Fact-checked by Grok 2 weeks ago

Hyperthyroidism

Hyperthyroidism is a common endocrine disorder in which the gland produces excessive amounts of , primarily thyroxine (T4) and (T3), leading to an acceleration of the body's metabolic processes and affecting nearly every . This overproduction, also known as an overactive , disrupts normal regulation of , body temperature, digestion, and energy use, resulting in a hypermetabolic state that can cause significant morbidity if untreated. The condition is classified as overt hyperthyroidism when (TSH) levels are low or suppressed alongside elevated T4 and T3, or subclinical when TSH is low but T4 and T3 remain normal. The most prevalent cause worldwide is , an autoimmune condition where antibodies stimulate the to overproduce hormones, accounting for approximately 50-80% of cases in iodine-sufficient regions like the United States. Other key etiologies include and , where autonomous nodules in the gland independently secrete hormones, particularly in older adults or iodine-deficient areas. , an inflammation of the often triggered by viral infections, postpartum changes, or medications like , can also release stored hormones temporarily. Less common triggers involve excessive iodine intake, which overwhelms the gland's regulatory mechanisms, or rare pituitary tumors secreting excess TSH. Risk factors include female sex (affecting women up to 10 times more than men), age over 60, family history of thyroid disorders, recent , and , which exacerbates . Globally, hyperthyroidism impacts about 1-2% of the population, with subclinical forms being more prevalent than overt cases. Symptoms typically develop gradually and include unintentional despite increased appetite, or irregular heartbeat, nervousness, , tremors, excessive sweating, , , , and menstrual irregularities in women. In , patients may also experience eye problems such as bulging eyes () or skin changes like . Older adults might present atypically with , , or unexplained rather than classic hypermetabolic signs. Diagnosis involves blood tests measuring TSH, free T4, and T3 levels, often supplemented by thyroid scans or tests to identify the underlying cause. Untreated hyperthyroidism raises risks for serious complications, including , due to accelerated bone turnover, and, in rare cases, —a life-threatening surge of hormones with high mortality. options are tailored to the cause, severity, and patient factors, encompassing antithyroid drugs like methimazole to inhibit hormone synthesis, beta-blockers for symptom relief, radioactive iodine ablation to destroy overactive thyroid tissue, or surgical in select cases. Early intervention is crucial to prevent long-term cardiovascular and skeletal damage.

Overview

Definition and Classification

Hyperthyroidism is defined as a condition characterized by excessive production of by the itself, resulting in elevated circulating levels of thyroxine (T4) and/or (T3). This overproduction leads to thyrotoxicosis, the clinical syndrome of thyroid hormone excess that manifests as a hypermetabolic state affecting multiple organ systems. In contrast, thyrotoxicosis can occur independently of hyperthyroidism when excess are derived from exogenous sources, such as factitious thyrotoxicosis caused by surreptitious ingestion of thyroid hormone medications, or from the release of preformed hormones in conditions like , where the is inflamed and damaged rather than overactive. Thyroid hormones T3 and T4 play essential roles in regulating , protein synthesis, and , influencing nearly every tissue in the body. Under normal conditions, thyroid function is tightly controlled by the hypothalamic-pituitary-thyroid (HPT) axis: (TRH) from the stimulates the to secrete (TSH), which in turn prompts the gland to synthesize and release T4 and T3; elevated thyroid hormone levels exert on the and pituitary to suppress further TRH and TSH secretion. Hyperthyroidism is classified in several ways to guide and . Based on origin, it is primarily thyroidal (primary hyperthyroidism), arising from intrinsic dysfunction, or rarely central (secondary or tertiary), due to inappropriate TSH secretion from pituitary adenomas or hypothalamic disorders, respectively. By severity, it is categorized as overt hyperthyroidism, marked by suppressed TSH levels alongside elevated free T4 and/or T3, or subclinical hyperthyroidism, featuring low TSH with normal levels, which may be asymptomatic or produce milder effects like subtle . Etiologically, common forms include autoimmune causes such as , autonomous nodules like , and iodine-induced cases such as the , where excess iodine exposure triggers hyperthyroidism in iodine-deficient individuals with underlying nodular .

Pathophysiology

Hyperthyroidism arises from excessive production or release of , primarily thyroxine (T4) and (T3), disrupting the normal hypothalamic-pituitary-thyroid axis. This excess can result from increased synthesis, often driven by stimulation of the (TSH) receptor; accelerated release of preformed hormones due to glandular destruction. In the feedback loop, elevated free T4 and T3 levels suppress TSH secretion via on the pituitary and , typically leading to low or undetectable TSH in overt hyperthyroidism, as described by the relationship TSH ∝ 1 / (T3 + T4) in simplified terms, where hormone levels inversely regulate pituitary output. At the cellular level, thyroid hormone synthesis begins with iodide uptake into thyrocytes via the sodium-iodide symporter (NIS), powered by the sodium-potassium ATPase, followed by transport into the colloid by . Thyroid peroxidase (TPO) then oxidizes iodide to iodine using hydrogen peroxide and catalyzes its incorporation onto tyrosine residues in , forming monoiodotyrosine () and diiodotyrosine (DIT). TPO further facilitates the coupling of these iodotyrosines—MIT with DIT to yield T3, or two DIT molecules to produce T4—before releases the hormones into circulation. Dysregulation, such as constitutive TSH receptor , amplifies these steps, leading to overproduction. Systemically, excess thyroid hormones bind nuclear receptors, upregulating genes that increase by enhancing Na+/K+-ATPase activity, thereby elevating oxygen consumption and heat production across tissues. This mimics beta-adrenergic stimulation, promoting , , and cardiovascular effects like increased , independent of catecholamines. Additionally, T3 stimulates activity via expression, accelerating and reducing bone mineral density. In specific etiologies, such as , TSH receptor-stimulating antibodies (TRAb) chronically activate the receptor, driving autonomous synthesis; toxic adenomas exhibit somatic TSH receptor mutations that confer nodular independence from TSH; and destructive processes in subacute or release stored hormones without new synthesis, causing transient excess.

Clinical Presentation

Signs and Symptoms

Hyperthyroidism typically presents with a range of symptoms resulting from excess thyroid hormone, affecting multiple organ systems and varying in severity based on the degree of hormonal elevation. Common general symptoms include , unintentional despite increased , , nervousness, , and menstrual irregularities in women. Cardiovascular manifestations are prominent and include , , widened , and, in advanced cases, due to increased metabolic demand. Neuromuscular symptoms often involve fine of the hands, proximal (), anxiety, irritability, , and , reflecting heightened sympathetic activity. Dermatological and gastrointestinal features encompass warm, moist skin, excessive sweating, thinning or brittle hair, , increased bowel frequency, and , stemming from accelerated and gastrointestinal motility. Ocular signs, generalizable across causes but more pronounced in , include and retraction, which may contribute to a staring appearance. A goiter, or enlarged , is present in the majority of cases, the most common etiology of hyperthyroidism, often appearing as diffuse neck swelling. In elderly patients, hyperthyroidism may manifest as apathetic hyperthyroidism, with subtler symptoms mimicking , such as weight loss, fatigue, apathy, and withdrawal, rather than classic hypermetabolic features. Symptom intensity generally correlates with elevated free T4 and T3 levels, though extreme exacerbations like represent acute beyond typical presentations.

Thyroid Storm

, also known as thyrotoxic crisis, is a rare but life-threatening endocrine emergency characterized by an acute exacerbation of hyperthyroidism, resulting from a sudden and excessive release of leading to multi-organ dysfunction. It typically occurs in patients with underlying hyperthyroidism, such as , and represents a decompensated state rather than a distinct entity. Common precipitants include infections (the most frequent trigger), (thyroid or non-thyroid), , iodine-containing contrast media, discontinuation of antithyroid therapy, and other stressors like parturition or burns. Clinically, thyroid storm manifests with severe systemic symptoms, including high fever exceeding 102°F (38.9°C), often reaching 104–106°F (40–41.1°C); profound tachycardia greater than 140 beats per minute; and central nervous system alterations ranging from agitation and confusion to delirium or coma. Gastrointestinal involvement is prominent, featuring nausea, vomiting, diarrhea, and occasionally jaundice due to hepatic dysfunction, while cardiovascular complications such as atrial fibrillation, heart failure, or shock may arise. These features reflect a hypermetabolic crisis with heightened sympathetic activity. Diagnosis relies on clinical assessment, as laboratory confirmation of hyperthyroidism alone is insufficient; the Burch-Wartofsky Point Scale provides a standardized scoring system, assigning points for thermoregulatory dysfunction (e.g., 30 points for temperature >104°F), central nervous system effects (e.g., 30 for coma), gastrointestinal symptoms (e.g., 20 for severe ), and cardiovascular issues (e.g., 15 for severe congestive ), among others, with a score greater than 45 indicating high likelihood of and 25–44 suggesting imminent risk. Elevated free thyroxine (T4) and (T3) levels with suppressed support the diagnosis but are not specific to the storm. Pathophysiologically, thyroid storm arises from an abrupt surge in circulating , often triggered by increased release or reduced binding to proteins during acute illness, coupled with enhanced end-organ sensitivity that amplifies hyperactivity, mimicking catecholamine excess and leading to a hyperadrenergic state. This cascade promotes widespread tissue oxygen demand, potentially culminating in multi-organ failure, including hepatic congestion, cardiac arrhythmias, and acute respiratory distress. Despite prompt intervention, mortality from has decreased to approximately 1-6% with modern management as of 2025, primarily due to cardiovascular collapse or infection-related complications. Its incidence is estimated at approximately 1.4 cases per 100,000 persons per year in females and 0.7 per 100,000 in males, with higher rates among hospitalized patients.

