You are on page 1of 9

Review Article

Obstetric Medicine
2020, Vol. 13(1) 5–13
The use of biologics for autoimmune ! The Author(s) 2019
Article reuse guidelines:
rheumatic diseases in fertility sagepub.com/journals-permissions
DOI: 10.1177/1753495X19841799
and pregnancy journals.sagepub.com/home/obm

May Ching Soh1,2,3 and Marcelo Moretto4,5

Abstract
In an age where autoimmune rheumatic diseases are successfully managed with biologics, their discontinuation in pregnancy is inadvisable without
careful forethought; maternal disease activity is associated with adverse pregnancy outcomes, which has long-term implications for both mother and
offspring. We aim to provide clinicians with the necessary tools to facilitate decision-making – when a biologic should be used, when it can be
discontinued in pregnancy if appropriate. The pathophysiology of these biologic molecules and their effect on fertility, pregnancy and parturition are
discussed. A summary of the 2016 international guidelines (European League Against Rheumatism and British Society in Rheumatology) on biologics in
pregnancy has been tabulated; more recent publications are discussed in depth. Data on transplacental-transfer ratios and breastmilk excretion rates
are also included. Biologic effects on organogenesis, their implications for the exposed infant in terms of infection risks and vaccination requirements
are included, and future directions for research proposed.

Keywords
Pregnancy, fertility, biologics, infection, vaccination

Date Received: 29 August 2018; accepted: 10 March 2019

What are ‘biologics’? failure during in vitro fertilisation (IVF), recurrent fetal loss and
Unlike chemically synthesised drugs with a known structure, biolog- other features of placental insufficiency such as pre-eclampsia and/
ics are complex molecules largely composed of immunoglobulin (Ig) or fetal growth restriction, often cumulating in preterm deliveries.7,8
G with modified Fc receptors or Fab fragments that bind and neu- TNF is associated with ovarian follicular and embryonic devel-
tralise their target molecule. Suffixes to names of biologics convey opment, activation of apoptotic pathways in fetal and placental cells
specific information concerning their origin and structure, e.g. ‘-mab’ and is likely to play a role in stimulation of protective mechanisms to
indicates monoclonal antibody (mAb), while ‘-cept’ denotes fusion of repair damages when structural anomalies are present or to trigger
a receptor to the Fc portion of human IgG1, ‘-ximab’ chimeric mAb apoptotic processes to prevent the ongoing development of embryos
and ‘-zumab’ humanised mAb1 (Table 1). with structural anomalies.9,10 A marked increase of inflammatory
A recent survey in Europe and the USA revealed that clinicians cytokines occurs during infection or inflammation and together
concerned that anti-tumour necrosis factor (TNF) causes poor preg- with other chemokines may play a role in preterm delivery.11,12
nancy outcomes have discouraged women from breastfeeding while Women with recurrent pregnancy loss had significantly higher
on an anti-TNF.2 With improved disease and pregnancy outcomes Th1/Th2 ratios of interferon-c/interleukin (IL)-4, TNF-a/IL-4 and
associated with ongoing biologic (especially anti-TNF) use during TNF-a/IL-10 in CD3þ/CD8 T helper cells than controls.
pregnancy, clinicians will therefore need to address these issues
with women of childbearing potential; discontinuation of biologics
1
during pregnancy is not advised without due consideration of the Department of Rheumatology, Tauranga Hospital, Bay of Plenty District
potential risks and benefits.3,4 Health Board, Tauranga, New Zealand
2
Department of Obstetrics and Gynaecology, Elizabeth Rothwell Building,
Waikato Hospital, Waikato District Health Board, Hamilton,
Pathophysiology of cytokines in fertility, New Zealand
3
pregnancy and delivery Women’s Health Academic Centre, King’s College London, London, UK
4
Department of Gynaecology, Hospital S~ao Lucas da Pontifıcia
In brief, pregnancy is an immune-tolerant state of the feto-maternal Universidade Cat olica do Rio Grande do Sul, Porto Alegre, Brazil
relationship in which T-helper (Th) cytokines play an important role. 5
Clınica Generar-Human Reproduction, Porto Alegre, Brazil
Normally, Th2 (humoral immunity) predominates over Th1 (cell-
mediated immunity) in early gestation. This balance will shift to a Corresponding author:
Th1-predominant state later in pregnancy.5,6 May Ching Soh, Department of Rheumatology, Tauranga Hospital, Bay of
Inflammation can shift this balance towards a Th1 dominance, Plenty District Health Board, 829 Cameron Road, Tauranga 3112,
resulting in a stronger cell-mediated response and cytokine produc- New Zealand.
tion. The Th1-predominant environment may lead to implantation Email: mayching.soh@doctors.org.uk
6 Obstetric Medicine 13(1)

Table 1. Biologic agents classified according to mode of action, used in the treatment of rheumatic diseases.
Mechanism of action Drug Structure

TNF inhibition Infliximab Chimeric mAb to TNFa


Etanercept Fusion protein with a modified Fc portion that binds to TNFa
Adalimumab Fully human monoclonal antibody to TNFa
Certolizumab mAb to TNFa antibody with a pegylated Fab portion without an Fc portion
Golimumab Fully human mAb to TNFa
IL-1 inhibition Anakinra Recombinant and modified human IL-1 receptor antagonist
Canakinumab Human mAb that binds specifically to IL-1ß
Rilonacept Fusion Fc receptor that binds IL-1ß
IL-6 inhibition Tocilizumab Humanised mAb against IL-6 receptors
Sarilumab Recombinant mAb to IL-6 receptors
IL-17 inhibition Secukinumab Human mAb neutralising the effects of IL-17A
Ixekizumab Humanised mAb against IL-17A
IL-12/23 inhibition Ustekinumab Fully human mAb binding specifically to IL-12/IL23 inhibiting its activity
Co-stimulatory blockade Abatacept Fusion Fc region to extra-cellular domain of CTLA-4 modifying T-cell response
B-cell depletion/inhibition Rituximab Chimeric mAb with murine anti-CD variable-sequence regions
Belimumab Fully humanised mAb directed against soluble B-lymphocyte stimulator

CTLA: cytotoxic T-lymphocyte-associated protein; CD: complementarity-determining; IL: interleukin; mAb: monoclonal antibody; TNF: tumour necrosis factor.

