Você está na página 1de 13

J Neurol (2013) 260:12021214

DOI 10.1007/s00415-012-6653-9

REVIEW

Multiple sclerosis and pregnancy: therapeutic considerations


Maria K. Houtchens Channa M. Kolb

Received: 22 May 2012 / Revised: 7 August 2012 / Accepted: 8 August 2012 / Published online: 25 August 2012
Springer-Verlag 2012

Abstract For women with multiple sclerosis (MS) who Keywords Multiple sclerosis  Pregnancy 
become pregnant, the risks and benefits of ongoing therapy Disease-modifying therapies  Management 
for the health of both the mother and the fetus must be Breastfeeding  Review
carefully considered. Based on a literature review and our
MS centers standard practices, we provide guidance to aid
clinical decision making in the absence of clear evidence- Introduction
based clinical practice guidelines. Women seeking to
achieve pregnancy should generally discontinue disease- Multiple sclerosis (MS) is an immune-mediated chronic
modifying therapy use prior to attempting conception. For neurodegenerative disease that may result in sustained
example, the immunosuppressant mitoxantrone is terato- physical and cognitive disability, reduction in quality of
genic and should be prescribed only with the assurance of life, and significant costs to families and society [14]. MS
effective contraception. Conception should be discouraged is the most common neurologic disease among young
for patients on fingolimod, because of the limited infor- adults, typically starting in the third and fourth decades of
mation available on human pregnancy outcomes. Current life. Women are more frequently affected than men, and
evidence, including data from pregnancy registries for MS can impact a womans reproductive years [5, 6]. Dis-
glatiramer acetate (GA), interferon beta-1a (IFNb-1a), and ease-modifying therapies (DMTs) and other medications
natalizumab, has not shown specific patterns of malfor- are widely used by women of childbearing potential. Thus,
mations suggesting teratogenicity. Pregnancy registry data women with MS who choose to become pregnant, experi-
have not been published for IFNb-1b. During breastfeed- ence an unplanned pregnancy or elect to breastfeed their
ing, intravenous immunoglobulin and corticosteroids are infants must consider the benefits and risks of MS therapy.
generally safe and may be associated with a reduction in The impact of pregnancy on MS has been reported in
postpartum relapses; however, a washout period is rec- various studies including the large, prospective Pregnancy in
ommended between corticosteroid administration and the Multiple Sclerosis (PRIMS) study. Pregnant women with
resumption of breastfeeding. Clinical data on the use of MS typically experience a gradual reduction in relapse rate
IFNb, GA, and natalizumab during lactation are limited. during the antenatal period, with an approximately 70 %
Mitoxantrone is contraindicated during breastfeeding, and reduction in relapse rate during the third trimester relative to
fingolimod should be avoided in nursing mothers, because the year before pregnancy [7]. By contrast, the relapse rate
of a lack of data. often increases to approximately 70 % above the prepre-
gnancy rate during the first postpartum trimester and 30 %
above the prepregnancy rate during postpartum trimesters
two and three before returning to prepregnancy levels [8, 9].
M. K. Houtchens (&)  C. M. Kolb While the complex mechanisms through which pregnancy
Department of Neurology, Partners Multiple Sclerosis Center,
may have positive effects on MS relapses are beyond the
Brigham and Womens Hospital, Harvard Medical School,
1 Brookline Place #225, Brookline, MA 02445, USA scope of the current review, high levels of estrogens have
e-mail: mhoutchens@partners.org been implicated in these therapeutic outcomes [10].

123
J Neurol (2013) 260:12021214 1203

There is currently no compelling evidence that preg- DMTs, and none of these therapies have been placed in the
nancy has any adverse effects on MS progression, even in safest pregnancy risk category (Table 3) [16]. The avail-
patients who experience postpartum exacerbations [1113]. able evidence for use of each therapy during pregnancy is
Although the impact of pregnancy on MS is well-estab- summarized below.
lished, there is little published information available on the
effects of MS treatments on the mother and the fetus/ GA
newborn. In addition, there are currently no evidence-based
clinical practice guidelines on clinical decision making and GA is the only DMT with an FDA pregnancy category B
therapeutic choices for women with MS who become rating [17]. This rating is based on the results of studies in
pregnant. rats and rabbits that show no adverse effects on offspring
In the US, current National MS Society recommenda- development at up to 36 times the therapeutic human dose.
tions [14] suggest that women stop their medication one However, since well-controlled studies of human preg-
full menstrual cycle prior to attempting conception. These nancy are lacking [17] and animal reproduction studies do
recommendations also allow resumption of therapy not always predict human response, the US prescribing
immediately after delivery, if breastfeeding is not planned. information for GA states that it should only be used during
Women on therapy for MS who unexpectedly become pregnancy, if clearly needed [17]. The EMA does not
pregnant are often advised to discontinue therapy as soon provide guidance on the use of GA during pregnancy.
as the pregnancy is recognized [12, 14]. The European However, the Association of British Neurologists guide-
Medicines Agency (EMA) has not issued general guide- lines for prescribing in MS state that GA is not licensed for
lines for MS care during pregnancy, but individual coun- use during pregnancy.
tries may provide guidance regarding the use of specific The GA manufacturers postmarketing surveillance
medications during pregnancy. Little guidance is available study, which was presented in 2003 [18] is the largest
for women who wish to continue their medication during source of data on GA safety during pregnancy. The overall
either pregnancy or breastfeeding or who want to better miscarriage rate (17 %) was close to the expected range in
understand the possible effects of prior therapy on their the US population (1015 %) [19], and there was no
pregnancy/newborn. apparent increased risk of adverse fetal or pregnancy out-
The purpose of this review is first to provide an comes. However, data on GA exposure throughout preg-
up-to-date summary of the available data on the utilization nancy are lacking, since most women discontinued
of DMTs and other medications for acute MS relapses treatment during the first trimester. Another study [20]
during pregnancy and breastfeeding. A second objective is reported a similar duration of GA exposure (median
to provide data- and experience-based practical guidance 6.9 weeks) based on registry data, with only 7 of the 31
on MS management in women on DMTs who plan to women in the analysis (23 %) receiving GA beyond week
become pregnant, in women who unexpectedly become 7. Again, GA exposure did not increase the risk for fetal
pregnant while receiving MS treatment, and in those who adverse effects, and birth weight was not significantly
elect to breastfeed. affected. There were also no observed complications in a
Brazilian study that included 20 patients on GA during
pregnancy [21]. However, the duration of GA usage during
MS therapies and pregnancy pregnancy and specific trimesters of exposure were not
reported. In addition, none of these studies reported patient
Therapies approved for use in MS patients relapse rates during or after pregnancy.
Three longer duration observational studies followed
US Food and Drug Administration (FDA) and EMA clas- women on GA during pregnancy and reported MS out-
sifications of pregnancy risk for medications are presented comes and relapse rates. In a retrospective assessment of 11
in Table 1. The FDA has created pregnancy risk categories women with MS on GA continuously for 7 months or more
for inclusion in consumer product information, while the during pregnancy [22], their offspring were fully evaluated
EMA has developed specific statements that are acceptable for complications after at least 1 year of age. There were
for use in the pregnancy section of a product monograph no observed drug-related malformations, neonatal compli-
[15]. There are currently seven approved MS therapies: cations or developmental abnormalities. The mean patient
glatiramer acetate (GA), subcutaneous (SC) interferon beta MS relapse rate of 1.6 in the 10 months prior to pregnancy
(IFNb)-1a, intramuscular (IM) IFNb-1a, SC IFNb-1b, fin- decreased to 0.4 (p = 0.005) during pregnancy and
golimod, natalizumab, and mitoxantrone (Table 2). To date remained low (0.6, p = 0.005 vs the prepregnancy rate) in
there are no reports of completed, adequate, well-con- the first 6 months post partum. Salminen et al. [23]
trolled prospective human pregnancy studies for these reported analogous findings in a prospective observational

