Annotated bibliography about 3 peers reviewed references

Develop an Annotated Bibliography for Use in Your Research Paper

Save Time On Research and Writing
Hire a Pro to Write You a 100% Plagiarism-Free Paper.
Get My Paper

Include the following in your annotated bibliography:

about 3 peer reviewed references

 

· APA citations and annotations for three (3) sources you deem relevant to your problem statement (thesis).

Save Time On Research and Writing
Hire a Pro to Write You a 100% Plagiarism-Free Paper.
Get My Paper

For each source:

· Cite the source in proper APA format. The citations should be organized in alphabetical order by author as in an APA References page. 

· Follow with a brief annotation that summarizes the source (approximately 3–5 sentences). You may quote from the source, but do not copy and paste from the abstract. 

· In 1 or 2 sentences, explain and evaluate the source’s relevance and significance to your research.  

· Use an academic tone and style. 

· It is a requirement that you include a copy of your working thesis statement at the top of your assignment.  

 

Simon et al. Arthritis Research & Therapy (2015) 17:212
DOI 10.1186/s13075-015-0728-9

RESEARCH ARTICLE Open Access

Incidence of malignancy in adult patients
with rheumatoid arthritis: a meta-analysis

Teresa A. Simon1*, Adam Thompson1, Kunal K. Gandhi1, Marc C. Hochberg2 and Samy Suissa3

Abstract

Introduction: Patients with rheumatoid arthritis (RA) are at an increased risk of malignancies compared with the
general population. This has raised concerns regarding these patients, particularly with the widespread use of
immunomodulating therapies, including biologic disease-modifying antirheumatic drugs (DMARDs). We performed
a systematic literature review and analysis to quantify the incidence of malignancies in patients with RA and the
general population to update previously published data.

Methods: A literature search was conducted that was consistent with and similar to that in a meta-analysis
published in 2008. MEDLINE, BIOSIS Previews, Embase, Derwent Drug File and SciSearch databases were searched
using specified search terms. Predefined inclusion criteria identified the relevant observational studies published
between 2008 and 2014 that provided estimates of relative risk of malignancy in patients with RA compared with
the general population. Risk data on overall malignancy and site-specific malignancies (lymphoma, melanoma and
lung, colorectal, breast, cervical and prostate cancer) were extracted. The standardized incidence ratios (SIRs; a
measure of risk) relative to the general population were evaluated and compared with published rates.

Results: A total of nine publications met the inclusion criteria. Seven of these reported SIRs for overall malignancy;
eight for lymphoma, melanoma, and lung, colorectal and breast cancer; seven for prostate cancer; and four for
cervical cancer. Compared with those in the general population, the SIR estimates for patients with RA suggest a
modest increased risk in overall malignancy, as previously observed. Patients with RA continued to show an increased
risk of lymphoma and lung cancer compared with the general population. Overall, SIR estimates for colorectal and
breast cancers continued to show a decrease in risk, whereas cervical cancer, prostate cancer and melanoma appeared
to show no consistent trend in risk among patients with RA compared with the general population.

Conclusions: The additional data evaluated here are consistent with previously reported data. Patients with RA are at
an increased risk of lung and lymphoma malignancies compared with the general population. Quantifying differences
in malignancy rates between non-biologic and biologic DMARD-treated patients with RA may further highlight which
malignancies may be related to treatment rather than to the underlying disease.

Introduction
Rheumatoid arthritis (RA) is a polygenic, multifactorial
and chronic immune-mediated disease characterized by
chronic joint inflammation, a predilection for develop-
ment of joint damage and deformity, and extraarticular
involvement [1]. The management of RA includes the
use of biologic and non-biologic disease-modifying anti-
rheumatic drugs (DMARDs) [2, 3], which act by directly
modifying immunologic pathways involved in the patho-
genesis of RA. The risk of malignancy among patients

* Correspondence: teresa.simon@bms.com
1Bristol-Myers Squibb, Princeton, NJ, USA
Full list of author information is available at the end of the article

© 2015 Simon et al. Open Access This articl
International License (http://creativecommo
reproduction in any medium, provided you
link to the Creative Commons license, and
Dedication waiver (http://creativecommons
article, unless otherwise stated.

with RA has been of ongoing interest and research be-
cause of the autoimmune pathogenesis that underlies
RA, the common etiology between rheumatic disease
and malignancy, and the use of immunomodulatory
therapy, such as DMARDs, that may alter normal immu-
nosurveillance and elevate the risk of malignancy [4, 5].
Understanding this potential therapeutic risk has be-
come more relevant with the increasing use of biologic
DMARDs as a routine therapeutic approach to RA man-
agement [3]. With more biologic treatment options avail-
able and initiation of biologic treatments occurring earlier,
it is important to understand the baseline risk of malig-
nancies in patients with RA. Furthermore, continuous

e is distributed under the terms of the Creative Commons Attribution 4.

0

ns.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
give appropriate credit to the original author(s) and the source, provide a
indicate if changes were made. The Creative Commons Public Domain
.org/publicdomain/zero/1.0/) applies to the data made available in this

http://crossmark.crossref.org/dialog/?doi=10.1186/s13075-015-0728-9&domain=pdf

mailto:teresa.simon@bms.com

http://creativecommons.org/licenses/by/4.0/

http://creativecommons.org/publicdomain/zero/1.0/

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 2 of 10

updates on the incidence of malignancies published in the
literature are crucial to allow better understanding of the
background rates for malignancy in clinical trials and ob-
servational research evaluating real-world practice.
Smitten et al. [6] performed a meta-analysis on the

risk of overall malignancy and several site-specific can-
cers, including overall lymphoma, Hodgkin disease, non-
Hodgkin lymphoma, lung cancer, colorectal cancer and
breast cancer, using data published between 1990 and
2007. Their meta-analysis suggested a small overall in-
crease in risk of malignancy, which was elevated for
lymphoma, Hodgkin disease, non-Hodgkin lymphoma
and lung cancer, but they found a decreased risk of colo-
rectal and breast cancer in patients with RA compared
with the general population [6]. The malignancies in-
cluded were prespecified and based on the most fre-
quently reported malignancies in an RA population.
In this article, we review the data on the incidence of

malignancy reported since the Smitten et al. [6] meta-
analysis and additionally evaluate the risk of other im-
portant site-specific malignancies, namely, melanoma,
cervical cancer and prostate cancer, which have been a
topic of discussion in recent publications [7–9].

Methods
Literature search
We conducted a search of the MEDLINE, BIOSIS
Previews, Embase, Derwent Drug File and SciSearch
databases for literature published between 1 January 2008
and 30 November 2014, using the following specified
search terms: cancer or tumor, tumour, malign* and rheu-
matoid arthritis or RA, and epidemiolog* or inciden*,
population, observation, retrospective or occurren*.

Exclusion algorithm
Predefined inclusion criteria were used to identify rele-
vant observational studies that provided estimates of
relative risk of malignancy in patients with RA compared
with the general population. The same criteria as de-
tailed by Smitten et al. [6] were applied. Studies were eli-
gible for inclusion if they fulfilled the following criteria:
(1) observational study design (including prospective,
retrospective, epidemiologic, database, survey, registry,
cohort and case–control), (2) reported malignancy out-
comes in patients with RA and a general population, (3)
enrolled more than 100 patients, (4) included only
patients older than 18 years of age, (5) covered any
geographic region and (6) were reported in English as a
full-length publication. Citations meeting the inclusion
criteria were obtained and screened for the outcomes of
interest, which included the observed incidence rates of
total malignancy and lymphoma, lung, colorectal, breast,
melanoma, prostate and cervical cancers in patients with
RA compared with the expected incidence rates in the

general population. Lymphoma was reported as Hodgkin
or non-Hodgkin where available. The selection of studies
for inclusion was made without regard to evaluation of
specific RA management strategies. We attempted to
avoid overlap by excluding studies for which updated
publications were available. We also excluded studies
that compared specific RA biologic treatments with
other RA treatments, unless overall RA and general
population rates were reported.

Data presentation
Citations meeting the inclusion criteria were screened for
outcomes of interest. Risk data on overall malignancy and
site-specific malignancies (overall lymphoma, Hodgkin
disease, non-Hodgkin lymphoma, melanoma, and lung,
colorectal, breast, cervical and prostate cancers) were
extracted independently by two people and compared for
accuracy. Study-specific estimates of relative risk were
measured mostly by overall and age- and sex-adjusted
standardized incidence ratios (SIRs) relative to non-RA
patients or the general population. In one study using a
case–control approach, researchers reported an odds
ratio, which is an accurate estimator of the relative risk
[10]. Thus, all measures of relative risk and their res-
pective 95 % confidence intervals (CIs) were then com-
pared with previously published rates. Pooled relative risks
and 95 % CIs were then computed using the DerSimonian
and Laird technique [11], in which a random-effects
model takes into consideration both within-study and
between-studies variation by incorporating the hetero-
geneity of the effects in the overall analysis. Some studies
in which authors reported data separately for colon and
rectal cancer, or for men and women, were considered
separate studies when we pooled relative risks.

Results
A total of 136 articles were identified using the defined
search criteria (Fig. 1). Following identification, a total of
33 potential publications were further analyzed; of these, 9
studies met all the inclusion criteria (Fig. 1) [10, 12–19].
These studies included population- and community-based
RA cohorts, examined data from 458 to 84,475 patients
and had mean follow-up periods ranging from 4 to
25 years. The relative risk of overall malignancy was
reported in seven studies [12–15, 17–19]; lymphoma
(overall lymphoma, Hodgkin disease or non-Hodgkin
lymphoma), lung, colorectal and breast cancers and mel-
anoma in eight [10, 12–17, 19]; prostate cancer in seven
[10, 12–17]; and cervical cancer in four [12, 14, 16, 17].
Overall risk by malignancy type was reported in eight
studies [10, 12–17, 19], both overall risk and risk by sex
were reported in three [15, 17, 18] and risk stratified by
sex only was reported in one [16].

136 publicatio ns identified

129 unique publications

1

21 publications

–7 duplicate publications

61 publications

33 publications

21 publications

15 publications

11 publications

10 publications met inclusion criteria
(1 publication was previously cited in

Smitten et al. [6] as in press and
therefore was not included)

–8 non-English publications

–60 not full manuscript publication s (i.e. meeting abstracts)

–28 not observational-type studies (i.e. reviews, meta-analyses, case reports)

–12 did not compare data to general population

–6 did not report malignancy outcome s

–4 did not report incidence data

–1 reported malignancy not evaluated in this publication

Fig. 1 Literature search data

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 3 of 10

Overall malignancies
In general, SIRs for overall malignancy across the studies
were similar. Six of seven studies reported a significant in-
crease in overall risk of malignancy (Fig. 2) [12–15, 17–32].
The total pooled SIR (95 % CI) for studies analyzed by
Smitten et al. (1.05 [1.01–1.09]) [6] and those included
here was significant: 1.09 (1.06–1.13). With the addition of
the newer studies, there was no change in the overall sum-
mary statistic showing that patients with RA have a 10 %
increase in overall malignancy risk compared with the
general population.

Lymphoma
In eight studies, authors reported outcomes as either
overall lymphoma, Hodgkin disease or non-Hodgkin
lymphoma [10, 12–17, 19]. Two studies reported only
lymphoma, whereas the remaining six studies reported
overall lymphoma, Hodgkin disease and non-Hodgkin
lymphoma rates, with all of the new studies reporting a sig-
nificant increase in risk of lymphoma in patients with RA
compared with the general population (Figs. 3, 4 and 5).
This ranged from almost a doubling (SIR 1.75) (Fig. 3)
[10, 15, 17, 21, 23, 24, 29, 30, 33] to a 12-fold increase

(SIR 12.82) (Fig. 4) [12, 14–16, 19, 22, 24, 25, 27–29, 34, 35]
in the risk of developing lymphoma. The pooled SIR for
Hodgkin disease (Fig. 4) was higher than for non-Hodgkin
lymphoma (Fig. 5) [12–16, 19, 20, 22, 24, 25, 27–29, 34–36].
The total pooled SIR (95 % CI) for the studies reviewed
here and by Smitten et al. [6] was 2.46 (2.05–2.96) for ma-
lignant lymphoma, 3.21 (2.42–4.27) for Hodgkin disease
and 2.26 (1.82–2.81) for non-Hodgkin lymphoma. The
overall risk with the addition of the new studies is con-
sistent with previously reported SIRs (2.08 [1.80–2.39],
3.29 [2.56–4.22] and 1.95 [1.70–2.24], respectively) [6];
however, a slight increase in risk for non-Hodgkin disease
was noted.

Lung cancer
In eight studies, authors reported that patients with RA
had an increased risk of lung cancer compared with the
general population [10, 12–17, 19]. The SIRs (95 % CI) for
lung cancer ranged from 1.36 (1.34–1.38) to 2.9 (1.6–4.8)
(Fig. 6) [10, 12–17, 19–22, 24–28, 32, 36–38]. The total
pooled SIR (95 % CI) for the studies reviewed here and by
Smitten et al. [6] was 1.64 (1.51–1.79) for lung cancer.
The risk of lung cancer in patients with RA compared

Source n/N

SIR (95% CI)

Matteson 1991 (DMARDs) [32] 20/530 1.52 (0.90, 2.6)†

Gridley 1993 [24] 840/11,683 0.95 (0.9, 1.0)
Moritomo 1995 [26] 26/655 1.38 (0.90, 2.02)

Kauppi 1996 [31] 540/9469 1.16 (1.07, 1.26)
Mellemkjaer 1996 [25] 1832/20,699 1.08 (1.03, 1.13)§

Cibere1997 [22] 136/862 0.8 (0.67, 0.95)
Thomas 2000 [27] 2029/26,623 1.01 (1.0, 1.1)
Ekstrom 2003 [29] 8896/76,527 1.07 (1.05, 1.09)
Askling 2005 [21] 3379/53,067 1.05 (1.01, 1.08)

Askling 2005 (early RA) [21] 138/3703 1.1 (0.9, 1.3)‡

Askling 2005 (anti-TNF) [21] 67/4160 0.9 (0.7, 1.2)‡

Geborek 2005 [30] 69/800 1.4 (1.1, 1.8)‡

Geborek 2005 (anti-TNF) [30] 16/757 1.1 (0.6, 1.8)
Abasolo 2007 [20] 25/789 1.2 (0.8, 1.8)†

Franklin 2007 [23] 92/1237 1.1 (0.9, 1.3)†

Wolfe 2007 [28] 543/13,869 1.0 (1.0, 1.1)†

Buchbinder 2008 [19] 64/458 1.5 (1.2, 1.9)
Hemminki 2008 [14] 4366/42,262 1.23 (1.19, 1.27)

Chen 2011 [12] 935/23,644 1.12 (1.11, 1.13)
Yamada 2011 [17] 173/7566 1.18 (1.02, 1.37)

Female* 1.13 (0.94, 1.36)
Male* 1.29 (0.99, 1.67)

Kim 2012 [18] 30/1501 0.86 (0.58, 1.23)
Female* 0.99 (0.65, 1.42)
Male* 0.31 (0.04, 1.11)

Dreyer 2013 [13] 128/3812 1.25 (1.05, 1.48)
Mercer 2013 [15] 182/3771 1.28 (1.10, 1.48)

Female* 1.38 (1.15, 1.64)
Male* 1.11 (0.85, 1.44)

Total pooled SIR 23 1.09 (1.06, 1.13)

0.1 10.01.0

New studies

Fig. 2 Relative risk of overall malignancy in patients with rheumatoid arthritis (RA) compared with the general population. CI, confidence interval;
DMARD, disease-modifying antirheumatic drug; n, number of malignancies; N, population size; RR, relative risk; SIR, standardized incidence ratio;
TNF, tumor necrosis factor. *SIRs by sex are not included in the total pooled SIR. †Excluding non-melanoma skin cancer. ‡All solid tumors. §Excluding
lymphatic and hematopoietic

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 4 of 10

with the general population in the contemporary study
is consistent with what was previously observed (1.63
[1.43–1.87]) [6].

Colorectal cancer
In seven studies [12, 14–17, 19], authors reported that
patients with RA had a decreased risk of colon or
colorectal cancer, with SIRs (95 % CI) ranging from
0.49 (0.26–0.83) to 0.96 (0.56–1.54) (Fig. 7) [10, 12–17,
19–22, 24–28, 31, 36]. However, in some studies, authors
reported SIRs >1, with the highest being 1.53 (0.69–3.42)

Source n/N
Gridley 1993 [24] 48/11,683

Ekstrom 2003 [29] 535/76,527

Askling 2005 [21] 319/53,067

Askling (early RA) [21] 11/3703

Askling 2005 (anti-TNF) [21] 9/4160

Geborek 2005 [30] 2/800

Geborek 2005 (anti-TNF) [30] 5/757

Franklin 2007 [23] 9/1237

Wolfe 2007 [33] 95/19,591

Hellgren 2010† [10] 19/6745

Yamada 2011 [17] 20/7566

Female*

Male*

Mercer 2013 [15] 21/3771

Female*
Male*

Total pooled SIR 12

0
New studies

Fig. 3 Relative risk of malignant lymphoma in patients with rheumatoid ar
interval; n, number of malignancies; N, population size; OR, odds ratio; SIR,
are not included in the total pooled SIR. †Reported as odds ratio

for rectal cancer [10, 13]. For colorectal cancer, the total
pooled SIR (95 % CI) for the studies reviewed by Smitten
et al. (0.77 [0.65–0.90]) [6] plus those included here was
0.78 (0.71–0.86).

Breast cancer
For breast cancer, there was generally no increase in
risk; estimated risk varied from 0.63 to 1.21 in the new
studies [10, 12–17, 19] (Fig. 8) [10, 12–17, 19–22,
24–28, 36]. The total pooled SIR (95 % CI) for the
studies reviewed by Smitten et al. (0.84 [0.79–0.90]) [6]

SIR (95% CI)

1.75

1.98 (1.5, 2.6)

2.00 (1.83, 2.17)

1.9 (1.7, 2.1)

2.0 (1.1, 3.5)

2.9 (1.3, 5.5)

1.3 (0.2, 4.5)

11.5 (3.7, 26.9)

2.94 (1.34, 5.57)

1.8 (1.5, 2.2)

(1.04, 2.96)

6.07 (3.71, 9.37)

6.00 (3.28, 10.07)

6.22 (2.28, 13.54)

3.81 (2.36, 5.82)

3.73 (1.98, 6.37)

3.95 (1.71, 7.79)
2.46 (2.05, 2.96)

30.01.0

thritis (RA) compared with the general population. CI, confidence
standardized incidence ratio; TNF, tumor necrosis factor. *SIRs by sex

Parikh-Patel 2009 [16]

Source n/N SIR (95% CI)

Gridley 1993 [24] 12/11,683 2.34 (1.2, 4.1)
Mellemkjaer 1996 [25] 14/20,699 3.4 (1.8, 5.6)

Cibere 1997 [22] 0/862 0.0 (0.00, 8.53)
Kauppi 1997 [34] 4/9469 2.2 (0.6, 5.7)

Thomas 2000 [27] 17/26,623 3.85 (2.2, 6.2)
Mariette 2002 (MTX) [35] 7/30,000 7.4 (3.0, 15.3)

Ekstrom 2003 [29] 77/76,527 3.06 (2.42, 3.83)
Wolfe 2007 [28] 4/13,869 3.0 (1.3, 6.8)

Buchbinder 2008 [19] 1/458 8.9 (0.2, 49.8)
Hemminki 2008 [14] 35/42,262 4.05 (2.82, 5.63)

25/84,475
Female* 1.62 (0.91, 2.68)
Male* 2.76 (1.32, 5.08)

Chen 2011 [12] 1/23,644 1.76 (1.45, 2.17)
Mercer 2013 [15] 5/3771 12.82 (4.16, 29.92)

Total pooled SIR 14 3.21 (2.42, 4.27)

0 50.01.0

New studies

Fig. 4 Relative risk of Hodgkin disease in patients with rheumatoid arthritis (RA) compared with the general population. CI, confidence interval;
n, number of malignancies; N, population size; MTX, methotrexate; SIR, standardized incidence ratio. *SIRs by sex for Parikh-Patel et al. [16] are included
in the total pooled SIR, as overall SIR was not available in their study

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 5 of 10

plus those included here was found to be 0.86 (0.73–1.01)
for breast cancer.

Melanoma and cervical and prostate cancers
As previously noted, we included three additional malig-
nancies (melanoma and cervical and prostate cancer) in
our assessment that were not included in the Smitten
et al. meta-analysis [6]. Figs. 9, 10 and 11 highlight the ar-
ticles analyzed by Smitten et al. [6] and the newer studies
included in our assessment. For melanoma, we reviewed
17 studies (9 from Smitten et al. [20–22, 24–28, 36] and 8
new [10, 12–17, 19]). Of these, the overall range of SIRs
was 0.3–8.83, and five studies [12, 14, 19, 28, 36] showed a
significant increase in the risk of melanoma in the RA
population, ranging from 1.29 to 3.0 (Fig. 9) [10, 12–17,
19–22, 24–28, 36]. The total pooled SIR was 1.23 (1.01–
1.49).
For cervical cancer, SIRs were reported in a total of 13

studies (nine from Smitten et al. [20–22, 24–28, 36] and

Source n/N
Gridley 1993 [24] 36/11,683

Mellemkjaer 1996 [25] 85/20,699
Cibere 1997 [22] 3/862
Kauppi 1997 [34] 34/9469

Mariette 2002 [35] 18/30,000
Thomas 2000 [27] 101/26,623
Ekstrom 2003 [29] 458/76,527

Setoguchi 2006 (DMARDs) [36] 58/7830
Abasolo 2007 [20] 3/789

Wolfe 2007 [28] 42/13,869

Buchbinder 2008 [19] 8/458
Hemminki 2008 [14] 280/42,262

Parikh-Patel 2009 [16] 325/84,475

Female*
Male*

Chen 2011 [12] 59/23,644
Dreyer 2013 [13] 5/3812
Mercer 2013 [15] 16/3771

Female*
Male*

Total pooled SIR 17

0.1

New studies

Fig. 5 Relative risk of non-Hodgkin lymphoma in patients with rheumatoid
interval; DMARD, disease-modifying antirheumatic drug; n, number of malig
sex included in total pooled SIR only if overall SIR was not available

four new [12, 14, 16, 17]). Overall, the range of SIRs was
0.43–2.15, and all confidence intervals were overlapping
(Fig. 10) [12, 14, 16, 17, 20–22, 24–28, 36]. In three
studies, authors reported a significant decrease in risk
compared with the RA population [12, 16, 36]. The total
pooled SIR for cervical cancer was 0.87 (0.72–1.05). For
prostate cancer, SIRs were reported in a total of 15
studies (8 from Smitten et al. [20–22, 24, 25, 27, 28, 36]
and 7 new [10, 12–17]). Overall, the range of SIRs was
0.35–3.20, and the total pooled SIR was 1.15 (0.98–1.34)
(Fig. 11) [10, 12–17, 20–22, 24, 25, 27, 28, 36].

