Graves Disease

Back

Background

Graves disease, named after Robert J. Graves, MD,[1] circa 1830s, is an autoimmune disease characterized by hyperthyroidism due to circulating autoantibodies. Thyroid-stimulating immunoglobulins (TSIs) bind to and activate thyrotropin receptors, causing the thyroid gland to grow and the thyroid follicles to increase synthesis of thyroid hormone. Graves disease, along with Hashimoto thyroiditis, is classified as an autoimmune thyroid disorder.

In some patients, Graves disease represents a part of more extensive autoimmune processes leading to dysfunction of multiple organs (eg, polyglandular autoimmune syndromes). Graves disease is associated with pernicious anemia, vitiligo, diabetes mellitus type 1, autoimmune adrenal insufficiency, systemic sclerosis, myasthenia gravis, Sjögren syndrome, rheumatoid arthritis, and systemic lupus erythematosus.[2]

Graves ophthalmopathy is shown below.



View Image

Graves disease. Varying degrees of manifestations of Graves ophthalmopathy.

 

Pathophysiology

In Graves disease, B and T lymphocyte-mediated autoimmunity are known to be directed at 4 well-known thyroid antigens: thyroglobulin, thyroid peroxidase, sodium-iodide symporter and the thyrotropin receptor. However, the thyrotropin receptor itself is the primary autoantigen of Graves disease and is responsible for the manifestation of hyperthyroidism. In this disease, the antibody and cell-mediated thyroid antigen-specific immune responses are well defined. Direct proof of an autoimmune disorder that is mediated by autoantibodies is the development of hyperthyroidism in healthy subjects by transferring thyrotropin receptor antibodies in serum from patients with Graves disease and the passive transfer of thyrotropin receptor antibodies to the fetus in pregnant women.

The thyroid gland is under continuous stimulation by circulating autoantibodies against the thyrotropin receptor, and pituitary thyrotropin secretion is suppressed because of the increased production of thyroid hormones. The stimulating activity of thyrotropin receptor antibodies is found mostly in the immunoglobulin G1 subclass. These thyroid-stimulating antibodies cause release of thyroid hormone and thyroglobulin that is mediated by 3,'5'-cyclic adenosine monophosphate (cyclic AMP), and they also stimulate iodine uptake, protein synthesis, and thyroid gland growth.

The anti-sodium-iodide symporter, antithyroglobulin, and antithyroid peroxidase antibodies appear to have little role in the etiology of hyperthyroidism in Graves disease. However, they are markers of autoimmune disease against the thyroid. Intrathyroidal lymphocytic infiltration is the initial histologic abnormality in persons with autoimmune thyroid disease and can be correlated with the titer of thyroid antibodies. Besides being the source of autoantigens, the thyroid cells express molecules that mediate T cell adhesion and complement regulation (Fas and cytokines) that participate and interact with the immune system. In these patients, the proportion of CD4 lymphocytes is lower in the thyroid than in the peripheral blood. The increased Fas expression in intrathyroidal CD4 T lymphocytes may be the cause of CD4 lymphocyte reduction in these individuals.

Several autoimmune thyroid disease susceptibility genes have been identified: CD40, CTLA-4, thyroglobulin, TSH receptor, and PTPN22.[3, 4] Some of these susceptibility genes are specific to either Graves disease or Hashimoto thyroiditis, while others confer susceptibility to both conditions. The genetic predisposition to thyroid autoimmunity may interact with environmental factors or events to precipitate the onset of Graves disease.

Two new susceptibility loci were found: the RNASET2-FGFR1OP-CCR6 region at 6q27 and an intergenic region at 4p14.[5] Moreover, strong associations of thyroid-stimulating hormone receptor and major histocompatibility complex class II variants with persistently thyroid stimulating hormone receptor autoantibodies (TRAb)-positive Graves disease were found.[6]

Graves disease patients a have higher rate of peripheral blood mononuclear cell conversion into CD34+ fibrocytes compared with healthy controls. These cells may contribute to the pathophysiology of ophthalmopathy by accumulating in orbital tissues and producing inflammatory cytokines, including TNF-alpha and IL-6.[7] In a genome-wide association study of more than 1500 Graves disease patients and 1500 controls, 6 susceptibility loci were found to be related to Graves disease (major histocompatibility complex, TSH receptor, CTLA4, FCRL3, RNASET2-FGFR1OP-CCR6 region at 6q27, and an intergenic region at 4p14.[8]

Pathophysiologic mechanisms are shown in the image below.



View Image

Pathophysiologic mechanisms of Graves disease relating thyroid-stimulating immunoglobulins to hyperthyroidism and ophthalmopathy. T4 is levothyroxine.....

Epidemiology

Frequency

United States

Graves disease is the most common cause of hyperthyroidism in the United States. A study conducted in Olmstead County, Minnesota estimated the incidence to be approximately 30 cases per 100,000 persons per year.[9] The prevalence of maternal thyrotoxicosis is approximately 1 case per 500 persons, with maternal Graves disease being the most common etiology. Commonly, patients have a family history involving a wide spectrum of autoimmune thyroid diseases, such as Graves disease, Hashimoto thyroiditis, or postpartum thyroiditis, among others.

International

Among the causes of spontaneous thyrotoxicosis, Graves disease is the most common. Graves disease represents 60-90% of all causes of thyrotoxicosis in different regions of the world. In the Wickham Study in the United Kingdom, the incidence was reported to be 100-200 cases per 100,000 population per year.[10] The incidence in women in the UK has been reported to be 80 cases 100,000 per year.[11]

Mortality/Morbidity

If left untreated, Graves disease can cause severe thyrotoxicosis. A life-threatening thyrotoxic crisis (ie, thyroid storm) can occur. Long-standing severe thyrotoxicosis leads to severe weight loss with catabolism of bone and muscle.[12] Cardiac complications and psychocognitive complications can cause significant morbidity. Graves disease is also associated with ophthalmopathy, dermopathy, and acropachy.

Thyroid storm is an exaggerated state of thyrotoxicosis.[13] It occurs in patients who have unrecognized or inadequately treated thyrotoxicosis and a superimposed precipitating event such as thyroid surgery, nonthyroidal surgery, infection, or trauma. When thyroid storm was first described, the acute mortality rate was nearly 100%. In current practice, with aggressive therapy and early recognition of the syndrome, the mortality rate is approximately 20%.[14]

Long-term excess of thyroid hormone can lead to osteoporosis in men and women. The effect can be particularly devastating in women, in whom the disease may compound the bone loss secondary to chronic anovulation or menopause. Bone loss is accelerated in patients with hyperthyroidism. The increase in bone loss can be demonstrated by increased urinary pyridinoline cross-link excretion. Serum calcium and phosphate, plasma FGF-23 were significantly higher in the patients with Graves disease than in healthy control subjects, suggesting that FGF-23 is physiologically related to serum phosphate homeostasis in untreated Graves disease.[15]

Hyperthyroidism increases muscular energy expenditure and muscle protein breakdown. These abnormalities may explain the sarcopenia and myopathy observed in patients with hyperthyroid Graves disease.

Cardiac hypertrophy has been reported in thyrotoxicosis of different etiologies. Rhythm disturbances such as extrasystolic arrhythmia, atrial fibrillation, and flutter are common. Cardiomyopathy and congestive heart failure can occur.[16]

Psychiatric manifestations such as mood and anxiety disorders are common.[17] Subjective cognitive dysfunction is often reported by Graves disease patients and may be due to affective and somatic manifestations of thyrotoxicosis, which remit after treatment of Graves thyrotoxicosis.[18]

Nonpitting edema is the most prevalent form of dermopathy (about 40%) and are primarily in the pretibial area. The nearly all (>95%) patients with dermopathy had ophthalmopathy.[19] Advanced forms of dermopathy are elephantiasis or thyroid acropachy. Severe acropachy can be disabling and can lead to total loss of hand function.

Progression of ophthalmopathy can lead to compromised vision and blindness. Visual loss due to corneal lesions or optic nerve compression can be seen in severe Graves ophthalmopathy.

In a study of 1128 patients with Graves ophthalmopathy, Kim et al found the prevalence of ocular hypertension (OHT) to be 6.8% and the prevalence of open-angle glaucoma (OAG) to be 1.6%. The prevalences were higher in patients over age 40 years, being 9.5% and 3.4%, respectively. The investigators also reported the prevalence of OHT in Graves ophthalmopathy to be associated with male sex, duration of the ophthalmopathy, a clinical activity score of 3 or above, extraocular muscle involvement, and lid retraction. Male sex and duration of the ophthalmopathy were associated with the prevalence of OAG in Graves ophthalmopathy.[20]

Maternal Graves disease can lead to neonatal hyperthyroidism by transplacental transfer of thyroid-stimulating antibodies. Approximately 1-5% of children of mothers with Graves disease (usually with high TSI titer) are affected. Usually, the TSI titer falls during pregnancy.