Causes

Autoimmune Causes

Autoimmune causes of hyperthyroidism primarily involve dysregulated immune responses targeting the gland, leading to excessive hormone production or release. The most common etiology is , an organ-specific autoimmune disorder characterized by the production of thyroid-stimulating immunoglobulins (TSI or TRAb) that bind to and activate the (TSHR) on thyroid follicular cells, mimicking the action of (TSH) and causing diffuse glandular and hyperfunction. These autoantibodies stimulate cyclic AMP production, promoting thyroid hormone synthesis and secretion, which accounts for 60-80% of hyperthyroidism cases in iodine-sufficient regions worldwide. The pathogenesis of Graves' disease involves both humoral and cellular immunity, with B cells producing the pathogenic TRAb and T cells providing helper functions through release that amplify the autoimmune response. Genetic susceptibility plays a key role, with associations to (HLA) alleles such as HLA-DR3, which influences and T-cell activation in tissue. Environmental factors can trigger or exacerbate the disease in genetically predisposed individuals, including cigarette smoking, which increases risk by promoting and immune dysregulation; , potentially via neuroendocrine pathways; and excess iodine intake, which may enhance autoantigen presentation. Graves' disease exhibits a marked female predominance, with a female-to-male of 5-10:1, likely influenced by estrogen-mediated immune modulation. It is also associated with other autoimmune conditions, such as and , reflecting shared genetic and immunological pathways that heighten overall risk. In some cases, TSHR autoantibodies cross-react with orbital antigens, contributing to extrathyroidal manifestations like ophthalmopathy through shared immunogenic epitopes. Other autoimmune etiologies include transient hyperthyroid phases in conditions like and . Hashitoxicosis represents an initial destructive hyperthyroid state in , where antibody-mediated (anti-thyroid peroxidase or anti-thyroglobulin) inflammation causes follicular disruption and release of preformed thyroid hormones, typically resolving into . , occurring in 5-10% of women within the first year after delivery, involves a similar autoimmune destructive process driven by rebound T-cell and activity following pregnancy-induced immune suppression, often presenting with a hyperthyroid phase due to hormone leakage before progressing to in many cases. These conditions highlight the spectrum of autoimmune thyroiditis, where initial hyperthyroidism stems from glandular destruction rather than stimulation.

Non-Autoimmune Causes

Non-autoimmune causes of hyperthyroidism encompass structural abnormalities in the gland, inflammatory conditions leading to hormone release, and exogenous factors that disrupt normal function. These differ from autoimmune etiologies by lacking antibody-mediated stimulation, instead involving autonomous hormone or leakage of pre-formed hormones. Toxic nodular goiter, also known as , arises from multiple autonomous nodules that independently produce excess , often appearing as "hot" areas on with suppressed uptake in surrounding . This condition is more prevalent in iodine-deficient regions, where the incidence is 1.5-18 cases per 100,000 person-years, compared to lower rates in iodine-sufficient areas. It typically affects older adults and develops gradually from longstanding non-toxic goiter. Toxic adenoma refers to a single hyperfunctioning that autonomously secretes , accounting for approximately 5-10% of hyperthyroidism cases, particularly in iodine-deficient populations. These benign tumors suppress TSH levels and function independently of regulatory signals, leading to clinical hyperthyroidism without systemic . They are more common in women and often present as a palpable solitary nodule. Thyroiditis variants represent inflammatory processes that cause transient hyperthyroidism through destructive release of stored , rather than increased synthesis. , often viral in origin and associated with upper respiratory infections, presents with painful enlargement, fever, and elevated inflammatory markers; it affects women more frequently and resolves spontaneously in most cases. , a painless form, involves lymphocytic infiltration and leakage, commonly seen postpartum but also in non-pregnant individuals without evident . Drug-induced , triggered by agents like or , disrupts follicular integrity; , for instance, can induce type 1 thyrotoxicosis via iodine excess in susceptible glands or type 2 through direct . Exogenous causes stem from external thyroid hormone or iodine overload, mimicking endogenous hyperthyroidism biochemically but with low or absent thyroidal radioiodine uptake. Iatrogenic hyperthyroidism results from excessive dosing in treatment, emphasizing the need for regular TSH monitoring to prevent overdose. Factitious hyperthyroidism, or thyrotoxicosis factitia, involves surreptitious ingestion of thyroid hormone preparations, often for or psychological reasons, and is characterized by suppressed levels. Iodine excess, from supplements, contrast agents, or medications like , can precipitate hyperthyroidism in predisposed individuals with underlying nodular disease, as high iodine loads overwhelm the gland's regulatory mechanisms. Rare non-autoimmune causes include , where markedly elevated (hCG) from molar pregnancies or choriocarcinomas cross-reacts with the TSH receptor, stimulating thyroid hormone production; this can occur in up to 20-50% of complete hydatidiform mole cases with hCG levels exceeding 100,000 IU/L. Rarely, TSH-secreting pituitary adenomas (TSHomas) cause central hyperthyroidism by autonomous TSH production, accounting for less than 1% of cases.

Diagnosis

Laboratory Tests

The diagnosis of hyperthyroidism begins with biochemical confirmation through , typically prompted by symptoms such as unexplained or . The primary screening test is (TSH), which is suppressed in hyperthyroidism due to from excess . A TSH level below 0.1 mU/L is highly suggestive of hyperthyroidism, particularly when accompanied by elevated free thyroxine (T4) or (T3). Overt hyperthyroidism is characterized by low TSH with elevated free T4 and/or total T3 levels, confirming excess thyroid hormone production. In subclinical hyperthyroidism, TSH is low or undetectable while free T4 and T3 remain within normal ranges, often representing an early or mild form of the condition. Some cases, particularly in , exhibit T3-predominant hyperthyroidism, where T3 levels are disproportionately elevated compared to T4. To identify the underlying , assays are essential. (TRAb), including thyroid-stimulating immunoglobulins (TSI), are positive in over 90% of cases and confirm autoimmune stimulation of the . In contrast, anti- peroxidase (anti-TPO) and anti-thyroglobulin (anti-Tg) may be elevated in destructive , indicating autoimmune-mediated damage rather than overproduction. Ancillary laboratory tests provide supportive evidence and assess complications. A complete blood count (CBC) often reveals mild due to increased turnover, while with a left shift may occur in , a severe manifestation of hyperthyroidism. Liver enzymes, such as (ALT) and aspartate aminotransferase (AST), are frequently elevated in 15-76% of untreated cases, reflecting direct effects of excess on hepatic function. Hypercalcemia, resulting from accelerated turnover and resorption, is observed in up to 20% of patients and can contribute to symptoms like . In amiodarone-induced hyperthyroidism, measurement of reverse T3 (rT3) is useful; elevated rT3 levels with high T4 but relatively low T3 help distinguish type 2 (destructive thyroiditis-like) from type 1 (iodine-induced overproduction). These tests collectively guide differentiation from other causes, with further imaging reserved for etiological confirmation.

Imaging and Other Studies

Thyroid scintigraphy serves as a primary imaging modality for evaluating the etiology of hyperthyroidism by assessing gland function and structure through the administration of radiotracers such as (123I) or pertechnetate (99mTc-pertechnetate). In , this test typically reveals diffusely increased radioiodine uptake, often exceeding 30% at 24 hours, reflecting enhanced activity, whereas uptake is low or suppressed in destructive due to impaired hormone synthesis. The scan also distinguishes hyperfunctioning "hot" nodules, indicative of toxic adenomas, from non-functioning "cold" nodules that may warrant further evaluation for . Additionally, whole-body scintigraphy can detect ectopic tissue, such as in , contributing to hyperthyroidism in rare cases. Ultrasound provides detailed anatomical assessment of the , particularly useful for characterizing nodules and evaluating parenchymal changes in hyperthyroidism. In , color often demonstrates markedly increased intrathyroidal vascularity, known as the "thyroid inferno" pattern, which correlates with disease activity. For nodular hyperthyroidism, the Thyroid Imaging Reporting and Data System (TIRADS) scoring system stratifies nodules based on ultrasound features like composition, , margins, calcifications, and shape to estimate malignancy risk and guide decisions. Computed tomography (CT) or (MRI) is employed when evaluating large goiters or retrosternal extension, providing critical information on tracheal compression, vascular involvement, and surgical planning. These modalities are particularly valuable in cases where is limited by anatomy or when assessing compressive symptoms. (PET), typically with 18F-fluorodeoxyglucose (FDG), is rarely indicated but may be used in suspected thyroid malignancy or to evaluate incidentalomas detected on other imaging. Functional tests like (TRH) stimulation, which provoke a blunted or absent (TSH) response in overt hyperthyroidism, have largely been supplanted by more sensitive laboratory assays and are now rarely performed.

Subclinical Hyperthyroidism

Subclinical hyperthyroidism is defined as a persistently suppressed (TSH) level, typically below 0.1 mU/L, accompanied by normal levels of free thyroxine (T4) and (T3). This condition reflects mild thyroid hormone excess without overt clinical symptoms, distinguishing it from manifest hyperthyroidism. Its prevalence in the general population ranges from 0.5% to 2%, with higher rates observed in older adults and regions with . In the United States, subclinical hyperthyroidism affects approximately 0.7% to 1.4% of individuals overall, rising to 1% to 8% among those over 65 years. The condition carries several health risks, particularly in vulnerable populations. It is associated with a 2- to 3-fold increased risk of , especially when TSH is below 0.1 mU/L and in patients over 60 years, contributing to higher rates of cardiovascular events such as coronary heart disease and . health is also affected, with accelerated bone loss and a higher incidence of and fractures in postmenopausal women. Additionally, subclinical hyperthyroidism has been linked to cognitive decline, reduced , and increased overall mortality, though these associations are more pronounced with prolonged duration and lower TSH levels. Recent data emphasize that cardiovascular risks escalate significantly with TSH suppression below 0.1 mU/L, independent of other factors. Progression to overt hyperthyroidism occurs at a rate of 2% to 5% per year on average, though this can reach up to 7% annually in cases with very low TSH or underlying , and is higher among the elderly. Conversely, spontaneous normalization of TSH levels happens in up to 12% of cases per year. Routine screening for subclinical hyperthyroidism is not recommended by major guidelines, including for high-risk groups such as individuals over years; however, if detected through testing prompted by symptoms or other indications, evaluation and management are advised. Management focuses on risk stratification rather than universal treatment. Intervention with antithyroid medications or other therapies is recommended for patients over 65 years with TSH below 0.1 mU/L, or those with comorbidities such as , heart disease, or , to mitigate associated risks. For milder cases (TSH 0.1-0.4 mU/L) or younger patients without symptoms, periodic monitoring of TSH levels every 6 to 12 months is sufficient, with reassessment for progression or complications. This approach balances potential benefits against treatment side effects, guided by clinical guidelines from endocrine societies.

