Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

REVIEW

Thyrotoxicosis: Diagnosis and Management


Anu Sharma, MBBS, and Marius N. Stan, MD

Abstract

Thyrotoxicosis is the clinical manifestation of excess thyroid hormone action at the tissue level due to
inappropriately high circulating thyroid hormone concentrations. Hyperthyroidism, a subset of
thyrotoxicosis, refers specifically to excess thyroid hormone synthesis and secretion by the thyroid
gland. We performed a review of the literature on these topics utilizing published data in PubMed and
MEDLINE. In this review, we discuss the more common etiologies of thyrotoxicosis, focusing on the
current approach to diagnosis and management, new trends in those directions, and potential up-
coming changes in the field.
ª 2018 Mayo Foundation for Medical Education and Research n Mayo Clin Proc. 2018;nn(n):1-17

From the Division of

T
hyrotoxicosis is the clinical manifes- of these etiologies, however, varies with the
Endocrinology, Meta-
tation of a group of disorders charac- level of iodine intake (nodular thyroid disease bolism and Diabetes, Uni-
terized by the presence of excess accounts for as much as 50% of cases in versity of Utah School of
thyroid hormone action at the tissue level iodine-deficient regions),5 the age of the popu- Medicine, Salt Lake City,
UT (A.S.); and Division of
and is the consequence of inappropriately lation (toxic nodular goiter is more common in Endocrinology, Diabetes,
high thyroid hormone concentrations.1 the elderly),6 and with the region studied Metabolism, and Nutri-
tion, Mayo Clinic, Roches-
Hyperthyroidism, a subset of thyrotoxicosis, (frequency of painless thyroiditis was 0.5% in ter, MN (M.N.S.).
refers specifically to excess thyroid hormone Denmark vs 22% in Wisconsin7,8).
synthesis and secretion by the thyroid gland.
The prevalence of hyperthyroidism is 1.3%
in the United States.2 It occurs more THYROTOXICOSIS WITH HYPERTHYROIDISM
commonly in women compared with men Graves Disease
(2% vs 0.2%),3 and the incidence increases History. Caleb Parry (1755-1822), a physi-
with age, iodine deficiency, and race (higher cian and farmer from Bath, England,
in whites compared with Hispanics and Afri- described 2 cases of goiter and palpitations.9
can Americans).4 We conducted a compre- His case records were published post-
hensive search of PubMed and MEDLINE humously in 1825 by his son.10 Robert J.
for English language articles using the terms Graves (1796-1853), chief physician at the
hyperthyroidism and thyrotoxicosis with no Meath Hospital in Dublin, Ireland, described
limits on date. The current review is based 3 cases of women with goiter and palpitations
on the most recent and highest-quality evi- in 1824-1825. In 1835, 4 cases were reported
dence available. (the fourth case had exophthalmos and was
added by his colleague William Stokes), and
ETIOLOGY in 1843 the entity of “GD” was described in
Thyrotoxicosis results from inappropriate acti- his book.11 In 1840, Karl von Basedow (1799-
vation at any level of the hypothalamic- 1854), a German physician, also reported a
pituitary-thyroid axis with increased thyroid case with palpitations, goiter, and exoph-
hormone production from thyroid follicles or thalmos (Merseburg triad).12 Graves over-
from release or ingestion of preformed thyroid looked Parry’s reports, and Basedow
hormone (Table 1). In iodine-sufficient areas, overlooked Graves’. The disease, therefore,
the most common cause of noniatrogenic has been referred to as Parry disease, GD, and
thyrotoxicosis is Graves disease (GD), which Basedow syndrome, depending on the
accounts for 80% of cases,5 followed by nodular geographic and cultural area involved. In
thyroid disease and thyroiditis. The frequency 1862, the eponymous honor was given to

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 1


www.mayoclinicproceedings.org n ª 2018 Mayo Foundation for Medical Education and Research
MAYO CLINIC PROCEEDINGS

mimicry with the TSH receptor), and stress


ARTICLE HIGHLIGHTS also play a role.19,20
d Thyrotropin receptor antibody assay, radioactive iodine uptake
and scan, and thyroid ultrasonography with color flow Doppler Diagnosis. Symptoms and signs of GD are
remain important tools for determining the cause of usually due to the underlying hyperthyroid-
thyrotoxicosis. ism or immune-mediated cellular infiltra-
tion. The most common manifestations are
d There is a changing trend in Graves disease therapy with
weight loss, fatigue, palpitations, tremor,
decreasing preference for radioactive iodine therapy and
and goiter. Atrial fibrillation is seen in 10%
increasing use of antithyroid drugs, including the use of long-term of individuals older than 60 years, whereas
antithyroid drug therapy, which is deemed safe and effective. a palpable goiter is more common in those
d Biotin use is a potential risk factor for spurious diagnosis of younger than 60 years.19 Clinically impor-
thyrotoxicosis. tant Graves orbitopathy (GO) occurs in
d New anticancer drugsdtyrosine kinase inhibitors and immune 25% of patients with GD; however, sub-
clinical eye involvement with extraocular
checkpoint inhibitorsdare causing thyroiditis, and thyroid func-
muscle enlargement can be seen radio-
tion monitoring should be considered in patients who take these
graphically in up to 70% of patients.21 Eye
drugs. lid retraction, exophthalmos, extraocular
d Immunomodulatory therapy is being tested in clinical trials to muscle dysfunction, and ocular pain are the
directly address the pathogenesis of Graves disease. most common manifestations of GO.22
Thyroid dermopathy is uncommon (occur-
ring in only 1%-4% of individuals with GD),
Graves because of Trousseau’s support at the is pathognomonic for thyroid autoimmu-
French Academy of Medicine.9 nity, and is almost always associated with
GO. Thyroid acropachy is even more rare,
Epidemiology and Pathogenesis. Graves dis- occurring in only 20% of patients with
ease is the most common cause of hyperthy- thyroid dermopathy.23
roidism. It has an annual incidence of 20 to Graves disease should be suspected in
50 cases per 100,000 persons and a lifetime the presence of clinical findings of thyrotox-
risk of 3% in women and 0.5% in men.13 The icosis combined with biochemical hyperthy-
pathogenesis of GD is strongly influenced by roidism (low serum TSH and high free
genetics. A family history of thyroid dysfunc- thyroxine [T4] or triiodothyronine [T3]). If
tion has been found in approximately half of pathognomonic signs are present (diffuse
individuals with GD.14 Twin concordance goiter with orbitopathy, dermopathy, or
studies suggest that up to 80% of risk is due to acropachy), no further testing is required
genetic factors.15 Many of the genes associated to establish the diagnosis. In the absence of
with an increased risk of GD overlap with these signs, measurement of TSH receptor
those associated with other autoimmune antibodies (TRAbs) (97% sensitivity and
diseases (rheumatoid arthritis, type 1 diabetes 98%-99% specificity for GD)24 can be help-
mellitus, and multiple sclerosis).16 Genes ful, especially in the setting of a nodular
conferring an increased risk for GD include goiter.25 Normal or increased radioactive
HLA-DRB1*03 (for expansion of gene sym- iodine (RAI) uptake (RAIU) with diffuse
bols, use search tool at www.genenames.org), distribution on the scan can also confirm
HLA-DRB1*08, thyroglobulin and thyrotropin the diagnosis and distinguishes GD from
(TSH) receptor, the protein tyrosine phos- other causes of thyrotoxicosis (Figure 1).
phatase nonreceptor type 22 (PTPN22), cyto- More recently, color flow Doppler sonogra-
toxic T-lymphocyte antigen 4 (CTLA4), CD25, phy (CFDS) with thyroid ultrasound has
and CD40.17,18 Environmental factors such as been employed with good accuracy for GD
cigarette smoking, dietary iodine, infection diagnosis as well (sensitivity of 87% and
with Yersinia enterocolitica (due to molecular specificity of 100%).26

n n
2 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

TABLE 1. Causes of Thyrotoxicosis


Etiology Mechanism
Thyrotoxicosis with hyperthyroidism (increased thyroid hormone synthesis)
Thyroidal origin
Graves disease TSHR-stimulating antibody
Toxic multinodular goiter Activating TSHR of G proteins
Solitary toxic adenoma Focal thyroid autonomy
TSH-secreting pituitary adenoma Autonomous TSH secretion
Neonatal Graves disease Thyroid-stimulating immunoglobulins
Congenital hyperthyroidism Activating mutations in TSHR
Familial gestational hyperthyroidism Activating mutations in TSHR
Pituitary resistance to thyroid hormone Mutated thyroid hormone receptor b
Choriocarcinoma Human chorionic gonadotropin secretion
Hyperemesis gravidarum Human chorionic gonadotropin secretion
Iodine, iodine-containing drugs (eg, amiodarone), Jod-Basedow phenomenon
radiographic contrast agents
Extrathyroidal origin
Struma ovarii Thyroid hormone production by dermoid tumor of ovary
Metastatic follicular thyroid cancer Extrathyroidal foci of thyroid hormone production
Thyrotoxicosis without hyperthyroidism (increased availability of preformed thyroid hormone)
Thyroidal origin
Silent (painless) and postpartum thyroiditis Release of stored thyroid hormone
Subacute thyroiditis Postviral infection
Drug-induced thyroiditis (eg, amiodarone-AIT2, Destruction of thyroid follicles
interferon-a, lithium, PD-1 inhibitors, TKI)
Acute infectious thyroiditis Bacterial or fungal infection
Radiation thyroiditis Thyroid cellular destruction and release of stored thyroid
hormone
Thyroid adenoma infarction Release of stored thyroid hormone
Extrathyroidal origin
Exogenous thyroid hormone Ingestion of thyroid hormone
AIT2 ¼ amiodarone-induced thyroiditis type 2; PD-1 ¼ programmed cell death 1; TKI ¼ tyrosine kinase inhibitor; TSH ¼ thyrotropin;
TSHR ¼ TSH receptor.

