Assessment and Treatment of Thyroid Disorders in Pregnancy and The Postpartum Period

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Reviews

Assessment and treatment of thyroid


disorders in pregnancy and the
postpartum period
Sun Y. Lee and Elizabeth N. Pearce ✉

Abstract | Thyroid disorders are prevalent in pregnant women. Furthermore, thyroid hormone
has a critical role in fetal development and thyroid dysfunction can adversely affect obstetric
outcomes. Thus, the appropriate management of hyperthyroidism, most commonly caused
by Graves disease, and hypothyroidism, which in iodine sufficient regions is most commonly
caused by Hashimoto thyroiditis, in pregnancy is important for the health of both pregnant
women and their offspring. Gestational transient thyrotoxicosis can also occur during pregnancy
and should be differentiated from Graves disease. Effects of thyroid autoimmunity and
subclinical hypothyroidism in pregnancy remain controversial. Iodine deficiency is the leading
cause of hypo­thyroidism worldwide. Despite global efforts to eradicate iodine deficiency disorders,
pregnant women remain at risk of iodine deficiency due to increased iodine requirements during
gestation. The incidence of thyroid cancer is increasing worldwide, including in young adults.
As such, the diagnosis of thyroid nodules or thyroid cancer during pregnancy is becoming more
frequent. The evaluation and management of thyroid nodules and thyroid cancer in pregnancy
pose a particular challenge. Postpartum thyroiditis can occur up to 1 year after delivery and must
be differentiated from other forms of thyroid dysfunction, as treatment differs. This Review provides
current evidence and recommendations for the evaluation and management of thyroid disorders in
pregnancy and in the postpartum period.

Thyroid disorders are fairly common in women of repro- and growth of thyroid nodules in pregnancy. The preva-
ductive age. Hashimoto thyroiditis and Graves disease, lence of thyroid nodules in pregnancy has been reported
both autoimmune thyroid disorders, are eight to ten times to be as high as 29%, depending on the iodine status of
more frequent in women than in men and have a peak the population studied6,7. Of note, a worldwide increase
prevalence in early adulthood1. Of note, hypothyroidism has occurred in the incidence of thyroid cancer in young
is estimated to occur in 4% of pregnancies (0.5% overt women in the past few decades. Thyroid cancer is now
and 3.5% subclinical hypothyroidism) and hyperthyroid- the most common type of cancer diagnosed in women
ism in 2.4% of pregnancies (0.6% overt and 1.8% subclin- aged 15–29 years and the second most common type in
ical hyperthyroidism)2. Thyroid hormone is important women aged 30–39 years8–10.
for both maternal and child health. Although the fetal In this Review, we present current evidence regarding
thyroid gland is present and functional by 10–12 weeks the diagnosis and management of thyroid disorders
gestation, it does not mature until 18–20 weeks3. Thus, during pregnancy and the postpartum period. We
the fetus depends on maternal thyroid hormone deliv- note that unless otherwise specified, the term ‘women’
ered via transplacental passage during a critical period refers to ciswomen, as the research highlighted in this
of development in early gestation3. Consequently, mater- Review included only ciswomen. There is currently a
nal thyroid dysfunction can lead to adverse pregnancy lack of available research regarding thyroid disease in
Section of Endocrinology, and child neurodevelopmental outcomes. In addition, pregnancy in transmen and non-​binary pregnant people.
Diabetes, and Nutrition, in the postpartum period, ~5% of women might expe-
Boston University School of
Medicine, Boston, MA, USA.
rience transient thyroid dysfunction from postpartum Thyroid dysfunction in pregnancy
✉e-​mail: elizabeth.pearce@ thyroiditis within 12 months of delivery4. Assessment of thyroid hormone levels in pregnancy
bmc.org Human chorionic gonadotropin (hCG), a hormone During pregnancy, the weakly thyrotrophic effect of
https://doi.org/10.1038/ produced by the placenta during pregnancy, has a weakly hCG on the thyroid gland increases thyroid hormone
s41574-021-00604-​z thyrotrophic effect5 and can promote the development production (Fig. 1), which in turn decreases thyroid

158 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

Key points
states such as pregnancy15. In 2018, Gronowski argued
that the decrease in serum levels of free T4 measured by
• Serum levels of thyroid stimulating hormone might be lower in pregnant women immunoassay seen in the second and third trimesters
during early gestation than levels outside the pregnancy setting, due to increased of pregnancy might be physiological16. Furthermore, a
production of thyroid hormone and stimulation from high levels of human chorionic 2021 study in about 300 euthyroid pregnant Spanish
gonadotropin.
women showed that serum levels of free T4 (measured
• Levothyroxine treatment for maternal subclinical hypothyroidism in pregnancy by two different immunoassays) decreased in the second
remains controversial because currently available studies have not shown a clear
and third trimesters17. Whether the decrease in free T4
benefit of treatment on obstetric or child neurodevelopmental outcomes.
observed in the second and third trimesters is actually
• Treatment of Graves disease in pregnancy requires careful consideration of the
physiological remains to be confirmed. Although levels
adverse effects of uncontrolled hyperthyroidism, antithyroid drugs and overtreatment
of hyperthyroidism on pregnancy outcomes.
of free T4 measured by immunoassays correlated well
with levels of free T4 measured by liquid chromatogra-
• Women who are planning to be pregnant, are pregnant or are breastfeeding should
take a daily oral supplement containing 150 µg of iodine to prevent adverse effects of
phy with tandem mass spectrometry (LC–MS–MS) in
iodine deficiency. the first trimester, the reference ranges were not inter-
• Treatment of differentiated thyroid cancer diagnosed in pregnancy can be safely
changeable at any time point and the immunoassay
deferred until after pregnancy with close monitoring for any tumour size increase or results were not correlated with LC–MS–MS results in
spread during pregnancy. the second and third trimesters. Serum levels of free T4
• Postpartum thyroiditis is a destructive thyroiditis that typically presents with measured by LC–MS–MS remain the gold standard for
thyrotoxicosis followed by transient hypothyroidism up to the first 12 months after assessing levels of free thyroid hormone, but this tech-
childbirth. nique is cumbersome and expensive. Although the mod-
ern immunoassays for free T4 measurements might be
more reliable than older immunoassays in pregnancy,
stimulating hormone (TSH) secretion from the pitui- the pregnancy-​specific, trimester-​specific reference
tary gland via negative feedback11. Thus, in early preg- ranges for each assay are needed. If a reliable assay for
nancy when levels of hCG are high, serum levels of free T4 or the free T4 index is not available, levels of total
TSH tend to shift downwards compared with those T4 or total T3 can be used as surrogates for the measure-
of non-​pregnant adults12. Serum levels of TSH typically ment of free hormone. Of note, between 7 and 16 weeks
increase slightly after the first trimester, largely due to a gestation, the non-​pregnancy assay reference ranges for
decrease in serum levels of hCG11. The levels of thyroid total T4 or T3 can be increased by 5% per week to account
hormone in pregnancy can differ by race and/or eth- for rises in serum levels of TBG. When serum levels of
nicity, and iodine status of populations. Therefore, the TBG stabilize beyond 16 weeks gestation, 150% of the
American Thyroid Association (ATA) and the American upper limit of non-​pregnant reference ranges for total
College of Obstetricians and Gynecologists (ACOG) T4 and T3 levels can be used18.
recommend using population-​specific, assay-​specific
and trimester-​specific ranges for thyroid function tests Hypothyroidism in pregnancy
in pregnant women6,13. If such ranges are not available, Overt hypothyroidism and obstetric outcomes. The
a TSH reference range of 0.1–4.0 mIU/l can be used adverse obstetric and perinatal effects of maternal overt
in early gestation (for context, the reference range for hyperthyroidism have been well documented. For exam-
non-​pregnant adults is usually 0.5–4.5 mIU/l)6,13. ple, increased rates of gestational hypertension (that is
Serum TSH should be used as the initial screening eclampsia, pre-​eclampsia and pregnancy-​induced hyper-
test for thyroid dysfunction in pregnancy, as TSH is the tension), placental abruption, postpartum haemorrhage,
most sensitive marker for primary thyroid dysfunction. preterm delivery, low birthweight and fetal death were
In addition, serum levels of free T4 can be used to distin- initially reported in women with overt hypothyroid-
guish between overt and subclinical thyroid dysfunction. ism and their infants in the 1980s to the 1990s19,20.
Overt hypothyroidism is defined by high serum levels More recently, a 2013 US national cohort study includ-
of TSH with low levels of free T4, whereas in subclinical ing 223,512 pregnant women similarly showed that
hypothyroidism serum levels of TSH are elevated but untreated maternal overt hypothyroidism increased the
levels of free T4 are normal. Similarly, women with overt risks of pre-​eclampsia, gestational diabetes mellitus, pre-
hyperthyroidism have low serum levels of TSH with ele- term birth, caesarean section and infant intensive care
vated levels of free T4 and those with subclinical hyper- unit admission21.
thyroidism have low serum levels of TSH with normal
levels of free T4. Overt hypothyroidism and child neurodevelopmental
Accurate measurement of serum levels of free T4 outcomes. Thyroid hormone affects neuronal migra-
is challenging in pregnant women. High levels of oes- tion, connection, myelination and synaptogenesis and
trogen in pregnancy cause a twofold increase in serum is thus essential for normal brain development22,23.
levels of thyroxine-​binding globulin (TBG)14. Indirect Thyroid hormone receptors are expressed during
free T4 immunoassays can be confounded by high TBG, cortex development24,25. The adverse effects of maternal
which can lead to falsely low results, especially in the thyroid hypofunction in child neurodevelopment were
second and third trimesters of pregnancy. The free T4 first reported in a series of studies in the 1970s26,27. In a
index, which utilizes assessment of levels of total T4 subsequent seminal study published in 1999, 62 children
and an index of binding to calculate free T4, might be born to hypothyroid women (median TSH 13.2 mIU/l)
more accurate than indirect immunoassays in high TBG had an average IQ that was four points lower at 7–9 years

