You are on page 1of 13

Endocrine

https://doi.org/10.1007/s12020-024-03718-2

REVIEW

Thyroid disorders induced by immune checkpoint inhibitors


Dimitra Karaviti 1 Eleni-Rafaela Kani 1 Eleftheria Karaviti 1 Eleni Gerontiti 1 Olympia Michalopoulou1
● ● ● ● ●

Katerina Stefanaki1 Paraskevi Kazakou1 Vasiliki Vasileiou2 Theodora Psaltopoulou1 Stavroula A. Paschou
● ● ● ●
1

Received: 13 December 2023 / Accepted: 30 January 2024


© The Author(s) 2024

Abstract
Immune checkpoint inhibitors (ICIs) are a revolutionary class of drugs that powerfully contribute to cancer therapy by
harnessing the immune system to fight malignancies. However, their successful use as anti-cancer drugs is accompanied by a
wide spectrum of immune-related adverse effects (irAEs), including endocrinopathies. Among them, thyroid dysfunction
stands out as one of the most common endocrinopathies induced by ICI therapy and surfaces as a prominent concern.
Destructive thyroiditis is the pathophysiological basis shared by the most common patterns of thyrotoxicosis followed by
hypothyroidism and isolated hypothyroidism. Diagnostic approach is guided by clinical manifestation, laboratory evaluation
1234567890();,:
1234567890();,:

and imaging modalities. Treatment approaches range from the substitution of levothyroxine to the utilization of beta
blockers, depending on the extent of thyroid dysfunction’s severity. While the medical community is dealing with the
evolution and complexities of immunotherapy, recognizing and effectively managing ICI-induced thyroid dysfunction
emerged as crucial for enhancing patient safety and achieving improved outcomes. The aim of this review is to navigate the
significance of ICI-induced thyroid dysfunction unraveling the various patterns, underlying mechanisms, diagnostic
approaches, and treatment strategies. It, also, highlights the impact of various factors such as cancer subtype, ICI dosage,
age, and genetic susceptibility on the risk of experiencing dysfunction.
Keywords Immune checkpoint inhibitors Immune-related adverse events Thyroid dysfunction Pathogenesis Diagnosis
● ● ● ● ●

Management

Introduction malignant melanoma and lung cancer, this technique


involves the administration of monoclonal antibodies that
In recent years, immune checkpoint inhibitor (ICI) therapy target specific cell proteins such as cytotoxic T lymphocyte-
has emerged as a promising approach in cancer treatment. associated protein 4 (CTLA-4), programmed cell death
This breakthrough approach leverages the body’s immune protein 1 (PD-1), and its ligand (PD-L1). Therefore, T cell
system to fight cancerous tumors, providing patients with activation against neoplasms is induced [2, 3]. However,
valuable therapeutic alternatives and improved survival alongside its benefits, the therapy comes with a range of
prospects [1]. Initially employed in the treatment of adverse effects, with thyroid disorders being among the
most prevalent endocrine complications. Thyroid dysfunc-
tion manifests with a wide clinical spectrum encompassing
thyroiditis, both hypo- and thyrotoxicosis, Graves’ disease
[4]. Its pathophysiological basis is considered to be
These authors contributed equally: Dimitra Karaviti, Eleni-Rafaela
destructive thyroiditis induced by a T cell-mediated acute
Kani
autoimmune response [1, 5–9]. However, studies con-
* Stavroula A. Paschou sistently highlight the role of autoantibodies against thyr-
s.a.paschou@gmail.com oglobulin (Tg), thyroid peroxidase (TPO) and thyroid
1 stimulating hormone (TSH) receptor and even the role of
Endocrine Unit and Diabetes Center, Department of Clinical
Therapeutics, Alexandra Hospital, School of Medicine, National cytokines in the pathogenesis of the disease [10, 11].
and Kapodistrian University of Athens, Athens, Greece Therefore, laboratory tests involving TSH, free thyroxine
2
Department of Endocrinology, Alexandra Hospital, (fT4), and antibody measurements carry immense impor-
Athens, Greece tance not only in accurate diagnosis, but also as check-up
Endocrine

prior to and during ICI therapy [12]. ICI-induced thyr- positivity [19]. The correlation between basal positivity of
oidopathy ranges in clinical presentation from asympto- TPOAb and TgAb and high risk of thyroid dysfunction after
matic cases to severe manifestations and death [13]. the initiation of ICIs therapy is also supported by the study of
Addressing these complications necessitates the prompt Zhou et al. [20]. Reportedly, the development of positive
diagnosis of the thyroid disorder, along with the imple- thyroid autoantibodies after initiation of ICI therapy is asso-
mentation of therapeutic strategies and drug dosages, tai- ciated with higher risk of ICI-induced thyroid dysfunction.
lored to the clinical manifestations and their severity. In In similar studies, stimulatory autoantibodies against
essence, close monitoring and collaboration between TSH receptor (TRAb) or thyroid-stimulating immunoglo-
oncologists and endocrinologists is required [13–15]. bulin (TSI) predominantly found negative in the majority of
cases. Case reports indicate that ICI therapy-induced thyroid
dysfunction may impact and abolish the thyroid stimulating
Pathogenesis effect of TSI. Notably, destructive thyroiditis may coexist
with Graves’ disease in a minority of cases, as suggested by
Thyroid dysfunction is the most common endocrine adverse TRAb positivity [10, 11].
effect associated with ICI therapy. Most studies report two Individual genetic susceptibility to thyroid dysfunction
patterns of ICI-related thyroid dysfunction: thyrotoxicosis also plays an important role. More specifically, thyroid
followed by hypothyroidism and isolated hypothyroidism. dysfunction has been shown to be associated with over-
However, the pathophysiological basis that appears to be expression of Human Leukocyte Antigen DR-isotype
common is destructive thyroiditis [5–7, 9]. The underlying (HLA-DR). Treatment with ICI therapy can change HLA-
pathophysiology is considered to be an immune-mediated DR expression, increasing T cell activation and leading to
acute inflammation followed by destruction of the thyroid thyroid autoimmune disease. Delivanis et al. conducted a
gland. ICI therapy induces autoimmune side effects through study examining healthy volunteers, patients with auto-
T cell activation and is characterized by intra-thyroidal immune thyroiditis, and individuals with pembrolizumab-
predominance of CD8+ and CD4-CD8- T lymphocytes induced thyroiditis [21]. The study revealed an elevated
[1, 8]. In a case report of a nivolumab-related hypothyr- count of CD56 + CD16+ Natural Killer (NK) cells and
oidism, the anti-PD-L1 therapy suppressed the inhibitory increased surface HLA DR expression on inflammatory
PD-1/PD-L1 signals on follicular helper T cells (Th), intermediate CD14 + /CD16+ monocytes in patients with
leading to increased proliferation and subsequent over- pembrolizumab-related thyroiditis. Comparing the PD-1
production of thyroid autoantibodies [16]. levels on peripheral T-cells among the three groups, they
In accordance with the above, recent studies suggest that were undetectable on the surface of T-cells in those with
autoantibodies against thyroid peroxidase (TPOAb) and pembrolizumab-induced thyroiditis, while they were com-
thyroglobulin (TgAb) have been found elevated at baseline in parable between healthy volunteers and patients with
some patients who develop thyroid dysfunction after ICI autoimmune thyroiditis. Thus, macrophage activation
immunotherapy. TPOAb and TgAb may be present at base- through up-regulation of HLA-DR may be a possible
line prior to or may develop after ICI therapy. Maekura et al. mechanism of pembrolizumab-induced thyroiditis. In addi-
studied the levels of TPOAb and TgAb in 53 patients treated tion to T and B lymphocytes, various cytokines play an
with nivolumab for non-small cell lung cancer (NSCLC) in essential role in the development of thyroid disorders. First
an attempt to predict the occurrence of hypothyroidism [17]. and foremost, elevated interleukin (IL)-2 levels facilitate the
Among the nine patients who tested positive for TPOAb at binding between HLA-II and thyroid cell autoantigen,
baseline, 44% (4 out of 9) developed ICI-related hypothyr- leading to stimulation of CD8+ cytotoxic T lymphocytes
oidism, compared to 2% (1 out of 44) in those who were TPO (CTL) and subsequent thyroid cell death. According to
Ab negative at baseline. Among the nine patients who had recent studies PD-L1 therapy increases CD4 + Th1 and
positive TgAb at baseline, 56% (5 out of 9) developed ICI- therefore the expression of interferon gamma (IFN-γ) and
related hypothyroidism, while no one out of the 44 patients IL-2, which leads to thyroid cell death. Kurimoto et al.
who were TgAb negative at baseline experienced the same. In measured the changes of various cytokines before and after
the Osorio et al. study, TPOAb and TgAb levels were ICIs treatment and identified that an increase in IL-2 and a
examined not only at baseline, but also during pem- decrease in granulocyte colony-stimulating factor (G-CSF)
brolizumab (anti-PD-1) treatment [18]. Additionally, accord- appeared to be associated with thyroid immune-related
ing to the study of Muir et al. anti-thyroid antibodies basal adverse events (irAEs) [22]. Regulatory T cells (Tregs)
positivity is associated with increased possibility of devel- enhance the response to anti-PD-L1 therapy by releasing
oping a thyroid disorder. The risk of overt thyrotoxicosis is cytokine IL-10. Conversely, Treg inhibition through ICI
higher when the title of TPOAb or/and TgAb is remarkably therapy is implicated in the development of autoimmune
elevated or when there is a newfound thyroid antibody thyroid diseases [23] (Table 1).
Table 1 Thyroid dysfunction linked with particular ICI-therapy for specific cancer subtypes
Study Publication Type of study Objective Cancer type Type of ICI-therapy Thyroid disorder induced Outcomes/Findings
Endocrine