Treatment. The management of GD is ATDs are favored)34; however, its use has
2-fold: symptom control and treatment of been decreasing.35,36 Interestingly, quality of
the underlying hyperthyroidism. Most of the life after treatment is no different regardless
symptoms are due to overstimulation of of treatment modality.32
b-adrenergic receptors. b-Blockade, there- Radioactive iodine I 131 is incorporated
fore, is the mainstay of symptom control. into thyroid tissue through the sodium-iodine
Nonselective propranolol offers the addi- symporter. The expression of the sodium-
tional benefit of decreasing the peripheral iodine symporter gene is dependent on TSH re-
conversion of T4 to T3.27 In a thyroid storm, ceptor activation. This occurs diffusely in GD,
glucocorticoids are also used for this and therefore, RAI I 131 is incorporated into
purpose.28 the entire thyroid gland. Tissue necrosis ensues
Radioactive iodine ablation, antithyroid over the subsequent 6 to 18 weeks, resulting in
drugs (ATDs), and thyroidectomy are the hypothyroidism in 80% to 90% patients after a
mainstay treatment options for the underlying single dose.33 As with many radiation-based
hyperthyroidism (Table 2). Radioactive iodine therapies, a negative pregnancy test result is
has been the preferred first-line approach in required before RAI treatment for all women
the United States (unlike Europe, where of childbearing potential.

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 3


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

with low-dose methimazole (5-10 mg daily),


moderate hyperthyroidism (frequently with
a free T4 level 1.5-2 times the upper limit
of normal) can be treated starting with 10
to 20 mg of methimazole, and severe hyper-
thyroidism (free T4 level 2-3 times the
upper limit of normal) is treated with high-
dose methimazole (20-40 mg daily) in
divided doses.39
Antithyroid drugs are typically given for
18 months and then withdrawn to determine
remission, which is defined as no recurrence
of GD after 12 months without treatment.
The remission rate with ATDs is 30% to
50% in the United States40,41 and 50% to
FIGURE 1. Radioactive iodine I 123 uptake 60% in Europe.42 The following factors
image showing diffuse thyroid uptake in a pa- decrease the likelihood of remission: male
tient with Graves disease. sex, age less than 40 years, previous recur-
rence of GD, cigarette smoking, large goiter,
orbitopathy, high ratio of free T3 to free T4,
high titers of TRAb at diagnosis, and the
Antithyroid drugs inhibit thyroid hormone end of the course of ATDs.43-46
synthesis by disrupting thyroid peroxidase Adverse reactions occur in 13% of patients
(TPO) iodination of tyrosine residues on thyro- taking ATDs. Cutaneous reactions are more
globulinda critical step in the formation of T3 common with methimazole; however, hepa-
and T4. Propylthiouracil (PTU) and methima- totoxicity is more common with PTU.29,47
zole are both available in the United States, Risk of hepatitis is ATD dose dependent,
with the former providing an additional benefit increasing considerably with higher doses of
of decreasing T4 to T3 conversion. However, both PTU and methimazole (low-dose PTU
methimazole is the preferred drug in the United vs high-dose PTU hazard ratio, 0.39; high-
States given the concerns about severe hepato- dose methimazole hazard ratio of 5.08 vs
toxicity associated with PTU.37 Carbimazole, a low-dose methimazole hazard ratio of 1.15
prodrug to methimazole, is used in the British compared with PTU).48 Long-term use of
Commonwealth with similar effects to methi- ATDs has been reported over the years49
mazole. For most patients, ATDs are utilized and is usually favored in patients with GO.
alone with the dose titrated to achieve euthyr- This trend has increased in recent years, and
oidism. It is worth noting that in some parts there is suggestion of benefits (ie, weight sta-
of the world outside the United States. an alter- bility, less frequent dysthyroidism, better GO
native approach called “block and replace” is outcome) from maintaining endogenous thy-
also employed in patients who have rapid fluc- roid hormone production with ATDs as
tuations between hypothyroidism and hyper- opposed to achieving euthyroidism with LT4
thyroidism: ATDs are used at a fixed dose and after RAI.50 Reassuringly, the adverse effects
combined with levothyroxine (LT4) to achieve associated with ATDs are very unlikely to
euthyroidism. This regimen might be associ- develop after the first 3 to 6 months of ther-
ated with less frequent clinic visits and labora- apy. Thus, the incidence of hepatotoxicity de-
tory testing.38 creases from 30 per 1000 patient-years at 30
Initial dosing of methimazole is selected days on methimazole to less than 1 per 1000
on the basis of the clinical and biochemical patient-years after 180 days on methima-
severity of thyrotoxicosis. Mild hyperthy- zole,48,49,51,52 and there has been only one
roidism (typically with a free T4 level 1-1.5 case of serious adverse effects that have
times the upper limit of normal) is treated occurred in adults after 12 months of ATD
n n
4 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

TABLE 2. Treatment of Graves Diseasea


Remission
Treatment Mechanism Dosing rate Adverse effects Pregnancy
ATDs Block TPO action Initial dose proportional to 30%-60% Dependent on MMI
d MMI degree of elevation dose and duration [ Risk of congenital
b
d PTU of thyroid hormones, of therapy (total, 13%)29 abnormalities; recommended
symptoms, and goiter size d Rash in 2nd and 3rd trimester31
d Elevated liver enzymes PTU
d Agranulocytosis (0.3%) Lower teratogenic risk but
c
d Vasculitis (<0.1%)
30
[ risk of hepatotoxicity;
recommended in 1st trimester
All ATDs
[ Risk of fetal hypothyroidism
Radioactive Radiation-induced Fixed or calculated 80%-90%32,33 Worsening thyrotoxicosis, Contraindicated in pregnancy
iodine I thyroid follicular dose based on radiation thyroiditis, and lactation
131 cell necrosis goiter size and uptake19 worsening orbitopathy
Thyroid Surgical removal of Total thyroid-ectomy 100% Hypoparathyroidism, Can be performed in
surgery visible thyroid tissue laryngeal nerve injury 2nd trimester1
a
ATDs ¼ antithyroid drugs; MMI ¼ methimazole; PTU ¼ propylthiouracil; TPO ¼ thyroid peroxidase; T3 ¼ triiodothyronine; T4 ¼ thyroxine; [ ¼ increased.
b
Blocks TPO and peripheral T4 to T3 conversion.
c
Reported with PTU.

therapy, a case of vasculitis that developed af- ATDs are favored in patients with a high
ter the use of PTU for 6 years.51 likelihood of remission or with severe GO.
If thyroidectomy is chosen, patients Thyroidectomy is preferred when there are
should be pretreated with ATDs and b- symptoms of compression from large goiters,
blockers to induce euthyroidism before sur- suspicious nodules, or moderate to severe
gery.53 Iodine solutions (saturated solution and active orbitopathy in patients who
of potassium iodide or Lugol solution) are cannot tolerate ATDs.1
taken for 10 days preoperatively to help Follow-up depends on the treatment mo-
normalize thyroid hormone levels, decrease dality chosen. At 6 to 10 weeks after RAI,
thyroid vascularity, and minimize surgical free T4 and total T3 should be assessed and
blood loss. In addition, calcium and vitamin then monitored every 2 to 4 weeks thereafter
D levels should be assessed and repleted until there is evidence of progression toward
before surgery to prevent development of hypothyroidism, when LT4 therapy should
symptomatic postoperative hypocalcemia.54 be initiated.1 If an ATD is chosen, measure-
The operation should be performed by a ment of free T4 and total T3 should be
high-volume thyroid surgeon (>25 thyroid- performed every 2 to 6 weeks until euthyroid-
ectomies per year), which has been associ- ism is achieved. The dose should be decreased
ated with improved patient outcomes.55 on the basis of declining free T4 and T3 levels
An individualized approach taking into and achievement of euthyroidism. Once the
account patient preference, clinical factors, lowest dose to maintain euthyroidism is
availability of and expertise with the treat- apparent, this dose should be continued with
ment options, and cost should be considered laboratory assessment every 2 to 3 months or
before deciding on the first treatment every 6 months if long-term ATD therapy has
approach. For example, in a young woman been chosen. Currently, there is insufficient
who desires future pregnancy, RAI and sur- evidence to support routine measurement of
gery are favored because of the increased white blood cell count and liver function tests
incidence of congenital malformations seen in the absence of clinical symptoms in patients
with ATDs during pregnancy.31,56 However, taking ATDs.1 Following thyroidectomy, LT4
Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 5
www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

should be initiated at 1.6 mg/kg per day with In the setting of a palpable thyroid nodule,
repeated TSH measurement at 6 to 8 weeks.1 a nodular goiter, or antibody-negative
The management of GD during pregnancy biochemical hyperthyroidism, a thyroid up-
is the subject of ongoing controversy.57 take and scan study should be obtained. In
Ideally, patients should be rendered stably TA, there is focal uptake in the toxic nodule
euthyroid before becoming pregnant. Howev- (so-called hot nodule) with decreased uptake
er, if hyperthyroidism develops or persists dur- in the surrounding thyroid tissue (Figure 2).
ing pregnancy, PTU is preferred during the In TMNG, the scan reveals multiple areas of
first trimester because of the increased risk of focal increased uptake interspersed with re-
serious birth defects associated with methima- gions of decreased uptake that represent unaf-
zole and carbimazole (odds ratio, 1.9 for fected thyroid tissue (Figure 3).
methimazole/carbimazole vs PTU).31 The Both TA and TMNG can be treated with
risk for teratogenicity declines after week 10 either RAI or thyroid surgery. Antithyroid
of pregnancy, and thus PTU therapy is usually drugs are unable to induce cure for these con-
converted to methimazole because of the ditions given their underlying mechanism for
increased risk of hepatotoxicity from PTU. hyperthyroidism. Lifelong ATDs are also not
Given the risk of dysthyroidism associated a cost-effective option in the setting of young
with these therapeutic changes and the known individuals with TA or TMNG,64 but this
teratogenicity of PTU itself,58 these changes approach might be a reasonable choice in in-
are still a matter of debate. Regardless of agent, dividuals with increased surgical risk and/or
the lowest dose of ATD needed to control limited life expectancy. For TA, risk of persis-
hyperthyroidism should be used in order to tent hyperthyroidism is 6% to 18% after RAI
prevent fetal hypothyroidism. and less than 1% after thyroid lobectomy or
isthmusectomy.65 There is a 20% to 55%
risk of hypothyroidism with hemithyroidec-
Toxic Adenoma and Toxic Multinodular tomy.66,67 With RAI, however, there is a pro-
Goiter gressive risk of hypothyroidism with up to
In both toxic adenoma (TA) and toxic multi- 60% of patients becoming hypothyroid at 20
nodular goiter (TMNG), there is focal or years after RAI.68 This risk is more likely in
multifocal hyperplasia of thyroid follicular older patients and those with thyroid
cells with unregulated thyroid hormone pro-
duction due to autonomy. Activating muta-
tions in the TSH receptor gene seems to be
the underlying mechanism resulting in
increased adenylyl cyclase production inde-
pendent of TSH.59,60 In TA, activating muta-
tions in the Gsa protein have also been
identified.61 Toxic multinodular goiter is
more common in iodine-deficient areas62;
however, no clinical factors have been found
to correlate with TA. Toxic multinodular
goiter is also more likely in the elderly, in
whom it tends to manifest as apathetic thyro-
toxicosis. This presentation is defined by the
absence of typical symptoms of hyperthyroid-
ism in the presence of new-onset cardiac
symptoms (heart failure and arrhythmias),
FIGURE 2. Radioactive iodine I 123 uptake
cognitive changes, hypercalcemia, weakness,
image showing focal uptake in a toxic adenoma
and lethargy.63 It can be encountered in up with decreased uptake in surrounding normal
to 15% of elderly patients in whom hyperthy- thyroid tissue.
roidism develops, regardless of cause.
n n
6 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

