Archives

Growth hormone Stimulation Tests in Assessing Adult Growth Hormone Deficiency

ABSTRACT

 

Adult growth hormone deficiency (GHD) is a clinical syndrome that can manifest either as isolated or associated with additional pituitary hormone deficiencies. Its clinical features are subtle and nonspecific, requiring GH stimulation testing to arrive at a correct diagnosis. However, diagnosing adult GHD can be challenging due to the episodic and pulsatile endogenous GH secretion, concurrently modified by age, gender, and body mass index. Hence, a GH stimulation test is often required to establish the diagnosis, and should only be considered if there is a clinical suspicion of GHD and the intention to treat if the diagnosis is confirmed. Currently, there is no ideal stimulation test and the decision to perform a GH stimulation test must factor in the validity of the chosen test, the appropriate GH cut-points, and the availability of local resources and expertise. For now, the insulin tolerance test remains the gold standard test, while the glucagon stimulation test and macimorelin test are reasonable alternatives to the insulin tolerance test, whereas the arginine test is no longer recommended because arginine is a poor GH secretagogue that requires a very low peak GH cut-point of 0.4 μg/L. In this chapter, we discuss published evidence of the GH stimulation tests used in the United States and the inherent caveats and limitations of each individual test. We propose utilizing the lower GH cut-point to 1mg/L for the glucagon stimulation test to improve its diagnostic accuracy in some overweight and all obese patients based on the clinical suspicion of having adult GHD, and summarize current knowledge and change of status of availability of the oral macimorelin test in the United States.

 

INTRODUCTION

 

Physiological growth hormone (GH) secretion from the anterior pituitary gland is episodic, pulsatile, and accounts for > 85% of total daily GH secretion (1). Due to its pulsatility, serum GH levels vary between peaks and troughs, with very low levels between pulses. Hypothalamic growth hormone–releasing hormone (GHRH) and somatostatin traverse the hypothalamic–pituitary portal system to stimulate and suppress GH production, respectively, by signaling through specific somatotroph cell-surface G protein-coupled receptors (2), while gastric-derived ghrelin also stimulates GH secretion and synergizes the action of GHRH (3). Additionally, other factors such as gender, nutritional status, sleep patterns, physical activity, and metabolic and hormonal signals from other endocrine glands, including glucocorticoids, thyroid hormones, and sex steroids, also play an important role in modulating day-to-day GH secretion (1). Growth hormone regulates its own secretion by a feedback mechanism that involves other peripheral mediators, such as insulin-like growth factor-I (IGF-I), free fatty acids, glucose, and insulin (4). Peripheral GH actions are primarily mediated through IGF-I synthesized mainly by the liver. Because IGF-I has a longer half-life in the circulation than GH, it is considered to provide an integrated measure of GH secretion. Like GH, serum IGF-I levels decline with aging (5), and tend to be low in obesity (6) and in patients with non-alcoholic fatty liver disease (7) that may overlap with the levels observed in younger GH–deficient patients. Hence, for these reasons, the diagnosis of adult GH deficiency (GHD) cannot be established in most patients by a random single measurement of serum GH or IGF-I level.

 

DIAGNOSIS OF ADULT GH DEFICIENCY: CURRENT PERSPECTIVE

 

Adult GHD is a rare heterogeneous disorder that commonly results from a variety of organic causes, including hypothalamic-pituitary tumors and/or their treatment, head trauma, and infiltrative diseases (8). This condition is characterized by decreased lean body mass and increased fat mass, dyslipidemia, cardiac dysfunction, decreased fibrinolysis and premature atherosclerosis, decreased muscle strength and exercise capacity, decreased bone mineral density, increased insulin resistance, and impaired quality of life (9). Treatment with GH replacement improves many, but not all, of these abnormalities (10, 11). However, due to the high cost of GH replacement (GH costs approximately $18,000 to $30,000 per year depending on the dose and brand used) (12) and concerns of potential long-term safety risks, particularly the development of diabetes mellitus, cancer and tumor recurrence, it is imperative that an accurate biochemical diagnosis is made so that appropriate GH replacement is offered to adults who are GH-deficient, and not for non-approved conditions (e.g., aging and sporting enhancement) (13, 14).

 

For the clinician, establishing the diagnosis of adult GHD is challenging because of the lack of a single biological end-point (e.g., growth failure in children with GHD). Other biochemical measurements like IGF-I, IGF-binding protein-3, or GH secretion over a 24-hour period have shown poor diagnostic value as there is an overlap between healthy and adults with GHD, particularly in adults > 40 years of age (5, 15). Hence, a GH stimulation test is often required to establish the diagnosis, and should only be considered if there is a clinical suspicion of GHD and the intention to treat if the diagnosis is confirmed. Currently, there is no ideal stimulation test as each test has its pros and cons, and the decision to consider performing a GH stimulation test to diagnose adult GHD must factor in the validity of the chosen test and its GH cut-points, and the availability of local resources and expertise.

 

Clinical practice guidelines recommend the evaluation of adult GHD to be based on medical history, clinical findings, and utilizing the appropriate GH stimulation test for biochemical confirmation (8, 16-18). The exception of when GH stimulation testing can be exempted include those with organic hypothalamic-pituitary disease with ≥ 3 pituitary hormone deficiencies and low serum IGF-I levels [< -2.0 standard deviation scores (SDS)] (19), patients with genetic defects affecting the hypothalamic-pituitary axes, and those with hypothalamic-pituitary structural brain defects (8, 16, 18). Evaluation for adult GHD should not be performed in patients with no evidence of a suggestive history, e.g., sellar/parasellar mass lesion or a history of a hypothalamic–pituitary insult, such as surgery, radiation therapy, head trauma, or brain tumor. Conversely, GH stimulation testing should not be performed in patients with commonly encountered, generalized, nonspecific symptoms of weakness, frailty, fatigue, or weight gain, without a history of organic hypothalamic/pituitary disease, as such patients are unlikely to benefit from GH therapy (8, 16, 18). These considerations are important for the clinician when deciding which patients to consider testing for possible adult GHD. 

 

All GH stimulation tests are based on the concept that a GH secretagogue agent acutely stimulates pituitary GH secretion, and peak serum GH levels are detected by sequential blood sampling of serum GH levels after administration of the agent. The desired criteria of an ideal GH stimulation test should include the following: the ability to accurately and reliably differentiate adults with GHD from GH-sufficient individuals, high reproducibility, safety with minimal side-effects, affordability, and short test duration. It should also not be unpleasant to the patient and it should be simple to perform.

 

The insulin tolerance test (ITT) has historically been accepted as the gold-standard test for the assessment of adult GHD provided adequate hypoglycemia (blood glucose <40 mg/dL) is achieved (8, 16, 17). However, multiple drawbacks associated with the ITT hamper its wider use (20), and they include the requirement of close medical supervision by a physician throughout the test, the possibility of inducing severe life-threatening hypoglycemia, and the potential of causing seizures and altered consciousness resulting from neuroglycopenia in certain susceptible sub-populations. This test is also contraindicated in the elderly (> 65 years of age) and in patients who are at risk of and/or with a history of cardio-/cerebrovascular disease and seizures.

 

Finding a reliable alternative to the ITT for the diagnosis of adult GHD has been challenging. When the GHRH-arginine test was available in the United States before EMD Serono discontinued manufacturing the GHRH analog (Geref@) in November 2008 (8, 16, 17),  GHRH-arginine test became the most acceptable alternative to the ITT. Since then, the glucagon stimulation test (GST) has grown in popularity replacing the GHRH-arginine test as the test of choice if the ITT cannot be performed or is contraindicated (21). Previous studies have examined the diagnostic utility of the GST for adult GHD, but these studies have either not taken body mass index (BMI) into consideration (22, 23) or included only controls with normal BMIs (24, 25). Several recent retrospective studies have questioned the diagnostic accuracy of the GST when the GH cut-point of 3mg/L is applied to overweight/obese adults (26-29) and in those with glucose intolerance (28, 29), while Hamrahian et al. (30) demonstrated in a prospective study of 28 patients by comparing the GST to the ITT that a lower GH cut-point of 1 mg/L improved its diagnostic accuracy with a 92% sensitivity and 100% specificity.

 

In this document, we will discuss published evidence of the GH stimulation tests used in the United States and the inherent caveats and limitations of each individual test. The lower GH cut-point of 1 mg/L for the GST should be utilized to improve its diagnostic accuracy in some overweight and all obese patients. We will also summarize current knowledge of the oral macimorelin test as the only approved diagnostic test for adult GHD by the United States Food and Drug Administration (FDA) and the European Medicines Agency, and its change in status of availability in the United States.

 

GENERAL LIMITATIONS AND IMPORTANT CAVEATS WHEN INTERPRETING GH STIMULATION TESTS

 

The responses to all GH stimulation tests show intra-individual variability, and the GH cut-points vary depending on the test used. For the ITT and GST, the cut-points advocated by previous consensus guidelines were 3-5 μg/L and 2.5-3 μg/L, respectively (8, 16). Other GH stimulatory agents such as clonidine, L-DOPA, and arginine are weaker GH secretagogues, and would require very low GH cut-points with utilization of sensitive GH assays to achieve adequate specificity (e.g., arginine of 0.4 μg/L) (31). Hence, these tests are not recommended in the United States (8, 16). Other limitations include the relative lack of validated normative data based on age, gender, BMI, glycemic status, and the paucity of data for specific etiologies of adult GHD that have recently been described, such as traumatic brain injury, subarachnoid hemorrhage, ischemic stroke, and central nervous system infections (32, 33).

 

One of the caveats in interpreting the results of GH stimulation tests is that adult GHD itself is complicated by an increased susceptibility to central obesity (34). Obesity per se is a state of relative GHD (35-40), and earlier physiologic studies in obese individuals have shown that spontaneous GH secretion is reduced, GH clearance is enhanced, and stimulated GH secretion is reduced (40-42). Conversely, serum IGF-I levels are unaffected, or even increased, and this discordance is related to the increased hepatic GH responsiveness (43). The decreased serum GH levels in obesity up-regulate GH receptor and sensitivity. Furthermore, non-alcoholic fatty liver disease and non-alcoholic steatohepatitis are now recognized as being highly prevalent in overweight and obese adults with GHD (44), with consequent lower serum IGF-I levels being associated with increased severity of the disease (7). Thus, these data suggest that BMI-specific cut-points should be considered when testing patients for adult GHD. Table 1summarizes the accepted GH cut-points for the GH stimulation tests used in the United States, as recommended by different consensus guidelines.

 

Table 1. Accepted GH Cut-Points (µg/L) for GH Stimulation Tests Used in the United States by Different Consensus Guidelines for Diagnosis of Adult GHD

 

GRS 2007

(17)

 

AACE 2009

(16)

ES 2011

(8)

AACE 2019

(18)

ITT

 

< 3.0

≤ 5.0

< 3.0 to 5.0

≤ 5.0

GHRH-arginine

- BMI < 25 kg/m2

- BMI 25-30 kg/m2

- BMI ≥ 30 kg/m2

 

 

< 11.0

< 8.0

< 4.0

 

≤ 11.0

≤ 8.0

≤ 4.0

 

< 11.0

< 8.0

< 4.0

No recommendation as not commercially available in the United States

Glucagon

- BMI < 25 kg/m2

- BMI 25-30 kg/m2

- BMI ≥ 30 kg/m2

 

 

< 3.0

< 3.0

< 3.0

 

≤ 3.0

≤ 3.0

≤ 3.0

 

< 3.0

< 3.0

< 3.0

 

≤ 3.0

≤ 3.01 or ≤ 1.02

≤ 1.0

Macimorelin

 

Not commercially available in 2007

 

Not commercially available in 2009

 

Not commercially available in 2011

≤ 2.8

Arginine

 

Not recommended to be used

≤ 0.4

 

Not recommended to be used

No longer recommended to be used

 

1GH cut-point of ≤ 3.0 µg/L for patients with a high pre-test probability; 2GH cut-point of ≤ 1.0 µg/L for patients with a low pre-test probability.

AACE, American Association of Clinical Endocrinologists; BMI, body mass index; ES, Endocrine Society; GHRH, growth hormone releasing hormone; GRS, Growth Hormone Research Society; ITT, insulin tolerance test.

 

GROWTH HORMONE STIMULATION TESTS USED IN DIAGNOSING ADULT GH DEFICIENCY

 

Insulin Tolerance Test

 

The ITT remains accepted as the gold standard test for the assessment of adult GHD, with a GH cut-point of 3-5 mg/L when adequate hypoglycemia (blood glucose < 40 mg/dL) is achieved (8, 16, 17). This GH cut-point was originally proposed by Hoffman et al. (45) in 1994 based on GH responses to insulin-induced hypoglycemia, mean 24-hour GH levels derived from 20-min sampling, and serum IGF-I and IGFBP-3 levels in 23 patients considered GH-deficient due to organic pituitary disease, and in 35 sex-matched normal subjects of similar age and BMI. The ranges of stimulated peak GH responses separated GH-deficient (0.2-3.1 mg/L) from GH-sufficient (5.3-42.5 mg/L) patients. However, an overlap in mean 24-hour GH, IGF-I, and IGFBP-3 levels was observed, demonstrating the challenge in utilizing random single serum GH, IGF-I and IGFBP-3 levels to accurately differentiate GH-sufficiency from GHD.

 

Disadvantages of the ITT include the requirement of close medical supervision, may be unpleasant, and cautioned in some patients because of potential adverse effects (e.g., seizures or loss of consciousness resulting from neuroglycopenia), and contraindicated in elderly patients and in patients at risk of and/or with a history of cardio-/cerebrovascular disease and seizures. Furthermore, normoglycemic and/or hyperglycemic obese patients with insulin resistance may fail to achieve adequate hypoglycemia (46), necessitating the use of higher insulin doses (0.15-0.2 IU/kg), thus increasing the risk of delayed hypoglycemia. Although the ITT demonstrates good sensitivity, its reproducibility is another major limitation. Differences in peak GH responses have been demonstrated in healthy subjects undergoing ITT at varying times (47) and in women at different times of their menstrual cycle (48).

 

Table 2. Recommended Protocol for Performing the ITT

CONTRAINDICATIONS:

History of epileptic seizures, coronary artery disease, pregnancy, or age > 55 years.

PRECAUTIONS:

Patients commonly develop neuroglycopenic symptoms during the test and should be encouraged to report these symptoms (administration of IV anti-emetics can be considered).

Late hypoglycemia may occur (patients should be advised to eat small and frequent meals after completion of the test).

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water (if the HPA axis is simultaneously assessed, then glucocorticoids should be withheld ≥ 12 hours before testing).

Weigh patient.

1Place IV cannula for IV access in both forearms.

2Administer IV human Regular insulin (standard dose: 0.05-0.1 units/kg for non-diabetic subjects with a BMI < 30 kg/m2 and high dose: 0.15-0.3 units/kg for subjects with a BMI ≥ 30 kg/m2).

SAMPLING AND MEASUREMENTS:

Baseline

Blood is drawn for glucose measurement with a glucometer.

Blood draw for baseline glucose, GH and IGF-I (cortisol and ACTH, if HPA axis is assessed simultaneously) levels will be sent to the laboratory for further analysis.

During the test

Blood samples are drawn from the IV line every 5-10 mins for measurement of glucose levels using a glucometer.

Signs and symptoms of neuroglycopenia are recorded.

When blood glucose levels from the glucometer approaches 45 mg/dL (2.5 mmol/L), blood samples are sent to the laboratory for measurements of blood glucose levels.

When symptomatic hypoglycemia is achieved (laboratory blood glucose < 40 mg/dL or 2.2 mmol/L), additional blood samples are collected to measure glucose and GH (+/- cortisol if the HPA axis is assessed simultaneously) levels at 20, 25, 30, 35, 40, 60 and 90 min.

The patient can begin drinking orange juice and eat to raise his/her blood glucose levels (IV 100 ml of 5% Dextrose can be administered if the patient cannot tolerate oral intake due to nausea or vomiting).

At the end of the test

Blood glucose levels measured from the glucometer should increase to levels > 70 mg/dL (3.9 mmol/L) before the patient is discharged from the testing unit.

INTERPRETATION:

If adequate (symptomatic) hypoglycemia is not achieved (< 40 mg/dL or 2.2 mmol/L), then adult GHD cannot be diagnosed.

Peak serum GH levels ≤ 5 µg/L at any time point during the hypoglycemic phase of the test is diagnostic of adult GHD.

CAUTION:

If adequate (symptomatic) hypoglycemia is not achieved (< 40 mg/dL or 2.2 mmol/L), then adult GHD cannot be diagnosed.

ACTH: adrenocorticotropic hormone, HPA: hypothalamic-pituitary-adrenal, IV: intravenous.

1Two IV lines are placed, one IV line is used for the administration of insulin bolus and possibly for administration of IV 5% Dextrose administration if the patient requires resuscitation from hypoglycemia, while the other IV line is used for repeated blood draws.

2In certain patients with BMIs > 30 kg/m2 who appear muscular with increased insulin sensitivity, clinical discretion is required in deciding the insulin dose for these patients. A dose of 0.05-0.1 units/kg may be more appropriate to prevent severe or delayed hypoglycemia.   

 

Glucagon Stimulation Test

 

Glucagon is reportedly to be more potent than arginine or clonidine in stimulating GH secretion (24, 25). Glucagon is also a more potent GH secretagogue when administered intramuscularly or subcutaneously compared to the intravenous route (49). However, the mechanism/s of glucagon-induced GH stimulation remains unclear, and one hypothesis is that glucagon decreases ghrelin-independent effects of glucose or insulin variations (50).

 

There have been three earlier studies that have assessed the GST in identifying adult GHD in patients with pituitary disorders (22, 23, 51). Gomez et al. (51) and Conceicao et al. (23) compared the diagnostic characteristics of GST to ITT and included a control group matched for age and sex in both studies, and for BMI in one study (51). Using receiver operating characteristic (ROC) analysis, both studies proposed that a GH cut-point of 3 mg/L provided optimal sensitivity and specificity (51, 52). Gomez et al. (51) also demonstrated an inverse correlation between age (R = - 0.389, P = 0.0075) and BMI (R = - 0.329, P = 0.025) with peak GH levels in healthy controls. These data suggest that there is a potential association between relative, but not organic, GHD in aging and obesity. However, this study was conducted in a European cohort, where the frequency and severity of obesity is generally to a lesser degree than in the United States (53). Conversely, Conceicao et al. (23) demonstrated that peak GH levels were unaffected by age in either the control or patient group, and neither were there any gender differences. Additionally, Gomez et al. (51)used intramuscular glucagon doses of 1 mg and 1.5 mg for body weights ≤ 90 kg and > 90 kg respectively, whereas Conceicao et al. (23) used intramuscular glucagon of 1 mg for all subjects. In another study, Berg et al. (22)demonstrated an optimal peak GH cut-point of 2.5 mg/L with 95% sensitivity and 79% specificity using ROC analysis. This study also reported lower peak GH levels with GST compared to ITT (5.1 vs 6.7 mg/L, P < 0.01) and a positive correlation between peak GH levels during ITT and GST (R = 0.88, P < 0.0001), but no correlation between BMI or age to peak GH responses (54, 55). However, these (22, 23, 51) and other earlier studies (24, 25, 49, 56) did not specifically evaluate patients with glucose intolerance; hence, the diagnostic accuracy of the GST in testing for GHD in this population remains unclear.

 

Advantages of the GST is its reproducibility, safety, and lack of influence by gender and hypothalamic GHD (21), whereas disadvantages include the lengthy test duration (3-4 hours), and the need for an intramuscular injection that might not appeal to some patients. Side-effects frequently reported include nausea, vomiting, and headaches ranging from < 10% (22) to 34% (54), mainly occur between 60-210 min and tend to resolve by 240 min into the test, and seem to be more pronounced in elderly subjects, where severe symptomatic hypotension, hypoglycemia, and seizures have been observed (57).    

 

However, since the publication of the 2009 American Association of Clinical Endocrinologists (AACE) (16) and 2011 Endocrine Society (8) Clinical Practice Guidelines, there have been several studies that have suggested that the fixed-dose GST using a GH cut-point of 3 mg/L may potentially over-diagnose adult GHD in a substantial number of overweight/obese subjects and in those with glucose intolerance. In two large retrospective studies, Toogood et al.(58) and Yuen et al. (29) found an inverse correlation between BMI and peak GH during the GST, and that this relationship appeared to be strongest with BMIs between 30 and 40 kg/m2 and seemed to plateau for those with BMIs > 40 kg/m2 (58). Alternatively, a negative correlation between BMI and peak GH following glucagon stimulation has been reported by Gomez et al. (51) in healthy subjects but not in patients with underlying pituitary disease. Dichtel et al. (26) evaluated 3 groups of overweight/obese men, i.e., controls who were younger than the patients, patients with 3-4 pituitary hormone deficits, and patients with 1-2 pituitary hormone deficits. Using ROC analysis, the GH cut-point of 0.94 mg/L provided the optimal sensitivity (90%) and specificity (94%), whereas BMI and amount of visceral adipose tissue inversely correlated with peak GH levels in controls. Almost half of the healthy overweight/obese individuals (45%) failed the GST using the 3 mg/L GH cut-point. Diri et al. (27) evaluated 216 patients with pituitary disease and 26 healthy controls and compared the GST to the ITT. These investigators used a GH cut-point of 3.0 mg/L for the ITT and two GH cut-points of 3.0 mg/L and 1.07 mg/L for the GST, yielding the diagnosis of adult GHD in 86.1%, 74.5%, and 54.2 % patients, respectively. Additionally, patient age, BMI, and number of pituitary hormone deficits correlated with IGF-I and peak GH levels. Twelve out of 26 (46.2 %) healthy subjects failed the GST using a GH cut-point of 3.0 mg/L, but none when the cut-point was lowered to 1.07 mg/L. Wilson et al. (28) studied 42 patients with a high pre-test probability of adult GHD. After excluding 10 patients with severe GHD based on peak GH levels ≤ 0.1 mg/L, these investigators found that body weight negatively correlated with GH area under the curve (AUC) (R = -0.45; P = 0.01) and peak GH response (R = -0.42; P = 0.02) and positively correlated with nadir blood glucose levels (R = 0.48; P < 0.01). Conversely, nadir blood glucose levels during GSTs inversely correlated with GH AUC (r= -0.38; p=0.03) and peak GH (r= -0.37; p=0.04), implying that patients with higher nadir blood glucose levels tended to have a lesser glucagon-induced GH response. Recently, Hamrahian et al. (30) compared the fixed-dose GST (1 mg or 1.5 mg in patients > 90 kg body weight) and weight-based GST (WB-GST: 0.03 mg/kg) with the ITT using a GH cut-point of 3.0 mg/L. Patients with hypothalamic-pituitary disease and 1-2 (n = 14) or ≥ 3 (n = 14) pituitary hormone deficiencies, and control subjects (n = 14) matched for age, sex, estrogen status and BMI undertook the ITT, GST and WB-GST in random order. Using ROC analyses, the optimal GH cut-point was 1.0 (92% sensitivity, 100% specificity) for fixed-dose GST and 2.0 mg/L (96% sensitivity and 100% specificity) for WB-GST. Therefore, lowering the GH cut-point from 3 mg/L to 1 mg/L is important to reduce misclassifying adult GHD in overweight (BMI 25-30 kg/m2) patients with a low pre-test probability and in obese (BMI > 30 kg/m2) patients.

 

It remains unclear whether hyperglycemia influences peak GH responses to glucagon stimulation, independent of central adiposity. No peak GH responses have been studied using the GST in normal controls > 70 years of age, and none of the previous studies included patients with poorly controlled diabetes mellitus. Studies by Yuen et al. (29) and Wilson et al. (28) demonstrated that higher fasting (range 90-316 mg/dL), peak (range 156-336 mg/dL), and nadir (range 52-200 mg/dL) blood glucose levels during the GST were associated with lower peak GH responses. Therefore, stratification of GH responsiveness by the degree of glycemia will be helpful to clinicians in interpreting the GST results in patients with impaired glucose tolerance and diabetes mellitus. Because these data are currently unavailable, caution should be exercised when interpreting abnormal GST results in these patients. Further larger prospective studies are needed to address the effects of varying degrees of hyperglycemia on the ability of glucagon to stimulate GH secretion.

 

Table 3.  Recommended Protocol for Performing the Glucagon Stimulation Test

CONTRAINDICATIONS:

Malnourished patients or patients who have not eaten for > 48 hours.

Severe fasting hyperglycemia > 180 mg/dL.

PRECAUTIONS:

Patients may feel nauseous during the test (administration of IV anti-emetics may be considered).

Late hypoglycemia may occur (patients should be advised to eat small and frequent meals after completion of the test).

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water.

Weigh patient.

Place IV cannula for IV access in one forearm.

Administer IM glucagon (1.0 mg if patient body weight ≤ 90 kg and 1.5 mg if patient body weight > 90 kg).

SAMPLING AND MEASUREMENTS:

Blood is drawn for measurements of serum GH1 and blood glucose2 levels at 0, 30, 60, 90, 120, 150, 180, 210 and 240 mins.

INTERPRETATION:

Peak GH levels ≤ 3.0 µg/L in normal-weight (BMI < 25 kg/m2) patients and in

overweight (BMI 25-30 kg/m2) patients with a high pre-test probability, and ≤ 1.0 ug/L in

overweight (BMI 25-30 kg/m2) patients with a low pre-test probability and in obese (BMI >

30 kg/m2) patients at any time point during testing are diagnostic of adult GHD.

CAUTION:

Clinical suspicion of pre-test probability should be taken into consideration when interpreting GST results in patients > 70 years of age and in patients with impaired glucose tolerance and poorly controlled diabetes mellitus, as no peak GH responses have been studied in these patients.

IM: intramuscular, IV: intravenous.

1Serum GH: peak GH levels tend to occur between 120-180 mins; 2blood glucose: usually peaks around 90 mins and then gradually declines (not a requirement to interpret the test).

 

Macimorelin Test

 

Growth hormone secretagogues (GHSs) are peptidyl (GH-releasing peptide [GHRP]) and nonpeptidyl molecules that exert strong dose-dependent and specific stimulatory effects on the animal and human somatotrope secretion (59). These agents act as functional somatostatin antagonists by binding to their specific GH secretagogue receptor-1a in the hypothalamus and pituitary. The natural ligand for this receptor is the gut peptide ghrelin (60). Growth hormone secretagogues are now considered as ghrelin mimetic agents and can be administered parenterally (e.g., GHRP-2, GHRP-6, hexarelin) or orally (e.g., MK-677 and macimorelin).

 

Macimorelin (formerly known as AEZS-130, ARD-07, and EP-01572) is a novel GH secretagogue that binds the GHS-R1a receptor and to pituitary and hypothalamic extracts with a similar affinity to ghrelin (61). In healthy volunteers, it is readily absorbed with good stability and oral bioavailability, and effectively stimulates endogenous GH secretion (61). An open-label, crossover, multicenter trial examined the diagnostic accuracy of a single oral dose of macimorelin (0.5 mg/kg) compared to GHRH plus arginine in adults with GHD and healthy matched controls (62). Peak GH levels were 2.36 ± 5.69 and 17.71 ± 19.11 mg/L in adults with GHD and healthy controls, respectively, with optimal GH cut-points ranging between 2.7 and 5.2 mg/L (62). Macimorelin showed good discrimination comparable to GHRH plus arginine, with peak GH levels that were inversely associated with BMI in controls. In a recent multicenter, open-label, randomized, two-way crossover study, oral macimorelin was compared to the ITT to validate its use for the diagnosis of adult GHD (63). The GH cut-point levels of 2.8 mg/L for macimorelin and 5.1 mg/L for ITT provided 95.4% (95% CI, 87% to 99%) negative agreement, 74.3% (95% CI, 63% to 84%) positive agreement, 87% sensitivity, and 96% specificity. In both studies (62, 63), macimorelin was well-tolerated, reproducible, and safe. In December 2017, the United States FDA approved macimorelin for use as a diagnostic test for adult GHD and mandated the GH cut-point of2.8 mg/L to be used to differentiate patients with normal GH secretion from those with GHD. However, in the study by Garcia et al. (63), when the GH cut-point was increased to 5.1 mg/L for both macimorelin and ITT, negative agreement and specificity was unchanged at 94% (95% CI, 85% to 98%) and 96%, respectively, but interestingly, positive agreement and sensitivity was higher at 82% (95% CI, 72% to 90%) and 92%. Because measured serum GH levels are dependent on the GH assays used, using the GH cut-point of 5.1 mg/L for macrimorelin that is identical to the cut-point accepted for the ITT could be considered in patients with peak serum GH levels between 2.8 mg/L to 5.1 mg/L, especially if the patient has a high pre-test probability, e.g., history of surgery on a sellar/parasellar mass with 1-2 other pituitary hormone deficiencies. It is important to note that this test is not affected by age, BMI, or sex indicating its robustness for diagnosing adult GHD (64).

 

Main advantages of macimorelin are that the drug is orally administered, unlike the ITT, GHRH plus arginine or GST, that requires intravenous or intramuscular administration, and no risk of causing hypoglycemia. In addition, the test only lasts 90 minutes with 3-4 blood sample collections required, in contrast to more blood sample collections over 2 hours for the ITT and 3-4 hours for the GST. The most commonly reported side effect was mild dysgeusia, which did not require any intervention and resolved spontaneously (63). One drug-related serious adverse event was reported; that was in a subject with an asymptomatic QT interval prolongation on the electrocardiogram that resolved spontaneously within 24 h (62). Thus, careful assessment of the patient’s concurrent medications is recommended as well as discontinuation of strong CYP3A4 inducers, provided this is considered safe by the prescribing physician and with sufficient washout time prior to testing.

 

However, in August 2022, a press announcement stated that Novo Nordisk Healthcare AG provided a 270-day notice period to terminate the amended development and commercialization license agreement for macimorelin (MacrilenÒ) in the United States (65). This means that as of May 23, 2023, Aerterna Zentaris regained its full rights in the United States and Canada to macimorelin but because it has yet to find a partner in the United States to market macimorelin, it was further announced that sales of macimorelin will be temporarily discontinued and use of the agent beyond May 2023 will continue until its supplies in the United States runs out (66).

 

Table 4.  Recommended Protocol for Performing the Macimorelin Test

CONTRAINDICATIONS:

Drugs that may increase its plasma levels and prolong QT.

PRECAUTIONS:

Dysgeusia.

PROCEDURE:

Fast from midnight for 8-10 hours.

All morning medications can be taken with water.

Weigh patient.

Place IV cannula for IV access in one forearm.

Dissolve in water 1 (120 ml) or 2 pouches (240 ml) of macimorelin (≤ 120 kg = 1 pouch; > 120 kg = 2 pouches)

Calculate macimorelin dose (0.5 mg/kg as a single oral dose) and volume of water required to reconstitute macimorelin solution (patient body weight X kg = X ml macimorelin solution, e.g., patient with a body weight of 70 kg would require 70 mL of reconstituted macimorelin solution)

After volume of macimorelin is calculated, stir the solution gently and thoroughly for 2-3 min, and use within 30 min of preparation.

Draw the exact macimorelin volume of solution into a needleless syringe, transfer the exact volume of into a drinking glass, and instruct the patient to drink the entire volume of solution within 30 seconds.

SAMPLING AND MEASUREMENTS:

Blood is drawn for measurements of serum GH levels at 30, 45, 60 and 90 min.

INTERPRETATION:

Peak serum GH levels tend to occur between 45-60 mins.

When used according to prescribing package label, peak GH levels ≤ 2.8 µg/L at any time point is diagnostic of adult GHD.

CAUTION:

Peak GH levels ≤ 5.1 µg/L at any time point may be considered in patients with a high-pre-test probability to diagnose adult GHD, as this higher GH cut-point limits the risk of a false-positive diagnosis and maintains a high detection rate for GH-deficient patients because of the more potent GH stimulatory effect of macimorelin compared with the ITT.

Safety and diagnostic performance in patients < 18 and > 65 years of age, and in patients with impaired glucose tolerance and poorly controlled diabetes mellitus, and BMI-adjusted peak GH cut-points for overweight and obese patients is not established.

 

Summary of Tests

 

Table 5 displays a summary of the desirable test characteristics of GH stimulation tests currently available in the United States.

 

Table 5. Summary of Desirable Test Characteristics of each GH Stimulation Test Currently Available in the United States

Test

Accurate?

Safe?

Tolerability?

Simple?

Quick?

Available?

Cost

ITT

Gold standard

No2

No4

No

No

Yes

$

GST

Yes1

Yes3

No3

Yes

No

Yes

$

Macimorelin

Yes

Yes

Yes

Yes

Yes

Yes/No

$$$

1if appropriate BMI-specific GH cut-points are used; 2contraindicated in patients with a history hypoglycemia, history of previous seizures, in the elderly (> 65 years of age), and in patients at risk of and/or with a history of cardio-/cerebrovascular disease; 3caution in patients with propensity for nausea and vomiting, and elderly patients who may be at risk of developing symptomatic hypotension and dizziness (57); 4patients may not tolerate severe symptomatic hypoglycemia. GST, glucagon stimulation test; ITT, insulin tolerance test.

 

STANDARDIZATION OF GH ASSAYS

 

Accurate measurement of GH levels is critical for establishing the diagnosis of adult GHD because the analytical method influences the results of GH stimulation tests, which is dependent on specific GH cut-point levels. However, circulating GH is present in several different isoforms and isomers, including the most common variant of 22 kDa, and other smaller molecules, such as the 20 kDa GH variant. Monoclonal antibodies binding to a specific molecular form of GH are used to limit detection to the 22 kDa GH, but will not detect other GH isoforms. Other molecules similar to GH (e.g., placental GH and prolactin) could potentially cross-react and affect the measurement of GH levels. Growth hormone binding protein, to which approximately 50% of circulating GH is bound, can also cause interference in a GH assay. Furthermore, substantial heterogeneity exists among currently utilized assays due to the use of different standard preparations for calibration of GH immunoassays, and lack of harmonization between various GH assays makes it difficult to directly compare diagnostic cut-points across different published studies. Another source of confusion when interpreting data of GH stimulation tests was that some laboratories reported GH levels in activity (mU/L), whereas others used mass units (mg/L) (67).

 

Due to the heterogeneity of GH assays, it is important that GH assays utilize a universal GH calibration standard 98/574 (National Institute for Biological Standards and Control), a recombinant pituitary GH preparation of high purity (68). All assay manufacturers should also specify the validation of their assay, which should include specification of the GH isoforms detected (20 kDa GH, 22 kDa GH, and other isoforms), the analyte being measured, the specificities of the antibodies used, and the presence or absence of growth hormone binding protein interference.

 

CONCLUSIONS

 

The decision to perform GH stimulation tests should be based on the clinical suspicion of the treating endocrinologist. If the clinical suspicion is high, such as in a patient with history of surgery on a sellar mass, concurrent 1-2 other pituitary hormone deficiencies, and a low (< -2 SDS) or low-normal (< 0 SDS) serum IGF-I level, then performing GH stimulation testing is recommended. If the clinical suspicion is low, such as in cases where there is no suggestive history, such as hypothalamic-pituitary disease, surgery or radiation therapy, head trauma, or childhood-onset GHD, then the diagnosis of adult GHD should not be pursued and GH stimulation testing should not be performed. For now, the ITT remains the gold standard GH stimulation test, and the GST and macimorelin test (where available) are reasonable alternatives to the ITT. As the reliability of the GST GH cut-point of 3 mg/L in overweight/obese subjects and in those with glucose intolerance can misclassify some patients, the utilization of GH cut-points of the GST is now based on the clinician’s level of suspicion of the patient’s pre-test probability and underlying BMI. Macimorelin, a drug administered orally that was approved by the United States FDA in December 2017 is an attractive test because it is easy to conduct with high reproducibility, safe, and has comparable diagnostic accuracy to the ITT and GHRH plus arginine test. The factors that limit its wider is its high cost (one 60 mg macimorelin packet costs approximately $4,500) (69) and the potential of drug-to-drug interactions that may cause QT prolongation. Following the announcement in August 2022 that macimorelin will be temporarily discontinued in the commercial market effective May 2023, after supplies of macimorelin runs out in the United States, the ITT and GST will only be the two GH stimulation tests available to clinicians, limiting the choices of tests that can be used.

 

REFERENCES

 

  1. Gunawardane K, Krarup Hansen T, Sandahl Christiansen J, Lunde Jorgensen JO. Normal physiology of growth hormone in adults. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, et al., editors. Endotext. South Dartmouth (MA) 2000.
  2. Hartman ML, Veldhuis JD, Thorner MO. Normal control of growth hormone secretion. Horm Res. 1993;40(1-3):37-47.
  3. Casanueva FF, Camina JP, Carreira MC, Pazos Y, Varga JL, Schally AV. Growth hormone-releasing hormone as an agonist of the ghrelin receptor GHS-R1a. Proc Natl Acad Sci U S A. 2008;105(51):20452-7.
  4. Muller EE, Locatelli V, Cocchi D. Neuroendocrine control of growth hormone secretion. Physiol Rev. 1999;79(2):511-607.
  5. Hilding A, Hall K, Wivall-Helleryd IL, Saaf M, Melin AL, Thoren M. Serum levels of insulin-like growth factor I in 152 patients with growth hormone deficiency, aged 19-82 years, in relation to those in healthy subjects. J Clin Endocrinol Metab. 1999;84(6):2013-9.
  6. Galli G, Pinchera A, Piaggi P, Fierabracci P, Giannetti M, Querci G, et al. Serum insulin-like growth factor-1 concentrations are reduced in severely obese women and raise after weight loss induced by laparoscopic adjustable gastric banding. Obes Surg. 2012;22(8):1276-80.
  7. Sumida Y, Yonei Y, Tanaka S, Mori K, Kanemasa K, Imai S, et al. Lower levels of insulin-like growth factor-1 standard deviation score are associated with histological severity of non-alcoholic fatty liver disease. Hepatol Res. 2015;45(7):771-81.
  8. Molitch ME, Clemmons DR, Malozowski S, Merriam GR, Vance ML. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96(6):1587-609.
  9. Simpson H, Savine R, Sonksen P, Bengtsson BA, Carlsson L, Christiansen JS, et al. Growth hormone replacement therapy for adults: into the new millennium. Growth Horm IGF Res. 2002;12(1):1-33.
  10. Jorgensen AP, Fougner KJ, Ueland T, Gudmundsen O, Burman P, Schreiner T, et al. Favorable long-term effects of growth hormone replacement therapy on quality of life, bone metabolism, body composition and lipid levels in patients with adult-onset growth hormone deficiency. Growth Horm IGF Res. 2011;21(2):69-75.
  11. Widdowson WM, Gibney J. The effect of growth hormone replacement on exercise capacity in patients with GH deficiency: a meta-analysis. J Clin Endocrinol Metab. 2008;93(11):4413-7.
  12. Cook D, Owens G, Jacobs M. Human growth hormone treatment in adults: balancing economics and ethics. Am J Manag Care. 2004;10(13 Suppl):S417-9.
  13. Bartke A. Growth hormone and aging: updated review. World J Mens Health. 2019;37(1):19-30.
  14. Siebert DM, Rao AL. The use and abuse of human growth hormone in sports. Sports Health. 2018:1941738118782688.
  15. Yuen KC, Cook DM, Sahasranam P, Patel P, Ghods DE, Shahinian HK, et al. Prevalence of GH and other anterior pituitary hormone deficiencies in adults with nonsecreting pituitary microadenomas and normal serum IGF-1 levels. Clin Endocrinol (Oxf). 2008;69(2):292-8.
  16. Cook DM, Yuen KC, Biller BM, Kemp SF, Vance ML, American Association of Clinical E. American Association of Clinical Endocrinologists medical guidelines for clinical practice for growth hormone use in growth hormone-deficient adults and transition patients - 2009 update. Endocr Pract. 2009;15 Suppl 2:1-29.
  17. Ho KKY on behalf of the 2007 GH Deficiency Consensus Workshop Participants. Consensus guidelines for the diagnosis and treatment of adults with GH deficiency II: a statement of the GH Research Society in association with the European Society for Pediatric Endocrinology, Lawson Wilkins Society, European Society of Endocrinology, Japan Endocrine Society, and Endocrine Society of Australia. Eur J Endocrinol. 2007;157(6):695-700.
  18. Yuen KCJ (chair of the 2019 AACE Growth Hormone Task Force), Biller BMK, Radovick S, Carmichael JD, Jasim S, Pantalone KM, et al. American Association of Clinical Endocrinologists and American College of Endocrinology Guidelines for management of growth hormone deficiency in adults and patients transitioning from pediatric to adult care. Endocr Pract. 2019 Nov;25(11):1191-1232.
  19. Hartman ML, Crowe BJ, Biller BM, Ho KK, Clemmons DR, Chipman JJ. Which patients do not require a GH stimulation test for the diagnosis of adult GH deficiency? J Clin Endocrinol Metab. 2002;87(2):477-85.
  20. Gordon MB, Levy RA, Gut R, Germak J. Trends in growth hormone stimulation testing and growth hormone dosing in adult growth hormone deficiency patients: results from the ANSWER Program. Endocr Pract. 2016;22(4):396-405.
  21. Yuen KC, Biller BM, Molitch ME, Cook DM. Clinical review: Is lack of recombinant growth hormone (GH)-releasing hormone in the United States a setback or time to consider glucagon testing for adult GH deficiency? J Clin Endocrinol Metab. 2009;94(8):2702-7.
  22. Berg C, Meinel T, Lahner H, Yuece A, Mann K, Petersenn S. Diagnostic utility of the glucagon stimulation test in comparison to the insulin tolerance test in patients following pituitary surgery. Eur J Endocrinol. 2010;162(3):477-82.
  23. Conceicao FL, da Costa e Silva A, Leal Costa AJ, Vaisman M. Glucagon stimulation test for the diagnosis of GH deficiency in adults. J Endocrinol Invest. 2003;26(11):1065-70.
  24. Aimaretti G, Baffoni C, DiVito L, Bellone S, Grottoli S, Maccario M, et al. Comparisons among old and new provocative tests of GH secretion in 178 normal adults. Eur J Endocrinol. 2000;142(4):347-52.
  25. Rahim A, Toogood AA, Shalet SM. The assessment of growth hormone status in normal young adult males using a variety of provocative agents. Clin Endocrinol (Oxf). 1996;45(5):557-62.
  26. Dichtel LE, Yuen KC, Bredella MA, Gerweck AV, Russell BM, Riccio AD, et al. Overweight/obese adults with pituitary disorders require lower peak growth hormone cutoff values on glucagon stimulation testing to avoid overdiagnosis of growth hormone deficiency. J Clin Endocrinol Metab. 2014;99(12):4712-9.
  27. Diri H, Karaca Z, Simsek Y, Tanriverdi F, Unluhizarci K, Selcuklu A, et al. Can a glucagon stimulation test characterized by lower GH cut-off value be used for the diagnosis of growth hormone deficiency in adults? Pituitary. 2015;18(6):884-92.
  28. Wilson JR, Utz AL, Devin JK. Effects of gender, body weight, and blood glucose dynamics on the growth hormone response to the glucagon stimulation test in patients with pituitary disease. Growth Horm IGF Res. 2016;26:24-31.
  29. Yuen KC, Biller BM, Katznelson L, Rhoads SA, Gurel MH, Chu O, et al. Clinical characteristics, timing of peak responses and safety aspects of two dosing regimens of the glucagon stimulation test in evaluating growth hormone and cortisol secretion in adults. Pituitary. 2013;16(2):220-30.
  30. Hamrahian AH, Yuen KC, Gordon MB, Pulaski-Liebert KJ, Bena J, Biller BM. Revised GH and cortisol cut-points for the glucagon stimulation test in the evaluation of GH and hypothalamic-pituitary-adrenal axes in adults: results from a prospective randomized multicenter study. Pituitary. 2016;19(3):332-41.
  31. Biller BM, Samuels MH, Zagar A, Cook DM, Arafah BM, Bonert V, et al. Sensitivity and specificity of six tests for the diagnosis of adult GH deficiency. J Clin Endocrinol Metab. 2002;87(5):2067-79.
  32. Pekic S, Popovic V. Diagnosis of endocrine disease: Expanding the cause of hypopituitarism. Eur J Endocrinol. 2017;176(6):R269-R82.
  33. Tanriverdi F, Kelestimur F. Classical and non-classical causes of GH deficiency in adults. Best Pract Res Clin Endocrinol Metab. 2017;31(1):3-11.
  34. Verhelst J, Abs R. Cardiovascular risk factors in hypopituitary GH-deficient adults. Eur J Endocrinol. 2009;161 Suppl 1:S41-9.
  35. Utz AL, Yamamoto A, Sluss P, Breu J, Miller KK. Androgens may mediate a relative preservation of IGF-I levels in overweight and obese women despite reduced growth hormone secretion. J Clin Endocrinol Metab. 2008;93(10):4033-40.
  36. Pijl H, Langendonk JG, Burggraaf J, Frolich M, Cohen AF, Veldhuis JD, et al. Altered neuroregulation of GH secretion in viscerally obese premenopausal women. J Clin Endocrinol Metab. 2001;86(11):5509-15.
  37. Makimura H, Stanley T, Mun D, You SM, Grinspoon S. The effects of central adiposity on growth hormone (GH) response to GH-releasing hormone-arginine stimulation testing in men. J Clin Endocrinol Metab. 2008;93(11):4254-60.
  38. Iranmanesh A, Lizarralde G, Veldhuis JD. Age and relative adiposity are specific negative determinants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half-life of endogenous GH in healthy men. J Clin Endocrinol Metab. 1991;73(5):1081-8.
  39. Beck P, Koumans JH, Winterling CA, Stein MF, Daughaday WH, Kipnis DM. Studies of insulin and growth hormone secretion in human obesity J Lab Clin Med. 1964;64:654-67.
  40. Langendonk JG, Meinders AE, Burggraaf J, Frolich M, Roelen CA, Schoemaker RC, et al. Influence of obesity and body fat distribution on growth hormone kinetics in humans. Am J Physiol. 1999;277(5):E824-9.
  41. Magiakou MA, Mastorakos G, Gomez MT, Rose SR, Chrousos GP. Suppressed spontaneous and stimulated growth hormone secretion in patients with Cushing's disease before and after surgical cure. J Clin Endocrinol Metab. 1994;78(1):131-7.
  42. Veldhuis JD, Iranmanesh A, Ho KK, Waters MJ, Johnson ML, Lizarralde G. Dual defects in pulsatile growth hormone secretion and clearance subserve the hyposomatotropism of obesity in man. J Clin Endocrinol Metab. 1991;72(1):51-9.
  43. Yuen KC, Cook DM, Rumbaugh EE, Cook MB, Dunger DB. Individual igf-I responsiveness to a fixed regimen of low-dose growth hormone replacement is increased with less variability in obese compared to non-obese adults with severe growth hormone deficiency. Horm Res. 2006;65(1):6-13.
  44. Nishizawa H, Iguchi G, Murawaki A, Fukuoka H, Hayashi Y, Kaji H, et al. Nonalcoholic fatty liver disease in adult hypopituitary patients with GH deficiency and the impact of GH replacement therapy. Eur J Endocrinol. 2012;167(1):67-74.
  45. Hoffman DM, O'Sullivan AJ, Baxter RC, Ho KK. Diagnosis of growth-hormone deficiency in adults. Lancet. 1994;343(8905):1064-8.
  46. Lee P, Greenfield JR, Ho KK. Factors determining inadequate hypoglycaemia during insulin tolerance testing (ITT) after pituitary surgery. Clin Endocrinol (Oxf). 2009;71(1):82-5.
  47. Pfeifer M, Kanc K, Verhovec R, Kocijancic A. Reproducibility of the insulin tolerance test (ITT) for assessment of growth hormone and cortisol secretion in normal and hypopituitary adult men. Clin Endocrinol (Oxf). 2001;54(1):17-22.
  48. Hoeck HC, Vestergaard P, Jakobsen PE, Laurberg P. Test of growth hormone secretion in adults: poor reproducibility of the insulin tolerance test. Eur J Endocrinol. 1995;133(3):305-12.
  49. Ghigo E, Bartolotta E, Imperiale E, Bellone J, Cardinale G, Aimaretti G, et al. Glucagon stimulates GH secretion after intramuscular but not intravenous administration. Evidence against the assumption that glucagon per se has a GH-releasing activity. J Endocrinol Invest. 1994;17(11):849-54.
  50. Arafat MA, Otto B, Rochlitz H, Tschop M, Bahr V, Mohlig M, et al. Glucagon inhibits ghrelin secretion in humans. Eur J Endocrinol. 2005;153(3):397-402.
  51. Gomez JM, Espadero RM, Escobar-Jimenez F, Hawkins F, Pico A, Herrera-Pombo JL, et al. Growth hormone release after glucagon as a reliable test of growth hormone assessment in adults. Clin Endocrinol (Oxf). 2002;56(3):329-34.
  52. Carmichael JD, Danoff A, Milani D, Roubenoff R, Lesser ML, Livote E, et al. GH peak response to GHRH-arginine: relationship to insulin resistance and other cardiovascular risk factors in a population of adults aged 50-90. Clin Endocrinol (Oxf). 2006;65(2):169-77.
  53. Mathus-Vliegen EM, Obesity Management Task Force of the European Association for the study of obesity. Prevalence, pathophysiology, health consequences and treatment options of obesity in the elderly: a guideline. Obes Facts. 2012;5(3):460-83.
  54. Leong KS, Walker AB, Martin I, Wile D, Wilding J, MacFarlane IA. An audit of 500 subcutaneous glucagon stimulation tests to assess growth hormone and ACTH secretion in patients with hypothalamic-pituitary disease. Clin Endocrinol (Oxf). 2001;54(4):463-8.
  55. Littley MD, Gibson S, White A, Shalet SM. Comparison of the ACTH and cortisol responses to provocative testing with glucagon and insulin hypoglycaemia in normal subjects. Clin Endocrinol (Oxf). 1989;31(5):527-33.
  56. Orme SM, Price A, Weetman AP, Ross RJ. Comparison of the diagnostic utility of the simplified and standard i.m. glucagon stimulation test (IMGST). Clin Endocrinol (Oxf). 1998;49(6):773-8.
  57. Tavares AB, Seixas-da-Silva IA, Silvestre DH, Paixao CM, Jr., Vaisman M, Conceicao FL. Potential risks of glucagon stimulation test in elderly people. Growth Horm IGF Res. 2015;25(1):53-6.
  58. Toogood A, Brabant G, Maiter D, Jonsson B, Feldt-Rasmussen U, Koltowska-Haggstrom M, et al. Similar clinical features among patients with severe adult growth hormone deficiency diagnosed with insulin tolerance test or arginine or glucagon stimulation tests. Endocr Pract. 2012;18(3):325-34.
  59. Bowers CY. GH releasing peptides - structure and kinetics. J Pediatr Endocrinol. 1993;6(1):21-31.
  60. Wang G, Lee HM, Englander E, Greeley GH, Jr. Ghrelin - not just another stomach hormone. Regul Pept. 2002;105(2):75-81.
  61. Piccoli F, Degen L, MacLean C, Peter S, Baselgia L, Larsen F, et al. Pharmacokinetics and pharmacodynamic effects of an oral ghrelin agonist in healthy subjects. J Clin Endocrinol Metab. 2007;92(5):1814-20.
  62. Garcia JM, Swerdloff R, Wang C, Kyle M, Kipnes M, Biller BM, et al. Macimorelin (AEZS-130)-stimulated growth hormone (GH) test: validation of a novel oral stimulation test for the diagnosis of adult GH deficiency. J Clin Endocrinol Metab. 2013;98(6):2422-9.
  63. Garcia JM, Biller BMK, Korbonits M, Popovic V, Luger A, Strasburger CJ, et al. Macimorelin as a diagnostic test for adult growth hormone deficiency. J Clin Endocrinol Metab. 2018;103(8):3083-93.
  64. Garcia JM, Biller BMK, Korbonits M, Popovic V, Luger A, Strasburger CJ, Chanson P, Swerdloff R, Wang C, Fleming RR, Cohen F, Ammer N, Mueller G, Kelepouris N, Strobl F, Ostrow V, Yuen KCJ. Sensitivity and specificity of the macimorelin test for diagnosis of AGHD. Endocr Connect. 2021 Jan;10(1):76-83.
  65. Global News Wire. https://www.globenewswire.com/en/news-release/2022/08/29/2505902/0/en/Aeterna-Zentaris-Set-to-Regain-Full-Rights-to-Macrilen-Macimorelin-in-U-S-and-Canada-from-Novo-Nordisk.html. Accessed August 6, 2023.
  66. Global News Wire.https://www.globenewswire.com/en/news-release/2023/04/05/2641603/0/en/Aeterna-Zentaris-Provides-Update-on-Macrilen-Macimorelin-Reiterates-Advancement-of-Pediatric-DETECT-Trial.html. Accessed August 6, 2023.
  67. Junnila RK, Strasburger CJ, Bidlingmaier M. Pitfalls of insulin-like growth factor-I and growth hormone assays. Endocrinol Metab Clin North Am. 2015;44(1):27-34.
  68. Clemmons DR. Consensus statement on the standardization and evaluation of growth hormone and insulin-like growth factor assays. Clin Chem. 2011;57(4):555-9.
  69. Monthly Prescribing Reference. Macrilen Rx. Available at: https://www.empr.com/drug/macrilen/. Accessed October 28, 2019.

Radiotherapy for Pituitary Tumors

ABSTRACT

 

Pituitary adenomas have been historically managed on a multidisciplinary level with surgery, medical therapy, and radiotherapy to control symptoms secondary to mass-effects and hypersecretion of hormones. While transsphenoidal surgery represents the standard initial approach in the majority of cases, radiotherapy is a valuable and effective treatment option for recurrent adenomas, or lesions not amenable to surgery or medical therapy. Following radiotherapy, tumor growth control (over 90% in most series), plus the normalization of hormones, occurs in a large proportion of treated patients, independent of tumor subtype. Over the last decades, radiotherapy technological advances have allowed the reduction of dose to uninvolved brain while maintaining an effective therapeutic dose to the tumor. This has generated debate on the superiority of some radiotherapy techniques over others. The clinical efficacy of conventionally-fractionated treatment (25 to 30 fractions delivered over 5 to 6 weeks), in the form of 3D-conformal radiotherapy (CRT) or intensity-modulated radiotherapy (IMRT) and the more refined “stereotactic” – highly conformal - fractionated radiotherapy (SFRT), can be compared to that provided by “radio-surgical” (SRS) techniques of irradiation (where the tumor is treated with single high dose of radiation). Due to the lack of randomized control trials addressing this issue, the evidence provided in retrospective studies of different radiotherapy technologies is critically reviewed in this chapter. 

 

INTRODUCTION

 

Pituitary adenomas are mostly benign tumors and comprise about 10% of all intracranial tumors [1, 2]. Radiotherapy has an important and long-established role as part of the multi-disciplinary management of both non-functioning and functioning adenomas. There has been a steady evolution in radiotherapy technologies since radiotherapy was first used to treat pituitary adenomas more than 100 years ago [3]. Despite decades of clinical experience, there remains a paucity of randomized clinical trials to enable a robust evidence-based approach to the optimal use of radiotherapy. This is to some extent compensated for by the large number of non-randomized largely retrospective case series which provide evidence on relevant clinical outcomes and toxicities associated with pituitary radiotherapy. Nevertheless, given the nature of the available data, there continue to be areas of controversy regarding the use of particular radiotherapy modalities. We review the available published data on modern radiotherapy techniques for the treatment of pituitary adenomas to provide a rational basis for the selection of radiotherapy technologies.

 

RATIONALE FOR PITUITARY RADIOTHERAPY

 

Traditional practice had been to use post-operative radiotherapy for all patients with a residual non-functioning pituitary adenoma after surgical resection, as it was considered that otherwise most would subsequently progress [4, 5]. With improvements in surgical techniques, and the development of magnetic resonance imaging (MRI), post-operative radiotherapy is no longer routinely used, even in the presence of residual tumor. The use of post-operative pituitary radiotherapy is now based on a risk assessment. In patients with non-functioning adenomas, radiotherapy is generally withheld until the time of progression, unless there are concerns of significant threat to function (vision) with tumor progression, or the histology raises concerns of earlier recurrence risk (e.g., atypical features, silent corticotroph adenoma). When radiotherapy is used for patients with progressive non-functioning adenomas, tumor control is achieved in over 90% of patients at 10 years, and in 85-92% at 20 years [5-13].

 

In patients with functioning adenomas, radiotherapy is used when surgery fails to achieve hormone normalization and/or when medical treatment is insufficient to control hormone secretion or is not considered appropriate, often due to toxicities. Hormone levels decline slowly following radiotherapy, consequently normalization may take from months to years to achieve. The time required to achieve hormone normalization is primarily related to the pre-treatment hormone levels. Nevertheless, despite this temporal delay, the majority of patients will eventually achieve normalization of excess pituitary hormone secretion following radiotherapy [14].

 

CURRENT TECHNIQUES OF PITUITARY RADIOTHERAPY

 

The principal aim of pituitary radiotherapy techniques has always been to deliver an effective treatment dose to the target tumor volume while at the same time minimizing the radiation dose delivered to surrounding normal tissues, thereby minimizing the risk of normal tissue damage. Improved radiotherapy treatment precision, with the use of the modern radiotherapy techniques described in this chapter, relies on the increased accuracy in tumor volume delineation achieved by using modern MRI imaging technology. Over the last twenty years there have been a number of developments in techniques for pituitary radiotherapy which have largely amounted to refinements of existing technologies. However, the overall success of modern high precision pituitary radiotherapy techniques is largely a function of the quality of a treatment center’s infrastructure and its expertise and accuracy in identifying the target tumor volume, rather than of the particular radiotherapy technique that is used to deliver treatment.

 

3D-Conformal RT

 

Until the last decade, the standard of care for pituitary radiotherapy was three-dimensional (3D) conformal radiotherapy (CRT). CRT uses pre-treatment computed tomography (CT) and MRI imaging for computerized 3D radiotherapy treatment planning. CRT treatment is planned and delivered using a non-invasive method of patient immobilization. The tumor is visualized on unenhanced magnetic resonance imaging co-registered with planning computed tomography (CT). The treatment target is delineated on the MRI scan (in the three orthogonal planes), while radiotherapy dosimetry is calculated using the CT scan data.

 

The treatment target comprises the visible residual tumor and also accounts for any pre-operative extension of disease whilst sparing the optic chiasm where possible after decompression. An isotropic margin of 5-10 mm is added to account for areas of uncertainty in volume delineation, the transsphenoidal surgical route and any set-up variation. The whole pre-operative extent of the tumor is not included within the treatment volume as debulking of large, and particularly cranially extending tumors, often leads to the return of normal anatomical structures to their pre-morbid positions with no residual tumor present. On the other hand, tumors are frequently not removed from the walls of the cavernous sinus, particularly if the sinus is involved, and so the lateral extent of the radiotherapy target does not tend to alter with surgery. The resulting volume outlined on the treatment planning system therefore encompasses both the visible tumor and also any regions of presumed residual tumor. Normal tissue structures adjacent to the pituitary, such as the optic chiasm and optic nerves, the brain stem and the hypothalamus, may also be outlined to aid in treatment planning, and also to enable the calculation and recording of normal tissue

dosimetry, although with conventional fractionated radiotherapy all the structures are treated to below the limits of radiation tolerance in terms of structural damage.

Figure 1. CT-MRI co-registration for planning purposes.

 

Reproducible patient immobilization is vital for the delivery of safe and accurate CRT. The immobilization system used should be well tolerated and must reliably minimize patient movement during both pre-treatment imaging and treatment delivery itself. The most commonly used system for immobilization for CRT is a custom-made closely fitting lightweight thermoplastic mask which is applied and molded directly to the patient’s face in the treatment planning process. The repositioning accuracy of this system is very good at around 3-5mm [15], and can be improved to 2-3mm, by using a tighter fitting but less comfortable mask [16].

 

CT imaging for CRT planning is performed with the patient lying in the radiotherapy treatment position within the immobilization system and co-registered with the MRI (Figure 1) 3D computerized radiotherapy planning is followed by robust quality assurance (QA) procedures to ensure the accuracy of the whole process both before and during treatment. The planning system defines the number, shape, and orientation of radiation beams to achieve uniform dose coverage of the target volume with the lowest possible dose to the surrounding normal tissues. As the dose to the tumor is below the radiation tolerance dose of the surrounding normal tissue structures, no specific measures are generally needed, or taken, during treatment planning to avoid the optic apparatus, hypothalamus, and brain stem. In any case, for many patients requiring pituitary irradiation, some of these entire normal structures lie within, or in close proximity to, the target volume and cannot be avoided without compromising the efficacy of treatment.

 

Localized irradiation is achieved using treatment in multiple beams each shaped to conform to the shape of the tumor using a multileaf collimator (MLC). Traditionally, beam arrangements used for CRT consisted of three fixed beams (an antero-superior beam and two lateral beams) (Figure 2).

 

Figure 2. Example of beam arrangement and dose distribution in a traditional CRT plan (one antero-superior beam and two lateral beams).

 

Intensity-Modulated RT

 

Techniques for varying the radiation dose intensity across a beam, by moving MLC leaves into the beam path, are now standard and are collectively referred to as intensity modulated radiotherapy (IMRT). IMRT is a form of 3D CRT which can spare critical structures, especially within a concave PTV. Although IMRT offers no significant advantage in comparison with CRT for target volume dose coverage [

], its improved conformality can allow for reduced radiation dose delivery to adjacent normal tissues. This can be of particular use in tumor with suprasellar extension, where the dose delivered to the medial temporal lobes can reduced. The technique of arcing IMRT (described as VMAT or RapidArc) offers a fast way of delivering complex IMRT and is increasingly used as an alternative to fixed-field techniques (Figure 3).

 

Figure 3. Example of beam arrangement and dose distribution in a static field IMRT plan (left) and in a VMAT plan (right) for the same patient. Note the better conformality of the high radiation dose region to the target volume in comparison with the CRT plan in Figure 2.

 

Patient immobilization and the imaging required for target volume definition are no different for IMRT treatment than for CRT as described above. Similarly, there are robust QA procedures to ensure the accuracy of IMRT treatment planning and delivery.

 

Stereotactic Radiotherapy Techniques

 

The term “stereotactic” is derived from long-established neurosurgical techniques, and denotes a method of determining the position of a lesion within the brain using an external 3D co-ordinate system based on a method of immobilization, usually an invasive neurosurgical stereotactic head frame [18-20]. Stereotactic radiotherapy originally referred to radiotherapy treatment delivered to an intracranial target lesion that was located by stereotactic means in a patient immobilized in a neurosurgical stereotactic head frame.

 

Stereotactic radiotherapy was first delivered with a multiheaded cobalt unit described as the gamma-knife (GK) which uses multiple cobalt-60 sources arranged in a hemispherical distribution with collimators to achieve a circumscribed spherical dose distribution of 4-18mm diameter [20]. Subsequent development of the GK has allowed larger non-spherical tumors to be treated by combining several radiation spheres using a multiple isocenter technique.

 

Due to the invasive nature of the GK stereotactic head frame (surgically fixed to the skull), GK radiation treatment is delivered as a single large dose during one combined treatment planning and delivery session. This single fraction stereotactic radiation technique was termed ‘radiosurgery’ [18]. The GK radiosurgical procedure aimed to create a non-invasive radiation-based analogue of an open neurosurgical ablation of an intra-cranial target lesion. It should be emphasized, however, that aside from the use of a surgically-fitted stereotactic frame, GK radiosurgery and open neurosurgery are quite distinct procedures, and GK radiosurgery is a radiotherapeutic rather than a surgicalintervention, particularly as the commonly used doses are not “ablative”.

 

Subsequently, linear accelerators (linacs) were adapted to deliver radiosurgery (single fraction radiation) using multiple arcs of rotation, achieving the same dose distribution as that delivered by the GK. With the introduction of non-invasive relocatable stereotactic head frames, which enabled stereotactic radiation to be given in a number of treatment sessions, stereotactic radiotherapy was delivered as fractionated treatment to conventional doses [21, 22]. Initially, specifically adapted linacs were required, but the precision of modern linacs is now such that they do not generally require modification for stereotactic radiotherapy. The improved patient immobilization, more accurate tumor target localization using cross-sectional image for treatment planning, and high precision radiation treatment delivery to the tumor target, enabled a reduction in the margins around the radiotherapy target volume (the gross tumor volume (GTV) to planning target volume (PTV) margin), therefore achieving greater sparing of surrounding normal tissues than can be obtained with standard CRT techniques.

 

The miniaturization of a 6MV linear accelerator has allowed for its mounting on a high precision industrial robotic arm, and this has been combined with real time kV imaging for target tracking during treatment to create a robotic frameless stereotactic radiotherapy machine that is commercially known as the Cyberknife (CK) [23]. The CK uses multiple narrow, low dose rate photon beams, which have to be summated, to create a dose distribution equivalent to that achieved with other techniques. The need to summate contributions from multiple narrow beams results in longer treatment times per fraction than with other techniques and requires that CK treatment be given as a single large fraction (SRS), or as a few large fractions delivered over the course of a week or so (hypofractionated stereotactic radiotherapy).

 

While the term stereotactic radiotherapy continues to be used, “stereotaxy” as initially used for neurosurgery and subsequently for target localization in radiotherapy is no longer necessary and not in routine use, as modern MR and CT imaging with on treatment image guidance allow for equivalent high-precision treatment delivery. The appropriate modern terminology for the best and most accurate techniques of treatment delivery should be high precision conformal radiotherapy. Nevertheless, the term stereotactic used in conjunction with fractionated treatment (see below), while largely outmoded, remains in use with no clear meaning other than presumably denoting accuracy. Stereotactic localization, however, largely remains the standard of practice with single fraction treatment (GK radiosurgery).

 

Radiotherapy Fractionation

 

TERMINOLOGY

 

The term ‘radiosurgery’ is used for radiation treatment that is given as a single large dose (a single fraction), and the term radiotherapy is used for treatment that is given as multiple, usually daily, small doses over a period of weeks (fractionated treatment). The fractionation of radiation treatment is a mechanism for protecting normal tissues, through recovery between fractions, and permits the delivery of higher total doses of radiation than can be given as single fractions [24].

 

Similarly, stereotactic radiotherapy to the pituitary can be given in multiple doses as fractionated stereotactic conformal radiotherapy (SCRT or fSRT), or as a single large dose when it is described as stereotactic radiosurgery (SRS). SCRT/fSRT is generally delivered using a linac. SRS has most frequently been delivered using a GK, but can also be delivered using a linac or a robotic arm mounted linac (CK). Treatment given in fewer large fractions is described as hypofractionated RT.

 

BIOLOGICAL RATIONALE

 

The use of single fraction SRS is based on a belief, prevalent in the literature, that there is greater clinical benefit from single fraction rather than fractionated irradiation for pituitary adenomas. This belief was based on radiobiological modelling which defines equivalent radiation doses and fractionation schemes through biologically derived parameters [24, 25], mainly from the radiobiology of malignant tumors and some normal tissues. Such models are not validated for single fraction treatments [26], and the corresponding biological parameters necessary to calculate equivalent radiation doses do not exist for benign tumors. Publications claiming theoretical benefit of single fraction radiosurgery over fractionated irradiation [25] are based on constants that are not derived from experimental data and may therefore be misleading.

 

The therapeutic effect of radiation on malignant tumors is thought to be due to tumor cell attrition, either as apoptosis, or reproductive cell death, secondary to radiation-induced DNA damage. As a consequence, the time taken for an irradiated tissue to manifest radiotherapy related effects is proportional to the rate of cell proliferation in the tissue. In tissues with rapidly proliferating cells (malignant tumors), radiation effects are expressed either during or immediately after a course of radiotherapy, while in a tissue with a slowly proliferating cell population, such as benign tumors, radiotherapy effects may take many months or years to manifest. It is assumed that the beneficial effects of radiation in pituitary adenomas conform to these same mechanistic principles with the radiation-induced depletion of pituitary adenoma tumor cells, and with the adenoma being considered a slowly proliferating tissue. As benign tumors are rarely grown in culture, the precise mechanism of the observed clinical benefit of irradiation is not elucidated and remains largely theoretical. The surrounding normal brain tissue is also considered to consist largely of slowly proliferating cell populations, although critical cell populations with faster turnover, such as blood vessels, are also present and are affected by radiation.

 

DOSE FRACTIONATION SCHEMES FOR PITUITARY ADENOMAS

 

Conventional CRT and fractionated SCRT are given to total dose of 45 to 50 Gy at 1.8 Gy per fractionation, once a day, five days per week. These treatment doses are below the tolerance of central nervous system neural tissue, and the risk of structural damage due to such treatment is <1% [27, 28]. While, theoretically, single large doses of radiation as used in SRS may result in a higher tumor cell kill than the equivalent total dose given over a small number of fractions, this is also true for the normal tissue cell population and leads to normal tissue toxicity which may not be acceptable if it affects critical regions such as optic chiasm [28].

 

As most pituitary adenomas requiring radiation treatment lie in close proximity to the optic apparatus, and to the cranial nerves in the cavernous sinus, SRS is suitable only for small lesions located away from critical structures, and the optic apparatus should not exceed single doses above 8Gy [28]. Fractionated SRT, using up to 5-fractions over a week course, is another feasible alternative fractionation scheme delivered by LINACs, Cyberknife or frameless radiosurgery.

 

For larger NFPA with chiasmatic involvement, hypofractionation can allow for safe delivery of enhanced biologically effective doses compared to conventional fractionation. The safety of this scheme has been recently reported in a cohort of NFPA, the majority with abutment or compression of optic chiasm, who had satisfactory local control compared to SRS with acceptable toxicity for visual preservation [29].

 

Linac Based SCRT/FSRT    

 

For fractionated stereotactic radiotherapy, patients are immobilized in a non-invasive relocatable frame with a relocation accuracy of 1-2mm [21, 22], or a precisely fitting thermoplastic mask system with an accuracy of 2-3mm [16]. Sub-millimeter repositioning accuracy can now be achieved with thermoplastic mask immobilization by means of image guidance techniques which can determine and apply daily online setup corrections [30]. As for conventional CRT, the GTV is outlined on an MRI scan co-registered with a CT scan. The PTV margin used for SCRT is smaller than for conventional CRT, typically in the region of 3-5mm based on the overall accuracy of the treatment system, the principal determinant of which is the repositioning accuracy of the patient in the immobilization device [31] and the ability to correct it with on treatment imaging (image guidance). For such precision treatment, accurate localization of the tumor volume is of paramount importance in order to avoid treatment failure due to exclusion of a part of the tumor from the treatment volume.

 

SCRT employs a larger number of radiotherapy beams than conventional CRT (usually 4-6). Each beam is conformed to the shape of the PTV using a narrow leaf MLC (5mm width known as mini MLC, or 3mm width known as micro MLC). MLC leaves can be used to modulate the intensity of the radiation beam during its delivery as in intensity-modulated radiotherapy (IMRT). More recently, arc-based or rotational techniques (volumetric modulated arc therapy or VMAT) have been introduced in the clinical practice to overcome some of the limitation of IMRT (complex planning and QA process). The continuous rotation of the radiation source allows the patient to be treated from a full 360° beam angle in a shorter time interval. Fractionated SCRT (fSRT) combines the precision of stereotactic patient positioning and treatment delivery with standard radiotherapy fractionation, which preferentially spares normal tissue. Complete avoidance of surrounding normal tissue structures, such as the optic apparatus, is not generally practiced, as the dose fractionation schemes used are below the radiation tolerance doses of the CNS. Nonetheless arc techniques are used to minimize the dose bilaterally to the temporal lobes with the aim of reducing the impact of treatment on patients’ cognitive function. The fractionated SCRT technique is suitable for pituitary adenomas of all sizes, regardless of their relationship to adjacent critical normal tissue structures.

 

Linac Based SRS

 

Linac based SRS can be delivered using either a relocatable or an invasive neurosurgical stereotactic frame. Use of an invasive neurosurgical frame necessitates that the treatment planning and delivery procedures are carried out and completed within a single day. Computerized treatment planning defines the arrangement of the radiation beams, as in SCRT. SRS can be planned either as multiple arcs of rotation, simulating GK SRS treatment, and producing small spherical dose distributions, or as multiple fixed conformal fields. Multiple arc SRS using a linear accelerator, employing multiple isocenters, is a cumbersome and rarely used technique. The use of multiple fixed fields is generally confined to fractionated treatment, although it can also be used for single fraction SRS. Because of the potentially damaging effect of large single fraction radiation doses on normal tissue structures, SRS is only suitable for small pituitary adenomas that are at least 3-5mm away from the optic chiasm.

 

Several dosimetry studies have shown that linear accelerators could deliver the same SRS doses to pituitary tumors as GK, with comparable conformity indices and OAR doses. Linac SRS has the advantage of being available, efficient with a less beam-on time, so could be considered for radiosurgery of pituitary adenomas [32]..

 

Gamma Knife SRS

 

For GK SRS, patients are immobilized in an invasive neurosurgical stereotactic frame. A relocatable non-invasive stereotactic frame has become available, enabling the delivery of hypofractionated stereotactic radiotherapy treatment in addition to SRS, and experience with this system is increasing [33, 34]. GK SRS delivers a single high dose, in a spherical distribution, of 4-18mm diameter. Larger, non-spherical tumors, which represent the majority of pituitary adenomas, are treated by combining several such spherical dose volumes using a multiple isocenter technique. The appropriate number and distribution of isocenters is defined using a 3D computer planning system which also allows for selective plugging of some of the cobalt source positions to enable shaping of the high dose volume envelope. The use of multiple isocenters results in dose inhomogeneity within the target volume, with small areas of high radiation dose (hot spots) in the regions of overlap of the radiation dose spheres. This may lead to radiation damage if critical normal structures, such as cranial nerves, lie within these hot spots. GK SRS is given to doses of 12 - 35Gy to the tumor margin with doses to the optic chiasm and the other cranial nerves in the cavernous sinus limited to 8-10Gy and 16-18 Gy respectively. royal sinus invasion has been reported as a significant predictor of poor outcomes after surgical resection. Different series have shown good local control using GK for positive residuals within the cavernous sinus after surgical resection [35-39].  

 

Although the total dose delivered with fractionated meanings of irradiation is largely consistent within different publications (45-50.4 Gy), the range of dose prescriptions between secretory and non-functioning adenomas treated with single fraction SRS tends to be different. The rationale behind this practice is based on the observation that a more rapid hormone normalization was reported in single studies using higher doses to treat secreting tumors [40, 41]. In absence of a strong radiobiological model and of prospective randomized studies in support, the relationship between dose and endocrine remission warrants further investigation.

 

Robotic Mounted Linac SRS

 

Cyberknife has been used to treat pituitary adenomas using a variety of dose/fractionation regimens, with a tendency to deliver treatment as hypofractionated radiotherapy in 3 to 5 fractions, rather than as single fraction SRS doses.

 

Proton Therapy

 

Proton beams, heavy charged particles with similar radiobiological effectiveness as photons, have been in use at a small number of centers with the relevant facilities since the late 1960s [42, 43]. Proton therapy was initially used in two US centers (Boston, MA, and Loma Linda, CA) and then subsequently in Europe (d’Orsay, France) and Japan (Tsukuba, Japan); these centers have reported the majority of the initial clinical results. The introduction of proton therapy had been underpinned by planning studies demonstrating, in selected cases, improved dose distribution of protons compared with photons.

 

The principal theoretical advantage of proton therapy over photon therapy is the deposition of energy at a defined depth in tissue (the Bragg peak) with little energy deposition beyond that point [44]. These properties make the use of protons appealing for tumors lying in close proximity to critical dose-limiting normal tissues, which is a bar to safe dose escalation using conventional photon radiotherapy, or when a reduction of low dose (the low dose radiation “bath” responsible for the late sequelae of radiotherapy) to the normal brain tissues is of particular clinical evidence, as in children.

 

Current indications for the use of protons within the UK Specialized Commissioning Team include the treatment of craniopharyngiomas and pituitary adenomas up to the age of 24 years old based on theoretical reduction in the possible late side effects of brain radiation, such as second malignancy, neuro-cognitive deficits and cerebrovascular disease [45].

 

Peptide Receptor Radionuclide Therapy (PRRT)

 

PRRT is a form of internal radiation therapy directed to the tumor tissues expressing peptide receptors using gamma emitting radiopharmaceuticals. It is typically used for neuroendocrine tumors; however, it was investigated as a treatment option for aggressive pituitary tumors refractory to other treatment modalities. Different pituitary tumors express somatostatin receptors and show uptake of radiolabeled somatostatin analogues like 68Ga-DOTATATE. The 2018 guidelines of the European Society of Endocrinology listed PRRT as an alternative treatment option for aggressive pituitary tumors refractory to other lines of treatment including temozolomide [46].The treatment doses and the type of nucleotide used varied in the available studies, with only small patient numbers being reported [47, 48].

 

CLINICAL OUTCOMES FOLLOWING PITUITARY RADIOTHERAPY

 

The clinical efficacy of radiotherapy for pituitary adenomas should be assessed by overall survival, actuarial tumor control (progression-free survival, PFS), and quality of life. Few publications focused on quality of life assessment after radiotherapy in pituitary tumors [49-51], while commonly reported endpoints for retrospective studies of radiation treatment for non-functioning pituitary adenomas are local tumor control, and long term morbidity.

 

In patients with functioning pituitary adenomas, the principal endpoint, in addition to PFS and morbidity, is the rate of normalization of elevated pituitary hormone levels. The rate of pituitary hormone decline after irradiation varies with the type of functioning tumor, and the time to reach normal hormone levels is dependent on the initial pre-treatment hormone levels [52]. The appropriate comparative measure for each pituitary hormone is the time to reach 50% of the pre-treatment hormone level, and this should be corrected for the confounding effect of medical treatment.

 

Surrogate endpoints such as ‘tumor control rate’ and the ‘proportion of patients achieving normal hormone levels’ do not, of themselves, provide adequate information on the efficacy of different pituitary irradiation techniques and are potentially misleading [53]. Tumor control rate must be quoted with an indication of the time or duration of follow-up required to achieve the stated level of control. Similarly, the proportion of patients achieving normal hormone levels following treatment is meaningful only when described in terms of the relationship to pre-treatment hormone levels. Due to the use of such surrogate endpoints in published retrospective series, inappropriate and incorrect claims have been made in the literature for superiority of one technique of irradiation over another.

 

Given that the published data on the efficacy of the various available techniques for pituitary irradiation consist entirely of retrospective case-series, the available data inevitably suffer from selection bias. While SCRT is suitable for the treatment of all pituitary tumors, irrespective of size, shape or proximity to critical normal tissue structures, SRS is only suitable for treatment of small tumors away from the optic chiasm. As a result, studies reporting the efficacy of SRS mostly deal with smaller tumors, which are typically associated with lower hormone levels if the adenomas are functioning. Therefore, the reported results of studies of SRS do not necessarily apply to the generality of pituitary adenomas and may give a false impression of greater efficacy if only more favorable cases are treated.

 

THE EFFICACY AND TOXICITIES OF TREATMENT

 

Conventional RT and CRT

 

The efficacy of modern stereotactic pituitary radiotherapy and pituitary radiosurgical techniques must be assessed in the light of the results achieved with standard treatment, which is conventional conformal radiotherapy. Large and mature case series provide data on the long-term effectiveness of CRT in controlling pituitary tumor growth and hormone secretion.

 

TUMOR CONTROL

 

The long-term results following pituitary CRT from case series published in the literature are shown in Table 1 [5-14, 17, 54-66]. The actuarial PFS is in the region of 80%-90% at 10 years and 75%-90% at 20 years [14, 55]. The single largest series of patients with pituitary adenomas treated with conventional fractionated radiotherapy is that from The Royal Marsden Hospital which reported a 10-year PFS of 92% and a 20-year PFS of 88% [8].

Post operative radiotherapy has been reported to provide excellent local control of non-functioning tumors when offered for progressive residual disease with almost no radiological evidence of tumor progression up to 15 years of follow-up [67].

 

ENDOCRINE CONTROL  

 

Fractionated irradiation leads to normalization of excess pituitary hormone secretion in the majority of patients, albeit with some time delay following treatment. For acromegaly, RT achieves normalization of GH/IGF-I levels in 30-50% of patients at 5-10 years, and in 75% of patients at 15 years, after treatment (Table 2) [14, 55]. As the time to normalization of GH levels is related to the pre-treatment GH level, the time to achieve a 50% reduction in GH levels, which takes into account the starting GH level, is in the region of 2 years, with IGF-1 reaching half of pre-treatment levels somewhat after the GH [58, 60].

 

A 10-year follow-up for more than 600 acromegaly patients was published by the Swedish Pituitary Register 2022. It has reported 78% of IGF-1 normalization rate with an annual rate of increased hormonal control of 1.23%. One third of the patients required bi-modality therapy to achieve hormonal control and 5% required triplet therapy i.e. surgical resection, medical treatment and radiotherapy with a trend towards reduced use of conventional radiotherapy doses [68]. 

 

Table 1. Summary of Results of Published Series on Conventional RT for Pituitary Adenomas

Authors

Type of adenoma

Number of patients

Follow-up

(median years)

Actuarial progression free survival (PFS) (%)

Late toxicity (%)

Visual Hypopituitarism

Grigby at al.,1989 [6]

NFA, SA

121

11.7

89.9 at 10 years

1.7

NA

McCollough et al., 1991 [7]

NFA, SA

105

7.8

95 at 10 years

NA

NA

Brada et al., 1993 [8]

NFA, SA

411

10.8

94 at 10 years

88 at 20 years

1.5

30 at 10 years

Tsang et al., 1994 [9]

NFA, SA

160

8.7

87 at 10 years

0

23**

Zierhut et al., 1995 [10]

NFA, SA

138

6.5

95 at 5 years

1.5

27**

Estrada et al., 1997 [56]

SA (ACTH)

30

3.5

73 at 2 years*

0

48**

Rush et al., 1997 [11]

NFA, SA

70

8

NA

NA

42**

Breen et al., 1998 [12]

NFA

120

9

87.5 at 10 years

1

NA

Gittoes et al., 1998 [5]

NFA

126

7.5

93 at 10 and 15 years

NA

NA

Barrande et al., 2000 [57]

SA (GH)

128

11

53 at 10 years*

0

50 at 10 years

Biermasz et al., 2000 [58]

SA (GH)

36

10

60 at 10 years*

0

54 at 10 years

Sasaki et al., 2000 [13]

NFA, SA

91

8.2

93 at 10 years

1

NA

Epaminonda et al., 2001 [59]

SA (GH)

67

10

65 at 15 years*

0

NA

Minniti et al., 2005 [60]

SA (GH)

45

12

52 at 10 years*

0

45 at 10 years

Langsenlehner et al., 2007 [61]

NFA, SA

87

15

93 at 15 years

 

0

88 at 10 years

Minniti et al., 2007 [62]

SA(ACTH)

40

9

78 and 84 at 5 and 10 years*

0

62 at 10 years

Rim et al., 2011 [63]

NFA, SA

60

5.6

96 at 10 years (NFA),

66 at 10 years (SA)

0

76 at 10 years

Kim et al., 2016 [65]

NFA, SA

73

8

98 at 10 years

0

NA

Patt et al., 2016 [66]

SA (GH)

36

4.9 (mean)

89 at 5 years

0

33

NFA, non-functioning adenoma; SA, secreting adenoma; NA, not assessed, ACTH-Cushing, GH- acromegaly, *hormone concentration normalization, **no time specified

 

After RT for Cushing’s disease, urinary free cortisol (UFC) is reduced to 50% of the pre-treatment levels after an interval of 6-12 months, and plasma cortisol after around 12 months [62]. The median time to cortisol level normalization is around 24 months after treatment [62]. The overall tumor and hormone control rates in the reported studies, after a median follow-up of 8 years, are 97% and 74% respectively [64]. Pituitary radiotherapy is rarely used to treat patients with prolactinoma. Occasional patients who fail surgery and medical therapy have been treated with RT, and the reported 10-year tumor and hormone control rates are 90% and 50% respectively [69-71].

 

TOXICITY

 

The toxicity of RT with total treatment doses of 45-50Gy with daily fraction sizes of < 2Gy is low. The principal toxicities reported in studies of CRT are described in Table 1.

 

Hypopituitarism  

 

Hypopituitarism is the most common long-term complication following RT, reported to occur in 30-60 % of patients by 10 years after treatment [8, 9, 14]. Pituitary hormone loss is observed to occur in a characteristic sequence, with GH secretion being affected most frequently, followed by the gonadotrophins, ACTH, and then TSH. Long term follow-up after pituitary irradiation, with intermittent testing for deficiency of all pituitary axes, is therefore an essential part of the post-treatment management of these patients.

 

Visual Pathways Deficit and Other Structural CNS Damage

 

The reported incidence of optic neuropathy resulting in visual deficit following CRT is 1-3% [8, 9]. The occurrence of necrosis of normal brain tissue is almost unknown following pituitary RT, although this complication has been reported to occur in 0.2% of patients [72].

 

Cerebrovascular Disease

 

Pituitary disease is, in itself, associated with increased mortality, principally due to vascular disease [73]. An increased incidence of stroke (relative to the general population) in patients treated with RT for both non-functioning and functioning pituitary adenomas has been reported in a number of retrospective cohort studies [74-77]. Whilst it is has long been known that radiotherapy can lead to vascular injury [78], it is not at present clear how much of the excess stroke risk following RT is attributable to radiotherapy, and how much may be due to other potential causes including the metabolic and cardiovascular consequences of hypopituitarism, the effects of associated endocrine syndromes, and the consequences of surgery.

 

In a retrospective cohort study of 342 patients treated with pituitary surgery and post-operative RT, 31 patients died from stroke after a median follow-up interval of 21 years (range, 2-33) [77] and in all cases the probable location of the stroke lesion was within the irradiated volume. Comparison of stroke patients with matched control patients without stroke drawn from the same cohort showed no significant differences in radiotherapy-dependent variables with the exception of the pre-treatment duration of symptoms of hypopituitarism. This suggests that untreated hormone deficiency may be a significant factor in the pathogenesis of stroke in patients treated for pituitary adenoma, rather than or in addition to treatment with radiotherapy. It is likely that the cause of stroke in patients treated with RT for pituitary adenoma is multi-factorial, and the relative contributions of the various possible contributory factors remains to be determined.

 

Second Brain Tumor

 

Intracranial radiotherapy is associated with the development of second, radiation-induced, brain tumors. The cumulative incidence of gliomas and meningiomas following radiotherapy for pituitary adenomas in retrospective case series is reported to be in the region of 2% at 20 years [77, 79-81]. A large retrospective study of patients who received radiotherapy for pituitary and sellar lesions has shown a relative risk of 3.34 (95% confidence interval 1.06-10.6) for development of malignant brain tumors and 4.06 (95% confidence interval 1.51-10.9) for development of meningiomas in comparison with patients who did not receive radiotherapy. Rates were higher in those treated with radiotherapy at a younger age, and there was no difference in incidence rates between conventional or stereotactic radiotherapy (70).

 

In another large retrospective cohort of more than 3600 patients from six adult endocrinology registries, incidence of secondary brain tumors was compared between irradiated and non-irradiated patients with pituitary adenomas and craniopharyngiomas. The relative risk of secondary brain tumors for irradiated patients was 2·18 (95% CI 1·31-3·62, p<0·0001). Cumulative probability of second brain tumor was 4% for the irradiated and 2·1% for the controls at 20 years. Radiotherapy exposure and older age at pituitary tumor detection were associated with increased risk of second brain tumor [82].

 

Cognitive Deficit

 

Radiotherapy treatment to significant volumes of normal brain in children is associated with subsequent neuro-cognitive impairment [27]. However, the evidence for the effect of radiotherapy treatment to small volumes of brain on neuro-cognitive function in adults is weak [27]. The effect of pituitary radiotherapy on neuro-cognitive function is particularly difficult to discern as this cannot be differentiated from the effect of other treatment interventions, and from the effects of the tumor itself [83-85].

 

A retrospective study of 84 patients following transsphenoidal surgery, of whom 39 received post-operative radiotherapy, compared neuro-cognitive function with a large reference sample, considered to be representative of normal population without pituitary disease. While the pituitary cohort had lower scores on the tests of both memory and executive function in comparison with the reference sample, patients who had received radiotherapy showed no significant difference compared to patients treated with surgery alone [86]. A dosimetric study did not find a correlation between radiotherapy dose to the hippocampus and pre-frontal cortex (brain regions known to be important in memory and executive function) and conformal technique of irradiation with cognitive performance [87].

 

Stereotactic Conformal Radiotherapy (SCRT/FSRT)

 

SCRT achieves tumor control and normalization of pituitary hormone hypersecretion at rates similar to the best reports following conventional RT. Longer duration follow-up is required to demonstrate the presumed lower incidence of long-term morbidity following SCRT compared to conventional RT. The results from reported studies of SCRT are summarized below.

 

TUMOR CONTROL

 

SCRT data for 1166 patients with either non-functioning or functioning pituitary adenomas have been reported in 21 studies to date (Table 2) [14, 17, 55, 64, 88-105]. Analysis of published data up to 2020 shows that, at a corrected median follow-up of 56 months (range 9-152 months), tumor control was achieved in 96% of patients. The 5-year actuarial PFS of 92 patients (67 non-functioning, 25 functioning) treated at The Royal Marsden Hospital was 97% [93]. These results are similar to the results seen in patient cohorts treated with conventional RT (Table 1).

 

ENDOCRINE CONTROL  

 

Detailed data on the rate of pituitary hormone decline are not available, although this is expected to be similar to that seen following conventional RT as the same dose-fractionation is used. In The Royal Marsden case series, 6 of 18 acromegalic patients (35%) had normalization of GH/IGF-I levels after a median follow-up of 39 months [93]. Similarly, in another single center study of 20 patients treated with SCRT, normalization of GH levels was reported in 70%, and local tumor control in 100% after a median follow-up of 26 months [90]. The data available on SCRT for patients with Cushing’s disease are limited. In a small series of 12 patients, control of elevated cortisol was reported in 9 out of 12 patients (75%) after a median follow-up of 29 months [92].

 

TOXICITIES

 

Following SCRT, hypopituitarism has been reported in 22% of patients after an overall corrected median follow-up of 57 months (Table 2). The length of follow-up after SCRT is shorter than reported for the mature cohorts treated with RT. It is likely that the rate of hypopituitarism following SCRT will continue to increase as the duration of follow-up increases particularly as the technique of SCRT generally does not avoid either the hypothalamus or the remaining pituitary gland. Other late complications have been rarely reported after SCRT. While the incidence of treatment-related morbidity with SCRT appears to be low, longer duration follow-up is necessary to detect normal tissue toxicity that may only become manifest at a low frequency many years after treatment.

 

Table 2. Summary of Results on Published Studies on SCRT for Pituitary Adenomas

Authors

Number of patients

Follow-up median (months)

Tumor growth control rate (%)

Late toxicity (%)

Visual  Hypopituitarism

Coke et al., 1997 [88]

19*

9

100

0

0

Mitsumori et al., 1998 [89]

30*

33

86 at 3 years

0

20

Milker-Zabel et al., 2001 [90]

68*

38

93 at 5 years

7

5

Paek et al., 2005 [91]

68

30

98 at 5 years

3

6

Colin et al., 2005 [92]

110*

48

99 at 5 years

2

29 at 4 years

Minniti et al., 2006 [93]

92*

32

98 at 5 years

1

22

Selch et al., 2006 [94]

39*

60

100

0

15

Kong et al., 2007 [95]

64*

37

97 at 4 years

0

11

Snead et al., 2008 [96]

100*

6.7 years

98 and 73 at 10 years for NFA and SA

1

35

Roug et al., 2010 [97]

34*

34

91 (50% hormonal normalization)

-

-

Schalin-Jantti et al., 2010 [98]

30

5.3 years

100

0

23

Weber et al., 2011 [99]

27*

72.4

96

4

8

Wilson et al., 2012 [100]

67

5.12 years

88

2

6

Kim et al., 2013 [101]

76*

6.8 years

97.1 at 7 years

0

48 (one or more hormone)

Kopp et al., 2013 [102]

37

57

91.9

5

43

Liao et al., 2014 [106]

34~

36.8 (mean)

100

0

NA

Minniti et al., 2015 [103]

68

75

97 and 91 at 5 and 10 years

0

26

Puataweepong et al., 2015 [107]

94*

72

95

3

9.6

Diallo et al., 2015 [104]

34*

152 (mean)

97

0

39

Barber et al., 2016 [105]

75*

47.5 (mean)

100

1.5

28

Lian et al., 2020 [108]

113*

36

99

0

28.3

* Case series includes secreting adenomas

 

Radiosurgery (SRS)

 

TUMOR CONTROL  

 

The published results of GK SRS for patients with non-functioning and functioning pituitary adenomas have been summarized in systematic reviews [14, 17, 55, 64] and an update with more recently published studies is given in Table 3 [14, 17, 35, 55, 64, 100, 109-130]. The majority of published reports provide information on tumor ‘control rate’, without specifying a time-frame, and therefore provide little useful information on the efficacy of GK SRS. The summary figure for the actuarial 5-year control rate (PFS) following GK SRS for non-functioning adenomas is 95% at 5 years (few 10-year results are available). This is a lower rate of tumor control than expected following RT & SCRT, particularly when it is considered that only small tumors suitable for GK SRS are treated, compared to that adenoma of all sizes treated with RT, CRT & SCRT.

 

Table 3. Summary of Results of Published Series on SRS for Non-Functioning Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Tumor control growth rate (%)

Late toxicity (%)

Visual Hypopituitarism

 

Martinez et al., 1998 [109]

14

26-45

100

0

0

 

Pan et al., 1998 [110]

17

29

95

0

0

 

Ikeda et al., 1998 [35]

13

45

100

0

0

 

Mokry et al., 1999 [111]

31

20

98

NA

NA

 

Sheehan et al., 2002 [112]

42

31*

97

2.3

0

 

Wowra et al., 2002 [113]

45

55

93 at 3 years

0

14

 

Petrovich et al., 2003 [114]

56

36

94 at 3 years

4

NA

 

Pollock et al., 2003 [115]

33

43

97 at 5 years

0

28 and 41 at 2 and 5 years

 

Losa et al., 2004 [116]

56

41*

88 at 5 years

0

24

 

Iwai et al., 2005 [117]

34

60

93 at 5 years

0

6

 

Mingione et al., 2006 [118]

100

45*

92

0

25

 

Liscak et al., 2007 [119]

140

60

100

0

2

 

Pollock et al., 2008 [120]

62

63

95 at 3 and 7 years

0

32 at 5 years

 

Kobayashi et al., 2009 [121]

60

>3 years

97

4.3

8.2 worsening

 

Gopalan et al., 2011 [122]

48

80.5

83

9.4

39

 

Park et al., 2011 [124]

125

62

94 at 5 years and

76 at 10 years

1

24 at 2 years

 

Wilson et al., 2012 [100]

51

4.17 years

100

0

0

 

Runge et al., 2012 [123]

61

83

98

0

9.8

 

Starke et al., 2012 [125]

140

4.2 years

97 at 5 and 87 at 10 years

12.8

30.3

 

El-Shehaby et al., 2012 [126]

38

44*

97

0

0

 

Sheehan et al., 2013+ [127]

512

36

95 at 5 years

6.6

21

 

Lee et al., 2014 [128]

41

48

94 at 5 and 85 at 10 years

2.4

24.4

 

Xu et al., 2014 [129]

34

56

73 at 3 years

24

29

 

Hasegawa et al., 2015 [130]

16

98

100

0

6

 

Graffeo et al., 2018[131]

57

48

99

NA

31 at 5years

 

Oh et al., 2018 [132]

76

53.5

96

NA

24.5

 

Cordeiro et al., 2018 [133]

410

51

94.4

NA

34.7

 

Narayan et al., 2018 [134]

87

48.2

90

8.1

20.7

 

Slavinsky P et al., 2022 [135]

 

28

63

94.2

NA

26%

Maldar AN, et al.,2022[136]

63

47

87.3

NA

26% at 5 years

29.7% at 10 years

*Mean follow-up; NA: not available, + multicenter study, 34 patients had prior CFSR

 

ENDOCRINE CONTROL WITH GK SRS

 

The reported endocrine outcomes following GK SRS for acromegaly are shown in Table 4 [14, 36, 40, 55, 64, 109-111, 114, 121, 137-163]. A summary analysis of the published literature up to 2020 shows that -41% of patients achieved normalization of serum GH, after a median follow-up of 46 months. The time to reach 50% of baseline serum GH, reported in only three studies, is in the region of 1.5-2 years with a slower reduction in IGF-I levels [147, 150, 164], which is similar to the rate reported following conventional RT/CRT.

 

Table 4. Summary of Results of Published Series on SRS for GH-Secreting Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Hormone normalization* (%)

Late toxicity (%)

Visual Hypopituitarism

 

Thoren et al., 1991 [137]

21

64

10

0

15

 

Martinez et al., 1998 [109]

7

26-45

NA

0

0

 

Pan et al., 1998 [110]

15

29

NA

0

0

 

Morange-Ramos et al., 1998 [138]

15

20

20

6

16

 

Lim et al., 1998 [139]

20

26

30

5

5

 

Kim et al., 1999 [165]

11

27

35

NA

NA

 

Landolt et al., 1998 [141]

16

17

50

0

16

 

Mokry et al., 1999 [111]

16

46

31

0

NA

 

Hayashi et al., 1999 [142]

22

>6

41

0

0

 

Inoue et al., 1999 [143]

12

>24

58

0

0

 

Zhang et al., 2000 [144]

68

>12

40

NA

NA

 

Izawa et al., 2000 [145]

29

>6

41

0

0

 

Pollock et al., 2002 [146]

26

36

47

4

16

 

Attanasio et al., 2003 [147]

30

46

23

0

6

 

Choi et al., 2003 [148]

12

43

30

0

0

 

Jane et al., 2003 [149]

64

>18

36

0

28

 

Petrovich et al., 2003 [114]

6

36

100

0

NA

 

Castinetti et al., 2005 [150]

82

49.5*

17

0

18

 

Gutt et al., 2005 [151]

44

22

48

NA

NA

 

Kobayashi et al., 2005 [152]

67

63

17

0

NA

 

Jezkova et al., 2006 [153]

96

54

50

0

26

 

Pollock et al., 2007 [154]

46

63

11 and 60 at 2 and 5 years

0

33 at 5 years

 

Jagannathan et al., 2009 [155]

95

57 *

53

5#

34 (new)

 

Kobayashi, 2009 [121]

49

63

17 (normal or nearly normal)

11

15

 

Wan et al., 2009 [156]

103

60 (minimum)

37

0

1.7**

 

Castinetti et al., 2009 [157]

27

60 (minimum)

42 at 50 months

1.3**

23**

 

Iwai et al., 2010 [158]

26

84

38

0

8

 

Hayashi et al., 2010 [36]

25

36*

40

0

0

 

Erdur et al., 2011 [159]

22

60

55

0

29

 

Sheehan et al., 2011 [40]

130

30

53 at 30 months

0

34

 

Franzin et al., 2012 [160]

103

71

56.9 at 5 years

0

7.8 (new)

 

Liu et al., 2012 [161]

40

72

57.5

0

40 (new)

 

Zeiler et al., 2013 [162]

21

33

30

3.9

13.2

 

Lee et al., 2014 [163]

136

61.5

64.5 and 82.6 at 4 and 8 years

3

33.1

 

Cordeiro et al., 2018 [133]

351

51

NA

NA

38.7

 

Gupta et al.,2018 [166]

25

69.5

28

NA

19.6

Ding et al., 2019 [167]

371

79

59 at 10 years

4

26

 

*mean follow-up; NA not assessed, #3 had previous RT, **whole series

 

A summary analysis of the published literature up to 2020, for patients with Cushing’s disease, shows that 52% achieved biochemical remission (as defined by plasma cortisol and 24-hour UFC level) at a corrected median follow-up of 50 months after SRS (Table 5) [14, 36, 40, 55, 64, 109-111, 114, 121, 138-140, 142, 143, 145, 146, 148, 149, 155-157, 162, 165, 168-178]. The reported time to hormonal normalization ranged from 3 months to 3 years, with no clear difference in the rate of decline of hormone level compared to RT/CRT. The largest single series of GK SRS for Cushing’s disease reported a remission rate of 54%, with 20% of patients who achieved remission subsequently relapsing, suggesting a higher failure rate following GK SRS than following RT/CRT [179].

 

Table 5. Summary of Results of Published Series on SRS for ACTH-Secreting Pituitary Adenomas

 

Authors

Number of patients

Follow-up median (months)

Tumor growth control rate (%)

Hormone normalization*%

Late toxicity

(%)

Visual    Hypopituitarism

 

Degerblad et al., 1986 [168]

29

3-9 years

76

48

NA

55

 

Ganz et al., 1993 [169]

4

18

NA

NA

0

NA

 

Seo et al., 1995 [170]

2

24

100

NA

0

NA

 

Martinez et al., 1998 [109]

3

26-45

100

100

0

0

 

Pan et al., 1998 [110]

4

29

95

NA

0

0

 

Morange-Ramos et al., 1998 [138]

6

20

100

66

0

16

 

Lim et al., 1998 [139]

4

26

NA

25

2

2

 

Mokry et al., 1999 [111]

5

26

93

20

0

2

 

Kim et al., 1999 [165]

8

26

100

60

NA

NA

 

Hayashi et al., 1999 [142]

10

>6

100

10

0

5

 

Inoue et al., 1999 [143]

3

>24

100

100

0

0

 

Izawa et al., 2000 [145]

12

>6

100

17

NA

0

 

Sheehan et al., 2000 [171]

43

44

100

63

2

16

 

Hoybye et al., 2001 [172]

18

17 years

100

83

0

66

 

Kobayashi et al., 2002 [173]

20

60

100

35

NA

NA

 

Pollock et al., 2002 [146]

11

36

85

35

35

8

 

Choi et al., 2003 [148]

9

43

100

55

0

0

 

Jane et al., 2003 [149]

45

>18

100

63

1

31

 

Petrovich et al., 2003 [114]

4

36

NA

50

0

NA

 

Devin et al.,  2004 [174]

35

35

91

49

0

40

 

Castinetti et al., 2007 [175]

40

54

100

42

0

NA

 

Jagannathan et al., 2009 [155]

90

45

96

54

6

22

 

Kobayashi, 2009 [121]

25

64(mean)

100

35

NA

NA

 

Wan et al., 2009 [156]

68

60(minimum)

90

28

0

1.7

 

Castinetti et al., 2009 [157]

18

60(minimum)

NA

50 at 28 months

1.3**

23**

 

Hayashi et al., 2010 [36]

13

36(mean)

97

38

0

0

 

Sicignano et al., 2012 [178]

15

60

97.7

64

NA

12.3

 

Zeiler et al., 2013 [162]

8

35

100

50

3.9

32

 

Sheehan et al., 2013 [177]

96

48

98

70

4

36

 

Marek et al., 2015 [176]

26

78

90.9 at 5 and 10 years

80.7

0

23

 

Cordeiro et al., 2018 [133]

262

51

95.8

NA

NA

26.6

 

Knappe et al., 2020 [180]

119

107

NA

78

NA

NA

 

Gupta et al., 2018  [166]

21

69.5

100

81%

NA

19.6%

*time not specified; NA not assessed

 

In patients with prolactinomas treated with GK SRS the reported time to hormonal response ranged from 5 months to 40 months (Table 6) [14, 40, 55, 64, 109-111, 114, 121, 138-140, 142, 143, 145, 146, 148, 149, 156, 157, 161, 169, 181-186]. At a corrected median follow-up of 43 months (median range 6-60 months), 33% of patients had normalization of serum prolactin concentrations following GK SRS [14]. One study of 35 patients reported a hormonal normalization of 80% after a median of 96 months and a tumor control rate of 97% [184]. There is insufficient information to assess the rate of decline of prolactin following GK SRS in comparison to that following CRT.

 

Table 6. Summary of Results of Published Series on SRS for Prolactin Secreting Pituitary Adenomas

 Authors

Number of patients

Follow-up median (months)

Hormone normalization*%

Late toxicity (%)

Visual        Hypopituitarism

Ganz et al., 1993 [169]

3

18

0

0

NA

Martinez et al., 1998 [109]

5

26-45

0

0

0

Pan et al., 1998 [110]

27

29

30

0

0

Morange-Ramos et al., 1998 [138]

4

20

0

0

16

Lim et al., 1998 [139]

19

26

50

NA

NA

Mokry et al., 1999 [111]

21

31

57

0

19

Kim et al., 1999 [165]

18

27

16

NA

NA

Hayashi et al., 1999 [142]

13

>6

15

NA

5

Inoue et al., 1999 [143]

2

>24

50

0

0

Landolt et al., 2000 [181]

20

29

25

0

NA

Pan et al., 2000 [182]

128

33

41

0

NA

Izawa et al., 2000 [145]

15

>6

16

0

NA

Pollock et al., 2002 [146]

7

26

29

14

16

Choi et al., 2003 [148]

21

43

23

0

0

Jane et al., 2003 [149]

19

>18

11

0

21

Petrovich et al., 2003 [114]

12

36

83

0

NA

Pouratian et al., 2006 [183]

23

55

26

7

28

Jezkova et al., 2009 [184]

35

96

80

NA

NA

Kobayashi, 2009 [121]

27

37(mean)

17

0

0

Wan et al., 2009 [156]

176

60 (minimum)

23

0

1.7

Castinetti et al., 2009 [157]

15

60 (minimum)

46 at 24 months

1.3**

23**

Liu et al., 2013 [185]

22

36

27

-

4.5

Cohen-Inbar et al., 2015 [186]

38

42.3

50

NA

30.3

Ježková et al., 2019 [187]

28

140

82.1

3.6

8.3

 

Early studies of linac based SRS reported results on small numbers of patients, but the available results are broadly equivalent to those reported for GK SRS [17]. In the largest linac based SRS study to date, which included 175 patients with both non-functioning and functioning pituitary adenomas treated using a single dose of 20 Gy, the local tumor control rate was 97% after a minimum of 12 months follow-up [188]. Actuarial 5-year PFS was not reported. Hormonal normalization rates were 47% for GH-secreting adenomas, 65% with Cushing’s disease, and 39% with prolactinomas. The mean time for hormone normalization was 36±24 months. Within the limited follow-up period, 12% developed additional pituitary dysfunction, 3% radiation-induced CNS tissue damage, and 1% radiation-induced optic neuropathy. These results from linac SRS are difficult to evaluate but are broadly similar to those achieved with GK SRS and appear inferior to those obtained with fractionated treatment.

 

TOXICITY

 

In common with other modalities of pituitary irradiation, the most commonly reported complication following GK SRS is hypopituitarism, with a crude incidence ranging from 0% to 66% [14, 55]; the actuarial incidence has not been defined. The expected frequency of visual complications would be low if GK SRS is only offered to patients with a pituitary adenoma at a safe distance from the optic chiasm and nerves (~ 5mm). However, one study in patients with Cushing’s disease reported a 10% incidence of new cranial nerve deficit, with a 6% incidence of optic neuropathy [155]. Similarly, a study reporting results of SRS for prolactinoma noted a 7% incidence of cranial nerve deficit [183]. Although the absolute numbers of patients treated in these studies of GK SRS were small, there is a suggestion that for some patients, possibly with larger tumors, the incidence of optic pathway toxicity with GK SRS is well above what is seen in patients following CRT. Long-term risks of cerebrovascular events and the incidence of second tumors following GK SRS are not yet defined. GK toxicity is expected to be higher when offered after surgical excision rather than as a primary treatment option. In a recently published systematic review and meta-analysis on 1381 patients with pituitary adenomas treated with GK SRS, rates of radiation-induced hypopituitarism were (11.4%) in primary GK compared to (18-32%) in post-operative GK SRS. This highlights the importance of long-term endocrinology follow-up [189].

 

Robotic SRS

 

A small number of retrospective case series on outcomes following CK SRS for pituitary adenomas have been published to date (Table 7) [190-197]. While the published results are comparable to the outcomes achieved with GK SRS, the same criticisms levelled at the GK SRS studies also apply to these early CK SRS series. The duration of follow-up in all the existing CK SRS series is too short to allow meaningful conclusions to be drawn with regard to both efficacy and toxicity outcomes.

