Вы находитесь на странице: 1из 11

CLINICAL OBSTETRICS AND GYNECOLOGY

Volume 59, Number 3, 487497


Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.

Hereditary
Thrombophilia and
Recurrent Pregnancy
Loss
ASHLEY M. PRITCHARD, MD,* PAUL W. HENDRIX, DO,*
and MICHAEL J. PAIDAS, MD*w
*Department of Obstetrics, Gynecology and Reproductive Sciences,
Division of Maternal Fetal Medicine, Yale School of Medicine; and
w Yale Women and Childrens Center for Blood Disorders and
Preeclampsia Advancement, New Haven, Connecticut

Abstract: The challenging nature of recurrent preg- and frustrating task for the obstetrician.
nancy loss (RPL) is multifactorial, but largely begins The challenging nature of this clinical
withdetermining whomeets diagnosticcriteria forRPL
as definitions vary and frequently change. Many problem is multifactorial, but largely be-
patients seek obstetrical intervention after losses, even gins with determining who meets diagnos-
if they do not meet the criteria for RPL, and even those tic criteria for RPL as definitions vary and
strictlymeeting criteriaoften presentaconundrumasto frequently change. Commonly used defi-
the etiology of their condition. The contribution of nitions include the following:
hereditary thrombophilia to RPL, the impact of each
disorder on the clotting cascade, available evidence  Two or more failed clinical pregnancies
regarding pregnancy outcomes, and current recom- as documented by ultrasonography or
mendations for evaluation and treatment is presented. histopathologic examination.1
Key words: recurrent pregnancy loss, hereditary throm-  Three consecutive pregnancy losses,
bophilia, clotting cascade which are not required to be
intrauterine.2
A retrospective cohort study of 587
women who had 3 or more consecutive
Introduction pregnancy losses before 12 weeks gesta-
Management of patients with recurrent tion demonstrated that biochemical preg-
pregnancy loss (RPL) can be an emotional nancies and pregnancies of unknown
location have a similarly negative impact
Correspondence: Ashley M. Pritchard, MD, Depart- on future live birth as intrauterine preg-
ment of Obstetrics, Gynecology and Reproductive Sci- nancy losses.3 Many patients seek obstet-
ences, Division of Maternal Fetal Medicine, Yale School
of Medicine, FMB 339B, New Haven, CT. E-mail: rical intervention after losses, even if they
ashley.pritchard@yale.edu do notmeet the above criteria for RPL, and
The authors declare that they have nothing to disclose. even those strictly meeting criteria often

CLINICAL OBSTETRICS AND GYNECOLOGY / VOLUME 59 / NUMBER 3 / SEPTEMBER 2016

www.clinicalobgyn.com | 487
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
488 Pritchard et al

present a conundrum as to the etiology of this enhanced hemostatic response.7 A


their condition. Approximately 15% of successful pregnancy is a complex balance
pregnantwomenwillexperienceasporadic of the risks of hypercoagulability and
loss of a clinically recognized pregnancy, hemorrhage from the moment of implan-
2% will have 2 consecutive pregnancy tation to the time of delivery. Hemorrhage
losses, and 1% or less will have 3 consec- must be avoided during implantation as
utive losses.4 Often, the cause of RPL cytotrophoblasts invade the maternal de-
cannot be determined; no discrete diag- cidual vessels to remodel the architecture
nosis can be made in upward of 50% of of the maternal spiral arteries. The uterine
patients.5 RPL is often multifactorial; an decidual layer plays a crucial role in the
extensive discussion of discrete causes can prevention of hemorrhage during embryo
be found elsewhere. In this chapter, we will implantation, placentation, and the third
focus on the contribution of hereditary stage of labor. As with the paradoxical
thrombophilia to RPL, including a review nature of hemostasis in pregnancy, decid-
of pertinent inherited thrombophilias, the ual tissue factor can also promote the
impact of each disorder on the clotting intense hypofibrinogenemia and dissemi-
cascade, available evidence regarding nated intravascular coagulation observed
pregnancy outcomes, and current recom- in placental abruption and the coagulop-
mendations for evaluation and treatment. athy seen in amniotic fluid embolism.8
Associated perils also include increased
risk for thrombotic complications, a risk
Pregnancy: A Prothrombotic further increased in women with thrombo-
State philias. Venous thromboembolism (VTE)
Pregnancy poses significant systemic he- and associated complications remain a
mostatic challenges, even in the healthiest leading cause of maternal morbidity and
of patients. The physiology of pregnancy is mortality accounting for an estimated
one primed for thrombosis. All of Virch- 9.3% of maternal deaths in the United
ows classic antecedents of thrombosis are States from 2006 to 2010.6 But with risks,
presentandevolveaspregnancycontinues. there are crucial benefits as this hyper-
The hormonal changes of pregnancy lead coagulable balance drives a rapid transi-
to pooling of venous return in the lower tion from 600 to 700 mL/minute of uterine
extremities, with venous stasis resulting blood flow at term to appropriate post-
from vasodilation. The gravid uterus fur- partum bleeding in a matter of minutes
ther promotes stasis within the lower during delivery and puerperium. Disrup-
extremities. Endothelial injury is a key tion in this tenuous balance can inhibit
component of implantation, and delivery implantation, initiate miscarriage, drive
represents profound endothelial damage. pathologic placentation, or result in
In combination with physiological risks, hemorrhage.
many of the procoagulant factors of the
clotting cascade progressively increase
during pregnancy including factor II, Inherited Thrombophilias in
VII, VIII, and X. Fibrinolysis is also sup- Pregnancy
pressed through a measurable decrease in Inherited dysfunction of the coagulation
protein S and resistance to activated pro- cascade results in a compounding effect of
tein C.6 thehypercoagulablestateofpregnancy.By
As a result, pregnancy can be referred affecting the quantity and/or function of
to as a prothrombotic state with the certain coagulation factors, inherited
high-volume, high-flow, low-resistance thrombophilias can disturb the coagula-
uteroplacental circulation necessitating tion system. There is little dispute