Discussion
Although individual studies can show considerable vari-
ation, the results of this updated meta-analysis of the
incidence of malignancy in adult patients with RA are
generally consistent with previously reported data for all
specific malignancies [6]. The updated data reported here
show that patients with RA have a modest increased risk

SIR (95% CI)
1.88 (1.3, 2.6)
2.4 (1.9, 2.9)
0.55 (0.11, 1.60)
2.2 (1.5, 3.1)
1.07 (0.6, 1.7)
2.13 (1.7, 2.6)
1.89 (1.72, 2.07)
2.2 (1.71, 2.87)
5.4 (1.1, 15.7)
1.7 (1.3, 2.2)

5.1 (2.2, 10.0)
2.34 (2.07, 2.63)

1.37 (1.19, 1.57)
2.07 (1.71, 2.48)
3.54 (3.45, 3.63)
2.27 (0.94, 5.45)
3.12 (1.79, 5.07)
3.39 (1.69, 6.07)
2.66 (0.86, 6.21)
2.26 (1.82, 2.81)

20.01.0

arthritis (RA) compared with the general population. CI, confidence
nancies; N, population size; SIR, standardized incidence ratio. *SIRs by

0.1 50.01.0

Source n/N SIR (95% CI)
Matteson 1991 (DMARDs) [32] 6/530 3.37 (1.58, 7.34)

McKendry 1993 (MTX) [37] 4/144 12.4 (3.38, 31.84)
Gridley 1993 [24] 68/11,683 1.31 (1.0, 1.7)

Mo ritomo 1995 [26] 0/655 0 (0, 1.82)
Kauppi 1996 [38] 73/8920 1.8 (1.4, 2.2)

Mellemkjaer 1996 [25] 308/20,699 1.5 (1.3, 1.7)
Cibere 1997 [22] 16/862 1.08 (0.61, 1.75)

Thomas 2000 [27] 472/26,623 1.39 (1.3, 1.5)
Askling 2005 [21] 330/53,067 1.48 (1.33, 1.65)

Askling 2005 (early RA) [21] 23/3703 2.4 (1.5, 3.6)
Askling 2005 (anti-TNF) [21] 10/4160 1.8 (0.9, 3.3)

Setoguchi 2006 [36] 169/7830 1.8 (1.52, 2.05)
Abasolo 2007 [20] 7/789 3.5 (1.4, 7.1)

Wolfe 2007 [28] 112/13,869 1.2 (1.0, 1.4)

Buchbinder 2008 [19] 14/458 2.9 (1.6, 4.8)
Hemminki 2008 [14] 448/42,262 1.73 (1.57, 1.89)

Parikh-Patel 2009 [16] 1178/84,475
Female* 1.28 (1.19, 1.38)
Male* 1.65 (1.49, 1.81)

Hellgren 2010† [10] 34/6745 2.24 (1.49, 3.36)
Chen 2011 [12] 123/23,644 1.36 (1.34, 1.38)

Ya mada 2011 [17] 34/7566 2.29 (1.57, 3.21)
Female* 1.66 (0.89, 2.85)
Male* 3.02 (1.85, 4.67)

Dreyer 2013 [13] 20/3812 1.67 (1.08, 2.59)
Mercer 2013 [15] 46/3771 2.39 (1.75, 3.19)

Female* 2.66 (1.79, 3.80)
Male* 2.01 (1.15, 2.36)

Total pooled SIR 23 1.64 (1.51, 1.79)

New studies

Fig. 6 Relative risk of lung cancer in patients with rheumatoid arthritis (RA) compared with the general population. CI, confidence interval;
DMARD, disease-modifying antirheumatic drug; MTX, methotrexate; n, number of malignancies; N, population size; SIR, standardized incidence
ratio; TNF, tumor necrosis factor. *SIRs by sex are included in total pooled SIR only if overall SIR was not available. †Reported as odds ratio

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 6 of 10

of overall malignancy as well as an increased risk of lung
cancer and lymphoma when compared with the general
population. Lymphoma was associated with the greatest
relative risk, especially Hodgkin disease, with a 12-fold in-
crease reported in one study [15] and an increase in
women versus men in another [16]. Lung cancer generally
showed a twofold increase in risk [10, 12–17, 19]. As

Fig. 7 Relative risk of colorectal cancer in patients with rheumatoid arthritis (R
DMARD, disease-modifying antirheumatic drug; n, number of malignancies; N
factor. *SIRs by sex are included in total pooled SIR only if overall SIR was not

previously noted, a decreased risk overall of colorectal
cancer in patients with RA compared with the general
population was found. For melanoma, the pooled SIR was
significantly greater than unity, although few of the indi-
vidual point estimates were statistically significant. Breast
and cervical cancers in general showed a decrease in risk
overall that failed to reach statistical significance.

A) compared with the general population. CI, confidence interval;
, population size; SIR, standardized incidence ratio; TNF, tumor necrosis
available. †Reported as odds ratio

Source n/N SIR (95% CI)
Gridley 1993 [24] 106/11,683 0.79 (0.6, 1.0)

Moritomo 1995 [26] 3/655 1.68 (0.34, 4.91)
Mellemkjaer 1996 [25] 186/20,699 0.8 (0.7, 0.9)

Cibere 1997 [22] 18/862 0.9 (0.46, 1.24)
Thomas 2000 [27] 249/26,623 0.95 (0.83, 1.07)
Askling 2005 [21] 471/53,067 0.83 (0.76, 0.91)

Askling 2005 (early RA) [21] 13/3703 0.6 (0.3, 1.0)
Askling 2005 (anti-TNF) [21] 8/4160 0.4 (0.2, 0.9)

Abasolo 2007 [20]2007 [20] 2/789 0.9 (0.1, 3.2)
Setoguchi 2006 (DMARDs) [36] 112/7830 0.9 (0.73, 1.06)

Wolfe 2007 [28] 102/13,869 0.8 (0.6, 0.9)

Buchbinder 2008 [19] 4/458 0.7 (0.2, 1.7)
Hemminki 2008 [14] 642/42,262 0.97 (0.90, 1.05)

Parikh-Patel 2009 [16] 842/84,475 0.63 (0.59, 0.67)
Hellgren 2010* [10] 42/6745 0.89 (0.64, 1.23)

Chen 2011 [12] 123/23,644 1.21 (1.19, 1.23)
Yamada 2011 [17] 20/7566 1.05 (0.64, 1.62)

Dreyer 2013 [13] 14/3812 0.89 (0.53, 1.51)
Mercer 2013 [15] 30/3771 1.07 (0.72, 1.52)

Total pooled SIR 19 0.86 (0.73, 1.01)

0.1 10.01.0
New studies

Fig. 8 Relative risk of breast cancer in patients with rheumatoid arthritis (RA) compared with the general population. CI, confidence interval;
DMARD, disease-modifying antirheumatic drug; n, number of malignancies; N, population size; SIR, standardized incidence ratio; TNF, tumor necrosis
factor. *Reported as odds ratio

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 7 of 10

Various explanations for the differences observed in
the risk of certain malignancies in patients with RA have
been proposed. It is thought that ongoing immunologic
stimulation over time may increase the risk of malignant
transformation of immune system cells and decrease the
number of T-suppressor lymphocytes, thus increasing
rates of lymphoma malignancy in patients with RA [4].
Chronic lung inflammation due to the disease in patients
with RA may explain the increased risk of lung cancer.
Smoking increases the risk of both RA and lung cancer;
however, the SIRs reported here were not adjusted for
smoking, so it is not possible to rule out the increase in

Askling 2005 (anti-TNF) [21]

Source n/N
Gridley 1993 [24] 12/11,683

Female*
Male*

Moritomo 1995 [26] 1/655
Female*

Mellemkjaer 1996 [25] 37/20,699
Female*
Male*

Cibere 1997 [22] 45/862
Thomas 2000 [27] 28/26,623

Female*
Male*

Askling 2005 [21] 120/53,067
Askling 2005 (early RA) [21] 4/3703

1/4160
Setoguchi 2006 (DMARDs) [36] 29/7830

Abasolo 2007 [20] 1/789
Male*

Wolfe 2007 [28] 32/13,869

Buchbinder 2008 [19] 7/458
Hemminki 2008 [14] 143/42,262

Parikh-Patel 2009 [16] 184/84,475
Female*
Male*

Hellgren 2010† [10] 11/6745
Chen 2011 [12] 3/23,644

Yamada 2011 [17] 4/7566
Female*
Male*

Dreyer 2013 [13] 3/3812
Mercer 2013 [15] 9/3771

Female*
Total pooled SIR 21

0
New studies

Fig. 9 Relative risk of melanoma in patients with rheumatoid arthritis (RA) co
disease-modifying antirheumatic drug; n, number of malignancies; N, populat
*SIRs by sex are included in total pooled SIR only if overall SIR was not availab

SIR being due to an indirect association. Thus, the risk
of both lymphoma and lung cancer has been hypothe-
sized to be dependent on the level of disease activity ex-
perienced by the patient [4, 6].
Patients with RA continued to show no increase in

overall risk of colorectal and breast cancers, in contrast
to their increased risk for lung cancer and lymphoma. In
patients with RA, any observed decreased risk in colo-
rectal malignancy may be due to the increased use of
non-steroidal anti-inflammatory drugs, which are known
to decrease this risk [4, 6]. Overall, the risk of malig-
nancy among patients with RA may be due in part to

SIR (95% CI)
0.93 (0.5, 1.16)
1.23 (0.6, 2.2)
0.25 (0, 1.4)
8.83 (0.12, 49.10)
13.3 (0.17, 74.0)
1.1 (0.8, 1.5)
1.1
1.3
0.83 (0.63, 1.16)

1.21 (0.79, 1.77)
0.34 (0.04, 1.22)
1.19 (0.99, 1.42)
0.9 (0.2, 2.2)
0.3 (0.0, 1.8)
2.3 (1.55, 3.22)
3.8 (0.1, 21.0)
17.1 (0.4, 95.2)
1.7 (1.3, 2.3)

3.0 (1.2, 6.2)
1.29 (1.09, 1.52)

0.63 (0.51, 0.76)
0.80 (0.63, 1.00)
1.04 (0.54, 1.99)
1.47 (1.31, 1.65)
2.34 (0.64, 6.00)
1.66 (0.20, 6.01)
3.96 (0.48, 14.30)
1.00 (0.32, 3.11)
2.05 (0.94, 3.90)
2.05 (0.75, 4.46)
1.23 (1.01, 1.49)

100.01.0

mpared with the general population. CI, confidence interval; DMARD,
ion size; SIR, standardized incidence ratio; TNF, tumor necrosis factor.
le. †Reported as odds ratio

Source n/N SIR (95% CI)
Gridley 1993 [24] 17/11,683 0.9 (0.5, 1.4)

Moritomo 1995 [26] 3/655 2.15 (0.43, 6.27)
Mellemkjaer 1996 [25] 40/20,699 1.1 (0.8, 1.5)

Cibere 1997 [22] 1/862 0.49 (0.06, 2.76)
Thomas 2000 [27] 26/26,623 0.89 (0.58, 1.31)
Askling 2005 [21] 33/53,067 1.03 (0.71, 1.45)

Askling 2005 (early RA) [21] 1/3703 0.8 (0.02, 4.3)
Askling 2005 (anti-TNF) [21] 1/4160 1.0 (0.0, 5.8)

Setoguchi 2006 (DMARDs) [36] 16/7830 0.5 (0.31, 0.82)
Abasolo 2007 [20] 1/789 1.0 (0.1, 22.7)

Wolfe 2007 [28] 4/13,869 0.8 (0.4, 1.9)

Hemminki 2008 [14] 58/42,262 1.26 (0.95, 1.62)
Parikh-Patel 2009 [16] 47/84,475 0.43 (0.31, 0.57)

Chen 2011 [12] 45/23,644 0.86 (0.84, 0.89)
Yamada 2011 [17] 4/7566 1.36 (0.37, 3.49)
Total pooled SIR 15 0.87 (0.72, 1.05)

0 30.0 1.0

New studies

Fig. 10 Relative risk of cervical cancer in patients with rheumatoid arthritis (RA) compared with the general population. CI, confidence interval;
DMARD, disease-modifying antirheumatic drug; n, number of malignancies; N, population size; SIR, standardized incidence ratio; TNF, tumor
necrosis factor

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 8 of 10

the autoimmune pathogenesis of RA and common
etiology between RA and malignancy, including genetic
factors, smoking-related tissue necrosis and viral infec-
tion [9]. The three other site-specific malignancies ex-
amined here but not studied previously by Smitten et al.
[6]—prostate cancer, cervical cancer and melanoma—are
all common cancers, with cervical cancer being attribut-
able to viral infection. In the present analysis, there did
not appear to be a consistent trend of increased or de-
creased risk of prostate cancer, cervical cancer or melan-
oma among patients with RA compared with the general
population. One review published in 2008 that did not
meet our criteria showed melanoma results that were con-
sistent with our present assessment. Some populations
showed an increase in risk, whereas others did not [8].
This review, like that of Smitten et al. [6], is based on

observational, real-world clinical data; however, these in-
dividual studies may have potential limitations. Although
most of the individual estimates were derived from simi-
lar database studies, their study designs (including popu-
lations, data sources, follow-up times, data analysis and

Fig. 11 Relative risk of prostate cancer in patients with rheumatoid arthritis
n, number of malignancies; N, population size; SIR, standardized incidence

procedures of reporting, particularly when linked to a
cancer registry) were a source of heterogeneity among
the studies and may be associated with bias of varying
magnitude. In addition, other potential sources of hetero-
geneity included geography and/or region, stage and se-
verity of RA, and treatment. Such heterogeneity should be
considered in any meta-analysis; the DerSimonian and
Laird technique used in this study [11] assumes hetero-
geneity across studies and incorporates it into the esti-
mation of the pooled SIRs and their confidence limits.
Therefore, although the variation observed among the in-
dividual SIRs reported should be treated with caution
when interpreting these data, it should be recognized that
this variation was accounted for in the analyses resulting
in the pooled SIRs.
For example, with regard to treatment, Dreyer et al. [13]

evaluated the effect of tumor necrosis factor (TNF) inhibi-
tor treatment on risk rate by examining the treated and
non-treated populations separately (only non-treated data
included in this analysis), whereas Mercer et al. [15]
evaluated biologic-naive patients. Buchbinder et al. [19]

(RA) compared with the general population. CI, confidence interval;
ratio. *Reported as odds ratio

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 9 of 10

examined malignancy risk only in patients with RA receiv-
ing methotrexate. However, the rest of the patients in
these studies are likely to have received treatments for
RA, therefore making it difficult to differentiate the effects
of treatment and disease. Nonetheless, to further under-
stand which malignancies may be related to treatment ver-
sus the underlying disease, it is important, where possible,
to quantify differences in malignancy rates between pa-
tients with RA treated with non-biologic DMARDs and
those treated with biologic DMARDs. Whether biologic
agents, predominantly TNF inhibitors, increase the risk of
malignancy has been debated [9]. Data have shown that
use of TNF inhibitors was not associated with a major
further increase in risk in the already elevated lymphoma
occurrence in patients with RA [39–44]. However, they do
appear to increase the risk of skin cancer, including
melanoma [39–43]. Furthermore, the data presented by
Simon et al. suggest that the observed incidence of total
malignancy and those for breast, colorectal and lung can-
cers and lymphoma in patients treated with abatacept was
largely consistent with that expected of patients with RA
treated with non-biologic DMARDs [45].

Conclusions
The additional data presented here are consistent with
previously reported data [6] for all specified malignancies.
For overall malignancy, the new studies analyzed here
show a similar increase in risk compared with that pre-
viously reported. In addition, patients with RA continue to
be at an increased risk of lung cancer and lymphoma when
compared with the general population. Further studies
examining specific aspects such as treatments, smoking or
other lifestyle factors are needed to investigate the under-
lying mechanisms for the increased or decreased risk of
specific cancers observed in patients with RA compared
with the general population. Also, it is important to quan-
tify any differences in malignancy risks between patients
with RA who are treated with non-biologic DMARDs and
those treated with biologic DMARDs, as understanding
which malignancies may be related to the treatment as
opposed to being related to underlying disease will allow
patients to be advised and monitored accordingly.

Abbreviations
CI: confidence interval; DMARD: disease-modifying anti-rheumatic drug;
MTX: methotrexate; NR: not reported; OR: odds ratio; RA: rheumatoid arthritis;
RR: relative risk; SIR: standardized incidence ratio; TNF: tumor necrosis factor.

Competing interests
TAS and KKG are employees and stockholders of Bristol-Myers Squibb. AT is
an intern at Bristol-Myers Squibb and a medical student at Rowan University
School of Osteopathic Medicine, Stratford, NJ, USA. MCH receives grant/
research support from the National Institutes of Health and is a consultant
for Bristol-Myers Squibb, Eli Lilly, EMD Serono, Genentech/Roche, Novartis
Pharma AG, Pfizer and UCB. SS is a consultant for Bristol-Myers Squibb and
Genentech/Roche. The authors declare that they have no non-financial
competing interests.

Authors’ contributions
TAS participated in the design and coordination of data collection,
contributed to analysis and interpretation of data and helped to draft the
manuscript. AT conducted the literature search and helped to draft the
manuscript. KKG contributed to the study design and data collection and
interpretation and helped to draft the manuscript. MCH contributed to the
study design and interpretation of the data and helped to draft the manuscript.
SS contributed to the study design, performed the meta-analysis, contributed
to data interpretation and helped to draft the manuscript. All authors read and
approved the final manuscript.

Acknowledgments
Bristol-Myers Squibb funded the study and reviewed and approved the
manuscript before its submission. The authors had ultimate control over the
decision to publish and the final version of the manuscript submitted for
publication. The authors thank Julia Chuang of Bristol-Myers Squibb for
providing research assistance. Professional medical writing and editorial
assistance was provided by Fiona Boswell at Caudex and was funded by
Bristol-Myers Squibb.

Author details
1Bristol-Myers Squibb, Princeton, NJ, USA. 2Departments of Medicine and
Epidemiology and Public Health, University of Maryland School of Medicine,
Baltimore, MD, USA. 3Division of Clinical Epidemiology, McGill University,
Montreal, QC, Canada.

Received: 13 May 2015 Accepted: 24 July 2015

References
1. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham 3rd CO, et al.

2010 rheumatoid arthritis classification criteria: an American College of
Rheumatology/European League Against Rheumatism collaborative
initiative. Arthritis Rheum. 2010;62:2569–81.

2. Smolen JS, Landewé R, Breedveld FC, Dougados M, Emery P, Gaujoux-Viala C,
et al. EULAR recommendations for the management of rheumatoid arthritis
with synthetic and biological disease-modifying antirheumatic drugs. Ann
Rheum Dis. 2010;69:964–75.

3. Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF, Kremer JM, et al. 2012
update of the 2008 American College of Rheumatology recommendations
for the use of disease-modifying antirheumatic drugs and biologic agents
in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken).
2012;64:625–39.

4. Love T, Solomon DH. The relationship between cancer and rheumatoid
arthritis: still a large research agenda. Arthritis Res Ther. 2008;10:109.

5. Cush JJ, Dao KH. Malignancy risks with biologic therapies. Rheum Dis Clin
North Am. 2012;38:761–70.

6. Smitten AL, Simon TA, Hochberg MC, Suissa S. A meta-analysis of the
incidence of malignancy in adult patients with rheumatoid arthritis. Arthritis
Res Ther. 2008;10:R45.

7. Kim SC, Glynn RJ, Giovannucci E, Hernández-Díaz S, Liu J, Feldman S, et al.
Risk of high-grade cervical dysplasia and cervical cancer in women with
systemic inflammatory diseases: a population-based cohort study. Ann
Rheum Dis. 2015;74:1360–7.

8. Chakravarty EF, Farmer ER. Risk of skin cancer in the drug treatment of
rheumatoid arthritis. Expert Opin Drug Saf. 2008;7:539–46.

9. Szekanecz Z, Szekanecz É, Bakó G, Shoenfeld Y. Malignancies in
autoimmune rheumatic disease – a mini-review. Gerontology. 2011;57:3–10.

10. Hellgren K, Smedby KE, Feltelius N, Baecklund E, Askling J. Do rheumatoid
arthritis and lymphoma share risk factors? A comparison of lymphoma and
cancer risks before and after diagnosis of rheumatoid arthritis. Arthritis
Rheum. 2010;62:1252–8.

11. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials.
1986;7:177–88.

12. Chen YJ, Chang YT, Wang CB, Wu CY. The risk of cancer in patients with
rheumatoid arthritis: a nationwide cohort study in Taiwan. Arthritis Rheum.
2011;63:352–8.

13. Dreyer L, Mellemkjær L, Andersen AR, Bennett P, Poulsen UE, Juulsgaard
Elligsen T, et al. Incidences of overall and site specific cancers in TNFα
inhibitor treated patients with rheumatoid arthritis and other arthritides – a
follow-up study from the DANBIO Registry. Ann Rheum Dis. 2013;72:79–82.

Simon et al. Arthritis Research & Therapy (2015) 17:212 Page 10 of 10

14. Hemminki K, Li X, Sundquist K, Sundquist J. Cancer risk in hospitalized
rheumatoid arthritis patients. Rheumatology. 2008;47:698–701.

15. Mercer L, Davies R, Galloway JB, Low A, Lunt M, Dixon WG, et al. Risk of
cancer in patients receiving non-biologic disease-modifying therapy for
rheumatoid arthritis compared with the UK general population.
Rheumatology (Oxford). 2013;52:91–8.

16. Parikh-Patel A, White RH, Allen M, Cress R. Risk of cancer among rheumatoid
arthritis patients in California. Cancer Causes Control. 2009;20:1001–10.

17. Yamada T, Nakajima A, Inoue E, Tanaka E, Taniguchi A, Momohara S, et al.
Incidence of malignancy in Japanese patients with rheumatoid arthritis.
Rheumatol Int. 2011;31:1487–92.

18. Kim YJ, Shim JS, Choi CB, Bae SC. Mortality and incidence of malignancy in
Korean patients with rheumatoid arthritis. J Rheumatol. 2012;39:226–32.

