You are on page 1of 14

Send Orders for Reprints to reprints@benthamscience.

ae 313
Current Vascular Pharmacology, 2017, 15, 313-326

REVIEW ARTICLE
ISSN: 1570-1611
eISSN: 1875-6212

Current Vascular
Pharmacology
Thrombotic Management of Antiphospholipid Syndrome: The journal for current and in-depth reviews on Vascular Pharmacology

Impact
Factor:
2.374

Towards Novel Targeted Therapies


BENTHAM
SCIENCE

Md. Asiful Islam1,#,*, Fahmida Alam1,#, Kah Keng Wong2, Mohammad Amjad Kamal3,4,5 and Siew Hua Gan1,*

1
Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan,
Malaysia; 2Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian,
Kelantan, Malaysia; 3King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, Saudi
Arabia; 4Enzymoics; 5Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW 2770,
Australia

Abstract: Antiphospholipid syndrome (APS) is a systemic autoimmune disease characterized by throm-


bosis and/or pregnancy morbidity with persistent levels of antiphospholipid antibodies (aPLs). The
development of thrombosis in APS is mediated by aPLs and contributes to the high mortality rate in
APS patients. However, although APS has been reported for more than 30 years, there has been no op-
ARTICLE HISTORY
timal regimen for its prevention or for the management of thrombosis, mainly because the mainstay
Received: May 30, 2016 treatment strategies for managing APS are not targeted towards aPL-mediated thrombotic pathophysiol-
Revised: December 07, 2016
Accepted: December 07, 2016
ogy. Instead, the treatments commonly used are aimed at general thrombotic disorders. Warfarin is the
most commonly used vitamin K antagonist (VKA), in addition to anti-platelet medications, such as aspi-
DOI:
Current Vascular Pharmacology

10.2174/1570161115666170105120931 rin and clopidogrel. Over the last decade, novel non-VKA oral anticoagulants, including rivaroxaban,
apixaban and dabigatran, as well as immunomodulatory agents, such as rituximab, eculizumab, hy-
droxychloroquine, statins and sirolimus, have also been used. In this review, we discuss the current
treatment strategies and future treatment outlook for thrombotic APS.

Keywords: Antiphospholipid syndrome, antiphospholipid antibodies, thrombosis, treatment, anticoagulant, anti-platelet agents.

1. INTRODUCTION thrombotic events in a study published in 1985 [5]. However,


in 2002, Asherson [6] reported that 55% of the APS patients
Antiphospholipid syndrome (APS) is a systemic autoim-
experienced deep vein thrombosis (DVT) and 40% experi-
mune disease characterized by recurrent vascular thrombosis enced pulmonary embolism (PE). According to the Euro-
(venous and/or arterial) and/or pregnancy morbidity, in addi-
Phospholipid project (involving 13 European countries with
tion to the presence of serum antiphospholipid antibodies
1,000 participants), thrombosis was the most frequently
(aPLs) [lupus anticoagulant (LA) antibody, anticardiolipin
(28.1%) observed clinical manifestation [7].
antibody (aCL) and anti-2 glycoprotein I (anti-2GPI) anti-
body] as laboratory features [1]. APS is classified into the To date, there is still no optimal treatment strategy to
following categories: 1) primary APS (at least one clinical combat thrombotic APS. Although the pathogenesis of
and one laboratory feature present), 2) APS associated with thrombi is mainly mediated by aPLs, most of the current
other diseases (for this category, an autoimmune disease, regimens are focused on preventing the recurrence of throm-
usually systemic lupus erythematosus (SLE), should be pre- bosis in such a way that they are more appropriate for gen-
sent in addition to the clinical and laboratory features of eral, non-APS thrombotic patients, rather than targeting dis-
APS), and, 3) catastrophic APS (the most aggressive form, ease-specific pathophysiology [8]. Anticoagulant drugs such
characterized by multiple organ failure) [2-4]. as warfarin and anti-platelet agents, including aspirin or
Thrombosis is one of the major clinical features of APS, clopidogrel, are most commonly used alone in patients with
and 30% of patients were documented to experience thrombotic APS or in combination with other drugs (ster-
oids, other anticoagulants, anti-platelet agents or immuno-
*Address correspondence to these authors at the Human Genome Centre, suppressive agents), based on the characteristics and degree
School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang of thrombosis [9-11]. In addition to the mainstay regimens, a
Kerian, Kelantan, Malaysia; Tel: +6097676797; Fax: +6097658914; substantial number of drugs, such as new non-vitamin K
E-mails: ayoncx70@yahoo.com, shgan@usm.my
#
These authors contributed equally to this work.
antagonist (non-VKA) oral anticoagulants (NOAC) and im-

1875-6212/17 $58.00+.00 © 2017 Bentham Science Publishers


314 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

munomodulatory agents, have been used in the last decade [36, 37] were reported to be the major immune-mediated
for the treatment of thrombosis in APS patients. phenomena contributing to thrombosis in APS patients.
To date, there is a lack of comprehensive reviews ad-
4. TREATMENT STRATEGIES
dressing the updates on all the current thrombotic treatment
strategies used in patients with APS. Therefore, in this re- To date, there is no single drug or recommended dosage
view, treatment strategies for managing thrombotic APS for the ideal management of thrombosis in patients with
patients with vitamin K antagonists (VKAs) as conventional APS. Single medications or combined therapeutic strategies
anticoagulants (warfarin), anti-platelet agents (aspirin and with different intensities and dosages have both been suc-
clopidogrel), novel NOACs (rivaroxaban, apixaban and cessful and unsuccessful for managing thrombosis in APS.
dabigatran) and immunomodulatory agents [rituximab, ecu- In addition to the use of conventional anticoagulants and
lizumab, hydroxychloroquine (HCQ), statins and sirolimus] anti-platelets, novel NOACs (Table 1) and targeted immu-
are discussed, based on both experimental and clinical evi- nomodulatory agents have recently been incorporated in the
dence. management of thrombotic APS (Fig. 1). The experimental
and clinical evidence indicating both the advantages and
2. THROMBOSIS AND APS pitfalls of the current treatment strategies for treating throm-
bosis in APS patients are considered in the subsequent sec-
According to the latest Sydney classification criteria [1],
tions.
thrombosis is one of the two clinical manifestations observed
in patients with APS. Among the other thrombophilic disor-
5. ANTICOAGULANTS
ders, both arterial and venous thromboses are observed in
APS patients, which contribute to the high mortality rate [10, 5.1. Warfarin
12]. A 10-year multicenter prospective study (n=1,000)
revealed that the most frequent causes of death in patients Warfarin (Coumadin®) is the most widely used oral anti-
with APS were severe thrombosis (36.5%) and thrombotic coagulant for the prevention of thrombosis and thromboem-
events (21.4%), including DVT, myocardial infarction (MI), bolism. Following its administration, warfarin is rapidly ab-
transient ischemic attacks (TIAs), stroke, PEs and superficial sorbed from the gastrointestinal (GI) tract and reaches its
thrombophlebitis, thus indicating that there is a strong maximal blood concentrations in healthy adults after 90 min
clinical impact and economic burden worldwide [7, 9]. [38, 39]. The half-lives of warfarin components, which con-
sists of a racemic mixture containing two active stereoi-
A thrombus can develop in any tissue and/or organ in somers (R and S), are 36 and 42 h, respectively. Warfarin
patients with APS, as confirmed by imaging, ultrasound or binds to the plasma proteins for circulation and accumulates
histopathology studies, except in the case of superficial ve- in the liver before being metabolically transformed via the
nous thrombosis [1]. In cases of catastrophic APS, the fre- cytochrome P450 enzymes [40]. Warfarin acts by inhibiting
quently reported cause of death is multiple organ failure due the vitamin K-dependent synthesis of calcium-dependent
to the formation of acute thrombi in the organs, which is blood clotting factors II, VII, IX and X, as well as some
believed to be the major cause of death (55.6%) [7]. regulatory factors, including proteins C, S and Z [41].

3. PATHOGENESIS OF THROMBOSIS IN APS Warfarin therapy is monitored based on the international


normalized ratio (INR). The INR is a mathematical conver-
Although the complete thrombotic pathophysiology of sion of the prothrombin time (PT) that is used to normalize
APS is still unclear, generally, the formation of thrombi in the results, irrespective of the different PT reagents (throm-
APS is mediated by aPL-induced dysregulation of cell acti- boplastins) [42]. Usually, the targeted INR following war-
vation, coagulation, the fibrinolytic pathway, the immune farin administration is between 2.0 and 3.0 [43]. A high INR
system, genetics and some other factors [8]. Activation of value is associated with increased risk of bleeding, whereas
platelet markers, such as CD63, procaspase activating com- INRs below the therapeutic values are indicative of an in-
pound-1 (PAC-1) [13], CD62 [14], and CD62P [15], the adequate warfarin dose to confer protection against throm-
formation of a thrombogenic complex between homo-
boembolism. The risk of bleeding is greatly increased when
tetrameric platelet factor 4 (PF4) and two dimerized mole-
the INR exceeds 4.5 [44].
cules of 2GPI [16], activation and elevation of the von
Willebrand factor (vWF) levels [17, 18], dysfunction of With the increased use of warfarin, the number of cases
ADAMTS13 (a protease enzyme) [19, 20], dysregulation of with bleeding has also increased and has contributed to a
prostaglandins [prostacyclin (PGI2) and thromboxane A 2 significant number of deaths. It was reported that the fre-
(TXA2)] [17, 21, 22], and annexin A2 [23] and toll-like re- quency of major bleeding due to the use of warfarin was as
ceptors (TLRs) [24] are involved in platelet activation, adhe- high as 16%. Therefore, in 2006, a ‘black box’ warning re-
sion and aggregation, leading to the generation of thrombi in garding the bleeding risk caused by warfarin was added to
APS patients. In addition to impaired fibrinolysis [25] in the the United States (US) product labels [45]. The warfarin
coagulation pathway, the involvement of tissue factors (TFs) dosage is complex because of the drug and food interactions
[26], annexin A5 [27], proteins C and S [28, 29], factor XI [46]. Drugs that are largely protein-bound compete with war-
[30], tissue factor pathway inhibitor (TFPI) [31] was also farin for protein binding, contributing to the increased INR
observed in patients with thrombotic APS. The activation of [47]. Since foods containing vitamin K1, such as green leafy
the complement system [32, 33] and the involvement of B vegetables (i.e. lettuce and spinach), are reported to interact
and T cells [34, 35] and tissue necrosis factor alpha (TNF-) with warfarin, it is recommended that its activity be
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 315

Table 1. Characteristics of the new non-vitamin K antagonist oral anticoagulants (NOACs) used for the treatment of thrombosis
in APS patients.

New NOACs Incorporated in the Treatment of Thrombotic APS


Characteristics Rivaroxaban Apixaban Dabigatran
(Xarelto®) (Eliquis ®) (Pradaxa®)

Bristol-Myers Squibb Company Boehringer Ingelheim


Manufacturer Janssen Pharmaceuticals Inc.
and Pfizer Inc. Pharmaceuticals Inc.