antibodies and prior treatment with


ATDs.68,69 Radioactive iodine is more likely
to induce euthyroidism in TA as opposed to
GD because the uptake of RAI is limited to
the adenoma while the remaining thyroid pa-
renchyma is suppressed and thus does not
incur any severe damage from RAI. In GD,
however, there is diffuse uptake throughout
the gland, thereby ablating the entire thyroid.
For TMNG, the risk of persistent hyper- FIGURE 3. Three radioactive iodine I 123 uptake images showing charac-
thyroidism is 11% to 20% for RAI and less teristic heterogeneous patchy thyroid uptake found in toxic multinodular
than 1% with near-total/total thyroidec- goiters.
tomy.70,71 Risk of hypothyroidism is 16% at
5 years after RAI72 vs 100% with near-total/
total thyroidectomy. Long-term ATD ther- effect)80 or hyperthyroidism (Jod-Basedow
apy is generally not recommended unless it phenomenon).81 There is an inversely propor-
is in the setting of decreased life expectancy tional effect of dietary iodine intake on thyroid
or contraindications to ablative therapy function in amiodarone-induced thyroid
(eg, pregnancy or major comorbidities). dysfunction. Thus, hypothyroidism is more
Additionally, where expertise is available, frequent in iodine-sufficient areas (22% hypo-
radiofrequency ablation can be used to treat thyroidism vs 2% hyperthyroidism in Massa-
TA. Radiofrequency ablation has a cure rate chusetts), and hyperthyroidism is more likely
of 82% for thyrotoxicosis and is also associ- in iodine-deficient areas (5% hypothyroidism
ated with a substantial decrease in nodule vs 9.6% hyperthyroidism in Pisa, Italy).82
size.73 This modality is able to avoid hypo- Although amiodarone-induced hypothy-
thyroidism because of preservation of the roidism is an entity that is easily managed
surrounding thyroid tissue. with LT4 therapy, amiodarone-induced
thyrotoxicosis (AIT) poses both diagnostic
Amiodarone-Induced Thyroid Dysfunction and therapeutic challenges. Amiodarone-
Amiodarone is the most widely prescribed induced thyrotoxicosis is classified as type 1
antiarrhythmic agent in the United States. (increased thyroid hormone synthesis due
It has little negative inotropic activity and to underlying GD or TMNG) and type 2
is, therefore, often preferred in patients (destructive thyroiditis when the increased
with heart failure.74 It contains iodinedeach thyroid levels are due to release of previously
100-mg amiodarone tablet provides 10 times stored hormone).83 Type 2 AIT is now more
the average daily iodine content in the Amer- common than type 1.63 The classic adrenergic
ican diet.75 Compounding this problem is symptoms of thyrotoxicosis are often masked
amiodarone’s long half-life of 100 days, because of the b-blocking activity of amiodar-
which is due to its lipophilic nature.76 one. Thus, patients usually exhibit apathetic
The effects of amiodarone on the thyroid thyrotoxicosis,63 presenting with unexplained
are 2-fold: intrinsic drug effects and effects weight loss, new-onset or recurrent arrhyth-
related to its iodine content. The combined mias, and/or worsening ischemic heart dis-
results of these actions are that amiodarone de- ease or heart failure.
creases the conversion of T4 to T3,77 inhibits Differentiating type 1 from type 2 AIT is
the binding of T3 to its nuclear receptors,78 important because their treatments differ;
and is directly toxic to thyroid follicular cells, however, both types can coexist. Table 3
resulting in a destructive thyroiditis.79 In the summarizes the differing clinical, laboratory,
setting of underlying thyroid disease, the and imaging findings in type 1 and type 2
excessive iodine load received after amiodar- AIT. Our diagnostic approach is based on
one ingestion results in either hypothyroidism the clinical history and goiter assessment.
(failure to escape from the Wolff-Chaikoff We use TRAb in addition to CFDS, which

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 7


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

pituitary adenomas (standardized incidence


TABLE 3. Amiodarone-Induced Thyrotoxicosis
rate 0.03 per 100,000).92,93 They are diag-
Characteristic Type 1 Type 2 nosed in the setting of an inappropriately
84
Onset of symptoms after starting amiodarone (mo), median 3.5 30 normal or high TSH level with a concomi-
Onset of symptoms after amiodarone discontinued84 No Yes tant high T4 concentration. Patients present
Goiter (diffuse or multinodular) þ  with a small goiter and symptoms of mild
Thyrotropin receptor antibody þ (GD)  hyperthyroidism. Frequently, there are also
 (TMNG) symptoms generated through local compres-
Thyroglobulin85 [[ [ sion by the tumor (headaches and visual
Interleukin 686 Y [ field defects).94 This diagnosis should also
CFDS87 [4 Y be considered in patients with presumed pri-
Radioactive iodine uptake [4Y YY mary hypothyroidism in whom TSH does
99m
Tc sestamibi88,89
[4 Y not normalize with LT4 therapy.95 Pituitary
CFDS ¼ color flow Doppler sonography; 99mTc sestamibi ¼ technetium Tc 99m sestamibi
imaging with magnetic resonance is the
scintigraphy with scan; þ ¼ present;  ¼ absent; [ ¼ increased; [[ ¼ significantly increased; next step, and the most common presenta-
Y ¼ decreased; YY ¼ significantly decreased; [4 ¼ increased or normal; [4Y ¼ increased, tion of a TSH-producing adenoma is a
normal, or decreased.
macroadenoma. Cavernous sinus invasion
is present in 23% of these lesions.96 It can
can successfully differentiate the 2 types in also cosecrete other pituitary hormones,
80% of cases,87 and have moved away from most commonly growth hormone or prolac-
RAIU given consistently low uptake in our tin (16% and 10%, respectively).96 The treat-
iodine-replete population. Technetium Tc ment approach should be geared toward
99m sestamibi thyroid uptake and scintig- achieving surgical cure, with 84% of cases
raphy have been shown in small studies experiencing remission postoperatively and
(n¼2089 and n¼1590) to correctly differen- 17% complicated by postoperative pituitary
tiate the type of AIT in 100% of cases as dysfunction.96
compared to CFDS or RAI88,89; however, Thyrotropin-secreting pituitary adenomas
further validation is needed. express somatostatin receptors 2 and 5,97 mak-
Type 1 AIT is treated with ATDs, used at ing somatostatin analogues an effective medi-
high doses and for a longer period to prevent cal option in patients who are unable to
relapse to hyperthyroidism. Potassium undergo surgical resection. Somatostatin ana-
perchlorate has been used with some efficacy logues can be used to decrease tumor size
but is not available in the United States, and either preoperatively or after incomplete surgi-
RAI therapy is impractical in this population. cal resection. It has also been associated with
In type 2 AIT, high-dose prednisone (40-60 normalization of thyroid function.96,98,99
mg/d) is used for 1 to 3 months and then Although some tumors also express dopamine
slowly tapered. When AIT type is unclear or receptors, the response to dopamine agonists
initial response is minimal, both prednisone has been variable.100 Conventional radio-
and methimazole should be used. Unfortu- therapy or radiosurgery are other options
nately, in about 7% of cases90 there is insuffi- when there is incomplete cure with surgery.101
cient response to medical therapy or medical Antithyroid drugs are avoided given the
therapy is not tolerated, and in those cases expected tumor growth if the residual feed-
thyroidectomy is required. For a good success back inhibition from thyroid hormones on
rate, it is necessary to select an experienced the tumor is eliminated.
thyroid surgeon and have available cardiac
anesthesia, given the nonnegligible mortality Hydatidiform Mole/Choriocarcinoma/
reported in this patient population.91 Testicular Germ Cell Tumors
Human chorionic gonadotropin (hCG)
Thyrotropin-Secreting Pituitary Adenoma weakly stimulates the TSH receptor due to
Thyrotropin-secreting pituitary adenomas ligand-receptor cross-reactivity between the b
are rare, accounting for less than 1% of all subunits of hCG and TSH. Hyperthyroidism
n n
8 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

may, therefore, be a manifestation from the step in diagnosis. Treatment is surgical


