Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 11/2023

Open Access 03.07.2023 | Guidelines

The EANM guideline on radioiodine therapy of benign thyroid disease

verfasst von: Alfredo Campennì, Anca M. Avram, Frederik A. Verburg, Ioannis Iakovou, Heribert Hänscheid, Bart de Keizer, Petra Petranović Ovčariček, Luca Giovanella

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 11/2023

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

This document provides the new EANM guideline on radioiodine therapy of benign thyroid disease. Its aim is to guide nuclear medicine physicians, endocrinologists, and practitioners in the selection of patients for radioiodine therapy. Its recommendations on patients’ preparation, empiric and dosimetric therapeutic approaches, applied radioiodine activity, radiation protection requirements, and patients follow-up after administration of radioiodine therapy are extensively discussed.
Hinweise

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Liability statement
This guideline summarizes the views of the EANM [Thyroid Committee]. It reflects recommendations for which the EANM cannot be held responsible. The recommendations should be taken into context of good practice of nuclear medicine and do not substitute for national and international legal or regulatory provisions.

Preamble

The European Association of Nuclear Medicine (EANM) is a professional non-profit medical association that facilitates communication worldwide among individuals pursuing clinical and research excellence in nuclear medicine. The EANM was founded in 1985.
These guidelines are intended to assist practitioners in providing appropriate nuclear medicine care for patients. They are not inflexible rules or requirements of practice and are not intended, nor should they be used, to establish a legal standard of care.
The ultimate judgment regarding the propriety of any specific procedure or course of action must be made by medical professionals taking into account the unique circumstances of each case. Thus, there is no implication that an approach differing from the guidelines, standing alone, is below the standard of care. To the contrary, a conscientious practitioner may responsibly adopt a course of action different from that set out in the guidelines when, in the reasonable judgment of the practitioner, such course of action is indicated by the condition of the patient, limitations of available resources, or advances in knowledge or technology subsequent to publication of the guidelines.
The practice of medicine involves not only the science but also the art of dealing with the prevention, diagnosis, alleviation, and treatment of disease. The variety and complexity of human conditions make it impossible to always reach the most appropriate diagnosis or to predict with certainty a particular response to treatment. Therefore, it should be recognized that adherence to these guidelines will not ensure an accurate diagnosis or a successful outcome.
All that should be expected is that the practitioner will follow a reasonable course of action based on current knowledge, available resources, and the needs of the patient to deliver effective and safe medical care. The sole purpose of these guidelines is to assist practitioners in achieving this objective.

Autoimmune hyperthyroidism (Graves’ disease) and non-autoimmune toxic nodular goiter/toxic multinodular goiter

Introduction

Radioactive iodine-131 (RAI) therapy has been used for the treatment of hyperthyroidism for more than 80 years since Doctor Saul Hertz successfully administered the first therapeutic activity of radioiodine-131 (I-131) to a patient with Graves’ disease (GD) in early 1941.
Since then, millions of patients worldwide have received RAI therapy for definitive treatment of hyperthyroidism due to either autoimmune (i.e., GD) or non-autoimmune toxic (multi)-nodular goiter (i.e., TMNG/TNG, respectively) [1]. Graves’ disease (GD) [Morbus Basedow in German-speaking countries] and autonomously functioning thyroid nodules (i.e., toxic nodular goiter, TNG; toxic multinodular goiter, TMNG) are the most common causes of hyperthyroidism (i.e., GD 70–80% of cases, TNG/TMNG 20–30% of cases).
RAI therapy has also been employed as an alternative to surgery for the treatment of patients affected by large non-toxic nodular goiter (NTG) to achieve a significant reduction of gland volume [25].
The use of RAI therapy has drastically diminished the number of patients requiring surgery for definitive treatment of hyperthyroidism, thus reducing both the risk of surgical complications (e.g., recurrent and superior laryngeal nerve palsy, hypoparathyroidism, hemorrhage) and healthcare costs [1]. RAI is the classic agent used for the diagnosis and treatment of benign thyroid disease based on sodium-iodine symporter (NIS) expression in normal thyroid tissue and its widespread medical use represents the first application of the theragnostic concept in medicine, remaining to this day the cornerstone of radionuclide therapy.
The accumulated clinical experience over the past eight decades has proven that RAI therapy is safe and highly effective, with current literature offering several schools of thought and differing insights on how to approach this treatment. Since the publication of the prior edition of this guideline [6], these insights have been significantly enriched. For instance, a recent meta-analysis of various dosimetry data from studies on RAI therapy for GD showed a dose–response relationship between the delivered radiation absorbed dose to the thyroid and the therapeutic outcome (i.e. rates of hyper-, eu-, and hypothyroidism after RAI treatment) [7]. Therefore, the current document presents an updated guideline for the use of RAI in the treatment of benign thyroid disease with the aim to guide/support nuclear medicine physicians, endocrinologists, and practitioners in selecting patients for RAI therapy. In addition, recommendations on patients’ preparation, strategy approaches (i.e., empiric vs. dosimetric therapy), radioiodine activity regimes, radiation protection requirements, and follow-up management are duly discussed.

Radionuclides of interest for thyroid imaging and therapy

Functional diagnostic imaging of patients with benign thyroid disease involves the use of 99mTc-pertechnetate (Na[99mTc]Tc04) and radioiodine-123 (Na[123I]I) due to their specificity for the sodium iodide symporter (NIS). Details on uptake mechanisms, pharmacokinetics, biodistribution, clinical indications of different tracers, and imaging procedures are out of the scope of the present guideline; readers are referred to the EANM practice guideline/SNMMI procedure standard for RAIU and thyroid scintigraphy [8].
The therapeutic radioisotope Na[131I]I has a half-life of 8.02 days and decays by β-electrons emission with a maximum energy of the main transition of 0.61 MeV, and an average energy release per decay of 0.192 MeV. The average range in the tissue of the β-particle is 0.4 mm. In addition, Na[131I]I emits an electromagnetic photon with a principal gamma-ray of 364 keV. Based on these physical characteristics, Na[131I]I was the first theragnostic radiopharmaceutical: its gamma-photon emission was used to observe and quantify iodine distribution and kinetics within the thyroid gland, while its β-particle emission (β-radiation) was used to obtain the therapeutic effect.

General considerations for RAI therapy

The rationale underlying RAI therapy is based on the ability of follicular thyroid cells to take up Na[131I]I, in the same fashion as stable iodine (127I) absorbed from dietary sources.
The therapeutic use of Na[131I]I has two main applications:
(i)
To treat hyperthyroidism in patients with GD or TNG/TMNG;
 
(ii)
To reduce the volume of the whole gland (e.g., GD or non-toxic goiter (NTG)) or hyperfunctioning nodule(s).
 
Patients affected by the above conditions referred for RAI therapy should be evaluated and eventually treated with I-131 according to international guidelines [6, 9] and local medical standards. Patients with absolute contraindications to RAI therapy must be promptly identified and excluded from this therapeutic procedure. The patients who qualify for RAI therapy require consultation with a nuclear medicine specialist to discuss specific information and preparation for this procedure based on the patient’s specific diagnosis, age, and comorbidities. Informed consent should be obtained before RAI therapy administration following national procedural recommendations. Detailed written instructions to reduce radiation exposure and the risk of contamination of family members and the general public must be given to patients before release after therapy.

RAI therapy for benign thyroid disease methods: patient preparation, dosimetry, and radiation protection requirements

Patient candidates for RAI therapy must be evaluated by a nuclear medicine specialist for discussing the indications, logistics, and expectations of RAI treatment. This consultation will also include a review of current medications and specific dietary preparation instructions in anticipation for RAI administration, as well as checking for current and/or prior use of iodine-containing products (Table 1] with resultant decreased radioiodine uptake (RAIU) in the thyroid gland which can reduce RAI therapy efficacy [1, 6, 1012]. Notably, RAIU testing and RAI therapy should be performed under the same condition (i.e., preparation protocol, medications).
Table 1
Thyroid drugs and iodide-containing substances that can reduce radioiodine thyroid uptake
Type of medication
Recommended time of withdrawal
Water-soluble intravenous radiographic contrast agents
6–8 wk*, assuming normal renal function
Lipophilic intravenous radiographic contrast agents
3–6 mo#
Thyroxine
3–4 wk*
Triiodothyronine
10–14 d§
Antithyroid drugs:
  Methimazole
  Propylthiouracil
2–5 d§ before RAI therapy
2–8 wk* if RAI therapy is performed by fixed-activity method
(4–7 d§ if RAI therapy is performed after personalized dosimetric approach)
Nutrition supplements containing iodide
7–10 d§
Kelp, agar, carrageenan, Lugol solution
2–3 wk*, depending on iodide content
Saturated solution of potassium iodide
2–3 wk*
Topical iodine (e.g., surgical skin preparation)
2–3 wk*
Amiodarone
3–6 mo# or longer
Adapted from SNMMI Procedure Standard 2012
§d days; *wk weeks; #mo months
Pregnancy is an absolute contraindication for RAI treatment. In all fertile females, it can be excluded by a careful history collection and demonstrated by obtaining hCG serum testing just before RAI administration (if not feasible within 72 h maximum). If there is a suspicion of early pregnancy (not detectable by beta-HCG) according to anamnestic data, RAI therapy should be postponed. Patients’ preparation protocols may differ depending on different thyroid diseases and treatment goals. Details are reported in the specific sections.

Radiation protection requirements

Any medical examination and treatment involving radiation exposure to patients, treating medical personnel, or members of the public should be carried out in accordance with the safety standards on radiation protection laid down in Chapter VII of the Council Directive 2013/59/Euratom [13].
With regard to the protection of patients treated with radiation, Article 56 of the directive is relevant: “For all medical exposure of patients for radiotherapeutic purposes, exposures of target volumes shall be individually planned and their delivery appropriately verified taking into account that doses to non-target volumes and tissues shall be as low as reasonably achievable and consistent with the intended radiotherapeutic purpose of the exposure.” [13].
Although the member states of the European Union were obliged to transpose the directive into national law, the requirement for individually planned treatments has not generally been adopted for nuclear medicine therapies. Irrespective of the present recommendations for action, medical practitioners must comply with the national formulations of the applicable laws, particularly when measurements of patient-specific biokinetics and retention time functions and verification of the administered radiation absorbed doses can be performed.
Depending on national regulations for treatments with unsealed radioactive agents, it may be necessary to have the treatment performed by specially trained personnel in a dedicated therapy ward with appropriately shielded rooms and suitable radiation protection equipment. Usually, such a ward has forced ventilation with adequate air exchange and is connected to a sewage system for the collection of contaminated wastewater. Treatment should be provided by a team of physicians, nurses, technologists, and medical physics experts with a high level of competence and a clear definition of tasks and responsibilities, considering legal requirements. Written instructions should be available for the handling of radioactive materials, waste, and treated patients. Adequate monitoring of personnel for exposure to direct radiation and/or contamination is required in line with national and regulatory requirements.

Dosimetry for therapy planning and dose verification

Dosimetry in conjunction with RAI therapy for benign thyroid disease can be performed either pre-therapeutically for determining the necessary Na[131I]I administered activity which will deliver the radiation absorbed dose to the target tissue that is expected to be effective in the particular type of disease, or post-therapeutically for evaluating the energy dose absorbed in tissue. The energy dose D from self-irradiation of a radioiodine accumulating target mass M, which can be a substructure of the thyroid gland such as a hyper-functioning adenoma or the whole thyroid gland such as in Graves’ disease or goiter, is given by the product of the mean energy Ē deposited in M per decay of 131I and the number of decays represented by the time integral of the activity A(t) in M. A(t) is often written as product Aa· RIU(t), where Aa is the administered activity and RIU(t) the radioiodine uptake, i.e., the fraction of the administered activity in M at a given time t after the administration. The activity necessary to achieve a specified radiation absorbed dose D in the target mass M is [14]:
$${A}_{a}\left[MBq\right]=\frac{1}{\overline{E} }\cdot \frac{M\left[g\right]\cdot D\left[Gy\right]}{\underset{0}{\overset{\infty }{\int }}RIU\left(t\right)dt\left[d\right]}$$
(1)
The constant value recommended in [14] for the mean energy deposited in the target tissue per decay of 131I,
$$\overline{E }=2.808\frac{Gy\cdot g}{MBq\cdot d}$$
(2)
which was calculated for thyroid with M = 20 g, will produce results with adequate accuracy for most of the patients. Mean energy deposition is about 5% higher due to increased photon absorption in goiters with M = 90 g.
Complete dosimetry requires measurements of the mass of the target tissue to be treated and the time function RIU(t) of iodine uptake in this mass. The activity required to achieve a specified absorbed dose is linearly dependent on the target mass. Any error in the mass estimate induces a correspondent error in the activity or absorbed dose calculations. For reliable dosimetry, morphological imaging should be used to measure the target mass. Two-dimensional ultrasound is inferior to three-dimensional ultrasound [1517]; however, it is easy to perform, cost-effective, and reliable in most patients [1720], and it is recommended in routine diagnostics. Computed tomography (CT) [21], magnetic resonance imaging (MRI) [20], and positron emission tomography (PET) [22] have been described as suitable, but as they are overly expensive and demanding, might however be considered in rare cases of large goiters with substernal extension. Planar and tomographic scintigraphies have limited spatial resolution and are sufficiently accurate only for target masses greater than approximately 20 g [20].
In particular, planar scintigraphy for mass estimation should be reserved for patients in whom the target tissue cannot be delineated or completely visualized in ultrasound, which is considerably more accurate for volume measurement. The measurement must then be performed after administration of Na[99mTc]TcO4 using a camera system with high spatial resolution and, after suitable contour finding, the volume must be estimated from the projection area of the accumulating gland [20]. Before planar scintigraphy is applied for volume determination, phantom studies and comparisons on patients with both scintigram and ultrasound measurement must be performed to verify that the contour finding and the equation for calculating volume from area provide sufficiently correct results for the gamma camera and acquisition parameters actually used.
From planar images with high spatial resolution, usually obtained by ultrasound, the mass M of a substructure such as a thyroid lobe or a nodule can be estimated from the lengths of the longest diameter A and 2 orthogonal axes B and C in the transverse section with the largest area as M = A,·B,·C/2, with A, B, and C, in centimeters and M in grams [14]. For large structures approximately shaped like an ellipsoid, the error of the mass estimate is typically less than 20%. Higher errors are possible for very small nodules, if the target mass is not clearly circumscribed in imaging, in patients with toxic goiters with several nodules or disseminated disease, or if the morphological imaging does not match the scintigraphically documented activity distribution. Morphological images used to deduce a mass estimate should always be compared to a Na[99mTc]TcO4 or Na[123I]I scintigram in order to verify that the measured volume corresponds to the accumulating target tissue.
The time integral of the uptake function determines the number of decays and thus the absorbed dose in the target mass per unit of activity administered. At least 3 uptake measurements are required to calculate the uptake function. Recommended time points for measurements are 4–6 h after the administration during the initial increase of uptake, after 1–2 days shortly after the time of maximum uptake, and after 5–8 days when the decrease of uptake allows for a reliable estimate of the effective half-life of the activity in the target tissue [14]. Making use of theoretical compartment model considerations [23, 24], the number of uptake assessments can be reduced to two measurements obtained after 1–2 days and after 5–8 days, or even to only one measurement obtained after 5–8 days with minor loss of accuracy [14, 2326]. To ensure reliable uptake measurements and consistent dose calculations, it is recommended to follow the standard operational procedures on pre-therapeutic dosimetry provided in [14].

Simplifying activity regimes

If instead of a late measurement of RIU(t), only the maximum uptake is estimated from an early assessment after time te, the measured value is not representative of the time integral in the denominator of Eq. (1]. The activity to be administered in such an approach should be calculated by [14]:
$${A}_{a}\left[MBq\right]=\frac{0.714}{\overline{E} }\cdot \frac{M\left[g\right]\cdot D\left[Gy\right]}{RIU\left({t}_{e}\right)\cdot {2}^{{t}_{e}/{T}_{est}}\cdot {T}_{est}\left[d\right]}$$
(3)
with a reasonable estimate Test of the effective half-life of the activity in the gland. As recommended in [14], a fixed value Test = 5.5 days might be used for all patients or disease-specific half-lives if they are known to be representative of the respective patient population. With Test = 5.5 days, the absorbed dose is a factor of 1.24 higher than expected if the actual effective half-life is 7 days and factors of 1.31 and 1.65 lower for 4 and 3 days actual effective half-life, respectively. Accuracy increases with the increasing time of the measurement after the administration of Na[131I]I. The potential for error is lower with a measurement after 2 days than with a measurement after one day or earlier.
Activity dosage linear to the mass M (g) without any pretherapeutic assessment of iodine kinetics in the target tissue assumes that the time integral over RIU(t) matches a predefined value. Possible overdosing is limited by the theoretically possible maximum value of the integral, which results from almost complete retention in M and decay with the physical half-life of 131I. Significant underdosing and thus ineffective treatment is possible in the case of unexpectedly low uptake and/or half-life.
The highest deviations from an adequate target dose D are possible when fixed activities are administered without considering any influence of thyroid tissue mass, radiation absorbed dose, and iodine kinetics on treatment outcome. Increased rates of residual or recurrent disease are expected with large masses and rapid iodine kinetics. Substantial overdosage is possible with small masses, e.g., a fixed activity of 555 MBq is more than twice as high as typically required to effectively treat autoimmune hyperthyroidism in a patient with 20-g thyroid gland and will result in a high radiation exposure that is medically unjustified and not “as low as reasonably achievable (ALARA)” [13]. Administration of fixed activities without any adjustment for the mass to be treated and the expected kinetics is not recommended.

Definition of Graves’ disease, toxic nodular goiter, toxic multinodular goiter

Hyperthyroidism can present as either persistent subclinical or overt thyrotoxicosis due to excessive synthesis and blood secretion of thyroid hormone(s) by either the whole thyroid gland or hyperfunctioning nodule(s) [1, 9].
Graves’ disease is autoimmune hyperthyroidism due to the presence of stimulating autoantibodies against the TSH receptor (TRAb) that promote thyroid growth and hyperfunction, frequently presenting as overt hyperthyroidism in adults living in iodine sufficient areas [1, 27, 28].
Conversely, the presence of hyperfunctioning thyroid nodule(s) (i.e., TNG or TMNG) is a frequent cause of hyperthyroidism in adults from non-iodine sufficient areas. Somatic gene mutations (i.e., TSH-R gene) have been described in TNG or TMNG, and the most common clinical presentation is subclinical hyperthyroidism [1]. Nevertheless, cardiovascular morbidity and mortality are increased in patients with TNG/TMNG despite the subclinical (or subtle) form of hyperthyroidism frequently encountered in such cases.

Diagnostic approaches and alternative treatments

Nuclear medicine physicians are overall responsible for the delivery of RAI therapy to patients affected by hyperthyroidism and non-toxic goiter.
A detailed personal and family history and an accurate clinical examination are mandatory [1]. GD patients are frequently young or young adults with a family history or past medical history of other autoimmune diseases (e.g., rheumatoid arthritis, vitiligo, coeliac disease) [1, 2931].
Non-autoimmune hyperthyroidism is typically diagnosed in older patients [9, 27, 32, 33]. In particular, TMNG patients frequently have a history of nontoxic goiter for many years or decades preceding hyperthyroidism onset. As a consequence of low iodine intake in regions with iodine insufficiency, chronic TSH stimulation can produce a progressive increase in size and number of nodules which may develop autonomous growth and function over time.
Accordingly, hyperthyroidism progresses gradually from subclinical to overt hyperthyroidism, frequently triggered by an iodine overload event (e.g., amiodarone treatment, iodinated contrast administration) [27, 32, 34].
Information about the current or prior use of substances able to reduce RAI uptake into the gland should be carefully obtained (Table 1]. Young patients with GD mainly exhibit symptoms of sympathetic activation (e.g., anxiety, hyperactivity, heat intolerance, and tremor), while older patients present more frequently with unexplained weight loss and cardiovascular symptoms/signs [27, 35]. Additionally, GD may be characterized by the involvement of peripheral tissues [27, 28, 34, 36, 37]. Graves’ orbitopathy (GO) is the most common and serious extra-thyroidal manifestation, affecting up to 50% of GD patients [27, 37]. GO may be present at the initial diagnosis of thyroid autoimmune disease, or may slowly develop after its onset [37].
Diagnosis of autoimmune hyperthyroidism relies on biochemical testing for anti-thyroid receptor antibody (TRAb) (99% sensitivity and specificity) which represents the diagnostic hallmark of GD. In TNG/TMNG patients, a T3-toxicosis (i.e., low/suppressed TSH with high FT3 and normal FT4) frequently represents the earliest stage of hyperthyroidism, while thyroid autoantibodies are usually absent [9, 27, 28, 32].
Thyroid ultrasonography (TUS) represents a useful tool for imaging evaluation of both GD and TNG/TMNG patients. In GD patients, TUS frequently demonstrates an enlarged gland with different ultrasonographic patterns, such as multiple hypoechoic areas, or diffuse hypoechoic gland, with or without nodules, with an important increase of vascularization on color flow Doppler, which helps distinguish between GD and destructive thyroiditis, particularly when radionuclide imaging is contraindicated, such as during pregnancy and lactation [9, 32]. In non-autoimmune patients, TUS provides information regarding nodule size, echographic features (frequently hyperfunctioning nodule is isoechoic with respect to the surrounding parenchyma), and vascular mapping of the nodule(s) on color flow Doppler evaluation.
Thyroid scintigraphy (TS) performed using Na[99mTc]TcO4 or Na[123I]I is recommended for evaluation of patients with low-normal serum TSH either for identifying hyperfunctioning nodule(s) and assessing the degree of suppression of extranodular thyroid parenchyma [1, 8, 9, 27, 32, 38] or if TRAb is negative and/or GD diagnosis is unclear. Moreover, Na[131I]I uptake (RAIU) is required for proceeding with dosimetry-guided RAI therapy. RAIU is usually elevated in patients with GD while thyroid scintigraphy shows diffusely increased radiotracer activity throughout both thyroid lobes [9, 27, 32]. RAIU values can be either normal or high in TNG/TMNG patients, while thyroid scintigraphy shows heterogenous radiotracer activity throughout the thyroid gland [1].
Fine needle cytology (FNC) is suggested for excluding malignancy for nodules with suspicious features at TUS (e.g., hypoechoic, blurred/irregular margins, microcalcifications (< 2 mm), taller than wide) [39] and hypofunctioning (i.e., cold nodule) at thyroid scintigraphy. TUS should always include imaging evaluation of the cervical lymph nodes [4042].
The medical treatment of autoimmune and non-autoimmune hyperthyroidism with anti-thyroid drugs (ATD; i.e., thionamides, carbimazole, methimazole, and propylthiouracil) is aimed to restore euthyroidism and can be used as a first-line treatment (achieving long-term remission in approximately 30% of GD cases) or to prepare patients before RAI therapy or surgery for reducing the risk of early side effects (mainly in elderly patients or in those with cardiovascular comorbidities) [6, 9, 42].
Finally, surgery which depending on the precise condition can be either a hemithyroidectomy or total thyroidectomy could be the first choice in (i) women planning a pregnancy in < 6 months, (ii) patients with symptomatic compression or large goiters (≥ 80 g), (iii) patients with low RAIU, (iv) patients with documented or suspected thyroid malignancy (including patients with nodules cytologically indeterminate) or large (≥ 4 cm) non-functioning thyroid nodule, and (v) patients with moderate to severe active GO [9]. In GD patients with moderate to severe GO, some authors suggested performing a rhTSH-aided RAI therapy after thyroidectomy for obtaining a total thyroid ablation. This would be more effective than thyroidectomy alone for improving GO [43, 44].

