lackner, K.J. & M€ uller-Calleja, N. (2016) Patho-
factors for thrombosis than anticardiolipin anti- glycoprotein I: mutation studies including resi- genesis of the antiphospholipid syndrome revis-
bodies in the antiphospholipid syndrome: a sys- dues R39 to R43. Arthritis and Rheumatism, 56, ited: time to challenge the dogma. Journal of
tematic review of the literature. Blood, 101, 280–290. Thrombosis and Haemostasis, 6, 1117–1120.
1827–1832. Ioannou, Y., Zhang, J.Y., Passam, F.H., Rahgozar, Lambrianides, A., Carroll, C.J., Pierangeli, S.S.,
Garcia, D., Akl, E.A., Carr, R. & Kearon, C. (2013) S., Qi, J.C., GiannakopoulosB, Qi M., Yu, P., Yu, Pericleous, C., Branch, W., Rice, J., Latchman,
Antiphospholipid antibodies and the risk of D.M., Hogg, P.J. & Krilis, S.A. (2010) Naturally D.S., Townsend, P., Isenberg, D.A., Rahman, A.
recurrence after a first episode of venous throm- occurring free thiols within b2-glycoprotein I & Giles, I.P. (2010) Effects of polyclonal IgG
boembolism: a systematic review. Blood, 122, in vivo: nitrosylation, redox modification by derived from patients with different clinical
817–824. endothelial cells and regulation of oxidative stress types of the antiphospholipid syndrome on
Girardi, G. (2010) Role of tissue factor in the induced cell injury. Blood, 116, 1961–1970. monocyte signaling pathways. Journal of
maternal immunological attack of the embryo in Ioannou, Y., Zhang, J.Y., Qi, M., Gao, L., Qi, J.C., Immunology, 184, 6622–6628.
the antiphospholipid syndrome. Clinical Reviews Yu, D.M., Lau, H., Sturgess, A.D., Vla- Levine, S.R., Brey, R.L., Tilley, B.C., Thompson,
in Allergy and Immunology, 39, 160–165. choyiannopoulos, P.G., Moutsopoulos, H.M., J.L., Sacco, R.L., Sciacca, R.R., Murphy, A., Lu,
Girardi, G., Berman, J., Redecha, P., Spruce, L., Rahman, A., Pericleous, C., Atsumi, T., Koike, Y., Costigan, T.M., Rhine, C., Levin, B., Triplett,
Thurman, J.M., Kraus, D., Hollmann, T.J., T., Heritier, S., Giannakopoulos, B. & Krilis, D.A. & Mohr, J.P. (2004) Antiphospholipid
Casali, P., Caroll, M.C., Wetsel, R.A., Lambris, S.A. (2011) Novel assays of thrombogenic antibodies and subsequent thrombo-occlusive
J.D., Holers, V.M. & Salmon, J.E. (2003) Com- pathogenicity in the antiphospholipid syndrome events in patients with ischemic stroke. JAMA,
plement C5a receptors and neutrophils mediate based on the detection of molecular oxidative 291, 576–584.
fetal injury in the antiphospholipid syndrome. modification of the major autoantigen beta2- Liestol, S., Sandset, P.M., Jacobsen, E.M., Mow-
Journal of Clinical Investigation, 112, 1644–1654. glycoprotein I. Arthritis and Rheumatism, 63, inckel, M.C. & Wisloff, F. (2007) Decreased
Girardi, G., Redecha, P. & Salmon, J.E. (2004) 2774–2782. anticoagulant response to tissue factor pathway
Heparin prevents antiphospholipid antibody- Johansson, E., Forsberg, K. & Johnsson, H. (1981) inhibitor type 1 in plasmas from patients with
induced fetal loss by inhibiting complement Clinical and experimental evaluation ofthe lupus anticoagulants. British Journal of Haema-
activation. Nature Medicine, 10, 1222–1226. thromboprophylactic effect of hydroxychloro- tology, 136, 131–137.