First FDA approval 2011 2012 2010

Yes
Any ongoing clinical trials with APS? Yes
(NCT02116036; No
(ClinicalTrials.govIdentifier) (NCT02295475)
NCT02157272)

Yes
Is this a prodrug? No No
(Dabigatran etexilate)

Required routine INR monitoring? No No No

Target Factor Xa Factor Xa Thrombin

Approved doses Fixed, once daily Fixed, once daily Fixed, twice daily
(mg) 10, 15, and 20 2.5 and 5 75 and 150

Molecular weight (g/mol) 435.9 459.5 627.8

10 mg dose: 80-100
Bioavailability (%) 50 3-7
20 mg dose: 66

Time to peak maximum


2-4 3-4 1-2
concentration (hours)

Protein binding (%) 92-95 87 35

Volume of distribution (L) 50 21 50-70

Half-life (hours) 5-9 Approximately 12 12-17

Major: CYP3A4;
CYP3A4/5, CYP2J2,
Metabolism Minor: CYP1A2, CYP2C8, Conjugation
and hydrolysis
CYP2C19, and CYP2J2

66% (Renal) 25% (Renal)


Elimination 80% (Renal)
33% (Fecal) 55% (Fecal)

References [166, 167] [168, 169] [170, 171]

monitored using the INR by conducting blood tests so that an However, Erkan et al. (2014) recommended the incorpora-
adequate and safe dose can be maintained [48]. tion of a longer duration of anticoagulation therapy to pre-
vent recurrent thrombosis among APS patients [51]. In an-
In APS patients, thrombosis leads to more deaths than
other study, a low dose of warfarin, when given in combina-
bleeding. In fact, according to a systematic review [49], 18
tion with aspirin, successfully prevented recurrent thrombo-
deaths among APS patients were reported to occur due to
recurrent thrombosis compared with only a single death due sis in an APS patient suffering from livedoid vasculitis [52].
to hemorrhage. According to the Euro-Phospholipid cohort According to a randomized, double-blind clinical trial of
study on APS patients (n=1,000), 36.5% of the total deaths APS patients (n=114), a moderate-intensity warfarin treat-
occurred due to thrombotic events during the follow-up pe- ment with a targeted INR between 2.0 and 3.0 was more
riod, whereas only 10.7% patients died due to hemorrhage effective at thromboprophylaxis than a high-intensity war-
[7]. farin treatment (INR 3.0-4.0). Recurrent thrombosis was
A higher targeted INR (>2.0-3.0) may be required for observed in 10.7% of patients receiving the high-intensity
warfarin [43] in APS patients. According to a retrospective warfarin treatment compared with patients receiving a mod-
study, anticoagulation therapy with a targeted INR of >3.0 is erate-intensity treatment, which reported only a 3.4% inci-
more protective in preventing recurrent thrombosis in APS dence [53]. Another randomized trial with APS patients
patients compared with anti-platelet therapy or standard anti- (n=109) also reported that the incidences of recurrent throm-
coagulation therapy that targets a lower INR (2.0-3.0) [50]. bosis were 11.1% and 5.5% among patients receiving war-
316 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

Antithrombotic Therapy in Antiphospholipid Syndrome

Anticoagulant Drugs Anti-platelet Drugs Immunomodulatory Drugs

Inhibits clotting Binds to the widely


Warfarin Rituximab expressed CD20
factors II, VII, IX, X Aspirin Clopidogrel
and regulatory protein on the B cell
proteins C and S surface
Inhibits both Inhibits the ADP
COX1 and COX2, receptor on Eculizumab
Directly inhibits Apixaban platelet Inhibits the cleavage
which produce
free or clot-bound membranes of C5 to C5a and C5b
inflammatory PGs
factor Xa

Directly inhibits Inhibits TLR9 and


Rivaroxaban HCQ reduces inflammatory
free or bound factor
Xa (prothrombinase processes
complex)
Statins Blocks cholesterol
synthesis by
Directly inhibits Dabigatran
inhibiting HMG-CoA
thrombin (factor IIa) reductase

Sirolimus Blocks B and T cell


activation by
inhibiting mTOR

Fig. (1). Summary of the types of antithrombotic therapy used to treat antiphospholipid syndrome and their protein targets or mechanisms of
actions.
COX: Cyclooxygenase; PGs: Prostaglandins; ADP: Adenosine diphosphate; C5: Complement 5; TLR: Toll-like receptor; HCQ: Hydroxy-
chloroquine; HMG-CoA: 3-hydroxy-3-methylglutaryl-coenzyme A; mTOR: Mammalian target of rapamycin.

farin at high (INR 3.0-4.5) and moderate intensities (INR recent (2016) prospective observational study [59] assessing
2.0-3.0), respectively [54]. the quality of anticoagulation therapy in APS patients,
warfarin was shown to be an inferior anticoagulation
A meta-analysis combining the results of these two ran-
therapy, due to the tendency to resist VKAs, in addition to
domized control trials did not establish any significant dif-
the lower therapeutic range (TTR) (<60%), indicating that a
ference in the rate of thrombosis recurrence between the two
considerable proportion of time is spent below the
regimens, although a higher thrombotic risk was observed
therapeutic range. The authors have recommended that pa-
following the use of a higher dose of warfarin [54]. A retro- tients with APS need a significantly higher weekly dosage of
spective study reported that higher doses of warfarin were
warfarin to maintain their targeted INR values. However,
more effective in preventing recurrent thrombosis among
higher intensity anticoagulation therapy is associated with
APS patients compared with either low doses of aspirin or
INR fluctuations and bleeding risk. Therefore, a larger num-
warfarin [55]. According to a familial case, the levels of
ber of prospective studies with larger sample sizes are re-
aPLs were decreased in three patients with thrombotic APS
quired to confirm these findings.
from the same family following prolonged warfarin interven-
tion that targeted moderate INR values (2.0-3.0) [56]. Taken
5.2. Rivaroxaban
together, these results suggest that moderate-intensity war-
farin treatments might be more effective than high-intensity Rivaroxaban (Xarelto®) is the first NOAC that directly
warfarin treatments in preventing thrombosis recurrence in inhibits factor Xa (FXa). The absorption of rivaroxaban is
APS patients. rapid, and food does not have any effects on its absorption or
In APS patients, the variable sensitivity of throm- pharmacokinetic properties. Rivaroxaban reaches its maxi-
mum plasma peak concentration at 1 to 4 h [60]. A bioavail-
boplastin reagents to LA may lead to an overestimated INR.
ability of approximately 80% can be achieved from a 10 mg
This overestimation may pose challenges in both the moni-
rivaroxaban tablet [61]. The half-life of plasma elimination is
toring and management of warfarin anticoagulation. In
5 to 9 h in young people and 11 to 13 h in aged people [60,
addition, the lack of information on PT measurements at
62].
baseline before starting the anticoagulation therapy may
complicate the situation further [57]. A cross-sectional study In 2011, rivaroxaban was initially approved by the US
suggested that a thrombin generation (TG) test should be Food and Drug Administration (FDA) and European
incorporated in future prospective studies to determine the Medicines Agency (EMA) for the treatment of DVT, which
appropriate anticoagulant intensity and address this chal- was later extended to use for the prevention of strokes
lenge, including achieving 3.5 INRs in APS patients to im- (2011), reduction of blood clot recurrences (2012), and
prove their anticoagulation treatment [58]. According to a inhibition of stent thromboses (2013) [63]. Rivaroxaban is
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 317

superior to the VKAs, as it has a fixed dose and does not 5.3. Apixaban
require routine monitoring of its coagulation profile, in addi-
Apixaban (Eliquis®) is another direct FXa-inhibiting
tion to fewer interactions with food, alcohol and drugs [64].
NOAC. After administration, the oral absorption of apixaban
In a Polish cohort of APS patients (n=12), only two APS is rapid, and it achieves the maximum plasma peak concen-
patients (associated with SLE in the presence of LA, aCL tration within 1 to 3 h [75]. The bioavailability is approxi-
and anti-2GPI) were reported to develop recurrent DVT mately 66%, and the elimination half-life is 8 to 15 h in
during treatment with rivaroxaban, although DVT, PE, young people [75, 76]. Apixaban was reported to have a few
stroke or miscarriage were reported as the initial clinical drug interactions; however, it does not interact with food and
manifestations [65]. In a French cohort of APS patients alcohol, as it is partially metabolized by cytochrome P450.
(n=26, duration of APS between 0 and 22 years), rivaroxa- Because of its reduced interactions with foods, alcohol or
ban was suggested (15-20 mg/day) for use in 15 patients who drugs compared with VKA antagonists, such as warfarin,
were suffering from vascular thrombotic events, such as apixaban exerts a more persistent anticoagulant intensities in
DVT, stroke, PE and arterial thrombosis. After 1 to 27 thrombotic patients, without the need for regular INR moni-
months of follow-up, a recurrence of thrombosis was not toring [9, 63]. Apixaban was licensed by the EMA and first
observed in 14 patients; however, three patients discontinued approved by the FDA in 2012. It is currently used to reduce
rivaroxaban treatment due to severe migraines, rectal bleed- the risk of cerebrovascular accidents and peripheral vascular
ing and the recurrence of microthromboses [66]. In another embolisms, including recurrent DVT, which can lead to PE
APS cohort (n=18, disease duration 8.8 ± 6.7 years) from the [77], as superior efficacy of different doses (2.5 and 5.0 mg,
UK, none of the patients exhibited vascular thromboem- twice daily) of apixaban was observed compared to the
bolisms after a mean of 12.9 months of rivaroxaban treat- placebo or low-molecular-weight heparin (LMWH) (1.0
ment, although 13 patients had a previous history of DVT mg/Kg every 12 h) [78, 79].
and five had a history of both DVT and PE [67].
A recent (2016) case report [80] did not observe a
However, a controversial scenario was observed in recurrence of thrombosis in a patient (female, 28 years old)
Brazilian APS patients (n=8) where all of the patients with primary APS [presenting venous thromboembolism
developed thrombi (DVT/PE/renal infarction/stroke) as the (VTE) in the presence of LA and aCL (both IgG and IgM)]
initial manifestations; six patients developed thrombotic following treatment with 5 mg of apixaban (3 times a day).
events (MI/DVT/stroke/arterial ischemia), and two patients Currently, a prospective, randomized, open-label, blinded
exhibited refractory migraines and headaches while being event pilot study is ongoing to assess the efficacy, effective-
treated with rivaroxaban [68]. According to a case report on ness and safety of apixaban (2.5 mg twice a day) for the sec-
an American patient with primary APS (female, 59 years ondary prevention of thrombosis in patients with APS
old), rivaroxaban failed to prevent recurrent thrombosis at a (n=668) compared with warfarin [81]. Therefore, before the
dose of 20 mg/day [69]. In another case report (male, 28 results of this clinical trial are available, the usability of
years old) of a patient with primary APS [70], thrombi apixaban as a secondary preventative measure for thrombosis
occurred in the right internal saphenous vein below the knee in APS patients will remain unclear, and it is not
(120 mm in length), in the middle of the thigh (30 mm), and recommended for use in patients with APS.
in the left internal vein (50 mm), in addition to the
presentation of relapsing superficial thrombophlebitis. When 5.4. Dabigatran
rivaroxaban was administered (20 mg/day), the relapsing Dabigatran (Pradaxa®) is a prodrug that is rapidly ab-
superficial thrombophlebitis was resolved in that patient. sorbed after oral administration, followed by direct binding
Therefore, rivaroxaban could be an effective treatment and thrombin (factor IIa) inhibition. It is completely
choice for APS patients suffering from superficial thrombo- converted into the active form in the plasma and the liver.
phlebitis or vascular thrombosis. However, there are contra- Dabigatran has a half-life of 12 to 14 h, and it achieves the
dictory results regarding the use of rivaroxaban in preventing maximum peak plasma concentration within 2 h after oral
thrombosis in patients with APS. The development of administration [82]. It is reported to have a low bioavailabil-
ity of approximately 6 to 7% [83].
thrombotic events and side-effects, as well as the failure of
rivaroxaban to prevent recurrent thrombosis, has raised ques- Dabigatran is not metabolized by the CYP enzymes.
tions about its safety and effectiveness. Currently, two clini- However, it is a substrate for the P-glycoprotein transporter
cal trials are ongoing (recruiting patients) in Canada [71] and and is mainly excreted by the kidneys [84, 85]. Dabigatran
Italy [72], in addition to one published protocol from the UK was licensed by the EMA in 2008 for the treatment of pa-
[73], to systematically assess the safety and efficacy of ri- tients with VTE, and the FDA initially approved its used for
varoxaban use in thrombotic patients with APS. Therefore, the prevention of stroke in 2010 [73].
before the results of the clinical trials are published, it is not In a French cohort of APS patients (n=26, duration of
recommended that rivaroxaban is used to treat thrombotic APS from 0 to 23 years) with an initial thrombotic history
patients with APS; however, this drug could be an option for (DVT, stroke, PE and arterial thrombosis), dabigatran was
exceptional cases only. In addition, if patients show moder- administered and followed up between 6 and 39 months (150
ate renal impairment (creatinine clearance: 30-49 mL/min), mg/twice daily) in 11 patients. Neither the recurrence of
the dose should be reduced from 20 mg/day to 15 mg/day, thrombotic events nor bleeding were observed in any of the
and rivaroxaban is not recommended in cases of severe renal patients [66]. Dabigatran was reported to be a successful
impairment (creatinine clearance: <15 mL/min) [74]. prolonged anticoagulation therapy that prevented the recur-
318 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