Elderly individuals may develop apathetic hyperthyroidism, and the only presenting features may be unexplained weight loss or cardiac symptoms such as atrial fibrillation and congestive heart failure.

Boelaert et al investigated the prevalence of and relative risks for coexisting autoimmune diseases in patients with Graves disease (2791 patients) or Hashimoto thyroiditis (495 patients). The authors found coexisting disorders in 9.7% of patients with Graves disease and in 14.3% of those with Hashimoto thyroiditis, with rheumatoid arthritis being the most common of these (prevalence = 3.15% and 4.24% in Graves disease and Hashimoto thyroiditis, respectively). Relative risks of greater than 10 were found for pernicious anemia, systemic lupus erythematosus, Addison disease, celiac disease, and vitiligo. The authors also reported a tendency for parents of patients with Graves disease or Hashimoto thyroiditis to have a history of hyperthyroidism or hypothyroidism, respectively.[21]

Race

In whites, autoimmune thyroid diseases are, based on linkage analysis, linked with the following loci: AITD1, CTLA4, GD1, GD2, GD3, HT1, and HT2. Different loci have been reported to be linked with autoimmune thyroid diseases in persons of other races.

Susceptibility is influenced by genes in the human leukocyte antigen (HLA) region on chromosome 6 and in CTLA4 on band 2q33. Association with specific HLA haplotypes has been observed and is found to vary with ethnicity.

Sex

As with most autoimmune diseases, susceptibility is increased in females. Hyperthyroidism due to Graves disease has a female-to-male ratio of 7-8:1.

The female-to-male ratio for pretibial myxedema is 3.5:1. Only 7% of patients with localized myxedema have thyroid acropachy.

Unlike the other manifestations of Graves disease, the female-to-male ratio for thyroid acropachy is 1:1.

Age

Typically, Graves disease is a disease of young women, but it may occur in persons of any age.

The typical age range is 20-40 years.

Most affected women are aged 30-60 years.

Prognosis

The natural history of Graves disease is that most patients become hypothyroid and require replacement. Similarly, the ophthalmopathy generally becomes quiescent. On occasion, hyperthyroidism returns because of persisting thyroid tissue after ablation and high antibody titers of anti-TSI. Further therapy may be necessary in the form of surgery or radioactive iodine ablation.

A study by Tun et al indicated that in patients with Graves disease receiving thionamide therapy, high thyrotropin receptor–stimulating antibody (TRab) levels at diagnosis of the disease and/or high TRab levels at treatment cessation are risk factors for relapse, particularly within the first two years. The study included 266 patients.[22]

A retrospective study by Rabon et al indicated that in children with Graves disease, antithyroid drugs usually do not induce remission, although most children who do achieve remission through these agents do not relapse. Of 268 children who were started on an antithyroid drug, 57 (21%) experienced remission, with 16 of them (28%) relapsing.[23]

Patient Education

Awareness of the symptoms related to hyperthyroidism and hypothyroidism is important, especially in the titration of antithyroid agents and in replacement therapy for hypothyroidism.

Patients also should be aware of the potential adverse effects of these medicines. They should watch for fever, sore throat, and throat ulcers.

Patients also must be instructed to avoid cold medicines that contain alpha-adrenergic agonists such as ephedrine or pseudoephedrine.

For patient education resources, see the Endocrine System Center, as well as Thyroid Problems.

History

Because Graves disease is an autoimmune disorder that also affects other organ systems, taking a careful patient history is essential to establishing the diagnosis.

In some cases, the history might suggest a triggering factor such as trauma to the thyroid, including surgery of the thyroid gland, percutaneous injection of ethanol, and infarction of a thyroid adenoma. Other factors might include interferon (eg, interferon beta-1b) or interleukin (IL-4) therapy.

Patients usually present with symptoms typical of thyrotoxicosis. Hyperthyroidism is characterized by both increased sympathetic and decreased vagal modulation.[24] Tachycardia and palpitation are very common symptoms.

Not all patients present with such classic features. In fact, a subset of patients with euthyroid Graves disease is described.

In elderly individuals, fewer symptoms are apparent to the patient. Clues may include unexplained weight loss, hyperhidrosis, or rapid heart beat.

Young adults of Southeast Asian descent may complain of sudden paralysis thought to be related to thyrotoxic periodic paralysis. There is an association of polymorphisms of the calcium channel alpha1-subunit gene with thyrotoxic periodic paralysis.[25] One third of patients with thyrotoxic hypokalemic periodic paralysis were found to have mutations in the inwardly rectifying potassium channel (Kir2.6).[26]

The symptoms of Graves disease, organized by systems, are as follows:

Physical

Most of the physical findings are related to thyrotoxicosis.

Physical findings that are unique to Graves disease but not associated with other causes of hyperthyroidism include ophthalmopathy and dermopathy. Myxedematous changes of the skin (usually in the pretibial areas) are described as resembling an orange peel in color and texture. Onycholysis can be seen usually in the fourth and fifth fingernails.

The presence of a diffusely enlarged thyroid gland, thyrotoxic signs and symptoms, together with evidence of ophthalmopathy or dermopathy, can establish the diagnosis.

Common physical findings, organized by anatomic regions, are as follows:

Ophthalmopathy is a hallmark of Graves disease. Approximately 25-30% of patients with Graves disease have clinical evidence of Graves ophthalmopathy. Progression from mild to moderate/severe ophthalmopathy occurs in about 3% of cases.[27] Thyrotropin receptor is highly expressed in the fat and connective tissue of patients with Graves ophthalmopathy. Measuring diplopia fields, eyelid fissures, range of extraocular muscles, visual acuity, and proptosis provides quantitative assessment to follow the course of ophthalmopathy. Signs of corneal or conjunctival irritation include conjunctival injection and chemosis. A complete ophthalmologic examination, including retinal examination and slit-lamp examination by an ophthalmologist, is indicated if the patient is symptomatic.

Although thyroid nodule(s) may be present, excluding multinodular toxic goiter (especially in older patients) as the cause of thyrotoxicosis is essential. The approach to treatment may be different. Excluding thyroid neoplasia is also important in these patients because reports have indicated that differentiated thyroid cancer is probably more common in patients with Graves disease and may also have a more aggressive course in these patients.[28]

Similarly, mortality has been reported to be increased in patients with Graves disease and differentiated thyroid carcinoma compared with euthyroid control patients with differentiated thyroid carcinoma.[29] Graves disease patients had also higher mortality rates compared with general population, with a particular increase in mortality due to cardiovascular and lung disorders, while hyperthyroid patients had increased mortality secondary to toxic nodules had increased mortality associated with malignancies.[30]

Causes

Graves disease is autoimmune in etiology, and the immune mechanisms involved may be one of the following:

The autoimmune process in Graves disease is influenced by a combination of environmental and genetic factors.

Several autoimmune thyroid disease susceptibility genes have been identified: CD40, CTLA-4, thyroglobulin, TSH receptor, and PTPN22.[3] Some of these susceptibility genes are specific to either Graves disease or Hashimoto thyroiditis, while others confer susceptibility to both conditions. HLA-DRB1 and HLA-DQB1 also appear to be associated with Graves disease susceptibility. Genetic factors contribute approximately 20-30% of overall disease susceptibility.

Environmental factors associated with susceptibility are largely unproven. Other factors include infection, iodide intake, stress, female sex, steroids, and toxins. Smoking has been implicated in the worsening of Graves ophthalmopathy.

Laboratory Studies

Ultrasensitive (third-generation) thyrotropin assays remain the best screening test for thyroid disorders.

Liver function test results should be obtained to monitor for liver toxicity caused by thioamides (antithyroid medications).

A CBC count with differential should be obtained at baseline and with the development of fever or symptoms of infection. Graves disease may be associated with normocytic anemia, low-normal to slightly depressed total WBC count with relative lymphocytosis and monocytosis, low-normal to slightly depressed platelet count. Thioamides may rarely cause severe hematologic side effects, but routine screening for these rare events is not cost-effective.

Investigation of gynecomastia associated with Graves disease may reveal increased sex hormone–binding globulin levels and decreased free testosterone levels.

Graves disease may worsen diabetes control and may be reflected by an increase in hemoglobin A1C in diabetic patients.

A fasting lipid profile may show decreased total cholesterol levels and decreased triglyceride levels.

Thyrotropin-releasing hormone testing has largely been replaced by third-generation thyrotropin assays.

A high titer of serum antibodies to collagen XIII is associated with active Graves ophthalmopathy.[40]

Imaging Studies

Radioactive iodine scanning and measurements of iodine uptake are useful in differentiating the causes of hyperthyroidism. In Graves disease, the radioactive iodine uptake is increased and the uptake is diffusely distributed over the entire gland.[39]

Ultrasounds with color-Doppler evaluation have been found to be cost-effective in hyperthyroid patients.[28, 41] A prospective trial showed that thyroid ultrasound findings are predictive of radioiodine treatment outcome, and, in patients with Graves disease, normoechogenic and large glands are associated with increased radioresistance.[42]

Computed tomography scanning or magnetic resonance imaging (of the orbits) may be necessary in the evaluation of proptosis. If routinely performed, most patients have evidence of ophthalmopathy, such as an increased volume of extraocular muscles and/or retrobulbar connective tissue. These techniques are useful to monitor changes over time or to ascertain the effects of treatment. Careful monitoring is required after using iodinated contrast agents as they may affect ongoing treatment plans.