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 159

0123456789();:
Reviews

of age than that of 124 children born to euthyroid high birthweight29–31. Furthermore, a meta-​analysis
women (median TSH 1.4 mIU/l)28. Furthermore, when and systematic review of ten cohort studies including
14 women with hypothyroidism who were treated with 48,684 women (which included some of the obser-
levothyroxine during pregnancy were excluded from the vational studies discussed in the previous sentence)
analysis, the difference in IQ was seven points. similarly showed no significant associations between
maternal subclinical hypothyroidism or isolated
Subclinical hypothyroidism, hypothyroxinaemia and hypothyroxinaemia and preterm birth32.
obstetric outcomes. The effects of maternal subclinical On the other hand, multiple other observational
hypothyroidism and isolated hypothyroxinaemia (that cohort studies have found associations between mater-
is, low serum levels of free T4 with normal levels of TSH) nal subclinical hypothyroidism and pregnancy loss
on obstetric outcomes have been debated. For example, or miscarriage33,34, prematurity or preterm birth35,36,
several observational studies have found no associa- placental abruption35 and neonatal intensive care unit
tions between maternal subclinical hypothyroidism and admission and respiratory distress syndrome in infants35.
outcomes including miscarriage, gestational diabetes A meta-​analysis of 18 cohort studies including almost
mellitus, gestational hypertension, pre-​eclampsia, pre- 4,000 women found that compared with euthyroidism,
term labour, premature delivery, low birthweight and maternal subclinical hyperthyroidism is associated with
increased risks of pregnancy loss (relative risk (RR) 2.01,
95% CI 1.66–2.44), placental abruption (RR 2.14, 95% CI
1.23–3.70), premature rupture of membranes (RR 1.43,
95% CI 1.04–1.95) and neonatal death (RR 2.58, 95% CI
1.41–4.73)37. In a 2020 retrospective study in 8,413 preg-
nant women, offspring of women with subclinical hypo-
thyroidism had increased risks of prematurity (RR 2.15,
95% CI 1.14–4.03) and neonatal respiratory distress
syndrome (RR 2.8, 95% CI 1.01–7.78) compared with
Iodine
Increased demand Increased maternal the risks in euthyroid women. Furthermore, the risks
for thyroid hormone Iodine Dietary dietary iodine of these adverse outcomes were even higher if subclin-
iodine requirements in
Thyrotropic hCG intake ical hypothyroidism was present in the first trimester38.
pregnancy
stimulation by hCG
Thyroid
In addition, an individual-​participant level meta-​analysis
of data published in 2019 from 19 cohort studies (includ-
ing 47,045 pregnant women) similarly showed increased
risks of preterm birth in women with subclinical hypo-
thyroidism (OR 2.9, 95% CI 1.01–1.64) and isolated
hypothyroxinaemia (OR 1.46, 95% CI 1.12–1.90)39.
Oestrogen-mediated
Iodine increase in TBG in These conflicting results regarding the effects of
hCG systemic circulation maternal subclinical hypothyroidism across cohort
studies might be due to variability in timing of TSH
measurements, differences in TSH cut-​off values used
for the diagnosis of subclinical hypothyroidism, and
Increased renal
iodine clearance variability in assessment of thyroid peroxidase (TPO)
Iodine antibody status. However, evidence is mounting that
Iodine
Kidney maternal subclinical hypothyroidism is associated with
Iodine miscarriage and preterm birth. The effects of isolated
Fetal thyroid maternal hypothyroxinaemia on pregnancy outcomes
hCG hormone
production remain unclear40.
Type 3 iodothyronine
deiodinase Subclinical hypothyroidism, hypothyroxinaemia and
child neurodevelopmental outcomes. Mild maternal
thyroid hypofunction has been shown to have a dele-
terious effect on child neurodevelopmental outcomes
Fig. 1 | Factors increasing maternal dietary iodine requirement in pregnancy. in multiple cohorts. In a study of 3,839 Dutch mother–
Maternal and fetal factors leading to increased iodine requirements in pregnancy are child pairs, both low and high maternal levels of free T4
described. During pregnancy, there is a ~50% increase in demand for thyroid hormone, in early pregnancy were associated with lower child IQ
which requires an additional 50–100 μg daily dietary iodine intake. Maternal thyroid and higher likelihood of an IQ of <85 at 6–8 years of age
hormone production is in part increased by the thyrotropic action of human chorionic compared with normal levels of maternal free T4 (ref.41).
gonadotropin (hCG), a hormone produced by the placenta. Oestrogen mediates a This association remained statistically significant even
twofold increase in circulating levels of thyroxine-​binding globulin (TBG), which binds
when overt hypothyroidism and overt hyperthyroid-
thyroid hormones in the maternal circulation, leading to a relative decrease in levels of
active free T4, which further promotes an increase in thyroid hormone production. ism were excluded. Of note, in this analysis, maternal
Furthermore, placental type 3 iodothyronine deiodinase deactivates T4 in the circulation serum TSH values did not predict child IQ. Brain MRI
to reverse T3. Increased maternal renal clearance of iodine (by 30–50%) also occurs due to has shown alterations in child neuroanatomy associated
the increase in maternal blood volume. Finally, iodine is transferred from the maternal with either low or high maternal thyroid function; the
systemic circulation (red box) to the fetus for fetal thyroid hormone production. strongest effect was observed when thyroid function was

160 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

measured at 8–14 weeks gestation. These findings sug- hypothyroidism or hypothyroxinaemia who were treated
gest that 8–14 weeks gestation is a critical period for the with 150 μg daily levothyroxine were compared with 404
effects of thyroid hormone on fetal brain development42. untreated pregnant women49. Treatment did not result
Of note, a longitudinal study in 4,615 UK mother–child in significant differences in mean child IQ or the pro-
pairs did not show significant associations between portion of children with an IQ of <85 at 3 years of age.
maternal levels of TSH or free T4 levels measured in the A follow-​up study of these children at 9 years of age again
first trimester and the standardized test scores of chil- showed no difference in IQ between children of treated
dren from the age of 54 months through to 15 years43. and children of untreated mothers50. Of note, the lev-
However, a 2018 meta-​analysis of 26 studies found sig- othyroxine dose employed in this study was high, poten-
nificant associations between maternal subclinical hypo- tially resulting in over-​treatment, although a secondary
thyroidism or hypothyroxinaemia and measures of child analysis including only those with high free T4 had sim-
intellectual disability such as low IQ, language delay or ilar results. A major limitation of both of these trials is
global developmental delay (OR 2.14, 95% CI 1.2–3.83, that the intervention was initiated well into the second
for subclinical hypothyroidism and OR 1.63, 95% CI trimester (median gestational age at randomization was
1.03–2.56, for hypothyroxinaemia, both compared with 16.7 weeks in the US study48 and 12.3 weeks in CATS)49,50.
children of euthyroid women)44. Although questions As the fetus depends entirely on maternal thyroid hor-
remain to be answered, observational data strongly sug- mone during the first trimester, both trials may have
gest an association between maternal levels of free T4 missed the critical window for assessing the potential
in early pregnancy and child developmental outcomes. benefits of maternal levothyroxine supplementation.
Overall, limited data from RCTs suggest that levothy-
Levothyroxine treatment for subclinical hypothyroidism roxine treatment in pregnant women with subclinical
and hypothyroxinaemia. Only a few randomized hypothyroidism, especially those with TSH >4 mIU/l,
controlled trials (RCTs) have assessed the benefit of might decrease risks of preterm delivery and pregnancy
levothyroxine treatment for maternal subclinical hypo- loss when initiated in early pregnancy. Levothyroxine
thyroidism and/or hypothyroxinaemia45. A RCT of treatment has not shown any notable benefit for child
366 TPO antibody-​negative pregnant Iranian women neurodevelopmental outcomes, but trials have been
with TSH 2.5–10 mIU/l assessed the effects of levothy- limited by initiation of therapy fairly late in gestation.
roxine treatment versus no therapy (randomization Additional RCTs are needed to determine the effect
at 11–12 weeks gestation) on preterm birth46. Overall, of levothyroxine treatment on adverse obstetric and
no difference was observed in the prevalence of pre- child neurodevelopment outcomes, when treatment is
term delivery between treated and untreated women. initiated in the first trimester.
However, in a sensitivity analysis in which the sample was
restricted to women with a baseline TSH of 4–10 mIU/l, Thyroid autoimmunity. Thyroid autoimmunity can
levothyroxine treatment did decrease preterm delivery occur in the presence or absence of thyroid dysfunction.
risk (7.3% versus 19%, P = 0.04). A 2019 meta-​analysis In regions with adequate iodine nutrition, autoimmune
of seven cohort studies and six RCTs (including women thyroiditis (Hashimoto thyroiditis) is the most common
undergoing assisted reproductive technology) showed a cause of hypothyroidism. TPO and thyroglobulin
17% decrease in risk of pregnancy loss in women with antibodies are frequently elevated in autoimmune
subclinical hypothyroidism who received levothyroxine thyroiditis. The prevalence of TPO and thyroglobulin
treatment (95% CI 0.72–0.97), although heterogeneity antibody positivity (indicators of thyroid autoimmunity)
was noted in the timing of intervention and the TSH in pregnant women is estimated to be 5–14% and 3–18%,
cut-​off values used47. respectively51. Underlying thyroid autoimmunity seems
A pair of RCTs in the USA randomized 677 preg- to have an additive or synergistic effect on miscarriage34
nant women with subclinical hypothyroidism (defined and prematurity36 risk, when combined with maternal
as TSH ≥4 mIU/l with normal free T4) and 526 women subclinical hypothyroidism. Thyroid autoimmunity
with hypothyroxinaemia (defined as free T4 <0.86 ng/dl) itself might also have adverse effects in pregnancy, even
to either levothyroxine or placebo treatment at a in the absence of thyroid dysfunction. Meta-​analyses of
median gestational age of 16.7 weeks48. No significant pregnancy in euthyroid women have shown a twofold
differences were noted in child neurodevelopmental to fourfold increase in the risk of recurrent miscarriage
scores at up to 60 months of age between children of the associated with thyroid autoimmunity52,53. Similarly,
levothyroxine-​treated women and the placebo-​treated euthyroid pregnant women with TPO antibody pos-
women. In addition, levothyroxine treatment for sub- itivity have been shown to have twofold to threefold
clinical hypothyroidism or hypothyroxinaemia did not increased risks of preterm birth34,39,52. The adverse effects
alter pregnancy outcomes, including preterm birth, of TPO antibody positivity might be due, at least in part,
gestational hypertension, pre-​eclampsia, gestational to increased risk of hypothyroidism as pregnancy
diabetes mellitus, stillbirth or miscarriage, low birth- progresses54. In a Dutch cohort study, TPO antibody-​
weight or respiratory distress syndrome. In this study, positive women had a higher risk of premature deliv-
levels of TSH were monitored throughout pregnancy and ery than TPO antibody-​negative women; however, this
the levothyroxine doses in the treatment groups were effect was negated if the TPO antibody-​positive women
adjusted for a goal TSH of 0.1–2.5 mIU/l. In another showed an appropriate increase in thyroid hormone
RCT, the Controlled Antenatal Thyroid Screening synthesis in response to hCG55. Of note, TPO antibody
Study (CATS), 390 pregnant women with subclinical positivity could potentially just be a marker for other

