Archives

Hyperthyroidism in Aging

ABSTRACT

 

Hyperthyroidism in the elderly is a serious clinical condition that is associated with significant morbidity. It may be difficult to diagnose due to the confounding effects of drugs and acute or chronic illnesses on the interpretation of thyroid function tests. In addition, there is a relative paucity of typical hyperadrenergic symptoms in older patients with hyperthyroidism, who instead may present with unexplained weight loss, neurocognitive changes, or cardiovascular effects. Of particular concern is the elevated risk of atrial fibrillation and cardiovascular complications in this age group. There is increasing evidence that even mild (subclinical) hyperthyroidism in the elderly is associated with these risks. Graves’ Disease and toxic multinodular goiter are the most common etiologies of hyperthyroidism in the elderly, although other causes of hyperthyroidism also occur. The use of amiodarone or administration of iodinated contrast agents can also lead to hyperthyroidism, and are commonly prescribed to older patients. Radioiodine or thionamide therapy are typically used to treat hyperthyroidism in older patients. Treatment decisions must be individualized, taking into account projected lifespan, comorbidities, and side effects of therapy.

 

PREVALENCE OF HYPERTHYROIDISM IN AGING

 

Hyperthyroidism is a common disorder (1); a population-based survey (2) conducted over 40 years ago revealed a prevalence in the general UK population of around 2.7% in women (10-fold less in men) and of undiagnosed disease in around 0.5% of women. A more recent population-based survey in the United States revealed a prevalence of hyperthyroidism of 1.3%, with no difference between men and women (3). This prevalence decreases to 0.4% if one excludes patients with known thyroid disease and those taking thyroid hormone preparations, indicating that many cases of hyperthyroidism are due to overtreatment with exogenous thyroid hormone.

 

A number of studies have reported the prevalence of hyperthyroidism specifically in elderly populations. Prevalence rates vary depending on whether patients taking thyroid hormone are included, but most surveys report that approximately 1–3% of subjects over the age of 60-65 years have hyperthyroidism (2-7).  If one excludes patients taking thyroid hormone, prevalence rates of hyperthyroidism appear similar in younger and older populations (3).

CLINICAL CONSEQUENCES OF HYPERTHYROIDISM

 

Classical symptoms and signs of thyrotoxicosis are shown in Table 1 (1).  While some or all of these may be present in elderly subjects with thyrotoxicosis, the clinical picture is often different in this age group (8,9). Problems such as weight loss and depression or agitation may predominate - so-called "apathetic" thyrotoxicosis, a condition in which more typical symptoms and signs reflecting sympathetic activation such as tremor and hyperactivity are absent (10-12). Instead, cardiovascular symptoms and signs often predominate in older patients, including atrial fibrillation. Other findings more common in older patients with hyperthyroidism include fatigue, anorexia, weight loss, apathy, agitation, or cognitive decline (11-14). Particularly in this age group, the diagnosis of thyrotoxicosis should also be considered in the presence of other symptoms and signs considered "non-specific" in nature, such as muscle weakness, persistent vomiting, hypercalcemia, and worsening osteoporosis.

 

Table 1. Symptoms and Signs in Hyperthyroidism

Symptoms

Signs

1. Weight loss

2. Sweating/heat intolerance

3. Nervousness/agitation

4. Tiredness

5. Muscle weakness

6. Palpitation

7. Shortness of breath

8. Tremor

 

 

1.Tremor

2. Hyperactivity

3. Proximal myopathy

4. Sinus tachycardia

5. Atrial fibrillation/atrial dysrhythmias

6. Systolic hypertension

7. Goiter

8. Lid lag/lid retraction

9. Ophthalmopathy*

10. Pretibial myxedema*

11. Thyroid acropachy*

 

* specific for Graves’ Disease

Cardiovascular Complications

 

Cardiovascular complications of thyrotoxicosis are especially common in the elderly and may be a cause of significant morbidity and mortality (1,15). A number of studies have reported increased all-cause and cardiovascular mortality, and increased risks of atrial fibrillation, arterial embolism, acute myocardial infarction, heart failure, venous thromboembolism, and stroke in hyperthyroid patients, compared to euthyroid controls (16-20). Risks are higher in older subjects and in untreated or undertreated groups, with a direct association between the duration of suppressed TSH levels and mortality in both untreated and treated patients (19). Risks decrease with treatment, regardless of treatment modality (21,22). All-cause mortality is also increased in treated hypothyroid patients with suppressed TSH levels, highlighting the importance of avoiding overtreatment.

 

In addition to classical findings of sinus tachycardia and systolic hypertension, it is well recognized that atrial fibrillation complicates thyrotoxicosis in about 15% of cases (23). The incidence of this complication rises with age, so it is observed more frequently in the elderly (24). It has been estimated that atrial fibrillation occurs at least three times more commonly in those with thyrotoxicosis than those without. Development of atrial fibrillation may itself lead to deteriorating cardiac status, especially in the presence of pre-existing heart disease, and it may also be associated with embolic complications, especially cerebral embolism (25). These influences probably contribute significantly to the increased cardiovascular and cerebrovascular mortality described above. Furthermore, the likelihood of spontaneous restoration of sinus rhythm in those with atrial fibrillation complicating thyrotoxicosis lessens with age, probably reflecting the presence of underlying ischemic, hypertensive, or valvular heart disease (26).

In view of these cardiovascular manifestations/complications, the diagnosis of thyrotoxicosis should be suspected in all subjects presenting with atrial fibrillation, worsening heart failure, systolic hypertension, and deteriorating ischemic heart disease. Nonetheless, case-finding studies have shown that thyrotoxicosis accounts for less than 5% of newly diagnosed cases of atrial fibrillation (23).

Bone Metabolism and Hyperthyroidism

 

The other significant consequence of thyrotoxicosis is its effect on bone metabolism. Overt hyperthyroidism is associated with increased bone turnover and reduction in bone mineral density (27). Meta-analysis of available data (28) has shown that this influence is especially marked in estrogen deficient postmenopausal women. While antithyroid treatment results in an improvement in bone mineral density, recovery is incomplete so risks of osteoporosis associated with aging, especially in women, are exacerbated (29). Several large-scale epidemiological studies (16,30) have revealed independent associations between a history of thyrotoxicosis and risk of fracture of the femur.

 DIAGNOSIS OF HYPERTHYROIDISM

 

It is essential that a clinical suspicion of thyrotoxicosis is confirmed or refuted by biochemical testing before further investigation or treatment is contemplated (1). The single most important biochemical test is measurement of serum TSH. If the serum TSH concentration is within the normal range, then a diagnosis of thyrotoxicosis is effectively ruled out. Exceptions to this rule are rare TSH-dependent causes of hyperthyroidism, such as TSH-secreting tumors of the pituitary and syndromes of thyroid hormone resistance, although these diagnoses are more typically associated with a modest rise in TSH (with raised serum thyroid hormones, as opposed to the usual pattern of raised TSH in conjunction with low thyroid hormone levels).

 

Studies of healthy elderly subjects have shown that serum concentrations of thyroxine (T4) and tri-iodothyronine (T3) are unchanged compared with younger age groups (31). Analysis of large U.S. population-based normative data suggest that there is a slight increase in the upper limit of normal TSH levels with aging, but the lower limit of normal TSH levels remains relatively unchanged (32). Therefore, in a healthy older patient, a low or suppressed TSH level suggests hyperthyroidism. On the other hand, "non-thyroidal" illnesses and drug therapies that alter tests of thyroid function are more common with increasing age. These effects typically lead to reduced peripheral conversion of T4 to T3 and reduction in serum T3 concentrations. Serum TSH may be unaffected by illness, although a reduction in TSH is commonly seen, as is a modest elevation in TSH particularly during the recovery phase of illness (33). Therefore, in an acutely or chronically ill older patient, interpretation of a low TSH level must be done with caution, as low serum TSH, especially if below the normal range but nonetheless detectable, often reflects a "non-thyroidal" illness or therapy with a wide variety of drugs (34) (Table 2). A diagnosis of thyrotoxicosis should be confirmed biochemically by measurement of serum free thyroxine (T4) (and in some cases T3 if free T4 is in the high/normal range and T3-toxicosis is therefore suspected).

 

Table 2. Effect of Drugs on Tests of Thyroid Function

Drug

 

Serum T4

 

Serum T3

 

Serum TSH

 

 Dopamine

 

¯, ®

 

 

¯, ®

 

¯

 

Glucocorticoids

 

¯, ®

 

¯, ®

 

¯

 

Estrogens

 

­ total T4

 

­ total T3

 

®

 

Anticonvulsants

 

¯, ®

 

¯, ®

 

®

 

Acetylsalicylic acid

 

­, ®

 

­, ®

 

¯ ®

 

Amiodarone

 

­

 

¯

 

variable

 

Heparin

 

­, ®

 

­, ®

 

¯, ®

 

Fenclofenac

 

¯, ®

 

¯, ®

 

®

 

Anabolic steroids

 

¯ total T4

 

¯ total T3

 

®

 

 

In the majority of cases of thyrotoxicosis, a typical biochemical picture of elevated free T4 and T3 with associated undetectable TSH will be observed. In some cases, a biochemical diagnosis of "T3-toxicosis" is evident, characterized by elevation of serum T3 in the absence of a rise in T4. This is typically observed in mild cases of toxic nodular hyperthyroidism and early in the course of Graves' hyperthyroidism. In some instances, the converse is true in that a rise in T3 is absent despite elevation in free T4 and suppression of TSH in a patient thought clinically to have thyrotoxicosis. This lack of rise in T3 may reflect the presence of another "non-thyroidal" illness, evident upon re-testing once the other morbidity is eliminated.

CAUSES OF THYROTOXICOSIS 

Graves' Disease and Toxic Nodular Hyperthyroidism

 

In iodine replete parts of the world, Graves' disease is the most common endogenous cause of thyrotoxicosis. In the elderly, however, toxic nodular hyperthyroidism becomes an important cause (1,35). In all age groups, toxic nodular hyperthyroidism is more common in areas of the world that are relatively iodine deficient (36). The natural history of goiter is of progression from the presence of diffuse thyroid enlargement to development of one or more nodules and eventual autonomous function of one or more of these nodules resulting in thyrotoxicosis. This natural history is typically long so the elderly patient presenting with thyrotoxicosis often describes the presence of a goiter for many years. A relatively rare cause is the presence of a single toxic adenoma - a benign tumor exhibiting autonomous secretion of thyroid hormones. This diagnosis accounts for less than 2% of cases of thyrotoxicosis occurring in the US (36). Biochemically, the development of autonomous function in a nodular goiter is first evidenced by suppression of serum TSH with normal serum concentrations of thyroid hormones ("subclinical" hyperthyroidism - see below), followed by elevation of serum T3 and free T4.

 

In many cases, the cause of thyrotoxicosis is obvious from the clinical picture (1,35). The diagnosis of Graves' disease may be evident by the presence of diffuse goiter and ophthalmopathy, whereas toxic nodular hyperthyroidism is characterized by the presence of a nodular goiter on examination of the neck. It should be noted, however, that the thyroid might be impalpable in about 30% of cases of Graves' disease or toxic nodular hyperthyroidism. If the cause of thyrotoxicosis is not obvious, further investigation may be warranted. The presence of thyroid autoantibodies (to thyroid peroxidase - TPO and/or thyroglobulin) is suggestive (but not diagnostic of) Graves' disease; TSH receptor antibodies are more specific for the diagnosis. Such antibodies are positive in 90% of Graves’ Disease cases, and are usually negative in cases of toxic nodular hyperthyroidism. If TSH receptor antibodies are positive in the presence of a nodular goiter, both conditions may co-exist. Radioisotope scanning, using technetium-99m or iodine-123, typically shows a diffuse pattern of uptake in Graves' disease, in contrast to the presence of multiple "hot" nodules with surrounding thyroid tissue not demonstrating any uptake in cases of toxic nodular hyperthyroidism (figure 1). Occasionally, a single "hot" nodule, with absent uptake elsewhere in the thyroid is observed. This finding suggests the presence of a toxic nodular adenoma.

Figure 1. Radionuclide imaging of the thyroid illustrating hot nodules in toxic nodular hyperthyroidism (right) which contrasts with a diffuse uptake in Graves' Disease (left)

 Other Causes of Thyrotoxicosis

 

Although Graves' disease and toxic nodular goiter are by far the most common causes of endogenous thyrotoxicosis in older patients, it is important to consider other diagnoses. As in other age groups, the elderly patient may develop transient thyroid hormone excess secondary to a temporary thyroiditis, i.e., destruction of the thyroid with release of pre-formed thyroid hormones (35). Sub-acute thyroiditis should be suspected if the patient complains of sore throat or neck tenderness, typically associated with symptoms of a viral illness or an upper respiratory tract infection. The diagnosis is confirmed by the finding of a raised erythrocyte sedimentation rate (ESR) and absent or very low uptake of iodine-123. This is an important diagnosis to make since antithyroid treatment with antithyroid drugs or radioiodine is inappropriate, because it is ineffective and because the condition resolves spontaneously (usually after a self-limiting period of hypothyroidism).  Silent thyroiditis has a similar clinical course as subacute thyroiditis, but the gland is not tender and there is no increase in ESR.  Both subacute and silent thyroiditis can occur in older patients, although the peak age range for these two conditions is among younger patients (37).

 

Iodine-induced thyroiditis should be considered in patients with a history of iodine ingestion (e.g., in the form of sea weed preparations or over the counter iodine containing compounds, such as expectorants) or after administration of iodine containing radiographic contrast agents (35). The diagnosis can be confirmed by the finding of low iodine uptake. This condition also remits spontaneously and radioiodine therapy is contraindicated. This diagnosis is more common in older patients, who are more likely to receive iodinated contrast agents and to have underlying multinodular goiters that predispose them to iodine-induced thyrotoxicosis.

 

Finally, it should be noted that exogenous thyrotoxicosis due to excessive doses of thyroid hormone in the treatment of hypothyroidism is quite common.  One study indicated that over 40% of older subjects taking thyroid hormone had low TSH levels, indicating excess thyroid hormone doses (38). A second study reported that iatrogenic thyrotoxicosis accounted for about 50% of low TSH events in a large cohort of subjects, with the highest rates in older women (39). A third study reported that thyroid hormone use increased 1.8-fold in the UK from 2001-2009, with decreasing TSH thresholds for initiating treatment. Of concern, 90% of treated subjects remained on L-T4 for > 5 years, and 16% had low or suppressed TSH levels, indicating excessive doses (40). A fourth study reported that thyroid hormone use doubled in the U.S. from 1997-2016, from 4% to 8%, while expenditures for thyroid hormone tripled. Thyroid hormone use was higher in women, older individuals, and non-Hispanic whites (41). These reports clearly indicate that thyroid hormone is being over-prescribed, with high risks of overtreatment and potential clinical consequences, particularly in older subjects who may have underlying cardiac issues or osteoporosis.

 Amiodarone Induced Thyrotoxicosis

 

The diagnosis of thyroid dysfunction should be considered in an elderly patient prescribed the antiarrhythmic agent amiodarone. This drug is widely used in the older age group for control of dysrhythmias, particularly those associated with poor left ventricular function. Amiodarone is an iodine-containing compound that affects the results of tests of thyroid function, even in those who are euthyroid (35,42). Typically, amiodarone, through its effect on peripheral conversion of T4 to T3, results in modest reduction in serum concentrations of T3 (often to below the normal range) and modest elevation in serum T4 (often to above the normal range). TSH is typically slightly elevated early after commencement of treatment and normalizes later in euthyroid patients.  Therefore, beginning 2-3 months after amiodarone is started, the serum TSH level is an accurate indication of thyroid function.

 

Although amiodarone results in overt thyroid dysfunction in 5-10% of cases, it is important not to over-interpret mildly abnormal results of tests of thyroid function. Thyrotoxicosis should only be diagnosed in the presence of significant elevation of free T4, together with elevation in serum T3 and suppression of TSH; sometimes serum T3 is at the upper range of normal rather than elevated, probably because of associated "non-thyroidal" illness in this age group, together with the block of T4 to T3 conversion seen with amiodarone.

 TREATMENT OF THYROTOXICOSIS

 Antithyroid Drugs

 

The thionamides – methimazole (or its precursor drug carbimazole) and propylthiouracil - represent the mainstay of drug treatment of thyrotoxicosis (1,35). These drugs inhibit the oxidation and organification of iodide and hence block the synthesis of T4 and T3 early in their biosynthetic pathway. They represent the most effective and rapid means of reducing circulating thyroid hormone concentrations. They can be used in several ways: short-term in preparation of the patient for definitive treatment with radioiodine or surgery, medium term in the hope of inducing remission in cases of thyrotoxicosis due to Graves' disease, or long-term for control of clinical and biochemical thyroid hormone excess.

 

In many elderly patients, thionamides are used short-term in the preparation for curative treatment. A typical starting dose of methimazole is 20-30 mg per day as a single daily dose. In contrast, propylthiouracil is typically given in divided doses, the equivalent to methimazole 20 mg being 200mg. Doses higher than this are rarely required, since high doses have not been shown to be more effective in terms of restoration of euthyroidism in prospective studies (43,44). Since compliance is better and side effects are less frequent, methimazole or carbimazole are considered the drugs of choice, in preference to propylthiouracil (35). Serum free T4 should be checked 4-6 weeks after beginning therapy and the thionamide dose adjusted accordingly. It is usually possible to render the patient euthyroid (or near euthyroid) after 2-3 months, so they can proceed to curative therapy.

 

Drug side effects are relatively uncommon, but it is essential that all subjects (in whichever age group) be warned (preferably in writing) of the potential risk of agranulocytosis so that they present urgently for a full blood count if they develop a fever or sore throat. Agranulocytosis often, but not always, occurs in the first few weeks after beginning thionamide therapy and is probably more common in those taking higher doses (35). The latter observation represents a relative contraindication to doses of methimazole/carbimazole of greater than 20-30 mg per day; doses higher than this are rarely necessary in the elderly.

 

Other serious side effects can occur, notably antineutrophil cytoplasmic antibody-associated-vasculitis (typically associated with prescription of propylthiouracil), hepatitis, or pancreatitis (35,44), although these are rare. These serious complications, together with agranulocytosis, represent absolute contraindications to further use of thionamides. Less serious side effects such as pruritic rash are more common and can usually be managed conservatively, although sometimes a change in drug therapy from one thionamide to another is required (ATA guidelines).

 ANTITHYROID DRUGS AND GRAVES’ DISEASE

 

In general, remission rates following thionamide therapy in Graves' hyperthyroidism are less than 50%, nonetheless, there is some evidence that the remission rate in Graves’ may be higher in the elderly age group, probably reflecting the presence of milder disease. If the objective is to achieve remission or "cure" of thyrotoxicosis secondary to Graves' disease, then thionamide treatment should be prescribed for a course of not less than 12 or 18 months, since shorter courses are associated with a lower rate of remission (35). Drug doses should be titrated according to serum concentrations of free T4 (serum TSH may remain suppressed for months); the majority of subjects will require a methimazole maintenance dose of 5-10 mg daily once normal fT4 levels are achieved (propylthiouracil 50-100mg daily in divided doses). Larger dose requirements are suggestive of poor compliance. Poor prognostic features for achieving long-term remission (35) (established in younger age groups) include male sex, the presence of a large goiter and biochemically severe disease at diagnosis. Most relapses of Graves' thyrotoxicosis occur 3-6 months after thionamide withdrawal.

 

Although standard recommendations for treating Graves’ disease with thionamides include a 12-18 month course of therapy, recent studies suggest that long term thionamide therapy is safe and efficacious (46,47). This option may be particularly useful in older patients with limited life expectancies, since it leads to more rapid attainment of euthyroidism and lower rates of hypothyroidism than radioactive iodine or surgery (48). Updated guidelines for treating hyperthyroidism now include the option for long-term thionamide therapy (35).

 ANTITHYROID DRUGS AND TOXIC NODULAR HYPERTHYROIDISM

 

Time-limited courses of thionamides virtually never result in remission or cure of thyrotoxicosis secondary to toxic nodular goiter, although some spontaneous fluctuation in the severity of the disease is seen. Thionamides may thus be used short-term (as above) to induce euthyroidism prior to definitive treatment, but a time-limited course should not be prescribed in the hope of inducing cure. Recent studies show that long-term thionamide therapy is safe and efficacious in toxic nodular hyperthyroidism (47,49).  Once biochemical control has been achieved, biochemical monitoring every 3-6 months is desirable.

BETA-ADRENERGIC BLOCKING AGENTS AND OTHER DRUGS AS ADJUNCTIVE THERAPIES

 

Beta adrenergic blockers are useful adjuncts to thionamides in the management of thyrotoxicosis. In cases of thyroiditis or mild cases of hyperthyroidism proceeding to radioiodine, they may be the only additional treatment required. Beta adrenergic blockers act promptly to reduce symptoms and signs of tremor and to improve tachycardia and associated palpitations (35). Such agents should be used cautiously in elderly subjects with heart failure (although a beneficial effect often results because of amelioration of some of the cardiovascular effects of thyroid hormone excess) and in those with asthma or chronic obstructive pulmonary disease. Propranolol has been widely used in thyrotoxic subjects but requires multiple daily dosing; longer acting beta adrenergic blockers such as atenolol (50-100mg daily) may therefore be preferred.

 

Other adjunctive therapies include salicylates for relief of local pain and tenderness in cases of subacute thyroiditis; occasionally glucocorticoids such as prednisolone are required short-term.

Anticoagulation with coumarin derivatives such as warfarin should be considered in elderly subjects with thyrotoxicosis complicated by atrial fibrillation. This is driven by evidence for embolic complications. There have been no controlled trials of the use of anticoagulants in thyrotoxic atrial fibrillation, but overwhelming evidence of their efficacy in other settings argues in favor of their use in this situation (50), unless contraindications exist. Therapy to restore sinus rhythm should be considered but not until the patient has been rendered euthyroid. This therapy may comprise pharmacological cardioversion (with agents such as sotalol) or electrical cardioversion. Restoration of sinus rhythm is more likely in those whose atrial fibrillation is of short duration and in those without underlying heart disease (23), although rates of restoration of sinus rhythm may be relatively low, even with cardiologic intervention (24).

 Radioiodine Therapy

 

Radioiodine (I-131) is a reasonable therapy in elderly hyperthyroid subjects, as it can be administered by mouth in the outpatient setting and is associated with few side effects. Some patients notice sore throat or neck tenderness (reflecting a radiation thyroiditis), but this is usually mild and transient. Its long-term efficacy is well established (35). Reports of potential risks of secondary cancers following radioactive iodine therapy for hyperthyroidism have been inconsistent, but long-term risks appear modest, and are likely to be of less importance in older subjects (51-53). There are few, if any, contraindications to radioiodine therapy apart from inability to comply with local radiation protection regulations. Such compliance may be difficult to achieve in hospital or nursing home residents, those with urinary incontinence, and those with significant mental impairment. In such cases, long-term thionamide therapy is often the best practical option (see above).

 

A relative contraindication to the use of radioiodine in cases of Graves' thyrotoxicosis is the presence of moderate or severe ophthalmopathy. There is a slightly increased risk of development or worsening of pre-existing thyroid eye disease in those treated with radioiodine compared with thionamides or surgery (35). Problematic eye disease is more likely in those with pre-existing ophthalmopathy, in smokers (smoking is an independent risk factor for development of ophthalmopathy in Graves’ disease), and those with severe biochemical disease. In view of evidence (35) that a course of glucocorticoid abolishes any increase in risk of ophthalmopathy in those receiving radioiodine, many experts prescribe a short course of prednisone/prednisolone at the time of therapy. Typical doses of prednisone are 0.4-0.5 mg/kg/day starting 1-3 days following I-131 therapy and continued for one month, with gradual tapering over the next two months. However, recent data suggest that a lower dose of prednisone of 0.2 mg/kg/day for 6 weeks may be equally efficacious (54).

 

In those with severe clinical and biochemical thyrotoxicosis it is desirable to restore euthyroidism before proceeding to radioiodine therapy. This is because of the theoretical risk of inducing "thyroid storm" due to thyroid destruction and release of pre-formed thyroid hormones following radioiodine administration, together with the need to stop thionamide therapy temporarily at the time of treatment. In mild cases (judged both clinically and biochemically), such pre-treatment with thionamides may be unnecessary and radioiodine may be given as initial therapy or after short-term preparation with beta-adrenergic blockers.

 RADIOIOIDINE DOSING

 

Many studies have attempted to define optimal radioiodine doses in the hope of inducing euthyroidism and avoiding iatrogenic hypothyroidism in hyperthyroid patients (35). Studies have examined attempts to titrate doses of radioiodine according to factors such as thyroid size (judged clinically or by imaging), isotope uptake, or isotope turnover in the thyroid. Older literature suggested that cases of toxic nodular hyperthyroidism require larger doses of radioiodine to induce euthyroidism than cases of Graves' disease. It is clear, however, that measures of thyroid size or isotope uptake/turnover generally do not allow effective "dose titration". Furthermore, the dose of radioiodine required to cure toxic nodular hyperthyroidism is not different from that required in Graves' disease in the majority of cases (55). In some subjects with large goiter, higher initial doses or multiple treatments are required.

 

Many large thyroid centers thus avoid attempts at radioiodine "dose titration" and administer empirical doses. Such an approach avoids the necessity for extra hospital visits to document isotope uptake into the gland or the need for other imaging. The dose of radioiodine administered varies between centers, and is determined in part by radiation protection restrictions that vary considerably around the world. Typically, a dose of radioiodine is chosen which can be administered in the outpatient setting and which results in cure of thyrotoxicosis in the majority after a single dose, while not inducing hypothyroidism in all. In iodine-replete parts of the world such as the US and UK, a standard dose of radioiodine is 10-15 mCi or 400-600 MBq. In a UK series (56) a dose of this size resulted in cure of thyrotoxicosis in more than two thirds, at a cost of early hypothyroidism in 50%. Some centers administer larger doses to those with large goiter or to men, in view of evidence of relative radioresistance in these groups. There is also evidence that use of thionamides, especially propylthiouracil, before and/or after radioiodine treatment also induces relative radioresistance and hence the need for repeat dosing or a larger initial dose (35). It has been suggested that large doses should be administered routinely to elderly subjects, particularly those with cardiovascular disease or complications, to be certain of rapid restoration of euthyroidism. This view is reinforced by evidence that effective cure as indicated by the development of hypothyroidism requiring thyroxine replacement therapy is associated with a lessening of vascular mortality (compared with those not rendered hypothyroid) (17) and more likely conversion to sinus rhythm in those with AF associated with hyperthyroidism (24).

 FOLLOW-UP AFTER RADIOIODINE THERAPY

 

Thionamide therapy should be withdrawn 3-7 days before radioiodine (to allow iodine uptake into the thyroid) and should be restarted after a similar period post-treatment if the elderly subject has severe disease, incomplete biochemical control, significant complications (e.g., atrial fibrillation), or has return of symptoms in the short period of thionamide withdrawal before radioiodine therapy. After therapy, clinical and biochemical assessment should be carried out every 4-6 weeks for the first few months so that thionamide doses may be adjusted (according to free T4) and hypothyroidism identified. A transient rise in serum TSH may be seen in the first few months after radioiodine and does not necessarily indicate permanent hypothyroidism, but more marked biochemical or symptomatic hypothyroidism usually indicates the need for life-long T4 therapy. Persistence of biochemical hyperthyroidism 6 months after radioiodine therapy usually indicates the need for re-dosing. Unless small empirical doses are administered, the vast majority of patients with either toxic nodular hyperthyroidism or Graves' disease are rendered euthyroid (off all treatment) or hypothyroid (on T4) with one, two or (uncommonly) three doses (56,57). Occasional cases of apparent resistance to radioiodine treatment are seen.

 

Long-term, patients treated with radioiodine require biochemical follow-up for detection of hypothyroidism. Such follow-up is essential since the incidence of hypothyroidism is significant even many years after radioiodine and eventually up to 90% of those treated in this way become hypothyroid (35). Hypothyroidism rates may be slightly lower in those with toxic nodular hyperthyroidism (56) because of relative sparing of normal thyroid tissue through concentration of isotope in "hot" autonomous nodules.

 Surgical Treatment of Thyrotoxicosis

 

Surgical treatment of thyrotoxicosis is a viable option in selected patients, and if experienced thyroid surgeons are available (35). However, there is a higher risk of complications of anesthetic and surgery in elderly subjects, which limits its utility in this population.

 

If surgery is contemplated, it is essential that clinical and biochemical euthyroidism are restored beforehand. This requires therapy with thionamides, ideally for 2-3 months prior to surgery, sometimes in conjunction with pre-operative preparation with beta-adrenergic blockers or Lugol's iodine (35). Thorough preparation is essential in order to avoid thyroid storm post-operatively, as well as other significant complications of thyroid hormone excess, especially cardiovascular complications.

 

There is on-going debate regarding the most appropriate surgical approach for treatment of thyrotoxicosis. Many large centers advocate total thyroidectomy for Graves' hyperthyroidism, since partial thyroidectomy is associated with significant rates of short - and long-term recurrence (35), while in expert hands surgical complication rates should be similar. Such complications include bleeding into the neck, hypoparathyroidism, and damage to recurrent laryngeal nerves. Hypothyroidism is inevitable after total thyroidectomy (the patient leaves the hospital on T4 therapy) but is also common after partial thyroidectomy. Life-long follow-up (as with cases treated with radioiodine) is essential for detection of hypothyroidism (and recurrence of hyperthyroidism) after partial thyroidectomy.

 

Cases of toxic nodular hyperthyroidism may be treated by thyroid lobectomy or excision of a single hot nodule. Such an approach has the theoretical advantage of avoidance of hypothyroidism, as well as improvement in cosmetic appearance in those with large goiter. It should be noted, however, that reduction in nodule/goiter size is also evident after radioiodine therapy, albeit after several months. Surgery may be considered appropriate if toxic nodular goiter is associated with obstructive symptoms or if there is concern about the presence of co-existent malignancy in the goiter/nodules.

 Treatment of Amiodarone-Induced Thyrotoxicosis (AIT)

 

This condition is difficult to treat and a cause of significant morbidity/mortality in patients with underlying cardiac disease (35,58).  AIT can be diagnosed many months after amiodarone has been discontinued, since it persists in the body for long periods of time.  AIT can be a life-threatening diagnosis, since it worsens arrhythmias and cardiac function in patients who already have compromised cardiovascular systems. 

 

There are two types of AIT. Type 1 AIT occurs in patients with pre-existing thyroid abnormalities such as nontoxic multinodular goiters or subclinical Graves’ Disease. This type is thought to be due to iodine overload, since amiodarone is 37% iodine by weight. Type 2 AIT is a destructive thyroiditis that causes thyrotoxicosis by the release of pre-formed thyroid hormone, which can be prolonged. Some experts report that these two types can be distinguished by measurement of serum interleukin-6 (raised in destructive thyroiditis) or by ultrasonographic definition of thyroid vascularity (35,58). These tests are not, however, routinely available, and it is increasingly recognized that these varieties may co-exist.

 

In general, thionamide therapy should be considered first line treatment of Type 1 AIT. High dose glucocorticoids are considered first-line therapy for Type 2 AIT, although they can have significant side effects in elderly patients. In practice, it can be difficult to distinguish Type 1 from Type 2 AIT, and in severe acute cases, both thionamides and prednisone are sometimes started simultaneously. Type 2 AIT responds more quickly to glucocorticoids than Type 1 AIT responds to methimazole, so a rapid response to therapy is an indirect indicator of Type 2 AIT.  Perchlorate may be a helpful adjunct therapy, although it is not commercially available in the U.S.

 

Withdrawal of amiodarone is often not possible because of the serious nature of underlying dysrhythmias leading to amiodarone treatment, although it should be carefully considered. In any case, the long half-life of the drug (around 50 days) determines that any effect of amiodarone withdrawal is slow. Because of the iodine content of the drug, radioiodine therapy is ineffective because the radioisotope is not taken up into the thyroid. Radioiodine treatment is typically not feasible until at least 6 months after amiodarone withdrawal. Several groups have described surgical treatment of AIT, with a recent report suggesting that patients treated with thyroidectomy had lower 5-year cardiovascular and 10-year all-cause mortality, compared to medically treated AIT patients (59). Restoration of euthyroidism with thionamides is preferable pre-operatively if possible.

 SUBCLINICAL HYPERTHYROIDISM

 

"Subclinical" hyperthyroidism is a biochemical diagnosis characterized by a low serum TSH with normal serum thyroid hormone concentrations. Many of the subjects included in the studies quoted at the beginning of this chapter had subclinical, rather than overt, hyperthyroidism, as subclinical hyperthyroidism is more common than overt disease. There is significant variation in the reported prevalence of subclinical hyperthyroidism in the elderly, with typically quoted prevalence of 0.8 – 2% (35). As with overt hyperthyroidism, prevalence rates are lower if one excludes subjects taking thyroid hormone preparations. The prevalence of endogenous subclinical hyperthyroidism in a population depends on age, gender and iodine intake (3,60,61).

 

The most common cause of suppression of TSH in the general population is exogenous thyroid hormone therapy, typically levothyroxine (LT4). Population surveys (62) have shown that approximately one quarter of those prescribed LT4 long-term display reduction in TSH suggestive of mild over-treatment; (this is deliberate in the relatively small number of patients with a history of thyroid cancer). Since LT4 is prescribed to many patients over 60 years old, this medication is a common cause of subclinical hyperthyroidism. In fact, a recent study showed that over 40% of patients over the age of 64 years treated with levothyroxine had low TSH levels, indicating overtreatment (38).

 

In patients not receiving exogenous thyroid hormone therapy, the differential diagnoses of a low or undetectable TSH includes nonthyroidal illness and medications (see above). Once these have been excluded, nodular goiter is the next most common cause of low serum TSH in this age group. In subjects with a nodular goiter, either detectable clinically or evident on isotope imaging, suppression of serum TSH represents the earliest biochemical marker of thyroid autonomy and onset of hyperthyroidism. Other causes of endogenous subclinical hyperthyroidism in the elderly include Graves’ Disease, subacute thyroiditis, and silent thyroiditis, as in younger patients, although these are less common.

 

The natural history of endogenous subclinical hyperthyroidism is variable, and depends on the underlying cause. Most patients have stable subclinical hyperthyroidism over years, but a sizable minority either progress to overt hyperthyroidism or normalize their thyroid function (35).  A low but detectable TSH probably has less pathophysiological significance than a completely suppressed TSH, in terms of clinical consequences as well as progression rates. In addition, endogenous subclinical hyperthyroidism, for example secondary to nodular goiter, is probably of greater significance than exogenous (due to levothyroxine therapy) since the former is associated with higher serum T3 concentrations.

 

There is little evidence to suggest that subclinical hyperthyroidism is associated with significant symptoms (63), but there is a growing body of evidence that low serum TSH is associated with adverse effects, particularly on heart, bone, and brain, and possibly increased all-cause and cardiovascular mortality.

 

An important study of the Framingham population of the US (64) first revealed a 3-fold increased incidence of atrial fibrillation in subjects aged over 60 with serum TSH of less than 0.1 mU/L, compared with those with normal serum TSH. The likelihood of developing atrial fibrillation was also increased, but less markedly, in those with low but detectable TSH. The group in this survey with low TSH was heterogeneous and included some subjects taking exogenous T4 therapy. Similar findings have been reported in larger population-based studies since this initial observation (35). 

 

Recent studies have also reported that subclinical hyperthyroidism is associated with increased mortality and cardiovascular events in subjects 65 years and older (19,21,65).  A meta-analysis of individual-level data from 52,674 participants pooled from 10 cohort studies concluded that subclinical hyperthyroidism confers a 24% increased risk of overall mortality and 29% increased risk of cardiovascular mortality (66). Some of these studies, including the meta-analysis, have also examined non-fatal cardiovascular events in subclinical hyperthyroidism, with similar increased risks (66-69). Data indicate that subclinical hyperthyroid subjects appear to be at particular risk for the development of heart failure (66,70,71), especially older subjects and those with lower TSH levels.

 

Adverse effects of subclinical hyperthyroidism on bone may occur. A recent meta-analysis of 6 prospective cohorts (5,458 subjects, median age 72 years, 5% with subclinical hyperthyroidism) reported that older subjects with subclinical hyperthyroidism had increased annual rates of bone loss at the femoral neck, especially if the TSH was less than 0.1 mU/L (72).  A second meta-analysis of 13 prospective cohorts (70,298 subjects, median age 64 years, median follow-up 12 years, 3% with subclinical hyperthyroidism) reported that subjects with subclinical hyperthyroidism had increased rates of hip fracture, clinical spine fracture, non-spine fracture, and any fracture (73).  Risks were greatest if the TSH was less than 0.1 mU/L.  There is also evidence for improvement in bone metabolism or BMD after treatment of endogenous subclinical hyperthyroidism (74). Finally, in hypothyroid subjects who were started on LT4 and followed for a mean of 7 years, the number of 6-month periods with low TSH levels increased the risk of hip and major osteoporotic fractures in post-menopausal women, but not in men (75).  This further illustrates the importance of avoiding overtreatment in hypothyroidism.

 

Mood and cognitive function have also been examined in older subjects with subclinical hyperthyroidism (76). A meta-analysis of 11 studies (16,805 subjects, mean age over 70 years, median follow-up 3.5 years) reported an increased risk of dementia in subclinical hyperthyroid subjects (77).  A more recent prospective cohort study (2,558 subjects ages 70-79 years, median follow-up 9 years) reported an increased risk of dementia if the TSH was suppressed, but not if the TSH was low but detectable (78).  Reports on associations between subclinical hyperthyroidism and rates of depression or anxiety have been variable, with some studies indicating no association in older subjects (35,79), while others report increased rates of depressive symptoms in subclinical hyperthyroidism (80).

 

Concerns about effects of mild thyroid hormone excess upon heart and bone, and more recently on cognitive function, have led to a trend towards treatment of this condition. In those taking exogenous thyroid hormone, management is relatively straightforward, namely reduction in prescribed dose and re-checking of serum TSH 6-8 weeks later. For those not taking T4, many experts administer either antithyroid drugs or radioiodine to those with persistent subclinical hyperthyroidism, especially in subjects with atrial fibrillation or other underlying cardiac disease. Prospective trials confirming benefit of such therapy have yet to be performed, but analysis of large datasets indicate that prolonged periods of undertreatment confer increased risks (19,21).  Based on this, consensus guidelines recommend that older subjects and those with AF or other vascular risk factors should be treated (35).

SCREENING FOR HYPERTHYROIDISM IN ELDERLY SUBJECTS

 

Several factors should be considered before a decision is made to institute either population or targeted screening for thyroid disorders in groups such as the elderly. Firstly, screening programs should be instituted only for those conditions in which the benefits of screening outweigh the costs. Whether benefits outweigh the costs depends on accurate quantification of these issues, then a judgment as to whether the costs of screening are justified. Although it is clear that hyperthyroidism is common, there are no data that demonstrate that identified subjects benefit from being diagnosed; it is not sufficient to demonstrate only that such subjects exist. Such benefits and costs should ideally be based upon the results of a randomized controlled trial in an appropriate sample of the relevant population. In considering costs, those incurred by those who do not themselves gain from the screening program should be considered. If, for example, the screening process uses a test such as serum TSH with occasional positives, then some patients may be exposed to investigations which are unnecessary, with accompanying risk and potential morbidity.

 

While overt and subclinical hyperthyroidism are common in older subjects, and while there is evidence for adverse consequences of these diagnoses, the evidence that treatment in a screened population improves morbidity/mortality, and that the risks of such treatment outweigh the costs, is currently inconclusive. There should, nonetheless, be a high index of suspicion for hyperthyroidism in this age group and a low threshold for biochemical testing, especially in those with a previous personal or family history of thyroid disease or those with conditions such as atrial fibrillation that may reflect hyperthyroidism. Care must also be taken to recognize the atypical presentations of hyperthyroidism that occur in this age group, including unexplained weight loss and psychiatric symptoms.

 REFERENCES

 

  1. Franklyn JA, Boelaert K. Thyrotoxicosis. Lancet. 2012;379:1155-66.
  2. Tunbridge WM, Evered DC, Hall R, Appleton D, Brewis M, Clark F et al. The spectrum of thyroid disease in a community: the Whickham survey. Clin.Endocrinol.(Oxf) 1977;7:481-93.
  3. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, Braverman LE. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab. 2002;87:489-99.
  4. Parle JV, Franklyn JA, Cross KW, Jones SC, Sheppard MC. Prevalence and follow-up of abnormal thyrotrophin (TSH) concentrations in the elderly in the United Kingdom. Clin.Endocrinol.(Oxf) 1991;34:77-83.
  5. Wilson S, Parle JV, Roberts LM, Roalfe AK, Hobbs FDR, Clark P, Sheppard MC, Gammage MD, Pattison HM, Franklyn JA. Prevalence of subclinical thyroid dysfunction in the elderly in England – the Birmingham Elderly Thyroid Study (BETS): a community based cross-sectional survey. J Clin Endocrinol Metab 2006;91:4809-4816
  6. de Jongh RT, Lips P, van Schoor NM, Rijs KJ, Deeg DJ, Comijs HC, Kramer MH, Vandenbroucke JP, Dekkers OM. Endogenous subclinical thyroid disorders, physical and cognitive function, depression, and mortality in older individuals. Eur J Endocrinol. 2011;165:545-54.
  7. Cappola AR, Fried LP, Arnold AM, Danese MD, Kuller LH, Burke GL, Tracy RP, Ladenson PW. Thyroid status, cardiovascular risk, and mortality in older adults. JAMA. 2006;295:1033-41.
  8. Boelaert K, Torlinska B, Holder RL, Franklyn JA. Older subjects with hyperthyroidism present with a paucity of symptoms and signs: a large cross-sectional study. J Clin Endocrinol Metab. 2010;95:2715-26.
  9. Goichot B, Caron P, Landron F, Bouee S.  Clinical presentation of hyperthyroidism in a large representative sample of outpatients in France: relationships with age, aetiology and hormonal parameters. Clin Endocrinol 2016;84(3):445-51.
  10. Nordyke RA, Gilbert FI, Jr., Harada AS. Graves' disease. Influence of age on clinical findings. Arch.Intern.Med. 1988;148:626-31.
  11. Trivalle C, Doucet J, Chassagne P, Landrin I, Kadri N, Menard JF, Bercoff E. Differences in the signs and symptoms of hyperthyroidism in older and younger patients. J Am Geriatr Soc. 1996;44:50-3.
  12. Mooradian AD. Asymptomatic hyperthyroidism in older adults: is it a distinct  clinical and laboratory entity? Drugs Aging. 2008;25:371-80.
  13. Ceresini G, Ceda GP, Lauretani F, Maggio M, Bandinelli S, Guralnik JM, Cappola AR, Usberti E, Morganti S, Valenti G, Ferrucci L. Mild thyroid hormone excess is associated with a decreased physical function in elderly men. Aging Male. 2011;14:213-9.
  14. Martin FI, Deam DR. Hyperthyroidism in elderly hospitalised patients. Clinical features and treatment outcomes. Med J Aust. 1996;164:200-3.
  15. Danzi S, Klein I.  Thyroid disease and the cardiovascular system.  Endocrinol Metabo Clin North Am 2014;43:517-28.
  16. Franklyn JA, Maisonneuve P, Sheppard MC, Betteridge J, Boyle P. Mortality after the treatment of hyperthyroidism with radioactive iodine. N.Engl.J.Med. 1998;338:712-8.
  17. Franklyn JA, Sheppard MC, Maisonneuve P. Mortality in subjects treated for hyperthyroidism – a prospective cohort study examining the influence of thyroid status. JAMA 2015;294:71-80.
  18.  Dekkers OM, Horváth-Puhó E, Cannegieter SC, Vandenbroucke J, Sørensen HT, Jorgensen JO. Acute cardiovascular events and all-cause mortality in patients with hyperthyroidism: population-based cohort study. Eur J Endocrinol. 2017;176(1):1-9.
  19. Lillevang-Johansen M, Abrahamsen B, Jørgensen HL, Brix TH, Hegedüs L. Excess mortality in treated and untreated hyperthyroidism is related to cumulative periods of low serum TSH. J Clin Endocrinol Metab. 2017;102(7):2301-2309.
  20. Giesecke P, Rosenqvist M, Frykman V, Friberg L, Wallin G, Höijer J, Lonn S, Törring O. Increased cardiovascular mortality and morbidity in patients treated for toxic nodular goiter compared to Graves disease and nontoxic goiter. Thyroid. 2017;27(7):878-885.
  21. Lillevang-Johansen M, Abrahamsen B, Jørgensen HL, Brix TH, Hegedüs L. Duration of hyperthyroidism and lack of sufficient treatment are associated with increased cardiovascular risk. Thyroid 2019;29:332–340.
  22. Okosieme OE, Tayler PN, Evans C, Thayer D, Chai A, Khan I, Draman MS, Tennant B, Geen J, Sayers A, French R, Lazarus JH, Premawardhana LD, Dayan CM. Primary therapy of Graves’ disease and cardiovascular morbidity and mortality: a linked-record cohort study. Lancet Diabetes Endocrinol 2019;7:278–287.
  23. Klein I,.Ojamaa K. Thyroid hormone and the cardiovascular system: from theory to practice. J.Clin.Endocrinol Metab 1994;78:1026-7.
  24. Osman F, Franklyn JA, Holder RL, Sheppard MC, Gammage MD. Cardiovascular manifestations of hyperthyroidism before and after antithyroid therapy; a matched case-control study. J Am Coll Cardiol 2007; 49:71-81
  25. Osman F, Gammage MD, Franklyn JA. Hyperthyroidism and cardiovascular morbidity and mortality. Thyroid 2002;12:483-7.
  26. Nakazawa HK, Sakurai K, Hamada N, Momotani N, Ito K. Management of atrial fibrillation in the post-thyrotoxic state. Am J.Med. 1982;72:903-6.
  27. Mosekilde L, Eriksen EF, Charles P. Effects of thyroid hormones on bone and mineral metabolism. Endocrinol Metab Clin.North Am 1990;19:35-63.
  28. Uzzan B, Campos J, Cucherat M, Nony P, Boissel JP, Perret GY. Effects on bone mass of long term treatment with thyroid hormones: a meta-analysis. J.Clin.Endocrinol.Metab 1996;81:4278-89.
  29. Franklyn JA, Betteridge J, Holder R, Sheppard MC. Effect of estrogen replacement therapy upon bone mineral density in thyroxine-treated postmenopausal women with a past history of thyrotoxicosis. Thyroid 1995;5:359-63.
  30. Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Ensrud KE et al. Risk factors for hip fracture in white women. Study of Osteoporotic Fractures Research Group. N.Engl.J.Med. 1995;332:767-73.
  31. Franklyn JA, Ramsden DB, Sheppard MC. The influence of age and sex on tests of thyroid function. Ann.Clin.Biochem. 1985;22 ( Pt 5):502-5.
  32. Surks MI, Boucai L. Age- and race-based serum thyrotropin reference limits. J Clin Endocrinol Metab. 2010;95:496-502.
  33. Franklyn JA, Black EG, Betteridge J, Sheppard MC. Comparison of second and third generation methods for measurement of serum thyrotropin in patients with overt hyperthyroidism, patients receiving thyroxine therapy, and those with nonthyroidal illness. J.Clin.Endocrinol.Metab 1994;78 :1368-71.
  34. Adler SM, Wartofsky L. The nonthyroidal illness syndrome. Endocrinol Metab Clin North Am. 2007;36:657-72.
  35. Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, Rivkees SA, Samuels M, Sosa JA, Stan MN, Walter MA.2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid. 2016;26:1343-1421.
  36. Hamburger JI. Evolution of toxicity in solitary nontoxic autonomously functioning thyroid nodules. J.Clin.Endocrinol Metab 1980; 50:1089-93.
  37. Samuels MH. Subacute, silent, and postpartum thyroiditis. Med Clin North Am. 2012;96:223-33.
  38. Somwaru LL, Arnold AM, Joshi N, Fried LP, Cappola AR. High frequency of and factors associated with thyroid hormone over-replacement and under-replacement in men and women aged 65 and over. J Clin Endocrinol Metab. 2009;94:1342-5.
  39. Mammen JS, McGready J, Oxman R, Chia CW, Ladenson PW, Simonsick EM. Thyroid Hormone Therapy and Risk of Thyrotoxicosis in Community-Resident Older Adults: Findings from the Baltimore Longitudinal Study of Aging. Thyroid. 2015;25(9):979-86.
  40. Taylor PN, Iqbal A, Minassian C, Sayers A, Draman MS, Greenwood R, Hamilton W, Okosieme O, Panicker V, Thomas SL, Dayan C. Falling threshold for treatment of borderline elevated thyrotropin levels-balancing benefits and risks: evidence from a large community-based study. JAMA Intern Med. 2014;174:32-9.
  41. Johansen ME, Marcinek JP, Doo Young Yun J. Thyroid Hormone Use in the United States, 1997-2016. J Am Board Fam Med. 2020;33:284-288.
  42. Goundan PN, Lee SL. Thyroid effects of amiodarone: clinical update. Curr Opin Endocrinol Diabetes Obes. 2020;27:329-334.
  43. Reinwein D, Benker G, Lazarus JH, Alexander WD. A prospective randomized trial of antithyroid drug dose in Graves' disease therapy. European Multicenter Study Group on Antithyroid Drug Treatment. J.Clin.Endocrinol Metab 1993;76:1516-21.
  44. Allannic H, Fauchet R, Orgiazzi J, Madec AM, Genetet B, Lorcy Y et al. Antithyroid drugs and Graves' disease: a prospective randomized evaluation of the efficacy of treatment duration. J.Clin.Endocrinol Metab 1990;70:675-9.
  45. Pecere A, Caputo M, Sarro A, Ucciero A, Zibetti A, Aimaretti G, Marzullo P, Barone-Adesi F 2020 Methimazole treatment and risk of acute pancreatitis: A population-based cohort study. J Clin Endocrinol Metab 105:dgaa544. PMID: 32813014.
  46. Azizi F, Amouzegar A, Tohidi M, Hedayati M, Khalili D, Cheraghi L, Mehrabi Y, Takyar M 2019 Increased remission rates after long-term methimazole therapy in patients with Graves’ disease. Thyroid. 2019;29:1192-1200.
  47. Azizi F, Abdi H, Cheraghi L, Amouzegar A. Treatment of subclinical hyperthyroidism in the elderly: Comparison of radioiodine and long-term methimazole treatment. Thyroid. 2021;31:545-551.
  48. Sjölin G, Holmberg M, Törring O, Byström K, Khamisi S, de Laval D, Abraham-Nordling M, Calissendorff J, Lantz M, Hallengren B, Filipsson Nyström H, Wallin G. The long-term outcomes of treatment for Graves’ hyperthyroidism. Thyroid. 2019;29:1545–1557.
  49. Azizi F, Takyar MA, Madreseh E, Amouzegar A. Treatment of toxic multinodular goiter: comparison of radioiodine and long-term methimazole treatment. Thyroid. 2019;29:625-630.
  50. Orsinelli DA. Current recommendations for the anticoagulation of patients with atrial fibrillation. Prog.Cardiovasc.Dis. 1996;39:1-20.
  51. Franklyn JA, Maisonneuve P, Sheppard M, Betteridge J, Boyle P. Cancer incidence and mortality after radioiodine treatment for hyperthyroidism: a population-based cohort study. Lancet 1999;353:2111-5.
  52. Kitahara CM, Preston DL, Sosa JA, Berrington de Gonzalez A. Association of Radioactive Iodine, Antithyroid Drug, and Surgical Treatments With Solid Cancer Mortality in Patients With Hyperthyroidism. JAMA Netw Open. 2020;3:e209660.
  53. Iakovou I, Giannoula E, Chatzipavlidou V, Sachpekidis C. Associating radioiodine therapy in hyperthyroidism with cancer mortality: robust or random results of a statistical analysis? Hell J Nucl Med. 2020;23:94-95.
  54. Stan MN, Garrity JA, Bahn RS.  The evaluation and treatment of graves ophthalmopathy. Med Clin North Am. 2012;96:311-28.
  55. Franklyn JA, Daykin J, Holder R, Sheppard MC. Radioiodine therapy compared in patients with toxic nodular or Graves' hyperthyroidism. QJM. 1995;88:175-80.
  56. Allahabadia A, Daykin J, Sheppard MC, Gough SC, Franklyn JA. Radioiodine treatment of hyperthyroidism-prognostic factors for outcome. J.Clin.Endocrinol.Metab 2001;86:3611-7.
  57. Roque C, Santos FS, Pilli T, Dalmazio G, Castagna MG, Pacini F. Long-term Effects of Radioiodine in Toxic Multinodular Goiter: Thyroid Volume, Function, and Autoimmunity. J Clin Endocrinol Metab. 2020;105:dgaa214.
  58. Bogazzi F, Bartalena L, Martino E. Approach to the patient with amiodarone-induced thyrotoxicosis. J Clin Endocrinol Metab. 2010;95:2529-35.
  59. Cappellani D, Papini P, Pingitore A, Tomisti L, Mantuano M, Di Certo AM, Manetti L, Marconcini G, Scattina I, Urbani C, Morganti R, Marcocci C, Materazzi G, Iervasi G, Martino E, Bartalena L, Bogazzi F. Comparison between total thyroidectomy and medical therapy for amiodarone-induced thyrotoxicosis. J Clin Endocrinol Metab 2020;105:242–251.
  60. Garmendia Madariaga A, Santos Palacios S, Guillén-Grima F, Galofré JC. The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab 2014;99:923-31.
  61. Schouten BJ, Brownlie BE, Frampton CM, Turner JG. Subclinical thyrotoxicosis in an outpatient population - predictors of outcome. Clin Endocrinol (Oxf) 2011;74:257-61.
  62. Canaris GJ, Manowitz NR, Mayor G, Ridgway EC. The Colorado thyroid disease prevalence study. Arch Intern Med. 2000;160:526-34.
  63. McCahon D, Haque  MS, Parle J, Hobbs FR, Roberts LM. Subclinical thyroid dysfunction symptoms in older adults: cross-sectional study in UK primary care.  Br J Gen Pract. 2020;70:e208-e214.
  64. Sawin CT, Geller A, Wolf PA, Belanger AJ, Baker E, Bacharach P et al. Low serum thyrotropin concentrations as a risk factor for atrial fibrillation in older persons. N.Engl.J.Med. 1994;331:1249-52.
  65. Grossman A, Weiss A, Koren-Morag N, Shimon I, Beloosesky Y, Meyerovitch J. Subclinical thyroid disease and mortality in the elderly: a retrospective cohort study. Am J Med. 2016;129:423-30.
  66. Collet TH, Gussekloo J, Bauer DC, den Elzen WP, Cappola AR, Balmer P, Iervasi G, Åsvold BO, Sgarbi JA, Völzke H, Gencer B, Maciel RM, Molinaro S, Bremner A, Luben RN, Maisonneuve P, Cornuz J, Newman AB, Khaw KT, Westendorp RG, Franklyn JA, Vittinghoff E, Walsh JP, Rodondi N; Thyroid Studies Collaboration Subclinical hyperthyroidism and the risk of coronary heart disease and mortality. Arch Intern Med. 2012;172:799-809.
  67. Vadiveloo T, Donnan PT, Cochrane L, Leese GP. The Thyroid Epidemiology, Audit, and Research Study (TEARS): morbidity in patients with endogenous subclinical hyperthyroidism. J Clin Endocrinol Metab. 2011;96:1344-51.
  68. Nanchen D, Gussekloo J, Westendorp RG, Stott DJ, Jukema JW, Trompet S, Ford I, Welsh P, Sattar N, Macfarlane PW, Mooijaart SP, Rodondi N, de Craen AJ; PROSPER Group. 2012 Subclinical thyroid dysfunction and the risk of heart failure in older persons at high cardiovascular risk. J Clin Endocrinol Metab. 2012;97:852-61.
  69. Yang LB, Jiang DQ, Qi WB, Zhang T, Feng YL, Gao L, Zhao J. Subclinical hyperthyroidism and the risk of cardiovascular events and all-cause mortality: an updated meta-analysis of cohort studies. Eur J Endocrinol. 2012;167:75-84.
  70. Selmer C, Olesen JB, Hansen ML, von Kappelgaard LM, Madsen JC, Hansen PR, Pedersen OD, Faber J, Torp-Pedersen C, Gislason GH. Subclinical and overt thyroid dysfunction and risk of all-cause mortality and cardiovascular events: a large population study. J Clin Endocrinol Metab. 2014;99:2372-82.
  71. Gencer B, Collet TH, Virgini V, Bauer DC, Gussekloo J, Cappola AR, Nanchen D, den Elzen WP, Balmer P, Luben RN, Iacoviello M, Triggiani V, Cornuz J, Newman AB, Khaw KT, Jukema JW, Westendorp RG, Vittinghoff E, Aujesky D, Rodondi N; Thyroid Studies Collaboration. Subclinical thyroid dysfunction and the risk of heart failure events: an individual participant data analysis from 6 prospective cohorts. Circulation. 2012;126:1040-9.
  72. Segna D, Bauer DC, Feller M, Schneider C, Fink HA, Aubert CE, Collet T-H, da Costa BR, Fischer K, Peeters RP, Cappola AR, Blum HR, van Dorland HA, Robbins J, Naylor K, Eastell R, Uitterlinden AG, Ramirez FR, Gogakos A, Gussekloo J, Williams GR, Schwartz A, Cauley JA, Aujesky DA, Bischoff-Ferrari HA, Rodondi N, Thyroid Studies Collaboration. Association between subclinical thyroid dysfunction and change in bone mineral density in prospective cohorts. J Intern Med. 2018;283:56-72.
  73. Blum MR, Bauer DC, Collet TH, Fink HA, Cappola AR, da Costa BR, Wirth CD, Peeters RP, Åsvold BO, den Elzen WP, Luben RN, Imaizumi M, Bremner AP, Gogakos A, Eastell R, Kearney PM, Strotmeyer ES, Wallace ER, Hoff M, Ceresini G, Rivadeneira F, Uitterlinden AG, Stott DJ, Westendorp RG, Khaw KT, Langhammer A, Ferrucci L, Gussekloo J, Williams GR, Walsh JP, Jüni P, Aujesky D, Rodondi N; Thyroid Studies Collaboration. Subclinical thyroid dysfunction and fracture risk: a meta-analysis. JAMA 2015;313:2055-65.
  74. Faber J, Jensen IW, Petersen L, Nygaard B , Hegedus L, Siersbaek-Nielsen K . Normalization of serum thyrotrophin by means of radioiodine treatment in subclinical hyperthyroidism: effect on bone loss in postmenopausal women. Clin.Endocrinol (Oxf) 1998; 48:285-90.
  75. Abrahamsen B, Jørgensen HL, Laulund AS, Nybo M, Bauer DC, Brix TH, Hegedüs L. The excess risk of major osteoporotic fractures in hypothyroidism is driven by cumulative hyperthyroid as opposed to hypothyroid time: an observational register-based time-resolved cohort analysis. J Bone Miner Res. 2015;30:898-905.
  76. Gan EH, Pearce SH. Clinical review: The thyroid in mind: cognitive function and low thyrotropin in older people. J Clin Endocrinol Metab. 2012;97:3438-49.
  77. Rieben C, Segna D, da Costa BR, Collet TH, Chaker L, Aubert CE, Baumgartner C, Almeida OP, Hogervorst E, Trompet S, Masaki K, Mooijaart SP, Gussekloo J, Peeters RP, Bauer DC, Aujesky D, Rodondi N. Subclinical Thyroid Dysfunction and the Risk of Cognitive Decline: a Meta-Analysis of Prospective Cohort Studies. J Clin Endocrinol Metab. 2016;101:4945-4954.
  78. Aubert CE, Bauer DC, da Costa BR, Feller M, Rieben C, Simonsick EM, Yaffe K, Rodondi N. The association between subclinical thyroid dysfunction and dementia: the Health, Aging and Body Composition (Health ABC) study. Clin Endocrinol (Oxf). 2017;87:617-626.
  79. Varella AC, Bensenor IM, Janovsky CCPS, Goulart AC, Birck MG, Santos IS, Brunoni AR, Lotufo PA Thyroid-stimulating hormone levels and incident depression: Results from the ELSA-Brasil study. Clin Endocrinol (Oxf). 2021;94:858-865.
  80. Blum MR, Wijsman LW, Virgini VS, Bauer DC, den Elzen WPJ, Jukema JW, Buckley BM, de Craen AJM, Kearney PM, Stott DS, Gussekloo J, Westendorp RGJ, Mooijaart SP, Rodondi N, PROSPER study group. Subclinical Thyroid Dysfunction and Depressive Symptoms among the Elderly: A Prospective Cohort Study. Neuroendocrinology. 2016;103:291-9.

 

 

 

 

Assessing Insulin Sensitivity and Resistance in Humans

ABSTRACT

 

In this chapter we discuss a representative variety of methods currently available for estimating insulin sensitivity/resistance. These range from complex, time consuming, labor-intensive, invasive procedures to simple tests involving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

INTRODUCTION

 

Insulin resistance plays a major pathophysiological role in type 2 diabetes and is tightly associated with major public health problems including obesity, hypertension, coronary artery disease, dyslipidemias, and a cluster of metabolic and cardiovascular abnormalities that define the metabolic syndrome (1, 2, 3).

 

 A global epidemic of obesity is driving the increased incidence and prevalence of type 2 diabetes and its cardiovascular complications (4). Insulin resistance is commonly associated with visceral adiposity, glucose intolerance, hypertension, dyslipidemia, hypercoagulable state, endothelial dysfunction, and/or elevated markers of inflammation. Therefore, the presence of these clinical abnormalities is usually characteristic of an insulin resistant state. In addition to clinical manifestations of the “Insulin Resistance Syndrome”, insulin resistance predisposes to accelerated cardiovascular disease (CVD). Therefore, it is of great importance to develop tools for quantifying insulin sensitivity/resistance in humans that may be used to appropriately investigate the epidemiology, pathophysiological mechanisms, outcomes of therapeutic interventions, and clinical course of patients with insulin resistance (5). In this chapter, we will discuss some currently used methods for assessing insulin sensitivity, their applications, merits, and limitations.

 

INSULIN SENSITIVITY AND RESISTANCE

 

Metabolic actions of insulin help to maintain glucose homeostasis and promote glucose utilization (6). Insulin increases glucose utilization in peripheral organs (e.g., skeletal muscle and adipose tissue) and suppresses hepatic glucose production (HGP) and adipose tissue lipolysis. In addition to these classical metabolic target tissues, insulin has many other important physiological targets. These include the brain, pancreatic β-cells, heart, and vascular endothelium that help to coordinate and couple metabolic and cardiovascular homeostasis under healthy conditions (6-9). Insulin has concentration-dependent saturable actions to increase whole-body glucose disposal. The maximal effect of insulin defines “insulin responsiveness” while the insulin concentration required for a half-maximal response defines “insulin sensitivity” (Fig. 1). Although, other actions of insulin on fat and amino-acid metabolism, cardiovascular, kidney, and brain function also exhibit a concentration-dependent response, the term “insulin sensitivity” typically refers to insulin’s metabolic actions to promote glucose disposal.

Figure 1. Schematic representation of concentration-response relationships between plasma insulin concentrations and insulin-mediated whole-body glucose disposal. Curve a: normal insulin sensitivity and responsiveness. Curve b: rightward shift in insulin concentration-response curve. This represents decreased insulin sensitivity (increased EC50) with normal insulin responsiveness. Curve c: Decreased insulin sensitivity (increased EC50) and reduced insulin responsiveness. Curve d: Leftward shift in the insulin concentration-response response curve. This represents increased insulin sensitivity (decreased EC50) with normal insulin responsiveness.

 

The concept of insulin resistance was proposed as early as 1936 to describe diabetic patients requiring high doses of insulin (10). Insulin resistance is typically defined as decreased sensitivity and/or responsiveness to insulin-mediated glucose disposal and/or inhibition of HGP and adipose tissue lipolysis. Rigorous evaluation of altered sensitivity and responsiveness therefore requires a comparison of insulin dose-response curves.

DIRECT MEASURES OF INSULIN SENSITIVITY

 

Hyperinsulinemic Euglycemic Glucose Clamp

 

PROCEDURE

 

The glucose clamp technique, originally developed by Andres and DeFronzo is widely accepted as the reference standard for directly determining metabolic insulin sensitivity in humans (11). After an overnight fast, insulin is infused intravenously at a constant rate that may range from 5 - 120 mU/m2/min (dose per body surface area per minute). This constant insulin infusion results in a new steady-state insulin level that is above the fasting level (hyperinsulinemic). As a consequence, glucose disposal in skeletal muscle and adipose tissue is increased while HGP is suppressed. Under these conditions, a bedside glucose analyzer is used to frequently monitor blood glucose levels at 5 – 10 min intervals while 20% dextrose is given intravenously at a variable rate in order to “clamp” blood glucose concentrations in the normal range (euglycemic). An infusion of potassium phosphate is also given to prevent hypokalemia resulting from hyperinsulinemia and increased glucose disposal. After several hours of constant insulin infusion, steady-state conditions are typically achieved for plasma insulin, blood glucose, and the glucose infusion rate (GIR). Assuming that the hyperinsulinemic state is sufficient to completely suppress hepatic glucose production, and since there is no net change in blood glucose concentrations under steady-state clamp conditions, the GIR must be equal to the glucose disposal rate (M) (Fig. 2). Thus, whole body glucose disposal at a given level of hyperinsulinemia can be directly determined. M is typically normalized to body weight or fat-free mass to generate an estimate of insulin sensitivity. Alternatively, an insulin sensitivity index derived from clamp data can be defined as SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fasting and steady-state plasma insulin concentrations) (12).

Figure 2. Schematic representation of the “steady state” dynamics of glucose and insulin during an euglycemic hyperinsulinemic glucose clamp.

 

The validity of glucose clamp measurements of insulin sensitivity depends on achieving steady-state conditions. “Steady-state” is often defined as a period greater than 30-min (at least 1 h after initiation of insulin infusion) during which the coefficient of variation for blood glucose, plasma insulin, and GIR are less than 5% (12, 13). It is possible to use stable isotope or radio-labeled glucose tracer under clamp conditions to estimate HGP so that appropriate corrections can be made to M in the event HGP is not completely suppressed (14, 15, 16,17). An alternative approach is to use an insulin infusion rate sufficiently high to completely suppress HGP according to the insulin sensitivity/resistance of the population to be studied. M is routinely obtained at only a single insulin infusion rate and therefore comparisons between M or SIClamp among different subjects is valid only if the same insulin infusion rate is used for all subjects. When glucose tracers are used during a clamp study, the tracer is infused at constant rate throughout the study. HGP estimated during the last 20 or 30 min of the clamp is a measure of insulin-mediated suppression of HGP, an estimate of hepatic insulin sensitivity. Similarly, lipolytic rates can be assessed at baseline and hyperinsulinemia during clamp by using isotopic tracers (e.g., palmitate). A single or multistep hyperinsulinemic euglycemic clamp can be used to measure adipose tissue insulin sensitivity. The linear relationship between log transformed rates of palmitate flux and plasma insulin concentrations provides an IC50 (pmol/L) for suppression of lipolysis (18).

 

ADVANTAGES AND LIMITATIONS

 

The principal advantage of the glucose clamp in humans is that it directly measures whole body glucose disposal at a given level of insulinemia under steady-state conditions. Conceptually, the approach is straightforward and there are a limited number of assumptions which are clearly defined. In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue (e.g., study population < 100) it is appropriate to use the reference standard glucose clamp technique. The main limitations of the clamp approach are that it is time-consuming, labor intensive, expensive, and requires an experienced operator to manage technical difficulties. Thus, for epidemiological studies, large clinical investigations, or routine clinical applications (e.g., following changes in insulin resistance after therapeutic intervention in individual patients) application of the glucose clamp is not feasible.

 

Insulin-Suppression Test (IST)

 

PROCEDURE

 

The insulin-suppression test, another method that directly measures metabolic insulin sensitivity/resistance, was introduced by Shen et. al. in 1970 and subsequently modified by Harano et. al. (19, 20). After an overnight fast, somatostatin (250 μg/h) or the somatostatin analogue octreotide (25 µg bolus, followed by 0.5 µg/min) (21) is intravenously infused to suppress endogenous secretion of insulin and glucagon. Simultaneously, insulin (25 mU/m2/min) and glucose (240 mg/m2/min) are infused into the same antecubital vein over 3 h. From the contralateral arm, blood samples for glucose and insulin determinations are taken every 30 min for 2.5 h and then at 10 min intervals from 150 - 180 min of the IST. The constant infusions of insulin and glucose determine steady-state plasma insulin (SSPI) and glucose (SSPG) concentrations. The steady-state period is assumed to be from 150 - 180 min after initiation of the IST. SSPI concentrations are generally (but not always) similar among subjects. Therefore, the SSPG concentration will be higher in insulin resistant subjects and lower in insulin sensitive subjects. That is, SSPG values are inversely related to insulin sensitivity. The IST provides a direct measure (SSPG) of the ability of exogenous insulin to mediate disposal of an intravenous glucose load under steady-state conditions where endogenous insulin secretion is suppressed.

 

ADVANTAGES AND LIMITATIONS

 

The SSPG is a highly reproducible direct measure of metabolic actions of insulin that is less labor-intensive and less technically demanding than the glucose clamp. Indeed, since there are no variable infusions with the IST, steady-state conditions are more easily achieved with the IST than with the glucose clamp. Estimates of insulin sensitivity determined by SSPG correlate well with reference standard glucose clamp estimates in normal subjects (r = 0.93) and in patients with type 2 diabetes mellitus (r = 0.91). (22, 23). Indeed, SSPG has positive predictive power for cardiovascular disease events and onset of type 2 diabetes (24, 25). In research settings where assessing insulin sensitivity/resistance is of primary interest and feasibility is not an issue, it is appropriate to use the IST. Moreover, the IST can be used for larger populations that may pose difficulties for application of the glucose clamp (26). Many of the limitations of the IST are similar to those described above for the glucose clamp (with the exception that the IST is less technically demanding). Thus, it is impractical to apply the IST in large epidemiological studies or in the clinical care setting. SSPG under ideal conditions determines primarily skeletal muscle insulin sensitivity and is not designed to reflect hepatic insulin sensitivity.

 

INDIRECT MEASURES OF INSULIN SENSITIVITY

 

Minimal Model Analysis of Frequently Sampled Intravenous Glucose Tolerance Test (FSIVGTT)

 

PROCEDURE

 

The minimal model, developed by Bergman, Cobelli and colleagues in 1979, provides an indirect measure of metabolic insulin sensitivity/resistance based on glucose and insulin data obtained during an FSIVGTT (27). After an overnight fast, an intravenous bolus of glucose (0.3 g/kg body weight) is infused over 2 min starting at time 0. Currently, a modified FSIVGTT is used where exogenous insulin (4 mU/kg/min) is also infused over 5 min beginning 20 min after the intravenous glucose bolus (28, 29,30). Some studies use tolbutamide instead of insulin in the modified FSIVGTT to stimulate endogenous insulin secretion (15, 29, 31, 32, 27). Blood samples are taken for plasma glucose and insulin measurements at -10, -1, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16, 20, 22, 23, 24, 25, 27, 30, 40, 50, 60, 70, 80, 90, 100, 120, 160, and 180 min. These data are then subjected to minimal model analysis using the computer program MINMOD to generate an index of insulin sensitivity (SI).

 

The minimal model is defined by two coupled differential equations with four model parameters (Fig. 3). The first equation describes plasma glucose dynamics in a single compartment. The second equation describes insulin dynamics in a “remote compartment”. The structure of the minimal model allows MINMOD to uniquely identify model parameters that determine a best fit to glucose disappearance during the modified FSIVGTT. SI is calculated from two of these model parameters and is defined as fractional glucose disappearance per insulin concentration unit. In addition to SI, other minimal model parameters may be used to estimate a “glucose effectiveness” index (SG). SG is defined as the ability of glucose per se to promote its own disposal and inhibit HGP in the absence of an incremental insulin effect (i.e., when insulin is at basal or constant concentrations).

 

Figure 3. Schematic, equations, and parameters for the minimal model of glucose metabolism. Differential equations describing glucose dynamics (G(t)) in a monocompartmental “glucose space” and insulin dynamics in a “remote compartment” (X(t)) are shown at the top. Glucose leaves or enters its space at a rate proportional to the difference between plasma glucose level, G(t) and the basal fasting level, Gb. In addition, glucose also disappears from its compartment at a rate proportional to insulin levels in the “remote” compartment (X(t)). In this model, t = time; G(t) = plasma glucose at time t; I(t) = plasma insulin concentration at time t; X(t) = insulin concentration in “remote” compartment at time t; Gb = basal plasma concentration; Ib = basal plasma insulin concentration; G(0) = G0 (assuming instantaneous mixing of the IV glucose load); p1, p2, p3, and G0 = unknown parameters in the model that are uniquely identifiable from FSIVGTT; glucose effectiveness, SG = p1; and insulin sensitivity, SI = p3/p2

 

Recently the minimal model has been used to assess free fatty acid (FFA) insulin sensitivity. Using a one compartment nonlinear model of FFA kinetics during FSIVGTT, showed that the FFA insulin sensitivity parameter correlated well with minimal model indices (33). Furthermore, this model also showed that glucose modulates disposal of FFAs.

 

ADVANTAGES AND LIMITATIONS

 

Minimal model analysis of the modified FSIVGTT is easier than the glucose clamp method because it is slightly less labor intensive, steady-state conditions are not required, and there are no intravenous infusions that require constant adjustment. Unlike the glucose clamp or IST, information about insulin sensitivity, glucose effectiveness, and β-cell function can be derived from a single dynamic test. The minimal model generates excellent predictions of glucose disappearance during the FSIVGTT. SI is a strong predictor of the development of diabetes in a prospective study of children of diabetic parents (34). Moreover, the insulin-modified FSIVGT may be used in relatively large-scale population studies (35). Therefore, in research settings where assessing insulin sensitivity along with glucose effectiveness and β-cell function is of interest, minimal model analysis of the insulin-modified FSIVGTT may be appropriate. The minimal model approach is simpler than direct methods for determining insulin sensitivity. Nevertheless, it still involves intravenous infusions with multiple blood sampling over a 3 h period that is nearly as labor intensive as the glucose clamp or IST. In addition, many limitations of minimal model analysis stem from the fact that the model oversimplifies the physiology of glucose homeostasis and is discussed in detail elsewhere (5).

 

Oral Glucose Tolerance Test (OGTT)

 

The oral glucose tolerance test (OGTT) is a simple test widely used in clinical practice to diagnose glucose intolerance and type 2 diabetes (36). After overnight fast, blood samples for determinations of glucose and insulin concentrations are taken at 0, 30, 60, and 120 min following a standard 75g oral glucose load. Oral glucose tolerance reflects the efficiency of the body to dispose of glucose after an oral glucose load or meal. The OGTT mimics the glucose and insulin dynamics of physiological conditions more closely than conditions of the glucose clamp, IST, or FSIVGTT. However, it is important to recognize that glucose tolerance and insulin sensitivity are not equivalent concepts. In addition to metabolic actions of insulin, insulin secretion, incretin effects, and other factors contribute importantly to glucose tolerance. Thus, the OGTT and meal tolerance tests provide useful information about glucose tolerance but not insulin sensitivity/resistance per se.

 

Intravenous and Oral Tracer Studies

 

The use of tracers for estimation of insulin sensitivity was first introduced in 1986 to overcome the shortcomings of FSIVGTT (37) The minimal model method does not allow segregation of glucose production from liver from exogenously administered glucose during calculations of insulin sensitivity and thus induces error in the insulin sensitivity calculations. Labeled intravenous glucose can be differentiated from endogenously produced glucose and thus use of labeled glucose during IVGTT provides more precise and accurate measurements (38,39) Similarly, labeled glucose has been used in oral glucose tolerance test and insulin sensitivity has been calculated by minimal model technique similar to FSIVGTT(40). There is a strong correlation of insulin sensitivity calculated from labeled oral minimal model with insulin sensitivity calculated from gold standard euglycemic hyperinsulinemic clamp, r=0.7, p<0.001 (41). There are dual tracer and triple tracer methods as well to estimate the hepatic/endogenous glucose production and discussion of these methods is beyond the scope of this review (42). Basal hepatic insulin resistance index can then be estimated as the product of HGP rate and the fasting plasma insulin concentration. Use of tracer definitely allows for improvement over the FSIVGTT. Use of labeled oral glucose allows for more precise measurements of insulin sensitivity and glucose disposal from a simple OGTT and this can be a useful tool in large studies. The triple tracer method is cumbersome and cannot be employed in large studies.

 

SIMPLE SURROGATE INDEXES FOR INSULIN SENSITIVITY/RESISTANCE

 

Surrogates Derived from Fasting Steady-state Conditions

 

PROCEDURE

 

After an overnight fast, a single blood sample is taken for determination of blood glucose and plasma insulin. In healthy humans, the fasting condition represents a basal steady-state where glucose is homeostatically maintained in the normal range such that insulin levels are not significantly changing and HGP is constant. That is, basal insulin secretion by pancreatic β cells determines a relatively constant level of insulinemia that will be lower or higher in accordance with insulin sensitivity/resistance such that HGP matches whole body glucose disposal under fasting conditions. Surrogate indexes based on fasting glucose and insulin concentrations reflect primarily hepatic insulin sensitivity/resistance. However, under most conditions, hepatic and skeletal muscle insulin sensitivity/resistance are proportional to each other. In the diabetic state with fasting hyperglycemia, fasting insulin levels are inappropriately low and insufficient to maintain euglycemia. Therefore, definitions of the more useful surrogate indexes take these considerations into account. Due to lack of a standardized insulin assay, it is not possible to use surrogate indexes to define universal cutoff points for insulin resistance.

 

ADVANTAGES AND LIMITATIONS

 

Simple surrogate indexes of insulin sensitivity/resistance are inexpensive quantitative tools that can be easily applied in almost every setting including epidemiological studies, large clinical trials, clinical research investigations, and clinical practice. If a direct measure of insulin sensitivity is not required, not feasible to obtain, or if insulin sensitivity is of secondary interest, it may be appropriate to use a surrogate index. The relative merits and limitations of individual surrogate indexes are discussed below.

 

The Homeostasis Model Assessment (HOMA)

 

HOMA, developed in 1985, is a model of interactions between glucose and insulin dynamics that is then used to predict fasting steady-state glucose and insulin concentrations for a wide range of possible combinations of insulin resistance and β-cell function (43). The model assumes a feedback loop between the liver and β-cell (43, 44, 45); glucose concentrations are regulated by insulin-dependent HGP while insulin levels depend on the pancreatic β-cell response to glucose concentrations. Thus, deficient β-cell function reflects a diminished response to glucose-stimulated insulin secretion. Likewise, insulin resistance is reflected by diminished suppressive effect of insulin on HGP. HOMA model describes this glucose-insulin homeostasis by a set of empirically derived non-linear equations. The model predicts fasting steady-state levels of plasma glucose and insulin for any given combination of pancreatic β-cell function and insulin sensitivity. Computer simulations have been used to generate a grid from which mathematical transformations of fasting glucose and insulin β-cell function (HOMA %B) from steady-state conditions. An important caveat for HOMA is that it imputes dynamic β-cell function (i.e., glucose-stimulated insulin secretion) from fasting steady-state data. In the absence of dynamic data, it is difficult, if not impossible, to determine the true dynamic function of β-cell insulin secretion.

 

In practical terms, most studies using HOMA employ an approximation described by a simple equation to determine a surrogate index of insulin resistance. This is defined by the product of the fasting glucose and fasting insulin divided by a constant. Thus, HOMA-IR = fasting insulin (μU/ml) × fasting glucose (mmol/l) / 22.5. The constant is a normalizing factor, the product of fasting plasma insulin of 5 µU/mL and plasma glucose of 4.5 mmol/L obtained from an “ideal” and “normal” individual. Therefore, for an individual with normal insulin sensitivity, HOMA-IR = 1. It is important to note that over wide ranges of insulin sensitivity/resistance Log (HOMA-IR), (which normalizes the skewed distribution of fasting insulin values) determines a much stronger linear correlation with glucose clamp estimates of insulin sensitivity (12). HOMA or Log (HOMA) is extensively used in large epidemiological studies, prospective clinical trials, and clinical research studies (45, 46, 47). In research settings where assessing insulin sensitivity/resistance is of secondary interest or feasibility issues preclude the use of direct measures by glucose clamp, it may be appropriate to use Log (HOMA-IR). However, as discussed below, other surrogate indexes have certain advantages over HOMA or Log (HOMA) in some circumstances.

 

Quantitative Insulin Sensitivity Check Index (QUICKI)

 

QUICKI is an empirically-derived mathematical transformation of fasting blood glucose and plasma insulin concentrations that provides a reliable, reproducible, and accurate index of insulin sensitivity with excellent positive predictive power (12, 48,13, 49, 50). Since fasting insulin levels have a non-normal skewed distribution, log transformation improves its linear correlation with SIclamp. However, as with 1/(fasting insulin) and the G/I ratio, this correlation is not maintained in diabetic subjects with fasting hyperglycemia and impaired β-cell function that is insufficient to maintain euglycemia. To accommodate these clinically important circumstances where fasting glucose is inappropriately high and insulin is inappropriately low, addition of log (fasting glucose) to log (fasting insulin) provides a reasonable correction such that the linear correlation with SIClamp is maintained in both diabetic and non-diabetic subjects. The reciprocal of this sum results in further transformation of the data generating an insulin sensitivity index that has a positive correlation with SIclamp. Thus, QUICKI = 1/Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose, mg/dl). Over a wide range of insulin sensitivity/resistance, QUICKI has a substantially better linear correlation with SIclamp (r ≈ 0.8 – 0.9) than SI derived from the minimal model or HOMA-IR (12, 48, 49). Log (HOMA) is roughly comparable to QUICKI in this regard. Multiple independent studies find excellent linear correlations between QUICKI and glucose clamp estimates (either GIR or SIClamp) in healthy subjects, obesity, diabetes, hypertension, and many other insulin-resistant states (49, 51, 52, 53, 54, 55, 56). QUICKI is among the most thoroughly evaluated and validated surrogate index for insulin sensitivity. As a simple, useful, inexpensive, and minimally invasive surrogate for glucose clamp-derived measures of insulin sensitivity, QUICKI is appropriate and effective for use in large epidemiological or clinical research studies, to follow changes after therapeutic interventions, and for use in studies where evaluation of insulin sensitivity is not of primary interest.

 

Adipose Tissue Insulin Resistance Index (Adipo-IR)

 

Adipo-IR is a measure similar to HOMA-IR in that it is obtained from a fasting level of FFA and insulin (product of FFA and insulin levels). Recent studies have shown that Adipo-IR correlates well with the gold standard measure of adipose tissue insulin sensitivity derived from one-step hyperinsulinemic-euglycemic clamp technique using a palmitate tracer (57). Age and physical fitness were however shown to affect the predictive values. Thus, Adipo-IR may be suitable for larger population studies, however the multistep pancreatic clamp technique is probably needed for mechanistic studies of adipose tissue insulin action.

 

Surrogates Derived from Dynamic Tests

 

PROCEDURE

 

Surrogate indexes of insulin sensitivity that use information derived from dynamic tests include OGTT, meal tolerance tests, and IVGTT. Procedures for these tests have been described in a previous section. Specific indexes including Matsuda index (58), Stumvoll index (59), Avignon index (60), oral glucose insulin sensitivity index (OGSI) (61), Gutt index (62), and Belfiore index (63) use particular sampling protocols during the OGTT or the meal. In addition, minimal model approaches have been used to model plasma glucose and insulin dynamics during an OGTT or a meal to determine insulin sensitivity/resistance (64). Glucose disposal of an oral glucose load or a meal is mediated by a complex dynamic process that includes gut absorption, glucose effectiveness, neurohormonal actions, incretin actions, insulin secretion, and metabolic actions of insulin that primarily determine the balance between peripheral glucose utilization and HGP. Surrogate indexes that depend on dynamic testing take into account both fasting steady-state and dynamic post-glucose load plasma glucose and insulin levels. After an oral glucose challenge, the HGP is maximally suppressed for approximately 60 min and remains suppressed at a constant level for the subsequent 60–120 min time period. Therefore, glucose uptake by peripheral tissues (e.g., muscle and adipose tissue) primarily determines the rate of decrease in plasma glucose concentration from its peak value to its nadir during an OGTT. Based on this observation, surrogate indices of hepatic and muscle insulin sensitivity/ resistance from an OGTT has been widely used (65). Recent studies comparing the OGTT-derived, tissue-specific surrogate indices hepatic insulin resistance index (HIRI) and muscle insulin sensitivity index (MISI) with clamp measurements showed that surrogate indices derived from an OGTT are accurate in predicting insulin sensitivity, but are not tissue-specific (66). Studies using oral tracers in OGTT, with measurement of insulin sensitivity from OGTT and then comparing these to clamp measurements, would be crucial to ascertain the validity these measures. Indeed, recent studies have shown that it is possible to measure hepatic insulin sensitivity in healthy volunteers and in prediabetes with the use of single tracer (67).

 

ADVANTAGES AND LIMITATIONS

 

Many surrogate measures derived from dynamic data correlate reasonably well with glucose clamp estimates of insulin sensitivity (58, 61,62). Estimates of insulin sensitivity derived from OGTT predict the development of type 2 diabetes in epidemiologic studies ( 50, 68, 65). The advantage of surrogates based on dynamic testing is that information about insulin secretion can be obtained at the same time as information about insulin action. However, if one is only interested in estimating insulin sensitivity/resistance, fasting surrogates may be preferable to dynamic surrogates because they are simpler to obtain. The oral route of glucose delivery is more physiological than intravenous glucose infusion. However, poor reproducibility of the OGTT and meal tolerance test due to variable glucose absorption, splanchnic glucose uptake, and additional incretin effects need to be considered. Thus, distinguishing direct metabolic actions of insulin following oral ingestion of glucose or a mixed meal is more problematic than after FSIVGTT. In addition, as with many other measures of insulin sensitivity, surrogates derived from dynamic testing generally incorporate both peripheral and hepatic insulin sensitivity. Although OGTT involves considerably less work than FSIVGTT, dynamic testing in general requires more effort and cost than fasting blood sampling.

 

ETHNIC DIFFERENCES

 

Hispanics, African Americans, and South Asians are highly prone to develop diabetes. A meta-analysis showed that non-diabetic Africans have lower insulin sensitivity and higher insulin response after an intravenous glucose load compared to Caucasians and East Asians  ( 69). In a study that compared euglycemic hyperinsulinemic clamp derived glucose disposal rates (GDR) with HOMA-IR, QUICKI, and OGTT-derived indices, fasting insulin levels and HOMA-IR did not correlate with GDR whereas Matsuda index derived from OGTT significantly correlated with GDR in African American men (70). Similarly, in another study in Afro-Caribbean adults, HOMA-IR did not correlate with insulin sensitivity calculated from FSIVGTT and M-value calculated from hyperinsulinemic euglycemic clamp (71). Likewise, IR predictive ability of QUICKI and HOMA-IR was limited in Asian-Indian men (72). Recent studies highlight that minimal model may underestimate insulin sensitivity between groups when acute insulin response (AIR) is higher in one group (73). African Americans have reduced insulin clearance and higher AIR than Whites, suggesting that the minimal model may underestimate insulin sensitivity in African Americans (73). These studies suggest that at least in some ethnic groups, QUICKI and HOMA-IR may only be useful as secondary outcome measurements in assessing insulin sensitivity/resistance and studies inferring lower insulin sensitivity in non-diabetic African Americans based on FSIVGTT and minimal modeling should be interpreted cautiously.

 

METABOLOMICS  

 

Metabolomics is an interrogation and quantification of small-molecule metabolites in body fluids and tissues. It aims at identifying and quantifying small molecules in the sample by either using mass spectrometry (MS) or nuclear magnetic resonance (NMR) spectroscopy. The details of the methodology and its application in diabetes research are beyond the scope of this chapter. In this chapter, we will focus on new markers of insulin resistance that have been discovered using this approach. Using a non-targeted approach, Gall et al. metabolically profiled fasting plasma samples from 399 non-diabetic, clinically healthy subjects (74). Insulin sensitivity was measured using euglycemic hyperinsulinemic clamps. Individuals in the bottom tertile of the cohort were designated as insulin-resistant. Among the 485 candidate biomarkers identified, plasma α-hydroxybutyrate levels were inversely related to insulin sensitivity and this association was independent of age, sex and BMI. Other metabolites such as linoleoyl-glycerophosphocholine (L-GPC), glycine, and creatine were also highly correlated with insulin sensitivity. Using 26 metabolites from this study, the group went on to develop a model called Quantose algorithm to predict insulin resistance. Fasting insulin, α-hydroxybutyrate, L-GPC and oleate levels were included in this model. Quantose IR as a fasting surrogate of insulin sensitivity was superior to other simple surrogate measures and was able to predict the progression from normal glucose tolerance to impaired glucose tolerance (75). Branched chain amino acids (BCAAs) were found to significantly increase in obese compared to lean subjects and a BCAA based index correlated with HOMA (76). The elevation of BCAA in subjects with impaired fasting glucose and diabetes has been confirmed in subsequent studies (77).

 

Lipoprotein insulin resistance score (LPIR) is a novel metabolomic biomarker based on nuclear magnetic resonance (NMR) quantification of lipoprotein levels and sizes. This index has been shown to predict future type 2 diabetes mellitus is some cohorts (78). LPIR is derived from the weighted score of six lipoproteins (VLDL, LDL, and HDL sizes and concentrations) that are more strongly related to IR than each of its individual subclasses (79). A risk score of between 0-100 is estimated, with a score of 100 denoting being most insulin resistant. These metabolomic studies are promising since they can measure hundreds of metabolites in a very small sample. However, the pricing, technology, and access, precludes its use clinically. Further studies using this approach are necessary in larger more heterogeneous cohorts to replicate and validate surrogate insulin resistance markers derived through metabolomics

 

Table 1. Methods for Assessing Insulin Sensitivity and Resistance in Humans

Method

Measure of Insulin sensitivity

Direct Measures

Hyperinsulinemic Euglycemic Glucose Clamp

Average glucose infusion rate (GIR) = glucose disposal rate (M). SIClamp = M/(G x ΔI), where M is normalized for G (steady-state blood glucose concentration) and ΔI (difference between fasting and steady-state plasma insulin concentrations)

Insulin-suppression Test (IST)

Steady-state plasma glucose (SSPG) concentrations during constant infusions of insulin and glucose with suppressed endogenous insulin secretion

   

Indirect Measures

Minimal Model Analysis of Frequently Sampled Intravenous Glucose Tolerance Test (FSIVGTT)

Minimal model uniquely identifies model parameters that determine a best fit to glucose disappearance during the modified FSIVGTT. SI : fractional glucose disappearance per insulin concentration unit; SG (glucose effectiveness): ability of glucose per se to promote its own disposal and inhibit HGP in the absence of an incremental insulin effect (i.e., when insulin is at basal levels).

Simple Surrogate Indexes

Surrogates Derived from Fasting Steady-state Conditions

The Homeostasis Model Assessment (HOMA)

HOMA-IR = [(Fasting Insulin (µU/mL)) X (Fasting Glucose (mmol/L))]/22.5

Quantitative Insulin Sensitivity Check Index (QUICKI)

QUICKI = 1/[Log (Fasting Insulin, µU/ml) + Log (Fasting Glucose, mg/dl)]

Surrogates Derived from Dynamic Tests (OGTT)

Matsuda Index

ISI(Matsuda) = 10000/√[(Gfasting (mg/dl) x Ifasting (µU/ml) x (Gmean x Imean)]

Gutt Index - ISI (0, 120) (mg.l2.mmol-1.mIU-1.min-1)

ISI (0, 120) = 75000 + (G0-G120)(mg/l) x 0.19 x BW / 120 x Gmean (0, 120min) (mmol/l) x Log [Imean (0, 120min)](mU/l)

Gmean, mean plasma glucose concentration during OGTT; Go, plasma glucose concentration during fasting; G120, plasma glucose concentration at 120 min; Gmean, mean plasma glucose concentration during OGTT; Imean, mean insulin concentration during OGTT; Io, plasma insulin concentration during fasting; I120, plasma insulin concentration at 120 min.

 

SUMMARY

 

In this chapter we have discussed a representative variety of methods currently available for estimating insulin sensitivity/resistance (but this is by no means an exhaustive review) (Table 1). These range from complex, time consuming, labor-intensive, invasive procedures to simple tests involving a single fasting blood sample. It is important to understand the physiological concepts informing each method so that relative merits and limitations of particular approaches are appropriately matched with proposed applications and data is interpreted correctly. The glucose clamp method is the reference standard for direct measurement of insulin sensitivity. Regarding simple surrogates, QUICKI and Log (HOMA) are among the best and most extensively validated. Dynamic tests are useful if information about both insulin secretion and insulin action are needed.

 

ACKNOWLEDGEMENTS

 

This work was supported by the Intramural Research Program, NIDDK, NIH

 

REFERENCES

 

  1. Reaven, G.M., The insulin resistance syndrome: definition and dietary approaches to treatment. Annu Rev Nutr, 2005. 25: p. 391-406.
  2. Petersen, K.F., et al., The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome. Proc Natl Acad Sci U S A, 2007. 104(31): p. 12587-94.
  3. DeFronzo, R.A. and E. Ferrannini, Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care, 1991. 14(3): p. 173-94.
  4. Poirier, P., et al., Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation, 2006. 113(6): p. 898-918.
  5. Muniyappa, R., et al., Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab, 2008. 294(1): p. E15-26.
  6. Accili, D., Lilly lecture 2003: the struggle for mastery in insulin action: from triumvirate to republic. Diabetes, 2004. 53(7): p. 1633-42.
  7. Prodi, E. and S. Obici, Minireview: the brain as a molecular target for diabetic therapy. Endocrinology, 2006. 147(6): p. 2664-9.
  8. Muniyappa, R., et al., Cardiovascular actions of insulin. Endocr Rev, 2007. 28(5): p. 463-91.
  9. Kahn, S.E., R.L. Hull, and K.M. Utzschneider, Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature, 2006. 444(7121): p. 840-6.
  10. Himsworth, H.P., Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types. 1936. Int J Epidemiol, 2013. 42(6): p. 1594-8.
  11. DeFronzo, R.A., J.D. Tobin, and R. Andres, Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol, 1979. 237(3): p. E214-23.
  12. Katz, A., et al., Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab, 2000. 85(7): p. 2402-10.
  13. Chen, H., G. Sullivan, and M.J. Quon, Assessing the predictive accuracy of QUICKI as a surrogate index for insulin sensitivity using a calibration model. Diabetes, 2005. 54(7): p. 1914-25.
  14. Rizza, R.A., L.J. Mandarino, and J.E. Gerich, Dose-response characteristics for effects of insulin on production and utilization of glucose in man. Am J Physiol, 1981. 240(6): p. E630-9.
  15. Finegood, D.T., R.N. Bergman, and M. Vranic, Estimation of endogenous glucose production during hyperinsulinemic-euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous glucose infusates. Diabetes, 1987. 36(8): p. 914-24.
  16. McMahon, M.M., et al., Underestimation of glucose turnover measured with 6-3H- and 6,6-2H- but not 6-14Cglucose during hyperinsulinemia in humans. Diabetes, 1989. 38(1): p. 97-107.
  17. Radziuk, J. and S. Pye, Quantitation of basal endogenous glucose production in Type II diabetes: importance of the volume of distribution. Diabetologia, 2002. 45(8): p. 1053-84.
  18. Sondergaard, E. and M.D. Jensen, Quantification of adipose tissue insulin sensitivity. J Investig Med, 2016. 64(5): p. 989-91.
  19. Shen, S.W., G.M. Reaven, and J.W. Farquhar, Comparison of impedance to insulin-mediated glucose uptake in normal subjects and in subjects with latent diabetes. J Clin Invest, 1970. 49(12): p. 2151-60.
  20. Harano, Y., et al., Glucose, insulin, and somatostatin infusion for the determination of insulin sensitivity in vivo. Metabolism, 1978. 27(9 Suppl 1): p. 1449-52.
  21. Pei, D., et al., Evaluation of octreotide to assess insulin-mediated glucose disposal by the insulin suppression test. Diabetologia, 1994. 37(8): p. 843-5.
  22. Greenfield, M.S., et al., Assessment of insulin resistance with the insulin suppression test and the euglycemic clamp. Diabetes, 1981. 30(5): p. 387-92.
  23. Mimura, A., et al., Insulin sensitivity test using a somatostatin analogue, octreotide (Sandostatin). Horm Metab Res, 1994. 26(4): p. 184-7.
  24. Yip, J., F.S. Facchini, and G.M. Reaven, Resistance to insulin-mediated glucose disposal as a predictor of cardiovascular disease. J Clin Endocrinol Metab, 1998. 83(8): p. 2773-6.
  25. Facchini, F.S., et al., Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab, 2001. 86(8): p. 3574-8.
  26. Yeni-Komshian, H., et al., Relationship between several surrogate estimates of insulin resistance and quantification of insulin-mediated glucose disposal in 490 healthy nondiabetic volunteers. Diabetes Care, 2000. 23(2): p. 171-5.
  27. Bergman, R.N., et al., Equivalence of the insulin sensitivity index in man derived by the minimal model method and the euglycemic glucose clamp. J Clin Invest, 1987. 79(3): p. 790-800.
  28. Finegood, D.T., I.M. Hramiak, and J. Dupre, A modified protocol for estimation of insulin sensitivity with the minimal model of glucose kinetics in patients with insulin-dependent diabetes. J Clin Endocrinol Metab, 1990. 70(6): p. 1538-49.
  29. Saad, M.F., et al., Differences between the tolbutamide-boosted and the insulin-modified minimal model protocols. Diabetes, 1997. 46(7): p. 1167-71.
  30. Quon, M.J., et al., Direct comparison of standard and insulin modified protocols for minimal model estimation of insulin sensitivity in normal subjects. Diabetes Res, 1994. 25(4): p. 139-49.
  31. Yang, Y.J., J.H. Youn, and R.N. Bergman, Modified protocols improve insulin sensitivity estimation using the minimal model. Am J Physiol, 1987. 253(6 Pt 1): p. E595-602.
  32. Beard, J.C., et al., The insulin sensitivity index in nondiabetic man. Correlation between clamp-derived and IVGTT-derived values. Diabetes, 1986. 35(3): p. 362-9.
  33. Stefanovski, D., et al., Insulin Action, Glucose Homeostasis and Free Fatty Acid Metabolism: Insights From a Novel Model. Front Endocrinol (Lausanne), 2021. 12: p. 625701.
  34. Martin, B.C., et al., Role of glucose and insulin resistance in development of type 2 diabetes mellitus: results of a 25-year follow-up study. Lancet, 1992. 340(8825): p. 925-9.
  35. Howard, G., et al., Insulin sensitivity and atherosclerosis. The Insulin Resistance Atherosclerosis Study (IRAS) Investigators. Circulation, 1996. 93(10): p. 1809-17.
  36. American Diabetes, A., Diagnosis and classification of diabetes mellitus. Diabetes Care, 2007. 30 Suppl 1: p. S42-7.
  37. Cobelli, C., et al., Estimation of insulin sensitivity and glucose clearance from minimal model: new insights from labeled IVGTT. Am J Physiol, 1986. 250(5 Pt 1): p. E591-8.
  38. Toffolo, G. and C. Cobelli, The hot IVGTT two-compartment minimal model: an improved version. Am J Physiol Endocrinol Metab, 2003. 284(2): p. E317-21.
  39. Krudys, K.M., et al., Integrated model of hepatic and peripheral glucose regulation for estimation of endogenous glucose production during the hot IVGTT. Am J Physiol Endocrinol Metab, 2005. 288(5): p. E1038-46.
  40. Cobelli, C., et al., The oral minimal model method. Diabetes, 2014. 63(4): p. 1203-13.
  41. Dalla Man, C., et al., Insulin sensitivity by oral glucose minimal models: validation against clamp. Am J Physiol Endocrinol Metab, 2005. 289(6): p. E954-9.
  42. Dube, S., et al., Assessment of insulin action on carbohydrate metabolism: physiological and non-physiological methods. Diabet Med, 2013. 30(6): p. 664-70.
  43. Matthews, D.R., et al., Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia, 1985. 28(7): p. 412-9.
  44. Levy, J.C., D.R. Matthews, and M.P. Hermans, Correct homeostasis model assessment (HOMA) evaluation uses the computer program. Diabetes Care, 1998. 21(12): p. 2191-2.
  45. Wallace, T.M., J.C. Levy, and D.R. Matthews, Use and abuse of HOMA modeling. Diabetes Care, 2004. 27(6): p. 1487-95.
  46. Haffner, S.M., H. Miettinen, and M.P. Stern, The homeostasis model in the San Antonio Heart Study. Diabetes Care, 1997. 20(7): p. 1087-92.
  47. Dabelea, D., et al., Birth weight, type 2 diabetes, and insulin resistance in Pima Indian children and young adults. Diabetes Care, 1999. 22(6): p. 944-50.
  48. Chen, H., et al., QUICKI is a useful index of insulin sensitivity in subjects with hypertension. Am J Physiol Endocrinol Metab, 2003. 284(4): p. E804-12.
  49. Mather, K.J., et al., Repeatability characteristics of simple indices of insulin resistance: implications for research applications. J Clin Endocrinol Metab, 2001. 86(11): p. 5457-64.
  50. Hanley, A.J., et al., Prediction of type 2 diabetes using simple measures of insulin resistance: combined results from the San Antonio Heart Study, the Mexico City Diabetes Study, and the Insulin Resistance Atherosclerosis Study. Diabetes, 2003. 52(2): p. 463-9.
  51. Bastard, J.P., et al., Is quantitative insulin sensitivity check index, a fair insulin sensitivity index in humans? Diabetes Metab, 2001. 27(1): p. 69-70.
  52. Yokoyama, H., et al., Quantitative insulin sensitivity check index and the reciprocal index of homeostasis model assessment are useful indexes of insulin resistance in type 2 diabetic patients with wide range of fasting plasma glucose. J Clin Endocrinol Metab, 2004. 89(3): p. 1481-4.
  53. Skrha, J., et al., Comparison of the insulin action parameters from hyperinsulinemic clamps with homeostasis model assessment and QUICKI indexes in subjects with different endocrine disorders. J Clin Endocrinol Metab, 2004. 89(1): p. 135-41.
  54. Rabasa-Lhoret, R., et al., Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states. J Clin Endocrinol Metab, 2003. 88(10): p. 4917-23.
  55. Katsuki, A., et al., QUICKI is useful for following improvements in insulin sensitivity after therapy in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab, 2002. 87(6): p. 2906-8.
  56. Uwaifo, G.I., et al., Indices of insulin action, disposal, and secretion derived from fasting samples and clamps in normal glucose-tolerant black and white children. Diabetes Care, 2002. 25(11): p. 2081-7.
  57. Sondergaard, E., et al., How to Measure Adipose Tissue Insulin Sensitivity. J Clin Endocrinol Metab, 2017. 102(4): p. 1193-1199.
  58. Matsuda, M. and R.A. DeFronzo, Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care, 1999. 22(9): p. 1462-70.
  59. Stumvoll, M., et al., Use of the oral glucose tolerance test to assess insulin release and insulin sensitivity. Diabetes Care, 2000. 23(3): p. 295-301.
  60. Avignon, A., et al., Assessment of insulin sensitivity from plasma insulin and glucose in the fasting or post oral glucose-load state. Int J Obes Relat Metab Disord, 1999. 23(5): p. 512-7.
  61. Mari, A., et al., A model-based method for assessing insulin sensitivity from the oral glucose tolerance test. Diabetes Care, 2001. 24(3): p. 539-48.
  62. Gutt, M., et al., Validation of the insulin sensitivity index (ISI(0,120)): comparison with other measures. Diabetes Res Clin Pract, 2000. 47(3): p. 177-84.
  63. Belfiore, F., S. Iannello, and G. Volpicelli, Insulin sensitivity indices calculated from basal and OGTT-induced insulin, glucose, and FFA levels. Mol Genet Metab, 1998. 63(2): p. 134-41.
  64. Cobelli, C., et al., Assessment of beta-cell function in humans, simultaneously with insulin sensitivity and hepatic extraction, from intravenous and oral glucose tests. Am J Physiol Endocrinol Metab, 2007. 293(1): p. E1-E15.
  65. Abdul-Ghani, M.A., et al., What is the best predictor of future type 2 diabetes? Diabetes Care, 2007. 30(6): p. 1544-8.
  66. Muniyappa, R., et al., Predictive Accuracy of Surrogate Indices for Hepatic and Skeletal Muscle Insulin Sensitivity. J Endocr Soc, 2019. 3(1): p. 108-118.
  67. Visentin, R., et al., Hepatic insulin sensitivity in healthy and prediabetic subjects: from a dual- to a single-tracer oral minimal model. Am J Physiol Endocrinol Metab, 2015. 309(2): p. E161-7.
  68. Hanson, R.L., et al., Evaluation of simple indices of insulin sensitivity and insulin secretion for use in epidemiologic studies. Am J Epidemiol, 2000. 151(2): p. 190-8.
  69. Kodama, K., et al., Ethnic differences in the relationship between insulin sensitivity and insulin response: a systematic review and meta-analysis. Diabetes Care, 2013. 36(6): p. 1789-96.
  70. Pisprasert, V., et al., Limitations in the use of indices using glucose and insulin levels to predict insulin sensitivity: impact of race and gender and superiority of the indices derived from oral glucose tolerance test in African Americans. Diabetes Care, 2013. 36(4): p. 845-53.
  71. Thompson, D.S., et al., Limitations of fasting indices in the measurement of insulin sensitivity in Afro-Caribbean adults. BMC Res Notes, 2014. 7: p. 98.
  72. Muniyappa, R., et al., Limited predictive ability of surrogate indices of insulin sensitivity/resistance in Asian-Indian men. Am J Physiol Endocrinol Metab, 2010. 299(6): p. E1106-12.
  73. Ha, J., et al., When MINMOD Artifactually Interprets Strong Insulin Secretion as Weak Insulin Action. Front Physiol, 2021. 12: p. 601894.
  74. Gall, W.E., et al., alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One, 2010. 5(5): p. e10883.
  75. Cobb, J., et al., A novel fasting blood test for insulin resistance and prediabetes. J Diabetes Sci Technol, 2013. 7(1): p. 100-10.
  76. Newgard, C.B., et al., A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab, 2009. 9(4): p. 311-26.
  77. Menni, C., et al., Biomarkers for type 2 diabetes and impaired fasting glucose using a nontargeted metabolomics approach. Diabetes, 2013. 62(12): p. 4270-6.
  78. Mackey, R.H., et al., Lipoprotein particles and incident type 2 diabetes in the multi-ethnic study of atherosclerosis. Diabetes Care, 2015. 38(4): p. 628-36.
  79. Shalaurova, I., et al., Lipoprotein insulin resistance index: a lipoprotein particle-derived measure of insulin resistance. Metab Syndr Relat Disord, 2014. 12(8): p. 422-9.