severely elevated concentrations of hCG in removal of the tumor. Preoperative medical
hydatidiform mole, choriocarcinoma, and management includes b-blockers and ATDs
testicular germ cell tumors.102,103 Radioactive (in the setting of moderate to severe hyper-
iodine uptake is normal or increased. Therapy thyroidism). The stromal tissue can also har-
is directed to the underlying tumor; however, bor differentiated thyroid carcinoma.
b-blockade and ATDs can be used for symp- Therefore, RAI has been used as adjunctive
tom control before definitive treatment is therapy in metastatic disease.112
performed.
Other
Metastatic Follicular Thyroid Cancer There are other rare causes of hyperthyroid-
Follicular thyroid cancer metastasizes via he- ism listed in Table 1 that are beyond the scope
matogenous spread. These metastases contain of this review, eg, neonatal GD, congenital
functional thyroid tissue that rarely (about 60 hyperthyroidism, familial gestational hyper-
reported cases in the PubMed literature104-108) thyroidism, pituitary resistance to thyroid
can result in hyperthyroidism. Tumor cells ex- hormone, and hyperemesis gravidarum.
press increased activity of type 1 and type 2 dei-
odinases.109 Triiodothyronine thyrotoxicosis,
THYROTOXICOSIS WITHOUT
therefore, predominates because of a combina-
HYPERTHYROIDISM
tion of increased T3 secretion and increased
conversion of exogenous T4 to T3. Therefore, Thyroiditis
in these cases it will be important to obtain T3 Thyroiditis refers to any disorder that results
and T4 values besides TSH measurement in or- from inflammation of the thyroid tissue with
der to guide the extent of thyroid hormone resultant thyrotoxicosis due to release of pre-
replacement. The intended TSH suppression formed thyroid hormone (Table 4).
can be achieved with probably much lower
doses of LT4, or at times LT4 therapy is unnec- Subacute Thyroiditis. Subacute (granuloma-
essary. Rarely, hyperthyroidism can be due to tous) thyroiditis, or de Quervain thyroiditis,
TSH receptorestimulating antibodies.110 If hy- is thought to be due to a viral infection or a
perthyroidism is manifesting beyond the postviral inflammatory process. Presumably
intended TSH suppression despite discontinua- the infection-related antigen possesses struc-
tion of LT4, then treatment requires a multi- tural similarity with thyroid follicular cells.
modal approach utilizing a combination of Binding of the antigen to HLA-B35 on mac-
ATDs, RAI, surgery, or radiation. rophages results in activation of cytotoxic
T cells. These T cells then invade the thyroid,
Struma Ovarii causing thyroid inflammation and proteolysis
Struma ovarii is a mesodermal benign or ma- of stored thyroglobulin. There is a resultant
lignant teratoma located in the ovary with surge in T3 and T4 values due to their release,
thyroid tissue comprising more than 50% of causing symptoms of thyrotoxicosis. New
its mass. Women most commonly present thyroid hormone production ceases because
with pain or a pelvic mass (either incidental of underlying thyroid gland inflammation
or symptomatic). Rarely, hyperthyroidism is and lack of TSH stimulation, which is sup-
a manifestation.111,112 In the absence of a pressed due to the high concentrations of T3
simultaneously enlarged thyroid, the mecha- and T4. Often, the thyrotoxicosis is followed
nism is unknown but probably involves mu- by a period of hypothyroidism until the thy-
tations leading to autonomy of the stromal roid gland recovers and TSH increases. In
tissue itself. The diagnosis should be consid- most cases, thyroid hormone synthesis re-
ered in any woman with biochemical hyper- sumes and euthyroidism is eventually ach-
thyroidism, absence of goiter, absent RAIU ieved after a period of 2 to 3 months.
in the neck, and increased thyroglobulin. Pel- The presenting symptom is usually ante-
vic ultrasonography should then be the next rior neck pain following an infection,
Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 9
www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

TABLE 4. Forms of Thyroiditisa


Subtype History Etiology TPO antibodies ESR RAIU
Subacute Anterior neck pain following viral Postviral inflammation due to Low titer or negative High Low
infection antigen similarity with thyroid
follicular cells
Painless Absence of neck pain, family Likely autoimmune High titer positive Low Low
history of thyroid disorder
Postpartum Pregnancy or miscarriage within Likely autoimmune High titer positive Low Low
the past year, absence of neck
pain, family history of thyroid
disorder
Drug induced History of offending drug, family Induced thyroid autoimmunity or Positive or negative Low Low
history of thyroid disorder direct toxic effect on thyroid
Infectious/suppurative Fever, neck pain, immunocompro- Acute bacterial thyroid infection Negative High Normal
mised
Riedel/fibrousb Neck tightness, dysphagia, Extensive fibrosis with lymphocyte Positive, high titer Normal Low or normal
hoarseness, diffusely hard goiter and eosinophil infiltration
a
ESR ¼ erythrocyte sedimentation rate; RAIU ¼ radioactive iodine update; TPO ¼ thyroid peroxidase.
b
This form of thyroiditis is not associated with thyrotoxicosis at any point but rather with a slowly progressive form of hypothyroidism.

occasionally associated with fever, fatigue, lymphocytic thyroiditis, accounts for approxi-
and myalgia. The thyroid is enlarged and mately 0.5% to 5% of cases of hyperthyroidism
tender, and there are signs of thyrotoxicosis. in iodine-sufficient areas.7,113 In this category,
In addition to a suppressed TSH concentra- we do not include the cases of thyroiditis
tion with elevated T4 and T3 levels, labora- diagnosed within the first year postpartum.
tory findings include the absence of thyroid Painless thyroiditis is thought to be part of the
antibodies (can occasionally be present in spectrum of autoimmune thyroid disorders,
the hypothyroid phase), increased thyro- affecting more women than men and often
globulin, and elevated erythrocyte sedimen- occurring in the presence of thyroid antibodies
tation rate or C-reactive protein. or a family history of thyroid autoimmu-
Radioactive iodine uptake is diffusely low, nity.114 The diagnosis relies on biochemical
an essential element in the diagnosis of thyrotoxicosis, a small nontender thyroid
thyroiditis, and CFDS reveals decreased gland, and absence of pathognomonic signs of
vascularity. GD. The erythrocyte sedimentation rate is
Treatment is supportive because the disease normal, and RAIU is decreased. Eventually,
is self-limiting. Nonsteroidal anti-inflammatory 20% of patients with painless thyroiditis will
agents can be used for neck pain. If this treat- become hypothyroid.115 It has a clinical course
ment is insufficient, prednisone can be given, similar to that of subacute thyroiditis with
which relieves the pain within a few days. It hyperthyroidism initially resulting from thy-
then should be tapered off and the dose titrated roid gland destruction, then hypothyroidism
in response to pain. b-Blockade is helpful for from decreased thyroid hormone stores and
symptoms of thyrotoxicosis. During the phase synthesis, and then either normal thyroid
of hypothyroidism, a short course of LT4 may function resumes or the patient remains hy-
be needed for moderate to severe symptoms. pothyroid. Treatment is supportive, consisting
Because the thyroid function usually recovers, mainly of b-blockade unless hypothyroidism
LT4 should be tapered off and thyroid function persists.
should be monitored every 3 to 4 weeks until
euthyroidism resumes. Postpartum Thyroiditis. Postpartum thyroid-
itis occurs in 5% to 7% of women within
Painless Thyroiditis. Painless thyroiditis, the first few months after delivery.116 It is
which is also called silent thyroiditis or more common in women with other
n n
10 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

autoimmune disorders, TPO antibodies, or a for this adverse effect are sunitinib (preva-
family history of thyroid disease.114 Patho- lence 10%)122 and sorafenib (prevalence
logic findings reveal lymphocytic infiltration, 3%),123 and thyroiditis related to these
which suggests that it is a variant of Hashi- agents can occur as early as 6 weeks into
moto thyroiditis.117 As with other forms of therapy. Repeated cycles of therapy have
thyroiditis, the typical 3 phases are to be been followed by recurrent episodes of
expected, starting with thyrotoxicosis 1 to 6 thyroiditis and then by permanent
months after delivery. There is a 70% risk of hypothyroidism.124
recurrent postpartum thyroiditis after the first Checkpoint inhibitor immunotherapy
episode.118 Permanent hypothyroidism can targeting cytotoxic T-lymphocyte antigen 4
eventually occur in up to 50% of women119 (eg, ipilimumab) and programmed cell death
and is more likely in multiparous women or 1 receptor (eg, pembrolizumab) are excellent
after spontaneous abortion.120 therapies for metastatic melanoma. They are,
The diagnosis can be made in any woman however, associated with a destructive
within 1 year of pregnancy, and it follows the thyroiditis. Thyrotoxicosis was observed in
approach described for silent thyroiditis. In 6% to 22%125,126 of patients treated with pem-
the breastfeeding period, thyroid uptake and brolizumab, occurring at a median of 6 weeks
scan studies are used sparingly. Because after initiation of therapy.126 The exact mech-
iodine is secreted in breast milk, nursing anism is unclear; however, it may be due to
mothers will need to pump and discard milk T-celle, natural killer celle, or monocyte-
for 2 days after RAI I 123 is administered. mediated pathways.127
Treatment is supportive, as described for For all these entities, removal of the
painless thyroiditis. Prevention of post- offending agent must be weighed against
partum thyroiditis in women with high TPO the benefit that the drug provides for the
antibody titers has been tried with selenium preexistent disease. The therapy is mainly
therapy, and although preliminary studies supportive by controlling the symptoms of
have found this intervention beneficial,121 thyrotoxicosis with b-blockers and initiating
confirmatory studies are not yet available. LT4 when hypothyroidism develops.