 

Table 7. Summary of Results of Published Series on Cyberknife SRS for Functioning & Non-Functioning Pituitary Adenomas

 

Author

Tumor type

Number of patients

Follow-up mean (months)

Tumor Control or Hormone normalization*

(%)

Late toxicity (%)

Visual Hypopituitarism

 

Kajiwara et al., 2005 [190]

14 NFA, 3 PRL, 2 GH, 2 ACTH

21

35.3

95.2TC, 50 HN

4.7

9.5

 

Adler et al., 2006 [191]

12 NFA, 4 GH, 2 ACTH, 1 PRL

19

46

18/19 TC

5.2

NA

 

Roberts et al., 2007 [192]

GH

9

25.4

44.4 HN

0

33

 

Killory et al., 2009 [193]

14 NFA, 4GH, 1 PRL, 1 TSH

20

26.6

100 TC

0

NA

 

Cho et al., 2009 [194]

17 NFA, 3 PRL, 6 GH

26

30

92.3 TC, 44 HN

7.6

0

 

Iwata et al., 2011 [195]

NFA

100

33 median

98 TC

1

4

 

Puataweepong et al., 2015[196]

27 NFA, 7 GH, 5 PRL, 1 ACTH

40

38.5 median

97.5 TC, 54 HN

0

0

 

Iwata et al., 2016 [197]

GH

52

60 median

100 TC, 20.4 HN

0

2.2

 

Plitt et al., 2019 [198]

NFA

53

32.5

98.1 TC

0

1.9

 

Romero-Gameros et al., 2023 [199]

GH

57

48

45.6% HN

0

24.5

                 

TC: Tumor Control; HN: hormone normalization

 

Proton Beam Therapy

 

An early study, published in 1989, of proton beam therapy for pituitary adenomas attempted to compare the effectiveness of this treatment modality to RT/CRT [200]. Follow-up after CRT in 17 patients and after proton therapy in 13 patients found a similar reduction of GH levels in both groups and the small number of patients does not allow for any statistically meaningful comparison. Nevertheless, treatment related side effects, including new hypopituitarism and oculomotor palsies, were more frequent in proton therapy group. Since the efficacy of both pituitary irradiation methods were similar, but proton therapy was associated with a higher incidence of serious side effects, the authors concluded that RT/CRT is the preferred treatment modality [200].

 

In a study from 2006, of 47 patients treated with fractionated proton therapy for both non-functioning and functioning pituitary adenomas reported tumor stabilization in 41 (87%) patients after a minimum 6-month follow-up; 1 patient developed temporal lobe necrosis, 3 developed new significant visual deficits, and 11 developed new hypopituitarism [201]. These are disappointing results suggesting considerably worse outcome both in terms of efficacy and toxicity than seen with photon irradiation.

 

A study of proton beam stereotactic radiosurgery in 22 patients with acromegaly reported normalization of GH in 59% after a median of 42 months. New pituitary deficiency was reported in 38% of patients, but no visual complications were reported [43]. The same group reported on the biochemical response in a larger population of secreting adenomas (74 ACTH-secreting, 50 GH-secreting, 9 PRL-secreting, 8 Nelson’s syndromes, 3 TSH-secreting) treated with the same technique. The study included 27 patients previously irradiated (14 pts) or treated with fractionated proton beam radiotherapy. At a median follow-up of 52 months, 42% of patients did not achieve endocrine control with patients with acromegaly having the longer time to biochemical response (49% at 5 years). The risk of developing hypopituitarism was 62% at 5 years and four patients (3%) experienced post treatment temporal lobe seizures, with associated temporal lobe changes on imaging (1 month to 9 years from proton treatment). [202]).

 

An evidence-based review of proton therapy from ASTRO’s emerging technology committee examined the evidence for proton therapy across multiple tumor sites and concluded that currently available evidence provides only limited indications for proton therapy [203]). The report recommended that robust prospective clinical trials be conducted to determine the appropriate clinical indications for proton therapy. In the present context, the available published reports of proton therapy for pituitary adenoma demonstrate disappointing efficacy and increased toxicity relative to much more readily available photon-based treatment. Also, in dosimetric comparisons, proton beam did not provide superior dose coverage advantage over photon radiation with comparable doses to OARs with both modalities [204]. Therefore, it seems difficult to justify proton therapy to the pituitary outside of the context of a clinical trial.

 

RE-IRRADIATION FOR RECURRENT DISEASE

 

Re-irradiation for progression of pituitary adenoma after previous pituitary radiotherapy is considered to be associated with a high risk of radiation-induced damage due to the presumed cumulative effect of radiation to the optic apparatus, the cranial nerves, and the normal brain tissues. However, re-irradiation using fractionated conventional or stereotactic techniques is feasible, with acceptable toxicity [53], provided that there has been at least a 3-4 year gap following primary radiotherapy treatment to doses below radiation tolerance of the CNS (which is the case for the conventional dose of 45Gy delivered at <1.8Gy per fraction). GK SRS has also been used to re-irradiate small recurrent lesions, particularly if they are not in close proximity to the optic apparatus [205].

 

While the current impression is that late toxicity following pituitary re-irradiation is uncommon, a high incidence of adverse side effects (13% radiation induced optic neuropathy and 13% of temporal lobe necrosis) was reported in a series of 15 patients re-irradiated with both single fraction and fractionated approaches (median time from previous RT 5.8 years) [206]. Nonetheless, there are at present insufficient long-term data to demonstrate the safety of pituitary re-irradiation for recurrent disease, although the use of high precision techniques and fractionation should theoretically reduce late toxicity.

 

With the lack of consensus, variations in the management of pituitary recurrences are discussed in MDT meetings and decisions vary based on expertise and scope of practicing physicians. For example, in a survey study for Canadian neurosurgeons and radiation oncologists, it was observed that physicians newer to practice had a greater tendency to advocate for stereotactic radiosurgery (SRS) or re-resection (54.5% and 36.4%, respectively), as compared to older surgeons who showed a higher propensity (22.2%) to advocate for observation. The presence of cavernous sinus extension encouraged radiation oncologists to offer earlier radiotherapy sooner (61.4%), compared to 40% of neurosurgeons [207].

 

OUTLOOK

 

The techniques of pituitary radiotherapy have gradually evolved over a number of decades with apparent choice between different technologies. All technologies share the aim of concentrating the radiation dose to the tumor with minimal dose to surrounding tissue and the irradiation is given in one, few or many fractions. There has been a lack of randomized comparative studies comparing the techniques to date. Systematic review of case series reported in the literature assessing the efficacy and toxicity provides a reasonably objective assessment of the techniques. While prospective randomized trials would provide the best objective comparative information, the beliefs of practitioners in particular treatment modalities, vested interests in technologies, and general difficulty of conducting studies in diseases with such long natural history make such comparative trials an unlikely prospect. This is compounded by the fact that new radiotherapy technologies continue to be introduced into clinical practice without the need for establishing efficacy as demanded for new drugs. Therefore, controversy will persist with regard to the appropriate and optimal methods for treating pituitary adenomas using radiation, and that all of the treatment modalities described here will continue in clinical use for the foreseeable future despite systematic reviews suggesting that some of the techniques may be less effective and potentially more toxic.

 

Conformal techniques of fractionated pituitary radiotherapy are standard practice, with many centers able to offer the additional accuracy of higher precision radiotherapy previously termed stereotactic but currently part of mainstream high-precision RT. Successful application of high-precision treatment is highly dependent on expertise in accurate target definition using modern MR imaging, on the precision of the immobilization system based on an exhaustive quality assurance program, and on infrastructure particularly in the form of expertise of staff in complex techniques of treatment planning and delivery. It seems most likely that it is the available expertise at all levels of staff in a treatment center that is the principal determinant of the success of pituitary radiotherapy rather than the choice of equipment and the precise treatment technique that is used.

 

SUMMARY

 

Fractionated radiotherapy is an effective treatment for pituitary adenomas, able to achieve excellent disease control and normalization of hormone levels. While the overall safety profile of this treatment modality is favorable, it is not devoid of side effects and it should only be employed when the risks from the disease itself are considered to outweigh the risks from the treatment. The balance of risks should take into account not only the early consequences of the disease and treatment, measured in terms of disease control and immediate morbidity, but also the long-term effects, particularly in terms of the influence of treatment on survival and quality of life, both of which are less well defined.

 

Residual pituitary adenomas, most of which have an indolent natural history, pose little threat to function, unless they lie close to the optic apparatus, or unless they destructively invade adjacent structures, which is an uncommon event. The risks of residual adenoma are therefore often minimal, and in the absence of progression or hormone hypersecretion, there is currently little justification for adjuvant radiation, whether in the form of fractionated or single fraction treatment. However, a policy of postoperative surveillance does require a program of close monitoring, usually in the form of annual MR imaging, and proceeding to timely irradiation if necessary, and certainly well before the need for further surgery. The aim of radiation treatment is to arrest tumor growth without the risks of re-operation.

 

For functioning tumors radiation treatment is generally offered to patients with persistent hormone elevation that is not decreasing at the expected rate following previous intervention of surgery and medical therapy. This usually means persistent hormone elevation in patients with acromegaly, Cushing’s disease, and other functioning adenomas, regardless of how far the actual hormone level is from normal, as the aim in most cases is to achieve normalization. In patients with acromegaly treated with somatostatin analogues, the expense and inconvenience of protracted systemic treatment also warrants early radiation treatment to allow for the withdrawal of medical treatment. The alternative is to continue medical management indefinitely without radiotherapy. It is not clear at present which policy is associated with better long-term survival and quality of life, and this should ideally be the subject of a prospective randomized trial.

 

Current clinical practice is therefore to offer treatment to patients with progressive non-functioning (or functioning) pituitary adenomas considered to be a threat to function, and to patients with functioning pituitary adenomas with persistent hypersecretion. Fractionated radiotherapy, as high-precision IMRT (previously considered as SCRT/fSRT), is the current standard of care for patients requiring radiation treatment for pituitary adenoma. Single fraction radiosurgery can be considered to treat small adenomas away from critical structures in view of the significant risk of radiation-induced damage carried by a high single dose of radiation. Long-term follow-up data are needed to fully evaluate the clinical efficacy of single fraction radiosurgery in comparison with fractionated radiotherapy.

 

ACKNOWLEDGEMENTS

 

MK and NF would like to thank Dr Francesca Solda, Dr Liam Welsh, Dr Thankamma Ajithkumar and Professor Michael Brada who authored previous versions of this review. MK is funded by the NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust and University College London.

 

REFERENCES

 

  1. Surawicz, T.S., et al., Descriptive epidemiology of primary brain and CNS tumors: results from the Central Brain Tumor Registry of the United States, 1990-1994. Neuro Oncol, 1999. 1(1): p. 14-25.
  2. Ezzat, S., et al., The prevalence of pituitary adenomas: a systematic review. Cancer, 2004. 101(3): p. 613-9.
  3. Beclere, J., The radiotherapeutic treatment of tumours of the hypophysis, gigantism and acromegaly. Archives of the Roentgen Ray, 1909. III(1): p. 8.
  4. Turner, H.E., et al., Audit of selected patients with nonfunctioning pituitary adenomas treated without irradiation - a follow-up study. Clin Endocrinol (Oxf), 1999. 51(3): p. 281-4.
  5. Gittoes, N.J., et al., Radiotherapy for non-function pituitary tumours. Clin Endocrinol (Oxf), 1998. 48(3): p. 331-7.
  6. Grigsby, P.W., et al., Prognostic factors and results of surgery and postoperative irradiation in the management of pituitary adenomas. Int J Radiat Oncol Biol Phys, 1989. 16(6): p. 1411-7.
  7. McCollough, W.M., et al., Long-term follow-up of radiotherapy for pituitary adenoma: the absence of late recurrence after greater than or equal to 4500 cGy. Int J Radiat Oncol Biol Phys, 1991. 21(3): p. 607-14.
  8. Brada, M., et al., The long-term efficacy of conservative surgery and radiotherapy in the control of pituitary adenomas. Clin Endocrinol (Oxf), 1993. 38(6): p. 571-8.
  9. Tsang, R.W., et al., Radiation therapy for pituitary adenoma: treatment outcome and prognostic factors. Int J Radiat Oncol Biol Phys, 1994. 30(3): p. 557-65.
  10. Zierhut, D., et al., External radiotherapy of pituitary adenomas. Int J Radiat Oncol Biol Phys, 1995. 33(2): p. 307-14.
  11. Rush, S. and P.R. Cooper, Symptom resolution, tumor control, and side effects following postoperative radiotherapy for pituitary macroadenomas. Int J Radiat Oncol Biol Phys, 1997. 37(5): p. 1031-4.
  12. Breen, P., et al., Radiotherapy for nonfunctional pituitary adenoma: analysis of long-term tumor control. J Neurosurg, 1998. 89(6): p. 933-8.
  13. Sasaki, R., et al., The efficacy of conventional radiation therapy in the management of pituitary adenoma. Int J Radiat Oncol Biol Phys, 2000. 47(5): p. 1337-45.
  14. Minniti, G., D.C. Gilbert, and M. Brada, Modern techniques for pituitary radiotherapy. Rev Endocr Metab Disord, 2009. 10(2): p. 135-44.
  15. Khoo, V.S., et al., Comparison of intensity-modulated tomotherapy with stereotactically guided conformal radiotherapy for brain tumors. Int J Radiat Oncol Biol Phys, 1999. 45(2): p. 415-25.
  16. Karger, C.P., et al., Three-dimensional accuracy and interfractional reproducibility of patient fixation and positioning using a stereotactic head mask system. Int J Radiat Oncol Biol Phys, 2001. 49(5): p. 1493-504.
  17. Ajithkumar, T. and M. Brada, Stereotactic linear accelerator radiotherapy for pituitary tumors. Treat Endocrinol, 2004. 3(4): p. 211-6.
  18. Leksell, L., The stereotaxic method and radiosurgery of the brain. Acta Chir Scand, 1951. 102(4): p. 316-9.
  19. Levy, R., A Short History of Stereotactic Neurosurgery. Cyber Museum of neurosurgery. 1999. Available from: http://www.neurosurgery.org/cybermuseum/stereotactichall/stereoarticle.html.
  20. Lasak, J.M. and J.P. Gorecki, The history of stereotactic radiosurgery and radiotherapy. Otolaryngol Clin North Am, 2009. 42(4): p. 593-9.
  21. Gill, S.S., et al., Relocatable frame for stereotactic external beam radiotherapy. Int J Radiat Oncol Biol Phys, 1991. 20(3): p. 599-603.
  22. Graham, J.D., et al., A non-invasive, relocatable stereotactic frame for fractionated radiotherapy and multiple imaging. Radiother Oncol, 1991. 21(1): p. 60-2.
  23. Adler, J.R., Jr., et al., The Cyberknife: a frameless robotic system for radiosurgery. Stereotact Funct Neurosurg, 1997. 69(1-4 Pt 2): p. 124-8.
  24. Steel, G.G., Basic Clinical Radiobiology. 3 ed. Vol. Arnold. 2002, London.
  25. Larson, D.A., J.C. Flickinger, and J.S. Loeffler, The radiobiology of radiosurgery. Int J Radiat Oncol Biol Phys, 1993. 25(3): p. 557-61.
  26. Park, C., et al., Universal survival curve and single fraction equivalent dose: useful tools in understanding potency of ablative radiotherapy. Int J Radiat Oncol Biol Phys, 2008. 70(3): p. 847-52.
  27. Lawrence, Y.R., et al., Radiation dose-volume effects in the brain. Int J Radiat Oncol Biol Phys, 2010. 76(3 Suppl): p. S20-7.
  28. Mayo, C., et al., Radiation dose-volume effects of optic nerves and chiasm. Int J Radiat Oncol Biol Phys, 2010. 76(3 Suppl): p. S28-35.
  29. Khattab, M.H., et al., Stereotactic Radiosurgery and Hypofractionated Stereotactic Radiotherapy for Nonfunctioning Pituitary Adenoma. J Neurol Surg B Skull Base, 2021. 82(Suppl 3): p. e51-e58.
  30. Rosenfelder, N.A., et al., Achieving the relocation accuracy of stereotactic frame-based cranial radiotherapy in a three-point thermoplastic shell. Clin Oncol (R Coll Radiol), 2013. 25(1): p. 66-73.
  31. Kumar, S., et al., Treatment accuracy of fractionated stereotactic radiotherapy. Radiother Oncol, 2005. 74(1): p. 53-9.
  32. Veselsky, T., et al., Pituitary adenoma treatment plan quality comparison between linear accelerator volumetric modulated arc therapy and Leksell Gamma Knife(R) radiosurgery. Med Dosim, 2021. 46(4): p. 440-448.
  33. Sayer, F.T., et al., Initial experience with the eXtend System: a relocatable frame system for multiple-session gamma knife radiosurgery. World Neurosurg, 2011. 75(5-6): p. 665-72.
  34. Zhao, K., et al., Fractionated Gamma Knife surgery for giant pituitary adenomas. Clin Neurol Neurosurg, 2016. 150: p. 139-142.
  35. Ikeda, H., H. Jokura, and T. Yoshimoto, Gamma knife radiosurgery for pituitary adenomas: usefulness of combined transsphenoidal and gamma knife radiosurgery for adenomas invading the cavernous sinus. Radiat Oncol Investig, 1998. 6(1): p. 26-34.
  36. Hayashi, M., et al., Gamma Knife robotic microradiosurgery of pituitary adenomas invading the cavernous sinus: treatment concept and results in 89 cases. J Neurooncol, 2010. 98(2): p. 185-94.
  37. Kim, E.H., et al., Postoperative Gamma Knife Radiosurgery for Cavernous Sinus-Invading Growth Hormone-Secreting Pituitary Adenomas. World Neurosurg, 2018. 110: p. e534-e545.
  38. Hayashi, M., et al., Gamma Knife Radiosurgery for Pituitary Adenomas Invading the Cavernous Sinus: Tokyo Women's Medical University Experience. Acta Neurochir Suppl, 2021. 128: p. 29-41.
  39. Lee, W.J., et al., Gamma Knife Radiosurgery as a Primary Treatment for Nonfunctioning Pituitary Adenoma Invading the Cavernous Sinus. Stereotact Funct Neurosurg, 2020. 98(6): p. 371-377.
  40. Sheehan, J.P., et al., Gamma Knife surgery for pituitary adenomas: factors related to radiological and endocrine outcomes. J Neurosurg, 2011. 114(2): p. 303-9.
  41. Grant, R.A., et al., Efficacy and safety of higher dose stereotactic radiosurgery for functional pituitary adenomas: a preliminary report. World Neurosurg, 2014. 82(1-2): p. 195-201.
  42. Kjellberg, R.N., et al., Proton-beam therapy in acromegaly. N Engl J Med, 1968. 278(13): p. 689-95.
  43. Petit, J.H., et al., Proton stereotactic radiosurgery in management of persistent acromegaly. Endocr Pract, 2007. 13(7): p. 726-34.
  44. Durante, M. and J.S. Loeffler, Charged particles in radiation oncology. Nat Rev Clin Oncol, 2010. 7(1): p. 37-43.
  45. Specialised Commissioning Team, N.E. B01/P/c Proton Beam Therapy for Cancer in Teenagers and Young Adults 2015; Available from: https://www.england.nhs.uk/commissioning/wp-content/uploads/sites/12/2015/10/b01-pc-prtn-bm-thrpy-teens-yng-oct15.pdf.
  46. Raverot, G., et al., European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol, 2018. 178(1): p. G1-G24.
  47. Giuffrida, G., et al., Peptide receptor radionuclide therapy for aggressive pituitary tumors: a monocentric experience. Endocr Connect, 2019. 8(5): p. 528-535.
  48. Maclean, J., et al., Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: variable clinical response in preliminary evaluation. Pituitary, 2014. 17(6): p. 530-8.
  49. Page, R.C., et al., An account of the quality of life of patients after treatment for non-functioning pituitary tumours. Clin Endocrinol (Oxf), 1997. 46(4): p. 401-6.
  50. van Beek, A.P., et al., Radiotherapy is not associated with reduced quality of life and cognitive function in patients treated for nonfunctioning pituitary adenoma. Int J Radiat Oncol Biol Phys, 2007. 68(4): p. 986-91.
  51. Yang, C.J., et al., Symptom distress and quality of life after stereotactic radiosurgery in patients with pituitary tumors: a questionnaire survey. PLoS One, 2014. 9(2): p. e88460.
  52. Jenkins, P.J., et al., Conventional pituitary irradiation is effective in lowering serum growth hormone and insulin-like growth factor-I in patients with acromegaly. J Clin Endocrinol Metab, 2006. 91(4): p. 1239-45.
  53. Brada, M. and P. Jankowska, Radiotherapy for pituitary adenomas. Endocrinol Metab Clin North Am, 2008. 37(1): p. 263-75, xi.
  54. Rowland, N.C. and M.K. Aghi, Radiation treatment strategies for acromegaly. Neurosurg Focus, 2010. 29(4): p. E12.
  55. Brada, M., T.V. Ajithkumar, and G. Minniti, Radiosurgery for pituitary adenomas. Clin Endocrinol (Oxf), 2004. 61(5): p. 531-43.
  56. Estrada, J., et al., The long-term outcome of pituitary irradiation after unsuccessful transsphenoidal surgery in Cushing's disease. N Engl J Med, 1997. 336(3): p. 172-7.
  57. Barrande, G., et al., Hormonal and metabolic effects of radiotherapy in acromegaly: long-term results in 128 patients followed in a single center. J Clin Endocrinol Metab, 2000. 85(10): p. 3779-85.
  58. Biermasz, N.R., H.V. Dulken, and F. Roelfsema, Postoperative radiotherapy in acromegaly is effective in reducing GH concentration to safe levels. Clin Endocrinol (Oxf), 2000. 53(3): p. 321-7.
  59. Epaminonda, P., et al., Efficacy of radiotherapy in normalizing serum IGF-I, acid-labile subunit (ALS) and IGFBP-3 levels in acromegaly. Clin Endocrinol (Oxf), 2001. 55(2): p. 183-9.
  60. Minniti, G., et al., The long-term efficacy of conventional radiotherapy in patients with GH-secreting pituitary adenomas. Clin Endocrinol (Oxf), 2005. 62(2): p. 210-6.
  61. Langsenlehner, T., et al., Long-term follow-up of patients with pituitary macroadenomas after postoperative radiation therapy: analysis of tumor control and functional outcome. Strahlenther Onkol, 2007. 183(5): p. 241-7.
  62. Minniti, G., et al., Long-term follow-up results of postoperative radiation therapy for Cushing's disease. J Neurooncol, 2007. 84(1): p. 79-84.
  63. Rim, C.H., et al., Radiotherapy for pituitary adenomas: long-term outcome and complications. Radiat Oncol J, 2011. 29(3): p. 156-63.
  64. Ajithkumar, T.B., M., Pituitary radiotherapy. Oxford Textbook of Endocrinology and Diabetes. 2011: Oxford Press University.
  65. Kim, M.Y., et al., Long-term outcomes of surgery and radiotherapy for secreting and non-secreting pituitary adenoma. Radiation Oncology Journal, 2016. 34(2): p. 121-127.
  66. Patt, H., et al., High-Precision Conformal Fractionated Radiotherapy Is Effective in Achieving Remission in Patients with Acromegaly after Failed Transsphenoidal Surgery. Endocr Pract, 2016. 22(2): p. 162-72.
  67. Hussein, Z., et al., Non-functioning pituitary macroadenoma following surgery: long-term outcomes and development of an optimal follow-up strategy. Front Surg, 2023. 10: p. 1129387.
  68. Arnardottir, S., et al., Long-term outcomes of patients with acromegaly: a report from the Swedish Pituitary Register. Eur J Endocrinol, 2022. 186(3): p. 329-339.
  69. Tsagarakis, S., et al., Megavoltage pituitary irradiation in the management of prolactinomas: long-term follow-up. Clin Endocrinol (Oxf), 1991. 34(5): p. 399-406.
  70. Johnston, D.G., et al., The long-term effects of megavoltage radiotherapy as sole or combined therapy for large prolactinomas: studies with high definition computerized tomography. Clin Endocrinol (Oxf), 1986. 24(6): p. 675-85.
  71. Mehta, A.E., F.I. Reyes, and C. Faiman, Primary radiotherapy of prolactinomas. Eight- to 15-year follow-up. Am J Med, 1987. 83(1): p. 49-58.
  72. Becker, G., et al., Radiation therapy in the multimodal treatment approach of pituitary adenoma. Strahlenther Onkol, 2002. 178(4): p. 173-86.
  73. Sherlock, M., et al., Mortality in patients with pituitary disease. Endocr Rev, 2010. 31(3): p. 301-42.
  74. Brada, M., et al., Cerebrovascular mortality in patients with pituitary adenoma. Clin Endocrinol (Oxf), 2002. 57(6): p. 713-7.
  75. Brada, M., et al., The incidence of cerebrovascular accidents in patients with pituitary adenoma. Int J Radiat Oncol Biol Phys, 1999. 45(3): p. 693-8.
  76. Tomlinson, J.W., et al., Association between premature mortality and hypopituitarism. West Midlands Prospective Hypopituitary Study Group. Lancet, 2001. 357(9254): p. 425-31.
  77. Erfurth, E.M., et al., Risk factors for cerebrovascular deaths in patients operated and irradiated for pituitary tumors. J Clin Endocrinol Metab, 2002. 87(11): p. 4892-9.
  78. Fajardo, L.F., The pathology of ionizing radiation as defined by morphologic patterns. Acta Oncol, 2005. 44(1): p. 13-22.
  79. Bliss, P., G.R. Kerr, and A. Gregor, Incidence of second brain tumours after pituitary irradiation in Edinburgh 1962-1990. Clin Oncol (R Coll Radiol), 1994. 6(6): p. 361-3.
  80. Tsang, R.W., et al., Glioma arising after radiation therapy for pituitary adenoma. A report of four patients and estimation of risk. Cancer, 1993. 72(7): p. 2227-33.
  81. Brada, M., et al., Risk of second brain tumour after conservative surgery and radiotherapy for pituitary adenoma. BMJ, 1992. 304(6838): p. 1343-6.
  82. Hamblin, R., et al., Risk of second brain tumour after radiotherapy for pituitary adenoma or craniopharyngioma: a retrospective, multicentre, cohort study of 3679 patients with long-term imaging surveillance. Lancet Diabetes Endocrinol, 2022. 10(8): p. 581-588.
  83. Peace, K.A., et al., Cognitive dysfunction in patients with pituitary tumour who have been treated with transfrontal or transsphenoidal surgery or medication. Clin Endocrinol (Oxf), 1998. 49(3): p. 391-6.
  84. Guinan, E.M., et al., Cognitive effects of pituitary tumours and their treatments: two case studies and an investigation of 90 patients. J Neurol Neurosurg Psychiatry, 1998. 65(6): p. 870-6.
  85. McCord, M.W., et al., Radiotherapy for pituitary adenoma: long-term outcome and sequelae. Int J Radiat Oncol Biol Phys, 1997. 39(2): p. 437-44.
  86. Brummelman, P., et al., Cognitive functioning in patients treated for nonfunctioning pituitary macroadenoma and the effects of pituitary radiotherapy. Clin Endocrinol (Oxf), 2011. 74(4): p. 481-7.
  87. Brummelman, P., et al., Cognitive performance after postoperative pituitary radiotherapy: a dosimetric study of the hippocampus and the prefrontal cortex. Eur J Endocrinol, 2012. 166(2): p. 171-9.
  88. Coke, C., et al., Multiple fractionated stereotactic radiotherapy of residual pituitary macroadenomas: initial experience. Stereotact Funct Neurosurg, 1997. 69(1-4 Pt 2): p. 183-90.
  89. Mitsumori, M., et al., Initial clinical results of LINAC-based stereotactic radiosurgery and stereotactic radiotherapy for pituitary adenomas. Int J Radiat Oncol Biol Phys, 1998. 42(3): p. 573-80.
  90. Milker-Zabel, S., et al., Fractionated stereotactically guided radiotherapy and radiosurgery in the treatment of functional and nonfunctional adenomas of the pituitary gland. Int J Radiat Oncol Biol Phys, 2001. 50(5): p. 1279-86.
  91. Paek, S.H., et al., Integration of surgery with fractionated stereotactic radiotherapy for treatment of nonfunctioning pituitary macroadenomas. Int J Radiat Oncol Biol Phys, 2005. 61(3): p. 795-808.
  92. Colin, P., et al., Treatment of pituitary adenomas by fractionated stereotactic radiotherapy: a prospective study of 110 patients. Int J Radiat Oncol Biol Phys, 2005. 62(2): p. 333-41.
  93. Minniti, G., et al., Fractionated stereotactic conformal radiotherapy for secreting and nonsecreting pituitary adenomas. Clin Endocrinol (Oxf), 2006. 64(5): p. 542-8.
  94. Selch, M.T., et al., Stereotactic radiotherapy for the treatment of pituitary adenomas. Minim Invasive Neurosurg, 2006. 49(3): p. 150-5.
  95. Kong, D.S., et al., The efficacy of fractionated radiotherapy and stereotactic radiosurgery for pituitary adenomas: long-term results of 125 consecutive patients treated in a single institution. Cancer, 2007. 110(4): p. 854-60.
  96. Snead, F.E., et al., Long-term outcomes of radiotherapy for pituitary adenomas. Int J Radiat Oncol Biol Phys, 2008. 71(4): p. 994-8.
  97. Roug, S., et al., Fractionated stereotactic radiotherapy in patients with acromegaly: an interim single-centre audit. Eur J Endocrinol, 2010. 162(4): p. 685-94.
  98. Schalin-Jantti, C., et al., Outcome of fractionated stereotactic radiotherapy in patients with pituitary adenomas resistant to conventional treatments: a 5.25-year follow-up study. Clin Endocrinol (Oxf), 2010. 73(1): p. 72-7.
  99. Weber, D.C., et al., Adjuvant or radical fractionated stereotactic radiotherapy for patients with pituitary functional and nonfunctional macroadenoma. Radiat Oncol, 2011. 6: p. 169.
  100. Wilson, P.J., et al., A single centre's experience of stereotactic radiosurgery and radiotherapy for non-functioning pituitary adenomas with the Linear Accelerator (Linac). J Clin Neurosci, 2012. 19(3): p. 370-4.
  101. Kim, J.O., et al., Long-term outcomes of fractionated stereotactic radiation therapy for pituitary adenomas at the BC Cancer Agency. Int J Radiat Oncol Biol Phys, 2013. 87(3): p. 528-33.
  102. Kopp, C., et al., Fractionated stereotactic radiotherapy in the treatment of pituitary adenomas. Strahlenther Onkol, 2013. 189(11): p. 932-7.
  103. Minniti, G., et al., Fractionated stereotactic radiotherapy for large and invasive non-functioning pituitary adenomas: long-term clinical outcomes and volumetric MRI assessment of tumor response. Eur J Endocrinol, 2015. 172(4): p. 433-41.
  104. Diallo, A.M., et al., Long-term results of fractionated stereotactic radiotherapy as third-line treatment in acromegaly. Endocrine, 2015. 50(3): p. 741-8.
  105. Barber, S.M., B.S. Teh, and D.S. Baskin, Fractionated Stereotactic Radiotherapy for Pituitary Adenomas: Single-Center Experience in 75 Consecutive Patients. Neurosurgery, 2016. 79(3): p. 406-17.
  106. Liao, H.I., et al., Fractionated stereotactic radiosurgery using the Novalis system for the management of pituitary adenomas close to the optic apparatus. J Clin Neurosci, 2014. 21(1): p. 111-5.
  107. Puataweepong, P., et al., Outcomes for Pituitary Adenoma Patients Treated with Linac- Based Stereotactic Radiosurgery and Radiotherapy: a Long Term Experience in Thailand. Asian Pac J Cancer Prev, 2015. 16(13): p. 5279-84.
  108. Lian, X., et al., Intensity-modulated Radiotherapy for Pituitary Somatotroph Adenomas. J Clin Endocrinol Metab, 2020. 105(12).
  109. Martinez, R., et al., Pituitary tumors and gamma knife surgery. Clinical experience with more than two years of follow-up. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 110-8.
  110. Pan, L., et al., Pituitary adenomas: the effect of gamma knife radiosurgery on tumor growth and endocrinopathies. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 119-26.
  111. Mokry, M., et al., A six year experience with the postoperative radiosurgical management of pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 88-100.
  112. Sheehan, J.P., et al., Radiosurgery for residual or recurrent nonfunctioning pituitary adenoma. J Neurosurg, 2002. 97(5 Suppl): p. 408-14.
  113. Wowra, B. and W. Stummer, Efficacy of gamma knife radiosurgery for nonfunctioning pituitary adenomas: a quantitative follow up with magnetic resonance imaging-based volumetric analysis. J Neurosurg, 2002. 97(5 Suppl): p. 429-32.
  114. Petrovich, Z., et al., Gamma knife radiosurgery for pituitary adenoma: early results. Neurosurgery, 2003. 53(1): p. 51-9; discussion 59-61.
  115. Pollock, B.E. and P.C. Carpenter, Stereotactic radiosurgery as an alternative to fractionated radiotherapy for patients with recurrent or residual nonfunctioning pituitary adenomas. Neurosurgery, 2003. 53(5): p. 1086-91; discussion 1091-4.
  116. Losa, M., et al., Gamma knife surgery for treatment of residual nonfunctioning pituitary adenomas after surgical debulking. J Neurosurg, 2004. 100(3): p. 438-44.
  117. Iwai, Y., K. Yamanaka, and K. Yoshioka, Radiosurgery for nonfunctioning pituitary adenomas. Neurosurgery, 2005. 56(4): p. 699-705; discussion 699-705.
  118. Mingione, V., et al., Gamma surgery in the treatment of nonsecretory pituitary macroadenoma. J Neurosurg, 2006. 104(6): p. 876-83.
  119. Liscak, R., et al., Gamma knife radiosurgery for endocrine-inactive pituitary adenomas. Acta Neurochir (Wien), 2007. 149(10): p. 999-1006; discussion 1006.
  120. Pollock, B.E., et al., Pituitary tumor type affects the chance of biochemical remission after radiosurgery of hormone-secreting pituitary adenomas. Neurosurgery, 2008. 62(6): p. 1271-6; discussion 1276-8.
  121. Kobayashi, T., Long-term results of stereotactic gamma knife radiosurgery for pituitary adenomas. Specific strategies for different types of adenoma. Prog Neurol Surg, 2009. 22: p. 77-95.
  122. Gopalan, R., et al., Long-term outcomes after Gamma Knife radiosurgery for patients with a nonfunctioning pituitary adenoma. Neurosurgery, 2011. 69(2): p. 284-93.
  123. Runge, M.J., et al., LINAC-radiosurgery for nonsecreting pituitary adenomas. Long-term results. Strahlenther Onkol, 2012. 188(4): p. 319-25.
  124. Park, K.J., et al., Long-term outcomes after gamma knife stereotactic radiosurgery for nonfunctional pituitary adenomas. Neurosurgery, 2011. 69(6): p. 1188-99.
  125. Starke, R.M., et al., Gamma Knife surgery for patients with nonfunctioning pituitary macroadenomas: predictors of tumor control, neurological deficits, and hypopituitarism. J Neurosurg, 2012. 117(1): p. 129-35.
  126. El-Shehaby, A.M., et al., Low-dose Gamma Knife surgery for nonfunctioning pituitary adenomas. J Neurosurg, 2012. 117 Suppl: p. 84-8.
  127. Sheehan, J.P., et al., Gamma Knife radiosurgery for the management of nonfunctioning pituitary adenomas: a multicenter study. J Neurosurg, 2013. 119(2): p. 446-56.
  128. Lee, C.C., et al., Initial Gamma Knife radiosurgery for nonfunctioning pituitary adenomas. J Neurosurg, 2014. 120(3): p. 647-54.
  129. Xu, Z., et al., Silent corticotroph adenomas after stereotactic radiosurgery: a case-control study. Int J Radiat Oncol Biol Phys, 2014. 90(4): p. 903-10.
  130. Hasegawa, T., et al., Stereotactic Radiosurgery as the Initial Treatment for Patients with Nonfunctioning Pituitary Adenomas. World Neurosurg, 2015. 83(6): p. 1173-9.
  131. Graffeo, C.S., et al., Hypopituitarism After Single-Fraction Pituitary Adenoma Radiosurgery: Dosimetric Analysis Based on Patients Treated Using Contemporary Techniques. Int J Radiat Oncol Biol Phys, 2018. 101(3): p. 618-623.
  132. Oh, J.W., et al., Hypopituitarism after Gamma Knife surgery for postoperative nonfunctioning pituitary adenoma. J Neurosurg, 2018. 129(Suppl1): p. 47-54.
  133. Cordeiro, D., et al., Hypopituitarism after Gamma Knife radiosurgery for pituitary adenomas: a multicenter, international study. J Neurosurg, 2018.
  134. Narayan, V., et al., Long-Term Outcome of Nonfunctioning and Hormonal Active Pituitary Adenoma After Gamma Knife Radiosurgery. World Neurosurg, 2018. 114: p. e824-e832.
  135. Slavinsky, P., et al., Gamma knife radiosurgery in pituitary adenomas. A single-center experience. Medicina (B Aires), 2022. 82(1): p. 111-116.
  136. Maldar, A.N., et al., Long-term hormonal and imaging outcomes of adjunctive gamma knife radiosurgery in non-functioning pituitary adenomas: a single center experience. J Neurooncol, 2022. 158(3): p. 423-433.
  137. Thoren, M., et al., Stereotactic radiosurgery with the cobalt-60 gamma unit in the treatment of growth hormone-producing pituitary tumors. Neurosurgery, 1991. 29(5): p. 663-8.
  138. Morange-Ramos, I., et al., Short-term endocrinological results after gamma knife surgery of pituitary adenomas. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 127-38.
  139. Lim, Y.L., et al., Four years' experiences in the treatment of pituitary adenomas with gamma knife radiosurgery. Stereotact Funct Neurosurg, 1998. 70 Suppl 1: p. 95-109.
  140. Kim, M.S., S.I. Lee, and J.H. Sim, Gamma Knife radiosurgery for functioning pituitary microadenoma. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 119-24.
  141. Landolt, A.M., et al., Stereotactic radiosurgery for recurrent surgically treated acromegaly: comparison with fractionated radiotherapy. J Neurosurg, 1998. 88(6): p. 1002-8.
  142. Hayashi, M., et al., Gamma Knife radiosurgery for pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 111-8.
  143. Inoue, H.K., et al., Pituitary adenomas treated by microsurgery with or without Gamma Knife surgery: experience in 122 cases. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 125-31.
  144. Zhang, N., et al., Radiosurgery for growth hormone-producing pituitary adenomas. J Neurosurg, 2000. 93 Suppl 3: p. 6-9.
  145. Izawa, M., et al., Gamma knife radiosurgery for pituitary adenomas. J Neurosurg, 2000. 93 Suppl 3: p. 19-22.
  146. Pollock, B.E., et al., Results of stereotactic radiosurgery in patients with hormone-producing pituitary adenomas: factors associated with endocrine normalization. J Neurosurg, 2002. 97(3): p. 525-30.
  147. Attanasio, R., et al., Gamma-knife radiosurgery in acromegaly: a 4-year follow-up study. J Clin Endocrinol Metab, 2003. 88(7): p. 3105-12.
  148. Choi, J.Y., et al., Radiological and hormonal responses of functioning pituitary adenomas after gamma knife radiosurgery. Yonsei Med J, 2003. 44(4): p. 602-7.
  149. Jane, J.A., Jr. and E.R. Laws, Jr., The management of non-functioning pituitary adenomas. Neurol India, 2003. 51(4): p. 461-5.
  150. Castinetti, F., et al., Outcome of gamma knife radiosurgery in 82 patients with acromegaly: correlation with initial hypersecretion. J Clin Endocrinol Metab, 2005. 90(8): p. 4483-8.
  151. Gutt, B., et al., Gamma-knife surgery is effective in normalising plasma insulin-like growth factor I in patients with acromegaly. Exp Clin Endocrinol Diabetes, 2005. 113(4): p. 219-24.
  152. Kobayashi, T., et al., Long-term results of gamma knife surgery for growth hormone-producing pituitary adenoma: is the disease difficult to cure? J Neurosurg, 2005. 102 Suppl: p. 119-23.
  153. Jezkova, J., et al., Gamma knife radiosurgery for acromegaly--long-term experience. Clin Endocrinol (Oxf), 2006. 64(5): p. 588-95.
  154. Pollock, B.E., et al., Radiosurgery of growth hormone-producing pituitary adenomas: factors associated with biochemical remission. J Neurosurg, 2007. 106(5): p. 833-8.
  155. Jagannathan, J., et al., Stereotactic radiosurgery for pituitary adenomas: a comprehensive review of indications, techniques and long-term results using the Gamma Knife. J Neurooncol, 2009. 92(3): p. 345-56.
  156. Wan, H., O. Chihiro, and S. Yuan, MASEP gamma knife radiosurgery for secretory pituitary adenomas: experience in 347 consecutive cases. J Exp Clin Cancer Res, 2009. 28: p. 36.
  157. Castinetti, F., et al., Long-term results of stereotactic radiosurgery in secretory pituitary adenomas. J Clin Endocrinol Metab, 2009. 94(9): p. 3400-7.
  158. Iwai, Y., et al., Gamma knife radiosurgery for growth hormone-producing adenomas. J Clin Neurosci, 2010. 17(3): p. 299-304.
  159. Erdur, F.M., et al., Gammaknife radiosurgery in patients with acromegaly. J Clin Neurosci, 2011. 18(12): p. 1616-20.
  160. Franzin, A., et al., Results of gamma knife radiosurgery in acromegaly. Int J Endocrinol, 2012. 2012: p. 342034.
  161. Liu, X., et al., Gamma knife radiosurgery for clinically persistent acromegaly. J Neurooncol, 2012. 109(1): p. 71-9.
  162. Zeiler, F.A., et al., Gamma knife in the treatment of pituitary adenomas: results of a single center. Can J Neurol Sci, 2013. 40(4): p. 546-52.
  163. Lee, C.C., et al., Stereotactic radiosurgery for acromegaly. J Clin Endocrinol Metab, 2014. 99(4): p. 1273-81.
  164. Jagannathan, J., et al., Gamma knife radiosurgery for acromegaly: outcomes after failed transsphenoidal surgery. Neurosurgery, 2008. 62(6): p. 1262-9; discussion 1269-70.
  165. Kim, S.H., et al., Gamma Knife radiosurgery for functioning pituitary adenomas. Stereotact Funct Neurosurg, 1999. 72 Suppl 1: p. 101-10.
  166. Gupta, A., et al., Upfront Gamma Knife radiosurgery for Cushing's disease and acromegaly: a multicenter, international study. J Neurosurg, 2018. 131(2): p. 532-538.
  167. Ding, D., et al., Stereotactic Radiosurgery for Acromegaly: An International Multicenter Retrospective Cohort Study. Neurosurgery, 2019. 84(3): p. 717-725.
  168. Degerblad, M., et al., Long-term results of stereotactic radiosurgery to the pituitary gland in Cushing's disease. Acta Endocrinol (Copenh), 1986. 112(3): p. 310-4.
  169. Ganz, J.C., E.O. Backlund, and F.A. Thorsen, The effects of Gamma Knife surgery of pituitary adenomas on tumor growth and endocrinopathies. Stereotact Funct Neurosurg, 1993. 61 Suppl 1: p. 30-7.
  170. Seo, Y., et al., Gamma knife surgery for Cushing's disease. Surg Neurol, 1995. 43(2): p. 170-5; discussion 175-6.
  171. Sheehan, J.M., et al., Radiosurgery for Cushing's disease after failed transsphenoidal surgery. J Neurosurg, 2000. 93(5): p. 738-42.
  172. Hoybye, C., et al., Adrenocorticotropic hormone-producing pituitary tumors: 12- to 22-year follow-up after treatment with stereotactic radiosurgery. Neurosurgery, 2001. 49(2): p. 284-91; discussion 291-2.
  173. Kobayashi, T., Y. Kida, and Y. Mori, Gamma knife radiosurgery in the treatment of Cushing disease: long-term results. J Neurosurg, 2002. 97(5 Suppl): p. 422-8.
  174. Devin, J.K., et al., The efficacy of linear accelerator radiosurgery in the management of patients with Cushing's disease. Stereotact Funct Neurosurg, 2004. 82(5-6): p. 254-62.
  175. Castinetti, F., et al., Gamma knife radiosurgery is a successful adjunctive treatment in Cushing's disease. Eur J Endocrinol, 2007. 156(1): p. 91-8.
  176. Marek, J., et al., Gamma knife radiosurgery for Cushing's disease and Nelson's syndrome. Pituitary, 2015. 18(3): p. 376-84.
  177. Sheehan, J.P., et al., Results of gamma knife surgery for Cushing's disease. J Neurosurg, 2013. 119(6): p. 1486-92.
  178. Sicignano, G., et al., Dosimetric factors associated with pituitary function after Gamma Knife Surgery (GKS) of pituitary adenomas. Radiother Oncol, 2012. 104(1): p. 119-24.
  179. Jagannathan, J., et al., Gamma Knife surgery for Cushing's disease. J Neurosurg, 2007. 106(6): p. 980-7.
  180. Knappe, U.J., et al., Fractionated radiotherapy and radiosurgery in acromegaly: analysis of 352 patients from the German Acromegaly Registry. Eur J Endocrinol, 2020. 182(3): p. 275-284.
  181. Landolt, A.M. and N. Lomax, Gamma knife radiosurgery for prolactinomas. J Neurosurg, 2000. 93 Suppl 3: p. 14-8.
  182. Pan, L., et al., Gamma knife radiosurgery as a primary treatment for prolactinomas. J Neurosurg, 2000. 93 Suppl 3: p. 10-3.
  183. Pouratian, N., et al., Gamma knife radiosurgery for medically and surgically refractory prolactinomas. Neurosurgery, 2006. 59(2): p. 255-66; discussion 255-66.
  184. Jezkova, J., et al., Use of the Leksell gamma knife in the treatment of prolactinoma patients. Clin Endocrinol (Oxf), 2009. 70(5): p. 732-41.
  185. Liu, X., et al., Gamma knife stereotactic radiosurgery for drug resistant or intolerant invasive prolactinomas. Pituitary, 2013. 16(1): p. 68-75.
  186. Cohen-Inbar, O., et al., Gamma Knife radiosurgery for medically and surgically refractory prolactinomas: long-term results. Pituitary, 2015. 18(6): p. 820-30.
  187. Jezkova, J., et al., Role of gamma knife radiosurgery in the treatment of prolactinomas. Pituitary, 2019. 22(4): p. 411-421.
  188. Voges, J., et al., Linear accelerator radiosurgery for pituitary macroadenomas: a 7-year follow-up study. Cancer, 2006. 107(6): p. 1355-64.
  189. Palmisciano, P., et al., Endocrine disorders after primary gamma knife radiosurgery for pituitary adenomas: A systematic review and meta-analysis. Pituitary, 2022. 25(3): p. 404-419.
  190. Kajiwara, K., et al., Image-guided stereotactic radiosurgery with the CyberKnife for pituitary adenomas. Minim Invasive Neurosurg, 2005. 48(2): p. 91-6.
  191. Adler, J.R., Jr., et al., Visual field preservation after multisession cyberknife radiosurgery for perioptic lesions. Neurosurgery, 2006. 59(2): p. 244-54; discussion 244-54.
  192. Roberts, B.K., et al., Efficacy and safety of CyberKnife radiosurgery for acromegaly. Pituitary, 2007. 10(1): p. 17.
  193. Killory, B.D., et al., Hypofractionated CyberKnife radiosurgery for perichiasmatic pituitary adenomas: early results. Neurosurgery, 2009. 64(2 Suppl): p. A19-25.
  194. Cho, C.B., et al., Stereotactic Radiosurgery with the CyberKnife for Pituitary Adenomas. J Korean Neurosurg Soc, 2009. 45(3): p. 157-63.
  195. Iwata, H., et al., Hypofractionated stereotactic radiotherapy with CyberKnife for nonfunctioning pituitary adenoma: high local control with low toxicity. Neuro Oncol, 2011. 13(8): p. 916-22.
  196. Puataweepong, P., et al., The Clinical Outcome of Hypofractionated Stereotactic Radiotherapy With CyberKnife Robotic Radiosurgery for Perioptic Pituitary Adenoma. Technol Cancer Res Treat, 2016. 15(6): p. NP10-NP15.
  197. Iwata, H., et al., Long-term results of hypofractionated stereotactic radiotherapy with CyberKnife for growth hormone-secreting pituitary adenoma: evaluation by the Cortina consensus. J Neurooncol, 2016. 128(2): p. 267-75.
  198. Plitt, A.R., et al., Fractionated CyberKnife Stereotactic Radiotherapy for Perioptic Pituitary Adenomas. World Neurosurg, 2019. 126: p. e1359-e1364.
  199. Romero-Gameros, C.A., et al., Efficiency and Safety of CyberKnife Robotic Radiosurgery in the Multimodal Management of Patients with Acromegaly. Cancers (Basel), 2023. 15(5).
  200. Ludecke, D.K., B.S. Lutz, and G. Niedworok, The choice of treatment after incomplete adenomectomy in acromegaly: proton--versus high voltage radiation. Acta Neurochir (Wien), 1989. 96(1-2): p. 32-8.
  201. Ronson, B.B., et al., Fractionated proton beam irradiation of pituitary adenomas. Int J Radiat Oncol Biol Phys, 2006. 64(2): p. 425-34.
  202. Wattson, D.A., et al., Outcomes of proton therapy for patients with functional pituitary adenomas. Int J Radiat Oncol Biol Phys, 2014. 90(3): p. 532-9.
  203. Allen, A.M., et al., An evidence based review of proton beam therapy: the report of ASTRO's emerging technology committee. Radiother Oncol, 2012. 103(1): p. 8-11.
  204. Sud, S., et al., Dosimetric Comparison of Proton Versus Photon Radiosurgery for Treatment of Pituitary Adenoma. Adv Radiat Oncol, 2021. 6(6): p. 100806.
  205. Edwards, A.A., F.M. Swords, and P.N. Plowman, Focal radiation therapy for patients with persistent/recurrent pituitary adenoma, despite previous radiotherapy. Pituitary, 2009. 12(1): p. 30-4.
  206. Verma, J., et al., Feasibility and outcome of re-irradiation in the treatment of multiply recurrent pituitary adenomas. Pituitary, 2014. 17(6): p. 539-45.
  207. Kasper, G., et al., Practice patterns in the management of recurrent and residual non-functioning pituitary adenomas: Results from a Canada-wide survey. eNeurologicalSci, 2021. 22: p. 100317.

Diabetic Striatopathy

ABSTRACT

 

Acute onset de novo movement disorders are increasingly being reported in the settings of hyperglycemia, particularly from Asian countries. Although hemichorea-hemiballism is the most common and classically described movement semiology in association with hyperglycemia, various other hyperkinetic (choreoathetosis, dystonia, tremors, akathisia, restless leg syndrome etc.) and hypokinetic (parkinsonism) movement disorders are recognized. Diabetic striatopathy (DS) is defined as the disease phenomenon characterized by either choreo-ballistic movement or suggestive signature changes in striatum on imaging or presence of both. DS is generally considered as the complication of long-standing, poorly controlled non-ketotic hyperglycemia with acute hyperglycemic surge, though it can also be the first presentation of previously undiagnosed diabetes. Thus, it is recommended to test for capillary blood glucose in every patient presenting with de novo acute onset movement disorders of any semiology irrespective of past history of diabetes. It is important to recognize that normal brain imaging does not exclude the diagnosis of DS (clinically isolated DS) because nearly 50% cases may not have any characteristic neuroradiological stigmata. There is also high prevalence of clinical-neuroradiological discordance in DS cases. Thus, while managing such patients’ priority should be imparted on bedside identification of the movement semiology accurately and aggressive treatment of hyperglycemia rather than ordering expensive neuroradiological investigation. Generally diabetic movement disorder carries excellent prognosis. The majority of cases rapidly resolves with insulin therapy alone with or without use of adjunctive neuroleptics.      

 

INTRODUCTION

 

Although a myriad of neurological complications resulting from chronic micro- and macroangiopathy and acute metabolic perturbations in diabetes mellitus (DM) had been well documented, structured studies on acute-onset movement disorders among patients with DM were surprisingly left out until recently (1,2). Movement disorders can manifest either as the first manifestation of undiagnosed DM or in later advanced stages of the disease (3-7). Genesis of these abnormal movements can directly be attributed to hyperglycemia or hypoglycemia, and may result from diabetic complications such as vasculopathy and neuropathy (2,8). Moreover, there are syndromes or conditions which can present as movement disorders alongside DM (8,9). Aggressive glycemic control is known to alleviate abnormal movements in most of the cases (1,2,8). Among all the movement semiologies discussed in literature, hemichorea-hemiballism is most frequently reported (1,2). Diabetic striatopathy (DS) is an umbrella term referring to a hyperglycemic condition associated with both or either one of the two following conditions: (1) acute onset chorea-ballism; (2) striatal hyperdensity on computed tomography (CT) or striatal hyperintensity on T1-weighted magnetic resonance imaging (MRI) (1,2,10). We herein briefly summarize the movement disorders in DM keeping DS at the center of discussion. Epidemiological and clinical spectrum, pathophysiology, neuroradiological conundrums, and available treatment are discussed. We also have tried to shed light upon the knowledge gaps in understanding of this particular disease that need to be addressed.

 

EPIDEMIOLOGY- MAGNITUDE OF THE PROBLEM

 

At present there is no prospective epidemiological study to assess the incidence or prevalence data available regarding movement disorders in diabetes. Few retrospective analyses with weak study methodology showed the prevalence of DS was in order of 1% or even less (11-13). On the other hand, a prospective study by Dubey et al revealed that 17.4% patients were diabetic among 552 patients presented with acute onset movement disorders and its mimics (including epilepsia partialis continua in a movement disorder clinic (1).

 

A systematic review of 176 patients observed that the lion share of DS cases was reported from Asian countries (2). Multiple factors like easy accessibility to healthcare, poor compliance to drugs, ethnicity, or genetic susceptibility might play roles, but it definitely requires more exploration. However, a study by Shafran et al revealed that DS was actually underdiagnosed in western populations leading to its underreporting (11).

 

Acute onset movement disorders in diabetes had been reported in a wide range of age groups ranging from first to ninth decade (2). The mean age of the patients was generally sixth to seventh decade observed in different case series or systematic reviews (2,14-19). Two studies from India reported a relatively younger mean age (fifth decade) of presentation (1,20). Chen et al analyzing only the cases of hemichorea-hemiballism with ketotic hyperglycemia also found a median age of 54 years (21).

 

Across different studies over the years, notably, a woman preponderance (nearly 2 times in most of the studies) of hyperglycemic hemichorea-hemiballism movements had been observed (2,14-21). The exact reason for this female predominance or the role of biological sex on hyperglycemia-induced acute movement disorders needs further study. Some have postulated that increased dopaminergic receptor sensitivity secondary to estrogen deficiency in the striatum among postmenopausal women might make them susceptible to hyperkinetic movement disorders (17,21). In contrast with this global scenario, the authors’ largest clinical series from India revealed a slight male predominance (52.5%) which needs further confirmation by replication in other independent studies (1).

 

CLINICAL PRESENTATION- SPECTRUM OF MOVEMENT DISORDERS IN DIABETES

 

Among different movement semiologies described among diabetics (table-1), hemichorea-hemiballism is the most common and classically described (1-8). See video 1-6 for different movement disorders associated with hyperglycemia.

 

VIDEOS

 

Table 1. Different Movement Semiologies Observed Among Patients with Diabetes

Choreic and ballistic movements

Non-choreoballistic movements

·       Choreoballism- hemi / mono / generalized

·       Pure chorea- hemi / mono / generalized

·       Pure ballism- hemi / mono / generalized

·       Choreoathetosis

 

·       Tremors

·       Hemifacial spasm

·       Parkinsonism

·       Myoclonus- focal, action, diaphragmatic, opsoclonus-myoclonus

·       Dystonia

·       Restless leg syndrome

·       Ataxia

·       Dyskinesia- Paroxysmal kinesigenic dyskinesia, paroxysmal non-kinesigenic dyskinesia, paroxysmal exertional dyskinesia

 

Dubey et al (1) in their largest clinical series showed that non-choreic, non-ballistic movements were present among 30.5% of 59 cases. Therefore, an immediate capillary blood glucose (CBG) measurement in all patients with any sort of acute onset movement disorders is of pivotal importance before ordering other costly and time-consuming investigations. Bilateral clinical involvement was identified in 37.2% of all patients and was significantly more common in non-choreoballistic movement disorders than choreoballism (1). In an analysis by Chua et al bilaterality was documented in 9.7% DS cases (2), whereas bilaterality was even more frequently (19.5%) observed in the series described by Dubey et al (1). The latter was the only study which systematically assessed both the hyperglycemia-associated choreoballism and non-choreoballistic movement disorders. It observed no statistically significant differences regarding demographic or clinical variables between these two types of movement disorders except bilaterality and delay in diagnosis (more frequent in non-choreoballism than choreoballism) (1).

 

Many a times seizures can mimic hyperkinetic movement disorders or sometimes both may coexist (3). Most commonly epilepsia partialis continua mimics a movement disorder. It is not conventionally categorized as a movement disorder; but is rather a type of simple focal motor status epilepticus with frequent repetitive muscle jerks, usually arrhythmic, that continues over prolonged periods. Moreover, the epilepsia partialis continua patients have electroencephalographic changes. The differentials to be considered are stroke, associated opposite hemispheric structural defect/s, and space-occupying lesions. Non-ketotic hyperglycemia is a well-known cause of reversible epilepsia partialis continua (22,23).

 

CORRELATION WITH MARKERS OF GLYCEMIA AND DIABETIC COMPLICATIONS

 

Movement disorders have been described in different types of DM, including type 1, type 2 and type 3c diabetes (1,2,5). DS is generally a complication of long-standing DM with poorly controlled glycemic status flared up by an acute hyperglycemic surge in a non-ketotic milieu. In the cohort of Dubey et al the mean duration of DM was 9.8 years and movement disorders were the presenting manifestation of previously undiagnosed DM in three cases (5.1%) (1). Patient-level meta-analysis of previously published cases has found a higher number (17%) of DS cases having previously undiagnosed DM (2). This discrepancy could be due to lack of screening or publication bias. Nonetheless, it is recommended to measure blood glucose levels at presentation among all patients with acute onset movement disorders irrespective of their past glycemic status. Importantly, the majority of the patients with DS bears the stigmata of other chronic microvascular diabetic complications (1).

 

PATHOPHYSIOLOGY- HOW METABOLIC MICROVASCULAR EFFECTS INFLUENCE MACRO-MOVEMENTS

 

From the various previously reported speculative pathophysiological basis of DS, Dubey et al (10) proposed "ominous octet" of pathogenesis of DS, which includes sequential occurrence of following factors: 1) gemistocytopathy, 2) petechial hemorrhage, 3) methemoglobin deposition, 4) mineral (calcium and magnesium) deposition, 5) cytotoxic edema, 6) myelinolysis, 7) gliosis, and 8) atrophy. The hyperglycemic state results in hyperosmolarity and hyperviscosity leading to reduced cerebral blood flow causing insult to the striatal astrocytes which are exquisitely sensitive to ischemia. These tumescent reactive astrocytes are known as gemistocytes, the most consistent finding gathered from limited number of biopsy studies (2).

 

Interestingly enough, genesis of majority of hyperglycemic movement disorders occurs in the background of non-ketotic milieu (1,2). In non-ketotic hyperglycemia brain metabolism is shifted towards the alternative anaerobic pathway in Krebs cycle causing depletion in gamma-aminobutyric acid (GABA), an inhibitory neurotransmitter. This leads to attenuated inhibition of the subthalamus by the medial globus pallidus resulting in hyperkinetic movements. Conversely, GABA can be readily re-synthesized from acetoacetate, which is in abundance in the ketotic milieu (8). Hence, in the latter state, hyperkinetic movements rarely occur unless some other sinister mechanisms (such as cerebrovascular insufficiency or ultrastructural changes in basal ganglia) are at play (1).

 

NEURORADIOLOGICAL AND CLINICAL CONUNDRUM OF DS

 

Although the sensitivity of MRI was observed to be higher than CT scan to detect DS (95.3% vs. 78.9%), the need for CT can’t be obviated in cases of negative MRI scans. There is plenty of cases where mismatch (defined as the complete absence of anomaly in basal ganglia on one imaging modality, but not the other) and incompatibility (defined as the difference in locations of striatal anomalies between CT and MRI) exist (2). Hyperdensity on CT or hyperintensity on T1-weighted MRI in contralateral (to the side of the abnormal movements) putamen surrounding edema or mass effect, along with hyperglycemia and hemichorea-hemiballism movements, is pathognomonic of DS (10). Putamen is the most commonly involved striatal structure, whereas isolated caudate or globus pallidus or subthalamic nucleus involvement seem to be less frequent (1,2). A significant portion of cases shows concomitant affliction of all three striatal components (putamen, caudate, and globus pallidus) (2). The reason behind putaminal vulnerability to hyperglycemia and how the same anatomical lesion causes such wide arrays of movement disorders remain elusive.

 

The pathological basis behind the striatal hyperintensity on T1-weighted MRI and hyperdensity on CT scan in these patients can be proved by histopathological evidence of petechial hemorrhages causing accumulation of methemoglobin (2). Unfortunately, this theory of microhemorrhages behind T1 hyperintensity can’t be substantiated well on corresponding gradient-echo images. In contrast, accumulation of gemistocytes due to ischemic events and neuronal dysfunction may partially explain the striatal hyperintensity on T1-weighted MRI, but not hyperdensity on CT (2,10). Few DS cases have documented restricted diffusion in diffusion-weighted imaging sequence (24). Advanced imaging modalities such as MR volumetry, spectroscopy, functional MRI, positron emission tomography (PET), single photon emission CT (SPECT), susceptibility weighted MR, perfusion imaging etc. although not routinely done in clinical practice for diagnosis of DS, might unveil its intricate pathophysiological basis (2,10).  

 

Previous studies showed that in different case series patients with choreo-ballistic movements did not have suggestive neuroimaging findings in 5-45% cases (clinically isolated DS) (2,14-17,25). Study focusing on both choreo-ballistic and non-choreo-ballistic movements revealed that only 44% cases had changes in brain MRI (1). This wide variability had been attributed to the varied use of MRI or CT and non-homogenous neuroradiological definition of DS applied among various studies (1,2,10). Moreover, neuroradiological changes lag behind the clinical manifestations. Nevertheless, it underscores the importance of initiating management by recognizing this disease phenomenon on the basis of clinical symptomatology (presence of acute onset movement disorders with concurrent hyperglycemia) without waiting for neuroimaging (1). On the contrary, 2% of patients may show radiological striatal lesions without any clinically manifested movement disorders (radiologically isolated DS) (2,26-28). There are also plenty of reports of clinical-radiological discordance or inconsistency in DS (1,2,14). Thus, striatopathy with clinically manifested movement disorders (symptomatic DS) can be subdivided into two groups, i.e., 1) concordant: bilateral involuntary movements with bilateral DS, or unilateral involuntary movements with contralateral DS (6); and 2) discordant: bilateral involuntary movements with unilateral DS or unilateral involuntary movements with bilateral or ipsilateral DS (10,29-31). This frequently observed clinical-radiological dissociation in DS is apparently contradictory with the classical concept of neurological localization of lesion-manifestation and requires further studies with newer neuroimaging modalities (1,10). Due to the controversial and ambiguous nature of the term "diabetic striatopathy" in literature (2), we had previously proposed a three-subset classification (10) (figure- 1).

 

Figure 1. Dubey’s classification schema of Diabetic Striatopathy (Adapted from: Dubey S, Biswas P, Ghosh R, Chatterjee S, Ray BK, Benito-León J. Neuroimaging of Diabetic Striatopathy: More Questions than Answers. Eur Neurol. 2022;85:371-6.)

Figure 2. Right striatal hyperintensity on T1 weighted MRI in a 56-yeald-old lady with previously undiagnosed diabetes presented with left hemichorea-hemiballism persisting for 1 week. Blood glucose was 453 mg/dl. Movement disorders abated with management of hyperglycemia with insulin therapy alone.

Figure 3. Left striatal hypodensity on non-contrast CT scan in a 68-year-old gentleman with diabetes presented with right hemichorea. Blood glucose was 356 mg/dl and HbA1c was 15.2%. Movement disorder was partially improved with glycemic control and needed haloperidol for complete recovery.

 

TREATMENT AND PROGNOSIS

 

Intensive management of hyperglycemia with insulin remains the pivotal measure to treat movement disorders associated with hyperglycemia (1,2). Some authors have speculated worsening of involuntary movements on aggressive lowering of blood glucose (analogous to diabetic retinopathy) (32-35), but this needs clarification by further reports. According to past studies, from one-fourth to almost half of the patients recover with insulin therapy alone (1,2,16,17) with a higher recovery rate in ketotic hyperglycemia cases (21). Additional therapies such as haloperidol, tetrabenazine, risperidone, tiapride (ballism and chorea), levodopa (parkinsonism), trihexyphenidyl, clonazepam (dystonia), pramipexole (restless leg syndrome), propranolol (tremor), carbamazepine (hemifacial spasm) etc. have been used with varying success rates (1,2). Whether the requirement of additional drugs may be attributed to late presentation or diagnostic delay needs further study (1,2,15,20). Surgical interventions such as pallidotomy, ventrolateral thalamotomy, transcranial magnetic stimulation, and globus pallidus internus deep brain stimulation had been tried for intractable symptoms (2,36-38).

 

In the study by Dubey et al (1), treatment of movement disorders was documented and followed up for at least three weeks. Patients who recovered fully from all involuntary movements within seven days were regarded as early responders, while the rest were taken as late responders. In that series the majority (47.5%) of the patients had early and complete resolution of symptoms, 28.8% responded late but had a complete reversal, while 23.7% cases recovered partially. Interestingly, in Chua et al’s analysis recovery was earlier among patients on glucose-therapy only (2 days) compared to those receiving additional anti-chorea medications (14 days), although median pre-treatment lag period was identical between those two groups (4 days) (2). Overall, the previous literature showed that recovery rate varied from 76.4% to 100% (2,14-21), which could be attributable to heterogeneity in the definition of recovery (clinical and/or neuroradiological) and duration of follow-up employed across different studies. During recovery, as expected, symptomatic improvement precedes abolition of neuroradiological stigmata. Minimum time period for radiological reversal noted in study by Chua et al were 10 days on CT and 60 days on MRI. On follow-up MRI scans progressive increase in striatal hyperintensity to reach its maximum limit was noted at around 90 days, whereas the mean periods of complete radiological reversal were around 60 and 180 days on CT and MRI, respectively. The median duration of discernible changes on CT and MRI were 24 and 120 days respectively (2). However, it is not at all uncommon to come across cases demonstrating persisting striatal anomalies on follow-up neuroimaging for months irrespective of symptomatic recovery (2,3,39). Currently there is paucity of studies which longitudinally evaluate the evolution of radiological changes over the course of disease process.

 

Despite having limited data regarding long-term follow-up, nearly 20% cases of DS clinically recurred even after initial resolution of striatal anomalies., which underscored the importance of periodic neuroradiological surveillance even after initial recovery. Recurrence rate did not differ across different treatment modalities (i.e., with or without additional use of anti-chorea medications) employed (2).

 

CONCLUSION

 

Acute onset or de novo movement disorder is one of the important neurological complications of DM, most prevalent but not limited to Asian population. Unfortunately, it is still less well-recognized among physicians, diabetologists, and endocrinologists leading to its diagnostic delay and probably poorer prognosis. Although DS and other movement disorders are generally complications of poorly controlled long-standing type 2 diabetes in the non-ketotic hyperglycemia state among elderly, it may be the first presentation of diabetes. Hence, clinicians must be aware of this entity so that crucial time is not wasted and readily available glucose measurement are ordered when dealing with such patients irrespective of their past glycemic status. Exact pathophysiological mechanisms, genetic basis, radiological correlates, and the explanation for the seemingly discordant clinical-radiological picture in hyperglycemia-induced movement disorders remain elusive. Much work needs to be done to determine the optimal management and prognostic indicators of this emerging disease entity.

 

ACKNOWLEDGMENTS

 

Informed consent was obtained from all patients whose videos are included in this chapter.

 

REFERENCES

 

  1. Dubey S, Chatterjee S, Ghosh R, Louis ED, Hazra A, Sengupta S, et al. Acute onset movement disorders in diabetes mellitus: A clinical series of 59 patients. Eur J Neurol. 2022;29:2241-8.
  2. Chua CB, Sun CK, Hsu CW, Tai YC, Liang CY, Tsai IT. "Diabetic striatopathy": clinical presentations, controversy, pathogenesis, treatments, and outcomes. Sci Rep. 2020;10:1594.
  3. Chatterjee S, Ghosh R, Ojha UK, Diksha, Biswas P, Benito-León J, et al. Recurrent facial focal seizure with chronic striatopathy and caudate atrophy- a double whammy in an elderly woman with diabetes mellitus. Neurohospitalist. 2022;12:147-50.
  4. Chatterjee S, Ghosh R, Kumari R, Ojha UK, Benito-León J, Dubey S. Faciobrachial myoclonus as the presenting manifestation of diabetic keto-acidosis. Tremor Other Hyperkinet Mov (N Y). 2021;11:9.
  5. Ghosh R, Roy D, Chatterjee S, Dubey S, Swaika BC, Mandal A, et al. Hemifacial spasm as the presenting manifestation of type 3c diabetes mellitus. Tremor Other Hyperkinet Mov (N Y). 2021;11:14.
  6. Ghosh R, Dubey S, Roy D, Ray A, Pandit A, Ray BK, et al. Choreo-ballistic movements heralding COVID-19 induced diabetic ketoacidosis. Diabetes Metab Syndr. 2021;15:913-7.
  7. Dubey S, Chatterjee S, Mukherjee D, Ghosh R, Sengupta S, Lahiri D, et al. “Dancing belly” in an old diabetic lady. J Family Med Prim Care. 2020;9:2580-2.
  8. Jagota P, Bhidayasiri R, Lang AE. Movement disorders in patients with diabetes mellitus. J Neurol Sci. 2012;314:5-11.
  9. Chakraborty PP, Ray S, Bhattacharjee R, Ghosh S, Mukhopadhyay P, Mukhopadhyay S, et al. First Presentation of Diabetes as Diabetic Ketoacidosis in a Case of Friedreich's Ataxia. Clin Diabetes. 2015;33:84-6.
  10. Dubey S, Biswas P, Ghosh R, Chatterjee S, Ray BK, Benito-León J. Neuroimaging of Diabetic Striatopathy: More Questions than Answers. Eur Neurol. 2022;85:371-6.
  11. Shafran I, Greenberg G, Grossman E, Leibowitz A. Diabetic striatopathy- Does it exist in non-Asian subjects? Eur J Intern Med. 2016;35:51-4.
  12. Ryan C, Ahlskog JE, Savica R. Hyperglycemic chorea/ballism ascertained over 15 years at a referral medical center. Parkinsonism Relat Disord. 2018;48:97-100.
  13. Ottaviani S, Arecco A, Boschetti M, Ottaviani E, Renzetti P, Marinelli L. Prevalence of diabetic striatopathy and predictive role of glycated hemoglobin level. Neurol Sci. 2022;43:6059-65.
  14. Gómez-Ochoa SA, Espín-Chico BB, Pinilla-Monsalve GD, Kaas BM, Téllez-Mosquera LE. Clinical and neuroimaging spectrum of hyperglycemia-associated chorea-ballism: systematic review and exploratory analysis of case reports. Funct Neurol. 2018;33:175-87.
  15. Cosentino C, Torres L, Núñez Y, Suárez R, Vélez M, Flores M. Hemichorea/hemiballism associated with hyperglycemia: report of 20 cases. Tremor other hyperkinet mov (N Y). 2016;6:402.
  16. Guo Y, Miao YW, Ji XF, Li M, Liu X, Sun XP. Hemichorea associated with nonketotic hyperglycemia: clinical and neuroimaging features in 12 patients. Eur Neurol. 2014;71:299-304.
  17. Lee SH, Shin JA, Kim JH. Chorea-ballism associated with nonketotic hyperglycaemia or diabetic ketoacidosis: characteristics of 25 patients in Korea. Diabetes Res Clin Pract. 2011;93:e80-3.
  18. Oh SH, Lee KY, Im JH, Lee MS. Chorea associated with nonketotic hyperglycemia and hyperintensity basal ganglia lesion on T1-weighted brain MRI study: a meta-analysis of 53 cases including four present cases. J Neurol Sci. 2002;200:57-62.
  19. Lee BC, Hwang SH, Chang GY. Hemiballismus-hemichorea in older diabetic women: a clinical syndrome with MRI correlation. Neurology. 1999;52:646-8.
  20. Prabhu S, Ramya N. Movement disorders and diabetes: a study of South India. Internet J Neurol. 2012;14:1-5.
  21. Chen C, Zheng H, Yang L, Hu Z. Chorea-ballism associated with ketotic hyperglycemia. Neurol Sci. 2014;35:1851-5.
  22. Paiboonpol S. Epilepsia partialis continua as a manifestation of hyperglycemia. J Med Assoc Thai. 2005;88:759-62.
  23. Shrivastava V, Burji NP, Basumatary LJ, Das M, Goswami M, Kayal AK. Etiological profile of epilepsia partialis continua among adults in a tertiary care hospital. Neurol India. 2013;61:156-60.
  24. Chu K, Kang DW, Kim DE, Park SH, Roh JK. Diffusion-weighted and gradient echo magnetic resonance findings of hemichoreahemiballismus associated with diabetic hyperglycemia: a hyperviscosity syndrome? Arch Neurol. 2002;59:448–52.
  25. Chen C, Zheng H, Yang L, Hu Z. Chorea-ballism associated with ketotic hyperglycemia. Neurol Sci. 2014;35:1851-5.
  26. Choi JY, Park JM, Kim KH, Park JS, Shin DW, Kim H. Radiographic basal ganglia abnormalities secondary to nonketotic hyperglycemia with unusual clinical features. Clin Exp Emerg Med. 2016;3:252-5.
  27. Hsu JL, Wang HC, Hsu WC. Hyperglycemia-induced unilateral basal ganglion lesions with and without hemichorea. A PET study. J Neurol. 2004;251:1486-90.
  28. Hansford BG, Albert D, Yang E. Classic neuroimaging findings of nonketotic hyperglycemia on computed tomography and magnetic resonance imaging with absence of typical movement disorder symptoms (hemichorea-hemiballism). J Radiol Case Rep. 2013;7:1-9.
  29. Danve A, Kulkarni S, Bhoite G. Non-ketotic hyperglycemia unmasks hemichorea. J Community Hosp Intern Med Perspect. 2015;5:27825.
  30. Lin JJ. Ipsilateral putamen hyperintensity on T1-weighted MRI in non-ketotic hyperglycemia with hemiballism-hemichorea: a case report. Parkinsonism Relat Disord. 2001;7:319-21.
  31. Fong SL, Tan AH, Lau KF, Ramli N, Lim SY. Hyperglycemia-associated hemichorea-hemiballismus with predominant ipsilateral putaminal abnormality on neuroimaging. J Mov Disord. 2019;12:187-9.
  32. Lim KX, Khaing Zin T, Yu Z, Peh WM. Delayed Presentation of Hemichorea in Diabetic Striatopathy. Cureus. 2022;14:e30219.
  33. Lizarraga KJ, Adams D, Post MJD, Skyler J, Singer C. Neurovascular uncoupling after rapid glycemic control as a trigger of the diabetic-uremic striatopallidal syndrome. Parkinsonism Relat Disord. 2017;39:89-90.
  34. Ando Y, Kadoya M, Kodera T. Involuntary Movements During Treatment for Hyperglycemia. AACE Clin Case Rep. 2022;9:21-2.
  35. Cho HS, Hong CT, Chan L. Hemichorea after hyperglycemia correction: A case report and a short review of hyperglycemia-related hemichorea at the euglycemic state. Medicine (Baltimore). 2018;97:e0076.
  36. Son BC, Choi JG, Ko HC. Globus Pallidus Internus Deep Brain Stimulation for Disabling Diabetic Hemiballism/Hemichorea. Case Rep Neurol Med. 2017;2017:2165905.
  37. De Vloo P, Breen DP, Milosevic L, Lee DJ, Dallapiazza RF, Hutchison WD, et al. Successful pallidotomy for post-hyperglycemic hemichorea-ballism. Parkinsonism Relat Disord. 2019;61:228-30.
  38. Kaseda Y, Yamawaki T, Ikeda J, Hayata M, Dohi E, Ohshita T, et al. Amelioration of persistent, non-ketotic hyperglycemia-induced hemichorea by repetitive transcranial magnetic stimulation. Case Rep Neurol. 2013;5:68-73.
  39. Lucassen EB, Delfyett WT, Stahl MC. Persistent Hemichorea and Caudate Atrophy in Untreated Diabetic Striatopathy: A Case Report. Case Rep Neurol. 2017;9:299-303.

 

Endocrine Testing Protocols: Hypothalamic Pituitary Adrenal Axis

ABSTRACT

 

Abnormalities in the hypothalamic pituitary adrenal (HPA) axis are identified by a careful analysis of both direct and non-stimulated measurements of the hormones as well as provocative tests.  Dynamic testing is useful to determine if elevated levels are suppressible and whether there is sufficient hormone reserve when low levels are measured under stimulation. A combination of all these analyses can distinguish between normal physiology and the consequences of clinical disease in the HPA axis.  While clinical suspicion drives the testing performed, arrival at the correct diagnosis by laboratory testing is crucial for cure of the patient.  Knowledge of the methodologies used in measuring cortisol and ACTH and associated hormones and binding proteins is essential for correct interpretation of the tests.  In this review we compare methodologies available, sensitivity and specificity of the various assays and volumes of sample needed. There are at least 7 different types of dexamethasone suppression testing and they are compared and described in detail. Confirmation of the anatomic source of the hormone is necessary. Petrosal sinus sampling and adrenal vein sampling are reviewed and the clinical indications for each discussed. Finally, once the endocrine diagnosis is reached based on endocrine testing, imaging studies are then reviewed which can confirm the endocrine diagnosis. An abnormality in the HPA axis is a laboratory diagnosis and radiologic imaging is reserved for the last step in the diagnosis of endocrine disease.

 

NONSTIMULATED HORMONE MEASUREMENTS

 

Overview

 

In evaluation of the hypothalamic pituitary adrenal (HPA) axis, static measurement of hormones is seldom useful due to the variable nature of cortisol and ACTH secretion in normal physiological states. In general, if one is suspicious of hypofunction of the HPA axis, then measurement of morning cortisol at 8 am when it is expected to be at its peaks is a good screening strategy. Depending on the result, this might need to be followed by dynamic testing to stimulate either adrenocorticotrophic hormone (ACTH) or cortisol for confirmatory purposes. On the other hand, if one is concerned about Cushing’s syndrome (CS), an overproduction of cortisol or ACTH, then measurement of cortisol should be performed late at night, when it is expected to be at its nadir. Alternatively, one could test cortisol’s response to suppression with dexamethasone.

 

The American Endocrine Society Clinical Guidelines recommend one of the following tests for the initial CS testing: at least two measurements of urinary-free cortisol (UFC), two measurements of late night salivary cortisol (LNSC), 1 mg overnight dexamethasone suppression test (DST) or a longer low-dose DST (1). Cortisol measurement (serum, UFC or salivary) is the end point for each recommended test.

 

Despite recent literature reports describing utility of direct salivary and urine cortisol measurements in CS diagnosis (2-4), most clinicians prefer provocative testing due to the variable nature of cortisol and ACTH secretion in normal physiological states. Cortisol is secreted under the direction of ACTH and follows a diurnal variation, with peak values at 08:00 h and a nadir at 22:00 h. In CS, diurnal variation is lost and PM cortisol level is inappropriately elevated. Superimposed on this diurnal pattern are 8-10 pulsatile peaks released during the course of a 24-hour period. Therefore, depending on the instance when blood is sampled, there can be significant variation in the absolute values of ACTH and serum cortisol. Due to this variability of cortisol and ACTH levels, it may be challenging to distinguish pituitary-dependent Cushing’s disease from pseudo-Cushing’s states. Cunningham et al conducted a study where blood was sampled and cortisol measured every 20 to 30 minutes for 24 hours. The group demonstrated that both circadian and pulse amplitudes of cortisol secretion were decreased in Cushing’s disease (5).

 

This section provides and overview of methodologies commonly used in clinical laboratories for direct determinations of cortisol and ACTH, regardless of whether they are a part of a provocative testing series or direct, non-stimulated hormone assessment.

 

Cortisol

 

Methods currently available for measuring serum cortisol levels include automated immunoassays and liquid chromatography-tandem mass spectrometry (LC-MS/MS).

 

CORTISOL IMMUNOASSAYS (TOTAL CORTISOL)

 

Cortisol immunoassays are widely available, have been in use for a long time, and automated methods provide high throughput with minimal manual sample manipulations. Virtually all immunoassay methods are based on the competitive binding principle, where cortisol from the patient sample and exogenous, labeled cortisol compete for the binding sites available on the anti-cortisol antibody. The major difference between the assays is in the label design and chemistry enabling antibody-antigen binding. All currently available cortisol methods have limit of detection below 1 µg/dL, providing sufficient sensitivity to support interpretation of CS dynamic testing results.

 

A widely recognized disadvantage of immunoassays is a potential of interferences from auto-, anti-animal or heterophilic antibodies. In addition, the older generations of cortisol assays had significant cross-reactivity with other steroids, such as 6-b-hydroxycortisol or prednisolone, due to the use of less specific polyclonal antibody in the assay formulation. However, the majority of current immunoassay methods have transitioned to a more specific monoclonal antibody format, minimizing or eliminating cross-reactivity with other steroids. It should also be noted that some immunoassay vendors use biotinylated antibodies in their assay design. In these instances, biotin may interfere with the assay, causing spuriously elevated cortisol measurement. The presence and magnitude of interference is vendor-specific and the potential of biotin interference should be checked with the laboratory that performs the testing. In general, none of the assays manufactured by Abbott use biotin in reagent formulation, while all assays manufactured by Roche do. The Roche cortisol assay should not be used to measure serum cortisol in a patient taking daily doses of biotin exceeding 5 mg, unless blood is obtained at least 8 hours following the last biotin ingestion.

 

LC-MS/MS CORTISOL ASSAYS

 

The LC-MS/MS assays utilize liquid chromatography to separate cortisol from other serum/plasma components and tandem mass spectrometry to detect and quantify compounds of interest. LC-MS/MS based methods offer superior analytical sensitivity and specificity over immunoassays.

 

Serum Free Cortisol

 

In conditions where CBG concentrations are affected, such as pregnancy or critical illness for example, total serum cortisol may not always reflect the true pituitary-adrenal status. In these cases, assessment of serum free cortisol is preferred. Free serum cortisol concentration are directly measured by separating free serum cortisol fraction using equilibrium dialysis (6) or ultrafiltration (7, 8) followed by cortisol determination, usually performed using LC-MS/MS method. Alternatively, free serum cortisol can be estimated by calculating the ratio of serum cortisol and CBG to obtain serum cortisol index (6).  Although not affected by CBG levels, free cortisol is also secreted in episodic fashion and thus not much more useful than random total serum cortisol levels in assessment of HPA axis functionality.

 

Urinary Free Cortisol

 

Cortisol is excreted in urine in an unbound (free) form and, like free serum cortisol is unaffected by fluctuations in CBG levels. Properly collected 24-hour urine specimens can be used to eliminate fluctuations that would affect serum cortisol levels, due to the pulsatile nature of its release. Therefore, measurement of UFC from 24 hour urine collections has become a valuable diagnostic tool for evaluation of adrenal cortical function and it is one of the first line tests recommended for CS diagnostic testing (1). In the unstressed patient, with normal renal function, elevation of UFC in 24-hour urine specimen is usually sufficient to diagnose CS. A normal result is strong evidence against that diagnosis. Although this test has long been used, its utility in CS diagnosis still remains somewhat controversial. Studies show wide variability in clinical utility of UFC for diagnosis of CS with clinical sensitivity ranging from 53% to over 90% and specificity ranging from 79% to 90% (2, 3, 9). These differences are due to differences in study design, cut-off, and methodology used. Furthermore, in a careful study of normal subjects de Boss Kuil et al found that urinary excretion of free cortisol can differ by as much as 50% between the two consecutive urine collections, while the creatinine values can differ by as much as five fold (10). Since the ratio of free cortisol/creatinine also varies considerably (range 1.0-3.7; median 1.3), intra-variation in urinary cortisol excretion could not be attributed to variation in creatinine excretion. In addition to biological variation, other factors include difficulty in over or under collection of urine. Given such wide discrepancies in reported clinical sensitivity and specificity of UFC measurements and significant intra-individual UFC variability, this test may not be an ideal choice for initial screening of CS.

 

Methodology used for UFC quantitation is the same as for serum cortisol. In terms of specimen collection, an 8:00 AM to the following day’s 8:00 AM collection is desirable. Samples should be refrigerated during collection and, while preservatives are not required, boric acid is usually acceptable. Quantitation of urine cortisol with a more sensitive and specific LC-MS/MS method is generally preferred over immunoassays. Typically, all the LC-MS/MS UFC assays involve a sample pretreatment with an organic solvent which removes the interfering substances. However, some UFC assays immunoassays either do not include this pretreatment step or offer it as an optional step to the user. As a result, UFC reference ranges vary widely between the assay manufacturers, methodologies, and different laboratories. To increase sensitivity, it is recommended that the upper limit of normal for any UFC assay be used as a positive test (5). It would be thus incorrect to make a diagnosis of adrenal insufficiency relying solely on 24-hour urine collections.

 

Salivary Cortisol

 

Late-night (23:00-24:00 h) salivary cortisol (LNSC) is one of the first line tests used to screen for CS. Most studies report high diagnostic sensitivity of this test (80-90%), but there are discrepancies in reported specificities (70-90%), resulting mostly from difference in methodologies and populations studied (2, 3, 11-13) Interestingly, mass spectrometry assays demonstrate high sensitivity, but low specificity (75%) for the diagnosis of CS (11). One potential explanation, as postulated by Raff, is that higher analytical specificity of mass spectrometry actually leads to lower diagnostic specificity, suggesting that cortisol metabolites and precursors picked up by immunoassays may be diagnostically relevant (14). Kannankeril et al recently reported that LNSC has excellent negative predictive value (99.8%) but poor positive predictive value (16.8%) for diagnosis of ACTH-dependent CS (12). Thus, a negative LNSC can be used to rule out ACTH-dependent CS, but complementary tests of adrenal function are needed to establish the diagnosis.

 

Salivary cortisol concentration is not dependent on CBG and could therefore be useful during an ACTH stimulation testing in patients with increased CBG concentrations due to increased estrogen or decreased plasma binding globulins due to critical illness.

 

Similar to UFC, the assay methodology remains the same as serum cortisol with the differences in specimen collection.

 

ACTH

 

ACTH measurements, while subject to the same circadian variability as cortisol (actually it is the variability of the ACTH that is directly responsible for the variability of the cortisol), are not subject to the effects of CBG. Values of ACTH > 100 pg/ml in the setting of possible adrenal insufficiency are usually suggestive of primary adrenal insufficiency, while values >500 pg/ml are diagnostic. Low concentrations of plasma ACTH are not diagnostic, except for the undetectable levels observed in patients with cortisol producing adrenal adenomas. Plasma ACTH concentration is also low in patients taking exogenous steroids.

 

Unlike widely available cortisol assays, the availability of clinical ACTH assays is limited. All currently available methods are immunoassays based on the “sandwich” principle, where two antibodies that recognize different ACTH epitopes are utilized. The first antibody, designated as capture antibody, detects one specific site on ACTH molecule and is used to pull the antigen from the patient’s plasma. The second antibody that detects a different ACTH epitope is then used to “sandwich” the antigen and generate a signal.

 

As is the case with any immunoassay, ACTH assays are susceptible to heterophilic antibody interferences. Several cases have been described in literature where aberrant, falsely elevated ACTH results were inconsistent with clinical picture and lead to unnecessary testing, misdiagnosis, and in some cases surgical interventions. These cases emphasize the importance of interaction between clinicians and the laboratory to identify any interference present and ensure that each patient is appropriately managed (15, 16). In addition, just as is the case with cortisol immunoassays, some vendors use biotinylated antibodies in the capture antibody design. Unlike cortisol, however, biotin interference may result in falsely decreased ACTH levels. The two most commonly used ACTH assays are manufactured by Siemens and Roche. Siemens ACTH assay is not affected by biotin, while the recommendation for Roche ACTH assay is not to use the test in patients ingesting >5 mg biotin daily, unless at least 8 hours had elapsed following the last biotin dose (cf. Roche Elecsys ACTH Package Insert V 12.0, 2020-11).

 

The preferred specimen for ACTH is EDTA plasma. ACTH is heat labile, and if not collected and preserved on ice, will lead to proteolysis, which can reduce the plasma concentration leading to falsely lower values.

 

Miscellaneous Non-Stimulated Measurements

 

CORTISOL BINDING GLOBULIN (CBG)

 

As mentioned earlier, the majority of cortisol (~92%) is bound to CBG, a serum protein. CBG levels increase in pregnancy and patients on oral contraceptives or supplemental estrogen. CBG is decreased in hyperinsulinemic states, nephrotic syndrome, starvation, severe illness, and chronic liver disease. This test is useful for the assessment of unexpected serum cortisol values. It is offered by large reference laboratories and uses a radioimmunoassay method.

 

11-DEOXYCORTISOL (COMPOUND S)

 

This is the immediate precursor of cortisol and is typically increased when ACTH is elevated or in 11 beta-hydroxylase deficiency. The method for 11-deoxycortisol measurement is now available by LC-MS/MS technology and is offered by most reference laboratories.

 

ANTI-ADRENAL ANTIBODIES

 

The measurement of anti-adrenal antibodies has been suggested to be useful in detecting early evidence of adrenal insufficiency, before cortisol values are decreased even in response to stimuli. The only test currently clinically available is a test that detects 21-hydroxylase autoantibodies, which are present in the common autoimmune form of Addison’s disease (17). This test is offered by major reference laboratories and is based on the radioimmunoassay format.

 

CORTICOTROPHIN RELEASING HORMONE (CRH)

 

Serum concentration of CRH is markedly elevated in pregnancy, presumably due to the production of CRH by the placenta. High levels are associated with high levels of CRH binding protein. Although mentioned as useful in the diagnosis of ectopic CRH syndromes, little data is available in this regard. CRH testing is not commonly done and we have not been able to find a commercial laboratory that is currently performing this test.

 

DYNAMIC TESTING

 

Glucocorticoid Deficiency

 

Adrenal insufficiency is a life-threatening disorder and prompt diagnosis is important because adequate hormonal replacement therapy can be lifesaving.

 

Despite that more than 35 years have elapsed since the initial description of the use of the insulin tolerance test (ITT) to diagnose adrenocortical deficiency (18), and more than 200 scientific publications in this area, clinicians today still argue as to which is the most sensitive and specific test to diagnose adrenocortical deficiency. The ITT is still regarded as the gold standard upon which to compare all other tests of HPA axis function. Unfortunately, this test has a considerable spectrum of intra-individual and inter-individual variation (19, 20). Therefore, when comparing other tests to the "gold standard", if the standard is not reliable, how can one determine the effectiveness of the other forms of testing? The problem lies in the ability of a single laboratory to know what the values are for their tests. Therefore, ranges from an ITT test response in normal subjects performed in one laboratory may not be normal for another laboratory. Taking this into account there are some general guidelines that are available for evaluating patients with suspected adrenal insufficiency.

 

PRIMARY ADRENAL INSUFFICIENCY

 

High Dose ACTH Stimulation Test

 

WHEN TO USE THIS TEST: Patients acutely ill in the hospital or clinic who present with signs and symptoms suggestive of primary adrenal insufficiency. Patients who are thermodynamically unstable should be resuscitated with crystalloids and given dexamethasone prior to testing if the diagnosis of primary adrenal insufficiency is being considered.

 

PROCEDURE: An intravenous (IV) line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. The IV line is to be kept open with 0.9% sodium chloride (NaCl) at a rate of 50 ml/hr. Blood is drawn at 0 min for ACTH (2 ml in a lavender top tube on ice) and cortisol (2 ml in a red top tube). Cosyntropin, 0.25 mg is administered as an IV bolus over 2 minutes. The cosyntropin comes as a lyophilized powder which should be reconstituted with 1 ml of 0.9% NaCl. Thirty min after the injection, blood is obtained from the IV line (2 ml) for cortisol. The same is repeated at 60 min (2 ml) for cortisol.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day. If the patient is receiving hydrocortisone or cortisone acetate, the medication should be held for at least 12 hours prior to testing (if possible). Although the test can be performed while the patient is receiving dexamethasone, there is some cross-reactivity in some assays and cortisol levels may not be accurate. Each laboratory should determine for itself, the effect of dexamethasone on their assay.

 

Patients with known sensitivity to cosyntropin or its preservatives should not have it administered. Oral estrogen use may result in elevation of the total serum cortisol level due to increased corticosteroid binding globulin (21). Patients with albumin <2.5 g/dL may also have a low cortisol level (21, 22).

 

CONTRAINDICATIONS: Hypersensitivity to cosyntropin or any component of the formulation.

 

WARNINGS / PRECAUTIONS: Use with caution in patients with pre-existing allergic disease or a history of allergic reactions to corticotropin. Class C in pregnancy.

 

ADVERSE REACTIONS 1% to 10%: Cardiovascular: Flushing. Central nervous system: Mild fever. Dermatologic: Pruritus. Gastrointestinal: Chronic pancreatitis. <1%: Hypersensitivity reactions

 

DRUG INTERACTIONS: Decreased effect: May decrease the effect of anticholinesterases in patients with myasthenia gravis; nondepolarizing neuromuscular blockers, phenytoin and barbiturates may decrease effect of cosyntropin

 

INTERPRETATION OF RESULTS: Baseline cortisol values <5 µg/dl and ACTH concentrations >100 pg/ml are usually diagnostic of primary adrenal insufficiency. The normal peak cortisol value post stimulation should be an increment no less than 7µg/dl. A peak stimulated cortisol value of >18 µg/dl at 30 min is considered normal. Since 37% of subjects had a peak response to cosyntropin at 30 min and 63% had a peak response at 60 min, both time points are analyzed in all patients and if either the 30 min or 60 min sample reaches the criteria as noted above, the test is considered normal (23).  However, there is some suggestion that new generation cortisol assays may have different cutoff values, but these have not been verified (24). 

 

Free cortisol, instead of total cortisol can be measured using a value of >1.2 µg/dl at 30 or 60 min as a normal result. This can be indicated in patient with albumin levels <2.5 g/dL or those with low cortisol binding globulin.

 

Serum aldosterone can be measured in 0 min, 30 min and 60 min blood samples as ACTH stimulation of the adrenal cortex will also stimulate aldosterone. It has been suggested that a normal aldosterone response to ACTH in the presence of a suboptimal cortisol response is diagnostic of secondary adrenal insufficiency (25).

 

Low dose ACTH stimulation Test

 

WHEN TO USE THIS TEST: Patients with subtle signs of adrenal insufficiency or patients who have been treated with glucocorticoids in whom determination of adrenal reserve is necessary. Patients who have autoimmune disease and may have early adrenocortical insufficiency may be best assessed with this test.

 

PROCEDURE: An intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. The IV line is to be kept open with 0.9% NaCl at a rate of 50 ml/hr. Blood is drawn at 0 min for ACTH (2 ml in a lavender top tube on ice) and cortisol (2 ml in a red top tube).

 

Cosyntropin, 1 µg is administered as an IV bolus over 2 minutes. The injection material was prepared according to the method of Dickstein as follows: The cosyntropin was diluted with 50 ml of sterile saline to a stock concentration of 5 µg/ml. Aliquots of 0.2 ml were added into sterile plastic tubes and kept at 4oC for a maximum of 4 months (26). Immediately prior to testing 0.8 ml of saline is added to the tube (final dilution 1 µg/ml) and 1 ml is injected into the patient. Thirty min after the injection blood is obtained from the IV line (2 ml) for cortisol. The same is repeated at 60 min (2 ml) for cortisol.

 

SPECIAL CONSIDERATIONS: Same as for high dose ACTH stimulation test, see above.

 

INTERPRETATION OF RESULTS: This test was originally developed to be more sensitive for diagnosing secondary adrenal insufficiency because it was more of a "physiologic" dose. It is much better at diagnosing secondary adrenal insufficiency than the high dose, although it is not at all recommended in acute or recent hypopituitarism when the intact adrenal glands can still respond normally to any dose of ACTH. Although probably not useful for the initial purpose of secondary adrenal insufficiency, it may be more sensitive at distinguishing milder forms of primary adrenal insufficiency (27). Furthermore, this low dose test was helpful in identifying mild adrenal suppression in asthmatic children being treated with inhaled steroids (28). As noted above, each laboratory should establish their normal values, however in general, a stimulated value at 30 or 60 min greater than 20 µg/dl would be considered normal.

 

A meta-analysis of 30 studies enrolling 1209 adults and 228 children with secondary adrenal insufficiency, evaluating the diagnostic accuracy of high and low dose ACTH stimulation concluded that they have similar diagnostic accuracy. They are both adequate to rule in, but not rule out, secondary adrenal insufficiency

 

SECONDARY ADRENAL INSUFFICIENCY (PITUITARY OR HYPOTHALAMIC)

 

Insulin Tolerance Testing (ITT)

 

WHEN TO USE THIS TEST: Patients in whom pituitary or hypothalamic disease may result in impaired corticotroph (or somatotroph) activity. Patients following pituitary surgery or pituitary radiation can be tested at any time, unlike the ACTH stimulation tests described above which are not useful in the acute setting. A random serum cortisol should be drawn prior to scheduling the test if the value is > 20 µg/dl, the test may not be necessary This test, can be performed in the outpatient clinic, however while relatively safe it requires a trained endocrine registered nurse to be present with the patient during the course of the test.

 

PROCEDURE: A 50 ml vial of 50% Dextrose should be at the patient's bedside in a syringe ready for injection before beginning the procedure.

 

An intravenous line is placed 30 minutes before the test for rapid phlebotomy, to eliminate a temporary rise in cortisol associated with a needle stick, and in order to have IV access for 50% Dextrose in the event of severe hypoglycemia. The IV line is to be kept open with 0.9% NaCl at a rate of 50 ml/hr. Blood is drawn at 0' for cortisol (2 ml in a red top tube) and glucose (1 ml in a gray top tube). Blood glucose is also checked at the bedside with a glucose monitor.

 

Regular (short acting) insulin is administered as an IV bolus at a dose of 0.1 units/kg. Blood is sampled for cortisol and glucose as noted above at 10min, 15min, 30min, 45min, 60min, 90min and 120min. A bedside nurse should monitor the blood glucose more frequently if glucose drops below 60 mg/dl on the glucometer or if the patient complains of neuroglycopenic symptoms, such as fatigue, diaphoresis, hunger, lightheadedness, or nausea. The test should continue until the blood glucose concentration drops below 40 mg/dl.

 

In patients with diabetes on insulin, consideration should be given that they may be insulin resistant. In which case, larger doses of insulin may be given. We usually begin with a single bolus of 0.1 U/kg and then re-bolus with insulin depending on the response to the initial dose (either give the same dose again if there was some response but insufficient, or double the dose if there was only minimal response to blood glucose, or give half the dose if the hypoglycemic response was close to the desired goal). This can be repeated several times until adequate hypoglycemia is reached.

 

Once the response goal of a glucose < 40 mg/dl is reached, patients can be fed a meal such as crackers and orange juice. Blood glucose should be checked at 5min, 10min and 15min post feeding. If there is no increase in glucose or a clinical response within 5min, intravenous glucose should be administered. If no response, then a repeat bolus of glucose is suggested. If no response or IV access is lost, glucagon 1 mg intramuscular can be given.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although due to the need for patients to be fasting it is most conveniently done in the morning. If the patient is receiving hydrocortisone or cortisone acetate, the medication should be held for at least 12 hours prior to testing (if possible). Unlike the ACTH stimulation tests, the ITT cannot be performed while the patient is receiving dexamethasone, due to suppression of the hypothalamic pathways necessary to respond to hypoglycemia.

 

In general ITT is not recommended in patients with uncontrolled seizure disorder or significant coronary artery disease.

 

In order to determine if the level of dysfunction is at the hypothalamus or at the pituitary this test is sometimes used in addition to the CRH stimulation test. When the ITT fails to stimulate cortisol, but the CRH test does stimulate it is likely that the patient is having hypothalamic dysfunction.

 

INTERPRETATION OF RESULTS: Serum cortisol should increase within 30 min of the hypoglycemic response to > 20 µg/dl. If the serum cortisol at baseline is 18 ug/dl the test may not be diagnostic. If the baseline serum cortisol is higher than 19 µg, adrenal insufficiency is unlikely. Although the response of cortisol is more reproducible than that of growth hormone in the ITT, intra-subject differences have been reported (20, 30).

 

Metyrapone Testing

 

WHEN TO USE THIS TEST: This test is perhaps the most sensitive to determine whether the HPA axis is intact. Although metyrapone is not generally available from your neighborhood pharmacy, it can be obtained by calling Novartis Pharmaceutical Corp. at 1-800-988-7768 on weekdays. Metyrapone blocks 11-b hydroxylase and results in the inhibition of conversion of 11-deoxycortisol to cortisol. Serum levels of cortisol decrease and concentration of 11-deoxycortisol increases, however 11-deoxycortisol does not down regulate ACTH. Therefore, in a normally functioning HPA axis there is an increase in 11-deoxycortisol. This metabolite can be directly measured in the serum or measured in the urine as 17-OH corticosteroids. This test can help differentiate primary adrenal deficiency from ACTH deficiency. It has a similar diagnostic performance to the ITT and it’s a potential alternative when there is a contraindication to ITT.

 

PROCEDURE: For assessment of adrenal or pituitary insufficiency the test can be performed as an overnight test. Metyrapone is given orally (30 mg/kg body weight, or 2 grams for <70 kg, 2.5 grams for 70 to 90 kg, and 3 grams for >90 kg body weight) at midnight with a glass of milk or a small snack (24). Serum 11-deoxycortisol and cortisol are measured at 8 AM the next morning; it is also recommended to measure plasma ACTH levels (31).

 

SPECIAL CONSIDERATIONS: The concurrent use of glucocorticoids will interfere with the test. Any medications that the patient is taking which increase the P450 enzymes will increase the metabolism and clearance of metyrapone (such as rifampin, phenobarbital, and phenytoin) (32). Similarly, hypothyroidism or hyperthyroidism will affect clearance of metyrapone and the adrenal responsiveness. Therefore, thyroid function tests should be measured prior to performing this test. Measurement of 11-deoxycortisol, like cortisol itself is dependent on CBG and drugs such as estrogens and oral contraceptives will falsely increase the concentrations of 11-deoxycortisol (33).

 

PREGNANCY IMPLICATIONS - Use during pregnancy only if clearly needed. Subnormal response may occur in pregnant women and the fetal pituitary may be affected.

 

LACTATION - Excretion in breast milk unknown/use caution

 

ADVERSE REACTIONS - Frequency not defined. Central nervous system: Headache, dizziness, sedation. Dermatologic: Allergic rash. Gastrointestinal: Nausea, vomiting, abdominal discomfort or pain. Hematologic: Rarely, decreased white blood cell count or bone marrow suppression.

 

INTERPRETATION OF RESULTS: 8 AM serum 11-deoxycortisol concentrations should be >7 µg/dL with serum cortisol less than 5 µg/dL (138 nmol/L), confirming adequate metyrapone blockade. The plasma ACTH concentration at 8 AM should exceed 75 pg/mL (17 pmol/L), confirming that any increases in serum 11-deoxycortisol concentrations are ACTH-dependent, thereby separating primary from secondary adrenal insufficiency (34, 35).

 

Glucocorticoid Excess

 

DEXAMETHASONE SUPPRESSION TEST

 

Measurement of endogenous cortisol production in response to exogenous dexamethasone suppression was the first provocative test and still remains among the most useful tests used for the evaluation of excess cortisol. Dexamethasone, due to its high affinity to the glucocorticoid receptor is a potent inhibitor of ACTH synthesis and release. In addition, most of modern immunoassays for cortisol (both urine and serum) utilize an antibody that does not cross-react with dexamethasone. Therefore, the combination of being able to use relatively low doses and at the same time not interfere with the measurement of cortisol make dexamethasone suppression useful for establishing the presence of a perturbation in the pituitary - adrenal axis and for diagnosing the etiology of hypercortisolism.

 

At least five different tests have been described using dexamethasone, which differ in the dose and timing of dexamethasone treatment and differ in whether there is measurement of urine or serum cortisol or 17-OH-corticostseroids (Table 1). Although the endocrine basis for the tests are in general the same, none are perfect. Confirming the diagnosis of patients with suspected hypercortisolism requires several tests for accurate diagnosis.

 

TABLE 1. Various Dexamethasone Suppression Tests

Dex Supp Test

Dex Dose

Time of Admin

Normal Response

Sens/Spec

Low dose Oral/Night

1 mg

@23:00 x1

<1.8 mcg/dl or <5 mcg/dl

87% / 100%

High dose Oral/Night

8 mg

@23:00 x 1

<50% basal

92% / 100%

Low Dose 2day

0.5 mg

q 6h x 2 days

<10 µg/24h in urine

74-98%/69-100%

High Dose 2 day

2.0 mg

q 6h x 2 days

<50% basal

79% / 100%

Very High dose

8 mg

q 6h x 1 day

<50% basal

74% / 100%

Note: To assure patient compliance and determine whether there is abnormal metabolism of the dexamethasone, serum levels of dexamethasone can be measured. However, this is not a common diagnostic test. Testing can be done by specialized laboratories, such as Esoterix inc. CA. The principle of the assay is RIA after chromatographic sample separation and requires 1 ml of serum sample.

 

All these tests require significant patient participation as the patients are required to self-administer the dexamethasone at inconvenient hours of the day (11PM) or up to 4 times a day. Sampling requires either collection of urine for 24 hours or coming to the physician's office at 8 AM for multiple blood sampling. Drugs that induce hepatic cytochrome P-450 enzymes, such as barbiturates, phenytoin, rifampin, and aminoglutethimide, increase the metabolism of dexamethasone and other steroids. Measurement of serum dexamethasone a few hours after the last dose will help determine if there is abnormal metabolism. All these caveats are in addition to the other problems associated with measurement of cortisol as noted above, including the variable diurnal variation as well as interference with concurrent administration of glucocorticoids, estrogen, or other medications that increase cortisol binding globulin.

 

A popular screening test for confirming hypercortisolism is the overnight 1 mg dexamethasone. A single dose of 1 mg is administered (or 0.3 mg/Kg for children (34) at 11PM and blood is obtained by 8 AM the following morning. The dexamethasone dose is given prior to the diurnal rise in endogenous ACTH release and therefore suppresses the early AM cortisol. A normal response would be a serum cortisol concentration of <1.8 mcg/dl, alternatively a cut point of < 5 µg/dl can be used which will yield more specificity with less sensitivity. If cortisol is >10 µg/dl the likelihood of hypercortisolism is high. The other dexamethasone suppression tests are reviewed in Table VIII. Patients with corticotroph macroadenomas or very active tumors, may have urine free cortisol in excess of 1000 µg/dl which will require higher doses of dexamethasone to confirm suppressibility and/or rule out ectopic ACTH production (36).

 

The two- day low dose dexamethasone suppression test can be used to differentiate Cushing’s syndrome from pseudo-Cushing’s which can present with many of the signs and symptoms associated with hypercortisolism in the setting of other clinical conditions such as depression, alcoholism, PCOS, obesity, and uncontrolled diabetes (37, 38). Dexamethasone 0.5 mg is delivered orally Q6 hours for 48 hours. Serum cortisol is measured 2 hours after the last dose and a cutoff level of <1.4 µg/dl is consistent with pseudo-Cushing’s. Measurement of 24 hour urine excretion of 17-hydroxycorticosteroid and creatinine during the administration of dexamethasone starting at 1200h, has also been suggested with a cut point of 11 umol/day or higher considered positive for Cushing’s syndrome (39). This test however, can misclassify as many as 15% of patients with Cushing’s syndrome and up to 15% of patients with pseudo Cushing’s.

 

The overnight high dose dexamethasone suppression test can help differentiate Cushing’s disease from ectopic ACTH syndrome in patients with ACTH-dependent Cushing’s syndrome. The basis for this differentiation is the fact that ACTH secretion in Cushing’s disease is only relatively resistant to glucocorticoid negative feedback inhibition. Cortisol levels will not suppress normally with overnight 1 mg but will suppress with a higher dose of 8 mg of dexamethasone. Serum cortisol concentration at 8 AM is <5 µg/dL in most patients with Cushing’s disease and is usually undetectable in normal individuals. A more than 50% decrease in cortisol on the day after taking 8 mg dexamethasone supports a diagnosis of Cushing’s disease over ectopic ACTH production. In patients with non-ACTH dependent hypercortisolism, a lack of suppression of cortisol by more than 50% with a low normal ACTH level (5-20 pg/ml) suggests an adrenal etiology.

 

CRH STIMULATION TEST

 

WHEN TO USE THIS TEST: This test is one of the most sensitive to determine if there is an abnormality in the HPA axis and for diagnosing the etiology of hypercortisolism in ACTH dependent Cushing’s.  Although CRH is expensive ($300), when one considers the cost of multiple urine collections and analyses of cortisol as well as the cost of a single MRI of the pituitary (which generally exceeds $1500), CRH is at least cost effective when one considers the overall expense in the evaluation of these patients.

 

PROCEDURE: An intravenous line is placed 30 min before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). CRH is then injected IV at a dose of 1 µg/Kg up to a maximum of 200 µg. Blood is obtained at 15, 30, 60, 90, 120, 180 and 210 min for cortisol and ACTH (2 ml in a lavender top tube on ice).

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although the initial studies describing the test have been done in the morning.

 

Side effects: The patient may experience slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes. Category C in pregnancy.