year

D.L. Morganstein 2016 Retrospective Evaluate thyroid dysfunction and its Melanoma Anti-PD-L1, anti-PD-1, anti-CTLA- Hypothyroidism, thyrotoxicosis TD can affect as many as half of the
et al. [5] observational study progression in ICI-treated melanoma 4 or combination of ipilimumab- patient. F > M
patients. nivolumab
Lee H. et al. [6] 2017 Cohort study Comparison of thyroid disorders between Solid and hematologic Anti-PD-1 or anti-PD-1 and anti- Hypothyroidism, thyrotoxicosis Most common ir-TD: thyroiditis.
patients receiving different treatment malignancies CTLA-4 combination Εarlier onset of thyrotoxicosis-,
regimens hypothyroidism later
Jeroen de Filette 2016 Observational, cohort Examine the occurrence and attributes of Melanoma Pembrolizumab (anti-PD-1) Thyrotoxicosis, hypothyroidism Thyrotoxicosis is associated with diffuse
et al. [9] study thyroid dysfunction related to increased 18 FDG uptake
pembrolizumab
DA Delivanis 2017 Single- Investigate the incidence and the possible Metastatic melanoma, Pembrolizumab (anti-PD-1) Destructive thyroiditis and overt The mechanism of thyroid damage seems
et al. [21] center, retrospective causes of thyroid irAEs triggered by anti- NSCLC hypothyroidism unrelated to thyroid autoantibodies and
cohort study PD-1 treatment might involve pathways influenced by T
cells, NK cells, and/or monocytes.
SA Paschou et al. [33] 2022 Cohort study Investigate the relation of endocrine irAE Various cancer types: Anti-PD1, anti-PD-L1 Non-specific Endocrinopathies induced by ICIs could
with PFS and OS bladder, renal, lung, potentially serve as indicators of a
ovarian cancer favorable response to immunotherapy.
Osorio JC et al. [18] 2016 Observational cohort Examine TD in NSCLC patients treated NSCLC Anti-PD-1 Transient thyrotoxicosis, Thyroid dysfunction occurs early in the
study with pembrolizumab, association with hypothyroidism pembrolizumab course and may be
antibodies and impact on survival associated with improved outcomes.
F. Guaraldi et al. [7] 2017 Long-term prospective Evaluation of the occurrence and Melanoma Anti-CTLA-4 (ipilimumab), Anti- Hypothyroidism, thyrotoxicosis ICIs are associated with high occurrences
single-arm study significance of ICI-induced thyroid PD1 (nivolumab, pembrolizumab) of severe autoimmune TD. Autoimmune
disorders in actual clinical practice conditions and BRAF mutation are linked
to improved clinical responses after anti-
CTLA-4 followed by anti-PD1
Olsson-Brown A 2020 Retrospective Characterization of the clinical patterns of Metastatic malignant Anti-PD1 mono/combination with hypothyroidism, thyrotoxicosis No correlation between clinical pattern of
et al. [25] observational study thyroid dysfunction to cancer patients melanoma anti-CTLA-4 followed by hypothyroidism dysfunction and thyroid antibodies,
treated with ICIs gender association: F > M
Difei Lu et al. [26] 2022 Retrospective analysis Analysis of ICI-TD prevalence and broad subtype of Anti-PD1, anti-PD-L1, anti-CTLA-4, Hypothyroidism, thyrotoxicosis ICI-TDs are presented as either
demographics, along with identification malignancies combination hypothyroidism or thyrotoxicosis,
of poor clinical outcome risk factors, particularly in individuals undergoing
using FAERS data. combination therapy.
Husebye ES et al. [1] 2022 Guideline Provision of evidence-based Broad subtype of tumors Anti-PD-1/PD-L1, anti-CTLA4 Hypothyroidism, thyrotoxicosis, Provision of practical guidance and
recommendations for treatment and as ICIs have been Graves’ disease and thyroid eye recommendations on the management of
follow-up for ICI-induced approved in 18 cancer disease patients with ICI-related endocrine
endocrinopathies addressing the gaps in types conditions.
existing guidelines
Toshiya Maekura 2017 Research study Evaluation of thyroid dysfunction and NSCLC Nivolumab (anti-PD1) Hypothyroidism Assessing TPO and TgAb at the outset
et al. [17] identification of the predictive factors of could be predictive for the development
nivolumab-induced hypothyroidism of hypothyroidism in NSCLC patients.
Yang S et al. [38] 2020 Observational cohort Examination of autoimmune disease risk Lung cancer Nivolumab, Pembrolizumab (PD-1- Hypothyroidism Patients under ICI-therapy were 1.97
study in lung cancer patients under ICI- inhibitors) more likely to experience autoimmune
treatment versus chemotherapy, with a diseases within the initial 6 months
specific focus on hypothyroidism compared to chemotherapy.
Anupam Kotwal 2020 Single-center Investigation of immune mechanisms and Diverse malignancies Anti-PD-1 Thyrotoxicosis, overt Thyroiditis induced by ICIs is associated
et al. [8] prospective cohort genetic factors associated with TD caused (most common: Anti-PD-L1 hypothyroidism following with a notable presence of CD8+ and
study by ICIs, focusing on PD-1 inhibitors melanoma) thyrotoxicosis, primary CD4 − CD8 − T lymphocytes to the
hypothyroidism thyroid tissue. HLA haplotypes might
play a role.
Kurimoto et al. [22] 2020 Prospective clinical Identification of predictive and sensitive Melanoma, RCC, Anti-PD1 (pembrolizumab, Thyrotoxicosis, hypothyroidism Potential biomarkers: serum Tg, TgAb,
observational research biomarkers in thyroid irAE. NSCLC, UTUC, gastric nivolumab), anti-CTLA-4 TPOAb, IL-1β, IL-2, GM-CSF, IL-8,
cancer (ipilimumab) or combination MCP-1

TD thyroid disorder, irAE immune-related adverse events, irTD immune related thyroid disorders, NK cells Natural killer cells, PFS progression-free survival, OS overall survival, Anti-PD1 anti-
programmed death r-1, Anti-PD-L1 anti-programmed death ligand-1, Anti-CTLA-4 anti-cytotoxic T-lymphocyte-associated protein 4, ICI-TD immune checkpoint inhibitor-related thyroid
dysfunction, TD thyroid disorders, FAERS FDA Adverse Event Reporting System, UTUC urothelial carcinoma, RCC renal cell carcinoma, NSCLC non-small cell lung cancer, F > M females>
Males, Tg thyroglobulin, TgAb thyroglobulin antibodies, TPOAb thyroperoxidase antibodies
Endocrine