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
Hereditary Thrombophilia and Recurrent Pregnancy Loss 489

regarding the relationship between inher- resistant to downregulation by protein C,


ited thrombophilia and thrombotic events and unable to modulate the activity of
including deep vein thrombosis and pul- factor VIII (Fig. 1).
monary embolism. Controversy remains In regard to recurrent fetal loss, data
regarding the connection between inherited surrounding FVL is the most robust com-
thrombophilia and adverse pregnancy out- pared with other inherited thrombo-
comes, including spontaneous abortion, philias. In a meta-analysis that included
fetal loss, preeclampsia, fetal growth re- 31 case-control, cohort, and cross-section-
striction, and placental abruption.8 Given al studies, Rey and colleagues determined
the emotionally charged nature of these an association between FVL and RPL
complications, inherited thrombophilias before 13 weeks (OR = 2.01; 95% CI,
continue to spur curiosity, with meta-anal- 1.13-3.58). Investigators also found a sim-
yses consistently finding data to support ilar association when evaluating studies
varying degrees of association between covering all gestational ages in which other
thrombophilias and pregnancy loss. Below causes for fetal loss were excluded (OR =
we will review the most common inherited 3.04;95%CI,2.16-4.3).Themeta-analysis
thrombophilias and available data regard- only included 1 study limited to recurrent
ing their association with adverse preg- fetal losses at >22 weeks, which found an
nancy outcomes. OR = 7.83 (95% CI, 2.83-21.67).10 Inter-
estingly, a cohort study by Roque et al11
determined FVL to have protective bene-
Factor V Leiden (FVL) fits from recurrent fetal loss starting before
The FVL carrier prevalence is approxi- 10weeksofgestation(OR = 0.23;95%CI,
mately 5% to 9% among whites of Euro- 0.07-0.77) but not from weeks 10 to 14
pean decent, making it the most common (OR = 1.07; 95% CI, 0.46-2.50). Rodger
inherited thrombophilia.8,9 The mutation and colleaguesprovidethe largest prospec-
is virtually absent in African, Chinese, tive data set evaluating FVL and fetal loss
Japanese, and other Asian populations; in a 2010 meta-analysis of 10 prospective
however, it is present in about 3% of cohort studies. Although RPLs were not
African Americans who are not recent evaluated, they found a small but statisti-
immigrants. The rate of homozygosity is cally significant association between FVL
approximately 1% among those with the and pregnancy loss at all gestational ages
gene mutation, resulting in a more severe (OR = 1.52; 95% CI, 1.06-2.19).12 Over-
phenotype. The condition results from a all, FVL appears to pose a small increased
point mutation on chromosome 1q23, riskof spontaneousabortionandfetalloss.
driving the substitution of arginine by Limited data regarding RPL and FVL also
glutamine at position 506 on the subse- suggest a modest association.
quent protein.5 This single substitution
creates a distorted protein C cleavage site,
making factor Va resistant to cleavage and
deactivation. In its intended state, factor V Prothrombin Gene Mutation
is activated by thrombin to factor Va. (PGM)
Factor Va is then responsible for the The prothrombin G20210A mutation re-
conversion of prothrombin to thrombin. sults from a single guanine to adenine
In the reverse fibrinolytic cascade, factor nucleotide point mutation in the untrans-
Va is cleaved by activated protein C and lated 30 region of the gene coding for
provides negative feedback for factor VIII. prothrombin. This alteration results in
In those affected by FVL, the distorted increased translation and elevated circulat-
cleavage site results in a factor Va that is ing prothrombin levels.8 The prevalence of