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 161

0123456789();:
Reviews

forms of autoimmunity and the observed increased rates and meta-​analysis of six RCTs assessing the effect of lev-
of pregnancy loss in TPO antibody-​positive women othyroxine treatment in euthyroid women with thyroid
could be mediated by the presence of non-​organ-​specific autoimmunity did not find any statistically significant
autoantibodies rather than by thyroid status51. differences in the relative risks of pregnancy achieved,
A few RCTs have assessed the benefits of levothy- miscarriage, preterm delivery or live birth60.
roxine treatment in euthyroid TPO antibody-​positive
pregnant women. A RCT of 393 euthyroid pregnant Current recommendations for screening for hypothyroid-
Italian women with positive TPO antibody titres found ism in pregnancy. There is general agreement that any
no significant differences in the rates of miscarriage and pregnant woman with symptoms and signs suggestive of
preterm delivery between levothyroxine-​t reated overt hypothyroidism should have her thyroid function
and untreated women. However, 49% of women ran- evaluated, although many symptoms might overlap the
domized to the no-​treatment group were then treated normal effects of pregnancy. The presence of underly-
with levothyroxine owing to a serum level of TSH of ing risk factors for thyroid dysfunction (Box 1) could be
≥3 mIU/l occurring in the second or third trimester56. helpful in determining who should be tested. However,
Both the TPO antibody-​positive levothyroxine-​treated universal screening for thyroid dysfunction in asympto-
and untreated groups had higher rates of miscarriage and matic pregnant women is controversial. Such universal
preterm delivery than the TPO antibody-​negative euthy- screening strategies would detect overt hypothyroidism,
roid women. Another RCT in 131 TPO antibody-​positive for which there is a clear treatment benefit, in about 0.5%
women showed a decrease in preterm delivery risk in of pregnant women. However, universal screening would
levothyroxine-​treated women compared with untreated also uncover far more cases of subclinical hypothyroid-
women (RR 0.3, 95% CI 0.1–0.85), but only in those with ism, in about 3.5% of pregnant women, for which the
a baseline serum TSH of >4 mIU/l57. benefits of treatment remain uncertain2. Therefore, there
Other studies have explored the impact of levothy- is currently no consensus regarding routine screening
roxine treatment initiated before conception. The for thyroid dysfunction in pregnant women.
Pregnancy Outcomes Study in Euthyroid Women The ATA and European Thyroid Association (ETA)
with Thyroid Autoimmunity after Levothyroxine both state that insufficient evidence exists to recom-
trial assessed the benefit of levothyroxine treatment mend for or against universal TSH screening at the first
in 565 Chinese TPO-​positive euthyroid women who trimester antenatal visit6,61. By contrast, ACOG and the
were undergoing in vitro fertilization58. Levothyroxine American Society of Reproductive Medicine recom-
treatment, initiated before conception and continued mend against universal screening for thyroid disease
through delivery, did not have a significant effect on in pregnant women13,62. Of note, the ATA does rec-
rates of miscarriage or live birth. Similarly, the Thyroid ommend aggressive case-​finding strategies to identify
Antibodies and Levothyroxine trial in 952 euthyroid pregnant women who are at increased risk for thyroid
UK women with positive TPO antibody titres and a hypofunction (Box 1). A retrospective single-​centre study
history of miscarriage or infertility showed no differ- has suggested that these case-​finding criteria identify
ences in the rates of live births, pregnancy loss, preterm fewer pregnant women with subclinical hypothyroid-
birth or neonatal outcomes when levothyroxine was ism but a comparable proportion with overt hypothy-
initiated before conception59. A 2021 systematic review roidism, compared with a universal screening strategy63.
On the other hand, several observational studies and a
meta-​analysis have suggested that such a case-​finding
Box 1 | Risk factors for developing thyroid
approach, even based on the recommended criteria by
hypofunction in pregnancy
the ATA, might miss up to 40–50% of pregnant women
The risk factors for developing thyroid hypofunction are with thyroid dysfunction in whom treatment would be
reviewed in the ATA guidelines6 and can be summarized recommended64–66.
as follows:
• History of thyroid dysfunction
Recommendations for treatment of hypothyroidism in
• Symptoms or signs of thyroid dysfunction pregnancy. Overt hypothyroidism should be treated to
• Presence of a goitre prevent adverse effects on pregnancy and child devel-
• Known thyroid antibody positivity opmental outcomes6,13. Regarding maternal subclinical
• Age >30 years hypothyroidism, the 2017 ATA guidelines recommend
• History of type 1 diabetes mellitus or other autoimmune utilizing TPO antibody status along with serum levels
disease of TSH to guide treatment decisions (Table 1). Neither
• History of head or neck radiation or prior thyroid surgery ATA nor ACOG currently recommends levothyroxine
• Family history of autoimmune thyroid disease or treatment for isolated hypothyroxinaemia6,13. However,
thyroid dysfunction ATA and ACOG do have differences in their recom-
• Presence of morbid obesity (defined as BMI ≥40 kg/m2) mendations due to differing interpretations of currently
• Use of amiodarone, lithium or administration of available evidence. For example, ACOG does not rec-
intravenous contrast agent within the past 3 months ommend treatment of subclinical hypothyroidism in
• Two or more prior pregnancies pregnancy due to the lack of clear benefit of levothyrox-
ine treatment for child neurodevelopmental outcomes.
• History of pregnancy loss, preterm delivery or infertility
By contrast, the ATA recommends treatment of subclin-
• Residing in area of moderate to severe iodine deficiency
ical hypothyroidism with levothyroxine, especially in the

162 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

Table 1 | Current US recommendations for treatment of thyroid hypofunction in pregnancy


Laboratory data ATA guidelines6 ACOG guidelines13
TPO antibody-​negative and TSH >10 mIU/l Treat with levothyroxine Treat with levothyroxine only
if free T4 is low
TPO antibody-​positive and TSH greater than the Treat with levothyroxine Treat with levothyroxine only
pregnancy-​specific range (or >4 mIU/l) if free T4 is low
TPO antibody-​positive and TSH >2.5 mIU/l but less than the Consider treatment with No treatment
pregnancy-​specific reference range (or <4 mIU/l) levothyroxine
TPO antibody-​negative and TSH greater than the Consider treatment with Treat with levothyroxine only
pregnancy-​specific range (or >4 mIU/l) but <10 mIU/l levothyroxine if free T4 is low
Isolated hypothyroxinaemia No treatment Not discussed
ACOG, American College of Obstetricians and Gynecologists; ATA, American Thyroid Association; TPO, thyroid peroxidase; TSH,
thyroid stimulating hormone.

presence of thyroid autoimmunity. This recommenda- also associated with hyperemesis75,76, GTT is usually
tion is based on the mounting observational evidence associated with symptoms of nausea and/or vomiting.
regarding increased risk of adverse obstetric outcomes in In both GTT and Graves disease, laboratory testing will
pregnant women with subclinical hypothyroidism, and demonstrate suppressed serum levels of TSH and ele-
data from small trials suggesting a treatment benefit. vated levels of thyroid hormone. Patients with Graves
Given the currently available evidence, and the safety disease often have an increased ratio of T3 to T4 (≥20:1),
and ease of levothyroxine treatment, the authors agree as preferential production of T3 occurs in Graves disease.
with the treatment of pregnant women with serum By contrast, serum levels of T3 are often lower in women
concentrations of TSH that are greater than pregnancy-​ with GTT in the setting of hyperemesis.
specific reference ranges or >4 mIU/l, regardless of TPO Of note, antibodies specific for the thyrotropin
antibody status. receptor (TRAb) are generally detectable in Graves
During pregnancy, levothyroxine monotherapy disease but are absent in GTT70. TRAb can be assessed
should be used for thyroid hormone replacement6,13. as TSH receptor-​binding immunoglobulin, which
The use of liothyronine, levothyroxine–liothyronine measures both stimulating and blocking antibodies
combination therapy, or desiccated thyroid is not recom- without distinction. Alternatively, thyroid-​stimulating
mended in pregnancy, as liothyronine does not cross the immuno­globulin can be measured, which specifically
blood–brain barrier to the fetal brain67. Consequently, measures stimulating TRAb77. Women with Graves
liothyronine-​c ontaining treatment options might disease typically report thyrotoxic symptoms, such as
normalize maternal serum levels of TSH but result in unintentional weight loss, palpitations, hand tremors
inadequate thyroid hormone availability for the fetus. and heat intolerance, which might have been present
Once levothyroxine therapy is started in a pregnant prior to pregnancy. By contrast, patients with GTT
woman, serum levels of TSH should be measured every rarely have severe thyrotoxic symptoms and they lack
4–6 weeks until mid-​gestation, then once in the second typical signs of Graves disease such as diffuse goitre and
and third trimesters for a goal serum level of TSH of Graves ophthalmopathy. GTT spontaneously resolves as
<2.5 mIU/l6,13. Most women who are on levothyroxine hCG levels decrease after about 10–12 weeks gestation
before conception will require a 25–30% increase in their and the condition is not associated with adverse preg-
levothyroxine dose once pregnant, in order to maintain nancy outcomes78,79. Therefore, patients with GTT can
euthyroidism in the setting of the oestrogen-​induced be treated with supportive measures for nausea, vomit-
increase in serum levels of TBG68,69. Levothyroxine ing and any electrolyte imbalance or volume depletion
can be decreased back to the pre-​pregnancy dose if they have concurrent hyperemesis gravidarum, but
after delivery. these patients do not require antithyroid drug therapy.

Hyperthyroidism in pregnancy Effects of hyperthyroidism on pregnancy outcomes.


Aetiology of hyperthyroidism in pregnancy. Graves dis- Other than GTT, untreated overt hyperthyroidism in
ease and gestational transient thyrotoxicosis (GTT) are pregnancy is associated with maternal and fetal com-
the two most common causes of hyperthyroidism in plications including pre-​eclampsia, pregnancy loss,
pregnancy. The prevalence of new-​onset Graves disease maternal and fetal congestive heart failure, maternal
in pregnant women is estimated to be 0.05%70, whereas thyroid storm, preterm labour, intrauterine growth
that of GTT ranges from 2% to 11%71,72. As the body’s retardation, low birthweight, and fetal and/or neonatal
autoimmune response generally decreases as pregnancy hyperthyroidism19,21,80,81. However, subclinical hyper-
progresses, the incidence of Graves hyperthyroidism thyroidism is not associated with such adverse obstetric
decreases in the second and third trimesters after a slight outcomes82.
increase in the first trimester73,74. Distinguishing between
the two forms of hyperthyroidism seen in pregnancy is Treatment options for Graves hyperthyroidism in
important, as the disease course and treatment options pregnancy. If treatment of overt hyperthyroidism is
differ. GTT is mediated by high serum levels of hCG indicated, antithyroid drugs or thyroidectomy can be
occurring in early pregnancy. As high levels of hCG are considered in pregnancy. Radioactive iodine ablation

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 163

0123456789();:
Reviews

is contraindicated during gestation given its teratogenic treatment, thyroidectomy can be considered. The sec-
effects6. Methimazole and propylthiouracil are antithy- ond trimester is the safest for thyroidectomy, given the
roid drugs available in the USA and Europe. In Europe, potential risk of teratogenicity from anaesthesia in
carbimazole (a drug that is rapidly metabolized to the first trimester and increased risk of preterm delivery
methimazole after ingestion) is also available. All in the third trimester6.
antithyroid drugs are equally effective in treating Graves Adequate thyroid hormone is critical for nor-
disease, but their adverse effects differ. Methimazole, mal fetal development, especially in early pregnancy.
carbimazole and propylthiouracil can increase serum Antithyroid drugs cross the placenta and can affect
levels of liver enzymes, which generally normalize after fetal thyroid hormone production once the fetal thy-
improvement of thyrotoxicosis83 or discontinuation of roid becomes functional at mid-​gestation. Therefore,
medication84. A meta-​analysis of 15 studies including it is important to avoid over-​treatment of hyperthy-
both non-​pregnant adults and pregnant women showed roidism in pregnancy. A Japanese study demonstrated
increased odds of elevated levels of liver enzymes associ- that antithyroid drugs might cause fetal hypothyroid-
ated with propylthiouracil compared with methimazole ism even when maternal thyroid function is normal93.
(OR 2.4, 95% CI 1.16–4.96)85. Propylthiouracil has also Thus, the ATA recommends targeting a maternal level
been associated with rare but potentially fatal fulminant of free T4 that is high–normal to mildly elevated when
hepatic failure, including in pregnant women86,87. Thus, treating hyperthyroidism in pregnancy. Furthermore,
methimazole or carbimazole is generally preferred over thyroid function should be monitored every 2–4 weeks
propylthiouracil in non-​pregnant patients. until a pregnant woman is on a stable dose of antithyroid
The risk of teratogenicity must be considered when drugs, then every 4 weeks thereafter6. Of note, thyroid
choosing antithyroid drugs in pregnancy and the pre- autoimmunity might improve in the second trimester
conception period. Previously, only methimazole or due to pregnancy-​induced immune tolerance, there-
carbimazole, but not propylthiouracil, was thought fore pregnant women might require decreasing doses
to cause birth defects. However, studies from the past of antithyroid drugs. The ‘block-​and-​replace’ strategy,
decade have shown that methimazole, carbimazole and using both antithyroid drugs and levothyroxine, is not
propylthiouracil are all teratogenic88–92. A Danish regis- recommended in pregnancy, as antithyroid drugs cross
try study found that 8–9% of children exposed to these the placenta to a much greater extent than levothyroxine
antithyroid drugs in utero had birth defects compared and this strategy can lead to fetal hypothyroidism.
with about 5–6% of non-​exposed children92. Similarly, a TRAb can cross the placenta and cause fetal hyper-
large Korean study found a 7.2% risk of birth defects with thyroidism and goitre when present in high levels.
antithyroid drug exposure in the first trimester com- Furthermore, TRAb titres can remain elevated for years
pared with 5.9% without antithyroid drug exposure91. after thyroidectomy or radioactive iodine ablation of
Compared with children of non-​exposed women (whose the thyroid gland. Therefore, all pregnant women with
rate of birth defects was 5.94 cases per 1,000 live births), active Graves hyperthyroidism or a history of ablative
the absolute risk of birth defects was higher in children of treatment for Graves disease should have serum TRAb
women exposed to antithyroid drugs during pregnancy: titres measured in the first trimester. If the TRAb titre is
8.81, 17.05 and 16.53 cases per 1,000 live births following more than 3 times the upper limit of the reference range,
exposure during the first trimester to propylthiouracil the measurement should be made again at 18–22 weeks
alone, methimazole alone and both propylthiouracil gestation when the fetal thyroid gland fully matures.
and methimazole, respectively91. A 2020 meta-​analysis When the TRAb titre remains elevated above 3 times
of seven studies of first-​trimester pregnant women also the upper limit of the reference range by mid-​gestation,
showed a higher risk of birth defects in women exposed consultation with maternal–fetal medicine and neona-
to methimazole or carbimazole compared with propylth- tology is warranted to monitor for fetal and neonatal
iouracil (OR 1.29, 95% CI 1.09–1.53)85. Methimazole or hyperthyroidism and goitre.
carbimazole exposure is associated with more severe
birth defects, including aplasia cutis, oesophageal or Preconception counselling and treatment options for
choanal atresia, and ventricular septal defects, whereas women with Graves disease. Women of reproductive
propylthiouracil exposure is associated with less severe age who are diagnosed with Graves disease should be
defects including cystic malformations of the face or counselled regarding plans for pregnancy, given the
neck, hydronephrosis, and cysts in the urinary tract88. potential adverse effects of both antithyroid drugs and
Given these findings, propylthiouracil is the preferred uncontrolled overt hyperthyroidism in pregnancy.
antithyroid drug in the first trimester. If a pregnant Pregnancy should be postponed until women are euthy-
woman continues to require antithyroid drugs beyond roid. Propylthiouracil might be preferred over methima-
16 weeks gestation, it is unclear whether the antithyroid zole in the preconception setting to avoid methimazole
drug should be changed to methimazole, given concern exposure during the period of organogenesis in early
for suboptimal control of hyperthyroidism during dose pregnancy6. In some women, antithyroid drugs can be
titration of the new antithyroid drug. If needed, a prop- discontinued as soon as pregnancy is confirmed, with
ylthiouracil to methimazole dose ratio of 20 to 1 can be close thyroid function monitoring undertaken, in the
used to switch6. hope that hyperthyroidism will not recur until the sec-
In pregnant women with severe hyperthyroidism ond trimester when the potential teratogenic effects of
who cannot tolerate antithyroid drugs, or when hyper- antithyroid drugs are no longer a concern. This strategy
thyroidism is not adequately controlled with medical is appropriate only in carefully selected women who