Clinical presentation as 3 years [6, 13, 21, 33–35]. Thyroid dysfunction is mainly
associated with anti-PD-1 monotherapy and its combination
Clinical diagnosis of thyroid toxicity is challenging, as with PD-L1 or/and anti-CTLA-4 therapy rather than with
patients may not exhibit any noticeable symptom or sign or anti-CTLA-4 or anti-PD-L1 monotherapy [19, 27, 36].
present with non-specific symptoms [24]. However, due to CTLA-4 inhibitors are mostly correlated with the possibility
the accessibility of thyroid function screening, thyroid dis- of developing hypothyroidism, while PD-1/PD-L1 inhibi-
orders are frequently identified at an early stage, even when tors can lead to thyrotoxicosis and hypothyroidism [19].
patients do not display typical symptoms. The spectrum of More specifically, thyrotoxicosis induced by anti-CTLA-4
thyroid disorders induced by ICIs includes thyroiditis, affects 0.2–1.7% of the patients, while thyrotoxicosis
hypothyroidism, Grave’s disease and thyrotoxicosis [6]. induced by anti-PD-1 affects o.6-3.7% of the patients. ICIs
Most cases involve thyroiditis processing to hypothyroid- combination is responsible for 8–11% of the cases of
ism [4]. Although hypothyroidism is typically permanent in thyrotoxicosis. Hypothyroidism is induced by CTLA-4
most cases, it is currently impossible to determine the ratio inhibitors in 2.5–5.2% of the cases, by PD-1/PD-L1 inhi-
of transient hypothyroidism compared to permanent cases bitors in 3.9-8.5% and by the combination of anti-CTLA-4
[25, 26]. and anti-PD-1 in 10.2–16.4% of the patients [37]. Various
For the majority of symptomatic patients, the first man- factors, including cancer subtype, the ICI dosage, and age,
ifestation is thyrotoxicosis [6]. Thyrotoxicosis usually pre- influence the risk of thyroid ICI side effects, with current
sents with weight loss, palpitation, tremors, anxiety, fatigue conflicting results regarding age and sex hormones [38, 39].
and sweating [27]. In addition, increased perspiration, heat ICIs induce thyroid dysfunction more frequently in women
intolerance, hyperdefecation and generally increased meta- than in men [28].
bolic activity are clinical manifestations that should raise
suspicion of thyrotoxicosis [28, 29]. Physical examination
sometimes reveals increased heart race and warm skin [13]. Thyroid disorders induced by ICIs and cancer
Atrial fibrillation may be seen, especially in older patients prognosis
[30]. Initial presentations of overt or subclinical thyrotox-
icosis typically resolve to euthyroidism or hypothyroidism Several studies have shown that cancer patients undergoing
within several weeks to months. ICI treatment who develop immune-related adverse effects
Primary hypothyroidism is a thyroid adverse effect that is (irAEs), particularly thyroid dysfunction, often exhibit
more commonly noticed in patients treated with PD-1 improved prognosis. Combining the results of the retro-
inhibitors [31]. Hypothyroidism can be detected during spective studies conducted by Prather et al. and Trudu et al.,
routine lab monitoring in asymptomatic patients or clini- it is suggested that lung cancer patients experiencing irAEs
cally presents with the typical symptoms such as weight had longer progression-free survival (PFS) and improved
gain, depression, profound fatigue, alopecia, cold intoler- overall survival compared to those without these side
ance, constipation, dry skin, bradycardia, periorbital edema effects. This implies that irAEs may serve as potential
and tongue swelling [4, 29, 30]. While most cases are mild indicators of enhanced treatment efficacy [40, 41]. Simi-
to moderate, untreated severe hypothyroidism can lead to larly, Zheng et al. reported that 47% of hepatocellular car-
myxedema coma, decreased mental status and often hypo- cinoma (HCC) patients treated with anti-PD-1 therapy
thermia [30]. developed thyroid dysfunction [42]. The survival rates
Graves’ disease induced by ICIs is primarily associated showed no significant difference between the group with
with CTLA‐4 gene polymorphisms [32]. While it is extre- normal thyroid function and the one with abnormal thyroid
mely rare, cases have been reported. It appears usually at the function [42]. Han-Sang Baek also revealed that individuals
beginning of the treatment. The presence of thyroid eye experiencing with irAEs, in particular hypothyroidism,
disease increases the possibility of Graves’ disease, while demonstrated a more favorable prognosis compared to those
signs such as orbitopathy or a large goiter enhance its without irAEs. This association remained irrespective of
diagnosis [27, 33]. factors such as age, sex, type of ICI used, and cancer type
[43]. Studies by Kotwal et al. and Lima Ferreira et al. also
observed improved survival in patients with thyroid dys-
Epidemiology function across different cancer types and ICI therapies
[44, 45]. However, these studies did not distinguish
Thyroid disorders may occur even after the first single between different types of thyroid dysfunction. In contrast
therapeutic dose of ICI therapy [6, 13]. The median time of to these findings, a case study highlighted a lung cancer
the onset is 6–10 weeks after the initiation, but it may patient who developed ICI-related thyroid dysfunction,
happen as early as 7 days post therapy initiation and as late leading to tumor progression and preventing surgical
Endocrine

intervention. This suggests that thyroid dysfunction does the diagnosis is established by the positivity TSI and/or
not uniformly indicate a better response to ICI treatments thyrotropin receptor antibodies [12]. In contrast to screening
[43, 46]. This becomes more intricate as studies propose of thyroid function, the screening for TPO-antibodies before
that thyrotoxicosis could potentially exacerbate cancer the initiation of ICIs is not recommended [34]. Additionally,
prognosis [47]. Von Itzstein et al. noted poorer outcomes in since some cases of Graves’ disease have been reported
patients with pre-existing thyroid dysfunction but also with normal levels of TSH receptor antibodies, ultra-
observed that initiating levothyroxine after beginning ICI sonography and scintigraphy/gamma scan are indicated
treatment improved overall survival [48]. This indicates that [12, 24]. Doppler ultrasound in Graves’ disease typically
pre-existing thyroid problems might negatively impact the reveals increased blood flow, presenting as high vascularity
effectiveness of ICI therapy and should be managed ade- in the thyroid gland [27]. Thyroid uptake tests are recom-
quately before starting ICI treatment. mended for patients with high likelihood of developing
Grave’s disease and who have not been exposed to intra-
venous CT contrast for at least 1 month, as this exposure
Laboratory and imaging evaluation may reduce thyroid iodine uptake [6, 27]. Radioactive
iodine uptake is a valuable tool in differentiating Graves’
The evaluation of TSH and fT4 is recommended before the disease from other causes of thyrotoxicosis such as
initiation of ICI therapy, in order to rule out the possibility destructive thyroiditis [51]. It involves administering a
of any preexisting thyroid disorder [12]. Thyroid function diagnostic dose of iodine-123 orally, followed by measuring
screening, involving TSH and fT4 measurements, should be its absorption by the thyroid gland either 6 or 24 h later.
conducted every 4–6 weeks, or more frequently, if neces- Graves’ disease is characterized by high, diffuse, homo-
sary, for all the patients undergoing ICIs treatment genous iodine uptake and elevated titers of TRAbs, while
[4, 12, 35]. Monitoring of the patients on ICIs immu- destructive thyroiditis is associated with low iodine uptake
notherapy should occur 4–6 weeks after the completion of and the presence, though not elevated, titers of TRAb
the last cycle [49]. Some clinicians suggest the measure of [15, 24, 27, 50]. Characteristic ultrasonic indicators of ICI-
TSH and fT4 before every circle of treatment [14, 35]. induced destructive thyroiditis include widespread thyroid
ICI-induced hypothyroidism is characterized by gland enlargement, decreased internal blood flow, and
decreased levels of free thyroxine (fT4) [28]. The assess- reduced internal echogenicity (Table 2).
ment of TSH is a more sensitive test [35]. Elevated TSH
with low or low-to-normal fT4 set the diagnosis of primary
hypothyroidism, while low or inappropriately low-to-mid- Treatment
normal TSH levels with low fT4 indicate secondary/central
hypothyroidism attributed to pituitary disorder, such as Multiple organizations have proposed various treatment
hypophysitis [12, 30, 50, 51]. In this case, measuring cor- approaches for thyroid immune-related adverse events
tisol is indicated to assess for adrenal insufficiency [30]. (irAEs) caused by ICIs, such as American Society of
Primary hypothyroidism may be subclinical. Subclinical Clinical Oncology (ASCO), National Comprehensive Can-
hypothyroidism is diagnosed when TSH is elevated but cer Network (NCCN), Society for Immunotherapy of
below 10 mIU/mL accompanied by a normal free T4 level Cancer (SITC) and European Society for Medical Oncology
[4]. For cases of hypothyroidism, including TPO-antibody (ESMO) (Table 3). These guidelines demonstrate a high
testing is prudent as it indicates an autoimmune origin [34]. level of agreement [13, 52]. The severity of each side effect
Overt thyrotoxicosis is characterized by suppressed TSH is classified into five grades based on the Common Termi-
serum levels and elevated fT4 and/or total triiodothyronine nology Criteria for Adverse Effects (CTCAE) established
(TT3), while subclinical thyrotoxicosis is defined as sup- by the National Cancer Institute (NCI) of the National
pressed TSH with normal fT4 and TT3 serum levels [9]. Institutes of Health (NIH).
Due to the potential process to subclinical hypothyroidism, In cases of hypothyroidism, asymptomatic patients with
a repeat thyroid function evaluation test should be per- mild thyroidopathy (TSH 4–10 mIU/l), normal FT4 (grade
formed 6 weeks after the initial diagnosis [4]. Guidelines 1) can continue ICI therapy, with regular TSH monitoring
recommend TSH receptor-antibody testing in case of thyr- every 4–6 weeks. Ιn patients having moderate thyroido-
otoxicosis [34]. pathy (TSH over 10 mIU/l or TSH 4–10 mIU/l) with low
Suspecting Graves’ disease is raised when thyroid hor- FT4, with or without symptoms, such as constipation, cold
mones (T4 and T3) levels are significantly high, initial intolerance, fatigue, disturbances in the menstrual cycle,
symptoms are prominent, thyrotoxic manifestations persist arthralgias, myopathy, pale and/or dry skin, thin brittle hair
for over 6 weeks and other characteristic signs, including or fingernails, depression signs, weight gain, weakness, etc
orbitopathy or a large goiter, are present. For those patients, (grade 2), ICI therapy should be ceased for a while, until the
Endocrine

