Sunteți pe pagina 1din 12

Clinic Rev Allerg Immunol

DOI 10.1007/s12016-016-8571-6

Additional Treatments for High-Risk Obstetric Antiphospholipid


Syndrome: a Comprehensive Review
Amelia Ruffatti 1,2 & Ariela Hoxha 1 & Maria Favaro 1 & Marta Tonello 1 & Anna Colpo 3 &
Umberto Cucchini 4 & Alessandra Banzato 4 & Vittorio Pengo 4

# Springer Science+Business Media New York 2016

Abstract Most investigators currently advocate prophylactic- Keywords Obstetric antiphospholipid syndrome .
dose heparin plus low-dose aspirin as the preferred treatment Treatment . Risk factor . Additional treatment
of otherwise healthy women with obstetric antiphospholipid
syndrome, whilst women with a history of vascular thrombo-
sis alone or associated with pregnancy morbidity are usually Introduction
treated with therapeutic heparin doses in association with low-
dose aspirin in an attempt to prevent both thrombosis and In 1983, the connection between pregnancy loss and the pres-
pregnancy morbidity. However, the protocols outlined above ence in the blood of anticardiolipin (aCL) antibodies was
fail in about 20 % of pregnant women with antiphospholipid made by Graham Hughes [1]. Since then, recurrent pregnancy
syndrome. Identifying risk factors associated with pregnancy loss has been considered as a clinical manifestation of
failure when conventional therapies are utilized is an impor- antiphospholipid syndrome (APS), and the Sydney update of
tant step in establishing guidelines to manage these high-risk the preliminary classification criteria of APS confirmed preg-
patients. Some clinical and laboratory risk factors have been nancy morbidity as a valuable clinical criterion for APS clas-
found to be related to maternal–foetal complications in preg- sification [2]. More specifically, APS pregnancy morbidity
nant women on conventional therapy. However, the most effi- includes three types of pregnancy complications: (a) one or
cacious treatments to administer to high-risk antiphospholipid more unexplained deaths of a morphologically normal foetus
syndrome women in addition to conventional therapy in order at or beyond the 10th week of gestation; or (b) one or more
to avoid pregnancy complications are as yet unestablished. This premature births of a morphologically normal neonate before
is a comprehensive review on this topic and an invitation to the 34th week of gestation because of eclampsia or severe
participate in a multicentre study in order to identify the best preeclampsia, or recognized features of placental insufficien-
additional treatments to be used in this subset of antiphospholipid cy; or (c) three or more unexplained consecutive spontaneous
syndrome patients. abortions before the 10th week of gestation, with maternal
anatomic or hormonal abnormalities and paternal and mater-
nal chromosomal causes excluded.
The hypotheses on the pathogenesis of lesions in APS
* Amelia Ruffatti pregnancy morbidity include the thrombogenic effect of
amelia.ruffatti@unipd.it
antiphospholipid antibodies which may cause slow progres-
sive thrombosis and infarction of the placenta. Since
1
Rheumatology Unit, Department of Medicine, University of Padua, antiphospholipid antibodies are associated with systemic
Padua, Italy thrombosis, obstetric complications in patients with
2
Reumatologia, Policlinico Universitario, Via Giustiniani, antiphospholipid antibodies were first attributed to thrombosis
2-35128 Padova, Italy and infarction at the maternal–foetal interface; indeed, early
3
Blood Transfusion Unit, University-Hospital of Padua, Padua, Italy reports of placenta from women with the lupus anticoagulant
4
Cardiology Unit, Department of Cardio-Thoracic and Vascular (LAC) exhibited ischemic infarcts [3]. However, large-scale
Sciences, University of Padua, Padua, Italy histological studies have then revealed that neither thrombosis
Clinic Rev Allerg Immunol

nor infarction is so common in the placenta of patients with findings from well-designed studies [10]. However, the prog-
APS [3]. Non-thrombotic mechanisms can be identified on nosis of pregnancies in patients with APS has greatly im-
both the maternal and foetal sides, causing the functional im- proved over the last two decades. Based on the results of some
pairment of decidua and trophoblast cells, respectively. In fact, trials [11–13] and in line with two meta-analyses [14, 15],
defective placentation may be mediated by the inhibition of most investigators currently advocate prophylactic-dose low-
endometrial angiogenesis and by the reduction of trophoblast molecular-weight heparin plus low-dose aspirin (LDA) as the
differentiation/invasiveness [4]. Furthermore, treatment of tro- preferred treatment of otherwise healthy women with obstetric
phoblast with antiphospholipid antibodies decreases the ex- APS, even if other authors found in this type of patient no
pression of matrix metalloproteinases and increases the ex- significant difference between treatment with low-molecular-
pression of inhibitors of metalloproteinases [4]. Combined, weight heparin plus low-dose aspirin and that with low-dose
these changes are likely to contribute to the decreased inva- aspirin alone [16]. Although specific clinical trials are lacking,
sion observed in trophoblasts treated with antiphospholipid women with a history of vascular thrombosis alone or associ-
antibodies. Moreover, it was found that complement- ated with pregnancy morbidity are usually treated with thera-
mediated placental inflammation plays a key role in an exper- peutic low-molecular-weight heparin doses in association
imental model of foetal loss [5]. There is strong evidence from with LDA in an attempt to prevent both thrombosis and preg-
murine models of APS that complement plays a crucial role in nancy morbidity.
mediating foetal demise in murine antiphospholipid antibody- The protocols outlined above fail in about 20 % of pregnant
affected pregnancies. The complement proteins C3 and C5 APS women [17], and additional treatments including intra-
have been reported to be crucial for mediating antiphospholipid venous immunoglobulins (IVIG) [18–28] (Table 1), low-dose
antibody-induced thrombosis in mice. Placenta from women prednisolone [29], or aphaeresis procedures such as plasma
with APS exhibit increased deposition of C4d and C3b, which exchange or immunoadsorption [30–37] have, at times, been
supports the hypothesis that complement activation may be combined with conventional therapies (Table 2). Due to the
involved in the pathogenesis of pregnancy complications in low number of treated cases, the efficacy of these protocols is
women with antiphospholipid antibodies [5]. However, there at present not well established.
are considerable differences in the control of the complement
system at the maternal–foetal interface between mice and wom- High-Risk Obstetric APS
en, and additional human studies are required before a defini-
tive conclusion regarding the role of complement in obstetric Identifying risk factors associated with pregnancy failure
APS can be made [6]. when conventional therapies are utilized is an important step
On the basis of these pathogenetic hypotheses, heparin and in establishing guidelines to manage these high-risk patients.
aspirin combination constitutes the most frequently applied Logically, the choice of treatment in pregnant patients with
treatment protocol in pregnant APS women. What is the ra- APS should be done on the basis of the degree of obstetric
tionale for the use of these drugs? According to the sugges- risk. But actually, maternal–foetal risk in APS pregnant wom-
tions in the literature, heparin, in addition to a direct effect on en has not yet been well defined.
the coagulation cascade, might protect pregnancy by reducing The consensus statement of the classification criteria for
the binding of antiphospholipid antibodies to trophoblast, re- definite APS [2] introduced the concept of stratification of
ducing inflammation, facilitating implantation and inhibiting the risk in APS patients on the basis of laboratory and clinical
complement activation [7]. Low-dose aspirin induces throm- characteristics. In particular, patients were allocated to classi-
boxane reduction not only at the platelet level but also at the fication categories on the basis of positivity to more than one
placental level. In vitro, aspirin suppresses the thromboxane test—category I—or to a single test—categories IIA (LAC
release of the normal placenta in the presence of LAC-positive alone), IIB (aCL antibodies alone) and IIC (anti-β2-glycopro-
IgG [8]. It increases the production of leukotrienes, which act tein I [anti-β2GPI] antibodies alone).
in stimulating interleukin-3 production. Interleukin-3 en- Concerning obstetric APS, several studies have attempted
hances placental and foetal development and was found in to identify variables predictive of complications during con-
low amounts in APS patients. Low-dose aspirin restores ventionally treated pregnancies (Table 3). Some investigators
interleukin-3 levels in animal models of APS [9]. Moreover, found a higher risk of maternal and foetal complications in
its suppression of nitric oxide synthetase activity prevents ox- pregnant APS women with previous thromboembolism [38],
idative stress able to induce trophoblast apoptosis. In fact, the and others reported the association of pregnancy loss with the
increased apoptosis of a trophoblastic cell line (BeWo), in- presence of multiple antiphospholipid positivity, i.e., for
duced by the serum of patients with early miscarriages, is anti-β2GPI antibodies and LAC [39, 40]. A history of throm-
significantly reduced by aspirin [7]. bosis [41, 42], some particular antiphospholipid antibody pro-
The treatment of pregnancy morbidity related to files such as the presence of LAC [43] or high titres of aCL or
antiphospholipid antibodies is not supported by consistent anti-β2GPI antibodies [44] and APS associated with systemic
Clinic Rev Allerg Immunol