 

INTERPRETATION OF RESULTS: The mean ACTH concentrations at 15 and 30 min after CRH should increase by at least 35% above the mean basal value at -15 and 0 min in patients with Cushing's disease, but not in patients with ectopic ACTH secretion. This measure gave the best sensitivity (93%) and specificity (100%) (40, 41).  The best cortisol criterion was a mean increase at 30 and 45 min of 20% or more above mean basal values, which gave a sensitivity of 91% and a specificity of 88%. It should be noted that the criterion for Cushing's disease is based on the presence of hypercortisolism. The CRH test will not adequately differentiate subjects with pseudo-Cushing’s and those with true pituitary dependent Cushing's disease.

 

CRH TEST WITH DEXAMETHSONE

 

WHEN TO USE THIS TEST: Several investigators have found that modifications of the CRH stimulation test can increase further the sensitivity and specificity in the diagnosis of the etiology of Cushing's disease. While the simultaneous use of vasopressin can augment the response to CRH, dexamethasone can be used to suppress all but pathologic responses to CRH stimulation [33]. Without dexamethasone the sensitivity and specificity of the CRH test is 65 and 100%, respectively, while with dexamethasone the CRH test is 100% sensitive and specific. This test is also particularly useful to differentiate true Cushing’s from pseudo-Cushing’s state.

 

PROCEDURE: Dexamethasone, 0.5 mg is self-administered orally by the patient every 6 hours for 2 days, at 6 AM, 12 Noon, 6 PM and midnight. On the morning of the 3rd day an additional dose of dexamethasone is given at 6 AM. The patient arrives at the testing center by 8 AM and an intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). CRH is then injected IV at a dose of 1 µg/Kg up to a maximum of 200 µg. Blood is obtained at 15, 30 60, 90 120, 180 and 210 min for cortisol and ACTH (2 ml in a lavender top tube on ice).

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although it is usually done in the morning.

 

Side effects that the patient may experience are: slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes.

 

Similar to the dexamethasone suppression test, the results should be interpreted with caution in patients taking estrogen therapy as they can present with falsely elevated cortisol levels due to an increase of cortisol-binding globulin. Drugs such as phenytoin, phenobarbitone, carbamazepine, rifampicin and alcohol induce hepatic enzymatic clearance of dexamethasone, mediated through CYP 3A4, thereby reducing the plasma concentration and may be associated with false positive results (42).

 

INTERPRETATION OF RESULTS: A normal response would be a plasma cortisol concentration less than 1.3 µg/dl measured 15 minutes after the administration of CRH.  Values of cortisol greater than 1.3 µg/dl correctly identified all cases of Cushing's syndrome and all cases of pseudo-Cushing's states (100% specificity, sensitivity, and diagnostic accuracy). While this is a general recommendation, each laboratory should confirm based on the sensitivity of the respective cortisol assay. Furthermore, it is important to confirm the serum level of dexamethasone at the time of the blood draw to assure patient compliance with the dexamethasone regimen.  Patients with ectopic ACTH production will have nonsuppressed cortisol and ACTH levels that are not stimulated by CRH.

 

DDAVP STIMULATION TEST

 

WHEN TO USE THIS TEST: This test can be used as part of the workup of ACTH dependent hypercortisolism. It can be used in addition to the CRH stimulation test as studies have shown that the combination of these two tests performs better than either of the tests separately. It can also be performed in lieu to the CRH test in situations in which CRH is not available. The aberrant expression of vasopressin V2 receptor in pituitary ACTH-secreting adenomas is the rationale for the use of the desmopressin test to differentiate corticotroph adenomas (which should respond to desmopressin injection) from ectopic ACTH secreting tumors or pseudo Cushing’s (which should not respond)(43-45).

 

PROCEDURE: An intravenous line is placed 30 minutes before the test for rapid phlebotomy and to eliminate a temporary rise in cortisol associated with a needle stick. Blood is drawn at -15' and 0' for cortisol and ACTH (2 ml in a lavender top tube on ice). DDAVP is then injected IV at a dose of 5 to 10 ug. Blood is obtained at 15, 30, 60, 90, 120, 180 and 210 minutes for cortisol and ACTH (2 ml in a lavender top tube on ice) (45).

 

INTERPRETATION OF RESULTS: No definitive cutoff values have been standardized for the interpretation of this test. The published established criteria for this test have generally been based on studies with small series of subjects. Malerbi et al. proposed a cortisol increase over baseline of 12% to be consistent with diagnosis of Cushing’s disease (46). An absolute ACTH increase over baseline equal or greater than 6 pmol/L yielded higher sensitivity and specificity to differentiate Cushing’s disease from pseudo Cushing’s in a different study. Alternatively the criteria used for the CRH stimulation test can be used in  the interpretation of the results (47).

 

INFERIOR PETROSAL SINUS SAMPLING (IPSS) WITH CRH STIMULATION

 

WHEN TO USE THIS TEST: Once the diagnosis of ACTH dependent Cushing's syndrome has been made based on endocrinologic testing, the next step in the evaluation of such patients should be an MRI of the pituitary to confirm the presence of a pituitary mass. Unfortunately, MRI imaging of the pituitary as a primary diagnostic tool is distinctly unhelpful due to the fact that 10% of all normal individuals may have slight abnormalities of their pituitary and that in many subjects with Cushing's disease, the tumor may be too small to be imaged with MRI scans. However, subjecting a patient to surgical pituitary exploration in the absence of a demonstrable mass is likely to result in an unsuccessful surgery. Furthermore, if previous dexamethasone and/or CRH testing is equivocal, then IPSS should be performed to further confirm the pituitary as the source of the ACTH (34). Although this test is less reliable in lateralizing the ACTH source (i.e., left versus right), than it is in confirming that the ACTH is central in origin, it can rule out ectopic ACTH production by a tumor (although ectopic CRH secreting tumors would be difficult to distinguish from true Cushings' disease based on IPSS). Simultaneous measurement of prolactin in the central samples can normalize the data if there is any difference in the location of the catheters (48).

 

It is recommended that active hypercortisolism is confirmed by measuring a 24-hour UFC or overnight UFC the day preceding IPSS. Misleading results have been reported when this test is performed “out of cycle” in patients with cyclical Cushing’s.

 

PROCEDURE: This test is done in conjunction with a skilled interventional neuroradiologist. It is important that the endocrinologist is personally present in the room during the procedure so that assurance can be made that the proper blood tests were drawn at the specified times. The patient is brought to the angiogram suite without sedation. A large bore IV line is placed in an antecubital fossa (to be certain there is access to peripheral blood sampling and CRH injection). Catheters (5 French) are placed in the femoral veins and threaded under fluoroscopic guidance to the inferior petrosal sinus. Injection of IV contrast confirms proper placement of the catheters.

 

Patients are on constant, pulse, blood pressure and oxygenation monitors during the course of the procedure. Test tubes are prechilled in ice and labeled so that during the rapid sampling period, blood can be placed in the tubes without delay.

 

It is recommended to routinely obtain 4 baseline measurements at -15, -10, -5 and at 0 minutes. This allows for practice allowing proper coordination between the radiologists drawing blood from the IPSS and the individual drawing blood from the brachial vein. Appropriate amounts of blood should be removed to discard the dead space of the catheter (this varies depending on the size of the catheter used). 2 ml of blood is obtained in lavender top vacutainer tubes on ice for measurement of cortisol (on peripheral samples); ACTH and prolactin (on central samples).

 

At 0' CRH is then injected as described above for the peripheral CRH test. Alternatively, a combination of CRH and 10ug of desmopressin can be used, especially if the patient has had a negative response to a prior CRH test. If CRH is not available, IPSS can also be performed with desmopressin alone per the protocol described above. Blood is then sampled from both central and peripheral lines at 2', 5' 10' and 15'. After the 15' time point and right before the IPSS catheters are removed, repeat fluoroscopic localization of the catheters should be performed to confirm that there was no displacement during the sampling. However, sampling on peripheral blood may continue as described in the CRH test discussed above.

 

SPECIAL CONSIDERATIONS: The test can be performed at any time of the day, although it is usually done in the morning.

 

Side effects that the patient may experience are: slight nausea, metallic taste, urgency to urinate, a change in blood pressure (either increase or decrease), a change in heart rate, headaches, abdominal discomfort, facial flushing, and lightheadedness. These side effects are mild and last for only few minutes.

 

Patients greater than 300 pounds in weight may not be able to be supported by the standard fluoroscopic table. Furthermore, such large patients may have an abdominal pannus that precludes reasonable access to the femoral veins. In such instances the IPSS can be performed via catheters placed in the antecubital vein with the patient immobilized in the sitting position.

 

Strokes have been reported in the literature as a potential complication (36). To minimize this possibility, it is recommended that the catheters remain in the petrosal sinus for no more than 30 min.

 

Freeze/thawing can decrease the ACTH concentration (see above); therefore, we recommend that the samples be brought to the endocrine lab and analyzed within 24 hours with the plasma separated and kept on ice during this time. If the analysis is not possible within 24 hours, the samples should be aliquoted and frozen to minimize the amount of freeze/thawing.

 

INTERPRETATION OF RESULTS: Plasma ACTH values are normalized to the prolactin value in order to correct for possible different localization of the catheters, or movement of the catheters during the study. The post CRH ACTH/Prolactin value of the central catheters should be >2.1 fold the ACTH/Prolactin value of the peripheral sample. In most cases of pituitary dependent Cushing’s, the increase is > 5.0-fold. A central to peripheral ACTH gradient higher or equal to 2 before CRH administration or higher or equal to 3, 10 min after CRH infusion is considered diagnostic of a pituitary source of ACTH (Cushing's disease). Lateralization would mean that the ratio of the left to right side is >2.0. Frequently the ratio criteria can be met without the need for CRH stimulation, however, the diagnostic accuracy increases from 86% to 90% with CRH (37).

 

The workup of ACTH dependent Cushing’s to differentiate Cushing’s disease from ectopic ACTH source can be quite challenging and often times requires combination of different dynamic testing in addition to imaging that often ultimately led to costly and invasive diagnostic procedures such as IPSS to be able to establish an accurate diagnosis. A retrospective study involving 167 patients with Cushing’s disease and 27 patients with ectopic Cushing’s found that using thresholds of a cortisol increase >17% with an ACTH increase >37% during CRH test and a cortisol increase >18% with an ACTH increase >33% during desmopressin test, the combination of both tests gave 73% sensitivity and 98% PPV of Cushing’s disease. The PPV was 100% in patients with positive response to both tests, with a negative pituitary MRI and whole-body CT scan. The NPV was 100% in patients with negative response to both tests, with negative pituitary MRI and positive whole body CT scan. This combination of dynamic tests with imaging studies is proposed as an accurate, cost-effective diagnostic strategy for the workup of ACTH depended Cushing’s that might minimize the need for IPSS which can be invasive, costly and unavailable in all institutions (43)

 

ADRENAL VEIN SAMPLING

 

WHEN TO USE THIS TEST: Patients diagnosed with ACTH independent Cushing’s and found to have bilateral adrenal tumors on imaging pose a particular challenge to the clinician. The differential diagnosis in these cases includes unilateral cortisol secreting adenoma (or carcinoma) with contralateral non-functioning cortical adenoma, bilateral cortisol secreting adenomas, macronodular adrenal hyperplasia, and primary pigmented nodular adrenocortical disease. Adrenal vein sampling measuring cortisol can be very helpful in this scenario and give valuable information to elucidate the proper diagnosis and guide therapy.

 

PROCEDURE: This test is done in conjunction with a skilled interventional radiologist under sedation. The procedure is usually performed early morning after an overnight fast on the second day of either a low dose (0.5 mg orally every 6 hours) or high dose (2 mg orally every 6 hours) of dexamethasone administration. This eliminates the probability of endogenous ACTH secretion causing interference with the interpretation of autonomous adrenal gland cortisol secretion. The adrenal veins can be catheterized by the percutaneous femoral vein approach, the position of the catheter tip should be verified by venogram. Concentrations of cortisol and aldosterone should be measured in blood obtained from both adrenal veins and the external iliac vein (for the detection of peripheral venous concentrations) (49).

 

SPECIAL CONSIDERATIONS: Potential complications include thrombosis with subsequent infarction or hemorrhage adrenal insufficiency and hypertensive crisis, however these are rare (48).

 

The aldosterone concentrations are usually much higher on the right adrenal vein compared to the left, this is presumably due to the anatomy differences and the catheter proximity to the right adrenal medulla. For this reason, although plasma epinephrine is measured to confirm success of adrenal vein catheterization, it cannot be used to correct for blood sample dilution between the 2 adrenal veins. There have been few case reports in which aldosterone has been used for side-to-side dilution differences, however whether it can be used for this purpose remains unclear (49, 50).

 

INTERPRETATION: Catheterization of each adrenal vein can be considered successful if plasma aldosterone concentration in the adrenal vein exceeds peripheral venous concentration by more than 100 pg/ml. An adrenal-to-peripheral venous cortisol gradient greater than 6.5 can be considered consistent with a cortisol secreting adenoma. Lateralization can be determined by measuring the side-to-side cortisol gradient (high-side to low-side). A ration of 2.3 or greater is consistent with autonomous cortical secretion from predominately 1 adrenal gland (49).

 

IMAGING STUDIES IN THE HPA AXIS EVALUATION

 

The evaluation of the HPA axis function should always be approached through biochemical measurements. With few exceptions, imaging studies provide no information about hormonal function but can be very useful for the localization of tumors or lesions. Once a biochemical diagnosis of either deficiency or excess of glucocorticoid production has been established, imaging studies can complement and assists the hormonal evaluation, providing valuable information about etiology, prognosis, and management.

 

Pituitary Imaging

 

In the vast majority of cases of ACTH dependent Cushing’s syndrome (CS), the source of ACTH is in the pituitary (Cushing’s Disease), so performing imaging studies of the pituitary gland in this scenario to try to localize a tumor is appropriate. However given the high incidence of non-functioning pituitary adenomas in the general population (up to 10%) (51) and the increasing sensitivity of the high resolution imaging modalities available that can lead to false positive results, it is important to perform a thorough dynamic testing evaluation of each case and consider inferior petrosal sinus sampling if appropriate (see section above), before committing a patient to pituitary surgery. Adrenocorticotropic pituitary tumors represent about 10% of all pituitary tumors (52) and  ACTH-secreting adenomas are most commonly microadenomas (<1cm). In cases of macroadenoma, assessment of extrasellar extension including chiasmatic compression and cavernous sinus involvement is imperative (53).

 

The other scenario in which pituitary imaging is indicated and can be useful in the evaluation of the HPA axis function, is in patients diagnosed with secondary adrenal insufficiency who have no history of recent exogenous glucocorticoid exposure or any other clear explanation for the clinical presentation. In these cases, a mass lesion disrupting the HPA function should be suspected, especially if the patient presents with deficiencies of other pituitary hormones and/or elevated prolactin, as isolated adrenal insufficiency from a non-functioning tumor affecting the pituitary is very rare.

 

PITUITARY MRI

 

Magnetic resonance imaging (MRI) is the mainstay of pituitary assessment. MRI is more sensitive than computed tomography (CT) in detecting corticotroph adenomas, but still detects only about 50% of these tumors (54) and has a false positive rate of 12-19% (55, 56)]. Standard pituitary imaging protocols typically include thin-section (2 or 3 mm) of T1-weighted (w) spin echo sequences (SE) performed both in coronal and sagittal planes through the pituitary fossa, which are repeated after administration of intravenous gadolinium contrast medium, associated with a T2-weighted sequence in the coronal plane (57, 58). High spatial detail can be achieved by using thin slices, a fine matrix size and a small field of view focused on the pituitary (58). The classic MR features of a corticotroph adenoma include a less than 1 cm focal area of lesser enhancement on T1-w images following contrast administration, hyperintense or hypointense on T2-w images as compared with the normal pituitary gland, remodeling of the pituitary sella floor and deformity of the gland contour (59). Acquiring dynamic sequences in the first 1-2 minutes after contrast injection can increase the sensitivity (60), but this technique has not been unequivocally demonstrated to improve the usefulness of MR in Cushing’s (61). The use of three-dimensional (3D) spoiled gradient recalled acquisition in the steady state (SPGR) sequence allows for superior soft tissue contrast compared to conventional spin echo sequences, this technique can be further optimized with thin-slice imaging (<1mm) (58).  Compared to T1-w SE sequence, SPGR has been reported to increase sensitivity but also has a higher false positive rate (62, 63).

 

PITUITARY CT SCAN

 

Pituitary computed tomography (CT) scanning is less sensitive than MRI for the detection of pituitary adenomas (64)and it is usually reserved for those patients who cannot safely undergo brain MRI. Acquisition of 1 mm (or less) axial sections through the pituitary fossa with coronal reconstructions can be helpful in the assessment of macroadenomas (57). It is also very helpful preoperatively in patients planned for transsphenoidal pituitary surgery to delineate the bony anatomy (65)

 

Adrenal Gland Imaging

 

There are a couple of scenarios in which adrenal gland imaging plays a role in the evaluation of the HPA axis. It is indicated and particularly important in the evaluation of patients diagnosed with ACTH independent CS, which is most commonly caused by adrenocortical adenomas or carcinomas and less frequently bilateral micronodular and macronodular hyperplasia. It can also be considered in cases of primary adrenal insufficiency. Tumors in the adrenal are fairly common in humans, they have been found to be present in 3% of autopsies performed in persons older than 50 years of age (66) and have been reported to be incidentally discovered in up to 5% of cross-sectional abdominal imaging carried out for unrelated problems (67). Most of these incidentally found adrenal tumors are nonfunctioning, 10 to 15% secrete excess amounts of hormones (68) of these, adrenocortical tumors are the most common. On the basis of imaging characteristics alone, no distinction can be made between a benign hyperfunctioning and a non-functioning adenoma, and this can only be differentiated based on clinical and biochemical diagnosis. Adrenal carcinoma represents <10% of adrenal tumors, 30 to 40% of these are hyperfunctioning in adults (69).  There are multiple important imaging characteristics that can help differentiate benign adrenal adenomas from pheochromocytomas, adrenocortical carcinomas and metastasis, like percentage of lipid content, tumor size, homogeneity, border regularity, presence of calcifications, invasion of surrounding tissue, and lymph node enlargements (Table 2).

 

ADRENAL CT SCAN

 

Unenhanced thin- section CT scan followed by contrast-enhanced examination is the cornerstone of imaging of adrenal tumors. Unenhanced CT is important to provide density measurements of lesions (70). The rich intracytoplasmic fat in adenomas results in a low attenuation on nonenhanced CT. The Hounsfield (HU) scale is a semiquantitative method to measure radiograph attenuation. If an adrenal mass measures <10 HU on unenhanced CT, the likelihood that it is a benign adenoma is nearly 100% (71).  However up to 30% of benign adenomas might not contain large amounts of lipid and present with higher HU on nonenhanced CT scan. This is when measuring the contrast washout on delayed images is very useful. Ten minutes after the administration of contrast, an absolute medium washout of more than 50% has been reported to be close to a 100% sensitive and specific for benign adenoma (72). Non-adenomas include metastases, pheochromocytomas and carcinomas.

 

Adrenal carcinomas usually appear as a unilateral mass, >4 cm in size with an inhomogeneous appearance due to necrosis, hemorrhage, fibrosis, and calcification. Careful assessment of the draining venous structures is essential on imaging, together with identification of direct infiltration of adjacent viscera (57).

 

ADRENAL MRI

 

When lesions cannot be characterized adequately with CT, MRI evaluation (with T1 and T2-weighted sequences, chemical shift and fat-suppression refinements) can be sought. Adrenal adenomas usually show low homogeneous signal on T1-weighted images and a signal intensity equivalent or higher than the liver on T2-weighted images. Chemical shift imaging will readily identify the lipid rich adenomas with signal loss on the out-of-phase sequences (73). This loss of signal can be measured using the adreno-splenic-ratio (ASR) and the signal intensity index (SII). An ASR ratio of <70% has been shown to be highly specific for adenomas and has a 78% sensitivity. Using the SII, a minimum of 5% signal loss characterizes an adrenal adenoma with accuracy of 100% [61]. MRI can also be particularly useful to evaluate for local and distant invasion of adrenocortical carcinomas.

 

Primary pigmented nodular adrenocortical disease is a rare cause of Cushing’s syndrome that has a female predilection and may be familial or associated with Carney complex. On imaging the adrenal glands may appear normal or minimally hyperplastic with multiple, usually <5 mm, unilateral or bilateral benign cortical nodules. The adrenal nodules are macroscopically pigmented; they demonstrate a lower T1 and T2 signal intensity on MRI compared to surrounding atrophic cortical tissue. When nodules are 1-2 cm in size, there might be atrophy of the intervening cortex, which helps distinguish this condition from ACTH- dependent hyperplasia (57).

 

Another rare cause of Cushing’s syndrome is ACTH-independent macronodular adrenal hyperplasia, which has a male predilection. The imaging appearance of the adrenal glands is striking with massive bilateral adrenal enlargement, nodularity. and distortion of adrenal contour. Nodules can measure 1 to 5.5 cm. On MRI they are hypointense relative to liver on T1-w images and hyperintense or isointense in T2-w images. On chemical shift imaging, nodules lose signal intensity on out-of-phase due to their high lipid content (57).

 

OTHER ADRENAL IMAGING MODALITIES

 

Patients that harbor adrenal masses, which are not adequately characterized by CT or MRI, can be further evaluated with functional nuclear medicine modalities that include single photon emission computed tomography (SPECT) scintigraphy with various radionuclide tracers, and positron emission tomography (PET) scintigraphy with various radionuclides.  PET images provide a higher spatial resolution compared to SPECT (70).

 

PET scan with either Fluorodeoxyglucose (FDG) or 11C-metomidate (MTO) can be useful in selected cases to differentiate benign adrenal adenomas from adrenocortical carcinomas. An elevated uptake on the FDG scan correlates with high metabolic activity and raises the suspicion for malignancy (74) with high sensitivity and specificity (75). Limitations of this technique include physiological excretion of FDG into renal inflammatory system and high metabolic uptake in inflammatory and infectious processes as well as in benign pheochromocytomas, leading to false positive results (64). Metomidate is an inhibitor of 11 beta-hydroxylase (CYP11B1) and aldosterone synthetase (CYP11B2), and based on this property its use can help differentiate tumors of adrenocortical origin from non-cortical lesions. Originally developed as a PET imaging agent radiolabeled with 11C, more recently it has been labeled with 18F and 123I, allowing SPECT and SPECT/CT imaging (76).

 

Integrated or “fused” PET-CT imaging allows to combine CT attenuation measurements with the intensity of FDG uptake, as described by the standardized uptake value (SUV), improving the performance of either imaging technique alone (77).

 

Scintigraphy with Iodine-131-Iodomethyl-19-norcholesterol (NP 59) is a functional nuclear medicine imaging modality that can be used to differentiate adrenal cortical adenomas from carcinomas. This is a labeled cholesterol analogue that specifically binds to low-density lipoproteins and after receptor-mediated uptake it is stored in the adrenocortical cells (70).  NP 59 uptake is regulated by ACTH and suppressed by dexamethasone, concentrating in hyperfunctioning cortisol and aldosterone secreting adenomas and showing low uptake in adrenocortical carcinomas because of the inefficient concentration of radiotracer by malignant tissue (78).

 

Other Imaging Modalities for Ectopic Cushing’s

 

Patients diagnosed with ACTH dependent Cushing’s whose biochemical dynamic tests suggests an ectopic source, pose a special challenge to the clinician. In 12 to 20% of these patients, the source remains undiscovered despite repeated biochemical and radiological investigations (55).

 

In the setting of ectopic ACTH production, imaging studies play a crucial role in trying to identify the source of the tumor causing the disease and guide management and prognosis. The optimal imaging study to detect these tumors has not been defined. CT, MRI, PET scan, 111In-pentetreotide (OCT) scintigraphy at conventional or higher radionuclide doses, as well as newer molecular imaging techniques like 131I/123-metaiodobenzylguanidine (MIBG), 18F-fluoro-2-2-deoxyglucose-positron emission tomography (FDG-PET), 18F-fluorodopa-PET (F-DOPA-PET), 68Ga-DOTATATE-PET/CT or 68Ga-DOTATOC-PET/CT scan (68Gallium-SSTR-PET/CT) are complementary and have been shown to be useful in different scenarios with variable sensitivity and specificity (79-83). For the most part at least two different imaging modalities are needed to establish a diagnosis and sometimes, repeated imaging over several months is required to identify the source. The choice of imaging modalities is guided by the sensitivity of the procedure balanced with the risk of false-positive findings (72).

 

A good approach is to start by obtaining images of the chest, since most ACTH-secreting tumors are located in this area. The most common causes are bronchial carcinoid tumors and small cell lung cancer. Other sources of excess ACTH production include neuroendocrine tumors of the thymus, bowel and pancreas, medullary carcinoma of the thyroid, pheochromocytomas, and mesotheliomas.

 

CT of the chest, abdomen and pelvis with intravenous contrast medium injection is the most commonly used initial imaging test performed and is very useful in many cases. In patients with equivocal CT imaging findings, MRI can be useful, particularly for tumors within the abdomen. It is recommended to follow CT and MRI imaging with a functional imaging modality, being OCT scintigraphy the most widely used. Functional imaging reduces false-positive results because it relies on the specific properties of tumor cells, not just their anatomic characteristics. However, tumors lacking the relevant receptor can have false negative results (83). Site-specific differences occur and different imaging modalities might have higher sensitivity and specificity depending on this. A recent systematic review showed that FDG-PET can be very sensitive in the detection of neuroendocrine tumors with high proliferation index, particularly in the pancreas. This review also showed that 68Gallium-SSTR-PET/CT had a 100% sensitivity but this is an imaging technique that has limited availability and was only performed in a minority of the patients in their series (80).

 

Table 2. Imaging Characteristics of Adrenal Tumors

Characteristic

Adenoma

Carcinoma

Pheochromocytoma

Metastasis

Size

<4 cm

>4 cm

Variable

>4 cm

Shape

Round

Irregular

Round

Irregular

Border

Smooth

Irregular

Well delineated

Irregular

Laterality

Unilateral

Unilateral

May be bilateral or unilateral

May be bilateral

Appearance

Round, homogeneous

Inhomogeneous with central necrosis. May have calcifications

Cystic and hemorrhagic changes.

Inhomogeneous

Vascularity

Normal

Increased

Increased

Increased

Growth rate

Slow (1 cm/year)

Fast (>2 cm/year)

Slow (0.5-1 cm/year)

Variable/Fast

Lipid content

Lipid rich or poor

Lipid poor

Lipid poor

Lipid poor

CT attenuation

 

<10 HU unenhanced.

>50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

>20 HU unenhanced.

<50% absolute washout.

MRI

Isointense with liver in T1 and T2-w.

Chemical shift

Hypointense compared to liver on T1-w

High to intermediate signal on T2-w

High signal intensity on T2-w

Hypointense compared to liver on T1-w

High to intermediate signal on T2-w

FDG-PET-CT

Low SUV

High SUV

Variable SUV

High SUV

Other

 

Evidence of invasion or metastasis

 

History of prior cancer

 

 REFERENCES

 

  1. Nieman LK, Biller BM, Findling JW, Newell-Price J, Savage MO, Stewart PM, Montori VM 2008 The diagnosis of Cushing's syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 93:1526-1540.
  2. Ceccato F, Barbot M, Zilio M, Frigo AC, Albiger N, Camozzi V, Antonelli G, Plebani M, Mantero F, Boscaro M, Scaroni C 2015 Screening Tests for Cushing's Syndrome: Urinary Free Cortisol Role Measured by LC-MS/MS. J Clin Endocrinol Metab 100:3856-3861.
  3. Elias PC, Martinez EZ, Barone BF, Mermejo LM, Castro M, Moreira AC 2014 Late-night salivary cortisol has a better performance than urinary free cortisol in the diagnosis of Cushing's syndrome. J Clin Endocrinol Metab 99:2045-2051.
  4. Pappachan JM, Hariman C, Edavalath M, Waldron J, Hanna FW 2017 Cushing's syndrome: a practical approach to diagnosis and differential diagnoses. J Clin Pathol 70:350-359.
  5. Cunningham JM, Buxton OM, Weiss RE 2002 Circadian variation in Cushing's disease and pseudo-Cushing states by analysis of F and ACTH pulsatility. J Endocrinol Invest 25:791-799.
  6. Bonte HA, van den Hoven RJ, van der Sluijs Veer G, Vermes I 1999 The use of free cortisol index for laboratory assessment of pituitary-adrenal function. Clin Chem Lab Med 37:127-132.
  7. Durber SM, Daly JR 1976 A simple method for the determination of free cortisol in plasma, with normal levels in men and women, including women taking low oestrogen contraceptives. Clin Chim Acta 68:43-48.
  8. McWhinney BC, Briscoe SE, Ungerer JP, Pretorius CJ 2010 Measurement of cortisol, cortisone, prednisolone, dexamethasone and 11-deoxycortisol with ultra high performance liquid chromatography-tandem mass spectrometry: Application for plasma, plasma ultrafiltrate, urine and saliva in a routine laboratory. J Chromatogr B Analyt Technol Biomed Life Sci 878:2863-2869.
  9. Aranda G, Careaga M, Hanzu FA, Patrascioiu I, Rios P, Mora M, Morales-Romero B, Jimenez W, Halperin I, Casals G 2016 Accuracy of immunoassay and mass spectrometry urinary free cortisol in the diagnosis of Cushing's syndrome. Pituitary 19:496-502.
  10. de Bos Kuil MJJ, Endert E, Fliers E, Prummel MF, Romijn JA, Wiersinga WM 1998 Establishment of reference values for endocrine tests.I:  Cushing's syndrome. Neth J Med 53:153-163.
  11. Erickson D, Singh RJ, Sathananthan A, Vella A, Bryant SC 2012 Late-night salivary cortisol for diagnosis of Cushing's syndrome by liquid chromatography/tandem mass spectrometry assay. Clin Endocrinol (Oxf) 76:467-472.
  12. Kannankeril J, Carroll T, Findling JW, Javorsky B, Gunsolus IL, Phillips J, Raff H 2020 Prospective Evaluation of Late-Night Salivary Cortisol and Cortisone by EIA and LC-MS/MS in Suspected Cushing Syndrome. J Endocr Soc 4:bvaa107.
  13. Raff H 2009 Utility of salivary cortisol measurements in Cushing's syndrome and adrenal insufficiency. J Clin Endocrinol Metab 94:3647-3655.
  14. Raff H 2013 Update on late-night salivary cortisol for the diagnosis of Cushing's syndrome: methodological considerations. Endocrine 44:346-349.
  15. Greene LW, Geer EB, Page-Wilson G, Findling JW, Raff H 2019 Assay-Specific Spurious ACTH Results Lead to Misdiagnosis, Unnecessary Testing, and Surgical Misadventure-A Case Series. J Endocr Soc 3:763-772.
  16. Morita K, Ogawa M, Kimura M, Okawa Y, Oki Y, Edo N, Tsukamoto K, Ishikawa T 2019 Falsely elevated plasma ACTH levels measured by the Elecsys assay related to heterophilic antibody in a case of secondary adrenocortical insufficiency. Endocr J 66:563-569.
  17. Husebye ES, Allolio B, Arlt W, Badenhoop K, Bensing S, Betterle C, Falorni A, Gan EH, Hulting AL, Kasperlik-Zaluska A, Kampe O, Lovas K, Meyer G, Pearce SH 2014 Consensus statement on the diagnosis, treatment and follow-up of patients with primary adrenal insufficiency. J Intern Med 275:104-115.
  18. Landon J, Greenwood FC, Stamp TC, Wynn V 1966 The plasma sugar, free fatty acid, cortisol, and growth hormone response to insulin, and the comparison of this procedure with other tests of pituitary and adrenal function. II. In patients with hypothalamic or pituitary dysfunction or anorexia nervosa. J Clin Invest 45:437-449.
  19. Dickstein G 2001 Hypothalamo-pituitary-adrenal axis testing:nothis is sacred and caution in interpretation is needed. Clin Endocrinol 54:15-16.
  20. Pfeifer M, Kanc K, Verhovec R, Kocijancic A 2001 Reproducibility of the insulin tolerance test (ITT) for assessment of growth hormone and cortisol secretion in normal and hypopituitary adult men. Clin Endocrinol (Oxf) 54:17-22.
  21. Hamrahian AH, Oseni TS, Arafah BM 2004 Measurements of serum free cortisol in critically ill patients. N Engl J Med 350:1629-1638.
  22. Klose M, Lange M, Rasmussen AK, Skakkebaek NE, Hilsted L, Haug E, Andersen M, Feldt-Rasmussen U 2007 Factors influencing the adrenocorticotropin test: role of contemporary cortisol assays, body composition, and oral contraceptive agents. J Clin Endocrinol Metab 92:1326-1333.
  23. Dokmetas HS, Colak R, Kelestimur F, Selcuklu A, Unluhizarci K, Bayram F 2000 A comparison between the 1-microg adrenocorticotropin (ACTH) test, the short ACTH (250 microg) test, and the insulin tolerance test in the assessment of hypothalamo-pituitary-adrenal axis immediately after pituitary surgery. J Clin Endocrinol Metab 85:3713-3719.
  24. Javorsky BR, Raff H, Carroll TB, Algeciras-Schimnich A, Singh RJ, Colon-Franco JM, Findling JW 2021 New Cutoffs for the Biochemical Diagnosis of Adrenal Insufficiency after ACTH Stimulation using Specific Cortisol Assays. J Endocr Soc 5:bvab022.
  25. Oilkers W, Diederich S, Bahr V 1992 Diagnosis and therapy surveillance in addison's disease:Rapid adrenocorticotropin (ACTH) test and measurement of plasma ACTH, renin activity, and aldosterone. J Clin Endocrinol Metab 75:259-264.
  26. Dickstein G 1997 Low-dose ACTH test- a word of caution to the word of caution:when ahd how to use it (Letter). J Clin Endocrinol Metab 82:322.
  27. Laureti S, Arvat E, Candeloro P, Di Vito L, Ghigo E, Santeusanio F, Falorni A 2000 Low dose (1 microg) ACTH test in the evaluation of adrenal dysfunction in pre-clinical Addison's disease. Clin Endocrinol (Oxf) 53:107-115.
  28. Kannisto S, Korppi M, Remes K, Voutilainen R 2000 Adrenal suppression, evaluated by a low dose adrenocorticotropin test, and growth in asthmatic children treated with inhaled steroids. J Clin Endocrinol Metab 85:652-657.
  29. Ospina NS, Al Nofal A, Bancos I, Javed A, Benkhadra K, Kapoor E, Lteif AN, Natt N, Murad MH 2016 ACTH Stimulation Tests for the Diagnosis of Adrenal Insufficiency: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 101:427-434.
  30. Dickstein G 2001 Peak cortisol response to insulin tolerance test: how reproducible is it really? Clin Endocrinol (Oxf) 55:819-820; author reply 820-811.
  31. Fiad TM, Kirby JM, Cunningham SK, McKenna TJ 1994 The overnight single-dose metyrapone test is a simple and reliable index of the hypothalamic-pituitary-axis. Clin Endocrinol 40:603-609.
  32. Jubiz W, Levinson RA, Meikle AW, West CD, Tyler FH 1970 Absorption and conjugation of metyrapone during diphenylhydantoin therapy:mechanism of the abnormal response to oral metyrapone. Endocrinology 86:328-331.
  33. Meikle AW, Jubiz W, Matsukura S, Harada G, West CD, Tyler FH 1970 Effect of estrogen on the metabolism of metyrapone and release of ACTH. J Clin Endocrinol Metab 30:259-263.
  34. Hindmarsh PC, Brook CGD 1985 Single dose dexamethasone suppression test in children:dose relationship to body size. Clin Endocrinol 23:67-.
  35. Mahajan DK, Wahlen JD, Tyler FH, West CD 1972 Plasma 11-deoxycortisol radioimmunoassay for metyrapone tests. Steroids 20:609.
  36. Al-Saadi N, Diederich S, Oelkers W 1998 A very high dose dexamethasone suppression test for the differential diagnosis of Cushing's syndrome. Clin Endocrinol 48:45-51.
  37. Alwani RA, Schmit Jongbloed LW, de Jong FH, van der Lely AJ, de Herder WW, Feelders RA 2014 Differentiating between Cushing's disease and pseudo-Cushing's syndrome: comparison of four tests. Eur J Endocrinol 170:477-486.
  38. Chabre O 2018 The difficulties of pseudo-Cushing's syndrome (or "non-neoplastic hypercortisolism"). Ann Endocrinol (Paris) 79:138-145.
  39. Yanovski JA, Cutler GB, Jr., Chrousos GP, Nieman LK 1998 The dexamethasone-suppressed corticotropin-releasing hormone stimulation test differentiates mild Cushing's disease from normal physiology. J Clin Endocrinol Metab 83:348-352.
  40. Chrousos GP, Schulte HM, Oldfield EH, Gold PW, Cutler GB, Jr., Loriaux DL 1984 The corticotropin-releasing factor stimulation test.An aid in the evaluation of patient's with Cushing's syndrome. N Engl J Med 310:622-626.
  41. Nieman LK, Oldfield EH, Wesley R, Chrousos GP, Loriaux DL, Cutler GB, Jr. 1993 A simplified morning ovine corticotropin-releasing hormone stimulation test for the differential diagnosis of adrenocorticotropin-dependent Cushing's syndrome. J Clin Endocrinol Metab 77:1308-1312.
  42. Kyriazopoulou V, Vagenakis AG 1992 Abnormal overnight dexamethasone suppression test in subjects receiving rifampicin therapy. J Clin Endocrinol Metab 75:315-317.
  43. Frete C, Corcuff JB, Kuhn E, Salenave S, Gaye D, Young J, Chanson P, Tabarin A 2020 Non-invasive Diagnostic Strategy in ACTH-dependent Cushing's Syndrome. J Clin Endocrinol Metab 105.
  44. Newell-Price J, Perry L, Medbak S, Monson J, Savage M, Besser M, Grossman A 1997 A combined test using desmopressin and corticotropin-releasing hormone in the differential diagnosis of Cushing's syndrome. J Clin Endocrinol Metab 82:176-181.
  45. Vassiliadi DA, Tsagarakis S 2018 DIAGNOSIS OF ENDOCRINE DISEASE: The role of the desmopressin test in the diagnosis and follow-up of Cushing's syndrome. Eur J Endocrinol 178:R201-R214.
  46. Malerbi DA, Mendonca BB, Liberman B, Toledo SP, Corradini MC, Cunha-Neto MB, Fragoso MC, Wajchenberg BL 1993 The desmopressin stimulation test in the differential diagnosis of Cushing's syndrome. Clin Endocrinol (Oxf) 38:463-472.
  47. Moro M, Putignano P, Losa M, Invitti C, Maraschini C, Cavagnini F 2000 The desmopressin test in the differential diagnosis between Cushing's disease and pseudo-Cushing states. J Clin Endocrinol Metab 85:3569-3574.
  48. Manni A, Latshaw RF, Page R, Santen RJ 1983 Simultanoues bilateral venous sampling for adrenocorticotropin in pituitary-dependent Cushing's disease:Evidence for lateralization of pituitary venous drainage. J Clin Endocrinol Metab 57:1070-1073.
  49. Wiggam MI, Heaney AP, McIlrath EM, McCance DR, Sheridan B, Hadden DR, Atkinson AB 2000 Bilateral inferior petrosal sinus sampling in the differential diganosis of adrenocorticotropin-dependent Cushing's syndrome:A comparison with other diagnostic tests. J Clin Endocrinol Metab 85:1525-1532.
  50. Obuobie K, Davies JS, DOgunko A, Scanlon MF 2000 Venous thrombo-emoblism following inferior petrosal sinus sampling in Cushing's disease. J Enocrinol Invest 23:542-544.
  51. Buurman H, Saeger W 2006 Subclinical adenomas in postmortem pituitaries: classification and correlations to clinical data. Eur J Endocrinol 154:753-758.
  52. Osamura RY, Kajiya H, Takei M, Egashira N, Tobita M, Takekoshi S, Teramoto A 2008 Pathology of the human pituitary adenomas. Histochem Cell Biol 130:495-507.
  53. Woo YS, Isidori AM, Wat WZ, Kaltsas GA, Afshar F, Sabin I, Jenkins PJ, Monson JP, Besser GM, Grossman AB 2005 Clinical and biochemical characteristics of adrenocorticotropin-secreting macroadenomas. J Clin Endocrinol Metab 90:4963-4969.
  54. Hall WA, Luciano MG, Doppman JL, Patronas NJ, Oldfield EH 1994 Pituitary magnetic resonance imaging in normal human volunteers: occult adenomas in the general population. Ann Intern Med 120:817-820.
  55. Ilias I, Torpy DJ, Pacak K, Mullen N, Wesley RA, Nieman LK 2005 Cushing's syndrome due to ectopic corticotropin secretion: twenty years' experience at the National Institutes of Health. J Clin Endocrinol Metab 90:4955-4962.
  56. Testa RM, Albiger N, Occhi G, Sanguin F, Scanarini M, Berlucchi S, Gardiman MP, Carollo C, Mantero F, Scaroni C 2007 The usefulness of combined biochemical tests in the diagnosis of Cushing's disease with negative pituitary magnetic resonance imaging. Eur J Endocrinol 156:241-248.
  57. Sahdev A, Reznek RH, Evanson J, Grossman AB 2007 Imaging in Cushing's syndrome. Arq Bras Endocrinol Metabol 51:1319-1328.
  58. Vitale G, Tortora F, Baldelli R, Cocchiara F, Paragliola RM, Sbardella E, Simeoli C, Caranci F, Pivonello R, Colao A, Group ABC 2017 Pituitary magnetic resonance imaging in Cushing's disease. Endocrine 55:691-696.
  59. Colombo N, Loli P, Vignati F, Scialfa G 1994 MR of corticotropin-secreting pituitary microadenomas. AJNR Am J Neuroradiol 15:1591-1595.
  60. Friedman TC, Zuckerbraun E, Lee ML, Kabil MS, Shahinian H 2007 Dynamic pituitary MRI has high sensitivity and specificity for the diagnosis of mild Cushing's syndrome and should be part of the initial workup. Horm Metab Res 39:451-456.
  61. Tabarin A, Laurent F, Catargi B, Olivier-Puel F, Lescene R, Berge J, Galli FS, Drouillard J, Roger P, Guerin J 1998 Comparative evaluation of conventional and dynamic magnetic resonance imaging of the pituitary gland for the diagnosis of Cushing's disease. Clin Endocrinol (Oxf) 49:293-300.
  62. Kasaliwal R, Sankhe SS, Lila AR, Budyal SR, Jagtap VS, Sarathi V, Kakade H, Bandgar T, Menon PS, Shah NS 2013 Volume interpolated 3D-spoiled gradient echo sequence is better than dynamic contrast spin echo sequence for MRI detection of corticotropin secreting pituitary microadenomas. Clin Endocrinol (Oxf) 78:825-830.
  63. Patronas N, Bulakbasi N, Stratakis CA, Lafferty A, Oldfield EH, Doppman J, Nieman LK 2003 Spoiled gradient recalled acquisition in the steady state technique is superior to conventional postcontrast spin echo technique for magnetic resonance imaging detection of adrenocorticotropin-secreting pituitary tumors. J Clin Endocrinol Metab 88:1565-1569.
  64. Escourolle H, Abecassis JP, Bertagna X, Guilhaume B, Pariente D, Derome P, Bonnin A, Luton JP 1993 Comparison of computerized tomography and magnetic resonance imaging for the examination of the pituitary gland in patients with Cushing's disease. Clin Endocrinol (Oxf) 39:307-313.
  65. Faje A, Tritos NA, Swearingen B, Klibanski A 2016 Neuroendocrine disorders: pituitary imaging. Handb Clin Neurol 136:873-885.
  66. Grumbach MM, Biller BM, Braunstein GD, Campbell KK, Carney JA, Godley PA, Harris EL, Lee JK, Oertel YC, Posner MC, Schlechte JA, Wieand HS 2003 Management of the clinically inapparent adrenal mass ("incidentaloma"). Ann Intern Med 138:424-429.
  67. Bovio S, Cataldi A, Reimondo G, Sperone P, Novello S, Berruti A, Borasio P, Fava C, Dogliotti L, Scagliotti GV, Angeli A, Terzolo M 2006 Prevalence of adrenal incidentaloma in a contemporary computerized tomography series. J Endocrinol Invest 29:298-302.
  68. Cawood TJ, Hunt PJ, O'Shea D, Cole D, Soule S 2009 Recommended evaluation of adrenal incidentalomas is costly, has high false-positive rates and confers a risk of fatal cancer that is similar to the risk of the adrenal lesion becoming malignant; time for a rethink? Eur J Endocrinol 161:513-527.
  69. Loh HH, Yee A, Loh HS, Sukor N, Kamaruddin NA 2017 The natural progression and outcomes of adrenal incidentaloma: a systematic review and meta-analysis. Minerva Endocrinol 42:77-87.
  70. Ilias I, Sahdev A, Reznek RH, Grossman AB, Pacak K 2007 The optimal imaging of adrenal tumours: a comparison of different methods. Endocr Relat Cancer 14:587-599.
  71. Dinnes J, Bancos I, Ferrante di Ruffano L, Chortis V, Davenport C, Bayliss S, Sahdev A, Guest P, Fassnacht M, Deeks JJ, Arlt W 2016 MANAGEMENT OF ENDOCRINE DISEASE: Imaging for the diagnosis of malignancy in incidentally discovered adrenal masses: a systematic review and meta-analysis. Eur J Endocrinol 175:R51-64.
  72. Hamrahian AH, Ioachimescu AG, Remer EM, Motta-Ramirez G, Bogabathina H, Levin HS, Reddy S, Gill IS, Siperstein A, Bravo EL 2005 Clinical utility of noncontrast computed tomography attenuation value (hounsfield units) to differentiate adrenal adenomas/hyperplasias from nonadenomas: Cleveland Clinic experience. J Clin Endocrinol Metab 90:871-877.
  73. Rockall AG, Babar SA, Sohaib SA, Isidori AM, Diaz-Cano S, Monson JP, Grossman AB, Reznek RH 2004 CT and MR imaging of the adrenal glands in ACTH-independent cushing syndrome. Radiographics 24:435-452.
  74. Groussin L, Bonardel G, Silvera S, Tissier F, Coste J, Abiven G, Libe R, Bienvenu M, Alberini JL, Salenave S, Bouchard P, Bertherat J, Dousset B, Legmann P, Richard B, Foehrenbach H, Bertagna X, Tenenbaum F 2009 18F-Fluorodeoxyglucose positron emission tomography for the diagnosis of adrenocortical tumors: a prospective study in 77 operated patients. J Clin Endocrinol Metab 94:1713-1722.
  75. Boland GW, Dwamena BA, Jagtiani Sangwaiya M, Goehler AG, Blake MA, Hahn PF, Scott JA, Kalra MK 2011 Characterization of adrenal masses by using FDG PET: a systematic review and meta-analysis of diagnostic test performance. Radiology 259:117-126.
  76. Hahner S, Stuermer A, Kreissl M, Reiners C, Fassnacht M, Haenscheid H, Beuschlein F, Zink M, Lang K, Allolio B, Schirbel A 2008 [123 I]Iodometomidate for molecular imaging of adrenocortical cytochrome P450 family 11B enzymes. J Clin Endocrinol Metab 93:2358-2365.
  77. Metser U, Miller E, Lerman H, Lievshitz G, Avital S, Even-Sapir E 2006 18F-FDG PET/CT in the evaluation of adrenal masses. J Nucl Med 47:32-37.
  78. Wong KK, Miller BS, Viglianti BL, Dwamena BA, Gauger PG, Cook GJ, Colletti PM, Rubello D, Gross MD 2016 Molecular Imaging in the Management of Adrenocortical Cancer: A Systematic Review. Clin Nucl Med 41:e368-382.
  79. de Herder WW, Krenning EP, Malchoff CD, Hofland LJ, Reubi JC, Kwekkeboom DJ, Oei HY, Pols HA, Bruining HA, Nobels FR, et al. 1994 Somatostatin receptor scintigraphy: its value in tumor localization in patients with Cushing's syndrome caused by ectopic corticotropin or corticotropin-releasing hormone secretion. Am J Med 96:305-312.
  80. Isidori AM, Sbardella E, Zatelli MC, Boschetti M, Vitale G, Colao A, Pivonello R, Group ABCS 2015 Conventional and Nuclear Medicine Imaging in Ectopic Cushing's Syndrome: A Systematic Review. J Clin Endocrinol Metab 100:3231-3244.
  81. Pacak K, Ilias I, Chen CC, Carrasquillo JA, Whatley M, Nieman LK 2004 The role of [(18)F]fluorodeoxyglucose positron emission tomography and [(111)In]-diethylenetriaminepentaacetate-D-Phe-pentetreotide scintigraphy in the localization of ectopic adrenocorticotropin-secreting tumors causing Cushing's syndrome. J Clin Endocrinol Metab 89:2214-2221.
  82. Santhanam P, Taieb D, Giovanella L, Treglia G 2015 PET imaging in ectopic Cushing syndrome: a systematic review. Endocrine 50:297-305.
  83. Zemskova MS, Gundabolu B, Sinaii N, Chen CC, Carrasquillo JA, Whatley M, Chowdhury I, Gharib AM, Nieman LK 2010 Utility of various functional and anatomic imaging modalities for detection of ectopic adrenocorticotropin-secreting tumors. J Clin Endocrinol Metab 95:1207-1219.

The Diabetic Foot

ABSTRACT

 

Diabetic foot ulcers (DFU) are associated with significant impairment of quality of life, increased morbidity and mortality, and are a huge drain on health care resources. In Western countries, the annual incidence of foot ulceration in the diabetic population is around 2%. DFUs develop as a consequence of a combination of factors, most commonly peripheral neuropathy (loss of the gift of pain), peripheral arterial disease (PAD), and some form of unperceived trauma. Recent studies emphasize the very high prevalence of foot ulceration in people with diabetes on dialysis as a consequence of end-stage renal disease. The mortality in this patient group is higher than for most forms of cancer. All patients with diabetes should have an annual screen to identify their foot ulcer risk status: those with any risk factors require specific foot care education as well as regular contact with a health care professional, usually a podiatrist. DFUs should heal if there is an adequate arterial inflow, infection is aggressively managed, and pressure is removed from the wound and its margins. In the management of plantar neuropathic ulcers, offloading is critical and all efforts must be made to enhance patient understanding of the need for offloading. Antibiotic usage should be guided by clinical signs of infection and microbiologic analysis of deep tissue specimens: evidence now exists to show that oral antibiotics are equally efficacious as intravenous in treating most cases of osteomyelitis in the diabetic foot. Most adjunctive therapies have little evidence to support their use although recent trials suggest efficacy for a number of topical therapies including LeucoPatch (3C patch) and sucrose octasulphate; and negative pressure wound therapy has also been shown to be helpful in certain cases. There is currently no indication for hyperbaric oxygen usage, whereas recent studies suggest that topical oxygen therapies help wound healing. Charcot neuroarthropathy (CN) should be easily preventable: most important is to treat any neuropathic patient with a warm swollen foot as having CN until proven otherwise.

 

INTRODUCTION

 

At the beginning of the 21st Century, diabetic foot problems, although eminently preventable, represent one of the commonest causes of hospital inpatient admission in Western countries. In 2005, the International Diabetes Federation realized the global importance of diabetic foot disease and chose to focus their campaign during the whole year on raising awareness with a worldwide campaign to “put feet first” and highlight the common problem of amputation amongst diabetic patients throughout the world. To coincide with World Diabetes Day 2005 (November 14, birth date of Frederick Banting), the Lancet elected to dedicate a whole issue to diabetic foot problems (1).

 

In this chapter, the global term “diabetic foot” refers to the variety of pathological conditions that might affect the feet in patients with diabetes. Foot ulcers are defined as lesions involving a skin break with loss of epithelium: they can extend into the dermis and deeper layers sometimes involving bone and muscle. Amputation is defined as “the removal of a terminal, non-viable portion of the limb”. The lifetime risk of a person with diabetes developing a foot ulcer (DFU) has been estimated to be as high as 34 % (2).

 

The suffering of affected individuals and the cost of DFUs are both equally staggering. Those individuals with DFUs usually have other complications of diabetes including nephropathy: data from the UK and the USA confirmed that the outlook for those people with foot complications who are on dialysis is very poor with a high mortality risk (1-3). Data from our group confirm that those people with diabetes who have had an amputation and who are on dialysis have a 75% two-year mortality; the majority of these were of cardiovascular etiology. Data such as these are worse than most malignant diseases, with the possible exception of lung and pancreas. There is therefore an urgent need for preventative strategies to reduce the incidence of foot complications amongst those with diabetes. With respect to costs, in 2008 Rogers et al (4) reported that in the US $18 billion was spent on the care of DFUs and US $11.7 billion on lower extremity amputations. More recently, data from the UK in 2019 suggest that a conservative estimate of the annual cost of diabetic foot problems exceeds UK £900 million which represents approximately 1% of the total budget of the National Health Service (5).

 

The importance of regular diabetic foot care in very high-risk patients is emphasized by an observational study from Arizona where the State decided to remove routine podiatry from high-risk patients to reduce their health budget. This led to an annual saving of US $351,000 but the cost of this action measured by increased hospitalization, length of stay, and amputations was $16.7 million (6).

 

This chapter will include a discussion on the epidemiology of foot problems including foot ulceration, amputations, and Charcot neuroarthropathy (CN). The etiopathogenesis will then be described and aspects of management of neuropathic, neuroischemic, and infected DFUs considered. The question of how to address primary and secondary prevention of diabetic foot problems will then be discussed followed by a section on Charcot neuroarthropathy. For more detailed discussion, the reader is referred to review articles on these topics (2,7-9).

 

EPIDEMIOLOGY OF THE DIABETIC FOOT

 

The study of the epidemiology of diabetic foot disease has been beset by numerous problems relating to both diagnostic tests used, and population selected. However, there is little doubt that foot complications are common. In the UK, the North West Diabetes Foot Care Study (a community-based study of over 15,000 people) reported that the annual incidence of foot problems amongst the population with diabetes was just under 2% (10), with similar results having been reported from the Netherlands. Similarly, when discussing amputations, the figures vary widely again due to diagnostic criteria as well as regional differences. It must be remembered that many individuals at diagnosis of type 2 diabetes have significant neuropathy: in the United Kingdom Prospective Diabetes Study, for example, 13% of patients at diagnosis had neuropathy of sufficient severity to put them at risk for foot ulceration (7).

 

With respect to ethnicity, studies from the UK suggest that foot ulcers and amputations appear to be less common in Asians of Indian sub-continental origin and Afro-Caribbean men. In contrast, reports from the USA suggest that amputation rates are more common amongst African-Americans with diabetes than amongst white Americans. Similarly, ulceration is much more common in Hispanic Americans and native Americans than in non-Hispanic whites (2). More recently, reviews have confirmed the importance of healthcare inequities in diabetic foot disease: race, ethnicity, socioeconomic status, and geography are powerful mediators of risk for DFU and lower-extremity amputation (2,11).

 

ETIOPATHOGENESIS OF DIABETIC FOOT ULCERATION

 

The foot does not break down spontaneously and in this section, the many warning signs that the feet are at risk of breakdown will be discussed. This was recognized by Elliott Joslin almost 90 years ago when he stated that “diabetic gangrene is not heaven-sent but is earth-borne” (12). It was previously believed that neuropathy, vascular disease, and infection were the main causes of ulceration: it is now recognized that infection occurs as a consequence of ulceration, and is not a cause thereof. There are many contributory factors to foot ulceration, the most important of which are diabetic neuropathy and peripheral arterial disease (PAD). These and other causative factors are listed in table 1.

 

Table 1. Risk Factors for Foot Ulceration

Peripheral neuropathy
Somatic
Autonomic

Peripheral arterial disease
Proximal and/or distal disease

Past history of foot ulcers/amputation

Other long-term complications
End-stage renal disease (especially on dialysis)
Post-transplant (including pancreas/kidney transplant)
Visual loss

Plantar callus

Elevated foot pressures

Foot deformity

Edema

Ethnic background

Poor social background

More common contributory factors shown in bold

 

Diabetic Neuropathy

 

Although the association between both somatic and autonomic neuropathy and foot ulceration has been recognized for many years, it is only in the last 20 years that prospective studies have confirmed these assumptions (2,8,10). It has been reported that the risk of developing the first foot ulcer is seven-fold higher in those with moderate to severe sensory loss compared with non-neuropathic diabetic individuals (13). Additionally, poor balance and instability as a consequence of loss of proprioception have been confirmed and are also likely contributory factors not only to foot ulceration, but also to Charcot neuroarthropathy (CN) (2,7,14,15).

 

Sympathetic autonomic neuropathy in the lower extremity leads to reduced sweating and dry skin that is prone to crack and fissure, and as well, in the absence of PVD, to increased blood flow, arterio-venous shunting, and the warm foot.

 

As will be discussed later, simple clinical tests may be used to identify the high-risk neuropathic foot (16). Most important in the identification of the high-risk neuropathic foot is good clinical observation and removal of the shoes and socks, with careful inspection of the feet as part of the routine follow up of all patients with diabetes.

 

Peripheral Arterial Disease (PAD)

 

A two-center study of causal pathways to foot ulceration reported that peripheral ischemia was a causal component in the pathway to ulceration in 35% of cases (17). In many Western countries, there has been an increase in the percentage of foot ulceration in which ischemia is a contributory factor (18). It is well recognized that patients with diabetes are more prone to distal arterial disease, which may be associated with a poorer outcome.

 

A detailed discussion of PAD in diabetes is outside the scope of this chapter and readers are directed to reviews on this topic (1920). A large follow-up study from Australia has confirmed that the strongest predictors of development of PAD in type 2 diabetes include microvascular complications (particularly macroalbuminuria and photocoagulation for retinopathy (21)).

 

Other Risk Factors

 

Of all the risk factors for foot ulceration (table 1), the most important is a past history of ulceration and/or amputation (2). In some series, the annual recurrence rate is up to 50%.

 

Other Long-term Complications

 

Those with other late complications particularly nephropathy, have an increased ulcer risk. Visual disturbance as a consequence of retinopathy is a confirmed risk factor; it is easy to understand why this should be. Those patients with sensory loss, particularly large fiber dysfunction, have poor balance and rely on vision as a secondary protective factor. Thus, those who have had for example extensive laser therapy and also have loss of proprioception, are at great risk of foot injury particularly when walking on uneven surfaces and in the hours of darkness.

 

A strong association between end-stage renal disease and foot ulceration has been emphasized in a number of studies. The temporal association between the start of dialysis treatment and foot ulceration was first confirmed by Game et al (22). A study comprising patients from both the US and the UK subsequently reported a very high prevalence of foot pathology in patients on dialysis, with 46% of patients having past or present foot ulceration and 18% were already amputees (23). The same group later confirmed that being on dialysis is an independent risk factor for foot ulceration in patients with diabetes (3,24). As noted above, preliminary data from the same group suggests that those patients who have already undergone amputation and who are on dialysis have a two-year mortality of up to 75%.

 

It must also be remembered that patients post-renal transplant or even post-simultaneous pancreas-kidney (SPK) transplant remain at very high risk of developing foot complications. There have been a number of reports of both foot ulceration and Charcot neuroarthropathy occurring in patients post-SPK (25). Theoretically, such subjects are “non-diabetic” but they remain at high risk because they invariably have a dense sensorimotor and autonomic peripheral neuropathy. They should remain under annual review and be coded as ‘diabetes in remission’.

 

Plantar Callus

 

Plantar callus forms under weight-bearing areas as a consequence of the dry skin (autonomic neuropathy), insensitivity, and repetitive moderate stress from high foot pressures. Callus itself acts as a foreign body and can cause ulceration in the insensate foot.

 

Elevated Foot Pressures

 

Numerous studies have confirmed the contributory role that abnormal plantar pressures play in the pathogenesis of foot ulceration (127). Most studies used sophisticated techniques such as pedobarography to assess foot pressures, but these are not required in day-to-day clinical practice.

 

Foot Deformities

 

A combination of motor neuropathy, cheiro-arthropathy, and altered gait pressures is thought to result in the “high-risk” neuropathic foot with clawing of the toes, prominent metatarsal heads, high arch, and small muscle wasting.

 

Demographics

 

In Western countries, the male sex has been associated with a 1.6-fold increased risk of foot ulcers (10). There is an increased risk of foot ulceration with increasing age and duration of diabetes.

 

Psychosocial Factors

 

There have been a few studies of psychosocial factors in the pathway to foot ulceration and it appears that patients’ behavior is not driven by the abstract designation of being “at risk”; it is driven by patients’ perception of their risk (26) . Thus, if a patient does not believe or understand that a foot ulcer lies on the path from neuropathy to amputation, are they likely to follow educational advice on how to reduce neuropathic ulcers? Moreover, a prospective study has confirmed that depression predicts first, although not recurrent, diabetic foot ulcers (27) .

 

THE PATHWAY TO FOOT ULCERATION IN DIABETES

 

As discussed and outlined in Figure 1, the pathway to ulceration is indeed complex and involves an interaction of numerous factors. Whereas none of the factors listed in the last section will alone result in ulceration, it is the interaction and combination of risk factors working together that leads to skin breakdown. In the prospective study of Reiber et al, 63% of all foot ulcers resulted from a combination of neuropathy, deformity, and trauma: in Western countries, the commonest cause of trauma is ill-fitting footwear (17). It must be remembered that as those with neuropathy have reduced sensory input, they will commonly be unable to feel the fit of a shoe until the pressure from the shoe is quite high. Thus, people with neuropathy frequently choose shoes that are too small. All such individuals should be advised to have their feet measured prior to the purchase of any “off the shelf” footwear.

 

Other simple examples of two risk factors working together in the pathway to ulceration are neuropathy and mechanical trauma (a common scenario is a neuropathic individual with a foreign body in the shoe), neuropathy and thermal trauma (holidays are particularly dangerous), and neuropathy and chemical trauma (such as inappropriate use of over-the-counter chemical corn treatments which should never be used in those with neuropathy).

 

In summary, whereas neuropathy was present in four out of five cases of new foot ulcers in the Reiber study (17), as noted above, the combination of neuropathy and ischemia is becoming more common in Western countries, and neuro-ischemic ulcers are the commonest type seen in 2023 in diabetic foot clinics.

 

FOOT ULCERATION

 

DFUs are common, associated with much morbidity and even mortality but should be eminently preventable. It used to be believed that diabetic foot ulcers were difficult to heal: this is not true: a foot ulcer will heal if it is permitted to do so and this requires attention to three factors-

A That there is adequate arterial inflow to the foot.

B That any infection is appropriately and aggressively managed.

C That all pressure is removed from the wound and its margins.

 

 

Figure 1. Pathways to Diabetic Foot Ulceration.

 

Despite increased knowledge of the pathogenesis and treatment of diabetic foot ulcers in recent years, it is still the third point, offloading the wound, that is poorly adhered to by health care professionals. Many forget that those with a neuropathic or neuroischemic ulcer have “lost the gift of pain”. That pain is a gift which is only realized when it is lost, as first described by Dr Paul Brand when studying leprosy (28). However, before going into more detail on management, it is important to classify wounds appropriately in order to guide therapeutic management.

 

CLASSIFICATION OF DIABETIC FOOT WOUNDS

 

Accurate and concise ulcer description and classification systems are required to improve multidisciplinary collaboration and communication, as well as for aiding treatment choices. For many years, the Meggitt-Wagner grading system was regarded as the gold standard. One problem with this system is that the ischemic status of the wound is not included. Thus, a number of new classification systems for diabetic foot wounds have been proposed and validated over the last 20 years. A commonly used system in the United States is the University of Texas Wound Classification System (29). This incorporates the Meggitt-Wagner grades but also enables the practitioner to stage the wound with respect to the presence or absence of infection and/or ischemia (Figure 2). In a comparative prospective study across two Centers, one in the UK and one in the US, the University of Texas Classification System was shown to be superior to the Meggitt-Wagner system at predicting outcomes (30). However, this study also showed that the traditional Meggitt-Wagner system was itself generally accurate in predicting outcomes.

 

Most recently, the WIFI (Wound Ischemia, Foot Infection) classification was introduced and is the most commonly used today in the USA, particularly in vascular clinics. This was developed and validated as a method to assess three variables – the wound, level of ischemia and the presence and severity of foot infection – to predict the risk of amputation (Figure 3)

 

Figure 2. The University of Texas Wound Classification System.

Figure 3. WIFI system. Wound, Ischemia, and Foot Infection (WIfI) Classification of Limb Threating diabetic foot disease, tissue loss, ischemia, and infection frequently overlap. However, one is frequently more dominant than the other at different times in the life cycle of an acute-on-chronic event. Here, the amount of tissue loss, ischemia, and foot infection can be ordinally graded to help predict outcome and assist in communicating a plan of action. aA higher score on the WIfI scale is associated with lower extremity amputation and morbidity and can be used to determine the need for revascularization. WIfI scores of 1, 2, 3, and 4 were associated with 1-year amputation rates of 0%, 8%, 11%, and 38%, respectively. Figure from JAMA 2023 Jul 3;330(1):62-75 with permission.

 

EVALUATION OF THE DIABETIC FOOT ULCER

 

Clinical evaluation of the foot wound should include a detailed description of the site, size, and depth of the wound. The neuropathic and vascular status of the wound should then be assessed (for details see below). In general, neuropathic ulcers typically occur in the warm but insensate foot, often under pressure bearing areas, and are surrounded by callus. In contrast, ischemic wounds tend to occur in the cool, poorly perfused foot, and are often at lateral fifth metatarsal head regions or the medial first metatarsal head regions. In a predominantly ischemic wound, callus tissue is uncommon. In a neuroischemic wound, the morphology will depend upon the predominance of each of these two pathologies. The correct identification of the degree of ischemia is of the utmost importance when evaluating a wound. If the foot is cool with impalpable pulses, then non-invasive Doppler ultrasound studies are indicated. Conventional methods of assessing tissue perfusion in the peripheral circulation may not be entirely reliable in patients with diabetes. For example, the Ankle Brachial Pressure Index, which is routinely used to screen for PAD in individuals without diabetes, may well be falsely elevated in the those with diabetes because of medial arterial calcification. Toe pressure indices may therefore be more reliable.

 

Peripheral Arterial Disease

 

A detailed discussion of vascular procedures is outside the scope of this review, although any person being considered for radiological or surgical procedures will require arteriography. Care must be taken in the use of certain contrast media in patients with chronic renal disease. A detailed discussion of the use of bedside investigations to diagnose PAD in people with diabetes is provided in the recently published guidelines by Fitridge et al (31).

 

Is Infection Present?

 

The correct diagnosis of infection in the diabetic foot wound is critical as it is often the combination of untreated infection and PAD that lead to amputation in the diabetic foot. A systematic review that was updated in 2020 still recommend that the diagnosis of infection requiring treatment is a clinical one (32). However, appropriate tissue specimens should be sent to the microbiological laboratory for culture and sensitivity. Superficial swabs are of little use: deep tissue specimens or if osteomyelitis is suspected, bone biopsies are recommended (32) .

 

A high index of suspicion for the presence of osteomyelitis is essential when assessing the diabetic foot wound. The “probe to bone” (PTB) is often used to diagnose osteomyelitis although there has been much discussion about its accuracy. A systematic review concluded that the PTB test can accurately diagnose osteomyelitis in high-risk patients, and rule out osteomyelitis in low-risk patients (33).

 

Role of Plain X-Ray in Diagnosing Osteomyelitis

 

The plain radiograph remains the commonest first radiological investigation of an acutely presenting diabetic foot problem. Despite this, it may be dismissed because of relatively low sensitivity for acute osteomyelitis, with literature over the last 10 years concentrating on CT scanning, MR scanning, and nuclear medicine studies (particularly Gallium Citrate, labelled leucocyte scans and recently PET, PET-CT, SPECT-CT and PET-MR). These latter studies are of limited availability and are expensive, and some carry a high radiation burden. They have their own sensitivity and specificity problems and may not be available in a timely manner. The initial sensitivity of the plain radiograph for acute osteomyelitis is improved by serial studies at one to two-week intervals, during which time therapy for presumed osteomyelitis may be instigated for clinical reasons and whilst awaiting the results of further “high tech” imaging (if still required). The plain radiographic findings could then be considered of high sensitivity and specificity, but with a two-week lag, both for diagnosis and for response to treatment. Appropriate clinical information for the reporting radiologist must include that the patient is diabetic, whether the foot is neuropathic, whether an ulcer is present and if so, its precise anatomical location, and whether it probes to bone. The radiologist should be aware that most sites of acute osteomyelitis in the diabetic foot occur in the floor of an ulcer that probes to bone and that if the foot is neuropathic there may be acute fractures without a history of trauma or acute Charcot neuroarthropathy may be present.

 

Whilst periosteal reaction is an early feature of osteomyelitis, it is not commonly seen around the small bones of the foot, and if present, is most often seen around metatarsals, and may be due to fracture rather than osteomyelitis.

 

The hallmark plain radiographic feature of osteomyelitis in the diabetic foot is focal loss of bone density, almost invariably adjacent to the floor of an ulcer. Whilst sometimes described as bone destruction, it is initially bone de-mineralization that causes this appearance, which can reverse on successful treatment, with radiographic re-appearance of the apparently destroyed bone (Figure 4). Obtaining the radiographic view most likely to demonstrate the bone in the floor of an ulcer is therefore an important consideration, often overlooked now that requests are electronic and radiographic views are selected from limited drop-down menus. For example, toe-tip ulcers and ulcers on the dorsum of the inter-phalangeal joints require lateral toe views - best obtained using dental radiographs if available; the inferior surfaces of metatarsal heads are best demonstrated on sesamoid views; the heel requires both lateral and axial views. As a general rule, radiographs tangential to the bone surface at the site of suspected osteomyelitis are ideal, in addition to the standard radiographs of the region. A dedicated team of radiographers familiar with these requirements will improve the relevance and quality of the resultant radiographs.

 

Plain radiology remains an important investigation in the diagnosis and management of diabetic foot osteomyelitis, but it needs to be of high quality, with appropriate views, and regularly repeated to fulfil its potential.

 

Figure 4. Acute presentation with an ulcer at the tip of the great toe, probing to bone. The terminal phalangeal tuft does show some irregularity (left panel). B) two weeks later there is marked bone demineralization consistent with osteomyelitis (middle panel). C) After 2 months of treatment there has been partial remineralization of the bone but with an underlying pathological fracture (right panel).

 

MANAGEMENT OF DIABETIC FOOT ULCERS

 

The principles of management of different types of foot ulcers will be discussed in brief in this section. The University of Texas Wound Classification System (Figure 2) will be used throughout.

 

Neuropathic Plantar Ulcers (UT 1A, 1B, 2A, 2B)

 

As noted above, neuropathic ulcers tend to occur under pressure areas, particularly at the plantar surface of the forefoot. Other recognized sites include the dorsal areas of the toes, particularly the distal inter-phalangeal joint if there is clawing of the toes. In patients with marked deformities such as those caused by Charcot neuroarthropathy, ulcers may occur at other pressure points, particularly in the plantar mid-foot due to, for example, a dropped cuboid bone. When lecturing on the management of neuropathic diabetic foot problems, one is often asked “what can one put on the wound to heal it?”. The answer is invariably that one should be asking “what should one take off the foot to help heal the ulcer?”. Thus, the management of a plantar neuropathic foot ulcer that is not infected is firstly sharp debridement of the ulcer down to bleeding healthy tissue with removal of all callus tissue over the wound and the edge, and secondly, the removal of pressure from the wound while the person is walking. Pain sensation normally protects wounds from further damage causing the non-neuropathic individual to limp. Any subject with a plantar ulcer who walks into the clinic without limping must, by definition, have loss of pain sensation. A neuropathic individual with a plantar ulcer will therefore walk on the ulcer as there is no warning symptom to inform him or her otherwise. Techniques for removing pressure include the use of casts (either removable or irremovable), boots, half shoes, sandals and felted foam dressings. The total contact cast (TCC) is regarded as the gold standard. Studies that randomize patients to an irremovable TCC, a removable cast walker (RCW), or other offloading devices invariably confirm that healing is fastest in the irremovable device (2,7). Although RCWs and irremovable casts (such as the TCC) offload equally well in the gait laboratory, the irremovable device is always associated with more rapid healing in clinical practice. The problem is that those with neuropathic foot ulcers have lost the sensory cue that tells them not to walk on their active ulcer. Studies suggest that individuals are compliant with wearing the offloading RCW during the day, but feel that home is safer and therefore tend to put slippers on, or even walk barefoot at home. A subsequent trial has confirmed that if the RCW is rendered irremovable by wrapping with scotch cast for example, then the outcome is that there is no difference in healing rates between the TCC and the RCW rendered irremovable (34) . Most people with simple neuropathic foot ulcers (UT grades 1A, 2A, 1B, 2B) generally heal in less than three months although of course this does vary with ulcer size. There is no contraindication to casting neuropathic individuals with mild foot infections (UT grades 2A, 2B). It is recommended that after the wound is healed, offloading should continue for a further four weeks to enable the scar tissue to firm up.

 

Wound dressings are important to keep the ulcer clean, but the placement of a large dressing on a wound may lead the person to a false sense of security by believing that dressing an ulcer is curative. Nothing could be further from the truth in the neuropathic ulcer. Unfortunately, there is little evidence from randomized controlled trials (RCTs) that any dressing is superior to another. Indeed, Jeffcoate et al (35)  randomized people to one of three dressings and could find no difference in outcome according to dressing used: the only difference was in cost. Thus, without an evidence-base, there is no indication to use some of the newer more expensive dressings.

 

Neuro-ischemic Ulcers

 

A neuro-ischemic ulcer is one occurring in a foot of a person who has both a neuropathic deficit and impaired arterial inflow: these would be classified UT 1C, 2C in the absence of infection, or 1D, 2D or 3D in the presence of infection. Such individuals warrant full vascular investigation as described above, and referral to the vascular surgery team. The principles of treatment are similar to those for neuropathic ulcers, and it has been confirmed that offloading can safely be used in non-infected neuro-ischemic ulcers under a weight-bearing area. However, antibiotics should be used if there is any suspicion of infection and casting only used with extreme caution in such cases (36). There is now evidence that one dressing, sucrose octasulphate, can improve the healing rates of neuroischemic ulcers in diabetic patients (for further details, see below under adjunctive treatments). With respect to the effectiveness of revascularization of the ulcerated foot in those with neuro-ischemic lesions, results showed that major outcomes following endovascular or open bypass surgery were similar amongst studies (37).

 

Management of Diabetic Foot Infections

 

Appropriate wound debridement and offloading together with antibiotics are important in the management of the infected neuropathic foot ulcer, although there are few data from randomized trials to guide the prescriber (32). There is however no evidence that clinically non-infected neuropathic ulcers warrant treatment with antibiotics. With respect to the choice of antibiotic therapy, the reader is directed to the helpful 2012 Infectious Diseases Society of America Clinical Practice Guideline (38).  Commonly used broad-spectrum antibiotics include Clindamycin, Cephalexin, Ciprofloxacin, and the Amoxycillin – Clavulanate potassium. Oral antibiotics usually suffice for mild infections, whereas more severe infections including cellulitis and osteomyelitis require intravenous antibiotic usage initially. Care should also be taken to optimize glycemic control, as hyperglycemia impairs leucocyte function.

 

The above statements on antibiotics refer to initial treatment: after starting with such broad-spectrum antibiotics, when the results of cultured deep tissue specimens are available, antibiotic therapy should be targeted at the likely primary infective organisms. Finally, with respect to duration of antibiotics, there are no data available from randomized trials to help guide the practitioner. Antibiotics should be continued until clinical signs of infection have resolved, but there is no indication to continue antibiotics beyond this period of time and certainly no indication to continue until the wound has healed. A recent review has identified the challenges facing us due to the increasing threat of multidrug-resistant pathogens (39) .

 

Osteomyelitis

 

Diagnosis of osteomyelitis has been discussed above both relating to the PTB test and also the use of plain radiographs. Although the treatment of osteomyelitis has traditionally been surgical, there is increasing evidence from case series and a RCT, that osteomyelitis localized to one or two bones, such as digits, may successfully be treated with antibiotics alone (40, 41). A randomized trial from Spain showed that antibiotics alone were not inferior to localized surgery (41). Again, with respect to duration of antibiotic therapy for osteomyelitis, there is no evidence-base to guide us though a recent trial suggests that six weeks’ antibiotic therapy for non-surgically treated diabetic foot osteomyelitis may be sufficient: traditionally, up to three months has been recommended (42). Lastly, many were surprised to read the results of the OVIVA (Oral Vs Intravenous Antibiotics) study (43) which randomized patients with osteomyelitis to oral vs intravenous antibiotics and showed no superiority of either delivery modality. These observations will certainly challenge the approach to osteomyelitis management in the future. A detailed updated review on infection management has been published by the American Diabetes Association in 2020 (44) .

 

Adjunctive Treatments

 

Adjunctive therapies are those which might be considered for complex diabetic foot wounds which fail to heal after 8-12 weeks of standard of care as discussed in the above sections. In recent years, many new such therapies, including skin substitutes, oxygen and other gases, products designed to correct abnormalities of wound biochemistry and cell biology associated with impaired wound healing, applications of cells, bioengineered skin and others, have been proposed to accelerate wound healing in the diabetic foot. Some years ago, an internationally conducted systemic review concluded that there was little published evidence from appropriately designed clinical trials to justify the use of such newer therapies (45).

 

However, there has been a renaissance in diabetic foot care with many RCTs of new therapies published since 2018 including topical therapies and oxygen-based treatments (46). A number of well-designed RCTs were published in 2018. The first proven therapy for neuro-ischemic ulcers, sucrose octasulfate dressings, was reported in the Explorer study (47). In the active group, 48% of wounds were closed after 20 weeks compared to 30% in the control dressing group (p<0.002). In the same issue of Lancet Diabetes Endocrinology, Game and colleagues reported the positive effect of the Leucopatch (3C Patch) device (a disc containing autologous platelets, leucocytes and fibrin) when applied to the surface of hard-to-heal foot ulcers (48).

 

Although, as noted above, the International Working Group on the Diabetic Foot (IWGDF) systematic review in 2016 (45) could not support the use of many of the therapies outlined above, this had changed by 2020 when three trials of placenta-derived products were considered (49). Although none was blinded, these were judged to be of low risk of bias as outcomes were assessed in a blinded manner. The first studied a cryo-preserved amniotic membrane allograft (50), the second an umbilical cord product (51), and the third a dehydrated amniotic membrane allograft (52): each showed significantly faster healing in the active treated group versus standard of care. Further details of all these studies referred to above can be found in the most recent American Diabetes Association (ADA) compendium on evidence-based management of complex diabetic foot wounds (53).

 

Hyperbaric and Topical Oxygen in the Diabetic Foot

 

HBO has been promoted as an effective treatment in diabetic foot wounds over many years (8). However, early RCTs have been criticized because of the small numbers of patients enrolled, and methodological and reporting inadequacies. A well designed and blinded RCT was conducted in Sweden some years ago suggesting the benefit of HBO in chronic neuro-ischemic infected foot ulcers with no possibility of revascularization (54). More recently, there have been two negative studies including a large retrospective cohort trial (55) and a multi-center Canadian study that showed no benefits of HBO whatsoever in any patient group (56). Thus, at present, the use of HBO in any diabetic foot wound has few data to support its efficacy and the multi-center trial from the Netherlands was also negative (57). The use of HBO in diabetic foot wounds was the topic of a recent debate (58).

 

There has been increasing interest in the use of topical oxygen-based therapies in wound healing in recent years. Whereas the latest studies of HBO have been negative, there have been interesting developments in the use of devices delivering topical oxygen. There is now evidence that both continuous (59) and cyclical (60) topical wound therapy may improve wound healing rates. A number of more recent studies now support the use of cyclical topical oxygen therapy (TWO2) (53) including some ‘real world’ data (61) and a number of meta-analyses and systematic reviews, the most recent of which has just been published (62). Thus, there is a body of evidence to support the use of TWO2 in the management of hard-to-heal diabetic foot ulcers that fail to respond to standard of care.

 

Negative Pressure Wound Therapy (NPWT)

 

The application of NPWT is believed to accelerate healing through reducing edema, removal of exudate, increased perfusion, self-proliferation, and the formation of granulation tissue (63). RCTs have suggested efficacy in rates of wound healing and reduced amputations, with the application of NPWT in both post-surgical and non-surgical chronic non-healing ulcers (64,65). A systematic review confirmed that there was some evidence to support the use of NPWT in post-operative wounds (49).

 

 ADA Standards of Care and IWDGF Guidelines 2023

 

The ADA publishes its standards of care and clinical practice guidelines each January in Diabetes Care. In 2023 (66), those adjunctive therapies for foot ulceration recommended and supported by level A evidence (based on large, well-designed randomized controlled trials or well-done meta-analyses of randomized controlled trials) included: negative-pressure wound therapy, placental membranes, bioengineered skin substitutes, several acellular matrices, autologous fibrin and leukocyte platelet patches, and topical oxygen therapy. The IWDGF guidelines are renewed every four years, and in 2023, for adjunctive therapies for foot ulceration, recommended, with variable levels of strength and certainty of evidence, that the following might be considered (67): - regular sharp debridement (strength of recommendation: strong), sucrose-octasulfate impregnated dressings, hyperbaric oxygen in neuro-ischemic or ischemic diabetes-related foot ulcers, topical oxygen therapy, the autologous leucocyte, platelet and fibrin patch (Leucopatch or 3C Patch), placental derived products and Negative Pressure Wound Therapy (only as an adjunct therapy to standard of care for the healing of postsurgical diabetes-related foot wounds).

 

CHARCOT NEUROARTHROPATHY (CN)

 

Charcot neuroarthropathy, although uncommon, is a potentially devastating late complication of diabetic neuropathy (68). Although the exact mechanisms resulting in the development of CN remain unclear, much progress has been made in our understanding of the etiopathogenesis of this disorder over the last two decades. CN occurs in a well-perfused foot with both somatic and autonomic neuropathy: the patient presenting with acute CN tends to be slightly younger than is usual for those presenting with foot ulcers. A history of trauma may be present though may be missed because of the severe sensory loss. Although, in its pathogenesis, there are many unanswered questions, improved understanding in recent years of the role of inflammatory pathways might lead to new pharmacologic approaches in the acute phase of the condition. The outcomes in terms of management of CN have been generally poor because of ignorance that leads to delayed diagnosis.

 

Most important in the management of this condition is recognition of the acute Charcot foot. Any patient with known neuropathy who presents with a warm, swollen foot of unknown causation should be presumed to have acute CN until proven otherwise. Contrary to earlier reports, many patients may present with painful, difficult to describe symptoms in the affected foot despite significant neuropathy.

 

In its early stages, all investigations may be normal, including the foot x-ray. The role of the radiologist in the diagnosis of acute and chronic CN is discussed in the next section.

 

Role of Radiologist in in Diagnosing CN

 

As with acute osteomyelitis (see above), the initial radiographs in acute CN may appear (almost) normal, though it is common for soft tissue swelling to be present and radiographically visible, usually over the dorsum of the foot. It is consequently imperative that both the clinician and the radiologist are aware of the possibility of this condition being present. The first more specific radiographic feature is bone demineralization, usually subchondral or periarticular, around the joint(s) involved by the acute CN process (in contrast to acute osteomyelitis, where it is related to the ulcer location). Focal peri-articular fractures may then develop (Figure 5). If CN is suspected, despite non-diagnostic initial radiographs, then the options are to treat as acute CN (see below) and perform serial radiographs at one-to-two-week intervals until the diagnosis is confirmed or no longer clinically suspected, or treat similarly whilst arranging urgent radiological investigation with a more sensitive test (whilst repeating the radiographs if the further tests are delayed). CT scanning may show small avulsion fractures around midfoot articulations that are invisible on plain radiographs, with minimal increase in the sensitivity and specificity over the plain radiograph, but MR scanning (to include fat suppressed sequences) is better, demonstrating soft tissue edema, bone marrow edema and/or ligamentous disruption. If the MR scan shows no marrow signal abnormality in the foot, acute CN is unlikely. Where the appearances or clinical presentation are complex, with both osteomyelitis and acute CN being suspected, Indium labelled white cell scans and PET/CT have a role, though both can be false positive for osteomyelitis in the presence of acute CN. In infection, MR may demonstrate soft tissue abscesses or sinus tracks that may extend to the (infected) bone surface.

 

In chronic inactive CN, plain radiographs demonstrate the features of joint distension, destruction, dislocation, disorganization, debris, increased bone density (sclerosis) and deformity. On MR scanning, marrow edema of acute CN is replaced by low signal from sclerosis of the bone. Acute osteomyelitis superimposed on chronic CN produces a mixed picture requiring careful clinical-radiological review.

 

Diagnosis of acute Charcot neuroarthropathy remains a synthesis of high clinical awareness, clinical findings and radiological findings. The latter should always include serial plain radiography and, where necessary, MR scans.

 

An overview of imaging in the Charcot foot is available online (69).

 

Figure 5. Acute Charcot neuroarthropathy. There is widening of the interosseous distance between the medial cuneiform and 2nd metatarsal (arrowheads), indicating disruption of the Lis-Franc ligament and a subtle flake fracture fragment (arrow).

 

Management of Charcot Neuroarthropathy

 

The treatment of CN depends upon the stage during which it is diagnosed. The essence of treatment in the acute phase remains non-weight bearing immobilization in a total contact or below-knee cast. Duration of treatment will depend upon response and it is recommended to continue casting until the temperature differential between the active and non-affected foot is down to approximately 1.5°C. As for the foot ulcer, it is recommended that treatment in a cast be continued for up to 4 weeks after the temperature differential has settled. At present, there are no proven medical or pharmacological approaches other than casting that have been shown to improve outcome. The management of advanced CN with bone deformity requiring reconstructive surgery is beyond the scope of this chapter and the reader is referred to a detailed review (70).

 

PREVENTION OF FIRST AND RECURRENT ULCERS

 

Prevention will only be successful with the early identification of those patients who have risk factors for foot ulceration. In the 1990s, the concept of the “annual review” was developed, and all those with diabetes should, at whatever stage, be screened for evidence of complications at least annually. The principle aim of such a review is to identify those with early signs of complications and institute appropriate management to prevent progression. The “Comprehensive Diabetic Foot Examination” (CDFE), was developed by a taskforce of the American Diabetes Association (ADA) that was charged with describing what should be included in the annual review for those at risk of foot complications (16). As noted above, the most important aspect of the annual foot review is the removal of shoes and socks with very careful inspection of both feet including between toes. Many neuropathic feet can be identified by this simple clinical observation, looking for features such as small muscle wasting, clawing of the toes, prominence of the metatarsal heads, distended dorsal foot pains (a sign of sympathetic autonomic neuropathy), dry skin, and callus formation. The key components of the diabetic foot annual examination are displayed in table 2.

 

The ADA Taskforce recommended that for evidence of neuropathy, that the perception of pressure using the 10g monofilament should be used at four sites in each foot (16). An additional test which might include a vibrating 128 Hz tuning fork or others outlined in table 2 should also be used to confirm any abnormality.

 

For the vascular assessment, foot pulse palpation is most important. Again, as noted above, the ankle brachial index may be falsely elevated in many people with diabetic neuropathy and therefore listening to the Doppler signal may be more helpful as may be a more detailed non-invasive vascular assessment.

 

More recently, other simple devices for clinical screening have been described. The simplest of all is the “Ipswich Touch Test” developed by Rayman et al in Ipswich, UK. This test simply assesses the ability of the patient to perceive the touch of a finger on the toes (71). The Vibratip which is a battery-operated disposable vibrating stylus can also be used to assess vibration sensation (72), and this has the advantage of using a forced-choice methodology. Both of these tests have been validated in clinical studies (71, 72).

 

Table 2. Key Components of the Diabetic Foot Exam. Adapted from Boulton (16)

Inspection
Evidence of past/present ulcers?
Foot shape?
Prominent metatarsal heads/claw toes
Hallux valgus
Muscle wasting
Charcot deformity
Dermatological?
Callus
Erythema
Sweating
Dystrophic nails

Neurological
10g monofilament at 4 sites on each foot + 1 of the following:
Vibration using 128 Hz tuning fork
Pinprick sensation
Ankle reflexes
Vibration perception threshold

Vascular
Foot pulses
Ankle Brachial Index, if indicated
Doppler wave forms, if indicated

 

Prevention of Diabetic Foot Ulcers

 

Surprisingly, there is no evidence from RCTs to confirm the efficacy of preventative foot care education either in the prevention of first foot ulcers or of recurrent foot ulceration (73). This, however, should be interpreted as lack of evidence rather than evidence of no effect. For those patients with no foot ulcer history found to have any of the risk factors listed above or in table 2, they require education in foot self-care and regular podiatric attention.

 

With respect to secondary prevention, a RCT that looked at the effect of a foot care education program in those with a history of foot ulcers could provide no evidence that such a program of targeted education led to clinical benefit when compared to the usual care (74). It seems likely that those with a history of foot ulcers have such predominant physical abnormalities (e.g., foot deformity, loss of sensation, etc.) that education alone in self-foot care management is insufficient to prevent recurrent ulceration. It may be the combination of foot care education and an intervention that the individual can perform may be more effective. Lavery and colleagues, in studies supported by other RCTs demonstrated in an RCT that patients with a history of neuropathic foot ulcers who were randomized for self-foot temperature monitoring did demonstrate a reduced recurrent ulceration rate. All patients in the active group received foot care education and were provided with a skin thermometer which they used twice a day to check the temperatures of both feet. Those patients who discovered increased unilateral foot temperatures were advised to stop walking and see their health care professional. In the active group there was a highly significant reduction in recurrent foot ulceration (75): however, not all subsequent studies have confirmed this observation (76).

 

Important in the prevention of foot complications in diabetes is the team approach: members of the team commonly include the diabetes specialists, orthopedic and vascular surgeons, podiatrists, nurse educators, physiotherapists, pedorthists, and others. A study from one district in the UK was able to confirm a 62% reduction in major amputations over an 11-year follow-up period: this decrease occurred after the establishment of such a multi-disciplinary diabetic foot care team (77).

 

One of the impacts of the recent Covid-19 pandemic has been an explosion in the use of telemedicine and remote monitoring in the care of the diabetic foot (78). A number of studies are currently ongoing looking at “smart technology” in the prevention of recurrent diabetic foot ulcers. These include the use of sensors in socks or shoes to detect pressure change and also various devices to measure differentials in skin temperature: each of these might alert patients in the pre-ulcerative phase with the hope of preventing the actual ulcer from developing. It has now clearly been confirmed that a temperature differential of 2.2C between the feet using remote at home monitoring in patients at high risk of plantar ulceration is a strong predictor of ulcer development (79). Similarly, intelligent pressure sensing insole systems can reduce the incidence of plantar ulcers in those with a past history of ulceration (80). However, face to face consultations remain crucial in the screening for PAD and neuropathy in people with diabetes.

 

The Foot in Remission

 

As a recurrence is so common after the healing of neuropathic or neuro-ischemic foot ulcers, it has been suggested that those with a history of foot ulcers should be described as having “a foot in remission” rather than healed. This might better communicate risk of recurrence not only to the patient, but also other healthcare professionals (8). It is hoped that, as in cancers, aggressive treatment during the active disease together with a focus on improving care in “remission” can help to maximize patients’ function and of course improve quality of life (8,26) .

 

CONCLUSIONS

 

Although there has been much progress in our understanding of the etiopathogenesis and management of diabetic foot disorders over the last 30 years, much of what we use in clinical practice today still lacks an evidence-base. This is particularly true for example for dressings. The International Working Group on the Diabetic Foot has reported on the details required in the planning and reporting of intervention studies in the prevention and management of diabetic foot lesions (81). Details of the necessary trial design, conduct, and reporting should be taken into account when assessing published studies on interventions in the diabetic foot. Most important of all however in the management of patients with diabetic foot disorders, is to remember that the patient has frequently lost the “gift of pain” that protects most of us from developing significant foot problems but, when absent, can lead to devastating consequences.

 

REFERENCES

 

  1. Boulton AJM, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J. The global burden of diabetic foot disease.  Lancet 2005; 366: 1719-24.
  2. Armstrong DG, Tan TW, Boulton AJM, Bus SA. Diabetic foot ulcers:  a review.  JAMA 2023; 330: 62-75. Doi: 10.1001/JAMA 2023.10578.
  3. Lavery LA, Hunt NA, Ndip A, et al. Impact of chronic kidney disease on survival after amputation in individuals with diabetes.  Diabetes Care 2010; 33: 2365-2369.
  4. Rogers LC, Lavery LA, Armstrong DG. The right to bear legs.  J Am Podiatr Med Assoc 2008; 98: 166-168.
  5. Kerr M, Barron E, Chadwick P, et al. The cost of diabetic foot ulcers and amputations to the National Health Service in England.  Diabetic Med 2019; 36: 995-1002.
  6. Skrepnek GH, Mills JL, Armstrong DG. Foot-in-Wallet Disease:  tripped up by “cost-saving” reductions?  Diabetes Care 2014; 37: 196-197.
  7. Boulton AJM. The diabetic lower limb – a forty-year journey: from clinical observation to clinical Science. Diabetic Medicine 2019; 36:1539-1549. Doi.org/10.1111/dme.13901.
  8. Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence.  New Engl J Med 2017; 376: 2367-2375.
  9. Zhang Y, Lazzarini PA, McPhail SM, et al. Global disability burdens of diabetes-related lower-extremity complications in 1990 and 2016.  Diabetes Care 2020; 43: 964-974. Doi: 10.2337/dc19-1614.  
  10. Abbott CA, Carrington AL, Ashe H, et al. The North-West Diabetes Foot Care Study:  incidence of, and risk factors for new diabetic foot ulceration in a community-based patient cohort.  Diabetic Med 2002; 19: 377-384.
  11. McDermott K, Fang M, Boulton AJM et al. Etiology, Epidemiology and Disparities in the Burden of Diabetic Foot Ulcers.  Diabetes Care 2023; 46: 209-221.  Doi: 10.2337/dci22-0043.
  12. Joslin EP. The menace of diabetic gangrene.  N Engl J Med 1934; 211: 16-20.
  13. Young MJ, Breddy JL, Veves A, et al. The prediction of diabetic neuropathic foot ulceration using vibration perception thresholds:  a prospective study.  Diabetes Care 1994; 17: 557-560.
  14. Brown SJ, Handsaker JC, Bowling FL, et al. Diabetic peripheral neuropathy compromises balance during daily activities.  Diabetes Care 2015; 38: 1116-22.
  15. Handsaker JC, Brown SJ, Bowling FL, et al. People with diabetic peripheral neuropathy display a decreased stepping accuracy during walking:  potential implications for risk of tripping.  Diabet Med 2016; 33: 644-649.
  16. Boulton AJM, Armstrong DG, Albert SF, et al. Comprehensive foot examination and risk assessment:  a report of the taskforce of the Foot Care Interest Group of the American Diabetes Association, with endorsement by the American Association of Clinical Endocrinologists.  Diabetes Care 2008; 31: 1679-1685.
  17. Reiber GE, Vileikyte L, Boyko EJ, et al. Causal pathways for incident lower-extremity ulcers in patients with diabetes from two settings.  Diabetes Care 1999; 22: 157-162.
  18. Prompers L, Huijberts M, Apelqvist J, et al. High prevalence of ischemia, infection and serious comorbidity in patients with diabetic foot disease in Europe.  Baseline results from the Eurodiale study.  Diabetologia 2007; 50: 18-25.
  19. Hinchliffe RJ, Forsythe RO, Apelqvist J, et al. Guidelines on the diagnosis, prognosis and management of peripheral artery disease in patients with foot ulcers and diabetes.  Diabetes Metab Res Rev 2020; 36 (suppl 1): e3276.doi10.1002/dmrr.3276.
  20. Forsythe RO, Hinchliffe RJ. Management of critical limb ischemia in the patient with diabetes.  J Cardiovasc Surg (Torino) 2016; 57: 273-281.
  21. Mohammedi K, Woodward M, Hirakawa Y, et al. Microvascular and macrovascular disease and risk for major peripheral arterial disease in patients with type 2 diabetes.  Diabetes Care 2016; 39: 1796-1803.
  22. Game FL, Chipchase SY, Hubbard R, et al. Temporal association between the incidence of foot ulceration and the start of dialysis in diabetes mellitus.  Nephrol Dial Transplant 2006; 21: 3207-3210.
  23. Ndip A, Lavery LA, Lafontaine J, et al. High levels of foot ulceration and amputation risk in a multiracial cohort of diabetic patients on dialysis therapy.  Diabetes Care 2010; 33: 878-880.
  24. Ndip A, Rutter MK, Vileikyte L, et al. Dialysis treatment is an independent risk factor for foot ulceration in patients with diabetes and stage 4 or 5 chronic kidney disease.  Diabetes Care 2010; 33: 1811-1866.
  25. Barrado FC, Kuypers DK, Matricali GA. Charcot neuroarthropathy after simultaneous pancreas-kidney transplantation:  risk factors, prevalence and outcome. Clin Transplant 2015; 29: 712-719.
  26. Vileikyte L, Pouwer F, Gonzalez JS. Psychosocial research in the diabetic foot:  are we making progress?  Diabetes Metab Res Rev 2020; 36 (suppl 1) e3257.doi10.1002/dmrr3257.
  27. Gonzalez JS, Vileikyte L, Ulbrecht JS, et al. Depression predicts first but not recurrent diabetic foot ulcers.  Diabetologia 2010; 53: 2241-2248.
  28. Boulton AJM. Diabetic Neuropathy: is pain God’s great gift to mankind?  Semin Vasc Surg 2012; 25: 61-65.
  29. Armstrong DG, Lavery LA, Harkless LB. Validation of a diabetic wound classification system.  The contribution of depth, infection, and ischemia to risk of amputation.  Diabetes Care 1998; 21: 855-859.
  30. Oyibo S, Jude EB, Tarawneh I, et al. A comparison of two diabetic foot ulcer classification systems:  the Wagner and the University of Texas wound classification system.  Diabetes Care 2001; 24: 84-88.
  31. Fitridge R, Chuter V, Mills JL et al.  . The 2023 intersocietal IWGDF, ESVS, SVS guidelines on peripheral arterial disease in patients with diabetes and a foot ulcer.  iwgdfguidelines.org. 2023 
  32. Senneville E, Lipsky BA, Abbas ZG, et al. Diagnosis of infection in the foot in diabetes:  a systematic review.  Diabetes Metab Res Rev 2020; 36 (suppl 1): e3281.doi:10.1002/dmrr3281.
  33. Lam K, van Asten SA, Nguyen T, et al. Diagnostic accuracy of probe to bone to detect osteomyelitis in the diabetic foot:  a systematic review.  Clin Infect Dis 2016 63: 944-948.
  34. Katz IA, Harlan A, Miranda-Palma B, et al. A randomized trial of two irremovable offloading devices in the management of plantar neuropathic diabetic foot ulcers.  Diabetes Care 2005; 28: 555-559.
  35. Jeffcoate WJ, Price PE, Phillips CJ, et al. Randomized controlled trial of the use of three dressing preparations in the management of chronic ulceration of the foot in diabetes.  Health Technol Assess 2009; 13: 1-86.
  36. Nabuurs-Fransen MH, Sleegers R, Huijberts HS, et al. Total contact casting of the diabetic foot in daily practice:  a prospective follow-up study.  Diabetes Care 2005; 28: 243-247.
  37. Hinchliffe RJ, Brownrigg JR, Andros G, et al. Effectiveness of revascularization of the ulcerated foot in patients with diabetes and peripheral artery disease:  a systematic review.  Diabetes Metab Res Rev 2016; 32 (suppl 1): 136-144.
  38. Lipsky BA, Berendt AR, Cornia PB, et al. 2012 Infectious Diseases Society of America clinical practice guideline for the diagnosis and treatment of diabetic foot infections.  Clin Infect Dis 2012; 54: e132-173.
  39. Ertugrul B, UcKay I, Schoni M, Peter-Riesch B, Lipsky BA. Management of diabetic foot infections in the light of recent literature and new international guidelines.  Expert Rev Anti-Infect Ther 2020; Feb 19 Epub.
  40. Game FL, Jeffcoate WJ. Primary non-surgical management of osteomyelitis of the foot   in diabetes.  Diabetologia 2008; 51: 962-967.
  41. Lázaro-Martinez JL, Aragón-Sánchez J, Garcia-Morales C. Antibiotics versus conservative surgery for treating diabetic foot osteomyelitis: a randomized comparative trial.  Diabetes Care 2014; 37: 789-795.
  42. Tone A, Nguyen S, Devemy F, et al. Six-week versus twelve-week antibiotic therapy for non-surgically treated diabetic foot osteomyelitis: a multicentre open-label controlled randomized study.  Diabetes Care 2015; 38: 302-307.
  43. Li HK, Rombach I, Zambellas R, et al. OVIVA Trial collaborators.  Oral Vs IntraVenous Antibiotics for bone and joint infection.  N Engl J Med 2019; 380: 425-436.
  44. Boulton AJM, Armstrong DG, Hardman MJ, et al. Diagnosis and management of diabetic foot infections.  Arlington (VA).  American Diabetes Association 2020.
  45. Game FL, Apelqvist J, Attinger C, et al. Effectiveness of interventions to enhance healing of chronic ulcers of the foot in diabetes:  a systematic review.  Diabetes Metab Res Rev 2016; 32 (suppl 1):  154-168.
  46. Edmonds ME. A renaissance in diabetic foot care:  new evidence-based treatments.  Lancet Diabet Endocrinol 2018; 6: 837-838.
  47. Edmonds ME, Lázaro-Martinez JL, Alfayate-Garcia JM, et al. Sucrose octasulfate dressing versus control dressing in patients with neuroischemic diabetic foot ulcers (Explorer):  an international, multicenter, double-blind, randomized controlled trial.  Lancet Diabetes Endocrinol 2018; 6: 186-196.
  48. Game F, Jeffcoate W, Tarnow L, et al. LeucoPatch system for the management of hard-to-heal diabetic foot ulcers in the UK, Denmark, and Sweden:  an observer-masked, randomized controlled trial.  Lancet Diabetes Endocrinol 2018; 6; 870-878.
  49. Rayman G, Vas P, Dharariya K, et al. International Working Group on the Diabetic Foot (IWGDF).  Guidelines on use of interventions to enhance healing of chronic foot ulcers in diabetes  (IWGDF 2019 update).  Diabetes Metab Res Rev 2020; 36 (suppl 1): 3283.
  50. Lavery LA, Fulmer J, Shebetka KA, et al. Grafix Diabetic Foot Ulcer Study Group.  The efficacy and safety of Grafix for the treatment of chronic diabetic foot ulcers:  results of a multi-centre, controlled, randomized, blinded, clinical trial.  Int Wound J 2014; 11: 554-560.
  51. Tettelbach W, Cazzell S, Sigal F, et al. A multicentre prospective randomized controlled comparative parallel study of dehydrated human umbilical word (EpiCord) allograft for the treatment of diabetic foot ulcers.  Int Wound J 2019; 16: 122-130.
  52. Tettelbach W, Cazzell S, Reyzelman AM, Sigal F, Caporusso JM, Agnew PS. A confirmatory study on the efficacy of dehydrated human amnion/chorion membrane dHACM allograft in the management of diabetic foot ulcers:  a prospective, multicentre, randomized, controlled study of 110 patients from 14 wound clinics.  Int Wound J 2019; 16: 19-29.
  53. Boulton AJM, Armstrong DG, Löndahl M, et al. New Evidence-Based Therapies for Complex Diabetic Foot Wounds. Arlington (VA) May 2022: American Diabetes Association. doi: 2337/db2022-02.
  54. Löndahl M, Katzman P, Nilsson A, et al. Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes.  Diabetes Care 2010; 33: 998-1003.
  55. Margolis DJ, Gupta J, Hoffstad O, et al. Lack of effectiveness of hyperbaric oxygen therapy for the treatment of diabetic foot ulcer and the prevention of amputation:  a cohort study.  Diabetes Care 2013; 36: 1961-1966.
  56. Fedorko L, Bowen JM, Jones W, et al. Hyperbaric oxygen therapy does not reduce indications for amputation in patients with diabetes with non-healing ulcers of the lower limb:  a prospective, double-blind, randomized controlled clinical trial.  Diabetes Care 2016; 39; 392-399.
  57. Santema KTB, Stoekenbroek RM, Koelemay MJW, et al. Hyperbaric oxygen therapy in the treatment of ischemic lower-extremity ulcers in patients with diabetes:  results of the DAMO2CLES multicenter randomized controlled trial.  Diabetes Care 2018; 41: 112-119.
  58. Löndahl M, Boulton AJM. Hyperbaric oxygen:  useless or useful?  A debate.  Diab Metab Res Rev 2020 Mar;36 Suppl 1:e3233. doi: 10.1002/dmrr.3233.
  59. Niederauer MQ, Michalek JE, Liu Q, et al. Continuous diffusion of oxygen improves diabetic foot ulcer healing when compared with a placebo control:  a randomized, double-blind, multicenter study.  J Wound Care 2018; 27 (suppl 9): S30-S45.
  60. Frykberg RG, Franks PJ, Edmonds ME, et al. A multinational, multicenter, randomized, double-blinded, placebo-controlled trial to evaluate the efficacy of cyclical topical wound oxygen therapy (TWO2) in the treatment of chronic diabetic foot ulcers:  the TWO2 study.  Diabetes Care 2020; 43: 616-624.
  61. Yellin JI, Gaebler JA, Zhou FF, et al. Reduced hospitalizations and amputations in patients with diabetic foot ulcers treated with cyclical pressurized topical wound oxygen therapy:  real-world outcomes.  Adv Wound Care (New Rochelle).  Online ahead of print on 6 December 2021 (doi: 10.1089/wound.2021.0118).
  62. Carter MJ, Frykberg RG, Oropallo A, et al. Efficacy of topical oxygen therapy in healing chronic diabetic foot ulcers:  systematic review and meta-analysis.  Adv Wound Care 2023, Apr; 12 (4): 177-186. doi: 10.1089/wound.2022.0041.
  63. Isaac AL, Armstrong DG. Negative pressure wound therapy and other new therapies for diabetic foot ulceration: the current state of play.  Med Clin North Amer 2013; 97: 899-909.
  64. Armstrong DG, Lavery LA. Negative pressure wound therapy after partial diabetic foot amputation:  a multicenter, randomized controlled trial.  Lancet 2005; 366: 1704-1710.
  65. Blume PA, Walters J, Payne W, et al. Comparison of negative pressure wound therapy using vacuum-assisted closure with advanced moist wound therapy in the treatment of diabetic foot ulcers:  a multicenter, randomized controlled trial.  Diabetes Care 2008; 31: 631-636.
  66. El Sayed NA, Aleppo G, Aroda VR, et al. Retinopathy, Neuropathy and Foot Care:  Standards of Care in Diabetes – 2023.  Diabetes Care 2023; 46 (Suppl 1): S203-S215.  doi: 10.2337/dc23-S012.
  67. Chen P, Vilorio NC, Dhatariya K, et al: Guidelines on interventions to enhance healing of foot ulcers in people with diabetes (IWGDF 2023 update).  Diab Metab Res Rev 2023; May 25; 3644 doi: 10.1002/dmrr.3644.
  68. Jeffcoate WJ. Charcot foot syndrome.  Diabetic Med 2015; 32: 760-770.
  69. Ergen FB, Sanverdi SE, Oznur A. Charcot foot in diabetes and an update on imaging.  Diabetic Foot Ankle 2013; 4: 10.3402/dfa.V4i0.21884.
  70. Shen W, Wukich D. Orthopedic surgery and the diabetic Charcot foot.  Med Clin North Am 2013; 97; 873-882.
  71. Rayman G, Vas PR, Baker N, et al. The Ipswich Touch Test:  a simple and novel method to identify inpatients with diabetes at risk of foot ulceration.  Diabetes Care 2011; 34: 1517-1518.
  72. Bowling FL, Abbott CA, Harris WE. A pocket-sized disposable device for testing the integrity of sensation in the outpatient setting.  Diabet Med 2012; 29: 1550-1552.
  73. Dorresteijn JA, Valk GD. Patient education for preventing diabetic foot ulceration.  Diabetes Metab Res Rev 2012; 28 (suppl 1): 101-106.
  74. Lincoln NB, Radford KA, Game FL, et al. Education for secondary prevention of foot ulcers in people with diabetes:  a randomized controlled trial.  Diabetologia 2008; 51 1954-1961.
  75. Lavery LA, Higgins KR, Lanctot DR, et al. Preventing diabetic foot ulcer recurrence in high-risk patients:  use of temperature monitoring as a self-assessment tool.  Diabetes Care 2007; 30: 14-20.
  76. Aan de Stegge WB, van Netten JJ, Bus SA. Does the skin heat up before it breaks down in diabetic foot ulceration?  Diabete Metab Res Rev 2023, 08 Feb 2023.  https://doi.org/10.1002/DMRR.3621.
  77. Krishnan S, Nash F, Baker N, et al. Reduction in diabetic amputations over eleven years in a defined UK population:  benefits of multidisciplinary teamwork and continuous prospective audit.  Diabetes Care 2008; 31: 99-101.
  78. Boulton AJM. Diabetic Foot Disease during the Covid-19 pandemic.  Medicina 2021; Jan 22; 57 (2): 97. Doi; 10.3390/medicina57020097.
  79. Frykberg RG, Gordon IL. Reyzelman AM, et al.  Feasibility and efficacy of a smart mat technology to predict development of diabetic plantar ulcers.  Diabetes Care 2017; 40: 973-980.doi: 10.2337/dc16-2294.
  80. Abbott CA, Chatwin KE, Foden P, et al. Innovative intelligent insole system reduces diabetic foot ulcer recurrence at plantar sites:  a prospective, randomized, proof-of-concept study.  Lancet Digit Health 2019; Oct 1 (6): e308-e318.
  81. Jeffcoate WJ, Bus SA, Game FL, et al. Reporting standards of studies and papers on the prevention and management of foot ulcers in diabetes:  required details and markers of good quality.  Lancet Diabetes Endocrinol 2016; 4: 781-788.