19. Buchbinder R, Barber M, Heuzenroeder L, Wluka AE, Giles G, Hall S, et al.
Incidence of melanoma and other malignancies among rheumatoid arthritis
patients treated with methotrexate. Arthritis Rheum. 2008;59:794–9.

20. Abásolo L, Júdez E, Descalzo MÁ, González-Álvaro I, Jover JA, Carmona L,
et al. Cancer in rheumatoid arthritis: occurrence, mortality and associated
factors in a South European population. Semin Arthritis Rheum.
2008;37:388–97.

21. Askling J, Fored CM, Brandt L, Baecklund E, Bertilsson L, Feltelius N, et al.
Risks of solid cancers in patients with rheumatoid arthritis and after
treatment with tumour necrosis factor antagonists. Ann Rheum Dis.
2005;64:1421–6.

22. Cibere J, Sibley J, Haga M. Rheumatoid arthritis and the risk of malignancy.
Arthritis Rheum. 1997;40:1580–6.

23. Franklin J, Lunt M, Bunn D, Symmons D, Silman A. Influence of inflammatory
polyarthritis on cancer incidence and survival: results from a community-based
prospective study. Arthritis Rheum. 2007;56:790–8.

24. Gridley G, McLaughlin JK, Ekbom A, Klareskog L, Adami HO, Hacker DG,
et al. Incidence of cancer among patients with rheumatoid arthritis. J Natl
Cancer Inst. 1993;85:307–11.

25. Mellemkjaer L, Linet MS, Gridley G, Frisch M, Møller H, Olsen JH. Rheumatoid
arthritis and cancer risk. Eur J Cancer. 1996;32A:1753–7.

26. Moritomo H, Ueda T, Hiyama T, Hosono N, Mori S, Komatsubara Y. The risk of
cancer in rheumatoid patients in Japan. Scand J Rheumatol. 1995;24:157–9.

27. Thomas E, Brewster DH, Black RJ, Macfarlane GJ. Risk of malignancy among
patients with rheumatic conditions. Int J Cancer. 2000;88:497–502.

28. Wolfe F, Michaud K. Biologic treatment of rheumatoid arthritis and the risk
of malignancy: analyses from a large US observational study. Arthritis
Rheum. 2007;56:2886–95.

29. Ekström K, Hjalgrim H, Brandt L, Baecklund E, Klareskog L, Ekbom A, et al.
Risk of malignant lymphomas in patients with rheumatoid arthritis and in
their first-degree relatives. Arthritis Rheum. 2003;48:963–70.

30. Geborek P, Bladström A, Turesson C, Gulfe A, Petersson IF, Saxne T, et al.
Tumour necrosis factor blockers do not increase overall tumour risk in
patients with rheumatoid arthritis, but may be associated with an increased
risk of lymphomas. Ann Rheum Dis. 2005;64:699–703.

31. Kauppi M, Pukkala E, Isomaki H. Low incidence of colorectal cancer in
patients with rheumatoid arthritis. Clin Exp Rheumatol. 1996;14:551–3.

32. Matteson EL, Hickey AR, Maguire L, Tilson HH, Urowitz MB. Occurrence of
neoplasia in patients with rheumatoid arthritis enrolled in a DMARD
Registry. Rheumatoid Arthritis Azathioprine Registry Steering Committee.
J Rheumatol. 1991;18:809–14.

33. Wolfe F, Michaud K. The effect of methotrexate and anti-tumor necrosis
factor therapy on the risk of lymphoma in rheumatoid arthritis in 19,562
patients during 89,710 person-years of observation. Arthritis Rheum.
2007;56:1433–9.

34. Kauppi M, Pukkala E, Isomäki H. Elevated incidence of hematologic
malignancies in patients with Sjögren’s syndrome compared with patients
with rheumatoid arthritis (Finland). Cancer Causes Control. 1997;8:201–4.

35. Mariette X, Cazals-Hatem D, Warszawki J, Liote F, Balandraud N, Sibilia J,
et al. Lymphomas in rheumatoid arthritis patients treated with
methotrexate: a 3-year prospective study in France. Blood. 2002;99:3909–15.

36. Setoguchi S, Solomon DH, Weinblatt ME, Katz JN, Avorn J, Glynn RJ, et al.
Tumor necrosis factor α antagonist use and cancer in patients with
rheumatoid arthritis. Arthritis Rheum. 2006;54:2757–64.

37. McKendry RJ, Dale P. Adverse effects of low dose methotrexate therapy in
rheumatoid arthritis. J Rheumatol. 1993;20:1850–6.

38. Kauppi M, Pukkala E, Isomäki H. Excess risk of lung cancer in patients with
rheumatoid arthritis. J Rheumatol. 1996;23:1484–5.

39. Mariette X, Matucci-Cerinic M, Pavelka K, Taylor P, van Vollenhoven R,
Heatley R, et al. Malignancies associated with tumour necrosis factor
inhibitors in registries and prospective observational studies: a systematic
review and meta-analysis. Ann Rheum Dis. 2011;70:1895–904.

40. Park HJ, Ranganathan P. TNF-α antagonism and cancer risk in rheumatoid
arthritis: is continued vigilance warranted? Discov Med. 2012;13:229–34.

41. Le Blay P, Mouterde G, Barnetche T, Morel J, Combe B. Risk of malignancy
including non-melanoma skin cancers with anti-tumor necrosis factor
therapy in patients with rheumatoid arthritis: a meta-analysis of registries
and systematic review. Clin Exp Rheumatol. 2012;30:756–64.

42. Askling J, Fahrbach K, Nordstrom B, Ross S, Schmid CH, Symmons D. Cancer
risk with tumor necrosis factor α (TNF) inhibitors: meta-analysis of
randomized controlled trials of adalimumab, etanercept, and infliximab
using patient level data. Pharmacoepidemiol Drug Saf. 2011;20:119–30.

43. Damjanov N, Nurmohamed MT, Szekanecz Z. Biologics, cardiovascular
effects and cancer. BMC Med. 2014;12:48. doi:10.1186/1741-7015-12-48.

44. Askling J, Baecklund E, Granath F, Geborek P, Fored M, Backlin C, et al.
Anti-tumour necrosis factor therapy in rheumatoid arthritis and risk of
malignant lymphomas: relative risks and time trends in the Swedish
Biologics Register. Ann Rheum Dis. 2009;68:648–53.

45. Simon TA, Smitten AL, Franklin J, Askling J, Lacaille D, Wolfe F, et al.
Malignancies in the rheumatoid arthritis abatacept clinical development
programme: an epidemiological assessment. Ann Rheum Dis. 2009;68:1819–26.

Submit your next manuscript to BioMed Central
and take full advantage of:

• Convenient online submission

• Thorough peer review

• No space constraints or color figure charges

• Immediate publication on acceptance

• Inclusion in PubMed, CAS, Scopus and Google Scholar

• Research which is freely available for redistribution

Submit your manuscript at
www.biomedcentral.com/submit

http://dx.doi.org/10.1186/1741-7015-12-48

Reproduced with permission of the copyright owner. Further reproduction prohibited without
permission.

  • c.ICABMDD_RRTH_20150101_B9B0CE553FC60B6A4530_893
  • Abstract
    Introduction
    Methods
    Results
    Conclusions
    Introduction
    Methods
    Literature search
    Exclusion algorithm
    Data presentation
    Results
    Overall malignancies
    Lymphoma
    Lung cancer
    Colorectal cancer
    Breast cancer
    Melanoma and cervical and prostate cancers
    Discussion
    Conclusions
    Abbreviations
    Competing interests
    Authors’ contributions
    Acknowledgments
    Author details
    References

REVIEW Open Access

Immunosuppressive drugs and fertility
Clara Leroy1,2, Jean-Marc Rigot2, Maryse Leroy3, Christine Decanter4, Kristell Le Mapihan1, Anne-Sophie Parent1,
Anne-Claire Le Guillou1, Ibrahim Yakoub-Agha5, Sébastien Dharancy6, Christian Noel7

and Marie-Christine Vantyghem1,8*

Abstract

Immunosuppressive drugs are used in the treatment of inflammatory and autoimmune diseases, as well as in
transplantation. Frequently prescribed in young people, these treatments may have deleterious effects on
fertility, pregnancy outcomes and the unborn child. This review aims to summarize the main gonadal side
effects of immunosuppressants, to detail the effects on fertility and pregnancy of each class of drug, and to
provide recommendations on the management of patients who are seen prior to starting or who are already
receiving immunosuppressive treatment, allowing them in due course to bear children. The recommendations
for use are established with a rather low level of proof, which needs to be taken into account in the patient
management. Methotrexate, mycophenolate, and le- and teri-flunomide, cyclophosphamide, mitoxanthrone are
contraindicated if pregnancy is desired due to their teratogenic effects, as well as gonadotoxic effects in the
case of cyclophosphamide. Anti-TNF-alpha and mTOR-inhibitors are to be used cautiously if pregnancy is desired, since
experience using these drugs is still relatively scarce. Azathioprine, glucocorticoids, mesalazine, anticalcineurins such as
cyclosporine and tacrolimus, ß-interferon, glatiramer-acetate and chloroquine can be used during pregnancy, bearing
in mind however that side effects may still occur. Experience is limited concerning natalizumab, fingolimod, dimethyl-
fumarate and induction treatments. Conclusion: At the time of prescription, patients must be informed of the possible
consequences of immunosuppressants on fertility and of the need for contraception. Pregnancy must be planned and
the treatment modified if necessary in a pre-conception time period adapted to the half-life of the drug, imperatively
in relation with the prescriber of the immunosuppressive drugs.

Keywords: Fertility, Pregnancy, Transplantation, Auto-immune diseases, Inflammatory diseases, Immunosuppressive
drugs: calcineurin inhibitor, Azathioprine, Corticosteroids, Mesalazine, Chloroquine, Cyclophosphamide, Methotrexate,
Mycophenolate, Leflunomide, Anti-TNF, mTOR inhibitors, Beta-interferon, Glatiramer, Natalizumab, Fingolimod,
Mitoxantrone, Dimethylfumarate

Disease name and definition
Immunosuppressive treatments, with their increasingly
varied mechanisms of action, are used in some inflam-
matory and autoimmune diseases, as well as in trans-
plantation. Frequently prescribed in young people, these
therapies can have deleterious effects on fertility, preg-
nancy outcomes and the unborn child. The aim of this
report is to summarise the main gonadal side effects of
immunosuppressive drugs, to detail the effects on fertil-
ity and pregnancy of each class of drug and to provide

practice guidelines on the management of patients who
are seen prior to starting or are already receiving im-
munosuppressive treatment.

Epidemiology
In Europe, close to 300,000 people have had transplant-
ation, with this number having risen by 45 % since 2000.
Autoimmune diseases have become the 3rd leading cause
of morbidity and mortality following cardiovascular dis-
ease and cancer.
The present review is based on a comprehensive PubMed

search between the dates of January 1, 1960, to October 1,
2014, using the search term fertility and pregnancy com-
bined with the different immunosuppressive drugs and
discussed according to the multidisciplinary clinical

* Correspondence: mc-vantyghem@chru-lille.fr
1Endocrinology and Metabolism, Hôpital Huriez, Lille University Hospital,
59037 Lille Cedex, France
8InsermU859 Biotherapies of Diabetes, Lille University Hospital, 59037 Lille
Cedex, France
Full list of author information is available at the end of the article

© 2015 Leroy et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136
DOI 10.1186/s13023-015-0332-8

http://crossmark.crossref.org/dialog/?doi=10.1186/s13023-015-0332-8&domain=pdf

mailto:mc-vantyghem@chru-lille.fr

http://creativecommons.org/licenses/by/4.0/

http://creativecommons.org/publicdomain/zero/1.0/

experience of the authors. Most of the published studies,
being retrospective, are observational studies without con-
trol groups, or are clinical case studies. Randomised, con-
trol studies are difficult to conduct for obvious ethical
reasons. There is very little data for some drugs, and they
are mainly based on animal studies.
Fertility after organ transplantation however has been

very well studied, but lacks the ability to distinguish the
roles played by immunosuppressive drugs from that
of improvement of general health after transplantation.
After liver or kidney transplantation, the rates of miscar-
riage range from 15 to 27 % [1–4], values that are
comparable to those of the general population, even if
considerably higher incidences (45 %) were observed
after kidney transplantation [5] between 1990 and 2003 in
the United States (US). The incidences of pre-eclampsia (5
to 15 %), intrauterine growth restriction (IUGR, 9 to
57 %), Caesarean sections (38 to 80 %), prematurity (4 to
50 %) and low-birth weight (32 %), heterogeneous accord-
ing to the studies and probably according to the under-
lying maternal condition, seem slightly higher than in the
general population [1, 2, 4]. Overall live birth rates, how-
ever, currently match that in the US population according
to the United States National Transplantation Pregnancy
and United Kingdom registries (80 %), as well as Desh-
pande’s meta-analysis (73.5 %) [4, 6]. The risk of foetal
anomalies is very depending on the type of drugs but this
risk can now be anticipated.

Clinical description of the consequences of
immunosuppressive drugs on fertility and
pregnancy
Most of the time, the intensity of immuno-suppression
decreases with time, especially after the first year in allo-
geneic organ transplantation. In addition, allogeneic
hematopoietic stem-cell transplant is preceded by the
eradication of diseased cells by chemotherapy, using drugs
such as cyclophosphamide, a cytotoxic and very gonado-
toxic immunosuppressive agent.

Immunosuppressive drugs and fertility
In transplanted male patients, there is a dose-dependent
decrease in plasma concentrations of testosterone, an in-
crease of gonadotrophins and an alteration of spermato-
genesis compared to the values of the general population.
These gonadal alterations however are less consider-
able than before the organ transplantation [7–9], as
in women [10, 11].

Immunosuppressive therapies and pregnancy
The efficacy of immuno-suppressive drugs in the treat-
ment of autoimmune diseases or transplants, the emer-
gence of new drugs, and a better knowledge of their side
effects, now make pregnancy possible where it was

contraindicated several years ago either because of the
teratogenic effect of the drugs or because of the under-
lying maternal condition. Steroids have been involved in
increased risk of premature membrane rupture, and ciclos-
porine in increased prematurity rates but the increased
risk of preterm birth in transplant recipient women is also
related to the maternal condition and not to immunosup-
pression. The risk of gestational diabetes and hypertension,
however, is amplified by immunosuppressive agents, par-
ticularly steroids and tacrolimus. Also, the risk of pre-
eclampsia is increased by creatininemia greater than
13 mg/L before pregnancy and/or the use of anticaci-
neurins. In addition, immunosuppressive drugs pose a
substantial maternal-foetal infectious risk (bacterial or
opportunistic infections especially cytomegalovirus or
BK reactivation). The risk of foetal transmission of hepa-
titis viruses, C in particular, is approximately 5 % and de-
pends on the mother’s viral load after liver transplantation.
Moreover, the risk of organ rejection or auto-immune/in-
flammatory disease reactivation related to the adjustment
of immunosuppressive drugs before or during pregnancy
is about 2 to 5 % [4]. Finally, breastfeeding should take into
account the passage of potential toxic metabolites into the
milk and therefore the neonate [12].
Most immunosuppressive agents cross the placental

barrier; some are permitted during pregnancy, others are
formally contraindicated due to the risk of foetal malfor-
mations. The risks of each of these therapeutic classes
are detailed in next section.

Diagnosis
The consequences of immunosuppressive drugs may be
anticipated or discovered at any time of a pregnancy in a
patient or even years after the birth in the child. The
diagnosis of these complications is made upon the med-
ical history (transplantation, rheumatoid arthritis, lupus,
bowel inflammatory diseases, multiple sclerosis…) and
the analysis of the previous treatment courses both in
mother and father before and during the pregnancy.
Diagnosis might be clinical by examination of a new-
born. Blood renal, haematological, infectious, hormonal
and immunologic investigations may me needed. Ultra-
sound examination is essential especially during preg-
nancy to screen for teratogenicity

Differential diagnosis
Immunosuppressive drugs are prescribed for a severe
disease and this underlying disease might be responsible
by itself for the anomaly. The genetic background may
also interfere. Recording all the cases of anomalies re-
corded with a given treatment may help to understand
the mechanisms and avoid further occurrence.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 2 of 15

Aetiologies or effects of the different classes of
immunosuppressive agents on fertility and
pregnancy
The different drugs are grouped according to their deleteri-
ous effects on gametogenesis and on the hypothalamic–pi-
tuitary–gonadal axis, followed by potential teratogenic
effects of each drug (Table 1).

Contraindicated drugs when pregnancy is desired (Table 1)
Methotrexate
Study results differ regarding the deleterious effect of
methotrexate on spermatogenesis. If real, this effect seems
to be reversible after 3 months of treatment discontinu-
ation. Due to the mutagenic risk, men are advised to wait
3 months after stopping treatment to conceive. There is
no evidence of a teratogenic effect [13].
The repercussions of methotrexate treatment on female

fertility appear to be slight and may even be nonexistent.
Serum concentrations of the anti-Müllerian hormone
(AMH) were not lower in women treated with methotrex-
ate for rheumatoid arthritis than in controls [14]. The
evaluation was done however 6 months after the start of
treatment, and the pregnancy rates subsequently obtained
were not reported. A poorer response was observed to
ovarian stimulation in the 18 months following metho-
trexate treatment, though it improved thereafter [15].
In contrast, the folic acid antagonist methotrexate has

been documented to be teratogenic if administered dur-
ing the first trimester.of pregnancy, even at doses lower
than 30 mg/week. Over 30 cases of foetal malformation
involving the central nervous system and the limbs were
reported in association with IUGR and failure to thrive,
etc. [16, 17]. The embryolethal effect of methotrexate is
otherwise used in the medical treatment of ectopic preg-
nancies [16]. The miscarriage rate on treatment is ap-
proximately 40 %, considerably higher than that seen in
the general population or in those with autoimmune dis-
eases [18]. During the second and third trimester, metho-
trexate administration is unrelated to a teratogenic effect
but could increase the risk of IUGR and low birth weight.
Administration apart from conception does not increase
the risk of malformations or miscarriage [18]. However a
3-month treatment-free interval between discontinuation
of methotrexate and conception is recommended.

Mycophenolate (purine synthesis inhibitor)
Mycophenolate, being non-diabetogenic, is one of the
most commonly used immunosuppressive drugs in
transplantation.
There is no data on the effects of mycophenolate on

male fertility. The 205 pregnancies involving 152 trans-
planted fathers who had been treated with mycophenolate
were associated with a similar risk of prematurity (10 %)
and malformations (3 %) as in the general population [19].

The AMH levels of female patients treated with myco-
phenolate for lupus were not lower than in a control
population [20]. A very considerable amount of myco-
phenolate crosses the placental barrier. In rats, there is
a teratogenic and mutagenic effect. In women, mycopheno-
late poses an increased risk of miscarriage (32 % to 45 %)
and multiple craniofacial congenital malformations (MMF-
associated embryopathy (EMFO tetrada: Ear, Mouth,
Fingers, Ocular/Organ malformation) in 26 % of cases after
first trimester exposure to MMF according to The
European Network of Teratology Information Services
[21]. Foetal toxicity is present throughout the first trimester
and seems to be cumulative depending on the clinical cases
reported. For this reason, treatment must imperatively be
modified in the event that pregnancy occurs, and the pre-
scription of mycophenolate should be avoided in young,
transplanted women with a potential desire to become preg-
nant. There are few data on the exposure to mycopheno-
late during the second part of pregnancy, but the treatment
could result in blood count abnormalities in exposed new-
borns. The U.S. Food and Drug Administration (FDA)
launched a systematic information programme for pa-
tients on the teratogenic risk including the issuance of a
written information document, signature of informed con-
sent before the prescription, and incentive to participate
in a registry of pregnancies occurring on mycophenolate
or within 6 weeks of its discontinuation.

Leflunomide and teriflunomide
In animal models, teriflunomide, an inhibitor of de novo
synthesis of pyrimidine, active metabolite of leflunomide
does not have adverse effects on male or female fertility,
but both drugs are embryotoxic and teratogenic, promot-
ing the development of malformations of the axial skel-
eton and head (microphthalmia, hydrocephaly). There is
little data from pregnant women, but cases of complex
morphological-functional abnormalities (congenital blind-
ness and perceptive deafness) were reported in children
with parents exposed to leflunomide in the pre-conception
period or during pregnancy [22, 23]. These drugs are there-
fore contraindicated in the pre-conception period (the 3½
months before pregnancy) and during pregnancy. A
washout procedure may be proposed with cholestyramine
(8 g, 3 times daily) or activated charcoal (50 g, 4 times
daily) for 10 days in order to shorten the time period
needed between discontinuation of the treatment and
conception, since the total elimination of the drug may
take 8 to 24 months. A sperm cryopreservation is recom-
mended before treatment.

Cyclophosphamide
Cyclophosphamide, a cytotoxic alkylating agent widely
used in allogeneic bone marrow transplantation, per-
manently alters the ovarian reserve in a manner that is

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 3 of 15

Table 1 Consequences of the main immunosuppressor on fertility and pregnancy

Immunosuppressants Hypothalamic–pituitary–
gonadal axis

Gametogenesis Mutagenesis Teratogenesis Pregnancy NN Management

A: Contraindicated drugs when pregnancy is desired

Methotrexate M: alteration debated,
reversible after stopping for 3
months

M: mutagenic F: teratogenic
without dose
effect, especially
between 6 and 8
weeks

Increased frequency
MC 23%

CI during pregnancy,
especially during the
first trimester.

W: No effect: AMH the same
at 6 months between treated
or non-treated population
but less response to ovarian
stimulation the first 18
months post-MTX

>30 cases of
malformation of
the CNS, skull,
limbs, IUGR,
cardiopathy

Discontinuate at least 3
months before
pregnancy in both
genders

Mycophenolate In rats: no effect on fertility mutagenic in vivo in
rats

W: clasto-carcino-
teratogenic:
multiple
craniofacial
anomalie,…

crosses placenta +++ NN heamato
monitoring if data at
2nd or 3rd trimester

Switch to another drug
before pregnancy

M: No effect increased risk of MC

Le- and teri-
flunomide inhibitor
of de novo synthesis
of pyrimidine

Total elimination of the
drug may take 8 to 24
months.