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
490 Pritchard et al

Intrinsic Pathway

XI
Extrinsic (Tissue Factor)
Antithrombin Pathway
XII XIIa X

XIa TF
VI
IX IXa VIIIa VIIa
I
PLT APC
PS

Antithrombin
Fibrinogen
(I)
Xa

TF = Tissue Factor Prothrombin Thrombin


PLT = Platelet ( II ) (IIa)
APC = Activated Protein C
PS = Protein S Va
= Intrinsic Pathway
Fibrin
= Extrinsic Pathway
(Ia)
= Common Pathway APC
= Inhibit Coagulation PS

FIGURE 1. Important factors of the coagulation system.

the mutation is between 2% and 4% among factor for thrombosis in pregnancy, data
European whites; it is less common among connecting this thrombophilia and preg-
women of African and Asian descent.9,13 nancy loss, recurrent or not, are contra-
Althoughthe connection between PGM dictory and definitive conclusions cannot
and VTE in pregnancy is clear, data be made based on available information.
regarding the impact of the PGM on early
and RPL is uncertain. On the basis of Reys
meta-analysis of 31 case-control, cohort, Protein C Deficiency
and cross-sectional studies, as outlined Although FVL and the prothrombin gene
above, the mutation was linked to recur- mutation G20210A often result in similar
rent first trimester loss (OR = 2.32; 95% or predictable phenotypes, protein C defi-
CI, 1.12-4.79) and recurrent fetal loss ciency has been linked to >160 distinct
before 25 weeks (OR = 2.56, 95% CI, mutations resulting in variable pheno-
1.04-6.29). There also was a significant types. The prevalence of protein C muta-
association with nonrecurrent fetal loss tion is estimated at 0.2% to 0.3% when
when studies across all gestational were determined by a functional assay with
included (OR = 2.05; 95% CI, 1.18- cutoff between 50% and 60%.8,13 The
3.54).10 In contrast, a systemic review that mutation may be more common among
included 4 prospective studies addressing those of Asian or African descent.9 Protein
PGM and fetal loss at all gestational ages C deficiency is inherited in an autosomal
failed to find an association (OR = 1.13; dominant pattern, with deficiency de-
95% CI, 0.64-2.01).12 Reviews of these scribed by 2 distinct subtypes, Type I and
data highlight the limitations and inherent Type II.TypeI is morecommonandresults
bias of the studies included. Overall, in both a decreased amount and activity of
although PGM is a common heritable risk protein C with extensive heterogeneity in

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
Hereditary Thrombophilia and Recurrent Pregnancy Loss 491

phenotype. Type II results in a normal level protein S is caused by a silenced gene or a


of protein C, but with decreased functional mutationthatresultsinreducedfreeprotein
activity. Protein C is a vitamin K-depend- S antigen levels and activity. There are
ent protein produced by the liver. When over 130 known mutations with variable
functional, it circulates as a zymogen and expression.8 Protein S deficiency is less
participates in anticoagulation following common with an estimated prevalence of
activation to become a serine protease, 0.03% to 0.13% among whites. However,
termedactivatedproteinC.Thisactivation true prevalence estimations may be limited
can be mediated by thrombin, or most bysubstantialvariabilityindetectionassays
efficiently when thrombin is bound to during pregnancy. For more reliable eval-
endothelial thrombomodulin. When acti- uation, screening is recommended outside
vated, protein C inactivates factor Va and of pregnancy, >6 weeks postpartum and
VIIIa. Downstream, this effect prevents in the absence of hormonal contraception.
activation of factor X and thrombin gen- Free protein S is significantly decreased
eration. The effect of protein C is enhanced in pregnancy. Levels decrease between
by protein S, which will be discussed in the second and third trimester, from
greater detail below. 38.9% 10.3% to 31.2% 7.4%.15 When
Protein C deficiency has not been clearly functioning, protein S accelerates protein Cs
linked to fetal loss or to RPL. The Euro- effect on factor Va and VIIIa, with down-
pean Prospective Cohort on Thrombo- stream suppression of thrombin formation.
philia (EPCOT) study evaluated 843 Protein S deficiency is divided into 3 major
women with varied thrombophilias and phenotypes.TypeIhaslowlevelsofproteinS
determinedtheORforproteinCdeficiency antigen, free and total, and decreased func-
andfetallossatanygestationalagetobe1.4 tion. Type II has normal free and total
(95% CI, 0.9-2.2), and 2.3 (95% CI, 0.6- antigen, but with impaired function. Type
8.3) for stillbirth defined as fetal loss >28 III has normal total antigen values, but low
weeks gestation, both statistically non- free protein S and decreased activity level.
significant findings.14 Rey et al10 found the Although few data exist regarding pro-
risk of RPL across all gestational ages to be tein S deficiency and RPL, some data
nonsignificant (OR = 1.57; 95% CI, 0.23- regarding nonrecurrent fetal loss are avail-
10.54), though the meta-analysis only in- able. Preston et al14 failed to find an
cluded small retrospective case-control association between pregnancy loss and
studies. Along the spectrum of maternal- protein S (OR = 1.3; 95% CI, 0.8-2.1).
fetal well-being, a rare fetal complication Conversely, a systematic review indicated
may result from homozygous or com- an association with stillbirth (defined as
pound heterozygous protein C deficiency unexplained fetal loss over 20-wk gestation
termed neonatal purpura fulminans. The with no fetal abnormalities) with an OR =
condition is characterized by disseminated 16.2 (95% CI, 5.0-52.3), though it was
intravascular coagulation and hemorrha- limitedbysmallstudysizes.16 Othersstudies
gic skin necrosis, caused by extremely low havefoundsimilarfindings.10 Givenlimited
levels of protein C. Despite this rare con- data to date, definitive conclusions regard-
dition, there remains no clear association ing the affect of protein S on RPL cannot be
between protein C deficiency and RPL. drawn and investigation continues.