123
1204 J Neurol (2013) 260:12021214

Table 1 US and EU pregnancy risk classifications for medications


FDA pregnancy risk categories
Category Description

A Adequate studies in pregnant women have failed to show a risk to the fetus in the first trimester, and there is no evidence of risk in
later trimesters
B Animal studies have failed to show a risk to the fetus, but there are no adequate studies in pregnant women or animal studies have
shown an adverse effect, but human studies have not shown a risk to the fetus in the first trimester, and there is no evidence of risk
in later trimesters
C Animal studies have shown an adverse effect on the fetus and there are no adequate studies in humans, but the benefits may outweigh
the risks or there are no adequate human studies
D There is positive evidence of human fetal risk, but the benefits may outweigh the risks
X Animal or human studies have shown fetal abnormalities or toxicity, and the risk outweighs the benefits
EMA pregnancy risk statements [15]
1. Based on human experience (specified), Drug X is suspected to cause congenital malformation (specified), when administered during
pregnancy
2. Drug X should not be used during pregnancy (trimester specified), unless the clinical condition of the woman requires treatment with Drug X
3. A moderate amount of data on pregnant women (between 300 and 1,000 pregnancy outcomes) indicates no malformative or feto/neonatal
toxicity for Drug X
4. No effects during pregnancy are anticipated, since systemic exposure to Drug X is negligible
FDA US Food and Drug Administration, EMA European Medicines Agency

case series of 13 women with highly active relapsing- states that initiation of IFNb treatment is contraindicated
remitting MS who were taking GA during pregnancy. The during pregnancy and that discontinuation of therapy
9 women who received GA throughout the antenatal period should be considered for women who become pregnant
all had live births with no reported birth defects. Finally, while taking IFNb.
Hellwig and Gold [24] followed 3 women taking GA In contrast, data from several large registry studies
throughout pregnancy and post partum. One case of penile suggest that IFNb-1a does not increase the human spon-
hypospadias was reported. With respect to disease activity, taneous abortion rate. Preliminary results from a pregnancy
no subjects had a relapse in the year before pregnancy, registry study of IM IFNb-1a treatment (the Avonex
during pregnancy or in the first 6 months post partum. pregnancy exposure registry [28]) that enrolled a total of
In summary, published open-label observations appear 329 patients showed that the spontaneous abortion rate
to confirm preclinical studies and suggest that GA is not (10.5 %) was in line with reported rates for the US general
associated with teratogenicity or a higher rate of miscar- population (1015 %) [19], with no increase in the pro-
riage [11, 18, 2024]. Although the potential relapse portion of births with major/serious malformations or
reduction with GA during pregnancy is interesting, there overrepresentation of specific malformations. Consistent
are only a small number of reported cases, and pregnancy findings were reported for an SC IFNb-1a pregnancy reg-
is normally associated with fewer MS relapses, particularly istry that included 1,022 cases of exposure to SC IFNb-1a
during the third trimester. These observations should and 679 documented outcomes [29]. In cases for which
therefore be interpreted with caution; current evidence information was available, the mean duration of fetal
supports no definitive conclusions about the therapeutic exposure before treatment discontinuation was 28 days.
efficacy of GA in pregnant women. Among prospectively studied pregnancies, 11.5 % resulted
in spontaneous abortions which is similar to the US rate
IFNb [19] and live birth major congenital anomalies were also in
line with the general population. Relapse rates were not
In primates, administration of up to 100 times the recom- reported in these registry studies. It should be noted that
mended weekly human dose of IFNb (based upon a body registries to date have only been published for IM and SC
surface area comparison) was not associated with terato- IFNb-1a, similar large-cohort data are not currently avail-
genicity or adverse effects on fetal development [2527]. able for IFNb-1b.
In spite of these data, the FDA has given IFNb drugs a Other reports also suggest that IFNb has no effect on
pregnancy category C rating based on experimental pri- fetal abnormalities and spontaneous abortions [21, 24, 30,
mate studies showing abortifacient activity at 2100 times 31]. One study [30] evaluating data on 396 pregnancies in
the corresponding human dose. The EMA guidance also 388 women (mean exposure 4.6 weeks) reported an 8 %

123
Table 2 DMTs currently approved for use in MS
Generic (brand) name Indication(s)a

GA (Copaxone, Teva Pharmaceutical Industries, FDA: To reduce relapse frequency in patients with relapsing-remitting MS and patients who have experienced a first clinical episode and
Petach Tikva, Israel) have MRI features consistent with MS
ABN guidelines: For the treatment of patients with a diagnosis of active MS with relapsing onset (relapsing-remitting MS) to reduce
relapses, active disease is defined by two clinically significant relapses in the previous 2 years, other patient categories can be
considered
IM IFNb-1a (Avonex, Biogen Idec, Weston, MA, FDA: To slow accumulation of physical disability and decrease frequency of clinical exacerbations in patients with relapsing forms of MS
USA) and patients who have experienced a first clinical episode and have MRI features consistent with MS
J Neurol (2013) 260:12021214

EMA: For the treatment of (1) patients diagnosed with relapsing MS, to slow the progression of disability and decrease the frequency of
relapses, and (2) patients with a single demyelinating event with an active inflammatory process, if it is severe enough to warrant
treatment with intravenous corticosteroids, if alternative diagnoses have been excluded, and if the patients are determined to be at high
risk of developing clinically definite MS
SC IFNb-1a (Rebif, Merck Serono, Geneva, FDA: To slow the accumulation of physical disability and decrease the frequency of clinical exacerbations in relapsing forms of MS
Switzerland) EMA: For the treatment of (1) patients with a single demyelinating event with an active inflammatory process, if alternative diagnoses
have been excluded and if the patients are determined to be at high risk of developing clinically definite MS, and (2) patients with
relapsing MS
IFNb-1b (Betaseron/Betaferon, Bayer FDA: To reduce the frequency of clinical exacerbations in patients with relapsing forms of MS and patients who have experienced a first
HealthCare Pharmaceuticals, clinical episode and have MRI features consistent with MS
Pine Brook, NJ, USA; Extavia, Novartis, Basel, EMA: For the treatment of (1) patients with a single demyelinating event with an active inflammatory process, if it is severe enough to
Switzerland) warrant treatment with intravenous corticosteroids, if alternative diagnoses have been excluded, and if the patients are determined to be
at high risk of developing clinically definite MS, (2) patients with relapsing-remitting MS and two or more relapses within the last
2 years and (3) patients with secondary progressive MS with active disease, as evidenced by relapses
TM
Fingolimod (Gilenya , Novartis) FDA: To reduce the frequency of clinical exacerbations and delay accumulation of physical disability in patients with relapsing forms of
MS
EMA: As monotherapy for adult patients with highly active relapsing-remitting MS despite treatment with a beta-interferon or with
rapidly evolving, severe relapsing-remitting MS, as defined by two or more disabling relapses in 1 year and one or more gadolinium-
enhancing lesions on brain MRI or a significant increase in T2 lesion load as compared to a previous recent MRI
Natalizumab (Tysabri, Biogen Idec) FDA: As monotherapy for relapsing forms of MS, to delay accumulation of physical disability and reduce frequency of clinical
exacerbations
EMA: As monotherapy for adult patients with highly active relapsing-remitting MS despite treatment with a beta-interferon or with
rapidly evolving, severe relapsing-remitting MS, as defined by two or more disabling relapses in 1 year and one or more gadolinium-
enhancing lesions on brain MRI or a significant increase in T2 lesion load as compared to a previous recent MRI
Mitoxantrone (Novantrone, EMD Serono, FDA: To reduce neurologic disability and/or the frequency of clinical relapses in secondary (chronic) progressive, progressive relapsing,
Darmstadt, Germany) or worsening relapsing-remitting MS
EMA: Not approved for MSa
DMT disease-modifying therapy, MS multiple sclerosis, GA glatiramer acetate, FDA US Food and Drug Administration, MRI magnetic resonance imaging, ABN association of british
neurologists, IM intramuscular, IFNb interferon beta, EMA European Medicines Agency, SC subcutaneous
a
Some indications may be country-specific. Please refer to country-specific guidelines for individual medications
1205

