ArticlePDF AvailableLiterature Review

Antiphospholipid syndrome: an update for clinicians and scientists

Authors:

Abstract and Figures

Purpose of review: Antiphospholipid syndrome (APS) is a leading acquired cause of thrombosis and pregnancy loss. Upon diagnosis (which is unlikely to be made until at least one morbid event has occurred), anticoagulant medications are typically prescribed in an attempt to prevent future events. This approach is not uniformly effective and does not prevent associated autoimmune and inflammatory complications. The goal of this review is to update clinicians and scientists on mechanistic and clinically relevant studies from the past 18 months, which have especially focused on inflammatory aspects of APS pathophysiology. Recent findings: How antiphospholipid antibodies leverage receptors and signaling pathways to activate cells is being increasingly defined. Although established mediators of disease pathogenesis (like endothelial cells and the complement system) continue to receive intensive study, emerging concepts (such as the role of neutrophils) are also receiving increasing attention. In-vivo animal studies and small clinical trials are demonstrating how repurposed medications (hydroxychloroquine, statins, and rivaroxaban) may have clinical benefit in APS, with these concepts importantly supported by mechanistic data. Summary: As anticoagulant medications are not uniformly effective and do not comprehensively target the underlying pathophysiology of APS, there is a continued need to reveal the inflammatory aspects of APS, which may be modulated by novel and repurposed therapies.
Content may be subject to copyright.
Antiphospholipid syndrome: an update for clinicians and
scientists
Andrew P. Vreede, M.D.1, Paula L. Bockenstedt, M.D.2, and Jason S. Knight, M.D., Ph.D.1
1Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical
School, Ann Arbor, Michigan, USA
2Division of Hematology, Department of Internal Medicine, University of Michigan Medical School,
Ann Arbor, Michigan, USA
Abstract
Purpose of review—Antiphospholipid syndrome (APS) is a leading acquired cause of
thrombosis and pregnancy loss. Upon diagnosis (which is not made until at least one morbid event
has occurred), anticoagulant medications are typically prescribed in an attempt to prevent future
events. This approach is not uniformly effective and does not prevent associated autoimmune and
inflammatory complications. The goal of this review is to update clinicians and scientists on
mechanistic and clinically-relevant studies from the past 18 months, which have especially
focused on inflammatory aspects of APS pathophysiology.
Recent findings—How antiphospholipid antibodies leverage receptors and signaling pathways
to activate cells are being increasingly defined. While established mediators of disease
pathogenesis (like endothelial cells and the complement system) continue to receive intensive
study, emerging concepts (such as the role of neutrophils) are also receiving increasing attention.
In vivo
animal studies and small clinical trials are demonstrating how repurposed medications
(hydroxychloroquine, statins, rivaroxaban) may have clinical benefit in APS, with these concepts
importantly supported by mechanistic data.
Summary—As anticoagulant medications are not uniformly effective and do not
comprehensively target the underlying pathophysiology of APS, there is a continued need to reveal
the inflammatory aspects of APS, which may be modulated by novel and repurposed therapies.
Keywords
Antiphospholipid syndrome; thrombosis; pregnancy loss; endothelial cells; neutrophils;
complement
Introduction
Vascular complications, including thrombotic events, are among the leading causes of
morbidity and mortality in lupus. Antiphospholipid antibodies (aPL), a major driver of
Correspondence: Jason S. Knight, M.D., Ph.D., 5520A MSRB 1, 1150 W Medical Center Drive, SPC 5680, Ann Arbor, MI
48109-5680, Tel: 734-936-3257, jsknight@umich.edu.
Conflicts of interest: The authors have no conflicts of interest to disclose.
HHS Public Access
Author manuscript
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Published in final edited form as:
Curr Opin Rheumatol
. 2017 September ; 29(5): 458–466. doi:10.1097/BOR.0000000000000410.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
thrombosis risk, are present in up to one-third of lupus patients. When aPL are associated
with certain clinical complications (either thrombotic or obstetric), a diagnosis of
antiphospholipid syndrome (APS) is assigned (Table 1) [1]. Beyond lupus-associated APS,
approximately half of APS cases will be diagnosed as a standalone syndrome (i.e., primary
APS) [2].
APS is a leading acquired cause of thrombosis and pregnancy loss, with an estimated
prevalence of 1 in 2,000 [3]. Framing this risk another way, aPL can be detected on the order
of 10% of the time in the setting of certain events including pregnancy morbidity, stroke,
myocardial infarction, and deep venous thrombosis (DVT) [4]. Emphasizing the systemic
nature of APS, the diagnosis also portends risk for cytopenias (especially hemolytic anemia
and thrombocytopenia), mitral and aortic valve lesions, seizure disorder, accelerated
cognitive decline, and nephropathy in the form of thrombotic microangiopathy [5]. The
approach to treatment is typically with anticoagulant drugs, which are not uniformly
effective in preventing recurrent aPL-mediated thrombosis and pregnancy loss, and offer
insufficient protection against the varied “non-criteria” manifestations of APS. Indeed, 44%
of “triple-positive” APS patients (positive testing for anticardiolipin, anti-beta-2-
glycoprotein I,
and
lupus anticoagulant) will develop recurrent thrombosis over a 10-year
follow-up period (even with the majority being prescribed anticoagulants) [6]. Furthermore,
at least 20% of obstetric APS patients have adverse outcomes in spite of therapy with aspirin
and low-molecular-weight heparin [7].
Despite its high prevalence and potential for devastating morbidity, APS pathophysiology
has yet to be fully defined. APS was historically viewed as a coagulation problem; however,
clinical observations and basic science discoveries are increasingly highlighting a more
multifaceted syndrome with an associated (and perhaps even central) inflammatory
component [8]. Herein we will discuss recent discoveries over the past 18 months, which
have continued to increase our understanding of APS pathophysiology. We will also discuss
how this improved basic understanding may translate to new and repurposed therapeutics for
APS (Table 2)
Cell activation and signaling pathways: new concepts
Understanding the cellular signaling pathways that mediate APS pathogenesis has remained
somewhat elusive, at least partially the consequence of study heterogeneity. Studies have
utilized different types of aPL (monoclonal vs. patient-derived; protein cofactor-dependent
vs. -independent) and have focused on a variety of cellular targets (endothelial cells,
platelets, monocytes, neutrophils, trophoblast cells, etc.).
Many (perhaps most) pathogenic antibodies in APS do not target phospholipids themselves,
but rather phospholipid-binding protein cofactors. The best characterized of these cofactors
is beta-2 glycoprotein I (β2GPI), a lipid-binding protein present at high levels in plasma
[22,23], albeit with largely unknown endogenous function. The mechanistic schema is that
anti-β2GPI antibodies potentiate thrombosis by engaging β2GPI protein that has been
recruited to cell surfaces—and thereby promote cell activation [24–26]. The mechanisms by
which anti-β2GPI antibodies activate cells have been recently reviewed [27], with roles
Vreede et al. Page 2
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
especially suggested for the cell surface proteins annexin A2, apolipoprotein E receptor 2
(ApoER2), Toll-like receptor 2 (TLR2), and TLR4 [27].
ApoER2 (also known as LDL receptor-related protein 8) is one receptor for β2GPI (and
consequently β2GPI-dependent aPL) on monocytes, endothelial cells, and platelets. Indeed,
in a 2011 study, Ramesh and colleagues demonstrated ApoER2−/− mice are relatively
resistant to thrombosis when confronted with aPL [28]. More recently, it has been revealed
that ApoER2 may play an important role in obstetric APS [29]. Specifically, Ulrich and
colleagues demonstrated enhanced placental trophoblast cell proliferation and migration
in
vitro
when aPL engage β2GPI/ApoER2 complexes on the trophoblast cell surface [29].
Extending these studies to an
in vivo
model of aPL-mediated pregnancy loss, they
demonstrated protection in ApoER2−/− mice [29]. In another recent study, Mineo and
colleagues developed a monoclonal antibody against β2GPI that prevents pathogenic aPL
binding, thereby protecting against aPL-mediated cell activation [30]*. The antibody
attenuated the association of β2GPI with ApoER2, thereby normalizing endothelial and
trophoblast cell function
in vitro
, as well as preventing thrombosis and fetal loss
in vivo
[30]*. Although further study is clearly needed, the intersection of aPL, β2GPI, and ApoER2
warrants further investigation as a potential therapeutic target in patients.
Since neither β2GPI itself, nor some β2GPI “receptors” such as annexin A2, have a
cytoplasmic domain to mediate signaling, there has been interest in additional partner
proteins that may convey activating signals to the cytoplasm. On this front, particular
attention has been given to the cell-surface TLRs, TLR2 and TLR4. In mouse models, TLR4
deletion protects against venous and arterial thrombosis in some [31–33], but not all [34]*,
studies (it is worth pointing out that the latter study utilized cofactor-independent aPL).
Studies of obstetric APS have also yielded mixed results with an older study demonstrating
no role for TLR4 in an
in vivo
model of pregnancy loss [35]. In contrast, Azuma and
colleagues recently suggested that, at least
in
vitro, TLR2 and TLR4 facilitate inflammatory
cytokine production by trophoblast cells in response to anti-β2GPI antibodies [36].
Signaling pathways downstream of the aforementioned receptors, at least as they relate to
APS pathogenesis, remain incompletely understood. Terrisse and colleagues recently
investigated downstream signaling pathways by which aPL (especially IgG isolated from
APS patients) activate platelets [37]*. The authors demonstrated that aPL potentiate
ex vivo
platelet activation through surface glycoprotein Ibα (the platelet receptor for von Willebrand
factor) and TLR2, via a mechanism involving class IA phosphoinositide 3-kinase (PI3K) α
and β isoforms [37]*. At least one downstream consequence of PI3K signaling is activation
of the serine/threonine kinase Akt, a pathway that supports cell survival, proliferation, and
migration [37]*. Indeed, PI3K inhibitors, which are being explored as potential drug targets
in other contexts [38], are effective at preventing aPL-mediated platelet activation [37]*.
Interestingly, another study has suggested that Akt activation is a downstream consequence
of trophoblast cell activation by aPL [29].
Beyond the engagement of aPL with cell surfaces, a recent report by Wu and colleagues
suggests an intriguing new mechanism by which aPL-activated endothelial cells may
propagate this activation in paracrine fashion to other endothelial cells [39]*. Anti-β2GPI
Vreede et al. Page 3
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
antibodies trigger the release of “extracellular vesicles” from endothelial cells, which the
authors define as inclusive of both microparticles and exosomes [39]*. These vesicles then
activate endothelial cells through a mechanism that is not dependent upon packaged
cytokines such as IL-1, but rather single-stranded RNA that signals through TLR7 in the
recipient cell [39]*. They also speculate that these vesicles may be a mechanism for delivery
of specific and functionally-relevant micro-RNA, although this hypothesis requires further
study.