Protein S Deficiency
Protein S is a vitamin K-dependent protein Antithrombin Deficiency
intimately involved in the protein C anti- Antithrombin deficiency was the first he-
coagulation mechanism. Deficiency in reditary thrombophilia identified; it is also

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
492 Pritchard et al

the most thrombogenic. There are >250 definitive causative relationship is not yet
associated mutations serving to decrease established.
gene transcription, leading to decreased
antithrombin antigen levels and activity or
normal antigen levels but decreased activ-
ity.8 Inheritance patterns are autosomal Methylenetatrahydrofolate
dominant with variable penetrance. Prev- Reductase (MTHFR)
alence of the mutation is estimated at 1 per MTHFR is an enzyme involved in folate
2500 patients, or a 0.02% to 1.15% among metabolism. MTHFRs role is to reduce
European whites. The prevalence may be 5,10 methylenetatrahydrofolate to 5-
higher in Asians with estimated prevalence methyltetrahydrofolate. The resulting en-
of 2%to 5%.9 Antithrombin is synthesized zyme acts to reduce homocysteine to
intheliverandendothelialcells;itsprimary methionine. Homozygosity for a MTHFR
mechanism of action is through an inhib- mutation is the most common cause of
itory effect on thrombin, working to pre- hyperhomocysteinemia. Homozygosity of
vent activated thrombins conversion of the MTHFR C677T and A1298C poly-
fibrinogen to fibrin. Antithrombin also morphisms is present in 10% to 16% and
acts as an inhibitor of clotting factors IX, 4% to 6% of all Europeans, respectively.
X, XI, XII and tissue-factor bound VIIa.5 The C677T allele has a carrier frequency
Similar to the thrombophilias outlined from 7% in sub-Saharan Africans to 44%
above, antithrombin deficiency is defined in Italians.17 The activity of the resulting
by subgroups. Type I is a quantitative enzyme is thermolabile, and is therefore
dysfunction and Type II is a qualitative less active at temperatures >371C. Homo-
dysfunction. Type II is again further div- zygotes for the C677T mutation demon-
ided into subcategories; Type IIa is char- strate elevated plasma homocysteine when
acterized by a defect in the reactive site of folate deficient, but normal levels when
the protein, and the resulting phenotype is folate replete. The second mutation,
often more thrombogenic. Type IIb has a A1298C, results in a more modest decrease
defect in the heparin-binding site, but the in enzyme function. Despite the potential
resulting phenotype is less thrombogenic. for decreased folate in individuals homo-
Type IIc includes both of the aforemen- zygous for these mutations, hyperhomo-
tioned defects. cysteinemia is quite rare given common
Given the rarity of antithrombin defi- folate food fortification and diets rich in
ciency, it is challenging to draw conclu- folate.
sions as to the thrombophilias effect on When homocysteinemia is present, it
pregnancy loss and related sequelae. The acts as a procoagulant. However, elevated
EPCOTstudyfounda modest increaserisk homocysteine levels remain a weak risk
in early fetal loss, as defined by gestational factor for VTE.8 Similarly, there does not
age <28 weeks (OR = 1.7, 95% CI, 1.0- seem to be connection between MTHFR
2.8). They also found a significant associ- and homocysteinemia with adverse preg-
ation with nonrecurrent losses at all gesta- nancy outcomes. At best, available data
tional ages (OR = 2.1; 95% CI, 1.2-3.6) are inconsistent with 1 meta-analysis find-
andafter28weeks(OR = 5.2;95%CI,1.5- ing a small link to recurrent fetal losses
18.1).14 Meta-analyses found no associa- <16 weeks (OR = 2.7; 95% CI, 1.4-5.2).18
tion with fetal loss, recurrent or not, but A 2000 study evaluating the pregnancies of
studies were limited by small numbers and 5883womenand14,492pregnanciesfound
retrospective design.10 In summary, data some correlation between elevated homo-
suggest an association between antithrom- cysteine and stillbirth, but the correlation
bin deficiency and pregnancy loss, but a did not meet statistical significance.19

