Archives

Mineralocorticoid Defects in Children

ABSTRACT

 

Isolated aldosterone deficiency in children related either to impaired secretion by the adrenal gland or to aldosterone resistance in target tissues is rare. The incidence is estimated to be <1:1,000,000 for congenital isolated primary hypoaldosteronism and 1:66,000 to 1:166,000 for congenital aldosterone resistance (1). Children may present with salt wasting, hyponatremia, hypotension, hyperkalemia, metabolic acidosis, and failure to thrive. There is a wide phenotypic spectrum based on the severity and etiology of aldosterone deficiency or action. In this chapter, we briefly discuss the physiology of mineralocorticoids in newborns, categorize the causes of isolated hypoaldosteronism, and review the etiologies to guide clinical and laboratory evaluation and treatment.

 

INTRODUCTION

 

Mineralocorticoids are a class of steroids produced in the zona glomerulosa in the adrenal cortex that regulate sodium, potassium and water balance; aldosterone is the primary mineralocorticoid. Its synthesis involves several enzymes within the adrenal, the final step regulated by aldosterone synthase (CYP11B2) (Figure 1). Aldosterone secretion involves an intricate feedback loop involving multiple organs including the adrenal glands, kidneys, liver, lungs, and blood vessels. The major regulators of aldosterone synthesis and secretion are the renin-angiotensin-aldosterone (RAA) axis and potassium (Figure 2).  Aldosterone binds to the mineralocorticoid receptor at the kidney to activate specific amiloride-sensitive sodium (ENaC) channels and a Na-K- ATPase pump. Through these actions, aldosterone promotes sodium reabsorption and urinary potassium excretion (Figure 2).

 

Mineralocorticoid deficiency (also referred to as hypoaldosteronism) refers to compromised aldosterone secretion from the adrenal glands or its cellular action. Hypoaldosteronism is observed as part of global adrenal cortex dysfunction in both congenital and acquired disorders, such as primary adrenal insufficiency (PAI), adrenal hypoplasia congenita (AHC), and congenital adrenal hyperplasia (CAH). In these disorders, hypoaldosteronism occurs together with glucocorticoid deficiency (i.e., adrenal insufficiency) and/or other deficient or dysregulated adrenal steroid secretion. While rare in children, hypoaldosteronism may occur  as an isolated condition, either congenital or acquired, and can be classified into 1) defective aldosterone biosynthesis 2) disturbances in stimulation of aldosterone secretion, and 3) impaired aldosterone action at the target tissue, mainly the kidneys (resistance) (2). The latter is also referred to as “pseudohypoaldosteronism” since circulating aldosterone levels are elevated despite clinical symptoms and signs of mineralocorticoid deficiency due to dysfunctional mineralocorticoid receptor or its downstream effects (2). In this chapter, we discuss isolated aldosterone-deficient conditions other than PAI, AHC, and CAH.  In depth coverage of adrenal insufficiency can be found in Endotext.org chapter: Adrenal Insufficiency in Children (3).

 

Normal aldosterone production, regulation, and action are essential in neonates, infants, and children for salt balance and overall growth. If untreated, defects in aldosterone secretion or action in children may lead to salt wasting, hypotension, hyperkalemia, metabolic acidosis, and failure to thrive. Severe hyponatremia (salt wasting) and metabolic acidosis can be life-threatening in newborns and infants. In depth coverage of mineralocorticoid deficiency and resistance can be found in Endotext.org chapter: Aldosterone Deficiency and Resistance (4). Our chapter focuses on isolated aldosterone defects in the pediatric population.

 

Figure 1. Enzyme defects related to aldosterone synthesis. Schematic of adrenal steroidogenesis demonstrating the various enzymes involved in aldosterone synthesis (large black box). The red lines indicate the specific enzymatic defects that result in defects in aldosterone synthesis. Cortisol circulates in the bloodstream at higher concentrations than aldosterone and it also interacts with MR. However, within the kidney and target tissues, there is selectivity of MR by aldosterone due to the enzyme 11βHSD2 that converts active cortisol to inactive cortisone (small black box). SCC: side-chain cleavage. HSD: hydroxysteroid dehydrogenase. MR: mineralocorticoid receptor. DHEA: dehydroepiandrosterone. Aldo: aldosterone.

 

PATHOPHYSIOLOGY

 

Aldosterone Synthesis

 

Aldosterone biosynthesis occurs at the zona glomerulosa, the outermost layer of the adrenal cortex, via the action of several enzymes: cholesterol desmolase [also known as cholesterol side-chain cleavage enzyme] (CYP11A1), 3β-hydroxysteroid dehydrogenase (HSD3B2), 21-hydroxylase (CYP21A2), 11-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) (Figure1). The first four enzymes are also expressed in the zona fasciculata and are involved in cortisol biosynthesis. Defects in any of these enzymes may lead to combined aldosterone/cortisol deficiencies as part of the syndromes seen in Congenital Adrenal Hyperplasia. Aldosterone synthase encoded by CYP11B2, the last enzymatic step in aldosterone biosynthesis, is expressed only at the zona glomerulosa and genetic defects in this gene result in isolated aldosterone deficiency (Figure 1).

 

Aldosterone synthesis involves two steps. The first includes the 18-hydroxylation of corticosterone to form 18-hydroxycorticosterone (18OH corticosterone) and the second is the 18-oxidation of 18OH corticosterone to form aldosterone. Although it was previously considered that these two steps are catalyzed by two different enzymes, it is now known to involve the same enzyme, aldosterone synthase (5). Based on the two final steps in aldosterone synthesis, two subtypes of aldosterone synthase deficiency (ASD) have been described; however, with further clarification of the enzymatic process this is now thought to be an overlapping clinical spectrum, depending on the degree of enzyme deficiency (5).

 

Aldosterone Regulation and Action

 

Serum potassium concentrations and the Renin, Angiotensin, Aldosterone (RAA) axis are the main regulators of aldosterone synthesis. Hyperkalemia has a direct stimulating effect independent of RAA axis (2). The RAA axis is a feedback loop that regulates sodium, potassium, water, fluid volume, and blood pressure (2). The cells in the macula densa of the juxtaglomerular apparatus are triggered to release renin in response to a drop in perfusion. Angiotensinogen is a protein produced from the liver that is cleaved to angiotensin I (Ang I) by renin (2). Angiotensin-converting enzyme (ACE) in vascular endothelium rapidly converts Ang I to Angiotensin II (Ang II).  Ang II is the most potent stimulus for aldosterone production and release (2). Of note, tissue and plasma peptidase inactivate angiotensin within minutes and circulating renin levels are the rate-limiting factor of this process (1).

 

Aldosterone mediates its effects by binding to the mineralocorticoid receptor (MR; aka NR3C2) at the distal convoluted tubules and collecting duct epithelial cells of the kidneys (Figure 2). The MR is a member of the nuclear receptor family, and along with the glucocorticoid and androgen receptors, forms the steroid receptor subfamily. In its unliganded state, the MR is located in the cytoplasm. Upon binding with its ligand, MR is translocated into the nucleus, where it modulates the transcription of several genes, such as those that encode the ENaC subunits (1). Mutations that inactivate the MR result in aldosterone resistance or pseudo-hypoaldosteronism type 1 (PHA1).

 

Aldosterone, 11-deoxycorticosterone (DOC), and cortisol are all endogenous agonists of the MR. Specifically, cortisol and aldosterone have an equal affinity for the mineralocorticoid receptor (2); however, selectivity of MR receptor for aldosterone is ensured in epithelial target tissues by 11βHSD2 enzyme that converts active cortisol to inactive cortisone (1) (Figure 1). This is of particular importance as cortisol circulates at concentrations 100 to 1,000-fold higher than aldosterone. Loss-of-function mutations of the kidney 11βHSD2 result in excessive cortisol-dependent MR activation and cause an autosomal recessive form of familial hypertension called apparent mineralocorticoid excess (6).

 

After binding to MR, aldosterone activates ENaC gene transcription, decreases ENaC degradation, and activates Na-K ATPase pump. ENaC, located at the apical membrane of epithelial cells, plays a crucial role in sodium reabsorption, potassium secretion, and subsequent volume expansion. ENaC consists of 3 subunits (a, b, and g) that are encoded by unique genes (SCNN1A, SCNN1B, SCNN1G, respectively) (1). Defects in these genes can impair ENaC function and lead also to aldosterone resistance or pseudo hypoaldosteronism type Ib. In addition to the epithelial cells of the distal convoluted tubule, ENaC is expressed at the epithelial cells of other tissues that are involved in salt conservation, such as colon, sweat glands, and lungs. Dysfunction of ENaC, therefore, has systemic manifestations from muti-organ water and salt loss.

 

Figure 2. Physiology of aldosterone secretion and action. The figure demonstrates the renin-angiotensin-aldosterone (RAA) system and its effects on sodium and potassium homeostasis, and blood pressure. Aldosterone secretion is regulated by decreased blood volume and hyponatremia via activation of the RAA axis, and indirectly, by hyperkalemia. Aldosterone then binds to the mineralocorticoid receptor (MR; aka NR3C2) at the distal convoluted tubules and collecting duct of the kidneys. Upon binding with aldosterone, MR translocate into the nucleus, where it modulates the transcription of the genes that encode the epithelial sodium channel (ENaC). ENaC is a sodium-selective ion channel that plays a crucial role in sodium reabsorption. Aldosterone action results in urinary potassium excretion and sodium reabsorption, and thus, increased blood volume.

 

Aldosterone Secretion in the Newborn

 

There are limited studies in infants investigating the interaction between water, sodium, and the renin-angiotensin-aldosterone system. Various changes related to water turnover, sodium metabolism, and kidney adaptation to extrauterine life occur in the neonatal period (1). The immediate postnatal phase in the first week of life is characterized by oliguria followed by a diuretic phase with extracellular contraction and net loss of sodium and water (1). Maximum weight loss occurs during this period (up to 10% of birth weight is considered normal). Kidneys in the neonate  exhibit tubular immaturity resulting in sodium wasting and impaired ability to reabsorb water (1). Additionally, aldosterone and renin concentrations are higher in the newborn period, whereas expression of renal MR is reduced, leading to transient renal resistance to aldosterone (7). In very preterm infants,  there is decreased activity of 11β-hydroxylase (CYP11B1) and low aldosterone synthase (CYP11B2) activity, possibly due to immaturity of these enzymes in the fetal adrenal cortex, leading to deficient aldosterone secretion (8, 9). After the first week of life, water losses decrease, and positive sodium balance is important for growth (1). It is essential to acknowledge these physiologic changes when evaluating mineralocorticoid function in the neonatal period.

 

CLINICAL PRESENTATION

 

The clinical presentation of aldosterone deficiency is variable depending on the etiology. Broadly, the signs of hypoaldosteronism include hypotension, hyponatremia (salt wasting), hyperkalemia, and metabolic acidosis. The symptoms that can be seen in infants and children related to these electrolyte derangements are dehydration, vomiting, irritability, weakness, seizures, and failure to thrive.

 

ETIOLOGY OF ISOLATED HYPOALDOSTERONISM

 

Isolated aldosterone disorders can be classified into disorders of defective synthesis, aldosterone resistance and diminished stimulation (Table 1).

 

Defective Aldosterone Synthesis

 

This refers to hyperreninemic hypoaldosteronism in which the renin production is intact, and the defect is at the level of the adrenal gland. The etiology of defective aldosterone synthesis can be separated into congenital and acquired causes. It is important to note that aldosterone deficiency due to defect in synthesis can be the first presenting sign of adrenal cortex failure and later progress to involve insufficient cortisol production. For descriptions of disorders involving adrenal cortical failure such as congenital adrenal hyperplasia and Addison’s disease, see Endotext.org chapter: Adrenal Insufficiency in Children (3).  

 

CONGENITAL CAUSES

 

Prematurity

 

Very preterm infants (<33 weeks’ gestation) have deficient aldosterone concentrations, thought to be related to both the general immaturity of the fetal adrenal cortex and specifically a defect in aldosterone production, perhaps due to low aldosterone synthase activity (10). This also aligns with other defects in adrenal steroidogenesis seen in preterm infants (e.g. low 11β-hydroxylation leading to high 17OHP and false positive on the newborn screening) (11).

 

Aldosterone Synthase Deficiency

 

Variants in the CYP11B2 gene result in variable loss of enzyme activity and aldosterone deficiency. As seen in Figure 1, aldosterone synthase is responsible for the hydroxylation of corticosterone to 18-hydroxycorticosterone followed by oxidation from 18-hydroxycorticosterone to aldosterone. Previously, these steps were thought to be controlled by 2 different enzymes and this disorder was called corticosterone methyl-oxidase (CMO) deficiency with 2 subtypes described (CMOI and CMOII) based on the aldosterone and precursor relative concentrations. These subtypes are now thought to be a spectrum of severity (12). Due to continued production of DOC and corticosterone, there is some mineralocorticoid activity. However, this may be insufficient in the setting of aldosterone resistance of the neonate and salt loss may occur in infancy. Children are more affected than adults who may even have normal renin levels as the mineralocorticoid sensitivity improves and exogenous salt from table food intake increases.

 

ACQUIRED CAUSES

 

Critical Illness

 

Despite intact ACTH and renin secretion as well as angiotensin II production, a portion of critically ill patients may  have low aldosterone levels (13, 14). This  is considered to represent a shift in the adrenal cortex prioritizing cortisol production to aid in recovery.

 

Adrenalectomy

 

Typically, unilateral adrenalectomy would not be expected to lead to a glucocorticoid or mineralocorticoid defect; however, this may occur in the setting of a hyperfunctioning defect in one adrenal with contralateral atrophy. In the case of mineralocorticoid function, a patient with Conn’s syndrome (also known as primary hyperaldosteronism) who undergoes unilateral adrenalectomy can experience signs and symptoms of hypoaldosteronism including hyperkalemia, with reports indicating this occurrence in 6-62% of patients (15-17). Post surgical monitoring is recommended, although few patients require medical treatment.  

 

Medication Induced

 

While other medications may lead to diminished aldosterone stimulation or resistance, heparin is  known to reduce aldosterone synthesis leading to natriuresis and hyperkalemia without an impact on corticosteroid production (18).

 

Aldosterone Resistance

 

This refers to impaired action of aldosterone at the level of the target tissue and can further be categorized into congenital and acquired causes.

 

CONGENITAL CAUSES

 

Pseudo-Hypoaldosteronism (PHA) Type 1

 

The genetic form of aldosterone resistance occurs due to a mutation impacting the mineralocorticoid receptor (19). Despite the prevalent consideration of PHA1 as a genetic form of type IV renal tubular acidosis (RTA), the biochemical profile can differ. Hyperkalemia, hyponatremia, and acidosis are universal; however, while RTA type IV involves a hyperchloremic non-anion gap acidosis, there are descriptions of both hyper and hypochloremia as well as an anion gap acidosis in PHA1 (20). PHA can be either autosomal dominant or recessive. The autosomal recessive disease (PHA1b) occurs due to a mutation in  the genes encoding one of the 3 ENaC subunits (SCNN1A, SCNN1B, SCNN1G) (21). The presentation of PHA1b is often severe given the systemic nature of ENaC outside of the kidney and in the epithelial cells of other tissues including colon, sweat glands, and lungs. The autosomal dominant disease (PHA1a) occurs due to a mutation in the gene encoding the mineralocorticoid receptor (NR3C2) and is restricted to the kidney (21, 22). This form is milder and tends to improve during childhood. However, despite its isolation to the kidney, the hyperkalemia that results can be devastating if not identified and treated early;  cases are described involving cardiac arrest and hypoxic ischemic encephalopathy as an outcome (23).  

 

Despite the classification of  PHA1 based on mutation (PHA1a vs. PHA1b), some individuals with features of PHA1 do not have identifiable molecular defects.  

 

ACQUIRED CAUSES

 

Secondary Pseudo-Hypoaldosteronism (PHA Type 3)

 

PHA Type 3 is often associated with urinary tract infections (UTI) and/or related to underlying urinary anomalies, primarily urinary tract obstruction, resulting in decreased aldosterone responsiveness (24). PHA Type 3 occurs frequently in male infants;  a recent systematic review identified 80% of cases in male babies under 4 months of age (25). Presentation can include failure to thrive and vomiting and laboratory evaluation reveals hyperreninemic, hyperaldosteronism with impaired responsiveness, and hyponatremic, hyperkalemic metabolic acidosis. Early identification allows for prevention of electrolyte related morbidity and expedited resolution of urinary obstruction through surgical management in over 40% of cases (24, 25).

 

 

Medications that block the ENaC channel (amiloride) or MR (spironolactone) will cause aldosterone resistance. These medications are used therapeutically in resistant hypertension and to prevent hypokalemia seen with other diuretics. Spironolactone is also used for its anti-androgenic properties and the potential for dehydration and hyperkalemia should be considered and monitored. Other ENaC blockers include triamterene, trimethoprim, and pentamidine, while other aldosterone antagonists include synthetic progestins and calcineurin inhibitors.  

 

Diminished Stimulation

 

Decreased renin or angiotensin II results in decreased aldosterone production due to diminished adrenal stimulation. When this hyporeninemic hypoaldosteronism occurs with hyperchloremia and non-anion gap metabolic acidosis, it is called Type 4 RTA. In adults, this is most often associated with nephropathy (diabetes, autonomic neuropathy, sickle cell disease, HIV, SLE) and medications (beta blockers, ACE inhibitors) (26). In children, Gordon Syndrome (Familial Hyperkalemic hypertension or pseudo-hypoaldosteronism type II [PHA2]) is rare and associated with low renin/low aldosterone (or inappropriately normal for degree of hyperkalemia) state with normal glomerular filtration. This is thought to be due to abnormal thiazide-sensitive sodium-chloride co-transporter in the distal nephron (mutations in WNK1, WNK4, CUL3, or KLHL3 genes) (27). The increased sodium and chloride reabsorption leads to hypertension, volume expansion, and decreased potassium and hydrogen excretion resulting in hyperkalemia and metabolic acidosis. In contrast to PHA1, PHA2 does have a biochemical profile that aligns with Type IV RTA including hyponatremic, hyperkalemic, and hyperchloremic non-anion gap metabolic acidosis (28). These causes of defective aldosterone stimulation are rare in the pediatric population, so when identified, affected children should be referred to the appropriate subspecialities such as nephrology for evaluation and treatment. Given the rare nature and lack of a primary endocrine etiology, these causes are not reviewed in the table below.

 

Table 1. CAUSES OF ISOLATED ALDOSTERONE DEFECTS IN CHILDREN

 

Condition

Cause

Presentation

Congenital – Aldosterone Synthesis

 

Prematurity (transient)

Immaturity of aldosterone synthase in very premature infants

HYPERreninemic,HYPOaldosteronism

 

Hyponatremia and hyperkalemia

Increased corticosterone

 

 

Aldosterone synthase deficiency

Formerly divided into:

CMO I deficiency (low 18-OH corticosterone)

-CMO II deficiency (high 18-OH corticosterone)

 

Autosomal recessive or autosomal dominant (mixed penetrance) variant in CYP11B2

Acquired – Aldosterone Synthesis

Critical illness

Thought to represent a shift in the adrenal cortex prioritizing cortisol production to aid in recovery

Adrenalectomy

Can occur in the setting of a hyperfunctioning lesion with contralateral atrophy

Medication induced

Heparin

Congenital – Aldosterone Resistance

Systemic Pseudo-hypoaldosteronism (PHA1b)

Autosomal recessive variant in ENaC gene (SCNN1A, SCNN1B, SCNN1G)

HYPERreninemicHYPERaldosteronism (pseudo-hypoaldosteronism)

 

Hyponatremia and hyperkalemia (electrolytes may be normal in mild cases)

 

Renal Pseudo-hypoaldosteronism (PHA1a)

Autosomal dominant variant in MR receptor gene (NR3C2)

Acquired – Aldosterone Resistance

Secondary Pseudo-hypoaldosteronism (type 3 PHA)

Associated with urinary tract infections

Secondary (medication induced) pseudo-hypoaldosteronism

Meds that block ENaC (amiloride, triamterene, trimethoprim, pentamidine), meds that block MR receptor (spironolactone, synthetic progestins, calcineurin inhibitors)

 

DIAGNOSTIC APPROACH

 

Defects of aldosterone synthesis or action in children should be suspected in the setting of dehydration, hyponatremia (salt wasting), and hyperkalemia. The clinical phenotype varies depending on the etiology and some infants or children may present only with mild electrolyte abnormalities and failure to thrive. Additionally, the causes of hyponatremia in children are broad, and may include iatrogenic causes due to hypotonic fluid, central nervous system or lung disease causing syndrome of inappropriate antidiuretic hormone (SIADH), excess ingestion of free water, and high salt losses due to diarrhea. Determining volume status and urinary sodium content are starting points for refining the etiology of hyponatremia. Mineralocorticoid deficiency is characterized by hypovolemic hyponatremia with high urine sodium. The other causes of hyponatremia will not be discussed in this chapter.

 

Differential Diagnosis and Laboratory Evaluation

 

The first step in the evaluation of a child with suspected mineralocorticoid deficiency is to determine whether there is associated adrenal insufficiency (figure 3). The evaluation for adrenal insufficiency includes measurement of serum cortisol (ideally morning level depending on the clinical scenario and age of patient), ACTH, 17-hydroxyprogesterone (17OHP), and possible provocative testing (ACTH stimulation test). Plasma renin activity and serum aldosterone should be measured to evaluate for mineralocorticoid deficiency. If there is global adrenal dysfunction resulting in both cortisol and aldosterone deficiency, the differential diagnosis can be narrowed to causes of primary adrenal insufficiency (see Endotext: Adrenal Insufficiency in Children) (3). It is critical to identify primary adrenal insufficiency, especially in infants, and promptly treat with hydrocortisone to avoid adrenal crisis.

 

If  an isolated aldosterone defect is considered, the second step is to evaluate whether the defect is at the level of the adrenal glands or kidneys. High renin and low aldosterone points to a defect at the level of the adrenal glands (defective aldosterone synthesis). High renin and high aldosterone points to a defect at the level of the kidneys causing resistance to aldosterone. The various causes of aldosterone resistance are detailed above in the section “Etiology”. Briefly, these include congenital (mutations in MR or ENaC channel) and acquired causes (medications, transient resistance in the setting of UTI, or renal tubular dysfunction). Low renin and low aldosterone states do not commonly occur in children, as they are often the consequence of chronic illness causing type IV RTA (i.e. in adults with diabetic nephropathy); however, there is also a genetic form, Gordon Syndrome or pseudo-hypoaldosteronism type 2 which is characterized by hypertension, hyperkalemia, and metabolic acidosis.

 

As  stated above, measurement of renin and aldosterone at the time of hyponatremia and hyperkalemia are important biochemical markers to differentiate the etiology of hypoaldosteronism. Furthermore, if there is suspicion for aldosterone synthase deficiency, corticosterone and 18-hydroxycorticosterone measurements can be useful (see figure 1). Values need to be interpreted according to age of the patient. Hemolyzed lblood may result in a falsely elevated potassium level and must be repeated to ensure accuracy of test values.

 

Figure 3. A proposed approach in the differential and diagnostic evaluation of children with suspected aldosterone deficiency.

 

Genetic Testing

 

In addition to biochemical evaluation, genetic testing is an invaluable tool to help guide treatment and prognosis, especially in infanta and children where the clinical manifestations of aldosterone defects vary widely (29). Genetic testing including whole exome sequencing or gene panels (for pseudo-hypoaldosteronism) may  clarify the diagnosis, treatment, and prognosis. Genes associated with hypoaldosteronism and pseudo-hypoaldosteronism include CYP11B2, NR3C2, SCNN1A, SCNN1B, SCNN1G, WNK1, WNK4, CUL3, KLHL3 (2).

 

TREATMENT

 

The initial management depends upon severity of presentation and etiology of the mineralocorticoid defect. Infants or children who are acutely ill with salt-wasting crisis must undergo fluid resuscitation to correct salt and water losses. It is essential to give stress dose corticosteroids (intramuscular or intravenous hydrocortisone 100mg/m2) if co-existing glucocorticoid deficiency exists. Hydrocortisone at high doses has mineralocorticoid effect, and fludrocortisone tablets may be added once hydrocortisone is weaned to be below about 50-60 mg/m2/day (3).

 

Oral treatment options for children with aldosterone defects include mineralocorticoid replacement (fludrocortisone), sodium chloride tablets, and sodium bicarbonate. The management plan depends on the underlying mineralocorticoid defect and is separated according to those children who are not able to produce aldosterone, and those who have resistance to its action.

 

Primary Hypoaldosteronism

 

Children with primary hypoaldosteronism (including those with adrenal insufficiency such as Addison’s Disease or CAH) should start mineralocorticoid replacement (fludrocortisone 0.05-0.2 mg/day). Infants and young children usually need higher doses of fludrocortisone in addition to sodium chloride supplementation due to renal resistance and general diet that is lower in salt. Sodium chloride is weaned over time as renin activity normalizes, and salt is incorporated into the diet. Fludrocortisone is continued for mineralocorticoid replacement and titrated based on normalization of blood pressure, electrolytes, and renin levels.

 

Aldosterone Resistance

 

Children with autosomal recessive (PHA1b) and autosomal dominant (PHA1a) pseudo-hypoaldosteronism are usually treated with high dose sodium chloride supplementation. Those who have PHA1a (mild form only affecting the kidneys) usually need lower doses of salt supplementation with gradual clinical improvement (typically no need for salt supplementation by 1-3 years of age) (30).  Infants and children with the severe/systemic form (PHA1b) are more difficult to manage given the need for higher doses of salt supplementation, potassium lowering agents, and potential for recurrent pulmonary infections (31). Some of these children might need gastrostomy tubes to allow for consistent high dose salt supplementation which is not always tolerated by mouth. Sodium bicarbonate is another medication used to improve metabolic acidosis which can impact growth and development if acidosis persists. Given the rarity of pseudo-hypoaldosteronism, the doses of sodium chloride and sodium bicarbonate are not well established and must be titrated based on serum sodium and bicarbonate concentrations. 

 

Table 2. SUMMARY OF TREATMENT OPTIONS FOR CHILDREN WITH ALDOSTERONE DEFECTS:

Treatment

Dose

Considerations

Fludrocortisone

0.05-0.2 mg/day

Once or twice daily

Doses titrated based on blood pressure, electrolytes, and renin levels.

Sodium chloride (salt tablets)

2 g/day or 2-5 mEq/kg daily

1-gram NaCl tablets = 17mEq

Higher/more frequent doses in babies and weaned down as they get older.

Doses titrated based on sodium levels.

Sodium bicarbonate or sodium citrate/citric acid

2-3 mEq/kg daily

Titrate based on bicarbonate levels

 

CONCLUSION

 

Isolated defects in aldosterone synthesis or action are rare in children; however, it is important to identify these disorders to prevent life-threatening complications. Infants may present with salt wasting crisis while older children may present with failure to thrive, mild hyponatremia, and metabolic acidosis. The two major categories of isolated hypoaldosteronism include aldosterone synthesis defects and aldosterone resistance. There are several genes associated with isolated hypoaldosteronism, and genetic testing is an important diagnostic tool. Treatment and prognosis depend on the underlying etiology.