164 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

require a low dose of antithyroid drugs to maintain Table 2 | Recommended daily iodine intakes by
euthyroidism (up to 5–10 mg of methimazole a day or life stage
100–200 mg of propylthiouracil a day), have been treated
Life stage Iodine intake
with antithyroid drugs for at least 6 months, do not have (μg per day)
Graves ophthalmopathy and have a low or negative
US Institute of Medicine97
TRAb titre6.
If women prefer to avoid antithyroid drug use during Age 0–6 months 110
pregnancy, definitive therapy with thyroidectomy or Age 7–12 months 130
radioactive iodine ablation can be considered before Age 1–8 years 90
conception. Serum levels of TRAb generally decrease
Age 9–13 years 120
within a year after thyroidectomy, but can transiently
increase and remain elevated for years after radioac- Age >13 years 150
tive iodine treatment94. After definitive treatment, it is During pregnancy 220
prudent to avoid conception until women are euthyroid During lactation 290
on a stable dose of levothyroxine. Pregnancy should be
WHO 98
avoided for 6 months after radioactive iodine treatment95.
Age 0–5 years 90
Iodine nutrition in pregnancy Age 6–12 years 120
Iodine requirements in pregnancy Age >12 years 150
Iodine is an essential micronutrient that is required During pregnancy 250
for thyroid hormone production. Iodine requirements
During lactation 250
increase in pregnancy due to several factors. First,
increased demand occurs for maternal thyroid hormone
production owing to the thyrotrophic effects of hCG and and stunted growth112,113. Meta-​analyses have shown that
oestrogen-​mediated increases in TBG. Second, iodine is children born to women with severe iodine deficiency
transferred to the fetus, especially after the fetal thyroid have an IQ lower by 7.4 to 12 points compared with
gland matures at 18–20 weeks gestation. Third, increases children born to iodine-​sufficient women114,115.
in renal clearance of iodine occur, owing to increased
blood volume and glomerular filtration in pregnancy96 Mild–moderate iodine deficiency in pregnancy
(Fig. 1). Thus, increased iodine intake from 150 μg per Many observational studies have found associations
day in non-​pregnant adults to 220–250 μg per day during between mild-​to-​moderate iodine deficiency in preg-
pregnancy is recommended97,98 (Table 2). nancy and risk of miscarriage116, preterm birth117–119,
Spot urinary iodine concentrations (UIC) can be pre-​eclampsia118, low birthweight117,118, and neonatal
used to assess iodine status at the population level99,100. intensive care unit admission119. However, these find-
A median UIC of 150–249 μg/l is consistent with adequate ings have not been universal120. The effects of maternal
iodine status99 (Table 3). Currently, no biomarker exists for mild-​to-​moderate iodine deficiency on child neurode-
chronic iodine nutritional status at the individual level, velopment are unclear. Observational studies have
as UIC only reflects recent (past 6–8 h) iodine intake101. shown lower verbal IQ and reading skills at 8 years of
age121 and lower language skills assessment score at
Epidemiology of iodine deficiency 9 years of age122 in children born to mothers with UIC
Given the increased iodine requirements in gestation, <150 μg/g creatinine or 150 μg/l compared with children
pregnant women might be at risk of iodine deficiency born to mothers with higher UIC levels. Other studies
even when the general population is iodine-​sufficient102. have shown that children born to mothers with maternal
Only one-​third of the 31 European countries with avail- mild-​to-​moderate iodine deficiency show poorer work-
able data report adequate iodine intake in pregnancy103. ing memory at 4 years of age123, lower scores in psycho-
In addition, pregnant women in the USA. are currently motor development assessment at 12 months of age124,
considered mildly iodine-​deficient, with a median UIC lower verbal IQ at 6–12 years of age125, and lower cogni-
of 144 μg/l in assessments performed during 2007–2014 tive, language and motor scores in children at 18 months
(refs104,105). A fourfold increase was noted in the propor- of age126, compared with those born to iodine-​sufficient
tion of US women of childbearing age with UIC <50 μg/l mothers. A 2019 meta-​analysis of individual data from
in assessments performed during 2005–2010 compared three population-​based cohorts including 6,180 mother–
with prior years104,106,107. child pairs also showed associations between maternal
UIC <150 μg/g creatinine or ≥500 μg/g creatinine with
Severe iodine deficiency in pregnancy decreased verbal IQ scores assessed at 1.5–8 years of
Severe iodine deficiency in pregnancy can cause mater- age127. However, other observational studies have failed
nal hypothyroidism. Severe iodine deficiency has been to show notable associations between maternal iodine
associated with increased risks of miscarriage, premature deficiency and child IQ127,128. The few available RCTs
birth and stillbirth108,109. Chronically low iodine intakes assessing iodine supplementation for women with
of <50 μg per day might also lead to maternal and fetal mild-​to-​moderate iodine deficiency in pregnancy have
goitre110,111. Iodine intakes of <25 μg per day in pregnancy not shown a clear benefit for child neurodevelopmen-
could lead to children being born with cretinism, char- tal outcomes. Of note, these studies have been limited
acterized by severe intellectual disability, deaf–mutism by non-​randomized or inadequately powered designs,

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 165

0123456789();:
Reviews

Table 3 | Median urinary iodine concentration ranges for assessment of ranges, serum levels of TSH should be checked in preg-
population-​level iodine status nant women with thyroid nodules to screen for hyper-
functioning nodules, which are unlikely to be malignant.
Group for assessment Median UIC (μg/l) Iodine status
Of note, nuclear thyroid scans using radioactive iodine
School-age children or <20 Severely insufficient tracer are contraindicated in pregnancy. In pregnant
non-pregnant, non-lactating women with thyroid nodules, a serum level of TSH that
adults 20–49 Moderately insufficient
remains suppressed beyond 16 weeks gestation suggests
50–99 Mildly insufficient the presence of a toxic adenoma (that is, a single hyper-
100–299 Adequate functioning thyroid nodule) and fine needle aspiration
(FNA) biopsy can be delayed until nuclear scanning is
≥300 Excessive
feasible after delivery. If toxic adenoma is unlikely and
Pregnant women <150 Insufficient the nodule meets criteria for cytological evaluation, FNA
150–249 Adequate biopsy of thyroid nodule(s) can be safely performed in
pregnancy138. Nodules that are cytologically benign
250–499 More than adequate can be managed in the same way as in non-​pregnant
≥500 Excessive adults. If cytology shows indeterminate results, includ-
Concentration ranges were obtained from ref.99 and ref.159. UIC, urinary iodine concentration. ing atypia of unknown significance or follicular lesion
of unknown significance, FNA biopsy can be repeated
for further evaluation. Molecular testing is not currently
initiation of supplements too late in pregnancy or by recommended in pregnancy, as it is not yet clear how
being conducted in areas of only borderline iodine pregnancy affects the results of molecular markers138. For
deficiency129,130. indeterminate nodules without characteristics sugges-
tive of an aggressive cancer, repeat FNA biopsy can be
Current recommendations for iodine intake deferred until after pregnancy.
Universal salt iodization is considered the most effective
way to improve iodine status at the population level131. Thyroid cancer in pregnancy and postpartum
However, salt iodization has not been mandated in Although the prevalence of thyroid cancer in pregnant
many countries including the USA and much of Europe. women is difficult to estimate, the incidence of thyroid
Therefore, many societies, including the ATA6, the cancer in young adults has been increasing in the last
Endocrine Society132, the US Teratology Society133, two to three decades, especially in young women of
the American Academy of Pediatrics134 and the ETA61, reproductive age8–10. A large retrospective study using
currently recommend that women who are planning to the 1991–1999 California Cancer Registry showed
be pregnant, are pregnant or are breastfeeding should that thyroid cancer was the second most common cancer
take a daily oral supplement containing 150 μg of iodine. in the perinatal period among all cancers recorded in the
registry, with a prevalence of 14.4%. This study showed
Thyroid nodules and cancer in pregnancy 3.3 occurrences of thyroid cancer per 100,000 cancer
Thyroid nodules in pregnancy cases found within 9 months before pregnancy, 3 occur-
The estimated prevalence of thyroid nodules in preg- rences of thyroid cancer per 100,000 cancer cases found
nant women ranges from 3% to 29%, depending on at delivery, and 10.8 occurrences of thyroid cancer per
iodine intake6,7. Older age, increased number of previ- 100,000 cancer cases found within 1 year of delivery139.
ous pregnancies, and low or excessive iodine nutritional An assessment of 595 women diagnosed with thyroid
status are associated with an increased risk of thyroid cancer between 9 months before to 12 months after
nodules in pregnancy135,136. A prospective cohort study delivery from the same cohort in 1991–1995 did not
of 221 pregnant Chinese women found a 15% preva- show any statistically significant differences in maternal
lence of thyroid nodules ≥2 mm in the first trimester, or neonatal outcomes compared with pregnant women
with a median increase of 5 mm in nodule size occur- without thyroid cancer140. In addition, no difference was
ring during pregnancy135. Nodule size might increase observed in survival between women with thyroid can-
during gestation due to the thyrotrophic effects of cer diagnosed in the perinatal period and non-​pregnant
hCG and due to TSH stimulation resulting from deple- women with thyroid cancer.
tion of iodine stores137,138. In addition to the structural Given the overall good prognosis of differentiated
similarity between hCG and TSH, structural simi- thyroid cancer (DTC) in young women and the potential
larities also exist between hCG and TSH receptors5. anxiety associated with a new cancer diagnosis141, FNA
Consequently, high serum levels of hCG can potentially biopsy of many thyroid nodules can be deferred until
stimulate TSH receptors on the thyroid gland; this effect after pregnancy, if the patient prefers. Small studies have
can lead to the development of thyroid nodules or gland shown that delaying diagnosis and treatment of thyroid
hyperplasia, in addition to increased thyroid hormone cancer until after pregnancy does not result in worse
production. outcomes. For example, no differences were observed
Thyroid ultrasonography is the best imaging modal- in long-​term survival142, changes in tumour volume143,144
ity for the evaluation of thyroid nodules found in preg- or development of lymph node metastases143,144 between
nant women. This modality can also be used for patient those who were treated during pregnancy and those who
risk stratification using the same characteristics as in deferred treatment. Therefore, the ATA recommends
non-​pregnant adults. Using pregnancy-​specific reference that most pregnant women with newly diagnosed DTC