Treatment

Antithyroid Medications

Antithyroid medications (ATDs), also known as thionamides, represent a cornerstone of first-line pharmacological for hyperthyroidism, particularly in cases like , by reversibly inhibiting hormone synthesis to restore euthyroidism. These agents are preferred initially due to their non-ablative nature, allowing for potential remission without permanent ablation. The primary ATDs are methimazole (MMI) and (PTU). Methimazole is the preferred agent in most non-pregnant adults, with an initial dose of 10-30 mg daily, titrated based on clinical response and . is recommended at 300-600 mg daily for severe hyperthyroidism or during the first trimester of , where it is favored over methimazole due to a lower risk of congenital anomalies. After the first trimester, switching to methimazole is often advised to minimize PTU-related risks. Both drugs exert their primary effect by inhibiting , the enzyme essential for iodination of residues and coupling to form thyroxine (T4) and (T3) within the gland. Uniquely, PTU also blocks peripheral deiodination of T4 to the more active T3 by inhibiting 5'-, providing an additional benefit in acute hyperthyroid states like . Treatment regimens involve dose titration to achieve normalization of (TSH) and free T4 levels, typically within 4-8 weeks, followed by dosing. Therapy duration is generally 12-18 months, after which discontinuation is attempted; remission rates in range from 30-50%, with predictors including lower pretreatment TSH receptor antibody levels and smaller goiter size. Adverse effects necessitate vigilant monitoring. Agranulocytosis, a severe reduction in neutrophils, occurs in 0.2-0.5% of patients and requires immediate drug cessation upon symptoms like or fever; routine monitoring is recommended, especially in the first 3 months. Hepatotoxicity is more common with PTU than methimazole, potentially leading to in rare cases, prompting baseline and periodic . Pruritic affects up to 5% of users and often resolves with antihistamines or dose adjustment. Recent studies from 2023-2025 have explored prolonged ATD use beyond , demonstrating and higher sustained remission rates—up to 60% with extended —particularly in patients with fluctuating activity. Additionally, mathematical models integrating patient-specific have been developed to predict free T4 trajectories and optimize dosing, enhancing precision in achieving euthyroidism while minimizing side effects. For patients unsuitable for long-term ATD therapy, radioactive iodine serves as a definitive alternative, though it carries a risk of permanent hypothyroidism. Symptomatic relief with beta-blockers may complement ATDs during .

Radioactive Iodine Therapy

Radioactive iodine therapy, utilizing (¹³¹I), serves as a definitive for hyperthyroidism by ablating overactive tissue. The procedure involves of ¹³¹I in the form of a capsule or liquid, which is selectively taken up by the sodium-iodide symporter in follicular cells. Once absorbed, the isotope emits beta particles that damage and destroy follicles, leading to reduced production over time. This non-invasive approach is typically performed on an outpatient basis, with patients advised to follow radiation safety precautions, such as limiting close contact with others for a few days to weeks post-treatment. Dosing strategies for ¹³¹I therapy include fixed doses, commonly 10-15 mCi (370-555 MBq), or calculated doses based on thyroid gland size, radioiodine uptake, and desired therapeutic outcome. The goal is often to induce in 80-90% of patients, as this ensures complete resolution of hyperthyroidism while allowing straightforward management with replacement. Fixed dosing is simpler and widely used for , while calculated approaches may be preferred for toxic nodules or multinodular goiter to minimize . Indications for radioactive iodine therapy primarily include and toxic thyroid nodules, where it provides a durable cure by permanently reducing thyroid function. It is contraindicated in and due to the risk of fetal or infant thyroid damage, with pregnancy delayed for at least 6-12 months afterward. Pretreatment with antithyroid drugs may be used to deplete thyroid hormone stores and prevent symptom exacerbation during therapy. Common side effects include a transient flare of hyperthyroidism in 5-10% of patients, occurring shortly after due to initial release from damaged cells, as well as inevitable typically onsetting within 2-3 months. Other potential issues encompass inflammation () or dryness from radiation uptake in salivary tissues, and mild neck tenderness managed with analgesics. Long-term, patients require monitoring for , which develops in the majority and necessitates lifelong replacement. Outcomes demonstrate remission of hyperthyroidism in 80-90% of patients after a single dose, with full effects manifesting over 2-6 months. Recent trends favor radioactive iodine over surgical options for most eligible cases, with reserved for approximately 5-10% of patients due to its invasiveness. This therapy's efficacy and safety profile make it a cornerstone of definitive , though repeated doses may be needed in 10-20% of non-responders.

Surgical Interventions

Surgical interventions for hyperthyroidism primarily involve , a procedure that removes part or all of the gland to provide a definitive cure by eliminating the source of excess hormone production. This approach is particularly valuable when medical therapies fail or are contraindicated, offering rapid resolution of symptoms compared to other modalities. is typically performed under general through a transverse incision in the , with careful preservation of the recurrent laryngeal and parathyroid glands to minimize complications. The choice of procedure depends on the underlying cause of hyperthyroidism. For , the most common etiology, total or near-total thyroidectomy is recommended to remove nearly all thyroid tissue, reducing the risk of recurrent hyperthyroidism to less than 1%. In contrast, for a solitary toxic nodule, a —removal of the affected thyroid lobe along with the —may be sufficient, preserving the contralateral lobe to avoid . These operations are ideally conducted by high-volume surgeons, defined as those performing more than 30 thyroidectomies annually, to optimize outcomes. Indications for thyroidectomy include failure or intolerance to antithyroid medications, such as methimazole or ; large goiters causing compressive symptoms like or airway obstruction; suspicion of thyroid malignancy based on ; moderate-to-severe , where radioactive iodine is contraindicated; and patient preference for a swift, permanent resolution of hyperthyroidism. It is also favored in scenarios like with severe hyperthyroidism unresponsive to medications, as it avoids potential fetal risks from prolonged antithyroid drug exposure. Additionally, surgery is considered for young patients or those with contraindications to radioactive iodine, such as desire for future . Preoperative preparation is essential to mitigate risks, beginning with achieving a euthyroid state using antithyroid drugs (e.g., methimazole 10-40 mg daily) combined with beta-blockers (e.g., 10-40 mg three to four times daily) to control symptoms like and prevent ; this typically takes 6 weeks to 3 months. In the 7-10 days prior to surgery, —such as Lugol's solution (5-7 drops three times daily) or saturated solution of (1-2 drops three times daily)—is administered to decrease thyroid vascularity, thereby reducing intraoperative blood loss by up to 40%. Calcium and supplementation may also be given prophylactically to address potential . Antithyroid drugs are discontinued on the day of , while beta-blockers are continued and tapered postoperatively. Complications of thyroidectomy for hyperthyroidism, though generally low in experienced hands, are slightly elevated in due to the gland's increased and . The following table summarizes key complication rates from a large single-center study of 594 patients undergoing total for :
ComplicationTransient RatePermanent Rate
Recurrent laryngeal nerve palsy5.2%0.16%
()40.6%0.5%
(requiring intervention)-0.5%
-1.8%
Other risks include (less than 1%) and , which may necessitate evacuation in up to 1-3% of cases. Transient often resolves with oral calcium and , but permanent requires lifelong management. Following total or near-total , patients require lifelong replacement to maintain euthyroidism. Surgery accounts for approximately 10-30% of definitive treatments for in various cohorts, with higher utilization in cases of severe ophthalmopathy or , where it may be preferred over alternatives for its immediacy and safety profile in these populations. In pregnant patients with refractory hyperthyroidism, is often performed in the second , achieving cure rates near 100% with low maternal and fetal risks when properly prepared.

Adjunctive and Symptomatic Management

Adjunctive therapies in hyperthyroidism primarily target symptom relief and support primary treatments by addressing adrenergic manifestations and accelerating hormone clearance. Beta-blockers, such as , are commonly employed to alleviate symptoms including , , , , and anxiety, providing rapid onset of action within minutes. , a nonselective beta-blocker, is preferred due to its ability to block peripheral conversion of T4 to T3 in addition to controlling and symptoms; typical dosing ranges from 10-40 mg orally three to four times daily, adjusted to 80-320 mg/day based on response, with caution in patients with , COPD, or . Other symptomatic agents include cholestyramine, a that binds in the intestine, reducing serum levels by up to 30% when used as an adjunct in refractory cases. Dosing typically starts at 4 g orally twice daily, increasing to 4 g three times daily for a total of 12 g/day, leading to normalization of free T4 within 12 days in reported cases of iodine-induced hyperthyroidism. Glucocorticoids, such as , are utilized in specific scenarios like or severe thyrotoxicosis to inhibit T4-to-T3 conversion and provide anti-inflammatory effects, with dosing at 40 mg daily for 1-2 weeks followed by taper in . In thyroid storm, a life-threatening exacerbation of hyperthyroidism, management emphasizes supportive measures alongside pharmacotherapy. Supportive care involves cooling with ice packs or blankets, intravenous fluids (dextrose-containing), electrolyte correction, and intensive care monitoring to address hyperthermia, dehydration, and organ dysfunction. The protocol includes beta-blockade with at 60-80 mg orally every 4-6 hours or intravenous 0.5-1 mg boluses, antithyroid drugs like 200 mg every 4 hours followed by iodine (e.g., Lugol's solution 10 drops every 8 hours, delayed at least 1 hour after antithyroid initiation to block hormone release), and glucocorticoids such as 100 mg intravenously every 8 hours for adrenal support and conversion inhibition. Aspirin is avoided as it may displace from binding proteins, exacerbating the condition. Recent guidelines, including the 2016 American Thyroid Association recommendations, stress rapid loading of antithyroid drugs with these adjuncts for optimal outcomes in storm. Dietary modifications support overall by mitigating hormone production and protecting against complications like bone loss. Iodine restriction is advised in cases of excess intake to prevent worsening hyperthyroidism, limiting foods such as iodized salt, , and to under 50 mcg/day if preparing for therapies or in iodine-induced states. Stimulants like from , , and energy drinks should be avoided to reduce anxiety and . For bone health, given the risk of from prolonged hyperthyroidism, supplementation or intake of calcium (e.g., from , ) and (e.g., from fortified non-iodized cereals) is recommended, particularly in at-risk patients.