123
1206 J Neurol (2013) 260:12021214

spontaneous abortion rate with IFNb-1a. An observational somewhat conflicting findings [20, 32, 33]. There is
study of 7 patients on IFNb throughout pregnancy and in inconsistent information as to whether IFNb leads to a
the postpartum period [24] also reported no increase in reduction in birth weight some studies found a reduction,
spontaneous abortions or malformations. Unexpectedly, while others reported no such effect. Women who become
disease activity increased, although these data should be pregnant or plan to become pregnant, while taking IFNb
interpreted cautiously given the small number of patients. should be informed of these findings and the risks and
No other studies to date have directly examined the impact benefits of treatment should be discussed.
of IFNb on relapse rate during pregnancy.
In contrast to large registry and observational studies, Fingolimod
some smaller studies have found higher rates of sponta-
neous abortion in patients treated with IFNb preparations The FDA has given fingolimod a pregnancy category C
[20, 32, 33]. Sandberg-Wollheim et al. [32] examined rating based on experimental rat and rabbit studies showing
individual patient data from eight clinical trials with SC developmental toxicity including teratogenicity (in rats),
IFNb-1a. Of the 41 pregnancies with in utero IFNb expo- and embryolethality following fingolimod administration to
sure (B16 weeks, with most women discontinuing at pregnant animals [35]. EMA product information recom-
B4 weeks), 19.5 % (8 of 41) spontaneously aborted. mends that women of childbearing potential be counseled
Boskovic et al. [33] followed 16 women (23 pregnancies) regarding the potential for serious risk to the fetus and the
and reported a spontaneous abortion rate of 39 % with need for effective contraception during treatment with
IFNb exposure (any formulation) versus 5 % in healthy fingolimod [36]. The sphingosine-1-phosphate receptors
non-MS controls; however, the duration of IFNb exposure affected by fingolimod have established functions in vas-
during pregnancy was not reported. Two major malfor- cular formation during embryogenesis. The most common
mations (Down syndrome and an abnormal X chromo- fetal visceral malformations in rats included ventricular
some; 8.7 %) were reported in the IFNb group and one septal defect and persistent truncus arteriosus, in which a
malformation (5.0 %) among healthy controls. By contrast, single arterial trunk arises from the two cardiac ventricles
Weber-Schoendorfer and Schaefer [20] found that IFNb-1a by means of a single semilunar valve. The highest no-effect
(SC or IM) was not associated with an increase in spon- dose in rat studies (0.03 mg/kg/day) was less than the
taneous losses. However, a higher miscarriage rate was recommended human dose of 0.5 mg/day on a body sur-
reported with IFNb-1b [28 % (5 of 18)] than with IFNb-1a face area (mg/m2) basis, while in rabbit studies, the no-
[5 % (2 of 42); p = 0.02] or GA [4 % (1 of 26); p = 0.02] effect dose (0.5 mg/kg/day) was approximately 20 times
or in healthy non-MS controls [9 % (138 of 1,515); the recommended human dose.
p = 0.03]. No malformations were reported for patients A pregnancy registry has been established to collect
taking IFNb-1a. Although important, these results should information on fingolimod use during pregnancy. As of
be considered together with the results from the larger trials January 2011, 47 pregnancies were reported in patients
described above. who were receiving or had received fingolimod therapy
Three of the studies mentioned above also indicated that [35]. Among the 35 known outcomes, there were 5 spon-
IFNb may be associated with a lower birth weight com- taneous abortions (14 %), 14 elective abortions, and 16
pared to unexposed control patients [20, 30, 33]. Con- successful births. One infant was born with unilateral
versely, other studies found no effect on birth weights [24, congenital posteromedial bowing of the tibia, and there was
26, 31, 34]. In women taking IFNb (n = 7) or GA (n = 3) one elective abortion following detection of tetralogy of
throughout pregnancy, birth weights were slightly higher in Fallot, the most common congenital cyanotic heart defect,
the IFNb-exposed pregnancies [24], but other small studies and one case of acrania in an ongoing pregnancy. The
found no statistical difference in birth weight with IFNb reported number of pregnancies is not sufficient to draw
exposure [34]. A retrospective analysis of 7 women who any definitive conclusions from these registry data.
became pregnant, while taking IFNb also did not detect an Fingolimod (at a 0.5 mg dose) does not significantly
effect of IFNb on birth weight [31]. None of these studies impact the pharmacokinetics of oral contraceptives and
distinguished among the different IFNb formulations. does not compromise contraceptive efficacy [37]. Based on
Taken together, this information indicates that IFNb is the potential risk of fetal harm with fingolimod, which may
not teratogenic. Collectively, reports on over 1,500 preg- take approximately 2 months to be completely eliminated
nant women [21, 24, 2831] indicate that IFNb exposure is from the body (Table 3), the US prescribing information
not associated with an increased risk of spontaneous advises women of childbearing potential to use effective
abortion. Other observational studies or analyses involving contraception to avoid pregnancy during and for 2 months
fewer than 120 patients treated with IFNb yielded after stopping fingolimod treatment [35].

123
J Neurol (2013) 260:12021214 1207

Table 3 DMTs currently approved for use in MS: general characteristics and pregnancy and lactation risks
Generic (brand) name Chemical structure T FDA EMA Lactation
(elimination) categorya statementb categoryc

GA (Copaxone, Teva Pharmaceutical L-glutamic acid polymer with *20 h B 2 L3


Industries) L-alanine, L-lysine, L-tyrosine,
molecular weight 59 kDa
IM IFNb-1a (Avonex, Biogen Idec) 166-aa glycoprotein, molecular *10 h C 2 L3
weight 23 kDa
SC IFNb-1a (Rebif, Merck Serono) 166-aa glycoprotein, molecular 69 37 h C 2 L3
weight 23 kDa
IFNb-1b (Betaseron/Betaferon, Bayer 165-aa protein product, molecular 8 min4.3 h C 2 L3
HealthCare Pharmaceuticals; Extavia, weight 19 kDa
Novartis)
TM
Fingolimod (Gilenya , Novartis) 2-amino-2-[2-(4-octylphenyl) ethyl] 69 days C 2 L4
propan-1,3-diol hydrochloride,
molecular weight 344 kDa
Natalizumab (Tysabri, Biogen Idec) Recombinant humanized IgG4j 11 4 days C 2 L3
monoclonal antibody, molecular
weight 149 kDa
Mitoxantrone (Novantrone, EMD Serono) Synthetic anthracenedione 23215 h D 2 L5
C22H28N4O62HCl, molecular weight
517 kDa
DMT disease-modifying therapy, MS multiple sclerosis, T half-life, FDA US Food and Drug Administration, EMA European Medicines
Agency, GA glatiramer acetate, kDa kilodaltons, IM intramuscular, IFNb interferon beta, aa amino acid, SC subcutaneous, IgG4j immuno-
globulin G4j
a
Based on risk of teratogenic effects, medications are classified by the FDA and listed in each drugs respective package insert as pregnancy
category A, B, C, D or X; please see each drugs respective package insert [16, 2426, 34, 38, 46]
b
Indicates the statement in Table 1 most similar to the statement in the product insert regarding pregnancy risk
c
Lactation risk categories are currently not listed in drug package inserts; DMT classifications based on lactation risk as L1 (safest), L2 (safer),
L3 (moderately safe), L4 (possibly hazardous) or L5 (contraindicated) are from Hale et al. [16]