166 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

can be monitored conservatively with serial thyroid Those with a history of thyroid cancer
ultrasonography during pregnancy. If evidence is found Having a history of thyroid cancer should not deter
of notable tumour growth by mid-​gestation or develop- patients from pursuing pregnancy. A large Korean
ment of metastatic disease, thyroid surgery can be con- study utilizing national insurance data from 2006–2014
sidered. When required, thyroidectomy is safest in the showed no statistically significant increase in adverse
second trimester138. No evidence is available to support pregnancy outcomes including caesarean section, pre-
the use of suppressive therapy with levothyroxine when term birth, pre-​eclampsia, placental abruption, pla-
thyroid cancer surgery is deferred. cental previa, stillbirth, low birthweight and large for
In a small study, stimulation of TSH and oestrogen gestational age, in women with a history of thyroid
receptors by hCG in pregnancy has been implicated in cancer148. In a study of 235 pregnant women in the USA
increased tumour aggressiveness in DTC145. Two Italian with a history of DTC, disease recurrence and progres-
studies found an increased risk of persistent or recur- sion were monitored using imaging (ultrasonography,
rent disease in patients when DTC is diagnosed during CT scan or radioactive iodine scans), serum levels of
pregnancy145,146. However, both of these studies defined thyroglobulin and thyroglobulin antibody, and clin-
recurrent or persistent disease based on serum levels of ical evaluation within 12 months before and after
thyroglobulin only, without evaluation for structural dis- pregnancy149. Women who had an excellent, indetermi-
ease. Furthermore, neither study showed an increased nate or biochemical incomplete response to treatment
risk of recurrent or persistent disease when surgery per the 2015 ATA guidelines138 prior to pregnancy had
was delayed until after pregnancy. In light of current no evidence of structural recurrence or progression
evidence regarding long-​term outcomes in patients of thyroid cancer on evaluation at 3–12 months after
with DTC diagnosed in pregnancy, we agree with the delivery, although a small number of women had an
current recommendation by the ATA for diagnosis and increase in serum levels of thyroglobulin and thyro­
treatment of DTC diagnosed in pregnancy as described globulin antibody. By contrast, 29% of women who
above. had a structural incomplete response to therapy prior
Studies assessing the potential effects of pregnancy to pregnancy had tumour progression, defined as a
on anaplastic or medullary thyroid cancer are lack- ≥3 mm increase in the size of pre-​existing disease or
ing. However, thyroid surgery in pregnancy should be identification of a new metastasis, at postpartum eval-
strongly considered given the aggressive nature of these uation. Therefore, women with a response to therapy
cancers. classified as excellent or indeterminate according to ATA
If radioactive iodine ablation is indicated as an guidelines prior to pregnancy do not require increased
adjunctive treatment for thyroid cancer, it should be surveillance, but those with an incomplete response
delayed until after delivery. Iodine is taken up into before pregnancy should be closely monitored during
the lactating mammary gland via the sodium iodide gestation. As subclinical hyperthyroidism is not associ-
symporter147. As such, women should be counselled to ated with adverse obstetric outcomes82, serum TSH goals
stop breastfeeding at least 6 weeks to 3 months prior for cancer survivors are the same in pregnant women as
to I131 radioactive iodine treatment in order to prevent in non-​pregnant adults138. Most women will require an
potential adverse effects of radiation on breast tissues increase in their levothyroxine dose by 25–30% during
and breastfed infants95. pregnancy to maintain TSH targets, due to increased
TBG levels.
Radioactive iodine uptake Postpartum thyroiditis
TSH
T4 Epidemiology
20% Postpartum thyroiditis is a destructive thyroiditis that
Increased occurs in 5–8% of women within the first 12 months
after delivery150. The aetiology is thought to be a rebound
of underlying thyroid autoimmunity occurring after the
Levels

immune tolerance induced by pregnancy has ended.


Normal 80%
This mechanism is proposed because postpartum thy-
roiditis is more commonly seen in women who are pos-
itive for TPO antibodies151 or those with a personal or
family history of autoimmune diseases152. Postpartum
Decreased
20% thyroiditis can also occur after miscarriage or pregnancy
loss, as well as in women on levothyroxine replacement
for underlying Hashimoto thyroiditis153.
0 1 2 3 4 5 6 7 8 9 10 11 12
Months after delivery Disease course
Postpartum thyroiditis classically presents as thyrotox-
Fig. 2 | Time course of postpartum thyroiditis. Typical time course in changes in serum
levels of thyroid stimulating hormone (TSH), T4 and radioactive iodine uptake in postpartum icosis within 4–8 weeks of delivery, due to release of
thyroiditis up to 12 months after delivery is depicted. Transient thyrotoxicosis is generally preformed thyroid hormones from an inflamed thyroid
followed by transient hypothyroidism, before full recovery to euthyroidism. However, about gland. This thyrotoxic phase typically lasts for 1–2 months
20% of women with postpartum thyroiditis might develop permanent hypothyroidism and can be followed by transient hypothyroidism last-
(as indicated by the dashed lines). ing 4–6 months until the thyroid gland recovers (Fig. 2).

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 167

0123456789();:
Reviews

However, the disease course can vary; ~25% of women supplementation in pregnancy have shown mixed
only have a thyrotoxic phase and 50% of women only results158. Thus, selenium supplementation for preven-
have a hypothyroid phase150. The majority of women tion or treatment of postpartum thyroiditis is currently
recover completely and return to a euthyroid state within not recommended6.
a year, but up to 50% will develop persistent hypothy-
roidism even after initial recovery154,155. Women who are Conclusions
positive for serum TPO antibodies are at increased risk Thyroid diseases are fairly common in pregnancy and
of developing permanent hypothyroidism after postpar- their management presents specific challenges. Due to
tum thyroiditis155. Postpartum thyroiditis recurs after up physiological changes in pregnancy, pregnancy-​specific
to 70% of subsequent pregnancies150. reference ranges should be used to assess thyroid func-
tion in pregnant women. Given the importance of nor-
Evaluation and treatment mal thyroid function for maternal and child health,
Distinguishing the thyrotoxic phase of postpartum both maternal and fetal outcomes should be consid-
thyroiditis from new or recurrent Graves disease is ered in making management decisions for treatment of
important, as treatment options differ. Antibody test- thyroid dysfunction in pregnancy. Similarly, the evalu-
ing for TRAb is helpful, as it is a highly specific and ation and treatment of thyroid nodules or thyroid can-
sensitive biomarker for Graves disease. By contrast, cer might require special considerations in pregnancy.
serum levels of TPO antibody can be elevated in both Thyroid dysfunction is also frequent in the immediate
postpartum thyroiditis and Graves hyperthyroidism156. postpartum period.
The ratio of T3 to T4 is often higher (>20:1) in Graves Many questions related to the care of pregnant
disease due to preferential T3 production157. Graves dis- women with thyroid disorders remain unanswered.
ease most frequently presents at 6–12 months after Levothyroxine treatment of maternal subclinical hypo-
delivery, compared with a presentation at 1–4 months thyroidism and hypothyroxinaemia initiated in the sec-
after delivery for postpartum thyroiditis152. Of note, I123 ond trimester has not shown any benefit; however, it is
radioactive iodine uptake and scan can be employed not known whether levothyroxine treatment initiated
to differentiate postpartum thyroiditis from Graves earlier in gestation would improve adverse obstetric
disease if needed. Iodine uptake is low in the thyro- and child neurodevelopmental outcomes. The mecha-
toxic phase of postpartum thyroiditis, whereas it is nism for the observed increased rates of pregnancy loss
elevated in Graves disease. However, TRAb measure- in TPO-​positive euthyroid pregnant women remains
ment is usually adequate to differentiate between the unknown; future studies will be needed to determine
two entities. whether there is an effective therapy for these women.
As the thyrotoxic phase of postpartum thyroiditis The safest approach to antithyroid drug use for overt
is transient and results from the release of preformed hyperthyroidism during gestation is also not entirely
hormones, rather than increased thyroid hormone clear. Although iodine supplementation for women in
synthesis, antithyroid drugs are not useful. Notable thy- mildly-​to-​moderately iodine-​deficient regions is now
rotoxic symptoms, such as palpitations or anxiety, can be widely recommended, clinical trials will be needed to
managed with β-​adrenergic agonists that are safe for use determine whether this supplementation improves
during lactation (for example, metoprolol or proprano- child developmental outcomes. Furthermore, the appro-
lol). Levothyroxine can be employed for the hypothyroid priate dose and timing of supplementation remains to
phase if women are symptomatic or are actively trying be elucidated. Regarding thyroid cancer, we do not
to conceive. Around 6–12 months after diagnosis, lev- fully understand the effects of pregnancy on the more
othyroxine treatment can be tapered as hypothyroidism aggressive types, such as anaplastic or medullary thyroid
is usually transient. However, given the increased risk cancer. Finally, it is not known whether any interven-
of recurrent hypothyroidism in women who recover tion could prevent progression of postpartum thyroidi-
from postpartum thyroiditis, these women should have tis to permanent hypothyroidism. Additional studies are
yearly monitoring of TSH and should be monitored needed to answer these questions and to further improve
for recurrent postpartum thyroiditis after subsequent the care of women with thyroid disease during gestation
pregnancies. Selenium supplementation has been and the postpartum period.
explored for its possible anti-​inflammatory effect in
autoimmune thyroiditis51. However, trials of selenium Published online 4 January 2022

1. Taylor, P. N. et al. Global epidemiology of 6. Alexander, E. K. et al. 2017 guidelines of the and mortality in the United States, 1974-2013. JAMA
hyperthyroidism and hypothyroidism. Nat. Rev. American Thyroid Association for the diagnosis 317, 1338–1348 (2017).
Endocrinol. 14, 301–316 (2018). and management of thyroid disease during 9. Kim, J., Gosnell, J. E. & Roman, S. A. Geographic
2. Dong, A. C. & Stagnaro-​Green, A. Differences in pregnancy and the postpartum. Thyroid 27, influences in the global rise of thyroid cancer. Nat. Rev.
diagnostic criteria mask the true prevalence of thyroid 315–389 (2017). Endocrinol. 16, 17–29 (2020).
disease in pregnancy: a systematic review and This paper describes the most current ATA 10. Miller, K. D. et al. Cancer statistics for adolescents and
meta-analysis. Thyroid 29, 278–289 (2019). guideline on diagnosis and management of young adults, 2020. CA Cancer J. Clin. 70, 443–459
3. Burrow, G. N., Fisher, D. A. & Larsen, P. R. Maternal thyroid disease in pregnancy and the postpartum (2020).
and fetal thyroid function. N. Engl. J. Med. 331, period. 11. Glinoer, D. et al. Regulation of maternal thyroid during
1072–1078 (1994). 7. Ollero, M. D. et al. Thyroid function reference pregnancy. J. Clin. Endocrinol. Metab. 71, 276–287
4. Stagnaro-​Green, A. Approach to the patient with values in healthy iodine-​sufficient pregnant women (1990).
postpartum thyroiditis. J. Clin. Endocrinol. Metab. 97, and influence of thyroid nodules on thyrotropin 12. Li, C. et al. Assessment of thyroid function during
334–342 (2012). and free thyroxine values. Thyroid 29, 421–429 first-trimester pregnancy: what is the rational upper
5. Yoshimura, M. & Hershman, J. M. Thyrotropic action (2019). limit of serum TSH during the first trimester in Chinese
of human chorionic gonadotropin. Thyroid 5, 8. Lim, H., Devesa, S. S., Sosa, J. A., Check, D. pregnant women? J. Clin. Endocrinol. Metab. 99,
425–434 (1995). & Kitahara, C. M. Trends in thyroid cancer incidence 73–79 (2014).