 

REFERENCES

 

  1. Bizzarri C, Pedicelli S, Cappa M, Cianfarani S. Water Balance and ‘Salt Wasting' in the First Year of Life: The Role of Aldosterone-Signaling Defects. Hormone Research in Paediatrics. 2016;86(3):143-53.
  2. Rajkumar V WM. Hypoaldosteronism. Treasure Island (FL): StatPearls Publishing; 2023.
  3. Kilberg M, Vogiatzi M. Adrenal Insufficiency in Children. EndoText. 2024.
  4. Arai K, Papadopoulou-Marketou N, Chrousos G. Aldosterone Deficiency and Resistance. EndoText. 2020.
  5. White PC. Aldosterone synthase deficiency and related disorders. Mol Cell Endocrinol. 2004;217(1-2):81-7.
  6. Carvajal CA, Tapia-Castillo A, Vecchiola A, Baudrand R, Fardella CE. Classic and Nonclassic Apparent Mineralocorticoid Excess Syndrome. J Clin Endocrinol Metab. 2020;105(4).
  7. Martinerie L, Viengchareun S, Delezoide AL, Jaubert F, Sinico M, Prevot S, et al. Low renal mineralocorticoid receptor expression at birth contributes to partial aldosterone resistance in neonates. Endocrinology. 2009;150(9):4414-24.
  8. Martinerie L, Pussard E, Yousef N, Cosson C, Lema I, Husseini K, et al. Aldosterone-Signaling Defect Exacerbates Sodium Wasting in Very Preterm Neonates: The Premaldo Study. The Journal of Clinical Endocrinology & Metabolism. 2015;100(11):4074-81.
  9. Travers S, Martinerie L, Boileau P, Lombes M, Pussard E. Alterations of adrenal steroidomic profiles in preterm infants at birth. Arch Dis Child Fetal Neonatal Ed. 2018;103(2):F143-F51.
  10. Martinerie L, Pussard E, Yousef N, Cosson C, Lema I, Husseini K, et al. Aldosterone-Signaling Defect Exacerbates Sodium Wasting in Very Preterm Neonates: The Premaldo Study. J Clin Endocrinol Metab. 2015;100(11):4074-81.
  11. Kamrath C, Hartmann MF, Boettcher C, Wudy SA. Reduced activity of 11beta-hydroxylase accounts for elevated 17alpha-hydroxyprogesterone in preterms. J Pediatr. 2014;165(2):280-4.
  12. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol. 2018;179(3):R125-R41.
  13. Findling JW, Waters VO, Raff H. The dissociation of renin and aldosterone during critical illness. J Clin Endocrinol Metab. 1987;64(3):592-5.
  14. Trimarchi T. Endocrine problems in critically ill children: an overview. AACN Clin Issues. 2006;17(1):66-78.
  15. Starker LF, Christakis I, Julien JS, Schwarz K, Graham P, Grubbs EG, et al. Considering Postoperative Functional Hypoaldosteronism after Unilateral Adrenalectomy. Am Surg. 2017;83(6):598-604.
  16. Queiroz NL, Stumpf MAM, Souza VCM, Maciel AAW, Fagundes GFC, Okubo J, et al. Renal Function Evolution and Hypoaldosteronism Risk After Unilateral Adrenalectomy for Primary Aldosteronism. Horm Metab Res. 2024;56(5):350-7.
  17. Shariq OA, Bancos I, Cronin PA, Farley DR, Richards ML, Thompson GB, et al. Contralateral suppression of aldosterone at adrenal venous sampling predicts hyperkalemia following adrenalectomy for primary aldosteronism. Surgery. 2018;163(1):183-90.
  18. Oster JR, Singer I, Fishman LM. Heparin-induced aldosterone suppression and hyperkalemia. Am J Med. 1995;98(6):575-86.
  19. Tajima T, Morikawa S, Nakamura A. Clinical features and molecular basis of pseudohypoaldosteronism type 1. Clin Pediatr Endocrinol. 2017;26(3):109-17.
  20. Adachi M, Nagahara K, Ochi A, Toyoda J, Muroya K, Mizuno K. Acid-Base Imbalance in Pseudohypoaldosteronism Type 1 in Comparison With Type IV Renal Tubular Acidosis. J Endocr Soc. 2022;6(12):bvac147.
  21. Furgeson SB, Linas S. Mechanisms of type I and type II pseudohypoaldosteronism. J Am Soc Nephrol. 2010;21(11):1842-5.
  22. Geller DS, Rodriguez-Soriano J, Vallo Boado A, Schifter S, Bayer M, Chang SS, et al. Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet. 1998;19(3):279-81.
  23. Tauber KA, Ermacor K, Listman J. Cardiac arrest in a newborn: A case of pseudohypoaldosteronism. Clin Case Rep. 2024;12(2):e8265.
  24. Moreno Sanchez A, Garcia Atares A, Molina Herranz D, Antonanzas Torres I, Romero Salas Y, Ruiz Del Olmo Izuzquiza JI. Secondary pseudohypoaldosteronism: a 15-year experience and a literature review. Pediatr Nephrol. 2024;39(11):3233-9.
  25. Betti C, Lavagno C, Bianchetti MG, Kottanattu L, Lava SAG, Schera F, et al. Transient secondary pseudo-hypoaldosteronism in infants with urinary tract infections: systematic literature review. Eur J Pediatr. 2024;183(10):4205-14.
  26. Mustaqeem R AA. Renal Tubular Acidosis. Treasure Island (FL): StatPearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK519044/.
  27. Manas F, Singh S. Pseudohypoaldosteronism Type II or Gordon Syndrome: A Rare Syndrome of Hyperkalemia and Hypertension With Normal Renal Function. Cureus. 2024 Jan 19;16(1).
  28. Adachi M, Motegi S, Nagahara K, Ochi A, Toyoda J, Mizuno K. Classification of pseudohypoaldosteronism type II as type IV renal tubular acidosis: results of a literature review. Endocr J. 2023;70(7):723-9.
  29. Turan I, Kotan LD, Tastan M, Gurbuz F, Topaloglu AK, Yuksel B. Molecular genetic studies in a case series of isolated hypoaldosteronism due to biosynthesis defects or aldosterone resistance. Clin Endocrinol (Oxf). 2018;88(6):799-805.
  30. Krishna S, Augustian M. Autosomal Dominant Pseudohypoaldosteronism Type 1 in a Newborn With Failure to Thrive. Cureus. 2024;16(4):e59356.
  31. Karacan Küçükali G ÇS, Tunç G, Oğuz MM, Çelik N, Akkaş KY, Şenel S, Güleray Lafcı N, Savaş Erdeve Ş. Clinical Management in Systemic Type Pseudohypoaldosteronism Due to SCNN1B Variant and Literature Review. J Clin Res Pediatr Endocrinol. 2021;13(4):446-51.

 

Normal Physiology of Growth Hormone in Adults

ABSTRACT

 

Growth hormone (GH) is an ancestral hormone, secreted episodically from somatotroph cells in the anterior pituitary. Since the recognition of its multiple and complex effects in the early 1960s, the physiology and regulation of GH has become a major area of research interest in the field of endocrinology. In adulthood its main role is to regulate metabolism. Pituitary synthesis and secretion of GH is stimulated by episodic hypothalamic secretion of GH releasing factor and inhibited by somatostatin. Insulin-like Growth Factor I (IGF-I) inhibits GH secretion by a negative loop at both hypothalamic and pituitary levels. In addition, age, gender, pubertal status, food, exercise, fasting, sleep, and body composition play important regulatory roles. GH acts both directly through its own receptors and indirectly through the induced production of IGF-I. Their effects may be synergic (stimulate growth) or antagonistic as for the effect on glucose metabolism: GH stimulates lipolysis and promotes insulin resistance, whereas IGF-I acts as an insulin agonist. The bioactivity of IGF-I is tightly controlled by a multitude of IGF-I binding globulins. The mechanisms to explain the insulin antagonist effect of GH in humans are causally linked to lipolysis and ensuing elevated levels of circulating free fatty acids (FFA). The nitrogen retaining properties of GH predominantly involve stimulation of protein synthesis, which could be either direct or mediated through IGF-I, insulin, or lipid intermediates. In this chapter the normal physiology of GH secretion and the effects of GH on intermediary metabolism throughout adulthood, focusing on human studies, are presented.

 

INTRODUCTION

 

Harvey Cushing proposed in 1912 in his monograph "The Pituitary Gland" the existence of a "hormone of growth” and was thereby among the first to indicate that the primary action of growth hormone (GH) was to control and promote skeletal growth. In clinical medicine GH (also called somatotrophin) was previously known for its role on promoting growth of hypopituitary children, and for its adverse effects in connection with hypersecretion as observed in acromegaly. The multiple and complex actions of human GH were, however, acknowledged shortly after the advent of pituitary derived preparation of the hormone in the late fifties, as reviewed by Raben in 1962 (1).

 

In the present chapter we will briefly review the normal physiology of GH secretion and the effects of GH on intermediary metabolism throughout adulthood. Other important physiological effects of GH are presented in the review on GH replacement in adults.

 

GROWTH HORMONE

 

GH is a single chain protein with 191 amino-acids and two disulfide bonds. The human GH gene is located on chromosome 17q22 as part of a locus that comprises five genes. In addition to two GH related genes (GH1 that codes for the main adult growth hormone, produced in the somatotrophic cells found in the anterior pituitary gland and, to a minor extent, in lymphocytes, and GH2 that codes for the placental GH), there are three genes coding for chorionic somatomammotropin (CSH1, CSH2 and CSHL) (also known as placental lactogen) genes (2,3). The GH1 gene encodes two distinct GH isoforms (22 kDa and 20 kDa). The principal and most abundant GH form in pituitary and blood is monomeric 22K-GH isoform, representing also the recombinant GH available for therapeutic use (and subsequently for doping purposes) (3). Administration of recombinant 22K-GH exogenously leads to a decrease in the 20K-GH isoform and testing both isoforms is used to detect GH doping in sports (4).

 

As already mentioned, GH is secreted by the somatotroph cells located primarily in the lateral wings of the anterior pituitary. A recent single cell RNA sequencing study performed in mice showed that GH-expressing cells, representing the somatotrophs, are the most abundant cell population in adult pituitary gland (5). The differentiation of somatotroph cell is governed by the pituitary transcription factor 1 (Pit-1). Data in mice suggests that the pituitary holds regenerative competence, the GH-producing cells being regenerated from the pituitary’s stem cells in young animals after a period of 5 months (6).

 

Physiological Regulation of GH Secretion

 

The morphological characteristics and number of somatotrophs are remarkably constant throughout life, while the secretion pattern changes. GH secretion occurs in a pulsatile fashion, and in a circadian rhythm with a maximal release in the second half of the night. So, sleep is an important physiological factor that increases GH release. Interestingly, the maximum GH levels occur within minutes of the onset of slow wave sleep and there is marked sexual dimorphism of the nocturnal GH increase in humans, constituting only a fraction of the total daily GH release in women, but the bulk of GH output in men (7).

 

GH secretion is also gender, pubertal status, and age dependent (Figure 1) (8). Integrated 24 h GH concentration is significantly greater in women than in men and greater in the young than in the old adults. The serum concentration of free estradiol, but not free testosterone, correlates with GH and when correcting for the effects of estradiol, neither gender nor age influence GH concentration. This suggests that estrogens play a crucial role in modulating GH secretion (8). During puberty, a 3-fold increase in pulsatile GH secretion occurs that peaks around the age of 15 years (yr) in girls and 1 yr later in boys (9).

 

Figure 1. The secretory pattern of GH in young and old females and males. In young individuals the GH pulses are larger and more frequent and females secrete more GH than men (modified from (8)).

 

Pituitary synthesis and secretion of GH is stimulated by episodic hypothalamic hormones. Growth hormone releasing hormone (GHRH) stimulates while somatostatin (SST) inhibits GH production and release. GH stimulates IGF-I production which in turn inhibits GH secretion at both hypothalamic and pituitary levels. The gastric peptide ghrelin is also a potent GH secretagogue, which acts to boost hypothalamic GHRH secretion and synergize with its pituitary GH-stimulating effects (Figure 2) (10). Interestingly, recently germline or somatic duplication of GPR101 constitutively activates the cAMP pathway in the absence of a ligand, leading to GH release. Although GPR101 physiology is unclear it is worth mentioning it since it clearly has an effect on GH physiology (11).

 

In addition, a multitude of other factors may impact the GH axis most probably due to interaction with GHRH, somatostatin, and ghrelin. Estrogens stimulate the secretion of GH but inhibit the action of GH on the liver by suppressing GH receptor (GHR) signaling. In contrast, androgens enhance the peripheral actions of GH (12). Exogenous estrogens potentiate pituitary GH responses to submaximal effective pulses of exogenous GHRH (13) and mutes inhibition by exogenous SST (14). Also, exogenous estrogen potentiates ghrelin action (15).

 

GH release correlates inversely with intraabdominal visceral adiposity via mechanisms that may depend on increased FFA flux, elevated insulin, or free IGF-I.

 

Figure 2. Factors that stimulate and suppress GH secretion under physiological conditions.

 

GROWTH HORMONE RELEASING HORMONE (GHRH)

 

GHRH is a 44 amino-acid polypeptide produced in the arcuate nucleus of the hypothalamus. These neuronal terminals secrete GHRH to reach the anterior pituitary somatotrophs via the portal venous system, which leads to GH transcription and secretion. Moreover, animal studies have demonstrated that GHRH plays a vital role in the proliferation of somatotrophs in the anterior pituitary, whereas the absence of GHRH leads to anterior pituitary hypoplasia (16). In addition, GHRH upregulates GH gene expression and stimulates GH release (17). The secretion of GHRH is stimulated by several factors including depolarization, α2-adrenergic stimulation, hypophysectomy, thyroidectomy, and hypoglycemia and it is inhibited by somatostatin, IGF-I, and activation of GABAergic neurons.

 

GHRH acts on the somatotrophs via a seven trans-membrane G protein-coupled stimulatory cell-surface receptor. This receptor has been extensively studied over the last decade leading to the identification of several important mutations. Point mutations in the GHRH receptors, as illustrated by studies done on the lit/lit dwarf mice, showed a profound impact on subsequent somatotroph proliferation leading to anterior pituitary hypoplasia (18). Unlike the mutations in the Pit-1 and PROP-1 genes, which lead to multiple pituitary hormone deficiencies and anterior pituitary hypoplasia, mutations in the GHRH receptor leads to profound GH deficiency with anterior pituitary hypoplasia. Subsequently to the first GHRH receptor mutation described in 1996 (19) an array of familial GHRH receptor mutations have been recognized over the last decade. These mutations account for almost 10% of the familial isolated GH deficiencies. An affected individual will present with short stature and a hypoplastic anterior pituitary. However, they lack certain typical features of GH deficiency such as midfacial hypoplasia, microphallus, and neonatal hypoglycemia (20).

 

SOMATOSTATIN  (SST)

 

SST is a cyclic peptide, encoded by a single gene in humans, which mostly exerts inhibitory effects on endocrine and exocrine secretions. Many cells in the body, including specialized cells in the anterior periventricular nucleus and arcuate nucleus, produce SST. These neurons secrete SST into the adenohypophyseal portal venous system, via the median eminence, to exert effect on the anterior pituitary. SST has a short half-life of approximately 2 minutes as it is rapidly inactivated by tissue peptidase in humans. The secretion of SST by the hypothalamic neurons is inhibited by high blood glucose and is stimulated by serum GH/IGF-I level, exercise, and immobilization (21).

 

SST acts via a seven trans-membrane, G protein coupled receptor and, thus far, five subtypes of the receptor have been identified in humans (SSTR1-5). Although all five receptor subtypes are expressed in the human fetal pituitary, adult pituitary only express 4 subtypes (SSTR1, SSTR2, SSTR3, SSTR5). Out of these four subtypes, somatotrophs exhibit more sensitivity to SSTR2 and SSTR5 ligands in inhibiting the secretion of GH in a synergistic manner (22).

 

GHRELIN

 

Ghrelin is a 28 amino-acid peptide that is the natural ligand for the GH secretagogue receptor. In fact, ghrelin and GHRH have a synergistic effect in increasing circulating GH levels (7). Ghrelin is primarily secreted by stomach and may be involved in the GH response to fasting and food intake.

 

Clinical Implications

 

GH LEVELS- INFLUENCE ON BODY COMPOSISTION, PHYSICAL FITNESS, AND AGE

 

With the introduction of dependable radioimmunological assays, it was recognized that circulating GH is blunted in obese subjects, and that normal aging is accompanied by a gradual decline in GH levels (23,24). It has been hypothesized that many of the senescent changes in body composition and organ function are related to or caused by decreased GH (25), also known as "the somatopause".

 

Studies done in the late 90s have uniformly documented that adults with severe GH deficiency are characterized by increased fat mass and reduced lean body mass (LBM) (26). It is also known that normal GH levels can be restored in obese subjects following massive weight loss (27), and that GH substitution in GH-deficient adults normalizes body composition. What remains unknown is the cause-effect relationship between decreased GH levels and senescent changes in body composition. Is the propensity for gaining fat and losing lean body mass initiated or preceded by a primary age-dependent decline in GH secretion and action? Alternatively, accumulation of fat mass secondary to non-GH dependent factors (e.g., lifestyle, dietary habits) results in a feedback inhibition of GH secretion. Moreover, little is known about possible age-associated changes in GH pharmacokinetics and bioactivity.

 

Cross sectional studies done to assess the association between body composition and stimulated GH release in healthy subjects show that, adult people (mean age 50 yr) have a lower peak GH response to secretagogues (clonidine and arginine), and females had a higher response to arginine when compared to males. Multiple regression analysis, however, reveal that intra-abdominal fat mass is the most important and negative predictor of peak GH levels as previously  mentioned (Figure 3) (28). In the same population, 24-h spontaneous GH levels also predominantly correlated inversely with intra-abdominal fat mass (29).

 

Figure 3. Correlation between intra-abdominal fat mass and 24-hour GH secretion.

 

A detailed analysis of GH secretion in relation to body composition in elderly subjects has, to our knowledge, not been performed. Instead, serum IGF-I has been used as a surrogate or proxy for GH status in several studies of elderly men (30-32). These studies comprise large populations of ambulatory, community-dwelling males aged between 50-90 yr. As expected, the serum IGF-I declined with age (Figure 4), but IGF-I failed to show any significant association with body composition or physical performance.

 

Figure 4. Changes in serum IGF-I with age; modified from (33).

 

GH ACTION - INFLUENCE OF AGE, SEX, AND BODY COMPOSITION

 

Considering the great interest in the actions of GH in adults, surprisingly few studies have addressed possible age associated differences in the responsiveness or sensitivity to GH. In normal adults the senescent decline in GH levels is paralleled by a decline in serum IGF-I, suggesting a down-regulation of the GH-IGF-I axis. Administration of GH to elderly healthy adults has generally been associated with predictable, albeit modest, effects on body composition and side effects in terms of fluid retention and modest insulin resistance (34). Whether this reflects an unfavorable balance between effects and side effects in older people or employment of excessive doses of GH is uncertain, but it is evident that older subjects are not resistant to GH. Short-term dose response studies clearly demonstrate that older patients require a lower GH dose to maintain a given serum IGF-I level (35,36), and it has been observed that serum IGF-I increases in individual patients on long-term therapy if the GH dosage remains constant. Moreover, patients with GH deficiency older than 60 yr are highly responsive to even a small dose of GH (37). Interestingly, there is a gender difference in response to GH treatment with men being more responsive in terms of IGF-I generation and fat loss during therapy, most probably due to men having lower estrogen levels that negatively impact the effect of GH on IGF-I generation in the liver (38).

 

The pharmacokinetics and short-term metabolic effects of a near physiological intravenous GH bolus (200 micrograms) were compared in a group of young (30 yr) and older (50 yr) healthy adults (39). The area under the GH curve was significantly lower in older subjects, whereas the elimination half-life was similar in the two groups, suggesting both an increased metabolic clearance rate and apparent distribution volume of GH in older subjects. Both parameters showed a strong positive correlation with fat mass, although multiple regression analysis revealed the age to be an independent positive predictor. The short-term lipolytic response to the GH bolus was higher in young as compared to older subjects. Interestingly, the same study showed that the GH binding protein (GHBP) correlated strongly and positively with abdominal fat mass (40).

 

A prospective long-term study of normal adults with serial concomitant estimations of GH status and adiposity would provide useful information about the cause-effect relationship between GH status and body composition as a function of age. In the meantime, the following hypothesis is proposed (Figure 5): 1.Changes in life-style and genetic predispositions promote accumulation of body fat with aging; 2. The increased fat mass increases FFA availability, inducing insulin resistance and hyperinsulinemia; 3. High insulin levels suppress IGF binding protein (BP)-1 resulting in a relative increase in free IGF-I levels; 4. Systemic elevations of FFA, insulin, and free IGF-I suppresses pituitary GH release, which further increases fat mass; 5. Endogenous GH is cleared more rapidly in subjects with high amount of fat tissue.

 

At present it is not justified to treat the age-associated deterioration in body composition and physical performance with GH also due to concern that the ensuing elevation of IGF-I levels may increase the risk for the development of neoplastic disease.

 

Figure 5. Hypothetical model for the association between low GH levels and increased visceral fat adults.

 

LIFE- LONG GH DEFICIENCY

 

A real-life model for the GH effects in human physiology is provided by the subjects with life-long severe reduction in GH signaling due to GHRH or GHRH receptor mutations, combined deficiency of GH, prolactin, and TSH, or global deletion of GHR. They show short stature, doll facies, high-pitched voices, central obesity, and are fertile (41). Despite central obesity and increased liver fat, they are insulin sensitive, partially protected from cancer, and present a major reduction in pro-aging signaling and perhaps increased longevity (42). The decrease in cancer risk in life-long GH deficiency together with reports on the GH permissive role for neoplastic colon growth (43), preneoplastic mammary lesions (44), and progression of prostate cancer (45) demands, at least, a careful tailoring of GH substitution dosage in the GH deficient patients. However, recent evidence suggests that the GH produced locally by the colon tumor cells, and not pituitary GH, acts in an autocrine and paracrine manner to suppress the tumor suppressor proteins and to increase nuclear β-catenin accumulation and epithelial–mesenchymal transition potentially participating in tumor progression (46,47).

 

GH AND IMMUNE SYSTEM

 

Although the majority of data on the relation between GH and the immune system are from animal studies, it seems that GH may pose immunomodulatory actions. Immune cells express receptors for growth hormone, and respond to GH stimulation (48). The GHR is expressed by several lymphocyte subpopulations. GH stimulates in vitro T and B-cell proliferation and immunoglobulin synthesis, enhances human myeloid progenitor cell maturation, and modulates in vivoTh1/Th2 (8) and humoral immune responses (49). It has been shown that GH can induce de novo T cell production and enhance CD4 recovery in HIV+ patients. Another study with possible clinical relevance showed that sustained GH expression reduced prodromal disease symptoms and eliminated progression to overt diabetes in mouse model of type 1 diabetes, a T-cell–mediated autoimmune disease. GH altered the cytokine environment, triggered anti-inflammatory macrophage (M2) polarization, maintained activity of the suppressor T-cell population, and limited Th17 cell plasticity (49). JAK/STAT signaling, the principal mediator of GHR activation, is well-known to be involved in the modulation of the immune system, so it is tempting to assume that GH may have a role too, but clear data in humans are needed.

 

Growth Hormone Signaling in Humans

 

GROWTH RECEPTOR ACTIVATION

 

GH receptor signaling is a separate and prolific research field by itself (50), so this section will focus on recent data obtained in human models.

 

GHR have been identified in many tissues including fat, lymphocytes, liver, muscle, heart, kidney, brain, and pancreas (51,52). Activation of receptor-associated Janus kinase (JAK) 2 is the critical step in initiating GH signaling. One GH molecule binds to two GHR molecules that exist as preformed homodimers. Following GH binding, the intracellular domains of the GHR dimer undergo rotation, which brings together the two intracellular domains, each of which bind one JAK2 molecule. This in turn induces cross-phosphorylation of tyrosine residues in the kinase domain of each JAK2 molecule followed by tyrosine phosphorylation of the GHR (51,53). Phosphorylated residues on GHR and JAK2 form docking sites for different signaling molecules including signal transducers and activators of transcription (STAT) 1, 3, 5a and 5b. STATs bound to the activated GHR-JAK2 complex are subsequently phosphorylated on a single tyrosine by JAK2 after which they dimerize and translocate to the nucleus, where they bind to DNA and act as gene transcription factors. A STAT5b binding site has been characterized in the IGF-I gene promoter region, which mediates GH-stimulated IGF-I gene activation (54). Attenuation of JAK2-associated GH signaling is mediated by a family of cytokine-inducible suppressors of cytokine signaling (SOCS) (55). SOCS proteins bind to phosphotyrosine residues on the GHR or JAK2 and suppress GH signaling by inhibiting JAK2 activity and competing with STATs for binding on the GHR. As an example, it has been reported that the inhibitory effect of estrogen on hepatic IGF-I production seems to be mediated via up regulation of SOCS-2 (56).

 

GH SIGNALING

 

Data on GHR signaling derive mainly from rodent models and experimental cell lines, although GH-induced activation of the JAK2/STAT5b and the MAPK pathways have been recorded in cultured human fibroblasts from healthy human subjects (57). STAT5b in human subjects is critical for GH-induced IGF-I expression and growth promotion as demonstrated by the identification of mutations in the STAT5b gene of patients presenting with severe GH insensitivity in the presence of normal GHR (58). GHR signaling in human models in vivo has been reported in a study in healthy young male subjects exposed to an intravenous GH bolus vs. saline (59). In muscle and fat biopsies significant tyrosine phosphorylation of STAT5b was recorded after GH exposure at 30-60 minutes. There was no evidence of GH-induced activation of PI 3-kinase, Akt/PKB, or MAPK in either tissue (59).

 

GH AND INSULIN SIGNALING

 

There is animal and in vitro evidence to suggest that insulin and GH share post-receptor signaling pathways (60). Convergence has been reported at the levels of STAT5 and SOCS3 (61) as well as on the major insulin signaling pathway: insulin receptor substrates (IRS) 1 and 2, PI 3-kinase, Akt, and extracellular regulated kinases (ERK) 1 and 2 (62,63). Studies in rodent models suggest that the insulin-antagonistic effects of GH in adipose and skeletal muscle involve suppression of insulin-stimulated PI3-kinase activity (60,64). One study assessed the impact of a GH infusion on insulin sensitivity and the activity of PI3-kinase as well as PKB/AKt in skeletal muscle in a controlled design involving healthy young subjects (65). The infusion of GH induced a sustained increase in FFA levels and subsequently insulin resistance as assessed by the euglycemic clamp technique, as expected, but was not associated with any changes in the insulin-stimulated increase in either IRS-1 associated PI3-kinase or PKB/Akt activity. It was subsequently assessed that insulin had no impact on GH-induced STAT5b activation or SOCS3 mRNA expression (66).

 

INSULIN-LIKE GROWTH FACTOR-I

 

Physiology of IGF-I

 

GH acts both directly through its own receptor and indirectly through the induced production of IGF-I. GH stimulates synthesis of IGF-I in the liver and many other GH target tissues (Figure 6); about 75% of circulating IGF-I is liver-derived.IGF-I is a 70 amino-acid peptide, found in the circulation, 99% bound to transport proteins.

 

Following the initial discovery of IGF-I, it was thought, that GH governs somatic growth only by IGF-I produced by the liver (67). However, in the 1980s this hypothesis was changed by the identification of IGF-I production in numerous tissues (Figure 6).  IGF-I is known as a global and tissue-specific growth factor as well as an endocrine factor. In some tissues IGF-I acts as a potent inhibitor of cellular apoptosis.

 

Figure 6. GH is produced in the pituitary gland. In the periphery, GH acts directly and indirectly through stimulation of IGF-I production. In the circulation, the liver is the most important source of IGF-I (75%) but other tissues (e.g. brain, adipose tissue, kidney, bone, and muscles) may contribute. Under GH stimulation the muscle, adipose tissue, and bone have been shown to secrete IGF-I that has a paracrine/autocrine effect.

 

Interestingly, insulin and IGF-I share many structural and functional similarities implying that they have originated from the same ancestral molecule. Both molecules could have been part of the cycle of food intake and consequent tissue growth. The IGF-I gene is a member of the insulin gene family and the IGF-I receptor is structurally similar to the insulin receptor in its tetrametric structure, with 2 alpha and 2 beta subunits (68). The alpha subunit binds IGF-I, IGF-II, and insulin; however, the subunit has a higher affinity towards IGF-I compared to IGF-II and insulin. Although insulin and IGF-I share many similarities, during evolution, the functionality of the two molecules has become more divergent, where insulin plays a more metabolic role and IGF-I plays a role in cell growth.

 

The IGF-I receptor is expressed in many tissues in the body. However, the receptor number on each cell is strictly regulated by several systemic and tissue factors including circulating GH, iodothyronines, platelet-derived growth factor, and fibroblast growth factor. Following the binding of the IGF-I molecule, the receptor undergoes a conformational change, which activates tyrosine kinase, leading to auto-phosphorylation of tyrosine. The activated receptor phosphorylates “insulin receptor substrate-2” (IRS-2), which in-turn activates the RAS activating protein SOS. This complex activates the mitogen activated protein kinase (MAP kinase) pathway. Thus activation of the MAP kinase pathway becomes vital in the stimulation of cell growth by IGF-I (69,70).

 

IGF-I is bound almost 100% to a family of binding proteins (IGFBP) in the circulation. The IGFBP family comprises six binding proteins (IGFBP 1-6) with a high affinity towards IGF-I and II. Apart from regulating the free plasma IGF fraction, IGFBPs also play an important role in the transport of IGF into different tissues and extravascular space. IGFBP-3 and IGFBP-2 are the most abundant forms seen in plasma and are saturated with IGF-I due to their high affinity. 75% of IGF-I is bound to IGFBP-3. Interestingly, similar to IGF-I, IGFBP-3 production is also regulated by GH. In the plasma, IGFBP-3 is bound to a protein called acid labile subunit (ALS), which stabilizes the “IGFBP3-IGF-I” complex, prolonging its half-life to approximately 16 hours (71). IGFBP-1, on the other hand, is present in lower concentration in plasma than IGFBP-2 and 3. However, due to lower affinity for IGF-I, IGFBP-1 is usually in an unsaturated state and changing plasma concentrations of IGFBP-1 becomes important in determining the unbound fraction of IGF-I. A recently new discovered player in the regulation of IGF-I bioavailability is the pregnancy-associated plasma protein-A2 (PAPP-A2) that cleaves IGFBP3 and 5 and releases IGF-I. Homozygous mutations in PAPP-A2 result in growth failure with elevated total but low free IGF-I (72). Low IGF-I bioavailability impairs growth and glucose metabolism in a mouse model of human PAPP-A2 deficiency and treatment with recombinant human IGF-I in PAPP-A2 deficient patients improves growth and bone mass and ameliorates glucose metabolism (72,73).