No adverse effect on male or
female, even in animals at
high doses

neither mutagenic
nor clastogenic

Teratogenic in
animals: head
malformations

insufficient human
data

one case of congenital
blindness

Stop ≥ 3.5 months
before conception or
Wash-out with
cholestyramine (8gx3/
day) or charcoal
(50gx4/day) – 10 days
to obtain
concentration< 0.02 mg/L

no studies in
humans

Sperm
cryopreservation
recommended before
treatment in men

Cyclophosphamide
cytotoxic alkylating
agent

W: FSH/LH increased,
even with short
exposures

Lasting alteration of ovarian
reserve that is dose-,
duration- and age-
dependent: low AMH

mutagenic embryolethal and
teratogenic
without dose
effect, especially if
early exposure:
limbs, dysmorphia,
eye,

CI during pregnancy
and breastfeeding
IUGR

more late exposure,
more significant risk
NN haemato

Effective contraception
to be continued until
end of treatment

Wait for one ovulation
cycle after
discontinuation before
conception

Lero
y
et

a
l.
O
rp
h
a
n
et

Jo
u
rn
a
l
o
f
R
a
re

D
isea

ses
(2

0
1

5
) 1

0
:1

3
6

P
ag

e
4
o
f
1
5

Table 1 Consequences of the main immunosuppressor on fertility and pregnancy (Continued)

Mitoxantrone anomalies of the
menstrual cycle or even
permanent amenorrhea
in 7 to 14% of treated
patients in correlation
with the cumulative
dose and the age of
exposure

deleterious effect on
spermatozoïds and ovocytes
leading to fertility alterations.
In association with other anti-
cancer drugs,

aneuploidism and
azoospermia
spontaneously
improved after 3 to 5
months of treatment
discontinuation

teratogenic in
animals and
humans

Contraindicated in
pregnancy .

A period of 6 months
is required after
treatment before
conception

Sperm
cryopreservation
recommended before
treatment in men and
contraception is
required in women.

Thalidomide teratogenic in
humans

B: Drugs to be used with caution if needed

mTOR inhibitors M: inhibitor M: oligoasthenosper mia,
reversible if stopped
(debated)

No mutagenic effect
in vitro/in vivo

embryo- and
foetotoxic in
animals

−16 reported
pregnancies

Substitute with other
drug and continue
contraception for 12
weeks after stopping
treatment

– sirolimus Testicular atrophy −3 MC and one child
with multiple
malformations
(+MMF)

No information on
breast feeding

– everolimus W: menstrual disorders 1st

year of graft, and 50-60%
ovarian cysts, diminishing
upon stopping or taking OP
in 80% of cases

temsirolimus

anti-TNFα M: no effect M: no effect non-mutagenic W: non-teratogenic
in animals and in
about 50
pregnancies

1st trimester: Few data
etanercept/
certolizumab ≠ inflixi/
adali:mumab NTR

Crosses placental
barrier ± (

infliximab

until 6 months in NN),
less for etanercept,
certolizumab

Not recommended to
discontinue therapy if
desire for pregnancy.

Anti-cytokine W: no studies FDA: increased risk
cardiac
malformations –
low level of proof

2nd3rd trim: 20 cases:
inflixim- adalim-
certoliz- umab-: NTR

CI live vaccines

etanercept

infliximab

adalimumab

certolizumab One child of mother
treated entire
pregnancy, died from
BCG (TB vaccine)
complications

Infections without
fever

golimumab

Lero
y
et
a
l.
O
rp
h
a
n
et
Jo
u
rn
a
l
o
f
R
a
re
D
isea
ses
(2
0
1
5
) 1
0
:1
3
6

P
ag

e
5
o
f
1
5

Table 1 Consequences of the main immunosuppressor on fertility and pregnancy (Continued)

C: Authorised drugs

Anti-inflammatory M and W: synthetics and
high dose inhibit
hypothalamic–pituitary–
gonadal axis

promotes apoptosis of germ
cells

not more cleft lip-
palate

IUGR, premature,
eclampsia >

– case of NN ACI after
high doses

Possible breast feeding

glucocorticoids W: In practice, no major
impact on fertility

actual role of GC or
underlying disease?

– predisposes to
unfavourable adult
metabolic profile

Anti-inflammatory M: oligoasthenoteratospermia
reversible after 2 months of
stopping; 60% infertility. W:
no effect

no effect increased prematurity No information on
breast feeding

Prescribe folates

Sulfasalazine
Mesalazine

Prefer Mesalazine

Anti-metabolites M and W: no effect few deleterious effects mutagenic in vivo /in
vitro

M and W: Non-
teratogenic

increased prematurity Breastfeeding CI ±
failure to thrive +
reversible
haematological NN risk

– Avoid use in a male
patient wishing a
conception

Azathioprine carcinogenic and
teratogenic in animals

Mutagenic
– Discontinuation 3
months before
conception

Prodrug of 6
mercaptopurine

– US survey of
pregnancy if
conception by a
treated father

– Sperm
cryopreservation
recommended

– Use possible
regardless of
pregnancy term.

– Possible breast
feeding

Beta interferon increase of LH Alterations of ovarian reserve

high molecular
weight and should
not cross the
placenta.

no teratogenic
effect

numerous reported
pregnancies with
either the father or
the mother treated :
no problem

lower birth weight and
higher rate of
spontaneous abortions,
in the treated mother,
even if the treatment
is stopped as soon as
the pregnancy is
known

– Discontinuation not
necessary in case of
pregnancyNo sperm alterations

– No long term
deleterious effects
have been reported in
the offspring

Glatiramer acetate increase of LH Decrease of ovarian surface
and number of antral follicles

high molecular
weight and should
not cross the
placenta.
no teratogenic
effect
numerous reported
pregnancies with
either the father or
the mother treated :
no problem

– Discontinuation not
necessary in case of
pregnancy

– No long term
deleterious effects
reported in the
offspring

Lero
y
et
a
l.
O
rp
h
a
n
et
Jo
u
rn
a
l
o
f
R
a
re
D
isea
ses
(2
0
1
5
) 1
0
:1
3
6

P
ag

e
6
o
f
1
5

Table 1 Consequences of the main immunosuppressor on fertility and pregnancy (Continued)

Chloroquine – No deleterious effects
on pregnancy or
breastfeeding

Calcineurin inhibitors W: no effect M: asthenoteratospermia if
dose > 2mg/kg/day

W: non-
teratogenic, limited
crossing to
placenta (5-20%)

increased infectious
risk (CMV)premature /
IUGR

passes into breast milk,
undetectable in
newborns

Use possible during
pregnancy; More
experience with
cyclosporine

Ciclosporine

Tacrolimus

non-teratogenic; 2
cases of congenital
malformations

30% premature/ low-
birth weight

no renal repercussions
in NN

Caution regarding
infections

Reversible involvement
of lymphocytes,
without clinical
repercussions.

Possible breast feeding

rare cases of transient
kidney failure in NN

D: Drugs with limited experience (biological therapies, inductors)

Tocilizumab Anti-IL-6
monoclonal Ab

poorly understood
effects on fertility
clearance: 2 weeks

Large molecule with limited
crossing to placenta and
breast milk

animal studies: no
lethal effect.

poorly understood
effects on both
fertility and
pregnancy

Very few pregnancies
reported.

Avoid pregnancy-
Limited experience-
Continue
contraception 3
months after stopping
(FDA).

Rituximab anti-CD20
monoclonal Ab, B
lymphocyte inhibitor

long half-life. Very limited transplacental
crossing (debated)

150 pregnancies,
some with early
exposure: NTR

Prematurity 15%
Haematological
anomalies, sometimes
infections up to 6–12
months after stopping

Avoid pregnancy less
than 12 months after
stopping

Abatacept (anti-
CD28 Ab or CTLA-4
fusion protein,
inhibiting the co-
stimulation pathway
of the T
lymphocytes (anti-T)

clearance: 2 ½ months
(half-life = 14 days)

-non-altered in animals large molecule: non-teratogenic in
animals

Several pregnancies
with early exposure:
NTR

Avoid
pregnancyLimited
experience- W: no studies

Anankira anti-IL-1
receptor

little teratogenicity
in the 1st trimester
since limited
crossing of
placenta ± end of
the 2nd trimester
when its crossing
increases.

<5 pregnancies reported: NTR

Administration in 3erd

trimester with
increased risk of NN
immunosuppression,
and CI live vaccines for
at least the first 6
months following
birth.

Avoid
pregnancyLimited
experience

Dimethyl fumarate inhibitor of immune
cells and an anti-oxidant

consequences on fertility not
known.

consequences on
pregnancy not
known.

Lero
y
et
a
l.
O
rp
h
a
n
et
Jo
u
rn
a
l
o
f
R
a
re
D
isea
ses
(2
0
1
5
) 1
0
:1
3
6

P
ag

e
7
o
f
1
5

Table 1 Consequences of the main immunosuppressor on fertility and pregnancy (Continued)

Natalizumab Fertility alterations in female
but not male animals

No teratogenic
effects – effects not
fully known yet

Discontinuation of
treatment
recommended 2
months before
conception in men.

Fingolimod or FTY
720 or Sphingosine
1-phosphate
receptor modulator

Effects on fertility not well
known, − Would have a
protective effect on ovarian
function at least in vitro

teratogenic effects,
in animals

Recommended to stop
the treatment 2
months before the
conception in men
and women.

Induction Unknown effects on
both fertility and
pregnancy

Avoid pregnancy
within 12 months of
stopping- anti-human

thymocyte Ig

– IL-2 receptor
inhibitors
daclizumab

– belatacept fusion
protein (Fc fragment
of human IgG1
+extracellular CTLA-4

Note the significant impact of cyclophosphamide on fertility If emergency use needed, start the treatment if possible after the 1st trimester
The website of the French Teratogenic Agent Information Centre [Centre de Référence sur les Agents Tératogènes (CRAT)] (http://www.lecrat.org/) can provide more information
Ab antibodies, CI contraindicated, MC miscarriage, FDA Food and Drug administration, W women, M men, HAS French National Authority for Health [Haute Autorité de Santé], ACI adrenocortical insufficiency, Ig
immunoglobulin, IL-2 interleukin-2, MMF mycophenolate, MTX methotrexate, NN neonatal, OP oestrogen-progestin contraceptive pills, NTR nothing to report, IUGR intrauterine growth restriction), US United States
teratogen substance that causes malformations in the foetus when administered to the mother, mutagen substance that increases the number of mutations in the genome, mutations that are likely to promote
malformations or an increased carcinogenesis risk, clastogen substance likely to induce chromosomal breaks and thus aberrations

Lero
y
et
a
l.
O
rp
h
a
n
et
Jo
u
rn
a
l
o
f
R
a
re
D
isea
ses
(2
0
1
5
) 1
0
:1
3
6

P
ag

e
8
o
f
1
5

dose-, duration- and age-dependent. This has been well
demonstrated through decreased plasma concentrations
of AMH and increases in gonadotrophins, with some
variations correlated to the cumulative dose of cyclo-
phosphamide, even for short exposures [19, 24].
Cyclophosphamide in animals is mutagenic, terato-

genic and embryolethal. In humans, it is a confirmed
teratogenic agent that promotes IUGR, malformation
of the extremities and the head (ocular involvement,
facial dysmorphia, craniostenosis, hydro- or micro-
cephaly). The reported cases are rare so that it is diffi-
cult to establish a dose–response relationship; some
malformations have been observed at low doses, while
newborns that had had high dosage exposures were
born healthy. The malformations seem to be secondary
to exposure during the 1st trimester. However, the
haematological side effects observed in the neonatal
period are all the more frequent the later the exposure
during the 2nd and 3rd trimesters of pregnancy. The
teratogenicity of cyclophosphamide absolutely contrain-
dicates its use in pregnancy. If its use is essential (e.g.,
breast cancer diagnosed during pregnancy), the treat-
ment will be prescribed after the 1st trimester if
possible. Conception is theoretically possible about two
days after the end of the treatment. In practice, effective
contraception must be continued until the end of treat-
ment, and a minimum of one ovulation cycle after the
end of treatment is recommended before considering
conception.

Mitoxantrone
Mitoxantrone (Table 1), sometimes used in multiple scler-
osis, also a chimiotherapy in leukemia and prostate cancer,
induces anomalies of the menstrual cycle or even perman-
ent amenorrhea in 7 to 14 % of treated patients The effects
are correlated with the cumulative dose and the age of ex-
posure [25, 26]. Mitoxantrone has a deleterious effect on
spermatozoïds and ovocytes leading to fertility alter-
ations. In association with other anti-cancer drugs,
mitoxanthrone may be responsible for aneuploidism
and azoospermia spontaneously improved after 3 to
5 months of treatment discontinuation [27, 28]. This
drug is teratogenic in animals and humans and is there-
fore contraindicated in pregnancy. Sperm cryopreservation
is recommended before treatment initiation in men and
contraception is recommended as well in women [29]. A
period of 6 months is required after treatment discontinu-
ation before conception.
In conclusion, these 5 immunosuppressive agents are

teratogens, and other immunosuppressive agents must
be used when pregnancy is desired. In addition, cyclo-
phosphamide alters the ovarian reserve, thus warranting
a consultation in a specialised centre for the purpose of
ovarian preservation before prescription of the drug.

Drugs to be used with caution (Table 1)
Anti-TNF-α (tumour necrosis factor alpha) (anti-cytokine)
The most commonly used anti-TNF alpha drugs are
first, etanercept, a TNF inhibitor acting as a soluble re-
ceptor, with a half-life of 72 h; and second, the anti-TNF
antibodies (infliximab, adalimumab, certolizumab, goli-
mumab), with a half-life up to 14 days.
Various andrological studies in men are not supportive

of spermatic alteration secondary to treatment with
TNF-alpha inhibitors. Therefore there are no recom-
mendations to stop treatment if conception is desired
[30]. Compared to case controls in spondyloarthritis, the
levels of inhibin B, testosterone and gonadotrophins are
unchanged by these drugs [30, 31], particularly etaner-
cept and adalimumab.
There have been no studies in women on the effects of

anti-TNF alpha agents on the hypothalamic–pituitary–
gonadal axis and the ovarian reserve.
Anti-TNF alpha agents are not teratogenic in animals,

nor mutagenic in pre-clinical tests.
These drugs cross the placenta − infliximab in particu-

lar, which has been detected up to 6 months after child-
birth in the child’s blood [32]. There are more reported
cases of exposure to infliximab and adalimumab in the
first trimester of pregnancy than to etanercept or certoli-
zumab. The reported cases of use of these drugs in the
second part of pregnancy are extremely rare. The data
however are reassuring and do not show malformations
in children born from these pregnancies [33]. A risk of
VACTERL association (anomalies of the vertebrae and/
or limbs, anal atresia, tracheoesophageal fistula, renal
malformation) and heart defects have been noted by the
Food and Drug Administration (FDA), but this report
has methodological biases [34].
The frequency of infections however, including reacti-

vation of tuberculosis in the mother, is higher, particularly
with certozilumab [35]. A case of severe tuberculosis that
resulted in death was reported after BCG vaccination in
one child.
In practical terms, considering the still limited experi-

ence available as to the use of these drugs, it is recom-
mended that contraception be continued in women
receiving TNF-alpha inhibitors, prolonged for 3 weeks
after stopping etanercept and 3 months after stopping
anti-TNF-alpha antibodies despite the lack of demon-
stration of teratogenic effects. In addition, infections may
not be accompanied by fever in patients using anti-TNF
alpha drugs. If the use of anti-TNF alpha treatment is
necessary, it is recommended that the last injection be
given at the start of the 3rd trimester of pregnancy due to
the long half-life of elimination. At birth, the child is
immunocompromised, and this may continue up until
6 months following the last maternal injection, making
the postponement of live vaccinations mandatory

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 9 of 15

(2013 HAS [French National Authority for Health]
Recommendations).

mTOR inhibitors: sirolimus, everolimus, temsirolimus
In addition to their immunosuppressive properties, the
mTOR inhibitors are also used as anti-tumour agents,
notably in pancreatic neuroendocrine tumours.
Sirolimus alters the functioning of the hypothalamic–

pituitary–gonadal axis [36, 37]. As a result, men with
kidney transplants who are treated with sirolimus have
lower serum testosterone levels and higher plasma con-
centrations of gonadotrophins than on other immuno-
suppressive drugs. These values nevertheless remain
within normal limits. These variations are positively cor-
related to the dose of sirolimus and are independent of
other variables, such as graft function, age of the patient,
body mass index, duration of dialysis, existence of post-
transplantation diabetes and use of steroids [38].
Similarly, high levels of everolimus (combined with
corticosteroids and calcineurin inhibitors) are the
cause of poorer recovery of testosterone, FSH and LH
levels after transplantation [39]. Sirolimus qualitatively
and quantitatively alters spermatogenesis in men, as
observed in 132 heart transplant patients [40–42].
This adverse effect is purportedly reversible upon
stopping the treatment [41, 43], although this revers-
ibility is controversial [42]. A testicular histological
study done in a patient treated with sirolimus who
needed surgical intervention for leydigioma showed
testicular atrophy and vacuolation of the seminiferous
epithelium [44], in agreement with the animal studies.
Young women treated with sirolimus and tacrolimus

have an increased risk of oligospanio- or amenorrhoea)
and ovarian cysts (50 to 62 % of cases) in the first year
of the transplant [45–47]. These cysts diminish in 80 %
of cases when sirolimus is stopped or with the use of
combined oestrogen plus progestin contraceptive pills.
Some cysts, such as mucinous cystadenomas or haemor-
rhagic cysts, require surgical cystectomy [47]. Their oc-
currence is allegedly promoted by insulin resistance
induced by immunosuppressive agents, but this hypoth-
esis has not been demonstrated.
In animal models (rats), treatment with tacrolimus, sir-

olimus or a combination of both lead to a decrease in
the ovarian surface, the number of ovulation cycles,
uterine size, and aromatase expression, although with
the corpora lutea present. Conversely, sirolimus treat-
ment allegedly protects the ovarian reserve and prevents
the transition of pre-antral follicles to antral follicles by
modulating the mTOR and signalling pathways [48].
In animals, sirolimus does not have an in vitro or in

vivo mutagenic effect, but it is embryo- and foetotoxic. In
the clinical setting, sixteen pregnancies were reported be-
tween 2004 and 2012 while using sirolimus or everolimus

[49]; of these, 3 resulted in miscarriage and one child had
many malformations but had received mycophenolate in
early pregnancy. Currently, experience with the use of this
treatment during pregnancy is insufficient. Even though the
published data seem to be reassuring, it is recommended
that another drug be used as a substitute for mTOR inhibi-
tors, and that contraception be continued for 12 weeks after
stopping treatment due to the clearance time.
Finally, these 2 therapeutic classes have little effect on

fertility and pregnancy, but the available experience of
use is still limited (about 10 years), and as a result their
prescription must be avoided when possible.

Treatments associated with a low risk of deleterious effects
on fertility and pregnancy (Table 1)
Glucocorticoids
The use of glucocorticoids has consequences on fertility
[50]. Therefore, synthetic (and even endogenous) corti-
costeroids have an inhibitor effect on the male hypothal-
amic–pituitary–gonadal axis: a) through direct and
indirect action on the level of transcription of the GnRH
receptor gene on the pituitary cells; b) by modulating
the expression of the LH receptor genes or of testoster-
one biosynthesis enzymes, whether through genomic ef-
fects or not; and c) through inhibition of the adrenal
synthesis of androgens. In the ovaries, low doses of corti-
costeroids play a positive role in steroidogenesis, the
maturation of oocytes, ovulation and maintenance of the
corpus luteum. However high-dose exogenous intake is
often deleterious.
In vitro, glucocorticoids exert a proapoptotic effect on

the germ cells of rat testes and ovaries of the female foetus
[51]. In practical terms however, there are no major conse-
quences on the fertilisation capacities in women receiving
corticosteroid treatment [52].
The incidence of IUGR, low-birth weight, pre-eclampsia,

hypertension, diabetes and prematurity is increased in
cases of exposure to corticosteroids during pregnancy
compared to the general population, regardless of the dos-
age and the type of corticosteroid use [53–55]. In addition,
high doses of glucocorticoids during intrauterine life risk
may cause neonatal adrenal insufficiency, but also dys-
function of the hypothalamic–pituitary–adrenal axis and
foetal “programming”, thus predisposing to an unfavour-
able metabolic profile as an adult [56].

Sulfasalazine
The use of sulfasalazine is the cause of oligo-astheno-
teratospermia and infertility in over 60 % of treated
men [57], although it is reversible after two months after
stopping treatment. However, the hypothalamic–pituit-
ary–gonadal axis changes little, as the plasma concentra-
tions of prolactin, testosterone and gonadotrophins are
normal [58]. In women, no effects of this treatment have

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 10 of 15

been reported on fertility parameters. There has been no
demonstration of congenital foetal anomalies on this ther-
apy [59], but the prescription of folate supplementation is
advised due to its mechanism of action.

Mesalazine
The hypothalamic–pituitary–gonadal axis and the sperm-
atic parameters are not altered with treatment using this
drug [60]. In fact, replacement of sulfasalazine by mesala-
zine improves the spermatic parameters [58]. There has
been no demonstration of congenital foetal anomalies
or maternal-foetal complications other than prematur-
ity, which was slightly increased with mesalazine [61].

Azathioprine (purine analog, precursor of 6- mercaptopurine)
Azathioprine is mutagenic. The rare andrological studies
are not supportive of alterations of fertility [62]. More
than 1000 pregnancies issued from men treated with
azathioprine or 6-mercaptopurine have been published
and no teratogenic effects have been reported. Neverthe-
less, chromosomal anomalies have been described on
spermatozoids of male subjects during the treatment
and the year following its discontinuation.
There is little published data in women, but azathio-

prine does not seem to have detrimental effects on fertility.
The AMH concentrations of patients treated with azathio-
prine, mycophenolate or calcineurin inhibitors for lupus
were not lower than in the control population [19].
Azathioprine is teratogenic in animals, as treated

pregnant females and their foetuses have presented
with chromosomal abnormalities on the circulating
lymphocytes.
Azathioprine is authorised during pregnancy. Although

it crosses the placental barrier, this agent cannot be con-
verted to an active metabolite, as the foetal liver lacks
the specific enzyme. No teratogenic risk has been dem-
onstrated in the many data, both in women and in men
[61, 63–65]. In contrast, prematurity [61], some cases of
immunosuppression that quickly resolve [66] and/or re-
versible involvement of the blood cell lineages have been
observed in newborns. Otherwise, breastfeeding is not
contraindicated, since the active metabolite, 6 mercapto-
purine, rarely passes, and if passes, remains at very low
level, into breast milk [67].
If azathioprine is required for control of the inflamma-

tory disease or the graft, it may be used during preg-
nancy, but an ultrasound monitoring should be proposed.
Amniocentesis may be discussed but can only discard
usual caryotypic alterations. A sperm cryopreservation is
recommended before instauration of the treatment in a
male subject because of the mutagenic effect. If a treated
male subject wishes a conception, the treatment should
be stopped if possible at least 3 months before conception
(a cycle of spermatogenesis).