Histologic Findings

In select cases in which thyroidectomy was performed for the treatment of severe hyperthyroidism, the thyroid glands from patients with Graves disease show lymphocytic infiltrates and follicular hypertrophy, with little colloid present.

Medical Care

Treatment involves alleviation of symptoms and correction of the thyrotoxic state. Adrenergic hyperfunction is treated with beta-adrenergic blockade. Correcting the high thyroid hormone levels can be achieved with antithyroid medications that block the synthesis of thyroid hormones or by treatment with radioactive iodine.

A study by Yasuda et al of pediatric patients with Graves disease found that a greater incidence and variety of adverse events occurred in those on a high dose of the antithyroid drug methimazole (0.7 or more mg/kg/day) than in those on a low dose (< 0.7 mg/kg/day), with the frequencies of adverse events being 50% and 20%, respectively. However, neutropenia and rash were found to manifest independently of dose.[43]

Radioactive iodine

The most commonly used therapy for Graves disease is radioactive iodine. Indications for radioactive iodine over antithyroid agents include a large thyroid gland, multiple symptoms of thyrotoxicosis, high levels of thyroxine, and high titers of TSI. Information and guidelines are as follows:

Graves ophthalmopathy

Graves ophthalmopathy can be divided into 2 clinical phases: the inflammatory stage and the fibrotic stage. The inflammatory stage is marked by edema and deposition of glycosaminoglycan in the extraocular muscles. This results in the clinical manifestations of orbital swelling, stare, diplopia, periorbital edema, and, at times, pain. The fibrotic stage is a convalescent phase and may result in further diplopia and lid retraction. Vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) levels may be mechanistically involved in Graves ophthalmopathy. Serum VEGF and b-FGF levels were higher in patients with Graves ophthalmopathy than in patients without, and they correlated with ophthalmopathy clinical activity scores.[50]

In a longitudinal cohort of 8404 adults with newly diagnosed Graves disease, 740 (8.8%) developed ophthalmopathy.[51] Graves ophthalmopathy improves spontaneously in 64% of patients. Approximately 10-20% of patients have gradual progression of disease over many years, followed by clinical stability. Approximately 2-5% have progressive worsening of the disease, with visual impairment in some.

Radioactive iodine therapy for Graves disease is a risk factor for Graves ophthalmopathy. Cholesterol-lowering drugs of the hydroxymethylglutarate-coenzyme A reductase inhibitor class were associated with a reduced risk of ophthalmopathy.[51] Ethnic factors are also important for Graves ophthalmopathy after radioactive iodine treatment; Japanese patients are less prone to Graves ophthalmopathy after radioactive iodine.[52]

Correction of both hyperthyroidism and hypothyroidism is important for the ophthalmopathy. Antithyroid drugs and thyroidectomy do not influence the course of the ophthalmopathy, whereas radioiodine treatment may exacerbate preexisting ophthalmopathy but can be prevented by glucocorticoids. However, Japanese patients may not respond well to prophylactic use of low-dose glucocorticosteroids.[52] No beneficial effect of glucocorticoid prophylaxis was found in patients without preexisting clinical evidence of ophthalmopathy.[53] In the long term, thyroid ablation may be beneficial for ophthalmopathy because of the decrease in antigens shared by the thyroid and the orbit in the autoimmune reactions. In general, treatment of hyperthyroidism is associated with an improvement of ophthalmopathy, but hypothyroidism must be avoided because it worsens ophthalmopathy.[54, 55, 56]

For mild-to-moderate ophthalmopathy, local therapeutic measures (eg, artificial tears and ointments, sunglasses, eye patches, nocturnal taping of the eyes, prisms, elevating the head at night) can control symptoms and signs. If the disease is active, the mainstays of therapy are (1) high-dose glucocorticoids,[57] (2) orbital radiotherapy, (3) both, or (4) orbital decompression.[58] A meta-analysis has shown that a 3-month course of prednisone (0.4-0.5 mg/kg) reduced the progression of preexisting mild-to-moderate ophthalmopathy.[53]

For severe or progressive disease, glucocorticoids at 40 mg/d (usual dose) may be tried. The drug should be continued until evidence of improvement and disease stability is observed. The dosage is then tapered over 4-12 weeks. High-dose pulse glucocorticoid therapy has also been used with good results but may be associated with a slightly increased risk of acute liver damage.[59]

A study by Liao and Huang evaluated the correlation of retrobulbar volume change, resected orbital fat volume, and proptosis reduction after surgical decompression in patients with Graves ophthalmopathy.[60] Decompression by resecting orbital fat was found to reduce proptosis in patients with disfiguring Graves ophthalmopathy.

If no response to therapy occurs in the inflammatory phase, orbital radiotherapy with or without steroids may be tried. Orbital radiotherapy did not increase the risk for radiation-induced tumors or retinopathy, except in patients with diabetes with possible or definite retinopathy.[61] Low-dose radiation from various sources (even if not aimed at the eyes) is linked to cataracts, which may be detected only after long term follow-up.[62] A meta-analysis found better outcome with combining steroids with radiotherapy compared with steroid therapy alone. However, quality-of-life scores were not different between the 2 groups.[63] Diuretics have a limited effect on the edema caused by venous engorgement of the orbit.

Gamma knife surgery has been attempted with success in a limited number of patients, but further studies are needed to validate this approach.

Surgical management is generally performed in the fibrotic phase, when the patient is euthyroid.

Novel treatments such as somatostatin analogs or intravenous immunoglobulins are under evaluation. Studies with octreotide LAR (long-acting, repeatable) show conflicting or marginal therapeutic benefit for patients with Graves ophthalmopathy.[64, 65, 66] Infliximab, an anti-tumour necrosis factor alpha (TNF-α) antibody, has been reported to successfully treat a case of sight-threatening Graves ophthalmopathy.[67] Rituximab, anti-CD20 monoclonal antibody, may transiently deplete B-lymphocytes and potentially suppress the active inflammatory phase of Graves ophthalmopathy.[68] However, clinical data concerning rituximab are still conflicting and controversial.[69, 70] A multicentered prospective pilot study suggests that periocular injection of triamcinolone may reduce diplopia and the size of extraocular muscles in patients with Graves ophthalmopathy of recent onset.[71] In a prospective randomized trial, pentoxifylline improved symptoms and proptosis in the inactive phase of Graves ophthalmopathy.[72]

Pretibial myxedema

Some degree of pretibial (localized dermopathy) myxedema is observed in 5-10% of patients, with 1-2% having cosmetically significant lesions. Affected patients tend to have more severe ophthalmopathy than those who are not affected.

It usually manifests as elevated, firm, nonpitting, localized thickening over the lateral aspect of the lower leg, with bilateral involvement. It also may involve the upper extremities.

Milder cases do not require therapy other than treatment of the thyrotoxicosis.

Therapy with topical steroids applied under an occlusive plastic dressing film (eg, Saran Wrap) for 3-10 weeks has been helpful. In severe cases, pulse glucocorticoid therapy may be tried.

Acropachy

Clubbing of fingers with osteoarthropathy, including periosteal new bone formation, may occur. This almost always occurs in association with ophthalmopathy and dermopathy. No therapy has been proven to be effective.

Inpatient Care

With the exception of thyroid storm, Graves disease generally is managed in an outpatient setting.

On occasion, patients may present with thyrotoxic heart disease, including congestive heart failure, atrial fibrillation, or other tachyarrhythmia, which requires inpatient management. Prompt recognition of thyrotoxicosis is required for optimal therapy. In certain cases, the patient may have to be admitted to the intensive care unit or critical care unit. Appropriate subspecialty consultations (eg, endocrinologist, cardiologist) are needed. Once patients' conditions are stabilized, they can be transferred to a regular room or discharged from the hospital.

In certain cases (ie, noncompliant patients, those who develop severe reactions to antithyroid drugs), radioiodine ablation therapy may be given in an inpatient setting.

Surgical Care

Indications and outcomes are as follows:

Procedures and preparations are as follows:

A literature review by Zhang et al comparing endoscopic with conventional open thyroidectomy for Graves disease reported that the endoscopic technique offers better cosmetic satisfaction and less blood loss, while open surgery is associated with reduced operation time. Complication rates for the two techniques with regard to transient recurrent laryngeal nerve palsy, recurrent hyperthyroidism, hypothyroidism, and transient hypocalcemia were equivalent.[81]

Ophthalmopathy is as follows:

Consultations

Consultation with an endocrinologist may be necessary for the management and regulation of thyroid hormone levels in atypical presentations, as follows:

Consultation with an ophthalmologist may be needed in the following situations:

Consultation with a dermatologist may be needed in patients with localized myxedema that is unresponsive to topical corticosteroids.