Moreover, those who experienced recurrent implantation failure after diagnosis of VACTERL-association.23 Multiple studies subsequently
IVF had a higher proportion of TNF-a producing CD3þ/CD8 cells failed to find any association. Therefore, many national and interna-
and higher Th1/Th2 ratios of TNF-a/IL-4 and TNF-a/IL-10 in tional guidelines support the use of anti-TNF agents in the first
CD3þ/CD8 cells when compared with controls.13 In sub-fertile trimester.24,25
women, the preconception ratio of TNF-a: IL-10 elevation has
been associated with high oocyte die-off ratio leading to lower success
rates during IVF.14,15 Adalimumab and immunoglobulins adminis- Biologics in the second and
tration have been associated with a reduction in the oocyte die-off
ratio and a greater chance of successful IVF.14–16
third trimesters
Overall, these observations suggest that targeting TNF-a may be From the second trimester onwards, transplacental passage of bio-
useful in treating women with recurrent pregnancy loss, recurrent logics is facilitated by Fc receptors on the fetal trophoblasts in
implantation failure and early, severe pre-eclampsia due to excessive tandem with increasing transplacental transfer of maternal IgG as
TNF-a production.17 pregnancy advances.26,27 Fetal IgG1 levels reach 50% of maternal
During delivery, IL-1, IL-6, IL-8 and TNF levels peak. levels at 28–32 weeks’ gestation and increase exponentially afterwards
Hypothetically, preterm labour is driven by overwhelming inflamma- with maximal transfer in the final 4 weeks prior to delivery.28 Hence,
tion.12 Using an experimental lipopolysaccharide-induced preterm at delivery, cord blood levels of neonates exposed to some of the
model, IL-6 inhibitors induced prolongation of the gestational biologics in late gestation are often much higher than maternal
period in mice (without adverse effects on pups) by significantly serum drug levels (Table 2). Cord blood levels probably depend on
inhibiting IL-6-induced prostaglandin E2 production.18 Therefore, it
several factors: (i) The gestation in which the drug was used – the
is possible that in the future preterm, labour might be manageable by
later in pregnancy, the higher the cord drug levels; (ii) structure of the
a combination of biologic agents that specifically targeted these path-
Fc receptors – biologics with modified Fc receptors (i.e. the ‘-cepts’)
ways of Th1 activation.
or those lacking an Fc portion altogether (e.g. certolizumab) are less
Clinical studies in men have reassuringly demonstrated that anti-
TNF agents do not impair sperm quality, and their use has not been efficiently transported across the placenta and can often be used
associated with teratogenicity in offspring.19,20 throughout pregnancy or at least until 34 weeks’ gestation (e.g. eta-
nercept); (iii) half-life of the drug – the longer its half-life, the more
likely its passage across the placenta. There are limited published
Biologics in early pregnancy studies on maternal to neonatal cord blood drug levels, with wide
variations in the very small cohorts sampled (Supplementary Table
Biologics are large molecular structures not easily transported across
1). In addition to the factors just listed, drug levels are also likely
the placenta, particularly during the first trimester when fetal troph-
influenced by the dosing interval (i.e. proximity of cord blood collec-
oblasts have yet to develop Fc receptors. Prior to 14 weeks’ gestation,
tion to administration of the last dose), dosages used (e.g. infliximab
these large molecules reach the fetus by passive diffusion; hence, only
can be dosed at 3–10 mg/kg at six to eight weekly intervals) and quite
limited amounts reach the fetus during the period of embryogenesis
(up to 12 weeks).21 possibly a multitude of other factors which remain poorly understood
An early and controversial study mining of the Food and Drug (e.g. drug antibodies). Apart from certolizumab – virtually none of
Administration database attempted to link anti-TNF use with which crosses the placenta – it remains unlikely that sufficient data
VACTERL (vertebral defects, anal atresia, cardiac defects, tracheo- will ever be available concerning the exact amount of drug trans-
esophageal fistula, renal anomalies, and limb abnormalities)- ferred to the neonate in utero. Available published data on cord
association congenital abnormalities.22 Individual anomalies that blood ratios have been summarised in Table 2. Furthermore, results
form the VACTERL are not uncommon – hence, the need for from animal studies should not be extrapolated since Fc receptors
three or more features is needed for the diagnosis. The cases reported and binding to neonatal trophoblasts may differ in animals.
in the study did not necessarily adhere to these criteria for the Additionally, doses tested in laboratory animals often exceed the
Soh and Moretto 7