The vessel wall: endothelial progenitors and interferons
Our group recently looked “upstream” of endothelial cells, asking whether a deficiency in
reparative, circulating endothelial progenitors might contribute to defective maintenance and
health of the endothelium over time. Indeed, a deficiency in the number and function of such
progenitors is a well-recognized aspect of both lupus and rheumatoid arthritis [40]. We
found that primary APS patients have a reduction in functional endothelial progenitors,
which was interestingly not dependent upon patient IgG; rather, we discovered a type I IFN
signature in the APS patients, abrogation of which could restore normal progenitor function
[41]*. These findings were recently replicated by van den Hoogen and colleagues, who
found that approximately 50% of primary APS patients have a type I IFN signature, which
was less likely to be present in patients taking either hydroxychloroquine or statins [20]**.
Interestingly, they also found that the IFN signature correlated with expansion of
“intermediate” and “non-classical” monocytes (which have been previously linked to
cardiovascular disease in lupus and rheumatoid arthritis) [20]**. How these monocytes
intersect with endothelial progenitors [42], and whether there is a role for anti-interferon
therapy in APS [43], are questions that deserve further consideration.
One potential consequence of endothelial cell (and progenitor) dysfunction is
atherosclerosis, an accelerated version of which is a well-known complication of lupus [44],
and which has also been reported in APS [45,46]. The recent work of Benagiano and
colleagues has examined the role of TH1 specific inflammatory responses to β2GPI in
established atherosclerotic lesions of primary APS patients. Their work demonstrated that
plaque-derived, β2GPI-specific CD4+ T lymphocytes facilitate perforin- and Fas ligand-
mediated cytotoxicity, pointing to a role for these autoreactive T cells in plaque
destabilization (and potentially the arterial thrombotic events that are known to occur at
higher frequency in APS) [47]**. They also demonstrated that β2GPI can induce
proliferation of (and IFN-γ expression by) plaque-derived T cell clones [47]**.
Furthermore, these T cells amplify monocyte responses, such as the production of tissue
factor and matrix metalloproteinases, which can be inhibited with an anti-IFN-γ antibody
[47]**.
Myeloid-lineage cells: neutrophil extracellular traps (NETs) and monocyte
NOX2
The role of neutrophils in APS pathogenesis has only recently been investigated. This
interest was precipitated by emerging descriptions of neutrophils as mediators of both
pathologic clotting and autoimmune diseases [48,49]. In particular, NETs (extracellular
Vreede et al. Page 4
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
chromatin-based structures released by activated neutrophils) have been described as triggers
of autoimmunity and tissue damage, as well as important instigators of thrombosis [50].
With this background in mind [51], our group recently identified increased levels of cell-free
DNA and NETs in the circulation of primary APS patients, as compared with healthy
controls [52]**. When APS neutrophils were cultured
in vitro
, they demonstrated an
enhanced propensity to spontaneously release NETs [52]**. Mechanistically, anti-β2GPI
IgG appears to be at least one factor in patient blood that supports NET release, with the
mechanism dependent upon both TLR4 and formation of reactive oxygen species [52]**.
Furthermore, the prothrombotic potential of aPL-mediated NETs was demonstrated in a
thrombin generation assay, with this potential abrogated by treatment with
deoxyribonuclease (DNase) [52]**. In parallel to our work, van den Hoogen and colleagues
reported increased levels of circulating “low-density granulocytes” or LDGs in patients with
primary APS [53]. This pro-inflammatory subset of neutrophils has been well characterized
in SLE and other autoimmune disorders, where they are reported to release NETs in
exaggerated fashion [54]. Whether LDGs are important sources of NETs in APS awaits
further study [55].
The
in vivo
relevance of NETs was recently confirmed by our group in a mouse model of
APS. In this model, IgG from triple-positive APS patients potentiated venous thrombosis in
mice that had been subjected to flow restriction in the inferior vena cava by a standard
surgical stenosis [56]*. As compared with control mice, mice treated with APS IgG were
twice as likely to develop macroscopic thrombi in response to flow restriction.
Mechanistically, APS thrombi were enriched for NETs, while patient IgG could be detected
on the surface of circulating neutrophils [56]*. Furthermore, APS IgG-mediated thrombosis
could be reversed by either neutrophil depletion or administration of systemic DNase [56]*.
Around the same time, Manukyan and colleagues published an elegant study demonstrating
that cofactor-independent aPL could similarly potentiate thrombosis in an inferior vena cava
flow-restriction model [34]*. Their interesting work found a major role for leukocyte
activation in thrombus formation, which could be abrogated by deletion of NOX2 (the
catalytic subunit of NADPH oxidase) from bone marrow-derived cells. While the authors’
primary interest was in monocyte NOX2 and its role in tissue factor expression, there is also
a well-accepted role for neutrophil NOX2 in NET formation [57]. Further studies may assess
the role of these cofactor-independent antiphospholipid antibodies in inducing NET release
in vitro
and
in vivo
.
Complement: at the intersection of coagulation and inflammation in APS
Animal models of APS have supported a role for complement activation in both thrombotic
events and pregnancy loss [58,59]. Studies in APS patients have demonstrated smoldering
activity of the complement cascade [60–62], while a recent case report revealed deposition
of β2GPI protein, IgG, and complement components C1q, C4, C3, and C5b-9 at the
endothelial surface of an occluded artery in an APS patient [63]. Furthermore, this patient,
who had suffered recurrent arterial occlusions, was successfully revascularized while under
treatment with eculizumab, a terminal complement inhibitor [63].
Vreede et al. Page 5
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
In lupus, antibodies to C1q (a complex that initiates the complement cascade in response to
immune complexes) amplify complement activation and strongly correlate with certain
clinical manifestations such as proliferative nephritis [64]. Oku and colleagues recently
investigated these antibodies in primary APS patients, demonstrating that 36% of patients
had detectable anti-C1q (compared to 55% of lupus patients) [65]. Interestingly titers of
anti-C1q were significantly higher in patients with refractory APS [65].
Rivaroxaban, a direct factor Xa inhibitor, has recently been proposed as an alternative agent
to vitamin K antagonists in APS. The first randomized, prospective study investigating use
of rivaroxaban in APS (RAPS trial) was recently published. In patients with a history of
venous thromboembolism (who had already demonstrated stable disease on warfarin), both
warfarin and rivaroxaban prevented new thrombotic events for 210 days in every study
patient [14]**. Bleeding events and overall adverse events were also similar between the
groups [14]**. While a full recounting of this important trial is beyond the scope of this
brief review, we would refer you to a detailed comment on the topic [66]. Related to our
discussion of the complement pathway, a post-hoc analysis of the RAPS trial revealed that,
prior to randomization, APS patients had significantly higher markers of complement
activation as compared with normal controls [19]*. While patients in the warfarin group
showed stable elevation of these markers over time, patients randomized to rivaroxaban
demonstrated decreased C3a, C5a, and soluble C5b-9 (all markers of classical pathway
activation) [19]*. In contrast, the alternative pathway marker, Bb, was unchanged with
rivaroxaban treatment [19]*. Whether direct oral anticoagulants have additional anti-
inflammatory properties is a topic that certainly warrants further study.
Repurposing medications: statins and hydroxychloroquine as adjuvant
therapies in APS?
HMG-CoA reductase inhibitors (or statins) have long been recognized to have pleotropic
anti-inflammatory effects supportive of vascular health, including reductions in
inflammation, oxidative stress, and coagulation [67]. Clinically, statins appear to reduce the
risk of venous thromboembolism in the general population [13]. In mouse models of APS,
statins mitigate aPL-mediated thrombotic events and fetal death [11,16]. Furthermore, when
administered to APS patients, statins decrease both prothrombotic and proinflammatory
biomarkers [68].
The standard of care for managing pregnancy complications in APS is the administration of
low-dose aspirin and low-molecular-weight heparin (the latter at either prophylactic or
therapeutic doses, depending on the patient’s thrombosis history) [69,70]. However, as
detailed in recent review articles [69,70], pregnancy complications in APS are often not
based in frank placental thrombosis, but rather spiral artery vasculopathy, as well as acute
and chronic inflammation—with increased infiltration of inflammatory cells and deposition
of complement in the placentae of women with APS [71–73]. Lefkou and colleagues
recently investigated the use of pravastatin in refractory obstetric APS [18]**. In their
clinical trial, 21 patients with refractory obstetric APS (emergence of preeclampsia and/or
intrauterine growth restriction [IUGR] despite treatment with low-dose aspirin and low-
Vreede et al. Page 6
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
molecular-weight heparin) were randomized either to continue standard therapy or to receive
pravastatin 20 mg/day at the onset of preeclampsia/IUGR [18]**. There was a remarkable
therapeutic benefit, with all the patients receiving pravastatin delivering healthy infants at
34–38 weeks [18]**. In contrast, the 10 patients who remained on standard therapy had
three stillbirths at 25–26 weeks, and seven pre-term Cesarean sections (resulting in two fetal
deaths) [18]**.
Hydroxychloroquine (which is nowadays prescribed to essentially all patients with lupus)
was utilized in the 1970s to reduce the risk of venous-thromboembolism in post-operative
patients [12]. In the 1990s, hydroxychloroquine was demonstrated to protect against aPL-
mediated thrombosis in mice [9]. Furthermore, there have been hints of a reduction in
thrombosis risk in lupus patients taking hydroxychloroquine, as compared to those who are
not [74,75]. Mechanistically, a recent study demonstrated that hydroxychloroquine inhibits
the translocation of monocyte NOX2 to the endosome in response to stimulants such as
TNFα, IL-1β, and aPL [10]**. This was accompanied by mitigation of aPL-induced,
NOX2-mediated thrombus formation
in vivo
[10]**. As the related drug chloroquine has
been shown to antagonize NET release [21], further studies should continue to explore the
intersection of hydroxychloroquine, activated monocytes/neutrophils, and APS.
Given its excellent safety profile in pregnancy [76], and its nearly standard-of-care
application in lupus pregnancies, hydroxychloroquine has been increasingly considered as
adjuvant therapy in APS pregnancies. Indeed, recent retrospective studies have suggested a
beneficial effect of hydroxychloroquine in APS pregnancies [7,17]. In a mouse model of
obstetric APS, Bertolaccini and colleagues recently demonstrated that hydroxychloroquine
prevents fetal death and placental metabolic changes [15]*. Going further, they
demonstrated that labeled aPL especially localize to the placenta and the developing fetal
brain, and that hydroxychloroquine mitigates complement deposition at both sites (which
correlated with lower levels of C3a and C5a in blood) [15]*. Intriguingly, C3a and C5a were
also reduced in the blood of APS patients after 6 months of hydroxychloroquine treatment
[15]*.