Diet

The amount of iodine in the diet can influence the hormone synthesis activity in the thyroid gland.

Iodine-containing food has different effects on thyroid uptake of131 I and technetium Tc 99m. Iodine-rich food decreases131 I uptake but increases99m Tc in most patients. However, the diagnostic value of a radioiodine uptake test to differentiate Graves disease and silent thyroiditis is not affected by dietary iodine intake.[83] Iodine restriction before a radioiodine uptake test is unnecessary.

Dietary iodine intake may influence the remission rate after antithyroid drug therapy. This is based on the observation that the outcome of antithyroid therapy in the older literature showed lower remission rates than it did in later studies and that the average dietary iodine content has been decreasing over the years. However, a direct causal relationship has not been established by clinical trials.

In addition, the use of antithyroid drug therapy for more than 2 years is a good predictor of Graves disease.[84] In pediatric patients with Graves disease, no difference was noted in remission rates between methimazole and PTU, while minor adverse effects were significantly increased in patients receiving PTU doses of 7.5 mg/kg or higher.[85]

Activity

Given the high-output state of the heart, strenuous exercise may be detrimental. The patient should be advised to avoid severe fatigue from exercise. Patients can use their pulse as a guide to activity.

Complications

Agranulocytosis is an idiosyncratic reaction to antithyroid drugs. The role of serial CBC counts to predict who will develop this serious adverse reaction is not well established.

In contrast to patients with Graves disease, preoperative iodine treatment should not be given to patients with toxic nodular goiters because it can exacerbate hyperthyroidism.

Prevention

Prevention is difficult because of the lack of knowledge regarding the pathogenesis of this condition.

Long-Term Monitoring

Hyperthyroidism represents a continuum of thyroid dysfunction. In the case of thyroid storm, decompensated patients with hyperthyroidism should be cared for in an institution with personnel familiar with this disease.

All patients should receive long-term follow-up, regardless of the mode of therapy (ie, surgery, radioiodine, antithyroid drugs).

Close follow-up visits with monitoring of examination findings, thyroid hormone levels, and thyrotropin levels are required.

If the patient is on antithyroidal medication (eg, thioamides), liver function tests and CBC counts with differentials should be monitored based on the clinical situation.

Examination of the eyes should be a routine part of follow-up of these patients, given the lack of predictability of ophthalmopathy.

Smoking cessation techniques should be continued.

Guidelines Summary

In 2016, the American Thyroid Association updated the 2011 hyperthyroidism/thyrotoxicosis guidelines it had codeveloped with the American Association of Clinical Endocrinologists. The following are a sampling of the 124 evidence-based recommendations included in the guideline update[86] :

Medication Summary

The goals of pharmacotherapy are to reduce morbidity and to prevent complications.

Propylthiouracil

Clinical Context:  Derivative of thiourea that inhibits organification of iodine by thyroid gland. Blocks oxidation of iodine in thyroid gland, thereby inhibiting thyroid hormone synthesis; inhibits T4-to-T3 conversion by blocking type I deiodinase (advantage over other agents). Usual course/duration of therapy is 1-2 y; sustained remission more likely after 1-2 y vs 3-6 mo of therapy.

Methimazole (Tapazole)

Clinical Context:  Inhibits thyroid hormone by blocking oxidation of iodine in thyroid gland; however, not known to inhibit peripheral conversion of thyroid hormone. Considerable debate surrounds optimal dosage/duration.

Class Summary

Thioamides function as antithyroid agents mainly by inhibiting iodide organification and coupling processes, thereby preventing synthesis of thyroid hormones. Half-life of T4 is 7 d in persons who are euthyroid and somewhat shorter in patients who are thyrotoxic. This accounts for a several-week delay in onset of clinical improvement in most patients. Agents have been reported to alter intrathyroidal immunoregulatory mechanisms. Only oral preparations are available, but they have been used as retention enemas in patients in whom oral intake is not possible or is contraindicated.

Although these agents fall under pregnancy category D, they have been used safely in many pregnant patients. Retrospective study indicates rate of major congenital malformations with PTU (3%) or methimazole (2.7%) was not significantly different from normal background rate (2-5%). Duration of treatment ranged from 0-23 wk, with doses ranging from 100-600 mg/d of PTU or 10-60 mg/d of methimazole.

Concentrations of methimazole are higher in breast milk; therefore, PTU is preferred in this patient population.

Risk of agranulocytosis is similar (0.2-0.5%) in members of this class. In general, PTU is associated with transaminase elevation in susceptible individuals, while methimazole may cause a cholestatic effect.[87]

The US Food and Drug Administration (FDA) added a boxed warning, the strongest warning issued by the FDA, to the prescribing information for PTU. The boxed warning emphasizes the risk for severe liver injury and acute liver failure, some of which have been fatal. The boxed warning also states that PTU should be reserved for use in patients who cannot tolerate other treatments, such as methimazole, radioactive iodine, or surgery.

Medically treated Graves disease has a significant risk of relapse (23% within 6 months of discontinuation of antithyroid medication and 42% within 5 years). The presence of goiter is associated with an increased risk of relapse after medical therapy.[88]

The decision to include a boxed warning was based on the FDA's review of postmarketing safety reports and on meetings held with the American Thyroid Association, the National Institute of Child Health and Human Development, and the pediatric endocrine clinical community.

The FDA has identified 32 cases (22 adult and 10 pediatric) of serious liver injury associated with PTU. Of the adults, 12 deaths and 5 liver transplants occurred, and among the pediatric patients, 1 death and 6 liver transplants occurred. PTU is indicated for hyperthyroidism due to Graves disease. These reports suggest an increased risk for liver toxicity with PTU compared with methimazole. Serious liver injury has been identified with methimazole in 5 cases (3 resulting in death).

PTU is considered to be a second-line drug therapy, except in patients who are allergic to or intolerant of methimazole, or in women who are in the first trimester of pregnancy. Rare cases of embryopathy, including aplasia cutis, have been reported with methimazole during pregnancy. The FDA recommends the following criteria be considered for prescribing PTU (for more information, see the FDA Safety Alert):

- Reserve PTU use during first trimester of pregnancy, or in patients who are allergic to or intolerant of methimazole.

- Closely monitor PTU therapy for signs and symptoms of liver injury, especially during the first 6 months after initiation of therapy.

- For suspected liver injury, promptly discontinue PTU therapy, evaluate the patient for evidence of liver injury, and provide supportive care.

- PTU should not be used in pediatric patients unless the patient is allergic to or intolerant of methimazole and no other treatment options are available.

- Counsel patients to promptly contact their health care provider for the following signs or symptoms: fatigue, weakness, vague abdominal pain, loss of appetite, itching, easy bruising, or yellowing of the eyes or skin.

Propranolol (Inderal, Inderal LA)

Clinical Context:  DOC in treating cardiac arrhythmias resulting from hyperthyroidism. Controls cardiac and psychomotor manifestations within minutes.

Drug completely absorbed from GI tract; because of extensive first-pass metabolism in liver, systemic bioavailability affected by hepatic blood flow, intrinsic clearance in liver, and genetic and age differences in individuals.

Dosage prediction for IV from prior PO difficult; therefore, careful titration of IV dose necessary.

Atenolol (Tenormin)

Clinical Context:  Selectively blocks beta1 receptors with little or no effect on beta2 types. Useful in treating cardiac arrhythmias resulting from hyperthyroidism.

Class Summary

Both cardioselective and noncardioselective types are important adjuncts in treating hyperthyroidism. Beta-blockade provides rapid relief of hyperadrenergic symptoms and signs of thyrotoxicosis (eg, palpitations, tremors, anxiety, heat intolerance, various eyelid signs) before any decrease in thyroid hormone levels demonstrated. Also useful in preventing episodes of hypokalemic periodic paralysis in susceptible individuals. DOC for thyroiditis, which is self-limiting. High-dose propranolol can inhibit peripheral T4-to-T3 conversion. Also useful in preparing thyrotoxic patients for surgery.

Potassium iodide (SSKI, Pima)

Clinical Context:  Inhibits thyroid hormone secretion.

Contains 5% iodine and 10% potassium iodide. Contains 8 mg of iodide per drop. May be mixed with juice or water for intake.

Diatrizoate (Hypaque sodium)

Clinical Context:  Blocks release of thyroid hormones.

Iopanoic acid (Telepaque)

Clinical Context:  Oral contrast agent for rapid and significant inhibition of peripheral T4-to-T3 conversion. Inorganic iodide released also blocks release of thyroid hormones.

Class Summary

Have long been used to treat thyrotoxicosis and are still important adjunctive therapy for hyperthyroidism in modern medicine. In pharmacologic concentrations (100-times normal plasma level), decrease activity of thyroid gland. Action involves decreasing thyroidal iodide uptake, decreasing iodide oxidation and organification, and blocking release of thyroid hormones (Wolff-Chaikoff effect).