Glotz, D., Russ, G., Rostaing, L., Legendre, C., quine sulfate after total hip replacement. Hae- Maiti, S.N., Balasubramanian, K., Ramoth, J.A. &
Chadban, S., Grinyo, J., Mamode, N., Cozzi, E., mostasis, 10, 89–96. Schroit, A.J. (2008) b-2-glycoprotein 1-depen-
Lebranchu, Y., Sandrini, S., Couzi, L. & Marks, Keeling, D., Mackie, I., Moore, G.W., Greer, I.A. & dent macrophage uptake of apoptotic cells:bind-
W. (2015) Eculizumab in Prevention of Acute Greaves, M. (2012) Guidelines on the investiga- ing to lipoprotein receptor-related protein
Antibody-Mediated Rejection in Sensitized tion and management of antiphospholipid receptor family members. Journal of Biological
Deceased-Donor Kidney Transplant Recipients: syndrome. British Journal of Haematology, 157, Chemistry, 283, 3761–3766.
1-Year Outcomes. American Transplant Con- 47–58. Mak, A., Cheung, M.W., Cheak, A.A. & Ho, R.C.
gress, Abstract number: 3039. Kumar, D. & Roubey, R.A. (2010) Use of ritux- (2010) Combination of heparin and aspirin is
Griffiths, M.H., Papadaki, L. & Neild, G.H. (2000) imab in the antiphospholipid syndrome. Current superior to aspirin alone in enhancing live births
The renal pathology of primary antiphospho- Rheumatology Reports, 12, 40–44. in patients with recurrent pregnancy loss and
lipid syndrome: a distinctive form ofendothelial Kutteh, W.H. (1996) Antiphospholipid antibody- positive anti-phospholipid antibodies: a meta-
injury. QJM : Monthly Journal of the Association associated recurrent pregnancy loss: treatment analysis of randomized controlled trials and
of Physicians, 93, 457–467. with heparin and low-dose aspirin is superior to meta-regression. Rheumatology (Oxford), 49,
Gris, J.C., Bouvier, S., Molinari, N., Galanaud, J.P., low-dose aspirin alone. American Journal of 281–288.
Cochery-Nouvellon, E., Mercier, E., Fabbro- Obstetrics and Gynecology, 174, 1584–1589. Malia, R.G., Kitchen, S., Greaves, M. & Preston,
Peray, P., Balducchi, J.P., Mares, P., Quere, I. & de Laat, H.B., Derksen, R.H., Urbanus, R.T., Roest, F.E. (1990) Inhibition of activated protein C
Dauzat, M. (2012) Comparative incidence of a M. & de Groot, P.G (2004) beta2-glycoprotein and its cofactor protein S by antiphospholipid
first thrombotic event in purely obstetric I-dependent lupus anticoagulant highly corre- antibodies. British Journal of Haematology, 76,
antiphospholipid syndrome with pregnancy loss: lates with thrombosis in the antiphospholipid 101–107.
the NOH-APS observational study. Blood, 119, syndrome. Blood, 104, 3598–3602. Manning, B.D. & Cantley, L.C. (2007) AKT/PKB
2624–2632. de Laat, B., Derksen, R.H., Urbanus, R.T. & de signaling: navigating downstream. Cell, 129,
de Groot, P.G. & Meijers, J.C. (2011) beta(2) -Gly- Groot, P.G. (2005) IgG antibodies that recognize 1261–1274.
coprotein I: evolution, structure and function. epitope Gly40-Arg43 in domain I of beta 2-gly- Manukyan, D., M€ uller-Calleja, N., J€ackel, S., Luch-
Journal of Thrombosis and Haemostasis, 9, 1275– coprotein I cause LAC, and their presence corre- mann, K., M€ onnikes, R., Kiouptsi, K., Rein-
1284. lates strongly with thrombosis. Blood, 105, hardt, C, Jurk, K., Walter, U. & Lackner, K.J.
de Groot, P.G., Lutters, B., Derksen, R.H., Lisman, 1540–1545. (2016) Cofactor-independent human antiphos-
T., Meijers, J.C. & Rosendaal, F.R. (2005) Lupus de Laat, B., Derksen, R.H., van Lummel, M., Pen- pholipid antibodies induce venous thrombosis
anticoagulants and the risk of a first episode of nings, M.T. & de Groot, P.G. (2006) Pathogenic in mice. Journal of Thrombosis and Haemostasis,
deep venous thrombosis. Journal of Thrombosis anti-beta2-glycoprotein I antibodies recognize 14, 1011–1020.