Conclusion
Since its description in the 1980s, APS has been managed primarily with anticoagulant
medications. These medications are not universally protective against subsequent thrombotic
events and pregnancy loss, and have little proven track record in treating “non-criteria”
manifestations of APS such as cytopenias and cardiac valvular disease. Basic science studies
continue to refine the signaling pathways, activated cells, and non-cellular effectors critical
for APS pathogenesis (Figure 1). In addition to a search for novel therapeutics, established
medications such as rivaroxaban, statins, and hydroxychloroquine are receiving increasing
interest as adjuvant therapies. In the near future, we hope to see more well-designed clinical
trials with both mechanistic and clinical endpoints.
Vreede et al. Page 7
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Acknowledgments
Financial support and sponsorship: JSK was supported by NIH K08AR066569 and career development awards
from the Burroughs Wellcome Fund, the Rheumatology Research Foundation, and the Arthritis National Research
Foundation.
References
1. Miyakis S, Lockshin MD, Atsumi T, Branch DW, Brey RL, Cervera R, Derksen RH, PGDEG, Koike
T, Meroni PL, et al. International consensus statement on an update of the classification criteria for
definite antiphospholipid syndrome (APS). J Thromb Haemost. 2006; 4:295–306. [PubMed:
16420554]
2. Bertolaccini ML, Amengual O, Andreoli L, Atsumi T, Chighizola CB, Forastiero R, de Groot P,
Lakos G, Lambert M, Meroni P, et al. 14th International Congress on Antiphospholipid Antibodies
Task Force. Report on antiphospholipid syndrome laboratory diagnostics and trends. Autoimmun
Rev. 2014; 13:917–930. [PubMed: 24824074]
3. Gomez-Puerta JA, Cervera R. Diagnosis and classification of the antiphospholipid syndrome. J
Autoimmun. 2014; 48–49:20–25.
4. Andreoli L, Chighizola CB, Banzato A, Pons-Estel GJ, Ramire de Jesus G, Erkan D. Estimated
frequency of antiphospholipid antibodies in patients with pregnancy morbidity, stroke, myocardial
infarction, and deep vein thrombosis: a critical review of the literature. Arthritis Care Res
(Hoboken). 2013; 65:1869–1873. [PubMed: 23861221]
5. Abreu MM, Danowski A, Wahl DG, Amigo MC, Tektonidou M, Pacheco MS, Fleming N,
Domingues V, Sciascia S, Lyra JO, et al. The relevance of “non-criteria” clinical manifestations of
antiphospholipid syndrome: 14th International Congress on Antiphospholipid Antibodies Technical
Task Force Report on Antiphospholipid Syndrome Clinical Features. Autoimmun Rev. 2015;
14:401–414. [PubMed: 25641203]
6. Pengo V, Ruffatti A, Legnani C, Gresele P, Barcellona D, Erba N, Testa S, Marongiu F, Bison E,
Denas G, et al. Clinical course of high-risk patients diagnosed with antiphospholipid syndrome. J
Thromb Haemost. 2010; 8:237–242. [PubMed: 19874470]
7. Mekinian A, Costedoat-Chalumeau N, Masseau A, Tincani A, De Caroli S, Alijotas-Reig J, Ruffatti
A, Ambrozic A, Botta A, Le Guern V, et al. Obstetrical APS: is there a place for
hydroxychloroquine to improve the pregnancy outcome? Autoimmun Rev. 2015; 14:23–29.
[PubMed: 25179813]
8. de Groot PG, Urbanus RT. Antiphospholipid Syndrome--Not a Noninflammatory Disease. Semin
Thromb Hemost. 2015; 41:607–614. [PubMed: 26276935]
9. Edwards MH, Pierangeli S, Liu X, Barker JH, Anderson G, Harris EN. Hydroxychloroquine
reverses thrombogenic properties of antiphospholipid antibodies in mice. Circulation. 1997;
96:4380–4384. [PubMed: 9416907]
10**. Muller-Calleja N, Manukyan D, Canisius A, Strand D, Lackner KJ. Hydroxychloroquine inhibits
proinflammatory signalling pathways by targeting endosomal NADPH oxidase. Ann Rheum Dis.
2016 This study demonstrates a new mechanism by which hydroxychloroquine may inhibit aPL-
mediated thrombosis, namely the inhibition of endosomal NADPH oxidase.
11. Ferrara DE, Liu X, Espinola RG, Meroni PL, Abukhalaf I, Harris EN, Pierangeli SS. Inhibition of
the thrombogenic and inflammatory properties of antiphospholipid antibodies by fluvastatin in an
in vivo animal model. Arthritis Rheum. 2003; 48:3272–3279. [PubMed: 14613293]
12. Carter AE, Eban R. Prevention of postoperative deep venous thrombosis in legs by orally
administered hydroxychloroquine sulphate. Br Med J. 1974; 3:94–95. [PubMed: 4853767]
13. Glynn RJ, Danielson E, Fonseca FA, Genest J, Gotto AM Jr, Kastelein JJ, Koenig W, Libby P,
Lorenzatti AJ, MacFadyen JG, et al. A randomized trial of rosuvastatin in the prevention of venous
thromboembolism. N Engl J Med. 2009; 360:1851–1861. [PubMed: 19329822]
14**. Cohen H, Hunt BJ, Efthymiou M, Arachchillage DR, Mackie IJ, Clawson S, Sylvestre Y,
Machin SJ, Bertolaccini ML, Ruiz-Castellano M, et al. Rivaroxaban versus warfarin to treat
patients with thrombotic antiphospholipid syndrome, with or without systemic lupus
erythematosus (RAPS): a randomised, controlled, open-label, phase 2/3, non-inferiority trial.
Vreede et al. Page 8
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Lancet Haematol. 2016; 3:e426–436. This is the first randomized controlled study investigating a
direct oral anticoagulant in APS patients. In a cohort of patients with history of venous
thromboembolism who already had stable disease on warfarin, both rivaroxaban and warfarin
prevented thrombosis over a 6-month period in all patients. [PubMed: 27570089]
15*. Bertolaccini ML, Contento G, Lennen R, Sanna G, Blower PJ, Ma MT, Sunassee K, Girardi G.
Complement inhibition by hydroxychloroquine prevents placental and fetal brain abnormalities
in antiphospholipid syndrome. J Autoimmun. 2016; 75:30–38. This study provides additional
mechanistic insights into how hydroxychloroquine may function as adjuvant therapy in obstetric
APS, namely by inhibition of complement activation and deposition. [PubMed: 27160365]
16. Redecha P, Franzke CW, Ruf W, Mackman N, Girardi G. Neutrophil activation by the tissue factor/
Factor VIIa/PAR2 axis mediates fetal death in a mouse model of antiphospholipid syndrome. J
Clin Invest. 2008; 118:3453–3461. [PubMed: 18802482]
17. Mekinian A, Lazzaroni MG, Kuzenko A, Alijotas-Reig J, Ruffatti A, Levy P, Canti V, Bremme K,
Bezanahary H, Bertero T, et al. The efficacy of hydroxychloroquine for obstetrical outcome in
anti-phospholipid syndrome: Data from a European multicenter retrospective study. Autoimmun
Rev. 2015; 14:498–502. [PubMed: 25617818]
18**. Lefkou E, Mamopoulos A, Dagklis T, Vosnakis C, Rousso D, Girardi G. Pravastatin improves
pregnancy outcomes in obstetric antiphospholipid syndrome refractory to antithrombotic therapy.
J Clin Invest. 2016; 126:2933–2940. This intriguing small clinical trial highlights the potential
role of statins in treatment of obstetric APS. [PubMed: 27454295]
19*. Arachchillage DR, Mackie IJ, Efthymiou M, Chitolie A, Hunt BJ, Isenberg DA, Khamashta M,
Machin SJ, Cohen H. Rivaroxaban limits complement activation compared with warfarin in
antiphospholipid syndrome patients with venous thromboembolism. J Thromb Haemost. 2016;
14:2177–2186. This study interestingly links coagulation and complement pathways,
demonstrating that the factor Xa inhibitor rivaroxaban suppresses smoldering complement
pathway activation in APS patients. [PubMed: 27541499]
20**. van den Hoogen LL, Fritsch-Stork RD, Versnel MA, Derksen RH, van Roon JA, Radstake TR.
Monocyte type I interferon signature in antiphospholipid syndrome is related to proinflammatory
monocyte subsets, hydroxychloroquine and statin use. Ann Rheum Dis. 2016 This study also
reports a type I IFN signature in primary APS patients, and finds that the signature is less likely
to be present in patients taking either hydroxychloroquine or statins. The authors also describe an
expansion of “non-classical” monocytes in the blood of primary APS patients; these cells have
been linked to cardiovascular damage in other autoimmune diseases.
21. Smith CK, Vivekanandan-Giri A, Tang C, Knight JS, Mathew A, Padilla RL, Gillespie BW,
Carmona-Rivera C, Liu X, Subramanian V, et al. Neutrophil extracellular trap-derived enzymes
oxidize high-density lipoprotein: an additional proatherogenic mechanism in systemic lupus
erythematosus. Arthritis Rheumatol. 2014; 66:2532–2544. [PubMed: 24838349]
22. Conti F, Sorice M, Circella A, Alessandri C, Pittoni V, Caronti B, Calderaro C, Griggi T, Misasi R,
Valesini G. Beta-2-glycoprotein I expression on monocytes is increased in anti-phospholipid
antibody syndrome and correlates with tissue factor expression. Clin Exp Immunol. 2003;
132:509–516. [PubMed: 12780700]
23. Caronti B, Calderaro C, Alessandri C, Conti F, Tinghino R, Palladini G, Valesini G. Beta2-
glycoprotein I (beta2-GPI) mRNA is expressed by several cell types involved in anti-phospholipid
syndrome-related tissue damage. Clin Exp Immunol. 1999; 115:214–219. [PubMed: 9933445]
24. Ma K, Simantov R, Zhang JC, Silverstein R, Hajjar KA, McCrae KR. High affinity binding of beta
2-glycoprotein I to human endothelial cells is mediated by annexin II. J Biol Chem. 2000;
275:15541–15548. [PubMed: 10809787]
25. Allen KL, Fonseca FV, Betapudi V, Willard B, Zhang J, McCrae KR. A novel pathway for human
endothelial cell activation by antiphospholipid/anti-beta2 glycoprotein I antibodies. Blood. 2012;
119:884–893. [PubMed: 22106343]
26. Sorice M, Longo A, Capozzi A, Garofalo T, Misasi R, Alessandri C, Conti F, Buttari B, Rigano R,
Ortona E, et al. Anti-beta2-glycoprotein I antibodies induce monocyte release of tumor necrosis
factor alpha and tissue factor by signal transduction pathways involving lipid rafts. Arthritis
Rheum. 2007; 56:2687–2697. [PubMed: 17665396]
Vreede et al. Page 9
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
27. Muller-Calleja N, Lackner KJ. Mechanisms of Cellular Activation in the Antiphospholipid
Syndrome. Semin Thromb Hemost. 2017
28. Ramesh S, Morrell CN, Tarango C, Thomas GD, Yuhanna IS, Girardi G, Herz J, Urbanus RT, de
Groot PG, Thorpe PE, et al. Antiphospholipid antibodies promote leukocyte-endothelial cell
adhesion and thrombosis in mice by antagonizing eNOS via beta2GPI and apoER2. J Clin Invest.