Drug-Induced Thyroiditis. There are a num- Other Forms of Thyrotoxicosis Without


ber of drugs that are known to cause Hyperthyroidism. There are other rare
thyroiditis, some with less prominent utiliza- causes of thyrotoxicosis listed in Table 1
tion now than in the past (interferon alfa, that are beyond the scope of this review
interleukin 2) while others are still a consis- (acute infectious thyroiditis, radiation-
tent choice for their main indication (amio- induced thyroiditis, and thyrotoxicosis
darone, lithium). These drugs are by now associated with adenoma infarction).
well known to most practitioners. However,
new drugs have been developed recently that Exogenous Thyrotoxicosis
have consistently demonstrated an ability to Exogenous hyperthyroidism is due to inten-
induce inflammation and destruction of the tional (thyrotoxicosis factitia) or accidental
thyroid parenchyma through a number of ingestion of excessive amounts of thyroid hor-
mechanisms, primarily autoimmune and/or mone. In conditions such as differentiated
ischemic. The most commonly encountered thyroid cancer, there is intentional overdosing
classes of these new drugs are tyrosine by clinicians aiming to solely suppress TSH
kinase inhibitors and immune checkpoint (while attempting to avoid hyperthyroxine-
inhibitors. mia) and hopefully prevent or decrease tumor
Tyrosine kinase inhibitors are used to growth.128 Patients may overdose intentionally
treat many types of cancer (eg, renal cell car- to achieve the adverse effect of weight loss or to
cinoma, gastrointestinal tumors, and thyroid increase energy or because of a psychiatric
cancer), and they have been associated with disorder. There may be unintentional overdos-
a form of destructive thyroiditis. Best known ing with weight-reducing supplements129 or
Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 11
www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

thyroid health supplements130 or the occur- thyroid testing is paramount in avoiding this
rence of “hamburger thyroiditis” (consump- pitfall.
tion of beef contaminated with thyroid tissue Another pitfall we noted in the diagnosis
due to incorporation of the cow’s neck tissue of hyperthyroidism relates to the use of thy-
in the patty).131 roid scan and uptake. It is important that
Laboratory findings reveal biochemical this test be interpreted in view of the simul-
thyrotoxicosis, low thyroglobulin level, and taneous TSH value as well as through the
decreased RAIU. Fecal T4 can be measured appreciation of iodine status. In patients
in difficult cases.132 Treatment includes with elevated iodine uptake and normal
b-blockade, iopanoic acid (not available in TSH/T3/T4 concentrations, iodine deficiency
the United States), which decreases T4 to T3 should be suspected rather than endogenous
conversion, and cholestyramine, which binds hyperthyroidism. These patients will have a
T4 and T3 in the intestine.133 In extreme cases, symmetric uptake that is elevated based on
therapeutic plasmapheresis can be used to our reference range but is reflective of their
achieve a rapid decrease in thyroid hormone iodine deficit. It is thus pertinent that in
levels if the aforementioned measures are countries where iodine deficiency is preva-
not effective. lent, the normal value for iodine uptake is
much higher than in the United States. The
explanation for the iodine deficit in patients
PITFALLS IMPACTING ASSESSMENT OF residing in iodine-sufficient areas usually re-
THE THYROTOXIC STATE lates to some very particular dietary habits
The diagnosis of thyrotoxicosis is fairly that are pertinent for avoidance of iodine-
straightforward in most cases. However, containing products. All ATD therapies
there are a number of instances in which should be avoided in these cases, and iodine
the diagnosis should be carefully considered. supplementation should be initiated.
These scenarios can present with a discrep- Occasionally, thyrotoxicosis is diagnosed
ancy between a biochemical picture of thyro- on the basis of a suppressed TSH level alone,
toxicosis and a clinical presentation that most commonly during follow-up of patients
does not support it. Alternatively, there receiving thyroid hormone replacement
might be notable inconsistencies between therapy. In these cases, a suppressed TSH
the various thyroid parameters combined level is interpreted as iatrogenic thyrotoxi-
with variable clinical presentations, neither cosis, and the dose of LT4 is decreased. In
conclusive for thyroid dysfunction. our experience, this is noted when patients
One scenario relates to the impact of biotin with central hypothyroidism change medical
on thyroid test results.134,135 Currently, biotin providers. However, physicians should also
is frequently used for touted benefits regarding consider the possibility that an additional
skin, hair, and nail health. Biotinylated prod- pathologic process affecting TSH production
ucts are also part of a number of hormonal as- (eg, hypophysitis, pituitary tumor, pituitary
says, specifically TSH, free T4 and T3, and apoplexy) might have developed in patients
TRAb. The way biotin impacts these assays with preexistent primary hypothyroidism.
(suppressing TSH value and elevating T4, T3, A similar situation is noted in patients who
and TRAb) can result in a spurious diagnosis use high-dose corticosteroids, which can
of GD. Therefore, in order to avoid misdiag- suppress TSH-releasing hormone and TSH
nosis, we recommend performing thyroid tests concentrations136,137 leading to a state of
after patients have not taken biotin for a mini- central hypothyroidism. This is a transient
mum of 12 hours and preferably 24 hours. It situation that will resolve with the discontin-
seems that the dose of biotin that will clearly uation of corticosteroids. Therefore, a clin-
impact the thyroid test results is 10 mg/d or ical correlation for the suppressed TSH
higher, but lower doses might be problematic should be sought, and if it is not indicative
as well. Obviously, the role of patient and physi- of thyrotoxicosis, further exploration of thy-
cian education about proper methodology for roid status with T3 and T4 measurements
n n
12 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

should be sought in order to make the cor- Thus, hopefully the long-overdue therapeutic
rect diagnosis. paradigm shift for this entity will reach the
horizon.
UNRESOLVED QUESTIONS AND POTENTIAL
FUTURE DEVELOPMENTS Abbreviations and Acronyms: AIT = amiodarone-induced
In recent years, there have been a number of thyrotoxicosis; ATD = antithyroid drug; CFDS = color flow
attempts to revamp the approach to hyper- Doppler sonography; GD = Graves disease; GO = Graves
orbitopathy; hCG = human chorionic gonadotropin; LT4 =
thyroidism, particularly to GD. Many are levothyroxine; PTU = propylthiouracil; RAI = radioactive
targeting the immune system. Rituximab iodine; RAIU = RAI uptake; T3 = triiodothyronine; T4 =
has been tried, but the results were not thyroxine; TA = toxic adenoma; TMNG = toxic multinodular
impressive138 and it has not been pursued goiter; TPO = thyroid peroxidase; TRAb = thyrotropin re-
ceptor antibody; TSH = thyrotropin
further. CFZ533 targets CD40-CD154 inter-
action and has been tested in a phase 2 clin-
ical trial (NCT02713256) with results yet to Potential Competing Interests: Dr Stan received support
be reported. Other studies are testing the from Novartis Pharmaceuticals Corporation for a clinical
trial that tested a product (CFZ533) for Graves disease
efficacy of a tolerogenic vaccine to prevent (funds paid to his institution), unrelated to the current
TSH receptor antibody formation, and work. Dr Sharma reports no competing interests.
another uses a monoclonal antibody that is
Correspondence: Address to Marius N. Stan, MD, Division
a TSH receptor blocker. Some approaches of Endocrinology, Diabetes, Metabolism, and Nutrition,
to hyperthyroidism therapy are ultrasound Mayo Clinic, 200 First St SW, Rochester, MN 55905 (stan.
based, such as high-intensity focused ultra- marius@mayo.edu).
sound and radiofrequency ablation. All these
therapies will have to undergo further evalu- REFERENCES
ations before they can be employed routinely 1. Ross DS, Burch HB, Cooper DS, et al. 2016 American Thy-
in decreasing thyroid hormone production. roid Association guidelines for diagnosis and management of
hyperthyroidism and other causes of thyrotoxicosis [published
correction appears in Thyroid. 2017;27(11):1462]. Thyroid.
CONCLUSION 2016;26(10):1343-1421.
The diagnostic evaluation of thyrotoxicosis 2. Hollowell JG, Staehling NW, Flanders WD, et al. Serum TSH,
T4, and thyroid antibodies in the United States population
should start from the clinical picture and then (1988 to 1994): National Health and Nutrition Examination
add biochemical testing, nuclear medicine Survey (NHANES III). J Clin Endocrinol Metab. 2002;87(2):
data, and ultrasound imaging, as appropriate. 489-499.
3. Tunbridge WM, Evered DC, Hall R, et al. The spectrum of
Biotin is being increasingly acknowledged as a thyroid disease in a community: the Whickham survey. Clin
factor leading to spurious biochemical results Endocrinol (Oxf). 1977;7(6):481-493.
4. Golden SH, Robinson KA, Saldanha I, Anton B, Ladenson PW.
and should be considered in laboratory inter- Prevalence and incidence of endocrine and metabolic disor-
pretation. Distinguishing increased thyroid ders in the United States: a comprehensive review. J Clin Endo-
hormone production from thyroiditis remains crinol Metab. 2009;94(6):1853-1878.
5. Laurberg P, Bülow Pedersen I, Knudsen N, Ovesen L,
essential in therapeutic selection. Graves dis- Andersen S. Environmental iodine intake affects the type of
ease and TMNG are the dominant causes of hy- nonmalignant thyroid disease. Thyroid. 2001;11(5):457-469.
6. Abraham-Nordling M, Byström K, Törring O, et al. Incidence
perthyroidism, and for GD, we are seeing an
of hyperthyroidism in Sweden. Eur J Endocrinol. 2011;165(6):
increasing tendency to use ATDs, sometimes 899-905.
long-term, with a simultaneous decrease in 7. Schwartz F, Bergmann N, Zerahn B, Faber J. Incidence rate of
symptomatic painless thyroiditis presenting with thyrotoxi-
the use of RAI. Regarding thyroiditis, there cosis in Denmark as evaluated by consecutive thyroid scintig-
remain multiple etiologies to be considered, raphies. Scand J Clin Lab Invest. 2013;73(3):240-244.
and some of the new oncological drugs have 8. Nikolai TF, Brosseau J, Kettrick MA, Roberts R, Beltaos E.
Lymphocytic thyroiditis with spontaneously resolving hyper-
added to our differential diagnosis. These devel- thyroidism (silent thyroiditis). Arch Intern Med. 1980;140(4):
opments might also shed more light on the 478-482.
9. Weetman AP. Grave’s disease 1835-2002. Horm Res. 2003;
mechanisms involved in thyroid autoimmu- 59(suppl 1):114-118.
nity. This new information should spur the 10. Parry C. Collections From the Unpublished Medical Writings
ongoing effort of identifying new agents that of the Late Caleb Hillier Parry. London, UK: Underwoods;
1845:478.
can tackle thyrotoxicosis, particularly GD, in a 11. Graves RJ. Clinical Lectures on the Practice of Medicine. Dublin,
modern way by addressing its pathophysiology. Ireland: Fannin and Co; 1864.