Emerging Therapies

Recent advances in the treatment of hyperthyroidism, particularly in , have focused on targeted immunotherapies aimed at modulating the underlying autoimmune response rather than solely suppressing thyroid hormone production, building on 2025 guidelines such as those from the Korean Thyroid Association that endorse extended low-dose ATD and fixed-dose radioactive iodine as bridges to novel options. Monoclonal antibodies such as rituximab, which depletes B-cells via anti-CD20 targeting, have shown remission rates of 40-48% in phase 2 trials among patients with low thyrotropin receptor antibody (TRAb) levels or younger demographics, offering a potential alternative for those intolerant to conventional antithyroid drugs. Similarly, teprotumumab, an insulin-like growth factor-1 receptor (IGF-1R) inhibitor primarily approved for thyroid eye disease, has demonstrated secondary benefits on thyroid autoimmunity by significantly reducing thyroid-stimulating immunoglobulin (TSI) from 1.90 IU/L to 0.69 IU/L and TRAb from 3.10 IU/L to 0.60 IU/L in studies of , though effects on thyroid hormones like free T4 and total T3 were variable and not always significant. Biologic agents targeting autoantibody production, particularly FcRn inhibitors, represent a promising class in ongoing trials as of 2025. Batoclimab, a subcutaneous anti-FcRn monoclonal antibody, achieved a 76% response rate (normalization of T3 and T4 without increased antithyroid drug dosing) at week 12 in a phase 2 trial of patients with uncontrolled Graves' hyperthyroidism, with 80% maintaining normal thyroid function and approximately 47% achieving antithyroid drug-free remission at six months post-treatment. Pivotal phase 3 trials for a next-generation agent, IMVT-1402, began enrollment in late 2024 with topline data expected in 2027. Efgartigimod, another FcRn inhibitor, is in phase 3 evaluation primarily for thyroid eye disease but shows potential for broader autoimmune thyroid modulation by reducing pathogenic IgG autoantibodies, though specific hyperthyroidism remission data remain limited to preclinical and early orbital studies. Veligrotug (VRDN-001), an IGF-1R antagonist similar to teprotumumab, met all primary endpoints in phase 3 trials for active thyroid eye disease in 2025, with a Biologics License Application submitted to the FDA in November 2025; it has indirect implications for associated hyperthyroidism through autoantibody reduction, but dedicated hyperthyroidism trials are not yet reported. Non-invasive ablation techniques, such as high-intensity focused ultrasound (HIFU), are under investigation for managing hyperfunctioning thyroid nodules contributing to hyperthyroidism, providing an outpatient alternative to . Systematic reviews indicate HIFU achieves a 75.8% success rate for benign nodule reduction at 6 months, with average volume decreases of 48.55% at 6 months and 55.02% at 12 months, and minimal complications like transient voice changes. Emerging enhancements, including thyroid-targeted nano-bombs (PSAPI) that encapsulate and for improved precision, have shown in preclinical models a significant increase in necrotic area and reduced relapse rates to 41.3% compared to standard HIFU, with high and lowered post-treatment inflammation. Other investigational approaches include TSH receptor (TSHR) and concepts to directly address autoimmune . The monoclonal TSHR K1-70 demonstrated symptom improvement without major adverse events in a phase 1 trial, while small-molecule variants like ANTAG-3 remain preclinical but show thyroid hormone reduction in animal models. For , antigen-specific immunotherapies like ATX-GD-59 achieved 50% normalization of T3 levels in a phase 1 trial, and preclinical TSHR-targeted CAR-T cells have eliminated TRAb-producing B-cells in mouse models, highlighting potential for durable remission but requiring further human validation. These therapies collectively aim for faster, etiology-specific remission rates of 30-50% in early data, surpassing the approximately 30% long-term remission with standard antithyroid drugs in recent 2024-2025 reviews.

Complications and Prognosis

Long-Term Complications

Untreated or inadequately managed hyperthyroidism can lead to various long-term organ-specific complications, primarily affecting the cardiovascular, skeletal, and ocular systems, with additional risks to muscle and structures. These sequelae arise from the sustained effects of excess on and function, and while often mitigates risks, some persist depending on disease duration and patient factors. Cardiovascular complications are among the most significant, with occurring in 10-15% of patients and carrying a of persistence even after achieving euthyroidism, particularly if has been prolonged. Excess thyroid hormone induces through increased and contractility, which can progress to and chronic in advanced cases, with an associated 60% higher mortality if untreated. Recent studies also link subclinical hyperthyroidism to a 20-80% increased incidence of cardiovascular morbidity and mortality, underscoring the need for early intervention. Skeletal effects manifest as accelerated and resorption, leading to and accelerated loss, particularly affecting postmenopausal women who face a higher risk. This loss contributes to reduced and increased fragility, with rates elevated due to the imbalance in bone turnover. In patients with , ocular complications such as develop in 20-30% of cases, featuring proptosis and from orbital inflammation and extraocular muscle involvement. significantly exacerbates the incidence and progression of this condition, increasing the risk up to sevenfold and impairing treatment response. Other complications include a low risk (3-5%) of in thyroid nodules associated with hyperthyroidism, necessitating evaluation for hyperfunctioning nodules. Proximal is common, but progression to remains rare, with only a handful of reported cases linked to thyrotoxicosis. Following radioactive iodine therapy, post-treatment develops in most patients within the first year, requiring regular monitoring of thyroid function every 4-6 weeks to initiate replacement promptly.

Prognosis and Outcomes

The prognosis of hyperthyroidism is generally favorable with appropriate treatment, with most patients achieving control of symptoms and normalization of thyroid function, though the specific outcomes depend on the underlying cause, such as or toxic nodular goiter, and the chosen therapy. Remission rates with antithyroid drugs (ATDs) typically range from 30% to 50%, but relapse occurs in approximately 50% of cases after discontinuation of therapy. In contrast, radioactive iodine () therapy and surgical interventions offer near-complete cure rates for hyperthyroidism, approaching 90-100%, though they often result in requiring lifelong thyroid hormone replacement. Several factors influence the likelihood of successful remission, particularly with ATDs; younger age and smaller goiter size are associated with better outcomes, while significantly worsens the prognosis for associated by increasing disease severity and reducing treatment efficacy. Recent 2024 studies indicate that 80-90% of patients achieve euthyroid status at one year following therapy, though up to 90% of those undergoing ablation require lifelong replacement due to induced . Emerging data on updated remission rates with novel adjunctive therapies, such as targeted immunomodulators, suggest potential improvements over traditional ATDs, addressing previous gaps in long-term control. Overall mortality is low, less than 1% in treated patients, with accounting for the majority of fatalities when it occurs, though prompt intervention reduces its impact. Cardiovascular events represent the primary long-term risk, particularly in untreated or inadequately managed cases. Post-remission monitoring includes annual TSH assessments to detect early, along with evaluations in at-risk populations, such as postmenopausal women, to mitigate from prior hyperthyroid effects.

Special Populations

Pregnancy

Hyperthyroidism during pregnancy is uncommon, with an incidence of 0.1% to 0.4% among pregnant individuals. In the first , mild hyperthyroidism-like changes are physiologically normal due to (hCG), which peaks around 8 to 10 weeks and weakly stimulates the gland via structural similarity to (TSH), suppressing TSH levels to 0.1 to 0.4 mU/L and mildly elevating free thyroxine (FT4) levels; these changes typically resolve by mid-pregnancy. Gestational transient thyrotoxicosis, affecting 1% to 3% of pregnancies and often linked to , must be distinguished from true hyperthyroid disease such as , as the former is self-limited and does not require antithyroid treatment. Untreated or poorly controlled , the most common cause of overt hyperthyroidism in (occurring in about 0.2% of cases), increases maternal risks including (up to twofold higher), , , , and . Fetal and neonatal risks include , , goiter, prematurity, and neonatal hyperthyroidism in 1% to 5% of cases, primarily due to transplacental passage of maternal thyroid-stimulating immunoglobulins (TRAb). Diagnosis involves clinical assessment, trimester-specific (suppressed TSH with elevated FT4 confirming overt disease), and TRAb measurement in the first trimester for those with known or suspected , with repeat testing at 18 to 22 weeks and 30 to 34 weeks if initially elevated to predict fetal . Radioactive iodine uptake scans and therapy are contraindicated due to fetal . Management prioritizes antithyroid drugs (ATDs), with (PTU) preferred in the first due to lower teratogenic compared to methimazole (3% versus 5% rate), followed by a switch to methimazole in the second and third if needed; the lowest effective dose is used to maintain FT4 in the upper normal range, with TSH levels that may remain suppressed. Beta-blockers such as may be used cautiously and short-term for symptomatic relief of or tremors until ATDs take effect, avoiding long-term use due to potential fetal growth restriction. For severe cases unresponsive to ATDs or with allergies, is considered in the second . The 2017 American Association (ATA) guidelines emphasize shared decision-making on ATD choice; liver function should be assessed if clinical suspicion of arises. Neonates of mothers with require monitoring for hyperthyroidism, particularly if maternal TRAb levels are high.

Other Animals

Hyperthyroidism is the most common endocrine disorder in , particularly affecting animals over 10 years of age, with a prevalence of approximately 10% in this population. In felines, the condition is primarily caused by adenomatous of the , leading to excessive production of . Clinical signs include progressive despite increased appetite, and , and potentially , which can manifest as heart murmurs or gallop rhythms. Among , the incidence ranges from 1% to 3%, with certain breeds such as showing a predisposition, possibly due to genetic factors influencing . In , hyperthyroidism is rare compared to , often resulting from in about 50% of cases or functional adenomas, with multinodular being less common than in felines. Signs are similar to those in , including , , and increased thirst, but frequently present with palpable masses due to the more pronounced enlargement of the gland. Diagnosis in both species relies on measuring elevated total thyroxine (T4) levels in , often confirmed by using pertechnetate to visualize hyperfunctional tissue. Treatment options mirror those used in humans, such as methimazole to inhibit hormone synthesis, surgical , or radioactive iodine () therapy, which is particularly preferred in for its high efficacy and non-invasive nature, achieving cure rates of up to 95% with a single dose. Recent advancements in veterinary protocols, including optimized dosing based on 2023 studies on body weight and renal function, have improved outcomes by reducing incidence to under 15% post-treatment. Unlike in humans, where drives autoimmune hyperthyroidism, the condition in cats and dogs is predominantly nodular or neoplastic, highlighting etiological differences that influence therapeutic approaches.