Osteoporosis and Bone Fragility in Children

ABSTRACT

 

Childhood is a unique time during which individuals accrue bone rapidly, and peak bone mass is achieved early in the third decade of life. Several factors may adversely influence bone accrual, including primary skeletal disorders as well as secondary causes of low bone density such as specific endocrinopathies, altered weight-bearing, and certain medications. Pediatric osteoporosis is defined by both: 1) a clinically significant fracture history; and 2) a low bone mineral density (BMD). Pragmatically, the diagnosis of osteoporosis is indicated by a BMD Z-score < -2.0 and a clinically significant fracture history, defined as two or more long bone fractures by age 10 years, or three or more long bone fractures at any age up to 19 years. Additionally, the finding of one or more vertebral non-traumatic compression fractures is diagnostic of osteoporosis independent of BMD. Notably, the diagnosis of pediatric osteoporosis should not be made based on densitometric criteria (i.e., DXA) alone. As childhood osteoporosis has several potential underlying etiologies, evaluation requires a careful assessment by a clinician with expertise in the possible mechanisms that may be contributing to the increased skeletal fragility. Both non-pharmacologic therapies as well as bone-active medications, such as bisphosphonates, increase bone mass and may lower the risk of fracture. The development of novel therapies that may restore physiologic anabolic bone activity in children with insufficient bone accrual from various causes has the potential to improve care for pediatric patients with osteoporosis. Prospective data acquisition to inform treatment strategies for primary prevention of fracture in children with osteoporosis, as is done in adult populations, is urgently needed to prevent the significant morbidity of fracture in this vulnerable population.

 

INTRODUCTION

 

Childhood and adolescence are unique time periods during which individuals accrue bone rapidly, and peak bone mass is only achieved early in the third decade of life. Several congenital or acquired factors may adversely influence bone accrual, including primary skeletal disorders, or secondary causes of low bone density such as delayed puberty, endocrinopathies, altered weight-bearing, and certain medications.

 

Pediatric osteoporosis is defined by both: 1) a clinically significant fracture history; and 2) a low bone mineral density (BMD). Pragmatically, the diagnosis of osteoporosis is indicated by a BMD Z-score < -2.0 and a clinically significant fracture history, defined as two or more long bone fractures by age 10 years, or three or more long bone fractures at any age up to 19 years. Additionally, the finding of one or more non-traumatic vertebral compression fractures is diagnostic of osteoporosis independent of BMD. Notably, the diagnosis of pediatric osteoporosis should not be made based only on densitometric criteria (i.e., DXA) (1).

 

ETIOLOGIES

 

Primary Causes of Low Bone Density or Osteoporosis

 

Genetic diversity overall accounts for 60-70% of the variability in bone mass, and numerous genes have been associated with bone density in genome-wide association studies and whole genome sequencing analyses (see Table 1). These include genes in the WNT signaling pathway (such as LRP4, LRP5, SOST, WNT4, WNT16, WLS), the osteoprotegerin-Receptor Activator of Nuclear factor Kappa-Β Ligand (RANKL) pathway (such as OPG, RANKL, RANK), TGFβ signaling (TGFBR3), mesenchymal stem cell differentiation pathway, endochondral ossification pathway (such as RUNX2), the pathway of collagen synthesis (such as COL1A1 and COL1A2) and other genes such as ESR1, CCDC170 (located adjacent to ESR1), VDR, and CALCR (2, 3). Further, several bone density associated loci have been associated with fracture risk, including FAM210A, SLC25A13, MEPE, SPTBN1, DKK1, LRP5, SOST, and  EN1(2-4).

 

In general, conditions that impair bone or connective tissue development can result in low bone density and increased fracture risk. Monogenic disorders leading to low bone density or osteoporosis include the various types of osteogenesis imperfecta, which are discussed in another chapter of Endotext (5) and will not be covered here. Marfan syndrome, caused by mutations in the gene coding for fibrillin-1 (FBN1), is a connective tissue disorder inherited in an autosomal dominant fashion. In addition to abnormalities in blood vessels, ligaments, muscles, and heart valves, the condition is associated with low bone density at multiple sites, particularly after adjusting for the taller height of these individuals, and with reduced bone accrual during adolescence, especially at the femoral neck (6-8). Similarly, certain forms of Ehlers-Danlos syndrome are associated with low bone density, such as those that result from mutations in COL5A1, COL5A2 and COL3A1 (8).  One study of adults with classical or hypermobility Ehlers-Danlos syndrome reported lower bone density, altered bone quality (as assessed by the trabecular bone score), and increased prevalence of morphometric vertebral fractures compared with controls (32% vs. 8% in controls) (9). Another study has reported lower bone density and strength estimates in individuals with hypermobile Ehlers-Danlos syndrome and generalized joint hypermobility spectrum disorder compared to controls (10). Other studies have reported similar data (11, 12). 

 

Low bone density is also observed in patients with homocystinuria with reported low bone density Z-scores in 38% of patients in one study (13) and lysinuric protein intolerance (14-16). High homocysteine levels have been demonstrated to have deleterious effects on osteoblasts and osteoclasts, to increase oxidative stress, disrupt cross-linking of collagen molecules, and increase levels of advanced glycation end products, all of which can reduce bone strength (17). Further, low lysine concentrations are related to growth failure and low bone density (18, 19).

 

Osteoporosis-pseudoglioma syndrome (OPPG) is a condition caused by homozygous or compound heterozygous inactivating mutations in the gene coding for low density lipoprotein related protein 5 (LRP5) (18-20). Wnt ligand binds to the Frizzled/LRP5 complex to activate the canonical Wnt signaling pathway and increase bone formation. Thus, inactivating mutations in LRP5 lead to severe juvenile osteoporosis. The condition is also associated with congenital vision loss that typically manifests in infancy and is a consequence of a spectrum of conditions ranging from phthisis bulbi to vitreoretinal dysplasia. Heterozygous carriers can have low bone density for age and gender norms, but do not demonstrate the eye findings (20, 21). 

 

Hypophosphatasia is a consequence of both recessive and dominant mutations in ALPL, the gene that codes for tissue nonspecific alkaline phosphatase (TNSALP), which is necessary for breaking down inorganic pyrophosphate in bone to enable bone mineralization (22). Reduced mineralization can lead to skeletal defects, altered growth plates resembling rickets, low bone density, and impaired cementum mineralization resulting in early loss of teeth. There are six major forms of hypophosphatasia ranging in severity from very severe (often associated with perinatal demise), to an infantile, juvenile, and adult form, to a very mild form that involves only the teeth (odontohypophosphatasia) and a rare form with normal ALP levels called pseudohypophosphatasia. The severity of the condition depends on the amount of ALP activity that results from the gene mutation.

 

Other primary conditions associated with fragility fractures but without low bone density include McCune-Albright syndrome, osteopetrosis, and pycnodysostosis (mutations in the gene coding for cathepsin-K). As these conditions are rarely associated with pediatric osteoporosis, they are not discussed further here.

 

Secondary Causes of Low Bone Density or Osteoporosis

 

NUTRITIONAL AND MALBSORPTIVE

 

Deficient Intake of Calcium and Vitamin D:

 

Insufficient intake of calcium and/or vitamin D can result in suboptimal bone mineralization, and the associated secondary hyperparathyroidism has the potential to cause deleterious effects on bone. However, data are conflicting regarding the impact of calcium or vitamin D deficiency and their replacement on bone mineral content (BMC) and BMD. One systematic review of dairy intake or calcium supplementation in children and adults 1-25 years old concluded that these measures provide no beneficial effect on bone mineralization or fracture risk (23). A meta-analysis of 21 randomized controlled trials (RCTs) found no significant change in total body BMC in those randomized to supplemental dairy or calcium alone regardless of baseline intake; however, the study did find an increase in whole body BMC in children with low baseline calcium intake who received high doses of dietary calcium supplements or dairy products with or without vitamin D supplementation (24). Another meta-analysis of 19 RCTs reported a small favorable effect on total body BMC and upper limb BMD with calcium supplementation, but no effect at the lumbar spine or femoral neck (25). A more recent meta-analysis of 15 RCTs and three non-randomized studies did find a positive impact of calcium supplementation on femoral neck BMD in children (26). Calcium supplementation has also been reported to have a beneficial effect on bone strength estimates in prepubertal and early pubertal children (27). Of note, maternal calcium supplementation during pregnancy has not been demonstrated to benefit offspring BMD even when baseline intake was low (28).  Overall, data suggest a possible small effect of dietary calcium or dairy supplementation on bone outcomes when baseline intake is low, with greatest effects at the whole body and femoral neck.

 

Similarly, data for the effects of vitamin D deficiency and supplementation are mixed, but overall suggest some effect on bone outcomes. One study reported that baseline 25-hydroxyvitamin D (25OHD) levels predict prospective changes in lumbar spine BMD over the next three years in peripubertal Finnish girls (29), and a case control study of 150 African American children 5-9 years old reported that those with forearm fractures were more likely to be vitamin D deficient (30). In Chinese adolescents, 25OHD levels 20-37 nmol/L (8-15 ng/mL) in girls and 33-39 nmol/L (13-16 ng/mL) in boys are reported to have positive effects on bone outcomes (31). The impact of race is interesting, with dark skinned children typically having lower 25OHD levels than light skinned children, but higher BMD measures. Further, data suggest that vitamin D supplementation in dark skinned (but not light skinned) children living in northern latitudes positively impact femoral neck BMC (32). Some data (but not all) suggest that maternal vitamin D status may predict offspring BMD, with low maternal 25OHD levels being concerning for low peak bone mass in their children (33). However, a meta-analysis of vitamin D supplementation during pregnancy and infancy reported no impact on subsequent bone health (34).

 

Conditions of Low Energy Availability or Energy Deficit

 

Conditions such as anorexia nervosa and the female athlete triad (Triad)/relative energy deficiency in sports (RED-S) are known to be associated with low BMD, reduced bone strength, and increased prevalence of fractures (35, 36), even in adolescents. Anorexia nervosa in both male and female adolescents is associated with low BMD (35, 37-39), and reduced bone accrual in adolescent girls with anorexia nervosa (40) raises significant concerns regarding peak bone mass acquisition. Young oligo-amenorrheic athletes have lower BMD than eumenorrheic athletes at the femoral neck and hip and lower strength estimates at the distal tibia; they also have lower spine BMD and lower strength estimates at the radius than non-athletes (41-43). Factors contributing to impaired bone outcomes include lower lean mass, lower BMI, hypogonadism, low levels of insulin like growth factor-1 (IGF-1), relatively high cortisol levels, and alterations in levels of appetite regulating hormones that also have an impact on bone health,  (such as insulin, leptin, peptide YY and oxytocin) (37, 44).

 

Conditions of Malabsorption

 

Conditions such as celiac disease, inflammatory bowel disease, cystic fibrosis, and biliary atresia are associated with malabsorption of essential nutrients, including vitamin D, that are important for optimizing bone health.

 

Celiac disease is associated with low BMD (45) and an increase in fracture risk (46). One meta-analysis reported that a lifetime history of bone fractures was twice as common in those with celiac disease versus controls, and that a baseline history of celiac disease is associated with a 30% increase in any fracture and 69% increase in spine fracture (46). The impact of celiac disease on bone health is related to a decrease in BMI and lean mass (in those who have poor weight gain or a decrease in body weight following diagnosis), malabsorption with reduced bioavailability of calcium and vitamin D, secondary hyperparathyroidism, an increase in intestinal production of inflammatory cytokines (IL-1β, IL-6 and TNF-α), and because of antibodies that may bind to bone tissue transglutaminase (47, 48). The institution of and adherence to a gluten free diet mitigates most of these factors.

 

Inflammatory bowel disease results in low bone mass in 30-50% of patients, who also demonstrate reduced rates of bone accrual during the adolescent years, resulting in compromised peak bone mass (49). Data for fractures are conflicting, with studies in children reporting modest to no increase in long bone fractures while studies in adults report a 32-40% increase in fracture risk (50-52); asymptomatic vertebral fractures are often missed in retrospective studies based on self-report. Some studies (but not all) report a higher risk of impaired bone health in children with Crohn’s disease (53, 54) compared to those with ulcerative colitis, likely because the former more commonly affects areas of the intestine responsible for absorption of vital nutrients and is more likely to be associated with use of glucocorticoids, but data are conflicting in this regard (55-57). In general, children with Crohn’s disease are most likely to present with growth and puberty delay. There are also conflicting data regarding whether or not bone compromise is more likely in boys (vs. girls) with inflammatory bowel disease (55, 57, 58).

 

In addition to malabsorption of vital nutrients, factors that contribute to low bone density and impaired bone accrual in inflammatory bowel disease include low BMI and reduced lean mass, associated pubertal delay and hypogonadism, poor nutritional intake, reduced physical activity, active inflammation (cytokine secretion by activated T-cells, and increased IFN-γ and TNF-α, which may inhibit osteoblastic activity and active osteoclasts both directly and via RANK ligand), and chronic use of glucocorticoids, which have anti-inflammatory effects (which helps with the condition) but affect bone metabolism at multiple steps (52-55, 59, 60). In general, the severity of disease correlates with the extent of bone compromise.

 

Cystic fibrosis results in low bone density in 30-60% of individuals with the condition, associated with increased fracture risk in adults (61, 62). A 100-fold increase in vertebral fractures and a 10-fold increase in rib fractures has been reported in adults with cystic fibrosis (61), and this can be problematic, as poor bone health can result in non-eligibility for a lung transplant in certain centers (63). Many factors contribute to low bone density including reduced BMI and lean mass when associated with low body weight, low levels of IGF-1, pubertal delay and hypogonadism, malabsorption of fat-soluble vitamins (including vitamins D and K), insufficient protein intake in diet, fecal calcium losses, secondary hyperparathyroidism, physical inactivity, increased secretion of inflammatory cytokines (IL-1β, IL-6 and TNF-α), chronic use of glucocorticoids, and direct effects of chloride channel defects (64-66).

 

Biliary atresia is also associated with malabsorption of fat-soluble vitamins, including vitamin D, and therefore, the condition can result in low bone density, which correlates with the severity of liver disease and jaundice (67, 68).

 

CONDITIONS OF REDUCED MECHANICAL BONE LOADING (INCLUDING DISUSE OR IMMOBILIZATION

 

Mechanical loading leads to a reduction in sclerostin secretion from osteocytes, the mechanosensors of bone, and increased signaling along the canonical Wnt pathway with an increase in bone formation (69). There are data to suggest that an optimal nutritional status and estrogen levels are permissive for these effects of mechanical loading on bone (70). Meta-analyses have demonstrated beneficial effects of bone loading activity on bone in children, particularly in the pre- and early pubertal years (71, 72). Therefore, conditions of reduced mechanical loading are associated with impaired bone accrual and low BMD. Similarly, the pull of muscle on bone is known to have bone anabolic effects. During periods of muscle disuse and prolonged immobilization, in addition to reduced osteoblastic activity (from increased production of sclerostin), there is an activation of osteoclastic bone resorption. Thus, conditions associated with hypotonia, spinal cord injury, spina bifida, muscular dystrophy, spinal muscular atrophy (SMA), and severe burns are associated with impaired bone accrual and low BMD in children and adolescents.

 

Patients with cerebral palsy (particularly those with limited ambulation) have low bone density and increased fracture risk associated with reduced mechanical loading of the skeleton, muscle wasting, suboptimal nutrition, and also vitamin D deficiency or impaired metabolism from concomitant use of certain anti-epileptic drugs (73-75). Studies have demonstrated that the severity of cerebral palsy predicts the severity of low BMD Z-scores in this condition (76, 77).

 

Duchenne and other muscular dystrophies are associated with reduced muscle mass, muscle strength, and muscle function resulting in low bone density and increased fracture risk (75, 78-82). Concomitant glucocorticoid therapy also impacts bone deleteriously, although amelioration of the underlying condition through glucocorticoid use may mitigate these effects to some extent (83). In one study, 53% of patients with Duchenne muscular dystrophy treated with deflazacort had vertebral fractures over a nine-year period (84). Another study has reported a 16-fold increased risk of fracture in patients taking daily deflazacort (85). Vamorolone, a dissociative steroidal anti-inflammatory drug, holds promise for use in this condition without significant bone effects (86). Studies have also reported impaired calcium metabolism and vitamin D status as well as high IL-6 levels in Duchenne muscular dystrophy, which could also contribute to impaired bone health (75, 79, 80, 87). Similarly, spinal muscular atrophy (SMA) is associated with low bone density and increased fracture risk (88-91). Low bone density in patients recovering from burns (92) is consequent to immobilization, muscle wasting, increased release of inflammatory cytokines that active osteoclastic activity and increase bone turnover, and low 25OHD levels (93). In one study, 27% of children with severe burns had low bone density Z-scores (92).

 

ENDOCRINE CONDITIONS

 

Many hormones have a direct impact on osteoblast and osteoclast activity (Figure 1); thus, a disruption in these hormone systems can have deleterious effects on bone.

Figure 1. Regulation of Bone Formation and Resorption. Osteoblasts are the primary bone-forming cells. Osteoblast anabolic activity is stimulated by testosterone and growth hormone and inhibited by cortisol and hyperglycemia. Osteoclasts mediate bone resorption. Thyroid hormone, parathyroid hormone, and inflammatory cytokines increase bone resorption, while estradiol inhibits osteoclast function. Osteocytes embedded within the bone matrix secrete sclerostin which inhibits osteoblast function; mechanical loading decreases sclerostin production thereby “releasing the brake” on osteoblast activity. Calcium deficiency, often as a result of vitamin D deficiency, leads to poorly mineralized bone matrix as well as secondary hyperparathyroidism.

 

Hypogonadism

 

Conditions of hypogonadism (Table 1) are associated with low bone density and impaired bone accrual given the critical role of the gonadal hormones on bone (70). Estradiol has anti-resorptive effects through its effects on the RANK-RANK-ligand-osteoprotegerin system. Estradiol also inhibits secretion of sclerostin, which otherwise inhibits the canonical Wnt signaling pathway and therefore osteoblast action, and also inhibits osteoclastic action (94). Testosterone has both direct bone anabolic and anti-resorptive effects, and also affects bone through its aromatization to estradiol. It increases periosteal bone apposition, while decreasing endosteal bone resorption, which collectively accounts for the larger size and thicker cortices of the male adult skeleton (70). During puberty, the rising levels of estradiol and testosterone are critical for adolescent bone accrual (95), and hypogonadism is therefore associated with reduced bone accrual, low bone density, and an increased risk of fracture (96, 97).

 

Hypercortisolemia

 

Chronic administration of glucocorticoids for underlying inflammatory or other conditions (such as inflammatory bowel disorders, chronic arthritis, Duchenne muscular dystrophy, renal conditions including post-transplant patients, and connective tissue disorders), and endogenous hypersecretion of cortisol (ACTH dependent or independent) can cause low bone density and increase the risk for fracture (98). Excessive exposure to glucocorticoids has multiple deleterious effects on bone. It inhibits osteoblastic activity (through direct effect on osteoblast precursors and stimulation of apoptosis of mature osteoblasts and osteocytes), reduces mechanosensing ability through its osteotoxic effects, increases osteoclast activity by decreasing osteoprotegerin and increasing RANK-ligand secretion from osteoblasts, impairs calcium absorption from the gut, impairs the renal handling of calcium, has an inhibitory effect on the growth hormone (GH)-IGF-1 axis, and leads to reduced muscle mass, impaired collagen formation, and suppression of the HPG axis (98, 99).

 

Importantly, hypercortisolemia is associated with increased fracture risk (particularly of the spine) independent of low BMD, related to the dose and duration of therapy (99). Low bone density can become evident within 3-6 months of therapy and improves in the first year after stopping glucocorticoids (particularly after the first six months). One study in children receiving glucocorticoids for three years for rheumatic disease reported an unadjusted vertebral fracture incidence rate of 4.4 per 100 person-years, and a 3-year incidence proportion of 12.4% (100). The highest annual incidence was in the first year, and every 0.5 mg/kg increase in glucocorticoid dose was associated with a doubling of fracture risk. Of concern, 50% of the fractures were asymptomatic and would have been missed without a lateral spine x-ray (100).  Importantly, recovery of vertebral shape and height appears possible in children affected in the pre- or early pubertal years and is unlikely in those who are mid to late pubertal (99, 101).

 

Chronic, Untreated Hyperthyroidism

 

Chronically high thyroid hormone levels, including at initial diagnosis of Graves’ disease, can lead to increased bone resorption and low BMD, particularly at cortical sites (102-104). An increase in IL-6 levels has been associated with this condition and contributes to increased bone resorption (105). Subclinical hyperthyroidism, as seen in survivors of pediatric differentiated thyroid carcinoma receiving levothyroxine at doses that suppress TSH (but with thyroid hormones levels still in the normal range), does not appear to have major negative effects on bone (106), indicating that high levels of thyroid hormones, but not suppressed TSH, account for the deleterious effects on bone in patients with hyperthyroidism.

 

Hyperparathyroidism

 

Primary hyperparathyroidism is rare in children and adolescents (and mostly associated with conditions such as MEN1 and MEN2), but secondary hyperparathyroidism occurs in conditions of hypocalcemia and vitamin D deficiency, and both secondary and tertiary hyperparathyroidism may be associated with chronic renal disease. The latter is discussed in a later section of this chapter. Chronic hyperparathyroidism causes increased bone resorption and results in low BMD, and the forearms (and primarily cortical sites) are involved to a greater extent that other parts of the body in this condition, with relative sparing of the spine (107).

 

Growth Hormone Deficiency and Resistance

 

Both GH and IGF-1 have multiple effects on bone. GH increases levels of osteoprotegerin, stabilizes the canonical Wnt signaling pathway, increases muscle mass and bone growth, while also stimulating local and hepatic IGF-1 secretion. The latter also stabilizes the canonical Wnt signaling pathway to activate osteoblasts, stimulate bone growth and an increase in muscle mass, increases 1-α hydroxylase activity, thus increasing intestinal absorption of calcium and phosphorus, increases tubular reabsorption of phosphorus, and increases RANK ligand activity. Thus, conditions of GH deficiency and resistance are at risk for low BMD. Adults with GH deficiency in the KIMS database had a 2.7 times higher fracture risk than the general population (108), and other studies have also reported an increased fracture risk in this population (109). However, a higher risk of fractures has not been observed in children with GH deficiency who received GH replacement therapy (110). Further, areal BMD in children with GH deficiency is often no longer low after adjusting for body size (111). Quantitative computed tomography studies have reported normal volumetric BMD, lower cortical thickness, and no differences for trabecular structure in children with GH deficiency vs. controls (112).  In conditions of undernutrition, an acquired state of GH resistance and low levels of IGF-1 contribute to low rates of bone accrual and low BMD (113).

 

Poorly Controlled Diabetes

 

While studies are still in the process of examining the effects of diabetes on bone in children, data seem quite clear that poorly controlled diabetes is associated with low BMD (114-117). This has been linked to low IGF-1 levels secondary to hypoinsulinemia resulting from poor diabetes control, increased markers of oxidative stress, and increased secretion of inflammatory cytokines (118).

 

CHRONIC MEDICAL CONDITIONS

 

Chronic inflammatory states such as inflammatory bowel disorders, connective tissue disorders, chronic arthritis and other inflammatory states are associated with low BMD for multiple reasons, including increased release of proinflammatory cytokines, which activate osteoclastic activity, chronic use of glucocorticoids and possibly some degree of undernutrition. Many of these conditions have been covered in previous sections.

 

Systemic Mastocytosis

 

Systemic mastocytosis is associated with low bone density in adults (119, 120), and BMD in these patients correlates with tryptase levels, mast cell proportion in bone marrow biopsies, and duration since diagnosis (119). Data are lacking in children.

 

Leukemia and Other Malignancies

 

Infiltrative conditions such as leukemia and other malignancies is associated with low bone density. Low bone density in patients with malignancy is a consequence of poor nutrition, malabsorption and diarrhea, vitamin D deficiency and associated hyperparathyroidism, release of inflammatory cytokines, chronic use of glucocorticoids, the effects of chemotherapy (gonadal failure, a direct suppressive effect of alkylating agents on bone marrow, and bone toxicity of high-dose methotrexate), as well as a direct effect of radiation therapy on osteoblasts. Pediatric acute lymphoblastic leukemia (ALL) is a known cause of low bone density and osteoporotic fractures (121-123). One study reported a cumulative fracture incidence of 32.5% for vertebral fractures and 23% for non-vertebral fractures in children with ALL over a six-year period, with 39% of children with vertebral fractures being asymptomatic (123). Vertebral reshaping occurred in younger children, but persistent vertebral deformity was noted in about 25%, particularly in older children and those with more severe vertebral collapse.

 

Beta-Thalassemia and Sickle Cell Disease

 

A large proportion of children with beta-thalassemia are reported to have low bone mass or bone density with reduced bone accrual compared to controls, regardless of transfusion and chelation regimens (124-126). One study reported BMC Z-scores of ≤ -2 in 61% of adolescents with beta-thalassemia (124). Another study reported that 82% and 52% of children and adolescents with transfusion dependent beta-thalassemia had low BMD Z-scores at the spine and the hip, respectively (125). Other studies, however, have reported much lower rates of low BMD in these children. Overall, nutritional status is a major determinant of bone outcomes in this condition.  Similarly, sickle cell disease in children and young adults has been associated with low bone density (127, 128), even after height adjustment (129), related to puberty, hip osteonecrosis, chronic pain, and hemoglobin values (129). Many of these children also have low calcium intake and low serum concentrations of 25OHD (127). Other predictors of low BMD include a low BMI, male sex, delayed puberty, and low serum zinc concentrations (128, 130).

 

Chronic Kidney Disease

 

Chronic kidney disease is a major risk factor for low BMD and fractures because of the associated secondary or tertiary hyperparathyroidism, hyperphosphatemia, reduced mineralization (reduced 1-α hydroxylase), increased cytokines such as TNF-α, and chronic use of glucocorticoids (131-133).

 

IATROGENIC CAUSES

 

Certain medications can also contribute to low bone density in children and adolescents. Antiepileptic medications such as phenytoin, primidone, phenobarbital, and carbamazepine impair vitamin D metabolism by stimulating hepatic microsomal cytochrome P450 enzymes, causing vitamin D deficiency, secondary hyperparathyroidism, and low BMD (134). Data also suggest that antiepileptic drugs may inhibit the cellular response to parathyroid hormone (PTH). Further, use of valproic acid has been associated with increased osteoclast activity and low BMD in some studies. In contrast, newer antiepileptic medications such as lamotrigine and topiramate have not been associated with impaired vitamin D metabolism or low bone density.

 

As already discussed, chronic glucocorticoid use has several deleterious effects on calcium absorption, retention, and osteoblast and osteoclast function. Methotrexate and certain antiviral drugs have also been demonstrated to contribute to impaired bone health. Further, medications that suppress the hypothalamic-pituitary-gonadal axis such as GnRH analogs and depo medroxyprogesterone acetate are associated with low BMD. Lastly, radiation therapy is known to have direct deleterious effects on osteoblasts.

 

LIFESTYLE FACTORS  

 

Cigarette smoking is believed to be a risk factor for low bone density (135), although it is difficult to sort out the effects of smoking on bone versus the contribution of risk factors common among smokers, which include a low BMI, greater intake of alcohol, lower levels of physical activity, and poor diet. The longer the duration of smoking and the greater the number of cigarettes consumed, the greater the risk of fracture. Further, healing following a fracture is slower in smokers than non-smokers, and complications during the healing process are more common in smokers. Even exposure to secondhand smoke has been related to suboptimal bone outcomes. Women who smoke often produce less estrogen and tend to enter menopause earlier, which would also contribute to increased bone loss. Further, levels of cortisol and free radicals are higher in smokers, which may also contribute, and nicotine and free radicals are toxic to osteoblasts. Importantly, quitting smoking does reduce the risk of low bone density and fractures. However, it may take several years to lower a former smoker’s risk.

 

Additionally, alcohol is deleterious to bone when consumed in excess (136); more than two alcoholic drinks per day are associated with low bone density, and may be related to decreased absorption of calcium, increased concentrations of cortisol and PTH, lower levels of estrogen, and alcohol per se is toxic to osteoblasts. Reduced bone density and impaired bone quality contribute to increased fracture risk (137).

 

Table 1. Causes of Low Bone Density or Osteoporosis

PRIMARY CAUSES OF LOW BONE DENSITY OR OSTEOPOROSIS

Osteogenesis imperfecta (COL1A1, COL1A2, IFITMF5, SERPINF1, CRTAP, LEPRE1 and other genes)

Marfan syndrome (FBN1)

Ehlers Danlos syndrome (COL5A, COL3A, non-monogenic forms)

Homocystinuria (CBS, MTHFR, MTR, MTRR, and MMADHC genes)

Lysinuric protein intolerance (SLC7A7)

Osteoporosis pseudoglioma syndrome (LRP5)

Idiopathic juvenile osteoporosis

Hypophosphatasia (ALPL)

Others

Other Primary Conditions Associated with Fragility Fractures but Without Low Bone Density

Polyostotic fibrous dysplasia (GNAS1)

Osteopetrosis (LRP5, CLCN6, CA2, others)

Pycnodysostosis (CTSK)

SECONDARY CAUSES OF LOW BONE DENSITY OR OSTEOPOROSIS

Nutritional and Malabsorptive Conditions

• Deficient intake of calcium and vitamin D

• Conditions of low energy availability or energy deficit (e.g., anorexia nervosa, Female Athlete Triad/Relative Energy Deficiency in Sports (RED-S))

• Conditions of malabsorption (e.g., celiac disease, inflammatory bowel disease, cystic fibrosis, biliary atresia)

Conditions of Reduced Mechanical Bone Loading (Including Disuse or Immobilization)

• Cerebral palsy

• Spinal cord injury

• Spina bifida

• Muscular dystrophy

• Spinal muscular atrophy

• Severe burns

• Conditions of prolonged immobilization

Endocrine Conditions

• Hypergonadotropic hypogonadism:

O     Primary ovarian insufficiency

O     Primary testicular insufficiency

•  Hypogonadotropic hypogonadism:

O     Isolated or combined pituitary hormone deficiencies (genetic and acquired causes)

O     Hyperprolactinemia

O     Functional hypothalamic amenorrhea (conditions of energy deficit, chronic stress)

O     Medications (e.g., depot medroxyprogesterone acetate, GnRH analog therapy)

• Hypercortisolemia:

O     Iatrogenic from prolonged use of glucocorticoids for underlying chronic conditions*

O     Endogenous: adrenal, pituitary and ectopic tumors

• Hyperthyroidism (chronic untreated)

• Hyperparathyroidism

• Growth hormone deficiency and resistance

• Diabetes (particularly when poorly controlled)

Chronic Medical Conditions

• Chronic inflammatory states**

• Mastocytosis

• Infiltrative conditions (e.g., leukemia and other malignancies)

• Thalassemia and sickle cell disease

• Chronic kidney disease

Iatrogenic

• Antiepileptic medications

• Glucocorticoids

• Methotrexate

• Antiretroviral drugs

• Depot medroxyprogesterone acetate

• GnRH analogs

• Radiation therapy

Lifestyle factors

smoking and chronic alcohol use

*Examples: Duchenne muscular dystrophy, inflammatory bowel disorders, chronic arthritis, renal conditions including post-transplant patients, connective tissue disorders, leukemia and other malignancies

**Examples: Inflammatory bowel disease, chronic arthritis, connective tissue disorders, nephrotic syndrome

 

EVALUATION

 

History and Physical Exam

 

Initial evaluation includes a thorough medical history, with special attention to aspects that may adversely affect bone health such as chronic immobility or oncology treatments; medication history, with a focus on past and current medications that may adversely influence bone health, such as glucocorticoids, anti-epileptic drugs, and hormonal contraception; pubertal history, dependent upon age; and family history of recurrent fractures, pre-menopausal osteoporosis, and bone disorders.

 

Additionally, a bone health assessment should include the following: 1) fracture history, including mechanism of injury (e.g., traumatic, fall from a standing height, etc.), treatment (e.g., cast, surgery, and if any complications such as prolonged healing); 2) dietary intake of calcium-rich foods, with quantification of the typical servings per day of dairy, and any supplementation of calcium and vitamin D, including type of supplementation (e.g., calcium carbonate, cholecalciferol, etc.) and dosages; 3) physical activity including sports, dance, gym class, physical therapy, and time in stander, as applicable.

 

The physical exam should be comprehensive and include a thyroid and pubertal exam, palpation of the spine, and assessment for hyper flexibility and any physical restrictions (i.e., contractures).

 

Laboratory Studies

 

Dependent upon the degree of clinical concern for osteoporosis or increased bone fragility, determined by the history, next steps could include assessment of bone mineral density by DXA, laboratory studies, and additional testing. If the bone fragility history is equivocal, it is reasonable to start with a DXA and perform additional evaluation if there is demonstrated low BMD. However, if the history is strongly suggestive of osteoporosis or increased bone fragility, the next steps are typically DXA and laboratory evaluation (see Table 2). It is helpful to assess calcium, magnesium, phosphorus, alkaline phosphatase, 25OHD, PTH, screening for celiac disease, and creatinine. Urinary calcium and creatinine ratio (spot) can be helpful to assess calcium status and there is an increased risk of hypercalciuria in non-ambulatory patients. Rarely, bone turnover markers, such as bone formation markers osteocalcin and bone specific alkaline phosphatase and the bone resorption marker c-telopeptide, may be helpful, if there is a clinical concern for a low bone turnover state. Also, 1,25-dihydroxyvitamin D (1-25OHD) is not routinely assessed, but is helpful if there is a clinical concern for a disorder of vitamin D metabolism. Additional biochemical assessments should be considered on a case-by-case basis. Notably, for all laboratory assessments, it is important to have pediatric reference ranges, and pubertal specific (i.e., Tanner stage) reference ranges when applicable, in order to properly interpret values.

 

Imaging Studies

 

EVALUATION OF BONE MINERAL DENSITY

 

DXA is the clinical gold standard for measuring BMD (138). The current International Society for Clinical Densitometry (ISCD) pediatric guidelines for optimal bone densitometry assessments include: for 4-15 years old total body less head and lumbar spine; 16 years and older lumbar spine and hip (1). By age 15 years, the skeletal landmarks at the hip that guide positioning are well-developed and enable the replication of positioning. In adolescent patients, it may be useful to perform a transition scan around 16 years old, including total body less head, lumbar spine, and hip, as this will enable assessment of interval change at two skeletal sites. The current ISCD pediatric guidelines suggest measurement of hip BMD in the school-age child if lack of weight-bearing and skeletal fragility are concerns. In certain clinical scenarios, it may be useful to obtain a distal lateral femur or forearm scans, such as in patients with neuromuscular disorder with impaired mobility (139). Distal lateral femur scans can be very informative in non-ambulatory patients. Forearm scans can be useful in patients who are unable to hold still, those with significant contractures, and in patients with orthopedic hardware that precludes scans of other skeletal sites.

 

It is helpful to assess interval change enabling evaluation of bone accrual and comparison of Z-scores over time (i.e., did they increase, remain the same, or decrease). The shortest interval usually assessed is one year, and dependent upon the clinical situation, it may be prudent to reassess after two years or longer, especially if it would not change clinical management. From a practical standpoint, insurance typically requires DXA scans to be performed at least one year apart (> 365 days from last DXA scan), but all scanning sites should be covered and are determined by medical necessity.

 

DXA is interpreted via areal BMD (g/cm2) calculated Z-scores which are age, sex, and ancestry matched and based on pediatric normative data (140). A Z-score less than -2.0 SD is low, and between -1.0 to -2.0 SD is considered borderline low and may be of clinical significance in patients with risk factors for increased bone fragility. Notably, in pediatrics, the diagnosis of osteoporosis requires both: 1) a clinically significant fracture history, which is defined as at least two long bone fractures in children less than 10 years old, at least three long bone fractures by 19 years old, or any vertebral fractures; and 2) low bone density, with BMD Z-scores < -2.0, assessed by DXA.

 

However, as a two-dimensional projected area of a three-dimensional structure, DXA is affected by bone size. For this reason, pediatric DXA derived areal BMD is affected by bone size and smaller bones may have an artifactually lower BMD Z-score. To take this into account, the BMD Z-score can be adjusted for height (i.e., height for age Z-score) in those with short stature (https://zscore.research.chop.edu/calcpedbonedens.php) or, in certain scenarios, adjusted for bone age if a patient has delayed puberty but is not short (141).

 

DXA only assesses bone mass and density and does not fully capture all factors contributing to bone fragility, such as volumetric BMD (g/cm3), bone microarchitecture (e.g., trabecular vs. cortical bone), bone quality, or bone strength. Additional research modalities allow for the assessment of bone microarchitecture, quality, and strength, such as peripheral quantitative computed tomography (pQCT), high resolution pQCT, and trabecular bone score (TBS), a measure of bone quality of the lumbar spine which correlates to bone microarchitecture. There are recently published pediatric TBS reference ranges (142). However, there are few pediatric normative data for these alternative modalities, currently limiting their use to primarily the research setting.

 

ASSESSING FOR COMPRESSION FRACTURES

 

It is important to evaluate the patient for compression fractures in patients with low bone density and clinical concern, such as back pain or unexplained decrease in physical activity. If there is acute concern for compression fractures, initial evaluation should include spine x-rays, typically two-view anteroposterior and lateral radiographs. If imaging is consistent with compression fracture(s), there should be prompt evaluation by orthopedics and endocrinology, as the disease course can be positively affected with appropriate treatment.

 

Vertebral fracture assessment (VFA) is an additional spine assessment that can be performed concurrently with DXA to assess for spine vertebral fractures. It is commonly used in adults but has only recently been utilized in children (143). Advantages of VFA over spine radiographs include lower radiation dose, logistics (done at the same visit as DXA), and lower cost. There is emerging evidence that it is useful to screen for vertebral fractures with VFA in high-risk pediatric populations, such as those with Duchenne muscular dystrophy and osteogenesis imperfecta.

 

RADIOGRAPHY

 

Radiographs (i.e., x-rays) do not quantify bone mass and are not a good screening tool for low bone mass. However, if radiographs are performed for other indications, such as a clinical concern for fracture or related to another medical evaluation, when there is at least 30-40% bone loss, there are typical findings, such as bone demineralization, gracile bones, and thin cortex. If there is radiographic concern for low bone mass, with concurrent risk factors for suboptimal bone accrual, this should be further evaluated with a DXA to quantify BMD. Radiographs can be useful to assess for specific findings in several bone disorders including rickets, which is a consequence of under-mineralization of bone in growing children; skeletal dysplasias, which are often diagnosed based on radiographs; osteopetrosis, with over-mineralization of bone; and osteogenesis imperfecta, of which several types have a substantial number of wormian bones on skull radiograph.

 

Consultative Services

 

Additional consultations may be required, depending upon the specific underlying etiology of the patient’s low bone density. For many patients, especially those who are underweight, intolerant of cow milk-based foods (i.e., milk protein allergy), or have an eating disorder, working closely with a nutritionist to ensure adequate caloric intake and calcium-rich foods is very useful. Patients with mobility challenges, such as hypermobility and non-ambulatory patients, often benefit from working with physical therapy. Dependent upon the suspected underlying disease, additional evaluation by other subspecialists, such as a geneticist or gastroenterologist, may be helpful.

 

Table 2. Laboratory and Imaging Evaluation for Increased Bone Fragility and Osteoporosis

Laboratory Studies

Standard Evaluation

calcium with albumin, magnesium, phosphorus, alkaline phosphatase, 25-hydroxyvitamin D, parathyroid hormone, tissue transglutaminase with IgA, creatinine

Consider*

genetic testing for osteogenesis imperfecta and possibly other genetic disorders (dependent upon family history), TSH/fT4, pubertal assessment (FSH, LH, estradiol, testosterone), bone turnover markers (osteocalcin, bone specific alkaline phosphatase, c-telopeptide), 1,25-dihydroxyvitamin D, erythrocyte sedimentation rate and c-reactive protein (if known or suspected chronic inflammatory disease), urine calcium and creatinine ratio (spot)

Imaging Assessments

Standard Evaluation

DXA – sites dependent upon age and logistics**

Consider*

Screening lateral spine x-rays in high-risk patients, VFA in high-risk patients, skull x-ray if concern for osteogenesis imperfecta, skeletal survey if concerned for skeletal dysplasia

* Consider additional assessment on a case-by-case basis

** See text for details regarding recommended sites

 

TREATMENT

 

Non-Pharmacologic Interventions to Optimize Bone Mineral Density and Bone Strength

 

CALCIUM AND VITAMIN D

 

Calcium is a critical component of bone, necessary for the formation of hydroxyapatite which confers strength to the bone matrix (144). Calcium is a threshold nutrient, meaning that once adequate intake is achieved to maximize calcium retention, further intake does not provide additional benefit to bone health (145). The optimal calcium intake for any individual depends on several factors: these include vitamin D status, given that vitamin D stimulates gut absorption of calcium, as well as other dietary factors such as sodium and protein intake which can alter renal calcium excretion (146). In addition to impairing skeletal mineralization, dietary calcium insufficiency may cause a secondary hyperparathyroidism, promoting bone resorption and phosphate excretion, further decreasing bone density. The United States National Academy of Sciences guidelines provide a Recommended Dietary Allowance (RDA) of calcium which is anticipated to meet the needs of 97.5% of the healthy population (Table 3) (147).

 

Table 3. Recommended Daily Intake of Calcium and Vitamin D by Age

Age

Calcium intake (mg)

Vitamin D intake (IU)

0-6 months

200

400

7-12 months

260

400

1-3 years

700

600

4-8 years

1000

600

9-18 years

1300

600

19-50 years

1000

600

 

1-25OHD, the active metabolite of vitamin D, is critical for optimal gastrointestinal absorption of calcium. Vitamin D sufficiency is assessed by circulating concentrations of 25OHD, though there is some controversy regarding the 25OHD threshold which reflects sufficiency. The Institute of Medicine (IOM) has defined sufficiency as 25OHD ≥ 20 ng/mL (147), based largely on bone biopsy evaluation of unmineralized osteoid accumulation (148), while other guidelines recommend a target of 30 ng/ml (149). To achieve these serum concentrations, the IOM recommends daily intake of 400 IU in the first year of life, and 600 IU from ages 1-70 years (Table 3). However, individual patients may require higher intakes to achieve vitamin D sufficiency, including those with malabsorptive conditions such as cystic fibrosis and inflammatory bowel disease (150, 151). In addition, individuals with obesity have a smaller incremental increase in 25OHD concentration with supplemental vitamin D and may require 2000 IU daily or more to achieve target serum concentrations (152).

 

For children on high-dose glucocorticoid therapy for underlying inflammatory or oncologic disease, several specific effects on calcium and vitamin D metabolism must be considered. Glucocorticoids can directly inhibit the gut absorption of calcium via decreased expression of epithelial calcium channels (153). In addition, glucocorticoids can inhibit the synthesis of 1-25OHD and accelerate the catabolism of vitamin D metabolites (154). Therefore, these patients may require higher than typical intake of calcium and vitamin D, which should be guided by monitoring circulating concentrations of 25OHD and PTH.

 

Studies of calcium supplementation in healthy children suggest that increases in BMD may be limited to prepubertal children and in those with low baseline calcium intake, again supporting the model of calcium as a threshold nutrient (155, 156). Long-term follow-up studies indicate that the effect of calcium supplementation wanes after discontinuation of the intervention (25, 157-160). Similarly, a meta-analysis of pediatric vitamin D supplementation studies indicated only modest effects on BMD which were limited to those with a baseline 25OHD <14 ng/mL (161). One follow-up study showed a loss of effect on BMD three years after completion of the supplementation intervention (162). These data suggest that, for children at risk for low calcium intake or low circulating vitamin D metabolites, optimization of these factors should be an ongoing process.

 

PHYSICAL ACTIVITY

 

Mechanical loading of the skeleton via high-impact physical activity promotes bone acquisition in growing children (71). Several observational studies have shown an association of childhood physical activity with increased BMD (163-167), with effects that persist into young adulthood (168, 169). A meta-analysis of RCTs confirmed a significant though small effect of physical activity on measures of bone mass, with increased responsiveness in pre-pubertal participants (170). Interestingly, this analysis also revealed a positive association of calcium intake with bone mineral content and density, suggesting that calcium enables or synergizes with the effects of weight-bearing (171, 172). Among healthy children, a threshold force of approximately 3 times the force of gravity, such as experienced during running or jumping, seems to be required to stimulate bone formation (173). Importantly, physical activity is beneficial even in healthy children with a high genetic risk for low BMD (174).

 

The role of weight-bearing activity in children with underlying musculoskeletal disease is less well-studied. Small studies of children with cerebral palsy have shown efficacy of increasing time of use in a stander (175) and a physical therapy program (176) to improve BMD. Whole-body vibration (WBV) has been studied in children with several conditions (177) including cerebral palsy (178-182), Duchenne muscular dystrophy (183-185), osteogenesis imperfecta (186, 187), and Down syndrome(188). Synthesis of the results of these studies is challenging due to methodological variation in the magnitude and frequency of vibration as well as the length of treatment sessions. In general, among children with cerebral palsy, WBV appears to have positive effects on bone density and bone strength estimates, while data are conflicting or limited in other conditions. 

 

Pharmacologic Intervention

 

WHOM TO TREAT WITH BONE ACTIVE MEDICATIONS

 

Selection of appropriate pediatric patients for pharmacological intervention is not straightforward (189). Unlike in adults, for whom low BMD alone suffices to confer a diagnosis of osteoporosis, the diagnosis of osteoporosis in children requires evidence of skeletal fragility, defined as multiple long-bone fractures or a vertebral compression fracture (190). Current evidence-based guidelines for the use of pharmacotherapy do not recommend prophylactic use of pharmacotherapy given the absence of robust prospective data enabling estimation of fracture risk in vulnerable children (191). This differs from guidelines for osteoporosis management in adults, in whom primary prevention of fracture is a goal (192). Given that, particularly in children with progressive neuromuscular disease, a single fracture can lead to permanent loss of ambulation (193, 194), further research to better define which pediatric patients may benefit from prophylactic therapy is urgently needed. Indeed, in certain particularly high-risk patients, such as those with spinal muscular atrophy (SMA), clinicians may choose to treat in advance of a patient meeting pediatric osteoporosis criteria (195).

 

Conversely, some children who fulfill criteria for osteoporosis may not warrant pharmacotherapy. Children with secondary causes of osteoporosis which may be transient, such as an inflammatory disease that responds to treatment, or glucocorticoids that are discontinued, have the potential to repair BMD losses as well as to spontaneously heal vertebral fractures. As an example, in a cohort of children with Crohn’s disease, initiation of anti-TNF-alpha therapy led to significant increases in BMC and BMD Z-scores over 12 months, indicating “catch-up” bone accrual (196). A study of fractures among children with acute lymphocytic leukemia (ALL) demonstrated that, while the cumulative incidence of vertebral compression fracture over 6 years was 33%, complete healing with restoration of normal vertebral shape was observed in 77% of those with fractures. Predictors of healing included younger age (mean 4.8 vs 8.0 years) and number and severity of fractures (123). Lower cumulative glucocorticoid doses may also correlate with greater chance of spontaneous healing in children with inflammatory disease (101). The decision whether or not to initiate pharmacotherapy thus depends on a careful weighing of each child’s individual clinical history and anticipated course of disease (189).

 

BISPHOSPHONATE TREATMENT

 

For children at high risk of fracture, bisphosphonates are the best-studied and most widely used pharmacologic treatment. Bisphosphonates are non-hydrolysable analogs of pyrophosphate that bind tightly to hydroxyapatite crystals and inhibit osteoclast-mediated bone resorption (197). Once embedded in bone, bisphosphonates are retained in the pediatric skeleton for several years, as evidenced by detectable urinary concentrations up to eight years after administration (198). Bisphosphonates are available in both oral and intravenous formulations. The bioavailability of oral bisphosphonates is extremely low, and there is significant intraindividual skeletal retention of bisphosphonates, which depends in part on endogenous bone turnover and renal function (199).

 

Table 4. Selected Bisphosphonates: Dosing and Examples of Pediatric Uses

Bisphosphonate

Administration

Typical Dosing Regimen

Use in Pediatrics

Alendronate

PO

5-10 mg daily(200) or 35 mg weekly(201)

·  Osteogenesis imperfecta(200, 202-204)

·  Glucocorticoid induced osteoporosis(205, 206)

·  Duchenne muscular dystrophy(207, 208)

·  Cerebral palsy(209)

·  Cystic Fibrosis(210)

·  Acute lymphoblastic leukemia(211)

·  Spinal cord injury(212)

·  Transplant(213)

Risedronate

PO

2.5-5 mg daily(214) or 15-30 mg once weekly(215)

·  Osteogenesis imperfecta(214-216)

·  Duchenne muscular dystrophy(217)

·  Cerebral palsy(218)

·  Non-ambulatory children(219)

 

Pamidronate

IV

9 mg/kg year, given as 0.25-1 mg/kg daily for 3 days every 2-4 months(220)

·  Osteogenesis imperfecta(204, 221, 222)

·  Glucocorticoid induced osteoporosis (223)

·  Cerebral palsy(224-226)

·  Acute lymphoblastic leukemia(227)

·  Idiopathic juvenile osteoporosis(228)

·  Burns(229)

 

Zoledronic acid

IV

0.05-0.1 mg/kg year, every 3-12 months(230, 231)

·  Osteogenesis imperfecta(230, 232-235)

·  Glucocorticoid induced osteoporosis(236)

·  Duchenne muscular dystrophy(237)

·  Cerebral palsy(238, 239)

·  Rett syndrome(239)

 

Neridronate

IV

2 mg/kg every 3-6 months(240)

·  Osteogenesis imperfecta(240, 241)

·  Osteoporosis pseudoglioma syndrome(242)

 

Early studies of bisphosphonate therapy in children focused on those with osteogenesis imperfecta. Treatment with both oral and intravenous formulations leads to significant increases in BMD at both the hip and spine (200, 214, 215, 243-245). Subsequent studies in other conditions including cystic fibrosis and glucocorticoid induced osteoporosis showed similar increases in BMD with bisphosphonate use (205, 210). This is a non-trivial result, given that skeletal fragility in osteogenesis imperfecta, as in most pediatric conditions, is not mediated by accelerated bone resorption. A primarily anti-resorptive medication thus does not address the underlying cause of low BMD. However, trans-iliac biopsy data demonstrate that bisphosphonate therapy in children leads to increases in cortical width, via modeling-based bone formation at the periosteal and endocortical surfaces (246). In addition, trabecular BMD increases via an increase in trabecular number but not in thickness. This effect is hypothesized to be due to a greater retention of primary trabeculae after new bone formation and subsequent incorporation into secondary spongiosa (246).

 

While the beneficial effects of bisphosphonates on bone density are well-documented, effects on the more critical outcomes of fracture and associated morbidities are less clear and almost exclusively limited to the osteogenesis imperfecta population (see Endotext chapter on osteogenesis imperfecta for details). In adults, data from a recent individual patient data meta-regression of osteoporosis trials revealed significant and strong correlations between increases in BMD and fracture risk reduction (247). Whether this result generalizes to growing children is uncertain. Small observational studies and randomized controlled trials suggest that bisphosphonates may reduce the incidence of vertebral fracture in glucocorticoid-treated children (101, 237, 248). Data regarding anti-fracture efficacy in other conditions including cerebral palsy is lacking (238).

 

The choice of which bisphosphonate to use, as well as the optimal dosing regimen and length of treatment, is challenging due to a limited number of trials as well as their relatively small size. Some studies suggest that intravenous (IV) agents may be more effective at promoting vertebral fracture healing (249), though a head-to-head study of alendronate vs. pamidronate in children with osteogenesis imperfecta showed no difference in BMD accrual, suppression of bone turnover markers, or fracture incidence (250). A major consideration is the risk of pill esophagitis with oral bisphosphonates, which is exacerbated by the presence of gastrointestinal reflux, leading to the recommendation that patients should swallow pills only with water and remain upright for at least 30 minutes after administration. Given the significant challenge this poses to many children with osteoporosis due to an underlying neuromuscular or other chronic disease, IV bisphosphonate therapy is often the most practical choice. Dosing regimens vary both by underlying condition and between institutions; typical dosing regimens are noted in Table 4.

 

How long to continue therapy once initiated is also not well-defined. Because growing children accrue new bone via modeling-based growth, intermittent dosing regimens result in new bone not being exposed to bisphosphonates. This leads to the classic “zebra-lines” seen on x-rays of children treated with IV bisphosphonates (251). Concern has arisen that the junction between regions of treated and non-treated bone may be at particularly high risk of fracture (252, 253). For children with primary osteoporosis, continuation of therapy until the completion of growth is thus typically recommended. Monitoring of BMD, via DXA, as well as careful assessment of fracture incidence both by history and spine imaging can guide the maintenance phase of therapy which may require decreases in the dose or frequency of administration (252, 253). In children with secondary osteoporosis in which the underlying condition has resolved or is well-controlled, discontinuation of treatment with close monitoring may be appropriate (191).

 

Short-term Adverse Effects of Bisphosphonates

 

Because the skeleton functions as a reservoir for calcium as well as phosphate, anti-resorptive therapy can lead to short-term hypocalcemia and hypophosphatemia, which typically presents in the first 1-3 days after infusion though may have a more delayed onset (234, 254, 255). While often asymptomatic, due to the possibility of symptomatic hypocalcemia requiring IV calcium infusion, it is critical to mitigate this risk by ensuring vitamin D sufficiency (i.e., 25OHD > 30 ng/mL) and optimization of oral calcium intake via diet or supplementation starting the night prior to infusion and continuing for the following 5-10 days. Due to the higher risk of electrolyte abnormalities with the first dose, a 50% reduction is commonly employed (256). Acute phase reaction characterized by myalgia, bone pain, fever, nausea, and headache is seen in 20-80% of patients following the first IV infusion (255-258) and can typically be managed with anti-pyretic, analgesic, and anti-nausea medication as needed.

 

Particular care must be taken with patients on glucocorticoid therapy who may have iatrogenic central adrenal insufficiency and may thus require stress-dose glucocorticoid treatment to provide 24-hour glucocorticoid coverage as well as careful anticipatory guidance about the risk of adrenal crisis in this setting. As bisphosphonates are renally cleared, it is critical to assess renal function in children prior to administration to prevent nephrotoxicity. For children with underlying musculoskeletal disease, serum creatinine may not be an accurate reflection of renal function, and measurement of cystatin C as an alternative assessment of renal function should be performed (259). While concerns about bisphosphonates interfering with fracture healing have been raised, this has not been borne out by evidence except in the special case of iatrogenic injury via osteotomy (260).

 

Other Adverse Effects of Bisphosphonates

 

In adults, particularly at the high doses used in malignancy, bisphosphonates have been reported to cause several rare but serious adverse events including atypical femoral fracture (AFF) and osteonecrosis of the jaw (ONJ). An AFF is a low-trauma, transverse fracture of the subtrochanteric femur, typically preceded by prodromal pain (261). While such fractures have been seen in children with osteogenesis imperfecta, this may reflect the natural history of the disease and not be related to bisphosphonate use (262, 263). Case reports of AFFs in bisphosphonate-treated children with other conditions including idiopathic juvenile osteoporosis (264) and SMA (195), suggest that anticipatory guidance regarding the possibility of AFF and early symptoms should be offered to patients. Several case-finding series have not identified bisphosphonate associated ONJ in children (265, 266). Several cases of an osteopetrosis-like phenomenon have been reported in children exposed to bisphosphonates; in all cases, these were at substantially higher bisphosphonate doses than are typically prescribed to children (267, 268). Finally, possible teratogenicity of bisphosphonates, particularly given their prolonged retention in and release from the skeleton has been raised as a potential concern. However, a case series of twenty one women exposed to bisphosphonates just prior to conception or during pregnancy did not demonstrate any concerning signal of fetal harm (269).

 

OTHER AGENTS

 

Denosumab is a humanized monoclonal antibody against RANKL, a critical factor for osteoclast differentiation and activation. As such, similar to bisphosphonates, denosumab is a potent anti-resorptive medication. The effective half-life of denosumab is much shorter than bisphosphonates, and a major clinical challenge in its use is the “rebound effect,” specifically an increase in bone turnover markers above pre-treatment baseline levels and a significant increase in vertebral fractures after discontinuation in adults (270, 271). In several case reports of denosumab use in children, this rebound can present as severe hypercalcemia within just several weeks following the previous dose (272-276). Given these considerations, denosumab is currently used only sparingly in pediatric populations with specific indications including osteogenesis imperfecta type 6 (277, 278) and giant cell tumors (279-281).

 

Given that most pediatric osteoporosis stems from insufficient bone accrual (i.e., decreased bone formation), the use of anabolic rather than anti-resorptive agents may offer better efficacy (282, 283). Sclerostin is an endogenous inhibitor of the canonical wnt-β-catenin signaling pathway, and romosozumab, an anti-sclerostin antibody, has been approved for women with post-menopausal osteoporosis (284, 285). An alternative sclerostin antibody, setrusumab, has been investigated in a phase 2 trial of adults with osteogenesis imperfecta (286), and pediatric studies of both antibodies in osteogenesis imperfecta are ongoing (Clinicaltrials.gov: NCT05768854, NCT05125809, and NCT04545554).

 

Teriparatide, the c-terminal portion of PTH, is also approved for post-menopausal osteoporosis and has potent osteoblast-stimulating activity. Until recently, the United States FDA included a black box warning about increased risk of osteosarcoma in patients treated with teriparatide based on pre-clinical models. While phase 4 data have not confirmed an excess risk in clinical patients and this black box warning was removed in 2020, persistent FDA guidance to avoid teriparatide in patients with open epiphyses has limited its use. A recent small study of adolescent boys with Duchenne muscular dystrophy suggested decreased fracture incidence with teriparatide and no significant adverse events (287). Most patients in this study were treated for two years and then transitioned to an anti-resorptive therapy to prevent the loss of BMD observed after discontinuation of teriparatide in adults (288, 289).

 

CONCLUSIONS

 

Childhood osteoporosis has several potential underlying etiologies, requiring a careful assessment by clinicians with expertise in the numerous mechanisms which can contribute to skeletal fragility. Both non-pharmacologic therapies as well as bone-active medications such as bisphosphonates increase bone mass and may lower the risk of fracture. The development of novel therapies that can restore physiologic anabolic bone activity in children with insufficient bone accrual of various causes has the potential to improve care for pediatric patients with osteoporosis. Prospective data acquisition to inform treatment strategies for primary prevention of fracture in children with osteoporosis, as is done in adult populations, is urgently needed to prevent the significant morbidity of fracture in this vulnerable population.

 

REFERENCES

 

  1. Shuhart, C.R., et al., Executive Summary of the 2019 ISCD Position Development Conference on Monitoring Treatment, DXA Cross-calibration and Least Significant Change, Spinal Cord Injury, Peri-prosthetic and Orthopedic Bone Health, Transgender Medicine, and Pediatrics. J Clin Densitom, 2019. 22(4): p. 453-471.
  2. Estrada, K., et al., Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture. Nat Genet, 2012. 44(5): p. 491-501.
  3. Mullin, B.H., et al., Genome-wide association study using family-based cohorts identifies the WLS and CCDC170/ESR1 loci as associated with bone mineral density. BMC Genomics, 2016. 17: p. 136.
  4. Zheng, H.F., et al., Whole-genome sequencing identifies EN1 as a determinant of bone density and fracture. Nature, 2015. 526(7571): p. 112-7.
  5. Marini, J.C. and A.N. Dang Do, Osteogenesis Imperfecta. 2000.
  6. Folkestad, L., et al., Bone Geometry, Density, and Microarchitecture in the Distal Radius and Tibia in Adults With Marfan Syndrome Assessed by HR-pQCT. J Bone Miner Res, 2020. 35(12): p. 2335-2344.
  7. Trifiro, G., et al., Areal bone mineral density in children and adolescents with Marfan syndrome: evidence of an evolving problem. Bone, 2015. 73: p. 176-80.
  8. Grover, M., et al., Assessment of bone mineral status in children with Marfan syndrome. Am J Med Genet A, 2012. 158A(9): p. 2221-4.
  9. Eller-Vainicher, C., et al., Bone involvement in adult patients affected with Ehlers-Danlos syndrome. Osteoporos Int, 2016. 27(8): p. 2525-31.
  10. Coussens, M., et al., Bone parameters in hypermobile Ehlers-Danlos syndrome and hypermobility spectrum disorder: A comparative cross-sectional study. Bone, 2023. 166: p. 116583.
  11. Mazziotti, G., et al., High prevalence of radiological vertebral fractures in adult patients with Ehlers-Danlos syndrome. Bone, 2016. 84: p. 88-92.
  12. Banica, T., et al., Higher fracture prevalence and smaller bone size in patients with hEDS/HSD-a prospective cohort study. Osteoporos Int, 2020. 31(5): p. 849-856.
  13. Weber, D.R., et al., Low bone mineral density is a common finding in patients with homocystinuria. Mol Genet Metab, 2016. 117(3): p. 351-4.
  14. Al-Qattan, S., C. Malcolmson, and S. Mercimek-Andrews, Lysinuric protein intolerance mimicking N-acetylglutamate synthase deficiency in a nine-year-old boy. Mol Genet Metab Rep, 2021. 27: p. 100741.
  15. Lukkarinen, M., et al., Effect of lysine infusion on urea cycle in lysinuric protein intolerance. Metabolism, 2000. 49(5): p. 621-5.
  16. Gomez, L., et al., Treatment of severe osteoporosis with alendronate in a patient with lysinuric protein intolerance. J Inherit Metab Dis, 2006. 29(5): p. 687.
  17. Saito, M. and K. Marumo, The Effects of Homocysteine on the Skeleton. Curr Osteoporos Rep, 2018. 16(5): p. 554-560.
  18. Abdel-Hamid, M.S., et al., Osteoporosis-pseudoglioma syndrome in four new patients: identification of two novel LRP5 variants and insights on patients' management using bisphosphonates therapy. Osteoporos Int, 2022. 33(7): p. 1501-1510.
  19. Tuysuz, B., et al., Osteoporosis-pseudoglioma syndrome: three novel mutations in the LRP5 gene and response to bisphosphonate treatment. Horm Res Paediatr, 2012. 77(2): p. 115-20.
  20. Balemans, W. and W. Van Hul, The genetics of low-density lipoprotein receptor-related protein 5 in bone: a story of extremes. Endocrinology, 2007. 148(6): p. 2622-9.
  21. Hartikka, H., et al., Heterozygous mutations in the LDL receptor-related protein 5 (LRP5) gene are associated with primary osteoporosis in children. J Bone Miner Res, 2005. 20(5): p. 783-9.
  22. NORD, https://rarediseases.org/rare-diseases/hypophosphatasia/.
  23. Lanou, A.J., S.E. Berkow, and N.D. Barnard, Calcium, dairy products, and bone health in children and young adults: a reevaluation of the evidence. Pediatrics, 2005. 115(3): p. 736-43.
  24. Huncharek, M., J. Muscat, and B. Kupelnick, Impact of dairy products and dietary calcium on bone-mineral content in children: results of a meta-analysis. Bone, 2008. 43(2): p. 312-321.
  25. Winzenberg, T., et al., Effects of calcium supplementation on bone density in healthy children: meta-analysis of randomised controlled trials. BMJ, 2006. 333(7572): p. 775.
  26. Cormick, G., et al., Effect of Calcium Fortified Foods on Health Outcomes: A Systematic Review and Meta-Analysis. Nutrients, 2021. 13(2).
  27. Macdonald, H., et al., Bone strength and its determinants in pre- and early pubertal boys and girls. Bone, 2006. 39(3): p. 598-608.
  28. Tihtonen, K., et al., Calcium supplementation during pregnancy and maternal and offspring bone health: a systematic review and meta-analysis. Ann N Y Acad Sci, 2022. 1509(1): p. 23-36.
  29. Lehtonen-Veromaa, M.K., et al., Vitamin D and attainment of peak bone mass among peripubertal Finnish girls: a 3-y prospective study. Am J Clin Nutr, 2002. 76(6): p. 1446-53.
  30. Ryan, L.M., et al., Bone mineral density and vitamin D status among African American children with forearm fractures. Pediatrics, 2012. 130(3): p. e553-60.
  31. Wu, F., L.L. Laslett, and Q. Zhang, Threshold Effects of Vitamin D Status on Bone Health in Chinese Adolescents With Low Calcium Intake. J Clin Endocrinol Metab, 2015. 100(12): p. 4481-9.
  32. Karlsland Akeson, P., et al., Vitamin D Intervention and Bone: A Randomized Clinical Trial in Fair- and Dark-skinned Children at Northern Latitudes. J Pediatr Gastroenterol Nutr, 2018. 67(3): p. 388-394.
  33. Zhu, K., et al., Maternal vitamin D status during pregnancy and bone mass in offspring at 20 years of age: a prospective cohort study. J Bone Miner Res, 2014. 29(5): p. 1088-95.
  34. Ma, K., et al., Effect of Vitamin D Supplementation in Early Life on Children's Growth and Body Composition: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients, 2021. 13(2).
  35. Faje, A.T., et al., Fracture risk and areal bone mineral density in adolescent females with anorexia nervosa. Int J Eat Disord, 2014. 47(5): p. 458-66.
  36. Ackerman, K.E., et al., Fractures in Relation to Menstrual Status and Bone Parameters in Young Athletes. Med Sci Sports Exerc, 2015. 47(8): p. 1577-86.
  37. Misra, M. and A. Klibanski, Endocrine consequences of anorexia nervosa. Lancet Diabetes Endocrinol, 2014. 2(7): p. 581-92.
  38. Misra, M., et al., Bone metabolism in adolescent boys with anorexia nervosa. J Clin Endocrinol Metab, 2008. 93(8): p. 3029-36.
  39. Misra, M., et al., Effects of anorexia nervosa on clinical, hematologic, biochemical, and bone density parameters in community-dwelling adolescent girls. Pediatrics, 2004. 114(6): p. 1574-83.
  40. Misra, M., et al., Weight gain and restoration of menses as predictors of bone mineral density change in adolescent girls with anorexia nervosa-1. J Clin Endocrinol Metab, 2008. 93(4): p. 1231-7.
  41. Christo, K., et al., Bone metabolism in adolescent athletes with amenorrhea, athletes with eumenorrhea, and control subjects. Pediatrics, 2008. 121(6): p. 1127-36.
  42. Ackerman, K.E., et al., Bone microarchitecture is impaired in adolescent amenorrheic athletes compared with eumenorrheic athletes and nonathletic controls. J Clin Endocrinol Metab, 2011. 96(10): p. 3123-33.
  43. Ackerman, K.E., et al., Cortical microstructure and estimated bone strength in young amenorrheic athletes, eumenorrheic athletes and non-athletes. Bone, 2012. 51(4): p. 680-7.
  44. Pedreira, C.C., J. Maya, and M. Misra, Functional hypothalamic amenorrhea: Impact on bone and neuropsychiatric outcomes. Front Endocrinol (Lausanne), 2022. 13: p. 953180.
  45. Fedewa, M.V., et al., Celiac Disease and Bone Health in Children and Adolescents: A Systematic Review and Meta-Analysis. J Clin Densitom, 2020. 23(2): p. 200-211.
  46. Heikkila, K., et al., Celiac disease and bone fractures: a systematic review and meta-analysis. J Clin Endocrinol Metab, 2015. 100(1): p. 25-34.
  47. Webster, J., et al., Prevalence of and Risk Factors for Low Bone Mineral Density in Children With Celiac Disease. Clin Gastroenterol Hepatol, 2019. 17(8): p. 1509-1514.
  48. Fouda, M.A., et al., Evaluation and management of skeletal health in celiac disease: position statement. Can J Gastroenterol, 2012. 26(11): p. 819-29.
  49. Laakso, S., et al., Compromised peak bone mass in patients with inflammatory bowel disease--a prospective study. J Pediatr, 2014. 164(6): p. 1436-43 e1.
  50. Hidalgo, D.F., et al., Inflammatory Bowel Disease and Risk of Osteoporotic Fractures: A Meta-Analysis. Cureus, 2019. 11(9): p. e5810.
  51. Bernstein, C.N., et al., The Impact of Inflammatory Bowel Disease in Canada 2018: Extra-intestinal Diseases in IBD. J Can Assoc Gastroenterol, 2019. 2(Suppl 1): p. S73-S80.
  52. Laakso, S., et al., Impaired bone health in inflammatory bowel disease: a case-control study in 80 pediatric patients. Calcif Tissue Int, 2012. 91(2): p. 121-30.
  53. Dubner, S.E., et al., Longitudinal assessment of bone density and structure in an incident cohort of children with Crohn's disease. Gastroenterology, 2009. 136(1): p. 123-30.
  54. Semeao, E.J., et al., Bone mineral density in children and young adults with Crohn's disease. Inflamm Bowel Dis, 1999. 5(3): p. 161-6.
  55. Gordon, R.J. and C.M. Gordon, Bone Health in Pediatric Patients with IBD: What Is New? Curr Osteoporos Rep, 2021. 19(4): p. 429-435.
  56. Jin, H.Y., et al., Growth, puberty, and bone health in children and adolescents with inflammatory bowel disease. BMC Pediatr, 2021. 21(1): p. 35.
  57. Schmidt, S., et al., Longitudinal assessment of bone mineral density in children and adolescents with inflammatory bowel disease. J Pediatr Gastroenterol Nutr, 2012. 55(5): p. 511-8.
  58. Sigurdsson, G.V., et al., Bone Mass Development from Childhood into Young Adulthood in Patients with Childhood-onset Inflammatory Bowel Disease. Inflamm Bowel Dis, 2017. 23(12): p. 2215-2226.
  59. Semeao, E.J., et al., Risk factors for low bone mineral density in children and young adults with Crohn's disease. J Pediatr, 1999. 135(5): p. 593-600.
  60. Lopes, L.H., et al., Risk factors for low bone mineral density in children and adolescents with inflammatory bowel disease. Dig Dis Sci, 2008. 53(10): p. 2746-53.
  61. Aris, R.M., et al., Increased rate of fractures and severe kyphosis: sequelae of living into adulthood with cystic fibrosis. Ann Intern Med, 1998. 128(3): p. 186-93.
  62. Anabtawi, A., et al., Cystic fibrosis bone disease: Pathophysiology, assessment and prognostic implications. J Cyst Fibros, 2019. 18 Suppl 2: p. S48-S55.
  63. O'Reilly, R., et al., Severe bone demineralisation is associated with higher mortality in children with cystic fibrosis. Ir Med J, 2009. 102(2): p. 47-9.
  64. Putman, M.S., et al., Cystic fibrosis bone disease treatment: Current knowledge and future directions. J Cyst Fibros, 2019. 18 Suppl 2: p. S56-S65.
  65. Javier, R.M. and J. Jacquot, Bone disease in cystic fibrosis: what's new? Joint Bone Spine, 2011. 78(5): p. 445-50.
  66. Sermet-Gaudelus, I., et al., Update on cystic fibrosis-related bone disease: a special focus on children. Paediatr Respir Rev, 2009. 10(3): p. 134-42.
  67. Homchan, K., et al., Low bone mineral density and the severity of cholestasis in biliary atresia. World J Hepatol, 2017. 9(16): p. 746-751.
  68. Honsawek, S., et al., Relationships between OPG, RANKL, bone metabolism, and bone mineral density in biliary atresia. Pediatr Surg Int, 2009. 25(3): p. 261-7.
  69. Omran, A., et al., Sclerostin: From Molecule to Clinical Biomarker. Int J Mol Sci, 2022. 23(9).
  70. Riggs, B.L., S. Khosla, and L.J. Melton, 3rd, Sex steroids and the construction and conservation of the adult skeleton. Endocr Rev, 2002. 23(3): p. 279-302.
  71. Tan, V.P., et al., Influence of physical activity on bone strength in children and adolescents: a systematic review and narrative synthesis. J Bone Miner Res, 2014. 29(10): p. 2161-81.
  72. Mello, J.B., et al., Exercise in school Physical Education increase bone mineral content and density: Systematic review and meta-analysis. Eur J Sport Sci, 2022. 22(10): p. 1618-1629.
  73. Mergler, S., et al., Epidemiology of low bone mineral density and fractures in children with severe cerebral palsy: a systematic review. Dev Med Child Neurol, 2009. 51(10): p. 773-8.
  74. King, W., et al., Prevalence of reduced bone mass in children and adults with spastic quadriplegia. Dev Med Child Neurol, 2003. 45(1): p. 12-6.
  75. Bianchi, M.L., et al., Bone mineral density and bone metabolism in Duchenne muscular dystrophy. Osteoporos Int, 2003. 14(9): p. 761-7.
  76. Henderson, R.C., et al., Predicting low bone density in children and young adults with quadriplegic cerebral palsy. Dev Med Child Neurol, 2004. 46(6): p. 416-9.
  77. Nazif, H., et al., Bone mineral density and insulin-like growth factor-1 in children with spastic cerebral palsy. Childs Nerv Syst, 2017. 33(4): p. 625-630.
  78. Bothwell, J.E., et al., Vertebral fractures in boys with Duchenne muscular dystrophy. Clin Pediatr (Phila), 2003. 42(4): p. 353-6.
  79. Soderpalm, A.C., et al., Low bone mineral density and decreased bone turnover in Duchenne muscular dystrophy. Neuromuscul Disord, 2007. 17(11-12): p. 919-28.
  80. Perera, N., et al., Fracture in Duchenne Muscular Dystrophy: Natural History and Vitamin D Deficiency. J Child Neurol, 2016. 31(9): p. 1181-7.
  81. Tian, C., et al., Bone health measures in glucocorticoid-treated ambulatory boys with Duchenne muscular dystrophy. Neuromuscul Disord, 2016. 26(11): p. 760-767.
  82. Del Rocio Cruz-Guzman, O., et al., Muscle function and age are associated with loss of bone mineral density in Duchenne muscular dystrophy. Muscle Nerve, 2019. 59(4): p. 417-421.
  83. Crabtree, N.J., et al., Regional changes in bone area and bone mineral content in boys with duchenne muscular dystrophy receiving corticosteroid therapy. J Pediatr, 2010. 156(3): p. 450-5.
  84. Singh, A., E.K. Schaeffer, and C.W. Reilly, Vertebral Fractures in Duchenne Muscular Dystrophy Patients Managed With Deflazacort. J Pediatr Orthop, 2018. 38(6): p. 320-324.
  85. Joseph, S., et al., Fractures and Linear Growth in a Nationwide Cohort of Boys With Duchenne Muscular Dystrophy With and Without Glucocorticoid Treatment: Results From the UK NorthStar Database. JAMA Neurol, 2019. 76(6): p. 701-709.
  86. Guglieri, M., et al., Efficacy and Safety of Vamorolone vs Placebo and Prednisone Among Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. JAMA Neurol, 2022. 79(10): p. 1005-1014.
  87. Rufo, A., et al., Mechanisms inducing low bone density in Duchenne muscular dystrophy in mice and humans. J Bone Miner Res, 2011. 26(8): p. 1891-903.
  88. Wasserman, H.M., et al., Low bone mineral density and fractures are highly prevalent in pediatric patients with spinal muscular atrophy regardless of disease severity. Neuromuscul Disord, 2017. 27(4): p. 331-337.
  89. Khatri, I.A., et al., Low bone mineral density in spinal muscular atrophy. J Clin Neuromuscul Dis, 2008. 10(1): p. 11-7.
  90. Baranello, G., et al., Evolution of bone mineral density, bone metabolism and fragility fractures in Spinal Muscular Atrophy (SMA) types 2 and 3. Neuromuscul Disord, 2019. 29(7): p. 525-532.
  91. Vai, S., et al., Bone and Spinal Muscular Atrophy. Bone, 2015. 79: p. 116-20.
  92. Klein, G.L., et al., Long-term reduction in bone mass after severe burn injury in children. J Pediatr, 1995. 126(2): p. 252-6.
  93. Klein, G.L., C.B. Langman, and D.N. Herndon, Vitamin D depletion following burn injury in children: a possible factor in post-burn osteopenia. J Trauma, 2002. 52(2): p. 346-50.
  94. Drake, M.T. and S. Khosla, Hormonal and systemic regulation of sclerostin. Bone, 2017. 96: p. 8-17.
  95. Soyka, L.A., W.P. Fairfield, and A. Klibanski, Clinical review 117: Hormonal determinants and disorders of peak bone mass in children. J Clin Endocrinol Metab, 2000. 85(11): p. 3951-63.
  96. Gordon, C.M., et al., Functional Hypothalamic Amenorrhea: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab, 2017. 102(5): p. 1413-1439.
  97. Misra, M., Effects of hypogonadism on bone metabolism in female adolescents and young adults. Nat Rev Endocrinol, 2012. 8(7): p. 395-404.
  98. Tack, L.J., et al., Effects of Glucocorticoids on Bone: What we can Learn from Pediatric Endogenous Cushing's Syndrome. Horm Metab Res, 2016. 48(11): p. 764-770.
  99. Ward, L.M., Glucocorticoid-Induced Osteoporosis: Why Kids Are Different. Front Endocrinol (Lausanne), 2020. 11: p. 576.
  100. LeBlanc, C.M., et al., Incident Vertebral Fractures and Risk Factors in the First Three Years Following Glucocorticoid Initiation Among Pediatric Patients With Rheumatic Disorders. J Bone Miner Res, 2015. 30(9): p. 1667-75.
  101. Ward, L.M., et al., Osteoporotic Fractures and Vertebral Body Reshaping in Children With Glucocorticoid-treated Rheumatic Disorders. J Clin Endocrinol Metab, 2021. 106(12): p. e5195-e5207.
  102. Lee, H.S., et al., Low Bone Mineral Density at Initial Diagnosis in Children and Adolescents with Graves' Disease. J Clin Densitom, 2021. 24(2): p. 275-280.
  103. Numbenjapon, N., et al., Low cortical bone density measured by computed tomography in children and adolescents with untreated hyperthyroidism. J Pediatr, 2007. 150(5): p. 527-30.
  104. Mora, S., et al., Longitudinal changes of bone density and bone resorption in hyperthyroid girls during treatment. J Bone Miner Res, 1999. 14(11): p. 1971-7.
  105. Lakatos, P., et al., Serum interleukin-6 and bone metabolism in patients with thyroid function disorders. J Clin Endocrinol Metab, 1997. 82(1): p. 78-81.
  106. Dekker, B.L., et al., Bone Mineral Density in Adult Survivors of Pediatric Differentiated Thyroid Carcinoma: A Longitudinal Follow-Up Study. Thyroid, 2021. 31(11): p. 1707-1714.
  107. Lewiecki, E.M. and P.D. Miller, Skeletal effects of primary hyperparathyroidism: bone mineral density and fracture risk. J Clin Densitom, 2013. 16(1): p. 28-32.
  108. Wuster, C., Fracture rates in patients with growth hormone deficiency. Horm Res, 2000. 54 Suppl 1: p. 31-5.
  109. Vestergaard, P., et al., Fracture risk is increased in patients with GH deficiency or untreated prolactinomas--a case-control study. Clin Endocrinol (Oxf), 2002. 56(2): p. 159-67.
  110. Hogler, W. and N. Shaw, Childhood growth hormone deficiency, bone density, structures and fractures: scrutinizing the evidence. Clin Endocrinol (Oxf), 2010. 72(3): p. 281-9.
  111. Gahlot, M., et al., The effect of growth hormone deficiency on size-corrected bone mineral measures in pre-pubertal children. Osteoporos Int, 2012. 23(8): p. 2211-7.
  112. Baer, T.G., et al., Deficits in Bone Geometry in Growth Hormone-Deficient Prepubertal Boys Revealed by High-Resolution Peripheral Quantitative Computed Tomography. Horm Res Paediatr, 2019. 92(5): p. 293-301.
  113. Misra, M., et al., Alterations in growth hormone secretory dynamics in adolescent girls with anorexia nervosa and effects on bone metabolism. J Clin Endocrinol Metab, 2003. 88(12): p. 5615-23.
  114. Dongare-Bhor, S., et al., Muscle and bone parameters in underprivileged Indian children and adolescents with T1DM. Bone, 2020. 130: p. 115074.
  115. Wierzbicka, E., et al., Skeletal Status, Body Composition, and Glycaemic Control in Adolescents with Type 1 Diabetes Mellitus. J Diabetes Res, 2018. 2018: p. 8121634.
  116. Mitchell, D.M., et al., Elevated HbA1c Is Associated with Altered Cortical and Trabecular Microarchitecture in Girls with Type 1 Diabetes. J Clin Endocrinol Metab, 2020. 105(4): p. e1648-56.
  117. Weber, D.R., et al., Poor Glycemic Control Is Associated With Impaired Bone Accrual in the Year Following a Diagnosis of Type 1 Diabetes. J Clin Endocrinol Metab, 2019. 104(10): p. 4511-4520.
  118. Napoli, N., et al., Mechanisms of diabetes mellitus-induced bone fragility. Nat Rev Endocrinol, 2017. 13(4): p. 208-219.
  119. Meyer, H.J., et al., Bone mineral density in patients with systemic mastocytosis: correlations with clinical and histopathological features. Clin Exp Rheumatol, 2021. 39(1): p. 52-57.
  120. Rossini, M., et al., Bone mineral density, bone turnover markers and fractures in patients with indolent systemic mastocytosis. Bone, 2011. 49(4): p. 880-5.
  121. Inaba, H., et al., Bone mineral density in children with acute lymphoblastic leukemia. Cancer, 2018. 124(5): p. 1025-1035.
  122. Ghassemi, A., et al., Evaluation of Bone Mineral Density in Children with Acute Lymphoblastic Leukemia (ALL) and Non-Hodgkin's Lymphoma (NHL): Chemotherapy with/without Radiotherapy. Int J Hematol Oncol Stem Cell Res, 2016. 10(3): p. 153-60.
  123. Ward, L.M., et al., Bone Morbidity and Recovery in Children With Acute Lymphoblastic Leukemia: Results of a Six-Year Prospective Cohort Study. J Bone Miner Res, 2018. 33(8): p. 1435-1443.
  124. Vogiatzi, M.G., et al., Low bone mineral density in adolescents with beta-thalassemia. Ann N Y Acad Sci, 2005. 1054: p. 462-6.
  125. Mirhosseini, N.Z., et al., Bone-related complications of transfusion-dependent beta thalassemia among children and adolescents. J Bone Miner Metab, 2013. 31(4): p. 468-76.
  126. Karimi, M., et al., Bone mineral density in beta-thalassemia major and intermedia. Indian Pediatr, 2007. 44(1): p. 29-32.
  127. Lal, A., et al., Bone mineral density in children with sickle cell anemia. Pediatr Blood Cancer, 2006. 47(7): p. 901-6.
  128. Miller, R.G., et al., High prevalence and correlates of low bone mineral density in young adults with sickle cell disease. Am J Hematol, 2006. 81(4): p. 236-41.
  129. Adesina, O.O., et al., Height-corrected low bone density associates with severe outcomes in sickle cell disease: SCCRIP cohort study results. Blood Adv, 2019. 3(9): p. 1476-1488.
  130. Giordano, P., et al., Mechanisms of Bone Impairment in Sickle Bone Disease. Int J Environ Res Public Health, 2021. 18(4).
  131. Wesseling-Perry, K. and I.B. Salusky, Chronic kidney disease: mineral and bone disorder in children. Semin Nephrol, 2013. 33(2): p. 169-79.
  132. Meza, K., et al., Tumor necrosis factor-alpha is associated with mineral bone disorder and growth impairment in children with chronic kidney disease. Pediatr Nephrol, 2021. 36(6): p. 1579-1587.
  133. Santos, F., et al., Bone Disease in CKD in Children. Calcif Tissue Int, 2021. 108(4): p. 423-438.
  134. Valsamis, H.A., et al., Antiepileptic drugs and bone metabolism. Nutr Metab (Lond), 2006. 3: p. 36.
  135. Weng, W., H. Li, and S. Zhu, An Overlooked Bone Metabolic Disorder: Cigarette Smoking-Induced Osteoporosis. Genes (Basel), 2022. 13(5).
  136. Cheraghi, Z., et al., The effect of alcohol on osteoporosis: A systematic review and meta-analysis. Drug Alcohol Depend, 2019. 197: p. 197-202.
  137. Gaddini, G.W., et al., Alcohol: A Simple Nutrient with Complex Actions on Bone in the Adult Skeleton. Alcohol Clin Exp Res, 2016. 40(4): p. 657-71.
  138. Binkovitz, L.A. and M.J. Henwood, Pediatric DXA: technique and interpretation. Pediatr Radiol, 2007. 37(1): p. 21-31.
  139. Weber, D.R., et al., The Utility of DXA Assessment at the Forearm, Proximal Femur, and Lateral Distal Femur, and Vertebral Fracture Assessment in the Pediatric Population: 2019 ISCD Official Position. J Clin Densitom, 2019. 22(4): p. 567-589.
  140. Zemel, B.S., et al., Revised reference curves for bone mineral content and areal bone mineral density according to age and sex for black and non-black children: results of the bone mineral density in childhood study. J Clin Endocrinol Metab, 2011. 96(10): p. 3160-9.
  141. Zemel, B.S., et al., Height adjustment in assessing dual energy x-ray absorptiometry measurements of bone mass and density in children. J Clin Endocrinol Metab, 2010. 95(3): p. 1265-73.
  142. Kalkwarf, H.J., et al., Trabecular Bone Score Reference Values for Children and Adolescents According to Age, Sex, and Ancestry. J Bone Miner Res, 2022. 37(4): p. 776-785.
  143. Crabtree, N.J., et al., Vertebral fractures assessment in children: Evaluation of DXA imaging versus conventional spine radiography. Bone, 2017. 97: p. 168-174.
  144. Boskey, A.L., Bone composition: relationship to bone fragility and antiosteoporotic drug effects. Bonekey Rep, 2013. 2: p. 447.
  145. Matkovic, V. and R.P. Heaney, Calcium balance during human growth: evidence for threshold behavior. Am J Clin Nutr, 1992. 55(5): p. 992-6.
  146. Greer, F.R., N.F. Krebs, and N. American Academy of Pediatrics Committee on, Optimizing bone health and calcium intakes of infants, children, and adolescents. Pediatrics, 2006. 117(2): p. 578-85.
  147. Ross, A.C. and Institute of Medicine (U. S.). Committee to Review Dietary Reference Intakes for Vitamin D and Calcium., Dietary reference intakes : calcium, vitamin D. 2011, Washington, DC: National Academies Press. xv, 536, 1075-1115 p.
  148. Priemel, M., et al., Bone mineralization defects and vitamin D deficiency: histomorphometric analysis of iliac crest bone biopsies and circulating 25-hydroxyvitamin D in 675 patients. J Bone Miner Res, 2010. 25(2): p. 305-12.
  149. Holick, M.F., et al., Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2011. 96(7): p. 1911-30.
  150. Pappa, H.M., R.J. Grand, and C.M. Gordon, Report on the vitamin D status of adult and pediatric patients with inflammatory bowel disease and its significance for bone health and disease. Inflamm Bowel Dis, 2006. 12(12): p. 1162-74.
  151. Tangpricha, V., et al., An update on the screening, diagnosis, management, and treatment of vitamin D deficiency in individuals with cystic fibrosis: evidence-based recommendations from the Cystic Fibrosis Foundation. J Clin Endocrinol Metab, 2012. 97(4): p. 1082-93.
  152. Sacheck, J.M., et al., Impact of Three Doses of Vitamin D3 on Serum 25(OH)D Deficiency and Insufficiency in At-Risk Schoolchildren. J Clin Endocrinol Metab, 2017. 102(12): p. 4496-4505.
  153. Huybers, S., et al., Prednisolone-induced Ca2+ malabsorption is caused by diminished expression of the epithelial Ca2+ channel TRPV6. Am J Physiol Gastrointest Liver Physiol, 2007. 292(1): p. G92-7.
  154. Roizen, J.D., et al., CYP3A4 mutation causes vitamin D-dependent rickets type 3. J Clin Invest, 2018. 128(5): p. 1913-1918.
  155. Johnston, C.C., Jr., et al., Calcium supplementation and increases in bone mineral density in children. N Engl J Med, 1992. 327(2): p. 82-7.
  156. Bonjour, J.P., et al., Calcium-enriched foods and bone mass growth in prepubertal girls: a randomized, double-blind, placebo-controlled trial. J Clin Invest, 1997. 99(6): p. 1287-94.
  157. Slemenda, C.W., et al., Reduced rates of skeletal remodeling are associated with increased bone mineral density during the development of peak skeletal mass. J Bone Miner Res, 1997. 12(4): p. 676-82.
  158. Bonjour, J.P., et al., Gain in bone mineral mass in prepubertal girls 3.5 years after discontinuation of calcium supplementation: a follow-up study. Lancet, 2001. 358(9289): p. 1208-12.
  159. Chevalley, T., et al., Skeletal site selectivity in the effects of calcium supplementation on areal bone mineral density gain: a randomized, double-blind, placebo-controlled trial in prepubertal boys. J Clin Endocrinol Metab, 2005. 90(6): p. 3342-9.
  160. Matkovic, V., et al., Calcium supplementation and bone mineral density in females from childhood to young adulthood: a randomized controlled trial. Am J Clin Nutr, 2005. 81(1): p. 175-88.
  161. Winzenberg, T., et al., Effects of vitamin D supplementation on bone density in healthy children: systematic review and meta-analysis. BMJ, 2011. 342: p. c7254.
  162. Zhu, K., et al., Growth, bone mass, and vitamin D status of Chinese adolescent girls 3 y after withdrawal of milk supplementation. Am J Clin Nutr, 2006. 83(3): p. 714-21.
  163. Bailey, D.A., et al., A six-year longitudinal study of the relationship of physical activity to bone mineral accrual in growing children: the university of Saskatchewan bone mineral accrual study. J Bone Miner Res, 1999. 14(10): p. 1672-9.
  164. Janz, K.F., et al., Everyday activity predicts bone geometry in children: the iowa bone development study. Med Sci Sports Exerc, 2004. 36(7): p. 1124-31.
  165. Farr, J.N., et al., Associations of physical activity duration, frequency, and load with volumetric BMD, geometry, and bone strength in young girls. Osteoporos Int, 2011. 22(5): p. 1419-30.
  166. McKay, H., et al., Physical activity positively predicts bone architecture and bone strength in adolescent males and females. Acta Paediatr, 2011. 100(1): p. 97-101.
  167. Gabel, L., et al., Physical Activity, Sedentary Time, and Bone Strength From Childhood to Early Adulthood: A Mixed Longitudinal HR-pQCT study. J Bone Miner Res, 2017. 32(7): p. 1525-1536.
  168. Baxter-Jones, A.D., et al., A longitudinal study of the relationship of physical activity to bone mineral accrual from adolescence to young adulthood. Bone, 2008. 43(6): p. 1101-7.
  169. Erlandson, M.C., et al., Higher premenarcheal bone mass in elite gymnasts is maintained into young adulthood after long-term retirement from sport: a 14-year follow-up. J Bone Miner Res, 2012. 27(1): p. 104-10.
  170. Behringer, M., et al., Effects of weight-bearing activities on bone mineral content and density in children and adolescents: a meta-analysis. J Bone Miner Res, 2014. 29(2): p. 467-78.
  171. Courteix, D., et al., Cumulative effects of calcium supplementation and physical activity on bone accretion in premenarchal children: a double-blind randomised placebo-controlled trial. Int J Sports Med, 2005. 26(5): p. 332-8.
  172. Bass, S.L., et al., Exercise and calcium combined results in a greater osteogenic effect than either factor alone: a blinded randomized placebo-controlled trial in boys. J Bone Miner Res, 2007. 22(3): p. 458-64.
  173. Deere, K., et al., A cross-sectional study of the relationship between cortical bone and high-impact activity in young adult males and females. J Clin Endocrinol Metab, 2012. 97(10): p. 3734-43.
  174. Mitchell, J.A., et al., Physical Activity Benefits the Skeleton of Children Genetically Predisposed to Lower Bone Density in Adulthood. J Bone Miner Res, 2016. 31(8): p. 1504-12.
  175. Caulton, J.M., et al., A randomised controlled trial of standing programme on bone mineral density in non-ambulant children with cerebral palsy. Arch Dis Child, 2004. 89(2): p. 131-5.
  176. Chad, K.E., et al., The effect of a weight-bearing physical activity program on bone mineral content and estimated volumetric density in children with spastic cerebral palsy. J Pediatr, 1999. 135(1): p. 115-7.
  177. Swolin-Eide, D. and P. Magnusson, Does Whole-Body Vibration Treatment Make Children's Bones Stronger? Curr Osteoporos Rep, 2020. 18(5): p. 471-479.
  178. Ward, K., et al., Low magnitude mechanical loading is osteogenic in children with disabling conditions. J Bone Miner Res, 2004. 19(3): p. 360-9.
  179. Ruck, J., G. Chabot, and F. Rauch, Vibration treatment in cerebral palsy: A randomized controlled pilot study. J Musculoskelet Neuronal Interact, 2010. 10(1): p. 77-83.
  180. Wren, T.A., et al., Effect of high-frequency, low-magnitude vibration on bone and muscle in children with cerebral palsy. J Pediatr Orthop, 2010. 30(7): p. 732-8.
  181. El-Shamy, S.M., Effect of whole-body vibration on muscle strength and balance in diplegic cerebral palsy: a randomized controlled trial. Am J Phys Med Rehabil, 2014. 93(2): p. 114-21.
  182. Saquetto, M., et al., The effects of whole body vibration on mobility and balance in children with cerebral palsy: a systematic review with meta-analysis. J Musculoskelet Neuronal Interact, 2015. 15(2): p. 137-44.
  183. Bianchi, M.L., et al., Low-Intensity Vibration Protects the Weight-Bearing Skeleton and Suppresses Fracture Incidence in Boys With Duchenne Muscular Dystrophy: A Prospective, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. JBMR Plus, 2022. 6(11): p. e10685.
  184. Soderpalm, A.C., et al., Whole body vibration therapy in patients with Duchenne muscular dystrophy--a prospective observational study. J Musculoskelet Neuronal Interact, 2013. 13(1): p. 13-8.
  185. Petryk, A., et al., Feasibility and tolerability of whole-body, low-intensity vibration and its effects on muscle function and bone in patients with dystrophinopathies: a pilot study. Muscle Nerve, 2017. 55(6): p. 875-883.
  186. Hoyer-Kuhn, H., et al., A specialized rehabilitation approach improves mobility in children with osteogenesis imperfecta. J Musculoskelet Neuronal Interact, 2014. 14(4): p. 445-53.
  187. Hogler, W., et al., The Effect of Whole Body Vibration Training on Bone and Muscle Function in Children With Osteogenesis Imperfecta. J Clin Endocrinol Metab, 2017. 102(8): p. 2734-2743.
  188. Matute-Llorente, A., et al., Effect of whole-body vibration training on bone mass in adolescents with and without Down syndrome: a randomized controlled trial. Osteoporos Int, 2016. 27(1): p. 181-91.
  189. Ward, L.M., Part 2: When Should Bisphosphonates Be Used in Children with Chronic Illness Osteoporosis? Curr Osteoporos Rep, 2021. 19(3): p. 289-297.
  190. Bishop, N., et al., Fracture prediction and the definition of osteoporosis in children and adolescents: the ISCD 2013 Pediatric Official Positions. J Clin Densitom, 2014. 17(2): p. 275-80.
  191. Simm, P.J., et al., Consensus guidelines on the use of bisphosphonate therapy in children and adolescents. J Paediatr Child Health, 2018. 54(3): p. 223-233.
  192. LeBoff, M.S., et al., The clinician's guide to prevention and treatment of osteoporosis. Osteoporos Int, 2022. 33(10): p. 2049-2102.
  193. Larson, C.M. and R.C. Henderson, Bone mineral density and fractures in boys with Duchenne muscular dystrophy. J Pediatr Orthop, 2000. 20(1): p. 71-4.
  194. Yildiz, S., et al., Retrospective Analysis of Fractures and Factors Causing Ambulation Loss After Lower Limb Fractures in Duchenne Muscular Dystrophy. Am J Phys Med Rehabil, 2020. 99(9): p. 789-794.
  195. Nasomyont, N., L.N. Hornung, and H. Wasserman, Intravenous bisphosphonate therapy in children with spinal muscular atrophy. Osteoporos Int, 2020. 31(5): p. 995-1000.
  196. DeBoer, M.D., et al., Increases in IGF-1 After Anti-TNF-alpha Therapy Are Associated With Bone and Muscle Accrual in Pediatric Crohn Disease. J Clin Endocrinol Metab, 2018. 103(3): p. 936-945.
  197. Drake, M.T., B.L. Clarke, and S. Khosla, Bisphosphonates: mechanism of action and role in clinical practice. Mayo Clin Proc, 2008. 83(9): p. 1032-45.
  198. Papapoulos, S.E. and S.C. Cremers, Prolonged bisphosphonate release after treatment in children. N Engl J Med, 2007. 356(10): p. 1075-6.
  199. Cremers, S.C., G. Pillai, and S.E. Papapoulos, Pharmacokinetics/pharmacodynamics of bisphosphonates: use for optimisation of intermittent therapy for osteoporosis. Clin Pharmacokinet, 2005. 44(6): p. 551-70.
  200. Ward, L.M., et al., Alendronate for the treatment of pediatric osteogenesis imperfecta: a randomized placebo-controlled study. J Clin Endocrinol Metab, 2011. 96(2): p. 355-64.
  201. Houston, C., K. Mathews, and A. Shibli-Rahhal, Bone density and alendronate effects in Duchenne muscular dystrophy patients. Muscle Nerve, 2014. 49(4): p. 506-11.
  202. Ward, L.M., et al., Single-dose pharmacokinetics and tolerability of alendronate 35- and 70-milligram tablets in children and adolescents with osteogenesis imperfecta type I. J Clin Endocrinol Metab, 2005. 90(7): p. 4051-6.
  203. Seikaly, M.G., et al., Impact of alendronate on quality of life in children with osteogenesis imperfecta. J Pediatr Orthop, 2005. 25(6): p. 786-91.
  204. Dimeglio, L.A., et al., A comparison of oral and intravenous bisphosphonate therapy for children with osteogenesis imperfecta. J Pediatr Endocrinol Metab, 2005. 18(1): p. 43-53.
  205. Rudge, S., et al., Effects of once-weekly oral alendronate on bone in children on glucocorticoid treatment. Rheumatology (Oxford), 2005. 44(6): p. 813-8.
  206. Bianchi, M.L., et al., Efficacy and safety of alendronate for the treatment of osteoporosis in diffuse connective tissue diseases in children: a prospective multicenter study. Arthritis Rheum, 2000. 43(9): p. 1960-6.
  207. Hawker, G.A., et al., Alendronate in the treatment of low bone mass in steroid-treated boys with Duchennes muscular dystrophy. Arch Phys Med Rehabil, 2005. 86(2): p. 284-8.
  208. Tian, C., et al., Oral bisphosphonate treatment in patients with Duchenne muscular dystrophy on long term glucocorticoid therapy. Neuromuscul Disord, 2020. 30(7): p. 599-610.
  209. Paksu, M.S., et al., Osteopenia in children with cerebral palsy can be treated with oral alendronate. Childs Nerv Syst, 2012. 28(2): p. 283-6.
  210. Bianchi, M.L., et al., Treatment of low bone density in young people with cystic fibrosis: a multicentre, prospective, open-label observational study of calcium and calcifediol followed by a randomised placebo-controlled trial of alendronate. Lancet Respir Med, 2013. 1(5): p. 377-85.
  211. MacDonald, P., et al., Safety and Efficacy of Alendronate to Treat Osteopenia in Children During Therapy for Acute Lymphoblastic Leukemia: A Retrospective Cohort Study of Sequential Outcomes. J Pediatr Hematol Oncol, 2023. 45(4): p. 200-206.
  212. Gilchrist, N.L., et al., Alendronate prevents bone loss in patients with acute spinal cord injury: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab, 2007. 92(4): p. 1385-90.
  213. El-Husseini, A.A., et al., Treatment of osteopenia and osteoporosis in renal transplant children and adolescents. Pediatr Transplant, 2004. 8(4): p. 357-61.
  214. Bishop, N., et al., Risedronate in children with osteogenesis imperfecta: a randomised, double-blind, placebo-controlled trial. Lancet, 2013. 382(9902): p. 1424-32.
  215. Rauch, F., et al., Risedronate in the treatment of mild pediatric osteogenesis imperfecta: a randomized placebo-controlled study. J Bone Miner Res, 2009. 24(7): p. 1282-9.
  216. Bishop, N., et al., A randomized, controlled dose-ranging study of risedronate in children with moderate and severe osteogenesis imperfecta. J Bone Miner Res, 2010. 25(1): p. 32-40.
  217. Srinivasan, R., et al., Prophylactic oral bisphosphonate therapy in duchenne muscular dystrophy. Muscle Nerve, 2016. 54(1): p. 79-85.
  218. Iwasaki, T., et al., Secondary osteoporosis in long-term bedridden patients with cerebral palsy. Pediatr Int, 2008. 50(3): p. 269-75.
  219. Cohran, V., et al., Oral risedronate sodium improves bone mineral density in non-ambulatory patients: a randomized, double-blind, placebo controlled trial. J Pediatr Rehabil Med, 2013. 6(2): p. 85-93.
  220. Rauch, F., et al., Osteogenesis imperfecta types I, III, and IV: effect of pamidronate therapy on bone and mineral metabolism. J Clin Endocrinol Metab, 2003. 88(3): p. 986-92.
  221. Glorieux, F.H., et al., Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. N Engl J Med, 1998. 339(14): p. 947-52.
  222. Plotkin, H., et al., Pamidronate treatment of severe osteogenesis imperfecta in children under 3 years of age. J Clin Endocrinol Metab, 2000. 85(5): p. 1846-50.
  223. Acott, P.D., et al., Pamidronate treatment of pediatric fracture patients on chronic steroid therapy. Pediatr Nephrol, 2005. 20(3): p. 368-73.
  224. Bachrach, S.J., et al., Decreased fracture incidence after 1 year of pamidronate treatment in children with spastic quadriplegic cerebral palsy. Dev Med Child Neurol, 2010. 52(9): p. 837-42.
  225. Henderson, R.C., et al., Bisphosphonates to treat osteopenia in children with quadriplegic cerebral palsy: a randomized, placebo-controlled clinical trial. J Pediatr, 2002. 141(5): p. 644-51.
  226. Plotkin, H., et al., Low doses of pamidronate to treat osteopenia in children with severe cerebral palsy: a pilot study. Dev Med Child Neurol, 2006. 48(9): p. 709-12.
  227. Goldbloom, E.B., E.A. Cummings, and M. Yhap, Osteoporosis at presentation of childhood ALL: management with pamidronate. Pediatr Hematol Oncol, 2005. 22(7): p. 543-50.
  228. Baroncelli, G.I., et al., Pamidronate treatment stimulates the onset of recovery phase reducing fracture rate and skeletal deformities in patients with idiopathic juvenile osteoporosis: comparison with untreated patients. J Bone Miner Metab, 2013. 31(5): p. 533-43.
  229. Klein, G.L., et al., The efficacy of acute administration of pamidronate on the conservation of bone mass following severe burn injury in children: a double-blind, randomized, controlled study. Osteoporos Int, 2005. 16(6): p. 631-5.
  230. Barros, E.R., et al., Safety and efficacy of a 1-year treatment with zoledronic acid compared with pamidronate in children with osteogenesis imperfecta. J Pediatr Endocrinol Metab, 2012. 25(5-6): p. 485-91.
  231. Ooi, H.L., et al., Zoledronic acid improves bone mineral density in pediatric spinal cord injury. J Bone Miner Res, 2012. 27(7): p. 1536-40.
  232. Li, L.J., et al., Effects of zoledronic acid on vertebral shape of children and adolescents with osteogenesis imperfecta. Bone, 2019. 127: p. 164-171.
  233. Saraff, V., et al., Efficacy and treatment costs of zoledronate versus pamidronate in paediatric osteoporosis. Arch Dis Child, 2018. 103(1): p. 92-94.
  234. Vuorimies, I., et al., Zoledronic acid treatment in children with osteogenesis imperfecta. Horm Res Paediatr, 2011. 75(5): p. 346-53.
  235. Brown, J.J. and M.R. Zacharin, Safety and efficacy of intravenous zoledronic acid in paediatric osteoporosis. J Pediatr Endocrinol Metab, 2009. 22(1): p. 55-63.
  236. Ward, L.M., et al., Zoledronic Acid vs Placebo in Pediatric Glucocorticoid-induced Osteoporosis: A Randomized, Double-blind, Phase 3 Trial. J Clin Endocrinol Metab, 2021. 106(12): p. e5222-e5235.
  237. Zacharin, M., et al., Randomized Controlled Trial Evaluating the Use of Zoledronic Acid in Duchenne Muscular Dystrophy. J Clin Endocrinol Metab, 2021. 106(8): p. 2328-2342.
  238. Granild-Jensen, J.B., et al., Cerebral palsy and bisphosphonates - and what can be learned from other types of secondary osteoporosis in children: A scoping review. Acta Paediatr, 2023. 112(4): p. 617-629.
  239. Wiedemann, A., et al., Annual Injection of Zoledronic Acid Improves Bone Status in Children with Cerebral Palsy and Rett Syndrome. Calcif Tissue Int, 2019. 104(4): p. 355-363.
  240. Gatti, D., et al., Intravenous neridronate in children with osteogenesis imperfecta: a randomized controlled study. J Bone Miner Res, 2005. 20(5): p. 758-63.
  241. Maines, E., et al., Children and adolescents treated with neridronate for osteogenesis imperfecta show no evidence of any osteonecrosis of the jaw. J Bone Miner Metab, 2012. 30(4): p. 434-8.
  242. Celli, M., et al., Clinical and biochemical response to neridronate treatment in a patient with osteoporosis-pseudoglioma syndrome (OPPG). Osteoporos Int, 2017. 28(11): p. 3277-3280.
  243. Adami, S., et al., Intravenous neridronate in adults with osteogenesis imperfecta. J Bone Miner Res, 2003. 18(1): p. 126-30.
  244. Gatti, D., et al., Intravenous bisphosphonate therapy increases radial width in adults with osteogenesis imperfecta. J Bone Miner Res, 2005. 20(8): p. 1323-6.
  245. Dwan, K., et al., Bisphosphonate therapy for osteogenesis imperfecta. Cochrane Database Syst Rev, 2016. 10(10): p. CD005088.
  246. Rauch, F., et al., The effects of intravenous pamidronate on the bone tissue of children and adolescents with osteogenesis imperfecta. J Clin Invest, 2002. 110(9): p. 1293-9.
  247. Black, D.M., et al., Bone mineral density as a surrogate biomarker for fracture risk reduction - Authors' reply. Lancet Diabetes Endocrinol, 2020. 8(11): p. 876.
  248. Nasomyont, N., et al., The effect of oral bisphosphonate therapy on vertebral morphometry and fractures in patients with Duchenne muscular dystrophy and glucocorticoid-induced osteoporosis. Muscle Nerve, 2021. 64(6): p. 710-716.
  249. Ward, L.M., V.N. Konji, and J. Ma, The management of osteoporosis in children. Osteoporos Int, 2016. 27(7): p. 2147-2179.
  250. DiMeglio, L.A. and M. Peacock, Two-year clinical trial of oral alendronate versus intravenous pamidronate in children with osteogenesis imperfecta. J Bone Miner Res, 2006. 21(1): p. 132-40.
  251. Loizidou, A., S. Andronikou, and C.P. Burren, Pamidronate "zebra lines": A treatment timeline. Radiol Case Rep, 2017. 12(4): p. 850-853.
  252. Rauch, F., et al., Long-bone changes after pamidronate discontinuation in children and adolescents with osteogenesis imperfecta. Bone, 2007. 40(4): p. 821-7.
  253. Biggin, A., et al., Fracture during intravenous bisphosphonate treatment in a child with osteogenesis imperfecta: an argument for a more frequent, low-dose treatment regimen. Horm Res Paediatr, 2014. 81(3): p. 204-10.
  254. Sbrocchi, A.M., et al., The use of intravenous bisphosphonate therapy to treat vertebral fractures due to osteoporosis among boys with Duchenne muscular dystrophy. Osteoporos Int, 2012. 23(11): p. 2703-11.
  255. George, S., et al., Short-Term Safety of Zoledronic Acid in Young Patients With Bone Disorders: An Extensive Institutional Experience. J Clin Endocrinol Metab, 2015. 100(11): p. 4163-71.
  256. Munns, C.F., et al., Acute phase response and mineral status following low dose intravenous zoledronic acid in children. Bone, 2007. 41(3): p. 366-70.
  257. Srivastava, T., et al., Serum 25-hydroxyvitamin D level and acute-phase reaction following initial intravenous bisphosphonate. J Bone Miner Res, 2011. 26(2): p. 437-8.
  258. Nasomyont, N., et al., Outcomes following intravenous bisphosphonate infusion in pediatric patients: A 7-year retrospective chart review. Bone, 2019. 121: p. 60-67.
  259. Viollet, L., et al., Utility of cystatin C to monitor renal function in Duchenne muscular dystrophy. Muscle Nerve, 2009. 40(3): p. 438-42.
  260. Munns, C.F., et al., Delayed osteotomy but not fracture healing in pediatric osteogenesis imperfecta patients receiving pamidronate. J Bone Miner Res, 2004. 19(11): p. 1779-86.
  261. Starr, J., Y.K.D. Tay, and E. Shane, Current Understanding of Epidemiology, Pathophysiology, and Management of Atypical Femur Fractures. Curr Osteoporos Rep, 2018. 16(4): p. 519-529.
  262. Vuorimies, I., et al., Bisphosphonate Treatment and the Characteristics of Femoral Fractures in Children With Osteogenesis Imperfecta. J Clin Endocrinol Metab, 2017. 102(4): p. 1333-1339.
  263. Trejo, P., et al., Diaphyseal Femur Fractures in Osteogenesis Imperfecta: Characteristics and Relationship With Bisphosphonate Treatment. J Bone Miner Res, 2017. 32(5): p. 1034-1039.
  264. Boyce, A.M., et al., A Subtrochanteric Femoral Stress Fracture following Bisphosphonate Treatment in an Adolescent Girl. Horm Res Paediatr, 2017. 87(1): p. 69-72.
  265. Neal, T.W. and T. Schlieve, Medication-Related Osteonecrosis of the Jaws in the Pediatric Population. J Oral Maxillofac Surg, 2022. 80(10): p. 1686-1690.
  266. Brown, J.J., L. Ramalingam, and M.R. Zacharin, Bisphosphonate-associated osteonecrosis of the jaw: does it occur in children? Clin Endocrinol (Oxf), 2008. 68(6): p. 863-7.
  267. Whyte, M.P., et al., Bisphosphonate-induced osteopetrosis. N Engl J Med, 2003. 349(5): p. 457-63.
  268. Whyte, M.P., et al., Drug-induced osteopetrosis. Bone, 2023. 173: p. 116788.
  269. Levy, S., et al., Pregnancy outcome following in utero exposure to bisphosphonates. Bone, 2009. 44(3): p. 428-30.
  270. Tsourdi, E., et al., Fracture risk and management of discontinuation of denosumab therapy: a systematic review and position statement by ECTS. J Clin Endocrinol Metab, 2020.
  271. Kim, A.S., C.M. Girgis, and M.M. McDonald, Osteoclast Recycling and the Rebound Phenomenon Following Denosumab Discontinuation. Curr Osteoporos Rep, 2022. 20(6): p. 505-515.
  272. Gossai, N., et al., Critical hypercalcemia following discontinuation of denosumab therapy for metastatic giant cell tumor of bone. Pediatr Blood Cancer, 2015. 62(6): p. 1078-80.
  273. Setsu, N., et al., Severe hypercalcemia following denosumab treatment in a juvenile patient. J Bone Miner Metab, 2016. 34(1): p. 118-22.
  274. Trejo, P., F. Rauch, and L. Ward, Hypercalcemia and hypercalciuria during denosumab treatment in children with osteogenesis imperfecta type VI. J Musculoskelet Neuronal Interact, 2018. 18(1): p. 76-80.
  275. Uday, S., et al., Osteonecrosis of the Jaw and Rebound Hypercalcemia in Young People Treated With Denosumab for Giant Cell Tumor of Bone. J Clin Endocrinol Metab, 2018. 103(2): p. 596-603.
  276. Seale, E., et al., Mitigating the Denosumab-Induced Rebound Phenomenon with Alternating Short- and Long-Acting Anti-resorptive Therapy in a Young Boy with Severe OI Type VI. Calcif Tissue Int, 2023. 112(5): p. 613-620.
  277. Semler, O., et al., First use of the RANKL antibody denosumab in osteogenesis imperfecta type VI. J Musculoskelet Neuronal Interact, 2012. 12(3): p. 183-8.
  278. Ward, L., et al., Osteogenesis Imperfecta Type VI in Individuals from Northern Canada. Calcif Tissue Int, 2016. 98(6): p. 566-72.
  279. Chawla, S., et al., Safety and efficacy of denosumab for adults and skeletally mature adolescents with giant cell tumour of bone: interim analysis of an open-label, parallel-group, phase 2 study. Lancet Oncol, 2013. 14(9): p. 901-8.
  280. Ferriero, K., et al., Case Report: Safety and Efficacy of Denosumab in Four Children With Noonan Syndrome With Multiple Giant Cell Lesions of the Jaw. Front Pediatr, 2020. 8: p. 515.
  281. Vanderniet, J.A., et al., Denosumab for central giant cell granuloma in an Australian tertiary paediatric centre. Bone, 2022. 159: p. 116395.
  282. Jacobsen, C.M., Application of anti-Sclerostin therapy in non-osteoporosis disease models. Bone, 2017. 96: p. 18-23.
  283. Ward, L.M. and F. Rauch, Anabolic Therapy for the Treatment of Osteoporosis in Childhood. Curr Osteoporos Rep, 2018. 16(3): p. 269-276.
  284. McClung, M.R. and A. Grauer, Romosozumab in postmenopausal women with osteopenia. N Engl J Med, 2014. 370(17): p. 1664-5.
  285. Cosman, F., et al., Romosozumab Treatment in Postmenopausal Women with Osteoporosis. N Engl J Med, 2016. 375(16): p. 1532-1543.
  286. Glorieux, F.H., et al., BPS804 Anti-Sclerostin Antibody in Adults With Moderate Osteogenesis Imperfecta: Results of a Randomized Phase 2a Trial. J Bone Miner Res, 2017. 32(7): p. 1496-1504.
  287. Nasomyont, N., et al., Safety and efficacy of teriparatide treatment for severe osteoporosis in patients with Duchenne muscular dystrophy. Osteoporos Int, 2020. 31(12): p. 2449-2459.
  288. Black, D.M., et al., One year of alendronate after one year of parathyroid hormone (1-84) for osteoporosis. N Engl J Med, 2005. 353(6): p. 555-65.
  289. Leder, B.Z., et al., Effects of teriparatide treatment and discontinuation in postmenopausal women and eugonadal men with osteoporosis. J Clin Endocrinol Metab, 2009. 94(8): p. 2915-21.