 

Anaerobic Infections and Endocrinology

ABSTRACT

 

Anaerobic bacteria are present as part of the normal microbial flora in the human body. These bacteria turn virulent whenever the host defense mechanisms are compromised. Diabetes and glucocorticoid abuse are the two common endocrine conditions that predisposes individuals to anaerobic infections. Anaerobic infections are common in tropical countries and can affect any tissue or gland resulting in severe organ dysfunction. Microbial endocrinology deals with the bidirectional interaction between the hormones and the microbes. The interaction is influenced by the virulence factors released from the microbes, inflammatory mediators, and the hormonal dysfunction. In this chapter, we shall discuss the various anaerobic bacterial infections relevant in endocrinology practices.

 

INTRODUCTION

 

The term “anaerobic” broadly denotes intolerance to oxygen. Anaerobic bacteria are the commonest bacteria in the bacterial flora present on the skin and mucous membranes (1). They are broadly divided into three types based on their relation to oxygen and growth potential as shown in figure 1.

Figure 1. Types of anaerobic bacteria

 

Virtually all anaerobic infections are derived from the normal bacterial flora of the body. The virulence characteristics of the organisms are kept in check by the defense mechanisms and a breach in the same may lead to infection. The risk of anaerobic infection is determined by the balance between the inoculum, virulence characteristics, and the host defenses. Previously, anaerobic infections were considered to be less prevalent due to the lack of identification techniques and the fastidious nature of the bacteria (2). Increased awareness, antibiotic misuse resulting in changing microbiome, ease of culture and diagnostic techniques helped in demonstrating that anaerobic infections also are frequent in clinical practice.

 

Microbial endocrinology is a term coined in 1992, to describe the bi-directional interplay between microbes and endocrine hormones (3). Endocrine glands are located deep in the human body with the exception of the thyroid gland. Most of the endocrine glands have a thick capsule protecting them from the contagious spread of infection. The endocrine glandular tissue is highly vascular, thereby not conducive for the growth of anaerobic bacteria. However, anaerobes can overcome the host defenses resulting in infection and breaks in the anatomic barrier can occur due to surgery, trauma, or the disease process itself from within. The predisposing factors for anaerobic infections include diabetes, immunosuppression, malignancy, neutropenia, and decreased redox potential in the tissues.

 

INTERPLAY BETWEEN ANAEROBIC BACTERIA AND HORMONES

 

The taxonomy of anaerobes has changed recently due to the improvement in diagnostic techniques. The development of advanced culture methods, next generation sequencing technology, and metagenomics has improved the understanding of anaerobic bacteria (4). Previously, the antibiotic susceptibility pattern of most of the anaerobes was not clear due to the difficulties in culture methods. Advanced diagnostic techniques like DNA hybridization, mass spectrometry, multiplex PCR, and oligonucleotide array technologies helped in improving the classification as well as the understanding of antibiotic susceptibility patterns of these bacteria. A simple taxonomical classification of anaerobic bacteria useful in clinical practice is shown in figure 2.

Figure 2. Types of anaerobic bacteria

 

Estrogen and Vaginal Flora

 

The healthy vaginal flora consists of Lactobacillus species and estrogen plays an important role in maintaining this flora (5). Estrogen increases vaginal epithelial activity resulting in a thickened layer of epithelium with glycogen deposition. The Lactobacilli breaks the glycogen into lactic acid and hydrogen peroxide locally, resulting in the vaginal pH being maintained in acidic range (< 4.5) to prevent the growth of anaerobic bacteria. Bacterial vaginosis is a common infection in women due to a shift of the vaginal microbiome from Lactobacillus flora to a mixture of facultative and obligatory anaerobic bacteria. The typical microorganisms include Gardnerella vaginalis, Mycoplasma hominis, and Atopobium vaginae. Postmenopausal females have a higher risk of bacterial vaginosis due to the precipitous decline in the concentration of estradiol. Evidence shows that topical estrogen therapy in these women normalize the vaginal flora and reduce the risk of anaerobic infections (6).

 

Adrenal Hormone and Anaerobes

 

Exposure to any form of stress elevates sympathetic nervous system activity and releases adrenaline and noradrenaline from the adrenal medulla. Prolonged stress induces a shift in immunity from Th1 linked cellular immunity to Th2 linked humoral immunity. In addition to many host tissues, microbes also respond to the catecholamines and increase their virulent characteristics (7). The hormonal communication between bacteria and humans involves the presence of interkingdom signaling receptors. Bacterial cell membrane bound histidine kinases (QseC and QseE) act as adrenergic sensors to detect the local hormone concentrations. QseC also modulate the expression of many genes that increase the virulence and inflammation. This is one of the mechanisms that interlink the immune-endocrine interactive pathway mediated by stress hormones.

 

Stress induced alterations in the anaerobes of the gingival flora led to the observation that noradrenaline and adrenaline act as environmental cues for bacteria (8). The spectrum of biological effects of the stress hormones on gingival flora could range from halitosis to atherosclerotic plaque rupture leading to acute coronary syndrome. These hormones affect the growth of Fusobacterium, Propionibacterium, and Prevotella and the hormonal effects are mostly species or strain specific. The biological adverse effects are mediated by changes in biofilms, bacterial adaptation techniques, bacterial adherence, and release of the cytotoxic enzymes.

 

DIABETES AND ANAEROBIC INFECTIONS

 

Diabetes mellitus (DM) is the most common metabolic and endocrine disorder that predisposes an individual to the development of infections. The defective immune responses seen in patients with DM could exacerbate the risk of anaerobic infections. Though many superficial and deep infections are common in patients with DM, few amongst them are unique in their description. The unique anaerobic infections seen in patients with DM include emphysematous cholecystitis and emphysematous pyelonephritis. Malignant otitis externa is also unique to DM but is mostly polymicrobial in origin.

 

Diabetic Foot Disease

 

Diabetic foot disease is the commonest cause of lower limb amputation in clinical practice. The lifetime risk for a diabetic foot disease is about 25% in certain patients with diabetes. The infections are usually polymicrobial in nature and lead to considerable morbidity and occasional mortality. Anaerobic infections are more common in wounds that are deep seated and are often resistant to the antibiotics and conservative measures (9). Peptostreptococcus and Bacteroides species are the two common anaerobic bacteria of the diabetic foot. Anaerobic bacteria could be either primary or secondary colonizers in the etiology of diabetic foot ulcers. The ischemic and necrotic wounds have a higher rate of anaerobic infection due to the associated low blood supply and low redox potential that facilitate the growth of these bacteria. There is an ethnic variation in the bacterial etiology of diabetic foot infections. Anaerobic osteomyelitis is typically seen associated with diabetic foot ulcers and presents with a chronic non-healing ulcer of the leg. Early surgical debridement, antibiotic therapy with a spectrum against anaerobes, foot revascularization along with proper foot care are the guiding principles in the management of diabetic foot disease. 

 

Fournier’s Gangrene

 

Fournier’s gangrene (FG), first described in 1883, is a rare necrotizing infection of the perineal and genital skin due to both aerobic and anaerobic organisms (10). There is a male preponderance and the disease is mostly described in middle age and elderly patients. The predisposing factors for FG include diabetes mellitus, immunosuppression, and alcoholism. Recently SGLT2 inhibitors have been linked with an increased risk of FG. The condition leads to microthrombi of the small subcutaneous vessels leading to local necrosis and gangrene which is a fertile nidus for anaerobic bacteria to spread rapidly in the subcutaneous tissues. Initially, the patient presents with cellulitis of the scrotal skin and progression of symptoms may lead to severe sepsis and death. The reported mortality rates with FG are about 25 – 30% and the management includes extensive surgical debridement along with broad spectrum antibiotics and hemodynamic supportive measures.

 

Necrotizing Fasciitis

 

Necrotizing fasciitis (NF) is a life-threatening soft tissue infection that causes local tissue destruction, necrosis, and severe sepsis (11). FG is also a form of NF restricted to the genital area. NF is divided into four types based on the etiological organisms. Type 1 NF is polymicrobial in origin including anaerobes, whereas, type 2 NF is due to either Streptococcus or Staphylococcus. Type 3 and 4 are less common and are due to Vibrio species and fungi respectively. The predisposing factors include DM, malignancy, immunosuppression, alcohol abuse, and systemic chronic debilitating disease. Initial presentation mimics that of cellulitis and early clues to the NF are pain and systemic features out of proportion to the local swelling and the presence of hemorrhagic bullae. Patients with diabetes and NF tend to have polymicrobial infections, severe renal impairment, delayed diagnosis. and multiple co-morbid ailments in comparison to NF patients without diabetes (12). Management principles are similar to FG and include surgical debridement, broad spectrum antibiotics, and supportive measures.

 

Periodontitis

 

Infection of the tissues surrounding the teeth are known as periodontitis and is usually caused by the anaerobic gram-negative bacteria. This is more common in patients with type 2 DM and this complication is often known as the “Sixth” complication of diabetes. The links between diabetes and periodontitis are mediated by oxidative stress, advanced glycation end products leading to immune dysfunction, inflammatory marker release, and increased tissue destruction (13). Periodontitis also exacerbates insulin resistance due to the release of cytokines and chemokines. DM is characterized by periapical bone destruction, poor wound healing, and also has a direct effect on the dental pulp integrity. Periodontitis is an independent marker of mortality in patients with T2DM and it is essential to treat these two conditions simultaneously for better outcomes.

 

ORGAN SPECIFIC ANAEROBIC INFECTIONS

 

Endocrine glands are usually resistant to localized infections due to their location, high vascularity, and in some glands the presence of a protective capsule preventing the local spread of infection. However, these natural barriers are broken in certain conditions leading to the development of infections.

 

Thyroid Gland

 

The thyroid gland is resistant to bacterial infection due to the high iodine content, blood supply, and thick capsule. Acute suppurative thyroiditis (AST) is a complication due to the anaerobic bacterial infection of the thyroid gland (14). Porphyromonas, Propionibacterium and Streptococcus are the common bacteria that have been reported to lead to AST. Many of these bacteria live as commensals in the gingival epithelium. These patients usually present with a tender neck mass and systemic features of inflammation, similar to the presentation of subacute thyroiditis (SAT). It is essential to differentiate between AST and SAT, as glucocorticoids worsen the former and are indicated in the later condition. The majority of the AST patients are euthyroid, whereas, the SAT presents with features of thyrotoxicosis. AST is seen involving the left side of thyroid gland, whereas, SAT involves both sides similarly. Ultrasonography and aspiration cytology aid in the confirmation of the diagnosis. Therapy consists of appropriate antimicrobial drugs and surgical drainage of an abscess if present.

 

Pituitary Gland

 

The intrasellar location and the high rate of blood flow per gram makes the pituitary gland resistant to the development of local infections. However, a few case reports have described anaerobic abscesses in the sella that could be due to blood stream infection (15). The patients present with features of a pituitary mass including local compression and hormonal dysfunction. Surgical drainage of the abscess along with prolonged anti-anaerobic therapy is essential for recovery. There may be residual hormonal dysfunction in patients necessitating long-term hormonal replacement.

 

Adrenal Gland

 

Adrenal gland infections are very rare in clinical practice and are usually predisposed by the presence of a blood collection in the gland. The presenting features include fever with chills, abdominal pain, and occasionally features of adrenal deficiency. The infection is mostly due to the aerobic bacilli, but polymicrobial infections are not uncommon. Recent reports suggest the beneficial role of metagenomic next generation sequencing (mNGS) that helps in the early identification of the anaerobic infection (16). mNGS technology helps in identification of multiple anaerobic bacteria simultaneously and the results are available in less than 48 hr, unlike conventional culture which takes more than a week. Management is similar to any other organ involvement with pus drainage and prolonged antibiotics.

 

INFERTILITY AND ANAEROBIC INFECTIONS

 

Infertility affects about 10 – 15% of couples and infections constitute one of the major contributory factors for infertility (17). Female and male factors account for about 40% of etiologies exclusively, whereas, both partners along with an idiopathic etiology account for the remaining 20%. Anaerobic infections constitute one of the common infectious causes of infertility, albeit, predominantly in females.

 

Female Infertility

 

Pelvic inflammatory disease (PID) is the commonest cause leading to female infertility due to tubal adhesions, mucosal damage, and tubal occlusion. PID is caused by multiple organisms which include Chlamydia, Neisseria, and anaerobes. Bacterial vaginosis is a major contributory factor in the pathogenesis of PID as evidenced by the identification of the similar microbial flora (18). Bacteria ascend the genital tract via the endocervical and endometrial epithelia including the lymphatics. Lower abdominal pain and vaginal discharge are the two common symptoms of PID. Early identification of PID, prompt antibiotic therapy, and surgical drainage of the pus result in the cure without residual tubal complications. Patients with recurrent abortions have also been shown to have vaginal colonization with Gardnerella vaginalis and facultative anaerobes (18). This indicates an association between the altered vaginal microflora, local and systemic inflammation, change in the immune mediators, chemokines and cytokines, impaired implantation, placentation, and blood vessel transformation culminating into the recurrent abortions.

 

Male Infertility

 

Anaerobic infections affect semen quality and the total sperm concentration leading to male infertility. The semen samples from sub-fertile men are characterized by the presence of a large number of pus cells and multiple bacteria (19). Anaerobic bacteria affect the ability of the sperm to penetrate the cervical mucosa by the release of microbial toxins. Anaerobic infections are not routinely identified with the standard methods of culture and should be ruled out in all patients with unexplained oligoasthenospermia along with the presence of pus cells in the semen. Positive microbial cultures, however do not convey the exact location of the infection as the semen consists of secretions from the multiple glands including the prostate. A classic four specimen technique could be helpful in the localization of the infection and these patients require long term antibiotic therapy.

 

GUT ANAEROBES AND METABOLIC DISORDERS

 

Gut microbes are essential for the host immune system and help in digestion and maintenance of local tissue integrity. The intestinal bacteria mediate their beneficial effects by breaking dietary constituents into various short chain fatty acids which act as beneficial signals in metabolism and immunomodulation (21). Though it’s very difficult to characterize the entire gut microbiome, parameters such as alpha species diversity, ratio between the beneficial (Akkermansia, Bifidobacterium, Lactobacillus etc.) and the harmful (Enterococcus, Bacteroides, Lachnospiraceae etc.) bacteria are used in laboratory evaluation. Recent reports have emerged that the gut microbiome plays an important role in the etiopathogenesis of metabolic disorders including type 2 DM and obesity.

 

Diet and environmental factors play an important role in shaping the gut microbiome. The diversity in the gut microbiota could also be a contributory factor in the prevalence of the metabolic disorders between different ethnic populations (22). Increasing use of the antibiotics, environmental pollution, and consumption of refined products have led to alterations in the microbial flora with a shift from a healthy flora to an unhealthy one. Proinflammatory molecules secreted from intestinal bacteria translocate to the blood stream triggering metabolic endotoxemia, which is described as the leaky gut syndrome. The gut-blood barrier is often broken with the colonization of the anaerobic bacteria in the gut replacing the normal flora.  

 

The microflora in individuals is a key determinant in directing the response to antibiotics and probiotics. The fecal samples of Japanese patients with T2DM showed lower bacterial counts of obligatory anaerobes and higher content of facultative anaerobes in comparison to the control population. There is also a higher percentage of gut bacteria in the circulation, thereby confirming the leaky-gut hypothesis (23).  Apart from metabolic disorders, the gut dysbiosis has not been shown to affect other endocrine disorders.

 

ENDOCRINE ISSUES WITH THE ANTIMICROBIALS USED AGAINST ANAEROBES

 

Antimicrobials are the cornerstone of therapy against the anaerobic infections. In a few cases, the antibiotic therapy is supplemented with the surgical drainage of the pus. The therapy is often prolonged due to the slow growth rate of the anaerobes, polymicrobial nature of the infection, and the development of antibiotic resistance (24). The commonly used antimicrobials against anaerobic infections include metronidazole, carbapenems, quinolones, beta-lactams, chloramphenicol, tigecycline, and clindamycin. Many of these drugs have no significant endocrine side-effects except for the dysglycemia with the use of quinolones. Other endocrine effects due to the protracted use of these drugs are summarized in the table 1.

 

Table 1. Endocrine Side-Effects of Antimicrobials used Against Anaerobic Infection

Drug

Endocrine side-effects

Metronidazole

Altered gut microbiome

Anterior pituitary inhibition

Quinolones

Dysglycemia,

Reduced absorption of levothyroxine

Seizures in thyrotoxicosis patients

Beta-lactams

Fractures

Tigecycline

Hypoglycemia

Chloramphenicol

Inhibition of thyroid hormones production

Clindamycin & Carbapenems

Nil

 

CONCLUSION

 

Anaerobic infections are common in clinical practice and diabetes is the most common endocrine condition predisposing for these infections. Anaerobic organisms have hormonal interactions with gonadal and adrenal hormones and the field of microbial endocrinology is expanding rapidly. Organ specific anaerobic infections may lead to endocrine dysfunction in the form of infertility, glandular abscess, and hypofunction of the involved endocrine axis. A high index of clinical suspicion is essential to identify anaerobic infections especially in the tropical countries. The principles of management are prolonged antibiotic therapy along with drainage of the pus. Systemic supportive therapy and extensive debridement is essential in life threatening anaerobic infections like necrotizing fasciitis.

 

REFERENCES

 

  1. Brook I. Spectrum and treatment of anaerobic infections. J Infect Chemother. 2016 Jan;22(1):1-13.
  2. Vena A, Muñoz P, Alcalá L, Fernandez-Cruz A, Sanchez C, Valerio M, Bouza E. Are incidence and epidemiology of anaerobic bacteremia really changing? Eur J Clin Microbiol Infect Dis. 2015 Aug;34(8):1621-9.
  3. Lyte M, Ernst S. Catecholamine induced growth of gram-negative bacteria. Life Sci. 1992;50(3):203-12.
  4. Lavigne JP, Sotto A, Dunyach-Remy C, Lipsky BA. New Molecular Techniques to Study the Skin Microbiota of Diabetic Foot Ulcers. Adv Wound Care (New Rochelle). 2015 Jan 1;4(1):38-49.
  5. Wilson JD, Lee RA, Balen AH, Rutherford AJ. Bacterial vaginal flora in relation to changing oestrogen levels. Int J STD AIDS. 2007 May;18(5):308-11. 
  6. Tidbury FD, Langhart A, Weidlinger S, Stute P. Non-antibiotic treatment of bacterial vaginosis-a systematic review. Arch Gynecol Obstet. 2021 Jan;303(1):37-45. 
  7. Boyanova L. Stress hormone epinephrine (adrenaline) and norepinephrine (noradrenaline) effects on the anaerobic bacteria. Anaerobe. 2017 Apr;44:13-19.
  8. Jentsch HF, März D, Krüger M. The effects of stress hormones on growth of selected periodontitis related bacteria. Anaerobe. 2013 Dec;24:49-54. 
  9. Charles PG, Uçkay I, Kressmann B, Emonet S, Lipsky BA. The role of anaerobes in diabetic foot infections. Anaerobe. 2015 Aug;34:8-13. 
  10. Montrief T, Long B, Koyfman A, Auerbach J. Fournier Gangrene: A Review for Emergency Clinicians. J Emerg Med. 2019 Oct;57(4):488-500.
  11. Shimizu T, Tokuda Y. Necrotizing fasciitis. Intern Med. 2010;49(12):1051-7.
  12. Tan JH, Koh BT, Hong CC, Lim SH, Liang S, Chan GW, Wang W, Nather A. A comparison of necrotising fasciitis in diabetics and non-diabetics: a review of 127 patients. Bone Joint J. 2016 Nov;98-B(11):1563-1568. 
  13. Lima SM, Grisi DC, Kogawa EM, Franco OL, Peixoto VC, Gonçalves-Júnior JF, Arruda MP, Rezende TM. Diabetes mellitus and inflammatory pulpal and periapical disease: a review. Int Endod J. 2013 Aug;46(8):700-9. 
  14. Sun JH, Chang HY, Chen KW, Lin KD, Lin JD, Hsueh C. Anaerobic thyroid abscess from a thyroid cyst after fine-needle aspiration. Head Neck. 2002 Jan;24(1):84-6. 
  15. Neelon FA, Mahaley MS Jr. Chiasmal syndrome due to intrasellar abscess. Arch Intern Med. 1976 Sep;136(9):1041-3. 
  16. Jin W, Miao Q, Wang M, Zhang Y, Ma Y, Huang Y, Wu H, Lin Y, Hu B, Pan J. A rare case of adrenal gland abscess due to anaerobes detected by metagenomic next-generation sequencing. Ann Transl Med. 2020 Mar;8(5):247.
  17. Rhoton-Vlasak A. Infections and infertility. Prim Care Update Ob Gyns. 2000 Sep 1;7(5):200-206.
  18. Hay PE. Bacterial vaginosis and miscarriage. Curr Opin Infect Dis. 2004 Feb;17(1):41-4.
  19. Kuon RJ, Togawa R, Vomstein K, Weber M, Goeggl T, Strowitzki T, Markert UR, Zimmermann S, Daniel V, Dalpke AH, Toth B. Higher prevalence of colonization with Gardnerella vaginalis and gram-negative anaerobes in patients with recurrent miscarriage and elevated peripheral natural killer cells. J Reprod Immunol. 2017 Apr;120:15-19.
  20. Eggert-Kruse W, Rohr G, Ströck W, Pohl S, Schwalbach B, Runnebaum B. Anaerobes in ejaculates of subfertile men. Hum Reprod Update. 1995 Sep;1(5):462-78.
  21. Hills RD Jr, Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients. 2019 Jul 16;11(7):1613. 
  22. Escobar JS, Klotz B, Valdes BE, Agudelo GM. The gut microbiota of Colombians differs from that of Americans, Europeans and Asians. BMC Microbiol. 2014 Dec 14;14:311.
  23. Sato J, Kanazawa A, Ikeda F, Yoshihara T, Goto H, Abe H, Komiya K, Kawaguchi M, Shimizu T, Ogihara T, Tamura Y, Sakurai Y, Yamamoto R, Mita T, Fujitani Y, Fukuda H, Nomoto K, Takahashi T, Asahara T, Hirose T, Nagata S, Yamashiro Y, Watada H. Gut dysbiosis and detection of "live gut bacteria" in blood of Japanese patients with type 2 diabetes. Diabetes Care. 2014 Aug;37(8):2343-50. 
  24. Brook I. Antimicrobials therapy of anaerobic infections. J Chemother. 2016 Jun;28(3):143-50. 

Androgen Physiology: Receptor and Metabolic Disorders

ABSTRACT

 

Androgens are an important class of C19 steroid hormones that control normal male development and reproductive function. The main circulating androgen is testosterone, which is produced in the Leydig cells of the testis and can also act as a pro-hormone after being metabolized to dihydrotestosterone (DHT) or estradiol (E2). The biological actions of testosterone and DHT are mediated by the androgen receptor, a member of the nuclear receptor superfamily, which in response to hormone regulates gene expression in target tissues. In this chapter the biosynthesis of androgens, receptor structure/function, and the consequences of genetic changes impacting on receptor expression and signaling in disorders of male development are discussed.

 

INTRODUCTION

 

Androgens are important hormones for expression of the male phenotype. They have characteristic roles during male sexual differentiation, but also during development and maintenance of secondary male characteristics and during initiation and maintenance of spermatogenesis (1, 2). The two most important androgens in this respect are testosterone and 5α-dihydrotestosterone [Figure 1].

Figure 1. Structure of testosterone and 5α-dihydrotestosterone and anti-androgens.

While acting through the same androgen receptor, each androgen has its own specific role during male sexual differentiation: testosterone is directly involved in development and differentiation of Wolffian duct derived structures (epididymides, vasa deferentia, seminal vesicles and ejaculatory ducts) [Figure 2A], whereas 5α-dihydrotestosterone, a metabolite of testosterone, is the active ligand in a number of other androgen target tissues, like urogenital sinus and tubercle and their derived structures (prostate gland, scrotum, urethra, penis) [Figure 2B] (3, 4).

Figure 2. Specific actions of testosterone (T) and 5α-dihydrotestosterone (DHT). A) Testosterone is synthesized in the testis under the control of luteinizing hormone (LH) from the pituitary. After entering target cells in the hypothalamus, pituitary, testis, and Wolffian duct, T binds to the androgen receptor (AR) and the T-AR complex binds to specific DNA sequences and regulates gene transcription, which can result in negative feedback regulation of gonadotrophin synthesis and secretion, in initiation and regulation of spermatogenesis, and in differentiation and development of Wolffian ducts. B) T is synthesized in the testis under the control of LH, enters target cells in urogenital sinus, urogenital tubercle, and several other androgen target tissues and is metabolized to DHT by the enzyme 5α-reductase type 2. DHT binds directly to the AR and the DHT-AR complex interacts with specific DNA sequences and regulates gene transcription resulting in differentiation and development of the prostate, the external genitalia, and during puberty several secondary male sex characteristics.

The interaction of both androgens with the androgen receptor is different. Testosterone has a twofold lower affinity than 5α-dihydrotestosterone for the androgen receptor, while the dissociation rate of testosterone from the receptor is five-fold faster than of 5α-dihydrotestosterone (5). However, testosterone can compensate for this "weaker" androgenic potency during sexual differentiation and development of Wolffian duct structures via high local concentrations due to diffusion from the nearby positioned testis. In more distally located structures, like the urogenital sinus and urogenital tubercle the testosterone signal is amplified via conversion to 5α-dihydrotestosterone.

 

ANDROGEN BIOSYNTHESIS

 

Androgens (testosterone and 5α-dihydrotestosterone) belong to the group of steroid hormones. The major circulating androgen is testosterone, which is synthesized from cholesterol in the Leydig cells in the testis. Testosterone production in the fetal human testis starts during the sixth week of pregnancy. Leydig cell differentiation and the initial early testosterone biosynthesis in the fetal testis are independent of luteinizing hormone (LH) (6-8). During testis development production of testosterone comes under the control of LH which is produced by the pituitary gland. Synthesis and release of LH is under control of the hypothalamus through gonadotropin-releasing hormone (GnRH) and inhibited by testosterone via a negative feedback mechanism [Figure 2A] (9).The biosynthetic conversion of cholesterol to testosterone involves several discrete steps, of which the first one includes the transfer of cholesterol from the outer to the inner mitochondrial membrane by the steroidogenic acute regulatory protein (Star) and the subsequent side chain cleavage of cholesterol by the enzyme P450scc (10). This conversion, resulting in the synthesis of pregnenolone, is the rate-limiting step in testosterone biosynthesis. Subsequent steps require several enzymes including, 3β-hydroxysteroid dehydrogenase, 17α-hydroxylase/C17-20-lyase and 17β-hydroxysteroid dehydrogenase type 3 [Figure 3] (11).

Figure 3. Biosynthetic pathways for testosterone and DHT synthesis. The classic pathway show testosterone synthesized from cholesterol with further metabolism to DHT. The alternative or “backdoor” pathway shows DHT production without going through testosterone. Note only some of the enzymes are shown for clarity.

METABOLISM OF TESTOSTERONE TO 5α-DIHYDRO-TESTOSTERONE

 

Metabolism of testosterone to 5α-dihydrotestosterone occurs through the classical pathway [Figure 3] and is essential for initiation of the differentiation and development of the urogenital sinus into the prostate [Figure 2B]. Differentiation of male external genitalia (penis, scrotum and urethra) also strongly depends on the conversion of testosterone to 5α-dihydrotestosterone in the urogenital tubercle, labioscrotal swellings, and urogenital folds (1). In recent research there has been considerable interest in the alternative or ‘backdoor’ pathway of DHT production (12 and references therein). This pathway has been found to have a significant role in the normal masculinization of the male fetus (see 13) and abnormal virilization of the female fetus in cases of congenital adrenal hyperplasia resulting from mutations in the enzyme P450 oxidoreductase (14).

 

The irreversible conversion of testosterone to 5α-dihydrotestosterone is catalyzed by the microsomal enzyme 5α-reductase type 2 (SRD5A2) and is NADPH dependent [Figure 4] (15). The cDNA of the gene for 5α-reductase type 2 codes for a protein of 254 amino acid residues with a predicted molecular mass of 28,398 Dalton (16, 17).

Figure 4. Metabolism of testosterone to DHT by the enzyme 5α-reductase type 2 (SDR5A2).

The NH2-terminal part of the protein contains a subdomain proposed to be involved in testosterone binding, while the COOH-terminal region is involved in NADPH-binding (3). The enzyme 5α-reductase type 2 belongs to the 5α-reductase family which is composed of 3 subfamilies with a total of 5 members (18). There are three isozymes: type 1, type 2 and the more recently discovered type 3, which has a role in the conversion of polyprenols to dolichols (important step in protein N-glycosylation) (19, 20). The other members are the proteins glycoprotein synaptic 2 (GPSN2) and glycoprotein synaptic 2 like (GPNS2L) and are most likely involved in double bond reduction during fatty acid elongation (21).

 

ANDROGEN ACTION

 

The Androgen Receptor and the Nuclear Receptor Family

 

Actions of androgens are mediated by the androgen receptor (NR3C4; Nuclear Receptor subfamily 3, group C, gene 4). This ligand-dependent transcription factor belongs to the superfamily of 48 known human nuclear receptors (22). This family includes receptors for steroid hormones, thyroid hormones, all-trans and 9-cis retinoic acid, 1,25 dihydroxy-vitamin D, ecdysone and activators of peroxisome proliferation (23-25). An increasing number of nuclear proteins have been identified with a protein structure homologous with that of nuclear receptors, but without a known ligand. These so-called "orphan" receptors form an important subfamily of transcription factors acting either in the absence of any ligand or with yet unknown endogenous ligands (26). Comparative structural and functional analysis of nuclear hormone receptors has revealed a common structural organization in 4 different functional domains: a NH2-Terminal Domain, a DNA-Binding Domain, a Hinge Region and a Ligand Binding Domain [Figure 5].

Figure 5. Steroid hormone receptor family. Sequence homologies between the human androgen receptor (hAR), human progesterone receptor (hPR), human glucocorticoid receptor (hGR), human mineralocorticoid receptor (hMR), and the human estrogen receptor alpha (hERα) and beta (hERβ).

The current model for androgen action involves a multi-step mechanism as depicted in Figure 6. Upon entry of testosterone into the androgen target cell, binding occurs to the androgen receptor either directly or after its conversion to 5α-dihydrotestosterone. Binding to the receptor is followed by dissociation of chaperone protein complexes (e.g., heat shock proteins) in the cytoplasm, simultaneously accompanied by a conformational change of the receptor protein resulting in a transformation and a translocation to the nucleus. The complex of chaperone and chaperone-associated proteins is collectively called the ‘foldosome’ and has functions beyond the classical role in the cytosol. The foldosome can for instance affect nuclear translocation and target gene expression (27, 28). Upon binding in the nucleus to specific DNA-sequences the receptor dimerizes with a second molecule and the homodimer entity recruits further additional proteins (e.g., coactivators, general transcription factors, RNA-polymerase II) via specific interaction motifs (29). This finally results in transcriptional activation or suppression of specific androgen responsive genes (30).

 

Figure 6. Simplified model of androgen action in an androgen target cell. The androgen receptor (AR) binds testosterone or its active metabolite DHT. After disassociation of heat shock proteins (hsp) the receptor enters the nucleus via an intrinsic nuclear localization signal and binds as a homodimer to specific DNA elements present as enhances upstream of androgen target genes. The next step is recruitment of coactivators, which form the communication bridge between the receptor and several components of the transcription machinery. The direct and indirect binding of the androgen receptor with several components of the transcription machinery (e.g., RNA polymerase II (RNA Pol II), general transcription factors (GTFs)) are key events in nuclear signaling. This communication triggers subsequent mRNA synthesis and consequently protein synthesis resulting in androgen responses. A non-genomic pathway involving the AR via cross-talk with the Src/Raf-1Erk-2 pathway is also known.

Interestingly androgen signaling via the androgen receptor can also occur in a non-genomic, rapid and sex-nonspecific way by crosstalk with the Scr, Raf-1, Erk-2 pathway [Figure 6] (31, 32). The classical androgen receptor is also involved in androgen-mediated induction of Xenopus oocyte maturation via the (MAPK)-signaling cascade in a transcription independent way (33, 34).

 

Cloning and Structural Organization of the Androgen Receptor Gene

 

Since cloning of the human androgen receptor cDNA our insights into the mechanism of androgen action have increased tremendously. Only one androgen receptor cDNA has been identified and cloned, despite the two different ligands (35-38). The concept of two hormones and one receptor to explain the different actions of androgens has been generally accepted and, according to the information available from the human genome project, it is very unlikely that additional genes exist coding for a functional nuclear receptor with androgen receptor-like properties (25).

 

The androgen receptor gene is located on the X-chromosome at Xq11.2 -12.  The gene spans 186,587 kilobases (kb) in total and codes for a protein with a molecular mass of approximately 110 kDa [Figure 7] (39, 40). The gene consists of 8 coding exons and the structural organization of the coding exons is essentially identical to those of the genes coding for the other steroid hormone receptors (e.g., exon/intron boundaries are highly conserved) and is characterized by unusually long 5’- and 3’-UTRs [Figure 7] (36, 41-43, 47). As a result of differential splicing in the 3' - untranslated region two androgen receptor mRNA species (of around 7.5 and 10 kb, respectively) have been identified in several human tissues and cell lines (36): only the larger transcript is seen in rodent tissues (36, 43, 47). There is no structural indication in the androgen receptor mRNA for any preferential use of either of the two transcripts or transcript specific functions, but it can be speculated that tissue-specific factors may determine which transcript is present in which androgen target tissue. In the human prostate and in genital skin fibroblasts the 10 kb size mRNA is predominantly expressed (43). It may also be significant that a number of micro-RNAs have been identified and validated that target the 3’-UTR that are likely to contribute to the regulation of receptor levels (44-46) [Figure 7].

Figure 7. Human androgen receptor gene was mapped to the long arm of the X chromosome. The human androgen receptor gene consists of coding exons and unusually long 5’- and 3’ UTRs. These have been shown to be important for transcriptional regulation (binding sites for both positive and negative regulatory factors) in the case of the 5’UTR. The 3’UTR region of the mRNA is targeted by a number of microRNAs (miRNAs). The androgen receptor has been shown to downregulate its own mRNA through response elements located in the 5’UTR and exon 2.

Regulation and Expression of the Androgen Receptor Gene

 

The promoter for the androgen receptor gene lacks TATA and CCAAT elements and transcription is driven primarily by the Zn-finger transcription factor Sp1. Sp1 binds to GC-boxes upstream of the transcription start site (-46 to -41 bps) and within the 5’UTR (+429 to +442) (47-52) [Figure 7]. In addition, the promoter and the region spanning the 5’-UTR and exon 1 contains a CpG island that demonstrates tissue-selective methylation patterns (53) and to be associated with loss of AR expression in prostate cancer (54).

 

Transcription of the receptor gene is under both positive and negative regulation (55, 58). Recent studies have focused on the auto-down regulation of the receptor mRNA in prostate cells. Balk and co-workers (56) identified, using chromatin immunoprecipitation (ChIP), binding sites for ligand bound androgen receptor within the second intron and a second negative androgen response elements has been characterized in the 5’UTR (+611 bp) of the human receptor gene (57). Unravelling the molecular mechanism(s) for androgen-dependent down regulation, including possible synergy between the identified elements, in different cell types and tissues is an active area of research (58).

 

In addition to regulation by hormone, recent work has also highlighted the importance of the balance between positive (Sp1) and negative (Purα) transcription factors binding to the 5’UTR of the human gene in determining the expression of receptor mRNA in different prostate cancer cell models (52 and references therein).

 

Androgen Receptor Polymorphisms

 

The androgen receptor DNA-binding and ligand-binding domains have a high homology with the corresponding domains of the other members of the steroid receptor subfamily (59) [Figure 5].

 

There is a remarkably low homology between the androgen receptor NH2-terminal domain and that of the other steroid receptors [Figure 5, see above] (60-65). A poly-glutamine stretch, encoded by a polymorphic (CAG)nCAA - repeat is present in the NH2-terminal domain (66). The length of the repeat has been used for identification of X-chromosomes for carrier detection in pedigree analyses (67, 68). Variation in length (9 - 38 glutamine residues) is observed in the normal population and has been suggested to be associated with a very mild modulation of androgen receptor activity (69). This assumption is based on in vitro experiments after transient transfection of androgen receptor cDNA's containing (CAG)nCAA - repeats of different lengths (70, 71). In translating this finding to the in vivo situation, it can be envisaged that either shorter or longer repeat lengths can result in a relevant biologic effect during life. This concept has been explored in epidemiological studies of men with prostate cancer or infertility. With respect to prostate cancer, a clear picture has not emerged, and controversy persists. In several studies, shortening of the (CAG)nCAA repeat length in exon 1 of the androgen receptor gene was found to correlate with an earlier age of onset of prostate cancer, and a higher tumor grade and aggressiveness (72-74). However, in other epidemiological studies in prostate cancer patients these associations were not confirmed (75, 76).

 

In several investigations in male infertile patients an association was found between a longer (CAG)nCAA repeat and the risk of defective spermatogenesis (77-79). This suggests that a less active androgen receptor, due to a moderate expanded repeat length, may be a factor in the etiology of male infertility.

 

The (CAG)nCAA - repeat in exon 1 of the androgen receptor gene is expanded in patients with spinal and bulbar muscular atrophy (SBMA) and varies between 38 and 75 repeat units (69, 80, 81). Spinal and bulbar muscular atrophy is characterized by progressive muscle weakness and atrophy and is associated with nuclear accumulation of androgen receptor protein with the expanded polyglutamine stretch in motor neurons. Clinical symptoms usually manifest in the third to fifth decade and result from severe depletion of lower motor nuclei in the spinal cord and brainstem (69, 82, 83). SBMA patients frequently exhibit endocrinological abnormalities including testicular atrophy, infertility, gynecomastia, and elevated LH, FSH and estradiol levels. Sex differentiation proceeds normally, and characteristics of mild androgen insensitivity appear later in life.

 

The neurotoxicity of the polyglutamine androgen receptor may involve generation of NH2-terminal truncation fragments, as such peptides occur in SBMA patients, but several other mechanisms have also been suggested for the molecular basis of SBMA (84, 85). Therapeutic approaches in SBMA are focusing on reducing nuclear localized mutant androgen receptor via enhanced mutant androgen receptor degradation or by disrupting the interaction with androgen receptor coregulators (86, 87). In a phase 3 clinical trial it was shown that the use of leuprorelin, a synthetic neuropeptide with an inhibitory action on LH secretion and consequently on testicular testosterone synthesis, is associated with improved swallowing function in SBMA patients, suggesting that interference by a pharmacon in testosterone-mediated AR aggregation can be a potential therapy in SBMA patients (88). The selective action of dutasteride (a 5α-reductase inhibitor) in motor neurons, by reducing significantly the formation of the active androgen 5α-dihydrotestosterone, resulted in a slowdown of the progression of SBMA and illustrated that active androgen depleting therapies can be promising in the treatment of SBMA (89).

 

In general patients with an expanded CAG repeat are expected to have a low incidence of prostate cancer. However, a rare case has been reported in which a high stage prostate cancer has been detected in a SBMA patient, which responded to a maximal androgen blockade therapy (90).

 

An important step in the receptor-mediated mechanism of action of androgens involves the NH2-terminal domain interacting with the COOH-terminal ligand binding domain (N/C interaction). (See details below under ‘Androgen Receptor Functional Domain Structure’). This N/C interaction is also a prerequisite for androgen receptor aggregation and toxicity in SBMA. Interference of the N/C interaction by selective androgen receptor modulators ameliorates aggregation and toxicity (91).

 

The androgen receptor is a substrate for numerous post-translational modifications (see below) and phosphorylation of serine 516 has been associated with cleavage of the receptor and cytotoxicity (92). In contrast, phosphorylation of serines 215 and 793, by Akt kinase, was found to prevent nuclear translocation and receptor transactivation (93). Interestingly, methylation on arginine residues 210, 212, 787, 789 enhanced cytotoxicity and the authors proposed that this was as a consequence of mutual antagonism of phosphorylation (serines 215, 792) and arginine methylation (94). Similarly, prevention of SUMOylation rescues the SBMA phenotype in a mouse model by enhancing receptor-dependent transcriptional activity (95).

 

The isoflavone genistein, which is derived from soy, is a potential therapeutic agent in SBMA, because this androgen receptor modulator can effectively disrupt the interaction between the co-regulator ARA70 and the androgen receptor and promotes the degradation of the mutant receptor in neuronal cells. (96). Similarly, targeting molecular chaperone complexes with small molecule modulators (e.g., 17-AAG, YM-1) has been shown to reduce neurotoxicity and enhance receptor-dependent degradation (reviewed in 81).

 

Several therapeutic approaches have been investigated at different levels in the androgen receptor signaling pathway and aggregation process, in SBMA mouse models. However, translating these results to the human situation in SBMA patients has its limitations and is far from a complete cure of SBMA patients (97, 98).

 

ANDROGEN RECEPTOR AMINO ACID NUMBERING

 

The current sequence of the androgen receptor cDNA and the amino acid numbering of the corresponding protein is based on the NCBI reference sequence NM_000044.3. This is different from the original numbering scheme used over the past 20 years that was based on Gen-Bank mRNA sequence M20132.1 (36).

 

In order to correctly identify mutations previously published, the following changes should be kept in mind: the variable polyglutamine tract length is two longer (23 instead of 21), whereas the variable polyglycine tract length is one shorter (23 instead of 24) for NM_000044.3 versus M20132.1, respectively. Consequently, the androgen receptor protein of the new reference sequence is one amino acid longer, that is, 920 residues, leading to a +2 shift in amino acid numbering between residues 78 and 449 and to a +1 shift between residues 472 and 919 compared with the previously used standard reference sequence. The +1 shift involves all the amino acid residues in the DNA-binding domain (DBD) and ligand-binding domain (LBD). The new reference numbering is further explained and illustrated in Figure 8 and will be used throughout the text.

Figure 8. Reference numbering of the androgen receptor (AR) of protein. The numbering of the amino acid residues is according to National Center for Biotechnology Information (NCBI) reference sequence number NM_000044.3, which refers to a gene size of 187,246 nucleotides and an AR protein of 920 amino acid residues with a polyglutamine tract of 23 and a polyglycine tract of 23 (110). Amino acid numbering +2 between 78 and 449; Amino acid numbering +1 between 472 and 919. In addition, a number of splice variants of the AR have been identified in prostate cancer cell lines and patient samples. These splice variants lack most or all of the LBD but retain a functional DBD and NTD with unique C-terminal sequences derived from cryptic exons (CE) (e.g., AR-v7).

ANDROGEN RECEPTOR: FUNCTIONAL DOMAIN STRUCTURE

 

The NH2-terminal Domain

 

The androgen receptor NH2-terminal domain (NTD) harbors the major transcription activation functions and several structural subdomains. The NTD of the androgen receptor, as that of the other steroid receptors, can be considered as an intrinsically disordered protein domain, existing as an ensemble of conformers. It has a structure between a fully unfolded state and a structured folded conformation: this molten-globule-like conformation has the propensity to form helical structures, despite its structurally plasticity (99-102). Within its 539 amino acids, two independent activation domains have been identified: activation function 1 (AF-1) (located between residues 103 and 372) that is essential for transcriptional activity of full-length androgen receptor, and activation function 5 (AF-5) (located between residues 362-486) that is required for transactivity of a constitutively active androgen receptor, which lacks its LBD (103). Evidence is available now that the AF-5 region in the receptor NH2-terminal domain interacts with a glutamine rich domain in p160 cofactors like SRC-1 and TIF2/GRIP1 and not with their LxxLL-like protein interacting motifs (104-107).

 

Recent years have seen further structural and functional insights into the intrinsically disordered NTD. Key discoveries include the high-resolution mapping of helical regions within the AF1 domain (108) that were in very good agreement with previous predictions (109); and the identification of a helical segment involving the WHTLF motif responsible for TFIIF binding (110). Also of note are helical regions mapping to the poly-Q and adjacent leucine stretch (111) and the sequence immediately preceding the DBD (112). Collectively, these studies emphasize the presence of helical regions within the NTD and its propensity to adopt a more helical structure underpinning function.

 

Another function of the androgen receptor NH2-terminal domain is its binding to the COOH-terminal LBD (N/C interaction) (113, 114). The NH2-terminal regions required for the binding of the LBD have been mapped to two essential units: the first 36 amino acids and residues 372-495 (115).

 

The hormone dependent interaction of the NH2-terminal domain with the ligand binding domain can play a role in stabilization of the androgen receptor dimer complex and in stabilization of the ligand receptor complex by slowing the rate of ligand dissociation and decreasing receptor degradation (116, 117). Agonists like T and DHT, but not antagonists like hydroxyflutamide or bicalutamide induce the N/C interaction in full length receptor. In a FRET (fluorescence resonance energy transfer) study it was clearly shown that the androgen receptor N/C interaction is rapidly initiated in the cytoplasm after hormone binding as an intramolecular interaction and is followed by an intermolecular N/C interaction in the nucleus, contributing to receptor dimerization (118). The N/C interaction occurs preferentially in the mobile androgen receptor, where it protects the coactivator binding groove for ultimately unfavorable protein-protein interactions. Specifically bound to DNA, the N/C interaction is lost allowing cofactor binding (119). Several mutations in the ligand binding domain, detected in patients with the syndrome of androgen insensitivity, negatively affect the interaction of the NH2-terminal domain with the ligand binding domain, while androgen binding was impaired, indicating the importance of this interaction (120).

 

In addition to the role of the NH2-terminal domain in protein-protein interactions it has also been reported to modulate DNA binding, leading to a lower apparent binding affinity for both selective and non-selective response elements (see also below) (121). These findings suggest a further role of the NH2-terminal domain, in interdomain interactions and allosteric regulation of receptor activity.

 

The DNA-binding Domain

 

The DNA-binding domain is the best conserved among the members of the receptor superfamily [Figure 5]. It is characterized by a high content of basic amino acids and by nine conserved cysteine residues [Figure 9A]. Detailed structural information has been published on the crystal structure of the DNA-binding domain of the glucocorticoid receptor complexed with DNA (122). 3D-information is also available for the androgen receptor-DNA interaction on an artificial DNA response element (123) [Figure 9B]. The folding of the DBD is similar to that reported for the glucocorticoid and estrogen receptor DBDs.

Figure 9. Structure of the DNA binding domain of the androgen receptor. A) The protein structure is represented in the one letter code. The domain consists of two zinc cluster modules, which are stabilized by the coordination binding of a zinc atom (red dot) by 4 cysteine residues (yellow). The first zinc cluster contains the P-box (proximal box) of which three residues determine androgen response element recognition. The second zinc cluster contains the D-box (distal box) in which amino acids are located that are involved in protein-protein interactions with a second receptor molecule in the homodimer complex. B) Structure of the AR-DBD bound to DNA (Pdb 1R41). C) Consensus androgen receptor response element.

Briefly, the DNA-binding domain has a compact, globular structure in which three substructures can be distinguished: two zinc clusters and a more loosely structured carboxy terminal extension (CTE) (124). Both zinc substructures contain centrally one zinc atom which interacts via coordination bonds with four cysteine residues [Figure 9].

 

The two zinc coordination centers are both C-terminally flanked by an α-helix (122, 123). The two zinc clusters are structurally and functionally different and are encoded by two different exons [see Figures 7 and 8]. The α-helix of the most N-terminal located zinc cluster interacts directly with nucleotides of the hormone response element in the major groove of the DNA. Three amino acid residues at the N-terminus of this α-helix are responsible for the specific recognition of the DNA-sequence of the responsive element [Figure 9A]. These three amino acid residues, the so-called P(proximal)-box [Gly; Ser; Val;] are identical in the androgen, progesterone, glucocorticoid and mineralocorticoid receptors, and differ from the residues at homologous positions in the estradiol receptor. It is not surprising therefore, that the androgen, progesterone, glucocorticoid and mineralocorticoid receptors can recognize the same response element. The receptor DNA binding domain requires a CTE of minimally four residues (amino acids 626 – TLGA – 629) for proper binding to an ARE (androgen response element) with an inverted repeat of high affinity ARE-half sites and a CTE of at least twelve residues (amino acids 626 – TLGARKLKKLGN – 637) for binding to an ARE with one high and one low affinity half site (125). For the hormone and tissue-specific responses of the different receptors additional determinants are needed. Important in this respect are DNA-sequences flanking the hormone response element, receptor interactions with other proteins and receptor concentrations. The second zinc cluster motif is involved in protein-protein interactions such as receptor dimerization via the so-called D(distal)-box [Figure 9A and B] (122, 123).

 

DNA Response Elements for the Androgen Receptor

 

In vitro the androgen receptor binds to 15 bp palindromic sequences [Figure 9C]. These non-selective elements are also recognized and bound by the glucocorticoid, mineralocorticoid and progesterone receptors. In contrast, androgen response elements demonstrate selectivity for the receptor. In an animal model, termed Specificity-affecting androgen receptor Knock-in or SPARKI, where the androgen receptor-DBD has been replaced by that of the glucocorticoid receptor-DBD, binding to selective AREs is disrupted (126). These mice have a reproductive phenotype, with male reproductive tissues having reduced weight and size and the animals showing reduced fertility. Interestingly the SPARKI males also demonstrated differential gene expression with the Rhox5 mRNA significantly reduced which correlated with a role for a selective ARE, necessary for receptor-dependent transcription of this gene (126).

 

More recently a number of genome-wide studies, using chromatin immunoprecipitation (ChIP), have increased our knowledge of androgen-regulated genes and have demonstrated a significant variability in DNA response element architecture, with imperfect palindromic sequences and half-sites identified as potential receptor binding sites (30, 127-131). These studies have also highlighted the enrichment of pioneering factors, such as FOXA1 and GATA2 in close proximity to receptor binding sites (30, 127-131).

 

The Hinge Region

 

Between the DNA-binding domain and the ligand binding domain is located a non-conserved hinge region, which is also variable in size in different steroid receptors [Figure 5]. The hinge region can be considered as a flexible linker between the ligand binding domain and the rest of the receptor molecule. The hinge region is important for nuclear localization and contains a bipartite nuclear localization signal. Co-repressor binding can also occur via the hinge region (125). In some nuclear receptors, including the androgen receptor, acetylation can occur in the hinge region at a highly conserved consensus site [KLLKK] [Figure 11, see below] (132, 133).

 

The Ligand Binding Domain

 

Finally, the second-best conserved region is the hormone binding domain. This domain is encoded by approximately 250 amino acid residues in the C-terminal end of the molecule [Figure 5, see above] (37, 60-63, 134). The crystal structure of the human androgen receptor ligand binding in complex with the synthetic ligand methyltrienolone (R1881) and 5α-dihydrotestosterone, respectively, have been determined [Figure 10A and B] (135, 136).

Figure 10. Structure of the ligand binding domain of the human androgen receptor. A) The crystal structure of the LBD with DHT bound (pdb 1137). Specific amino acid- hormone interactions are illustrated in the right-hand panel. B) The LBD structure with the synthetic agonist R1881 and a coactivator peptide with an FxxLF motif bound to AF2 region (pink oval) (pdb 1XOW). C) Structure of the LBD showing the location of the BF3 pocket (blue oval) with triiodothyroacetic acid/TRIAC bound (pdb 2PKL).

The 3-dimensional structure has the typical nuclear receptor ligand binding domain fold (59). Interestingly the ligand binding pocket consists of 18 amino acid residues interacting more or less directly with the bound ligand, with a relatively small number of specific hydrogen-bonds and hydrophobic interactions determining hormone-selectivity [Figure 10A] (135). The ligand binding pocket is somewhat flexible and can accommodate ligands with different structures. The structural data are being used in designing optimized selective androgen receptor modulators (SARMs) (137, 138). Several AR mutations found in prostate tumors have been investigated functionally, including T878S, T878A, H875T, V716M, W742C, and L702H as a single mutation or in combination with T878A. Similar to T878A these AR mutations have a broadened ligand specificity and are activated by different low affinity ligands like estradiol, progesterone, glucocorticoids and different partial and full antagonists (139-146).

 

Crystallographic data on the ligand binding domain complexed with agonist predict 11 helices (no helix 2) with two anti-parallel β-sheets arranged in a so-called helical sandwich pattern. In the agonist-bound conformation the carboxy-terminal helix 12 is positioned in an orientation allowing a closure of the ligand binding pocket. Upon hormone binding the fold of the ligand binding domain results in a globular structure with an interaction surface for binding of interacting proteins like co-activators (AF2) [Figure 10B]. In this way the androgen receptor selectively recruits a number of proteins and can communicate with other partners of the transcription initiation complex. Crystallization studies of wild type androgen receptor ligand binding domain with antagonists have not been reported so far. However, the structural consequences of surface modulatory compounds on the receptor LBD crystals complexed with DHT are promising for future developments of new androgen receptor modulators including a new type of androgen receptor antagonists (147).

 

The androgen receptor can use different transactivation domains (AF1 and AF5, respectively, in the NH2-terminal domain and AF2 in the COOH-terminal domain) depending on the "form" of the receptor protein [Figure 8, see above] (103). The AF2 function in the ligand binding domain is strongly dependent on the presence of nuclear receptor coactivators. In vivo experiments favor a ligand-dependent functional interaction between the AF-2 region in the ligand-binding domain with the NH2-terminal domain (113, 115). In fact, the AF2 surface demonstrates a preference for more bulky hydrophobic amino acids over the LxxLL motif and the structural basis for this has been described (148-150). Thus, the receptor NTD FxxLF motif [Figure 10B] is more effective at forming a charge clamp with Glu898 and Lys721 and burying the phenylalanine residues into the AF2 pocket, whereas peptides containing the sequence LxxLL make weaker and fewer contacts with the LBD.

 

Interestingly, a previously unknown regulatory surface cleft, named BF-3, has been identified in the receptor LBD (147) [Figure 10C]. BF-3 comprises of Ile-673, Phe-674, Pro-724, Gly-725, Asn-728, Phe-827, Glu-830, Asn-834, Glu-838 and Arg-841. The androgen receptor transcriptional activity and co-activator binding can be decreased by binding of thyroid hormones triiodothyronine (T3) and TRIAC and three non-steroidal anti-inflammatory drugs to the BF-3 pocket. In addition, several mutations of the amino acid residues of BF-3 have been found in subjects with either androgen insensitivity syndrome (AIS, loss of function mutation) or in prostate cancer (gain of function mutation) (151). Mutational analyses have shown the requirement of several of these amino acid residues for receptor-dependent transcriptional activity. However, these analyses have been performed only in the presence of DHT (147). The influence of each of these residues in the presence of T3, TRIAC or other nonsteroidal anti-inflammatory drugs is therefore unknown.

 

A long-standing question in the field concerning dimerization of the AR-LBD has recently been resolved with new crystallographic studies (152, 153). The work from the Estébanez-Perpiñá group identified sequences in helix 5 as novel dimerization interface. Significantly, a number of point mutations associated with androgen insensitivity or prostate cancer map to this region emphasizing its functional importance for AR signaling (152).

 

Androgen Receptor Splice Variants Lacking the LBD

 

Deletions in the ligand binding domain abolish hormone binding completely (154). Deletions in the N-terminal domain and DNA-binding domain do not affect hormone binding. Deletion of the ligand binding domain leads to a constitutively active androgen receptor protein with trans-activation capacity comparable to the full-length androgen receptor (154). Thus, it appears that the hormone binding domain acts as a repressor of the trans-activation function in the absence of hormone. This regulatory function of the androgen receptor ligand binding domain in the absence of hormone, is not unique for the androgen receptor and has been reported also for the glucocorticoid receptor (155).

 

The generation of NH2-terminal splice variants involves the use of cryptic exons (AR-v1and -v7) or exon skipping (AR-v12) [Figure 8] (156). Androgen receptor variants have been shown to regulate similar patterns of gene expression to the full-length hormone-bound receptor (157). However, intriguingly there are a growing number of studies reporting unique sets of genes expressed by AR-v7 (157, 158), both expected and variant-specific target genes for AR-v12 (159) or differential regulation of classical androgen receptor-target genes (160). Importantly, these constitutively active splice variants have been identified in prostate cancer cell-lines, xenographs and prostate cancer patients undergoing androgen ablation therapy (157-159, 161-163).

 

Structural Insights from a DNA-bound Complex of Full-length Androgen Receptor

 

A major development in our understanding of AR mechanism of action has been the recent description of the structure of the full-length receptor bound to DNA and co-regulatory proteins (SRC-3 and p300) (164). The structures of the receptor alone or in a transcriptional active complex were solved by cryo-EM at resolution of 12 to 20 Å and reveal several interesting features. Particularly striking is the folding of the NTD of each monomer into a ‘life buoy ring’ surround the LBD-DBD dimer, creating a platform for SRC-3 and p300 binding, as well as N/C interaction and contacts between the NTDs (164). In contrast to a similar structure of the estrogen receptor α (165), only one molecule of SRC-3 is bound to the AR and the conformation of each NTD is proposed to be different based on visualizing antibodies recognizing the very N-terminus and AF1 regions resulting in an asymmetric appearance. This could have implications for protein-protein interactions and transcriptional regulation: for, example does the conformation of the NTDs change depending on the nature of the DNA binding site? It was also of note that the binding of p300 was enhanced in the presence of SRC-3, suggesting the latter stabilized the binding of the former. However, it is worth noting that previous biochemical studies demonstrated folding of the AR-AF1, using a chemical chaperone (TMAO) or an SRC-1 polypeptide, similarly enhanced subsequent co-regulatory protein binding (166) supporting a model of induced folding of AF1 and assembly of transcription complexes.

 

ANDROGEN RECEPTOR POSTTRANSLATIONAL MODIFICATIONS

 

Methylation, Acetylation, Ubiquitination and SUMOylation

 

The androgen receptor protein can be extensively covalently modified either by methylation, acetylation, ubiquitination, SUMOylation or phosphorylation [Figure 11] (132, 133, 167-171).

Figure 11. Post-translational modifications of the human androgen receptor. AC, acetylation of lysine residues (631, 633, and 634); CH3, methylation of lysine (633); P, phosphorylation of serines (16, 83, 96, 215, 258, 310, 426, 516, 651, and 792); SUMO-1, sumoylation on lysines (388 and 521); Ub, ubiquitination of lysines (846 and 848).

All these reactions are reversible and consequently enzymes that mediate dephosphorylation, deacetylation, deubiquitination, demethylation and de-SUMOylation are also potential regulators of androgen receptor activity. A total of 23 sites in the androgen receptor protein have been identified undergoing direct modification (170). These posttranslational modifications can contribute significantly to androgen receptor structure, activity and stability. It has been shown for instance that the histone methyltransferase SET9 is able to methylate the receptor in the hinge region at the Lysine residues 631 and 633 resulting in enhancement of transcriptional activity of the receptor (172, 173). The same Lysine residues together with Lysine 634 can be acetylated and the acetylation-deficient mutants have a decreased transcriptional activity, while the acetylation-mimetic mutations showed an enhanced transcriptional activity (132, 174). Recently, phosphorylation of serine 83 was observed to result in recruitment of the histone acetyltransferase p300, acetylation of the receptor and enhanced receptor stabilization and transcriptional activity (175).

 

Conversely, disruption of acetylation, through mutating the lysine residues or knock-down of p300 resulted in receptor ubiquitination and degradation. This study elegantly demonstrates how different post-translational modifications of the androgen receptor can work in concert to regulate receptor expression and activity. RNF6 dependent ubiquitination of Lysine residues 846 and 848 in the receptor protein results in recruitment of the coregulator ARA54 by the androgen receptor and directly determines promoter selectivity and specificity of the receptor (176).

 

SUMOylation of the androgen receptor occurs at two sites Lysine residues 388 and 521, but SUMOylation only at Lysine 388 results in a significant reduction of transcriptional activity (177). However, recent, whole genome analysis revealed that SUMOylation regulated both receptor recruitment to DNA and target gene selection (178). Significantly, the physiological importance of SUMOylation has been demonstrated in a knock-in mouse model, ARKI, where the SUMOylation sites were mutated to arginine (179). Male animals developed normally but were found to be infertile due to defects in epididymal sperm maturation. Crucially, In the ARKI animals the AR-dependent transcriptional activity was impaired in the epididymis and there was an absence of receptor SUMOylation linking this PTM to normal male reproduction and fertility (179).

 

Phosphorylation

 

The androgen receptor can be phosphorylated at serine, threonine and tyrosine residues (170, 171, 180, 181). Immediately after translation the androgen receptor becomes phosphorylated resulting in the appearance of two isoforms separable by SDS-polyacrylamide gel electrophoresis (182). The non-phosphorylated faster migrating 110 kDa isoform is converted into a 112 kDa phospho-isoform. Mutational analysis of serine 83 or serine 96 in the androgen receptor NH2-terminal domain abolishes this up-shift indicating that phosphorylation of these serine residues likely contributes to the phosphorylation of the 112 kDa androgen receptor isoform (70, 183). Phosphorylation of Serine 83 by CDK9 stabilizes androgen receptor chromatin binding, mediates transcriptional activity and can influence prostate cancer cell growth (184, 185). This serine residue is also phosphorylated after stimulation of Plexin-B1 resulting in nuclear translocation of the receptor protein (186). Three other androgen receptor phosphorylation sites have been identified using mutational analysis and trypsin-digestion of 32P-labelled androgen receptor followed by HPLC analysis and Edman degradation (183, 187, 188). These include the serine residues at position 516, 651, and 663. Ser-516 phosphorylation by MAP kinase is linked to altering the nuclear cytoplasmic shuttling and to the EGF-induced increase in androgen receptor transcriptional activity (189). Furthermore, androgen receptor intranuclear localization and transcriptional activity has been correlated with phosphorylation of serine 310 by CDK1, demonstrating a role for phosphorylation in regulating the receptor in a cell-cycle-dependent manner (181, 190). Transcription factor TFIIH also phosphorylates the receptor at Ser516 and is an essential partner in the cyclic recruitment of the transcription machinery (191). Substitution of serine 651 reduced androgen receptor activity by up to 30%. Furthermore, dephosphorylation of receptor phosphorylated at serine 651 by protein phosphatase 1 (PP1) can modulate androgen receptor translocation to the nucleus (192). More recently, PP1α has been shown to bind to the receptor-LBD and prevent ubiquitination and receptor degradation (193). Several other sites have been identified in the NH2-terminal domain at positions S16, S215, S258, S310, and S426 (180, 194-196). The function of phosphorylation of these sites is in the majority of the cases unknown or controversial. Two additional sites (S579 and S792) have been identified and characterized in the DNA-binding and ligand binding domains, respectively (189, 197). Phosphorylation of serine 579 by PKC kinase alters the nuclear cytoplasmic shuttling and elimination of phosphorylation at serine 579 eliminates EGF-induced transcriptional activation (189).

 

Besides the basal phosphorylation resulting in the 110-112 kDa doublet, addition of androgen induces another shift and the generation of a 110-112-114 kDa androgen receptor triplet (70). This triplet is the result of both an addition and a redistribution of phosphorylated sites, however, it is unknown which exact residues are involved (198). Interestingly, mutations that inactivate androgen receptor function, such as mutations resulting in loss of DNA binding or transactivation, inhibit the formation of the 114 kDa isoform. This suggests that part of the androgen - induced phosphorylation occurs during or after androgen receptor transcription regulation (70).

 

Functional phosphorylation at three tyrosine residues has also been demonstrated and extensively studied. The androgen receptor tyrosine residue 536 is highly phosphorylated. This phosphorylation is induced by EGF via activation of Src tyrosine kinase and may be important for prostate cancer cell growth under androgen-depleted conditions (199, 200). Activation of Cdc42-associated tyrosine kinase Ack1 can result in phosphorylation of tyrosine residues 269 and 365 enhancing androgen receptor transcriptional function and promoting androgen independent prostate cell growth (200, 201) and disrupting phosphorylation primarily of tyrosine 269 results in impaired nuclear localization (202). Recently it was reported that threonine phosphorylation of the receptor can also occur. Aurora A induces androgen receptor transactivation activity by phosphorylation of Threonine residue 284 (203).

 

In conclusion, phosphorylation of the androgen receptor can occur at serine, threonine and tyrosine residues by specific kinases and can be directly or indirectly linked to activation upon hormone binding, altering of nuclear cytoplasmic shuttling, modulation of DNA binding and transcriptional activity (168, 170, 181, 199, 204). Furthermore, phosphorylation of the androgen receptor can play an essential role in the hormone-independent activation of the androgen receptor by protein kinases in the MAPK and AKT (protein kinase B) signaling pathways, activated either through HER-2/neu or growth factors (205, 206).

 

ANTI-ANDROGENS AND SELECTIVE ANDROGEN RECEPTOR MODULATORS

 

Androgen receptor antagonists are compounds that interfere in some way in the biological effects of androgens and are frequently used in the treatment of androgen-based pathologies. The steroidal anti-androgens, cyproterone acetate (CPA) and RU38486 (RU486; mifepristone), have partial agonistic and antagonistic actions. Interestingly both compounds also display partial progestational and glucocorticoid actions and are therefore not considered to be pure anti-androgens. The non-steroidal anti-androgens hydroxyflutamide, nilutamide and bicalutamide [see Figure 1] are pure antiandrogens (207-209). Recent developments have led to the generation and marketing of second-generation non-steroidal antiandrogens, such as enzalutamide (formerly called MDV3100) [Figure 1], which have been reported to be more effective at blocking receptor nuclear translocation and activity (210). Recently, two new anti-androgens apalutamide and darolutamide have received FDA approval for treatment of non-metastatic castrate resistant prostate cancer (see 211, 212). However, resistance to enzalutamide has now also been identified as a result of an Phe876Leu point mutation in the LBD (213) and the expression of NH2-terminal domain splice variants (163) in CRPC, emphasizing the need for continued research and development of strategies to switch off androgen receptor signaling.

 

Mechanism of Action of Antiandrogens

 

In contrast to the full antagonists hydroxyflutamide and bicalutamide, CPA and RU486 can partially activate the androgen receptor with respect to transcription activation (214). With a limited proteolytic protection assay, it was demonstrated that binding of androgens by the androgen receptor results in two consecutive conformational changes of the receptor molecule. Initially, a fragment of 35 kDa, spanning the complete ligand binding domain and part of the hinge region, is protected from digestion by the ligand. After prolonged incubation times with the ligand a second conformational change occurs resulting in protection of a smaller fragment of 29 kDa (214, 215). In the presence of several anti-androgens (e.g., cyproterone acetate, hydroxyflutamide and bicalutamide) only the 35 kDa fragment is protected from proteolytic digestion, and no smaller fragments are detectable upon longer incubations. Obviously, the 35 kDa fragment can be associated with an inactive conformation, whereas the second conformational change, only inducible by agonists and considered as the necessary step for transcription activation, is lacking upon binding of anti-androgens.

 

During treatment of advanced prostate cancers, resistance develops to several of the above-mentioned anti-androgens, mostly due to mutations rendering the receptor protein less sensitive to anti-androgens. Promising results were reported for a newly developed second generation of antiandrogens for castration resistant prostate cancer (CRPC): ASC-J19, enzalutamide (MDV3100), apalutamide (ARN-509), AZD 3514, Compound30 and VPC-3033. (87, 210, 216-220). Characteristics of this new generation of anti-androgens are androgen displacement, inhibition of receptor- mediated transcription and enhancement of androgen receptor degradation. Clinical applications in prostate cancer were reported for enzalutamide (221-223). However, resistance to enzalutamide and apalutamide has been reported in prostate cancer due to a mutation at residue Phe877Leu (213, 224). Interestingly this mutation is located in a residue next to the LNCaP prostate cancer cell line mutation Thr878Ala (139, 225), supporting the view that this region in the ligand binding domain of the androgen receptor is very susceptible to mutagenesis in prostate cancer, which may lead to the tumor becoming resistant to hormone-based therapies.

 

Selective Androgen Receptor Modulation (SARMs)

 

Androgen signaling via the androgen receptor can occur in a non-genomic, rapid and sex-nonspecific way by crosstalk with the Scr, Raf-1, Erk-2 pathway [Figure 6, see above] (31, 32, 226). The anti-apoptotic action via the androgen receptor in bone cells (osteocytes, osteoblasts), and also in HeLa cells, could be induced by androgens and estrogens and inhibited by antiandrogens as well as anti-estrogens. The anti-apoptotic action appeared to be dissociated from the genomic action of the androgen receptor. The progesterone-induced oocyte maturation in Xenopus laevis also appears to be mediated in a non-genomic way by androgens and the androgen receptor via activating the MAPK pathway after the rapid conversion of progesterone to androstenedione and testosterone (33). These findings stimulated the development of new compounds (SARMs) which can selectively activate the androgen receptor either in a non-genomic pathway or in a genotropic transcriptional activation pathway. The term SARM (= Selective Androgen Receptor Modulator) was introduced in 1999 in analogy of the term SERM (Selective Estrogen Receptor Modulator) (227). A SARM can be defined as a molecule that targets the androgen receptor, and elicits a biological response, in a tissue-specific way. In a sense, anti-androgens (molecules that specifically target the androgen receptor pathway resulting in inhibition of the biological effects of androgens) can be considered as a special subtype of SARMs. Extensive overviews of current clinical trials with newly developed SARMs by several different pharmaceutical companies have been presented (228-230).

 

The structural basis for SARM binding and activity has been reviewed (138). Based on the conformational changes of the androgen receptor ligand binding domain induced by androgens or anti-androgens, it can be concluded that the different transcriptional activities displayed by either full agonists (testosterone, 5α-dihydrotestosterone, methyltrienolone), partial agonists (RU486 and CPA) or full antagonists (hydroxyflutamide, bicalutamide, enzalutamide) are the result of recruitment of a different repertoire of co-regulators (coactivators or corepressors) as a consequence of these conformational changes. The differential recruitment of co-regulators can be considered as a special form of ligand-selective modulation of the androgen receptor ligand binding domain and can also be applied in a broader sense to the tissue selective modulation of androgen action, where levels of co-activators and co-repressors may ultimately determine the final activity (229-232).

 

TISSUE-SPECIFIC ANDROGEN RECEPTOR MEDIATED ACTIONS IN MOUSE MODELS

 

Genetic mouse models in which the androgen receptor gene has been inactivated (so-called ARKO [androgen receptor knock-out] mouse models) are valuable tools to understand in detail the role of receptor-mediated pathways in male and female reproductive functions. For this purpose several different mouse models have been developed for studying androgen receptor mediated tissue-specific action in almost all known androgen target tissues, although the application of the mouse findings to the human situation has its limitation (233-238). Furthermore, the development of a mouse model for imaging of luciferase activity under control of endogenous androgen receptor activity has contributed to a further elucidation of tissue-specific receptor action (239).

 

ANDROGEN RECEPTOR DISORDERS

 

There is growing evidence for the involvement of the androgen receptor in the gender biases seen in a wide range of pathological conditions, from cancers of non-reproductive tissues (i.e., bladder, liver) (see 240, 241) to cardiovascular and metabolic disease (see 242-244). However, in this review we will focus on receptor mutations leading to defects of male development and fertility.

 

Androgen Insensitivity Syndrome

 

It has been known for quite some time that defects in male sexual differentiation in 46, XY individuals have an X-linked pattern of inheritance. It was Reifenstein who reported in 1947 on families with severe hypospadias, infertility, and gynecomastia (245). The end-organ resistance to androgens has been designated as androgen insensitivity syndrome (AIS) and is distinct from other XY disorders of sex development (XY, DSD; formerly named male pseudohermaphroditism) like 17β-hydroxy-steroid dehydrogenase type 3 deficiency or 5α-reductase type 2 deficiency (3, 246-248). It is generally accepted that defects in the androgen receptor gene can prevent the normal development of both internal and external male structures in 46, XY individuals and information on the molecular structure of the human androgen receptor gene has facilitated the study of molecular defects associated with androgen insensitivity. Due to the X-linked character of the syndrome, only 46, XY individuals are affected, while in female carriers only sporadic reports are available on delayed menarche (249). Naturally occurring mutations in the androgen receptor gene are an interesting source for the investigation of receptor structure-function relationships. In addition, the variation in clinical phenotypes provides the opportunity to correlate a mutation in the androgen receptor structure with the impairment of a specific physiological function.

 

Clinical Features of the Complete Androgen Insensitivity Syndrome (CAIS)

 

The main phenotypic characteristics of individuals with the complete androgen insensitivity syndrome (CAIS) are: female external genitalia, a short, blind ending vagina, absence of Wolffian duct derived structures like epididymides, vasa deferentia, and seminal vesicles, the absence of a prostate, the absence of pubic and axillary hair and the development of gynecomastia (250, 251). Müllerian duct derived structures are usually absent because anti-Mullerian hormone action is normal due to the presence of both testes in the abdomen or in the inguinal canals. Usually, testosterone levels are within the normal range (10 - 40 nmol/L) or elevated, while elevated luteinizing hormone (LH) levels (> 10 IU/L) are also found indicating androgen resistance at the hypothalamic-pituitary level. The high testosterone levels are also substrate for aromatase activity, resulting in substantial amounts of estrogens, which are responsible for further feminization in CAIS individuals.

 

Clinical Features of the Partial Androgen Insensitivity Syndrome (PAIS)

 

In the partial androgen insensitivity syndrome (PAIS) several phenotypes ranging from individuals with predominantly a female appearance (e.g., external female genitalia and pubic hair at puberty, or with mild clitoromegaly, and some fusion of the labia) to persons with ambiguous genitalia or individuals with a predominantly male phenotype (also called Reifenstein syndrome) (250, 251). Patients from this latter group can present with a micropenis, perineal hypospadias, and cryptorchidism. In the group of PAIS individuals, Wolffian duct derived structures can be partially to fully developed, depending on the biochemical phenotype of the androgen receptor mutation. At puberty, elevated luteinizing hormone, testosterone, and estradiol levels are observed, but in general, the degree of feminization is less as compared to individuals with CAIS. Individuals with mild symptoms of undervirilization (mild androgen insensitivity syndrome) and infertility have been described as well. Phenotypic variation between individuals in different families has been described for several mutations (251-254). However, in cases of CAIS no phenotypic variation has been described within one single family, in contrast to families with individuals with PAIS (255).

 

Genetics of Androgen Insensitivity Syndrome (AIS)

 

Since the cloning of the androgen receptor cDNA in 1988 and the subsequent elucidation of the genomic organization of the androgen receptor gene, molecular diagnostic tools have been available for the molecular analysis of the androgen receptor gene in individuals with AIS. In addition to endocrinology data, such as levels of testosterone, luteinizing hormone, androstenedione, and 5α-dihydrotestosterone, which can vary widely in AIS individuals, the most reliable approach is the sequencing of each individual androgen receptor exon and the flanking intron sequences. In general, AIS can be routinely analyzed and separated from entirely different syndromes presenting with similar phenotypes including testicular enzyme deficiencies, 5α-reductase type 2 deficiency, and Leydig cell hypoplasia due to inactivating luteinizing hormone receptor mutations. Furthermore, in pedigree analysis intragenic polymorphisms like the highly polymorphic (CAG)nCAA repeat encoding a poly-glutamine stretch, the polymorphic GGN repeat encoding a poly-glycine stretch, the HindIII polymorphism [Figure 8, see above] (39) and the StuI polymorphism (256), can be used as X-chromosomal markers (67, 257, 258). Extensive general information can be obtained at the internet site, www.genecards.org for the androgen receptor gene (NR3C4) and on the 233 identified single nucleotide polymorphisms (SNP’s).

 

Mutations in the Androgen Receptor Gene

 

In the androgen receptor gene, 4 different types of mutations have been detected in 46, XY individuals with AIS: single point mutations resulting in amino acid substitutions or premature stop codons, nucleotide insertions or deletions most often leading to a frame shift and premature termination, complete or partial gene deletions (>10 nucleotides), and intronic mutations in either splice donor or splice acceptor sites which affect the splicing of androgen receptor RNA (151). In general, in 70% of the cases, androgen receptor gene mutations are transmitted in an X-linked recessive manner, but in 30% the mutations arise de novo. When de novo mutations occur after the zygotic stage, they result in somatic mosaicisms (259). The most recent update on androgen receptor gene mutations is available at http://www.mcgill.ca/androgendb/ (151).

 

MUTATIONS IN THE NH2-TERMINAL DOMAIN

 

Mutations in the NH2-terminal domain (exon 1 of the gene) do not occur frequently and the vast majority of the mutations result directly in a stop codon or in premature termination due to frameshifts caused by nucleotide insertions or deletions. Mutations in 103 different codons have been reported in the NH2-terminal domain, which is approximately 18 % of all codons in exon 1 (http://androgendb.mcgill.ca/ ) (151, 260-264).

 

An interesting mutation is described in the fourth nucleotide, which results in a decreased translational efficiency of the androgen receptor mRNA in an individual with PAIS (265). Three other missense mutations were reported in combination with mosaicism or with a mutation in another region of the gene. In a family with PAIS associated with severe hypospadias, the length of the androgen receptor NH2-terminal poly-glutamine repeat has been reported to be shortened to only 12 glutamine residues (266). The shortened glutamine stretch as such is not the cause for the androgen resistance, but it seems to increase the thermolability of the androgen receptor in combination with a point mutation in exon 5 (Y764C) in the ligand binding domain. This point mutation causes rapid dissociation of hormone, but no thermolability. These data support a functional interaction of the two separated regions in the androgen receptor and indicates further that the defect becomes critical in only some of the androgen target tissues because of the partial character of the androgen resistance found in this family (266).

 

MUTATIONS IN THE DNA-BINDING DOMAIN  

 

In general, mutations in the DNA binding domain (e.g., single nucleotide substitutions) result in a normal hormone-binding protein, which is defective in DNA-binding/dimerization and consequently in transcription activation. In total, 71 different mutations have been reported in 38 different codons in the DNA-binding domain, which is approximately 43% of all codons in exons 2 and 3 (http://androgendb.mcgill.ca/ ) (151, 260, 264, 267, 268). Thirty-four mutations were observed in the first zinc cluster and thirty-two in the second zinc cluster. Since the 3D structure of the DNA-binding domain of several nuclear receptors have been published earlier than that of the androgen receptor DNA-binding domain, the consequence of several mutations in the androgen receptor DNA-binding domain have been predicted initially on basis of the structure of the glucocorticoid receptor DNA-binding domain (122, 123).This is illustrated in two studies in which 3D-modelling of the mutated DNA binding domain of the androgen receptor predicts the functional activity of mutant receptors (269, 270). A mutation (G578R) in the so-called P-box [Figure 9, see above], which is involved in androgen response element recognition, was found in a PAIS individual. This mutation differentially affects transactivation of several natural and synthetic promoters, suggesting that androgen target genes may be differentially affected by this mutation (271). An interesting observation was made with respect to the second zinc cluster in which either one of two adjacent arginine residues (Arg608 & Arg609) were found to be mutated in PAIS individuals who developed breast cancer [Figure 9, see above] (272, 273). It is speculated that a decrease in androgen action within the breast cells could account for the development of male breast cancer by the loss of a protective effect of androgens. However, the same mutations in several other PAIS individuals did not result in breast cancer development.

 

The mutation Ala597Thr in the second zinc cluster in the so-called D-box resulted in abolishment of dimerization in a PAIS individual [Figure 9, see above] (274). A similar mutation at an identical position in the second zinc cluster of the glucocorticoid receptor DNA-binding domain has been created to discriminate between dimerization/DNA binding of the glucocorticoid receptor and protein-protein interactions with other transcription factors such as the AP-1 transcription complex (275). It appeared that the dimerization mutant did not affect the cross-talk with other transcription factors. In this way, a tissue-specific response can be influenced by a single amino acid change and if this is also true for the mutant androgen receptor then the partial phenotype can be explained. Interestingly a Ser580Arg, also located in the D-box can cause significantly different phenotypes ranging from under-virilization to a normal male phenotype (276).

 

MUTATIONS IN THE HINGE REGION  

 

In the so-called hinge region, located between amino acid residues 623 and 671 [Figure 8, see above], only nine mutations have been reported. The relatively low number of mutations in the hinge region (only in 18 % of all codons) indicates that this region might be very flexible and that some variation in composition and length of this region is not detrimental for androgen receptor function (http://www.mcgill.ca/androgendb/) (151).  Four amino acid substitutions within the hinge region have been described that resulted in CAIS, four in PAIS and one in MAIS (http://www.mcgill.ca/androgendb/ (151). The Ile665Asn substitution on the border of the hinge region and ligand-binding domain, resulted in a decreased hormone binding (277).

 

MUTATIONS IN THE LIGAND-BINDING DOMAIN  

 

It can be expected that mutations in the ligand binding domain might affect different functional aspects (e.g., loss of ligand binding, changes in ligand binding affinity and specificity, changes in co-activator receptor interactions, changes in receptor stability and thermolability). A large number of mutations (283 different mutations in 164 codons, which is in 66 % of all codons of the ligand binding domain) in the ligand binding domain have been reported in all 5 exons in individuals with either CAIS, PAIS or MAIS (http://androgendb.mcgill.ca/ ) (151, 260, 265, 278-286). Most mutations are located in exons 4 (62 mutations), 5 (77 mutations) and 7 (54 mutations). Interestingly mutations are found in 13 of the 18 amino acid residues considered to interact with the ligand directly (120). For some mutations (in total 25, distributed over the whole ligand binding domain) either a complete (CAIS) as well as a partial (PAIS) phenotype (13 cases) or a CAIS and a PAIS and a mild (MAIS) phenotype (4 cases) or a PAIS and a MAIS phenotype (8 cases) has been described, indicating that phenotype does not always match with genotype. In the AF-2 core region (894-EMMAEIIS-901) of the androgen receptor ligand-binding domain a relatively low number of mutations have been reported [see Figure 10 for location of AF-2]. At positions methionine 895 (deletion), Met896, Ala897, Glu898 and Ile899 (all substitutions) have been described in individuals with the complete syndrome (287, 288). It can be speculated that in this part of helix 12 mutations in the androgen receptor ligand-binding domain are very deleterious for androgen receptor function as well as those in helix 5 and in the β-turn, wherein almost every amino acid residue has been found to be mutated in AIS individuals (http://androgendb.mcgill.ca/ ) (151). Functional analysis of an androgen receptor mutation, Gln903Lys in helix 12, in an individual with partial androgen insensitivity, indicated that this residue is important for modulation of NH2/COOH terminal interaction and TIF-2 activation (289). Interestingly a mutation, Phe827Leu, found in a PAIS patient, displayed an unexpected increased N/C interaction and TIF2 coactivation (290). An explanation for the phenotype of the patient could be that the receptor mutant recruits a different repertoire of co-activators absent in genital tissues. Alternatively, an altered conformation of the ligand binding domain may enhance preferential recruitment of co-repressors.

 

Several reports have established the pathogenic nature of androgen receptor mutations found in AIS individuals with different functional assays (260, 289-292). In order to optimize molecular diagnosis an extensive functional analysis of receptor mutations is desired. For counselling strategies and for future outcome predictions a correct functional diagnosis is very important as well as for prognosis on the risks of gonadal malignancy (293). A combination of different functional analyses, designed to test androgen receptor mutations at different stages in receptor functioning (e.g., hormone binding, transcriptional activation, cofactor binding, translocation to the nucleus and nuclear dynamics) will provide a more accurate prediction of androgen receptor action and will help to establish a more exact phenotypic characterization.

 

DELETIONS AND DUPLICATIONS OF THE ANDROGEN RECEPTOR GENE  

 

Only a few cases (8 different deletions in 15 different patients) have been reported on partial or complete androgen receptor gene deletions, indicating the relatively low frequency of this type of androgen receptor defect (http://androgendb.mcgill.ca/) (151, 294). All cases reported are found in CAIS individuals, with the exception of two cases, one in which an exon 4 deletion was found in a person with azoospermia (295) and another one in which a large intron 2 deletion of at least 6 kb was reported involving a branch point site, which resulted in a partial exon 3 skipping during the splicing process (294).

 

Deletion of either exon 3 or exon 4 occur both in-frame and result in a non-functional protein lacking either the second zinc cluster or the hinge region and the NH2-terminal part of the ligand-binding domain [see Figure 7 for genomic organization of the androgen receptor gene]. In case of an exon 3 deletion, an intact and functional ligand-binding domain is present [Figure 7]. So far, functionally significant mutations in the androgen receptor promoter region or in the 5'- and 3'- untranslated regions of the gene have not been reported.

 

SPLICE SITE MUTATIONS AFFECTING ANDROGEN RECEPTOR RNA SPLICING  

 

A special group of interesting, but rare, mutations are the splice donor and splice acceptor site mutations in the androgen receptor gene in AIS individuals (http://androgendb.mcgill.ca/ ) (151). For all splice donor sites in the gene, the consensus splice donor site sequence GTAAG/A is present. The twelve reported mutations in donor splice sites are all substitutions either at position +1 (G  A or G  T), position +2 (T  C), position +3 (A  T), position + 4 (AT) or position + 5 (G  A) and result in defective splicing with the consequence of one or more exons spliced out, or the use of a cryptic splice donor site within the preceding exon (264, 296-301). In 11 of the reported cases, the phenotype is complete androgen insensitivity. In one case, an insertion of one nucleotide (T) at position + 4 in the splice donor site of intron 6 has been reported, resulting in a partial androgen insensitive phenotype (300). Only 5 mutations have been reported in splice acceptor sites, which all affect the splicing of the androgen receptor RNA. Interestingly, a substitution at position -11 (T G) has been found in the pyrimidine-rich region of the splice acceptor site of intron 2, resulting in the activation of a cryptic splice acceptor site at position -70/-69 and consequently in the insertion of 69 nucleotides (corresponding to 23 additional amino acid residues) in the mRNA between exons 2 and 3 (302). The corresponding protein is defective in DNA-binding because the insertion has occurred between the first and second zinc cluster. In another CAIS patient a splice junction mutation at the intron2/exon3 splice acceptor site resulted in the utilization of the same cryptic splice acceptor site and also in the insertion of 69 bp in the mRNA, predicting the insertion of 23 amino acid residues in frame between the two zinc clusters (303).

 

Androgen Receptor Gene Mutations in Cancers

 

Mutations in the androgen receptor gene have also been reported to be associated with prostate cancers, breast cancers, larynx cancers, liver cancers and testicular cancers (http://androgendb.mcgill.ca/ ) (151).

 

ANDROGEN METABOLISM DISORDERS

 

The metabolism of testosterone to 5α-dihydrotestosterone by the enzyme 5α-reductase type 2 (SRD5A2) is essential for the initiation of the differentiation and development of the urogenital sinus into the prostate. The differentiation of the male external genitalia (penis, scrotum and urethra) also strongly depends on the conversion of testosterone to 5α-dihydrotestosterone in the urogenital tubercle, labioscrotal swellings and urogenital folds, respectively [Figure 2B, see above] (3, 4). Interestingly in the SPARKI mouse expression of Srd5α2 gene is significantly impaired in the epididymis and the androgen-regulation of the gene was demonstrated to involve three selective AREs (304). 

 

Clinical Features of the Syndrome of 5α-reductase Type 2 Deficiency

 

46, XY individuals with impairment of 5α-reductase type 2 have normally virilized Wolffian duct derived structures, with seminal vesicles (although small seminal vesicles have been reported as well), with vasa deferentia, epididymides and ejaculatory ducts and no Mullerian duct derived structures (3, 305, 306). However, differentiation of the urogenital sinus and genital tubercle is not observed, resulting in absence of the prostate and in ambiguous or in female external genitalia at birth (3, 305, 306). Affected 46, XY individuals are therefore often raised as girls. At puberty all affected individuals show some or a severe degree of virilization often resulting in deepening of the voice, an increased muscle mass, growth of the penis, scrotal development, testicular descent and sometimes leading to a gender change (3, 307).

 

Gynecomastia in adulthood does not occur. The additional virilization may result from the action of testosterone because testosterone is available at high levels during puberty. In addition, some testosterone may be converted to 5α-dihydrotestosterone by some residual 5α-reductase activity and by the action of 5α-reductase type 1, which is expressed in non-genital skin, pubic skin, liver and certain brain regions. In the affected 46, XY individuals a typical female pubic hair pattern develops, while the facial and body hair amount is absent or reduced (4). This last observation points to a role for 5α-reductase type 2 in the normal development of this type of body hair. Male pattern baldness has never been observed. 5α-reductase type 2 deficient patients are usually infertile due to the absence or underdevelopment of the prostate and seminal vesicles, in addition to oligospermia or azoospermia due to maldescent of the testes. However, paternity has been reported in some cases, either by intrauterine insemination or after in vitro fertilization in combination with intracytoplasmic sperm injection (3, 305, 308-310). 46, XX female carriers have normal fertility, decreased body hair and delayed menarche, normal sebum production but no history of acne (3, 305). This suggests a role of 5α-reductase type 2 enzyme in females in the physiology and pathophysiology of body hair growth, menarche and follicular development (305).

 

Molecular Basis for the Syndrome of 5α-Reductase Type 2 Deficiency

 

A reflection of defective or absence 5α-reductase type 2 enzyme activity can be obtained in patients’ serum and urine samples by measuring testosterone levels (elevated), 5α-dihydrotestosterone levels (decreased) and by measuring the ratio of testosterone/5α-dihydrotestosterone (increased after hCG stimulation) (3). Furthermore, in cultured genital skin fibroblasts (if available) the conversion of testosterone to 5α-dihydrotestosterone can be assessed and is an option for establishing a defective enzyme. In broken cell preparations at pH 5.5, the type 2 isozyme activity is measured more specifically and can be compared with a preparation from a normal person (3).

 

Genetic analysis of 5α-reductase type 2 deficiency has become possible since the cloning of the cDNA (17). The gene is located on chromosome 2 at locus 2p23. The enzyme is encoded by 5 exons and the most reliable approach to detect gene mutations is the sequencing of each individual exon and the flanking intron sequences [Figure 12]. A relatively large number of loss of function mutations in the type 2 steroid 5α-reductase has been identified in 46XY individuals with this rare autosomal recessive disorder of sex development (46XY, DSD).

 

Interestingly worldwide 87 different mutations have been detected in the 5α-reductase type 2 gene in patients with the syndrome of 5α-reductase type 2 deficiency in several different ethnic groups [Figure 12] (3, 4, 285, 305-307, 311-339). Identical mutations have been reported in different ethnic groups and some of them can be considered to be due to a founder effect and some to have occurred de novo (340-342). The mutations were found in all five exons of the gene, although the majority of the mutations are reported in exons 1 and 4 [Figure 12].

 

The mutations comprise of 57 amino acid substitutions (65.5%), one complete gene deletion (3, 306), one complete exon 1 deletion (16), one substitution at stop codon 255 resulting in a Serine residue (336), ten small deletions resulting in either a premature stop codon or in an in-frame amino acid residue deletion, four small insertions (335), nine nonsense mutations and four splice site mutations, resulting in aberrant splicing [Figure 12]. The majority of the reported patients are homozygous for one of the mutations. A smaller number of patients appeared to be compound heterozygous, while a small group of patients are heterozygous (331, 340, 341).

 

In general male carriers of a single mutant allele have normal fertility as is the case for female carriers. The largest investigated kindreds were found in the Dominican Republic, in Turkey and in New Guinea (3, 305, 333). In all three kindreds the high incidence can be directly related to a founder affect in geographical isolated populations with a high degree of inbreeding. For other cases also a large incidence of proven consanguinity is reported (3, 305).

 

In prostate cancer de novo mutations in the 5α-reductase type 2 have been reported, resulting in increased 5α-reductase activity (317, 333, 343, 344).  This finding indicates a role for increased 5α-dihydrotestosterone levels in the prostate, during prostate cancer progression in a subset of patients. The V89L mutant significantly reduced SRD5A2 enzymatic activity by almost 30% (316, 342, 343). The rare allele frequency of the V89L variant is 22%, 23,5%, and 46,1% for African Americans, Caucasians, and Asians, respectively, paralleling a substantial racial/ethnic variation in prostate cancer risk, indicating that this polymorphism might be implicated in prostate cancer carcinogenesis (343-346).

 

CONCLUSIONS-KEY POINTS

 

Androgenic steroids are important for normal development and function of male reproductive tissues and for anabolic actions in muscle and bone. The multiple actions of the main circulating androgen testosterone and the more potent metabolite DHT are mediated by a single intracellular receptor protein, the androgen receptor. The hormone-bound receptor acts primarily to differentially regulate gene expression in target tissues and its encoding gene is located on the X chromosome, making it a single-copy gene in males. Thus, genetic changes affecting expression or structure/function of the receptor protein will lead to a range of diseases associated with loss or impaired androgen signaling, including disruption of male development, infertility or a late onset neurodegenerative disease (SBMA). Furthermore, altered expression and genetic changes in the receptor are also key drivers in progression of prostate cancer to a therapy-resistant stage.

 

Since the first cloning of the androgen receptor cDNA, over thirty years ago, considerable progress has been made in our understanding of receptor structure and function. Advances include: the availability of 3D-structures of the isolated LBD with different ligands bound and the isolated DBD; structural characterization of the intrinsically disordered NH2-terminal domain; first glimpse of the structure of full-length AR transcriptional complex on DNA; the identification of a plethora of co-regulatory proteins binding to the ligand- and NH2-terminal domains; identification of gene regulatory pathways in target cells; and a better understanding of the impact of genetic changes affecting receptor structure/function. Future research will likely focus on the mechanisms determining cell/tissue-selective expression and function of the androgen receptor in both normal and pathophysiological conditions and a more complete structural descriptions of the full-length receptor bound to different DNA response elements and co-regulatory proteins.