Epidemiology and History

Epidemiology

Hyperthyroidism affects approximately 1-2% of the global population, with overt cases comprising about 0.5% and subclinical cases ranging from 0.7% to 1.7%. The condition is significantly more prevalent in women, with a female-to-male of 5-10:1, reflecting hormonal and genetic influences that predispose females to autoimmune thyroid disorders like , the leading cause. In the United States, the and Nutrition Examination Survey (NHANES III) estimated an overall prevalence of 1.3%, including 0.5% overt and 0.7% subclinical hyperthyroidism. The annual incidence of , the most common etiology, ranges from 20 to 50 cases per 100,000 population in iodine-sufficient regions. Incidence of toxic nodular goiter varies by iodine status, occurring at 3-6 cases per 100,000 per year in iodine-replete areas and rising to 20-40 cases per 100,000 in iodine-deficient regions.02016-0/fulltext) Recent data from 2024 indicate an increasing trend in hyperthyroidism incidence in iodine-sufficient populations, potentially linked to improved diagnostics and environmental factors. Key risk factors include advanced age, with prevalence doubling in individuals over 60 years compared to younger adults. Female sex amplifies susceptibility, while smoking doubles the odds of developing ( 2.5). Iodine excess in replete areas heightens risk for toxic nodules, and genetic factors contribute substantially, with family history conferring up to a 30% increased familial risk through estimated at 79% for . Geographically, toxic nodular goiter is more prevalent in iodine-deficient and , while overt hyperthyroidism prevalence is 0.75% in and 0.78% in . In the , NHANES data confirm a 1.2% overall prevalence. Ethnically, rates are higher among Asians and than in Caucasians. Post-2020 trends show rising subclinical cases, possibly influenced by COVID-19-related thyroid disruptions. Recent studies from 2023-2025 link hyperthyroidism to elevated cardiovascular risk, with prevalence up to 17.5% among acute patients.

History

The earliest descriptions of hyperthyroidism emerged in the early , with English physician Caleb Hillier Parry providing the first detailed account of exophthalmic goiter in a posthumously published work in 1825, based on clinical observations dating back to 1786.00769-7/fulltext) In 1835, Irish physician independently described the condition in a series of clinical lectures, highlighting the triad of goiter, , and in female patients, which he termed a "newly observed affection of the thyroid gland." Shortly thereafter, in 1840, German physician Carl Adolph von Basedow reported similar cases in Europe, emphasizing the systemic effects including eye protrusion and cardiac symptoms, leading to the eponym "Basedow's disease" on the continent.00769-7/fulltext) Key milestones in understanding hyperthyroidism followed in the early , including the isolation of thyroxine by Edward Calvin Kendall at the on December 25, 1914, which confirmed the role of in metabolic regulation and paved the way for hormone-based therapies. Surgical interventions, such as subtotal thyroidectomy, had become the primary treatment by the late 1800s, refined by pioneers like Theodor Kocher, but carried high risks of mortality and complications until preoperative iodine preparation was introduced in the 1920s.00769-7/fulltext) The advent of medical therapies transformed management in the 1940s, with antithyroid drugs (ATDs) like derivatives discovered through wartime research on sulfur compounds, enabling non-surgical control of thyroid overactivity and gaining widespread adoption post-World War II in the late 1940s and 1950s. A pivotal advancement was radioactive iodine (RAI) therapy, pioneered by American endocrinologist Saul Hertz, who administered the first therapeutic dose to a patient with in 1941, leveraging iodine's selective uptake by the to ablate overactive tissue—a method that became standard in the . In the 1970s, the discovery of thyroid receptor antibodies (TRAb), particularly thyroid-stimulating immunoglobulins (TSI), provided insight into the autoimmune basis of the disease, with studies in 1974 demonstrating that Graves' IgG competed with TSH for receptor binding, shifting paradigms toward immune-targeted diagnostics. The evolution of hyperthyroidism treatment has progressed from predominantly surgical approaches in the late 19th and early 20th centuries, which addressed goiter but not underlying , to multifaceted medical options by mid-century, including ATDs for reversible control and for definitive ablation.00769-7/fulltext) This shift reduced operative risks and improved outcomes, though early applications highlighted gaps in trial ethics, such as inadequate follow-up on effects.