Oral contrast agents ipodate or iopanoic acid also shown to be potent inhibitors of T4-to-T3 conversion, making them ideal for severe or decompensated thyrotoxicosis. Generally administered after thioamide is started. Also used as preoperative preparation for thyroid surgery for Graves disease.

In combination with thioamides and/or propranolol, iodines are used routinely before thyroidectomy. Iodines are given for 2-3 weeks before surgery and decrease vascularity of hyperthyroid gland. Making patient euthyroid before surgery prevents intraoperative and postoperative complications.

Cholestyramine (Questran)

Clinical Context:  Can be used to lower serum thyroid hormone levels. This cholesterol-lowering resin has been used as adjunctive therapy in management of hyperthyroid Graves disease. Proved to be effective and well-tolerated adjunctive therapy, leading to a more rapid reduction of thyroid hormone levels.

Class Summary

Based on the observation that a small portion of L-thyroxine is usually reabsorbed in the bowel and recycled in the enterohepatic circulation, exchange resins have been used to bind thyroid hormones in the GI tract. Enterohepatic circulation of thyroxine is increased in cases of hyperthyroidism.

Lithium (Lithotabs, Eskalith, Lithobid)

Clinical Context:  Patients intolerant to iodine can be treated with lithium, which also impairs thyroid hormone release. Can be used in patients who cannot take PTU or MMI. Use of iodine alone is debatable.

Class Summary

Act in a manner similar to iodine but is not routinely used because of transient effect and risk of potentially serious adverse effects. Now primarily used as a backup agent when other first-line agents are contraindicated because of hypersensitivity or toxicity.

Amiodarone (Cordarone)

Clinical Context:  Case report described successful normalization of thyroid hormone level in a patient with Graves disease who had fulminant PTU-induced hepatitis. However, experience and information in treatment of Graves disease is scant.

Class Summary

Amiodarone, an iodinated benzofuran, is an important antiarrhythmic medication that also alters thyroid hormone metabolism. High iodine content of this molecule (37.5%) is responsible for hypothyroidism. On the other hand, amiodarone can lead to hyperthyroidism through 2 complex mechanisms. Type I amiodarone-induced thyrotoxicosis is due to increased thyroid hormone synthesis and release in patients with multinodular goiter or Graves disease, while type II amiodarone-induced thyrotoxicosis is a destructive thyroiditis with release of preformed thyroid hormone.

Prednisone (Sterapred)

Clinical Context:  Has been customarily used in management of Graves ophthalmopathy. Other oral glucocorticoids at equipotent doses may also be effective.

Methylprednisolone (Solu-Medrol)

Clinical Context:  Has been customarily used for high-dose pulse steroid therapy in management of Graves ophthalmopathy. Other glucocorticoids at equipotent doses may also be effective. Intravenous high dose glucocorticoid therapy may be more effective and better tolerated than oral steroid therapy in the management of Graves ophthalmopathy (Aktaran, 2007).

Dexamethasone (Decadron)

Clinical Context:  In healthy persons, induces decrease in serum T3 levels without a change in serum T4 levels, suggesting an effect of dexamethasone on peripheral T3-to-T4 conversion.

In patients with Graves hyperthyroidism, induces rapid fall in serum thyroid hormone levels. Changes are too rapid to be explained by a steroid-induced fall in the level of a circulating IgG thyroid stimulator (TSI). Mechanism for this observation is unclear.

Class Summary

Graves disease is an autoimmune disease. Although glucocorticoids have been shown to decrease T4-to-T3 conversion and decrease thyroid hormones by yet undiscovered mechanisms, the adverse effect profile of long-term glucocorticoid therapy makes it unattractive for long-term management of Graves hyperthyroidism. However, glucocorticoids may have a role in rapidly lowering thyroid hormone levels in the clinical setting of thyroid storm. With regard to Graves ophthalmopathy, current evidence indicates that glucocorticoids represent the only class of drug therapy that, either alone or combined with other therapies, has an unequivocal role in management.

Metoprolol (Lopressor, Toprol XL)

Clinical Context:  Selective beta1-adrenergic receptor blocker that decreases automaticity of contractions. Useful in treating cardiac arrhythmias resulting from hyperthyroidism. During IV administration, carefully monitor BP, heart rate, and ECG.

What is Graves disease?Which other autoimmune diseases are associated with Graves disease?What is the pathophysiology of Graves disease?What is the role of the thyroid gland in the pathophysiology of Graves disease?Which genes are implicated in the pathogenesis of in Graves disease?What are the pathophysiologic mechanisms for Graves disease?What is the prevalence of Graves disease in the US?What is the global prevalence of Graves disease?What are complications of Graves disease?What is the pathogenesis of osteoporosis in Graves disease?What is the pathogenesis of hyperthyroidism in Graves disease?What are the cardiac complications of Graves disease?What are the psychiatric manifestations of Graves disease?What are the dermatologic manifestations of Graves disease?What are the ophthalmologic manifestations of Graves disease?What are the complications of Graves disease during pregnancy?What are the signs and symptoms of apathetic hyperthyroidism in Graves disease?What is the prevalence of coexisting autoimmune disorders in Graves disease?What are the racial predilections for Graves disease?How does the prevalence of Graves disease vary by sex?In which age groups is Graves disease most prevalent?What is the prognosis of Graves disease?What is included in the patient education information for Graves disease?What should be the focus of the history in suspected Graves disease?What are the classic features of Graves disease?Which symptoms may be present in young adults of Southeast Asian descent with Graves disease?What are the symptoms of Graves disease?Which medical condition is most apparent in the physical findings of Graves disease?Which physical findings are characteristic of Graves disease?Which physical findings are diagnostic of Graves disease?What are the common physical findings in Graves disease?What are the ophthalmologic findings suggestive of Graves disease?What are the endocrine symptoms in Graves disease?Which factors increase the mortality rate for Graves disease?Which immune mechanisms are etiologic factors in Graves disease?Which factors influences the autoimmune process Graves disease?What is the role of genetics in Graves disease?What is the role of environmental factors in the etiology of Graves disease?Which disorders should be included in the differential diagnoses for Graves disease?What are the differential diagnoses for Graves Disease?How are patients screened for Graves disease?What is the role of liver function test results in the diagnosis of Graves disease?What is the role of CBC count in the diagnosis of Graves disease?What are the significance of a finding of gynecomastia in Graves disease?What is the relationship between Graves disease and diabetes?What is the role of a fasting lipid profile in the diagnosis of Graves disease?What is the role of thyrotropin-releasing hormone testing in the diagnosis of Grave disease?What is the role of serum antibodies testing in the diagnosis of Graves disease?What is the role of radioactive iodine scanning in the diagnosis of Graves disease?What is the role of ultrasound in the diagnosis of Graves disease?What is the role of CT scanning and MRI in the diagnosis of Graves disease?Which histologic findings are characteristic of Graves disease?What are the goals of treatment for Graves disease?Which treatments for children with Graves disease have the greatest incidence of adverse events?What are the guidelines for radioactive iodine therapy in the treatment of Graves disease?What are the clinical phases of Graves ophthalmopathy?What is the frequency of disease progression for Graves ophthalmopathy?What are the treatment options for Graves ophthalmopathy?How are hyperthyroidism and hypothyroidism managed in patients with Graves ophthalmopathy?What are the treatment options for mild-to-moderate Graves ophthalmopathy?Which medications are used to treat severe or progressive Graves ophthalmopathy?What is the role of surgical interventions in the treatment of Graves ophthalmopathy?What is the role of orbital radiotherapy in the treatment of Graves ophthalmopathy?What is the role of gamma knife surgery in the treatment of Graves ophthalmopathy?When is surgical management of Graves ophthalmopathy performed?Which novel treatments are under investigation for Graves ophthalmopathy?What is the prevalence of pretibial (localized dermopathy) myxedema in Graves disease?What are the clinical manifestations of pretibial (localized dermopathy) myxedema in Graves disease?What is the treatment for mild pretibial (localized dermopathy) myxedema in Graves disease?What is the role of steroidal therapy in the treatment of pretibial (localized dermopathy) myxedema in Graves disease?What is acropachy in Graves disease?In what setting are patients with Graves disease usually treated?What is the role of inpatient care for the management of Graves disease?What are the indications for surgical care in the treatment of Graves disease?What in the preoperative preparation for thyroidectomy in patients with Graves disease?What are the risks and benefits of endoscopic surgery compared to open surgery for the treatment of Graves disease?What are the surgical options for the treatment of Graves ophthalmopathy?When is consultation with an endocrinologist indicated in the treatment of Graves disease?When is consultation with an ophthalmologist indicated for the treatment of Graves disease?When is consultation with a dermatologist indicated for the treatment of Graves disease?What is the role of dietary iodine intake in Graves disease?What is the role of dietary iodine in the treatment of Graves disease?What is the role of dietary iodine in the remission of Graves disease?What are the activity recommendations for patients with Graves disease?What are the possible complications of treatment for Graves disease?How is Graves disease prevented?What is included in the long-term monitoring of patients with Graves disease?What are the ATA/AACE guidelines key recommendations for the treatment and management of Graves disease?What are the goals of drug treatment for Graves disease?Which medications in the drug class Beta-adrenergic Blocker are used in the treatment of Graves Disease?Which medications in the drug class Glucocorticoids are used in the treatment of Graves Disease?Which medications in the drug class Antiarrhythmics are used in the treatment of Graves Disease?Which medications in the drug class Antidepressants are used in the treatment of Graves Disease?Which medications in the drug class Bile acid sequestrants are used in the treatment of Graves Disease?Which medications in the drug class Iodines are used in the treatment of Graves Disease?Which medications in the drug class Beta-adrenergic blocker are used in the treatment of Graves Disease?Which medications in the drug class Antithyroid agents are used in the treatment of Graves Disease?