 

 

 

 

I agree w/ Madhu. Also, I think, in general, people are doing less of a PE, so this is a reminder

 

Carney Complex

ABSTRACT

 

Carney complex (CNC) is a rare dominantly inherited syndrome of multiple neoplasias combined with cardio-cutaneous manifestations. Approximately 70% of index cases have a familial history, while the remaining 30% have a de novo germline mutation. Hitherto, two loci have been principally involved in the genetics of CNC: the CNC1 gene, located on 17q22-24, which is coding the regulatory subunit (R1a) of the protein kinase A (PRKAR1A) and is responsible for 2/3 of cases, whereas the putative “CNC2” gene at the 2p16 locus has not been identified as yet. As most of the identified PRKAR1A mutations are nonsense and lead to a lack of detectable mutant protein, no genotype-phenotype correlations are generally observed. Cutaneous lesions (lentigines, nevi, and myxomas), although with minimal clinical impact, are the most common and occasionally specific findings, assisting early diagnosis. Cardiac myxomas show an atypical presentation and contribute substantially to mortality. Among several associated endocrine neoplasias, Primary Pigmented Nodular Adrenal Dysplasia is the one most frequently observed, followed by thyroid nodules, somatomammotrope adenomas, and testicular tumors. The diagnosis is principally set by 12 major clinical criteria and 2 supplemental criteria, including molecular testing and family history. Molecular testing, which has a mutation detection rate of approximately 60%, cannot currently be recommended for all patients. If testing is performed and a mutation is detected, genetic screening is recommended for first-degree relatives. Surveillance for all the manifestations of CNC should be performed at least annually, starting in infancy. As CNC is generated by a constitutional genetic defect, no etiologic therapy is available yet. The therapeutic approach should target each clinical manifestation and treat accordingly.

 

INTRODUCTION - HISTORICAL OVERVIEW

 

Carney complex (CNC - Online Mendelian Inheritance in Man 160980, 608837) is a dominantly inherited syndrome of multiple neoplasias combined with cardiocutaneous manifestations. The neoplastic lesions are both endocrine (adrenal, pituitary, thyroid, testicular tumors) and non-endocrine (myxomas, schwannomas). The skin lesions are divided into two major types: a) pigmented such as lentigines and blue nevi that can be observed on the face, neck, and trunk and b) not pigmented such as cutaneous myxomas (1) (Figure 1).

Figure 1. Spotty pigmentation of the face. With permission http://ugen.nichd.nih.gov.

This syndrome was first described by J. Carney in 1985, as “the complex of myxomas, spotty pigmentation, and endocrine overactivity”. In the original study, 40 patients were included, and a familial distribution was reported in 10 of them. Additional evidence for unifying this coexistence of otherwise rare conditions in an inherited clinical entity was the young age at presentation and the unusual type of involvement of most affected sites, which tended to be multicentric (heart and skin) and bilateral in paired organs (adrenal, breast, and testis) (2). One year later Carney reported observations consistent with Mendelian dominant inheritance of the syndrome (3) that in the meanwhile was designated as “Carney complex” (CNC) by Bain (4). This new entity included patients manifesting cardiocutaneous lesions, previously diagnosed as LAMB (lentigines, atrial myxoma, mucocutaneous myxoma, blue nevi) (5) and NAME (nevi, atrial myxoma, myxoid neurofibroma, ephelides) (6).

 

In 1996, linkage analysis studies by Stratakis et al. (7) demonstrated a locus potentially linked to CNC on chromosome 2p16, in proximity to the gene encoding proopiomelanocortin and the DNA-mismatch repair genes hMSH2 and hMSH6. However, the syndrome was later shown to be genetically heterogeneous (8), and in 1998, a second possible locus located on chromosome 17q2 was detected (9). In 2000, two different research teams demonstrated that germline mutations in the gene coding the alpha regulatory subunit (R1a) of protein kinase A (PKAR1A) located on the locus 17q22-24 were responsible for several phenotypes of CNC (10,11). More recently, mutations of other genes, which encode the catalytic subunit α or β of the PKA, and phosphodiesterases 11A and 8B have been reported in CNC patients (1).

 

Nowadays, diagnosis of the syndrome is feasible in clinically asymptomatic patients by commercially available molecular genetic assays and next-generation sequencing techniques. Notably, Carney’s complex should not be confused with Carney’s triad, a completely different entity consisting of the triad of gastric leiomyosarcoma, pulmonary chondroma, and extra-adrenal paraganglioma.

 

EPIDEMIOLOGY AND INHERITANCE

 

Carney’s complex is a rare disease and the majority of cases have been registered by the NIH-Mayo Clinic (USA) and the Cochin Center (Paris, France) consortium (12,13). Approximately 70% of individuals diagnosed with CNC have a familial history, while the remaining 30% present as a de novo germline mutation. In large series, a predilection of female over male gender has been observed (63% vs. 37% respectively), whereas there is no apparent predilection concerning ethnicity (14).

 

CNC is inherited as a dominant trait, although transmission through a female affected parent is almost 5-fold more frequent than the male. A possible explanation for this discrepancy might be the fact that male patients often harbor Large Cell Calcified Sertoli Cell Tumors (LCCSCT), which may hamper fertility (15). Moreover, data from animal models correlate haploinsufficiency at the PRKR1A gene locus with male infertility, independently of LCCSCT (16). The median age of diagnosis is 20 years; however, in many cases, a diagnostic delay of decades is reported. In general, the penetrance of CNC is 70%-80% by the age of 40 years, as clinical manifestations accumulate during the lifespan. The maximum number of affected generations reported in one kindred is 5 (9).

 

MOLECULAR GENETICS AND PATHOPHYSIOLOGY

 

 Hitherto, two loci have been principally involved in the genetics of CNC: 17q22-24 and 2p16.

 

CNC1 Gene

 

The CNC1 gene, located on 17q22-24, is 21 kb long and contains 11 exons, coding the regulatory subunit (R1a) of the protein kinase A (PRKAR1A), a protein of 384 amino acids (17). Protein Kinase A (PKA) is an enzyme involved in G protein-coupled intracellular pathways and serves as a mediator of c-AMP actions promoting cell metabolism, proliferation, and apoptosis. Its quaternary structure consists of 4 peptide chains that form a tetramer of two regulatory (R) subunits, each bound to one catalytic (C) subunit (18). So far four subtypes of regulatory (RIα, RIβ, RIIα, and RIIβ) and four subtypes of catalytic subunits (Cα, Cβ, Cγ, and Prkx) have been identified. A corresponding gene is coding each R (PRKR1A, PRKR1B, PRKR2A, PRKR2B) and each C subunit (PRKACA, PRKACB, PRKACG, PRKX) respectively (19). When c-AMP binds to the regulatory subunits, their conformation is altered, causing the dissociation of each active C subunit from the dimer with the corresponding R subunit. The free catalytic subunits then phosphorylate serine and threonine residues of proteins critical to the activation of downstream processes, such as cAMP response-binding protein (CREB) (Figure 2).

 

Figure 2. The G protein-coupled intracellular pathways and the defect in CNC patients: PRKAR1A mutations result in deficient / inefficient regulatory subunits, resulting in constitutional activation of C subunits (http://prkar1a.nichd.nih.gov).

 

Heterozygous inactivating mutations of PRKAR1A have been detected in more than 70% of affected individuals. Interestingly, in patients presenting with Cushing's syndrome (CS) this frequency rises to about 80%. (20). Up to date, 140 different PRKAR1A mutations have been registered at the CNC consortium database (http://prkar1a.nichd.nih.gov), and they are distributed among the 11 exons of the PRKAR1A gene, showing a predilection for exons 2, 3, 5 and 7, which are more often mutated, whereas exon 1, is non-coding and rarely mutated.  Most of them are family or patient-specific; however, certain hot-spot mutations have been identified such as the  c.709- 7del6 in intron 7, c.491-492delTG in exon 5, and c.82C > T in exon 2 (21).

 

The penetrance for CNC due to PRKAR1A mutations is higher than that encountered in CNC due to other genetic defects, reaching 98% (12). The vast majority of mutations (83%) lead to a premature stop codon (nonsense) and thus, short mutant mRNAs that are eliminated by selective degradation, a phenomenon known as nonsense-mediated mRNA decay (NMD) (17). The result is a lack of detectable mutant protein and a reduction of RIα protein levels by 50%. The rest of the mutations (17%) result in the expression of an altered protein (missense) that may be associated with more severe phenotypes (22). Large PRKAR1A deletions have also been detected in a proportion of CNC patients, who also express a more severe phenotype with unusual features. These deletions are more prominent (21.6%) among patients negative by conventional Sanger sequencing, rendering array-based studies necessary for diagnostic confirmation of such cases (23). The structure of the PRKR1A gene and the location of detected mutations are shown in Figure 3.

 

Figure 3. Schematic presentation of the PRKR1A gene and detected mutations in relation to their exon location.

 

Germline haploinsufficiency of PRKAR1A leads to a deficiency of the R1a subunits, which in turn results in enhanced intracellular signaling by PKA due to unhindered activation of the catalytic (C) subunits, as evidenced by an almost 2-fold greater response to c-AMP in CNC tumors and cell lines (24,25). How this PKA overactivity leads to tumor development has not been fully elucidated. According to previous studies, PKA-enhanced activity may trigger pathways that favor cell proliferation as the upregulation of D-type cyclins (26) or activation of the mTOR pathway (27). Recent studies in adrenocortical cell lines have confirmed the accumulation of cyclin D1 and further suggest Bcl-xL upregulation, which is associated with resistance to apoptosis (28). Consistent with the Knudson two-hit model of hereditary tumorigenesis, PRKAR1A haploinsufficiency (first hit) has been considered as a predisposition for tumorigenesis, which when combined with loss of heterozygosity (LOH) at 17q22- 24 (second hit), may lead to the development of tumors in CNC patients (29). Interestingly, tumors that do not present inactivation of the remaining wild-type allele have also been described, implying that the coexistence of PRKAR1A haploinsufficiency with defects of other tumor suppressor genes or proto-oncogenes may act synergistic for tumorigenesis (30). Accordingly, activating somatic mutations of the beta-catenin gene (CTNNB1) have been detected in adrenocortical tumors of CNC patients, carriers of a PRKAR1A mutation (31).

 

These findings were supported by experiments on Prkar1a +/- knockout mice, the genotypic animal model of Carney’s complex. These mice were developed by inserting an antisense transgene for Prkar1a exon 2 and present with many of the manifestations of CNC, such as adrenocortical hyperplasia with cortisol hypersecretion, thyroid follicular neoplasia non-pigmented schwannomas and bone lesions (32). On the contrary, mice with complete loss of Prkar1a were not viable as this genotype leads to early embryonic demise due to failure of heart tube development (33). Eventually, the development of thyroid and pituitary cell tumors as well as heart myxomas was achieved by inducing tissue-specific complete ablation of Prkar1a (34,35). Moreover, mice double heterozygote for Prkar1a and Trp53 or Rb1 developed more sarcomas and grew more, and larger pituitary and thyroid tumors compared to the single Prkar1a heterozygotes (36).

 

Other Loci

 

Approximately 30% of the families affected with CNC are not related to defective PRKAR1A. The putative “CNC2” gene located at the 2p16 locus is linked to the majority of them; nevertheless, it has not yet been identified. These patients present with a milder phenotype, they are diagnosed later in life and are usually sporadic cases. Initial studies demonstrated amplification of a 10 Mb region at the 2p16–23 locus in PRKAR1A-negative CNC patients. Moreover, somatic alterations of the 2p16 region have been reported in CNC tumors which are usually gene amplifications, whereas, tumor-specific LOH has not been a consistent feature of CNC2 (37). These data suggested that the gene located at 2p16 is a potential oncogene that may code a PKA catalytic subunit.

 

Recently, alterations of PRKACA and PRKACB resulting in a gain of function of the catalytic subunits α and β of PKA respectively, have been associated with components of CNC (38,39). Similarly, inactivating mutations of the phosphodiesterase 11A (PDE11A) gene (located at 2q31.2) and PDE8B, which result in augmented cAMP signaling, have been demonstrated in isolated PPNAD patients (40,41), while CNC patients present a high frequency of PDE11A sequence variants (42).

 

GENOTYPE AND PHENOTYPE CORRELATIONS

 

 Efforts have been made to relate specific phenotypes to corresponding genotypes. A study analyzing 353 patients and 80 different genotypes demonstrated that individuals carrying a PRKAR1A mutation tended to present manifestations earlier and were more likely to have pigmentary disorders, myxomas, and thyroid as well as gonadal tumors. Mutations located in exons were more often associated with acromegaly, myxomas, lentigines, and schwannomas, while intronic mutations had a less serious phenotype  (21). As most of the identified PRKAR1A mutations are nonsense and lead to a lack of detectable mutant protein due to NMD, no genotype-phenotype correlations are expected to be seen. However, specific hot-spot mutations show some genotype-phenotype correlation, such as c.709-7del6, which is associated with the development of PPNAD and c.491-492del, which is frequently associated with cardiac myxomas, lentigines, and thyroid tumors. Regarding those few missense mutations that lead to the expression of a mutant protein, they are related to more severe forms of CNC syndrome, suggesting that NMD may play a protective role against the deleterious effects of mutant products (43).

 

CLINICAL MANIFESTATIONS

 

Carney’s complex is a constellation of clinical manifestations that shows significant variability between patients, even among members of the same family. Some of these features are quite specific, like PPNAD, while others are not, such as thyroid nodules or blue nevi (1). The maximum number of conditions reported to be present together in a single patient is five. Skin disorders are the most common, followed by cardiac myxomas and PPNAD (44,45). These data are summarized in Table 1.

 

Table 1. Clinical Manifestations of CNC

 

A

B1

B2

Adrenal

 

 

 

·       PPNAD

26.0

54.3

57.1

·       Possible PPNAD

 

 

11.4

Cardiac myxoma

53.0

18.6

22.9

Skin

 

37.1

60.0

·       Lentigines

77.0

30.0

55.7

·       Skin myxomas

33.0

14.3

20.0

·       Blue nevi

 

11.4

17.1

Pituitary

 

 

 

·       Certain hypersomatotropism

10.0

8.6

18.6

·       Possible hypersomatotropism

 

-

30.0

Multiple thyroid nodules or carcinoma

5.0

7.1

11.4

Testicular calcifications/LCCST

33.0

20.0

35.0

Breast

 

 

 

·       Benign lesions

3.0

 

42.0

·       Myxomatosis

 

2.0

0

·       Adenoma

 

4.0

0

·       ACR 2-3

 

n/a

0

·       Carcinoma

 

6.0

10.0

Schwannomas

10.0

 

 

·       Confirmed by histology

 

4.3

4.3

·       Suspected

 

0

5.7

Osteochondromyxomas

 

 

 

·       Confirmed by histology

 

2.9

2.9

·       Suspected

 

0

2.9

  1. A) at the time of presentation among 338 patients from an older study (44) and B) from a recent prospective study including 70 patients at the time of presentation (B1), and after three years of follow-up (B2); (45)

 

Most often clinical signs appear in the teen years and early adulthood, with a median age of diagnosis at 20 years of age, while, evidence of the disease, especially cutaneous lesions, can be found even in newborns. During infancy, the most common tumors encountered are cardiac and cutaneous myxomas, as well as PPNAD, while LCCSCT and thyroid nodules appear somewhat later. Acromegaly is clinically evident during the third and fourth decade of life, while cardiac myxomas are equally distributed during the life span (46).

 

The average historic adjusted life expectancy of CNC patients has been reported to be 50-55 years, principally due to individuals who succumb from early cardiovascular sudden death: complications due to cardiac myxoma (myxoma emboli, cardiomyopathy, cardiac arrhythmia, surgical intervention) comprise the major factor of mortality for CNC patients (43). Other less important factors are metastatic or intracranial PMS, thyroid carcinomas, and metastatic pancreatic and testicular tumors (1,44).

 

Cutaneous Pigmentary Disorders

 

These lesions may appear either as multiple lentigines or as blue nevi. They may be present at birth; however, they acquire their typical intensity and distribution around puberty when they increase in number and appear anywhere on the body. Typically, they fade after the fourth decade, although they have been reported in individuals as old as 70 years. Occasionally café au lait spots and depigmented lesions may also be observed (47).

 

LENTIGINES

 

They are the most common cutaneous manifestation of CNC patients (70-75%) and usually present as multiple small (0.2 to 2 mm) brown to black macules that can practically appear on any part of the body with areas of confluence and foci of deeper pigmentation. They are typically located around the orifices of the body, such as on the vermilion border of the lips, on the eyelids, ears, and the genital area (Figure 1). Macroscopically, lentigines are flat, poorly circumcised macules, though in African- Americans, they may be slightly raised, similar to nevi. They may look like solar lentigines; however, they differ as they develop predominantly in areas that have not been exposed to sunlight (e.g., genitalia) and do not change with sun exposure. Histologically, the hyperpigmentation of CNC lesions is associated with melanocytic hyperplasia and hypertrophy, rather than increased melanin production as observed in solar lentigines (47).

 

BLUE NEVI

 

These are larger lesions (up to 8 mm), blue to black, and dome-shaped. They are less common than lentigines but still represent the second most frequent skin manifestation in CNC. They may be multiple with a variable distribution. Histologically, they may present features of epithelioid, junctional, or even compound nevi. Epithelioid blue nevi, currently known as Pigmented Epithelioid Melanocytomas, comprise a class of uncommon melanocytic tumors of intermediate malignancy, which may frequently present lymph nodes metastasis but rarely disseminate to distantorgans (48).

 

Myxomas

 

CUTANEOUS MYXOMAS

 

The skin myxomas present as non-pigmented subcutaneous nodules with a smooth surface and may look white, flesh-colored, opalescent, or pink (Figure 4). They are generally asymptomatic and appear up to the fourth decade. Myxomas can emerge on the face and trunk, while typical sights in CNC are the eyelids (the most common site), external ear canal, and nipples. Less common sites of myxoma formation include the oropharynx (tongue, hard palate, and pharynx) and the female genital tract (uterus, cervix, and vagina). Interestingly, hands and feet are preserved (47). Clinical diagnosis is quite difficult as they are often confused with common “skin tags” and other overgrowths, thus histological confirmation is usually required. Lesions can be localized to the upper dermis and subcutis and consist of polygonal to stellate dermal fibroblasts scattered singly or in non-encapsulated clusters against an abundant basophilic myxoid matrix (2). Although cutaneous myxomas have minimal impact on the clinical course of CNC, their recognition is crucial since they are the most specific manifestation of CNC and may herald the presence of a potentially fatal cardiac myxoma (49)

 

Figure 4. Cutaneous myxoma on the right flank of a CNC patient. With permission from Dermatology Online Journal 2004; 10 (3): 11.

 

CARDIAC MYXOMAS

 

Although these tumors are benign, they are responsible for the majority of deaths (>50%) related to CNC mainly due to cardiovascular complications. In a recent prospective study of 319 CNC patients, 42.6% developed cardiac myxomas and the mean age at diagnosis was in the 3rd decade of life, occasionally presenting as early as at 4 years. The risk of developing cardiac myxomas was elevated among patients already presenting thyroid lesions or breast myxomas. They can develop in any cardiac chamber, with a predilection in the left atrial septum, while they may be multiple and recurrent, therefore, their resection cannot guarantee a permanent cure. Almost half of the patients harboring a cardiac myxoma will experience recurrence, and the risk is increased among women, lasting up to 20 years after the initial detection (49). Of notice, the detection of an apparently sporadic cardiac myxoma should alert the physician, as CNC-associated myxomas represent a significant proportion (7%) of these rare tumors. However, there are significant differences between cardiac myxomas in CNC and their sporadic counterparts regarding their epidemiology, distribution, and biological behavior. Sporadic myxomas emerge most commonly in middle-aged women and are almost exclusively localized at the left atrial aspect of the interatrial septum, at the fossa ovalis. In addition, most of them are cured by surgical resection and do not recur (50).

 

Heart myxomas typically present with a triad of symptoms: a) Symptoms related to myxoma embolization (e.g., stroke, peripheral artery occlusions), b) Heart failure due to reduced cardiac output (complete occlusion of a valvular orifice can lead to sudden death) c) Constitutional symptoms (emaciation, recurrent fevers) probably related to production of cytokines [e.g., interleukin (IL-6)], by the tumor (51). Their size ranges from a few millimeters to 8 cm in diameter and can be partially calcified. They can be depicted sonographically as isoechoic (compared with the heart wall) masses inside the cardiac chambers. They can be studied further with magnetic resonance imaging (MRI), where they appear as hyperintense lesions on T2-weighted images (52). Histologically, the tumors have a gelatinous or hemorrhagic appearance and arise from a population of multipotent subendocardial mesenchymal precursor cells (53)

 

BREAST MYXOMAS (MYXOID FIBROADENOMAS)

 

These lesions are observed in about a fifth of women with Carney complex and are generally considered benign breast tumors. They usually occur in females after puberty and can be multicentric as well as bilateral (Figure 5). Their size ranges from 2mm to 2cm in diameter and may be pink or white with a mucoid appearance. Physical examination of the breast is indicative of diffuse nodularity without dominant masses. Nipple discharge, breast skin abnormalities, or sentinel lymphadenopathy are not features of breast myxomas (54). Histologically, breast myxomas appear as lobulated mesenchymal lesions, characterized by accumulations of large amounts of ground substance in the lobules, as well as in the interlobular stroma. The tumors may or may not be encapsulated (2).

 

Figure 5. Breast multiple myxomas in a patient with Carney complex. Mammogram (A), shows typical dense breasts in a younger woman with no evidence of tumor. However, in the fat-suppressed magnetic resonance image (B) shown on the right, the presence of multiple small myxomas is clearly seen. With permission http://ugen.nichd.nih.gov.

 

When detected in mammography, they appear as well-defined, non-calcified, isodense, or hypodense lesions. Occasionally, they may have an irregular contour, a worrisome finding that warrants fine-needle aspiration (FNA), even in proven CNC patients. However, the imaging modality of choice is MR mammography as it has greater sensitivity compared to ultrasonography or conventional mammography. The number of myxoid lesions depicted with this technique is usually numerous (more than 58 per breast in a case) and shows a homogeneous increase of the signal intensity, a situation characteristic of CNC, also referred to as “breast myxomatosis” (52).

 

OSTEOCHONDROMYXOMAS

 

Osteochondromyxomas or Carney bone tumors are rare myxomatous tumors of the bone that principally affect nasal sinuses and long bones. They have been described in a few cases (1-5%) and exhibit benign behavior; however, they can occasionally cause bone erosion and extend into soft tissues (55). Radiologically they can present as osteolytic lesions with aggressive periosteal new bone formation or as an expansive bone area with mixed sclerotic and lucent regions). These lesions often get the characteristic appearance of “ring sign”, which is evident in plain radiographs, computed tomography, and MRI and are quite specific for CNC (56). Complete resection of the tumor is usually curative. Experiments in rodents demonstrated the osteoblastic origin of the lesion and that the knockdown of PRKAR1A disrupts the differentiation of osteoblasts (56,57).

 

Endocrine Tumors and Overactivity

 

PPNAD (PRIMARY PIGMENTED NODULAR ADRENAL DYSPLASIA) PPNAD

 

This is the endocrine disorder most frequently observed in individuals with CNC. It bilaterally affects the adrenal glands and, according to recent data, can cause clinically overt Cushing’s Syndrome in more than 50% of patients with CNC (45). However, autopsy studies have provided histological evidence that PPNAD is present in almost every CNC individual (2). In 12% of the CNC patients, isolated PPNAD is the only manifestation. A bimodal age distribution is observed: a first peak occurs during infancy, while a second one that includes the majority of cases takes place between the second and third decade of life. The median age at diagnosis is 34 years and it is predominantly observed in females (sex ratio 2.4:1) (44). Histologically, the adrenal cortex is dominated by small pigmented micronodules with an average size of less than 10mm (Figure 6). Although not encapsulated, the nodules are sharply demarcated from the remainder of the cortex and most of them appear to originate deep in the cortex almost at the level of the medulla. A brown-pigmented substance, lipofuscin, is contained in many of the tumor cells and is responsible for the characteristic color of the lesions. Interestingly, tumor cells stain positively for neuroendocrine markers (e.g., Synaptophysin), while normal cortical cells don’t (58). Internodular cortical atrophy is typical, thus the overall weight of the adrenal gland remains more or less normal (2).

 

Figure 6. Macroscopic and CT-scan findings in primary pigmented nodular adrenocortical disease (PPNAD). A: Macroscopic appearance of the adrenal gland where multiple pigmented micronodules are evident at the cut surface. B: Adrenal CT scan revealed a micronodule on the external limb of the left adrenal (see red arrow). Copyright © 2006 Bertherat; licensee BioMed Central Ltd.

 

Radiological and scintigraphic findings are not specific, since the adrenals may appear bilaterally or unilaterally enlarged but in most cases, they appear normal (59). Computed Tomography (CT) is the most appropriate examination for depicting adrenal lesions in PPNAD. Particularly, images obtained with a slice thickness of 3 mm or less, before and after intravenous (IV) injection of contrast are preferable as they might reveal subtle contour irregularities and the presence of hypodense spots that correspond to small, pigmented nodules. The characteristic picture is that of “beads on a string” (52). The type of hypercortisolism observed in this disorder is that of ACTH-independent adrenal hyperfunction. However, demonstrating cortisol overproduction can be challenging because it may develop progressively over the years. Moreover, intermittent, or cyclic forms of hypercortisolism have been reported (60). Clinical manifestations are non-specific and similar to those observed in patients with Cushing syndrome (CS) of other etiologies (central obesity, hypertension, myopathy), with a predisposition to osteoporosis. A 6-day Liddle’s test (low dose dexamethasone for 2 days followed by high dose dexamethasone for 2 days) has been used for the distinction of PPNAD from CS caused by other primary adrenal disorders (61). A paradoxical increase of UFC and/or 17-hydroxysteroids of more than 50% on the second day after high-dose dexamethasone administration is indicative of PPNAD. However, recent reports have argued against the utility of Liddle’s test by demonstrating low sensitivity (39%) and specificity (45). Initial screening with overnight dexamethasone suppression test and urine free cortisol is suggested instead.

 

Notably, reports from independent groups describe the development of adrenocortical cancer (ACC) in the context of CNC (62,63). In both reports, the patients carried PRKAR1A mutations and ACC developed on the background of PPNAD. This observation together with previous reports of benign macronodules (between 1 and 3.5 cm) in adrenal glands affected with PPNAD implies a continuum of tumorigenesis from adrenal hyperplasia to benign nodules, and then cancer, associated with alterations in other tumor suppressor genes apart from PRKAR1A (64).

 

GROWTH HORMONE (GH)-SECRETING PITUITARY ADENOMAS (ACROMEGALY)

 

Clinically evident acromegaly due to a pituitary GH-secreting tumor has been observed in approximately 10-12% of patients with CNC, whereas, gigantism, resulting from excessive GH secretion prior to puberty, is quite rare (44). Data from a recent prospective study, raise this figure to 18%, with a median age at diagnosis 34.5 years (45).

 

The usual underlying pathology is a solitary pituitary adenoma, while cases of multiple adenomas or even diffuse somatomammotrope hyperplasia, a possible precursor of GH-producing adenomas, have been demonstrated in CNC patients (65) as well as in specific Prkr1a knockout mice (66). Pituitary adenomas usually stain positively for both GH and PRL and are occasionally accompanied by mild hyperprolactinemia (67). However, almost a third of CNC patients present asymptomatic disturbances of the somatotroph axis, without meeting the diagnostic criteria of acromegaly, even without pituitary MRI findings (45).

 

THYROID NODULES

 

Seventy-five percent of CNC patients present with thyroid nodules, most of them being benign, non-toxic follicular adenomas. Thyroid nodules usually appear during the first ten years of life in CNC patients. Occasionally, patients (~3%) present with papillary or follicular carcinoma, particularly after a long history of multiple thyroid adenomas (21). In contrast to experimental data and what is observed in CNC patients with adrenal and pituitary tumors, thyroid nodules do not appear to have a predilection for hyperfunction (68).

 

TESTICULAR TUMORS

 

These tumors are of three types: A) Large Cell Calcifying Sertoli Cell Tumors (LCCSCT), B) Leydig cell tumors, and C) adrenocortical rest tumors. So far, the two latter types have been observed only in patients in whom LCCSCT had already been diagnosed.

 

LCCSCTs are observed in 20-50% of affected CNC males at the time of presentation, however, most males will develop such tumors in their adult life (21,45). These tumors are rarely observed in sporadic forms (<1% of testicular tumors), however, they are common in syndromes such as CNC and Peutz-Jeghers, where they are often multicentric and bilateral. They are almost always benign; malignancy has been rarely reported, particularly in tumors exceeding 6 cm in diameter. Nevertheless, their local expansion results in the replacement and compression of the normal testicular tissue (69). Occasionally (25%), LCCSCT may be hormone-producing and demonstrate increased P-450 aromatase expression (15). LCCSCT often presents as rock-hard and non-tender testicular masses and in ultrasonography, they appear as heterogeneous lesions of increased echogenicity with large areas of calcification (70). Macroscopically they are well-demarcated, yellow, and calcified tumors. Clinically, these hormone-producing tumors may cause sexual precocity in young males with low gonadotropin levels, as well as gynecomastia that may result from aromatase overactivity. Typically, fertility is impaired due to obstruction of the seminiferous tubules (16).

 

Leydig cell tumors and adrenocortical rests are both steroid-producing tumors and macroscopically are quite similar, characterized by a brownish hue and relatively soft texture. Leydig cell tumors may show malignant behavior, thus radical resection has been typically recommended. On the contrary, adrenal rests are benign lesions that do not require resection but can lead to recurrent Cushing’s syndrome after adrenalectomy (71). The histological distinction between these two types of tumors can be difficult and a useful feature is the detection of crystalloids of Reinke that are present solely in Leydig cell tumors. However, these crystalloids are not a constant finding. In such case, testicular vein sampling can be helpful, as it may demonstrate cortisol gradient between peripheral and testicular venous blood.

 

OVARIAN LESIONS

 

Eight to 14% of female patients with CNC may present with ovarian lesions, either cystic or solid tumors of the ovarian surface epithelium, such as serous cystadenomas and teratomas. The percentage of ovarian lesions rises up to 60% in autopsy series (72). Ovarian cysts are usually clinically insignificant, whereas, tumors may progress, occasionally, to ovarian carcinoma, particularly in the elderly.

 

Psammomatous Melanotic Schwannomas

 

Psammomatous Melanotic Schwannomas (PMS) are encapsulated tumors of the peripheral nerve sheath and are observed in less than 10% of individuals with CNC, usually in the fourth decade of life (21). Other hereditary syndromes that may present with PMS are neurofibromatosis and isolated familial schwannomatosis. Schwannomas in CNC are heavily pigmented and present frequently with calcifications and multicentricity. Their dark, brownish pigmentation is attributed to elongated spindle-shaped Schwann cells with melanogenic potential. Calcifications are encountered in a laminated form called psammomas and may be accompanied by hemorrhage and necrosis (73). PMS can develop anywhere in the central and peripheral nervous system; however. the most frequent locations are the nerves of the gastrointestinal tract and the paraspinal sympathetic chain (28% of cases). Other sights involved are the chest wall with involvement of the adjacent ribs and the trigeminal ganglion.

 

The initial presentation is usually characterized by local compression; whenever located in the gastrointestinal tract or within soft tissues they may evoke pain and discomfort. If they develop in the spine they may present as radiculopathy. Schwannomas are among the most difficult tumors to treat, especially when they emerge around nerve roots along the spine, a location that makes excision not feasible. In addition, in rare cases (10%), they can be malignant and then often metastasize to the lungs, liver, or brain (74). Unfortunately, there is a paucity of effective treatments for metastatic PMS. Promising results have been recently published for the combination of the check-point inhibitor Nivolumab along with concurrent external beam radiotherapy (75).

 

OTHER MANIFESTATIONS

 

Apart from the major clinical manifestations, there are many other features suggestive of CNC, however, they are not present in a constant manner to set the diagnosis (44). These features are listed in Table 2.

 

Table 2. Findings Suggestive or Possibly Associated with CNC, but Not Diagnostic for the Disease.

1. Intense freckling (without darkly pigmented spots or typical distribution).

2. Blue nevus, usual type (if multiple).

3. Café-au-lait spots or other "birthmarks".

4. Elevated IGF-I levels, abnormal OGTT, or paradoxical GH responses to TRH testing in the absence of clinical acromegaly.

5. Cardiomyopathy.

6. Pilonidal sinus.

7. History of Cushing’s syndrome, acromegaly, or sudden death in extended family.

8. Multiple skin tags and other skin lesions; lipomas.

9. Colonic polyps (usually in association with acromegaly).

10. Hyperprolactinemia (usually mild and almost always in association with clinical or subclinical acromegaly).

11. Single, benign thyroid nodule in a young patient; multiple thyroid nodules in an older patient (detected by ultrasonography).

12. Family history of carcinoma, in particular of the thyroid, colon, pancreas, and ovary; other multiple benign or malignant tumors.

 

Breast ductal adenomas are benign tumors of the mammary gland ducts that may also develop in the context of CNC and can be multiple and bilateral as well. Coexistence with breast myxomas can be observed (54). They are palpable, painless masses that usually appear near the areola and can produce bloody nipple discharge. Radiologically their appearance varies from well-delineated and spherical to completely irregular lesions and they always contain calcifications (52). These calcifications may be coarse (typically benign) or microcalcifications, which are often encountered in adenocarcinomas. Moreover, a possible association between CNC and breast cancer has been demonstrated in the most recent prospective study, affecting up to 13.5% of female patients at an unusually young age (<50 years) (45). Consequently, the differential diagnosis is difficult, and FNA is always recommended.

 

Other tumors reported in around 2,5-4,5% of CNC patients are pancreatic neoplasms including acinar cell carcinoma, adenocarcinoma, and intraductal pancreatic mucinous neoplasia (45,76). In addition, CNC is associated with increased detection of liver lesions, including hepatocellular adenomas, carcinomas, and fibrolamellar carcinomas (77,78).

 

DIAGNOSIS

 

The diagnosis of CNC is principally set by clinical criteria and can be confirmed by molecular testing, which has a mutation detection rate of approximately 60-70%. Genetic testing currently can only be recommended either as an adjunctive test for individuals who meet the clinical criteria or for the detection of affected members of families where the index case harbors a known mutation, in order to avoid unnecessary medical surveillance of non-carriers (1).

 

The following clinical criteria were initially proposed in 1998, were revised in 2001, and have not been modified since then. They yield a sensitivity of nearly 98%. They include 12 clinical manifestations that set the major criteria for diagnosis, as well as 2 supplemental criteria regarding molecular testing and family history. At least two major criteria need to be present to establish the diagnosis of CNC and their occurrence has to be confirmed either biochemically, histologically, or by imaging as indicated. In the presence of one supplemental criterion, a single clinical manifestation is sufficient to establish the diagnosis (44).

 

Major Criteria

 

Skin pigmentation disorders

  1. Spotty skin pigmentation with a typical distribution (vermilion border of the lips, conjunctiva, and inner or outer canthi, vaginal and penile mucosa)
  2. Blue nevus, epithelioid blue nevus (multiple)*

Myxomas

  1. Cutaneous and mucosal myxomas*
  2. Cardiac myxomas*
  3. Breast myxomatosis* or fat-suppressed magnetic resonance imaging findings suggestive of this diagnosis
  4. Osteochondromyxoma*

Endocrine tumors / Overactivity

  1. Primary pigmented nodular adrenal dysplasia (PPNAD)* or a paradoxical positive response of urinary glucocorticosteroids to dexamethasone administration during Liddle’s test
  2. Acromegaly due to GH-producing adenoma or evidence of excess GH production
  3. Large-Cell Calcifying Sertoli Cell Tumor (LCCSCT)* or characteristic calcification on testicular ultrasonography
  4. Thyroid carcinoma* or multiple, hypoechoic nodules on thyroid ultrasonography, in a young patient

Miscellaneous

  1. Psammomatous Melanotic Schwannoma*
  2. Breast ductal adenoma*

* histologically confirmed

 

Supplemental Criteria

  1. Affected first-degree relative
  2. Inactivating mutation of the PRKAR1A gene

 

MANAGEMENT

 

So far, no evidence-based monitoring schedule has been established for CNC; however, clinical work-up for all the manifestations of CNC should be performed at least once a year in all patients, and asymptomatic known mutation carriers, should start in infancy.

 

Surveillance

 

Prepubertal children should be screened as follows:

  • Cardiac ultrasound should start as soon as the diagnosis is made, based either on clinical or genetic grounds, and be performed at least once a year thereafter. In patients with a history of cardiac myxoma, screening should be more frequent, optimally every 6 months, due to the increased risk of recurrence (49).
  • Patients should undergo an initial thyroid ultrasound within the first decade of age, and then repeated according to findings.
  • Screening for the other manifestations should be performed in patients under 5 years of age only by clinical examination. Especially for males, testicular ultrasonography is recommended at the initial evaluation and if microcalcifications are present it should be repeated yearly. Regarding ovarian and breast imaging of female patients, these may be deferred until after puberty (1).
  • Pubertal staging and growth rate should be monitored as pediatric patients with CNC may present with failure to thrive, a possible outcome of various CNC components, such as Cushing’s syndrome due to PPNAD or hepatic involvement (79). On the other hand, the presence of a functional LCCSCT may be associated with growth and maturation acceleration.

 

In post-pubertal adolescents and adults, the following investigations should be performed at initial diagnosis and annually thereafter, including screening for:

  • Cardiac myxomas by echocardiography, which if positive, should be repeated bi-annually. The possibility of first occurrence decreases with age and is exceptional after the age of 50 (49).
  • PPNAD by measurement of urinary free cortisol, ACTH, and overnight suppression with 1 mg Dexamethasone, followed by a formal Low Dose Dexamethasone Test if abnormal. If this suggests cortisol hypersecretion, the diagnosis may be supported by a 6-day Liddle’s test and an adrenal CT scan.
  • Acromegaly by measurement of serum GH, PRL, and Insulin-Like-Growth-Factor I (IGF I). In case of abnormal findings, confirmation of GH hypersecretion with an oral glucose suppression test (OGTT) and imaging of the pituitary region with MRI is suggested.
  • Thyroid nodules by ultrasonography and further evaluation with FNA as needed, according to the relevant guidelines for the general population.
  • LCCSCT in males by testicular ultrasound, especially when small-sized calcifications are found. Follow-up may be less frequent than annual due to the slow progression of these tumors (45)
  • PMS with spine MRI once at baseline and thereafter when clinical signs suggest the presence of this tumor.
  • Breast myxomas as well as ductal adenomas in females should be screened and followed up in the context of screening for breast cancer including self-examination, clinical evaluation, mammography, and ultrasound, starting earlier than in the general population, maybe earlier than the age of 40 (45). In the case of findings, MRI of the breast is more sensitive in mapping the lesions (52).
  • Ovarian lesions by transabdominal ultrasonography during the first evaluation. The test should be repeated due to the low risk of ovarian malignancy (72).

 

Treatment

 

As CNC is generated by a constitutional genetic defect, no etiologic therapy is available yet. The therapeutic approach should target each clinical manifestation and treat accordingly.

  • Cardiac myxomas require surgical removal. However, due to the high recurrence rate, re-operation is usually indicated (49)
  • Cutaneous and mammary myxomas may be surgically removed, mainly for cosmetic and/or diagnostic purposes.
  • Regarding PPNAD, bilateral adrenalectomy has been typically suggested if overt Cushing’s syndrome is evident. Some institutions though have reported treatment with a low dose regimen (0,5-4 g daily) of O,p'-dichlorodiphenyldichloroethane (Mitotane) (80,81) with long term effects; however, the possible significant adverse events of such an approach should be considered.
  • LCCSCT has been traditionally treated with orchiectomy; however, the fact that these tumors often occur bilaterally and are grossly benign has raised an issue to consider treatment options that might preserve fertility. Such an approach is testicular-sparing surgery, followed by strict monitoring of growth and pubertal staging and administration of anti-estrogen drugs in case of recurrence (82). Alternatively, successful treatment of prepubertal gynecomastia and growth acceleration by exclusively using aromatase inhibitors has been reported; however long-term efficacy and safety data are still lacking (83). Similarly, management of Leydig tumors, which often present as small non-palpable testicular lesions, tends to change with the implementation of advanced imaging modalities (magnetic resonance, contrast-enhanced ultrasonography, strain elastography) which may allow a more conservative approach, including surveillance and testis-sparing surgery (84)
  • Pituitary adenomas should be removed by transsphenoidal or transcranial approach, according to their size and extension as in sporadic tumors. Alternatively, long-term medical treatment can be offered.
  • Thyroid nodules should be evaluated and treated surgically according to current guidelines.
  • PMS: surgery to remove primary and/or metastatic lesions.

 

Genetic Counseling

 

Genetic analysis may be suggested for CNC index cases, taking into consideration the fact that the mutation detection rate of PRKR1A testing with standard sequencing is at present approximately 60%. Therefore, a negative test does not exclude CNC in an individual who meets clinical criteria. In such cases, copy number variant (CNV) analysis by comparative genomic hybridization (CGH) and/or PRKAR1A gene deletion testing may be suggested to rule out a PRKAR1A defect. Currently, genetic diagnosis may be assisted by NGS techniques. If all testing for PRKAR1A defects is negative, screening for other candidate genes or loci, including the PRKACA, PRKACB and the phosphodiesterase 8 and 11 genes has been suggested.

 

In those cases where a mutation is detected, genetic screening (specific sequencing) is recommended for first-degree relatives (parents, siblings, and offspring). In case of a positive test, mutation carriers should undergo the same follow-up and management as that suggested for CNC patients. The first cardiac ultrasound should be performed at the same time as the molecular testing.

 

Genetic counseling should include the following general information:

  • If a parent of the index case is affected, the risk to his siblings is 50%. On the contrary, in case of a de novo mutation, this risk falls to approximately 1%.
  • Each child of an individual with CNC has a 50% chance of being affected.
  • Fertility may be impaired in males with CNC. Contrary to male patients, CNC is not specifically associated with female infertility and successful pregnancies and deliveries of female CNC patients have been reported (85).
  • Most tumors of CNC are in general benign except for thyroid nodules and schwannomas; however, they are associated with significant morbidity. Prenatal testing is available by chorionic villous sampling (CVS) at approximately ten to 12 weeks of gestation or amnioparacentesis at 15-18 weeks of gestation. Pre-implantation genetic diagnosis (PGD) is available for PRKAR1A mutation carriers and in conjunction with in-vitro fertilization allows the selection of disease-free embryos for implantation.

 

FUTURE PERSPECTIVES

 

Although remarkable progress has been made since CNC was first described, several issues need to be answered. There are still CNC families that do not carry a PRKAR1A gene mutation and cannot be assigned to CNC2 either. The CNC2 gene located at the 2p16 locus remains to be determined.

 

Researchers put efforts in the search for a more specific therapy for CNC. An older study demonstrated that 8-Cl-adenosine (8-Cl-ADO), a cAMP analog was able to, inhibit in vitro the proliferation induced by G protein-coupled receptors (86); however, no further research has been published on this substance. Moreover, PRKAR1A haploinsufficiency has been shown to induce cyclooxygenase-2 (COX2) activation and prostaglandin E2 (PGE2) overproduction, a disorder that has been associated with the abnormal proliferation of adult bone stromal cells (ABSCs) seen in osteochondromyxomas of CNC patients (87). Experimental administration of celecoxib, a COX2 inhibitor, in mice with PKA defects decreased PGE2 and associated proliferation of ABSCs, resulting in a substantial reduction of bone tumor growth and improved organization of cortical bone that was adjacent to the tumor. Based on the same principle, experiments with celecoxib on adrenocortical cell lines and in a mouse model of PPNAD demonstrated in vitro and in vivo reduction of steroid secretion and cell proliferation (88). Recent advances in genomics and pharmaceutical technologies are promising for timely diagnosis and “etiologic” cure of this syndrome.

 

REFERENCES

 

  1. Bouys L, Bertherat J. MANAGEMENT OF ENDOCRINE DISEASE: Carney complex: clinical and genetic update 20 years after the identification of the CNC1 (PRKAR1A) gene. Eur. J. Endocrinol. 2021;184(3):R99–R109.
  2. Carney JA, Gordon H, Carpenter PC, Shenoy B V, Go VL. The complex of myxomas, spotty pigmentation, and endocrine overactivity. Med. 1985;64(4):270–283.
  3. Carney JA, Hruska LS, Beauchamp GD, Gordon H. Dominant inheritance of the complex of myxomas, spotty pigmentation, and endocrine overactivity. Mayo Clin Proc 1986;61(3):165–172.
  4. Bain J. “Carney’s complex.” Mayo Clin Proc 1986;61(6):508.
  5. Rhodes AR, Silverman RA, Harrist TJ, Perez-Atayde AR. Mucocutaneous lentigines, cardiomucocutaneous myxomas, and multiple blue nevi: the “LAMB” syndrome. J Am Acad Dermatol 1984;10(1):72–82.
  6. Atherton DJ, Pitcher DW, Wells RS, MacDonald DM. A syndrome of various cutaneous pigmented lesions, myxoid neurofibromata and atrial myxoma: the NAME syndrome. Br J Dermatol 1980;103(4):421–429.
  7. Stratakis CA, Carney JA, Lin JP, Papanicolaou DA, Karl M, Kastner DL, Pras E, Chrousos GP. Carney complex, a familial multiple neoplasia and lentiginosis syndrome. Analysis of 11 kindreds and linkage to the short arm of chromosome 2. J Clin Invest 1996;97(3):699–705.
  8. Basson CT, MacRae CA, Korf B, Merliss A. Genetic heterogeneity of familial atrial myxoma syndromes (Carney complex). Am J Cardiol 1997;79(7):994–995.
  9. Casey M, Mah C, Merliss AD, Kirschner LS, Taymans SE, Denio AE, Korf B, Irvine AD, Hughes A, Carney JA, Stratakis CA, Basson CT. Identification of a novel genetic locus for familial cardiac myxomas and Carney complex. Circulation 1998;98(23):2560–2566.
  10. Casey M, Vaughan CJ, He J, Hatcher CJ, Winter JM, Weremowicz S, Montgomery K, Kucherlapati R, Morton CC, Basson CT. Mutations in the protein kinase A R1alpha regulatory subunit cause familial cardiac myxomas and Carney complex. J Clin Invest 2000;106(5):R31-8.
  11. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet 2000;26(1):89–92.
  12. Horvath A, Bertherat J, Groussin L, Guillaud-Bataille M, Tsang K, Cazabat L, Libé R, Remmers E, René-Corail F, Faucz FR, Clauser E, Calender A, Bertagna X, Carney JA, Stratakis CA. Mutations and polymorphisms in the gene encoding regulatory subunit type 1-alpha of protein kinase A (PRKAR1A): an update. Hum. Mutat.2010;31(4):369–79.
  13. Stratakis CA. Carney complex: A familial lentiginosis predisposing to a variety of tumors. Rev. Endocr. Metab. Disord. 2016;17(3):367–371.
  14. Espiard S, Bertherat J. Carney complex. Front Horm Res 2013;41:50–62.
  15. Premkumar A, Stratakis CA, Shawker TH, Papanicolaou DA, Chrousos GP. Testicular ultrasound in Carney complex: report of three cases. J Clin Ultrasound 1997;25(4):211–214.
  16. Burton KA, McDermott DA, Wilkes D, Poulsen MN, Nolan MA, Goldstein M, Basson CT, McKnight GS.Haploinsufficiency at the protein kinase A RI alpha gene locus leads to fertility defects in male mice and men. Mol Endocrinol 2006;20(10):2504–2513.
  17. Rothenbuhler A, Stratakis CA. Clinical and molecular genetics of Carney complex. Best Pr. Res Clin Endocrinol Metab 2011;24(3):389–399.
  18. Scott JD. Cyclic nucleotide-dependent protein kinases. Pharmacol Ther 1991;50(1):123–145.
  19. Almeida M, Stratakis CA. How does cAMP/protein kinase A signaling lead to tumors in the adrenal cortex and other tissues? Mol. Cell. Endocrinol. 2011;336(1–2):162–8.
  20. Cazabat L, Ragazzon B, Groussin L, Bertherat J. PRKAR1A mutations in primary pigmented nodular adrenocortical disease. Pituitary 2006;9(3):211–9.
  21. Bertherat J, Horvath A, Groussin L, Grabar S, Boikos S, Cazabat L, Libe R, Rene-Corail F, Stergiopoulos S, Bourdeau I, Bei T, Clauser E, Calender A, Kirschner LS, Bertagna X, Carney JA, Stratakis CA. Mutations in regulatory subunit type 1A of cyclic adenosine 5’-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients and 80 different genotypes. J Clin Endocrinol Metab 2009;94(6):2085–2091.
  22. Groussin L, Kirschner LS, Vincent-Dejean C, Perlemoine K, Jullian E, Delemer B, Zacharieva S, Pignatelli D, Carney JA, Luton JP, Bertagna X, Stratakis CA, Bertherat J. Molecular analysis of the cyclic AMP-dependent protein kinase A (PKA) regulatory subunit 1A (PRKAR1A) gene in patients with Carney complex and primary pigmented nodular adrenocortical disease (PPNAD) reveals novel mutations and clues for pathophysiology: . Am J Hum Genet 2002;71(6):1433–1442.
  23. Salpea P, Horvath A, London E, Faucz FR, Vetro A, Levy I, Gourgari E, Dauber A, Holm IA, Morrison PJ, Keil MF, Lyssikatos C, Smith ED, Sanidad MA, Kelly JC, Dai Z, Mowrey P, Forlino A, Zuffardi O, Stratakis CA.Deletions of the PRKAR1A locus at 17q24.2-q24.3 in Carney complex: genotype-phenotype correlations and implications for genetic testing. J. Clin. Endocrinol. Metab. 2014;99(1):E183-8.
  24. Stergiopoulos SG, Stratakis CA. Human tumors associated with Carney complex and germline PRKAR1A mutations: a protein kinase A disease! FEBS Lett 2003;546(1):59–64.
  25. Cazabat L, Ragazzon B, Varin A, Potier-Cartereau M, Vandier C, Vezzosi D, Risk-Rabin M, Guellich A, Schittl J, Lechêne P, Richter W, Nikolaev VO, Zhang J, Bertherat J, Vandecasteele G. Inactivation of the Carney complex gene 1 (PRKAR1A) alters spatiotemporal regulation of cAMP and cAMP-dependent protein kinase: a study using genetically encoded FRET-based reporters. Hum. Mol. Genet. 2014;23(5):1163–74.
  26. Nadella KS, Kirschner LS. Disruption of protein kinase a regulation causes immortalization and dysregulation of D-type cyclins. Cancer Res 2005;65(22):10307–10315.
  27. Mavrakis M, Lippincott-Schwartz J, Stratakis CA, Bossis I. Depletion of type IA regulatory subunit (RIalpha) of protein kinase A (PKA) in mammalian cells and tissues activates mTOR and causes autophagic deficiency. Hum Mol Genet 2006;15(19):2962–2971.
  28. Basso F, Rocchetti F, Rodriguez S, Nesterova M, Cormier F, Stratakis CA, Ragazzon B, Bertherat J, Rizk-Rabin M. Comparison of the effects of PRKAR1A and PRKAR2B depletion on signaling pathways, cell growth, and cell cycle control of adrenocortical cells. Horm. Metab. Res. 2014;46(12):883–8.
  29. Knudson AG. Hereditary cancer: two hits revisited. J Cancer Res Clin Oncol 1996;122(3):135–140.
  30. Tsilou ET, Chan C-C, Sandrini F, Rubin BI, Shen DF, Carney JA, Kaiser-Kupfer M, Stratakis CA. Eyelid myxoma in Carney complex without PRKAR1A allelic loss. Am. J. Med. Genet. A 2004;130A(4):395–7.
  31. Gaujoux S, Tissier F, Groussin L, Libé R, Ragazzon B, Launay P, Audebourg A, Dousset B, Bertagna X, Bertherat J. Wnt/beta-catenin and 3’,5’-cyclic adenosine 5’-monophosphate/protein kinase A signaling pathways alterations and somatic beta-catenin gene mutations in the progression of adrenocortical tumors. J. Clin. Endocrinol. Metab. 2008;93(10):4135–40.
  32. Griffin KJ, Kirschner LS, Matyakhina L, Stergiopoulos S, Robinson-White A, Weinberg F, Meoli E, Bornstein SR, Stratakis CA. A mouse model for Carney complex. Endocr Res 2004;30(4):903–911.
  33. Amieux PS, Howe DG, Knickerbocker H, Lee DC, Su T, Laszlo GS, Idzerda RL, Stanley McKnight G. Increased basal cAMP-dependent protein kinase activity inhibits the formation of mesoderm-derived structures in the developing mouse embryo. J. Biol. Chem. 2002;277(30):27294–27304.
  34. Pringle DR, Yin Z, Lee AA, Manchanda PK, Yu L, Parlow AF, Jarjoura D, La Perle KM, Kirschner LS. Thyroid-specific ablation of the Carney complex gene, PRKAR1A, results in hyperthyroidism and follicular thyroid cancer. Endocr Relat Cancer 2012;19(3):435–446.
  35. Kirschner LS. Use of mouse models to understand the molecular basis of tissue-specific tumorigenesis in the Carney complex. J Intern Med 2009;266(1):60–68.
  36. Almeida MQ, Muchow M, Boikos S, Bauer AJ, Griffin KJ, Tsang KM, Cheadle C, Watkins T, Wen F, Starost MF, Bossis I, Nesterova M, Stratakis CA. Mouse Prkar1a haploinsufficiency leads to an increase in tumors in the Trp53+/- or Rb1+/- backgrounds and chemically induced skin papillomas by dysregulation of the cell cycle and Wnt signaling. Hum. Mol. Genet. 2010;19(8):1387–98.
  37. Matyakhina L, Pack S, Kirschner LS, Pak E, Mannan P, Jaikumar J, Taymans SE, Sandrini F, Carney JA, Stratakis CA. Chromosome 2 (2p16) abnormalities in Carney complex tumours. J. Med. Genet. 2003;40(4):268–77.
  38. Berthon AS, Szarek E, Stratakis CA. PRKACA: the catalytic subunit of protein kinase A and adrenocortical tumors. Front. cell Dev. Biol. 2015;3:26.
  39. Forlino A, Vetro A, Garavelli L, Ciccone R, London E, Stratakis CA, Zuffardi O. PRKACB and Carney Complex . N. Engl. J. Med. 2014;370(11):1065–1067.
  40. Horvath A, Giatzakis C, Robinson-White A, Boikos S, Levine E, Griffin K, Stein E, Kamvissi V, Soni P, Bossis I, de Herder W, Carney JA, Bertherat J, Gregersen PK, Remmers EF, Stratakis CA. Adrenal hyperplasia and adenomas are associated with inhibition of phosphodiesterase 11A in carriers of PDE11A sequence variants that are frequent in the population. Cancer Res 2006;66(24):11571–11575.
  41. Horvath A, Giatzakis C, Tsang K, Greene E, Osorio P, Boikos S, Libè R, Patronas Y, Robinson-White A, Remmers E, Bertherat J, Nesterova M, Stratakis CA. A cAMP-specific phosphodiesterase (PDE8B) that is mutated in adrenal hyperplasia is expressed widely in human and mouse tissues: a novel PDE8B isoform in human adrenal cortex. Eur. J. Hum. Genet. 2008;16(10):1245–53.
  42. Libe R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin-Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA. Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab 2010;96(1):E208-14.
  43. Correa R, Salpea P, Stratakis CA. Carney complex: an update. Eur. J. Endocrinol. 2015;173(4):M85–M97.
  44. Stratakis CA, Kirschner LS, Carney JA. Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. J Clin Endocrinol Metab 2001;86(9):4041–4046.
  45. Espiard S, Vantyghem M-C, Assié G, Cardot-Bauters C, Raverot G, Brucker-Davis F, Archambeaud-Mouveroux F, Lefebvre H, Nunes M-L, Tabarin A, Lienhardt A, Chabre O, Houang M, Bottineau M, Stroër S, Groussin L, Guignat L, Cabanes L, Feydy A, Bonnet F, North MO, Dupin N, Grabar S, Duboc D, Bertherat J. Frequency and Incidence of Carney Complex Manifestations: A Prospective Multicenter Study With a Three-Year Follow-Up. J. Clin. Endocrinol. Metab. 2020;105(3):e436–e446.
  46. Wilkes D, McDermott DA, Basson CT. Clinical phenotypes and molecular genetic mechanisms of Carney complex. Lancet Oncol 2005;6(7):501–508.
  47. Mateus C, Palangie A, Franck N, Groussin L, Bertagna X, Avril MF, Bertherat J, Dupin N. Heterogeneity of skin manifestations in patients with Carney complex. J Am Acad Dermatol 2008;59(5):801–810.
  48. Cohen J, Nassau DE, Patel P, Ramasamy R. Low Testosterone in Adolescents & Young Adults. Front. Endocrinol. (Lausanne). 2020;10:916.
  49. Pitsava G, Zhu C, Sundaram R, Mills JL, Stratakis CA. Predicting the risk of cardiac myxoma in Carney complex. Genet. Med. 2021;23(1):80–85.
  50. Reynen K. Cardiac myxomas. N Engl J Med 1995;333(24):1610–1617.
  51. Kamilaris CDC, Faucz FR, Voutetakis A, Stratakis CA. Carney Complex. Exp. Clin. Endocrinol. Diabetes2019;127(2–3):156–164.
  52. Courcoutsakis NA, Tatsi C, Patronas NJ, Lee CC, Prassopoulos PK, Stratakis CA. The complex of myxomas, spotty skin pigmentation and endocrine overactivity (Carney complex): imaging findings with clinical and pathological correlation. Insights Imaging 2013;4(1):119–133.
  53. Lie JT. The identity and histogenesis of cardiac myxomas. A controversy put to rest. Arch Pathol Lab Med1989;113(7):724–726.
  54. Carney JA, Toorkey BC. Ductal adenoma of the breast with tubular features. A probable component of the complex of myxomas, spotty pigmentation, endocrine overactivity, and schwannomas. Am J Surg Pathol1991;15(8):722–731.
  55. Carney JA, Boccon-Gibod L, Jarka DE, Tanaka Y, Swee RG, Unni KK, Stratakis CA. Osteochondromyxoma of bone: a congenital tumor associated with lentigines and other unusual disorders. Am J Surg Pathol2001;25(2):164–176.
  56. Yu W, Zhang Z-Z, Wang O, Huang M-Q, Xia W-B, Guermazi A. Ring sign: an imaging sign for osteochondromyxoma in Carney complex. Quant. Imaging Med. Surg. 2019;9(12):1958–1965.
  57. Pavel E, Nadella K, Towns W. H. 2nd, Kirschner LS. Mutation of Prkar1a causes osteoblast neoplasia driven by dysregulation of protein kinase A. Mol Endocrinol 2008;22(2):430–440.
  58. Stratakis CA, Carney JA, Kirschner LS, Willenberg HS, Brauer S, Ehrhart-Bornstein M, Bornstein SR.Synaptophysin immunoreactivity in primary pigmented nodular adrenocortical disease: neuroendocrine properties of tumors associated with Carney complex. J Clin Endocrinol Metab 1999;84(3):1122–1128.
  59. Vezzosi D, Tenenbaum F, Cazabat L, Tissier F, Bienvenu M, Carrasco CA, Laloi-Michelin M, Barrande G, Lefebvre H, Hiéronimus S, Tabarin A, Bertagna X, Legmann P, Vantyghem M-C, Bertherat J. Hormonal, Radiological, NP-59 Scintigraphy, and Pathological Correlations in Patients With Cushing’s Syndrome Due to Primary Pigmented Nodular Adrenocortical Disease (PPNAD). J. Clin. Endocrinol. Metab. 2015;100(11):4332–8.
  60. Gunther DF, Bourdeau I, Matyakhina L, Cassarino D, Kleiner DE, Griffin K, Courkoutsakis N, Abu-Asab M, Tsokos M, Keil M, Carney JA, Stratakis CA. Cyclical Cushing syndrome presenting in infancy: an early form of primary pigmented nodular adrenocortical disease, or a new entity? J Clin Endocrinol Metab 2004;89(7):3173–3182.
  61. Stratakis CA, Sarlis N, Kirschner LS, Carney JA, Doppman JL, Nieman LK, Chrousos GP, Papanicolaou DA.Paradoxical response to dexamethasone in the diagnosis of primary pigmented nodular adrenocortical disease. Ann Intern Med 1999;131(8):585–591.
  62. Anselmo J, Medeiros S, Carneiro V, Greene E, Levy I, Nesterova M, Lyssikatos C, Horvath A, Carney JA, Stratakis CA. A large family with Carney complex caused by the S147G PRKAR1A mutation shows a unique spectrum of disease including adrenocortical cancer. J Clin Endocrinol Metab 2012;97(2):351–359.
  63. Morin E, Mete O, Wasserman JD, Joshua AM, Asa SL, Ezzat S. Carney complex with adrenal cortical carcinoma. J Clin Endocrinol Metab 2011;97(2):E202-6.
  64. Bertherat J. Adrenocortical cancer in Carney complex: a paradigm of endocrine tumor progression or an association of genetic predisposing factors? J Clin Endocrinol Metab 2012;97(2):387–390.
  65. Lonser RR, Mehta GU, Kindzelski BA, Ray-Chaudhury A, Vortmeyer AO, Dickerman R, Oldfield EH. Surgical Management of Carney Complex-Associated Pituitary Pathology. Neurosurgery 2017;80(5):780–786.
  66. Yin Z, Williams-Simons L, Parlow AF, Asa S, Kirschner LS. Pituitary-specific knockout of the Carney complex gene Prkar1a leads to pituitary tumorigenesis. Mol Endocrinol 2008;22(2):380–387.
  67. Raff SB, Carney JA, Krugman D, Doppman JL, Stratakis CA. Prolactin secretion abnormalities in patients with the “syndrome of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas” (Carney complex). J Pediatr Endocrinol Metab 2000;13(4):373–379.
  68. Stratakis CA, Courcoutsakis NA, Abati A, Filie A, Doppman JL, Carney JA, Shawker T. Thyroid gland abnormalities in patients with the syndrome of spotty skin pigmentation, myxomas, endocrine overactivity, and schwannomas (Carney complex). J Clin Endocrinol Metab 1997;82(7):2037–2043.
  69. Nogales FF, Andujar M, Zuluaga A, Garcia-Puche JL. Malignant Large Cell Calcifying Sertoli Cell Tumor of the Testis. J. Urol. 1995;153(6):1935–1937.
  70. Isidori AM, Lenzi A.Scrotal Ultrasound: Morphological and Functional Atlas. Genova: Accademia Nazionale di Medicina; 2008.
  71. Shawker TH, Doppman JL, Choyke PL, Feuerstein IM, Nieman LK. Intratesticular masses associated with abnormally functioning adrenal glands. J Clin Ultrasound 1992;20(1):51–58.
  72. Stratakis CA, Papageorgiou T, Premkumar A, Pack S, Kirschner LS, Taymans SE, Zhuang Z, Oelkers WH, Carney JA. Ovarian lesions in Carney complex: clinical genetics and possible predisposition to malignancy. J. Clin. Endocrinol. Metab. 2000;85(11):4359–66.
  73. Carney JA. Psammomatous melanotic schwannoma. A distinctive, heritable tumor with special associations, including cardiac myxoma and the Cushing syndrome. Am J Surg Pathol 1990;14(3):206–222.
  74. Shields LBE, Glassman SD, Raque GH, Shields CB. Malignant psammomatous melanotic schwannoma of the spine: A component of Carney complex. Surg. Neurol. Int. 2011;2(1):136.
  75. Bajpai J, Kapoor A, Jalali R, Gounder MM. Checkpoint inhibitors and radiotherapy in refractory malignant melanocytic schwannoma with Carney complex: first evidence of efficacy. BMJ Case Rep. 2021;14(5):e240-296.
  76. Gaujoux S, Tissier F, Ragazzon B, Rebours V, Saloustros E, Perlemoine K, Vincent-Dejean C, Meurette G, Cassagnau E, Dousset B, Bertagna X, Horvath A, Terris B, Carney JA, Stratakis CA, Bertherat J. Pancreatic ductal and acinar cell neoplasms in Carney complex: a possible new association. J. Clin. Endocrinol. Metab.2011;96(11):E1888-95.
  77. Tirosh A, Hamimi A, Faucz F, Aharon-Hananel G, Zavras PD, Bonella B, Auerbach A, Gillis D, Lyssikatos C, Belyavskaya E, Stratakis CA, Gharib AM. Liver findings in patients with Carney complex, germline PRKAR1A pathogenic variants, and link to cardiac myxomas. Endocr. Relat. Cancer 2020;27(6):355–360.
  78. Graham RP, Lackner C, Terracciano L, González-Cantú Y, Maleszewski JJ, Greipp PT, Simon SM, Torbenson MS. Fibrolamellar carcinoma in the Carney complex: PRKAR1A loss instead of the classic DNAJB1-PRKACA fusion. Hepatology 2018;68(4):1441–1447.
  79. Tirosh A, Auerbach A, Bonella B, Zavras PD, Belyavskaya E, Lyssikatos C, Meir K, Weiss R, Daum H, Lodish MB, Gillis D, Stratakis CA. Failure to Thrive in the Context of Carney Complex. Horm. Res. Paediatr.2018;89(1):38–46.
  80. Campo MR, Lamacchia O, Farese A, Conserva A, Picca G, Grilli G, Cignarelli M. Mitotane and Carney Complex: ten years follow-up of a low-dose mitotane regimen inducing a sustained correction of hypercortisolism. Hormones (Athens). 2015;14(2):300–4.
  81. Cignarelli M, Picca G, Campo M, Margaglione M, Marino A, Logoluso F, Giorgino F. A six month mitotane course induced sustained correction of hypercortisolism in a young woman with PPNAD and Carney complex. J Endocrinol Invest 2005;28(1):54–60.
  82. Brown B, Ram A, Clayton P, Humphrey G. Conservative management of bilateral Sertoli cell tumors of the testicle in association with the Carney complex: a case report. J Pediatr Surg 2007;42(9):E13-5.
  83. Crocker MK, Gourgari E, Lodish M, Stratakis CA. Use of aromatase inhibitors in large cell calcifying sertoli cell tumors: effects on gynecomastia, growth velocity, and bone age. J. Clin. Endocrinol. Metab. 2014;99(12):E2673-80.
  84. Isidori AM, Pozza C, Gianfrilli D, Giannetta E, Lemma A, Pofi R, Barbagallo F, Manganaro L, Martino G, Lombardo F, Cantisani V, Franco G, Lenzi A. Differential diagnosis of nonpalpable testicular lesions: qualitative and quantitative contrast-enhanced US of benign and malignant testicular tumors. Radiology 2014;273(2):606–18.
  85. Ralser DJ, Strizek B, Kupczyk P, Stoffel-Wagner B, Altengarten J, Müller A, Woelfle J, Gembruch U, Klingmueller D, Merz WM, Paschkowiak-Christes A. Obstetric and Neonatal Outcome of Pregnancy in Carney Complex: A Case Report. Front. Endocrinol. (Lausanne). 2020;11:296.
  86. Robinson-White AJ, Bossis I, Hsiao HP, Nesterova M, Leitner WW, Stratakis CA. 8-Cl-adenosine inhibits proliferation and causes apoptosis in B-lymphocytes via protein kinase A-dependent and independent effects: implications for treatment of Carney complex-associated tumors. J Clin Endocrinol Metab 2009;94(10):4061–4069.
  87. Saloustros E, Liu S, Mertz EL, Bhattacharyya N, Starost MF, Salpea P, Nesterova M, Collins M, Leikin S, Stratakis CA. Celecoxib treatment of fibrous dysplasia (FD) in a human FD cell line and FD-like lesions in mice with protein kinase A (PKA) defects. Mol. Cell. Endocrinol. 2017;439:165–174.
  88. Liu S, Saloustros E, Berthon A, Starost MF, Sahut-Barnola I, Salpea P, Szarek E, Faucz FR, Martinez A, Stratakis CA. Celecoxib reduces glucocorticoids in vitro and in a mouse model with adrenocortical hyperplasia. Endocr. Relat. Cancer 2016;23(1):15–25.

Monitoring Technologies – Continuous Glucose Monitoring, Mobile Technology, Biomarkers of Glycemic Control

ABSTRACT

 

It is recognized that traditional measures of glucose control (such as hemoglobin A1c [A1C]) provide little information regarding the need for day-to-day changes in therapies. While intermittent self-monitored blood glucose (SMBG) provides additional information with which to make treatment decisions, significant barriers to its use exist, such asinconvenience and lack of timely and regular feedback. Furthermore, important information regarding glucose trends may be missed. Continuous glucose monitoring (CGM) has become increasingly reliable and has demonstrated efficacy in terms of improving A1C, reducing hypoglycemia, and improving the time in target glucose range. Incremental progress continues to be made toward a fully functional artificial pancreas, of which CGM will play a vital role. As more and more data are presented to patients and providers, it has become increasingly paramount that the data are organized in a standardized way and that communication of data is streamlined using patients’ mobiledevices where available and within the existing clinic infrastructure. Systems that provide immediate feedback to patients and decision support tools for patients and providers have demonstrated superior outcomes compared to routine SMBG alone. Alternate markers of glucose control may provide complementary information about glucose control and long-term prognosis. This chapter will review the latest evidence for use of professional and personal CGM, mobile glucose monitoring approaches, and biomarkers of glycemic control.

 

INTRODUCTION

 

The current technology for monitoring of glucose levels has been well established since the 1980′s. This practice is beneficial to patients with diabetes from both a clinical and an economic standpoint when used optimally. Knowledgeof the glucose levels that are measured can allow a patient to select an appropriate dose of insulin or implement dietary or other lifestyle changes to regulate their glucose levels. Expert groups provide recommendations for glucose targets, including A1C, self-monitored blood glucose (SMBG), and interstitial glucose (1,2). Although targets vary, expert groups recommend individualization based upon risk of hypoglycemia, polypharmacy, comorbidities, and other characteristics that may affect long-term benefit and individual patient characteristics. The ADA has expanded recommendations for assessing overall glucose levels to include the A1C or CGM metrics such as % Time in Range(TIR, the % of time spent 70-180 mg/dl), or the Glucose Management Indicator (GMI), which is an estimate of A1C that is derived from a 14-day CGM report for routine assessment of glucose levels (1).

 

The landscape of glucose monitoring technologies is expanding and rapidly changing. For a full review of glucose monitoring technologies, the reader is referred to one of many excellent reviews referenced throughout this chapter. Several trends are emerging in glucose monitoring and will be reviewed in more detail in this chapter:

 

  • CGM: This practice is becoming more widely established as evidence supporting its use has accumulated. The data available through CGM can permit significantly more fine-tuned adjustments in insulin dosing and other therapies than spot testing from self-monitoring of blood glucose (SMBG) can CGM technologies forautomatic collection of data have spurred interest in noninvasive glucose monitoring as an additional tool for obtaining information about glucose levels.
  • Closed loop control (CLC): Also known as an “artificial” or “bionic” pancreas, this technology links CGM withautomatically controlled insulin The first steps toward CLC are now in use.
  • Mobile Technology and Decision Support: In recent years, increasing connectivity between glucose monitoringtechnologies and mobile devices has facilitated ongoing improvements in self-care and communication of data.
  • Alternate Markers of Glucose Control: Finally, the use of additional analytes besides glucose is still being established.

 

This chapter analyzes the technology, benefits, and problems with the use of intermittent SMBG and CGM, mobile technology and decision support, and alternate biomarkers of glycemic control.

 

CONTINUOUS GLUCOSE MONITORS

 

CGM measures glucose levels (typically interstitial glucose) continuously and updates the glucose level display every 5 minutes. Most CGMs consist of 1) a monitor to display the information (in some cases, this is the patient’s mobile device), 2) a sensor that is usually inserted into the subcutaneous tissue, and 3) a transmitter that transmits the sensor data to the monitor. Previously, all devices were approved for adjunctive use only due to limitations in accuracy; in this case patients must still perform fingerstick glucose monitoring in order to guide therapy and perform calibrations. However, in 2016, the FDA approved the use of the Dexcom G5 as the first CGM for stand-alone use.Newer technologies have eliminated the requirements for calibration of CGM with a fingerstick glucose. The accuracy of all commercially available CGMs is still the lowest in the hypoglycemic range, which is where the need for sensitivity and specificity is great in terms of serving as an alarm for hypoglycemia.

 

CGM can provide both retrospective as well as real-time information to detect: 1) hypoglycemic and hyperglycemic excursions; 2) predict impending hypoglycemia; and 3) wide fluctuations in glucose levels, also known as glycemicvariability. 24-hour telephone support is available for all FDA approved CGM devices. Use of CGM can help both thepatient and their medical provider make fine tune adjustments to medication therapy and provide insight to the patient on behavioral changes to achieve glycemic control. Additionally, current efforts to link CGM measurement with automatically controlled insulin delivery, has progressed incrementally toward a fully functional artificial pancreas.Systems can be divided according to their intended use as professional CGM (which is a clinic-owned device and provides either retrospective or real-time glucose data) and personal CGM (which is patient-owned and provides real-time glucose data).

 

PROFESSIONAL CGM

 

Professional CGM describes CGM data that are obtained via healthcare provider owned equipment. It does not necessarily provide the glucose results in real time, but downloads the readings after they have been collected, similar to a 24-hour cardiac Holter monitor that provides information about cardiac rhythms after they have occurred. This allows the health care provider to obtain relatively unbiased glucose patterns during typical everyday life. The Endocrine Society recommendations state that professional CGM may be of benefit in adults with diabetes to detectnocturnal hypoglycemia, dawn phenomenon, postprandial hyperglycemia and to assist in management of diabetes therapies (3). Professional CGM is more readily reimbursed than personal CGM, but interpretation of both personal and professional CGM reports by qualified healthcare professionals may be reimbursed on a monthly basis.

 

Some personal CGM systems can be operated in a blinded fashion in order to provide professional glucose data. These systems will be discussed in more detail later (see “Personal [Real-time] Continuous Glucose Monitoring”). The first device for reading blood glucose levels continuously was a professional CGM that was approved by the FDA inJune 1999. This device was the Continuous Glucose Monitor System (CGMS) manufactured by Medtronic MiniMed (Medtronic Diabetes, Northridge, CA) (4). Since then, newer models have shown improvements in accuracy and patient acceptance. In a meta-analysis of 22 articles, professional CGM resulted in a greater reduction in A1c(-0.28%, 95% CI -0.36% to -0.21%, P < 0.00001) as well as TIR (5.59%, 95% CI 0.12 to 11.06, P = 0.05) compared to usual practice (5).

 

FreeStyle Libre Pro

 

The FreeStyle Libre Pro utilizes the same sensor as the Libre personal CGM. The Libre is factory calibrated andtherefore does not require self-monitored blood glucose calibrations. This may be a potential advantage since capillary blood glucose testing is subject to various system and user errors, which in addition to the physiologic lag time between blood and interstitial glucose (which is magnified in the postprandial period) could contribute to CGM error. Itcollects up to 14 days of glucose readings, which are recorded every 15 minutes. The glucose sensor is fully disposable and a single reader is used to activate and scan multiple devices, allowing multiple patients in one office to undergo the procedure simultaneously. Reports are obtained through the LibreView website, which offers a secure cloud-based system, or the FreeStyle Libre desktop reporting software. Reports provide daily patterns, an assessmentof glucose variability and hypoglycemia risk, a daily glucose report, and an overall snapshot report.

 

The overall MARD (Mean Absolute Relative Difference which is calculated by averaging the absolute values of relative differences between CGM measurement results and corresponding comparison method results) for the FreeStyle Libre is 11.4%, 86.7% of readings were in Zone A of the Consensus Error Grid analysis, and 99.7% of results were in Zones A and B (6). It is important to note that sensor accuracy is lower on day 1 and in thehypoglycemia range (MARD 20.3% for values <72 mg/dl in one study) (7). Accuracy improves and remains steady over the 14-day wear period. The Libre utilizes glucose oxidase in a “direct signaling” approach that is not dependenton oxygen and minimizes interference by other substances, such as acetaminophen, which may falsely elevated readings on other devices.

 

Dexcom Professional

 

The Dexcom G6 Pro was approved by the FDA in March 2018 and is available in blinded or unblinded mode depending upon whether the goal is to observe glucose patterns without intervention, to provide immediate feedback to educate and inform patients about their medications and behaviors, or to facilitate decisions about pursuingpersonal CGM. The sensor, transmitter, and receiver are essentially identical to the personal Dexcom G6 system and features expedited startup time and no calibration. The device measures interstitial glucose levels every 5 minutes and is approved for 10 days of use. The device is downloaded using Dexcom CLARITY, a web-based software program that is also used to download and review personal data.

 

Analysis of Retrospective Data

 

Data from all CGM devices can be studied retrospectively after downloading (8). It is recommended that diet, activity, symptom, and insulin data are collected during professional CGM to assist with interpretation, either via patient diary,direct entry of events into the device, or use of an accompanying app, depending on the system. Three time periods should be analyzed. These are:

 

  • Overnight: Out-of-target overnight glucose levels can be modified by adjusting the basal insulin dose.
  • Pre-prandial Period: Out-of-target pre-prandial glucose levels can be modified by adjusting the previous meal bolus, meal, or exercise pattern.
  • Post-prandial period: Out-of-target postprandial glucose levels can be modified by adjusting the immediate meal bolus, meal, or exercise pattern.

 

In certain special situations, targets may need to be adjusted. Other important elements of a professional CGM analysis are shown in Table 1. An example of a patient who used CGM is presented in Figure 1. The CGMdemonstrated high glucose levels from 6:00 PM to 11:00 PM post-supper and low glucose levels from 12:00 AM to 2AM. Recognition of these patterns allowed appropriately timed treatment interventions.

 

Table 1. Elements of Professional Continuous Glucose Monitoring Analysis

Overall Control

Mean Glucose

Glucose Variability (Standard Deviation, Coefficient of Variation)

Daily Detail

Diurnal Patterns: dawn phenomenon, overnight Meal effects

Correction Exercise effects

Other patterns (work days vs. weekend, menstrual cycles)

Hypoglycemia

Precipitating factors

Corresponding meter glucose (recognition)

 

 

Ambulatory Glucose Profile

 

The ambulatory glucose profile (AGP, Figure 2) is a standardized reporting format for glucose data that was developed by an expert panel of diabetes specialists and sponsored by the Helmsley Charitable Trust and is customized for insulin pumps or injection therapy (9). The universal report is intended to simplify and facilitate interpretation of otherwise complex and lengthy reports with varying terminology. It is anticipated that a standardized report would “help clinicians develop expertise in CGM use, enhance quality of care through enhanced pattern recognition, improve practice efficiencies with minimal disruption of workflow, and engage patients, thereby reinforcing consistent use of CGM technology.” A single page report that the medical team can view and file into a patient’s electronic medical record and that can be used as a shared decision-making tool with people with diabetes wasconsidered to be of great value in the report of the 12th International Conference on Advanced Technologies & Treatments for Diabetes (ATTD 2019) (10). The AGP is currently employed by many reporting systems and consists of 3 components:

 

  • Statistical Summary, which utilizes standard metrics and terminology to summarize the number of values,percentage of values, and time in target, above target, and below target, as well as an assessment of glucose variability.
  • Modal day report which collapses data from days or weeks to a single day in order to identify patterns by time of day. Data are presented graphically as 5 distribution curves, representing the median, interquartile range, and10th to 90th percentiles, on the backdrop of target range.
  • Daily View, which facilitates review of within day

 

Composite Metrics

 

As a measure of the quality of glycemia, the time in range (TIR), similar to the A1C is limited in its assessment of hypoglycemia. Multiple composite metrics have thus been reported (11). However, the use of multiple metricsincreases complexity and is subject to issues with collinearity. The Glycemia Risk Index (GRI) is a composite metricthat was developed using input from 330 clinical experts who analyzed 14-day tracings from 225 adults with diabetes (12). GRI more heavily weights very high or very low glucose values and correlates with clinician rankings more closely than TIR or time below range (%time < 70 mg/dl, TBR) alone.

 

Figure 2. Ambulatory Glucose Profile for Insulin Pumps.
Glucose Statistics: Metrics include mean glucose, estimated A1C, glucose ranges, coefficient of variation and standard deviation.
Glucose Profile: Daily glucose profiles are combined to make a one-day (24-hour) picture. Ideally, lines would stay within grey shaded area (target range).
Orange: median (middle) glucose line.
Blue: area between blue lines shows 50% of the glucose values.
Green: 10% of values are above (90% top line) and 10% are below (10% bottom line). Insulin Profile Graph: Shows basal insulin pump settings over a 24-hour period.
Bolus Insulin Graph: Combines all bolus insulin doses into one graph to make a one-day (24-hour) picture. Each box on the graph covers 60 minutes of doses.
Orange: median (middle) dot.
Blue: shaded box shows 50% of the bolus dosages in the hour.
Green: lines above and below the shaded box (whiskers) show how many of the bolus dosages per hour were between 75 - 90% and between 10 - 25%.

 

PERSONAL REAL-TIME CGM (RT-CGM) OR INTERMITTENTLY SCANNED CGM (IS-CGM)

 

RT-CGM devices not only display the current glucose every few minutes, but may also alert the patient for impending (projected alert) or actual (threshold alert) hyperglycemia or hypoglycemia or rate of change in glucose. Bycomparison, is-CGM requires patient interaction with the device to obtain readings but may still provide alerts for hypoglycemia or hyperglycemia. While few head to head studies are available, some studies suggest greater reduction in hypoglycemia and improvement in TIR with RT-CGM compared to is-CGM in persons with type 1 diabetes (13,14), even up to 24 months (15).

 

Over time, accuracy with RT-CGM and is-CGM has improved substantially (16,17,18). In fact, some devices, including the Dexcom and Freestyle Libre are approved for stand-alone use, meaning that under specified conditions, the device may be used to make treatment decisions without confirmatory blood glucose measure. However, the user will still experience a tradeoff between a high alarm sensitivity and specificity for detecting hypoglycemic events, particularly where glucose levels are changing rapidly (Figure 3). Current and recent glucose levels, trend information,and a visual alarm are all presented so that a patient can predict future low or high glucose excursions. Using this information will allow the patient to take actions to spend more time in the euglycemic range and less time in the hypoglycemic or hyperglycemic ranges. This potential decrease in glycemic variability will not necessarily be reflected in an improved A1C value, which reflects mean glycemic levels.

 

Figure 3. Tradeoffs between emphasis on high sensitivity compared to emphasis on high specificity in a hypoglycemic alarm that is part of a continuous glucose monitor.

 

Evidence- Type 1 Diabetes

 

Studies may be divided according to background therapies (insulin pump or injection therapy).

 

STUDIES UTILIZING EITHER INSULIN PUMP OR INJECTIONS AS BACKGROUND THERAPIES

 

  • The seven-country GuardControl Study was the first randomized controlled trial to ever demonstrate a statisticallysignificant improvement in A1C levels with the use of RT-CGM (19). The Guardian RT was used either continuously or biweekly for three months and both regimens were compared to control treatment which did not include use of CGM. At one month and at three months the continuous users had significantly lower A1C levelsthan the controls. The biweekly users had intermediate improvement which did not reach statistical significance compared to the outcomes in the control group.
  • In 2008, the Juvenile Diabetes Research Foundation Continuous Glucose Monitoring Study Group evaluated 322 adults and children with type 1 diabetes (either injection or insulin pump therapy) and A1C 7-10% who wererandomized to either RT-CGM or usual care (20). RT-CGM was associated with a 53% reduction in A1Ccompared to usual care (p<0.001), but was only significant among subjects over age 24 due to lack of consistent use in younger patients. Hypoglycemia was infrequent and was not different between groups.
  • In 2011, 120 children and adults with type 1 diabetes on insulin pump or injection therapy and A1C <7.5% wererandomly assigned to RT-CGM (Freestyle Navigator—not available in the US) or masked CGM every other week (21). The time spent in hypoglycemia was reduced over 50% at 26 weeks, and patients spent more time in 70-180 mg/dl range.
  • In the IMPACT trial, 241 adults with type 1 diabetes with an A1C less than or equal to 7.5% were randomly assigned to Freestyle flash glucose monitoring (described in more detail under “Overview of Stand-Alone Personal CGM systems”) vs. SMBG. In this group 68% of the patients were treated with multiple daily injections and 32% with CSII. The amount of time spent in hypoglycemia was decreased by nearly 90 minutes per day (P<0.0001) when patients had access to CGM data (22). It must be noted that this technology does not provide real-time alerts for impending hypoglycemia or hyperglycemia and data are accessed via a hand-held device on Ina small study of patients with hypoglycemia unawareness or recent severe hypoglycemia, RT-CGM more effectively reduced the time spent in hypoglycemia compared to flash glucose monitoring (23).
  • The CITY study was a randomized study among 153 adolescents and young adults with type 1 diabetes. CGM resulted in a -0.37% greater reduction in A1C compared to usual care (p=0.01) (24). In this study, only 68% of participants used CGM at least 5 days per week in month 6, which is significantly lower than studies reported inadults (25). However, this is more than twice that reported in the pivotal JDRF study of 2008 (20). Moreover, this study utilized an earlier generation CGM which required twice daily calibration; thus, it is possible that newer technologies may support greater persistence with use.
  • Among 203 older adults (median age 68) with type 1 diabetes randomized to CGM or usual care, CGM resulted in less hypoglycemia at 26 weeks (estimated treatment difference 27 minutes/day, p<0.001) as well as modest improvement in A1C (estimated treatment difference -0.3%, p<0.001) (26). The improvement in hypoglycemia was sustained over 52 weeks, at which point CGM use was still >90% (27).

 

STUDIES UTILIZING INSULIN PUMP THERAPY AS BACKGROUND

 

  • In the largest study to date, the STAR3 study, 485 adults and children with A1C 4-9.5% were randomized to sensor-augmented pump therapy (Medtronic Paradigm Revel) or multiple daily injections per day (28). Sensor-augmented pump therapy resulted in better A1C reduction with between-group difference of 0.6%, p<0.001.Hypoglycemia did not differ between groups, but only short-term CGM data were available for comparison and patients with a history of severe hypoglycemia were excluded.

 

STUDIES UTILIZING INJECTION THERAPY AS BACKGROUND

 

  • In 2016, a 6-month randomized controlled trial, the DIAMOND study, compared RT-CGM (using Dexcom G4system) versus SMBG in 158 patients with type 1 diabetes on multi-dose injection therapy and demonstrated a significantly lower A1C (between group difference 0.6%, p<0.0001), decrease in hypoglycemia (43 minutes vs. 80 minutes per day, p=0.0002) and less glucose variability with RT-CGM compared to SMBG. This study did not address hypoglycemia frequency in the two groups (25).
  • The GOLD trial studied 161 patients with type 1 diabetes receiving multiple daily injections with either RT-CGM (Dexcom G4) or standard care in a random order cross-over trial. The mean difference in A1C was 0.43% (p<0.001), favoring RT-CGM. One subject in the CGM group compared to 5 subjects in the standard care group experienced a severe hypoglycemic event. The percentage of time spent in hypoglycemia numerically favored the CGM group but statistical analyses were not presented. There was a significant reduction in standard deviationand MAGE (measures of glucose variability). Overall well-being, diabetes treatment satisfaction, and fear of hypoglycemia improved (29).
  • In the FLASH-UK study, 156 participants with type 1 diabetes were randomized to intermittently scanned glucosemonitoring or usual care (30). The intervention group had a significantly greater reduction in HbA1 (adjusted treatment difference -0.5%, p<0.001), higher % TIR, and lower % TBR.
  • A randomized controlled trial among 104 adults with type 1 diabetes found that intermittently scanned glucose monitoring improved A1c (estimated treatment difference 0.3% [95% CI, 0.0%-0.6%; P = 0.04) and TIR but not TBR compared to blood glucose monitoring (31).

 

META-ANALYSES

 

A Cochrane review and another meta-analysis found modest A1c reductions, particularly among patients who were not using insulin pumps, patients under age 18, and among patients with lower adherence (32). The results were heavily influenced by the STAR3 trial, and the JDRF study did not report a difference between pump users and patients using multiple dose injection therapy. Severe hypoglycemia rates did not differ. However, the quality of most studies was limited due to small sample size, lack of blinding, and lack of sufficient data to compare hypoglycemia rates. Meta-analyses may be hampered by the inclusion of studies with obsolete technology or lack of consideration for the intended use of the device in the study (33,34). In another meta-analysis, studies that specifically enrolled patients at risk for hypoglycemia and used blinded CGM to assess it did show improvement in hypoglycemia (35).

 

More recently, a meta-analysis of 21 studies published between 2011-2020 encompassing 2149 individuals with type 1 diabetes revealed that CGM led to a significant reduction in A1C by 0.23% (p=0.0005), with larger treatment effect at higher baseline A1C (>8%), and no effect on severe hypoglycemia or DKA (36). However, the meta-analysis did not report CGM derived metrics such as TIR or TBR or clinically significant hypoglycemia. In a 2023 meta-analysis of 22 randomized controlled trials that included participants with type 1 diabetes, there was an overall improvement in A1c,TIR and TBR (37). Reduction in A1C was limited to nonadjunctive devices but all devices resulted in improvement in TIR.

 

PATIENTS WITH HYPOGLYCEMIC UNAWARENESS

 

Many older studies specifically excluded patients with a history of severe hypoglycemia or were underpowered todetect significant hypoglycemia. Recent studies have examined the use of RT- CGM in patients with hypoglycemiaunawareness, which is a risk factor for severe hypoglycemia (events requiring outside assistance to treat).

 

  • In the HypoCOMPaSS trial, 96 patients with a history of hypoglycemia unawareness determined by the GOLD Score of at least 4 or more were randomly assigned in a 2x2 factorial design to insulin pump or injection therapy, both with access to a bolus insulin calculator, and either RT-CGM (Medtronic Continuous Glucose Monitoring System) or SMBG. All patients had diabetes education with a goal toward hypoglycemia avoidance (38). Theresults demonstrated a similar reduction in severe hypoglycemia and improvement in hypoglycemia unawareness and fear of hypoglycemia without a significant treatment interaction between insulin or glucose monitoring interventions. Treatment satisfaction was higher with insulin pump compared to injection therapy but similar between RT-CGM and
  • The IN CONTROL trial evaluated patients with Type 1 diabetes and hypoglycemia unawareness receiving either injection or insulin pump therapy in a crossover study comparing RT-CTM (Medtronic Paradigm Veo system with a MiniLink transmitter and an Enlite glucose sensor) or SMBG (39). Hypoglycemia was significantly reduced with RT-CGM compared to SMBG (including a 9.8% reduction in events <70 mg/dl and 44% reduction in events <40 mg/dl). Severe hypoglycemic events were significantly reduced but hypoglycemia unawareness was unchanged.
  • In a smaller study of 52 adults with type 1 diabetes and problematic hypoglycemia, immediate randomization to CGM was more effective for preventing severe hypoglycemia (39% fewer events, p<0.05) than a dedicated hypoglycemia avoidance education program alone (40). CGM also lead to greater reduction in A1c (treatment difference -0.47%, p<0.05), but impaired awareness was restored in 31% of both groups, supporting the conceptthat CGM assists in earlier recognition and treatment of impending hypoglycemia as opposed to effecting fundamental change in counterregulatory responses.

 

Differences between studies may be explained by differences in populations and the technologies utilized. In the InCONTROL study, contact with patients was less frequent, sensor use was greater (89 vs. 57% in HypoCOMPaSS) and there were no insulin adjustment protocols. Therefore, more studies are needed to understand the potential role of background therapy, other technologies, and clinical support.

 

PATIENT REPORTED OUTCOMES

 

Generic Quality of life scores generally do not improve with RT-CGM but treatment-specific measures, such asdiabetes distress, hypoglycemic confidence, fear of hypoglycemia and to a lesser extent, measures of convenience, efficacy and performance, may be improved (28,41,42).

 

Evidence- Type 2 Diabetes

 

In patients with type 2 diabetes, even in patients not on insulin, RT-CGM may act as a motivator and positive influence for patients to improve lifestyle. The change in behavior can potentially lead to better glycemic control and weight loss(43). Moreover, periodic (every 3 months) short- term (14 day) use of real-time CGM may be sufficient to achieve andmaintain clinically relevant improvements in A1c in this population (44).

 

  • In 2012, Vigersky et al. randomized 100 patients with type 2 diabetes on basal insulin and anti-hyperglycemic agents into either a group that used real-time RT-CGM intermittently (2 weeks on, 1 week off) or a group that recorded SMBG four times per day for 12 weeks. At 12 weeks, they found a statistically significantly greater reduction in A1c by 1.0% in the CGM group compared to 5% reduction in the SMBG group. The effect persistedup to the 40-week follow-up, 0.8% and 0.5% reduction in A1c in the RT-CGM versus SMBG group respectively (45).
  • In 2017, Beck et al conducted a randomized study to evaluate benefit of RT-CGM use in 158 patients with type 2 diabetes with mean A1C of 8.5% treated using multiple daily injections (46). Over a 24-week period the A1C decreased to 7.7% in the RT-CGM group compared to 8% in the group with usual care (mean difference -0.3%,p=0.022). RT-CGM derived hypoglycemia and quality of life did not differ.
  • The Dexcom MOBILE study assessed patients with type 2 diabetes on basal insulin randomly assigned to the Dexcom G6 or usual care for 8 months and reported a significant reduction in A1C, improved TIR andhypoglycemia (47). This was accomplished without an appreciable change in insulin or other medication use, indicating that CGM improves glucose levels by facilitating behavioral changes. Moreover, subsequent discontinuation of CGM for 6 months resulted in loss of about half of the improvement in TIR (48). Moreover, the benefit was similar in older (≥65 years old) vs. younger adults (49).
  • In a 10-week study of 101 patients with type 2 diabetes on multiple daily injections of insulin, patients randomized flash glucose monitoring (Freestyle Libre) had greater A1C reduction (-0.82 vs -0.33%, p=0.005), found their treatment to be significantly more flexible and were more likely to recommend it to others (50).
  • Among 141 adults with type 2 diabetes treated with insulin or sulfonylurea and recent myocardial infarction, thoserandomized to intermittently scanned glucose monitoring had significantly less TBR (-80 minutes, 95% CI -118, -43 minutes) at 90 days, but marginal difference in A1c or TIR, and the intervention was reported to be cost-effective (51).
  • In a randomized trial of 116 adults with type 2 diabetes using non-insulin therapies, intermittently scanned glucose monitoring in combination with diabetes self-management education demonstrated superior A1c reduction at 16 weeks (treatment difference 0.3%, 95% CI 0 to 7%, p=0.048), larger increase in TIR (9.9%, p<0.01), and greatersatisfaction compared to education alone (52).

 

Real World Outcomes

 

  • In a study of over 29,000 pediatric patients with type 1 diabetes in the Type 1 diabetes Exchange Registry or the German/Austrian DPV Initiative, pediatric CGM use was associated with lower mean A1C regardless of insulindelivery modality (pump or injection) (53).
  • In a study of 106 UK hospitals incorporating 16,427 participants, 1241 with repeated TIR data, improvements inTIR were associated with improvement in hypoglycemia unawareness and diabetes related Moreover, TIR>70% was associated with reduced resource utilization (hospital admissions for hypoglycemia or hyperglycemia, paramedic visits, and severe hypoglycemia (54).
  • In the Swedish National Diabetes Registry that included 14,372 adults with type 1 diabetes, intermittently scanned glucose monitoring was associated with a small (0.11%, p<0.0001) reduction in A1C after 15-24 months and reduction in severe hypoglycemic episodes (OR 0.79, 95% CI 0.69-0.91) (55).
  • Using the French national claims database, a total of 74,011 patients with type 1 or type 2 diabetes initiatedintermittently scanned glucose monitoring and over 98% persisted with the device at 12 months (56). Following initiation of the device, patients had a 39-49% reduction in hospitalizations for acute complications and a 32-40%reduction in diabetes-related Moreover, the reduction in hospitalizations persisted after 2 years (57).
  • In Belgium, a study of 1913 adults with type 1 diabetes were studied before and after nationwide reimbursement of intermittently scanned continuous glucose monitoring (58). Following the policy change, treatment satisfaction improved, there was a significant reduction in admissions for acute complications (severe hypoglycemia orketoacidosis), and there were fewer absences from work.
  • Among 41,753 patients with insulin requiring diabetes in an integrated health care delivery system, 3806 patientsinitiated CGM, which was associated with a greater reduction in A1C (adjusted treatment difference 0.40%, p<0.001), emergency department or hospitalization for hypoglycemia (adjusted difference -2.7%, p=0.001), areduction in number of outpatient visits and an increase in telephone visits (59). However, there was no difference in hospitalizations for hyperglycemia or ketoacidosis.
  • In a Medicare supplemental and commercial claims database study of 2463 patients with type 2 diabetes on multiple injections of insulin/day, intermittently scanned CGM was associated with a reduction in acute diabetes events (HR 0.39, 95% CI 0.30-0.51) and all cause hospitalizations (HR 68, 95% CI 0.59-0.78) at 6 monthscompared to the 6 months prior to initiation (60).

 

Recommendations

 

Patients should be adequately informed of the benefits and limitations of this technology, particularly with respect to the role for SMBG. At a minimum, structured education programs encompassing concepts such as carbohydrate counting and active insulin time (insulin on board) should be completed prior to considering RT-CGM, and payers mayrequire that patients demonstrate that they can reliably and consistently perform SMBG (61). Several expert groups have issued guidance in the use of RT-CGM.

 

  • In 2016, the Endocrine Society, co-sponsored by The American Association for Clinical Chemistry, the American Association of Diabetes Educators, and the European Society of Endocrinology, published guidelines for use ofinsulin pumps and CGM. The guidelines recommended RT-CGM in adults with type 1 diabetes and any A1C who are willing and able to use the devices nearly daily. The panel suggested short-term intermittent use for patientswith type 2 diabetes (not requiring prandial insulin) who had an A1C ≥7% and are willing and able to use the device (3).
  • The American Diabetes Association (ADA) Standards of Care recommend CGM in adults with type 1 diabetes andthose with hypoglycemia unawareness or frequent hypoglycemia (Table 2a). Among pediatric patients, the ADAnotes that CGM may reduce missed school days with regular usage (1).

 

Table 2a. ADA 2023 Recommendations for CGM

Group

Recommendation (Level of Evidence)

 

Real-time CGM

Intermittently Scanned CGM

 

Adults

Youth

Adults

Youth

MDI or CSII

insulin use

Should be offered (A)

Should be offered(B-T1D,

E-T2D)

Should be offered(B)

Should be offered (E-

T1D)

 

Should be used as close to daily as

possible (A)

Should be scanned frequently, at

least every 8 hours (A)

Basal insulin use

A

NA

C

NA

All

·       Devices are recommended for individuals or caregivers who can use the devices safely

·       The choice of device should be individualized based on patient centered factors.

·       People should have uninterrupted access to supplies to minimize gaps in monitoring (A)

·       Periodic RT-CGM, intermittently scanned CGM, or professional CGM

can be helpful where continuous use is not possible (C)

Diabetes and

pregnancy

CGM can help to achieve A1C targets in pregnancy when used as an

adjunct to pre- and postprandial SMBG (B)

A=Clear evidence from well-conducted, generalizable randomized controlled trials that are adequately powered;B=Supportive evidence from well-conducted cohort studies; C= Supportive evidence from poorly controlled or uncontrolled studies; E=expert consensus.

T1D=type 1 diabetes, T2D=type 2 diabetes, SMBG=self-monitored blood glucose

 

Table 2b. AACE Recommendations for CGM by Methodology

Method

Background/Therapy

Evidence*

BEL*

Grade^

RT-CGM

•       Problematic hypoglycemia

•       Lifestyle and other factors should also be considered

Low- Intermediate

1

B

isCGM

•       Newly diagnosed T2D

•       Non-hypoglycemic therapies

•       Motivated to scan device several times/day

•       Low hypoglycemia risk, desire for more data

Low/Expert Opinion

4

D

Diagnostic/ professional CGM

•       Newly diagnosed T2D

•       problematic hypoglycemia, but no access to personal CGM

•       non-insulin therapies as an educational tool

•       Trial use

Intermediate

1

B

Intermittent CGM

persons …who are reluctant or unable to commit to routine CGM use.

Intermediate

1

C

* Level of Evidence:

  • High (1) = randomized controlled trial (RCT) or meta-analysis of RCT
  • Intermediate (2) = meta-analysis including nonrandomized studies, network meta-analysis, nonrandomizedcontrolled trial, prospective cohort, case control, cross-sectional, hypothesis driven epidemiologic, open label extension, post-hoc analysis
  • Weak (3) = discovery/exploratory, economic, consecutive case series, case report, safety/feasibility, high impact basic research
  • None (4) = consensus, position, policy, guideline, any highly flawed study, lower impact basic science

BEL=best evidence level

^Grade is based upon evidence level, recommendation qualifiers, subjective factors, and consensus

 

Table 2c. AACE Recommendations for CGM—Patient Characteristics

Background/Therapy

Evidence rating

BEL

Grade*

3+ injections/day or CSII

High

1

A

Frequent/severe or nocturnal hypoglycemia or unawareness

Intermediate-High

1

A

Children/adolescents with T1D

Intermediate-High

1

A

Pregnant, 3+ injection/day

Intermediate-High

1

A

Gestational DM on insulin

Intermediate

1

A

Gestational DM no insulin

Intermediate

1

B

T2D, on insulin

Intermediate

1

B

* Level of Evidence:

  • High (1) = randomized controlled trial (RCT) or meta-analysis of RCT
  • Intermediate (2) = meta-analysis including nonrandomized studies, network meta-analysis, nonrandomizedcontrolled trial, prospective cohort, case control, cross-sectional, hypothesis driven epidemiologic, open label extension, post-hoc analysis
  • Weak (3) = discovery/exploratory, economic, consecutive case series, case report, safety/feasibility, high impact basic research
  • None (4) = consensus, position, policy, guideline, any highly flawed study, lower impact basic science

BEL=best evidence level

^Grade is based upon evidence level, recommendation qualifiers, subjective factors, and consensus

 

The 2021 American Association of Clinical Endocrinologists recommendations for use are summarized in Table 2band 2c. These include all adults and children with type 1 diabetes, especially those with severe hypoglycemia or hypoglycemia unawareness, and all patients with type 2 diabetes on multiple insulin injections, basal insulin, or sulfonylureas who are at risk for hypoglycemia (2).