168 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

13. [No authors listed] Thyroid disease in pregnancy: 37. Maraka, S. et al. Subclinical hypothyroidism in 53. Xie, J. et al. Effect of antithyroid antibodies on women
ACOG Practice Bulletin, Number 223. Obstet. pregnancy: a systematic review and meta-​analysis. with recurrent miscarriage: a meta-​analysis. Am. J.
Gynecol. 135, e261–e274 (2020). Thyroid 26, 580–590 (2016). Reprod. Immunol. 83, e13238 (2020).
This paper describes the most recent ACOG This systematic review and meta-​analysis 54. Glinoer, D., Riahi, M., Grün, J. P. & Kinthaert, J.
recommendations on the diagnosis and management summarizes results of randomized trials and cohort Risk of subclinical hypothyroidism in pregnant women
of thyroid disease in pregnancy. studies on the effects of subclinical hypothyroidism with asymptomatic autoimmune thyroid disorders.
14. Krassas, G. E., Poppe, K. & Glinoer, D. Thyroid function in pregnancy published up to 2015. J. Clin. Endocrinol. Metab. 79, 197–204 (1994).
and human reproductive health. Endocr. Rev. 31, 38. Lee, S. Y., Cabral, H. J., Aschengrau, A. & Pearce, E. N. 55. Korevaar, T. I. M. et al. Thyroid autoimmunity impairs
702–755 (2010). Associations between maternal thyroid function in the thyroidal response to human chorionic
This is a comprehensive review on the interactions pregnancy and obstetric and perinatal outcomes. gonadotropin: two population-​based prospective
between thyroid function and human reproduction. J. Clin. Endocrinol. Metab. 105, e2015–e2023 (2020). cohort studies. J. Clin. Endocrinol. Metab. 102,
15. Lee, R. H. et al. Free T4 immunoassays are flawed 39. Consortium on Thyroid and Pregnancy — Study Group 69–77 (2017).
during pregnancy. Am. J. Obstet. Gynecol. 200, on Preterm Birth. et al. Association of thyroid function This cohort study showed a suboptimal thyroidal
260.e1–250.e6 (2009). test abnormalities and thyroid autoimmunity with response to hCG stimulation in pregnant women
16. Gronowski, A. M. Evaluation of thyroid function during preterm birth: a systematic review and meta-​analysis. with positive TPO antibody status.
pregnancy: have we taken a wrong turn? Clin. Chem. JAMA 322, 632–641 (2019). 56. Negro, R., Schwartz, A. & Stagnaro-​Green, A. Impact
64, 439–441 (2018). This study assesses risk of preterm birth associated of levothyroxine in miscarriage and preterm delivery
17. Hernández, J. M. et al. Reference intervals of thyroid with maternal thyroid hypofunction and thyroid rates in first trimester thyroid antibody-​positive
function tests assessed by immunoassay and mass autoimmunity using individual data from 47,045 women with TSH less than 2.5 mIU/L. J. Clin.
spectrometry in healthy pregnant women living in pregnant women from 19 cohort studies. Endocrinol. Metab. 101, 3685–3690 (2016).
Catalonia. J. Clin. Med. 10, 2444 (2021). 40. Ramezani Tehrani, F., Nazarpour, S. & 57. Nazarpour, S. et al. Effects of levothyroxine treatment
18. Weeke, J. et al. A longitudinal study of serum TSH, Behboudi-Gandevani, S. Isolated maternal on pregnancy outcomes in pregnant women with
and total and free iodothyronines during normal hypothyroxinemia and adverse pregnancy outcomes: autoimmune thyroid disease. Eur. J. Endocrinol. 176,
pregnancy. Acta Endocrinol. 101, 531–537 (1982). a systematic review. J. Gynecol. Obstet. Hum. Reprod. 253–265 (2017).
19. Davis, L. E., Leveno, K. J. & Cunningham, F. G. 50, 102057 (2021). 58. Wang, H. et al. Effect of levothyroxine on miscarriage
Hypothyroidism complicating pregnancy. Obstet. 41. Korevaar, T. I. M. et al. Association of maternal thyroid among women with normal thyroid function and
Gynecol. 72, 108–112 (1988). function during early pregnancy with offspring IQ and thyroid autoimmunity undergoing in vitro fertilization
20. Leung, A. S., Millar, L. K., Koonings, P. P., Montoro, M. brain morphology in childhood: a population-​based and embryo transfer: a randomized clinical trial. JAMA
& Mestman, J. H. Perinatal outcome in hypothyroid prospective cohort study. Lancet Diabetes Endocrinol. 318, 2190–2198 (2017).
pregnancies. Obstet. Gynecol. 81, 349–353 (1993). 4, 35–43 (2016). 59. Dhillon-​Smith, R. K. et al. Levothyroxine in women
21. Männistö, T. et al. Thyroid diseases and adverse 42. Jansen, T. A. et al. Maternal thyroid function during with thyroid peroxidase antibodies before conception.
pregnancy outcomes in a contemporary US cohort. pregnancy and child brain morphology: a time N. Engl. J. Med. 380, 1316–1325 (2019).
J. Clin. Endocrinol. Metab. 98, 2725–2733 (2013). window-​specific analysis of a prospective cohort. This UK randomized controlled trial assessed
22. Howdeshell, K. L. A model of the development of the Lancet Diabetes Endocrinol. 7, 629–637 (2019). the effects of levothyroxine treatment starting
brain as a construct of the thyroid system. Environ. This study assessed offspring brain morphology preconception for euthyroid women with positive
Health Perspect. 110, 337–348 (2002). by MRI at a median age of 10 years in 1,981 TPO antibodies and a history of miscarriage or
23. Bernal, J., Guadaño-​Ferraz, A. & Morte, B. Perspectives mother–child pairs with maternal thyroid function infertility on live birth rates.
in the study of thyroid hormone action on brain measurements during pregnancy. 60. Lau, L., Benham, J. L., Lemieux, P., Yamamoto, J.
development and function. Thyroid 13, 1005–1012 43. Nelson, S. M. et al. Maternal thyroid function and & Donovan, L. E. Impact of levothyroxine in women
(2003). child educational attainment: prospective cohort with positive thyroid antibodies on pregnancy
24. Moog, N. K. et al. Influence of maternal thyroid study. BMJ 360, k452 (2018). outcomes: a systematic review and meta-​analysis
hormones during gestation on fetal brain development. 44. Thompson, W. et al. Maternal thyroid hormone of randomised controlled trials. BMJ Open 11,
Neuroscience 342, 68–100 (2017). insufficiency during pregnancy and risk of neuro­ e043751 (2021).
25. de Escobar, G. M., Obregón, M. J. & del Rey, F. E. developmental disorders in offspring: a systematic 61. Lazarus, J. et al. 2014 European Thyroid Association
Maternal thyroid hormones early in pregnancy and review and meta-​analysis. Clin. Endocrinol. 88, guidelines for the management of subclinical
fetal brain development. Best. Pract. Res. Clin. 575–584 (2018). hypothyroidism in pregnancy and in children.
Endocrinol. Metab. 18, 225–248 (2004). 45. Yamamoto, J. M., Benham, J. L., Nerenberg, K. A. Eur. Thyroid. J. 3, 76–94 (2014).
26. Man, E. B., Jones, W. S., Holden, R. H. & Mellits, E. D. & Donovan, L. E. Impact of levothyroxine therapy on 62. Practice Committee of the American Society for
Thyroid function in human pregnancy. 8. Retardation obstetric, neonatal and childhood outcomes in women Reproductive Medicine. Subclinical hypothyroidism
of progeny aged 7 years; relationships to maternal age with subclinical hypothyroidism diagnosed in pregnancy: in the infertile female population: a guideline.
and maternal thyroid function. Am. J. Obstet. Gynecol. a systematic review and meta-​analysis of randomised Fertil. Steril. 104, 545–553 (2015).
111, 905–916 (1971). controlled trials. BMJ Open 8, e022837 (2018). 63. Sitoris, G. et al. Screening for thyroid dysfunction in
27. Man, E. B. & Serunian, S. A. Thyroid function in 46. Nazarpour, S. et al. Effects of levothyroxine on pregnancy with targeted high-​risk case finding: can
human pregnancy. IX. Development or retardation pregnant women with subclinical hypothyroidism, it be improved? J. Clin. Endocrinol. Metab. 104,
of 7-year-​old progeny of hypothyroxinemic women. negative for thyroid peroxidase antibodies. J. Clin. 2346–2354 (2019).
Am. J. Obstet. Gynecol. 125, 949 (1976). Endocrinol. Metab. 103, 926–935 (2018). 64. Pop, V. J., Broeren, M. A., Wiersinga, W. M.
28. Haddow, J. E. et al. Maternal thyroid deficiency 47. Nazarpour, S., Ramezani Tehrani, F., Amiri, M., & Stagnaro-​Green, A. Thyroid disease symptoms
during pregnancy and subsequent neuropsychological Bidhendi Yarandi, R. & Azizi, F. Levothyroxine during early pregnancy do not identify women
development of the child. N. Engl. J. Med. 341, treatment and pregnancy outcomes in women with with thyroid hypofunction that should be treated.
549–555 (1999). subclinical hypothyroidism: a systematic review and Clin. Endocrinol. 87, 838–843 (2017).
29. Cleary-​Goldman, J. et al. Maternal thyroid hypofunction meta-​analysis. Arch. Gynecol. Obstet. 300, 805–819 65. Rosario, P. W. Selective screening for thyroid
and pregnancy outcome. Obstet. Gynecol. 112, 85–92 (2019). dysfunction in pregnant women: how often do
(2008). This systematic review and meta-​analysis low-risk women cease to be treated following the
30. Männistö, T. et al. Perinatal outcome of children born summarizes results of 13 randomized controlled new guidelines of the American Thyroid Association?
to mothers with thyroid dysfunction or antibodies: trials and cohort studies on the effects of Arch. Endocrinol. Metab. 62, 641–643 (2018).
a prospective population-​based cohort study. J. Clin. levothyroxine treatment for maternal subclinical 66. Jouyandeh, Z., Hasani-​Ranjbar, S., Qorbani, M.
Endocrinol. Metab. 94, 772–779 (2009). hypothyroidism on pregnancy outcomes. & Larijani, B. Universal screening versus selective
31. Männistö, T. et al. Thyroid dysfunction and 48. Casey, B. M. et al. Treatment of subclinical case-​based screening for thyroid disorders in
autoantibodies during pregnancy as predictive factors hypothyroidism or hypothyroxinemia in pregnancy. pregnancy. Endocrine 48, 116–123 (2015).
of pregnancy complications and maternal morbidity in N. Engl. J. Med. 376, 815–825 (2017). 67. Calvo, R., Obregón, M. J., Ruiz de Oña, C.,
later life. J. Clin. Endocrinol. Metab. 95, 1084–1094 This US randomized controlled trial assessed the Escobar del Rey, F. & Morreale de Escobar, G.
(2010). effects of levothyroxine treatment of maternal Congenital hypothyroidism, as studied in rats.
32. Sheehan, P. M., Nankervis, A., Araujo Júnior, E. thyroid hypofunction on pregnancy outcomes and Crucial role of maternal thyroxine but not of
& Da Silva Costa, F. Maternal thyroid disease and child neurodevelopmental outcomes at a median 3,5,3′-triiodothyronine in the protection of the fetal
preterm birth: systematic review and meta-​analysis. age of 5 years. brain. J. Clin. Invest. 86, 889–899 (1990).
J. Clin. Endocrinol. Metab. 100, 4325–4331 (2015). 49. Lazarus, J. H. et al. Antenatal thyroid screening and 68. Arafah, B. M. Increased need for thyroxine in women
33. Negro, R. et al. Increased pregnancy loss rate in childhood cognitive function. N. Engl. J. Med. 366, with hypothyroidism during estrogen therapy. N. Engl.
thyroid antibody negative women with TSH levels 493–501 (2012). J. Med. 344, 1743–1749 (2001).
between 2.5 and 5.0 in the first trimester of This UK randomized controlled trial assessed the 69. Yassa, L., Marqusee, E., Fawcett, R. & Alexander, E. K.
pregnancy. J. Clin. Endocrinol. Metab. 95, E44–E48 effects of levothyroxine treatment of maternal Thyroid hormone early adjustment in pregnancy
(2010). thyroid hypofunction on child neurodevelopmental (the THERAPY) trial. J. Clin. Endocrinol. Metab. 95,
34. Liu, H. et al. Maternal subclinical hypothyroidism, outcomes at 3 years of age. 3234–3241 (2010).
thyroid autoimmunity, and the risk of miscarriage: 50. Hales, C. et al. Controlled antenatal thyroid screening 70. Cooper, D. S. & Laurberg, P. Hyperthyroidism in
a prospective cohort study. Thyroid 24, 1642–1649 II: effect of treating maternal suboptimal thyroid pregnancy. Lancet Diabetes Endocrinol. 1, 238–249
(2014). function on child cognition. J. Clin. Endocrinol. Metab. (2013).
35. Casey, B. M. et al. Subclinical hypothyroidism and 103, 1583–1591 (2018). 71. Glinoer, D. The regulation of thyroid function in
pregnancy outcomes. Obstet. Gynecol. 105, 239–245 51. De Leo, S. & Pearce, E. N. Autoimmune thyroid pregnancy: pathways of endocrine adaptation from
(2005). disease during pregnancy. Lancet Diabetes Endocrinol. physiology to pathology. Endocr. Rev. 18, 404–433
36. Korevaar, T. I. M. et al. Hypothyroxinemia and 6, 575–586 (2018). (1997).
TPO-antibody positivity are risk factors for premature 52. Thangaratinam, S. et al. Association between thyroid 72. Yeo, C. P. et al. Prevalence of gestational thyrotoxicosis
delivery: the generation R study. J. Clin. Endocrinol. autoantibodies and miscarriage and preterm birth: in Asian women evaluated in the 8th to 14th weeks
Metab. 98, 4382–4390 (2013). meta-​analysis of evidence. BMJ 342, d2616 (2011). of pregnancy: correlations with total and free beta