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 13


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

12. Von Basedow CA. Exophthalmos durch Hypertrophie des randomized to treatment with antithyroid drugs, radioiodine,
Zellgewebes in der Augenhohle. Wchenscher Heilkunde. or surgery. Thyroid. 2005;15(11):1279-1286.
1840;6:197-202. 33. Dobyns BM, Vickery AL, Maloof F, Chapman EM. Functional
13. Zimmermann MB, Boelaert K. Iodine deficiency and thyroid and histologic effects of therapeutic doses of radioactive
disorders. Lancet Diabetes Endocrinol. 2015;3(4):286-295. iodine on the thyroid of man. J Clin Endocrinol Metab. 1953;
14. Manji N, Carr-Smith JD, Boelaert K, et al. Influences of age, 13(5):548-567.
gender, smoking, and family history on autoimmune thyroid 34. Bartalena L, Burch HB, Burman KD, Kahaly GJ. A 2013 Euro-
disease phenotype. J Clin Endocrinol Metab. 2006;91(12): pean survey of clinical practice patterns in the management of
4873-4880. Graves’ disease. Clin Endocrinol (Oxf). 2016;84(1):115-120.
15. Brix TH, Kyvik KO, Christensen K, Hegedüs L. Evidence for a 35. Burch HB, Burman KD, Cooper DS. A 2011 survey of clinical
major role of heredity in Graves’ disease: a population-based practice patterns in the management of Graves’ disease. J Clin
study of two Danish twin cohorts. J Clin Endocrinol Metab. Endocrinol Metab. 2012;97(12):4549-4558.
2001;86(2):930-934. 36. Brito JP, Schilz S, Singh Ospina N, et al. Antithyroid drugsdthe
16. Brand OJ, Gough SC. Genetics of thyroid autoimmunity and most common treatment for Graves’ disease in the United
the role of the TSHR. Mol Cell Endocrinol. 2010;322(1-2): States: a nationwide population-based study [letter]. Thyroid.
135-143. 2016;26(8):1144-1145.
17. Chen QY, Huang W, She JX, Baxter F, Volpe R, Maclaren NK. 37. Bahn RS, Burch HS, Cooper DS, et al. The role of propylth-
HLA-DRB1*08, DRB1*03/DRB3*0101, and DRB3*0202 are iouracil in the management of Graves’ disease in adults: report
susceptibility genes for Graves’ disease in North American of a meeting jointly sponsored by the American Thyroid As-
Caucasians, whereas DRB1*07 is protective. J Clin Endocrinol sociation and the Food and Drug Administration. Thyroid.
Metab. 1999;84(9):3182-3186. 2009;19(7):673-674.
18. Tomer Y. Mechanisms of autoimmune thyroid diseases: from 38. Vaidya B, Wright A, Shuttleworth J, et al. Block & replace
genetics to epigenetics. Annu Rev Pathol. 2014;9:147-156. regime versus titration regime of antithyroid drugs for the
19. Smith TJ, Hegedüs L. Graves’ disease [letter]. N Engl J Med. treatment of Graves’ disease: a retrospective observational
2017;376(2):185. study. Clin Endocrinol (Oxf). 2014;81(4):610-613.
20. Marinò M, Latrofa F, Menconi F, Chiovato L, Vitti P. Role of 39. Nakamura H, Noh JY, Itoh K, Fukata S, Miyauchi A,
genetic and non-genetic factors in the etiology of Graves’ dis- Hamada N. Comparison of methimazole and propylthiouracil
ease. J Endocrinol Invest. 2015;38(3):283-294. in patients with hyperthyroidism caused by Graves’ disease.
21. Enzmann DR, Donaldson SS, Kriss JP. Appearance of Graves’ J Clin Endocrinol Metab. 2007;92(6):2157-2162.
disease on orbital computed tomography. J Comput Assist 40. Klein I, Becker DV, Levey GS. Treatment of hyperthyroid dis-
Tomogr. 1979;3(6):815-819. ease. Ann Intern Med. 1994;121(4):281-288.
22. Bartley GB, Fatourechi V, Kadrmas EF, et al. Clinical features of 41. Sundaresh V, Brito JP, Thapa P, Bahn RS, Stan MN. Compar-
Graves’ ophthalmopathy in an incidence cohort. Am J Ophthal- ative effectiveness of treatment choices for Graves’ hyperthy-
mol. 1996;121(3):284-290. roidism: a historical cohort study. Thyroid. 2017;27(4):497-505.
23. Fatourechi V. Thyroid dermopathy and acropachy. Best Pract 42. Mazza E, Carlini M, Flecchia D, et al. Long-term follow-up of
Res Clin Endocrinol Metab. 2012;26(4):553-565. patients with hyperthyroidism due to Graves’ disease treated
24. Tozzoli R, Bagnasco M, Giavarina D, Bizzaro N. TSH receptor with methimazole: comparison of usual treatment schedule
autoantibody immunoassay in patients with Graves’ disease: with drug discontinuation vs continuous treatment with low
improvement of diagnostic accuracy over different genera- methimazole doses; a retrospective study. J Endocrinol Invest.
tions of methods; systematic review and meta-analysis. Auto- 2008;31(10):866-872.
immun Rev. 2012;12(2):107-113. 43. Abraham P, Avenell A, McGeoch SC, Clark LF, Bevan JS. Anti-
25. Pedersen IB, Knudsen N, Perrild H, Ovesen L, Laurberg P. thyroid drug regimen for treating Graves’ hyperthyroidism.
TSH-receptor antibody measurement for differentiation of Cochrane Database Syst Rev. 2010;(1):CD003420.
hyperthyroidism into Graves’ disease and multinodular toxic 44. Kimball LE, Kulinskaya E, Brown B, Johnston C, Farid NR. Does
goitre: a comparison of two competitive binding assays. Clin smoking increase relapse rates in Graves’ disease? J Endocrinol
Endocrinol (Oxf). 2001;55(3):381-390. Invest. 2002;25(2):152-157.
26. Hiraiwa T, Tsujimoto N, Tanimoto K, Terasaki J, 45. Allahabadia A, Daykin J, Holder RL, Sheppard MC, Gough SC,
Amino N, Hanafusa T. Use of color Doppler ultrasonog- Franklyn JA. Age and gender predict the outcome of treat-
raphy to measure thyroid blood flow and differentiate ment for Graves’ hyperthyroidism. J Clin Endocrinol Metab.
Graves’ disease from painless thyroiditis. Eur Thyroid J. 2000;85(3):1038-1042.
2013;2(2):120-126. 46. Carella C, Mazziotti G, Sorvillo F, et al. Serum thyrotropin re-
27. Cooper DS, Daniels GH, Ladenson PW, Ridgway EC. Hyper- ceptor antibodies concentrations in patients with Graves’ dis-
thyroxinemia in patients treated with high-dose propranolol. ease before, at the end of methimazole treatment, and after
Am J Med. 1982;73(6):867-871. drug withdrawal: evidence that the activity of thyrotropin re-
28. Williams DE, Chopra IJ, Orgiazzi J, Solomon DH. Acute effects ceptor antibody and/or thyroid response modify during the
of corticosteroids on thyroid activity in Graves’ disease. J Clin observation period. Thyroid. 2006;16(3):295-302.
Endocrinol Metab. 1975;41(2):354-361. 47. Otsuka F, Noh JY, Chino T, et al. Hepatotoxicity and cuta-
29. Sundaresh V, Brito JP, Wang Z, et al. Comparative effective- neous reactions after antithyroid drug administration. Clin
ness of therapies for Graves’ hyperthyroidism: a systematic re- Endocrinol (Oxf). 2012;77(2):310-315.
view and network meta-analysis. J Clin Endocrinol Metab. 2013; 48. Wang M-T, Lee W-J, Huang T-Y, Chu C-L, Hsieh C-H. Anti-
98(9):3671-3677. thyroid drug-related hepatotoxicity in hyperthyroidism pa-
30. Andersen SL, Olsen J, Laurberg P. Antithyroid drug side effects tients: a population-based cohort study. Br J Clin Pharmacol.
in the population and in pregnancy. J Clin Endocrinol Metab. 2014;78(3):619-629.
2016;101(4):1606-1614. 49. Azizi F, Malboosbaf R. Long-term antithyroid drug treatment:
31. Song R, Lin H, Chen Y, Zhang X, Feng W. Effects of methima- a systematic review and meta-analysis. Thyroid. 2017;27(10):
zole and propylthiouracil exposure during pregnancy on the 1223-1231.
risk of neonatal congenital malformations: a meta-analysis. 50. Villagelin D, Romaldini JH, Santos RB, Milkos AB, Ward LS.
PLoS One. 2017;12(7):e0180108. Outcomes in relapsed Graves’ disease patients following
32. Abraham-Nordling M, Törring O, Hamberger B, et al. Graves’ radioiodine or prolonged low dose of methimazole treat-
disease: a long-term quality-of-life follow up of patients ment. Thyroid. 2015;25(12):1282-1290.