References

  1. [1]
    Hyperthyroidism - Symptoms and causes - Mayo Clinic
    Nov 30, 2022 · Hyperthyroidism happens when the thyroid gland makes too much thyroid hormone. This condition also is called overactive thyroid.
  2. [2]
    Hyperthyroidism (Overactive Thyroid) - NIDDK
    Hyperthyroidism, an overactive thyroid gland, can speed up your body's functions. Learn about its causes, complications, symptoms, diagnosis, and treatment.
  3. [3]
    Hyperthyroidism - StatPearls - NCBI Bookshelf - NIH
    Hyperthyroidism is a common thyroid disorder with multiple underlying etiologies. This disease is characterized by excess thyroid hormone production.Continuing Education Activity · Treatment / Management · Differential Diagnosis
  4. [4]
    Thyrotoxicosis - StatPearls - NCBI Bookshelf - NIH
    Jan 18, 2025 · Thyrotoxicosis is a clinical condition characterized by excessive thyroid hormone activity, primarily triiodothyronine (T 3 ) and thyroxine (T 4 ).Thyrotoxicosis · Etiology · Treatment / Management
  5. [5]
    Physiology, Thyroid Stimulating Hormone - StatPearls - NCBI - NIH
    May 1, 2023 · Primary thyroid disease refers to problems arising from the thyroid gland itself. In contrast, secondary thyroid disease refers to central ...Missing: classification | Show results with:classification
  6. [6]
    The Pathology of Hyperthyroidism - PMC - PubMed Central - NIH
    Dec 3, 2018 · Hyperthyroidism is a clinical syndrome characterized by hypermetabolic state due to the increased free serum thyroxine (T4) and/or free ...Toxic Goiter · Drug Associated... · Amiodarone Associated...<|control11|><|separator|>
  7. [7]
    Jod-Basedow Syndrome - StatPearls - NCBI Bookshelf - NIH
    Jod-Basedow syndrome, also known as iodine-induced hyperthyroidism, is a rare cause of thyrotoxicosis typically seen after administering exogenous iodine.Continuing Education Activity · Etiology · Pathophysiology · Differential Diagnosis
  8. [8]
    Physiology, Thyroid Hormone - StatPearls - NCBI Bookshelf
    Jun 5, 2023 · Thyroid hormone increases the basal metabolic rate. It increases the gene expression of Na+/K+ ATPase in different tissues leading to increased ...
  9. [9]
    Hyperthyroidism: A Review - PMC - PubMed Central
    Apr 17, 2024 · Hyperthyroidism affects 2.5% of adults worldwide and is associated with osteoporosis, heart disease, and increased mortality.
  10. [10]
    Overview of the clinical manifestations of hyperthyroidism in adults
    Oct 10, 2024 · Outline · Bone · Serum lipids · Hyperglycemia · Adrenal function · Gonadal function · Respiratory · Gastrointestinal · Thymic enlargement ...
  11. [11]
    Graves Disease - StatPearls - NCBI Bookshelf - NIH
    Graves' disease is an autoimmune disease which primarily affects the thyroid gland. It may also affect multiple other organs including eyes and skin.
  12. [12]
    Thyroid Storm - StatPearls - NCBI Bookshelf
    A thyroid storm, also known as a thyrotoxic crisis, is an acute, life-threatening complication of hyperthyroidism that presents with multi-system involvement.
  13. [13]
    Thyroid Storm: Clinical Manifestation, Pathophysiology, and Treatment
    Thyroid Storm: Clinical Manifestation, Pathophysiology, and Treatment. Written By. Rahul Pandey, Sanjeev Kumar and Narendra Kotwal. Submitted: 06 July 2019 ...
  14. [14]
    How common is thyroid storm and which groups have the highest ...
    The estimated occurrence of thyroid storm was 1.4 cases per 100,000 people per year in females and 0.7 cases per 100,000 people per year in males.
  15. [15]
    Graves' Disease Mechanisms: The Role of Stimulating, Blocking ...
    Stimulating antibodies bind only to the naturally conformed TSHR and compete for TSH binding to the receptor site. These antibodies induce cyclic AMP generation ...
  16. [16]
    Hyperthyroidism: Diagnosis and Treatment - AAFP
    Aug 15, 2005 · Graves' disease is the most common cause of hyperthyroidism, accounting for 60 to 80 percent of all cases. It is an autoimmune disease caused ...
  17. [17]
    Characterizing the Interplay of Lymphocytes in Graves' Disease - PMC
    Apr 6, 2023 · The disease is primarily mediated by B cells, which produce autoantibodies against the thyroid-stimulating hormone receptor (TSHR), chronically stimulating it.
  18. [18]
    Role of the T and B lymphocytes in pathogenesis of autoimmune ...
    Feb 13, 2018 · In Graves' Disease, B cells play a vital role as they are the source of pathognomonic activating autoantibodies (TRAb) against thyroid ...
  19. [19]
    Association of HLA-DR and -DQ genes with Graves disease in ...
    Graves disease (GD) is an autoimmune thyroid disease and is associated with human leukocyte antigen (HLA)-DR3 and DQA1*0501 in Caucasians.
  20. [20]
    Smoking and Other Lifestyle Factors and the Risk of Graves ...
    Jul 25, 2005 · Postulated environmental and lifestyle risk factors include cigarette smoking, stress and adverse life events, and high dietary iodine intake.
  21. [21]
    Environmental Issues in Thyroid Diseases - Frontiers
    Radiation exposure, and increased iodine intake, selenium, and vitamin D deficiency are environmental factors increasing the AITD risk. GO and GD are associated ...Environmental Factors · Drugs · Heavy Metals
  22. [22]
    role of TRAbs | BMC Endocrine Disorders
    Dec 21, 2024 · GD affects women more often, with a female: male ratio of around 5–10:1 [1]. While reliable regional data from other countries in the South ...
  23. [23]
    Evaluating and managing patients with thyrotoxicosis - RACGP
    Graves disease, TSH receptor Ab increases thyroid hormone production and causes thyroid hyperplasia, Female:male ratio 5–10:1. Peak onset 40–60 years. Diffuse ...
  24. [24]
    The association of other autoimmune diseases in patients with ...
    A significant percentage (16.7%) of GD patients had another associated autoimmune disease such as vitiligo, autoimmune gastritis, rheumatoid arthritis, ...
  25. [25]
    Thyroid associated orbitopathy - PMC - NIH
    Immunoglobulins from patients with Graves' disease induce hyaluronan synthesis in their orbital fibroblasts through the self-antigen, insulin-like growth factor ...
  26. [26]
    Hashimoto Thyroiditis - StatPearls - NCBI Bookshelf
    Feb 9, 2025 · Hashimoto thyroiditis is an autoimmune disease that destroys thyroid follicular cells through cell- and antibody-mediated immune processes.
  27. [27]
    Prolonged Duration of Hashitoxicosis in a Patient with Hashimoto's ...
    Jun 14, 2018 · Hashitoxicosis is the initial hyperthyroid phase of patients with Hashimoto's thyroiditis and, usually, this phase lasts for one to two ...
  28. [28]
    Postpartum Thyroiditis - StatPearls - NCBI Bookshelf - NIH
    Postpartum thyroiditis (PPT) is a destructive autoimmune disease occurring in the first year after delivery in women without a history of thyroid disease ...Continuing Education Activity · Introduction · Evaluation · Treatment / Management
  29. [29]
    Thyroiditis: Evaluation and Treatment - AAFP
    Postpartum thyroiditis is an autoimmune-mediated destructive thyroiditis ... phase includes Graves disease, which can also present during the postpartum period.Hashimoto Thyroiditis (Chronic... · Postpartum Thyroiditis · Subacute Thyroiditis
  30. [30]
    Plummer Disease - StatPearls - NCBI Bookshelf - NIH
    Mar 11, 2023 · Plummer disease, or toxic multi-nodular goiter (MNG), is a hormonally active MNG with hyperthyroidism.
  31. [31]
    Toxic Thyroid Adenoma Presenting as Apathetic Hyperthyroidism
    May 29, 2024 · The overall prevalence of toxic adenomas is between 5-7%, accounting for approximately 1-2% of all hyperthyroid cases in men and women [10]. It ...
  32. [32]
    Hyperthyroidism in Aging - Endotext - NCBI Bookshelf - NIH
    Aug 9, 2021 · A relatively rare cause is the presence of a single toxic adenoma - a benign tumor exhibiting autonomous secretion of thyroid hormones. This ...
  33. [33]
    Subacute Thyroiditis - Endotext - NCBI Bookshelf - NIH
    Jun 12, 2018 · Subacute thyroiditis (SAT) is an inflammatory condition of the thyroid with characteristic presentations and clinical course.CLINICAL RECOGNITION · PATHOPHYSIOLOGY · DIAGNOSIS AND... · THERAPY
  34. [34]
    Thyroiditis - StatPearls - NCBI Bookshelf
    Drug-induced thyroiditis: The most common drugs causing thyroid dysfunction are amiodarone, lithium, and tyrosine kinase inhibitors. Amiodarone can cause both ...
  35. [35]
    Amiodarone-induced thyrotoxicosis: A review - PMC - NIH
    Amiodarone-induced thyrotoxicosis (AIT) develops in 3% of amiodarone-treated patients in North America. AIT is classified as type 1 or type 2.
  36. [36]
    Hyperthyroidism and human chorionic gonadotrophin production in ...
    It is thought that the HCG produced in woman with GTD has enhanced thyrotrophic activity compared with HCG in normal pregnancy.
  37. [37]
    Gestational Trophoblastic Disease - StatPearls - NCBI Bookshelf
    Gestational trophoblastic disease (GTD) is a group of tumors defined by abnormal trophoblastic proliferation involving both benign and malignant entities.
  38. [38]
    Thyroid Function Tests
    The best way to initially test thyroid function is to measure the TSH level in a blood sample. Changes in TSH can serve as an “early warning system.”
  39. [39]
    Subclinical Hyperthyroidism: When to Consider Treatment - AAFP
    Jun 1, 2017 · The ATA recommends treating patients with TSH levels persistently less than 0.1 mIU per L if they are 65 years or older; if they are younger ...
  40. [40]
    Hyperthyroidism: Diagnosis and Treatment - AAFP
    Mar 1, 2016 · The diagnostic workup for hyperthyroidism includes measuring thyroid-stimulating hormone, free thyroxine (T4), and total triiodothyronine (T3) ...
  41. [41]
    Hyperthyroidism | American Thyroid Association
    ### Diagnostic Lab Tests for Hyperthyroidism
  42. [42]
    Thyroid Antibodies: MedlinePlus Medical Test
    Aug 7, 2023 · This test looks for thyroid antibodies in your blood. They can be a sign of a thyroid disorder, including Graves or Hashimoto's disease.
  43. [43]
    Hyperthyroidism | Annals of Internal Medicine - ACP Journals
    Jul 3, 2012 · Other common laboratory abnormalities include anemia, leukocytosis, hyperglycemia, azotemia, hypercalcemia, and elevated liver enzymes. Under ...
  44. [44]
    Hyperthyroidism and Liver Dysfunction: A Review of a Common ...
    Feb 7, 2022 · Deranged liver enzymes due to hyperthyroidism rather than intrinsic liver pathology are not uncommon. The reported prevalence of liver ...
  45. [45]
    An Unusual Case of Hypercalcemia Associated with Graves ... - NIH
    Asymptomatic serum calcium elevation has been documented in up to 20% of patients with hyperthyroidism and is related to increased bone resorption. We ...
  46. [46]
    Amiodarone and thyroid dysfunction - UpToDate
    May 20, 2024 · Amiodarone-induced thyrotoxicosis · - Clinical manifestations ... reverse T3 accumulates since it is not metabolized to T2 [3].
  47. [47]
    Thyroid Uptake and Scan - StatPearls - NCBI Bookshelf - NIH
    Oct 3, 2022 · The thyroid uptake and scan is a radiologic diagnostic tool used to determine thyroid function and pathologies.Introduction · Procedures · Potential Diagnosis · Normal and Critical Findings
  48. [48]
    The Role of Nuclear Medicine in the Clinical Management of Benign ...
    Mar 1, 2021 · Thyroid scintigraphy with either radioiodine or 99m Tc-pertechnetate is useful to characterize different forms of hyperthyroidism and provides information for ...
  49. [49]
    Significance of multiple uptake measurements in I-123 thyroid ...