Radioiodine therapy in autoimmune and non-autoimmune hyperthyroid patients

Patient facility

RAI treatment delivery depends on national legislation regarding the therapeutic use of Na[131I]I. In many countries, RAI therapy is performed in an outpatient setting, reserving an inpatient facility for patients affected by important comorbidities and/or in whom severe complications can be expected, and/or according to Na[131I]I activity administrated and local legislation. Finally, an inpatient facility should be preferred in cases of bladder or bowel incontinence since it allows for safe disposal of excreta.
RAI therapy must be performed by a trained nuclear medicine physician in the nuclear medicine unit, with the availability of nursing and medical physics support (mainly if a dosimetric approach is planned).

Patient preparation, information, and instructions

Adequate preparation before performing RAI therapy is very important for reducing the risk of early side effects, such as exacerbation of thyrotoxicosis (i.e., transient rise in FT3 and FT4 levels) due to radiation-induced thyroiditis, occurring in about 10% of all treated patients or, more rarely, the so-called “thyroid storm” (more common in GD patients with poorly controlled hyperthyroidism) [9, 10, 45, 46]. Thus, patients should be ideally treated in a euthyroid state (overt hyperthyroidism should be avoided mainly in patients with active GO). Accordingly, ATD should be stopped 2–5 days before RAI therapy. However, longer withdrawal periods (2–8 weeks) are advised when propylthiouracil (PTU) is used due to its more pronounced radioprotective effect on thyroid tissue than methimazole’s [1, 6, 4751]. However, high cure rates were reported after short PTU withdrawal before RAI therapy in patients treated with personalized dosimetric approaches [52]. Using a dosimetry strategy with an intended absorbed dose of 250 Gy (and achieved dose > 200 Gy) excellent ablative results for GD were obtained with only a 2-day PTU withdrawal period before RAIU testing and subsequent RAI therapy delivery [52].
Performing RAI therapy in euthyroid status may be more important in patients with overt hyperthyroidism and/or in elderly patients and/or in patients with comorbidities (e.g., cardiovascular, cerebrovascular, systemic, or pulmonary hypertension, renal failure) [9, 10, 45, 46, 5355] for avoiding increased heart rate (due to the so-called T3-toxicosis) and possible, even if rare, supraventricular tachycardia including atrial fibrillation or flutter [9].
If possible, ATD medication should not be used in the days/weeks after RAI therapy administration since it can reduce therapy efficacy [10]. However, in elderly patients with pre-existing heart disease, even if asymptomatic, post-therapy use of ATD(s) could be considered starting 3–7 days after RAI therapy delivery [9].
Patient preparation before administration of RAI therapy includes discontinuation of iodine-containing products (e.g., toothpaste, disinfectant, hair dye) since they can reduce RAIU and, as per consequence, the success rate of RAI therapy.
RAI therapy must be postponed for at least 6 months in patients who received amiodarone treatment, and for several weeks to months for patients who received radiographic contrast agent, depending on the type of substance used, e.g., 6–8 weeks for water-soluble agents, vs. 3–6 months for lipophilic agents. In such patients, RAIU is obligatory before RAI therapy [11, 12] while an iodine urine measurement could be useful as well [6]. However, it is not available in everyday clinical practice of many nuclear medicine departments.
The ß-adrenergic blockade should be used both before and after RAI therapy in patients with severe and symptomatic hyperthyroidism, especially in elderly patients and those with cardiovascular disease, for avoiding side effects such as tachycardia or cardiac arrhythmia [6, 56, 57].
In NTG/TMNG patients, RAI therapy should be postponed if serum TSH is high-normal or elevated for avoiding the effect of RAI on normal thyroid tissue since it increases the risk of developing hypothyroidism [9, 58].
In GD patients with active and mild GO and without risk factors like high TRAb level (based on institutional assay and thresholds) and/or smoking, RAI therapy without steroid therapy should be considered as an acceptable approach mainly in patients with comorbidities such as diabetes and/or osteoporosis. Steroid therapy is recommended in such patients if risk factors are present [9]. In addition, in GD patients with active moderate to severe GO in whom RAI therapy is chosen since other treatments are not feasible, steroid treatment is recommended for minimizing the risk of GO worsening, mainly if concomitant risk factors (e.g., smoking) are present [1, 9, 59]. In such patients, intravenous administration of steroids is recommended using a starting dose of 0.5 g once weekly for 6 weeks, followed by 0.25 g once weekly for 6 weeks [58, 59].
For patients referred for RAI therapy who are at high risk of development/worsening of GD orbitopathy, prophylactic steroids oral administration is recommended using a daily dose of 0.2 mg prednisone (prednisolone)/kg body weight for 6 weeks after RAI therapy [59] or alternatively 0.3–0.5 mg/kg/bodyweight as an initial dose, subsequently gradually tapered and discontinued [58].
Steroid therapy was debated for a long time [60] but is not recommended for patients with present but inactive GO, and non-smoking GD patients without apparent GO by current EUGOGO recommendations [58]. Steroid therapy should not be started before RAI therapy being able to reduce RAIU and consequently the efficacy of the treatment [61]. Accordingly, steroid therapy should be started 1–3 days after RAI therapy and continued for several weeks [1, 46, 9].
Lithium therapy is not frequently used in hyperthyroid patients. However, being able to lengthen 131I half-life in the gland (without reducing iodine uptake), it may be used in GD patients with low (< 20%) RAIU starting immediately after RAI therapy and continued until 7 days post-therapy. Presently, its clinical significance on RAI therapy effectiveness is not completely clear even if some literature data showed that adjuvant short-term (7 days) use of lithium (900 mg/day) increases RAI therapy efficacy without an increase in side effects (e.g., orbitopathy worsening in GD patients) [62, 63]. Since side effects are currently reported in about 10% of patients, the use of lithium should be evaluated case by case and considered only if RAI therapy is not feasible without it, and alternative treatment options are not available or not feasible [6].
In general, no special diet is strictly required before RAIU testing and subsequent RAI therapy administration. However, it seems sensible to avoid iodinated salt and reduce iodine-rich food (e.g., seaweeds, fish, milk, and eggs), respectively, for at least 7 days before RAIU [1]. In this light, an iodine urine measurement may be useful in selected cases (i.e., patients whose diet is mainly based on seaweed or is rich in supplements containing iodide) [9].
Pregnancy is an absolute contraindication for RAI treatment. In all fertile females, it can be excluded by obtaining hCG serum testing within 72 h before RAI administration. If there is a suspicion of early pregnancy (not detectable by beta-HCG) according to anamnestic data, RAI therapy should be postponed. Finally, pregnancy must be avoided for almost 4 months after RAI therapy [64]. Breastfeeding must have been discontinued for at least 6 weeks before RAI therapy administration for allowing time for mammary tissue involution with the goal of avoiding high radioiodine uptake and, consequently inadvertent radiation to hypertrophied mammary gland tissue [6, 9, 11, 65].
The nuclear medicine physician must provide verbal and written information to patients prior to therapy administration, addressing the following:
(i)
Aim and strategy of RAI therapy;
 
(ii)
Final functional outcome including the risk of persistent/recurrent disease with possible subsequent re-treatment;
 
(iii)
Medical therapy to use before and/or after RAI therapy;
 
(iv)
Possible early and/or late side effects and their possible treatments;
 
(v)
Radiation precaution instructions according to national laws
 
(vi)
Clinical management, laboratory tests, and imaging studies during follow-up.
 
The patient’s written informed consent must be obtained before RAI therapy administration.

Procedure and Na[131I]I activity

Na[131I]I is preferentially administered orally as a capsule since it allows an easier and safer treatment than using the liquid form or intravenous administration, thus reducing the risk of contamination. Intravenous administration can be used in patients with parenteral nutrition, vomiting, or other causes of the inability to process iodine through the gastrointestinal tract. The liquid form can be used in patients with severe swallowing difficulties; on the other hand, oral administration is not feasible in some countries due to legislative compliance, and therefore, intravenous administration may be required even for patients with swallowing difficulties. In addition, liquid Na[131I]I is generally supplied for intravenous injection, and therefore, administering liquid orally would be classed as an unlicensed way of administering Na[131I]I (off-label) with different absorbed doses to the mouth. If oral administration is chosen, as happen in most cases, patients have to be fasting overnight before RAI therapy and 1–2 h after treatment. Following RAI therapy administration, patients should be encouraged to drink a sufficient quantity of fluid for 24–48 h for reducing the radiation absorbed dose to the bladder (critical organ) and minimize body radiation exposure by enhancing urinary RAI excretion.
The administered Na[131I]I therapeutic activity depends both on the aim (e.g., ablative vs. functional therapy in GD patients) and the strategy (fixed-activity method vs. dosimetric approach) of RAI treatment.
Accordingly, the patients’ restrictions regarding work and with respect to family members and the general population will be different. In general, patients have to be advised to increase the distance and reduce the contact time when interacting with others (mainly children, pregnant, and breastfeeding women) for reducing radiation exposure that in Europe should not exceed 1 mSv cumulative dose per year [6, 13, 64, 66–68].

RAI therapy for patients with autoimmune hyperthyroidism (Graves’ disease)

Introduction

Graves’ disease (GD) is the most common cause of persistent hyperthyroidism in adults living in iodine-sufficient regions and is more prevalent among smokers [1]. GD patients mostly suffer from overt hyperthyroidism.

Definition and epidemiology

GD has an autoimmune etiology due to the loss of immune tolerance to thyroid self-antigens with consequent production of organ-specific auto-antibodies targeted against the TSH receptor (TSH-R) expressed on thyrocyte membrane, the so-called TRAb [27, 28].
GD is the most common form of hyperthyroidism in iodine-sufficient areas accounting for about 80% of all cases. Autoimmune hyperthyroidism has a peak incidence between 30 and 60 years of age, with a higher prevalence in women (1:5–7) [27, 28, 33, 34]. The annual incidence of GD is estimated at 20 − 50 cases per 100,000 individuals [27, 28].

Indication and contraindications (absolute and relative)

a.
RAI therapy is considered the first line treatment for GD patients when:
(i)
There are contraindications to the use of an anti-thyroid drug (ATD);
 
(ii)
Increased surgical risk is documented (e.g., heart failure, laryngeal nerve palsy, pulmonary hypertension)
 
(iii)
Recurrent hyperthyroidism occurs after ATD withdrawal or sub-optimal thyroid surgery;
 
(iv)
Patients are elderly with comorbidities;
 
(v)
Patients do not have access to a high volume thyroid surgeon (especially in pediatric patients);
 
(vi)
Thyroid remnant tissue remained and malignant GO occurred/persisted after surgery [6, 9, 69].
 
 
b.
RAI therapy may be considered a first line treatment for GD patients when:
(i)
They are > 10 years, regardless of gender, having small to medium goiter and inactive GO;
 
(ii)
They are > 10 years, regardless of gender, having small to medium goiter and low to mild active GO; Conversely, RAI therapy is contraindicated for the following GD patients:
1)
Absolute contraindications:
(1)
Pregnant or breastfeeding patients
 
(2)
Patients with the (multi)-nodular variant of GD with suspected or confirmed thyroid cancer
 
(3)
patients ≤ 5 years old
 
 
2)
Relative contraindications:
(1)
Patients with very large goiters (≥ 80 g)
 
(2)
Patients with active moderate-severe GO and high TRAb levels (mainly if smokers) [9, 6975].
 
 
 
 
In such cases, a careful consideration of pros and cons of RAI therapy, surgery, and long-term ATD treatment is advised, taking into account local resources and patients’ preferences, in order to customize patients’ management.
RAI therapy should be delayed as much as possible in children between 5 and 10 years of age [1, 9].

Concepts for RAI therapy delivery in Graves’ disease: functional vs. ablative

There are 2 concepts for RAI therapy delivery in GD patients:
  • The functional dose concept in which the aim of RAI therapy is to correct subclinical or overt hyperthyroidism with the goal of reaching euthyroidism as soon as possible.
  • The ablative dose concept in which the aim of RAI therapy is to achieve hypothyroidism as soon as possible, based on evidence that the definitive success rate in controlling hyperthyroidism is much higher than that obtained with the functional dose concept (> 90% vs. < 70%, respectively) [ 6, 9, 52, 76, 77]. In addition, ablative RAI therapy reduces the volume of the gland correcting both mechanical issues (e.g., dyspnea, dysphagia) and aesthetic features of the neck [ 78].
RAI therapy success rate depends on both radioiodine-administered activity (and its kinetics, i.e., uptake and effective half-life in the gland) and thyroid volume [6, 9, 49, 50, 79, 80]. A recent meta-analysis showed that the delivered radiation absorbed dose to the thyroid is directly related to outcome [7].
A thyroid radiation-absorbed dose of 150 Gy is necessary to obtain euthyroidism according to the functional dose concept [6, 14, 75], while it is necessary to deliver a radiation-absorbed dose to the thyroid ranging between 200 and 300 Gy according to the ablative dose concept [6, 14, 76, 81] (Table 2]. The frequency of persistent hyperthyroidism is very low (8%) when a thyroidal radiation-absorbed dose of approximately 300 Gy is delivered [1, 76].
Table 2
A summary of the intended absorbed dose with respect to thyroid disorders and purpose of RAI therapy
Disease
Absorbed dose
Aim
GD
100–150 Gy
Normalize thyroid function (functional dose concept)
GD
200–300 Gy
Hypothyroidism and replacement (ablative dose concept)
TNG
300–400 Gy
Normalize thyroid function (functional dose concept)
TMNG
150–300 Gy
Normalize thyroid function (functional dose concept)
To date, the unsatisfactory long-term outcome of GD patients treated according to the functional dose concept resulting in a higher incidence of recurrent hyperthyroidism and the risk of possible GO worsening is the main reason for the currently preferred ablative dose concept [6, 77]. Accordingly, our panel members favor the implementation of the ablative dose concept for all GD patients.
In the recently published European Thyroid Association guidelines on the management of GD in pediatric patients [82], the ablative dose concept was also advised as the strategy of choice in pediatric patients since it reduces the risks of both persistent/recurrent hyperthyroidism and delayed radiogenic-induced thyroid neoplasms within residual non-ablated thyroid tissue [6, 8387].

Fixed-activity method

The administration of fixed RAI activity is the easiest and most commonly used method for performing RAI therapy in GD patients for the definitive treatment of hyperthyroidism.
The fixed-activity method was originally based on the administration of a fixed activity to all patients. Moreover it may be refined by estimating the thyroid gland size by palpation, morphological, or functional imaging (i.e., TUS or thyroid scintigraphy, respectively) (i.e., fixed administered activity per mass unit) [1, 6]. In addition, Na[131I]I thyroid gland maximum uptake and turnover should be empirically taken into account when therapeutic RAI activity is planned [88].
Most often, fixed activities between 370 and 555 MBq are employed to treat GD patients [1, 9, 11, 75]. However, higher activities may be required when the thyroid volume is higher than 40 ml. In this setting, a dosimetric approach should be considered [89].
In daily clinical routine, the main advantage of the fixed-activity method is the simplicity of pre-treatment logistics. However, a potential disadvantage of the fixed-activity method is the potential risk of over-treatment, associated with unnecessarily high radiation exposure to the patient, or under-treatment with a higher cumulative incidence of persistent/recurrent hyperthyroidism [1, 78, 90, 91]. Notably, however, neither randomized clinical trials nor retrospective comparison studies demonstrated the inferiority of the fixed-activity approach compared to dosimetric approach.

Dosimetric approach

Results of studies reporting the relationship between the thyroid radiation absorbed dose and treatment outcome are conflicting [92]. However, a multicentric prospective randomized trial [93] and a systematic review and meta-analysis including the most relevant retrospective studies [7] on RAI therapy of Graves’ disease with complete dosimetry showed that there is a significant dependence of treatment success rates on the actual thyroid radiation absorbed dose. According to the combined analysis [7], the delivery of 150 Gy, 200 Gy, and 300 Gy to the thyroid gland is expected to eliminate hyperthyroidism in 74%, 81%, and 88% of cases, and restore euthyroidism in 38%, 35%, and 29% of cases, respectively. Besides the thyroidal radiation absorbed dose, a larger size of the thyroid gland (often associated with fast iodine kinetics) appears to be an independent risk factor for residual disease [76, 93, 94].
A target dose of at least 200 Gy is recommended for the definitive treatment of patients with refractory Graves’ disease. Up to 300 Gy is appropriate especially for large glands and very pronounced hyperthyroidism if a more reliable therapeutic effect is desired in the short term. If restoration of a euthyroid state is reasonably possible in milder disease with a low risk of recurrence and is desired by the patient, approximately 150 Gy target dose should be aimed for. The success of the therapy is not guaranteed due to uncertainties in determining the therapeutic RAI activity and individual variables that have not yet been sufficiently investigated. Patients should be informed that treatments with lower target radiation absorbed dose are more likely to require retreatment and that the likelihood of hypothyroidism requiring lifelong thyroid hormone replacement increases with an escalation of the target radiation absorbed dose. Even if euthyroidism is successfully restored after treatment, regular long-term monitoring of hormonal status remains necessary because the rate of hypothyroidism increases continuously over years following RAI therapy.
If the therapeutic RAI activity is estimated from an early uptake measurement according to Eq. (3], an estimate of the effective half-life of Test = 5.5 days suggests that approximately 6, 8, and 12 MBq multiplied by the mass in gram and divided by the 24-h Na[131I]I uptake (RAIU) are to be administered for target doses of 150, 200, and 300 Gy, respectively.
Example: A patient with 35-g thyroid mass and 65% 24-h Na[131I]I uptake, RAIU(t = 24 h) = 0.65, who is to be treated with a thyroid absorbed dose of 200 Gy, should be given Aa = 8·35/0.65 MBq = 430 MBq. In the case of fast iodine kinetics, recognizable by an early and high iodine retention and high Technetium Thyroid Uptake (TcTU) in the Na[99mTc]TcO4 scintigram, a shorter half-life in the tissue and therefore underestimation of the absorbed dose is to be expected. Especially with large thyroid glands, which might be less sensitive to radiation, this can lead to high failure rates [94] justifying to aim for absorbed doses higher than 200 Gy.
If a pre-therapeutic assessment of the kinetics is omitted, activity dosage should be linear to the mass M assuming that about half of the administered activity decays in the gland, corresponding, e.g., to iodine kinetics with 79%, 67%, or 57% 24-h Na[131I]I uptake and 4.5, 5.5, or 6.5 days, respectively, the effective half-life of the activity in the gland. The therapeutic activity is calculated as 9, 12, and 18 MBq multiplied by the mass in grams for target doses of about 150, 200, and 300 Gy, respectively. Example: A thyroid gland with 20-g mass is to be treated with Aa = 9·20 MBq = 180 MBq for an absorbed dose of 150 Gy.

RAI therapy for patients affected by non-autoimmune toxic nodular goiter/toxic multinodular goiter

Introduction

Hyperfunctioning thyroid nodules — either isolated (TNG) or in the context of a multinodular goiter (TMNG) — represent approximately 5–10% of all thyroid nodules, and they are the most common cause of hyperthyroidism in elderly patients who come from iodine-deficient areas [38, 9597]. The degree of hyperfunction is variable in such nodules. Accordingly, they can produce either sub-clinical or overt hyperthyroidism [98102].

Definition and epidemiology

A somatic activating mutation affecting the thyrotropin receptor (TSHR) gene and the gene encoding for the stimulatory GPT-binding protein (Gsα) have been reported as the main cause of TNG and have also been described in many TMNG patients [1, 103105].
In iodine deficiency areas, TNG and TMNG represent the main cause of hyperthyroidism accounting for approximately 60% of all cases while their prevalence significantly decreases in the iodine-sufficient areas (e.g., USA) being 15–30% of hyperthyroidism cases [106].

Indications and contraindications (absolute and relative)

RAI therapy is a safe and effective therapeutic option for the definitive treatment of hyperthyroidism in patients affected by TNG or TMNG [1, 107].
RAI therapy is considered the first-line treatment option for patients affected by subclinical or overt hyperthyroidism in the following circumstances: (i) a solitary hyperfunctioning nodule (i.e., Plummer disease); (ii) one or multiple hyperfunctioning nodules in multinodular goiter without suspected or confirmed thyroid cancer; (iii) advanced age; (iv) comorbidities (e.g., cardiovascular, cerebrovascular, systemic, or pulmonary hypertension) resulting in higher surgical risks; (v) previous history of neck surgery and/or irradiation; (vi) limited or no access to a high volume thyroid surgeon [1, 9].
Conversely, RAI therapy is contraindicated for the following TNG or TMNG patients:
I.
Absolute contraindications:
1)
Pregnancy or breastfeeding
 
2)
Suspected or confirmed thyroid cancer
 
 
II.
Relative contraindications:
1)
Large TMNG
 
2)
Compressive neck symptoms and/or signs [1, 9].
 
 

RAI therapy for non-autoimmune toxic nodular goiter /toxic multinodular goiter

In TNG or TMNG patients, the first goal of RAI therapy is to correct subclinical or overt hyperthyroidism, preferably restoring euthyroidism. The success of RAI therapy is mainly related to radioiodine-administered activity and its kinetics in TNG or TMNG and nodule(s) volume [1, 9, 108].
In this light, even if some authors suggested lower absorbed doses in the past [109] an absorbed dose to the target(s) ranging from 150 to 300 Gy is currently recommended to correct hyperthyroidism [6, 110113], while higher absorbed doses (i.e., up to 400 Gy) were reported to slightly improve RAI therapy success rate [113] (Table 2].
Overall, the success rate is very high ranging from 81 to 94% of TMNG and TNG patients, respectively [1, 6, 9, 78, 110113]. The risk of persistent or recurrent hyperthyroidism is higher in TMNG than in TNG patients, reaching up to 20% of all treated patients [1, 9, 78, 114116]. Conversely, the risk of developing hypothyroidism is higher for patients > 45 years old and/or treated with higher radioiodine activities and/or with higher RAIU and/or partial suppression of extra toxic nodule(s) parenchyma and/or pre-treated with ATD [1, 9, 117, 118]. The incidence of hypothyroidism after RAI therapy ranges between 20 and 60% depending on disease (TNG or TMNG), degree of extra-nodular suppression, and length of follow-up [1, 90, 92, 112, 117121].
The second aim of RAI therapy in such patients, mainly in those with larger goiters, is to reduce the volume of hyperfunctioning nodule(s) and/or goiter (if extra-nodular thyroid parenchyma is not suppressed), thus alleviating mechanical complaints (e.g., dysphagia, dyspnea) that affect a significant number of such patients. Toxic thyroid nodule(s) volume reduction is 35% within 3 months and up to 45% after 24 months following RAI therapy administration [1, 78, 114, 122].

Fixed-activity method

As for GD patients, the administration of predefined fixed RAI therapeutic activities is the easiest and most commonly used method for the delivery of RAI therapy in TNG and TMNG patients.
According to this method, the estimation of hyperfunctioning nodule(s) volume is obtained by palpation or preferably by TUS or TS [1, 6] and 131I nodule(s) maximum uptake and turnover (evaluated by RAIU trend) should be considered for RAI therapy planning.
However, the fixed-activity method has limited accuracy [1, 91], and consequently, the cumulative incidence of persistent/recurrent hyperthyroidism (mainly in TMNG patients) or, conversely, of hypothyroidism (mainly in TNG patients whose extranodular parenchyma is not suppressed) is not negligible [1, 78, 90, 118].
Most often, fixed activities ranging from 370 to 740 MBq are used to treat TNG or TMNG patients [1, 9, 11].
RAI therapy efficacy is limited when hyperfunctioning nodule(s) volume is > 26 ml. In such patients, a dosimetric approach is considered more useful for obtaining an effective treatment [89].
The fixed-dose method offers the same advantages and limitations already discussed for RAI therapy in GD patients.