Table 2 Clinical presentation, laboratory and imaging evaluation of thyroid disorders


Thyroid Clinical symptoms Physical *TSH **fT4, ***T3 Anti-thyroid antibodies Imaging study
disorder examination levels

Hypothyroidism Fatigue, weight gain, cold Bradycardia, Elevated Normal, low Anti-TPO (thyroid (not necessary)
intolerance, constipation, hypothermia TSH f4, fT3 peroxidase) testing is
depression, dry skin, facial recommended as it
puffiness, periorbital edema, suggests an autoimmune
tongue swelling origin
Thyrotoxicosis Weight loss, anxiety, fatigue, Tremors, Decreased Elevated fT4 Possibility of positive Radioiodine uptake testing:
increased frequency of bowel warm and or and/or T3 (+) TPO-TG Ab decreased uptake
movements smooth skin, suppressed [levels of fT4 TSH receptor antibodies
palpitations, TSH are more testing is recommended
lid lag elevated
compared to
T3 levels]
Graves´ disease Ophthalmopathy/ orbitopathy Thyroid bruit, Low TSH significantly **** [Positive (+) TSH Doppler ultrasound reveals
(conjunctival redness, eye pain) large goiter elevated receptor antibodies increased blood flow.
(TRAb)/TSI], Radioactive iodine uptake:
possibility of positive increasing and
(+) TPO-TG Ab homogeneously diffuse

TSH thyroid-stimulating hormone, fT4 free thyroxine, T3 Triiodothyronine


*TSH normal levels: 0.4–4 mUI/L
**fT4normal levels: 12–22 pmol/L
***T3 normal levels: 3.1–6.8 pmol/L
**** Cases of Graves’ disease without elevated levels of TSH have been described

irAEs subside [13]. These patients should start with a use of I-131 [13]. For cases of thyrotoxicosis, individuals
replacement dose of approximately 1,3 μg/kg/day levo- with mild thyroidopathy (TSH less than 0.4 mIU/l), whether
thyroxine. Special attention should be paid to the elderly symptomatic or asymptomatic (grade 1), can proceed with
patients or those with cardiovascular compromise, begin- ICIs therapy, with regular monitoring of TSH, FT4, T3 every
ning with a lower daily dose of 25–50 μg and gradually 2–3 weeks [13, 53]. Patients with moderate thyroidopathy,
increasing it or simply reducing the default dose by low TSH and moderate symptoms, such as heat intolerance,
approximately 10%, in order to avoid the risk of thyrotox- attention deficit disorder, gastrointestinal (GI) hypermotility,
icosis [13, 14]. TSH should be assessed every 6 weeks to insomnia, hair loss, tremor, anxiety etc., (grade 2) should
determine the appropriate dose, whereas FT4 can be eval- temporarily discontinue ICIs until the resolution of adverse
uated to certify the adequacy of the default dose [13, 53]. effects and simultaneously initiate a beta blocker therapy
Patients with severe thyroidopathy and symptoms like [13, 53]. Methimazole or propylthiouracil (PTU) may be
puffiness of face, low temperature, low heart rate, slow administered if thyrotoxicosis persists for over 6 weeks in
speech (grade 3) are managed similarly to patients in grade these patients [13, 53]. In case of persistent thyrotoxicosis, it
2 with the only difference that expert opinion from an is also prudent to investigate Graves’ disease (TSH receptor
endocrinologist is necessary, while at previous grades just autoantibodies-TRAb). TSH, FT4, T3 should be measured at
recommended. Patients with life-threatening thyroidopathy, these patients every 3–4 weeks and the expert’s care
exhibiting severe symptoms that could lead to death (grade (endocrinologist) should be provided [53]. The management
4), are treated as mentioned above [13]. However, physi- of patients with severe thyroidopathy and severe symptoms
cians remain particularly vigilant for signs of myxedema, like tachycardia and palpitations (grade 3) follows the same
such as progressive weakness, stupor, hypothermia, hypo- approach as for the previous stage. The first line treatment
ventilation, hypoglycemia and hyponatremia. In such cases, remains beta blocker and PTU/methimazole [13]. Corticos-
hospitalization for intravenous treatment is necessary. teroids are also acceptable at this stage, despite the lack of
Levothyroxine treatment is recommended until the TSH strong evidence for their efficacy [53]. Patients with life-
levels are restored to normal. Adrenal insufficiency threatening thyroidopathy and symptoms severe enough to
should be ruled out if suspected [14, 51]. potentially lead to death (grade 4) are treated similarly, with
the distinction that hospitalization may be warranted, when
suspecting an upcoming thyroid storm [13]. Furthermore, a
If the physician suspects an underlying thyrotoxicosis, common and widely accepted approach at this point involves
patients should undergo thorough monitoring to exclude the the administration of 1–2 mg/kg/d or equivalent prednisone
possibility of destructive thyroiditis, which requires a distinct during a brief titrating period of 1–2 weeks. Saturated
management approach. In such instances, due to their solution of potassium iodide or PTU/methimazole can also
potential to turn to hypothyroidism, it is prudent to avoid the be used [53].
Table 3 Guidelines for ICI-induced thyroid disorders’ treatment
ESE 2022 [1] ESMO 2021 ASCO 2022[15] SCIT 2017 [54] British Society IDSC 2018 [52] French Endocrine Society 2019 NCCN 2019 [14] Japanese Endocrine
Endocrine

[13] for [56] Society 2019 [55]


Endocrinology
2018 [56]

Hypo- Search for TPO-Abs Grade 1*: Grade 1*: Grade 3*** and 4 ****: If myxedema is If ICI therapy ICI therapy is not contraindicated, Grade 1*: Continue Hold ICI therapy;
thyroid- (autoimmunity) ; Give Continue ICI Continue ICI Cease ICIs; suspected, includes CTLA4 but can be carried over for later; If ICI therapy; monitor Administer an initial
ism levothyroxine until TSH therapy; monitor therapy; monitor hospitalization may be advise an blockade and TSH TSH > 10 miU/L, administer TSH every 4–6 dose of 25–50 μg
levels are normal TSH every 4–6 TSH every 4–6 necessary for grade 4**** endocrinologist; is normal or low, levothyroxine at an initial dose weeks; levothyroxine per day
weeks. weeks. patients. Exclude first exclude firstly 1–1.6 µg/kg/day; Adjust the dose Grade 2: ** except from elderly or
Grade 2: ** Grade 2: ** Grade 2: ** adrenal crisis secondary adrenal according to patient’s age, Cease ICIs until patients with cardiac
Cease ICIs until Hold ICIs until Postpone ICIs for symptomatic and then insufficiency, after comorbidities and survival adverse effects problems who should
adverse effects adverse effects patients, until symptoms investigate T4; euthyroid sick prognosis; If TSH 5–10 miU/L and subside; Start start with a dose of
subside; start subside; start resolve. If in doubt, treat syndrome; anti-TPO antibodies or symptoms ~1.6 mcg/kg/day 12,5 μg; Continue
~1.1 μg/kg/day levothyroxine if Give 1.6 mcg/kg levothyroxine for cortisol otherwise are present, consider giving levothyroxine (For treatment adjusting
levothyroxine. symptomatic or if (except from elderly and deficiency levothyroxine is levothyroxine; In case of elderly and patients the dose in regard to
(For elderly or TSH >10 mIU/L patients with cardiovascular indicated hypothyroidism following a with cardiovascular the serum TSH levels
cardiovascular persists, monitor comorbidities who should start destructive thyroiditis, thyroid comorbidities:
comorbidities: TSH every 6 with a lower dose of 25–50 μg) hormones may be needed lifelong reduce dose by
gradually weeks, adjusting and monitor TSH and fT4 ~10%); Advise
increase the dose until every 6-8 weeks until normal endocrinologist;
25–50 μg initial TSH returns to TSH levels achieved; If TSH Grade 3: ***
dose); monitor normal. remains high, increase Hold ICIs in any case
TSH every 6 check FT4 to levothyroxine dose by and manage similarly
weeks and FT4 confirm dose 12.5 mcg to 25 mcg; maintain as mentioned right
to confirm dose sufficiency. the dose; reassure after 12 above;
sufficiency. Once successfully months or earlier if the Grade 4: ****
Grade 3: *** treated, resume patient’s Manage similarly as
Manage ICI therapy. condition alters mentioned right
similarly as in Monitor THs above;
Grade 2**; every 6 weeks
advise during therapy
endocrinologist. and yearly after
Grade 4: **** therapy.
Manage Grade 3: ***
similarly as in Manage similarly
Grade 2**; as in Grade 2**;
hospitalization advise
may be endocrinologist.
necessary for Grade 4: ****
i.v. treatment in Manage similarly
the case of as in Grade 2**;
myxedema. hospitalization
may be necessary
for i.v. treatment
in case of
myxedema.
Thyro- Search for TRabs - Grade 1*: Grade 1*: If thyrotoxicosis is suspected, If thyrotoxicosis Differentiate Treatment based on appointment Give non-selective b Hold ICI therapy;
toxico- exclude Grave’s; Continue ICI Continue ICI exclude Grave’s disease firstly is suspected or thyroiditis from between the oncologist and blockers to alleviate give b blockers as
sis administer non-selective therapy. therapy, (requires thionamides); is present, Grave’s; Monitor endocrinologist; ICI therapy is not symptoms, symptoms relievers
beta-blocker to alleviate Monitor TSH, monitor TSH, Management without many provide closely TSH, T4, contraindicated, but can be carried glucocorticoids in e.g., propranolol
symptoms;Give FT4, T3 every FT4, T3 every medical interventions supportive care T3 in case of over for later; high dosages and 30 mg per day
corticosteroids in high 2–3 weeks. 2–3 weeks, effectively; give non-selective in an ICU/ a hypothyroidism if asymptomatic, no intervention, PTU. Monitor TSH
dosages; if symptoms Recognize recognize b blocker (with alpha blocker HDU and appearance or just close monitoring; and FT4 levels 4–6
persist ( > 6 weeks), destructive destructive action preferably) in endocrinolo- persistence of if symptoms are present, administer weeks after the crisis;
search again for TRAb, thyroiditis or thyroiditis or symptomatic patient; TFT test gist’s thyrotoxicosis; if b blocker: consider corticosteroids if abnormal,
test fT3, conduct persistent persistent every 2 weeks; in case supervision thyrotoxicosis for severe cases. administer I-123 or
ultrasound or scintigraphy thyrotoxicosis thyrotoxicosis evolvement into persists, check for Give levothyroxine while scan to identify the
to exclude Graves’ ; if Grade 2**: Grade 2**: hypothyroidism, TSI or scan for continuing the ICI therapy after cause of
Hold ICIs until Hold ICIs until levothyroxine; iodine uptake thyrotoxicosis, reduce gradually the thyrotoxicosis and
Table 3 (continued)
ESE 2022 [1] ESMO 2021 ASCO 2022[15] SCIT 2017 [54] British Society IDSC 2018 [52] French Endocrine Society 2019 NCCN 2019 [14] Japanese Endocrine
[13] for [56] Society 2019 [55]
Endocrinology
2018 [56]