rence of thrombosis after orthotopic liver transplantation in a aspirin with other nonselective, reversible COX inhibitors,
22-year-old Russian male with primary APS co-existing with such as ibuprofen and naproxen, impairs its efficacy, as other
Budd-Chiari syndrome [86]. drugs compete with aspirin for a common docking site (ar-
ginine 120) within the COX channel [96].
A 43-year-old female with primary APS was initially
managed with warfarin; however, due to heavy menstrual Several clinical studies have reported the efficacy of aspi-
bleeding and difficulty in managing her INR, she was di- rin for managing thrombosis in patients with APS. In 2001,
rected to take dabigatran at a dose of 150 mg/day. Eventu- Erkan et al. reported that APS patients who had pregnancy
ally, it failed to prevent the recurrence of vascular thrombo- loss as the only APS manifestation were at high risk of de-
sis [69]. Win et al. [87] reported a study of a 53-year-old veloping subsequent vascular thrombosis and suggested that
female patient with severe APS and a history of multiple aspirin treatment would be an effective prophylaxis against
thrombotic complications [recurrent DVT and TIA]. It was thrombosis [97]. In 2005, an open-label RCT with APS
suggested that she switch from warfarin to dabigatran (150 patients (n=109) reported that the standard treatment (aspirin
mg twice daily) because of her uncontrollable INR due to alone at a dose of 100 mg/day or warfarin at doses adjusted
genetic limitations in warfarin metabolism (heterozygous for to an INR range 2.0-3.0) was superior to high-intensity
CYP2C9 and homozygous for the vitamin K epoxide warfarin (INR range 3.0-4.5) for preventing recurrent
reductase complex subunit 1). Although a recurrence of thrombosis, where warfarin was also found to be linked with
thrombotic events was not observed later, neurologic abnor- an increase in minor hemorrhagic complications [54]. In
malities, such as memory loss, transient weakness, acute 2008, a retrospective study by Hereng et al. on asymptomatic
vision changes and presyncopal events, were apparent. Be- aPL-positive carriers (n=103) associated with different dis-
cause existing studies are not conclusive in regards to the eases reported that SLE patients (4/10) who were not taking
safety and efficacy of dabigatran, larger cohort studies are aspirin developed thrombosis compared to those SLE pa-
needed to obtain a clear picture of the usage of dabigatran in tients (3/27) who were taking aspirin. Patients with autoim-
APS patients with thrombotic manifestations. mune thrombocytopenia (4/6) who were not taking aspirin
developed thrombotic events compared with patients who
5.5. Anti-Platelet Drugs were taking aspirin (1/10). The study results supported the
use of aspirin to prevent thrombotic events as APS manifes-
5.5.1. Aspirin
tation in aPL-positive patients with SLE and autoimmune
Aspirin (acetylsalicylic acid) is a platelet aggregation thrombocytopenia [98].
inhibiting agent that is usually used to prevent thrombosis. In Based on the data from a randomized, double-blind, pla-
addition, aspirin has a substantial role in the treatment of cebo-controlled trial in 2007, Erkan et al. suggested that low-
other health conditions, such as fever, rheumatic fever, colo- dose aspirin (81 mg/day) did not protect against primary
rectal, breast and prostate cancers, pain, neuropsychiatric thrombosis in asymptomatic, persistently aPL-positive indi-
disorders, cardiovascular disease and inflammation-related viduals (without any vascular and/or pregnancy events) [99].
diseases, including rheumatoid arthritis and pericarditis [88]. However, a recent (2014) meta-analysis reported that low-
Aspirin is administered via the oral route. After inges- dose aspirin treatment had a protective effect on the risk of
tion, aspirin takes approximately 30 to 40 min to reach to its primary arterial thrombosis in SLE patients and asympto-
peak plasma level. It has been reported to be rapidly ab- matic aPL individuals compared to HCQ [100]. A prospec-
sorbed in the stomach and upper GI tract [89]. The half-life tive, open RCT also reported the efficacy of low-dose aspirin
of aspirin in the plasma is only 20 min. Over a wide range of alone in comparison with the combination of low-intensity
doses, approximately 40% to 50% bioavailability is reported warfarin plus low-dose aspirin in causing fewer thrombotic
for regular aspirin tablets [90]. and bleeding episodes in aPL-positive patients with SLE
and/or obstetric morbidity [101].
Aspirin exerts its anti-platelet activity by permanently
inactivating the key platelet enzyme cyclooxygenase (COX) Combination treatments consisting of aspirin with other
[91], although nascent platelet generation can reverse this anticoagulants have been reported to have positive outcomes
effect. It has been reported that platelet turnover helps to in several clinical studies. In 2004, a prospective study re-
recover COX activity by approximately 10% per day after ported that aspirin (325 mg/day) and heparin (1 mg/kg
ingestion of a single dose of aspirin [92]. Aspirin-mediated enoxaparin once daily) treatment of asymptomatic aPL-
inhibition of platelet function is measurable within 60 min of carriers (n=178) who were at high risk of thrombosis re-
administration [93]. However, enteric-coated aspirin requires sulted in no episodes of thrombosis in any of the carriers
3 to 4 h to achieve the peak plasma effect [94]. Randomized during the follow-up period of 36 months. On the other hand,
controlled trials (RCTs) reported that aspirin doses between APS patients (n=226) who received the dicumarin treatment
50 and 100 mg/day effectively act as an antithrombotic exhibited a recurrence of venous and arterial thrombosis,
agent, although dosages as low as 30 mg/day are suggested indicating that this treatment was an insufficient anticoagula-
to be highly effective [93]. tion therapy [102].
Aspirin administration is reported to be adversely From our observations, most of the existing evidence
associated with an increased risk of bleeding, specifically, GI supports the use of aspirin alone, rather than in combination
tract bleeding [93]. The combined administration of aspirin with other drugs. It is also observed that the combination of
with other non-steroidal anti-inflammatory drugs (NSAIDs), aspirin with other COX inhibitors leads to competition for
clopidogrel or warfarin has reported to further increase the the common binding sites, whereas combinations with anti-
bleeding risk in the upper GI tract [95]. Co-administration of coagulants increase the bleeding risk, although controversial
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 319

results are also reported. The selection of an effective dose of C levels were adequate. However, the oral administration of
aspirin is also a controversial issue. Therefore, more investi- clopidogrel in association with subcutaneous LMWH
gations are required to make a conclusive decision about the (enoxaparin) and oral aspirin induced the rapid and remark-
effective dose and efficacy of aspirin either alone or in com- able resolution of the skin lesions and improved the clinical
bination with other anti-thrombotic drugs. condition [119].

5.5.2. Clopidogrel In 2009, another 35-year-old male APS patient was diag-
nosed with MI and exhibited a recanalized thrombus in the
Clopidogrel (Plavix®) is a thienopyridine class anti- left anterior descending artery. Treatment with aspirin,
platelet drug. Oral administration of 75 mg clopidogrel clopidogrel and enoxaparin for 2 months managed the clini-
reaches to its maximum peripheral blood concentration cal state of the patient [120]. In 2010, a male patient (59
within 1.5 h which declines to undetectable values in most years old) who was suspected to have APS and presented MI
cases within 6 h [103]. Age and food do not significantly developed multiple recurrent stent thrombosis. The patient
influence its bioavailability [104]. However, interindividual showed mildly elevated titers [12 IgG phospholipid (GPL)
differences may affect the peak plasma concentration and the unit] of the aCL antibody and resistance to clopidogrel.
time to peak plasma concentration [105]. Clopidogrel is rap- However, a 6-week combination treatment with LMWH,
idly absorbed in the intestine and is then activated in the liver clopidogrel and aspirin effectively resolved the chest pain
by cytochrome P450 enzymes, mainly the CYP2C19 [106]. and thrombosis. Undetectable titers of IgG, IgA, IgM aCL
Mechanistically, clopidogrel irreversibly inhibits the P2Y12 and LA were present [121]. In 2011, a female APS patient
subtype of adenosine diphosphate (ADP) chemoreceptors with SLE who had a past history of PE reported a sudden
(present on platelet cell membranes) by forming a disulfide onset of chest pain and shortness of breath. The patient was
bridge. Inhibition of P2Y12 by clopidogrel further inhibits diagnosed with a thrombus in the proximal left anterior de-
platelet activation, which thereby inhibits fibrin-mediated scending artery. However, daily treatment with full-dose
cross-linking and prevents the formation of blood clots [44]. aspirin, clopidogrel (75 mg), continuous heparin infusions,
It is mainly used to treat peripheral vascular, cerebrovascular and a 24-h continuous eptifibatide infusion resolved the
and coronary arterial diseases [107]. Clopidogrel is also very chest pain and breathing problems and showed effective re-
effective in preventing MI and stroke [108]. However, a gression of the thrombus [122]. In 2012, a retrospective
3.58-fold greater risk of major cardiovascular events (stroke single-center study of APS patients (n=81) with arterial
and heart attack) has been reported in patients who express a thromboses revealed that a dual anti-platelet treatment
variant allele of CYP2C19 [109]. In these patients, lower (n=13) with aspirin and ticlopidine/clopidogrel prevented the
levels of the active metabolite of clopidogrel were observed, recurrence of arterial thrombosis in 54% of APS patients
which failed to inhibit a sufficient number of platelets. The
[123]. In 2013, an 18-year-old female APS patient was re-
risk of adverse events is greatest in CYP2C19 poor metabo-
ported to develop recurrent DVT and was treated with long-
lizers [110]. In 2010, the US FDA alerted consumers about
term warfarin therapy. However, the combination of clopi-
the reduced effectiveness of clopidogrel in people with a
dogrel and HCQ instead of warfarin succeeded in preventing
poor capacity to metabolize the drug [111, 112].
the recurrence of thrombotic or bleeding events in that pa-
Bleeding is considered as one of the most common ad- tient [124].
verse effects caused by clopidogrel [113]. In addition to
To the best of our knowledge, most of the evidence
bleeding, thrombotic thrombocytopenic purpura is also
reported as an adverse effect [114]. Co-administration of mentioned above is confined to case reports in which clopi-
aspirin has been reported to increase the rate of hemorrhage dogrel was effective in combination with other anti-
[115]. In 2009, the FDA announced that patients who use thrombotic drugs. Therefore, more prospective studies with
proton pump inhibitors, such as omeprazole or esomepra- larger sample sizes and in vivo studies are required to evalu-
zole, should use caution when they are already taking clopi- ate the safety and efficacy of clopidogrel in APS patients,
dogrel [116] as these drugs can completely inhibit the and its synergistic interaction with other well-documented
CYP2C19 isoenzyme preventing the active metabolite for- anti-thrombotic medications, respectively.
mation of clopidogrel [117].
5.6. Immunomodulatory Drugs
In previous years, several clinical reports revealed the
efficacy of clopidogrel in combination with other agents in 5.6.1. Rituximab
APS or non-APS patients. According to a recent (2014) case
report, clopidogrel in combination with rivaroxaban signifi- Rituximab is a chimeric monoclonal antibody that binds
cantly reduced the symptoms and improved the prognosis of to the B cell surface protein CD20 (anti-CD20 antibody) and
a 55-year-old female (diagnosed with acute PE, acute MI and induces apoptosis [125]. Initially, rituximab was approved
a cerebral infarction) patient who did not have any major for the treatment of B-cell lymphomas, and it has since been
bleeding episodes [118]. successfully used to treat systemic autoimmune diseases,
including APS, with a consistent safety profile [126]. In
According to a case report published in 2003, a 31-year- APS, the pathogenic role of B cells in the generation of
old male APS (associated with SLE) patient developed thrombi is assumed [8]. Therefore, rituximab is used to at-
erythematous purpuric plaques. Biopsies of the lesioned skin tenuate the titer of aPLs and ultimately prevent aPL-
showed intravascular EC proliferation associated with the mediated thromboses in patients with APS, although the ef-
formation of microthrombi, which failed to be resolved by ficiencies were heterogeneous [127, 128]. For the first time,
warfarin treatment, although the therapeutic INR and protein Ahn et al. [129] reported the use of rituximab in an APS
320 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