Natalizumab (natalizumab) Pregnancy Exposure Registry (TPER) sug-


gests that treatment has no apparent adverse effect on
The FDA has given natalizumab a pregnancy category C pregnancy outcomes in humans [44]. As of 23 May 2011, a
rating based on a guinea pig study showing a small total of 341 pregnant patients were enrolled prospectively
reduction in pup survival, when the drug was administered in TPER, there were 277 known pregnancy outcomes. The
at 30 mg/kg (7 times the human dose based on body reported 11 % preliminary rate of spontaneous abortions
weight). In addition, a primate study showed hematologic [44] is consistent with the expected US population rate of
effects on the fetus (mild anemia, reduced platelet count, 15 % for spontaneous pregnancy loss in recognized preg-
increased spleen weight, and reduced liver and thymus nancies at \20 weeks gestation [19]. Malformations
weights) when the drug was administered at 2.3 times the occurred in 23 pregnancy outcomes in 21 of 277 women.
human dose [3840]. These hematological effects resolved Malformations of the male genitalia, umbilical hernias,
following natalizumab discontinuation. Both studies plagiocephaly, hemangioma, and cardiovascular defects
revealed no evidence of teratogenic effects at seven times (ventricular septal defect, patent ductus arteriosus, and
the human dose (30 mg/kg) [4042]. tetralogy of Fallot) were recorded, but these cases were
The EMA guidance on natalizumab use during preg- isolated and some were confounded by other risk factors,
nancy states that data on the use of natalizumab in pregnant they did not suggest any drug-related pattern [44].
women are currently inadequate and that natalizumab Data from this registry are consistent with other findings
should not be used during pregnancy unless the clinical reported in a small number of pregnant women with
condition of the woman requires natalizumab treatment natalizumab exposure. Hoevenaren et al. [45] reported on
[43]. the pregnancy outcomes of 2 women with MS. One patient
Although there are no well-controlled studies of natal- used natalizumab, while trying to become pregnant and the
izumab efficacy and safety in pregnant women, a pre- other received natalizumab prior to becoming pregnant and
liminary review of data from the prospective Tysabri throughout gestation. The newborns did not have any

123
1208 J Neurol (2013) 260:12021214

abnormalities at birth and showed normal development at developing fetus have been observed with structurally
6 weeks follow-up. The patient who received continuous related agents [49].
natalizumab experienced no MS relapses during preg- In preclinical models, administration of mitoxantrone to
nancy. In another recently published article, Hellwig et al. rats during the organogenesis period of pregnancy was
[46] followed 35 women with MS who accidentally associated with fetal growth retardation at doses of
became pregnant, while taking natalizumab. All patients [0.1 mg/kg/day (0.01 times the recommended human dose
stopped natalizumab as soon as they became aware of their on a mg/m2 basis) [49]. Similarly, when pregnant rabbits
pregnancy. Patients included in the study had received were treated during organogenesis, mitoxantrone treatment
natalizumab for at least 8 weeks prior to their last menses. increased the rate of premature delivery at doses of
The proportion of spontaneous abortions was 14 % (5 of [0.1 mg/kg/day (0.01 times the recommended human dose
35), which is similar to what was seen in the natalizumab on a mg/m2 basis). Although no teratogenic effects were
registry [40] and is in line with the US population rates observed in these studies, the maximum doses tested were
[19]. All of the remaining 29 pregnancies were healthy well below the recommended human dose (0.02 and 0.05
newborns, except for a boy born with hexadactyly. times the recommended human dose on a mg/m2 basis in
Although the babies from natalizumab-exposed pregnan- rats and rabbits, respectively).
cies tended to have a lower mean birth weight than those Just three case reports on clinical outcomes during
from a non DMT-treated control group (3,159 vs 3,406 g), pregnancy with mitoxantrone have been published, and
all newborns had birth weights within the normal range. only two of these were in women with MS [5052]. In the
Interestingly, after natalizumab was withdrawn, MS first case, a woman with MS received mitoxantrone until
activity did not rebound either during pregnancy or post the 29th week of her pregnancy [50]. Fetal growth was
partum. Furthermore, natalizumab-exposed women tended restricted; however, the baby had no evidence of fetal
to have fewer relapses than non DMT-exposed women malformations. In the second case, a patient with MS was
during pregnancy, while the number of relapses after exposed to mitoxantrone during the conception period and
pregnancy did not differ significantly between the two delivered a child with Pierre Robin sequence, a syndrome
groups. characterized by glossoptosis, micrognathia, and cleft pal-
Three cases of first-trimester exposure to natalizumab ate [50]. The authors concluded that a causal relation with
were recently reported [47, 48]. All three pregnancies mitoxantrone could be possible. In the third case, involving
developed without any complications, with normal fetus a non-MS patient with acute myeloid leukemia, the new-
growth and full-term delivery (one spontaneous birth and born showed no congenital malformations [51].
two cesarean births). Two of the women experienced As a consequence of the available information with
relapses in the immediate postpartum period, but reinitia- mitoxantrone, the US prescribing information states that all
tion of treatment with natalizumab stopped disease activity. women of childbearing potential should have a pregnancy
In addition, 2 patients from the authors clinic were test performed prior to each dose of the drug and should be
exposed to natalizumab during the first 6 weeks of preg- advised against becoming pregnant while taking this
nancy. One pregnancy resulted in a healthy delivery, the cytotoxic agent.
other ended in miscarriage (M. Houtchens, unpublished
data). Collectively, there are only a few published cases on Effects of approved MS therapies on fertility
the usage of natalizumab during pregnancy. Monitoring
patients through the prospective natalizumab pregnancy The potential influence of DMTs for MS on fertility was
registry should provide needed information on the safety of also reviewed. GA has no known effects on human fertility.
natalizumab exposure during pregnancy. In a multigeneration reproduction and fertility study in rats,
GA at SC doses of up to 36 mg/kg (18 times the human
Mitoxantrone therapeutic dose on a mg/m2 basis) had no adverse effects
on reproductive parameters including fertility [16]. Simi-
Mitoxantrone is a cytotoxic chemotherapeutic agent larly, IFNb-1a has no known effects on human fertility.
approved for the treatment of secondary progressive MS. It Subcutaneously administered IFNb-1a showed no adverse
intercalates with DNA and inhibits topoisomerase II, and it effects on fertility in rhesus monkeys at approximately nine
is considered a potential human teratogen. The FDA has times the recommended human dose [25, 26]. Following
given mitoxantrone a pregnancy category D rating, and the administration of oral fingolimod (0, 1, 3 or 10 mg/kg/day)
products US package insert states that women who are to male and female rats prior to and during mating, and
pregnant or are trying to become pregnant should not use continuing to day 7 of gestation in females, no effect on
mitoxantrone [49]. The EMA has not approved mitoxan- fertility was observed up to the highest fingolimod dose
trone for the treatment of MS. Adverse effects on the tested (10 mg/kg, which is approximately 200 times the