n n
14 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

51. Laurberg P, Berman DC, Andersen S, Bülow Pedersen I. Sus- thyroid nodules: a 20 years’ retrospective study. Clin Endocrinol
tained control of Graves’ hyperthyroidism during long-term (Oxf). 2005;62(3):331-335.
low-dose antithyroid drug therapy of patients with severe 69. Nygaard B, Hegedüs L, Nielsen KG, Ulriksen P, Hansen JM.
Graves’ orbitopathy. Thyroid. 2011;21(9):951-956. Long-term effect of radioactive iodine on thyroid function
52. Elbers L, Mourits M, Wiersinga W. Outcome of very long- and size in patients with solitary autonomously functioning
term treatment with antithyroid drugs in Graves’ hyperthy- toxic thyroid nodules. Clin Endocrinol (Oxf). 1999;50(2):197-
roidism associated with Graves’ orbitopathy. Thyroid. 2011; 202.
21(3):279-283. 70. Erickson D, Gharib H, Li H, van Heerden JA. Treatment of pa-
53. Langley RW, Burch HB. Perioperative management of the thy- tients with toxic multinodular goiter. Thyroid. 1998;8(4):277-
rotoxic patient. Endocrinol Metab Clin North Am. 2003;32(2): 282.
519-534. 71. Kang AS, Grant CS, Thompson GB, van Heerden JA. Current
54. Oltmann SC, Brekke AV, Schneider DF, Schaefer SC, Chen H, treatment of nodular goiter with hyperthyroidism (Plummer’s
Sippel RS. Preventing postoperative hypocalcemia in patients disease): surgery versus radioiodine. Surgery. 2002;132(6):916-
with Graves disease: a prospective study. Ann Surg Oncol. 923.
2015;22(3):952-958. 72. Yano Y, Sugino K, Akaishi J, et al. Treatment of autonomously
55. Sosa JA, Bowman HM, Tielsch JM, Powe NR, Gordon TA, functioning thyroid nodules at a single institution: radioiodine
Udelsman R. The importance of surgeon experience for clin- therapy, surgery, and ethanol injection therapy. Ann Nucl Med.
ical and economic outcomes from thyroidectomy. Ann Surg. 2011;25(10):749-754.
1998;228(3):320-330. 73. Ha EJ, Baek JH, Kim KW, et al. Comparative efficacy of radio-
56. Seo GH, Kim TH, Chung JH. Antithyroid drugs and congenital frequency and laser ablation for the treatment of benign thy-
malformations: a nationwide Korean cohort study. Ann Intern roid nodules: systematic review including traditional pooling
Med. 2018;168(6):405-413. and Bayesian network meta-analysis. J Clin Endocrinol Metab.
57. Alexander EK, Pearce EN, Brent GA, et al. 2017 Guidelines of 2015;100(5):1903-1911.
the American Thyroid Association for the diagnosis and man- 74. Goldschlager N, Epstein AE, Naccarelli GV, et al. A practical
agement of thyroid disease during pregnancy and the post- guide for clinicians who treat patients with amiodarone:
partum [published correction appears in Thyroid. 2017;27(9): 2007 [published correction appears in Heart Rhythm. 2007;
1212]. Thyroid. 2017;27(3):315-389. 4(12):1590]. Heart Rhythm. 2007;4(9):1250-1259.
58. Andersen SL, Olsen J, Wu CS, Laurberg P. Severity of birth 75. Kennedy RL, Griffiths H, Gray TA. Amiodarone and the thy-
defects after propylthiouracil exposure in early pregnancy. roid. Clin Chem. 1989;35(9):1882-1887.
Thyroid. 2014;24(10):1533-1540. 76. Latini R, Tognoni G, Kates RE. Clinical pharmacokinetics of
59. Führer D, Holzapfel HP, Wonerow P, Scherbaum WA, amiodarone. Clin Pharmacokinet. 1984;9(2):136-156.
Paschke R. Somatic mutations in the thyrotropin receptor 77. Rao RH, McCready VR, Spathis GS. Iodine kinetic studies dur-
gene and not in the Gsa protein gene in 31 toxic thyroid nod- ing amiodarone treatment. J Clin Endocrinol Metab. 1986;62(3):
ules. J Clin Endocrinol Metab. 1997;82(11):3885-3891. 563-568.
60. Holzapfel HP, Führer D, Wonerow P, Weinland G, 78. Franklyn JA, Davis JR, Gammage MD, Littler WA,
Scherbaum WA, Paschke R. Identification of constitutively Ramsden DB, Sheppard MC. Amiodarone and thyroid hor-
activating somatic thyrotropin receptor mutations in a subset mone action. Clin Endocrinol (Oxf). 1985;22(3):257-264.
of toxic multinodular goiters. J Clin Endocrinol Metab. 1997; 79. Roti E, Minelli R, Gardini E, Bianconi L, Braverman LE. Thyro-
82(12):4229-4233. toxicosis followed by hypothyroidism in patients treated with
61. Parma J, Duprez L, Van Sande J, et al. Diversity and prevalence amiodarone: a possible consequence of a destructive process
of somatic mutations in the thyrotropin receptor and Gsa in the thyroid. Arch Intern Med. 1993;153(7):886-892.
genes as a cause of toxic thyroid adenomas. J Clin Endocrinol 80. Braverman LE, Ingbar SH, Vagenakis AG, Adams L, Maloof F.
Metab. 1997;82(8):2695-2701. Enhanced susceptibility to iodide myxedema in patients with
62. Laurberg P, Pedersen KM, Vestergaard H, Sigurdsson G. High Hashimoto’s disease. J Clin Endocrinol Metab. 1971;32(4):
incidence of multinodular toxic goitre in the elderly popula- 515-521.
tion in a low iodine intake area vs. high incidence of Graves’ 81. Stanbury JB, Ermans AE, Bourdoux P, et al. Iodine-induced hy-
disease in the young in a high iodine intake area: comparative perthyroidism: occurrence and epidemiology. Thyroid. 1998;
surveys of thyrotoxicosis epidemiology in East-Jutland 8(1):83-100.
Denmark and Iceland. J Intern Med. 1991;229(5):415-420. 82. Martino E, Safran M, Aghini-Lombardi F, et al. Environmental
63. Bogazzi F, Bartalena L, Dell’Unto E, et al. Proportion of type 1 iodine intake and thyroid dysfunction during chronic amiodar-
and type 2 amiodarone-induced thyrotoxicosis has changed one therapy. Ann Intern Med. 1984;101(1):28-34.
over a 27-year period in Italy. Clin Endocrinol (Oxf). 2007; 83. Bogazzi F, Bartalena L, Martino E. Approach to the patient
67(4):533-537. with amiodarone-induced thyrotoxicosis. J Clin Endocrinol
64. Vidal-Trecan GM, Stahl JE, Durand-Zaleski I. Managing toxic Metab. 2010;95(6):2529-2535.
thyroid adenoma: a cost-effectiveness analysis. Eur J Endocrinol. 84. Tomisti L, Rossi G, Bartalena L, Martino E, Bogazzi F. The
2002;146(3):283-294. onset time of amiodarone-induced thyrotoxicosis (AIT) de-
65. Vidal-Trecan GM, Stahl JE, Eckman MH. Radioiodine or sur- pends on AIT type. Eur J Endocrinol. 2014;171(3):363-368.
gery for toxic thyroid adenoma: dissecting an important deci- 85. Bartalena L, Brogioni S, Grasso L, Bogazzi F, Burelli A,
sion; a cost-effectiveness analysis. Thyroid. 2004;14(11):933- Martino E. Treatment of amiodarone-induced thyrotoxicosis,
945. a difficult challenge: results of a prospective study. J Clin Endo-
66. Verloop H, Louwerens M, Schoones JW, Kievit J, Smit JW, crinol Metab. 1996;81(8):2930-2933.
Dekkers OM. Risk of hypothyroidism following hemithyroi- 86. Bartalena L, Grasso L, Brogioni S, Aghini-Lombardi F,
dectomy: systematic review and meta-analysis of prognostic Braverman LE, Martino E. Serum interleukin-6 in
studies. J Clin Endocrinol Metab. 2012;97(7):2243-2255. amiodarone-induced thyrotoxicosis. J Clin Endocrinol Metab.
67. Ahn D, Sohn JH, Jeon JH. Hypothyroidism following hemithyr- 1994;78(2):423-427.
oidectomy: incidence, risk factors, and clinical characteristics. 87. Bogazzi F, Martino E, Dell’Unto E, et al. Thyroid color flow
J Clin Endocrinol Metab. 2016;101(4):1429-1436. doppler sonography and radioiodine uptake in 55 consecutive
68. Ceccarelli C, Bencivelli W, Vitti P, Grasso L, Pinchera A. patients with amiodarone-induced thyrotoxicosis. J Endocrinol
Outcome of radioiodine-131 therapy in hyperfunctioning Invest. 2003;26(7):635-640.