patient (female, 30 years old) with a thrombotic history of the evidence us from case reports/series and pilot clinical
over 10 years (recurrent and spontaneous DVT, PE and studies; hence, large, well-designed, prospective RCTs
stroke) and the persistent presence of aPLs (LA, aCL and should be conducted to evaluate the efficacy and safety of
anti-2GPI). Following four weekly infusions of rituximab rituximab as a secondary thromboprophylaxis and for the
(375 mg/m2), the LA titers became persistently negative, and treatment of ‘non-criteria’ manifestations in patients with
the aCL IgM titer was temporarily negative and later APS.
reverted to a positive, low titer. Although there was no sig-
nificant change in the aCL (IgG) and anti-2GPI (IgM) lev- 5.6.2. Eculizumab
els, an improvement of her thrombocytopenic condition was Eculizumab (Soliris®) is a humanized monoclonal
observed, as well as a remission from thrombosis. Rituximab antibody (IgG2/4k) that inhibits the activation of C5
(375 mg/m2 weekly x 4 doses) was successful in resolving
complement by preventing the cleavage of C5 into C5a and
cerebral venous sinus thrombosis and in a complete visual
C5b, followed by inhibiting the generation of the membrane
recovery in an APS patient, with no side effects [130]. Stud-
ies reported a reduction in the aPL levels and a reduced fre- attack complex (MAC) [137, 138], which plays a major role
quency of recurrent thrombotic events in patients with APS in the pathogenesis of thrombi formation in APS [8]. Eculi-
who were treated with rituximab and followed for as long as zumab was approved by the FDA and the EMA in 2007 for
41 months [131, 132]. According to a recent (2014) system- the treatment of paroxysmal nocturnal hemoglobinuria
atic analysis of the literature [133], the clinical manifesta- (PHN) and was later approved in 2011 for the treatment of
tions were improved and/or no recurrence of thrombosis was atypical hemolytic uremic syndrome (aHUS) [139].
observed in 23 pediatric APS patients (13 ± 4.6 years) who To date, eculizumab has mainly been used in refractory
were treated with rituximab in combination with immuno- patients with catastrophic APS [140]. In 2012, eculizumab
suppressive regimens, anticoagulants and/or steroids. How- was administered (six doses of 600 mg approximately every
ever, none of the patients who were positive for aPLs (LA, week) to a 28-year-old young man suffering from cata-
aCL and anti-2GPI) became negative for these antibodies strophic APS and during the more than three-year follow-up
after the rituximab treatment. period, no recurrence of any forms of thrombotic events was
According to Berman et al. [134], among the 20 observed in addition to the reversal of his thrombocytopenia
refractory catastrophic APS patients from the “European [141]. Eculizumab was also successful in another cata-
CAPS registry”, 16 recovered (80%) from acute episodes of strophic APS patient with thrombotic microangiopathy
catastrophic APS after treatment with different dosages of (male, 51 years old) who underwent renal transplantation
rituximab [8 (40%) and 12 (60%) patients received rituximab [142]. Recently (2015), a 47-year-old primary catastrophic
as the first-line and second-line treatments, respectively], but male APS patient was treated with eculizumab, which pro-
4 (20%) patients died during the follow-up. Rituximab was gressively improved both the clinical and laboratory features
also used to treat APS patients who exhibited ‘non-criteria’ during the over 16-month follow-up period [143].
manifestations, including thrombocytopenia, skin ulcers and
To date, histories of success for eculizumab treatments in
hemolytic anemia with persistent levels of aPLs [127, 135].
APS patients are very limited and are only confined to case
Recently (2016), a 43-year-old triple aPL-positive (with high
reports. Currently, a clinical trial investigating the efficacy of
titers) woman was treated with rivaroxaban, tinzaparin (a
LMWH), warfarin and aspirin for her recurrent DVT and eculizumab in kidney-transplanted patients with catastrophic
multifocal cerebral ischemic symptoms. Despite the use of APS is ongoing (ClinicalTrials.gov Identifier: NCT01029587)
these regimens, she experienced recurrent transient stroke [144]. In addition, large prospective cohort studies are war-
episodes. Therefore, in addition to her existing medications, ranted to investigate the safety and efficacy of eculizumab in
HCQ, methylprednisolone and rituximab were added. After 3 patients with thrombotic APS.
months of follow-up, she did not experience further throm- 5.6.3. Hydroxychloroquine (HCQ)
botic events [136]. Therefore, rituximab may be another
emerging regimen that can be used in combination with other HCQ (Plaquenil®) is mainly an antimalarial regimen that
anticoagulants, anti-platelets, anti-inflammatory drugs and possesses anti-inflammatory, immunomodulatory and anti-
appropriate steroids to restrain the recurrence of thrombosis thrombotic activities. It is widely used for the treatment of
in APS patients. rheumatoid arthritis, SLE and APS [145].
The abovementioned evidence is from case reports/series According to in vitro studies, a significant reduction of
and pilot clinical studies; therefore, large, well-designed, binding between anti-2GPI and the phospholipid bilayer
prospective RCTs should be conducted to evaluate the effi- [146], inhibition of GPIIb/IIIa on aPL-activated platelets
cacy and safety of rituximab as a secondary thromboprophy- [147] and prevention of aPL-induced disruption of the
laxis and for the treatment of ‘non-criteria’ manifestations in annexin A5 shield [148] were observed upon treatment with
patients with APS. effective concentrations of HCQ as low as 1, 25 and 1
Based on our observations, rituximab is more effective in μg/mL, respectively. HCQ significantly diminished the sizes
combination with other regimens than alone. Therefore, of the thrombi in male CD-1 mice that were injected with
more studies of rituximab are warranted to evaluate its effi- human IgG aCL, possibly by reversing the thrombogenic
cacy in combination with anticoagulants, anti-platelet drugs, properties of aPLs [149]. A cross-sectional study observed a
anti-inflammatory drugs and appropriate steroids to restrain possible role of HCQ in protecting asymptomatic aPL-
the recurrence of thrombosis in APS patients. Currently, all positive subjects from thrombosis [150].
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 321