and Haemostasis, 3, 1993–1997. domain I of beta2-glycoprotein I only after a Mcneil, H.P., Simpson, R.J., Chesterman, C.N. &
Holers, V.M., Girardi, G., Mo, L., Guthridge, J.M., conformational change. Blood, 107, 1916–1924. Krilis, S.A. (1990) Anti-phospholipid antibodies
Molina, H., Pierangeli, S.S., Espinola, R., Xiao- de Laat, B., Pengo, V., Pabinger, I., Musial, J., Vos- are directed against a complex antigen that
wei, L.E., Mao, D., Vialpando, C.G. & Salmon, kuyl, A.E., Bultink, I.E., Ruffatti, A., Rozman, includes a lipid-binding inhibitor of coagulation:

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 13


Review

beta 2-glycoprotein I (apolipoprotein H). Pro- antiphospholipid antibodies, and pregnancy. aspirin plus heparin in pregnant women with
ceedings of the National Academy of Sciences of Annals of Internal Medicine, 110, 495–496. recurrent miscarriage associated with phospho-
the United States of America, 87, 4120–4124. Pelkmans, L., Kelchtermans, H., de Groot, P.G., lipid antibodies (or antiphospholipid antibod-
Mekinian, A., Lazzaroni, M.G., Kuzenko, A., Alijo- Zuily, S., Regnault, V., Wahl, D., Pengo, V. & ies). BMJ, 314, 253–257.
tas-Reig, J., Ruffatti, A., Levy, P., Canti, V., de Laat, B. (2013) Variability in exposure of epi- Rand, J.H., Wu, X.X., Andree, H.A., Lockwood,
Bremme, K., Bezanahary, H., Bertero, T., Dhote, tope G40-R43 of domain i in commercial anti- C.J., Guller, S., Scher, J. & Harpel, P.C. (1997)
R., Maurier, F., Andreoli, L., Benbara, A., Tiga- beta2-glycoprotein I IgG ELISAs. PLoS ONE, 8, Pregnancy loss in the antiphospholipid-antibody
zin, A., Carbillon, L., Nicaise-Roland, P., Tin- e71402. syndrome–a possible thrombogenic mechanism.
cani, A. & Fain, O. (2015) The efficacy of Pengo, V., Biasiolo, A., Pegoraro, C., Cucchini, U., The New England Journal of Medicine, 337, 154–
hydroxychloroquine for obstetrical outcome in Noventa, F. & Iliceto, S. (2005) Antibody pro- 160.
anti-phospholipid syndrome: data from a Euro- files for the diagnosis of antiphospholipid syn- Rand, J.H., Wu, X.X., Quinn, A.S. & Taatjes, D.J.
pean multicenter retrospective study. Autoimmu- drome. Thrombosis and Haemostasis, 93, 1147– (2010a) The annexin A5-mediated pathogenic
nity Reviews, 14, 498–502. 1152. mechanism in the antiphospholipid syndrome:
Miyakis, S., Lockshin, M.D., Atsumi, T., Branch, Pengo, V., Biasiolo, A., Gresele, P., Marongiu, F., role in pregnancy losses and thrombosis. Lupus,
D.W., Brey, R.L., Cervera, R., Derksen, R.H., DE Erba, N., Veschi, F., Ghirarduzzi, A., Barcellona, 19, 460–469.
Groot, P.G., Koike, T., Meroni, P.L., Reber, G., D. & Tripodi, A. (2007) A comparison of lupus Rand, J.H., Wu, X.X., Quinn, A.S., Ashton, A.W.,
Shoenfeld, Y., Tincani, A., Vlachoyiannopoulos, anticoagulant-positive patients with clinical pic- Chen, P.P., Hathcock, J.J., Andree, H.A. & Taat-
P.G. & Krilis, S.A. (2006) International consen- ture of antiphospholipid syndrome and those jes, D.J. (2010b) Hydroxychloroquine protects
sus statement on an update of the classification without. Arteriosclerosis, Thrombosis, and Vascu- the annexin A5 anticoagulant shield from dis-
criteria for definite antiphospholipid syndrome lar Biology, 27, e309–e310. ruption by antiphospholipid antibodies: evi-
(APS). Journal of Thrombosis and Haemostasis: Pengo, V., Ruffatti, A., Legnani, C., Gresele, P., dence for a novel effect for an old antimalarial
JTH, 4, 295–306. Barcellona, D., Erba, N., Testa, S., Marongiu, F., drug. Blood, 115, 2292–2299.