Grave’s give adverse effects adverse effects Cease ICIs at grade 3*** and dose as long as the therapy comes rule out Graves’
antithyroid drugs subside; subside; grade 4**** patient. At to an end; simultaneously close disease; if
administer a administer beta- grade2**, hold ICIs until monitor TSH levels thyrotoxicosis turns
beta-blocker and blocker and adverse effects subside; Advise into hypothyroidism,
methimazole if methimazole if endocrinologist; give 1.6 mcg/kg/day
thyrotoxicosis needed, levothyroxine orally
persists; measure TSH, until TSH reaches
measure TSH, FT4, T3 every normal levels
FT4, T3 every 3–4 weeks,
4–6 weeks; advise
advise endocrinologist;
endocrinologist. hydrate and
Grade 3 ***: supportive care;
Treat similarly Investigate for
as Grave’s if
mentioned abov- thyrotoxicosis
e. persists > 6
Consider weeks;
corticosteroids. Grade 3 ***:
Grade 4 ****: Treat as in Grade
Hospitalization 2**, consider
may be needed corticosteroids.
for i.v. Grade 4 ****:
treatment. Hospitalization
Start 1 to 2 mg/ may be necessary,
kg/d prednisone start 1 to 2 mg/kg/
while titrating d prednisone
for 1 to 2 weeks. while titrating for
Consider 1 to 2 weeks;
saturated Saturated solution
solution of of potassium
potassium iodide or PTU/
iodide or PTU/ methimazole can
methimazole. also be used
Cease ICI at
grade 3 and
grade 4****.
Thyr- n/a recommendation for Hold ICIs only n/a n/a n/a n/a Hold ICI therapy in case of severe n/a n/a
oid-like holding or continuing in severe forms orbitopathy; the decision to
eye ICIs; Give systemic of TED; give continue the therapy should be
disease corticosteroids in high systemic taken personalized for each case
(TED) dosages; corticosteroids
in high dosages;

Severity grades are based on the Common Terminology Criteria for Adverse Effects (CTCAE) established by the National Cancer Institute (NCI) of the National Institutes of Health (NIH):
* TSH < 4-10mIU/l, FT4: normal and without symptoms
** TSH >10mIU/l or TSH 4–10 mIU/l with low FT4 with or without symptoms
*** TSH>10mIU/l or TSH 4–10 mIU/l with low FT4 and severe symptoms
**** TSH > 10mIU/l with very severe symptoms (life threatening)
n/a non-available, ICU intensive care units, HDU high-dependency care units, PTU propylthiouracil, ICI immune checkpoint inhibitor, FT4 free thyroxine, TSH thyroid-stimulating hormone, T3
triiodothyronine, TSI thyroid-stimulating immunoglobulin, TRab TSH Receptor antibody, fT3 free triiodothyronine
Endocrine
Endocrine

Fig. 1 Classification of thyroid adverse effects after ICIs therapy


Endocrine

Table 4 Recommendations for managing ICI-induced thyroid disorders


Side effect Severity Management recommendations

Hypothyroidism • TSH 4–10 mIU/l, FT4: Continue ICI therapy; Monitor TSH every 4–6 weeks
normal
• without symptoms
• TSH > 10 mIU/l or TSH Stop ICIs if symptoms are present until they subside; Start levothyroxine;
4–10 mIU/l, low FT4 Measure TSH every 6 weeks for dose adjustment; Monitor fT4 in the meantime (every
• with or without symptoms 2 weeks) to ensure that the management is appropriate
• TSH > 10 mIU/l or TSH Manage similarly as right above; Advise endocrinologist
4–10 mIU/l, low FT4
• severe symptoms
• TSH > 10 mIU/l Manage similarly as right above; Hospitalization may be needed for i.v. treatment if
• very severe symptoms (life myxedema appears
threatening)
Thyrotoxicosis • TSH < 0.4 mIU/l Continue ICI therapy; Monitor TSH, fT4, T3 every 2–3 weeks (for early diagnosis of
• without or with mild destructive thyroiditis or persistent thyrotoxicosis)
symptoms
• TSH < 0.4 mlU/l Stop ICIs if symptoms are present until they subside; Consider b blockers and methimazole
• moderate symptoms If thyrotoxicosis persists (>6 weeks), investigate for Grave’s searching TRab, testing fT3,
conducting thyroid scintigraphy or ultrasound; hydrate and provide supportive care
• TSH < 0.4 mlU/l Manage similarly as mentioned right above; Corticosteroids may be used
• severe symptoms
• TSH < 0.4 mlU/l Manage similarly as mentioned right above; Hospitalization may be needed for thyroid
• very severe symptoms (life storm concern
threatening)
Thyrotoxic crisis Hold ICIs; Give non-selective b blockers, high-dose glucocorticoids and PTU; monitor
TSH and FT4 levels 4–6 weeks after the crisis; If normal, the management is completed. If
not, search for TRAb, even test fT3, administer I-123 or conduct a scan to identify the cause
of persistent (>6 weeks) thyrotoxicosis and exclude Graves’ disease
Thyroid-Like Eye Hold ICIs only in severe forms of TED; Give systemic corticosteroids in high dosages;
Disease (TED) Canthotomy/cantholysis for severe cases
PTU propylthiouracil, fT4 free thyroxine, TSH thyroid-stimulating hormone, ICI immune checkpoint inhibitor, TRab TSH Receptor antibody, fT3
free triiodothyronine

In case of thyroid storm, it is recommended to administer a scan to identify the cause of persistent (>6 weeks) thyrotox-
non-selective beta blocker, typically propranolol, at doses of 40 icosis exclude Graves’ disease [13, 14, 52, 54]. In case of
to 80 mg every four to six hours, high dose glucocorticoids, to Grave’s, antithyroid drugs (methimazole or PTU in the first
prevent the peripheral conversion of T4 to T3, and a starting semester of pregnancy) and beta blocker should be adminis-
dose of 500 to 1000 mg of propylthiouracil (PTU) continuing tered—avoid the use of I-131 as it increases the risk of
with a subsequent dose of 250 mg every four hours [13, 14]. hypothyroidism. If thyrotoxicosis turns into hypothyroidism,
Both PTU and methimazole block the production of thyroid levothyroxine should be given at a dose of 1.6 mcg/kg/day
hormones. Nonetheless, PTU is preferred over methimazole in orally until TSH reaches normal levels [12, 14, 54]. Another
this case, because of its additional effect of blocking peripheral approach of managing a thyrotoxic crisis involves minimal
conversion of T4 to T3. Immunotherapy can be continued only medical interventions, with beta blockers administered only in
if the patient is asymptomatic, while TSH and FT4 levels presence of symptoms and more frequent reassessments of
should be monitored 4–6 weeks after the crisis [14]. If they TSH, FT4, i.e., typically every 2–3 weeks [54–57].
normalize, the management can be considered complete. If not, In case of thyroid-like eye disease (TED), high dose
it is recommended to search again for TSH receptor auto- systemic corticosteroids are strongly recommended for their
antibodies-TRAb, even test fT3, administer I-123 or conduct a potent anti-inflammatory effect. In instances where the
Endocrine

orbital thyroidopathy persists, canthotomy/cantholysis may Funding Open access funding provided by HEAL-Link Greece.
be considered. As for the ICIs treatment, it should only be
paused in cases of severe TED [58] (Fig. 1 and Table 4). Compliance with ethical standards

Conflict of interest The authors declare no competing interests.