 

Effects of IGF-I

 

Studies on hypophysectomized animals overexpressing IGF-I demonstrate the independent anabolic effects of IGF-I (74). IGF-I plays a key role in growth, where it acts not only as a determinant of postnatal growth, but also as an intra-uterine growth promoter. Total inactivation of the IGF-I gene in mice produce a perinatal mortality of 80% with the surviving animal showing significant growth retardation compared to controls (75). Human IGF-I deficiency can be either due to GH deficiency, GHR inactivation, or IGF-I gene mutation. Interestingly, infants with congenital GH deficiency and GHR mutations present with only minor growth retardation, whereas the rare patient with IGF-I deficiency, secondary to a homozygous partial deletion of the IGF-I gene, presents with severe pre and postnatal growth failure, mental retardation, sensorineural deafness, and microcephaly (76-78). The differences in the clinical presentation are most likely due to the fact that some degree of IGF-I production is present in patients with GH deficiency, GHR, and GHRH defects. More detailed studies on transgenic mice have clearly demonstrated this fact with selective deletion of IGF-I gene expression only in the liver, showing low serum IGF-I concentrations with only 6-8% postnatal growth retardation. In contrast, animals with total IGF-I deletion or only peripherally produced IGF-I deletion showed marked growth retardation (79).

 

Both elevated and reduced levels of serum IGF-I are associated with excess mortality in human adults (80). In addition, it is well recognized in many species including worms, flies, rodents, and primates that a reciprocal relationship exists between longevity and activation of the insulin/IGF axis (80). The underlying mechanisms are subject to continued scrutiny and are likely to be complex. In this regard, it is noteworthy that calorie restriction is associated with increased longevity and reduced insulin/IGF activity in many species (81) albeit GH levels are increased by calorie restriction and fasting (82).

 

In the context of GH and IGF-I physiology it can be concluded that 1) during childhood and adolescence the combined actions of GH and IGF-I in the presence of sufficient nutrition promote longitudinal growth and somatic maturation, 2) continued excess IGF-I activity in adulthood increases the risk for cardiovascular and neoplastic diseases and hence reduces longevity, 3) calorie restriction, which suppresses IGF-I activity and stimulates GH secretion, may promote longevity  in human adults (82).

 

METABOLIC EFFECTS OF GROWTH HORMONE

 

Nutritional status dictates GH effects. In the state of feast and sufficient nutrient intake where insulin is increased in the liver and IGF-I production is stimulated, GH promotes protein anabolism. Whereas, in the state with decreased nutrient intake and during sleep and exercise, the direct effect of GH are more predominant and this is mainly characterized by stimulation of lipolysis.

 

Glucose Homeostasis and Lipid Metabolism

 

The involvement of the pituitary gland in the regulation of substrate metabolism was originally detailed in the classic dog studies by Houssay (83). Fasting hypoglycemia and pronounced sensitivity to insulin were distinct features of hypophysectomized animals. These symptoms were readily corrected by the administration of anterior pituitary extracts. It was also noted that pancreatic diabetes was alleviated by hypophysectomy. Finally, excess of anterior pituitary lobe extracts aggravated or induced diabetes in hypophysectomized dogs. Furthermore glycemic control deteriorates following exposure to a single supraphysiological dose of human GH in hypophysectomized adults with type 1 diabetes mellitus (84). Somewhat surprisingly, only modest effects of GH on glucose metabolism were recorded in the first metabolic balance studies involving adult hypopituitary patients (85,86).

 

More recent studies on glucose homeostasis in GH deficient adults have generated results, which at first glance may appear contradictory. Insulin resistance may be more prevalent in untreated GH deficient adults, whereas the impact of GH replacement on this feature seems to depend on the duration and the dose (87).

 

Below, some of the metabolic effects of GH in human subjects, with special reference to the interaction between glucose and lipid metabolism, will be reviewed.

 

STUDIES IN NORMAL ADULTS  

 

More than fifty years ago, it was shown that infusion of high dose GH into the brachial artery of healthy adults reduced forearm glucose uptake in both muscle and adipose tissue, which was paralleled by increased uptake and oxidation of FFA (88). This pattern was opposite to that of insulin, and GH in the same model abrogated the metabolic actions of insulin.

 

Administration of a GH bolus in the post absorptive state stimulates lipolysis following a lag time of 2-3 hours (89). Plasma levels of glucose and insulin, on the other hand, change very little. This is associated with small reductions in muscular glucose uptake and oxidation, which could reflect substrate competition between glucose and fatty acids (i.e., the glucose/fatty acid cycle) (Figure 7). In line with this, sustained exposure to high GH levels induces both hepatic and peripheral (muscular) resistance to the actions of insulin on glucose metabolism together with increased (or inadequately suppressed) lipid oxidation. However, GH excess reduces intrahepatic lipid content suggesting that GH-induced insulin resistance is not associated with hepatic lipid accumulation (90). Apart from enhanced glucose/fatty acid cycling, it has been shown that GH induced insulin resistance is accompanied by reduced muscle glycogen synthase activity (91) and diminished glucose dependent glucose disposal (92). Bak et al. also demonstrated insulin binding and insulin receptor kinase activity from muscle biopsies to be unaffected by GH (91).

 

Undoubtedly, a causal link exists between GH-induced lipolysis and insulin resistance (93). Acute GH exposure in healthy individuals downregulates important suppressors of lipolysis, the G0/G1 switch gene (G0S2) and fat specific protein 27 (FSP27), in addition to regulating the suppressor of the insulin signaling, phosphatase and tensin homolog (PTEN) (94).

 

LESSONS FROM ACROMEGALY

 

Active acromegaly clearly unmasks the diabetogenic properties of GH. In the basal state plasma glucose is elevated despite compensatory hyperinsulinemia. In the basal and insulin-stimulated state (euglycemic glucose clamp) hepatic and peripheral insulin resistance is associated with enhanced lipid oxidation and energy expenditure (95). There is evidence to suggest that this hyper-metabolic state ultimately leads to beta cell exhaustion and overt diabetes mellitus (96), but it is also demonstrated that the abnormalities are completely reversed after successful surgery (95). Conversely, it has been shown that only two weeks of the administration of GH in supraphysiological doses induces comparable acromegaloid, and reversible abnormalities in substrate metabolism and insulin sensitivity (97).

 

Interaction of Glucose and Lipid Metabolism

 

Relatively few studies have scrutinized the exact modes of action of GH on glucose metabolism. There is no evidence of a GH effect on insulin binding to the receptor (91,98), which obviously implies post receptor metabolic effects. The effect of FFA on the partitioning of intracellular glucose fluxes was originally described by Randle et al. (99). According to his hypothesis (the glucose/fatty acid cycle), oxidation of FFA initiates an upstream, chain-reaction-like inhibition of glycolytic enzymes, which ultimately inhibits glucose uptake (Figure 7).

 

Figure 7. The glucose fatty-acid cycle.

 

Randle proposed in 1963 that increased FFA compete with and displace glucose utilization leading to a decreased glucose uptake. The hypothesis stated that an increase in fatty acid oxidation in muscle and fat results in higher acetyl CoA in mitochondria leading to inactivation of two rate-limiting enzymes of glycolysis (i.e., phosphofructokinase (PFK) and pyruvate dehydrogenase (PDH) complex). A subsequent increase in intracellular glucose-6-phosphate (glucose 6-P) results in high intracellular glucose concentrations and decreased glucose uptake by muscle and fat.

 

However, in contrast to the proposed hypothesis by Randle, studies using MR spectroscopy have shown reductions in intramyocellular glucose 6-P and glucose concentrations and have led to an alternative hypothesis. The new hypothesis proposes that a transient increase of intracellular diacylglycerol (DAG) activates theta isoform of protein kinase C (PKCθ) that causes increased serine phosphorylation of IRS-1/2 and consecutively decrease PI3K activation and glucose-transport activity leading to decrease intracellular glucose concentrations

 

When considering the pronounced lipolytic effects of GH the Randle hypothesis remains an appealing model to explain the insulin-antagonistic effects of GH. In support of this, experiments have shown that co-administration of anti-lipolytic agents and GH reverses GH-induced insulin resistance. Similar conclusions were drawn from a recent study in GH deficient adults, which showed that insulin sensitivity was restored when acipimox (a nicotinic acid derivative) was co-administered with GH (100). We have also shown that GH-induced insulin resistance is associated with suppressed pyruvate dehydrogenase activity in skeletal muscle (101). It has, however, also been reported that GH-induced insulin resistance precedes the increase in circulating levels of fatty acids and forearm uptake of lipid intermediates (102). This early effect of GH on muscular glucose uptake could reflect intramyocytic FFA release and oxidation and thus be compatible with the Randle hypothesis. According to the Randle hypothesis the fatty acid-induced insulin resistance will result in elevated intracellular levels of both glucose and glucose-6-phosphate (Figure 7). By contrast, muscle biopsies from GH deficient adults after GH treatment have revealed increased glucose but low-normal glucose-6-phosphate levels (103). Moreover, NMR spectroscopy studies in healthy adults indicate that FFA infusion results in a drop in the levels of both glucose and glucose-6-phosphate (104). The latter study, which did not involve GH administration, reported that FFA suppressed the activity of PI-3 kinase, an enzyme stimulated by insulin, which is considered essential for glucose transportation into skeletal muscle via translocation of glucose transporter activity (GLUT 4). A more recent study showed that GH infusion does not impact insulin-stimulated PI-3 kinase activity (65).

 

IMPLICATIONS FOR GH REPLACEMENT  

 

Regardless of the exact mechanisms, the insulin antagonistic effects may cause concern when replacing adult GH deficient patients with GH, since some of these patients are insulin resistant in the untreated state. There is evidence to suggest that the direct metabolic effects on GH may be balanced by long-term beneficial effects on body composition and physical fitness, but some studies report impaired insulin sensitivity in spite of favorable changes in body composition. There is little doubt that these effects of GH are dose-dependent and may be minimized or avoided if an appropriately low replacement dose is used. Still, the pharmacokinetics of subcutaneous (s.c.) GH administration is unable to mimic the endogenous GH pattern with suppressed levels after meals and elevations only during post absorptive periods, such as during the night. This may be considered the natural domain of GH action, which coincides with minimal beta-cell challenge. This reciprocal association between insulin and GH and its potential implications for normal substrate metabolism was initially described by Rabinowitz & Zierler (105). The problem arises when GH levels are elevated during repeated prandial periods. The classic example is active acromegaly, but prolonged high dose s.c. GH administration may cause similar effects. Administration of GH in the evening probably remains the best compromise between effects and side effects (106), but it is far from physiological.

Long-acting GH analogues have been developed to improve adherence and compliance. The clinical experience is limited now but seem not to impact adversely the glucose metabolism compared with daily GH (107). However, long-term surveillance data are required to consolidate its safety profile (108).

 

Effects of GH on Muscle Mass and Function

 

The anabolic nature of GH is clearly evident in patients with acromegaly and vice versa in patients with GH deficiency. A large number of in vitro and animal studies throughout several decades have documented stimulating effects of GH on skeletal muscle growth. The methods employed to document in vivo effects of GH on muscle mass in humans have been exhaustive including whole body retention of nitrogen and potassium, total and regional muscle protein metabolism using labeled amino acids, estimation of lean body mass by total body potassium or dual x-ray absorptiometry, and direct calculation of muscle area or volume by computerized tomography (CT) and magnetic resonance imaging.

 

EFFECT OF GH ON SKELETAL MUSCLE METABOLISM IN VITRO AND IN VIVO

 

The clinical picture of acromegaly and gigantism includes increased lean body mass of which skeletal muscle mass accounts for approximately 50%. Moreover, retention of nitrogen was one of the earliest observed and most reproducible effects of GH administration in humans (1). Thoroughly conducted studies with GH administration in GH deficient children using a variety of classic anthropometric techniques strongly suggested that skeletal muscle mass increased significantly during treatment (109,110). Indirect evidence of an increase in muscle cell number following GH treatment was also presented (110).

 

These early clinical studies were paralleled by experimental studies in rodent models. GH administration in hypophysectomized rats increased not only muscle mass, but also muscle cell number (i.e., muscle DNA content) (110). Interestingly, the same series of experiments revealed that work-induced muscle hypertrophy could occur in the absence of GH. The ability of GH to stimulate RNA synthesis and amino acid incorporation into protein of isolated rat diaphragm suggested direct mechanisms of actions, whereas direct effects of GH on protein synthesis could not be induced in liver cell cultures (111). Another important observation of that period was made by Goldberg, who studied protein turnover in skeletal muscle of hypophysectomized rats with 3H-leucine tracer techniques. In these studies it was convincingly demonstrated that GH directly increased the synthesis of both sarcoplasmic and myofibrillar protein without affecting proteolysis (112).

 

The most substantial recent contributions within the field derive from human in vivo studies of the effects of systemic and local GH and IGF-I administration on total and regional protein metabolism by means of amino acid isotope dilution techniques. Systemic GH administration for 7 days in normal adults increased whole body protein synthesis without affecting proteolysis (113), and similar data were subsequently obtained in GH deficient adults (114). Systemically infused GH for 8 hours in normal adults lead to an acute stimulation of forearm (muscle) protein synthesis without any effects on whole body protein synthesis (115). By contrast in a design that also included co-administration of somatostatin to suppress insulin, an acute stimulatory effect of GH on whole body protein synthesis was observed, but no stimulatory effect on leg protein synthesis (116), Finally, infusion of GH into the brachial artery was accompanied by a local increase in forearm muscle protein synthesis (117).

 

Based on these studies it seems that the nitrogen retaining properties of GH predominantly involve stimulation of protein synthesis without affecting (lowering) proteolysis. Theoretically, the protein anabolic effects of GH could be either direct or mediated through IGF-I, insulin, or lipid intermediates. GHR are present in skeletal muscle (52), which combined with Fryburg’s intra-arterial GH studies, makes a direct GH effect conceivable. An alternative interpretation could be that GH stimulates local muscle IGF-I release, which subsequently acts in an autocrine/paracrine manner. The effects of systemic IGF-I administration on whole body protein metabolism seem to depend on ambient amino acid levels in the sense that IGF-I administered alone suppresses proteolysis (118) whereas IGF-I in combination with an amino acid infusion increase protein synthesis (119). Moreover, intra-arterial IGF-I in combination with systemic amino acid infusion increased protein synthesis (120). It is therefore likely that the muscle anabolic effects of GH, at least to some extent, are mediated by IGF-I. By contrast, it is repeatedly shown that insulin predominantly acts through suppression of proteolysis and this effect(s) appears to be blunted by co-administration of GH (121). The degree to which mobilization of lipids contributes to the muscle anabolic actions of GH has so far not been specifically investigated.

 

In conclusion several experimental lines of evidence strongly suggest that GH stimulates muscle protein synthesis. This effect is presumably in part mediated through binding of GH to GHR in skeletal muscle. This does not rule out a significant role of IGF-I being produced either systematically or locally.

 

An interesting discovery has been that infusion of GH and IGF-I into the brachial artery increases forearm blood flow several fold (117,122). This effect appears to be mediated through stimulation of endothelial nitric oxide release leading to local vasodilatation (123,124). Thus, it appears that an IGF-I mediated increase in muscle nitric oxide release accounts for some of the effects of GH on skeletal muscle protein synthesis. These intriguing observations may have many other implications. It is, for instance, tempting to speculate that this increase in skeletal muscle blood flow contributes to the GH induced increase in resting energy expenditure, since skeletal muscle metabolism is a major determinant of resting energy expenditure (24). Moreover, it is plausible that the reduction in total peripheral resistance seen after GH administration in adult growth hormone deficiency is mediated by nitric oxide (124).

 

EFFECTS OF GH ADMINISTRATION ON MUSCLE MASS AND FUNCTION IN ADULTS WITHOUT GH DEFICIENCY  

 

As previously mentioned, the ability of acute and more prolonged GH administration to retain nitrogen in healthy adults has been known for decades and more recent studies have documented a stimulatory effect on whole body and forearm protein synthesis.

 

Rudman et al. was the first to suggest that the senescent changes in body composition were causally linked to the concomitant decline in circulation GH and IGF-I levels (24). This concept has been recently reviewed (125) and a number of studies with GH and other anabolic agents for treating the sarcopenia of ageing are currently in progress.

 

Placebo-controlled GH administration in young healthy adults (21-34 yr) undergoing a resistance exercise program for 12 weeks showed a GH induced increase in lean body mass (LBM), whole body protein balance, and whole body protein synthesis, whereas quadriceps muscle protein synthesis rate and muscle strength increased to the same degree in both groups during training (126). In a similar study in older men (67 yr) GH also increased LBM and whole body protein synthesis, without significantly amplifying the effects of exercise on muscle protein synthesis or muscle strength (127). An increase in LBM but unaltered muscle strength following 10 weeks of GH administration plus resistance exercise training was also recorded (128). A more recent study of 52 older men (70-85 yr) treated with either GH or placebo for 6 months, without concomitant exercise, observed a significant increase (4.4 %) in LBM with GH, but no significant effects on muscle strength (129). A meta-analysis of studies administering GH to healthy adult subjects demonstrate that it increases lean body mass and reduces fat mass without improving muscle strength or aerobic exercise capacity (130).

 

Numerous studies have evaluated the effects of GH administration in chronic and acute catabolic illness. A comprehensive survey of the prolific literature within this field is beyond the scope of this review, but it is noteworthy, that HIV-associated body wasting is a licensed indication for GH treatment in the USA. In this patient category GH treatment for 12 weeks has been associated with significant increments in LBM and physical fitness (131,132).

 

CONCLUSIONS

 

GH/IGF-I axis is specifically regulated and is involved in a multitude of processes during all aspects of life from intrauterine growth, to childhood and puberty, adulthood, and lastly elderly periods. GH actions directly or via its principal metabolite, IGF-I have a wide range of physiological roles being a metabolic active hormone in adulthood. Nutritional status of an organism dictates the effects of GH, either an impairment of insulin action (fasted state) or promoting protein anabolism (feed state). As our knowledge of the GH normal physiology increases, our ability to understand and specifically target the GH/IGF-I pathway for a diverse range of therapeutic purposes should also increase.

 

REFERENCES

 