2011; 121:120–131. [PubMed: 21123944]
29. Ulrich V, Gelber SE, Vukelic M, Sacharidou A, Herz J, Urbanus RT, de Groot PG, Natale DR,
Harihara A, Redecha P, et al. ApoE Receptor 2 Mediation of Trophoblast Dysfunction and
Pregnancy Complications Induced by Antiphospholipid Antibodies in Mice. Arthritis Rheumatol.
2016; 68:730–739. [PubMed: 26474194]
30*. Mineo C, Lanier L, Jung E, Sengupta S, Ulrich V, Sacharidou A, Tarango C, Osunbunmi O, Shen
YM, Salmon JE, et al. Identification of a Monoclonal Antibody That Attenuates
Antiphospholipid Syndrome-Related Pregnancy Complications and Thrombosis. PLoS One.
2016; 11:e0158757. The authors describe a novel monoclonal antibody that targets β2GPI and
thereby prevents aPL-mediated cell activation. The study further extends our mechanistic
understanding of the association between β2GPI and ApoER2. [PubMed: 27463336]
31. Pierangeli SS, Vega-Ostertag ME, Raschi E, Liu X, Romay-Penabad Z, De Micheli V, Galli M,
Moia M, Tincani A, Borghi MO, et al. Toll-like receptor and antiphospholipid mediated
thrombosis: in vivo studies. Ann Rheum Dis. 2007; 66:1327–1333. [PubMed: 17261530]
32. Xie H, Kong X, Zhou H, Xie Y, Sheng L, Wang T, Xia L, Yan J. TLR4 is involved in the
pathogenic effects observed in a murine model of antiphospholipid syndrome. Clin Immunol.
2015; 160:198–210. [PubMed: 26065621]
33. Laplante P, Fuentes R, Salem D, Subang R, Gillis MA, Hachem A, Farhat N, Qureshi ST, Fletcher
CA, Roubey RA, et al. Antiphospholipid antibody-mediated effects in an arterial model of
thrombosis are dependent on Toll-like receptor 4. Lupus. 2016; 25:162–176. [PubMed: 26391610]
34*. Manukyan D, Muller-Calleja N, Jackel S, Luchmann K, Monnikes R, Kiouptsi K, Reinhardt C,
Jurk K, Walter U, Lackner KJ. Cofactor-independent human antiphospholipid antibodies induce
venous thrombosis in mice. J Thromb Haemost. 2016; 14:1011–1020. This work supports a role
for “cofactor-independent” aPL in APS pathogenesis. These aPL mediate thrombosis through a
mechanism that relies upon the catalytic subunit of NADPH oxidase, NOX2. [PubMed:
26786324]
35. Girardi G, Mackman N. Tissue factor in antiphospholipid antibody-induced pregnancy loss: a pro-
inflammatory molecule. Lupus. 2008; 17:931–936. [PubMed: 18827058]
36. Azuma H, Yamamoto T, Chishima F. Effects of anti-beta2-GPI antibodies on cytokine production
in normal first-trimester trophoblast cells. J Obstet Gynaecol Res. 2016; 42:769–775. [PubMed:
27098191]
37*. Terrisse AD, Laurent PA, Garcia C, Gratacap MP, Vanhaesebroeck B, Sie P, Payrastre B. The
class I phosphoinositide 3-kinases alpha and beta control antiphospholipid antibodies-induced
platelet activation. Thromb Haemost. 2016; 115:1138–1146. This study provides important clues
to ApoER2-mediated intracellular signaling in platelets, with a particular focus on PI3K-related
pathways. [PubMed: 26818901]
38. Nylander S, Kull B, Bjorkman JA, Ulvinge JC, Oakes N, Emanuelsson BM, Andersson M, Skarby
T, Inghardt T, Fjellstrom O, et al. Human target validation of phosphoinositide 3-kinase
(PI3K)beta: effects on platelets and insulin sensitivity, using AZD6482 a novel PI3Kbeta inhibitor.
J Thromb Haemost. 2012; 10:2127–2136. [PubMed: 22906130]
39*. Wu M, Barnard J, Kundu S, McCrae KR. A novel pathway of cellular activation mediated by
antiphospholipid antibody-induced extracellular vesicles. J Thromb Haemost. 2015; 13:1928–
1940. This work demonstrates a new mechanism by which aPL activate endothelial cells, namely
paracrine signaling via vesicle-delivered single-stranded RNA. [PubMed: 26264622]
40. Westerweel PE, Verhaar MC. Endothelial progenitor cell dysfunction in rheumatic disease. Nat
Rev Rheumatol. 2009; 5:332–340. [PubMed: 19434075]
41*. Grenn RC, Yalavarthi S, Gandhi AA, Kazzaz NM, Nunez-Alvarez C, Hernandez-Ramirez D,
Cabral AR, McCune WJ, Bockenstedt PL, Knight JS. Endothelial progenitor dysfunction
associates with a type I interferon signature in primary antiphospholipid syndrome. Ann Rheum
Dis. 2016 This work demonstrates for the first time a type I IFN signature in primary APS
Vreede et al. Page 10
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
patients, which promotes dysfunction of circulating endothelial progenitors. Mechanistically, this
could contribute to accrual of endothelial damage over time.
42. Yalavarthi S, Grenn RC, Knight JS. Response to: ‘Monocyte type I interferon signature in
antiphospholipid syndrome is related to pro-inflammatory monocyte subsets, hydroxychloroquine
and statin use’ by van den Hoogen et al. Ann Rheum Dis. 2016
43. Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, Illei GG, Drappa J, Wang L,
Yoo S, et al. Anifrolumab, an Anti-Interferon-alpha Receptor Monoclonal Antibody, in Moderate-
to-Severe Systemic Lupus Erythematosus. Arthritis Rheumatol. 2017; 69:376–386. [PubMed:
28130918]
44. Knight JS, Kaplan MJ. Cardiovascular disease in lupus: insights and updates. Curr Opin
Rheumatol. 2013; 25:597–605. [PubMed: 23846339]
45. Denas G, Jose SP, Bracco A, Zoppellaro G, Pengo V. Antiphospholipid syndrome and the heart: a
case series and literature review. Autoimmun Rev. 2015; 14:214–222. [PubMed: 25461836]
46. Kazzaz NM, Wilson AM, Kado R, Barnes GD, Knight JS. A 37-year-old man with primary
antiphospholipid syndrome presenting with respiratory distress and worsening toe ischemia.
Arthritis Care Res (Hoboken). 2016
47**. Benagiano M, Gerosa M, Romagnoli J, Mahler M, Borghi MO, Grassi A, Della Bella C, Emmi
G, Amedei A, Silvestri E, et al. beta2 Glycoprotein I Recognition Drives Th1 Inflammation in
Atherosclerotic Plaques of Patients with Primary Antiphospholipid Syndrome. J Immunol. 2017
This interesting study examines the inflammatory milieu of atherosclerotic lesions in APS, with a
special focus on TH1 cell-mediated inflammation and interferon signaling.
48. Grayson PC, Kaplan MJ. At the Bench: Neutrophil extracellular traps (NETs) highlight novel
aspects of innate immune system involvement in autoimmune diseases. J Leukoc Biol. 2016;
99:253–264. [PubMed: 26432901]
49. Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein
thrombosis. Arterioscler Thromb Vasc Biol. 2012; 32:1777–1783. [PubMed: 22652600]
50. Rao AN, Kazzaz NM, Knight JS. Do neutrophil extracellular traps contribute to the heightened risk
of thrombosis in inflammatory diseases? World J Cardiol. 2015; 7:829–842. [PubMed: 26730289]
51. Leffler J, Stojanovich L, Shoenfeld Y, Bogdanovic G, Hesselstrand R, Blom AM. Degradation of
neutrophil extracellular traps is decreased in patients with antiphospholipid syndrome. Clin Exp
Rheumatol. 2014; 32:66–70. [PubMed: 24295292]
52**. Yalavarthi S, Gould TJ, Rao AN, Mazza LF, Morris AE, Nunez-Alvarez C, Hernandez-Ramirez
D, Bockenstedt PL, Liaw PC, Cabral AR, et al. Release of Neutrophil Extracellular Traps by
Neutrophils Stimulated With Antiphospholipid Antibodies: A Newly Identified Mechanism of
Thrombosis in the Antiphospholipid Syndrome. Arthritis Rheumatol. 2015; 67:2990–3003. This
study demonstrates the ability of aPL to trigger NET release
in vitro
, through a pathway that
involves TLR4 and reactive oxygen species. Furthermore, these aPL-triggered NETs promote
thrombin generation. [PubMed: 26097119]
53. van den Hoogen LL, Fritsch-Stork RD, van Roon JA, Radstake TR. Low-Density Granulocytes Are
Increased in Antiphospholipid Syndrome and Are Associated With Anti-beta2 -Glycoprotein I
Antibodies: Comment on the Article by Yalavarthi et al. Arthritis Rheumatol. 2016; 68:1320–
1321. [PubMed: 26749538]
54. Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W,
Olsen SH, Klinker M, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and
expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011;
187:538–552. [PubMed: 21613614]
55. Yalavarthi S, Knight JS. Reply. Arthritis Rheumatol. 2016; 68:1321–1322. [PubMed: 26748667]
56*. Meng H, Yalavarthi S, Kanthi Y, Mazza LF, Elfline MA, Luke CE, Pinsky DJ, Henke PK, Knight
JS. In vivo role of neutrophil extracellular traps in antiphospholipid antibody-mediated venous
thrombosis. Arthritis Rheumatol. 2016 This work demonstrates the potential
in vivo
relevance of
NETs in APS. NETs are present in exaggerated fashion in APS thrombi, while thrombosis is
prevented by NET-disrupting treatments.