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 15


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

88. Wang J, Zhang R. Evaluation of 99mTc-MIBI in thyroid gland 107. Nishihara E, Amino N, Miyauchi A. Fractionated radioiodine
imaging for the diagnosis of amiodarone-induced thyrotoxi- therapy for hyperthyroidism caused by widespread metastatic
cosis. Br J Radiol. 2017;90(1071):20160836. follicular thyroid carcinoma [letter]. Thyroid. 2010;20(5):569-
89. Piga M, Cocco MC, Serra A, Boi F, Loy M, Mariotti S. The use- 570.
fulness of 99mTc-sestaMIBI thyroid scan in the differential diag- 108. Azevedo MF, Casulari LA. Hyperfunctioning thyroid cancer: a
nosis and management of amiodarone-induced thyrotoxicosis. five-year follow-up. Arq Bras Endocrinol Metabol. 2010;54(1):
Eur J Endocrinol. 2008;159(4):423-429. 78-80.
90. Tomisti L, Materazzi G, Bartalena L, et al. Total thyroidectomy 109. Miyauchi A, Takamura Y, Ito Y, et al. 3,5,3’-Triiodothyronine
in patients with amiodarone-induced thyrotoxicosis and se- thyrotoxicosis due to increased conversion of administered
vere left ventricular systolic dysfunction. J Clin Endocrinol levothyroxine in patients with massive metastatic follicular thy-
Metab. 2012;97(10):3515-3521. roid carcinoma. J Clin Endocrinol Metab. 2008;93(6):2239-
91. Houghton SG, Farley DR, Brennan MD, van Heerden JA, 2242.
Thompson GB, Grant CS. Surgical management of 110. Kasagi K, Takeuchi R, Miyamoto S, et al. Metastatic thyroid
amiodarone-associated thyrotoxicosis: Mayo Clinic experi- cancer presenting as thyrotoxicosis: report of three cases.
ence. World J Surg. 2004;28(11):1083-1087. Clin Endocrinol (Oxf). 1994;40(3):429-434.
92. Tjörnstrand A, Gunnarsson K, Evert M, et al. The incidence 111. Ayhan A, Yanik F, Tuncer R, Tuncer ZS, Ruacan S. Struma
rate of pituitary adenomas in western Sweden for the period ovarii. Int J Gynaecol Obstet. 1993;42(2):143-146.
2001-2011. Eur J Endocrinol. 2014;171(4):519-526. 112. DeSimone CP, Lele SM, Modesitt SC. Malignant struma ovarii:
93. Raappana A, Koivukangas J, Ebeling T, Pirilä T. Incidence of pi- a case report and analysis of cases reported in the literature
tuitary adenomas in Northern Finland in 1992-2007. J Clin with focus on survival and I131 therapy. Gynecol Oncol. 2003;
Endocrinol Metab. 2010;95(9):4268-4275. 89(3):543-548.
94. Amlashi FG, Tritos NA. Thyrotropin-secreting pituitary ade- 113. Ross DS. Syndromes of thyrotoxicosis with low radioactive
nomas: epidemiology, diagnosis, and management. Endocrine. iodine uptake. Endocrinol Metab Clin North Am. 1998;27(1):
2016;52(3):427-440. 169-185.
95. Tjörnstrand A, Nyström HF. DIAGNOSIS OF ENDOCRINE 114. Pearce EN, Farwell AP, Braverman LE. Thyroiditis [published
DISEASE: Diagnostic approach to TSH-producing pituitary ad- correction appears in N Engl J Med. 2003;349(6):620].
enoma. Eur J Endocrinol. 2017;177(4):R183-R197. N Engl J Med. 2003;348(26):2646-2655.
96. Yamada S, Fukuhara N, Horiguchi K, et al. Clinicopathological 115. Nikolai TF, Coombs GJ, McKenzie AK. Lymphocytic thyroiditis
characteristics and therapeutic outcomes in thyrotropin- with spontaneously resolving hyperthyroidism and subacute
secreting pituitary adenomas: a single-center study of 90 cases. thyroiditis: long-term follow-up. Arch Intern Med. 1981;
J Neurosurg. 2014;121(6):1462-1473. 141(11):1455-1458.
97. Yoshihara A, Isozaki O, Hizuka N, et al. Expression of type 5 116. Muller AF, Drexhage HA, Berghout A. Postpartum thyroiditis
somatostatin receptor in TSH-secreting pituitary adenomas: a and autoimmune thyroiditis in women of childbearing age:
possible marker for predicting long-term response to octreo- recent insights and consequences for antenatal and postnatal
tide therapy. Endocr J. 2007;54(1):133-138. care. Endocr Rev. 2001;22(5):605-630.
98. Caron P, Arlot S, Bauters C, et al. Efficacy of the long-acting 117. Mizukami Y, Michigishi T, Nonomura A, et al. Postpartum
octreotide formulation (octreotide-LAR) in patients with thyroiditis: a clinical, histologic, and immunopathologic study
thyrotropin-secreting pituitary adenomas. J Clin Endocrinol of 15 cases. Am J Clin Pathol. 1993;100(3):200-205.
Metab. 2001;86(6):2849-2853. 118. Lazarus JH, Ammari F, Oretti R, Parkes AB, Richards CJ,
99. Kuhn JM, Arlot S, Lefebvre H, et al. Evaluation of the treat- Harris B. Clinical aspects of recurrent postpartum thyroiditis.
ment of thyrotropin-secreting pituitary adenomas with a Br J Gen Pract. 1997;47(418):305-308.
slow release formulation of the somatostatin analog lanreo- 119. Premawardhana LD, Parkes AB, Ammari F, et al. Postpartum
tide. J Clin Endocrinol Metab. 2000;85(4):1487-1491. thyroiditis and long-term thyroid status: prognostic influence
100. Brucker-Davis F, Oldfield EH, Skarulis MC, Doppman JL, of thyroid peroxidase antibodies and ultrasound echogenicity.
Weintraub BD. Thyrotropin-secreting pituitary tumors: diag- J Clin Endocrinol Metab. 2000;85(1):71-75.
nostic criteria, thyroid hormone sensitivity, and treatment 120. Browne-Martin K, Emerson CH. Postpartum thyroid dysfunc-
outcome in 25 patients followed at the National Institutes tion. Clin Obstet Gynecol. 1997;40(1):90-101.
of Health. J Clin Endocrinol Metab. 1999;84(2):476-486. 121. Negro R, Greco G, Mangieri T, Pezzarossa A, Dazzi D,
101. Malchiodi E, Profka E, Ferrante E, et al. Thyrotropin-secreting Hassan H. The influence of selenium supplementation on
pituitary adenomas: outcome of pituitary surgery and irradia- postpartum thyroid status in pregnant women with thyroid
tion. J Clin Endocrinol Metab. 2014;99(6):2069-2076. peroxidase autoantibodies. J Clin Endocrinol Metab. 2007;
102. Walkington L, Webster J, Hancock BW, Everard J, 92(4):1263-1268.
Coleman RE. Hyperthyroidism and human chorionic gonado- 122. Ahmadieh H, Salti I. Tyrosine kinase inhibitors induced thy-
trophin production in gestational trophoblastic disease. Br J roid dysfunction: a review of its incidence, pathophysiology,
Cancer. 2011;104(11):1665-1669. clinical relevance, and treatment. Biomed Res Int. 2013;2013:
103. Oosting SF, de Haas EC, Links TP, et al. Prevalence of 725410.
paraneoplastic hyperthyroidism in patients with metastatic 123. Tamaskar I, Bukowski R, Elson P, et al. Thyroid function test
non-seminomatous germ-cell tumors. Ann Oncol. 2010;21(1): abnormalities in patients with metastatic renal cell carcinoma
104-108. treated with sorafenib. Ann Oncol. 2008;19(2):265-268.
104. Karimifar M. A case of functional metastatic follicular thyroid 124. Desai J, Yassa L, Marqusee E, et al. Hypothyroidism after suni-
carcinoma that presented with hip fracture and hypercalcemia. tinib treatment for patients with gastrointestinal stromal tu-
Adv Biomed Res. 2018;7:92. mors. Ann Intern Med. 2006;145(9):660-664.
105. Aoyama M, Takizawa H, Tsuboi M, Nakagawa Y, Tangoku A. 125. Morganstein DL, Lai Z, Spain L, et al. Thyroid abnormalities
A case of metastatic follicular thyroid carcinoma complicated following the use of cytotoxic T-lymphocyte antigen-4 and
with Graves’ disease after total thyroidectomy. Endocr J. 2017; programmed death receptor protein-1 inhibitors in the treat-
64(12):1143-1147. ment of melanoma. Clin Endocrinol (Oxf). 2017;86(4):614-620.
106. Danilovic DL, de Camargo RY, Castro G Jr, Papadia C, 126. de Filette J, Jansen Y, Schreuer M, et al. Incidence of thyroid-
Marui S, Hoff AO. Rapid control of T3 thyrotoxicosis in pa- related adverse events in melanoma patients treated with
tients with metastatic follicular thyroid cancer treated with len- pembrolizumab. J Clin Endocrinol Metab. 2016;101(11):4431-
vatinib [letter]. Thyroid. 2015;25(11):1262-1264. 4439.

n n
16 Mayo Clin Proc. XXX 2018;nn(n):1-17 https://doi.org/10.1016/j.mayocp.2018.10.011
www.mayoclinicproceedings.org
THYROTOXICOSIS

127. Delivanis DA, Gustafson MP, Bornschlegl S, et al. Pembrolizu- 133. Shakir KM, Michaels RD, Hays JH, Potter BB. The use of
mab-induced thyroiditis: comprehensive clinical review and in- bile acid sequestrants to lower serum thyroid hormones
sights into underlying involved mechanisms. J Clin Endocrinol in iatrogenic hyperthyroidism. Ann Intern Med. 1993;
Metab. 2017;102(8):2770-2780. 118(2):112-113.
128. Haugen BR, Alexander EK, Bible KC, et al. 2015 American 134. Kummer S, Hermsen D, Distelmaier F. Biotin treatment
Thyroid Association management guidelines for adult patients mimicking Graves’ disease [letter]. N Engl J Med. 2016;
with thyroid nodules and differentiated thyroid cancer: the 375(7):704-706.
American Thyroid Association Guidelines Task Force on Thy- 135. Sharma A, Baumann NA, Shah P. Biotin-induced biochemical
roid Nodules and Differentiated Thyroid Cancer. Thyroid. Graves disease: a teachable moment. JAMA Intern Med. 2017;
2016;26(1):1-133. 177(4):571-572.
129. Ohye H, Fukata S, Kanoh M, et al. Thyrotoxicosis caused by 136. Alkemade A, Unmehopa UA, Wiersinga WM, Swaab DF,
weight-reducing herbal medicines. Arch Intern Med. 2005; Fliers E. Glucocorticoids decrease thyrotropin-releasing hor-
165(8):831-834. mone messenger ribonucleic acid expression in the paraven-
130. Kang GY, Parks JR, Fileta B, et al. Thyroxine and triiodothyro- tricular nucleus of the human hypothalamus. J Clin Endocrinol
nine content in commercially available thyroid health supple- Metab. 2005;90(1):323-327.
ments. Thyroid. 2013;23(10):1233-1237. 137. Shimokaze T, Toyoshima K, Shibasaki J, et al. TSH sup-
131. Kinney JS, Hurwitz ES, Fishbein DB, et al. Community pression after intravenous glucocorticosteroid administra-
outbreak of thyrotoxicosis: epidemiology, immunogenetic tion in preterm infants. J Pediatr Endocrinol Metab. 2012;
characteristics, and long-term outcome. Am J Med. 1988; 25(9-10):853-857.
84(1):10-18. 138. El Fassi D, Nielsen CH, Bonnema SJ, Hasselbalch HC,
132. Bouillon R, Verresen L, Staels F, Bex M, De Vos P, De Roo M. Hegedüs L. B lymphocyte depletion with the monoclonal anti-
The measurement of fecal thyroxine in the diagnosis of thyro- body rituximab in Graves’ disease: a controlled pilot study.
toxicosis factitia. Thyroid. 1993;3(2):101-103. J Clin Endocrinol Metab. 2007;92(5):1769-1772.

Mayo Clin Proc. n XXX 2018;nn(n):1-17 n https://doi.org/10.1016/j.mayocp.2018.10.011 17


www.mayoclinicproceedings.org

You might also like