123
J Neurol (2013) 260:12021214 1209

recommended human dose on a mg/m2 basis) [35]. With fluorine at the 9a position, cross the placenta with minimal
natalizumab, reductions in female guinea pig fertility have metabolism, leading to likely direct full-dose effects on the
been observed at dose levels of 30 mg/kg, but not at the fetus [56].
10 mg/kg dose level (2.3 times the clinical dose) [42]. Intravenous immunoglobulin (IVIG) administration has
Natalizumab did not affect male guinea pig fertility at been reported to reduce the MS relapse rate during preg-
doses up to seven times the recommended human dose. No nancy and post partum, allowing concomitant breastfeed-
controlled studies have reported the effects of natalizumab ing with no serious adverse effects on the infant [57]. The
on human fertility [40]. Finally, in four randomized, con- Gammaglobulin Post Partum (GAMPP) study investigated
trolled clinical trials of mitoxantrone in MS patients, two IVIG doses (avoiding placebo control, because of
infertility and amenorrhea in female patients were among ethical concerns) and concluded that IVIG application
the most frequently reported short-term adverse events likely had beneficial effects, since the relapse rate did not
[53]. increase post partum [58]. More recently, Hellwig et al.
[59] have assessed the comparative efficacy of IVIG
Therapies not approved for use in MS patients application, treatment with other immunomodulatory
compounds and no treatment on the postpartum relapse
A number of therapeutic agents are used off-label for the rate. The reduction in relapse rate showed a trend in favor
treatment of MS including intravenous immunoglobulin of the IVIG-treated women. However, analyzing the
therapy, immunosuppressants, and monoclonal antibodies. expected number of relapses, IVIG-treated patients had
These agents are generally approved for the treatment of significantly fewer postpartum relapses than the untreated
other conditions and have been or are being investigated control group matched for disease activity before and
for MS. As such, they are typically utilized with patients during pregnancy (Chi-square test, p = 0.013).
who have failed to adequately respond to an approved
DMT. Table 3 summarizes the FDA pregnancy risk cate-
gory and EMA pregnancy risk statement for each thera- MS therapies and breastfeeding
peutic. There are only a few clinical reports on the use of
these agents during pregnancy that specify adverse out- Breastfeeding is not contraindicated in women with MS.
comes. Data on the effects of these agents on fertility in Most studies have reported that breastfeeding has no effect
MS patients are also not yet available. on MS relapses [9, 6063]. In other studies by Langer-
Gould et al. [64] and Hellwig et al. [65, 66], exclusive
Corticosteroid therapy for acute relapse treatment breastfeeding in the postpartum period actually reduced the
risk of postpartum relapses. A detailed discussion of the
In MS, intravenous corticosteroids are frequently used for potential influence of breastfeeding on postpartum MS
the treatment of acute relapses. Adequate human repro- relapses is beyond the scope of the current review, which
duction studies have not been performed with corticoste- focuses on MS drug exposure and pregnancy.
roids. However, they are broadly used in obstetric medicine Although a 24-h suspension of breastfeeding has his-
to treat fetuses at risk of preterm birth to prevent respira- torically been recommended following exposure to intra-
tory distress syndrome and to aid lung maturity [54]. Their venous contrast agents such as gadolinium-based agents as
use in this context does not appear to be associated with a a precautionary measure, because of the lack of data on
negative impact on the development of the fetus [55], and levels of gadolinium excreted in breast milk and the risk to
intravenous corticosteroids are thus probably safe in the newborns, recent evidence-based guidelines for the use of
second and third trimesters of pregnancy. Steroids cross the computerized tomography, magnetic resonance imaging
placental barrier and may increase the risk of cleft palate (MRI), and contrast media during pregnancy state that it
when used in the first trimester [56] or may cause low birth seems to be safe to continue breastfeeding immediately
weight [55], and they could in theory delay healing for the after receiving iodinated contrast or gadolinium [67, 68].
mother after birth. Therefore, corticosteroids should prob- Therefore, the mother should generally be allowed to
ably be reserved for treatment of serious acute exacerba- choose, whether or not to breastfeed based on her personal
tions. Prednisone, prednisolone, and methylprednisolone preference as long as her illness has not been particularly
can be administered with low levels of fetal exposure aggressive prior to conception. In women who were taking
and may be preferred for use during pregnancy [55, 56]. a DMT before pregnancy, we advise not resuming therapy
These agents are metabolized to inactive forms by 11-b- until after the mother has finished breastfeeding her infant
hydroxysteroid dehydrogenase in the placenta, allowing [14]. In a woman with difficult-to-control disease prior to
less than 10 % of the maternal dose to reach the fetus. In pregnancy, an argument can be made in favor of rapid
contrast, betamethasone and dexamethasone, which have reintroduction of immunomodulating therapy in order to

123
1210 J Neurol (2013) 260:12021214

Table 4 DMTs not currently approved for use in MS


Generic (brand) name Approved indication(s) FDA EMA Lactation
categorya statementb categoryc

Antibodies
Alemtuzumab (Campath/MabCampath, Genzyme Corporation, B cell lymphocytic leukemia C 1 L4
Cambridge, MA, USA)
Daclizumab (Zenapax, Hoffmann-La Roche, Basel, Switzerland) Prophylaxis of organ rejection in C 1d Unknown
renal transplants
Rituximab (MabThera, Hoffmann-La Roche; Rituxan, Non-Hodgkin lymphoma, C 1 L3
Biogen Idec) follicular lymphoma, RA
Immunoglobulin therapy
Intravenous immunoglobulin (various) Immune deficiencies, autoimmune C 2 L2
disorders, acute infections
Immunosuppressants
Azathioprine (Imuran, GlaxoSmithKline, London, UK; Azasan, Autoimmune diseases, RA, D 1 L3
Salix Pharmaceuticals, Morrisville, NJ, USA) prevention of organ and tissue
rejection
Cyclophosphamide (Endoxan, Baxter Oncology, Halle Malignancies, bone marrow D 1 L5
Westfalen, Germany; Cytoxan, E.R. Squibb and Sons, transplantation
Plainsboro, NJ, USA; Neosar, Pfizer, New York, NY,
USA; Procytox, Baxter Corporation, Mississauga,
TM
Ontario, Canada; Revimmune , Accentia Biopharmaceuticals,
Tampa, FL, USA)
Methotrexate (Rheumatrex, Dava Pharmaceuticals, Fort Lee, NJ, Neoplastic diseases, psoriasis, RA X 1 L3
TM
USA; Trexall , Duramed Pharmaceuticals, Pomona, NY, USA)
Mycophenolate mofetil (Cellcept, Genentech, South San Prophylaxis of tissue graft D 1 L4
Francisco, CA, USA) rejections
DMT disease-modifying therapy, MS multiple sclerosis, FDA US Food and Drug Administration, EMA European Medicines Agency,
RA rheumatoid arthritis
a
Based on risk of teratogenic effects, medications are classified by the FDA and listed in each drugs respective package insert as pregnancy
category A, B, C, D or X; please see each drugs respective package insert [17, 2527, 35, 40, 49]
b
Indicates the statement in Table 1 most similar to the statement in the product insert regarding pregnancy risk
c
Lactation risk categories are currently not listed in drug package inserts; DMT classifications based on lactation risk as L1 (safest), L2 (safer),
L3 (moderately safe), L4 (possibly hazardous) or L5 (contraindicated) are from Hale et al. [16]
d
The marketing authorization for Zenapax has been withdrawn at the request of the marketing authorization holder

avoid the compounded risk of postpartum exacerbation and 30 lg/week showed that even using the highest value
a rapid return to an active prepregnancy disease state. measured in breast milk (179 pg/mL), the estimated rel-
ative infant dose would be 0.006 % of the maternal dose.
Approved DMTs and breastfeeding No side effects were noted in any of the breastfed infants
[69]. These data suggest only subclinical levels of IFNb-1a
Lactation risk categories are not currently included in US levels in human milk following IM dosing.
or EMA drug package inserts. As an alternative, the lac- However, clinical data on the use of GA or other IFNb
tation risk categories for each approved DMT were formulations during breastfeeding are limited. Fragoso
obtained from the authoritative resource Medications and et al. [22] reported on 9 mothers who breastfed their babies
Mothers Milk [16] and are presented in Table 3. Excretion for a mean period of 3.6 months while taking GA. No
of GA, IM IFNb-1a, SC IFNb-1a or SC IFNb-1b in human infections, signs of inadequate digestion or other significant
milk is not well established [17, 2527]. The molecular ill effects were observed in these breastfed children during
weight of these compounds may limit their transfer to or after breastfeeding. Hellwig and Gold [24] followed
maternal milk [24] and they are likely to be depolymerized, 3 mothers taking GA and 1 mother taking IFNb during
if ingested orally so toxicity is unlikely [23]. In one recent breastfeeding, again without any noticeable problems.
report [69], systematic assessment of IM IFNb-1a transfer Although approved injectable DMTs are unlikely to have a
to human breast milk in 6 women receiving IM IFNb-1a negative effect during breastfeeding, the decision on using

123
J Neurol (2013) 260:12021214 1211

Table 5 Author recommendations on the use of MS therapies during pregnancy and breastfeeding
Guidance for physicians counseling women receiving MS treatment who want to become pregnant