57. Singel KL, Segal BH. NOX2-dependent regulation of inflammation. Clin Sci (Lond). 2016;
130:479–490. [PubMed: 26888560]
Vreede et al. Page 11
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
58. Salmon JE, Girardi G. Antiphospholipid antibodies and pregnancy loss: a disorder of
inflammation. J Reprod Immunol. 2008; 77:51–56. [PubMed: 17418423]
59. Pierangeli SS, Girardi G, Vega-Ostertag M, Liu X, Espinola RG, Salmon J. Requirement of
activation of complement C3 and C5 for antiphospholipid antibody-mediated thrombophilia.
Arthritis Rheum. 2005; 52:2120–2124. [PubMed: 15986360]
60. Devreese KM, Hoylaerts MF. Is there an association between complement activation and
antiphospholipid antibody-related thrombosis? Thromb Haemost. 2010; 104:1279–1281.
[PubMed: 20838748]
61. Sarmiento E, Dale J, Arraya M, Gallego A, Lanio N, Navarro J, Carbone J. CD8+DR+ T-Cells and
C3 Complement Serum Concentration as Potential Biomarkers in Thrombotic Antiphospholipid
Syndrome. Autoimmune Dis. 2014; 2014:868652. [PubMed: 24982803]
62. Davis WD, Brey RL. Antiphospholipid antibodies and complement activation in patients with
cerebral ischemia. Clin Exp Rheumatol. 1992; 10:455–460. [PubMed: 1458697]
63. Meroni PL, Macor P, Durigutto P, De Maso L, Gerosa M, Ferraresso M, Borghi MO, Mollnes TE,
Tedesco F. Complement activation in antiphospholipid syndrome and its inhibition to prevent
rethrombosis after arterial surgery. Blood. 2016; 127:365–367. [PubMed: 26644452]
64. Stojan G, Petri M. Anti-C1q in systemic lupus erythematosus. Lupus. 2016; 25:873–877.
[PubMed: 27252264]
65. Oku K, Amengual O, Hisada R, Ohmura K, Nakagawa I, Watanabe T, Bohgaki T, Horita T, Yasuda
S, Atsumi T. Autoantibodies against a complement component 1 q subcomponent contribute to
complement activation and recurrent thrombosis/pregnancy morbidity in anti-phospholipid
syndrome. Rheumatology (Oxford). 2016; 55:1403–1411. [PubMed: 27084309]
66. Dufrost V, Risse J, Zuily S, Wahl D. Direct Oral Anticoagulants Use in Antiphospholipid
Syndrome: Are These Drugs an Effective and Safe Alternative to Warfarin? A Systematic Review
of the Literature. Curr Rheumatol Rep. 2016; 18:74. [PubMed: 27812956]
67. Girardi G. Can statins prevent pregnancy complications? J Reprod Immunol. 2014; 101–102:161–
167.
68. Erkan D, Willis R, Murthy VL, Basra G, Vega J, Ruiz-Limon P, Carrera AL, Papalardo E,
Martinez-Martinez LA, Gonzalez EB, et al. A prospective open-label pilot study of fluvastatin on
proinflammatory and prothrombotic biomarkers in antiphospholipid antibody positive patients.
Ann Rheum Dis. 2014; 73:1176–1180. [PubMed: 23933625]
69. Schreiber K, Hunt BJ. Pregnancy and Antiphospholipid Syndrome. Semin Thromb Hemost. 2016;
42:780–788. [PubMed: 27652601]
70. Meroni PL. Prevention & treatment of obstetrical complications in APS: Is hydroxychloroquine the
Holy Grail we are looking for? J Autoimmun. 2016; 75:1–5. [PubMed: 27496152]
71. Stone S, Pijnenborg R, Vercruysse L, Poston R, Khamashta MA, Hunt BJ, Poston L. The placental
bed in pregnancies complicated by primary antiphospholipid syndrome. Placenta. 2006; 27:457–
467. [PubMed: 16005063]
72. Shamonki JM, Salmon JE, Hyjek E, Baergen RN. Excessive complement activation is associated
with placental injury in patients with antiphospholipid antibodies. Am J Obstet Gynecol. 2007;
196:167e161–165. [PubMed: 17306667]
73. Marder W, Knight JS, Kaplan MJ, Somers EC, Zhang X, O’Dell AA, Padmanabhan V, Lieberman
RW. Placental histology and neutrophil extracellular traps in lupus and pre-eclampsia pregnancies.
Lupus Sci Med. 2016; 3:e000134. [PubMed: 27158525]
74. Jung H, Bobba R, Su J, Shariati-Sarabi Z, Gladman DD, Urowitz M, Lou W, Fortin PR. The
protective effect of antimalarial drugs on thrombovascular events in systemic lupus erythematosus.
Arthritis Rheum. 2010; 62:863–868. [PubMed: 20131232]
75. Tektonidou MG, Laskari K, Panagiotakos DB, Moutsopoulos HM. Risk factors for thrombosis and
primary thrombosis prevention in patients with systemic lupus erythematosus with or without
antiphospholipid antibodies. Arthritis Rheum. 2009; 61:29–36. [PubMed: 19116963]
76. Kaplan YC, Ozsarfati J, Nickel C, Koren G. Reproductive outcomes following hydroxychloroquine
use for autoimmune diseases: a systematic review and meta-analysis. Br J Clin Pharmacol. 2016;
81:835–848. [PubMed: 26700396]
Vreede et al. Page 12
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Key points
1. Current standard-of-care therapy for APS does not explicitly target
inflammatory aspects of APS pathophysiology.
2. A better understanding of inter- and intra-cellular signaling pathways in APS
has revealed potential drug targets (i.e., interferons, phosphoinositide 3-
kinase, etc.).
3. In addition to the well-established cellular mediators of APS pathogenesis
(endothelial cells, platelets, etc.), there is emerging interest in the contribution
of myeloid-lineage cells to APS pathogenesis. The role of neutrophil
extracellular trap release, in particular, warrants further study.
4. Complement activation and deposition continue to be recognized for their role
in APS pathogenesis. Activity of this pathway may be mitigated by several
medications including rivaroxaban and hydroxychloroquine.
5. Adjuvant therapeutics including statins and hydroxychloroquine have the
potential to improve APS pregnancy outcomes, based upon animal studies
and small clinical trials.
Vreede et al. Page 13
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 1.
Recent mechanistic insights into the pathophysiology of antiphospholipid antibodies (aPL)
and APS. Starting at the bottom of the figure and moving roughly clockwise: In the vessel
wall of atherosclerotic plaques, beta-2 glycoprotein I (β2GPI)-specific TH1 cells trigger cell
death and release interferons (IFNs). Endothelial cells (ECs) release vesicles (like
microparticles) that activate TLR7 in other ECs by delivery of single-stranded RNA. aPL-
mediated platelet activation relies on phosphoinositide 3-kinase (PI3K). Type I IFNs reduce
the function of restorative circulating endothelial progenitors, which may lead to the accrual
of endothelial damage over time. Cofactor-independent aPL activate monocytes via
endosomal reactive oxygen species (ROS), resulting in increased expression of tissue factor
(TF). In response to aPL, neutrophils release neutrophil extracellular traps (NETs), which
help facilitate thrombin activation. Complement activation, especially through the classical
pathway, leads to the assembly of the membrane attack complex (MAC) on the endothelial
surface, while also facilitating the recruitment and activation of inflammatory cells.
Vreede et al. Page 14
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Vreede et al. Page 15
Table 1
Classification Criteria for Antiphospholipid Syndrome [1]
APS is present if 1 of the clinical criteria and 1 of the laboratory criteria are met
Clinical criteria 1. Vascular thrombosis ≥1 clinical episode of arterial, venous, or small-vessel thrombosis.
2. Pregnancy morbidity a. ≥1 unexplained death of a morphologically normal fetus at ≥10 weeks of
gestation
b. ≥1 premature delivery of a morphologically normal fetus at <34 weeks
gestation because of:
i. Severe pre-eclampsia or eclampsia defined according to standard
definition
ii. Recognized features of placental insufficiency
c. ≥3 unexplained consecutive miscarriages at <10 weeks gestation, with
maternal and paternal factors (anatomic, hormonal or chromosomal
abnormalities) excluded
Laboratory criteria The presence of antiphospholipid antibodies on ≥2 occasions ≥12 weeks apart:
a. Presence of lupus anticoagulant in plasma
b. Medium- to high-titer anticardiolipin antibodies of IgG or IgM isoforms
c. Medium- to high-titer anti-beta-2 glycoprotein-I (anti-β2GPI) antibodies of IgG or IgM isoforms
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Vreede et al. Page 16
Table 2
Summary of efficacy and mechanisms by which adjuvant therapeutics could potentially benefit APS patients
Hydroxychloroquine Statins Rivaroxaban
Summary of efficacy:
Thrombotic risk
Mouse models Protects [9,10] Protects [11]
APS patients No prospective studies in APS, but
protects in post-operative setting [12]
No studies in APS, but
protects in the general
population [13]
Efficacy may be similar to
warfarin (although further study
is needed) [14]
Obstetric events
Mouse models Prevents fetal death and metabolic
changes [15] Prevents fetal death [16]
APS patients May prevent pregnancy loss [7,17] May prevent fetal
morbidity and mortality
[18]
Potential anti-inflammatory mechanisms:
Complement Inhibits activation and deposition [15] Decreases activation [19]
Type I IFN signature Decreases [20] Decreases [20]
NET release Possibly inhibits [21]
Curr Opin Rheumatol
. Author manuscript; available in PMC 2018 September 01.
... Antiphospholipid syndrome (APLS), a complex autoimmune disorder characterized by recurrent thrombosis and obstetric morbidity, presents a significant challenge for clinicians and patients due to its multifaceted nature [1]. APLS and systemic lupus erythematosus (SLE) are rare autoimmune disorders that affect a small fraction of the global female population, impacting fewer than 5% [2]. ...
Article
Full-text available
Antiphospholipid syndrome (APLS) and systemic lupus erythematosus (SLE) are rare autoimmune disorders that afflict a small percentage of the global female population. The complexity of these conditions is further exacerbated by their propensity to give rise to recurrent thrombosis and obstetric morbidity, thereby posing intricate challenges for clinicians and patients alike. One of the most concerning aspects of these diseases is the heightened risk they confer for accelerated atherosclerosis, which can ultimately culminate in the development of acute coronary syndrome (ACS). This case report describes a 27-year-old female with APLS, SLE, and lupus nephritis. She suffered from a catastrophic antiphospholipid syndrome (CAPS) episode and simultaneously developed ACS. She also had a patent foramen ovale (PFO) shunt.