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 169

0123456789();:
Reviews

human chorionic gonadotrophin. Clin. Endocrinol. 55, 96. Leung, A. M., Pearce, E. N. & Braverman, L. E. Iodine and adverse pregnancy outcomes in the Norwegian
391–398 (2001). nutrition in pregnancy and lactation. Endocrinol. Mother, Father and Child Cohort Study. BMC Med.
73. Andersen, S. L., Olsen, J., Carlé, A. & Laurberg, P. Metab. Clin. North Am. 40, 765–777 (2011). 18, 211 (2020).
Hyperthyroidism incidence fluctuates widely in and 97. Institute of Medicine (US) Panel on Micronutrients. 119. Gargari, S. S. et al. Maternal and neonatal outcomes
around pregnancy and is at variance with some other Dietary Reference Intakes for Vitamin A, Vitamin K, and determinants of iodine deficiency in third
autoimmune diseases: a Danish population-​based Arsenic, Boron, Chromium, Copper, Iodine, Iron, trimester of pregnancy in an iodine sufficient area.
study. J. Clin. Endocrinol. Metab. 100, 1164–1171 Manganese, Molybdenum, Nickel, Silicon, Vanadium, BMC Pregnancy Childbirth 20, 174 (2020).
(2015). and Zinc (National Academies Press (US), 2001). 120. Torlinska, B., Bath, S. C., Janjua, A., Boelaert, K.
74. Laurberg, P. & Andersen, S. L. Endocrinology in 98. WHO Secretariat, Andersson, M., de Benoist, B., & Chan, S.-Y. Iodine status during pregnancy in a
pregnancy: pregnancy and the incidence, diagnosing Delange, F. & Zupan, J. Prevention and control of region of mild-​to-moderate iodine deficiency is not
and therapy of Graves’ disease. Eur. J. Endocrinol. iodine deficiency in pregnant and lactating women associated with adverse obstetric outcomes; results
175, R219–R230 (2016). and in children less than 2-years-​old: conclusions and from the Avon Longitudinal Study of Parents and
75. Goodwin, T. M., Montoro, M., Mestman, J. H., recommendations of the Technical Consultation. Children (ALSPAC). Nutrients 10, 291 (2018).
Pekary, A. E. & Hershman, J. M. The role of chorionic Public Health Nutr. 10, 1606–1611 (2007). 121. Bath, S. C., Steer, C. D., Golding, J., Emmett, P.
gonadotropin in transient hyperthyroidism of This documents reviews recommendations regarding & Rayman, M. P. Effect of inadequate iodine status in
hyperemesis gravidarum. J. Clin. Endocrinol. Metab. iodine intake in pregnant and lactating women and UK pregnant women on cognitive outcomes in their
75, 1333–1337 (1992). children to prevent iodine deficiency. children: results from the Avon Longitudinal Study
76. Goodwin, T. M., Montoro, M. & Mestman, J. H. 99. WHO, UNICEF, ICCIDD. Assessment of iodine deficiency of Parents and Children (ALSPAC). Lancet 382,
Transient hyperthyroidism and hyperemesis gravidarum: disorders and monitoring their elimination: a guide for 331–337 (2013).
clinical aspects. Am. J. Obstet. Gynecol. 167, 648–652 programme managers. 3rd ed. http://whqlibdoc.who.int/ 122. Hynes, K. L., Otahal, P., Hay, I. & Burgess, J. R. Mild
(1992). publications/2007/9789241595827_eng.pdf (2007). iodine deficiency during pregnancy is associated with
77. Kahaly, G. J., Diana, T. & Olivo, P. D. TSH receptor 100. Andersen, S., Karmisholt, J., Pedersen, K. M. & reduced educational outcomes in the offspring: 9-year
antibodies: relevance & utility. Endocr. Pract. 26, Laurberg, P. Reliability of studies of iodine intake and follow-​up of the gestational iodine cohort. J. Clin.
97–106 (2020). recommendations for number of samples in groups Endocrinol. Metab. 98, 1954–1962 (2013).
78. Bouillon, R. et al. Thyroid function in patients with and in individuals. Br. J. Nutr. 99, 813–818 (2008). 123. Mil, N. H. V. et al. Low urinary iodine excretion during
hyperemesis gravidarum. Am. J. Obstet. Gynecol. 101. Pearce, E. N. & Caldwell, K. L. Urinary iodine, thyroid early pregnancy is associated with alterations in
143, 922–926 (1982). function, and thyroglobulin as biomarkers of iodine executive functioning in children. J. Nutr. 142,
79. Kinomoto-​Kondo, S. et al. The effects of gestational status. Am. J. Clin. Nutr. 104, 898S–901S (2016). 2167–2174 (2012).
transient thyrotoxicosis on the perinatal outcomes: 102. Wong, E. M., Sullivan, K. M., Perrine, C. G., 124. Costeira, M. J. et al. Psychomotor development of
a case-​control study. Arch. Gynecol. Obstet. 295, Rogers, L. M. & Peña-​Rosas, J. P. Comparison of children from an iodine-​deficient region. J. Pediatr.
87–93 (2017). median urinary iodine concentration as an indicator 159, 447–453 (2011).
80. Sheffield, J. S. & Cunningham, F. G. Thyrotoxicosis and of iodine status among pregnant women, school-​age 125. Moleti, M. et al. Effects of maternal iodine nutrition
heart failure that complicate pregnancy. Am. J. Obstet. children, and nonpregnant women. Food Nutr. Bull. and thyroid status on cognitive development in
Gynecol. 190, 211–217 (2004). 32, 206–212 (2011). offspring: a pilot study. Thyroid 26, 296–305 (2016).
81. Millar, L. K. et al. Low birth weight and preeclampsia 103. Zimmermann, M. B., Gizak, M., Abbott, K., 126. Zhou, S. J. et al. Association between maternal iodine
in pregnancies complicated by hyperthyroidism. Andersson, M. & Lazarus, J. H. Iodine deficiency in intake in pregnancy and childhood neurodevelopment
Obstet. Gynecol. 84, 946–949 (1994). pregnant women in Europe. Lancet Diabetes Endocrinol. at age 18 months. Am. J. Epidemiol. 188, 332–338
82. Casey, B. M. et al. Subclinical hyperthyroidism and 3, 672–674 (2015). (2019).
pregnancy outcomes. Obstet. Gynecol. 107, 337–341 104. Caldwell, K. L. et al. Iodine status in pregnant 127. Levie, D. et al. Association of maternal iodine status
(2006). women in the National Children’s Study and in with child IQ: a meta-​analysis of individual participant
83. Scappaticcio, L. et al. Abnormal liver blood tests in U.S. women (15-44 years), National Health and data. J. Clin. Endocrinol. Metab. 104, 5957–5967
patients with hyperthyroidism: systematic review and Nutrition Examination Survey 2005-2010. Thyroid (2019).
meta-​analysis. Thyroid 31, 884–894 (2021). 23, 927–937 (2013). 128. Ghassabian, A. et al. Maternal urinary iodine
84. Suzuki, N. et al. Analysis of antithyroid drug-​induced 105. Perrine, C. G., Herrick, K. A., Gupta, P. M. concentration in pregnancy and children’s cognition:
severe liver injury in 18,558 newly diagnosed & Caldwell, K. L. Iodine status of pregnant women results from a population-​based birth cohort in an
patients with Graves’ disease in Japan. Thyroid 29, and women of reproductive age in the United States. iodine-​sufficient area. BMJ Open 4, e005520 (2014).
1390–1398 (2019). Thyroid 29, 153–154 (2019). 129. Nazeri, P., Shariat, M. & Azizi, F. Effects of iodine
85. Yu, W. et al. Side effects of PTU and MMI in the 106. Caldwell, K. L., Makhmudov, A., Ely, E., Jones, R. L. supplementation during pregnancy on pregnant
treatment of hyperthyroidism: a systematic review and & Wang, R. Y. Iodine status of the U.S. population, women and their offspring: a systematic review and
meta-​analysis. Endocr. Pract. 26, 207–217 (2020). National Health and Nutrition Examination Survey, meta-​analysis of trials over the past 3 decades. Eur. J.
86. Rivkees, S. A. & Szarfman, A. Dissimilar hepatotoxicity 2005–2006 and 2007–2008. Thyroid 21, 419–427 Endocrinol. 184, 91–106 (2021).
profiles of propylthiouracil and methimazole in children. (2011). 130. Dineva, M., Fishpool, H., Rayman, M. P., Mendis, J.
J. Clin. Endocrinol. Metab. 95, 3260–3267 (2010). 107. Caldwell, K. L., Miller, G. A., Wang, R. Y., Jain, R. B. & Bath, S. C. Systematic review and meta-​analysis
87. Cooper, D. S. & Rivkees, S. A. Putting propylthiouracil & Jones, R. L. Iodine status of the U.S. population, of the effects of iodine supplementation on thyroid
in perspective. J. Clin. Endocrinol. Metab. 94, National Health and Nutrition Examination Survey function and child neurodevelopment in mildly-​
1881–1882 (2009). 2003-2004. Thyroid 18, 1207–1214 (2008). to-moderately iodine-​deficient pregnant women.
88. Andersen, S. L., Olsen, J., Wu, C. S. & Laurberg, P. 108. Yarrington, C. & Pearce, E. N. Iodine and pregnancy. Am. J. Clin. Nutr. 112, 389–412 (2020).
Birth defects after early pregnancy use of antithyroid J. Thyroid Res. 2011, 934104 (2011). This systematic review and meta-​analysis
drugs: a Danish nationwide study. J. Clin. Endocrinol. 109. Abuye, C. & Berhane, Y. The goitre rate, its association summarizes findings of 37 randomized controlled
Metab. 98, 4373–4381 (2013). with reproductive failure, and the knowledge of iodine trials, intervention studies, and observational
89. Andersen, S. L., Olsen, J., Wu, C. S. & Laurberg, P. deficiency disorders (IDD) among women in Ethiopia: studies on the effects of iodine supplementation
Severity of birth defects after propylthiouracil exposure cross-​section community based study. BMC Public in mildly-​to-moderately iodine deficiency pregnant
in early pregnancy. Thyroid 24, 1533–1540 (2014). Health 7, 316 (2007). women on maternal and infant thyroid function and
90. Andersen, S. L., Lönn, S., Vestergaard, P. & Törring, O. 110. Pearce, E. N. Effects of iodine deficiency in pregnancy. child neurodevelopmental outcomes.
Birth defects after use of antithyroid drugs in early J. Trace Elem. Med. Biol. 26, 131–133 (2012). 131. World Health Organization. Iodine supplementation
pregnancy: a Swedish nationwide study. Eur. J. 111. Glinoer, D. The importance of iodine nutrition during in pregnant and lactating women. WHO http://www.
Endocrinol. 177, 369–378 (2017). pregnancy. Public Health Nutr. 10, 1542–1546 (2007). who.int/elena/titles/iodine_pregnancy/en/ (2021).
91. Seo, G. H., Kim, T. H. & Chung, J. H. Antithyroid drugs 112. Pharoah, P. O., Ellis, S. M., Ekins, R. P. & Williams, E. S. 132. De Groot, L. et al. Management of thyroid dysfunction
and congenital malformations: a nationwide Korean Maternal thyroid function, iodine deficiency and fetal during pregnancy and postpartum: an Endocrine
cohort study. Ann. Intern. Med. 168, 405–413 (2018). development. Clin. Endocrinol. 5, 159–166 (1976). Society clinical practice guideline. J. Clin. Endocrinol.
92. Andersen, S. L., Knøsgaard, L., Olsen, J., 113. Pharoah, P. O., Buttfield, I. H. & Hetzel, B. S. Metab. 97, 2543–2565 (2012).
Vestergaard, P. & Andersen, S. Maternal thyroid Neurological damage to the fetus resulting from 133. Obican, S. G., Jahnke, G. D., Soldin, O. P. & Scialli, A. R.
function, use of antithyroid drugs in early pregnancy, severe iodine deficiency during pregnancy. Lancet 1, Teratology public affairs committee position paper:
and birth defects. J. Clin. Endocrinol. Metab. 104, 308–310 (1971). iodine deficiency in pregnancy. Birth Defects Res. A Clin.
6040–6048 (2019). 114. Bougma, K., Aboud, F. E., Harding, K. B. & Mol. Teratol. 94, 677–682 (2012).
This large study assessed the effects of antithyroid Marquis, G. S. Iodine and mental development of 134. Leung, A. M., Pearce, E. N., Braverman, L. E.
drug use in early pregnancy on risk of birth defects children 5 years old and under: a systematic review & Stagnaro-​Green, A. AAP recommendations on
utilizing the Danish national registry. and meta-​analysis. Nutrients 5, 1384–1416 (2013). iodine nutrition during pregnancy and lactation.
93. Momotani, N., Noh, J. Y., Ishikawa, N. & Ito, K. 115. Qian, M. et al. The effects of iodine on intelligence Pediatrics 134, e1282 (2014).
Effects of propylthiouracil and methimazole on fetal in children: a meta-​analysis of studies conducted in 135. Kung, A. W. C., Chau, M. T., Lao, T. T., Tam, S. C. F.
thyroid status in mothers with Graves’ hyperthyroidism. China. Asia Pac. J. Clin. Nutr. 14, 32–42 (2005). & Low, L. C. K. The effect of pregnancy on thyroid
J. Clin. Endocrinol. Metab. 82, 3633–3636 (1997). 116. Jiskra, J. et al. Mild iodine deficiency in women after nodule formation. J. Clin. Endocrinol. Metab. 87,
94. Laurberg, P. et al. TSH-​receptor autoimmunity in Graves’ spontaneous abortions living in an iodine-​sufficient 1010–1014 (2002).
disease after therapy with anti-​thyroid drugs, surgery, area of Czech Republic: prevalence and impact on 136. Gao, M. et al. Excessive iodine intake is associated
or radioiodine: a 5-year prospective randomized study. reproductive health. Clin. Endocrinol. 80, 452–458 with formation of thyroid nodules in pregnant Chinese
Eur. J. Endocrinol. 158, 69–75 (2008). (2014). women. Nutr. Res. 66, 61–67 (2019).
95. American Thyroid Association Taskforce on 117. Charoenratana, C., Leelapat, P., Traisrisilp, K. 137. Sahin, S. B. et al. Alterations of thyroid volume and
Radioiodine Safety. et al. Radiation safety in the & Tongsong, T. Maternal iodine insufficiency and nodular size during and after pregnancy in a severe
treatment of patients with thyroid diseases by adverse pregnancy outcomes. Matern. Child Nutr. 12, iodine-​deficient area. Clin. Endocrinol. 81, 762–768
radioiodine 131I: practice recommendations of the 680–687 (2016). (2014).
American Thyroid Association. Thyroid 21, 335–346 118. Abel, M. H. et al. Insufficient maternal iodine intake is 138. Haugen, B. R. et al. 2015 American Thyroid Association
(2011). associated with subfecundity, reduced foetal growth, management guidelines for adult patients with thyroid