Nayar, R. & Lage, J.M. (1996) Placental changes in Bison, E., Denas, G., Banzato, A., Padayattil, J.S. Reynaud, Q., Lega, J.C., Mismetti, P., Chapelle, C.,
a first trimester missed abortion in maternal sys- & Iliceto, S. (2010) Clinical course of high-risk Wahl, D., Cathebras, P. & Laporte, S. (2014)
temic lupus erythematosus with antiphospho- patients diagnosed with antiphospholipid syn- Risk of venous and arterial thrombosis accord-
lipid syndrome; a case report and review of the drome. Journal of Thrombosis and Haemostasis, ing to type of antiphospholipid antibodies in
literature. Human Pathology, 27, 201–206. 8, 237–242. adults without systemic lupus erythematosus: a
Noel, N., Dutasta, F., Costedoat-Chalumeau, N., Pengo, V., Ruffatti, A., Legnani, C., Testa, S., systematic review and meta-analysis. Autoimmu-
Bienvenu, B., Mariette, X., Geffray, L., Sene, D., Fierro, T., Marongiu, F., De Micheli, V., Gresele, nity Reviews, 13, 595–608.
Chaidi, R.B., Michot, J.M., Fain, O., Darnige, L., P., Tonello, M., Ghirarduzzi, A., Bison, E., Ridker, P.M. (2009) The JUPITER trial: results,
Ankri, A., Cacoub, P., Piette, J.C. & Saadoun, D. Denas, G., Banzato, A., Padayattil, J.S. & Iliceto, controversies, and implications for prevention.
(2015) Safety and efficacy of oral direct inhibitors S. (2011) Incidence of a first thromboembolic Circulation: Cardiovascular Quality and Out-
of thrombin and factor Xa in antiphospholipid event in asymptomatic carriers of high-risk comes, 2, 279–285.
syndrome. Autoimmunity Reviews, 14, 680–685. antiphospholipid antibody profile: a multicenter Rodriguez-Garcia, V., Ioannou, Y., Fernandez-
Nojima, J., Kuratsune, H., Suehisa, E., Iwatani, Y. prospective study. Blood, 118, 4714–4718. Nebro, A., Isenberg, D.A. & Giles, I.P. (2015)
& Kanakura, Y. (2005) Acquired activated pro- Pengo, V., Ruffatti, A., Tonello, M., Cuffaro, S., Examining the prevalence of non-criteria anti-
tein C resistance associated with IgG antibodies Banzato, A., Bison, E., Denas, G. & Padayattil, phospholipid antibodies in patients with anti-
against beta2-glycoprotein I and prothrombin as J.S. (2015) Antiphospholipid syndrome: anti- phospholipid syndrome: a systematic review.
a strong risk factor for venous thromboem- bodies to Domain 1 of beta2-glycoprotein 1 cor- Rheumatology (Oxford), 54, 2042–2050.
bolism. Clinical Chemistry, 51, 545–552. rectly classify patients at risk. Journal of Romay-Penabad, Z., Carrera Marin, A.L., Willis,
Oku, K., Atsumi, T., Bohgaki, M., Amengual, O., Thrombosis and Haemostasis, 13, 782–787. R., Weston-Davies, W., Machin, S., Cohen, H.,
Kataoka, H., Horita, T., Yasuda, S. & Koike, T. Pericleous, C., Ruiz-Limon, P., Romay-Penabad, Brasier, A. & Gonzalez, E.B. (2014) Complement
(2009) Complement activation in patients with Z., Marin, A.C., Garza-Garcia, A., Murfitt, L., C5-inhibitor rEV576 (coversin) ameliorates in-
primary antiphospholipid syndrome. Annals of Driscoll, P.C., Latchman, D.S., Isenberg, D.A., vivo effects of antiphospholipid antibodies.
the Rheumatic Diseases, 6, 1030–1035. Giles, I., Ioannou, Y., Rahman, A. & Pierangeli, Lupus, 23, 1324–1326.
van Os, G.M., Meijers, J.C., Agar, C., Seron, M.V., S.S. (2015) Proof-of-concept study demonstrat- Ruffatti, A., Del, R.T., Ciprian, M., Bertero, M.T.,
Marquart, J.A., Akesson, P., Urbanus, R.T., ing the pathogenicity of affinity-purified IgG Sciascia, S., Scarpato, S., Montecucco, C., Rossi,
Derksen, R.H., Herwald, H., Morgelin, M. & antibodies directed to domain I of beta2-glyco- S., Caramaschi, P., Biasi, D., Doria, A., Ram-
Groot Pg, D.E. (2011) Induction of anti-beta2 - protein I in a mouse model of anti-phospholi- pudda, M., Monica, N., Fischetti, F., Picillo, U.,
glycoprotein I autoantibodies in mice by protein pid antibody-induced thrombosis. Rheumatology Brucato, A., Salvan, E., Vittorio, P., Meroni, P.