... APS is an important cause of acquired thromboembolic complications in any vasculature: arterial, venous, or small vessels, and it has come to be subcategorized into primary, secondary, and catastrophic APS syndrome. Primary is utilized when there is no associated disorder, secondary is with an associated autoimmune disorder such as systemic lupus erythematosus, and catastrophic when thrombosis occurs at multiple sites in a short space of time [1][2][3]. Once APS is diagnosed, long-term treatment with anticoagulant drugs is considered because the risk of recurrent thrombosis is high [4]. Since the clinical symptoms of thrombosis and pregnancy morbidity in APS are non-specific, accurate detection of aPL is critical for an effective diagnosis and patient management. ...
Article
Full-text available
Antiphospholipid syndrome (APS) is defined as an autoimmune and prothrombotic disorder in patients with the persistent presence of antiphospholipid antibodies (aPLs). In the classification criteria, aPL expresses lupus anticoagulant (LA) activity, which is detected by prolongation of coagulation assays. The LA detection algorithm is a sequential flow including screening tests, mixing tests, and confirmatory tests to differentiate between LA-positive and other anticoagulant abnormalities. Two types of assays are used, like dilute Russell’s viper venom time (dRVVT) and activated partial thromboplastin time (APTT) because no single test is sensitive to all LAs. The anticoagulant drugs prescribed for the prevention and treatment of thrombosis disorders can interfere with the assays, and it is important to know the effects of these drugs in the assays. Especially, new generation anticoagulant drugs, called direct oral anticoagulants (DOACs), affect the results. In this review, the following points are discussed: i) LA detection flow and data interpretation, ii) the principles of coagulation assays proposed and their characteristics, and iii) the effects of anticoagulant drugs in LA detection.
... Positivity for polyreactive aCL antibodies is a criterion in the classification of two autoimmune conditions: APS and SLE 251,252 . aCL antibody titres in these diseases are associated with thrombosis and pregnancy complications 253 . AMA-M5 are complement-fixing autoantibodies that target unknown mitochondrial antigens. ...
Article
The mitochondrion is an intracellular organelle thought to originate from endosymbiosis between an ancestral eukaryotic cell and an α-proteobacterium. Mitochondria are the powerhouses of the cell, and can control several important processes within the cell, such as cell death. Conversely, dysregulation of mitochondria possibly contributes to the pathophysiology of several autoimmune diseases. Defects in mitochondria can be caused by mutations in the mitochondrial genome or by chronic exposure to pro-inflammatory cytokines, including type I interferons. Following the release of intact mitochondria or mitochondrial components into the cytosol or the extracellular space, the bacteria-like molecular motifs of mitochondria can elicit pro-inflammatory responses by the innate immune system. Moreover, antibodies can target mitochondria in autoimmune diseases, suggesting an interplay between the adaptive immune system and mitochondria. In this Review, we discuss the roles of mitochondria in rheumatic diseases such as systemic lupus erythematosus, antiphospholipid syndrome and rheumatoid arthritis. An understanding of the different contributions of mitochondria to distinct rheumatic diseases or manifestations could permit the development of novel therapeutic strategies and the use of mitochondria-derived biomarkers to inform pathogenesis.
... Antiphospholipid antibodies (aPL) are autoantibodies that bind to phospholipid-binding proteins and can provoke tissue pathology [1][2][3][4][5]. The diagnosis of antiphospholipid syndrome (APS) is reserved for patients who develop vascular thrombosis in association with aPL or a lupus anticoagulant (LAC), which screens indirectly for the presence of aPL. ...
Article
Full-text available
Background Non-criteria antiphospholipid antibodies (NC-aPL) are a relatively undefined subgroup of antiphospholipid antibodies (aPL). Knowledge about NC-aPL in adults is limited and even less is known in pediatric patients. Routine tests for antiphospholipid syndrome (APS)—a clinical state marked by the presence of aPL in association with vascular thrombosis—usually include lupus anticoagulant (LAC), anti-cardiolipin (aCL) and -beta-2 glycoprotein I (aβ2GPI). LAC is a functional screen for prothrombotic aPL, while the latter tests identify specific autoantibodies. Specific targets of NC-aPL include, but are not limited to, phosphatidylethanolamine, phosphatidylserine, and prothrombin. Presentation of cases We present single-center data from eight pediatric patients with NC-aPL identified during a three-year period. All patients had presenting features raising suspicion for APS. Most patients were female with a primary rheumatic disease. One patient had a stroke. Another patient had alveolar hemorrhage and pulmonary hypertension. Raynaud’s phenomenon, rashes involving distal extremities, and headaches were common. Most patients had a positive LAC, yet their routine aPL tests were negative, prompting testing for NC-aPL. Conclusions Our findings suggest NC-aPL are associated with typical signs and symptoms of APS in pediatric patients. Pediatricians and pediatric subspecialists should consider NC-aPL when clinical suspicion is high and routine aPL tests are negative, particularly when LAC is positive. While guidelines for NC-aPL do not yet exist for children or adults, these autoantibodies have pathogenic potential. Actionable items could include evaluation for the presence of other (primary) rheumatic diseases, and consultation with hematologists and/or obstetricians regarding anticoagulation/platelet inhibition and thrombosis education. Future guidelines regarding NC-aPL will only be generated by gathering more data, ideally prospectively.
... From the clinical perspective, the heart valve disease was progressive despite anticoagulation [18]. Clinicians should be alert to the underlying SLE in patients with those non-criteria manifestations, for whom anticoagulants alone may offer insufficient protection [19] and for those with a severe condition immunosuppressive therapy besides anticoagulation may be necessary. Further search is needed to investigate whether non-criteria manifestations can predict future SLE in aPL-positive patients, while it is certain that non-criteria manifestations should be taken into account in the APS assessment [20]. ...
Article
Full-text available
Background Antiphospholipid syndrome (APS) is an autoimmune disease characterized by persistent antiphospholipid antibodies (aPLs) positivity with a wide manifestation spectrum. A risk stratification is needed for management guidance and prognosis assessment. We aimed to identify phenotypes among aPL-positive patients and assess the prognosis of each phenotype. Methods This was a single-center, prospective cohort study of aPL-positive patients presented to Peking Union Medical College Hospital from 2012 to 2020. Demographic characteristics, aPL-related manifestations, cardiovascular risk factors, and antibodies profiles were recorded. The primary endpoint was defined as a combination of newly onset thrombosis, major bleeding events, non-criteria manifestations, and all-cause death. Hierarchical cluster analysis and Kaplan-Meier survival analysis were performed. Results Four clusters among 383 patients (70.2% female; mean age 37.7 years) were identified. Cluster 1 (n = 138): patients with systemic lupus erythematosus (SLE) and non-criteria manifestations; cluster 2 (n = 112): patients with multiple cardiovascular risk factors; cluster 3 (n = 83): female patients with obstetric morbidity; cluster 4 (n = 50): patients with isolated lupus anticoagulant (LA) positivity. Non-criteria manifestations were found aggregated with SLE from cluster analysis of variables. Cluster 3 showed the best outcome, while cluster 2 suffered highest frenquency of newly onset arterial thrombosis. Conclusions We identified 4 clinical phenotypes of aPL-positive patients. Non-criteria manifestations may indicate underlying SLE, for which immunosuppressive therapy besides anticoagulation may be necessary. Patients with isolated LA positivity suffered similar risks with secondary APS and patients with multiple cardiovascular risk factors. Attention should be paid to male patients, and the screening of cardiovascular risk factors should never be ignored.
... While various proteins involved in the mitochondrial processing of pyruvate (e.g., periosteum-derived cell E2), sulfite oxidase, and glycogen phosphorylase are mitochondrial proteins known to be targeted by AMAs (subtypes M2, M4, and M9, respectively) in PBC (11), limited knowledge is available concerning the extent of the mitochondrial proteome targeted by AMAs in SLE. Antibodies to cardiolipin, a phospholipid uniquely synthetized in the mitochondria of humans, are known to be associated with vascular and obstetric events in SLE and APS (16,34,35). To date, the only mitochondrial protein with autoantibodies associated with disease manifestations in SLE is HSP60 (17). ...
Article
Full-text available
Objective Mitochondria are organelles that exhibit several bacterial features, such as a double‐stranded genome with hypomethylated CpG islands, formylated proteins, and cardiolipin‐containing membranes. In systemic lupus erythematosus (SLE), mitochondria and their inner components are released into the extracellular space, potentially eliciting a proinflammatory response from the immune system. While cardiolipin and mitochondrial DNA and RNA are confirmed targets of autoantibodies, other antigenic mitochondrial proteins in SLE remain to be identified. The present study was undertaken to characterize the protein repertoire recognized by antimitochondrial antibodies (AMAs) in patients with SLE. Methods Using shotgun proteomic profiling, we identified 1,345 proteins, 431 of which were associated with the mitochondrial proteome. Immunoreactivities to several of these candidate proteins were assessed in serum samples from a local cohort (n = 30 healthy donors and 87 patients with SLE) using enzyme‐linked immunosorbent assay, and further analyzed for associations with demographic and disease characteristics. Results We determined that IgG antibodies to the complement component C1q binding protein were significantly elevated in the patients with SLE (P = 0.049) and were also associated with lupus anticoagulant positivity (P = 0.049). Elevated levels of IgG antibodies against mitochondrial protein mitofusin 1 (MFN‐1) were promising predictors of SLE diagnosis in our cohort (adjusted odds ratio 2.99 [95% confidence interval 1.39–6.43], P = 0.0044). Moreover, increased levels of anti–MFN‐1 were associated with the presence of antiphospholipids (P = 0.011) and anti–double‐stranded DNA (P = 0.0005). Conclusion In this study, we characterized the mitochondrial repertoire targeted by AMAs in the setting of SLE. Our results indicate that autoantibodies can recognize secreted and/or surface proteins of mitochondrial origin. Profiling of the AMA repertoire in large prospective cohorts may improve our knowledge of mitochondrial biomarkers and their usefulness for patient stratification.
... However, recent animal model and human studies indicate a greater role for complement activation, endothelial progenitor cell deficiency, and activation of neutrophils with release of neutrophil extracellular traps by APLA, suggesting that APS is more an autoimmune thrombo-inflammatory disorder rather than simply a disorder of coagulation. This has opened up a plethora of potential new therapeutic targets as diseasemodifying therapy in severe or anticoagulant refractory APS (7)(8)(9). ...