 

REFERENCES

 

  1. George FW, Wilson JD 1994 Sex determination and differentiation. In: Knobil E, Neill JD, eds. The Physiology of Reproduction. New York.: Raven Press Ltd.; Chapter 1
  2. Brinkmann AO 2011 Molecular mechanisms of androgen action--a historical perspective. Methods Mol Biol 776:3-24
  3. Wilson JD, Griffin JE, Russell DW 1993 Steroid 5 alpha-reductase 2 deficiency. Endocr Rev 14(5):577-593
  4. Randall VA 1994 Role of 5 alpha-reductase in health and disease. Baillieres Clin Endocrinol Metab 8(2):405-431
  5. Grino PB, Griffin JE, Wilson JD 1990 Testosterone at high concentrations interacts with the human androgen receptor similarly to dihydrotestosterone. Endocrinology 126(2):1165-1172
  6. El-Gehani F, Zhang FP, Pakarinen P, Rannikko A, Huhtaniemi I 1998 Gonadotropin-independent regulation of steroidogenesis in the fetal rat testis. Biol Reprod 58(1):116-123
  7. Brinkmann AO, van Straalen RJ 1979 Development of the LH-response in fetal guinea pig testes. Biol Reprod 21(4):991-997
  8. O'Shaughnessy PJ, Baker P, Sohnius U, Haavisto AM, Charlton HM, Huhtaniemi I 1998 Fetal development of leydig cell activity in the mouse is independent of pituitary gonadotroph function. Endocrinology 139(3):1141-1146
  9. Curtin D, Jenkins S, Farmer N, Anderson AC, Haisenleder DJ, Rissman E, Wilson EM, Shupnik MA 2001 Androgen suppression of GnRH-stimulated rat LHbeta gene transcription occurs through Sp1 sites in the distal GnRH-responsive promoter region. Mol Endocrinol 15(11):1906-1917
  10. Stocco DM, Clark BJ 1996 Regulation of the acute production of steroids in steroidogenic cells. Endocr Rev 17(3):221-244
  11. Miller WL 1988 Molecular biology of steroid hormone synthesis. Endocr Rev 9(3):295-318
  12. Fukami M, Homma K, Hasegawa T, Ogata T. 2013 Backdoor pathway for dihydrotestosterone biosynthesis: implications for normal and abnormal human sex development. Dev Dyn.242(4):320-9.
  13. O'Shaughnessy PJ, Antignac JP, Le Bizec B, Morvan ML, Svechnikov K, Söder O, Savchuk I, Monteiro A, Soffientini U, Johnston ZC, Bellingham M, Hough D, Walker N, Filis P, Fowler PA. 2019 Alternative (backdoor) androgen production and masculinization in the human fetus. PLoS Biol. 17(2):e3000002.
  14. Reisch N, Taylor AE, Nogueira EF, Asby DJ, Dhir V, Berry A, Krone N, Auchus RJ, Shackleton CHL, Hanley NA, Arlt W. 2019 Alternative pathway androgen biosynthesis and human fetal female virilization. Proc Natl Acad Sci U S A. 116(44):22294-22299.
  15. Russell DW, Wilson JD 1994 Steroid 5 alpha-reductase: Two genes/two enzymes. Annu Rev Biochem 63:25-61
  16. Andersson S, Berman DM, Jenkins EP, Russell DW 1991 Deletion of steroid 5 alpha-reductase 2 gene in male pseudohermaphroditism. Nature 354(6349):159-161
  17. Andersson S, Bishop RW, Russell DW 1989 Expression cloning and regulation of steroid 5 alpha-reductase, an enzyme essential for male sexual differentiation. J Biol Chem 264(27):16249-16255
  18. Azzouni F, Godoy A, Li Y, Mohler J 2012 The 5 alpha-reductase isozyme family: A review of basic biology and their role in human diseases. Adv Urol 2012:530121
  19. Stiles AR, Russell DW 2010 SRD5A3: A surprising role in glycosylation. Cell 142(2):196-198
  20. Cantagrel V, Lefeber DJ, Ng BG, Guan Z, Silhavy JL, Bielas SL, Lehle L, Hombauer H, Adamowicz M, Swiezewska E, De Brouwer AP, Blumel P, Sykut-Cegielska J, Houliston S, Swistun D, Ali BR, Dobyns WB, Babovic-Vuksanovic D, van Bokhoven H, Wevers RA, Raetz CR, Freeze HH, Morava E, Al-Gazali L, Gleeson JG 2010 SRD5A3 is required for converting polyprenol to dolichol and is mutated in a congenital glycosylation disorder. Cell 142(2):203-217
  21. Moon YA, Horton JD 2003 Identification of two mammalian reductases involved in the two-carbon fatty acyl elongation cascade. J Biol Chem 278(9):7335-7343
  22. Nuclear Receptors Nomenclature Committee 1999 A unified nomenclature system for the nuclear receptor superfamily. Cell 97(2):161-163
  23. Evans RM 1988 The steroid and thyroid hormone receptor superfamily. Science 240(4854):889-895
  24. Laudet V, Hanni C, Coll J, Catzeflis F, Stehelin D 1992 Evolution of the nuclear receptor gene superfamily. EMBO J 11(3):1003-1013
  25. Robinson-Rechavi M, Carpentier AS, Duffraisse M, Laudet V 2001 How many nuclear hormone receptors are there in the human genome? Trends Genet 17(10):554-556
  26. Enmark E, Gustafsson JA 1996 Orphan nuclear receptors--the first eight years. Mol Endocrinol 10(11):1293-1307
  27. Hutchison KA, Dittmar KD, Pratt WB 1994 All of the factors required for assembly of the glucocorticoid receptor into a functional heterocomplex with heat shock protein 90 are preassociated in a self-sufficient protein folding structure, a "foldosome". J Biol Chem 269(45):27894-27899
  28. Cano LQ, Lavery DN, Bevan CL 2013 Mini-review: Foldosome regulation of androgen receptor action in prostate cancer. Mol Cell Endocrinol 369(1-2):52-62
  29. Heemers HV, Tindall DJ 2007 Androgen receptor (AR) coregulators: A diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev 28(7):778-808
  30. Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, Chen Z, Beroukhim R, Wang H, Lupien M, Wu T, Regan MM, Meyer CA, Carroll JS, Manrai AK, Janne OA, Balk SP, Mehra R, Han B, Chinnaiyan AM, Rubin MA, True L, Fiorentino M, Fiore C, Loda M, Kantoff PW, Liu XS, Brown M 2009 Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138(2):245-256
  31. Migliaccio A, Castoria G, Di Domenico M, de Falco A, Bilancio A, Lombardi M, Barone MV, Ametrano D, Zannini MS, Abbondanza C, Auricchio F 2000 Steroid-induced androgen receptor-oestradiol receptor beta-src complex triggers prostate cancer cell proliferation. EMBO J 19(20):5406-5417
  32. Kousteni S, Bellido T, Plotkin LI, O'Brien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC 2001 Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: Dissociation from transcriptional activity. Cell 104(5):719-730
  33. Lutz LB, Cole LM, Gupta MK, Kwist KW, Auchus RJ, Hammes SR 2001 Evidence that androgens are the primary steroids produced by xenopus laevis ovaries and may signal through the classical androgen receptor to promote oocyte maturation. Proc Natl Acad Sci U S A 98(24):13728-13733
  34. Sen A, Prizant H, Hammes SR 2011 Understanding extranuclear (nongenomic) androgen signaling: What a frog oocyte can tell us about human biology. Steroids 76(9):822-828
  35. Chang CS, Kokontis J, Liao ST 1988 Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science 240(4850):324-326
  36. Lubahn DB, Joseph DR, Sar M, Tan J, Higgs HN, Larson RE, French FS, Wilson EM 1988 The human androgen receptor: Complementary deoxyribonucleic acid cloning, sequence analysis and gene expression in prostate. Mol Endocrinol 2(12):1265-1275
  37. Trapman J, Klaassen P, Kuiper GG, van der Korput JA, Faber PW, van Rooij HC, Geurts van Kessel A, Voorhorst MM, Mulder E, Brinkmann AO 1988 Cloning, structure and expression of a cDNA encoding the human androgen receptor. Biochem Biophys Res Commun 153(1):241-248
  38. Tilley WD, Marcelli M, Wilson JD, McPhaul MJ 1989 Characterization and expression of a cDNA encoding the human androgen receptor. Proc Natl Acad Sci U S A 86(1):327-331
  39. Brown CJ, Goss SJ, Lubahn DB, Joseph DR, Wilson EM, French FS, Willard HF 1989 Androgen receptor locus on the human X chromosome: Regional localization to Xq11-12 and description of a DNA polymorphism. Am J Hum Genet 44(2):264-269
  40. van Laar JH, Bolt-de Vries J, Voorhorst-Ogink MM, Brinkmann AO 1989 The human androgen receptor is a 110 kDa protein. Mol Cell Endocrinol 63(1-2):39-44
  41. Kuiper GG, Faber PW, van Rooij HC, van der Korput JA, Ris-Stalpers C, Klaassen P, Trapman J, Brinkmann AO 1989 Structural organization of the human androgen receptor gene. J Mol Endocrinol 2(3):R1-4
  42. Lubahn DB, Brown TR, Simental JA, Higgs HN, Migeon CJ, Wilson EM, French FS 1989 Sequence of the intron/exon junctions of the coding region of the human androgen receptor gene and identification of a point mutation in a family with complete androgen insensitivity. Proc Natl Acad Sci U S A 86(23):9534-9538
  43. Gaspar ML, Meo T, Tosi M. 1990 Structure and size distribution of the androgen receptor mRNA in wild-type and Tfm/Y mutant mice. Mol Endocrinol. 4(10):1600-10.
  44. Ebron JS, Shukla GC 2016 Molecular characterization of a novel androgen receptor transgene responsive to MicroRNA mediated post-transcriptional control exerted via 3'-untranslated region. Prostate. 76(9):834-44.
  45. Östling P, Leivonen SK, Aakula A, Kohonen P, Mäkelä R, Hagman Z, Edsjö A, Kangaspeska S, Edgren H, Nicorici D, Bjartell A, Ceder Y, Perälä M, Kallioniemi O 2011 Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Res. 71(5):1956-67.
  46. Liu C, Chen Z, Hu X, Wang L, Li C, Xue J, Zhang P, Chen W, Jiang A2015 MicroRNA-185 downregulates androgen receptor expression in the LNCaP prostate carcinoma cell line.Mol Med Rep. 11(6): 4625-32.
  47. Faber PW, King A, van Rooij HC, Brinkmann AO, de Both NJ, Trapman J 1991 The mouse androgen receptor. functional analysis of the protein and characterization of the gene. Biochem J 278 ( Pt 1)(Pt 1):269-278
  48. Faber PW, van Rooij HC, van der Korput HA, Baarends WM, Brinkmann AO, Grootegoed JA, Trapman J 1991 Characterization of the human androgen receptor transcription unit. J Biol Chem 266(17):10743-10749
  49. Faber PW, van Rooij HC, Schipper HJ, Brinkmann AO, Trapman J 1993 Two different, overlapping pathways of transcription initiation are active on the TATA-less human androgen receptor promoter. the role of Sp1. J Biol Chem 268(13):9296-9301
  50. Takane KK, McPhaul MJ 1996 Functional analysis of the human androgen receptor promoter. Mol Cell Endocrinol 119(1):83-93
  51. Wang LG, Ferrari AC 2006 Mithramycin targets sp1 and the androgen receptor transcription level-potential therapeutic role in advanced prostate cancer. Transl Oncogenomics 1:19-31
  52. Hay CW, Hunter I, MacKenzie A, McEwan IJ 2015 An Sp1 modulated regulatory region unique to higher primates regulates human androgen receptor promoter activity in prostate cancer cells. PLoS One 10(10):e0139990
  53. Jarrard DF, Kinoshita H, Shi Y, Sandefur C, Hoff D, Meisner LF, Chang C, Herman JG, Isaacs WB, Nassif N.1998 Methylation of the androgen receptor promoter CpG island is associated with loss of androgen receptor expression in prostate cancer cells.Cancer Res. 58(23): 5310-4.
  54. Kinoshita H, Shi Y, Sandefur C, Meisner LF, Chang C, Choon A, Reznikoff CR, Bova GS, Friedl A, Jarrard DF.2000 Methylation of the androgen receptor minimal promoter silences transcription in human prostate cancer.Cancer Res. 60(13):3623-30
  55. Burnstein KL 2005 Regulation of androgen receptor levels: Implications for prostate cancer progression and therapy. J Cell Biochem 95(4):657-669
  56. Cai C, Chen S, Ng P, Bubley GJ, Nelson PS, Mostaghel EA, Marck B, Matsumoto AM, Simon NI, Wang H, Chen S, Balk SP 2011 Intratumoral de novo steroid synthesis activates androgen receptor in castration-resistant prostate cancer and is upregulated by treatment with CYP17A1 inhibitors. Cancer Res 71(20):6503-6513
  57. Hay CW, Watt K, Hunter I, Lavery DN, MacKenzie A, McEwan IJ 2014 Negative regulation of the androgen receptor gene through a primate-specific androgen response element present in the 5' UTR. Horm Cancer 5(5):299-311
  58. Hunter I, Hay CW, Esswein B, Watt K, McEwan IJ. 2018 Tissue control of androgen action: The ups and downs of androgen receptor expression. Mol Cell Endocrinol. 465:27-35.
  59. Huang P, Chandra V, Rastinejad F 2010 Structural overview of the nuclear receptor superfamily: Insights into physiology and therapeutics. Annu Rev Physiol 72:247-272
  60. Hollenberg SM, Weinberger C, Ong ES, Cerelli G, Oro A, Lebo R, Thompson EB, Rosenfeld MG, Evans RM 1985 Primary structure and expression of a functional human glucocorticoid receptor cDNA. Nature 318(6047):635-641
  61. Green S, Walter P, Kumar V, Krust A, Bornert JM, Argos P, Chambon P 1986 Human oestrogen receptor cDNA: Sequence, expression and homology to v-erb-A. Nature 320(6058):134-139
  62. Misrahi M, Atger M, d'Auriol L, Loosfelt H, Meriel C, Fridlansky F, Guiochon-Mantel A, Galibert F, Milgrom E 1987 Complete amino acid sequence of the human progesterone receptor deduced from cloned cDNA. Biochem Biophys Res Commun 143(2):740-748
  63. Arriza JL, Weinberger C, Cerelli G, Glaser TM, Handelin BL, Housman DE, Evans RM 1987 Cloning of human mineralocorticoid receptor complementary DNA: Structural and functional kinship with the glucocorticoid receptor. Science 237(4812):268-275
  64. Faber PW, Kuiper GG, van Rooij HC, van der Korput JA, Brinkmann AO, Trapman J 1989 The N-terminal domain of the human androgen receptor is encoded by one, large exon. Mol Cell Endocrinol 61(2):257-262
  65. Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA 1996 Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci U S A 93(12):5925-5930
  66. Sleddens HF, Oostra BA, Brinkmann AO, Trapman J 1992 Trinucleotide repeat polymorphism in the androgen receptor gene (AR). Nucleic Acids Res 20(6):1427
  67. Boehmer AL, Brinkmann AO, Niermeijer MF, Bakker L, Halley DJ, Drop SL 1997 Germ-line and somatic mosaicism in the androgen insensitivity syndrome: Implications for genetic counseling. Am J Hum Genet 60(4):1003-1006
  68. Li SL, Ting SS, Lindeman R, Ffrench R, Ziegler JB 1998 Carrier identification in X-linked immunodeficiency diseases. J Paediatr Child Health 34(3):273-279
  69. Nance MA 1997 Clinical aspects of CAG repeat diseases. Brain Pathol 7(3):881-900
  70. Jenster G, de Ruiter PE, van der Korput HA, Kuiper GG, Trapman J, Brinkmann AO 1994 Changes in the abundance of androgen receptor isotypes: Effects of ligand treatment, glutamine-stretch variation, and mutation of putative phosphorylation sites. Biochemistry 33(47):14064-14072
  71. Kazemi-Esfarjani P, Trifiro MA, Pinsky L 1995 Evidence for a repressive function of the long polyglutamine tract in the human androgen receptor: Possible pathogenetic relevance for the (CAG)n-expanded neuronopathies. Hum Mol Genet 4(4):523-527
  72. Hardy DO, Scher HI, Bogenreider T, Sabbatini P, Zhang ZF, Nanus DM, Catterall JF 1996 Androgen receptor CAG repeat lengths in prostate cancer: Correlation with age of onset. J Clin Endocrinol Metab 81(12):4400-4405
  73. Giovannucci E, Stampfer MJ, Krithivas K, Brown M, Dahl D, Brufsky A, Talcott J, Hennekens CH, Kantoff PW 1997 The CAG repeat within the androgen receptor gene and its relationship to prostate cancer. Proc Natl Acad Sci U S A 94(7):3320-3323
  74. Cude KJ, Montgomery JS, Price DK, Dixon SC, Kincaid RL, Kovacs KF, Venzon DJ, Liewehr DJ, Johnson ME, Reed E, Figg WD 2002 The role of an androgen receptor polymorphism in the clinical outcome of patients with metastatic prostate cancer. Urol Int 68(1):16-23
  75. Correa-Cerro L, Wohr G, Haussler J, Berthon P, Drelon E, Mangin P, Fournier G, Cussenot O, Kraus P, Just W, Paiss T, Cantu JM, Vogel W 1999 (CAG)nCAA and GGN repeats in the human androgen receptor gene are not associated with prostate cancer in a french-german population. Eur J Hum Genet 7(3):357-362
  76. Freedman ML, Pearce CL, Penney KL, Hirschhorn JN, Kolonel LN, Henderson BE, Altshuler D 2005 Systematic evaluation of genetic variation at the androgen receptor locus and risk of prostate cancer in a multiethnic cohort study. Am J Hum Genet 76(1):82-90
  77. Dowsing AT, Yong EL, Clark M, McLachlan RI, de Kretser DM, Trounson AO 1999 Linkage between male infertility and trinucleotide repeat expansion in the androgen-receptor gene. Lancet 354(9179):640-643
  78. Mifsud A, Sim CK, Boettger-Tong H, Moreira S, Lamb DJ, Lipshultz LI, Yong EL 2001 Trinucleotide (CAG) repeat polymorphisms in the androgen receptor gene: Molecular markers of risk for male infertility. Fertil Steril 75(2):275-281
  79. Wallerand H, Remy-Martin A, Chabannes E, Bermont L, Adessi GL, Bittard H 2001 Relationship between expansion of the CAG repeat in exon 1 of the androgen receptor gene and idiopathic male infertility. Fertil Steril 76(4):769-774
  80. La Spada AR, Wilson EM, Lubahn DB, Harding AE, Fischbeck KH 1991 Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy. Nature 352(6330):77-79
  81. Orafidya FO, McEwan IJ 2015 Trinucleotide repeats and protein folding and disease: The perspective from studies with the androgen receptor. Future Science OA 1:DOI 10.4155
  82. Kennedy WR, Alter M, Sung JH 1968 Progressive proximal spinal and bulbar muscular atrophy of late onset. A sex-linked recessive trait. Neurology 18(7):671-680
  83. Robitaille Y, Lopes-Cendes I, Becher M, Rouleau G, Clark AW 1997 The neuropathology of CAG repeat diseases: Review and update of genetic and molecular features. Brain Pathol 7(3):901-926
  84. Young JE, Garden GA, Martinez RA, Tanaka F, Sandoval CM, Smith AC, Sopher BL, Lin A, Fischbeck KH, Ellerby LM, Morrison RS, Taylor JP, La Spada AR 2009 Polyglutamine-expanded androgen receptor truncation fragments activate a bax-dependent apoptotic cascade mediated by DP5/Hrk. J Neurosci 29(7):1987-1997
  85. Beitel LK, Alvarado C, Mokhtar S, Paliouras M, Trifiro M 2013 Mechanisms mediating spinal and bulbar muscular atrophy: Investigations into polyglutamine-expanded androgen receptor function and dysfunction. Front Neurol 4:53
  86. Adachi H, Waza M, Tokui K, Katsuno M, Minamiyama M, Tanaka F, Doyu M, Sobue G 2007 CHIP overexpression reduces mutant androgen receptor protein and ameliorates phenotypes of the spinal and bulbar muscular atrophy transgenic mouse model. J Neurosci 27(19):5115-5126
  87. Yang Z, Chang YJ, Yu IC, Yeh S, Wu CC, Miyamoto H, Merry DE, Sobue G, Chen LM, Chang SS, Chang C 2007 ASC-J9 ameliorates spinal and bulbar muscular atrophy phenotype via degradation of androgen receptor. Nat Med 13(3):348-353
  88. Banno H, Katsuno M, Suzuki K, Tanaka F, Sobue G 2012 Pathogenesis and molecular targeted therapy of spinal and bulbar muscular atrophy (SBMA). Cell Tissue Res 349(1):313-320
  89. Fernandez-Rhodes LE, Kokkinis AD, White MJ, Watts CA, Auh S, Jeffries NO, Shrader JA, Lehky TJ, Li L, Ryder JE, Levy EW, Solomon BI, Harris-Love MO, La Pean A, Schindler AB, Chen C, Di Prospero NA, Fischbeck KH 2011 Efficacy and safety of dutasteride in patients with spinal and bulbar muscular atrophy: A randomised placebo-controlled trial. Lancet Neurol 10(2):140-147
  90. Kosaka T, Miyajima A, Kikuchi E, Takahashi S, Suzuki N, Oya M 2012 A case of spinal and bulbar muscular atrophy with high-stage and high-gleason score prostate cancer responded to maximal androgen blockade therapy. J Androl 33(4):563-565
  91. Orr CR, Montie HL, Liu Y, Bolzoni E, Jenkins SC, Wilson EM, Joseph JD, McDonnell DP, Merry DE 2010 An interdomain interaction of the androgen receptor is required for its aggregation and toxicity in spinal and bulbar muscular atrophy. J Biol Chem 285(46):35567-35577
  92. LaFevre-Bernt MA, Ellerby LM 2003 Kennedy's disease. phosphorylation of the polyglutamine-expanded form of androgen receptor regulates its cleavage by caspase-3 and enhances cell death. J Biol Chem 278(37):34918-34924
  93. Palazzolo I, Burnett BG, Young JE, Brenne PL, La Spada AR, Fischbeck KH, Howell BW, Pennuto M 2007 Akt blocks ligand binding and protects against expanded polyglutamine androgen receptor toxicity. Hum Mol Genet 16(13):1593-1603
  94. Scaramuzzino C, Casci I, Parodi S, Lievens PM, Polanco MJ, Milioto C, Chivet M, Monaghan J, Mishra A, Badders N, Aggarwal T, Grunseich C, Sambataro F, Basso M, Fackelmayer FO, Taylor JP, Pandey UB, Pennuto M 2015 Protein arginine methyltransferase 6 enhances polyglutamine-expanded androgen receptor function and toxicity in spinal and bulbar muscular atrophy. Neuron 85(1):88-100
  95. Chua JP, Reddy SL, Yu Z, Giorgetti E, Montie HL, Mukherjee S, Higgins J, McEachin RC, Robins DM, Merry DE, Iniguez-Lluhi JA, Lieberman AP 2015 Disrupting SUMOylation enhances transcriptional function and ameliorates polyglutamine androgen receptor-mediated disease. J Clin Invest 125(2):831-845
  96. Qiang Q, Adachi H, Huang Z, Jiang YM, Katsuno M, Minamiyama M, Doi H, Matsumoto S, Kondo N, Miyazaki Y, Iida M, Tohnai G, Sobue G 2013 Genistein, a natural product derived from soybeans, ameliorates polyglutamine-mediated motor neuron disease. J Neurochem 126(1):122-130
  97. Ranganathan S, Fischbeck KH 2010 Therapeutic approaches to spinal and bulbar muscular atrophy. Trends Pharmacol Sci 31(11):523-527
  98. Rocchi A, Pennuto M 2013 New routes to therapy for spinal and bulbar muscular atrophy. J Mol Neurosci 50(3):514-523
  99. Lavery DN, McEwan IJ 2008 Structural characterization of the native NH2-terminal transactivation domain of the human androgen receptor: A collapsed disordered conformation underlies structural plasticity and protein-induced folding. Biochemistry 47(11):3360-3369
  100. Kumar R, Thompson EB 2003 Transactivation functions of the N-terminal domains of nuclear hormone receptors: Protein folding and coactivator interactions. Mol Endocrinol 17(1):1-10
  101. McEwan IJ 2012 Intrinsic disorder in the androgen receptor: Identification, characterisation and drugability. Mol Biosyst 8(1):82-90
  102. Kumar R, McEwan IJ 2012 Allosteric modulators of steroid hormone receptors: Structural dynamics and gene regulation. Endocr Rev 33(2):271-299
  103. Jenster G, van der Korput HA, Trapman J, Brinkmann AO 1995 Identification of two transcription activation units in the N-terminal domain of the human androgen receptor. J Biol Chem 270(13):7341-7346
  104. Christiaens V, Bevan CL, Callewaert L, Haelens A, Verrijdt G, Rombauts W, Claessens F 2002 Characterization of the two coactivator-interacting surfaces of the androgen receptor and their relative role in transcriptional control. J Biol Chem 277(51):49230-49237
  105. Claessens F, Denayer S, Van Tilborgh N, Kerkhofs S, Helsen C, Haelens A 2008 Diverse roles of androgen receptor (AR) domains in AR-mediated signaling. Nucl Recept Signal 6:e008
  106. Reid J, Murray I, Watt K, Betney R, McEwan IJ 2002 The androgen receptor interacts with multiple regions of the large subunit of general transcription factor TFIIF. J Biol Chem 277(43):41247-41253
  107. Lavery DN, McEwan IJ 2008 Functional characterization of the native NH2-terminal transactivation domain of the human androgen receptor: Binding kinetics for interactions with TFIIF and SRC-1a. Biochemistry 47(11):3352-3359
  108. De Mol E, Fenwick RB, Phang CT, Buzón V, Szulc E, de la Fuente A, Escobedo A, García J, Bertoncini CW, Estébanez-Perpiñá E, McEwan IJ, Riera A, Salvatella X. 2016 EPI-001, A Compound Active against Castration-Resistant Prostate Cancer, Targets Transactivation Unit 5 of the Androgen Receptor. ACS Chem Biol. 11(9): 2499-505. 
  109. Reid J, Kelly SM, Watt K, Price NC, McEwan IJ. 2002 Conformational analysis of the androgen receptor amino-terminal domain involved in transactivation. Influence of structure-stabilizing solutes and protein-protein interactions. J Biol Chem. 277(22):20079-86.
  110. De Mol E, Szulc E, Di Sanza C, Martínez-Cristóbal P, Bertoncini CW, Fenwick RB, Frigolé-Vivas M, Masín M, Hunter I, Buzón V, Brun-Heath I, García J, De Fabritiis G, Estébanez-Perpiñá E, McEwan IJ, Nebreda ÁR, Salvatella X. 2018 Regulation of Androgen Receptor Activity by Transient Interactions of Its Transactivation Domain with General Transcription Regulators. Structure. 26(1):145-152.
  111. Eftekharzadeh B, Piai A, Chiesa G, Mungianu D, García J, Pierattelli R, Felli IC, Salvatella X. 2016 Sequence Context Influences the Structure and Aggregation Behavior of a PolyQ Tract. Biophys J. 110(11): 2361-2366.
  112. Meyer S, Ying-Hui Wang Y-H, Pau Pérez-Escrivà P, Bruno Kieffer B. 2016 Backbone 1H, 15N, 13C NMR assignment of the 518-627 fragment of the androgen receptor encompassing N-terminal and DNA binding domains. Biomol NMR Assign 10(1):175-8.
  113. Langley E, Zhou ZX, Wilson EM 1995 Evidence for an anti-parallel orientation of the ligand-activated human androgen receptor dimer. J Biol Chem 270(50):29983-29990
  114. Doesburg P, Kuil CW, Berrevoets CA, Steketee K, Faber PW, Mulder E, Brinkmann AO, Trapman J 1997 Functional in vivo interaction between the amino-terminal, transactivation domain and the ligand binding domain of the androgen receptor. Biochemistry 36(5):1052-1064
  115. Berrevoets CA, Doesburg P, Steketee K, Trapman J, Brinkmann AO 1998 Functional interactions of the AF-2 activation domain core region of the human androgen receptor with the amino-terminal domain and with the transcriptional coactivator TIF2 (transcriptional intermediary factor2). Mol Endocrinol 12(8):1172-1183
  116. Zhou ZX, Lane MV, Kemppainen JA, French FS, Wilson EM 1995 Specificity of ligand-dependent androgen receptor stabilization: Receptor domain interactions influence ligand dissociation and receptor stability. Mol Endocrinol 9(2):208-218
  117. Centenera MM, Harris JM, Tilley WD, Butler LM 2008 The contribution of different androgen receptor domains to receptor dimerization and signaling. Mol Endocrinol 22(11):2373-2382
  118. Schaufele F, Carbonell X, Guerbadot M, Borngraeber S, Chapman MS, Ma AA, Miner JN, Diamond MI 2005 The structural basis of androgen receptor activation: Intramolecular and intermolecular amino-carboxy interactions. Proc Natl Acad Sci U S A 102(28):9802-9807
  119. van Royen ME, Cunha SM, Brink MC, Mattern KA, Nigg AL, Dubbink HJ, Verschure PJ, Trapman J, Houtsmuller AB 2007 Compartmentalization of androgen receptor protein-protein interactions in living cells. J Cell Biol 177(1):63-72
  120. Thompson J, Saatcioglu F, Janne OA, Palvimo JJ 2001 Disrupted amino- and carboxyl-terminal interactions of the androgen receptor are linked to androgen insensitivity. Mol Endocrinol 15(6):923-935
  121. Brodie J, McEwan IJ 2005 Intra-domain communication between the N-terminal and DNA-binding domains of the androgen receptor: Modulation of androgen response element DNA binding. J Mol Endocrinol 34(3):603-615
  122. Luisi BF, Xu WX, Otwinowski Z, Freedman LP, Yamamoto KR, Sigler PB 1991 Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature 352(6335):497-505
  123. Shaffer PL, Jivan A, Dollins DE, Claessens F, Gewirth DT 2004 Structural basis of androgen receptor binding to selective androgen response elements. Proc Natl Acad Sci U S A 101(14):4758-4763
  124. Jakob M, Kolodziejczyk R, Orlowski M, Krzywda S, Kowalska A, Dutko-Gwozdz J, Gwozdz T, Kochman M, Jaskolski M, Ozyhar A 2007 Novel DNA-binding element within the C-terminal extension of the nuclear receptor DNA-binding domain. Nucleic Acids Res 35(8):2705-2718
  125. Haelens A, Tanner T, Denayer S, Callewaert L, Claessens F 2007 The hinge region regulates DNA binding, nuclear translocation, and transactivation of the androgen receptor. Cancer Res 67(9):4514-4523
  126. Schauwaers K, De Gendt K, Saunders PT, Atanassova N, Haelens A, Callewaert L, Moehren U, Swinnen JV, Verhoeven G, Verrijdt G, Claessens F 2007 Loss of androgen receptor binding to selective androgen response elements causes a reproductive phenotype in a knockin mouse model. Proc Natl Acad Sci U S A 104(12):4961-4966
  127. Massie CE, Adryan B, Barbosa-Morais NL, Lynch AG, Tran MG, Neal DE, Mills IG 2007 New androgen receptor genomic targets show an interaction with the ETS1 transcription factor. EMBO Rep 8(9):871-878
  128. Bolton EC, So AY, Chaivorapol C, Haqq CM, Li H, Yamamoto KR 2007 Cell- and gene-specific regulation of primary target genes by the androgen receptor. Genes Dev 21(16):2005-2017
  129. Eacker SM, Shima JE, Connolly CM, Sharma M, Holdcraft RW, Griswold MD, Braun RE 2007 Transcriptional profiling of androgen receptor (AR) mutants suggests instructive and permissive roles of AR signaling in germ cell development. Mol Endocrinol 21(4):895-907
  130. Wang Q, Li W, Liu XS, Carroll JS, Janne OA, Keeton EK, Chinnaiyan AM, Pienta KJ, Brown M 2007 A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol Cell 27(3):380-392
  131. Chen Z, Lan X, Thomas-Ahner JM, Wu D, Liu X, Ye Z, Wang L, Sunkel B, Grenade C, Chen J, Zynger DL, Yan PS, Huang J, Nephew KP, Huang TH, Lin S, Clinton SK, Li W, Jin VX, Wang Q 2015 Agonist and antagonist switch DNA motifs recognized by human androgen receptor in prostate cancer. EMBO J 34(4):502-516
  132. Fu M, Wang C, Zhang X, Pestell RG 2004 Acetylation of nuclear receptors in cellular growth and apoptosis. Biochem Pharmacol 68(6):1199-1208

133   Clinckemalie L, Vanderschueren D, Boonen S, Claessens F 2012 The hinge region in androgen receptor control. Mol Cell Endocrinol 358(1):1-8

  1. Brinkmann AO, Trapman J 2000 Genetic analysis of androgen receptors in development and disease. Adv Pharmacol 47:317-341
  2. Matias PM, Donner P, Coelho R, Thomaz M, Peixoto C, Macedo S, Otto N, Joschko S, Scholz P, Wegg A, Basler S, Schafer M, Egner U, Carrondo MA 2000 Structural evidence for ligand specificity in the binding domain of the human androgen receptor. implications for pathogenic gene mutations. J Biol Chem 275(34):26164-26171
  3. Sack JS, Kish KF, Wang C, Attar RM, Kiefer SE, An Y, Wu GY, Scheffler JE, Salvati ME, Krystek SR,Jr, Weinmann R, Einspahr HM 2001 Crystallographic structures of the ligand-binding domains of the androgen receptor and its T877A mutant complexed with the natural agonist dihydrotestosterone. Proc Natl Acad Sci U S A 98(9):4904-4909
  4. Pereira de Jesus-Tran K, Cote PL, Cantin L, Blanchet J, Labrie F, Breton R 2006 Comparison of crystal structures of human androgen receptor ligand-binding domain complexed with various agonists reveals molecular determinants responsible for binding affinity. Protein Sci 15(5):987-999
  5. McEwan IJ 2013 Androgen receptor modulators: A marriage of chemistry and biology. Future Med Chem 5(10):1109-1120
  6. Veldscholte J, Berrevoets CA, Ris-Stalpers C, Kuiper GG, Jenster G, Trapman J, Brinkmann AO, Mulder E 1992 The androgen receptor in LNCaP cells contains a mutation in the ligand binding domain which affects steroid binding characteristics and response to antiandrogens. J Steroid Biochem Mol Biol 41(3-8):665-669
  7. Taplin ME, Bubley GJ, Shuster TD, Frantz ME, Spooner AE, Ogata GK, Keer HN, Balk SP 1995 Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer. N Engl J Med 332(21):1393-1398
  8. Taplin ME, Bubley GJ, Ko YJ, Small EJ, Upton M, Rajeshkumar B, Balk SP 1999 Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Res 59(11):2511-2515
  9. Zhao XY, Boyle B, Krishnan AV, Navone NM, Peehl DM, Feldman D 1999 Two mutations identified in the androgen receptor of the new human prostate cancer cell line MDA PCa 2a. J Urol 162(6):2192-2199
  10. Zhao XY, Malloy PJ, Krishnan AV, Swami S, Navone NM, Peehl DM, Feldman D 2000 Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor. Nat Med 6(6):703-706
  11. Krishnan AV, Zhao XY, Swami S, Brive L, Peehl DM, Ely KR, Feldman D 2002 A glucocorticoid-responsive mutant androgen receptor exhibits unique ligand specificity: Therapeutic implications for androgen-independent prostate cancer. Endocrinology 143(5):1889-1900
  12. Shi XB, Ma AH, Xia L, Kung HJ, de Vere White RW 2002 Functional analysis of 44 mutant androgen receptors from human prostate cancer. Cancer Res 62(5):1496-1502
  13. Hay CW, McEwan IJ 2012 The impact of point mutations in the human androgen receptor: Classification of mutations on the basis of transcriptional activity. PLoS One 7(3):e32514
  14. Estebanez-Perpina E, Arnold LA, Nguyen P, Rodrigues ED, Mar E, Bateman R, Pallai P, Shokat KM, Baxter JD, Guy RK, Webb P, Fletterick RJ 2007 A surface on the androgen receptor that allosterically regulates coactivator binding. Proc Natl Acad Sci U S A 104(41):16074-16079
  15. Dubbink HJ, Hersmus R, Verma CS, van der Korput HA, Berrevoets CA, van Tol J, Ziel-van der Made AC, Brinkmann AO, Pike AC, Trapman J 2004 Distinct recognition modes of FXXLF and LXXLL motifs by the androgen receptor. Mol Endocrinol 18(9):2132-2150
  16. He B, Gampe RT,Jr, Kole AJ, Hnat AT, Stanley TB, An G, Stewart EL, Kalman RI, Minges JT, Wilson EM 2004 Structural basis for androgen receptor interdomain and coactivator interactions suggests a transition in nuclear receptor activation function dominance. Mol Cell 16(3):425-438
  17. Hur E, Pfaff SJ, Payne ES, Gron H, Buehrer BM, Fletterick RJ 2004 Recognition and accommodation at the androgen receptor coactivator binding interface. PLoS Biol 2(9):E274
  18. Gottlieb B, Beitel LK, Nadarajah A, Paliouras M, Trifiro M 2012 The androgen receptor gene mutations database: 2012 update. Hum Mutat 33(5):887-894
  19. Nadal M, Prekovic S, Gallastegui N, Helsen C, Abella M, Zielinska K, Gay M, Vilaseca M, Taulès M, Houtsmuller AB, van Royen ME, Claessens F, Fuentes-Prior P, Estébanez-Perpiñá E 2017 Structure of the homodimeric androgen receptor ligand-binding domain. .Nat Commun. 8:14388.
  20. Jiménez-Panizo A, Pérez P, Rojas AM, Fuentes-Prior P, Estébanez-Perpiñá E 2019 Non-canonical dimerization of the androgen receptor and other nuclear receptors: implications for human disease. Endocr Relat Cancer. 26(8): R479-R497.
  21. Jenster G, van der Korput HA, van Vroonhoven C, van der Kwast TH, Trapman J, Brinkmann AO 1991 Domains of the human androgen receptor involved in steroid binding, transcriptional activation, and subcellular localization. Mol Endocrinol 5(10):1396-1404
  22. Hollenberg SM, Evans RM 1988 Multiple and cooperative trans-activation domains of the human glucocorticoid receptor. Cell 55(5):899-906
  23. Lu J, Van der Steen T, Tindall DJ 2015 Are androgen receptor variants a substitute for the full-length receptor? Nat Rev Urol 12(3):137-144
  24. Hu R, Isaacs WB, Luo J 2011 A snapshot of the expression signature of androgen receptor splicing variants and their distinctive transcriptional activities. Prostate
  25. Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, Chen H, Kong X, Melamed J, Tepper CG, Kung HJ, Brodie AM, Edwards J, Qiu Y 2009 A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res 69(6):2305-2313
  26. Sun S, Sprenger CC, Vessella RL, Haugk K, Soriano K, Mostaghel EA, Page ST, Coleman IM, Nguyen HM, Sun H, Nelson PS, Plymate SR 2010 Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clin Invest 120(8):2715-2730
  27. Krause WC, Shafi AA, Nakka M, Weigel NL 2014 Androgen receptor and its splice variant, AR-V7, differentially regulate FOXA1 sensitive genes in LNCaP prostate cancer cells. Int J Biochem Cell Biol 54:49-59
  28. Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ 2008 Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res 68(13):5469-5477
  29. Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, Widmark A, Bergh A, Wikstrom P 2011 Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival. PLoS One 6(4):e19059
  30. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, Chen Y, Mohammad TA, Chen Y, Fedor HL, Lotan TL, Zheng Q, De Marzo AM, Isaacs JT, Isaacs WB, Nadal R, Paller CJ, Denmeade SR, Carducci MA, Eisenberger MA, Luo J 2014 AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 371(11):1028-1038
  31. Yu X, Yi P, Hamilton RA, Shen H, Chen M, Foulds CE, Mancini MA, Ludtke SJ, Wang Z, O'Malley BW. 2020 Structural Insights of Transcriptionally Active, Full-Length Androgen Receptor Coactivator Complexes. Mol Cell. 79(5):812-823.e4.
  32. Yi P, Wang Z, Feng Q, Chou CK, Pintilie GD, Shen H, Foulds CE, Fan G, Serysheva I, Ludtke SJ, Schmid MF, Hung MC, Chiu W, O'Malley BW. 2017 Structural and Functional Impacts of ER Coactivator Sequential Recruitment. Mol Cell. ;67(5):733-743.
  33. Kumar R, Betney R, Li J, Thompson EB, McEwan IJ. 2004 Induced alpha-helix structure in AF1 of the androgen receptor upon binding transcription factor TFIIF. Biochemistry 43(11):3008-13. 
  34. Lavery DN, Bevan CL 2011 Androgen receptor signalling in prostate cancer: The functional consequences of acetylation. J Biomed Biotechnol 2011:862125
  35. Anbalagan M, Huderson B, Murphy L, Rowan BG 2012 Post-translational modifications of nuclear receptors and human disease. Nucl Recept Signal 10:e001
  36. Coffey K, Robson CN 2012 Regulation of the androgen receptor by post-translational modifications. J Endocrinol 215(2):221-237
  37. Gioeli D, Paschal BM 2012 Post-translational modification of the androgen receptor. Mol Cell Endocrinol 352(1-2):70-78
  38. van der Steen T, Tindall DJ, Huang H 2013 Posttranslational modification of the androgen receptor in prostate cancer. Int J Mol Sci 14(7):14833-14859
  39. Gaughan L, Stockley J, Wang N, McCracken SR, Treumann A, Armstrong K, Shaheen F, Watt K, McEwan IJ, Wang C, Pestell RG, Robson CN 2011 Regulation of the androgen receptor by SET9-mediated methylation. Nucleic Acids Res 39(4):1266-1279
  40. Ko S, Ahn J, Song CS, Kim S, Knapczyk-Stwora K, Chatterjee B 2011 Lysine methylation and functional modulation of androgen receptor by Set9 methyltransferase. Mol Endocrinol 25(3):433-444
  41. Fu M, Rao M, Wang C, Sakamaki T, Wang J, Di Vizio D, Zhang X, Albanese C, Balk S, Chang C, Fan S, Rosen E, Palvimo JJ, Janne OA, Muratoglu S, Avantaggiati ML, Pestell RG 2003 Acetylation of androgen receptor enhances coactivator binding and promotes prostate cancer cell growth. Mol Cell Biol 23(23):8563-8575
  42. Zhong J, Ding L, Bohrer LR, Pan Y, Liu P, Zhang J, Sebo TJ, Karnes RJ, Tindall DJ, van Deursen J, Huang H 2014 p300 acetyltransferase regulates androgen receptor degradation and PTEN-deficient prostate tumorigenesis. Cancer Res 74(6):1870-1880
  43. Xu K, Shimelis H, Linn DE, Jiang R, Yang X, Sun F, Guo Z, Chen H, Li W, Chen H, Kong X, Melamed J, Fang S, Xiao Z, Veenstra TD, Qiu Y 2009 Regulation of androgen receptor transcriptional activity and specificity by RNF6-induced ubiquitination. Cancer Cell 15(4):270-282
  44. Kaikkonen S, Jaaskelainen T, Karvonen U, Rytinki MM, Makkonen H, Gioeli D, Paschal BM, Palvimo JJ 2009 SUMO-specific protease 1 (SENP1) reverses the hormone-augmented SUMOylation of androgen receptor and modulates gene responses in prostate cancer cells. Mol Endocrinol 23(3):292-307
  45. Sutinen P, Malinen M, Heikkinen S, Palvimo JJ 2014 SUMOylation modulates the transcriptional activity of androgen receptor in a target gene and pathway selective manner. Nucleic Acids Res 42(13):8310-8319
  46. Zhang FP, Malinen M, Mehmood A, Lehtiniemi T, Jääskeläinen T, Niskanen EA, Korhonen H, Laiho A, Elo LL, Ohlsson C, Kotaja N, Poutanen M, Sipilä P, Palvimo JJ. 2019 Lack of androgen receptor SUMOylation results in male infertility due to epididymal dysfunction. Nat Commun. 10(1):777.
  47. Gioeli D, Ficarro SB, Kwiek JJ, Aaronson D, Hancock M, Catling AD, White FM, Christian RE, Settlage RE, Shabanowitz J, Hunt DF, Weber MJ 2002 Androgen receptor phosphorylation. regulation and identification of the phosphorylation sites. J Biol Chem 277(32):29304-29314
  48. Koryakina Y, Ta HQ, Gioeli D 2014 Androgen receptor phosphorylation: Biological context and functional consequences. Endocr Relat Cancer 21(4):T131-45
  49. Kuiper GG, de Ruiter PE, Grootegoed JA, Brinkmann AO 1991 Synthesis and post-translational modification of the androgen receptor in LNCaP cells. Mol Cell Endocrinol 80(1-3):65-73
  50. Zhou ZX, Kemppainen JA, Wilson EM 1995 Identification of three proline-directed phosphorylation sites in the human androgen receptor. Mol Endocrinol 9(5):605-615
  51. Gordon V, Bhadel S, Wunderlich W, Zhang J, Ficarro SB, Mollah SA, Shabanowitz J, Hunt DF, Xenarios I, Hahn WC, Conaway M, Carey MF, Gioeli D 2010 CDK9 regulates AR promoter selectivity and cell growth through serine 81 phosphorylation. Mol Endocrinol 24(12):2267-2280
  52. Chen S, Gulla S, Cai C, Balk SP 2012 Androgen receptor serine 81 phosphorylation mediates chromatin binding and transcriptional activation. J Biol Chem 287(11):8571-8583
  53. Williamson M, de Winter P, Masters JR 2016 Plexin-B1 signalling promotes androgen receptor translocation to the nucleus. Oncogene 35(8):1066-1072
  54. Blok LJ, de Ruiter PE, Brinkmann AO 1998 Forskolin-induced dephosphorylation of the androgen receptor impairs ligand binding. Biochemistry 37(11):3850-3857
  55. Wong HY, Burghoorn JA, Van Leeuwen M, De Ruiter PE, Schippers E, Blok LJ, Li KW, Dekker HL, De Jong L, Trapman J, Grootegoed JA, Brinkmann AO 2004 Phosphorylation of androgen receptor isoforms. Biochem J 383(Pt 2):267-276
  56. Ponguta LA, Gregory CW, French FS, Wilson EM 2008 Site-specific androgen receptor serine phosphorylation linked to epidermal growth factor-dependent growth of castration-recurrent prostate cancer. J Biol Chem 283(30):20989-21001

190   Koryakina Y, Knudsen KE, Gioeli D 2015 Cell-cycle-dependent regulation of androgen receptor function. Endocr Relat Cancer 22(2):249-264

  1. Chymkowitch P, Le May N, Charneau P, Compe E, Egly JM 2011 The phosphorylation of the androgen receptor by TFIIH directs the ubiquitin/proteasome process. EMBO J 30(3):468-479
  2. Chen S, Kesler CT, Paschal BM, Balk SP 2009 Androgen receptor phosphorylation and activity are regulated by an association with protein phosphatase 1. J Biol Chem 284(38):25576-25584
  3. Liu X, Han W, Gulla S, Simon NI, Gao Y, Cai C, Yang H, Zhang X, Liu J, Balk SP, Chen S 2016 Protein phosphatase 1 suppresses androgen receptor ubiquitylation and degradation. Oncotarget 7(2):1754-1764
  4. Yang CS, Xin HW, Kelley JB, Spencer A, Brautigan DL, Paschal BM 2007 Ligand binding to the androgen receptor induces conformational changes that regulate phosphatase interactions. Mol Cell Biol 27(9):3390-3404
  5. Taneja SS, Ha S, Swenson NK, Huang HY, Lee P, Melamed J, Shapiro E, Garabedian MJ, Logan SK 2005 Cell-specific regulation of androgen receptor phosphorylation in vivo. J Biol Chem 280(49):40916-40924
  6. Zong H, Chi Y, Wang Y, Yang Y, Zhang L, Chen H, Jiang J, Li Z, Hong Y, Wang H, Yun X, Gu J 2007 Cyclin D3/CDK11p58 complex is involved in the repression of androgen receptor. Mol Cell Biol 27(20):7125-7142
  7. Wen Y, Hu MC, Makino K, Spohn B, Bartholomeusz G, Yan DH, Hung MC 2000 HER-2/neu promotes androgen-independent survival and growth of prostate cancer cells through the akt pathway. Cancer Res 60(24):6841-6845
  8. Kuiper GG, De Ruiter PE, Brinkmann AO 1993 Androgen receptor phosphorylation. Ann N Y Acad Sci 684:224-226
  9. Guo Z, Dai B, Jiang T, Xu K, Xie Y, Kim O, Nesheiwat I, Kong X, Melamed J, Handratta VD, Njar VC, Brodie AM, Yu LR, Veenstra TD, Chen H, Qiu Y 2006 Regulation of androgen receptor activity by tyrosine phosphorylation. Cancer Cell 10(4):309-319
  10. Liu Y, Karaca M, Zhang Z, Gioeli D, Earp HS, Whang YE 2010 Dasatinib inhibits site-specific tyrosine phosphorylation of androgen receptor by Ack1 and src kinases. Oncogene 29(22):3208-3216
  11. Mahajan NP, Liu Y, Majumder S, Warren MR, Parker CE, Mohler JL, Earp HS, Whang YE 2007 Activated Cdc42-associated kinase Ack1 promotes prostate cancer progression via androgen receptor tyrosine phosphorylation. Proc Natl Acad Sci U S A 104(20):8438-8443
  12. Karaca M, Liu Y, Zhang Z, De Silva D, Parker JS, Earp HS, Whang YE 2015 Mutation of androgen receptor N-terminal phosphorylation site tyr-267 leads to inhibition of nuclear translocation and DNA binding. PLoS One 10(5):e0126270
  13. Shu SK, Liu Q, Coppola D, Cheng JQ 2010 Phosphorylation and activation of androgen receptor by aurora-A. J Biol Chem 285(43):33045-33053
  14. Koryakina Y, Ta HQ, Gioeli D. (2014) Phosphorylation of the Androgen Receptor. Endocr Relat Cancer. 2014 Aug; 21(4): T131–T145.
  15. Craft N, Shostak Y, Carey M, Sawyers CL 1999 A mechanism for hormone-independent prostate cancer through modulation of androgen receptor signaling by the HER-2/neu tyrosine kinase. Nat Med 5(3):280-285
  16. Yeh S, Lin HK, Kang HY, Thin TH, Lin MF, Chang C 1999 From HER2/Neu signal cascade to androgen receptor and its coactivators: A novel pathway by induction of androgen target genes through MAP kinase in prostate cancer cells. Proc Natl Acad Sci U S A 96(10):5458-5463
  17. Neumann F, Topert M 1986 Pharmacology of antiandrogens. J Steroid Biochem 25(5B):885-895
  18. Raynaud JP, Ojasoo T 1986 The design and use of sex-steroid antagonists. J Steroid Biochem 25(5B):811-833
  19. Furr BJ, Valcaccia B, Curry B, Woodburn JR, Chesterson G, Tucker H 1987 ICI 176,334: A novel non-steroidal, peripherally selective antiandrogen. J Endocrinol 113(3):R7-9
  20. Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, Wasielewska T, Welsbie D, Chen CD, Higano CS, Beer TM, Hung DT, Scher HI, Jung ME, Sawyers CL 2009 Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324(5928):787-790
  21. Rathkopf DE, Scher HI. 2018 Apalutamide for the treatment of prostate cancer. Expert Rev Anticancer Ther. 18(9):823-836.
  22. Sugawara T, Baumgart SJ, Nevedomskaya E, Reichert K, Steuber H, Lejeune P, Mumberg D, Haendler B. 2019 Darolutamide is a potent androgen receptor antagonist with strong efficacy in prostate cancer models. Int J Cancer. 145(5):1382-1394.
  23. Korpal M, Korn JM, Gao X, Rakiec DP, Ruddy DA, Doshi S, Yuan J, Kovats SG, Kim S, Cooke VG, Monahan JE, Stegmeier F, Roberts TM, Sellers WR, Zhou W, Zhu P 2013 An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide). Cancer Discov 3(9):1030-1043
  24. Kuil CW, Mulder E 1994 Mechanism of antiandrogen action: Conformational changes of the receptor. Mol Cell Endocrinol 102(1-2):R1-5
  25. Kuil CW, Berrevoets CA, Mulder E 1995 Ligand-induced conformational alterations of the androgen receptor analyzed by limited trypsinization. studies on the mechanism of antiandrogen action. J Biol Chem 270(46):27569-27576

216   Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, Chen Y, Grillot K, Bischoff ED, Cai L, Aparicio A, Dorow S, Arora V, Shao G, Qian J, Zhao H, Yang G, Cao C, Sensintaffar J, Wasielewska T, Herbert MR, Bonnefous C, Darimont B, Scher HI, Smith-Jones P, Klang M, Smith ND, De Stanchina E, Wu N, Ouerfelli O, Rix PJ, Heyman RA, Jung ME, Sawyers CL, Hager JH 2012 ARN-509: A novel antiandrogen for prostate cancer treatment. Cancer Res 72(6):1494-1503

  1. Lin TH, Lee SO, Niu Y, Xu D, Liang L, Li L, Yeh SD, Fujimoto N, Yeh S, Chang C 2013 Differential androgen deprivation therapies with anti-androgens casodex/bicalutamide or MDV3100/Enzalutamide versus anti-androgen receptor ASC-J9(R) lead to promotion versus suppression of prostate cancer metastasis. J Biol Chem 288(27):19359-19369
  2. Loddick SA, Ross SJ, Thomason AG, Robinson DM, Walker GE, Dunkley TP, Brave SR, Broadbent N, Stratton NC, Trueman D, Mouchet E, Shaheen FS, Jacobs VN, Cumberbatch M, Wilson J, Jones RD, Bradbury RH, Rabow A, Gaughan L, Womack C, Barry ST, Robson CN, Critchlow SE, Wedge SR, Brooks AN 2013 AZD3514: A small molecule that modulates androgen receptor signaling and function in vitro and in vivo. Mol Cancer Ther 12(9):1715-1727
  3. Kuruma H, Matsumoto H, Shiota M, Bishop J, Lamoureux F, Thomas C, Briere D, Los G, Gleave M, Fanjul A, Zoubeidi A 2013 A novel antiandrogen, compound 30, suppresses castration-resistant and MDV3100-resistant prostate cancer growth in vitro and in vivo. Mol Cancer Ther 12(5):567-576
  4. Li H, Hassona MD, Lack NA, Axerio-Cilies P, Leblanc E, Tavassoli P, Kanaan N, Frewin K, Singh K, Adomat H, Bohm KJ, Prinz H, Guns ET, Rennie PS, Cherkasov A 2013 Characterization of a new class of androgen receptor antagonists with potential therapeutic application in advanced prostate cancer. Mol Cancer Ther 12(11):2425-2435
  5. Scher HI, Beer TM, Higano CS, Anand A, Taplin ME, Efstathiou E, Rathkopf D, Shelkey J, Yu EY, Alumkal J, Hung D, Hirmand M, Seely L, Morris MJ, Danila DC, Humm J, Larson S, Fleisher M, Sawyers CL, Prostate Cancer Foundation/Department of Defense Prostate Cancer Clinical Trials Consortium 2010 Antitumour activity of MDV3100 in castration-resistant prostate cancer: A phase 1-2 study. Lancet 375(9724):1437-1446
  6. Hoffman-Censits J, Kelly WK 2013 Enzalutamide: A novel antiandrogen for patients with castrate-resistant prostate cancer. Clin Cancer Res 19(6):1335-1339
  7. Menon MP, Higano CS 2013 Enzalutamide, a second generation androgen receptor antagonist: Development and clinical applications in prostate cancer. Curr Oncol Rep 15(2):69-75
  8. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Maneval EC, Chen I, Darimont B, Hager JH 2013 A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discov 3(9):1020-1029
  9. Veldscholte J, Berrevoets CA, Brinkmann AO, Grootegoed JA, Mulder E 1992 Anti-androgens and the mutated androgen receptor of LNCaP cells: Differential effects on binding affinity, heat-shock protein interaction, and transcription activation. Biochemistry 31(8):2393-2399
  10. Lamont KR, Tindall DJ 2011 Minireview: Alternative activation pathways for the androgen receptor in prostate cancer. Mol Endocrinol 25(6):897-907
  11. Negro-Vilar A 1999 Selective androgen receptor modulators (SARMs): A novel approach to androgen therapy for the new millennium. J Clin Endocrinol Metab 84(10):3459-3462
  12. Narayanan R, Mohler ML, Bohl CE, Miller DD, Dalton JT 2008 Selective androgen receptor modulators in preclinical and clinical development. Nucl Recept Signal 6:e010
  13. Haendler B, Cleve A 2012 Recent developments in antiandrogens and selective androgen receptor modulators. Mol Cell Endocrinol 352(1-2):79-91
  14. Berrevoets CA, Umar A, Trapman J, Brinkmann AO 2004 Differential modulation of androgen receptor transcriptional activity by the nuclear receptor co-repressor (N-CoR). Biochem J 379(Pt 3):731-738
  15. Augello MA, Hickey TE, Knudsen KE 2011 FOXA1: Master of steroid receptor function in cancer. EMBO J 30(19):3885-3894
  16. Grosse A, Bartsch S, Baniahmad A 2012 Androgen receptor-mediated gene repression. Mol Cell Endocrinol 352(1-2):46-56
  17. Wang RS, Yeh S, Tzeng CR, Chang C 2009 Androgen receptor roles in spermatogenesis and fertility: Lessons from testicular cell-specific androgen receptor knockout mice. Endocr Rev 30(2):119-132
  18. Walters KA, Simanainen U, Handelsman DJ 2010 Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Hum Reprod Update 16(5):543-558
  19. De Gendt K, Verhoeven G 2012 Tissue- and cell-specific functions of the androgen receptor revealed through conditional knockout models in mice. Mol Cell Endocrinol 352(1-2):13-25
  20. Robins DM 2012 Androgen receptor gene polymorphisms and alterations in prostate cancer: Of humanized mice and men. Mol Cell Endocrinol 352(1-2):26-33
  21. Matsumoto T, Sakari M, Okada M, Yokoyama A, Takahashi S, Kouzmenko A, Kato S 2013 The androgen receptor in health and disease. Annu Rev Physiol 75:201-224
  22. O'Hara L, Smith LB. (2016) Development and Characterization of Cell-Specific Androgen Receptor Knockout Mice. Methods Mol Biol. 1443:219-48.
  23. Dart DA, Waxman J, Aboagye EO, Bevan CL 2013 Visualising androgen receptor activity in male and female mice. PLoS One 8(8):e71694
  24. Zhang H, Li XX, Yang Y, Zhang Y, Wang HY, Zheng XFS  2018 Significance and mechanism of androgen receptor overexpression and androgen receptor/mechanistic target of rapamycin cross-talk in hepatocellular carcinoma. Hepatology. 67(6):2271-2286.
  25. Li P, Chen J, Miyamoto H2017 Androgen Receptor Signaling in Bladder Cancer..Cancers (Basel). 9(2):20.
  26. Huang CK, Lee SO, Chang E, Pang H, Chang C 2016 Androgen receptor (AR) in cardiovascular diseases. J Endocrinol. 229(1):R1-R16.
  27. Agiannitopoulos K, Bakalgianni A, Marouli E, Zormpa I, Manginas A, Papamenzelopoulos S, Lamnissou K.J 2016 Gender Specificity of a Genetic Variant of Androgen Receptor and Risk of Coronary Artery Disease. Clin Lab Anal. 30(3):204-7. 
  28. Takov K, Wu J, Denvir MA, Smith LB, Hadoke PWF 2018 The role of androgen receptors in atherosclerosis. Mol Cell Endocrinol. 465: 82-91.
  29. Reifenstein EC,Jr 1947 Hereditary familial hypogonadism. Proc Am Fed Clin Res 3:86
  30. Wilson JD, Harrod MJ, Goldstein JL, Hemsell DL, MacDonald PC 1974 Familial incomplete male pseudohermaphroditism, type 1: evidence for androgen resistance and variable clinical manifestations in a family with the reifenstein syndrome. N Engl J Med 290(20):1097-1103
  31. Geissler WM, Davis DL, Wu L, Bradshaw KD, Patel S, Mendonca BB, Elliston KO, Wilson JD, Russell DW, Andersson S 1994 Male pseudohermaphroditism caused by mutations of testicular 17 beta-hydroxysteroid dehydrogenase 3. Nat Genet 7(1):34-39
  32. Hughes IA 2008 Disorders of sex development: A new definition and classification. Best Pract Res Clin Endocrinol Metab 22(1):119-134
  33. Sai TJ, Seino S, Chang CS, Trifiro M, Pinsky L, Mhatre A, Kaufman M, Lambert B, Trapman J, Brinkmann AO 1990 An exonic point mutation of the androgen receptor gene in a family with complete androgen insensitivity. Am J Hum Genet 46(6):1095-1100
  34. Quigley CA, De Bellis A, Marschke KB, el-Awady MK, Wilson EM, French FS 1995 Androgen receptor defects: Historical, clinical, and molecular perspectives. Endocr Rev 16(3):271-321
  35. Boehmer AL, Brinkmann AO, Nijman RM, Verleun-Mooijman MC, de Ruiter P, Niermeijer MF, Drop SL 2001 Phenotypic variation in a family with partial androgen insensitivity syndrome explained by differences in 5alpha dihydrotestosterone availability. J Clin Endocrinol Metab 86(3):1240-1246
  36. Batch JA, Davies HR, Evans BA, Hughes IA, Patterson MN 1993 Phenotypic variation and detection of carrier status in the partial androgen insensitivity syndrome. Arch Dis Child 68(4):453-457
  37. Imasaki K, Hasegawa T, Okabe T, Sakai Y, Haji M, Takayanagi R, Nawata H 1994 Single amino acid substitution (840Arg-->His) in the hormone-binding domain of the androgen receptor leads to incomplete androgen insensitivity syndrome associated with a thermolabile androgen receptor. Eur J Endocrinol 130(6):569-574
  38. Evans BA, Hughes IA, Bevan CL, Patterson MN, Gregory JW 1997 Phenotypic diversity in siblings with partial androgen insensitivity syndrome. Arch Dis Child 76(6):529-531
  39. Boehmer AL, Brinkmann O, Bruggenwirth H, van Assendelft C, Otten BJ, Verleun-Mooijman MC, Niermeijer MF, Brunner HG, Rouwe CW, Waelkens JJ, Oostdijk W, Kleijer WJ, van der Kwast TH, de Vroede MA, Drop SL 2001 Genotype versus phenotype in families with androgen insensitivity syndrome. J Clin Endocrinol Metab 86(9):4151-4160
  40. Lu J, Danielsen M 1996 A stu I polymorphism in the human androgen receptor gene (AR). Clin Genet 49(6):323-324
  41. Davies HR, Hughes IA, Patterson MN 1995 Genetic counselling in complete androgen insensitivity syndrome: Trinucleotide repeat polymorphisms, single-strand conformation polymorphism and direct detection of two novel mutations in the androgen receptor gene. Clin Endocrinol (Oxf) 43(1):69-77
  42. Ris-Stalpers C, Hoogenboezem T, Sleddens HF, Verleun-Mooijman MC, Degenhart HJ, Drop SL, Halley DJ, Oosterwijk JC, Hodgins MB, Trapman J 1994 A practical approach to the detection of androgen receptor gene mutations and pedigree analysis in families with x-linked androgen insensitivity. Pediatr Res 36(2):227-234
  43. Kohler B, Lumbroso S, Leger J, Audran F, Grau ES, Kurtz F, Pinto G, Salerno M, Semitcheva T, Czernichow P, Sultan C 2005 Androgen insensitivity syndrome: Somatic mosaicism of the androgen receptor in seven families and consequences for sex assignment and genetic counseling. J Clin Endocrinol Metab 90(1):106-111
  44. Elfferich P, van Royen ME, van de Wijngaart DJ, Trapman J, Drop SL, van den Akker EL, Lusher SJ, Bosch R, Bunch T, Hughes IA, Houtsmuller AB, Cools M, Faradz SM, Bisschop PH, Bunck MC, Oostdijk W, Bruggenwirth HT, Brinkmann AO 2013 Variable loss of functional activities of androgen receptor mutants in patients with androgen insensitivity syndrome. Sex Dev 7(5):223-234
  45. Topcu V, Ilgin-Ruhi H, Siklar Z, Karabulut HG, Berberoglu M, Hacihamdioglu B, Savas-Erdeve S, Aycan Z, Peltek-Kendirci HN, Ocal G, Tukun FA 2015 Investigation of androgen receptor gene mutations in a series of 21 patients with 46,XY disorders of sex development. J Pediatr Endocrinol Metab 28(11-12):1257-1263
  46. Phelan N, Williams EL, Cardamone S, Lee M, Creighton SM, Rumsby G, Conway GS 2015 Screening for mutations in 17beta-hydroxysteroid dehydrogenase and androgen receptor in women presenting with partially virilised 46,XY disorders of sex development. Eur J Endocrinol 172(6):745-751
  47. Paris F, Gaspari L, Mbou F, Philibert P, Audran F, Morel Y, Biason-Lauber A, Sultan C 2016 Endocrine and molecular investigations in a cohort of 25 adolescent males with prominent/persistent pubertal gynecomastia. Andrology 4(2):263-269
  48. Doehnert U, Bertelloni S, Werner R, Dati E, Hiort O 2015 Characteristic features of reproductive hormone profiles in late adolescent and adult females with complete androgen insensitivity syndrome. Sex Dev 9(2):69-74
  49. Choong CS, Quigley CA, French FS, Wilson EM 1996 A novel missense mutation in the amino-terminal domain of the human androgen receptor gene in a family with partial androgen insensitivity syndrome causes reduced efficiency of protein translation. J Clin Invest 98(6):1423-1431
  50. McPhaul MJ, Marcelli M, Tilley WD, Griffin JE, Isidro-Gutierrez RF, Wilson JD 1991 Molecular basis of androgen resistance in a family with a qualitative abnormality of the androgen receptor and responsive to high-dose androgen therapy. J Clin Invest 87(4):1413-1421
  51. Dougan GC, Uli N, Shulman DI 2014 Progressive central puberty in a toddler with partial androgen insensitivity. J Pediatr 164(3):655-657
  52. de Silva KS, Sirisena ND, Wijenayaka HK, Cooray JG, Jayasekara RW, Dissanayake VH 2015 Androgen insensitivity syndrome in a cohort of sri lankan children with 46, XY disorders of sex development (46, XY DSD). Ceylon Med J 60(4):139-142
  53. Lobaccaro JM, Poujol N, Chiche L, Lumbroso S, Brown TR, Sultan C 1996 Molecular modeling and in vitro investigations of the human androgen receptor DNA-binding domain: Application for the study of two mutations. Mol Cell Endocrinol 116(2):137-147
  54. Bruggenwirth HT, Boehmer AL, Lobaccaro JM, Chiche L, Sultan C, Trapman J, Brinkmann AO 1998 Substitution of Ala564 in the first zinc cluster of the deoxyribonucleic acid (DNA)-binding domain of the androgen receptor by asp, asn, or leu exerts differential effects on DNA binding. Endocrinology 139(1):103-110
  55. Nguyen D, Steinberg SV, Rouault E, Chagnon S, Gottlieb B, Pinsky L, Trifiro M, Mader S 2001 A G577R mutation in the human AR P box results in selective decreases in DNA binding and in partial androgen insensitivity syndrome. Mol Endocrinol 15(10):1790-1802
  56. Wooster R, Mangion J, Eeles R, Smith S, Dowsett M, Averill D, Barrett-Lee P, Easton DF, Ponder BA, Stratton MR 1992 A germline mutation in the androgen receptor gene in two brothers with breast cancer and reifenstein syndrome. Nat Genet 2(2):132-134
  57. Lobaccaro JM, Lumbroso S, Belon C, Galtier-Dereure F, Bringer J, Lesimple T, Heron JF, Pujol H, Sultan C 1993 Male breast cancer and the androgen receptor gene. Nat Genet 5(2):109-110
  58. Gast A, Neuschmid-Kaspar F, Klocker H, Cato AC 1995 A single amino acid exchange abolishes dimerization of the androgen receptor and causes reifenstein syndrome. Mol Cell Endocrinol 111(1):93-98
  59. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, Gass P, Schmid W, Herrlich P, Angel P, Schutz G 1998 DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93(4):531-541
  60. Giwercman YL, Ivarsson SA, Richthoff J, Lundin KB, Giwercman A 2004 A novel mutation in the D-box of the androgen receptor gene (S597R) in two unrelated individuals is associated with both normal phenotype and severe PAIS. Horm Res 61(2):58-62
  61. Pinsky L, Trifiro M, Kaufman M, Beitel LK, Mhatre A, Kazemi-Esfarjani P, Sabbaghian N, Lumbroso R, Alvarado C, Vasiliou M 1992 Androgen resistance due to mutation of the androgen receptor. Clin Invest Med 15(5):456-472
  62. Lottrup G, Jorgensen A, Nielsen JE, Jorgensen N, Duno M, Vinggaard AM, Skakkebaek NE, Rajpert-De Meyts E 2013 Identification of a novel androgen receptor mutation in a family with multiple components compatible with the testicular dysgenesis syndrome. J Clin Endocrinol Metab 98(6):2223-2229
  63. Tordjman KM, Yaron M, Berkovitz A, Botchan A, Sultan C, Lumbroso S 2014 Fertility after high-dose testosterone and intracytoplasmic sperm injection in a patient with androgen insensitivity syndrome with a previously unreported androgen receptor mutation. Andrologia 46(6):703-706
  64. Petroli RJ, Hiort O, Struve D, Maciel-Guerra AT, Guerra-Junior G, Palandi de Mello M, Werner R 2014 Preserved fertility in a patient with gynecomastia associated with the p.Pro695Ser mutation in the androgen receptor. Sex Dev 8(6):350-355
  65. Li Y, Qu S, Li P 2015 A novel mutation of the androgen receptor gene in familial complete androgen insensitivity syndrome. Eur Rev Med Pharmacol Sci 19(21):4146-4152
  66. Rajender S, Gupta NJ, Chakrabarty B, Singh L, Thangaraj K 2013 L712V mutation in the androgen receptor gene causes complete androgen insensitivity syndrome due to severe loss of androgen function. Steroids 78(12-13):1288-1292
  67. Franasiak JM, Yao X, Ashkinadze E, Rosen T, Scott RT,Jr 2015 Discordant embryonic aneuploidy testing and prenatal ultrasonography prompting androgen insensitivity syndrome diagnosis. Obstet Gynecol 125(2):383-386
  68. Nam H, Kim CH, Cha MY, Kim JM, Kang BM, Yoo HW 2015 Polycystic ovary syndrome woman with heterozygous androgen receptor gene mutation who gave birth to a child with androgen insensitivity syndrome. Obstet Gynecol Sci 58(2):179-182
  69. Akcay T, Fernandez-Cancio M, Turan S, Guran T, Audi L, Bereket A 2014 AR and SRD5A2 gene mutations in a series of 51 turkish 46,XY DSD children with a clinical diagnosis of androgen insensitivity. Andrology 2(4):572-578
  70. Mazen I, Soliman H, El-Gammal M, Torky A, Mekkawy M, Abdel-Hamid MS, Essawi M 2014 A novel mutation (c.2735_2736delTC) in the androgen receptor gene in 46,XY females with complete androgen insensitivity syndrome in an egyptian family. Horm Res Paediatr 82(6):411-414
  71. Hiort O, Sinnecker GH, Holterhus PM, Nitsche EM, Kruse K 1998 Inherited and de novo androgen receptor gene mutations: Investigation of single-case families. J Pediatr 132(6):939-943
  72. Lundberg Giwercman Y, Nikoshkov A, Lindsten K, Bystrom B, Pousette A, Chibalin AV, Arvidsson S, Tiulpakov A, Semitcheva TV, Peterkova V, Hagenfeldt K, Ritzen EM, Wedell A 1998 Functional characterisation of mutations in the ligand-binding domain of the androgen receptor gene in patients with androgen insensitivity syndrome. Hum Genet 103(4):529-531
  73. Umar A, Berrevoets CA, Van NM, van Leeuwen M, Verbiest M, Kleijer WJ, Dooijes D, Grootegoed JA, Drop SL, Brinkmann AO 2005 Functional analysis of a novel androgen receptor mutation, Q902K, in an individual with partial androgen insensitivity. J Clin Endocrinol Metab 90(1):507-515
  74. Wong HY, Hoogerbrugge JW, Pang KL, van Leeuwen M, van Royen ME, Molier M, Berrevoets CA, Dooijes D, Dubbink HJ, van de Wijngaart DJ, Wolffenbuttel KP, Trapman J, Kleijer WJ, Drop SL, Grootegoed JA, Brinkmann AO 2008 A novel mutation F826L in the human androgen receptor in partial androgen insensitivity syndrome; increased NH2-/COOH-terminal domain interaction and TIF2 co-activation. Mol Cell Endocrinol 292(1-2):69-78
  75. Elfferich P, Juniarto AZ, Dubbink HJ, van Royen ME, Molier M, Hoogerbrugge J, Houtsmuller AB, Trapman J, Santosa A, de Jong FH, Drop SL, Faradz SM, Bruggenwirth H, Brinkmann AO 2009 Functional analysis of novel androgen receptor mutations in a unique cohort of indonesian patients with a disorder of sex development. Sex Dev 3(5):237-244
  76. Tadokoro R, Bunch T, Schwabe JW, Hughes IA, Murphy JC 2009 Comparison of the molecular consequences of different mutations at residue 754 and 690 of the androgen receptor (AR) and androgen insensitivity syndrome (AIS) phenotype. Clin Endocrinol (Oxf) 71(2):253-260
  77. Cools M, Wolffenbuttel KP, Drop SL, Oosterhuis JW, Looijenga LH 2011 Gonadal development and tumor formation at the crossroads of male and female sex determination. Sex Dev 5(4):167-180
  78. Ris-Stalpers C, Verleun-Mooijman MC, de Blaeij TJ, Degenhart HJ, Trapman J, Brinkmann AO 1994 Differential splicing of human androgen receptor pre-mRNA in X-linked reifenstein syndrome, because of a deletion involving a putative branch site. Am J Hum Genet 54(4):609-617
  79. Akin JW, Behzadian A, Tho SP, McDonough PG 1991 Evidence for a partial deletion in the androgen receptor gene in a phenotypic male with azoospermia. Am J Obstet Gynecol 165(6 Pt 1):1891-1894
  80. Ris-Stalpers C, Kuiper GG, Faber PW, Schweikert HU, van Rooij HC, Zegers ND, Hodgins MB, Degenhart HJ, Trapman J, Brinkmann AO 1990 Aberrant splicing of androgen receptor mRNA results in synthesis of a nonfunctional receptor protein in a patient with androgen insensitivity. Proc Natl Acad Sci U S A 87(20):7866-7870
  81. Ahmed SF, Cheng A, Dovey L, Hawkins JR, Martin H, Rowland J, Shimura N, Tait AD, Hughes IA 2000 Phenotypic features, androgen receptor binding, and mutational analysis in 278 clinical cases reported as androgen insensitivity syndrome. J Clin Endocrinol Metab 85(2):658-665
  82. Yong EL, Chua KL, Yang M, Roy A, Ratnam S 1994 Complete androgen insensitivity due to a splice-site mutation in the androgen receptor gene and genetic screening with single-stranded conformation polymorphism. Fertil Steril 61(5):856-862
  83. Avila DM, Wilson CM, Nandi N, Griffin JE, McPhaul MJ 2002 Immunoreactive AR and genetic alterations in subjects with androgen resistance and undetectable AR levels in genital skin fibroblast ligand-binding assays. J Clin Endocrinol Metab 87(1):182-188
  84. Trifiro MA, Lumbroso R, Beitel LK, Vasiliou DM, Bouchard J, Deal C, Van Vliet G, Pinsky L 1997 Altered mRNA expression due to insertion or substitution of thymine at position +3 of two splice-donor sites in the androgen receptor gene. Eur J Hum Genet 5(1):50-58
  85. Infante JB, Alvelos MI, Bastos M, Carrilho F, Lemos MC 2016 Complete androgen insensitivity syndrome caused by a novel splice donor site mutation and activation of a cryptic splice donor site in the androgen receptor gene. J Steroid Biochem Mol Biol 155(Pt A):63-66
  86. Bruggenwirth HT, Boehmer AL, Ramnarain S, Verleun-Mooijman MC, Satijn DP, Trapman J, Grootegoed JA, Brinkmann AO 1997 Molecular analysis of the androgen-receptor gene in a family with receptor-positive partial androgen insensitivity: An unusual type of intronic mutation. Am J Hum Genet 61(5):1067-1077
  87. Jaaskelainen J, Mongan NP, Harland S, Hughes IA 2006 Five novel androgen receptor gene mutations associated with complete androgen insensitivity syndrome. Hum Mutat 27(3):291
  88. Kerkhofs S, Dubois V, De Gendt K, Helsen C, Clinckemalie L, Spans L, Schuit F, Boonen S, Vanderschueren D, Saunders PT, Verhoeven G, Claessens F 2012 A role for selective androgen response elements in the development of the epididymis and the androgen control of the 5alpha reductase II gene. FASEB J 26(10):4360-4372
  89. Zhu YS, Katz MD, Imperato-McGinley J 1998 Natural potent androgens: Lessons from human genetic models. Baillieres Clin Endocrinol Metab 12(1):83-113
  90. Sinnecker GH, Hiort O, Dibbelt L, Albers N, Dorr HG, Hauss H, Heinrich U, Hemminghaus M, Hoepffner W, Holder M, Schnabel D, Kruse K 1996 Phenotypic classification of male pseudohermaphroditism due to steroid 5 alpha-reductase 2 deficiency. Am J Med Genet 63(1):223-230
  91. Forti G, Falchetti A, Santoro S, Davis DL, Wilson JD, Russell DW 1996 Steroid 5 alpha-reductase 2 deficiency: Virilization in early infancy may be due to partial function of mutant enzyme. Clin Endocrinol (Oxf) 44(4):477-482
  92. Katz MD, Kligman I, Cai LQ, Zhu YS, Fratianni CM, Zervoudakis I, Rosenwaks Z, Imperato-McGinley J 1997 Paternity by intrauterine insemination with sperm from a man with 5alpha-reductase-2 deficiency. N Engl J Med 336(14):994-997
  93. Matsubara K, Iwamoto H, Yoshida A, Ogata T 2010 Semen analysis and successful paternity by intracytoplasmic sperm injection in a man with steroid 5alpha-reductase-2 deficiency. Fertil Steril 94(7):2770.e7-2770.10
  94. Kang HJ, Imperato-McGinley J, Zhu YS, Cai LQ, Schlegel P, Palermo G, Rosenwaks Z 2011 The first successful paternity through in vitro fertilization-intracytoplasmic sperm injection with a man homozygous for the 5alpha-reductase-2 gene mutation. Fertil Steril 95(6):2125.e5-2125.e8
  95. Canto P, Vilchis F, Chavez B, Mutchinick O, Imperato-McGinley J, Perez-Palacios G, Ulloa-Aguirre A, Mendez JP 1997 Mutations of the 5 alpha-reductase type 2 gene in eight mexican patients from six different pedigrees with 5 alpha-reductase-2 deficiency. Clin Endocrinol (Oxf) 46(2):155-160
  96. Hiort O, Sinnecker GH, Willenbring H, Lehners A, Zollner A, Struve D 1996 Nonisotopic single strand conformation analysis of the 5 alpha-reductase type 2 gene for the diagnosis of 5 alpha-reductase deficiency. J Clin Endocrinol Metab 81(9):3415-3418
  97. Boudon C, Lobaccaro JM, Lumbroso S, Ogur G, Ocal G, Belon C, Sultan C 1995 A new deletion of the 5 alpha-reductase type 2 gene in a turkish family with 5 alpha-reductase deficiency. Clin Endocrinol (Oxf) 43(2):183-188
  98. Vilchis F, Mendez JP, Canto P, Lieberman E, Chavez B 2000 Identification of missense mutations in the SRD5A2 gene from patients with steroid 5alpha-reductase 2 deficiency. Clin Endocrinol (Oxf) 52(3):383-387
  99. Silver RI, Rodriguez R, Chang TS, Gearhart JP 1999 In vitro fertilization is associated with an increased risk of hypospadias. J Urol 161(6):1954-1957
  100. Anwar R, Gilbey SG, New JP, Markham AF 1997 Male pseudohermaphroditism resulting from a novel mutation in the human steroid 5 alpha-reductase type 2 gene (SRD5A2). Mol Pathol 50(1):51-52
  101. Makridakis N, Ross RK, Pike MC, Chang L, Stanczyk FZ, Kolonel LN, Shi CY, Yu MC, Henderson BE, Reichardt JK 1997 A prevalent missense substitution that modulates activity of prostatic steroid 5alpha-reductase. Cancer Res 57(6):1020-1022
  102. Mazen I, Gad YZ, Hafez M, Sultan C, Lumbroso S 2003 Molecular analysis of 5alpha-reductase type 2 gene in eight unrelated egyptian children with suspected 5alpha-reductase deficiency: Prevalence of the G34R mutation. Clin Endocrinol (Oxf) 58(5):627-631
  103. Sasaki G, Nakagawa K, Hashiguchi A, Hasegawa T, Ogata T, Murai M 2003 Giant seminoma in a patient with 5 alpha-reductase type 2 deficiency. J Urol 169(3):1080-1081
  104. Hiort O, Schutt SM, Bals-Pratsch M, Holterhus PM, Marschke C, Struve D 2002 A novel homozygous disruptive mutation in the SRD5A2-gene in a partially virilized patient with 5alpha-reductase deficiency. Int J Androl 25(1):55-58
  105. Hafez M, Mazen I, Ghali I, Sultan C, Lumbroso S 2003 A new mutation of 5-alpha-reductase type 2 (A62E) in a large egyptian kindred. Horm Res 59(6):281-284
  106. Bahceci M, Ersay AR, Tuzcu A, Hiort O, Richter-Unruh A, Gokalp D 2005 A novel missense mutation of 5-alpha reductase type 2 gene (SRD5A2) leads to severe male pseudohermaphroditism in a turkish family. Urology 66(2):407-410
  107. Bertelloni S, Scaramuzzo RT, Parrini D, Baldinotti F, Tumini S, Ghirri P 2007 Early diagnosis of 5alpha-reductase deficiency in newborns. Sex Dev 1(3):147-151
  108. Baldinotti F, Majore S, Fogli A, Marrocco G, Ghirri P, Vuerich M, Tumini S, Boscherini B, Vetri M, Scommegna S, Rinaldi R, Simi P, Grammatico P 2008 Molecular characterization of 6 unrelated italian patients with 5alpha-reductase type 2 deficiency. J Androl 29(1):20-28
  109. Chan AO, But BW, Lau GT, Lam AL, Ng KL, Lam YY, Lee CY, Shek CC 2009 Diagnosis of 5alpha-reductase 2 deficiency: A local experience. Hong Kong Med J 15(2):130-135
  110. Sahakitrungruang T, Wacharasindhu S, Yeetong P, Snabboon T, Suphapeetiporn K, Shotelersuk V 2008 Identification of mutations in the SRD5A2 gene in thai patients with male pseudohermaphroditism. Fertil Steril 90(5):2015.e11-2015.e15
  111. Sahu R, Boddula R, Sharma P, Bhatia V, Greaves R, Rao S, Desai M, Wakhlu A, Phadke S, Shukla M, Dabadghao P, Mehrotra RN, Bhatia E 2009 Genetic analysis of the SRD5A2 gene in indian patients with 5alpha-reductase deficiency. J Pediatr Endocrinol Metab 22(3):247-254
  112. Vilchis F, Valdez E, Ramos L, Garcia R, Gomez R, Chavez B 2008 Novel compound heterozygous mutations in the SRD5A2 gene from 46,XY infants with ambiguous external genitalia. J Hum Genet 53(5):401-406
  113. Kim SH, Kim KS, Kim GH, Kang BM, Yoo HW 2006 A novel frameshift mutation in the 5alpha-reductase type 2 gene in korean sisters with male pseudohermaphroditism. Fertil Steril 85(3):750.e9-750.e12
  114. Di Marco C, Bulotta AL, Varetti C, Dosa L, Michelucci A, Baldinotti F, Meucci D, Castagnini C, Lo Rizzo C, Di Maggio G, Simi P, Mari F, Bertelloni S, Renieri A, Messina M 2013 Ambiguous external genitalia due to defect of 5-alpha-reductase in seven iraqi patients: Prevalence of a novel mutation. Gene 526(2):490-493
  115. Zhang M, Yang J, Zhang H, Ning G, Li X, Sun S 2011 A novel SRD5A2 mutation with loss of function identified in chinese patients with hypospadias. Horm Res Paediatr 76(1):44-49
  116. Berra M, Williams EL, Muroni B, Creighton SM, Honour JW, Rumsby G, Conway GS 2011 Recognition of 5alpha-reductase-2 deficiency in an adult female 46XY DSD clinic. Eur J Endocrinol 164(6):1019-1025
  117. Maimoun L, Philibert P, Cammas B, Audran F, Bouchard P, Fenichel P, Cartigny M, Pienkowski C, Polak M, Skordis N, Mazen I, Ocal G, Berberoglu M, Reynaud R, Baumann C, Cabrol S, Simon D, Kayemba-Kay's K, De Kerdanet M, Kurtz F, Leheup B, Heinrichs C, Tenoutasse S, Van Vliet G, Gruters A, Eunice M, Ammini AC, Hafez M, Hochberg Z, Einaudi S, Al Mawlawi H, Nunez CJ, Servant N, Lumbroso S, Paris F, Sultan C 2011 Phenotypical, biological, and molecular heterogeneity of 5alpha-reductase deficiency: An extensive international experience of 55 patients. J Clin Endocrinol Metab 96(2):296-307
  118. Maimoun L, Philibert P, Bouchard P, Ocal G, Leheup B, Fenichel P, Servant N, Paris F, Sultan C 2011 Primary amenorrhea in four adolescents revealed 5alpha-reductase deficiency confirmed by molecular analysis. Fertil Steril 95(2):804.e1-804.e5
  119. Maimoun L, Philibert P, Cammas B, Audran F, Pienkowski C, Kurtz F, Heinrich C, Cartigny M, Sultan C 2010 Undervirilization in XY newborns may hide a 5alpha-reductase deficiency: Report of three new SRD5A2 gene mutations. Int J Androl 33(6):841-847
  120. Fenichel P, Paris F, Philibert P, Hieronimus S, Gaspari L, Kurzenne JY, Chevallier P, Bermon S, Chevalier N, Sultan C 2013 Molecular diagnosis of 5alpha-reductase deficiency in 4 elite young female athletes through hormonal screening for hyperandrogenism. J Clin Endocrinol Metab 98(6):E1055-9
  121. Nie M, Zhou Q, Mao J, Lu S, Wu X 2011 Five novel mutations of SRD5A2 found in eight chinese patients with 46,XY disorders of sex development. Mol Hum Reprod 17(1):57-62
  122. Savas Erdeve S, Aycan Z, Berberoglu M, Siklar Z, Hacihamdioglu B, Sipahi K, Akar N, Ocal G 2010 A novel mutation of 5alpha-steroid reductase 2 deficiency (CD 65 ALA-PRO) with severe virilization defect in a turkish family and difficulty in gender assignment. Eur J Pediatr 169(8):991-995
  123. Tsai MC, Chou YY, Lin SJ, Tsai LP 2012 A novel SRD5A2 mutation in a taiwanese newborn with ambiguous genitalia. Kaohsiung J Med Sci 28(4):231-235
  124. Shabir I, Khurana ML, Marumudi E, Khadgawat R, Ammini AC 2013 Novel nucleotide insertions in two unrelated indian patients with 5alpha reductase 2 deficiency leading to premature termination of SRD5A2 enzyme. Steroids 78(12-13):1159-1163
  125. Shabir I, Khurana ML, Joseph AA, Eunice M, Mehta M, Ammini AC 2015 Phenotype, genotype and gender identity in a large cohort of patients from india with 5alpha-reductase 2 deficiency. Andrology 3(6):1132-1139
  126. Zhu H, Liu W, Han B, Fan M, Zhao S, Wang H, Lu Y, Pan C, Chen F, Chen M, Song H, Cheng K, Qiao J 2014 Phenotypic and molecular characteristics in eleven chinese patients with 5alpha-reductase type 2 deficiency. Clin Endocrinol (Oxf) 81(5):711-720
  127. Makridakis NM, Ross RK, Pike MC, Crocitto LE, Kolonel LN, Pearce CL, Henderson BE, Reichardt JK 1999 Association of mis-sense substitution in SRD5A2 gene with prostate cancer in african-american and hispanic men in los angeles, USA. Lancet 354(9183):975-978
  128. Makridakis N, Akalu A, Reichardt JK 2004 Identification and characterization of somatic steroid 5alpha-reductase (SRD5A2) mutations in human prostate cancer tissue. Oncogene 23(44):7399-7405
  129. Makridakis NM, di Salle E, Reichardt JK 2000 Biochemical and pharmacogenetic dissection of human steroid 5 alpha-reductase type II. Pharmacogenetics 10(5):407-413
  130. Wang C, Tao W, Chen Q, Hu H, Wen XY, Han R 2010 SRD5A2 V89L polymorphism and prostate cancer risk: A meta-analysis. Prostate 70(2):170-178

 

Treatment of Diabetes mellitus in Children and Adolescents

ABSTRACT

The incidence of Type 1 Diabetes continues to increase around the world. Advances in technology of insulin delivery systems including closed loop and continuous glucose monitoring are improving the possibilities of maintaining desirable glucose control. Type 2 Diabetes is increasing in the adolescent age groups across the world, in certain populations especially including Native Americans, Pacific Islanders, Hispanics, African Americans, and South East Asians. For Type 2 Diabetes, the pharmacological armamentarium has markedly increased by the addition of GLP-1 agonists, DPP4 antagonists, and SGLT2 inhibitors, each of which has improved metabolic control and cardiovascular outcomes. To date, these newer modalities are being tested in adolescents with T2DM but several are not yet officially approved for this age group. Diabetic Ketoacidosis (DKA) remains the initial presentation of some 30%-40% of pediatric patients, and DKA remains the leading cause of death, sometimes associated with Cerebral Edema; complications are also very high in children/adolescents presenting with Hyperglycemia/Hyperosmolar syndrome in the context of a T2DM clinical picture. Appropriate treatment in medical centers with trained personnel and modern laboratory facilities has markedly reduced the mortality and morbidity associated with DKA and Hyperglycemic-Hyperosmolar Syndrome.

 

DIABETIC KETOACIDOSIS

 

Pathophysiology

 

Diabetic ketoacidosis (DKA) is a life-threatening metabolic decompensation considered to be a medical emergency and caused by a combination of insulin deficiency and the action of counter-regulatory hormones(1). The biochemical, metabolic and acid-base abnormalities that occur have been extensively documented at a physiologic level and to some extent at a molecular level (2-4).  Briefly, deficiency of insulin prevents the entry of glucose into insulin-sensitive cells in tissues such as liver, muscle, and fat and its appropriate metabolism. Sensing intra-cellular glucopenia, the organism responds by increased secretion of the 4 counter-regulatory hormones, glucagon, cortisol, growth hormone, and catecholamines. Acting synergistically, these hormones increase glucose production via glycogen breakdown and gluconeogenesis, induce lipolysis and ketogenesis and result in hyperglycemia, osmotic polyuria, dehydration, increased thirst, and acidosis from the accumulation of ketoacids, principally β-hydroxybutyrate, (B-OHB) which exceed buffering capacity, as well as lactic acidosis from the ensuing dehydration and limited tissue perfusion. Hence, the symptoms and signs are polyuria, polydipsia, dehydration, tachycardia, deep sighing respiration (Kussmaul breathing), and the smell of acetone on the breath, and abdominal pain and nausea imitating an acute abdominal condition; paradoxically, despite dehydration, blood pressure may be normal or elevated reflecting the effects of catecholamines (Table 1). These manifestations develop over hours or days, in contrast to hypoglycemia which can occur suddenly. In cases of new diabetes, weight loss, increased appetite, and nocturia, or enuresis in previously toilet-trained child, are almost universally present if a careful history is elicited. Left untreated, clouding of consciousness due decreased cerebral oxygen perfusion, acidosis, and neural biochemical changes lead to coma and eventually death. Absolute insulin deficiency occurs most often at onset of evolving T1DM, but it may also occur after deliberate or inadvertent omission of insulin in a child or adolescent responsible for their own care, or with kinking or obstruction of tubing in insulin pumps. Relative insulin deficiency occurs with major physiological stressors such as sepsis, infection, or severe trauma that result in profound increased secretion of the counter-regulatory hormones which overwhelm the actions of insulin. Recurrent episodes of DKA are almost the result of psycho-social mal-adjustment. These concepts are summarized in figure 1.

 

TABLE 1. Clinical and Biochemical Manifestations of Diabetic Ketoacidosis

Clinical

Biochemical

Dehydration

Hyperglycemia (11-50mmol/l)

Rapid, deep, sighing (Kussmaul respiration)

Variable degrees of acidosis (PH<7.3; HCO3 <15meq/l)

Nausea, vomiting, and abdominal pain mimicking an acute abdomen

Ketosis-serum BOHB (>3mmol/l)

Progressive obtundation and loss of consciousness

Elevation of BUN and Creatinine

Increased leukocyte count with left shift

Fever only when infection is present

Non-specific elevation of serum amylase

Figure 1. Pathophysiology of diabetic ketoacidosis(7). Copyright © 2006 American Diabetes Association. From Diabetes Care, Vol. 29, 2006:1150-1159. Reprinted with permission of The American Diabetes Association

Criteria for Defining DKA

 

The criteria for a diagnosis of DKA are hyperglycemia with glucose ≥200mg/dl (≥11mmol/l), pH ≤7.30 or bicarbonate (HCO3) ≤15mmol/l, and ketonuria or B-OHB ≥2.0mmol/l. Severity of DKA is defined by the degree of acidosis; mild=pH 7.20-7.30, HCO3 10-15mmol/l; moderate=pH 7.1-7.2, HCO3 5-10mmol/l; severe =pH<7.1, HCO3<5 mmol/l. Hyperglycemia usually ranges between 200-1000mg/dl (16.6-50.5mmol/l);values >1000mg/dl should raise the possibility of hyperosmolar hyperglycemia, separately discussed below. The reported frequency of DKA varies from about 13% to 80% in various countries and is generally higher in less developed countries; it is inversely related to socio-economic development, level of education of the family, and to the incidence of diabetes mellitus in the location. In the United States, about 30-40% of newly diagnosed patients with DM present in DKA, reflecting delay in establishing the diagnosis of diabetes in a child, particularly in children <5years of age (8-11).

 

Table 2 illustrates the average losses of fluids and electrolytes in diabetic ketoacidosis and maintenance requirements in normal children adapted from references (4, 7, 9).

 

TABLE 2.  Fluid and Electrolyte Losses and Maintenance Requirements in DKA

 

Average (range) losses

24-hour maintenance requirements

Water

70 mL/kg (30-100)

≤10 kg: 100 mL/kg/24hrs

11-20 kg: 1000 mL+ 50 mL/kg/24 hr for each kg from 11-20

>20 kg: 1500 mL+ 20 mL/kg/24 hr for each kg >20

Sodium

6 mmol/kg (5-13)

2-4 mmol/kg

Potassium

5 mmol/kg (3-6)

2-3 mmol/kg

Chloride

4 mmol/kg (3-9)

2-3 mmol/kg

Phosphate

1 mmol/kg (0.5-2.5)

1-2 mmol/kg

 

Principles of Treatment  

 

The principles of treatment enunciated here are based on those of the International Society for Pediatric and Adolescent Diabetes, the American Diabetes Association, and the Pediatric Endocrine Societies of Europe and the USA (4, 7, 11).

 

Mild cases of DKA such as might occur in a patient using an insulin pump in which the tubing has become obstructed, or mild upper respiratory or mild abdominal infection without significant vomiting or diarrhea in an educated patient and strong family support might be managed via telephone instructions. When fluids are tolerated, 3-4 ounces of clear fluids (approximately 100ml) can be offered hourly. In addition, rapid acting insulin 0.1-0.2U/kg is given every 2-4 hours, glucose levels are checked via home meters and urinary ketones are checked via strips. Resolution of hyperglycemia and ketonuria, and tolerance to oral fluid intake indicates successful management and return to customary regimens including pump settings and/or subcutaneous basal-bolus insulin regimens.

 

For new onset patients, those that cannot tolerate oral fluid intake, and those with moderate to severe DKA, we recommend admission to a unit with capabilities similar to those of an ICU, possessing written guidelines on management, physicians and nursing staff trained in the management of DKA, bedside glucose and blood gas monitoring, vital sign monitoring (pulse, blood pressure, respiration), and laboratory back-up of acid-base and electrolyte status. A thorough physical examination including level of consciousness should determine the overall clinical status, degree of dehydration, and consider the need to evaluate for infection. Supplemental oxygen may be provided via mask or nasal cannula, and a gastric tube passed if the patient is vomiting. Urine output should be measured via bag collection and catheterization avoided if possible.  A blood sample should be obtained for measurement of glucose, electrolytes, β-OHB, acid-base status, hematocrit, and complete blood count; blood cultures and imaging studies should be considered in cases of suspected sepsis as the precipitating cause and appropriate antibiotics given. A modest increase in WBC with neutrophil predominance may reflect an acute phase response rather than sepsis; even if an acute abdominal condition is suspected, surgery should be deferred until several hours of resuscitation with fluids and electrolytes has occurred. Access for IV infusion should be established; this or preferably a separate IV site can serve as source of blood sampling.

 

The initial resuscitation consists of intravenous saline bolus infusion at 10-20 ml/kg over 1-2 hours depending on the degree of dehydration. Clinical assessment of dehydration is based on physical findings such as heart rate, blood pressure, speed of capillary refill, tissue turgor, and dry coated tongue and is generally rated as 5% (mild),10% (moderate) or greater than 10% (severe). Urine output is not a reliable sign as it usually continues due to the osmotic diuresis of hyperglycemia; diminished urine output may reflect evolving renal failure. Clinical judgment of degree of dehydration is notoriously inaccurate and can result in over or under estimation; hematocrit or a very recent weight may aid in assessment of the degree of dehydration so as to more accurately guide the amount and composition of fluids to be infused and in turn reduce the risk of the occurrence of cerebral edema (12, 13). Hence initial estimates of dehydration and osmolality of plasma based on glucose and electrolyte status, as well as acid base resolution, require re-assessment as treatment progresses. The initial resuscitation period with normal (0.9%) sodium chloride solution provides an opportunity to elicit a careful history and formulate the plan of management focused on provision of fluid, electrolytes, insulin, and monitoring to anticipate and correct complications.

 

FLUID

 

The amount of fluid to be administered is based on the estimated degree of dehydration, e.g., 5% of body weight in Kg, plus daily maintenance (see table 2) evenly infused over 24-36 hours, subtracting the fluid administered as resuscitation. For example, a 30kg child with estimated dehydration of 5% would require 1500 ml for deficit plus 1700 ml for daily maintenance, yields a total of 3200 ml (table2); subtracting the 20ml/kg bolus of normal 0.9% saline from this total (600ml), leaves 2600 ml, or approximately 100 ml/hour over the initial 24 hours with adjustments made according to response. The total daily fluid infused should rarely exceed 1.5-2.0 times daily maintenance.

 

COMPOSITION OF FLUID

 

Sodium Chloride

 

Normal isotonic saline is the initial crystalloid of choice to be given over the initial 4-6 hours. This fluid is hypotonic relative to the osmolality of the patient’s plasma which can be calculated as 2(Na meq/dl + K meq/dl) plus glucose in mmol, or mg/dl divided by 18. Assuming a Na of 140meq, K 4.0meq and glucose of 450 mg/dl, the osmolality is 284+25 = 309 mosm. Normal (0.9%) saline has an osmolality of 286meq/l; the difference between the osmolality of the infused saline and patient’s plasma becomes greater as the glucose concentration in plasma rises. But it is important to note that the infusate remains hypotonic relative to plasma as long as the hyperglycemia persists; decline in plasma osmolality must be carefully monitored to avoid rapid osmotic shifts that facilitate entry of water to the intracellular /intracerebral compartment. After the first 4-6 hours, 0.5-0.75 N saline plus added potassium(K) as the phosphate, acetate or chloride maintains an osmolality of the infusate close to that of the patient’s plasma. Because of the concerns regarding use of chloride in worsening acidosis, some have recommended the use of lactated Ringers solution or sodium acetate in lieu of normal saline (7).

 

Potassium

 

During acidosis K moves from the intra cellular to the extracellular compartment and considerable K is then lost in urine. As a result, total body K stores are almost always depleted and with correction of acidosis, K returns to the intracellular compartment resulting in hypokalemia, which may precipitate cardiac arrhythmia. Hyperkalemia is less common and may reflect impaired renal function. Hence, after initial resuscitation, as soon as urine output is documented, K should be added to the infusate at a concentration of 20-40 meq/l. The potassium may be in the form of potassium chloride, but this adds to the hyperchloremia which may result in persistent hyperchloremic acidosis. Hence, some recommend that the K may be administered, at least in part, as the acetate or phosphate, which may have additional benefit as described below. Total amounts of potassium replacement should not exceed 0.5mm/kg/hour. Potassium replacement should continue throughout the period of IV therapy to assist in the repletion of potassium stores. This may not be fully accomplished during IV therapy and continues when oral intake is resumed. The measurement of K concentration is an essential component of biochemical monitoring as described below; additional rapid monitoring of K concentration in plasma is in the evaluation of the EKG which may show high peaked T waves with hyperkalemia and low amplitude of T waves, T wave inversion, prolonged PR interval and prominent U waves with hypokalemia. 

 

Phosphate

 

As with potassium, phosphate stores are depleted in ketoacidosis and further losses occur with ongoing diuresis during treatment and the effects of insulin in promoting intracellular entry. Severe hypophosphatemia (<1mg/dl) may be associated with depletion of ATP and the resultant deleterious effects on any energy requiring processes, including muscle function, CNS disturbances, hemolysis, and rhabdomyolysis. In addition, phosphate participates in the regulation of the oxygen dissociation curve, so depletion impairs oxygen release to tissues and further exacerbates acidosis by promoting lactate accumulation. On the other hand, infusing phosphate is associated with hypocalcemia and limited trials have not shown consistent beneficial effects in the treatment of DKA. An advantage however, is its cautious use in limiting hyperchloremia and hence acidosis by providing some of the potassium requirement as phosphate rather than chloride, alternating KCl with KPO4 and monitoring calcium concentration to avoid or treat hypocalcemia. We use this approach in our practice recommendations.