Author

Sai-Ching Jim Yeung, MD, PhD, FACP, Professor of Medicine, Department of Emergency Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Celgene, Inc.<br/>Received research grant from: DepoMed and Bristol-Myer-Squibb.

Coauthor(s)

Alice Cua Chiu, MD, Associate Affiliate, Department of Internal Medicine, Division of Endocrinology, Bayshore Medical Center

Disclosure: Nothing to disclose.

Mouhammed Amir Habra, MD, Endocrine Fellow, Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Kent Wehmeier, MD, Professor, Department of Internal Medicine, Division of Endocrinology, Diabetes, and Metabolism, St Louis University School of Medicine

Disclosure: Nothing to disclose.

Chief Editor

Romesh Khardori, MD, PhD, FACP, Professor of Endocrinology, Director of Training Program, Division of Endocrinology, Diabetes and Metabolism, Strelitz Diabetes and Endocrine Disorders Institute, Department of Internal Medicine, Eastern Virginia Medical School

Disclosure: Nothing to disclose.

Additional Contributors

Steven R Gambert, MD, Professor of Medicine, Johns Hopkins University School of Medicine; Director of Geriatric Medicine, University of Maryland Medical Center and R Adams Cowley Shock Trauma Center

Disclosure: Nothing to disclose.

References

  1. Ellis H. Robert Graves: 1796-1852. Br J Hosp Med (Lond). 2006 Jun. 67(6):313. [View Abstract]
  2. Cruz AA, Akaishi PM, Vargas MA, de Paula SA. Association between thyroid autoimmune dysfunction and non-thyroid autoimmune diseases. Ophthal Plast Reconstr Surg. 2007 Mar-Apr. 23(2):104-8. [View Abstract]
  3. Jacobson EM, Tomer Y. The CD40, CTLA-4, thyroglobulin, TSH receptor, and PTPN22 gene quintet and its contribution to thyroid autoimmunity: back to the future. J Autoimmun. 2007 Mar-May. 28(2-3):85-98. [View Abstract]
  4. Iwama S, Ikezaki A, Kikuoka N, et al. Association of HLA-DR, -DQ genotype and CTLA-4 gene polymorphism with Graves' disease in Japanese children. Horm Res. 2005. 63(2):55-60. [View Abstract]
  5. Chu X, Pan CM, Zhao SX, et al. A genome-wide association study identifies two new risk loci for Graves' disease. Nat Genet. 2011 Aug 14. 43(9):897-901. [View Abstract]
  6. Bell L, Hunter AL, Kyriacou A, Mukherjee A, Syed AA. Clinical diagnosis of Graves' or non-Graves' hyperthyroidism compared to TSH receptor antibody test. Endocr Connect. 2018 Mar 12. 2017:7354673. [View Abstract]
  7. Douglas RS, Afifiyan NF, Hwang CJ, et al. Increased generation of fibrocytes in thyroid-associated ophthalmopathy. J Clin Endocrinol Metab. 2010 Jan. 95(1):430-8. [View Abstract]
  8. Chu X, Pan CM, Zhao SX, Liang J, Gao GQ, Zhang XM, et al. A genome-wide association study identifies two new risk loci for Graves' disease. Nat Genet. 2011 Aug 14. 43(9):897-901. [View Abstract]
  9. Furszyfer J, Kurland LT, McConahey WM, Elveback LR. Graves' disease in Olmsted County, Minnesota, 1935 through 1967. Mayo Clin Proc. 1970 Sep. 45(9):636-44. [View Abstract]
  10. Tunbridge WM, Evered DC, Hall R, et al. The spectrum of thyroid disease in a community: the Whickham survey. Clin Endocrinol (Oxf). 1977 Dec. 7(6):481-93. [View Abstract]
  11. Vanderpump MP, Tunbridge WM, French JM, et al. The incidence of thyroid disorders in the community: a twenty-year follow-up of the Whickham Survey. Clin Endocrinol (Oxf). 1995 Jul. 43(1):55-68. [View Abstract]
  12. Riis AL, Jørgensen JO, Gjedde S, et al. Whole body and forearm substrate metabolism in hyperthyroidism: evidence of increased basal muscle protein breakdown. Am J Physiol Endocrinol Metab. 2005 Jun. 288(6):E1067-73. [View Abstract]
  13. Nayak B, Burman K. Thyrotoxicosis and thyroid storm. Endocrinol Metab Clin North Am. 2006 Dec. 35(4):663-86, vii. [View Abstract]
  14. Burch HB, Wartofsky L. Life-threatening thyrotoxicosis. Thyroid storm. Endocrinol Metab Clin North Am. 1993 Jun. 22(2):263-77. [View Abstract]
  15. Park SE, Cho MA, Kim SH, Rhee Y, Kang ES, Ahn CW. The adaptation and relationship of FGF-23 to changes in mineral metabolism in Graves' disease. Clin Endocrinol (Oxf). 2007 Jun. 66(6):854-8. [View Abstract]
  16. Uchida T, Takeno K, Goto M, et al. Superior thyroid artery mean peak systolic velocity for the diagnosis of thyrotoxicosis in Japanese patients. Endocr J. 2010 Mar 6. [View Abstract]
  17. Bunevicius R, Prange AJ Jr. Psychiatric manifestations of Graves' hyperthyroidism: pathophysiology and treatment options. CNS Drugs. 2006. 20(11):897-909. [View Abstract]
  18. Vogel A, Elberling TV, Hørding M, Dock J, Rasmussen AK, Feldt-Rasmussen U. Affective symptoms and cognitive functions in the acute phase of Graves' thyrotoxicosis. Psychoneuroendocrinology. 2007 Jan. 32(1):36-43. [View Abstract]
  19. Schwartz KM, Fatourechi V, Ahmed DD, Pond GR. Dermopathy of Graves' disease (pretibial myxedema): long-term outcome. J Clin Endocrinol Metab. 2002 Feb. 87(2):438-46. [View Abstract]
  20. Kim JW, Ko J, Woo YJ, Bae HW, Yoon JS. Prevalence of Ocular Hypertension and Glaucoma as well as Associated Factors in Graves' Orbitopathy. J Glaucoma. 2018 Mar 19. [View Abstract]
  21. Boelaert K, Newby PR, Simmonds MJ, et al. Prevalence and relative risk of other autoimmune diseases in subjects with autoimmune thyroid disease. Am J Med. 2010 Feb. 123(2):183.e1-9. [View Abstract]
  22. Tun NN, Beckett G, Zammitt NN, Strachan MW, Seckl JR, Gibb FW. Thyrotropin Receptor Antibody Levels at Diagnosis and After Thionamide Course Predict Graves' Disease Relapse. Thyroid. 2016 Jul 6. [View Abstract]
  23. Rabon S, Burton AM, White PC. Graves' Disease in Children: Long Term Outcomes of Medical Therapy. Clin Endocrinol (Oxf). 2016 May 12. [View Abstract]
  24. Chen JL, Chiu HW, Tseng YJ, Chu WC. Hyperthyroidism is characterized by both increased sympathetic and decreased vagal modulation of heart rate: evidence from spectral analysis of heart rate variability. Clin Endocrinol (Oxf). 2006 Jun. 64(6):611-6. [View Abstract]
  25. Kung AW. Clinical review: Thyrotoxic periodic paralysis: a diagnostic challenge. J Clin Endocrinol Metab. 2006 Jul. 91(7):2490-5. [View Abstract]
  26. Ryan DP, da Silva MR, Soong TW, et al. Mutations in potassium channel Kir2.6 cause susceptibility to thyrotoxic hypokalemic periodic paralysis. Cell. 2010 Jan 8. 140(1):88-98. [View Abstract]
  27. Tanda ML, Piantanida E, Liparulo L, Veronesi G, Lai A, Sassi L, et al. Prevalence and Natural History of Graves' Orbitopathy in a Large Series of Patients with Newly Diagnosed Graves' Hyperthyroidism Seen at a Single Center. J Clin Endocrinol Metab. 2013 Feb 13. [View Abstract]
  28. Chung JO, Cho DH, Chung DJ, et al. Ultrasonographic features of papillary thyroid carcinoma in patients with Graves' disease. Korean J Intern Med. 2010 Mar. 25(1):71-6. [View Abstract]
  29. Pellegriti G, Mannarino C, Russo M, Terranova R, Marturano I, Vigneri R. Increased Mortality in Patients with Differentiated Thyroid Cancer Associated With Graves' Disease. J Clin Endocrinol Metab. 2013 Jan 24. [View Abstract]