Table 2. The half-life of biologics and transplacental There were no reports of serious infections or developmental
transfer ratio. delay in a median follow-up of 18 months in neonates exposed to
either canakinumab or anakinra.48 However, data on vaccinations
Transplacental transfer were lacking, especially for neonates exposed in the third trimester.49
ratio (cord blood: Canakinumab has a transplacental transfer ratio of 2.11 (Table 2)
Drug t1=2 (days) References maternal drug level)a based on a single case where the cord canakinumab level was quan-
30,31,32,33,34 tified in an infant exposed until 34 weeks’ gestation and delivered at
Infliximab 8–10 2.23
35,36 39 weeks’ gestation.42 Hitherto, despite multiple clinical cases pub-
Etanercept 4–5 0.06
37,38,33,39,34 lished in the literature, anakinra has never been quantified in cord
Adalimumab 10–20 1.11
blood, and its transplacental transfer ratio is unknown.49–52 Since
Golimumab 12–15 NA –
40,33,41 anakinra has a half-life of 4–6 h compared to 22 days in canakinu-
Certolizumab 14 0.04
42 mab, it would seem reasonable to switch to anakinra in pregnancy if
Canakinumab 22 2.11
clinically indicated. Nevertheless, this short half-life entails daily
Tocilizumab 8–14 NA –
Ustekinumab 15–32 43
1.86 administration – rather than the four- to eight-weekly dosing interval
Abatacept 8–25 NA – of canakinumab. Hence, quantification of anakinra cord, maternal
Rituximab 18–22 44,45,46
1.69 and infant serum drug levels would be very helpful in informing
Belimumab 19–20 NA clinicians and prospective mothers on the preferred IL-1 inhibitor
to use in pregnancy. Although, cord serum quantification of anakinra
Note: NA: not available in human subjects. (as a slight modification of the natural IL-1 receptor antagonist) may
a
Mean value from cumulative studies. require a special test that prevents measuring both the native-
occurring IL-1 and anakinra together.
Cases of rilonacept use during pregnancy have yet to
recommended therapeutic doses used in human subjects26,29 (full data be published.
in Supplementary Table 1).
Hence, most studies on biologics use in pregnancy have focused
on clinical outcomes in exposed offspring.
IL-6 inhibitors: Tocilizumab and sarilumab
Since EULAR and BSR guidelines, there have been four additional
publications including registry data from the Tocilizumab manufac-
Latest updates on biologic agents turer’s global safety database.53,54,55,56 There have been 358 observed
While data remain limited, there are no signals of increased rates of pregnancies; the miscarriage rate was 22.9%, though concurrent
malformations or adverse pregnancy outcomes from the various reg- methotrexate (and occasionally leflunomide) use was prevalent in
istries set up specifically to look at the use of biologics in pregnancy. these cohorts.55,56 The livebirth rate was only 60%, due to a high
This review paper will endeavour to summarise the European rate of miscarriage and elective termination of pregnancy (18.4%) in
League Against Rheumatism (EULAR) and British Society in the cohorts studied. Six malformations were noted in the literature,
but without any distinguishing pattern of malformation to suggest
Rheumatology (BSR) guidelines 2016 shown in Table 324,25,47 and
teratogenicity.55,56 Tocilizumab use (>90% in four-weekly 8mg/kg
focus on developments since publication of the guidelines.
infusions) occurred predominantly during pre-conception and the
Additional information on breastmilk excretion is available in
first trimester, the drug being discontinued upon confirmation of
Supplementary Table 1.
pregnancy for the majority of women; tocilizumab was used
beyond the first trimester in only 15 pregnancies.55,56 Of the 15 neo-
IL-1 inhibitors: Canakinumab and anakinra nates exposed to tocilizumab later in pregnancy, none had their cord
tocilizumab level quantified.
An international retrospective study of IL-1 inhibitors anakinra and
A case series of two women showed that breastmilk concentra-
canakinumab was collated by members of the International Society
tions of tocilizumab peaked at day three post-administration but
for Systemic Autoinflammatory Diseases.48 Among the eight women
remained 1/500 to 1/1000 compared to maternal serum concentra-
exposed to canakinumab during pregnancy (plus five paternal expo-
tions. Both breastfed infants remained well; they received all routine
sures in the preconception period), no congenital abnormalities were
vaccinations, including Bacillus Calmette–Guérin (BCG) and rotavi-
observed. A single early miscarriage occurred at six weeks’ gestation
rus without problems.57
in a woman with refractory Cogan’s syndrome. The seven infants Studies on sarilumab use in pregnancy have yet to be published.
exposed to canakinumab were delivered at term with normal birth The manufacturer has set up an observational study (commencing 1
weights. Maternal disease remained in remission during pregnancy, February 2018) to recruit at least 300 women (and infants) for
despite only four women continuing on canakinumab beyond the the duration of pregnancy and one year follow-up post-partum
period of organogenesis (12 weeks’ gestation); others either stopped (see: https://clinicaltrials.gov/ct2/show/NCT03378219).
treatment or switched to anakinra (n ¼ 2). Four infants were
breastfed with no adverse effects. Follow-up time of infants varied
from seven days to four years. IL-17A inhibitors: Secukinumab and ixekizumab
In the same study, 23 women (and 6 men) were exposed to ana- An abstract on secukinumab in 66 pregnancies from the manufac-
kinra. A single case of renal agenesis and ectopic neurohypophysis turer’s global safety database was recently presented.58 There were
with growth hormone deficiency was noted in an infant whose mother 8 (12.1%) miscarriages, only 15 reported livebirths (without any con-
was treated from 9 weeks’ gestation till delivery; miscarriage at genital malformation) and 11 ongoing pregnancies. Close to half
12 weeks was recorded in a woman with active Cogan’s syndrome.48 (48%) had either an elective termination of pregnancy (without any
An abstract summarising the experience of five women who received listed cause) or information on the pregnancy was missing. No infor-
anakinra 100 mg into the third trimester found no malformations or mation was provided on 18 pregnancies with paternal exposure.
neonatal problems – including the three infants breastfed while their More recently, the experience of six women with psoriatic arthritis
mothers continued treatment with anakinra.49 on secukinumab during pregnancy was published as an abstract.59
8 Obstetric Medicine 13(1)

Table 3. Summary from EULAR points to consider and BSR guidelines for use during pregnancy.
Compatible with
Compatible for use in early pregnancy What gestation to discontinue breastfeeding

Agent EULAR BSR EULAR BSR EULAR BSR

Infliximab   Up to 20 weeks a
Stop at 16 weeks  
Etanercept   Up to 32 weeksa Not compatible with  
third trimester
Adalimumab   Up to 20 weeksa Not compatible with  
third trimester
Certolizumab   Use throughout Compatible with  No data
pregnancy third trimester
Golimumab No increased  Use another agent § No data  No data
malformations §
Anakinra  ‡ Only if no other   No data No data
options
Tocilizumab § Stop three Avoid §   No data No data
months prior ‡
Ustekinumab No increased – Use another agent § –  No data –
malformations §
Abatacept §  §  No data  No data No data
Rituximab  Can be used Stop six Not specified. †   No data No data
in exceptional months prior ‡
circumstances.
Belimumab No increased ‡ §   No data No data
malformations §

Note: : yes; : avoid/not recommended; No data: no published data available; §: insufficient evidence; †: neonatal B cell depletion if used in late pregnancy; ‡: insufficient
data to recommend drug, but unintentional exposure in the first trimester is unlikely to be harmful; NA: not applicable; – not covered by publication. EULAR: European
League Against Rheumatism; BSR: British Society in Rheumatology.
a
Can be used throughout pregnancy if indicated.

Secukinumab was discontinued upon confirmation of pregnancy and remained well, and one even had all his ‘routine’ vaccinations without
no malformation, adverse maternal or neonatal outcomes occurred. problems.69 Overall, the livebirth rate across all published cases was
Maternal psoriatic arthritis remained in remission throughout preg- 57.1%, and miscarriage was low at 15.9%. Miscarriage occurred at
nancy even off secukinumab.59 12 weeks in one mother who was a heavy smoker.64 Only one case
Similarly, data on ixekizumab from the manufacturer’s Safety had concurrent DMARD documented (i.e. azathioprine 100mg
Systems (from seven clinical trials) were limited.60 Of 18 pregnancies, daily). Since these were largely studies for Crohn’s and psoriasis,
8 (44.4%) resulted in livebirths and 5 (27.8%) in miscarriages or some women may have been co-prescribed methotrexate. No reasons
elective terminations. One third of cases (n ¼ 6) had no recorded out- were given for elective terminations, and no congenital malforma-
come; it was unclear if this represented missing information or any tions were reported. In the three cases involving third trimester expo-
other adverse pregnancy outcome (e.g. stillbirth, late pregnancy loss, sure, neither cord drug levels nor drug excretion into breastmilk were
etc.). No congenital malformations were reported. Of the 34 cases quantified. In one case, despite ustekinumab use until 33 weeks’ ges-
with paternal exposure to ixekizumab, the vast majority (82.4%) tation, the mother was advised not to breastfeed the infant once she
resulted in livebirths, without any cases of congenital malformation. resumed ustekinumab post-partum.68 None of the published cases
For all studies, information on concurrent disease-modifying anti- included paternal exposure to ustekinumab.
rheumatic drugs (DMARD) use, duration of exposure to the IL-17A Human data from published studies do not appear to show an
inhibitor, breastfeeding or reasons for termination of pregnancy were increased risk of congenital malformations or miscarriages in the
not available from the abstracts. exposed group. However, ustekinumab use in later pregnancy, trans-
placental transfer ratios, neonatal infections and responses to vacci-
nations were not recorded in the reports.
IL-12/23 inhibitor: Ustekinumab
Only 10 published papers and abstracts exist on the use of ustekinu-
mab during pregnancy. Two of the larger studies were derived from
B-cell inhibitors: Belimumab
Phase II studies on Crohn’s and psoriasis where ustekinumab was Most of the data on belimumab in pregnancy come from the phar-
discontinued upon confirmation of pregnancy.61,62 To date, 63 preg- maceutical company’s registry (see: http://www.bprgsk.com, current-
nancies with exposure to ustekinumab and known outcomes have ly collecting information till 2021).71 Data on 66 pregnancies were
been described.61–70 Ustekinumab exposure occurred within three presented as an abstract in EULAR in 2013 related to an open label
months pre-conceptually or in the first trimester in the majority of Phase IV (long-term extension) clinical trial and showed a lower rate
cases (59/63). Three women who were prescribed ustekinumab in the of fetal loss in women treated with belimumab compared to placebo
third trimester also had good neonatal outcomes.63,68,69 While the and a livebirth rate of only 50%. Exact information on DMARDs
dosage and dosing intervals for ustekinumab were different in the was not available; there were two cases of malformations – one
two of the women in the third trimester, their exposed offspring Dandy–Walker syndrome, and one bilaterally enlarged kidneys.72
Soh and Moretto 9