Table 1 Effect of IVIG administered in addition to conventional therapy in high-risk pregnancies of patients with antiphospholipid syndrome

Authors Year Clinical IVIG protocols Outcome


(reference) reports

Carreras et al. [78] 1988 Case report 400 mg kg−1 day−1 for 5 days started at week 17 and followed by 2-day Favourable
courses at 22 and 27 week
Parke et al. [18] 1989 Case report 300 mg kg−1 month−1 for 6 months (before conception); 4-day induction of Favourable
400 mg kg−1 day−1 (after conception) continued at 600 mg day−1 month−1
Wapner et al. [19] 1989 Case reports Pt 1: 1 g kg−1 month−1 from week 9 to 34 weeks Favourable
Pt 2: 1 g kg−1 month−1 from week 10 to 33 weeks
Ron-El et al. [20] 1993 Case report 400 mg kg−1 month−1 (during the pregnancy) Favourable
Kaaja et al. [21] 1993 Case series 1 g kg−1 month−1 Live birth rate 100 %,
1 case of preeclampsia
Arnout et al. [22] 1994 Case report 400 mg/kg for 5 days with a 4-week interval (during the pregnancy for Live birth and placental
6 months) abruption
Somerset et al. 1998 Case report 500 mg kg−1 day−1 for 4 days at weeks 23 and 27 Favourable
[23]
Branch et al. [24] 2000 Multicenter, Group I: LDA (81 mg/day) and unfractionated heparin (7500 U) every 12 h, Fewer cases of IUGR and
placebo- increased to 10,000 U/12 h in the second trimester + IVIG: 1 g kg−1 day−1 neonatal intensive care
controlled for 2 days every month through 36 weeks unit admissions amongst
pilot study Group II: LDA and heparin as above + placebo the IVIG-treated cases
Stojanovich 2007 Case report 5 g kg−1 month−1 for 9 times during the pregnancy Favourable
et al. [25]
Mar et al. [26] 2014 Case report 0.5 mg kg−1 month−1 from week 8 (7 total doses) Favourable
Watanabe et al. 2014 Case series 400 mg kg−1 day−1 for 5 days from week 6 or 7 Live birth rate 100 %
[27]
Rose et al. [28] 2014 Case report 1 g kg−1 week−1 from week 9 IUGR, stillbirth at
21 weeks

IVIG intravenous immunoglobulins, LDA low-dose aspirin, IUGR intrauterine growth restriction

lupus erythematosus or other systemic autoimmune diseases component levels have, moreover, been found to be indepen-
[43, 45] have been found to be related to maternal–foetal dent predictors of lower neonatal birth weight and of birth at
complications in pregnant APS women. The persistence of an earlier gestational age, whilst a false-positive IgM TORCH
uterine artery notching and lower complement C3 and C4 (Toxoplasma, rubella, cytomegalovirus and herpes simplex

Table 2 Effect of apheresis procedures administered in addition to other treatments in high-risk pregnancies of patients with antiphospholipid
syndrome

Authors (reference) Year Clinical reports Treatment protocols Outcome

Frampton et al. [30] 1987 Case report Plasma exchange, low-dose aspirin, dipyridamole and low-dose prednisolone Favourable
Kobayashi et al. 1992 Case reports Immunoadsorption, low-dose aspirin and low-dose prednisolone Favourable
[31]
Nakamura et al. 1999 Case series Immunoadsorption, low-dose aspirin and low-dose prednisolone Live birth rate 7/8
[32] (87.5 %)
El-Haieg et al. [33] 2007 Case series Plasma exchange and low-dose prednisone Live birth rate 18/18
(100 %)
Ruffatti et al. [34] 2007 Case series Plasma exchange, therapeutic heparin ± low-dose aspirin; in two cases + IVIG Live birth rate 5/7
(71.4 %)
Bortolati et al. [35] 2009 Case reports Pt 1: Plasma exchange plus IVIG, therapeutic heparin and low-dose aspirin Favourable
Pt 2: Immunoadsorption plus IVIG, therapeutic heparin and low-dose aspirin
Rose et al. [28] 2014 Case reports Plasma exchange, therapeutic heparin, low-dose aspirin, HCQ, low-dose Live birth, preeclampsia
prednisolone and azathioprine
Mayer-Pickel et al. 2015 Case report Plasma exchange, therapeutic heparin and low-dose aspirin Favourable
[36]
Ruffatti et al. [37] 2016 Case series Plasma exchange plus IVIG, therapeutic heparin and low-dose aspirin Live birth rate 17/18
(94.4 %)

IVIG intravenous immunoglobulins, HCQ hydroxychloroquine


Clinic Rev Allerg Immunol

Table 3 Risk factors for pregnancy failure on conventional treatment in patients with antiphospholipid syndrome

Authors (reference) Year Risk factors

Lima et al. [38] 1996 Previous thromboembolism


Ruffatti et al. [69] 2006 Previous thromboembolism and/or triple antiphospholipid antibody positivity
Sailer et al. [39] 2006 Multiple antiphospholipid antibody positivity
Danowski et al. [45] 2009 Association with SLE or other SAD
Nodler et al. [53] 2009 High antiphospholipid antibody titres
Ruffatti et al. [70] 2009 Antiphospholipid antibody category I (Sydney criteria) or triple antiphospholipid antibody positivity
Bramham et al. [41] 2010 Previous thrombosis
Ruffatti et al. [71] 2011 Association with SLE or other SAD or previous thrombosis and pregnancy morbidity or triple antiphospholipid positivity
Simchen et al. [44] 2011 High titres of anticardiolipin or anti-β2 glycoprotein I antibodies
Fischer-Betz et al. [42] 2012 Previous ischaemic cerebral events and multiple antiphospholipid antibody tests
Lockshin et al. [43] 2012 Lupus anticoagulant positivity or association with SLE or other SAD
de Jesus et al. [54] 2014 Previous thrombosis
Matsuki Y et al. [40] 2015 Multiple antiphospholipid antibody positivity