Conclusion
Publisher’s note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
The ICI therapy has revolutionized cancer treatment by
harnessing the immune system to fight malignancies. While Open Access This article is licensed under a Creative Commons
offering promising therapeutic results and improved survi- Attribution 4.0 International License, which permits use, sharing,
val rates for cancer patients, the utilization of ICIs is asso- adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
ciated with various adverse effects, among which thyroid source, provide a link to the Creative Commons licence, and indicate if
disorders are notably prevalent. The pathogenesis of ICI- changes were made. The images or other third party material in this
induced thyroid dysfunction encompasses immune- article are included in the article’s Creative Commons licence, unless
mediated acute inflammation leading to destructive thyr- indicated otherwise in a credit line to the material. If material is not
included in the article’s Creative Commons licence and your intended
oiditis. T cell activation, alongside the involvement of use is not permitted by statutory regulation or exceeds the permitted
various antibodies and cytokines, plays a significant role in use, you will need to obtain permission directly from the copyright
both initiating and progressing the disease [1, 8, 16, 17]. holder. To view a copy of this licence, visit http://creativecommons.
Diagnosing ICI-related thyroid disorders requires vigi- org/licenses/by/4.0/.
lance and regular thyroid function screening, utilizing
measurements of TSH, fT4, and TRAb, TPO-antibodies and
TSH receptor antibodies, according to specific guidelines References
[13, 14, 53–57]. Clinical presentations vary from asymp-
1. E.S. Husebye, F. Castinetti, S. Criseno, G. Curigliano, B. Dec-
tomatic cases to severe manifestations of thyrotoxicosis, allonne, M. Fleseriu, C.E. Higham, I. Lupi, S.A. Paschou, M.
hypothyroidism, and even Graves’ disease [24, 30, 32]. Toth, M. van der Kooij, O.M. Dekkers, Endocrine-related adverse
Early detection demands a thorough examination encom- conditions in patients receiving immune checkpoint inhibition: An
passing both clinical and laboratory evaluation. ESE clinical practice guideline. Eur. J. Endocrinol. 187(6),
G1–G21 (2022). https://doi.org/10.1530/EJE-22-0689
Understanding the pathophysiological mechanisms under- 2. J. Moslehi, A.H. Lichtman, A.H. Sharpe, L. Galluzzi, R.N. Kitsis,
lying these adverse effects is vital for the development of Immune checkpoint inhibitor-associated myocarditis: Manifesta-
effective treatment strategies. The management of these tions and mechanisms. J. Clin. Investig. 131(5), e145186, 145186
thyroid-related side effects necessitates an individualized (2021). https://doi.org/10.1172/JCI145186
3. N. Okura, M. Asano, J. Uchino, Y. Morimoto, M. Iwasaku, Y.
approach that considers the severity of the condition, the Kaneko, T. Yamada, M. Fukui, K. Takayama, Endocrinopathies
patient’s clinical state, and the stage of any malignancy associated with immune checkpoint inhibitor cancer treatment: A
involved. As such, a collaborative effort between various review. J. Clin. Med. 9(7), Article 7 (2020). https://doi.org/10.
medical professionals is essential to ensure optimal care 3390/jcm9072033
4. Z. Cardona, J.A. Sosman, S. Chandra, W Huang, Endocrine side
[13, 14, 53, 54, 56]. Guidelines established by prominent effects of immune checkpoint inhibitors. Front Endocrinol 14 (2023).
organizations such as ASCO, NCCN, SITC, and ESMO serve https://www.frontiersin.org/articles/10.3389/fendo.2023.1157805
as a valuable resource for healthcare providers, highlighting the 5. D.L. Morganstein, Z. Lai, L. Spain, S. Diem, D. Levine, C. Mace,
significance of tailored treatments based on the severity grade. M. Gore, J. Larkin, Thyroid abnormalities following the use of
cytotoxic T-lymphocyte antigen-4 and programmed death receptor
Close monitoring, prompt diagnosis, and personalized protein-1 inhibitors in the treatment of melanoma. Clin. Endo-
treatment strategies are crucial for addressing the com- crinol. 86(4), 614–620 (2017). https://doi.org/10.1111/cen.13297
plexities of ICI-induced thyroid disorders. Collaboration 6. H. Lee, F.S. Hodi, A. Giobbie-Hurder, P.A. Ott, E.I. Buchbinder,
between healthcare professionals and continuous research R. Haq, S. Tolaney, R. Barroso-Sousa, K. Zhang, H. Donahue, M.
Davis, M.E. Gargano, K.M. Kelley, R.S. Carroll, U.B. Kaiser, L.
are essential for the formulation of future guidelines, the Min, Characterization of thyroid disorders in patients receiving
implementation of tailored treatment and the enhancement immune checkpoint inhibition therapy. Cancer Immunol. Res.
of ICI therapy. 5(12), 1133–1140 (2017). https://doi.org/10.1158/2326-6066.
CIR-17-0208
Author contributions S.A.P., D.K., E.-R.K., E.K. conceived the idea 7. F. Guaraldi, R. La Selva, M.T. Samà, V. D’Angelo, D. Gori, P.
of this review article. Material preparation, data collection and analysis Fava, M.T. Fierro, P. Savoia, E. Arvat, Characterization and
were performed by D.K., E.-R.K. and E.K. The first draft of the implications of thyroid dysfunction induced by immune checkpoint
manuscript was written by D.K., E.-R.K. and E.K. and E.G. All inhibitors in real-life clinical practice: A long-term prospective
authors critically revised the manuscript and approved the final study from a referral institution. J. Endocrinol. Investig. 41(5),
manuscript. 549–556 (2018). https://doi.org/10.1007/s40618-017-0772-1
Endocrine