H of Streptococcus pyogenes. Journal of Throm- (Oxford), 54, 722–727. & Tincani, A. (2011) Risk factors for a first
bosis and Haemostasis, 9, 2447–2456. Pierangeli, S.S., Liu, X.W., Barker, J.H., Anderson, thrombotic event in antiphospholipid antibody
Ostertag, M.V., Liu, X., Henderson, V. & Pieran- G. & Harris, E.N. (1995) Induction of thrombo- carriers: a prospective multicentre follow-up
geli, S.S. (2006) A peptide that mimics the Vth sis in a mouse model by IgG, IgM and IgA study. Annals of the Rheumatic Diseases, 70,
region of beta-2-glycoprotein I reverses immunoglobulins from patients with the 1083–1086.
antiphospholipid-mediated thrombosis in mice. antiphospholipid syndrome. Thrombosis and Ruiz-Irastorza, G., Khamashta, M.A., Hunt, B.J.,
Lupus, 15, 358–365. Haemostasis, 5, 1361–1367. Escudero, A., Cuadrado, M.J. & Hughes, G.R.
Out, H.J., Kooijman CD, F.A.U., Bruinse HW, Pierangeli, S.S., Chen, P.P., Raschi, E., Scurati, S., (2002) Bleeding and recurrent thrombosis in
F.A.U. & Derksen, R.H. (1991) Histopathologi- Grossi, C., Borghi, M.O., Palomo, I., Harris, definite antiphospholipid syndrome: analysis of
cal findings in placentae from patients with E.N. & Meroni, P.L. (2008) Antiphospholipid a series of 66 patients treated with oral anticoag-
intra-uterine fetal death and anti-phospholipid antibodies and the antiphospholipid syndrome: ulation to a target international normalized ratio
antibodies. European Journal of Obstetrics, Gyne- pathogenic mechanisms. Seminars in Thrombosis of 3.5. Archives of Internal Medicine, 162, 1164–
cology, and Reproductive Biology, 41, 179–186. and Hemostasis, 34, 236–250. 1169.
Parke, A., Maier, D., Wilson, D., Andreoli, J. & Rai, R., Cohen, H., Dave, M. & Regan, L. (1997) Ruiz-Irastorza, G., Hunt, B.J. & Khamashta, M.A.
Ballow, M. (1989) Intravenous gamma-globulin, Randomised controlled trial of aspirin and (2007) A systematic review of secondary

14 ª 2017 John Wiley & Sons Ltd, British Journal of Haematology


Review

thromboprophylaxis in patients with antiphos- Sciascia, S., Hunt, B.J., Talavera-Garcia, E., Lliso, induced murine fetal loss: novel protective effect
pholipid antibodies. Arthritis and Rheumatism, G., Khamashta, M.A. & Cuadrado, M.J. (2016) of a peptide targeting the beta2 glycoprotein I
57, 1487–1495. The impact of hydroxychloroquine treatment on phospholipid-binding site. Implications for
Ruiz-Irastorza, G., Ramos-Casals, M., Brito-Zeron, pregnancy outcome in women with antiphos- human fetal loss. Journal of Autoimmunity, 38,
P. & Khamashta, M.A. (2010) Clinical efficacy pholipid antibodies. American Journal of Obstet- J209–J215.
and side effects of antimalarials in systemic rics and Gynecology, 214, 273.e1-8. Tripodi, A., Chantarangkul, V., Clerici, M., Negri,
lupus erythematosus: a systematic review. Annals Shapira, I., Andrade, D., Allen, S.L. & Salmon, J.E. B., Galli, M. & Mannucci, P.M. (2001) Labora-
of the Rheumatic Diseases, 69, 20–28. (2012) Brief report: induction of sustained tory control of oral anticoagulant treatment by
Ruiz-Irastorza, G., Cuadrado, M.J., Ruiz-Arruza, I., remission in recurrent catastrophic antiphos- the INR system in patients with the antiphos-
Brey, R., Crowther, M., Derksen, R., Erkan, D., pholipid syndrome via inhibition of terminal pholipid syndrome and lupus anticoagulant.