 

INSULIN THERAPY

 

Fluid therapy alone incompletely corrects many of the biochemical features of DKA, but full resolution of DKA requires insulin to switch off ketogenesis, restore acid-base balance, and resume anabolic processes. For moderate to severe acidosis, we recommend a starting dose of insulin(regular) at 0.1 U/Kg/hour, infused intravenously until acidosis is curtailed; insulin should be continued even if the blood glucose concentration has declined to ~300mg/dl or less and additional glucose provided as 5%-10% solution to maintain glucose at ~300mg/dl. Temporarily switching off the insulin infusion may result in rebound or persistence of acidosis, as insulin is essential to curtail keto-acid production and enable metabolism of keto-acids to bicarbonate. It is permissible to reduce the insulin dose to 0.05U/Kg/hr if there is difficulty in maintaining glucose at ~300mg/dl, even with additional glucose infusion, but insulin infusion should not stop until acidosis is resolved and pH is 7.3 or higher. In those admitted with mild acidosis, or those who administered basal insulin prior to admission, the starting dose of insulin should be 0.05U/Kg/hr, in order to avoid too rapid decline in the glucose concentration. Monitoring of blood glucose decline may require upward adjustment of the insulin dose if glucose is not declining at least 50 mg/dl/hour. An intravenous insulin bolus of insulin is not recommended to be given at the start of therapy and may not be effective as acidosis promotes dissociation of hormone binding to its cognate receptor. Where venous access is not possible, IM or SQ fast acting insulin (aspart or lispro) may be given at a starting dose of 0.2-0.3U/Kg and doses of 0.1-0.2 U/kg repeated 1-2 hours apart depending on response in terms of decline in glucose and correction of acidosis.

 

BICARBONATE THERAPY

 

In controlled trials in adults, bicarbonate therapy has not been effective in shortening the time of acidosis; bicarbonate actually may cause harm. Harm may occur because HCO3- combines with the H+ to form H2CO3 which dissociates to H2O and CO2.Whereas HCO3- does not cross the blood-brain barrier, CO2 diffuses readily across the blood-brain barrier and may exacerbate acidosis. In addition, large doses of bicarbonate may induce alkalosis and promote hypokalemia. Although controlled trials have not been performed in children, observational outcomes in pediatric studies show resolution of acidosis with provision of fluids and insulin; bicarbonate therapy is not recommended in published guideline (4, 7, 12). In severe acidosis, with pH <7.0, myocardial contractility may be impaired and here bicarbonate may be helpful. In these circumstances, bicarbonate may be infused at 1-2mmol/Kg over 60 minutes and acid -base status reassessed thereafter. Bicarbonate therapy may be useful in treatment of severe hyperkalemia. Bicarbonate must not be given as a bolus in treating DKA.

 

An example of the losses and management of DKA in a child with weight 30kg (Surface area 1M2) are shown in Tables 3 and 4.

 

TABLE 3. Fluid and Electrolyte losses Based on Assumed 7% Dehydration in a Child with Diabetic Ketoacidosis*

Fluid and electrolyte

Approximate accumulated losses with 7% dehydration

Approximate requirements for maintenance (36hrs)

Approximate working total

Water (mL)

2100

2550

4650

Sodium (mEq)

180

180

360

Potassium (mEq)

120

90

210

Chloride (mEq)

120

90

210

Phosphate (mEq)

30

45

75

*Weight 30 kg; surface area 1 M2; See tables 2 and 3, references (4, 7, 12) and text for source of losses of water and electrolytes       

 

TABLE 4. Replacement Therapy for a Child with Assumed 7% Dehydration and DKA

Duration

Fluid composition/amount

 

Sodium (mEq)

 

Chloride (mEq)

 

Potassium (mEq)

Phosphate (mEq)

 

Hour 0-2:

500 mL N. SALINE (0.9%NaCl)

75

 

75

 

0

0

Hour 2-6:

150mL/hr

INSULIN

0.1 U/kg/hr

600mL N. SALINE

+ 40mEq KCl/L

 

90

115

25

0

Hour 6-12:

150mL/hr

INSULIN

0.1 U/kg/hr

900ML 0.5N. SALINE +40mEq  KCl/L

 

~70

105

35

0

Subtotal: initial 12 hr

2000ML

 

235

295

60

0

Next 24 hr:

100mL/hr

INSULIN

0.1 U/kg/hr

2400 mL 0.5N SALINE

1STLITER ADD KPO4 40mEq

2ndLITER ADD KCl    20 mEq

3rdLITER ADD KPO4 20 mEq

 

75

 

75

 

30

 

75

 

95

 

30

 

40

 

20

 

8

 

40

 

0

 

8

Total 36 hr:

4400 ml

415

495

128

48

*Weight 30 kg; surface area 1 M2; In this formulation, calculated fluid deficit has been corrected by about 12 hours and basal requirement over the ensuing 24 hours; total fluid over the 36 hours has not exceeded 2 times daily maintenance. Total sodium infused only modestly exceeds the calculated deficit, but total chloride excess is considerable and may be associated with persistent (hyperchloremic) acidosis. Potassium and phosphate repletion is incomplete and continues after transition to oral intake of nutrition and subcutaneous insulin therapy. This example is for illustrative purposes only; the actual amount and composition of infused fluids is dictated by the biochemical responses monitored and recorded during therapy. Detailed discussion of electrolyte replacement can be found in references (12) and (13).

 

MONITORING

 

A flow sheet to record clinical and biochemical progress is an essential component of therapy. Actual real-time monitoring of vital signs should be complemented by hourly recordings. Initial chemical laboratory tests must include blood glucose, serum electrolytes with emphasis on sodium, chloride, and potassium, as well as phosphate, calcium, pH, pCO2, HCO3, base excess, BUN and creatinine as indices of renal function and β-hydroxybutyrate(B-OHB) as a measure of ketosis. Measurement of urine output, urine glucose and ketones also must be recorded. The urine ketone measurement uses the sodium nitroprusside reaction which measures aceto-acetic acid and weakly acetone, but not B-OHB, the predominant ketone in blood. Hence, the major contributor to ketoacidosis is not reflected in the urinary ketone measurement. Bedside blood glucose, electrolyte, and acid base, and ketone meters are very useful but must be verified by periodic formal laboratory measurements. Initially, hourly measurement of glucose, electrolytes, and acid base status are recommended for the first 4 hours and 2-4 hourly thereafter depending on indices of improvement and resolution of acidosis, defined as pH≥7.3 or bicarbonate ≥15 mm/l. At this time transition to oral intake and discontinuation of IV therapy can be undertaken; absence of ketonuria should not be a criterion as this may continue for some time due to conversion of B-OHB to aceto-acetate as ketosis resolves. After the first day, once daily measurement of electrolytes, acid base and renal function should be performed until restoration of normal function is confirmed.

 

TRANSITION TO ORAL INTAKE

 

Oral intake may be begun when clinical recovery has occurred even if the acid base status and ketonuria have not completely resolved. Oral sips of clear liquids precede the introduction of oral fluids to gradually supplant the IV provision and total daily fluid restricted to no more than 1.5 times calculated daily maintenance. The first dose of regular or fast acting insulin is given subcutaneously approximately 1-2 hours before discontinuing the IV insulin to allow for absorption. For patients on a basal-bolus insulin regimen, the first dose of basal insulin may be administered in the evening while the IV insulin is maintained till the morning and then discontinued.

 

Mortality and Morbidity of DKA

 

Mortality of DKA has declined markedly in the past 2 decades largely due to greater referral of patients to specialized centers (5, 6, 14). Cerebral edema (CE) is responsible for the majority of deaths and survivors of CE may have severe or mild residual impairment of CNS function including memory impairment (15-17). Several other causes of mortality and morbidity occur, but each is individually rare and include venous and arterial CNS thromboses, pulmonary embolus, rhabdomyolysis, pancreatitis, ARDS, and infections such as rhino-cerebral mucormycosis and other rare entities. These rarer complications are more fully described in prior reviews (4, 7, 16).

 

CEREBRAL EDEMA

 

Cerebral edema is the most feared complication of DKA occurring either early (cerebral ischemia/reperfusion injury) or later during the course of therapy; mechanisms have not been clearly defined and whether the composition of IV fluids and their rate of administration contribute to or may prevent this complication is hotly debated (12-19). New onset, younger age and indices of severity have been associated with greater risk of this complication (20). Symptoms and signs include severe headache and development of bradycardia and hypertension as evidence of raised intracranial pressure, restlessness and irritability, localizing neurological features such as nystagmus and incontinence or polyuria without glucosuria as indicators of evolving diabetes insipidus, as well as evidence of papilledema.  Clinical diagnosis based on bedside evaluation of neurological state as shown below has been proposed (16). In this formulation, one diagnostic criterion, two major criteria, or one major and two minor criteria have a sensitivity of 92% and a false positive rate of only 4% (16).  Signs that occur before treatment should not be included in the diagnosis of cerebral edema. Diagnostic criteria include abnormal motor or verbal response to pain; decorticate or decerebrate posture; cranial nerve palsy (especially III, IV, and VI), and abnormal neurogenic respiratory pattern such as grunting, or Cheyne-Stokes respiration. Major criteria include altered mentation/fluctuating level of consciousness; sustained heart rate deceleration (decrease more than 20 beats per minute) not attributable to improved intravascular volume or sleep state; and age-inappropriate incontinence with a rise in serum sodium indicative of loss of free water(diabetes insipidus).Minor criteria include vomiting, headache; lethargy or not easily arousable; diastolic BP >90 mm Hg; young age( <5 years) (16). The mechanisms responsible for the development of cerebral edema in DKA appear to be both osmotic (12-14, 18, 20) and vasogenic (21, 22), and the timing of appearance as early or late in the course of treatment may depend in part on the major contribution of the mechanism involved. Treatment should begin with reduction in the rate of fluid administration, elevating the head of the bed, administration of mannitol, 0.5-1 g/kg IV over 10-15 minutes, and repeating the dose of mannitol if there is no initial response in 30 minutes to 2 hours. Hypertonic saline (3%), at a dose 2.5-5 mL/kg over 10-15 minutes, may be used as an alternative to mannitol, especially if there is no initial response to mannitol. After these measures have begun, imaging of the CNS should be arranged to identify intracranial pathology such as thrombosis and treat as appropriate.

 

Caveats

 

  1. Ketone bodies measured in urine grossly underestimate the degree of ketosis, because the common method uses sodium nitroprusside which reacts strongly with aceto-acetate, weakly with acetone, and not at all with βOHB. Yet the actual amount of βOHB may be 5 times or more than aceto-acetate, especially in the presence of acidosis. As acidosis is corrected and more of the βOHB is converted to aceto-acetate, it appears as if the ketosis is getting worse, when in fact acidosis and clinical parameters are improving. Measurements of βOHB via bedside meters or formal laboratory methods are better means to monitor “ketone” status.
  2. After commencing treatment, acidosis may appear to worsen initially for 3 reasons (4). First, dilution of the total bicarbonate in the expanding fluid volume lowers the apparent bicarbonate concentration because the HCO3 is expressed as mmol/L, and while the total mmols may not have changed, they are distributed in a greater volume. Second, with initially rapid rehydration, accumulated lactic acid enters into the circulation. Third, the βOHB acid is excreted in urine after it is converted to Na Butyrate; the Na derives from NaHCO3, leaving bicarbonate which combines with H+ to yield CO2 and H2O and permits loss of the CO2 in respiration. In these processes, bicarbonate (HCO3) and Na are lost, further depleting the bicarbonate content of plasma. With rehydration and insulin, which together curtail ketogenesis, acid base balance gradually returns to normal (4).
  3. The use of phosphate as potassium phosphate or acetate rather than KCl, may reduce the large amount of chloride used and hence reduce hyperchloremic acidosis, as well as improving oxygen dissociation to enable lactate to be converted to pyruvate. However, this is not accepted by all authorities and some claim no additional benefit from using phosphate. In addition, the use of phosphate may result in hypocalcemia. However, with severe phosphate depletion, the use of phosphate is indicated and likely to be beneficial.

 

Recurring Episodes of DKA

 

A small subset of patients experience repeat episodes of DKA, and with each episode the prognosis for short-term and long-term outcome worsens. Recent data confirm that the majority of such recurrences reflect psycho-social maladjustments that require careful attention via medical and social support to avoid disastrous consequences (5, 6).

 

HYPERGLYCEMIA HYPEROSMOLAR SYNDROME (HHS)

 

The hyperglycemia-hyperosmolar syndrome (HHS) is characterized by blood glucose concentrations >600mg/dl (>33.3 mmol/l), serum hyper-osmolality ≥330mmol/l, and minor acidosis and ketosis; serum bicarbonate remains >15meq/l and urinary “ketones” (aceto-acetate) are usually negative or only trace positive on testing urine via dipstick (7, 21, 22). Hospital admissions for HHS are increasing in incidence, have high morbidity, and though classically considered to occur in obese patients with T2DM it may occur in T1DM as frequently as in T2DM (23). Although there are similarities to diabetic keto-acidosis, the fundamental difference is a greater degree of dehydration and less acidosis, so that treatment should focus on fluid and electrolyte replacement, and less on provision of insulin; indeed, insulin should be withheld initially to prevent a too rapid fall in serum glucose and lowering of serum osmolality which might result in fluid shifts into the cerebral compartment and cerebral edema (CE). However, CE is rarer in HHS than in DKA. The degree of insulin deficiency and the magnitude of counter-regulatory response appear to be less severe, so that the symptoms and signs of DKA are absent or less pronounced; abdominal pain and Kussmaul respiration are absent, and vomiting is less severe. These milder features also lead to greater time in evolution, greater degrees of dehydration and electrolyte losses resulting from the polyuria, and are often compounded by intake of highly glucose-enriched carbonated soft drinks consumed due to thirst. Glucose concentrations commonly exceed 1000mg/dl, dehydration may be as much as twice that occurring in DKA and may be difficult to estimate due to co-existing obesity and hypertonicity which retains fluid in the intra-vascular compartment. Persistence of the polyuria due to the persistence of glucose concentrations exceeding renal threshold of ~200mg/dl during treatment, requires careful monitoring of clinical status and fluid replacement to avoid dehydration and vascular collapse. The risk of thrombosis is greater in HHS than in DKA, possibly as a result of osmotic disruption of endothelial cells, with release of thromboplastins facilitating coagulation.

 

Treatment should assume dehydration of 10%-15% and initially isotonic (normal) saline should be provided at 20ml/kg bolus infusions to restore fluid deficits and maintain vascular volume with assessment of serum chemistries every 1-2 hours; subsequently, 0.5-0.75 N saline, with added potassium and phosphate should be infused to replace calculated losses over 24-48 hours, guided by laboratory chemistry every 2-4 hours and ongoing clinical monitoring performed in an ICU or equivalent setting. The aim should be to control the decline in blood glucose to 100 mg/dl per hour; if glucose is not declining at a rate of at least 50 mg/dl, or ketosis is more than mild, insulin at a rate of only 0.025-0.05U/kg/hour may be used with caution and careful clinical and laboratory monitoring. Potassium, phosphate and magnesium losses may be considerable; potassium should be infused at 40meq/l added to each liter of saline, with balanced mixtures of potassium chloride and potassium phosphate, the latter to replete phosphate depletion which may predispose to rhabdomyolysis and hemolytic anemia. As in DKA, use of bicarbonate is not recommended. Magnesium also may be severely depleted in HHS and predispose to hypocalcemia; the recommended doses of magnesium replacement are 25-50mg/kg/dose given every 4-6 hours at a maximum infusion rate of 150mg/min(2gm/hr.) for 3-4 doses. In addition to cerebral edema, thrombosis, and rhabdomyolysis, malignant hyperthermia is reported as a complication. Monitoring for these complications is based in part on clinical anticipation e.g., hyperthermia, and supplemented by appropriate biochemical testing e.g., serum creatinine kinase for rhabdomyolysis. Some patients have features that combine DKA and HHS that reflect the degree of insulin deficiency; clinical acumen, earlier use of insulin, and careful monitoring of the patient’s vital signs and chemistries guide treatment, especially the earlier use of insulin in appropriate doses. This syndrome of HHS in adolescents and young adults was classically considered a feature of T2DM (24), an entity that is increasing at an annual rate of 4.8% in the obese population of the USA (25). Hence, the frequency of HHS as a presenting feature is also likely to increase, so that physicians caring for these patients in an ICU or equivalent setting must be alert to the differences in management with the greater focus on fluid and electrolyte replacement in HHS rather than the use of insulin as in DKA.  

 

Table 5. Monitoring of Patients with HHS in the ICU (1)

A.    Continuous cardiac, respiratory and blood pressure monitoring

B.    Hourly glucose and clinical assessment

C.    2-4hourly assessment of fluid balance(input/output); serum electrolytes, BUN, creatinine, CPK (creatine-phospho-kinase)

D.    4-6 hourly Calcium, phosphate, magnesium

E.    Be alert to complications-thrombosis, rhabdomyolysis, hyperpyrexia, cerebral edema.

 

ROUTINE MANAGEMENT OF DIABETES

 

The goals of treating diabetes mellitus in children are to maintain metabolism as near to normal by the appropriate provision of insulin, maintaining nutrition by meeting caloric requirements and balanced composition of food choices within the cultural preferences of the family, and to balance both insulin and nutrition with recommended exercise and activity to allow normal growth and development. In order to prevent diabetes related complications, especially long-term microvascular disease, glycemic control is crucial.  This optimal diabetes regimen requires intensive management by patients and their families along with a multidisciplinary approach with psychosocial support.  Glycemic control is assessed by periodic measurement of hemoglobin A1C levels.

 

Table 6. The American Diabetes Association Guidelines for the Target Glucose and HbA1C Levels (26)

A1C

<7%

Pre-prandial plasma glucose

90-130 mg/dl

Overnight plasma glucose

70-180 mg/dl

 

TYPE 1 DIABETES

 

Insulin Therapy

 

The management of diabetes can be cumbersome.  In caring for children and adolescents with Type 1 diabetes, providers must take into account unique factors such as a child’s pubertal stage and growth, ability to provide self-care, supervision of care, school environment, and neurological vulnerability of hypoglycemia in young children. 

 

However, it is crucial to normalize glucose levels in order to prevent long-term consequences of diabetes especially from microvasculopathies, leading to neuropathy, renal failure, and blindness. In 1993, The Diabetes Control and Complications Trial (DCCT) reported results demonstrating that the intensive therapy of T1DM reduces the risk of development and progression of microvascular complications. Furthermore, these benefits outweighed the increased risk of hypoglycemia that accompanied intensive diabetes therapy (27). Thereafter, The Epidemiology of Diabetes Interventions and Complications (EDIC) study assessed whether these benefits persisted after the end of DCCT. The findings of this study provide further support for the DCCT recommendation that most adolescents with T1D receive intensive therapy aimed at achieving glycemic control as close to normal as possible to reduce the risk of microvascular complications (28). This goal is not easily achieved even with a multi-dose insulin regimen of basal and short acting insulin that attempt to mimic normal patterns (Figure 2).

 

Figure 2. Normal Glucose and Insulin Profiles

This pattern is difficult to achieve because the perfect alignment of glucose and insulin in the normal person depends on a complex interaction of neural, hormonal, and nutritional signals that are absent in type 1 DM. Moreover, the “first pass” of endogenous insulin is via the portal vein to the liver, whereas SQ insulin injections first reach the liver via the systemic circulation. Hence the common problem of post-prandial hyperglycemia due to delay in the action of insulin during and immediately after a meal, and rebound hypoglycemia sometime after the meal. The following describes the insulin regimens recommended as standard of care attempting to reproduce the normal situation.

 

Figure 3. Time Course of Rapid and Long-Acting Insulin

 

Insulin Glargine and Detemir provide day-long basal insulin without significant peaks of action. In some children, however, Glargine may not be fully effective for a whole 24 hours and for this reason is usually given at night. This synthetic insulin cannot be safety mixed with other insulins in the same syringe due to pH incompatibilities. Throughout the day, short acting insulin preparations such as Insulin Aspart, Lispro, and Glulisine are given to normalize blood glucose levels and cover calories consumed during meals and as possible snacks. As 3 meals are eaten by most on a daily basis, short acting insulins should be given at least 3 times daily to prevent excessive hyperglycemic excursions.

 

Prepubertal children typically require a total daily insulin dose of ~ 0.7 -1 U/kg/day whereas pubertal children may require total daily insulin doses up to 1.2-1.5 U/kg/day; greater than 2.0 U/kg/day suggest extreme insulin resistance or non-compliance (29).  Of the total daily insulin dose, 40-60% should be given as basal insulin. The actual dose depends upon the level of glycemia and the quantity of calories and carbohydrates consumed. A fast-acting insulin bolus is given to cover meals, calculated from an estimation of the carbohydrate content in grams and an individual factor (insulin to carbohydrate ratio) relating insulin dosage to the amount of carbohydrate to be consumed. When using carbohydrate counting, the ‘500-rule’ can be used to obtain an initial carbohydrate ratio by dividing 500 by the total daily insulin dose.(29) The range is from 1 unit per 10-50 grams.  In addition to the meal bolus, the difference between the blood glucose recorded immediately before the meal and the target glucose concentration (120 mg/dl for older children; 150 mg/dl for younger children) is used to calculate a correction bolus, based on a theoretical Insulin Sensitivity Factor or ISF. This may range from 1 unit for 10- 200 mg/dl blood glucose depending upon age and body size. The ‘1800-rule’ can be used to obtain an initial ISF by dividing 1800 by the total daily insulin dose.

 

Table 7. Types of Insulin Preparations and Action Profiles (30)

Insulin

Onset

Peak

Duration

Insulin lispro

(rapid acting)

15-30 mins

0.5-2 hrs

2-5 hrs

Insulin aspart

(rapid acting)

15 mins

1-3 hrs

3-5 hrs

Insulin glulisine

(rapid acting)

12-30 mins

1.5 hrs

5-6 hrs

Fiasp

(ultra-rapid acting)

5 mins

0.5 hrs

3-5 hrs

Regular

(short acting)

0.5-1 hrs

2-4 hrs

5-8 hrs

NPH

(medium acting)

2-4 hrs

4-10 hrs

8-16 hrs

Insulin glargine

(long acting)

2-4 hrs

None

24 hrs

Insulin detemir

(long acting)

1-2 hrs

6-12 hrs

20-24 hrs

Insulin degludec

(ultra-long acting)

0.5-1.5 hrs

none

42 hrs

Insulin glargine U300

(ultra-long acting)

6 hrs

none

24-36 hrs

 

The intermittent, short acting insulin given as a bolus should be injected 15 minutes before the meal in order to have its full effect when glucose rises during and after the meal. For infants and toddlers, these short acting insulin doses may be given after the meal if food consumption has been found to be unpredictable. In addition to the 3 meals, additional amounts of short acting insulin may be taken to cover snacks, and to reduce blood glucose concentration as appropriate at bedtime. An insulin "pen" is a convenient way of carrying multiple doses in a single dispenser but this pen device does not reduce the burden of multiple insulin injections.  Super long-acting insulin preparations e.g. Degludec or super-fast acting insulin e.g. FIAsp(Ultra-Fast acting insulin Aspart) are available and may have advantages in specific situations. In particular, a super-fast acting insulin would allow more rapid equilibration between the systemic and portal circulations, so offering advantages to prevent excessive rise in glucose after a meal and avoiding later development of post-prandial hypoglycemia (31, 32).  The half-life of insulin degludec exceeds the dosing interval resulting in a very low peak:trough ratio at steady state.

 

An alternative and better method of insulin delivery is the use of continuous subcutaneous insulin infusion (CSII) through use of an insulin pump. In this case, only short acting insulin e.g., lispro, glulisine), aspart, or FiAsp is taken as a continuous basal infusion and multiple boluses of insulin are given as above. The newer generation of CSII pumps automatically calculate meal and/or correction boluses based on input insulin-to carbohydrate ratios and insulin sensitivity factors plus estimates of the amount of carbohydrate consumed. The infusion site is best changed every 2 days to avoid skin infections. The advantages of CSII when used correctly are that insulin is delivered only as needed, and not in an anticipatory fashion as with long-acting insulin. Insulin boluses are also delivered only as needed, nutritional intake may be more liberal, glycemic excursions both as hyperglycemic and hypoglycemic episodes can be reduced, and the system is convenient and portable. Patients receiving insulin via CSII and their parents have generally reported improved treatment satisfaction. The one disadvantage of CSII is that since only short acting insulin is used (effects of rapid acting insulins are dissipated within 3 hours), any blockage in the tubing or pump failure can lead to rapid onset of hyperglycemia, accumulation of serum ketones, and an uncontrolled diabetic state. Thus, patients and their care-givers must be educated on treatment of hyperglycemia with an insulin pen or syringe in case of suspected pump malfunction which can be a common cause of DKA. 

 

With advanced technology, insulin therapy is becoming more physiologic. Continuous subcutaneous insulin infusion (CSII) therapy is transforming care of T1DM while continuous glucose monitoring (CGM) of interstitial fluid has become widely available and increasingly used in the US. Without continuous glucose monitoring, manual adjustment of insulin doses in response to changes in blood glucose are based only upon intermittent blood glucose testing and corrections.  Moreover, as previously mentioned, insulin injections or infusions are given subcutaneously and initially enter the systemic circulation, whereas endogenous insulin is secreted into the portal vein and act directly and immediately on the liver.

 

Currently, we are entering a new era of diabetes care for children, with the adoption of closed loop systems (33). All systems in development rely on glucose measurements via CGM transmitted to an insulin delivery system (pump) which uses a computerized algorithm to adjust insulin infusion based on upper and lower limits and the rate of change in increase or decrease of glucose values (34-36).  Modified versions of the closed-loop system that are semi-automated are FDA approved for use in children.  Semi-automated closed-loop systems, also known as hybrid closed loop systems, are characterized by the combination of automated insulin delivery via an algorithm for basal requirement and user-initiated insulin delivery for meals.  A setting to suspend insulin delivery when glucose is low or rapidly downward trending can be utilized to avoid hypoglycemia. To improve ease of mobility, a tubeless insulin pump that can be operated by a third party through a wireless receiver is available and useful for very young children with control exercised by a parent.    

 

Bi-hormonal systems (insulin and glucagon) can rapidly alter high or low glucose values and be used with safety during normal activity and exercise (37, 38).This system has not yet been approved for commercial use. However, the next decade should see application of these tools to an increasing number of patients with T1DM.

 

The clinical management for patients with T1DM is based on adequate insulin replacement matched to food intake and modified by exercise. Insulin is required throughout the whole day to prevent development of a starvation state and ketosis i.e., the basal insulin requirement.

 

Recurrent hypoglycemia represents a mismatch between insulin provided and caloric expenditure, which occurs as a result of not covering the basal amount or a bolus with appropriate food intake, malabsorption of food e.g., celiac disease, or exercise without adjustment in the insulin dose or omission of additional calories before or after the exercise. unexplained episodes of hypoglycemia require re-evaluation of the insulin regimen, exclusion of concurrent conditions such as acute illness with diminished food intake, and testing for celiac disease, hypothyroidism, and Addison’s disease. 

 

In special circumstances where adherence to recommended regimens is not being followed due to various psycho-social limitations, a twice daily regimen of pre-mixed insulin (NPH/Reg70/30) may be prescribed, though it is known not to be ideal. In developing countries with limited resources to treat diabetes, this twice daily dosing regimen of NPH and regular continues to be vital to avoid DKA but is associated with less optimal glycemic control. 

 

Glucose Monitoring

 

Patients on insulin regimens are encouraged to check glucose levels prior to meals and snacks, at bedtime, prior to exercise, and when they suspect low blood glucose.  This typically amounts to 4-6 glucose levels per day.  Blood glucose monitoring allows patients and families to evaluate their individual response to therapy, assess whether achieving glycemic targets, and help guide treatment decisions.

 

Continuous glucose monitors and intermittently scanned monitors measures interstitial glucose which correlates well with plasma glucose.  Time in range is defined as percentage of the day with blood glucose levels between 70 and 180 mg/dL.  A time in range target of more than 70% is recommended (39).  Time in range is becoming a key glycemic metric, in addition to glycosylated hemoglobin (HbA1c) (39).  A correlation between improved HbA1c levels and fewer diabetes complications has been demonstrated (40).  Percent time in range provides real-time insights into glycemic variability and frequency of hypoglycemia and hyperglycemia. Time in range also correlates with complications of diabetes and HbA1c values and is being used to compare diabetes technology (40).

 

Nutrition

 

The goal of nutrition is to support normal growth and development, improve diabetes outcomes, and reduce cardiovascular risk factors.  Dietary recommendations should be based on healthy eating principles appropriate for all children and their families. Typically, distribution of energy sources recommended is 50-55% carbohydrates, <35% fat, and 15-20% protein (41).  Regular meals are recommended.  A commonly prescribed meal plan consists of 20% of calories at breakfast, 30% at lunch and 30% at dinner with 2 snacks of 10% each one of which is at bedtime to avoid nocturnal hypoglycemia. In the basal-bolus insulin regimen, insulin doses are matched to the amount of carbohydrates consumed during each meal. 

 

Protein and fat are not typically accounted for in the meal time insulin dose calculation though this issue is controversial and some authorities recommend that these protein-fat derived calories must be included. The predominant effect of dietary fats and protein is late postprandial hyperglycemia. Bolus corrections for insulin pump use when eating fatty meals have been devised and recommended (42-44).  Studies have found that lower glycemic index diets improved glycemic control compared to traditional higher glycemic index diets (45, 46).  Low glycemic index foods include whole-grain breads, pasta, temperate fruits, and dairy products

 

Dietary advice should be given in the context of cultural, ethnic and family traditions to be successful. Continuous nutritional education regarding a healthy diet and carbohydrate counting is recommended. Food labeling requirements have simplified the process, as many foods are clearly labeled with the amount of carbohydrate grams per serving. Additionally, apps, such as Calorie King, provide information on carbohydrate content of foods provided by many large restaurants.

 

Exercise

 

Establishing and maintaining an active lifestyle should be the goal for all children. Increased physical activity is associated with better glucose utilization and increased insulin sensitivity leading to lower insulin requirements. However, blood glucose levels can be difficult to regulate during these intervals of exercise. Hypoglycemia is common during exercise and can possibly last up to 24 hours afterwards, due to increased insulin sensitivity (47). This increases the risk of nocturnal hypoglycemia. Factors during exercise frequently associated with hypoglycemia are excessive insulin dosing prior to exercise, prolonged duration, and higher intensity aerobic exercise (47).

 

To reduce the risk of hypoglycemia during prolonged exercise, reductions in bolus and basal insulin are typically needed. In children using CSII pumps, simply suspending or reducing the basal infusion rate can markedly reduce the risk of hypoglycemia during exercise. If insulin doses prior to exercise are not reduced, a snack of 1-1.5 grams of carbohydrates per kilogram is recommended (47). Meals with high carbohydrate content should be consumed shortly after exercise. As the effects of exercise can be prolonged, blood glucose should be measured before bed and a decrease in basal insulin (either long acting or overnight basal) should be considered after exercise later in the day. 

 

Any exercise should be avoided if blood glucose prior to exercise are high (>250 mg/dl) and associated with ketonuria. Exercise during such insulinopenic states is dangerous owing to the effects of uninhibited counterregulatory hormones and may precipitate diabetic ketoacidosis. 

 

Sick Day Management

 

Children with intercurrent illnesses such as fever or vomiting, should be closely monitored for the development of hyperglycemia and ketonuria. On sick days, blood glucose levels should be checked every 2-3 hours when not tolerating food and urine should be checked for the presence of ketones with every void. Correction doses with rapid-acting insulin should be given approximately every 3 hours. Persistent vomiting and/or ketonuria are signs of diabetic ketoacidosis; patients with such signs and symptoms should be evaluated in an emergency department immediately. 

 

Adequate fluid intake is crucial to preventing dehydration and accumulation of ketones. For blood glucose >200 mg/dl, rehydration with sugar free fluids is recommended. Sugar containing fluids such as flat soda or diluted juice may be necessary to maintain normoglycemia if blood glucose is <140 mg/dl. 

 

Management of Co-Morbid Conditions

 

Besides insulin replacement therapy for T1DM, co-existing hypertension and dyslipidemia should be aggressively treated; it is important to use age-appropriate references for determining the presence of hypertension and upper levels of acceptable lipid values of LDL and triglycerides. 

 

Increased urinary protein excretion is the earliest clinical finding of diabetic nephropathy. Measurement of the urine albumin-to-creatinine ratio in an untimed urinary sample is the preferred screening strategy for moderately increased albuminuria in all patients with diabetes and should be repeated yearly. Screening for increased urinary albumin excretion can be deferred for five years after the onset of disease in patients with type 1 diabetes because increased albumin excretion is uncommon before this time; screening should begin at diagnosis in patients with type 2 diabetes because many have had diabetes for several years before diagnosis. Abnormal results should be confirmed by repeat testing before establishing a diagnosis because of the large number of false positives that can occur. The normal ratio of microalbumin to creatinine is less than 30 mg/g.  Thus, a persistently elevated ratio of 30-300 mg/g signifies microalbuminuria. Microalbuminuria and/or hypertension should be a call for use of angiotensin converting enzyme (ACE) inhibitors to minimize progression to chronic glomerulosclerotic damage. ACE inhibitors may induce angio-edema and can produce a troublesome dry cough.

 

Poorly controlled diabetes induces an increase in VLDL and triglyceride levels, when acute or chronic, pancreatitis may be induced. Diet reduced in animal fat and administration of fibrates (e.g., gemfibrozil or fenofibrate) may be used to combat hypertriglyceridemia. Co- existing Hashimoto's thyroiditis should be periodically sought through thyroid autoantibody analyses, and hypothyroidism when identified by elevated TSH levels, treated by thyroid hormone replacement. Celiac disease also should be regularly checked via titers of tissue transglutaminase antibody titers, and treated via gluten exclusion when diagnosis is confirmed by endoscopically obtained biopsy specimens. Addison's disease and atrophic gastritis/pernicious anemia should always be considered in patients with T1DM, especially with unexplained frequent episode of hypoglycemia, and if found, treated accordingly.

 

TYPE 2 DIABETES

 

The increased incidence of T2DM is attributed to the increase in obesity worldwide. Approximately 3700 youths are diagnosed with T2DM every year in the US (25) and it is estimated that the number of youths with T2DM will almost quadruple from 22,820 in 2010 to be approximately 85,000 adolescents with T2DM by 2050 (48). Similar rates of increases in youths with T2DM are reported from the UK, India, China and Japan (48). The child with T2DM as part of the insulin resistance syndrome (IRS) should be aggressively treated to prevent the burgeoning complications of the condition. The development of complications associated with T2DM is accelerated in youth, with reported rates of 6% with renal failure within 5 years of diagnosis, and 2.3% end stage renal disease by 10 years.  

 

Initial education for T2DM should focus on dietary and lifestyle modifications and this education should continue to be reinforced with the goal being to decrease insulin resistance. The approaches should include an exercise program such as walking or swimming for 30-40 minutes most days of the week, since at the level of the muscle, exercise provokes glucose entry into muscle without the involvement of insulin. Sedentary time including homework, computer and phone related activities, and video games should be assessed and established for appropriateness in each family setting. Caloric restriction, particularly of carbohydrates, is the key to reducing weight, a task that has proven resistant to success in many instances.  Elimination of sugar containing sodas and juices has been shown to result in significant weight loss (49). Barriers include older age at diagnosis, difference in socioeconomic status, and poor diet within the household (50). Also, clinicians should understand the health beliefs and behaviors of the family and community and take into account cultural food preferences and the use of food during celebrations and cultural festivals in order to collaborate with the family on diabetes management.  

 

The use of metformin as first-line therapy is based on its glucose-lowering efficacy, safety profile, weight neutrality, and reasonable cost. In most countries, metformin is currently approved for use in children. Metformin is approved for the treatment of T2DM in children, but is also the drug of choice for insulin resistance syndrome, also known as metabolic syndrome (IRS) and impaired glucose tolerance because of its property in improving insulin sensitivity. Monotherapy with metformin was associated with durable glycemic control in approximately half of children and adolescents with T2DM (51). Some suggest that it is the gastro- intestinal side effects of the drug that accounts for much of its beneficial effects. However, the drug is effective in T2DM even without weight loss, an action attributed to reduced hepatic glucose output.

 

The guidelines from the ADA and EASD indicate that any FDA-approved second agent can be used in combination with metformin to improve glycemic control, whereas the American Association of Clinical Endocrinologists recommends either incretin-based therapy or sodium glucose transporter 2 (SGLT2) inhibition agents (52). Sulfonylureas are approved for use in adolescents in some countries; these agents bind to receptors on the K+/ATP channel complex resulting in insulin secretion. The PPAR-γ agonists are effective at insulin sensitization but are less useful in supporting weight loss. Further, they promote salt retention and a tendency for edema. A new class of drugs which inhibit the sodium co-transporter 2 (SGLT2) resulting in glycosuria at a lower blood glucose threshold than normal have become available, though not currently approved for use in children (canagliflozin, dapagliflozin, empagliflozin). Use of SGLT2 inhibitors has been associated with an increase in fungal infections of the genital areas and missed symptoms of evolving keto-acidosis.

 

Two drug classes were developed that target the incretin system and increase endogenous insulin secretion: glucagon-like peptide (GLP)-1 receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors. GLP-1 receptor agonists (e.g., liraglutide and exenatide) resist degradation by DPP-4 resulting in increased circulating levels of the administered drug (53). DDP-4 inhibitors (e.g., sitagliptin, vildagliptin and saxagliptin) reduce endogenous GLP-1 degradation, thereby maintaining circulating levels of GLP-1 with biological effect. Both these classes of drugs improve glycemic control with a low incidence of hypoglycemia because of their glucose-dependent mechanism of action. In addition to their effects on improving insulin secretion, these drugs lower glucagon and delay gastric emptying, and potentiate weight loss, in part through decreased appetite.

 

Whereas the glucagon like peptide one (GLP-1) analogue exenatide given by subcutaneous injection will lower blood glucose levels and complement metformin in provoking weight loss, it should be reserved for more severely diabetic adults and teenagers who have become unresponsive to diet and exercise programs.  Some formulations of GLP-1 analogues can be given once weekly.  In 2019, liraglutide was approved for adjunct use to improve glycemic control in pediatric patients 10 years and older with T2DM. (54, 55) Sitagliptin blocks the dipeptidyl peptidase-4 (DPP-4) enzyme preventing it from inactivating GLP-1, thus prolonging the action of GLP-1 once induced by a meal. Whereas the latter agent is in general weight neutral, it can be of adjunctive help in lowering hyperglycemic excursions. When these additional agents also fail to maintain near normoglycemia, then insulin should be given instead of the secretagogues.

 

Table 8. Glucose Management for Adult Patients with Type 2 Diabetes (52)

IRS/IGT

 

 

Diabetes

Mild

 

 

 

 

 

 

 

 

 

 

 

 

Severe

Step 1: Dietary and lifestyle education- 3-5% weight loss; 150 min/week exercise

Step 2: Addition of metformin- Maximum daily dose of 2000 mg

Step 3: Addition of second antihyperglycemic drug

 

Pioglitazone

DPP4 inhibitor

GLP-1 agonist

SGLT2

inhibitor

Sulfonylurea

HbA1C

¯

¯

¯

¯

¯

Weight

 

--

¯

¯

 

Hypoglycemia

--

--

--

--

 

Major CV events

--

--

¯

¯

-

Heart failure

 

*

--

¯

-

Step 4: Addition of insulin

Basal insulin with or without prandial insulin

*Saxagliptin is a DPP4 inhibitor associated with heart failure. Other DPP4 inhibitors have not been shown to cause heart failure. Whereas studies of these agents are under investigation, only a GLP-1 agonist is currently approved by the FDA for use in children and adolescents

 

One of the main goals of therapy in IRS/T2DM should be to achieve an ideal body mass index (kg/m2) for age and gender. This is not readily achievable with lifestyle modification and medical therapy in many subjects; bariatric surgery is emerging as a successful and durable treatment in adults and adolescents with IRS, obesity, and their associated complications (56-58). In adults the ADA recommends bariatric surgery in those with BMI of 30 kg/m2 or greater and poorly controlled DM (59). Bariatric surgery is an effective treatment for severe obesity that results in the improvement or remission of many obesity-related comorbid conditions, as well as sustained weight loss and improvement in quality of life. Mortality owing to cardiovascular diseases, diabetes, and respiratory conditions is reduced after bariatric surgery (60). A prospective follow up studies of bariatric surgery in adolescents with severe obesity showed a substantial and durable weight reduction and cardio-metabolic benefits (57, 58). Changes in glucoregulatory hormones produced by the gastrointestinal tract, bile acid metabolism, and GI tract nutrient sensing and glucose utilization are proposed mechanism for improvement in glycemic control after bariatric surgery (61). Currently, bariatric surgery is considered only in children with BMI ≥ 40 kg/m2 with comorbidities or BMI ≥ 50 kg/m2regardless of comorbidities (62, 63). Indications in adults are much less stringent; adults with BMI ≥ 35 kg/m2 with comorbidities are candidates for these procedures. Updated recommendations for adolescents provide more aggressive recommendations similar to those for adults (64). Bariatric surgery is now safe, with mortality comparable to common elective general surgical operations. Level 1 evidence show that bariatric surgery provides superior short-term and long-term weight loss and improvement of T2DM compared with conventional medical therapy. However, patients require life-long follow up and monitoring for nutritional deficiencies and abdominal issues, and to date, results in adolescents are relatively short term.  Pediatric patients who are being considered for bariatric surgery should be evaluated by a multidisciplinary team dedicated to providing long-term follow- up care postoperatively. In addition, selection criteria often exclude the population most in need of this proven procedure.

 

Treatment of Associated Comorbidities

 

The typical dyslipidemia associated with IRS and T2DM should be treated by reduced intake of animal fat and a fibrate such as gemfibrozil or fenofibrate. Where there is an increased level of triglycerides, restriction of animal fats and simple sugars should be recommended. However, those patients who have prominent elevations in LDL-cholesterol should be treated with a statin. The mixed use of a statin and a fibrate should be undertaken cautiously since the risks of muscle necrosis (rhabdomyolysis) with renal failure has been. In patients taking a statin gemfibrozil should not be used and fenofibrate is the fibrate of choice as the risk of myositis is less. Hypertension and microalbuminuria, when present, should be aggressively treated, preferably with angiotensin converting enzyme inhibitors (ACE) and angiotensin receptor blockers (ARBs), at least initially. 

 

Oral contraceptive agents are often prescribed in IRS when there is evidence of hyper-androgenization, where they may counteract the effects of androgens. However, oral contraceptives also increase the level of hormonal binding globulins, including sex hormone binding globulin that binds testosterone, thereby lowering the level of free and bio-available testosterone. Estrogen containing therapies in a prepubertal patient increases the risk of premature closure of the epiphyses and hence risks loss of adult height; they also promote thrombosis and mitigate against weight loss.

 

TREATMENT OF MONOGENIC FORMS OF DIABETES

 

Monogenic forms of diabetes constitute a heterogeneous group of disorders classified according to clinical features that suggest possible type 1 diabetes, type 2 diabetes, and neonatal diabetes, all in the absence of markers of autoimmunity such as circulating antibodies to various islet antigens. The genes responsible for these forms share a role in the formation or function of the pancreatic β-cell, limiting normal insulin secretion that depending on severity, and under certain conditions, results in clinical diabetes. Increasingly, it is being recognized that there is a continuum in the spectrum of these disorders such that the severity of the genetic defect responsible for insulin secretion or action determines the clinical pattern (65-68). This is perhaps best exemplified in the genetic defects of the ATP-regulated potassium channel (KATP) involving the ABCC8 gene coding for the sufonylurea receptor SUR1, and KCNJ11 coding for the subunits of Kir, the inward rectifying potassium channel itself. Severe activating mutations in these genes maintain the KATP in an open state and result in permanent neonatal diabetes, sometimes associated with developmental delay and epilepsy (DEND). Progressively less severe functional mutations may result in transient neonatal diabetes, or in a form of maturity onset diabetes of youth (MODY), or in T2DM.These activating mutations typically respond to sulfonylurea therapy, high dose for the severe mutations and lower doses for the less severe mutations, inducing endogenous insulin secretion mediated in part by GLP-1, and improved metabolic control superior to that obtained by exogenous insulin injection. Similar considerations apply to transcription factors such as hepatocyte nuclear factor1α (HNF1A) and hepatocyte nuclear factor 4α (HNF4A), respectively responsible for MODY3 and MODY1, which may respond to oral sulfonylurea drugs, avoiding the need for injected insulin, at least initially. Heterozygous inactivating mutations in the glucokinase gene responsible for MODY2 delay insulin secretion and result in a mild diabetes that is not associated with an increased risk of macrovascular or microvascular complications, so that treatment with exogenous insulin or other drug therapies is not indicated (69). Hence, knowledge of the genetic mutation drives therapy, permits more precise genetic counselling, and may indicate prognosis. In an era of precision medicine and progressive decline in the cost of sequencing, genetic testing should be considered in those with a strong family history of diabetes, early onset diabetes, and in children or adolescents who present with features suggestive of T1DM, are negative for islet auto-antibodies, and have residual c-peptide secretion as determined by measurement or reflected in persistently low insulin requirements extending beyond 1 year after diagnosis. These concepts are discussed in greater detail in several recent publications (65-69).      

 

Maturity Onset Diabetes of Youth (MODY)

 

The term MODY refers to Maturity Onset Diabetes of Youth, a term coined by Fajans and Tattersall for a mild type of diabetes with autosomal dominant inheritance and varying degrees of impaired insulin secretion (70). The molecular basis for these entities was discovered initially to be due to transcription factors or the enzyme glucokinase responsible for phosphorylating glucose to enable its metabolism to yield ATP; numbering followed the timing of discovery of the genetic-molecular basis (70). There are now 14 entities considered to be MODY as listed in the table in our chapter on the “Etiology and Pathogenesis of Diabetes Mellitus in Children and Adolescents” in Endotext. Of the original 6 “classical” MODY entities, MODY3 (HNF1A mutation), MOY2 (GCK (glucokinase) mutation) and MODY1 (HNF4A mutation) constitute about 85% of all MODY cases (65-68). With rare exceptions, these patients present as milder types of diabetes before age 30-35 years, with a positive family history involving at least 2-3 generations, and negative for islet cell antibodies; a daily dose of insulin less than 0.5U/Kg /day after 1 year of diagnosis should raise suspicion for MODY. MODY affects both sexes and is found in all races, with a prevalence of ~2%-4% of patients diagnosed with diabetes ≤ 30 years (71, 72). The majority are misdiagnosed as type1 or type 2 diabetes and incorrectly and unnecessarily treated with insulin or ineffective drugs such as metformin. MODY 2 affects about 1:1000 people and in females may be noted for the first time during oral glucose tolerance testing in pregnancy, again resulting in inappropriate classification and treatment (73). Biomarkers such as the urinary C-peptide to creatinine ratio (≥0.2nmol/mmol), and negative islet cell antibodies (GAD and IA2) should lead to molecular genetic testing (72). Using this approach, the minimum prevalence of monogenic diabetes was found to be 3.6% of patients diagnosed ≤30years of age with diabetes (72). In a study screening for MODY in all antibody negative children with diabetes in a national population-based registry in Norway, the prevalence of MODY was found to be 6.5%, and in a study from Japan, 11/89 children with insulin requiring diabetes but negative for islet cell antibodies were found to have monogenic forms of diabetes involving mutations in INS, the insulin gene, and in HNF1A or HNF4A (74, 75). Mutations in HNF4A may be associated with large size at birth and neonatal hypoglycemia with hyperinsulinemia that resolves spontaneously, only later becoming manifest as diabetes. Family history is helpful but not essential; de novo mutations occur.

 

In summary, there should be a high index of suspicion for MODY in milder forms of diabetes and in those children who are islet cell negative; using biomarkers followed by molecular diagnostics, the yield becomes quite high for discovering a form of MODY. As the cost of molecular diagnostics declines, and newer algorithms to apply these tools to differentiate apparent type1 from monogenic forms of diabetes are being developed (76, 77), it is becoming apparent that some of these mutations also contribute to the genetics of apparent type 2 diabetes (78, 79). For MODY3 and MODY1, oral sufonylurea medication (Glipizide) is likely to be effective inducing endogenous insulin secretion; MODY2 does not require treatment. Genetic counselling should inform patients of the 50% likelihood of each of their offspring having MODY, so that inappropriate diagnosis and treatment is avoided. In addition, the prognosis for vascular complications is improved especially in MODY2, though not absolute in MODY3. MODY12 (ABCC8) and MODY13 (KCNJ11) are also responsive to oral sufonylurea medication, but may require careful upward titration. For the remaining forms of MODY, insulin is likely to be necessary to control diabetes. In particular, these less frequent forms of MODY may have involvement of other organ systems, e.g., kidney cysts and dysfunction in MODY5(HNF1B), gastrointestinal involvement in MODY8 (CEL), exocrine pancreatic disturbances in MODY4 (PDX-1), blood abnormalities in MODY11(BLK), as well as other abnormalities (see references (65-68) for details). Thus, establishing a diagnosis for a form of MODY has several important consequences. First, it guides treatment, obviating the need for insulin with its costs and discomforts in several forms of MODY, as well as anticipation for possible associated abnormalities. Second, it permits a more accurate prediction of the course and prognosis for complications, e.g., MODY2, which in turn has ramifications on the cost and ability to obtain life insurance policy, or the choice of occupations which may be restricted to a person with T1DM. Third, it permits precise genetic counselling for risk of occurrence in offspring, and targeted molecular screening for the existence of the mutation in suspected family members.    

 

NEONATAL DIABETES MELLITUS 

 

Figure 4. Causes of Neonatal Diabetes

Neonatal diabetes mellitus (NDM) is defined as diabetes occurring in the first 6 months of life; for some authorities, the window extends to 9 months of age, but several of the mutations may manifest only later (65, 80, 81). For convenience, NDM is classified into 3 categories; transient NDM which constitutes about 45%, permanent NDM also constituting about 45%, and NDM associated with various other syndromic features, about 10% (Figure 4).

 

Transient Neonatal Diabetes

 

The transient forms are characterized by a period of remission during which glucose tolerance is normal, but diabetes usually recurs later in life. Of these transient forms, about 2/3rd involve methylation abnormalities in chromosome 6q24 which lead to malfunction of imprinted genes PLAGL, also known as ZAC, and HYMAI that arise by the mechanisms listed in the Figure. These infants display small size at birth due to inadequate in utero secretion of insulin, a major regulator of anabolic growth; there is rapid catch-up growth when insulin is provided by sub-cutaneous injection or via pump therapy with insulin diluted 1:10 so that 1 ml contains 10 U rather than the standard 100U/ml. Hyperglycemia and glucosuria are present but may be missed if not sought. Rare variants of these methylation defects may have initial hypoglycemia and devolve into hyperglycemia. Most are sporadic, but duplication of paternal chromosome 6 leads to dominant transmission (see figure). Of the remaining 1/3rd of TNDM, the majority harbor mutations in the KATP genes ABCC8 and KCNJ11 which respond to therapy via oral hypoglycemic agents such as glipizide; dosage requires titration to individual responsiveness. Approximately 5% of transient cases involve mutation in the insulin gene INS, the β-cell glucose transporter SLCA2A, or other genes as listed in Figure 4.

 

Permanent Neonatal Diabetes

 

Permanent NDM primarily involves 3 genes; severe mutations in KCNJ11 or ABCC8, and the insulin gene INS. Because the KATP channel and its’ genes are also expressed in the CNS, severe mutations also may affect neural function and development. Developmental delay, Epilepsy, and Neonatal Diabetes constitute the DEND syndrome, with associated physical and neuropsychological features; early treatment with oral sulfonylurea medication benefits neuropsychological function and timing of treatment influences outcome, i.e., the earlier the better (82-84). There is debate whether treatment with oral sulfonylurea should be started before confirmation of the genetic defect, but we recommend that it not be started, as the transition to oral agents with concomitant reduction in the injected insulin dose, essential to control the severe hyperglycemia and sometimes associated with DKA, is potentially dangerous and the dose of sufonylurea needed is much higher than that used in adults with T2DM. If successful, transition to oral therapy is associated with remarkable improvement of metabolic control due to stimulation of endogenous insulin secretion and neuropsychological improvement; it is also easier and less traumatic to the patient than insulin injection (80). We therefore recommend that such transitions be performed in a hospital setting according to a published protocol (85). When insulin therapy is used, either in mutations of the KATP channel or where it must be in mutations of INS which do not respond to sulfonylurea, using continuous subcutaneous infusion via a pump and diluted insulin, appears to be the best option (86).

 

Neonatal Diabetes and Associated Syndromes

 

About 10% of cases of NDM are associated with a spectrum of syndromic disorders; the more common ones are listed in the figure 4 and greater details can be found in the references (65-68, 77, 80, 81).  In all forms of neonatal diabetes, children are born small for gestational age; the smaller the child, the more severe the defect in insulin synthesis, secretion or action is likely to be. The associated abnormalities provide clinical clues, and it was the clinical associations that often defined the syndrome, before the genetic mutation was known. Next generation sequencing with a panel specifically designed for NDM can provide a rapid diagnosis and guide therapy, predict associated abnormalities, and infer possible interventions before some of the classical features have evolved (77, 81). Indeed, this is the approach now recommended, i.e., non-selective genetic testing in any case of neonatal diabetes. In addition, exome sequencing of unusual cases not covered by the panel may uncover new entities, as recently described for a form of autoimmunity associated with NDM that is responsive to a CTL4 mimetic (87). For most of the syndromic forms, insulin is the required therapy to control diabetes; an exception may be thiamine responsive megaloblastic anemia and diabetes which is due to mutation in the thiamine transporter SLCA29 and initially responsive to thiamine replacement (77, 80).

 

REFERENCES

 

  1. Jensen, E.T., et al., Increase in Prevalence of Diabetic Ketoacidosis at Diagnosis Among Youth With Type 1 Diabetes: The SEARCH for Diabetes in Youth Study. Diabetes Care, 2021.
  2. Foster, D.W. and J.D. McGarry, The metabolic derangements and treatment of diabetic ketoacidosis. N Engl J Med, 1983. 309(3): p. 159-69.
  3. Palmer, B.F. and D.J. Clegg, Electrolyte and Acid-Base Disturbances in Diabetes Mellitus. N Engl J Med, 2015. 373(25): p. 2482-3.
  4. Kamel, K.S. and M.L. Halperin, Acid-base problems in diabetic ketoacidosis. N Engl J Med, 2015. 372(20): p. 1969-70.
  5. Sperling, M.A., Diabetes: Recurrent DKA - for whom the bell tolls. Nat Rev Endocrinol, 2016. 12(10): p. 562-4.
  6. Gibb, F.W., et al., Risk of death following admission to a UK hospital with diabetic ketoacidosis. Diabetologia, 2016. 59(10): p. 2082-7.
  7. Wolfsdorf, J.I., et al., ISPAD Clinical Practice Consensus Guidelines 2018: Diabetic ketoacidosis and the hyperglycemic hyperosmolar state. Pediatr Diabetes, 2018. 19 Suppl 27: p. 155-177.
  8. Usher-Smith, J.A., et al., Variation between countries in the frequency of diabetic ketoacidosis at first presentation of type 1 diabetes in children: a systematic review. Diabetologia, 2012. 55(11): p. 2878-94.
  9. Jensen ET,Stafford JM,Saydah S, et al., Increase in the prevalence of ketoacidosis at diabetes diagnosis in youth with Type1 Diabetes: the SEARCH for diabetes in youth study. Diabetes Care 2021; June7 on line, doi10:2337 dc20-0389.
  10. Rewers, M., et al., Beta-cell autoantibodies in infants and toddlers without IDDM relatives: diabetes autoimmunity study in the young (DAISY). J Autoimmun, 1996. 9(3): p. 405-10.
  11. Szypowska, A. and A. Skorka, The risk factors of ketoacidosis in children with newly diagnosed type 1 diabetes mellitus. Pediatr Diabetes, 2011. 12(4 Pt 1): p. 302-6.
  12. Halperin, M.L., et al., Strategies to diminish the danger of cerebral edema in a pediatric patient presenting with diabetic ketoacidosis. Pediatr Diabetes, 2006. 7(4): p. 191-5.
  13. Carlotti, A.P., et al., Occult risk factor for the development of cerebral edema in children with diabetic ketoacidosis: possible role for stomach emptying. Pediatr Diabetes, 2009. 10(8): p. 522-33.
  14. White, P.C. and B.A. Dickson, Low morbidity and mortality in children with diabetic ketoacidosis treated with isotonic fluids. J Pediatr, 2013. 163(3): p. 761-6.
  15. Watts, W. and J.A. Edge, How can cerebral edema during treatment of diabetic ketoacidosis be avoided? Pediatr Diabetes, 2014. 15(4): p. 271-6.
  16. Muir, A.B., et al., Cerebral edema in childhood diabetic ketoacidosis: natural history, radiographic findings, and early identification. Diabetes Care, 2004. 27(7): p. 1541-6.
  17. Sperling, M.A., Cerebral edema in diabetic ketoacidosis: an underestimated complication? Pediatr Diabetes, 2006. 7(2): p. 73-4.
  18. Hsia, D.S., et al., Fluid management in pediatric patients with DKA and rates of suspected clinical cerebral edema. Pediatr Diabetes, 2015. 16(5): p. 338-44.
  19. White, P.C., Optimizing fluid management of diabetic ketoacidosis. Pediatr Diabetes, 2015. 16(5): p. 317-9.
  20. Glaser, N., et al., Risk factors for cerebral edema in children with diabetic ketoacidosis. The Pediatric Emergency Medicine Collaborative Research Committee of the American Academy of Pediatrics. N Engl J Med, 2001. 344(4): p. 264-9.
  21. Tasker, R.C. and C.L. Acerini, Cerebral edema in children with diabetic ketoacidosis: vasogenic rather than cellular? Pediatr Diabetes, 2014. 15(4): p. 261-70.
  22. Glaser, N.S., et al., Regional Brain Water Content and Distribution During Diabetic Ketoacidosis. J Pediatr, 2017. 180: p. 170-176.
  23. Bagdure, D., et al., Epidemiology of hyperglycemic hyperosmolar syndrome in children hospitalized in USA. Pediatr Diabetes, 2013. 14(1): p. 18-24.
  24. Fourtner, S.H., S.A. Weinzimer, and L.E. Levitt Katz, Hyperglycemic hyperosmolar non-ketotic syndrome in children with type 2 diabetes*. Pediatr Diabetes, 2005. 6(3): p. 129-35.
  25. Mayer-Davis, E.J., et al., Incidence Trends of Type 1 and Type 2 Diabetes among Youths, 2002-2012. N Engl J Med, 2017. 376(15): p. 1419-1429.
  26. American Diabetes Association., Summary of Revisions: Standards of Medical Care in Diabetes-2021. Diabetes Care, 2021. 44(Suppl 1): p. S4-S6.
  27. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med, 1993. 329(14): p. 977-86.
  28. American Diabetes Association., Standards of Medical Care in Diabetes-2021 Abridged for Primary Care Providers. Clin Diabetes, 2021. 39(1): p. 14-43.
  29. Danne, T., et al., ISPAD Clinical Practice Consensus Guidelines 2018: Insulin treatment in children and adolescents with diabetes. Pediatr Diabetes, 2018. 19 Suppl 27: p. 115-135.
  30. Hirsch, I.B., et al., The Evolution of Insulin and How it Informs Therapy and Treatment Choices. Endocr Rev, 2020. 41(5).
  31. Biester, T., et al., Pharmacokinetic and prandial pharmacodynamic properties of insulin degludec/insulin aspart in children, adolescents, and adults with type 1 diabetes. Pediatr Diabetes, 2016. 17(8): p. 642-649.
  32. Fath, M., et al., Faster-acting insulin aspart provides faster onset and greater early exposure vs insulin aspart in children and adolescents with type 1 diabetes mellitus. Pediatr Diabetes, 2017.
  33. Tauschmann, M. and R. Hovorka, Technology in the management of type 1 diabetes mellitus - current status and future prospects. Nat Rev Endocrinol, 2018. 14(8): p. 464-475.
  34. Bally, L., et al., Day-and-night glycaemic control with closed-loop insulin delivery versus conventional insulin pump therapy in free-living adults with well controlled type 1 diabetes: an open-label, randomised, crossover study. Lancet Diabetes Endocrinol, 2017. 5(4): p. 261-270.
  35. Anderson, S.M., et al., Multinational Home Use of Closed-Loop Control Is Safe and Effective. Diabetes Care, 2016. 39(7): p. 1143-50.
  36. Breton, M.D., et al., A Randomized Trial of Closed-Loop Control in Children with Type 1 Diabetes. N Engl J Med, 2020. 383(9): p. 836-845.
  37. Russell, S.J., et al., Blood glucose control in type 1 diabetes with a bihormonal bionic endocrine pancreas. Diabetes Care, 2012. 35(11): p. 2148-55.
  38. El-Khatib, F.H., et al., Home use of a bihormonal bionic pancreas versus insulin pump therapy in adults with type 1 diabetes: a multicentre randomised crossover trial. Lancet, 2017. 389(10067): p. 369-380.
  39. Battelino, T., et al., Clinical Targets for Continuous Glucose Monitoring Data Interpretation: Recommendations From the International Consensus on Time in Range. Diabetes Care, 2019. 42(8): p. 1593-1603.
  40. Pease, A., et al., Time in Range for Multiple Technologies in Type 1 Diabetes: A Systematic Review and Network Meta-analysis. Diabetes Care, 2020. 43(8): p. 1967-1975.
  41. Smart, C.E., et al., ISPAD Clinical Practice Consensus Guidelines 2018: Nutritional management in children and adolescents with diabetes. Pediatr Diabetes, 2018. 19 Suppl 27: p. 136-154.
  42. Bell, K.J., et al., Estimating insulin demand for protein-containing foods using the food insulin index. Eur J Clin Nutr, 2014. 68(9): p. 1055-9.
  43. Bao, J., et al., Improving the estimation of mealtime insulin dose in adults with type 1 diabetes: the Normal Insulin Demand for Dose Adjustment (NIDDA) study. Diabetes Care, 2011. 34(10): p. 2146-51.
  44. Pankowska, E., M. Blazik, and L. Groele, Does the fat-protein meal increase postprandial glucose level in type 1 diabetes patients on insulin pump: the conclusion of a randomized study. Diabetes Technol Ther, 2012. 14(1): p. 16-22.
  45. Gilbertson, H.R., et al., The effect of flexible low glycemic index dietary advice versus measured carbohydrate exchange diets on glycemic control in children with type 1 diabetes. Diabetes Care, 2001. 24(7): p. 1137-43.
  46. Gilbertson, H.R., et al., Effect of low-glycemic-index dietary advice on dietary quality and food choice in children with type 1 diabetes. Am J Clin Nutr, 2003. 77(1): p. 83-90.
  47. Adolfsson, P., et al., ISPAD Clinical Practice Consensus Guidelines 2018: Exercise in children and adolescents with diabetes. Pediatr Diabetes, 2018. 19 Suppl 27: p. 205-226.
  48. Imperatore, G., et al., Projections of type 1 and type 2 diabetes burden in the U.S. population aged <20 years through 2050: dynamic modeling of incidence, mortality, and population growth. Diabetes Care, 2012. 35(12): p. 2515-20.
  49. Barlow, S.E. and C. Expert, Expert committee recommendations regarding the prevention, assessment, and treatment of child and adolescent overweight and obesity: summary report. Pediatrics, 2007. 120 Suppl 4: p. S164-92.
  50. Zeitler, P., et al., ISPAD Clinical Practice Consensus Guidelines 2018: Type 2 diabetes mellitus in youth. Pediatr Diabetes, 2018. 19 Suppl 27: p. 28-46.
  51. Group, T.S., et al., A clinical trial to maintain glycemic control in youth with type 2 diabetes. N Engl J Med, 2012. 366(24): p. 2247-56.
  52. Reusch, J.E. and J.E. Manson, Management of Type 2 Diabetes in 2017: Getting to Goal. JAMA, 2017. 317(10): p. 1015-1016.
  53. Samson, S.L. and A.J. Garber, A Plethora of GLP-1 Agonists: Decisions About What to Use and When. Curr Diab Rep, 2016. 16(12): p. 120.
  54. Tamborlane, W.V., et al., Liraglutide in Children and Adolescents with Type 2 Diabetes. N Engl J Med, 2019. 381(7): p. 637-646.
  55. Kelly, A.S., et al., A Randomized, Controlled Trial of Liraglutide for Adolescents with Obesity. N Engl J Med, 2020. 382(22): p. 2117-2128.
  56. Inge, T.H., et al., Weight Loss and Health Status 3 Years after Bariatric Surgery in Adolescents. N Engl J Med, 2016. 374(2): p. 113-23.
  57. Inge, T.H., et al., Long-term outcomes of bariatric surgery in adolescents with severe obesity (FABS-5+): a prospective follow-up analysis. Lancet Diabetes Endocrinol, 2017. 5(3): p. 165-173.
  58. Olbers, T., et al., Laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity (AMOS): a prospective, 5-year, Swedish nationwide study. Lancet Diabetes Endocrinol, 2017. 5(3): p. 174-183.
  59. Mechanick, J.I., et al., Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient--2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring), 2013. 21 Suppl 1: p. S1-27.
  60. Lent, M.R., et al., All-Cause and Specific-Cause Mortality Risk After Roux-en-Y Gastric Bypass in Patients With and Without Diabetes. Diabetes Care, 2017.
  61. Batterham, R.L. and D.E. Cummings, Mechanisms of Diabetes Improvement Following Bariatric/Metabolic Surgery. Diabetes Care, 2016. 39(6): p. 893-901.
  62. Mirensky, T.L., Bariatric Surgery in Youth. Endocrinol Metab Clin North Am, 2016. 45(2): p. 419-31.
  63. Khattab, A. and M.A. Sperling, Obesity in Adolescents and Youth: The Case for and against Bariatric Surgery. J Pediatr, 2019. 207: p. 18-22.
  64. Michalsky, M., et al., ASMBS pediatric committee best practice guidelines. Surg Obes Relat Dis, 2012. 8(1): p. 1-7.
  65. Sperling MA, G.A., Monogenic Forms of Diabetes, in Diabetes in America. 2016. p. 7-1-7-27.
  66. Yang, Y. and L. Chan, Monogenic Diabetes: What It Teaches Us on the Common Forms of Type 1 and Type 2 Diabetes. Endocr Rev, 2016. 37(3): p. 190-222.
  67. Flannick, J., S. Johansson, and P.R. Njolstad, Common and rare forms of diabetes mellitus: towards a continuum of diabetes subtypes. Nat Rev Endocrinol, 2016. 12(7): p. 394-406.
  68. Bonnefond, A. and P. Froguel, Rare and common genetic events in type 2 diabetes: what should biologists know? Cell Metab, 2015. 21(3): p. 357-68.
  69. Steele, A.M., et al., Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA, 2014. 311(3): p. 279-86.
  70. Fajans, S.S. and G.I. Bell, MODY: history, genetics, pathophysiology, and clinical decision making. Diabetes Care, 2011. 34(8): p. 1878-84.
  71. Pihoker, C., et al., Prevalence, characteristics and clinical diagnosis of maturity onset diabetes of the young due to mutations in HNF1A, HNF4A, and glucokinase: results from the SEARCH for Diabetes in Youth. J Clin Endocrinol Metab, 2013. 98(10): p. 4055-62.
  72. Shields, B.M., et al., Population-Based Assessment of a Biomarker-Based Screening Pathway to Aid Diagnosis of Monogenic Diabetes in Young-Onset Patients. Diabetes Care, 2017. 40(8): p. 1017-1025.
  73. Carmody, D., et al., GCK-MODY in the US National Monogenic Diabetes Registry: frequently misdiagnosed and unnecessarily treated. Acta Diabetol, 2016. 53(5): p. 703-8.
  74. Johansson, B.B., et al., Targeted next-generation sequencing reveals MODY in up to 6.5% of antibody-negative diabetes cases listed in the Norwegian Childhood Diabetes Registry. Diabetologia, 2017. 60(4): p. 625-635.
  75. Ushijima, K., et al., Comprehensive screening for monogenic diabetes in 89 Japanese children with insulin-requiring antibody-negative type 1 diabetes. Pediatr Diabetes, 2017.
  76. Patel, K.A., et al., Type 1 Diabetes Genetic Risk Score: A Novel Tool to Discriminate Monogenic and Type 1 Diabetes. Diabetes, 2016. 65(7): p. 2094-2099.
  77. Hattersley, A.T. and K.A. Patel, Precision diabetes: learning from monogenic diabetes. Diabetologia, 2017. 60(5): p. 769-777.
  78. Fuchsberger, C., et al., The genetic architecture of type 2 diabetes. Nature, 2016. 536(7614): p. 41-47.
  79. Rich, S.S., Diabetes: Still a geneticist's nightmare. Nature, 2016. 536(7614): p. 37-8.
  80. Hattersley, A.T., et al., ISPAD Clinical Practice Consensus Guidelines 2018: The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes, 2018. 19 Suppl 27: p. 47-63.
  81. De Franco, E., et al., The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study. Lancet, 2015. 386(9997): p. 957-63.
  82. Bowman, P., et al., Neuropsychological impairments in children with KCNJ11 neonatal diabetes. Diabet Med, 2017. 34(8): p. 1171-1173.
  83. Beltrand, J., et al., Sulfonylurea Therapy Benefits Neurological and Psychomotor Functions in Patients With Neonatal Diabetes Owing to Potassium Channel Mutations. Diabetes Care, 2015. 38(11): p. 2033-41.
  84. Thurber, B.W., et al., Age at the time of sulfonylurea initiation influences treatment outcomes in KCNJ11-related neonatal diabetes. Diabetologia, 2015. 58(7): p. 1430-5.
  85. Pearson, E.R., et al., Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med, 2006. 355(5): p. 467-77.
  86. Rabbone, I., et al., Insulin therapy in neonatal diabetes mellitus: a review of the literature. Diabetes Res Clin Pract, 2017. 129: p. 126-135.
  87. Johnson, M.B., et al., Recessively Inherited LRBA Mutations Cause Autoimmunity Presenting as Neonatal Diabetes. Diabetes, 2017. 66(8): p. 2316-2322.

Fungi and Endocrine Dysfunction

ABSTRACT

 

Fungi are ubiquitous microbes and form a fraction of the symbiotic human microbiome. Transition from normal commensals to opportunistic mycoses can occur in immunocompromised hosts. Endemic mycoses are caused by fungi that are acquired from environmental sources. Fungal infections can be classified based on the depth of tissue invasion. Superficial diseases are limited to skin, nails, and mucous membrane while systemic dissemination can affect multiple organs including endocrine glands. Fungal involvement of the adrenals, pituitary, thyroid, pancreas, and gonads is well recognized. On the other hand, individual with diabetes mellitus and Cushing’s syndrome are susceptible to fungal disease as a result of immune dysfunction. Mucormycosis, candidiasis, and dermatophytosis occur more commonly in diabetes. Exogenous as well as endogenous Cushing’s syndrome is another endocrine disorder that predisposes to systemic fungal diseases. High index of suspicion is necessary to recognise these infections as clinical manifestations can be masked due to the anti-inflammatory properties of glucocorticoids. Autoimmune polyendocrine syndrome type I (APS-1) is a unique genetic disease where autoimmune damage predisposes to chronic mucocutaneous candidiasis (CMC) and a multitude of endocrine anomalies. Antifungal agents like azoles and polyenes can adversely affect the normal functioning of various endocrine pathways. Errors in diagnosis and treatment of the fungal infections of the endocrine glands can be critical. Equally important is to identify the various fungal diseases occurring in diabetes and other endocrine disorders. Conditions that predispose to fungal diseases such as diabetes and immunosuppressed states in organ-transplant recipients are becoming increasingly prevalent. Understanding of the critical interplay between the endocrine system and fungal pathogens are imperative for optimal patient outcomes in modern medicine.

INTRODUCTION

 

Fungi are classified as a separate kingdom that consists of single-celled or complex multicellular organisms. They are heterotrophs and unlike autotrophic plants, fungi lack chlorophyll and cannot synthesize their own food. They acquire nutrients from the surrounding media by osmosis.

 

Fungi are ubiquitous, transient, or persistent human colonizers which form the fungal microbiota or mycobiome. The human microbiota consists of a diverse array of microorganisms such as viruses, bacteria, fungi, protozoa, and parasites that reside in and around the human body. Fungi comprise ≤0.1% of the total human microbiota, but it still plays a crucial role in human health and disease (1).

 

Fungal species have complex interactions with the human host, which can be viewed as a spectrum of symbiotic relationships. The association can be mutualistic where it is advantageous to both, or commensal where only one profits but the other is unharmed.  On the other hand, the connection can be parasitic where the fungi are benefitted with a damaging effect on the human host, or amensalistic where one organism is harmed but the other remains unaffected. These human fungal symbionts can transition from commensalism to parasitism within the body. Immune dysfunction is one of the common factors that influence this conversion. Endocrine diseases like diabetes mellitus, Cushing’s syndrome, and autoimmune polyglandular syndrome type 1 (APS1) are prone to fungal infections due to immune dysfunction.

 

The prevalence of superficial fungal infection is 20-25% (2). On the other hand, fungal infections tend to spread in individuals with low immunity such as patients with cancer or acquired immunodeficiency syndrome (AIDS) and recipients of immunosuppressive drugs. The reported incidence of invasive fungal disease is 5.9 cases per thousand per year (3). The dissemination may affect various endocrine glands leading to their dysfunction, the adrenal gland being the one most commonly involved. Endocrine system involvement in fungal infections would extend to the adverse effect of various antifungal therapy too. Azoles are the most frequently described class affecting the endocrine system and, adrenal glands and gonads are their primary targets.

 

The diverse aspects of this complex relationship between fungi and the endocrine system are described in this chapter.

 

TYPES OF FUNGAL INFECTION

 

Fungal infections have been classified based on both anatomic location and epidemiology. They can also be classified on the basis of morphological structure of the fungus.

 

Anatomical Categories

 

MUCOCUTANEOUS INFECTIONS

 

Mucocutaneous infections is a heterogeneous group characterized by infections of the skin, mucous membranes, and the nails. These infections are confined to the cutaneous surface, with little propensity for systemic dissemination. The effect can vary from mild to severe depending on the extent of involvement but are rarely fatal.

 

DEEP ORGAN INFECTIONS

Fungal infections can sometimes cause deep tissue involvement and have the potential for hematogenous and systemic spread. Dissemination of fungal infections is usually observed in immunocompromised conditions. If untreated, deep organ or systemic fungal affection can be fatal.

 

Epidemiological Categories

 

ENDEMIC MYCOSES

 

Endemic mycoses include infections caused by fungi that do not belong to the normal human microbiota but rather are acquired from environmental sources. In endemic mycosis, deep organ infection is almost exclusively caused by inhalation, whereas cutaneous disease is most often caused by direct contact with soil but can also occasionally result from hematogenous dissemination. Dermatophytid fungi are mainly acquired by environmental contact however, human-to-human transmission has been reported. Examples of endemic mycoses include coccidioidomycosis, paracoccidioidomycosis, histoplasmosis, blastomycosis, penicilliosis, phaeohyphomycosis, sporotrichosis, and adiaspiromycosis.

 

OPPORTUNISTIC MYCOSES

 

Opportunistic fungi can be normal human microbiota components, but in the immunocompromised state, these organism transition from harmless commensals to invasive pathogens. These fungi invade the host from the usual sites of colonization, typically the mucous membranes or the gastrointestinal tract. Typical examples are candidiasis, aspergillosis, mucormycosis (zygomycosis), cryptococcosis, scedosporiosis, trichosporonosis, fusariosis, and pneumocystosis. Fungi that are reported to affect the various endocrine glands are shown in table 1.

 

Table 1. Fungi Affecting Specific Endocrine Glands

Type of fungus

Organs affected     

Aspergillosis

Pituitary, Thyroid, Pancreas, Adrenal

Zygomycosis

Thyroid

Candidiasis

Pituitary, Thyroid, Pancreas, Testis

Cryptococcosis

Thyroid, Pancreas, Adrenal, Testis

Histoplasmosis

Thyroid, Adrenal, Ovaries,

Blastomycosis

Testis, Ovaries

Coccidioidomycosis

Thyroid, Adrenal,

Paracoccidioidomycosis

Thyroid, Adrenal

Pneumocystis jirovecii

Pituitary, Thyroid, Parathyroid, Pancreas, Adrenal

 

Based on Morphology

 

YEASTS

 

Yeast are found as single rounded cells or as budding organisms. Examples are Saccharomyces cerevisiae, Candida albicans, and Leucosporidium frigidum.

 

MOLDS

 

Molds grow in filamentous forms called hyphae both at room temperature and in invaded tissue. The common molds are aspergillus (A. fumigatus, A flavus, and A brasiliensis), penicillium and rhizopus.

 

DIMORPHIC

 

Dimorphic fungi grow as yeasts or large spherical structures in the tissue but as filamentous forms at room temperature in the environment. These include histoplasma (H. capsulatum), blastomyces (B. dermatitidis), paracoccidioides (P. brasiliensis), coccidioides (C. immitis), penicillium (P. marneffei), and sporothrix (S schenckii).

Figure 1. Classification of Fungal Infections

FUNGAL DISEASES OF MAJOR ENDOCRINE GLANDS

 

Fungal infections are more prevalent in the immunocompromised state (table 2). There is a tendency for fungal infections to disseminate in such cases and affect endocrine organs like the pituitary, thyroid, parathyroid, pancreas, adrenal glands, and gonads. The involvement of these endocrine glands may lead to deficient hormone secretion. The clinical manifestations, diagnosis, and management of fungal infection of the major endocrine glands are discussed below.

 

Table 2. Conditions Predisposing to Systemic Fungal Infections

A.    Endocrine diseases

1.     Diabetes mellitus

2.     Cushing’s syndrome

3.     Autoimmune polyendocrine syndrome-1

4.     STAT5b deficiency (Congenital Insulin-like Growth Factor-1 Deficiency)

B.    Immunosuppressed states

1.     Cancer

2.     Acquired immunodeficiency syndrome

3.     Acute leukemia

4.     Hematopoietic stem cell transplant recipients

5.     Solid-organ transplant recipients

6.     Recipients of immunosuppressive drugs in conditions like connective tissue diseases

 

Pituitary Fungal Infections

 

ETIOLOGY

 

Pituitary infections or abscesses are rare and account for less than 1% of pituitary lesions (4). Even among them, fungal infections are extremely unusual and occur predominantly in immunocompromised states. The mode of spread could be hematogenous, extension from adjacent structures like meninges, sphenoid sinus, cavernous sinus, and skull base, or iatrogenic during transsphenoidal procedures. Fungal infection of the pituitary can occur in the presence of underlying lesions like pituitary adenoma, Rathke’s cleft cyst, etc. Cushing’s syndrome, resulting from an adrenocorticotrophic hormone (ACTH) secreting pituitary adenoma, itself causes immunosuppression and further predisposes to fungal disease (5). Aspergillus is the most frequently reported fungal infection of the pituitary (6–8). Other fungi described to infect the pituitary include candida (9,10), Pneumocystis jirovecii (in HIV/AIDS) (11,12), and coccidia (13). In a review of 13 cases of pituitary aspergillus infection, five were immunosuppressed (14).

 

CLINICAL FEATURES

 

The clinical presentation of fungal infection of the pituitary can be variable (table 3).  Symptoms from mass effects such as headache, visual disturbances (due to optic chiasma compression), and ophthalmoplegia are the usual presenting features. Features suggestive of infection, such as fever, leukocytosis, and meningismus were absent in most of the reported cases (8,15,16). Aspergillus is known to cause angioinvasion and vasculitis, and thus can be additionally associated with features arising from cerebrovascular infarcts (8,14). Pituitary insufficiency can acutely manifest as hypotension and shock primarily from secondary hypoadrenalism (9). Gonadotrophin and other hormone secretion can be affected as a delayed sequalae,  but such reports are very rare (17). Pituitary stalk compression can induce hyperprolactinemia (18). Diabetes insipidus (DI) occur more frequently than seen with pituitary adenomas (10).

 

Table 3. Clinical Profile of Recently Reported Cases of Pituitary Aspergillus Infection

Author, year

Clinical setting

Symptoms

Diagnosis

Management/

outcome

Moore, 2016 (8)

74-year old male,

CAD, CKD, AHA hypertension

Right eye pain, headaches, 10 months of worsening left hemiparesis

 

Imaging - right ICA occlusion, acute right pontine stroke, smaller infarcts in the right MCA territory

Fatal outcome, autopsy findings revealed fungal hyphae in pituitary

Choi, 2021(15)

75-year old male, DM, hypertension, lung aspergillosis

Headache, visual disturbance, hyponatremia

MRI - bilateral sphenoid sinusitis and pituitary involvement, transsphenoidal biopsy demonstrated invasive aspergillus

Endoscopic debridement of sinuses. Oral voriconazole given, gradual improvement

Saffarian,2018 (16)

60-year old male

DM, hypertension, sphenoid aspergilloma

Headache, progressive visual loss, 4thcranial nerve palsy

MRI findings, endoscopy by nasal approach demonstrated aspergillus in biopsy

Endoscopic drainage, intravenous amphotericin, responded to treatment

Ouyang, 2015 (18)

55-year old female,

no comorbidities

Headache, dizziness, and decreased visual acuity

 

MRI - sellar and sphenoid sinus

mass

Prolactin - 815 ng/mL

Transnasal, transsphenoidal removal of the mass and oral voriconazole – resolution of symptoms

 

Vijay-vargiya, 2013 (14)

68-year old female,

kidney transplant recipient

Headache, left temporal hemianopsia, ptosis.

MRI – sellar mass

Intraoperative frozen

section showed organisms consistent with aspergillus

Transsphenoidal resection, voriconazole, Developed ACA ischemic stroke, died.

CAD – coronary artery disease, AHA – autoimmune hemolytic anemia, ICA – internal carotid artery, MCA – middle cerebral artery, ACA – anterior cerebral artery, DM – diabetes mellitus, MRI – magnetic resonance imaging

 

DIAGNOSIS

 

Fungal pituitary infections usually present with symptoms of headache, visual disturbance, and ophthalmoplegia and are often misdiagnosed as tumors (14). Identification of a mass in the sellar region in an immunocompromised state should raise suspicion of fungal etiology.  T1-weighted magnetic resonance imaging (MRI) of fungal abscess of pituitary shows nonspecific isointensity or hypointensity (4). Pituitary abscess of any etiology including fungal may demonstrate peripheral rim enhancement and calcifications on T2-weighted images. Low signals due to iron deposition are however indicative of fungal involvement (19). Involvement of the adjacent sinuses is another pointer for fungal disease (15,16). It is difficult to distinguish fungal pituitary infections from intrasellar bacterial infections and tumors, and the diagnosis is often confirmed during surgery or autopsy. Histopathological examination can reveal hyphae and fungal spores. Silver impregnation stains such as Grocott or Gomori methenamine silver, fungal culture, or fungal polymerase chain reaction (PCR) can confirm the diagnosis (4). Serum 1,3-β-D-glucan is positive in a broad range of invasive fungal infections, including candida (19). Serum galactomannan is however, a specific marker for invasive aspergillosis (20).

 

TREATMENT

 

Treatment includes antifungal therapy and drainage of the abscess by transsphenoidal endoscopic approach (14). Craniotomy is discouraged due to fear of intracranial dissemination. Deficiency of pituitary hormones may necessitate replacement (9). Voriconazole is the preferred therapeutic agent for aspergillus infection. Other medical options are liposomal amphotericin B, posaconazole, isavuconazole, and echinocandins (21). The recommended dose of voriconazole for central nervous system (CNS) aspergillosis is intravenous loading with 6 mg/kg every 12 hour for two doses followed by 4 mg /kg every 12 hour. The oral loading dose is 400 mg every 12 hour for two doses, followed by 200 mg twice daily (22). Oral treatment may be required for months. The exact duration of therapy is not established and depends on the clinical parameters. Antifungal therapy for other varieties of fungus should be administered as per standard practice. Mortality rates are high in disseminated disease with vascular invasion, immunosuppressed state, and in cases of a delayed diagnosis (14).

 

Thyroid Disorders

 

ETIOLOGY

 

Infections of the thyroid are rare as its rich blood supply, iodine content, and capsule are protective against microbial invasion (23). Fungi form a small subset among the microbial pathogens infecting the thyroid. A. fumigatus is the predominant fungi in general, whereas P. jirovecii is the most common cause of fungal thyroiditis in patients with AIDS (24,25). Table 4 enumerates the fungal infections reported to infect the thyroid. These infections are primarily seen in immunocompromised patients and usually is a part of disseminated infection. Both hematogenous and lymphatic spread can occur.  Direct invasion of the thyroid by fungal infection is also reported. Mycotoxin secreted by the fungus may affect thyroid function, however the evidence in humans is not definitive (26).

 

Table 4. Predisposing Conditions Where Fungus Affects the Thyroid Gland

Type of fungus

Predisposing condition

Aspergillus

Organ transplant (27,28), AML (29), ALL (30),  MDS (31), NHL (32), SLE (24,33), cryoglobulinemic vasculitis (34), AIDS, normal immune status with MNG (35)

Pneumocystis

AIDS (25), Thymic alymphoplasia (36)

Candida

ALL (37), AML (38)

Coccidiodes

SLE on corticosteroids (39), sarcoidosis on corticosteroids, PAN on corticosteroids (40)

Histoplasmosis

NHL (41)

AML – acute myeloid leukemia, ALL – acute lymphoblastic leukemia, MDS – myelodysplastic syndrome, NHL- Non-Hodgkin’s Lymphoma, SLE- systemic lupus erythematosus, AIDS – acquired immunodeficiency syndrome, MNG – multinodular goiter, PAN – polyarteritis nodosa

 

CLINICAL FEATURES

 

Fungal infection of the thyroid usually occurs in presence of underlying critical illness. The symptoms of thyroid infection can get masked by the primary disease. Thyroid involvement can be often detected post-mortem in cases of disseminated fungal disease (42). Common clinical presentations include pain, swelling of the thyroid gland, and fever, often mimicking subacute thyroiditis. In severe cases, thyroid enlargement may cause dysphagia and respiratory distress due to esophageal and tracheal obstruction, respectively (25,42,43). Fungal thyroiditis typically follows the course of a brief phase of thyrotoxicosis followed by hypothyroidism. Recovery of thyroid function generally takes place in weeks to months. Sick euthyroid syndrome, which sometimes occurs in disseminated fungal infections, may confound thyroid function testing. The clinical presentation of different varieties of fungal infections is similar.

 

Aspergillus

 

A review of 28 cases of aspergillus thyroiditis by Tan et al. revealed that 12 (43%) patients had a primary thyroid infection. The rest had aspergillus infection elsewhere (usually lungs and airways). Fever, dyspnea, and neck swelling were the usual presentation. Dysphagia and airway obstruction resulted from mass effect and was fatal in two cases. The overall mortality rate was high (64%) (24).

 

Pneumocystis

 

Zavascki et al. described 15 cases of P. jirovecii thyroiditis. Most of the cases were reported in individuals with AIDS. It should be suspected if neck pain and swelling occur in presence of a CD4 count < 200/µL. Compressive symptoms such as odynophagia, dysphagia, dysarthria, and hoarseness have been reported. Extra-thyroid disease was present in 53% (8/15) of cases and documented usually on post-mortem studies. Most of the cases were euthyroid, three were hypothyroid, and one developed transient thyrotoxicosis (25).

 

Others

 

There are reports of infection of the thyroid with candida, histoplasma, coccidiodes, and, paracoccidiodes in immunocompromised hosts (37–41). The different varieties of fungal thyroiditis are clinically indistinguishable from each other.

 

DIAGNOSIS

 

Thyroid infection should be suspected in immunocompromised hosts who present with swelling and pain in the region of the thyroid gland. The thyroid involvement not uncommonly remains asymptomatic and gets detected post-mortem (42). Imaging of the neck by ultrasonography can be useful to define the morphology of the lesion. Computed tomography of the chest additionally identifies fungal lesions in the lungs, the usual site of primary or secondary infection. Fungal staining and culture of the lesion obtained by fine needle aspiration (FNA) of the thyroid gland can confirm the diagnosis. Results of thyroid function testing can be normal or may reveal thyrotoxicosis or hypothyroidism.

 

TREATMENT

 

Antifungal therapy is the mainstay of treatment. Voriconazole is the first line agent for invasive aspergillus infection. Adding echinocandin (capsofungin or antidulafungin) along with voriconazole may provide marginally better outcomes in patients who are immunocompromised (44,45). Cotrimoxazole is the preferred therapy for pneumocystis infection. The choice of antifungal therapy depends on the type of fungus and the prevalent pattern of antifungal resistance. Surgical debridement may be required especially if there is a possibility of tracheal compression due to mass effect. Symptomatic treatment may be required in the thyrotoxic phase resulting from acute damage to the gland. The thyroid gland fails to recover in a minority of patients. They should be treated with thyroid hormone replacement. Outcome of fungal thyroiditis has improved over the last two decades with advances in antifungal therapy (43).  

 

Disorders of Calcium Metabolism

 

Fungal infections can alter calcium and vitamin D metabolism. The common metabolic bone disorders are described in the following section.

 

MONOCYTE 1α HYDROXYLASE MEDIATED HYPERCALCEMIA

 

Etiology and Pathogenesis

 

Conversion to the active 1,25-dihydroxyvitamin D [1,25(OH)2D] from 25-hydroxyvitamin D [25(OH)D] occurs primarily in the kidney. The renal enzyme 25(OH)D-1α hydroxylase (CYP27B1) responsible for the conversion, is tightly regulated by parathyroid hormone (PTH), fibroblast growth factor 23 (FGF-23), and the serum 1,25(OH)2D concentration. The activated mononuclear cells and macrophages also exhibit 25(OH)D-1α-hydroxylase activity. The 1,25(OH)2D synthesized in these cells normally exert a paracrine effect on growth and differentiation of cells. In granulomatous disorders, such as sarcoidosis, tuberculosis, and fungal infections, the 1,25(OH)2D production in monocytes is dysregulated resulting in hypercalcemia. The monocyte 25(OH)D-1α-hydroxylase is resistant to the regulatory mechanisms and the lack of calcium-mediated negative feedback predisposes to hypercalcemia  (46). PTH-independent hypercalcemia is described in chronic fungal infections, such as histoplasmosis, coccidioidomycosis, para-coccidioidomycosis, candidiasis, cryptococcosis, and pneumocystis.

 

Clinical Profile

 

The fungal infections associated with 1α-hydroxylase mediated hypercalcemia can occur in both immunocompromised and immunocompetent hosts. In a review summarizing 16 cases of histoplasmosis induced hypercalcemia, 68.7% (11/16) were immunosuppressed. The common presentations were with polyuria, constipation, altered sensorium, and renal insufficiency (47). Hypercalcemia is also reported in cryptococcus and pneumocystis infections in individuals with HIV/AIDS (48–50). Hypercalcemia can be an early marker of pneumocystis pneumonia in renal transplant recipients (51,52).  

 

Laboratory Features

 

Patients present with elevated serum calcium and phosphate levels, suppressed PTH values, normal 25(OH)D, and increased 1,25(OH)2D concentrations. Serum angiotensin-converting enzyme (ACE) levels can be elevated (47).

 

Treatment

 

Hypercalcemia resolves with resolution of the infection after institution of successful antifungal therapy. Hydration, calcitonin, and bisphosphonates can be considered to lower calcium till the effect of antifungal medication occurs (47). Steroids can be used in resistant cases but should be initiated only under appropriate antifungal coverage. Fatalities have been reported when the cases have been misdiagnosed as sarcoidosis and steroids initiated without antifungal drugs (53,54). Some cases show transient worsening of hypercalcemia probably mediated by immune reconstitution inflammatory syndrome (55). Also, initiation of antiretroviral therapy in patients with HIV/AIDS infected with cryptococcus, might cause hypercalcemia. This may be due to restoration of granulomatous host response (56).

 

PARATHYROID HORMONE REALTED PROTEIN (PTHrP) MEDIATED HYPERCALCEMIA

 

Coccidioidomycosis infection is associated with hypercalcemia. However, the mechanism of hypercalcemia in coccidioidomycosis is not related to autonomous 1,25(OH)2D production. It could be due to osseous coccidioidomycosis in some cases, but in the majority of cases it occurs without bony lesions. Serum PTH levels and 1,25(OH2)D levels were either suppressed or normal (57).  Expression of PTHrP by the granulomatous tissue has been documented in coccidioidomycosis. The serum PTHrP levels are elevated in cases with hypercalcemia and presumed to be the possible mechanism. The PTHrP levels return to normal along with resolution of hypercalcemia after successful antifungal treatment  (58).

 

OTHER DISORDERS OF CALCIUM METABOLISM

 

Histoplasmosis-induced hypercalcemia has been postulated to result from excess expression and secretion of osteopontin by histiocytes in granulomas (59). Osteopontin can activate osteoclasts and subsequently lead to bone resorption (60). However, currently there is insufficient evidence to support this hypothesis. 

 

Hypoparathyroidism has also been described in HIV/AIDS with pneumocystis infiltrating the parathyroid glands. It causes hypocalcemia and hyperphosphatemia (61).

 

Fungal Infection of the Adrenal Gland

 

The adrenal gland is the commonest endocrine organ to be affected by infections including mycosis. Adrenal fungal infection can be asymptomatic and get detected as an incidental finding during radiological imaging, or can manifest with symptoms of adrenal insufficiency (62,63).

 

ETIOLOGY AND PATHOGENESIS

 

Unlike the other endocrine organs, isolated adrenal involvement can be seen as a manifestation of endemic mycoses in immunocompetent hosts by histoplasmosis, paracoccidioidomycosis, blastomycosis, and other fungal organisms (64–66). The susceptibility to develop primary adrenal infection or disseminated fungal disease is however more often seen in the immunocompromised individuals with HIV/AIDS, or in those receiving immunosuppressive therapy such as solid organ transplant recipients (67). Predisposition of the adrenal glands to fungal infections is postulated to be due to suppression of cell-mediated local immunity caused by high local glucocorticoid levels (68). More often than isolated involvement, the adrenal gland is involved as a part of disseminated infection. Histoplasmosis and paracoccidioidomycosis are the commonest fungal infections reported to have adrenal disease at autopsy (67,69).

 

Affinity for different adrenal zones might vary for different fungal infections. Paracoccidioides species has affinity for zona reticularis as well as zona glomerulosa leading to decreased dehydroepiandrosterone sulfate and aldosterone levels, respectively (59,70,71). The large fungal cells cause embolic infection of the small vessels of the gland subsequently leading to endovasculitis, granuloma formation and caseous necrosis (67,72). In patients with histoplasmosis, zona fasciculata and reticularis are preferentially affected owing to the presence of high concentration of cortisol (73). Vasculitis of downstream medullary vessels starting from zona fasciculata induce glandular destruction and subsequent caseation necrosis  (68,74).

 

CLINICAL FEATURES

 

The spectrum of manifestations of fungal adrenal involvement can vary from asymptomatic cases detected incidentally to frank adrenal crisis. Occasionally, adrenal involvement can get masked by the disseminated fungal disease or the underlying immunocompromised state (67). Many of the patients despite bilateral adrenal infection do not develop adrenal insufficiency, as destruction of more than 90% of adrenal cortex is required for the disease to manifest (59). Some studies have observed lower prevalence of adrenal involvement in immunocompromised hosts, presumably due to the inability to launch a granulomatous response in the gland (75,76).

 

Addison’s disease is most frequently reported with histoplasmosis and paracoccidioidomycosis, given their high affinity for adrenal glands. In a review of 252 cases of adrenal histoplasmosis, adrenal hypofunction was confirmed in 41.3%. Almost all the cases were secondary to chronic disseminated pulmonary histoplasmosis although isolated adrenal involvement has also been reported (77). A study of 546 cases of paracoccidioidomycosis from Brazil documented adrenal involvement in only 5% (n = 27) (78). Another review revealed partial adrenal insufficiency in 33–40% of cases, and frank symptoms in 3–10% cases (79).  Patients with diminished adrenal reserve often require glucocorticoid supplementation during periods of stress or after initiating antifungal agents known to affect steroidogenesis. There are reports of blastomycosis, pneumocystis, and cryptococcus causing adrenal insufficiency as well (80–82). The clinical features of primary adrenal insufficiency include fatigue, loss of appetite, weight loss, orthostatic hypotension, and hyperpigmentation (66,83).

 

DIAGNOSIS

 

Fungal infection of the adrenal glands can be asymptomatic and detected incidentally on abdominal imaging. Radiographically bilateral symmetric adrenal enlargement is seen with histoplasmosis whereas paracoccidioidomycosis and blastomycosis cause asymmetric and occasionally unilateral involvement (81,84–86). Other radiographical features include peripheral enhancement, central hypoattenuation, preserved contour, and calcifications (66,67). These features help to differentiate from other disorders such as metastatic disease where the adrenal contour is distorted and autoimmune adrenalitis, where the glands are atrophic (66,67,87,88).

 

The laboratory findings such as hyponatremia and hyperkalemia are often seen but the diagnosis of adrenal insufficiency is confirmed with the short Synacthen test (SST) or cosyntropin test (250 ug of Synacthen, im or iv) in chronic and stable cases. In a patient with suspected Addisonian crisis, a blood sample collected for estimation of serum cortisol and adrenocorticotrophic hormone (ACTH) before initiating glucocorticoid replacement can be helpful. A formal evaluation by SST can be performed later. Simultaneous estimation of plasma renin and aldosterone to determine mineralocorticoid reserve can be considered. (66).

 

The confirmation of fungal etiology might necessitate fungal staining or culture of the biopsied material. In disseminated disease, a more accessible site like skin lesion or affected lymph node can be biopsied instead of the adrenal gland.

 

MANGEMENT AND PROGNOSIS

 

Initiation of antifungal therapy at the earliest is essential to salvage adrenal function. Recovery has been reported in a few cases with histoplasmosis and paracoccidioidomycosis (59). However, frequently adrenal insufficiency is irreversible and lifelong glucocorticoid replacement is required. Mineralocorticoid replacement with fludrocortisone may additionally be necessary (83). Onset of  adrenal insufficiency in paracoccidioidomycosis can occur after initiation of antifungal therapy from the fibrosis that occurs during recovery (79,89).

 

Fungal Infection of the Pancreas

 

The pancreas is normally resistant to fungal infection. Fungal affection of the pancreas usually occurs in an inflamed gland in the presence of underlying necrosis. Although rare, the prevalence of fungal pancreatitis is on the rise.

 

ETIOLOGY AND PATHOGENESIS

 

Candida (C. albicans and C. glabrata) is the most common etiology responsible for fungal pancreatic infections (90). Pneumocystis, aspergillosis, and cryptococcosis have also been reported to affect the pancreas (91–93). The risk factors for fungal infection are necrotizing pancreatitis, use of broad-spectrum antibiotics, abdominal surgery, prolonged total parenteral nutrition, indwelling catheters, and an immunosuppressed state. The mode of spread could be translocation from the gut, hematogenous spread, or external seeding (90).

 

CLINICAL COURSE AND MANAGEMENT

 

The clinical features of fungal infection of the pancreas are non-specific. Abdominal pain, fever, and a palpable abdominal mass can occur (94). Most cases of fungal pancreatitis occur on the backdrop of recent necrotizing pancreatitis (90,94). In a study of 92 patients with necrotizing pancreatitis, 22 (24%) had evidence of candida infection in the surgical necrosectomy material (95). Candida was demonstrated in 27% of aspirates from walled-off necrosis occurring after acute pancreatitis (96).  Rare cases of recurrent pancreatitis from candida have also been described (97,98).

 

Fungal culture and staining of percutaneous aspirates, or necrosed tissue obtained during surgery, are necessary to establish the diagnosis. Antifungal therapy and surgical drainage and debridement are the mainstay of therapy. Mortality rates are higher if candida infection is present (95).

 

 

Fungal Infection of the Testis

 

ETIOLOGY AND PATHOGENESIS

 

Fungal epididymo-orchitis can occur in isolation or as a part of disseminated infection. The fungi reported to infect testis and epididymis include candida, blastomycosis, histoplasma, aspergillus, and cryptococcus (99–103). Both C. albicans and C. glabatra can cause epididymo-orchitis by retrograde transport from infection in the urinary tract. Risk factors comprise diabetes mellitus, instrumentation of the urinary tract, urinary obstruction, or recent antibiotic usage (104). The majority of blastomycosis infections were associated with systemic diseases (105). Granulomatous epididymo-orchitis can also occur as a part of disseminated histoplasmosis in immunocompromised state (106).  

 

CLINICAL COURSE AND MANAGEMENT

 

Most patients present with unilateral or bilateral pain and swelling of the scrotum. Onset can be acute or insidious with duration of symptoms lasting for days to months (104). In contrast, bacterial infection is almost always unilateral with an acute onset of scrotal swelling, redness, and pain. Some fungal infections may remain asymptomatic and get detected on autopsy (102). Fungal epididymo-orchitis is also recognized as a cause of azoospermia and infertility (107). This is mainly due to direct gonadal invasion but can also be due to anti-sperm effects induced by fungi and by secreted mycotoxins (59). C. guilliermondii and C. albicans can affect sperm viability and motility (108). Antifungal agents are the mainstay of treatment. Surgery may be required in some cases.