SLE systemic lupus erythematosus, SAD systemic autoimmune diseases

virus tests) was an independent predictor of lower neonatal study conducted in Florida on 141,286 women who delivered
birth weight [46]. However, a subsequent study failed to re- in 2001 showed that women with high antiphospholipid anti-
port any association between hypocomplementemia and ob- body titres had an increased risk of preeclampsia or eclampsia
stetric complications [47]. (adjusted odds ratio [AOR] = 2.93), placenta insufficiency
Conventional treatment of patients with APS during preg- (AOR = 4.58) and prolonged length of stay at hospital
nancy can significantly improve live birth rates, so reaching (>3 days, AOR = 3.93) [53]. Moreover, APS patients with
more than 70 % of live birth rate [48], but other complications history of cerebral ischemic events are at increased risk of
remain high despite treatment. Disorders linked to APS during preeclampsia (34.8 %), especially those positive for multiple
pregnancy include thrombotic events as well as specific ob- antiphospholipid antibody test, and new cerebral ischemia can
stetrical complications. The coexistence of both thrombosis occur if preeclampsia develops (OR = 7.0) [42]. Considering
and miscarriage is estimated at 2.5–5 % of APS pregnancies patients with definite APS, there are no randomized controlled
[49]. trials that included patients with exclusively severe pre-
Thrombotic events are major problems during pregnancy eclampsia or eclampsia and premature birth before the 34th
because of the management they implicate and the risk of week, as stated by the classification criteria [2]. Therefore, the
complications, such as pulmonary embolism. Interestingly, recommended treatment for this group of patients (LDA plus
the Nimes Obstetricians and Hematologists APS (NOH- heparin) is the same as for patients with pregnancy loss; how-
APS) observational study compared the incidence of throm- ever, its real efficacy is unknown. In fact, preeclampsia is still
botic events in 517 women with purely obstetrical APS to 796 common in patients with APS receiving LDA and heparin,
seronegative women with a history of pregnancy loss. The resulting in significant maternal and foetal morbidity in these
annual rate of thrombotic complications, defined by deep vein pregnancies. Clinicians should be aware of obstetric APS-
thrombosis (1.46 %), pulmonary embolism (0.43 %), superfi- related events even in patients with history of only thrombotic
cial vein thrombosis (0.44 %) and cerebrovascular events manifestations of the syndrome [54].
(0.32 %), was found to be higher in obstetrical APS women There is another severe pregnancy complication in preg-
than in control patients (0.43, 0.12, 0.14 and 0.09 %, respec- nant APS women. It is characterized by hemolysis, elevated
tively) [50]. The most serious thrombotic disorder also affect- liver enzymes and low platelet count (HELLP syndrome)
ing APS patients during pregnancy or puerperium is cata- [55]. Several case reports have described the association be-
strophic APS (CAPS). CAPS represents 1 % of APS and tween antiphospholipid antibodies/APS and HELLP syn-
can occur inside or outside of pregnancy. CAPS is defined drome [55]. In two case series, the authors concluded that
as a Bthrombotic storm^ secondary to microangiopathic dif- antiphospholipid antibodies were associated with increased
fuse thrombosis leading to multi-organ failure. Six per cent of prevalence and severity of HELLP syndrome [56, 57].
CAPS seems to be associated with pregnancy and postpartum, Moreover, HELLP/CAPS overlap has also been reported in
but this is probably underestimated [51]. More specific obstet- both case reports [58–60] and case series [51]. HELLP syn-
rical manifestations include severe preeclampsia, which gen- drome is defined as an endothelium-based disorder [61, 62].
erally affects 2–8 % of pregnancies [52]. A cross-sectional The increased prevalence of HELLP syndrome suggested by
Clinic Rev Allerg Immunol

some authors in the setting of APS can be explained in part by In a second study, we related laboratory classification cat-
the interplay between antiphospholipid antibodies and endo- egories with pregnancy outcome in patients with primary APS
thelial cells, leading to thrombosis and microangiopathic man- prescribed antithrombotic therapy. Ninety-seven pregnancies
ifestations [56, 57, 61]. of 79 APS women affected with pregnancy morbidity and/or
As well as preeclampsia, intrauterine growth restriction thromboembolism were followed from December 1995 to
(IUGR) is commonly found in pregnant women with APS, June 2007. According to their laboratory characteristics, the
often associated to oligohydramnios with an amniotic fluid patients have been divided into categories I, IIA, IIB and IIC.
index of 5 cm or less. Both disorders represent different signs The Kaplan–Meier survival curves showed the cumulative
of placental insufficiency and can be found in the same pa- proportion of unsuccessful pregnancies in triple-positive,
tient. Isolated foetal impairment, however, is possible and may double-positive or single-positive women. Triple-positive pa-
develop with absent or minimal maternal signs, like small tients had a significantly higher cumulative proportion of un-
uterine height [63]. Some investigators found almost 40 % successful pregnancies than double-positive women (p =
of small for gestational age neonates (below the 10th percen- 0.03). Thus, we concluded that poor pregnancy outcomes oc-
tile) in patients with thrombotic APS compared to 16 % of cur more frequently in category I than in category II primary
patients with obstetric APS [41]. Other authors reported APS patients. However, it has been seen that a greater predict-
28.6 % of small for gestational age in pregnancies after cere- ability is achieved when category I patients are grouped into
bral ischemic events related to antiphospholipid antibodies triple and double positivity states. Triple-positive patients
[42]. A paper evaluating the efficacy of prophylaxis using might be a specific and separated category of patients at very
LDA and heparin in the prevention of IUGR in patients with high risk of pregnancy loss [70].
APS provided negative results; of the APS patients, 32.3 % In this context, a case–control study drawing on a large
had low birth weight newborns (below the 10th percentile) multicentre cohort of conventionally treated pregnancies was
compared to 2.5 % of the control group [64]. Once again, conducted to verify whether specific laboratory profiles and/or
treatment for this presentation of APS is disappointing, with clinical characteristics are predictive of unsuccessful pregnan-
a higher frequency of IUGR in APS patients. Premature birth cy outcome during conventional treatments. We retrospective-
is reported in 20–25 % of APS patients (range = 11–66 % ly considered 410 pregnancies of women diagnosed with
based on different studies) [65–68], mainly caused by preterm APS. The study focused on 57 unsuccessful pregnancies (the
delivery due to preeclampsia, HELLP syndrome or IUGR. study population) and 57 successful pregnancies (the control
Some authors described higher rates of preterm delivery in population) matched for age and therapy. All the pregnant
patients with thrombotic APS when compared to women with patients were treated with conventional protocol treatments
history of recurrent abortion only [41]. including LDA and/or heparin. The clinical and laboratory
In an attempt to evaluate the influence of clinical and lab- features of the two groups of APS women were statistically
oratory maternal features on pregnancy outcome, we studied compared. The most statistically significant risk factors for
the pregnancies of 53 APS women with pregnancy morbidity pregnancy failure were: (a) association of APS with systemic
during a mean follow-up period of 6.3 years. The study group lupus erythematosus or other systemic autoimmune diseases
included only APS women in laboratory classification catego- (OR = 6.9, CI = 2.7–17.8); (b) a history of both thrombosis
ry I. Obstetric history included foetal loss in 72 % of cases, and pregnancy morbidity (OR = 12.7, CI = 2.8–57.9); and (c)
spontaneous abortions in 22 % and preeclampsia or eclampsia triple (LAC plus aCL plus anti-β2GPI antibodies)
in 6 %. Laboratory diagnosis showed double antiphospholipid antiphospholipid antibody positivity (OR = 9.2, CI = 2.8–
antibody positivity in 70 % of cases and triple positivity in 30.9; Fig. 1). The probability of pregnancy failure estimated
30 %. Previous thromboembolism was found in 30 % of the by logistic regression analysis was 93 % in APS women with
women (6 % pulmonary embolism, 9 % deep vein thrombosis two or more of these features, 63 % in those with only one
and 15 % stroke). The results of the statistical analysis dem- feature and 21 % in those with none. It was seen at univariate
onstrated that only previous thromboembolism and triple analysis that APS patients diagnosed on the basis of a single-
antiphospholipid antibody positivity were significantly asso- positive test and/or history of pregnancy morbidity alone gen-
ciated with a new unsuccessful pregnancy. Moreover, only the erally had successful pregnancies (Fig. 1). It would seem from
same two parameters were significantly associated with a new these findings that the risk of pregnancy complications in APS
thromboembolism during the next pregnancy. In this study, women planning to conceive can be stratified on the basis of
we concluded that in primary APS with pregnancy morbidity some specific clinical and laboratory features [71].
in classification category I, quite different groups of patients
may be identified on the basis of laboratory tests and clinical Additional Treatments in High-Risk Obstetric APS
characteristics. Triple antibody positivity and/or a history of
thromboembolism predict new unsuccessful pregnancy and For the moment, there are no guidelines on the ideal additional
new thromboembolism [69]. treatment strategy in APS patients at high risk of pregnancy
Clinic Rev Allerg Immunol