Counsel all women of childbearing age on family planning and conception planning
Provide expedited referral to infertility specialists, if a woman is unable to conceive and is remaining off therapy for extended periods of
time while attempting pregnancy
Consider monthly intravenous high-dose steroid treatment to coincide with the menstrual period in a woman who is attempting conception
and is off standard DMTs
IFNb and GA therapies should be discontinued for 1 month (elimination half-life 8 min4.5 days) prior to attempting conception. A longer
period (at least 23 months; elimination half-life 615 days) is recommended for natalizumab, fingolimod, and mitoxantrone. (See Table 1)
Women who wish to continue taking a DMT during pregnancy should avoid mitoxantrone and fingolimod. For the other DMTs, women and
their clinicians should weigh up the risks and benefits based on the following information:
Preliminary data on GA, IFNb, and natalizumab exposure and pregnancy outcomes have not shown any specific patterns of malformations
suggestive of teratogenicity, but there is currently insufficient evidence for definite conclusions
Among the IFNb formulations, IM or SC IFNb-1a is preferred due to the availability of safety data from pregnancy registries, which are
lacking for IFNb-1b
Limited preclinical data and findings in a limited number of patients suggest that IFNb may be associated with an increase in the rate of
spontaneous abortions. However, this effect was not seen in over 1,500 women enrolled in pregnancy registries and other studies
GA and natalizumab may be associated with reduced relapses during pregnancy, according to limited evidence. Comparable data are not
available for IFNb. Natalizumab should be used during pregnancy only, if the potential maternal benefit justifies the potential risk to the
fetus
Intravenous prednisone, prednisolone, and methylprednisolone are moderately safe in the second and third trimesters of pregnancy for the
treatment of serious acute exacerbations
Reassure patients that methods of obstetrical anesthesia and mode of delivery (vaginal versus cesarean section) are not related to the
likelihood of postpartum relapse [78, 79]
Guidance for physicians counseling women receiving MS treatment who unexpectedly become pregnant
Women should be advised to discontinue their DMT treatment if they unexpectedly become pregnant. This particularly applies to
mitoxantrone and fingolimod. Women taking GA and IFNb should be counseled that preliminary data on GA, IFNb, and natalizumab
exposure and pregnancy outcomes have not shown any specific patterns of malformations suggestive of teratogenicity, but there is currently
insufficient evidence for definite conclusions
If there is documented exposure to high-risk approved or off-label compounds in the first trimester, the patient should be expeditiously
referred to a high-risk maternal fetal medicine specialist for ongoing pregnancy follow-up
The same recommendations outlined in the section above apply to those who unexpectedly become pregnant and wish to continue taking
their DMT
Guidance for physicians counseling women with MS treatment who wish to breastfeed their infant
The transfer of GA into breast milk is unlikely, and IFNb was reported to be present only in extremely small quantities [66]. Natalizumab
has also been detected in human breast milk [38]. However, clinical data on the use of these drugs during breastfeeding and potential effects
on infants are currently lacking. Therefore, women considering taking one of these agents during breastfeeding should weigh the potential
risks as well as the benefits
In patients with a high risk of postpartum relapse, intravenous immunoglobulin is safe during breastfeeding and may be associated with
reduction in postpartum relapses [56, 80, 81]
Corticosteroids are moderately safe for women experiencing an acute relapse while breastfeeding, and they may reduce relapse rates when
used as monthly high-dose infusions. However, a washout period should be applied
For high intravenous doses, mothers should pump and discard breast milk for 2448 h after their infusion before resuming nursing
Fingolimod and mitoxantrone should be avoided during breastfeeding
It is safe to continue breastfeeding after receiving iodinated or gadolinium-contrast agents for MRI [64, 65]
DMT disease-modifying therapy, IFNb interferon beta, GA glatiramer acetate, IM intramuscular, SC, subcutaneous, MRI magnetic resonance
imaging

IFNb or GA during lactation should be made jointly by the excretion into milk comes from experiments in treated rats
neurologist and the obstetrician, and the current practice [35]. For mitoxantrone and natalizumab, excretion into
standards advise against the use of any injectable MS milk has been observed in humans [40, 49]. As mitoxan-
therapies in breastfeeding mothers. trone is a cytotoxic agent, it has a high risk of causing
There is some evidence that other approved agents significant negative effects on the infant and its use is
(fingolimod, natalizumab, and mitoxantrone) are excreted contraindicated during lactation. The use of fingolimod
into milk [35, 40, 49]. For fingolimod, the evidence for during breastfeeding should also be avoided, given that it is

123
1212 J Neurol (2013) 260:12021214

orally bioavailable and takes a long time to clear. Although at high risk for postpartum exacerbation, but choose to
natalizumab is not orally bioavailable, it has been detected breastfeed their infants and are not comfortable returning to
in breast milk [40] and the effects of exposure on new- their platform prepregnancy therapy while nursing.
borns/infants are unknown, so caution is required.

Other therapies and breastfeeding Conclusions

The lactation risk categories for other common therapies Historically, women with MS were discouraged from
that are not approved for MS are summarized in Table 4. having children based on unsubstantiated concerns about
Of these therapies, intravenous immunoglobulin may be the potential effects of pregnancy on the mothers disease
particularly useful during breastfeeding to reduce post- and the potential effects of MS on the fetus. Mounting
partum relapse rate without causing adverse effects to the evidence has shown that pregnancy has no long-term
infant [57, 58, 70]. A double-blind, randomized trial in negative effects on disease course and that maternal MS is
Europe investigated intravenous immunoglobulin in 168 not associated with an increased risk of birth defects or
postpartum mothers with MS [57]. Mothers who breastfed developmental delays [6, 7780]. Though data on the
for 3 months or longer while taking this therapy were more efficacy and/or safety of MS treatments during pregnancy
likely to be relapse-free during the study than those who and breastfeeding remain limited, completed or ongoing
did not breastfeed or who breastfed for less than 3 months. registry studies are providing essential real-world data
No adverse effects on the breastfed infants were reported. allowing healthcare professionals and patients to weigh the
In addition, a case series in 43 breastfeeding women with benefit of specific MS treatments against the potential risks
MS who received intravenous immunoglobulin within to the fetus. Based on data from these registries, other
3 days of delivery and monthly thereafter found that the published studies and analyses, and our own clinical
relapse rate was lower in the studied patients than in his- experience, we have developed clinical considerations on
torical controls [71]. The only reported adverse effect was the use of DMTs and corticosteroids during pregnancy and
a transient rash, possibly caused by the administration of breastfeeding (Table 5). Ultimately, the treating physician
immunoglobulin. The potential benefits of postpartum and the patient involved must carefully consider the deci-
intravenous immunoglobulin on relapse rate have also been sion to either discontinue or continue using a particular
seen in another smaller study [72]. agent during pregnancy or breastfeeding.
Breastfeeding by women on glucocorticoid therapy is
generally considered safe, and there appears to be minimal Acknowledgments Medical writing assistance was provided by
Christopher Barnes and editorial support was provided by Joshua
excretion of corticosteroids into breast milk [7375]. In one Safran, both of Infusion Communications. Their work was funded by
study, it was calculated that small-to-moderate doses of Biogen Idec Inc. The authors were not compensated and retained full
corticosteroids ingested from milk (e.g., prednisolone editorial control.
80 mg daily) would add, at most, 10 % to the infants
Conflicts of interest Dr. Houtchens has served on scientific advi-
endogenous corticosteroid production, which was expected sory boards and received compensation for her work as a consultant
to have little clinical significance [74]. However, since for EMD Serono, Biogen Idec, Novartis, Acorda, and Teva Pharma-
steroids can cause adverse events in children including ceutical Industries. Dr. Kolb has received compensation as a paid
growth retardation [55], a washout period should be consultant for Teva Neuroscience.
applied between dosing and nursing. Steroid levels in
breast milk peak 2 h after administration and decline rap-
idly thereafter. Therefore, exposure may be minimized, if References
nursing is performed 4 h after a low-to-moderate dose is
1. Grima DT, Torrance GW, Francis G, Rice G, Rosner AJ,
taken [55]. For higher intravenous doses (e.g., of monthly Lafortune L (2000) Cost and health related quality of life con-
methylprednisolone), it may be prudent for mothers to wait sequences of multiple sclerosis. Mult Scler 6:9198
2448 h after their infusion before nursing, using a breast 2. Naci H, Fleurence R, Birt J, Duhig A (2010) Economic burden of
pump in the interim. These women should be instructed to multiple sclerosis: a systematic review of the literature. Phar-
macoeconomics 28:363379
plan ahead and save breast milk ahead of time to continue 3. Asche CV, Singer ME, Jhaveri M, Chung H, Miller A (2010) All-
to use it, while they are transiently unable to nurse. cause health care utilization and costs associated with newly
In the postpartum period, pulses of monthly intravenous diagnosed multiple sclerosis in the United States. J Manag Care
methylprednisolone for 6 months led to a reduced rate of Pharm 16:703712
4. Naci H, Fleurence R, Birt J, Duhig A (2010) The impact of
relapses relative to an untreated group (p = 0.018), sug- increasing neurological disability of multiple sclerosis on health
gesting a beneficial effect for this therapy in this setting utilities: a systematic review of the literature. J Med Econ
[76]. A similar approach can be utilized in women who are 13:7889