    Normal 24 hour uptake values have a range of 10-30%. Hypothyroidism is considered any percentage less than 10; whereas uptake values greater than 30 indicate ...
  50. [50]
    Scintigraphic Manifestations of Thyrotoxicosis1 - RSNA Journals
    We recommended to the patient that any treatment for Graves disease be deferred pending repeated thyroid function testing 4-6 weeks later to definitely exclude ...
  51. [51]
    Struma Ovarii With Hyperthyroidism - Clinical Nuclear Medicine
    In patients with extrathyroidal hyperthyroidism, 123I WBS with SPECT/CT imaging of the pelvis provides accurate identification of ectopic thyroid tissue for ...
  52. [52]
    Ultrasonography of the Thyroid - Endotext - NCBI Bookshelf - NIH
    Apr 11, 2020 · In one investigation, the American College of Radiology TIRADS structured reporting improved the “quality” of thyroid ultrasound reports.
  53. [53]
    Thyroid inferno | Radiology Reference Article | Radiopaedia.org
    Nov 9, 2023 · Thyroid inferno refers to the color Doppler appearance of the thyroid gland in active Graves disease (inclusive of variants such as Marine-Lenhart syndrome)
  54. [54]
    Thyroid Imaging Reporting and Data System (TI-RADS)
    Jun 27, 2025 · TI-RADS refers to any of several risk stratification systems for thyroid lesions, usually based on ultrasound features, with a structure modelled off BI-RADS.
  55. [55]
    Risk Stratification of Thyroid Nodules: From Ultrasound Features to ...
    Jan 30, 2022 · The TIRADS classification is a point scale that categorizes the US of thyroid nodules from low to high suspicion, based on the number and ...
  56. [56]
    Thyroid computed tomography imaging: pictorial review of variable ...
    In this pictorial review, we cover a wide spectrum of common and uncommon, incidental and non-incidental thyroid findings from CT scans.
  57. [57]
    Computed Tomography and Magnetic Resonance Imaging of the ...
    CT imaging of a retrosternal goiter to identify the extent of tracheal compression and deviation.
  58. [58]
    PET/CT in the Management of Thyroid Cancers | AJR
    CT and MRI are useful in the workup for locally advanced thyroid cancers to assess tumor extension into surrounding structures and cervical lymph node ...
  59. [59]
    jnumed.120.251504.full.pdf - Journal of Nuclear Medicine
    Feb 12, 2021 · Focal 18F-FDG uptake in the thyroid gland can be found in nodular goiter, and about 35% of these “incidentalomas” are thyroid cancer (32).
  60. [60]
    TRH Stimulation When Basal TSH is Within the Normal Range
    ... TRH is no longer used commonly for the detection of primary hypothyroidism or hyperthyroidism. The reason has been that this test has been replaced by the ...
  61. [61]
    The Differential Diagnosis of Discrepant Thyroid Function Tests
    Jul 6, 2020 · ... hyperthyroidism can cause a blunted response to TRH. For example, one RTHβ patient with an underlying “warm” thyroid nodule showed a blunted ...
  62. [62]
    Management of Subclinical Hyperthyroidism - PMC - PubMed Central
    Grade I subclinical hyperthyroidism is 3 to 4 times more common than Grade II subclinical hyperthyroidism. The risk of progression from Grade I to overt ...Missing: secondary | Show results with:secondary
  63. [63]
    [PDF] THYROID - Endocrine Society
    Prevalence of overt and subclinical hyperthyroidism by race/ethnicity in the US. RACE/ETHNICITY. HYPERTHYROIDISM. OVERALL. OVERT. SUBCLINICAL. All. 1.3%. 0.5%.
  64. [64]
    A Stepwise Approach to the Evaluation and Treatment of ...
    The clinical consequences of subclinical hyperthyroidism such as higher mortality, atrial dysrhythmia, accelerated bone loss, and fractures are age and severity ...
  65. [65]
    Approach to the Patient with Subclinical Hyperthyroidism
    Subclinical hyperthyroidism has been associated with an increased risk of atrial fibrillation and mortality, decreased bone mineral density in postmenopausal ...
  66. [66]
    A systematic review of subclinical hyperthyroidism guidelines - NIH
    Jun 13, 2024 · Progression to overt hyperthyroidism occurs in up to 7% of patients per year, and reversion to normal occurs in up to 12% of patients per year ( ...
  67. [67]
    2022 Update on Clinical Management of Graves' Disease and ... - NIH
    Methimazole (MMI) should be used in patients who choose ATD therapy for GD, except (a) during the first trimester of pregnancy when propylthiouracil (PTU) is ...
  68. [68]
    Most patients with Graves' disease treated with antithyroid drugs ...
    Of the patients who relapsed, 49% had a second course of ATDs and of those, 29% achieved remission. Of all patients treated with ATDs who remained in remission ...
  69. [69]
    Propylthiouracil (PTU) - StatPearls - NCBI Bookshelf
    Propylthiouracil is an anti-thyroid drug used to manage Graves disease and hyperthyroidism. This activity will highlight the mechanism of action, adverse event ...
  70. [70]
    Management of hyperthyroidism during pregnancy and lactation
    Both methimazole (MMI) and propylthiouracil (PTU) may be used during pregnancy; however, PTU is preferred in the first trimester and should be replaced by MMI ...
  71. [71]
    Methimazole - StatPearls - NCBI Bookshelf - NIH
    Sep 13, 2023 · Methimazole is an antithyroid medication used to treat hyperthyroidism and is categorized within the thioamide drug class.
  72. [72]
    2018 European Thyroid Association Guideline for the Management ...
    The initial dose of MMI is usually 10–30 mg once daily depending on the severity of hyperthyroidism (CBZ 15–40 mg/day). PTU is given at a dose of 100 mg every 8 ...
  73. [73]
    Graves' Disease: Can It Be Cured? - Endocrinology and Metabolism
    Mar 21, 2019 · The incidence is 20 to 30 cases per year per 100,000 persons [23]. Approximately 3% of women and 0.5% of men develop Graves' disease during ...Thyroidectomy · Radioactive Iodine · Antithyroid Drugs<|control11|><|separator|>
  74. [74]
    Increased Risk of Antithyroid Drug Agranulocytosis Associated with ...
    Background: Agranulocytosis occurs in 0.2-0.5% of patients treated with the antithyroid drugs (ATDs) methimazole and propylthiouracil. The objectives of ...
  75. [75]
    Antithyroid drug-related hepatotoxicity in hyperthyroidism patients
    The examined hepatotoxicity consisted of cholestasis, non-infectious hepatitis, acute liver failure and liver transplant, with the incidences and relative risks ...
  76. [76]
    Antithyroid drugs - Knowledge @ AMBOSS
    May 8, 2023 · Propylthiouracil also lowers peripheral conversion of T4 to T3 by inhibiting 5'-deiodinase. Inhibit proteolytic cleavage of T3 and T4 from ...
  77. [77]
    A short review of current knowledge regarding long-term treatment ...
    Dec 5, 2024 · Compared to the conventional treatment, long-term antithyroid treatment has been associated with a higher chance of Graves' disease remission.<|separator|>
  78. [78]
    Mathematical Modeling of Free Thyroxine Concentrations During ...
    May 31, 2022 · Our mathematical model produced valid FT4 predictions during MMI treatment in GD and provided the basis for the DigiThy application already ...
  79. [79]
    Hyperthyroidism treatment and risk for heart problems
    Methimazole is used to treat hyperthyroidism, especially when it is caused by Graves' disease. Propylthiouracil (PTU): an antithyroid medication that blocks the ...
  80. [80]
    Radioactive Iodine Therapy - StatPearls - NCBI Bookshelf
    Radioactive iodine therapy is used in the management and treatment of hyperthyroidism. This activity describes the indications, actions, and contraindications ...
  81. [81]
    Guidelines for the Use of Radioiodine in the Management of ... - NIH
    Radioiodine ( 131 I) therapy is indicated in patients with nearly all causes of hyperthyroidism. It may safely be given to patients of all age groups.
  82. [82]
    2025 Korean Thyroid Association Management Guidelines for ...
    Jun 24, 2025 · A fixed dose of 10 to 15 mCi is recommended. Although a strict low-iodine diet is unnecessary, iodine-rich foods should be avoided for at least ...
  83. [83]
    Radioactive Iodine | American Thyroid Association
    RAI is a safe and effective treatment for thyroid problems. If you get RAI for your hyperthyroidism, you may become hypothyroid.
  84. [84]
    Radioiodine I-131 For The Therapy Of Graves' Disease - PMC - NIH
    The aim of therapy is to treat hyperthyroidism by destroying sufficient thyroid tissue to render the patient either euthyroid or hypothyroid. Iodine-131 is a ...
  85. [85]
    Surgical Management of Graves' Disease
    Thyroidectomy should be considered for anyone with Graves' disease (GD) and moderate-to-severe eye disease, or smokers with GD due to increased risk of ...
  86. [86]
    Thyroidectomy - StatPearls - NCBI Bookshelf - NIH
    Nov 25, 2024 · Symptomatic goiter: Surgery is indicated for enlarged thyroid glands that cause symptoms such as dysphagia, dyspnea, chronic cough, obstructive ...Continuing Education Activity · Introduction · Indications · Technique or Treatment
  87. [87]
    Considerations for Thyroidectomy as Treatment for Graves Disease
    Apr 24, 2019 · Indications for thyroidectomy include large goiters, goiters causing airway obstruction/dysphagia, moderate to severe ophthalmopathy (because ...
  88. [88]
    Preoperative management in patients with Graves' disease - PMC
    The main clinical situations that favor the choice of surgery as the treatment for Graves' hyperthyroidism include: intolerance, ineffectiveness or recurrence ...Missing: interventions | Show results with:interventions
  89. [89]
    The value of total thyroidectomy as the definitive treatment for ... - NIH
    Feb 7, 2019 · However, the long-term remission rate does not exceed 40–50% of patients, and it was not improved even after >8 months of therapy. Therefore, ...
  90. [90]
    Clinical and Socioeconomic Factors influence treatment decisions in ...
    427 patients underwent definitive treatment for Graves' disease: 300 (70%) underwent RAI while 127 (30%) underwent surgery. Table 1 shows a comparison of all ...
  91. [91]
    2016 American Thyroid Association Guidelines for Diagnosis and ...
    Patients with overt Graves' hyperthyroidism should be treated with any of the following modalities: RAI therapy, ATDs, or thyroidectomy. Strong recommendation, ...
  92. [92]
    Role of Cholestyramine in Refractory Hyperthyroidism: A Case ... - NIH
    Cholestyramine is an effective additional treatment for hyperthyroidism and may be an effective treatment for refractory iodine-induced hyperthyroidism.
  93. [93]
    Thyroid Storm Treatment & Management - Medscape Reference
    Apr 2, 2024 · Glucocorticoids should be weaned and stopped and the dose of thioamides adjusted to maintain thyroid function in the normal range. Beta-blockers ...Approach Considerations · Medical Care · Surgical Care
  94. [94]
    Foods to eat and avoid with hyperthyroidism - MedicalNewsToday
    A person with hyperthyroidism should avoid eating excessive amounts of iodine-rich foods, such as: iodized salt. fish and shellfish.Diet and hyperthyroidism · Foods to eat · Foods to avoid
  95. [95]
    Graves' Disease: Is It Time for Targeted Therapy? A Narrative Review
    Mar 13, 2025 · Current therapies for Graves' disease (GD) primarily aim to manage hyperthyroidism through synthetic antithyroid drugs, radioiodine, or surgery.
  96. [96]
    Effects of Teprotumumab and Role of Human Leukocyte Antigens ...
    This study showed that teprotumumab significantly reduced thyroid stimulating immunoglobin, glycated hemoglobin A1c levels, proptosis, clinical activity score.
  97. [97]
    Immunovant Provides Update on Graves' Disease Development ...