Ciclosporine (calcineurin inhibitor)
No significant difference in the hormonal parameters
(FSH, LH, testosterone and prolactin) were observed be-
tween kidney transplants treated with ciclosporine and
those treated with tacrolimus [68]. However, rats treated
with ciclosporine had irreversible testicular damage [7].
In men, doses greater than 2 mg/kg/day [9] have been
implicated in asthenoteratospermia. No significant differ-
ence was seen in men receiving lower doses compared
to the control group. Therefore it is not recommended
that treatment be discontinued if pregnancy is desired.
Plasma concentrations of AMH in patients treated with
calcineurin inhibitors for lupus were not lower than in a
control population [20].
Limited amounts of ciclosporine cross the placenta

(5–20 %), and it does not result in teratogenic effects [69].
No renal repercussions attributable to ciclosporine were

observed during the follow-up of several hundred children
exposed in utero. Reversible involvement of B or T lympho-
cytes without clinical consequences, prematurity, and
IUGR have sometimes been reported. These neonatal ef-
fects may be attributed to ciclosporine but also to the dis-
ease and/or associated treatments.

Tacrolimus (calcineurin inhibitor)
There are a large number of published data on pregnant
women exposed to tacrolimus, and they show no evi-
dence of an increase in malformations [70]. The experi-
ence acquired over 13 years [71] in 37 women treated
with tacrolimus after liver transplantation and who gave
birth to 49 children resulted in:

– 30 % premature births [72]
– 32 % birth weight below the 50th percentile
– 5 to 6 % congenital malformations [70]
– rare cases of transient hyperkalaemia and kidney

failure in the child

Beta interferon and glatiramer acetate
Studies are scarce but significant alterations of ovarian
reserve have been reported both with beta interferon and
glatiramer acetate used in the treatment of multilocular
sclerosis [73]. These 2 molecules have a high molecular
weight and should not cross the placenta. Neither sperm-
atic alterations nor teratogenic effects have been observed
in the numerous reported pregnancies with either the
father or the mother treated with these 2 molecules
[74, 75]. Interferon is however associated with a lower
birth weight and a higher rate of spontaneous abortions, in
the treated mother, even if the treatment is stopped as soon
as the pregnancy is known [76, 77]. The discontinuation of
these treatments 3 months before conception is recom-
mended each time the disease is not a frequent relapsing
form. In other cases, the treatment might be stopped as

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 11 of 15

soon as the conception is proven. If necessary, the treat-
ment might be pursued during the whole pregnancy. No
long-term deleterious effects have been reported in the off-
spring [78–81].

Chloroquine
Chloroquine appears to have a deleterious effect on sperm
quality in vitro and in vivo in animal studies. Little data are
available in males, unlike in pregnant women in which
there is a large amount of reassuring data. The teratogenic
risk related to the use of hydroxychloroquine is very
low [82].
Chloroquine may be used at prophylactic doses for

malaria. In other situations, if continuation of the ther-
apy is required for good control of the treated disease, the
lowest possible dose should be used during pregnancy.
In conclusion, if these drugs are required for control

of the inflammatory disease or maintenance of the graft,
they may be used during pregnancy.

Recent drugs in which the effects are not fully known
(Table 1)
Biological therapies
Biological therapies (tocilizumab, rituximab, abatacept, ana-
nkira) seem to have few teratogenic effects in the first tri-
mester, since a very limited amount cross the placental
barrier. This is in contrast to the end of the 2nd trimester,
in which crossing is increased [83–85]. If administered in
the 3rd trimester, some of these drugs carry an increased
risk of neonatal immunosuppression, and their use contra-
indicates the use of live vaccines for at least the first
6 months after birth. Abatacept and anankira appear to be
associated to a lesser extent with side effects than the
anti-TNF alpha agents, infliximab in particular [34, 86].
Very few pregnancies have been reported. Due to this

scarcity of experience, the prescription of biological ther-
apies is discouraged if pregnancy is desired, both in women
and men due to the still poorly understood long-term side
effects [87]. Continued use of contraception is advised
for 3 months following discontinuation of these treat-
ments due to their long half-life.

Immunomodulators in multiple sclerosis
Besides beta interferon and glatiramer acetate use
(Table 1), and mitoxantrone (Table 1), dimethyl fu-
marate acts as an inhibitor of immune cells and an
anti-oxidant. Its consequences on fertility and preg-
nancy are not known.
Natalizumab alters fertility in female, but not male,

animals. No teratogenic effects have been observed in
women but the effects are not fully known yet [88, 89]. The
discontinuation of treatment is recommended 2 months
before conception in men.

The effects of fingolimod on fertility are not well known,
but fingolimod seems to have a protective effect on ovar-
ian function at least in vitro despite its teratogenic effects
in animals [90, 91]. Therefore, it is recommended to stop
the treatment 2 months before the conception in men as
well as in women [92].
Except for beta interferon and glatiramer acetate, which

might be prescribed if necessary, the other immunomodu-
lators used in multiple sclerosis should be stopped two
(natalizumab, fingolimod) to six months (mitoxanthrone)
before conception, with a possible wash-out procedure for
teriflunomide.

Induction immunosuppressive treatments
The effects of induction treatments (rabbit anti-human
thymocyte immunoglobulins; interleukin-2 receptor an-
tagonists such as daclizumab; belatacept) on fertility and
pregnancy have not been studied.

Management of patients using
immunosuppressive drugs
The management prior to the patient starting treatment
with an immunosuppressive agent, or when the patient is
already receiving this immunosuppressor treatment is sum-
marized in Tables 1 and 2 [93–95].
Pregnancy, when an option, must be planned under

conditions of strict monitoring, when the disease is not
in an acute phase. These findings prompt the recom-
mendation to delay pregnancy up to l to 2 years after
the transplant, when the immunosuppressive doses will
be lower and the menstrual cycles will have spontan-
eously re-established [14].
Most immunosuppressants cross the placental barrier

and, depending on the foetal malformation risk, some
are authorised during pregnancy (azathioprine, corti-
costeroids, anticalcineurins, ß interferon, glatiramer
acetate, chloroquine), and others are formally contra-
indicated (methotrexate, mycophenolate, le- and teri-
flunomide, cyclophosphamide, mitoxantrone). If the
latter treatments (cyclophosphamide in particular) need to
be prescribed for emergency purposes, it is preferable that
they be started after the 1st trimester of pregnancy. For
some drugs that have fewer data but seem reassuring, the
use may be continued during the pregnancy if needed
(anti-TNF alpha, mTOR inhibitors).
The preconception time period for stopping varies with

each drug and depends on its clearance time (5 half-lives):
12 weeks for the m-TOR inhibitors; 4 weeks to 2 years for
le- and teri-flunomide (according to whether or not the
wash-out protocol with charcoal and cholestyramine is
used); 12 weeks in men and in women for methotrexate;
one ovulation cycle for cyclophosphamide; and 6 weeks
for mycophenolate.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 12 of 15

Prognosis and unresolved questions
In the 21st century, immunosuppressive drugs are indi-
cated in many different diseases due to their efficacy but
also due to a better understanding of their mechanisms of
action and their side effects. This efficacy has enabled im-
provement of underlying diseases, making parenthood an
option in some young patients who previously had defini-
tive life-threatening conditions. Nevertheless, these treat-
ments remain powerful therapies that require skilful
handling; therefore it should be borne in mind that
the long-term consequences are still not fully under-
stood. As an example, the marketing authorisation for
mTOR inhibitors only dates back 10–15 years. Although
childbirth is possible with these treatments, this undertak-
ing is never without risk to the mother and child. It must
be anticipated very far in advance, before the prescription
of the immunosuppressants, in order to preserve fertility,
and then before conception in order to adjust the immuno-
suppressive treatment. Close monitoring of the pregnancy
in conjunction with the prescriber of the immunosuppres-
sion and long-term follow-up of children from these
pregnancies is recommended. The potential conse-
quences of immunosuppressive drugs in children who
have received this exposure during pregnancy must still
be evaluated once they are adults. Recommendations
for use are established with a rather low level of proof,
which needs to be taken into account when informing
and monitoring patients. This multidisciplinary manage-
ment for planned parenthood must be discussed with the

couple so that they understand and accept the challenges
and obligations.

Abbreviations
Ab: Antibodies; CI: Contraindicated; MC: Miscarriage; FDA: Food and Drug
administration; HAS: French National Authority for Health [Haute Autorité de
Santé]; ACI: Adrenocortical insufficiency; Ig: Immunoglobulin; IL-2: Interleukin-
2; IUGR: Intrauterine growth restriction; MMF: Mycophenolate;
MTX: Methotrexate; NN: Neonatal; OP: Oestrogen-progestin
contraceptive pills; NTR: Nothing to report; W: Women; M: Men.

Competing interests
The authors declare that they have no competing interests.

Authors’ contributions
CL, JMR, ML, CD, SD, MCV analyzed the literature data; KLM, ASP, ACLG, IYA,
CN discussed the litterature according to their experience; CL and MCV
wrote the manuscript. All authors gave final approval of the manuscript.

Acknowledgement
The authors thank Prof J. Orgiazzi for his attentive expertise.

Author details
1Endocrinology and Metabolism, Hôpital Huriez, Lille University Hospital,
59037 Lille Cedex, France. 2Andrology, Hôpital Calmette, Lille University
Hospital, 59037 Lille Cedex, France. 3Gynaecology –Obstetrics, Hôpital Jeanne
de Flandres, Lille University Hospital, 59037 Lille Cedex, France. 4Endocrine
Gynaecology, Hôpital Jeanne de Flandres, Lille University Hospital, 59037 Lille
Cedex, France. 5Hematology, Hôpital Huriez, Lille University Hospital, 59037
Lille Cedex, France. 6Liver Diseases and Gastroenterology, Hôpital Huriez, Lille
University Hospital, 59037 Lille Cedex, France. 7Nephrology Hôpital Huriez,
Lille University Hospital, 59037 Lille Cedex, France. 8InsermU859 Biotherapies
of Diabetes, Lille University Hospital, 59037 Lille Cedex, France.

Received: 9 May 2015 Accepted: 30 August 2015

Table 2 Management of patients prior to starting or already taking immunosuppressive drugs

A: Management prior to starting immunosuppressive treatment

1. Patient information to clarify the consequences of the planned treatment on future fertility and the couple desire for subsequent fertility
(”Bioethics Law of 2004 and 2011 France (Art. L. 2141-)11)

2. Verify the absence of pregnancy.

3. Send to a fertility preservation centre, particularly if cyclophosphamide (see Table 3).

4. Effective contraception until desire for pregnancy, even in case of supposedly already altered ovarian reserve if the treatment is teratogenic and/or
pregnancy would be at risk due to the underlying disorder. The metabolic effects of calcineurins and mTOR inhibitors as well as glucocorticoids will
require the prescription of a progestin-only rather than a combined oestrogen-progestin contraceptive.

5. Screening for potential gynaecological neoplasias (cervical-vaginal smear, mammography) that could be worsened by immunosuppressive
treatment

B Pre-conception management of a parent treated with immunosuppressive agent

1. Pre-conception consultation to discuss stopping/changing immunosuppressive drug with referring team.

2. Preconception consultation with the couple: an explanation will be offered concerning the possible strategies and obligations in order to minimise
the maternal-foetal risks if pregnancy is possible; these include the need for a stable disease, multidisciplinary management, a possible period of
weaning from the treatment or immuno-suppression switch, the risk of organ rejection and/ or reactivation of an underlying disease, the risk of
transmitting a genetic disorder causing organ failure (Wilson’s disease, polycystic kidney disease, etc.).

3. The pregnancy will need to be planned, and the treatment will need to be modified if necessary in a pre-conception time period adapted to its
half-life (See Table 1, last column).

4. If pregnancy inadvertently occurs while taking immunosuppressive treatment, reassure the patient, test for known complications (ultrasound fetal
examination) and consider stopping or replacing the treatment according to the context.

5. During a planned pregnancy while on authorised treatment, the dosages of immunosuppressive agents should be closely monitored in order to
avoid overdose situations that are shared with the foetus, but also to avoid underdosing, which would compromise the control of the disease or the
function of the graft. During pregnancy there are variations in absorption and metabolism of the drugs.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 13 of 15

References
1. Alvaro E, Jimenez LC, Palomo I, Manrique A, Alegre C, Garcia M, et al.

Pregnancy and orthotopic liver transplantation. Transplant Proc.
2013;45:1966–8.

2. Hammoud GM, Almashhrawi AA, Ahmed KT, Rahman R, Ibdah JA. Liver
diseases in pregnancy: liver transplantation in pregnancy. World J
Gastroenterol. 2013;19:7647–51.

3. Blume C, Sensoy A, Gross MM, Guenter HH, Haller H, Manns MP, et al.
A comparison of the outcome of pregnancies after liver and kidney
transplantation. Transplantation. 2013;95:222–7.

4. Westbrook RH, Yeoman AD, Agarwal K, Aluvihare V, O’Grady J, Heaton N, et
al. Outcomes of pregnancy following liver transplantation: The king’s
college hospital experience. Liver Transpl. 2015. doi: 10.1002/lt.24182

5. Gill JS, Zalunardo N, Rose C, Tonelli M. The pregnancy rate and live birth
rate in kidney transplant recipients. Am J Transplant. 2009;9:1541–9.

6. Deshpande NA, James NT, Kucirka LM, Boyarsky BJ, Garonzik-Wang JM,
Montgomery RA, et al. Pregnancy outcomes in kidney transplant recipients:
a systematic review and meta-analysis. Am J Transplant. 2011;11:2388–404.

7. Seethalakshmi L, Menon M, Malhotra RK, Diamond DA. Effect of
cyclosporine A on male reproduction in rats. J Urol. 1987;138(4 Pt 2):991–5.

8. Haberman J, Karwa G, Greenstein SM, Soberman R, Glicklich D, Tellis V, et al.
Male fertility in cyclosporine-treated renal transplant patients. J Urol.
1991;145:294–6.

9. Xu LG, Yang YR, Wang HW, Qiu F, Peng WL, Xu HM, et al. Characteristics of
male fertility after renal transplantation. Andrologia. 2011;43:203–7.

10. Canobbio MM, Perloff JK, Rapkin AJ. Gynecological health of females with
congenital heart disease. Int J Cardiology. 2005;98:379–87.

11. De Koning ND, Haagsma EB. Normalization of menstrual pattern after liver
transplantation: consequences for contraception. Digestion. 1990;46:239–41.

12. Constantinescu S, Pai A, Coscia LA, Davison JM, Moritz MJ, Armenti VT.
Breast-feeding after transplantation. Best Pract Res Clin Obstet Gynaecol.
2014;28:1163–73.

13. Weber-Schoendorfer C, Hoeltzenbein M, Wacker E, Meister R, Schaefer C. No
evidence for an increased risk of adverse pregnancy outcome after paternal
low-dose methotrexate: an observational cohort study. Rheumatology
(Oxford). 2014;53:757–63.

14. Brouwer J, Laven JSE, Hazes JMW, Schipper I, Dolhain RJEM. Levels of serum
anti-Müllerian hormone, a marker for ovarian reserve, in women with
rheumatoid arthritis. Arthritis Care Res. 2013;65:1534–8.

15. McLaren JF, Burney RO, Milki AA, Westphal LM, Dahan MH, Lathi RB. Effect
of methotrexate exposure on subsequent fertility in women undergoing
controlled ovarian stimulation. Fertil Steril. 2009;92:515–9.

16. Practice Committee of American Society for Reproductive Medicine. Medical
treatment of ectopic pregnancy: a committee opinion. Fertil Steril.
2013;100:638–44.

17. Nurmohamed L, Moretti ME, Schechter T, Einarson A, Johnson D, Lavigne
SV, et al. Outcome following high-dose methotrexate in pregnancies
misdiagnosed as ectopic. Am J Obst Gynecol. 2011;205:533. e1-e3.

18. Weber-Schoendorfer C, Chambers C, Wacker E, Beghin D, Bernard N, Network
of French Pharmacovigilance Centers. Pregnancy outcome after methotrexate
treatment for rheumatic disease prior to or during early pregnancy: a
prospective multicenter cohort study. Arthritis Rheumatol. 2014;66:1101–10.

19. Jones A, Clary MJ, McDermott E, Coscia LA, Constantinescu S, Moritz MJ, et
al. Outcomes of pregnancies fathered by solid-organ transplant recipients
exposed to mycophenolic acid products. Prog Transpl. 2013;23:153–7.

20. Mok CC, Chan PT, To CH. Anti-müllerian hormone and ovarian reserve in
systemic lupus erythematosus. Arthritis & Rheumatol. 2013;65:206–10.

21. Kim M, Rostas S, Gabardi S. Mycophenolate fetal toxicity and risk evaluation
and mitigation strategies. Am J Transplant. 2013;13:1383–9.

22. Cassina M, Johnson DL, Robinson LK, Braddock SR, Xu R, Jimenez JL, et al.
Pregnancy outcome in women exposed to leflunomide before or during
pregnancy. Arthritis& Rheumatol. 2012;64:2085–94.

23. Warnke C, Stüve O, Kieseier BC. Teriflunomide for the treatment of multiple
sclerosis. Clin Neurol Neurosurg. 2013;115 Suppl 1:S90–4.

24. Clowse MEB, Copland SC, Hsieh T-C, Chow S-C, Hoffman GS, Merkel PA, et al.
Ovarian reserve diminished by oral cyclophosphamide therapy for
granulomatosis with polyangiitis (Wegener’s). Arthritis Care Res.
2011;63:1777–81.

25. Cocco E, Sardu C, Gallo P, Capra R, Amato MP, Trojano M, et al.
Frequency and risk factors of mitoxantrone-induced amenorrhea in
multiple sclerosis: the FEMIMS study. Mult Scler. 2008;14:1225–33.

26. Le Page E, Leray E, Edan G, Group FMS. Long-term safety profile of mitoxantrone
in a French cohort of 802 multiple sclerosis patients: a 5-year prospective study.
Mult Scler. 2011;17:867–75.

27. Hellwig K, Schimrigk S, Chan A, Epplen J, Gold R. A newborn with Pierre
Robin sequence after preconceptional mitoxantrone exposure of a female
with multiple sclerosis. J Neurol Sci. 2011;307:164–5.

28. Frias S, Van Hummelen P, Meistrich ML, Lowe XR, Hagemeister FB, Shelby
MD, et al. NOVP chemotherapy for Hodgkin’s disease transiently induces
sperm aneuploidies associated with the major clinical aneuploidy syndromes
involving chromosomes X, Y, 18, and 21. Cancer Res. 2003;63:44–51.

29. Dubey P, Wilson G, Mathur KK, Hagemeister FB, Fuller LM, Ha CS, et al.
Recovery of sperm production following radiation therapy for Hodgkin’s disease
after induction chemotherapy with mitoxantrone, vincristine, vinblastine, and
prednisone (NOVP). Int J Rad Oncol &Biol &Physics. 2000;46:609–17.

30. Ramonda R, Foresta C, Ortolan A, Bertoldo A, Oliviero F, Lorenzin M, et al.
Influence of tumor necrosis factor α inhibitors on testicular function and
semen in spondyloarthritis patients. Fertil Steril. 2014;101:359–65.

31. Almeida BP, Saad CGS, Souza FHC, Moraes JCB, Nukumizu LA, Viana VST,
et al. Testicular Sertoli cell function in ankylosing spondylitis. Clin Rheumatol.
2013;32:1075–9.

32. Mahadevan U, Wolf DC, Dubinsky M, Cortot A, Lee SD, Siegel CA, et al. Placental
transfer of anti-tumor necrosis factor agents in pregnant patients with
inflammatory bowel disease. Clin Gastroenterol & Hepatol. 2013;11:286–92.

33. Gisbert JP, Chaparro M. Safety of anti-TNF agents during pregnancy and
breastfeeding in women with inflammatory bowel disease. Am J
Gastroenterol. 2013;108:1426–38.

34. Koren G, Inoue M. Do tumor necrosis factor inhibitors cause malformations
in humans? J Rheumatol. 2009;36:465–6.

35. Singh JA, Wells GA, Christensen R, Tanjong Ghogomu E, Maxwell L, Macdonald
JK, et al. Adverse effects of biologics: a network meta-analysis and Cochrane
overview. Cochrane Database Syst Rev. 2011;2:CD008794.

36. Framarino-dei-Malatesta M, Derme M, Manzia TM, Iaria G, De Luca L,
Fazzolari L, et al. Impact of mTOR-I on fertility and pregnancy: state of the
art and review of the literature. Expert Rev Clin Immunol. 2013;9:781–9.

37. Tondolo V, Citterio F, Panocchia N, Nanni G, Castagneto M. Sirolimus
impairs improvement of the gonadal function after renal transplantation.
Am J Transplant. 2005;5:197.

38. Huyghe E, Zairi A, Nohra J, Kamar N, Plante P, Rostaing L. Gonadal impact of
target of rapamycin inhibitors (sirolimus and everolimus) in male patients:
an overview. Transpl Int. 2007;20:305–11.

39. Krämer BK, Neumayer H-H, Stahl R, Pietrzyk M, Krüger B, Pfalzer B, et al. Graft
function, cardiovascular risk factors, and sex hormones in renal transplant
recipients on an immunosuppressive regimen of everolimus, reduced dose
of cyclosporine, and basiliximab. Transplant Proc. 2005;37:1601–4.

40. Kaczmarek I, Groetzner J, Adamidis I, Landwehr P, Mueller M, Vogeser M, et
al. Sirolimus impairs gonadal function in heart transplant recipients. Am J
Transplant. 2004;4:1084–8.

41. Bererhi L, Flamant M, Martinez F, Karras A, Thervet E, Legendre C.
Rapamycin-induced oligospermia. Transplantation. 2003;76:885–6.

42. Zuber J, Anglicheau D, Elie C, Bererhi L, Timsit M-O, Mamzer-Bruneel M-F, et
al. Sirolimus may reduce fertility in male renal transplant recipients. Am J
Transplant. 2008;8:1471–9.

43. Rovira J, Diekmann F, Ramírez-Bajo MJ, Bañón-Maneus E, Moya-Rull D,
Campistol JM. Sirolimus-associated testicular toxicity: detrimental but
reversible. Transplantation. 2012;93:874–9.

44. Deutsch MA, Kaczmarek I, Huber S, Schmauss D, Beiras-Fernandez A,
Schmoeckel M, et al. Sirolimus-associated infertility: case report and
literature review of possible mechanisms. Am J Transplant. 2007;7:2414–21.