 

Fungal Infection of the Ovary

 

ETIOLOGY AND PATHOGENESIS

 

Pelvic inflammatory disease (PID) refers to infection of the upper genital tract usually occurring in reproductive age females. A tubo-ovarian abscess (TOA) is a sequela of PID. It is a complex adnexal mass resulting from ascent of the infection through the fallopian tube (109). Though the common causative organisms are bacteria such as Chlamydia trachomatis and Neisseria gonorrhoeae, fungal infections are also recognized as an important etiological agent (110). It can also be a part of disseminated infection (111–113). C. albicans as well as other candida species such as C. glabrata and C. keyfr have been described to cause TOA (114–116). Intrauterine devices, diabetes, and morbid obesity are the typical risk factors (114,117). There are rare reports of female genital coccidioidomycosis (112,113,118).

 

CLINICAL COURSE AND MANAGEMENT

 

The usual presentation is that of a pelvic infection not responding to conventional antibiotics (117). Presenting symptoms can be dysmenorrhea, menstrual irregularities, menorrhagia, anovulation, and infertility. Occasional patients present with severe lower abdominal pain, fever and vomiting (116).  

 

Fusarium toxin zearalenone and its metabolite zearalenol can be present as a contaminant in cereals and usually enter the food chain as pesticide. It is a non-steroidal estrogen mycotoxin with strong affinity for estrogen receptors (119). The resulting hyperestrogenism has the potential to cause infertility by suppressing luteinizing hormone (LH) and progesterone secretion and also can have a carcinogenic effect on the breast (120).

 

FUNGAL INFECTIONS OCCURING IN ENDOCRINE DISORDERS

 

Individuals with certain endocrine disorders such as diabetes mellitus and Cushing’s syndrome are predisposed to fungal infections as a result of the associated immune dysfunction. Both pathogenic and opportunistic fungi can cause infection in these conditions. APS1 is an endocrine syndrome characterized by CMC (121). The common fungal infections occurring in individuals with endocrine dysfunction are discussed below. Other fungal infections like coccidioidomycosis and aspergillosis are also known to occur at a higher frequency in individuals with diabetes.

 

Fungal Infections in Patients with Diabetes

 

Diabetes is known to affect both innate and adaptive immunity. Hyperglycemia also induces critical alterations in cytokine signaling (122). Fungal infections in general occur at a slightly increased frequency in diabetes, especially if glycemic control is poor. However, certain fungal infections like mucocutaneous candidiasis and invasive mucormycosis have a strong association with diabetes (123).

 

CANDIDIASIS

 

Infection with candida is common in individuals with diabetes (124) . Genital candidiasis is often an indicator for undetected or poorly controlled diabetes. Increased hydrolytic enzyme activity and hydrophobicity along with altered biofilm formation have been proposed as possible mechanisms that favor candida infection in diabetes (125,126). The common sites and clinical characteristics of candida infection in diabetes are summarized in table 5.

 

Table 5. Candida Infections in Diabetes

Site

Usual species

Predisposing factors

Clinical features

Diagnosis

Treatment

Oral candidiasis

C. albicans

C. glabrata

C. tropicalis

C. krusei

C. dubliniensis C. parapsilosis

(124)

Uncontrolled hyperglycemia, dentures, xerostomia, inhaled corticosteroids (127)

Types of lesions: Pseudo-membranous

Hyperplastic

Erythematous

Atrophic (denture stomatitis)

Angular cheilitis

Median rhomboid glossitis (128)

Compatible clinical findings; Confirmation by Gram stain or KOH preparation or fungal culture of the scrapings (129)

Oral hygiene

Topical: Clotrimazole, miconazole, nystatin, amphotericin B suspension

Oral: Fluconazole, itraconazole

(129)

Vulvo-vaginal candidiasis

C. albicans

C. glabrata (124)

 

Uncontrolled hyperglycemia, pregnancy and hyper-estrogenemic state, SGLT2 inhibitor therapy, immunosuppression (130)

Thick white cottage cheese-like discharge, itching, pain, redness, burning, edema and dyspareunia

Clinical findings, Vaginal swab – acidic pH, KOH or fungal staining, fungal culture in selected cases

Glycemic control

Vaginal: Clotrimazole, miconazole, tioconazole, terconazole, butoconazole

Oral: Fluconazole (150 mg single dose ) (131)

 

Balanoposthitis

C. albicans

C. glabrata

Uncontrolled hyperglycemia,  SGLT2 inhibitor therapy, uncircumcised men, immunosuppression (132,133)

Sore, pruritic erythematous rash with small papules, erosions or dry dull areas with glazed appearance (134)

Clinical findings, KOH or fungal stain of scrapings in rare cases

Glycemic control

Topical: Clotrimazole, miconazole

Oral: Fluconazole (150 mg single dose), Itraconazole

Esophageal candidiasis

C. albicans,  C. dubliniensis (124)

Old age, HIV/AIDS, corticosteroid use, COPD, PPI use, esophageal dysmotility (135)

Odynophagia, dysphagia, retrosternal pain, usually associated with oral thrush (136)

Endoscopy - white mucosal plaque-like lesions. Biopsy – confirmatory. Culture rarely required (136)

Initial therapy: Oral fluconazole

Second-line therapy: Itraconazole,

voriconazole

isavuconazole,

echinocandin,

liposomal amphotericin B

Urinary tract candidiasis

C. albicans,

C. glabrata,

C. tropicalis (137)

Hyperglycemia, urinary retention and stasis, renal transplantation, long-term urinary catheterization, hospitalization (138)

Asymptomatic, symptoms of lower and upper urinary tract involvement mimic bacterial infection (139)

Urinalysis and culture of urine, Imaging when indicated (139)

Asymptomatic candiduria needs treatment in neutropenic patients, before urological procedures.

First line: Fluconazole

Second line: Flucytosine, amphotericin B (138)

Onychomycosis

C. albicans,

C. parapsilosis

C. tropicalis (124)

Age, nail disorders, frequent exposure to moisture (124)

Nail discoloration, subungual hyperkeratosis, onycholysis, splitting, and nail plate destruction

Clinical findings, KOH preparations, fungal cultures, histopathologic examination with a PAS stain and PCR testing (140)

Oral itraconazole treatment of choice.

Terbinafine might also be efficacious (141)

Systemic candidiasis

C. albicans,

C. parapsilosis, C. krusei,

C. tropicalis,

C. glabrata (142)

New onset hemodialysis, use of TPN, or receipt of broad-spectrum antibiotic (143)

Can vary from minimal fever to a full-blown sepsis

Blood culture. 1,3-β-d-glucan assay may assist in the diagnosis

Preferred therapy Echinocandin: anidulafungin, capsofungin, micofungin

Alternative: Amphotericin B

Step down therapy: Fluconazole if susceptibility results support (144)

KOH - potassium hydroxide, SGLT2 - Sodium-glucose cotransporter-2, COPD – chronic obstructive pulmonary disease, PPI – proton-pump inhibitor, PAS – Periodic Acid Schiff, PCR – polymerase chain reaction, TPN – total parenteral nutrition.

 

MUCORMYCOSIS

 

Mucormycosis refers to a group of infections caused by fungi of the order Mucorales present ubiquitously in the environment. Individuals with uncontrolled diabetes or those who are immunosuppressed are characteristically affected. The most common presentation is rhino-orbital-cerebral mucormycosis, though pulmonary, gastrointestinal, cutaneous, and renal infection can also occur (145). Several cases of mucormycosis have been reported recently following SARS COV-2 disease (146). Around 40% of the patients had received corticosteroids within the month before the diagnosis of mucormycosis. Diabetes with ketoacidosis (DKA) is 50% more likely to develop mucormycosis than without DKA. The prognosis is poor and mortality rates remain high. The rhino-orbital-cerebral form is characteristically associated with diabetes and detailed below.

 

Pathogenic Organisms

 

The pathogenic fungi belonging to order Mucorale customarily associated with human infections are Rhizopus, Mucor,and Lichtheimia (formerly Absidia and Mycocladus). The rarer pathogens include Rhizomucor, Cunninghamella, Apophysomyces, and Saksenaea (147).  Infection occurs presumably from inhalation of spores.

 

Pathogenesis

 

Patients with diabetes, defects in phagocytic function (such as neutropenia or glucocorticoid treatment), and/or elevated levels of free iron which supports fungal growth in serum and tissues are prone to mucormycosis. DKA is a risk factor for developing rhino-orbital-cerebral mucormycosis, as acidosis leads to dissociation of iron from sequestering proteins, which aids increased fungal survival and virulence (148). Moreover, the ketoacid -hydroxybutyrate facilitates fungal adherence and penetration into tissues, by increased expression of fungal receptors (149). Apart from ketoacidosis, hyperglycemia itself may contribute to the risk of mucormycosis by four possible mechanisms: (i) disruption of normal iron sequestration due to hyper-glycation of iron-sequestering proteins; (ii) phagocytic dysfunction; (iii) enhanced expression of a mammalian cell receptor (GRP78) that binds to Mucorales, enabling tissue penetration; (iv) enhanced expression of CotH, a Mucorales-specific protein that binds to  GRP78 and mediates host cell invasion (150). The risk factors for mucormycosis are summarized in table 6.

 

Table 6. Risk Factors for Mucormycosis

Uncontrolled diabetes mellitus especially if associated with ketoacidosis

Underlying malignancy receiving chemotherapy or immunotherapy

Solid organ transplant

Hematopoietic stem cell transplant

Treatment with deferoxamine

Iron overload

Corticosteroid therapy

Trauma or burns

Malnutrition

Coronavirus disease 2019

 

Clinical Features

 

Rhino-orbital-cerebral mucormycosis is the most common form of the disease whereas lung, gastrointestinal, renal, and cutaneous involvement are less frequent (145). Initial symptoms of rhino-orbital-cerebral mucormycosis include eye or facial pain and facial numbness followed by conjunctival suffusion and blurring of vision. Facial erythema with or without edema may be present. Fever occurs in only half of the cases (151). Black, necrotic eschar develops over the palate or in the nasal mucosa. In untreated cases, infection spreads from the ethmoid sinus to the orbit which involvement of extra-ocular muscles. It results in proptosis, typically with chemosis. Infection might further extend from the orbit to the frontal lobe of the brain via hematogenous route or contiguous dissemination. It may extend to cavernous sinus as well via venous drainage (147). The clinical features are summarized in table 7.

 

Table 7. Clinical Features of Rhino-Orbital-Cerebral Mucormycosis

Site

Symptoms

Signs

Paranasal sinuses

Nasal congestion, purulent nasal discharge or post-nasal drip, loss of smell, headache, pain over the sinuses

Swelling, redness, ulceration and blackening of overlying skin and nasal mucosa

Systemic features

Fever

Fever

Orbit

Red eyes, pain, visual blurring, loss of vision, bulging of eyes

Periorbital swelling, chemosis, proptosis, loss of visual acuity

Cavernous sinus

Headache, periorbital swelling and pain, diplopia, and visual loss

Periorbital swelling, chemosis, ptosis, proptosis, restricted or painful eye movement, diminished facial sensation

Palate

Ulceration, pain, swelling

Ulceration, eschar formation

Central nervous system

Headache, drowsiness, seizures, hemiparesis, obtundation, coma

Focal seizures, hemiparesis, altered sensorium

Vascular invasion

Black eschars over skin, nasal mucosa, palate and involved areas, symptoms related to stroke

Black eschars (from cutaneous necrosis), focal neurological deficit (also from mycotic aneurysm)

 

Diagnosis

 

Clinical features, mycological, and histological investigations and imaging with CT or MRI are necessary for establishing the diagnosis and assessing the extent of spread. If sinusitis is suspected, endoscopy should be performed. Histopathological examination of infected tissue demonstrates characteristic wide, thick walled, ribbon like, aseptate hyphal elements that branch at right angles. Fungal culture of specimens is strongly recommended for genus and species identification, and for antifungal susceptibility testing (145). PCR-based technique and matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) can assist to confirm fungal etiology if cultures are negative (145,152). MRI of the cranium including the sinuses and orbit should be done to delineate the extent of involvement (145). CT scan can help to assess the extent of bony erosion and can be considered if MRI is not readily available.

 

Treatment and Prognosis

 

Surgical debridement of the necrotic tissue in combination with intravenous lipid preparations of amphotericin B are the mainstay of therapy. It is also important to restore euglycemia and correct acidosis as soon as possible. The recommended dose of lipid formulation amphotericin B is 5mg/kg/day. There is evidence to support a higher dose of 10 mg/kg/day in cases of CNS involvement. There is no consensus on total duration of therapy but it usually takes weeks to months for completely cure these infections. It is critically important to monitor for adverse effects of amphotericin B especially nephrotoxicity and electrolyte imbalance. Posaconazole or isavuconazole can be considered as oral step down therapy, as salvage therapy, or if amphotericin B related adverse effects precludes its further use (145). Repeat surgery may be necessary if the infection progresses. Prognosis is poor especially if there is associated CNS involvement.

 

DERMATOPHYTES

 

Dermatophytosis are caused by filamentous fungi belonging to the genera Microsporum, Epidermophyton, and Trichophyton. Dermatophytes cause infection of skin, hairs, and nails and derive nutrition from keratin present in these tissues. Dermatophytosis is known to occur commonly in individuals with diabetes. Infection of the hair is referred to as tinea capitis (scalp) and tinea barbae (beard). Infection of the body surface in general is called tinea corporis while that of groin is known as tinea cruris.

 

Skin infection with dermatophytes occurring over the feet is called tinea pedis. It can cause micro-fissuring that may predispose to secondary bacterial infection and subsequently to diabetic foot. The other form of dermatophyte infection affecting feet is onychomycosis or tinea unguium (153). Tinea pedis and onychomycosis are commonly causes by the anthropophilic dermatophytes T. rubrum, T. interdigitale and E. floccosum (154). Uremic patients on hemodialysis more often have dystrophic nail changes and are at increased risk of developing onychomycosis (155). Dystrophic nails in onychomycosis look thick, brittle and discolored, often with a yellow shade. It may also lead to separation of the nail plate from the nail bed (onycholysis). Paronychial inflammation of the nail edge surrounding skin is a characteristic feature (156). Early recognition and treatment of tinea pedis and onychomycosis can prevent ominous complications like diabetic foot.

 

Clinical features along with KOH preparation of scrapings from affected area are usually adequate to establish the diagnosis. Treatment mainly includes topical and oral agents with activity against dermatophytes. The commonly applied topical agents includes azoles, allylamines, butenafine, ciclopirox, and tolnaftate. Oral therapy usually involves use of terbinafine, itraconazole or fluconazole (157).

 

Fungal Infections in Cushing’s Syndrome

 

The susceptibility of individuals with Cushing’s syndrome to fungal infection is well recognized. Both endogenous and exogenous hypercortisolism are associated with opportunistic fungal diseases. Hypercortisolism induces immune dysfunction by multiple mechanisms (158).  The major defects induced by excess cortisol are depicted in table 8. Among the subtypes of endogenous Cushing’s syndrome, fungal infections are more commonly seen in the syndrome of ectopic ACTH secretion. Propensity for fungal infections in exogenous Cushing’s syndrome depends on both, the intensity of glucocorticoid therapy and relative virulence of the offending fungus. With respect to glucocorticoids, it depends on administration route, dose, potency, and duration of treatment (159). The commonly reported fungal infections in Cushing’s syndrome are discussed below.

 

Table 8. Hypercortisolemia-Induced Immune Dysfunctions Increasing Susceptibility to Fungal Infections

Cell/Mediator

Dysfunction

Innate immunity

Neutrophils

Impaired neutrophil adherence to endothelium

Monocytes and macrophages

Decreased circulating monocytes

Decreased degranulation capacity

Decreased phagocyte function

Natural Killer cells

Suppressed cytotoxic activity

Adaptive immunity

T Cells

Lymphopenia due to a reduction in CD4+ subset

 

Influences the Th1/Th2 balance

 

Induces apoptosis in mature T lymphocytes

Cytokines

Cytokines

Down-regulates multiple cytokines by inactivating key proinflammatory transcription factors (e.g., NF kappa B)

CD - cluster of differentiation, Th – T helper cells, NF – nuclear factor

 

CANDIDIASIS

 

In immunocompromised states such as Cushing’s syndrome, candida species may cause superficial infections like cutaneous candidiasis, oropharyngeal candidiasis, esophagitis, or vulvovaginitis. Cases of candida endophthalmitis have also been described (160). It may also disseminate in the bloodstream to cause candidemia. Glucocorticoid may augment biological fitness of candida species, by enhancing its adhesion to epithelial cells. C. albicans is the most common species reported though infection with C. glabrata, C. parapsilosis and C. tropicalis can also occur (159).

 

ASPERGILLUS

 

Aspergillus is associated with invasive fungal infection in endogenous Cushing’s syndrome as well as in those receiving exogenous glucocorticoids (161) . Most common species to cause invasive infection are A. fumigatus, followed by A. flavus, A. terreus, and A. niger. The usual portal of entry for aspergillus is typically the pulmonary tract. However, later it might get disseminated systemically and severe cases requiring emergency bilateral adrenalectomy for control of hypercortisolism has been reported (162). Apart from immune dysfunction, glucocorticoids can induce alterations in the biology of aspergillus species to increase its invasiveness. For example A. fumigatus and A. flavusshowed increased growth on in-vitro exposure to pharmacological doses of hydrocortisone (163).

 

PNEUMOCYSTIS

P.  jirovecii is usually seen in immunocompromised patients. Severe P. jirovecii pneumonia even leading to fatal outcome are described in cases of endogenous Cushing’s syndrome (164). The infection is often unmasked once treatment for hypercortisolism is commenced. The restoration of immune response with lowering of cortisol levels presumably induce the inflammatory changes and result in manifest disease (165,166). A review of 15 cases of P. jirovecii pneumonia, reiterated the same observation of immune reconstitution related worsening of symptoms after treatment initiation. In 13 of these cases symptoms were triggered after cortisol-lowering therapy was started. Interestingly, all but one if these patients had ectopic Cushing’s syndrome. All the cases were characterized by severe hypercortisolemia and the outcome was fatal in 11 cases (167). Patients with Cushing’s syndrome, especially those with severe hypercortisolemia might benefit from prophylaxis with cotrimoxazole before beginning cortisol-lowering therapy.

 

CRYPTOCOCCOSIS

C. neoformans is another opportunistic infection where Cushing’s syndrome is a predisposing factor. The route of entry is inhalational. It may cause pneumonitis or disseminate systemically to cause more severe infections, such as meningitis and meningoencephalitis (168). Fatal cases have been reported (169,170). The presence of coexisting diabetes might further increase the risk of infection (171).

 

Glucocorticoid-induced immunosuppression has a few unique characteristics noted with cryptococcosis. For example, alveolar macrophage capacity to attach to and ingest is diminished by cortisone acetate, which potentially may lead to dissemination of the fungus (172). Moreover, chemotactic activity of cerebrospinal fluid toward polymorphonuclear (PMN) leucocytes and monocytes, is substantially reduced by glucocorticoid administration. This leads to lack of PMN leucocyte influx in cerebrospinal fluid and subsequent inability to eradicate fungi like C. neoformans with tropism for the CNS. Glucocorticoid-induced abnormalities of microglial cells further intensify this attenuation. Thus, individuals with hypercortisolemia are predisposed to cryptococcal meningitis (173).

 

HISTOPLASMOSIS

 

Pulmonary histoplasmosis has been reported in association with endogenous Cushing’s syndrome (174). Patients receiving glucocorticoids may develop primary or reactivated infections by endemic fungi (175). There are reports of pulmonary histoplasmosis after prolonged glucocorticoid therapy from non-endemic countries as well (176). H. capsulatum, the usual pathogen in most cases of histoplasmosis, enters through the respiratory tract and causes pulmonary histoplasmosis but can also disseminate to cause systemic infection. Pathological features of histoplasmosis are atypical in patients treated with glucocorticoids. Discrete granuloma formation is prevented by the anti-inflammatory properties of glucocorticoids (175).

 

OTHER FUNGAL INFECTIONS

 

Other fungal infections reported with hypercortisolemia are C. immitis, mucor, fusarium and blastomyces (159). Besides the heightened risk of fungal inspection in hypercortisolemia, the other concerning issue is masking of the signs and symptoms of infections due to the anti-inflammatory properties of glucocorticoids. Recognition of infections may be delayed in presence of hypercortisolemia, and a high index of suspicion is required for early diagnosis. Treatment of fungal infection must include prompt correction of hypercortisolism and aggressive antifungal therapy.

 

Chronic Mucocutaneous Candidiasis in Autoimmune Polyendocrine Syndrome Type 1

 

Autoimmune polyendocrine syndrome type 1 (APS1) is characterized by the classical triad of chronic mucocutaneous candidiasis (CMC), autoimmune hypoparathyroidism, and Addison’s disease. Two of the three classic features should be present to establish the diagnosis of APS1. However, there is a risk of the development of autoimmune diseases affecting almost every organ. APS1 is also known as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) with ectodermal dysplasia occurring in a third of the patients. Ectodermal dystrophy is not related to candidiasis (121). CMC commonly occurs sporadically secondary to AIDS, diabetes, and immunosuppressive treatment (177). C. albicans is the predominant pathogen but infection with other candida species is also described.

 

PATHOGENESIS

 

APS1 is an autosomal recessive disease caused by mutations in the autoimmune regulator (AIRE) gene, located on the short arm of chromosome 21. The functioning of following pathways can be altered in APS1, though the specific contribution in increasing susceptibility to candida infection is not well defined.

  1. Defects in AIRE gene are associated with autoantibodies to interleukin (IL) 17A, IL17F and IL22, which are key cytokines for the function of the T-helper (Th) 17 cell subset. Loss of function of these cytokines increase susceptibility to candida infections (177).
  2. Autoimmunity to mediators involved in antigen presentation by B cells may be an additional factor responsible for susceptibility. This is further corroborated by the response to rituximab (anti-CD 20 antibody that prevents B cell function) to certain components of the disease in individuals with AIRE deficiency (178).
  • A defect in Dectin-1, a β-glucan receptor, has been shown to diminish tumor necrosis factor α production in APS-1. Innate immune response is affected as a result (179).

 

CLINICAL SPECTRUM

 

CMC is the most common component of APS-1. It has been reported in 80-100% of cases in different series (121,177). Onset of CMC is usually in the first decade and cases can be seen in the very first year of life. Mouth, nails and, less frequently, skin, vagina and the esophagus are affected. The infection tends to be persistent or recurrent. Severity of the infection in variable, however disseminated disease is rare (177).

 

The oral mucosa is the usual site of infection. All spectra of infection starting from localized ulceration, and redness in mild cases to involvement of entire mouth is described. White or grey membrane covering the tongue or mucosa are visible in the hyperplastic form. Cracks (angular cheilitis or perlèche) occurring at the angle of the mouth is common. The atrophic form has areas of leukoplakia, which is a significant risk factor for carcinoma of the oral mucosa. The finger nails are the other site which is commonly affected. There can be an associated paronychia. Onychomycosis in CMC is particularly resistant to treatment (121,180).

 

TREATMENT

 

Oral fluconazole is the preferred therapy. Some patients require suppressive treatment with fluconazole 100 mg three times a week. Emergence of resistance remains a possibility with suppressive therapy. Alternatives for fluconazole refractory disease includes itraconazole, Posaconazole, or voriconazole. Rare cases of systemic disease not responding to azoles might require a lipid formulation of amphotericin B or echinocandins (144).

 

ADVERSE ENDOCRINE EFFECTS OF ANTIFUNGAL AGENTS

 

The antifungal drugs such as polyenes, azoles and echinocandins can impact the function of endocrine glands. Azoles are recognized for their adverse effect on adrenal cortex and the gonads. The other drugs are also known to cause endocrine dysfunction though less frequently. These important adverse endocrine consequences of the different antifungal agents are discussed below.

 

Azoles

 

The azoles are the one of the most frequently administered systemic antifungal agents. They can be divided into two groups on the basis of their structure. Ketoconazole, which belongs to the imidazole group, is associated with multiple endocrine adverse effect, but seldom used orally as an antifungal agent currently. The newer azoles belonging to the triazole group include fluconazole, itraconazole, voriconazole, posaconazole, and isavuconazole. Endocrine dysfunction also occurs with the triazoles but is less frequent (181).

 

ADRENAL GLAND

 

The azoles exert their antifungal effect by inhibiting the cytochrome P450 (CYP450) enzyme lanosterol 14-α-demethylase (CYP51) mediated conversion of lanosterol to ergosterol, a critical constituent of fungal cell wall.  Mammals do not have this enzyme, but azoles can block the synthesis of glucocorticoids, mineralocorticoids, and sex steroids by blocking CYP450 dependent enzymes involved in steroidogenesis (182).

 

Ketoconazole

 

Ketoconazole is a dose-dependent reversible inhibitor of cholesterol desmolase, 17,20-lyase, 11β-hydroxylase, 17α-hydroxylase, and 18-hydroxylase (183). Ketoconazole at doses of more than 200 mg daily can impair glucocorticoid synthesis. Overt adrenal insufficiency is relatively infrequent however it can be seen with doses of 600 to 1200 mg/day, which are often used in the medical management of Cushing’s syndrome (59,184,185). Apart from inhibiting enzymes involved in steroidogenesis, ketoconazole is also a dose-dependent, reversible, competitive antagonist at the glucocorticoid receptor level (186). The inhibitory effect of ketoconazole on adrenal steroid synthesis has been utilized for  the medical management of Cushing’s  syndrome (187).

 

Fluconazole and Posaconazole

 

Adrenal insufficiency has been reported with the imidazole derivatives itraconazole, fluconazole, voriconazole, and posaconazole (188–192). Primary adrenal insufficiency induced by fluconazole has been observed in critically ill patient as a result of CYP450 inhibition (193). Fluconazole has been employed for the medical management of Cushing’s syndrome (194). Posaconazole-induced primary adrenal insufficiency resulting from a similar mechanism has been described (190,192).

 

Itraconazole and Voriconazole

 

Itraconazole and voriconazole (also ketoconazole) are potent inhibitors of CYP3A4, the enzyme that partially metabolizes glucocorticoids. The resultant decrease in glucocorticoid clearance produces supraphysiological levels of glucocorticoid from inhaled, nasal or oral steroids (195). The clinical profile resembles that of an iatrogenic Cushing’s syndrome later progressing to secondary or central adrenal insufficiency consequent to suppression of the hypothalamic-pituitary-adrenal (HPA) axis (196). Secondary adrenal insufficiency following combined use of glucocorticoids and itraconazole or voriconazole have been described (188,191). Steroids that are predominantly metabolized by the CYP3A4 pathway include methylprednisolone, fluticasone, budesonide and triamcinolone. It may be prudent to consider alternative glucocorticoids such as prednisolone, beclomethasone, or flunisolide that are not predominantly metabolized by CYP3A4 enzymes when voriconazole or itraconazole is being administered (190,191).

 

Pseudohyperaldosteronism

 

Posaconazole and itraconazole has been associated with a syndrome of mineralocorticoid excess manifested by low-renin low-aldosterone hypertension and hypokalemia (197). Two distinct mechanisms are implicated in the pathogenesis with significant interindividual differences. Posaconazole can inhibit the enzyme 11 β-hydroxylase (CYP11B1) and prevent the conversion of 11-deoxycortisol to cortisol. Diminished cortisol synthesis triggers adrenal steroidogenesis as a result of loss of feedback inhibition of the HPA axis and causes accumulation of 11-deoxycortisol (and 11-deoxycorticosterone). Even though aldosterone production is reduced due to posaconazole-induced aldosterone synthase (CYP11B2) inhibition, very high levels of 11-deoxycortisol and 11-deoxycorticosterone can overcome that and produce a state of mineralocorticoid excess (197,198). The other mechanism incriminated is blockage of the peripheral cortisol metabolizing enzyme 11 β-hydroxysteroid dehydrogenase 2 (11β-HSD2) leading to an increased ratio of active to inactive cortisol metabolite. Elevated ratios of cortisol to corticosterone and their tetrahydro-metabolites are observed in such individuals (198). There are few case reports of itraconazole and several reports of posaconazole-induced pseudohyperladosteronism (199–202). Therapeutic options include lowering the dose of azoles or changing to alternatives like isavuconazole (198).

 

GONADS

 

Male Sexual Dysfunction

 

Inhibition of 17,20-lyase by ketoconazole impairs production of testosterone in the male gonads (203). The effect can be seen even at a single dose of 200mg, however lower testosterone levels and longer duration of suppression can be seen with an increasing dose (204). Oligospermia and azoospermia as well as decreased libido and impotence have been reported at doses more than 800mg/day (181). Reversible gynecomastia is another manifestation seen due to increase in the estradiol:testosterone ratio partially attributed to displacement of estrogen from sex-hormone binding globulin by the drug (205).

 

Ketoconazole also binds to androgen receptors thereby blocking androgen signaling (206). Antiandrogenic properties of ketoconazole have been used in the treatment of prostate cancer, autonomous Leydig cell hyperactivity in children with precocious puberty, and topical therapy for androgenetic alopecia (207–209).

 

Fluconazole in contrast to ketoconazole does not affect testosterone synthesis (210). A single case of posaconazole induced gynecomastia has been reported. Inhibition of the CYP11B1 enzyme by the drug stimulates compensatory adrenal steroidogenesis. Increased peripheral conversion of adrenal androgens to estrogen was presumed to induce gynecomastia after posaconazole use. The other possible hypothesis could be reduced catabolism of estrogen in the liver due to blocking of CYP3A4 and CYP3A7 (211).

 

Female Reproductive Dysfunction

 

Ketoconazole reduces estrogen levels in females. Reduction of estrogen levels could be due to aromatase inhibition or androgen synthesis blockade. Estrogen precursor deprivation from decreased androgen synthesis is likely to be the predominant mechanism (59). In animal studies, ovarian progesterone production is impaired thereby preventing uterine implantation (212). Ketoconazole has been used in treatment of polycystic ovarian syndrome and ovarian hyperthecosis, given its ability to substantially block ovarian androgen synthesis (213). Itraconazole when co-prescribed with simvastatin, induced metrorrhagia in a 69-year old lady, presumably occurring as result of low-estrogen breakthrough bleeding (214). Itraconazole can also enhance estrogen metabolism interfering with efficacy of oral contraceptives (215). Fluconazole on the other hand can increase estrogen levels by inhibiting its metabolism and is not associated with risk of contraceptive failure (216).

 

HYPONATREMIA

 

Voriconazole use has been associated with severe hyponatremia. The median time to onset of hyponatremia is 6-26 days (217). Severe hyponatremia, volume depletion, elevated antidiuretic hormone (ADH), and plasma renin activity along with high urinary sodium suggestive of salt-losing nephropathy were observed after voriconazole administration (218). Syndrome of inappropriate ADH secretion (SIADH) has been implicated as another possible mechanism and euvolemia is the critical distinguishing feature from salt-losing nephropathy (219). The toxic effect of voriconazole is concentration-dependent and therapeutic drug monitoring has been found to be useful for prevention and dose adjustment for hyponatremia (220). The risk of hyponatremia increased with trough concentrations > 7 mg/L and the dose should be modified to maintain levels below that threshold (181). An interesting observation was predisposition to develop voriconazole induced hyponatremia among Asians, in whom polymorphism of CYP2C19 is more common (221). CYP2C19 is the enzyme that metabolizes voriconazole and dosing depending on genotype has been proposed as a means to avert its adverse effects including hyponatremia (222,223).

 

FLUORIDE-INDUCED PERIOSTITIS

 

There are several reports of voriconazole-induced periostitis presumably related to excess fluoride released from the three fluorine atoms present in the molecule (224–228). A 400 mg tablet of voriconazole contains approximately 65 mg of fluoride, however only 5% of the fluoride is generated from the drug in free form (181,224). The other fluorinated azoles fluconazole and posaconazole contain two atoms of fluorides and have not been associated with fluorosis and periostitis (225).

 

A review summarizing 98 cases of periostitis, reported the median age to be 59 years with onset of symptoms between 6 weeks to 8 years after drug exposure. Presenting features are muscle and bone pain. Affection of almost any skeletal site has been described (229). Ribs and ulna are the most common site of involvement. The other involved sites include tibia, clavicle, femur, radius, fibula, scapula, and humerus (224,229).

 

The serum fluoride and alkaline phosphatase levels are significantly higher in those with periostitis compared to those without (224). The plain radiograph reveals multiple areas of periosteal thickening along with formation of new bones which may take the form of an exostoses or can be fluffy. The radiological findings are analogous to periostitis deformans observed in fluoride intoxication (230).  Bone scan shows increased tracer uptake but unlike hypertrophic osteoarthropathy tend to be asymmetric (224). Discontinuation of voriconazole usually results in improvement in the majority of cases. Substitution by a non-fluorinated azole such as itraconazole can be considered when continued antifungal coverage is necessary. Replacement by posaconazole has also been beneficial (228). 

 

OTHER ENDOCRINE ABNORMALITIES

 

High dose ketoconazole (1200mg/day) may rarely cause hypothyroidism by interference with iodine and thyroid peroxidase (231). Ketoconazole is also an inhibitor of 25(OH)D-1α hydroxylase (CYP27B1) leading to decreased 1,25(OH)2D levels (232). Hypercalcemia induced by sarcoidosis, tuberculosis and other granulomatous disorders respond to treatment with ketoconazole (233,234). Both ketoconazole and fluconazole are treatment options for idiopathic infantile hypercalciuria that occurs from CYP24A1 (24-hydroxylase) gene mutations (235,236). The effects of ketoconazole on enzymes regulating vitamin D has also been explored for treatment of prostate cancer (208,237).   

 

There are rare reports of pancreatitis with fluconazole, itraconazole, and voriconazole (181). Voriconazole, ketoconazole, and fluconazole have been implicated as a cause of hypoglycemia (238,239). The hypoglycemia could be due to hyperinsulinemia resulting from decreased degradation of insulin (240). The metabolism of sulfonylureas can be inhibited by fluconazole thereby increasing the risk of hypoglycemia in individuals receiving both these drugs (241,242).

 

Polyenes

 

The polyenes currently in medical use are nystatin and amphotericin B. Use of nystatin is limited to topical application. Amphotericin B deoxycholate is associated with higher risk of toxicity as compared to its lipid preparation. The lipid formulations of amphotericin B are expensive but the risk of adverse effect is less. Electrolyte abnormalities resulting from tubular damage is the predominant endocrine dysfunction described with amphotericin B. Rare cases of pancreatitis have occurred with liposomal amphotericin B (243).  

 

TUBULAR DAMAGE

 

Clinical manifestations of amphotericin B induced nephrotoxicity include renal insufficiency, hypokalemia, hypomagnesemia, metabolic acidosis resulting from distal renal tubular acidosis, and polyuria due to nephrogenic diabetes insipidus (DI) (244–246). The mechanism for DI involves a decrease in aquaporin 2 expression in the kidney medulla, that makes the collecting tubules insensitive to ADH (244). Although the risk of nephrogenic DI with lipid preparations of  amphotericin B is significantly less, cases have still been described (247). Nephrogenic DI can be managed by amiloride plus hydrochlorothiazide, or indomethacin (248).

 

Nephrogenic DI can also be induced by hypokalemia caused by amphotericin B (249). Hypokalemia is more common with amphotericin B deoxycholate but is also recognized  with lipid preparations of amphotericin B (250). Amphotericin B can induce apoptosis of renal tubular cells and also enhance tubular permeability by damage to lining epithelium (251). Renal magnesium loss can also result from amphotericin B. PTH secretion is  affected by hypomagnesemia and that may subsequently lead to hypocalcemia (252). Monitoring and supplementing potassium and magnesium is an important adjunct to prevent adverse consequences of amphotericin B therapy (253).

 

Echinocandins

 

Capsofungin, micofungin and antidulafungin are the three echinocandins currently in clinical use.  These agents, unlike azoles or amphotericin B, do not usually cause adverse endocrine effects. Micafungin is rarely reported to cause pancreatitis (254). Caspofungin has been reported to induce hypercalcemia in an infant by an undefined mechanism (255).

 

Other Agents

 

Oral potassium iodide is used in treatment of cutaneous sporotrichosis (256). It may precipitate thyrotoxicosis in patients with incipient Graves’ disease or multinodular goiter in areas of relative iodine deficiency (Jod-Basedow disease).  Hypothyroidism can occur in those with excessive autoregulation on prolonged exposure (Wolff-Chaikoff effect) (257).

 

CONCLUSION

 

Although fungi are ubiquitous within the environment, very few are considered true pathogens and affect healthy individuals only in limited circumstances. The majority of fungi are opportunistic and immune dysfunction in endocrine disorders increase susceptibility to fungal infection. On the other hand, fungal diseases especially in immunocompromised host can disseminate and affect various endocrine glands thereby impairing their function. Antifungal therapies too contribute to endocrine adverse effects. Moreover, in few endocrinological conditions like Cushing’s syndrome, signs and symptoms of fungal infection can be masked due to effect of hypercortisolemia. A high index of suspicion is mandated in such cases, as delayed or missed diagnosis could dramatically influence the outcome. An understanding of the complex relationship between fungal infection and endocrine disorders is necessary in modern-day medicine as both these conditions are increasingly prevalent.

 

REFERENCES

 

  1. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010 Mar 4;464(7285):59–65.
  2. Havlickova B, Czaika VA, Friedrich M. Epidemiological trends in skin mycoses worldwide. Mycoses. 2008 Sep;51 Suppl 4:2–15.
  3. Bitar D, Lortholary O, Le Strat Y, Nicolau J, Coignard B, Tattevin P, et al. Population-based analysis of invasive fungal infections, France, 2001-2010. Emerg Infect Dis. 2014 Jul;20(7):1149–55.
  4. Pekic S, Miljic D, Popovic V. Infections of the Hypothalamic-Pituitary Region. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Mar 12]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK532083/
  5. Catarino D, Ribeiro C, Gomes L, Paiva I. Corticotroph adenoma and pituitary fungal infection: a rare association. Endocrinol Diabetes Metab Case Rep. 2020 Mar 25;2020.
  6. Al-Mendalawi MD. Pituitary aspergillosis: A report and review of the literature. Neurol India. 2018 Oct;66(5):1525.
  7. Iplikcioglu AC, Bek S, Bikmaz K, Ceylan D, Gökduman CA. Aspergillus pituitary abscess. Acta Neurochir (Wien). 2004 May;146(5):521–4.
  8. Moore LA, Erstine EM, Prayson RA. Pituitary aspergillus infection. J Clin Neurosci Off J Neurosurg Soc Australas. 2016 Jul;29:178–80.
  9. Strickland BA, Pham M, Bakhsheshian J, Carmichael J, Weiss M, Zada G. Endoscopic Endonasal Transsphenoidal Drainage of a Spontaneous Candida glabrata Pituitary Abscess. World Neurosurg. 2018 Jan;109:467–70.
  10. Heary RF, Maniker AH, Wolansky LJ. Candidal pituitary abscess: case report. Neurosurgery. 1995 May;36(5):1009–12; discussion 1012-1013.
  11. Sano T, Kovacs K, Scheithauer BW, Rosenblum MK, Petito CK, Greco CM. Pituitary pathology in acquired immunodeficiency syndrome. Arch Pathol Lab Med. 1989 Sep;113(9):1066–70.
  12. Bartlett JA, Hulette C. Central nervous system pneumocystosis in a patient with AIDS. Clin Infect Dis Off Publ Infect Dis Soc Am. 1997 Jul;25(1):82–5.
  13. Scanarini M, Rotilio A, Rigobello L, Pomes A, Parenti A, Alessio L. Primary intrasellar coccidioidomycosis simulating a pituitary adenoma. Neurosurgery. 1991 May;28(5):748–51.
  14. Vijayvargiya P, Javed I, Moreno J, Mynt MA, Kotapka M, Zaki R, et al. Pituitary aspergillosis in a kidney transplant recipient and review of the literature. Transpl Infect Dis Off J Transplant Soc. 2013 Oct;15(5):E196-200.
  15. Choi E, Kim SB, Kim JH, Yoon YK. Lung aspergilloma with pituitary invasive aspergillosis presenting as headache and hyponatraemia. BMJ Case Rep. 2021 Jan 27;14(1).
  16. Saffarian A, Derakhshan N, Taghipour M, Eghbal K, Roshanfarzad M, Dehghanian A. Sphenoid Aspergilloma with Headache and Acute Vision Loss. World Neurosurg. 2018 Jul;115:159–61.
  17. Stalldecker G, Molina HA, Antelo N, Arakaki T, Sica RE, Basso A. [Hypopituitarism caused by colonic carcinoma metastasis associated with hypophysial aspergillosis]. Medicina (Mex). 1994;54(3):248–52.
  18. Ouyang T, Zhang N, Wang L, Jiao J, Zhao Y, Li Z, et al. Primary Aspergillus sellar abscess simulating pituitary tumor in immunocompetent patient. J Craniofac Surg. 2015 Mar;26(2):e86-88.
  19. Hong W, Liu Y, Chen M, Lin K, Liao Z, Huang S. Secondary headache due to aspergillus sellar abscess simulating a pituitary neoplasm: case report and review of literature. SpringerPlus. 2015;4:550.
  20. Fontana C, Gaziano R, Favaro M, Casalinuovo I, Pistoia E, Di Francesco P. (1-3)-β-D-Glucan vs Galactomannan Antigen in Diagnosing Invasive Fungal Infections (IFIs). Open Microbiol J. 2012 Aug 24;6:70–3.
  21. Patterson TF, Thompson GR, Denning DW, Fishman JA, Hadley S, Herbrecht R, et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis Off Publ Infect Dis Soc Am. 2016 Aug 15;63(4):e1–60.
  22. Miceli MH. Central Nervous System Infections Due to Aspergillus and Other Hyaline Molds. J Fungi [Internet]. 2019 Aug 30 [cited 2021 Mar 21];5(3). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6787746/
  23. Shrestha RT, Hennessey J. Acute and Subacute, and Riedel’s Thyroiditis. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Mar 21]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK285553/
  24. Tan J, Shen J, Fang Y, Zhu L, Liu Y, Gong Y, et al. A suppurative thyroiditis and perineal subcutaneous abscess related with aspergillus fumigatus: a case report and literature review. BMC Infect Dis. 2018 Dec 27;18(1):702.
  25. Zavascki AP, Maia AL, Goldani LZ. Pneumocystis jiroveci thyroiditis: report of 15 cases in the literature. Mycoses. 2007 Nov;50(6):443–6.
  26. Mori T, Matsumura M, Yamada K, Irie S, Oshimi K, Suda K, et al. Systemic aspergillosis caused by an aflatoxin-producing strain of Aspergillus flavus. Med Mycol. 1998 Apr;36(2):107–12.
  27. Cicora F, Mos F, Paz M, Roberti J. Successful treatment of acute thyroiditis due to Aspergillus spp. in the context of disseminated invasive aspergillosis in a kidney transplant patient. Nefrol Publicacion Of Soc Espanola Nefrol. 2013;33(4):618–9.
  28. Solak Y, Atalay H, Nar A, Ozbek O, Turkmen K, Erekul S, et al. Aspergillus thyroiditis in a renal transplant recipient mimicking subacute thyroiditis. Transpl Infect Dis Off J Transplant Soc. 2011 Apr;13(2):178–81.
  29. Badawy SM, Becktell KD, Muller WJ, Schneiderman J. Aspergillus thyroiditis: first antemortem case diagnosed by fine-needle aspiration culture in a pediatric stem cell transplant patient. Transpl Infect Dis Off J Transplant Soc. 2015 Dec;17(6):868–71.
  30. Phulware RH, Gupta B, Sahoo B, Agarwal S, Mathur S. Aspergillus thyroiditis: In an immunocompromised young adult. Diagn Cytopathol. 2019 Apr;47(4):362–4.
  31. Santiago M, Martinez JH, Palermo C, Figueroa C, Torres O, Trinidad R, et al. Rapidly growing thyroid mass in an immunocompromised young male adult. Case Rep Endocrinol. 2013;2013:290843.
  32. Kishi Y, Negishi M, Kami M, Hamaki T, Miyakoshi S, Ueyama J, et al. Fatal airway obstruction caused by invasive aspergillosis of the thyroid gland. Leuk Lymphoma. 2002 Mar;43(3):669–71.
  33. Marui S, de Lima Pereira AC, de Araújo Maia RM, Borba EF. Suppurative thyroiditis due to aspergillosis: a case report. J Med Case Reports. 2014 Nov 21;8:379.
  34. Nicolè S, Lanzafame M, Cazzadori A, Vincenzi M, Mangani F, Colato C, et al. Successful Antifungal Combination Therapy and Surgical Approach for Aspergillus fumigatus Suppurative Thyroiditis Associated with Thyrotoxicosis and Review of Published Reports. Mycopathologia. 2017 Oct;182(9–10):839–45.
  35. Erdem H, Uzunlar AK, Yildirim U, Yildirim M, Geyik MF. Diffuse infiltration of Aspergillus hyphae in the thyroid gland with multinodular goiter. Indian J Pathol Microbiol. 2011 Dec;54(4):814–6.
  36. Rahimi SA. Disseminated Pneumocystis carinii in thymic alymphoplasia. Arch Pathol. 1974 Mar;97(3):162–5.
  37. Niles D, Boguniewicz J, Shakeel O, Margolin J, Chelius D, Gupta M, et al. Candida tropicalis Thyroiditis Presenting With Thyroid Storm in a Pediatric Patient With Acute Lymphocytic Leukemia. Pediatr Infect Dis J. 2019 Oct;38(10):1051–3.
  38. Fernandez JF, Anaissie EJ, Vassilopoulou-Sellin R, Samaan NA. Acute fungal thyroiditis in a patient with acute myelogenous leukaemia. J Intern Med. 1991 Dec;230(6):539–41.
  39. Berry CZ, Goldberg LC, Shepard WL. Systemic lupus erythematosus complicated by coccidioidomycosis. JAMA. 1968 Oct 28;206(5):1083–5.
  40. Smilack JD, Argueta R. Coccidioidal infection of the thyroid. Arch Intern Med. 1998 Jan 12;158(1):89–92.
  41. Goldani LZ, Klock C, Diehl A, Monteiro AC, Maia AL. Histoplasmosis of the thyroid. J Clin Microbiol. 2000 Oct;38(10):3890–1.
  42. Goldani LZ, Zavascki AP, Maia AL. Fungal thyroiditis: an overview. Mycopathologia. 2006 Mar;161(3):129–39.
  43. Nguyen J, Manera R, Minutti C. Aspergillus thyroiditis: a review of the literature to highlight clinical challenges. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2012 Dec;31(12):3259–64.
  44. Lee D-G, Lee H-J, Yan JL, Lin SS-F, Aram JA. Efficacy and safety of combination antifungal therapy in Korean haematological patients with invasive aspergillosis. Mycoses. 2019 Oct;62(10):969–78.
  45. Marr KA, Schlamm HT, Herbrecht R, Rottinghaus ST, Bow EJ, Cornely OA, et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med. 2015 Jan 20;162(2):81–9.
  46. Goltzman D. Approach to Hypercalcemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 May 18]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279129/
  47. Gurram PR, Castillo NE, Esquer Garrigos Z, Vijayvargiya P, Abu Saleh OM. A Dimorphic Diagnosis of a Pleomorphic Disease: An Unusual Cause of Hypercalcemia. Am J Med. 2020 Nov;133(11):e659–62.
  48. Spindel SJ, Hamill RJ, Georghiou PR, Lacke CE, Green LK, Mallette LE. Case report: vitamin D-mediated hypercalcemia in fungal infections. Am J Med Sci. 1995 Aug;310(2):71–6.
  49. Ahmed B, Jaspan JB. Case report: hypercalcemia in a patient with AIDS and Pneumocystis carinii pneumonia. Am J Med Sci. 1993 Nov;306(5):313–6.
  50. Huang J-C, Kuo M-C, Hwang S-J, Hwang D-Y, Chen H-C. Vitamin D-mediated hypercalcemia as the initial manifestation of pulmonary cryptococcosis in an HIV-uninfected patient. Intern Med Tokyo Jpn. 2012;51(13):1793–6.
  51. Hamroun A, Lenain R, Bui Nguyen L, Chamley P, Loridant S, Neugebauer Y, et al. Hypercalcemia is common during Pneumocystis pneumonia in kidney transplant recipients. Sci Rep. 2019 Aug 29;9(1):12508.
  52. Giordani MC, Villamil Cortez SK, Diehl M, Barcan LA, Rosa-Diez G, Groppa SR, et al. Hypercalcemia as an Early Finding of Opportunistic Fungal Pneumonia in Renal Transplantation: A Case Series Report. Transplant Proc. 2020 May;52(4):1178–82.
  53. Walker JV, Baran D, Yakub N, Freeman RB. Histoplasmosis with hypercalcemia, renal failure, and papillary necrosis. Confusion with sarcoidosis. JAMA. 1977 Mar 28;237(13):1350–2.
  54. Murray JJ, Heim CR. Hypercalcemia in disseminated histoplasmosis. Aggravation by vitamin D. Am J Med. 1985 May;78(5):881–4.
  55. Chalhoub E, Elhomsy G, Brake M. Hypercalcemia in histoplasmosis aggravated with antifungal treatment. J Med Liban. 2012 Sep;60(3):165–8.
  56. Lawn SD, Macallan DC. Hypercalcemia: a manifestation of immune reconstitution complicating tuberculosis in an HIV-infected person. Clin Infect Dis Off Publ Infect Dis Soc Am. 2004 Jan 1;38(1):154–5.
  57. Caldwell JW, Arsura EL, Kilgore WB, Reddy CM, Johnson RH. Hypercalcemia in patients with disseminated coccidioidomycosis. Am J Med Sci. 2004 Jan;327(1):15–8.
  58. Fierer J, Burton DW, Haghighi P, Deftos LJ. Hypercalcemia in disseminated coccidioidomycosis: expression of parathyroid hormone-related peptide is characteristic of granulomatous inflammation. Clin Infect Dis Off Publ Infect Dis Soc Am. 2012 Oct;55(7):e61-66.
  59. Lionakis MS, Samonis G, Kontoyiannis DP. Endocrine and Metabolic Manifestations of Invasive Fungal Infections and Systemic Antifungal Treatment. Mayo Clin Proc. 2008 Sep;83(9):1046–60.
  60. Reinholt FP, Hultenby K, Oldberg A, Heinegård D. Osteopontin--a possible anchor of osteoclasts to bone. Proc Natl Acad Sci U S A. 1990 Jun;87(12):4473–5.
  61. Siddiqi A, Goh BT, Brown CL, Hillman RJ, Monson JP. Hypothyroidism and hypoparathyroidism associated with Pneumocystis carinii infection in a patient with AIDS. Int J STD AIDS. 1998 Feb;9(2):108–10.
  62. Wheat LJ, Azar MM, Bahr NC, Spec A, Relich RF, Hage C. Histoplasmosis. Infect Dis Clin North Am. 2016 Mar;30(1):207–27.
  63. Kauffman CA. Endemic mycoses: blastomycosis, histoplasmosis, and sporotrichosis. Infect Dis Clin North Am. 2006 Sep;20(3):645–62, vii.
  64. Sharma B, Nehara HR, Bhavi VK, Maan P, Saran S. Adrenal histoplasmosis in immunocompetent individuals a case series from the North-Western part of India, Rajasthan province: An emerging endemic focus. Indian J Med Microbiol. 2020 Dec;38(3 & 4):485–8.
  65. Pereira GH, Lanzoni VPB, Beirão EM, Timerman A, Melhem M de SC. DISSEMINATED FUNGAL INFECTION WITH ADRENAL INVOLVEMENT: REPORT OF TWO HIV NEGATIVE BRAZILIAN PATIENTS. Rev Inst Med Trop Sao Paulo. 2015 Dec;57(6):527–30.
  66. Alexandraki KI, Grossman A. Adrenal Insufficiency. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 May 23]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279122/
  67. Paolo WF, Nosanchuk JD. Adrenal infections. Int J Infect Dis. 2006 Sep;10(5):343–53.
  68. Frenkel JK. Role of corticosteroids as predisposing factors in fungal diseases. Lab Investig J Tech Methods Pathol. 1962 Nov;11:1192–208.
  69. Roubsanthisuk W, Sriussadaporn S, Vawesorn N, Parichatikanond P, Phoojaroenchanachai M, Homsanit M, et al. Primary adrenal insufficiency caused by disseminated histoplasmosis: report of two cases. Endocr Pract Off J Am Coll Endocrinol Am Assoc Clin Endocrinol. 2002 Jun;8(3):237–41.
  70. Leal AMO, Magalhães PKR, Martinez R, Moreira AC. Adrenocortical hormones and interleukin patterns in paracoccidioidomycosis. J Infect Dis. 2003 Jan 1;187(1):124–7.
  71. Del Negro G, Melo EH, Rodbard D, Melo MR, Layton J, Wachslicht-Rodbard H. Limited adrenal reserve in paracoccidioidomycosis: cortisol and aldosterone responses to 1-24 ACTH. Clin Endocrinol (Oxf). 1980 Dec;13(6):553–9.
  72. Cherri J, Freitas MA, Llorach-Velludo MA, Piccinato CE. Paracoccidioidomycotic aortitis with embolization to the lower limbs. Report of a case and review of the literature. J Cardiovasc Surg (Torino). 1998 Oct;39(5):573–6.
  73. Disseminated Histoplasmosis and Rare Adrenal Involvement: Evidence of Absence or Absence of Evidence. Front Cell Infect Microbiol [Internet]. 2021 Mar 15 [cited 2021 May 23];11. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8005706/
  74. Goodwin RA, Shapiro JL, Thurman GH, Thurman SS, Des Prez RM. Disseminated histoplasmosis: clinical and pathologic correlations. Medicine (Baltimore). 1980 Jan;59(1):1–33.
  75. Gupta A, Ghosh A, Singh G, Xess I. A Twenty-First-Century Perspective of Disseminated Histoplasmosis in India: Literature Review and Retrospective Analysis of Published and Unpublished Cases at a Tertiary Care Hospital in North India. Mycopathologia. 2017 Dec;182(11–12):1077–93.
  76. Sifuentes-Osornio J, Corzo-León DE, Ponce-de-León LA. Epidemiology of Invasive Fungal Infections in Latin America. Curr Fungal Infect Rep. 2012 Mar;6(1):23–34.
  77. Carvalho FP de F de, Curiati JAE, Mauad T, Incerti MM, Jacob Filho W. Bilateral adrenal [corrected] nodules due to histoplasmosis in an elderly. Braz J Infect Dis Off Publ Braz Soc Infect Dis. 2007 Feb;11(1):160–2.
  78. Peçanha PM, Batista Ferreira ME, Massaroni Peçanha MA, Schmidt EB, Lamas de Araújo M, Zanotti RL, et al. Paracoccidioidomycosis: Epidemiological and Clinical Aspects in 546 Cases Studied in the State of Espírito Santo, Brazil. Am J Trop Med Hyg. 2017 Sep;97(3):836–44.
  79. Bocca AL, Amaral AC, Teixeira MM, Sato PK, Sato P, Shikanai-Yasuda MA, et al. Paracoccidioidomycosis: eco-epidemiology, taxonomy and clinical and therapeutic issues. Future Microbiol. 2013 Sep;8(9):1177–91.
  80. Sharma N, Ahlawat RS, Singh H, Sharma C, Anuradha S. Pneumocystis jirovecii infection of bilateral adrenal glands in an immunocompetent adult: a case report. J R Coll Physicians Edinb. 2019 Sep;49(3):222–4.
  81. Kumar A, Sreehari S, Velayudhan K, Biswas L, Babu R, Ahmed S, et al. Autochthonous blastomycosis of the adrenal: first case report from Asia. Am J Trop Med Hyg. 2014 Apr;90(4):735–9.
  82. Ito M, Hinata T, Tamura K, Koga A, Ito T, Fujii H, et al. Disseminated Cryptococcosis with Adrenal Insufficiency and Meningitis in an Immunocompetent Individual. Intern Med Tokyo Jpn. 2017;56(10):1259–64.
  83. Koene RJ, Catanese J, Sarosi GA. Adrenal hypofunction from histoplasmosis: a literature review from 1971 to 2012. Infection. 2013 Aug;41(4):757–9.
  84. Porntharukchareon T, Khahakaew S, Sriprasart T, Paitoonpong L, Snabboon T. Bilateral Adrenal Histoplasmosis. Balk Med J. 2019 Dec;36(6):359–600.
  85. Ahuja A, Mathur SR, Iyer VK, Sharma SK, Kumar N, Agarwal S. Histoplasmosis presenting as bilateral adrenal masses: cytomorphological diagnosis of three cases. Diagn Cytopathol. 2012 Aug;40(8):729–31.
  86. de Morais RQ, Salomon MFB, Corbiceiro WCH, de Melo ASA, Corrêa DG. Imaging contribution for the diagnosis of disseminated paracoccidioidomycosis. Int J Infect Dis IJID Off Publ Int Soc Infect Dis. 2020 Dec;101:206–9.
  87. Gupta P, Bhalla A, Sharma R. Bilateral adrenal lesions. J Med Imaging Radiat Oncol. 2012 Dec;56(6):636–45.
  88. Dhamija E, Panda A, Das CJ, Gupta AK. Adrenal imaging (Part 2): Medullary and secondary adrenal lesions. Indian J Endocrinol Metab. 2015;19(1):16–24.
  89. Gaspar GG, Cocio TA, Guioti-Puga F, Nascimento E, Fabro AT, Kress MR von Z, et al. Paracoccidioidomycosis due to Paracoccidioides lutzii complicated with adrenal injury and pulmonary arterial hypertension. Rev Inst Med Trop São Paulo [Internet]. [cited 2021 May 24];62. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7669275/
  90. Shanmugam N, Isenmann R, Barkin JS, Beger HG. Pancreatic fungal infection. Pancreas. 2003 Aug;27(2):133–8.
  91. Karam MB, Mosadegh L. Extra-pulmonary Pneumocystis jiroveci infection: a case report. Braz J Infect Dis Off Publ Braz Soc Infect Dis. 2014 Dec;18(6):681–5.
  92. Guice KS, Lynch M, Weatherbee L. Invasive aspergillosis: an unusual cause of hemorrhagic pancreatitis. Am J Gastroenterol. 1987 Jun;82(6):563–5.
  93. Cappell MS, Hassan T. Pancreatic disease in AIDS--a review. J Clin Gastroenterol. 1993 Oct;17(3):254–63.
  94. Trikudanathan G, Navaneethan U, Vege SS. Intra-abdominal fungal infections complicating acute pancreatitis: a review. Am J Gastroenterol. 2011 Jul;106(7):1188–92.
  95. Isenmann R, Schwarz M, Rau B, Trautmann M, Schober W, Beger HG. Characteristics of infection with Candida species in patients with necrotizing pancreatitis. World J Surg. 2002 Mar;26(3):372–6.
  96. Sahar N, Kozarek RA, Kanji ZS, Chihara S, Gan SI, Irani S, et al. The microbiology of infected pancreatic necrosis in the era of minimally invasive therapy. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2018 Jul;37(7):1353–9.
  97. Tange K, Yokota T, Sunago K, Aono M, Ochi H, Takechi S, et al. A rare case of acute pancreatitis caused by Candida Albicans. Clin J Gastroenterol. 2019 Feb;12(1):82–7.
  98. Chung RT, Schapiro RH, Warshaw AL. Intraluminal pancreatic candidiasis presenting as recurrent pancreatitis. Gastroenterology. 1993 May;104(5):1532–4.
  99. Giannopoulos A, Giamarellos-Bourboulis EJ, Adamakis I, Georgopoulou I, Petrikkos G, Katsilambros N. Epididymitis caused by Candida glabrata: a novel infection in diabetic patients? Diabetes Care. 2001 Nov;24(11):2003–4.
  100. Seo R, Oyasu R, Schaeffer A. Blastomycosis of the epididymis and prostate. Urology. 1997 Dec;50(6):980–2.
  101. Botero-García CA, Faccini-Martínez ÁA, Uribe E, Calixto O-J, Pérez-Díaz CE, Osejo-Diago PP, et al. Epididymo-orchitis caused by Histoplasma capsulatumin a Colombian patient. Rev Soc Bras Med Trop. 2017 Dec;50(6):868–70.
  102. Hood SV, Bell D, McVey R, Wilson G, Wilkins EG. Prostatitis and epididymo-orchitis due to Aspergillus fumigatus in a patient with AIDS. Clin Infect Dis Off Publ Infect Dis Soc Am. 1998 Jan;26(1):229–31.
  103. James CL, Lomax-Smith JD. Cryptococcal epididymo-orchitis complicating steroid therapy for relapsing polychondritis. Pathology (Phila). 1991 Jul;23(3):256–8.
  104. Jenkin GA, Choo M, Hosking P, Johnson PD. Candidal epididymo-orchitis: case report and review. Clin Infect Dis Off Publ Infect Dis Soc Am. 1998 Apr;26(4):942–5.
  105. Eickenberg H-Unull, Amin M, Lich R. Blastomycosis of the genitourinary tract. J Urol. 1975 May;113(5):650–2.
  106. Tichindelean C, East JW, Sarria JC. Disseminated histoplasmosis presenting as granulomatous epididymo-orchitis. Am J Med Sci. 2009 Sep;338(3):238–40.
  107. Fijak M, Pilatz A, Hedger MP, Nicolas N, Bhushan S, Michel V, et al. Infectious, inflammatory and ‘autoimmune’ male factor infertility: how do rodent models inform clinical practice? Hum Reprod Update. 2018 Jul;24(4):416–41.
  108. Nagy B, Sutka P. Demonstration of antibodies against Candida guilliermondii var. guilliermondii in asymptomatic infertile men. Mycoses. 1992 Oct;35(9–10):247–50.
  109. Lareau SM, Beigi RH. Pelvic inflammatory disease and tubo-ovarian abscess. Infect Dis Clin North Am. 2008 Dec;22(4):693–708.
  110. Curry A, Williams T, Penny ML. Pelvic Inflammatory Disease: Diagnosis, Management, and Prevention. Am Fam Physician. 2019 Sep 15;100(6):357–64.
  111. Saccente M, Woods GL. Clinical and laboratory update on blastomycosis. Clin Microbiol Rev. 2010 Apr;23(2):367–81.
  112. Chowfin A, Tight R. Female genital coccidioidomycosis (FGC), Addison’s disease and sigmoid loop abscess due to Coccidioides immites; case report and review of literature on FGC. Mycopathologia. 1999;145(3):121–6.
  113. Bæk O, Astvad K, Serizawa R, Wheat LJ, Brenøe PT, Hansen A-BE. Peritoneal and genital coccidioidomycosis in an otherwise healthy Danish female: a case report. BMC Infect Dis. 2017 Jan 31;17(1):105.
  114. To V, Gurberg J, Krishnamurthy S. Tubo-Ovarian Abscess Caused by Candida Albicans in an Obese Patient. J Obstet Gynaecol Can JOGC J Obstet Gynecol Can JOGC. 2015 May;37(5):426–9.
  115. Toy EC, Scerpella EG, Riggs JW. Tuboovarian abscess associated with Candida glabrata in a woman with an intrauterine device. A case report. J Reprod Med. 1995 Mar;40(3):223–5.
  116. Okmen F, Ekici H, Ari SA. Case Report of a Tubo-ovarian Abscess Caused by Candida kefyr. J Obstet Gynaecol Can JOGC J Obstet Gynecol Can JOGC. 2018 Nov;40(11):1466–7.
  117. Hsu W-C, Lee Y-H, Chang D-Y. Tuboovarian abscess caused by Candida in a woman with an intrauterine device. Gynecol Obstet Invest. 2007;64(1):14–6.
  118. Bylund DJ, Nanfro JJ, Marsh WL. Coccidioidomycosis of the female genital tract. Arch Pathol Lab Med. 1986 Mar;110(3):232–5.
  119. Tatay E, Meca G, Font G, Ruiz M-J. Interactive effects of zearalenone and its metabolites on cytotoxicity and metabolization in ovarian CHO-K1 cells. Toxicol Vitro Int J Publ Assoc BIBRA. 2014 Feb;28(1):95–103.
  120. Minervini F, Giannoccaro A, Cavallini A, Visconti A. Investigations on cellular proliferation induced by zearalenone and its derivatives in relation to the estrogenic parameters. Toxicol Lett. 2005 Dec 15;159(3):272–83.
  121. Sperling MA, Angelousi A, Yau M. Autoimmune Polyglandular Syndromes. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 May 30]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279152/
  122. Zhou K, Lansang MC. Diabetes Mellitus and Infections. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 May 31]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK569326/
  123. Higa M. [Clinical epidemiology of fungal infection in diabetes]. Nihon Rinsho Jpn J Clin Med. 2008 Dec;66(12):2239–44.
  124. Rodrigues CF, Rodrigues ME, Henriques M. Candida sp. Infections in Patients with Diabetes Mellitus. J Clin Med [Internet]. 2019 Jan 10 [cited 2021 May 31];8(1). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6352194/
  125. Tsang CSP, Chu FCS, Leung WK, Jin LJ, Samaranayake LP, Siu SC. Phospholipase, proteinase and haemolytic activities of Candida albicans isolated from oral cavities of patients with type 2 diabetes mellitus. J Med Microbiol. 2007 Oct;56(Pt 10):1393–8.
  126. Mba IE, Nweze EI. Mechanism of Candida pathogenesis: revisiting the vital drivers. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2020 Oct;39(10):1797–819.
  127. Rautemaa R, Ramage G. Oral candidosis--clinical challenges of a biofilm disease. Crit Rev Microbiol. 2011 Nov;37(4):328–36.
  128. Axéll T, Samaranayake LP, Reichart PA, Olsen I. A proposal for reclassification of oral candidosis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1997 Aug;84(2):111–2.
  129. Millsop JW, Fazel N. Oral candidiasis. Clin Dermatol. 2016 Aug;34(4):487–94.
  130. Unnikrishnan AG, Kalra S, Purandare V, Vasnawala H. Genital Infections with Sodium Glucose Cotransporter-2 Inhibitors: Occurrence and Management in Patients with Type 2 Diabetes Mellitus. Indian J Endocrinol Metab. 2018;22(6):837–42.
  131. Dovnik A, Golle A, Novak D, Arko D, Takač I. Treatment of vulvovaginal candidiasis: a review of the literature. Acta Dermatovenerol Alp Pannonica Adriat. 2015;24(1):5–7.
  132. Nyirjesy P, Sobel JD. Genital mycotic infections in patients with diabetes. Postgrad Med. 2013 May;125(3):33–46.
  133. Nyirjesy P, Sobel JD, Fung A, Mayer C, Capuano G, Ways K, et al. Genital mycotic infections with canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes mellitus: a pooled analysis of clinical studies. Curr Med Res Opin. 2014 Jun;30(6):1109–19.
  134. Kalra S, Chawla A. Diabetes and balanoposthitis. JPMA J Pak Med Assoc. 2016 Aug;66(8):1039–41.
  135. Weerasuriya N, Snape J. Oesophageal candidiasis in elderly patients: risk factors, prevention and management. Drugs Aging. 2008;25(2):119–30.
  136. Mohamed AA, Lu X-L, Mounmin FA. Diagnosis and Treatment of Esophageal Candidiasis: Current Updates. Can J Gastroenterol Hepatol. 2019;2019:3585136.
  137. Yismaw G, Asrat D, Woldeamanuel Y, Unakal C. Prevalence of candiduria in diabetic patients attending Gondar University Hospital, Gondar, Ethiopia. Iran J Kidney Dis. 2013 Mar;7(2):102–7.
  138. Alfouzan WA, Dhar R. Candiduria: Evidence-based approach to management, are we there yet? J Mycol Medicale. 2017 Sep;27(3):293–302.
  139. Kauffman CA. Diagnosis and management of fungal urinary tract infection. Infect Dis Clin North Am. 2014 Mar;28(1):61–74.
  140. Saunte DML, Piraccini BM, Sergeev AY, Prohić A, Sigurgeirsson B, Rodríguez-Cerdeira C, et al. A survey among dermatologists: diagnostics of superficial fungal infections - what is used and what is needed to initiate therapy and assess efficacy? J Eur Acad Dermatol Venereol JEADV. 2019 Feb;33(2):421–7.
  141. Gupta AK, Gregurek-Novak T, Konnikov N, Lynde CW, Hofstader S, Summerbell RC. Itraconazole and terbinafine treatment of some nondermatophyte molds causing onychomycosis of the toes and a review of the literature. J Cutan Med Surg. 2001 Jun;5(3):206–10.
  142. Pfaller MA, Jones RN, Doern GV, Fluit AC, Verhoef J, Sader HS, et al. International surveillance of blood stream infections due to Candida species in the European SENTRY Program: species distribution and antifungal susceptibility including the investigational triazole and echinocandin agents. SENTRY Participant Group (Europe). Diagn Microbiol Infect Dis. 1999 Sep;35(1):19–25.
  143. Borg-von Zepelin M, Kunz L, Rüchel R, Reichard U, Weig M, Gross U. Epidemiology and antifungal susceptibilities of Candida spp. to six antifungal agents: results from a surveillance study on fungaemia in Germany from July 2004 to August 2005. J Antimicrob Chemother. 2007 Aug;60(2):424–8.
  144. Pappas PG, Kauffman CA, Andes DR, Clancy CJ, Marr KA, Ostrosky-Zeichner L, et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis Off Publ Infect Dis Soc Am. 2016 Feb 15;62(4):e1-50.
  145. Cornely OA, Alastruey-Izquierdo A, Arenz D, Chen SCA, Dannaoui E, Hochhegger B, et al. Global guideline for the diagnosis and management of mucormycosis: an initiative of the European Confederation of Medical Mycology in cooperation with the Mycoses Study Group Education and Research Consortium. Lancet Infect Dis. 2019 Dec;19(12):e405–21.
  146. Ravani SA, Agrawal GA, Leuva PA, Modi PH, Amin KD. Rise of the phoenix: Mucormycosis in COVID-19 times. Indian J Ophthalmol. 2021 Jun;69(6):1563–8.
  147. Petrikkos G, Skiada A, Lortholary O, Roilides E, Walsh TJ, Kontoyiannis DP. Epidemiology and clinical manifestations of mucormycosis. Clin Infect Dis Off Publ Infect Dis Soc Am. 2012 Feb;54 Suppl 1:S23-34.
  148. Artis WM, Fountain JA, Delcher HK, Jones HE. A mechanism of susceptibility to mucormycosis in diabetic ketoacidosis: transferrin and iron availability. Diabetes. 1982 Dec;31(12):1109–14.
  149. Ibrahim AS, Kontoyiannis DP. Update on mucormycosis pathogenesis. Curr Opin Infect Dis. 2013 Dec;26(6):508–15.
  150. Morales-Franco B, Nava-Villalba M, Medina-Guerrero EO, Sánchez-Nuño YA, Davila-Villa P, Anaya-Ambriz EJ, et al. Host-Pathogen Molecular Factors Contribute to the Pathogenesis of Rhizopus spp. in Diabetes Mellitus. Curr Trop Med Rep. 2021 Jan 22;1–12.
  151. Yohai RA, Bullock JD, Aziz AA, Markert RJ. Survival factors in rhino-orbital-cerebral mucormycosis. Surv Ophthalmol. 1994 Aug;39(1):3–22.
  152. Skiada A, Lass-Floerl C, Klimko N, Ibrahim A, Roilides E, Petrikkos G. Challenges in the diagnosis and treatment of mucormycosis. Med Mycol. 2018 Apr 1;56(suppl_1):93–101.
  153. Lipner SR, Scher RK. Onychomycosis: Treatment and prevention of recurrence. J Am Acad Dermatol. 2019 Apr;80(4):853–67.
  154. Nenoff P, Krüger C, Ginter-Hanselmayer G, Tietz H-J. Mycology - an update. Part 1: Dermatomycoses: causative agents, epidemiology and pathogenesis. J Dtsch Dermatol Ges J Ger Soc Dermatol JDDG. 2014 Mar;12(3):188–209; quiz 210, 188–211; 212.
  155. Falodun O, Ogunbiyi A, Salako B, George AK. Skin changes in patients with chronic renal failure. Saudi J Kidney Dis Transplant Off Publ Saudi Cent Organ Transplant Saudi Arab. 2011 Mar;22(2):268–72.
  156. Westerberg DP, Voyack MJ. Onychomycosis: Current trends in diagnosis and treatment. Am Fam Physician. 2013 Dec 1;88(11):762–70.
  157. Ely JW, Rosenfeld S, Seabury Stone M. Diagnosis and management of tinea infections. Am Fam Physician. 2014 Nov 15;90(10):702–10.
  158. Hasenmajer V, Sbardella E, Sciarra F, Minnetti M, Isidori AM, Venneri MA. The Immune System in Cushing’s Syndrome. Trends Endocrinol Metab TEM. 2020 Sep;31(9):655–69.
  159. Lionakis MS, Kontoyiannis DP. Glucocorticoids and invasive fungal infections. Lancet Lond Engl. 2003 Nov 29;362(9398):1828–38.
  160. Lee EK, Kim JH, Yu HG. Candida albicans endophthalmitis in a patient with a non-functioning pituitary adenoma evolving into Cushing׳s disease: A case report. Med Mycol Case Rep. 2014 Oct;6:37–41.
  161. Kansagara DL, Tetrault J, Hamill C, Moore C, Olson B. Fatal factitious Cushing’s syndrome and invasive aspergillosis: case report and review of literature. Endocr Pract Off J Am Coll Endocrinol Am Assoc Clin Endocrinol. 2006 Dec;12(6):651–5.
  162. Joubert M, Reznik Y, Verdon R. “Rescue” bilateral adrenalectomy in paraneoplastic Cushing’s syndrome with invasive Aspergillus fumigatus infection. Am J Med Sci. 2007 Dec;334(6):497–8.
  163. Ng TT, Robson GD, Denning DW. Hydrocortisone-enhanced growth of Aspergillus spp.: implications for pathogenesis. Microbiol Read Engl. 1994 Sep;140 ( Pt 9):2475–9.
  164. Arlt A, Harbeck B, Anlauf M, Alkatout I, Klöppel G, Fölsch UR, et al. Fatal pneumocystis jirovecii pneumonia in a case of ectopic Cushing’s syndrome due to neuroendocrine carcinoma of the kidney. Exp Clin Endocrinol Diabetes Off J Ger Soc Endocrinol Ger Diabetes Assoc. 2008 Oct;116(9):515–9.
  165. Oosterhuis JK, van den Berg G, Monteban-Kooistra WE, Ligtenberg JJM, Tulleken JE, Meertens JHJM, et al. Life-threatening Pneumocystis jiroveci pneumonia following treatment of severe Cushing’s syndrome. Neth J Med. 2007 Jun;65(6):215–7.
  166. Gabalec F, Zavrelová A, Havel E, Cerman J, Radocha J, Svilias I, et al. Pneumocystis pneumonia during medicamentous treatment of Cushing’s syndrome--a description of two cases. Acta Medica (Hradec Kralove). 2011;54(3):127–30.
  167. van Halem K, Vrolijk L, Pereira AM, de Boer MGJ. Characteristics and Mortality of Pneumocystis Pneumonia in Patients With Cushing’s Syndrome: A Plea for Timely Initiation of Chemoprophylaxis. Open Forum Infect Dis [Internet]. 2017 Jan 30 [cited 2021 Jun 7];4(1). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5414011/
  168. Lu L, Zhao YY, Yang HB, Tian XL, Xu ZJ, Lu ZL. Cushing’s disease with pulmonary Cryptococcus neoformans infection in a single center in Beijing, China: A retrospective study and literature review. J Formos Med Assoc Taiwan Yi Zhi. 2019 Jan;118(1 Pt 2):285–90.
  169. Fujikawa H, Araki M. Disseminated cryptococcosis in a patient with adrenocortical carcinoma and Cushing’s syndrome. J Infect Chemother Off J Jpn Soc Chemother. 2020 Dec;26(12):1301–4.
  170. Lacativa PGS, Donangelo I, Wagman MB, Sieiro Neto L, Caldas CR, Violante AHD, et al. [Pseudotumoral pulmonary cryptococcosis in association with Cushing’s syndrome]. Arq Bras Endocrinol Metabol. 2004 Apr;48(2):318–23.
  171. Thangakunam B, Christopher DJ, Kurian S, Thomas R, James P. ENDOGENOUS EXCESS CORTISOL PRODUCTION AND DIABETES MELLITUS AS PREDISPOSING FACTORS FOR PULMONARY CRYPTOCOCCOSIS: A CASE REPORT AND LITERATURE REVIEW. Lung India Off Organ Indian Chest Soc. 2008;25(4):155–7.
  172. Gross NT, Chinchilla M, Camner P, Jarstrand C. Anticryptococcal activity by alveolar macrophages from rats treated with cortisone acetate during different periods of time. Mycopathologia. 1996;136(1):1–8.
  173. Nidhi A, Meena A, Sreekumar A, Daga MK. Corticosteroid-induced cryptococcal meningitis in patient without HIV. BMJ Case Rep. 2017 Jan 4;2017.
  174. Kosseifi SG, Nassour DN, Shaikh MA, Sarubbi FA, Jordan RM, Peiris AN. Nodular pulmonary histoplasmosis in Cushing’s disease: a case report and literature review. Tenn Med J Tenn Med Assoc. 2007 Dec;100(12):44–6.
  175. Dismukes WE, Royal SA, Tynes BS. Disseminated histoplasmosis in corticosteroid-treated patients. Report of five cases. JAMA. 1978 Sep 29;240(14):1495–8.
  176. Jung EJ, Park DW, Choi J-W, Choi WS. Chronic Cavitary Pulmonary Histoplasmosis in a Non-HIV and Immunocompromised Patient without Overseas Travel History. Yonsei Med J. 2015 May 1;56(3):871–4.
  177. Humbert L, Cornu M, Proust-Lemoine E, Bayry J, Wemeau J-L, Vantyghem M-C, et al. Chronic Mucocutaneous Candidiasis in Autoimmune Polyendocrine Syndrome Type 1. Front Immunol. 2018;9:2570.
  178. Gavanescu I, Benoist C, Mathis D. B cells are required for Aire-deficient mice to develop multi-organ autoinflammation: A therapeutic approach for APECED patients. Proc Natl Acad Sci U S A. 2008 Sep 2;105(35):13009–14.
  179. Pedroza LA, Kumar V, Sanborn KB, Mace EM, Niinikoski H, Nadeau K, et al. Autoimmune regulator (AIRE) contributes to Dectin-1-induced TNF-α production and complexes with caspase recruitment domain-containing protein 9 (CARD9), spleen tyrosine kinase (Syk), and Dectin-1. J Allergy Clin Immunol. 2012 Feb;129(2):464–72, 472.e1-3.
  180. Eyerich K, Eyerich S, Hiller J, Behrendt H, Traidl-Hoffmann C. Chronic mucocutaneous candidiasis, from bench to bedside. Eur J Dermatol EJD. 2010 Jun;20(3):260–5.
  181. Benitez LL, Carver PL. Adverse Effects Associated with Long-Term Administration of Azole Antifungal Agents. Drugs. 2019 Jun;79(8):833–53.
  182. Loose DS, Kan PB, Hirst MA, Marcus RA, Feldman D. Ketoconazole blocks adrenal steroidogenesis by inhibiting cytochrome P450-dependent enzymes. J Clin Invest. 1983 May;71(5):1495–9.
  183. Sonino N. The use of ketoconazole as an inhibitor of steroid production. N Engl J Med. 1987 Sep 24;317(13):812–8.
  184. Best TR, Jenkins JK, Murphy FY, Nicks SA, Bussell KL, Vesely DL. Persistent adrenal insufficiency secondary to low-dose ketoconazole therapy. Am J Med. 1987 Mar 23;82(3 Spec No):676–80.
  185. Khosla S, Wolfson JS, Demerjian Z, Godine JE. Adrenal crisis in the setting of high-dose ketoconazole therapy. Arch Intern Med. 1989 Apr;149(4):802–4.
  186. Duret C, Daujat-Chavanieu M, Pascussi J-M, Pichard-Garcia L, Balaguer P, Fabre J-M, et al. Ketoconazole and miconazole are antagonists of the human glucocorticoid receptor: consequences on the expression and function of the constitutive androstane receptor and the pregnane X receptor. Mol Pharmacol. 2006 Jul;70(1):329–39.
  187. Pivonello R, De Leo M, Cozzolino A, Colao A. The Treatment of Cushing’s Disease. Endocr Rev. 2015 Aug;36(4):385–486.
  188. Skov M, Main KM, Sillesen IB, Müller J, Koch C, Lanng S. Iatrogenic adrenal insufficiency as a side-effect of combined treatment of itraconazole and budesonide. Eur Respir J. 2002 Jul;20(1):127–33.
  189. Santhana Krishnan SG, Cobbs RK. Reversible acute adrenal insufficiency caused by fluconazole in a critically ill patient. Postgrad Med J. 2006 Sep;82(971):e23.
  190. Miller A, Brooks LK, Poola-Kella S, Malek R. Posaconazole-Induced Adrenal Insufficiency in a Case of Chronic Myelomonocytic Leukemia. Case Rep Endocrinol. 2018;2018:2170484.
  191. Duman AK, Fulco PP. Adrenal Insufficiency With Voriconazole and Inhaled/Intranasal Corticosteroids: Case Report and Systematic Review. J Pharm Pract. 2017 Aug;30(4):459–63.
  192. Araque DP, Zuniga G, Ayala AR. PRIMARY ADRENAL INSUFFICIENCY SECONDARY TO CHRONIC POSACONAZOLE USE. AACE Clin Case Rep. 2020 Apr;6(2):e62–4.
  193. Albert SG, DeLeon MJ, Silverberg AB. Possible association between high-dose fluconazole and adrenal insufficiency in critically ill patients. Crit Care Med. 2001 Mar;29(3):668–70.
  194. Zhao Y, Liang W, Cai F, Wu Q, Wang Y. Fluconazole for Hypercortisolism in Cushing’s Disease: A Case Report and Literature Review. Front Endocrinol. 2020;11:608886.
  195. Lebrun-Vignes B, Archer VC, Diquet B, Levron JC, Chosidow O, Puech AJ, et al. Effect of itraconazole on the pharmacokinetics of prednisolone and methylprednisolone and cortisol secretion in healthy subjects. Br J Clin Pharmacol. 2001 May;51(5):443–50.
  196. Varis T, Kivistö KT, Backman JT, Neuvonen PJ. Itraconazole decreases the clearance and enhances the effects of intravenously administered methylprednisolone in healthy volunteers. Pharmacol Toxicol. 1999 Jul;85(1):29–32.
  197. Beck KR, Odermatt A. Antifungal therapy with azoles and the syndrome of acquired mineralocorticoid excess. Mol Cell Endocrinol. 2021 Mar 15;524:111168.
  198. Thompson GR, Beck KR, Patt M, Kratschmar DV, Odermatt A. Posaconazole-Induced Hypertension Due to Inhibition of 11β-Hydroxylase and 11β-Hydroxysteroid Dehydrogenase 2. J Endocr Soc. 2019 Jul 1;3(7):1361–6.
  199. Hoffmann WJ, McHardy I, Thompson GR. Itraconazole induced hypertension and hypokalemia: Mechanistic evaluation. Mycoses. 2018 May;61(5):337–9.
  200. Boughton C, Taylor D, Ghataore L, Taylor N, Whitelaw BC. Mineralocorticoid hypertension and hypokalaemia induced by posaconazole. Endocrinol Diabetes Metab Case Rep. 2018;2018.
  201. Agarwal N, Apperley L, Taylor NF, Taylor DR, Ghataore L, Rumsby E, et al. Posaconazole-Induced Hypertension Masquerading as Congenital Adrenal Hyperplasia in a Child with Cystic Fibrosis. Case Rep Med. 2020;2020:8153012.
  202. Barton K, Davis TK, Marshall B, Elward A, White NH. Posaconazole-induced hypertension and hypokalemia due to inhibition of the 11β-hydroxylase enzyme. Clin Kidney J. 2018 Oct;11(5):691–3.
  203. Sikka SC, Swerdloff RS, Rajfer J. In vitro inhibition of testosterone biosynthesis by ketoconazole. Endocrinology. 1985 May;116(5):1920–5.
  204. Pont A, Williams PL, Azhar S, Reitz RE, Bochra C, Smith ER, et al. Ketoconazole blocks testosterone synthesis. Arch Intern Med. 1982 Nov;142(12):2137–40.
  205. Pont A, Goldman ES, Sugar AM, Siiteri PK, Stevens DA. Ketoconazole-induced increase in estradiol-testosterone ratio. Probable explanation for gynecomastia. Arch Intern Med. 1985 Aug;145(8):1429–31.
  206. Eil C. Ketoconazole binds to the human androgen receptor. Horm Metab Res Horm Stoffwechselforschung Horm Metab. 1992 Aug;24(8):367–70.
  207. Hugo Perez BS. Ketocazole as an adjunct to finasteride in the treatment of androgenetic alopecia in men. Med Hypotheses. 2004;62(1):112–5.
  208. Patel V, Liaw B, Oh W. The role of ketoconazole in current prostate cancer care. Nat Rev Urol. 2018 Oct;15(10):643–51.
  209. Almeida MQ, Brito VN, Lins TSS, Guerra-Junior G, de Castro M, Antonini SR, et al. Long-term treatment of familial male-limited precocious puberty (testotoxicosis) with cyproterone acetate or ketoconazole. Clin Endocrinol (Oxf). 2008 Jul;69(1):93–8.
  210. Hanger DP, Jevons S, Shaw JT. Fluconazole and testosterone: in vivo and in vitro studies. Antimicrob Agents Chemother. 1988 May;32(5):646–8.
  211. Thompson GR, Surampudi PN, Odermatt A. Gynecomastia and hypertension in a patient treated with posaconazole. Clin Case Rep. 2020 Sep 28;8(12):3158–61.
  212. Cummings AM, Hedge JL, Laskey J. Ketoconazole impairs early pregnancy and the decidual cell response via alterations in ovarian function. Fundam Appl Toxicol Off J Soc Toxicol. 1997 Dec;40(2):238–46.
  213. Pepper G, Brenner SH, Gabrilove JL. Ketoconazole use in the treatment of ovarian hyperandrogenism. Fertil Steril. 1990 Sep;54(3):438–44.
  214. P B, Sj G, D G, A C. Iatrogenic metrorrhagia after the use of itraconazole for onychomycosis. Indian J Pharmacol [Internet]. 2017 Dec [cited 2021 Jun 11];49(6). Available from: https://pubmed.ncbi.nlm.nih.gov/29674803/
  215. Pillans PI, Sparrow MJ. Pregnancy associated with a combined oral contraceptive and itraconazole. N Z Med J. 1993 Oct 13;106(965):436.
  216. Hilbert J, Messig M, Kuye O, Friedman H. Evaluation of interaction between fluconazole and an oral contraceptive in healthy women. Obstet Gynecol. 2001 Aug;98(2):218–23.
  217. Levine MT, Chandrasekar PH. Adverse effects of voriconazole: Over a decade of use. Clin Transplant. 2016 Nov;30(11):1377–86.
  218. Teranishi J, Nagatoya K, Kakita T, Yamauchi Y, Matsuda H, Mori T, et al. Voriconazole-associated salt-losing nephropathy. Clin Exp Nephrol. 2010 Aug;14(4):377–80.
  219. Isobe K, Muraoka S, Sugino K, Yamazaki Y, Kikuchi N, Hamanaka N, et al. [Case of pulmonary aspergillosis associated with inappropriate antidiuretic hormone syndrome caused by voriconazole therapy]. Nihon Kokyuki Gakkai Zasshi. 2007 Jun;45(6):489–93.
  220. Xu R-A, Zheng S-L, Xiao L-L, Cai X-D, Lai X-X, Lin G-Y, et al. Therapeutic drug monitoring in voriconazole-associated hyponatremia. Med Mycol Case Rep. 2013 Jun 19;2:134–6.
  221. Matsumoto K, Ikawa K, Abematsu K, Fukunaga N, Nishida K, Fukamizu T, et al. Correlation between voriconazole trough plasma concentration and hepatotoxicity in patients with different CYP2C19 genotypes. Int J Antimicrob Agents. 2009 Jul;34(1):91–4.
  222. Ho S, Rawlins M, Ingram P, Boan P. Voriconazole-induced hyponatraemia associated with homozygous CYP2C19*2 genotype. J Chemother Florence Italy. 2017 Oct;29(5):325–6.
  223. Lin X-B, Li Z-W, Yan M, Zhang B-K, Liang W, Wang F, et al. Population pharmacokinetics of voriconazole and CYP2C19 polymorphisms for optimizing dosing regimens in renal transplant recipients. Br J Clin Pharmacol. 2018 Jul;84(7):1587–97.
  224. Moon WJ, Scheller EL, Suneja A, Livermore JA, Malani AN, Moudgal V, et al. Plasma fluoride level as a predictor of voriconazole-induced periostitis in patients with skeletal pain. Clin Infect Dis Off Publ Infect Dis Soc Am. 2014 Nov 1;59(9):1237–45.
  225. Thompson GR, Bays D, Cohen SH, Pappagianis D. Fluoride excess in coccidioidomycosis patients receiving long-term antifungal therapy: an assessment of currently available triazoles. Antimicrob Agents Chemother. 2012 Jan;56(1):563–4.
  226. Wermers RA, Cooper K, Razonable RR, Deziel PJ, Whitford GM, Kremers WK, et al. Fluoride excess and periostitis in transplant patients receiving long-term voriconazole therapy. Clin Infect Dis Off Publ Infect Dis Soc Am. 2011 Mar 1;52(5):604–11.
  227. Skiles JL, Imel EA, Christenson JC, Bell JE, Hulbert ML. Fluorosis because of prolonged voriconazole therapy in a teenager with acute myelogenous leukemia. J Clin Oncol Off J Am Soc Clin Oncol. 2011 Nov 10;29(32):e779-782.
  228. Poinen K, Leung M, Wright AJ, Landsberg D. A vexing case of bone pain in a renal transplant recipient: Voriconazole-induced periostitis. Transpl Infect Dis Off J Transplant Soc. 2018 Oct;20(5):e12941.
  229. Adwan MH. Voriconazole-induced periostitis: a new rheumatic disorder. Clin Rheumatol. 2017 Mar;36(3):609–15.
  230. Chitkara M, Rackoff PJ, Beltran LS. Multiple painless masses: periostitis deformans secondary to fluoride intoxication. Skeletal Radiol. 2014 Apr;43(4):529–30, 555–6.
  231. Tanner AR. Hypothyroidism after treatment with ketoconazole. Br Med J Clin Res Ed. 1987 Jan 10;294(6564):125.
  232. Glass AR, Eil C. Ketoconazole-induced reduction in serum 1,25-dihydroxyvitamin D. J Clin Endocrinol Metab. 1986 Sep;63(3):766–9.
  233. G S, S B, Gi B, G DN. Ketoconazole decreases the serum ionized calcium and 1,25-dihydroxyvitamin D levels in tuberculosis-associated hypercalcemia. Am J Dis Child 1960 [Internet]. 1993 Mar [cited 2021 Jun 13];147(3). Available from: https://pubmed.ncbi.nlm.nih.gov/8438806/
  234. Ar G, Jm C, W E, J L, Rw G, C E. Ketoconazole reduces elevated serum levels of 1,25-dihydroxyvitamin D in hypercalcemic sarcoidosis. J Endocrinol Invest [Internet]. 1990 May [cited 2021 Jun 13];13(5). Available from: https://pubmed.ncbi.nlm.nih.gov/2166103/
  235. Sayers J, Hynes AM, Srivastava S, Dowen F, Quinton R, Datta HK, et al. Successful treatment of hypercalcaemia associated with a CYP24A1 mutation with fluconazole. Clin Kidney J. 2015 Aug;8(4):453–5.
  236. Nguyen M, Boutignon H, Mallet E, Linglart A, Guillozo H, Jehan F, et al. Infantile hypercalcemia and hypercalciuria: new insights into a vitamin D-dependent mechanism and response to ketoconazole treatment. J Pediatr. 2010 Aug;157(2):296–302.
  237. Peehl DM, Seto E, Feldman D. Rationale for combination ketoconazole/ vitamin D treatment of prostate cancer. Urology. 2001 Aug;58(2 Suppl 1):123–6.
  238. Ghatak T, Singh RK, Baronia AK. Enteral voriconazole induced hypoglycemia: A potentially life threatening complication. Indian J Pharmacol. 2012;44(1):138–9.
  239. Lobo BL, Miwa LJ, Jungnickel PW. Possible ketoconazole-induced hypoglycemia. Drug Intell Clin Pharm. 1988 Aug;22(7–8):632.
  240. Lyoen M, Rostain F, Grimault A, Minello A, Sgro C. VFEND® (voriconazole)-associated hypoglycaemia without identified drug interaction. Fundam Clin Pharmacol. 2013 Oct;27(5):570–1.
  241. Niemi M, Backman JT, Neuvonen M, Laitila J, Neuvonen PJ, Kivistö KT. Effects of fluconazole and fluvoxamine on the pharmacokinetics and pharmacodynamics of glimepiride. Clin Pharmacol Ther. 2001 Apr;69(4):194–200.
  242. Gunaratne K, Austin E, Wu PE. Unintentional sulfonylurea toxicity due to a drug-drug interaction: a case report. BMC Res Notes. 2018 May 21;11(1):331.
  243. Stuecklin-Utsch A, Hasan C, Bode U, Fleischhack G. Pancreatic toxicity after liposomal amphotericin B. Mycoses. 2002 Jun;45(5–6):170–3.
  244. Kim SW, Yeum CH, Kim S, Oh Y, Choi KC, Lee J. Amphotericin B decreases adenylyl cyclase activity and aquaporin-2 expression in rat kidney. J Lab Clin Med. 2001 Oct;138(4):243–9.
  245. Metzger NL, Varney Gill KL. Nephrogenic diabetes insipidus induced by two amphotericin B liposomal formulations. Pharmacotherapy. 2009 May;29(5):613–20.
  246. Laniado-Laborín R, Cabrales-Vargas MN. Amphotericin B: side effects and toxicity. Rev Iberoam Micol. 2009 Dec 31;26(4):223–7.
  247. Canada TW, Weavind LM, Augustin KM. Possible liposomal amphotericin B-induced nephrogenic diabetes insipidus. Ann Pharmacother. 2003 Jan;37(1):70–3.
  248. Bockenhauer D, Bichet DG. Pathophysiology, diagnosis and management of nephrogenic diabetes insipidus. Nat Rev Nephrol. 2015 Oct;11(10):576–88.
  249. Ishikawa S. Amphotericin B-induced nephrogenic diabetes insipidus. Intern Med Tokyo Jpn. 2005 May;44(5):403.
  250. Kobayashi R, Keino D, Hori D, Sano H, Suzuki D, Kishimoto K, et al. Analysis of Hypokalemia as a Side Effect of Liposomal Amphotericin in Pediatric Patients. Pediatr Infect Dis J. 2018 May;37(5):447–50.
  251. Zager RA, Bredl CR, Schimpf BA. Direct amphotericin B-mediated tubular toxicity: assessments of selected cytoprotective agents. Kidney Int. 1992 Jun;41(6):1588–94.
  252. Barton CH, Pahl M, Vaziri ND, Cesario T. Renal magnesium wasting associated with amphotericin B therapy. Am J Med. 1984 Sep;77(3):471–4.
  253. Johansen HK, Gøtzsche PC. Amphotericin B lipid soluble formulations versus amphotericin B in cancer patients with neutropenia. Cochrane Database Syst Rev [Internet]. 2014 Sep 4 [cited 2021 Jun 13];2014(9). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6457843/
  254. Sato K, Hayashi M, Utsugi M, Ishizuka T, Takagi H, Mori M. Acute pancreatitis in a patient treated with micafungin. Clin Ther. 2007 Jul;29(7):1468–73.
  255. Smith PB, Steinbach WJ, Cotten CM, Schell WA, Perfect JR, Walsh TJ, et al. Caspofungin for the treatment of azole resistant candidemia in a premature infant. J Perinatol Off J Calif Perinat Assoc. 2007 Feb;27(2):127–9.
  256. Xue S, Gu R, Wu T, Zhang M, Wang X. Oral potassium iodide for the treatment of sporotrichosis. Cochrane Database Syst Rev. 2009 Oct 7;(4):CD006136.
  257. Costa RO, de Macedo PM, Carvalhal A, Bernardes-Engemann AR. Use of potassium iodide in Dermatology: updates on an old drug. An Bras Dermatol. 2013;88(3):396–402.

 

Hypoglycemia During Therapy of Diabetes

ABSTRACT

Hypoglycemia, caused by treatment with a sulfonylurea, a glinide, or insulin coupled with compromised defenses against the resulting falling plasma glucose concentrations, is the limiting factor in the glycemic management of diabetes. It causes recurrent morbidity in most people with type 1 diabetes mellitus (T1DM) and many with advanced type 2 diabetes mellitus (T2DM) and is sometimes fatal; it limits maintenance of physiologic normoglycemia over a lifetime of diabetes; and it impairs physiological and behavioral defenses against subsequent hypoglycemia. In addition to drug selection and application of diabetes treatment technologies, minimizing hypoglycemia in diabetes includes acknowledging the problem, considering each of the risk factors, and applying the principles of intensive glycemic therapy. For most people with diabetes who are at risk for, or suffering from, iatrogenic hypoglycemia these principles include selecting appropriate individualized glycemic goals and providing structured patient education that will often reduce the incidence of hypoglycemia. That is typically coupled with short-term scrupulous avoidance of hypoglycemia that will often reverse impaired awareness of hypoglycemia.

THE CLINICAL PROBLEM OF HYPOGLYCEMIA IN DIABETES

 

The problem of iatrogenic hypoglycemia in diabetes has been reviewed in detail (1-7).

Glycemic Control

In the context of comprehensive treatment, including weight, blood pressure, and blood lipid control among other measures, glycemic control makes a difference for people with diabetes. Improved glycemic control reduces microvascular complications (retinopathy, nephropathy, and neuropathy) in both type 1 diabetes mellitus (T1DM) (8) and type 2 diabetes mellitus (T2DM) (9, 10). Follow-up of patients with T1DM (11) and T2DM (12) suggests that an improved earlier period of glycemic control may also reduce subsequent macrovascular complications. Clearly, maintenance of physiologic normoglycemia over a lifetime of diabetes would be in the best interest of people with diabetes if that could be accomplished safely.

The Limiting Factor

Iatrogenic hypoglycemia, fundamentally but not exclusively usually results from treatment with an insulin secretagogue or insulin, and is the major limiting factor in the goal of near normoglycemia in the management of diabetes (1, 2). Iatrogenic hypoglycemia causes recurrent morbidity in most people with T1DM and many with advanced T2DM, and is sometimes fatal (5). It impairs defenses against subsequent falling plasma glucose concentrations and thus causes a vicious cycle of recurrent hypoglycemia. It generally precludes maintenance of euglycemia over a lifetime of diabetes and, thus, full realization of the benefits of glycemic control.

Type 1 and Type 2 Diabetes

Iatrogenic hypoglycemia commonly occurs in the overwhelming majority of people with T1DM who must, of course, be treated with insulin. Most have untold numbers of episodes of asymptomatic hypoglycemia. These are not benign since they impair defenses against subsequent hypoglycemia (1, 2). They suffer an average of two episodes of symptomatic hypoglycemia per week – thousands of such episodes over a lifetime of diabetes – and about one episode of temporarily disabling severe (i.e., requiring third party assistance) hypoglycemia per year. Hypoglycemia causes brain fuel deprivation that, if unchecked, results in functional brain failure that is typically corrected after the plasma glucose concentration is raised (13). Rarely, if it is profound and prolonged, can result in brain death (13). Hypoglycemia may lead to cardiac arrhythmias, especially in patients with preexisting cardiac abnormalities (14, 15). Additionally, hypoglycemia has been demonstrated to be pro-coagulant and pro-atherothrombotic (16, 17). Furthermore, severe hypoglycemia has been associated with increased risk of death extending many months and up to one year after the sentinel episode (18). Early reports suggested that 2 to 4% of deaths of people with diabetes, largely T1DM, were the result of hypoglycemia (2, 5). More recent reports suggest that 6 to 10% of deaths of people with T1DM are the result of hypoglycemia (5, 6, 15, 19, 20). Regardless of the actual rate, the fact that there is an iatrogenic hypoglycemia mortality rate is alarming.