According to the first prospective, non-randomized study isomerase (PDI)] that were responsible for the induction of
of patients with primary APS, co-administration of oral anti- thrombosis in patients with APS were observed [159].
coagulants and HCQ protected against recurrent thrombosis Although the use of statins to prevent the recurrence of
[151]; however, a recent meta-analysis (2015) found that thrombosis in APS patients might be beneficial (because of
there was no substantial protective effect of HCQ on throm- the lipid-lowering activity of statins), clinical evidence of the
bosis [100]. According to a recent (2015) observational effects of statins on APS patients is substantively limited.
study, 26 patients with thrombotic APS were treated with Therefore, RCTs are warranted to determine the safety and
NOACs, rivaroxaban or dabigatran, and HCQ was efficacy of using statins to prevent thrombosis in patients
administered to 50% of the patients (n=13) as an adjunct with APS.
regiment [66]. Among the 13 patients, a recurrence of
thrombosis was not observed in 11 subjects, whereas 2 dis- 5.6.5. Sirolimus
continued the treatment due to severe migraines and recur-
Sirolimus (Rapamune®), also known as rapamycin, is an
rent rectal bleeding (no information was available about their immunosuppressor that acts on the mammalian target of
recurrence of thrombosis). Therefore, concurrent treatment rapamycin (mTOR, a kinase that integrates a variety of
strategies with NOACs and HCQ could be an interesting signaling pathways to regulate cellular growth, proliferation
clinical aspect of determining whether HCQ exerts any and survival) and thereby inhibits the activation of B and T
synergistic effect on preventing the recurrence of thrombosis cells by inhibiting IL-2 [160, 161]. Sirolimus has success-
in APS patients. fully been used to prevent rejection in organ transplantation,
As the results of existing studies are contradictory, it especially renal transplantations [162]. In 2014, Canaud
would be interesting to investigate the effects of concurrent et al. [163] reported that patients with APS nephropathy who
treatment strategies using NOACs and HCQ to observe required renal transplantation were treated with sirolimus
whether HCQ exerts any synergistic effect on preventing and did not show a recurrence of vascular lesions compared
recurrence of thrombosis in APS patients. Currently, a clini- with patients who were not treated with sirolimus. In
cal trial investigating the efficacy of HCQ on the endothelial addition, 70% of transplanted patients who were treated with
dysfunction in patients with APS is ongoing (ClinicalTri- sirolimus had a functioning renal allograft (144-month fol-
als.gov Identifier: NCT02595346) [152], and the results of low-up) compared with 11% of patients who were not re-
ceiving sirolimus. Therefore, sirolimus could be a potential
this trial are expected to clarify the contradiction regarding
immunosuppressive drug for renal transplanted patients with
the efficiency of using HCQ to treat thrombotic patients with
APS; however, well-designed RCTs are required to confirm
APS.
these findings.
5.6.4. Statins
6. FUTURE DIRECTIONS AND CONCLUSION
Statins are a class of lipid-lowering drugs that regulate
cholesterol levels by inhibiting 3-hydroxy-3-methylglutaryl- Although the thrombotic pathogenesis of APS has been
coenzyme A (HMG-CoA) reductase [153]. A meta-analysis studied extensively, further studies are required to identify
revealed that use of statins is significantly associated with novel molecular targets and develop precise treatment strate-
gies for thrombotic APS.
reduced frequencies of VTE [154]. Based on in vitro studies,
statins (fluvastatin, simvastatin and rosuvastatin) diminished As the mechanism of thrombi generation in APS is
the aPL-mediated upregulation of TFs and adhesion mole- unique compared to general thrombi formation, new thera-
cules in ECs [155, 156]. A similar type of ameliorating effect peutic approaches targeting pathogenic molecules are en-
was observed in an in vivo study of aPL-induced throm- couraged for the management of thrombosis in APS. In addi-
bogenic and proinflammatory biomarkers [157], indicating tion to the existing VKA anticoagulants and anti-platelet
that statins have the potential to regulate inflammation and agents, new NOACs and immunomodulatory agents seem to
thrombosis. have the potential to prevent thrombosis or manage the re-
currence of thrombosis. However, firm clinical safety and
According to a recent (2014) 3 month, open-label, efficacy data are required before wide therapeutic use is
prospective pilot intervention trial (ClinicalTrials.gov practiced (except for exceptional refractory cases in which
identifier: NCT00674297) with aPL-positive subjects (n=41, all the currently approved drugs are ineffective). Upon the
39 had history of thrombotic events) [158], 40 mg/day completion of current, ongoing clinical trials investigating
fluvastatin significantly (p<0.05) diminishes 50% of the APS and newer drugs in the thrombotic management of
proinflammatory and prothrombotic biomarkers [interleukin APS, novel information is expected to add guidance for gov-
(IL)-1, vascular endothelial growth factor (VEGF), TNF-, erning the thrombotic management of APS in a better direc-
inducible protein-10 (IP-10), soluble CD40 ligand (sCD40L) tion. Currently, our Cochrane systematic review and meta-
and soluble TF] that are generally upregulated in patients analysis [164] assessing the efficacy and safety of anti-
with APS. A case-control study with APS patients (n=42) platelet and anticoagulant agents at preventing the recurrence
observed that upon administration of 20 mg of of any peripheral vascular thrombosis (arterial or venous, or
fluvastatin/day for 30 days, a significant reduction (p<0.05) both) in patients with APS is ongoing and is expected to
in the levels of several monocyte-mediated inflammatory clarify the appropriate type of regimens to use in the
molecules [TF, protein activator receptors (PAR) 1, 2 and thrombotic management of APS patients. Combined thera-
VEGF] and altered expression of some thrombogenic peutic approaches targeting different unregulated thrombotic
biomarkers (annexin A2, RhoA and protein disulfide pathways in APS can be a potential management strategy;
322 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

therefore, well-designed RCTs are required to investigate the [11] Bittar M, Uthman I. Antiphospholipid syndrome novel therapies.
efficacy and safety of combined therapy compared with pla- EMJ Rheum 2014; 1(1): 113-21.
[12] Santamaria JR, Badziak D, Barros MF, Mandelli FL, Cavalin LC,
cebo or other currently used individual antithrombotic regi- Sato MS. Antiphospholipid syndrome. An Bras Dermatol 2005;
mens. In addition to the current antithrombotic regimens, the 80(3): 225-39.
development of new antithrombotic drugs from natural com- [13] Joseph JE, Harrison P, Mackie IJ, Isenberg DA, Machin SJ. In-
pounds is encouraged [165], which can be another potential creased circulating platelet-leucocyte complexes and platelet acti-
vation in patients with antiphospholipid syndrome, systemic lupus
treatment for APS patients in the future. erythematosus and rheumatoid arthritis. Br J Haematol 2001;
115(2): 451-9.
LIST OF ABBREVIATIONS [14] Fanelli A, Bergamini C, Rapi S, et al. Flow cytometric detection of
circulating activated platelets in primary antiphospholipid syn-
APS = Antiphospholipid Syndrome drome. Correlation with thrombocytopenia and anticardiolipin an-
tibodies. Lupus 1997; 6(3): 261-7.
aPL = Antiphospholipid Antibody [15] Jy W, Tiede M, Bidot CJ, et al. Platelet activation rather than endo-
DVT = Deep Vein Thrombosis thelial injury identifies risk of thrombosis in subjects positive for
antiphospholipid antibodies. Thromb Res 2007; 121(3): 319-25.
HCQ = Hydroxychloroquine [16] Vlachoyiannopoulos PG, Routsias JG. A novel mechanism of
thrombosis in antiphospholipid antibody syndrome. J Autoimmun
MI = Myocardial Infarction 2010; 35(3): 248-55.
[17] Lindsey NJ, Dawson RA, Henderson FI, Greaves M, Hughes P.
PE = Pulmonary Embolism Stimulation of von willebrand factor antigen release by immuno-
globulin from thrombosis prone patients with systemic lupus
SLE = Systemic Lupus Erythematosus erythematosus and the antiphospholipid syndrome. Rheumatology
1993; 32(2): 123-6.
CONFLICT OF INTEREST [18] Cugno M, Borghi MO, Lonati LM, et al. Patients with antiphos-
pholipid syndrome display endothelial perturbation. J Autoimmun
The authors confirm that this article content has no con- 2010; 34(2): 105-10.
flict of interest. [19] Crawley JT, de Groot R, Xiang Y, Luken BM, Lane DA. Unravel-
ing the scissile bond: How adamts13 recognizes and cleaves von
willebrand factor. Blood 2011; 118(12): 3212-21.
ACKNOWLEDGEMENTS [20] Amoura Z, Costedoat-Chalumeau N, Veyradier A, et al. Throm-
We would like to acknowledge the Universiti Sains Ma- botic thrombocytopenic purpura with severe adamts-13 deficiency
in two patients with primary antiphospholipid syndrome. Arthritis
laysia (USM) Vice-Chancellor Award (2015/2016) and the Rheum 2004; 50(10): 3260-4.
USM Global Fellowship (2014/2015) awarded to Md. Asiful [21] Årfors L, Vesterqvist O, Johnsson H, Green K. Increased throm-
Islam and Fahmida Alam, respectively, to pursue their PhD boxane formation in patients with antiphospholipid syndrome. Eur
degrees. J Clin Invest 1990; 20(6): 607-12.
[22] Carreras L, Vermylen J. “Lupus” anticoagulant and thrombosis--
possible role of inhibition of prostacyclin formation. Thromb Hae-
REFERENCES most 1982; 48(1): 38-40.
[23] Ao W, Zheng H, Chen X-W, Shen Y, Yang C-D. Anti-annexin ii
[1] Miyakis S, Lockshin MD, Atsumi T, et al. International consensus
antibody is associated with thrombosis and/or pregnancy morbidity
statement on an update of the classification criteria for definite an-
in antiphospholipid syndrome and systemic lupus erythematosus
tiphospholipid syndrome (APS). J Thromb Haemost 2006; 4(2):
with thrombosis. Rheumatol Int 2011; 31(7): 865-9.
295-306. [24] Benhamou Y, Bellien J, Armengol G, et al. Role of toll-like recep-
[2] Asherson RA, Khamashta MA, Ordi-ros J, et al. The "primary" tors 2 and 4 in mediating endothelial dysfunction and arterial re-
antiphospholipid syndrome: Major clinical and serological features. modeling in primary arterial antiphospholipid syndrome. Arthritis
Medicine (Baltim) 1989; 68(6): 366-74. Rheumatol 2014; 66(11): 3210-20.
[3] Alarcón-Segovia D, Delezé M, Oria CV, et al. Antiphospholipid [25] Atsumi T, Khamashta MA, Andujar C, et al. Elevated plasma lipo-
antibodies and the antiphospholipid syndrome in systemic lupus protein (a) level and its association with impaired fibrinolysis in pa-
erythematosus a prospective analysis of 500 consecutive patients. tients with antiphospholipid syndrome. J Rheumatol 1998; 25(1):
Medicine (Baltim) 1989; 68(6): 353-65. 69-73.
[4] Cervera R, Bucciarelli S, Plasín MA, et al. Catastrophic antiphos- [26] López-Pedrera C, Buendía P, Barbarroja N, Siendones E, Velasco
pholipid syndrome (CAPS): Descriptive analysis of a series of 280 F, Cuadrado MJ. Antiphospholipid-mediated thrombosis: Interplay
patients from the “caps registry”. J Autoimmun 2009; 32(3): 240-5. between anticardiolipin antibodies and vascular cells. Clin Appl
[5] Lechner K, Pabinger-Fasching I. Lupus anticoagulants and throm- Thromb Hemost 2006; 12(1): 41-5.
bosis. Pathophysiol Haemost Thromb 1985; 15(4): 254-62. [27] Rand JH, Wu XX, Quinn AS, Taatjes DJ. Resistance to annexin a5
[6] Asherson RA. The antiphospholipid syndrome ii: Autoimmune anticoagulant activity: A thrombogenic mechanism for the an-
thrombosis., 1st ed. Elsevier Science: Amsterdam 2002; pp. 478. tiphospholipid syndrome. Lupus 2008; 17(10): 922-30.
[7] Cervera R, Serrano R, Pons-Estel GJ, et al. Morbidity and mortality [28] Keeling DM, Wilson AJ, Mackie IJ, Isenberg DA, Machin SJ. Role
in the antiphospholipid syndrome during a 10-year period: A multi- of beta 2-glycoprotein i and anti-phospholipid antibodies in activa-
centre prospective study of 1000 patients. Ann Rheum Dis 2014; tion of protein c in vitro. J Clin Pathol 1993; 46(10): 908-11.
74(6): 1011-8. [29] Erkan D, Zhang HW, Shriky RC, Merrill JT. Dual antibody reactiv-
[8] Islam MA, Alam F, Sasongko TH, Gan SH. Antiphospholipid ity to 2-glycoprotein i and protein s: Increased association with
antibody-mediated thrombotic mechanisms in antiphospholipid thrombotic events in the antiphospholipid syndrome. Lupus 2002;
syndrome: Towards pathophysiology-based treatment. Curr Pharm 11(4): 215-20.
Des 2016; 22(28): 4451-69. [30] Giannakopoulos B, Gao L, Qi M, et al. Factor xi is a substrate for
[9] Chighizola CB, Ubiali T, Meroni PL. Treatment of thrombotic oxidoreductases: Enhanced activation of reduced fxi and its role in
antiphospholipid syndrome: The rationale of current management- antiphospholipid syndrome thrombosis. J Autoimmun 2012; 39(3):
an insight into future approaches. J Immunol Res 2015; 2015: 1-20. 121-9.
[10] Nalli C, Andreoli L, Casu C, Tincani A. Management of recurrent [31] Adams M, Breckler L, Stevens P, Thom J, Baker R, Oostryck R.
Anti-tissue factor pathway inhibitor activity in subjects with an-
thrombosis in antiphospholipid syndrome. Curr Rheumatol Rep
tiphospholipid syndrome is associated with increased thrombin
2014; 16(3): 1-8.
generation. Haematologica 2004; 89(8): 985-90.
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 323