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
Hereditary Thrombophilia and Recurrent Pregnancy Loss 493

More recent work has not indicated a link demonstrated success in prolonging preg-
between MTHFR homozygosity or ele- nancies in early-onset severe preeclampsia
vated homocysteine with adverse preg- with antithrombin treatment. Currently,
nancy outcomes. there is a large multicenter double-blinded
randomized controlled trial underway
studying recombinant antithrombin ther-
apy for severe preeclampsia between 23 and
Other Adverse Pregnancy 30 weeks. Nonetheless, there has been no
Outcomes clear connection or causative relationship
Although the focus of this chapter remains demonstrated between any of the inherited
the impact of inherited thrombophilias on thrombophilias and preeclampsia.21
RPL, data suggest these conditions may
impact ongoing pregnancies, both in terms FETAL GROWTH RESTRICTION
of fetal and maternal health. The condi- Fetal growth restriction has links to nu-
tions outlined below are important causes merous maternal, fetal, and placental con-
of perinatal morbidity and they share ditions. Thus, a link between fetal growth
pathophysiology with some cases of preg- restriction and inherited thrombophilia
nancy loss. has been extensively studied. A meta-
analysis that evaluated the relationship
PREECLAMPSIA between fetal growth restriction, homozy-
Preeclampsia remains an increasingly com- gous and heterozygous FVL, homozygous
mon, but poorly understood disease of and heterozygous PGM, and MTHFR
pregnancy. Given the high degree of ma- homozygosity found no relationship or a
ternal morbidity and mortality, copious weak relationship between these entities
ongoing studies strive to understand risk and fetal growth restriction.22
factors for disease development as well as
treatment. Although case-control studies PLACENTAL ABRUPTION
suggest an association between FVL and A consistent association between any in-
preeclampsia, meta-analysis of prospective herited thrombophilia and placental
studiesfailtoshowasignificantrelationship abruption has not been demonstrated.
(OR = 1.23; 95% CI, 0.89-1.70).12 Similar This may be in part due to the relatively
studies have been conducted among those rare occurrence of placental abruption in
withthePGM.Anestedcase-controlcohort combination with the rarity of diagnosed
evaluation through the Danish National inherited thrombophilias, making it chal-
Birth Cohort found no association between lenging to conduct well-designed studies in
PGMandpreeclampsiawithseverefeatures this area.
(OR = 0.81; 95% CI, 0.29-2.30).20 One
meta-analyses suggest a connection be-
tween protein C and protein S deficiency Who Should be Tested and
andpreeclampsia.However,studiesremain
few and small in participant numbers.16 Should Treatment be
Although antithrombin deficiency has Considered?
not been found to be a risk factor for When a patient experiences RPL, there is
preeclampsia, antithrombin is currently temptation to consider broad diagnostic
being evaluated as a therapy for pre- testing in an effort to offer some explan-
eclampsia. Antithrombin has both antico- ation for this emotional outcome. How-
agulant and anti-inflammatory properties, ever, given the clinical evidence to
making it a plausible candidate for thera- date, routine inherited thrombophilia
peutic intervention. Several smaller studies screening is not recommended for those

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
494 Pritchard et al

TABLE 1. Association Between Hereditary Thrombophilias and Selected Pregnancy


Complications
Nonrecurrent Late Nonrecurrent Recurrent First Recurrent
Pregnancy Loss Pregnancy Loss* Trimester Lossw Pregnancy Lossw z
FVL 1.52 (1.06-2.19)12z 2.06 (1.1-3.86)23 1.91 (1.01-3.61)23 3.04 (2.16-4.3)10
PGM 1.13 (0.64-2.01)12z 2.66 (1.28-5.53)23 2.70 (1.37-5.34)23 2.05 (1.18-3.54)10
Protein C 1.4 (0.9-2.2)14 2.3 (0.6-8.3)14 N/A 1.57 (0.23-10.54)10
deficiency
Protein S 1.3 (0.8-2.1)14 7.39 (1.28-42.83)10 N/A 14.72(0.99-218.01)10
deficiency
Antithrombin 2.1 (1.2-3.6)14 5.2 (1.5-18.1)14 N/A N/A
deficiency

Data are odds ratio (95% confidence interval).