Fig. 1 Pregnancy outcome in antiphospholipid syndrome women on features. aOn the basis of univariate analysis. bOn the basis of logistic
conventional therapy. Stratification of the risk of pregnancy regression analysis. aPL antiphospholipid antibodies, SLE systemic lupus
complications on the basis of some specific clinical and laboratory erythematosus, SAD systemic autoimmune diseases

failure/complications during conventional treatment, probably of IVIG in APS may be due to the presence of anti-idiotypic
because of the rarity of this disorder. antibodies to antiphospholipid antibodies in IVIG or the inac-
Whilst steroids and aphaeresis procedures are currently tivation of B cell clones, leading to subsequently decreased
considered additional therapies [72, 73], data on the useful- autoantibody production [77]. In addition, IVIG infusion was
ness of IVIG in obstetric APS are conflicting [18–28, 74–76]. shown to prevent obstetric complications in APS animal
The use of IVIG in pregnant APS women is rare and is gen- models, possibly preventing the binding of antiphospholipid
erally considered a second-line treatment when other more antibodies to the trophoblast [84]. It is interesting that IVIG
standardized therapies have failed [77]. The first description resulted beneficial in most cases [18–27, 78] when adminis-
of a case of obstetric APS patient treated with IVIG was that tered in addition to the first-line therapy, but it had only a
by Carreras et al. in 1988 [78]. Treatment with IVIG at the slight [74] or no effect at all [75, 76] when used alone as the
standard dose of 400 mg kg−1 day−1 for 5 days started at first-line treatment. However, as the literature shows hetero-
week 17 and followed by 2-day courses at 22 and 27 weeks geneous data with respect to IVIG dosing, timing of adminis-
of gestation in a 28-year-old woman who suffered from nine tration and concomitant treatment used (heparin, aspirin or
early spontaneous abortions, two intrauterine deaths and a steroids), it is difficult to draw well definite conclusions about
perinatal death at 26 weeks. The patient delivered by caesar- IVIG benefits in obstetric APS. In general, IVIG therapy is
ean section a healthy girl at week 34. IVIG are blood products well tolerated and adverse reactions are usually mild and re-
prepared from the serum of a large number of donors, and versible [85]. The use of IVIG infusion is, in any case, limited
their use has rapidly grown to treat a wide variety of autoim- because of its high cost and short supply.
mune diseases [79]. The mechanisms in which IVIG exert As complement system activation and inflammatory
their immunomodulatory and anti-inflammatory effects re- changes seem to induce disorders in trophoblast invasion
main unclear, with many pathways in the innate and adaptive and placentation, it is possible that steroids are beneficial in
immune systems being potentially targeted. The many expla- APS women with recurrent pregnancy loss [86]. Whilst high
nations for these actions include modulation of Fc receptor doses of prednisolone (40–60 mg/day) administered in com-
expression on leukocytes and endothelial cells, interaction bination with LDA have been associated with too many high
with complement proteins, modulation of the synthesis and rates of pregnancy complications [86], low doses of prednis-
release of cytokines and chemokines, modulation of cell pro- olone (10 mg/day) combined with heparin/aspirin from the
liferation and apoptosis, remyelinization, neutralization of cir- time a pregnancy test resulted positive until the 14th week of
culating antibodies, selection of immune repertoires, interac- gestation produced encouraging results in APS women with
tion with other cell surface molecules expressed on lympho- previous pregnancy morbidity, but not in those also suffering
cytes and monocytes, modulation of dendritic cell maturation from previous thromboembolism and triple antiphospholipid
and restoration of idiotypic–anti-idiotypic networks [80, 81]. antibody positivity [29].
Particularly in APS, IVIG have been shown to inhibit both The safety of both plasma exchange or immunoadsorption
the action of antiphospholipid antibodies and their production procedures during pregnancy is well documented [30–37]. In
[79]. Indeed, some investigators [82] reported that F(ab′)2 order to avoid bleeding during plasmapheresis treatment, as
fragments from IVIG can neutralize the binding of aCL anti- plasmapheresis temporarily removes blood clotting factors, in
bodies to cardiolipin in a dose-dependent manner and other heparin-treated patients, it is important that the morning hep-
authors [83] that the same fragment can also inhibit LAC arin, performed after the session, was halved [37]. Both extra-
activity. The mechanisms responsible for the beneficial effects corporeal blood purification techniques are known to lower
Clinic Rev Allerg Immunol

antibody levels in APS patients [87, 88]. Their use in pregnant into a more effective treatment. On the other hand, plasma
APS patients has, nevertheless, been circumscribed, and de- exchange removal of activated complement components, coag-
scriptions in the literature are limited to case reports [28, 30, ulation factors and microparticles may also be relevant. The use
31, 35, 36] and case series [32–34, 37]. Both aphaeresis pro- of immunoadsorption procedure is, in any case, limited because
cedures have, in any case, always been administered in addi- of its very high cost and long duration of treatment. Thus,
tion to other treatments, including steroids, heparin/LDA or according to our experience, the use of immunoadsorption
IVIG, and have been associated to a live birth rate ranging technique could be considered in high-risk APS pregnancies
between 71.4 and 100 % of the treated cases [33, 34, 37]. with IgG antiphospholipid antibody isotype as a treatment
During the first period in which plasma exchange was used when a previous pregnancy failed on plasma exchange or as
by us, several questions were raised [34]. More precisely, it has substitutive treatment of plasma exchange at early occurrence
been seen that the replacement fluid used during therapeutic of pregnancy complications [37, 90]. For both plasma ex-
plasma exchange can be an important factor influencing its change and immunoadsorption, it is important to use only sub-
results. By evaluating the pathophysiology of the underlying cutaneous arm veins as blood access points in order to avoid
disease, it is possible to choose different options, including 4 % catheter-related complications.
albumin solution and fresh frozen plasma. In one of our patient, Whilst the true relevance of these treatments has still to be
suffering from catastrophic APS soon after delivery, the infu- confirmed, other suggestions related to experimental models
sion of fresh frozen plasma containing amounts of coagulation are rising. Amongst these, hydroxychloroquine might be the
factors was associated with severe thrombotic microangiopa- first drug to be largely administered in addition to convention-
thy. This disorder immediately disappeared when plasma ex- al treatment because of its well-documented safety in pregnant
change was administered using 4 % albumin in saline as the patients [91]. In vitro studies indicate that hydroxychloroquine
replacement fluid. The temporary reduction of coagulation pa- may be beneficial to APS patients. In fact, hydroxychloroquine
rameters that was observed after albumin infusion may have was able to reduce TLR4 mRNA and protein expression and
benefited this thrombophilic APS patient [89]. On the other to finally restore trophoblast function, suggesting the use
hand, the deficiency in natural anticoagulants related to albu- of this drug in obstetric APS [92]. Some authors [93]
min infusion was balanced by the administration of antithrom- showed that hydroxychloroquine can dissociate IgG
bin III at the end of the plasma exchange session. The best antiphospholipid–β2GP1 complexes and reduce the amount
timing for beginning plasma exchange was an important con- of IgG antiphospholipid antibodies that binds to phospho-
sideration for us as we treated these women. It has been seen lipid bilayers trapping annexin A5, a potent natural antico-
that plasma exchange was completely ineffective in the first agulant with high affinity for anionic phospholipids.
treated patient, when it was begun in the presence of late Annexin A5 is required for the maintenance of placental
foetal-placental damage. Plasma exchange treatment in high- integrity in mice and plays a thrombomodulatory role at the
risk pregnancies should be initiated in the presence of early maternal–foetal interface within the placentary blood cir-
complications including bilateral notching or a gradual platelet culation by shielding the apical membrane phospholipids
fall (>20 %) occurring during the first trimester, or possibly of placental villous syncytiotrophoblasts. The reversion of
when foetal heart rate is documented and before complications IgG antiphospholipid–β2GPI complexes mediated by
occur, so as to remove the offending antiphospholipid antibod- hydroxychloroquine permits more annexin A5 to crystal-
ies and thus avoid its injuries. Due to the after delivery maternal lize and may also ameliorate some of the other proposed
microangiopathy, plasma exchange could be restarted after de- thrombogenic effects. The same authors [93] also found
livery. The high probability of bleeding when plasma exchange that hydroxychloroquine promotes the formation of second
is administered soon after surgery, i.e. caesarean section, was layers of annexin A5 crystal Bpatches^ over areas where
found by us when we were treating the first patient. In fact, the mentioned immune complexes had disrupted crystalli-
plasma exchange was begun again in this patient a few hours zation, thereby further reducing the exposure of
after caesarean section, and almost immediately, a life- thrombogenic phospholipids. However, to the best of our
threatening subfascial hematoma took place. The other three knowledge, there is only a retrospective study concerning
patients who instead began plasma exchange again several days the use of hydroxychloroquine in obstetric APS [94].
after surgery had no bleeding [34]. The advantage of Prospective randomized trials should be designed to eval-
immunoadsorption as compared to plasma exchange would uate its efficacy in patients with obstetric APS as additional
be the more selective removal of large amounts of IgG antibod- therapy of the current treatment with heparin and LDA.
ies without significantly affecting other plasma components Recently, a large multicentre, observational, retrospective
such as hormones, natural anticoagulants, cytokines and pro- study from a European Forum on antiphospholipid antibody
teins whose lack might have bad influence on the course of project was undertaken to investigate the effect of various
pregnancy. Moreover, immunoadsorption actually treats a larg- treatments on pregnancy outcomes in APS women with di-
er plasma volume than plasma exchange, which might transfer verse risk factors for pregnancy failure. Between 1996 and
Clinic Rev Allerg Immunol