[32] Pierangeli SS, Vega-Ostertag M, Liu X, Girardi G. Complement [55] Rosove MH, Brewer PM. Antiphospholipid thrombosis: Clinical
activation: A novel pathogenic mechanism in the antiphospholipid course after the first thrombotic event in 70 patients. Ann Intern
syndrome. Ann NY Acad Sci 2005; 1051(1): 413-20. Med 1992; 117(4): 303-8.
[33] Breen KA, Seed P, Parmar K, Moore GW, Stuart-Smith SE, Hunt [56] Islam MA, Wong KK, Sasongko TH, Gan SH, Wong JS. Familial
BJ. Complement activation in patients with isolated antiphosphol- primary antiphospholipid syndrome: A report of co-occurrence in
ipid antibodies or primary antiphospholipid syndrome. Thromb three malaysian family members. Eur J Rheumatol 2015; 3(3): 139-
Haemost 2012; 107(3): 423-9. 41.
[34] Simonin L, Pasquier E, Leroyer C, et al. Among patients with [57] Tripodi A, Chantarangkul V, Clerici M, Negri B, Galli M, Man-
venous thrombo-embolism, B cell subset disturbances characterise nucci PM. Laboratory control of oral anticoagulant treatment by the
those with primary antiphospholipid syndrome. Ann Rheum Dis inr system in patients with the antiphospholipid syndrome and lu-
2015; 74(1): 4. pus anticoagulant. Results of a collaborative study involving nine
[35] Conti F, Spinelli FR, Alessandri C, et al. Subclinical atherosclero- commercial thromboplastins. Br J Haematol 2001; 115(3): 672-8.
sis in systemic lupus erythematosus and antiphospholipid syndrome [58] Efthymiou M, Lawrie AS, Mackie I, et al. Thrombin generation
focus on 2GPI-specific T cell response. Arterioscler Thromb Vasc and factor x assays for the assessment of warfarin anticoagulation
Biol 2014; 34(3): 661-8. in thrombotic antiphospholipid syndrome. Thromb Res 2015;
[36] Swadzba J, Iwaniec T, Musial J. Increased level of tumor necrosis 135(6): 1191-7.
factor- in patients with antiphospholipid syndrome: Marker not [59] Pastori D, Parrotto S, Vicario T, et al. Antiphospholipid syndrome
only of inflammation but also of the prothrombotic state. Rheuma- and anticoagulation quality: A clinical challenge. Atherosclerosis
tol Int 2011; 31(3): 307-13. 2016; 244(1): 48-50.
[37] Bearevi M, Ignjatovi S, Majki-Singh N. Deterioration of [60] Kubitza D, Becka M, Wensing G, Voith B, Zuehlsdorf M. Safety,
thromboses in primary antiphospholipid syndrome: Tnf-alpha and pharmacodynamics, and pharmacokinetics of bay 59-7939-an oral,
anti-annexin a5 antibodies. Clin Lab 2012; 58(9-10): 1079-84. direct factor xa inhibitor-after multiple dosing in healthy male sub-
[38] O'Reilly RA. Vitamin k and the oral anticoagulant drugs. Annu Rev jects. Eur J Clin Pharmacol 2005; 61(12): 873-80.
Med 1976; 27(1): 245-61. [61] Eriksson BI, Quinlan DJ, Weitz JI. Comparative pharmacodynam-
[39] Breckenridge A. Oral anticoagulant drugs: Pharmacokinetic as- ics and pharmacokinetics of oral direct thrombin and factor xa in-
pects. Semin Hematol 1978; 15(1): 19-26. hibitors in development. Clin Pharmacokinet 2009; 48(1): 1-22.
[40] Kaminsky LS, Zhang ZY. Human p450 metabolism of warfarin. [62] Mueck W, Borris LC, Dahl OE, et al. Population pharmacokinetics
Pharmacol Ther 1997; 73(1): 67-74. and pharmacodynamics of once-and twice-daily rivaroxaban for the
[41] Freedman MD. Oral anticoagulants: Pharmacodynamics, clinical prevention of venous thromboembolism in patients undergoing to-
indications and adverse effects. J Clin Pharmacol 1992; 32(3): 196- tal hip replacement. Thromb Haemost 2008; 100(3): 453-61.
209. [63] Sciascia S, Lopez-Pedrera C, Cecchi I, Pecoraro C, Roccatello D,
[42] Funk DMA. Coagulation assays and anticoagulant monitoring. Cuadrado MJ. Non-vitamin k antagonist oral anticoagulants and
Hematology 2012; 2012(1): 460-5. antiphospholipid syndrome. Rheumatology 2016; 55(10): 1726-35.
[43] Hirsh J, Dalen JE, Anderson DR, et al. Oral anticoagulants: [64] Agnelli G, Becattini C, Franco L. New oral anticoagulants for the
Mechanism of action, clinical effectiveness, and optimal therapeu- treatment of venous thromboembolism. Best Pract Res Clin Hae-
tic range. Chest 2001; 119(1): 8-21. matol 2013; 26(2): 151-61.
[44] Brown DG, Wilkerson EC, Love WE. A review of traditional and [65] Son M, Wypasek E, Celinska-Lowenhoff M, Undas A. The use of
novel oral anticoagulant and antiplatelet therapy for dermatologists rivaroxaban in patients with antiphospholipid syndrome: A series
and dermatologic surgeons. J Am Acad Dermatol 2015; 72(3): 524- of 12 cases. Thromb Res 2015; 135(5): 1035-6.
34. [66] Noel N, Dutasta F, Costedoat-Chalumeau N, et al. Safety and effi-
cacy of oral direct inhibitors of thrombin and factor xa in antiphos-
[45] Wysowski DK, Nourjah P, Swartz L. Bleeding complications with
pholipid syndrome. Autoimmun Rev 2015; 14(8): 680-5.
warfarin use: A prevalent adverse effect resulting in regulatory ac-
[67] Sciascia S, Hunt B. Rivaroxaban use in patients with antiphosphol-
tion. Arch Intern Med 2007; 167(13): 1414-9.
ipid syndrome patients and previous poor anticoagulation control
[46] Holbrook AM, Pereira JA, Labiris R, et al. Systematic overview of
with vitamin k antagonists. Arthritis Rheumatol 2014; 66(1): 7.
warfarin and its drug and food interactions. Arch Intern Med 2005;
[68] Signorelli F, Nogueira F, Domingues V, Mariz HA, Levy RA.
165(10): 1095-106.
Thrombotic events in patients with antiphospholipid syndrome
[47] Gage BF, Fihn SD, White RH. Management and dosing of warfarin
treated with rivaroxaban: A series of eight cases. Clin Rheumatol
therapy. Am J Med 2000; 109(6): 481-8.
2015; 35(3): 801-5.
[48] Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G.
[69] Schaefer JK, McBane RD, Black DF, Williams LN, Moder KG,
Pharmacology and management of the vitamin k antagonists:
Wysokinski WE. Failure of dabigatran and rivaroxaban to prevent
American college of chest physicians evidence-based clinical prac-
thromboembolism in antiphospholipid syndrome: A case series of
tice guidelines. Chest 2008; 133(6): 160-98. three patients. Thromb Haemost 2014; 112(5): 947-50.
[49] RuizIrastorza G, Hunt BJ, Khamashta MA. A systematic review of [70] Bachmeyer C, Elalamy I. Rivaroxaban as an effective treatment for
secondary thromboprophylaxis in patients with antiphospholipid recurrent superficial thrombophlebitis related to primary antiphos-
antibodies. Arthritis Care Res 2007; 57(8): 1487-95. pholipid syndrome. Clin Exp Dermatol 2014; 39(7): 840-1.
[50] Khamashta MA, Cuadrado MJ, Mujic F, Taub NA, Hunt BJ, [71] St. Joseph's Healthcare Hamilton. Rivaroxaban in antiphospholipid
Hughes GRV. The management of thrombosis in the antiphosphol- syndrome pilot study: A multicenter feasibility study of rivaroxa-
ipid-antibody syndrome. N Engl J Med 1995; 332(15): 993-7. ban for patients with antiphospholipid syndrome and prior arterial
[51] Erkan D, Aguiar CL, Andrade D, et al. 14th international congress or venous thrombosis. In: Clinicaltrials.Gov. Available from:
on antiphospholipid antibodies task force report on antiphosphol- https://www.clinicaltrials.gov/ct2/show/NCT02116036. [Accessed
ipid syndrome treatment trends. Autoimmun Rev 2014; 13(6): 685- on 30 July, 2016].
96. [72] University of Padova. A prospective, randomized clinical trial
[52] So BJ, Park JB, Yoo MG, Kim IH, Son SW. Successful treatment comparing rivaroxaban vs warfarin in high risk patients with an-
of livedoid vasculitis with primary antiphospholipid syndrome by tiphospholipid syndrome. In: Clinicaltrials.Gov. Available from:
using aspirin and low dose warfarin combination therapy. Ann https://www.clinicaltrials.gov/ct2/show/NCT02157272. [Accessed
Dermatol 2015; 27(5): 614-5. on 30 July, 2016].
[53] Crowther MA, Ginsberg JS, Julian J, et al. A comparison of two [73] Cohen H, Doré CJ, Clawson S, et al. Rivaroxaban in antiphosphol-
intensities of warfarin for the prevention of recurrent thrombosis in ipid syndrome (RAPS) protocol: A prospective, randomized con-
patients with the antiphospholipid antibody syndrome. N Engl J trolled phase ii/iii clinical trial of rivaroxaban versus warfarin in
Med 2003; 349(12): 1133-8. patients with thrombotic antiphospholipid syndrome, with or with-
[54] Finazzi G, Marchioli R, Brancaccio V, et al. A randomized clinical out sle. Lupus 2015; 24(10): 1087-94.
trial of high-intensity warfarin vs. Conventional antithrombotic [74] Braham S, Bucciarelli P, Moia M. Treatment of thrombosis in
therapy for the prevention of recurrent thrombosis in patients with antiphospholipid syndrome. In: Antiphospholipid antibody syn-
the antiphospholipid syndrome (APS). J Thromb Haemost 2005; drome: From bench to bedside, 1st ed. Springer International Pub-
3(5): 848-53. lishing: Switzerland 2015; pp. 185-92.
324 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