Overall, for a given individual, iatrogenic hypoglycemia is less frequent in T2DM (1, 2, 21, 22). However, due to the greatly increased numbers of individuals with T2DM, the prevalence of hypoglycemic episodes is actually greater than in T1DM. Drugs that can cause endogenous or exogenous (insulin) hyperinsulinemia unregulated by glucose can cause hypoglycemia. On the other hand, insulin sensitizers (metformin or a thiazolidinedione), α-glucosidase inhibitors, sodium glucose cotransporter 2 inhibitors, and even those drugs that cause glucose-dependent hyperinsulinemia (glucagon-like peptide-1 receptor agonists or dipeptidyl peptidase-IV inhibitors) among other actions should not, and probably do not, cause hypoglycemia. They do, however, increase the risk of hypoglycemia if used with an insulin secretagogue or with insulin. Even during treatment of T2DM with insulin, hypoglycemia event rates are about one-third of those in T1DM overall (21). However, for reasons discussed shortly (see Glucose Counterregulatory Physiology and its Pathophysiology in Diabetes), the incidence of iatrogenic hypoglycemia increases over time, approaching that in T1DM, as people approach the insulin deficient end of the spectrum of T2DM (22). Because T2DM is roughly 20-fold more prevalent than T1DM and many, perhaps most, people with T2DM ultimately require treatment with insulin, most episodes of hypoglycemia, including those of severe hypoglycemia, occur in individuals with T2DM. Insulin secretagogue and insulin induced hypoglycemia can be fatal in T2DM although precise hypoglycemic mortality rates are as yet known. As many as 10% of patients with severe sulfonylurea-induced hypoglycemia die (23, 24).

DEFINITION AND CLASSIFICATION OF HYPOGLYCEMIA (25, 26)

The American Diabetes Association and the International Hypoglycemia Study Group (Table 1) define clinically significant hypoglycemia as a blood glucose <54 mg/dl (3.0 mmol/L) which is detected by individual’s self-monitoring blood glucose (SMBG) as well as by continuous glucose monitoring ((CGM), glucose values of <54 mg/dl (3.0 mmol/L) for at least 20 min), or laboratory measurement of plasma glucose which is sufficiently low to indicate clinically significant hypoglycemia (25, 26). Blood glucose ≤70 mg/dl (3.9 mmol/L) is considered a hypoglycemia alert value, which represents an important lower glucose cutoff value for treatment with fast acting carbohydrates and dose adjustments of antidiabetic medications. Severe hypoglycemia is defined as a low glucose value with severe cognitive impairment which requires assistance from another person in order to achieve recovery (27). Relative hypoglycemia or pseudohypoglycemia represents an event during which the person with diabetes reports any of the typical symptoms of hypoglycemia and interprets those as indicative of hypoglycemia with a measured plasma glucose concentration >70 mg/dL (>3.9 mmol/L).

Table 1. Classification of Hypoglycemia in Diabetes (25, 26)

Level

Glycemic criteria

 

Hypoglycemia alert value (level 1)

≤70 mg/dl (3.9 mmol/L)

Sufficiently low for treatment with fast acting carbohydrate and dose adjustment of glucose lowering therapy

Clinically significant hypoglycemia (level 2)

<54 mg/dl (3.0 mmol/L)

Sufficiently low to indicate serious, clinically important hypoglycemia

Severe hypoglycemia (level 3)

No specific glucose threshold

Hypoglycemia associated with severe cognitive impairment requiring external assistance for recovery

COMPLICATIONS OF HYPOGLYCEMIA

Increased mortality has been observed in randomized controlled trials during more aggressive compared with less aggressive glucose-lowering therapy in patients with T2DM (28) and in patients with hypoglycemia in intensive care units (29). The associations between increased hypoglycemia and increased mortality during aggressive glycemic therapy in these and other (19, 30, 31) trials have been thought to be multifactorial (32). Although central to the defense against a falling glucose, the release of catecholamines in response to hypoglycemia may lead to increased cardiac workload, which may worsen an already compromised heart, especially in patients with preexisting coronary artery disease (32). Hypoglycemia has also been found to cause abnormal cardiac repolarization with a prolongation of QTc (33), an increase that may lead to severe cardiac arrhythmias (34). Acute hypoglycemia has also been demonstrated to result in an increased pro-coagulant state, release of inflammatory cytokines, and ultimately to endothelial dysfunction and vascular injury (35, 36). The clinical implication of these findings is that overly aggressive glucose-lowering therapy of diabetes, with currently available methods, may cause excess mortality.

The association of hypoglycemia with cognitive function appears to be more complicated. Among older individuals with type 2 diabetes, a history of severe hypoglycemia was associated with greater risk of dementia (37). The ACCORD study reported that cognitive impairment at baseline and a continuing decline in cognitive function among individuals was found to be associated with greater risk for dementia following hypoglycemia (38). It should be noted however that in DCCT/EDIC, which involved much younger participants, no association of severe hypoglycemia and cognitive decline was found (25, 39).

GLUCOSE COUNTERREGULATORY PHYSIOLOGY AND ITS PATHOPHYSIOLOGY IN DIABETES

Physiology

In nondiabetic individuals, there are a number of physiological defenses against falling plasma glucose concentrations. These include reductions in insulin secretion, which occur as glucose levels decline within the physiological range. This allows for increased hepatic (and renal) glucose production, and increments in glucagon and epinephrine secretion, which occur as glucose levels fall just below the physiological range and stimulate hepatic glucose production (1-3) (Figure 1). Increased epinephrine levels also normally mobilize gluconeogenic precursors from muscle and fat, stimulate renal glucose production, limit glucose utilization by muscle and fat, and limit insulin secretion (3). The behavioral defense against falling plasma glucose concentrations is carbohydrate ingestion prompted largely by the perception of neurogenic (autonomic) symptoms (e.g., palpitations, tremor, and anxiety/arousal which are catecholamine-mediated or adrenergic and sweating, hunger, and paresthesias which are sympatho-adrenal mediated or cholinergic) (40, 41) (Figure 1). These are largely sympathetic neural, rather than adrenomedullary, in origin (41). The extent to which mild neuroglycopenic symptoms such as altered mentation or psychomotor changes contribute to the patient’s recognition of hypoglycemia is unclear; awareness of hypoglycemia is largely prevented by pharmacological antagonism of neurogenic symptoms (40). Severe neuroglycopenic symptoms include frank confusion, acute focal or central neurologic deficits, seizure and/or loss of consciousness. All of these defenses can be compromised in T1DM and advanced T2DM (1, 2, 42, 43).

Pathophysiology

Episodes of therapeutic hyperinsulinemia, the result of glucose unregulated delivery of endogenous (insulin secretagogue therapy) or exogenous (insulin therapy) insulin into the circulation, initiate the sequence that may, or may not, culminate in an episode of hypoglycemia (1, 2). Absolute therapeutic insulin excess of sufficient magnitude can cause isolated episodes of hypoglycemia despite intact glucose counterregulatory defenses against hypoglycemia (Figure 2). But that is an uncommon event. Iatrogenic hypoglycemia is typically the result of the interplay of mild-moderate absolute therapeutic insulin excess, reduced glucose intake, exercise, increased insulin sensitivity, sleep, and existing or induced compromised physiological and behavioral defenses against falling plasma glucose concentrations in T1DM (1, 2, 42) and T2DM (1, 2, 43). In T1DM, because of β-cell failure, insulin levels do not decrease as glucose levels fall; the first physiological defense is lost. Furthermore, glucagon levels do not increase as glucose levels fall (44); the second physiological defense is lost. That, too, is possibly attributable to a β-cell signaling failure since a decrease in β-cell secretion, coupled with a low α-cell glucose concentration, normally signals α-cell glucagon secretion (4, 45, 46). Finally, the increase in epinephrine levels as glucose levels fall is also attenuated (1, 2, 43); and thus, the three major physiological defenses are compromised.

Figure 1. Physiological and Behavioral Defenses Against Hypoglycemia in Humans. ACH, acetylcholine; NE, norepinephrine; PNS, parasympathetic nervous system; SNS, sympathetic nervous system. From reference (47).

Although it is often caused by recent antecedent hypoglycemia (42, 48) or by prior exercise (49) or sleep (50-52), the mechanism of the attenuated sympathoadrenal response to falling glucose levels is unknown (4). Nonetheless, the attenuated epinephrine response is a marker of an attenuated sympathetic neural response (41) and the latter largely results in the reduction of the symptoms of hypoglycemia causing hypoglycemia unawareness (or impaired awareness of hypoglycemia) and thus loss of the behavioral defense, i.e., carbohydrate ingestion. In the setting of therapeutic hyperinsulinemia, falling plasma glucose concentrations, absent decrements in insulin, absent increments in glucagon, and attenuated increments in epinephrine cause the clinical syndrome of defective glucose counter-regulation (1, 2, 42), which is associated with a 25-fold (53) or greater (54) increased risk of iatrogenic hypoglycemia. The attenuated sympathoadrenal, particularly the attenuated sympathetic neural response, causes the clinical syndrome of hypoglycemia unawareness (1, 2) which is associated with a 6-fold increased risk of iatrogenic hypoglycemia (55).

The pathophysiology of glucose counter-regulation is the same in T1DM and T2DM albeit with different time courses. β-cell failure, and therefore loss of the insulin and glucagon responses to falling plasma glucose concentrations, develops early in T1DM but more gradually in T2DM. Thus, iatrogenic hypoglycemia, becomes a common problem early in T1DM and later in T2DM.

The concept of hypoglycemia-associated autonomic failure (HAAF) in diabetes (1, 2, 4, 6, 42, 43) (Figure 2) posits that recent antecedent hypoglycemia, as well as prior exercise or sleep, causes both defective glucose counter-regulation (by reducing increments in epinephrine in the setting of absent decrements in insulin and absent increments in glucagon during subsequent hypoglycemia) and hypoglycemia unawareness (by reducing sympathoadrenal and resulting neurogenic symptom responses during subsequent hypoglycemia) and, therefore, a vicious cycle of recurrent hypoglycemia. Supporting this concept is the finding, that as little as 2-3 weeks of scrupulous avoidance of hypoglycemia reverses hypoglycemia unawareness and improves the attenuated epinephrine component of defective glucose counter-regulation in most affected patients. (56-59).

The mechanism(s) of the attenuated sympathoadrenal response to falling glucose levels, the key feature of HAAF, is not known (4). It must involve the central nervous system or the afferent of efferent components of the sympathoadrenal system. Theories include increased blood-to-brain transport of a metabolic fuel, effects of a systemic mediator such as cortisol on the brain, altered hypothalamic mechanisms, and activation of an inhibitory cerebral network mediated through the thalamus (4).

Figure 2. Schematic Diagram of HAAF in Diabetes. From reference (2). 

RISK FACTORS FOR HYPOGLYCEMIA IN DIABETES

Conventional Risk Factors

The conventional risk factors are based on the premise that relative to low rates of glucose delivery into the circulation, high rates of glucose efflux out of the circulation, or both, or absolute therapeutic hyperinsulinemia is the sole determinant of risk (1, 2). They include (but are not limited to):

  1. Insulin (or insulin secretagogue) doses are excessive, ill-timed, or of the wrong type.
  2. Exogenous glucose delivery is decreased (as following missed meals and during the overnight fast, with gastroparesis or celiac disease).
  3. Glucose utilization and sensitivity to insulin are increased (as during and shortly after exercise, in the middle of the night, following weight loss, or improved glycemic control).
  4. Endogenous glucose production is decreased (as following alcohol ingestion or in liver failure).
  5. Insulin clearance is decreased (as in renal failure).
  6. Classical diabetic autonomic neuropathy.

Patients with diabetes and their caregivers must consider each of these risk factors carefully whenever hypoglycemia is a problem. (60).

Risk Factors Indicative of Hypoglycemia-Associated Autonomic Failure (HAAF)

These risk factors stem directly from the pathophysiology of glucose counter-regulation and the concept of HAAF in diabetes (1, 2, 42, 43). They include:

  1. The degree of absolute endogenous insulin deficiency. This determines the extent to which insulin levels will not decrease and glucagon levels will not increase as plasma glucose concentrations fall in response to therapeutic hyperinsulinemia. It is in part a function of the duration of diabetes.
  2. A history of severe hypoglycemia, hypoglycemia unawareness, or both as well as recent antecedent hypoglycemia, prior exercise or sleep.
  3. Aggressive glycemic therapy per se (lower A1C levels, lower glycemic goals). Studies with a control group treated to higher mean glycemia consistently document higher rates of hypoglycemia in the group treated to lower mean glycemia (e.g. (5)). Mean glycemia is a risk factor for hypoglycemia. However, hypoglycemia can occur in individuals with any A1C level, and the fact that mean glycemia is a risk factor does not mean that one cannot both lower mean glycemia and reduce the risk of hypoglycemia in individual patients (7).

PREVENTION OF HYPOGLYCEMIA IN DIABETES

Obviously, it is preferable to prevent, rather than treat, hypoglycemia in people with diabetes. The prevention of hypoglycemia requires the practice of hypoglycemia risk reduction (1, 2, 7, 61). That involves four steps: 1) Acknowledge the problem. 2) Consider the conventional risk factors in diabetes. 3) Consider the risk factors indicative of HAAF in diabetes. 4) Apply the relevant principles of intensive glycemic therapy of diabetes.

Acknowledge the Problem

The issue of hypoglycemia should be addressed at every contact with a patient treated with an insulin secretagogue or with insulin (7). In addition to the patient’s comments and review of the individual’s SMBG data (as well as any CGM data) we find it especially helpful to inquire what is the glucose level when each patient can detect hypoglycemia and what are the symptoms and signs at various hypoglycemic levels. It is also often helpful to question close associates of the patient since they may have observed clues to episodes of hypoglycemia. Patient concerns about the reality, or even the possibility, of hypoglycemia can be a barrier to glycemic control (62, 63). Their concerns need to be discussed and addressed if hypoglycemia is a real or perceived problem.

Consider the Conventional Risk Factors for Hypoglycemia in Diabetes

Each of the risk factors that result in relative or absolute therapeutic hyperinsulinemia, as just discussed, should be considered carefully in any patient with iatrogenic hypoglycemia. Those include the dose, timing, and type of the insulin secretagogue or insulin preparations(s) used, and conditions in which exogenous glucose delivery or endogenous glucose production is decreased, glucose utilization or insulin sensitivity is increased or insulin clearance is decreased.

Consider the Risk Factors Indicative of HAAF in Diabetes

As detailed earlier, the risk factors indicative of HAAF include the degree of absolute endogenous insulin deficiency, a history of severe hypoglycemia, impaired awareness of hypoglycemia, or both as well as any relationship between iatrogenic hypoglycemia and recent antecedent hypoglycemia, prior exercise or sleep, and lower glycemic goals. A history of severe hypoglycemia is a clinical red flag. Without a fundamental adjustment of the treatment regimen, the likelihood of another episode is high (8, 64).

Apply the Relevant Principles of Intensive Glycemic Therapy

The principles of intensive glycemic therapy relevant to minimizing the risk of iatrogenic hypoglycemia in diabetes include drug selection, selective application of diabetes treatment technologies, individualized glycemic goals, structured patient education, and short-term scrupulous avoidance of hypoglycemia (7). Based on the premise that the risk of hypoglycemia is modifiable, the International Hypoglycaemia Study Group recommended that people with diabetes treated with a sulfonylurea, a glinide, or insulin should be educated about hypoglycemia, should treat self-monitoring of plasma glucose (SMPG) <70 mg/dL (<3.9 mmol/L) to avoid progression to clinical iatrogenic hypoglycemia, and should regularly be queried about hypoglycemia, including the glucose level at which symptoms develop (7).

Drug selection relevant to minimizing the risk of hypoglycemia includes avoidance, if possible, of sulfonylureas or glinides, the use of more physiological insulin regimens (65), and the use of long-acting or even ultra-long-acting basal insulin analogues and rapid-acting prandial insulin analogues (66-69). Insulin analogues reduce the frequency of at least nocturnal hypoglycemia (66-68) including severe nocturnal hypoglycemia (68) compared to human insulins. In insulin-requiring T2DM, basal insulins are associated with less hypoglycemia than prandial insulin regimens. Furthermore, (and perhaps in the future in T1DM) the combination of a long-acting basal insulin with a glucose-lowering drug that does not cause hypoglycemia (e.g., a GLP-1 receptor agonist) may result in less hypoglycemia than with the use of basal-bolus insulin therapy (70).

Relevant diabetes treatment technologies include continuous subcutaneous insulin infusion (CSII), continuous glucose monitoring (CGM), and combinations of CSII and CGM. Although earlier meta-analyses disclosed little (71) or no (72) advantage of CSII, recent evidence suggest that CSII treatment is superior in achieving glucose control compared to multiple daily injections (73, 74). CGM devices alone have been shown to improve glycemic control and decrease duration of hypoglycemia in patients with diabetes mellitus (75, 76). As their accuracy is continuously improving, several CGM systems have been approved by the FDA, and other regulatory authorities to even replace point of care blood glucose testing (77, 78). Real-time CGM systems have been also found to improve hypoglycemia awareness, without a change in A1C,  in a small group of patients with T1DM (79).  A favorable experience with CSII has been also reported (80, 81). The combination of CSII and real-time CGM – sensor augmented pump therapy, particularly that including an insulin pump programmed to stop insulin infusion for up to two hours when CGM values fall to a selected glucose level (“low glucose suspend”) – has been reported to reduce the frequency of severe hypoglycemia in T1DM (82-84). Recent innovations have included cessation of insulin delivery during hypoglycemia. Several promising studies have investigated approaches for leading closed-loop insulin (or insulin and glucagon) replacement. The development of automated closed-loop insulin pumps represents an area of ongoing research and fully closed-loop insulin (85) or insulin and glucagon replacement (86) and pancreatic islet transplantation (87) will undoubtedly eliminate hypoglycemia and improve overall glycemic control. A hybrid-not fully automated -system (as only basal insulin is automatically adjusted) has received approval by the FDA (88).

Special circumstances relevant to drug selection and treatment technologies in the prevention of hypoglycemia in diabetes include exercise, the overnight period, the elderly, drivers, and pregnancy. Especially in insulin-treated patients hypoglycemia can occur during or shortly after exercise (89) or late after exercise (90, 91). Measures to avoid early-onset exercise hypoglycemia include interspersing episodes of intense exercise (which tends to raise plasma glucose concentrations), adding carbohydrate ingestion, and reducing insulin doses (92). A consistent observation since the DCCT (8) is that more than half of episodes of hypoglycemia, including severe hypoglycemia, occur during the night. That is typically the longest interval between meals and between SMPG and includes the time of maximal sensitivity to insulin. In addition to the use of insulin analogues, sensor augmented pump therapy or close-loop insulin or insulin and glucagon replacement, all discussed earlier, approaches to the prevention of nocturnal hypoglycemia include attempts to produce sustained delivery of exogenous carbohydrate or sustained endogenous glucose production (93). With respect to the former approach, a conventional bedtime snack or bedtime administration of uncooked cornstarch have not been found to be consistently effective (93). With respect to the latter approach an experimental treatment is bedtime administration of a β2­-adrenergic agonist such as terbutaline (93-95). In addition to HAAF, comorbidities including renal insufficiency, polypharmacy, and impaired cognition are more relevant to the development of hypoglycemia in older individuals (96). Drivers with diabetes and a history of recurrent hypoglycemia-related driving mishaps have been found to have greater driving simulator impairments (97). Finally, up to 45% of pregnant women with type 1 diabetes experience severe hypoglycemia especially in the first trimester (98).

Individualized Glycemic Goal

Glycemic goals should be individualized in patients with diabetes (5, 99). The selection of a glycemic goal in a person with diabetes is a trade-off between the benefits of glycemic control – partial prevention or delay of microvascular complications – and the risk of recurrent morbidity, and potential mortality, of hypoglycemia (5). A reasonable individualized glycemic goal is the lowest A1C that does not cause severe hypoglycemia and preserves awareness of hypoglycemia, preferably with little or no symptomatic or even asymptomatic hypoglycemia, at a given stage in the evolution of the individual’s diabetes (5). Thus, the glycemic goal should be linked not only to the level of glycemic control (i.e., the A1C) but also to the risk of hypoglycemia, specifically the drugs used (a sulfonylurea, a glinide, or insulin), the degree of endogenous insulin deficiency, and the anticipated benefit of the targeted level of glycemic control. A nondiabetic A1C would be reasonable in a patient with early T2DM treated effectively with lifestyle changes and/or drugs that do not cause hypoglycemia. For the majority of non-pregnant adults a reasonable goal for an A1C is <7% (53 mmol/mol). For selected individuals with long life expectancy, without significant comorbidities (especially cardiovascular disease), stringent A1c goals (<6.5% (48 mmol/mol)) should be targeted, if this can be achieved without significant hypoglycemia (25).  For children and adolescents, an A1C of <7.5% (58 mmol/mol) should be the goal, although a lower target (<7% (53 mmol/mol)) should be reasonable if it can be achieved without excessive hypoglycemia (100). However much higher levels of A1C (7.5%-8.0% (58-64 mmol/mol)) may be appropriate in elderly patients where hypoglycemia may be harmful.  Even higher targets (A1C<8.5% (69 mmol/mol)) may be appropriate in individuals with very limited life expectancy (101).

Of note, it needs to be underscored that severe hypoglycemia can and does occur at A1C levels between 8-10% (64-86 mmol/mol) in either T1DM or T2DM.  Thus, severe hypoglycemia is not just a consequence of “low or near normal” A1C values.  Of concern are recent data that severe hypoglycemia occurring in T2DM individuals >60 years with elevated A1C may have greater serious adverse events and increased mortality compared to individuals with improved glycemic control and lower A1C values.

Thus, attempts to improve glycemic control with insulin in T2DM individuals that have been resistant or proven challenging to strategies to lower glucose levels may be at greater risk for severe hypoglycemia and associated serious adverse events (19, 28, 30, 31).

Structured Patient Education

The core approach, applicable to virtually all patients with diabetes treated with a sulfonylurea, a glinide, or insulin in whom hypoglycemia becomes a problem, is thorough, structured patient education (often re-education) that teaches the patient how and when their drugs can cause hypoglycemia, how to adjust their medications, meal plans and exercise to optimize glycemic control and minimize hypoglycemia, and how to recognize and treat hypoglycemia (7). Based conceptually on earlier inpatient education programs (102), there is increasing evidence that outpatient structured education programs decrease hypoglycemia, often with a decrease in A1C (103-107). For example, a structured patient education program in flexible insulin therapy led to a reduction of impaired awareness of hypoglycemia (45% of those with impaired awareness initially were aware at one year) and a reduction in severe hypoglycemia (from 1.9 to 0.6 episodes per patient-year and a small but significant decrease in A1C in patients with type 1 diabetes (104). Patient education needs to cover a broad range of information and skill training and often include a motivational element (7).

Short-Term Scrupulous Avoidance of Hypoglycemia

In patients with impaired awareness of hypoglycemia structured patient education should be combined with 2- to 3-weeks of scrupulous avoidance of hypoglycemia – which may require acceptance of somewhat higher glycemic goals in the short-term – since that can be expected to restore awareness of hypoglycemia in most affected patients (56-59).

In summary, people with diabetes treated with a sulfonylurea, a glinide, or insulin should be educated about hypoglycemia, should treat SMPG (or CGM) glucose levels <70 mg/dL (<3.9 mmol/L) to avoid progression to clinical iatrogenic hypoglycemia, and should regularly be queried about hypoglycemia, including the SMPG (or CGM) level at which symptoms develop (7).

TREATMENT OF HYPOGLYCEMIA IN DIABETES

Most episodes of asymptomatic hypoglycemia, detected by routine SMBG or CGM, or of mild-moderate symptomatic hypoglycemia are effectively self-treated by ingestion of glucose tablets or carbohydrate containing juice, soft drinks, candy, other snacks, or a meal (1, 2, 108). A reasonable dose is 20 g of carbohydrate (108). The dose can be repeated in 15 to 20 minutes, if necessary. Since the glycemic response to oral glucose is transient – roughly two hours in the setting of ongoing hyperinsulinemia (108) – the ingestion of a more substantial snack or meal shortly after the plasma glucose level is raised is generally advisable.

When a hypoglycemic patient is unwilling (because of neuroglycopenia) or unable to take carbohydrate orally, parenteral therapy is required. That is often glucagon injected subcutaneously or intramuscularly by an associate of the patient who has been trained to recognize and treat severe hypoglycemia. The usual glucagon dose is 1.0 mg; that can be life-saving although it causes substantial, albeit transient, hyperglycemia (108) and can cause nausea, and even vomiting. Smaller doses (e.g., 150 mcg), repeated if necessary, have been found to be effective without side effects in adolescents (109). Recent advances include 1) approval of nasal glucagon and of a device to deliver glucagon intranasally (111), that would obviate the need for parenteral injection and 2) a glucagon that is stable in solution (110), that would obviate the need to reconstitute the drug prior to administration. Because it also stimulates insulin secretion, glucagon might be less effective in patients with early T2DM. In a medical setting intravenous glucose, 25 g initially, is the standard parenteral therapy (1, 2). The glycemic response to intravenous glucose is, of course, transient. A subsequent glucose infusion is generally needed, and food should be provided as soon as the patient is able to ingest it safely.

The duration of a hypoglycemic episode is a function of its cause. While that caused by a short-acting insulin secretagogue or a rapid-acting insulin can be measured in hours, that caused by a long-acting insulin secretagogue or insulin can last for days requiring hospitalization for prolonged therapy. The duration of secretagogue-induced hypoglycemia can be shortened by administration of octreotide (112, 113).

In the UK, the Joint British Diabetes Societies for Inpatient Care have produced guidance on the management of hypoglycemia for hospital inpatients, although these can be used in the community setting as necessary (114).

ACKNOWLEDGMENTS AND DISCLOSURES

Hugh A. Davis has no disclosures to report.

Elias K. Spanakis has received research support (CGM supplies) from DEXCOM (San Diego, CA) for the conduction of inpatient CGM clinical studies.

Stephen N. Davis- This work has received support from the NIH, NHLBI, NIDDK, JDRF and VA.

REFERENCES

  1. Cryer, P.E., The barrier of hypoglycemia in diabetes. Diabetes, 2008. 57(12): p. 3169-76.
  2. Cryer, P.E., Hypoglycemia in Diabetes: Pathophysiology, Prevalence and Prevention. 2nd ed. 2013, Alexandria, VA: American Diabetes Association.
  3. Cryer, P.E., The Prevention and correction of hypoglycemia., in Handbook of Physiology. Volume II. The Endocrine Pancreas and Regulation of Metaboism, L.S. Jefferson and A.D. Cherrington, Editors. 2001, Oxford University Press: New York. p. 1057-1092.
  4. Cryer, P.E., Mechanisms of hypoglycemia-associated autonomic failure in diabetes. N Engl J Med, 2013. 369(4): p. 362-72.
  5. Cryer, P.E., Glycemic goals in diabetes: trade-off between glycemic control and iatrogenic hypoglycemia. Diabetes, 2014. 63(7): p. 2188-95.
  6. Cryer, P.E., Hypoglycemia-associated automatic failure in diabetes: maladaptive, adaptive, or both? Diabetes, 2015: p. In Press.
  7. International Hypoglycaemia Study Group, Minimizing hypoglycemia in diabetes. Diabetes Care, 2015: p. In Press.
  8. The Diabetes Control and Complications Trial Research Group, The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med, 1993. 329(14): p. 977-86.
  9. UK Prospective Diabetes Study (UKPDS) Group, Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet, 1998. 352(9131): p. 837-53.
  10. UK Prospective Diabetes Study (UKPDS) Group, Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet, 1998. 352(9131): p. 854-65.
  11. The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group, et al., Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med, 2005. 353(25): p. 2643-53.
  12. Holman, R.R., et al., 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med, 2008. 359(15): p. 1577-89.
  13. Cryer, P.E., Hypoglycemia, functional brain failure, and brain death. J Clin Invest, 2007. 117(4): p. 868-70.
  14. Stahn, A., et al., Relationship between hypoglycemic episodes and ventricular arrhythmias in patients with type 2 diabetes and cardiovascular diseases: silent hypoglycemias and silent arrhythmias. Diabetes Care, 2014. 37(2): p. 516-20.
  15. Chow, E., et al., Risk of cardiac arrhythmias during hypoglycemia in patients with type 2 diabetes and cardiovascular risk. Diabetes, 2014. 63(5): p. 1738-47.
  16. Frier, B.M., G. Schernthaner, and S.R. Heller, Hypoglycemia and cardiovascular risks. Diabetes Care, 2011. 34 Suppl 2: p. S132-7.
  17. Jialal, I. and S. Dhindsa, Hypoglycemia and the predisposition to cardiovascular disease: Is the pro-inflammatory-pro-coagulant diathesis a plausible explanation? Atherosclerosis, 2016. 251: p. 504-506.
  18. Lee, A.K., et al., The Association of Severe Hypoglycemia With Incident Cardiovascular Events and Mortality in Adults With Type 2 Diabetes. Diabetes Care, 2018. 41(1): p. 104-111.
  19. Mellbin, L.G., et al., Does hypoglycaemia increase the risk of cardiovascular events? A report from the ORIGIN trial. Eur Heart J, 2013. 34(40): p. 3137-44.
  20. Reno, C.M., et al., Severe hypoglycemia-induced lethal cardiac arrhythmias are mediated by sympathoadrenal activation. Diabetes, 2013. 62(10): p. 3570-81.
  21. Donnelly, L.A., et al., Frequency and predictors of hypoglycaemia in Type 1 and insulin-treated Type 2 diabetes: a population-based study. Diabet Med, 2005. 22(6): p. 749-55.
  22. U.K. Hypoglycaemia Study Group, Risk of hypoglycaemia in types 1 and 2 diabetes: effects of treatment modalities and their duration. Diabetologia, 2007. 50(6): p. 1140-7.
  23. Gerich, J.E., Oral hypoglycemic agents. N Engl J Med, 1989. 321(18): p. 1231-45.
  24. Holstein, A. and E.H. Egberts, Risk of hypoglycaemia with oral antidiabetic agents in patients with Type 2 diabetes. Exp Clin Endocrinol Diabetes, 2003. 111(7): p. 405-14.
  25. American Diabetes, Association., 6. Glycemic Targets: Standards of Medical Care in Diabetes-2018. Diabetes Care, 2018. 41(Suppl 1): p. S55-S64.
  26. International Hypoglycaemia Study Group., Glucose concentrations of less than 3.0 mmol/l (54 mg/dl) should be reported in clinical trials: a joint position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia, 2017. 60(1): p. 3-6.
  27. Seaquist, E.R., et al., Hypoglycemia and diabetes: a report of a workgroup of the American Diabetes Association and the Endocrine Society. Diabetes Care, 2013. 36(5): p. 1384-95.
  28. ACCORD Study Group, et al., Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med, 2008. 358(24): p. 2545-59.
  29. Finfer, S., et al., Intensive versus conventional glucose control in critically ill patients. N Engl J Med, 2009. 360(13): p. 1283-97.
  30. Zoungas, S., et al., Severe hypoglycemia and risks of vascular events and death. N Engl J Med, 2010. 363(15): p. 1410-8.
  31. Duckworth, W., et al., Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med, 2009. 360(2): p. 129-39.
  32. Davis, I.C., et al., Understanding the impact of hypoglycemia on the cardiovascular system. Expert Rev Endocrinol Metab, 2017. 12(1): p. 21-33.
  33. Robinson, R.T., et al., Mechanisms of abnormal cardiac repolarization during insulin-induced hypoglycemia. Diabetes, 2003. 52(6): p. 1469-74.
  34. Barr, C.S., et al., QT dispersion and sudden unexpected death in chronic heart failure. Lancet, 1994. 343(8893): p. 327-9.
  35. Joy, N.G., et al., Effects of equivalent sympathetic activation during hypoglycemia on endothelial function and pro-atherothrombotic balance in healthy individuals and obese standard treated type 2 diabetes. Metabolism, 2016. 65(12): p. 1695-1705.
  36. Joy, N.G., et al., Effects of Acute and Antecedent Hypoglycemia on Endothelial Function and Markers of Atherothrombotic Balance in Healthy Humans. Diabetes, 2015. 64(7): p. 2571-80.
  37. Whitmer, R.A., et al., Hypoglycemic episodes and risk of dementia in older patients with type 2 diabetes mellitus. JAMA, 2009. 301(15): p. 1565-72.
  38. Punthakee, Z., et al., Poor cognitive function and risk of severe hypoglycemia in type 2 diabetes: post hoc epidemiologic analysis of the ACCORD trial. Diabetes Care, 2012. 35(4): p. 787-93.
  39. Diabetes, C., et al., Long-term effect of diabetes and its treatment on cognitive function. N Engl J Med, 2007. 356(18): p. 1842-52.
  40. Towler, D.A., et al., Mechanism of awareness of hypoglycemia. Perception of neurogenic (predominantly cholinergic) rather than neuroglycopenic symptoms. Diabetes, 1993. 42(12): p. 1791-8.
  41. DeRosa, M.A. and P.E. Cryer, Hypoglycemia and the sympathoadrenal system: neurogenic symptoms are largely the result of sympathetic neural, rather than adrenomedullary, activation. Am J Physiol Endocrinol Metab, 2004. 287(1): p. E32-E41.
  42. Dagogo-Jack, S.E., S. Craft, and P.E. Cryer, Hypoglycemia-associated autonomic failure in insulin-dependent diabetes mellitus. J Clin Invest, 1993. 91(3): p. 819-28.
  43. Segel, S.A., D.S. Paramore, and P.E. Cryer, Hypoglycemia-associated autonomic failure in advanced type 2 diabetes. Diabetes, 2002. 51(3): p. 724-33.
  44. Gerich, J.E., et al., Lack of glucagon response to hypoglycemia in diabetes: evidence for an intrinsic pancreatic alpha cell defect. Science, 1973. 182(4108): p. 171-3.
  45. Raju, B. and P.E. Cryer, Loss of the decrement in intraislet insulin plausibly explains loss of the glucagon response to hypoglycemia in insulin-deficient diabetes. Diabetes, 2005. 54(3): p. 757-64.
  46. Cooperberg, B.A. and P.E. Cryer, Beta-cell-mediated signaling predominates over direct alpha-cell signaling in the regulation of glucagon secretion in humans. Diabetes Care, 2009. 32(12): p. 2275-80.
  47. Cryer, P.E., Mechanisms of sympathoadrenal failure and hypoglycemia in diabetes. J Clin Invest, 2006. 116(6): p. 1470-3.
  48. Heller, S.R. and P.E. Cryer, Reduced neuroendocrine and symptomatic responses to subsequent hypoglycemia after 1 episode of hypoglycemia in nondiabetic humans. Diabetes, 1991. 40(2): p. 223-6.
  49. Ertl, A.C. and S.N. Davis, Evidence for a vicious cycle of exercise and hypoglycemia in type 1 diabetes mellitus. Diabetes Metab Res Rev, 2004. 20(2): p. 124-30.
  50. Jones, T.W., et al., Decreased epinephrine responses to hypoglycemia during sleep. N Engl J Med, 1998. 338(23): p. 1657-62.
  51. Banarer, S. and P.E. Cryer, Sleep-related hypoglycemia-associated autonomic failure in type 1 diabetes: reduced awakening from sleep during hypoglycemia. Diabetes, 2003. 52(5): p. 1195-203.
  52. Schultes, B., et al., Defective awakening response to nocturnal hypoglycemia in patients with type 1 diabetes mellitus. PLoS Med, 2007. 4(2): p. e69.
  53. White, N.H., et al., Identification of type I diabetic patients at increased risk for hypoglycemia during intensive therapy. N Engl J Med, 1983. 308(9): p. 485-91.
  54. Bolli, G.B., et al., A reliable and reproducible test for adequate glucose counterregulation in type I diabetes mellitus. Diabetes, 1984. 33(8): p. 732-7.
  55. Geddes, J., et al., Prevalence of impaired awareness of hypoglycaemia in adults with Type 1 diabetes. Diabet Med, 2008. 25(4): p. 501-4.
  56. Fanelli, C.G., et al., Meticulous prevention of hypoglycemia normalizes the glycemic thresholds and magnitude of most of neuroendocrine responses to, symptoms of, and cognitive function during hypoglycemia in intensively treated patients with short-term IDDM. Diabetes, 1993. 42(11): p. 1683-9.
  57. Fanelli, C., et al., Long-term recovery from unawareness, deficient counterregulation and lack of cognitive dysfunction during hypoglycaemia, following institution of rational, intensive insulin therapy in IDDM. Diabetologia, 1994. 37(12): p. 1265-76.
  58. Cranston, I., et al., Restoration of hypoglycaemia awareness in patients with long-duration insulin-dependent diabetes. Lancet, 1994. 344(8918): p. 283-7.
  59. Dagogo-Jack, S., C. Rattarasarn, and P.E. Cryer, Reversal of hypoglycemia unawareness, but not defective glucose counterregulation, in IDDM. Diabetes, 1994. 43(12): p. 1426-34.
  60. The Diabetes Control and Complications Trial Research Group, Epidemiology of severe hypoglycemia in the Diabetes Control and Complications Trial. Am J Med, 1991. 90(4): p. 450-9.
  61. Cryer, P.E., S.N. Davis, and H. Shamoon, Hypoglycemia in diabetes. Diabetes Care, 2003. 26(6): p. 1902-12.
  62. Gonder-Frederick, L.A., et al., Predictors of fear of hypoglycemia in adolescents with type 1 diabetes and their parents. Pediatr Diabetes, 2006. 7(4): p. 215-22.
  63. Nordfeldt, S. and J. Ludvigsson, Fear and other disturbances of severe hypoglycaemia in children and adolescents with type 1 diabetes mellitus. J Pediatr Endocrinol Metab, 2005. 18(1): p. 83-91.
  64. Cox, D.J., et al., Prediction of severe hypoglycemia. Diabetes Care, 2007. 30(6): p. 1370-3.
  65. Rossetti, P., et al., Prevention of hypoglycemia while achieving good glycemic control in type 1 diabetes. Diabetes Care, 2008. 31 (Suppl 2): p. S113-S120.
  66. Siebenhofer, A., et al., Short acting insulin analogues versus regular human insulin in patients with diabetes mellitus. Cochrane Database Syst Rev, 2006(2): p. CD003287.
  67. Horvath, K., et al., Long-acting insulin analogues versus NPH insulin (human isophane insulin) for type 2 diabetes mellitus. Cochrane Database Syst Rev, 2007(2): p. CD005613.
  68. Pedersen-Bjergaard, U., et al., Effect of insulin analogues on risk of severe hypoglycaemia in patients with type 1 diabetes prone to recurrent severe hypoglycaemia (HypoAna Trial): a prospective, randomised, open-label, blinded-endpoint crossover trial. Lancet Diabetes Endocrinol, 2014. 2(7): p. 553-61.
  69. Garber, A.J., et al., Insulin degludec, an ultra-longacting basal insulin, versus insulin glargine in basal-bolus treatment with mealtime insulin aspart in type 2 diabetes (BEGIN Basal-Bolus Type 2): a phase 3, randomised, open-label, treat-to-target non-inferiority trial. Lancet, 2012. 379(9825): p. 1498-507.
  70. Eng, C., et al., Glucagon-like peptide-1 receptor agonist and basal insulin combination treatment for the management of type 2 diabetes: a systematic review and meta-analysis. Lancet, 2014. 384(9961): p. 2228-34.
  71. Fatourechi, M.M., et al., Clinical review: Hypoglycemia with intensive insulin therapy: a systematic review and meta-analyses of randomized trials of continuous subcutaneous insulin infusion versus multiple daily injections. J Clin Endocrinol Metab, 2009. 94(3): p. 729-40.
  72. Yeh, H.C., et al., Comparative effectiveness and safety of methods of insulin delivery and glucose monitoring for diabetes mellitus: a systematic review and meta-analysis. Ann Intern Med, 2012. 157(5): p. 336-47.
  73. Pickup, J.C., Y. Reznik, and A.J. Sutton, Glycemic Control During Continuous Subcutaneous Insulin Infusion Versus Multiple Daily Insulin Injections in Type 2 Diabetes: Individual Patient Data Meta-analysis and Meta-regression of Randomized Controlled Trials. Diabetes Care, 2017. 40(5): p. 715-722.
  74. Benkhadra, K., et al., Continuous subcutaneous insulin infusion versus multiple daily injections in individuals with type 1 diabetes: a systematic review and meta-analysis. Endocrine, 2017. 55(1): p. 77-84.
  75. Beck, R.W., et al., Effect of Continuous Glucose Monitoring on Glycemic Control in Adults With Type 1 Diabetes Using Insulin Injections: The DIAMOND Randomized Clinical Trial. JAMA, 2017. 317(4): p. 371-378.
  76. Beck, R.W., et al., Continuous Glucose Monitoring Versus Usual Care in Patients With Type 2 Diabetes Receiving Multiple Daily Insulin Injections: A Randomized Trial. Ann Intern Med, 2017. 167(6): p. 365-374.
  77. FDA expands indication for continuous glucose monitoring system, first to replace fingerstick testing for diabetes treatment decisions. 2016; Available from: https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm534056.htm.
  78. FDA approves first continuous glucose monitoring system for adults not requiring blood sample calibration. 2017; Available from: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm577890.htm.
  79. Rickels, M.R., et al., Continuous Glucose Monitoring for Hypoglycemia Avoidance and Glucose Counterregulation in Long-Standing Type 1 Diabetes. J Clin Endocrinol Metab, 2018. 103(1): p. 105-114.
  80. Cooper, M.N., et al., A population-based study of risk factors for severe hypoglycaemia in a contemporary cohort of childhood-onset type 1 diabetes. Diabetologia, 2013. 56(10): p. 2164-70.
  81. Johnson, S.R., et al., Long-term outcome of insulin pump therapy in children with type 1 diabetes assessed in a large population-based case-control study. Diabetologia, 2013. 56(11): p. 2392-400.
  82. Bergenstal, R.M., et al., Threshold-based insulin-pump interruption for reduction of hypoglycemia. N Engl J Med, 2013. 369(3): p. 224-32.
  83. Ly, T.T., et al., Effect of sensor-augmented insulin pump therapy and automated insulin suspension vs standard insulin pump therapy on hypoglycemia in patients with type 1 diabetes: a randomized clinical trial. JAMA, 2013. 310(12): p. 1240-7.
  84. Maahs, D.M., et al., A randomized trial of a home system to reduce nocturnal hypoglycemia in type 1 diabetes. Diabetes Care, 2014. 37(7): p. 1885-91.
  85. Leelarathna, L., et al., Day and night home closed-loop insulin delivery in adults with type 1 diabetes: three-center randomized crossover study. Diabetes Care, 2014. 37(7): p. 1931-7.
  86. Russell, S.J., et al., Outpatient glycemic control with a bionic pancreas in type 1 diabetes. N Engl J Med, 2014. 371(4): p. 313-25.
  87. Ang, M., et al., Magnitude and mechanisms of glucose counterregulation following islet transplantation in patients with type 1 diabetes suffering from severe hypoglycaemic episodes. Diabetologia, 2014. 57(3): p. 623-32.
  88. FDA approves first automated insulin delivery device for type 1 diabetes. 2016; Available from: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/UCM522974.htm.
  89. Tansey, M.J., et al., The effects of aerobic exercise on glucose and counterregulatory hormone concentrations in children with type 1 diabetes. Diabetes Care, 2006. 29(1): p. 20-5.
  90. MacDonald, M.J., Postexercise late-onset hypoglycemia in insulin-dependent diabetic patients. Diabetes Care, 1987. 10(5): p. 584-8.
  91. Tsalikian, E., et al., Impact of exercise on overnight glycemic control in children with type 1 diabetes mellitus. J Pediatr, 2005. 147(4): p. 528-34.
  92. Gallen, I.W. Hypoglycemia associated with exercise in people with type 1 diabetes. Diabetic Hypoglycemia Online, 2014. 7, 3.
  93. Raju, B., et al., Nocturnal hypoglycemia in type 1 diabetes: an assessment of preventive bedtime treatments. J Clin Endocrinol Metab, 2006. 91(6): p. 2087-92.
  94. Cooperberg, B.A., et al., Terbutaline and the prevention of nocturnal hypoglycemia in type 1 diabetes. Diabetes Care, 2008. 31(12): p. 2271-2.
  95. Taplin, C.E., et al., Preventing post-exercise nocturnal hypoglycemia in children with type 1 diabetes. J Pediatr, 2010. 157(5): p. 784-8.
  96. Kirkman, M.S., et al., Diabetes in older adults. Diabetes Care, 2012. 35(12): p. 2650-64.
  97. Cox, D.J., H. Singh, and D. Lorber, Diabetes and driving safety: science, ethics, legality and practice. Am J Med Sci, 2013. 345(4): p. 263-5.
  98. Nielsen, L.R., et al., Hypoglycemia in pregnant women with type 1 diabetes: predictors and role of metabolic control. Diabetes Care, 2008. 31(1): p. 9-14.
  99. Inzucchi, S.E., et al., Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care, 2015. 38(1): p. 140-9.
  100. American Diabetes Association, 12. Children and Adolescents: Standards of Medical Care in Diabetes-2018. Diabetes Care, 2018. 41(Suppl 1): p. S126-S136.
  101. American Diabetes Association, 11. Older Adults: Standards of Medical Care in Diabetes-2018. Diabetes Care, 2018. 41(Suppl 1): p. S119-S125.
  102. Sämann, A., et al., Glycaemic control and severe hypoglycaemia following training in flexible, intensive insulin therapy to enable dietary freedom in people with type 1 diabetes: a prospective implementation study. Diabetologia, 2005. 48(10): p. 1965-70.
  103. Little, S.A., et al., Recovery of hypoglycemia awareness in long-standing type 1 diabetes: a multicenter 2 x 2 factorial randomized controlled trial comparing insulin pump with multiple daily injections and continuous with conventional glucose self-monitoring (HypoCOMPaSS). Diabetes Care, 2014. 37(8): p. 2114-22.
  104. Hopkins, D., et al., Improved biomedical and psychological outcomes 1 year after structured education in flexible insulin therapy for people with type 1 diabetes: the U.K. DAFNE experience. Diabetes Care, 2012. 35(8): p. 1638-42.
  105. Leelarathna, L., et al., Restoration of self-awareness of hypoglycemia in adults with long-standing type 1 diabetes: hyperinsulinemic-hypoglycemic clamp substudy results from the HypoCOMPaSS trial. Diabetes Care, 2013. 36(12): p. 4063-70.
  106. de Zoysa, N., et al., A psychoeducational program to restore hypoglycemia awareness: the DAFNE-HART pilot study. Diabetes Care, 2014. 37(3): p. 863-6.
  107. Elliott, J., et al., Substantial reductions in the number of diabetic ketoacidosis and severe hypoglycaemia episodes requiring emergency treatment lead to reduced costs after structured education in adults with Type 1 diabetes. Diabet Med, 2014. 31(7): p. 847-53.
  108. Wiethop, B.V. and P.E. Cryer, Alanine and terbutaline in treatment of hypoglycemia in IDDM. Diabetes Care, 1993. 16(8): p. 1131-6.
  109. Haymond, M.W. and B. Schreiner, Mini-dose glucagon rescue for hypoglycemia in children with type 1 diabetes. Diabetes Care, 2001. 24(4): p. 643-5.
  110. Chabenne, J., et al., A glucagon analog chemically stabilized for immediate treatment of life-threatening hypoglycemia. Mol Metab, 2014. 3(3): p. 293-300.
  111. Pontiroli, A.E., Intranasal glucagon: a promising approach for treatment of severe hypoglycemia. J Diabetes Sci Technol, 2015. 9(1): p. 38-43.
  112. Boyle, P.J., et al., Octreotide reverses hyperinsulinemia and prevents hypoglycemia induced by sulfonylurea overdoses. J Clin Endocrinol Metab, 1993. 76(3): p. 752-6.
  113. McLaughlin, S.A., C.S. Crandall, and P.E. McKinney, Octreotide: an antidote for sulfonylurea-induced hypoglycemia. Ann Emerg Med, 2000. 36(2): p. 133-8.
  114. Graveling, A., Walden, E., Flanangan, D., et. al., Hospital management of hypoglycaemia in adults with diabetes. Joint British Diabetes Societies for Inpatient Care. 2021; Available from: https://abcd.care/resource/hospital-management-hypoglycaemia-adults-diabetes.

 

Etiology and Pathogenesis of Diabetes Mellitus in Children and Adolescents

ABSTRACT

 

In this chapter, we review the etiology and pathogenesis of Type 1 diabetes mellitus (T1DM), with particular emphasis on the most common immune mediated form. Whereas Type 2 diabetes (T2DM) appears to be an increasing price paid for worldwide societal affluence, there is also evidence worldwide of a rising tide of T1DM. The increase in understanding of the pathogenesis of T1DM has made it possible to consider interventions to slow the autoimmune disease process in an attempt to delay or even prevent the onset or slow the progression of hyperglycemia. Although the prevention of T1DM is still at the stage of research trials, the trials are often mentioned in the lay press.  Current investigations will determine if antigen-based therapies can in fact abrogate ongoing autoimmunity via immuno-stimulation and ultimately prevent diabetes in humans without the risks of general immunosuppression.  We also review the etiology and pathogenesis of T2DM and monogenic forms of diabetes that may be confused with T1DM or T2DM. 

 

INTRODUCTION

 

Diabetes Mellitus (DM) is a syndrome of disturbed metabolism involving carbohydrate, protein, and fat which results from the degree of insulin deficiency (absolute or relative) and tissue sensitivity to its actions. The combination(s) of insulin deficiency and sensitivity to its actions bring about distinct clinical phenotypes with varying severity of disturbed metabolism, most conveniently monitored by the degree of hyperglycemia. Absolute insulin deficiency (Type 1 DM) occurs with autoimmune destruction of insulin secreting β-cells (Type 1A DM) and other congenital (genetic defects in the formation or function of the endocrine pancreas), or acquired (relapsing pancreatitis and pancreatectomy) conditions. Absolute deficiency of insulin action also can occur in the total absence of insulin receptors, a rare event. Relative insulin deficiency occurs with genetic or acquired defects in insulin synthesis or secretion that are inadequate to overcome the resistance caused by fewer functioning insulin receptors, or resistance to insulin action induced by stress, drugs, and most commonly obesity (Type 2 DM).The acute clinical manifestations are those related to hyperglycemia which exceeds renal threshold to result in polyuria, increased thirst, dehydration, electrolyte disturbances, weight loss, and metabolic decompensation, in extreme degree known as diabetic ketoacidosis and non-ketotic hyperosmolar coma. The chronic complications include macrovascular (CAD, CVD, amputations) and microvascular (retinopathy, nephropathy, neuropathy) lesions.  Both the acute and chronic complications are inversely related to the degree of metabolic control achieved.  These brief introductory comments form the basis for the etiology, pathogenesis, classification and diagnosis of diabetes mellitus.

 

Classification and Diagnosis of Diabetes

 

The American Diabetes Association Standards of Medical Care for Diabetes 2021(1) proposes the following classification (Table 1).

 

Table 1. Classification of Diabetes

Type 1 Diabetes owing to autoimmune destruction of insulin secreting β-cells leading to insulin deficiency

Type 2 Diabetes owing to inadequate insulin secretion that cannot overcome the existing degree of insulin resistance

Gestational diabetes (diabetes diagnosed in the second or third trimester of pregnancy that is not clearly overt diabetes)

Diabetes owing to other causes

- Monogenetic diabetes syndromes (neonatal diabetes, maturity-onset diabetes of the young [MODY])

- Disease of the exocrine pancreas (cystic fibrosis, pancreatitis, pancreatectomy)

- Medication induced (glucocorticoids, treatment of HIV/AIDS, immunosuppressants, chemotherapeutic agents)

 

Criteria for the Diagnosis of Diabetes Mellitus

 

The Expert Committee on the Diagnosis and Classification of Diabetes Mellitus recommends the following criteria for diagnosing DM (1).  Two replicate fasting glucose levels that exceed 126 mg/dl (>7 mmol/L) is consistent with diabetes even in the absence of symptoms. Normal fasting blood glucose levels of 100 mg/dl or above are considered impaired fasting glucose (IFG). Persons with IFG levels (FPG= 100-125 mg/dl (5.66.9 mmol/l) and/or with impaired glucose tolerance test (IGT) (2hour post-load glucose 140-199 mg/dl (78.8 mmol/L-11.1 mmol/L) are at risk of diabetes and should be observed periodically to detect hyperglycemic progression. Replicate, two-hour glycemic responses >200 mg/dl (>11.1 mmol/L) after a standard oral glucose tolerance test also indicate diabetes. This stage is often reached before the fasting glucose levels rise in T2DM and post-prandial hyperglycemia may precede fasting hyperglycemia by months to years. The reliance on only fasting glucose levels is generally more useful for identification of impending T1D but not for T2D.

 

The ADA now recommends that measurement of HbA1c levels can be used in clinical practice for the diagnosis of diabetes, since the onset is seldom so acute that it will not be reflected in elevated HbA1c levels Table 2 (1).

 

Table 2. The American Diabetes Association Diagnostic Guidelines (1,2)

Stage

Latent

Impaired glucose tolerance

Diabetes

Diagnostic criteria

Presence of 2 or more autoantibodies

AND

Normal glucose levels

Fasting plasma glucose: 100-125 mg/dl

OR

2hour plasma glucose during OGTT*: 140-199 mg/dl

OR

HbA1C+: 5.7-6.4%

Fasting plasma glucose: ≥126 mg/dL

OR

2hour plasma glucose during OGTT*: ≥200 mg/dl

OR

Random plasma glucose: ≥200 mg/dl with symptoms of polyuria, and weight loss.

OR

HbA1C+ ≥6.5%.

*The OGTT should be performed as described by the World Health Organization (1.75 gm/kg up to 75 gm, using a glucose load containing anhydrous glucose dissolved in water).

 

ETIOLOGIC CLASSIFICATION

 

Type 1 Diabetes Mellitus

 

Type 1 diabetes mellitus (T1DM) comprises several diseases of the pancreatic ß cells which lead to an absolute insulin deficiency. This is usually considered to be the result of an autoimmune destruction of the pancreatic ß cells (type 1A). Some patients with T1DM with no evidence of ß cell autoimmunity have underlying defects in insulin secretion often from inherited defects in pancreatic ß cell glucose sensing and from other genetic or acquired diseases.

 

Type 2 Diabetes Mellitus

 

Type 2 diabetes mellitus (T2DM) is by far the more common type of diabetes and is characterized by insulin resistance resulting from defects in the action of insulin on its target tissues (muscle, liver, and fat), but complicated by varying and usually progressive failure of beta cells’ insulin secretary capacity. Most patients with T2DM in the US and Europe are overweight or obese, however in India and China, most T2DM patients have a lean body mass index (BMI), albeit with increased visceral and hepatic fat.

 

Monogenic Diabetes

 

Monogenic forms of diabetes are characterized by impaired secretion of insulin from pancreatic β cells caused by a single gene mutation. These forms comprise a genetically heterogenous group of diabetes including, maturity onset diabetes of the young (MODY), permanent or transient neonatal diabetes, and mitochondrial diabetes. MODY is the most common form of monogenic diabetes, with autosomal dominant transmission of one of several genes encoding a primary defect in insulin secretion.

 

TYPE 1 DIABETES MELLITUS

 

Epidemiology of Type 1 Diabetes

 

T1DM is one of the most common chronic diseases of childhood and is classified as an autoimmune disease. Most common autoimmune disorders predominantly affect females, but, T1DM equally affects males and females with a slight male predominance in younger children. This and other inconsistencies have raised questions as to whether T1DM is a “pure” auto-immune disease or whether the auto-immune component is a marker of a separate primary trigger (3,4).  We discuss these issues later in this chapter. 

 

The incidence and prevalence of T1DM vary by age, season, geographic location, and within different racial and ethnic groups. Of cases diagnosed before the age of 20, however, two peaks of T1DM presentation are observed; one between 5 and 7 years of age, and the other during puberty at the mid-teens (5). However, first presentation of T1DM actually is as common in adulthood as it is in childhood and is characterized by a milder course in adults; the term LADA, (Latent, Auto-immune, Diabetes of Adults) is used to describe this entity. A seasonal variation in the incidence of T1DM is also observed; the majority of new cases of T1DM are diagnosed mostly in autumn and winter (6).  Findings from large T1DM registry studies such as the World Health Organization Multinational Project for Childhood Diabetes, known as the DIAMOND Project, EURODIAB   and others monitor incidence and other epidemiological markers.

 

The World Health Organization Multinational Project for Childhood Diabetes, known as the DIAMOND Project (in 50 countries), EURODIAB (in Europe), and SEARCH for Diabetes in Youth (in the USA) were established to address the implications of diabetes in youth and describe the incidence of T1DM. Wide variations in incidence of T1DM exist throughout the world, lowest in China and Venezuela (0.1 per 100,000 per year) and highest in Finland and Sardinia (50-60 per 100,000 per year) (7). A multicenter study focusing on identifying the prevalence and incidence of diabetes by type, age, gender, and ethnicity found a 1.8% annual increase in the prevalence of T1DM among American youth from 2002-2003 to 2011-2012, whereas T2DM had increased 4.8% annually from 2002-2003 to 2011-2012 (Table 3) (8).  The greatest increase was seen in youth of minority racial/ethnic groups (8).  Similar rates of increase in T2DM in teens are reported from the UK, India, China and Japan.

 

Table 3. Incidence of T1DM in the USA (per 100,000/year)

 

Age Group

 

0-4 yr

5-9 yr

10-14 yr

15-19 yr

Non-Hispanic White

18.6

28.1

32.9

15.1

African American

9.7

16.2

19.2

11.1

Hispanic American

9.1

15.7

17.6

12.1

American Indian

4.1

5.5

7.1

4.8

Asian and Pacific Islander American

6.1

8.0

8.3

6.8

All

14.3

22.1

25.9

33.1

 

Although, there is a wide variance in the incidence and prevalence of diabetes throughout the world, the number of youths who are being diagnosed with T1DM has been growing at an annual rate of about 3 percent (9) and a similar increased annual rate was also observed among U.S. youth (10). This rising incidence of T1DM in children across the world in a short period of time clearly cannot be explained by genetic factors. Analytical epidemiological studies suggest that environmental risk factors, operating early in life, might be contributing to the increasing trend in incidence of T1DM (11,12).

 

On the basis of estimates for the number of people with diabetes in 2014, the cost of health care of diabetes in the US is estimated to be $105 billion per annum and the direct annual cost of diabetes in the world is $825 billion (13). However studies indicate that many more diabetic adults diagnosed as having T2DM phenotype actually have T1DM  as defined by the presence of antibodies to islet cell components (14,15); the term LADA, Latent Autoimmune Diabetes of Adults, is often used to describe this group (16).

 

Natural History of Type 1 Diabetes 

 

After immune activation in the setting of genetic susceptibility, the disease progresses through pre-symptomatic stages identified by presence of autoantibodies and impaired glucose intolerance, arising from further loss of β-cell function and ultimately resulting in clinical diabetes. (Figure 1)

Figure 1. Type 1 diabetes disease progression (17)

Pancreatic ß cells secrete insulin and are found in the islets of Langerhans. These islets are specialized groups of a few hundred to a few thousand endocrine cells that are anatomically and functionally discrete from pancreatic exocrine tissue, the primary function of which is to secrete pancreatic enzymes into the duodenum. Normal subjects have about one million islets, which in total weigh only 1-2 grams and constitute less than 1% of the mass of the pancreas. Furthermore, islets are composed of various types of cells that are interconnected as a regulatory network to regulate the disposition of nutrients and their utilization for energy use and tissue growth and repair. At least 70% are ß cells localized in the core of the islets, surrounded by α-cells that secrete glucagon, δ-cells that secrete somatostatin, and PP cells that secrete pancreatic polypeptide. All the cells communicate with each other through their extracellular spaces and through gap junctions; communication is further modulated by a rich network of sympathetic and para sympathetic innervation.

 

Insulin, a peptide hormone composed of 51 amino acids is synthesized, packaged and secreted in pancreatic ß cells. Insulin is synthesized as preproinsulin in the ribosomes of rough endoplasmic reticulum. The preproinsulin is then cleaved to proinsulin that is transported to the Golgi apparatus where it is packaged into secretory granules. Most of the proinsulin is cleaved into equimolar amounts of insulin and connecting (or C)-peptide in the secretory granules. Because the C-peptide sequence differs from that of insulin, and because, unlike insulin, it is not extracted by the liver, it is possible to estimate β-cell insulin secretion by measuring C-peptide, even in the presence of insulin antibodies resulting from insulin replacement therapy that impair the ability to measure insulin directly. Similarly, because C-peptide is an index of endogenous insulin secretion, and because C-peptide is not extracted by the liver, the ratio of C-peptide: insulin should exceed 1; when it is less than 1, implying a high insulin value, exogenous insulin may have been used. This has diagnostic and forensic utility in diagnosing causes of hypoglycemia.

 

Glucose is a major regulator of insulin secretion (Figure 2). When extracellular fluid glucose concentrations rise after a meal, glucose is taken up by the ß cells via glucose transporters, GLUT2 and GLUT1. Glucose is then phosphorylated into glucose-6-phosphate by islet specific glucokinase and metabolized, thereby increasing cellular ATP concentrations. The rise in ATP raises the resting ratio of ATP:ADP, that closes ATP dependent potassium channels (K-ATP) in the β-cell membrane, resulting in accumulation of intracellular potassium, causing membrane depolarization and influx of calcium via a voltage gated calcium channel. The rise in intracellular free calcium in ß-cells promotes margination of the secretory granules, their fusion with the cell membrane, and release of cell contents which include insulin into the extracellular space. An immediately releasable pool of insulin granules adjacent to the plasma membrane is responsible for an acute (first phase) insulin response; with ongoing stimulation, a pool of granules in the interior of the cell is mobilized and released as the “second phase” response. Amino acids also stimulate insulin release by a similar mechanism that involves the enzyme glutamate dehydrogenase which enables metabolism and ATP production by certain amino acids. Defects in the genes regulating these processes may result in diabetes if the K-ATP channel is prevented from closing normally (activating mutations) or syndromes of hyperinsulinemic hypoglycemia if the K-ATP channel is prevented from opening (inactivating mutations).  These aspects are discussed in greater detail in the section on Monogenic forms of diabetes (see below).

Figure 2. Insulin secretion by Pancreatic β cells. In the stimulated state, glucose is transported into the β cell by the GLUT2 transporter which undergoes phosphorylation by glucokinase and glucose is then metabolized. This results in an increase in the ATP/ADP ratio and initiation of a cascade of events that is characterized by closure of the K-ATP channel, decreased flux of potassium across the membrane, membrane depolarization, and calcium influx. This cascade ultimately results in insulin release from storage granules. The K-ATP channel shown is composed of four small subunits, Kir6.2, that surround a central pore and four larger regulatory subunits constituting SUR1. In the resting state, the potassium channel is open, modulated by the ratio of ATP to ADP. Leucine also stimulates insulin secretion by allosterically activating GDH and by increasing the oxidation of glutamate; this then increases the ATPADP ratio leading to the cascade of events beginning with closure of the KATP channel.
MCT-1: Monocarboxylate transporter-1, SCHAD: Short chain 3-hydroxyacyl-CoA dehydrogenase, SUR1: Sulfonylurea receptor 1, Kir 6.2: Potassium Inward Rectifying Channel 6.2, UCP-2: Uncoupling protein 2, HNF4α: Hepatocyte Nuclear Factor 4α, HNF1 α: Hepatocyte Nuclear Factor 4α, K+: Potassium, ATP: Adenosine Triphosphate, GDH: Glutamate Dehydrogenase, GLUT-2: Glucose Transporter 2

Metabolic Derangements of Type 1 Diabetes

 

As the pancreatic ß cell mass declines in an islet cell antibody (ICA) positive person, the first metabolic abnormality discernable is a decline in the first phase of insulin release (FPIR) to an IVGTT (18). The insulin level after a 3-4 minute infusion of glucose at 0.5Gms/kg rises abruptly in normal children at about 8 years of age, perhaps coincident with the onset of adrenarche (19). In the relatives and children from the general population with positive ICA, a decline in the FPIR is a strong predictive marker of evolving diabetes (19-21).

 

Subsequently, in evolving T1DM there is a rise in the fasting glucose level followed by an inability to keep the two-hour, post-OGTT glucose level below 200mg/dl (11.1mM). Transient insulin resistance also occurs in untreated T1DM and is due to raised levels of free fatty acids (FFAs) from uncontrolled lipolysis (22), as well as decreased levels of hepatic glucokinase and insulin regulated GLUT 4 glucose transporters in adipocytes which contribute to  the onset of symptomatic diabetes (23-25). Prolonged hyperglycemia itself likely impairs the ability to secrete insulin and when insulin replacement therapy begins, there is usually some recovery in the patient's ability to secrete insulin (the "honeymoon" period). However, within months to years, this partial recovery in endogenous insulin secretion ultimately fails. If it does not fail after 2 years, another form of diabetes, such as MODY should be suspected. Initially, the glucagon secreting cells within the pancreatic islets remain relatively preserved, resulting in excessive secretion of glucagon relative to insulin after protein meals (26). These elevated glucagon levels exacerbate the effects of the insulin deficiency, and promote lipolysis and ketogenesis, effects that can be partially reversed by an infusion of somatostatin (27). As the mass of islet cells decline, there is also loss of amylin, an islet cell hormone that down-regulates glucagon secretion. Thus, an analogue of amylin (pramlintide- marketed under the trade name Symlin) can be used as adjunctive therapy with insulin replacement. In time, with continued loss of islets, glucagon deficiency develops in established long standing T1DM, rendering patients more susceptible to insulin-induced hypoglycemia (26,28).  

 

Insulin is the hormone of "feasting", promoting utilization and deposition of ingested nutrients into body stores, as well as having multiple anabolic effects in many tissues. Progressive insulin deficiency thus induces a starvation like state, associated with excessive hepatic and renal gluconeogenesis, decreased peripheral utilization of glucose, hyperglycemia with resultant glycosuria, loss of water and sodium salts, and proteolysis in muscle liberating amino acids such as alanine and glutamine as substrates for gluconeogenesis (29-31). Uncontrolled lipolysis leads to the rapid mobilization of fatty acids from adipose tissue and the increased delivery of fatty acids to the liver leading to the increased synthesis of triglycerides and secretion of very low-density lipoprotein (VLDL).

 

With severe insulin deficiency the fatty acids delivered to the liver are metabolized to yield beta hydroxybutyric and aceto-acetic acids (ketone bodies) and contribute to keto-acidosis. Ketoacidosis is a life-threatening metabolic decompensation that is characterized by hyperglycemia, dehydration, metabolic acidosis and ketosis, all the result of the effects of severe insulin deficiency as well as the counter-regulatory stress hormones, cortisol, growth hormone, catecholamines and glucagon. Specifically, hepatic glucokinase levels fall with insulinopenia, synthesis of hepatic triglyceride and glycogen levels decline, malonyl CoA falls and thereby carnitine palmitoyl transferase-I levels rise promoting the transport of fatty acyl-CoA into mitochondria with the formation of acetyl-CoA (32-34).  In the liver, acetyl-CoA is converted into ß-hydroxybutyrate and acetoacetate in a proportion that depends upon the prevailing redox state, which provide an additional fuel substrates for muscle and brain (31,35,36). Lipoprotein lipases are also inactivated, leading to reduced hydrolysis of triglycerides that, if severe, may turn the serum milky with increased VLDL characteristic of the type 4 lipemic phenotype (37-39).

 

Genetic Susceptibility to Type 1 Diabetes

 

Individuals with autoimmune T1DM have inherited a number of quantitative trait loci (QTL) that encode protective and predisposing alleles which have exceeded the net genetic threshold required to predispose them to the disease (40). However, this genetic threshold (penetrance) is dependent in turn on chance interactions with greater predisposing than protective environmental forces. The multiple genetic influences in T1DM comprise a major effect from DR/DQ genotypes of the HLA complex (some 50% of the genetic effect), coupled to several other QTLs with minor influences (Table 4). All of the latter QTLs are not obligatory genetic elements themselves since they are of minor-influence, but they collectively interact to create additive influences on the genetic threshold. Siblings of a diabetic patient develop T1DM at about 15-fold greater frequency than persons in the general population (prevalence 1:250-300), vs. a value of 15. The HLA predisposition to T1DM is encoded by cis- and trans complementation DQA1*/DQB1* heterodimers which have an arginine at residue 52 of the A chain and a neutral amino acid (DQB1*0302, *0201) rather than a charged aspartic acid at residue 57 of the B chain (DQB1*0602/3 and DQB1*0301) (40), as modified by DRB1*04 subtypes (*0401 and *0405 are susceptible and *0403 and 6 are resistant types) (41) in the HLA genotype. Further, HLA-DP alleles have also been implicated, even though they are at a considerable recombination frequency away from the closely linked DR/DQ loci (42). Other genes involved include the variable number of tandem repeat (VNTR) alleles 5' to the insulin (INS) gene on chromosome 11p15, where the protective class III alleles (>200 repeats) are associated with increased expression of insulin in the thymus, leading to a more efficient eradication of insulin autoreactive T cells than class I alleles (26-63 repeats) that confer susceptibility to develop diabetes (43,44). There are also CTLA-4 gene polymorphisms on chromosome 2q that are associated with T1DM. CTLA-4 is an induced accessory molecule that is expressed on activated T cells. CTLA-4 interacts with B7.2 expressed by antigen presenting cells (APC), signaling apoptosis of T cells that become activated as part of an immune response, thereby confining the immune response. The non-obese diabetic (NOD) mouse, a model for autoimmune diabetes, has an enlarged lymphoid mass because of resistance of their T cells to undergo apoptosis, as do CTLA-4 knockout mice, which readily develop lymphocytic organ infiltrates like NOD mice. These genes thus collectively affect the general ability to be tolerant to "self" antigens. Another susceptibility locus, (the IDDM 4) in the genomic interval on chromosome 11q13harbors the high affinity IgE Fc receptor gene that has been linked to atopy and asthma, which are characterized byTh2 responses that may protect individuals against the development of anti- islet Th1 responses, and thereby protect against T1DM. There are other genomic intervals associated with or linked to T1DM that have been putatively mapped, but these mostly lack plausible candidate genes in the DNA region, and pathogenic mechanisms for them cannot yet be offered. The NOD mouse however has been subjected to extensive genetic mapping studies, in the hopes that genomic intervals harboring susceptibility or protective genes which are syntenic to humans will be discovered, thus hastening the identification of equivalent defective genes.

 

Table 4. Genotypes of the HLA Complex Associated with Diabetes Mellitus

Locus

Chromosome

Candidate Genes/Microsatellites

References

IDDM1

6p21.3*

HLA-DQ/DR

(45,46)

IDDM2

11p15*

INS VNTR

(47,48)

IDDM3

15q26

D15s107

(49)

IDDM4

11q13

MDU1, ZFM1, RT6, FADD/MORT1, LRP5

(50,51)

IDDM5

6q24-27

ESR, MnSOD

(52)

IDDM6

18q12-q21

D18s487, D18s64, JK (Kidd locus)

(53)

IDDM7

2q31

D2s152, IL-1, NEUROD, GALNT3

(54)

IDDM8

6q25-27

D6s264, D6s446, D6s281

(52)

IDDM9

3q21-25

D3s1303

(55)

IDDM10

10p11-q11

D10s193, D10s208, D10s588

(56)

IDDM11

14q24.3-q31

D14s67

(57)

IDDM12

2q33*

CTLA-4, CD28

(58)

IDDM13

2q34

D2s137, D2s164, IGFBP2, IGFBP5

(59)

IDDM14

?

NCBI# 3413

 

IDDM15

6q21

D6s283, D6s434, D6s1580

(52)

IDDM16

?

NCBI# 3415

 

IDDM17

10q25

D10s1750- D10s1773

(60)

2p12

EIF2AK3

 

(61)

5p11-q13

 

 

(62)

16p

 

D16s405- D16s207

(62)

16q22-q24

 

D16s515- D16s520

(55)

1q42

 

D1s1617

(63)

Xp11

 

DXS1068

(64)

 

In summary, T1DM is a complex, multifactorial disease involving genetic predisposition and an environmental triggering event, of which viral causes have been proposed. Although more than 50 loci have been identified, genes involved in immune regulation including HLA subtypes, VNTR in insulin itself, CTLA4, PTPN22, AIRE, and IL2R remain most prominent (65,66). The HLA association, especially class II, remains the strongest predictor of T1DM risk. The heterozygous DR3/DR4 genotype carries the highest genetic risk for T1DM in non-Hispanic whites (45-70).  In conclusion, insulin expressing islets from recent-onset T1D subjects show overexpression of interferon stimulated genes (ISGs), with an expression pattern similar to that seen in islets infected with virus or exposed to IFN-γ/interleukin-1β or IFN-α.

 

Autoantigens and Autoantibodies in Type 1 Diabetes

 

The Doniach group in London, first reported islet cell autoantibodies in patients with autoimmune polyglandular syndromes (APSs) (71), especially in those with APS type-1 (APS-1) (72), even though such patients did not often develop diabetes. Lendrum and colleagues, having failed to find serological evidence for an autoimmune basis for chronic pancreatitis, did succeed in finding Islet Cell Antibodies (ICA) detectable by indirect immunofluorescence in patients with T1DM. Islet cell surface reactive autoantibodies and autoreactive peripheral blood T cells were also reported (73,74). Over the years that followed, the presence of ICA in US patients was confirmed but with distinctly lower frequencies of ICA among African American diabetic patients (75). Insulin autoantibodies (IAA) were discovered in patients with T1DM before their first dose of insulin replacement had been received (76). The presence of IAA together with ICA identified a group of non-diabetic relatives of probands with T1DM, that were at high risk for T1DM themselves (77). Insulin itself is not an ICA antigen that can be detected by the indirect immunofluorescent technique. Subsequently, much of the antigenic nature of the ICA reactivity has become clearer. It was recognized that many patients with "stiff" man syndrome who were prone to develop diabetes, also had ICA and autoantibodies to glutamic acid decarboxylase (GAD65). These GAD autoantibodies penetrated the blood brain barrier. High concentrations of GAD in the cerebellum reduce brain levels of the inhibitory neurotransmitter gamma aminobutyric acid (GABA), thereby causing the appearance of temporal lobe epilepsy, depressed cognition, muscle spasms, cerebellar incoordination and motor dysfunctions. That GAD65 was the antigen that accounted for the 64 KDa islet cell protein previously discovered by Baekkeskov to react with autoantibodies in T1DM, was later confirmed by the same investigator (78). Antibodies to recombinant GAD65 and GAD67 in T1DM patients were soon reported (79). The autoantibodies reacted to the antigens by conformational rather than linear epitopes, and thus with native rather than denatured antigens. Therefore, they were best detected by liquid phase assays such as radioimmunoassay, rather than by an ELISA technique. In stiff-man syndrome, the predominant GAD autoantibodies reacted with linear epitopes. It became known that besides islet cell 64 KDa sized proteins, autoantibodies in the sera of T1DM patients also precipitated islet cell proteins of 50, 40 and 37 KDa as well (80).

 

The next islet cell antigen discovered was one of the two-dozen tyrosine phosphatases expressed in islet cells, insulinoma antigen-2 (IA-2) (81). This antigen shared structural homologies with the ICA-512 antigen (82). A second tyrosine phosphatase named IA-2ß was discovered next (83). These additional tyrosine phosphatase antigens allowed for the matching of the islet cell proteins previously identifiable only by their molecular weights. Thus, GAD65 and its tryptic fragment explained the 64 and 50 KDa proteins, while tryptic fragments of IA-2 and IA- 2ß were identical with the 40 KDa and the 37 KDa islet precipitable proteins respectively (84). The tyrosine phosphatases are a family of transmembrane enzymes of which only these two are expressed by the pancreatic islets and react with T1DM autoantibodies. The reactivity is almost exclusively with the internal domains of these molecules, suggesting that they arise as a consequence of islet cell damage from autoimmunity. Antibodies to IA-2 cross-react with those of IA-2ß in about 50% of the patient sera. Some unusual patient sera however react exclusively with IA-2ß. The question of why only these two members of the tyrosine phosphatase family are targets of islet cell autoimmunity has been answered by the finding that they are relatively resistant to proteolytic enzymatic digestion, and once released from islet cells after their lysis, are insoluble and thus become better antigens for auto-immunization, than those that remain soluble and are more rapidly digested (85).

 

Recently, another antigen of 38KDa size (GLIMA) was added to the islet cell group, albeit only a minority of patient's sera reacts to it (86). Still more islet cell autoantigens are likely to be discovered. The detection of islet cell autoantibodies is useful for differentiating T1DM from diabetes of other causes, and can be used to predict onset of diabetes months to years before onset of the clinical disease (20,21,87,88) in non-diabetic relatives of probands with T1DM.  Importantly, the clinical onset of the disease is often long preceded by the appearance of autoantibodies reactive to islet cells (ICA) (88) and to insulin (77), as independent age-related variables in predicting a diabetic outcome (89). Islet cell autoantibodies (ICA) also show a strong tendency to disappear after diabetes onset when all ß cells are destroyed (90,91).

 

Studies in mice demonstrated a critical role of autoantibodies to GAD65 in the induction of autoimmune diabetes in NOD mice. In humans, the German BABY-DIAB study and the Finnish TRIGR study showed that islet autoantibodies which are mostly IgG class can be transferred through the placenta from islet antibody-positive mothers to their offspring (92,93). Most of the antibodies, however, disappeared from the circulation of the infant within the first year of life, indicating that they represent maternal antibodies and unlikely that they are markers of fetal induction of B-cell autoimmunity (93). In the German BABY-DIAB study, it was demonstrated that 729 offspring of mothers with T1DM had significantly lower risk of developing multiple islet autoantibodies (5 year risk 1.3%) and diabetes (8-year risk 1.1%) when they were GAD or IA-2 positive, than offspring who were islet autoantibody negative at birth (94). These findings suggest that fetal exposure to islet autoantibodies may protect from future diabetes. Furthermore, the German BABY-DIAB study finding is consistent with the overall decreased risk of development of diabetes in offspring of mother with T1DM compared with that of offspring of fathers with T1DM and nondiabetic mothers (95).

 

The timing of the appearance of the autoantibodies seems to be important. It was found that progression to multiple islet autoantibodies was fastest in children who were antibody positive by age 2 years and that progression to diabetes was inversely related to the age of first positivity for multiple autoantibodies (96).

 

The presence of multiple autoantibodies strikingly increases the risk of diabetes, whereas one of the above autoantibodies in the absence of all of the others when tested for, denotes only a modestly increased risk (20,21). This suggests that antigenic epitope spreading is involved in a sustained or accelerated autoimmune attack (72) (97). Besides autoimmunity to islet cell autoantigens, patients with T1DM are subject to other autoimmunities. Thus T1DM is a component part of the autoimmune polyglandular syndromes, commonly in APS-2  (Diabetes Mellitus, Addison Disease, Hypothyroidism) and with less frequency in APS-1(AIRE gene mutations) (72). Accordingly, patients with T1DM have high rates of thyroid autoimmunity, especially if they are females (98) (99), and are at increased risk for Addison's disease (99), atrophic gastritis (100), pernicious anemia (98), celiac disease (101), and vitiligo (102).

 

Table 5. Autoantibody Targets in Type 1 Diabetes

glutamic acid decarboxylase 65

Islet cells

Insulin

Zinc Transporter 8

 

Antigen Specific Cellular Immunity in Type1 Diabetes

 

Autoreactive T cells that develop in impending T1DM, localize to the pancreatic islets where they become a component part of the evolving insulitis lesions. Thus, circulating autoreactive T cells are relatively sparse in impending T1DM. Nevertheless, antigen specific T cells are identifiable through prolonged in-vitro cultures in the presence of purified or recombinant islet cell autoantigens such as GAD (103) (104) and IA-2 (105). In fact, autoreactivity to a large number of autoantigens have been reported in both human and murine diabetes (106). T cell proliferative responses to insulin and GAD65, and more generally to islet extracts, have been repeatedly reported in both patients with T1DM (107,108) and NOD mice. However, both in humans and NOD mice, reports of spontaneous proliferative responses have been difficult to reproduce and validate, probably because of the relative paucity of autoreactive T cells in peripheral blood samples, and the ready contamination of recombinant "test" antigens by lymphotoxin or lipopolysaccharide (LPS), that by itself, can produce proliferative responses even when present in trace amounts. Furthermore, significant T cell responses to insulin, proinsulin or GAD65 antigen were reported, in some normal controls as well as in T1DM patients (109-111). Numerous laboratories have reported T cell reactivity in diabetic patients against GAD65 and IA-2 and their peptides with variable results (105,107,112-117). However, in established diabetes, the loss of the majority of ß cell mass resulting in associated loss of GAD65 and other ß cell antigens, in turn leads to the inactivation of T cells due to the loss of the peptide antigens that were driving the response. Thus, antigenic/epitopic spreading is an undesirable phenomenon associated with progression in autoimmune diseases like T1DM to a clinically significant outcome.

 

Pathogenesis of Type 1 Diabetes

 

The availability of Biobreeding (BB) rats and nonobese diabetic (NOD) mice, the rodent models of T1DM, has greatly enhanced our understanding of the possible pathogenic mechanisms involved (Fig. 3). Recently, it has become possible to compare these findings with findings in human islets, obtained from post mortem specimens of the pancreas through the network of Pancreatic Organ Donors (nPOD) and from patients with recent onset DM via endoscopic pancreatic biopsy (DiViD study, Norway) (86,118,119). In addition, epidemiological studies aimed at the prediction and prevention of T1DM permit a picture of the natural history to emerge. The process of destruction of β-cells is chronic in nature, often beginning during infancy and continuing over the many months or years that follow. At the time of clinical diagnosis of T1DM, about +80% of the β- cells have been destroyed, the islets are infiltrated with chronic inflammatory mononuclear cells (insulitis), including CD8+ cytotoxic T cells. Once islet cell autoimmunity has begun, progression to islet cell destruction is quite variable, with some patients rapidly progressing to clinical diabetes, while others remain in a non-progressive state.

Figure 3. The pathogenesis of islet cell destruction. Islet cell proteins are presented by antigen presenting cells (APCs) to naïve Th0 type CD4+ T cells in association with MHC class II molecules. Interleukin (IL)-12 is thus secreted by APCs that promotes the differentiation of Th0 cells to Th1 type cells. Th1 cells secrete IL-2 and IFN-γ that further stimulate CD8+ cytotoxic T cells or macrophages to release free radicals (super-oxides) or perforin/granzymes, leading to ß cell apoptosis or death. CD8+ cytotoxic T cells further mediate ß cell death by Fas mediated mechanisms. Interleukin (IL)-4, on the other hand, secreted mainly by natural killer T (NKT) cells drives Th0 cell to Th2 pathway leading to benign insulitis.

Diabetes risk and time to diabetes in relatives of patients directly correlates with the number of different autoantibodies present. The pathogenesis of T1DM has been extensively studied, but the exact mechanism involved in the initiation and progression of β-cell destruction is still unclear. The presentation of beta cell-specific autoantigens by antigen- presenting cells (APC) [macrophages or dendritic cells (DC)] to CD4+ helper T cells in association with MHC class II molecules is considered to be the first step in the initiation of the disease process. Macrophages secrete interleukin (IL)-12, stimulating CD4 + T cells to secrete interferon (IFN)-γ and IL-2. IFN-γ stimulates other resting macrophages to release other cytokines such as IL-1β, tumor necrosis factor (TNF-α) and free radicals, which are toxic to pancreatic β-cells. During this process, cytokines induce the migration of β-cell autoantigen specific CD8+ cytotoxic T cells. On recognizing specific autoantigen on ß cells in association with class I molecules, these CD8+ cytotoxic T cells cause ß cell damage by releasing perforin and granzyme and by Fas-mediated apoptosis of the beta cells. Continued destruction of beta cells eventually results in the clinical onset of diabetes.

 

Recently, these concepts derived from studies in the rodent models have been challenged as having the same pathologic process that occur in humans. Analysis of variations in histopathology observed from these organ donors provide mechanistic differences related to etiological agents and serve an important function in terms of identifying the heterogeneity of T1D (120). The findings are not always consistent with those of the rodent models. For example, the dense infiltration of islets by T-cells is evident in the pancreas of those who succumb to DKA at onset, but more chronic cases show a patchy distribution of destroyed and functioning islets containing beta cells with insulin suggesting a defect in secretion rather than synthesis. In the DiViD (Diabetes Virus Detection) study, expression of inflammatory markers, predominance of Class I antigens (rather than expression of Class 2 antigens) in islets, and actual viral isolations suggest a more acute process. Taken together, the studies suggest that T1DM may be a heterogeneous group of conditions in which auto-immunity may be a consequence or companion rather than the initiating mechanism. These findings begin to explain why prediction of developing T1DM in those from affected families considered at risk has become quite accurate, whereas prevention or reversal of DM by immune intervention or modulation has failed repeatedly (3,4,121).

 

The Indian uctive Event in Type 1 Diabetes

 

Various mechanisms have been proposed:

 

MOLECULAR MIMCRY

 

In antigenic molecular mimicry, cross-reactive immune responses occur due to significant structural homologies shared by molecules encoded by dissimilar genes.

 

The incidence of T1DM has increased over the last three to four decades in Europe, and the clinical disease exhibits preferential seasonal onset (122). These observations emphasize the role of environmental factors in the disease process. It has long been suggested that T1DM in humans is caused by viral infections (123-125). However, despite a vast increase in the information regarding the various genetic factors controlling the disease, little is known about the role of the putative environmental factors that might provide a more direct approach to therapy (8). Specifically, allegations that childhood vaccines could be causal have not been upheld by more extensive controlled studies.

 

The disease pathogenesis may involve multiple factors including the genetics of the host, strain of the virus, activation status of the autoreactive T cells, upregulation of pancreatic MHC class I antigens, molecular mimicry between viral and ß cell epitopes and direct islet cell destruction by viral cytolysis. Viruses, as one of the environmental factors affecting the induction of T1DM, may act as triggering agents of autoimmunity or as primary injurious agents, which directly damage pancreatic ß cells. Immune responses against a determinant shared by host cells and a virus could cause a tissue-specific immune response by generation of cytotoxic cross-reactive effector lymphocytes or antibodies that recognize self-proteins located on the target cells.

Monoclonal antibodies against viruses have been observed to be capable of cross-reacting with host determinants (126).

 

Several studies in humans also point to viruses as triggers of the disease (127). Coxsackie B4 virus and rubella virus have been linked with T1DM. In a few instances, Coxsackie B4 virus has even been directly isolated from pancreatic tissues of individuals with acute T1DM. Inoculation of this virus into mice, in one report, produced diabetes (128). The possibility that viruses might cause some cases of T1DM by infecting and destroying pancreatic ß-cells has received considerable attention. However, it is difficult to demonstrate in-vivo that viruses replicate in human ß-cells and/or produce diabetes in man. An in-vitro system was therefore developed to determine whether viruses are capable of destroying human β-cells in culture (129,130). By this method, it was clearly shown that several common human viruses, including mumps virus (131), Coxsackie B3 virus(132), Coxsackie B4 virus (128), reovirus type 3 (133), could infect human ß-cells. In addition, by radioimmunoassay, it was shown that the infection markedly decreased the insulin content of the ß-cells.

 

A strong correlation was found between the CMV genome in the immunocytes and the islet cell autoantibodies in the sera from diabetic patients (134). About 15% of newly diagnosed autoimmune T1DM patients have been reported to have persistent CMV infections.

Furthermore, it has been proposed that a molecular mimicry between protein 2C (p2C) of Coxsackie virus B4 and the autoantigen GAD65 may play a role in pathogenesis of T1DM. Kaufman et al (135) and Vreugdenhil et al (125), showed that the amino acid sequence of p2C shares a striking homology with a sequence in GAD65 (PEVKEK) and is highly conserved in Coxsackie virus B4 isolates as well as in different viruses of the subgroup of Coxsackie B-like viruses. These are the most prevalent enteroviruses and therefore the exposure to the mimicry motif should be a frequent event throughout the life. Furthermore, they suggested that molecular mimicry might be limited to the HLA-DR3 subpopulation of the T1D patients.

 

Although numerous sequence similarities between viral proteins and ß-cell autoantigens are plausible, the relationship between Coxsackie virus infection and GAD65 autoimmunity has received the most attention.

 

Glutamate Decarboxylase (GAD)

 

The finding by Kauffman et al (135), of a striking sequence homology of 18 amino acid peptide between human GAD65 and the Coxsackie virus p2-C protein, enhanced the evidence of a specific molecular mimicry model involving GAD. In addition, this specific region of GAD65 contains a T cell epitope involved in the GAD cellular autoimmunity in humans with immune mediated diseases (103)  and this region is an early target of the cellular immunity in NOD mice (136,137). GAD catalyzes the formation of the inhibitory neurotransmitter γ-amino butyric acid (GABA) from glutamine (104). Two forms of GAD exist (GAD65 and GAD67). GAD65 is the predominant form within the human pancreatic islet cells, while GAD67 predominates in mouse islets. Within the islets, GAD is predominantly observed within the ß-cells, while its roles in the inhibition of somatostatin and glucagon secretion and in the regulation of proinsulin synthesis and insulin secretion, have also been suggested (138).

 

Another study further supports a link between Coxsackie virus and T1DM, associating IgM antibodies to Coxsackie B virus as a marker of recent exposure to the virus in newly diagnosed IMD patients and age/sex-matched controls (139). In that report, humoral immunity to Coxsackie virus and GAD appeared to cluster, even in people without diabetes. A series of overlapping synthetic GAD65 peptides were used to study the most reactive T cell determinants in individuals at increased risk for T1DM, i.e., autoantibody positive, first degree relatives of T1DM patients. Elevated in vitro T cell responses were observed to GAD65 peptides (amino acids 247-266 and 260-279) in newly diagnosed T1DM patients and autoantibody positive at- risk individuals (140). The sequence of this region of GAD65 (amino acids 250-273) is significantly similar to the p2-C protein of Coxsackie B virus (123). However, not all published reports have demonstrated a linkage between immunity to GAD and Coxsackie virus. For example, one study identified a non-Coxsackie-homologous region of GAD65 as a predominant cellular immune epitope while studying the polyclonal human T cell responses (115).

 

Insulinoma Antigen Two (IA-2)

 

Tyrosine phosphatase IA-2 is another molecular target of pancreatic islet autoimmunity in T1DM. In one recent study, the epitope spanning 805-820 amino acid elicited maximum T-cell responses in all at-risk relatives, out of a total of 68 overlapping, synthetic peptides encompassing the intracytoplasmic domain of IA-2 (141). This epitope was found to have 56% identity and 100% similarity over 9 amino acids with a sequence in VP7, a major immunogenic protein of human rotavirus. This dominant epitope also has 75-45% identity and 88-64% similarity over 8-14 amino acids to sequences in Dengue, cytomegalovirus, measles, hepatitis C and canine distemper viruses and the bacterium Haemophilus influenzae.

 

Furthermore, three other IA-2 epitope peptides have 71-100% similarity over 7-12 amino acid stretch to herpes, rhino-, hanta- and flavi-viruses. Two others have 80-82% similarity with dietary proteins of milk, wheat and bean proteins. These molecular mimicries could lead to triggering or exacerbation of ß-cell autoimmunity.

 

SUPERANTIGENS

 

Besides molecular mimicry, retroviral expression of superantigens (Sags) may be able to activate clonal expansion of autoreactive T cell clones. Superantigens have been implicated in the pathogenesis of the various autoimmune diseases (142,143). Originally described as minor-lymphocyte stimulating antigens, retroviral Sags expressed by B cells interact with the development of T helper cells of both Th1 and Th2 subtypes in mice. A study in patients with T1DM demonstrated that two thirds of IAA positive sera also reacted with p73 (144). Conrad et al (145)  isolated a novel mouse mammary tumor virus-related human endogenous retrovirus (HERV), in patients suffering from acute onset T1DM. He termed them the HERV IDDMK1,2 22 subtype. They further showed that the N-terminal moiety of the envelope (env) gene encoded an MHC class II-dependent superantigen. He proposed that expression of this Sag, induced extra-pancreatically and by professional antigen-presenting cells, could lead to ß-cell destruction via the systemic activation of autoreactive T cells. He further reported the selective expansion of Vß7+ T cells in the islet cell infiltrates from two patients with recent onset IMD was associated with extensive junctional diversity of Vß7+ T cell clones. These investigators demonstrated that islet cell membrane preparations preferentially expanded Vß7+ T cells from non-diabetic peripheral blood mononuclear cells (146). However, other investigators were unable to confirm T1DM specificity of the IDDMK1,2 22, since it was equally recoverable as viremia from controls as well as patients (147). Furthermore, both patients and controls made antibodies to env proteins.

 

In order to establish molecular mimicry as a mechanism responsible for the autoimmune diseases it is important to identify the precise epitope that initiates the putative cross-reactive immune response. Additional complexity that has come to various animal studies is that of

epitope spreading (148). An increasing array of autoantigens or autoantigenic peptides reactive with autoantibodies develop over time. Both intramolecular and intermolecular epitope spreading has been described in NOD mice (136,149). These studies demonstrated that T- cell responses in NOD mice expand in vivo against a defined group of islet cell antigens in an orderly sequential manner. These responses in the young NOD mice first show a strong reactivity to GAD enzyme and not to other islet cell antigens. Furthermore, the initial response to GAD is first limited to one region of the protein only. Gradually, this response spreads intramolecularly to involve other regions of the protein. Eventually, after the destructive islet cell inflammation (insulitis) as a result of autoimmunity to ß-cells, the T-cell responses spread intermolecularly to involve other islet cell proteins (e.g., heat shock protein 60, carboxypeptidase H and insulin) as well (150). This epitope spreading makes it difficult to predict which putative cross-reactions, if any, are important in terms of disease induction, and which do not give rise to autoimmune pathology, particularly in humans who are exposed to many infections.

 

Deficiencies in immunoregulation in Type 1 Diabetes

 

There is both evidence for and speculation about defective central and peripheral mechanisms of immunoregulation in the autoimmune form of T1DM. Deletion of autoreactive T cells in the thymus, is one mechanism for the induction of tolerance to self-antigens (central deletion). This may involve diminished expression of insulin in the thymus of susceptible individuals due to the presence of class I VNTR alleles 5' to the insulin gene as already discussed. Others have suggested that it is the ineffective antigenic binding of the T1DM-prone HLA-DQ or -DR that promotes islet cell autoimmunity, since this permits autoreactive T cells to escape thymic ablation and pass into circulation.

 

In addition to clonal T cell deletion and anergy in thymus, peripheral regulatory T (Treg) cells are essential for the down regulation of T cell responses to both foreign and self-antigens, and for the prevention of autoimmunity. Various studies have identified defects in the peripheral Treg cells in T1DM patients (151,152) as well as in NOD mice affecting both NKT cells (153,154) as well as CD4+CD25+ suppressor T cells (155). Since these Treg cells are not absent in either species, ways to stimulate them should be actively sought to provide novel therapies for the future. The possibility of future therapeutic use of Treg cells in human autoimmune diseases lies heavily on basic studies that are designed to elucidate the mechanisms of induction and function of these cells. Therapy with immunomodulatory compounds that specifically target endogenous pools of Treg cells can be envisioned (156). This approach requires a more detailed investigation into the intracellular and extracellular events that regulate the differentiation and expansion of these cells in-vivo.

 

Of great interest has been the emergence of immune mediated T1DM in patients treated with checkpoint inhibitors for various cancers (157).  Unlocking the immune response via drugs that block the molecules programmed death (PD1) or its ligand, PDL1, as well as CTL4, may result in immunotoxicity with emergence of autoimmunity affecting various organs, including endocrine tissues such as the thyroid, adrenal and pancreas causing a form of T1DM (158). Indeed, autoimmunity has been called the “Achilles’ Heel” of immunotherapy, with increasing reports of its association with T1DM (159).

 

Environmental Factors in Type 1 Diabetes

 

Besides the familial predispositions, much evidence points to a major role of environmental factors in the disease pathogenesis. More than 60% of identical twins affected by T1DM are discordant for the disease and most of the non-diabetic twins lack islet cell autoantibodies. Over the past 3 decades, the disease frequency is on a steep rise in Western countries that cannot be explained by the accumulation of the susceptible genes. Africans, who dominate the tropics, and Chinese, both have low frequencies of the susceptible genes and low incidence rates of T1DM (75), except where there has been a high rate of Caucasian genetic admixture.

 

More persuasively, migrants from countries with low hygiene and low incidence rates of T1DM to countries with high hygiene and high incidence become as susceptible as the natives within a generation (160). Animals reared in sterile environments have early onsets and increased frequencies of diabetes while those infected with a variety of micro-organisms and parasites become protected (161-165). The hygiene hypothesis was proposed.  A strong causal relationship between prevailing level of community hygiene, especially with respect to drinking water and the dramatic increase in the incidence of autoimmune diseases such as T1DM in the modern world, has been referred to as the hygiene hypothesis.

 

ROLE OF DIET

 

Despite persuasive epidemiological evidence for environmental factors that precipitate T1DM in genetically susceptible individuals, their identity remains elusive. This may be due to long period between exposure and the onset of hyperglycemia, the complex genetics of the disease, and the likely multiple insults of perhaps different derivation involved in the initiation of the insulitis and subsequent ß cell destruction. Dietary habits such as consumption of dairy products and early weaning of infants, and dietary toxins such as nitrates and nitrites have been associated with this autoimmune disease (166,167).

 

Close correlations between per capita consumption of unfermented milk proteins and the incidence of diabetes between countries(168-170) and within a country have been reported (171). The claimed negative association between diabetes incidence and a high frequency and long duration of breast-feeding is more controversial (166) and has not been confirmed by reports from Germany (172) and the United States. Several studies have found associations between the consumption of foods rich in nitrates (or nitrites), which is reduced to nitrite in the gut, and the occurrence of T1DM (173,174). The active species is believed to be N-Nitroso compounds that can be formed from the reaction of nitrite with amines (175). Most recently, the gut microbiome and its modulation by dietary factors, has been implicated in the causality of T1DM (176).

 

The incidence of T1DM varies worldwide according to dietary patterns. In-depth exploration of dietary risk factors during pregnancy and early neonatal life is warranted to confirm whether and to what extent diet cooperates with genetic susceptibility in the early onset of T1DM.

 

Screening Methods for Type 1 Diabetes

 

T1DM is by far the most common chronic metabolic disease of childhood and adolescence and its prevalence and incidence has been increasing worldwide (96). This increase of incidence is the highest among the children under 5 years of age (177). Prevention of T1DM would constitute a major advance in the lives of pre-diabetic individuals and significantly relieve a major current and predicted burden on both the individual and the health care system. Identifying individuals at risk developing the disease and the prevention of the disease progression are two important steps before the onset of disease. The presence of islet autoantibodies, as well as the genetic predisposition with specific HLA haplotypes are known risk factors associated with the development of diabetes. Most studies have been carried out on first-degree relatives of T1DM patients who have 15-fold increased risk of the developing diabetes in comparison to the general population. However, more than 90% of all patients developing T1DM do not have an affected family member. Therefore, it is crucial to establish a standardized screening method which will efficiently identify individuals at high risk in a general population. School children between 5-18 years of age were screened to evaluate the predictive value of autoantibodies over a period of 6-12 years (178). This study indicated that the risk of developing T1DM when ICA is detected in the absence of other autoantibodies is low, whereas with more than one autoantibody (either GAD65A, IAA, IA-2A or IA-2ßA) the risk of developing T1DM in a general population is high. Similar findings were also reported in other studies (179-181). These results support the value of multiple autoantibodies as good predictive markers for T1DM not only in first degree relatives but also in the generalpopulation.  Consequently, the American Diabetes Association now considers the presence of 2 or more autoantibodies as form of early presymptomatic diabetes (182).

 

Prevention Trials in Type 1 Diabetes

 

The elucidation of the natural history of pre-diabetes has allowed for the characterization of those individuals at greatest risk for developing autoimmune T1DM, through the use of genetic, immunologic and metabolic markers. This predictive ability has become possible in both high- risk relatives and the general population as mentioned above. The subclinical autoimmune destruction of ß-cells in the pancreas may last from a few months to several years. This pre- diabetic period has allowed investigators to test prevention strategies, which mainly have focused in modulation of autoimmune process (183). A number of studies initiated with general immunosuppressive agents, such as cyclosporin-A, azathioprine and prednisone in patients with new clinical onset T1DM, positive results in that insulin free remission rates were increased and endogenous insulin (C-peptide) reserves were improved (121). However, despite continued immunotherapy with the attendant risks of renal damage and lymphomas at higher doses, relapses proved to be the rule and such treatments were abandoned. Cyclosporin given at a prediabetic phase of the disease delayed but did not prevent diabetes (184,185).

 

With the observation that nicotinamide prevents pancreatic ß cell destruction from streptozotocin by raising otherwise depleted levels of islet cell NAD as a result of superoxide induced DNA breaks and repair, the vitamin was subjected to a large European and Canadian trial called The European Nicotinamide Diabetes Intervention Trial (ENDIT). However, nicotinamide failed to prevent progression to diabetes (186). In addition, a  study in Germany (DENIS)   was completed without any effect of nicotinamide on prevention of T1DM.(187).More recent studies have used Anti CD21(Rituximab), Anti CD3, Anti CTLA-4, oral insulin,GAD65 peptides, and infusions of Treg cells  with early encouraging results in preserving insulin secretion, but without durable effects (188). These results in humans were often based on animal studies in NOD mice (189-191). In stark contrast to these encouraging studies in NOD mice, where a variety of interventions induce long lasting remissions, none of the studies in humans has so far yielded long-lasting remissions in humans (183,188).