Presumably belimumab was discontinued upon confirmation of preg- The accumulation of biologics in offspring exposed in utero can
nancy; a large number of women also elected to have their pregnancy persist for up to a year. Much depends on the timing of drug admin-
terminated.72 istration, the degree of transplacental transfer and the drug’s half-life.
Two additional published case reports that described the use of For instance, infliximab transfers efficiently starting from the second
belimumab beyond the first trimester of pregnancy did not demon- trimester (transplacental transfer 2.23) and remains in the infant for a
strate any adverse outcomes in either mother or neonate.73,71 There is median period of 7.3 months (although persisting up until 12 months
a single published case report of maternal use of belimumab till 26 of age), while adalimumab has a transplacental transfer of 1.11 and
weeks’ gestation for severe systemic lupus erythematosus (SLE). At remains in the infant for a median of 4 months (Table 3).34 It is
40 weeks, she delivered a healthy male infant who subsequently postulated that ineffective clearance by the neonate’s immature retic-
received all routine immunisations, including the rotavirus live vac- uloendothelial system could contribute to the persistence of detect-
cine at 6 weeks, without problems. He was also noted to mount a able drug levels in the infant’s serum.33 Recent research has
good response to the pneumococcal vaccine at seven months of age.74 demonstrated that TNF remains in complex with adalimumab even
six months after discontinuation of the drug in adults.81 Whether
persistence of the drug has any adverse effect on the neonate has
Abatacept not been adequately studied.
To date, 162 pregnancies with abatacept exposure in early pregnancy Primate studies have shown that TNFs (particularly superfamily
(152 maternal and 10 paternal) have been described.75,76 The manu- members lymphotoxin (LT)a and LTb) are dispensable for the devel-
facturer’s registry revealed 151 maternal cases resulting in 86 (59.5%) opment of the neonatal immune system.11 Golimumab in primate
live births, 49 miscarriages (including one late loss at 21 weeks’ ges- studies has not demonstrated a reduction in lymphoid tissue in
tation) and 19 elective terminations.76 Methotrexate was co- macaque offspring exposed in utero.11 This laboratory observation
administered in 13.2%. Seven cases (8.1%) of congenital malforma- seems to be borne out by clinical findings – infants exposed in utero
tions were recorded, including a single case of Down’s syndrome to anti-TNF agents are not at greater risk of infections, particularly if
(terminated at 17 weeks’ gestation due to spontaneous rupture of anti-TNF agents are used as monotherapy.34,82
membranes). The cases of malformation were isolated and did not Nevertheless, there have been rare reports of neonatal neutrope-
reflect a specific pattern of abnormalities.76 In all published cases of nia attributed to anti-TNF exposure in utero (women were treated
maternal exposure, abatacept was discontinued upon confirmation of with infliximab throughout pregnancy for inflammatory bowel dis-
pregnancy in the first trimester.75,76 Paternal exposure to abatacept ease (IBD)) – a known adverse effect of the drug.83,84 However, when
was not associated with any congenital anomalies in 9/10 ongoing combined with other immunomodulators, in particular thiopurines,
pregnancies.76 Information concerning the abatacept transplacental one international cohort study involving 80 women with IBD
transfer ratio, use in late pregnancy, childhood infections and vacci- found that infants exposed to both thiopurines and anti-TNF
nations has yet to be published, though follow-up in 16 of the off- agents demonstrated a three-fold increased risk of childhood infec-
spring exposed in early pregnancy has not demonstrated any immune tions, compared to those whose mothers were on monotherapy with
deficiencies.76 an anti-TNF agent (relative risk 2.7; 95% confidence interval 1.09–
6.78; p ¼ 0.02).34 The Groupe d’Étude Thérapeutique des Affections
du Tube Digestif cohort (n ¼ 232) found that the risk of infection and
Childhood infections and vaccinations pregnancy complications was similar in two groups of pregnant
women with IBD which, coincidentally, had similar rates of thiopur-
If a biologic transfers efficiently across the placenta, and maternal use ine use, although one group was on anti-TNF therapy and the other
has continued beyond 28 weeks, the exposed neonate is assumed to served as non-TNF control (n ¼ 99).85
have cord drug levels exceeding those of maternal serum drug levels. Rituximab causes B-cell depletion, has a high transplacental
The drug’s effects on the neonate will resemble those in the mother, transfer rate of 1.7 (Table 3) and is known to cause B-cell depletion
i.e. potent immunosuppression. Hence, similar precautions are and transient cytopenias in exposed infants.44 In the global safety
required as those taken in the case of the mother, e.g. prompt assess- database for rituximab, 21 women receiving rituximab (for a variety
ment and treatment of infections, avoidance of live vaccines (at least of causes, including chemotherapy) had infants (n ¼ 11) with
for the first six to seven months of the neonate’s life). These early recorded haematologic abnormalities that included B-cell depletion,
vaccines to be avoided include rotavirus and BCG in certain coun- lymphopenia, neutropenia, anaemias and thrombocytopenias.86
tries. All other non-live vaccines are safe to be administered. Most spontaneously recovered in weeks to months post-partum,
There is emerging evidence that rotavirus vaccine is safe even in apart from one fatal case of cerebral haemorrhage in an exposed
infants exposed to maternal anti-TNF use in the third trimester.77 infant born with neonatal thrombocytopenia at 39 weeks’ gestation
Nevertheless, many will recall the solitary published case of the following the administration of rituximab to her mother at her sev-
demise of a 4.5 month infant from disseminated non-caseating gran- enth month of gestation for idiopathic thrombocytopaenic purpura.
ulomatous disease (likely mycobacteria, though Ziehl–Neelsen stain- Suppressed B-cell development and depletion have not given rise to
ing for acid-fast bacilli and tuberculosis polymerase chain reaction an increase in reported infectious complications amongst the affected
methods were deemed equivocal) following BCG vaccination at 3 neonates, and most of those studied continue to exhibit a robust
months of age; the mother had received infliximab therapy through- immune response to vaccinations given.44–46,86
out pregnancy for her underlying Crohn’s, with her last infusion just Women on biologics planning pregnancy should be made aware
two weeks prior to delivery.78 Since TNF is integral to formation of that up until the present there have been no long-term outcome stud-
the granuloma, the question then arises as to whether BCG vaccina- ies of children following in utero exposure. A theoretical risk has
tion would be safe in neonates exposed in the third trimester to other always existed of exposed offspring developing autoimmune diseases
biologics (non-TNF-inhibitors).79,80 While there has been no signifi- (e.g. anti-TNF-induced lupus), or being prone to allergies, malignan-
cant increase in neonatal infectious complications, carers should be cies, etc. However, fear of the unknown in the long term needs to be
advised to remain vigilant for possible infections. With dampened carefully balanced against the shorter term gain of maintaining
cytokine release in the presence of biologics, infants may not neces- maternal health and well-being, thereby ensuring an optimal preg-
sarily exhibit the same signs of infection that clinicians are accus- nancy outcome. Mothers with active inflammatory/autoimmune dis-
tomed to seeing. eases are at greater risk of disease flares leading to placental
10 Obstetric Medicine 13(1)