[75] Frost C, Wang J, Nepal S, et al. Apixaban, an oral, direct factor xa [99] Erkan D, Harrison MJ, Levy R, et al. Aspirin for primary thrombo-
inhibitor: Single dose safety, pharmacokinetics, pharmacodynamics sis prevention in the antiphospholipid syndrome: A randomized,
and food effect in healthy subjects. Br J Clin Pharmacol 2013; double-blind, placebocontrolled trial in asymptomatic antiphos-
75(2): 476-87. pholipid antibody-positive individuals. Arthritis Rheum 2007;
[76] Frosi C, Yu Z, Nepal S, et al. Apixaban, a direct factor xa lnhibitor: 56(7): 2382-91.
Single-dose pharmacokinetics and pharmacodynamics of an intra- [100] Arnaud L, Mathian A, Devilliers H, et al. Patient-level analysis of
venous formulation. J Clin Pharmacol 2008; 48(9): 1132. five international cohorts further confirms the efficacy of aspirin
[77] Mandernach MW, Beyth RJ, Rajasekhar A. Apixaban for the pro- for the primary prevention of thrombosis in patients with antiphos-
phylaxis and treatment of deep vein thrombosis and pulmonary pholipid antibodies. Autoimmun Rev 2015; 14(3): 192-200.
embolism: An evidence-based review. Ther Clin Risk Manag 2015; [101] Cuadrado MJ, Bertolaccini ML, Seed PT, et al. Low-dose aspirin
26(11): 1273-82. vs low-dose aspirin plus low-intensity warfarin in thromboprophy-
[78] Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treat- laxis: A prospective, multicentre, randomized, open, controlled trial
ment of acute venous thromboembolism. N Engl J Med 2013; in patients positive for antiphospholipid antibodies (ALIWAPAS).
369(9): 799-808. Rheumatology 2014; 53(2): 275-84.
[79] Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended [102] Girón-González JA, del Río EG, Rodríguez C, Rodríguez-Martorell
treatment of venous thromboembolism. N Engl J Med 2013; J, Serrano A. Antiphospholipid syndrome and asymptomatic carri-
368(8): 699-708. ers of antiphospholipid antibody: Prospective analysis of 404 indi-
[80] Betancur JF, Bonilla-Abadía F, Hormaza AA, Jaramillo FJ, Cañas viduals. J Rheumatol 2004; 31(8): 1560-7.
CA, Tobón GJ. Direct oral anticoagulants in antiphospholipid syn- [103] Sangkuhl K, Klein TE, Altman RB. Clopidogrel pathway. Pharma-
drome: A real life case series. Lupus 2016; 25(6): 658-62.
cogen Genomics 2010; 20(7): 463-5.
[81] Woller SC, Stevens SM, Kaplan DA, et al. Apixaban for the sec-
[104] McEwen J, Strauch G, Perles P, et al. Clopidogrel bioavailability:
ondary prevention of thrombosis among patients with antiphos-
Absence of influence of food or antacids. Semin Thromb Hemost
pholipid syndrome study rationale and design (ASTRO-APS). Clin
1998; 25(2): 47-50.
Appl Thromb Hemost 2015; 22(3): 239-47.
[82] Stangier J, Stähle H, Rathgen K, Fuhr R. Pharmacokinetics and [105] Caplain H, Donat F, Gaud C, Necciari J. Pharmacokinetics of
pharmacodynamics of the direct oral thrombin inhibitor dabigatran clopidogrel. Semin Thromb Hemost 1998; 25(2): 25-8.
in healthy elderly subjects. Clin Pharmacokinet 2008; 47(1): 47-59. [106] Liu Q, Dang DS, Chen YF, Yan M, Shi GB, Zhao QC. The influ-
[83] Blech S, Ebner T, Ludwig-Schwellinger E, Stangier J, Roth W. The ence of omeprazole on platelet inhibition of clopidogrel in various
metabolism and disposition of the oral direct thrombin inhibitor, cyp2c19 mutant alleles. Genet Test Mol Biomarkers 2012; 16(11):
dabigatran, in humans. Drug Metab Dispos 2008; 36(2): 386-99. 1293-7.
[84] Brighton T. New oral anticoagulant drugs–mechanisms of action. [107] Tran H, Anand SS. Oral antiplatelet therapy in cerebrovascular
Aust Prescr 2010; 33(2): 38-41. disease, coronary artery disease, and peripheral arterial disease.
[85] Chighizola CB, Moia M, Meroni PL. New oral anticoagulants in JAMA 2004; 292(15): 1867-74.
thrombotic antiphospholipid syndrome. Lupus 2014; 23(12): 1279- [108] Trialists’Collaboration A. Collaborative meta-analysis of random-
82. ised trials of antiplatelet therapy for prevention of death, myocar-
[86] Reshetnyak TM, Seredavkina NV, Satybaldyeva MA, Nasonov EL, dial infarction, and stroke in high risk patients. BMJ 2002;
Reshetnyak VI. Liver transplantation in a patient with primary an- 324(7329): 71-86.
tiphospholipid syndrome and budd-chiari syndrome. World J Hepa- [109] Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants
tol 2015; 7(19): 2229-36. of response to clopidogrel and cardiovascular events. N Engl J Med
[87] Win K, Rodgers GM. New oral anticoagulants may not be effective 2009; 360(4): 363-75.
to prevent venous thromboembolism in patients with antiphosphol- [110] Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 poly-
ipid syndrome. Am J Hematol 2014; 89(10): 1017. morphisms and response to clopidogrel. N Engl J Med 2009;
[88] Berk M, Dean O, Drexhage H, et al. Aspirin: A review of its neu- 360(4): 354-62.
robiological properties and therapeutic potential for mental illness. [111] Food Drug Administration. FDA announces new boxed warning on
BMC Med 2013; 11(74): 1-17. plavix: Alerts patients, health care professionals to potential for re-
[89] Benedek IH, Joshi AS, Pieniaszek HJ, King SYP, Kornhauser DM. duced effectiveness, published in 2010. Press release. Retrieved on
Variability in the pharmacokinetics and pharmacodynamics of low 30 May 2016.
dose aspirin in healthy male volunteers. J Clin Pharmacol 1995; [112] Food Drug Administration. FDA drug safety communication: Re-
35(12): 1181-6. duced effectiveness of plavix (clopidogrel) in patients who are poor
[90] McAdam B, Catella-Lawson F, Mardini I, Kapoor S, Lawson J, metabolizers of the drug, revised 2010.
FitzGerald G. Systemic biosynthesis of prostacyclin by cyclooxy- [113] Eikelboom JW, Mehta SR, Anand SS, Xie C, Fox KA, Yusuf S.
genase (COX)-2: The human pharmacology of a selective inhibitor Adverse impact of bleeding on prognosis in patients with acute
of cox-2. Proc Natl Acad Sci USA 1999; 96(1): 272-7. coronary syndromes. Circulation 2006; 114(8): 774-82.
[91] Roth GJ, Stanford N, Majerus PW. Acetylation of prostaglandin [114] Zakarija A, Bandarenko N, Pandey DK, et al. Clopidogrel-
synthase by aspirin. Proc Natl Acad Sci USA 1975; 72(8): 3073-6. associated ttp an update of pharmacovigilance efforts conducted by
[92] Burch J, Stanford N, Majerus P. Inhibition of platelet prostaglandin independent researchers, pharmaceutical suppliers, and the food
synthetase by oral aspirin. J Clin Invest 1978; 61(2): 314-9. and drug administration. Stroke 2004; 35(2): 533-7.
[93] Patrono C, Coller B, Dalen JE, et al. Platelet-active drugs: The [115] Diener HC, Bogousslavsky J, Brass LM, et al. Aspirin and clopi-
relationships among dose, effectiveness, and side effects. Chest dogrel compared with clopidogrel alone after recent ischaemic
2001; 119(1): 39-63. stroke or transient ischaemic attack in high-risk patients (MATCH):
[94] Patrono C, Coller B, FitzGerald GA, Hirsh J, Roth G. Platelet- Randomised, double-blind, placebo-controlled trial. Lancet 2004;
active drugs: The relationships among dose, effectiveness, and side 364(9431): 331-7.
effects: The seventh accp conference on antithrombotic and throm- [116] Food Drug Administration. Public health advisory: Updated safety
bolytic therapy. Chest 2004; 126(3): 234-64. information about a drug interaction between clopidogrel bisulfate
[95] Sørensen HT, Mellemkjaer L, Blot WJ, et al. Risk of upper gastro- (marketed as plavix) and omeprazole (marketed as prilosec and
intestinal bleeding associated with use of low-dose aspirin. Am J prilosec OTC), published in 2010. PublicHealthAdvi-
Gastroenterol 2000; 95(9): 2218-24.
sories/ucm190825.htm [Accessed on 30 May, 2016].
[96] Catella-Lawson F, Reilly MP, Kapoor SC, et al. Cyclooxygenase
[117] Ly V, Czosnowski L, McCluggage LK, Spinler S. Interaction be-
inhibitors and the antiplatelet effects of aspirin. N Engl J Med
tween clopidogrel and proton-pump inhibitors. Expert Rev Clin
2001; 345(25): 1809-17.
Pharmacol 2010; 3(1): 89-102.
[97] Erkan D, Merrill JT, Yazici Y, Sammaritano L, Buyon JP, Lock-
shin MD. High thrombosis rate after fetal loss in antiphospholipid [118] Xie Xj, Jiang Jb, Jiang J, Wang JA. Concurrent pulmonary throm-
syndrome: Effective prophylaxis with aspirin. Arthritis Rheum bosis with systemic embolism: A case report. J Zhejiang Uni Sci B
2001; 44(6): 1466-7. 2014; 15(9): 838-44.
[98] Hereng T, Lambert M, Hachulla E, et al. Influence of aspirin on the [119] Thai KE, Barrett W, Kossard S. Reactive angioendotheliomatosis
clinical outcomes of 103 anti-phospholipid antibodies-positive pa- in the setting of antiphospholipid syndrome. Australas J Dermatol
tients. Lupus 2008; 17(1): 11-5. 2003; 44(2): 151-5.
Thrombotic Management of Antiphospholipid Syndrome Current Vascular Pharmacology, 2017, Vol. 15, No. 4 325