  1. Raben MS. Growth hormone. 1. Physiologic aspects. The New England journal of medicine. 1962;266:31-35.
  2. Petronella N, Drouin G. Gene conversions in the growth hormone gene family of primates: stronger homogenizing effects in the Hominidae lineage. Genomics. 2011;98(3):173-181.
  3. Baumann GP. Growth hormone doping in sports: a critical review of use and detection strategies. Endocrine reviews. 2012;33(2):155-186.
  4. Holt RIG, Ho KKY. The Use and Abuse of Growth Hormone in Sports. Endocrine reviews. 2019;40(4):1163-1185.
  5. Cheung LYM, George AS, McGee SR, et al. Single-Cell RNA Sequencing Reveals Novel Markers of Male Pituitary Stem Cells and Hormone-Producing Cell Types. Endocrinology. 2018;159(12):3910-3924.
  6. Willems C, Fu Q, Roose H, et al. Regeneration in the pituitary after cell-ablation injury: time-related aspects and molecular analysis. Endocrinology. 2015:en20151741.
  7. Ribeiro-Oliveira A, Barkan AL. Growth Hormone Pulsatility and its Impact on Growth and Metabolism in Humans. in K. Ho (ed.), Growth Hormone Related Diseases and Therapy: A Molecular and Physiological Perspective for the Clinician, Contemporary Endocrinology. 2011:33-56.
  8. Ho KY, Evans WS, Blizzard RM, et al. Effects of sex and age on the 24-hour profile of growth hormone secretion in man: importance of endogenous estradiol concentrations. The Journal of clinical endocrinology and metabolism. 1987;64(1):51-58.
  9. Leung KC, Johannsson G, Leong GM, Ho KK. Estrogen regulation of growth hormone action. Endocrine reviews. 2004;25(5):693-721.
  10. Kargi AY, Merriam GR. Diagnosis and treatment of growth hormone deficiency in adults. Nature reviews. Endocrinology. 2013;9(6):335-345.
  11. Iacovazzo D, Hernandez-Ramirez LC, Korbonits M. Sporadic pituitary adenomas: the role of germline mutations and recommendations for genetic screening. Expert review of endocrinology & metabolism. 2017;12(2):143-153.
  12. Birzniece V, Ho KKY. Sex steroids and the GH axis: Implications for the management of hypopituitarism. Best practice & research. Clinical endocrinology & metabolism. 2017;31(1):59-69.
  13. Veldhuis JD, Evans WS, Bowers CY, Anderson S. Interactive regulation of postmenopausal growth hormone insulin-like growth factor axis by estrogen and growth hormone-releasing peptide-2. Endocrine. 2001;14(1):45-62.
  14. Bray MJ, Vick TM, Shah N, et al. Short-term estradiol replacement in postmenopausal women selectively mutes somatostatin's dose-dependent inhibition of fasting growth hormone secretion. The Journal of clinical endocrinology and metabolism. 2001;86(7):3143-3149.
  15. Anderson SM, Shah N, Evans WS, Patrie JT, Bowers CY, Veldhuis JD. Short-term estradiol supplementation augments growth hormone (GH) secretory responsiveness to dose-varying GH-releasing peptide infusions in healthy postmenopausal women. The Journal of clinical endocrinology and metabolism. 2001;86(2):551-560.
  16. Murray PG, Higham CE, Clayton PE. 60 YEARS OF NEUROENDOCRINOLOGY: The hypothalamo-GH axis: the past 60 years. The Journal of endocrinology. 2015;226(2):T123-140.
  17. Bonnefont X, Lacampagne A, Sanchez-Hormigo A, et al. Revealing the large-scale network organization of growth hormone-secreting cells. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(46):16880-16885.
  18. Le Tissier PR, Carmignac DF, Lilley S, et al. Hypothalamic growth hormone-releasing hormone (GHRH) deficiency: targeted ablation of GHRH neurons in mice using a viral ion channel transgene. Molecular endocrinology. 2005;19(5):1251-1262.
  19. Wajnrajch MP, Gertner JM, Harbison MD, Chua SC, Jr., Leibel RL. Nonsense mutation in the human growth hormone-releasing hormone receptor causes growth failure analogous to the little (lit) mouse. Nature genetics. 1996;12(1):88-90.
  20. Alatzoglou KS, Dattani MT. Genetic causes and treatment of isolated growth hormone deficiency-an update. Nature reviews. Endocrinology. 2010;6(10):562-576.
  21. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocrine reviews. 1998;19(6):717-797.
  22. Ren SG, Taylor J, Dong J, Yu R, Culler MD, Melmed S. Functional association of somatostatin receptor subtypes 2 and 5 in inhibiting human growth hormone secretion. The Journal of clinical endocrinology and metabolism. 2003;88(9):4239-4245.
  23. Copinschi G, Wegienka LC, Hane S, Forsham PH. Effect of arginine on serum levels of insulin and growth hormone in obese subjects. Metabolism: clinical and experimental. 1967;16(6):485-491.
  24. Rudman D, Kutner MH, Rogers CM, Lubin MF, Fleming GA, Bain RP. Impaired growth hormone secretion in the adult population: relation to age and adiposity. The Journal of clinical investigation. 1981;67(5):1361-1369.
  25. Rudman D. Growth hormone, body composition, and aging. Journal of the American Geriatrics Society. 1985;33(11):800-807.
  26. Jorgensen JO, Vahl N, Hansen TB, Thuesen L, Hagen C, Christiansen JS. Growth hormone versus placebo treatment for one year in growth hormone deficient adults: increase in exercise capacity and normalization of body composition. Clinical endocrinology. 1996;45(6):681-688.
  27. Williams T, Berelowitz M, Joffe SN, et al. Impaired growth hormone responses to growth hormone-releasing factor in obesity. A pituitary defect reversed with weight reduction. The New England journal of medicine. 1984;311(22):1403-1407.
  28. Vahl N, Jorgensen JO, Jurik AG, Christiansen JS. Abdominal adiposity and physical fitness are major determinants of the age associated decline in stimulated GH secretion in healthy adults. The Journal of clinical endocrinology and metabolism. 1996;81(6):2209-2215.
  29. Vahl N, Jorgensen JO, Skjaerbaek C, Veldhuis JD, Orskov H, Christiansen JS. Abdominal adiposity rather than age and sex predicts mass and regularity of GH secretion in healthy adults. The American journal of physiology. 1997;272(6 Pt 1):E1108-1116.
  30. Papadakis MA, Grady D, Tierney MJ, Black D, Wells L, Grunfeld C. Insulin-like growth factor 1 and functional status in healthy older men. Journal of the American Geriatrics Society. 1995;43(12):1350-1355.
  31. Goodman-Gruen D, Barrett-Connor E. Epidemiology of insulin-like growth factor-I in elderly men and women. The Rancho Bernardo Study. American journal of epidemiology. 1997;145(11):970-976.
  32. Kiel DP, Puhl J, Rosen CJ, Berg K, Murphy JB, MacLean DB. Lack of an association between insulin-like growth factor-I and body composition, muscle strength, physical performance or self-reported mobility among older persons with functional limitations. Journal of the American Geriatrics Society. 1998;46(7):822-828.
  33. Juul A, Bang P, Hertel NT, et al. Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, sex, stage of puberty, testicular size, and body mass index. The Journal of clinical endocrinology and metabolism. 1994;78(3):744-752.
  34. Rudman D, Feller AG, Nagraj HS, et al. Effects of human growth hormone in men over 60 years old. The New England journal of medicine. 1990;323(1):1-6.
  35. Jorgensen JO, Flyvbjerg A, Lauritzen T, Alberti KG, Orskov H, Christiansen JS. Dose-response studies with biosynthetic human growth hormone (GH) in GH-deficient patients. The Journal of clinical endocrinology and metabolism. 1988;67(1):36-40.
  36. Moller J, Jorgensen JO, Lauersen T, et al. Growth hormone dose regimens in adult GH deficiency: effects on biochemical growth markers and metabolic parameters. Clinical endocrinology. 1993;39(4):403-408.
  37. Toogood AA, Shalet SM. Growth hormone replacement therapy in the elderly with hypothalamic-pituitary disease: a dose-finding study. The Journal of clinical endocrinology and metabolism. 1999;84(1):131-136.
  38. Burman P, Johansson AG, Siegbahn A, Vessby B, Karlsson FA. Growth hormone (GH)-deficient men are more responsive to GH replacement therapy than women. The Journal of clinical endocrinology and metabolism. 1997;82(2):550-555.
  39. Vahl N, Moller N, Lauritzen T, Christiansen JS, Jorgensen JO. Metabolic effects and pharmacokinetics of a growth hormone pulse in healthy adults: relation to age, sex, and body composition. The Journal of clinical endocrinology and metabolism. 1997;82(11):3612-3618.
  40. Fisker S, Vahl N, Jorgensen JO, Christiansen JS, Orskov H. Abdominal fat determines growth hormone-binding protein levels in healthy nonobese adults. The Journal of clinical endocrinology and metabolism. 1997;82(1):123-128.
  41. Aguiar-Oliveira MH, Bartke A. Growth Hormone Deficiency: Health and Longevity. Endocrine reviews. 2019;40(2):575-601.
  42. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Science translational medicine. 2011;3(70):70ra13.
  43. Chesnokova V, Zonis S, Zhou C, et al. Growth hormone is permissive for neoplastic colon growth. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(23):E3250-3259.
  44. Kleinberg DL, Wood TL, Furth PA, Lee AV. Growth hormone and insulin-like growth factor-I in the transition from normal mammary development to preneoplastic mammary lesions. Endocrine reviews. 2009;30(1):51-74.
  45. Slater MD, Murphy CR. Co-expression of interleukin-6 and human growth hormone in apparently normal prostate biopsies that ultimately progress to prostate cancer using low pH, high temperature antigen retrieval. Journal of molecular histology. 2006;37(1-2):37-41.
  46. Khan J, Pernicova I, Nisar K, Korbonits M. Mechanisms of ageing: growth hormone, dietary restriction, and metformin. The lancet. Diabetes & endocrinology. 2023;11(4):261-281.
  47. Chesnokova V, Zonis S, Apostolou A, et al. Local non-pituitary growth hormone is induced with aging and facilitates epithelial damage. Cell reports. 2021;37(11):110068.
  48. Bodart G, Farhat K, Charlet-Renard C, Salvatori R, Geenen V, Martens H. The Somatotrope Growth Hormone-Releasing Hormone/Growth Hormone/Insulin-Like Growth Factor-1 Axis in Immunoregulation and Immunosenescence. Frontiers of hormone research. 2017;48:147-159.
  49. Villares R, Kakabadse D, Juarranz Y, Gomariz RP, Martinez AC, Mellado M. Growth hormone prevents the development of autoimmune diabetes. Proceedings of the National Academy of Sciences of the United States of America. 2013.
  50. Lanning NJ, Carter-Su C. Recent advances in growth hormone signaling. Rev Endocr Metab Disord. 2006;7(4):225-235.
  51. Dehkhoda F, Lee CMM, Medina J, Brooks AJ. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Frontiers in endocrinology. 2018;9:35.
  52. Kelly PA, Djiane J, Postel-Vinay MC, Edery M. The prolactin/growth hormone receptor family. Endocrine reviews. 1991;12(3):235-251.
  53. Brooks AJ, Dai W, O'Mara ML, et al. Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science (New York, N.Y.). 2014;344(6185):1249783.
  54. Woelfle J, Chia DJ, Rotwein P. Mechanisms of growth hormone (GH) action. Identification of conserved Stat5 binding sites that mediate GH-induced insulin-like growth factor-I gene activation. The Journal of biological chemistry. 2003;278(51):51261-51266.
  55. Wormald S, Hilton DJ. Inhibitors of cytokine signal transduction. The Journal of biological chemistry. 2004;279(2):821-824.
  56. Leung KC, Doyle N, Ballesteros M, et al. Estrogen inhibits GH signaling by suppressing GH-induced JAK2 phosphorylation, an effect mediated by SOCS-2. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(3):1016-1021.
  57. Silva CM, Kloth MT, Whatmore AJ, et al. GH and epidermal growth factor signaling in normal and Laron syndrome fibroblasts. Endocrinology. 2002;143(7):2610-2617.
  58. Hwa V, Little B, Adiyaman P, et al. Severe growth hormone insensitivity resulting from total absence of signal transducer and activator of transcription 5b. The Journal of clinical endocrinology and metabolism. 2005;90(7):4260-4266.
  59. Jorgensen JO, Jessen N, Pedersen SB, et al. GH receptor signaling in skeletal muscle and adipose tissue in human subjects following exposure to an intravenous GH bolus. American journal of physiology. Endocrinology and metabolism. 2006;291(5):E899-905.
  60. Dominici FP, Argentino DP, Munoz MC, Miquet JG, Sotelo AI, Turyn D. Influence of the crosstalk between growth hormone and insulin signalling on the modulation of insulin sensitivity. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2005;15(5):324-336.
  61. Emanuelli B, Peraldi P, Filloux C, Sawka-Verhelle D, Hilton D, Van Obberghen E. SOCS-3 is an insulin-induced negative regulator of insulin signaling. The Journal of biological chemistry. 2000;275(21):15985-15991.
  62. Ridderstrale M, Degerman E, Tornqvist H. Growth hormone stimulates the tyrosine phosphorylation of the insulin receptor substrate-1 and its association with phosphatidylinositol 3-kinase in primary adipocytes. The Journal of biological chemistry. 1995;270(8):3471-3474.
  63. Thirone AC, Carvalho CR, Saad MJ. Growth hormone stimulates the tyrosine kinase activity of JAK2 and induces tyrosine phosphorylation of insulin receptor substrates and Shc in rat tissues. Endocrinology. 1999;140(1):55-62.
  64. del Rincon JP, Iida K, Gaylinn BD, et al. Growth hormone regulation of p85alpha expression and phosphoinositide 3-kinase activity in adipose tissue: mechanism for growth hormone-mediated insulin resistance. Diabetes. 2007;56(6):1638-1646.
  65. Jessen N, Djurhuus CB, Jorgensen JO, et al. Evidence against a role for insulin-signaling proteins PI 3-kinase and Akt in insulin resistance in human skeletal muscle induced by short-term GH infusion. American journal of physiology. Endocrinology and metabolism. 2005;288(1):E194-199.
  66. Nielsen C, Gormsen LC, Jessen N, et al. Growth hormone signaling in vivo in human muscle and adipose tissue: impact of insulin, substrate background, and growth hormone receptor blockade. The Journal of clinical endocrinology and metabolism. 2008;93(7):2842-2850.
  67. Salmon WD, Jr., Daughaday WH. A hormonally controlled serum factor which stimulates sulfate incorporation by cartilage in vitro. The Journal of laboratory and clinical medicine. 1957;49(6):825-836.
  68. Werner H, Weinstein D, Bentov I. Similarities and differences between insulin and IGF-I: structures, receptors, and signalling pathways. Archives of physiology and biochemistry. 2008;114(1):17-22.
  69. Denley A, Cosgrove LJ, Booker GW, Wallace JC, Forbes BE. Molecular interactions of the IGF system. Cytokine & growth factor reviews. 2005;16(4-5):421-439.
  70. Kim JJ, Accili D. Signalling through IGF-I and insulin receptors: where is the specificity? Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2002;12(2):84-90.
  71. Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocrine reviews. 2002;23(6):824-854.
  72. Cabrera-Salcedo C, Mizuno T, Tyzinski L, et al. Pharmacokinetics of IGF-1 in PAPP-A2-Deficient Patients, Growth Response, and Effects on Glucose and Bone Density. The Journal of clinical endocrinology and metabolism. 2017;102(12):4568-4577.
  73. Fujimoto M, Andrew M, Liao L, et al. Low IGF-I Bioavailability Impairs Growth and Glucose Metabolism in a Mouse Model of Human PAPPA2 p.Ala1033Val Mutation. Endocrinology. 2019;160(6):1363-1376.
  74. Behringer RR, Lewin TM, Quaife CJ, Palmiter RD, Brinster RL, D'Ercole AJ. Expression of insulin-like growth factor I stimulates normal somatic growth in growth hormone-deficient transgenic mice. Endocrinology. 1990;127(3):1033-1040.
  75. Powell-Braxton L, Hollingshead P, Giltinan D, Pitts-Meek S, Stewart T. Inactivation of the IGF-I gene in mice results in perinatal lethality. Annals of the New York Academy of Sciences. 1993;692:300-301.
  76. Gluckman PD, Gunn AJ, Wray A, et al. Congenital idiopathic growth hormone deficiency associated with prenatal and early postnatal growth failure. The International Board of the Kabi Pharmacia International Growth Study. The Journal of pediatrics. 1992;121(6):920-923.
  77. Savage MO, Blum WF, Ranke MB, et al. Clinical features and endocrine status in patients with growth hormone insensitivity (Laron syndrome). The Journal of clinical endocrinology and metabolism. 1993;77(6):1465-1471.
  78. Woods KA, Camacho-Hubner C, Savage MO, Clark AJ. Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. The New England journal of medicine. 1996;335(18):1363-1367.
  79. Lupu F, Terwilliger JD, Lee K, Segre GV, Efstratiadis A. Roles of growth hormone and insulin-like growth factor 1 in mouse postnatal growth. Developmental biology. 2001;229(1):141-162.
  80. Bartke A, Sun LY, Longo V. Somatotropic signaling: trade-offs between growth, reproductive development, and longevity. Physiological reviews. 2013;93(2):571-598.
  81. Fontana L, Partridge L, Longo VD. Extending healthy life span--from yeast to humans. Science (New York, N.Y.). 2010;328(5976):321-326.
  82. Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocrine reviews. 2009;30(2):152-177.
  83. BA H. The hypophysis and metabolism. The New England journal of medicine. 1936;214:961-985.
  84. Luft R, Ikkos D, Gemzell CA, Olivecrona H. Effect of human growth hormone in hypophysectomised diabetic subjects. Lancet. 1958;1(7023):721-722.
  85. Raben MS, Hollenberg CH. Effect of growth hormone on plasma fatty acids. The Journal of clinical investigation. 1959;38(3):484-488.
  86. Henneman DH, Henneman PH. Effects of human growth hormone on levels of blood urinary carbohydrate and fat metabolites in man. The Journal of clinical investigation. 1960;39:1239-1245.
  87. Hew FL, Koschmann M, Christopher M, et al. Insulin resistance in growth hormone-deficient adults: defects in glucose utilization and glycogen synthase activity. The Journal of clinical endocrinology and metabolism. 1996;81(2):555-564.
  88. Rabinowitz D, Klassen GA, Zierler KL. Effect of human growth hormone on muscle and adipose tissue metabolism in the forearm of man. The Journal of clinical investigation. 1965;44:51-61.
  89. Moller N, Jorgensen JO, Schmitz O, et al. Effects of a growth hormone pulse on total and forearm substrate fluxes in humans. The American journal of physiology. 1990;258(1 Pt 1):E86-91.
  90. Arlien-Søborg MC, Madsen MA, Dal J, et al. Ectopic lipid deposition and insulin resistance in patients with GH disorders before and after treatment. European journal of endocrinology / European Federation of Endocrine Societies. 2023;188(1).
  91. Bak JF, Moller N, Schmitz O. Effects of growth hormone on fuel utilization and muscle glycogen synthase activity in normal humans. The American journal of physiology. 1991;260(5 Pt 1):E736-742.
  92. Orskov L, Schmitz O, Jorgensen JO, et al. Influence of growth hormone on glucose-induced glucose uptake in normal men as assessed by the hyperglycemic clamp technique. The Journal of clinical endocrinology and metabolism. 1989;68(2):276-282.
  93. Hjelholt AJ, Charidemou E, Griffin JL, et al. Insulin resistance induced by growth hormone is linked to lipolysis and associated with suppressed pyruvate dehydrogenase activity in skeletal muscle: a 2 × 2 factorial, randomised, crossover study in human individuals. Diabetologia. 2020;63(12):2641-2653.
  94. Hjelholt AJ, Lee KY, Arlien-Søborg MC, et al. Temporal patterns of lipolytic regulators in adipose tissue after acute growth hormone exposure in human subjects: A randomized controlled crossover trial. Molecular metabolism. 2019;29:65-75.
  95. Moller N, Schmitz O, Joorgensen JO, et al. Basal- and insulin-stimulated substrate metabolism in patients with active acromegaly before and after adenomectomy. The Journal of clinical endocrinology and metabolism. 1992;74(5):1012-1019.
  96. Sonksen PH, Greenwood FC, Ellis JP, Lowy C, Rutherford A, Nabarro JD. Changes of carbohydrate tolerance in acromegaly with progress of the disease and in response to treatment. The Journal of clinical endocrinology and metabolism. 1967;27(10):1418-1430.
  97. Moller N, Moller J, Jorgensen JO, et al. Impact of 2 weeks high dose growth hormone treatment on basal and insulin stimulated substrate metabolism in humans. Clinical endocrinology. 1993;39(5):577-581.
  98. Rosenfeld RG, Wilson DM, Dollar LA, Bennett A, Hintz RL. Both human pituitary growth hormone and recombinant DNA-derived human growth hormone cause insulin resistance at a postreceptor site. The Journal of clinical endocrinology and metabolism. 1982;54(5):1033-1038.
  99. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1(7285):785-789.
  100. Nielsen S, Moller N, Christiansen JS, Jorgensen JO. Pharmacological antilipolysis restores insulin sensitivity during growth hormone exposure. Diabetes. 2001;50(10):2301-2308.
  101. Nellemann B, Vendelbo MH, Nielsen TS, et al. Growth hormone-induced insulin resistance in human subjects involves reduced pyruvate dehydrogenase activity. Acta physiologica (Oxford, England). 2014;210(2):392-402.
  102. Jensen RB, Thankamony A, Day F, et al. Genetic markers of insulin sensitivity and insulin secretion are associated with spontaneous postnatal growth and response to growth hormone treatment in short SGA children: the North European SGA Study (NESGAS). The Journal of clinical endocrinology and metabolism. 2015;100(3):E503-507.
  103. Christopher M, Hew FL, Oakley M, Rantzau C, Alford F. Defects of insulin action and skeletal muscle glucose metabolism in growth hormone-deficient adults persist after 24 months of recombinant human growth hormone therapy. The Journal of clinical endocrinology and metabolism. 1998;83(5):1668-1681.
  104. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of clinical investigation. 2000;106(2):171-176.
  105. Rabinowitz D, Zierler KL. A METABOLIC REGULATING DEVICE BASED ON THE ACTIONS OF HUMAN GROWTH HORMONE AND OF INSULIN, SINGLY AND TOGETHER, ON THE HUMAN FOREARM. Nature. 1963;199:913-915.
  106. Jorgensen JO. Human growth hormone replacement therapy: pharmacological and clinical aspects. Endocrine reviews. 1991;12(3):189-207.
  107. Takahashi Y, Biller BMK, Fukuoka H, et al. Weekly somapacitan had no adverse effects on glucose metabolism in adults with growth hormone deficiency. Pituitary. 2023;26(1):57-72.
  108. Ho KK, O'Sullivan AJ, Burt MG. The physiology of growth hormone (GH) in adults: translational journey to GH replacement therapy. The Journal of endocrinology. 2023;257(2).
  109. Tanner JM, Hughes PC, Whitehouse RH. Comparative rapidity of response of height, limb muscle and limb fat to treatment with human growth hormone in patients with and without growth hormone deficiency. Acta endocrinologica. 1977;84(4):681-696.
  110. DB C. Effect of growth hormone on cell and somatic growth. . In: Handbook of physiology (Eds. Knobil and Sawyer)Washington DC. 1974:159-186.
  111. Korner A. Growth hormone control of biosynthesis of protein and ribonucleic acid. Recent progress in hormone research. 1965;21:205-240.
  112. Goldberg AL. Protein turnover in skeletal muscle. I. Protein catabolism during work-induced hypertrophy and growth induced with growth hormone. The Journal of biological chemistry. 1969;244(12):3217-3222.
  113. Horber FF, Haymond MW. Human growth hormone prevents the protein catabolic side effects of prednisone in humans. The Journal of clinical investigation. 1990;86(1):265-272.
  114. Russell-Jones DL, Weissberger AJ, Bowes SB, et al. The effects of growth hormone on protein metabolism in adult growth hormone deficient patients. Clinical endocrinology. 1993;38(4):427-431.
  115. Fryburg DA, Barrett EJ. Growth hormone acutely stimulates skeletal muscle but not whole-body protein synthesis in humans. Metabolism: clinical and experimental. 1993;42(9):1223-1227.
  116. Copeland KC, Nair KS. Acute growth hormone effects on amino acid and lipid metabolism. The Journal of clinical endocrinology and metabolism. 1994;78(5):1040-1047.
  117. Fryburg DA, Gelfand RA, Barrett EJ. Growth hormone acutely stimulates forearm muscle protein synthesis in normal humans. The American journal of physiology. 1991;260(3 Pt 1):E499-504.
  118. Turkalj I, Keller U, Ninnis R, Vosmeer S, Stauffacher W. Effect of increasing doses of recombinant human insulin-like growth factor-I on glucose, lipid, and leucine metabolism in man. The Journal of clinical endocrinology and metabolism. 1992;75(5):1186-1191.
  119. Russell-Jones DL, Umpleby AM, Hennessy TR, et al. Use of a leucine clamp to demonstrate that IGF-I actively stimulates protein synthesis in normal humans. The American journal of physiology. 1994;267(4 Pt 1):E591-598.
  120. Fryburg DA, Jahn LA, Hill SA, Oliveras DM, Barrett EJ. Insulin and insulin-like growth factor-I enhance human skeletal muscle protein anabolism during hyperaminoacidemia by different mechanisms. The Journal of clinical investigation. 1995;96(4):1722-1729.
  121. Fryburg DA, Louard RJ, Gerow KE, Gelfand RA, Barrett EJ. Growth hormone stimulates skeletal muscle protein synthesis and antagonizes insulin's antiproteolytic action in humans. Diabetes. 1992;41(4):424-429.
  122. Copeland KC, Nair KS. Recombinant human insulin-like growth factor-I increases forearm blood flow. The Journal of clinical endocrinology and metabolism. 1994;79(1):230-232.
  123. Fryburg DA. NG-monomethyl-L-arginine inhibits the blood flow but not the insulin-like response of forearm muscle to IGF- I: possible role of nitric oxide in muscle protein synthesis. The Journal of clinical investigation. 1996;97(5):1319-1328.
  124. Boger RH, Skamira C, Bode-Boger SM, Brabant G, von zur Muhlen A, Frolich JC. Nitric oxide may mediate the hemodynamic effects of recombinant growth hormone in patients with acquired growth hormone deficiency. A double-blind, placebo-controlled study. The Journal of clinical investigation. 1996;98(12):2706-2713.
  125. Bartke A, Darcy J. GH and ageing: Pitfalls and new insights. Best practice & research. Clinical endocrinology & metabolism. 2017;31(1):113-125.
  126. Yarasheski KE, Campbell JA, Smith K, Rennie MJ, Holloszy JO, Bier DM. Effect of growth hormone and resistance exercise on muscle growth in young men. The American journal of physiology. 1992;262(3 Pt 1):E261-267.
  127. Yarasheski KE, Zachwieja JJ, Campbell JA, Bier DM. Effect of growth hormone and resistance exercise on muscle growth and strength in older men. The American journal of physiology. 1995;268(2 Pt 1):E268-276.
  128. Taaffe DR, Pruitt L, Reim J, et al. Effect of recombinant human growth hormone on the muscle strength response to resistance exercise in elderly men. The Journal of clinical endocrinology and metabolism. 1994;79(5):1361-1366.
  129. Papadakis MA, Grady D, Black D, et al. Growth hormone replacement in healthy older men improves body composition but not functional ability. Annals of internal medicine. 1996;124(8):708-716.
  130. Hermansen K, Bengtsen M, Kjaer M, Vestergaard P, Jorgensen JOL. Impact of GH administration on athletic performance in healthy young adults: A systematic review and meta-analysis of placebo-controlled trials. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society. 2017;34:38-44.
  131. Waters D, Danska J, Hardy K, et al. Recombinant human growth hormone, insulin-like growth factor 1, and combination therapy in AIDS-associated wasting. A randomized, double-blind, placebo-controlled trial. Annals of internal medicine. 1996;125(11):865-872.
  132. Schambelan M, Mulligan K, Grunfeld C, et al. Recombinant human growth hormone in patients with HIV-associated wasting. A randomized, placebo-controlled trial. Serostim Study Group. Annals of internal medicine. 1996;125(11):873-882.

 

Multiple Endocrine Neoplasia Type 4

ABSTRACT

MEN4 (OMIM #610755) has many similarities with MEN1 but is caused by germline mutations in CDKN1B. MEN4 is rarer than MEN1. Clinical manifestations of MEN4 encompass primary hyperparathyroidism, pituitary adenomas, and gastroenteropancreatic neuroendocrine neoplasms. In line with MEN1 other neoplasms may occur.

INTRODUCTION

MEN4 (OMIM #610755) was initially named MENX and was first described in rats (1-3). MEN4 is caused by germline mutations in CDKN1B (Cdkn1b in rats), a tumor suppression gene encoding for the protein p27Kip1 (commonly referred to as p27 or as KIP1) (4). The CDKN1B gene is located on chromosome 12p13.1 (5). p27 is a member of the cyclin-dependent kinase inhibitor (CDKI) family which regulates the cell cycle (6, 7). Germline mutations in CDKN1B lead to reduced expression of p27, thereby resulting in uncontrolled cell cycle progression. To date, most of the reported human mutations were missense. These mutations were deemed pathogenic due to their in vivo or in vitro effects on the function of p27. In humans, two CDKI families have been identified: the INK4a/ARF family and the Cip/Kip family (8). Natalia Pellegata and colleagues reported in 2006 a three-generation family with apparently MEN1-related tumors, but this kindred turned out to become the first reported cases of MEN4 in humans (2). The incidence of CDKN1B mutations in patients with a MEN1-related phenotype is likely to be in the range of 1-4% (9-11). MEN4 screening has been recommended for all patients with a MEN1-related phenotype without the presence of a MEN1 gene mutation, but the yield seems to be extremely low (< 0.1%) (12, 13). All first-degree relatives of patients with MEN4 should be offered genetic testing (14-16). The offspring of an individual with MEN4 has a 50% chance of inheriting the CDKN1B pathogenic variant (17). Possible genotype-phenotype correlations might exist (18).

CLINICAL FEATURES OF MEN4

Primary Hyperparathyroidism

Primary hyperparathyroidism has been reported in up to 80%-90% of cases with MEN4 (3). The indications for parathyroid surgery in MEN4 are the same as for MEN1 and the approach in MEN4-related primary hyperparathyroidism may be similar to that in MEN1 (19-22). It is suggested that screening for hyperparathyroidism with serum calcium measurements (and parathyroid hormone levels (PTH) if indicated) should start at the age of 15 years in MEN4 mutation carriers (23, 24).

Pituitary Adenomas

Pituitary involvement in MEN4 is the second most common manifestation of the disease, affecting approximately 1/3 of the reported cases to date. The types of pituitary disorders in MEN4 include: nonfunctional pituitary adenoma, acromegaly and gigantism, prolactinoma, or Cushing’s disease (16, 22, 25-36). Pituitary tumors in MEN4 generally present with less aggressiveness and smaller size compared to those in MEN1 (28). The management of pituitary tumors in MEN4 is similar to other sporadic or familial cases (19). Routine surveillance for the development of pituitary tumors in patients with MEN4 should be performed on a case-by-case basis and follow the current guidelines for MEN1 (19, 24).

 

Gastroenteropancreatic Neuroendocrine Neoplasms (GEP NENs)

The prevalence of GEP NENs in MEN4 is approximately 25%. These include gastroduodenal or pancreatic NENs (panNENs), which are either nonfunctioning or secreting several peptides and hormones, including gastrin, insulin, adrenocorticotropic hormone (ACTH), or vasoactive intestinal polypeptide (VIP) (11, 20, 22, 25, 37-39). It appears that there is a decreased penetrance of gastroduodenal NENs or panNENs in MEN4 as compared to MEN1. The clinical syndromes associated with these hormonal overproductions can be found elsewhere in Endotext (40-43). The diagnosis and management of panNENs in MEN4 is similar to that in MEN1 (19). Screening for gastroduodenal NENs and panNENs should be initiated according to MEN1 screening protocols (19).

Other Neoplasms

Cervical neuroendocrine carcinoma (NEC), secreting and nonsecreting adrenal tumors, testicular cancer, breast cancer, papillary and medullary thyroid cancer, colon cancer, thymic and lung carcinoids, and meningioma have been reported incidentally in MEN4 cases (2, 9, 11, 15, 22, 23, 34, 36, 44, 45).

REFERENCES

  1. Fritz A, Walch A, Piotrowska K, Rosemann M, Schäffer E, Weber K, et al. Recessive transmission of a multiple endocrine neoplasia syndrome in the rat. Cancer Res. 2002;62(11):3048-51.
  2. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Höfler H, et al. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A. 2006;103(42):15558-63.
  3. Lee M, Pellegata NS. Multiple endocrine neoplasia type 4. Front Horm Res. 2013;41:63-78.
  4. Bencivenga D, Stampone E, Azhar J, Parente D, Ali W, Del Vecchio V, et al. p27(Kip1) and Tumors: Characterization of CDKN1B Variants Identified in MEN4 and Breast Cancer. Cells. 2025;14(3).
  5. Philipp-Staheli J, Payne SR, Kemp CJ. p27(Kip1): regulation and function of a haploinsufficient tumor suppressor and its misregulation in cancer. Exp Cell Res. 2001;264(1):148-68.
  6. Hengst L, Reed SI. Translational control of p27Kip1 accumulation during the cell cycle. Science. 1996;271(5257):1861-4.
  7. Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P, et al. Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell. 1994;78(1):59-66.
  8. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13(12):1501-12.
  9. Georgitsi M, Raitila A, Karhu A, van der Luijt RB, Aalfs CM, Sane T, et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J Clin Endocrinol Metab. 2007;92(8):3321-5.
  10. Molatore S, Marinoni I, Lee M, Pulz E, Ambrosio MR, degli Uberti EC, et al. A novel germline CDKN1B mutation causing multiple endocrine tumors: clinical, genetic and functional characterization. Hum Mutat. 2010;31(11):E1825-35.
  11. Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. J Clin Endocrinol Metab. 2009;94(5):1826-34.
  12. Chevalier B, Coppin L, Romanet P, Cuny T, Maïza JC, Abeillon J, et al. Beyond MEN1, When to Think About MEN4? Retrospective Study on 5600 Patients in the French Population and Literature Review. J Clin Endocrinol Metab. 2024;109(7):e1482-e93.
  13. Faggiano A, Fazzalari B, Mikovic N, Russo F, Zamponi V, Mazzilli R, et al. Clinical Factors Predicting Multiple Endocrine Neoplasia Type 1 and Type 4 in Patients with Neuroendocrine Tumors. Genes (Basel). 2023;14(9).
  14. de Laat JM, van der Luijt RB, Pieterman CR, Oostveen MP, Hermus AR, Dekkers OM, et al. MEN1 redefined, a clinical comparison of mutation-positive and mutation-negative patients. BMC Med. 2016;14(1):182.
  15. Alrezk R, Hannah-Shmouni F, Stratakis CA. MEN4 and CDKN1B mutations: the latest of the MEN syndromes. Endocr Relat Cancer. 2017;24(10):T195-t208.
  16. Schernthaner-Reiter MH, Trivellin G, Stratakis CA. MEN1, MEN4, and Carney Complex: Pathology and Molecular Genetics. Neuroendocrinology. 2016;103(1):18-31.
  17. Brock P, Kirschner L. Multiple Endocrine Neoplasia Type 4. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Amemiya A, editors. GeneReviews(®). Seattle (WA): University of Washington, Seattle; 1993.
  18. Halperin R, Arnon L, Nasirov S, Friedensohn L, Gershinsky M, Telerman A, et al. Germline CDKN1B variant type and site are associated with phenotype in MEN4. Endocr Relat Cancer. 2023;30(1).
  19. Pieterman CRC, van Leeuwaarde RS, van den Broek MFM, van Nesselrooij BPM, Valk GD. Multiple Endocrine Neoplasia Type 1. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  20. Tonelli F, Giudici F, Giusti F, Marini F, Cianferotti L, Nesi G, et al. A heterozygous frameshift mutation in exon 1 of CDKN1B gene in a patient affected by MEN4 syndrome. Eur J Endocrinol. 2014;171(2):K7-k17.
  21. Mazarico-Altisent I, Capel I, Baena N, Bella-Cueto MR, Barcons S, Guirao X, et al. Novel germline variants of CDKN1B and CDKN2C identified during screening for familial primary hyperparathyroidism. J Endocrinol Invest. 2023;46(4):829-40.
  22. Seabrook A, Wijewardene A, De Sousa S, Wong T, Sheriff N, Gill AJ, et al. MEN4, the MEN1 Mimicker: A Case Series of three Phenotypically Heterogenous Patients With Unique CDKN1B Mutations. J Clin Endocrinol Metab. 2022;107(8):2339-49.
  23. Frederiksen A, Rossing M, Hermann P, Ejersted C, Thakker RV, Frost M. Clinical Features of Multiple Endocrine Neoplasia Type 4: Novel Pathogenic Variant and Review of Published Cases. J Clin Endocrinol Metab. 2019;104(9):3637-46.
  24. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4). Mol Cell Endocrinol. 2014;386(1-2):2-15.
  25. Occhi G, Regazzo D, Trivellin G, Boaretto F, Ciato D, Bobisse S, et al. A novel mutation in the upstream open reading frame of the CDKN1B gene causes a MEN4 phenotype. PLoS Genet. 2013;9(3):e1003350.
  26. Crona J, Gustavsson T, Norlén O, Edfeldt K, Åkerström T, Westin G, et al. Somatic Mutations and Genetic Heterogeneity at the CDKN1B Locus in Small Intestinal Neuroendocrine Tumors. Ann Surg Oncol. 2015;22 Suppl 3:S1428-35.
  27. Sambugaro S, Di Ruvo M, Ambrosio MR, Pellegata NS, Bellio M, Guerra A, et al. Early onset acromegaly associated with a novel deletion in CDKN1B 5'UTR region. Endocrine. 2015;49(1):58-64.
  28. Stratakis CA, Tichomirowa MA, Boikos S, Azevedo MF, Lodish M, Martari M, et al. The role of germline AIP, MEN1, PRKAR1A, CDKN1B and CDKN2C mutations in causing pituitary adenomas in a large cohort of children, adolescents, and patients with genetic syndromes. Clin Genet. 2010;78(5):457-63.
  29. Ikeda H, Yoshimoto T, Shida N. Molecular analysis of p21 and p27 genes in human pituitary adenomas. Br J Cancer. 1997;76(9):1119-23.
  30. Dahia PL, Aguiar RC, Honegger J, Fahlbush R, Jordan S, Lowe DG, et al. Mutation and expression analysis of the p27/kip1 gene in corticotrophin-secreting tumours. Oncogene. 1998;16(1):69-76.
  31. Lindberg D, Akerström G, Westin G. Mutational analysis of p27 (CDKN1B) and p18 (CDKN2C) in sporadic pancreatic endocrine tumors argues against tumor-suppressor function. Neoplasia. 2007;9(7):533-5.
  32. Takeuchi S, Koeffler HP, Hinton DR, Miyoshi I, Melmed S, Shimon I. Mutation and expression analysis of the cyclin-dependent kinase inhibitor gene p27/Kip1 in pituitary tumors. J Endocrinol. 1998;157(2):337-41.
  33. Chasseloup F, Pankratz N, Lane J, Faucz FR, Keil MF, Chittiboina P, et al. Germline CDKN1B Loss-of-Function Variants Cause Pediatric Cushing's Disease With or Without an MEN4 Phenotype. J Clin Endocrinol Metab. 2020;105(6):1983-2005.
  34. Tichomirowa MA, Lee M, Barlier A, Daly AF, Marinoni I, Jaffrain-Rea ML, et al. Cyclin-dependent kinase inhibitor 1B (CDKN1B) gene variants in AIP mutation-negative familial isolated pituitary adenoma kindreds. Endocr Relat Cancer. 2012;19(3):233-41.
  35. De Sousa SMC. From bench to bedside in the sella: translational developments in pituitary tumour genetics. Endocr Relat Cancer. 2025.
  36. Mercè F, Asla Q, Illana FJ, Victòria F, Javier HL, Marta S, et al. A novel likely pathogenic germline variant in CDKN1B in a patient with MEN4 and medullary thyroid cancer. Fam Cancer. 2025;24(1):24.
  37. Malanga D, De Gisi S, Riccardi M, Scrima M, De Marco C, Robledo M, et al. Functional characterization of a rare germline mutation in the gene encoding the cyclin-dependent kinase inhibitor p27Kip1 (CDKN1B) in a Spanish patient with multiple endocrine neoplasia-like phenotype. Eur J Endocrinol. 2012;166(3):551-60.
  38. Belar O, De La Hoz C, Pérez-Nanclares G, Castaño L, Gaztambide S. Novel mutations in MEN1, CDKN1B and AIP genes in patients with multiple endocrine neoplasia type 1 syndrome in Spain. Clin Endocrinol (Oxf). 2012;76(5):719-24.
  39. Han HJ, Moalem J, Shih AR, Gigliotti BJ. Insulinoma: A Novel Presentation of Multiple Endocrine Neoplasia 4. AACE Clin Case Rep. 2025;11(2):93-7.
  40. Jensen RT, Ito T. Gastrinoma. In: Feingold KR, Ahmed SF, Anawalt B, Blackman MR, Boyce A, Chrousos G, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  41. de Herder WW, Hofland J. Insulinoma. In: Feingold KR, Ahmed SF, Anawalt B, Blackman MR, Boyce A, Chrousos G, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  42. de Herder WW, Hofland J. Vasoactive Intestinal Peptide-Secreting Tumor (VIPoma). In: Feingold KR, Ahmed SF, Anawalt B, Blackman MR, Boyce A, Chrousos G, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  43. Tsoli M, Dimitriadis GK, Androulakis, II, Kaltsas G, Grossman A. Paraneoplastic Syndromes Related to Neuroendocrine Tumors. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
  44. Desrosiers-Battu LR, Lee JH, Tarasiewicz I, Gilbert AR, Galvan EM, Singh AK, et al. Anaplastic meningioma in a 6-year-old with somatic YAP1::MAML2 fusion and multiple endocrine neoplasia type 4 (MEN4) syndrome. Cancer Genet. 2025;292-293:106-10.
  45. Singeisen H, Renzulli MM, Pavlicek V, Probst P, Hauswirth F, Muller MK, et al. Multiple endocrine neoplasia type 4: a new member of the MEN family. Endocr Connect. 2023;12(2).