Dosimetric approach

In non-autoimmune toxic goiter, dosimetry should ideally only take into account the kinetics in the autonomously functioning tissue, whereby not all, but only part of the thyroid gland parenchyma is affected, typically in the form of one or more nodules. With uptake probes usually used to measure activity kinetics [14], dosimetry is reliably possible if the mass of the autonomic tissue can be measured and the healthy thyroid tissue is completely suppressed. Underdosing and the consequent risk of persistent or recurrent hyperthyroidism occurs when a non-negligible fraction of the measured activity is located in unsuppressed healthy tissue or autonomously functioning tissue outside of the nodules taken into account in the mass determination. The main risks for developing hypofunction are incomplete suppression of the remainder of the gland and a high targeted absorbed dose in a large target volume. In toxic goiter with a single or only a few reliably measurable nodules, RAI treatment based on complete dosimetry of the autonomously functioning volume eliminates hyperthyroidism in 85–100% of individuals treated with 300–400 Gy target dose with rates of hypothyroidism typically between 10 and 20% [112, 113].
When calculating the target dose for the treatment of toxic nodule, the measured iodine uptake must be corrected by the nodule/whole thyroid uptake ratio obtained from the scintigram. The target dose for the nodule is 300–400 Gy.
When the estimation of a target mass is not possible, absorbed doses of 100–150 Gy to the mass of the entire gland should be applied to calculate the therapeutic RAI activity [92, 110, 112].
If complete dosimetry is omitted, activity dosing should be based on an early uptake measurement, as indicated by a meta-analysis [123]. Equation (3] suggests administering approximately 4, 6, 8, 12, or 16 MBq multiplied by the mass in gram and divided by the 24-h Na[131I]I uptake for target doses of 100, 150, 200, 300, or 400 Gy, respectively. The suggested value of 5.5 days for Test is about 10% less than the mean effective half-lives typically observed in most toxic nodular goiters [110, 113] but limits the potential error to ± 25% for the vast majority of individuals [124]. Examples: A patient with a 10-g nodule with 30% 24-h Na[131I]I uptake should be administered Aa = 12·10/0.3 MBq = 400 MBq for a nodule absorbed dose of 300 Gy. Another patient with multinodular goiter in a gland with 30-g total mass and 40% 24-h Na[131I]I uptake needs Aa = 6·30/0.4 MBq = 450 MBq for a mean thyroid absorbed dose of 150 Gy.
The dosimetric approach following the European standard operational procedure provides the most accurate results regarding achieved and intended target doses. The Marinelli formula seems to enable minimal deviations of achieved doses from target doses in TMNG, although other individualized strategies taking into account at least the thyroid volume also result in low deviations of presumed achieved dose from the intended dose [14, 125].

RAI therapy for patients with non-toxic goiter

Introduction

RAI therapy has been used for the volumetric reduction of goiter and/or thyroid nodule(s) in patients with benign non-toxic nodular goiter (NTG) for the past 30 years [92, 126, 127]. The current guidelines support the use of RAI therapy as an alternative to thyroid surgery, particularly for patients considered at high surgical risk due to significant comorbidities, or in patients who wish to avoid surgery [3, 126, 128, 129].

Definition and epidemiology

NTG is defined as thyroid gland enlargement, with or without intrathyroidal nodules, associated with TSH, FT3, and FT4 values within the reference range.
It is one of the most prevalent thyroid disorders, particularly in iodine-insufficient regions, with a worldwide population health impact.
Thyroid nodules represent a very common clinical presentation, being diagnosed by clinical evaluation and/or high-resolution TUS in up to 60–70% of the overall population, with increased prevalence in female patients and the elderly [9, 102, 130132].
The incidence of NTG in iodine-sufficient areas is about 0.1/350.000 new diagnoses per year. However, the incidence is higher (2/350.000 per year) in the population that underwent radiation exposure of the neck [133].

Indications and contraindications for RAI therapy in NTG

RAI therapy is indicated for NTG patients with compressive symptoms and a total thyroid volume < 100 ml, particularly if elderly and/or with comorbidities (e.g., cardiovascular) and/or with a history of previous neck surgery [126]. In addition, RAI therapy is indicated for the treatment of large NTG with intrathoracic extension when surgery cannot be performed or is refused by the patient [126].
Of course, the choice to treat such patients using RAI depends on the local expertise, radiation regulations, and patient’s preference [126].
Absolute contraindications to RAI therapy include the following: (i) pregnancy or lactation (however such patients are generally elderly); (ii) cases with suspected or confirmed thyroid cancer, and (iii) patients in whom a fast resolution of tracheal compression is required. Relative contraindications to RAI therapy include (i) patients with 24 h RAIU value < 15% (however, rhTSH may be selectively used in such cases), (ii) thyroid volume > 100 ml; (iii) when the nodule responsible for symptoms (e.g., dyspnea and/or dysphagia) is scintigraphically hypofunctional/cold [126].

RAI therapy approach for NTG patients

Since clinical, diagnostic, and therapeutic management of NTG patients is controversial, nuclear physicians must perform an accurate patient selection for RAI therapy taking into consideration, the risk of treatment failure, goiter recurrence/persistence, and patient’s preferences. Accordingly, a detailed personal and familiar history, clinical examination, laboratory tests, and imaging evaluation are needed.
Thyroid ultrasonography is used to obtain information regarding nodule(s) size and topography within the thyroid gland, echographic features, and vascular mapping on color flow Doppler evaluation. In addition, TUS-guided FNC is a fundamental tool for ruling out malignancy [9].
Second-level morphological imaging (i.e., CT without contrast or MRI) should be considered in NTG patients with (very)-large goiter and/or with intrathoracic extension for whom high-resolution three-dimensional visualization of the thyroid is required to calculate gland volume and to evaluate its relationship with the surrounding anatomical structures [134]. CT or MR-based estimations of the smallest cross-sectional tracheal area permit comparison between baseline and post-therapeutic measurements to assess RAI therapy results [134138]. However, the agreement between CT and MR in evaluating both thyroid gland volume and the smallest cross-sectional tracheal area is not known. As per consequence, the same imaging modality should be used before and after RAI therapy [134].
Thyroid scintigraphy performed using Na[99mTc]TcO4 or Na[123I]I is indicated for patients with low-normal serum TSH (especially in iodine deficiency areas) both for excluding hyperfunctioning nodule(s) and for identifying cold nodule(s) and their topography within the gland [102], and for evaluating radiotracer distribution heterogeneity throughout the thyroid gland, which may decrease RAI therapy efficacy [122, 134, 139]. RAIU is a fundamental diagnostic tool for the evaluation of NTG patients since the prescribed therapeutic RAI activity should be adjusted according to RAIU values [74, 102, 122].
Currently, no worldwide consensus exists regarding the management of NTG patients recognizing that no ideal treatment is available. As per consequence, more than 30% of clinicians prefer an observational approach avoiding any type of treatment, especially for patients with mild or moderate compressive symptoms in whom malignancy has been ruled out [24, 102, 134].
Iodine supplementation has been suggested as an adequate and feasible approach for the prevention of NTG since goiter development is strongly associated with iodine deficiency, even if mild [134, 140]. However, there is limited information regarding its efficacy in patients with already established goiter, while its use has been associated with an increased incidence of Hashimoto’s thyroiditis and papillary thyroid carcinoma [134, 141]. However, these possible effects are overlooked regarding the routine use of iodine supplementation in a large part of Europe and in North America [3, 4, 134].
Levothyroxine therapy is generally the preferred choice of many members of both the European Thyroid Association and the American Thyroid Association (ATA) [3, 4]. However, there is no agreement regarding its efficacy, and several authors have demonstrated that a sufficient goiter reduction can be observed in only a limited number of patients [134, 142144]. In particular, goiter reduction observed in patients on levothyroxine therapy is lower than that observed in patients who underwent RAI therapy [140]. Moreover, the gland volume tends to return to baseline values when levothyroxine therapy is discontinued [142].
Finally, for obtaining goiter reduction, levothyroxine therapy aimed at suppressing the serum TSH level is often prescribed resulting in sub-clinical or overt hyperthyroidism, which should be avoided, especially in elderly (> 60 years) patients due to untoward side effects on the cardiovascular and skeleton systems [134, 145, 146].
Based on the above considerations, levothyroxine therapy should be suggested only in young NTG patients with modestly increased thyroid volume [134].
Surgical removal of the thyroid gland [especially (near)-total-thyroidectomy] should be the preferred therapy in selected NTG patients (see the specific paragraph). The main advantage of surgery is related to the immediate and significant goiter reduction with prompt relief of compressive symptoms and histopathological analysis of the thyroid tissue [134].
However, some potential, permanent or temporary, side effects must be taken into account when the surgical approach is planned [134, 143, 147]. In particular, recurrent laryngeal nerve palsy, hypoparathyroidism, respiratory complications, and tracheomalacia can occur in a not negligible number of patients, mainly in those with larger NTG with substernal extension [134, 148152].
In addition, goiter recurrence can be observed in NTG patients who underwent less than subtotal thyroidectomy [134, 153156]. In such patients, RAI therapy represents a favorable alternative to reoperation [134, 153156] for avoiding the risk of permanent vocal cord paralysis and/or hypoparathyroidism that is 3-to-tenfold higher than at initial operation [134, 148, 149, 157].
For reducing the risk of surgical complications, thyroid surgery should be always performed by expert surgeons in high-volume centers [102].

RAI therapy delivery for treatment of non-toxic goiter

The main goal of RAI therapy in NTG is to reduce the volume of the thyroid gland [3, 92, 126, 128, 129]. Following RAI therapy administration goiter size volumetric reduction of approximately 40–50% after 1 year, and up to 60% after 2–5 years has been reported [92, 122, 126, 138, 144, 158165]. The response to RAI therapy is individual and may be different among patients but symptoms most often improve and patient satisfaction is high [126, 138, 160, 165, 166].
No consensus exists regarding the prescribed therapeutic RAI activity for the treatment of NTG [92, 126, 167]. Based on published clinical experiences, a therapeutic RAI activity ranging from 3.7 to 14 MBq/g thyroid tissue corrected for RAIU has been recommended, to impart 100–175 Gy radiation absorbed dose to the thyroid tissue [122, 126, 138, 158, 159, 161165]. Certainly, the most pronounced thyroid volume reduction rate was observed when the highest thyroid radiation absorbed dose (i.e., 175 Gy) was used [126, 168], but this approach increases the whole-body radiation exposure [126, 169].
However, RAI therapy efficacy is inversely correlated to initial NTG volume. As per consequence, in patients with very large NTG (i.e., > 100 ml) in whom RAI therapy is chosen as the preferred treatment, the volume reduction rate is around 35% after 1 year [92, 122, 126]. Likewise, efficacy is reduced in NTG patients with low RAIU values [92, 126].
Repeated courses of RAI therapy have been reported to result in significantly larger rates of goiter reduction in patients with very large goiters, and this option should be considered especially for patients unable or unwilling to undergo reductive thyroid surgery. Furthermore, while resolving cosmetic concerns may take a more extensive volumetric reduction, quite often the reduction in volume seen after a single RAI therapy in very large goiters is still sufficient to alleviate compressive complaints to such an extent that a second RAI therapy is obviated.

Patient facility

The patient facility can be different according to national legislation on the therapeutic use of 131I. Please refer to the paragraph on RAI therapy for hyperthyroidism.

Patient preparation, information, and instruction

As per hyperthyroid patients, iodine-containing products such as administration of amiodarone or radiographic contrast agents should be discontinued well in advance of therapy, or better avoided altogether since they reduce RAIU and, consequently, RAI therapy efficacy can be decreased [1, 6, 912]. Patients with (very)-large NTNG (≥ 100 ml) and severe trachea narrowing (especially if < 1 cm) should always be treated under steroid coverage (e.g., prednisolone, 25 mg daily for 14 days) [6, 126]. Moreover, consideration for inpatient RAI therapy, mainly for elderly patients with comorbidities (e.g., heart failure), will ensure a hospital setting for addressing possible post-treatment complications [6, 144, 158, 170].
Patient preparation, logistical information, and instructions for the delivery of RAI therapy for NTG patients are similar to hyperthyroid patients regarding:
(i)
The advice to avoid/reduce the intake of iodized salt, seaweed, iodine-containing nutritional supplements, and iodine-rich foods such as sea fish, milk, and eggs for at least 7 days before RAIU and subsequent RAI therapy [1]. Urinary iodine measurement should be performed before RAIU/RAI therapy in patients whose diet is mainly based on seaweed or is rich in iodide-containing nutritional supplements [9];
 
(ii)
Pregnancy is an absolute contraindication for RAI treatment. In all fertile females, it can be excluded by obtaining hCG serum testing within 72 h before RAI administration. If there is a suspicion of early pregnancy (not detectable by beta-HCG) according to anamnestic data, RAI therapy should be postponed. In addition, pregnancy must be avoided for 4–6 months after RAI therapy, while breastfeeding must be discontinued for at least 6 weeks before RAI therapy administration for avoiding high radioiodine uptake and, consequently, radiation absorbed dose to the breasts [6, 11].
 
(iii)
The instructions that nuclear medicine physicians must provide to patients before RAI therapy.
 
(iv)
The requirement for obtaining written informed consent from patients.
 

Procedure and Na[131I]I activity

As for hyperthyroid patients, Na[131I]I is preferentially administered orally in the form of a capsule but liquid form or intravenous administration can be used in patients with severe swallowing difficulties or patients with parental nutrition, respectively.
For rapid 131-I absorption, it is recommended that oral RAI therapy is administered on an empty stomach and this condition must be maintained in the following 1–2 h.
To reduce the radiation absorbed dose to critical organs and diminish radiation exposure to the entire body, patients should be encouraged to drink a large quantity of fluid for at least 24–48 h following RAI therapy.
The prescribed Na[131I]I therapeutic activity largely depends on the target thyroid volume, the RAIU values, and radioiodine distribution in the gland.
Accordingly, the instructions regarding patients’ restrictions on work, with family members, and with respect to the general public will vary depending on the amount of prescribed radioactivity.
Prior to RAI therapy administration all patients must receive detailed information for reducing both radiation exposure and contamination risk to others, particularly to children, pregnant, and breastfeeding women [6, 13, 6466, 67,68].

Fixed-activity method

The easiest and most common method used for performing RAI therapy in NTNG patients is the administration of fixed RAI activities.
According to this method, the estimation of the target treatment volume is obtained by palpation and/or TUS and/or CT or MR (in cases with sub-sternal extension), functional mapping of the goiter is obtained by TS, and RAIU testing should be performed for RAI therapy planning [1, 6, 136, 160, 171178].
There is no consensus regarding the fixed RAI activity prescribed for NTG treatment and a wide range of RAI activities have been reported [92, 126, 136, 160, 171179]. Most often, fixed activities ranging from 600 to 1110 MBq are used to treat NTNG patients [126, 136, 159, 160, 171177].

Dosimetric approach

Although the volumes of euthyroid goiters often are substantially increased, the biokinetics usually corresponds approximately to those of healthy thyroids with normal or only moderately increased radioiodine uptake and a long half-life within the gland [168]. After pre-therapeutic dosimetry, treatment should target a mean thyroid absorbed dose of about 100 Gy [134], in selected cases up to 150 Gy. An often-used activity of 3.7 MBq multiplied by the mass in gram and divided by the 24-h 131I uptake administers a mean thyroid absorbed dose of 120 Gy to a large goiter treated for size reduction if the effective half-life of the activity in the gland is 7 days.
The dosimetric approach following the European standard operational procedure provides the most accurate results regarding achieved and intended target doses [14, 125].

rhTSH-aided RAI therapy for NTG

Although rhTSH use was initially restricted to thyroid cancer patients, it has been used since the early 2000s to increase thyroidal RAIU in patients with NTG [173, 180, 181]. Administration of rhTSH is associated with goiter reduction enhancement by 35–55% as compared with Na[131I]I therapy without rhTSH stimulation. The adjuvant role of rhTSH in RAI therapy for NTG has been studied mainly for patients with very large goiters and in those with a low baseline RAIU [179]. However, rhTSH is not yet approved for this indication, and administration in a patient with NTG represents an off-label use. Therefore, the current guidelines cannot endorse the general use of rhTSH for the treatment of NTG, unless rhTSH-aided RAI therapy is regulated according to local rules (e.g., by authorization of the local Ethical Committee) and cautiously chosen individually for each patient in view of the risks, benefits, experience of the treating physician, and patient’s personal preference.
Declining efficacy with increasing goiter size and low thyroidal RAIU are the major challenges in conventional RAI therapy for NTG, and these factors represent the main indications for rhTSH-stimulated RAI therapy [182]. The potential benefits of rhTSH-aided RAI therapy are based on the marked increase in thyroidal RAIU, resulting in increased radiation absorbed dose to the thyroid without increasing the administered RAI activity (superiority strategy) or reduction of the administered RAI activity while maintaining the same thyroidal radiation absorbed dose (dose-reduction or equality strategy) [183]. According to published literature, rhTSH should be administrated 24 h before RAI therapy (optimal time interval) rather than 2 h, 48 h, or 72 h for maximizing the thyroidal RAIU increase [92, 126, 168, 184]; however, no consensus exists about the dose of rhTSH for aiding RAI therapy. Although in most studies a dose of 0.1–0.3 mg has been given, a wide range of doses (i.e., 0.005–0.9 mg) have been used [92, 126, 174, 185]. However, the choice of rhTSH dose to use for aiding RAI therapy is strongly related to the purpose of the treatment. Indeed, a dose of 0.1 mg is recommended in the equality strategy, whereas a higher rhTSH dose is employed (i.e., 0.3 mg) coupled with higher prescribed therapeutic RAI activity in the superiority strategy [92], resulting in a higher goiter reduction as compared to the equality approach [92]. All in all, based on the present knowledge the optimal rhTSH dose for aiding RAI therapy is most likely in the range of 0.03–0.1 mg. Indeed, in this dose range, a significant improvement of the thyroidal RAIU is obtained while minimizing the risk of side effects [126].
The rationale for using rhTSH-aided RAI therapy in NTG is to obtain an enhanced goiter volume reduction (GVR) which is maintained for several years after therapy with the goal of reducing the need for additional therapy, such as a second RAI treatment or thyroid surgery. However, little is known about the long-term risk of goiter recurrence after RAI therapy, whether with or without rhTSH stimulation [186]. Reported adverse events include administered RAI activity-dependent increased risk of thyroid hyperfunction and acute thyroid swelling when RAI therapy is preceded by rhTSH stimulation. While other serious acute side effects seem of little concern when using low rhTSH doses, the more pronounced goiter reduction, seen with rhTSH-stimulated RAI therapy, is unfortunately achieved at the expense of an up to fivefold increase in the rate of permanent hypothyroidism [175, 182].
Pretreatment with a single, low rhTSH dose allows a reduction of the prescribed RAI activity in patients with nodular goiter and significantly enhances goiter volume reduction in some studies. Notably, however, available evidences do not support its routine clinical use, and the rhTSH is not EMA-approved for this indication. Its out-of-label use requires strict compliance with local laws and recommendations under the responsibility of nuclear medicine physicians.

RAI therapy in pediatric patients

Special considerations for RAI therapy in children

There are special considerations regarding RAI therapy in pediatric patients, especially in pre-pubertal children who are much more susceptible to the harmful effects of ionizing radiation. Furthermore, the developing body of pediatric patients requires consideration of body size before adapting RAI therapy concepts that were designed for fully-grown adults. The special considerations applicable to pediatric RAI therapy are discussed below:

Disease spectrum

Autonomous nodules or obstructive goiter are very rare in children; nearly all patients referred for RAI therapy have been diagnosed with GD refractory to medical therapy with ATD. Therefore, unless explicitly stated otherwise, the recommendations and considerations in this section will pertain to GD.

Age

The reluctance for employing RAI therapy in pediatric GD increases with decreasing patient’s age. According to the recently published European Thyroid Association Guidelines [82] on the treatment of Graves’ disease in children and adolescents, Na[131I]I can theoretically be used in any patient with GD. However, a very young age (< 5 years; because of a greater long-term theoretical risk of malignancy) is seen as a near-absolute contraindication. Age 5–10 years is regarded as a relative contraindication. Interestingly, an update in this guideline is a recommendation that ATD should be maintained for multiple years in pediatric GD patients so that rather than being referred for surgery, children with a near-absolute or relative age-based contraindication for RAI therapy patients can often be kept on ATD until they reach an age adequate for undergoing RAI therapy.

Patient preparation

Other than the standard preparations as described for adults (e.g., stopping ATD), no special medical preparations are necessary for pediatric patients. Of course, depending on the age of the patient and the possibilities allowed by local radiation protection regulations, it may be possible or even necessary to allow a parent to be co-hospitalized with the patient after appropriate preparation of the parent/caregiver.

Therapeutic concepts

For pediatric GD patients, the aim of RAI therapy is (functional) thyroid ablation in order to minimize the risk of GD recurrence and possible future malignant transformation of persistent, viable cells with radiogenic-induced genetic damage. To achieve this aim, a number of concepts have been described in pediatric populations, including a fixed activity approach with activities ranging from 200 to 800 MBq [6], a weight-based approach of 15 MBq Na[131I]I per gram thyroid tissue (as estimated by ultrasound) and a dosimetric concept aiming at a delivered dose to the thyroid of at least 300 Gy [6, 12].
There is no definitive literature evidence demonstrating the superiority of one of these concepts in a pediatric population, although a recent systematic review in adults was able to clearly demonstrate that higher delivered radiation absorbed doses are associated with higher rates of successful functional thyroid ablation [7]. Although fixed-activity concepts have been demonstrated to be successful, they do carry a risk of over-treatment: the activity given might be higher than necessary for achieving an ablative dose to the thyroid, exposing non-target tissue to unwarranted high radiation doses, which is particularly unjustifiable in pediatric patients. Conversely, there is also a risk of undertreatment, thus exposing non-target tissues to radiation whilst not achieving the therapeutic goal. Therefore, for optimizing the chance of success, while at the same time minimizing excessive radiation exposure to non-target tissues, we recommend performing thyroid dosimetry if available. This recommendation also applies to the treatment of autonomous nodules in children.
If hyperthyroidism persists after the first RAI therapy course, repeat RAI treatment after a sufficiently long interval (i.e., 3–6 months) is possible, similar to adult patients.

Side effects in pediatric patients

Side effects following RAI treatment in pediatric patient population are very rare and include occasional mild neck tenderness over the thyroid area in the first-week post-RAI therapy administration. Notwithstanding comparatively high administered RAI activities, for RAI therapy aimed at functional thyroid ablation (i.e., hypothyroidism) in pediatric patients, observational studies found no evidence of increased risk of malignancy or fertility problems after almost 40 years of clinical follow-up [86, 187189].

Alternative treatment

For pediatric patients, the selection of RAI therapy versus total thyroidectomy is often a contentious choice. Local opinion and expertise will also play a role. Of course, each pediatric case necessitates interdisciplinary consultation including a pediatric endocrinologist, thyroid surgeon, and nuclear medicine physician specialized in thyroid disease. The definitive treatment choice involves shared decision-making with the patient and the parents/legal guardians. An absolute or relative contraindication for surgery or RAI therapy such as very young age, pregnancy, or a markedly elevated risk of perioperative morbidity may direct the choice of treatment modality. Surgery remains the preferred definitive treatment option for GD patients < 10 years of age, patients with other (relative) contraindications for RAI therapy, those with a large or nodular goiter, and those with active Graves’ ophthalmopathy necessitating quick complete removal of thyroid tissue.

Adverse effects of RAI therapy

Although RAI therapy is safe and usually well-tolerated, either acute or late side effects may occur, mainly in patients with uncontrolled severe hyperthyroidism and/or active Graves’ orbitopathy [1, 92]. However, patients with overt and uncontrolled hyperthyroidism should not be treated, while those with severe active thyroid orbitopathy should be firstly considered for surgery treatment [1, 190].
The main side effects are summarized in Table 3 and briefly reported below.
Table 3
Summary of early and late side effects
Side effect
Onset
Pathophysiology
Symptoms
Thyroid swelling
Early
Inflammatory reaction to irradiation
Thyroid pain
Sensation of thyroid growth
Dyspnea in patients with a large goiter
Radiation thyroiditis and post-therapy thyrotoxicosis
Early
Transient rise in fT3 and fT4 levels
Exacerbation of hyperthyroidism symptoms
Thyroid storm (rare):
-high fever
-central nervous system manifestations, gastrointestinal and hepatic manifestations
-heart failure
Radioiodine-Induced sialadenitis
Early/late
Concentration in salivary of iodide due to the sodium iodine symporter expression
Swelling,
Periductal pressure Duct constriction Pain
Xerostomia
Taste dysfunctions
Immunogenic effects
Early
Release of thyroid antigens from destroyed follicular cells with increase of TRAb,
RAI therapy causes the transient increase of TRAb
Hypothyroidism: transient or persistent
Late
Transient hypothyroidism: unclear cause
Persistent hypothyroidism:
Thyroid irradiation
Transient hypothyroidism: no symptoms or signs, only biochemical
Persistent hypothyroidism:
Typical sign of hypothyroidism
Graves’ orbitopathy
Late
B-cells and macrophages activation with cytokines secretion
Worsening or appearance of orbitopathy

Early side effects

Early side effects can occur either promptly or during the first weeks after RAI therapy. In general, they are especially correlated to initial thyroid volume and hyperthyroid status before RAI administration.