45. Alfadhli E, Koh A, Albaker W, Bhargava R, Ackerman T, McDonald C, et al. High
prevalence of ovarian cysts in premenopausal women receiving sirolimus and
tacrolimus after clinical islet transplantation. Transpl Int. 2009;22:622–5.

46. Braun M, Young J, Reiner CS, Poster D, Krauer F, Kistler AD, et al. Low-dose
oral sirolimus and the risk of menstrual-cycle disturbances and ovarian cysts:
analysis of the randomized controlled SUISSE ADPKD trial. PLoS One.
2012;7:e45868.

47. Cure P, Pileggi A, Froud T, Norris PM, Baidal DA, Cornejo A, et al.
Alterations of the female reproductive system in recipients of islet
grafts. Transplantation. 2004;78:1576–81.

48. Zhang X, Li L, Xu J, Wang N, Liu W, Lin X, et al. Rapamycin preserves the
follicle pool reserve and prolongs the ovarian lifespan of female rats via
modulating mTOR activation and sirtuin expression. Gene. 2013;523:82–7.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 14 of 15

49. Carta P, Caroti L, Zanazzi M. Pregnancy in a kidney transplant patient
treated with everolimus. Am J Kidney Dis. 2012;60:329.

50. Whirledge S, Cidlowski JA. Glucocorticoids, stress, and fertility. Minerva
Endocrinol. 2010;35:109–25.

51. Poulain M, Frydman N, Duquenne C, N’Tumba-Byn T, Benachi A, Habert R,
et al. Dexamethasone induces germ cell apoptosis in the human fetal ovary.
J Clin Endocrinol Metab. 2012;97:E1890–7.

52. Boomsma CM, Keay SD, Macklon NS. Peri-implantation glucocorticoid
administration for assisted reproductive technology cycles. Cochrane
Database Syst Rev. 2012;6:CD005996.

53. Källén B. Maternal drug use and infant cleft lip/palate with special reference
to corticoids. Cleft Palate-Craniofacial J. 2003;40:624–8.

54. Chi C-C, Wang S-H, Mayon-White R, Wojnarowska F. Pregnancy outcomes
after maternal exposure to topical corticosteroids: a UK population-based
cohort study. JAMA Dermatol. 2013;149:1274–80.

55. Schatz M, Zeiger RS, Harden K, Hoffman CC, Chilingar L, Petitti D. The safety
of asthma and allergy medications during pregnancy. J Allergy Clin
Immunol. 1997;100:301–6.

56. Ng PC, Wong GW, Lam CW, Lee CH, Fok TF, Wong MY, et al. Effect of multiple
courses of antenatal corticosteroids on pituitary-adrenal function in preterm
infants. Arch Dis Child Fetal Neonatal Ed. 1999;80:F213–6.

57. Vermeire S, Carbonnel F, Coulie PG, Geenen V, Hazes JMW, Masson PL, et al.
Management of inflammatory bowel disease in pregnancy. J Crohns Colitis.
2012;6:811–23.

58. Zelissen PM, van Hattum J, Poen H, Scholten P, Gerritse R, te Velde
ER. Influence of salazosulphapyridine and 5-aminosalicylic acid on
seminal qualities and male sex hormones. Scand J Gastroenterol.
1988;23:1100–4.

59. Nørgård B, Czeizel AE, Rockenbauer M, Olsen J, Sørensen HT. Population-
based case control study of the safety of sulfasalazine use during
pregnancy. Aliment Pharmacol Ther. 2001;15:483–6.

60. Riley SA, Lecarpentier J, Mani V, Goodman MJ, Mandal BK, Turnberg LA.
Sulphasalazine induced seminal abnormalities in ulcerative colitis: results of
mesalazine substitution. Gut. 1987;28:1008–12.

61. Akbari M, Shah S, Velayos FS, Mahadevan U, Cheifetz AS. Systematic review
and meta-analysis on the effects of thiopurines on birth outcomes from
female and male patients with inflammatory bowel disease. Inflamm Bowel
Dis. 2013;19:15–22.

62. Dejaco C, Mittermaier C, Reinisch W, Gasche C, Waldhoer T, Strohmer H, et
al. Azathioprine treatment and male fertility in inflammatory bowel disease.
Gastroenterology. 2001;121:1048–53.

63. Francella A, Dyan A, Bodian C, Rubin P, Chapman M, Present DH. The safety
of 6-mercaptopurine for childbearing patients with inflammatory bowel
disease: a retrospective cohort study. Gastroenterology. 2003;124:9–17.

64. Moskovitz DN, Bodian C, Chapman ML, Marion JF, Rubin PH, Scherl E, et al.
The effect on the fetus of medications used to treat pregnant inflammatory
bowel-disease patients. Am J Gastroenterol. 2004;99:656–61.

65. Hoeltzenbein M, Weber-Schoendorfer C, Borisch C, Allignol A, Meister R,
Schaefer C. Pregnancy outcome after paternal exposure to azathioprine/6-
mercaptopurine. Reprod Toxicol (Elmsford, NY). 2012;34:364–9.

66. Coté CJ, Meuwissen HJ, Pickering RJ. Effects on the neonate of prednisone
and azathioprine administered to the mother during pregnancy. J Pediatry.
1974;85:324–8.

67. Sau A, Clarke S, Bass J, Kaiser A, Marinaki A, Nelson-Piercy C. Azathioprine
and breastfeeding: is it safe. BJOG. 2007;14:498–501.

68. Kantarcı G, Şahin S, Uras A, Ergin H. Effects of different calcineurin inhibitors
on sex hormone levels in transplanted male patients. Transplant Proc.
2004;36:178–9.

69. Bourget P, Fernandez H, Bismuth H, Papiernik E. Transplacental passage of
cyclosporine after liver transplantation. Transplantation. 1990;49:663.

70. Kainz A, Harabacz I, Cowlrick IS, Gadgil S, Hagiwara D. Analysis of 100
pregnancy outcomes in women treated systemically with tacrolimus.
Transplantation. 2000;70:1718–21.

71. Jain AB, Reyes J, Marcos A, Mazariegos G, Eghtesad B, Fontes PA, et al.
Pregnancy after liver transplantation with tacrolimus immunosuppression:
a single center’s experience update at 13 years. Transplantation. 2003;76:827–32.

72. Ecevit Ç, Ünal F, Baran M, Aydoğdu S. Parenthood in pediatric liver transplant
patients. Pediatr Transplant. 2012;16:346–9.

73. Cil AP, Leventoğlu A, Sönmezer M, Soylukoç R, Oktay K. Assessment of ovarian
reserve and Doppler characteristics in patients with multiple sclerosis using
immunomodulating drugs. J Turkish Ger Gynecol Assoc. 2009;10:213–9.

74. Dung AA, Panda AK. Interferon β-1a therapy for multiple sclerosis during
pregnancy: an unresolved issue. BMJ Case Report. 2014. doi: 10.1136/bcr-
2013-201273

75. Houtchens MK, Kolb CM. Multiple sclerosis and pregnancy: therapeutic
considerations. J Neurol. 2013;260:1202–14.

76. Ghezzi A, Annovazzi P, Portaccio E, Cesari E, Amato MP. Current
recommendations for multiple sclerosis treatment in pregnancy and
puerperium. Expert Rev Clin Immunol. 2013;9:683–91.

77. Boskovic R, Wide R, Wolpin J, Bauer DJ, Koren G. The reproductive effects of
beta interferon therapy in pregnancy: a longitudinal cohort. Neurology.
2005;65:807–11.

78. Fragoso YD, Adoni T, Alves-Leon SV, Azambuja Jr ND, Barreira AA, Brooks JB,
et al. Long-term effects of exposure to disease-modifying drugs in the
offspring of mothers with multiple sclerosis: a retrospective chart review.
CNS Drugs. 2013;27:955–61.

79. Fragoso YD. Glatiramer acetate to treat multiple sclerosis during pregnancy
and lactation: a safety evaluation. Expert Opin Drug Saf. 2014;13:1743–8.

80. Pecori C, Giannini M, Portaccio E, Ghezzi A, Hakiki B, Pastò L, et al. Paternal
therapy with disease modifying drugs in multiple sclerosis and pregnancy
outcomes: a prospective observational multicentric study. BMC Neurol.
2014;14:114.

81. Lu E, Zhu F, Zhao Y, van der Kop M, Synnes A, Dahlgren L, et al. Birth outcomes
in newborns fathered by men with multiple sclerosis exposed to disease-
modifying drugs. CNS Drugs. 2014;28:475–82.

82. Costedoat-Chalumeau N, Amoura Z, Huong DLT, Lechat P, Piette J-C. Safety
of hydroxychloroquine in pregnant patients with connective tissue diseases.
Review of the literature. Autoimmun Rev. 2005;4:111–5.

83. Ojeda-Uribe M, Afif N, Dahan E, Sparsa L, Haby C, Sibilia J, et al. Exposure to
abatacept or rituximab in the first trimester of pregnancy in three women
with autoimmune diseases. Clin Rheumatol. 2013;32:695–700.

84. Hyrich KL, Verstappen SM. Biologic therapies and pregnancy: the story so
far. Rheumatology (Oxford). 2014;53:1377–85.

85. Chakravarty EF, Murray ER, Kelman A, Farmer P. Pregnancy outcomes after
maternal exposure to rituximab. Blood. 2011;117:1499–506.

86. Fischer-Betz R, Specker C, Schneider M. Successful outcome of two pregnancies
in patients with adult-onset Still’s disease treated with IL-1 receptor antagonist
(anakinra). Clin Exp Rheumatol. 2011;29:1021–3.

87. Ostensen M, Förger F. Treatment with biologics of pregnant patients with
rheumatic diseases. Curr Opin Rheumatol. 2011;23:293–8.

88. Lu E, Wang BW, Guimond C, Synnes A, Sadovnick D, Tremlett H. Disease-
modifying drugs for multiple sclerosis in pregnancy: a systematic review.
Neurology. 2012;79:1130–5.

89. Ebrahimi N, Herbstritt S, Gold R, Amezcua L, Koren G, Hellwig K.
Pregnancy and fetal outcomes following natalizumab exposure in
pregnancy. A prospective, controlled observational study. Mult Scler.
2014;21:198–205.

90. Karlsson G, Francis G, Koren G, Heining P, Zhang X, Cohen JA, et al.
Pregnancy outcomes in the clinical development program of
fingolimod in multiple sclerosis. Neurology. 2014;82:674–80.

91. Soleimani R, Heytens E, Oktay K. Enhancement of neoangiogenesis and
follicle survival by sphingosine-1-phosphate in human ovarian tissue
xenotransplants. PLoS One. 2011;6:e19475.

92. Zelinski MB, Murphy MK, Lawson MS, Jurisicova A, Pau KY, Toscano NP, et al.
In vivo delivery of FTY720 prevents radiation-induced ovarian failure and
infertility in adult female nonhuman primates. Fertil Steril. 2011;95:1440–5.

93. Poels J, Abou-Ghannam G, Herman S, Van Langendonckt A, Wese FX, Wyns
C. In Search of Better Spermatogonial Preservation by Supplementation of
Cryopreserved Human Immature Testicular Tissue Xenografts with N-
acetylcysteine and Testosterone. Front Surg. 2014;1:47. e collection 2014.

94. Su HI, Maas K, Sluss PM, Chang RJ, Hall JE, Joffe H. The impact of depot
GnRH agonist on AMH levels in healthy reproductive-aged women. J Clin
Endocrinol Metab. 2013;98:E1961–6.

95. Del Mastro L, Ceppi M, Poggio F, Bighin C, Peccatori F, Demeestere I, et al.
Gonadotropin-releasing hormone analogues for the prevention of
chemotherapy-induced premature ovarian failure in cancer women:
systematic review and meta-analysis of randomized trials. Cancer Treat
Rev. 2014;40:675–83.

Leroy et al. Orphanet Journal of Rare Diseases (2015) 10:136 Page 15 of 15

Reproduced with permission of the copyright owner. Further reproduction prohibited without
permission.

  • c.ICABMDD_OJRD_20150101_32084110BF24CE40D324_10862
  • Abstract
    Disease name and definition
    Epidemiology
    Clinical description of the consequences of immunosuppressive drugs on fertility and pregnancy
    Immunosuppressive drugs and fertility
    Immunosuppressive therapies and pregnancy
    Diagnosis
    Differential diagnosis
    Aetiologies or effects of the different classes of immunosuppressive agents on fertility and pregnancy
    Contraindicated drugs when pregnancy is desired (Table 1)
    Methotrexate
    Mycophenolate (purine synthesis inhibitor)
    Leflunomide and teriflunomide
    Cyclophosphamide
    Mitoxantrone
    Drugs to be used with caution (Table 1)
    Anti-TNF-α (tumour necrosis factor alpha) (anti-cytokine)
    mTOR inhibitors: sirolimus, everolimus, temsirolimus
    Treatments associated with a low risk of deleterious effects on fertility and pregnancy (Table 1)
    Glucocorticoids
    Sulfasalazine
    Mesalazine
    Azathioprine (purine analog, precursor of 6- mercaptopurine)
    Ciclosporine (calcineurin inhibitor)
    Tacrolimus (calcineurin inhibitor)
    Beta interferon and glatiramer acetate
    Chloroquine
    Recent drugs in which the effects are not fully known (Table 1)
    Biological therapies
    Immunomodulators in multiple sclerosis
    Induction immunosuppressive treatments

    Management of patients using immunosuppressive drugs
    Prognosis and unresolved questions
    Abbreviations
    Competing interests
    Authors’ contributions
    Acknowledgement
    Author details
    References

ITALIAN JOURNAL
OF PEDIATRICS

Davanzo et al. Italian Journal of Pediatrics 2013, 39:50
http://www.ijponline.net/content/39/1/50

RESEARCH Open Access

Antiepileptic drugs and breastfeeding
Riccardo Davanzo, Sara Dal Bo, Jenny Bua, Marco Copertino, Elisa Zanelli and Lorenza Matarazzo*

Abstract

Introduction: This review provides a synopsis for clinicians on the use of antiepileptic drugs (AEDs) in the
breastfeeding mother.

Methods: For each AED, we collected all retrievable data from Hale’s “Medications and Mother Milk” (2012), from
the LactMed database (2013) of the National Library of Medicine, and from a MedLine Search of relevant studies in
the past 10 years.

Results: Older AEDs, such as carbamazepine, valproic acid, phenytoin, phenobarbital, primidone are considered to
have a good level of safety during lactation, due to the long term clinical experience and the consequent amount
of available data from the scientific literature. On the contrary, fewer data are available on the use of new AEDs.
Therefore, gabapentin, lamotrigine, oxcarbazepine, vigabatrin, tiagabine, pregabalin, leviracetam and topiramate are
compatible with breastfeeding with a less documented safety profile. Ethosuximide, zonisamide and the continue
use of clonazepam and diazepam are contraindicated during breastfeeding.

Conclusions: Although the current available advice on the use of AEDs during breastfeeding, given by different
accredited sources, present some contradictions, most AEDs can be considered safe according to our review.

Keywords: Antiepileptic drugs, Breastfeeding, Lactation risk

Introduction
Breastfeeding is known for its beneficial effects on both
mothers and infants [1,2]. Nevertheless, in mothers suf-
fering from epilepsy or bipolar disorders treated with
antiepileptic drugs (AEDs), some concerns on infant
health may raise. The decision to encourage breast-
feeding in those women should be taken after a careful
evaluation of the possible side-effects on the infant
caused by the indirect exposure to AEDs via breast milk.
Data on the use of AEDs by the nursing woman are
mainly represented by single pharmacologic or pharma-
cokinetic studies and/or by case reports or case series on
the side-effects attributed to their presence in breast
milk. Moreover, toxicological and clinical data on AEDs
during breastfeeding are dispersed in the scientific litera-
ture and consequently not easily accessible to health
professionals called to give an evidence-based clinical
advice. The present paper aims at providing the clinician
with an updated synopsis of the lactation risk of AEDs.

* Correspondence: lorenza.matarazzo@gmail.com
Division of Neonatology, Institute for Maternal and Child Health – IRCCS
“Burlo Garofolo”, Via dell’Istria 65/1, Trieste 34100, Italy

© 2013 Davanzo et al.; licensee BioMed Centr
Commons Attribution License (http://creativec
reproduction in any medium, provided the or

Methods
Drugs considered for the present review have been se-
lected as the most frequently used among those catego-
rized as major antiepileptic (Code N03) by the Anatomical
Therapeutic Chemical Classification System (ATC) [3]
together with the anxiolytic diazepam (N05BA01). Before
assessing the lactation risk of each AED, we have collected
information on their main pharmacokinetic parameters:
plasma protein binding, half-life, milk-to-plasma ratio,
oral bioavailability (see Table 1 for definitions). We de-
cided to present data on AEDS pharmacokinetic parame-
ters as they represent the theoretical basis on which the
lactation risk assessment should be developed. As an ex-
ample, we can assume that if a drug has a short half-life
(<3 hours), its level in the maternal plasma will be declin- ing when the infant feeds again, considering a 3-hour interval. Moreover, if the drug is highly protein bound, it cannot enter the milk compartment easily. Milk/plasma ratio has the primary use of quantifying the extent of drug transfer into the milk; nevertheless, its use in assessing a lactation risk is limited as the amount of drug transfer into milk is mainly determined by the maternal plasma level. Medications for the breastfeeding mother should have a low oral bioavailability, as the result of either a poor gut

al Ltd. This is an Open Access article distributed under the terms of the Creative
ommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
iginal work is properly cited.