Pituitary Gigantism

ABSTRACT

 

Pituitary gigantism in a child is an extraordinarily rare condition that results from excessive production of growth hormone. It can present as early as infancy or not until adolescence. It may be congenital or acquired, occurring as a sporadic condition or in the context of a known syndrome in which hypersecretion of GH is a feature. Conditions in which GH excess occurs include Neurofibromatosis Type 1, McCune-Albright syndrome, Multiple Endocrine Neoplasia Type 1, Carney Complex, Isolated Familial Somatotropinomas, and X-Linked Acrogigantism. Therapeutic modalities for the treatment of pituitary gigantism are the same as those for acromegaly (adult-onset GH excess) and include surgery, medication, and radiation. Great strides have been made in identification of the molecular genetic basis for pituitary gigantism, affording novel insights into the mechanisms underlying normal and abnormal growth. Etiologies, phenotypic features, and diagnostic and treatment considerations are reviewed in this chapter.

 

ILLUSTRATIVE CASE

 

A 13 year 6-month-old boy presents for evaluation of rapid growth. Parents report that he was always tall as a child, but they have noticed that he is now taller than most classmates. He developed signs of puberty (body odor, pubic hair) a year ago coincident with the onset of rapid growth. His parents are concerned and want to make sure “everything is normal”. He is asymptomatic other than periodic headaches that developed during the last year.

 

He was born appropriate for gestational age (AGA) at term following an uncomplicated pregnancy. By 1 year of age he was noted to be tall for his age, but this was attributed to the tall stature of his parents. Father stands 6’2” and Mother is 5’8”. They are both healthy. He is an only child.

 

Upon review of his medical record he has a growth velocity of 19 cm/year (7.5 in/year) over the last calendar year; last year at the PCP the height was 160 cm, which is at 82.7% (0.9SDS)

 

He is currently at the 99.0 % for height at 179 cm/70.5 inches (+2.36 SDS) thus confirming the rapid gain in height. (See attached growth curves. Figure 1) On physical examination he is tall, but proportionate. Visual field testing shows normal vision in all fields. Thyroid examination is normal. There are no areas of skin hyperpigmentation and no obvious skeletal abnormalities other than acral enlargement. Pubic hair is Tanner stage 3 and testicular volumes are 10 and 12 cc.

Figure 1. Growth curves

Bone Age is 14 years yielding a predicted adult height of 193.1 cm (76 inches) which, at +2.35 SDS, is above his family genetic height potential. A random serum GH concentration in the morning is 15 ng/ml with a corresponding IGF1 level of 720 ng/ml. (normal range for age and pubertal status in a male: 123-701 ng/ml). Because of the excessive growth and elevated IGF1, a GH suppression test was conducted. GH concentration 120 min after 75g of glucose administered orally was 4 ng/ml. An MRI of the brain was ordered.

 

Approach

  

Statural growth is a dynamic process that varies in children during development. Unlike adults who reach a final height greater than 2 SDS for their genetic, sex, and ethnic population of origin, the definition of gigantism in children must include a growth pattern that diverges from normal. This would include the child who exceeds expected growth curve (moving up from established percentiles) or has a growth velocity exceeding the normal range for sex, pubertal stage, and age. Once the growth rate is determined to be significantly greater than normal, establishing biochemical evidence of growth hormone hypersecretion is critical to the evaluation. Measuring IGF1 levels and assessing the suppressibility of GH following a glucose load are the most useful biochemical tests. Prompt MRI imaging evaluating size, invasiveness, and extrasellar extension of a pituitary adenoma is key. Since close to 50% of patients with pituitary gigantism have a discernable genetic cause, genetic counseling and testing are helpful in management. The case is continued at the end of the chapter.

 

INTRODUCTION

 

The association between gigantism and acromegaly was recognized as early as the late 1880’s (1), when it was noted that pituitary giants invariably developed acromegalic features such as progressive enlargement of the head, face, hands, and feet (2). (See Appendix) The major difference between these two conditions is that pituitary gigantism results from excessive GH production during the period of active skeletal growth whereas acromegaly results from GH excess ensuing after epiphyseal fusion. A further distinction relates to the overall incidence of these disorders. While acromegaly is uncommon, occurring at an estimated worldwide annual rate of 2.8-4 cases per million (3), pituitary gigantism is extremely rare, with an estimated incidence of 8 per million person-years and the total number of reported cases thus far numbering only in the hundreds. Despite these disparities, a degree of clinical overlap is evident by the observation that 10% of patients with acromegaly have tall stature (4), indicating that the onset of GH excess pre-dated epiphyseal fusion in many.

 

GH hypersecretion may occur sporadically or within a constellation of abnormalities in the setting of several well- recognized syndromes. Conversely, a genetic predilection to the development of GH-secreting pituitary adenomas only may be present, as is the case in kindreds with isolated familial somatotropinomas. In recent years there has been increased recognition of the underlying molecular genetic abnormalities that lead to pituitary gigantism, one of which can be identified in approximately 50% of cases (5). Regardless of the underlying etiology, the clinical manifestations of chronic GH hypersecretion in childhood are indistinguishable, and the initial diagnostic evaluation standardized. The various categories and sources of GH excess along with their associated genetic abnormalities are discussed individually.

 

IDIOPATHIC SPORADIC FORMS OF PITUITARY GIGANTISM

 

Unlike in acromegalic adults, in whom discreet pituitary adenomas are present in the overwhelming majority (6), several different pathologic mechanisms underly childhood GH hypersecretion. These relate to the concept that pituitary gigantism represents a distinct entity, with different characteristics in terms of pituitary morphology and function. Supporting this view are reports of diffuse pituitary hyperplasia in the setting of early-onset gigantism in which congenital growth hormone releasing-hormone (GHRH) excess has been proposed as the inciting cause (7;8). Additionally, the nearly ubiquitous finding of combined GH and prolactin over-secretion in nearly all cases of early childhood gigantism, a feature not universally present in acromegaly, suggests separate pathologic processes. This dual hormonal secretion has been attributed to the presence of mammo-somatotrophs (9;10), which are rare in adults but predominate in fetal life. Even in cases of apparent pituitary microadenomas or macroadenomas arising during early childhood, this unique biochemical feature has been present (11;12). In contrast, prolactin levels are usually normal in cases of pituitary GH-secreting adenomas originating during adolescence, which may be thought of as existing within the spectrum of adult GH hypersecretion. Interestingly, a reversible transformation of pituitary somatotrophs into bi-hormonal mammo-somatotrophs when exposed to ectopic overproduction of GHRH has been observed, lending additional support to the concept that hypothalamic GHRH excess may play a pivotal role in the genesis of early-onset gigantism (13).

 

GH-secreting tumors are all derived from PIT1-lineage cells. Those composed of somatotrophs may be densely granulated, resembling normal somatotrophs, or sparsely granulated with unusual fibrous bodies. As mentioned above, those composed of mammo-somatotrophs also produce prolactin whereas rare pluri-hormonal tumors composed of cells that resemble mammo-somatotrophs also produce TSH. Some pituitary neuroectodermal tumors (PitNETs) composed of immature PIT1-lineage cells that do not resemble differentiated somatotrophs, mammo-somatotrophs, lactotroph, or thyrotrophs may also cause GH excess. An unusual oncocytic PIT1-lineage tumor known as the acidophil stem cell tumor is predominantly a lactotroph tumor but may express GH. Immature PIT1-lineage cells that express variable amounts of hormones alone or in combination can also sometimes cause GH excess (14)

 

An additional cause of sporadic pituitary gigantism linked to CNS pathology is that which occurs in the setting of a hypothalamic gangliocytoma or neurocytoma. These rare tumors, comprised of large hypothalamic-like ganglion cells, produce GHRH (15;16) and are found in close proximity to pituitary growth hormone-secreting adenomas (17). Normalization of serum growth hormone levels following resection of the hypothalamic tumor in some patients further supports a central role for abnormal GHRH secretion in the development of gigantism or acromegaly in these cases (18).

 

SYNDROMIC AND FAMILIAL FORMS OF PITUITARY GIGANTISM

 

A second major category of childhood GH hypersecretion is that which occurs in the setting of a recognized syndrome. In these cases, gigantism may be the sole presenting feature or it may be detected during clinical follow-up for endocrine or nonendocrine problems. Alternatively, biochemical evidence of sub-clinical GH excess may be revealed through routine surveillance in a child known to be at risk for the development of gigantism. As is the case in sporadic GH hypersecretion, a variety of different morphologic abnormalities involving the pituitary gland may be found. Paracrine pituitary GHRH secretion has also been implicated by the discovery of GHRH expression from clusters of cells in the hyperplastic pituitaries of two boys from a family with hereditary early-onset gigantism (19). Syndromes that are associated with the development of childhood GH excess are reviewed below. Table 1 outlines the characteristics of the GH excess and other clinical features in these disorders.

 

Table 1. Clinical Characteristics in Syndromic and Familial Pituitary Gigantism

Disorder

Mode ofInheritance

Clinical Features

Frequency ofGigantism

Typical Age of Presentation

 

PituitaryMorphology

Screening

Neurofibromatosis -1

Autosomal Dominant or Sporadic

·       Optic gliomas

·       Café au lait skin pigmentation

Extremely rare

6 months on

Optic pathway tumor with normal to small pituitary

Not routine

McCune- AlbrightSyndrome

Sporadic

·       Precocious Puberty

·       Café au lait skin pigmentation

·       Fibrous bone dysplasia

·       Multiple endocrinopathies

15-20%

Early childhood on

Pituitary adenomas or diffuse pituitary hyperplasia or no visible abnormality

Annually

Multiple Endocrine Neoplasia Type 1

Autosomal Dominant or Sporadic

Pituitary, pancreatic and parathyroid adenomas

10-60%

10% by

age 40 but has occurred as early as age 5

Pituitary adenoma

Annually beginning at age 5

Multiple Endocrine Neoplasia Type 4

Autosomal Dominant or Sporadic

Pituitary, pancreatic and parathyroid adenomas

Unknown

Unknown

Pituitary adenoma

Not established

Carney Complex

Autosomal Dominant or Sporadic

Multiple endocrine tumors

Skin lentigines

Cardiac myxomas

Neural sheath tumors

10%

Usually 3rd & 4th decade

Pituitary adenoma or pituitary hyperplasia

Annually beginning post-pubertally

3PA Association

Autosomal Dominant or Sporadic

Pheochromocytoma, paraganglioma, pituitary adenoma

Unknown

Usually 3rd & 4th decade

Pituitary adenoma with intracytoplasmic vacuoles

As clinically indicated in unaffected family members

Isolated Familial Somatotropinomas

Autosomal Dominant or Sporadic

Isolated GH- secreting pituitary adenomas

100%

Before 3rd decade and as early as age 5

Pituitary adenoma

As clinically indicated in unaffected family members

X-linked Acrogigantism

Sporadic or X- linked

Isolated GH excess

100%

Early childhood with onset in late infancy or onset during adolescence

Pituitary adenoma or pituitary hyperplasia or both

As clinically indicated in unaffected family members

 

Neurofibromatosis-1 (NF-1)

 

  Beginning in the 1970’s, reports of gigantism occurring in young children with NF-1 have appeared in the medical literature (20). In these cases, excessive growth has been noted as early as 6 months of life (21).  Neuroimaging in these patients typically reveals an optic glioma (22), usually with infiltration into the medial temporal lobe. However, growth hormone excess has frequently been reported to be a transient phenomenon in children with NF-1, raising questions as to the necessity of treatment (23,24). Several investigations aimed at identifying the precise etiology of the gigantism in these children have been conducted, but in all cases in which tumor tissue has been available, immunostaining for GH, GHRH, and somatostatin has been uniformly negative (25;26). This, in conjunction with the known temporal lobe location of somatostatin-producing neurons, led to the hypothesis that GH excess in these patients was the result of a hypothalamic regulatory defect. Specifically, tumor infiltration of somatostatinergic pathways would presumably result in reduced somatostatin tone leading to overproduction of GHRH-mediated pituitary GH. Despite this plausible explanation, arginine-induced GH stimulation in a patient with gigantism in the setting of NF-1 showed an increase in GH secretion, contrary to the expected lack of response to arginine, which acts through somatostatin inhibition (27). Thus, the precise pathogenesis of gigantism in NF-1 remains unclear. Little information is available regarding the overall incidence of GH hypersecretion in patients with NF-1 and optic gliomas, although studies have suggested that it may occur in over 10% of affected patients, some of whom have concurrent central precocious puberty (28). Interestingly, all affected patients had a tumor involving the optic chiasm, without pituitary involvement. Optic pathway tumors are usually identified on magnetic resonance image scans as a contrast enhancing mass. (28). Interestingly, growth hormone excess has also been reported in children with sporadic optic pathway tumors without associated NF-1 (29). Figure 2 demonstrates the linear growth acceleration and figure 3 the café-au-lait pigmentation observed in a young boy with NF-1 and gigantism.

Figure 2. Growth acceleration seen in neurofibromatosis and gigantism.

Figure 3. Characteristic “coast of California” café au lait macules in a child with neurofibromatosis and gigantism.

McCune-Albright Syndrome (MAS)

 

MAS is a complex and heterogenous disorder in which GH excess typically arises in conjunction with additional endocrinopathies and other abnormalities. In the classic form, MAS displays the triad of precocious puberty, café-au-lait skin pigmentation, and fibrous dysplasia of bone. It has long been recognized, however, that individuals with MAS have a propensity to develop several additional endocrine disorders including gigantism or acromegaly (30).

 

  Elucidation of the molecular genetic defect in MAS in the early 1990’s (31) illuminated the mechanism underlying the abnormal hormone secretion. Activating mutations of Gsα, the stimulatory subunit of the heterotrimeric G-protein complex involved in intracellular signaling, are the basis for nearly all of the clinical manifestations of MAS (32). These mutations, which typically involve substitution of arginine at the 201 position with cysteine or histidine, result in unregulated signal transduction leading to increased intracellular cAMP accumulation and downstream gene transcription. All affected individuals are mosaic for the mutation, which may make confirmation with a molecular diagnosis challenging. The precise timing of the mutation during embryologic life, which occurs in a post-zygotic cell line, will ultimately determine the extent of abnormal cells and severity of the resultant clinical phenotype. The incidence of GH excess in classic MAS has been reported to be 15-21% (33.34) and may be more common in males (34). However, enhanced recognition of the frequency of atypical or forme fruste variants of MAS have the potential to increase the estimated frequency. Indeed, several historical reports of extreme gigantism where fibrous bone dysplasia was also present strongly suggest a diagnosis of MAS in these individuals, a hypothesis confirmed by molecular genetic analysis in at least one case (35.36). Subclinical growth hormone excess has also been reported in MAS, in which the only clinical manifestation may be the presence of normal stature as an adult (rather than short stature) in the context of a history of untreated precocious puberty. Additional phenotypic features in this subgroup of patients with MAS include a higher incidence of vision and hearing deficits, a rise in serum GH following a TRH test, and hyperprolactinemia (37). Growth hormone excess in MAS is typically accompanied by skull base fibrous dysplasia and is notorious for increasing craniofacial morbidity and macrocephaly (38). Early diagnosis and treatment have been found to decrease the risk of optic neuropathy in these patients (39).

 

A variety of pituitary morphologic abnormalities are found on histology and imaging in MAS patients with GH hypersecretion (40), ranging from discrete pituitary adenomas (41,42) to diffuse pituitary hyperplasia (7), to no discernible radiographic abnormality (43). Of note is the fact that the Gsα mutation found in MAS is identical to that implicated in the pathogenesis of sporadic GH-secreting pituitary adenomas, where it results in the formation of the GSP oncogene. Up to 40% of somatotroph adenomas in adults contain either an Arg201 activating mutation, or a related point substitution of glutamine at position 227 (44). Interestingly, these sporadic tumors, as well as those from patients with MAS and acromegaly, display the Gsα mutation exclusively from the maternal allele, providing evidence that the GNAS1 gene is subject to imprinting (45). Figure 4 demonstrates an area of classic café au lait skin pigmentation that crosses midline and has serrated edges in a patient with MAS.

Figure 4. Café au lait pigmentation in the typical “coast of Maine” configuration in an individual with McCune-Albright syndrome.

Multiple Endocrine Neoplasia-Type I (MEN1)

 

  MEN1 is a familial cancer syndrome characterized by autosomal dominant inheritance and multi-endocrine gland involvement. Although significant clinical heterogeneity exists in terms of specific tumor combinations, the most frequent manifestations of MEN1 are parathyroid, pancreatic, and pituitary adenomas (46). The gene for MEN1, which had previously been mapped to chromosomal locus 11q13, encodes the 610 amino acid nuclear protein, menin (47). Many different molecular genetic abnormalities within the menin gene have been identified in kindreds with MEN1, including nonsense, missense, deletion, insertion, and donor-splice mutations (48); genotype/phenotype correlations have not been observed. In all cases of MEN1, the development of neoplasia is thought to arise from a defect in normal tumor suppression via a 2-hit hypothesis. The first hit represents inheritance of a germline MEN1 mutation, leading to a heterozygous loss of the MEN1 gene in every cell (49). As menin is believed to function as a tumor suppressor protein, the second hit involves a somatic MEN1 mutation in one cell, with subsequent abnormal cellular transformation and clonal expansion. Indeed, somatic biallelic MEN1 mutations have been demonstrated to be present in at least 15% of sporadic pituitary adenomas, including somatotroph tumors (50). Anterior pituitary adenomas in individuals with known MEN1 have a reported prevalence of 10-60% and are thought to represent the first clinical manifestation of the disease in up to 25% of sporadic cases (51). Of these, the majority are prolactinomas, with GH-secreting adenomas developing in approximately 10% of individuals with MEN1 by age 40. The youngest reported case of gigantism in MEN1 occurred in a 5-year-old boy, who presented with growth acceleration and a GH-secreting mammo-somatotroph adenoma in the context of a family history of MEN1 (52). Molecular genetic analysis confirmed the germline and tumor tissue MEN1 mutations but failed to reveal an etiology for the accelerated presentation in this case. Nonetheless, current recommendations include screening for anterior pituitary hormone excess beginning at age 5 in all individuals with MEN1, as well as ascertaining MEN1 carrier status by germline mutation testing in several clinical situations (53). Interestingly, GH excess due to ectopic elaboration of GHRH from a pancreatic neuroendocrine tumor has also been reported in several individuals with MEN1 (54).

 

Multiple Endocrine Neoplasia-Type 4 (MEN4)

 

MEN4 is caused by germline mutations in the CDKN1B gene which encodes the putative tumor suppressor p27Kip1 (55). Affected patients are typically heterozygous for mutations in CDKN1B and exhibit a phenotype similar to that seen in MEN1. Because of the low number of individuals diagnosed with MEN4, screening protocols for patients and their family members have not yet been established (56).

 

Carney Complex (CNC)

 

Initially described in 1985 (57), CNC is a rare autosomal dominant disorder in which the cardinal features include multiple endocrine tumors, skin lentigines (spotty pigmentation), cardiac myxomas and neural sheath tumors. The condition shares characteristics with several other syndromes, including MEN1 (multiple endocrine tumors), MAS (endocrine hyperfunction and skin pigmentation) and Peutz-Jeghers syndrome (mucosal lentiginoses and gonadal tumors), but has a unique clinical and molecular genetic identity. Two distinct genetic abnormalities have been implicated in the pathogenesis of CNC. The first is found on 2p16 (58), although a specific candidate gene within this region has not been identified. The second involves mutations in the gene encoding the protein kinase A regulatory subunit (1α) (PRKAR1A) and explains 35-44% of both familial and sporadic cases of CNC (59). This protein, which is intricately involved in endocrine cell signaling pathways, is thought to function as a tumor suppressor. Supporting this theory has been the observation that tumors from patients with CNC (in which diminished levels of PRKAR1A are present) exhibit a 2-fold increase in cAMP responsiveness compared with control tumors (60).The identical mutation has also been found in some sporadic endocrine tumors. As with MEN1, a germline mutation is thought to be the inciting event for eventual development of the disease. The clinical presentation of CNC is extremely heterogeneous,as is the age at diagnosis. The development of GH excess is rare, occurring usually during the 3rd   and 4th decades of life, and typically found in only 10% of patients at the time of presentation (61). Thus, annual screening for GH hypersecretion is recommended only in post pubertal patients. As in cases of gigantism/acromegaly in the setting of MAS, diffuse pituitary hyperplasia (62) and concomitant hyperprolactinemia (63) are frequently seen in individuals with CNC and GH excess.

 

3PA Association

 

The constellation of paraganglioma, pheochromocytoma, and pituitary adenoma is termed 3PA Association and has been shown to be due to germline mutations in subunits of succinate dehydrogenase (56;64). Growth hormone excess typically occurs in the 3rd and 4th decades of life (65). To date, no pediatric patients with pituitary gigantism in the setting of the 3PA phenotype have been reported.

 

Familial Somatotropinomas

 

  It has long been recognized that isolated pituitary gigantism or acromegaly may occur in a familial pattern. This condition, “Familial Isolated Pituitary Adenomas” (FIPA), is defined as “the development of pituitary adenomas of any type in two or more members of a family in the absence of clinical and genetic evidence of other known syndromic diseases”.  At least 46 different affected kindreds have been reported (66). Unlike in MEN1 and CNC, GH excess tends to arise early in life, with 70% of those with the disorder diagnosed before the 3rd decade. Early childhood gigantism in this setting has also occurred, involving sisters with abnormal linear growth since age 5 (67) and a more virulent course than is seen in sporadic somatotropinomas has been suggested by a case series (68). Once assumed to represent a variant of MEN1, mutations within the menin gene as the etiology for FIPA were conclusively excluded (69;70). However, the precise molecular genetic basis for the development of pituitary GH-secreting adenomas in the majority of affected families has eluded detection. Initial investigation revealed loss of heterozygosity and linkage to a 9.7 Mb region of 11q13, suggesting the presence of an additional putative tumor suppressor gene in this region,distinct from that involved in MEN1. Subsequent studies identified inactivating mutations in the gene encoding aryl hydrocarbon receptor interacting protein (AIP) at 11q13.3 in 15%-25% of families with FIPA (71-73) making it the most common genetic defect found in these kindreds. Although the mechanism by which these mutations cause pituitary adenomas is unknown, the resulting phenotype is characterized by early-onset and aggressive disease. In an amazing case of medical sleuthing, a germline AIP mutation identified in DNA from the preserved teeth of an 18th century Irish giant was found to be an exact match for the mutation harbored by four contemporary Irish families with FIPA, indicating a common ancestor dating back more than 50 generations! Interestingly, a second potential locus for FIPA has been mapped to 2p12-16, very close to the region implicated in several kindreds with CNC (66). Additional molecular genetic analysis performed in these patients has included a search for germline mutations within the GHRH receptor gene, Gsα and Gi2α genes, all of which were normal. Similar to observations in MEN1, patients with FIPA have discreet pituitary adenomas, the majority of which are comprised solely of somatotrophs (75). However, prolactinomas, gonadotropinomas, and silent pituitary adenomas may occur in different members of the same kindred (76;77) . Macroadenomas with invasion into the cavernous sinus are common in the setting of FIPA, and treatment is notoriously difficult (77).

 

X-Linked Acrogigantism

 

An additional cause of familial gigantism and acromegaly is due to microduplication of Xq26.3 and termed X-linked acrogigantism (X-LAG). This genomic duplication was initially identified in 14 patients with gigantism and is associated with both sporadic and familial cases (78; 79). Of the four genes contained in the duplicated region, the growth hormone excess appears to result from an abnormality of GPR101, a gene that encodes for an orphan G-protein coupled receptor. This gene is markedly over-expressed in pituitary tissue from affected patients. The condition can result from either germline or somatic duplications in GPR101 and has a female predominance (80, 81). That more girls than boys have X-LAG might be related to their greater number of X chromosomes. However, a potentially lethal effect of an Xq26.3 microduplication on hemizygous male embryos is also a proposed explanation (82). Mosaicism for GPR101 duplication resulting in X-LAG has also been reported in sporadic cases involving boys (83). Patients harboring the Xq26.3 microduplication exhibit a distinct phenotype characterized by strikingly early gigantism with a median age of onset of 12 months. In addition to hypersecretion of GH, elevated circulating GHRH and prolactin have also been noted (84). Both pituitary adenomas and pituitary hyperplasia have been seen among cases testing positive for X-LAG. This discovery highlights new biological processes that will undoubtedly lead to novel insights regarding the central regulation of human growth.

 

A summary of the genetic abnormalities causing gigantism and their putative abnormalities is shown in figure 5.

Figure 5. Schematic of disorders leading to pituitary gigantism, genetic loci, and their putative targets. NF1: Neurofibromatosis type 1; XLAG: X-linked acrogigantism; MAS: McCune-Albright syndrome; CNC1: Carney complex type 1; FIPA: Familial isolated pituitary adenomatosis; MEN1: Multiple endocrine neoplasia syndrome type 1; MEN4: Multiple endocrine neoplasia syndrome type 4. The MEN syndromes display unrestrained cell replication due to lack of a tumor suppressor whereas the others affect the GH secretory pathway at the points shown. See text above for details.

CLINICAL AND BIOCHEMICAL FEATURES OF GIGANTISM

 

As would be predicted, linear growth acceleration is the cardinal feature of excessive GH production in a child or adolescent. However, the excessive linear growth observed in young children with gigantism may be accompanied or even preceded by macrocephaly and or increased weight for height. (9;11). In a large international study of patients with pituitary gigantism, the median onset of rapid growth was 13 years and occurred earlier in girls than in boys (85). Additional clinical features frequently encountered include frontal bossing, broad nasal bridge, prognathism, excessive sweating, voracious appetite, coarse facial features, and enlargement of the hands and feet. Bone age radiographs in these patients have been reported to be normal or advanced, even in the complete absence of sex steroid production. Figure 6 demonstrates the prognathism, coarse facial features and typical tall stature seen in a 12-year-old boy with gigantism, and Figure 7 illustrates enlargement of the hands in this same patient.