Thyroid swelling

Thyroid swelling is an early side effect that can be observed in patients with large toxic or, more frequently, non-toxic goiter. Patients could manifest thyroid pain and sensation of anterior neck discomfort due to the inflammation process caused by irradiation of thyroid tissue (i.e., radiation-induced thyroiditis). There are rare reported cases of elderly patients with very-large nodular goiters experiencing rapid thyroid enlargement due to edema which may cause tracheal compression and dyspnea. This possible side effect should always be considered, particularly when rhTSH-aided RAI therapy is planned in patients with large NTG or patients with known tracheomalacia [1, 160].
The aforementioned symptoms are generally mild or moderate. As per consequence, medical therapy is not usually required or may be considered to alleviate local pain symptoms (e.g., paracetamol). However, in patients with higher risk factors (e.g., larger goiter size), they can be prevented/treated using a non-steroidal anti-inflammatory agent for 24–48 h after RAI administration while corticosteroids should only be used in selected patients since it can reduce RAI therapy efficacy [1, 61].

Radiation thyroiditis and post-therapy thyrotoxicosis

The prevalence of radiation thyroiditis is very low with about 1% of all treated patients. It may occur during the first weeks after RAI therapy and can produce a transient rise of serum thyroid hormone levels due to follicular cell damage.
As per consequence, a mild to a severe worsening of thyrotoxicosis may occur in about 10% of patients, mainly among those with poorly controlled hyperthyroidism before RAI therapy [54]. Conversely, the so-called thyroid storm is a rare but life-threatening condition that requires immediate diagnosis, comprehensive, and advanced medical treatment using anti-thyroid drugs (ATDs), inorganic iodide, beta-adrenergic receptor antagonists, corticosteroids, and antipyretic drugs [191] and, if all else fails, emergency thyroidectomy.
A careful selection of the optimal time point for performing RAI therapy and correct patients’ premedication are mandatory for avoiding/reducing the risk of thyrotoxicosis worsening and, especially, thyroid storm condition [1].

Radioiodine-induced sialadenitis

Although it is a more frequent complication in case of RAI therapy for differentiated thyroid cancer, radioiodine-induced sialadenitis can also occur in hyperthyroid patients, especially if treated with a high RAI activity (e.g., ≥ 1000 MBq), both as an acute and a late side effect [1]. Iodine is concentrated in the salivary gland by the NIS and then secreted into saliva. During this process, salivary glands are exposed to dose-dependent radiation damage [192]. Clinical symptoms include salivary gland swelling (frequently transient), local pain, xerostomia, or dysgeusia (taste dysfunction), which may occur in as many as 20–30% of patients [192, 193]. The use of lemon juice and/or salivation-inducing snacks (like lemon/orange candy), starting 24 h following RAI therapy is a valid radioprotective procedure to diminish RAI damage to salivary parenchyma [1, 194196].

Immunogenic effects

The so-called Marine-Lenhart syndrome [197] is a rare side effect of RAI treatment observed in TNG patients during the first days after RAI therapy, with a reported prevalence ranging from 2.7 to 4.1% [198, 199]. However, its incidence is significantly higher in patients with elevated serum levels of thyroid peroxidase antibody (TPOAb) before RAI therapy, as well as in patients who became TPOAb positive after RAI therapy [198, 200203]. These observations support the hypothesis that immunogenic hyperthyroidism in such patients represents an exacerbation of a pre-existing and occult immunogenic thyroid disorder [1].
RAI administration, as well as surgery, could produce a transient increase of TRAb due to the release of thyroid antigens from damaged follicular cells. As per consequence, autoimmune hyperthyroidism can be observed in such patients, but no correlation has been found between serum TRAb rise and RAI activity administered [1, 204].
The most serious consequence of immunogenic side effects is the activation or new onset of Graves’ orbitopathy (GO) [92].

Late side effects

Hypothyroidism: transient or persistent

Persistent hypothyroidism can be counted among side effects of RAI therapy for TNG or NTNG patients [1, 92]. However, in these patients, thyroid surgery will produce hypothyroidism too [92]. Conversely, hypothyroidism represents the optimal result (i.e., main goal) when RAI therapy is performed in GD patients according to the newest ablative dose concept [1, 6, 9, 52, 76, 77]. However, some patients experience transient hypothyroidism that may manifest 2–5 months after RAI therapy and spontaneously recover within a few months without the development of symptoms or signs of hypothyroidism [1, 205]. Differentiating between transient and permanent hypothyroidism in the early months following RAI therapy remains a clinical conundrum that requires adequate clinical and laboratory follow-up to establish the optimal treatment plan, especially for GD patients with orbitopathy [1, 206, 207].

Graves’ orbitopathy

Several studies demonstrated a significant association between RAI therapy and the development or worsening of GO [1, 208]. In a systematic review that included nine studies comprising a total of 1773 patients, Li et al. reported that RAI therapy is a significant risk for new-onset or worsened orbitopathy as compared to ATD treatment (p < 0.00001), while prophylactic administration of steroids has been demonstrated to be an effective method for preventing this event (p = 0.002), especially in patients with moderate orbitopathy [208]. High TRAb levels before RAI administration, smoking, and uncorrected post-treatment hypothyroidism are associated with a more aggressive orbitopathy and a worse prognosis [70]. For this latter reason, thyroid function should be always evaluated in the first weeks after RAI therapy to avoid the risk of development or worsening of GO due to uncorrected hypothyroidism. However, early (i.e., after 2 weeks following RAI therapy) levothyroxine substitutive treatment has been already proposed for preventing hypothyroidism [207]. In GD patients at high risk for the development/worsening of orbitopathy following RAI therapy, the administration of prophylactic oral steroids is recommended using a daily dose of 0.2 mg prednisone/kg body weight for 6 weeks after RAI therapy [59]. Conversely, intravenous administration of steroids is recommended in GD patients with moderate to severe active GO using a starting dose of 0.5 g once weekly for 6 weeks, followed by 0.25 g once weekly for 6 weeks [59].

Cancer incidence

There is no convincing evidence for an increased risk of secondary cancers and/or increased mortality in patients treated by RAI therapy [209211].

Pregnancy following RAI treatment

Women should be recommended to avoid pregnancy 4–6 months after radioiodine treatment for benign thyroid disease, mainly to ensure to avoid pregnancy in uncontrolled hypothyroidism while radioprotection concerns are limited in such cases.
Specific counseling about pregnancy and fatherhood should be provided to patients planning a pregnancy after RAI therapy. Strict communication between the patients, the referring physician, and the nuclear medicine physician is pivotal to provide the best service.