Krilis, S., Machin, S., Pengo, V., Pierangeli, S., complement with eculizumab. Arthritis and Results of a collaborative study involving nine
Tektonidou, M. & Khamashta, M. (2011) Evi- Rheumatism, 64, 2719–2723. commercial thromboplastins. British Journal of
dence-based recommendations for the preven- Signorelli, F., Nogueira, F., Domingues, V., Mariz, Haematology, 115, 672–678.
tion and long-term management of thrombosis H.A. & Levy, R.A. (2016) Thrombotic events in Willis, R., Gonzalez, E.B. & Brasier, A.R. (2015)
in antiphospholipid antibody-positive patients: patients with antiphospholipid syndrome treated The journey of antiphospholipid antibodies
report of a task force at the 13th International with rivaroxaban: a series of eight cases. Clinical from cellular activation to antiphospholipid syn-
Congress on antiphospholipid antibodies. Lupus, Rheumatology, 35, 801–805. drome. Current Rheumatology Reports, 17, 16–
20, 206–218. Son, M., Wypasek, E., Celinska-Lowenhoff, M. & 0485.
Salmon, J.E., Girardi, G. & Holers, V.M. (2003) Undas, A. (2015) The use of rivaroxaban in Win, K. & Rodgers, G.M. (2014) New oral antico-
Activation of complement mediates antiphos- patients with antiphospholipid syndrome: a ser- agulants may not be effective to prevent venous
pholipid antibody-induced pregnancy loss. ies of 12 cases. Thrombosis Research, 135, 1035– thromboembolism in patients with antiphospho-
Lupus, 12, 535–538. 1036. lipid syndrome. American Journal of Hematology,
Schaefer, J.K., Mcbane, R.D., Black, D.F., Williams, Sperber, K., Quraishi, H., Kalb, T.H., Panja, A., 89, 1017.
L.N., Moder, K.G. & Wysokinski, W.E. (2014) Stecher, V. & Mayer, L. (1993) Selective regula- Xu, C., Mao, D., Holers, V.M., Palanca, B., Cheng,
Failure of dabigatran and rivaroxaban to prevent tion of cytokine secretion by hydroxychloro- A.M. & Molina, H. (2000) A critical role for
thromboembolism in antiphospholipid syn- quine: inhibition of interleukin 1 alpha (IL-1- murine complement regulator crry in fetomater-
drome: a case series of three patients. Thrombo- alpha) and IL-6 in human monocytes and T nal tolerance. Science, 287, 498–501.
sis and Haemostasis, 112, 947–950. cells. Journal of Rheumatology, 20, 803–808. Yu, P., Passam, F.H., Yu, D.M., Denyer, G. & Kri-
Schmidt-Tanguy, A., Voswinkel, J., Henrion, D., The PROMISE study (Predictors of pRegnancy lis, S. (2008) b2-glycoprotein I inhibits vascular
Subra, J.F., Loufrani, L., Rohmer, V., Ifrah, N. & outcome: biomarkers In APS and Systemic lupus endothelial growth factor and basic fibroblast
Belizna, C. (2013) Antithrombotic effects of Erythematosus). Available at: https://clinicaltria growth factor induced angiogenesis through its
hydroxychloroquine in primary antiphospho- ls.gov/ct2/show/NCT00198068. (accessed 20 June amino terminaldomain. Journal of Thrombosis
lipid syndrome patients. Journal of Thrombosis 2016). and Haemostasis, 6, 1215–1223.
and Haemostasis, 11, 1927–1929. de la Torre, Y.M., Pregnolato, F., D’Amelio, F., Zhang, J. & Mccrae, K.R. (2005) Annexin A2
Sciascia, S., Breen, K. & Hunt, B.J. (2015) Rivarox- Grossi, C., Di, S.N., Pasqualini, F., Nebuloni, mediates endothelial cell activation by antiphos-
aban use in patients with antiphospholipid syn- M., Chen, P., Pierangeli, S., Bassani, N., pholipid/anti-beta2 glycoprotein I antibodies.
drome and previous venous thromboembolism. Ambrogi, F., Borghi, M.O., Vecchi, A., Locati, Blood, 105, 1964–1969.
Blood Coagulation & Fibrinolysis, 26, 476–477. M. & Meroni, P.L. (2012) Anti-phospholipid

ª 2017 John Wiley & Sons Ltd, British Journal of Haematology 15

You might also like