170 | March 2022 | volume 18 www.nature.com/nrendo

0123456789();:
Reviews

nodules and differentiated thyroid cancer: The 148. Cho, G. J. et al. Risk of adverse obstetric outcomes 156. Ide, A. et al. Differentiation of postpartum Graves’
American Thyroid Association Guidelines Task Force and the abnormal growth of offspring in women with thyrotoxicosis from postpartum destructive
on Thyroid Nodules and Differentiated Thyroid Cancer. a history of thyroid cancer. Thyroid 29, 879–885 thyrotoxicosis using antithyrotropin receptor
Thyroid 26, 1–133 (2016). (2019). antibodies and thyroid blood flow. Thyroid 24,
139. Smith, L. H., Danielsen, B., Allen, M. E. & Cress, R. 149. Rakhlin, L., Fish, S. & Tuttle, R. M. Response to 1027–1031 (2014).
Cancer associated with obstetric delivery: results therapy status is an excellent predictor of pregnancy-​ 157. Amino, N. et al. Serum ratio of triiodothyronine
of linkage with the California cancer registry. Am. J. associated structural disease progression in patients to thyroxine, and thyroxine-​binding globulin
Obstet. Gynecol. 189, 1128–1135 (2003). previously treated for differentiated thyroid. cancer. and calcitonin concentrations in Graves’ disease and
140. Yasmeen, S. et al. Thyroid cancer in pregnancy. Thyroid 27, 396–401 (2017). destruction-​induced thyrotoxicosis. J. Clin. Endocrinol.
Int. J. Gynaecol. Obstet. 91, 15–20 (2005). This study assessed structural disease recurrence Metab. 53, 113–116 (1981).
141. Pitt, S. C. et al. Patients’ reaction to diagnosis with and progression during pregnancy in women with 158. Hubalewska-​Dydejczyk, A., Duntas, L. &
thyroid cancer or an indeterminate thyroid nodule. a history of papillary thyroid cancer. Gilis-Januszewska, A. Pregnancy, thyroid, and the
Thyroid 31, 580–588 (2021). 150. Pearce, E. N. Management of thyrotoxicosis: potential use of selenium. Hormones 19, 47–53
142. Moosa, M. & Mazzaferri, E. L. Outcome of preconception, pregnancy, and the postpartum period. (2020).
differentiated thyroid cancer diagnosed in pregnant Endocr. Pract. 25, 62–68 (2019). 159. UNICEF. Guidance on the monitoring of salt iodization
women. J. Clin. Endocrinol. Metab. 82, 2862–2866 151. Premawardhana, L. D. K. E., Parkes, A. B., John, R., programmes and determination of population
(1997). Harris, B. & Lazarus, J. H. Thyroid peroxidase iodine status. UNICEF https://www.ign.org/
143. Oh, H.-S. et al. Serial neck ultrasonographic antibodies in early pregnancy: utility for prediction p142003099.html?from=0142002801 (2018).
evaluation of changes in papillary thyroid carcinoma of postpartum thyroid dysfunction and implications
during pregnancy. Thyroid 27, 773–777 (2017). for screening. Thyroid 14, 610–615 (2004). Author contributions
144. Ito, Y. et al. Effects of pregnancy on papillary 152. Amino, N. & Arata, N. Thyroid dysfunction following The authors contributed equally to all aspects of the article.
microcarcinomas of the thyroid re-​evaluated in the pregnancy and implications for breastfeeding. Best.
entire patient series at Kuma hospital. Thyroid 26, Pract. Res. Clin. Endocrinol. Metab. 34, 101438 Competing interests
156–160 (2016). (2020). The authors declare no competing interests.
145. Vannucchi, G. et al. Clinical and molecular features 153. Moleti, M. et al. Postpartum thyroiditis in women
of differentiated thyroid cancer diagnosed during with euthyroid and hypothyroid hashimoto’s thyroiditis Peer review information
pregnancy. Eur. J. Endocrinol. 162, 145–151 antedating pregnancy. J. Clin. Endocrinol. Metab. 105, Nature Reviews Endocrinology thanks W. Teng and the other,
(2010). e2421–e2428 (2020). anonymous, reviewer(s) for their contribution to the peer
146. Messuti, I. et al. Impact of pregnancy on prognosis 154. Lucas, A. et al. Postpartum thyroiditis: long-​term review of this work.
of differentiated thyroid cancer: clinical and follow-​up. Thyroid 15, 1177–1181 (2005).
molecular features. Eur. J. Endocrinol. 170, 659–666 155. Premawardhana, L. D. et al. Postpartum thyroiditis Publisher’s note
(2014). and long-​term thyroid status: prognostic influence Springer Nature remains neutral with regard to jurisdictional
147. Tazebay, U. H. et al. The mammary gland iodide of thyroid peroxidase antibodies and ultrasound claims in published maps and institutional affiliations.
transporter is expressed during lactation and in echogenicity. J. Clin. Endocrinol. Metab. 85, 71–75
breast cancer. Nat. Med. 6, 871–878 (2000). (2000). © Springer Nature Limited 2022

NaTure RevIeWS | EndOCRInOLOGy volume 18 | March 2022 | 171

0123456789();:

You might also like