    Sep 9, 2024 · High dose batoclimab achieved 76% response rate in patients uncontrolled on antithyroid drugs (ATDs) at week 12; High dose batoclimab ...
  98. [98]
    New Therapeutic Horizons for Graves' Hyperthyroidism - PMC
    The 2 most widely studied compounds that target FcRn are efgartigimod and rozanolixizumab (64), both of which are currently in phase 3 studies for autoimmune ...
  99. [99]
    News Details - Viridian Therapeutics, Inc.
    Sep 10, 2024 · Veligrotug (VRDN-001) achieved all primary and secondary endpoints in THRIVE, the largest phase 3 trial conducted to date of an anti-IGF-1R antibody in thyroid ...Missing: Graves | Show results with:Graves
  100. [100]
    Efficacy of HIFU for the treatment of benign thyroid nodules - PubMed
    The success rate of HIFU treatment for thyroid nodules is 75.8% at 6 months. Average volume reduction ratios are 48.55% and 55.02% at 6 and 12 months.
  101. [101]
    Thyroid‐Targeted Nano‐Bombs Empower HIFU for Graves' Disease
    Jan 22, 2025 · Subsequent studies showed that HIFU ablation caused thyroid nodule destruction through irreversible tissue necrosis within the targeted area.
  102. [102]
    Hyperthyroidism and cardiovascular complications: a narrative ...
    This review summarizes the cardiovascular consequences of hyperthyroidism with underlying mechanisms.
  103. [103]
    Atrial Fibrillation and Hyperthyroidism - PMC - PubMed Central - NIH
    Oct 1, 2005 · Atrial fibrillation occurs in 10 - 15% of patients with hyperthyroidism. Low serum thyrotropin concentration is an independent risk factor for atrial ...
  104. [104]
    Hyperthyroidism and the Heart - PMC - PubMed Central - NIH
    28 Subclinical hyperthyroidism is further classified into two categories ... Subclinical and overt thyroid dysfunction and risk of all-cause mortality ...Hyperthyroidism And The... · Heart Failure In... · Atrial Fibrillation In...<|control11|><|separator|>
  105. [105]
    The impact of thyroid disorder on cardiovascular disease
    Oct 23, 2024 · Specifically, subclinical thyroid dysfunction has been linked to a 20 % to 80 % higher incidence of vascular morbidity and mortality [3], [5].
  106. [106]
    Evaluation and Management of Bone Health in Patients with Thyroid ...
    Apr 27, 2023 · Overt hyperthyroidism is an established cause of high bone turnover with accelerated bone loss, leading to osteoporosis and increased fracture ...
  107. [107]
    The Influence of Thyroid Pathology on Osteoporosis and Fracture Risk
    Mar 7, 2020 · Hyperthyroidism is generally accepted to decreases bone mineral density (BMD) and increases the risk of fractures. On the other hand, ...
  108. [108]
    Prevalence of thyroid eye disease in Graves' disease: A meta ...
    Jul 21, 2020 · A substantial proportion of patients with GD have TED and often manifest as lid retraction, proptosis and diplopia.Missing: exacerbation | Show results with:exacerbation
  109. [109]
    Prevention of Graves' ophthalmopathy - ScienceDirect.com
    Smoking is the most important risk factor for the occurrence/progression of Graves' ophthalmopathy (GO), as well as for its lower/slower response to ...
  110. [110]
    Malignancy risk of hyperfunctioning thyroid nodules compared ... - NIH
    Feb 25, 2021 · Overall incidence of malignancy in all hot thyroid nodules ranged from 5 to 100% in comparison with non-toxic nodules, 3.8–46%.
  111. [111]
    Myopathy after rapid correction of hyperthyroidism - NIH
    Jan 17, 2020 · Myopathy can be caused by a rapid reduction of thyroid hormone during the treatment of hyperthyroidism.Missing: nodule malignancy
  112. [112]
    What should you expect after radioactive iodine therapy for Graves ...
    The American Thyroid Association recommends testing thyroid hormone (free T4 and total T3) and TSH levels for the first 2 months then to continue every 4 – 6 ...
  113. [113]
    Antithyroid Drug Treatment in Graves' Disease
    Jun 16, 2021 · In the United States, the remission rate was 20% to 30% after ATD treatment for 12 to 18 months, whereas that in Europe was 50% to 60% for 5 to ...
  114. [114]
    Long-term management of Graves disease: a narrative review - PMC
    An 123I or 99mTc pertechnetate scan should be obtained when the clinical presentation suggests thyroiditis or toxic adenoma (Fig. 1A-1C). Thyroid ...Missing: 99mTc- | Show results with:99mTc-
  115. [115]
    Cure Rates After a Single Dose of Radioactive Iodine to Treat ... - NIH
    Aug 23, 2022 · Radioactive iodine (RAI) has been used to treat hyperthyroidism for more than 70 years. Cure rates after RAI therapy range between 80% and 100%, ...Missing: trends | Show results with:trends
  116. [116]
    Graves' Disease: Can It Be Cured? - Endocrinology and Metabolism
    Mar 21, 2019 · Only 27% was in remission, and might be considered cured. If the definition of “cure” would also include the disappearance of thyroid antibodies ...
  117. [117]
    Predictors of Early Remission of Hyperthyroidism in Children1
    We conclude that, of multiple clinical and laboratory variables associated with early remission, BMI sd score and goiter size are independent predictors. ...
  118. [118]
    Influence of biological sex, age and smoking on Graves' orbitopathy
    Apr 3, 2023 · Smoking is the strongest habitual risk factor and showed in a meta-analysis a 4-fold increased risk of GO occurrence (23, 25, 29, 30).
  119. [119]
    Predictive Factors for the Efficacy of Radioactive Iodine Treatment of ...
    Nov 8, 2024 · In our study, the response rate for Graves' hyperthyroidism treated with RAI for the first time was 91.2%, which is similar to the results of ...
  120. [120]
    Predictors of Remission in Graves' Disease Patients Treated With ...
    Jan 13, 2025 · Several factors were identified as predictors of remission, including gender, BMI, and maximum TSH levels. Male patients had a higher remission ...
  121. [121]
    Thyroid Function and Mortality in Patients Treated for Hyperthyroidism
    Jul 6, 2005 · Results In 15 968 person-years of follow-up, 554 died vs 487 expected deaths (standardized mortality ratio [SMR], 1.14; 95% confidence interval ...
  122. [122]
    The impact of thyroid disorder on cardiovascular disease
    Current evidences of thyroid dysfunctions on cardiovascular diseases. Overt hyperthyroidism is associated with a 13% increase in overall mortality and a 21 % ...
  123. [123]
    2018 European Thyroid Association Guideline for the Management ...
    Thyroid function tests are reviewed 3–4 weeks after starting treatment, and the dose is titrated based on free T4 and free T3 levels. A substantial proportion ...
  124. [124]
    Evaluation and Management of Bone Health in Patients with Thyroid ...
    Overt hyperthyroidism is an established cause of high bone turnover with accelerated bone loss, leading to osteoporosis and increased fracture risk.
  125. [125]
    Hyperthyroidism in Pregnancy - StatPearls - NCBI Bookshelf - NIH
    Sep 14, 2025 · Hyperthyroidism is uncommon during pregnancy.[1] The condition is characterized by elevated levels of circulating thyroid hormones, ...
  126. [126]
    Hyperthyroidism in Pregnancy | American Thyroid Association
    It is recommended that the lowest possible dose of ATD be used to control maternal hyperthyroidism in order to minimize the development of hypothyroidism in the ...
  127. [127]
    Graves' Disease and Pregnancy - Everyday Health
    Graves' disease is associated with a higher risk of miscarriage, preterm birth, and high blood pressure in the mother. Learn how to lower these risks.
  128. [128]
    Pregnancy Outcomes among Women with Graves' Hyperthyroidism
    Sep 29, 2021 · This study demonstrated that pregnant women with thyrotoxicosis had an increased risk of preterm birth, low birth weight, preeclampsia and ...Missing: miscarriage | Show results with:miscarriage
  129. [129]
    Update Coming for Thyroid Disease in Pregnancy Guidelines
    Nov 8, 2024 · A preview of much-anticipated updates to guidelines on managing thyroid disease in pregnancy shows key changes to recommendations in the evolving field.
  130. [130]
    2017 Guidelines of the American Thyroid Association for the ...
    Results: The revised guidelines for the management of thyroid disease in pregnancy include recommendations regarding the interpretation of thyroid function ...
  131. [131]
    Global epidemiology of hyperthyroidism and hypothyroidism - PubMed
    In this report, we review the global incidence and prevalence of hyperthyroidism and hypothyroidism, highlighting geographical differences and the effect of ...
  132. [132]
    Serum TSH, T4, and Thyroid Antibodies in the United States ...
    Hypothyroidism was found in 4.6% of the U.S. population (0.3% clinical and 4.3% subclinical) and hyperthyroidism in 1.3% (0.5% clinical and 0.7% subclinical). ( ...<|control11|><|separator|>
  133. [133]
    Smoking as a risk factor for Graves' disease, toxic nodular ... - PubMed
    There was an increased risk of both Graves' disease (odds ratio [OR] = 2.5, 95% confidence interval [CI]: 1.8-3.5), toxic nodular goiter (OR = 1.7, 95% CI: 1.1 ...
  134. [134]
    Is hyperthyroidism genetic? Causes and risk factors
    Nov 22, 2022 · The same study notes that genetic factors might account for 79% of an individual's predisposition to develop Graves' disease. Graves' disease ...Is it genetic? · Risk factors · During pregnancy · Complications
  135. [135]
    Variation in Rates of Autoimmune Thyroid Disease by Race/Ethnicity ...
    Apr 16, 2014 · To our knowledge, this is the first report to identify that Graves disease is more common in blacks and Asian/Pacific Islanders compared with ...
  136. [136]
    Thyroid function during COVID-19 and post-COVID complications in ...
    Feb 3, 2025 · Several studies have reported that COVID-19 can induce thyroid dysfunction, leading to conditions such as thyroiditis and alterations in thyroid hormone levels.
  137. [137]
    Graves' Disease - The New England Journal of Medicine
    Oct 26, 2000 · Robert Graves first identified the association of goiter, palpitations, and exophthalmos in 1835, although Caleb Parry had published details ...
  138. [138]
    The discovery of thyroid replacement therapy. Part 3 - NIH
    Thyroxine was isolated in 1915 by Kendall (actually, on Christmas Day, 1914), its chemical structure determined in 1926 by Harington, and it was synthesized by ...
  139. [139]
    The Origin of Antithyroid Drugs - PubMed
    Aug 18, 2023 · When the antithyroid drugs were discovered in the early 1940s, they were immediately recognized as a revolutionary new treatment for hyperthyroidism.Missing: post WWII 50s
  140. [140]
    Saul Hertz, MD, and the birth of radionuclide therapy - PMC - NIH
    Apr 27, 2017 · Saul Hertz first using radioiodine to treat a patient with thyroid disease. In honor of this anniversary, we have spent a bit of time ...
  141. [141]
    Autoantibodies to the TSH Receptor—from discovery to ... - J-Stage
    In 1974 Graves' IgG was shown to compete with 125I-labelled TSH for the TSH receptor providing good evidence that Graves' hyperthyroidism was caused by TSH ...
  142. [142]
    A Review of the History of Radioactive Iodine Theranostics - NIH
    Oct 19, 2020 · Saul Hertz's success with RAI in thyrotoxicosis fueled a seamless transition to Samuel Seidlin's investigations with RAI in thyroid cancer.
  143. [143]
    The history of Graves' disease discovery and treatment - Mattioli 1885
    Feb 16, 2022 · Indeed, it was the Englishman Caleb Hillier Parry to firstly report a case of hyperthyroidism and goiter in 1786, but his report was not ...