Table 4. Practical points for consideration when facing a woman on a new biologic contemplating pregnancy.
1. The structure of the biologic and the likelihood of its passage across the placenta
2. When (or if) the biologic should be discontinued during pregnancy
3. Clinically significant and longer term effects on the neonate exposed in utero, the risks being balanced against the immediate short-term risk of a
maternal disease flare or active disease in pregnancy resulting in an adverse outcome, preterm delivery or longer term maternal morbidity

insufficiency clinically presenting as pre-eclampsia and fetal growth Biosimilars are flooding the market. Studies looking at their
restriction – factors that in turn lead to preterm deliveries (and all the effects are therefore necessary given that it remains unclear how
associated morbidity accompanying a preterm growth-restricted (dis)similar these molecular structures are and what potential
infant).87,88 Moreover, the mothers themselves are also at risk since impact they might have on pregnancy.
long-term population-based studies have demonstrated that women The ongoing accumulation and sharing of information on biolog-
with autoimmune diseases (in particular SLE) who have pregnancies ic use in pregnancy are of paramount importance. It is vital to publish
complicated by placental insufficiency or maternal-placental syn- case reports of early biologic use in pregnancy to facilitate informa-
drome and preterm delivery prior to 34 weeks’ gestation are at a tion sharing.
two-fold increased risk of accelerated development of cardiovascular In addition, the reporting of pregnancy complications and their
events and premature cardiovascular death.89–91 outcomes should be standardised. Little information can be gleaned
from published statements such as ‘delivery of a normal infant at
term’. Hence, the collaborations of rheumatologists, gastroenterolo-
Breastfeeding gists, oncologists, neurologists, obstetricians, neonatologists and pae-
It is unlikely that comprehensive studies will ever be carried out on all diatricians are crucial for more comprehensive data collection that
biologics and the peak time for breastmilk excretion from time of would benefit multiple subspecialties. Long-term studies to determine
drug administration (Supplementary Table 1). However, basic phys- the safety of these drugs in exposed offspring are urgently needed.
iology indicates that breastmilk is predominantly IgA (not IgG1); the Would better disease control in pregnancy mean better long-term
large structure of most biologic molecules means that they are less outcomes for both mother and child? Collaborative efforts and the
likely to be excreted in breastmilk. Additionally, such large protein- establishment of long-term registries for both the disease and the
aceous molecules are denatured and digested by passage through the drugs they are exposed to are essential for a holistic view of how
infant’s gastrointestinal tract and therefore do not attain significant these biologics influence pregnancy and perhaps even the
levels in breastfed infants,47 and hence why parenteral administration
next generation.
is necessary to achieve a therapeutic response. All breastmilk studies
completed to date have borne this out by consistently showing a
steady decline in breastfed infant drug levels despite ongoing mater- Acknowledgements
nal dosing. These data seem fairly reassuring should a woman desire The authors thanks to Mrs. Brina Roshd who proof-read and edited
to breastfeed even while taking a biologic. this manuscript comprehensively.

Future directions Declaration of conflicting interests


To date, published data seem largely reassuring: rates of congenital The author(s) declared no potential conflicts of interest with respect
anomalies in offspring exposed in utero are similar to those in the to the research, authorship, and/or publication of this article.
general population, and they have achieved normal milestones in the
first few years of life without an increased prevalence of allergies or Funding
life-threatening infections.
Although data concerning many newer biologics remain limited, The author(s) received no financial support for the research, author-
the knowledge base is expanding. Pharmaceutical companies are ship, and/or publication of this article.
encouraged to include pregnant women in drug trials, and pregnancy
registries have been established for various drugs.92 Ethical approval
It is highly desirable that pharmaceutical companies will in future
Not applicable.
invest in developing of biologics suitable for use in pregnancy.
Perhaps biologics could be created from IgG2 molecules instead –
the latter would give rise to minimal transfer compared to other IgG Informed consent
molecules. Alternatively, an expansion of the current repertoire of Not applicable.
larger molecule biologics (e.g. certolizumab) whose size prohibits effi-
cient transplacental transfer or modification of Fc receptors that are
not conducive to transplacental transport would also be very wel- Guarantor
come.28,93 Companies could also offer complimentary maternal MCS.
drug serum testing and cord blood testing for all infants exposed
and such data should be shared in their registries and freely accessible
Contributorship
to all clinicians caring for (and counselling) women who are pregnant
or planning pregnancy. Additional studies looking at the combined MM contributed to the sections on reproduction and fertility; MCS is
effects of biologic agents and other immunomodulators such ashy- responsible for the concept and initial drafts of the manuscript.
droxychloroquine or sulfasalazine on the risk of infection in neonates Both authors have contributed to the drafting and approval of
are required to further delineate the contributory immunosuppressive the final version of the manuscript and have agreed to be accountable
effects of these drugs on the risk of infection in both mother for all aspects pertaining to the accuracy and integrity of the
and neonate. entire manuscript.
Soh and Moretto 11