*Studies varied in definition of late from >20 to >28 weeks gestation.
w Meta-analysis data, studies varied in definition of recurrent to include Z2 or Z3 prior events.
z Meta-analysis data, gestational age range varied in individual studies, all gestational ages included.
FVL indicates factor V Leiden; PGM, prothrombin gene mutation.

experiencing RPL8 (Table 1). In addition, receiving pharmacologic intervention, 32


routine screening among women with (22%) experienced pregnancy loss as
adverse pregnancy outcomes including compared with 29 (20%) women rece-
preeclampsia, IUGR, and placental iving close pregnancy surveillance only,
abruption is not recommended. In cases resulting in an OR of 0.91 (95% CI, 0.52-
of stillbirth, evaluation for thrombophilia 1.59).24 Of note, this study evaluated
may be considered if the suspected etiology women with unexplained RPL, and not
of the stillbirth includes a possible throm- those with concurrent inherited thrombo-
botic event. Discussion of further evalua- philia. A 2014 systematic review of 9
tion of RPL, preeclampsia, IUGR, and clinical trials among women with RPL
placental abruption includes evaluation (defined as 2 or more consecutive miscar-
of acquired thrombophilia, which is dis- riages) with or without concurrent inher-
cussed elsewhere. ited thrombophilia also evaluated the role
There have been a number of small of anticoagulation. Included trials were
observational trials evaluating pharmaco- bothrandomizedandquasi-randomizedin
logic intervention among this population; nature, with interventions including un-
however, comparison of outcomes is lim- fractionated heparin, lowmolecular-
ited by study heterogeneity of inclusion weight heparin (LMWH), and aspirin.
and exclusion criteria, and treatment regi- Although the studies were heterogenous
men, in addition to small sample size and in terms of design, treatment regimen, and
resulting confounding error. The 2010 proclivity for bias, among the 1228 women
SPIN study assessed pregnancy outcomes studied, anticoagulation did not have a
among 294 women with at least 2 consec- beneficial effect on live birth, regardless of
utive pregnancy losses in the United King- which anticoagulant was evaluated;
dom and New Zealand. Women were among women who received aspirin com-
randomized to receive enoxaparin versus pared with placebo the RR for live birth
low-dose aspirin, in addition to close was 0.94 (95% CI, 0.80-1.11; n = 256), in
pregnancy surveillance, in parallel to a women who received LMWH compared
control group receiving close pregnancy with aspirin RR was 1.08 (95% CI, 0.93-
surveillance only in a multicenter random- 1.26; n = 239), and in women who received
ized controlled trial. Of the 147 women LMWH and aspirin compared with no

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
Hereditary Thrombophilia and Recurrent Pregnancy Loss 495