 

In 2017, the Advanced Technologies & Treatments for Diabetes (ATTD) Congress organized an international consensus panel, consisting of physicians, researchers, and individuals with diabetes to analyze the existing literature and to provide guidance for utilizing, interpreting, and reporting CGM data (62). This was updated in 2019 (Table 3 and 4). These recommendations are supported by recent data from the DCCT demonstrating that a 10% reduction in time in target glucose range derived from 7-point self-monitored glucose profiles is associated with a 40% reduction in risk of microalbuminuria and a 64% reduction in risk of incident or progressive retinopathy (63).

 

Table 3. CGM-Based Targets for Different Diabetes Populations

Glucose Range

%Time in Range

 

Non-Pregnant Patients

Type 1 and Type 2 Diabetes

Older/High Risk Diabetes

>250 mg/dl (13.9 mmol/L)

<5%

<10%

>180 mg/dl (10 mmol/L)*

<25%

<50%

70-180 mg/dl (3.9-10 mmol/L)

>70%

>50%

<70 mg/dl (3.9 mmol/L)**

<4%

<1%

<54 mg/dl (3.0 mmol/L)

<1%

 

 

Pregnant Patients

Type 1 Diabetes

Gestational and Type 2 Diabetes #

>140 mg/dl (7.8 mmol/L)

<25%

-

63-140 mg/dl (3.5-7.8 mmol/L)

>70%

-

<65 mg/dl (3.5 mmol/L)

<4%

-

<54 mg/dl (3.0 mmol/L)

<1%

-

*Includes time >250, **Includes time <54 mg/dl, #Insufficient data

 

Table 4. Summary of ATTD Recommendations for CGM

Limitations of A1C

CGM should be utilized when there is a discrepancy in A1C and other measures of glucose control.

CGM should be utilized to assess hypoglycemia and glucose variability.

Guiding management and assessing outcomes

CGM should be considered for patients with type 1 diabetes and insulintreated type 2 diabetes who are not achieving targets or those with hypoglycemia.

All patients should receive training education regarding how to interpret andrespond to their data, utilizing standardized programs with follow-up.

Performance

No accepted standard exists for CGM system performance. However, a mean absolute relative difference ≤10% provides little additional benefit forinsulin dosing.

Definition and assessment of hypoglycemia

Clinical classification

Level 1: 54-70 mg/dl with or without symptoms

Level 2: <54 mg/dl with or without symptoms (clinically significant)

Level 3: cognitive impairment requiring external assistance for recovery

Quantification using CGM

% of values or time below a given threshold (54 or 70 mg/dl) Number of events (defined as CGM readings persistently below threshold for at least15 min. with recovery defined as persistent readings over the threshold forat least 15 min.) over a given reporting period)

Glycemic Variability

Coefficient of Variation should be the primary measure

Time in Range

The % time in hyperglycemia, hypoglycemia, and target range should

be reported.

CGM Metrics

Standardized reporting using the AGP and integration into electronic health records is recommended.

Key metrics:

·         Number of days worn (14 days recommended)

·         % time CGM is active (70% of data from 14 days recommended)

·         Mean glucose

·         Glucose Management Indicator

·         Glycemic variability (%CV, target <36%)

·         % Time in Range (TIR): 70-180 mg/dl (3.9-10.0 mmol/L)

%Time Above Range (TAR): 181-250 mg/dl (10.1-13.9 mmol/L), and >250 mg/dl (>13.9 mmol/L)

·         %Time Below Range (TBR): 54-69 mg/dl (3.0-3.8 mmol/L), and

<54 mg/dl (3.0 mmol/L)

 

Also, in 2017 the ADA and the European Association for the Study of Diabetes published a joint statement providing recommendations for systematic improvements in clinical use and regulatory handling of CGM devices (64).

 

Hospital Use

 

CGM is not currently approved for use in the hospital setting. However, during the COVID-19 pandemic, the FDA announced that it would not object to their use in an effort to support reduction in use of personal protective equipmentand risk of exposures to staff. Thus, there has been increasing interest in their use. Moreover, an increasing number of patients are using these devices in the ambulatory setting and want to continue their use in the hospital. Recent randomized trials support the use of CGM on the hospital wards, where it has been shown to be safe, and may reduce the frequency of hypoglycemia (65,66). In the ICU, there is concern that CGM may be less accurate due to factors such as edema, hypoperfusion, and acidosis but preliminary studies suggest use of CGM in conjunction with periodic point of care (POC) blood glucose (BG) within well-established protocols is safe and may reduce the need for POC BG (67).

 

In 2020, the Diabetes Technology Society sponsored a panel of experts in inpatient diabetes management to reviewthe evidence for us of CGM in the hospital (68). The panel agreed that CGM had the potential to improve clinicaloutcomes, particularly for patients who are unable to communicate signs or symptoms of hypoglycemia, but use is limited by lack of data demonstrating accuracy (particularly in the hypoglycemic range or in case of diabetic ketoacidosis, poor perfusion, or acetaminophen use) and clinical utility, and a lack of decision support systems, including infrastructure for communicating results to care teams and to the electronic medical record. The panelagreed that patients who are admitted with personal CGM devices should be allowed to continue use of such devices under the condition that they are able to self-manage the devices on their own and are followed by an endocrinologist or experienced practitioner who is specifically trained in their use. In particular, the panel advised implementing institutional policies that recommend continued capillary or blood glucose monitoring, ensuring that CGM data are not used for inpatient insulin dosing (since no CGM device is FDA approved in the inpatient setting), and requiring patients to sign safety waivers which illustrate the potential risks and benefits of continued use. Devices must beremoved for any MR or CT imaging. The panel made the specific recommendations for clinical care including:

  • Consider use of CGM to reduce exposures (such as for point of care glucose) and need for personal protective equipment in persons with highly contagious diseases.
  • Barring use in the setting of highly contagious disease, CGM values should be confirmed with point of care (POC) glucose prior to making treatment decisions.
  • Hospitals should develop implementation plans which include a process map, protocol, provider/staff/patient education and order sets.
  • Providers should recognize CGM pattern caused by compression of the device, which can cause a falsely low value.
  • Providers should ensure patients are not taking medications or supplements that can interfere with CGM.
  • Nurses should be adequately trained on use of CGM, inspect the insertion site every shift, and set expectations that POC values are still necessary to support ongoing use of CGM (typically every 6 hours).
  • Hospitals need to develop security protocols, data storage, visualization tools, and integration within the electronic medical record to support the use of CGM.
  • Hospitals need to identify CGM values in the electronic medical record to distinguish values from blood glucose values.
  • Hospitals need to adopt the Unique Device Identifier (UDI) to track devices in the electronic medical record.

 

Limitations of Use

 

It should be emphasized that most prospective randomized controlled trials enroll highly motivated patients. In the real-world setting, there are concerns about limited resources for training, and less motivated patients may be overwhelmed with the additional data, particularly where complex algorithms are required. Nevertheless, in the Type 1 Diabetes Exchange Registry, CGM use increased from 7% in 2010-2012 to 30% in 2016-2018, and rose more than 10-fold in children (69). A1C levels were lower in CGM users compared to nonusers. While CGM use has improved substantially over time, more than half of respondents cited cost or insurance coverage as a significant barrier to use (70). Moreover, disparities in prescribing patterns and implicit bias have been described (71,72,73). Modifiablereasons for avoiding use include the hassle of devices (47%) and aversion to having a device attached to the body (35%). Skin reactions and/or difficulty with adhesion are well known and are an important cause of discontinuation (74). Methods of addressing this barrier such as use of barriers, overlay patches, or topical antihistamines and corticosteroids have been described but additional research is needed (75).

 

In a multi-national study of 263 patients, persistent sensor use for 12 months was only 30% (76). Improvement in A1C was associated with higher A1C at baseline, older age, and more frequent sensor use. Diabetes related hospital admissions were reduced following the initiation of sensor augmented pump therapy and fear of hypoglycemia improved. In the 6-month follow- up phase of the JDRF-CGM trial, RT-CGM was initiated in the control group in a manner that more closely approximates clinical practice (77). Investigators found a significant reduction in CGM use inall age groups over time. However, increasing sensor use was associated with A1C reduction. It is likely that adherence will improve as technologies improve.

 

Other limitations include possible interference with acetaminophen, ascorbate, and other active agents in glucose-oxidase based electrochemical sensors. They are also dependent on both the sensitivity and specificity on the enzyme availability on the electrode surface. There are well known delay artifacts due to the time lag between glucose concentration in the interstitial fluid and blood glucose. These time ranges, often between 5 and 10 minutes, are not crucial to analyzing retrospective data, but can be critical when CGM is indicated for real-time decision making (78).

 

Daily Use

 

Patients must be aware that sensor readings can deviate from actual blood glucose measurements, particularly during rapid glucose changes such as that which occurs post-meal or during exercise. Calibration, where necessary, should not be performed when trend arrows indicate rapid swings in glucose. While systems are becoming more reliable, patients may need to verify sensor readings before taking action such as meal boluses or treatment of hypoglycemia depending on the device, even if a device is approved for nonadjunctive use.

 

Alarm thresholds should be set in order to maximize patient compliance, keeping in mind that the sensitivity fordetecting hypoglycemia decreases as the threshold is reduced below 70 mg/dl. Conversely, specificity improves to amuch smaller degree at lower thresholds, and thus false alarms may not be reduced substantially.

 

Several algorithms have been published that provide specific guidance to patients for responding to trend arrows and alarms and are summarized below. All algorithms are complex and are not integrated within bolus calculators ofexisting insulin pumps. Therefore, they should only be implemented in patients who have demonstrated anunderstanding of CGM technology, including lag times between CGM and BGM, calibration procedures, alerts andtrend arrows, as well as understanding of insulin action time and the risks of insulin stacking. In one small study, trend arrows were accurate approximately 79% of the time outside of mealtime windows (30 minutes before and 120 min after carbohydrate intake) but this dropped to ~60% within mealtime windows (80). Thus, algorithms are not intended for use post-meal. The use of automated insulin delivery systems should increase safety and efficacy and reduce the complexity of the trend arrow approach.

 

  • The algorithm by Jenkins et al. provides tiered recommendations that are based upon the meter glucose and sensor trend arrows (81). In addition, the algorithm advises patients how to review downloads of the data periodically (weekly) and make adjustments. Patients who were randomly assigned to sensor augmented pump with the algorithm had lower A1C and reported better quality of life at 16 weeks compared to patients who did not get the The effect on quality of life persisted at the 32-week follow-up, and was associated with A1Creduction. Importantly, patients who received the algorithm at 16 weeks after initiating sensor augmented pump did not benefit.
  • The DirecNet study algorithm (for use with the Navigator system) recommended that patients increase or decreasethe meal + correction bolus by 10-20% based upon the rate of change and provided specific instructions for responding to alarms (82). Algorithm use was high in the first 3 weeks but dropped off by week 13, despite increasing insulin self- adjustments, possibly as patients became more independent over time.
  • Subsequent methods recommended adjustment of only the correction insulin dose by the amount needed to cover a glucose level that is incrementally higher or lower than the current glucose, based upon the trend arrow (83,84).
  • Klonoff and Kerr proposed a more straightforward correction dose (in 5-unit increments), based upon the trend arrow and the patient’s insulin sensitivity (85).
  • A consensus statement facilitated by the Endocrine Society provides expert guidance on the use of trend arrows for making treatment decisions (86). The guidance recommends adjustment of boluses pre-meal and no sooner than 4 hours post-meal in 0.5-unit increments based upon the trend arrow and the patient’s sensitivity. The statement recommends no additional treatment within 2 hours of a previous meal bolus, and correction bolus using the bolus calculator or usual correction dose only in the 4 hours after a meal. Similar expert guidance has been developed for the Freestyle Libre system (87).
  • A more recent adaptation of the Endocrine Society guidance incorporated pre-meal glucose levels in addition to theinsulin sensitivity (88) and a small randomized study demonstrated it was more effective than the incorporation of insulin sensitivity alone, particularly among insulin pump patients (89).

 

Overview of Stand-alone Personal RT-CGM and IS-CGM Systems

 

The first RT-CGM (Guardian, MedtronicR) was approved in 2004. Since then, additional models and other devices have entered the market, and accuracy and patient satisfaction have improved. Several personal continuous glucosemonitors have been approved by the US Food and Drug Administration (FDA) for use in the United States or carry CE marking for use in Europe and are currently on the market (Table 5). For a full review of regulatory requirements for glucose monitoring devices, the reader is referred to one of several excellent reviews (90).

 

Table 5. Comparison of Subcutaneous Continuous Glucose Monitoring Devices

 

 

 

Calibration required

Confirmatory Fingersticks requiredprior to

treatment

 

 

Real-time alerts

 

 

Sensor Life (days)

 

 

Warm-up (hrs)

 

Removefor MRI, CT

diathermy

 

 

Acetaminophen interference

Dexcom G5

Y

N

Y

7

2

Y

Y

Dexcom G6

N

N

Y

10

2

Y

N

Dexcom G7

N

N

Y

10

0.5

Y

N

Medtronic

Guardian 3

 

Y

 

Y

 

Y

 

7

 

2

 

Y

 

Y

FreeStyle Libre

14 day

 

N

 

N

 

N

 

14

 

1

 

Y

 

N

FreeStyle Libre 2

N

N

Y

14

1

Y

N

FreeStyle Libre 3

N

N

Y

14

1

Y

N

Eversense

(surgical implant)

 

Y

 

N

 

Y

 

180

 

NA

 

Y

 

No

 

GUARDIAN CONNECT

 

The Guardian Connect utilizes the Medtronic Guardian Sensor 3, the Guardian Connect transmitter, and the GuardianConnect app to transmit data via Bluetooth every 5 minutes to the user’s smart phone or device (initially only available on iOS devices) via the Guardian Connect App on smartphones and via CareLink personal and professional software. A separate receiver is not available with this system. Data can be shared with others remotely, and SMS messages can be sent in times of hypoglycemia. The system is only approved for adjunctive use and at least 2 daily fingerstick calibrations are required.

 

DEXCOM G6

 

The Dexcom CGM utilizes a glucose oxidase sensor at the tip of a wire that is implanted in the subcutaneous space. The data are transmitted wirelessly and are displayed on a separate receiver (personal smartphone or device specificreceiver). The Dexcom G6 has a sensor life of 10 days, no longer requires calibrations, and minimizes interference by acetaminophen. G6 is also associated with a smartphone app that allows the patient to log activity, set reminders or alarms, and physically see their glucose levels and trends throughout their time wearing the device. The Dexcom CLARITY Diabetes Management Software organizes and presents the patient’s blood glucose data. Dexcom SHARE app allows users to share data with up to 10 other individuals.

 

DEXCOM G7

 

The Dexcom G7 features a 60% smaller size (the size of 3 stacked quarters) vs. the G6, is fully disposable, and has a shorter 30-minute warm up time and a 12-hour grace period to replace completed sensors. The overall MARD was reported to be 8.2% with the abdomen and 9.1% on the arm (17). Consistent with previous studies, accuracy is lower at lower sensor glucose, higher glucose rate of change, and on day one of wear.

 

FREESTYLE LIBRE 14 Day

 

The sensor utilizes Wired Enzyme™ technology in which the enzyme and mediator are co- immobilized on the sensor. It offers factory calibration, and therefore nearly eliminates the need for fingerstick monitoring. However, patients are still advised to perform SMBG whenever an alert appears on the reader display (which occurs when the glucose isrising or falling rapidly) or whenever the glucose value does not fit the patient’s symptoms. The reader contains a built-in meter for this purpose. The sensor is FDA approved for 14 days of use. The system is not approved for use in children under age 18, or during pregnancy or in persons requiring hemodialysis. The Libre has minimized the interference by acetaminophen which is present in other devices but interference from other substances such as ascorbic acid or aspirin may be possible. The Libre 14 day differs from other CGM devices in that the system does not alert the user for glucose values surpassing a high or low threshold. In addition, glucose values are not automatically made available to the user but are easily and instantly accessed by scanning the sensor with a handheld reader or theassociated app FreeStyle LibreLink. However, this product may be attractive option for patients who are averse to the hassle imposed by other RT-CGM devices. Glucoses are measured every minute and recorded every 15 minutes. Data can be accessed using the reader or downloaded to LibreView cloud based online management system, or using the FreeStyle Libre desktop software. The MARD is reported by the manufacturer to be 9.7% overall, and as with other CGM devices, less accurate on day 1 of wear and in hypoglycemia range (91).

 

FREESTYLE LIBRE 2

 

The FreeStyle Libre 2 system offers real-time alerts for high or low glucose values and improved accuracy, approved for ages 4 years and older (92). However, users must continue to scan the device to obtain glucose readings.Moreover, similar to the Libre 14 day, the sensor memory is only 8 hours and glucose data are lost if the sensor is scanned less frequently.

 

FREESTYLE LIBRE 3

 

The FreeStyle Libre 3 is even smaller than other devices (the size of 2 stacked pennies), does not require scanningunlike older models, but does require the use of a compatible smartphone. The bluetooth range is improved from 20 to 33 feet. Accuracy is improved compared to the Libre 2, with an overall MARD of 9.2% in adults and 9.7% in children (16).

 

EVERSENSE

 

The Eversense system (Senseonics) is a 90-day implantable sensor that uses fluorescent technology to sendmeasures via a rechargeable transmitter which rests just above the skin to a smartphone app titled Eversense NOW (93). In a pivotal clinical study of 71 patients with type 1 and type 2 diabetes, there were no device-related serious adverse events, and the MARD was 11.1%, with over 99% of samples in clinically acceptable error zones A and B of Clarke Error Grid Analysis (94). One study reported interference with tetracycline and mannitol, but not with acetaminophen or ascorbic acid (95). The Eversense XL CGM system consists of a 180-day implantable sensor thathas been shown to have acceptable safety and accuracy with an overall MARD of 9.1% (96). This option may be particularly useful for patients with privacy concerns, physical disability, needle phobia, allergies or other difficulty with adhesion, or activities or professions that may be barriers to external wear (97).

 

Sensor Augmented Pumps

 

To date the largest A1C reductions have been observed when sensors are initiated with insulin pump technology. In the observational (nonrandomized) COMISAIR study, patients initiating CGM (with or without insulin pump) achieved significantly larger reductions in A1C (-1.2%) compared to subjects initiating insulin pump alone (-0.6%) or remaining on injections alone (- 0.3%) (98). There was no difference in outcomes between the DexCom G4 and Enlite sensor.

 

A reduction in time spent in hypoglycemia was observed only in patients using CGM (8% vs 6%, p<0.001).

 

STEPS TOWARDS AN ARTIFICIAL PANCREAS

 

Until recently, RT-CGM technology has operated completely independently of insulin delivery. By combining continuous basal insulin delivery during fasting periods with discrete bolus doses of insulin at mealtimes, insulindelivery can be crafted to mimic the natural pattern of pancreatic insulin release. An artificial pancreas consists of: 1) an automatic and continuous glucose monitor; 2) an implanted continuous insulin delivery system; 3) a control processor to link the insulin delivery rate to the glucose level; and 4) a signal to send the glucose level to the body surface for continuous display onto a monitor. Limitations to full implementation include sensor accuracy and lag time, inadequate onset and offset of currently available rapid acting insulin analogs, meal challenges, and changes in insulinsensitivity due to circadian rhythms, exercise, menstrual cycles, and intercurrent illness (99). However, even incremental advances improve glucose control without increasing the complexity of decision-making on the part of the patient. These include:

  • Low glucose (threshold) suspend: the insulin pump suspends when the glucose decreases below a pre-set value.
  • Suspend before low: insulin pump suspends when hypoglycemia is
  • Hybrid closed loop: insulin delivery increases or decreases based upon the sensor glucose value but meal boluses are still required.
  • Closed loop control: fully closed loop delivery without the need for meal boluses
  • Dual hormone systems: these are hybrid closed loop or closed loop control systems that utilize glucagon or other peptides (such as amylin) in an effort to more closely mimic the physiology of the endocrine pancreas.

 

The long-term safety, efficacy, cost, and cost-effectiveness of an artificial pancreas are still largely unknown at this time. However, the urgency of this technology is demonstrated by the #WeAreNotWaiting movement, which has given rise to home-grown, crowd-sourced, patient driven systems that utilize existing devices which are linked by open-source software, such as Open Artificial Pancreas System, and Loop. Recently Tidepool Loop received FDA approval (100). A retrospective observational study of patients with Type 1 diabetes demonstrated lower mean glucose, higher time in target range, and less time in hypoglycemia using Open Artificial Pancreas Systems (OpenAPS) compared to sensor augmented pump use alone (101). In general, open-source systems carry safety concerns, particularly among less tech-savvy patients, in the absence of regulatory approval (102). However, the healthcare provider can providesafety recommendations as well as a back-up plan in case of system failure (103). The reader is referred to one of several reviews as a detailed review is beyond the scope of this chapter (104,105).

 

Threshold Suspend

 

Progress is expected toward a fully functional closed loop system in incremental steps. The first step toward a fully automated “artificial pancreas” is the low glucose suspend feature, which is now available. The Medtronic 530Gsystem, containing the Veo insulin pump and Enlite sensor, is the first sensor augmented pump with low threshold suspend and uses the same sensor as the more recent 630G system. The Enlite sensor accuracy is significantly improved over the previous Sof-sensorR, with a MARD of 13.6% when used with the 530G (106). The Enlite is also one-third of the size of Sof-sensor and the filament is 38% shorter. The Enlite sensor may be worn up to six days. The low threshold suspend SmartGardTM technology suspends the pump for up to two hours in the event of sensor detected hypoglycemia in which the user does not respond to the alarm. Prior to suspension, a “siren” sounds which is distinct from other high or low alerts, and the suspension can be overridden at any time. The MiniMed Connect mobile accessory sends sensor data to an app on a mobile device where data can be viewed (available only with the 530G system). A study that enrolled 247 patients with type 1 diabetes and documented nocturnal hypoglycemia to sensor-augmented pump with or without a low-glucose threshold-suspend feature demonstrated similar A1C between groups at 3 months but lower frequency of nocturnal hypoglycemia (107). Similar findings were demonstrated in an Australian study of 95 patients, in which the incidence rate ratio for hypoglycemia was 3.6 (95% CI 1.7-7.5, p<0.001) (108). There were no reports of DKA in either study.

 

Suspend Before Low

 

The next incremental step in closed loop systems is the suspend before low feature, currently available in theMedtronic 640G (approved only in Europe) and the 670G systems. This feature automatically suspends insulin delivery 30 minutes before a low glucose threshold is predicted and resumes delivery once the glucose recovers, without alerting the patient. In a 6-month randomized study of 154 children and adolescents with type 1 diabetes, the 640G system reduced the time spent in hypoglycemia from 2.6 to 1.5% without causing a change in A1C (109). The t:slim X2 Insulin Pump incorporates Basal IQ technology with predictive low glucose suspend using the Dexcom G5 or G6 sensor. In a randomized cross-over study of 103 participants with type 1 diabetes age 6-72 years of age, predictivelow glucose suspend resulted in a 31% reduction in time spent in hypoglycemia < 70 mg/dl without a change in meanglucose or time in hyperglycemia (110).

 

Hybrid Closed Loop (HCL)

 

This step refers to sensor glucose driven automatic adjustment of basal insulin with or without additional auto boluses,and still requires the patient to bolus for meals. In a recent consensus statement, an ideal candidate for automated insulin delivery systems (103):

  • Is technically capable of managing a pump, has basic carbohydrate counting skills, and is able to implement a back-up plan (including the use of manual injections).
  • Has realistic expectations of system In particular, several situations that are unique to HCL are worth emphasizing:
    • Bolusing: pre-blousing approximately 15 minutes prior to meals is critical to maintain In many systems,delayed boluses not only cause early postmeal hyperglycemia but also precipitate delayed hypoglycemia as the system has already begun to augment insulin delivery in response to hyperglycemia.
    • Exercise management: Similarly, carbohydrate loading prior to exercise while using HCL systems will only stimulate insulin delivery and thus is recommended that users implement other means for management such as setting a higher target, typically with a designated exercise mode, or exiting to manual mode with temporarybasal insulin reduction or temporary suspension of the pump.
    • Hypoglycemia management: HCL users typically need fewer carbohydrates (about half) to manage hypoglycemia since the pump has generally already suspended insulin delivery based upon glucose trends.
  • Has adequate support, including diabetes education, insurance coverage, and caregiver or other social support where relevant.
  • Has the ability to transmit data to the healthcare
  • Is mentally and psychologically able to implement AID

 

On the other hand, there does not appear to be an ideal threshold A1c for determining candidates for HCL therapy, asthose with lower A1c may benefit by reducing TBR and those with higher A1c benefit from reductions in hyperglycemia without the perceived risk of ketoacidosis traditionally attributed to initiation of insulin pump therapy (111). Thus, less ideal candidates may obtain the greatest benefit in terms of achieving glycemic targets.

 

HCL demonstrates improvements in a range of glycemic outcomes and may confer psychological benefits as well.Most studies have enrolled patients with type 1 diabetes. More data are needed for special populations including type 2 diabetes, especially those with very high insulin requirements, pregnancy, acute illness, steroid use, renaldysfunction, and persons in assisted living facilities (112).

 

As with CGM, it is important to evaluate these systems in the real-world setting, where user experience can differ from that of the highly controlled and supportive research environment. Cost and insurance hassles, as well as user wear issues are the most commonly reported barriers to use of any diabetes related device. These barriers contribute todiabetes distress and depressive symptoms which can impede self-management behaviors (70,112). HCL systems improve glucose control but may also introduce additional alarms or alerts which are needed for safety (such as threshold alerts for hyperglycemia or hypoglycemia or HCL mode exits due to insulin delivery exceeding the system’s guardrail) or ongoing functionality such as calibration of the CGM (113). Newer systems have attempted to address many of these barriers through improved algorithms or other features (Table 6). Devices differ considerably with respect to algorithms used for insulin adjustment and a number of other features (Table 6) (114,115,116). There are few head-to-head studies comparing the efficacy and safety of available HCL systems. Details of select systems are presented next.

 

MEDTRONIC 670G

 

The first system to gain FDA approval is the Medtronic 670G, which adjusts basal insulin delivery every 5 minuteswhen in auto mode. This system utilizes the Guardian sensor 3, which offers enhanced sensor accuracy, with an overall MARD of 9.64% (117). The system was associated with a reduction in A1C from 7.4 to 6.9% and there were trends in improvement of time in target range and hypoglycemia in a non-randomized study of 124 patients with type 1 diabetes (118). A subsequent randomized trial of 151 adults and children demonstrated a significant reduction in A1c and TBR compared to insulin pump without CGM (119). There are few studies addressing long-term use however. In a 1-year prospective observational study of 84 patients, 28% stopped using auto mode by 3 months, and 33% discontinued by 12 months (120). The most common reasons for discontinuation included sensor issues (62%) and difficulty obtaining supplies (12%), fear of hypoglycemia (12%), and preference for injections (8%) or sports (8%). In astudy of 92 youth, 30% discontinued HCL, typically between 3 and 6 months after initiation, due to issues such asdifficulty with calibrations, alarms, and extra time needed for operation (121).

 

MEDTRONIC ADVANCED HYBRID CLOSED LOOP (AHCL) SYSTEM (780G)

 

This HCL system is approved for use in Europe and features substantially reduce frequency of alerts, improved time in auto mode, remote software updating, an adjustable target setting as low as 100 mg/dl and enable users to view data via an app on mobile devices (122). In a single arm study of 157 adolescents and adults the 780G system resulted in nearly 95% time in automated mode with 1.2 exits per week, improved A1C, TIR, and TBR (123). In a randomized study of 82 persons with type 1 diabetes using multiple injections per day and isCGM, AHCL resulted in improvements in A1C (-1·42%, 95% CI -1·74 to -1·10; p<0·0001) and TIR, but no difference in hypoglycemia (124). There was an improvement in treatment satisfaction, fear of hypoglycemia, and similar diabetes quality of life. By comparison in a randomized study of 41 participants with type 1 diabetes who were naïve to both CGM or insulin pump technologies, AHCL also resulted in improvements in TBR (125). In a real-world study of 3211 youth (<age 15 years) and 8874 individuals >age 15 years, ACHL demonstrated >90% treatment persistence over 6 months (126). The ACHL system was reported to have better glucose monitoring treatment satisfaction but similar diabetes distress, technology attitudes, and fear of hypoglycemia compared to the 670G system (127).

 

TANDOM CONTROL-IQ

 

This system utilizes a t:slim X2 insulin pump with a calibration-free Dexcom G6 sensor (128). In a 6-month trial of 168 patients (age 14-71) randomized 2:1 to hybrid closed loop vs. sensor augmented pump alone, the % time in target 70-180 mg/dl was increased by 11% more in the hybrid closed loop group compared to sensor augmented pump(p<0.0001), with improvements in hypoglycemia, mean glucose and A1c. Moreover, real-world outcomes among 1435 persons with type 1 diabetes included reduced impact of diabetes on life, improved device related treatment satisfaction, and improved emotional well-being (129).

 

OMNIPOD 5

 

The Omnipod 5 is a HCL system that uses the Omnipod DASH platform (130). In a single arm study, the Omnipod 5 demonstrated a reduction in A1C of 0.38%, increase in TIR of 9.3% and decrease in TBR of 1.6% (131).

 

Table 6. Comparison of Hybrid Closed Loop Systems

 

Medtronic 670G/770G

Medtronic780G

Tandem T:Slim with Control IQ

Omnipod 5

iLet BionicPancreas

Insulin delivery

Tubing

Tubing

Tubing

Tubeless (pod)

Tubing

CGM

Guardian 3

Guardian 4

Dexcom G6

Dexcom G6

Dexcom G6

Reservoir capacity(unit)

300

300

300

200

180

Calibration needed

Yes

No

No

No

No

Supplies

DME

company

DME

company

DME company

Pharmacy

To be determined

Control via smartphone

Dataviewable from Smartphone

Data viewablefrom Smartphone

Smartphone bolus

Compatible smartphone controller

No

Algorithm initiation

48 hours in manualmode to estimate

TDI

48 hours in manual mode to

estimate TDI

Weight and TDI entry with maximumdelivery of 50% TDI over 2 hr

TDI estimated from programmed

basal rates

Weight entry

Bolus automation

No

Yes, every 5 minutes

up to 1 auto-correction bolus//hr if glucose predicted >180 mg/dl

No

unknown

Other inputs

CIR, AIT

Unable to overridebolus dose

CIR, AIT

CIR, ISF, AIT is fixedat 5 hours

CIR, ISF, AIT

Boluscalculator uses CGM rate of change

“Usual for Me”, “More”, or“Less” customized by

meal

Extended bolus

No

No

Yes, up to 2 hr

No

No

Algorithm adjustment

Every 6 days

Every 6days

TDI used to scalebasal changes

Every 3 days with pod change

Continuously or based on

change in entered weight

Target

120 mg/dl

100, 110,

120 mg/dl

112.5-160 mg/dl

110, 120, 130,

140, 150 mg/dl, customizableby time of day

100, 110, 120,

130 mg/dl, customizable by time of day

Exercise

Temp target 150 mg/dl for

2-12 hour

Temp target 150 mg/dlfor

2-12 hour

Exercise target 140-160 mg/dl—cannotprogram duration

Sleep mode: target 112.5-120 mg/dlwithout auto-correction bolus, programmable

Target 150 mg/dl, “less aggressive”,for 1-24 hr

None

Safety mode*

Yes, for 670/770G

results in forced exits from HCL

Yes, exits to Safe Basal up to 4hours (5.7 vs. 1.7

x/week with 670G) (130).

Not applicable(defaults to basal rate settings)

Yes, forced exits “rare”

Yes, BG-run mode uses manuallyentered BG upto 72 hours, forces switch to

alternate therapy.

PID=proportional integral derivative (system with continual change in response to error between actual and targetvalues). MPC=model predictive control (dynamic reference model serves as a basis. TDI=total daily insulin dose,CIR=carbohydrate to insulin ratio, AIT=active insulin time, ISF=insulin sensitivity factor. *Safety mode provides amechanism to ensure insulin delivery in case of loss of sensor input or threshold for insulin delivery guardrail is reached.

 

Closed Loop Systems (CLC)

 

Additional steps toward closed loop control (CLC) insulin delivery require algorithmic insulin adjustments, whicharguably present additional safety concerns. Overnight CLC insulin delivery is relatively straightforward, whereas post-meal control and exercise effects remain the most challenging of events to manage. Until recently, most randomized studies have been small and reported only short-term outcomes, often in controlled settings.

 

ILET BIONIC PANCREAS

 

The iLet Bionic pancreas was approved by the FDA in 2023. This insulin pump is initiated using the patient’s body weight and requires meal announcements (designated as small, medium, or large) but not formal carb counting and thus represents an incremental step toward a fully closed loop insulin pump. In A 13-week multi-center randomized study of 219 participants with type 1 diabetes demonstrated a greater reduction in A1c with the iLet bionic pancreas (-0.5%, [95% CI -0.6 to -0.3; P<0.001) but no difference in hypoglycemia compared to standard care (132).

 

OTHER SYSTEMS

 

Systems have utilized single hormone (rapid acting insulin only) or dual hormone (both fast- acting insulin analog and glucagon to imitate normal physiology) as directed by a computer algorithm (Figure 4) (133). At this time, there areinsufficient data demonstrating the superiority of one system or algorithm compared to others. The three most common algorithms are:

  • Model Predictive Control (MPC): predicts future glucose levels and adjusts insulin delivery in response.
  • Proportional Integral Derivative (PID): calculates the deviation of glucose from target to determine insulin delivery.
  • Fuzzy Logic (FL): mimics insulin dosing based upon clinical

 

A meta-analysis of 40 randomized studies (35 studies using insulin alone and 9 dual hormone studies) including 1027participants with type 1 diabetes demonstrated a significant increase in % time in target range (70-180 mg/dl, weighted mean difference 9.6%, 95% CI 7.5-12%), as well as less time in hypoglycemia or hyperglycemia, regardless of type of system (134). In another meta-analysis of 24 studies and 585 participants (7 studies using dual-hormone therapy and 20 studies of insulin only) reported greater improvement in time in target with artificial pancreas systems (12.6%, 95% CI 9.0-16.2, p<0.0001), and greater improvement with dual hormone compared to single hormone systems (135). Another meta-analysis of studies with at least 8 weeks duration confirmed these findings (136). A systematic review and meta-analysis of 25 studies in 504 children demonstrated superior %TIR with CLC and bi-hormonal systems vs. single hormone systems (137).

 

Figure 4. Dual hormone Closed Loop Control system.

 

MINIMALLY INVASIVE AND NON-INVASIVE GLUCOSE MONITORS

 

Continuous hypoglycemia detection systems using current sensing technology must be either implanted (fully or partially, either subcutaneously or into a blood vessel). Implantation is more secure, but may be associated with biocompatibility problems or local irritation. Less invasive methods may be categorized as minimally invasive or noninvasive. Minimally invasive techniques extract fluid (tears or interstitial fluid) while noninvasive technologies do not.

 

Minimally invasive methods include electrical, nanotechnology, and optical approaches while noninvasive techniquesrely on some form of radiation without the need to access bodily fluids. Noninvasive methods frequently incorporate electric, thermal, optical, or nanotechnology methods for detection. Many noninvasive devices under development are aimed for non- continuous monitoring as they often require controlled surroundings including factors such as light, motion and temperature.

  • Optical approaches utilize reflective, absorptive, or refractive properties of infrared and optical bands of the light spectrum to detect glucose. Pure optical methods under development utilize Raman and Near infra-red spectroscopy.
  • Thermal methods detect glucose via the far-infrared band of the spectrum and provide noninvasive approaches for glucose monitoring.
  • Electric methods use electromagnetic radiation, currents, or ultrasound approaches to detect dielectric properties of Reverse iontophoresis has been employed with early minimally invasive approaches while bioimpedance spectroscopy has been used in recent noninvasive approaches.
  • Nanotechnologies aim to miniaturize existing technologies, including fluorescence and surface plasmon resonance (SPR) approaches (138).

 

Few devices (other than interstitial CGMs discussed above) have demonstrated high levels of accuracy recommendedby expert groups, though several have been approved by CE or FDA (139).

 

MOBILE TECHNOLOGY AND DECISION SUPPORT

 

It has become increasingly clear that the isolated use of glucose monitoring technologies without a specific plan toaddress the data provides minimal benefit, particularly among patients with type 2 diabetes or who are not using insulin (140). In order for glucose monitoring to provide the most benefit, patients and providers must be able to easilyobtain and communicate the data. Data must be organized in such a way that patterns can be identified, and patients must receive feedback at the point of care. The widespread use of mobile devices provides opportunities for data collection, analysis, and communication of results with health care providers as well as facilitates digital or remote clinical models of care (141). Finally, as healthcare providers are inundated with more data and spend increasingamounts of time using electronic medical records, it has also become paramount that devices and or reports from the devices communicate or interface with these systems (142).

 

Hurdles to wider implementation of mobile technology include the lack of usability (both for patients, as well as providers who may be expected to review and act upon reports), safety, efficacy (including long-term adherence), and cost-effectiveness studies (143). The lack of data is in part due to the rapidly changing technology itself, which rendersthe technology obsolete by the time a vigorous clinical trial is conducted and published. The fee for service model is amajor barrier to adapting many glucose monitoring technologies, which often require frequent feedback and treatment adjustments, efforts that are not reimbursed without an actual office visit. Finally, cyber security is a big concern for all medical devices, especially for devices that are controlled by a smartphone (144).

 

Device Downloading, Connectivity, and Interoperability

 

Manual recording of glucose data is fraught with inaccuracies (145). Most monitors can be downloaded, via a tethering cable or wireless connection, either by the patient or healthcare provider. Each glucose monitoring devicegenerally works with its own proprietary management software. However, several programs (Tidepool,Glooko/Diasend, Carelink by Medtronic, Accu- chek) are capable of downloading and organizing data for multiple different devices (146).

 

Reports are standardized across all device downloads, facilitating efficient and actionable patient and healthcare provider review. These programs also facilitate population health and telehealth strategies (discussed below). The Nightscout Project is a crowd sourced application that provides a free mobile technology platform for patients whowant to access their devices in real time on any mobile device (147). Recent data suggest that retrospective weekly review of data is associated with improved TIR (148,149) as well as patient reported outcomes including confidence in avoiding hypoglycemia, overall well-being and diabetes distress (150).

 

Direct connectivity of blood glucose or CGM levels to cell phones or other devices not only improves data integrity but may also simplify the assimilation of glucose levels with other data such as insulin use, carbohydrate intake, andactivity levels for the purpose of facilitating insulin dose adjustments in real time or retrospectively. Cell phone connectivity may also improve communication with providers. A few meters with direct cellular capability are available.

 

Devices with direct cellular or Bluetooth connections may be paired with apps that facilitate collection, communication, and analysis of a variety of data and provide tools for education (such as nutrition information) at the point of care.

 

Currently, both the Tandem t:slim X2 and Insulet’s Omnipod 5 System are FDA approved for remote blousing via a cell phone app (151,152). A regulatory pathway has been developed for alternate controller enabled (ACE) infusion pumps which can be operated in conjunction with interchangeable components, particularly CGMs (153). In 2019, theFDA approved the first such devices (Tandem t:Slim X2 and Omnipod DASH system).

 

Diabetes Apps

 

A variety of stand-alone smart phone applications that support glucose monitoring are also available. Most provide information and track data (usually manually entered), some allow insulin or carbohydrate documentation, facilitate carbohydrate or calorie counting, promote weight loss, track or promote physical activity, enhance medication adherence, and use motivational or self-efficacy approaches, and a few provide an insulin dosing calculator. Simple apps provide information or tracking functions while more sophisticated approaches incorporate gaming theory and machine learning approaches that learn from the user’s previous experiences to optimize interactions. Apps have shown limited magnitude and sustainability of effect due to a variety of factors, including user fatigue, require continuous data entry (e.g., most apps do not connect directly with a glucose meter), and lack of integration with the health care team. Moreover, most apps have not been evaluated by the FDA or other regulatory agencies. Data privacy is also a concern, as no federal regulations currently prevent app developers from disclosing data to third parties. Most apps (81% in one survey of Android apps) do not have privacy policies, and of those that do, 49% share user data with third parties (154). Expert groups have developed policy or guidance statements to improvestandardization and functionality (155,156,157).

 

Efficacy

 

While the data are still evolving with respect to mobile diabetes applications, several systematic reviews and meta-analyses demonstrate modest (~0.5%) reductions in A1C in persons with type 2 diabetes, especially among younger patients, apps that provide healthcare provider feedback, or had other features including wireless entry of data (158,159,160,161). The Agency for Healthcare Research and Quality published a systematic review of comparative effectiveness studies assessing apps or programs available through a mobile device for the purpose of diabetes self-management (162). For type 1 diabetes, 6 apps were identified, 3 of which were associated with improvement in A1c, 2 of which were associated with improvement in hypoglycemia. Five apps for patients with type 2 diabetes were identified, 3 of which were associated with improvement in A1c. Efficacy is variable, in part because app features vary but also because apps are often studied as part of a multi-component intervention, making it difficult to assess individual elements, particularly the effect of additional health care provider support. Other researchers have focusedon identifying standard evidence-based features that should be included in diabetes apps, such as education, glucose monitoring, and reminders (163,164).

 

Usability

 

In a systematic review of 20 studies, only one third of the 20 apps met the authors’ health literacy standards (165).Usability was measured in 7 studies through satisfaction surveys from patients and experts, and ranged from 38-80%. The most common usability problems were multi-step tasks, limited functionality, and poor system navigation. While many apps are rated high quality for performing a single task, most do not address diabetes self-management tasks comprehensively (166) or otherwise do not function properly (165,167).

 

Decision Support

 

The use of pattern management software improves health care provider efficiency and accuracy in identifying needed therapeutic adjustments (168,169). Software programs provide graphs or charts and may in some cases provide dosing advice, either for the healthcare provider or directly to the patient.

 

Insulin Dosing Calculators

 

Insulin dosing calculators have been used for years as a means of incorporating glucose measures into routine practice, largely in concert with continuous insulin infusion pumps. While numerous apps have become available forbolus insulin calculation and basal insulin titration, it is important to note that only a few have been formally evaluated and approved by regulatory agencies. In addition, many still require manual data entry, few integrate within existing electronic medical records, and published evidence for efficacy is limited (170). All approved insulin calculators or dose titration apps require a prescription or need to be set up by a healthcare provider. Many such apps operate in conjunction with connected meters and insulin pens, which are subject to regulatory oversight and long-term support (171). Such support ensures safety and that software is updated to address any problems with operation and device compatibility. The functionality of connected pens ranges from insulin tracking functions, including insulin on board calculations and reminders to smart insulin pens which feature bolus dose calculators and more advanced decisionsupport such as dose titration and coaching features (172). A full review of insulin dosing apps is beyond the scope of this chapter.

 

Bolus calculators are known to substantially improve dosing accuracy and glycemic control in outpatients with type 1 diabetes (173,174,175). Bolus calculators might be particularly helpful for patients with poor numeracy. A number of stand-alone smart-phone apps for bolus insulin calculation have been developed but safety and efficacy remain a concern (176,177). Though algorithms typically incorporate the current glucose level, active insulin time, and carbohydrate intake, some do not account for activity or illness. Applications that improve the accuracy of carbohydrate counting, which is a major source of error (regardless of educational level), are desirable (178). Reports from connected pens provide insight into missed or altered insulin doses and when integrated with CGM data can alsofacilitate the evaluation of timing of boluses.

 

Likewise, basal insulin calculators have been developed to recommend ongoing adjustments in therapy, either fortitration or for mealtime insulin calculations. Unfortunately, efficacy and safety studies are not currently available for most apps. Most basal insulin titration apps account only for fasting glucose measures and not overnight trends.

 

Although there are a plethora of apps available, the ultimate choice should be individualized to the needs of thepatient. Those patients only needing a resource that assists with carbohydrate counting can be referred to common apps like MyFitnessPal or Calorie King. For glucose monitoring, apps that require manual entry of data should beminimized as they are not likely to be utilized long-term. Universal platforms that can download multiple devices can increase clinic efficiency. Where possible, patients should be invited to directly link with their clinic. This is particularly useful for telehealth visits. Smart insulin pens provide assistance with insulin dosing and can also be downloaded using some universal platforms.

 

Integration within the Electronic Health Record (EHR)

 

The major limitation of patient generated data is that it does not integrate within the EHR in a meaningful way. Someopportunities exist with the integration of Apple Health Kit and Samsung S-Health which can transmit data from a variety of apps but this process requires multiple steps and can be cumbersome (179,180). Recently, a consensus report from the Integration of Continuous Glucose Monitoring Data into the Electronic Health Record (iCoDE) project was published, setting standards for integration of CGM data within the EHR (181). Under these standards, data would be accessed by placing an order in the EHR. This would generate a notice to the patient via email or electronic message to obtain consent for sharing data. Once approved, standardized report is uploaded to the EHR. Importantly,none of these mobile health tools replace frequent patient contact and feedback (182).

 

BIOMARKERS OF GLYCEMIC CONTROL

 

Hemoglobin A1c (A1C)

 

A1C is the best biomarker indicator of glycemic control over the past 2-3 months due to strong data predicting complications (1,2). In addition, the American Diabetes Association has recommended its use for the diagnosis of diabetes (1).

 

Hemoglobin A1c refers to the nonenzymatic addition of glucose to the N-terminal valine of the hemoglobin beta chain. Assays are based upon charge and structural differences between hemoglobin molecules (183,184). Therefore, variants in hemoglobin molecules may lead to analytic interferences. It should be noted that some homozygous hemoglobin variants (HbC or HbD, or sickle cell disease) also alter erythrocyte life span and therefore, even if theassay does not show analytic interference, other methods of monitoring glycemia should be utilized, as A1C will be falsely low. Individual assay interferences are available at the National Glycohemohemoglobin Standardization Program website: www.ngsp.org (185). Several commercial home monitoring kits are also available (186). The two reference methods used to standardize A1c levels are 1) HPLC and electrospray ionization mass spectrometry or 2) a two- dimensional approach using HPLC and capillary electrophoresis with UV-detection (187). A brief summary of assay methods is described below.

 

  • HPLC methods utilize the fact that glycated hemoglobin has a lower isoelectric point and migrates faster than otherhemoglobin As such it has variable interference with hemoglobinopathies that alter the charge of the molecule (such as HbF and carbamylated Hb), but these may be revealed through individual inspection of the chromatograms.
  • Boronate affinity methods are based upon glucose binding to m-aminophenylboronic acid and measures glycation on the N-terminal valine on the beta chain but also glycation at other sites. There is minimal interference from hemoglobinopathies but this assay is not widely available.
  • Immunoassays make use of antibody binding to glucose and N-terminal amino acids on the beta chain and therefore may be affected by hemoglobinopathies with structural changes at these sites, including HbF but notHbE, HbD, or carbamylated Some newer assays have attempted to correct for these interferences.
  • Enzymatic methods lyse whole blood, releasing glycated N-terminal valines which are detected using achromogenic reaction and are not affected by hemoglobin

 

An Organization with links to governmental regulatory agencies, the National Glycohemoglobin Standardization Program (NGSP) (<http://www.ngsp.org/news.asp >), evaluates every laboratory and home test for A1C, sets accuracy standards, and certifies which methods meet their standards (188). The trend in industry is for monitors to become increasingly more accurate and the trend in regulatory organizations is to require increasing accuracy for ongoing certification.

 

A1C is an analyte found within red blood cells, comprised of glycated Hemoglobin. The glycation gap (formerly known as the glycosylation gap) (GG), based on fructosamine measurement, and the Hemoglobin Glycation Index (HGI), based on mean blood glucose, are two indices of between-individual differences in glycated hemoglobin adjusted forglycemia. GG is the difference between the measured A1C test and the A1C test result predicted from serum fructosamine testing based on a population regression equation of A1C on fructosamine (189). and HGI is the difference between the measured A1C test and A1C results predicted from the mean blood glucose level (calculated from self-monitored blood glucose tests) based on a population regression equation of A1C tests on mean blood glucose levels (190). These two indices are consistent within an individual over time and reflect an inherent tendency for an individual’s proteins to glycate (191,192). Patients with high GG and HGI indices might have falsely high A1C test results and might also be at increased risk of basement membrane glycosylation and development of microvascular complications. Whether between-individual biological variation in Hemoglobin A1c is an independent risk factor, distinct from that attributable to mean blood glucose or fructosamine levels, for diabetic microvascular complications is controversial (193).

 

Because the A1C test is supposed to reflect the mean level of glycemia, attempts have been made to correlate thiswidely-accepted measure with empirically measured mean blood glucose levels. In 2008, the A1c-Derived Average Glucose (ADAG) study compared A1C and continuous glucose monitoring derived mean glucose and 7-point glucose profiles among 507 patients with type 1 and type 2 diabetes and without diabetes from 10 international centers to derive an estimated average glucose (eAG) from A1C levels using the following equation: eAG(mg/dl) = (28.7* A1C)-46.7 (Table 6).

 

Table 6. A1C and Estimated Average Glucose

A1C (%)

eAG (mg/dl)

eAG (mmol/l)

5

97

5.4

6

126

7.0

7

154

8.6

8

183

10.2

9

212

11.8

10

240

13.4

11

269

14.9

12

298

16.5

 

Several lines of evidence support this disconnect from a tight correlation between mean glycemia and A1C levels. First, improvements in mean glycemia may not necessarily be reflected by improvements in A1C in intensively treated patients (194). A1C does not reflect short-term changes in glucose control, and therefore can be misleading wherethere have been recent changes in the clinical condition. In addition, glucose fluctuations, compared to chronic sustained hyperglycemia, have been shown to exhibit a more specific triggering effect on oxidative stress and endothelial function (195,196). Glycemic variability cannot be assessed by a global measure of mean glycemia, such as A1C, but requires multiple individual glucose values, such as what can be obtained from continuous glucose monitoring or from seven-point- per-day (or greater) self-glucose testing. Third, A1C does not permit specific adjustments in therapy, particularly among patients requiring insulin titration. Finally, A1C reliability may be affected by several conditions that alter red blood cell lifespan and its use in these circumstances can be misleading. Acomparison of the features and limitations in glucose markers is presented in Table 7 (197,198,199).

 

Table 7. Comparison of Markers of Glycemic Control

 

Biomarker mechanism

Interval of time reflecting glucose

control

Cautions/Interferences

A1C

Hemoglobin glycation

3 months

Hemoglobinopathy (↑/↓*)

Decrease in RBC survival (hemolysis, splenomegaly, pregnancy, drugs) (↓)

Increase in RBC survival (Erythropoietin, iron replacement) (↑)

Transfusion (↓)

Fructosamine

Protein glycation

2 weeks

Conditions resulting in hypoproteinemia (severe cirrhosis, nephrotic syndrome, enteropathy) (↓)

High dose Vitamin C, severe hyperbilirubinemia/uremia/ hypertriglyceridemia (↑)

1,5-AG

Renal clearance

1 week

Chronic kidney disease (stage 4, 5) (↓)

Glucosuria (pregnancy, renal tubular disorders SGLT2 inhibitors) (↓)

Advanced cirrhosis (↓)

High soy diet (↑)

*Assay-dependent

 

Ethnic differences in A1C have also been reported (200). For example, recent data from the Type 1 Diabetes Exchange demonstrates a 0.4% higher A1C at a given mean glucose among black patients compared to whitepatients with type 1 diabetes, but no effect of race on glycated albumin or fructosamine (201). However, NHANES data do not demonstrate an effect of ethnicity on the association between A1C and retinopathy (202). Data from the ARIC study demonstrated that A1C, fructosamine, glycated albumin, and 1,5-AG were consistent with residualhyperglycemia among blacks compared to whites, and the prognostic value for incident cardiovascular disease, end stage renal disease and retinopathy were similar by race (203). It should be noted that the range of available A1C was relatively narrow in NHANES and ARIC, and further data across an expansive range is needed. In relation to CGMs, utility of A1C is further enhanced when used as a complement to glycemic data measured by CGM (10). Other biomarkers are becoming more widely used, however, A1C remains the most common biomarker. Other measures of average glycemia such as fructosamine and 1,5-anhydroglucitol are available, but their translation into average glucose levels and prognostic significance are not as clear as for A1C (1).

 

Fructosamine

 

A short to medium-term marker (reflecting the average glucose control over the past few weeks) may be useful for determining control over a period of days to weeks since A1C does not reflect recent changes in glucose control. Alternate markers may also be useful in patients with discrepant A1C and self-monitored blood glucose readings as well as patients with other hematologic conditions known to affect A1C. Fructosamine is a term that refers to a family of glycated serum proteins and this family is comprised primarily of albumin and to a lesser extent, globulins, and to an even lesser extent, other circulating serum proteins. No product exists for home use that measures serum fructosamine. A home blood fructosamine monitor, Duet Glucose Control System, was marketed in the early 2000′sand then withdrawn from the market. No subsequent home fructosamine test has been available since then. Randomized controlled trials have reported inconsistent effects of frequent monitoring on A1C lowering, possibly due to differences in execution of therapeutic interventions (204,205). Serial monitoring of short-term markers may also facilitate timely elective surgery in patients whose procedure is delayed due to an elevated A1C. In a recent study, fructosamine was a better predictor of post-operative complications in patients undergoing primary total joint arthroplasty (206).

 

GLYCATED ALBUMIN

 

The largest constituent of fructosamine is glycated albumin. Several investigators and companies are developing portable assays for glycated albumin to assess overall control during periods of rapidly changing glucose levels. In these situations, an A1C test may change too slowly to capture a sudden increase or decrease in mean glycemia. The components of the necessary technology appear to be in place to build a commercial instrument for home testing of glycated albumin. However, there is no randomized controlled trial showing that the measurement of glycated albumin improves outcomes. In the ARIC study, fructosamine, glycated albumin, and 1,5-AG were associated with incident diabetes, even after adjustment for baseline A1C and fasting glucose. In the ARIC study, both fructosamine and glycated albumin predicted incident retinopathy and nephropathy, even after adjusting for A1C (207). However, in adults with severe chronic kidney disease, none of the markers, including A1C, fructosamine, or glycated albumin were very highly correlated with fasting glucose, and there did not appear to be an advantage of one marker over another (208). In addition, baseline glycated albumin and fructosamine were associated with cardiovascular outcomes over a 20-year follow-up period after adjusting for other risk factors, but the overall magnitude of associations was similar to A1C (209). In the Diabetes Control and Complications Trial (DCCT), glycated albumin had a similar association with retinopathy and nephropathy as A1C, but the combination of both markers provided even better prediction (210). Short-term markers are also of interest for use in pregnancy, where glucose levels are changing more quickly than can be reflected by A1C. Unfortunately, glycated albumin does not predict gestational diabetes more effectively than A1C or fasting glucose (211). However, other preliminary data suggests that glycated albumin may be a better predictor of pregnancy complications than A1C (212).

 

1,5-Anhyroglucitol

 

The aforementioned biomarkers for measuring glycemic control, (A1C, fructosamine, and glycated albumin) only reflect mean levels of glycemia. These measures can fail to portray hyperglycemic excursions if they are balanced by hypoglycemic excursions. Plasma 1,5- anhydroglucitol (1,5-AG) is a naturally occurring dietary monosaccharide, witha structure similar to that of glucose (Figure 5). This analyte has been proposed as a marker for postprandial hyperglycemia (213). An automated laboratory grade assay named Glycomark is approved in the U.S. for measuring 1,5-AG as a short-term marker for glycemic control. A similar laboratory assay has been used in Japan. During normoglycemia, 1,5-AG is maintained at constant steady-state levels because of a large body pool compared with the amount of intake and because this substance is metabolically inert. Normally, 1,5-AG is filtered and completely reabsorbed by the renal tubules. During acute hyperglycemia when the blood glucose levels exceed 180 mg/dl, whichis the renal threshold for spilling glucose into the urine, serum 1,5-AG falls. This fall occurs due to competitive inhibition of renal tubular reabsorption by filtered glucose. The greater the amount of glucose in renal filtrate (due to hyperglycemia), the less 1, 5-AG is reabsorbed by the kidneys. The 1,5-AG levels respond sensitively and rapidly torises in serum glucose and a fall in the serum level of this analyte can indicate transient elevations of serum glucose occurring over as short a period as a few days. Measurement of 1,5-AG can be useful in assessing the prior 1-2 weeks for: 1) the degree of postprandial hyperglycemia; and 2) the mean short-term level of glycemia. This assay might prove useful in assessing the extent of glycemic variability that is present in an individual with a close-to-normal A1C level, but who is suspected to be alternating between frequent periods of hyperglycemia and hypoglycemia. In such a patient, the 1,5-AG level would be low, which would indicate frequent periods of hyperglycemia, whereas in a patient with little glycemic variability, the 1,5-AG levels would not be particularly depressed because of a lack of frequent hyperglycemic periods. In the ARIC study, 1,5-AG was associated with severe hypoglycemia after adjustmentfor other variables, an observation which is consistent with the role of 1,5-AG in reflecting glycemic variability, a known risk factor for hypoglycemia (214).

 

Longitudinal data from the ARIC study showed that 1,5-AG was associated with ESRD over a 19-year follow-up period, but the relationship was no longer significant after adjusting for glucose control with other markers (215). Among participants with diabetes and A1C <7%, each 5 mcg/mL decrease in 1,5-AG was associated with an increase in dementia risk by 16%, and at A1C >7%, there was also a significant association over a median 21-year follow-up period (216). There was also an association of 1,5-AG and cardiovascular outcomes in ARIC, which persisted, thoughwere attenuated after adjusting for A1C (217). Therefore, it is not yet clear whether 1,5-AG, as a measure of glucoseexcursions, provides incremental value beyond A1C for predicting long-term complications.

 

Figure 5. Structure of glucose (left) and 1,5-anhydroglucitol (right).

 

CONCLUSIONS

 

Many new types of technology are increasingly being developed and applied to fight diabetes and its complications. New technologies will improve the lives of people with diabetes by measuring glucose and other biomarkers of glycemic control and linking glucose levels with insulin delivery to improve the lives of people with diabetes.

 

REFERENCES

  1. ElSayed NA, Aleppo G, Aroda VR, et , American Diabetes Association. 7. Diabetes technology: Standards of Care in Diabetes—2023. Diabetes Care 2023;46(Suppl. 1): S111–S127.
  2. Grunberger G, Sherr J, Allende M, Blevins T, Bode B, Handelsman Y, Hellman R, Lajara R, Roberts VL, Rodbard D, Stec C, Unger J. American Association of Clinical Endocrinology Clinical Practice Guideline: The Use of Advanced Technology in the Management of Persons With Diabetes Mellitus. Endocr Pract. 2021Jun;27(6):505-537. doi: 10.1016/j.eprac.2021.04.008.
  3. Peters AL, Ahmann AJ, Battelino T, Evert A, Hirsch IB, Murad MH, Winter WE, Wolpert DiabetesTechnology-Continuous Subcutaneous Insulin Infusion Therapy and Continuous Glucose Monitoring in Adults: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016;101(11):3922-3937.
  4. Gross TM, Bode BW, Einhorn D, Kayne DM, Reed JH, White NH, Mastrototaro JJ: Performance evaluation ofthe MiniMed continuous glucose monitoring system during patient home use. Diabetes Technol Ther. 2000;2:49-56.
  5. Di Molfetta S, Caruso I, Cignarelli A, Natalicchio A, Perrini S, Laviola L, Giorgino F. Professional continuous glucose monitoring in patients with diabetes mellitus: A systematic review and meta-analysis. Diabetes Obes 2023;25(5):1301-1310.
  6. Bailey T, Bode BW, Christiansen MP, Klaff LJ, Alva The Performance and Usability of a Factory-Calibrated Flash Glucose Monitoring System. Diabetes Technol Ther. 2015;17(11):787-94.
  7. Ólafsdóttir AF, Attvall S, Sandgren U, Dahlqvist S, Pivodic A, Skrtic S, Theodorsson E, Lind A Clinical Trialof the Accuracy and Treatment Experience of the Flash Glucose Monitor FreeStyle Libre in Adults with Type 1 Diabetes. Diabetes Technol Ther. 2017;19(3):164-172.
  8. Miele A, Weiland K, Dungan Clinical outcomes associated with referral-based continuous glucosemonitoring using a central standardized interpretation strategy. Diabetes Technol Ther. 2012;14(9):765-71.
  9. Bergenstal RM, Ahmann AJ, Bailey T, Beck RW, Bissen J, Buckingham B, Deeb L, Dolin RH, Garg SK, Goland R, Hirsch IB, Klonoff DC, Kruger DF, Matfin G, Mazze RS, Olson BA, Parkin C, Peters A, Powers MA,Rodriguez H, Southerland P, Strock ES, Tamborlane W, Wesley DM. Recommendations for standardizing glucose reporting and analysis to optimize clinical decision making in diabetes: the Ambulatory Glucose Profile (AGP). Diabetes Technol Ther. 2013;15(3):198-211.
  10. Battelino T, Danne T, Bergenstal RM, et al. Clinical Targets for Continuous Glucose Monitoring DataInterpretation: Recommendations From the International Consensus on Time in Range. Diabetes Care. 2019;42(8):1593-1603.
  11. Rodbard D. Quality of Glycemic Control: Assessment Using Relationships Between Metrics for Safety and Diabetes Technol Ther. 2021 Oct;23(10):692-704. doi: 10.1089/dia.2021.0115.
  12. Klonoff DC, Wang J, Rodbard D, Kohn MA, Li C, Liepmann D, Kerr D, Ahn D, Peters AL, Umpierrez GE, Seley JJ, Xu NY, Nguyen KT, Simonson G, Agus MSD, Al-Sofiani ME, Armaiz-Pena G, Bailey TS, Basu A, BattelinoT, Bekele SY, Benhamou PY, Bequette BW, Blevins T, Breton MD, Castle JR, Chase JG, Chen KY, ChoudharyP, Clements MA, Close KL, Cook CB, Danne T, Doyle FJ 3rd, Drincic A, Dungan KM, Edelman SV, Ejskjaer N, Espinoza JC, Fleming GA, Forlenza GP, Freckmann G, Galindo RJ, Gomez AM, Gutow HA, Heinemann L, Hirsch IB, Hoang TD, Hovorka R, Jendle JH, Ji L, Joshi SR, Joubert M, Koliwad SK, Lal RA, Lansang MC, Lee WA, Leelarathna L, Leiter LA, Lind M, Litchman ML, Mader JK, Mahoney KM, Mankovsky B, Masharani U, Mathioudakis NN, Mayorov A, Messler J, Miller JD, Mohan V, Nichols JH, Nørgaard K, O'Neal DN, Pasquel FJ, Philis- Tsimikas A, Pieber T, Phillip M, Polonsky WH, Pop-Busui R, Rayman G, Rhee EJ, Russell SJ, Shah VN, Sherr JL, Sode K, Spanakis EK, Wake DJ, Waki K, Wallia A, Weinberg ME, Wolpert H, Wright EE, Zilbermint M, Kovatchev B. A Glycemia Risk Index (GRI) of Hypoglycemia and Hyperglycemia for Continuous Glucose Monitoring Validated by Clinician Ratings. J Diabetes Sci Technol. 2022 Mar 29:19322968221085273. doi: 1177/19322968221085273.
  13. Hásková A, Radovnická L, Petruželková L, Parkin CG, Grunberger G, Horová E, Navrátilová V, Kádě O,Matoulek M, Prázný M, Šoupal Real-time CGM Is Superior to Flash Glucose Monitoring for Glucose Control in Type 1 Diabetes: The CORRIDA Randomized Controlled Trial. Diabetes Care. 2020 Nov;43(11):2744-2750. doi: 10.2337/dc20-0112.
  14. Visser MM, Charleer S, Fieuws S, De Block C, Hilbrands R, Van Huffel L, Maes T, Vanhaverbeke G, Dirinck E, Myngheer N, Vercammen C, Nobels F, Keymeulen B, Mathieu C, Gillard P. Comparing real-time andintermittently scanned continuous glucose monitoring in adults with type 1 diabetes (ALERTT1): a 6-month,prospective, multicentre, randomised controlled trial. Lancet. 2021 Jun 12;397(10291):2275-2283. doi: 1016/S0140-6736(21)00789-3.
  15. Visser MM, Charleer S, Fieuws S, De Block C, Hilbrands R, Van Huffel L, Maes T, Vanhaverbeke G, Dirinck E, Myngheer N, Vercammen C, Nobels F, Keymeulen B, Mathieu C, Gillard P. Effect of switching from intermittently scanned to real-time continuous glucose monitoring in adults with type 1 diabetes: 24-monthresults from the randomised ALERTT1 trial. Lancet Diabetes Endocrinol. 2023;11(2):96-108.
  16. Alva S, Bailey T, Brazg R, Budiman ES, Castorino K, Christiansen MP, Forlenza G, Kipnes M, Liljenquist DR, Liu H. Accuracy of a 14-Day Factory-Calibrated Continuous Glucose Monitoring System With AdvancedAlgorithm in Pediatric and Adult Population With Diabetes. J Diabetes Sci Technol. 2022 Jan;16(1):70-77. doi: 1177/1932296820958754.
  17. Garg SK, Kipnes M, Castorino K, Bailey TS, Akturk HK, Welsh JB, Christiansen MP, Balo AK, Brown SA, Reid JL, Beck SE. Accuracy and Safety of Dexcom G7 Continuous Glucose Monitoring in Adults with Diabetes. Diabetes Technol 2022 Jun;24(6):373-380. doi: 10.1089/dia.2022.0011.
  18. Luijf YM, Mader JK, Doll W, Pieber T, Farret A, Place J, Renard E, Bruttomesso D, Filippi A, Avogaro A,Arnolds S, Benesch C, Heinemann L, DeVries AP@home consortium. Accuracy and reliability of continuous glucose monitoring systems: a head-to-head comparison. Diabetes Technol Ther. 2013;15(8):722-7
  19. Deiss D, Bolinder J, Riveline JP, Battelino T, Bosi E, Tubiana-Rufi N, Kerr D, Phillip Improved glycemiccontrol in poorly controlled patients with type 1 diabetes using real- time continuous glucose monitoring. Diabetes Care. 2006; 29:2730-2732.
  20. Juvenile Diabetes Research Foundation Continuous Glucose Monitoring Study Group, Tamborlane WV, Beck RW, Bode BW, Buckingham B, Chase HP, Clemons R, Fiallo- Scharer R, Fox LA, Gilliam LK, Hirsch IB, Huang ES, Kollman C, Kowalski AJ, Laffel L, Lawrence JM, Lee J, Mauras N, O'Grady M, Ruedy KJ, Tansey M,Tsalikian E, Weinzimer S, Wilson DM, Wolpert H, Wysocki T, Xing D. Continuous glucose monitoring and intensive treatment of type 1 diabetes. N Engl J Med. 2008;359(14):1464-76.
  21. Battelino T, Phillip M, Bratina N, Nimri R, Oskarsson P, Bolinder J. Effect of continuous glucose monitoring onhypoglycemia in type 1 Diabetes Care. 2011;34(4):795- 800.
  22. Bolinder J, Antuna R, Geelhoed-Duijvestijn P, Kröger J, Weitgasser R. Novel glucose- sensing technology andhypoglycaemia in type 1 diabetes: a multicentre, non-masked, randomised controlled trial. Lancet. 2016;388(10057):2254-2263.
  23. Reddy M, Jugnee N, El Laboudi A, Spanudakis E, Anantharaja S, Oliver A randomized controlled pilot study of continuous glucose monitoring and flash glucose monitoring in people with Type 1 diabetes and impaired awareness of hypoglycaemia. Diabet Med. 2017;35(4): 483-90.
  24. Laffel LM, Kanapka LG, Beck RW, Bergamo K, Clements MA, Criego A, DeSalvo DJ, Goland R, Hood K, Liljenquist D, Messer LH, Monzavi R, Mouse TJ, Prahalad P, Sherr J, Simmons JH, Wadwa RP, Weinstock RS,Willi SM, Miller KM; CGM Intervention in Teens and Young Adults with T1D (CITY) Study Group; CDE10. Effect of Continuous Glucose Monitoring on Glycemic Control in Adolescents and Young Adults With Type 1Diabetes: A Randomized Clinical Trial. JAMA. 2020 Jun 16;323(23):2388-2396. doi: 1001/jama.2020.6940.
  25. Beck RW, Riddlesworth T, Ruedy K, Ahmann A, Bergenstal R, Haller S, Kollman C, Kruger D, McGill JB, Polonsky W, Toschi E, Wolpert H, Price D; DIAMOND Study 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):371-378.
  26. Pratley RE, Kanapka LG, Rickels MR, Ahmann A, Aleppo G, Beck R, Bhargava A, Bode BW, Carlson A, Chaytor NS, Fox DS, Goland R, Hirsch IB, Kruger D, Kudva YC, Levy C, McGill JB, Peters A, Philipson L, Philis-Tsimikas A, Pop-Busui R, Shah VN, Thompson M, Vendrame F, Verdejo A, Weinstock RS, Young L, Miller KM; Wireless Innovation for Seniors With Diabetes Mellitus (WISDM) Study Group. Effect of Continuous Glucose Monitoring on Hypoglycemia in Older Adults With Type 1 Diabetes: A Randomized Clinical Trial. JAMA. 2020 Jun 16;323(23):2397-2406. doi: 10.1001/jama.2020.6928.
  27. Miller KM, Kanapka LG, Rickels MR, Ahmann AJ, Aleppo G, Ang L, Bhargava A, Bode BW, Carlson A,Chaytor NS, Gannon G, Goland R, Hirsch IB, Kiblinger L, Kruger D, Kudva YC, Levy CJ, McGill JB, O'Malley G, Peters AL, Philipson LH, Philis-Tsimikas A, Pop- Busui R, Salam M, Shah VN, Thompson MJ, Vendrame F, Verdejo A, Weinstock RS, Young L, Pratley R. Benefit of Continuous Glucose Monitoring in Reducing Hypoglycemia Is Sustained Through 12 Months of Use Among Older Adults with Type 1 Diabetes. Diabetes Technol Ther. 2022 Jun;24(6):424-434. doi: 10.1089/dia.2021.0503.
  28. Bergenstal RM, Tamborlane WV, Ahmann A, Buse JB, Dailey G, Davis SN, Joyce C, Peoples T, Perkins BA, Welsh JB, Willi SM, Wood MA; STAR 3 Study Group. Effectiveness of sensor-augmented insulin-pump therapyin type 1 N Engl J Med. 2010;363(4):311-20.
  29. Lind M, Polonsky W, Hirsch IB, Heise T, Bolinder J, Dahlqvist S, Schwarz E, Ólafsdóttir AF, Frid A, Wedel H, Ahlén E, Nyström T, Hellman J. Continuous Glucose Monitoring vs Conventional Therapy for Glycemic Controlin Adults With Type 1 Diabetes Treated With Multiple Daily Insulin Injections: The GOLD Randomized Clinical Trial. JAMA. 2017;317(4):379-387.
  30. Leelarathna L, Evans ML, Neupane S, Rayman G, Lumley S, Cranston I, Narendran P, Barnard-Kelly K,Sutton CJ, Elliott RA, Taxiarchi VP, Gkountouras G, Burns M, Mubita W, Kanumilli N, Camm M, Thabit H,Wilmot EG; FLASH-UK Trial Study Group. Intermittently Scanned Continuous Glucose Monitoring for Type 1 Diabetes. N Engl J Med. 2022 Oct 20;387(16):1477-1487. doi: 10.1056/NEJMoa2205650.
  31. Yan J, Zhou Y, Zheng X, Zheng M, Lu J, Luo S, Yang D, Deng H, Xu W, Bi Y, Bao W, Weng J. Effects of intermittently scanned continuous glucose monitoring in adult type 1 diabetes patients with suboptimalglycaemic control: A multi-centre randomized controlled trial. Diabetes Metab Res Rev. 2023;39(4):e3614.
  32. Langendam M, Luijf YM, Hooft L, Devries JH, Mudde AH, Scholten RJ. Continuous glucose monitoringsystems for type 1 diabetes Cochrane Database Syst Rev. 2012;1:CD008101.
  33. Yeh HC, Brown TT, Maruthur N, Ranasinghe P, Berger Z, Suh YD, Wilson LM, Haberl EB, Brick J, Bass EB, Golden SH. 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):336-47.
  34. Pickup The evidence base for diabetes technology: appropriate and inappropriate meta-analysis. J Diabetes Sci Technol. 2013;7(6):1567-74.
  35. Pickup JC, Freeman SC, Sutton Glycaemic control in type 1 diabetes during real time continuous glucose monitoring compared with self monitoring of blood glucose: meta- analysis of randomised controlled trials using individual patient data. BMJ. 2011;343:d3805.
  36. Teo E, Hassan N, Tam W, Koh S. Effectiveness of continuous glucose monitoring in maintaining glycaemiccontrol among people with type 1 diabetes mellitus: a systematic review of randomised controlled trials and meta-analysis. Diabetologia. 2022 Apr;65(4):604-619. doi: 10.1007/s00125-021-05648-4.
  37. Elbalshy M, Haszard J, Smith H, Kuroko S, Galland B, Oliver N, Shah V, de Bock MI, Wheeler Effect ofdivergent continuous glucose monitoring technologies on glycaemic control in type 1 diabetes mellitus: A systematic review and meta-analysis of randomised controlled trials. Diabet Med. 2022;39(8):e14854.
  38. Little SA, Leelarathna L, Walkinshaw E, Tan HK, Chapple O, Lubina-Solomon A, Chadwick TJ, Barendse S, Stocken DD, Brennand C, Marshall SM, Wood R, Speight J, Kerr D, Flanagan D, Heller SR, Evans ML, Shaw JA. Recovery of hypoglycemia awareness in long-standing type 1 diabetes: a multicenter 2 × 2 factorial randomized controlled trial comparing insulin pump with multiple daily injections and continuous with conventional glucose self-monitoring (HypoCOMPaSS). Diabetes 2014;37(8):2114- 22.
  39. van Beers CA, DeVries JH, Kleijer SJ, Smits MM, Geelhoed-Duijvestijn PH, Kramer MH, Diamant M, Snoek FJ, Serné EH. Continuous glucose monitoring for patients with type 1 diabetes and impaired awareness of hypoglycaemia (IN CONTROL): a randomised, open- label, crossover trial. Lancet Diabetes Endocrinol.2016;4(11):893-902.
  40. Serné EH, van den Berg IK, Racca C, van Raalte DH, Kramer MHH, de Wit M, Snoek Improved Effectiveness of Immediate Continuous Glucose Monitoring in Hypoglycemia- Prone People with Type 1 Diabetes Compared with Hypoglycemia-Focused Psychoeducation Following a Previous Structured Education: A Randomized Controlled Trial. Diabetes Technol Ther. 2023;25(1):50-61.
  41. Rubin RR, Peyrot M; STAR 3 Study Group. Health-related quality of life and treatment satisfaction in theSensor-Augmented Pump Therapy for A1C Reduction 3 (STAR 3) Diabetes Technol Ther. 2012;14(2):143-51.
  42. Polonsky WH, Hessler D, Ruedy KJ, Beck RW; DIAMOND Study Group. The Impact of Continuous Glucose Monitoring on Markers of Quality of Life in Adults With Type 1 Diabetes: Further Findings From the DIAMONDRandomized Clinical Diabetes Care. 2017;40(6):736-741.
  43. Choe HJ, Rhee EJ, Won JC, Park KS, Lee WY, Cho YM. Effects of Patient-Driven Lifestyle Modification Using Intermittently Scanned Continuous Glucose Monitoring in Patients With Type 2 Diabetes: Results From theRandomized Open-label PDF Diabetes Care. 2022 Oct 1;45(10):2224-2230.
  44. Moon SJ, Kim KS, Lee WJ, Lee MY, Vigersky R, Park Efficacy of intermittent short- term use of a real-time continuous glucose monitoring system in non-insulin-treated patients with type 2 diabetes: A randomizedcontrolled trial. Diabetes Obes Metab. 2023 Jan;25(1):110-120.
  45. Vigersky RA, Fonda SJ, Chellappa M, Walker MS, Ehrhardt Short- and long-term effects of real-time continuous glucose monitoring in patients with type 2 diabetes. Diabetes Care. 2012 Jan;35(1):32-8.
  46. Beck RW, Riddlesworth TD, Ruedy K, Ahmann A, Haller S, Kruger D, McGill JB, Polonsky W, Price D, Aronoff S, Aronson R, Toschi E, Kollman C, Bergenstal R; DIAMOND Study Group. Continuous Glucose Monitoring Versus Usual Care in Patients With Type 2 Diabetes Receiving Multiple Daily Insulin Injections: A Randomized Trial. Ann Intern 2017 Sep 19;167(6):365-374.
  47. Martens T, Beck RW, Bailey R, Ruedy KJ, Calhoun P, Peters AL, Pop-Busui R, Philis- Tsimikas A, Bao S,Umpierrez G, Davis G, Kruger D, Bhargava A, Young L, McGill JB, Aleppo G, Nguyen QT, Orozco I, Biggs W, Lucas KJ, Polonsky WH, Buse JB, Price D, Bergenstal RM; MOBILE Study Group. Effect of Continuous Glucose Monitoring on Glycemic Control in Patients With Type 2 Diabetes Treated With Basal Insulin: A Randomized Clinical Trial. JAMA. 2021 Jun 8;325(22):2262-2272. doi: 1001/jama.2021.7444.
  48. Aleppo G, Beck RW, Bailey R, Ruedy KJ, Calhoun P, Peters AL, Pop-Busui R, Philis- Tsimikas A, Bao S, Umpierrez G, Davis G, Kruger D, Bhargava A, Young L, Buse JB, McGill JB, Martens T, Nguyen QT, Orozco I, Biggs W, Lucas KJ, Polonsky WH, Price D, Bergenstal RM; MOBILE Study Group; Type 2 Diabetes BasalInsulin Users: The Mobile Study (MOBILE) Study Group:. The Effect of Discontinuing Continuous Glucose Monitoring in Adults With Type 2 Diabetes Treated With Basal Insulin. Diabetes Care. 2021 Dec;44(12):2729-2737. doi: 10.2337/dc21-1304.
  49. Bao S, Bailey R, Calhoun P, Beck Effectiveness of Continuous Glucose Monitoring in Older Adults with Type 2 Diabetes Treated with Basal Insulin. Diabetes Technol Ther. 2022 May;24(5):299-306. doi: 10.1089/dia.2021.0494.
  50. Yaron M, Roitman E, Aharon-Hananel G, Landau Z, Ganz T, Yanuv I, Rozenberg A, Karp M, Ish-Shalom M, Singer J, Wainstein J, Raz I. Effect of Flash Glucose Monitoring Technology on Glycemic Control and Treatment Satisfaction in Patients With Type 2 Diabetes. Diabetes Care. 2019 Jul;42(7):1178-1184. doi: 10.2337/dc18-0166.
  51. Ajjan RA, Heller SR, Everett CC, Vargas-Palacios A, Higham R, Sharples L, Gorog DA, Rogers A, Reynolds C,Fernandez C, Rodrigues P, Sathyapalan T, Storey RF, Stocken Multicenter Randomized Trial of Intermittently Scanned Continuous Glucose Monitoring Versus Self-Monitoring of Blood Glucose in Individuals With Type 2 Diabetes and Recent-Onset Acute Myocardial Infarction: Results of the LIBERATES Trial.Diabetes Care. 2023 Feb 1;46(2):441-449.
  52. Aronson R, Brown RE, Chu L, Bajaj HS, Khandwala H, Abitbol A, Malakieh N, Goldenberg IMpact of flash glucose Monitoring in pEople with type 2 Diabetes Inadequately controlled with non-insulin AntihyperglycaemicThErapy (IMMEDIATE): A randomized controlled trial. Diabetes Obes Metab. 2023;25(4):1024-1031.
  53. DeSalvo DJ, Miller KM, Hermann JM, Maahs DM, Hofer SE, Clements MA, Lilienthal E, Sherr JL, Tauschmann M, Holl RW; T1D Exchange and DPV Registries. Continuous glucose monitoring and glycemic control amongyouth with type 1 diabetes: International comparison from the T1D Exchange and DPV Initiative. Pediatr Diabetes. 2018;19(7):1271-1275.
  54. Deshmukh H, Wilmot E, Pieri B, Choudhary P, Shah N, Gregory R, Kilvert A, Lumb A, Christian P, Barnes D, Patmore J, Walton C, Ryder REJ, Sathyapalan T. Time in range following flash glucose monitoring:Relationship with glycaemic control, diabetes-related distress and resource utilisation in the Association of British Clinical Diabetologists national audit. Diabet Med. 2022 Nov;39(11):e14942. doi: 10.1111/dme.14942.
  55. Nathanson D, Svensson AM, Miftaraj M, Franzén S, Bolinder J, Eeg-Olofsson Effect of flash glucose monitoring in adults with type 1 diabetes: a nationwide, longitudinal observational study of 14,372 flash users compared with 7691 glucose sensor naive controls. Diabetologia. 2021 Jul;64(7):1595-1603. doi: 10.1007/s00125-021-05437-z.
  56. Roussel R, Riveline JP, Vicaut E, de Pouvourville G, Detournay B, Emery C, Levrat- Guillen F, Guerci Important Drop Rate of Acute Diabetes Complications in People With Type 1 or Type 2 Diabetes After Initiation of Flash Glucose Monitoring in France: The RELIEF Study. Diabetes Care. 2021 Apr 20:dc201690. doi: 10.2337/dc20-1690.
  57. Riveline JP, Roussel R, Vicaut E, de Pouvourville G, Detournay B, Emery C, Levrat- Guillen F, Guerci B. Reduced Rate of Acute Diabetes Events with Flash Glucose Monitoring Is Sustained for 2 Years After Initiation: Extended Outcomes from the RELIEF Study. Diabetes Technol Ther. 2022 Sep;24(9):611-618. doi: 10.1089/dia.2022.0085.
  58. Charleer S, De Block C, Van Huffel L, Broos B, Fieuws S, Nobels F, Mathieu C, Gillard Quality of Life and Glucose Control After 1 Year of Nationwide Reimbursement of Intermittently Scanned Continuous Glucose Monitoring in Adults Living With Type 1 Diabetes (FUTURE): A Prospective Observational Real-World Cohort Study. Diabetes Care. 2020 Feb;43(2):389-397. doi: 10.2337/dc19-1610.
  59. Karter AJ, Parker MM, Moffet HH, Gilliam LK, Dlott Association of Real-time Continuous Glucose Monitoring With Glycemic Control and Acute Metabolic Events Among Patients With Insulin-Treated Diabetes. JAMA. 2021 Jun 8;325(22):2273-2284. doi: 10.1001/jama.2021.6530.
  60. Bergenstal RM, Kerr MSD, Roberts GJ, Souto D, Nabutovsky Y, Hirsch IB. Flash CGM Is Associated WithReduced Diabetes Events and Hospitalizations in Insulin-Treated Type 2 Diabetes. J Endocr Soc. 2021 Feb 2;5(4):bvab013. doi: 10.1210/jendso/bvab013. PMID: 33644623; PMCID: PMC7901259.
  61. Yeoh E, Choudhary P, Nwokolo M, Ayis S, Amiel SA. Interventions That Restore Awareness of Hypoglycemiain Adults With Type 1 Diabetes: A Systematic Review and Meta-analysis. Diabetes Care. 2015 Aug;38(8):1592-609.
  62. Danne T, Nimri R, Battelino T, Bergenstal RM, Close KL, DeVries JH, Garg S, Heinemann L, Hirsch I, Amiel SA, Beck R, Bosi E, Buckingham B, Cobelli C, Dassau E, Doyle FJ 3rd, Heller S, Hovorka R, Jia W, Jones T, Kordonouri O, Kovatchev B, Kowalski A, Laffel L, Maahs D, Murphy HR, Nørgaard K, Parkin CG, Renard E, Saboo B, Scharf M, Tamborlane WV, Weinzimer SA, Phillip International Consensus on Use of Continuous Glucose Monitoring. Diabetes Care. 2017 Dec;40(12):1631-1640.
  63. Beck RW, Bergenstal RM, Riddlesworth TD, et Validation of time in range as an outcome measure for diabetes clinical trials. Diabetes Care 2019;42:400-405.
  64. Petrie JR, Peters AL, Bergenstal RM, Holl RW, Fleming GA, Heinemann Improving the Clinical Value and Utility of CGM Systems: Issues and Recommendations: A Joint Statement of the European Association for the Study of Diabetes and the American Diabetes Association Diabetes Technology Working Group. Diabetes Care. 2017;40(12):1614-1621.
  65. Spanakis EK, Urrutia A, Galindo RJ, Vellanki P, Migdal AL, Davis G, Fayfman M, Idrees T, Pasquel FJ, Coronado WZ, Albury B, Moreno E, Singh LG, Marcano I, Lizama S, Gothong C, Munir K, Chesney C, Maguire R, Scott WH, Perez-Guzman MC, Cardona S, Peng L, Umpierrez GE. Continuous Glucose Monitoring-Guided Insulin Administration in Hospitalized Patients With Diabetes: A Randomized Clinical Trial. Diabetes Care. 2022 Oct 1;45(10):2369-2375. doi: 10.2337/dc22-0716.
  66. Fortmann AL, Spierling Bagsic SR, Talavera L, Garcia IM, Sandoval H, Hottinger A, Philis- Tsimikas A. Glucose as the Fifth Vital Sign: A Randomized Controlled Trial of Continuous Glucose Monitoring in a Non-ICU Hospital Setting. Diabetes Care. 2020 Nov;43(11):2873- 2877. doi: 10.2337/dc20-1016.
  67. Buschur EO, Faulds E, Dungan CGM in the Hospital: Is It Ready for Prime Time? Curr Diab Rep. 2022 Sep;22(9):451-460. doi: 10.1007/s11892-022-01484-x.
  68. Galindo RJ, Umpierrez GE, Rushakoff RJ, Basu A, Lohnes S, Nichols JH, Spanakis EK, Espinoza J, Palermo NE, Awadjie DG, Bak L, Buckingham B, Cook CB, Freckmann G, Heinemann L, Hovorka R, Mathioudakis N,Newman T, O'Neal DN, Rickert M, Sacks DB, Seley JJ, Wallia A, Shang T, Zhang JY, Han J, Klonoff Continuous Glucose Monitors and Automated Insulin Dosing Systems in the Hospital Consensus Guideline. J Diabetes Sci Technol. 2020 Nov;14(6):1035-1064. doi: 10.1177/1932296820954163.
  69. Foster NC, Beck RW, Miller KM, Clements MA, Rickels MR, DiMeglio LA, Maahs DM, Tamborlane WV,Bergenstal R, Smith E, Olson BA, Garg State of Type 1 Diabetes Management and Outcomes from the T1D Exchange in 2016-2018. Diabetes Technol Ther. 2019;21(2):66-72.
  70. Tanenbaum ML, Hanes SJ, Miller KM, Naranjo D, Bensen R, Hood KK. Diabetes Device Use in Adults WithType 1 Diabetes: Barriers to Uptake and Potential Intervention Diabetes Care. 2017;40(2):181-187.
  71. Odugbesan O, Addala A, Nelson G, Hopkins R, Cossen K, Schmitt J, Indyk J, Jones NY, Agarwal S, Rompicherla S, Ebekozien O. Implicit Racial-Ethnic and Insurance-Mediated Bias to Recommending Diabetes Technology: Insights from T1D Exchange Multicenter Pediatric and Adult Diabetes Provider Cohort. Diabetes Technol Ther. 2022;24(9):619-
  72. Majidi S, Ebekozien O, Noor N, et al.: Inequities in health outcomes in children and adults with type 1 diabetes: data from the T1D Exchange Quality Improvement Collaborative. Clin Diabetes 2021;39:278–283.
  73. Agarwal S, Kanapka LG, Raymond JK, et al.: Racial-ethnic inequity in young adults with type 1 diabetes. J Clin Endocrinol Metab 2020;105:e2960–e2969.
  74. Rigo RS, Levin LE, Belsito DV, Garzon MC, Gandica R, Williams KM. Cutaneous Reactions to Continuous Glucose Monitoring and Continuous Subcutaneous Insulin Infusion Devices in Type 1 Diabetes JDiabetes Sci Technol. 2021 Jul;15(4):786- 791. doi: 10.1177/1932296820918894.
  75. Messer LH, Berget C, Beatson C, Polsky S, Forlenza Preserving Skin Integrity with Chronic Device Use in Diabetes. Diabetes Technol Ther. 2018 Jun;20(S2):S254-S264. doi: 10.1089/dia.2018.0080.
  76. Nørgaard K, Scaramuzza A, Bratina N, Lalić NM, Jarosz-Chobot P, Kocsis G, Jasinskiene E, De Block C, Carrette O, Castañeda J, Cohen O; Interpret Study Group. Routine sensor-augmented pump therapy in type 1diabetes: the INTERPRET Diabetes Technol Ther. 2013;15(4):273-80.
  77. Juvenile Diabetes Research Foundation Continuous Glucose Monitoring Study Group. Effectiveness of continuous glucose monitoring in a clinical care environment: evidence from the Juvenile Diabetes ResearchFoundation continuous glucose monitoring (JDRF- CGM) trial. Diabetes Care. 2010;33(1):17-22.
  78. Cappon G, Vettoretti M, Sparacino G, Facchinetti A. Continuous Glucose Monitoring Sensors for DiabetesManagement: A Review of Technologies and Diabetes and Metabolism Journal. 2019 Aug; 43(4): 383-397. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6712232/
  79. Bay C, Kristensen PL, Pedersen-Bjergaard U, Tarnow L, Thorsteinsson B. Nocturnal continuous glucose monitoring: accuracy and reliability of hypoglycemia detection in patients with type 1 diabetes at high risk ofsevere Diabetes Technol Ther. 2013;15(5):371-7.
  80. Freckmann G, Link M, Westhoff A, Kamecke U, Pleus S, Haug C. Prediction Quality of Glucose Trend Indicators in Two Continuous Tissue Glucose Monitoring Systems. Diabetes Technol Ther. 2018 Aug;20(8):550-556.
  81. Jenkins AJ, Krishnamurthy B, Best JD, Cameron FJ, Colman PG, Hamblin PS, O'Connell MA, Rodda C, Teede H, O'Neal DN. An algorithm guiding patient responses to real-time- continuous glucose monitoring improves quality of life. Diabetes Technol Ther. 2011 Feb;13(2):105-9.
  82. Diabetes Research In Children Network (DirecNet) Study Group, Buckingham B, Xing D, Weinzimer S, Fiallo-Scharer R, Kollman C, Mauras N, Tsalikian E, Tamborlane W, Wysocki T, Ruedy K, Beck R. Use of the DirecNet Applied Treatment Algorithm (DATA) for diabetes management with a real-time continuous glucose monitor (the FreeStyle Navigator). Pediatr Diabetes. 2008;9(2):142-7.
  83. Scheiner Practical CGM: Improving Patient Outcomes Through Continuous Glucose Monitoring. 4th ed. Alexandria, VA: American Diabetes Association; 2015.
  84. Pettus J, Edelman SV. Recommendations for using real-time continuous glucose monitoring (rtCGM) data forinsulin adjustments in type 1 J Diabetes Sci Technol. 2017;11(1):138–147.
  85. Klonoff DC, Kerr D. A simplified approach using rate of change arrows to adjust insulin with real-timecontinuous glucose J Diabetes Sci Technol. 2017;11(6):1063– 1069.
  86. Aleppo G, Laffel LM, Ahmann AJ, Hirsch IB, Kruger DF, Peters A, Weinstock RS, Harris DR. A PracticalApproach to Using Trend Arrows on the Dexcom G5 CGM System for the Management of Adults with Diabetes. Journal of the Endocrine Society 2017;1(12):1445-1460.
  87. Ajjan RA, Cummings MH, Jennigs P, Leelarathna L, Rayman G, Wilmot E. Optimising use of rate-of-changetrend arrows for insulin dosing decisions using the FreeStyle Libre Flash Glucose Monitoring System. Diabetes and Vascular Disease Research. 2018;16(1):3-12.
  88. Ziegler, R.; von Sengbusch, S.; Kröger, J.; Schubert, O.; Werkmeister, P.; Deiss, D.; Siegmund, TherapyAdjustments Based on Trend Arrows Using Continuous Glucose Monitoring Systems. J. Diabetes Sci. Technol. 2019, 13, 763–773.
  89. Parise M, Di Molfetta S, Graziano RT, Fiorentino R, Cutruzzolà A, Gnasso A, Irace C. A Head-to-HeadComparison of Two Algorithms for Adjusting Mealtime Insulin Doses Based on CGM Trend Arrows in AdultPatients with Type 1 Diabetes: Results from an Exploratory Study. Int J Environ Res Public Health. 2023;20(5):3945.
  90. Gonzales WV, Mobashsher AT, Abbosh A. The Progress of Glucose Monitoring—A Review of Invasive toMinimally and Non-Invasive Techniques, Devices, and Sensors. 2019;19(4):800.
  91. FreeStyle Libre User Manual. https://freestyleserver.com/Payloads/IFU/2017_sep/ART38553-001_rev-C-Web.pdf. Accessed June 29, 2023.
  92. https://www.freestylelibre.us/system-overview/freestyle-libre-html?msclkid=4cedd02409fe16f5d98e8f09162393b1. Site accessed June 29, 2023.
  93. Aronson R, Abitbol A, Tweden First assessment of the performance of an implantable continuous glucose monitoring system through 180 days in a primarily adolescent population with type 1 diabetes. Diabetes Obes Metab. 2019;21(7):1689-1694.
  94. Kropff J, Choudhary P, Neupane S, Barnard K, Bain SC, Kapitza C, Forst T, Link M, Dehennis A, DeVries Accuracy and Longevity of an Implantable Continuous Glucose Sensor in the PRECISE Study: A 180-Day, Prospective, Multicenter, Pivotal Trial. Diabetes Care. 2017 Jan;40(1):63-68.
  95. Lorenz C, Sandoval W, Mortellaro M. Interference Assessment of Various Endogenous and ExogenousSubstances on the Performance of the Eversense Long-Term Implantable Continuous Glucose Monitoring System. Diabetes Technol Ther. 2018;20(5):344-352.
  96. Garg SK, Liljenquist D, Bode B, Christiansen MP, Bailey TS, Brazg RL, Denham DS, Chang AR, Akturk HK, Dehennis A, Tweden KS, Kaufman FR. Evaluation of Accuracy and Safety of the Next-Generation Up to 180-Day Long-Term Implantable Eversense Continuous Glucose Monitoring System: The PROMISE Diabetes Technol Ther. 2022 Feb;24(2):84-92. doi: 10.1089/dia.2021.0182.
  97. Deiss D, Szadkowska A, Gordon D, Mallipedhi A, Schütz-Fuhrmann I, Aguilera E, Ringsell C, De Block C, Irace C. Clinical Practice Recommendations on the Routine Use of Eversense, the First Long-Term Implantable Continuous Glucose Monitoring System. Diabetes Technol Ther. 2019;21(5):254-264.
  98. Šoupal J, Petruželková L, Flekač M, Pelcl T, Matoulek M, Daňková M, Škrha J, Svačina Š, Prázný M. Comparison of Different Treatment Modalities for Type 1 Diabetes, Including Sensor-Augmented Insulin Regimens, in 52 Weeks of Follow-Up: A COMISAIR Study. Diabetes Technol Ther. 2016 Sep;18(9):532-8.
  99. Boughton CK, Hovorka Is an artificial pancreas (closed-loop system) for Type 1 diabetes effective? Diabet Med. 2019;36(3):279-286.
  100. Braune K, Hussain S, Lal The First Regulatory Clearance of an Open-Source Automated Insulin Delivery Algorithm. J Diabetes Sci Technol. 023:19322968231164166.
  101. Melmer A, Züger T, Lewis DM, Leibrand S, Stettler C, Laimer Glycaemic Control in Individuals with Type 1 Diabetes using an artificial open source pancreas system (OpenAPS). Diabetes Obesity and Metabolism. 2019; 21(10):2333-2337.
  102. https://www.fda.gov/news-events/press-announcements/fda-warns-against-use-unauthorized-devices-diabetes-management. Accessed June 29,
  103. Jennifer Sherr, Lutz Heinemann, G. Alexander Fleming, Richard M. Bergenstal, Daniela Bruttomesso, Hélène Hanaire, Reinhard W. Holl, John R. Petrie, Anne L. Peters, Mark Evans; Automated Insulin Delivery: Benefits, Challenges, and Recommendations. A Consensus Report of the Joint Diabetes Technology Working Group of the European Association for the Study of Diabetes and the American Diabetes Association. Diabetes Care 1 December 2022; 45 (12): 3058–3074. https://doi.org/10.2337/dci22-0018
  104. Lewis History and Perspective on DIY Closed Looping. J Diabetes Sci Technol. 2019;13(4):790-793.
  105. Hernando ME, Garcia-Sáez G, Gómez EJ, Pérez-Gandia C, Rodriguez-Herrero A. Automated InsulinDelivery: The Artificial Pancreas Technical American Journal of Therapeutics. 2019; 1075-2765
  106. Bailey TS, Ahmann A, Brazg R, Christiansen M, Garg S, Watkins E, Welsh JB, Lee SW. Accuracy andacceptability of the 6-day Enlite continuous subcutaneous glucose Diabetes Technol Ther. 2014;16(5):277-83.
  107. Bergenstal RM, Klonoff DC, Garg SK, Bode BW, Meredith M, Slover RH, Ahmann AJ, Welsh JB, Lee SW, Kaufman FR; ASPIRE In-Home Study Group. Threshold-based insulin-pump interruption for reduction of N Engl J Med. 2013;369(3):224- 32.
  108. Ly TT, Nicholas JA, Retterath A, Lim EM, Davis EA, Jones TW. Effect of sensor- augmented insulin pump therapy and automated insulin suspension vs standard insulin pump therapy on hypoglycemia in patientswith type 1 diabetes: a randomized clinical JAMA. 2013;310(12):1240-7.
  109. Abraham MB, Nicholas JA, Smith GJ, Fairchild JM, King BR, Ambler GR, Cameron FJ, Davis EA, JonesTW; PLGM Study Reduction in Hypoglycemia With the Predictive Low-Glucose Management System: A Long-term Randomized Controlled Trial in Adolescents With Type 1 Diabetes. Diabetes Care. 2018;41(2):303-310.
  110. Predictive Low-Glucose Suspend Reduces Hypoglycemia in Adults, Adolescents, and Children WithType 1 Diabetes in an At-Home Randomized Crossover Study: Results of the PROLOG Trial. Forlenza GP, Li Z, Buckingham BA, Pinsker JE, Cengiz E, Wadwa RP, Ekhlaspour L, Church MM, Weinzimer SA, Jost E, Marcal T, Andre C, Carria L, Swanson V, Lum JW, Kollman C, Woodall W, Beck RW. Diabetes Care. 2018;41(10):2155-2161.
  111. Ekhlaspour L, Town M, Raghinaru D, Lum JW, Brown SA, Buckingham BA. Glycemic Outcomes inBaseline Hemoglobin A1C Subgroups in the International Diabetes Closed- Loop Trial. Diabetes Technol Ther. 2022;24(8):588-591.
  112. Messer LH, Tanenbaum ML, Cook PF, et al. Cost, hassle, and on-body experience: barriers to diabetesdevice use in adolescents and potential intervention Diabetes Technol Ther 2020;22:760–767.
  113. Shivers JP, Mackowiak L, Anhalt H, Zisser “Turn it off!”: diabetes device alarm fatigue considerations for the present and the future. J Diabetes Sci Technol 2013;7:789–794.
  114. Zhou K, Isaacs Closed-Loop Artificial Pancreas Therapy for Type 1 Diabetes. Curr Cardiol Rep. 2022 Sep;24(9):1159-1167. doi: 10.1007/s11886-022-01733-1.
  115. Cobry EC, Berget C, Messer LH, Forlenza GP. Review of the Omnipod® 5 Automated Glucose ControlSystem Powered by Horizon™ for the treatment of Type 1 Ther Deliv. 2020 Aug;11(8):507-519. doi: 10.4155/tde-2020-0055.
  116. Nallicheri A, Mahoney KM, Gutow HA, Bellini N, Isaacs Review of Automated Insulin Delivery Systems for Type 1 Diabetes and Associated Time in Range Outcomes. touchREV Endocrinol. 2022 Jun;18(1):27-34. doi: 10.17925/EE.2022.18.1.27.
  117. Christiansen MP, Garg SK, Brazg R, Bode BW, Bailey TS, Slover RH, Sullivan A, Huang S, Shin J, LeeSW, Kaufman Accuracy of a Fourth-Generation Subcutaneous Continuous Glucose Sensor. Diabetes Technol Ther. 2017;19(8):446-456.
  118. Bergenstal RM, Garg S, Weinzimer SA, Buckingham BA, Bode BW, Tamborlane WV, Kaufman Safety of a Hybrid Closed-Loop Insulin Delivery System in Patients With Type 1 Diabetes. JAMA. 2016;316(13):1407-1408.
  119. Garg SK, Grunberger G, Weinstock R, Lawson ML, Hirsch IB, DiMeglio LA, Pop-Busui R, Philis-TsimikasA, Kipnes M, Liljenquist DR, Brazg RL, Kudva YC, Buckingham BA, McGill JB, Carlson AL, Criego AB, Christiansen MP, Kaiserman KB, Griffin KJ, Forlenza GP, Bode BW, Slover RH, Keiter A, Ling C, Marinos B,Cordero TL, Shin J, Lee SW, Rhinehart AS, Vigersky RA; Adult and Pediatric MiniMed™ HCL Outcomes 6-month RCT: HCL versus CSII Control Study Group. Improved Glycemia with Hybrid Closed-Loop Versus Continuous Subcutaneous Insulin Infusion Therapy: Results from a Randomized Controlled Trial. Diabetes Technol Ther. 2023;25(1):1-12.
  120. Lal RA, Basina M, Maahs DM, Hood K, Buckingham B, Wilson One Year Clinical Experience of the First Commercial Hybrid Closed-Loop. Diabetes Care. 2019;42(12):2190-2196.
  121. Messer LH, Berget C, Vigers T, et al. Real world hybrid closed-loop discontinuation: predictors andperceptions of youth discontinuing the 670G system in the first 6 Pediatr Diabetes 2020;21:319–327.
  122. https://www.medtronicdiabetes.com/products/minimed-780g- system?utm_source=google&utm_campaign=Pumps+-+BRAND+-+Core+-+Exact++-+LP_Test_fy24_pumps_asbly&utm_medium=cpc&gclid=Cj0KCQjw1_SkBhDwARIsANbGpFvAwdARyEPkREpyy8EWXp5ysEutJ5OXqYmzQbyqn3raueXzLYRYSecaAssxEALw_wc B&gclsrc=aw.ds. Site accessed June 29,2023.
  123. Carlson AL, Sherr JL, Shulman DI, Garg SK, Pop-Busui R, Bode BW, et al. Safety and GlycemicOutcomes During the MiniMed™ Advanced Hybrid Closed-Loop System Pivotal Trial in Adolescents and Adults with Type 1 Diabetes. Diabetes Technol Ther. 2022;24(3):178–189. https:// doi. org/ 10. 1089/ dia. 2021. 0319.
  124. Choudhary P, Kolassa R, Keuthage W, Kroeger J, Thivolet C, Evans M, Ré R, de Portu S, Vorrink L, Shin J, Habteab A, Castañeda J, da Silva J, Cohen O; ADAPT study Group. Advanced hybrid closed loop therapy versus conventional treatment in adults with type 1 diabetes (ADAPT): a randomised controlled study. Lancet Diabetes Endocrinol. 2022 Oct;10(10):720-731. doi: 10.1016/S2213-8587(22)00212-1.
  125. Matejko B, Juza A, Kieć-Wilk B, Cyranka K, Krzyżowska S, Chen X, Cohen O, Da Silva J, Malecki MT, Klupa T. Transitioning of People With Type 1 Diabetes From Multiple Daily Injections and Self-Monitoring of Blood Glucose Directly to MiniMed 780G Advanced Hybrid Closed-Loop System: A Two-Center, Randomized, Controlled Study. Diabetes Care. 2022;45(11):2628-2635.
  126. Arrieta A, Battelino T, Scaramuzza AE, Da Silva J, Castañeda J, Cordero TL, Shin J, Cohen Comparison of MiniMed 780G system performance in users aged younger and older than 15 years: Evidencefrom 12 870 real-world users. Diabetes Obes Metab. 2022 Jul;24(7):1370-1379. doi: 10.1111/dom.14714.
  127. Hood KK, Laffel LM, Danne T, Nimri R, Weinzimer SA, Sibayan J, Bailey RJ, Schatz D, Bratina N, Bello R, Punel A, Calhoun P, Beck RW, Bergenstal RM, Phillip M. Lived Experience of Advanced Hybrid Closed-Loop Versus Hybrid Closed-Loop: Patient- Reported Outcomes and Diabetes Technol Ther. 2021Dec;23(12):857-861. doi: 10.1089/dia.2021.0153.
  128. Brown SA, Kovatchev BP, Raghinaru D, et Six-month randomized, multicenter trial of closed-loop control in type 1 diabetes. N Engl J Med 2019;381:1707-1717.
  129. Pinsker JE, Müller L, Constantin A, Leas S, Manning M, McElwee Malloy M, Singh H, Habif S. Real-World Patient-Reported Outcomes and Glycemic Results with Initiation of Control-IQ DiabetesTechnol Ther. 2021 Feb;23(2):120-127. doi: 10.1089/dia.2020.0388.
  130. https://www.myomnipod.com/healthcareproviders/about-omnipod/innovation. Site accessed June29,2023.
  131. Brown SA, Forlenza GP, Bode BW, et al. Multicenter trial of a tubeless, on-body automated insulindelivery system with customizable glycemic targets in pediatric and adult participants with type 1 diabetes. Diabetes Care. 2021;44:1630–40.
  132. Bionic Pancreas Research Group; Russell SJ, Beck RW, Damiano ER, El-Khatib FH, Ruedy KJ, BalliroCA, Li Z, Calhoun P, Wadwa RP, Buckingham B, Zhou K, Daniels M, Raskin P, White PC, Lynch J, Pettus J, Hirsch IB, Goland R, Buse JB,
  133. Kruger D, Mauras N, Muir A, McGill JB, Cogen F, Weissberg-Benchell J, Sherwood JS, Castellanos LE, Hillard MA, Tuffaha M, Putman MS, Sands MY, Forlenza G, Slover R, Messer LH, Cobry E, Shah VN, PolskyS, Lal R, Ekhlaspour L, Hughes MS, Basina M, Hatipoglu B, Olansky L, Bhangoo A, Forghani N, Kashmiri H, Sutton F, Choudhary A, Penn J, Jafri R, Rayas M, Escaname E, Kerr C, Favela-Prezas R, Boeder S, Trikudanathan S, Williams KM, Leibel N, Kirkman MS, Bergamo K, Klein KR, Dostou JM, Machineni S, YoungLA, Diner JC, Bhan A, Jones JK, Benson M, Bird K, Englert K, Permuy J, Cossen K, Felner E, Salam M, Silverstein JM, Adamson S, Cedeno A, Meighan S, Dauber A. Multicenter, Randomized Trial of a Bionic Pancreas in Type 1 N Engl J Med. 2022 Sep 29;387(13):1161-1172.
  134. DeSalvo D, Buckingham Continuous glucose monitoring: current use and future directions.Curr Diab Rep. 2013;13(5):657-62.
  135. Bekiari E, Kitsios K, Thabit H, Tauschmann M, Athanasiadou E, Karagiannis T, Haidich AB, Hovorka R, Tsapas A. Artificial pancreas treatment for outpatients with type 1 diabetes: systematic review and meta-analysis. BMJ. 2018;361:k1310.
  136. Weisman A, Bai JW, Cardinez M, Kramer CK, Perkins BA. Effect of artificial pancreas systems onglycaemic control in patients with type 1 diabetes: a systematic review and meta-analysis of outpatient randomised controlled trials. Lancet Diabetes Endocrinol 2017;5:501-
  137. Jiao X, Shen Y, Chen Better TIR, HbA1c, and less hypoglycemia in closed-loop insulin system in patients with type 1 diabetes: a meta-analysis. BMJ Open Diabetes Res Care. 2022 Apr;10(2):e002633. doi: 10.1136/bmjdrc-2021-002633.
  138. Alexandraki K, Tentolouris N, Stefanadis C, Chrousos GP, Tousoulis Effectiveness of artificial pancreas in the non-adult population: A systematic review and network meta- analysis. Metabolism. 2019 Jan;90:20-30. doi: 10.1016/j.metabol.2018.10.002.
  139. Bailey TS, Grunberger G, Bode BW, Handelsman Y, Hirsch IB, Jovanovič L, Roberts VL, Rodbard D,Tamborlane WV, Walsh J; American Association of Clinical Endocrinologists (AACE); American College of Endocrinology (ACE). American Association of Clinical Endocrinologists and American College of Endocrinology 2016 Outpatient Glucose Monitoring Consensus Statement. Endocr Pract. 2016;22(2):231-61.
  140. Ma R, Shao R, An X, Zhang Q, Sun S. Recent advancements in noninvasive glucose monitoring andclosed-loop management systems for J Mater Chem B. 2022 Jul 27;10(29):5537-5555. doi: 10.1039/d2tb00749e.
  141. Malanda UL, Welschen LM, Riphagen II, Dekker JM, Nijpels G, Bot SD. Self-monitoring of blood glucose in patients with type 2 diabetes mellitus who are not using insulin. Cochrane Database Syst Rev. 2012;1:CD005060.
  142. Cahn A, Akirov A, Raz Digital health technology and diabetes management. J Diabetes. 2018;10(1):10-17.
  143. Dinh-Le C, Chuang R, Chokshi S, Mann D. Wearable Health Technology and Electronic Health RecordIntegration: Scoping Review and Future JMIR Mhealth Uhealth. 2019;7(9):e12861.
  144. Klonoff DC, Kerr Overcoming Barriers to Adoption of Digital Health Tools for Diabetes. J Diabetes Sci Technol. 2018;12(1):3-6.
  145. Klonoff DC, Kerr D, Kleidermacher Now Is the Time for a Security and Safety Standard for Consumer Smartphones Controlling Diabetes Devices. J Diabetes Sci Technol. 2017;11(5):870-873.
  146. Blackwell M, Wheeler Clinical review: the misreporting of logbook, download, and verbal self-measured blood glucose in adults and children with type I diabetes. Acta Diabetol. 2017;54(1):1-8.
  147. Beck Downloading Diabetes Device Data: Empowering Patients to Download at Home to Achieve Better Outcomes. Diabetes Technol Ther. 2015;17(8):536-7.
  148. Lee JM, Hirschfeld E, Wedding A Patient-Designed Do-It-Yourself Mobile Technology System for Diabetes: Promise and Challenges for a New Era in Medicine. JAMA. 2016 315(14):1447-8.
  149. van der Linden J, Welsh JB, Walker Sustainable Use of a Real-Time Continuous Glucose Monitoring System from 2018 to 2020. Diabetes Technol Ther. 2021 Jul;23(7):508-511. doi: 10.1089/dia.2021.0014.Akturk HK, Dowd R, Shankar K, Derdzinski M. Real-World Evidence and Glycemic Improvement Using Dexcom G6Features. Diabetes Technol Ther. 2021 Mar;23(S1):S21- S26. doi: 10.1089/dia.2020.0654.
  150. Polonsky WH, Soriano EC, Fortmann AL. The Role of Retrospective Data Review in the Personal Use ofReal-Time Continuous Glucose Monitoring: Perceived Impact on Quality of Life and Health Outcomes. Diabetes Technol Ther. 2022 Jul;24(7):492-501. doi: 1089/dia.2021.0526.
  151. https://investors.insulet.com/news/news-details/2019/Insulet-Omnipod-DASH-System-
  152. Mobile-Apps-Now-Available-for-iOS-Devices/default.aspx. Site accessed June 29,
  153. Tandem Diabetes Care announces FDA clearance for the t:slim X2 insulin pump to bolus using the t:connect Mobile App [press release]. San Diego, CA: Tandem Diabetes Care, Inc; February 16, 2022. Site accessed June 29, 2023. https://www.fda.gov/news-events/press-announcements/fda-authorizes-first-interoperable- insulin-pump-intended-allow-patients-customize-treatment-through. Site accessed June 29, 2023.
  154. Blenner SR, Köllmer M, Rouse AJ, Daneshvar N, Williams C, Andrews Privacy Policies of Android Diabetes Apps and Sharing of Health Information. JAMA 2016;315:1051-2.
  155. Jacques Rose K, Petrut C, L'Heveder R, de Sabata IDF Europe position on mobile applications in diabetes. Diabetes Res Clin Pract. 2019;149:139-46.
  156. Klonoff DC, Kerr D, Wong JC, Pavlovic Y, Koliwad S, Hu J, Salber P, Aguilera A, Long W, Hamilton G, Chen KY, Adi S. Digital Diabetes Congress 2017. J Diabetes Sci Technol. 2017;11(5):1045-1052.
  157. Rose KJ, Petrut C, L'Heveder R, de Sabata IDF Europe's position on mobile applications in diabetes. Diabetes Res Clin Pract. 2019;149:39-46.
  158. Hou C, Carter B, Hewitt J, Francisa T, Mayor S. Do Mobile Phone Applications Improve Glycemic Control (HbA1c) in the Self-management of Diabetes? A Systematic Review, Meta-analysis, and GRADE of 14Randomized Diabetes Care. 2016;39(11):2089- 2095.
  159. Cui M, Wu X, Mao J, Wang X, Nie M. T2DM Self-Management via Smartphone Applications: ASystematic Review and Meta-Analysis. PLoS One 2016;11(11):e0166718.
  160. Bonoto BC, de Araújo VE, Godói IP, de Lemos LL, Godman B, Bennie M, Diniz LM, Junior Efficacy of Mobile Apps to Support the Care of Patients With Diabetes Mellitus: A Systematic Review and Meta-Analysisof Randomized Controlled Trials. JMIR Mhealth Uhealth. 2017;5(3):e4.
  161. Wu Y, Yao X, Vespasiani G, Nicolucci A, Dong Y, Kwong J, Li L, Sun X, Tian H, Li S. Mobile App-Based Interventions to Support Diabetes Self-Management: A Systematic Review of Randomized Controlled Trials toIdentify Functions Associated with Glycemic Efficacy. JMIR Mhealth Uhealth. 2017;5(3):e35.
  162. Veazie S, Winchell K, Gilbert J, Paynter R, Ivlev I, Eden K, Nussbaum K, Weiskopf N, Guise JM, Helfand Mobile Applications for Self-Management of Diabetes [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2018 May.
  163. Adu MD, Malabu UH, Callander EJ, Malau-Aduli AE, Malau-Aduli Considerations for the Development of Mobile Phone Apps to Support Diabetes Self-Management: Systematic Review. JMIR Mhealth Uhealth. 2018 ;6(6):e10115.
  164. Martinez-Millana A, Jarones E, Fernandez-Llatas C, Hartvigsen G, Traver V. App Features for Type 1Diabetes Support and Patient Empowerment: Systematic Literature Review and Benchmark Comparison. JMIR Mhealth Uhealth. 2018;6(11):e12237.
  165. Fu H, McMahon SK, Gross CR, Adam TJ, Wyman Usability and clinical efficacy of diabetes mobile applications for adults with type 2 diabetes: A systematic review. Diabetes Res Clin Pract. 2017;131:70-81.
  166. Chavez S, Fedele D, Guo Y, Bernier A, Smith M, Warnick J, Modave Mobile Apps for the Management of Diabetes. Diabetes Care. 2017;40(10):e145-e146.
  167. Brzan PP, Rotman E, Pajnkihar M, Klanjsek Mobile Applications for Control and Self Management of Diabetes: A Systematic Review. J Med Syst. 2016;40(9):210.
  168. Katz LB, Dirani RG, Li G, Randoll RA, Mahoney Automated glycemic pattern analysis can improve health care professional efficiency and accuracy. J Diabetes Sci Technol. 2013;7(1):163-6.
  169. Parkin CG, Davidson Value of self-monitoring blood glucose pattern analysis in improving diabetes outcomes. J Diabetes Sci Technol. 2009;3(3):500-8.
  170. Eiland L, McLarney M, Thangavelu T, Drincic App-Based Insulin Calculators: Current and Future State. Curr Diab Rep. 2018;18(11):123.
  171. Ahn Automated Bolus Calculators and Connected Insulin Pens: A Smart Combination for Multiple Daily Injection Insulin Therapy. J Diabetes Sci Technol. 2022 May;16(3):605- 609. doi: 10.1177/19322968211062624.
  172. MacLeod J, Vigersky A Review of Precision Insulin Management With Smart Insulin Pens: Opening Up the Digital Door to People on Insulin Injection Therapy. J Diabetes Sci Technol. 2023 Mar;17(2):283-289. doi: 10.1177/19322968221134546.
  173. Shashaj B, Busetto E, Sulli Benefits of a bolus calculator in pre- and postprandial glycaemic control and meal flexibility of paediatric patients using continuous subcutaneous insulin infusion (CSII). Diabet Med. 2008;25(9):1036–42.
  174. Sussman A, Taylor EJ, Patel M, Ward J, Alva S, Lawrence A, Ng R. Performance of a glucose meterwith a built-in automated bolus calculator versus manual bolus calculation in insulin-using subjects. J Diabetes Sci Technol. 2012;6(2):339-44.
  175. Maurizi AR, Lauria A, Maggi D, Palermo A, Fioriti E, Manfrini S, Pozzilli A novel insulin unit calculator for the management of type 1 diabetes. Diabetes Technol Ther. 2011;13(4):425–8.
  176. Huckvale, K, Adomaviciute, S, Prieto, JT, Leow, MK, Car, Smartphone apps for calculating insulin dose: a systematic assessment. BMC Med. 2015;13(1):106.
  177. Ahn Automated Bolus Calculators and Connected Insulin Pens: A Smart Combination for Multiple Daily Injection Insulin Therapy. J Diabetes Sci Technol. 2022 May;16(3):605- 609. doi: 10.1177/19322968211062624.
  178. Meade LT, Rushton Accuracy of carbohydrate counting in adults. Clin Diabetes. 2016;34:142–7.
  179. Weatherly J, Kishnani S, Aye T. Challenges with Patient Adoption of Automated Integration of BloodGlucose Meter Data in the Electronic Health Diabetes Technol Ther. 2019;21:671-674.
  180. Jung SY, Kim JW, Hwang H, et al. Development of Comprehensive Personal Health Records IntegratingPatient-Generated Health Data Directly From Samsung S-Health and Apple Health Apps: Retrospective Cross-Sectional Observational Study. JMIR Mhealth Uhealth. 2019;7:e12691.
  181. Espinoza J, Klonoff D, Vidmar A, et al. 2022 iCoDE Report: CGM-EHR Integration Standards and Diabetes Technol Soc. Published online November 7, 2022. Accessed March 14, 2023. https://www.diabetestechnology.org/icode/
  182. Lee EY, Cha SA, Yun JS, Lim SY, Lee JH, Ahn YB, Yoon KH, Hyun MK, Ko SH. Efficacy of PersonalizedDiabetes Self-care Using an Electronic Medical Record-Integrated Mobile App in Patients With Type 2 Diabetes: 6-Month Randomized Controlled Trial. J Med Internet Res. 2022 Jul 28;24(7):e37430. doi: 10.2196/37430.
  183. Gallagher EJ, Le Roith D, Bloomgarden Review of hemoglobin A(1c) in the management of diabetes. Journal of diabetes 2009;1:9-17.
  184. Little RR, Roberts A review of variant hemoglobins interfering with hemoglobin A1c measurement. Journal of diabetes science and technology 2009;3:446-51.
  185. http://www.ngsp.org. Site accessed June 29,
  186. Knaebel J, Irvin BR, Xie Accuracy and clinical utility of a point-of-care HbA1c testing device.Postgrad Med. 2013;125(3):91-8.
  187. Jeppsson JO, Kobold U, Barr J et Approved IFCC reference method for the measurement of HbA1cin human blood. Clin Chem Lab Med. 2002;40:78–89.
  188. Miedema Standardization of HbA1c and Optimal Range of Monitoring. Scand J Clin Lab Invest Suppl. 2005;240:61-72.
  189. Cohen RM, Snieder H, Lindsell CJ, Beyan H, Hawa MI, Blinko S, Edwards R, Spector TD, Leslie RD. Evidence for independent heritability of the glycation gap (glycosylation gap) fraction of HbA1c in nondiabetic twins. Diabetes Care. 2006; 29:1739-1743
  190. Chalew SA, McCarter RJ, Thomas J, Thomson JL, Hempe JM. A comparison of the Glycosylation Gapand Hemoglobin Glycation Index in patients with J Diabetes Complications. 2005;19:218-222.
  191. Rodríguez-Segade S, Rodríguez J, García Lopez JM, Casanueva FF, Camina F. Estimation of theglycation gap in diabetic patients with stable glycemic Diabetes Care 2012;35:2447 –2450.
  192. Nayak AU, Holland MR, Macdonald DR, Nevill A, Singh Evidence for consistency of the glycation gap in diabetes. Diabetes Care. 2011;34(8):1712-6.
  193. Sacks DB, Nathan DM, Lachin Gaps in the glycation gap hypothesis. Clin Chem 2011;57:150–152.
  194. Kilpatrick ES, Rigby AS, Atkin Variability in the Relationship between Mean Plasma Glucose and HbA1c: Implications for the Assessment of Glycemic Control. Clin Chem. 2007;53(5):897-901
  195. Quagliaro L, Piconi L, Assaloni R, Martinelli L, Motz E, Ceriello A. Intermittent high glucose enhances apoptosis related to oxidative stress in human umbilical vein endothelial cells: the role of protein kinase C andNAD(P)H-oxidase Diabetes 2003;52(11):2795-2804.
  196. Inhat M, Green D, Ross K, et al. Reduced antioxidant response of retinal and endothelial cells inresponse to chronic oscillating glucose Diabetes 2005;54(Suppl. 1):2314A.
  197. Radin Pitfalls in hemoglobin A1c measurement: when results may be misleading. J Gen Intern Med. 2014 Feb;29(2):388-94.
  198. Parrinello CM, Selvin Beyond HbA1c and glucose: the role of nontraditional glycemic markers in diabetes diagnosis, prognosis, and management. Curr Diab Rep. 2014;14(11):548.
  199. Danese E, Montagnana M, Nouvenne A, Lippi Advantages and pitfalls of fructosamine and glycatedalbumin in the diagnosis and treatment of diabetes. J Diabetes Sci Technol. 2015;9(2):169-76.
  200. Herman WH, Cohen Racial and ethnic differences in the relationship between HbA1c and blood glucose: implications for the diagnosis of diabetes. J Clin Endocrinol Metab. 2012;97(4):1067-72.
  201. Bergenstal RM, Gal RL, Connor CG, Gubitosi-Klug R, Kruger D, Olson BA, Willi SM, Aleppo G, Weinstock RS, Wood J, Rickels M, DiMeglio LA, Bethin KE, Marcovina S, Tassopoulos A, Lee S, Massaro E, Bzdick S, Ichihara B, Markmann E, McGuigan P, Woerner S, Ecker M, Beck RW; T1D Exchange RacialDifferences Study Racial Differences in the Relationship of Glucose Concentrations and HemoglobinA1c Levels. Ann Intern Med. 2017;167(2):95-102.
  202. Bower JK, Brancati FL, Selvin E. No ethnic differences in the association of glycated hemoglobin withretinopathy: the national health and nutrition examination survey 2005- 2008. Diabetes Care. 2013;36(3):569-73.
  203. Parrinello CM, Sharrett AR, Maruthur NM, Bergenstal RM, Grams ME, Coresh J, Selvin Racial Differences in and Prognostic Value of Biomarkers of Hyperglycemia. Diabetes Care. 2016;39(4):589-95.
  204. Lindsey CC, Carter AW, Mangum S, Greene D, Richardson A, Brown SJ, Essary JL, McCandless Aprospective, randomized, multicentered controlled trial to compare the annual glycemic and quality outcomes of patients with diabetes mellitus monitored with weekly fructosamine testing versus usual care. Diabetes TechnolTher. 2004;6(3):370-7.
  205. Edelman SV, Bell JM, Serrano RB, Kelemen Home testing of fructosamine improves glycemic control in patients with diabetes. Endocr Pract. 2001;7(6):454-8.
  206. Shohat N, Tarabichi M, Tischler EH, Jabbour S, Parvizi Serum Fructosamine: A Simple and Inexpensive Test for Assessing Preoperative Glycemic Control. J Bone Joint Surg Am. 2017;99(22):1900-1907.
  207. Selvin E, Rawlings AM, Grams M, Klein R, Sharrett AR, Steffes M, Coresh J. Fructosamine and glycatedalbumin for risk stratification and prediction of incident diabetes and microvascular complications: a prospective cohort analysis of the Atherosclerosis Risk in Communities (ARIC) study. Lancet Diabetes Endocrinol. 2014;2(4):279-88.
  208. Jung M, Warren B, Grams M, Kwong YD, Shafi T, Coresh J, Rebholz CM, Selvin E. Performance of non-traditional hyperglycemia biomarkers by chronic kidney disease status in older adults with diabetes: Resultsfrom the Atherosclerosis Risk in Communities Study. J Diabetes. 2018;10(4):276-285.
  209. Selvin E, Rawlings AM, Lutsey PL, Maruthur N, Pankow JS, Steffes M, Coresh J. Fructosamine andGlycated Albumin and the Risk of Cardiovascular Outcomes and Death. Circulation. 2015;132(4):269-77.
  210. Nathan DM, McGee P, Steffes MW, Lachin JM; DCCT/EDIC Research Group. Relationship of glycatedalbumin to blood glucose and HbA1c values and to retinopathy, nephropathy, and cardiovascular outcomes in the DCCT/EDIC study. Diabetes. 2014;63(1):282-90.
  211. Zhu J, Chen Y, Li C, Tao M, Teng The diagnostic value of glycated albumin in gestational diabetes mellitus. J Endocrinol Invest. 2018;41(1):121-128.
  212. Shimizu I, Hiramatsu Y, Omori Y, Nakabayashi M. Comparison of HbA1c and glycated albumin as acontrol marker for newborn complications in diabetic women in a multicentre study in Japan (Japan glycated albumin study group: study 2). Ann Clin Biochem. 2018;55(6):639-646.
  213. Dungan KM, Buse JB, Largay J, Kelly MM, Button EA, Kato S, Wittlin S. 1,5- anhydroglucitol andpostprandial hyperglycemia as measured by continuous glucose monitoring system in moderately controlled patients with diabetes. Diabetes Care. 2006;29:1214-1219.
  214. Lee AK, Lee CJ, Huang ES, Sharrett AR, Coresh J, Selvin Risk Factors for Severe Hypoglycemia in Black and White Adults With Diabetes: The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care. 2017;40(12):1661-1667.
  215. Rebholz CM, Grams ME, Chen Y, Gross AL, Sang Y, Coresh J, Selvin Serum Levels of 1,5-Anhydroglucitol and Risk of Incident End-Stage Renal Disease. Am J Epidemiol. 2017;186(8):952-960.
  216. Rawlings AM, Sharrett AR, Mosley TH, Ballew SH, Deal JA, Selvin Glucose Peaks and the Risk ofDementia and 20-Year Cognitive Decline. Diabetes Care. 2017;40(7):879-886.
  217. Selvin E, Rawlings A, Lutsey P, Maruthur N, Pankow JS, Steffes M, Coresh Association of 1,5-Anhydroglucitol With Cardiovascular Disease and Mortality. Diabetes. 2016;65(1):201-8.