Figure 6. Twelve-year-old boy with pituitary gigantism measuring 6’5” with his mother. Note the coarse facial features and prominent jaw.

Figure 7. Enlarged hand of the same patient in comparison with the hand of an adult male with a height of 6’1”. The patient’s middle digit has a circumference of 9 centimeters.

The most consistent biochemical abnormality observed in patients with gigantism is an elevated IGF-1, which is known to exhibit an excellent correlation with 24-hour GH secretion (86). As previously mentioned, hyperprolactinemia is extremely common in early-onset GH hypersecretion. Depending on the individual situation, the additional pituitary screening evaluation may be normal, indicative of hypopituitarism, or central precocious puberty. Concurrent endocrinopathies may also be present, particularly in patients with syndromes such as MAS or MEN1. Rarely, alterations in glucose tolerance brought about by GH excess may result in the development of overt diabetes, leading to transient diabetic ketoacidosis (87-89) which may even be the presenting feature in rare instances (90). An additional physiologic effect of GH excess that may have clinical significance is that of increased erythropoiesis, as demonstrated by a case of acromegaly-induced polycythemia vera that resolved following surgical resection of the GH-secreting adenoma (91). The importance of GH in the regulation of red blood cell production has further been supported by the observation that pre- treatment hemoglobin concentrations in children with idiopathic growth hormone deficiency are lower than controls (92)

 

DIAGNOSTIC EVALUATION OF GH EXCESS

 

The gold standard for making the diagnosis of GH excess relies on the inability to suppress serum GH concentration following an oral glucose load. While the OGTT has been the diagnostic test of choice for many years, numeric guidelines for the expected degree of suppression in a normal individual have steadily decreased. This trend is the direct result of newer assays with an improved threshold of sensitivity for detection (93).  A normal response to a standardized glucose bolus (1.75 gm/kg up to 75 grams) utilizing the newer IRMA/ICMA assays is a GH level below 1 ng/ml (94). However, given the observation that recurrence of GH excess may be detected in patients with a GH nadir less than 1 ng/ml, and that healthy subjects nearly always suppress to below 0.14 ng/ml, some investigators have suggested that the 1 ng/ml cut-off is too liberal (95). The nadir in serum GH is typically occurs within the first 2 hours of the test. Occasionally, 24-hour integrated GH assessment may be helpful in cases in which an equivocal response to OGTT is seen (96). Despite the development of highly sensitive GH assays, generalizability of results across institutions or regions is hampered by significant heterogeneity in the availability of reference preparations and methods used by specific laboratories (97). Depending on the individual circumstance, measurement of peripheral GHRH may also be indicated to investigate the possibility of ectopic GHRH secretion. Once biochemical evidence of GH excess has been demonstrated, MRI scanning of the H-P region is obviously the next step. Figure 8 illustrates the typical appearance of a GH-secreting pituitary macroadenoma in an adolescent with gigantism.

Figure 8. Pituitary somatotroph macroadenoma in an adolescent with gigantism.

A potential pitfall in the evaluation of gigantism in adolescents is the fact that significant elevations of IGF-1 may be present during normal puberty (98). Moreover, growth hormone response to an oral glucose load in normal children has been found to be gender and pubertal-stage specific, with the highest nadir GH occurring in Tanner stage 2-3 girls (99). The effect of sex steroids on IGF-1 and GH suppression must also be considered when a diagnosis of gigantism is being considered in a child with concurrent precocious puberty, as may be the case in NF-1 or MAS. Adding to the possible diagnostic ambiguity is the fact that a significant percentage of normal tall adolescents fail to suppress serum GH in response to oral glucose testing (100). Therefore, both screening and definitive testing for GH excess should be performed in the context of high clinical suspicion, and IGF-1 levels interpreted according to age and pubertal stage-adjusted normal ranges (see figure 9).

Figure 9. Schematic evaluation of patients with suspected pituitary gigantism

TREATMENT

 

No large-scale studies evaluating various therapeutic approaches to the treatment of GH excess in pediatric patients are available. Therefore, the optimal treatment of gigantism has traditionally been extrapolated from the adult literature as well as case reports or small series involving children. As is the case in adults, the three separate modalities available for the treatment of children and adolescents are surgery, radiation, and medical therapy. Of these, the greatest recent advances by far have occurred in the realm of pharmacologic agents, resulting in an exciting armamentarium of drugs promising truly enhanced efficacy and excellent safety. Regardless of the individual treatment strategy, the goals of therapy remain the same, namely the restoration of GH and IGF-1 levels to normal (101). Of all parameters investigated, GH levels themselves appear to correlate best with overall morbidity and mortality in acromegaly (102). Table 2 summarizes the current therapeutic options as they pertain to pediatric patients, each of which is discussed below.

 

Table 2. Therapeutic Modalities in GH Excess in Pediatric Patients

 

Modality

Specific Options

Current Indications

Pediatric Experience

Surgery

Transphenoidal resection

Pituitary microadenoma or macroadenoma

Performed safely in children as young as 2 years old

 

Radiation

 

Conventional radiation

Adjuvant to surgical or medical therapy

Typically avoided if at all possible, but has been used as adjuvant therapy

Stereotactic radiosurgery,ex: gamma knife

Adjuvant therapy in patients with residual GH hypersecretion

No experience with use in children

Medical Therapy

Somatostatin analogues

·       Octreotide (Sandostatin)

·       Lanreotide

·       Primary therapy in cases of diffuse pituitary hyperplasia or severe bone disease

·       Adjuvant to surgery or radiation

·       Ectopic GH excess

Used safely in children with both sporadic and syndromic gigantism for extended periods of time alone and in combination with dopamine analogues

Depot somatostatin analogues

Sandostatin LAR

SR-lanreotide

·       Same as above

Safety and efficacy appear equivalent to non-depotpreparations

Dopamine agonists

·       Bromocriptine

Cabergoline

·       Adjuvant to somatostatin analogues and other therapies

·       Particularly useful when concurrenthyperprolactinemia is present

Used safely in children in combination with somatostatin analogues

GH receptor antagonists

Pegvisomant

·       Particularly useful for treatment of refractory disease

Has been used alone and in combination with somatostatin analogues Preliminary experience in children appears promising

 

Surgery

 

Transphenoidal resection is the treatment of choice for discreet pituitary microadenomas or macroadenomas (103), with the objective being preservation of pituitary function in association with the elimination of the GH excess, as evidenced by a rapid normalization of serum GH levels (often within one hour) and response to OGTT.  Not surprisingly, the expertise of the individual surgeon impacts the likelihood of success (104). However, while surgery cures the majority of patients with microadenomas, less than 50% of patients with macroadenomas are cured of their disease (105, 106). Moreover, extended post-operative follow-up has revealed a gradual return of GH excess over time in a substantial number of patients in whom the disease was previously deemed to be well controlled (107;108). In one large retrospective study of 208 patients with pituitary gigantism, long-term control of GH/IGF1 was achieved in only 39% (108). Experience with surgical treatment of gigantism in children and adolescents has been comparable to that observed in adults (109;110), and it has been employed safely in patients as young as 24 months (12). Although further investigation is needed, a potential role for pre-operative medical therapy has been suggested by studies indicating higher surgical remission rates and lower anesthesia risk following a several month course of a somatostatin analogue (111).

 

Radiation

 

Although traditionally included as a therapeutic option, significant problems exist with the use of conventional radiotherapy in gigantism or acromegaly. These include a low level of efficacy, delayed normalization of GH levels, and a high incidence of hypopituitarism. In the setting of MAS, radiation therapy for GH hypersecretion may contribute to malignant transformation of dysplastic bone tissue (112). Additional concerns particularly relevant to children include potential adverse neurocognitive effects and the possible development of hypothalamic obesity, both of which have been linked to cranial irradiation in pediatric patients (112;113). Therefore, radiation therapy would be considered a last resort. Improved precision and safety are observed with use of stereotactic radiosurgery in the form of the gamma knife technique, which has been successfully employed as adjuvant therapy in adults with acromegaly (112;114-116).

 

Medical Therapy

 

Although most commonly considered adjunctive to surgery or radiation, a primary role for medical therapy has always existed for those patients with diffuse pituitary hyperplasia or severe bony deformities precluding a surgical approach. As tremendous improvements in the pharmacologic agents available for use in GH excess continues to evolve (117), the number of patients offered medical therapy as first-line treatment will surely expand. The three currently existing classes of drugs for suppression of GH and IGF-1 levels are reviewed below.

 

SOMATOSTATIN ANALOGUES

 

Ever since their development in the mid-1980’s, long-acting analogues of somatostatin have held a pivotal place in the medical treatment of GH excess. These agents act by binding to somatostatin receptors within somatotroph adenomas (118). By far the greatest experience in the United States has been with octreotide, which is typically administered subcutaneously in three divided doses daily. Short-term administration of octreotide decreases GH levels within one hour in > 90% of patients with acromegaly (119), while sustained use normalizes GH and IGF-1 levels in up to 65% of patients (120). Experience with the use of octreotide in children has been similarly favorable, where it has been beneficial in the treatment of sporadic as well as syndromic gigantism (121;122). Continuous subcutaneous infusion of octreotide has also resulted in superior efficacy in controlling GH hypersecretion in a pubertal patient (123). Long-acting depot preparations of octreotide in the form of Sandostatin LAR and SR-lanreotide are also available, in which a slow release of drug is achieved through degradation of a polymer in which microspheres are encapsulated (124). This allows for monthly IM administration, resulting in a safety and efficacy profile that is comparable to or improved in contrast to traditional dosing (125). Both slow-release preparations have also been used in the treatment forms of GH excess due to ectopic GHRH secretion (126) and in MAS associated gigantism (127-129), and have been noted to have equivalent safety and efficacy (130). The development of novel somatostatin analogues has the potential to improve efficacy over existing agents (131). The major side effect of all the somatostatin analogues is an increased risk of biliary sludge and gallstones after sustained use, necessitating periodic ultrasound examinations in patients treated long-term (132).

 

DOPAMINE AGONISTS  

 

Although rarely effective alone, dopamine agonists have a valuable role as adjunctive agents in the treatment of GH excess. Due to their suppressive effects on prolactin, these drugs are particularly advantageous when hyperprolactinemia is also present, as is often the case in childhood-onset gigantism. Both bromocriptine and the more potent dopamine agonists such as cabergoline have been administered to children in combination with octreotide long-term with no apparent adverse effects (128).

 

GH RECEPTOR ANTAGONISTS    

 

The latest development in the realm of medical therapy has been the emergence of pegvisomant, a genetically engineered human GH analogue that acts as a highly selective GH antagonist (133). This is achieved through alterations in GH structure altering receptor binding compared to the native GH molecule (121), resulting in prevention of the normal extracellular dimerization of the growth hormone receptor. Administration of pegvisomant long-term to adults with acromegaly has been shown to result in normalization of serum IGF-1 levels in 97% of patients (134). Despite these extremely promising results, the implications of the nearly ubiquitous elevations in serum GH levels observed in conjunction with pegvisomant treatment initially created some concerns. Although early reports recounted an increase in tumor volume and abnormal liver enzymes in association with pegvisomant use (135;136), long-term follow has demonstrated that these complications are rare and that efficacy is very good (137;138). Combination therapy using pegvisomant along with a dopamine agonist or somatostatin analogue also appears promising (137). Thus far, preliminary experience with the use of pegvisomant alone or in combination with a somatostatin analogue for the treatment of gigantism in children also appears favorable (139). This approach resulted in successful normalization of IGFI levels in a 4 year old with NF-1 (140), a 12 year old with MAS (141), and a couple of children with persistent GH hypersecretion following surgical removal of a pituitary adenoma who had failed a somatostatin analogue (142;143). Even more reassuring is a report of long-term (up to 3.5 years) treatment using pegvisomant in 3 children with gigantism, all of whom experienced a decline in growth velocity and resolution of acromegalic features(144).

 

Treatment of Tall Stature

 

Medical treatment of children and adolescents with tall stature was more common in the past (145), particularly for girls, but is now strongly discouraged except in exceptional cases. This is because of increased cultural acceptance of tall stature and recognition of side effects of treatment, which include reduced fertility (146) and increased prevalence of depression (147) not related to adult height. Depending on the absolute height and the degree of growth potential remaining, one of the goals in the treatment of gigantism may be prevention of further linear growth in these exceptional cases. When this is the case, acceleration of epiphyseal fusion can be achieved with exogenous sex steroids (145). Short-term administration of both high dose testosterone and estrogen have been utilized for this purpose in children with gigantism, resulting in significant improvements in terms of adult height (148;149). However, such an approach would require great caution given reports of subfertility in women who were treated with high dose estrogen during adolescence with the goal of attenuating growth in the setting of constitutional tall stature (150;151).

 

CONCLUSION

 

The differential diagnosis of pituitary gigantism includes a significant number of heterogeneous disorders exhibiting a vast array of clinical and genetic features (66). In most cases, the history, physical examination and adjunctive biochemical, imaging, and/or molecular genetic testing will ultimately reveal the diagnosis. Albeit rare, pituitary gigantism affords the unique opportunity for a glimpse into the complex mechanisms of growth regulation. Thus, continued clinical and scientific investigation will enhance not only individual patient care, but also collective insight into the intricacies of the fundamental processes of human growth.

 

CASE OUTCOME

 

The MRI revealed a pituitary macroadenoma after which he underwent transsphenoidal surgery. Histopathological diagnosis was mammosomatotropic adenoma. Three months after surgery, IGF-1 normalized, nadir GH during OGTT suppressed to less than 1 ng/mL and no residual tumor was found on the MRI. Genetic testing identified a mutation in the AIP gene. This case points out the importance of early diagnosis of gigantism, as treatment delay increases long-term morbidity.

 

KEY LEARNING POINTS

 

  • Pituitary gigantism is rare but important condition resulting from excessive secretion of GH (and therefore IGF1) before fusion of epiphyseal growth plates leading to tall stature, acral enlargement, facial changes, headaches, and excessive sweating.
  • Excessive linear growth is the cardinal feature of excessive GH production in children and adolescents who have open epiphyseal growth plates.
  • There is a male preponderance (78%) in pituitary gigantism in contrast to the slight female predominance (54.5%) observed in acromegaly.
  • Once growth hormone (GH) hypersecretion has been established, prompt studies to examine pituitary anatomy and define the etiology via family history and genetic testing should be performed.
  • Normalization of GH and IGF-1 levels is the goal of therapy
  • Because nearly 50% of patients with pituitary gigantism have a known underlying genetic cause, these patients should receive genetic counseling and testing for mutations.
  • Somatotropinomas in pituitary gigantism are usually large (macroadenomas) and difficult to cure with surgery or medical therapy alone.
  • Patients with large tumors and multiple surgeries and radiotherapy are often left with multiple pituitary hormone deficiencies.

 

REFERENCES

1.       Dana C. L. Giants and Gigantism Scribner' Magazine 17,1719-185 (1895)  Giants and giantism - Digital Collections - National Library of Medicine (nih.gov)        
2.       Daughaday WH. Pituitary gigantism. Endocrinol Metab Clin North Am 1992; 21(3):633-647.

  1. Etxabe J, Gaztambide S, Latorre P, Vazquez JA. Acromegaly: an epidemiological study. J Endocrinol Invest 1993; 16(3):181-187.
  2. Sotos JF. Overgrowth. Hormonal Causes. Clin Pediatr (Phila) 1996; 35(11):579-590.
  3. Lodish MB, Trivellin G, Stratakis CA. Pituitary gigantism: update on molecular biology and management. Curr Opin Endocrinol Diabetes Obes 2016; 23(1):72-80.
  4. Melmed S. Acromegaly. N Engl J Med 1990; 322(14):966-977.
  5. Moran A, Asa SL, Kovacs K et al. Gigantism due to pituitary mammosomatotroph hyperplasia. N Engl J Med 1990; 323(5):322-327.
  6. Zimmerman D, Young WF, Jr., Ebersold MJ et al. Congenital gigantism due to growth hormone-releasing hormone excess and pituitary hyperplasia with adenomatous transformation. J Clin Endocrinol Metab 1993; 76(1):216-222.
  7. Dubuis JM, Deal CL, Drews RT et al. Mammosomatotroph adenoma causing gigantism in an 8-year old boy: a possible pathogenetic mechanism. Clin Endocrinol (Oxf) 1995; 42(5):539-549.
  8. Felix IA, Horvath E, Kovacs K, Smyth HS, Killinger DW, Vale J. Mammosomatotroph adenoma of the pituitary associated with gigantism and hyperprolactinemia. A morphological study including immunoelectron microscopy. Acta Neuropathol (Berl) 1986; 71(1-2):76-82.
  9. Blumberg DL, Sklar CA, David R, Rothenberg S, Bell J. Acromegaly in an infant. Pediatrics 1989; 83(6):998-1002.
  10. Gelber SJ, Heffez DS, Donohoue PA. Pituitary gigantism caused by growth hormone excess from infancy. J Pediatr 1992; 120(6):931-934.
  11. Vidal S, Horvath E, Kovacs K, Lloyd RV, Smyth HS. Reversible transdifferentiation: interconversion of somatotrophs and lactotrophs in pituitary hyperplasia. Mod Pathol 2001; 14(1):20-28.
  12. Asa SL, Kucharczyk W, Ezzat S. Pituitary acromegaly: not one disease. Endocr Relat Cancer. 2017 Mar;24(3):C1-C4. doi: 10.1530/ERC-16-0496. Epub 2017 Jan 25. PMID: 28122798.
  13. Araki Y, Sakai N, Andoh T, Yoshimura S, Yamada H. Central neurocytoma presenting with gigantism: case report. Surg Neurol 1992; 38(2):141-145.
  14. Isidro ML, Iglesias DP, Matias-Guiu X, Cordido F. Acromegaly due to a growth hormone-releasing hormone-secreting intracranial gangliocytoma. J Endocrinol Invest 2005; 28(2):162-165.
  15. Asada H, Otani M, Furuhata S, Inoue H, Toya S, Ogawa Y. Mixed pituitary adenoma and gangliocytoma associated with acromegaly--case report. Neurol Med Chir (Tokyo) 1990; 30(8):628-632.
  16. Asa SL, Scheithauer BW, Bilbao JM et al. A case for hypothalamic acromegaly: a clinicopathological study of six patients with hypothalamic gangliocytomas producing growth hormone-releasing factor. J Clin Endocrinol Metab 1984; 58(5):796-803.
  17. Glasker S, Vortmeyer AO, Lafferty AR et al. Hereditary pituitary hyperplasia with infantile gigantism. J Clin Endocrinol Metab 2011; 96(12):E2078-E2087.
  18. Costin G, Fefferman RA, Kogut MD. Hypothalamic gigantism. J Pediatr 1973; 83(3):419-425.
  19. Drimmie FM, MacLennan AC, Nicoll JA, Simpson E, McNeill E, Donaldson MD. Gigantism due to growth hormone excess in a boy with optic glioma. Clin Endocrinol (Oxf) 2000; 53(4):535-538.
  20. Duchowny MS, Katz R, Bejar RL. Hypothalamic mass and gigantism in neurofibromatosis: treatment with bromocriptine. Ann Neurol 1984; 15(3):302-304.
  21. Josefson JL, Listernick R, Charrow J, Habiby RL. Growth Hormone Excess in Children with Optic Pathway Tumors Is a Transient Phenomenon. Horm Res Paediatr 2016; 86(1):35-38.
  22. Sani I, Albanese A. Endocrine Long-Term Follow-Up of Children with Neurofibromatosis Type 1 and Optic Pathway Glioma. Horm Res Paediatr 2017; 87(3):179-188.
  23. Fuqua JS, Berkovitz GD. Growth hormone excess in a child with neurofibromatosis type 1 and optic pathway tumor: a patient report. Clin Pediatr (Phila) 1998; 37(12):749-752.
  24. Manski TJ, Haworth CS, Duval-Arnould BJ, Rushing EJ. Optic pathway glioma infiltrating into somatostatinergic pathways in a young boy with gigantism. Case report. J Neurosurg 1994; 81(4):595-600.
  25. Waguespack SG, Eugster EA, Pescovitz OH. Growth hormone (GH) excess in a child with neurofibromatosis type 1 (NF1) an optic pathway glioma. Pediatric Research 49[6 Suppl 2 of 2], 82A. 2001.Ref Type: Abstract
  26. Cambiaso P, Galassi S, Palmiero M et al. Growth hormone excess in children with neurofibromatosis type-1 and optic glioma. Am J Med Genet A 2017; 173(9):2353-2358.
  27. Josefson JL, Listernick R, Charrow J, Habiby RL. Growth Hormone Excess in Children with Optic Pathway Tumors Is a Transient Phenomenon. Horm Res Paediatr 2016; 86(1):35-38.
  28. Salenave S, Boyce AM, Collins MT, Chanson P. Acromegaly and McCune-Albright syndrome. J Clin Endocrinol Metab 2014; 99(6):1955-1969.
  29. Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med 1991; 325(24):1688-1695.
  30. Lumbroso S, Paris F, Sultan C. McCune-Albright syndrome: molecular genetics. J Pediatr Endocrinol Metab 2002; 15 Suppl 3:875-882.
  31. Christoforidis A, Maniadaki I, Stanhope R. McCune-Albright syndrome: growth hormone and prolactin hypersecretion. J Pediatr Endocrinol Metab 2006; 19 Suppl 2:623-625.
  32. Yao Y, Liu Y, Wang L et al. Clinical characteristics and management of growth hormone excess in patients with McCune-Albright syndrome. Eur J Endocrinol 2017; 176(3):295-303.
  33. Tinschert S, Gerl H, Gewies A, Jung HP, Nurnberg P. McCune-Albright syndrome: clinical and molecular evidence of mosaicism in an unusual giant patient. Am J Med Genet 1999; 83(2):100-108.
  34. Vogl TJ, Nerlich A, Dresel SH, Bergman C. CT of the "Tegernsee Giant": juvenile gigantism and polyostotic fibrous dysplasia. J Comput Assist Tomogr 1994; 18(2):319-322.
  35. Akintoye SO, Chebli C, Booher S et al. Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. J Clin Endocrinol Metab 2002; 87(11):5104-5112.
  36. Collins MT, Singer FR, Eugster E. McCune-Albright syndrome and the extraskeletal manifestations of fibrous dysplasia. Orphanet J Rare Dis 2012; 7 Suppl 1:S4.
  37. Boyce AM, Glover M, Kelly MH et al. Optic neuropathy in McCune-Albright syndrome: effects of early diagnosis and treatment of growth hormone excess. J Clin Endocrinol Metab 2013; 98(1):E126-E134.
  38. Vortmeyer AO, Glasker S, Mehta GU et al. Somatic GNAS mutation causes widespread and diffuse pituitary disease in acromegalic patients with McCune-Albright syndrome. J Clin Endocrinol Metab 2012; 97(7):2404-2413.
  39. Dotsch J, Kiess W, Hanze J et al. Gs alpha mutation at codon 201 in pituitary adenoma causing gigantism in a 6-year-old boy with McCune-Albright syndrome. J Clin Endocrinol Metab 1996; 81(11):3839-3842.
  40. Zumkeller W, Jassoy A, Lebek S, Nagel M. Clinical, endocrinological and radiography features in a child with McCune-Albright syndrome and pituitary adenoma. J Pediatr Endocrinol Metab 2001; 14(5):553-559.
  41. Cuttler L, Jackson JA, Saeed uz-Zafar M, Levitsky LL, Mellinger RC, Frohman LA. Hypersecretion of growth hormone and prolactin in McCune-Albright syndrome. J Clin Endocrinol Metab 1989; 68(6):1148-1154.
  42. Shimon I, Melmed S. Genetic basis of endocrine disease: pituitary tumor pathogenesis. J Clin Endocrinol Metab 1997; 82(6):1675-1681.
  43. Mantovani G, Bondioni S, Lania AG et al. Parental origin of Gsalpha mutations in the McCune-Albright syndrome and in isolated endocrine tumors. J Clin Endocrinol Metab 2004; 89(6):3007-3009.
  44. Skogseid B, Rastad J, Oberg K. Multiple endocrine neoplasia type 1. Clinical features and screening. Endocrinol Metab Clin North Am 1994; 23(1):1-18.
  45. Guru SC, Goldsmith PK, Burns AL et al. Menin, the product of the MEN1 gene, is a nuclear protein. Proc Natl Acad Sci U S A 1998; 95(4):1630-1634.
  46. Bassett JH, Forbes SA, Pannett AA et al. Characterization of mutations in patients with multiple endocrine neoplasia type 1. Am J Hum Genet 1998; 62(2):232-244.
  47. Mutch MG, Dilley WG, Sanjurjo F et al. Germline mutations in the multiple endocrine neoplasia type 1 gene: evidence for frequent splicing defects. Hum Mutat 1999; 13(3):175-185.
  48. Boggild MD, Jenkinson S, Pistorello M et al. Molecular genetic studies of sporadic pituitary tumors. J Clin Endocrinol Metab 1994; 78(2):387-392.
  49. Carty SE, Helm AK, Amico JA et al. The variable penetrance and spectrum of manifestations of multiple endocrine neoplasia type 1. Surgery 1998; 124(6):1106-1113.
  50. Stratakis CA, Schussheim DH, Freedman SM et al. Pituitary macroadenoma in a 5-year-old: an early expression of multiple endocrine neoplasia type 1. J Clin Endocrinol Metab 2000; 85(12):4776-4780.
  51. Brandi ML, Gagel RF, Angeli A et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab 2001; 86(12):5658-5671.
  52. Sala E, Ferrante E, Verrua E et al. Growth hormone-releasing hormone-producing pancreatic neuroendocrine tumor in a multiple endocrine neoplasia type 1 family with an uncommon phenotype. Eur J Gastroenterol Hepatol 2013; 25(7):858-862.
  53. Lee M, Pellegata NS. Multiple endocrine neoplasia type 4. Front Horm Res 2013; 41:63-78.
  54. Lodish MB, Trivellin G, Stratakis CA. Pituitary gigantism: update on molecular biology and management. Curr Opin Endocrinol Diabetes Obes 2016; 23(1):72-80.
  55. Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL. The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine (Baltimore) 1985; 64(4):270-283.
  56. Stratakis CA, Carney JA, Lin JP et al. 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.
  57. Kirschner LS, Carney JA, Pack SD et al. 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.
  58. Sandrini F, Stratakis C. Clinical and molecular genetics of Carney complex. Mol Genet Metab 2003; 78(2):83-92.
  59. 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.
  60. Pack SD, Kirschner LS, Pak E, Zhuang Z, Carney JA, Stratakis CA. Genetic and histologic studies of somatomammotropic pituitary tumors in patients with the "complex of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas" (Carney complex). J Clin Endocrinol Metab 2000; 85(10):3860-3865.
  61. 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.
  62. O'Toole SM, Denes J, Robledo M, Stratakis CA, Korbonits M. 15 YEARS OF PARAGANGLIOMA: The association of pituitary adenomas and phaeochromocytomas or paragangliomas. Endocr Relat Cancer 2015; 22(4):T105-T122.

65      Denes J, Swords F, Rattenberry E et al. Heterogeneous genetic background of the association of pheochromocytoma/paraganglioma and pituitary adenoma: results from a large patient cohort. J Clin Endocrinol Metab 2015; 100(3):E531-E541.