 

Table 6. Prevention Trials (121)

Study and Phase

Drug

Age

Eligibility

Ref

TRIGR

Cow’s milk hydrolysate

0-7 days

First Degree relatives, High-risk HLA

(192)

BABY DIET

Gluten-free diet

Younger than 3 months

Relatives, high risk HLA DR, DQ

(193)

TrialNet NIP

Docosahexaenoic acid

>24 weeks gestation- newborn

Relatives, HLA DR3 or DR4

(194)

TrialNet Teplizumab

Teplizumab

8-45 years

At least 2 confirmed autoantibodies and abnormal glucose tolerance

 (195,196)

DIAPREV-IT

GAD-alum

4-18 years

Islet autoantibody positive

(197)

TrialNet Oral Insulin, Phase III

Human insulin

1-45 years

Relatives, 2+islet antibodies including to insulin

(198)

INIT I/II,

 

Intranasal insulin

4-30 years

Relatives, 2+islet antibodies, HLA not DR2, DQ6

(199)

Pre-Point, Phase I/II

Human insulin

1.5-7 years

First degree relatives,

>50% risk of T1DM

(200)

FINDIA

Insulin-free whey- based formula

Infants

General population, high-risk HLA DQ

(201)

Teplizumab

Teplizumab

</=18 years of age

Relatives

(202)

Golimumab

Golimumab

6 to 21 years

Newly diagnosed T1DM

(203)

 

TYPE 2 DIABETES MELLITUS

 

As the US passed into the 21st century, the epidemic of obesity and T2DM continues unabated, affecting more younger adults and children than in the past.  They will spend longer periods of their life with the disease. Perhaps in part under pressure of commercial interests, we as a nation have learned to eat too fast, too much, and the wrong foods.  However, the problem of obesity and its consequences is pervasive globally, affecting developing as well as economically developed countries.  For those with the energy conserving "thrifty" genes of insulin resistance syndrome (IRS), this excess of food and especially of the insulin provoking carbohydrates, leads to obesity, an IRS phenotype and T2DM. Nearly half of the new cases of diabetes in teens can be termed T2DM (204).  Currently, in some US states where there are large numbers of ethnic groups prone to IRS and T2DM (Hispanics, American Indians, Asian Indians, African Americans), the number of children with T2DM is beginning to rival if not surpass the number with T1DM. It is estimated that 1 in 3 people born in the US in the year of 2000 will develop T2DM sometime in their lifetime (205).

 

The increased incidence of T2DM is attributed to the increase in obesity worldwide. Approximately 3700 youths are diagnosed with T2DM every year in the US (206) and it is estimated that the number of youth with T2DM will almost quadruple from 22,820 in 2010 to  approximately 85,000 adolescents with T2DM by 2050 (10). Similar rates of increased in youths with T2DM are reported from the UK, India, China and Japan (10).

 

Pathophysiology of Type 2 Diabetes

 

T2DM is characterized by insulin resistance in peripheral tissues (muscle, fat, and liver) with progressive β cell failure, ,especially manifest with defective insulin secretion in response to a glucose stimulus, increased glucose production by the liver, and no markers of pancreatic autoimmunity (207). The progressive decline in β cell function is more rapid in youths at 20-30% decline per year versus 7-11% decline per year in adults, even with aggressive medical therapy.

 

Table 7. Pathophysiologic Factors

Obesity/Insulin resistance (IR)

See IRS

Intrauterine environment

Epidemiological studies have shown a strong association between poor intrauterine growth and the subsequent development of the Metabolic Syndrome. It was suggested that the effects of poor nutrition in early life impair the development of pancreas and resulting permanent changes in glucose- insulin metabolism (208).

Gestational diabetes

Studies in Pima Indian women showed significant increased risk of developing T2DM in offspring of women with diabetes during pregnancy compared to non-diabetic mothers (209).

Ethnicity

There is a significant increase risk in certain ethnic/race groups (205).

Gender and puberty

Puberty is a state of IR brought about by the increased secretion of GH during this process. There is a 30%-50% decrease in insulin sensitivity and compensatory increase in insulin secretion. Those that have an inherent defect in insulin secretion and inadequate response to the resistance develop DM. The mean age at diagnosis of T2DM in children is 13.5 years, corresponding to the time of peak adolescent growth and development.

Girls are 1.5-3 times more likely than boys to develop T2D as children or adolescents (270).

Family History

Between 74-100% of children with T2DM have a first or second-degree relative with T2DM. The lifetime risk is 40% if one parent is affected and 70% if both parents are affected (210).

Genetics

Genome-wide studies led the discovery of single- nucleotide polymorphisms (SNPs) at several loci regulating insulin secretion.  To date, more than 30 diabetes-related SNPS (diabetoSNPs) have been identified (211).

Several genes have been found to be associated with T2D;

1.     1) Peroxisome Proliferator-Activated Receptor-γ2 (PPAR-γ2) Gene: An important regulator of lipid and glucose homeostasis. Missense mutation Pro12Ala in PPAR-γ2 is associated with decreased risk for T2DM.

2.     2) Kir6.2 Gene (KCNJ11): The missense mutationGlu23Lys in the Kir6.2 gene has been associated with increased risk of T2DM.

3.     3) MODY genes (HNF4α and HNF1β)

4.     4) Transcription Factor 7-like (TCF7L2) Gene: A product of HMG box containing transcription factors that play role in the glucose homeostasis. Specific polymorphisms in the TCF7L2 gene increase the risk of progression from IGT toT2DM.

5.     5) Calpain-10 Gene: Calpains are Ca+2 dependent cysteine proteases and play a role in regulating insulin secretion and action.

 

 

The natural history of progression to T2DM is that a person with IRS begins to decompensate, with a fall in the disposition index (the amount of insulin produced for the degree of insulin resistance). Subsequently levels of blood glucose rise after feeding; elevations in fasting blood glucose levels occur later. At this early stage, diet, exercise and insulin sensitizers are indicated.

 

INSULIN RESISTANCE SYNDROME (IRS)

 

This syndrome complex is centered upon genetic predispositions to insulin resistance and the hyperinsulinemia that results from it. This medical state is also named syndrome X and the metabolic syndrome, however the descriptive term insulin resistance syndrome (IRS) is the one increasingly used in the literature (207,212). In IRS, there are poorly understood genetic lesions that lead to insulin resistance from early life if not during embryogenesis. In many affected families, the disease occurrences suggest a dominant mode of transmission. In rare families, mutations affecting insulin receptors, or peroxisome proliferators-gamma (PPAR- gamma) expression may be the cause of it (213). IRS is the association of insulin and leptin resistance with obesity (typically with increased visceral fat), functional adrenal hyper-androgenism, functional ovarian hyperandrogenism, hypersecretion of pituitary LH, dyslipidemia, hypertension, and features of hyperinsulinemia such as late reactive hypoglycemia and acanthosis nigricans. When the compensation by increased insulin secretion fails, glucose intolerance and T2DM result.

 

Natural History of Insulin Resistance Syndrome

 

Several studies indicate that many children and adults with T2DM were born small for gestational age. This suggests that the insulin resistant state existed in-utero since it is insulin rather than pituitary growth hormone that is the principal growth-promoting hormone of the unborn child, and decreased insulin action might be anticipated to impair embryonic growth. After birth, premature pubarche resulting from excessive adrenal androgens such as dihydroepiandrosterone (DHEA) may occur, even before obesity has appeared. Thus, it has been proposed by some that obesity may be the result of insulin resistance, and not its cause. Excessive DHEA may be seen best after ACTH injection leading to a clinical suspicion that the 3ß hydroxysteroid dehydrogenase enzyme is underactive. Obesity can begin from infancy but often dates from about 8 years of age when physiological pubarche occurs. Early onset obesity raises the possibility of a genetic satiety causation such as the Prader-Willi Syndrome or deficiency of MC4R. Acanthosis nigricans resulting from increased keratinocytes in certain areas of skin is thought to result from insulin stimulation of insulin-like growth factor 1 (IGF-1) receptors and often manifests during puberty Menarche may be delayed in age at onset or menses may be missed after menarche, or else there can be dysfunctional bleeding resulting from anovulatory cycles.

 

Hirsutism often becomes bothersome during adolescence, as may male pattern hair thinning, persistent acne and development of polycystic ovaries. An increase in very low-density lipoprotein (VLDL) secretion by the liver is observed with increasing age, associated with diminished, atherogenesis protective, high density lipoprotein cholesterol (HDL-C), a dyslipidemic profile that promotes early and progressive onset of atherosclerosis, predisposing to coronary heart disease (CHD), stroke, and peripheral vascular diseases in later life. The latter problems are compounded by the appearance of hypertension and type-2 diabetes. The glucose intolerance that precedes type-2 diabetes often first involves post-prandial glucose levels or the two-hour time point of the OGTT as discussed above, but later induces a rise in fasting glucose (impaired fasting glucose) levels as well. The mechanism is thought to be ß cell exhaustion or more likely a glucosamine and lipid mediated islet cell toxicity. Once this stage is reached, damage to the islets can become irreversible, resulting in the dual problems of insulin resistance and insulinopenia, both of which need to be addressed in therapeutic strategies.  In children and adolescents, the progression of impaired insulin secretion and its complications including the appearance of albuminuria, exhibits a faster tempo than that of adults presenting later in life. Hence, these adolescents may more rapidly progress to requiring insulin therapy.

 

Table 8. Clinical features of IRS. Adapted from refs (210,213,214).

Clinical Features

 

Infancy

Family history of obesity and T2DM, SGA, LGA

Gestational Diabetes

Childhood/Adolescence

Acanthosis nigricans Premature adrenarche, Obesity, Pseudoacromegaly, Striae, Skin tags, Amenorrhea

Adulthood

Tall Stature, Pseudoacromegaly Fatty liver, Focal glomerulosclerosis

Hirsutism, Ovarian hyperandrogenism, PCOS

Endothelial dysfunction, Atherosclerosis, Increased carotid wall thickness, Stroke CHD

Glucose intolerance, T2DM

 

Table 9. Laboratory Features of IRS

↓IGFBP-1, ↓SHBG, ↑free testosterone

↓CBG, ↑free cortisol

↑VLDL, ↑TG, ↓HDL, ↑ small dense LDL

Increased PAI-1, CRP, fibrinogen

Adhesion molecules and uric acid

Decrease first phase insulin response

Increased decompensated insulin resistance

Postprandial hyperglycemia

Fasting hyperglycemia

Diabetes

 

Underlying Mechanisms of Insulin Resistance

 

OBESITY

 

Affected patients commonly show polyphagia, and may have voracious appetites that are characteristically resistant to dietary advice. When leptin deficiency was discovered in Ob/Ob mice and leptin receptor deficiency discovered in Db/Db mice, the adipocyte became to be appreciated as an endocrine cell rather than one that was an inert repository of triglycerides. However, the promise of a breakthrough in the understanding of human obesity was quickly dissipated when such lesions proved to be rare in humans. Obese patients with their greater degrees of adiposity also have the highest levels of leptin as expected, however these high levels do not reduce the appetites of IRS patients (215). Thus, such patients are also leptin resistant. Early trials of leptin therapy have not affected weight loss. However, patients with lipodystrophy who have leptin deficiency develop insulin resistance, hyper-insulinemia, dyslipidemia and T2DM, all of which respond dramatically to leptin given as therapy (216,217).   Deficiencies in other appetite suppressing hormones such as resistin have more recently been implicated but not yet shown to have therapeutic relevance. Hyperinsulinemia itself is a compounding variable, in that excessive carbohydrate containing diets stimulate the highest levels of insulin and the greatest degrees of adiposity. Therapies such as metformin that improve insulin sensitivity when combined with a diet restricted in low amounts of simple carbohydrates and exercise, can dramatically lower weight in children with IRS when they adhere to therapeutic guidelines. However, failure to adhere to instructions is a common problem in adolescents (218,219).

 

HYPERANDROGENISM

 

It is uncertain as to the degree to which the pituitary abnormality of increased LH secretion leads to the androgenic excess or vice versa. Probably, both are responses to the insulin resistance and hyperinsulinemia of IRS by mechanisms that have yet to be clearly understood. Androgens of ovarian origins usually predominate over those of the adrenal gland, albeit both are often found to be elevated. Sex hormone binding globulins in the circulation are often low, resulting in increased free androgens with their increased bio-availability (220). This is often seen with testosterone, which can be raised or normal in hirsute girls whereas increased free testosterone levels are common.

 

Interestingly, we hold that there is a clinical overlap between Cushing's syndrome and IRS (221). Both tend to have visceral (central) obesity and striae suggestive of glucocorticoid excess. However, whereas the patient with Cushing's syndrome has high levels of serum cortisol, the patient with IRS has low normal levels, albeit both have increased levels of urinary free cortisol. Again, the explanation may lie in the low levels of corticosteroid binding globulins found in IRS where circulating cortisol is disproportionately free. Some investigators have suggested that there is an impaired conversion of cortisol to the metabolically inactive cortisone in IRS. Further, the child with Cushing's syndrome is invariably growth retarded in contrast to the child with IRS whose linear growth tends to be excessive. In IRS and obesity, the GH levels during stimulation tests are suppressed implying a diagnosis of GH deficiency which likely is not the case as these children tend to be tall. IGFBP levels in serum are depressed, resulting in an excessive free IGF-1 level, albeit the total IGF-1 concentration is usually normal. The pseudo-acromegaly observed in severely affected children with IRS may be occurring via this mechanism. In addition, high concentrations of insulin interact with the IGF-I receptor, thereby promoting growth (222).

 

ACANTHOSIS NIGRICANS

 

Stimulation of the IGF-1 receptors of skin keratinocytes by high levels of circulating insulin is thought to explain their hyperplasia and excessive laying down of keratin in the skin of the neck, axillae, elbows and knees, skin creases and indeed most areas of skin (223). In addition, excessive free IGF-1 may have the same effect, albeit the greater the degree of insulin resistance, the higher the insulin levels, the more striking the acanthosis nigricans. Increased bioavailability of IGF-1 (high IGF-1 and low IGFBP-1) are directly correlated with the severity of acanthosis nigricans

 

GLUCOSE INTOLERANCE AND T2DM

 

Children and young adults affected by IRS are often hyperinsulinemic. In such persons, stimulation of insulin secretion by carbohydrates alone or with protein can induce an excessive but delayed rise in insulin secretion, reflected in an early excessive rise in glucose, followed by an excessive fall in glucose levels 3-5 hours afterwards, of sufficient severity to provoke symptoms of hypoglycemia. As the ability to secrete insulin declines, impaired glucose intolerance appears first. Later in the evolution of T2DM, the 2-hour criteria for diabetes during OGTT become apparent, followed later by impaired fasting hyperglycemia and finally by fasting hyperglycemia that meets the criteria for the diagnosis of diabetes. An HbA1c level can be used to screen diabetes as recommended by the American Diabetes Association.

 

Table 10. Criteria for Increased Risk of Diabetes (1)

Fasting plasma glucose

100 – 125 mg/dl

2-hour plasma glucose after OGTT

140 – 199 mg/dl

HbA1C

5.7 – 6.4%

 

NON-ALCOHOLIC STEATOHEPATITIS (NASH)

 

It is also known as fatty liver or hepatic steatosis. The incidence of fatty liver among obese children was 2.6% in one study (224), and hyperinsulinemia was found to be the major contributor for its’ development (225). A number of factors may play a role in the development of fatty liver including, induction of cytochrome P4502E1 during obesity, which is capable of generating free radicals, while the high level of dietary intake of polyunsaturated fatty acids or low intake of nutritional antioxidants contributes to the oxidative stress. Fatty liver alone appears to be a relatively benign disease, and can be reversible. However, it may progress over years to hepatic cirrhosis, liver failure, or hepatocellular carcinoma. The onset of disease is usually insidious. Laboratory evaluation indicates mild to moderate elevation of serum aminotransferases in most children and serum alanine aminotransferase (ALT) levels had been shown a useful screening for fatty liver in obese children (226). The ratio of aspartate aminotransferase (AST) to ALT is usually less than 1, but this ratio increases as fibrosis advances. Serum aminotransferases, alkaline phosphatase and gamma glutamyl transferase (GGT) levels are proposed surrogate markers of fatty liver (227,228).

 

RENAL INVOLVEMENT

 

A form of focal glomerulosclerosis (often with IgA deposition) appears to be associated with IRS, leading to microalbuminuria. Hypertension becomes increasingly common through adolescence and beyond. The mechanisms responsible have not been elucidated.

 

INFLAMMATION

 

IRS and T2DM have increased markers of inflammation. This takes the form of increased levels of C-reactive protein, raised erythrocyte sedimentation rates (ESR) and increased cytokine (TNF-α) levels.  Obese patients also have abnormalities of thyroid function suggestive of primary thyroid deficiency with modestly elevated TSH but normal or slightly elevated fT4 and fT3.These abnormalities resolve with weight loss and have therefore been interpreted as representing an adaptive response to obesity i.e., by raising TSH and free T3, caloric expenditure would increase (229-231). Obese patients are thus often unnecessarily treated for hypothyroidism they do not have. They may however develop true hypothyroidism on the basis of associated Hashimoto's disease.

 

ATYPICAL DIABETES

 

Genetic Defects of ß-cell Function (Monogenic Diabetes)

 

Monogenic forms of diabetes are characterized by impaired secretion of insulin from pancreatic β cells caused by a single gene mutation. These forms comprise a genetically heterogenous group of diabetes including, maturity onset diabetes of the young (MODY), permanent or transient neonatal diabetes (NDM), and mitochondrial diabetes. MODY is the most common form of monogenic diabetes, with autosomal dominant transmission of a gene encoding a primary defect in insulin secretion (232-235).

 

Approximately 1 to 2 % of diabetes in Europe is MODY (236). The clinical characteristics of these patients are heterogeneous, and not reliable in predicting the underlying pathogenesis (237,238). It is often misdiagnosed as T1DM or T2DM. Several genetic abnormalities have been found that account for the disorder. Some members of an affected family may have the genetic defect but not develop the diabetes phenotype. Whether this is due to modifying genes or environmental factors is unclear. MODY differs from the classical immunological T1DM in several ways. With MODY, a dominant family history of diabetes (if known) is always present.  However, de novo mutations can occur.  Hyperglycemia is mostly mild with a minimal tendency to ketosis before the age of 25 years, the insulin secretion in response to oral (OGTT) or intravenous (IVGTT) glucose administration is modestly decreased, and evidence of islet cell autoimmunity is absent. It is estimated that more than 80% of patients with monogenic diabetes are either not diagnosed or are misclassified as type 1 or type 2 DM (239).

 

The underlying genetic defects of the many MODY subtypes have been identified, as indicated below (Table 11). To date, fourteen genetic forms of MODY are recognized. MODY resulting from defects in the glucokinase gene (GCK) and hepatocyte nuclear factor-1-alpha (HNF-1α) are the most common types seen during childhood (MODY-2) and post puberty (MODY-3), respectively.  MODY Types 2 and 3 together constitute 80% of all cases of MODY syndromes.

 

MODY 2 is the most common form of MODY with a prevalence of about 1:1000 people. It is caused by a dominant heterozygous inactivating mutation in glucokinase, the enzyme that phosphorylates glucose to permit its oxidation to ATP and hence insulin release. Insulin is released but at higher glucose concentration-the curve is right shifted but otherwise normal. Thus, fasting glucose is in the range of ~95-110 mg/dl and may remain above 140 mg/dl at 2 hours post prandial but returns to normal thereafter. HbA1c is in the range of 5.8-7.6% and generally remains in the low- mid 6% range. Patients are rarely symptomatic and may be discovered by chance when a blood glucose is obtained. Treatment is not necessary except during pregnancy in some cases; there is a very low prevalence of micro-macrovascular disease even after almost 50 years of follow-up. Young women are often discovered to have mid hyperglycemia when tested during pregnancy and erroneously labeled as having gestational diabetes. The non-affected fetus of an affected Mother may have some macrosomia in utero-the result of extra insulin secretion by the fetus in response to the maternal hyperglycemia (240).

 

MODY3 is the next most common form of MODY caused by a heterozygous mutation in HNF-1α, necessary for normal insulin secretion. Onset is usually in the teen years and glucose is in the mid-200s with mild to moderate symptoms. Patients may respond to sulfonylurea drugs initially, but later may go on to insulin dependence and more severe hyperglycemia. As with other MODY forms, a family history of diabetes is often obtained, with a diagnosis of T2DM common for older patients and T1DM in younger patients. Confirmation of the diagnosis by molecular testing is essential for recommending treatment and family counseling (241).

 

Defects in four pancreatic ß cell-specific transcription factor genes, HNF-1β (MODY5), HNF-(MODY1), pancreatic and duodenal homeobox 1 gene (PDX1) [previously termed insulin promoter factor-1 (IPF-1)] (MODY4) and neurogenic differentiation 1 gene (NeuroD1) and BETA2 (MODY6) are responsible for others. In contrast to MODY-2, patients with heterozygous mutations in the HNF1A, HNF4A, or HNF1B and more rarely in PDX1 or NEUROD1 have progressive deterioration in glucose tolerance and are at risk for developing complications of diabetes (242).

 

More recently, mutations in the tumor suppressor protein KLF-11 (MODY7), the carboxyl ester lipase CEL (MODY8), the transcription factor, paired box gene 4, PAX-4 (MODY9), the insulin gene, INS (MODY10), and tyrosine kinase, B-lymphocyte specific gene, BLK (MODY11) have been described.  MODY 12 and MODY 13 are due to mutations in the ABCC8 and KCNJ11 genes, respectively. Mutations in these 2 genes also have been reported in neonatal diabetes.  They are very rare and represent fewer than 1% of all MODY cases.

 

Table 11. Classification of MODY

MODY Type

Gene

Gene Loci

Incidence

Age at Diagnosis

Primary Defect

Associated Features

Severity of   Diabetes

Ref

1

HNF-4α 20q

Rare

Postpubertal

Transcription gene defects in ß-cells lead to impaired metabolic signaling of insulin secretion.

-

Severe

(242)

2

Glucokinase

7p

10-60%

Childhood

impairment of ß-cells sensitivity to glucose and; defect in hepatic glycogenesis

Reduced birth weight

Mild

(243)

3

HNF-1α

12q

20-60%

Postpubertal

Similar to MODY1

Renal glucosuria

Severe

(242-246)

4

PDX1 (IPF-1)13q

Rare

Early adulthood

Defects in transcription factors during embryogenesis lead to abnormal ß-cell development and function

-

Mild

(247)

5

HNF-1β 17cen- q21.3

Unknown

Postpubertal

Similar to MODY 1 and 3

Glomerulocystic kidney disease, female genital malformations, Hyperuricemia, abnormal liver function tests

Mild

(248)

6

NeuroD1/BETA2

2q32

Rare

Early adulthood

Defect in this gene causes abnormal development of ß cell and function

-

Unknown

(249)

7

KLF11  

2p25

Very Rare

Early adulthood

Reduced glucose sensitivity of the beta cell

Phenotype similar to T2D

Unknown

(250)

8

CEL      

9q34

Very Rare

 

<20 years

Impaired endocrine and exocrine pancreatic function

Exocrine pancreatic dysfunction

Unknown

(251)

9

PAX4   

7q32

Very Rare

<20 years

Impaired gene transcription in pancreatic beta cells on apoptosis and proliferation

-

DKA is possible

(252,253)

10

INS      

11p15.5

Very Rare

<20 years

Defect in this gene may result the loss of beta cell mass through apoptosis

-

Unknown

(254)

11

BLK      

8p23

Very Rare

<20 years

decreases insulin synthesis and secretion in response to glucose by up- regulating transcription factors

Higher incidence in obese individuals

Unknown

(255)

12

ABCC8 

11p15.1

< 1%

<35 years

Inactivating mutations cause impaired secretion mild mode

 

 

(255)

 

13

KCNJ11

11p15.1

<1%

 

 

 

 

 

14

APPL1  

3p14.3

<1%

 

adapter protein, phosphotyrosine interacting with pH domain and leucine zipper

 

 

(256,257)

 

Neonatal Diabetes

 

Neonatal diabetes is a rare disorder with an incidence of 1:100,000-1:200,000 live births (232,258).  It presents in first 6 months of life and its’ severity depends on the underlying mutation in that it is either transient or permanent. Almost 50% of cases with neonatal diabetes are permanent (PND) while the remainder are “transient” (TNDM) in that they remit, but may reappear and become apparent later in life or at times of stress. Heterozygous activating mutations in KCNJ11 and ABCC8 —which encode the Kir6.2 and SUR1 subunits, respectively, of the ATP-sensitive potassium channel, are the most common causes of PND. Missense mutations in the INS gene are also identified in patients with PND and they may have an autosomal dominant or recessive inheritance pattern (232,254,258). Genetic diagnosis is important since the KCNJ11 and ABCC8 mutations respond to treatment by sulfonylureas, possibly without need for additional insulin therapy because these drugs can close the β cell potassium channel by an ATP-independent route (259). It is increasingly apparent that the same mutations can become manifest for the first time well beyond infancy and diagnosed as T2DM or rarely T1DM. Severe mutations in the KATP genes, especially KCNJ11 also may present with a neurological component in a syndrome known as DEND (Developmental delay, Epilepsy, Neonatal Diabetes); early diagnosis and treatment with sulfonylurea drugs is reported to ameliorate the neurological manifestations as the KATP channels are expressed in the brain. The major form of   transient neonatal diabetes results from anomalies of the imprinted region on chromosome 6q24,but mutations in KCNJ11 or ABCC8 can also cause TNDM (232).  Various rare forms of syndromic disease which include NDM are described; early diagnosis may diminish or delay the hitherto described natural history and consequences (258).

 

Mitochondrial Diabetes

 

Point mutations in mitochondrial m.3243A→G cause another form of diabetes with an insulin secretory defect that is commonly associated with neuro-sensory hearing impairment and a strict maternal mode of inheritance (260). In addition, genetic abnormalities that result in the inability to convert pro-insulin to insulin (261), or the production of mutant insulin molecules (262), are other examples of specific genetic defects in ß cell function which are rare causes of diabetes.

 

Chronic Illnesses

 

Hemochromatosis is a progressively more common recognized cause of diabetes with aging, and does not present in a pediatric age group. However repeated blood transfusions for conditions such as thalassemia major can lead to diabetes associated with hemosiderosis.

Many patients with cystic fibrosis develop a form of T1DM often during their teenage years which may require insulin replacement and is labeled “cystic fibrosis related diabetes (CFRD)” (263).  Most CF patients now live long enough for this to have become a more common problem with impact on overall well-being and severity of symptoms ascribed to CF and partially responsive to insulin therapy. DKA is rare in CFRD, perhaps because of the concurrent effects on the α-cell secreting glucagon as well as the β-cell secreting insulin. Patients with Gitelman’s syndrome develop diabetes which resolves when they are adequately replaced with magnesium, excessively lost through the kidneys in this syndrome. Gitelman syndrome is a recessively inherited genetic entity, but the presentation of DM is usually not until later midlife (264).

 

Genetic Defects in Insulin Action

 

There are a series of rare genetic abnormalities in the insulin receptor, or in the signal transduction events which follow insulin docking to its receptor resulting in diabetes. The recessive DNA breakage disease (Bloom’s syndrome) is associated with mild diabetes due to severe insulin resistance, with very high levels of circulating insulin and insulin like growth factor one (IGF-1). Progeria and lipodystrophy are other such causes (232). In the latter case, the absolute deficiency of leptin leads to uncontrolled lipolysis resulting in severe insulin resistance, which is partially reversible by leptin administration (232)/

 

Endocrinopathies Associated with Hyperglycemia

 

Several hormones, such as epinephrine, glucagon, cortisol, and growth hormone, antagonize the action of insulin. Whereas release of these hormones constitutes the protective counter regulatory response to hypoglycemia, primary over secretion of these hormones can result in glucose intolerance or overt diabetes.

 

  • Cushing's syndrome, due to pituitary and ACTH secreting adenomas or adrenal hyperplastic disease or to exogenous glucocorticoid administration, can lead to diabetes (265). Steroid-induced diabetes is most often seen when there is pre- existing insulin resistance or a defect in insulin synthesis/secretion unmasked by the inability to increase insulin secretion to overcome the resistance to its actions induced by glucocorticoids.
  • Acromegaly is associated with overt diabetes in 10 to 15% of cases, and impaired glucose tolerance in a further 50% (266,267). In acromegaly, there is marked insulin resistance and hyperinsulinemic responses; DM occurs only when the hyperinsulinemic response cannot match the requirement to overcome the degree of resistance.
  • Pheochromocytomas are associated with both inhibition of insulin secretion and an increase in hepatic glucose output (268). These changes lead to impaired glucose tolerance, the severity of which is directly related to the magnitude of catecholamine production (269). When seen in children, these are usually a component of the Von Hippel-Lindau syndrome, MEN2, and NF1.
  • Glucagon-secreting tumors (glucagonoma) are associated with an unusual constellation of clinical features, including skin rash, weight loss, anemia, and thromboembolic problems. Approximately 80% of these patients have either impaired glucose tolerance or diabetes (270).
  • Somatostatin-secreting tumors (somatostatinomas) are typically associated with the triad of diabetes mellitus, cholelithiasis, and diarrhea with steatorrhea (271).
  • Although thyroxine is not a counter regulatory hormone, hyperthyroidism can interfere with glucose metabolism. It is associated with both increased sensitivity of pancreatic ß cells to glucose, resulting in increased insulin secretion, and antagonism to the peripheral action of insulin. The latter effect usually predominates, leading to impaired glucose tolerance in some untreated patients (272).

 

Drug- or Chemical-induced Diabetes 

 

A large number of drugs can impair glucose tolerance; they may act by decreasing insulin secretion, increasing hepatic glucose production, and/or by causing resistance to the action of insulin (273). Included in this list are several classes of antihypertensive drugs, such as beta blockers (274), protease inhibitors used for the treatment of HIV infection (275), and tacrolimus and cyclosporine used primarily to prevent transplant rejection (276,277). Drugs of the serotonin re-uptake inhibitor (SSRIs) class can lead to obesity, impaired glucose intolerance and T2DM, especially if individuals were already insulin resistant before they started such medications.

 

There is a common association between obesity, insulin resistance, hypertension, and dyslipidemia, which has been called syndrome X or the metabolic syndrome (207,212,278,279). The administration of a thiazide diuretic or a ß-blocker to such patients can exacerbate the insulin resistance and may bring on hyperglycemia (274). In comparison, angiotensin-converting enzyme (ACE) inhibitors and alpha-adrenergic antagonists (such as doxazosin) may improve insulin sensitivity. Because the former also protect against renal disease, they are the drugs of choice for diabetic patients with hypertension.

 

Viral Infections

 

Certain viruses e.g., Coxsackie B4, have been implicated to cause diabetes, either through direct ß cell destruction or possibly by inducing autoimmune damage. The direct proof of this however remains tenuous. Chronic hepatitis C virus infection is associated with an increased incidence of diabetes, but it remains uncertain as yet if there is a cause-and-effect relationship.

 

Uncommon Forms of Immune-Mediated Diabetes

 

Several uncommon forms of immune-mediated diabetes have been identified.

 

  • The stiff-man syndrome is an autoimmune disorder of the central nervous system, which is characterized by progressive muscle stiffness, rigidity, and spasms involving the axial muscles, with impairment of ambulation (280). Patients characteristically have high titers of glutamic acid decarboxylase (GAD65) autoantibodies and diabetes occurs in at least one-third of cases. Graves’ disease is also common in the syndrome. Presentation is usually in early
  • Anti-insulin receptor antibodies can bind to insulin receptors and either act as an agonist, leading to hypoglycemia, or block the binding of insulin and cause diabetes (281). This so-called type B insulin resistance is more common in females who show other signs of autoimmunity including systemic lupus erythematosus (SLE). However one study found that almost 10% of young patients with insulin resistance in the absence of autoimmune stigmata were also positive for insulin receptor autoantibodies (282).

 

REFERENCES

 

  1. American Diabetes Association. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2021. Diabetes Care 2021; 44:S15-S33
  2. American Diabetes Association. Standards of Medical Care in Diabetes-2021 Abridged for Primary Care Providers. Clin Diabetes 2021; 39:14-43
  3. Soleimanpour SA, Stoffers DA. The pancreatic beta cell and type 1 diabetes: innocent bystander or active participant? Trends Endocrinol Metab 2013; 24:324-331
  4. Atkinson MA, Bluestone JA, Eisenbarth GS, Hebrok M, Herold KC, Accili D, Pietropaolo M, Arvan PR, Von Herrath M, Markel DS, Rhodes CJ. How does type 1 diabetes develop?: the notion of homicide or beta-cell suicide revisited. Diabetes 2011; 60:1370-1379
  5. Harjutsalo V, Sjoberg L, Tuomilehto J. Time trends in the incidence of type 1 diabetes in Finnish children: a cohort study. Lancet 2008; 371:1777-1782
  6. Moltchanova EV, Schreier N, Lammi N, Karvonen M. Seasonal variation of diagnosis of Type 1 diabetes mellitus in children worldwide. Diabet Med 2009; 26:673-678
  7. Diabetes Group. Incidence and trends of childhood Type 1 diabetes worldwide 1990- 1999. Diabet Med 2006; 23:857-866
  8. Mayer-Davis EJ, Lawrence JM, Dabelea D, Divers J, Isom S, Dolan L, Imperatore G, Linder B, Marcovina S, Pettitt DJ, Pihoker C, Saydah S, Wagenknecht L, Study SfDiY. Incidence Trends of Type 1 and Type 2 Diabetes among Youths, 2002-2012. N Engl J Med 2017; 376:1419-1429
  9. Patterson CC, Gyurus E, Rosenbauer J, Cinek O, Neu A, Schober E, Parslow RC, Joner G, Svensson J, Castell C, Bingley PJ, Schoenle E, Jarosz-Chobot P, Urbonaite B, Rothe U, Krzisnik C, Ionescu-Tirgoviste C, Weets I, Kocova M, Stipancic G, Samardzic M, de Beaufort CE, Green A, Dahlquist GG, Soltesz G. Trends in childhood type 1 diabetes incidence in Europe during 1989-2008: evidence of non-uniformity over time in rates of increase. Diabetologia 2012; 55:2142-2147
  10. Imperatore G, Boyle JP, Thompson TJ, Case D, Dabelea D, Hamman RF, Lawrence JM, Liese AD, Liu LL, Mayer-Davis EJ, Rodriguez BL, Standiford D, Group SfDiYS. Projections of type 1 and type 2 diabetes burden in the U.S. population aged <20 years through 2050: dynamic modeling of incidence, mortality, and population growth. Diabetes Care 2012; 35:2515-2520
  11. Gillespie KM, Bain SC, Barnett AH, Bingley PJ, Christie MR, Gill GV, Gale EA. The rising incidence of childhood type 1 diabetes and reduced contribution of high-risk HLA haplotypes. Lancet 2004; 364:1699-1700
  12. VanBuecken D, Lord S, Greenbaum CJ. Changing the Course of Disease in Type 1 Diabetes. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, eds. Endotext. South Dartmouth (MA)2000.
  13. Collaboration NCDRF. Worldwide trends in diabetes since 1980: a pooled analysis of 751 population-based studies with 4.4 million participants. Lancet 2016; 387:1513-1530
  14. American Diabetes Association. 2. Classification and Diagnosis of Diabetes. Diabetes Care 2017; 40:S11-S24
  15. Standards of Medical Care in Diabetes-2017: Summary of Revisions. Diabetes Care 2017; 40:S4-S5
  16. Ostergaard JA, Laugesen E, Leslie RD. Should There be Concern About Autoimmune Diabetes in Adults? Current Evidence and Controversies. Curr Diab Rep 2016; 16:82
  17. Greenbaum CJ, Speake C, Krischer J, Buckner J, Gottlieb PA, Schatz DA, Herold KC, Atkinson MA. Strength in Numbers: Opportunities for Enhancing the Development of Effective Treatments for Type 1 Diabetes-The TrialNet Experience. Diabetes 2018; 67:1216-1225
  18. Ganda OP, Srikanta S, Brink SJ, Morris MA, Gleason RE, Soeldner JS, Eisenbarth GS. Differential sensitivity to beta-cell secretagogues in "early," type I diabetes mellitus. Diabetes 1984; 33:516-521
  19. Cantor AB, Krischer JP, Cuthbertson DD, Schatz DA, Riley WJ, Malone J, Schwartz S, Quattrin T, Maclaren NK. Age and family relationship accentuate the risk of insulin-dependent diabetes mellitus (IDDM) in relatives of patients with IDDM. J Clin Endocrinol Metab 1995; 80:3739-3743
  20. Rewers M, Norris JM, Eisenbarth GS, Erlich HA, Beaty B, Klingensmith G, Hoffman M, Yu L, Bugawan TL, Blair A, Hamman RF, Groshek M, McDuffie RS, Jr. Beta-cell autoantibodies in infants and toddlers without IDDM relatives: diabetes autoimmunity study in the young (DAISY). J Autoimmun 1996; 9:405-410
  21. Maclaren N, Lan M, Coutant R, Schatz D, Silverstein J, Muir A, Clare-Salzer M, She JX, Malone J, Crockett S, Schwartz S, Quattrin T, DeSilva M, Vander Vegt P, Notkins A, Krischer J. Only multiple autoantibodies to islet cells (ICA), insulin, GAD65, IA-2 and IA-2beta predict immune-mediated (Type 1) diabetes in relatives. J Autoimmun 1999; 12:279-287
  22. Randle PJ, Kerbey AL, Espinal J. Mechanisms decreasing glucose oxidation in diabetes and starvation: role of lipid fuels and hormones. Diabetes Metab Rev 1988; 4:623-638
  23. Exton JH. Mechanisms of hormonal regulation of hepatic glucose metabolism. Diabetes Metab Rev 1987; 3:163-183
  24. El-Maghrabi MR, Claus TH, McGrane MM, Pilkis SJ. Influence of phosphorylation on the interaction of effectors with rat liver pyruvate kinase. J Biol Chem 1982; 257:233-240
  25. Pilkis SJ, Granner DK. Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Annu Rev Physiol 1992; 54:885-909
  26. Cryer PE. Minireview: Glucagon in the pathogenesis of hypoglycemia and hyperglycemia in diabetes. Endocrinology 2012; 153:1039-1048
  27. Gerich JE, Langlois M, Noacco C, Karam JH, Forsham PH. Lack of glucagon response to hypoglycemia in diabetes: evidence for an intrinsic pancreatic alpha cell defect. Science 1973; 182:171-173
  28. Unger RH, Cherrington AD. Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover. J Clin Invest 2012; 122:4-12
  29. Kimball SR, Jefferson LS. Cellular mechanisms involved in the action of insulin on protein synthesis. Diabetes Metab Rev 1988; 4:773-787
  30. Kimball SR, Jefferson LS. Regulation of initiation of protein synthesis by insulin in skeletal muscle. Acta Diabetol 1991; 28:134-139
  31. Cahill GF, Jr. Starvation in man. Clin Endocrinol Metab 1976; 5:397-415
  32. McGarry JD. Lilly Lecture 1978. New perspectives in the regulation of ketogenesis. Diabetes 1979; 28:517-523
  33. Wolfsdorf J, Glaser N, Sperling MA, American Diabetes A. Diabetic ketoacidosis in infants, children, and adolescents: A consensus statement from the American Diabetes Association. Diabetes Care 2006; 29:1150-1159
  34. Foster DW, McGarry JD. The metabolic derangements and treatment of diabetic ketoacidosis. N Engl J Med 1983; 309:159-169
  35. Owen OE, Felig P, Morgan AP, Wahren J, Cahill GF, Jr. Liver and kidney metabolism during prolonged starvation. J Clin Invest 1969; 48:574-583
  36. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill GF, Jr. Brain metabolism during fasting. J Clin Invest 1967; 46:1589-1595
  37. Ashby P, Robinson DS. Effects of insulin, glucocorticoids and adrenaline on the activity of rat adipose-tissue lipoprotein lipids. Biochem J 1980; 188:185-192
  38. Parkin SM, Walker K, Ashby P, Robinson DS. Effects of glucose and insulin on the activation of lipoprotin lipase and on protein-synthesis in rat adipose tissue. Biochem J 1980; 188:193-199
  39. Tavangar K, Murata Y, Pedersen ME, Goers JF, Hoffman AR, Kraemer FB. Regulation of lipoprotein lipase in the diabetic rat. J Clin Invest 1992; 90:1672-1678
  40. Todd JA. Genetics of type 1 diabetes. Pathol Biol (Paris) 1997; 45:219-227
  41. She JX. Susceptibility to type I diabetes: HLA-DQ and DR revisited. Immunol Today 1996; 17:323-329
  42. Noble JA, Cavalli AS, Erlich HA. DPB1*5901a: a novel HLA-DPB1 allele from a Caucasian family with insulin-dependent diabetes mellitus. Tissue Antigens 1996; 47:159-162
  43. Pugliese A, Zeller M, Fernandez A, Jr., Zalcberg LJ, Bartlett RJ, Ricordi C, Pietropaolo M, Eisenbarth GS, Bennett ST, Patel DD. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet 1997; 15:293-297
  44. Vafiadis P, Bennett ST, Todd JA, Nadeau J, Grabs R, Goodyer CG, Wickramasinghe S, Colle E, Polychronakos C. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet 1997; 15:289-292
  45. Nerup J, Platz P, Andersen OO, Christy M, Lyngsoe J, Poulsen JE, Ryder LP, Nielsen LS, Thomsen M, Svejgaard A. HL-A antigens and diabetes mellitus. Lancet 1974; 2:864-866
  46. Pociot F. Insulin-dependent diabetes mellitus--a polygenic disorder? Dan Med Bull 1996; 43:216-248
  47. Bell GI, Horita S, Karam JH. A polymorphic locus near the human insulin gene is associated with insulin-dependent diabetes mellitus. Diabetes 1984; 33:176-183
  48. Julier C, Hyer RN, Davies J, Merlin F, Soularue P, Briant L, Cathelineau G, Deschamps I, Rotter JI, Froguel P, et al. Insulin-IGF2 region on chromosome 11p encodes a gene implicated in HLA-DR4-dependent diabetes susceptibility. Nature 1991; 354:155-159
  49. Field LL, Tobias R, Magnus T. A locus on chromosome 15q26 (IDDM3) produces susceptibility to insulin-dependent diabetes mellitus. Nat Genet 1994; 8:189-194
  50. Davies JL, Kawaguchi Y, Bennett ST, Copeman JB, Cordell HJ, Pritchard LE, Reed PW, Gough SC, Jenkins SC, Palmer SM, et al. A genome-wide search for human type 1 diabetes susceptibility genes. Nature 1994; 371:130-136
  51. Hashimoto L, Habita C, Beressi JP, Delepine M, Besse C, Cambon-Thomsen A, Deschamps I, Rotter JI, Djoulah S, James MR, et al. Genetic mapping of a susceptibility locus for insulin-dependent diabetes mellitus on chromosome 11q. Nature 1994; 371:161-164
  52. Delepine M, Pociot F, Habita C, Hashimoto L, Froguel P, Rotter J, Cambon-Thomsen A, Deschamps I, Djoulah S, Weissenbach J, Nerup J, Lathrop M, Julier C. Evidence of a non-MHC susceptibility locus in type I diabetes linked to HLA on chromosome 6. Am J Hum Genet 1997; 60:174-187
  53. Merriman T, Twells R, Merriman M, Eaves I, Cox R, Cucca F, McKinney P, Shield J, Baum D, Bosi E, Pozzilli P, Nistico L, Buzzetti R, Joner G, Ronningen KS, Thorsby E, Undlien D, Pociot F, Nerup J, Bain S, Barnett A, Todd J. Evidence by allelic association-dependent methods for a type 1 diabetes polygene (IDDM6) on chromosome 18q21. Hum Mol Genet 1997; 6:1003-1010
  54. Owerbach D, Gabbay KH. The HOXD8 locus (2q31) is linked to type I diabetes. Interaction with chromosome 6 and 11 disease susceptibility genes. Diabetes 1995; 44:132-136
  55. Mein CA, Esposito L, Dunn MG, Johnson GC, Timms AE, Goy JV, Smith AN, Sebag-Montefiore L, Merriman ME, Wilson AJ, Pritchard LE, Cucca F, Barnett AH, Bain SC, Todd JA. A search for type 1 diabetes susceptibility genes in families from the United Kingdom. Nat Genet 1998; 19:297-300
  56. Reed P, Cucca F, Jenkins S, Merriman M, Wilson A, McKinney P, Bosi E, Joner G, Ronningen KS, Thorsby E, Undlien D, Merriman T, Barnett A, Bain S, Todd J. Evidence for a type 1 diabetes susceptibility locus (IDDM10) on human chromosome 10p11-q11. Hum Mol Genet 1997; 6:1011-1016
  57. Field LL, Tobias R, Thomson G, Plon S. Susceptibility to insulin-dependent diabetes mellitus maps to a locus (IDDM11) on human chromosome 14q24.3-q31. Genomics 1996; 33:1-8
  58. Copeman JB, Cucca F, Hearne CM, Cornall RJ, Reed PW, Ronningen KS, Undlien DE, Nistico L, Buzzetti R, Tosi R, et al. Linkage disequilibrium mapping of a type 1 diabetes susceptibility gene (IDDM7) to chromosome 2q31-q33. Nat Genet 1995; 9:80-85
  59. Morahan G, Huang D, Tait BD, Colman PG, Harrison LC. Markers on distal chromosome 2q linked to insulin-dependent diabetes mellitus. Science 1996; 272:1811-1813
  60. Verge CF, Vardi P, Babu S, Bao F, Erlich HA, Bugawan T, Tiosano D, Yu L, Eisenbarth GS, Fain PR. Evidence for oligogenic inheritance of type 1 diabetes in a large Bedouin Arab family. J Clin Invest 1998; 102:1569-1575
  61. Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, Julier C. EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 2000; 25:406-409
  62. Nerup J, Pociot F, European Consortium for IS. A genomewide scan for type 1-diabetes susceptibility in Scandinavian families: identification of new loci with evidence of interactions. Am J Hum Genet 2001; 69:1301-1313
  63. Concannon P, Gogolin-Ewens KJ, Hinds DA, Wapelhorst B, Morrison VA, Stirling B, Mitra M, Farmer J, Williams SR, Cox NJ, Bell GI, Risch N, Spielman RS. A second-generation screen of the human genome for susceptibility to insulin-dependent diabetes mellitus. Nat Genet 1998; 19:292-296
  64. Cucca F, Goy JV, Kawaguchi Y, Esposito L, Merriman ME, Wilson AJ, Cordell HJ, Bain SC, Todd JA. A male-female bias in type 1 diabetes and linkage to chromosome Xp in MHC HLA-DR3-positive patients. Nat Genet 1998; 19:301-302
  65. Onengut-Gumuscu S, Chen WM, Burren O, Cooper NJ, Quinlan AR, Mychaleckyj JC, Farber E, Bonnie JK, Szpak M, Schofield E, Achuthan P, Guo H, Fortune MD, Stevens H, Walker NM, Ward LD, Kundaje A, Kellis M, Daly MJ, Barrett JC, Cooper JD, Deloukas P, Type 1 Diabetes Genetics C, Todd JA, Wallace C, Concannon P, Rich SS. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet 2015; 47:381-386
  66. Barrett JC, Clayton DG, Concannon P, Akolkar B, Cooper JD, Erlich HA, Julier C, Morahan G, Nerup J, Nierras C, Plagnol V, Pociot F, Schuilenburg H, Smyth DJ, Stevens H, Todd JA, Walker NM, Rich SS, Type 1 Diabetes Genetics C. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet 2009; 41:703-707
  67. Eisenbarth GS. Banting Lecture 2009: An unfinished journey: molecular pathogenesis to prevention of type 1A diabetes. Diabetes 2010; 59:759-774
  68. Krogvold L, Edwin B, Buanes T, Frisk G, Skog O, Anagandula M, Korsgren O, Undlien D, Eike MC, Richardson SJ, Leete P, Morgan NG, Oikarinen S, Oikarinen M, Laiho JE, Hyoty H, Ludvigsson J, Hanssen KF, Dahl-Jorgensen K. Detection of a low-grade enteroviral infection in the islets of langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes 2015; 64:1682-1687
  69. Richardson SJ, Rodriguez-Calvo T, Gerling IC, Mathews CE, Kaddis JS, Russell MA, Zeissler M, Leete P, Krogvold L, Dahl-Jorgensen K, von Herrath M, Pugliese A, Atkinson MA, Morgan NG. Islet cell hyperexpression of HLA class I antigens: a defining feature in type 1 diabetes. Diabetologia 2016; 59:2448-2458
  70. Lundberg M, Krogvold L, Kuric E, Dahl-Jorgensen K, Skog O. Expression of Interferon-Stimulated Genes in Insulitic Pancreatic Islets of Patients Recently Diagnosed With Type 1 Diabetes. Diabetes 2016; 65:3104-3110
  71. Bottazzo GF, Florin-Christensen A, Doniach D. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet 1974; 2:1279-1283
  72. Neufeld M, Maclaren N, Blizzard R. Autoimmune polyglandular syndromes. Pediatr Ann 1980; 9:154-162
  73. Huang W, Connor E, Rosa TD, Muir A, Schatz D, Silverstein J, Crockett S, She JX, Maclaren NK. Although DR3-DQB1*0201 may be associated with multiple component diseases of the autoimmune polyglandular syndromes, the human leukocyte antigen DR4-DQB1*0302 haplotype is implicated only in beta-cell autoimmunity. J Clin Endocrinol Metab 1996; 81:2559-2563
  74. Maclaren NK, Huang SW, Fogh J. Antibody to cultured human insulinoma cells in insulin-dependent diabetes. Lancet 1975; 1:997-1000
  75. Neufeld M, Maclaren NK, Riley WJ, Lezotte D, McLaughlin JV, Silverstein J, Rosenbloom AL. Islet cell and other organ-specific antibodies in U.S. Caucasians and Blacks with insulin-dependent diabetes mellitus. Diabetes 1980; 29:589-592
  76. Palmer JP, Asplin CM, Clemons P, Lyen K, Tatpati O, Raghu PK, Paquette TL. Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science 1983; 222:1337-1339
  77. Atkinson MA, Maclaren NK, Riley WJ, Winter WE, Fisk DD, Spillar RP. Are insulin autoantibodies markers for insulin-dependent diabetes mellitus? Diabetes 1986; 35:894-898
  78. Baekkeskov S, Aanstoot HJ, Christgau S, Reetz A, Solimena M, Cascalho M, Folli F, Richter-Olesen H, De Camilli P. Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature 1990; 347:151-156
  79. Atkinson MA, Kaufman DL, Newman D, Tobin AJ, Maclaren NK. Islet cell cytoplasmic autoantibody reactivity to glutamate decarboxylase in insulin-dependent diabetes. J Clin Invest 1993; 91:350-356
  80. Christie MR, Genovese S, Cassidy D, Bosi E, Brown TJ, Lai M, Bonifacio E, Bottazzo GF. Antibodies to islet 37k antigen, but not to glutamate decarboxylase, discriminate rapid progression to IDDM in endocrine autoimmunity. Diabetes 1994; 43:1254-1259
  81. Lu J, Li Q, Xie H, Chen ZJ, Borovitskaya AE, Maclaren NK, Notkins AL, Lan MS. Identification of a second transmembrane protein tyrosine phosphatase, IA-2beta, as an autoantigen in insulin-dependent diabetes mellitus: precursor of the 37-kDa tryptic fragment. Proc Natl Acad Sci U S A 1996; 93:2307-2311
  82. Solimena M, Dirkx R, Jr., Hermel JM, Pleasic-Williams S, Shapiro JA, Caron L, Rabin DU. ICA 512, an autoantigen of type I diabetes, is an intrinsic membrane protein of neurosecretory granules. EMBO J 1996; 15:2102-2114
  83. Lan MS, Wasserfall C, Maclaren NK, Notkins AL. IA-2, a transmembrane protein of the protein tyrosine phosphatase family, is a major autoantigen in insulin-dependent diabetes mellitus. Proc Natl Acad Sci U S A 1996; 93:6367-6370
  84. Notkins AL, Lu J, Li Q, VanderVegt FP, Wasserfall C, Maclaren NK, Lan MS. IA-2 and IA-2 beta are major autoantigens in IDDM and the precursors of the 40 kDa and 37 kDa tryptic fragments. J Autoimmun 1996; 9:677-682
  85. Lan MS, Maclaren NK. Cryptic epitope and autoimmunity. Diabetes Metab Rev 1998; 14:333-334
  86. Aanstoot HJ, Kang SM, Kim J, Lindsay LA, Roll U, Knip M, Atkinson M, Mose-Larsen P, Fey S, Ludvigsson J, Landin L, Bruining J, Maclaren N, Akerblom HK, Baekkeskov S. Identification and characterization of glima 38, a glycosylated islet cell membrane antigen, which together with GAD65 and IA2 marks the early phases of autoimmune response in type 1 diabetes. J Clin Invest 1996; 97:2772-2783
  87. Bonifacio E, Bingley PJ, Shattock M, Dean BM, Dunger D, Gale EA, Bottazzo GF. Quantification of islet-cell antibodies and prediction of insulin-dependent diabetes. Lancet 1990; 335:147-149
  88. Riley WJ, Atkinson MA, Schatz DA, Maclaren NK. Comparison of islet autoantibodies in 'pre-diabetes' and recommendations for screening. J Autoimmun 1990; 3 Suppl 1:47-51
  89. Krischer JP, Schatz D, Riley WJ, Spillar RP, Silverstein JH, Schwartz S, Malone J, Shah S, Vadheim CM, Rotter JI, et al. Insulin and islet cell autoantibodies as time-dependent covariates in the development of insulin-dependent diabetes: a prospective study in relatives. J Clin Endocrinol Metab 1993; 77:743-749
  90. Irvine WJ, McCallum CJ, Gray RS, Campbell CJ, Duncan LJ, Farquhar JW, Vaughan H, Morris PJ. Pancreatic islet-cell antibodies in diabetes mellitus correlated with the duration and type of diabetes, coexistent autoimmune disease, and HLA type. Diabetes 1977; 26:138-147
  91. Riley WJ, Maclaren NK, Krischer J, Spillar RP, Silverstein JH, Schatz DA, Schwartz S, Malone J, Shah S, Vadheim C, et al. A prospective study of the development of diabetes in relatives of patients with insulin-dependent diabetes. N Engl J Med 1990; 323:1167-1172
  92. Roll U, Christie MR, Fuchtenbusch M, Payton MA, Hawkes CJ, Ziegler AG. Perinatal autoimmunity in offspring of diabetic parents. The German Multicenter BABY-DIAB study: detection of humoral immune responses to islet antigens in early childhood. Diabetes 1996; 45:967-973
  93. Hamalainen AM, Ronkainen MS, Akerblom HK, Knip M. Postnatal elimination of transplacentally acquired disease-associated antibodies in infants born to families with type 1 diabetes. The Finnish TRIGR Study Group. Trial to Reduce IDDM in the Genetically at Risk. J Clin Endocrinol Metab 2000; 85:4249-4253
  94. Koczwara K, Bonifacio E, Ziegler AG. Transmission of maternal islet antibodies and risk of autoimmune diabetes in offspring of mothers with type 1 diabetes. Diabetes 2004; 53:1-4
  95. Ziegler AG, Hummel M, Schenker M, Bonifacio E. Autoantibody appearance and risk for development of childhood diabetes in offspring of parents with type 1 diabetes: the 2-year analysis of the German BABYDIAB Study. Diabetes 1999; 48:460-468
  96. Onkamo P, Vaananen S, Karvonen M, Tuomilehto J. Worldwide increase in incidence of Type I diabetes--the analysis of the data on published incidence trends. Diabetologia 1999; 42:1395-1403
  97. Kumar V, Sercarz E. Induction or protection from experimental autoimmune encephalomyelitis depends on the cytokine secretion profile of TCR peptide-specific regulatory CD4 T cells. J Immunol 1998; 161:6585-6591
  98. Irvine WJ, Clarke BF, Scarth L, Cullen DR, Duncan LJ. Thyroid and gastric autoimmunity in patients with diabetes mellitus. Lancet 1970; 2:163-168
  99. Riley WJ, Maclaren NK, Neufeld M. Adrenal autoantibodies and Addison disease in insulin-dependent diabetes mellitus. J Pediatr 1980; 97:191-195
  100. Riley WJ, Toskes PP, Maclaren NK, Silverstein JH. Predictive value of gastric parietal cell autoantibodies as a marker for gastric and hematologic abnormalities associated with insulin-dependent diabetes. Diabetes 1982; 31:1051-1055
  101. Rapoport MJ, Bistritzer T, Vardi O, Broide E, Azizi A, Vardi P. Increased prevalence of diabetes-related autoantibodies in celiac disease. J Pediatr Gastroenterol Nutr 1996; 23:524-527
  102. Song YH, Connor E, Li Y, Zorovich B, Balducci P, Maclaren N. The role of tyrosinase in autoimmune vitiligo. Lancet 1994; 344:1049-1052
  103. Atkinson MA, Bowman MA, Campbell L, Darrow BL, Kaufman DL, Maclaren NK. Cellular immunity to a determinant common to glutamate decarboxylase and coxsackie virus in insulin-dependent diabetes. J Clin Invest 1994; 94:2125-2129
  104. Ellis TM, Atkinson MA. The clinical significance of an autoimmune response against glutamic acid decarboxylase. Nat Med 1996; 2:148-153
  105. Ellis TM, Schatz DA, Ottendorfer EW, Lan MS, Wasserfall C, Salisbury PJ, She JX, Notkins AL, Maclaren NK, Atkinson MA. The relationship between humoral and cellular immunity to IA-2 in IDDM. Diabetes 1998; 47:566-569
  106. Bach JF, Chatenoud L. Immunosuppression in insulin-dependent diabetes mellitus: from cellular selectivity towards autoantigen specificity. Chem Immunol 1995; 60:32-47
  107. Atkinson MA, Kaufman DL, Campbell L, Gibbs KA, Shah SC, Bu DF, Erlander MG, Tobin AJ, Maclaren NK. Response of peripheral-blood mononuclear cells to glutamate decarboxylase in insulin-dependent diabetes. Lancet 1992; 339:458-459
  108. Harrison LC, Honeyman MC, DeAizpurua HJ, Schmidli RS, Colman PG, Tait BD, Cram DS. Inverse relation between humoral and cellular immunity to glutamic acid decarboxylase in subjects at risk of insulin-dependent diabetes. Lancet 1993; 341:1365-1369
  109. Semana G, Gausling R, Jackson RA, Hafler DA. T cell autoreactivity to proinsulin epitopes in diabetic patients and healthy subjects. J Autoimmun 1999; 12:259-267
  110. Alleva DG, Crowe PD, Jin L, Kwok WW, Ling N, Gottschalk M, Conlon PJ, Gottlieb PA, Putnam AL, Gaur A. A disease-associated cellular immune response in type 1 diabetics to an immunodominant epitope of insulin. J Clin Invest 2001; 107:173-180
  111. Roep BO, Atkinson MA, van Endert PM, Gottlieb PA, Wilson SB, Sachs JA. Autoreactive T cell responses in insulin-dependent (Type 1) diabetes mellitus. Report of the first international workshop for standardization of T cell assays. J Autoimmun 1999; 13:267-282
  112. Endl J, Otto H, Jung G, Dreisbusch B, Donie F, Stahl P, Elbracht R, Schmitz G, Meinl E, Hummel M, Ziegler AG, Wank R, Schendel DJ. Identification of naturally processed T cell epitopes from glutamic acid decarboxylase presented in the context of HLA-DR alleles by T lymphocytes of recent onset IDDM patients. J Clin Invest 1997; 99:2405-2415
  113. Honeyman MC, Stone N, de Aizpurua H, Rowley MJ, Harrison LC. High T cell responses to the glutamic acid decarboxylase (GAD) isoform 67 reflect a hyperimmune state that precedes the onset of insulin-dependent diabetes. J Autoimmun 1997; 10:165-173
  114. Lohmann T, Leslie RD, Hawa M, Geysen M, Rodda S, Londei M. Immunodominant epitopes of glutamic acid decarboxylase 65 and 67 in insulin-dependent diabetes mellitus. Lancet 1994; 343:1607-1608
  115. Lohmann T, Leslie RD, Londei M. T cell clones to epitopes of glutamic acid decarboxylase 65 raised from normal subjects and patients with insulin-dependent diabetes. J Autoimmun 1996; 9:385-389
  116. Panina-Bordignon P, Lang R, van Endert PM, Benazzi E, Felix AM, Pastore RM, Spinas GA, Sinigaglia F. Cytotoxic T cells specific for glutamic acid decarboxylase in autoimmune diabetes. J Exp Med 1995; 181:1923-1927
  117. Weiss U, Manfras BJ, Terjung D, Eiermann T, Wolpl A, Loliger C, Kuhnl P, Boehm BO. In vitro stimulation with glutamic acid decarboxylase (GAD65) leads to an oligoclonal response of peripheral T-cells in an IDDM patient. Scand J Immunol 1995; 42:673-678
  118. Pugliese A, Yang M, Kusmarteva I, Heiple T, Vendrame F, Wasserfall C, Rowe P, Moraski JM, Ball S, Jebson L, Schatz DA, Gianani R, Burke GW, Nierras C, Staeva T, Kaddis JS, Campbell-Thompson M, Atkinson MA. The Juvenile Diabetes Research Foundation Network for Pancreatic Organ Donors with Diabetes (nPOD) Program: goals, operational model and emerging findings. Pediatr Diabetes 2014; 15:1-9
  119. Krogvold L, Skog O, Sundstrom G, Edwin B, Buanes T, Hanssen KF, Ludvigsson J, Grabherr M, Korsgren O, Dahl-Jorgensen K. Function of Isolated Pancreatic Islets From Patients at Onset of Type 1 Diabetes: Insulin Secretion Can Be Restored After Some Days in a Nondiabetogenic Environment In Vitro: Results From the DiViD Study. Diabetes 2015; 64:2506-2512
  120. Campbell-Thompson M. Organ donor specimens: What can they tell us about type 1 diabetes? Pediatr Diabetes 2015; 16:320-330
  121. Michels A, Zhang L, Khadra A, Kushner JA, Redondo MJ, Pietropaolo M. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes 2015; 16:465-484
  122. Karvonen M, Tuomilehto J, Libman I, LaPorte R. A review of the recent epidemiological data on the worldwide incidence of type 1 (insulin-dependent) diabetes mellitus. World Health Organization DIAMOND Project Group. Diabetologia 1993; 36:883-892
  123. Marron MP, Raffel LJ, Garchon HJ, Jacob CO, Serrano-Rios M, Martinez Larrad MT, Teng WP, Park Y, Zhang ZX, Goldstein DR, Tao YW, Beaurain G, Bach JF, Huang HS, Luo DF, Zeidler A, Rotter JI, Yang MC, Modilevsky T, Maclaren NK, She JX. Insulin-dependent diabetes mellitus (IDDM) is associated with CTLA4 polymorphisms in multiple ethnic groups. Hum Mol Genet 1997; 6:1275-1282
  124. See DM, Tilles JG. The pathogenesis of viral-induced diabetes. Clin Diagn Virol 1998; 9:85-88
  125. Vreugdenhil GR, Geluk A, Ottenhoff TH, Melchers WJ, Roep BO, Galama JM. Molecular mimicry in diabetes mellitus: the homologous domain in coxsackie B virus protein 2C and islet autoantigen GAD65 is highly conserved in the coxsackie B-like enteroviruses and binds to the diabetes associated HLA-DR3 molecule. Diabetologia 1998; 41:40-46
  126. Srinivasappa J, Saegusa J, Prabhakar BS, Gentry MK, Buchmeier MJ, Wiktor TJ, Koprowski H, Oldstone MB, Notkins AL. Molecular mimicry: frequency of reactivity of monoclonal antiviral antibodies with normal tissues. J Virol 1986; 57:397-401
  127. Jenson AB, Rosenberg HS, Notkins AL. Pancreatic islet-cell damage in children with fatal viral infections. Lancet 1980; 2:354-358
  128. Yoon JW, Onodera T, Notkins AL. Virus-induced diabetes mellitus. XV. Beta cell damage and insulin-dependent hyperglycemia in mice infected with coxsackie virus B4. J Exp Med 1978; 148:1068-1080
  129. Yoon JW, Selvaggio S, Onodera T, Wheeler J, Jenson AB. Infection of cultured human pancreatic B cells with reovirus type 3. Diabetologia 1981; 20:462-467
  130. Prince GA, Jenson AB, Billups LC, Notkins AL. Infection of human pancreatic beta cell cultures with mumps virus. Nature 1978; 271:158-161
  131. Parkkonen P, Hyoty H, Koskinen L, Leinikki P. Mumps virus infects beta cells in human fetal islet cell cultures upregulating the expression of HLA class I molecules. Diabetologia 1992; 35:63-69
  132. Yoon JW, Onodera T, Jenson AB, Notkins AL. Virus-induced diabetes mellitus. XI. Replication of coxsackie B3 virus in human pancreatic beta cell cultures. Diabetes 1978; 27:778-781
  133. Campbell IL, Harrison LC, Ashcroft RG, Jack I. Reovirus infection enhances expression of class I MHC proteins on human beta-cell and rat RINm5F cell. Diabetes 1988; 37:362-365
  134. Pak CY, Eun HM, McArthur RG, Yoon JW. Association of cytomegalovirus infection with autoimmune type 1 diabetes. Lancet 1988; 2:1-4
  135. Kaufman DL, Erlander MG, Clare-Salzler M, Atkinson MA, Maclaren NK, Tobin AJ. Autoimmunity to two forms of glutamate decarboxylase in insulin-dependent diabetes mellitus. J Clin Invest 1992; 89:283-292
  136. Kaufman DL, Clare-Salzler M, Tian J, Forsthuber T, Ting GS, Robinson P, Atkinson MA, Sercarz EE, Tobin AJ, Lehmann PV. Spontaneous loss of T-cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabetes. Nature 1993; 366:69-72
  137. Tisch R, Yang XD, Singer SM, Liblau RS, Fugger L, McDevitt HO. Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice. Nature 1993; 366:72-75
  138. Tian J, Lehmann PV, Kaufman DL. T cell cross-reactivity between coxsackievirus and glutamate decarboxylase is associated with a murine diabetes susceptibility allele. J Exp Med 1994; 180:1979-1984
  139. Szopa TM, Titchener PA, Portwood ND, Taylor KW. Diabetes mellitus due to viruses--some recent developments. Diabetologia 1993; 36:687-695
  140. Schloot NC, Roep BO, Wegmann DR, Yu L, Wang TB, Eisenbarth GS. T-cell reactivity to GAD65 peptide sequences shared with coxsackie virus protein in recent-onset IDDM, post-onset IDDM patients and control subjects. Diabetologia 1997; 40:332-338
  141. Honeyman MC, Stone NL, Harrison LC. T-cell epitopes in type 1 diabetes autoantigen tyrosine phosphatase IA-2: potential for mimicry with rotavirus and other environmental agents. Mol Med 1998; 4:231-239
  142. Sutkowski N, Palkama T, Ciurli C, Sekaly RP, Thorley-Lawson DA, Huber BT. An Epstein-Barr virus-associated superantigen. J Exp Med 1996; 184:971-980
  143. White J, Herman A, Pullen AM, Kubo R, Kappler JW, Marrack P. The V beta-specific superantigen staphylococcal enterotoxin B: stimulation of mature T cells and clonal deletion in neonatal mice. Cell 1989; 56:27-35
  144. Hao W, Serreze DV, McCulloch DK, Neifing JL, Palmer JP. Insulin (auto)antibodies from human IDDM cross-react with retroviral antigen p73. J Autoimmun 1993; 6:787-798
  145. Conrad B, Weissmahr RN, Boni J, Arcari R, Schupbach J, Mach B. A human endogenous retroviral superantigen as candidate autoimmune gene in type I diabetes. Cell 1997; 90:303-313
  146. Conrad B, Weidmann E, Trucco G, Rudert WA, Behboo R, Ricordi C, Rodriquez-Rilo H, Finegold D, Trucco M. Evidence for superantigen involvement in insulin-dependent diabetes mellitus aetiology. Nature 1994; 371:351-355
  147. Lan MS, Mason A, Coutant R, Chen QY, Vargas A, Rao J, Gomez R, Chalew S, Garry R, Maclaren NK. HERV-K10s and immune-mediated (type 1) diabetes. Cell 1998; 95:14-16; discussion 16
  148. Lehmann PV, Sercarz EE, Forsthuber T, Dayan CM, Gammon G. Determinant spreading and the dynamics of the autoimmune T-cell repertoire. Immunol Today 1993; 14:203-208
  149. Serreze DV, Leiter EH, Kuff EL, Jardieu P, Ishizaka K. Molecular mimicry between insulin and retroviral antigen p73. Development of cross-reactive autoantibodies in sera of NOD and C57BL/KsJ db/db mice. Diabetes 1988; 37:351-358
  150. Naserke HE, Ziegler AG, Lampasona V, Bonifacio E. Early development and spreading of autoantibodies to epitopes of IA-2 and their association with progression to type 1 diabetes. J Immunol 1998; 161:6963-6969
  151. Wilson SB, Kent SC, Patton KT, Orban T, Jackson RA, Exley M, Porcelli S, Schatz DA, Atkinson MA, Balk SP, Strominger JL, Hafler DA. Extreme Th1 bias of invariant Valpha24JalphaQ T cells in type 1 diabetes. Nature 1998; 391:177-181
  152. Kukreja A, Cost G, Marker J, Zhang C, Sun Z, Lin-Su K, Ten S, Sanz M, Exley M, Wilson B, Porcelli S, Maclaren N. Multiple immuno-regulatory defects in type-1 diabetes. J Clin Invest 2002; 109:131-140
  153. Baxter AG, Kinder SJ, Hammond KJ, Scollay R, Godfrey DI. Association between alphabetaTCR+CD4-CD8- T-cell deficiency and IDDM in NOD/Lt mice. Diabetes 1997; 46:572-582
  154. Hammond KJ, Poulton LD, Palmisano LJ, Silveira PA, Godfrey DI, Baxter AG. alpha/beta-T cell receptor (TCR)+CD4-CD8- (NKT) thymocytes prevent insulin-dependent diabetes mellitus in nonobese diabetic (NOD)/Lt mice by the influence of interleukin (IL)-4 and/or IL-10. J Exp Med 1998; 187:1047-1056
  155. Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity 2000; 12:431-440
  156. Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, Herold KC, Lares A, Lee MR, Li K, Liu W, Long SA, Masiello LM, Nguyen V, Putnam AL, Rieck M, Sayre PH, Tang Q. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015; 7:315ra189
  157. Stamatouli AM, Quandt Z, Perdigoto AL, Clark PL, Kluger H, Weiss SA, Gettinger S, Sznol M, Young A, Rushakoff R, Lee J, Bluestone JA, Anderson M, Herold KC. Collateral Damage: Insulin-Dependent Diabetes Induced With Checkpoint Inhibitors. Diabetes 2018; 67:1471-1480
  158. Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 2018; 378:158-168
  159. June CH, Warshauer JT, Bluestone JA. Is autoimmunity the Achilles' heel of cancer immunotherapy? Nat Med 2017; 23:540-547
  160. Bodansky HJ, Staines A, Stephenson C, Haigh D, Cartwright R. Evidence for an environmental effect in the aetiology of insulin dependent diabetes in a transmigratory population. BMJ 1992; 304:1020-1022
  161. Wilberz S, Partke HJ, Dagnaes-Hansen F, Herberg L. Persistent MHV (mouse hepatitis virus) infection reduces the incidence of diabetes mellitus in non-obese diabetic mice. Diabetologia 1991; 34:2-5
  162. Takei I, Asaba Y, Kasatani T, Maruyama T, Watanabe K, Yanagawa T, Saruta T, Ishii T. Suppression of development of diabetes in NOD mice by lactate dehydrogenase virus infection. J Autoimmun 1992; 5:665-673
  163. Martins TC, Aguas AP. Mechanisms of Mycobacterium avium-induced resistance against insulin-dependent diabetes mellitus (IDDM) in non-obese diabetic (NOD) mice: role of Fas and Th1 cells. Clin Exp Immunol 1999; 115:248-254
  164. Oldstone MB. Viruses as therapeutic agents. I. Treatment of nonobese insulin-dependent diabetes mice with virus prevents insulin-dependent diabetes mellitus while maintaining general immune competence. J Exp Med 1990; 171:2077-2089
  165. Cooke A, Tonks P, Jones FM, O'Shea H, Hutchings P, Fulford AJ, Dunne DW. Infection with Schistosoma mansoni prevents insulin dependent diabetes mellitus in non-obese diabetic mice. Parasite Immunol 1999; 21:169-176
  166. Akerblom HK, Knip M. Putative environmental factors in Type 1 diabetes. Diabetes Metab Rev 1998; 14:31-67
  167. Atkinson MA, Eisenbarth GS. Type 1 diabetes: new perspectives on disease pathogenesis and treatment. Lancet 2001; 358:221-229
  168. Dahl-Jorgensen K, Joner G, Hanssen KF. Relationship between cows' milk consumption and incidence of IDDM in childhood. Diabetes Care 1991; 14:1081-1083
  169. Scott FW. Cow milk and insulin-dependent diabetes mellitus: is there a relationship? Am J Clin Nutr 1990; 51:489-491
  170. Patterson CC, Dahlquist G, Soltesz G, Green A, Europe EASG, Diabetes. Is childhood-onset type I diabetes a wealth-related disease? An ecological analysis of European incidence rates. Diabetologia 2001; 44 Suppl 3:B9-16
  171. Fava D, Leslie RD, Pozzilli P. Relationship between dairy product consumption and incidence of IDDM in childhood in Italy. Diabetes Care 1994; 17:1488-1490
  172. Norris JM, Beaty B, Klingensmith G, Yu L, Hoffman M, Chase HP, Erlich HA, Hamman RF, Eisenbarth GS, Rewers M. Lack of association between early exposure to cow's milk protein and beta-cell autoimmunity. Diabetes Autoimmunity Study in the Young (DAISY). JAMA 1996; 276:609-614
  173. Kostraba JN, Gay EC, Rewers M, Hamman RF. Nitrate levels in community drinking waters and risk of IDDM. An ecological analysis. Diabetes Care 1992; 15:1505-1508
  174. Virtanen SM, Jaakkola L, Rasanen L, Ylonen K, Aro A, Lounamaa R, Akerblom HK, Tuomilehto J. Nitrate and nitrite intake and the risk for type 1 diabetes in Finnish children. Childhood Diabetes in Finland Study Group. Diabet Med 1994; 11:656-662
  175. Like AA, Rossini AA. Streptozotocin-induced pancreatic insulitis: new model of diabetes mellitus. Science 1976; 193:415-417
  176. Knip M, Siljander H. The role of the intestinal microbiota in type 1 diabetes mellitus. Nat Rev Endocrinol 2016; 12:154-167
  177. Neu A, Willasch A, Ehehalt S, Kehrer M, Hub R, Ranke MB. Diabetes incidence in children of different nationalities: an epidemiological approach to the pathogenesis of diabetes. Diabetologia 2001; 44 Suppl 3:B21-26
  178. Maclaren NK, Lan MS, Schatz D, Malone J, Notkins AL, Krischer J. Multiple autoantibodies as predictors of Type 1 diabetes in a general population. Diabetologia 2003; 46:873-874
  179. Samuelsson U, Sundkvist G, Borg H, Fernlund P, Ludvigsson J. Islet autoantibodies in the prediction of diabetes in school children. Diabetes Res Clin Pract 2001; 51:51-57
  180. LaGasse JM, Brantley MS, Leech NJ, Rowe RE, Monks S, Palmer JP, Nepom GT, McCulloch DK, Hagopian WA, Washingtno State Diabetes Prediction S. Successful prospective prediction of type 1 diabetes in schoolchildren through multiple defined autoantibodies: an 8-year follow-up of the Washington State Diabetes Prediction Study. Diabetes Care 2002; 25:505-511
  181. Kimpimaki T, Kulmala P, Savola K, Kupila A, Korhonen S, Simell T, Ilonen J, Simell O, Knip M. Natural history of beta-cell autoimmunity in young children with increased genetic susceptibility to type 1 diabetes recruited from the general population. J Clin Endocrinol Metab 2002; 87:4572-4579
  182. Roden M. [Diabetes mellitus: definition, classification and diagnosis]. Wien Klin Wochenschr 2016; 128 Suppl 2:S37-40
  183. Beauchamp G, Haller MJ. Can we prevent type 1 diabetes? Curr Diab Rep 2015; 15:86
  184. De Filippo G, Carel JC, Boitard C, Bougneres PF. Long-term results of early cyclosporin therapy in juvenile IDDM. Diabetes 1996; 45:101-104
  185. Carel JC, Boitard C, Eisenbarth G, Bach JF, Bougneres PF. Cyclosporine delays but does not prevent clinical onset in glucose intolerant pre-type 1 diabetic children. J Autoimmun 1996; 9:739-745
  186. European Nicotinamide Diabetes Intervention Trial G. Intervening before the onset of Type 1 diabetes: baseline data from the European Nicotinamide Diabetes Intervention Trial (ENDIT). Diabetologia 2003; 46:339-346
  187. Lampeter EF, Klinghammer A, Scherbaum WA, Heinze E, Haastert B, Giani G, Kolb H. The Deutsche Nicotinamide Intervention Study: an attempt to prevent type 1 diabetes. DENIS Group. Diabetes 1998; 47:980-984
  188. Lernmark A, Larsson HE. Immune therapy in type 1 diabetes mellitus. Nat Rev Endocrinol 2013; 9:92-103
  189. Muir A, Peck A, Clare-Salzler M, Song YH, Cornelius J, Luchetta R, Krischer J, Maclaren N. Insulin immunization of nonobese diabetic mice induces a protective insulitis characterized by diminished intraislet interferon-gamma transcription. J Clin Invest 1995; 95:628-634
  190. Ramiya VK, Shang XZ, Pharis PG, Wasserfall CH, Stabler TV, Muir AB, Schatz DA, Maclaren NK. Antigen based therapies to prevent diabetes in NOD mice. J Autoimmun 1996; 9:349-356
  191. Daniel D, Wegmann DR. Protection of nonobese diabetic mice from diabetes by intranasal or subcutaneous administration of insulin peptide B-(9-23). Proc Natl Acad Sci U S A 1996; 93:956-960
  192. Group TS, Akerblom HK, Krischer J, Virtanen SM, Berseth C, Becker D, Dupre J, Ilonen J, Trucco M, Savilahti E, Koski K, Pajakkala E, Fransiscus M, Lough G, Bradley B, Koski M, Knip M. The Trial to Reduce IDDM in the Genetically at Risk (TRIGR) study: recruitment, intervention and follow-up. Diabetologia 2011; 54:627-633
  193. Schmid S, Buuck D, Knopff A, Bonifacio E, Ziegler AG. BABYDIET, a feasibility study to prevent the appearance of islet autoantibodies in relatives of patients with Type 1 diabetes by delaying exposure to gluten. Diabetologia 2004; 47:1130-1131
  194. Chase HP BD, Rodriguez H, Donaldson D, Chritton S, Rafkin-Mervis L, Krischer J, Skyler JS, Clare-Salzler M. Type 1 Diabetes TrialNet Nutritional Intervention to Prevent (NIP) Type 1 Diabetes Study Group. Pediatr Diabetes 2015; 16:271-279
  195. Atkinson MA, von Herrath M, Powers AC, Clare-Salzler M. Current concepts on the pathogenesis of type 1 diabetes--considerations for attempts to prevent and reverse the disease. Diabetes Care 2015; 38:979-988
  196. Wicklow BA, Taback SP. Feasibility of a type 1 diabetes primary prevention trial using 2000 IU vitamin D3 in infants from the general population with increased HLA-associated risk. Ann N Y Acad Sci 2006; 1079:310-312
  197. Andersson C, Carlsson A, Cilio C, Cedervall E, Ivarsson SA, Jonsdottir B, Jonsson B, Larsson K, Neiderud J, Lernmark A, Elding Larsson H, Di A-ITSG. Glucose tolerance and beta-cell function in islet autoantibody-positive children recruited to a secondary prevention study. Pediatr Diabetes 2013; 14:341-349
  198. Chaillous L, Lefevre H, Thivolet C, Boitard C, Lahlou N, Atlan-Gepner C, Bouhanick B, Mogenet A, Nicolino M, Carel JC, Lecomte P, Marechaud R, Bougneres P, Charbonnel B, Sai P. Oral insulin administration and residual beta-cell function in recent-onset type 1 diabetes: a multicentre randomised controlled trial. Diabete Insuline Orale group. Lancet 2000; 356:545-549
  199. Castellana M, Cignarelli A, Brescia F, Perrini S, Natalicchio A, Laviola L, Giorgino F. Efficacy and safety of GLP-1 receptor agonists as add-on to SGLT2 inhibitors in type 2 diabetes mellitus: A meta-analysis. Sci Rep 2019; 9:19351
  200. Bonifacio E, Ziegler AG, Klingensmith G, Schober E, Bingley PJ, Rottenkolber M, Theil A, Eugster A, Puff R, Peplow C, Buettner F, Lange K, Hasford J, Achenbach P, Pre PSG. Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes: the Pre-POINT randomized clinical trial. JAMA 2015; 313:1541-1549
  201. Vaarala O, Ilonen J, Ruohtula T, Pesola J, Virtanen SM, Harkonen T, Koski M, Kallioinen H, Tossavainen O, Poussa T, Jarvenpaa AL, Komulainen J, Lounamaa R, Akerblom HK, Knip M. Removal of Bovine Insulin From Cow's Milk Formula and Early Initiation of Beta-Cell Autoimmunity in the FINDIA Pilot Study. Arch Pediatr Adolesc Med 2012; 166:608-614
  202. Herold KC, Bundy BN, Long SA, Bluestone JA, DiMeglio LA, Dufort MJ, Gitelman SE, Gottlieb PA, Krischer JP, Linsley PS, Marks JB, Moore W, Moran A, Rodriguez H, Russell WE, Schatz D, Skyler JS, Tsalikian E, Wherrett DK, Ziegler AG, Greenbaum CJ, Type 1 Diabetes TrialNet Study G. An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med 2019; 381:603-613
  203. Quattrin T, Haller MJ, Steck AK, Felner EI, Li Y, Xia Y, Leu JH, Zoka R, Hedrick JA, Rigby MR, Vercruysse F, Investigators TGS. Golimumab and Beta-Cell Function in Youth with New-Onset Type 1 Diabetes. N Engl J Med 2020; 383:2007-2017
  204. Shah AS, D'Alessio D, Ford-Adams ME, Desai AP, Inge TH. Bariatric Surgery: A Potential Treatment for Type 2 Diabetes in Youth. Diabetes Care 2016; 39:934-940
  205. Dagogo-Jack S. Predicting diabetes: our relentless quest for genomic nuggets. Diabetes Care 2012; 35:193-195
  206. Writing Group for the SfDiYSG, Dabelea D, Bell RA, D'Agostino RB, Jr., Imperatore G, Johansen JM, Linder B, Liu LL, Loots B, Marcovina S, Mayer-Davis EJ, Pettitt DJ, Waitzfelder B. Incidence of diabetes in youth in the United States. JAMA 2007; 297:2716-2724
  207. DeFronzo RA, Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care 1991; 14:173-194
  208. Hales CN, Barker DJ. The thrifty phenotype hypothesis. Br Med Bull 2001; 60:5-20
  209. Pettitt DJ, Aleck KA, Baird HR, Carraher MJ, Bennett PH, Knowler WC. Congenital susceptibility to NIDDM. Role of intrauterine environment. Diabetes 1988; 37:622-628
  210. Type 2 diabetes in children and adolescents. American Diabetes Association. Diabetes Care 2000; 23:381-389
  211. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 2007; 316:1341-1345
  212. Reaven GM. Role of insulin resistance in human disease (syndrome X): an expanded definition. Annu Rev Med 1993; 44:121-131
  213. Kahn CR, Flier JS, Bar RS, Archer JA, Gorden P, Martin MM, Roth J. The syndromes of insulin resistance and acanthosis nigricans. Insulin-receptor disorders in man. N Engl J Med 1976; 294:739-745
  214. Ten S, Bhangoo A, Ramchandani N, Mueller C, Vogiatzi M, New M, Lesser M, Maclaren N. Characterization of insulin resistance syndrome in children and young adults. When to screen for prediabetes? J Pediatr Endocrinol Metab 2007; 20:989-999
  215. Chadt A, Scherneck S, Joost HG, Al-Hasani H. Molecular links between Obesity and Diabetes: "Diabesity". In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, eds. Endotext. South Dartmouth (MA)2000.
  216. Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, Wagner AJ, DePaoli AM, Reitman ML, Taylor SI, Gorden P, Garg A. Leptin-replacement therapy for lipodystrophy. N Engl J Med 2002; 346:570-578
  217. Sperling MA, Garg A. Monogenic Forms of Diabetes. In: rd, Cowie CC, Casagrande SS, Menke A, Cissell MA, Eberhardt MS, Meigs JB, Gregg EW, Knowler WC, Barrett-Connor E, Becker DJ, Brancati FL, Boyko EJ, Herman WH, Howard BV, Narayan KMV, Rewers M, Fradkin JE, eds. Diabetes in America. Bethesda (MD)2018.
  218. Laffel L, Chang N, Grey M, Hale D, Higgins L, Hirst K, Izquierdo R, Larkin M, Macha C, Pham T, Wauters A, Weinstock RS, Group TS. Metformin monotherapy in youth with recent onset type 2 diabetes: experience from the prerandomization run-in phase of the TODAY study. Pediatr Diabetes 2012; 13:369-375
  219. Allen DB. TODAY--a stark glimpse of tomorrow. N Engl J Med 2012; 366:2315-2316
  220. Galloway PJ, Donaldson MD, Wallace AM. Sex hormone binding globulin concentration as a prepubertal marker for hyperinsulinaemia in obesity. Arch Dis Child 2001; 85:489-491
  221. Karnieli E, Cohen P, Barzilai N, Ish-Shalom Z, Armoni M, Rafaelov R, Barzilai D. Insulin resistance in Cushing's syndrome. Horm Metab Res 1985; 17:518-521
  222. Low L, Chernausek SD, Sperling MA. Acromegaloid patients with type A insulin resistance: parallel defects in insulin and insulin-like growth factor-I receptors and biological responses in cultured fibroblasts. J Clin Endocrinol Metab 1989; 69:329-337
  223. Rogers DL. Acanthosis nigricans. Semin Dermatol 1991; 10:160-163
  224. Tominaga K, Kurata JH, Chen YK, Fujimoto E, Miyagawa S, Abe I, Kusano Y. Prevalence of fatty liver in Japanese children and relationship to obesity. An epidemiological ultrasonographic survey. Dig Dis Sci 1995; 40:2002-2009
  225. Kawasaki T, Hashimoto N, Kikuchi T, Takahashi H, Uchiyama M. The relationship between fatty liver and hyperinsulinemia in obese Japanese children. J Pediatr Gastroenterol Nutr 1997; 24:317-321
  226. Tazawa Y, Noguchi H, Nishinomiya F, Takada G. Serum alanine aminotransferase activity in obese children. Acta Paediatr 1997; 86:238-241
  227. Marchesini G, Brizi M, Bianchi G, Tomassetti S, Zoli M, Melchionda N. Metformin in non-alcoholic steatohepatitis. Lancet 2001; 358:893-894
  228. Fan Y, Fang X, Tajima A, Geng X, Ranganathan S, Dong H, Trucco M, Sperling MA. Evolution of hepatic steatosis to fibrosis and adenoma formation in liver-specific growth hormone receptor knockout mice. Front Endocrinol (Lausanne) 2014; 5:218
  229. Reinehr T, Isa A, de Sousa G, Dieffenbach R, Andler W. Thyroid hormones and their relation to weight status. Horm Res 2008; 70:51-57
  230. Reinehr T. Obesity and thyroid function. Mol Cell Endocrinol 2010; 316:165-171
  231. Reinehr T. Thyroid function in the nutritionally obese child and adolescent. Curr Opin Pediatr 2011; 23:415-420
  232. Sperling MA GA. Monogenic Forms of Diabetes. Diabetes in America. 3rd ed2016:7-1-7-27.
  233. Fajans SS, Bell GI. MODY: history, genetics, pathophysiology, and clinical decision making. Diabetes Care 2011; 34:1878-1884
  234. Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 2001; 345:971-980
  235. Pihoker C, Gilliam LK, Ellard S, Dabelea D, Davis C, Dolan LM, Greenbaum CJ, Imperatore G, Lawrence JM, Marcovina SM, Mayer-Davis E, Rodriguez BL, Steck AK, Williams DE, Hattersley AT, Group SfDiYS. Prevalence, characteristics and clinical diagnosis of maturity onset diabetes of the young due to mutations in HNF1A, HNF4A, and glucokinase: results from the SEARCH for Diabetes in Youth. J Clin Endocrinol Metab 2013; 98:4055-4062
  236. Ellard S, Bellanne-Chantelot C, Hattersley AT, European Molecular Genetics Quality Network Mg. Best practice guidelines for the molecular genetic diagnosis of maturity-onset diabetes of the young. Diabetologia 2008; 51:546-553
  237. Dussoix P, Vaxillaire M, Iynedjian PB, Tiercy JM, Ruiz J, Spinas GA, Berger W, Zahnd G, Froguel P, Philippe J. Diagnostic heterogeneity of diabetes in lean young adults: classification based on immunological and genetic parameters. Diabetes 1997; 46:622-631
  238. Tuomi T, Miettinen PJ, Hakaste L, Groop L. Atypical Forms of Diabetes. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, eds. Endotext. South Dartmouth (MA)2000.
  239. Shields BM, Hicks S, Shepherd MH, Colclough K, Hattersley AT, Ellard S. Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 2010; 53:2504-2508
  240. Chakera AJ, Steele AM, Gloyn AL, Shepherd MH, Shields B, Ellard S, Hattersley AT. Recognition and Management of Individuals With Hyperglycemia Because of a Heterozygous Glucokinase Mutation. Diabetes Care 2015; 38:1383-1392
  241. Hattersley AT, Greeley SAW, Polak M, Rubio-Cabezas O, Njolstad PR, Mlynarski W, Castano L, Carlsson A, Raile K, Chi DV, Ellard S, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2018: The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes 2018; 19 Suppl 27:47-63
  242. Yamagata K, Furuta H, Oda N, Kaisaki PJ, Menzel S, Cox NJ, Fajans SS, Signorini S, Stoffel M, Bell GI. Mutations in the hepatocyte nuclear factor-4alpha gene in maturity-onset diabetes of the young (MODY1). Nature 1996; 384:458-460
  243. Froguel P, Zouali H, Vionnet N, Velho G, Vaxillaire M, Sun F, Lesage S, Stoffel M, Takeda J, Passa P, et al. Familial hyperglycemia due to mutations in glucokinase. Definition of a subtype of diabetes mellitus. N Engl J Med 1993; 328:697-702
  244. Hansen T, Eiberg H, Rouard M, Vaxillaire M, Moller AM, Rasmussen SK, Fridberg M, Urhammer SA, Holst JJ, Almind K, Echwald SM, Hansen L, Bell GI, Pedersen O. Novel MODY3 mutations in the hepatocyte nuclear factor-1alpha gene: evidence for a hyperexcitability of pancreatic beta-cells to intravenous secretagogues in a glucose-tolerant carrier of a P447L mutation. Diabetes 1997; 46:726-730
  245. Peters AL, Davidson MB, Schriger DL, Hasselblad V. A clinical approach for the diagnosis of diabetes mellitus: an analysis using glycosylated hemoglobin levels. Meta-analysis Research Group on the Diagnosis of Diabetes Using Glycated Hemoglobin Levels. JAMA 1996; 276:1246-1252
  246. Pontoglio M, Prie D, Cheret C, Doyen A, Leroy C, Froguel P, Velho G, Yaniv M, Friedlander G. HNF1alpha controls renal glucose reabsorption in mouse and man. EMBO Rep 2000; 1:359-365
  247. Stoffers DA, Ferrer J, Clarke WL, Habener JF. Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat Genet 1997; 17:138-139
  248. Horikawa Y, Iwasaki N, Hara M, Furuta H, Hinokio Y, Cockburn BN, Lindner T, Yamagata K, Ogata M, Tomonaga O, Kuroki H, Kasahara T, Iwamoto Y, Bell GI. Mutation in hepatocyte nuclear factor-1 beta gene (TCF2) associated with MODY. Nat Genet 1997; 17:384-385
  249. Naya FJ, Huang HP, Qiu Y, Mutoh H, DeMayo FJ, Leiter AB, Tsai MJ. Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/neuroD-deficient mice. Genes Dev 1997; 11:2323-2334
  250. Neve B, Fernandez-Zapico ME, Ashkenazi-Katalan V, Dina C, Hamid YH, Joly E, Vaillant E, Benmezroua Y, Durand E, Bakaher N, Delannoy V, Vaxillaire M, Cook T, Dallinga-Thie GM, Jansen H, Charles MA, Clement K, Galan P, Hercberg S, Helbecque N, Charpentier G, Prentki M, Hansen T, Pedersen O, Urrutia R, Melloul D, Froguel P. Role of transcription factor KLF11 and its diabetes-associated gene variants in pancreatic beta cell function. Proc Natl Acad Sci U S A 2005; 102:4807-4812
  251. Raeder H, Johansson S, Holm PI, Haldorsen IS, Mas E, Sbarra V, Nermoen I, Eide SA, Grevle L, Bjorkhaug L, Sagen JV, Aksnes L, Sovik O, Lombardo D, Molven A, Njolstad PR. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction. Nat Genet 2006; 38:54-62
  252. St-Onge L, Sosa-Pineda B, Chowdhury K, Mansouri A, Gruss P. Pax6 is required for differentiation of glucagon-producing alpha-cells in mouse pancreas. Nature 1997; 387:406-409
  253. Edghill EL, Flanagan SE, Patch AM, Boustred C, Parrish A, Shields B, Shepherd MH, Hussain K, Kapoor RR, Malecki M, MacDonald MJ, Stoy J, Steiner DF, Philipson LH, Bell GI, Neonatal Diabetes International Collaborative G, Hattersley AT, Ellard S. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes 2008; 57:1034-1042
  254. Borowiec M, Liew CW, Thompson R, Boonyasrisawat W, Hu J, Mlynarski WM, El Khattabi I, Kim SH, Marselli L, Rich SS, Krolewski AS, Bonner-Weir S, Sharma A, Sale M, Mychaleckyj JC, Kulkarni RN, Doria A. Mutations at the BLK locus linked to maturity onset diabetes of the young and beta-cell dysfunction. Proc Natl Acad Sci U S A 2009; 106:14460-14465
  255. Bonnefond A, Froguel P. Rare and common genetic events in type 2 diabetes: what should biologists know? Cell Metab 2015; 21:357-368
  256. Hattersley AT, Patel KA. Precision diabetes: learning from monogenic diabetes. Diabetologia 2017; 60:769-777
  257. Prudente S, Jungtrakoon P, Marucci A, Ludovico O, Buranasupkajorn P, Mazza T, Hastings T, Milano T, Morini E, Mercuri L, Bailetti D, Mendonca C, Alberico F, Basile G, Romani M, Miccinilli E, Pizzuti A, Carella M, Barbetti F, Pascarella S, Marchetti P, Trischitta V, Di Paola R, Doria A. Loss-of-Function Mutations in APPL1 in Familial Diabetes Mellitus. Am J Hum Genet 2015; 97:177-185
  258. De Franco E, Flanagan SE, Houghton JA, Lango Allen H, Mackay DJ, Temple IK, Ellard S, Hattersley AT. The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study. Lancet 2015; 386:957-963
  259. Pearson ER, Flechtner I, Njolstad PR, Malecki MT, Flanagan SE, Larkin B, Ashcroft FM, Klimes I, Codner E, Iotova V, Slingerland AS, Shield J, Robert JJ, Holst JJ, Clark PM, Ellard S, Sovik O, Polak M, Hattersley AT, Neonatal Diabetes International Collaborative G. Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 2006; 355:467-477
  260. Johns DR. Seminars in medicine of the Beth Israel Hospital, Boston. Mitochondrial DNA and disease. N Engl J Med 1995; 333:638-644
  261. Robbins DC, Shoelson SE, Rubenstein AH, Tager HS. Familial hyperproinsulinemia. Two cohorts secreting indistinguishable type II intermediates of proinsulin conversion. J Clin Invest 1984; 73:714-719
  262. Nishi M, Nanjo K. Insulin gene mutations and diabetes. J Diabetes Investig 2011; 2:92-100
  263. Hameed S, Jaffe A, Verge CF. Advances in the detection and management of cystic fibrosis related diabetes. Curr Opin Pediatr 2015; 27:525-533
  264. Ren H, Qin L, Wang W, Ma J, Zhang W, Shen PY, Shi H, Li X, Chen N. Abnormal glucose metabolism and insulin sensitivity in Chinese patients with Gitelman syndrome. Am J Nephrol 2013; 37:152-157
  265. Boyle P. Cushing's disease, glucocorticoid excess, glucocorticoid deficiency and diabetes. Diabetes Review 1993; 1
  266. Ganda OP. Growth hormone, acromegaly and diabetes. Diabetes Reviews 1993; 1
  267. Wass JA, Cudworth AG, Bottazzo GF, Woodrow JC, Besser GM. An assessment of glucose intolerance in acromegaly and its response to medical treatment. Clin Endocrinol (Oxf) 1980; 12:53-59
  268. Cryer P. Catecholamines, pheochromocytomas and diabetes. Diabetes Review 1993; 1
  269. Stenstrom G, Sjostrom L, Smith U. Diabetes mellitus in phaeochromocytoma. Fasting blood glucose levels before and after surgery in 60 patients with phaeochromocytoma. Acta Endocrinol (Copenh) 1984; 106:511-515
  270. Boden G. RJ CX. Glucagonoma syndrome, glucagon and glucose tolerance. Diabetes Review 1993; 1
  271. Vinik A, Feliberti E, Perry RR. Glucagonoma Syndrome. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A, eds. Endotext. South Dartmouth (MA)2000.
  272. Andersen OO, Friis T, Ottesen B. Glucose tolerance and insulin secretion in hyperthyroidism. Acta Endocrinol (Copenh) 1977; 84:576-587
  273. Repaske DR. Medication-induced diabetes mellitus. Pediatr Diabetes 2016; 17:392-397
  274. Houston MC. The effects of antihypertensive drugs on glucose intolerance in hypertensive nondiabetics and diabetics. Am Heart J 1988; 115:640-656
  275. Carr A, Samaras K, Burton S, Law M, Freund J, Chisholm DJ, Cooper DA. A syndrome of peripheral lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS 1998; 12:F51-58
  276. Johnson C, Ahsan N, Gonwa T, Halloran P, Stegall M, Hardy M, Metzger R, Shield C, 3rd, Rocher L, Scandling J, Sorensen J, Mulloy L, Light J, Corwin C, Danovitch G, Wachs M, van Veldhuisen P, Salm K, Tolzman D, Fitzsimmons WE. Randomized trial of tacrolimus (Prograf) in combination with azathioprine or mycophenolate mofetil versus cyclosporine (Neoral) with mycophenolate mofetil after cadaveric kidney transplantation. Transplantation 2000; 69:834-841
  277. First MR, Gerber DA, Hariharan S, Kaufman DB, Shapiro R. Posttransplant diabetes mellitus in kidney allograft recipients: incidence, risk factors, and management. Transplantation 2002; 73:379-386
  278. Jarrett RJ. Why is insulin not a risk factor for coronary heart disease? Diabetologia 1994; 37:945-947
  279. Reaven GM, Chen YD. Role of insulin in regulation of lipoprotein metabolism in diabetes. Diabetes Metab Rev 1988; 4:639-652
  280. Helfgott SM. Stiff-man syndrome: from the bedside to the bench. Arthritis Rheum 1999; 42:1312-1320
  281. Taylor SI. Lilly Lecture: molecular mechanisms of insulin resistance. Lessons from patients with mutations in the insulin-receptor gene. Diabetes 1992; 41:1473-1490
  282. Zhou P, Ten S, Sinha S, Ramchandani N, Vogiatzi M, Maclaren N. Insulin receptor autoimmunity and insulin resistance. J Pediatr Endocrinol Metab 2008; 21:369-375

 

 

Sexual Dysfunction in Diabetes

ABSTRACT

 

Diabetes is an increasingly prevalent problem that has been associated very strongly with sexual problems in both men and women.  Diabetes has numerous end organ effects and also exerts a substantial psychological toll which may predispose diabetic people to sexual problems.  Erectile Dysfunction (ED) is common in men with diabetes; these men tend to present with more severe and refractory ED compared to non-diabetic peers.  While ED is the best-established diabetes-related sexual dysfunction, ejaculatory and sexual desires issues may also occur in men with diabetes.  Women with diabetes are also at risk for sexual dysfunction.  Sexual health inquiry is an important aspect of diabetes care. Importantly, lifestyle change and close management of diabetes has been associated with improvements in sexual function.