treatmentRRwas1.01(95%CI,0.87-1.16; RPL and known inherited thrombophilia,


n = 322).25 with varying outcome; overall there re-
Given the available data, treatment mains an inability to generalize results.
among those with RPL and inherited Giventhelackofconsistentevidenceatthis
thrombophilia is often considered on a time, anticoagulation among women with
case-by-case basis, extrapolating from the inherited thrombophilia in an effort to
treatment of those with APLS, in whom improve pregnancy outcome is not recom-
heparin or aspirin is frequently used.5 mended. There is significant data support-
Support for antithrombotic treatment in ing treatment for deep vein thrombosis
those with inherited thrombophilia and prevention among this same population,
RPL was first published by Brenner and which is discussed in detail elsewhere.
colleagues in a study of 61 pregnancies in 50
women. Enoxaparin was prescribed
throughout the pregnancy and 4 to 6 weeks
into the postpartum period. About 75% of
the pregnancies resulted in a live birth Areas of Ongoing Research
compared with a success rate of 20% in As our knowledge of thrombosis and
previous pregnancies without anticoagula- inflammation expands, it is apparent both
tion.Thesameinvestigatorslaterwentonto relatedentitiesareimportantforsuccessful
study the efficacy and role of varying doses embryonic implantation and develop-
of enoxaparin.26 The 2014 TIPPS trial ment. Preimplantation factor (PIF) is a
evaluated the use of antepartum dalteparin 15 amino-acid embryo-derived product
for the prevention of VTE, pregnancy loss, integral to maternal adaptation to preg-
and placental-mediated pregnancy compli- nancy and communication between the
cationamongwomenwithinheritedthrom- mother and embryo.29 It is secreted by
bophilia. Women were randomized to viable gestations detectable at the 2-cell
receive dalteparin prophylactic dosing or stage in human pregnancies. It not only has
no dalteparin from the day of random- utility in diagnostic applications, specifi-
ization (<20 wk gestation) to 37 weeks cally in predicting successful pregnancy in
gestation. Findings indicate that dalteparin the bovine model, but also it is a promising
did not reduce the occurrence of VTE, therapeutic immunomodulator.30,31 PIF
pregnancy loss, or placenta-mediated com- promotes endometrial receptivity by upre-
plications.27 Among this same population, gulating a2b-3 integrin expression and
the EAGeR trial investigated the associa- promotes embryo decidual adhesion.29,32
tion between daily aspirin therapy and very PIFhaspleiotropiceffectsbeyondimmune
early pregnancy losses or euploid losses modulation including modulation of lym-
among women with 1 to 2 prior losses. phocyte proliferation, inhibition of natu-
Women were randomized to daily aspirin ral killer cell activity, and promotion of
and folic acid versus folic acid only in this both neuroprotection and neuroregenera-
block-randomized, double-blind, placebo- tion.33 PIFs properties and safety profile
controlled trial. Medication was continued hasallowedittobefasttrackedbytheFDA
for r6 menstrual cycles, or throughout for clinical trial in its first human trial,
pregnancy if they conceived, through 36 treating autoimmune hepatitis (Clinical-
weeks. Results indicated no association Trials.gov Identifier: NCT02239562). The
between aspirin therapy and clinically rec- field continues to investigate potential
ognized pregnancy losses or implantation diagnostic andtreatmentmodalitiesaimed
failures.28 atfosteringimprovedpregnancyoutcomes
A number of subsequent small studies for common reproductive and obstetric
have been conducted among women with complications.34