8. A. Kotwal, M.P. Gustafson, S. Bornschlegl, L. Kottschade, D.A. Pembrolizumab-induced thyroiditis: Comprehensive clinical
Delivanis, A.B. Dietz, M. Gandhi, M. Ryder, Immune checkpoint review and insights into underlying involved mechanisms. J.
inhibitor-induced thyroiditis is associated with increased intra- Clin. Endocrinol. Metab. 102(8), 2770–2780 (2017). https://
thyroidal T lymphocyte subpopulations. Thyroid 30(10), doi.org/10.1210/jc.2017-00448
1440–1450 (2020). https://doi.org/10.1089/thy.2020.0075 22. C. Kurimoto, H. Inaba, H. Ariyasu, H. Iwakura, Y. Ueda, S.
9. J. de Filette, Y. Jansen, M. Schreuer, H. Everaert, B. Velkeniers, Uraki, K. Takeshima, Y. Furukawa, S. Morita, Y. Yamamoto, S.
B. Neyns, B. Bravenboer, Incidence of thyroid-related adverse Yamashita, M. Katsuda, A. Hayata, H. Akamatsu, M. Jinnin, I.
events in melanoma patients treated with pembrolizumab. J. Clin. Hara, H. Yamaue, T. Akamizu, Predictive and sensitive bio-
Endocrinol. Metab. 101(11), 4431–4439 (2016). https://doi.org/ markers for thyroid dysfunctions during treatment with immune-
10.1210/jc.2016-2300 checkpoint inhibitors. Cancer Sci. 111(5), 1468–1477 (2020).
10. U. Azmat, D. Liebner, A. Joehlin-Price, A. Agrawal, F. Nabhan, https://doi.org/10.1111/cas.14363
Treatment of ipilimumab induced graves’ disease in a patient with 23. L. Zhan, H. Feng, H. Liu, L. Guo, C. Chen, X. Yao, S Sun,
metastatic melanoma. Case Rep. Endocrinol. 2016, e2087525 Immune checkpoint inhibitors-related thyroid dysfunction: Epi-
(2016). https://doi.org/10.1155/2016/2087525 demiology, clinical presentation, possible pathogenesis, and
11. E.H. Gan, A.L. Mitchell, R. Plummer, S. Pearce, P. Perros, management. Front. Endocrinol. 12 (2021). https://www.
Tremelimumab-induced graves hyperthyroidism. Eur. Thyroid J. frontiersin.org/articles/10.3389/fendo.2021.649863
6(3), 167–170 (2017). https://doi.org/10.1159/000464285 24. A. Brancatella, N. Viola, S. Brogioni, L. Montanelli, C. Sardella,
12. M.V. Deligiorgi, S. Sagredou, L. Vakkas, D.T. Trafalis, The P. Vitti, C. Marcocci, I. Lupi, F. Latrofa, Graves’ disease induced
continuum of thyroid disorders related to immune checkpoint by immune checkpoint inhibitors: A case report and review of the
inhibitors: STill Many Pending Queries. Cancers 13(21), Article literature. Eur. Thyroid J. 8(4), 192–195 (2019). https://doi.org/10.
21 (2021). https://doi.org/10.3390/cancers13215277 1159/000501824
13. S.A. Paschou, K. Stefanaki, T. Psaltopoulou, M. Liontos, K. 25. A. Olsson-Brown, R. Lord, J. Sacco, J. Wagg, M. Coles, M.
Koutsoukos, F. Zagouri, I. Lambrinoudaki, M.-A. Dimopoulos, Pirmohamed, Two distinct clinical patterns of checkpoint
How we treat endocrine complications of immune checkpoint inhibitor-induced thyroid dysfunction. Endocr. Connect. 9(4),
inhibitors. ESMO Open 6(1), 100011 (2021). https://doi.org/10. 318–325 (2020). https://doi.org/10.1530/EC-19-0473
1016/j.esmoop.2020.100011 26. D. Lu, J. Yao, G. Yuan, Y. Gao, J. Zhang, X. Guo, Immune
14. J.A. Thompson, B.J. Schneider, J. Brahmer, S. Andrews, P. checkpoint inhibitor-related new-onset thyroid dysfunction: A
Armand, S. Bhatia, L.E. Budde, L. Costa, M. Davies, D. Dun- retrospective analysis using the US FDA adverse event reporting
nington, M.S. Ernstoff, M. Frigault, B. Hoffner, C.J. Hoimes, M. system. Oncologist 27(2), e126–e132 (2022). https://doi.org/10.
Lacouture, F. Locke, M. Lunning, N.A. Mohindra, J. Naidoo, J.L. 1093/oncolo/oyab043
Scavone, Management of immunotherapy-related toxicities, Ver- 27. M. Stelmachowska-Banaś, I. Czajka-Oraniec, Management of
sion 1.2019, NCCN clinical practice guidelines in oncology. J. endocrine immune-related adverse events of immune checkpoint
Natl. Compr. Cancer Netw. 17(3), 255–289 (2019). https://doi. inhibitors: An updated review. Endocr. Connect. 9(10),
org/10.6004/jnccn.2019.0013 R207–R228 (2020). https://doi.org/10.1530/EC-20-0342
15. K.C.J. Yuen, S.L. Samson, I. Bancos, A.R. Gosmanov, S. Jasim, 28. I. Goyal, M.R. Pandey, R. Sharma, A. Chaudhuri, P. Dandona,
L.A. Fecher, J.S. Weber, American association of clinical endo- The side effects of immune checkpoint inhibitor therapy on the
crinology disease state clinical review: Evaluation and manage- endocrine system. Indian J. Med. Res. 154(4), 559 (2021). https://
ment of immune checkpoint inhibitor-mediated endocrinopathies: doi.org/10.4103/ijmr.IJMR_313_19
A practical case-based clinical approach. Endocr. Pract. 28(7), 29. J. Del Rivero, L.M. Cordes, J. Klubo‐Gwiezdzinska, R.A. Madan,
719–731 (2022). https://doi.org/10.1016/j.eprac.2022.04.010 L.K. Nieman, J.L. Gulley, Endocrine‐related adverse events
16. K. Torimoto, Y. Okada, S. Nakayamada, S. Kubo, Y. Tanaka, related to immune checkpoint inhibitors: Proposed algorithms for
Anti-PD-1 antibody therapy induces hashimoto’s disease with an management. Oncologist 25(4), 290–300 (2020). https://doi.org/
increase in peripheral blood follicular helper T cells. Thyroid 10.1634/theoncologist.2018-0470
27(10), 1335–1336 (2017). https://doi.org/10.1089/thy.2017.0062 30. L.-S. Chang, R. Barroso-Sousa, S.M. Tolaney, F.S. Hodi, U.B.
17. T. Maekura, M. Naito, M. Tahara, N. Ikegami, Y. Kimura, S. Kaiser, L. Min, Endocrine toxicity of cancer immunotherapy
Sonobe, T. Kobayashi, T. Tsuji, S. Minomo, A. Tamiya, S. Atagi, targeting immune checkpoints. Endocr. Rev. 40(1), 17–65 (2019).
Predictive factors of nivolumab-induced hypothyroidism in https://doi.org/10.1210/er.2018-00006
patients with non-small cell lung cancer. Vivo 31(5), 1035–1039 31. J. Villadolid, A. Amin, Immune checkpoint inhibitors in clinical
(2017). https://doi.org/10.21873/invivo.11166 practice: Update on management of immune-related toxicities.
18. J.C. Osorio, A. Ni, J.E. Chaft, R. Pollina, M.K. Kasler, D. Ste- Transl. Lung Cancer Res. 4(5) (2015). https://doi.org/10.3978/j.
phens, C. Rodriguez, L. Cambridge, H. Rizvi, J.D. Wolchok, T. issn.2218-6751.2015.06.06
Merghoub, C.M. Rudin, S. Fish, M.D. Hellmann, Antibody- 32. L. Duan, L. Wang, H. Wang, X. Si, L. Zhang, X. Liu, Y. Li, X.
mediated thyroid dysfunction during T-cell checkpoint blockade Guo, J. Zhou, H. Zhu, L. Zhang, Clinical diagnosis and treatment
in patients with non-small-cell lung cancer. Ann. Oncol. 28(3), of immune checkpoint inhibitors-related endocrine dysfunction.
583–589 (2017). https://doi.org/10.1093/annonc/mdw640 Thorac. Cancer 11(4), 1099–1104 (2020). https://doi.org/10.1111/
19. C.A. Muir, A.M. Menzies, R. Clifton-Bligh, V.H.M. Tsang, 1759-7714.13347
Thyroid toxicity following immune checkpoint inhibitor treatment 33. S. A. Paschou, M. Liontos, E. Eleftherakis-Papaiakovou, K. Ste-
in advanced cancer. Thyroid 30(10), 1458–1469 (2020). https:// fanaki, C. Markellos, K. Koutsoukos, F. Zagouri, T. Psaltopoulou,
doi.org/10.1089/thy.2020.0032 M.-A Dimopoulos, Oncological patients with endocrine compli-
20. X. Zhou, S. Iwama, T. Kobayashi, M. Ando, H. Arima, Risk of cations after immunotherapy with checkpoint inhibitors present
thyroid dysfunction in PD-1 blockade is stratified by the pattern of longer progression-free and overall survival. Front. Oncol. 12
TgAb and TPOAb positivity at baseline. J. Clin. Endocrinol. (2022). https://www.frontiersin.org/articles/10.3389/fonc.2022.
Metab. 108(10), e1056–e1062 (2023). https://doi.org/10.1210/ 847917
clinem/dgad231 34. B. Anderson, D.L. Morganstein, Endocrine toxicity of cancer
21. D.A. Delivanis, M.P. Gustafson, S. Bornschlegl, M.M. Merten, immunotherapy: Clinical challenges. Endocr. Connect. 10(3),
L. Kottschade, S. Withers, A.B. Dietz, M. Ryder, R116–R124 (2021). https://doi.org/10.1530/EC-20-0489
Endocrine