Acknowledgements

The guidelines were brought to the attention of the relevant EANM Committees and the National Societies of Nuclear Medicine. The comments and suggestions from the EANM [Physics Committee, Dosimetry Committee, Radiation Protection Committee, Radiopharmaceutical Sciences Committee, Oncology & Theranostics Committee, Paediatrics Committee, Technologist Committee] and the Austrian, Slovak, Italian, Dutch, Serbian, Hellenic, Romanian, Turkish, Latvian, Estonian, Polish, Danish, Hungarian, British, French, North Macedonia, and Belgian National Societies are highly appreciated and have been considered for this Guideline.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Liability statement
This guideline summarizes the views of the EANM [Thyroid Committee]. It reflects recommendations for which the EANM cannot be held responsible. The recommendations should be taken into context of good practice of nuclear medicine and do not substitute for national and international legal or regulatory provisions.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Giovanella L, editor. Nuclear medicine therapy: side effects and complications. 1st ed. Nuclear Medicine Therapy: Springer International Publishing; 2019. Giovanella L, editor. Nuclear medicine therapy: side effects and complications. 1st ed. Nuclear Medicine Therapy: Springer International Publishing; 2019.
2.
Zurück zum Zitat Diehl LA, Garcia V, Bonnema SJ, Hegedüs L, Albino CC, Graf H. Management of the nontoxic multinodular goiter in Latin America: comparison with North America and Europe, an electronic survey. J Clin Endocrinol Metab. 2005;90(1):117–23.PubMedCrossRef Diehl LA, Garcia V, Bonnema SJ, Hegedüs L, Albino CC, Graf H. Management of the nontoxic multinodular goiter in Latin America: comparison with North America and Europe, an electronic survey. J Clin Endocrinol Metab. 2005;90(1):117–23.PubMedCrossRef
3.
Zurück zum Zitat Bonnema SJ, Bennedbæk FN, Wiersinga WM, Hegedüs L. Management of the nontoxic multinodular goitre: a European questionnaire study. Clin Endocrinol (Oxf). 2000;53(1):5–12.PubMedCrossRef Bonnema SJ, Bennedbæk FN, Wiersinga WM, Hegedüs L. Management of the nontoxic multinodular goitre: a European questionnaire study. Clin Endocrinol (Oxf). 2000;53(1):5–12.PubMedCrossRef
4.
Zurück zum Zitat Bonnema SJ, Bennedbæk FN, Ladenson PW, Hegedudies L. Management of the nontoxic multinodular goiter: a North American survey. J Clin Endocrinol Metab. 2002;87(1):112–7.PubMedCrossRef Bonnema SJ, Bennedbæk FN, Ladenson PW, Hegedudies L. Management of the nontoxic multinodular goiter: a North American survey. J Clin Endocrinol Metab. 2002;87(1):112–7.PubMedCrossRef
5.
Zurück zum Zitat Bhagat MC, Dhaliwal SS, Bonnema SJ, Hegedüs L, Walsh JP. Differences between endocrine surgeons and endocrinologists in the management of non-toxic multinodular goitre. Br J Surg. 2003;90(9):1103–12.PubMedCrossRef Bhagat MC, Dhaliwal SS, Bonnema SJ, Hegedüs L, Walsh JP. Differences between endocrine surgeons and endocrinologists in the management of non-toxic multinodular goitre. Br J Surg. 2003;90(9):1103–12.PubMedCrossRef
6.
Zurück zum Zitat Stokkel MPM, Handkiewicz Junak D, Lassmann M, Dietlein M, Luster M. EANM procedure guidelines for therapy of benign thyroid disease. Eur J Nucl Med Mol Imaging. 2010;37(11):2218–28.PubMedCrossRef Stokkel MPM, Handkiewicz Junak D, Lassmann M, Dietlein M, Luster M. EANM procedure guidelines for therapy of benign thyroid disease. Eur J Nucl Med Mol Imaging. 2010;37(11):2218–28.PubMedCrossRef
7.
Zurück zum Zitat Taprogge J, Gape PM, Carnegie-Peake L, Murray I, Gear JI, Leek F, et al. A systematic review and meta-analysis of the relationship between the radiation absorbed dose to the thyroid and response in patients treated with radioiodine for Graves’ disease. Thyroid. 2021;31(12):1829–38.PubMedPubMedCentralCrossRef Taprogge J, Gape PM, Carnegie-Peake L, Murray I, Gear JI, Leek F, et al. A systematic review and meta-analysis of the relationship between the radiation absorbed dose to the thyroid and response in patients treated with radioiodine for Graves’ disease. Thyroid. 2021;31(12):1829–38.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Giovanella L, Avram AM, Iakovou I, Kwak J, Lawson SA, Lulaj E, et al. EANM practice guideline/SNMMI procedure standard for RAIU and thyroid scintigraphy. Eur J Nucl Med Mol Imaging. 2019;46(12):2514–25.PubMedCrossRef Giovanella L, Avram AM, Iakovou I, Kwak J, Lawson SA, Lulaj E, et al. EANM practice guideline/SNMMI procedure standard for RAIU and thyroid scintigraphy. Eur J Nucl Med Mol Imaging. 2019;46(12):2514–25.PubMedCrossRef
9.
Zurück zum Zitat Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, et al. 2016 American Thyroid Association Guidelines for diagnosis and management of hyperthyroidism and other causes of thyrotoxicosis. Thyroid. 2016;26(10):1343–421.PubMedCrossRef Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, et al. 2016 American Thyroid Association Guidelines for diagnosis and management of hyperthyroidism and other causes of thyrotoxicosis. Thyroid. 2016;26(10):1343–421.PubMedCrossRef
10.
Zurück zum Zitat Walter MA, Briel M, Christ-Crain M, Bonnema SJ, Connell J, Cooper DS, et al. Effects of antithyroid drugs on radioiodine treatment: systematic review and meta-analysis of randomised controlled trials. BMJ. 2007;334(7592):514.PubMedPubMedCentralCrossRef Walter MA, Briel M, Christ-Crain M, Bonnema SJ, Connell J, Cooper DS, et al. Effects of antithyroid drugs on radioiodine treatment: systematic review and meta-analysis of randomised controlled trials. BMJ. 2007;334(7592):514.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Silberstein EB, Alavi A, Balon HR, Clarke SEM, Divgi C, Gelfand MJ, et al. The SNMMI practice guideline for therapy of thyroid disease with 131I 3.0. J Nucl Med. 2012;53(10):1633–51.PubMedCrossRef Silberstein EB, Alavi A, Balon HR, Clarke SEM, Divgi C, Gelfand MJ, et al. The SNMMI practice guideline for therapy of thyroid disease with 131I 3.0. J Nucl Med. 2012;53(10):1633–51.PubMedCrossRef
13.
Zurück zum Zitat Council Directive 2013/59/Euratom of 5 December 2013 laying down basic safety standards for protection against the dangers arising from exposure to ionising radiation, and repealing Directives 89/618/Euratom, 90/641/Euratom, 96/29/Euratom, 97/43/Euratom a. Off J EU. 2013;L(13):1–73. Council Directive 2013/59/Euratom of 5 December 2013 laying down basic safety standards for protection against the dangers arising from exposure to ionising radiation, and repealing Directives 89/618/Euratom, 90/641/Euratom, 96/29/Euratom, 97/43/Euratom a. Off J EU. 2013;L(13):1–73.
14.
Zurück zum Zitat Hänscheid H, Canzi C, Eschner W, Flux G, Luster M, Strigari M, et al. 2013 EANM Dosimetry committee series on standard operational procedures for pre-therapeutic dosimetry II. Dosimetry prior to radioiodine therapy of benign thyroid diseases. Eur J Nucl Med Mol Imaging. 2013;40(7):1126–34.PubMedCrossRef Hänscheid H, Canzi C, Eschner W, Flux G, Luster M, Strigari M, et al. 2013 EANM Dosimetry committee series on standard operational procedures for pre-therapeutic dosimetry II. Dosimetry prior to radioiodine therapy of benign thyroid diseases. Eur J Nucl Med Mol Imaging. 2013;40(7):1126–34.PubMedCrossRef
15.
Zurück zum Zitat Schlögl S, Werner E, Lassmann M, Terekhova J, Muffert S, Seybold S, et al. The use of three-dimensional ultrasound for thyroid volumetry. Thyroid. 2001;11(6):569–74.PubMedCrossRef Schlögl S, Werner E, Lassmann M, Terekhova J, Muffert S, Seybold S, et al. The use of three-dimensional ultrasound for thyroid volumetry. Thyroid. 2001;11(6):569–74.PubMedCrossRef
16.
Zurück zum Zitat Nygaard B, Nygaard T, Court-Payen M, Ingeman Jensen L, Søe-Jensen P, Gerhard Nielsen K, et al. Thyroid volume measured by ultrasonography and CT. Acta Radiol. 2002;43(3):269–74.PubMedCrossRef Nygaard B, Nygaard T, Court-Payen M, Ingeman Jensen L, Søe-Jensen P, Gerhard Nielsen K, et al. Thyroid volume measured by ultrasonography and CT. Acta Radiol. 2002;43(3):269–74.PubMedCrossRef
17.
Zurück zum Zitat Andermann P, Schlögl S, Mäder U, Luster M, Lassmann M, Reiners C. Intra- and interobserver variability of thyroid volume measurements in healthy adults by 2D versus 3D ultrasound. Nuklearmedizin. 2007;24(1):1–7. Andermann P, Schlögl S, Mäder U, Luster M, Lassmann M, Reiners C. Intra- and interobserver variability of thyroid volume measurements in healthy adults by 2D versus 3D ultrasound. Nuklearmedizin. 2007;24(1):1–7.
18.
Zurück zum Zitat Brunn J, Block U, Ruf G, Bos I, Kunze WP, Scriba PC. Volumetric analysis of thyroid lobes by real-time ultrasound (author’s transl). Dtsch Med Wochenschr. 1981;106(41):1338–40.PubMedCrossRef Brunn J, Block U, Ruf G, Bos I, Kunze WP, Scriba PC. Volumetric analysis of thyroid lobes by real-time ultrasound (author’s transl). Dtsch Med Wochenschr. 1981;106(41):1338–40.PubMedCrossRef
19.
Zurück zum Zitat Park CS, Kim SH, Jung SL, Kang BJ, Kim JY, Choi JJ, et al. Observer variability in the sonographic evaluation of thyroid nodules. J Clin Ultrasound. 2010;38(6):287–93.PubMed Park CS, Kim SH, Jung SL, Kang BJ, Kim JY, Choi JJ, et al. Observer variability in the sonographic evaluation of thyroid nodules. J Clin Ultrasound. 2010;38(6):287–93.PubMed
20.
Zurück zum Zitat Van Isselt JW, De Klerk JMH, Van Rijk PP, Van Gils APG, Polman LJ, Kamphuis C, et al. Comparison of methods for thyroid volume estimation in patients with Graves’ disease. Eur J Nucl Med Mol Imaging. 2003;30(4):525–31.PubMedCrossRef Van Isselt JW, De Klerk JMH, Van Rijk PP, Van Gils APG, Polman LJ, Kamphuis C, et al. Comparison of methods for thyroid volume estimation in patients with Graves’ disease. Eur J Nucl Med Mol Imaging. 2003;30(4):525–31.PubMedCrossRef
21.
Zurück zum Zitat Hermans R, Bouillon R, Laga K, Delaere PR, De Foer B, Marchal G, et al. Estimation of thyroid gland volume by spiral computed tomography. Eur Radiol. 1997;7(2):214–6.PubMedCrossRef Hermans R, Bouillon R, Laga K, Delaere PR, De Foer B, Marchal G, et al. Estimation of thyroid gland volume by spiral computed tomography. Eur Radiol. 1997;7(2):214–6.PubMedCrossRef
22.
Zurück zum Zitat Eschmann SM, Reischl G, Bilger K, Kupferschläger J, Thelen MH, Dohmen BM, et al. Evaluation of dosimetry of radioiodine therapy in benign and malignant thyroid disorders by means of iodine-124 and PET. Eur J Nucl Med Mol Imaging. 2002;29(6):760–7.PubMedCrossRef Eschmann SM, Reischl G, Bilger K, Kupferschläger J, Thelen MH, Dohmen BM, et al. Evaluation of dosimetry of radioiodine therapy in benign and malignant thyroid disorders by means of iodine-124 and PET. Eur J Nucl Med Mol Imaging. 2002;29(6):760–7.PubMedCrossRef
23.
Zurück zum Zitat Hänscheid H, Laßmann M, Reiners C. Dosimetry prior to I-131-therapy of benign thyroid disease. Z Med Phys. 2011;21(4):250–7.PubMedCrossRef Hänscheid H, Laßmann M, Reiners C. Dosimetry prior to I-131-therapy of benign thyroid disease. Z Med Phys. 2011;21(4):250–7.PubMedCrossRef
24.
Zurück zum Zitat Merrill S, Horowitz J, Traino AC, Chipkin SR, Hollot CV, Chait Y. Accuracy and optimal timing of activity measurements in estimating the absorbed dose of radioiodine in the treatment of Graves’ disease. Phys Med Biol. 2011;56(3):557–71.PubMedCrossRef Merrill S, Horowitz J, Traino AC, Chipkin SR, Hollot CV, Chait Y. Accuracy and optimal timing of activity measurements in estimating the absorbed dose of radioiodine in the treatment of Graves’ disease. Phys Med Biol. 2011;56(3):557–71.PubMedCrossRef
25.
Zurück zum Zitat Bockisch A, Jamitzky T, Derwanz R, Biersack HJ. Optimized dose planning of radioiodine therapy of benign thyroidal diseases. J Nucl Med. 1993;34(10):1632–8.PubMed Bockisch A, Jamitzky T, Derwanz R, Biersack HJ. Optimized dose planning of radioiodine therapy of benign thyroidal diseases. J Nucl Med. 1993;34(10):1632–8.PubMed
26.
Zurück zum Zitat Jönsson H, Mattsson S. Single uptake measurement for absorbed dose planning for radioiodine treatment of hyperthyroidism. Cancer Biother Radiopharm. 2003;18(3):473–9.PubMed Jönsson H, Mattsson S. Single uptake measurement for absorbed dose planning for radioiodine treatment of hyperthyroidism. Cancer Biother Radiopharm. 2003;18(3):473–9.PubMed
28.
Zurück zum Zitat Ruggeri R, Giuffrida G, Campennì A. Autoimmune endocrine disease. Minerva Endocrinol. 2018;43(3):305–22.PubMedCrossRef Ruggeri R, Giuffrida G, Campennì A. Autoimmune endocrine disease. Minerva Endocrinol. 2018;43(3):305–22.PubMedCrossRef
29.
Zurück zum Zitat Cooper G, Bynum M, Somers E. Recent insights in the epidemiology of autoimmune diseases: improved prevalence estimates and understanding of clustering of diseases. J Autoimmun. 2009;33(3–4):197–207.PubMedPubMedCentralCrossRef Cooper G, Bynum M, Somers E. Recent insights in the epidemiology of autoimmune diseases: improved prevalence estimates and understanding of clustering of diseases. J Autoimmun. 2009;33(3–4):197–207.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Boelaert K, Newby P, Simmonds M, Holder R, Carr-Smith J, Heward J, et al. Prevalence and relative risk of other autoimmune diseases in subjects with autoimmune thyroid disease. Am J Med. 2010;123(2):183.e1-9.PubMedCrossRef Boelaert K, Newby P, Simmonds M, Holder R, Carr-Smith J, Heward J, et al. Prevalence and relative risk of other autoimmune diseases in subjects with autoimmune thyroid disease. Am J Med. 2010;123(2):183.e1-9.PubMedCrossRef
31.
Zurück zum Zitat Ruggeri R, Trimarchi F, Giuffrida G, Certo R, Cama E, Campennì A, et al. Autoimmune comorbidities in Hashimoto’s thyroiditis: different patterns of association in adulthood and childhood/adolescence. Eur J Endocrinol. 2017;176(2):133–41.PubMedCrossRef Ruggeri R, Trimarchi F, Giuffrida G, Certo R, Cama E, Campennì A, et al. Autoimmune comorbidities in Hashimoto’s thyroiditis: different patterns of association in adulthood and childhood/adolescence. Eur J Endocrinol. 2017;176(2):133–41.PubMedCrossRef
33.
Zurück zum Zitat Laurberg P, Bülow Pedersen I, Knudsen N, Ovesen L, Andersen S. Environmental iodine intake affects the type of non-malignant thyroid disease. Thyroid. 2001;11(5):457–69.PubMedCrossRef Laurberg P, Bülow Pedersen I, Knudsen N, Ovesen L, Andersen S. Environmental iodine intake affects the type of non-malignant thyroid disease. Thyroid. 2001;11(5):457–69.PubMedCrossRef
34.
Zurück zum Zitat Bartalena L, Masiello E, Magri F, Veronesi G, Bianconi E, Zerbini F, et al. The phenotype of newly diagnosed Graves’ disease in Italy in recent years is milder than in the past: results of a large observational longitudinal study. J Endocrinol Invest. 2016;39(12):1445–51.PubMedCrossRef Bartalena L, Masiello E, Magri F, Veronesi G, Bianconi E, Zerbini F, et al. The phenotype of newly diagnosed Graves’ disease in Italy in recent years is milder than in the past: results of a large observational longitudinal study. J Endocrinol Invest. 2016;39(12):1445–51.PubMedCrossRef
35.
Zurück zum Zitat Boelaert K, Torlinska B, Holder R, Franklyn J. Older subjects with hyperthyroidism present with a paucity of symptoms and signs: a large cross-sectional study. J Clin Endocrinol Metab. 2010;95(6):2715–26.PubMedCrossRef Boelaert K, Torlinska B, Holder R, Franklyn J. Older subjects with hyperthyroidism present with a paucity of symptoms and signs: a large cross-sectional study. J Clin Endocrinol Metab. 2010;95(6):2715–26.PubMedCrossRef
36.
Zurück zum Zitat Burch HB. Overview of the clinical manifestations of thyrotoxicosis. In: Braverman LE, Cooper DS, Werner SC, Ingbar SH, editors. Werner & Ingbar’s the thyroid : a fundamental and clinical text. 10th ed. Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins; 2013. p. 434–40. Burch HB. Overview of the clinical manifestations of thyrotoxicosis. In: Braverman LE, Cooper DS, Werner SC, Ingbar SH, editors. Werner & Ingbar’s the thyroid : a fundamental and clinical text. 10th ed. Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins; 2013. p. 434–40.
38.
Zurück zum Zitat Giovanella L, D’Aurizio F, Campenni’ A, Ruggeri R, Baldari S, Verburg F, et al. Searching for the most effective thyrotropin (TSH) threshold to rule-out autonomously functioning thyroid nodules in iodine deficient regions. Endocrine. 2016;54(3):757–61.PubMedCrossRef Giovanella L, D’Aurizio F, Campenni’ A, Ruggeri R, Baldari S, Verburg F, et al. Searching for the most effective thyrotropin (TSH) threshold to rule-out autonomously functioning thyroid nodules in iodine deficient regions. Endocrine. 2016;54(3):757–61.PubMedCrossRef
39.
Zurück zum Zitat Schenke S, Klett R, Seifert P, Kreissl MC, Görges R, Zimny M. Diagnostic performance of different thyroid imaging reporting and data systems (Kwak-TIRADS, EU-TIRADS and ACR TI-RADS) for risk stratification of small thyroid nodules (≤10 mm). J Clin Med. 2020;9(1):236.PubMedPubMedCentralCrossRef Schenke S, Klett R, Seifert P, Kreissl MC, Görges R, Zimny M. Diagnostic performance of different thyroid imaging reporting and data systems (Kwak-TIRADS, EU-TIRADS and ACR TI-RADS) for risk stratification of small thyroid nodules (≤10 mm). J Clin Med. 2020;9(1):236.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat You E, Mascarella MA, Al Jassim A, Forest VI, Hier MP, Tamilia M, et al. Prevalence and aggressiveness of papillary thyroid carcinoma in surgically-treated graves’ disease patients: a retrospective matched cohort study. J Otolaryngol - Head Neck Surg. 2019;48(1):40.PubMedPubMedCentralCrossRef You E, Mascarella MA, Al Jassim A, Forest VI, Hier MP, Tamilia M, et al. Prevalence and aggressiveness of papillary thyroid carcinoma in surgically-treated graves’ disease patients: a retrospective matched cohort study. J Otolaryngol - Head Neck Surg. 2019;48(1):40.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Boutzios G, Vasileiadis I, Zapanti E, Charitoudis G, Karakostas E, Ieromonachou P, et al. Higher incidence of tall cell variant of papillary thyroid carcinoma in Graves’ disease. Thyroid. 2014;24(2):347–54.PubMedCrossRef Boutzios G, Vasileiadis I, Zapanti E, Charitoudis G, Karakostas E, Ieromonachou P, et al. Higher incidence of tall cell variant of papillary thyroid carcinoma in Graves’ disease. Thyroid. 2014;24(2):347–54.PubMedCrossRef
42.
Zurück zum Zitat Campennì A, Barbaro D, Guzzo M, Capoccetti F, Giovanella L. Personalized management of differentiated thyroid cancer in real life - practical guidance from a multidisciplinary panel of experts. Endocrine. 2020;70(2):280–91.PubMedPubMedCentralCrossRef Campennì A, Barbaro D, Guzzo M, Capoccetti F, Giovanella L. Personalized management of differentiated thyroid cancer in real life - practical guidance from a multidisciplinary panel of experts. Endocrine. 2020;70(2):280–91.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Moleti M, Violi MA, Montanini D, Trombetta C, Di Bella B, Sturniolo G, et al. Radioiodine ablation of postsurgical thyroid remnants after treatment with recombinant human TSH (rhTSH) in patients with moderate-to-severe Graves’ orbitopathy (GO): a prospective, randomized, single-blind clinical trial. J Clin Endocrinol Metab. 2014;99(5):1783–9.PubMedCrossRef Moleti M, Violi MA, Montanini D, Trombetta C, Di Bella B, Sturniolo G, et al. Radioiodine ablation of postsurgical thyroid remnants after treatment with recombinant human TSH (rhTSH) in patients with moderate-to-severe Graves’ orbitopathy (GO): a prospective, randomized, single-blind clinical trial. J Clin Endocrinol Metab. 2014;99(5):1783–9.PubMedCrossRef
44.
Zurück zum Zitat Menconi F, Marinò M, Pinchera A, Rocchi R, Mazzi B, Nardi M, et al. Effects of total thyroid ablation versus near-total thyroidectomy alone on mild to moderate Graves’ orbitopathy treated with intravenous glucocorticoids. J Clin Endocrinol Metab. 2007;92(5):1653–8.PubMedCrossRef Menconi F, Marinò M, Pinchera A, Rocchi R, Mazzi B, Nardi M, et al. Effects of total thyroid ablation versus near-total thyroidectomy alone on mild to moderate Graves’ orbitopathy treated with intravenous glucocorticoids. J Clin Endocrinol Metab. 2007;92(5):1653–8.PubMedCrossRef
45.
Zurück zum Zitat McDermott M, Kidd G, Dodson LJ, Hofeldt F. Radioiodine-induced thyroid storm. Case report and literature review. Am J Med. 1983;75(2):353–9.PubMedCrossRef McDermott M, Kidd G, Dodson LJ, Hofeldt F. Radioiodine-induced thyroid storm. Case report and literature review. Am J Med. 1983;75(2):353–9.PubMedCrossRef
46.
Zurück zum Zitat Akamizu T, Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, et al. Diagnostic criteria, clinical features, and incidence of thyroid storm based on nationwide surveys. Thyroid. 2012;22(7):661–79.PubMedPubMedCentralCrossRef Akamizu T, Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, et al. Diagnostic criteria, clinical features, and incidence of thyroid storm based on nationwide surveys. Thyroid. 2012;22(7):661–79.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Moka D, Dietlein M, Schicha H. Radioiodine therapy and thyrostatic drugs and iodine. Eur J Nucl Med Mol Imaging. 2002;29(Suppl 2):S486–91.PubMedCrossRef Moka D, Dietlein M, Schicha H. Radioiodine therapy and thyrostatic drugs and iodine. Eur J Nucl Med Mol Imaging. 2002;29(Suppl 2):S486–91.PubMedCrossRef
48.
Zurück zum Zitat Santos RB, Romaldini JH, Ward LS. Propylthiouracil reduces the effectiveness of radioiodine treatment in hyperthyroid patients with Graves’ disease. Thyroid. 2004;14(7):525–30.PubMedCrossRef Santos RB, Romaldini JH, Ward LS. Propylthiouracil reduces the effectiveness of radioiodine treatment in hyperthyroid patients with Graves’ disease. Thyroid. 2004;14(7):525–30.PubMedCrossRef
49.
Zurück zum Zitat Bonnema SJ, Bennedbæk FN, Veje A, Marving J, Hegedüs L. Propylthiouracil before 131I therapy of hyperthyroid diseases: effect on cure rate evaluated by a randomized clinical trial. J Clin Endocrinol Metab. 2004;89(9):4439–44.PubMedCrossRef Bonnema SJ, Bennedbæk FN, Veje A, Marving J, Hegedüs L. Propylthiouracil before 131I therapy of hyperthyroid diseases: effect on cure rate evaluated by a randomized clinical trial. J Clin Endocrinol Metab. 2004;89(9):4439–44.PubMedCrossRef
50.
Zurück zum Zitat Braga M, Walpert N, Burch HB, Solomon BL, Cooper DS. The effect of methimazole on cure rates after radioiodine treatment for Graves’ hyperthyroidism: a randomized clinical trial. Thyroid. 2002;12(2):135–9.PubMedCrossRef Braga M, Walpert N, Burch HB, Solomon BL, Cooper DS. The effect of methimazole on cure rates after radioiodine treatment for Graves’ hyperthyroidism: a randomized clinical trial. Thyroid. 2002;12(2):135–9.PubMedCrossRef
51.
Zurück zum Zitat Hancock LD, Tuttle RM, LeMar H, Bauman J, Patience T. The effect of propylthiouracil on subsequent radioactive iodine therapy in Graves’ disease. Clin Endocrinol (Oxf). 1997;47(4):425–30.PubMedCrossRef Hancock LD, Tuttle RM, LeMar H, Bauman J, Patience T. The effect of propylthiouracil on subsequent radioactive iodine therapy in Graves’ disease. Clin Endocrinol (Oxf). 1997;47(4):425–30.PubMedCrossRef
52.
Zurück zum Zitat Kobe C, Weber I, Eschner W, Sudbrock F, Schmidt M, Dietlein M, et al. Graves’ disease and radioiodine therapy. Is success of ablation dependent on the choice of thyreostatic medication? Nuklearmedizin. 2008;47(4):153–6.PubMedCrossRef Kobe C, Weber I, Eschner W, Sudbrock F, Schmidt M, Dietlein M, et al. Graves’ disease and radioiodine therapy. Is success of ablation dependent on the choice of thyreostatic medication? Nuklearmedizin. 2008;47(4):153–6.PubMedCrossRef
53.
Zurück zum Zitat Shafe R, Nuttall F. Acute changes in thyroid function in patients treated with radioactive iodine. Lancet. 1975;2(7936):635–7.CrossRef Shafe R, Nuttall F. Acute changes in thyroid function in patients treated with radioactive iodine. Lancet. 1975;2(7936):635–7.CrossRef