2011, 196 pregnancies of 156 APS women attending 16 generally occur in high-risk pregnant APS women despite
European centres were retrospectively considered. The conventional treatment [41, 42].
study’s inclusion criteria were one or more of the following Given that the rarity of high-risk APS pregnancies does not
risk factors: a history of thrombosis, systemic lupus erythema- allow performing clinical trials, thus, for the time being, a new
tosus and triple antiphospholipid antibody positivity. The fol- multicentre, retrospective study on the framework of the
lowing treatments were considered in the study: LDA alone, European Forum on antiphospholipid antibodies focusing on
prophylactic dose of heparin + LDA, therapeutic dose of hep- the efficacy and safety of the different additional treatments in
arin + LDA and additional treatments including IVIG infu- high-risk pregnant APS women is ongoing in order to identify
sions, weekly aphaeresis procedures and low-dose predniso- the best treatments to be used in addition to conventional
lone (10 mg/day), alone or combined. The primary outcome of therapy in this subset of patients.
the study was the live birth rate. Seventy-six per cent of preg-
nancies ended successfully, producing 150 live infants, in- Study Design
cluding one set of twins. Mean week of gestation at delivery,
mean birth weight in percentiles and mean Apgar score at Only pregnant patients who fulfilled—at the time they were
5 min were quite good. Pregnancy loss took place in 24 % diagnosed with primary APS—the clinical and laboratory
of cases. Maternal–foetal complications were noted in 25 % of classification criteria established by an International
cases. It is interesting to observe that no important side effects Consensus in 2004 [2] are retrospectively enrolled in the
were recorded during all types of treatments. The primary study.
outcome, i.e. live birth rate, was analysed in relation to patient Both the following two inclusion criteria have to be present
risk profiles and treatment strategies. The only risk profile in at the time of the considered pregnancy:
which a significant difference was found between treatments
was the thrombosis plus triple antiphospholipid antibody pos- 1. Laboratory criteria: (a) positivity to all three antiphospholipid
itivity group. Backward conditional logistic regression, ad- antibody assays, referred to LAC plus aCL plus a-β2GPI
justed for potential confounding risk factors, analysed all the antibodies, or (b) positivity to LAC alone or associated either
treatments in this profile and found additional treatments as to aCL or to anti-ß2GPI antibodies
the only independent factor associated with a favourable out- 2. Clinical criteria: (a) a history of maternal thrombosis and/
come of pregnancy (OR = 9.7, CI = 1.1–88.9, p < 0.05). We or (b) one or more of the following severe obstetric com-
can conclude that APS patients with thrombosis and triple plications: eclampsia, preeclampsia (arterial pressure
antiphospholipid antibody positivity could be treated during ≥160/110 and proteinuria ≥5 g in a 24-h urine sample),
pregnancy with a second-line therapy in addition to conven- HELLP syndrome, intrauterine growth restriction (birth
tional treatment as a higher live birth rate on additional therapy weight <10th percentile), prematurity (delivery <34th
rather than on conventional therapy alone has been demon- week of gestation) and/or (c) an unexplained foetal death
strated in this subset of patients. However, in this study, it was at or beyond the 10th week of gestation during conven-
impossible to examine the additional therapies singularly and tional treatment
to identify the benefits and limits of different additional treat-
ment strategies and indicate which is associated with the best Each patient to be included in the study must have at least
pregnancy outcome as the number of patients studied was so one laboratory criterion in combination with at least one clin-
small [95]. ical criterion.
All the following exclusion criteria had to be absent at the
Future Direction time of the considered pregnancy:

According to the above-mentioned reports, primary APS pa- 1. The previous pregnancy occurred successfully on conven-
tients with a well-defined antiphospholipid antibody profile tional therapy
(triple antiphospholipid antibody or LAC positivity) in asso- 2. The presence of a well-defined associated autoimmune
ciation with a history of thrombotic events and/or previous systemic disease
severe maternal–foetal complications develop more pregnan- 3. Age ≥ 45 years
cy failure/complications compared to pregnant APS women 4. Single- or multiple-organ failure
exclusively affected by previous miscarriages or foetal loss. 5. Severe pulmonary hypertension
Thus, treatment during pregnancy of this subset of APS pa- 6. Other known causes of pregnancy failure
tients should be differentiated because it should improve live
birth rates and reduce obstetric complications such as maternal All the women have to be treated with conventional plus
thrombosis, eclampsia, severe preeclampsia, HELLP syn- additional treatments within the 12th week of gestation.
drome, foetal growth restriction and prematurity, which However, additional treatments can start later due to
Clinic Rev Allerg Immunol