[120] Adler A, Afek A, Levy Y, Keren G, George J. Antiphospholipid [141] Shapira I, Andrade D, Allen SL, Salmon JE. Brief report: Induction
antibodies from a patient with primary antiphospholipid syndrome of sustained remission in recurrent catastrophic antiphospholipid
enhance experimental atherosclerosis. Nat Clin Pract Cardiovasc syndrome via inhibition of terminal complement with eculizumab.
Med 2009; 6(3): 215-8. Arthritis Rheum 2012; 64(8): 2719-23.
[121] Middlebrooks EH, Panda M. Multiple recurrent stent thrombosis in [142] Lonze BE, Singer AL, Montgomery RA. Eculizumab and renal
a patient with coexisting clopidogrel resistance and increased anti- transplantation in a patient with caps. N Engl J Med 2010; 362(18):
cardiolipin antibodies: A case report. Case Rep Med 2010; 1744-5.
2010(2010): 1-5. [143] Zikos TA, Sokolove J, Ahuja N, Berube C. Eculizumab induces
[122] Patel PJ, Matthai Jr WH, Untereker WJ. Effective management of sustained remission in a patient with refractory primary cata-
acute coronary thrombosis in a young woman with lupus using ag- strophic antiphospholipid syndrome. J Clin Rheumatol 2015; 21(6):
gressive medical therapy. Cardiology 2011; 118(1): 45-9. 311-3.
[123] Fujieda Y, Amengual O, Watanabe T, et al. Dual antiplatelet ther- [144] Johns Hopkins University. Phase 2 study of the use of eculizumab
apy as prophylaxis of recurrent arterial thrombosis in patients with to prevent thrombosis after renal transplantation in patients with a
antiphospholipid syndrome. Arthritis Rheumatol 2012; 64(10): history of catastrophic antiphospholipid antibody syndrome
1036. (CAPS). In: Clinicaltrials.Gov [internet]. Available from:
[124] Park IC, Baek YH, Han SY, et al. Simultaneous intrahepatic and https://www.clinicaltrials.gov/ct2/show/NCT01029587. [Accessed
subgaleal hemorrhage in antiphospholipid syndrome following an- on 30 July, 2016].
ticoagulation therapy. World J Gastroenterol 2013; 19(38): 6494-9. [145] Belizna C. Hydroxychloroquine as an anti-thrombotic in antiphos-
[125] Edwards JC, Szczepaski L, Szechiski J, et al. Efficacy of B-cell- pholipid syndrome. Autoimmun Rev 2015; 14(4): 358-62.
targeted therapy with rituximab in patients with rheumatoid arthri-
[146] Rand JH, Wu XX, Quinn AS, Chen PP, Hathcock JJ, Taatjes DJ.
tis. N Engl J Med 2004; 350(25): 2572-81.
Hydroxychloroquine directly reduces the binding of antiphosphol-
[126] Rossi A, Colorio C, Puente D, Forastiero R. An update on current
ipid antibody-2-glycoprotein i complexes to phospholipid bilay-
management strategies for antiphospholipid syndrome-related
ers. Blood 2008; 112(5): 1687-95.
thrombosis. Clin Lipidol 2014; 9(1): 91-100.
[147] Espinola RG, Pierangeli SS, Ghara AE, Harris EN. Hydroxy-
[127] Erkan D, Vega J, Ramón G, Kozora E, Lockshin MD. A pilot
chloroquine reverses platelet activation induced by human igg an-
open-label phase ii trial of rituximab for non-criteria manifestations
of antiphospholipid syndrome. Arthritis Rheum 2013; 65(2): tiphospholipid antibodies. Thromb Haemost 2002; 87(3): 518-22.
464-71. [148] Rand JH, Wu XX, Quinn AS, et al. Hydroxychloroquine protects
[128] Erre GL, Pardini S, Faedda R, Passiu G. Effect of rituximab on the annexin a5 anticoagulant shield from disruption by antiphos-
clinical and laboratory features of antiphospholipid syndrome: A pholipid antibodies: Evidence for a novel effect for an old antima-
case report and a review of literature. Lupus 2008; 17(1): 50-5. larial drug. Blood 2010; 115(11): 2292-9.
[129] Ahn ER, Lander G, Bidot CJ, Jy W, Ahn YS. Long-term remission [149] Edwards MH, Pierangeli S, Liu X, Barker JH, Anderson G, Harris
from life-threatening hypercoagulable state associated with lupus EN. Hydroxychloroquine reverses thrombogenic properties of an-
anticoagulant (la) following rituximab therapy. Am J Hematol tiphospholipid antibodies in mice. Circulation 1997; 96(12):
2005; 78(2): 127-9. 4380-4.
[130] Chalam KV, Gupta SK, Agarwal S. Rituximab effectively reverses [150] Erkan D, Yazici Y, Peterson MG, Sammaritano L, Lockshin MD.
papilledema associated with cerebral venous sinus thrombosis in A cross-sectional study of clinical thrombotic risk factors and pre-
antiphospholipid antibody syndrome. Eur J Ophthalmol 2007; ventive treatments in antiphospholipid syndrome. Rheumatology
17(5): 867. 2002; 41(8): 924-9.
[131] Veneri D, Ambrosetti A, Franchini M, Mosna F, Poli G, Pizzolo G. [151] Schmidt-Tanguy A, Voswinkel J, Henrion D, et al. Antithrombotic
Remission of severe antiphospholipid syndrome associated with effects of hydroxychloroquine in primary antiphospholipid syn-
non-hodgkin's B-cell lymphoma after combined treatment with ri- drome patients. J Thromb Haemost 2013; 11(10): 1927-9.
tuximab and chemotherapy. Haematologica 2005; 90(37): 37. [152] University Hospital Rouen. Efficiency of hydroxychloroquine on
[132] Rubenstein E, Arkfeld DG, Metyas S, Shinada S, Ehresmann S, the endothelial dysfunction in antiphospholipid syndrome
Liebman HA. Rituximab treatment for resistant antiphospholipid (APLAQUINE). In: Clinicaltrials.Gov [internet]. [Available from:
syndrome. J Rheumatol 2006; 33(2): 355-7. https://www.clinicaltrials.gov/ct2/show/NCT02595346. Accessed
[133] Aguiar C, Lehman T, Erkan D. A106: Rituximab use in pediatric on 30 July, 2016].
antiphospholipid antibody positive patients. Arthritis Rheumatol [153] Prospective Studies Collaboration, Lewington S, Whitlock G, et al.
2014; 66(3): 142. Blood cholesterol and vascular mortality by age, sex, and blood
[134] Berman H, Rodríguez-Pintó I, Cervera R, et al. Rituximab use in pressure: A meta-analysis of individual data from 61 prospective
the catastrophic antiphospholipid syndrome: Descriptive analysis of studies with 55,000 vascular deaths. Lancet 2007; 370(9602):
the caps registry patients receiving rituximab. Autoimmun Rev 1829-39.
2013; 12(11): 1085-90. [154] Pai M, Evans NS, Shah SJ, Green D, Cook D, Crowther MA. Stat-
[135] Trappe R, Loew A, Thuss-Patience P, Dörken B, Riess H. Success- ins in the prevention of venous thromboembolism: A meta-analysis
ful treatment of thrombocytopenia in primary antiphospholipid an- of observational studies. Thromb Res 2011; 128(5): 422-30.
tibody syndrome with the anti-cd20 antibody rituximab-monitoring [155] Meroni PL, Raschi E, Testoni C, et al. Statins prevent endothelial
of antiphospholipid and anti-gp antibodies: A case report. Ann cell activation induced by antiphospholipid (anti-2-glycoprotein I)
Hematol 2006; 85(2): 134-5. antibodies: Effect on the proadhesive and proinflammatory pheno-
[136] O'Connor A, Murphy G, Cronin S. Antiphospholipid syndrome- type. Arthritis Rheum 2001; 44(12): 2870-8.
associated crescendo stroke events treated with rituximab. Neurol- [156] Ferrara DE, Swerlick R, Casper K, et al. Fluvastatin inhibits up-
ogy 2016; 86(16): 351. regulation of tissue factor expression by antiphospholipid antibod-
[137] Chighizola CB, Favalli EG, Meroni PL. Novel mechanisms of ies on endothelial cells. J Thromb Haemost 2004; 2(9): 1558-63.
action of the biologicals in rheumatic diseases. Clin Rev Allergy [157] Ferrara DE, Liu X, Espinola RG, et al. Inhibition of the throm-
Immunol 2014; 47(1): 6-16. bogenic and inflammatory properties of antiphospholipid antibod-
[138] Schmidtko J, Peine S, El-Housseini Y, Pascual M, Meier P. Treat- ies by fluvastatin in an in vivo animal model. Arthritis Rheum
ment of atypical hemolytic uremic syndrome and thrombotic mi-
2003; 48(11): 3272-9.
croangiopathies: A focus on eculizumab. Am J Kidney Dis 2013;
[158] Erkan D, Willis R, Murthy VL, et al. A prospective open-label
61(2): 289-99.
pilot study of fluvastatin on proinflammatory and prothrombotic
[139] Kaplan BS, Ruebner RL, Copelovitch L. Eculizumab treatment of
biomarkers in antiphospholipid antibody positive patients. Ann
atypical hemolytic uremic syndrome. Expert Opin Orphan Drugs
Rheum Dis 2014; 73(6): 1176-80.
2013; 1(2): 167-76.
[159] López-Pedrera C, Ruiz-Limón P, Aguirre MÁ, et al. Global effects
[140] Kronbichler A, Frank R, Kirschfink M, et al. Efficacy of eculizu-
mab in a patient with immunoadsorption-dependent catastrophic of fluvastatin on the prothrombotic status of patients with antiphos-
antiphospholipid syndrome: A case report. Medicine (Baltimore) pholipid syndrome. Ann Rheum Dis 2011; 70(4): 675-82.
2014; 93(26): 143.
326 Current Vascular Pharmacology, 2017, Vol. 15, No. 4 Islam et al.

[160] Canaud G, Knebelmann B, Harris PC, et al. Therapeutic mtor inhi- http://www.drugs.com/newdrugs/fda-approves-xarelto-rivaroxaban-
bition in autosomal dominant polycystic kidney disease: What is prevent-deep-vein-thrombosis-patients-undergoing-knee-hip-2747.
the appropriate serum level? Am J Transplant 2010; 10(7): 1701-6. html. [Accessed: 30 July, 2016].
[161] Walker NM, Belloli EA, Stuckey L, et al. Mechanistic target of [167] Rivaroxaban (xarelto) Prescribing Information. Janssen pharmaceu-
rapamycin complex 1 (mTORC1) and mTORC2 as key signaling ticals, inc. Titusville, nj, revised: May 2016. http://www. xarel-
intermediates in mesenchymal cell activation. J Biol Chem 2016; tohcp.com/shared/product/xarelto/prescribing-information.pdf.
291(12): 6262-71. [Accessed on 30 July, 2016].
[162] El-Agroudy A, Alarrayed S, Al-Ghareeb S, Farid E, Alhelow H, [168] FDA Approves Eliquis. FDA approves eliquis to reduce the risk of
Abdulla S. Efficacy and safety of early tacrolimus conversion to si- stroke, blood clots in patients with non-valvular atrial fibrillation,
rolimus after kidney transplantation: Long-term results of a pro- published: December 28, 2012. http://www.drugs.com/newdrugs/
spective randomized study. Ind J Nephrol 2016; 1-9. fda-approves-eliquis-reduce-risk-stroke-blood-clots-patients-non-
[163] Canaud G, Bienaimé F, Tabarin F, et al. Inhibition of the mtorc valvular-atrial-fibrillation-3618.html. [Accessed on 30 July, 2016].
pathway in the antiphospholipid syndrome. N Engl J Med 2014; [169] Apixaban (eliquis) Prescribing Information. Bristol-myers squibb,
371(4): 303-12. princeton nj and pfizer inc. New york, ny, revised 2015.
[164] Islam MA, Alam F, Sasongko TH, et al. Antiplatelet and antico- http://packageinserts.bms.com/pi/pi_eliquis.pdf. [Accessded on 30
agulant agents for preventing recurrence of peripheral vascular July, 2016].
thrombosis in patients with antiphospholipid syndrome. Cochrane [170] FDA Approves Pradaxa. FDA approves pradaxa to prevent stroke
Database Syst Rev 2016; CD012198(5): 1-19. in people with atrial fibrillation, published: October 19, 2010.
[165] Islam MA, Alam F, Khalil MI, Sasongko TH, Gan SH. Natural http://www.drugs.com/newdrugs/fda-approves-pradaxa-prevent-
products towards the discovery of potential future antithrombotic stroke-atrial-fibrillation-2370.html. [Accessed on 30 July, 2016].
drugs. Curr Pharm Des 2016; 22(20): 2926-46. [171] Dabigatran (pradaxa) Prescribing Information. Boehringer ingel-
[166] FDA Approves Xarelto. Fda approves xarelto (rivaroxaban) to heim pharmaceuticals, inc. Ridgefield, ct, revised april 2013.
prevent deep vein thrombosis in patients undergoing knee or hip http://docs.boehringer-ingelheim.com/Prescribing%20Information/
replacement surgery, published: July 1, 2011. PIs/Pradaxa/ Pradaxa.pdf. [Accessed on 30 July, 2016].

You might also like