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
496 Pritchard et al

References 16. AlfirevicZ,RobertsD,MartlewV.Howstrongisthe


1. Practice Committee of the American Society for association between maternal thrombophilia and
Reproductive Medicine. Definitions of infertility adverse pregnancy outcome?: a systematic review.
andrecurrentpregnancyloss:acommitteeopinion. Eur J Obstet Gynecol Reprod Biol. 2002;101:614.
Fertil Steril. 2013;99:63. 17. Botto LD, Yang Q. 5, 10-methylenetetrahydrofo-
2. Jauniaux E, Farquharson RG, Christiansen OB, late reductase gene variants and congenital anoma-
et al. Evidence-based guidelines for the investiga- lies: a HuGE review. Am J Epidemiol. 2000;151:
tion and medical treatment of recurrent miscar- 862877.
riage. Hum Reprod. 2006;21:22162222. 18. Nelen WL, Blom HJ, Steegers EA, et al. Hyper-
3. Kolte A, van Oppenraaij R, Quenby S, et al. Non- homocysteinemia and recurrent early pregnancy
visualized pregnancy losses are prognostically loss: a meta-analysis. Fertil Steril. 2000;74:
important for unexplained recurrent miscarriage. 11961199.
Hum Reprod. 2014;29:931937. 19. Vollset SE, Refsum H,Irgens LM, et al. Plasmatotal
4. Salat-Baroux J. Recurrent spontaneous abortions. homocysteine, pregnancy complications, and ad-
Reprod Nutr Dev. 1988;28:15551568. verse pregnancy outcomes: the Hordaland Homo-
5. McNamee K, Dawood F, Farquharson RG. cysteine Study. Am J Clin Nutr. 2000;71:962968.
Thrombophilia and early pregnancy loss. Best 20. Lykke J, Bare L, Olsen J, et al. Thrombophilias and
Pract Res Clin Obstet Gynaecol. 2012;26:91102. adverse pregnancy outcomes: results from the
6. Hendrix PW, Paidas MJ. Thrombophila and Danish National Birth Cohort. Thromb Haemost.
pregnancy. In: Malvasi A, Tinelli A, Di Renzo 2012;10:13201325.
GC, eds. Management and Therapy of Early 21. Paidas MJ, Sibai BM, Triche EW, et al. Exploring
Pregnancy Complications. Cham, Switzerland: the role of antithrombin replacement for the
Springer International; 2016. treatment of preeclampsia: a prospective random-
7. Pettker CM, Lockwood CJ. Thromboembolic ized evaluation of the safety and efficacy of
disorders. In: Gabbe SG, Niebyl JR, Galan recombinant antithrombin in very preterm pree-
HL, et al, ?eds. Obstetrics: Normal and Problem clampsia (PRESERVE-1). Am J Reprod Immunol.
Pregnancies. Atlanta, GA: Elsevier Health Scien- 2013;69:539544.
ces; 2012. 22. Facco F, You W, Grobman W. Genetic thrombo-
8. American Congress of Obstetrics & Gynecology. philias and intrauterine growth restriction: a meta-
ACOG practice bulletin no. 138. Practice bulletin analysis. Obstet Gynecol. 2009;113:12061216.
no. 138: inherited thrombophilias in pregnancy. 23. Robertson L, Wu O, Langhorne P, et al. Throm-
Obstet Gynecol. 2013;122:706716. bophilia in pregnancy: a systematic review. Br J
9. Margaglione M, Grandone E. Population genetics Haematol. 2006;132:171196.
of venous thromboembolism. Thromb Haemost. 24. Clark P, Walker ID, Langhorne P, et al. SPIN
2011;105:221231. (Scottish Pregnancy Intervention) study: a multi-
10. Rey E, Kahn SR, David M, et al. Thrombophilic center, randomized controlled trial of low-molec-
disorders and fetal loss: a meta-analysis. Lancet. ular-weightheparinandlow-doseaspirininwomen
2003;361:901908. with recurrent miscarriage. Blood. 2010;115:
11. Roque H, Paidas MJ, Funai EF, et al. Maternal 41624167.
thrombophilias are not associated with early 25. de Jong PG, Kaandorp S, Di Nisio M, et al. Aspirin
pregnancy loss. Thromb Haemost. 2004;91: and/or heparin for women with unexplained re-
290295. current miscarriage with or without inherited
12. Rodger MA, Betancourt MT, Clark P, et al. The thrombophilia. Cochrane Database Syst Rev.
association of factor V Leiden and prothrombin 2014;7:CD004734.
gene mutation and placenta-mediated pregnancy 26. Brenner B. Antithrombotic prophylaxis for wom-
complications: a systematic review and meta- en with thrombophilia and pregnancy
analysis of prospective cohort studies. PLoS Med. complicationsyes. Thromb Haemost. 2003;1:
2010;7:728. 20702072.
13. Franco RF, Reitsma PH. Genetic risk factors of 27. Rodger MA, Hague WM, Kingdom J, et al.
venous thrombosis. Hum Genet. 2001;109: Antepartum dalteparin versus no antepartum
369384. dalteparin for the prevention of pregnancy com-
14. Preston FE, Rosendaal FR, Walker ID, et al. plications in pregnant women with thrombophilia
Increased fetal loss in women with heritable (TIPPS): a multinational open-label randomised
thrombophilia. Lancet. 1996;348:913916. trial. Lancet. 2014;384:16731683.
15. PaidasMJ,KuDH,LeeMJ,etal.ProteinZ,protein 28. Schisterman EF, Silver RM, Lesher LL, et al.
S levels are lower in patients with thrombophilia Preconception low-dose aspirin and pregnancy
and subsequent pregnancy complications. Thromb outcomes: results from the EAGeR randomised
Haemost. 2005;3:497501. trial. Lancet. 2014;384:2936.

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.
Hereditary Thrombophilia and Recurrent Pregnancy Loss 497

29. Paidas MJ, Krikun G, Huang SJ, et al. A genomic implantation: increases endometrial integrin-
and proteomic investigation of the impact of a2b3, amphiregulin and epiregulin while reducing
preimplantation factor on human decidual cells. betacellulin expression via MAPK in decidua.
Am J Obstet Gynecol. 2010;202:459.e1459.e8. Reprod Biol Endocrinol. 2012;10:50.
30. Ramu S, Stamatkin C, Timms L, et al. PreImplan- 33. Barnea E, Almogi-Hazan O, Or R, et al. Immune
tation factor (PIF) detection in maternal circulation regulatory and neuroprotective properties of pre-
in early pregnancy correlates with live birth (bovine implantation factor: From newborn to adult.
model). Reprod Biol Endocrinol. 2013;11:105114. Pharmacol Ther. 2015;156:1025.
31. Barnea ER, Kirk D, Ramu S, et al. PreImplanta- 34. Ornaghi S, Mueller M, Barnea ER, et al. Throm-
tion Factor (PIF) orchestrates systemic antiin- bosis during pregnancy: Risks, prevention, and
flammatory response by immune cells: effect on treatment for mother and fetusharvesting the
peripheral blood mononuclear cells. Am J Obstet power of omic technology, biomarkers and in vitro
Gynecol. 2012;207:e1e11. or in vivo models to facilitate the treatment of
32. Barnea ER, Kirk D, Paidas MJ. Preimplantation thrombosis. Birth Defects Res C Embryo Today.
factor (PIF) promoting role in embryo 2015;105:209225.

www.clinicalobgyn.com
Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.

Вам также может понравиться