54.
Zurück zum Zitat Burch HB, Solomon BL, Cooper DS, Ferguson P, Walpert N, Howard R. The effect of antithyroid drug pretreatment on acute changes in thyroid hormone levels after 131I ablation for Graves’ disease. J Clin Endocrinol Metab. 2001;86(7):3016–21.PubMed Burch HB, Solomon BL, Cooper DS, Ferguson P, Walpert N, Howard R. The effect of antithyroid drug pretreatment on acute changes in thyroid hormone levels after 131I ablation for Graves’ disease. J Clin Endocrinol Metab. 2001;86(7):3016–21.PubMed
55.
Zurück zum Zitat Andrade V, Gross J, Maia A. Effect of methimazole pretreatment on serum thyroid hormone levels after radioactive treatment in Graves’ hyperthyroidism. J Clin Endocrinol Metab. 1999;84(11):4012–6.PubMed Andrade V, Gross J, Maia A. Effect of methimazole pretreatment on serum thyroid hormone levels after radioactive treatment in Graves’ hyperthyroidism. J Clin Endocrinol Metab. 1999;84(11):4012–6.PubMed
56.
57.
Zurück zum Zitat Klein I, et al. Endocrine disorders and cardiovascular disease. In: Libby P, et al., editors. Braunwald’s heart disease: a textbook of cardiovascular medicine. 8th ed. Philadelphia: Saunders/Elsevier; 2008. p. 2033–47. Klein I, et al. Endocrine disorders and cardiovascular disease. In: Libby P, et al., editors. Braunwald’s heart disease: a textbook of cardiovascular medicine. 8th ed. Philadelphia: Saunders/Elsevier; 2008. p. 2033–47.
58.
Zurück zum Zitat Bartalena L, Kahaly GJ, Baldeschi L, Dayan CM, Eckstein A, Marcocci C, et al. The 2021 European group on Graves’ orbitopathy (EUGOGO) clinical practice guidelines for the medical management of Graves’ orbitopathy. Eur J Endocrinol. 2021;185(4):G43-67.PubMedCrossRef Bartalena L, Kahaly GJ, Baldeschi L, Dayan CM, Eckstein A, Marcocci C, et al. The 2021 European group on Graves’ orbitopathy (EUGOGO) clinical practice guidelines for the medical management of Graves’ orbitopathy. Eur J Endocrinol. 2021;185(4):G43-67.PubMedCrossRef
59.
Zurück zum Zitat Bartalena L, Baldeschi L, Boboridis K, Eckstein A, Kahaly GJ, Marcocci C, et al. The 2016 European Thyroid Association/European Group on Graves’ orbitopathy guidelines for the management of Graves’ orbitopathy. Eur Thyroid J. 2016;5(1):9–26.PubMedPubMedCentralCrossRef Bartalena L, Baldeschi L, Boboridis K, Eckstein A, Kahaly GJ, Marcocci C, et al. The 2016 European Thyroid Association/European Group on Graves’ orbitopathy guidelines for the management of Graves’ orbitopathy. Eur Thyroid J. 2016;5(1):9–26.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Dederichs B, Dietlein M, Jenniches-Kloth B, Schmidt M, Theissen P, Moka D, et al. Radioiodine therapy of Graves’ hyperthyroidism in patients without pre-existing ophthalmopathy: can glucocorticoids prevent the development of new ophthalmopathy? Exp Clin Endocrinol Diabetes. 2006;114(7):366–70.PubMedCrossRef Dederichs B, Dietlein M, Jenniches-Kloth B, Schmidt M, Theissen P, Moka D, et al. Radioiodine therapy of Graves’ hyperthyroidism in patients without pre-existing ophthalmopathy: can glucocorticoids prevent the development of new ophthalmopathy? Exp Clin Endocrinol Diabetes. 2006;114(7):366–70.PubMedCrossRef
61.
Zurück zum Zitat Halstenberg J, Kranert WT, Korkusuz H, Mayer A, Ackermann H, Grünwald F, et al. Influence of glucocorticoid therapy on intratherapeutic biodistribution of 131I radioiodine therapy in Graves’ disease. Nuklearmedizin. 2018;57(2):43–9.PubMed Halstenberg J, Kranert WT, Korkusuz H, Mayer A, Ackermann H, Grünwald F, et al. Influence of glucocorticoid therapy on intratherapeutic biodistribution of 131I radioiodine therapy in Graves’ disease. Nuklearmedizin. 2018;57(2):43–9.PubMed
62.
Zurück zum Zitat Sekulić V, Rajić M, Vlajković M, Ilić S, Stević M, Kojić M. The effect of short-term treatment with lithium carbonate on the outcome of radioiodine therapy in patients with long-lasting Graves’ hyperthyroidism. Ann Nucl Med. 2017;31(10):744–51.PubMedCrossRef Sekulić V, Rajić M, Vlajković M, Ilić S, Stević M, Kojić M. The effect of short-term treatment with lithium carbonate on the outcome of radioiodine therapy in patients with long-lasting Graves’ hyperthyroidism. Ann Nucl Med. 2017;31(10):744–51.PubMedCrossRef
63.
Zurück zum Zitat Sekulić V, Rajić MP, Vlajković MŽ, Ilić SS, Stević ML, Kojić MM. Short term treatment with lithium carbonate as adjunct to radioiodine treatment for long-lasting Graves’ hyperthyroidism. Hell J Nucl Med. 2015;18(3):186–8.PubMed Sekulić V, Rajić MP, Vlajković MŽ, Ilić SS, Stević ML, Kojić MM. Short term treatment with lithium carbonate as adjunct to radioiodine treatment for long-lasting Graves’ hyperthyroidism. Hell J Nucl Med. 2015;18(3):186–8.PubMed
64.
Zurück zum Zitat International Commission on Radiological Protection. Release of patients after therapy with unsealed radionuclides. Ann ICRP. 2004;34(2):v–vi, 179. https://doi.org/10.1016/j.icrp.2004.08.001. Erratum in: Ann ICRP. 2004;34(3-4):281. Erratum in: Ann ICRP. 2006;36(4):77. Erratum in: Ann ICRP. 2013 Aug;42(4):341. International Commission on Radiological Protection. Release of patients after therapy with unsealed radionuclides. Ann ICRP. 2004;34(2):v–vi, 179. https://​doi.​org/​10.​1016/​j.​icrp.​2004.​08.​001. Erratum in: Ann ICRP. 2004;34(3-4):281. Erratum in: Ann ICRP. 2006;36(4):77. Erratum in: Ann ICRP. 2013 Aug;42(4):341.
65.
Zurück zum Zitat Brzozowska M, Roach PJ. Timing and potential role of diagnostic I-123 scintigraphy in assessing radioiodine breast uptake before ablation in postpartum women with thyroid cancer: a case series. Clin Nucl Med. 2006;31(11):683–7.PubMedCrossRef Brzozowska M, Roach PJ. Timing and potential role of diagnostic I-123 scintigraphy in assessing radioiodine breast uptake before ablation in postpartum women with thyroid cancer: a case series. Clin Nucl Med. 2006;31(11):683–7.PubMedCrossRef
66.
Zurück zum Zitat Cappelen T, Unhjem JF, Amundsen AL, Kravdal G, Følling I. Radiation exposure to family members of patients with thyrotoxicosis treated with iodine-131. Eur J Nucl Med Mol Imaging. 2006;33(1):81–6.PubMedCrossRef Cappelen T, Unhjem JF, Amundsen AL, Kravdal G, Følling I. Radiation exposure to family members of patients with thyrotoxicosis treated with iodine-131. Eur J Nucl Med Mol Imaging. 2006;33(1):81–6.PubMedCrossRef
67.
Zurück zum Zitat Koch W, Rosa F, Knesewitsch P, Hahn K. Guideline on radiation protection in medicine requires documentation of radioiodine therapy and follow-up: what are the benefits of an electronic database? Nuklearmedizin. 2005;44(2):49–55.PubMedCrossRef Koch W, Rosa F, Knesewitsch P, Hahn K. Guideline on radiation protection in medicine requires documentation of radioiodine therapy and follow-up: what are the benefits of an electronic database? Nuklearmedizin. 2005;44(2):49–55.PubMedCrossRef
68.
Zurück zum Zitat Dillehay GL, Ellerbroek NA, Balon H, Brill DR, Grigsby PW, MacKlis RM, et al. Practice guideline for the performance of therapy with unsealed radiopharmaceutical sources. Int J Radiat Oncol Biol Phys. 2006;64(5):1299–307.PubMedCrossRef Dillehay GL, Ellerbroek NA, Balon H, Brill DR, Grigsby PW, MacKlis RM, et al. Practice guideline for the performance of therapy with unsealed radiopharmaceutical sources. Int J Radiat Oncol Biol Phys. 2006;64(5):1299–307.PubMedCrossRef
69.
Zurück zum Zitat Bartalena L, Chiovato L, Vitti P. Management of hyperthyroidism due to Graves’ disease: frequently asked questions and answers (if any). J Endocrinol Invest. 2016;39(10):1105–14.PubMedCrossRef Bartalena L, Chiovato L, Vitti P. Management of hyperthyroidism due to Graves’ disease: frequently asked questions and answers (if any). J Endocrinol Invest. 2016;39(10):1105–14.PubMedCrossRef
70.
Zurück zum Zitat Bahn RS, Burch HB, Cooper DS, Garber JR, Greenlee MC, Klein I, et al. Hyperthyroidism and other causes of thyrotoxicosis: management guidelines of the American Thyroid Association and American Association of Clinical Endocrinologists. Thyroid. 2011;21(6):593–646.CrossRef Bahn RS, Burch HB, Cooper DS, Garber JR, Greenlee MC, Klein I, et al. Hyperthyroidism and other causes of thyrotoxicosis: management guidelines of the American Thyroid Association and American Association of Clinical Endocrinologists. Thyroid. 2011;21(6):593–646.CrossRef
71.
Zurück zum Zitat Träisk F, Tallstedt L, Abraham-Nordling M, Andersson T, Berg G, Calissendorff J, et al. Thyroid-associated ophthalmopathy after treatment for Graves’ hyperthyroidism with antithyroid drugs or iodine-131. J Clin Endocrinol Metab. 2009;94(10):3700–7.PubMedCrossRef Träisk F, Tallstedt L, Abraham-Nordling M, Andersson T, Berg G, Calissendorff J, et al. Thyroid-associated ophthalmopathy after treatment for Graves’ hyperthyroidism with antithyroid drugs or iodine-131. J Clin Endocrinol Metab. 2009;94(10):3700–7.PubMedCrossRef
72.
Zurück zum Zitat Eckstein AK, Plicht M, Lax H, Neuhäuser M, Mann K, Lederbogen S, et al. Thyrotropin receptor autoantibodies are independent risk factors for Graves’ ophthalmopathy and help to predict severity and outcome of the disease. J Clin Endocrinol Metab. 2006;91(9):3464–70.PubMedCrossRef Eckstein AK, Plicht M, Lax H, Neuhäuser M, Mann K, Lederbogen S, et al. Thyrotropin receptor autoantibodies are independent risk factors for Graves’ ophthalmopathy and help to predict severity and outcome of the disease. J Clin Endocrinol Metab. 2006;91(9):3464–70.PubMedCrossRef
73.
74.
Zurück zum Zitat Ron E, Doody MM, Becker DV, Brill AB, Curtis RE, Goldman MB, et al. Cancer mortality following treatment for adult hyperthyroidism. Cooperative Thyrotoxicosis Therapy Follow-up Study Group. JAMA. 1998;280(4):347–55.PubMedCrossRef Ron E, Doody MM, Becker DV, Brill AB, Curtis RE, Goldman MB, et al. Cancer mortality following treatment for adult hyperthyroidism. Cooperative Thyrotoxicosis Therapy Follow-up Study Group. JAMA. 1998;280(4):347–55.PubMedCrossRef
75.
Zurück zum Zitat De Jong J, Da Huysmans C, de Keizer B. 131I Therapy in primary hyperthyroidism and non-toxic (multi)nodular goitre. Dutch medical guidelines database. 2016. De Jong J, Da Huysmans C, de Keizer B. 131I Therapy in primary hyperthyroidism and non-toxic (multi)nodular goitre. Dutch medical guidelines database. 2016.
76.
Zurück zum Zitat Reinhardt M, Brink I, Joe A, Von Mallek D, Ezziddin S, Palmedo H, et al. Radioiodine therapy in Graves’ disease based on tissue-absorbed dose calculations: effect of pre-treatment thyroid volume on clinical outcome. Eur J Nucl Med. 2002;29(9):1118–24.CrossRef Reinhardt M, Brink I, Joe A, Von Mallek D, Ezziddin S, Palmedo H, et al. Radioiodine therapy in Graves’ disease based on tissue-absorbed dose calculations: effect of pre-treatment thyroid volume on clinical outcome. Eur J Nucl Med. 2002;29(9):1118–24.CrossRef
77.
Zurück zum Zitat Dunkelmann S, Neumann V, Staub U, Groth P, Kuenstner H, Schuemichen C. Results of a risk adapted and functional radioiodine therapy in Graves’ disease. Nuklearmedizin. 2005;44(6):238–42.PubMedCrossRef Dunkelmann S, Neumann V, Staub U, Groth P, Kuenstner H, Schuemichen C. Results of a risk adapted and functional radioiodine therapy in Graves’ disease. Nuklearmedizin. 2005;44(6):238–42.PubMedCrossRef
78.
Zurück zum Zitat Tarantini B, Ciuoli C, Di Cairano G, Guarino E, Mazzucato P, Montanaro A, et al. Effectiveness of radioiodine (131-I) as definitive therapy in patients with autoimmune and non-autoimmune hyperthyroidism. J Endocrinol Invest. 2006;29(7):594–8.PubMedCrossRef Tarantini B, Ciuoli C, Di Cairano G, Guarino E, Mazzucato P, Montanaro A, et al. Effectiveness of radioiodine (131-I) as definitive therapy in patients with autoimmune and non-autoimmune hyperthyroidism. J Endocrinol Invest. 2006;29(7):594–8.PubMedCrossRef
79.
Zurück zum Zitat Kung AWC, Yau CC, Cheng ACK. The action of methimazole and L-thyroxine in radioiodine therapy: a prospective study on the incidence of hypothyroidism. Thyroid. 1995;5(1):7–12.PubMedCrossRef Kung AWC, Yau CC, Cheng ACK. The action of methimazole and L-thyroxine in radioiodine therapy: a prospective study on the incidence of hypothyroidism. Thyroid. 1995;5(1):7–12.PubMedCrossRef
80.
Zurück zum Zitat Santos RB, Romaldini JH, Ward LS. A randomized controlled trial to evaluate the effectiveness of 2 regimens of fixed iodine (131I) doses for Graves disease treatment. Clin Nucl Med. 2012;37(3):241–4.PubMedCrossRef Santos RB, Romaldini JH, Ward LS. A randomized controlled trial to evaluate the effectiveness of 2 regimens of fixed iodine (131I) doses for Graves disease treatment. Clin Nucl Med. 2012;37(3):241–4.PubMedCrossRef
81.
Zurück zum Zitat Willegaignon J, Sapienza M, Buchpiguel C. Radioiodine therapy for Graves disease: thyroid absorbed dose of 300 Gy-tuning the target for therapy planning. Clin Nucl Med. 2013;38(4):231–6.PubMedCrossRef Willegaignon J, Sapienza M, Buchpiguel C. Radioiodine therapy for Graves disease: thyroid absorbed dose of 300 Gy-tuning the target for therapy planning. Clin Nucl Med. 2013;38(4):231–6.PubMedCrossRef
82.
Zurück zum Zitat Mooij CF, Cheetham TD, Verburg FA, Eckstein A, Pearce SH, Léger J, van Trotsenburg ASP. 2022 European Thyroid Association Guideline for the management of pediatric Graves' disease. Eur Thyroid J. 2022;11(1):e210073. https://doi.org/10.1530/ETJ-21-0073. Mooij CF, Cheetham TD, Verburg FA, Eckstein A, Pearce SH, Léger J, van Trotsenburg ASP. 2022 European Thyroid Association Guideline for the management of pediatric Graves' disease. Eur Thyroid J. 2022;11(1):e210073. https://​doi.​org/​10.​1530/​ETJ-21-0073.
83.
Zurück zum Zitat Rivkees SA. Controversies in the management of Graves’ disease in children. J Endocrinol Invest. 2016;39(11):1247–57.PubMedCrossRef Rivkees SA. Controversies in the management of Graves’ disease in children. J Endocrinol Invest. 2016;39(11):1247–57.PubMedCrossRef
84.
Zurück zum Zitat Dobyns BM, Sheline GE, Workman JB, Tompkins EA, McConahey WM, Becker DV. Malignant and benign neoplasms of the thyroid in patients treated for hyperthyroidism: a report of the cooperative thyrotoxicosis therapy follow-up study. J Clin Endocrinol Metab. 1974;38(6):976–98.PubMedCrossRef Dobyns BM, Sheline GE, Workman JB, Tompkins EA, McConahey WM, Becker DV. Malignant and benign neoplasms of the thyroid in patients treated for hyperthyroidism: a report of the cooperative thyrotoxicosis therapy follow-up study. J Clin Endocrinol Metab. 1974;38(6):976–98.PubMedCrossRef
85.
Zurück zum Zitat Sheline GE, Lindsay S, McCormack KR, Galante M. Thyroid nodules occurring late after treatment of thyrotoxicosis with radioiodine. J Clin Endocrinol Metab. 1962;22:8–18.PubMedCrossRef Sheline GE, Lindsay S, McCormack KR, Galante M. Thyroid nodules occurring late after treatment of thyrotoxicosis with radioiodine. J Clin Endocrinol Metab. 1962;22:8–18.PubMedCrossRef
86.
Zurück zum Zitat Rivkees SA, Dinauer C. An optimal treatment for pediatric Graves’ disease is radioiodine. J Clin Endocrinol Metab. 2007;92(3):797–800.PubMedCrossRef Rivkees SA, Dinauer C. An optimal treatment for pediatric Graves’ disease is radioiodine. J Clin Endocrinol Metab. 2007;92(3):797–800.PubMedCrossRef
87.
Zurück zum Zitat Chao M, Jiawei X, Guoming W, Jianbin L, Wanxia L, Driedger A, et al. Radioiodine treatment for pediatric hyperthyroid Grave’s disease. Eur J Pediatr. 2009;168(10):1165–9.PubMedCrossRef Chao M, Jiawei X, Guoming W, Jianbin L, Wanxia L, Driedger A, et al. Radioiodine treatment for pediatric hyperthyroid Grave’s disease. Eur J Pediatr. 2009;168(10):1165–9.PubMedCrossRef
88.
Zurück zum Zitat Arora S, Bal C. Is there any need for adjusting 131 I activity for the treatment of high turnover Graves’ disease compared to normal turnover patients? Results from a retrospective cohort study validated by propensity score analysis. Nucl Med Mol Imaging (2010). 2021;55(1):15–26.CrossRef Arora S, Bal C. Is there any need for adjusting 131 I activity for the treatment of high turnover Graves’ disease compared to normal turnover patients? Results from a retrospective cohort study validated by propensity score analysis. Nucl Med Mol Imaging (2010). 2021;55(1):15–26.CrossRef
89.
Zurück zum Zitat Vija Racaru L, Fontan C, Bauriaud-Mallet M, Brillouet S, Caselles O, Zerdoud S, et al. Clinical outcomes 1 year after empiric 131I therapy for hyperthyroid disorders: real life experience and predictive factors of functional response. Nucl Med Commun. 2017;38(9):756–63.PubMedCrossRef Vija Racaru L, Fontan C, Bauriaud-Mallet M, Brillouet S, Caselles O, Zerdoud S, et al. Clinical outcomes 1 year after empiric 131I therapy for hyperthyroid disorders: real life experience and predictive factors of functional response. Nucl Med Commun. 2017;38(9):756–63.PubMedCrossRef
90.
Zurück zum Zitat Metso S, Jaatinen P, Huhtala H, Luukkaala T, Oksala H, Salmi J. Long-term follow-up study of radioiodine treatment of hyperthyroidism. Clin Endocrinol (Oxf). 2004;61(5):641–8.PubMedCrossRef Metso S, Jaatinen P, Huhtala H, Luukkaala T, Oksala H, Salmi J. Long-term follow-up study of radioiodine treatment of hyperthyroidism. Clin Endocrinol (Oxf). 2004;61(5):641–8.PubMedCrossRef
91.
Zurück zum Zitat Allahabadia A, Daykin J, Sheppard MC, Gough SCL, Franklyn JA. Radioiodine treatment of hyperthyroidism-prognostic factors for outcome. J Clin Endocrinol Metab. 2001;86(8):3611–7.PubMed Allahabadia A, Daykin J, Sheppard MC, Gough SCL, Franklyn JA. Radioiodine treatment of hyperthyroidism-prognostic factors for outcome. J Clin Endocrinol Metab. 2001;86(8):3611–7.PubMed
92.
Zurück zum Zitat Bonnema SJ, Hegedüs L. Radioiodine therapy in benign thyroid diseases: effects, side effects, and factors affecting therapeutic outcome. Endocr Rev. 2012;33(6):920–80.PubMedCrossRef Bonnema SJ, Hegedüs L. Radioiodine therapy in benign thyroid diseases: effects, side effects, and factors affecting therapeutic outcome. Endocr Rev. 2012;33(6):920–80.PubMedCrossRef
93.
Zurück zum Zitat Peters H, Fischer C, Bogner U, Reiners C, Schleusener H. Radioiodine therapy of Graves’ hyperthyroidism: standard vs. calculated 131iodine activity. Results from a prospective, randomized, multicentre study. Eur J Clin Invest. 1995;25(3):186–93.PubMedCrossRef Peters H, Fischer C, Bogner U, Reiners C, Schleusener H. Radioiodine therapy of Graves’ hyperthyroidism: standard vs. calculated 131iodine activity. Results from a prospective, randomized, multicentre study. Eur J Clin Invest. 1995;25(3):186–93.PubMedCrossRef
94.
Zurück zum Zitat De Jong JAF, Verkooijen HM, Valk GD, Zelissen PMJ, De Keizer B. High failure rates after (131)I therapy in Graves hyperthyroidism patients with large thyroid volumes, high iodine uptake, and high iodine turnover. Clin Nucl Med. 2013;38(6):401–6.PubMedCrossRef De Jong JAF, Verkooijen HM, Valk GD, Zelissen PMJ, De Keizer B. High failure rates after (131)I therapy in Graves hyperthyroidism patients with large thyroid volumes, high iodine uptake, and high iodine turnover. Clin Nucl Med. 2013;38(6):401–6.PubMedCrossRef
95.
Zurück zum Zitat Aghini-Lombardi F, Antonangeli L, Martino E, Vitti P, Maccherini D, Leoli F, et al. The spectrum of thyroid disorders in an iodine-deficient community: the Pescopagano survey. J Clin Endocrinol Metab. 1999;84(2):561–6.PubMed Aghini-Lombardi F, Antonangeli L, Martino E, Vitti P, Maccherini D, Leoli F, et al. The spectrum of thyroid disorders in an iodine-deficient community: the Pescopagano survey. J Clin Endocrinol Metab. 1999;84(2):561–6.PubMed
96.
Zurück zum Zitat Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr Rev. 2008;29(1):76–131.PubMedCrossRef Biondi B, Cooper DS. The clinical significance of subclinical thyroid dysfunction. Endocr Rev. 2008;29(1):76–131.PubMedCrossRef
97.
Zurück zum Zitat Ruggeri R, Campennì A, Sindoni A, Baldari S, Trimarchi F, Benvenga S. Association of autonomously functioning thyroid nodules with Hashimoto’s thyroiditis: study on a large series of patients. Exp Clin Endocrinol Diabetes. 2011;119(10):621–7.PubMedCrossRef Ruggeri R, Campennì A, Sindoni A, Baldari S, Trimarchi F, Benvenga S. Association of autonomously functioning thyroid nodules with Hashimoto’s thyroiditis: study on a large series of patients. Exp Clin Endocrinol Diabetes. 2011;119(10):621–7.PubMedCrossRef
98.
Zurück zum Zitat Lau LW, Ghaznavi S, Frolkis AD, Stephenson A, Robertson HL, Rabi DM, et al. Malignancy risk of hyperfunctioning thyroid nodules compared with non-toxic nodules: systematic review and a meta-analysis. Thyroid Res. 2021;14(1):3.PubMedPubMedCentralCrossRef Lau LW, Ghaznavi S, Frolkis AD, Stephenson A, Robertson HL, Rabi DM, et al. Malignancy risk of hyperfunctioning thyroid nodules compared with non-toxic nodules: systematic review and a meta-analysis. Thyroid Res. 2021;14(1):3.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Corvilain B. The natural history of thyroid autonomy and hot nodules. Ann Endocrinol. 2003;64(1):17–22. Corvilain B. The natural history of thyroid autonomy and hot nodules. Ann Endocrinol. 2003;64(1):17–22.
100.
Zurück zum Zitat Moreno-Reyes R, Kyrilli A, Lytrivi M, Bourmorck C, Chami R, Corvilain B. Is there still a role for thyroid scintigraphy in the workup of a thyroid nodule in the era of fine needle aspiration cytology and molecular testing? F1000Research. 2016;5:F1000 Faculty Rev-763. Moreno-Reyes R, Kyrilli A, Lytrivi M, Bourmorck C, Chami R, Corvilain B. Is there still a role for thyroid scintigraphy in the workup of a thyroid nodule in the era of fine needle aspiration cytology and molecular testing? F1000Research. 2016;5:F1000 Faculty Rev-763.
101.
Zurück zum Zitat Sandrock D, Olbricht T, Emrich D, Benker G, Reinwein D. Long-term follow-up in patients with autonomous thyroid adenoma. Acta Endocrinol (Copenh). 1993;128(1):51–5.PubMed Sandrock D, Olbricht T, Emrich D, Benker G, Reinwein D. Long-term follow-up in patients with autonomous thyroid adenoma. Acta Endocrinol (Copenh). 1993;128(1):51–5.PubMed
102.
Zurück zum Zitat Gharib H, Papini E, Garber JR, Duick DS, Harrell RM, Hegedüs L, et al. American Association of Clinical Endocrinologists, American College of Endocrinology, and Associazione Medici Endocrinologi Medical Guidelines for Clinical Practice for the Diagnosis and Management of Thyroid Nodules - 2016 Update Appendix. Endocr Pract. 2016;22(1):1–60.CrossRef Gharib H, Papini E, Garber JR, Duick DS, Harrell RM, Hegedüs L, et al. American Association of Clinical Endocrinologists, American College of Endocrinology, and Associazione Medici Endocrinologi Medical Guidelines for Clinical Practice for the Diagnosis and Management of Thyroid Nodules - 2016 Update Appendix. Endocr Pract. 2016;22(1):1–60.CrossRef
103.
Zurück zum Zitat Vicchio TM, Giovinazzo S, Certo R, Cucinotta M, Micali C, Baldari S, et al. Lack of association between autonomously functioning thyroid nodules and germline polymorphisms of the thyrotropin receptor and Gαs genes in a mild to moderate iodine-deficient Caucasian population. J Endocrinol Invest. 2014;37(7):625–30.PubMedCrossRef Vicchio TM, Giovinazzo S, Certo R, Cucinotta M, Micali C, Baldari S, et al. Lack of association between autonomously functioning thyroid nodules and germline polymorphisms of the thyrotropin receptor and Gαs genes in a mild to moderate iodine-deficient Caucasian population. J Endocrinol Invest. 2014;37(7):625–30.PubMedCrossRef
104.
Zurück zum Zitat Tonacchera M, Chiovato L, Pinchera A, Agretti P, Fiore E, Cetani F, et al. Hyperfunctioning thyroid nodules in toxic multinodular goiter share activating thyrotropin receptor mutations with solitary toxic adenoma. J Clin Endocrinol Metab. 1998;83(2):492–8.PubMed Tonacchera M, Chiovato L, Pinchera A, Agretti P, Fiore E, Cetani F, et al. Hyperfunctioning thyroid nodules in toxic multinodular goiter share activating thyrotropin receptor mutations with solitary toxic adenoma. J Clin Endocrinol Metab. 1998;83(2):492–8.PubMed