pregnancy complications occurring during conventional ther- IgM anti-β2GPI). In accordance with the Sydney update of the
apy alone in patients not included in the study from the begin- classification criteria [2], the cutoff values for medium/high
ning of pregnancy. Any additional treatments administered titres for both aCL and anti-β2GPI antibodies will be calculat-
alone or combined is considered in the study, including: low ed using the 99th percentile obtained by testing 120 age-
dose of steroids, equivalent to 10 mg/daily prednisolone; matched healthy women. Alternatively, centres could assess
hydroxychloroquine between 200 and 400 mg/daily accord- IgG and IgM aCL and IgG and IgM anti-β2GPI antibodies
ing to body weight; IVIG in a systematic way with a total using commercial kits following the manufacturers’ instruc-
amount of 2 g/kg per month, divided into two to five doses; tions. LAC is assessed by multiple coagulation tests using
apheresis procedures including plasma exchange or platelet-poor plasma samples following the revised interna-
immunoadsorption administered in a systematic way accord- tionally accepted guidelines [97].
ing to a well-defined timetable; other treatments in accordance Comparisons of the data will be performed using Pearson’s
with the choice of the physician. chi-square test, Fisher’s exact test and the Mann–Whitney test.
The study’s primary outcome is the live birth rate, i.e. the Logistic regression and backward conditional logistic regres-
number of live newborns surviving for the first 27 days after sion analyses will assess the independent effect of the treat-
birth (perinatal period), whilst maternal thrombosis, eclamp- ments and yield the odds ratio (OR) for treatments adjusted for
sia, severe preeclampsia, HELLP syndrome, intrauterine confounding variables. A p value <0.05 is considered signif-
growth restriction or prematurity are the secondary outcomes. icant. A power analysis will be performed to calculate the
The medical records of the pregnant APS patients are hypothetical number of pregnancies required in each sub-
reviewed after an informed consent was obtained from all group and in each treatment group in order to obtain reliable
women. results. The assumption will be made that women who re-
Baseline data collected for analysis include: age; disease ceived conventional treatments had a 60–70 % chance of
duration; history and type of pregnancy morbidity; achieving live births. The analysis will be performed to detect
antiphospholipid antibody profile; other autoantibodies (anti- an absolute increase of 20–30 percentage points in the live
nuclear, anti-double-strand DNA, anti-extractable nuclear an- birth rates and a decrease of 20–30 percentage points in the
tigens and anti-thyroid antibodies); presence of genetic risk pregnancy complications during conventional plus different
factors for thrombosis (factor V Leiden, prothrombin additional treatments. The power of the statistical test will be
G20210A mutation, decrease in C and S protein antigens 80 % and the significance level 5 %.
and activities, antithrombin III deficiency and increased acti- The results of the study will be reported and discussed in a
vated protein C resistance); acquired risk factors for thrombo- paper.
sis (blood pressure >140/90 mmHg or use of antihypertensive
drugs, total cholesterol ≥ 250 mg/dl, diabetes mellitus, body Compliance with Ethical Standards
mass index ≥30 kg m−2 and cigarette smoking); and low C3
Conflict of Interest The authors declare that they have no conflict of
and/or C4 levels (below 80 and 10 mg/dL, respectively). interest.
The collected pregnancy-related data include: the type and
dosage of therapy prescribed; when therapy was begun; low
C3 and/or C4 levels; maternal and foetal complications in-
cluding maternal thrombosis, eclampsia, preeclampsia,
HELLP syndrome, premature rupture of membranes, placenta References
abruptio, oligohydramnios, intrauterine growth restriction, ab-
normalities of Doppler waveform of the uterine arteries and/or 1. Hughes GR (1984) The Prosser–White oration 1983. Connective
umbilical artery; pregnancy outcome (live birth or foetal death tissue disease and the skin. Clin Exp Dermatol 9:535–544
2. Miyakis S, Lockshin MD, Atsumi T et al (2006) International con-
or early miscarriage); the week of gestation when delivery
sensus statement on an update of the classification criteria for def-
took place; newborn’s sex; birth weight in grams and in per- inite antiphospholipid syndrome (APS). J Thromb Haemost 4:295–
centiles; Apgar score at 5 min; and neonatal complications. 306
ACL and anti-β2GPI of IgG and IgM isotypes are deter- 3. Sebire NJ, Fox H, Backos M, Rai R, Paterson C, Regan L (2002)
mined using a homemade enzyme-linked immunosorbent as- Defective endovascular trophoblast invasion in primary
antiphospholipid antibody syndrome-associated early pregnancy
say following internationally accepted guidelines [96]. The failure. Hum Reprod 17:1067–1071
results of aCL testing were expressed as immunoglobulin G 4. Di Simone N, Marana R, Castellani R et al (2010) Decreased ex-
phospholipid or immunoglobulin M phospholipid units using pression of heparin-binding epidermal growth factor-like growth
international reference material. The results of anti-β2GPI as- factor as a newly identified pathogenic mechanism of
antiphospholipid-mediated defective placentation. Arthritis
says were calculated as arbitrary units using a standard curve Rheum 62:1504–1512
obtained from a pool of positive samples calibrated to Koike’s 5. Shamonki JM, Salmon JE, Hyjek E, Baergen RN (2007) Excessive
monoclonal antibodies (HCAL for the IgG and EY2C9 for the complement activation is associated with placental injury in
Clinic Rev Allerg Immunol

patients with antiphospholipid antibodies. Am J Obstet Gynecol 23. Somerset DA, Raine-Fenning N, Gordon C, Weaver JB, Kilby MD
196:161–165 (1998) Intravenous immunoglobulin therapy in compromised preg-
6. Tong M, Viall CA, Chamley LW (2015) Antiphospholipid antibod- nancies associated with antiphospholipid antibodies and systemic
ies and the placenta: a systematic review of their in vitro effects and lupus erythematosus. Eur J Obstet Gynecol Reprod Biol 79:227–
modulation by treatment. Hum Reprod Update 21:97–118 229
7. Bose P, Black S, Kadyrov M et al (2005) Heparin and aspirin 24. Branch DW, Peaceman AM, Druzin M et al (2000) A multicenter,
attenuate placental apoptosis in vitro: implications for early preg- placebo-controlled pilot study of intravenous immune globulin
nancy failure. Am J Obstet Gynecol 192:23–30 treatment of antiphospholipid syndrome during pregnancy. The
8. Peaceman AM, Rehnberg KA (1995) The effect of aspirin and Pregnancy Loss Study Group. Am J Obstet Gynecol 182:122–127
indomethacin on prostacyclin and thromboxane production by pla- 25. Stojanovich L, Mikovic Z, Mandic V, Popovich-Kuzmanovich D
cental tissue incubated with immunoglobulin G fractions from pa- (2007) Treatment of antiphospholipid syndrome in pregnancy with
tients with lupus anticoagulant. Am J Obstet Gynecol 173:1391– low doses of intravenous immunoglobulin. Isr Med Assoc J 9:555–
1396 556
9. Fishman P, Falach-Vaknin E, Sredni B et al (1996) Aspirin- 26. Mar N, Kosowicz R, Hook K (2014) Recurrent thrombosis preven-
interleukin-3 interrelationships in patients with anti-phospholipid tion with intravenous immunoglobulin and hydroxychloroquine
syndrome. Am J Reprod Immunol 35:80–84 during pregnancy in a patient with history of catastrophic
10. de Jesus GR, Agmon-Levin N, Andrade CA et al (2014) 14th antiphospholipid syndrome and pregnancy loss. J Thromb
International Congress on Antiphospholipid Antibodies Task Thrombolysis 38:196–200
Force report on obstetric antiphospholipid syndrome. Autoimmun 27. Watanabe N, Yamaguchi K, Motomura K, HisanoM SH,
Rev 13:795–813 Murashima A (2014) Combination therapy with anticoagulants,
11. Kutteh WH (1996) Antiphospholipid antibody-associated recurrent corticosteroids and intravenous immunoglobulin for women with
pregnancy loss: treatment with heparin and low-dose aspirin is su- severe obstetric antiphospholipid syndrome. Clin Exp Rheumatol
perior to low-dose aspirin alone. Am J Obstet Gynecol 174:1584– 32:299–300
1589 28. Rose HL, Ho WK (2014) Management of very high risk pregnancy
12. Rai R, Cohen H, Dave M et al (1997) Randomised controlled trial with secondary anti-phospholipid sindrome and triple positivity to
of aspirin and aspirin plus heparin in pregnant women with recur- the anti-phospholipid antibodies. J Throm Thrombolysis 38:453–
rent miscarriage associated with phospholipid antibodies (or 456
antiphospholipid antibodies). BMJ 314:253–257 29. Bramham K, Thomas M, Nelson-Piercy C et al (2011) First-
13. Franklin RD, Kutteh WH (2002) Antiphospholipid antibodies trimester low-dose prednisolone in refractory antiphospholipid
(APA) and recurrent pregnancy loss: treating a unique APA positive antibody-related pregnancy loss. Blood 117:6948–6951
population. Hum Reprod 17:2981–2985 30. Frampton G, Cameron JS, Thom M et al (1987) Successful removal
14. Empson M, Lassere M, Craig J, Scott J (2005) Prevention of recur- of anti-phospholipid antibody during pregnancy using plasma ex-
rent miscarriage for women with antiphospholipid antibody or lu- change and low-dose prednisolone. Lancet 2:1023–1024
pus anticoagulant. Cochrane Database Syst Rev 2:CD002859 31. Kobayashi S, Tamura N, Tsuda H et al (1992) Immunoadsorbent
15. Mak A, Cheung MW, Cheak AA, Ho RC (2010) Combination of plasmapheresis for a patient with antiphospholipid syndrome dur-
heparin and aspirin is superior to aspirin alone in enhancing live ing pregnancy. Ann Rheum Dis 51:399–401
births in patients with recurrent pregnancy loss and positive anti- 32. Nakamura Y, Yoshida K, Itoh S et al (1999) Immunoadsorption
phospholipid antibodies: a meta-analysis of randomized controlled plasmapheresis as a treatment for pregnancy complicated by sys-
trials and meta-regression. Rheumatology 9:281–288 temic lupus erythematosus with positive antiphospholipid antibod-
16. Farquharson RG, Quenby S, Greaves M (2002) Antiphospholipid ies. Am J Reprod Immunol 41:307–311
syndrome in pregnancy: a randomized, controlled trial of treatment. 33. El-Haieg DO, Zanati MF, El-Foual FM (2007) Plasmapheresis and
Obstet Gynecol 100:408–413 pregnancy outcome in patients with antiphospholipid syndrome. Int
17. Lassere M, Empson M (2004) Treatment of antiphospholipid syn- J Gynaecol Obstet 99:236–241
drome in pregnancy—a systematic review of randomized therapeu- 34. Ruffatti A, Marson P, Pengo V et al (2007) Plasma exchange in the
tic trials. Thromb Res 114:419–426 management of high risk pregnant patients with primary
18. Parke A, Maier D, Wilson D, Andreoli J, Ballow M (1989) antiphospholipid syndrome. A report of 9 cases and a review of
Intravenous gamma-globulin, antiphospholipid antibodies, and the literature. Autoimmun Rev 6:196–202
pregnancy. Ann Intern Med 110:495–496 35. Bortolati M, Marson P, Chiarelli S et al (2009) Case reports of the
19. Wapner RJ, Cowchock FS, Shapiro SS (1989) Successful treatment use of immunoadsorption or plasma exchange in high-risk pregnan-
in two women with antiphospholipid antibodies and refractory cies of women with antiphospholipid syndrome. Ther Apher Dial
pregnancy losses with intravenous immunoglobulin infusions. Am 13:157–160
J Obstet Gynecol 161:1271–1272 36. Mayer-Pickel K, Horn S, Lang U, Cervar-Zivkovic M (2015)
20. Ron-el R, Vinder A, Golan A et al (1993) The use of intravenous Response to plasmapheresis measured by angiogenic factors in a
gammaglobulin, heparin and aspirin in the maintenance of pregnan- woman with antiphospholipid syndrome in pregnancy. Case Rep
cy of freeze thawed embryo in a patient with lupus-type anticoag- Obstet Gynecol Article ID 123408:1–6
ulant. Eur J Obstet Gynecol Reprod Biol 52:131–133 37. Ruffatti A, Favaro M, Hoxha A et al (2016) Apheresis and intrave-
21. Kaaja R, Julkunen H, Ammälä P et al (1993) Intravenous immuno- nous immunoglobulins used in addition to conventional therapy to
globulin treatment of pregnant patients with recurrent pregnancy treat high-risk pregnant antiphospholipid antibody syndrome pa-
losses associated with antiphospholipid antibodies. Acta Obstet tients. A prospective study. J Reprod Immunol 115:14–19
Gynecol Scand 72:63–66 38. Lima F, Khamashta MA, Buchanan NM, Kerslake S, Hunt BJ,
22. Arnout J, Spitz B, Wittevrongel C, Vanrusselt M, Van Assche A, Hughes GR (1996) A study of sixty pregnancies in patients with
Vermylen J (1994) High-dose intravenous immunoglobulin treat- the antiphospholipid syndrome. Clin Exp Rheumatol 14:131–136
ment of a pregnant patient with an antiphospholipid syndrome: 39. Sailer T, Zoghlami C, Kurz C et al (2006) Anti-beta2-glycoprotein I
immunological changes associated with a successful outcome. antibodies are associated with pregnancy loss in women with the
Thromb Haemost 71:741–747 lupus anticoagulant. Thromb Haemost 95:796–801
Clinic Rev Allerg Immunol