ORCID iD 19. Micu MC, Micu R, Surd S, et al. TNF-alpha inhibitors do not
May Ching Soh http://orcid.org/0000-0002-7160-7550 impair sperm quality in males with ankylosing spondylitis after
short-term or long-term treatment. Rheumatology (Oxford)
2014; 53: 1250–1255.
References 20. Larsen MD, Friedman S, Magnussen B, et al. Birth outcomes in
1. Lamberg L. A host of novel agents for treating psoriasis, psori- children fathered by men treated with anti-TNF-alpha agents
atic arthritis stir interest. JAMA 2003; 289: 2779–2783. before conception. Am J Gastroenterol 2016; 111: 1608–1613.
2. Tincani A, Taylor P, Fischer-Betz R, et al. FRI0692 Anti-TNF 21. Hyrich KL and Verstappen SM. Biologic therapies and pregnan-
treatments for women with chronic inflammatory diseases: com- cy: the story so far. Rheumatology (Oxford) 2014; 53: 1377–1385.
paring attitudes and perceptions of physicians in Europe and the 22. Carter JD, Ladhani A, Ricca LR, et al. A safety assessment of
US. Ann Rheum Dis 2018; 77: 865–865. tumor necrosis factor antagonists during pregnancy: a review of
3. Ursin K, Lydersen S, Skomsvoll J, et al. THU0673 disease activ- the Food and Drug Administration database. J Rheumatol 2009;
ity during and after pregnancy in women with psoriatic arthritis. 36: 635–641.
Ann Rheum Dis 2018; 77: 530–530. 23. Østensen M. Are TNF inhibitors safe in pregnancy? Nat Rev
4. F€orger F, Bandoli G, Luo Y, et al. FRI0119 Effect of discontin- Rheumatol 2009; 5: 184–185.
uing TNF inhibitors during pregnancy on the course of rheuma- 24. Gotestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The
toid arthritis and juvenile idiopathic arthritis. Ann Rheum Dis EULAR points to consider for use of antirheumatic drugs
2018; 77: 604–604. before pregnancy, and during pregnancy and lactation. Ann
5. Chaouat G. The Th1/Th2 paradigm: still important in pregnan- Rheum Dis 2016; 75: 795–810.
cy? Semin Immunopathol 2007; 29: 95–113. 25. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on
6. Ghaebi M, Nouri M, Ghasemzadeh A, et al. Immune regulatory prescribing drugs in pregnancy and breastfeeding-Part I: stan-
network in successful pregnancy and reproductive failures. dard and biologic disease modifying anti-rheumatic drugs and
Biomed Pharmacother 2017; 88: 61–73. corticosteroids. Rheumatology (Oxford) 2016; 55: 1693–1697.
7. Challis JR, Lockwood CJ, Myatt L, et al. Inflammation and 26. Kuo TT, Baker K, Yoshida M, et al. Neonatal Fc receptor: from
pregnancy. Reprod Sci 2009; 16: 206–215. immunity to therapeutics. J Clin Immunol 2010; 30: 777–789.
8. Kalu E, Bhaskaran S, Thum MY, et al. Serial estimation of Th1: 27. Fouda GG, Martinez DR, Swamy GK, et al. The impact of IgG
th2 cytokines profile in women undergoing in-vitro fertilization- transplacental transfer on early life immunity. Immunohorizons
embryo transfer. Am J Reprod Immunol 2008; 59: 206–211.
2018; 2: 14–25.
9. Toder V, Fein A, Carp H, et al. TNF-alpha in pregnancy loss
28. Saji F, Samejima Y, Kamiura S, et al. Dynamics of immunoglo-
and embryo maldevelopment: a mediator of detrimental stimuli
bulins at the feto-maternal interface. Rev Reprod 1999; 4: 81–89.
or a protector of the fetoplacental unit? J Assist Reprod Genet
29. Chambers CD, Tutuncu ZN, Johnson D, et al. Human pregnan-
2003; 20: 73–81.
cy safety for agents used to treat rheumatoid arthritis: adequacy
10. Robertson SA, Chin P-Y, Femia JG, et al. Embryotoxic cyto-
of available information and strategies for developing post-
kines – potential roles in embryo loss and fetal programming.
marketing data. Arthritis Res Ther 2006; 8: 215.
J Reprod Immunol 2018; 125: 80–88.
30. Vasiliauskas EA, Church JA, Silverman N, et al. Case report:
11. Arsenescu R, Arsenescu V and de Villiers WJ. TNF-alpha and
evidence for transplacental transfer of maternally administered
the development of the neonatal immune system: implications for
infliximab to the newborn. Clin Gastroenterol Hepatol 2006;
inhibitor use in pregnancy. Am J Gastroenterol 2011;
4: 1255–1258.
106: 559–562.
31. Mahadevan U, Terdiman JP, Church J. Infliximab levels in
12. Di Renzo GC, Tosto V and Giardina I. The biological basis and
infants born to women with inflammatory bowel disease.
prevention of preterm birth. Best Pract Res Clin Obstet Gynaecol
2018; 52: 13–22. Gastroenterology 2007; 132: A144.
13. Kwak-Kim J. Increased T helper 1 cytokine responses by circu- 32. Zelinkova Z, de Haar C, de Ridder L, et al. High intra-uterine
lating T cells are present in women with recurrent pregnancy exposure to infliximab following maternal anti-TNF treatment
losses and in infertile women with multiple implantation failures during pregnancy. Aliment Pharmacol Ther 2011; 33: 1053–1058.
after IVF. Hum Reprod 2003; 18: 767–773. 33. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer
14. Winger EE, Reed JL, Ashoush S, et al. Degree of TNF-alpha/IL- of anti-tumor necrosis factor agents in pregnant patients with
10 cytokine elevation correlates with IVF success rates in women inflammatory bowel disease. Clin Gastroenterol Hepatol 2013;
undergoing treatment with Adalimumab (Humira) and IVIG. 11: 286—292.
Am J Reprod Immunol 2011; 65: 610–618. 34. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations
15. Winger EE, Reed JL, Ashoush S, et al. Die-off ratio correlates of Adalimumab and Infliximab in Mothers and Newborns, and
with increased TNF-alpha:IL-10 ratio and decreased IVF success Effects on Infection. Gastroenterology 2016; 151: 110–119.
rates correctable with humira. Am J Reprod Immunol 2012; 35. Murashima A, Watanabe N, Ozawa N, et al. Etanercept during
68: 428–437. pregnancy and lactation in a patient with rheumatoid arthritis:
16. Winger EE, Reed JL, Ashoush S, et al. Treatment with adalimu- drug levels in maternal serum, cord blood, breast milk and the
mab (Humira) and intravenous immunoglobulin improves preg- infant’s serum. Ann Rheum Dis 2009; 68: 1793–1794.
nancy rates in women undergoing IVF. Am J Reprod Immunol 36. Berthelsen BG, Fjeldsoe-Nielsen H, Nielsen CT, et al. Etanercept
2009; 61: 113–120. concentrations in maternal serum, umbilical cord serum, breast
17. Alijotas-Reig J, Esteve-Valverde E, Ferrer-Oliveras R, et al. milk and child serum during breastfeeding. Rheumatology
Tumor necrosis factor-alpha and pregnancy: focus on biologics. (Oxford) 2010; 49: 2225–2227.
An updated and comprehensive review. Clinic Rev Allerg 37. Mahadevan U, Miller JK, Wolf DC. Adalimumab Levels
Immunol 2017; 53: 40–53. Detected in Cord Blood and Infants Exposed in Utero.
18. Wakabayashi A, Sawada K, Nakayama M, et al. Targeting Gastroenterology 2011; 140: S-61–S-62.
interleukin-6 receptor inhibits preterm delivery induced by 38. Julsgaard M, Brown S, Gibson P, et al. Adalimumab levels in an
inflammation. Mol Hum Reprod 2013; 19: 718–726. infant. Journal of Crohn’s & colitis 2013; 7: 597–598.
12 Obstetric Medicine 13(1)