105.
Zurück zum Zitat Gozu HI, Lublinghoff J, Bircan R, Paschke R. Genetics and phenomics of inherited and sporadic non-autoimmune hyperthyroidism. Mol Cell Endocrinol. 2010;322(1–2):125–34.PubMedCrossRef Gozu HI, Lublinghoff J, Bircan R, Paschke R. Genetics and phenomics of inherited and sporadic non-autoimmune hyperthyroidism. Mol Cell Endocrinol. 2010;322(1–2):125–34.PubMedCrossRef
107.
Zurück zum Zitat Ross DS. Radioiodine therapy for hyperthyroidism. N Engl J Med. 2011;364(20):1978–9.CrossRef Ross DS. Radioiodine therapy for hyperthyroidism. N Engl J Med. 2011;364(20):1978–9.CrossRef
108.
Zurück zum Zitat Amato E, Campennì A, Leotta S, Ruggeri RM, Baldari S. Treatment of hyperthyroidism with radioiodine targeted activity: a comparison between two dosimetric methods. Phys Med. 2016;32(6):847–53.PubMedCrossRef Amato E, Campennì A, Leotta S, Ruggeri RM, Baldari S. Treatment of hyperthyroidism with radioiodine targeted activity: a comparison between two dosimetric methods. Phys Med. 2016;32(6):847–53.PubMedCrossRef
109.
Zurück zum Zitat Kahraman D, Keller C, Schneider C, Eschner W, Sudbrock F, Schmidt M, et al. Development of hypothyroidism during long-term follow-up of patients with toxic nodular goitre after radioiodine therapy. Clin Endocrinol (Oxf). 2012;76(2):297–303.PubMedCrossRef Kahraman D, Keller C, Schneider C, Eschner W, Sudbrock F, Schmidt M, et al. Development of hypothyroidism during long-term follow-up of patients with toxic nodular goitre after radioiodine therapy. Clin Endocrinol (Oxf). 2012;76(2):297–303.PubMedCrossRef
110.
Zurück zum Zitat Reinhardt MJ, Joe A, Von Mallek D, Zimmerlin M, Manka-Waluch A, Palmedo H, et al. Dose selection for radioiodine therapy of borderline hyperthyroid patients with multifocal and disseminated autonomy on the basis of 99mTc-pertechnetate thyroid uptake. Eur J Nucl Med Mol Imaging. 2002;29(4):480–5.PubMedCrossRef Reinhardt MJ, Joe A, Von Mallek D, Zimmerlin M, Manka-Waluch A, Palmedo H, et al. Dose selection for radioiodine therapy of borderline hyperthyroid patients with multifocal and disseminated autonomy on the basis of 99mTc-pertechnetate thyroid uptake. Eur J Nucl Med Mol Imaging. 2002;29(4):480–5.PubMedCrossRef
111.
Zurück zum Zitat Dunkelmann S, Endlicher D, Prillwitz A, Rudolph F, Groth P, Schümichen C. Ergebnisse der TcTUs-optimierten Radioiodtherapie bei multifokaler und disseminierter Autonomie [Results of TcTUs-optimized radioiodine therapy in multifocal and disseminated autonomy]. Nuklearmedizin. 1999;38(5):131–9.PubMedCrossRef Dunkelmann S, Endlicher D, Prillwitz A, Rudolph F, Groth P, Schümichen C. Ergebnisse der TcTUs-optimierten Radioiodtherapie bei multifokaler und disseminierter Autonomie [Results of TcTUs-optimized radioiodine therapy in multifocal and disseminated autonomy]. Nuklearmedizin. 1999;38(5):131–9.PubMedCrossRef
112.
Zurück zum Zitat Reiners C, Schneider P. Radioiodine therapy of thyroid autonomy. Eur J Nucl Med Mol Imaging. 2002;29(Suppl 2):S471–8.PubMedCrossRef Reiners C, Schneider P. Radioiodine therapy of thyroid autonomy. Eur J Nucl Med Mol Imaging. 2002;29(Suppl 2):S471–8.PubMedCrossRef
113.
Zurück zum Zitat Reinhardt MJ, Biermann K, Wissmeyer M, Juengling FD, Brockmann H, Von Mallek D, et al. Dose selection for radioiodine therapy of borderline hyperthyroid patients according to thyroid uptake of 99mTc-pertechnetate: applicability to unifocal thyroid autonomy? Eur J Nucl Med Mol Imaging. 2006;33(5):608–12.PubMedCrossRef Reinhardt MJ, Biermann K, Wissmeyer M, Juengling FD, Brockmann H, Von Mallek D, et al. Dose selection for radioiodine therapy of borderline hyperthyroid patients according to thyroid uptake of 99mTc-pertechnetate: applicability to unifocal thyroid autonomy? Eur J Nucl Med Mol Imaging. 2006;33(5):608–12.PubMedCrossRef
114.
Zurück zum Zitat Nygaard B, Hegedüs L, Ulriksen P, Nielsen KG, Hansen JM. Radioiodine therapy for multinodular toxic goiter. Arch Intern Med. 1999;159(12):1364–8.PubMedCrossRef Nygaard B, Hegedüs L, Ulriksen P, Nielsen KG, Hansen JM. Radioiodine therapy for multinodular toxic goiter. Arch Intern Med. 1999;159(12):1364–8.PubMedCrossRef
115.
Zurück zum Zitat Erickson D, Gharib H, Li H, Van Heerden JA. Treatment of patients with toxic multinodular goiter. Thyroid. 1998;8(4):277–82.PubMedCrossRef Erickson D, Gharib H, Li H, Van Heerden JA. Treatment of patients with toxic multinodular goiter. Thyroid. 1998;8(4):277–82.PubMedCrossRef
116.
Zurück zum Zitat Kang AS, Grant CS, Thompson GB, Van Heerden JA, Pasieka JL, LoGerfo P, et al. Current treatment of nodular goiter with hyperthyroidism (Plummer’s disease): surgery versus radioiodine. Surgery. 2002;132(6):916–23.PubMedCrossRef Kang AS, Grant CS, Thompson GB, Van Heerden JA, Pasieka JL, LoGerfo P, et al. Current treatment of nodular goiter with hyperthyroidism (Plummer’s disease): surgery versus radioiodine. Surgery. 2002;132(6):916–23.PubMedCrossRef
117.
Zurück zum Zitat Zakavi SR, Mousavi Z, Davachi B. Comparison of four different protocols of I-131 therapy for treating single toxic thyroid nodule. Nucl Med Commun. 2009;30(2):169–75.PubMedCrossRef Zakavi SR, Mousavi Z, Davachi B. Comparison of four different protocols of I-131 therapy for treating single toxic thyroid nodule. Nucl Med Commun. 2009;30(2):169–75.PubMedCrossRef
118.
Zurück zum Zitat Ceccarelli C, Bencivelli W, Vitti P, Grasso L, Pinchera A. Outcome of radioiodine-131 therapy in hyperfunctioning thyroid nodules: a 20 years’ retrospective study. Clin Endocrinol (Oxf). 2005;62(3):331–5.PubMedCrossRef Ceccarelli C, Bencivelli W, Vitti P, Grasso L, Pinchera A. Outcome of radioiodine-131 therapy in hyperfunctioning thyroid nodules: a 20 years’ retrospective study. Clin Endocrinol (Oxf). 2005;62(3):331–5.PubMedCrossRef
119.
Zurück zum Zitat Holm L-E, Lundell G, Israelsson A, Dahlqvist I. Incidence of hypothyroidism occurring long after iodine-131 therapy for hyperthyroidism. J Nucl Med. 1982;23(2):103–7.PubMed Holm L-E, Lundell G, Israelsson A, Dahlqvist I. Incidence of hypothyroidism occurring long after iodine-131 therapy for hyperthyroidism. J Nucl Med. 1982;23(2):103–7.PubMed
120.
Zurück zum Zitat Yano Y, Sugino K, Akaishi J, Uruno T, Okuwa K, Shibuya H, et al. Treatment of autonomously functioning thyroid nodules at a single institution: radioiodine therapy, surgery, and ethanol injection therapy. Ann Nucl Med. 2011;25(10):749–54.PubMedCrossRef Yano Y, Sugino K, Akaishi J, Uruno T, Okuwa K, Shibuya H, et al. Treatment of autonomously functioning thyroid nodules at a single institution: radioiodine therapy, surgery, and ethanol injection therapy. Ann Nucl Med. 2011;25(10):749–54.PubMedCrossRef
121.
Zurück zum Zitat Nygaard B, Hegedüs L, Nielsen KG, Ulriksen P, Hansen JM. Long-term effect of radioactive iodine on thyroid function and size in patients with solitary autonomously functioning toxic thyroid nodules. Clin Endocrinol (Oxf). 1999;50(2):197–202.PubMedCrossRef Nygaard B, Hegedüs L, Nielsen KG, Ulriksen P, Hansen JM. Long-term effect of radioactive iodine on thyroid function and size in patients with solitary autonomously functioning toxic thyroid nodules. Clin Endocrinol (Oxf). 1999;50(2):197–202.PubMedCrossRef
122.
Zurück zum Zitat Bonnema SJ, Bertelsen H, Mortensen J, Andersen PB, Knudsen DU, Bastholt L, et al. The feasibility of high dose iodine 131 treatment as an alternative to surgery in patients with a very large goiter: effect on thyroid function and size and pulmonary function. J Clin Endocrinol Metab. 1999;84(10):3636–41.PubMed Bonnema SJ, Bertelsen H, Mortensen J, Andersen PB, Knudsen DU, Bastholt L, et al. The feasibility of high dose iodine 131 treatment as an alternative to surgery in patients with a very large goiter: effect on thyroid function and size and pulmonary function. J Clin Endocrinol Metab. 1999;84(10):3636–41.PubMed
123.
Zurück zum Zitat Rokni H, Sadeghi R, Moossavi Z, Treglia G, Zakavi SR. Efficacy of different protocols of radioiodine therapy for treatment of toxic nodular goiter: systematic review and meta-analysis of the literature. Int J Endocrinol Metab. 2014;12(2): e14424.PubMedPubMedCentralCrossRef Rokni H, Sadeghi R, Moossavi Z, Treglia G, Zakavi SR. Efficacy of different protocols of radioiodine therapy for treatment of toxic nodular goiter: systematic review and meta-analysis of the literature. Int J Endocrinol Metab. 2014;12(2): e14424.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Kobe C, Eschner W, Wild M, Rahlff I, Sudbrock F, Schmidt M, et al. Radioiodine therapy of benign thyroid disorders: what are the effective thyroidal half-life and uptake of 131I? Nucl Med Commun. 2010;31(3):201–5.PubMedCrossRef Kobe C, Eschner W, Wild M, Rahlff I, Sudbrock F, Schmidt M, et al. Radioiodine therapy of benign thyroid disorders: what are the effective thyroidal half-life and uptake of 131I? Nucl Med Commun. 2010;31(3):201–5.PubMedCrossRef
125.
Zurück zum Zitat Hammes J, van Heek L, Hohberg M, Reifegerst M, Stockter S, Dietlein M, Wild M, Drzezga A, Schmidt M, Kobe C. Impact of different approaches to calculation of treatment activities on achieved doses in radioiodine therapy of benign thyroid diseases. EJNMMI Phys. 2018;5(1):32. https://doi.org/10.1186/s40658-018-0231-x. Hammes J, van Heek L, Hohberg M, Reifegerst M, Stockter S, Dietlein M, Wild M, Drzezga A, Schmidt M, Kobe C. Impact of different approaches to calculation of treatment activities on achieved doses in radioiodine therapy of benign thyroid diseases. EJNMMI Phys. 2018;5(1):32. https://​doi.​org/​10.​1186/​s40658-018-0231-x.
126.
Zurück zum Zitat Bonnema SJ, Fast S, Hegedüs L. The role of radioiodine therapy in benign nodular goitre. Best Pract Res Clin Endocrinol Metab. 2014;28(4):619–31.PubMedCrossRef Bonnema SJ, Fast S, Hegedüs L. The role of radioiodine therapy in benign nodular goitre. Best Pract Res Clin Endocrinol Metab. 2014;28(4):619–31.PubMedCrossRef
127.
Zurück zum Zitat Hegedus L, Molholm Hansen B, Knudsen N, Molholm HJ. Reduction of size of thyroid with radioactive iodine in multinodular non-toxic goitre. Br Med J. 1988;297(6649):661–2.CrossRef Hegedus L, Molholm Hansen B, Knudsen N, Molholm HJ. Reduction of size of thyroid with radioactive iodine in multinodular non-toxic goitre. Br Med J. 1988;297(6649):661–2.CrossRef
128.
Zurück zum Zitat Gharib H, Papini E, Paschke R, Duick DS, Valcavi R, Hegedüs L, et al. American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and EuropeanThyroid Association Medical Guidelines for Clinical Practice for the Diagnosis and Management of Thyroid Nodules. Endocr Pract. 2010;16(Suppl 1):1–43.PubMedCrossRef Gharib H, Papini E, Paschke R, Duick DS, Valcavi R, Hegedüs L, et al. American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and EuropeanThyroid Association Medical Guidelines for Clinical Practice for the Diagnosis and Management of Thyroid Nodules. Endocr Pract. 2010;16(Suppl 1):1–43.PubMedCrossRef
129.
Zurück zum Zitat Paschke R, Hegedüs L, Alexander E, Valcavi R, Papini E, Gharib H. Thyroid nodule guidelines: agreement, disagreement and need for future research. Nat Rev Endocrinol. 2011;7(6):354–61.PubMedCrossRef Paschke R, Hegedüs L, Alexander E, Valcavi R, Papini E, Gharib H. Thyroid nodule guidelines: agreement, disagreement and need for future research. Nat Rev Endocrinol. 2011;7(6):354–61.PubMedCrossRef
130.
Zurück zum Zitat Atkins D, Best D, Briss PA, Eccles M, Falck-Ytter Y, Flottorp S, et al. Grading quality of evidence and strength of recommendations. BMJ. 2004;328(7454):1490.PubMedCrossRef Atkins D, Best D, Briss PA, Eccles M, Falck-Ytter Y, Flottorp S, et al. Grading quality of evidence and strength of recommendations. BMJ. 2004;328(7454):1490.PubMedCrossRef
131.
132.
133.
Zurück zum Zitat Sherman SI, Tuttle RM, Ball DW, Byrd D, Clark OH, Daniels GH, et al. NCCN clinical practice guidelines in oncology-thyroid carcinoma. Vol. 1. www.nccn.org; 2009. Accessed 14 Dec 2021. Sherman SI, Tuttle RM, Ball DW, Byrd D, Clark OH, Daniels GH, et al. NCCN clinical practice guidelines in oncology-thyroid carcinoma. Vol. 1. www.​nccn.​org; 2009. Accessed 14 Dec 2021.
134.
Zurück zum Zitat Hegedüs L, Bonnema SJ, Bennedbæk FN. Management of simple nodular goiter: current status and future perspectives. Endocr Rev. 2003;24(1):102–32.PubMedCrossRef Hegedüs L, Bonnema SJ, Bennedbæk FN. Management of simple nodular goiter: current status and future perspectives. Endocr Rev. 2003;24(1):102–32.PubMedCrossRef
135.
Zurück zum Zitat Bonnema SJ, Nielsen VE, Boel-Jørgensen H, Grupe P, Andersen PB, Bastholt L, et al. Recombinant human thyrotropin-stimulated radioiodine therapy of large nodular goiters facilitates tracheal decompression and improves inspiration. J Clin Endocrinol Metab. 2008;93(10):3981–4.PubMedCrossRef Bonnema SJ, Nielsen VE, Boel-Jørgensen H, Grupe P, Andersen PB, Bastholt L, et al. Recombinant human thyrotropin-stimulated radioiodine therapy of large nodular goiters facilitates tracheal decompression and improves inspiration. J Clin Endocrinol Metab. 2008;93(10):3981–4.PubMedCrossRef
136.
Zurück zum Zitat Medeiros-Neto G, Marui S, Knobel M. An outline concerning the potential use of recombinant human thyrotropin for improving radioiodine therapy of multinodular goiter. Endocrine. 2008;33(2):109–17.PubMedCrossRef Medeiros-Neto G, Marui S, Knobel M. An outline concerning the potential use of recombinant human thyrotropin for improving radioiodine therapy of multinodular goiter. Endocrine. 2008;33(2):109–17.PubMedCrossRef
137.
Zurück zum Zitat Melissant CF, Smith SJ, Perlberger R, Verschakelen J, Lammers JWJ, Demedts M. Lung function, CT-scan and X-ray in upper airway obstruction due to thyroid goitre. Eur Respir J. 1994;7(10):1782–7.PubMedCrossRef Melissant CF, Smith SJ, Perlberger R, Verschakelen J, Lammers JWJ, Demedts M. Lung function, CT-scan and X-ray in upper airway obstruction due to thyroid goitre. Eur Respir J. 1994;7(10):1782–7.PubMedCrossRef
138.
Zurück zum Zitat Huysmans DA, Hermus AR, Corstens FH, Barentsz JO, Kloppenborg PW. Large, compressive goiters treated with radioiodine. Ann Intern Med. 1994;121(10):757–62.PubMedCrossRef Huysmans DA, Hermus AR, Corstens FH, Barentsz JO, Kloppenborg PW. Large, compressive goiters treated with radioiodine. Ann Intern Med. 1994;121(10):757–62.PubMedCrossRef
139.
Zurück zum Zitat Nielsen VE, Bonnema SJ, Boel-Jørgensen H, Veje A, Hegedüs L. Recombinant human thyrotropin markedly changes the 131I kinetics during 131I therapy of patients with nodular goiter: an evaluation by a randomized double-blinded trial. J Clin Endocrinol Metab. 2005;90(1):79–83.PubMedCrossRef Nielsen VE, Bonnema SJ, Boel-Jørgensen H, Veje A, Hegedüs L. Recombinant human thyrotropin markedly changes the 131I kinetics during 131I therapy of patients with nodular goiter: an evaluation by a randomized double-blinded trial. J Clin Endocrinol Metab. 2005;90(1):79–83.PubMedCrossRef
140.
Zurück zum Zitat Medeiros-Neto G. Iodine deficiency disorders. In: Jameson JL, De Groot LJ, editors. Endocrinology. Philadelphia: WB Saunders; 2000. p. 1529–39. Medeiros-Neto G. Iodine deficiency disorders. In: Jameson JL, De Groot LJ, editors. Endocrinology. Philadelphia: WB Saunders; 2000. p. 1529–39.
141.
Zurück zum Zitat Harach HR, Williams ED. Thyroid cancer and thyroiditis in the goitrous region of Salta, Argentina, before and after iodine prophylaxis. Clin Endocrinol (Oxf). 1995;43(6):701–6.PubMedCrossRef Harach HR, Williams ED. Thyroid cancer and thyroiditis in the goitrous region of Salta, Argentina, before and after iodine prophylaxis. Clin Endocrinol (Oxf). 1995;43(6):701–6.PubMedCrossRef
142.
Zurück zum Zitat Berghout A, Wiersinga WM, Touber JL, Smits NJ, Drexhage HA. Comparison of placebo with L-thyroxine alone or with carbimazole for treatment of sporadic non-toxic goitre. Lancet (London, England). 1990;336(8709):193–7.PubMedCrossRef Berghout A, Wiersinga WM, Touber JL, Smits NJ, Drexhage HA. Comparison of placebo with L-thyroxine alone or with carbimazole for treatment of sporadic non-toxic goitre. Lancet (London, England). 1990;336(8709):193–7.PubMedCrossRef
143.
Zurück zum Zitat Lima N, Knobel M, Cavaliere H, Sztejnsznajd C, Tomimori E, Medeiros-Neto G. Levothyroxine suppressive therapy is partially effective in treating patients with benign, solid thyroid nodules and multinodular goiters. Thyroid. 1997;7(5):691–7.PubMedCrossRef Lima N, Knobel M, Cavaliere H, Sztejnsznajd C, Tomimori E, Medeiros-Neto G. Levothyroxine suppressive therapy is partially effective in treating patients with benign, solid thyroid nodules and multinodular goiters. Thyroid. 1997;7(5):691–7.PubMedCrossRef
144.
Zurück zum Zitat Wesche MF, Tiel-v Buul MM, Lips P, Smits NJ, Wiersinga WM. A randomized trial comparing levothyroxine with radioactive iodine in the treatment of sporadic nontoxic goiter. J Clin Endocrinol Metab. 2001;86(3):998–1005.PubMedCrossRef Wesche MF, Tiel-v Buul MM, Lips P, Smits NJ, Wiersinga WM. A randomized trial comparing levothyroxine with radioactive iodine in the treatment of sporadic nontoxic goiter. J Clin Endocrinol Metab. 2001;86(3):998–1005.PubMedCrossRef
145.
Zurück zum Zitat Parle JV, Maisonneuve P, Sheppard MC, Boyle P, Franklyn JA. Prediction of all-cause and cardiovascular mortality in elderly people from one low serum thyrotropin result: a 10-year cohort study. Lancet (London, England). 2001;358(9285):861–5.PubMedCrossRef Parle JV, Maisonneuve P, Sheppard MC, Boyle P, Franklyn JA. Prediction of all-cause and cardiovascular mortality in elderly people from one low serum thyrotropin result: a 10-year cohort study. Lancet (London, England). 2001;358(9285):861–5.PubMedCrossRef
146.
147.
Zurück zum Zitat Schumm-Draeger PM. Drug therapy of goiter. Iodine, thyroid hormones or combined therapy. Z Gesamte Inn Med. 1993;48(12):592–8.PubMed Schumm-Draeger PM. Drug therapy of goiter. Iodine, thyroid hormones or combined therapy. Z Gesamte Inn Med. 1993;48(12):592–8.PubMed
148.
Zurück zum Zitat al-Suliman NN, Ryttov NF, Qvist N, Blichert-Toft M, Graversen HP. Experience in a specialist thyroid surgery unit: a demographic study, surgical complications, and outcome. Eur J Surg. 1997;163(1):13–20.PubMed al-Suliman NN, Ryttov NF, Qvist N, Blichert-Toft M, Graversen HP. Experience in a specialist thyroid surgery unit: a demographic study, surgical complications, and outcome. Eur J Surg. 1997;163(1):13–20.PubMed
149.
Zurück zum Zitat Thomusch O, Machens A, Sekulla C, Ukkat J, Lippert H, Gastinger I, et al. Multivariate analysis of risk factors for postoperative complications in benign goiter surgery: prospective multicenter study in Germany. World J Surg. 2000;24(11):1335–41.PubMedCrossRef Thomusch O, Machens A, Sekulla C, Ukkat J, Lippert H, Gastinger I, et al. Multivariate analysis of risk factors for postoperative complications in benign goiter surgery: prospective multicenter study in Germany. World J Surg. 2000;24(11):1335–41.PubMedCrossRef
150.
Zurück zum Zitat Abdel Rahim AA, Ahmed ME, Hassan MA. Respiratory complications after thyroidectomy and the need for tracheostomy in patients with a large goitre. Br J Surg. 1999;86(1):88–90.PubMedCrossRef Abdel Rahim AA, Ahmed ME, Hassan MA. Respiratory complications after thyroidectomy and the need for tracheostomy in patients with a large goitre. Br J Surg. 1999;86(1):88–90.PubMedCrossRef
151.
Zurück zum Zitat Maruotti RA, Zannini P, Viani MP, Voci C, Pezzuoli G. Surgical treatment of substernal goiters. Int Surg. 1991;76(1):12–7.PubMed Maruotti RA, Zannini P, Viani MP, Voci C, Pezzuoli G. Surgical treatment of substernal goiters. Int Surg. 1991;76(1):12–7.PubMed
152.
Zurück zum Zitat Torre G, Borgonovo G, Amato A, Arezzo A, Ansaldo G, De Negri A, et al. Surgical management of substernal goiter: analysis of 237 patients. Am Surg. 1995;61(9):826–31.PubMed Torre G, Borgonovo G, Amato A, Arezzo A, Ansaldo G, De Negri A, et al. Surgical management of substernal goiter: analysis of 237 patients. Am Surg. 1995;61(9):826–31.PubMed
153.
154.
Zurück zum Zitat Mishra A, Agarwal A, Agarwal G, Mishra SK. Total thyroidectomy for benign thyroid disorders in an endemic region. World J Surg. 2001;25(3):307–10.PubMedCrossRef Mishra A, Agarwal A, Agarwal G, Mishra SK. Total thyroidectomy for benign thyroid disorders in an endemic region. World J Surg. 2001;25(3):307–10.PubMedCrossRef
155.
Zurück zum Zitat Pappalardo G, Guadalaxara A, Frattaroli FM, Illomei G, Falaschi P. Total compared with subtotal thyroidectomy in benign nodular disease: personal series and review of published reports. Eur J Surg. 1998;164(7):501–6.PubMedCrossRef Pappalardo G, Guadalaxara A, Frattaroli FM, Illomei G, Falaschi P. Total compared with subtotal thyroidectomy in benign nodular disease: personal series and review of published reports. Eur J Surg. 1998;164(7):501–6.PubMedCrossRef
156.
Zurück zum Zitat Hisham AN, Azlina AF, Aina EN, Sarojah A. Total thyroidectomy: the procedure of choice for multinodular goitre. Eur J Surg. 2001;167(6):403–5.PubMedCrossRef Hisham AN, Azlina AF, Aina EN, Sarojah A. Total thyroidectomy: the procedure of choice for multinodular goitre. Eur J Surg. 2001;167(6):403–5.PubMedCrossRef
157.
Zurück zum Zitat Seiler CA, Glaser C, Wagner HE. Thyroid gland surgery in an endemic region. World J Surg. 1996;20(5):593–7.PubMedCrossRef Seiler CA, Glaser C, Wagner HE. Thyroid gland surgery in an endemic region. World J Surg. 1996;20(5):593–7.PubMedCrossRef
158.
Zurück zum Zitat Bachmann J, Kobe C, Bor S, Rahlff I, Dietlein M, Schicha H, et al. Radioiodine therapy for thyroid volume reduction of large goitres. Nucl Med Commun. 2009;30(6):466–71.PubMedCrossRef Bachmann J, Kobe C, Bor S, Rahlff I, Dietlein M, Schicha H, et al. Radioiodine therapy for thyroid volume reduction of large goitres. Nucl Med Commun. 2009;30(6):466–71.PubMedCrossRef
159.
Zurück zum Zitat Nielsen VE, Bonnema SJ, Boel-Jørgensen H, Grupe P, Hegedüs L. Stimulation with 0.3-mg recombinant human thyrotropin prior to iodine 131 therapy to improve the size reduction of benign nontoxic nodular goiter: a prospective randomized double-blind trial. Arch Intern Med. 2006;166(14):1476–82.PubMedCrossRef Nielsen VE, Bonnema SJ, Boel-Jørgensen H, Grupe P, Hegedüs L. Stimulation with 0.3-mg recombinant human thyrotropin prior to iodine 131 therapy to improve the size reduction of benign nontoxic nodular goiter: a prospective randomized double-blind trial. Arch Intern Med. 2006;166(14):1476–82.PubMedCrossRef
160.
Zurück zum Zitat Bonnema SJ, Nielsen VE, Boel-Jørgensen H, Grupe P, Andersen PB, Bastholt L, et al. Improvement of goiter volume reduction after 0.3 mg recombinant human thyrotropin-stimulated radioiodine therapy in patients with a very large goiter: a double-blinded, randomized trial. J Clin Endocrinol Metab. 2007;92(9):3424–8.PubMedCrossRef Bonnema SJ, Nielsen VE, Boel-Jørgensen H, Grupe P, Andersen PB, Bastholt L, et al. Improvement of goiter volume reduction after 0.3 mg recombinant human thyrotropin-stimulated radioiodine therapy in patients with a very large goiter: a double-blinded, randomized trial. J Clin Endocrinol Metab. 2007;92(9):3424–8.PubMedCrossRef
161.
Zurück zum Zitat Verelst J, Bonnyns M, Glinoer D. Radioiodine therapy in voluminous multinodular non-toxic goitre. Acta Endocrinol (Copenh). 1990;122(4):417–21.PubMed Verelst J, Bonnyns M, Glinoer D. Radioiodine therapy in voluminous multinodular non-toxic goitre. Acta Endocrinol (Copenh). 1990;122(4):417–21.PubMed
162.
Zurück zum Zitat Nygaard B, Hegedus L, Gervil M, Hjalgrim H, Soe-Jensen P, Molholm HJ. Radioiodine treatment of multinodular non-toxic goitre. BMJ. 1993;307(6908):828–32.PubMedPubMedCentralCrossRef Nygaard B, Hegedus L, Gervil M, Hjalgrim H, Soe-Jensen P, Molholm HJ. Radioiodine treatment of multinodular non-toxic goitre. BMJ. 1993;307(6908):828–32.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Huysmans D, Hermus A, Edelbroek M, Barentsz J, Corstens F, Kloppenborg P. Radioiodine for nontoxic multinodular goiter. Thyroid. 1997;7(2):235–9.PubMedCrossRef Huysmans D, Hermus A, Edelbroek M, Barentsz J, Corstens F, Kloppenborg P. Radioiodine for nontoxic multinodular goiter. Thyroid. 1997;7(2):235–9.PubMedCrossRef