  1. Vasilev V, Daly AF, Trivellin G, et al. HEREDITARY ENDOCRINE TUMOURS: CURRENT STATE-OF-THE-ART AND RESEARCH OPPORTUNITIES: The roles of AIP and GPR101 in familial isolated pituitary adenomas (FIPA). Endocr Relat Cancer. 2020 Aug;27(8):T77-T86.
  2. Matsuno A, Teramoto A, Yamada S et al. Gigantism in sibling unrelated to multiple endocrine neoplasia: case report. Neurosurgery 1994; 35(5):952-955.
  3. Nozieres C, Berlier P, Dupuis C et al. Sporadic and genetic forms of paediatric somatotropinoma: a retrospective analysis of seven cases and a review of the literature. Orphanet J Rare Dis 2011; 6:67.
  4. Gadelha MR, Prezant TR, Une KN et al. Loss of heterozygosity on chromosome 11q13 in two families with acromegaly/gigantism is independent of mutations of the multiple endocrine neoplasia type I gene. J Clin Endocrinol Metab 1999; 84(1):249-256.
  5. Jorge BH, Agarwal SK, Lando VS et al. Study of the multiple endocrine neoplasia type 1, growth hormone-releasing hormone receptor, Gs alpha, and Gi2 alpha genes in isolated familial acromegaly. J Clin Endocrinol Metab 2001; 86(2):542-544.
  6. Beckers A, Aaltonen LA, Daly AF, Karhu A. Familial isolated pituitary adenomas (FIPA) and the pituitary adenoma predisposition due to mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene. Endocr Rev 2013; 34(2):239-277.
  7. Daly AF, Vanbellinghen JF, Khoo SK et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab 2007; 92(5):1891-1896.
  8. Martucci F, Trivellin G, Korbonits M. Familial isolated pituitary adenomas: an emerging clinical entity. J Endocrinol Invest 2012; 35(11):1003-1014.
  9. Chahal HS, Stals K, Unterlander M et al. AIP mutation in pituitary adenomas in the 18th century and today. N Engl J Med 2011; 364(1):43-50.
  10. Gadelha MR, Une KN, Rohde K, Vaisman M, Kineman RD, Frohman LA. Isolated familial somatotropinomas: establishment of linkage to chromosome 11q13.1-11q13.3 and evidence for a potential second locus at chromosome 2p16-12. J Clin Endocrinol Metab 2000; 85(2):707-714.
  11. Raverot G, Arnous W, Calender A et al. Familial pituitary adenomas with a heterogeneous functional pattern: clinical and genetic features. J Endocrinol Invest 2007; 30(9):787-790.
  12. Cansu GB, Taskiran B, Trivellin G, Faucz FR, Stratakis CA. A novel truncating AIP mutation, p.W279*, in a familial isolated pituitary adenoma (FIPA) kindred. Hormones (Athens) 2016; 15(3):441-444.
  13. Trivellin G, Daly AF, Faucz FR et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med 2014; 371(25):2363-2374.
  14. Iacovazzo D, Korbonits M. Gigantism: X-linked acrogigantism and GPR101 mutations. Growth Horm IGF Res 2016; 30-31:64-69.
  15. Iacovazzo D, Caswell R, Bunce B et al. Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study. Acta Neuropathol Commun 2016; 4(1):56.
  16. Rodd C, Millette M, Iacovazzo D et al. Somatic GPR101 Duplication Causing X-Linked Acrogigantism (XLAG)-Diagnosis and Management. J Clin Endocrinol Metab 2016; 101(5):1927-1930.
  17. Beckers A, Lodish MB, Trivellin G, et al. X-linked acrogigantism syndrome: clinical profile and therapeutic responses. Endocr Relat Cancer. 2015 Jun;22(3):353-67. doi: 10.1530/ERC-15-0038. Epub 2015 Feb 24.
  18. Daly AF, Yuan B, Fina F et al. Somatic mosaicism underlies X-linked acrogigantism syndrome in sporadic male subjects. Endocr Relat Cancer 2016; 23(4):221-233.
  19. Daly AF, Lysy PA, Desfilles C et al. GHRH excess and blockade in X-LAG syndrome. Endocr Relat Cancer 2016; 23(3):161-170.
  20. Rostomyan L, Daly AF, Petrossians P et al. Clinical and genetic characterization of pituitary gigantism: an international collaborative study in 208 patients. Endocr Relat Cancer 2015; 22(5):745-757.
  21. Barkan AL, Beitins IZ, Kelch RP. Plasma insulin-like growth factor-I/somatomedin-C in acromegaly: correlation with the degree of growth hormone hypersecretion. J Clin Endocrinol Metab 1988; 67(1):69-73.
  22. Ali O, Banerjee S, Kelly DF, Lee PD. Management of type 2 diabetes mellitus associated with pituitary gigantism. Pituitary 2007; 10(4):359-364.
  23. Alvi NS, Kirk JM. Pituitary gigantism causing diabetic ketoacidosis. J Pediatr Endocrinol Metab 1999; 12(6):907-909.
  24. Kuzuya T, Matsuda A, Sakamoto Y, Yamamoto K, Saito T, Yoshida S. A case of pituitary gigantism who had two episodes of diabetic ketoacidosis followed by complete recovery of diabetes. Endocrinol Jpn 1983; 30(3):329-334.
  25. Kuo SF, Chuang WY, Ng S et al. Pituitary gigantism presenting with depressive mood disorder and diabetic ketoacidosis in an Asian adolescent. J Pediatr Endocrinol Metab 2013; 26(9-10):945-948.
  26. Grellier P, Chanson P, Casadevall N, Abboud S, Schaison G. Remission of polycythemia vera after surgical cure of acromegaly. Ann Intern Med 1996; 124(5):495-496.
  27. Eugster EA, Fisch M, Walvoord EC, DiMeglio LA, Pescovitz OH. Low hemoglobin levels in children with in idiopathic growth hormone deficiency. Endocrine 2002; 18(2):135-136.
  28. Chapman IM, Hartman ML, Straume M, Johnson ML, Veldhuis JD, Thorner MO. Enhanced sensitivity growth hormone (GH) chemiluminescence assay reveals lower postglucose nadir GH concentrations in men than women. J Clin Endocrinol Metab 1994; 78(6):1312-1319.
  29. Melmed S, Jackson I, Kleinberg D, Klibanski A. Current treatment guidelines for acromegaly. J Clin Endocrinol Metab 1998; 83(8):2646-2652.
  30. Freda PU, Nuruzzaman AT, Reyes CM, Sundeen RE, Post KD. Significance of "abnormal" nadir growth hormone levels after oral glucose in postoperative patients with acromegaly in remission with normal insulin-like growth factor-I levels. J Clin Endocrinol Metab 2004; 89(2):495-500.
  31. Patel YC, Ezzat S, Chik CL et al. Guidelines for the diagnosis and treatment of acromegaly: a Canadian perspective. Clin Invest Med 2000; 23(3):172-187.
  32. Bidlingmaier M, Strasburger CJ. Growth hormone assays: current methodologies and their limitations. Pituitary 2007; 10(2):115-119.
  33. Le Roith D. Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors. N Engl J Med 1997; 336(9):633-640.
  34. Misra M, Cord J, Prabhakaran R, Miller KK, Klibanski A. Growth hormone suppression after an oral glucose load in children. J Clin Endocrinol Metab 2007; 92(12):4623-4629.
  35. Holl RW, Bucher P, Sorgo W, Heinze E, Homoki J, Debatin KM. Suppression of growth hormone by oral glucose in the evaluation of tall stature. Horm Res 1999; 51(1):20-24.
  36. Giustina A, Barkan A, Casanueva FF et al. Criteria for cure of acromegaly: a consensus statement. J Clin Endocrinol Metab 2000; 85(2):526-529.
  37. Rajasoorya C, Holdaway IM, Wrightson P, Scott DJ, Ibbertson HK. Determinants of clinical outcome and survival in acromegaly. Clin Endocrinol (Oxf) 1994; 41(1):95-102.
  38. Nomikos P, Buchfelder M, Fahlbusch R. The outcome of surgery in 668 patients with acromegaly using current criteria of biochemical 'cure'. Eur J Endocrinol 2005; 152(3):379-387.
  39. Gittoes NJ, Sheppard MC, Johnson AP, Stewart PM. Outcome of surgery for acromegaly--the experience of a dedicated pituitary surgeon. QJM 1999; 92(12):741-745.
  40. Jane JA, Jr., Starke RM, Elzoghby MA et al. Endoscopic transsphenoidal surgery for acromegaly: remission using modern criteria, complications, and predictors of outcome. J Clin Endocrinol Metab 2011; 96(9):2732-2740.
  41. Swearingen B, Barker FG, Katznelson L et al. Long-term mortality after transsphenoidal surgery and adjunctive therapy for acromegaly. J Clin Endocrinol Metab 1998; 83(10):3419-3426.
  42. Laws ER, Jr., Thapar K. Pituitary surgery. Endocrinol Metab Clin North Am 1999; 28(1):119-131.
  43. Rostomyan L, Daly AF, Petrossians P et al. Clinical and genetic characterization of pituitary gigantism: an international collaborative study in 208 patients. Endocr Relat Cancer 2015; 22(5):745-757.
  44. Abe T, Tara LA, Ludecke DK. Growth hormone-secreting pituitary adenomas in childhood and adolescence: features and results of transnasal surgery. Neurosurgery 1999; 45(1):1-10.
  45. Williams F, Hunter S, Bradley L et al. Clinical experience in the screening and management of a large kindred with familial isolated pituitary adenoma due to an aryl hydrocarbon receptor interacting protein (AIP) mutation. J Clin Endocrinol Metab 2014; 99(4):1122-1131.
  46. Katznelson L, Atkinson JL, Cook DM, Ezzat SZ, Hamrahian AH, Miller KK. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the diagnosis and treatment of acromegaly--2011 update. Endocr Pract 2011; 17 Suppl 4:1-44.
  47. Liu F, Li W, Yao Y et al. A case of McCune-Albright syndrome associated with pituitary GH adenoma: therapeutic process and autopsy. J Pediatr Endocrinol Metab 2011; 24(5-6):283-287.
  48. Sklar C, Boulad F, Small T, Kernan N. Endocrine complications of pediatric stem cell transplantation. Front Biosci 2001; 6:G17-G22.
  49. Attanasio R, Epaminonda P, Motti E et al. Gamma-knife radiosurgery in acromegaly: a 4-year follow-up study. J Clin Endocrinol Metab 2003; 88(7):3105-3112.
  50. Castinetti F, Taieb D, Kuhn JM et al. Outcome of gamma knife radiosurgery in 82 patients with acromegaly: correlation with initial hypersecretion. J Clin Endocrinol Metab 2005; 90(8):4483-4488.
  51. Swords FM, Allan CA, Plowman PN et al. Stereotactic radiosurgery XVI: a treatment for previously irradiated pituitary adenomas. J Clin Endocrinol Metab 2003; 88(11):5334-5340.
  52. Grasso LF, Pivonello R, Colao A. Investigational therapies for acromegaly. Expert Opin Investig Drugs 2013; 22(8):955-963.
  53. Patel YC, Greenwood M, Panetta R et al. Molecular biology of somatostatin receptor subtypes. Metabolism 1996; 45(8 Suppl 1):31-38.
  54. Ezzat S, Snyder PJ, Young WF et al. Octreotide treatment of acromegaly. A randomized, multicenter study. Ann Intern Med 1992; 117(9):711-718.
  55. Newman CB, Melmed S, George A et al. Octreotide as primary therapy for acromegaly. J Clin Endocrinol Metab 1998; 83(9):3034-3040.
  56. Feuillan PP, Jones J, Ross JL. Growth hormone hypersecretion in a girl with McCune-Albright syndrome: comparison with controls and response to a dose of long-acting somatostatin analog. J Clin Endocrinol Metab 1995; 80(4):1357-1360.
  57. Schoof E, Dorr HG, Kiess W et al. Five-year follow-up of a 13-year-old boy with a pituitary adenoma causing gigantism--effect of octreotide therapy. Horm Res 2004; 61(4):184-189.
  58. Nanto-Salonen K, Koskinen P, Sonninen P, Toppari J. Suppression of GH secretion in pituitary gigantism by continuous subcutaneous octreotide infusion in a pubertal boy. Acta Paediatr 1999; 88(1):29-33.
  59. Freda PU. Somatostatin analogs in acromegaly. J Clin Endocrinol Metab 2002; 87(7):3013-3018.
  60. Flogstad AK, Halse J, Bakke S et al. Sandostatin LAR in acromegalic patients: long-term treatment. J Clin Endocrinol Metab 1997; 82(1):23-28.
  61. Drange MR, Melmed S. Long-acting lanreotide induces clinical and biochemical remission of acromegaly caused by disseminated growth hormone-releasing hormone-secreting carcinoid. J Clin Endocrinol Metab 1998; 83(9):3104-3109.
  62. Ciresi A, Amato MC, Galluzzo A, Giordano C. Complete biochemical control and pituitary adenoma disappearance in a child with gigantism: efficacy of octreotide therapy. J Endocrinol Invest 2011; 34(2):162-163.
  63. Tajima T, Tsubaki J, Ishizu K, Jo W, Ishi N, Fujieda K. Case study of a 15-year-old boy with McCune-Albright syndrome combined with pituitary gigantism: effect of octreotide-long acting release (LAR) and cabergoline therapy. Endocr J 2008; 55(3):595-599.
  64. Zacharin M. Paediatric management of endocrine complications in McCune-Albright syndrome. J Pediatr Endocrinol Metab 2005; 18(1):33-41.
  65. Murray RD, Melmed S. A critical analysis of clinically available somatostatin analog formulations for therapy of acromegaly. J Clin Endocrinol Metab 2008; 93(8):2957-2968.
  66. Hofland LJ, van der HJ, van Koetsveld PM et al. The novel somatostatin analog SOM230 is a potent inhibitor of hormone release by growth hormone- and prolactin-secreting pituitary adenomas in vitro. J Clin Endocrinol Metab 2004; 89(4):1577-1585.
  67. Schmidt K, Leuschner M, Harris AG et al. Gallstones in acromegalic patients undergoing different treatment regimens. Clin Investig 1992; 70(7):556-559.
  68. Trainer PJ, Drake WM, Katznelson L et al. Treatment of acromegaly with the growth hormone-receptor antagonist pegvisomant. N Engl J Med 2000; 342(16):1171-1177.
  69. van der Lely AJ, Hutson RK, Trainer PJ et al. Long-term treatment of acromegaly with pegvisomant, a growth hormone receptor antagonist. Lancet 2001; 358(9295):1754-1759.
  70. Bernabeu I, Marazuela M, Lucas T et al. Pegvisomant-induced liver injury is related to the UGT1A1*28 polymorphism of Gilbert's syndrome. J Clin Endocrinol Metab 2010; 95(5):2147-2154.
  71. Buhk JH, Jung S, Psychogios MN et al. Tumor volume of growth hormone-secreting pituitary adenomas during treatment with pegvisomant: a prospective multicenter study. J Clin Endocrinol Metab 2010; 95(2):552-558.
  72. Higham CE, Atkinson AB, Aylwin S et al. Effective combination treatment with cabergoline and low-dose pegvisomant in active acromegaly: a prospective clinical trial. J Clin Endocrinol Metab 2012; 97(4):1187-1193.
  73. van der Lely AJ, Biller BM, Brue T et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab 2012; 97(5):1589-1597.
  74. Mangupli R, Rostomyan L, Castermans E et al. Combined treatment with octreotide LAR and pegvisomant in patients with pituitary gigantism: clinical evaluation and genetic screening. Pituitary 2016; 19(5):507-514.
  75. Main KM, Sehested A, Feldt-Rasmussen U. Pegvisomant treatment in a 4-year-old girl with neurofibromatosis type 1. Horm Res 2006; 65(1):1-5.
  76. Rix M, Laurberg P, Hoejberg AS, Brock-Jacobsen B. Pegvisomant therapy in pituitary gigantism: successful treatment in a 12-year-old girl. Eur J Endocrinol 2005; 153(2):195-201.
  77. Bergamaschi S, Ronchi CL, Giavoli C et al. Eight-year follow-up of a child with a GH/prolactin-secreting adenoma: efficacy of pegvisomant therapy. Horm Res Paediatr 2010; 73(1):74-79.
  78. Daniel A, d'Emden M, Duncan E. Pituitary gigantism treated successfully with the growth hormone receptor antagonist, pegvisomant. Intern Med J 2013; 43(3):345-347.
  79. Goldenberg N, Racine MS, Thomas P, Degnan B, Chandler W, Barkan A. Treatment of pituitary gigantism with the growth hormone receptor antagonist pegvisomant. J Clin Endocrinol Metab 2008; 93(8):2953-2956.
  80. Drop SL, De Waal WJ, De Muinck Keizer-Schrama SM. Sex steroid treatment of constitutionally tall stature. Endocr Rev 1998; 19(5):540-558.
  81. Venn A, Bruinsma F, Werther G, et al.  Oestrogen treatment to reduce the adult height of tall girls: long-term effects on fertility. Lancet. 2004 Oct 23-29;364(9444):1513-8.
  82. Bruinsma FJ, Venn AJ, Patton GC, et al. Concern about tall stature during adolescence and depression in later life. J Affect Disord. 2006 Apr;91(2-3):145-52. 
  83. Lu PW, Silink M, Johnston I, Cowell CT, Jimenez M. Pituitary gigantism. Arch Dis Child 1992; 67(8):1039-1041.
  84. Minagawa M, Yasuda T, Someya T, Kohno Y, Saeki N, Hashimoto Y. Effects of octreotide infusion, surgery and estrogen on suppression of height increase and 20K growth hormone ratio in a girl with gigantism due to a growth hormone-secreting macroadenoma. Horm Res 2000; 53(3):157-160.
  85. Albuquerque EV, Scalco RC, Jorge AA. MANAGEMENT OF ENDOCRINE DISEASE: Diagnostic and therapeutic approach of tall stature. Eur J Endocrinol 2017; 176(6):R339-R353.
  86. Hendriks AE, Drop SL, Laven JS, Boot AM. Fertility of tall girls treated with high-dose estrogen, a dose-response relationship. J Clin Endocrinol Metab 2012; 97(9):3107-3114.

APPENDIX  

Research into the function of the pituitary, and GH in particular, started with clinical observations and ana­tomical descriptions of people with gigantism and adults with acromegalic features (1). In 1884, the Swiss general physician Fritsche reported in great detail the history of a 44‑year-old man developing the characteristic features of acromegaly — a term later coined by Pierre Marie in 1886 (2) — and an enlarged pituitary, which was observed post-mortem (3). Minkowski proposed the connection between the pituitary and acromegaly before eosinophilic tumors of the anterior pituitary emerged as the anatomical basis of gigantism and acromegaly (4).

REFERENCES

  1. de Herder, W. W. Acromegaly and gigantism in the medical literature. Case descriptions in the era before and the early years after the initial publication of Pierre Marie (1886). Pituitary 12, 236–244 (2009).
  2. Marie, P. Sur deux cas d’acromégalie. Revue Med. Paris 6, 297–333 (1886).
  3. Fritsche, C. F. & Klebs, E. Ein Beitrag zur Pathologie des Riesenwuchses. Klinische und Pathologisch Anatomische Untersuchungen (Vogel, FCW, 1884).
  4. Minkowski, O. Übereinen fall von akromegalie. Berlin Klin. Wochenschr. 24, 371–374 (1887).

 

Disorders of Growth Hormone in Childhood

ABSTRACT

 

Growth is a fundamental process of childhood and growth disorders remain one of the commonest reasons for referral to a pediatric endocrinologist. Growth can be divided into four phases – fetal, infancy, childhood and the pubertal phase with different hormonal components influencing growth at each stage. The GH-IGF1 axis plays a major role in the childhood phase of growth with a significant role alongside sex steroids during puberty while in infancy thyroid hormone and nutrition are vital. Although an uncommon cause of short stature disorders of the GH-IGF1 axis are extremely important due to the effectiveness of recombinant human growth hormone therapy for the child with GH deficiency (GHD). Here we review the diagnosis of growth hormone deficiency through a combination of auxology, biochemistry, imaging, and genetic testing. Particular focus is given to the accuracy of IGF-1/BP3 for diagnosis as well as the known problems with GH stimulation tests and GH assays. Isolated GHD is caused by mutations in GH1, BTK, and RNPC3 while GHD seen as part of multiple pituitary hormone deficiency is known to be caused by mutations in a wide variety of genes. A variety of structural malformations of the brain can be associated with congenital GHD with the commonest being the presence of an ectopic posterior pituitary or Septo-optic dysplasia. Acquired GHD is rarer and caused by tumors, radiotherapy, hypophysitis, and traumatic brain injury.  Treatment with recombinant human GH is highly efficacious in improving height in children with GH deficiency and extremely safe. Short stature disorders are, rarely, also caused by a variety of other disorders of the GH-IGF1 axis. Resistance to growth hormone is seen in Laron syndrome and in mutations in IGF1 and IGF1R while decreased bioavailability of IGF1 is seen in ALS deficiency and PAPPA2 deficiency. Treatment with recombinant human IGF1 (rhIGF1) is available for those with IGF-I deficiency caused by either Laron syndrome or IGF1 mutations. rhIGF1 is effective in improving height but treatment is less effective than the use of GH to treat GH deficiency.  The role of IGF1 in pre-natal growth is highlighted by the phenotype of patients with IGF1R or IGF1 mutations where pre-natal growth is commonly impaired and children born small for gestational age. GH excess is much rarer than GH deficiency in childhood and can be caused by pituitary adenomas, optic nerve gliomas (seen predominantly with NF1), McCune Albright syndrome, or Carney complex. Treatment is with surgery, somatostatin analogs, or GH receptor antagonists.

 

CASE STUDY

 

 A 5-year-old girl was referred to her local community pediatrician by her health visitor with concerns about growth and poor calorie intake. Height at presentation was 91.5 cm (-4.1 SD) with weight 12.5 kg (-3.4 SD) and head circumference 48.8 cm (-2.5 SD).  Her teeth were affected by multiple caries which made chewing hard foods painful and she therefore ate only soft foods. Development was reported to be normal and she was performing well in school. Her parents had noticed loud snoring and tonsils were enlarged on examination.

 

She was born at term by vaginal delivery with a birth weight of 3.5kg and was the youngest of 6 children. The parents were consanguineous (first cousins) and there was a family history of short stature in distant cousins. Mother was 147 cm tall (-2.7 SD) and father 165.1 cm (-1.5 SD). There was a history of diabetes mellitus type 2, diabetic nephropathy and thalassemia in mother and the father had a history of recurrent kidney stones. 

 

On review in the endocrinology clinic prominent forehead, depressed nasal bridge and a high-pitched voice were noted.  General investigations (detailed below) were normal; however, IGF-I and IGFBP-3 concentrations were low with high basal GH and peak GH concentrations (the latter >40µg/L). The combination of low IGF-I with raised GH concentrations suggested a diagnosis of GH insensitivity. In view of the history of snoring the patient was referred to an ENT surgeon who noted large prolapsing tonsils with mild apneic episodes on sleep study. Due to the propensity of IGF-I therapy to induce tonsillar hypertrophy, she underwent tonsillectomy.

 

Treatment with recombinant human IGF-I was started at the age of 6 years and 1 month initially with 0.6 mg (38 mcg/kg/) BD, increasing after 1 week to 1.1 mg (70 mcg/kg) BD and then to 1.7 mg (108 mcg/kg) BD. There were no problems with hypoglycemia. Height velocity increased from 3.6 cm/year to 10.3 cm/year over the first year of treatment. Sequencing of the GH receptor identified a known intronic point (A>G) mutation between exons 6 and 7 in which leads to inclusion of a pseudoexon and an additional 36 amino acids in the extracellular domain of the GHR.

 

At the age of 9 years and 3 months she was noted to be at breast stage 3 and in order to preserve height potential she has been treated with GnRH analogue (Zoladex LA). The IGF-I dose has been increased to maintain dose in the range 100 – 120 mcg/kg/BD and at 10 years 3 months height is 125.8 cm (-2.1 SD) with weight 32 kg (-0.2 SD). There has been some lipophypertrophy around the injection sites and she required an adenoidectomy due to a recurrence of her snoring (with daytime somnolescence) caused by a large obstructing adenoidal pad.

 

Baseline Investigations

Serum electrolytes, urea, creatinine, liver function tests, calcium, phosphate, hemoglobin – all normal

Karyotype 46 XX

TSH 2.2 mU/L (0.3 -5.0) free T4 17 pmol/L (11 - 24)

Prolactin 174 mU/l (85 – 250)

IGF-I <25 ng/mL (55 – 280)

IGFBP-3 0.7 mg/L (1.5 – 3.4)

ALS 3.2 mg/L (2.3 – 11)

Fasting glucose 4.0 mmol/L Insulin 2.1 mIU/L (2.3 - 26)

Skeletal survey – no evidence of skeletal dysplasia

Bone Age delayed by 18 months

 

Arginine stimulation Test

Time (min)       Growth Hormone (µg/L)

-15                   19.3

0                      4.0

15                    4.8

30                    14

60                    >40

90                    >40

120                  15.6

 

Standard Synacthen Test

Time (min)       Cortisol (nmol/L)

0 min               213

30 min             624

60 min             742

 

GnRH Test at age 5 years

Time                LH (IU/L)         FSH (IU/L)

0                      <0.1                 1.7

30                    2.7                   14

60                    3.3                   18

 

INTRODUCTION

 

Growth is a fundamental process of childhood. It can be divided into four phases – fetal, infancy, childhood, and pubertal growth. Although growth occurs as a continuum, the endocrine control of each phase is distinct. The fetal phase includes the fastest period of growth with a crown-rump velocity of 62cm/year during the second trimester. Growth during this phase is dependent upon placental function and maternal nutrition in addition to hormonal factors especially IGF-I, IGF-II and insulin (1,2).  Although size at birth (and hence fetal growth) is profoundly affected by IGF-I deficiency during fetal life (3), the effects of congenital GH deficiency are much less marked with a mild reduction in birth size (4). 

 

Fetal Phase

 

During the first year of life, growth declines from an initial velocity of around 25cm/year to around 10cm/year. Previously it has been thought that during this period growth hormone did not have a significant influence on growth however it is now clear that children with growth hormone deficiency display reduced height velocity from birth (5). In addition to growth hormone, thyroid hormone and adequate nutrition are vital for normal growth during infancy.

 

Infancy Phase

 

During the first two years of life there is a significant period of catch-up or catch-down growth so while size at birth is not well correlated with parental height, by two years of age the correlation between parental and child heights significantly improves (6). It has been hypothesized that this catch up growth is the result of a central mechanism which detects the difference between the actual and expected size and acts to increase growth velocity (7). No experimental evidence exists for this hypothesis. The second hypothesis on the origin of this catch up/down growth is that it arises from alterations in growth plate senescence. Catch down growth is associated with a reduction in the number of stem cell divisions within the growth plate while catch up growth would be due to a compensatory increase in the number of stem cell divisions within the growth plate (8).

 

Childhood Phase

 

There is a gradual transition from the infancy phase into the childhood phase of growth from 6 months to 3 years of age. Prepubertal growth velocity is relatively constant between 4-7 cm/year with the lowest growth velocity of life occurring immediately before the onset of puberty. During childhood growth is mainly controlled by the influence of the GH-IGF-I axis along with thyroid hormone.

 

Pubertal Growth

 

The final phase of growth is puberty – the period of transition from the pre-pubertal state to the full development of secondary sexual characteristics and achievement of final height. Puberty begins with the onset of activity within the hypothalamic-pituitary-gonadal axis leading to the production of androgens (in males) and estrogen (in females). In males the first sign of pubertal development is enlargement of the testes while in females it is development of breast buds. The production of androgens and estrogen is associated with an increase in activity within the GH-IGF-I axis. Administration of testosterone to boys increased both GH and IGF-I concentrations (9) but this effect is dependent upon aromatization as co-administration of an estrogen receptor antagonist (10) or administration of dihydrotestosterone (11) (the active form of testosterone that cannot be aromatized) does not lead to an increase in GH or IGF-I concentrations. In girls there is also an increase in IGF-I levels and GH secretion during puberty but the mechanisms underlying this are less clear. Administration of oral or transdermal estrogen induces a decline in serum IGF-I concentrations and a consequent increase in GH secretion (12).  

 

Fusion of the epiphyseal growth plates is induced by the activity of estrogen on ERα as patients with mutations in the genes encoding Erα (13) or aromatase enzyme (14) result in failure of fusion of the epiphyses and tall stature.

 

This chapter will firstly discuss the physiology of the GH-IGF-I axis along with signal transduction of GH and IGF-I and then consider the diagnosis and treatment of growth hormone deficiency before discussing individual pathological conditions associated with both GH deficiency and GH excess. Disorders leading to GH deficiency have been divided into congenital and acquired. 

 

GH-IGF-I AXIS

 

Physiology of the GH-IGF-I Axis

 

Release of Growth Hormone Releasing Hormone (GHRH) from the hypothalamus regulates the secretion of GH from the anterior pituitary both by increasing GH1 gene transcription and by promoting the secretion of stored GH. GHRH release is pulsatile and influenced by somatostatin and Ghrelin. Ghrelin is a 28 amino acid peptide produced in the stomach (15) and acts via the GH secretagogue receptor (GHSR). The active hormone is the octanoylated form produced by Ghrelin O-acetyltransferase(16) and is cleaved from the 117 amino acid preprohormone. In addition to the role in GH secretion Ghrelin also acts as an appetite stimulant (17) and stimulates the secretion of insulin (18), ACTH (19), and prolactin (19). In vivo the action of Ghrelin requires an intact GHRH system to influence GH secretion (20) but in vitro is capable of directly stimulating GH (15). Somatostatin is a peptide derived from pre-pro-somatostatin within neurons of the anterior periventricular nucleus which project to the median eminence. There are two main forms of somatostatin – 14 and 28 amino acid variants.  It acts via the somatostatin receptors of which there are 5 subtypes (SSTR1-5). The anterior pituitary expresses SSTR1, 2, 3 and 5 (21). Somatostatin acts to decrease the secretion of GH by inhibiting GHRH secretion, directly inhibiting GH secretion in the anterior pituitary (22), antagonizing the activity of Ghrelin (20) as well as inhibiting its secretion (23). Somatostatin tone determines trough levels of GH and reductions in somatostatin tone are a major factor in determining the time of a pulse of GH.  GH secretion is also stimulated by hypoglycemia and exercise. A summary of the factors influencing GH secretion is given in Figure 1.