  30. Brandt F, Thvilum M, Almind D, Christensen K, Green A, Hegedus L, et al. Graves´ disease and toxic nodular goiter are both associated with increased mortality but differ with respect to the cause of death. A Danish population-based register study. Thyroid. 2012 Dec 20. [View Abstract]
  31. Zaletel K, Krhin B, Gaberscek S, Pirnat E, Hojker S. The influence of the exon 1 polymorphism of the cytotoxic T lymphocyte antigen 4 gene on thyroid antibody production in patients with newly diagnosed Graves' disease. Thyroid. 2002 May. 12(5):373-6. [View Abstract]
  32. Zaletel K, Krhin B, Gaberscek S, Hojker S. Thyroid autoantibody production is influenced by exon 1 and promoter CTLA-4 polymorphisms in patients with Hashimoto's thyroiditis. Int J Immunogenet. 2006 Apr. 33(2):87-91. [View Abstract]
  33. Wang PW, Chen IY, Liu RT, Hsieh CJ, Hsi E, Juo SH. Cytotoxic T lymphocyte-associated molecule-4 gene polymorphism and hyperthyroid Graves' disease relapse after antithyroid drug withdrawal: a follow-up study. J Clin Endocrinol Metab. 2007 Jul. 92(7):2513-8. [View Abstract]
  34. Ban Y, Tozaki T, Taniyama M, Tomita M, Ban Y. Association of a C/T single-nucleotide polymorphism in the 5' untranslated region of the CD40 gene with Graves' disease in Japanese. Thyroid. 2006 May. 16(5):443-6. [View Abstract]
  35. Heward JM, Brand OJ, Barrett JC, Carr-Smith JD, Franklyn JA, Gough SC. Association of PTPN22 haplotypes with Graves' disease. J Clin Endocrinol Metab. 2007 Feb. 92(2):685-90. [View Abstract]
  36. Minich WB, Dehina N, Welsink T, Schwiebert C, Morgenthaler NG, Köhrle J. Autoantibodies to the IGF1 Receptor in Graves' Orbitopathy. J Clin Endocrinol Metab. 2013 Feb. 98(2):752-60. [View Abstract]
  37. Benvenga S, Guarneri F, Vaccaro M, et al. Homologies between proteins of Borrelia burgdorferi and thyroid autoantigens. Thyroid. 2004. 14:964-6. [View Abstract]
  38. Gangi E, Kapatral V, El-Azami El-Idrissi M, et al. Characterization of a recombinant Yersinia enterocolitica lipoprotein; implications for its role in autoimmune response against thyrotropin receptor. Autoimmunity. 2004 Sep-Nov. 37(6-7):515-20. [View Abstract]
  39. Al-Muqbel KM, Tashtoush RM. Patterns of thyroid radioiodine uptake: Jordanian experience. J Nucl Med Technol. 2010 Mar. 38(1):32-6. [View Abstract]
  40. De Bellis A, Sansone D, Coronella C, et al. Serum antibodies to collagen XIII: a further good marker of active Graves' ophthalmopathy. Clin Endocrinol (Oxf). 2005 Jan. 62(1):24-9. [View Abstract]
  41. Cappelli C, Pirola I, De Martino E, Agosti B, Delbarba A, Castellano M. The role of imaging in Graves' disease: A cost-effectiveness analysis. Eur J Radiol. 2007 Apr 23. [View Abstract]
  42. Markovic V, Eterovic D. Thyroid echogenicity predicts outcome of radioiodine therapy in patients with graves' disease. J Clin Endocrinol Metab. 2007 Sep. 92(9):3547-52. [View Abstract]
  43. Yasuda K, Miyoshi Y, Tachibana M, et al. Relationship between dose of antithyroid drugs and adverse events in pediatric patients with Graves' disease. Clin Pediatr Endocrinol. 2017 Jan. 26 (1):1-7. [View Abstract]
  44. Kubota S, Ohye H, Yano G, Nishihara E, Kudo T, Ito M. Two-day thionamide withdrawal prior to radioiodine uptake sufficiently increases uptake and does not exacerbate hyperthyroidism compared to 7-day withdrawal in Graves' disease. Endocr J. 2006 Oct. 53(5):603-7. [View Abstract]
  45. Bonnema SJ, Bennedbaek FN, Veje A, et al. Propylthiouracil before 131I therapy of hyperthyroid diseases: effect on cure rate evaluated by a randomized clinical trial. J Clin Endocrinol Metab. 2004. 89:4439-44. [View Abstract]
  46. Read CH Jr, 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 Sep. 89(9):4229-33. [View Abstract]
  47. Ceccarelli C, Canale D, Battisti P, Caglieresi C, Moschini C, Fiore E. Testicular function after 131I therapy for hyperthyroidism. Clin Endocrinol (Oxf). 2006 Oct. 65(4):446-52. [View Abstract]
  48. Rivkees SA, Dinauer C. An optimal treatment for pediatric Graves' disease is radioiodine. J Clin Endocrinol Metab. 2007 Mar. 92(3):797-800. [View Abstract]
  49. Chen YK, Lin CL, Chang YJ, Cheng FT, Peng CL, Sung FC. Cancer risk in patients with Graves' disease: A nationwide cohort study. Thyroid. 2013 Feb 19. [View Abstract]
  50. Ye X, Liu J, Wang Y, Bin L, Wang J. Increased serum VEGF and b-FGF in Graves' ophthalmopathy. Graefes Arch Clin Exp Ophthalmol. 2014 Oct. 252 (10):1639-44. [View Abstract]
  51. Stein JD, Childers D, Gupta S, Talwar N, Nan B, Lee BJ, et al. Risk factors for developing thyroid-associated ophthalmopathy among individuals with Graves disease. JAMA Ophthalmol. 2015 Mar. 133 (3):290-6. [View Abstract]
  52. Watanabe N, Noh JY, Kozaki A, Iwaku K, Sekiya K, Kosuga Y, et al. Radioiodine-Associated Exacerbation of Graves' Orbitopathy in the Japanese Population: Randomized Prospective Study. J Clin Endocrinol Metab. 2015 Jul. 100 (7):2700-8. [View Abstract]
  53. Shiber S, Stiebel-Kalish H, Shimon I, Grossman A, Robenshtok E. Glucocorticoid regimens for prevention of Graves' ophthalmopathy progression following radioiodine treatment: systematic review and meta-analysis. Thyroid. 2014 Oct. 24 (10):1515-23. [View Abstract]
  54. Bartalena L, Marcocci C, Bogazzi F, et al. Relation between therapy for hyperthyroidism and the course of Graves' ophthalmopathy. N Engl J Med. 1998 Jan 8. 338(2):73-8. [View Abstract]
  55. Bartalena L, Marcocci C, Bogazzi F, Panicucci M, Lepri A, Pinchera A. Use of corticosteroids to prevent progression of Graves' ophthalmopathy after radioiodine therapy for hyperthyroidism. N Engl J Med. 1989 Nov 16. 321(20):1349-52. [View Abstract]
  56. Bartalena L, Tanda ML, Piantanida E, Lai A, Pinchera A. Relationship between management of hyperthyroidism and course of the ophthalmopathy. J Endocrinol Invest. 2004 Mar. 27(3):288-94. [View Abstract]
  57. Macchia PE, Bagattini M, Lupoli G, et al. High-dose intravenous corticosteroid therapy for Graves' ophthalmopathy. J Endocrinol Invest. 2001. 24:152-8. [View Abstract]
  58. Gibson A, Czyz CN. Graves Disease, Orbital Decompression. 2018 Jan. [View Abstract]
  59. Sisti E, Coco B, Menconi F, Leo M, Rocchi R, Latrofa F, et al. Intravenous glucocorticoid therapy for Graves' ophthalmopathy and acute liver damage: an epidemiological study. Eur J Endocrinol. 2015 Mar. 172 (3):269-76. [View Abstract]
  60. Liao SL, Huang SW. Correlation of retrobulbar volume change with resected orbital fat volume and proptosis reduction after fatty decompression for Graves ophthalmopathy. Am J Ophthalmol. 2011 Mar. 151(3):465-9.e1. [View Abstract]
  61. Wakelkamp IM, Tan H, Saeed P, et al. Orbital irradiation for Graves' ophthalmopathy: Is it safe? A long-term follow-up study. Ophthalmology. 2004 Aug. 111(8):1557-62. [View Abstract]
  62. Seals KF, Lee EW, Cagnon CH, Al-Hakim RA, Kee ST. Radiation-Induced Cataractogenesis: A Critical Literature Review for the Interventional Radiologist. Cardiovasc Intervent Radiol. 2015 Sep 24. [View Abstract]
  63. Rajendram R, Bunce C, Lee RW, Morley AM. Orbital radiotherapy for adult thyroid eye disease. Cochrane Database Syst Rev. 2012 Jul 11. 7:CD007114. [View Abstract]