... From the clinical perspective, the heart valve disease was progressive despite anticoagulation [18]. Clinicians should be alert to the underlying SLE in patients with those non-criteria manifestations, for whom anticoagulants alone may offer insu cient protection [19] and for those with a severe condition immunosuppressive therapy besides anticoagulation may be necessary. Further search is needed to investigate whether non-criteria manifestations can predict future SLE in aPL-positive patients, while it is certain that non-criteria manifestations should be taken into account in the APS assessment [20]. ...
Preprint
Full-text available
Background: Antiphospholipid syndrome (APS) is an autoimmune disease characterized by persistent antiphospholipid antibodies (aPLs) positivity with a wide manifestation spectrum. A risk stratification is needed for management guidance and prognosis assessment. We aimed to identify phenotypes among aPL-positive patients and assess the prognosis of each phenotype. Methods: This was a single-center, prospective cohort study of aPL-positive patients presented to Peking Union Medical College Hospital from 2012 to 2020. Demographic characteristics, aPL-related manifestations, cardiovascular risk factors and antibodies profiles were recorded. The primary endpoint was defined as a combination of newly-onset thrombosis, major bleeding events, non-criteria manifestations and all-cause death. Hierarchical cluster analysis and Kaplan-Meier survival analysis were performed. Results: Four clusters among 383 patients (70.2% female; mean age 37.7 years) were identified. Cluster 1 (n=138): patients with systemic lupus erythematosus (SLE) and non-criteria manifestations; Cluster 2 (n=112): patients with multiple cardiovascular risk factors; Cluster 3 (n=83): female patients with obstetric morbidity; Cluster 4 (n=50): patients with isolated lupus anticoagulant (LA) positivity. Non-criteria manifestations were found aggregated with SLE from cluster analysis of variables. Cluster 3 showed the best outcome, while cluster 2 suffered highest frenquency of newly-onset arterial thrombosis. Conclusions: We identified 4 clinical phenotypes of aPL-positive patients. Non-criteria manifestations may indicate underlying SLE, for which immunosuppressive therapy besides anticoagulation may be necessary. Patients with isolated LA positivity suffered similar risks with secondary APS and patients with multiple cardiovascular risk factors. Attention should be paid to male patients, and the screening of cardiovascular risk factors should never be ignored.
Preprint
Full-text available
Bis(monoacylglycero)phosphate (BMP) is a major phospholipid constituent of intralumenal membranes in late endosomes/lysosomes, where it regulates the degradation and sorting of lipid cargo. Recent observations suggest that the Batten disease - associated protein CLN5 functions as lysosomal BMP synthase. Here, we show that transacylation reactions catalyzed by cytosolic and secreted enzymes enhance BMP synthesis independently of CLN5. The transacylases identified in this study are capable of acylating the precursor lipid phosphatidylglycerol (PG), generating acyl-PG, which is subsequently hydrolyzed to BMP. Extracellularly, acyl-PG and BMP are generated by endothelial lipase in cooperation with other serum enzymes of the pancreatic lipase family. The intracellular acylation of PG is catalyzed by several members of the cytosolic phospholipase A2 group IV (PLA2G4) family. Overexpression of secreted or cytosolic transacylases was sufficient to correct BMP deficiency in HEK293 cells lacking CLN5. Collectively, our observations suggest that functionally overlapping pathways promote BMP synthesis in mammalian cells.
Article
Background: Thrombotic antiphospholipid syndrome (t-APS) is characterized by arterial, venous or microvascular occlusions, which are explained, in part, by the presence of antiphospholipid antibodies (aPL). Although there are several evidences indicating that isolated aPL antibodies increase the activity of platelets obtained from healthy volunteers, platelet function in t-PAPS has not been as widely studied. Objective: To evaluate platelet reactivity in t-PAPS patients. Methods: Platelet aggregation, protein expression and cyclic nucleotide levels were carried out in platelet rich plasma (PRP) or washed platelets (WP) obtained from t-PAPS or healthy volunteers. Results: ADP-induced aggregation was significantly higher in PRP obtained from t-PAPS than obtained from the control. The protein expression of P2Y12 receptor and Gs alpha was significantly higher and lower, respectively in WP from t-PAPS patients. In PRP incubated with iloprost or sodium nitroprusside, the residual platelet reactivity induced by ADP was still higher in PRP from t-PAPS than from the control. Lower intracellular levels of cAMP and cGMP were observed in unstimulated PRP from t-PAPS patients. The protein expression of soluble guanylate cyclase subunits and phosphodiesterases type 3 and 5 did not differ. The antiplatelet activity of ticagrelor was similar between the groups and cilostazol significantly potentiated this response. Isolated aPL antibodies obtained from t-PAPS patients potentiated ADP-induced aggregation in healthy platelets, but did not affect the inhibitory responses induced by iloprost or SNP. Conclusions: The overexpression of P2Y12 receptor, accompanied by lower levels of cAMP and cGMP levels produced greater amplitude of ADP aggregation in platelets from t-PAPS patients.
Article
Full-text available
Antiphospholipid syndrome (APS) is characterized by recurrent arterial/venous thrombosis and miscarriages in the persistent presence of autoantibodies against phospholipid-binding proteins (aPLs), such as β2 glycoprotein I (β2GPI). In addition to the aPL thrombophilic effect, arterial thrombosis was related to accelerated atherosclerosis in animal models; however, contrasting findings were reported in primary APS patients with regard to the increased number of plaques or abnormal arterial wall thickness. We investigated the cytokine production induced by β2GPI in activated T cells that infiltrate in vivo atherosclerotic lesions of primary APS patients with atherothrombosis. We also examined the helper function of β2GPI-specific T cells for monocyte matrix metalloproteinase-9 and tissue factor production, as well as their cytolytic potential and their helper function for Ab production. APS patients with atherothrombosis harbor in vivo-activated CD4(+) T cells that recognize β2GPI in atherothrombotic lesions. β2GPI induces T cell proliferation and IFN-γ expression in plaque-derived T cell clones. β2GPI-specific T cells display helper function for monocyte matrix metalloproteinase-9 and tissue factor production and promote Ig production in autologous B cells. Moreover, plaque-derived β2GPI-specific CD4(+) T lymphocytes express perforin-mediated and Fas/Fas ligand-mediated cytotoxicity. β2GPI, and especially the DI domain, drive a local Th1 inflammatory response, with subsequent plaque instability that eventually favors atherothrombosis. This finding may explain the association between aPLs and arterial thrombosis, despite the lack of evidence of surrogate markers for atherosclerosis in primary APS.
Article
Full-text available
Objective To assess the efficacy and safety of anifrolumab, a type I interferon (IFN) receptor antagonist, in a phase IIb, randomized, double‐blind, placebo‐controlled study of adults with moderate‐to‐severe systemic lupus erythematosus (SLE). Methods Patients (n = 305) were randomized to receive intravenous anifrolumab (300 mg or 1,000 mg) or placebo, in addition to standard therapy, every 4 weeks for 48 weeks. Randomization was stratified by SLE Disease Activity Index 2000 score (<10 or ≥10), oral corticosteroid dosage (<10 or ≥10 mg/day), and type I IFN gene signature test status (high or low) based on a 4‐gene expression assay. The primary end point was the percentage of patients achieving an SLE Responder Index (SRI[4]) response at week 24 with sustained reduction of oral corticosteroids (<10 mg/day and less than or equal to the dose at week 1 from week 12 through 24). Other end points (including SRI[4], British Isles Lupus Assessment Group [BILAG]–based Composite Lupus Assessment [BICLA], modified SRI[6], and major clinical response) were assessed at week 52. The primary end point was analyzed in the modified intent‐to‐treat (ITT) population and type I IFN–high subpopulation. The study result was considered positive if the primary end point was met in either of the 2 study populations. The Type I error rate was controlled at 0.10 (2‐sided), within each of the 2 study populations for the primary end point analysis. Results The primary end point was met by more patients treated with anifrolumab (34.3% of 99 for 300 mg and 28.8% of 104 for 1,000 mg) than placebo (17.6% of 102) (P = 0.014 for 300 mg and P = 0.063 for 1,000 mg, versus placebo), with greater effect size in patients with a high IFN signature at baseline (13.2% in placebo‐treated patients versus 36.0% [P = 0.004] and 28.2% [P = 0.029]) in patients treated with anifrolumab 300 mg and 1,000 mg, respectively. At week 52, patients treated with anifrolumab achieved greater responses in SRI(4) (40.2% versus 62.6% [P < 0.001] and 53.8% [P = 0.043] with placebo, anifrolumab 300 mg, and anifrolumab 1,000 mg, respectively), BICLA (25.7% versus 53.5% [P < 0.001] and 41.2% [P = 0.018], respectively), modified SRI(6) (28.4% versus 49.5% [P = 0.002] and 44.7% [P = 0.015], respectively), major clinical response (BILAG 2004 C or better in all organ domains from week 24 through week 52) (6.9% versus 19.2% [P = 0.012] and 17.3% [P = 0.025], respectively), and several other global and organ‐specific end points. Herpes zoster was more frequent in the anifrolumab‐treated patients (2.0% with placebo treatment versus 5.1% and 9.5% with anifrolumab 300 mg and 1,000 mg, respectively), as were cases reported as influenza (2.0% versus 6.1% and 7.6%, respectively), in the anifrolumab treatment groups. Incidence of serious adverse events was similar between groups (18.8% versus 16.2% and 17.1%, respectively). Conclusion Anifrolumab substantially reduced disease activity compared with placebo across multiple clinical end points in the patients with moderate‐to‐severe SLE.
Article
Full-text available
Background The cornerstone of thrombotic antiphospholipid syndrome (APS) patients’ management is to prevent recurrent thrombosis by long-term anticoagulation. Purpose of reviewThe purpose of the review is to summarize available literature on direct oral anticoagulants (DOACs) use in APS patients through a systematic review and to determine factors associated with thrombosis recurrence. Recent findingsThe recent RAPS trial demonstrated that APS patients treated with rivaroxaban had a significant twofold-increased thrombin potential, suggesting a higher thrombotic risk, in comparison with warfarin users. Furthermore, several reports of APS patients treated with DOACs have raised safety issues. Our systematic review identified 122 published APS patients treated with DOACs; among them, 19 experienced a recurrent thrombosis while on DOACs. Of note, triple positivity (positivity of all three laboratory criteria for APS) was associated with a 3.5-fold increased risk for recurrent thrombosis. SummaryIn conclusion, DOACs should be used with caution in APS patients and randomized control trials with clinical primary endpoints assessing clinical efficacy and safety are awaited to establish whether the prescription of DOACs could be a safe alternative to warfarin.