123
J Neurol (2013) 260:12021214 1213

5. Weinshenker BG, Bass B, Rice GP et al (1989) The natural 26. Avonex (intramuscular interferon beta-1a) prescribing informa-
history of multiple sclerosis: a geographically based study I. tion (2011) http://www.avonex.com/pdfs/pi-powder.pdf. Accessed
Clinical course and disability. Brain 112:133146 4 November 2011
6. Confavreux C, Aimard G, Devic M (1980) Course and prognosis 27. Betaseron (interferon beta-1b) prescribing information (2011)
of multiple sclerosis assessed by the computerized data pro- http://berlex.bayerhealthcare.com/html/products/pi/Betaseron_PI.pdf.
cessing of 349 patients. Brain 103:281300 Accessed 4 November 2011
7. Confavreux C, Hutchinson M, Hours MM, Cortinovis-Tourniaire 28. Richman S, Wallace K, Liu S, Sperling B (2012) Pregnancy
P, Moreau T (1998) Rate of pregnancy-related relapse in multiple outcomes from the Avonex (interferon beta-1a) pregnancy
sclerosis. Pregnancy in multiple sclerosis group. N Engl J Med exposure registry. Neurology 78:P06.191
339:285291 29. Sandberg-Wollheim M, Alteri E, Moraga MS, Kornmann G
8. Vukusic S, Hutchinson M, Hours M et al (2004) Pregnancy and (2011) Pregnancy outcomes in multiple sclerosis following sub-
multiple sclerosis (the PRIMS study): clinical predictors of post- cutaneous interferon beta-1a therapy. Mult Scler 17:423430
partum relapse. Brain 127:13531360 30. Amato MP, Portaccio E, Ghezzi A et al (2010) Pregnancy and
9. Vukusic S, Confavreux C (2006) Pregnancy and multiple sclerosis: fetal outcomes after interferon-beta exposure in multiple sclero-
the children of PRIMS. Clin Neurol Neurosurg 108:266270 sis. Neurology 75:17941802
10. Niino M, Hirotani M, Fukazawa T, Kikuchi S, Sasaki H (2009) 31. Patti F, Cavallaro T, Lo Fermo S et al (2008) Is in utero early-
Estrogens as potential therapeutic agents in multiple sclerosis. exposure to interferon beta a risk factor for pregnancy outcomes
Cent Nerv Syst Agents Med Chem 9:8794 in multiple sclerosis? J Neurol 255:12501253
11. Stenager E, Stenager EN, Jensen K (1994) Effect of pregnancy on 32. Sandberg-Wollheim M, Frank D, Goodwin TM et al (2005)
the prognosis for multiple sclerosis: a 5-year follow up investi- Pregnancy outcomes during treatment with interferon beta-1a in
gation. Acta Neurol Scand 90:305308 patients with multiple sclerosis. Neurology 65:802806
12. Dhooghe MB, Nagels G, Uitdehaag BM (2010) Long-term 33. Boskovic R, Wide R, Wolpin J, Bauer DJ, Koren G (2005) The
effects of childbirth in MS. J Neurol Neurosurg Psychiatry reproductive effects of beta interferon therapy in pregnancy:
81:3841 a longitudinal cohort. Neurology 65:807811
13. National MS Society (2011) Pregnancy, delivery, and the post- 34. Hellwig K, Haghikia A, Gold R (2010) Parenthood and immuno-
partum period: information for women with MS to share with modulation in patients with multiple sclerosis. J Neurol 257:580583
their physicians. http://www.nationalmssociety.org/living-with- 35. Gilenya (fingolimod) US prescribing information (2011)
multiple-sclerosis/healthy-living/pregnancy/index.aspx. Accessed http://www.pharma.us.novartis.com/product/pi/pdf/gilenya.pdf.
12 August 2011 Accessed 4 November 2011
14. Coyle PK, Christie S, Fodor P et al (2004) Multiple sclerosis 36. Gilenya (fingolimod) European public assessment report (2012)
gender issues: clinical practices of women neurologists. Mult http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/
Scler 10:582588 human/medicines/002202/human_med_001433.jsp&mid=WC0b
15. Davis DB (2010) Drugs in pregnancythe issues for 2010. 01ac058001d124. Accessed 22 June 2012
J Popul Ther Clin Pharmacol 17:e332e335 37. Collins W, Francis G, Koren G et al (2011) Lack of interaction
16. Hale TW (2010) Medications and mothers milk, 14th edn. Hale between fingolimod (FTY720) and oral contraceptives, and
Publishing, Amarillo pregnancy experience in the clinical program of fingolimod in
17. Copaxone (glatiramer acetate) prescribing information (2011) multiple sclerosis. Neurology 76(Suppl 4):A609
http://www.sharedsolutions.com/pdfs/ 38. Wehner NG, Shopp G, Osterburg I, Fuchs A, Buse E, Clarke J
prescribinginformation.aspx. Accessed 4 November 2011 (2009) Postnatal development in cynomolgus monkeys following
18. Coyle P, Johnson K, Pardo L (2003) Pregnancy outcomes in prenatal exposure to natalizumab, an alpha4 integrin inhibitor.
patients with multiple sclerosis treated with glatiramer acetate Birth Defects Res B Dev Reprod Toxicol 86:144156
(Copaxone). 19th Congress of the European Committee for 39. Wehner NG, Shopp G, Oneda S, Clarke J (2009) Embryo/fetal
Treatment and Research in Multiple Sclerosis (ECTRIMS), development in cynomolgus monkeys exposed to natalizumab, an
Milan, 1720 September 2003 alpha4 integrin inhibitor. Birth Defects Res B Dev Reprod Tox-
19. Ventura SJ, Curtin SC, Abma JC (2012) Estimated pregnancy icol 86:117130
rates and rates of pregnancy outcomes for the United States, 40. Tysabri (natalizumab) prescribing information (2011) http://
19902008. Nat Vital Statistics Rep 60:122 www.accessdata.fda.gov/drugsatfda_docs/label/2011/125104s06
20. Weber-Schoendorfer C, Schaefer C (2009) Multiple sclerosis, 58lbl.pdf. Accessed 6 December 2011
immunomodulators, and pregnancy outcome: a prospective 41. Wehner NG, Shopp G, Rocca MS, Clarke J (2009) Effects of
observational study. Mult Scler 15:10371042 natalizumab, an alpha4 integrin inhibitor, on the development of
21. Finkelsztejn A, Fragoso YD, Ferreira ML et al (2011) The Bra- Hartley guinea pigs. Birth Defects Res B Dev Reprod Toxicol
zilian database on pregnancy in multiple sclerosis. Clin Neurol 86:98107
Neurosurg 113:277280 42. Wehner NG, Skov M, Shopp G, Rocca MS, Clarke J (2009)
22. Fragoso YD, Finkelsztejn A, Kaimen-Maciel DR et al (2010) Effects of natalizumab, an alpha4 integrin inhibitor, on fertility in
Long-term use of glatiramer acetate by 11 pregnant women with male and female guinea pigs. Birth Defects Res B Dev Reprod
multiple sclerosis: a retrospective, multicentre case series. CNS Toxicol 86:108116
Drugs 24:969976 43. Tysabri (natalizumab) European public assessment report (2011)
23. Salminen HJ, Leggett H, Boggild M (2010) Glatiramer acetate http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/
exposure in pregnancy: preliminary safety and birth outcomes. human/medicines/000603/human_med_001119.jsp&mid=WC0b
J Neurol 257:20202023 01ac058001d124. Accessed 22 June 2012
24. Hellwig K, Gold R (2011) Glatiramer acetate and interferon-beta 44. Cristiano LM, Bozic C, Bloomgren G (2011) Preliminary eval-
throughout gestation and postpartum in women with multiple uation of pregnancy outcomes from the Tysabri (natalizumab)
sclerosis. J Neurol 258:502503 pregnancy exposure registry. Mult Scler 17:S457
25. Rebif (subcutaneous interferon beta-1a) prescribing information 45. Hoevenaren IA, de Vries LC, Rijnders RJ, Lotgering FK (2011)
(2011) http://www.emdserono.com/cmg.emdserono_us/en/images/ Delivery of healthy babies after natalizumab use for multiple
rebif_tcm115_19765.pdf. Accessed 4 November 2011 sclerosis: a report of two cases. Acta Neurol Scand 123:430433