  64. Dickinson AJ, Vaidya B, Miller M, Coulthard A, Perros P, Baister E. Double-blind, placebo-controlled trial of octreotide long-acting repeatable (LAR) in thyroid-associated ophthalmopathy. J Clin Endocrinol Metab. 2004 Dec. 89(12):5910-5. [View Abstract]
  65. Wemeau JL, Caron P, Beckers A, et al. Octreotide (long-acting release formulation) treatment in patients with graves' orbitopathy: clinical results of a four-month, randomized, placebo-controlled, double-blind study. J Clin Endocrinol Metab. 2005. 90:841-8. [View Abstract]
  66. Stan MN, Garrity JA, Bradley EA, Woog JJ, Bahn MM, Brennan MD. Randomized, double-blind, placebo-controlled trial of long-acting release octreotide for treatment of Graves' ophthalmopathy. J Clin Endocrinol Metab. 2006 Dec. 91(12):4817-24. [View Abstract]
  67. Durrani OM, Reuser TQ, Murray PI. Infliximab: a novel treatment for sight-threatening thyroid associated ophthalmopathy. Orbit. 2005 Jun. 24(2):117-9. [View Abstract]
  68. Salvi M, Vannucchi G, Campi I, Currò N, Dazzi D, Simonetta S. Treatment of Graves' disease and associated ophthalmopathy with the anti-CD20 monoclonal antibody rituximab: an open study. Eur J Endocrinol. 2007 Jan. 156(1):33-40. [View Abstract]
  69. Stan MN, Garrity JA, Carranza Leon BG, Prabin T, Bradley EA, Bahn RS. Randomized controlled trial of rituximab in patients with Graves' orbitopathy. J Clin Endocrinol Metab. 2015 Feb. 100 (2):432-41. [View Abstract]
  70. Salvi M, Vannucchi G, Currò N, Campi I, Covelli D, Dazzi D, et al. Efficacy of B-cell targeted therapy with rituximab in patients with active moderate to severe Graves' orbitopathy: a randomized controlled study. J Clin Endocrinol Metab. 2015 Feb. 100 (2):422-31. [View Abstract]
  71. Ebner R, Devoto MH, Weil D, et al. Treatment of thyroid associated ophthalmopathy with periocular injections of triamcinolone. Br J Ophthalmol. 2004 Nov. 88(11):1380-6. [View Abstract]
  72. Finamor FE, Martins JR, Nakanami D, Paiva ER, Manso PG, Furlanetto RP. Pentoxifylline (PTX)--an alternative treatment in Graves' ophthalmopathy (inactive phase): assessment by a disease specific quality of life questionnaire and by exophthalmometry in a prospective randomized trial. Eur J Ophthalmol. 2004 Jul-Aug. 14(4):277-83. [View Abstract]
  73. Grodski S, Stalberg P, Robinson BG, Delbridge LW. Surgery versus Radioiodine Therapy as Definitive Management for Graves' Disease: The Role of Patient Preference. Thyroid. 2007 Feb. 17(2):157-60. [View Abstract]
  74. Genovese BM, Noureldine SI, Gleeson EM, Tufano RP, Kandil E. What is the best definitive treatment for graves' disease? A systematic review of the existing literature. Ann Surg Oncol. 2013 Feb. 20(2):660-7. [View Abstract]
  75. Pradeep PV, Agarwal A, Baxi M, Agarwal G, Gupta SK, Mishra SK. Safety and efficacy of surgical management of hyperthyroidism: 15-year experience from a tertiary care center in a developing country. World J Surg. 2007 Feb. 31(2):306-12; discussion 313. [View Abstract]
  76. Panzer C, Beazley R, Braverman L. Rapid preoperative preparation for severe hyperthyroid Graves' disease. J Clin Endocrinol Metab. 2004 May. 89(5):2142-4. [View Abstract]
  77. Piantanida E. Preoperative management in patients with Graves' disease. Gland Surg. 2017 Oct. 6 (5):476-81. [View Abstract]
  78. Erbil Y, Ozluk Y, Giris M, Salmaslioglu A, Issever H, Barbaros U. Effect of lugol solution on thyroid gland blood flow and microvessel density in the patients with Graves' disease. J Clin Endocrinol Metab. 2007 Jun. 92(6):2182-9. [View Abstract]
  79. Randle RW, Bates MF, Long KL, Pitt SC, Schneider DF, Sippel RS. Impact of potassium iodide on thyroidectomy for Graves' disease: Implications for safety and operative difficulty. Surgery. 2018 Jan. 163 (1):68-72. [View Abstract]
  80. Calissendorff J, Falhammar H. Lugol's solution and other iodide preparations: perspectives and research directions in Graves' disease. Endocrine. 2017 Dec. 58 (3):467-73. [View Abstract]
  81. Zhang Y, Dong Z, Li J, Yang J, Yang W, Wang C. Comparison of endoscopic and conventional open thyroidectomy for Graves' disease: A meta-analysis. Int J Surg. 2017 Feb 22. 40:52-9. [View Abstract]
  82. Alsuhaibani AH, Carter KD, Policeni B, Nerad JA. Effect of orbital bony decompression for Graves' orbitopathy on the volume of extraocular muscles. Br J Ophthalmol. 2011 Sep. 95(9):1255-8. [View Abstract]
  83. Hiraiwa T, Ito M, Imagawa A, et al. High diagnostic value of a radioiodine uptake test with and without iodine restriction in Graves' disease and silent thyroiditis. Thyroid. 2004 Jul. 14(7):531-5. [View Abstract]
  84. Anagnostis P, Adamidou F, Polyzos SA, Katergari S, Karathanasi E, Zouli C, et al. Predictors of long-term remission in patients with Graves' disease: a single center experience. Endocrine. 2013 Feb 11. [View Abstract]
  85. Sato H, Sasaki N, Minamitani K, Minagawa M, Kazukawa I, Sugihara S, et al. Higher dose of methimazole causes frequent adverse effects in the management of Graves' disease in children and adolescents. J Pediatr Endocrinol Metab. 2012. 25(9-10):863-7. [View Abstract]
  86. [Guideline] Ross DS, Burch HB, Cooper DS, et al. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid. 2016 Oct. 26 (10):1343-1421. [View Abstract]
  87. Rivkees SA, Stephenson K, Dinauer C. Adverse events associated with methimazole therapy of Graves' disease in children. Int J Pediatr Endocrinol. 2010. 2010:176970. [View Abstract]
  88. Mohlin E, Filipsson Nyström H, Eliasson M. Long-term prognosis after medical treatment of Graves' disease in a northern Swedish population 2000-2010. Eur J Endocrinol. 2014 Mar. 170 (3):419-27. [View Abstract]
  89. Liu X, Shi B, Li H. Valuable predictive features of relapse of Graves' disease after antithyroid drug treatment. Ann Endocrinol (Paris). 2015 Oct 26. [View Abstract]
  90. Villagelin D, Romaldini JH, Santos RB, Milkos AB, Ward LS. Outcomes in Relapsed Graves' Disease Patients Following Radioiodine or Prolonged Low Dose of Methimazole Treatment. Thyroid. 2015 Oct 20. [View Abstract]
  91. Salvi M, Campi I. Medical Treatment of Graves' Orbitopathy. Horm Metab Res. 2015 Sep. 47 (10):779-88. [View Abstract]
  92. Prasek K, Płazińska MT, Krolicki L. Diagnosis and treatment of Graves' disease with particular emphasis on appropriate techniques in nuclear medicine. General state of knowledge. Nucl Med Rev Cent East Eur. 2015. 18 (2):110-6. [View Abstract]
  93. Jankauskiene J, Jarusaitiene D. The Influence of Juvenile Graves' Ophthalmopathy on Graves' Disease Course. J Ophthalmol. 2017. 2017:4853905. [View Abstract]

Graves disease. Varying degrees of manifestations of Graves ophthalmopathy.

Pathophysiologic mechanisms of Graves disease relating thyroid-stimulating immunoglobulins to hyperthyroidism and ophthalmopathy. T4 is levothyroxine. T3 is triiodothyronine.

Pathophysiologic mechanisms of Graves disease relating thyroid-stimulating immunoglobulins to hyperthyroidism and ophthalmopathy. T4 is levothyroxine. T3 is triiodothyronine.

Graves disease. Varying degrees of manifestations of Graves ophthalmopathy.