40. Matsuki Y, Atsumi T, Yamaguchi K et al (2015) Clinical features 59. Sinha J, Chowdhry I, Sedan S, Barland P (2002) Bone marrow
and pregnancy outcome in antiphospholipid syndrome with history necrosis and refractory HELLP syndrome in a patient with cata-
of severe pregnancy complications. Mod Rheumatol 25:215–218 strophic antiphospholipid antibody syndrome. J Rheumatol 29:
41. Bramham K, Hunt BJ, Germain S et al (2010) Pregnancy outcome 195–197
in different clinical phenotypes of antiphospholipid syndrome. 60. Koenig M, Roy M, Baccot S, Cuilleron M, de Filippis JP, Cathédras
Lupus 19:58–64 P (2005) Trombotic microangiopathy with liver, gut and bone in-
42. Fischer-Betz R, Specker C, Brinks R et al (2012) Pregnancy out- farction (catastrophic antiphospholipid syndrome) associated with
come in patients with antiphospholipid syndrome after cerebral HELLP syndrome. Clin Rheumatol 2:166–168
ischaemic events: an observational study. Lupus 21:1183–1189 61. Asherson RA, Cervera R, Pierangeli SS (2007) Microangiopathic
43. Lockshin MD, Kim M, Laskin CA et al (2012) Prediction of ad- antiphospholipid syndromes (MAPS) revisited—new concepts re-
verse pregnancy outcome by the presence of lupus anticoagulant, lating to antiphospholipid antibodies and syndromes. J Rheumatol
but not anticardiolipin antibody, in patients with antiphospholipid 34:1793–1795
antibodies. Arthritis Rheum 64:2311–2318 62. Steegers EA, von Dadelszen P, Duvekot JJ, Pijnenborg R (2010)
44. Simchen MJ, Dulitzki M, Rofe G et al (2011) High positive anti- Preeclampsia. Lancet 376:631–644
body titers and adverse pregnancy outcome in women with 63. de Jesus GR, dos Santos FC, Oliveira CS et al (2012) Management
antiphospholipid syndrome. Acta Obstet Gynecol Scand 90:1428– of obstetric antiphospholipid syndrome. Curr Rheumatol Rep 14:
1433 79–86
45. Danowski A, de Azevedo MN, de Souza Papi JA et al (2009) 64. Spegiorin LC, Galão EA, De Godoy JM et al (2007)
Determinants of risk for venous and arterial thrombosis in primary Antiphospholipid antibodies and growth retardation in intrauterine
antiphospholipid syndrome and in antiphospholipid syndrome with development. Prague Med Rep 108:185–190
systemic lupus erythematosus. J Rheumatol 36:1195–1199 65. Dadhwal V, Sharma AK, Deka D et al (2011) The obstetric outcome
46. De Carolis S, Botta A, Santucci S et al (2010) Predictors of preg- following treatment in a cohort of patients with antiphospholipid
nancy outcome in antiphospholipid syndrome: a review. Clin Rev antibody syndrome in a tertiary care center. J Postgrad Med 57:16–
Allergy Immunol 38:116–124 19
47. Reggia R, Ziglioli T, Andreoli L et al (2012) Primary 66. Serrano F, Nogueira I, Borges A, Branco J (2009) Primary
antiphospholipid syndrome: any role for serum complement levels antiphospholipid syndrome: pregnancy outcome in a Portuguese
in predicting pregnancy complications? Rheumatology 51:2186– population. Acta Reumatol Port 34:492–497
2190
67. Cervera R, Piette JC, Font J et al (2002) Antiphospholipid syn-
48. Danza A, Rui z-Irastorza G, Khamashta MA (2012)
drome: clinical and immunologic manifestations and patterns of
Antiphospholipid sindrome in obstetrics. Best Pract Res Clin
disease expression in a cohort of 1,000 patients. Arthritis Rheum
Obstet Gynaecol 26:65–76
46:1019–1027
49. Cervera R, Khamashta MA, Shoenfeld Yet al (2009) Morbidity and
68. Andreoli L, Fredi M, Nalli C et al (2012) Pregnancy implications
mortality in the antiphospholipid syndrome during a 5-year period:
for systemic lupus erythematosus and the antiphospholipid syn-
a multicentre prospective study of 1000 patients. Ann Rheum Dis
drome. J Autoimmun 38:197–208
68:1428–1432
50. Gris JC, Bouvier S, Molinari N et al (2012) Comparative incidence 69. Ruffatti A, Tonello M, Del Ross T et al (2006) Antibody profile and
of a first thrombotic event in purely obstetric antiphospholipid syn- clinical course in primary antiphospholipid syndrome with preg-
drome with pregnancy loss: the NOH-APS observational study. nancy morbidity. Thromb Haemost 96:337–341
Blood 119:2624–2632 70. Ruffatti A, Tonello M, Cavazzana A et al (2009) Laboratory clas-
51. Gomez-Puerta JA, Sanin-Blair J, Galarza-Maldonado C (2009) sification categories and pregnancy outcome in patients with prima-
Pregnancy and catastrophic antiphospholipid syndrome. Clin Rev ry antiphospholipid syndrome prescribed antithrombotic therapy.
Allergy Immunol 36:85–90 Thromb Res 123:482–487
52. Costedoat-Chalumeau N, Guettrot-Imbert G, Leguern et al (2012) 71. Ruffatti A, Tonello M, Visentin MS et al (2011) Risk factors for
Pregnancy and antiphospholipid syndrome. Rev Med Interne 33: pregnancy failure in patients with anti-phospholipid syndrome
209–216 treated with conventional therapies: a multicentre, case-control
53. Nodler J, Moolamalla SR, Ledger EM, Nuwayhid BS, Mulla ZD study. Rheumatology 50:1684–1689
(2009) Elevated antiphospholipid antibody titers and adverse preg- 72. Wijetilleka S, Scoble T, Khamashta M (2012) Novel insights into
nancy outcomes: analysis of a population-based hospital dataset. pathogenesis, diagnosis and treatment of antiphospholipid syn-
BMC Pregnancy Childbirth 9:11 drome. Curr Opin Rheumatol 24:473–481
54. de Jesus GR, Rodrigues G, de Jesus NR, Levy RA (2014) 73. Alijotas-Reig J (2013) Treatment of refractory obstetric
Pregnancy morbidity in antiphospholipid syndrome: what is the antiphospholipid syndrome: the state of the art and new trends in
impact of treatment? Curr Rheumatol Rep 16:403 the therapeutic management. Lupus 22:6–17
55. Appenzeller S, Souza FH, Wagner Silva de Souza A, Shoenfeld Y, 74. Vaquero E, Lazzarin N, Valensise H et al (2001) Pregnancy out-
de Carvalho JF (2011) HELLP syndrome and its relationship with come in recurrent spontaneous abortion associated with
antiphospholipid syndrome and antiphospholipid antibodies. Semin antiphospholipid antibodies: a comparative study of intravenous
Arthritis Rheum 41:517–523 immunoglobulin versus prednisone plus low-dose aspirin. Am J
56. Pauzner R, Dulitzky M, Carp H et al (2003) Hepatic infarctions Reprod Immunol 45:174–179
during pregnancy are associated with the antiphospholipid syn- 75. Triolo G, Ferrante A, Ciccia F et al (2003) Randomized study of
drome and in addition with complete or incomplete HELLP syn- subcutaneous low molecular weight heparin plus aspirin versus
drome. J Thromb Haemost 1:1758–1763 intravenous immunoglobulin in the treatment of recurrent foetal
57. Le Thi TD, Tieulié N, Costedoat N et al (2005) The HELLP syn- loss associated with antiphospholipid antibodies. Arthritis Rheum
drome in the antiphospholipid syndrome: retrospective study of 16 48:728–731
cases in 15 women. Ann Rheum Dis 64:273–278 76. Dendrinos S, Sakkas E, Makrakis E (2009) Low-molecular-weight
58. Hochfeld M, Druzin ML, Maia D, Wright J, Lambert E, McGuire J heparin versus intravenous immunoglobulin for recurrent abortion
(1994) Pregnancy complicated by primary antiphospholipid anti- associated with antiphospholipid antibody syndrome. Int J
body syndrome. Obstet Gynecol 83:804–805 Gynaecol Obstet 104:223–225
Clinic Rev Allerg Immunol