Disorders in The Action of Vitamin D

ABSTRACT

 

Vitamin D is produced in the skin under the influence of UVB portion of the light spectrum in sunlight. This form of vitamin D is cholecalciferol or vitamin D3. Vitamin D may also be part of the diet either in certain foods such as fatty fish or as supplements either ingested separately or in supplemented food such as milk. Such supplements can be either vitamin D3 or vitamin D2, the form produced in plants from UVB radiation of ergosterol. For the purposes of this chapter, we will make no distinction between these forms of vitamin D, although the differences in the side chain (double bond at C22-23 and methyl group at C24 of vitamin D2) do alter both the binding of vitamin D2 to vitamin D binding protein (DBP) and the metabolism of vitamin D2. Vitamin D then must be metabolized to its most active form, 1,25 dihydroxyvitamin D (1,25(OH)2D), by a series of steps involving first the conversion to 25 hydroxyvitamin D (25OHD), the principal circulating form of vitamin D, principally by the enzyme CYP2R1 in the liver, then to 1,25(OH)2D principally in the kidney by the enzyme CYP27B1. That said there are a number of enzymes with 25-hydroxylase activity found in a number of tissues, and although CYP27B1 is essentially the only 1-hydroxylase, this enzyme is found in numerous tissues where it is thought to play primarily a paracrine/autocrine role. Putting a check to the production of 25OHD and 1,25(OH)2D is the 24-hydroxylase CYP24A1, that metabolizes both 25OHD and 1,25(OH)2D to inactive products. Like CYP27B1, CYP24A1 is widely distributed. Vitamin D and its metabolites are carried in blood tightly bound to DBP, such that only a small fraction (<1%) is free to enter most cells unless those cells express the megalin/cubilin complex which facilitates the transport of the DBP bound metabolites into the cell. DBP is produced in the liver, and its levels can vary in patients with limited hepatic synthetic capacity. Moreover, DBP is an acute phase reactant so a variety of conditions can alter DBP levels and thus the measurement of vitamin D and its metabolites.  1,25(OH)2D is the major ligand for the vitamin D receptor (VDR), a nuclear transcription factor found in most if not all cells of the body and known to regulate thousands of genes in a cell specific manner.  Although numerous physiologic functions have been attributed to vitamin D and its metabolites, clinically its major action is to control the availability of calcium and phosphate from the diet for the proper mineralization of the skeleton. Thus, disorders in vitamin D availability, metabolism, and action manifest first and foremost in the skeleton that when severe result in rickets in growing children and osteomalacia in adults in whom the growth plates have closed. After a brief review of vitamin D metabolism and molecular mechanisms of action this chapter will describe the pathologic changes in bone when the supply of mineral to bone is inadequate due to disorders in vitamin D action whether from dietary deficiency, metabolism, or mechanism of action. We will then look in depth at the causes of vitamin D deficiency including mutations resulting in altered metabolism, then discuss the range of effects different mutations in the VDR have on its function including the example of alopecia, which is best known function of VDR that is independent of its ligand 1,25(OH)2D.

 

INTRODUCTION

 

Vitamin D derived from endogenous production in the skin or absorbed from the gut is transformed into its active form by two successive steps: hydroxylation in the liver to 25-hydroxyvitamin D [25(OH)D] primarily by the enzyme CYP2R1 followed by 1a-hydroxylation in the renal proximal tubule to 1,25-dihydroxyvitamin D (1,25(OH)2D-calcitriol) by the enzyme CYP27B. Other cells exhibit 1α-hydroxylase activity including placental decidual cells, epithelial cells including keratinocytes, macrophages, dendritic cells, parathyroid cells, and some tumor cells. The role of the extrarenal production of 1,25(OH)2D is thought to play a paracrine and/or autocrine function and under normal conditions does not significantly contribute to the circulating levels of the hormone. Hydroxylation at carbon 24 to produce 24,25-dihydroxyvitamin D [24,25(OH)2D] or 1,24,25-trihydroxyvitamin D by the enzyme CYP24A1 is performed in a wide range of normal tissues and is believed to be important in the removal of vitamin D metabolites. CYP27B1 and CYP24A1 are mitochondrial mixed function oxidase containing cytochrome P450 with ferredoxin and heme-binding domains whereas CYP2R1 is likewise a P450 mixed function oxidase in the microsomes. The sequences of these genes are known contributing to an understanding of the mutations that affect their function (1-9).

 

The 25-hydroxylation of vitamin D has been thought to be primarily substrate dependent but recently studies found that it is under metabolic control (10). Obesity induced by a high fat diet reduces the levels of hepatic CYP2R1 leading to reduced levels of 25OHD (11). CYP27B1 is stringently regulated by parathyroid hormone (PTH) through a cAMP mediated pathway; calcitonin in a different region of the proximal tubule, apparently not via a rise in cAMP; 1,25(OH)2D through its receptor; calcium in part via its regulation of PTH and by phosphorus mainly via Fibroblast Growth Factor 23 (FGF-23) production in osteocytes and osteoblasts. Of these the stimulation by PTH and inhibition by FGF23 and 1,25(OH)2D are best studied and likely the dominant regulators. CYP24A1is likewise regulated by PTH and FGF23 but opposite to that of CYP27B1 and is markedly induced by 1,25(OH)2D. All vitamin D metabolites are fat soluble and circulate principally bound to vitamin D binding protein (DBP), an a-globulin produced primarily in the liver. Most of the body pool of vitamin D is in the body fat while only a small fraction of the pools of 25(OH)D or 1,25(OH)2D are in fat. Normal daily turnover of vitamin D and 25(OH)D are approximately 30 and 15 mg per day respectively with 1,25(OH)2D daily production about 1 mg. The fractional conversion of vitamin D to 1,25(OH)2D is much higher in vitamin D deficient states. It is difficult to measure vitamin D in blood. Circulating vitamin D metabolites measured in clinical practice are 25(OH)D and 1,25(OH)2D. As 25(OH)D synthesis is primarily substrate dependent, serum levels of this metabolite are taken as a measure of vitamin D status.  

 

1,25(OH)2D biologic actions are mediated via a high-affinity intracellular vitamin D receptor (VDR). VDR acts as a ligand-modulated transcription factor that belongs to the steroid, thyroid, and retinoic acid receptors gene family (12-15). VDR is found in most if not all tissues, leading to the increased recognition of multiple target organs and actions of the hormone. However, the degree to which vitamin D impacts physiologic processes outside the musculoskeletal system remains a subject of substantial investigation and controversy.

 

1,25(OH)2D is the most powerful physiological agent that stimulates active transport of calcium, and to a lesser degree phosphorus and magnesium, across the small intestine (16,17). Thus, disorders in vitamin D action will lead to a decrease in the net flux of mineral to the extracellular compartment causing hypocalcemia and secondary hyperparathyroidism. Hypophosphatemia will also ensue, as a result of both increased renal phosphate clearance due to secondary hyperparathyroidism and reduced absorption of phosphorus due to deficient 1,25(OH)2D action on the gut. Low concentrations of calcium and phosphorus in the extracellular fluid will lead to defective mineralization of organic bone matrix. Defective bone matrix mineralization of the newly formed bone and growth plate cartilage will produce the characteristic morphological and clinical signs of rickets, while at sites of bone remodeling, it will cause osteomalacia. Disorders in vitamin D action may impair the differentiation of osteoblasts and thus their functional capacity to mineralize bone matrix; this may be an additional mechanism of minor importance that contributes to rickets and osteomalacia (18,19). Continuous bone remodeling takes place in the growing skeleton, thus children with rickets will have osteomalacia as well. The clinical term used in this chapter to describe defective bone matrix mineralization in general will be rickets and/or osteomalacia.

 

Mineralization of the newly formed organic matrix of bone is a complex and highly ordered process. The essential components for normal mineralization include appropriate extracellular concentrations of calcium, phosphorous and normal functions of the bone forming cells. Disturbances in any of these components will lead to a stereotypic response of disturbed mineralization. The histological, radiological and most of the clinical features characteristic of rickets and/or osteomalacia will be the same regardless of the primary disorder. Thus, defective bone matrix mineralization can be caused by: a) calcium deficiency (20) (e.g. a rare nutritional deficiency of the element seen primarily in children or the more common form of calcium deficiency secondary to disorders of vitamin D metabolism); b) phosphorous deficiency (e.g. increased renal phosphate clearance as in X-linked hypophosphatemia (for detailed discussion see Endotext chapter entitled Primary Disorders of Phosphate Metabolism); c) primary defects in local bone processes (e.g. hypophosphatasia).

 

Serum concentrations of calcium, phosphorous, PTH, 25(OH)D, biochemical markers of bone turnover and 24 hour urinary calcium excretion will differentiate among the different primary disturbances leading to defective bone matrix mineralization (Table 1).

 

Table 1. Biochemical Parameters of Mineral and Bone Metabolism in Patients with Rickets and/or Osteomalacia, by Etiology

 

Serum levels

24h urinary calcium excretion

Etiology

Calcium

Phosphorous

iPTH

Bone specific alk.phos*

 

Hypocalcemic e.g., vitamin D deficiency

Low to low normal

Low

Elevated

Elevated

Low

Hypophosphatemic e.g., X-linked hypophosphatemia

Normal

Low

Normal to
low normal

Elevated

Low to elevated

Primary defects in bone e.g., hypophosphatasia

Normal

Normal

Normal

Low

Normal

* Alk. phos. alkaline phosphatase activity

 

This chapter will deal with hypocalcemic rickets and/or osteomalacia. A primary calcium deficiency state due to extremely low calcium intake was reported in growing children (20) but is very uncommon. Thus, the discussion will concentrate on calcium deficiency secondary to disorders in vitamin D action. Such disorders can be due to disturbances along the cascade of 1,25(OH)2D synthesis leading to a deficiency of the hormonal form of the vitamin (vitamin D deficiency states) or defects in the interaction of 1,25(OH)2D and its target tissues (resistance to 1,25(OH)2D).

 

As all disorders in vitamin D action will lead to the same clinical, radiological, histological, and most of the biochemical aberrations, those common features will be discussed first.

 

CLINICAL AND RADIOLOGICAL FEATURES OF RICKETS AND OSTEOMALACIA

 

The clinical features of rickets due to disorders in vitamin D action are weakness, bone pain, bone deformity, and fracture. The most rapidly growing bones show the most striking abnormalities. Thus, the localization and severity of the clinical features will depend on the age of onset. In children with acquired disorders in vitamin D action, signs and symptoms of rickets will vary with the age in which the deficiency is being manifested. Children with hereditary disorders of vitamin D action will appear normal at birth as calcium and phosphorous levels in fetal plasma are sustained by placental transport from maternal plasma that is not regulated by the fetal vitamin D system. These children usually develop the characteristic features of rickets within the first 2 years of life. Defects in bone mineralization are particularly evident in regions of rapid bone growth, including, during the first year of life, the cranium, wrist, and ribs. Rickets at this time will lead to widened cranial sutures, frontal bossing, posterior flattening of the skull (craniotabes), widening of the wrists, bulging of costochondral junction (rachitic rosary), and indentation of the ribs at the diaphragmatic insertion (Harrison’s groove). The rib cage may be so deformed that it contributes to respiratory failure. Dental eruption is delayed, and teeth show enamel hypoplasia. Muscle weakness and hypotonia are severe and result in a protuberant abdomen, that may contribute to respiratory failure, and may result in the inability to walk without support. After the first year of life with the acquisition of erect posture and rapid linear growth, the deformities are most severe in the legs. Bow legs (genu varum) or knock-knee (genu valgum) deformities of various severity develop as well as widening of the end of long bones. If not treated, rickets may cause severe lasting deformities, compromise adult height, and increase susceptibility to pathological fractures (Figure 1).

 

Figure 1. Teenage male with rickets. Note deformities of legs (bow legs) and compromised height.

 

The specific radiographic features of rickets reflect the failure of cartilage calcification and endochondral ossification and therefore are best seen in the metaphysis of rapidly growing bones (Figures 2 and 3). The metaphyses are widened, uneven, concave, or cupped and because of the delay in or absence of calcification, the metaphyses could become partially or totally invisible on x-ray. In more severe forms or in patients untreated for prolonged periods, rarefaction and thinning of the cortex of the entire shaft, sparse bone trabecularization, and bone deformities will become evident. Greenstick fractures may appear as well.

 

Figure 2. Radiograph of distal femur and proximal tibia and fibula of a patient with rickets. Note widening of epiphysis, resorption of provisional zone of calcification, flaring of metaphysis and bone deformity.

Figure 3. Radiograph of the wrist and hand of a patient with rickets. Note scalloping, widening and irregularity of metaphyseal end of the ulna; apparent widening of the joint space as a result of absence of epiphyseal provisional calcification.

 

The clinical features of osteomalacia in adults are subtle and could be manifested as bone pain or low back pain of varying severity in some cases. Severe muscle weakness and hypotonia may be a prominent feature in adults with vitamin D deficiency. Improvement of myopathy occurs after low doses of vitamin D.  

 

The first clinical presentation could be an acute fracture of the long bones, pubic ramii, ribs, or spine. The radiographic manifestations could be mild, e.g., generalized, nonspecific osteopenia or more specific, such as pseudofractures, commonly seen at the medial edges of the shafts of long bones (Figure 4).

 

Figure 4. Young male with osteomalacia. Note a pseudofracture in the medial edge of the upper femoral shaft (arrow).

 

In hypocalcemic rickets and/or osteomalacia, as is the case in disorders in vitamin D action, there may exist radiographic features of secondary hyperparathyroidism such as subperiosteal resorption and cysts of the long bones.

 

HISTOLOGICAL FEATURES OF RICKETS AND OSTEOMALACIA

 

The characteristic histological feature of rickets and osteomalacia is deficiency or lack of mineralization of the organic matrix of bone. Osteomalacia is defined as excess osteoid (hyperosteoidosis) and a quantitative dynamic proof of defective bone matrix mineralization obtained by analysis of time-spaced tetracycline labeling (21,22).

 

Bone Biopsy and Bone Histomorphometry

 

GENERAL

 

Transiliac bone biopsy is taken at a standard location: 2cm behind the antero-superior iliac spine and just below the crest, with a terpine 7.5mm inner diameter or 5mm for children. The biopsy should contain the inner and outer cortex and intact trabecular bone in between. Double tetracycline labeling is performed by giving the drug: e.g., dimethyl-chlortetracycline 1g/day in two divided doses, for 2 days, followed by a second administration typically of a different tetracycline with different fluorescent color after 10 to 14 days of drug free interval. The bone biopsy is performed 5 to 7 days following the last dose of tetracycline. The biopsy is sectioned undecalcified and is evaluated unstained or with different stains and techniques to obtain qualitative and quantitative histomorphometric parameters (Figure 5). The following are the commonly used quantitative histomorphometric parameters (22): trabecular bone volume (BV/TV), osteoid volume (OV/BV), osteoid surface (OS/BS), osteoid thickness (O. Th), osteoblast surface (Ob.s/BS), osteoclast surface (Oc.S/BS), osteoclast number (N.Oc/TA), double labeled surface (dLS/BS), single labeled surface (sLS/BS), mineral appositional rate (MAR), bone formation rate (BFR/BS), and mineralization lag time (MLT).

 

Figure 5. Photomicrographs of transilial bone biopsies of patients with rickets and osteomalacia (A, C and E) and siblings without bone disease (B, D and F) (Courtesy of Drs. D. Gazit and I.A. Bab). A and B: Modified Mason stain; magnification x130. Note in A: broad osteoid seams (arrow), osteoid trabeculae (heavy arrow) and irregular mineralization front (rectangular arrow).C and D: Polarized light; Von Kossa toluidine blue stain; magnification x360. Note in C: increased number of osteoid lamellae (arrows). E and F: Fluorescent photomicrograph, unstained; magnification x200. Note in E wide fluorescent bands (arrows), no double or single tetracycline labels and ground glass appearance.

 

OSTEOMALACIA

 

Histologically patients with osteomalacia with or without rickets will represent an abundance of unmineralized matrix, sometimes to the extent that whole trabeculae appear to be composed of only osteoid (Figure 5A). This will be depicted by quantitative histomorphometry as increases in osteoid volume, surface and thickness (OV/BV, OS/BS and O.TH). However, hyperosteoidosis could be observed in a number of bone diseases with a high turnover. The osteomalacic nature of the hyperosteoidosis is being demonstrated by defective mineralization; irregularity of mineralization fronts (Figure 5A); high number of osteoid lamellae (Figure 5C); broad single tetracycline fluorescent labels (Figure 5E) or no label at all, in contrast to the normal double tetracycline fluorescent labels (Figure5F). These qualitative observations have to be supported by the unequivocal changes in quantitative histomorphometry, i.e., decreases in a double and single tetracycline labeled surface (dLS/BS and sLS/BS) and in mineral apposition rate (MAR) as well as prolongation of mineralization lag time (MLT).

 

BIOCHEMICAL FEATURES OF RICKETS AND OSTEOMALACIA

 

The biochemical parameters characterizing disorders in vitamin D action can be divided into those associated with vitamin D status, the primary disturbance in mineral homeostasis and the respective compensatory mechanisms, and changes in bone metabolism. Changes in mineral and bone metabolism will be similar in all states of disorders in vitamin D action, while serum levels of vitamin D metabolites will characterize each of the classes delineated in the introduction (Table 2).

 

Table 2. Serum Levels of Vitamin D Metabolites in Patients with Disorders in Vitamin D Action, by Etiology

 

Serum levels

*Etiology

25(OH)D

1,25(OH)2D

Vitamin D deficiency

Low

Low to normal

1,25(OH)2D deficiency

Normal to elevated

Very low

Resistance to 1,25(OH)2D

Normal to elevated

Markedly elevated

* See section on Definitions and Terminology

 

As previously discussed, disorders in vitamin D action will lead to hypocalcemia and secondary hyperparathyroidism. Thus, the characteristic biochemical features are low to low-normal concentrations of serum calcium (depending on compensatory parathyroid activity), low urinary calcium excretion, hypophosphatemia, increased serum immunoreactive parathyroid hormone (iPTH) levels, increased urinary cyclic AMP excretion, and decreased tubular reabsorption of phosphate (the last two measures reflecting the biological activity of elevated iPTH). Biochemical markers associated with increased osteoid production as bone specific alkaline phosphatase and osteocalcin will be elevated in states of rickets and osteomalacia (23).

 

DEFINITIONS AND TERMINOLOGY

 

Terminology applied to disorders in vitamin D action has led to much confusion. This is principally because the terms were coined prior to our detailed understanding of vitamin D metabolism and mechanism of action. The following will define and clarify the terms deficiency and resistant and point to the limitations and ambiguity of these terms as being currently used as well as additional widely used terms such as pseudodeficiency and dependency.

 

Deficiency States

 

It is important to specify the localization of a defect as precisely as possible. The description of a deficiency state should indicate the most proximal metabolite in the vitamin D biosynthetic pathway that is deficient, emphasizing, if possible, that the concentrations of the immediate precursor of the metabolite specified are normal. For example, low serum levels of 1,25(OH)2D could be the end result of vitamin D deficiency or defects in the activity of the renal tubular enzyme 25 (OH)D1α-hydroxylase. Only in the latter case will it be appropriate to define it as calcitriol deficiency. Based on our current understanding of vitamin D biosynthesis, three states of deficiency could be envisioned involving the major metabolites: vitamin D, 25 (OH) D, and 1,25(OH)2D. As mentioned, before it is difficult to measure vitamin D in blood. Thus, in clinical practice the diagnosis of vitamin D deficiency states is usually established by measurements of serum 25(OH)D levels. 25(OH)D measurements as a marker of vitamin D status is due to relatively long half-life of 25(OH)D and levels in the blood relative to vitamin D plus a conversion of vitamin D to 25(OH)D that has been regarded as not tightly regulated. That said, recent findings indicate that CYP2R1, the major hepatic 25-hydroxylase, is influenced by disorders in metabolism (10) such as the negative impact of obesity induced by a diet high in fat on CYP2R1 expression. (24). Moreover, mutations in CYP2R1 have also been described leading to osteomalacia accompanied by low 25(OH)D but generally normal 1,25(OH)2D (25).

 

Based on the commonly available measurement of only two vitamin D metabolites, two deficiency states could be defined in clinical practice: low serum 25(OH) levels implying vitamin D and/or 25(OH)D deficiency, but referred mainly to define a vitamin D deficiency with the limitations mentioned above; and calcitriol deficiency, based on low 1,25(OH)2D and normal or high 25(OH)D serum levels (Table 2).

 

In theory, the pathogenic mechanism leading to deficiency of any of these metabolites could be categorized to either defective synthesis or increased clearance, with the exception of the parent vitamin D in which the additional possibility of deficient intake has to be taken into account. Each category: deficient synthesis or increased clearance could have a hereditary or acquired etiology and each subcategory could be further divided into simple (implying that the defect is localized only to vitamin D metabolism) or complex (implying that the defect is a part of a more generalized disturbance affecting other metabolic pathways).

 

To demonstrate the practical use and usefulness of the terminology, the following are some examples. Calcitriol deficiency could be caused by hereditary or acquired defects of the renal tubular enzyme 25(OH)D 1α-hydroxylase. If a hereditary defect involves only the vitamin D metabolic pathway it will be termed simple hereditary calcitriol deficiency. If the defect is a part of a more generalized hereditary disturbance, involving other metabolic pathways as well, e.g., Fanconi syndrome, renal tubular acidosis, X-linked hypophosphatemic rickets, or all states of increased FGF-23 secretion, the disorder will be defined as a complex hereditary 1,25(OH)2D deficiency. Decreased calcitriol synthesis could be caused by acquired diseases that usually affect additional metabolic pathways, e.g., chronic renal failure, hypoparathyroidism, tumor induced osteomalacia, thus the appropriate term will be complex acquired 1,25(OH)2D deficiency due to defective synthesis. Some drugs as barbiturates and the antiepileptic-hydantoin may affect the liver cytochrome P450 enzyme system and increase vitamin D catabolism. It may accelerate vitamin D deficiency usually in patients that have vitamin D insufficiency. Such disturbances will be termed complex acquired vitamin D deficiency due to increased clearance. Intestinal malabsorption either hereditary or acquired interferes with the absorption and enterohepatic recirculation of vitamin D and its metabolites and thus may lead to complex hereditary or acquired vitamin D deficiency due to decreased input. In the skin the substrate for vitamin D, 7-dehydrocholesterol, (7-DHC), is on the Kandutsch-Russell pathway of cholesterol synthesis with the last step of conversion of 7-DHC to cholesterol being performed by 7-DHC reductase (DHCR7). Mutations of this enzyme including polymorphisms as well a number metabolites including vitamin D and cholesterol regulate this enzyme thus also providing for complex hereditary of acquired alterations in vitamin D availability due to changes in its precursor 7-DHC (25).

 

Resistant States

 

Resistance to a factor may be defined as a state where normal levels of the factor are associated with subnormal bioeffects. This definition has two important determinants, the factor and the bioeffect. In the case of the vitamin D system, the factor can be the parent vitamin D, the 25-hydroxylated metabolite, or calcitriol. Based on vitamin D metabolism and mechanism of action, the term resistance should only be used with the most distal metabolite or the active hormonal form of vitamin D, i.e., 1,25(OH)2D. For example, patients with a renal defect in calcitriol synthesis are resistant to vitamin D or 25(OH)D caused by defective calcitriol production, but have a completely normal response to physiological replacement doses of 1a-hydroxylated vitamin D metabolites.

 

Several bioeffects in vivo have potential relevance in evaluating states of resistance to 1,25(OH)2D. These include the effects on bone matrix mineralization and the effect on calcium transport through the small intestine. However, as discussed before, rickets and/or osteomalacia are stereotypic responses of the mineralizing bone matrix to a variety of perturbations not all of which are correlated to disturbances in 1,25(OH)2D action. Two clinical examples are presented to clarify this point: a. X-linked hypophosphatemic rickets (XLH) was and still is termed vitamin D resistant rickets. However, the unresponsiveness to vitamin D of the bone disease in XLH is due to the fact that the defective mineralization is caused mainly by phosphorus deficiency though there is some suppression of calcitriol synthesis caused by increased FGF-23 secretion; b. Hereditary hypophosphatemic rickets with hypercalciuria is a disease characterized by defective bone matrix mineralization, low serum phosphorus and elevated serum 1,25(OH)2D levels, taken together, it may look as a resistance to the hormone. However, these patients have increased intestinal absorption of calcium and hypercalciuria. The primary defect in this disease is an isolated defect in a renal phosphate transporter leading to increased renal phosphate clearance, hypophosphatemia, increased 1,25(OH)2D production and the appropriate physiological response i.e., increased intestinal calcium absorption. The rickets and/or osteomalacia are the result of phosphorus deficiency rather than unresponsiveness to calcitriol (for details see Endotext chapter Primary Disorders of Phosphate Metabolism).

 

There is no doubt about the pivotal role of calcitriol on net calcium transport across the intestinal epithelium. Thus, intestinal calcium absorption measured directly or assessed by secondary changes in serum and urine calcium and iPTH levels can be utilized as a bioeffect in evaluating states of resistance to 1,25(OH)2D. When the defect in bone matrix mineralization is caused by calcium deficiency, rickets and/or osteomalacia could be used as an additional bioeffect to define calcitriol resistance.

 

Resistance to 1,25(OH)2D is defined here as a state in which normal or high levels of the hormone are associated with hypo-or normocalcemia, secondary hyperparathyroidism, rickets and/or osteomalacia caused by calcium deficiency.

 

Pseudovitamin D Deficiency and Vitamin D Dependency

 

The term pseudovitamin D deficiency refers to a state with biochemical and tissue features of vitamin D deficiency (calcium deficiency, secondary hyperparathyroidism, impaired bone matrix mineralization) with no history of vitamin D or calcium deficiency or low serum levels of 25(OH)D. This is an ambiguous term as it includes two different diseases:1,25(OH)2D deficiency and resistance to 1,25(OH)2D, the so-called pseudovitamin D deficiency type I and II respectively, and does not include the known etiology and pathogenesis of these disturbances.

 

The term vitamin D dependency has been used interchangeably with pseudovitamin D deficiency. It meant to describe patients capable or responding to, and thus dependent on, supraphysiological doses of vitamin D. This is the situation in patients with simple hereditary 1,25(OH)2D deficiency due to defects in the renal enzyme 25(OH)vitamin D 1α-hydroxylase. Patients with this disease have a complete clinical remission on physiological replacement doses of calcitriol. The term vitamin D dependency type II was applied to describe patients with simple hereditary resistance to 1,25(OH)2D due to mutations in the vitamin D receptor (VDR), the majority of whom are unresponsive to any dose of vitamin D or its active metabolites, and therefore are not dependent on vitamin D.

 

DISORDERS IN VITAMIN D ACTION

 

Three disorders in vitamin D action will be discussed in this chapter: vitamin D deficiency, mainly simple acquired vitamin D deficiency (i.e., involving only the vitamin D metabolic pathway); simple hereditary 1,25(OH)2D deficiency resulting from defects in calcitriol biosynthesis; simple hereditary resistance to 1,25(OH)2D caused by defects in calcitriol receptor-effector system. While acquired vitamin D deficiency is the most common disorder, the other two rare inborn errors in vitamin D metabolism will be discussed because of its immense contribution to the understanding of vitamin D metabolism and mode of action in human beings.

 

Vitamin D Deficiency

 

PATHOGENESIS

 

Vitamin D deficiency can be caused by decreased input or increased clearance. Decreased input of vitamin D may be the end result of: a) defective intake due to dietary (nutritional) deficiency or intestinal malabsorption; b) deficient photosynthesis of vitamin D in the skin. Increased vitamin D clearance could result from increased catabolism, mainly in the liver, or increased loss via the kidneys or intestine. As discussed, each of these disturbances could be, at least in theory, hereditary or acquired, and further subcategorized as simple (involving only the vitamin D metabolic pathway) or complex (implying a more generalized disturbance). Thus, whenever the diagnosis of vitamin D deficiency is established, there is a need to define the etiology and pathophysiology of the disorder and categorize it accordingly.

 

Vitamin D deficiency due to increased clearance is relatively uncommon in clinical practice and usually is part of more general diseases (i.e., complex disturbances) e.g., protein losing nephropathy, intestinal malabsorption, and increased liver catabolism sometimes caused by certain drugs (e.g., barbiturates, antiepileptics). The rest of this section will deal with vitamin D deficiency caused by decreased input.

 

Vitamin D content of various unfortified food substances is very low with the exception of fatty fish such as herring, mackerel, or cod liver oil. It is estimated that under normal unfortified food consumption, less than 20% of the total circulating 25(OH)D is contributed by nutritional vitamin D. In some countries the dietary content of vitamin D is higher due to vitamin D supplementation of some food products. In the United States, milk is being fortified by 400IU per quart. Increased vitamin D intake could result from habitual use of multivitamins that usually contain 400IU of vitamin D per tablet, or some calcium salt preparations that contain vitamin D as well. These supplements will of course increase the relative contribution of dietary vitamin D intake to the total body vitamin D pool in general, and when cutaneous production of vitamin D is limited, in particular.

 

Gastrointestinal malabsorption will interfere with vitamin D input from the gut and it may affect the enterohepatic recirculation of vitamin D metabolites as well. Thus, intestinal malabsorption may contribute to vitamin D deficiency by decreasing the input and increasing the clearance of the vitamin and its metabolites. Vitamin D deficiency and its clinical and biochemical consequences may be the first sign of occult-malabsorption due, for example, to non-tropical sprue.

 

Vitamin D synthesis in the skin is produced from 7-DHC under the influence of UV light with a maximal effective wave length between 290-310 nm. Thus, levels of 7-DHC regulated by DHCR7 influences the amount of vitamin D that can be produced.  Cutaneous vitamin D production is also affected by the intensity of the UV light that reaches the body, the surface area of the skin exposed and intrinsic properties of the epidermis. In northern latitudes, during the winter, almost no UV reaches the ground. In the northern parts of the United States, Canada and Northwestern Europe, between October to March, very little or practically no vitamin D is produced in exposed skin. Clothing affected by religious or cultural habits, glass, plastic and sunscreens that are widely used due to concerns of skin cancer effectively block UV radiation and prevent cutaneous vitamin D synthesis. Vitamin D production is much less effective in dark skin, melanin absorbs UV radiation, and in elderly relative to young people. However, even in the elderly, dermal vitamin D production remains very effective. It has been estimated that in summer, a 10-minute exposure, three times a week of the unprotected skin of the head and arms is adequate to prevent vitamin D deficiency in the elderly (for detailed discussion see Endotext chapter Vitamin D: Production, Metabolism, and Mechanisms of Actionand references (26-28).

 

DIAGNOSIS

 

The diagnosis of vitamin D deficiency is established by low serum concentrations of 25(OH)D. As discussed, circulating levels of 25(OH)D are a good and reliable measure of vitamin D status in most clinically relevant situations although as mentioned obesity and diabetes mellitus can alter the conversion from vitamin D to 25(OH)D. All additional biochemical parameters as well as clinical signs and symptoms reflect the primary and secondary perturbations in mineral and bone metabolism caused by vitamin D deficiency and are common to all disorders in vitamin D action and calcium deficiency (Tables 1,2). Those parameters include low to low normal serum calcium levels, hypocalciuria, secondary hyperparathyroidism, hypophosphatemia, increased levels of biochemical markers of bone turnover, rickets and/or osteomalacia. Therefore, all these measures can be used to support but not to establish the diagnosis, and mainly to assess the relative severity of the vitamin D deficiency and the response to treatment. It is important to note that in vitamin D deficiency, circulating levels of 1,25(OH)2D could vary from low to elevated (Table 2) and thus are useless for the diagnosis.

 

25(OH)D serum levels are currently being determined in clinical practice by methods that have become more accurate and reproducible, though still with some variability. Reference values of serum 25(OH)D that were population based introduce uncertainty. These reference values differed according to geography, season, dress habits, bed or housebound situations, and age, all of which may affect sunshine exposure and thus vitamin D synthesis, as well as eating habits, local regulation on food fortification, and customs of vitamin supplementation, all of which will affect vitamin D intake. An alternative approach to obtain clinically meaningful reference values was to define health-based parameters, i.e., values of serum 25(OH)D levels below which adverse health outcome may occur. In actuality it is an intervention threshold reference value, below which therapy may prevent adverse effects on the musculoskeletal system (29). This approach is based on the current notion that though severe vitamin D deficiency will lead to rickets and/or osteomalacia, milder vitamin D deficiency states will not affect bone matrix mineralization, but via its effect on mineral homeostasis will cause secondary hyperparathyroidism, increased bone turnover and bone loss. These aberrations in mineral and bone metabolism will contribute to the development and acceleration of osteoporosis and together with the non-skeletal effects of vitamin D on physical function, will increase the risk of low impact skeletal fractures (28,30). The relationship between serum concentrations of 25(OH) D and iPTH in various vitamin D states was analyzed in multiple studies (28,31-33). The aim was to define serum 25(OH)D concentrations below which serum iPTH levels started to increase, or base line 25(OH)D levels above which vitamin D supplementation caused a significant decrease of iPTH serum concentrations. Both approaches yielded similar functional thresholds of serum 25(OH)D levels that affect circulating iPTH concentrations, although the relationship between 25(OH)D and PTH levels is substantially affected by calcium intake.

 

Based on these studies, a diagnostic staging of vitamin D deficiency states, based on serum 25(OH)D levels and secondary perturbation in mineral and bone metabolism has been proposed (28). Mild vitamin D deficiency (or vitamin D insufficiency) is diagnosed when serum 25(OH)D levels are below 50 nmol/liter (20 ng/ml). This is associated with mild elevations of serum iPTH and biochemical markers of bone turnover. Moderate vitamin D deficiency is defined when serum 25(OH)D levels are below 25 nmol/liter (10 ng/ml). Serum iPTH concentration is moderately increased with high bone turnover. Severe vitamin D deficiency occurs when serum 25(OH)D levels are lower than 12.5 nmol/liter (5 ng/ml), iPTH circulating concentration may be markedly increased and rickets and/or osteomalacia may occur. It is obvious that according to this scheme, vitamin D sufficiency is defined as serum 25(OH)D levels above 50 nmol/liter (20 ng/ml). That said these cut off values remain controversial.

 

A somewhat similar definition was suggested recently by an expert panel for the Institute of Medicine (IOM) (current name National Academy of Medicine (NAM)) recommending that a level of 50 nmol/liter (20 ng/ml) of 25(OH)D was sufficient for 97.5% of the adult USA population, although up to 125 nmol/liter (50 ng/ml) was considered safe (34). At the same time, another group of experts, from the Endocrine Society, suggested that the most advantageous value of 25(OH)D for musculoskeletal health is 75 nmol/liter (30 ng/ml) (35).

 

PREVALENCE

 

Serum 25(OH)D levels vary widely among different populations. On average serum 25(OH)D concentration are lower in European countries than in the USA and even lower in some countries in the Middle East and the Asia-Pacific region. A multinational study (with the exception of North America), in post-menopausal women treated for osteoporosis, revealed that 30-90% (depending on location and season), had serum levels of 25(OH)D below 75 nmol/liter (30 ng/ml), (the highest frequency of women below this level was observed in the Middle East and Asia (two countries in each region)). If the threshold is taken as 50 nmol/liter (20 ng/ml), 31% of the women fell below that threshold. Based on more than 40 studies from Europe, North America, the Middle East and the Asian-Pacific regions (28), it seems that a gradual decline in mean serum 25(OH)D levels is apparent starting from healthy adults to independent elderly, institutionalized elderly, and the lowest value in patients with hip fracture. Several studies of patients hospitalized for non-traumatic fractures, revealed that few (about 5%) of these subjects had serum 25(OH)D levels above 50 nmol/liter (20 ng/ml); the majority of them had vitamin D deficiency of various severity.

 

As discussed, vitamin D status will be determined by cutaneous photosynthesis and intake. The populations at risk to develop vitamin D deficiency are those who have limited sunshine exposure and no vitamin D supplementation. Thus, vitamin D insufficiency and deficiency will be more prevalent in populations that are not exposed to sunshine due to geography-latitude, cultural, religious, and life-style habits, or are less efficient in cutaneous vitamin D synthesis due to age or pigmentation. The most vulnerable populations are those who are unable to move freely, bedridden or their mobility is severely diminished. This is more common at the beginning or the end of the life cycle, i.e., infants and the elderly, but of course it can happen at any age when free movement is limited due to physical or mental handicap.

 

The prevalence of osteomalacia is much lower and will depend on the criteria used for diagnosis, i.e., clinical, biochemical, bone histology or quantitative bone histomorphometry. In a review by Lips (28) of 19 publications describing bone histomorphometry of the femoral head or iliac crest biopsy in a total of about 1400 patients with hip fracture, the frequency of osteomalacia ranged from none to more than 30%. This difference may reflect different populations but also the histological criteria used to define osteomalacia. The very high incidence of vitamin D deficiency recorded in patients with hip fractures supports the notion that increased fracture risk is not just the outcome of frank osteomalacia but may result from secondary hyperparathyroidism, increased bone turnover, bone loss, and increased risk of low impact fractures caused by vitamin D deficiency. This is further supported by the positive relationship observed between serum 25(OH)D levels (below a certain threshold) and hip BMD (28,36,37), and the negative relationship observed between hip BMD and serum iPTH (28,36-38). Moreover, a daily dose of 700-800 IU and above of vitamin D and calcium supplementation caused a significant reduction in the incidence of hip and other nonvertebral fractures in women with post-menopausal osteoporosis, including nursing home residents (29,30,39).

 

TREATMENT

 

Adequate intake of vitamin D was defined recently in the USA by two working groups (NAM and the Endocrine Society) as discussed above. Both reports agree on the recommended dose for infants (400 IU/day) and children (600 IU/day). There is a difference in the recommendations for adults (600 IU/day) until the age of 70 (including pregnant and lactating women) and 800 IU/day above this age (NAM report), while the Endocrine Society recommendations are 1,500-2,000 IU/day for all of the adult population (34,35). In Europe, vitamin D intake recommendations are similar or somewhat lower than the NAM. The upper tolerable daily doses of vitamin D were suggested to be 4,000 IU, 10,000 IU, and 2,000 IU by the NAM, the Endocrine Society guidelines and the European Commission, respectively. There is an unresolved debate on the equipotency of vitamin D2 versus D3, but as vitamin D3 is widely available, all recommendations are actually for vitamin D3. These recommended intakes of vitamin D are usually unachievable if widely consumed food substances are not fortified with vitamin D. Thus, the population at large and the elderly population in particular are dependent on adequate cutaneous vitamin D synthesis, i.e., sunshine exposure, or vitamin D supplementation. In the United States and a few additional countries, milk is fortified with vitamin D, 400 IU per quart or liter, and the consumption of multivitamins containing vitamin D as well is relatively common, but not uniform or mandatory. Thus, in most countries and even in the USA, the elderly and especially the immobile, housebound elderly are prone to develop vitamin D deficiency.

 

Treatment has to be targeted towards the population with the highest risk to develop vitamin D deficiency. For a long period, vitamin D supplementation for infants up to 1 year of age, is mandatory in many countries and the daily dose recommendations was increased from 200 IU/day to 400 IU/day (34,35). Unfortunately, this has not yet become common practice for the elderly population. As discussed, nursing home residents, institutionalized and hospitalized elderly, and patients with hip and other non-traumatic fractures and neurological disorders are among the ones with the highest risk of having and developing vitamin D deficiency. Treatment with the recommended doses of vitamin D and calcium (see below) can be initiated even before biochemical screening of 25(OH)D serum levels and indices of mineral and bone metabolism is obtained. This is based on the very high incidence of vitamin D deficiency in this population of patients and on the fact that with the recommended vitamin D doses and the tight physiological control of 1.25(OH)2D production, no toxicity is likely to occur at doses of up to 4,000 IU/day for a limited period.

 

It is important to remember that in concordance with vitamin D treatment, the recommended daily calcium allowance must be achieved as well, usually by calcium salt supplementation (see Endotext chapter Osteoporosis: Prevention and Treatment).

 

The usual recommended oral doses of vitamin D as discussed above are 600 to 1,500 IU daily. In severe vitamin D deficiency, doses of 4,000 IU- to 6,000 IU per day (or the equivalent weekly or twice weekly dose) could be given for the first 4-6 weeks, followed by dose adjustment in accordance with the biochemical response, with the final aim to achieve the recommended maintenance dose. Because vitamin D is stored in fat and released slowly and the half-life of 25(OH)D is 2-3 weeks, the vitamin can be given orally once a week. Studies with administration of vitamin D in megadoses at intervals greater than monthly have shown increased risk of falls and should be avoided (28,40-43).

 

The response to treatment with vitamin D will depend on the degree and severity of vitamin D deficiency and the secondary changes in mineral and bone metabolism. In severe vitamin D deficiency with osteomalacia, a dramatic response in the signs, symptoms, and laboratory parameters will be observed. Bone pain and muscle weakness will improve quickly, pseudofractures will show signs of healing on x-ray, and serum calcium, iPTH and biochemical markers of bone turnover will return towards the normal range. In moderate or mild vitamin D deficiency or insufficiency, the response to treatment is more subtle. Muscle weakness and bone pain may improve, serum 25(OH)D levels will increase towards the normal, serum iPTH and biochemical markers of bone turnover will return towards normal (this will be a function of the severity of the initial vitamin D deficiency). In the long run, bone mineral density may increase somewhat and the incidence of fractures may decrease. These results are based on responses of groups of patients in clinical trials and not individuals (28).

 

1,25 Dihydroxyvitamin D Deficiency

 

As discussed, 1,25(OH)2D deficiency is defined as low circulating levels of this metabolite with normal or elevated (depending on preceding vitamin D therapy) serum concentrations of 25(OH)D. In theory, it could be the end result of decreased production or increased clearance. Increased clearance of 1,25(OH)2D is uncommon and usually a part of increased clearance of additional vitamin D metabolites such as 25(OH)D, thus it would fit the definition of vitamin D deficiency.

 

Decreased production of 1,25(OH)2D could be hereditary or acquired and each one can be subcategorized as simple or complex. Acquired 1,25(OH)2D deficiency due to defective synthesis is usually part of a more general disease (i.e., complex) as chronic renal failure, acquired Fanconi syndrome, hypoparathyroidism, or tumor induced osteomalacia, that will affect mineral and bone metabolism in a multiple and complex ways that is beyond the scope of this chapter. Thus, the entity to be discussed here will be simple hereditary 1,25(OH)2D deficiency caused by defective synthesis of calcitriol.

 

Prader et al. (44) were the first to describe two young children who showed all the usual clinical features of vitamin D deficiency despite adequate intake of the vitamin, thus coining the name “pseudovitamin D deficiency.” Complete remission was dependent on continuous therapy with high doses of vitamin D, thus the term “vitamin D-dependent rickets.” However, remission of the disease could be achieved by continuous therapy with physiological (microgram) doses of 1a-hydroxylated vitamin D metabolites (45,46).

 

Family studies have revealed it to be an autosomal recessive disease. Linkage analysis in a subset of French-Canadian families assigned the gene responsible for the disease to chromosome 12q13 (47,48). The gene encoding the 25(OH)D-1α-hydroxylase (CYP27B1) of mouse kidney, human keratinocytes, and peripheral mononuclear cells was localized on chromosome 12q 13.1-13.3 which maps to the disease locus of simple hereditary 1,25(OH)2D deficiency (49-55). Cloning and sequencing of the enzyme has enabled the demonstration of its decreased expression in patients with this disease (56) as well as enabling studies showing that the same enzyme is widely distributed in tissues and not limited to the kidney (56,57). The sequence of the human CYP27B1 gene from keratinocytes and peripheral blood mononuclear cells has been shown to be identical with the renal gene (49,51-55), thus supporting the use of these accessible cells as a proxy to study the renal tubular enzymatic defect. That said regulation of extrarenal CYP27B1 differs from that of the kidney, with its regulation in keratinocytes and macrophages responding to cytokines rather than PTH and FGF23 (25). Subjects with mutations in CYP27B1 have circulating levels of 25(OH)D that are normal or elevated, depending on previous vitamin D treatment; serum concentrations of 1,25(OH)2D are very low (Table 2); while massive doses (100-300 times the daily recommended dose) of vitamin D or 25(OH)D are required to maintain remission of rickets, physiological replacement doses of calcitriol are sufficient to achieve the same effect.  The 1α-hydroxylase gene from more than 25 families with simple hereditary 1,25(OH)2D deficiency and some of their first-degree healthy relatives were analyzed by direct sequencing, site directed mutagenesis, and cDNA expression in transfected cells (49,51-55). All patients had homozygous mutations while parents or other healthy siblings were heterozygous for the mutation. Most patients of French-Canadian origin had the same mutation causing a frameshifting and a premature stop codon in the putative heme-binding domain (51). The same mutation was observed in additional families of diverse origin (55). All other patients had either a base-pair deletion causing premature termination codon upstream from the putative ferredoxin and heme-binding domains, or missense mutations (56,58-61). No 1α-hydroxylase activity was detected when the mutant enzyme was expressed in various cells. Taken together, these observations support the notion that the etiology of this hereditary disease is a defect in CYP27B1.

 

The beneficial therapeutic effect of high serum concentrations of 25(OH)D in patients treated with massive doses of vitamin D, while 1,25(OH)2D levels remain low, may have several possible explanations. First, high levels of 25(OH)D may activate the VDR whose affinity for this metabolite is approximately two orders of magnitude lower than for 1,25(OH)2D. Second, a metabolite of 25(OH)D may act directly on target tissue, and finally, high levels of 25(OH)D may drive the production of 1,25(OH)2D if the mutated enzyme has some residual function.

 

The differential diagnosis of simple hereditary 1,25(OH)2D deficiency from other hereditary or acquired forms of hypocalcemic rickets and/or osteomalacia especially the one associated with defects in the vitamin D receptor-effector system is based on serum concentrations of calcitriol and the response to treatment with 1-alpha hydroxylated vitamin D metabolites (Table 2).

 

A similar syndrome has been described and studied in a mutant strain of pigs where the mode of inheritance as well as the clinical and biochemical features are similar to the human disease (62,63). Piglets affected by the disease have rickets, elevated 25(OH)D with low or undetectable 1,25(OH)2D serum concentrations, normal specific tissue binding sites for tritiated 1,25(OH)2D, and no detectable activity of 25(OH)D-1α-hydroxylase in renal cortical homogenates. Thus, there is strong evidence that the disease state in the pig is caused solely by an inherited defect in the porcine CYP27B1. An animal model in which the gene encoding 25(OH)D-1α-hydroxylase was knocked out by homologous recombination reproduced all the clinical and biochemical features of this disease including undetectable serum 1,25(OH)2D levels (64).

 

Simple Hereditary Resistance to 1,25 Dihydroxy-Vitamin D

 

This is a rare disorder and only about 60 patients have been reported (65) Brooks et al. (66) described an adult patient with hypocalcemic osteomalacia and elevated serum concentration of 1,25(OH)2D. Treatment with vitamin D, causing a further increase in serum calcitriol levels, cured the patient. The term vitamin D-dependent rickets type II was suggested to describe this disorder. However, reports on additional patients, about half of whom did not respond to vitamin D therapy, as well as in vivo and in vitro studies to be discussed below, seem to prove that vitamin D dependency is a misnomer. The term hereditary vitamin D resistant rickets (HVDRR) is the current most widely used name of this disorder.

 

CLINICAL AND BIOCHEMICAL FEATURES

 

General Features

 

The clinical, radiological, histological, and biochemical features (except serum levels of vitamin D metabolites) are typical of hypocalcemic rickets and/or osteomalacia as previously discussed with one exception. Many of these patients develop alopecia in early childhood, not seen in even severe cases of vitamin D deficiency. In these patients there is no history of vitamin D deficiency and no clinical or biochemical response to physiological doses of vitamin D or its 1a-hydroxylated active metabolites. Serum levels of 25(OH)D are normal or elevated (depending on preceding vitamin D therapy); 1,25(OH)2D concentrations are markedly elevated before or during therapy with vitamin D preparations (Table 2).

 

The disease manifests itself as an active metabolic bone disease in early childhood. However, late onset at adolescence and adulthood was documented in several sporadic cases including the first report by Brooks et al. (66,67). These patients represented the mildest form of the disease and had a complete remission when treated with vitamin D or its active metabolites. It is unclear if the adult-onset patients belong to the same hereditary entity, as no further studies on their VDR status have been published.

 

Ectodermal Anomalies  

 

A peculiar clinical feature of the patients, appearing in more than half of the subjects, is total alopecia or sparse hair (Figure 6). Alopecia usually appears during the first year of life and in one patient, at least, has been associated with additional ectodermal anomalies (68) (Figure 6).

 

Figure 6. Patient with mutation in VDR demonstrating both alopecia and changes in dentition.

 

It seems that alopecia is a marker of a more severe form of the disease as judged by the earlier onset, the severity of the clinical features, the proportion of patients who do not respond to treatment with high doses of vitamin D or its active metabolites, and the extremely elevated levels of serum 1,25(OH)2D recorded during therapy (69,70). Though some patients with alopecia could achieve clinical and biochemical remission of their bone disease, none have shown hair growth. The notion that total alopecia is probably a consequence of a defective vitamin D receptor-effector system is supported by the following: alopecia has only been associated with hereditary defects in the VDR system, i.e., with end-organ resistance to the action of the hormone, and has not been recorded with hereditary deficiency in 1,25(OH)2D synthesis, i.e., low circulating levels of the hormone (71); alopecia is present in kindreds with different defects in the VDRs; high-affinity uptake of tritiated 1,25(OH)2D3 in the nucleus of the outer root sheath of the hair follicle of rodents has been demonstrated by autoradiography (72). Of interest is that mutations in Hairless likewise cause alopecia with histologic features similar to those seen with VDR mutations. Hairless and VDR interact in the keratinocyte suggesting their codependence for regulation of hair follicle cycling (73). High dietary or infusions of calcium can reverse the skeletal changes but do not reverse the alopecia (74). Finally, alopecia developed in homozygote VDR knockout mice (75-77). Taken together, it could be hypothesized that an intact VDR-effector system is important for the differentiation of the hair follicle in the fetus, which is unrelated to mineral homeostasis.

 

VITAMIN D METABOLISM

 

Serum concentrations of 1,25(OH)2D range from upper normal values to markedly elevated before therapy, but on vitamin D treatment may reach the highest levels found in any living system (100 times and more than the upper normal range) (70). These values may represent the end results of four different mechanisms acting synergistically to stimulate strongly the renal 25(OH)D-1α-hydroxylase. Three of the mechanisms are hypocalcemia, secondary hyperparathyroidism, and hypophosphatemia. The fourth mechanism may be a failure of the negative feedback loop by which the hormone inhibits the renal enzyme activity caused by the basic defect in the VDR-effector system. This was demonstrated in a patient in remission (normal serum levels of calcium, phosphorus and PTH) in whom a load of 25(OH)D3 had caused a marked increase in serum 1,25(OH)2D3 concentration (69,78). It was reported that the 1α-hydroxylase gene expression was not suppressed by 1,25(OH)2D3 in renal tubular cells from VDR knock out mice while it was suppressed in cells with normal VDR or heterozygote for the null mutation (6,79).

 

MODE OF INHERITANCE

 

In approximately half of the reported kindreds, parental consanguinity and multiple siblings with the same defect suggest autosomal recessive mode of inheritance (69). Parents or siblings of patients who are expected to be obligate heterozygotes have been reported to be normal, i.e., no bone disease or alopecia and normal blood biochemistry. However, studies on cells (cultured dermal fibroblasts, Epstein-Barr transformed lymphoblasts, and mitogen-stimulated lymphocytes) obtained from parents or siblings of affected children revealed decreased bioresponses, decreased normal VDR protein and its mRNA, and a heterozygote genotype exhibiting both normal and mutant DNA alleles (80-83). There is a striking clustering of patients around the Mediterranean, including patients reported from Europe and America who originated from the same area. A notable exception is a cluster of some kindreds from Japan (67,84-89).

 

CELLULAR AND MOLECULAR DEFECTS

 

Methods  

 

The near ubiquity of a similar if not identical VDR-effector system among various cell types including cells originating from tissues easily accessible for sampling made feasible studies on the nature of the intracellular and molecular defects in patients with simple hereditary resistance to 1,25(OH)2D. The cells used were mainly fibroblasts derived from skin biopsies (69,78,80-82,87,90-104) and peripheral blood mononuclear (PBM) cells. PBM cells contain high-affinity receptors for 1,25(OH)2D3 that are expressed constitutively in monocytes and are induced in mitogen-stimulated T-lymphocytes and Epstein-Barr (EB) transformed lymphoblasts. All cells have been used to assess most of the steps in 1,25(OH)2D3 action from cellular and subcellular uptake of the hormone to bioresponse as well as to elucidate the molecular aberrations in the VDR protein, RNA, and DNA.

 

The hormone-receptor interaction has been analyzed by several methods including binding characteristics of 3H 1,25(OH)2D3 to intact cells, nuclei or high salt cellular soluble extracts (69,78,80-83,90-92,95-99,105-110); measurements of VDR protein content by monoclonal antibodies with radiological immunoassay or Western blot analysis (81,82); by immunocyto-chemical methods in whole cells (111); characterization of the hormone-receptor complex on continuous sucrose gradient and nonspecific DNA-cellulose columns (69,78,80,90-92,94-97,106,110,112).

 

The cloning and nucleotide sequencing of the human VDR gene made feasible studies of the molecular defects in patients with this disease. The methods used included, among others, isolation, amplification, and sequencing of genomic VDR DNA, as well as cloning and sequencing of VDR complementary DNA (cDNA). The mutant DNA was recreated in vitro and was transfected into cells that do not express endogenous VDR. Post-transcriptional action of 1,25(OH)2D was tested in cells originating from patients or in cells co-transfected with VDR (either mutant or wild type) fused to a promoter containing vitamin D response element (VDRE).

 

Studies with the above-mentioned methods in cells originating from a variety of patients revealed heterogeneity of the cellular and molecular defects in the VDR-effector system. Based on the known functional properties of the VDR, different classes of defects could be identified.

 

Defect in the Hormone Binding Region (Including Heterodimerization)

 

Deficient Hormone Binding. There are three subgroups in this class:

 

(i) No Hormone Binding. This is the most common abnormality observed and is characterized by unmeasurable specific binding of 3H 1,25(OH)2D3 to either intact cells, nuclei, or cell extracts (69,78,81,82,91,96,100-102,105,107,112-116). Studies in several kindreds with this defect (including an extended kindred with 8 patients studied) revealed undetectable levels of VDR by immunoblots on an immunoradiometric assay in most kindreds (81,82,102,116,117). DNA from these affected subjects exhibited a single base mutation that was different in each kindred resulting in a premature stop codon. The truncated VDRs produced lacked hormone binding or both hormone and DNA binding domains (Figure 7) (113-115,117). The recreated mutant VDR cDNA was expressed in mammalian cells, and the resulting mutant VDR was demonstrated to be the truncated protein that exhibited no specific hormone binding. In cells cultured from parents of some patients, expected to be obligate heterozygotes, binding of 3H 1,25(OH)2D3, VDR protein, and mRNA content of cells ranged from the lower limit of normal to about half the normal level.

 

Figure 7. Schematic presentation of homozygous mutation in the VDR protein in simple hereditary resistance to 1,25(OH)2D. The asterisks depict sites of amino acid substitutions due to point mutation and codon changes, using the numbering system of Baker et (13); fs-frame shift.

 

In one patient representing a kindred with no hormone binding, a missense mutation resulted in the substitution of the hydrophobic basic arginine 274 by the hydrophilic nonpolar leucine in the hormone binding region (116) (Figure 7). In this patient, normal transcription could be elicited by 1000-fold higher concentrations of calcitriol than needed for the wild-type receptor. However, no in vivo or in vitro stimulation of 25(OH)D-24-hydroxylase could be obtained by high concentrations of 1,25(OH)2D3 (see below).

 

Two siblings without alopecia and no response to any dose of 1,25(OH)2D in vivo and in vitro (118) had a missense mutation that caused a substitution of tryptophan by arginine at amino acid 286 of the VDR (102). This substitution in a normal size VDR abolished completely the binding of 1,25(OH)2D to its receptor. The tryptophan in this position is critical for the positioning of calcitriol in the VDR as was unveiled by the three-dimensional arrangement of the VDR and its ligand based on its crystal structure.

 

(ii) Defective Hormone Binding Capacity. In a patient representing one kindred, the number of binding sites in nuclei and soluble cell extracts was 10% of control, with an apparent normal affinity  (78,105,112). Recently, a boy with total alopecia, severe rickets and growth retardation was found to have two heterozygote different molecular defects in the ligand binding domain (9). The patient’s VDR had a low hormone binding capacity, 10% to 30% of controls, with normal affinity and a markedly deficient stimulation of 25(OH)D3-24-hydroxylase. The recreated mutations, each one tested separately in vitro, showed also deficient heterodimerization as well as different transactivation of two gene promoters. This patient, similar to another one described more than 20 years ago, could be completely cured by very high doses of 25(OH) vitamin D, 250 μg a day initially, followed by 100 μg/day and then 75 μg/day as a maintenance dose for years, plus modest calcium supplementation. In both patients, it could be shown that during remission (normocalcemia, normophosphatemia, normal iPTH), 1,25(OH)2D production is driven by the substrate, i.e., 25(OH) vitamin D concentrations.

 

An additional patient with hereditary resistance to 1,25(OH)2D and alopecia was found to have compound heterozygous mutations in the VDR (119). This girl’s cultured fibroblasts were found to have about 30% binding sites of 1,25(OH)2D in comparison to her parents or normal controls. However, RXR heterodimerization, co-activator interaction and gene transactivation were completely abolished, and no response in vitro to high doses of 1,25(OH)2D3 was observed. Treatment with 3,000mg of calcium carbonate orally per day and 3μg/kg of calcitriol caused some improvement in the clinical features and biochemical parameters.

 

(iii) Defective Hormone Binding Affinity. Binding affinity of tritiated calcitriol was reduced 20- to 30-fold, with normal capacity in soluble dermal fibroblast extracts from one kindred (98). An additional patient, representing a different kindred had a modest decrease of VDR affinity when measured at 0°C (120).

 

Deficient Nuclear Uptake. The following features characterize the hormone-receptor-nuclear interaction in this defect: normal or near normal binding capacity and affinity of 3H 1,25(OH)2D3 to soluble cell extracts with low to unmeasurable hormone uptake into nuclei of intact cells (87,90,105,121,122). These features were demonstrated in skin-derived fibroblasts in all kindreds, in cells cultured from a bone biopsy of one patient (94), and in EB-transformed lymphoblasts of one patient (121). Occupied VDR obtained from fibroblast extracts of two kindreds demonstrated normal binding to nonspecific DNA cellulose (80). Immuno-cytological studies in fibroblasts of a patient with this defect showed that immediately after 1,25(OH)2D3 treatment, VDR accumulated along the nuclear membrane with no nuclear translocation (123). Patients with this defect included a kindred with normal hair and several kindreds with total alopecia. Finally, almost all patients responded with a complete clinical remission to high doses of vitamin D and its active 1a-hydroxylated metabolites.

 

Attempts to characterize the molecular defect were carried in six kindreds. In three of them, no mutation in the coding region of the VDR gene was observed (121). Studies in three kindreds, revealed a normal molecular mass and quantitative expression of the VDR (122). Complete sequencing of the VDR coding region revealed a different single nucleotide mutation in each kindred in a region that is considered to play a role in heterodimerization of VDR with RXR (Figure 7) (115,122). Thus, it has been suggested that these patients’ receptors have defects that compromise RXR heterodimerization, which is essential for nuclear localization and probably for recognition of the vitamin D responsive element as well. The fact that no mutation in the VDR coding region was observed in three additional kindreds with the same phenotypical defect may suggest that the genetic defect affects another component of the receptor effector system that is essential for the VDR function as a nuclear transcription factor. It has been shown that coactivation complexes are essential for the ligand induced transactivation of VDR (124).

 

Deficient coactivators of the calcitriol-VDR complex. A patient with simple hereditary resistance to 1,25(OH)2D without alopecia was described (104). Sequencing of the VDR-DNA revealed a missense mutation in the ligand binding domain that caused a substitution of glutamic acid to lysine at amino acid 120. This receptor exhibits many normal properties including calcitriol binding, dimerization and binding to vitamin D response elements in the DNA, but a marked impairment in binding coactivators that are essential for the transactivation of the hormone receptor complex and the initiation of the physiological response (104).

 

Defects in the DNA Binding Region  

 

Deficient Binding to DNA. Cell preparations derived from patients with this defect demonstrate normal or near normal binding capacity and affinity for 3H 1,25(OH)2D3 to nuclei of intact cells and soluble cell extracts, as well as normal molecular size. Hormone receptor complexes, however, have decreased affinity to nonspecific DNA (88,89,97,108,109). A single nucleotide missense mutation within exon 2 or 3, encoding the DNA binding domain of the VDR, was demonstrated in genomic DNA isolated from dermal fibroblasts and/or EB-transformed lymphoblasts from ten unrelated kindreds (80,88,89,95,110,114,125,126). Eight different single nucleotide mutations were found in the ten kindreds (Figure 7). Two apparently unrelated kindreds share the same mutation (88,110). All point mutations caused a single substitution of an amino acid that resides in the region of the two zinc fingers of the VDR protein that are essential for the functional interaction of the hormone-receptor complex with DNA. Interestingly, all these altered amino acids are highly conserved in the steroid receptor superfamily that includes the receptors for steroid hormones, thyroid hormones and retinoic acid.

 

Studies on cells obtained from parents and some of the siblings of these patients revealed, as expected, a heterozygous state, i.e., expression of both normal and defective forms of VDR as well as normal and mutant gene sequences (80,102,109), but without any clinical and biochemical abnormalities.

 

In Vitro Post Transcriptional Effect of 1,25(OH)2D3

 

In vitro bioeffects of the hormone on various cells in patients with simple hereditary resistance to 1,25(OH)2D have been assayed mainly by two procedures, induction of 25(OH)D-24-hydroxylase and inhibition of mitogen stimulated PBM cells.

 

1,25(OH)2D3 induces 25(OH)D-24 hydroxylase activity in skin-derived fibroblasts (78,81,82,89,91-93,96-104,109,110,121,126,127), mitogen-stimulated lymphocytes (83), and cells originating from bone (94,128) in a dose-dependent manner. In cells from normal subjects, maximal and half-maximal induction of the enzyme was achieved by 10-8and 10-9M concentrations of 1,25(OH)2D3, respectively. Dermal fibroblast or PBM cells from patients with no calcemic response to maximal doses of vitamin D or its metabolites in vivo did not show any 25(OH)D-24-hydroxylase response to very high concentration of 1,25(OH)2D3 in vitro, while dermal fibroblasts from patients with a calcemic response to high doses of vitamin D or its metabolites in vivo showed inducible 24-hydroxylase with supraphysiological concentrations of 1,25(OH)2D3 in vitro. Physiological concentrations of 1,25(OH)2D3 partially inhibit mitogen-induced DNA synthesis in peripheral lymphocytes with a half maximal inhibition achieved at 10-10M of the hormone (85,107). Mitogen stimulated lymphocytes from several kindreds with defects characterized as no hormone binding or deficient binding to DNA, with no calcemic response to high doses of vitamin D and its metabolites in vivo, showed no inhibition of lymphocyte proliferation in vitro with concentrations of up to 10-6M 1,25(OH)2D3. Additional methods to measure bioeffects of 1,25(OH)2D3 on various cells in vitro were carried out only in a few patients and included inhibition of dermal fibroblast proliferation (93), induction of osteocalcin synthesis in cells derived from bone (128), a mitogenic effect on dermal fibroblasts (123), and stimulation of cGMP production in cultured skin fibroblasts (129). It is noteworthy that in all assays mentioned and without exception, each patient’s cell showed severely deficient responses.

 

With the elucidation of the molecular defects in simple hereditary resistance to 1,25(OH)2D, the transactivation abilities of naturally occurring mutant or recreated mutant VDRs were evaluated in a transcriptional activation assay. In this assay a gene promoter responsive to VDR is fused to a gene reporter that its message is easily measured, and the plasmid is transfected into patients or normal cells (81,82,88,89,109,116,121). Treating normal transfected cells with 1,25(OH)2D3 caused a concentration-dependent induction of transcription. No induction of transcription was observed in cells originating from patients with defects characterized as no hormone binding (130,131)(116) or deficient binding to DNA (88,89,109,110). Moreover, in a cotransfection assay, the addition of a normal human VDR cDNA expression vector to the transfected plasmid that directed synthesis of a normal VDR, restored hormone responsiveness of resistant cells. Finally, in a patient characterized as deficient nuclear uptake defect, no mutation was identified within the coding region of the VDR gene; no induction of 25(OH)D-24-hydroxylase activity by up to 10-6 1,25(OH)2D3 was observed in cultured skin fibroblasts, but there was normal transactivation by 1,25(OH)2D3 in the transcriptional activation assay (121).

 

CELLULAR DEFECTS AND CLINICAL FEATURES

 

Normal hair was described with most phenotypes of the cellular defects, the exception being patients with deficient hormone binding capacity and affinity, but this could be due to the fact that only one or two kindreds were described per subgroup. Normal hair is usually associated with a milder form of the disease, as judged by the age of onset, severity of the clinical features, and usually the complete clinical and biochemical remission on high doses of vitamin D or its metabolites. Notable exceptions are 3 kindreds (4 patients), two of them without alopecia that displayed resistance both in vivo (no clinical remission on circulating calcitriol level up to 100 times the mean normal adult values) and in vitro (no induction of 25(OH)D-24-hydroxylase activity in dermal fibroblasts by up to 10-8 M 1,25(OH)2D3) (99,102,104). Only approximately half of the patients with alopecia have shown satisfactory clinical and biochemical remission to high doses of vitamin D or its active 1a-hydroxylated metabolites, but the dose requirement is ~10-fold higher than in patients with normal hair (70).

 

It seems that patients’ defects characterized as deficient hormone binding affinity and deficient nuclear uptake achieve complete clinical and biochemical remission on high doses of vitamin D or its active 1a-hydroxylated metabolites. Most of the patients with other types of defects could not be cured with high doses of vitamin D or its metabolites. However, it should be emphasized that not all of the patients received treatment for a long enough period of time and with sufficiently high doses (see Treatment, below).

 

DIAGNOSIS

 

Clinical features of early onset rickets with no history of vitamin D deficiency, total alopecia, parental consanguinity, additional siblings with the same disease, serum biochemistry of hypocalcemic rickets, elevated circulating levels of 1,25(OH)2D, and normal to high levels of 25(OH)D (Table 2) support the diagnosis of simple hereditary resistance to 1,25(OH)2D. The issue becomes more complicated when the clinical features are atypical, i.e., late onset of the disease, sporadic cases, and normal hair. Failure of a therapeutic trial with calcium and/or physiological replacement doses of vitamin D or its metabolites may support the diagnosis but the final direct proof requires the demonstration of a cellular, molecular, and functional defect in the VDR-effector system.

 

Based on the clinical and biochemical features, the following additional disease states should be considered: (1) extreme calcium deficiency: a seemingly rare situation described in a group of children from a rural community in South Africa, who consumed an exceptionally low calcium diet of 125 mg/day (20). All had severe bone disease with histologically proven osteomalacia, biochemical features of hypocalcemic rickets with elevated serum levels of 1,25(OH)2D, and sufficient vitamin D. Calcium repletion caused complete clinical and biochemical remission. Nutritional history and the response to calcium supplementation support this diagnosis. (2) Severe vitamin D deficiency: during initial stages of vitamin D therapy in children with severe vitamin D-deficient rickets, the biochemical picture may resemble 1,25(OH)2D resistance, i.e., hypocalcemic rickets with elevated serum calcitriol levels. This may represent a “hungry bone syndrome,” i.e., high calcium demands of the abundant osteoid tissue becoming mineralized. This is a transient condition that may be differentiated from simple hereditary resistance to 1,25(OH)2D by a history of vitamin D deficiency and the final therapeutic response to replacement doses of vitamin D.

 

TREATMENT

 

In about half of the kindreds with simple hereditary resistance to 1,25(OH)2D, the bioeffects of 1,25(OH)2D3 were measured in vitro (see above). An invariable correlation (with one exception) was documented between the in vitro effect and the therapeutic response in vivo; i.e., patients with no calcemic response to high levels of serum calcitriol showed no effects of 1,25(OH)2D3 on their cells in vitro (either induction of 25(OH))D-24-hydroxylase or inhibition of lymphocyte proliferation) and vice versa. If the predictive therapeutic value of the in vitro cellular response to 1,25(OH)2D3 could be substantiated convincingly, it may eliminate the need for time consuming and expensive therapeutic trials with massive doses of vitamin D or its active metabolites. In the meantime, it is mandatory to treat every patient with this disease irrespective of the type of receptor defect.

 

An adequate therapeutic trial must include vitamin D at a dose that is sufficient to maintain high serum concentrations of 1,25(OH)2D3 as the patients can produce high hormone levels if supplied with enough substrate. If high serum calcitriol levels are not achieved, it is advisable to treat with 1a-hydroxylated vitamin D metabolites in daily doses of up to 6 mg/kg weight or a total of 30-60 mg and calcium supplementation of up to 3 g of elemental calcium daily; therapy must be maintained for a period sufficient to mineralize the abundant osteoid (usually 3-5 months). Therapy may be considered a failure if no change in the clinical, radiological, or biochemical parameters occurs during continuous and frequent follow up while serum 1,25(OH)2D concentrations are maintained at ~100 times the mean normal range.

 

In some patients with no response to adequate therapeutic trials with vitamin D or its metabolites, a remarkable clinical and biochemical remission of their bone disease, including catch-up growth, was obtained by treatment with large amounts of calcium as noted previously. This was achieved by long-term (months) intracaval infusions of up to 1000 mg of calcium daily (102,128,132-134). Another way to increase calcium input into the extra cellular compartment is to increase net gut absorption, independent of vitamin D, by increasing calcium intake (135). This approach is limited by dose and patient tolerability and was actually used successfully in only a few patients.

 

Several patients have shown unexplained fluctuations in response to therapy or in presentation of the disease. One patient after a prolonged remission became completely unresponsive to much higher doses of active 1a-hydroxylated vitamin D metabolites (78), and another patient seemed to show amelioration of resistance to 1,25(OH)2D3 after a brief therapeutic trial with 24,25(OH)2D (68). In several patients, spontaneous healing occurred in their teens (106) or rickets did not recur for 14 years after cessation of therapy (87).

 

ANIMAL MODELS

 

Some New World primates (marmoset and tamarins) that develop osteomalacia in captivity are known to have high nutritional requirements for vitamin D and maintain high serum levels of 1,25(OH)2D, thus exhibiting a form of end-organ resistance to 1,25(OH)2D (136-139). Cultured dermal fibroblasts and EB-virus transformed lymphoblast have shown deficient hormone binding capacity and affinity (136,137). It has been observed that marmoset lymphoblasts contain a soluble protein of 50-60 kDa that binds 1,25(OH)2D3 with a low affinity but high capacity and thus may serve as a sink that interferes with the hormone binding and its cognate receptor (140). The same group described another protein present in the nuclear extract of these cells capable of inhibiting normal VDR-RXR binding to the vitamin D response element (141). It is of interest that these New World primates also exhibit a compensated hereditary end-organ resistance to the true steroid hormones including glucocorticoids, estrogens, and progestins (142). This of course raises the interesting possibility that the defect in the hormone-receptor-effector system involves an element shared by all the members of this superfamily of ligand modulated transcription factors. Subsequent studies have identified the proteins involved as members of the heterogeneous nuclear ribonucleoprotein family (143-145).

 

VDR knock out mice have been created by targeted ablation of the first or second zinc finger (75,76). Only the homozygote mice were affected. Though phenotypically normal at birth, after weaning however, they become hypocalcemic, develop secondary hyperparathyroidism, rickets, osteomalacia and progressive alopecia. The female mice with ablation of the first zinc finger are infertile and show uterine hypoplasia and impaired folliculogenesis. Otherwise, both VDR cell mutant mice show clinical, radiological, histological and biochemical features that are identical to the human disease. Supplementation with a calcium enriched diet can prevent or treat most of the disturbances in mineral and bone metabolism in these animal models except alopecia (77).

 

It is of interest that targeting expression of the human VDR to keratinocytes of VDR null mice prevented alopecia without correcting the mineral disorder (146). Further evidence that the role of VDR in hair follicle cycling is ligand independent comes from the observation that mice in which Cyp27b1, the enzyme solely responsible for producing 1,25(OH)2D, has been deleted do not lose hair (147)

 

CONCLUDING REMARKS

 

Acquired vitamin D deficiency, especially moderate or mild (sometimes referred to as vitamin D insufficiency), is much more common than previously appreciated. It may affect more than 50% of populations with limited sunshine exposure and no vitamin D supplementation. In the less severe forms of vitamin D deficiency, there is no defective bone matrix mineralization, but an increased bone loss secondary to the perturbations in calcium homeostasis and secondary hyperparathyroidism, which accelerates the development of osteoporosis in post-menopausal women, the elderly population at large in general, and some subgroups in particular such as those on drugs that contribute to bone loss or deficient in calcium intake. Osteomalacia, which marks severe vitamin D deficiency, may affect some of these patients as well. Based on these observations, it is highly recommended for anyone taking care of the elderly, especially the house or bed bound, institutionalized or patients with physical or mental deficiencies that limit their free movement, to consider and evaluate their vitamin D status. It seems to be good clinical practice and cost effective to recommend vitamin D supplementation for populations at risk, as a measure to prevent the deleterious effects of vitamin D deficiency on the musculoskeletal system.

 

Hereditary deficiencies in vitamin D action are rare disorders. The importance of studying these diseases stems from the fact that they represent a naturally occurring experimental model that helps to elucidate the function and importance of vitamin D and the VDR-effector system in human beings in vivo.

 

VDRs are abundant and widely distributed among most tissues studied and multiple effects of calcitriol are observed on various cell functions in vitro. Yet, the clinical and biochemical features in patients with simple hereditary 1,25(OH)2D deficiency and resistance seems to demonstrate that the most important disturbances of clinical relevance are perturbations in mineral and bone metabolism. This emphasizes the pivotal role of 1,25(OH)2D in transepithelial net calcium fluxes. Moreover, the fact that in patients with extreme end-organ resistance to calcitriol, calcium infusions correct the disturbances in mineral homeostasis and cure the bone disease may support the notion that defective bone matrix mineralization is secondary to disturbances in mineral homeostasis. That said, the widespread distribution of the VDR and the enzymes metabolizing vitamin D to its active metabolites have led to numerous studies into its non-skeletal actions with the hope that vitamin D may play a role in the treatment of diseases such as cancer, heart disease, respiratory illness, inflammatory diseases, and diabetes. Characterization of the molecular, cellular, and functional defects of the different natural mutants of the human VDR in simple hereditary 1,25(OH)2D deficiency and resistance, demonstrates the essentiality of the VDR as the mediator of calcitriol action and the importance and function of its different domains, i.e., binding of the hormone, an RXR isoform, and binding to a specifically defined DNA region, as well as coactivators and corepressor complexes. Thus, studies in patients with hereditary deficiencies in vitamin D action are the essential link between molecular defects and physiological relevant effects in human beings.