123
1214 J Neurol (2013) 260:12021214

46. Hellwig K, Haghikia A, Gold R (2011) Pregnancy and natal- 63. Jalkanen A, Airas L, Marttila RJ (2011) To breastfeed or not to
izumab: results of an observational study in 35 accidental preg- breastfeed: effect of breastfeeding on postpartum disease activity
nancies during natalizumab treatment. Mult Scler 17:958963 in multiple sclerosis revisited. Neurology 76(Suppl 4):A610
47. Mattioda A, Masera S, Romagnolo A et al (2011) Healthy baby 64. Langer-Gould A, Huang SM, Gupta R et al (2009) Exclusive
delivery after conception during natalizumab exposure: a case breastfeeding and the risk of postpartum relapses in women with
report. Mult Scler 17:S455S456 multiple sclerosis. Arch Neurol 66:958963
48. Totaro R, Rossi M, Casalena A, Carolei A (2011) Pregnancy, 65. Hellwig K, Kuge M, Gold R, Langer-Gould A (2011) Exclusive
delivery, and birth outcome after natalizumab use for multiple breastfeeding reduces the risk of postpartum relapses a prospec-
sclerosis: a report of two cases. Mult Scler 17:S227S228 tive study from the German MS and pregnancy registry. Neu-
49. Novantrone (mitoxantrone) prescribing information (2011) http:// rology 76(Suppl 4):A273
www.accessdata.fda.gov/drugsatfda_docs/label/2010/019297s03 66. Hellwig K, Haghikia A, Gold R (2011) Multiple sclerosis and
3s034lbl.pdf. Accessed 4 November 2011 pregnancy experience from a nationwide database. Neurology
50. De Santis M, Straface G, Cavaliere AF, Rosati P, Batocchi AP, 76(Suppl 4):A273
Caruso A (2007) The first case of mitoxantrone exposure in early 67. Webb JA, Thomsen HS, Morcos SK, Members of Contrast Media
pregnancy. Neurotoxicology 28:696697 Safety Committee of European Society of Urogenital Radiology
51. Baumgartner AK, Oberhoffer R, Jacobs VR et al (2009) (ESUR) (2005) The use of iodinated and gadolinium contrast
Reversible foetal cerebral ventriculomegaly and cardiomyopathy media during pregnancy and lactation. Eur Radiol 15:12341240
under chemotherapy for maternal AML. Onkologie 32:4043 68. Chen MM, Coakley FV, Kaimal A, Laros RK Jr (2008) Guide-
52. Hellwig K, Schimrigk S, Chan A, Epplen J, Gold R (2011) lines for computed tomography and magnetic resonance imaging
A newborn with Pierre Robin sequence after preconceptional use during pregnancy and lactation. Obstet Gynecol 112(2 Pt
mitoxantrone exposure of a female with multiple sclerosis. 1):333340
J Neurol Sci 307:164165 69. Hale TW, Siddiqui AA, Baker TE (2012) Transfer of interferon
53. Martinelli V, Radaelli M, Straffi L, Rodegher M, Comi G (2009) b-1a into human breastmilk. Breastfeed Med 7:123125
Mitoxantrone: benefits and risks in multiple sclerosis patients. 70. Ferrero S, Esposito F, Pretta S, Ragni N (2006) Fetal risks related
Neurol Sci 30(Suppl 2):S167S170 to the treatment of multiple sclerosis during pregnancy and
54. Roberts D, Dalziel S (2006) Antenatal corticosteroids for accel- breastfeeding. Exp Rev Neurotherapeutics 6:18231831
erating fetal lung maturation for women at risk of preterm birth. 71. Haas J (2000) High dose IVIG in the post partum period for
Cochrane Database Sys Rev 3:CD004454 prevention of exacerbations in MS. Mult Scler 6(Suppl 2):S18
55. Hoes JN, Jacobs JW, Boers M et al (2007) EULAR evidence- S20 (discussion S33)
based recommendations on the management of systemic gluco- 72. Orvieto R, Achiron R, Rotstein Z, Noy S, Bar-Hava I, Achiron A
corticoid therapy in rheumatic diseases. Annals Rheumatic Dis (1999) Pregnancy and multiple sclerosis: a 2-year experience. Eur
66:15601567 J Obstet Gynecol Reprod Biol 82:191194
56. Elliott AB, Chakravarty EF (2010) Immunosuppressive medica- 73. McKenzie SA, Selley JA, Agnew JE (1975) Secretion of pred-
tions during pregnancy and lactation in women with autoimmune nisolone into breast milk. Arch Dis Child 50:894896
diseases. Womens Health 6:431440 74. Ost L, Wettrell G, Bjorkhem I, Rane A (1985) Prednisolone
57. Achiron A, Kishner I, Dolev M et al (2004) Effect of intravenous excretion in human milk. J Pediatrics 106:10081011
immunoglobulin treatment on pregnancy and postpartum-related 75. Greenberger PA, Odeh YK, Frederiksen MC, Atkinson AJ Jr
relapses in multiple sclerosis. J Neurol 251:11331137 (1993) Pharmacokinetics of prednisolone transfer to breast milk.
58. Haas J, Hommes OR (2007) A dose comparison study of IVIG in Clin Pharmacol Therapeut 53:324328
postpartum relapsing-remitting multiple sclerosis. Mult Scler 76. de Seze J, Chapelotte M, Delalande S, Ferriby D, Stojkovic T,
13:900908 Vermersch P (2004) Intravenous corticosteroids in the post-
59. Hellwig K, Beste C, Schimrigk S, Chan A (2009) Immunomod- partum period for reduction of acute exacerbations in multiple
ulation and postpartum relapses in patients with multiple scle- sclerosis. Mult Scler 10:596597
rosis. Ther Adv Neurol Disord 2:711 77. Bennett KA (2005) Pregnancy and multiple sclerosis. Clinical
60. Portaccio E, Ghezzi A, Hakiki B et al (2011) Breastfeeding is not Obstet Gynecol 48:3847
related to postpartum relapses in multiple sclerosis. Neurology 78. Houtchens MK (2007) Pregnancy and multiple sclerosis. Semin
77:145150 Neurol 27:434441
61. Nelson LM, Franklin GM, Jones MC (1988) Risk of multiple 79. Jalkanen A, Alanen A, Airas L (2010) Pregnancy outcome in
sclerosis exacerbation during pregnancy and breast-feeding. women with multiple sclerosis: results from a prospective
JAMA 259:34413443 nationwide study in Finland. Mult Scler 16:950955
62. Hellwig K, Haghikia A, Agne H, Beste C, Gold R (2009) Pro- 80. Stuart M, Bergstrom L (2011) Pregnancy and multiple sclerosis.
tective effect of breastfeeding in postpartum relapse rate of J Midwifery Womens Health 56:4147
mothers with multiple sclerosis. Arch Neurol 66:15801581 81. Ferrero S, Pretta S, Ragni N (2004) Multiple sclerosis: manage-
(author reply 1581) ment issues during pregnancy. Eur J Obstet Gynecol Reprod Biol
115:39

123

Você também pode gostar