INTRODUCTION

Diabetes mellitus (DM) may lead to disruption of normal sexual function in both men and women via diabetic-induced end organ damage and psychological stress.  There is a strong association between diabetes and erectile dysfunction (ED) in men; ED is the best studied sexual dysfunction but the sexual health ramifications of diabetes extend well beyond erectile pathophysiology. In the Endotext chapter on Male Endocrinology “Medical and Surgical Therapy of Erectile Dysfunction”, Shindel, et al review the pathophysiology, work-up, and treatments for erectile dysfunction of any cause. In this chapter, we will focus specifically on sexual dysfunction in people with diabetes, with particular emphasis on practical information for clinicians.

 

EPIDEMIOLOGY

 

Sexual dysfunction is a common problem that is particularly prevalent in men and women with diabetes.  The presence of sexual dysfunction in type I diabetes has been associated with markedly lower quality of life and psychological distress (1). While, the incidence of sexual problems increases with age (particularly in men but also in women), this is driven primarily by comorbid conditions associated with aging. Examples include smoking, heart disease, high blood pressure, high cholesterol, and diabetes (2). The prevalence of ED in men with diabetes is approximately three and a half times higher than in the general population (3,4). ED may also be the presenting symptom for DM and may predict later neurologic sequelae (5).

 

PATHOGENESIS

 

The pathophysiology of ED in DM is multifactorial, consisting of both vascular, hormonal, and neurologic insults (6). Diabetic neuropathy may impair autonomic and somatic nerve processes essential for erections. Diabetes is also associated with impaired relaxation of cavernosal smooth muscle due to endothelial-derived nitric oxide induced by  glycosylation products (7-8).  A variety of serum markers (e.g., E-selectin, Interleukin-10, reactive oxygen species) have been linked to diabetes-related ED. The clinical utility of these remains ambiguous but they may have future utility as biomarkers for incipient ED pending further study (9).

 

New evidence has suggested that men with diabetes may also be at increased risk of low serum testosterone levels (10,11). The etiology of low T in diabetic men remains unclear but may be secondary to a decline in the levels of pituitary hormones responsible for stimulating testicular production of testosterone (12). Low levels of testosterone may lead to a decline in sexual desire and, directly or indirectly, to ED (13).

 

Men with diabetes should be screened for the presence of low testosterone by checking serum total testosterone. Sex hormone binding globulin and albumin may also be tested to permit assessment for free and bioavailable testosterone (14). The clinical utility of free and bioavailable testosterone remains controversial. The most recent guidelines on testosterone issued by the American Urological Association do not recommend use of free or bioavailable testosterone in clinical decision making (10). The most recent Endocrine Society Guideline states that free/bioavailable testosterone may be worth assessing (via equilibrium dialysis or an accurate estimator) in men with symptoms and low-normal total testosterone (14).

 

Testing for hypogonadism should be performed in the morning hours (between 8 and 11 AM) when serum testosterone is highest (14). The appropriate assay and biochemical cut-off values for “low” testosterone are controversial; generally speaking, symptoms of hypogonadism are progressively more common in men with total testosterone levels less than 320 ng/dL and free testosterone levels lower than 64 pg/mL (15). When assessing a patient with a single report of low testosterone, providers should consider confirmatory testing to include repeat testosterone as well as pituitary hormones (FSH, LH, and prolactin) to rule out central causes of hypogonadism (10,14). Only those patients with biochemically low testosterone AND symptoms potentially referable to hypogonadism (decreased libido, ED, fatigue, decreased bone mineral density, depressed mood, etc.) in which alternative etiologies for symptoms are not readily apparent should be considered for treatment (14).  

 

TREATMENT OF ED WITH PHOSPHODIESTERASE TYPE 5 INHIBITORS (PDE5I)

 

The treatment of ED in general was revolutionized by the introduction of the PDE5 inhibitor (PDE5I) class of medications. The first of PDE5I to obtain United States Food and Drug Administration (FDA) approval was of sildenafil (Viagra®), followed by vardenafil (Levitra®/Staxyn®), tadalafil (Cialis®), and avanafil (Stendra®).

 

All PDE5I are dependent on function of the NO/cGMP pathway. Sexual stimulation provokes the release of nitric oxide (NO) from cavernous nerves and endothelial cells. NO leads to activation of guanylate cyclase, which catalyzes the transformation of GTP to cyclic guanosine monophosphate (cGMP). By a variety of downstream mechanisms, cGMP triggers decreased intracellular calcium with subsequent relaxation of actin/myosin cross bridges and penile smooth muscle relaxation. cGMP is deactivated by conversion to 5 prime guanosine monophosphate, a process mediated by phosphodiesterase type 5 (PDE5)- the predominant functional PDE type found in the penis (16). 

 

PDE5I block the inactivation of cGMP, leading to persistently elevated levels of cGMP and continued smooth muscle relaxation(16). Since the release of NO is mediated by both neuronal and endothelial Nitric Oxide Synthase (NOS), neuropathy and endothelial disease (as may occur with diabetes) blunts the efficacy of PDE5I. This is confirmed clinically as men with diabetes have a poorer response overall to PDE5I than men with ED of other etiologies.

 

A prospective, multi-center, randomized, controlled, double-blinded (RCDB) trial of vardenafil in men with diabetes was carried out by Goldstein, et al (17). The study consisted of 430 men with chronic ED, a hemoglobin A1c (HbA1c) of <12%, and no other serious confounding causes of ED (e.g., radical pelvic surgery, spinal cord injury, etc.). Additionally, patients were excluded if they had unstable coronary disease or other contraindications to PDE5I use. The patients were evaluated using the erectile function (EF) domain of the 15 item International Index of Erectile Function (IIEF), 2 diary questions regarding the patient’s ability to penetrate (SEP2) and have successful intercourse (SEP3), and a global assessment question (GAQ) about whether or not the treatment had improved their erections. There were statistically and clinically significant improvements in all of the evaluated endpoints, with most of the improvements demonstrating a dose-relation. With 20 mg of vardenafil, the EF score was 19 (out of a total possible of 25) and 54% of men were able to complete intercourse, with an overall responder rate (as measured by the GAQ) of 72%. The effect was attenuated in patients with severe underlying ED but improvement remained significant. There was no correlation noted between different strata of HgA1c levels. The drug was well-tolerated with few patients discontinuing the study due to adverse side-effects.

 

A similar RCDB trial of tadalafil in men with diabetes was performed by Saenz de Tejada, et al (18). A total of 191 patients completed this study; evaluated parameters were very similar to the vardenafil study above. Exclusion criteria were also similar to the vardenafil study, except that patient with hypertension and hypercholesterolemia were also excluded in the tadalafil study. As in the vardenafil study, statistically and clinically significant improvements were noted in all of the evaluated parameters for men using tadalafil, regardless of severity of underlying DM or level of HgA1c, with an overall responder rate (as assessed by GAQ) of 64% by those using 20 mg. The drug was also well-tolerated with few discontinuations.

 

A unique study from Denmark attempted to assess the “real-life” use of sildenafil in men with diabetes and ED in terms of how many patients wanted to try an agent, how many were eligible to do so, and how efficacious the medicine was (19).  Examining a population of 326 men seen in an outpatient diabetes clinic, 192 (59%) self-reported ED and 187 of these were over 40 years old. Of these 187 patients, 79 (42%) were excluded because of medical or pharmacologic contraindications to sildenafil use. A further 63 patients either declined to participate in the study or did not respond. This left 45 patients for the study (23% of those patients with self-reported ED). Of these, 10 dropped out due to lack of sexual partner and 2 others without recorded reason. Sixty-one percent of the remaining patients self-titrated to a maximum dose of 100 mg. Of the 33 patients remaining, 36% noted consistent improvement, 27% noted variable improvement, and 36% felt they had no improvement; overall, 54% felt that the medicine had met their expectations. Essentially, just 18 of 187 (9.6%) men over age 40 with DM and ED felt that the medicine met their expectations. This real-world experience should inform conversations regarding PDE5i efficacy in men with DM and ED.

 

In 2008 the US Food and Drug Administration (FDA) approved low-dose (2.5-5 mg) tadalafil as a daily treatment for ED.  Hatzichristou et al. enrolled 298 men with diabetes (89% type 2) and ED in a RCDB lasting 12 weeks and assessed clinical response using the sexual encounter profile questions 2 and 3.  At baseline 38%, 42%, and 32% of men reported the ability to attain an erection sufficient for vaginal penetration (SEP2) in the placebo, 2.5 mg, and 5 mg groups, respectively. The percentages of men in the same groups able to maintain erection until the completion of satisfactory intercourse (SEP3) were 20%, 20% and 16%, respectively. At the completion of the study, men treated with either the 2.5 mg or 5 mg dose of tadalafil manifested greater improvements in SEP 2 (increase from baseline of 5%, 20%, and 29%) and SEP3 (28%, 46%, 41%).  The lower success rate in the 5 mg group was likely accounted for by relatively worse diabetic disease at baseline in that group. Patients treated with tadalafil reported improvements in erection (based on IIEF scores) irrespective of baseline IIEF scores. Patients were significantly more likely to prefer tadalafil treatment compared to placebo (20).

 

In addition to daily dosing as an alternative to on-demand dosing for PDE5I, there has been great interest in recent years in the use of PDE5I not just as a therapy to produce erections but as a means to halt or even reverse the penile tissue damage that leads to ED. Studies in animals with a form of experimentally induced diabetes most similar to diabetes mellitus type 1 have demonstrated enhancement of erectile function and preservation of penile tissue health when treated with either vardenafil or SK-3530 (a novel PDE5I that has not yet been approved for routine in humans) (21,22).  A preliminary study of routine dose sildenafil vs. placebo for 4 weeks in 292 men with type 2 diabetes and ED revealed some improvements in blood tests used to measure oxidative stress in men treated with sildenafil. Unfortunately, there were some differences between the placebo and sildenafil group at baseline and there were no significant erectile function differences after the 4-week course of daily treatment was completed (23). Another study in 20 men with type 2 diabetes but no ED indicated that treatment with sildenafil 25 mg three times a day led to improved vascular function and a decline in blood markers for various types of inflammation and oxidative stress.  The ultimate clinical relevance of these findings is unclear (24). 

 

These encouraging preliminary results will require further assessment before the routine use of PDE5I for reversal of tissue damage can be recommended routinely. A degree of caution is required since, despite a series of encouraging pre-clinical animal studies, routine dose PDE5I for the management of ED related to pelvic surgery has not been proven beneficial for recovery of spontaneous erection responses (25,26).

 

TREATMENT OF ED WITH OTHER MODALITIES

 

Direct administration of vasodilators to the erectile tissue of the penis is a well-established modality for management of ED dating back more than three decades. Commonly used agents include papaverine, phentolamine, and prostaglandin E-1 (PgE1) (27). These agents are often used as combinations (e.g., bimix or trimix) to reduce the adverse effects of each specific agent. 

 

Only PgE-1 has received formal FDA approval for management of ED. Intracavernosal PgE1 injection therapy in men with diabetes and ED was evaluated in a large, multicenter trial by Heaton, et al (28). Over 300 men entered the trial; 83% completed the titration period and proceeding to home use. Of those patients using the medication at home, 79% required 30 micrograms/dose or less, and 72% remained satisfied with the initial dose during the follow-up period (6 months). There were 2 instances of priapism (sustained erection of greater than 4 hours unaccompanied by sexual stimuli) neither of which required intervention, 1 patient developed a penile nodule, and 24% of patients reported penile pain with injection; the pain led to patient drop-out in 5% of the treatment group. A smaller, more recent study with longer follow-up (10 years) found that men with diabetes and ED using penile injections tended to shift towards decreased frequency of use but preferred stronger agents (mixtures of alprostadil with papaverine and/or phentolamine), with men with type 1 diabetes and ED stabilizing their doses within 5 years and men with type 2 diabetes and ED stabilizing within 9-10 years (29).

 

Prostaglandin may also be administered via an intraurethral route; the Medicated Urethral Suppository for Erections (MUSE®) is a urethral prostaglandin suppository.  This treatment has FDA approval and has been used with some success by men with ED.  Side effects include urethral burning, pain, and irritation of the sexual partner’s mucous membranes (30).

 

In patients for whom injection or intraurethral therapy does not work vacuum erection devices (VED) may be useful. There is a paucity of data specifically evaluating the use of VED in men with diabetes and ED but the drop-out rate for patients is generally quite high, even for patients who are able to achieve a rigid erection with the device. One subset analysis found that despite a good response (i.e., firm erection) using VED, only 50% of those couples found the treatment to be satisfactory. This may be due to difficult operating the device and/or a feeling that it is a cumbersome interruption of sexual activity.  Possible local side effects include petechiae (small red dots from broken capillaries), a feeling of having a cold penis, and abnormal sensation of ejaculation (31). Many men also report that their erectile rigidity is sub-optimal with the VED.

 

PENILE PROSTHETICS

 

Penile prostheses are an excellent option for diabetic men with ED refractory to medical management and/or those who cannot tolerate medical management of ED. Prosthesis surgery is irreversible in that the corporal tissue is permanently altered; if the prosthesis is removed without replacement complete ED will almost certainly result. While a variety of exotic materials, flaps, and grafts have been used in the past, most contemporary prostheses are either hollow silicone cylinders that are inflated with saline via pump action or semi-rigid rods (32,33). Of all modalities for management of ED, prostheses have the highest satisfaction rates, with 2 large studies demonstrating greater than 95% satisfaction (34,35). While this high rate of satisfaction is encouraging it must be understood that the population of men who are motivated enough to undergo surgery for erectile function may not be representative of the larger population of ED patients.

 

Although some studies suggest that elevated HbA1c levels may predict a higher rate of infections in men with diabetes having penile prosthesis surgery, more recent studies refute this (36). A large study from Wilson, et al demonstrated that neither diabetic status nor preoperative HgA1c were risk factors for prosthesis infection. A more recent study confirmed that elevated HbA1c is not a risk factor for infection; however, short-term poor glucose control (defined as morning fast glucose levels >200 ng/ml) was associated with more complications (37,38).

 

EXPERIMENTAL THERAPIES FOR ED

 

Low-intensity shock wave therapy (LiESWT) has attracted great interest over the past decade as a novel treatment modality for ED. A number of randomized controlled studies in the general ED population have suggested modest but significant short-term benefit with minimal to no side effect profile (39). 

 

A pooled analysis from 5 double-blind, sham-controlled trials of LiESWT reported on 61 men with diabetes and ED responsive to PDE5I and another 48 men with diabetes and ED NOT responsive to PDe5I. Clinically significant improvements in erectile function were noted in 80%, 77%, and 66% of the PDe5I responsive treated patients at 1-, 6-, and 12-months post therapy.  Importantly, over half (55%) of treated men who had been non-responders to PDE5I were able to achieve erection sufficient for penetration with PDE5I post-treatment (40).

 

These encouraging data merit further research, preferably in a dedicated study of men with diabetes-related ED.  Despite encouraging preliminary data this therapy remains experimental and is currently not recommended outside a clinical trial setting conducted at no or minimal cost to patients (26).

 

TREATMENT OF LOW TESTOSTERONE LEVELS

 

Although there is some controversy over what constitutes a true "low" testosterone level and the best way to measure it, some studies have indicated that men with low levels of testosterone and symptoms consistent with low testosterone (e.g., decreased libido, decreased energy, depression, anxiety, fatigue, weight gain) may benefit from testosterone replacement therapy. The general efficacy of testosterone in improving sexual function (particularly sexual desire and response to PDE5I in cases of initial failure to respond) in appropriately selected patients has been established (41). In addition to improving sexual symptoms in these men, testosterone supplementation may have beneficial effects with respect to lean body mass and insulin sensitivity in diabetic men with hypogonadism (42,43).  A recent small RCDB indicated that 40 weeks of testosterone supplementation did not produce a significant improvement in either sexual desire or erectile dysfunction for obese men with type 2 diabetes (44). A more nuanced finding in a larger population suggested that the testosterone supplementation provides benefit for men with sexual dysfunction and severe testosterone deficiency (defined here as less than 8 nmol/L, approximately 230 ng/dL) who are treated such that trough levels approach 15 nmol/L (approximately 432 ng/dL) (45).

 

A number of different testosterone formulations are available, including intramuscular injections, transdermal creams/gels, buccal tablets, and subcutaneous depots (see the Male Reproduction Section of Endotext for a complete discussion of testosterone replacement therapy). 

 

EJACULATORY DYSFUNCTION

 

Men with diabetes may have sexual disorders other than erectile dysfunction. Examples include diminished sexual desire, lack of ejaculation with sexual climax (anejaculation or retrograde ejaculation), and premature ejaculation. Successful antegrade ejaculation depends on the coordination of three neurologic events: seminal emission, bladder neck closure, and contraction of the muscles of the pelvic floor (e.g., bulbocavernosus, ischiocavernous, etc.) (46). In diabetes, derangements of the nerves controlling closure of the connection between the bladder and urethra may disrupt normal ejaculation. In this situation ejaculate is deposited in the innermost portion of the urethra but the connection between the bladder and urethra does not close. Since the bladder neck is open, some or all of the ejaculate may leak backwards into the bladder during the muscle contractions that normally expel the semen from the penis. In the most severe cases there may be total lack of seminal emission. Either of these conditions will impact fertility.  It may also be a source of psychological disturbance to the man; indeed, some men report that they are not able to fully enjoy orgasm in the absence of ejaculation. 

 

From a fertility standpoint, sperm may be retrieved from post-ejaculate urine and then used for artificial insemination. Alternative strategies to overcome retrograde ejaculation generally focus on attempts to help the bladder neck close.  A variety of pharmacologic agents have also been used, including anticholinergics, antihistamines, and alpha-adrenergics (47,48).  Evidence for efficacy of these interventions in management of retrograde/anejaculation is scant.

 

FEMALE SEXUAL DYSFUNCTION

 

Our understanding of the medical and physiological aspects of female sexual function is poor relative to our understanding of men's sexual physiology and function. It is recognized that diabetes can be detrimental to female sexuality in a multifactorial manner, including both psychologic and physiologic dimensions (49,50).

 

In much of the published literature “Female Sexual Dysfunction” is treated as unitary diagnosis in and of itself.  It is more appropriate to consider that this overarching term encompasses several specific (and overlapping) concerns related to sexual function.

 

The International Society for the Study of Women’s Sexual Health describes: (51)

 

  • Hypoactive Sexual Desire Disorder (HSDD, decreased interest in sex and/or receptivity to sexual initiation by a partner)
  • Female Sexual Arousal Disorder, which can be sub-divided into Female Cognitive Arousal Disorder (difficulty with maintaining mental/emotional arousal responses) and Female Genital Arousal Disorder (difficulty with maintaining genital arousal responses).
  • Persistent Genital Arousal Disorder (unwanted and intrusive feelings of genital arousal)
  • Female Orgasm Disorder (compromise of orgasm frequency or intensity).

 

There are similarities between the molecular processes that mediate both male and female genital engorgement with arousal although the tissue effects of course differ (e.g., vasocongestion of erectile tissues leads to penile erection in men and vaginal engorgement/transudate in women) (52). Caruso et al (53) undertook a RCDB trial of 100 mg sildenafil in type 1 diabetic women with sexual dysfunction. Of the 28 women who completed the trial, significant improvement was seen in both subjective and objective parameters. Subjectively, arousal, orgasm, and dyspareunia were all improved in those taking sildenafil in comparison to baseline and those taking placebo. Color Doppler ultrasonography was performed on the clitoral arteries, revealing an increase in blood flow in these women. The clinical utility of ultrasonography in the evaluation of women with sexual dysfunction is unclear; these results should be interpreted with caution.

 

THE IMPORTANCE OF MANAGING LIFESTYLE FACTORS IN TREATING SEXUAL PROBLEMS IN DIABETES

 

As with most aspects of diabetes care, routine exercise, careful monitoring of glucose levels, and usage of appropriate therapies to prevent hyperglycemia are key to preventing progression of diabetes-induced sexual problems. Weight management and dietary prudence are also critical in the management of diabetes. There is evidence to suggest weight loss may reverse erectile dysfunction in some men. In a study of 65 obese men with ED and the Metabolic Syndrome (MetS, obesity with at abnormalities of blood pressure, abnormal glucose level/diabetes, and abnormal cholesterol levels), eating a "Mediterranean diet" (emphasizing fresh fruit and vegetables) for two years led to normalization of erectile function (as determined by an International Index of Erectile function score greater than 22) in 13 of 35 men compared to 2 of 30 men in the group that did not have dietary manipulation (54). 

 

A similar study in women with sexual dysfunction and MetS showed a significant improvement in mean sexual function (mean increase on the Female Sexual Function Index from 19.7 to 26.1 in the treatment group vs. no change from baseline in the control group). Also noted in both of these studies were improvements in serum insulin and glucose level in men and women who consumed a “Mediterranean” diet (55). A multi-center randomized controlled trial of intensive lifestyle intervention in obese women with type 2 diabetes confirmed that women who had the intervention were: 1) more likely to remain sexually active at one year (83% versus 64% for the intervention versus control group, respectively), 2) improve specific domains of sexual function, and 3) to obtain composite scores on the Female Sexual Function Index that were consistent with low risk for sexual dysfunction (28% of intervention patients versus 11% of controls) (56).

 

CONCLUSION

 

Sexual dysfunctions are common in people with diabetes and may arise from a variety of vascular, neurologic, and hormonal derangements. In terms of managing ED, PDE5I are the first-line agents of choice although the failure rate is higher when compared to men with non-diabetic ED.  Second and third line options may be considered should PDE5I fail. Sexual problems related to diabetes extend beyond ED to include sexual desire and ejaculatory dysfunction in men and a variety of sexual concerns in women. In addition to therapy specifically tailored to sexual concerns, management of underlying diabetic condition may markedly improve sexual quality of life in people with diabetes.

 

SUMMARY

 

  • The cause of ED in men with diabetes is multifactorial, including neuropathy, vasculopathy, and endocrinopathy
  • Men with diabetes should be routinely screened for the presence of low testosterone

 

  • Non-ED sexual dysfunctions are common in people with diabetes

 

  • Medical therapies for ED in men with diabetes are not as successful as in men with ED of other etiologies

 

REFERENCES

 

  1. Jacobson, A.M., Braffett, B.H., Cleary, P.A., Dunn, R.L., Larkin, M.E., Wessells, H., et al.Relationship of urologic complications with health-related quality of life and perceived value of health in men and women with type I diabetes: the Diabetes Control and Complications Trial/Epidemiology of Interventions and Complications (DCCT/EDIC) cohort.  Diabetes Care 2015 Oct;38(10):1904-12.
  2. Feldman, H.A., et al., Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging Study. J Urol, 1994. 151(1): p. 54-61.
  3. Kouidrat et al. High prevalence of erectile dysfunction in diabetes: a systematic review and meta-analysis of 145 studies. Diabet Med 2017;34:1185-1192
  4. Kamenov, Z.A.A comprehensive review of erectile dysfunction in men with diabetes.  Exp Clin Endocrinol Diabetes 2015 Mar;123(3):141-58.
  5. McCulloch, D.K., et al., The natural history of impotence in diabetic men. Diabetologia, 1984. 26(6): p. 437-40. -
  6. Gandhi J.The Role of Diabetes Mellitus in Sexual and Reproductive Health: An Overview of Pathogenesis, Evaluation, and Management. CUrr Diabetes Rev 2017;13:573-581
  7. Cartledge, J.J., I. Eardley, and J.F. Morrison, Advanced glycation end-products are responsible for the impairment of corpus cavernosal smooth muscle relaxation seen in diabetes. BJU Int, 2001. 87(4): p. 402-7. -
  8. Saenz de Tejada, I., et al., Impaired neurogenic and endothelium-mediated relaxation of penile smooth muscle from diabetic men with impotence. N Engl J Med, 1989. 320(16): p. 1025-30.
  9. Patel DP. et al. Serum Biomarkers of Erectile Dysfunction in Diabetes Mellitus: A Systematic Review of Current Literature. Sex Med Rev 2017;5:339-348
  10. Mulhall JP. et al. Evaluation and Management of Testosterone Deficiency: AUA Guideline.J Urol. 2018 Aug;200(2):423-432
  11. Kapoor D, et al. Erectile dysfunction is associated with low bioactive testosterone levels and visceral adiposity in men with type 2 diabetes. Int J Androl. 2007 Dec;30(6):500-507 -
  12. Dandona P, et al. Hypogonadotrophic hypogonadism in type 2 diabetes. Aging Male. 2008 Sep;11(3):107-17 -
  13. Morelli A, et al. Which patients with sexual dysfunction are suitable for testosterone replacement therapy. J Endocrinol Invest. 2007 Nov;30(10):880-888
  14. Bhasin S. et alTestosterone Therapy in Men With Hypogonadism: An Endocrine Society Clinical Practice Guideline J Clin Endocrinol Metab. 2018 May 1;103(5):1715-1744
  15. Wu F, et al. Identification of late-onset hypogonadism, in middle-aged and elderly men. N Engl J Med. 2010 Jul 8;363(2):123-35
  16. Dean R, et al. Physiology of penile erection and pathophysiology of erectile dysfunction. Urol Clin NA 2005 Nov;32(4):379-95
  17. Goldstein, I., et al., Vardenafil, a new phosphodiesterase type 5 inhibitor, in the treatment of erectile dysfunction in men with diabetes: a multicenter double-blind placebo-controlled fixed-dose study. Diabetes Care, 2003. 26(3): p. 777-83. -
  18. Saenz de Tejada, I., et al., Effects of tadalafil on erectile dysfunction in men with diabetes. Diabetes Care, 2002. 25(12): p. 2159-64. -
  19. Behrend, L., J. Vibe-Petersen, and H. Perrild, Sildenafil in the treatment of erectile dysfunction in men with diabetes: demand, efficacy and patient satisfaction. Int J Impot Res, 2005. 17(3): p. 264-9. -
  20. Hatzichristou D, et al. Efficacy of tadalafil once daily in men with diabetes mellitus and erectile dysfunction. Diabet Med. 2008 Feb;25(2):138-46 -
  21. Park K, et al. Chronic treatment with a type 5 Phosphodiesterase inhibitor suppresses apoptosis of corporal smooth muscle by potentiating Akt signaling in a rat model of diabetic erectile dysfunction. Eur Urol. 2008;53:1282-1289 -
  22. DeYoung LX, et al. Endothelial rehabilitation: the impact of chronic PDE5 inhibitors on erectile function and protein alteration in cavernous tissue of diabetic rats. Eur Urol. 2008;54:213-220 -
  23. Burnett AL, et al. Serum biomarker measurements of endothelial function and oxidative stress after daily dosing of sildenafil in type 2 diabetic men with erectile dysfunction. J Urol. 2009;181:245-251. -
  24. Aversa A, et al. Chronic administration of Sildenafil improves markers of endothelial function in men with Type 2 diabetes. Diabet Med. 2008 Jan;25(1):37-44 -
  25. Montorsi F, Brock G, Lee J, Shapiro J, van Poppel H, Graefen M, et al. Effect of nightly versus on-demand vardenafil on recovery of erectile function in men following bilateral nerve-sparing radical prostatectomy. Eur Urol. 2008;54:924-931
  26. Burnett AL et al. Erectile Dysfunction: AUA Guideline.J Urol 2018 Sep;200(3):633-641
  27. Bella AJ, Brock GB.Intracavernous pharmacotherapy for erectile dysfunction. Endocrine 2004;23:149-55
  28. Heaton, J.P., et al., Intracavernosal alprostadil is effective for the treatment of erectile dysfunction in diabetic men. Int J Impot Res, 2001. 13(6): p. 317-21 -
  29. Perimenis, P., et al., Long-term treatment with intracavernosal injections in diabetic men with erectile dysfunction. Asian J Androl, 2006. 8(2): p. 219-24. -
  30. Costa A, et al. Intraurethral alprostadil for erectile dysfunction: a review of the literature. Drugs. 2012 Dec 3;72(17):2243-54
  31. Ryder, R.E., et al., Impotence in diabetes: aetiology, implications for treatment and preferred vacuum device. Diabet Med, 1992. 9(10): p. 893-8. -
  32. Akoz, T., et al., The use of iliac bone flap as a penile stiffener in a diabetic patient with erectile dysfunction. Plast Reconstr Surg, 1999. 103(7): p. 1975-8. -
  33. Mulcahy, J.J., et al., The penile implant for erectile dysfunction. J Sex Med, 2004. 1(1): p. 98-109. -
  34. Levine, L.A., C.R. Estrada, and A. Morgentaler, Mechanical reliability and safety of, and patient satisfaction with the Ambicor inflatable penile prosthesis: results of a 2 center study. J Urol, 2001. 166(3): p. 932-7. -
  35. Montorsi, F., et al., AMS three-piece inflatable implants for erectile dysfunction: a long-term multi-institutional study in 200 consecutive patients. Eur Urol, 2000. 37(1): p. 50-5. -
  36. Bishop, J.R., et al., Use of glycosylated hemoglobin to identify diabetics at high risk for penile periprosthetic infections. J Urol, 1992. 147(2): p. 386-8. -
  37. Wilson, S.K., et al., Quantifying risk of penile prosthesis infection with elevated glycosylated hemoglobin. J Urol, 1998. 159(5): p. 1537-9; discussion 1539-40. -
  38. Cakan, M., et al., Risk factors for penile prosthetic infection. Int Urol Nephrol, 2003. 35(2): p. 209-13. –
  39. Campbell JD et al. Meta-analysis of randomized controlled trials that assess the efficacy of low-intensity shockwave therapy for the treatment of erectile dysfunction. Ther Adv Urol. 2019 Mar 29;11:1756287219838364. doi: 10.1177/1756287219838364.
  40. Spivak et al. Low-Intensity Extracorporeal Shockwave Therapy for Erectile Dysfunction in Diabetic Patients. Sex Med Rev. 2019 Aug 1:S2050-0521(19)30072-1. doi: 10.1016/j.sxmr.2019.06.007
  41. Buvat J et al. Significance of hypogonadism in erectile dysfunction. World J Urol. 2006 Dec;24(6):657-67. -
  42. Kapoor D et al. Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. Eur J Endocrinol. 2006 Jun;154(6):899-906 -
  43. Kapoor D, et al. The effect of testosterone replacement therapy on adipocytokines and C-reactive protein in hypogonadal men with type 2 diabetes. Eur j Endocrinol. 2007 May;156(5):595-602 –
  44. Gianatti, E.J., Dupuis, P., Hoermann, R., Zajac, J.D., Grossmann, M.Effects of testosterone treatment on constitutional and sexual symptoms in men with type 2 diabetes in a randomized, placebo-controlled clinical trial.  J Clin Endocrinol Metab. 2014 Oct;99(10);3821-8.
  45. Hackett, G., Cole, N., Bhartia, M., Kennedy, D., Raju, J., Wilkinson, P., et al.The response to testosterone undecanoate in men with type 2 diabetes is dependent on achieving threshold serum levels (the BLAST study).  Int J Clin Pract 2014 Feb;68(2):203-15.
  46. Clement P, Giuliano F.Physiology and Pharmacology of Ejaculation. Basic Clin Pharmacol Toxicol 2016 Oct;119 suppl 3:18-25
  47. Kamischke, A. and E. Nieschlag, Update on medical treatment of ejaculatory disorders. Int J Androl, 2002. 25(6): p. 333-44. -
  48. Tomasi, P.A., G. Fanciulli, and G. Delitala, Successful treatment of retrograde ejaculation with the alpha1-adrenergic agonist methoxamine: case study. Int J Impot Res, 2005. 17(3): p. 297-9. -
  49. Giraldi A., Kristensen E..Sexual dysfunction in women with diabetes mellitus. J Sex Res 2010;47:199-211
  50. Bitzer J., Alder J.: Diabetes and female sexual health. Women's health 2009;5:629-636-
  51. Parish SJ. et al. The Evolution of the Female Sexual Disorder/Dysfunction Definitions, Nomenclature, and Classifications: A Review of DSM, ICSM, ISSWSH, and ICD. Sex Med Rev 2021;9:36-56
  52. Yang, C.C., et al., Sexually responsive vascular tissue of the vulva. BJU Int, 2006. 97(4): p. 766-72. -
  53. Caruso, S., et al., Sildenafil improves sexual functioning in premenopausal women with type 1 diabetes who are affected by sexual arousal disorder: a double-blind, crossover, placebo-controlled pilot study. Fertil Steril, 2006 -
  54. Esposito K, et al. Mediterranean diet improves erectile function in subjects with the metabolic syndrome. Int J Impot Res. 2006 Jul-Aug;18(4):405-10. -
  55. Esposito K, et al. Mediterranean diet improves sexual function in women with the metabolic syndrome. Int J Impot Res. 2007 Sep-Oct;19(5):486-91. –
  56. Wing, R.R., Bond, D.S., Gendrano, I.N 3rd, Wadden, T., Bahnson, J., Lewis, C.E., et al.Effect of intensive lifestyle intervention on sexual dysfunction in women with type 2 diabetes: results from an ancillary Look AHEAD study.  Diabetes Care. 2013 Oct;26(10):29

Osteoporosis: Clinical Evaluation

ABSTRACT

The identification of a patient at high risk of fracture should be followed by evaluation for factors contributing to low bone mass, skeletal fragility, falls, and fractures. Components of the evaluation include a bone density test, osteoporosis-directed medical history and physical exam, laboratory studies, and possibly skeletal imaging. A bone density test with dual-energy X-ray absorptiometry (DXA) helps with diagnostic classification, assessment of fracture risk, and provides a baseline for monitoring the skeletal effects of treatment. FRAX is a fracture risk algorithm that includes input of femoral neck bone mineral density measured by DXA. The DXA T-score, prior fracture history, and FRAX estimation of fracture risk are used with clinical practice guidelines to determine whether treatment is indicated. The medical history may reveal underlying causes of osteoporosis (e.g., nutritional deficiencies, gastric surgery, medications with adverse skeletal effects) and important risk factors for fracture (e.g., past history of fracture, family history of osteoporosis, or recent falls). Physical exam may show skeletal deformities due to unrecognized fractures (e.g., loss of height, kyphosis, or diminished rib-pelvis space), identify possible secondary causes of skeletal fragility (e.g., blue sclera with osteogenesis imperfecta, urticarial pigmentosa with systemic mastocytosis, dermatitis herpetiformis with celiac disease, or bone tenderness with osteomalacia), and help to recognize patients with poor balance and frailty that might lead to falls. Laboratory studies may show potentially reversible abnormalities (e.g., vitamin D deficiency, hypocalcemia, or impaired kidney function) that must be assessed and corrected, if possible, before starting pharmacological therapy. Disorders other than osteoporosis, requiring other types of treatment, may be found; for example, low serum alkaline phosphatase suggests hypophosphatasia, M-component may be due to myeloma, or hypocalciuria due to celiac disease. There are important safety considerations that can be derived from a pre-treatment assessment, as well. A patient with a blood clotting disorder should not be treated with raloxifene, a history of esophageal stricture is a contraindication for oral bisphosphonates, and previous skeletal radiation therapy precludes treatment with teriparatide or abaloparatide. Skeletal imaging may be helpful when a fracture, malignancy, or Paget’s disease of bone is suspected. Bone biopsy is rarely performed in clinical practice, but may be helpful in some situations, such as when it is necessary to determine the underlying bone disease in a patient with severe chronic kidney disease.

INTRODUCTION

Osteoporosis is a common disease characterized by low bone strength that results in an increased risk of fracture (1). Fractures are associated with serious clinical consequences, including pain, disability, loss of independence, and death, as well as high healthcare costs. Early identification and intervention with patients at high risk for fracture is needed to reduce the burden of osteoporotic fractures (2). The management of a patient with a confirmed diagnosis of osteoporosis or low bone mass (osteopenia) includes assessment of fracture risk, evaluation for secondary causes of skeletal fragility, making decisions on initiation of treatment, and identification of all relevant clinical factors that may influence patient management. This is a review of the key components in the care of patients with osteoporosis prior to treatment.

DIAGNOSIS OF OSTEOPOROSIS

The World Health Organization (WHO) diagnostic classification (Table 1) (3) is made by bone mineral density (BMD) testing with dual-energy X-ray absorptiometry (DXA) using the T-score, calculated by subtracting the mean BMD (in g/cm2) of a young-adult reference population from the patient’s BMD and dividing by the standard deviation (SD) of the young-adult reference population. The International Society for Clinical Densitometry (ISCD) recommends that BMD be measured at the lumbar spine (ideally L1-L4), total hip, and femoral neck, with the 33% radius (1/3 radius) being measured when the lumbar spine and/or hip cannot be measured (e.g., obese patient who exceeds weight limit of table) or is invalid (e.g., patient with lumbar laminectomy or bilateral total hip replacements) (4). Osteoporosis cannot be diagnosed by BMD measurement at skeletal sites other than lumbar spine, total hip, femoral neck, and 33% radius or with technologies other than DXA, except for total hip and femoral neck T-scores calculated from 2D projections of quantitative computed tomography (QCT) data. The quality of DXA instrument maintenance, acquisition, analysis, interpretation, and reporting is important in obtaining valid results that can be used for making appropriate clinical decisions (4,5). In a patient with a fragility fracture, a clinical diagnosis of osteoporosis may be considered independently of BMD results, assuming that other causes of skeletal fragility (e.g., osteomalacia) are not responsible for the fracture. Establishing a diagnosis of osteoporosis is clinically useful because it facilitates communication among healthcare providers and patients concerning a disease with potentially serious consequences; in some countries, such as the United States (US), a diagnosis is necessary in order to select a numerical code for submission of insurance claims for reimbursement for medical services. The US National Bone Health Alliance (6) has recommended that osteoporosis be diagnosed in postmenopausal women and men over the age of 50 years in any of the following circumstances: T-score ≤ −2.5 at the lumbar spine or hip; low-trauma hip fracture; osteopenia by BMD with a low-trauma vertebral, proximal humerus, pelvis, or, in some cases, distal forearm fracture; and when FRAX shows fracture risk above the country-specific threshold for treatment (for the US, this is 10-year probability of major osteoporotic fracture ≥ 20% or 10-year probability of hip fracture ≥ 3%).

Table 1. World Health Organization criteria for classification of patients with bone mineral density measured by dual-energy X-ray absorptiometry (3).

 

Classification

T-score

Normal

-1.0 or greater

Low bone mass (osteopenia)

Between 1-.0 and -2.5

Osteoporosis

-2.5 and below

Severe osteoporosis

-2.5 and below + fragility fracture

The National Osteoporosis Foundation (NOF) indications for BMD testing in the US (7), which are similar to the ISCD Official Positions (4), are listed in Table 2. BMD testing should be done when it is likely to have an influence on patient management decisions. Other organizations and other countries with different economic resources and health care priorities have used a variety of methodologies to develop alternative recommendations (8-10).

Table 2. National Osteoporosis Foundation recommends that bone mineral density testing be performed at DXA facilities using accepted quality assurance procedures for the following individuals (7).

 

All women age 65 years and older and men age 70 years and older

Postmenopausal women and men above age 50-69 years, based on risk factor profile

Postmenopausal women and men over age 50 years who have had an adult-age fracture, to diagnose and determine the degree of osteoporosis

FRACTURE RISK ASSESSMENT

There is a robust correlation between BMD and fracture risk, with approximately a 2-fold increase in fracture risk for every 1 standard deviation (SD) decrease in BMD (11). However, many or most patients with a hip fracture have a T-score better than -2.5 (12); although fracture risk is higher in patients with very low BMD, there are numerically many more patients with a T-score better than -2.5 than with a T-score of -2.5 or worse, therefore numerically more fractures in those with higher T-scores. The presence of clinical risk factors (CRFs) that are independent of BMD, particularly advancing age, prior fracture, and recency of fracture, can identify patients at high risk for fracture by providing information on fracture risk that is complementary to BMD. The NOF has provided an extensive list of CRFs (Table 3) for osteoporosis and fractures. Since most fractures occur as a result of a fall, it is helpful to recognize risk factors for falling (Table 4) so that appropriate interventions can be made, when possible, to reduce the chances of falling.

Table 3. Conditions, diseases and medications that cause or contribute to osteoporosis and fractures (7).

Lifestyle Factors

Low Calcium Intake

Vitamin D Insufficiency

Excess Vitamin A

High Caffeine Intake

High Salt Intake

Aluminum (in antacids)

Inadequate Physical Activity

Immobilization

Smoking

Falling

Thinness

Alcoholism

Genetic Factors

Cystic Fibrosis

Homocystinuria

Osteogenesis Imperfecta

Ehlers-Danlos Syndrome

Hypophosphatasia

Gaucher’s Disease

Idiopathic Hypercalciuria

Porphyria

Glycogen storage diseases

Marfan Syndrome

Riley-Day Syndrome

Hemochromatosis

Menkes Steely Hair Syndrome

Parental History of Hip Fracture

Androgen Insensitivity

Turner’s & Klinefelter’s Syndromes

Endocrine Disorders

Adrenal Insufficiency

Diabetes Mellitus

Hyperthyroidism

Cushing’s Syndrome

Hyperparathyroidism

Hypogonadal States

Panhypopituitarism

Athletic Amenorrhea

Anorexia Nervosa and Bulimia

Hyperprolactinoma

Premature Ovarian Failure

 

Gastrointestinal disorders

Celiac Disease

Inflammatory Bowel Disease

Primary Biliary Cirrhosis

Gastric Bypass

Malabsorption

GI Surgery

Pancreatic Disease

 

Hematologic Disorders

Hemophilia

Multiple Myeloma

Systemic Mastocytosis

Leukemia

Lymphoma

Sickle Cell Disease

Thalassemia

 

Rheumatic and Autoimmune Diseases

Ankylosing Spondylitis

Lupus

Rheumatoid Arthritis

 

Miscellaneous Conditions and Diseases

Chronic Obstructive Pulmonary Disease

Muscular Dystrophy

Amyloidosis

End Stage Renal Disease

Parenteral Nutrition

Chronic Metabolic Acidosis

Epilepsy

Post-Transplant Bone Disease

Congestive Heart Failure

Idiopathic Scoliosis

Prior Fracture as an Adult

Depression

Multiple Sclerosis

Sarcoidosis

HIV/AIDS

 

Medications

Anticoagulants (heparin)

Cancer Chemotherapeutic Drugs

Gonadotropin Releasing Hormone Agonists

Anticonvulsants

Lithium

Aromatase Inhibitors

Depo-medroxyprogesterone

Barbiturates

Glucocorticoids (> 5mg of prednisone or equivalent for > 3 months)

Cyclosporine A

Tacrolimus

 

 

Table 4. Risk factors for falls adapted from guidelines of the National Osteoporosis Foundation (7).

Environmental Risk Factors

Lack of assistive devices in bathrooms, loose throw rugs, low level lighting, obstacles in the walking path, slippery outdoor conditions

Medical Risk Factors

Age, anxiety and agitation, arrhythmias, dehydration, depression, female gender, impaired transfer and mobility, malnutrition, orthostatic hypotension, poor vison and use of bifocals, previous fall, reduced mental acuity and diminished cognitive skills, urgent urinary incontinence, Vitamin D insufficiency (serum 25-OH-D < 30ng/ml (75nmol/l)), medications causing over-sedation (narcotic analgesics, anticonvulsants, psychotropics), diabetes

Neurological and Musculoskeletal Risk Factors

Kyphosis, poor balance, reduced proprioception, weak muscles

Other Risk Factors; Fear of falling

The presence of any of these risk factors should trigger consideration of further evaluation and treatment to reduce the risk of falls and fall-related injuries.

VERTEBRAL FRACTURE ASSESSMENT (VFA)

VFA is a method for imaging the thoracic and lumbar spine by DXA for the purpose of detecting vertebral fracture deformities. Identification of a previously unrecognized vertebral fracture may alter diagnostic classification, change estimation of fracture risk, and influence treatment decisions (13). VFA compares favorably with standard radiographs of the spine, with good correlation for detecting moderate (grade 2) and severe (grade 3) vertebral fractures, a smaller dose of ionizing irradiation, greater patient convenience (i.e., it may be done at the same visit and with the same instrument as BMD testing by DXA), and lower cost. In a study of women age 65 and older, using the Genant semi-quantitative (SC) method of classifying vertebral deformities (14), the sensitivity of VFA for diagnosing moderate and severe vertebral fractures was 87-93%, with a specificity of 93-95% (15). Indications for vertebral imaging are listed in Table 5. Optimal use of DXA and VFA requires training and adherence to well established quality standards (4).

Table 5. International Society for Clinical Densitometry (ISCD) indications for lateral spine imaging by standard radiography or vertebral fracture assessment (VFA).

The ISCD Official Positions (4) state that vertebral imaging is indicated when the T-score is < -1.0 and one or more of the following is present:

Women > 70 years of age or men > 80 years of age

Historical height loss > 4cm (1.5 inches)

Self-reported but undocumented prior vertebral fracture

Glucocorticoid therapy equivalent to ≥ 5 mg of prednisone or equivalent per day for ≥ 3 months

QUALITY OF DXA AND VFA

DXA and VFA should be performed by well-trained and experienced staff operating an instrument that has been maintained and calibrated according to the manufacturer’s standards. Precision assessment and least significant change (LSC) calculation by each DXA technologist are required in order to make quantitative comparisons of serial BMD measurements. The LSC is the smallest change in BMD that is statistically significant, usually with a 95% level of confidence. The use of the correct scan modes, proper patient positioning, consistent vertebral body labeling, and bone edge detection are among the essential elements for serial comparisons of BMD. VFA should be done by a technologist properly trained in acquisition techniques and interpreted by a clinician familiar with methods of diagnosing vertebral fractures using this technology. Bone densitometry facilities should be supervised by a clinician who knows current methods for BMD measurement and fully understands the standards for quality control, interpretation, and reporting of the findings. Poor quality studies may result in inappropriate clinical decisions, generate unnecessary healthcare expenses, and be harmful to patients (5). Assurances of high quality DXA can be attained through education, training, and certification of DXA technologists and interpreters by organizations such as the ISCD. DXA facilities should understand and adhere to ISCD Official Positions and DXA Best Practices; facility accreditation provides assurance of adherence to DXA quality standards (4,16,17).

TECHNOLOGIES FOR ASSESSMENT OF SKELETAL HEALTH

Dual-Energy X-Ray Absorptiometry (DXA)

Devices that measure or estimate BMD differ according to their clinical utility, cost, portability, and use of ionizing radiation (Table 6). DXA is the “gold standard” method for measuring bone density in clinical practice (18). There is a strong correlation between mechanical strength and BMD measured by DXA biomechanical studies (19). In observational studies of untreated patients, there is a robust relationship between fracture risk and BMD measured by DXA (11). The WHO diagnostic classification of osteoporosis is based primarily on reference data obtained by DXA (3), and femoral neck BMD provides input into the FRAX algorithm. Most randomized clinical trials showing reduction in fracture risk with pharmacological therapy have selected study participants according to BMD measured by DXA (20). There is a relationship between fracture risk reduction with drug therapy and increases in BMD measured by DXA (21). Accuracy and precision of DXA are excellent (22). Radiation exposure with DXA is very low (23). BMD of the 33% (one-third) radius, measured either by a dedicated peripheral DXA (pDXA) device or a central DXA instrument with appropriate software, may be used for diagnostic classification with the WHO criteria and to assess fracture risk, but is generally not clinically useful in monitoring the effects of treatment (23). DXA measures bone mineral content (BMC in grams [g]) and bone area (cm2), then calculates areal BMD in g/cm2 and derives parameters, such as the T-score and Z-score. DXA is used for diagnostic classification, assessment of fracture risk, and for monitoring changes in BMD over time.

Table 6. Devices for measuring or estimating bone mineral density (BMD)

 

DXA

pDXA

QUS

QCT

pQCT

Diagnostic classification*

Yes

Limited**

No

Yes***

No

Measurement

Areal BMD

Areal BMD

SOS, BUA

Volumetric BMD

Volumetric BMD

Prediction of fracture risk

Yes

Yes

Yes

Yes

Yes

Monitoring changes over time

Yes

No

No

Yes

No

Ionizing radiation

++

+

0

+++

++

Cost

++

+

+

+++

++

Clinical applications of different technologies are listed with approximate comparison of associated radiation exposure and cost, with 0 = none, + = low, ++ moderate, +++ = highest.

DXA = dual-energy X-ray absorptiometry; pDXA = peripheral DXA; QUS = quantitative ultrasound; QCT = quantitative computed ultrasound; pQCT = peripheral QCT; SOS = speed of sound; BUA = broadband ultrasound attenuation; * World Health Organization classification; **pDXA of the distal one-third radius (33% radius) may be used with the WHO classification; *** Total hip and femoral neck T-scores calculated from 2D projections of QCT data may be used with the WHO classification

Quantitative Ultrasound (QUS)

QUS devices emit inaudible high frequency sound waves in the ultrasonic range, typically between 0.1 and 1.0 megahertz (MHz). The sound waves are produced and detected by means of high-efficiency piezoelectric transducers, which must have good acoustical contact with the skin over the bone being tested. Technical differences among QUS systems are great, with different instruments using variable frequencies, different transducer sizes, and sometimes measuring different regions of interest, even at the same skeletal site. The calcaneus is the skeletal site most often tested, although other bones, including the radius, tibia, and finger phalanges, can be used. Commercial QUS systems usually measure two parameters- the speed of sound (SOS) and broadband ultrasound attenuation (BUA). A proprietary value, such as the “quantitative ultrasound index” (QUI) with the Hologic Sahara or “stiffness index” with the GE Healthcare Achilles Express, may be calculated from a combination of these measurements. SOS varies according to the type of bone, with a typical range of 3000-3600 meters per second (m/sec) with cortical bone and 1650-2300 m/sec for trabecular bone (24). A higher bone density is associated with a higher SOS. BUA, reported as decibels per megahertz (dB/MHz), is a measurement of the loss of energy, or attenuation, of the sound wave as it passes through bone. As with SOS, a higher bone density is associated with a higher BUA. Values obtained from calculations using ultrasound parameters may be used to generate an estimated BMD and a T-score. The T-score derived from a QUS measurement is not the same as a T-score from a DXA. QUS cannot be used for diagnostic classification and is not clinically useful to monitor the effects of therapy (25).

Quantitative Computed Tomography (QCT) and Peripheral QCT (pQCT)

QCT and pQCT measure trabecular and cortical volumetric BMD at the axial skeleton and peripheral skeletal sites, respectively. QCT is a useful research tool to enhance understanding of the pathophysiology of osteoporosis and the mechanism of action of pharmacological agents used to treat osteoporosis. QCT predicts fracture risk, with the correlation varying according to skeletal site and bone compartment measured, type of fracture predicted, and population assessed (26). The ISCD Official Positions state that “spinal trabecular BMD as measured by QCT has at least the same ability to predict vertebral fractures as AP spinal BMD measured by central DXA in postmenopausal women with lack of sufficient evidence to support this position in men; pQCT of the forearm at the ultra-distal radius predicts hip, but not spine, fragility fractures in postmenopausal women with lack of sufficient evidence to support this position in men (26).” QCT is more expensive than DXA and QUS and uses higher levels of ionizing radiation than DXA. T-scores by QCT are typically lower than with DXA (27), thereby overestimating the prevalence of osteoporosis, with the exception of total hip and femoral neck T-scores calculated from 2D projections of QCT data, which are similar to DXA-derived T-scores at the same regions of interest and may be used for diagnosis of osteoporosis in accordance with the WHO criteria. T-scores and femoral neck BMD derived from 2D projections of QCT data may also be used as input for the FRAX algorithm to estimated 10-year fracture probabilities.

FRACTURE RISK ASSESSMENT TOOL (FRAX®)

The combination of BMD and clinical risk factors (CRFs) predicts fracture risk better than BMD or CRFs alone (28,29) (2). A fracture risk assessment tool (FRAX) combines CRFs and femoral neck BMD in a computer-based algorithm that estimates the 10-year probability of hip fracture and major osteoporotic fracture (i.e., clinical spine, hip, proximal humerus, and distal forearm fracture). FRAX can be accessed online at http://www.shef.ac.uk/FRAX (Figure 1), on most software versions of DXA systems, and on smartphones. FRAX is based on analysis of data from 12 large prospective observational studies in about 60,000 untreated men and women in different world regions, having over 250,000 person-years of observation and more than 5,000 reported fractures reported.

Figure 1. FRAX online for US Caucasian patients. This example shows a 65-year-old woman who has no clinical risk factors for fracture and a femoral neck BMD of 0.582 g/cm2 with a Hologic instrument. The 10-year probability of major osteoporotic fracture is 11% and the 10-year probability of hip fracture is 2.2%. These levels do not meet the National Osteoporosis Foundation guidelines for initiation of pharmacological therapy in the US (7). Image reproduced with permission of the World Health Organization.

The input for FRAX is the patient’s age, sex, height, weight, a “yes” or “no” response indicating the presence or absence for each of 7 CRFs: 1. previous ‘spontaneous’ or fragility fracture as an adult; 2. parent with hip fracture; 3. current tobacco smoking; 4. ever use of chronic glucocorticoids at least 5 mg prednisolone for at least 3 months; 5. confirmed rheumatoid arthritis; 6. secondary osteoporosis, such as type 1 diabetes, osteogenesis imperfecta in adults, untreated longstanding hyperthyroidism and hypogonadism, or premature menopause (note: this is a “dummy” risk factor that has no effect on the fracture risk calculation unless no femoral neck BMD value is entered); 7. alcohol intake greater than 3 units per day, with a unit of alcohol defined as equivalent to a glass of beer, an ounce of spirits or a medium-sized glass of wine), and if available, femoral neck BMD and trabecular bone score (TBS). Since the introduction of FRAX, upgrades have been introduced to correct errors, enhance its usability, and incorporate new data that have become available.

Benefits of FRAX

The use of FRAX provides a quantitative estimation of fracture risk that is based on robust data in large populations of men and women with ethnic and geographic diversity. Expression of fracture risk as a probability provides greater clinical utility for than relative risk. When combined with cost-utility analysis, a fracture risk level at which it is cost-effective to treat may be derived. FRAX can be used to estimate fracture probability without femoral neck BMD, allowing it to be used when DXA in unavailable or inaccessible. FRAX is incorporated into many clinical practice guidelines.

Limitations of FRAX

To generate a valid FRAX output, the responses to CRF questions must be correct; for example, an incorrect entry of self-reported rheumatoid arthritis or use of glucocorticoids could skew the results toward overestimation of fracture risk. FRAX may underestimate or overestimate fracture risk due to dichotomized (yes or no) input for CRFs that in reality are associated with a range of risk that varies according to dose, duration of exposure, or severity; for example, fracture risk may be underestimated when a patient is on high-dose glucocorticoid therapy or has had multiple recent fragility fractures, even when a “yes” response is entered for these CRFs. FRAX is validated only in untreated patients and may overestimate fracture risk when the patient is being treated; the NOF/ISCD guidance on FRAX suggests that “untreated” may be interpreted as never treated or if previously treated, no bisphosphonate for the past 2 years (unless it is an oral agent taken for less than 2 months); and no estrogen, raloxifene, calcitonin, or denosumab for the past 1 year (7). In this context, calcium and vitamin D do not constitute treatment. FRAX in the US allows input for 4 ethnicities (Caucasian, Black, Hispanic, Asian); it is not clear how to use FRAX for patients of other ethnicities or a mix of these ethnicities. Answering “yes” for the category of secondary osteoporosis has no effect on the fracture risk calculation as long as a value for femoral neck BMD is entered. The range of error for a fracture probability generated by FRAX is unknown but may be substantial in some cases. Some important risk factors, such as falls and frailty, are not directly entered into FRAX, although they are indirectly included insofar as they are a component of aging. FRAX may underestimate fracture risk when the lumbar spine BMD is substantially lower than femoral neck BMD, as may occur in about 15% of patients (30).

Despite the numerous limitations of FRAX, it is a helpful clinical tool when used with a good understanding of factors that may result in underestimation or overestimation of fracture risk. FRAX may enhance discussion of risk with the patient and help to identify those who are at sufficiently high for fracture to benefit from therapy.

MEDICAL HISTORY

A thorough medical history may identify risk factors for osteoporosis and fractures, suggesting that a bone density test and/or further evaluation is indicated. The medical history may also reveal symptoms of potentially correctable causes of skeletal fragility (e.g., gluten intolerance with celiac disease) or co-morbidities that could influence treatment decisions (e.g., esophageal stricture suggests that oral bisphosphonates should not be given). A history of falls is a predictor of future falls, with that risk potentially modifiable though appropriate interventions. Finally, some symptoms may trigger further evaluation for the presence of fractures (e.g., historical height loss or development of kyphotic posture suggests the possibility of vertebral fractures that may warrant spine imaging). Table 7 provides examples of helpful information that might be obtained from a thoughtful interactive discussion with the patient.

Medical History for Patients with Osteoporosis

A thorough review of systems and history of relevant familial disorders, previous surgical procedures, medications, dietary supplements, food intolerances, and lifestyle provide helpful information in the management of patients with osteoporosis. Such historical information may play a role in determining who should have a bone density test, assessing fracture risk, providing input for FRAX®, evaluating for secondary causes of osteoporosis, selecting the most appropriate treatment to reduce fracture risk, and finding factors contributing to suboptimal response to therapy. Listed here are key components of the skeletal health history and examples of the potential impact on patient care.

Table 7. Clinical Utility of the Medical History

Clinical Utility

Medical History

Assist in determining who need a bone density test

See Table 3

Assessing fracture risk

See Table 3 and 4

Input for FRAX®

Age, sex, weight, height, previous fracture, parent with hip fracture, current tobacco smoking, ever use of glucocorticoids, rheumatoid arthritis, secondary osteoporosis, alcohol intake 3 or more units per day, and if available, femoral neck bone mineral density and trabecular bone score

Evaluating for secondary causes of osteoporosis

See Table 3

Selecting most appropriate treatment

Identify co-morbidities of clinical significance. For example, high risk of breast cancer favors raloxifene use, while history of thrombophlebitis suggests that raloxifene should not be used; esophageal stricture is a contraindication for oral bisphosphonate use; a patient with a skeletal malignancy should not be treated with teriparatide.

Factors contributing to suboptimal response to therapy

Compliance and persistence to therapy; adequacy of calcium and vitamin D; comorbidities listed in Table3.

PHYSICAL EXAM

Findings of importance on the physical exam of a patient with osteoporosis may be the sequelae of old fractures (e.g., kyphosis due to old vertebral fractures), a consequence of a recent fracture (e.g., localized vertebral spinous process tenderness with a new vertebral fracture), or abnormalities suggestive of a secondary cause of osteoporosis (e.g., thyromegaly with thyrotoxicosis). An accurate measurement of height with a wall-mounted stadiometer is a helpful office tool for evaluating patients at risk for fracture. A height loss of 1.5 inches (4.0 cm) or more compared to the historical maximum (31,32) or a loss of 0.75 inches (2.0 cm) or more compared to a previous measured height (33) suggests a high likelihood of vertebral fracture. Body weight measurement is part of the osteoporosis evaluation because low body weight (less than 127 lbs) (34), low BMI (20 kg/m2 or less) (35), and weight loss of 5% or more (36) are associated with increased risk of fracture. Localized tenderness of the spine, kyphosis, or diminished distance between the lower ribs and the pelvic brim may be the result of one or more vertebral fractures. Abnormalities of gait, posture, balance, muscle strength, or the presence of postural hypotension or impaired level of consciousness may be associated with increased risk of falling. Bone tenderness may be the caused by osteomalacia. Atrophic testicles suggest hypogonadism. Patients should be observed for stigmata of hyperthyroidism or Cushing’s syndrome. Blue sclera, hearing loss, and yellow-brown teeth are suggestive of osteogenesis imperfecta. Joint hypermobility and skin fragility could be due to Ehlers-Danlos syndrome. Urticaria pigmentosa may occur with systemic mastocytosis. Table 8 shows examples of abnormal physical exam findings with osteoporosis.

Table 8. Focused Physical Examination in a Patient with Osteoporosis

Component of physical exam

Example of finding of potential skeletal importance

Potential clinical implications for skeletal health

Vital signs

Low body weight or body mass index

Anorexia nervosa

Loss of height

Vertebral fracture

Loss of weight

Malignancy, malabsorption

Skin

Urticaria pigmentosa

 

Dermatitis herpetiformis

Systemic mastocytosis

 

Celiac disease

Striae, acne

Cushing’s syndrome, exogenous glucocorticoids

Head

Cranial dysostosis

Hypophosphatasia

Eyes

Blue sclera

Osteogenesis imperfect

Ears

Hearing loss

Osteogenesis imperfecta, sclerosteosis

Nose

Anosmia

Kallmann syndrome

Throat

Poor dentition

Increased risk of osteonecrosis of the jaw

Neck

Thyromegaly

Thyrotoxicosis

Lungs

Decreased breath sounds

Chronic obstructive pulmonary disease

Heart

Aortic insufficiency

Marfan’s syndrome

Musculoskeletal

Kyphosis

Vertebral fractures

Spinous process tenderness

Acute vertebral fracture

Decreased space between lower ribs and pelvis

Vertebral fractures

Tender bones

Osteomalacia

Inflammatory joint disease

Rheumatoid arthritis

Hypermobility of joints

Ehlers-Danlos syndrome

Muscle weakness

Vitamin D deficiency, osteomalacia

Abdomen

Hepatomegaly

Chronic liver disease

Surgical scars

Bariatric surgery, gastrectomy

Genitalia

Testicular atrophy

Hypogonadism

Neurological

Poor balance

High fall risk, vitamin D deficiency

Dementia

Poor adherence to therapy, high fall risk

This table provides examples of findings on physical exam that may be helpful in the evaluation of skeletal health. It is not intended to show all findings of importance.

EVALUATION FOR SECONDARY CAUSES OF OSTEOPOROSIS

The possibility of previously unrecognized causes of skeletal fragility should be considered in every patient with osteoporosis (37). After an initial medical history is taken and physical exam is performed, appropriate laboratory testing and imaging may provide information that is critical for ongoing patient care. Osteoporosis is commonly divided into two categories according to etiology. “Primary osteoporosis” is due to time-appropriate postmenopausal estrogen deficiency (type I osteoporosis, preferentially involving trabecular bone loss) or to aging in men and women (type II osteoporosis, with a combination of trabecular and cortical bone loss). “Secondary osteoporosis” is osteoporosis caused by conditions, diseases, or medications other than estrogen deficiency or aging.  

The reported prevalence of secondary osteoporosis varies depending on the study population, the extent of the medical evaluation, and definitions for laboratory abnormalities. It is likely that many or most patients with primary osteoporosis have clinically significant contributing factors that may influence patient management. In a study of North American women receiving osteoporosis therapy, it was found that 52% had vitamin D inadequacy, defined as serum 25-hydroxyvitamin D (25-OH-D) levels less than 30 ng/ml (38). In another study of patients referred to an osteoporosis clinic, over 60% were found to have elements of secondary osteoporosis when vitamin D deficiency was very conservatively defined as serum 25-OH-D level less than 12.5 ng/ml (39,40). In the same study, the number of patients with secondary osteoporosis was much higher when vitamin D inadequacy was more appropriately defined as serum 25-OH-D less than 33 ng/ml (41,42).

It has been proposed by some that a bone density that is less than expected compared to an age- and sex-matched population, as represented by a low Z-score (e.g., less than -2.0), suggests a high likelihood of secondary osteoporosis and should be one of the triggers for further investigation (43,44). While there may be some merit to this concept, there are few if any studies validating the use of a Z-score cutoff for this purpose. Since secondary osteoporosis is common, a more effective strategy is to screen all patients with osteoporosis for contributing factors (45). The results of a metabolic evaluation may identify previously unrecognized diseases and conditions that require treatment in addition to, or instead of, standard osteoporosis pharmacological therapy.

Depending on the patient population being studied, different causes of secondary osteoporosis may predominate. Calcium deficiency, vitamin D deficiency, and sedentary lifestyle are common contributing factors for all patients. In women referred to an osteoporosis clinic with previously recognized medications or diseases contributing to osteoporosis, the most common were history of glucocorticoid use (36%), premature ovarian failure (21%), history of unintentional weight loss (10%), history of alcoholism (10%), and history of liver disease (10%) (39). When patients without previously recognized contributing factors were evaluated at the same specialty clinic, most (55%) were found to have vitamin D deficiency or insufficiency (serum 25-OH-D less than 33 ng/ml) (42), while 10% had hypercalciuria, 8% had malabsorption, and 7% had primary or secondary hyperparathyroidism (39). In men, the most common secondary causes of osteoporosis are long-term glucocorticoid use, hypogonadism, and alcoholism (46,47). The increasing use of aromatase inhibitor therapy for breast cancer in women (48) and androgen deprivation therapy for prostate cancer in men (49) is now recognized as an important factor in the development of osteoporosis in these patients. Other common causes for low BMD and fractures include multiple myeloma (50), gastric bypass surgery (51) and gastric resection (52). Treatable but easily missed secondary causes of osteoporosis include asymptomatic primary hyperparathyroidism (53), subclinical hyperthyroidism (54), mild Cushing’s syndrome (55), and malabsorption due to unrecognized celiac disease (56). Table 9 lists some of the causes of low BMD by category.

Table 9. Causes of Low Bone Mineral Density

Inherited

Nutritional

Endocrine

Drugs

Other

Osteogenesis imperfecta

Malabsorption

Hypogonadism

Glucocorticoids

Multiple myeloma

Homocystinuria

Chronic liver disease

Hyperthyroidism

Anticonvulsants

Rheumatoid arthritis

Marfan’s syndrome

Alcoholism

Hyperparathyroidism

Long-term heparin

Systemic mastocytosis

Hypophosphatasia

Calcium deficient diet

Cushing’s syndrome

Excess thyroid

Immobilization

 

Vitamin D deficiency

Eating disorder

GnRH agonists

 
     

Aromatase inhibitors

 

A variety of testing strategies have been proposed as screening for all patients with osteoporosis (37,39,42,45,57,58). A minimal cost-effective work-up for all patients consists of a complete blood count (CBC), serum calcium, phosphorus, creatinine with calculated or measured creatinine clearance, alkaline phosphatase, 24-hour urinary calcium, and serum 25-OH-D. Other laboratory tests may be indicated according to the patient’s clinical profile and the practice setting. A summary of useful common and uncommon laboratory studies with comments on their possible skeletal significance is provided below.

CLINICAL CASE

A 52-year-old postmenopausal woman with a history of irritable bowel syndrome (IBS) and a family history of osteoporosis (mother with hip fracture) is found to have osteoporosis on a DXA study. Evaluation for secondary causes of osteoporosis is unremarkable except for mild iron deficiency anemia (a long-standing problem, previously attributed to heavy menses) and a low 24-hour urinary calcium of 30 mg, with adequate calcium intake and normal renal function. Serum 25-OH-D is 29 ng/ml. Additional work-up shows a high titer of IgA endomysial antibodies consistent with celiac disease. This diagnosis is confirmed by a small bowel biopsy showing villous atrophy. She is started on a gluten-free diet, resulting in resolution of her “IBS” symptoms and correction of her anemia. One year later, with no pharmacological therapy for osteoporosis, there is a statistically significant BMD increase of 9% at the lumbar spine.

Celiac disease may result in osteoporosis due to calcium malabsorption, even in the absence of gastrointestinal symptoms. Treatment is strict lifelong adherence to a gluten-free diet, which may sometimes be followed by a substantial increase in BMD, as seen in this patient. A 24-hour urinary calcium is an inexpensive screening test for calcium malabsorption that should be considered a routine part of the initial evaluation of osteoporosis.

BASIC BLOOD TESTS

CBC- Anemia may be seen in patients with myeloma or malnutrition

Sedimentation rate- May be elevated with myeloma.

Calcium- Among the many causes of hypercalcemia are primary and secondary hyperparathyroidism, hyperthyroidism, renal failure, vitamin D intoxication, and Paget’s disease. Hypocalcemia may be seen with vitamin D deficiency and hyperphosphatemia.

Phosphorus- Hyperphosphatemia may occur with hypoparathyroidism, renal failure, and possibly with bisphosphonate therapy. Hypophosphatemia may be seen with primary or secondary hyperparathyroidism, vitamin D deficiency, tumor induced osteomalacia, and X-linked hypophosphatemia.

Alkaline phosphatase- High values can be seen with healing fractures, osteomalacia, and Paget’s disease, as well as occurring normally in growing children. Low values occur with hypophosphatasia, a rare genetic disorder that causes impaired mineralization of bone and dental tissue.

Vitamin D- The test that best reflects vitamin D stores is the serum 25-OH-D. While there is no consensus on the optimal range of serum 25-OH-D, a reasonable target for good skeletal health is approximately 30-50 ng/ml. This is likely to maximize intestinal absorption of calcium and minimize serum PTH levels. Interpretation of serum 25-OH-D levels is confounded by assay variability (59). Serum 1,25-(OH)2-D3 is usually not helpful in the evaluation of osteoporosis patients, unless there are concerns regarding renal conversion of 25-OH-D to 1,25-(OH)2-D3. Deficiency or insufficiency of vitamin D is very common and play a role in the pathogenesis of osteoporosis and osteomalacia.

Creatinine- Chronic kidney disease may cause an elevated creatinine level and renal osteodystrophy. Elderly patients with small muscle mass may have impaired renal function with a “normal” serum creatinine. An estimated glomerular filtration rate can be calculated using one of many formulae, such as that of Cockcroft and Gault (60) or modification of diet in renal disease study equation (61). Impaired renal function not only has adverse skeletal effects but also raises considerations regarding the type and dose of pharmacologic agents used.

TSH- Hyperthyroidism from any cause, including excess thyroid replacement, can usually be recognized by a low TSH. High bone turnover associated hyperthyroidism is associated with loss of bone mass.

Liver enzymes- Abnormalities may be caused by chronic liver disease, which is a risk factor for osteoporosis.

BASIC URINE TESTS

Urinalysis. Proteinuria may occur with multiple myeloma or chronic kidney disease. Abnormal cells may suggest kidney disease.

24-hour urine for calcium- A well-collected 24-hour urine for calcium is a helpful screening test for identifying patients with common disorders of calcium metabolism. The “normal” range of urinary calcium is not well established and varies according to many dietary factors and estrogen status in women (62,63). As a “rule of thumb,” urinary calcium may be considered elevated when it is greater than 250 mg per 24 hours in women; greater than 300 mg per 24 hours in men; or greater than 4 mg/kg body weight per 24 hours in either sex. It has been proposed that hypercalciuria can be easily classified as “renal” (renal calcium leak), “resorptive” (excess skeletal loss of calcium) or “absorptive” (increased intestinal absorption of calcium) (64). However, in clinical practice, these distinctions are not so easily established. Idiopathic hypercalciuria, perhaps the most common type of hypercalciuria (65), may be diagnosed if there are no underlying medical disorders (e.g., hyperparathyroidism, vitamin D toxicity, Paget’s disease of bone, multiple myeloma, sarcoidosis) and no obvious dietary excesses (e.g., calcium, sodium, protein, carbohydrates, alcohol) or deficiencies (e.g., phosphate, potassium) that are associated with hypercalciuria (63). In the absence of dietary calcium deficiency, vitamin D deficiency, malabsorption, liver disease, or chronic renal failure, low urinary calcium (less than 50 mg per 24 hours in women or men) is suggestive of calcium malabsorption and warrants further investigation. Celiac disease is a common (66) cause of asymptomatic malabsorption in osteoporosis that is treatable with a gluten-free diet (56).

ADDITIONAL STUDIES IN SELECTED PATIENTS

Celiac antibodies- Anti-endomysial antibody and tissue transglutaminase antibody are currently the serological markers of choice, with a higher sensitivity and specificity than anti-gliadin antibody and anti-reticulin antibody (67). If a serological marker is abnormal, or if there is a high clinical suspicion for celiac disease, the patient should be referred for endoscopy and small bowel biopsy.

Intact PTH- This may be elevated in patients with primary hyperparathyroidism, vitamin D deficiency, or renal failure.

Serum and urine protein electrophoresis- These are helpful tests to screen for possible multiple myeloma. If an M-component is identified, referral for bone marrow aspiration may be indicated.

Dexamethasone suppression test or 24-hour urinary free cortisol- This is helpful to evaluate patients with suspected Cushing’s syndrome.

Serum total or free testosterone level- May be helpful in the assessment of men with osteoporosis.

Serum homocysteine- Elevated circulating homocysteine levels are associated with increased risk of fracture (68,69). It is unknown whether reduction of homocysteine levels by increasing dietary intake of folic acid and vitamins B6 and B12 reduces the risk of fracture.

Serum tryptase and 24-hour urine for N-methylhistamine- Systemic mastocytosis is a rare cause of osteoporosis that can be diagnosed by a biopsy of typical skin lesions of urticaria pigmentosa, when present. Patients with systemic mastocytosis may sometimes present with osteoporosis and no other manifestations of the disease (70,71). When this disorder is suspected but skin lesions are not present, the finding of an elevated serum tryptase and/or urinary N-methyl histamine can be helpful, especially during or soon after a symptomatic episode of histamine release. However, normal values do not exclude the diagnosis. Bone marrow aspiration or biopsy, or non-decalcified double tetracycline labeled transiliac bone biopsy, may be necessary to confirm the diagnosis.

Serum bicarbonate- Renal tubular acidosis (RTA) has been associated with osteoporosis (72). With distal (type I) RTA, the serum bicarbonate is usually less than 15 mmol/l with a urine pH greater than 5.5.

BONE TURNOVER MARKERS

Bone turnover markers (BTMs) are noninvasive laboratory tests of serum and urine that are readily available in clinical practice. While BTMs cannot be used to diagnose osteoporosis or determine the cause to osteoporosis, they have been very helpful in the research to understand the pathophysiology of osteoporosis and other skeletal diseases and the mechanism of action of interventions used in the treatment of osteoporosis. In clinical practice, BTMs offer the potential of predicting fracture risk independently of BMD and may be useful in monitoring the metabolic effects of therapy (73). Drugs that are approved for the management of osteoporosis modulate bone remodeling in ways that are reflected by changes in BTMs. A decrease in BTMs with antiresorptive therapy is predictive of a subsequent increase in BMD (74) and reduction in fracture risk (75-77). The magnitude of BTM decrease with antiresorptive therapy is significantly associated with the level of fracture risk reduction, although the proportion of treatment effect due to the reduction in BTMs appears to vary according to the type of drug used (79). With teriparatide, a bone anabolic agent, an early increase in BTM levels is predictive of a subsequent increase in BMD (80).

Markers of bone resorption are mostly fragments of type I collagen, the main component of the organic bone matrix, that are released during osteoclastic bone resorption. These are measured in the serum or urine, with those available for clinical use including N-telopeptide of type I collagen (NTX), C-telopeptide of type I collagen (CTX), deoxypyridinoline (DPD), and pyridinoline (PYD). Bone formation markers are proteins secreted by osteoblasts or byproducts of type I collagen production by osteoblasts. They are measured in the serum and include bone specific alkaline phosphatase (BSAP), N-terminal propeptide of type I collagen (P1NP), and osteocalcin. CTX and P1NP have been proposed as the reference BTMs for clinical trials.

(Samuel Vasikaran 1, Cyrus Cooper, Richard Eastell, Andrea Griesmacher, Howard A Morris, Tommaso Trenti, John A Kanis, International Osteoporosis Foundation and International Federation of Clinical Chemistry and Laboratory Medicine position on bone marker standards in osteoporosis, Clin Chem Lab Med. 2011 Aug;49(8):1271-4)

Clinical use of BTMs requires knowledge of their limitations as well as benefits. BTMs are subject to pre-analytical (biological) and analytical variability. Uncontrollable sources of pre-analytical variability include age, sex, menopausal status, pregnancy, lactation, fractures, co-existing diseases (e.g., diabetes mellitus, impaired renal function, and liver disease), drugs (e.g., glucocorticoids, anticonvulsants, and gonadotropin hormone releasing agonists) and immobility (81). Controllable pre-analytical sources of variability include time of day (circadian variability), fasting status, and exercise (81). Analytical sources of variability include specimen processing (e.g., collection, handling, and storage) (82). Between-laboratory variability may be large (reported to be as much as a 7.3-fold difference), casting doubt on the validity of comparing specimens sent to different labs (83). Reference ranges for BTMs are not well established and may vary according to the population tested, the type of BTM, and the circumstances under which it is collected and processed.

In order to compare BTMs measurements longitudinally, it would be ideal to know the least significant change (LSC) and use this in a manner similar to what should be (but is probably not) common practice with DXA. However, the standards for calculating an LSC for a BTM are not as clear as with DXA, and the opportunity to do precision assessment for a BTM may not present itself. The NOF recommends calculating the LSC with a 95% level of confidence for each BTM used by multiplying the laboratory-provided precision error by 2.77 (84). The NOF also recommends that specimens be obtained in the early morning following an overnight fast to reduce biological variability, with serial measurements to be obtained at the same time of day and ideally during the same season of the year. The Belgian Bone Club suggests using an estimated LSC of assuming an LSC of about 30% for serum BTMs and about 50-60% for urine BTMs (73). While the LSC for BTMs is almost always greater than for DXA, the magnitude of likely change (85) is greater than DXA, with the ‘signal to noise ratio’ that may be as good or even better than DXA. One strategy for the use of BTMs to monitor patients on antiresorptive therapy is to use absolute values rather that percent changes, as follows: treatment effect can be considered optimal when serum CTX has decreased by 100 ng/L or is below 280 ng/L, or when P1NP has decreased by 10 mcg/L or is less than 35 mcg/L. (Andreas Fontalis, Richard Eastell, The challenge of long-term adherence: The role of bone turnover markers in monitoring bisphosphonate treatment of osteoporosis, Bone. 2020 Jul;136:115336).

Evidence-based guidelines for the clinical use of BTMs have been developed by organizations that include the NOF (7), Belgian Bone Club (73), and the Japan Osteoporosis Society (86). The NOF guidelines state that “suppression of biochemical markers of bone turnover after 3-6 months of specific antiresorptive osteoporosis therapies, and biochemical marker increases after 1-3 months of specific anabolic therapies, have been predictive of greater BMD responses and in some cases fracture risk reduction in large clinical trials. Biochemical marker changes in individuals must exceed the LSC in order to be clinically meaningful.” The Belgian Bone Club suggests that “early changes in BTM can be used to measure the clinical efficacy of an antiresorptive treatment and to reinforce patient compliance,” with goal of decreasing the BTM to the premenopausal range or at least achieving a decrease as great as the LSC. The Japanese guidelines indicate that “the argument for measuring bone turnover markers to evaluate the therapeutic effects of bone antiresorptive medications can be justified,” but go on to state that there is insufficient evidence for their use with medications having other mechanisms of action (86).

A significant change of a BTM level in the appropriate direction following therapy is evidence that the patient is taking the drug regularly, taking it correctly, and that it is being absorbed and having the expected effect in modulating bone remodeling. Failure to achieve such a change in the BTM level is cause for concern and suggests that evaluation and possibly a reconsideration of treatment should be considered (87). The use of BTMs allows assessment of drug effect sooner than with DXA, so that evaluation and corrective action, if needed, can be taken early in the course of therapy rather than later. Monitoring BTMs, especially in association with regular contact by a healthcare provider, may improve persistence with therapy (88). Despite the well-described limitations of BTMs (89), there is emerging support for their use in clinical practice, particularly in the assessment of response to therapy (90,91). Clinicians who are familiar with the benefits and limitations of BTMs may find them a helpful tool, in association with BMD testing, for managing patients with osteoporosis.

IMAGING STUDIES

Standard X-rays are used to diagnose fractures of all types and may sometimes suggest secondary causes of osteoporosis. Pseudofractures (Looser’s zones) are radiolucent lines running perpendicular to the bone cortex that may be seen in patients with osteomalacia. These probably represent stress fractures that have healed with poorly mineralized osteoid. Punctate radiolucencies may be seen in bone X-rays of patients with systemic mastocytosis. Primary hyperparathyroidism may cause bone cysts, subperiosteal bone resorption, brown tumors, and demineralization (‘salt and pepper’ pattern) of the skull. MRI, CT scanning, or nuclear imaging may be used to detect stress fractures not visible on X-ray. MRI of the spine is commonly used prior to vertebroplasty or kyphoplasty to determine the age of the fracture, the likelihood of the fracture being from causes other than osteoporosis, and whether there is retropulsion of bony fragments than could impair neurological function.

BONE BIOPSY

Non-decalcified double tetracycline labeled iliac crest bone biopsy is rarely used in clinical practice but may be helpful with difficult diagnostic problems. In the evaluation of renal osteodystrophy, a bone biopsy can distinguish between high turnover and low turnover bone disease, and possibly be an aid in the selection of therapy. With infiltrative disorders of bone, such as systemic mastocytosis, a bone biopsy or bone marrow aspiration may sometimes be the only way to make the diagnosis. In patients who are not responding to therapy as expected, or in patients with unusual presentations of osteoporosis, a bone biopsy may be indicated. Bone biopsies are required by the FDA for safety monitoring in clinical trials of osteoporosis drugs.

SUMMARY

Osteoporosis is a common skeletal disease with serious clinical consequences. Effective management of skeletal health includes appropriate selection of patients for bone density testing and assessment of risk factors for fracture. Prior to treatment, and when response to treatment is suboptimal, patients should be evaluated for secondary causes of osteoporosis. All reversible factors should be corrected and treatment should be individualized based on the clinical circumstances.

REFERENCES

  1. Klibanski A, Adams-Campbell L, Bassford T, Blair SN, Boden SD, Dickersin K, Gifford DR, Glasse L, Goldring SR, Hruska K, Johnson SR, McCauley LK, Russell WE, Osteopor NCDP 2001 Osteoporosis prevention, diagnosis, and therapy. JAMA 285(6):785-795.
  2. US Department of Health and Human Services 2004 Bone Health and Osteoporosis: A Report of the Surgeon General. US Department of Health and Human Services, Office of the Surgeon General, Rockville, MD.
  3. World Health Organisation 1994 Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. WHO, Geneva, Switzerland.
  4. Shuhart CR, Yeap SS, Anderson PA, Jankowski LG, Lewiecki EM, Morse LR et al. Executive Summary of the 2019 ISCD Position Development Conference on Monitoring Treatment, DXA Cross-calibration and Least Significant Change, Spinal Cord Injury, Periprosthetic and Orthopedic Bone Health, Transgender Medicine, and Pediatrics. J Clin Densitom. 2019;22(4):453-71. doi:10.1016/j.jocd.2019.07.001.
  5. Lewiecki EM, Binkley N, Petak SM 2006 DXA Quality Matters. J Clin Densitom 9(4):388-392.
  6. Siris ES, Adler R, Bilezikian J, Bolognese M, Dawson-Hughes B, Favus MJ, Harris ST, Jan de Beur SM, Khosla S, Lane NE, Lindsay R, Nana AD, Orwoll ES, Saag K, Silverman S, Watts NB 2014 The clinical diagnosis of osteoporosis: a position statement from the National Bone Health Alliance Working Group. Osteoporos Int 25(5):1439-43.
  7. Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, Lindsay R 2014 Clinician's Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int 25(10):2359-81.
  8. Qaseem A, Snow V, Shekelle P, Hopkins R, Jr., Forciea MA, Owens DK 2008 Screening for osteoporosis in men: a clinical practice guideline from the American College of Physicians. Ann.Intern.Med 148(9):680-684.
  9. Kanis JA, McCloskey EV, Johansson H, Strom O, Borgstrom F, Oden A 2008 Case finding for the management of osteoporosis with FRAX--assessment and intervention thresholds for the UK. Osteoporos.Int. 19(10):1395-1408.
  10. Kanis JA, Burlet N, Cooper C, Delmas PD, Reginster JY, Borgstrom F, Rizzoli R 2008 European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int 19(4):399-428.
  11. Marshall D, Johnell O, Wedel H 1996 Meta-analysis of how well measures of bone mineral density predict occurrence of osteoporotic fractures. BMJ 312(7041):1254-1259.
  12. Wainwright SA, Marshall LM, Ensrud KE, Cauley JA, Black DM, Hillier TA, Hochberg MC, Vogt MT, Orwoll ES 2005 Hip fracture in women without osteoporosis. J Clin Endocrinol Metab 90(5):2787-2793.
  13. Lewiecki EM, Laster AJ 2006 Clinical applications of vertebral fracture assessment by dual-energy X-ray absorptiometry. J Clin Endocrinol Metab 91(11):4215-4222.
  14. Genant HK, Wu CY, Van Kuijk C, Nevitt MC 1993 Vertebral fracture assessment using a semiquantitative technique. J Bone Miner Res 8(9):1137-1148.
  15. Schousboe JT, DeBold CR 2006 Reliability and accuracy of vertebral fracture assessment with densitometry compared to radiography in clinical practice. Osteoporos Int 17(2):281-289.
  16. Lewiecki EM, Binkley N, Morgan SL, Shuhart CR, Camargos BM, Carey JJ, et al 2016  Best Practices for Dual-Energy X-ray Absorptiometry Measurement and Reporting: International Society for Clinical Densitometry Guidance.  J Clin Densitom19 (2) : 127-40.
  17. International Society for Clinical Densitometry, Facility Accreditation.  Available at www.iscd.org/accreditation.
  18. Lewiecki EM 2005 Update on bone density testing. Curr.Osteoporos Rep. 3(4):136-142.
  19. Lotz JC, Cheal EJ, Hayes WC 1991 Fracture prediction for the proximal femur using finite element models: Part I--Linear analysis. J Biomechan.Eng. 113:353-360.
  20. Cranney A, Tugwell P, Wells G, Guyatt G 2002 Systematic reviews of randomized trials in osteoporosis: Introduction and methodology. Endocrine Reviews 23(4):497-507.
  21. Wasnich RD, Miller PD 2000 Antifracture efficacy of antiresorptive agents are related to changes in bone density. Journal of Clinical Endocrinology and Metabolism 85(1):231-236.
  22. Mazess R, Chesnut CH, III, McClung M, Genant H 1992 Enhanced precision with dual-energy X-ray absorptiometry. Calcif.Tissue Int 51(1):14-17.
  23. Njeh CF, Fuerst T, Hans D, Blake GM, Genant HK 1999 Radiation exposure in bone mineral density assessment. Applied Radiation & Isotopes. 50(1):215-236.
  24. Bonnick SL 2004 Bone densitometry in clinical practice- application and interpretation, 2nd ed ed. Humana Press, Totowa, N.J.
  25. Lewiecki EM, Richmond B, Miller PD 2006 Uses and misuses of quantitative ultrasonography in managing osteoporosis. Cleve.Clin J Med 73(8):742-752.
  26. Engelke K, Adams JE, Armbrecht G, Augat P, Bogado CE, Bouxsein ML, Felsenberg D, Ito M, Prevrhal S, Hans DB, Lewiecki EM 2008 Clinical use of quantitative computed tomography and peripheral quantitative computed tomography in the management of osteoporosis in adults: the 2007 ISCD Official Positions. J.Clin.Densitom. 11(1):123-162.
  27. Faulkner KG, von Stetten E, Miller P 1999 Discordance in patient classification using T-scores. J.Clin.Densitom. 2:343-350.
  28. Kanis JA, Oden A, Johnell O, Johansson H, De Laet C, Brown J, Burckhardt P, Cooper C, Christiansen C, Cummings S, Eisman JA, Fujiwara S, Gluer C, Goltzman D, Hans D, Krieg MA, La CA, McCloskey E, Mellstrom D, Melton LJ, III, Pols H, Reeve J, Sanders K, Schott AM, Silman A, Torgerson D, van ST, Watts NB, Yoshimura N 2007 The use of clinical risk factors enhances the performance of BMD in the prediction of hip and osteoporotic fractures in men and women. Osteoporos.Int. 18(8):1033-1046.
  29. Kanis JA, on behalf of the World Health Organization Scientific Group 2007 Assessment of osteoporosis at the primary health-care level. Technical Report. World Health Organization Collaborating Centre for Metabolic Bone Diseases, University of Sheffield, UK: Printed by the University of Sheffield.
  30. Leslie WD, Lix LM, Johansson H, Oden A, McCloskey E, Kanis JA 2011 Spine-hip discordance and fracture risk assessment: a physician-friendly FRAX enhancement. Osteoporos Int 22:839-847.
  31. Vogt TM, Ross PD, Palermo L, Musliner T, Genant HK, Black D, Thompson DE 2000 Vertebral fracture prevalence among women screened for the Fracture Intervention Trial and a simple clinical tool to screen for undiagnosed vertebral fractures. Fracture Intervention Trial Research Group. Mayo Clin Proc. 75(9):888-896.
  32. Gunnes M, Lehmann EH, Mellstrom D, Johnell O 1996 The relationship between anthropometric measurements and fractures in women. Bone 19(4):407-413.
  33. Siminoski K, Jiang G, Adachi JD, Hanley DA, Cline G, Ioannidis G, Hodsman A, Josse RG, Kendler D, Olszynski WP, Ste Marie LG, Eastell R 2005 Accuracy of height loss during prospective monitoring for detection of incident vertebral fractures. Osteoporos Int 16(4):403-410.
  34. Margolis KL, Ensrud KE, Schreiner PJ, Tabor HK 2000 Body size and risk for clinical fractures in older women. Study of Osteoporotic Fractures Research Group. Ann.Intern Med 133(2):123-127.
  35. de Laet C, Kanis JA, Oden A, Johanson H, Johnell O, Delmas P, Eisman JA, Kroger H, Fujiwara S, Garnero P, McCloskey EV, Mellstrom D, Melton LJ, III, Meunier PJ, Pols HA, Reeve J, Silman A, Tenenhouse A 2005 Body mass index as a predictor of fracture risk: a meta-analysis. Osteoporos.Int. 16(11):1330-1338.
  36. Ensrud KE, Ewing SK, Stone KL, Cauley JA, Bowman PJ, Cummings SR, Gr SOFR 2003 Intentional and unintentional weight loss increase bone loss and hip fracture risk in older women. Journal of the American Geriatrics Society 51(12):1740-1747.
  37. Lewiecki EM 2013 Evaluation of the patient at risk for osteoporosis. In: Marcus R, Feldman D, Dempster DW, Luckey M, Cauley JA (eds.) Osteoporosis, Fourth ed., vol. 2. Elsevier, Waltham, Massachusetts, USA, pp 1481-1504.
  38. Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, Petruschke RA, Chen E, De Papp AE 2005 Prevalence of Vitamin D inadequacy among postmenopausal North American women receiving osteoporosis therapy. J Clin Endocrinol Metab 90(6):3215-3224.
  39. Tannenbaum C, Clark J, Schwartzman K, Wallenstein S, Lapinski R, Meier D, Luckey M 2003 Yield of laboratory testing to identify secondary contributors to osteoporosis in otherwise healthy women. J Clin Endocrinol Metab 87:4431-4437.
  40. Wagman RB, Marcus R 2002 Beyond bone mineral density-navigating the laboratory assessment of patients with osteoporosis. J Clin Endocrinol Metab 87(10):4429-4430.
  41. Barzel US 2003 Recommended testing in patients with low bone density. J Clin Endocrinol Metab 88(3):1404-1405.
  42. 42Luckey MM, Tannenbaum C 2003 Authors' response: Recommended testing in patients with low bone density. J Clin Endocrinol Metab 88(3):1405.
  43. Harper KD, Weber TJ 1998 Secondary osteoporosis - Diagnostic considerations. Endocrinology and Metabolism Clinics of North America 27(2):325-348.
  44. Eastell R 1998 Treatment of postmenopausal osteoporosis. N.Engl.J.Med. 338(11):736-746.
  45. Favus MJ 2005 Postmenopausal osteoporosis and the detection of so-called secondary causes of low bone density. J Clin Endocrinol Metab 90(6):3800-3801.
  46. Looker AC, Orwoll ES, Johnston CCJ, Lindsay RL, Wahner HW, Dunn WL, Calvo MS, Harris TB, Heyse SP 1997 Prevalence of low femoral bone density in older U.S. adults from NHANES III. J.Bone Miner.Res. 12:1761-1768.
  47. Orwoll ES, Klein RF 1995 Osteoporosis in men. Endocr.Rev. 16:87-116.
  48. Lester J, Coleman R 2005 Bone loss and the aromatase inhibitors. Br.J Cancer 93 Suppl 1:S16-S22.
  49. Daniell HW 2001 Osteoporosis due to androgen deprivation therapy in men with prostate cancer. Urology 58(2 Suppl 1):101-107.
  50. Melton LJ, III, Kyle RA, Achenbach SJ, Oberg AL, Rajkumar SV 2005 Fracture risk with multiple myeloma: a population-based study. J Bone Miner Res 20(3):487-493.
  51. De Prisco C, Levine SN 2005 Metabolic bone disease after gastric bypass surgery for obesity. Am J Med Sci. 329(2):57-61.
  52. Adachi Y, Shiota E, Matsumata T, Iso Y, Yoh R, Kitano S 2000 Osteoporosis after gastrectomy: bone mineral density of lumbar spine assessed by dual-energy X-ray absorptiometry. Calcif.Tissue Int 66(2):119-122.
  53. Cormier C, Souberbielle JC, Kahan A 2004 Primary hyperparathyroidism and osteoporosis in 2004. Joint Bone Spine 71(3):183-189.
  54. Földes J, Tarján G, Szathmari M, Varga F, Krasznai I, Horvath C 1993 Bone mineral density in patients with endogenous subclinical hyperthyroidism:  Is this thyroid status a risk factor for osteoporosis. Clin.Endocrinol.(Oxf.) 39:521-527.
  55. Mancini T, Doga M, Mazziotti G, Giustina A 2004 Cushing's Syndrome and Bone. Pituitary. 7:249-252.
  56. Bianchi ML, Bardella MT 2002 Bone and celiac disease. Calcified Tissue International 71(6):465-471.
  57. Freitag A, Barzel US 2002 Differential diagnosis of osteoporosis. Gerontology 48(2):98-102.
  58. Orlic ZC, Raisz LG 1999 Causes of secondary osteoporosis. J Clin Densitom 2(1):79-92.
  59. Binkley N, Krueger D, Cowgill CS, Plum L, Lake E, Hansen KE, Deluca HF, Drezner MK 2004 Assay variation confounds the diagnosis of hypovitaminosis D: a call for standardization. J Clin Endocrinol Metab 89(7):3152-3157.
  60. Cockcroft DW, Gault MH 1976 Prediction of creatinine clearance from serum creatinine. Nephron 16(1):31-41.
  61. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, Kusek JW, Van LF 2006 Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann.Intern.Med. 145(4):247-254.
  62. Heaney RP, Recker RR, Ryan RA 1999 Urinary calcium in perimenopausal women: normative values. Osteoporos Int 9(1):13-18.
  63. Audran M, Legrand E 2000 Hypercalciuria. Joint Bone Spine 67(6):509-515.
  64. Pak CY, Kaplan R, Bone H, Townsend J, Waters O 1975 A simple test for the diagnosis of absorptive, resorptive and renal hypercalciurias. N.Engl.J Med 292(10):497-500.
  65. Pacifici R 1997 Idiopathic hypercalciuria and osteoporosis--distinct clinical manifestations of increased cytokine-induced bone resorption? J Clin Endocrinol Metab 82(1):29-31.
  66. Fasano A, Berti I, Gerarduzzi T, Not T, Colletti RB, Drago S, Elitsur Y, Green PH, Guandalini S, Hill ID, Pietzak M, Ventura A, Thorpe M, Kryszak D, Fornaroli F, Wasserman SS, Murray JA, Horvath K 2003 Prevalence of celiac disease in at-risk and not-at-risk groups in the United States: a large multicenter study. Arch.Intern.Med. 163(3):286-292.
  67. Alaedini A, Green PH 2005 Narrative review: celiac disease: understanding a complex autoimmune disorder. Ann.Intern Med 142(4):289-298.
  68. McLean RR, Jacques PF, Selhub J, Tucker KL, Samelson EJ, Broe KE, Hannan MT, Cupples LA, Kiel DP 2004 Homocysteine as a predictive factor for hip fracture in older persons. N.Engl.J Med 350(20):2042-2049.
  69. van Meurs JB, Dhonukshe-Rutten RA, Pluijm SM, van der KM, de JR, Lindemans J, de Groot LC, Hofman A, Witteman JC, van Leeuwen JP, Breteler MM, Lips P, Pols HA, Uitterlinden AG 2004 Homocysteine levels and the risk of osteoporotic fracture. N.Engl.J Med 350(20):2033-2041.
  70. Chines A, Pacifici R, Avioli LV, Teitelbaum SL, Korenblat PE 1991 Systemic mastocytosis presenting as osteoporosis: a clinical and histomorphometric study. J Clin Endocrinol Metab 72(1):140-144.
  71. Lidor C, Frisch B, Gazit D, Gepstein R, Hallel T, Mekori YA 1990 Osteoporosis as the sole presentation of bone marrow mastocytosis. J Bone Miner Res 5(8):871-876.
  72. Weger M, Deutschmann H, Weger W, Kotanko P, Skrabal F 1999 Incomplete renal tubular acidosis in 'primary' osteoporosis. Osteoporos Int 10(4):325-329.
  73. Bergmann P, Body JJ, Boonen S, Boutsen Y, Devogelaer JP, Goemaere S, Kaufman JM, Reginster JY, Gangji V 2009 Evidence-based guidelines for the use of biochemical markers of bone turnover in the selection and monitoring of bisphosphonate treatment in osteoporosis: a consensus document of the Belgian Bone Club. Int J Clin Pract 63(1):19-26.
  74. Greenspan SL, Parker RA, Ferguson L, Rosen HN, Maitland-Ramsey L, Karpf DB 1998 Early changes in biochemical markers of bone turnover predict the long-term response to alendronate therapy in representative elderly women: a randomized clinical trial. J Bone Miner Res 13:1431-1438.
  75. Eastell R, Barton I, Hannon RA, Chines A, Garnero P, Delmas PD 2003 Relationship of early changes in bone resorption to the reduction in fracture risk with risedronate. J Bone Miner Res 18:1051-1056.
  76. Bauer DC, Black DM, Garnero P, Hochberg M, Ott S, Orloff J, Thompson DE, Ewing SK, Delmas PD 2004 Change in bone turnover and hip, non-spine, and vertebral fracture in alendronate-treated women: the fracture intervention trial. J Bone Miner Res 19(8):1250-1258.
  77. Sarkar S, Reginster JY, Crans GG, Diez-Perez A, Pinette KV, Delmas PD 2004 Relationship between changes in biochemical markers of bone turnover and BMD to predict vertebral fracture risk. J Bone Miner Res 19(3):394-401.
  78. Hochberg MC, Greenspan S, Wasnich RD, Miller P, Thompson DE, Ross PD 2002 Changes in bone density and turnover explain the reductions in incidence of nonvertebral fractures that occur during treatment with antiresorptive agents. J Clin Endocrinol.Metab 87(4):1586-1592.
  79. Bouxsein ML, Delmas PD 2008 Considerations for development of surrogate endpoints for antifracture efficacy of new treatments in osteoporosis: a perspective. J Bone Miner Res 23(8):1155-1167.
  80. Chen P, Satterwhite JH, Licata AA, Lewiecki EM, Sipos AA, Misurski DM, Wagman RB 2005 Early changes in biochemical markers of bone formation predict BMD response to teriparatide in postmenopausal women with osteoporosis. J Bone Miner Res 20(6):962-970.
  81. Hannon R, Eastell R 2000 Preanalytical variability of biochemical markers of bone turnover. Osteoporos Int 11 Suppl 6:S30-S44.
  82. Seibel MJ 2005 Biochemical markers of bone turnover: part I: biochemistry and variability. Clin Biochem.Rev 26(4):97-122.
  83. Seibel MJ, Lang M, Geilenkeuser WJ 2001 Interlaboratory variation of biochemical markers of bone turnover. Clin Chem. 47(8):1443-1450.
  84. National Osteoporosis Foundation 2013, Version 3, Released February 25, 2014 Clinician's Guide to Prevention and Treatment of Osteoporosis, .
  85. Srivastava AK, Vliet EL, Lewiecki EM, Maricic M, Abdelmalek A, Gluck O, Baylink DJ 2005 Clinical use of serum and urine bone markers in the management of osteoporosis. Curr.Med Res Opin. 21(7):1015-1026.
  86. Nishizawa Y, Nakamura T, Ohta H, Kushida K, Gorai I, Shiraki M, Fukunaga M, Hosoi T, Miki T, Chaki O, Ichimura S, Nakatsuka K, Miura M 2005 Guidelines for the use of biochemical markers of bone turnover in osteoporosis (2004). J Bone Miner Metab 23(2):97-104.
  87. Lewiecki EM, Watts NB 2008 Assessing response to osteoporosis therapy. Osteoporos Int 19(10):1363-1368.
  88. Clowes JA, Peel NF, Eastell R 2004 The impact of monitoring on adherence and persistence with antiresorptive treatment for postmenopausal osteoporosis: a randomized controlled trial. J Clin Endocrinol Metab 89(3):1117-1123.
  89. Meier C, Seibel MJ, Kraenzlin ME 2009 Use of bone turnover markers in the real world: are we there yet? J Bone Miner Res 24(3):386-388.
  90. Lewiecki EM, Baim S, Bilezikian JP, Eastell R, LeBoff MS, Miller PD 2009 2008 Santa Fe Bone Symposium: update on osteoporosis. J Clin Densitom 12(2):135-157.
  91. Singer FR, Eyre DR 2008 Using biochemical markers of bone turnover in clinical practice. Cleve.Clin J Med 75(10):739-750.