39. Zelinkova Z, van der Ent C, Bruin KF, et al. Effects of discon- 257). Australasian College of Dermatologists 2017 New South
tinuing anti-tumor necrosis factor therapy during pregnancy on Wales, Australia, 2017.
the course of inflammatory bowel disease and neonatal exposure. 59. Meroni M, Generali E, Guidelli GM, et al. THU0319 Overall
Clin Gastroenterol Hepatol 2013; 11: 318–321. safety of 7-week secukinumab exposure during pregnancy in
40. Mahadevan U, Abreu MT. 960 Certolizumab Use in Pregnancy: women with psoriatic arthritis. Ann Rheum Dis 2018;
Low Levels Detected in Cord Blood. Gastroenterology 2009; 136: 77: 377–378.
A–146. 60. Feldman S, Pangallo B, Xu W, et al. Ixekizumab and pregnancy
41. Mariette X, Forger F, Abraham B, et al. Lack of placental trans- outcome. Journal of the American Academy of Dermatology 2017;
fer of certolizumab pegol during pregnancy: results from CRIB, a 76: AB419.
prospective, postmarketing, pharmacokinetic study. Ann Rheum 61. Schaufelberger BW, Horn E, Cather JC, et al. Pregnancy out-
Dis 2018; 77: 228–233. comes in women exposed to ustekinumab in the psoriasis clinical
42. Egawa M, Imai K, Mori M, et al. Placental Transfer of development program. Journal of the American Academy of
Canakinumab in a Patient with Muckle-Wells Syndrome. J Dermatology 2014; 70: AB178.
Clin Immunol 2017; 37: 339—341 62. Scherl E, Jacobstein D, Murphy C, et al. A109 pregnancy out-
43. Rowan CR, Cullen G, Mulcahy HE, et al. Ustekinumab Drug comes in women exposed to Ustekinumab in the Crohn’s disease
Levels in Maternal and Cord Blood in a Woman With Crohn’s clinical development program. Journal of the Canadian
Disease Treated Until 33 Weeks of Gestation. J Crohns Colitis Association of Gastroenterology 2018; 1: 166.
2018; 12: 376-3-78. 63. Andrulonis R, Ferris LK. Treatment of severe psoriasis with
44. Klink DT, van Elburg RM, Schreurs MW, et al. Rituximab ustekinumab during pregnancy. Journal of drugs in dermatology:
administration in third trimester of pregnancy suppresses neona- JDD 2012; 11: 1240.
tal B-cell development. Clin Dev Immunol 2008; 2008: 271363. 64. Fotiadou C, Lazaridou E, Sotiriou E, et al. Spontaneous abor-
45. Decker M, Rothermundt C, Hollander G, et al. Rituximab plus tion during ustekinumab therapy. J Dermatol Case Rep 2012;
CHOP for treatment of diffuse large B-cell lymphoma during 6: 105–107.
second trimester of pregnancy. The lancet oncology 2006; 65. Sheeran C, Nicolopoulos J. Pregnancy outcomes of two patients
7: 693–694. exposed to ustekinumab in the first trimester. Australas J
46. Friedrichs B, Tiemann M, Salwender H, et al. The effects of Dermatol 2014; 55: 235–236.
rituximab treatment during pregnancy on a neonate. 66. Alsenaid A, Prinz JC. Inadvertent pregnancy during ustekinu-
Haematologica 2006; 91: 1426–1427. mab therapy in a patient with plaque psoriasis and
47. Soh MC, MacKillop L. Biologics in pregnancy - for the obste- impetigo herpetiformis. J Eur Acad Dermatol Venereol 2016;
trician. The Obstetrician & Gynaecologist 2016; 18: 25–32. 30: 488–490.
48. Youngstein T, Hoffmann P, Gul A, et al. International multi- 67. Rocha K, Piccinin MC, Kalache LF, et al. Pregnancy during
centre study of pregnancy outcomes with interleukin-1 inhibitors. Ustekinumab Treatment for Severe Psoriasis. Dermatology
Rheumatology (Oxford) 2017; 56: 2102–2108. (Basel, Switzerland) 2015; 231(2): 103–104.
49. Smith CJF, Chambers C. FRI0107 Five successful pregnancies 68. Galli-Novak E, Mook SC, Büning J, et al. Successful pregnancy
with antenatal anakinra exposure. Ann Rheum Dis 2018; 77: 598. outcome under prolonged ustekinumab treatment in a patient
50. Berger CT, Recher M, Steiner U, et al. A patient’s wish: anakinra with Crohn’s disease and paradoxical psoriasis. Journal of the
in pregnancy. Ann Rheum Dis 2009; 68: 1794–1795. European Academy of Dermatology and Venereology 2016; 30:
51. Chang Z, Spong CY, Jesus AA, et al. Anakinra use during preg- e191—e192.
nancy in patients with cryopyrin-associated periodic syndromes 69. Cortes X, Borrás-Blasco J, Antequera B, et al. Ustekinumab
(CAPS). Arthritis & rheumatology (Hoboken, NJ) 2014; therapy for Crohn’s disease during pregnancy: a case report
66: 3227–3232. and review of the literature. Journal of Clinical Pharmacy and
52. Ilgen U, Kucuksahin O. Anakinra use during pregnancy: Report Therapeutics 2017; 42: 234–236.
of a case with Familial Mediterranean Fever and infertility. Eur J 70. Lund T, Thomsen SF. Use of TNF-inhibitors and ustekinumab
Rheumatol 2017; 4: 66–67. for psoriasis during pregnancy: A patient series. Dermatol Ther
53. Kaneko K, Sugitani M, Goto M, et al. Tocilizumab and preg- 2017; 30. ePub ahead of print. DOI: 10.1111/dth.12454.
nancy: Four cases of pregnancy in young women with rheuma- 71. Danve A, Perry L, Deodhar A. Use of belimumab throughout
toid arthritis refractory to anti-TNF biologics with exposure to pregnancy to treat active systemic lupus erythematosus-A case
tocilizumab. Mod Rheumatol 2016; 26: 672–675. report. Semin Arthritis Rheum 2014; ePub ahead of print. DOI:
54. Weber-Schoendorfer C, Schaefer C. Pregnancy outcome after 10.1016/j.semarthrit.2014.05.006.
tocilizumab therapy in early pregnancy-a case series from the 72. Powell M, Hill D, Eudy A, et al. OP0041 Pregnancy Outcomes
German Embryotox Pharmacovigilance Center. Reprod Toxicol for Systemic Lupus Erythematosus (SLE) Subjects with
2016; 60: 29–32. Conception during Belimumab Intravenous (IV) and
55. Nakajima K, Watanabe O, Mochizuki M, et al. Pregnancy out- Subcutaneous (SC) Placebo-Controlled Clinical Trials and
comes after exposure to tocilizumab: A retrospective analysis of Long Term Extension Trials. Ann Rheum Dis 2014; 73: 75–76.
61 patients in Japan. Mod Rheumatol 2016; 26: 667–671. 73. Emmi G, Silvestri E, Squatrito D, et al. Favorable pregnancy
56. Hoeltzenbein M, Beck E, Rajwanshi R, et al. Tocilizumab use in outcome in a patient with systemic lupus erythematosus treated
pregnancy: Analysis of a global safety database including data with belimumab: A confirmation report. Semin Arthritis Rheum
from clinical trials and post-marketing data. Semin Arthritis 2016; 45: e26—e27.
Rheum 2016; 46: 238–245. 74. Bitter H, Bendvold AN, Ostensen ME. Lymphocyte changes and
57. Saito J, Yakuwa N, Takai C, et al. Tocilizumab concentrations in vaccination response in a child exposed to belimumab during
maternal serum and breast milk during breastfeeding and a safety pregnancy. Ann Rheum Dis 2018; ePub ahead of print. DOI:
assessment in infants: a case study. Rheumatology (Oxford) 10.1136/annrheumdis-2018-213004.
2018; ePub ahead of print. DOI: 10.1093/rheumatology/key091. 75. Ojeda-Uribe M, Afif N, Dahan E, et al. Exposure to abatacept or
58. Sullivan J, Warren RB, Reich K, et al. Secukinumab in pregnan- rituximab in the first trimester of pregnancy in three women with
cy: Outcomes from the global safety database (Abstract Number: autoimmune diseases. Clin Rheumatol 2013; 32: 695–700.
Soh and Moretto 13