 

REFERENCES

 

  1. Ohyama Y, Noshiro M, Eggertsen G, Gotoh O, Kato Y, Bjoerkhem I, Okuda K. Structural characterization of the gene encoding rat 25-hydroxyvitamin D3 24-hydroxylase. Biochemistry. 1993;32(1):76-82.
  2. Akeno N, Saikatsu S, Kawane T, Horiuchi N. Mouse Vitamin D-24-Hydroxylase: Molecular Cloning, Tissue Distribution, and Transcriptional Regulation by 1α,25-Dihydroxyvitamin D3*. Endocrinology. 1997;138(6):2233-2240.
  3. Fu GK, Portale AA, Miller WL. Complete Structure of the Human Gene for the Vitamin D 1α-Hydroxylase, P450c1α. DNA and Cell Biology. 1997;16(12):1499-1507.
  4. Monkawa T, Yoshida T, Wakino S, Shinki T, Anazawa H, Deluca HF, Suda T, Hayashi M, Saruta T. Molecular Cloning of cDNA and Genomic DNA for Human 25-hydroxyvitamin D31α-hydroxylase. Biochemical and Biophysical Research Communications. 1997;239(2):527-533.
  5. Shinki T, Shimada H, Wakino S, Anazawa H, Hayashi M, Saruta T, DeLuca HF, Suda T. Cloning and expression of rat 25-hydroxyvitamin D3-1alpha-hydroxylase cDNA. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(24):12920-12925.
  6. Takeyama K-i, Kitanaka S, Sato T, Kobori M, Yanagisawa J, Kato S. 25-Hydroxyvitamin D <sub>3</sub> 1α-Hydroxylase and Vitamin D Synthesis. Science. 1997;277(5333):1827-1830.
  7. Jehan F, Ismail R, Hanson K, DeLuca HF. Cloning and expression of the chicken 25-hydroxyvitamin D3 24-hydroxylase cDNA. Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression. 1998;1395(3):259-265.
  8. Jones G, Ramshaw H, Zhang A, Cook R, Byford V, White J, Petkovich M. Expression and Activity of Vitamin D-Metabolizing Cytochrome P450s (CYP1α and CYP24) in Human Nonsmall Cell Lung Carcinomas*. Endocrinology. 1999;140(7):3303-3310.
  9. Nguyen M, d'Alesio A, Pascussi JM, Kumar R, Griffin MD, Dong X, Guillozo H, Rizk-Rabin M, Sinding C, Bougnères P, Jehan F, Garabédian M. Vitamin D-Resistant Rickets and Type 1 Diabetes in a Child With Compound Heterozygous Mutations of the Vitamin D Receptor (L263R and R391S): Dissociated Responses of the CYP-24 and rel-B Promoters to 1,25-Dihydroxyvitamin D3. Journal of Bone and Mineral Research.2006;21(6):886-894.
  10. Bouillon R, Bikle D. Vitamin D Metabolism Revised: Fall of Dogmas. J Bone Miner Res. 2019;34(11):1985-1992.
  11. Roizen JD, Long C, Casella A, O'Lear L, Caplan I, Lai M, Sasson I, Singh R, Makowski AJ, Simmons R, Levine MA. Obesity Decreases Hepatic 25-Hydroxylase Activity Causing Low Serum 25-Hydroxyvitamin D. J Bone Miner Res. 2019;34(6):1068-1073.
  12. Evans RM. The steroid and thyroid hormone receptor superfamily. Science. 1988;240(4854):889-895.
  13. Baker AR, McDonnell DP, Hughes M, Crisp TM, Mangelsdorf DJ, Haussler MR, Pike JW, Shine J, O'Malley BW. Cloning and expression of full-length cDNA encoding human vitamin D receptor. Proceedings of the National Academy of Sciences of the United States of America. 1988;85(10):3294-3298.
  14. Burmester JK, Maeda N, DeLuca HF. Isolation and expression of rat 1,25-dihydroxyvitamin D3 receptor cDNA. Proceedings of the National Academy of Sciences of the United States of America. 1988;85(4):1005-1009.
  15. Ozono K, Sone T, Pike JW. Perspectives: The genomic mechanism of action of 1,25-dihydroxyvitamin D3. Journal of Bone and Mineral Research. 2009;6(10):1021-1027.
  16. Deluca HF. The Metabolism, Physiology, and Function of Vitamin D. Vitamin D: Springer US; 1984:1-68.
  17. Mayer E, Kadowaki S, Williams G, Norman AW. Mode of Action of 1α,25-Dihydroxyvitamin D. Vitamin D: Springer US; 1984:259-302.
  18. Reichel H, Koeffler HP, Norman AW. The Role of the Vitamin D Endocrine System in Health and Disease. New England Journal of Medicine. 1989;320(15):980-991.
  19. Owen TA, Aronow MS, Barone LM, Bettencourt B, Stein GS, Lian JB. Pleiotropic Effects of Vitamin D on Osteoblast Gene Expression Are Related to the Proliferative and Differentiated State of the Bone Cell Phenotype: Dependency upon Basal Levels of Gene Expression, Duration of Exposure, and Bone Matrix Competency in Normal Rat Osteoblast Cultures*. Endocrinology. 1991;128(3):1496-1504.
  20. Pettifor JM, Ross FP, Travers R, Glorieux FH, Deluca HF. Dietary calcium deficiency : A syndrome associated with bone deformities and elevated serum 1,25-Dihyroxyvitamin D concentrations. Metabolic Bone Disease and Related Research. 1981;2(5):301-305.
  21. Teitelbaum SL, Bullough PG. The pathophysiology of bone and joint disease. Am J Pathol. 1979;96(1):282-354.
  22. Parfitt AM, Drezner MK, Glorieux FH, Kanis JA, Malluche H, Meunier PJ, Ott SM, Recker RR. Bone histomorphometry: Standardization of nomenclature, symbols, and units: Report of the asbmr histomorphometry nomenclature committee. Journal of Bone and Mineral Research. 2009;2(6):595-610.
  23. Cole DEC, Carpenter TO, Gundberg CM. Serum osteocalcin concentrations in children with metabolic bone disease. The Journal of Pediatrics. 1985;106(5):770-776.
  24. Roizen JD, Levine MA. Response to: Obesity and Vitamin D Metabolism Modifications. J Bone Miner Res.2019;34(7):1384.
  25. Bikle DD. Vitamin D: Newer Concepts of Its Metabolism and Function at the Basic and Clinical Level. J Endocr Soc. 2020;4(2):bvz038.
  26. Holick MF. McCollum Award Lecture, 1994: Vitamin D—new horizons for the 21st century. The American Journal of Clinical Nutrition. 1994;60(4):619-630.
  27. Holick MF. Environmental factors that influence the cutaneous production of vitamin D. The American Journal of Clinical Nutrition. 1995;61(3):638S-645S.
  28. Lips P. Vitamin D Deficiency and Secondary Hyperparathyroidism in the Elderly: Consequences for Bone Loss and Fractures and Therapeutic Implications. Endocrine Reviews. 2001;22(4):477-501.
  29. Bischoff-Ferrari HA, Giovannucci E, Willett WC, Dietrich T, Dawson-Hughes B. Estimation of optimal serum concentrations of 25-hydroxyvitamin D for multiple health outcomes. The American Journal of Clinical Nutrition.2006;84(1):18-28.
  30. Bischoff-Ferrari HA, Willett WC, Wong JB, Stuck AE, Staehelin HB, Orav EJ, Thoma A, Kiel DP, Henschkowski J. Prevention of Nonvertebral Fractures With Oral Vitamin D and Dose Dependency. Archives of Internal Medicine. 2009;169(6):551.
  31. Krall EA, Sahyoun N, Tannenbaum S, Dallal GE, Dawson-Hughes B. Effect of Vitamin D Intake on Seasonal Variations in Parathyroid Hormone Secretion in Postmenopausal Women. New England Journal of Medicine.1989;321(26):1777-1783.
  32. Thomas MK, Lloyd-Jones DM, Thadhani RI, Shaw AC, Deraska DJ, Kitch BT, Vamvakas EC, Dick IM, Prince RL, Finkelstein JS. Hypovitaminosis D in Medical Inpatients. New England Journal of Medicine.1998;338(12):777-783.
  33. Lips P, Duong T, Oleksik A, Black D, Cummings S, Cox D, Nickelsen T. A Global Study of Vitamin D Status and Parathyroid Function in Postmenopausal Women with Osteoporosis: Baseline Data from the Multiple Outcomes of Raloxifene Evaluation Clinical Trial. The Journal of Clinical Endocrinology &amp; Metabolism.2001;86(3):1212-1221.
  34. Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 Dietary Reference Intakes for Calcium and Vitamin D: What Dietetics Practitioners Need to Know⁎⁎This article is a summary of the Institute of Medicine report entitled Dietary Reference Intakes for Calcium and Vitamin D (available at http://www.iom.edu/Reports/2010/Dietary-Reference-Intakes-for-Calcium-and-Vitamin-D.aspx) for dietetics practitioners; a similar summary for clinicians has also been published (Ross AC, Manson JE, Abrams SA, Aloia JF, Brannon PM, Clinton SK, Durazo-Arvizu RA, Gallagher JC, Gallo RL, Jones G, Kovacs CS, Mayne ST, Rosen CJ, Shapses SA. The 2011 report on Dietary Reference Intakes for calcium and vitamin D from the Institute of Medicine: What clinicians need to know. J Clin Endocrinol Metab. 2011;96:53-58). Journal of the American Dietetic Association. 2011;111(4):524-527.
  35. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP, Murad MH, Weaver CM. Evaluation, Treatment, and Prevention of Vitamin D Deficiency: an Endocrine Society Clinical Practice Guideline. The Journal of Clinical Endocrinology &amp; Metabolism. 2011;96(7):1911-1930.
  36. Khaw KT, Sneyd MJ, Compston J. Bone density parathyroid hormone and 25-hydroxyvitamin D concentrations in middle aged women. BMJ. 1992;305(6848):273-277.
  37. Mezquita-Raya P, Muñoz-Torres M, De Dios Luna J, Luna V, Lopez-Rodriguez F, Torres-Vela E, Escobar-Jiménez F. Relation Between Vitamin D Insufficiency, Bone Density, and Bone Metabolism in Healthy Postmenopausal Women. Journal of Bone and Mineral Research. 2001;16(8):1408-1415.
  38. Collins D, Jasani C, Fogelman I, Swaminathan R. Vitamin D and bone mineral density. Osteoporosis International. 1998;8(2):110-114.
  39. Chapuy MC, Arlot ME, Duboeuf F, Brun J, Crouzet B, Arnaud S, Delmas PD, Meunier PJ. Vitamin D<sub>3</sub>and Calcium to Prevent Hip Fractures in Elderly Women. New England Journal of Medicine.1992;327(23):1637-1642.
  40. Davies M, Mawer EB, Hann JT, Stephens WP, Taylor JL. VITAMIN D PROPHYLAXIS IN THE ELDERLY: A SIMPLE EFFECTIVE METHOD SUITABLE FOR LARGE POPULATIONS. Age and Ageing. 1985;14(6):349-354.
  41. Weisman Y, Schen RJ, Eisenberg Z, Amarilio N, Graff E, Edelstein-Singer M, Goldray D, Harell A. Single Oral High-Dose Vitamin D<sub>3</sub>Prophylaxis in the Elderly. Journal of the American Geriatrics Society.1986;34(7):515-518.
  42. Khaw KT, Scragg R, Murphy S. Single-dose cholecalciferol suppresses the winter increase in parathyroid hormone concentrations in healthy older men and women: a randomized trial. The American Journal of Clinical Nutrition. 1994;59(5):1040-1044.
  43. Heikinheimo RJ, Inkovaara JA, Harju EJ, Haavisto MV, Kaarela RH, Kataja JM, Kokko AM-L, Kolho LA, Rajala SA. Annual injection of vitamin D and fractures of aged bones. Calcified Tissue International. 1992;51(2):105-110.
  44. Prader A, Illig R, Heierli E. [An unusual form of primary vitamin D-resistant rickets with hypocalcemia and autosomal-dominant hereditary transmission: hereditary pseudo-deficiency rickets]. Helv Paediatr Acta.1961;16:452-468.
  45. Fraser D, Kooh SW, Kind HP, Holick MF, Tanaka Y, DeLuca HF. Pathogenesis of Hereditary Vitamin-D-Dependent Rickets. New England Journal of Medicine. 1973;289(16):817-822.
  46. Delvin EE, Glorieux FH, Marie PJ, Pettifor JM. Vitamin D dependency: Replacement therapy with calcitriol. The Journal of Pediatrics. 1981;99(1):26-34.
  47. Labuda M, Morgan K, Glorieux FH. Mapping autosomal recessive vitamin D dependency type I to chromosome 12q14 by linkage analysis. Am J Hum Genet. 1990;47(1):28-36.
  48. Braekeleer MD, Larochelle J. Population genetics of vitamin D-dependent rickets in northeastern Quebec. Annals of Human Genetics. 1991;55(4):283-290.
  49. Fu GK, Lin D, Zhang MYH, Bikle DD, Shackleton CHL, Miller WL, Portale AA. Cloning of Human 25-Hydroxyvitamin D-1α-Hydroxylase and Mutations Causing Vitamin D-Dependent Rickets Type 1. Molecular Endocrinology. 1997;11(13):1961-1970.
  50. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, Glorieux FH. The 25-Hydroxyvitamin D 1-Alpha-Hydroxylase Gene Maps to the Pseudovitamin D-Deficiency Rickets (PDDR) Disease Locus. Journal of Bone and Mineral Research. 1997;12(10):1552-1559.
  51. Wang JT, Lin CJ, Burridge SM, Fu GK, Labuda M, Portale AA, Miller WL. Genetics of vitamin D 1alpha-hydroxylase deficiency in 17 families. Am J Hum Genet. 1998;63(6):1694-1702.
  52. Yoshida T, Monkawa T, Tenenhouse HS, Goodyer P, Shinki T, Suda T, Wakino S, Hayashi M, Saruta T. Two novel 1α-hydroxylase mutations in French-Canadians with vitamin D dependency rickets type I. Kidney International. 1998;54(5):1437-1443.
  53. Kitanaka S, Takeyama K-i, Murayama A, Sato T, Okumura K, Nogami M, Hasegawa Y, Niimi H, Yanagisawa J, Tanaka T, Kato S. Inactivating Mutations in the 25-Hydroxyvitamin D<sub>3</sub>1α-Hydroxylase Gene in Patients with Pseudovitamin D–Deficiency Rickets. New England Journal of Medicine. 1998;338(10):653-662.
  54. Kitanaka S, Murayama A, Sakaki T, Inouye K, Seino Y, Fukumoto S, Shima M, Yukizane S, Takayanagi M, Niimi H, Takeyama K-i, Kato S. No Enzyme Activity of 25-Hydroxyvitamin D3 1α-Hydroxylase Gene Product in Pseudovitamin D Deficiency Rickets, Including That with Mild Clinical Manifestation. The Journal of Clinical Endocrinology &amp; Metabolism. 1999;84(11):4111-4117.
  55. Smith SJ, Rucka AK, Berry JL, Davies M, Mylchreest S, Paterson CR, Heath DA, Tassabehji M, Read AP, Mee AP, Mawer EB. Novel Mutations in the 1α-Hydroxylase (P450c1) Gene in Three Families with Pseudovitamin D-Deficiency Rickets Resulting in Loss of Functional Enzyme Activity in Blood-Derived Macrophages. Journal of Bone and Mineral Research. 1999;14(5):730-739.
  56. Fu GK, Lin D, Zhang MY, Bikle DD, Shackleton CH, Miller WL, Portale AA. Cloning of human 25-hydroxyvitamin D-1 alpha-hydroxylase and mutations causing vitamin D-dependent rickets type 1. Mol Endocrinol. 1997;11(13):1961-1970.
  57. Glorieux FH, Arabian A, Delvin EE. Pseudo-vitamin D deficiency: absence of 25-hydroxyvitamin D 1 alpha-hydroxylase activity in human placenta decidual cells. The Journal of Clinical Endocrinology &amp; Metabolism. 1995;80(7):2255-2258.
  58. Kitanaka S, Takeyama K, Murayama A, Sato T, Okumura K, Nogami M, Hasegawa Y, Niimi H, Yanagisawa J, Tanaka T, Kato S. Inactivating mutations in the 25-hydroxyvitamin D3 1alpha-hydroxylase gene in patients with pseudovitamin D-deficiency rickets. N Engl J Med. 1998;338(10):653-661.
  59. Yoshida T, Monkawa T, Tenenhouse HS, Goodyer P, Shinki T, Suda T, Wakino S, Hayashi M, Saruta T. Two novel 1alpha-hydroxylase mutations in French-Canadians with vitamin D dependency rickets type I1. Kidney Int. 1998;54(5):1437-1443.
  60. Kitanaka S, Murayama A, Sakaki T, Inouye K, Seino Y, Fukumoto S, Shima M, Yukizane S, Takayanagi M, Niimi H, Takeyama K, Kato S. No enzyme activity of 25-hydroxyvitamin D3 1alpha-hydroxylase gene product in pseudovitamin D deficiency rickets, including that with mild clinical manifestation. J Clin Endocrinol Metab.1999;84(11):4111-4117.
  61. Smith SJ, Rucka AK, Berry JL, Davies M, Mylchreest S, Paterson CR, Heath DA, Tassabehji M, Read AP, Mee AP, Mawer EB. Novel mutations in the 1alpha-hydroxylase (P450c1) gene in three families with pseudovitamin D-deficiency rickets resulting in loss of functional enzyme activity in blood-derived macrophages. J Bone Miner Res. 1999;14(5):730-739.
  62. Winkler I, von Grabe C, Harmeyer J. Pseudo vitamin D deficiency rickets in pigs: in vitro measurements of renal 25-hydroxycholecalciferol-1-hydroxylase activity. Zentralbl Veterinarmed A. 1982;29(2):81-88.
  63. Fox J, Maunder EMW, Randall VA, Care AD. Vitamin D-dependent rickets type I in pigs. Clinical Science.1985;69(5):541-548.
  64. Dardenne O, Prud’homme Je, Arabian A, Glorieux FH, St-Arnaud R. Targeted Inactivation of the 25-Hydroxyvitamin D3-1α-Hydroxylase Gene (CYP27B1) Creates an Animal Model of Pseudovitamin D-Deficiency Rickets*. Endocrinology. 2001;142(7):3135-3141.
  65. Malloy PJ, Tasic V, Taha D, Tutunculer F, Ying GS, Yin LK, Wang J, Feldman D. Vitamin D receptor mutations in patients with hereditary 1,25-dihydroxyvitamin D-resistant rickets. Mol Genet Metab. 2014;111(1):33-40.
  66. Brooks MH, Bell NH, Love L, Stern PH, Orfei E, Queener SF, Hamstra AJ, DeLuca HF. Vitamin-D-Dependent Rickets Type II. New England Journal of Medicine. 1978;298(18):996-999.
  67. Fujita T, Nomura M, Okajima S, Furuya H. Adult-Onset Vitamin D-Resistant Osteomalacia with the Unresponsiveness to Parathyroid Hormone. The Journal of Clinical Endocrinology &amp; Metabolism.1980;50(5):927-931.
  68. Liberman UA, Halabe A, Samuel R, Kauli R, Edelstein S, Weisman Y, Papapoulos SE, Fraher LJ, Clemens TL, O'Riordan JLH. END-ORGAN RESISTANCE TO 1,25-DIHYDROXYCHOLECALCIFEROL. The Lancet.1980;315(8167):504-507.
  69. Marx SJ, Liberman UA, Eil C, Gamblin GT, Degrange DA, Balsan S. Hereditary Resistance to 1,25-Dihydroxyvitamin D. Proceedings of the 1983 Laurentian Hormone Conference: Elsevier; 1984:589-620.
  70. Marx SJ, Bliziotes MM, Nanes M. ANALYSIS OF THE RELATION BETWEEN ALOPECIA AND RESISTANCE TO 1,25-DIHYDROXYVITAMIN D. Clinical Endocrinology. 1986;25(4):373-381.
  71. Bikle DD, Chang S, Crumrine D, Elalieh H, Man MQ, Choi EH, Dardenne O, Xie Z, Arnaud RS, Feingold K, Elias PM. 25 Hydroxyvitamin D 1 α-Hydroxylase Is Required for Optimal Epidermal Differentiation and Permeability Barrier Homeostasis. Journal of Investigative Dermatology. 2004;122(4):984-992.
  72. Stumpf WE, Sar M, Reid FA, Tanaka Y, DeLuca HF. Target Cells for 1,25-Dihydroxyvitamin D <sub>3</sub> in Intestinal Tract, Stomach, Kidney, Skin, Pituitary, and Parathyroid. Science. 1979;206(4423):1188-1190.
  73. Hsieh JC, Sisk JM, Jurutka PW, Haussler CA, Slater SA, Haussler MR, Thompson CC. Physical and functional interaction between the vitamin D receptor and hairless corepressor, two proteins required for hair cycling. J Biol Chem. 2003;278(40):38665-38674.
  74. Balsan S, Garabedian M, Larchet M, Gorski AM, Cournot G, Tau C, Bourdeau A, Silve C, Ricour C. Long-term nocturnal calcium infusions can cure rickets and promote normal mineralization in hereditary resistance to 1,25-dihydroxyvitamin D. J Clin Invest. 1986;77(5):1661-1667.
  75. Li YC, Pirro AE, Amling M, Delling G, Baron R, Bronson R, Demay MB. Targeted ablation of the vitamin D receptor: an animal model of vitamin D-dependent rickets type II with alopecia. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(18):9831-9835.
  76. Yoshizawa T, Handa Y, Uematsu Y, Takeda S, Sekine K, Yoshihara Y, Kawakami T, Arioka K, Sato H, Uchiyama Y, Masushige S, Fukamizu A, Matsumoto T, Kato S. Mice lacking the vitamin D receptor exhibit impaired bone formation, uterine hypoplasia and growth retardation after weaning. Nature Genetics.1997;16(4):391-396.
  77. Li YC, Amling M, Pirro AE, Priemel M, Meuse J, Baron R, Delling G, Demay MB. Normalization of mineral ion homeostasis by dietary means prevents hyperparathyroidism, rickets, and osteomalacia, but not alopecia in vitamin D receptor-ablated mice. Endocrinology. 1998;139(10):4391-4396.
  78. Balsan S, Garabedian M, Liberman UA, Bourdeau A, Eil C, Guillozo H, Grimberg R, Deunff MJL, Lieberherr M, Guimbaud P, Broyer M, Marx SJ. Rickets and Alopecia with Resistance to 1,25-Dihydroxyvitamin D: Two Different Clinical Courses with Two Different Cellular Defects*. The Journal of Clinical Endocrinology &amp; Metabolism. 1983;57(4):803-811.
  79. Murayama A, Takeyama K-i, Kitanaka S, Kodera Y, Kawaguchi Y, Hosoya T, Kato S. Positive and Negative Regulations of the Renal 25-Hydroxyvitamin D3 1α-Hydroxylase Gene by Parathyroid Hormone, Calcitonin, and 1α,25(OH)2D3 in Intact Animals*. Endocrinology. 1999;140(5):2224-2231.
  80. Malloy PJ, Hochberg Z, Pike JW, Feldman D. Abnormal Binding of Vitamin D Receptors to Deoxyribonucleic Acid in a Kindred With Vitamin D-Dependent Rickets, Type II*. The Journal of Clinical Endocrinology &amp; Metabolism. 1989;68(2):263-269.
  81. Malloy PJ, Hochberg Z, Tiosano D, Pike JW, Hughes MR, Feldman D. The molecular basis of hereditary 1,25-dihydroxyvitamin D3 resistant rickets in seven related families. The Journal of clinical investigation.1990;86(6):2071-2079.
  82. Ritchie HH, Hughes MR, Thompson ET, Malloy PJ, Hochberg Z, Feldman D, Pike JW, O'Malley BW. An ochre mutation in the vitamin D receptor gene causes hereditary 1,25-dihydroxyvitamin D3-resistant rickets in three families. Proceedings of the National Academy of Sciences of the United States of America. 1989;86(24):9783-9787.
  83. Takeda E, Yokota I, Ito M, Kobashi H, Saijo T, Kuroda Y. 25-Hydroxyvitamin D-24-Hydroxylase in Phytohemagglutinin-Stimulated Lymphocytes: Intermediate Bioresponse to 1,25-Dihydroxyvitamin D<sub>3</sub>of Cells from Parents of Patients with Vitamin DDependent Rickets type II*. The Journal of Clinical Endocrinology &amp; Metabolism. 1990;70(4):1068-1074.
  84. Tsuchiya Y, Matsuo N, Cho H, Kumagai M, Yasaka A, Suda T, Orimo H, Shiraki M. An Unusual Form of Vitamin D-Dependent Rickets in a Child: Alopecia and Marked End-Organ Hyposensitivity to Biologically Active Vitamin D. The Journal of Clinical Endocrinology &amp; Metabolism. 1980;51(4):685-690.
  85. Takeda E, Kuroda Y, Saijo T, Toshima K, Naito E, Kobashi H, Iwakuni Y, Miyao M. Rapid diagnosis of vitamin D-dependent rickets type II by use of phytohemagglutinin-stimulated lymphocytes. Clinica Chimica Acta.1986;155(3):245-250.
  86. Takeda E, Kuroda Y, Saijo T, Naito E, Kobashi H, Yokota I, Miyao M. 1α-Hydroxyvitamin D3 Treatment of Three Patients With 1,25-Dihydroxyvitamin D-Receptor-Defect Rickets and Alopecia. Pediatrics.1987;80(1):97-101.
  87. Takeda E, Yokota I, Kawakami I, Hashimoto T, Kuroda Y, Arase S. Two siblings with vitamin-D-dependent rickets type II: No recurrence of rickets for 14 years after cessation of therapy. European Journal of Pediatrics.1989;149(1):54-57.
  88. Saijo T, Ito M, Takeda E, Huq AH, Naito E, Yokota I, Sone T, Pike JW, Kuroda Y. A unique mutation in the vitamin D receptor gene in three Japanese patients with vitamin D-dependent rickets type II: utility of single-strand conformation polymorphism analysis for heterozygous carrier detection. Am J Hum Genet.1991;49(3):668-673.
  89. Yagi H, Ozono K, Miyake H, Nagashima K, Kuroume T, Pike JW. A new point mutation in the deoxyribonucleic acid-binding domain of the vitamin D receptor in a kindred with hereditary 1,25-dihydroxyvitamin D-resistant rickets. The Journal of Clinical Endocrinology &amp; Metabolism. 1993;76(2):509-512.
  90. Eil C, Liberman UA, Rosen JF, Marx SJ. A Cellular Defect in Hereditary Vitamin-D-Dependent Rickets Type II: Defective Nuclear Uptake of 1,25-Dihydroxyvitamin D in Cultured Skin Fibroblasts. New England Journal of Medicine. 1981;304(26):1588-1591.
  91. Feldman D, Chen T, Cone C, Hirst M, Shani S, Benderli A, Hochberg, Zeev. VITAMIN D RESISTANT RICKETS WITH ALOPECIA: CULTURED SKIN FIBROBLASTS EXHIBIT DEFECTIVE CYTOPLASMIC RECEPTORS AND UNRESPONSIVENESS TO 1,25(OH)2D3. The Journal of Clinical Endocrinology &amp; Metabolism. 1982;55(5):1020-1022.
  92. Clemens TL, Adams JS, Horiuchi N, Gilchrest BA, Cho H, Tsuchiya Y, Matsuo N, Suda T, Holick MF. Interaction of 1,25-Dihydroxyvitamin-D<sub>3</sub>with Keratinocytes and Fibroblasts from Skin of Normal Subjects and a Subject with Vitamin-D-Dependent Rickets, Type II: A Model for Study of the Mode of Action of 1,25-Dihydroxyvitamin D<sub>3</sub>*. The Journal of Clinical Endocrinology &amp; Metabolism.1983;56(4):824-830.
  93. Griffin JE, Zerwekh JE. Impaired stimulation of 25-hydroxyvitamin D-24-hydroxylase in fibroblasts from a patient with vitamin D-dependent rickets, type II. A form of receptor-positive resistance to 1,25-dihydroxyvitamin D3. The Journal of clinical investigation. 1983;72(4):1190-1199.
  94. Liberman UA, Eil C, Holst P, Rosen JF, Marx SJ. Hereditary Resistance to 1,25-Dihydroxyvitamin D: Defective Function of Receptors for 1,25-Dihydroxy vitamin D in Cells Cultured from Bone*. The Journal of Clinical Endocrinology &amp; Metabolism. 1983;57(5):958-962.
  95. Malloy PJ, Weisman Y, Feldman D. Hereditary 1 alpha,25-dihydroxyvitamin D-resistant rickets resulting from a mutation in the vitamin D receptor deoxyribonucleic acid-binding domain. The Journal of Clinical Endocrinology &amp; Metabolism. 1994;78(2):313-316.
  96. Chen TL, Hirst MA, Cone CM, Hochberg Z, Tietze H-U, Feldman D. 1,25-Dihydroxyvitamin D Resistance, Rickets, and Alopecia: Analysis of Receptors and Bioresponse in Cultured Fibroblasts from Patients and Parents*. The Journal of Clinical Endocrinology &amp; Metabolism. 1984;59(3):383-388.
  97. Hirst MA, Hochman H, Feldman D. Vitamin D Resistance and Alopecia: A Kindred with Normal 1,25-Dihydroxyvitamin D Binding, but Decreased Receptor Affinity for Deoxyribonucleic Acid*. The Journal of Clinical Endocrinology &amp; Metabolism. 1985;60(3):490-495.
  98. Castells S, Greig F, Fusi MA, Finberg L, Yasumura S, Liberman UA, Eil C, Marx SJ. Severely Deficient Binding of 1,25-Dihydroxyvitamin D to Its Receptors in a Patient Responsive to High Doses of This Hormone*. The Journal of Clinical Endocrinology &amp; Metabolism. 1986;63(1):252-256.
  99. Fraher LJ, Karmali R, Hinde FRJ, Hendy GN, Jani H, Nicholson L, Grant D, O'Riordan JLH. Vitamin D-dependent rickets type II: extreme end organ resistance to 1,25-dihydroxy vitamin D3 in a patient without alopecia. European Journal of Pediatrics. 1986;145(5):389-395.
  100. Zhu W, Malloy PJ, Delvin E, Chabot G, Feldman D. Hereditary 1,25-Dihydroxyvitamin D-Resistant Rickets Due to an Opal Mutation Causing Premature Termination of the Vitamin D Receptor. Journal of Bone and Mineral Research. 1998;13(2):259-264.
  101. Malloy PJ, Zhu W, Zhao X-Y, Pehling GB, Feldman D. A Novel Inborn Error in the Ligand-Binding Domain of the Vitamin D Receptor Causes Hereditary Vitamin D-Resistant Rickets. Molecular Genetics and Metabolism.2001;73(2):138-148.
  102. Nguyen TM, Adiceam P, Kottler ML, Guillozo H, Rizk-Rabin M, Brouillard F, Lagier P, Palix C, Garnier JM, Garabedian M. Tryptophan Missense Mutation in the Ligand-Binding Domain of the Vitamin D Receptor Causes Severe Resistance to 1,25-Dihydroxyvitamin D. Journal of Bone and Mineral Research.2002;17(9):1728-1737.
  103. Malloy PJ, Pike JW, Feldman D. The Vitamin D Receptor and the Syndrome of Hereditary 1,25-Dihydroxyvitamin D-Resistant Rickets*. Endocrine Reviews. 1999;20(2):156-188.
  104. Malloy PJ, Xu R, Peng L, Clark PA, Feldman D. A Novel Mutation in Helix 12 of the Vitamin D Receptor Impairs Coactivator Interaction and Causes Hereditary 1,25-Dihydroxyvitamin D-Resistant Rickets without Alopecia. Molecular Endocrinology. 2002;16(11):2538-2546.
  105. Liberman UA, Eil C, Marx SJ. Resistance to 1,25-dihydroxyvitamin D. Association with heterogeneous defects in cultured skin fibroblasts. The Journal of clinical investigation. 1983;71(2):192-200.
  106. Hochberg Z, Benderli A, Levy J, Vardi P, Weisman Y, Chen T, Feldman D. 1,25-dihydroxyvitamin D resistance, rickets, and alopecia. The American Journal of Medicine. 1984;77(5):805-811.
  107. Koren R, Ravid A, Liberman UA, Hochberg Z, Weisman Y, Novogrodsky A. Defective binding and function of 1,25-dihydroxyvitamin D3 receptors in peripheral mononuclear cells of patients with end-organ resistance to 1,25-dihydroxyvitamin D. The Journal of clinical investigation. 1985;76(5):2012-2015.
  108. Liberman UA, Eil C, Marx SJ. Receptor-Positive Hereditary Resistance to 1,25-Dihydroxyvitamin D: Chromatography of Hormone-Receptor Complexes on Deoxyribonucleic Acid-Cellulose Shows Two Classes of Mutation*. The Journal of Clinical Endocrinology &amp; Metabolism. 1986;62(1):122-126.
  109. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA, Pike JW, Feldman D, O'Malley BW. Point mutations in the Human Vitamin D Receptor Gene Associated with Hypocalcemic Rickets. Science. 1988;242(4886):1702-1705.
  110. Sone T, Marx SJ, Liberman UA, Pike JW. A Unique Point Mutation in the Human Vitamin D Receptor Chromosomal Gene Confers Hereditary Resistance to 1,25-Dihydroxyvitamin D<sub>3</sub>. Molecular Endocrinology. 1990;4(4):623-631.
  111. Barsony J, Pike JW, DeLuca HF, Marx SJ. Immunocytology with microwave-fixed fibroblasts shows 1 alpha,25-dihydroxyvitamin D3-dependent rapid and estrogen-dependent slow reorganization of vitamin D receptors. J Cell Biol. 1990;111(6 Pt 1):2385-2395.
  112. Sone T, Scott RA, Hughes MR, Malloy PJ, Feldman D, O'Malley BW, Pike JW. Mutant vitamin D receptors which confer hereditary resistance to 1,25-dihydroxyvitamin D3 in humans are transcriptionally inactive in vitro. Journal of Biological Chemistry. 1989;264(34):20230-20234.
  113. Hawa NS, Cockerill FJ, Vadher S, Hewison M, Rut AR, Pike JW, O'Riordan JLH, Farrow SM. Identification of a novel mutation in hereditary vitamin D resistant rickets causing exon skipping. Clinical Endocrinology.1996;45(1):85-92.
  114. Mechica JB, Leite MOR, Mendonca BB, Frazzatto EST, Borelli A, Latronico AC. A Novel Nonsense Mutation in the First Zinc Finger of the Vitamin D Receptor Causing Hereditary 1,25-Dihydroxyvitamin D<sub>3</sub>-Resistant Rickets. The Journal of Clinical Endocrinology &amp; Metabolism. 1997;82(11):3892-3894.
  115. Cockerill FJ, Hawa NS, Yousaf N, Hewison M, O’Riordan JLH, Farrow SM. Mutations in the Vitamin D Receptor Gene in Three Kindreds Associated with Hereditary Vitamin D Resistant Rickets. The Journal of Clinical Endocrinology &amp; Metabolism. 1997;82(9):3156-3160.
  116. Kristjansson K, Rut AR, Hewison M, O'Riordan JL, Hughes MR. Two mutations in the hormone binding domain of the vitamin D receptor cause tissue resistance to 1,25 dihydroxyvitamin D3. The Journal of clinical investigation. 1993;92(1):12-16.
  117. Wiese RJ, Goto H, Prahl JM, Marx SJ, Thomas M, Al-Aqeel A, DeLuca HF. Vitamin D-dependency rickets type II: truncated vitamin D receptor in three kindreds. Molecular and Cellular Endocrinology. 1993;90(2):197-201.
  118. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D. The Crystal Structure of the Nuclear Receptor for Vitamin D Bound to Its Natural Ligand. Molecular Cell. 2000;5(1):173-179.
  119. Zhou Y, Wang J, Malloy PJ, Dolezel Z, Feldman D. Compound heterozygous mutations in the vitamin D receptor in a patient with hereditary 1,25-dihydroxyvitamin D-resistant rickets with alopecia. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.2009;24(4):643-651.
  120. Malloy PJ, Eccleshall TR, Gross C, Van Maldergem L, Bouillon R, Feldman D. Hereditary vitamin D resistant rickets caused by a novel mutation in the vitamin D receptor that results in decreased affinity for hormone and cellular hyporesponsiveness. The Journal of clinical investigation. 1997;99(2):297-304.
  121. Hewison M, Rut AR, Kristjansson K, Walker RE, Dillon MJ, Hughes MR, O'Riordan JLH. Tissue resistance to 1,25-dihydroxyvitamin D without a mutation of the vitamin D receptor gene. Clinical Endocrinology.1993;39(6):663-670.
  122. Whitfield GK, Selznick SH, Haussler CA, Hsieh JC, Galligan MA, Jurutka PW, Thompson PD, Lee SM, Zerwekh JE, Haussler MR. Vitamin D receptors from patients with resistance to 1,25-dihydroxyvitamin D3: point mutations confer reduced transactivation in response to ligand and impaired interaction with the retinoid X receptor heterodimeric partner. Molecular Endocrinology. 1996;10(12):1617-1631.
  123. Barsony J, McKoy W, DeGrange DA, Liberman UA, Marx SJ. Selective expression of a normal action of the 1,25-dihydroxyvitamin D3 receptor in human skin fibroblasts with hereditary severe defects in multiple actions of that receptor. The Journal of clinical investigation. 1989;83(6):2093-2101.
  124. Freedman LP. Increasing the Complexity of Coactivation in Nuclear Receptor Signaling. Cell. 1999;97(1):5-8.
  125. Lin NU, Malloy PJ, Sakati N, al-Ashwal A, Feldman D. A novel mutation in the deoxyribonucleic acid-binding domain of the vitamin D receptor causes hereditary 1,25-dihydroxyvitamin D-resistant rickets. The Journal of Clinical Endocrinology &amp; Metabolism. 1996;81(7):2564-2569.
  126. Rut AR, Hewlson Y, Kristjansson K, Luisl B, Hughes MR, O'Riordan JLH. Two mutations causing vitamin D resistant rickets: modelling on the basis of steroid hormone receptor DNA-binding domain crystal structures. Clinical Endocrinology. 1994;41(5):581-590.
  127. Gamblin GT, Liberman UA, Eil C, Downs RW, Jr., DeGrange DA, Marx SJ. Vitamin D-dependent rickets type II. Defective induction of 25-hydroxyvitamin D3-24-hydroxylase by 1,25-dihydroxyvitamin D3 in cultured skin fibroblasts. The Journal of clinical investigation. 1985;75(3):954-960.
  128. Balsan S, Garabédian M, Larchet M, Gorski AM, Cournot G, Tau C, Bourdeau A, Silve C, Ricour C. Long-term nocturnal calcium infusions can cure rickets and promote normal mineralization in hereditary resistance to 1,25-dihydroxyvitamin D. The Journal of clinical investigation. 1986;77(5):1661-1667.
  129. Barsony J, Marx SJ. Receptor-mediated rapid action of 1 alpha,25-dihydroxycholecalciferol: increase of intracellular cGMP in human skin fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 1988;85(4):1223-1226.
  130. Ritchie HH, Hughes MR, Thompson ET, Malloy PJ, Hochberg Z, Feldman D, Pike JW, O'Malley BW. An ochre mutation in the vitamin D receptor gene causes hereditary 1,25-dihydroxyvitamin D3-resistant rickets in three families. Proc Natl Acad Sci U S A. 1989;86(24):9783-9787.
  131. Malloy PJ, Hochberg Z, Tiosano D, Pike JW, Hughes MR, Feldman D. The molecular basis of hereditary 1,25-dihydroxyvitamin D3 resistant rickets in seven related families. J Clin Invest. 1990;86(6):2071-2079.
  132. Lin JP, Uttley WS. Intra-atrial calcium infusions, growth, and development in end organ resistance to vitamin D. Arch Dis Child. 1993;69(6):689-692.
  133. Bliziotes M, Yergey AL, Nanes MS, Muenzer J, Begley MG, Vieira NE, Kher KK, Brandi ML, Marx SJ. Absent Intestinal Response to Calciferols in Hereditary Resistance to 1,25-Dihydroxyvitamin D: Documentation and Effective Therapy With High Dose Intravenous Calcium Infusions*. The Journal of Clinical Endocrinology &amp; Metabolism. 1988;66(2):294-300.
  134. Weisman Y, Bab I, Gazit D, Spirer Z, Jaffe M, Hochberg Z. Long-term intracaval calcium infusion therapy in end-organ resistance to 1,25-dihydroxyvitamin D. The American Journal of Medicine. 1987;83(5):984-990.
  135. Sakati N, Woodhouse NJY, Niles N, Harfi H, de Grange DA, Marx S. Hereditary Resistance to 1,25-Dihydroxyvitamin D: Clinical and Radiological Improvement during High-Dose Oral Calcium Therapy. Hormone Research. 1986;24(4):280-287.
  136. Adams JS, Gacad MA. Phenotypic Diversity of the Cellular 1,25-Dihydroxyvitamin D<sub>3</sub>-Receptor Interaction Among Different Genera of New World Primates. The Journal of Clinical Endocrinology &amp; Metabolism. 1988;66(1):224-229.
  137. Liberman UA, de Grange D, Marx SJ. Low affinity of the receptor for 1α, 25-dihydroxyvitamin D<sub>3</sub> in the marmoset, a new world monkey. FEBS Letters. 1985;182(2):385-388.
  138. Shinki T, Shiina Y, Takahashi N, Tanioka Y, Koizumi H, Suda T. Extremely high circulating levels of 1α,25-dihydroxyvitamin D3 in the marmoset, a new world monkey. Biochemical and Biophysical Research Communications. 1983;114(2):452-457.
  139. Takahashi N, Suda S, Shinki T, Horiuchi N, Shiina Y, Tanioka Y, Koizumi H, Suda T. The mechanism of end-organ resistance to 1 alpha,25-dihydroxycholecalciferol in the common marmoset. Biochem J.1985;227(2):555-563.
  140. Gacad MA, Adams JS. Identification of a competitive binding component in vitamin D-resistant new world primate cells with a low affinity but high capacity for 1,25-dihydroxyvitamin D3. Journal of Bone and Mineral Research. 2009;8(1):27-35.
  141. Arbelle JE, Chen H, Gaead MA, Allegretto EA, Pike JW, Adams JS. Inhibition of vitamin D receptor-retinoid X receptor-vitamin D response element complex formation by nuclear extracts of vitamin D-resistant New World primate cells. Endocrinology. 1996;137(2):786-789.
  142. Lipsett MB, Chrousos GP, Tomita M, Brandon DD, Loriaux DL. The Defective Glucocorticoid Receptor in Man and Nonhuman Primates. Proceedings of the 1984 Laurentian Hormone Conference: Elsevier; 1985:199-247.
  143. Gacad MA, Chen H, Arbelle JE, LeBon T, Adams JS. Functional characterization and purification of an intracellular vitamin D-binding protein in vitamin D-resistant new world primate cells. Amino acid sequence homology with proteins in the hsp-70 family. J Biol Chem. 1997;272(13):8433-8440.
  144. Chen H, Hu B, Gacad MA, Adams JS. Cloning and expression of a novel dominant-negative-acting estrogen response element-binding protein in the heterogeneous nuclear ribonucleoprotein family. J Biol Chem.1998;273(47):31352-31357.
  145. Chen H, Hewison M, Hu B, Adams JS. Heterogeneous nuclear ribonucleoprotein (hnRNP) binding to hormone response elements: a cause of vitamin D resistance. Proc Natl Acad Sci U S A. 2003;100(10):6109-6114.
  146. Chen CH, Sakai Y, Demay MB. Targeting Expression of the Human Vitamin D Receptor to the Keratinocytes of Vitamin D Receptor Null Mice Prevents Alopecia. Endocrinology. 2001;142(12):5386-5389.
  147. Bikle DD, Chang S, Crumrine D, Elalieh H, Man MQ, Choi EH, Dardenne O, Xie Z, Arnaud RS, Feingold K, Elias PM. 25 Hydroxyvitamin D 1 alpha-hydroxylase is required for optimal epidermal differentiation and permeability barrier homeostasis. J Invest Dermatol. 2004;122(4):984-992.

Pathogenesis of Type 1 Diabetes

Dana VanBuecken and Sandra Lord contributed equally to this submission

ABSTRACT

 

Type 1A diabetes (T1D) represents an autoimmune disorder that can affect individuals from within a year of birth until age 60. A number of genes strongly influence the development of disease, including genes found within the human lymphocyte antigen (HLA) complex. The role of non-HLA genes is being defined in recent studies, and we are beginning to identify pathways that lead to autoimmunity and eventually pancreatic islet cell destruction. Although genes can predispose one to type 1A diabetes, environmental factors may also play a significant role in the pathogenesis. These as-yet-undefined factors appear to have accelerated the onset and markedly increased the frequency of disease in many populations around the world over the last 30 years. The development of ever more sophisticated immunoassays to detect antibodies directed against pancreatic antigens have helped define the autoimmune nature of the disorder, but as importantly have also provided an opportunity to identify those individuals with prediabetes and to stratify their risk of developing overt hyperglycemia. Immunologic assays as well as intervention trials are allowing us to learn more about the immune pathways that are disordered and offer hope for future therapeutic approaches to prevent and reverse type 1A diabetes.

 

INTRODUCTION

 

In the U.S. alone, more than one million people are living with type 1 diabetes (TID) and approximately 80 people per day, or 30,000 individuals per year, are newly diagnosed (1, 2). Recent epidemiological studies demonstrate that the global T1D incidence is increasing at a rate of approximately 3-4% per year, notably among younger children (3, 4). Despite improvements in insulins, insulin delivery methods, and home glucose monitoring, the vast majority of those with T1D do not achieve recommended levels of glycemic control.  This is particularly true in childhood and adolescence, where a recent U.S. study reported mean HbA1c values exceeding 9.5%, and a high frequency of both DKA and severe hypoglycemia (5). In addition to the increased risk of morbidity and mortality, TID places significant emotional and financial burdens on individuals, families, and society. These realities highlight the need for both better TID therapies and the continued push towards the prevention of TID. In recent decades, research efforts have described the natural history of type 1 diabetes and expanded the ability to identify individuals at risk for the disease even before clinical onset, via the recognition of genetic markers or TID-specific autoantibodies. The increasing ability to identify the at-risk population affords researchers the opportunity to intervene at progressively earlier stages in the disease.  With the understanding that established islet autoimmunity, confirmed by the presence of multiple T1D autoantibodies, inevitably leads to clinical TID, investigative efforts are shifting towards the prevention or modification of autoimmunity.  Furthermore, with the mounting evidence that any amount of residual C-peptide improves long term clinical outcomes in TID, some therapies aim to preserve remaining beta cell function in those with clinical disease. In this chapter, we review the epidemiology of TID and the genetic and environmental risk factors for T1D.

 

 

EPIDEMIOLOGY OF DIABETES

 

T1D, or autoimmune diabetes, represents 5-10% of diabetes, and like autoimmunity in general, TID is increasing worldwide. The increase likely is attributable to environmental factors or epigenetic changes, as genetic changes don’t occur rapidly enough to explain such a dramatic increase. The SEARCH for Diabetes in Youth Study is a multicenter observational study investigating trends in incidence and prevalence of diabetes in American youth < age 20.  SEARCH data suggests that the prevalence of TID among non-Hispanic white youth is ~1/300 in the US by age 20 years (6). Between 2002 and 2009, the incidence of TID among non-Hispanic white youth < age 20 years increased by an average of 2.7% per year (7). Similarly, the EURODIAB study evaluated TID incidence trends in 17 European countries from 1989-2003 in youth < age 15 years, and found an average annual incidence increase of 3.9%. This trend predicts a 70% increase in TID prevalence between 2005-2020 among European youth < 15 years old (8) with the peak of diagnosis between ages 10-14 (9). While incidence and prevalence are well documented in children, TID occurs in adults as well, at a frequency that is less certain; estimates are that 25-50% of all TID cases are diagnosed in adulthood. The uncertainty likely is due to a less dramatic clinical presentation than is typically seen in children who present with TID. The incidence of TID varies tremendously by geographic location, with higher rates generally seen in countries located farther from the equator. Worldwide incidence data was reported in 2000 by the DIAMOND project (10), a WHO-sponsored effort to address the public health implications of TID. The incidence of TID between 1990 and 1994 in 50 countries is shown in Figure 1. Between 1990 and 1994, the incidence of TID in individuals aged 0-14 years in both Finland and Sardinia was 37/100,000 individuals, whereas the incidence in both China and Venezuela was 0.1/100,000 individuals, a 350-fold difference. The increased incidence coupled with reduced early mortality has contributed to the increasing prevalence of disease. 

 

Figure 1. Worldwide incidence of TID 1990-1994, used with permission from International Diabetes Federation.

 

 

 

WHAT IS THE RISK OF TYPE 1 DIABETES?

 

As is true for Cindy, 85% of individuals who develop TID have no family history of TID; nonetheless, a family history of the disease does increase an individual’s relative risk.  The prevalence of TID in the US non-Hispanic white population by age 20 is ~0.3%, as compared with ~5% of those with a relative with TID, a 15-fold increase in relative risk.   This relative risk is depicted in Figure 2.

 

Figure 2. Among 300 people without a family member with diabetes, 1 will have TID. Among 300 people with a family member with diabetes, 15 will have TID.

 

The risk of TID among family members varies depending on who the affected family member is, as shown in Table 1.  

 

Table 1. Prevalence of TID in Individuals with a Family History of TID

Relative with TID

Prevalence at age 20

Reference

Mother

2%

(11, 12)

Father

6%

(11, 12)

Non-twin sibling

6%

(13)

Dizygotic (fraternal) twin

10%

(13, 14)

Monozygotic (identical) twin

>50%

(15)

 

The heritability pattern suggests that both genes and environment contribute to risk.  Curiously, the risk of TID in offspring is higher if the father has TID (~6%) as compared to if the mother has TID (~2%) (11, 12). Moreover, the risk to a dizygotic twin is slightly higher (~10%) than is the risk to a non-twin sibling with similar HLA risk genes (~6%) (13, 14) suggesting that the intrauterine environment and/or similar early life exposures may be important. Lastly, the risk to a monozygotic twin is upwards of ~50%; surprisingly the second twin’s diagnosis may occur many decades after the index twin, highlighting the complexities of gene and environmental interactions that underlie the disease (15).

 

 

 

THE NATURAL HISTORY TYPE 1 DIABETES

 

It is now understood that TID is an immune-mediated disease that begins in the setting of genetic predisposition and then progresses along a predictable path: early islet autoimmunity (one autoantibody), established islet autoimmunity (two or more autoantibodies), abnormal glucose tolerance, clinical TID with some remaining beta cell function, and finally, little or no remaining beta cell function. This understanding comes from decades of effort by multiple investigators and from participation by thousands of patients with TID and their family members.  George Eisenbarth’s description of TID as a chronic autoimmune disease, manifested by autoimmunity and a gradual linear fall in beta cell function until there is insufficient beta cell mass to suppress symptomatic hyperglycemia, has served for decades as the TID natural history paradigm (16). The “Eisenbarth” model has undergone refinements in recent years; namely, although autoimmunity and beta cell dysfunction do appear prior to diagnosis, these changes are often step-wise and non-linear.  Furthermore, beta cell destruction may not be absolute.  Nonetheless, the paradigm is largely correct and serves as the underlying rationale for TID trials. 

 

The long pre-symptomatic natural history of TID presents an opportunity to intervene earlier than is done currently. Diabetes-specific autoantibodies can appear many years before clinical diagnosis and may reliably be used to predict disease progression. In 2015, JDRF, the Endocrine Society, and the American Diabetes Association proposed a new TID staging system which underscores that TID begins with islet autoimmunity rather than with symptomatic hyperglycemia (17). Stage 1 TID is defined as the presence of 2 or more autoantibodies with normoglycemia; stage 2 TID is 2 or more autoantibodies, impaired glucose tolerance, and no symptoms; stage 3 TID is clinical disease. The staging system is depicted in figure 3.  

 

HOW TO DETERMINE RISK OF TID

 

Risk of TID may be determined by the identification of autoantibodies, usually in those identified as having genetic risk through HLA testing or by family history. Autoantibodies are detectable years before the onset of clinical TID. 

 

Determining Risk: Genes

 

With the knowledge that TID runs in families and with advances in technology, investigators have described the genetic risk of TID.  TID risk is strongly linked to HLA class II DR3 and DR4 haplotypes, with the highest risk in those with the DR3/DR4 genotype.  The importance of HLA genes to TID risk highlights the role of the adaptive immune system in the development of autoimmunity.  Newer studies have discovered multiple other genes that also contribute to TID risk (18). They are largely genes known also to impact immune function; however, their contribution is dwarfed by the impact of HLA genes. Interestingly, recent work suggests that HLA genes primarily contribute to development of autoantibodies, while non-HLA genes and environmental factors may be more important in the progression from autoantibodies to clinically overt disease (19, 20). The description of non-HLA risk genes (such as the genes for insulin, a major TID autoantigen) highlights other potential pathways to disease and potential therapies. 

Although the contribution of HLA class II risk genes overwhelms the contribution of non-HLA risk genes, the HLA contribution may be decreasing as the overall incidence of TID increases.  This suggests that in a population with non-HLA genetic susceptibility, the environment may have become more conducive to the development of TID. This was reported in a 2004 Lancet article by Gillespie, et al., in which the investigators compared the frequency of HLA class II haplotypes in a UK cohort of 194 individuals diagnosed with TID between 1922-1946 (the Golden Years cohort) to a cohort of 582 individuals diagnosed between 1985-2002 (the BOX cohort) (21). In this comparison, shown in Figure 4, 47% of individuals in the Golden Years cohort were positive for the highest risk genotype DR3-DQ2/DR4-DQ8, compared to 35% of individuals in the BOX cohort.

 

Figure 4. Decreased contribution of high-risk HLA haplotypes over time. HLA class II haplotypes in Golden Years and BOX cohorts, adapted from Gillespie et.al Lancet 2004 (21).

 

Determining Risk: Family History And Islet Cell Autoantibodies

 

Natural history studies of relatives such as Diabetes Prevention Trial (DPT-1) and Diabetes TrialNet Pathway to Prevention have helped define the risk of TID in those with a family history of TID.  Since 2000, Diabetes TrialNet has screened over 200,000 relatives of people with TID, aiming to enroll at-risk individuals in prevention trials.  Among relatives of people with TID, ~5% will have at least one of five islet autoantibodies (22). TrialNet screens for islet cell antibodies (ICA), autoantibodies to insulin (IAA or mIAA), antibodies to a tyrosine phosphatase (IA-2; previously ICA512), antibodies to glutamic acid decarboxylase (GAD), and antibodies to a zinc transporter (ZnT8).  With each additional autoantibody, the risk of TID increases predictably. Unsurprisingly, those with islet autoimmunity and abnormal glucose tolerance are at an even further increased risk of symptomatic T1D. The TrialNet strategy to identify islet autoimmunity among relatives of individuals with TID is shown in Figure 5. There are many other screening efforts ongoing outside of TrialNet. (23-25)

 

Figure 5. Diabetes TrialNet process for identifying relatives with islet autoimmunity.

 

Natural history studies have shown not only that islet autoimmunity predicts TID risk, but also that islet autoantibodies usually appear early in life; 64% of babies destined to develop T1D before puberty will have antibodies by age 2 and 95% by age 5 (26). Furthermore, the data from both prospective birth cohort studies (27) and cross-sectional studies (28-31) is remarkably consistent and suggests that the risk of progression from established autoimmunity to clinical TID is in the range of 40% after 5 years, 70% after 10 years, and 85% after 15 years. This risk over time is depicted in Figure 6. The key understanding from natural history studies is that essentially all individuals with confirmed islet autoimmunity will eventually develop clinical T1D at a rate of 11% per year.

 

Figure 6. Established islet autoimmunity inevitably progresses to clinical T1D. Extrapolated data from multiple studies in genetically at-risk individuals; Ziegler et al. JAMA 2013; DPT-1 Study Group Diabetes 1997; Sosenko et al. Diabetes Care 2014; Mahon et al. Pediatric Diabetes 2009.

 

Identifying individuals with islet autoimmunity has two potential benefits; namely, the opportunity to monitor closely for disease progression, conferring a reduced risk of morbidity and mortality at the time of TID diagnosis, and the identification of individuals who are eligible for prevention trials.  It is perhaps underappreciated that there is potentially a direct clinical benefit to identifying those with islet autoimmunity.  Individuals with islet autoimmunity followed regularly until clinical diagnosis present with lower HbA1c and experience less DKA than those diagnosed in the community (Table 2) (32-36). For this reason, since 2009, the ADA has recommended that all individuals with a relative with T1D be counseled about the opportunity to be screened for diabetes autoantibodies in the context of a clinical research trial (37).

 

Table 2.  Individuals Diagnosed with T1D While Enrolled in a Clinical Trial have Less Morbidity at the Time of Diagnosis. (32-36)

 

STUDY

HbA1c at time of TID diagnosis

% with DKA at time of TID diagnosis

 

Enrolled in study

Usual care

Enrolled in study

Usual care

SEARCH

 

 

 

25.5%

BABYDIAB

8.6%

11.0%

3.3%

29.1%

DPT-1

6.4%

 

3.7%

 

DAISY

7.2%

10.9%

< 4%

 

TEDDY < age 5

 

 

13.1%

 

SEARCH < age 5

 

 

 

36.4%

BABYDIAB < age 5

 

 

 

32.3%

 

STRATEGIES TO BRING SCREENING FOR RISK TO CLINICAL PRACTICE

 

Screening relatives does identify a population of those at risk for clinical T1D; however, at least 85% who get T1D have no relatives with disease.  Thus, to truly prevent all T1D, testing of the general population would have to occur. This could be done with current technology by testing all babies for genetic (HLA) risk at birth and then following with antibody testing. The Population Level Estimate of type 1 Diabetes risk Genes in children (PLEDGE) study enrolls newborns from the general population and offers one-time genetic testing and follow-up autoantibody testing at 2 and 4 years of age (38). The study aims to demonstrate feasibility and to develop evidence to support eventual inclusion of a T1D screening program in standard primary care.

 

Other studies, such as The Environmental Determinants of Diabetes in the Young (TEDDY) study, the Diabetes Autoimmunity Study in the Young (DAISY), and the Global Platform for the Prevention of Autoimmune Diabetes (GPPAD) are exploring similar methodologies to screen and monitor for risk (24, 39, 40).  However, with an increasing number of individuals developing T1D even without the high-risk HLA types, such approaches may still miss some destined to develop disease. 

 

An alternative risk detection strategy for those without a family history may be to perform point-of-care antibody testing in a routine pediatric visit.  Since almost all who will develop diabetes before puberty will have antibodies by age 5; such testing could be done at age 4-5 and perhaps once again in the teenage years.  This method will still miss those who develop T1D before this age, but would likely be a cost-effective approach to finding those at risk.  If these at-risk subjects are monitored regularly until development of clinical disease they would benefit from reduced morbidity at time of diagnosis even if a prevention therapy were not yet available.

 

There are many ongoing projects aimed at screening members of the general population for diabetes autoantibodies even without prior HLA testing (23, 25, 41, 42).

 

As risk-screening programs employ varying assays and recruit from different populations, interpretation and translation of results is unclear. It is not yet known whether those found to be autoantibody positive through one program will experience the same rates of T1D progression and/or benefit from the same therapies as individuals who have participated in other screening and intervention efforts.

 

Source: (37).

 

 

PRENATAL INFLUENCES  

 

The prenatal environment can have profound effects on the developing fetus. With the recognition that antibodies often develop early in life and that essentially all those with established islet autoimmunity (two or more autoantibodies) will eventually develop TID, investigators have looked to the prenatal period to search for factors that could contribute to disease development in utero.  As shown in Table 3, decades of observational studies have yielded inconsistent results.  Yet this remains an important area of investigation and one that may lead to primary prevention strategies for T1D. The Environmental Determinants of Islet Autoimmunity (ENDIA) study is an ongoing prospective birth cohort study in Australia that enrolled infants and unborn infants of first degree relatives with T1D. Biologic samples including blood, stool, and saliva will be collected longitudinally for investigation of factors including viral exposures during pregnancy and early childhood, maternal and fetal microbiome, delivery method, maternal and early infant nutrition, pregnancy and early childhood body weight, and both innate and adaptive immune function. In 2018, the ENDIA study completed target enrollment of ~1500 subjects, who will be followed regularly until the development of islet autoimmunity (43).

 

Table 3.  Potential Prenatal Influences on TID Risk

Pre-natal or intrauterine exposure

Relative risk to offspring

Reference

Maternal age

Inconsistent data

(44-46)

Birth weight > 2 SD above norm (~4000g)

Inconsistent data

(47-51)

Birth weight < 2 SD below norm (~2500g)

Inconsistent data

(49-51)

Birth order: second and later born

Inconsistent data

(46, 52, 53)

Birth interval < 3 years

Inconsistent data

(46, 54)

Caesarean delivery

Inconsistent data

(51, 55, 56)

Pre-eclampsia

Inconsistent data

(51, 57)

Pre-term delivery (<37 weeks gestation)

Inconsistent data

(51, 58)

Maternal vitamin D supplementation

Inconsistent data

(59-62)

Maternal antibiotic use

No association

(53, 63)

maternal BMI/pregnancy weight gain

No association

(51, 64)

Maternal omega 3 fatty acid supplementation

No association

(60, 65, 66)

 

Source: (67).

 

 

Investigators also have studied the early childhood period for clues to the causes of islet autoimmunity and TID; these have included both observational studies and randomized clinical trials. Such influences might be divided into early nutritional exposures and early microbial/infectious exposures, both of which can affect development of the normal immune system.

 

The inconsistent findings relating to environmental factors reported from observational studies and clinical trials led to the design and implementation of a large international comprehensive evaluation of genetically at-risk babies using cutting edge technologies to study genetics, genomics (gene function), metabolomics, and the microbiome. The Environmental Determinants of Diabetes in the Young (TEDDY) is an international prospective birth cohort study that recruited almost 8,000 babies at increased risk for TID (based on HLA and family history) from Finland, Germany, Sweden, and the US from 2004-2010.  Information on environmental exposures such as diet (including breastfeeding history), infections, vaccinations, and psychosocial stressors will be collected. Participants will be followed until the age of 15 for the development of islet autoimmunity or TID. The wealth of data from this study will provide a foundation for future randomized clinical trials (24). One interesting finding reported in December 2019 is that there are subtle differences in the gut microbiome—such as, persistent stool enterovirus B species--in children who develop islet autoimmunity compared to children who do not develop autoimmunity (68).

 

EARLY NUTRITIONAL EXPOSURES

 

Breastfeeding

 

The hypothesis that human breastmilk may protect against future TID development was presented as early as 1984 (69). Since then, there have been several prospective cohort studies to suggest that breastmilk lowers the risk of islet autoimmunity and TID, including the German BABYDIAB/BABYDIET study (70), the Colorado-based DAISY study (71), and the Norwegian MIDIA study (72), but others show no effect (73).  Although the data on whether breastmilk is protective against TID isn’t clear, it certainly isn’t harmful.  Given the well-established general benefits of breastfeeding, patients may safely be advised to follow the American Academy of Pediatrics’ guidelines related to infant feeding. The mechanism by which breastmilk may lower the risk of TID is uncertain, but one theory suggests that breastmilk has positive effects on the infant microbiome. The microbiome is discussed in greater detail below.  

 

Cow’s Milk And Bovine Insulin Exposure

 

In contrast to considering breastfeeding as potentially beneficial in protecting against autoimmunity, it was hypothesized that early introduction of cow’s milk or cow protein might accelerate disease.  This concept was tested in the Trial to Reduce IDDM in the Genetically at Risk (TRIGR) which asked whether weaning to hydrolyzed casein (which is free of bovine proteins including insulin) formula (n=1081) instead of regular cow’s milk formula (n=1078) in genetically at-risk infants could prevent or delay TID.  Though the TRIGR pilot study was suggestive of benefit, no benefit was seen in the fully powered study (74) (75). Similarly, The Finnish Dietary Intervention Trial for the Prevention of Type 1 Diabetes of (FINDIA) suggested that weaning to hydrolyzed cow’s milk formula was not effective in reducing the appearance of autoantibodies, though they did report that a patented cow’s milk formula specifically removing bovine insulin appeared to be beneficial in this pilot study (76).  While additional studies may be informative, current data does not support that weaning to hydrolyzed cow’s milk formula is protective against islet autoimmunity. 

 

Gluten Exposure

 

Both BABYDIAB (77) and DAISY (78) were observational studies that suggested an association between introduction of gluten and islet autoimmunity.  However, these studies had different results as to the timing of gluten introduction. Similarly, no effect was found in the BABYDIET study; a randomized controlled trial that asked whether delayed introduction of gluten to 6 vs 12 months would affect the risk of diabetes autoimmunity (79, 80).

 

Vitamin D And/Or Omega 3 Fatty Acids

 

Vitamin D is an important component of a normal immune response; moreover, the higher incidence of TID in northern climates suggests that vitamin D deficiency could contribute to autoimmunity and TID.  However, data from observational studies is mixed on whether vitamin D and/or omega 3 supplementation is beneficial or not (60, 81-86). A pilot randomized trial of omega 3 supplementation to pregnant mothers and infants failed to demonstrate a profound immunologic effect of treatment (87). With routine vitamin D supplementation recommended for infants (88), it is unlikely that a fully powered randomized trial would be feasible to assess the impact on autoimmunity. 

 

MICROBIAL EXPOSURES

 

The Hygiene Hypothesis

 

Parallel to the rising incidence of TID and other autoimmune diseases, there has been a worldwide trend towards urbanization, increased standard of living, smaller family sizes, less crowded living conditions, safer water and food supplies, less cohabitation with animals, wide use of antibiotics, childhood vaccination, etc.  While these trends are generally considered improvements in human existence, the so-called “hygiene hypothesis,” proposed by Strachan in 1989 (89) suggests a possible downside; that is, that early microbial exposures might have a protective effect via the early education of the immune system and the development of normal tolerance to self-antigens. Data cited in support of the hygiene hypothesis comes from comparisons between eastern Finland and Russian Karelia (Figure 7) (90-92).

 

Figure 7. Border between Finland and Russian Karelia, with a 6-fold difference in the incidence of TID, from "Karelia today”. The countries share a common border and ancestry and thus have similar geography, climate, vitamin D levels, and prevalence of HLA risk haplotypes. However, Finland has 6-fold higher incidence of TID. This markedly higher rate of TID is accompanied by a much lower rate of infectious disease. In Finland as compared to Karelia 2% vs 24% had hepatitis A; 5% vs 24% had toxoplasma gondii; and 5% vs. 73% for helicobacter pylori. There is an ongoing study aiming to better understand the mechanisms that may underlie these differences.

 

The Microbiome

 

Another possible interface between microbial exposure and human disease is through the microbiome; that is the gut flora established within the first 3 years of life (93).  It has been hypothesized that perturbations in normal early microbiome development might pre-dispose to disease whether through direct modulation of innate immunity or via alteration of intestinal permeability and the downstream effects on adaptive immunity. Interestingly, it appears that the gut microbiome is less diverse and less “protective” in individuals with islet autoimmunity or recent onset TID (94-96).  Whether this difference is cause, effect, or correlation isn’t known. Nonetheless, multiple factors might affect the early intestinal microbiome, some of which also have been shown to correlate with risk of islet autoimmunity and TID.  For example, breastfeeding can alter the intestinal microbiome of the infant by increasing the number and diversity of beneficial microbiota (97, 98). As previously discussed, multiple prospective observational studies suggest that breastfeeding protects against future development of islet autoimmunity and TID, but there’s no evidence to connect this directly to the infant microbiome.

  

Viral Infections

 

A viral etiology for initiation of autoimmunity is an attractive idea; a beta cell trophic virus could contribute to disease by directly killing beta cells, by leading to a chronic infection which triggers an immune response, or by molecular mimicry in which self-antigens are erroneously recognized as viral epitopes targeted for destruction.  Notably, these possible mechanisms would not necessarily point to a particular virus; any virus widespread in a population could theoretically lead to autoimmunity in genetically susceptible individuals if encountered at a vulnerable time in immune system or beta cell development.  With the notable exception of congenital rubella which is associated with type 1 diabetes (99), other data relating viruses to initiation of autoimmunity is less conclusive.  While some studies have reported viral “footprints” in islets from individuals who have died from TID, these have not been consistently confirmed.  Similarly, many studies have focused on enteroviruses, including coxsackie B, due to observations suggesting seasonal variation in antibody development that is reminiscent of the timing of such infections (100) (101), yet this remains controversial.  Aside from a viral role in the initiation of autoimmunity, others have proposed that acute viral infections may impact the transition from islet autoimmunity to clinical TID due to increased insulin demand during infections.  Patients commonly report an acute viral illness preceding the diagnosis of TID, and the clinical onset of TID more commonly presents in the fall and winter months in both the northern and southern hemispheres (102); but this does not imply a causal relationship.

 

Vaccinations

 

In recent decades, an increasing number of parents in Western countries have declined routine childhood vaccination of their children, which has created a situation with significant personal and public health consequences.  Multiple high-quality studies have thoroughly investigated vaccinations and TID, and none have found any association with islet autoimmunity or TID (103-107)

 

Sources: (88, 103-108).

 

FUTURE CONSIDERATIONS

 

Despite advances in glucose monitoring and insulin delivery, the daily psychological and financial burden of disease on individuals, their families, and society together with the persistence of complications and reduced life span demand a paradigm shift.

 

As of 2021, we know much about the natural history of disease. We know that antibodies can develop early in life and that essentially all of those with established islet autoimmunity will develop clinically overt disease. We also know that identifying these individuals is of significant clinical benefit. Those with islet autoimmunity followed carefully until diagnosis have markedly less morbidity at the time of diagnosis and lower HbA1c values. Family members of T1D probands should be made aware of their disease risk and should be offered autoantibody screening and enrollment in monitoring trials. Correspondingly, patients with TID should be informed of the opportunity to have their relatives screened for TID risk in the setting of a clinical research study.

 

While the interaction of humans with their environment must contribute to disease; how this occurs is still being elucidated. It is likely that there are many different paths by which individual gene/environment interactions result in T1D; suggesting that dissecting this heterogeneity will provide better insights and therapies.

 

REFERENCES

 

  1. JDRF Fact Sheet (accessed 2021 Dec 14) Available from: https://www.jdrf.org/t1d-resources/about/facts/.
  2. Centers for Disease Control and Prevention, National Diabetes Statistics Report, 2014. 2014; (accessed 2021 Dec 14)Available from: http://www.cdc.gov/diabetes/pubs/statsreport14/national- diabetes-report-web.pdf.
  3. Patterson, C., et al., 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(8): p. 2142-2147.
  4. Bell, R., et al., Diabetes in non-Hispanic white youth:prevalence, incidence, and clinical characteristics: the SEARCH for diabetes in youth study. Diabetes Care, 2009. 32(Suppl 2): p. S102-S111.
  5. Miller, K.M., et al., Current State of Type 1 Diabetes Treatment in the U.S.: Updated Data From the T1D Exchange Clinic Registry. Diabetes Care, 2015. 38(6): p. 971-978.
  6. Liese, A.D., R.B. D'Agostino, Jr., and R. Hamman, The burden of diabetes mellitus among US youth; prevalence estimates from the SEARCH for diabetes in youth study. Pediatrics, 2006. 118(4): p. 1510-18.
  7. Lawrence, J., et al., Trends in incidence of type 1 diabetes among non-Hispanic white youth in the U.S., 2002-2009 Diabetes, 2014. 63(11): p. 3938-3945.
  8. Patterson, C., et al., Incidence trends for childhood type 1 diabetes in Europe during 1989-2003 and predicted new cases 2005-20: a multicentre prospective registration study. Lancet, 2009. 373(9680): p. 2027-2033.
  9. Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabet. Med, 2006. 23(8): p. 857-866.
  10. Karvonen, M., et al., Incidence of childhood type 1 diabetes worldwide. Diabetes Mondiale (DiaMond) Project Group. Diabetes Care, 2000. 23(10): p. 1516-1526.
  11. Warram, J.H., et al., Differences in risk of insulin- dependent diabetes in offspring of diabetic mothers and diabetic fathers. N Engl J Med, 1984. 311(3): p. 149-52.
  12. Pociot, F., et al., A nationwide population-based study of the familial aggregation of type 1 (insulin-dependent) diabetes mellitus in Denmark. Danish Study Group of Diabetes in Childhood. Diabetologia, 1993. 36(9): p. 870-875.
  13. Nistico, L., et al., Emerging effects of early environmental factors over genetic background for type 1 diabetes susceptibility: evidence from a Nationwide Italian Twin Study. J Clin Endocrinol Metab, 2012. 97(8): p. E1483- E1491.
  14. Kyvik, K., A. Green, and H. Beck-Nielsen, Concordance rates of insulin dependent diabetes mellitus: a population based study of young Danish twins. BMJ, 1995. 311(7010): p. 913-917.
  15. Redondo, M., et al., Concordance for islet autoimmunity among monozygotic twins. N Engl J Med, 2008. 359(26): p. 2849-2850.
  16. Eisenbarth, G.S., Type I diabetes mellitus. A chronic autoimmune disease. N. Engl. J. Med, 1986. 314(21): p. 1360-1368.
  17. Insel, R.A., et al., Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care, 2015. 38(10): p. 1964-74.
  18. Concannon, P., S.S. Rich, and G.T. Nepom, Genetics of type 1A diabetes. N. Engl. J. Med, 2009. 360(16): p. 1646- 1654.
  19. Knip, M., Can we predict type 1 diabetes in the general population? Diabetes Care, 2002. 25(3): p. 623-5.
  20. Steck, A.K., et al., Predictors of Progression From the Appearance of Islet Autoantibodies to Early Childhood Diabetes: The Environmental Determinants of Diabetes in the Young (TEDDY). Diabetes Care, 2015. 38(5): p. 808-13.
  21. Gillespie, K.M., et al., The rising incidence of childhood type 1 diabetes and reduced contribution of high-risk HLA haplotypes. Lancet, 2004. 364(9446): p. 1699-1700.
  22. Mahon, J.L., et al., The TrialNet Natural History Study of the Development of Type 1 Diabetes: objectives, design, and initial results. Pediatr. Diabetes, 2008.
  23. JDRF. T1Detect: Learn why you should be screened. (accessed 2021 Dec 14); Available from: https://www.jdrf.org/t1d-resources/t1detect/.
  24. Hagopian, W.A., et al., TEDDY--The Environmental Determinants of Diabetes in the Young: an observational clinical trial. Ann N Y Acad Sci, 2006. 1079: p. 320-6.
  25. Combined Antibody Screening for Celiac and Diabetes Evaluation (CASCADE). 2020.
  26. Parikka V., N.-S.K., Saarinen M, Simell T, Ilonen J, Hyöty H, Veijola R, Knip M, Simell O., Early seroconversion and rapidly increasing autoantibody concentrations predict prepubertal manifestation of type 1 diabetes in children at genetic risk. Diabetologia, 2012. 55(7): p. 1926-36.
  27. Vehik, K., et al., Methods, quality control and specimen management in an international multicentre investigation of type 1 diabetes: TEDDY. Diabetes Metab Res Rev, 2013. 29(7): p. 557-67.
  28. Ziegler, A.G., et al., Seroconversion to Multiple Islet Autoantibodies and Risk of Progression to Diabetes in Children. Journal of the American Medical Association, 2013. 309(23): p. 2473-9.
  29. DPT-1 Study Group, Demographics of relatives screened and ICA positive in the diabetes prevention trial-type 1 diabetes (DPT-1). Diabetes, 1997. 46 (Suppl 1): p. 142A.
  30. Sosenko, J., et al., Use of the Diabetes Prevention Trial- Type 1 Risk Score (DPTRS) for improving the accuracy of the risk classification of type 1 diabetes. Diabetes Care, 2014. 37(4): p. 979-984.
  31. Mahon, J., et al., The TrialNet Natural History Study of the Development of Type 1 Diabetes: Objectives, design, and initial results. Pediatr Diabetes, 2009. 10(2): p. 97-104.
  32. Winkler, C., et al., Markedly reduced rate of diabetic ketoacidosis at onset of type 1 diabetes in relatives screened for islet autoantibodies. Pediatric Diabetes, 2012. 13(4): p. 308-313.
  33. Triolo, T., et al., Diabetic subjects diagnosed through the Diabetes Prevention Trial-Type 1 (DPT-1) are often asymptomatic with normal A1C at diabetes onset. Diabetes Care, 2009. 32: p. 769-773.
  34. Elding Larsson, H., et al., Reduced prevalence of diabetic ketoacidosis at diagnosis of type 1 diabetes in young children participating in longitudinal follow-up. Diabetes Care, 2011. 34(11): p. 2347-52.
  35. Rewers, A., et al., Presence of diabetic ketoacidosis at diagnosis of diabetes mellitus in youth: the Search for Diabetes in Youth Study. Pediatrics, 2008. 121: p. e1258- e1266.
  36. Barker, J., et al., Clinical characteristics of children diagnosed with type 1 diabetes through intensive screening and follow-up. Diabetes Care, 2004. 27(6): p. 1399-404.
  37. American Diabetes Association, I., American Diabetes Association Standards of Medical Care in Diabetes- 2021. Diabetes Care, 2021. 44 (Supplement 1): p. S18.
  38. General Population Level Estimation for Type 1 Diabetes Risk in Children 0-5 Years Old During Routine Care Delivery (PLEDGE). 2021 October 27, 2020 (accessed 2021 Dec 14); Available from: https://clinicaltrials.gov/ct2/show/NCT04477928?term=PL EDGE+study&cond=t1d&draw=2&rank=1.
  39. Rewers, M., et al., Newborn screening for HLA markers associated with IDDM: diabetes autoimmunity study in the young (DAISY). Diabetologia, 1996. 39(7): p. 807-12.
  40. Munchen, T.U., GPPAD-POInT (Global Platform of Autoimmune Diabetes - Primary Oral Insulin Trial).
  41. Barbara Davis Center for Diabetes, Autoimmunity Screening for Kids. 2019, March 13 (accessed 2021 Dec 14); Available from: https://www.askhealth.org/.
  42. Puff, R., et al., (Early diagnosis, early care--"Fr1da" screening of children for type 1 diabetes). MMW Fortschr Med, 2016. 158(4): p. 65-6.
  43. Megan AS Penno, J.J.C., Maria E Craig, et al. ENDIA Study Group, Environmental determinants of islet autoimmunity (ENDIA): a pregnancy to early life cohort study in children at-risk of type 1 diabetes, BMC Pediatr, 2013. 13-124: p. 1471-2431.
  44. Cardwell, C., et al., Maternal age at birth and childhood type 1 diabetes: a pooled analysis of 30 observational studies. Diabetes, 2010. 59(2): p. 486-494.
  45. Flood, T., S. Brink, and R. Gleason, Increased incidence of type 1 diabetes in children of older mothers. Diabetes Care, 1982. 5(6): p. 571-573.
  46. Warram, J.H., B.C. Martin, and A.S. Krolewski, Risk of IDDM in children of diabetic mothers decreases with increasing maternal age at pregnancy. Diabetes, 1991. 40(12): p. 1679-1684.
  47. Cardwell, C., et al., Birthweight and the risk of childhood- onset type 1 diabetes: a meta-analysis of observational studies using individual patient data. Diabetologia, 2010. 53(4): p. 641-651.
  48. Stene, L., et al., Birth weight and childhood onset type 1 diabetes: population-based cohort study. BMJ, 2001. 322(7291): p. 889-892.
  49. Dahlquist, G., S. Bennich, and B. Kallen, Intrauterine growth pattern and risk of childhood onset insulin dependent (type 1) diabetes: population based case- control study. BMJ, 1996. 313(7066): p. 1174-1177.
  50. Harder, T., et al., Birth weight, early weight gain, and subsequent risk of type 1 diabetes: systematic review and meta-analysis. Am J Epidemiol, 2009. 169(12): p. 1428-1436.
  51. Robertson, L. and K. Harrild, Maternal and neonatal risk factors for childhood type 1 diabetes: a Matched case- control study. BMC Public Health, 2010. 10: p. 281.
  52. Cardwell, C., et al., Birth order and childhood type 1 diabetes risk: a pooled analysis of 31 observational studies. Int J Epidemiol, 2011. 40(2): p. 363-374.
  53. Virtanen, S.M., et al., Microbial exposure in infancy and subsequent appearance of type 1 diabetes mellitus- associated autoantibodies: a cohort study. JAMA Pediatr, 2014. 168(8): p. 755-63.
  54. Cardwell, C., et al., Interbirth interval is associated with childhood type 1 diabetes risk. Diabetes, 2012. 61(3): p. 702-707.
  55. Cardwell, C., et al., Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: a meta-analysis of observational studies. Diabetologia, 2008. 51(5): p. 726-735.
  56. Khashan, A., et al., Mode of obstetrical delivery and type 1 diabetes: a sibling design study. Pediatrics, 2014. 134(3): p. e806-e813.
  57. Dahlquist, G. and B. Kallen, Maternal-child blood goup incompatibility and other perinatal events increase the risk for early-onset type 1 (insulin-dependent) diabetes mellitus. Diabetologia, 1992. 35(7): p. 671-675.
  58. Zhang, L., et al., Preterm birth and risk of type 1 and type 2 diabetes: systematic review and meta-analysis. Obes. Rev., 2014. 15(10): p. 804-811.
  59. Granfors, M., et al., No association between use of multivitamin supplement containing vitamin D during pregnancy and risk of Type 1 Diabetes in the child. Pediatr Diabetes, 2016. 17(7): p. 525-530.
  60. Stene, L.C. and G. Joner, Use of cod liver oil during the first year of life is associated with lower risk of childhood-onset type 1 diabetes: a large, population-based, case-control study. Am. J. Clin. Nutr, 2003. 78(6): p. 1128-1134.
  61. Fronczak, C., et al., In utero dietary exposures and risk of islet autoimmunity in children. Diabetes Care, 2003. 26(12): p. 3237-3242.
  62. Marjamaki, L., et al., Maternal intake of vitamin D during pregnancy and risk of advanced beta cell autoimmunity and type 1 diabetes in offspring. Diabetologia, 2010. 53(8): p. 1599-607.
  63. Kikkinen, A., et al., Use of antimicrobials and risk of type 1 diabetes in a population-based mother-child cohort. Diabetalogia, 2006. 49(1): p. 66-70.
  64. Arkkola, T., et al., Relationship of maternal weight status and weight gain rate during pregnancy to the development of advanced beta cell autoimmunity in the offspring: a prospective birth cohort study. Pediatr Diabetes, 2011. 12(5): p. 478-84.
  65. Niinisto, S., et al., Fatty acid status in infancy is associated with the risk of type 1 diabetes-associated autoimmunity. Diabetologia, 2017. 60(7): p. 1223-1233.
  66. Sorenson, I., et al., Serum long chain n-3 fatty acids (EPA and DHA) in the pregnant mother are independent of tisk of type 1 diabetes in the offspring. Diabetes Metab Res Rev, 2012. 28(5): p. 431-438.
  67. Ross, A., et al., The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab, 2011. 96(1): p. 53-38.
  68. Vehik. Kendra; Lynch, K.W., Matthew; et al. TEDDY Study Group, Prospective virome analyses in young children at increased genetic risk for type 1 diabetes. Nature Medicine, 2019. 25(12):1865-1872.
  69. Borch-Johnsen, K., et al., Relation between breast-feeding and incidence rates of insulin-dependent diabetes mellitus: A hypothesis. Lancet, 1984. 2(8411): p. 1083-1086.
  70. Chimel, R., et al., Early infant feeding and risk of developing islet autoimmunity and type 1 diabetes. Acta Diabetol, 2015 Jun;52(3): p. 621-624.
  71. Frederiksen, B., et al., Infant exposures and development of type 1 diabetes mellitus: The Diabetes Autoimmunity Study in the Young (DAISY). JAMA Pediatr, 2013. 167(9): p. 808-815.
  72. Lund-Blix, N., et al., Infant feeding in relation to islet autoimmunity and type 1 diabetes in genetically susceptible children: the MIDIA Study. Diabetes Care, 2015. 38(2): p. 257-263.
  73. Couper, J.J., et al., Lack of association between duration of breast-feeding or introduction of cow's milk and development of islet autoimmunity. Diabetes, 1999. 48(11): p. 2145-2149.
  74. Knip, M., et al., Hydrolyzed infant formula and early beta- cell autoimmunity: a randomized clinical trial. JAMA, 2014. 311(22): p. 2279-87.
  75. Knip, M., Writing group for teh TRIGR Study Group, Effect of Hydrolyzed Infant Formula vs Conventional Formula on Risk of Type 1 Diabetes. JAMA, 2018 Jan. 319(1): p. 38-48.
  76. Vaarala, O., et al., 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(7): p. 608-14.
  77. Ziegler, A.G., et al., Early infant feeding and risk of developing type 1 diabetes-associated autoantibodies. JAMA, 2003. 290(13): p. 1721-1728.
  78. Norris, J.M., et al., Timing of initial cereal exposure in infancy and risk of islet autoimmunity. JAMA, 2003. 290(13): p. 1713-1720.
  79. Hummel S, P.M., Hummel M, Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study. Diabetes Care, 2011. 34(6): p. 1302-1305.
  80. Beyerlein, A., et al., Timing of gluten introduction and islet autoimmunity in young children: updated results from the BABYDIET study. Diabetes Care, 2014. 37(9): p. e194- e195.
  81. Brekke, H. and J. Ludvigsson, Vitamin D supplementation and diabetes-related autoimmunity in the ABIS study. Pediatr Diabetes, 2007. 8(1): p. 11-14.
  82. Vitamin D supplement in early childhood and risk for Type I (insulin-dependent) diabetes mellitus. The EURODIAB Substudy 2 Study Group. Diabetologia, 1999. 42(1): p. 51-54.
  83. Hypponen, E., et al., Intake of vitamin D and risk of type 1 diabetes: a birth-cohort study. Lancet, 2001. 358(9292): p. 1500-1503.
  84. Zipitis, C. and A. Akobeng, Vitamin D supplementation in early childhood and risk of type 1 diabetes: a systematic review and meta-analysis. Arch Dis Child, 2008. 93(6): p. 512-517.
  85. Simpson, M., et al., No association of vitamin D intake or 25-hydroxyvitamin D levels in childhood with risk of islet autoimmunity and type 1 diabetes: the Diabetes Autoimmunity Study in the Young (DAISY). Diabetologia, 2011. 54(11): p. 2779-2788.
  86. Norris, J., et al., Omega-3 polyunsaturated fatty acid intake and islet autoimmunity in children at increased risk for type 1 diabetes. JAMA, 2007. 298(12): p. 1420-1428.
  87. Chase, H., et al., Effect of docosahexaenoic acid supplementation on inflammatory cytokine levels in infants at high risk for type 1 diabetes. Pediatr Diabetes, 2014.
  88. Wagner, C. and F. Greer, Prevention of rickets and vitamin D deficiency in infants, children, and adolescents. Pediatrics, 2008. 122(5): p. 1142-1152.
  89. Strachan, D.P., Hay fever, hygiene, and household size. BMJ, 1989. 299(6710): p. 1259-1260.
  90. Karelia map. 2015; (accessed 2021 Dec 14) Available from: https://commons.wikimedia.org/wiki/File:Karelia_today.pn g#/media/File:Karelia_today.png.
  91. Kondrashova, A., et al., The 'Hygiene hypothesis' and the sharp gradient in the incidence of autoimmune and allergic diseases between Russian Karelia and Finland. APMIS, 2013. 121(6): p. 478-93.
  92. Seiskari, T., et al., Allergic senstization and microbial load--a comparison between Finland and Russian Karelia. Clin Exp Immunol, 2007. 148(1): p. 47-52.
  93. Yatsunenko, T., et al., Human gut microbiome viewed across age and geography. Nature, 2012. 486(7402): p. 222-227.
  94. Giongo, A., et al., Toward defining the autoimmune microbiome for type 1 diabetes. ISME J, 2011. 5(1): p. 82-91.
  95. Brown, C.T., et al., Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One, 2011. 6(10): p. e25792.
  96. de Goffau, M.C., et al., Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes, 2013. 62(4): p. 1238-44.
  97. Bezirtzoglou, E., A. Tsiotsias, and G.W. Welling, Microbiota profile in feces of breast- and formula-fed newborns by using flourescence in situ hybridization (FISH). Anaerobe, 2011. 2011(17): p. 6.
  98. Stark, P. and A. Lee, The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J Med Microbiol, 1982. 15(2): p. 189-203.
  99. Menser, M., J. Forrest, and R. Bransby, Rubella infection and diabetes mellitus. Lancet, 1978. 1: p. 57-60.
  100. Oikarinen, S., et al., Enterovirus RNA in blood is linked to the development of type 1 diabetes. Diabetes, 2011. 60(1): p. 276-9.
  101. Kimpimaki, T., et al., The first signs of beta-cell autoimmunity appear in infancy in genetically susceptible children from the general population: the Finnish Type 1 Diabetes Prediction and Prevention Study. J Clin Endocrinol Metab, 2001. 86(10): p. 4782-8.
  102. Moltchanova, E., et al., Seasonal variation of diagnosis of type 1 diabetes mellitus in children worldwide. Diabet Med, 2009. 26(7): p. 673-678.
  103. Duderstadt, S., et al., Vaccination and risk of type 1 diabetes mellitus in active component U.S. military,

2002-2008. Vaccine, 2012. 30(4): p. 813-819.

  1. Graves, P., et al., Lack of association between early childhood immunizations and beta-cell autoimmunity. Diabetes Care, 1999. 22(10): p. 1694-1697.
  2. DeStefano, F., et al., Childhood vaccinations, vaccination timing, and risk of type 1 diabetes mellitus. Pediatrics, 2001. 108(6): p. E112.
  3. Hviid, A., et al., Childhood vaccination and type 1 diabetes. N Engl J Med, 2004. 350(14): p. 1398-1404.
  4. Elding Larsson, H., et al., Pandemrix(R) vaccination is not associated with increased risk of islet autoimmunity or type 1 diabetes in the TEDDY study children. Diabetologia, 2018. 61(1): p. 193-202.
  5. Breastfeeding and the use of human milk. Pediatrics, 2012. 129(3): p. e827-e841.

 

 

ADDITIONAL INFORMATION

(From prior chapter by Aaron W. Michels, MD and Peter Gottlieb, MD)

 

INTRODUCTION

 

Type 1 diabetes mellitus is defined as immune mediated diabetes mellitus (1-6). It can become manifest with hyperglycemia presenting in the first days of life or in adults over the age of 60. Current estimates indicate that immune mediated diabetes represents approximately 5 to 10% of the diabetes developing in adults and that approximately as many individuals develop this form of diabetes as adults as do children (7-9). In the United States the great majority (>90%) of Caucasian children developing diabetes have type 1 diabetes; whereas, approximately 50% of African American and Hispanic American children developing diabetes lack the autoantibody and immunogenetic markers of typical type 1 diabetes (10-12). Most of these latter children appear to have variants of type 2 diabetes with a small number having specific characteristic genetic syndromes (e.g. MODY: Maturity Onset Diabetes of Youth) with identified mutations of genes such as glucokinase and HNF (Hepatic Nuclear Factors) (13). In addition, studies of the pathology of the pancreas of Hispanic and African American children who lack islet autoantibodies show that all islets have some beta cells, but in decreased numbers (11). In contrast, in the pancreas of patients with type 1 diabetes, there is lobular loss of beta cells (termed pseudoatrophic islets) (11).

 

When an individual presents with type 1 diabetes it indicates that they and their relatives have an increased risk of having or developing a series of autoimmune disorders (12). Celiac disease, hypothyroidism, hyperthyroidism, Addison's disease, and pernicious anemia are some of the most prominent associated diseases. For example, approximately 1/20 patients with type 1 diabetes have celiac disease (14,15). Most of these patients are asymptomatic and the disorder is only discovered if anti-transglutaminase autoantibodies are measured and individuals with positive antibodies biopsied. In that the therapy for celiac disease, namely gluten avoidance, is highly effective, and we routinely screen all type 1 diabetic patients. We also screen for thyroid disease, which has an incidence of approximately 20% in type 1 diabetes, with yearly TSH measurements and for Addison's disease (21-hydroxylase autoantibodies) (16).

 

GENETIC SUSCEPTIBILITY

 

Type 1 diabetes is itself heterogeneous, with several forms of immune mediated diabetes with known genetic causes as parts of autoimmune syndromes (thus likely to be classified as other Specific Forms of Diabetes). In particular, patients develop immune mediated diabetes when they have mutations of the AIRE (Autoimmune Regulator) gene (21). Mutations of the AIRE gene result in Autoimmune Polyendocrine Syndrome Type I (23,24). Most forms of type 1 diabetes are polygenic in etiology, and polymorphisms of genes within the major histocompatibility complex (HLA genes) play a major role in determining disease susceptibility (27,28).

 

The alleles of different HLA genes (e.g., DRB1 and DQB1) are non-randomly associated with each other, such that with DRB1*0401 one usually finds one of three DQ alleles (e.g., DQB1*0301, DQB1*0302, DQB1*0303) rather than any one of more than forty different DQB molecules. Such non-random association of alleles of different genes on the same chromosome is termed linkage disequilibrium. The histocompatibility complex is divided into three regions, class II, class III and class I. The most important determinants of type 1 diabetes are the HLA DQ and DR alleles. These molecules on the surface of antigen presenting cells (e.g., macrophages) bind and present short peptides that are recognized by T cell receptors of T lymphocytes (27,35,36). They are termed immune response genes in that the specific amino acid sequence of these molecules determines which peptides will be bound and to a large extent determine which peptides an individual will respond to. Each different amino acid sequence is given a number. For the DQ molecules both its alpha and beta chain gene are polymorphic, and thus to specify a DQ molecule one must specify both chains. For DR molecules only the DRB chain is polymorphic and thus only this chain is specified. Each number after the star indicates a specific amino acid sequence of the HLA allele and the letters and first number the gene (e.g., DRB1*0401, DR B chain gene number 1, allele 0401).

 

There is a tremendous spectrum of diabetes risk associated with different DR and DQ genotypes (37-39) (Figure 8). For Caucasians with type 1 diabetes the most common diabetes-associated haplotypes are DR3 and DR4. More than 90% of patients with type 1A diabetes have one or both of these alleles versus approximately 40% of the general U.S. population. With the finer sequence information that is now available, DR4 haplotypes are subdivided based on specific variants of DRB1 and DQB1. The highest risk DR4 haplotypes have DRB1*0401, DRB1*0402, DRB1*0405, while DRB1*0403 is moderately protective. The highest risk DR4 haplotypes have DQB1*0302, with DQB1*0301 and DQB1*0303 of lower risk. Thus, both DR and DQ alleles contribute to diabetes risk. DR3 haplotypes are almost always conserved with DRB1*03 combined with DQA1*0501, DQB1*0201 (40). The highest risk genotype has both DR4/DR3 DQB1*0302/DQB1*0201. This genotype occurs in 2.4% of newborns in Denver, Colorado, and between 30 and 50% of children developing type 1 diabetes. Approximately 50% of children developing type 1 diabetes early (i.e., less than age 5) are DR3/4 heterozygotes versus 30% of young adults presenting with type 1A diabetes.

 

Figure 8. Hierarchy of diabetes risk with examples of haplotypes that lead to diabetes susceptibility, are neutral, or protective. Modified from teaching slides www.barbaradaviscenter.org

 

There are three HLA molecules that provide dominant protection. The most common is DQB1*0602 that occurs in approximately 20% of U.S. individuals (41-43). Protection is not absolute, but less than 1% of children with type 1 diabetes have this molecule. DQA1*0201 with DQB1*0303 and DRB1*1401 also provide dramatic protection, rarely being found in patients with type 1 diabetes and rarely transmitted from a parent with the alleles to their diabetic offspring (38,39). It is noteworthy that both DR and DQ alleles can protect. The specific mechanism underlying both susceptibility and protection are not fully understood. One attractive hypothesis is that protective alleles when expressed within the thymus lead to deletion of T cells with receptors that recognize a critical islet peptide (44). With deletion of such T cells, the risk of diabetes would be reduced. In addition, it is likely that high-risk HLA alleles present specific peptides of target islet molecules to T lymphocytes (28).

 

Multiple additional loci (Figure 7) have been implicated with estimates that approximately 50% of the familial aggregation of type 1 diabetes is attributable to the HLA region, perhaps 10% to the insulin locus, with all other loci contributing much less, though in aggregate their contribution is important. In the Cox analysis (Figure 7) of approximately 700 sibling pairs the only significant LOD score was for a locus on chromosome 16q that was not given an iddm designation with earlier genome screens. Several areas implicated in the past had suggestive scores, but there is overlap with the families from which the original evidence was generated. It is likely that contributing loci may differ between populations contributing to the initial difficulty of replicating putative loci in different studies (56,57). More than 40 genetic loci contributing to diabetes risk have been implicated (Figure 7). Polymorphisms of the insulin gene are well established as contributing to risk. A repeat sequence upstream (5') of the insulin gene termed a Variable nucleotide tandem repeat or VNTR, is divided into three general repeat sizes with the longest set of repeats associated with protection from diabetes (46-48). This set of alleles is also associated with greater thymic production of insulin messenger RNA (49), leading to the hypothesis that greater thymic message and presumably greater proinsulin production dampens anti-insulin autoimmunity (49-51). A functional polymorphism of the LYP gene (Lymphocyte Specific Phosphatase; PTPN22- Protein Tyrosine Phosphatase) has been associated with type 1 diabetes, rheumatoid arthritis, and lupus erythematosus (52-54). The R620W missense mutation (tryptophan replacing arginine) disrupts the binding of the phosphatase to the molecule Csk and this blocks its ability to down-regulate T cell receptor signaling. With an odds ratio of between 1.7 and 2.0 of the "autoimmunity” allele which is relatively common (5-10% allele frequency) there is a large genetic effect that is much greater than CTLA-4 polymorphisms associated with diabetes risk (55). Combining known diabetogenic polymorphisms of LYP, the insulin gene, alleles of DP, DQ, and DR class II immune response genes, as well all of the new loci account for approximately 48% of the familial aggregation of type 1A diabetes, with DR and DQ loci accounting for 41% of this 48% (45).  A recent study suggests that for a major subset of individuals with the highest risk HLA genotype (DR3/4-DQ2/DQ8 heterozygotes) who share both HLA haplotypes with a diabetic sibling, risk of activating anti-islet autoimmunity is as high as 80% (33).

 

AUTOIMMUNITY

 

Insulin autoantibodies are usually the first autoantibody to appear in children followed from birth for the development of type 1 diabetes (84,85). These autoantibodies can appear in the first six months of life. Once insulin autoantibodies appear in such young children there is a high risk of development of additional anti-islet autoantibodies and progression to diabetes. More than 90% of children developing type 1 diabetes prior to age 5 have insulin autoantibodies while less than 50% of children developing diabetes after age 12 have such autoantibodies (86). Therapy with human insulin induces insulin antibodies that cannot at present be distinguished from insulin autoantibodies. Thus, if an individual has been treated with insulin for more than several weeks, positive insulin autoantibodies are not interpretable. For all autoantibodies measured in the first 9 months of life, the antibodies may be transplacental in origin, a particular problem if a mother has type 1 diabetes and is treated with insulin.

 

There are a number of important caveats in the utilization of anti-islet autoantibody assays. The field developed from the initial observation that patient's sera "stained” islets of cut sections of human pancreas, the cytoplasmic islet cell antibody (ICA) assay (83). This assay, given its utilization of human pancreas from cadaveric donation and subjective reading of slides, has proven the most difficult to standardize (69). The assay predominantly detects antibodies reacting with GAD65, IA-2 and ZnT8, but does not detect anti-insulin autoantibodies. Given the difficulty in standardization, reliability over time, and major overlap with defined autoantibody assays, a number of investigators no longer utilize this assay. For research purposes and potentially in older adults with what has been termed LADA (latent autoimmune diabetes of adults) the ICA assay may have utility in that there is evidence of one or more additional autoantibodies detected with this assay and not with GAD65, IA-2, ZnT8 and insulin autoantibody determination.

 

A single autoantibody, even when present on multiple occasions, is associated with only a modest risk of progression to diabetes: approximately 10% (87,88). Once two or more anti-islet autoantibodies are present in children, progression to diabetes is very high, approaching almost 100% after 15 years of follow-up (89). In addition, once multiple autoantibodies are present it is very unusual for an individual to lose all expression of autoantibodies prior to the development of overt diabetes. Following the development of diabetes, IA-2 and more slowly GAD65 (over decades) autoantibodies wane. Following islet or pancreatic transplantation expression of GAD65 and IA-2 autoantibodies can be induced in patients with long-standing diabetes (90).

 

The most specific of the autoantibodies react with the molecule IA-2, but IA-2 autoantibodies are usually detected following the appearance of insulin and/or GAD65 autoantibodies (84). Even with IA-2 autoantibodies, however, there are apparent "false” positives in terms of diabetes risk. We evaluated approximately 10 individuals with either transient IA-2 autoantibodies or normal controls with IA-2 autoantibodies. None of these individuals expressed an additional anti-islet autoantibody. In contrast to patients diagnosed with or developing type 1 diabetes, the ICA512/IA-2 autoantibodies of nine out of ten of these normal individuals did not recognize multiple ICA512 epitopes and did not react with the dominant ICA512 autoantigenic domain (91). This indicates that even with a highly specific radioassay, if one screens tens of thousands of sera, one can find sera that presumably by chance cross-react with some epitope of the IA-2 molecule. It is much less likely to find an individual with antibodies that by chance react with two different islet autoantigens using fluid phase radioassays set with specificity at the 99th percentile of controls.

 

LOSS OF INSULIN SECRETION

 

At present, beta cell mass is not readily measured over time in humans, so it is not possible to absolutely define progression of beta cell loss. There is however no doubt that measurable anti-islet autoimmunity precedes the development of diabetes in terms of anti-islet autoantibodies in humans, and autoantibodies and T cell invasion in animal models. In the NOD mouse there is evidence of some beta cell destruction and beta cell regeneration prior to the onset of diabetes (92). There is also evidence for a change in the immune system close to the time of onset of diabetes (i.e., Th2 to Th1) (93-96). This change is associated with more rapid disease progression, ability to transfer diabetes by T cells, and a time window during which a specific immunotherapy (monoclonal anti-CD3 antibodies) is effective (97). In humans the best evidence for progressive loss of beta cell function comes from studies of insulin and C-peptide secretion (98). C-peptide, the connecting peptide of proinsulin, is secreted in equimolar amount to insulin, but C-peptide is not present in insulin preparations utilized to treat diabetes. Thus, C-peptide has become an important indicator of remaining beta cell function. Following the onset of diabetes, it has long been appreciated that C-peptide secretion progressively declines, until for most patients with type 1 diabetes C-peptide becomes non-detectable, associated with true insulin dependence. In a similar manner, first phase insulin secretion following a bolus of glucose on intravenous glucose tolerance testing is progressively lost for relatives followed to the development of type 1 diabetes (99). Such metabolic abnormalities may result in part from functional inhibition of beta cell secretion, but pathologic studies indicate that beta cell mass is normal for identical twins of patients that have not activated anti-islet autoimmunity, and for new onset patients that bulk of beta cells are destroyed (100). Within the pancreas of a patient with type 1 diabetes there is heterogeneity of islet lesions, with most islets lacking all beta cells and with no lymphocytic infiltrates (pseudoatrophic islets), few normal islets with no infiltrates, and few islets with remaining beta cells and infiltrates. This is perhaps analogous to the progressive development of vitiligo in patients, with patches of skin with all melanocytes destroyed, whereas other skin is normal.

 

OVERT DIABETES

 

The development of type 1 diabetes is usually perceived as an abrupt event, and some individuals may rapidly manifest severe hyperglycemia. Now that we can follow individuals to the development of type 1 diabetes, we can see that anti-islet autoantibodies can precede hyperglycemia by years, and there is usually some deterioration in glucose tolerance more than one year prior to diabetes onset (particularly with intravenous glucose tolerance testing) (101). The majority of individuals identified to be diabetic following autoantibody testing are found to have a diabetic 2-hour glucose on oral glucose tolerance testing (>200mg/dl) rather than fasting hyperglycemia. The acute presentation with severe hyperglycemia and ketoacidosis is life threatening, and it is estimated that approximately 1/200 children die at the onset of type 1 diabetes (102,103). Such children typically have a medical history where the first health care providers have failed to make the diagnosis of diabetes; the child then presents again later and dies with cerebral edema. The classic symptoms of polyuria, polydipsia, and weight loss are usually present but the initial diagnosis is still missed. The alternative diagnosis of nausea and vomiting due to viral illness is the most common mistaken diagnosis, and with the ready availability of glucose determination from a finger or heel stick, there should be a low threshold in emergency rooms and physicians’ offices for ruling out diabetes. Though transient hyperglycemia can occur, such children obviously need close follow up. We usually arrange glucose monitoring for children thought to have transient hyperglycemia, and measure anti-islet autoantibodies (104). Of those with anti-islet autoantibodies and transient hyperglycemia, almost all progress to type 1 diabetes within several months.

 

At the onset of type 1 diabetes, almost all individuals have residual insulin secretion, and there is convincing evidence that residual insulin secretion as measured by C-peptide secretion is of clinical benefit (less hypoglycemia, less microvascular complications, and much easier diabetes management).

 

REFERENCES

 

  1. Eisenbarth GS. Banting Lecture 2009: An unfinished journey: molecular pathogenesis to prevention of type 1A diabetes. Diabetes 2010; 59:759-774
  2. Todd JA. Etiology of type 1 diabetes. Immunity 2010; 32:457-467
  3. Stadinski B, Kappler J, Eisenbarth GS. Molecular targeting of islet autoantigens. Immunity 2010; 32:446-456
  4. Skyler JS. Immunomodulation for type 1 diabetes mellitus. International journal of clinical practice Supplement 2010:59-63
  5. Wong FS, Wen L. The study of HLA class II and autoimmune diabetes. Current molecular medicine 2003; 3:1-15
  6. Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet 2014; 383:69-82
  7. Lorenzen T, Pociot F, Hougaard P, et al. Long-term risk of IDDM in first-degree relatives of patients with IDDM. Diabetologia 1994; 37:321-327
  8. Janzon L, Bergentz SE, Ericsson BF, et al. The arm-ankle pressure gradient in relation to cardiovascular risk factors in intermittent claudication. Circulation 1981; 63:1339-1341
  9. Zimmet P, Turner R, McCarty D, et al. Crucial points at diagnosis. Type 2 diabetes or slow type 1 diabetes. Diabetes care 1999; 22 Suppl 2:B59-64
  10. Pinhas-Hamiel O, Dolan LM, Daniels SR, et al. Increased incidence of non-insulin-dependent diabetes mellitus among adolescents. The Journal of pediatrics 1996; 128:608-615
  11. Gianani R, Campbell-Thompson M, Sarkar SA, et al. Dimorphic histopathology of long-standing childhood-onset diabetes. Diabetologia 2010; 53:690-698
  12. Triolo TM, Armstrong TK, McFann K, et al. Additional Autoimmune Disease Found in 33% of Patients at Type 1 Diabetes Onset. Diabetes care 2011; 34:1211-1213
  13. McCarthy MI, Hattersley AT. Learning from molecular genetics: novel insights arising from the definition of genes for monogenic and type 2 diabetes. Diabetes 2008; 57:2889-2898
  14. Bao F, Yu L, Babu S, et al. One third of HLA DQ2 homozygous patients with type 1 diabetes express celiac disease-associated transglutaminase autoantibodies. Journal of autoimmunity 1999; 13:143-148
  15. Hoffenberg EJ, Bao F, Eisenbarth GS, et al. Transglutaminase antibodies in children with a genetic risk for celiac disease. The Journal of pediatrics 2000; 137:356-360
  16. Baker PR, Baschal EE, Fain PR, et al. Dominant suppression of Addison's disease associated with HLA-B15. The Journal of clinical endocrinology and metabolism 2011; 96:2154-2162
  17. Atkinson MA, Eisenbarth GS. Type 1 diabetes: new perspectives on disease pathogenesis and treatment. Lancet 2001; 358:221-229
  18. Eisenbarth GS. Type I diabetes mellitus. A chronic autoimmune disease. The New England journal of medicine 1986; 314:1360-1368
  19. Diagnosis and classification of diabetes mellitus. Diabetes care 2004; 27 Suppl 1:S5-S10
  20. Imagawa A, Hanafusa T, Miyagawa J, et al. A novel subtype of type 1 diabetes mellitus characterized by a rapid onset and an absence of diabetes-related antibodies. Osaka IDDM Study Group. The New England journal of medicine 2000; 342:301-307
  21. Robles DT, Eisenbarth GS, Ikegami H, et al. Endocrinology and metabolism clinics of North America. Philadelphia: W.B. Saunders; 2002.
  22. Patel DD. Escape from tolerance in the human X-linked autoimmunity-allergic disregulation syndrome and the Scurfy mouse. The Journal of clinical investigation 2001; 107:155-157
  23. Bjorses P, Aaltonen J, Horelli-Kuitunen N, et al. Gene defect behind APECED: a new clue to autoimmunity. Human molecular genetics 1998; 7:1547-1553
  24. Michels AW, Eisenbarth GS. Autoimmune polyendocrine syndrome type 1 (APS-1) as a model for understanding autoimmune polyendocrine syndrome type 2 (APS-2). Journal of internal medicine 2009; 265:530-540
  25. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature immunology 2003; 4:330-336
  26. Owen CJ, Jennings CE, Imrie H, et al. Mutational analysis of the FOXP3 gene and evidence for genetic heterogeneity in the immunodysregulation, polyendocrinopathy, enteropathy syndrome. The Journal of clinical endocrinology and metabolism 2003; 88:6034-6039
  27. Klein J, Sato A. The HLA system. First of two parts. The New England journal of medicine 2000; 343:702-709
  28. Lee KH, Wucherpfennig KW, Wiley DC. Structure of a human insulin peptide-HLA-DQ8 complex and susceptibility to type 1 diabetes. Nature immunology 2001; 2:501-507
  29. Mordes JP, Greiner DL, Rossini AA. Animal models of autoimmune diabetes mellitus. . In: LeRoith D, Taylor SI, Olefsky JM, eds. Diabetes mellitus : a fundamental and clinical text. Philadelphia: Lippincott-Raven; 1996:349-360.
  30. Martin AM, Maxson MN, Leif J, et al. Diabetes-prone and diabetes-resistant BB rats share a common major diabetes susceptibility locus, iddm4: additional evidence for a "universal autoimmunity locus" on rat chromosome 4. Diabetes 1999; 48:2138-2144
  31. Yu B, Gauthier L, Hausmann DH, et al. Binding of conserved islet peptides by human and murine MHC class II molecules associated with susceptibility to type I diabetes. European journal of immunology 2000; 30:2497-2506
  32. Redondo MJ, Jeffrey J, Fain PR, et al. Concordance for islet autoimmunity among monozygotic twins. The New England journal of medicine 2008; 359:2849-2850
  33. Aly TA, Ide A, Jahromi MM, et al. Extreme genetic risk for type 1A diabetes. Proceedings of the National Academy of Sciences of the United States of America 2006; 103:14074-14079
  34. Redondo MJ, Fain PR, Eisenbarth GS. Genetics of type 1A diabetes. Recent progress in hormone research 2001; 56:69-89
  35. Kwon OJ, Brautbar C, Weintrob N, et al. Immunogenetics of HLA class II in Israeli Ashkenazi Jewish, Israeli non-Ashkenazi Jewish, and in Israeli Arab IDDM patients. Human immunology 2001; 62:85-91
  36. Undlien DE, Lie BA, Thorsby E. HLA complex genes in type 1 diabetes and other autoimmune diseases. Which genes are involved? Trends in genetics : TIG 2001; 17:93-100
  37. Lie BA, Ronningen KS, Akselsen HE, et al. Application and interpretation of transmission/disequilibrium tests: transmission of HLA-DQ haplotypes to unaffected siblings in 526 families with type 1 diabetes. American journal of human genetics 2000; 66:740-743
  38. Redondo MJ, Kawasaki E, Mulgrew CL, et al. DR- and DQ-associated protection from type 1A diabetes: comparison of DRB1*1401 and DQA1*0102-DQB1*0602*. The Journal of clinical endocrinology and metabolism 2000; 85:3793-3797
  39. Kawasaki E, Noble J, Erlich H, et al. Transmission of DQ haplotypes to patients with type 1 diabetes. Diabetes 1998; 47:1971-1973
  40. Erlich H, Valdes AM, Noble J, et al. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes 2008; 57:1084-1092
  41. Baisch JM, Weeks T, Giles R, et al. Analysis of HLA-DQ genotypes and susceptibility in insulin-dependent diabetes mellitus. The New England journal of medicine 1990; 322:1836-1841
  42. Pugliese A, Gianani R, Moromisato R, et al. HLA-DQB1*0602 is associated with dominant protection from diabetes even among islet cell antibody-positive first-degree relatives of patients with IDDM. Diabetes 1995; 44:608-613
  43. Pugliese A, Kawasaki E, Zeller M, et al. Sequence analysis of the diabetes-protective human leukocyte antigen-DQB1*0602 allele in unaffected, islet cell antibody-positive first degree relatives and in rare patients with type 1 diabetes. The Journal of clinical endocrinology and metabolism 1999; 84:1722-1728
  44. Barrett JC, Clayton DG, Concannon P, et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nature genetics 2009; 41:703-707
  45. Todd JA, Walker NM, Cooper JD, et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nature genetics 2007; 39:857-864
  46. 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
  47. Undlien DE, Bennett ST, Todd JA, et al. Insulin gene region-encoded susceptibility to IDDM maps upstream of the insulin gene. Diabetes 1995; 44:620-625
  48. Barratt BJ, Payne F, Lowe CE, et al. Remapping the insulin gene/IDDM2 locus in type 1 diabetes. Diabetes 2004; 53:1884-1889
  49. Pugliese A, Brown D, Garza D, et al. Self-antigen-presenting cells expressing diabetes-associated autoantigens exist in both thymus and peripheral lymphoid organs. The Journal of clinical investigation 2001; 107:555-564
  50. Pugliese A, Zeller M, Fernandez A, Jr., et al. 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. Nature genetics 1997; 15:293-297
  51. Bennett ST, Wilson AJ, Esposito L, et al. Insulin VNTR allele-specific effect in type 1 diabetes depends on identity of untransmitted paternal allele. The IMDIAB Group. Nature genetics 1997; 17:350-352
  52. Bottini N, Musumeci L, Alonso A, et al. A functional variant of lymphoid tyrosine phosphatase is associated with type I diabetes. Nature genetics 2004; 36:337-338
  53. Kyogoku C, Langefeld CD, Ortmann WA, et al. Genetic association of the R620W polymorphism of protein tyrosine phosphatase PTPN22 with human SLE. American journal of human genetics 2004; 75:504-507
  54. Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. American journal of human genetics 2004; 75:330-337
  55. Ueda H, Howson JM, Esposito L, et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003; 423:506-511
  56. Noble JA, Valdes AM, Varney MD, et al. HLA class I and genetic susceptibility to type 1 diabetes: results from the Type 1 Diabetes Genetics Consortium. Diabetes 2010; 59:2972-2979
  57. Burren OS, Adlem EC, Achuthan P, et al. T1DBase: update 2011, organization and presentation of large-scale data sets for type 1 diabetes research. Nucleic acids research 2011; 39:D997-1001
  58. Abiru N, Maniatis AK, Yu L, et al. Peptide and major histocompatibility complex-specific breaking of humoral tolerance to native insulin with the B9-23 peptide in diabetes-prone and normal mice. Diabetes 2001; 50:1274-1281
  59. Heath VL, Hutchings P, Fowell DJ, et al. Peptides derived from murine insulin are diabetogenic in both rats and mice, but the disease-inducing epitopes are different: evidence against a common environmental cross-reactivity in the pathogenicity of type 1 diabetes. Diabetes 1999; 48:2157-2165
  60. Wen L, Chen NY, Tang J, et al. The regulatory role of DR4 in a spontaneous diabetes DQ8 transgenic model. The Journal of clinical investigation 2001; 107:871-880
  61. Ellerman KE, Like AA. Susceptibility to diabetes is widely distributed in normal class IIu haplotype rats. Diabetologia 2000; 43:890-898
  62. Ellerman KE, Richards CA, Guberski DL, et al. Kilham rat triggers T-cell-dependent autoimmune diabetes in multiple strains of rat. Diabetes 1996; 45:557-562
  63. Wucherpfennig KW, Eisenbarth GS. Type 1 diabetes. Nature immunology 2001; 2:767-768
  64. Stadinski BD, Zhang L, Crawford F, et al. Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register. Proceedings of the National Academy of Sciences of the United States of America 2010; 107:10978-10983
  65. Michels AW. Targeting the trimolecular complex. Clin Immunol 2013; 149:339-344
  66. Ginsberg-Fellner F, Witt ME, Yagihashi S, et al. Congenital rubella syndrome as a model for type 1 (insulin-dependent) diabetes mellitus: increased prevalence of islet cell surface antibodies. Diabetologia 1984; 27 Suppl:87-89
  67. Shaver KA, Boughman JA, Nance WE. Congenital rubella syndrome and diabetes: a review of epidemiologic, genetic, and immunologic factors. American annals of the deaf 1985; 130:526-532
  68. Rabinowe SL, George KL, Loughlin R, et al. Congenital rubella. Monoclonal antibody-defined T cell abnormalities in young adults. The American journal of medicine 1986; 81:779-782
  69. Lonnrot M, Korpela K, Knip M, et al. Enterovirus infection as a risk factor for beta-cell autoimmunity in a prospectively observed birth cohort: the Finnish Diabetes Prediction and Prevention Study. Diabetes 2000; 49:1314-1318
  70. Graves PM, Norris JM, Pallansch MA, et al. The role of enteroviral infections in the development of IDDM: limitations of current approaches. Diabetes 1997; 46:161-168
  71. Rewers M, Norris JM. Epidemiology of type I diabetes. In: Eisenbarth GS, Lafferty KJ, eds. Type I diabetes : molecular, cellular, and clinical immunology. New York: Oxford University Press; 1996:172-208.
  72. Martin JM, Trink B, Daneman D, et al. Milk proteins in the etiology of insulin-dependent diabetes mellitus (IDDM). Annals of medicine 1991; 23:447-452
  73. Norris JM, Beaty B, Klingensmith G, et al. 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
  74. Norris JM, Barriga K, Klingensmith G, et al. Timing of initial cereal exposure in infancy and risk of islet autoimmunity. Jama 2003; 290:1713-1720
  75. Ziegler AG, Schmid S, Huber D, et al. Early infant feeding and risk of developing type 1 diabetes-associated autoantibodies. Jama 2003; 290:1721-1728
  76. Yang Z, Wang K, Li T, et al. Childhood diabetes in China. Enormous variation by place and ethnic group. Diabetes care 1998; 21:525-529
  77. Onkamo P, Vaananen S, Karvonen M, et al. Worldwide increase in incidence of Type I diabetes--the analysis of the data on published incidence trends. Diabetologia 1999; 42:1395-1403
  78. Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. The New England journal of medicine 2002; 347:911-920
  79. Wenzlau JM, Moua O, Sarkar SA, et al. SlC30A8 is a major target of humoral autoimmunity in type 1 diabetes and a predictive marker in prediabetes. Annals of the New York Academy of Sciences 2008; 1150:256-259
  80. Bonifacio E, Atkinson M, Eisenbarth G, et al. International Workshop on Lessons From Animal Models for Human Type 1 Diabetes: Identification of Insulin but Not Glutamic Acid Decarboxylase or IA-2 as Specific Autoantigens of Humoral Autoimmunity in Nonobese Diabetic Mice. Diabetes 2001; 50:2451-2458
  81. Margolis DJ, Gupta J, Hoffstad O, et al. Lack of effectiveness of hyperbaric oxygen therapy for the treatment of diabetic foot ulcer and the prevention of amputation: a cohort study. Diabetes care 2013; 36:1961-1966
  82. Miao D, Guyer KM, Dong F, et al. GAD65 autoantibodies detected by electrochemiluminescence assay identify high risk for type 1 diabetes. Diabetes 2013; 62:4174-4178
  83. Bottazzo GF, Florin-Christensen A, Doniach D. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet 1974; 2:1279-1283
  84. Bonifacio E, Scirpoli M, Kredel K, et al. Early autoantibody responses in prediabetes are IgG1 dominated and suggest antigen-specific regulation. J Immunol 1999; 163:525-532
  85. Yu L, Robles DT, Abiru N, et al. Early expression of antiinsulin autoantibodies of humans and the NOD mouse: evidence for early determination of subsequent diabetes. Proceedings of the National Academy of Sciences of the United States of America 2000; 97:1701-1706
  86. Vardi P, Ziegler AG, Mathews JH, et al. Concentration of insulin autoantibodies at onset of type I diabetes. Inverse log-linear correlation with age. Diabetes care 1988; 11:736-739
  87. Verge CF, Gianani R, Kawasaki E, et al. Prediction of type I diabetes in first-degree relatives using a combination of insulin, GAD, and ICA512bdc/IA-2 autoantibodies. Diabetes 1996; 45:926-933
  88. Bingley PJ, Bonifacio E, Williams AJ, et al. Prediction of IDDM in the general population: strategies based on combinations of autoantibody markers. Diabetes 1997; 46:1701-1710
  89. Ziegler AG, Rewers M, Simell O, et al. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. Jama 2013; 309:2473-2479
  90. Bosi E, Braghi S, Maffi P, et al. Autoantibody response to islet transplantation in type 1 diabetes. Diabetes 2001; 50:2464-2471
  91. Clark CM, Jr. How should we respond to the worldwide diabetes epidemic? Diabetes care 1998; 21:475-476
  92. Sreenan S, Pick AJ, Levisetti M, et al. Increased beta-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. Diabetes 1999; 48:989-996
  93. Gazda LS, Charlton B, Lafferty KJ. Diabetes results from a late change in the autoimmune response of NOD mice. Journal of autoimmunity 1997; 10:261-270
  94. Dilts SM, Lafferty KJ. Autoimmune diabetes: the involvement of benign and malignant autoimmunity. Journal of autoimmunity 1999; 12:229-232
  95. Andre I, Gonzalez A, Wang B, et al. Checkpoints in the progression of autoimmune disease: lessons from diabetes models. Proceedings of the National Academy of Sciences of the United States of America 1996; 93:2260-2263
  96. Shimada A, Charlton B, Taylor-Edwards C, et al. Beta-cell destruction may be a late consequence of the autoimmune process in nonobese diabetic mice. Diabetes 1996; 45:1063-1067
  97. Chatenoud L, Primo J, Bach JF. CD3 antibody-induced dominant self tolerance in overtly diabetic NOD mice. J Immunol 1997; 158:2947-2954
  98. Rosenbauer J, Herzig P, von Kries R, et al. Temporal, seasonal, and geographical incidence patterns of type I diabetes mellitus in children under 5 years of age in Germany. Diabetologia 1999; 42:1055-1059
  99. Chase HP, Cuthbertson DD, Dolan LM, et al. First-phase insulin release during the intravenous glucose tolerance test as a risk factor for type 1 diabetes. The Journal of pediatrics 2001; 138:244-249
  100. Foulis AK, McGill M, Farquharson MA, et al. A search for evidence of viral infection in pancreases of newly diagnosed patients with IDDM. Diabetologia 1997; 40:53-61
  101. Srikanta S, Ganda OP, Rabizadeh A, et al. First-degree relatives of patients with type I diabetes mellitus. Islet-cell antibodies and abnormal insulin secretion. The New England journal of medicine 1985; 313:461-464
  102. Laing SP, Swerdlow AJ, Slater SD, et al. The British Diabetic Association Cohort Study, II: cause-specific mortality in patients with insulin-treated diabetes mellitus. Diabetic medicine : a journal of the British Diabetic Association 1999; 16:466-471
  103. Laing SP, Swerdlow AJ, Slater SD, et al. The British Diabetic Association Cohort Study, I: all-cause mortality in patients with insulin-treated diabetes mellitus. Diabetic medicine : a journal of the British Diabetic Association 1999; 16:459-465
  104. Herskowitz RD, Wolfsdorf JI, Ricker AT, et al. Transient hyperglycemia in childhood: identification of a subgroup with imminent diabetes mellitus. Diabetes Res 1988; 9:161-167
  105. Barker JM, Goehrig SH, Barriga K, et al. Clinical characteristics of children diagnosed with type 1 diabetes through intensive screening and follow-up. Diabetes care 2004; 27:1399-1404