Table 1 Definitions and clinical relevance of the pharmacokinetic parameters used to assess the lactation risk
following maternal intake of medications
Pharmacokinetic parameter Definition
Half-life or “T ½” The half-life of a substance is the time it takes for its plasma concentration to halve. If the half-life is long (>12-24 hrs),
drugs may accumulate in maternal plasma and the time of drug transfer from plasma to breast milk is longer.
Maternal plasma protein
binding (PB)
This parameter is expressed in percentage. The higher the percentage of the drug bound to the maternal plasma
proteins, the less the drug passes into breast milk. An ideal drug to be taken during breastfeeding should have a
plasma protein binding > 80%.
Milk-to-plasma ratio (M/P) It denotes the ratio of the drug concentration in the mother’s milk (M) divided by its concentration in the mother’s
plasma (P). It is an indicator of drug transfer into breast milk. A M/P ratio greater than 1.0 suggests that the drug may
be present in breast milk in high concentrations.
Oral bioavailability It describes the fraction of one orally administered dose of a drug that reaches the systemic circulation. It is expressed
as a percentage of the administered dose. When the intestinal absorption of a drug is impaired, the risk of adverse
effects may be lower.
Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 2 of 11
http://www.ijponline.net/content/39/1/50
absorption, or the liver sequestration prior to entering the
plasma compartment. Table 2 summarises each AED main
pharmacokinetic characteristics.
Beyond presenting pharmacokinetic parameters on
AEDs, we chose to collect more relevant clinical param-
eters such as the theoretical infant dose (TID), the thera-
peutic dose in the neonatal period and the relative infant
dose (RID). All these parameters were reported in a table
(Table 3) together with the assessment of the lactation
Table 2 Main pharmacokinetic characteristics of antiepileptic
PB† Oral
Bioavailability ‡
(%) (%)
Carbamazepine 74 100
Clonazepam 50 – 86 100
Diazepam 99 100
Ethosuximide NA 100
Gabapentin < 3 50 – 60 Lamotrigine 55 98 Levetiracetam < 10 100 Oxcarbazepine 40 100 Phenobarbital 51 80 – 100 Phenytoin 89 70 – 100 Pregabalin NA 90 Primidone 25 90 Tiagabine 96 90 Topiramate 15 75 Valproate 94 100 Vigabatrin NA 50 Zonisamide 40 NA † PB: maternal plasma protein binding expressed as percentage. ‡ Oral Bioavailability: intestinal absorption after oral administration expressed as pe § T ½: half-life of the drug. ¶ M/P: milk to plasma ratio of a drug concentration. Data drawn from references 1–8 except where otherwise specified. NA: indicates th risk (see below), in order to have a clinical overview of each AED. TID is the maximum estimated amount of ingested drug with breast milk; in other words, it is the estimation in milligrams per kilogram per day of the theoretical infant dose (TID). We calculated it by using the formula by Atkinson (TID = daily breast milk intake (150 ml/kg/day) × maximum breast milk concentration of medication). [4] As AEDs may be used in the neonatal period as therapy, drugs T ½ § M/P¶ (hours) 18 – 54 0.69 18 – 50 0.33 43 0.2 – 2.7 30 – 60 0.94 5 – 7 0.7 – 1.3 29 0.057 – 1.47 [33] 6 – 8 0.76 – 1.55 [37,38] 9 (oxcarbazepine metabolyte) 0.5 20 – 133 0.4 – 0.6 (45–500 in newborns) [41] 6 – 24 0.18 – 0.45 (20–160 in preterm infants) [48] 6 NA 5 – 18 0.72 7 – 9 NA 18 – 24 0.86 – 1.1 [55] 14 0.42 7 < 1 63 0.93 rcentage of the administered dose. at no data are available. Table 3 Clinical relevant parameters for each antiepileptic drug and assessment of their lactation risk Drug TID† Neonatal therapeutic oral dose RID‡ Assessment of the lactation risk according to: (mg/kg/day) (%) Hale 2012§ LactMed 2013 Present study (mg/kg/day) (including adverse drug reactions) Carbamazepine 0.7 10 - 20 3.8 - 5.9 L2 CBZ levels are relatively high in breast milk Safe ▪ Breastfed infants have serum levels that are usually below the therapeutic range. ▪ Side effects were rarely reported as sedation, decreased suckling, withdrawal reactions and 3 cases of liver dysfunction. ▪ Infant should be monitored for jaundice, drowsiness, adequate weight gain, and developmental milestones especially in premature infants, exclusively breastfed and in combination with other antipsychotics. Clonazepam 0.002 0.1 - 0.2 2.8 L3 ▪ Monitor growth, sedation, developmental milestones, especially in preterm neonates, exclusively breastfed infants and if mother is receiving psychotropic drugs. Contraindicated ▪ Monitoring of serum concentration in breastfed infant, if excessive sedation occurs. Diazepam 0.05 IV dose available: 7.1 L3 ▪ Accumulates in maternal milk and serum of breastfed infant. Other agents are preferred, especially while nursing a newborn or preterm infant. Contraindicated 0.1- 0.3 Oral dose ▪ Single dose does not require delaying feeding.0.5 – 1 [76] Ethosuximide 11.5 15 – 40 31.4-73.5 L4 ▪ Monitor infant for drowsiness, adequate weight gain and psychomotor development. Contraindicated ▪ Measurement of an infant serum level might help rule out toxicity, if there is a concern. Gabapentin 1.7 Only paediatric dose available: 1.3 - 6.6 L2 ▪ Monitor the infant for drowsiness, adequate weight gain, and developmental milestones, especially in younger, exclusively breastfed infants and when using combinations of anticonvulsant or psychotropic drugs. Moderately safe 10-15 Lamotrigine 0.7 1-6 with valproate, 5- 15 with enzyme inducing AEDs 9.2 L3 ▪ It is not necessary to discontinue breastfeeding, but any adverse effects such as apnoea, rash, drowsiness, decreased sucking are to be monitored and serum levels are to be measured. Moderately safe (testo: 9.2-18.3) ▪ Monitoring of the platelet count may also be advisable. Levetiracetam 3.9 5 - 10 [78] 3.4 - 7.8 L3 ▪ Monitor infant for the appearance of sleepiness, increase appropriate weight, normal psychomotor development. Moderately safe Oxcarbazepine NA 27.7 – 50 1.5-1.7 L3 ▪ Monitor the infant for drowsiness and decreased feeding, and developmental milestones especially in the first 2 months of life. Moderately safe (<18 years) [72] Phenobarbital 0.4 3-4 24 L3 ▪ The presence of phenobarbital in breast milk may mitigate possible neonatal abstinence. Safe ▪ Monitor the breastfed infant for the possible onset of drowsiness, adequate weight gain and developmental milestones, especially in younger, exclusively breastfed infants and antiepileptic polytherapy. ▪ Measurement of the infant’s serum drug concentration might help rule out toxicity. Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 3 of 11 http://www.ijponline.net/content/39/1/50 Table 3 Clinical relevant parameters for each antiepileptic drug and assessment of their lactation risk (Continued) Phenytoin 0.4 5-8 0.6-7.7 L2 ▪ The proportion ingested by infants is small and generally brings about no problems except in rare cases of idiosyncratic reactions. Safe Pregabalin NA 5-14 [79] NA L3 ▪ Compatible with breastfeeding. Moderately Safe ▪ An alternate drug may be preferred, especially while nursing a newborn or preterm infant. Primidone 0.9 12-20 8.4-8.6 L3 ▪ The presence of phenobarbital in breast milk may mitigate possible neonatal abstinence. ▪ Monitor the breastfed infant for the possible onset of drowsiness, adequate weight gain and developmental milestones, especially in younger, exclusively breastfed infants and antiepileptic polytherapy. Safe ▪ Measurement of the infant’s serum drug concentration might help rule out toxicity. Tiagabine NA <12 years: limited data available. NA L3 ▪ Monitor the infant for the onset of drowsiness, for adequate weight gain and for developmental milestones especially in younger, exclusively breastfed infants and when using combinations of anticonvulsant or psychotropic drugs. Moderately safe ▪ Other drugs should be preferred especially while nursing a newborn or preterm infant. Topiramate 0.3 1 - 6 24.5 L3 ▪ Monitor the infant for the onset of diarrhea, drowsiness, increase appropriate weight and psychomotor development. Moderately safe (<2 years) [80] Valproate 0.7 Limited data available in the neonatal period. 1.4-1.7 L3 ▪ Breastfed infants are at risk for hepatotoxicity. Safe ▪ Monitor the infant for unusual bleeding (a case of thrombocytopenia has been reported). Vigabatrin 0.1 25-50 1.5 – 2.7 L3 ▪ Until more data are available, vigabatrin should only be used with careful monitoring during breastfeeding. Moderately safe S-enantiomer Zonisamide 1.9 5 – 8 28.9 -36.8 L4 ▪ Monitor infant for drowsiness, adequate weight gain and psychomotor development. Contraindicated ▪ Measurement of an infant serum level might help rule out toxicity if there is a concern. † TID theoretical infant dose. ‡ RID relative infant dose. § Hale lactaction risk categories: L1: safe drugs at the highest level, L2: safe, L3: moderately safe; L4: possibly dangerous, L5: contraindicated. Present study risk categories: The moderately safe category has a less documented safety profile due to a short clinical experience and lack of studies. The moderately safe AEDs can be used, but the lowest dose of the drug should be chosen and the nursing infant should be clinically monitored and, when possible, his/her plasma level should be checked. Data drawn from reference 1,41 except where otherwise specified. NA: indicates that no data are available. Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 4 of 11 http://www.ijponline.net/content/39/1/50 we presented their therapeutic dose, expressed in mg/kg/ day in order to have a direct comparison with the TID. A therapeutic dose which is higher than the TID reassures of the drug safety during breastfeeding. RID represents one of the most useful parameters for assessing the lactation risk. RID is calculated by dividing the infant dose via milk in “mg/kg/day” by maternal dose in “mg/kg/day”, assum- ing a 70 kg weight. [5] Many authors agree that anything less than 10% of the maternal dose is considered probably safe [5]. To review the lactation risk of AEDs, we consulted the following two most accredited English sources: Medica- tions and Mother’s Milk 2012, a Manual of Lactational Pharmacology [1], and the 2013 Lactmed database (in TOXNET) [6] of the National Library of Medicine. In his textbook, updated every two years, Hale collects data on many current medications and their use during breastfeeding. After evaluating information on pharma- cokinetics and what is currently published in the scien- tific literature for each drug, including its reported side effects, he makes a personal recommendation, using a 5 categories of lactation risk: L1: safe drugs at the highest level, L2: safe drugs, L3: moderately safe drugs, L4: drugs possibly dangerous, L5: contraindicated drugs. LactMed database is part of the National Library of Medicine’s Toxicology Data Network (TOXNET), and it includes information on the levels of drugs and other chemicals to which breastfeeding mothers may be exposed in Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 5 of 11 http://www.ijponline.net/content/39/1/50 breast milk and infant blood, together with the possible adverse effects in the nursing infant. All data published in LactMed are derived from the scientific literature and fully referenced. To complete our synopsis, we have also performed a non-systematic Medline search of the literature with the keywords “antiepileptic drugs” AND “breastfeeding”, re- trieving studies from 2004 to April 9th 2013 , including the most relevant and up-dated studies on lactation risk. Data from the Committee on Drugs of the American Academy of Pediatrics (AAP) [7], and the Goodman & Gilman’s Textbook of Pharmacology [8] were also included in our review. For each AED we organized the relevant data into small summaries. As the result of our review we classified AEDs in 3 categories: safe, moderately safe and contraindicated during breastfeeding. The moderately safe category has a less documented safety profile due to a short clinical experience and lack of studies. However, the moderately safe AEDs can be used with caution. The lowest dose of the drug should be chosen and the nursing infant should be monitored and, when possible, his/her plasma level should be checked. Results Benzodiazepines Benzodiazepines (BDZ) are multiple-action psychoactive compounds. Therefore, they may be used as anxiolytics, antiepileptics, sedatives, hypnotics, muscle relaxants, and as coadjuvants in anaesthesia induction. Simplifying, we can state that all BDZ produce all these effects, albeit with different expression of their principal effect (relative selectivity). In general, the first step to judge the safety of BDZ during lactation is to know their half-life (short, inter- mediate, long). Independently of other kinetic character- istics, the longer is a drug half-life, the longer is its passage into maternal milk, and the greater is the infant metabolic effort to metabolize the drug. Both diazepam and clonazepam have long half-life (diazepam 43 hours, clonazepam 18–50 hours) [1]. Consequently, their pri- mary risk to the infant is drug accumulation, which also depends on the infant metabolism (i.e. slower in preterms) and the therapy duration. Intermittent, short-term therapy (24–72 h), carries a negligible risk of accumulation, whereas prolonged use of a long half-life BDZ such as diazepam and clonazepam, carries a greater risk. In the meanwhile, there is no lactation risk after a single dose of BZD, as no drug accumulation occurs. Diazepam is highly protein bound and its transfer into breast milk is variable. Concentrations of diazepam and its metabolite desmethyldiazepam in maternal milk vary between 7.7 and 87 ng/L and 19.2 and 77 ng/L, respectively [9]. Its active metabolite tends to accumulate during prolonged lactation. Lethargy and difficult feeding have been attributed to diazepam during breastfeeding [10]. Maternal plasma peak usually occurs within 2 h. By delaying feeding, the infant exposure to diazepam with maternal milk can be reduced. Despite its scarce transfer into maternal milk, clonaze- pam was reported to cause irregular breathing, apnea and cyanosis in the first 10 days of life in an infant whose mother had been taking the drug also during pregnancy. The child psychomotor development at 5 months was normal [11]. On the contrary, in the study of Birnbaum, no side effects on breastfed infants were reported by mothers treated with benzodiazepines in association with antidepressants [12]. Clonazepam has a later plasma peak (2–6 hours) which limits the feasibility of delaying feeding in order to reduce the infant exposure. Carbamazepine Carbamazepine (CBZ) is a broad-spectrum anticonvul- sant, also used in psychiatric disorders (such as schizo- phrenia) and in trigeminal neuralgia. If taken during pregnancy, its concentration compared to the maternal serum level ranges 60-76% in the umbilical cord and 32- 80% in breast milk [13]. Most available studies on carba- mazepine in human milk usually refer to women under anticonvulsant polytherapy with possible interference between drugs. The CBZ and its active epoxide metabolite (ECBZ) are poorly excreted into breast milk (M/P: 0.69 and 0.79 re- spectively), partly due to the good plasma protein bind- ing. CBZ concentration in breast milk in the current literature is widely variable (0.34-6 mg/L) [14–17]. Its RID is low (3.8-5.9%) [1] and serum levels in babies are usually low, while ECBZ is not detected at all [18–20]. LactMed Database suggests to monitor breastfed in- fants for adequate growth and possible onset of sedation and jaundice, given some case reports of liver dysfunc- tion with cholestasis (increase in transaminases and gammaGT) [21,22]. Nevertheless, the drug is considered compatible with breastfeeding [7,22–25]. In fact, it is ad- ministered, with a good safety profile, directly to patients of pediatric age. Ethosuximide Ethosuximide is used in the treatment of absence sei- zures. Protein binding of the drug is insignificant. Its half-life is shorter in children (30–60 h) than in adults (about 45 h) and the M/P ratio is 0.94 [1]. The amount of ethosuximide excreted in breast milk leads to a rele- vant RID (31.4-73.5%) [1], which explains the significant serum concentrations in breastfed infants (15–40 mg/L) [26]. Although the study of Rane reported some neuro- logical symptoms in breastfed infants (hyperexcitability, Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 6 of 11 http://www.ijponline.net/content/39/1/50 sucking difficulty), this could have been due to the antiepileptic polytherapy of their mothers and/or to the combination with withdrawal symptoms [27]. Ethosu- ximide is considered to be potentially dangerous during lactation [1] and monitoring breastfed infant has been suggested [26]. Gabapentin Gabapentin is used in the treatment of partial epilepsy, cluster headache [28], neuralgia, post-caesarean delivery pain [2] and some psychosis. Gabapentin has a medium half-life and is believed to accumulate in the fetus, while it does not concentrate in breast milk (average M/P = 0.7-1.3) [1,29], leading to low serum levels and no side effects in breastfed infants [29,30]. Lamotrigine In fetuses exposed to lamotrigine, concentrations are highest at birth and then gradually decline over time, more quickly if the infant is not breastfed [31]. Some of the pharmacokinetic characteristics of lamotrigine, such as the long half life (29 hours) and the low protein bind- ing (55%) [1] together with a recent reported case of severe toxicity [32], warrant attention towards its use during breastfeeding. The anecdotal case by Nordmo refers to a 16 days-old-infant whose mother had shown visual disturbances and dizziness, after taking a high dose of lamotrigine (850 mg/day) [32]. Soon after, the baby had a series of episodes of apnea followed by a cyanotic crisis which requested resuscitation. Plasma level of lamotrigine in that infant was consistently high (4.87 μg/mL). After discontinuing the drug, the infant recovered. The excretion of lamotrigine in breast milk is largely variable. A study on 30 breastfeeding women, treated for more than 7 days with lamotrigine at doses 300– 450 mg/day, showed that breast milk samples collected over 24 h contained 0.5-18.1 μg/mL of lamotrigine [33]. The M/P ratio ranged from 0.057 to 1.47. [1,33] The dose that the breastfed infant would take has been calcu- lated between 0.37 and 0.65 mg/kg/day, well below the dose given to infants affected by epilepsy resistant to common antiepileptic drugs [34,35]. The average RID is 9.2 (33). Liporace reports that serum concentrations of lamotrigine in breastfed children in some cases reach therapeutic ranges [36]. These high levels may be explained by the genetic variability in the neonatal glycuroconjugation responsible for lamotrigine metabolism [36]. When maternal plasma levels are 4.5-13.4 μg/mL, breastfed infant plasma levels are 0.6-1.8 μg/mL, with a M/P ratio 0.413% on average [33]. Moreover laboratory tests (electrolytes, hepatic function tests, complete blood count) have been all normal, except from a modest thrombocytosis (range: 329.000–652.000/mm3 PLT, in 7 out of 8 infants tested) [33]. Eventually, Newport et al. reassure on lamotrigine use while breastfeeding, as no adverse effects was reported in the nursing infants in their study [33]. In conclusion, lamotrigine is considered moderately safe during breastfeeding [1,30], and LactMed recom- mends to check its plasma level and to carry out a platelet count in infants whose mothers are on lamotrigine [6]. Levetiracetam Levetiracetam is a new AED, and is usually added to other drugs in case of inadequate control of seizures. It significantly passes into breast milk (M/P: 0.76-1.55) [5,37,38], it is completely absorbed orally and it has a pediatric half-life of about 18 hours with a low RID (3.4- 7.8%) [1]. Serum levels of levetiracetam in breastfed infants are low (<21 μmol/L) and no side-effects have been reported when doses between 1.3 and 3 g/day are administered to their mothers [37,38]. Consequently, levetiracetam is usually considered com- patible with breastfeeding [1,37,38], even if associated to other AEDs such as primidone and phenobarbital [39]. Oxcarbazepine Oxcarbazepine (OXC) is a pro-drug which is converted in its active metabolite (10-OH-carbazepine) (10-OH-CBZ) in the liver. Reports of its use while breastfeeding are lim- ited. Most information is obtained by the paper of Lutz: oxcarbazepine has a long half-life (9 h), a low M/P ratio (0.5) and a low concentration in human milk (<11 μg/mL) with a low RID (1.5-1.7%) [40]. It lacks side effects in breastfed infants. Blood levels in infants of OCX and 10- OH-CBZ are negligible (both <0.2 μg/mL). Therefore, we can consider OXC moderately safe in the breastfeeding mother. Phenobarbital Phenobarbital is widely used in both adults and children. Its metabolism is mainly hepatic. The extremely long half-life in the pediatric age (20–133 hours in infants and up to 500 hours in newborns) [41] and the lower plasma protein binding in neonates compared to adults (3-43% vs 51%) could explain why its blood levels may be higher in newborns than in their mothers. The main, yet rare, side-effect attributed to phenobar- bital is sedation, holding true in adults as well as in breastfed babies. This occurrence is reported more fre- quently in infants whose mothers are in polytherapy, because they are likely to be exposed to interactions be- tween drugs with possible enhancement of both clinical and side effects [39]. Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 7 of 11 http://www.ijponline.net/content/39/1/50 Phenobarbital is commonly and safely used at a daily dosage 5–7 mg/kg/day in newborns affected by seizures or drug-abstinence syndrome. This dose is far higher than the estimated dosage (2–4 mg/day) received by a breastfed infant by a woman taking 150 mg (a high dose) phenobarbital per day. Given these data, there is no need to discontinue breastfeeding in mothers treated with phenobarbital. However, AAP is more cautious and recommends to monitor all breastfed infants whose mothers are on phenobarbital [7]. In premature babies or in infants with drowsiness, difficulty in sucking or poor weight gain, it is recommended to monitor its plasma levels [6,42–44]. Phenytoin Despite the formerly reported methemoglobinemia in in- fants [45], phenytoin is considered to be safe in lactation [5,46,47], as it is highly bound to plasma proteins (89%) [1], it scarcely passes into breast milk (M/P: 0.18-0.45) [1,48,49] and has low concentration in human milk [49]. In fact, milk concentration of phenytoin has been reported to be only 1.9 mg/L following maternal intakes equal to 300–600 mg [50]. The amount ingested by nursing infants is usually low (RID 0.6-7.7%) [1]. Pregabalin Pregabalin is used to treat postoperative [51] and neuro- pathic pain and some psychosis. There are no studies on the passage of pregabalin into human milk. The absence of binding to plasma proteins and its excellent oral bio- availability [52] suggest that it can pass into the mother’s milk and into the circulation of the nursing infants. Numerous side effects such as dizziness, drowsiness, impaired vision have been observed in adults. It is rated as moderately safe during breastfeeding [1]. Primidone Primidone is metabolized to phenobarbital and other derivates. Serum levels of primidone and its metabolites in breastfed infants could be close to the therapeutic range values and cases of sedation and poor feeding are reported in literature. It should be used with caution during breastfeeding [14,42,43,53,54], also according to the AAP [7]. The clinical assessment of its use during breastfeeding is similar to phenobarbital. Tiagabine Tiagabine oral absorption is almost complete and it is highly bound to plasma proteins [41]. There are no stud- ies on its use during breastfeeding, leading to possibly prefer other antiepileptic drugs. If taken by the nursing mother, infant should be monitored [46]. Topiramate Topiramate use is increasingly prescribed, being effective and well-tolerated by epileptic patients. It is rapidly absorbed, it has a low plasma protein binding, a rela- tively long half-life and a significant excretion into breast milk (M/P:0.86-1.1) [1,55] with a high RID (24.5%) [1]. Despite these characteristics may raise some concerns on its use during breastfeeding, breastfed infants have been found to have very low serum levels (<2.8 μmol/L) in the first 3 months of life after maternal intakes of 150–200 mg/day of topiramate in association with CBZ [55]. These low serum levels seem to depend on the in- fant good capability to eliminate topiramate, possibly facilitated by CBZ enzyme induction [55]. Moreover, infants who were breastfed by mothers treated with topiramate did not show side-effects. Notwithstanding, they should be monitored [55,56]. Valproate Patients taking valproate may develop hepatotoxicity, thrombocytopenia and anemia. The limited passage of valproate into breast milk (the drug is almost completely bound to plasma proteins) make it safe in lactation. [23,24,46,47] Serum levels in breastfed infants are low [18]. However, some controversies exist on its safety profile. The teratogenic effect of valproate exposure in pregnancy [57], together with the decrease of methyla- tion in the DNA extracted from the umbilical cord blood [58] and a report of a breastfed infant with thrombocyto- penia, purpura and anemia [59], induce a cautious use of this AED during breastfeeding [1]. Although there is no indication to perform routine laboratory