164.
Zurück zum Zitat de Klerk JM, van Isselt JW, van Dijk A, Hakman ME, Pameijer FA, Koppeschaar HP, et al. Iodine-131 therapy in sporadic nontoxic goiter. J Nucl Med. 1997;38(3):372–6.PubMed de Klerk JM, van Isselt JW, van Dijk A, Hakman ME, Pameijer FA, Koppeschaar HP, et al. Iodine-131 therapy in sporadic nontoxic goiter. J Nucl Med. 1997;38(3):372–6.PubMed
165.
Zurück zum Zitat Le Moli R, Wesche MF, Tiel-Van Buul MM, Wiersinga WM. Determinants of longterm outcome of radioiodine therapy of sporadic non-toxic goitre. Clin Endocrinol (Oxf). 1999;50(6):783–9.PubMedCrossRef Le Moli R, Wesche MF, Tiel-Van Buul MM, Wiersinga WM. Determinants of longterm outcome of radioiodine therapy of sporadic non-toxic goitre. Clin Endocrinol (Oxf). 1999;50(6):783–9.PubMedCrossRef
166.
Zurück zum Zitat Bonnema SJ, Nielsen VE, Hegedüs L. Long-term effects of radioiodine on thyroid function, size and patient satisfaction in non-toxic diffuse goitre. Eur J Endocrinol. 2004;150(4):439–45.PubMedCrossRef Bonnema SJ, Nielsen VE, Hegedüs L. Long-term effects of radioiodine on thyroid function, size and patient satisfaction in non-toxic diffuse goitre. Eur J Endocrinol. 2004;150(4):439–45.PubMedCrossRef
167.
Zurück zum Zitat Greenlee C, Burmeister LA, Butler RS, Edinboro CH, Morrison SMI, Milas M. Current safety practices relating to I-131 administration for diseases of the thyroid: a survey of physicians and allied practitioners. Thyroid. 2011;21(2):151–60.PubMedCrossRef Greenlee C, Burmeister LA, Butler RS, Edinboro CH, Morrison SMI, Milas M. Current safety practices relating to I-131 administration for diseases of the thyroid: a survey of physicians and allied practitioners. Thyroid. 2011;21(2):151–60.PubMedCrossRef
168.
Zurück zum Zitat Fast S, Nielsen VE, Grupe P, Bonnema SJ, Hegedüs L. Optimizing 131I uptake after rhTSH stimulation in patients with nontoxic multinodular goiter: evidence from a prospective, randomized, double-blind study. J Nucl Med. 2009;50(5):732–7.PubMedCrossRef Fast S, Nielsen VE, Grupe P, Bonnema SJ, Hegedüs L. Optimizing 131I uptake after rhTSH stimulation in patients with nontoxic multinodular goiter: evidence from a prospective, randomized, double-blind study. J Nucl Med. 2009;50(5):732–7.PubMedCrossRef
169.
Zurück zum Zitat Huysmans DAKC, Buijs WCAM, van de Ven MTP, van den Broek WJM, Kloppenborg PWC, Hermus ARMM, et al. Dosimetry and risk estimates of radioiodine therapy for large, multinodular goiters. J Nucl Med. 1996;37(12):2072–9.PubMed Huysmans DAKC, Buijs WCAM, van de Ven MTP, van den Broek WJM, Kloppenborg PWC, Hermus ARMM, et al. Dosimetry and risk estimates of radioiodine therapy for large, multinodular goiters. J Nucl Med. 1996;37(12):2072–9.PubMed
170.
Zurück zum Zitat Papini E, Petrucci L, Guglielmi R, Panunzi C, Rinaldi R, Bacci V, et al. Long-term changes in nodular goiter: a 5-year prospective randomized trial of levothyroxine suppressive therapy for benign cold thyroid nodules. J Clin Endocrinol Metab. 1998;83(3):780–3.PubMedCrossRef Papini E, Petrucci L, Guglielmi R, Panunzi C, Rinaldi R, Bacci V, et al. Long-term changes in nodular goiter: a 5-year prospective randomized trial of levothyroxine suppressive therapy for benign cold thyroid nodules. J Clin Endocrinol Metab. 1998;83(3):780–3.PubMedCrossRef
171.
Zurück zum Zitat Giusti M, Cappi C, Santaniello B, Ceresola E, Augeri C, Lagasio C, et al. Safety and efficacy of administering 0.2 mg of recombinant human TSH for two consecutive days as an adjuvant to therapy with low radioiodine doses in elderly out-patients with large nontoxic multinodular goiter. Minerva Endocrinol. 2006;31(3):191–209.PubMed Giusti M, Cappi C, Santaniello B, Ceresola E, Augeri C, Lagasio C, et al. Safety and efficacy of administering 0.2 mg of recombinant human TSH for two consecutive days as an adjuvant to therapy with low radioiodine doses in elderly out-patients with large nontoxic multinodular goiter. Minerva Endocrinol. 2006;31(3):191–209.PubMed
172.
Zurück zum Zitat Cohen O, Ilany J, Hoffman C, Olchovsky D, Dabhi S, Karasik A, et al. Low-dose recombinant human thyrotropin-aided radioiodine treatment of large, multinodular goiters in elderly patients. Eur J Endocrinol. 2006;154(2):243–52.PubMedCrossRef Cohen O, Ilany J, Hoffman C, Olchovsky D, Dabhi S, Karasik A, et al. Low-dose recombinant human thyrotropin-aided radioiodine treatment of large, multinodular goiters in elderly patients. Eur J Endocrinol. 2006;154(2):243–52.PubMedCrossRef
173.
Zurück zum Zitat Nieuwlaat WA, Huysmans DA, Van Den Bosch HC, Sweep CG, Ross HA, Corstens FH, et al. Pretreatment with a single, low dose of recombinant human thyrotropin allows dose reduction of radioiodine therapy in patients with nodular goiter. J Clin Endocrinol Metab. 2003;88(7):3121–9.PubMedCrossRef Nieuwlaat WA, Huysmans DA, Van Den Bosch HC, Sweep CG, Ross HA, Corstens FH, et al. Pretreatment with a single, low dose of recombinant human thyrotropin allows dose reduction of radioiodine therapy in patients with nodular goiter. J Clin Endocrinol Metab. 2003;88(7):3121–9.PubMedCrossRef
174.
Zurück zum Zitat Duick DS, Baskin HJ. Utility of recombinant human thyrotropin for augmentation of radioiodine uptake and treatment of nontoxic and toxic multinodular goiters. Endocr Pract. 2003;9(3):204–9.PubMedCrossRef Duick DS, Baskin HJ. Utility of recombinant human thyrotropin for augmentation of radioiodine uptake and treatment of nontoxic and toxic multinodular goiters. Endocr Pract. 2003;9(3):204–9.PubMedCrossRef
175.
Zurück zum Zitat Silva MNC, Rubió IGS, Romão R, Gebrin EMMS, Buchpiguel C, Tomimori E, et al. Administration of a single dose of recombinant human thyrotrophin enhances the efficacy of radioiodine treatment of large compressive multinodular goitres. Clin Endocrinol (Oxf). 2004;60(3):300–8.PubMedCrossRef Silva MNC, Rubió IGS, Romão R, Gebrin EMMS, Buchpiguel C, Tomimori E, et al. Administration of a single dose of recombinant human thyrotrophin enhances the efficacy of radioiodine treatment of large compressive multinodular goitres. Clin Endocrinol (Oxf). 2004;60(3):300–8.PubMedCrossRef
176.
Zurück zum Zitat Albino CC, Mesa CO, Olandoski M, Ueda CE, Woellner LC, Goedert CA, et al. Recombinant human thyrotropin as adjuvant in the treatment of multinodular goiters with radioiodine. J Clin Endocrinol Metab. 2005;90(5):2775–80.PubMedCrossRef Albino CC, Mesa CO, Olandoski M, Ueda CE, Woellner LC, Goedert CA, et al. Recombinant human thyrotropin as adjuvant in the treatment of multinodular goiters with radioiodine. J Clin Endocrinol Metab. 2005;90(5):2775–80.PubMedCrossRef
177.
Zurück zum Zitat Paz-Filho GJ, Mesa-Junior CO, Olandoski M, Woellner LC, Goedert CA, Boguszewski CI, et al. Effect of 30 mCi radioiodine on multinodular goiter previously treated with recombinant human thyroid-stimulating hormone. Brazilian J Med Biol Res = Rev Bras Pesqui medicas e Biol. 2007;40(12):1661–70.CrossRef Paz-Filho GJ, Mesa-Junior CO, Olandoski M, Woellner LC, Goedert CA, Boguszewski CI, et al. Effect of 30 mCi radioiodine on multinodular goiter previously treated with recombinant human thyroid-stimulating hormone. Brazilian J Med Biol Res = Rev Bras Pesqui medicas e Biol. 2007;40(12):1661–70.CrossRef
178.
Zurück zum Zitat Nielsen VE, Bonnema SJ, Hegedüs L. Transient goiter enlargement after administration of 0.3 mg of recombinant human thyrotropin in patients with benign nontoxic nodular goiter: a randomized, double-blind, crossover trial. J Clin Endocrinol Metab. 2006;91(4):1317–22.PubMedCrossRef Nielsen VE, Bonnema SJ, Hegedüs L. Transient goiter enlargement after administration of 0.3 mg of recombinant human thyrotropin in patients with benign nontoxic nodular goiter: a randomized, double-blind, crossover trial. J Clin Endocrinol Metab. 2006;91(4):1317–22.PubMedCrossRef
179.
Zurück zum Zitat Bonnema SJ, Hegedüs L. A 30-year perspective on radioiodine therapy of benign nontoxic multinodular goiter. Curr Opin Endocrinol Diabetes Obes. 2009;16(5):379–84.PubMedCrossRef Bonnema SJ, Hegedüs L. A 30-year perspective on radioiodine therapy of benign nontoxic multinodular goiter. Curr Opin Endocrinol Diabetes Obes. 2009;16(5):379–84.PubMedCrossRef
180.
Zurück zum Zitat Huysmans DA, Nieuwlaat W-A, Erdtsieck RJ, Schellekens AP, Bus JW, Bravenboer B, et al. Administration of a single low dose of recombinant human thyrotropin significantly enhances thyroid radioiodide uptake in nontoxic nodular goiter. J Clin Endocrinol Metab. 2000;85(10):3592–6.PubMed Huysmans DA, Nieuwlaat W-A, Erdtsieck RJ, Schellekens AP, Bus JW, Bravenboer B, et al. Administration of a single low dose of recombinant human thyrotropin significantly enhances thyroid radioiodide uptake in nontoxic nodular goiter. J Clin Endocrinol Metab. 2000;85(10):3592–6.PubMed
181.
Zurück zum Zitat Woodmansee WW, Haugen BR. Uses for recombinant human TSH in patients with thyroid cancer and nodular goiter. Clin Endocrinol (Oxf). 2004;61(2):163–73.PubMedCrossRef Woodmansee WW, Haugen BR. Uses for recombinant human TSH in patients with thyroid cancer and nodular goiter. Clin Endocrinol (Oxf). 2004;61(2):163–73.PubMedCrossRef
182.
Zurück zum Zitat Bonnema SJ, Fast S, Hegedüs L. Non-surgical approach to the benign nodular goiter: new opportunities by recombinant human TSH-stimulated 131I-therapy. Endocrine. 2011;40(3):344–53.PubMedCrossRef Bonnema SJ, Fast S, Hegedüs L. Non-surgical approach to the benign nodular goiter: new opportunities by recombinant human TSH-stimulated 131I-therapy. Endocrine. 2011;40(3):344–53.PubMedCrossRef
183.
Zurück zum Zitat Fast S, Bonnema SJ, Hegedüs L. Radioiodine therapy of benign non-toxic goitre. Potential role of recombinant human TSH. Ann Endocrinol (Paris). 2011;72(2):129–35.PubMedCrossRef Fast S, Bonnema SJ, Hegedüs L. Radioiodine therapy of benign non-toxic goitre. Potential role of recombinant human TSH. Ann Endocrinol (Paris). 2011;72(2):129–35.PubMedCrossRef
184.
Zurück zum Zitat Pena S, Arum S, Cross M, Magnani B, Pearce EN, Oates ME, et al. 123I thyroid uptake and thyroid size at 24, 48, and 72 hours after the administration of recombinant human thyroid-stimulating hormone to normal volunteers. J Clin Endocrinol Metab. 2006;91(2):506–10.PubMedCrossRef Pena S, Arum S, Cross M, Magnani B, Pearce EN, Oates ME, et al. 123I thyroid uptake and thyroid size at 24, 48, and 72 hours after the administration of recombinant human thyroid-stimulating hormone to normal volunteers. J Clin Endocrinol Metab. 2006;91(2):506–10.PubMedCrossRef
185.
Zurück zum Zitat Cubas ER, Paz-Filho GJ, Olandoski M, Goedert CA, Woellner LC, Carvalho GA, et al. Recombinant human TSH increases the efficacy of a fixed activity of radioiodine for treatment of multinodular goitre. Int J Clin Pract. 2009;63(4):583–90.PubMedCrossRef Cubas ER, Paz-Filho GJ, Olandoski M, Goedert CA, Woellner LC, Carvalho GA, et al. Recombinant human TSH increases the efficacy of a fixed activity of radioiodine for treatment of multinodular goitre. Int J Clin Pract. 2009;63(4):583–90.PubMedCrossRef
186.
Zurück zum Zitat Fast S, Nielsen VE, Grupe P, Boel-Jørgensen H, Bastholt L, Andersen PB, et al. Prestimulation with recombinant human thyrotropin (rhTSH) improves the long-term outcome of radioiodine therapy for multinodular nontoxic goiter. J Clin Endocrinol Metab. 2012;97(8):2653–60.PubMedCrossRef Fast S, Nielsen VE, Grupe P, Boel-Jørgensen H, Bastholt L, Andersen PB, et al. Prestimulation with recombinant human thyrotropin (rhTSH) improves the long-term outcome of radioiodine therapy for multinodular nontoxic goiter. J Clin Endocrinol Metab. 2012;97(8):2653–60.PubMedCrossRef
187.
Zurück zum Zitat Read CH, Tansey MJ, Menda Y. A 36-year retrospective analysis of the efficacy and safety of radioactive iodine in treating young Graves’ patients. J Clin Endocrinol Metab. 2004;89(9):4229–33.PubMedCrossRef Read CH, Tansey MJ, Menda Y. A 36-year retrospective analysis of the efficacy and safety of radioactive iodine in treating young Graves’ patients. J Clin Endocrinol Metab. 2004;89(9):4229–33.PubMedCrossRef
188.
Zurück zum Zitat Mizokami T, Hamada K, Maruta T, Higashi K, Tajiri J. Long-term outcomes of radioiodine therapy for juvenile Graves disease with emphasis on subsequently detected thyroid nodules: a single institution experience from Japan. Endocr Pract. 2020;26(7):729–37.PubMedCrossRef Mizokami T, Hamada K, Maruta T, Higashi K, Tajiri J. Long-term outcomes of radioiodine therapy for juvenile Graves disease with emphasis on subsequently detected thyroid nodules: a single institution experience from Japan. Endocr Pract. 2020;26(7):729–37.PubMedCrossRef
189.
Zurück zum Zitat Lutterman SL, Zwaveling-Soonawala N, Verberne HJ, Verburg FA, Van Trotsenburg ASP, Mooij CF. The efficacy and short- and long-term side effects of radioactive iodine treatment in pediatric Graves’ disease: a systematic review. Eur Thyroid J. 2021;10(5):353–63.PubMedPubMedCentralCrossRef Lutterman SL, Zwaveling-Soonawala N, Verberne HJ, Verburg FA, Van Trotsenburg ASP, Mooij CF. The efficacy and short- and long-term side effects of radioactive iodine treatment in pediatric Graves’ disease: a systematic review. Eur Thyroid J. 2021;10(5):353–63.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat De LS, Lee SY, Braverman LE. Hyperthyroidism. Lancet. 2016;388(10047):906–18.CrossRef De LS, Lee SY, Braverman LE. Hyperthyroidism. Lancet. 2016;388(10047):906–18.CrossRef
191.
Zurück zum Zitat Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, Tsuboi K, et al. 2016 Guidelines for the management of thyroid storm from The Japan Thyroid Association and Japan Endocrine Society (First edition). Endocr J. 2016;63(12):1025–64.PubMedCrossRef Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, Tsuboi K, et al. 2016 Guidelines for the management of thyroid storm from The Japan Thyroid Association and Japan Endocrine Society (First edition). Endocr J. 2016;63(12):1025–64.PubMedCrossRef
192.
Zurück zum Zitat Mandel SJ, Mandel L. Radioactive iodine and the salivary glands. Thyroid. 2003;13(3):265–71.PubMedCrossRef Mandel SJ, Mandel L. Radioactive iodine and the salivary glands. Thyroid. 2003;13(3):265–71.PubMedCrossRef
193.
Zurück zum Zitat Grewal RK, Larson SM, Pentlow CE, Pentlow KS, Gonen M, Qualey R, et al. Salivary gland side effects commonly develop several weeks after initial radioactive iodine ablation. J Nucl Med. 2009;50(10):1605–10.PubMedCrossRef Grewal RK, Larson SM, Pentlow CE, Pentlow KS, Gonen M, Qualey R, et al. Salivary gland side effects commonly develop several weeks after initial radioactive iodine ablation. J Nucl Med. 2009;50(10):1605–10.PubMedCrossRef
194.
Zurück zum Zitat Van Nostrand D, Bandaru V, Chennupati S, Wexler J, Kulkarni K, Atkins F, et al. Radiopharmacokinetics of radioiodine in the parotid glands after the administration of lemon juice. Thyroid. 2010;20(10):1113–9.PubMedCrossRef Van Nostrand D, Bandaru V, Chennupati S, Wexler J, Kulkarni K, Atkins F, et al. Radiopharmacokinetics of radioiodine in the parotid glands after the administration of lemon juice. Thyroid. 2010;20(10):1113–9.PubMedCrossRef
195.
Zurück zum Zitat Nakada K, Ishibashi T, Takei T, Hirata K, Shinohara K, Katoh S, et al. Does lemon candy decrease salivary gland damage after radioiodine therapy for thyroid cancer? J Nucl Med. 2005;46(2):261–6.PubMed Nakada K, Ishibashi T, Takei T, Hirata K, Shinohara K, Katoh S, et al. Does lemon candy decrease salivary gland damage after radioiodine therapy for thyroid cancer? J Nucl Med. 2005;46(2):261–6.PubMed
196.
Zurück zum Zitat Jentzen W, Balschuweit D, Schmitz J, Freudenberg L, Eising E, Hilbel T, et al. The influence of saliva flow stimulation on the absorbed radiation dose to the salivary glands during radioiodine therapy of thyroid cancer using 124I PET(/CT) imaging. Eur J Nucl Med Mol Imaging. 2010;37(12):2298–306.PubMedCrossRef Jentzen W, Balschuweit D, Schmitz J, Freudenberg L, Eising E, Hilbel T, et al. The influence of saliva flow stimulation on the absorbed radiation dose to the salivary glands during radioiodine therapy of thyroid cancer using 124I PET(/CT) imaging. Eur J Nucl Med Mol Imaging. 2010;37(12):2298–306.PubMedCrossRef
197.
Zurück zum Zitat Marine D, Lenhart CH. Pathological anatomy of exophthalmic goiter: the anatomical and physiological relations of the thyroid gland to the disease; the treatment. Arch Intern Med. 1911;VIII(3):265–316.CrossRef Marine D, Lenhart CH. Pathological anatomy of exophthalmic goiter: the anatomical and physiological relations of the thyroid gland to the disease; the treatment. Arch Intern Med. 1911;VIII(3):265–316.CrossRef
198.
Zurück zum Zitat Giuffrida G, Giovinazzo S, Certo R, Vicchio TM, Baldari S, Campennì A, et al. An uncommon case of Marine-Lenhart syndrome. Arq Bras Endocrinol Metabol. 2014;58(4):398–401.PubMedCrossRef Giuffrida G, Giovinazzo S, Certo R, Vicchio TM, Baldari S, Campennì A, et al. An uncommon case of Marine-Lenhart syndrome. Arq Bras Endocrinol Metabol. 2014;58(4):398–401.PubMedCrossRef
199.
Zurück zum Zitat Charkes ND. Graves’ disease with functioning nodules (Marine-Lenhart Syndrome). J Nucl Med. 1972;13(12):885–92.PubMed Charkes ND. Graves’ disease with functioning nodules (Marine-Lenhart Syndrome). J Nucl Med. 1972;13(12):885–92.PubMed
200.
Zurück zum Zitat Chiovato L, Santini F, Vitti P, Bendinelli G, Pinchera A. Appearance of thyroid stimulating antibody and Graves’ disease after radioiodine therapy for toxic nodular goitre. Clin Endocrinol (Oxf). 1994;40(6):803–6.PubMedCrossRef Chiovato L, Santini F, Vitti P, Bendinelli G, Pinchera A. Appearance of thyroid stimulating antibody and Graves’ disease after radioiodine therapy for toxic nodular goitre. Clin Endocrinol (Oxf). 1994;40(6):803–6.PubMedCrossRef
201.
Zurück zum Zitat Schmidt M, Gorbauch E, Dietlein M, Faust M, Stützer H, Eschner W, et al. Incidence of postradioiodine immunogenic hyperthyroidism/Graves’ disease in relation to a temporary increase in thyrotropin receptor antibodies after radioiodine therapy for autonomous thyroid disease. Thyroid. 2006;16(3):281–8.PubMedCrossRef Schmidt M, Gorbauch E, Dietlein M, Faust M, Stützer H, Eschner W, et al. Incidence of postradioiodine immunogenic hyperthyroidism/Graves’ disease in relation to a temporary increase in thyrotropin receptor antibodies after radioiodine therapy for autonomous thyroid disease. Thyroid. 2006;16(3):281–8.PubMedCrossRef
202.
Zurück zum Zitat Boddenberg B, Voth E, Schicha H. Immunogenic hyperthyroidism following radioiodine ablation of focal autonomy. Immunogene Hyperthyreose nach Radiojod-Ablation einer fokalen Autonomie. Nukl. 1993;32(1):18–22. Boddenberg B, Voth E, Schicha H. Immunogenic hyperthyroidism following radioiodine ablation of focal autonomy. Immunogene Hyperthyreose nach Radiojod-Ablation einer fokalen Autonomie. Nukl. 1993;32(1):18–22.
203.
Zurück zum Zitat Custro N, Ganci A, Scafidi V. Relapses of hyperthyroidism in patients treated with radioiodine for nodular toxic goiter: relation to thyroid autoimmunity. J Endocrinol Invest. 2003;26(2):106–10.PubMedCrossRef Custro N, Ganci A, Scafidi V. Relapses of hyperthyroidism in patients treated with radioiodine for nodular toxic goiter: relation to thyroid autoimmunity. J Endocrinol Invest. 2003;26(2):106–10.PubMedCrossRef
204.
Zurück zum Zitat Chiappori A, Villalta D, Bossert I, Ceresola EM, Lanaro D, Schiavo M, et al. Thyrotropin receptor autoantibody measurement following radiometabolic treatment of hyperthyroidism: comparison between different methods. J Endocrinol Invest. 2010;33(3):197–201.PubMedCrossRef Chiappori A, Villalta D, Bossert I, Ceresola EM, Lanaro D, Schiavo M, et al. Thyrotropin receptor autoantibody measurement following radiometabolic treatment of hyperthyroidism: comparison between different methods. J Endocrinol Invest. 2010;33(3):197–201.PubMedCrossRef
205.
Zurück zum Zitat Aizawa Y, Yoshida K, Kaise N, Fukazawa H, Kiso Y, Sayama N, et al. The development of transient hypothyroidism after iodine-131 treatment in hyperthyroid patients with Graves’ disease: prevalence, mechanism and prognosis. Clin Endocrinol (Oxf). 1997;46(1):1–5.PubMedCrossRef Aizawa Y, Yoshida K, Kaise N, Fukazawa H, Kiso Y, Sayama N, et al. The development of transient hypothyroidism after iodine-131 treatment in hyperthyroid patients with Graves’ disease: prevalence, mechanism and prognosis. Clin Endocrinol (Oxf). 1997;46(1):1–5.PubMedCrossRef
206.
Zurück zum Zitat Bartalena L, Marcocci C, Tanda ML, Manetti L, Dell’Unto E, Bartolomei MP, et al. Cigarette smoking and treatment outcomes in Graves ophthalmopathy. Ann Intern Med. 1998;129(8):632–5.PubMedCrossRef Bartalena L, Marcocci C, Tanda ML, Manetti L, Dell’Unto E, Bartolomei MP, et al. Cigarette smoking and treatment outcomes in Graves ophthalmopathy. Ann Intern Med. 1998;129(8):632–5.PubMedCrossRef
208.
Zurück zum Zitat Li HX, Xiang N, Hu WK, Jiao XL. Relation between therapy options for Graves’ disease and the course of Graves’ ophthalmopathy: a systematic review and meta-analysis. J Endocrinol Invest. 2016;39(11):1225–33.PubMedCrossRef Li HX, Xiang N, Hu WK, Jiao XL. Relation between therapy options for Graves’ disease and the course of Graves’ ophthalmopathy: a systematic review and meta-analysis. J Endocrinol Invest. 2016;39(11):1225–33.PubMedCrossRef
209.
Zurück zum Zitat Tulchinsky M, Brill AB. Spotlight on the association of radioactive iodine treatment with cancer mortality in patients with hyperthyroidism is keeping the highest risk from antithyroid drugs in the blind spot. Clin Nucl Med. 2019;44(10):789–91.PubMedCrossRef Tulchinsky M, Brill AB. Spotlight on the association of radioactive iodine treatment with cancer mortality in patients with hyperthyroidism is keeping the highest risk from antithyroid drugs in the blind spot. Clin Nucl Med. 2019;44(10):789–91.PubMedCrossRef
210.
Zurück zum Zitat Verburg FA, Hoffmann M, Iakovou I, Konijnenberg MW, Mihailovic J, Gabina PM, et al. Errare humanum est, sed in errare perseverare diabolicum: methodological errors in the assessment of the relationship between I-131 therapy and possible increases in the incidence of malignancies. Eur J Nucl Med Mol Imaging. 2019;47(3):519–22.CrossRef Verburg FA, Hoffmann M, Iakovou I, Konijnenberg MW, Mihailovic J, Gabina PM, et al. Errare humanum est, sed in errare perseverare diabolicum: methodological errors in the assessment of the relationship between I-131 therapy and possible increases in the incidence of malignancies. Eur J Nucl Med Mol Imaging. 2019;47(3):519–22.CrossRef
211.
Zurück zum Zitat Hindié E, Ain KB, Zerdoud S, Avram AM. Association of radioactive iodine treatment of hyperthyroidism with cancer mortality: an unjustified warning? J Clin Endocrinol Metab. 2020;105(4):e1901–2.CrossRef Hindié E, Ain KB, Zerdoud S, Avram AM. Association of radioactive iodine treatment of hyperthyroidism with cancer mortality: an unjustified warning? J Clin Endocrinol Metab. 2020;105(4):e1901–2.CrossRef
Metadaten
Titel
The EANM guideline on radioiodine therapy of benign thyroid disease
verfasst von
Alfredo Campennì
Anca M. Avram
Frederik A. Verburg
Ioannis Iakovou
Heribert Hänscheid
Bart de Keizer
Petra Petranović Ovčariček
Luca Giovanella
Publikationsdatum
03.07.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 11/2023
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-023-06274-5

Weitere Artikel der Ausgabe 11/2023

European Journal of Nuclear Medicine and Molecular Imaging 11/2023 Zur Ausgabe