77. Sherer Y, Levy Y, Shoenfeld Y (2000) Intravenous immunoglobu- in pregnant patients with antiphospholipid syndrome. J Clin Apher
lin therapy of anti-phospholipid syndrome. Rheumatology (Oxford) 27:200–204
39:421–426 89. Marson P, Bagatella P, Bortolati M et al (2008) Plasma exchange for
78. Carreras LD, Perez GN, Vega HR, Casavilla F (1988) Lupus anti- the management of the catastrophic antiphospholipid syndrome:
coagulant and recurrent fetal loss: successful treatment with importance of the type of fluid replacement. J Intern Med 264:
gammaglobulin. Lancet 2:393–394 201–203
79. Tenti S, Cheleschi S, Guidelli GM, Galeazzi M, Fioravanti A 90. Ruffatti A, Favaro M, Brucato A et al (2015) Apheresis in high risk
(2015) Intravenous immunoglobulins and antiphospholipid syn- antiphospholipid syndrome pregnancy and autoimmune congenital
drome: how, when and why? A review of the literature. heart block. Transfus Apher Sci 53:269–278
Autoimmun Rev 15:226–235 91. Mekinian A, Costedoat-Chalumeau N, Masseau A et al (2015)
80. Gelfand EW (2012) Intravenous immune globulin in autoimmune Obstetrical APS: is there a place for hydroxychloroquine to im-
and inflammatory diseases. N Engl J Med 367:2015–2025 prove the pregnancy outcome? Autoimmun Rev 14:23–29
81. Guidelli GM, Tenti S, Pascarelli NA, Galeazzi M, Fioravanti A 92. Marchetti T, Ruffatti TA, Wuillemin C, de Moerloose P, Cohen M
(2015) Granulomatosis with polyangiitis and intravenous immuno- (2014) Hydroxychloroquine restores trophoblast fusion affected by
globulins: a case series and review of the literature. Autoimmun antiphospholipid antibodies. J Thromb Haemost 12:910–920
Rev 14:659–664
93. Rand JH, Wu XX, Quinn AS, Taatjes DJ (2010) The annexin A5-
82. Caccavo D, Vaccaro F, Ferri GM, Amoroso A, Bonomo L (1994)
mediated pathogenic mechanism in the antiphospholipid syndrome:
Anti-idiotypes against antiphospholipid antibodies are present in
role in pregnancy losses and thrombosis. Lupus 19:460–469
normal polyspecific immunoglobulins for therapeutic use. J
Autoimmun 7:537–548 94. Mekinian A, Lazzaroni MG, Kuzenko A (2015) The efficacy of
83. Galli M, Cortelazzo S, Barbui T (1991) In vivo efficacy of intrave- hydroxychloroquine for obstetrical outcome in anti-phospholipid
nous gammaglobulins in patients with lupus anticoagulant is not syndrome: data from a European multicenter retrospective study.
mediated by an anti-idiotypic mechanism. Am J Hematol 38:184– Autoimmun Rev 14:498–502
188 95. Ruffatti A, Salvan E, Del Ross T et al (2014) Treatment strategies
84. Bakimer R, Guilburd B, Zurgil N, Shoenfeld Y (1993) The effect of and pregnancy outcomes in antiphospholipid syndrome patients
intravenous gammaglobulin on the induction of experimental with thrombosis and triple antiphospholipid positivity. A
antiphospholipid syndrome. Clin Immunol Immunopathol 69:97– European multicentre retrospective study. Thromb Haemost 112:
100 727–735
85. Cherin P, Marie I, Michallet M et al (2016) Management of adverse 96. Devreese KMJ, Pierangeli SS, De Laat B, Tripodi A, Atsumi T,
events in the treatment of patients with immunoglobulin therapy: a Ortel TL (2014) For the Subcommittee on Lupus Anticoagulant/
review of evidence. Autoimmun Rev 15:71–81 Phospholipid/Dependent Antibodies. Testing for antiphospholipid
86. Scoble T, Wijetilleka S, Khamashta MA (2011) Management of antibodies with solid phase assays: guidance from the SSC of the
refractory anti-phospholipid syndrome. Autoimmun Rev 10:669– ISTH. J Thromb Haemost 12:792–795
673 97. Pengo V, Tripodi A, Reber G et al (2009) Update of the guidelines
87. Hauser AC, Hauser L, Pabinger-Fasching I et al (2005) The course for lupus anticoagulant detection. Subcommittee on Lupus
of anticardiolipin antibody levels under immunoadsorption therapy. Anticoagulant/Antiphospholipid Antibody of the Scientific and
Am J Kidney Dis 46:446–454 Standardisation Committee of the International Society on
88. Bontadi A, Ruffatti A, Marson P et al (2012) Plasma exchange and Thrombosis and Haemostasis. J Thromb Haemost 7:1737–1740
immunoadsorption effectively remove antiphospholipid antibodies

S-ar putea să vă placă și