investigations, in case of late jaundice, it is reasonable to assess the hepatobiliary function and to test its plasma levels in the breastfed infant [23,46,57–62]. Vigabatrin Vigabatrin is commonly used for multi-resistant epi- lepsy. The maternal plasma concentration is 93 μmol/L following an intake of 1.5 g vigabatrin. There are no pre- cise data about its passage into breast milk. Since no information is available about its use during lactation, the nursing infants should be monitored [46]. Zonisamide Zonisamide is a broad-spectrum anticonvulsant with a half-life of nearly 63 hours [1], even if some authors re- port a longer one (109 hours) [63]. It easily passes into breast milk (M/P: 0.93) [6,64]. Although no studies document side-effects in children of mothers receiving zonisamide, due to a very high RID (28.9-36.8%), it has been considered contraindicated by some authors [1]. Others authors simply recommend measurement of Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 8 of 11 http://www.ijponline.net/content/39/1/50 infant serum level to rule out toxicity, if there is a con- cern [6]. Table 2 summarises each AED main pharmacokinetic characteristics. Table 3 gives an overview of the clinical relevant parameters for each antiepileptic drug (TID, therapeutic dose, RID) and the assessment of their lacta- tion risk according to the two main sources (Hale and LactMed) and to our study group. Discussion The use of AEDs is often unavoidable for most mothers with epilepsy. Moreover, the post partum period is a vul- nerable time for women with epilepsy, owing to the changes in the drug metabolism and possibly to sleep deprivation. Both these conditions may trigger an epilep- tic crisis [65]. As the newborn might be indirectly exposed to AEDs via breast milk, pharmacologically treated epilepsy has been and sometimes is still considered a contraindica- tion to breastfeeding, irrespective of which AED is taken by the mother [66]. On the contrary, health professionals have currently become more aware of the need to weigh the short and long-term risks for the nursing infant against the well demonstrated nutritional, immuno- logical, developmental, economic and ecological benefits of breastfeeding [1,2,67]. When evaluating the safety profile of an AED during lactation, the physician should collect and process data which are often dispersed in the scientific literature and difficult to retrieve [66]. We have therefore completed the present review in order to facili- tate the consultancy on the use of AEDs during breast- feeding. The comparison of advices from the main and most recent accredited literature sources has highlighted the existence of a certain degree of inconsistency [66], which is probably the consequence of the combination of lim- ited scientific evidence and of a certain degree of arbi- trariness. This heterogeneity of advice is not specific of AEDs. We have previously documented a metavariability in the assessment of the lactation risk of other classes of drugs, such as beta-blockers [68], corticosteroids [69] and antidepressants [70]. Evaluating the lactation risk of any drug is complex and should take into account several aspects. In our re- view, we have shown that neither pharmacokinetic infor- mation nor more clinical parameters (such as TID and RID) are alone good predictors of the lactation risk. As an example, a drug such as phenobarbital, which is poorly bound to the mother plasma proteins and has a relatively long half-life and an high RID, can still be con- sidered compatible during breastfeeding [1]. When evaluating the lactation risk of a drug, we should always check whether or not toxicity in breastfed infants is reported. AEDs are expected to determine in the nursing infant a series of symptoms related to their pharmacological effects on the central nervous system, such as altered sleep patterns, abnormal tone, poor feed- ing and possibly poor growth secondary to inadequate breast sucking. However, the clinical relevance of these attributed side-effects vary widely. As an example, the lactation risk of infant apnea after maternal use of clo- nazepam [11] deserves a higher level of caution than the risk of infant sedation after maternal use of CBZ. Another variable to take into account when consider- ing the infant tolerance to AEDs, is his/her ability to metabolize the drug. This is much lower when the ma- ternal treatment begins before or during pregnancy (as the exposure of the baby through breast milk adds to the one through the placenta), or in case of prematurity and during the first 2 months of life [71]. Maternal polytherapy is also expected to increase the lactation risk, as the result of the possible synergistic interaction of different medications [39]. We might also speculate that the alleged long-term nega- tive effects on child psychomotor development resulting from the passage of small amounts of the drug through breast milk, though up to now not well-documented, could be well-balanced by the benefits that breast- feeding confers to the cognitive, social and relational development. This hypothesis seems to be supported by the results from a study conducted in the US on 199 children exposed in uterus to AEDs (phenytoin, carba- mazepine, valproate and lamotrigine) and followed in the first 3 years of life. In this cohort, there was no sig- nificant difference regarding cognitive scores between breastfed and not breastfed children [24]. Breastfeeding mothers should be provided sound and clear information on the lactation risk of the prescribed medications. Nevertheless, breastfeeding mothers taking AEDs happen to receive inconsistent and sometimes conflicting advices on whether or not to breastfeed from different clinicians (general physician, neurologist, pedia- trician, obstetrician, etc.). Unluckily, many such given ad- vices are not evidence-based, but simply emphasize not circumstantial assessments of the lactation risk. We believe that mothers treated with AEDs should be encouraged to start breastfeeding immediately after child- birth, even if a definite and complete evidence-based in- formation has yet not been collected. In fact, breast milk should not be considered as a “presumed guilty”, because, after a complete scrutiny, only very few AEDs resulted contraindicated during breastfeeding [24,72,73]. The present study is not without limitations. First of all, we did not conduct a systematic review of the pub- lished literature, but we offered a synopsis of the two most updated and authoritative clinical sources on the lactation risk (namely: Hale and Toxnet). Secondly, the existing reports on the lactation risks of medications in Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 9 of 11 http://www.ijponline.net/content/39/1/50 general [73], and particularly of AEDs [44] , are usually anecdotal or based on case series and therefore of poor methodological quality. However, we should remember that any recorded side-effect, while biologically and pharmacologically plausible in the nursing infant, is rarely attributable with certainty to a particular drug taken by her/his mother [74]. Conclusions According to the present review, we grouped AEDs into 3 main classes of lactation risk: 1. AEDs safe to use during lactation. Carbamazepine, valproic acid and phenytoin are compatible with breastfeeding. Phenobarbital and primidone are also compatible, although particular attention should be paid on infant monitoring, possibly measuring the infant plasma drug level. 2. AEDs with a less documented safety profile during lactation (moderately safe). Gabapentin, lamotrigine, levetiracetam, oxcarbazepine, pregabalin and vigabatrin may be used during lactation, but their lowest dose should be prescribed. The nursing infant should be carefully monitored and, if required, the infant drug plasma-level tested. When lamotrigine is taken by the mother, it is advisable to check the presence of thrombocytopenia in the nursing infant. Although based on scarce available data, tiagabine and topiramate can be considered compatible with breastfeeding. 3. AEDs contraindicated during lactation. Ethosuximide should be used only when there are no alternatives and after informed consent of the mother. Lastly, zonisamide and the continuous use of diazepam or clonazepam should be avoided. Even if an AED can be safely taken by the breastfeeding mother, it is a good clinical practice to call the mother attention on the behavior, sleep, feeding patterns and growth of the nursing infant, especially in the first 2 months of life [75]. When possible, we might advice to monitor the infant plasma levels, especially for the moder- ately safe AEDs. However, we are aware that there is no clear agreement on when to test them. We believe that after 4–8 weeks is a reasonable time window, as, if the child is fully breastfed, breast milk would have reached its highest production and the consequent intake by the nursing infant would be at the maximum. Competing interests This study was performed without any funding or grants. All the authors declare that they have no competing interest and do not have any financial relationships with any biotechnology and/or pharmaceutical manufacturers. Authors’ contributions RD conception and design. LM, SDB, RD analysis and interpretation of the data. LM, SDB RD drafting of the article. RD critical revision of the article for important intellectual content. All authors listed here have seen and approved the final version of the report. JB administrative, technical, or logistic support. MC, EZ collection and assembly of data. Acknowledgements We thank Dr. Gian Paolo Chiaffoni (Pediatric Department, Conegliano, Italy) and Prof. Giuliana Decorti (Institute of Pharmacology, University of Trieste, Italy) for helpful advice. Received: 23 April 2013 Accepted: 10 August 2013 Published: 28 August 2013 References 1. Hale TW: Medications and Mother’s milk 2012. 15th edition. 1712 N Forest Amarillo TX: A Manual of Lactational Pharmacology; 2012:79106–7017. Pages 184-186, 264-265, 325-326, 426-427, 495-496, 651-653, 664-665, 870-871, 905-906, 910-911, 938-939, 940-941, 1077, 1091-1092, 1126-1127, 1136-1137, 1161-1162. 2. Section on Breastfeeding: Breastfeeding and the use of human milk: an analysis of the American Academy of Pediatrics. Pediatrics 2012, 129(3):e827–841. 3. WHO: Collaborating centre for drug statistics methodology, Norwegian institute of public health. Olso: Guidelines for ATC classification and DDD assignment; 2012:15–21. 4. Atkinson HC, Begg EJ, Darlow BA: Drugs in human milk. Clinical pharmacokinetic considerations. Clin Pharmacokinet 1988, 14(4):217–40. 5. Ip S, Chung M, Raman G, Trikalinos TA, Lau J: A summary of the agency for healthcare research and quality’s evidence report on breastfeeding in developed countries. Breastfeed Med 2009, 4(Suppl 1):S17–30. 6. Drugs and lactation database (LactMed). united states national library of medicine; 2013. http://toxnet.nlm.nih.gov/cgi-bin/sis/htmlgen?LACT; accessed on April 7th, 2013. 7. Ward RM, Bates BA, Benitz WE, Burchfield DJ, Ring JC, American Academy of Pediatrics Committee on Drugs, et al: Transfer of drugs and other chemicals into human milk. Pediatrics 2001, 108(3):776–89. 8. Goodman LS, Hardman JG, Limbird LE, Goodman Gilman A: Goodman & Gilman’s The Pharmacological Basis of Therapeutics. 10th edition. New York: Mc GrawHill; 2001. General Principles, Drugs acting on the Central Nervous System (Drugs Effective in the Theraphy of the Epilepsis Pages 1-112, 521-548. 9. Wesson DR, Camber S, Harkey M, Smith DE: Diazepam and desmetyldiazepam in breast milk. J Psychoactive Drugs 1985, 17(1):55–56. 10. Patrick M, Tilstone WJ, Reavey P: Diazepam and breastfeeding. Lancet 1972, 1(7749):542–3. 11. Fisher JB, Edgren BE, Mammel MC, Coleman JM: Neonatal apnea associated with maternal clonazepam therapy: a case report. Obstet Gynecol 1985, 66(3 Suppl):34S–35S. 12. Birnbaum CS, Cohen LS, Bailey JW, Grush LR, Robertson LM, et al: Serum concentrations of antidepressants and benzodiazepines in nursing infants: a case series. Pediatrics 1999, 104(1):e11. 13. Meyer FP, Quednow B, Potrakfki A, Walther H, et al: Pharmacokinetics of anticonvulsants in the perinatal period. Zantralbl GyneKol 1988, 110(19):1195–205. 14. Kaneko S, Sato T, Suzuki K: The levels of anticonvulsants in breast milk. Br J Clin Pharmacol 1979, 7(6):624–7. 15. Shimoyama R, Ohkubo T, Sugawara K: Monitoring of carbamazepine and carbamazepine 10,11-epoxide in breast milk and plasma by high- performance liquid chromatography. Ann Clin Biochem 2000, 37(Pt 2):210–5. 16. Zhao M, Yang L, Wei Y, Xiong X, Zhou Y, et al: A case report of monitoring on carbamazepine in breastfeeding woman. Beeijing Da Xue Xue Bao 2010, 42(5):602–3. 17. Froescher W, Eichelbaum M, Niesen M, Dietrich K, Rausch P: Carbamazepine levels in breast milk. Ther Drug Monit 1984, 6(3):266–71. 18. Wisner KL, Perel J: Serum levels of valproate and carbamazepine in breastfeeding mother-infant pairs. J Clin Psychopharmacol 1998, 18(2):167–169. 19. Kuhnz W, Jäger-Roman E, Rating D, Deichl A, Kunze J, et al: Carbamazepine and carbamazepine-10,11-epoxide during pregnancy and postnatal period in epileptic mother and their nursed infants: pharmacokinetics and clinical effects. Pediatr Pharmacol (New York) 1983, 3(3–4):199–208. 20. Pynnonen S, Kanto J, Sillanpää M, Erkkola R: Carbamazepine: placental transport, tissue concentration in foetus and newborn and level in milk. Acta Pharmacol Toxicol (Copenh). 1977, 41(3):244–253. Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 10 of 11 http://www.ijponline.net/content/39/1/50 21. Merlob P, Mor N, Litwin A: Transient hepatic dysfunction in an infant of an epileptic mother treated with carbamazepine during pregnancy and breastfeeding. Ann Pharmacother 1992, 26(12):1563–5. 22. Frey B, Braegger CP, Ghelfi D: Neonatal cholestatic hepatitis from carbamazepine exposure during pregnancy and breast feeding. Ann Pharmacother 2002, 36(4):644–7. 23. Chaudron LH, Jefferson JW: Mood stabilizers during breastfeeding: a review. J Clin Psychiatry 2000, 61(2):79–90. 24. Meador KJ, Baker GA, Browning N, Clayton-Smith J, Combs-Cantrell DT, et al: NEAD Study Group. Effects of breastfeeding in children of women taking antiepileptic drugs. Neurology 2010, 75(22):1954–60. 25. Frey B, Schubiger G, Musy JP: Transient cholestatic hepatitis in a neonate associated with carbamazepine exposure during pregnancy and breastfeeding. Eur J Pediatr 1990, 150(2):136–8. 26. Kuhnz W, Koch S, Jacob S, Hartmann A, Helge H, et al: Ethosuximide in epileptic women during pregnancy and lactation period. Placental transfer, serum concentration in nursed infants. Br J Clin Pharmacol 1984, 18(5):671–7. 27. Rane A, Tunell R: Ethosuximide in human milk and in plasma of a mother and her nursed infant. Br J Clin Pharmacol 1981, 12(6):855–8. 28. Jürgens TP, Schaefer C, May A: Treatment of cluster headache in pregnancy and lactation. Cephalalgia 2009, 29(4):391–400. 29. Ohman I, Vitols S, Tomson T: Pharmacokinetics of gabapentin during delivery, in the neonatal period, and lactation: does a fetal accumulation occur during pregnancy? Epilepsia 2005, 46(10):1621–4. 30. Kristensen JH, Ilett KF, Hackett LP, Kohan R: Gabapentin and breastfeeding: a case report. J Hum Lact 2006, 22(4):426–8. 31. Madadi P, Ito S: Perinatal exposure to maternal lamotrigine. Canadian Family Physician 2010, 56(11):1132–4. 32. Nordmo E, Aronsen L, Wasland K, Småbrekke L, Vorren S: Severe apnea in an infant exposed to lamotrigine in breast milk. Ann Pharmacother 2009, 43(11):1893–7. 33. Newport DJ, Pennell PB, Calamaras MR, Ritchie JC, Newman M, et al: Lamotrigine in breast milk and nursing infants: determination of exposure. Pediatrics 2008, 122(1):e223–31. 34. Mikati MA, Fayad M, Koleilat M, Mounla N, Hussein R, et al: Efficacy, tolerability, and kinetics of lamotrigine in infants. J Pediatr 2002, 141(1):31–35. 35. Piña-Garza JE, Levisohn P, Gucuyener K, Mikati MA, Warnock CR, et al: Adjunctive lamotrigine for partial seizures in patients aged 1 to 24 months. Neurology 2008, 70(22 Pt 2):2099–108. 36. Liporace J, Kao A, D’Abreu A: Concerns regarding lamotrigine and breastfeeding. Epilepsy Behav 2004, 5(1):102–5. 37. Johannessen SI, Helde G, Brodtkorb E: Levetiracetam concentrations in serum and in breast milk at birth and during lactation. Epilepsia 2005, 46(5):775–7. 38. Tomson T, Palm R, Källén K, Ben-Menachem E, Söderfeldt B, et al: Pharmacokinetics of levetiracetam during pregnancy, delivery, in the neonatal period, and lactation. Epilepsia 2007, 48(6):1111–6. 39. Rauchenzauner M, Kiechl-Kohlendorfer U, Rostasy K, Luef G: Old and new antiepileptic drugs during pregnancy and lactation–report of a case. Epilepsy Behav 2011, 20(4):719–20. 40. Lutz UC, Wiatr G, Gaertner H, Bartels M: Oxcarbazepine treatment during breastfeeding: a case report. J Clin Psychopharmacol 2007, 27(6):730–2. 41. Taketomo CK, Hodding JH, Kraus DM: Pediatric and Neonatal Dosage Handbook. 19th edition. Lexicomp Drug Reference Handbooks; 2012. Pages 303-307, 413-415, 517-521, 668-669, 777-779, 981-987, 997-1001, 1277-1280, 1344-1347, 1353-1357, 1413-1415, 1642-1646, 1659-1662, 1705-1709, 1735-1737, 1774-1777. 42. Kaneko S, Suzuki K, Sato T, et al: The problems of antiepileptic medication in the neonatal period: is breastfeeding advisable? In Epilepsy, pregnancy and the child. Edited by Janz D, Dam M, Richens A, et al. New York: Raven Press; 1982:343–8. 43. Granstrom ML, Bardy AH, Hiilesmaa VK: Prolonged feeding difficulties of infants of primidone mothers during neonatal period. In preliminary results from the Helsinki study. Edited by Janz D, et al. New York: Epilepsy, pregnancy and the child; 1982:357–358. Raven Press. 44. Schiavetti B, Clavenna A, Fortinguerra F, Miglio D, Bonati M: Psicofarmaci in Allattamento. Roma: Guida pratica basata sulle evidenze. Il Pensiero scientifico editore; 2007. 45. Finch E, Lorber J: Methaemoglobinaemia in the newborn. Probably due to phenytoin excreted in human milk. J Obstet Gynaecol Br Emp 1954, 61(6):833–4. 46. Bar-Oz B, Nulman I, Koren G, Ito S: Anticonvulsants and breastfeeding: a critical review. Paediatr Drugs 2000, 2(2):113–126. 47. Hagg S, Spigset O: Anticonvulsant use during lactation. Drug Saf 2000, 22(6):425–40. 48. Shimoyama R, Ohkubo T, Sugawara K, Ogasawara T, Ozaki T, et al: Monitoring of phenytoin in human breast milk, maternal plasma and cord blood plasma by solid-phase extraction and liquid chromatography. J Pharm Biomed Anal 1998, 17(4–5):863–9. 49. Harden CL, Pennel PB, Koppel BS, Hovinga CA, Gidal B, et al: Practice parameter update: management issues for women with epilepsy- focus on pregnancy (an evidence-based review): vitamn K, folic acid, blood levels, and breastfeeding: report of the quality standards subcommittee and therapeutics and technology assesment subcomittee of the American academy of neurology and American epilepsy society. Neurology 2009, 73(2):142–9. 50. Mirkin BL: Diphenylhydantoin: placental transport, fetal localization, neonatal metabolism, and possible teratogenic effects. J Pediatr 1971, 78(2):329–37. 51. Durkin B, Page C, Glass P: Pregabalin for the treatment of postsurgical pain. Expert Opin Pharmacother 2010, 11(16):2751–8. 52. Patsalos PN, Berry DJ, Bourgeois BF, Cloyd JC, Glauser TA, et al: Antiepileptic drugs-best practices guidelines for therapeutic drug monitoring: a position paper by the subcommission on therapeutic drug monitoring, ILAE Commission on Therapeutic Strategies. Epilepsia 2008, 49(7):1239–76. 53. Tomson T: Gender aspects of phamacokinetics of new and old AEDs: pregnancy and brest-feeding. Ther Drug Monit 2005, 27(6):718–21. 54. Espir MLE, Benton P, Will E, et al: Sodium valproate (Epilim) - some clinical and pharmacological aspects. In Clinical and pharmacological aspects of sodium valproate in the treatment of epilepsy: proceedings of a symposium. Edited by Legg NJ. Available from P.O Box 14, Tunbridge Wells, Kent TN30BA; 1976:145–151. 55. Ohman I, Vitols S, Luef G, Söderfeldt B, Tomson T: Topiramate kinetics during delivery, lactation and in the neonate: preliminary observations. Epilepsia 2002, 43(10):1157–60. 56. Gentile S: Topiramate in pregnancy and breastfeeding. Clin Drug Investig 2009, 29(2):139–41. 57. Orny A: Valporic acid in pregnancy: how much are we endangering the embryo and the fetus? Reprod Toxicol 2009, 28(1):1–10. 58. Smith AK, Conneely KN, Newport DJ, Kilaru V, Schroeder JW, et al: Prenatal antiepileptic exposure associates with neonatal DNA methylation differences. Epigenetics 2012, 7(5):458–63. 59. Stahl MM, Neiderud J, Vinge E: Thrombocytopenic purpura and anemia in a breastfed infant whose mother was treated with valproic acid. J Paediatr 1997, 130(6):1001–3. 60. Wallace SJ: A comparative review of the adverse effects of anticonvulsants in children with epilepsy. Drug Saf 1996, 15(6):378–93. 61. Perucca E: Pharmacological and therapeutic properties of valproate: a summary after 35 years of clinical experience. CNS Drugs 2002, 16(10):695–714. 62. Eberhard-Gran M, Eskild A, Opjordsmoen S: Use of Psychotropic Medications in treating mood disorders during lactation: practical recommendations. CNS Drugs 2006, 20(3):187–98. 63. Kawada K, Itoh S, Kusaka T, Isobe K, Ishii M: Pharmacokinetics of zonisamide in perinatal period. Brain Dev 2002, 24(2):95–7. 64. Shimoyama R, Ohkubo T, Sugawara K: Monitoring of zonisamide in human breast milk and maternal plasma by solid-phase extraction HPLC method. Biomed Chromatogr 1999, 13(5):370–2. 65. Klein A: The postpartum period in women with epilepsy. Neurol Clin 2012, 30(3):867–75. 66. Hussainy SY, Dermele N: Knowledge, attitudes and practices of health professionals and women towards medication use in breastfeeding: A review. Int Breastfeed J 2011, 6:11. 67. Crawford PM: Managing epilepsy in women of childbearing age. Drug Saf 2009, 32(4):293–307. 68. Davanzo R, Rubert L, Oretti C: Meta-variability of advice on drugs in the breastfeeding mother: the example of beta-blockers. Arch Dis Child Fetal Neonatal Ed 2008, 93(3):F249–50. 69. Davanzo R, Roncoroni I, Rubert L: Valutazione dei farmaci in corso di allattamento al seno: i corticosteroidi. Medico e Bambino 2007, 26(8):527–530. Davanzo et al. Italian Journal of Pediatrics 2013, 39:50 Page 11 of 11 http://www.ijponline.net/content/39/1/50 70. Davanzo R, Copertino M, De Cunto A, Minen F, Amaddeo A: Antidepressant drugs and breastfeeding: a review of the literature. Breastfeed Med 2011, 6(2):89–98. 71. Hale TW, Hartmann P: The transfer of medications into human milk. In Hale & Hartmann’s Textbook of Human Lactation. Texas: Hale Publishing Amarillo; 2007. Antidepressants and antipsychotics. Pages 535-556. 72. Bang L, Goa K: Oxcarbazepine: a review of its use in children with epilepsy. Paediatr Drugs 2003, 5(8):557–73. 73. Schirm E, Schwagermann MP, Tobi H, de Jong-van den Berg LT: Drug use during breastfeeding. A survey from the Netherlands. Eur J Clin Nutr 2004, 58(2):386–90. 74. Anderson A: Breastfeeding: societal encouragement needed. J Hum Nutr Diet 2003, 16(4):217–8. 75. Sabers A, Tomson T: Managing antiepileptic drugs during pregnancy and lactation. Curr Opin Neurol 2009, 22(2):157–61. 76. Langslet A, Meberg A, Bredesen JE, Lunde PK: Plasma concentrations of diazepam and N-desmethyldiazepam in newborn infants after intravenous, intramuscular, rectal and oral administration. Acta Paediatr Scand 1978, 67(6):699–704. 77. Jain R, Mishra D, Juneja M: Add-on lamotrigine in pediatric epilepsy in India. Indian Pediatr 2011, 48(1):55–8. 78. Sharpe CM, Capparelli EV, Mower A, Farrell MJ, Soldin SJ, et al: A seven-day study of the pharmacokinetics of intravenous levetiracetam in neonates: marked changes in pharmacokinetics occur during the first week of life. Pediatr Res 2012, 72(1):43–9. 79. Jan MM, Zuberi SA, Alsaihati BA: Pregabalin: preliminary experience in intractable childhood epilepsy. Pediatr Neurol 2009, 40(5):347–50. 80. Mahmoud AA, Rizk TM, Mansy AA, Ali JA, Al-Tannir MA: Ineffectiveness of topiramate and levetiracetam in infantile spasms non-responsive to steroids. Open labeled randomized prospective study. Neurosciences (Riyadh) 2013, 18(2):143–6. doi:10.1186/1824-7288-39-50 Cite this article as: Davanzo et al.: Antiepileptic drugs and breastfeeding. Italian Journal of Pediatrics 2013 39:50. Submit your next manuscript to BioMed Central and take full advantage of: • Convenient online submission • Thorough peer review • No space constraints or color figure charges • Immediate publication on acceptance • Inclusion in PubMed, CAS, Scopus and Google Scholar • Research which is freely available for redistribution Submit your manuscript at www.biomedcentral.com/submit Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1824-7288-39-50

Copertino, M., Bua, J., Zanelli, E., Davanzo, R., Dal Bo, S., & Matarazzo, L. (2013). Antiepileptic drugs and breastfeeding. Italian Journal of Pediatrics, 39(1), 1-11. doi:10.1186/1824-7288-39-50

Rigot, J., Le Mapihan, K., Parent, A., Leroy, C., Decanter, C., Yakoub-Agha, I., . . . Dharancy, S. (2015). Immunosuppressive drugs and fertility. Orphanet Journal of Rare Diseases, 10(1), 1-15. doi:10.1186/s13023-015-0332-8

Simon, T. A., Thompson, A., Gandhi, K. K., Hochberg, M. C., & Suissa, S. (2015). Incidence of malignancy in adult patients with rheumatoid arthritis: A meta-analysis. Arthritis Research & Therapy, 17 Retrieved from

https://search-proquest-com.westcoastuniversity.idm.oclc.org/docview/1772399081?accountid=162765

Remember these have to in alphabetic order

Still stressed with your coursework?
Get quality coursework help from an expert!