Article
Full-text available
Primary antiphospholipid syndrome (APS) is an autoimmune disorder of unknown cause, characterised by not only thrombotic events and pregnancy morbidity but also accelerated atherosclerosis. We reviewed with great interest the letter by van den Hoogen et al ,1 which comments on our recent paper.2 In our study, we discovered a defect in endothelial progenitor function in patients with primary APS—such defects have been described as predecessors to atherosclerosis. We subsequently revealed a type I interferon (IFN) signature in peripheral blood mononuclear cells of the same patients. Importantly, inhibition of the type I IFN receptor restored normal differentiation of endothelial progenitors into endothelial cells in vitro. Further, we detected the IFN signature not only in our local primary APS cohort but also in a cohort of patients from Mexico. van den Hoogen et al 1 have now detected a type I IFN signature in a third cohort of patients with primary APS, this one recruited in the Netherlands. Going further, they reanalysed …
Article
Full-text available
Objective: Antiphospholipid syndrome (APS) is a leading acquired cause of thrombotic events. Although antiphospholipid antibodies have been shown to promote thrombosis in mice, the role of neutrophils has not been explicitly studied. The aim of this study was to characterize neutrophils in the context of a new model of antiphospholipid antibody-mediated venous thrombosis. Methods: Mice were administered fractions of IgG obtained from patients with APS. At the same time, blood flow through the inferior vena cava was reduced by induction of stenosis. Resulting thrombi were characterized for size and neutrophil content. Circulating factors and the vessel wall were also assessed. Results: As measured by both thrombus weight and thrombosis frequency, mice treated with IgG from patients with APS (APS IgG) demonstrated exaggerated thrombosis as compared with control IgG-treated mice. Thrombi in mice treated with APS IgG were enriched for citrullinated histone H3 (a marker of neutrophil extracellular traps [NETs]). APS IgG-treated mice also demonstrated elevated levels of circulating cell-free DNA and human IgG bound to the neutrophil surface. In contrast, circulating neutrophil numbers and markers of vessel wall activation were not appreciably different between APS IgG-treated mice and control mice. Treatment with either DNase (which dissolves NETs) or a neutrophil-depleting antibody reduced thrombosis in APS IgG-treated mice to the level in control mice. Conclusion: These data support a mechanism whereby circulating neutrophils are primed by antiphospholipid antibodies to accelerate thrombosis. This line of investigation suggests new, immunomodulatory approaches for the treatment of APS.
Article
It is long known that antiphospholipid antibodies (aPL) induce proinflammatory and procoagulant cellular responses. The underlying signal transduction has been a major focus of research and is the topic of this review. An amazingly heterogeneous panel of signaling pathways has been described and it turns out that at least some of this heterogeneity can be explained by effects of distinct aPL species. On the one hand, there are antibodies against β2-glycoprotein I (β2GPI) which appear to exert their cellular effects only as a complex of β2GPI/anti-β2GPI. Their major targets are low-density lipoprotein-receptor related protein 8 (LRP8), annexin A2 (ANXA2), toll-like receptor 4 (TLR4), and possibly TLR2. The other relevant aPL species are antibodies against cardiolipin which are internalized into endosomes and induce cellular responses via activation of endosomal NADPH-oxidase. Their cell surface target is still unknown. Another important issue relates to the role of complement. It has been shown in vivo that certain pathogenic effects of aPL depend on complement activation, but the exact interplay with the signaling pathways described earlier needs to be elucidated. Thus, while there has been tremendous progress over the past decade, many open questions remain to be answered.
Article
Objectives Hydroxychloroquine (HCQ) has been used for decades to treat patients with rheumatic diseases, for example, systemic lupus erythematosus (SLE), rheumatoid arthritis or the antiphospholipid syndrome (APS). We hypothesise that HCQ might target endosomal NADPH oxidase (NOX), which is involved in the signal transduction of cytokines as well as antiphospholipid antibodies (aPL). Methods For in vitro experiments, monocytic cells were stimulated with tumour necrosis factor α (TNFα), interleukin-1β (IL-1β) or a human monoclonal aPL and the activity of NOX was determined by flow cytometry. The expression of genes known to be induced by these stimuli was quantified by quantitative reverse transcription PCR. Live cell imaging was performed by confocal laser scanning microscopy. Finally, the effects of HCQ on NOX-induced signal transduction were analysed in an in vivo model of venous thrombosis. Results HCQ strongly reduces or completely prevents the induction of endosomal NOX by TNFα, IL-1β and aPL in human monocytes and MonoMac1 cells. As a consequence, induction of downstream genes by these stimuli is reduced or abrogated. This effect of HCQ is not mediated by direct interference with the agonists but by inhibiting the translocation of the catalytic subunit of NOX2 (gp91phox) into the endosome. In vivo, HCQ protects mice from aPL-induced and NOX2-mediated thrombus formation. Conclusions We describe here a novel mechanism of action of HCQ, that is, interference with the assembly of endosomal NOX2. Since endosomal NOX2 is involved in many inflammatory and prothrombotic signalling pathways, this activity of HCQ might explain many of its beneficial effects in rheumatic diseases including the APS.
Article
Background Increased activity of the type I interferon (IFN) pathway is central to the pathogenesis of SLE. Blocking the type I receptor may reduce SLE activity more effectively than inhibiting IFN-α alone. Objectives Efficacy and safety of anifrolumab were assessed in a Phase IIb, randomized, double-blind, placebo-controlled study of adults with moderate to severe SLE (the MUSE study). Methods 305 patients were treated for 48 weeks with intravenous anifrolumab (300 mg or 1000 mg) or placebo, in addition to standard-of-care medications. Randomization was stratified by SLE Disease Activity Index 2000 (SLEDAI-2K) score (<10 or ≥10), oral corticosteroid (OCS) dose (<10 or ≥10 mg/day), and IFN gene signature status (high vs. low) based on a 4-gene expression assay. The primary endpoint was the percentage of patients achieving an SLE Responder Index [SRI(4)] response at Day 169 with sustained reduction of OCS (<10 mg/day and ≤Day 1 dose from Day 85 to 169). Results The primary endpoint was met by more anifrolumab-treated patients [placebo vs. 300 mg vs. 1000 mg: 17.6%, 34.3% (p=0.014), 28.8% (p=0.063)] with greater effect sizes observed in the 75% of patients who had a high baseline IFN signature [13.2%, 36.0% (p=0.004), 28.2% (p=0.029)]. At Day 365 more anifrolumab-treated patients achieved SRI(4) responses [40.2%, 62.6%, (p<0.001), 53.8%, (p=0.043)], BILAG-based Composite Lupus Assessment (BICLA) [25.7%, 53.5% (p<0.001), 41.2% (p=0.018)], modified SRI(6) [28.4%, 49.5% (p=0.002), 44.7% (p=0.015)], and SLEDAI-2K ≤2 [17.6%, 35.4% (p=0.004), 32.7% (p=0.012)]. Major clinical response (BILAG “C” or better in all domains at Day 169 maintained to Day 365) was achieved by 6.9%, 19.2% (p=0.012), and 17.3% (p=0.025) of patients. BILAG “A” flares were reported in more placebo- vs. anifrolumab-treated patients (16.7%, 9.1%, 10.6%). In patients with baseline Cutaneous Lupus Erythematosus Disease Area and Severity Index activity score ≥10, more anifrolumab-treated patients attained a ≥50% reduction by Day 365 [30.8%, 63.0% (p=0.013), 58.3% (p=0.077)]. In patients with ≥8 swollen and ≥8 tender joints at baseline more anifrolumab-treated patients achieved ≥50% decrease in joint count [48.6%, 69.6% (p=0.038), 64.6% (p=0.156)]. Although steroid tapering was not mandated, OCS reduction to ≤7.5 mg/d at Day 365 was achieved by 26.6%, 56.4%, and 31.7% of patients. The observed benefits were driven by results in the IFN-high subpopulation (figure). Median suppression of 21 IFN-regulated genes was ∼90% for both doses of anifrolumab. Patients in the placebo group had the lowest incidence of Herpes zoster (2.0%, 5.1%, 9.5%) and cases reported as influenza (2.0%, 6.1%, 7.6%); there were no differences in the incidence of serious adverse events (18.8%, 16.2%, 17.1%). The incidence of infusion-related reactions was similar (5.9%, 2.0%, 3.8%). View larger version: • In a new window • Download as PowerPoint Slide Conclusions Anifrolumab significantly reduced disease activity across all clinical endpoints. Enhanced effects in IFN-high patients support the pathobiology of this treatment strategy. Acknowledgement Funded by MedImmune. Editorial assistance: K Alexander, QXV Comms, an Ashfield business, UK Disclosure of Interest R. Furie Consultant for: MedImmune, J. Merrill Grant/research support from: MedImmune; Genentech/Roche, Consultant for: Medimmune, Genentech/Roche, Neovacs, V. Werth Consultant for: MedImmune, M. Khamashta: None declared, K. Kalunian Grant/research support from: MedImmune, Consultant for: AstraZeneca, P. Brohawn Shareholder of: AstraZeneca, Employee of: MedImmune, G. Illei Shareholder of: AstraZeneca, Employee of: MedImmune, J. Drappa Shareholder of: AstraZeneca, Employee of: MedImmune, L. Wang Employee of: MedImmune, S. Yoo Shareholder of: AstraZeneca; Regenx Bio, Consultant for: Regenx Bio
Article
Several autoimmune diseases, most notably systemic lupus erythematosus (SLE), show an overexpression of type I interferon (IFN)-inducible genes, termed the IFN signature.1 The IFN signature is associated with endothelial progenitor cells (EPC) dysfunction in SLE2 and the recent report by Grenn et al 3 provides evidence for this link in antiphospholipid syndrome (APS) as well. The authors show that patients with primary APS (PAPS) have a reduced number of circulating EPC that differentiate into endothelial cell like cells. This is supported by a similar reduction in EPC differentiation when EPC from healthy controls (HC) are cultured with serum from patients with PAPS. Whereas depletion of antiphospholipid antibodies (aPL) has no effect, blockade of the receptor for type I IFN restores EPC differentiation. Hence, targeting type I IFN might mitigate vascular disease in patients with APS by restoring EPC differentiation. In support of this assumption, the authors demonstrate the presence of an IFN signature in peripheral blood mononuclear cells (PBMC) of patients with PAPS. Previous hints in the literature further confirm the presence of an IFN signature in PBMC of patients with PAPS.4 ,5 Interestingly, a microarray study on isolated monocytes of patients with SLE, SLE+APS and PAPS reported the presence of an IFN signature only in monocytes of patients with SLE and SLE+APS, not in PAPS.6 Monocytes are considered key players in the pathogenesis of APS4 among others due to an overexpression of tissue factor (TF), the main initiator of humoral coagulation, while shifts in monocyte subsets are linked to vascular disease in rheumatic diseases.7 ,8 Here we report the …