35. J.J. Wright, A.C. Powers, D.B. Johnson, Endocrine toxicities of 48. M.S. von Itzstein, A.S. Gonugunta, Y. Wang, T. Sheffield, R. Lu, S.
immune checkpoint inhibitors. Nat. Rev. Endocrinol. 17(7), Ali, F.J. Fattah, D. Xie, J. Cai, Y. Xie, D.E. Gerber, Correction to:
Article 7 (2021). https://doi.org/10.1038/s41574-021-00484-3 Divergent prognostic effects of pre-existing and treatment-emergent
36. E. Kassi, A. Angelousi, N. Asonitis, P. Diamantopoulos, A. thyroid dysfunction in patients treated with immune checkpoint
Anastasopoulou, G. Papaxoinis, M. Kokkinos, I. Giovanopoulos, inhibitors. Cancer Immunol., Immunotherapy: CII 71(9), 2183–2184
G. Kyriakakis, F. Petychaki, A. Savelli, O. Benopoulou, H. (2022). https://doi.org/10.1007/s00262-022-03218-0
Gogas, Endocrine-related adverse events associated with immune- 49. S. Coniac, M. Stoian, Updates in endocrine immune-related
checkpoint inhibitors in patients with melanoma. Cancer Med. adverse events in oncology immunotherapy. Acta Endocrinolo-
8(15), 6585–6594 (2019). https://doi.org/10.1002/cam4.2533 gica (Buchar., Rom.: 2005) 17(2), 286–289 (2021). https://doi.
37. A. Chera, A.L. Stancu, O. Bucur, Thyroid-related adverse events org/10.4183/aeb.2021.286
induced by immune checkpoint inhibitors. Front. Endocrinol. 13, 50. D.J. Byun, J.D. Wolchok, L.M. Rosenberg, M. Girotra, Cancer
1010279 (2022). https://doi.org/10.3389/fendo.2022.1010279 immunotherapy—Immune checkpoint blockade and associated
38. S. Yang, K.-H. Yu, N. Palmer, K. Fox, S.C. Kou, I.S. Kohane, endocrinopathies. Nat. Rev. Endocrinol. 13(4), Article 4 (2017).
Autoimmune effects of lung cancer immunotherapy revealed by https://doi.org/10.1038/nrendo.2016.205
data-driven analysis on a nationwide cohort. Clin. Pharmacol. 51. O.-P.R. Hamnvik, P.R. Larsen, E. Marqusee, Thyroid dysfunction
Ther. 107(2), 388–396 (2020). https://doi.org/10.1002/cpt.1597 from antineoplastic agents. J. Natl. Cancer Inst. 103(21),
39. M.R. Migden, D. Rischin, C.D. Schmults, A. Guminski, A. Haus- 1572–1587 (2011). https://doi.org/10.1093/jnci/djr373
child, K.D. Lewis, C.H. Chung, L. Hernandez-Aya, A.M. Lim, A.L.S. 52. M. Girotra, A. Hansen, A. Farooki, D.J. Byun, L. Min, B.C. Creelan,
Chang, G. Rabinowits, A.A. Thai, L.A. Dunn, B.G.M. Hughes, N.I. M.K. Callahan, M.B. Atkins, E. Sharon, S.J. Antonia, P. West, A.E.
Khushalani, B. Modi, D. Schadendorf, B. Gao, F. Seebach, M.G. Gravell; Investigational Drug Steering Committee (IDSC) Immu-
Fury, PD-1 blockade with cemiplimab in advanced cutaneous notherapy Task Force collaboration, The current understanding of the
squamous-cell carcinoma. N. Engl. J. Med. 379(4), 341–351 (2018). endocrine effects from immune checkpoint inhibitors and recom-
https://doi.org/10.1056/NEJMoa1805131 mendations for management. JNCI Cancer Spectr. 2(3), pky021
40. L. Trudu, G. Guaitoli, F. Bertolini, M. Maur, C. Santini, V. R. (2018). https://doi.org/10.1093/jncics/pky021
Papapietro, S. Talerico, S. Natalizio, C. Isca, M. Dominici, F. 53. J.R. Brahmer, C. Lacchetti, B.J. Schneider, M.B. Atkins, K.J.
Barbieri, Thyroid function impairment after chemo- Brassil, J.M. Caterino, I. Chau, M.S. Ernstoff, J.M. Gardner, P.
immunotherapy for advanced NSCLC: A single institutional ret- Ginex, S. Hallmeyer, J. Holter Chakrabarty, N.B. Leighl, J.S.
rospective report. Immunotherapy (2022). https://doi.org/10.2217/ Mammen, D.F. McDermott, A. Naing, L.J. Nastoupil, T. Phillips,
imt-2021-0165 L.D. Porter; National Comprehensive Cancer Network, Manage-
41. L.L. Prather, A. Ali, H. Wang, W. Du, N. Chen, A. Sheth, V. San- ment of immune-related adverse events in patients treated with
dulache, A.L. Sabichi, J.O. Kemnade, D.Y. Wang, Immune-related immune checkpoint inhibitor therapy: American society of clinical
adverse events and immunotherapy efficacy in patients with cancer: A oncology clinical practice guideline. J. Clin. Oncol 36(17),
retrospective study. J. Clin. Oncol. 40(16_suppl), 2654–2654 (2022). 1714–1768 (2018). https://doi.org/10.1200/JCO.2017.77.6385
https://doi.org/10.1200/JCO.2022.40.16_suppl.2654 54. I. Puzanov, A. Diab, K. Abdallah, C.O. Bingham, C. Brogdon, R.
42. X. Zheng, H. Xiao, J. Long, Q. Wei, L. Liu, L. Zan, W. Ren, Dadu, L. Hamad, S. Kim, M.E. Lacouture, N.R. LeBoeuf, D.
Dynamic follow-up of the effects of programmed death 1 inhibitor Lenihan, C. Onofrei, V. Shannon, R. Sharma, A.W. Silk, D.
treatment on thyroid function and sonographic features in patients Skondra, M.E. Suarez-Almazor, Y. Wang, K. Wiley, P. Zheng,
with hepatocellular carcinoma. Endocr. Connect. 11(5), e220065 Managing toxicities associated with immune checkpoint inhibi-
(2022). https://doi.org/10.1530/EC-22-0065 tors: Consensus recommendations from the Society for Immu-
43. H.-S. Baek, C. Jeong, K. Shin, J. Lee, H. Suh, D.-J. Lim, M.I. notherapy of Cancer (SITC) Toxicity Management Working
Kang, J. Ha, Association between the type of thyroid dysfunction Group. J. Immunother. Cancer 5(1), 95 (2017). https://doi.org/10.
induced by immune checkpoint inhibitors and prognosis in cancer 1186/s40425-017-0300-z
patients. BMC Endocr. Disord. 22(1), 89 (2022). https://doi.org/ 55. H. Arima, S. Iwama, H. Inaba, H. Ariyasu, N. Makita, M. Otsuki,
10.1186/s12902-022-01004-8 K. Kageyama, A. Imagawa, T. Akamizu, Management of
44. A. Kotwal, L. Kottschade, M. Ryder, PD-L1 inhibitor-induced immune-related adverse events in endocrine organs induced by
thyroiditis is associated with better overall survival in cancer immune checkpoint inhibitors: Clinical guidelines of the Japan
patients. Thyroid 30(2), 177–184 (2020). https://doi.org/10.1089/ Endocrine Society. Endocr. J. 66(7), 581–586 (2019). https://doi.
thy.2019.0250 org/10.1507/endocrj.EJ19-0163
45. J. Lima Ferreira, C. Costa, B. Marques, S. Castro, M. Victor, J. 56. F. Castinetti, F. Albarel, F. Archambeaud, J. Bertherat, B.
Oliveira, A.P. Santos, I.L. Sampaio, H. Duarte, A.P. Marques, I. Bouillet, P. Buffier, C. Briet, B. Cariou, P. Caron, O. Chabre, P.
Torres, Improved survival in patients with thyroid function test Chanson, C. Cortet, C.D. Cao, D. Drui, M. Haissaguerre, S.
abnormalities secondary to immune-checkpoint inhibitors. Cancer Hescot, F. Illouz, E. Kuhn, N. Lahlou, F. Borson-Chazot, French
Immunol. Immunotherapy 70(2), 299–309 (2021). https://doi.org/ Endocrine Society Guidance on endocrine side effects of immu-
10.1007/s00262-020-02664-y notherapy. Endocr.-Relat. Cancer 26(2), G1–G18 (2019). https://
46. X. Li, X. Wang, S. Wang, Y. Liu, R. Wang, Y. Liu, L. Huang, Y. doi.org/10.1530/ERC-18-0320
Feng, X. Xie, L. Shi, Thyroid dysfunction induced by immune 57. C.E. Higham, A. Olsson-Brown, P. Carroll, T. Cooksley, J. Lar-
checkpoint inhibitors and tumor progression during neoadjuvant kin, P. Lorigan, D. Morganstein, P.J. Trainer, Society for endo-
therapy of non‑small cell lung cancer: A case report and literature crinology endocrine emergency guidance: Acute management of
review. Oncol. Lett. 26(5), 496 (2023). https://doi.org/10.3892/ol. the endocrine complications of checkpoint inhibitor therapy.
2023.14083 Endocr. Connect. 7(7), G1–G7 (2018). https://doi.org/10.1530/
47. P. Petranović Ovčariček, F.A. Verburg, M. Hoffmann, I. Iakovou, EC-18-0068
J. Mihailovic, A. Vrachimis, M. Luster, L. Giovanella, Correction 58. C.W. Yu, M. Yau, N. Mezey, I. Joarder, J.A. Micieli, Neuro-
to: Higher thyroid hormone levels and cancer. Eur. J. Nucl. Med. ophthalmic complications of immune checkpoint inhibitors: A
Mol. Imaging 48(3), 951–953 (2021). https://doi.org/10.1007/ systematic review. Eye Brain 12, 139–167 (2020). https://doi.org/
s00259-020-05052-x 10.2147/EB.S277760

You might also like