76. Kumar M, Ray L, Vemuri S, et al. Pregnancy outcomes follow- inflammatory bowel disease. Aliment Pharmacol Ther 2014;
ing exposure to abatacept during pregnancy. Semin Arthritis 40(4): 363–373.
Rheum 2015; 45: 351–356. 86. Chakravarty EF, Murray ER, Kelman A, et al. Pregnancy out-
77. Bond SJ, Young L, Poulsen K, et al. FRI0141 Vaccination deci- comes after maternal exposure to rituximab. Blood 2011;
sions and incidence of neonatal infections in mothers exposed to 117: 1499–1506.
biologicals during pregnancy. Ann Rheum Dis 2018; 77: 614–615. 87. Skorpen CG, Lydersen S, Gilboe I-M, et al. Influence of disease
78. Cheent K, Nolan J, Shariq S, et al. Case Report: Fatal case of activity and medications on offspring birth weight, pre-eclampsia
disseminated BCG infection in an infant born to a mother taking and preterm birth in systemic lupus erythematosus: a population-
infliximab for Crohn’s disease. Journal of Crohn’s & Colitis 2010; based study. Ann Rheum Dis 2018; 77: 264–269.
4: 603–605. 88. Soh MC, Nelson-Piercy C. High-risk pregnancy and the rheuma-
79. Silva D, Silva MVD, Barros CCO, et al. TNF-alpha blockade tologist. Rheumatology 2015; 54: 572–587.
impairs in vitro tuberculous granuloma formation and down 89. Soh MC, Nelson-Piercy C, Dib F, et al. Association between
modulate Th1, Th17 and Treg cytokines. PloS one 2018; 13: pregnancy outcomes and cardiovascular deaths in women with
e0194430. ePub ahead of print. DOI: 10.1371/journal. systemic lupus erythematousus utilising Swedish population reg-
pone.0194430. istries. Arthritis & rheumatology (Hoboken, NJ) 2015;
80. Ehlers S. Tumor necrosis factor and its blockade in granuloma- 67: 2376–2382.
tous infections: differential modes of action of infliximab and 90. Soh MC, Dib F, Nelson-Piercy C, et al. Maternal-placental syn-
etanercept? Clinical infectious diseases : an official publication of drome and future risk of accelerated cardiovascular events in
the Infectious Diseases Society of America 2005; 41 Suppl Parous Swedish women with systemic lupus erythematosus - a
3: S199–S203. population-based retrospective cohort study with time-to-event
81. l’Ami MJ, Berkhout LC, Nurmohamed MT, et al. THU0211 Six analysis. Rheumatology (Oxford) 2016; Jul 55: 1235–1242.
months after treatment discontinuation tnf is still in complex 91. Soh MC, Nelson-Piercy C, Westgren M, et al. Do adverse preg-
with adalimumab. Ann Rheum Dis 2018; 77: 325. nancy outcomes contribute to accelerated cardiovascular events
82. Mahadevan U, Martin CF, Sandler RS, et al. 865 PIANO: A seen in young women with systemic lupus erythematosus? Lupus
1000 Patient Prospective Registry of Pregnancy Outcomes in 2017; 26: 1351–1367.
Women With IBD Exposed to Immunomodulators and 92. Kavanaugh A, Cush JJ, Ahmed MS, et al. Proceedings from the
Biologic Therapy. Gastroenterology 2012; 142(5): S–149. American College of Rheumatology Reproductive Health
83. Sebastian S, Ashton K, Houston Y, et al. Anti-TNF therapy Summit: the management of fertility, pregnancy, and lactation
induced immune neutropenia in Crohn’s disease- report of 2 in women with autoimmune and systemic inflammatory diseases.
cases and review of literature. J Crohns Colitis 2012; 6: 713–716. Arthritis Care Res (Hoboken) 2015; 67: 313–325.
84. Guiddir T, Fremond ML, Triki TB, et al. Anti-TNF-alpha ther- 93. Malek A. Role of IgG antibodies in association with placental
apy may cause neonatal neutropenia. Pediatrics 2014; 134: function and immunologic diseases in human pregnancy. Expert
e1189—e1193. Review of Clinical Immunology 2013; 9: 235–249.
85. Seirafi M, de Vroey B, Amiot A, et al. Factors associated with
pregnancy outcome in anti-TNF treated women with

You might also like