 

GH is released from the somatotrophs of the anterior pituitary in a pulsatile manner with the pulses predominantly overnight, increasing in amplitude with age (24). The pulse amplitude is maximal in the pubertal years consistent with the raised IGF-I levels and growth velocity at this time (25).  In males there is greater diurnal variation in peak amplitude, with higher peaks overnight and a lower baseline GH level compared to females. Overall GH production is higher in females. GH peak amplitude is linked to IGF-I concentrations while nadir GH is linked to waist-hip ratio (26).  

 

Growth Hormone and GH signal Transduction

 

Growth Hormone (GH) is encoded  by the GH1 gene located at chromosome 17q23.3 and is a 191 amino acid single chain polypeptide (27). There are 20 and 22kDa isoforms of GH generated by alternative splicing (the smaller isoform lacks amino acids 32-46) with the 20kDa accounting for around 10-20% of circulating GH (28).  While GH1 is expressed within the anterior pituitary a 20kDa variant of GH is encoded by the GH2 gene but this is expressed in placenta and not in the pituitary (29).

Figure 1. Physiology of the GH-IGF-I Axis. Release of GHRH from the hypothalamus is under the control of somatostatin (inhibitory) and Ghrelin (stimulatory). Alterations in GHRH tone led to pulsatile release of GH from the anterior pituitary. GH has widespread effects on muscle, fat and in the growth plate. IGF-I is produced in liver and in local tissues in response to GH stimulation. Red lines indicate feedback loops. Figure reproduced and adapted from Butcher I Molecular and Metabolomic Mechanisms Affecting Growth in Children Born Small for Gestational Age PhD thesis University of Manchester 2013.

In the circulation GH is bound to Growth Hormone Binding Protein (GHBP). GHBP is generated either by proteolysis cleavage of the extracellular domain of the growth hormone receptor (GHR) by metzincin metalloproteinase tumor necrosis factor-α converting enzyme (30) or by alternative splicing of the GHR (31). The 22kDa isoform of GH has the highest affinity for GHBP with the 20kDa and placental GH having a lower affinity (32).  GHBP has a molecular mass of 60kDa and acts to prolong the half-life of GH with an increase from 11 minutes to 80 minutes (33). GHBP also acts to maintain the circulating pool of GH within the vasculature (34), reducing the ability of the circulating pool of GH to bind to peripheral GHRs.

 

The actions of GH are mediated via the GHR, a 620 amino acid protein containing a 246-residue extracellular domain, a single24 amino acid transmembrane helix and a 350 amino acid intracellular domain. The GHR gene is located on chromosome 5p13 and contains 10 exons. The GHR exists in a pre-dimerized form on the cell surface. In contrast to previous models, it is now recognized that dimerization per se is insufficient to initiate signaling (35).  GH binds to the GHR via two binding sites – initial binding is via the high affinity site 1 followed by binding to the low affinity binding site 2 (36). GH binding induces a conformational change in the dimerized GHR including rotation of one of the GHR subunits (see Figure 2).  This results in locking together of the extracellular receptor-receptor interaction domain and repositioning of the box 1 motifs in the intracellular domain increasing the distance between them. In turn this leads to repositioning of tyrosine kinases, including JAK2 (37). This repositioning is crucial to JAK2 activation. In the inactive state two JAK2 molecules (each attached to one of two dimerized GHRs) are positioned so that the kinase domain of one JAK2 molecule interacts with the inhibitory pseudokinase domain of the other JAK2 molecule. After repositioning, due to the conformational change induced by GH binding, the inhibitory kinase-pseudokinase interaction is lost and the kinase domains of each JAK2 molecule interact with each other leading to JAK2 activation (38).

 

Activation of the GHR results in JAK2 mediated phosphorylation of the signal transducers and activator of transcription proteins (STAT), including STAT1, STAT3, STAT5A and STAT5B. STAT5A and 5B are recruited to the phosphorylated GHR where their Src homology 2 (SH2) domain is phosphorylated by JAK2. STAT5A/B then homo- or heterodimers and translocate to the nucleus (37,39) (see Figure 3). Activation of STAT1 and STAT3 is also via phosphorylation by JAK2 but this does not require recruitment to the GHR.  JAK2 also phosphorylates the Src homology domain of SHC (leading to activation of the mitogen activated protein kinase pathway) and the insulin receptor substrates (IRS-1, IRS-2 and IRS-3), which, in turn activate phosphatidylinositol-3 kinase and induces translocation of GLUT4 to the membrane. In addition to activation of JAK2, activation of the GHR also leads to direct activation of the Src family kinases, which are capable of activating the mitogen activated protein kinase pathway (40), and activation of protein kinase C via phospholipase C. Activation of protein kinase C stimulates lipogenesis, c-fos expression and increases intracellular calcium levels by activating type 1 calcium channels.

Figure 2. Growth hormone binding to the extracellular domain of the growth hormone receptor reorients the pre-existing homodimer so that one growth hormone receptor subunit rotates relative to the other. This structural reorientation is transmitted through the transmembrane domain resulting in a repositioning of tyrosine kinases bound to the cytoplasmic domain of the receptors. The distance between the box 1 motifs increases between inactive and active states and this movement is fundamental to activation of JAK2. Phosphorylation of JAK2 in turn leads to phosphorylation of STAT molecules, activation of the MAPK cascade and activation of IRS-1. STAT5a and STAT5b homo/heterodimerize and translocate to the nucleus. Figure kindly supplied by Dr Andrew Brooks, Institute for Molecular Bioscience, The University of Queensland.

GH signal transduction is regulated via several mechanisms: JAK2 is autoinhibitory with the pseudokinase domain inhibiting the catalytic domain (41), SHP1 binds to and dephosphorylates JAK2 in response to GH and GH also phosphorylates the transmembrane signal regulatory glycoprotein SIRPα1 which dephosphorylates JAK2 and the GHR.

 

The net result of GH signal transduction is the transcription of a set of GH dependent genes and the production of IGF-I the combination of which mediates the actions of GH including effects on cell proliferation, bone density, glucose homeostasis and serum lipids.

 

Insulin Like Growth Factors, Their Binding Proteins and Signal Transduction

 

INSULIN LIKE GROWTH FACTORS

 

The two insulin-like growth factors, IGF-I and IGF-II, are single chain polypeptide hormones sharing 50% homology with insulin. IGF-I is a 70 amino acid 7.5 kDa protein with four domains – A, B, C and D. The prohormone also contains a c-terminal peptide that is cleaved in the Golgi apparatus before secretion. IGF-II is a 67 amino acid peptide also with a molecular weight of 7.5 kDa. The mitogenic and, in part, the metabolic effects of GH are mediated via IGF-I rather than IGF-II.  The IGFs circulate bound to the IGF binding proteins (IGFBPs), of which there are six classical high affinity IGFBPs. The IGFs form a ternary complex with an IGFBP and the Acid Labile Subunit (ALS), an 85kDa protein secreted by the liver.  99% of serum IGF-I is bound to a ternary complex which acts to prolong the half-life of IGF-I (42). IGF-I is produced in both the liver and in peripheral tissues and thus can act in an autocrine and paracrine manner.

 

IGF RECEPTORS

 

The IGF-1R is a transmembrane heterotetramer consisting of consisting of two extracellular α chains and two membrane-spanning β chains linked by several disulphide bonds (43). Ligand binding sites are present in the α subunits while the β subunits contain the juxtamembrane domain, tyrosine kinase domain and a carboxy terminal domain (44). Ligand binding to the α subunit activates the intrinsic tyrosine kinase activity of the β subunit which leads to autophosphorylation of tyrosine kinases in the juxtamembrane, tyrosine kinase and carboxy terminal domains. This autophosphorylation provides docking sites for substrates including the insulin receptor substrates (IRS-1, -2, -3, -4) and Shc. IRS-1 and Shc recruit the growth factor receptor bound protein 2 that associates with son of sevenless to activate the MAPK pathway. IRS-1 also activates PI3K via its regulatory subunit, p85, leading to activation of AKT which phosphorylates BAD and activates mTOR leading to inhibition of apoptosis and stimulation of proliferation. A summary of IGF-I signal transduction is given in Figure 3.

 

Mouse studies have delineated the relative contribution to growth of the GH-IGF system – deletion of Igf1 or Igf2 results in a 40% reduction in birth weight with a reduction of 55% where Igf1r is deleted (45). Deletion of Igf1 with Igf1r or Igf2 leads to a 70% reduction in birth weight and death from respiratory distress at birth (45) whereas the Igf2r appears to negatively regulate growth as deletion of this gene results in an increase in size to 130% of wild type. IGF-I is produced in both the liver and in peripheral tissues and thus can act in an autocrine and paracrine manner. It appears that autocrine/paracrine IGF-I is more important for growth than liver derived IGF-I as a hepatic specific deletion of Igf1 in mouse resulted in no impairment of growth despite a 75% reduction in serum IGF-I concentrations (46) while a triple liver specific deletion of Igf1/Igfals/Igfbp3 resulted in a 97.5% reduction in circulating IGF-I concentrations with a 6% decrease in body length (47).

Figure 3. IGF-I Signal Transduction. Binding of IGF-I leads to phosphorylation and activation of IRS-1 which, in turn, activates the PI3K and MAPK pathways.

DIAGNOSIS OF GROWTH HORMONE DEFICIENCY IN CHILDHOOD

 

The diagnosis of growth hormone deficiency in childhood is multifactorial process and includes 1) auxological assessment 2) biochemical tests of the GH-IGF-I axis and 3) radiological evaluation of the hypothalamus and pituitary (normally with MR imaging). Prior to evaluation of the GH-IGF-I axis in a short child other diagnosis such as familial short stature, hypothyroidism, Turner syndrome, chronic illness such as Crohn’s disease and skeletal dysplasias should be considered and evaluated appropriately. Patients to be evaluated for growth hormone deficiency include (48,49):

 

  1. Severe short stature (defined height >3 SD below mean)
  2. Height more than 1.5 SD below mid parental height
  3. Height >2 SD below mean with height velocity over 1 year >1 SD below the mean for chronological age or a decrease of more than 0.5 SD in height over 1 year in children aged >2 years
  4. In the absence of short stature – a height velocity more than 2 SD below mean over 1 year or >1.5 SD below mean sustained over 2 years
  5. Signs indicative of an intracranial lesion or history of brain tumor, cranial irradiation, or other organic pituitary abnormality.
  6. Radiological evidence of a pituitary abnormality
  7. Signs and/or symptoms of neonatal GHD

 

Etiology

 

Disorders of GH can be divided into those that cause growth hormone deficiency or growth hormone excess. In childhood growth hormone deficiency is rare with an incidence of 1 in 4000 while the incidence of childhood GH excess is not known but only around 200 cases have been reported in the literature (50).  Causes of GH deficiency are listed in Table 1.

 

Table 1. Causes of Growth Hormone Deficiency

Cause

Examples

Idiopathic

 

 

Genetic

GHRHR mutations

GH1 mutations

Structural brain malformations

Pituitary stalk interruption syndrome

Rathke’s cyst

Agenesis of corpus callosum

Septo-optic dysplasia

Holoprosencephaly

Midline Tumors

Craniopharyngioma

Optic nerve Glioma

Germinoma

Pituitary adenoma

Cranial Irradiation

 

 

Traumatic Brain Injury

Road Traffic Accident

 

CNS infections

 

 

Inflammation and Auto-immunity

Sarcoidosis

Langerhans Cell Histiocytosis

Hypophysitis

Psychosocial deprivation

 

 

Clinical Presentation of GH Deficiency

 

GH deficiency can present either in isolation (isolated GHD - IGHD) or in combination with other pituitary hormone insufficiencies (multiple pituitary hormone deficiency - MPHD). In the neonatal period MPHD typically presents with reduced penile size, episodes of hypoglycemia, and prolonged unconjugated hyperbilirubinemia. MPHD is associated with breech delivery, adverse incidents in pregnancy, and admission to the newborn intensive care unit (51).  Children with severe growth hormone deficiency often appear young for their age and have midface hypoplasia and increased truncal adiposity (see Figure 4). The major clinical feature of GH deficiency is growth failure; typically, this occurs after the first year of life but may be apparent earlier in severe GHD. The earliest manifestations are a reduction in height velocity followed by a reduction in height standard deviation score (SDS) adjusted for mean parental height SDS. The child’s height SDS will ultimately fall below -2SD with the time taken to achieve this depending on the severity and duration of GHD.

Figure 4. Child with Laron syndrome. Short stature with typical facial appearance of GH insensitivity with midface hypoplasia, this finding is common to GH deficiency as well.

Biochemical Assessment of the GH-IGF-I Axis

 

Multiple assays have been developed to measure GH in serum. A consensus statement of the GH-IGF-I research society in 2000 recommended that assays used should use monoclonal antibodies to measure the 22kDa variant of human GH and that the reference preparation should be the WHO standard 88/624 (a recombinant human 22kDa GH at 3 IU = 1mg) (48,52).

 

Growth Hormone Stimulation Tests and GH Profiles

 

A number of growth hormone stimulation tests have been developed and can be divided into screening tests or definitive tests. Screening tests include exercise, fasting, levodopa, and clonidine and are characterized by low toxicity, ease of administration but low specificity. Definitive tests include the insulin tolerance test, glucagon, and arginine stimulation tests. Using the auxological criteria above a peak GH concentration below 10µg/L has traditionally been used to support the diagnosis of GHD. GHD is not a dichotomous state but exists as a continuum from severe GHD to normality and there is known to be an overlap in peak GH concentrations between normal children and those with GHD.  For this reason, and due to the advent of more sensitive monoclonal antibodies based on the recombinant human GH reference standard, some units will use a more stringent cut-off for the diagnosis of GHD e.g., 7µg/L. Where the diagnosis is isolated idiopathic GHD two pharmacological tests are required. Only one provocative test of GH secretion is required in children with one or more of the following criteria:

 

  1. Central nervous system pathology affecting the pituitary or hypothalamus
  2. A history of cranial irradiation
  3. An identified pathological genetic variant known to be associated with GHD
  4. Multiple pituitary hormone deficiency

 

INSULIN TOLERANCE TEST

 

The gold standard test is considered to be the Insulin Tolerance Test.  This test relies upon an intravenous dose of insulin to induce hypoglycemia with a subsequent rise in GH expected as part of the counter regulatory response to hypoglycemia (53). Cortisol secretion also rises in response to hypoglycemia and thus this test also assesses the hypothalomo- pituitary-adrenal axis. The patient is required to fast overnight and, in the morning, a reliable intravenous line is inserted following which an insulin dose of 0.1units/kg is administered. The dose is reduced to 0.05 units/kg in children under 4 and where there is known or likely multiple pituitary hormone deficiency. This test is generally not recommended for infants and in this group the dose of insulin would be reduced further to 0.01units/kg. After administration of insulin there is careful bedside monitoring of blood glucose concentration and once the blood glucose has reached <2.6 mmol/L (47 mg/dL) the patient eats a high carbohydrate meal. Administration of 10% glucose at 2ml/kg may be required in order to restore adequate blood glucose concentrations. This should be prepared in advance of the start of the test along with an appropriate dose of IV hydrocortisone (this should be given after hypoglycemia where there is known adrenal insufficiency or where hypoglycemia is more severe or prolonged than expected). 50% dextrose is recommended by some for the correction of hypoglycemia during the test but administration of such hyperosmolar solutions has been associated with adverse outcome (54) including cerebral edema. Due to the risks associated with this test it should only ever be performed in a center with appropriate experience.

 

GLUCAGON TEST

 

The glucagon test is one of a number of safer alternative GH provocation tests. Intramuscular administration of glucagon leads to an increase in GH due to a rise in insulin levels compensating for the increase in serum glucose (55). Maximum GH peak occurs 2-3 hours after injection of glucagon. Although less common than with the insulin tolerance test hypoglycemia can occur with the glucagon stimulation test where there is an excessive insulin response. There should therefore be blood glucose monitoring throughout the test and a meal consumed at the end of the test. Nausea and vomiting are other common side effects.

 

ARGININE STIMULATION TEST

 

Arginine administration stimulates the release of GH by inhibiting somatostatin release. Following an overnight fast arginine is administered intravenously at 0.5g/kg (maximum dose 30g) over 30 minutes. Unlike glucagon or insulin, arginine does not directly cause hypoglycemia and thus the arginine stimulation test may be safer, particularly for those patients with predisposition to hypoglycemia. Examples of patients where an arginine test would be suitable where the insulin or glucagon-based tests would not be suitable include patients with diabetes and a history of seizures or children with disorders of cerebral glucose uptake (GLUT2 deficiency) where the patient should be continuously ketotic. Arginine can be combined with L-dopa or GHRH. For combined tests, particularly the arginine-GHRH test it is important to have a test specific cut off for the diagnosis of GHD as with a powerful stimulus of GH secretion a higher cut off is required (a normal peak GH response for arginine-GHRH has been defined at 19-120 µg/L(56)).  GHRH can be used on its own as a provocative agent but is greatly affected by variations in somatostatin tone leading to a highly variable response. In addition, false negative tests may occur in children with hypothalamic damage.

 

Oral agents used in GH stimulation tests include clonidine and L-Dopa. Both clonidine and L-Dopa act by increasing adrenergic tone to increase GHRH and decrease somatostatin levels. A fast of 6 hours is required prior to the test. Since clonidine is a drug used to lower blood pressure hypotension is a potential side effect. Drowsiness is also a frequent occurrence during this test.

 

INTERPRETATION

 

Significant problems exist with GH stimulation tests – peak GH varies according to the stimulus used (57), false positive results in normal pre-pubertal children are frequent (56), the tests have poor reproducibility and there is also variability in GH level with GH assay used (58). Peak GH is also reduced in obesity and for adults BMI specific cut-offs for the diagnosis of GHD have been developed (59).

 

Low GH levels to provocation tests frequently occur in the immediate peripubertal period. Given the known action of the sex steroids to augment endogenous GH secretion this has led some pediatric endocrinologists to prime children of peripubertal age but without clinical signs of puberty undergoing GH stimulation testing with exogenous sex steroids (diethylstilbestrol, ethinylestradiol and testosterone can be used). Around 50% of pediatric endocrinologists routinely use priming for GH stimulation tests(60). Some endocrinologists will prime boys >9 years and girls >8 years others will prime only those with a delayed puberty >13-14 years in boys and > 11 or 12 years in girls. In one study by Marin et al(61) where 61% of healthy prepubertal children failed to demonstrate a peak GH >7µg/L to three GH provocative tests (exercise, insulin and arginine) but after administration of estrogen 95% of these children demonstrated a peak GH >7 µg/L. Multiple other studies have confirmed this result in healthy peripubertal children with growth impairment (62).  Thus, the argument in favor of priming is that it prevents false positive diagnoses of GHD in this group. The concerns about priming are that it only briefly augments the GH response which then returns to suboptimal levels which may be insufficient for normal growth. Thus priming may result in failure to treat children with transient peripubertal GH deficiency who would have benefitted from treatment (62).

 

24 hour or overnight 12-hour GH profiles with measurement of serum GH every 20 minutes have been proposed as an alternative assessment of GH secretion. The obvious disadvantages are the large number of samples required and costs, particularly of the overnight hospital admission. While a 24 hour GH profile has a high reproducibility there is also a large degree of inter individual variability limiting the usefulness of the procedure as a diagnostic test (63).

 

A diagnosis of GH neurosecretory dysfunction can be made where the patient presents with signs/symptoms of GHD with low IGF-I concentration, a normal peak GH level to pharmacological stimulation but absence of spontaneous GH peaks on 24 hour serum GH profile (64). This diagnosis has not been identified in adults and given the interindividual variability in 24-hour GH profiles caution should be made before coming to GH neurosecretory dysfunction as a diagnosis, particularly where there is no history of cranial irradiation.

 

Measurement of IGF-I and/or IGFBP-3

 

IGF-I and IGFBP-3 are, unlike GH, present at relatively constant concentrations in serum throughout the day and can therefore be measured by a simple blood test without the need for pharmacological stimulation. IGF-I is suppressed in states of poor nutrition and both IGF-I and IGFBP-3 concentrations vary with age and pubertal stage, thus normative ranges taking into account age, Tanner stage, and BMI have been recommended (52). The majority of IGF-I exists bound in the ternary IGF-I/IGFBP-3/ALS complex (thus free IGF-I is very low and difficult to measure) and assays therefore require a step to remove the IGF binding proteins before measurement of total IGF-I. Incomplete removal of IGF-I can potentially lead to false low IGF-I concentrations. Both IGF-I and IGFBP-3 have a low sensitivity (~50%) with a high specificity (97%) (65,66) and thus are of limited value in isolation. They do, however, form a vital component of the assessment of a child for GHD combined with auxological, other biochemical and radiological data.

 

Neuroimaging

 

Identifying abnormalities of the hypothalamo-pituitary axis provides powerful evidence for the diagnosis of GH deficiency in the short child. The most common abnormality identified in congenital GHD is the so-called pituitary stalk interruption syndrome consisting of a variable combination of anterior pituitary hypoplasia, ectopic posterior pituitary, and thinning or interruption of the pituitary stalk (67). Loss of the vascular pituitary stalk increases the risk of MPHD 27-fold but required gadolinium-DTPA administration to reliably distinguish presence/absence of vascular stalk (68). Other potential findings in congenital GHD include

 

  1. Septo-optic dysplasia – combination of absence of septum pellucidium, optic nerve hypoplasia and hypopituitarism. May be associated with an ectopic posterior pituitary and anterior pituitary hypoplasia.
  2. Abnormalities of the corpus callosum – agenesis, corpus callosum cysts
  3. Holoprosencephaly
  4. Eye abnormalities – microphthalmia or anophthalmia (GLI2 or OTX2 mutations)
  5. Absent olfactory bulbs (FGFR1, FRF8 and PROKR2 mutations)
  6. Pituitary hyperplasia (seen in patients with PROP1 mutations)
  7. Hypothalamic hamartoma (Pallister-Hall syndrome)
  8. Empty sella
  9. Absence of the internal carotid artery
  10. Arnold-Chiari malformations
  11. Arachnoid cysts
  12. Syringomyelia

 

In acquired GHD tumors affecting the hypothalamo-pituitary axis will frequently be identified – craniopharyngiomas, adenomas, and germinomas. Thickening of the pituitary stalk may be identified in Langerhans cell histiocytosis.

 

As well as a role in the diagnosis of GH deficiency MR imaging can also help predict which patients will require re-testing of growth hormone status at the end of growth. Young adults with MRI abnormalities have an increased risk of persisting GHD into adulthood (69).

 

GH Therapy

 

All children diagnosed with GH deficiency should be treated with recombinant human growth hormone as soon as possible after the diagnosis is made. The aim of treatment is to normalize height – both to within the normal range for the population and to achieve a height within the child’s target range. GH is administered as a once daily subcutaneous injection in the evening. Starting dose is usually in the range of 25-35µg/kg/day with maximum dose being 50µg/kg/day. In children with more severe GHD (evidence by a lower peak GH level, more severe presentation, MRI abnormality) the response to GH is better and often height can be normalized with lower doses of GH e.g., 17-35 µg/kg/day (70). Prediction models (discussed below) are available and in GHD have been shown to reduce variability in response but do not improve height gain (71). Children receiving GH therapy should be seen every 3-6 months and the GH dose titrated to height velocity and height gain. Monitoring of IGF-I concentrations is recommended to avoid prolonged periods of supraphysiological IGF-I levels. In general, IGF-I should be measured at least annually but can be measured more frequently particularly where there has been a recent increase in dose. A reduction in dose would normally be considered were two consecutive IGF-I levels were above +2 SD. As a guide to dose adjustment a 20% alteration in dose leads, on average, to a 1 SD change in IGF-I concentration (72).  Treatment is continued until the child is post-pubertal and growth is either completely ceased or is <2cm per year.  A growth chart from a child with congenital GHD treated with recombinant human GH therapy is shown in Figure 5.

 

Currently there is no single accepted definition of poor response to GH treatment with suggestions including change in height SDS <0.3 or 0.5 during the first year of treatment, change in height velocity <+3cm/year during 1st year of treatment, change in height velocity <+1SD or a height velocity <-1 SD during the first year of therapy.  Depending on the definition used 20-35% of patients display a poor response (73). It is important to discuss the possibility of a poor response with the family prior to staring therapy.

Figure 5. Growth Chart from child with GH deficiency. GH therapy is started at age 4 with height SDS -3.7 SD. There is a sustained improvement in height velocity leading to a final height of +1.5 SD.

Multiple long-acting preparations of growth hormone are at various stages of development (74). A phase three trial in adults with GHD have been completed and has demonstrated similar efficacy with a once weekly injection of a long-acting GH compared to conventional daily GH (75). Trials in children are currently ongoing.

 

Prediction of Response to GH Therapy and the d3-Growth Hormone Receptor Polymorphism

 

Initial work predicting the response to GH therapy was based on auxological and biochemical data, particularly from the Kabi International Growth Study (KIGS), a large surveillance study of over 62,000 patients treated with GH in childhood. Prediction models developed included models for idiopathic isolated GH deficiency (76) and early onset isolated GH deficiency (77).  For the idiopathic isolated GH deficiency prediction model the model explained 61% of the variability on GH response. Factors included in the prediction model were peak GH during stimulation test, age at start of GH therapy, height SDS minus mean parental height SDS, growth hormone dose and weight SDS. Other prediction models derived from alternative datasets have also been produced for GHD (78,79).

 

Around 50% of the European population are homo- or heterozygous for a polymorphism of the GHR that leads to deletion of exon 3 and 22 amino acid residues near the N-terminal. In 2004 it was reported that GH signaling via the GHR with the d3 was increased and that children treated with GH under the SGA license or with idiopathic short stature showed an increased first year growth velocity where they were homo- or heterozygous for the d3 polymorphism (80). Since this original report there have been many studies assessing the effect on the d3 polymorphism on response to GH therapy in GH deficiency, Turner syndrome, SGA children and in children with idiopathic short stature. A meta-analysis of these studies in 2011 indicated that, compared to children homozygous for the full-length allele, children homozygous for the d3 polymorphism have an increase in 1st year height velocity SDS of 0.14 SD and children heterozygous for the d3 polymorphism has an increase of 0.09 SD (81).  Thus, it appears that the d3 polymorphism has a modest effect mediating the response to GH therapy.

 

The PREDICT study was a large international observational study which assessed the contribution of single nucleotide polymorphisms in over 100 candidate genes to GH response in a cohort of children with GH deficiency or Turner syndrome (82,83). GH response was assessed by change in IGF-I concentrations over 1 month and by height velocity change over the first year of treatment. Carriage of 10 polymorphisms within 7 different genes, related in particular to cell signaling, were identified to be associated with change in IGF-I over the first month of GH treatment and height velocity over the first year of treatment. In addition to assessing association between genotype and response to GH therapy the PREDICT study also assessed the use of basal gene expression in peripheral blood mononuclear cells to predict GH response. There were 1188 genes where the expression level was associated with low response and 865 genes where expression level was associated with a high response to GH therapy (83). Network analysis of the human interactome associated with these genes indicated that glucocorticoid, estrogen, and insulin receptor signaling, and protein ubiquitination pathways were most represented by the genes where association was linked to high or low response to GH therapy.

 

A recent genome wide association study examining GH responsiveness did not identify any significant SNPs in their primary analysis (the primary analysis utilized all diagnostic groups for GH treatment together) (84).  They did identify 4 SNPs in a secondary analysis stratifying by diagnosis and limiting to European ancestry – the closest associate genes are UBE4B, LAPTM4B, COL1A1/NT5DC1 and CLEC7A/OLR1(84).

 

INHERITED DISORDERS OF THE GH-IGF-I AXIS

 

Genetic Disorders Causing Isolated Growth Hormone Deficiency

 

Initial reports suggested that only around 12% of cases of isolated growth hormone deficiency were associated with abnormalities of the hypothalamus or pituitary on MR imaging (85). More recent studies have indicated that up to 26% of cases of isolated GHD are associated with MR abnormalities (86), particularly anterior pituitary hypoplasia and ectopic posterior pituitary.  Within the remaining cohort of patients with IGHD an increasing number of genetic causes have been identified.

 

IGHD TYPE 1

 

IGHD type 1a is inherited in an autosomal recessive manner and is due to homozygous deletions and nonsense mutations in the GH1 gene leading to a complete absence of the GH protein from serum. The clinical presentation is with severe growth hormone deficiency and growth failure from 6 months of life with height SDS >4.5 SD below mean. Typically patients respond well to initial therapy with GH but then develop anti-GH antibodies leading to a loss of efficacy (87). Treatment with IGF-I is an option for such patients.

 

IGHD type 1b is also autosomal recessive and caused by mutations in the GH1 gene – either mis-sense, splice site or nonsense or by mutations within the GHRHR (the gene encoding the GHRH receptor). The clinical phenotype in IGHD type 1b is milder than that of IGHD 1a with the presence of low but detectable levels of GH to stimulation tests. These patients show a good response to treatment with GH without the development of anti-GH antibodies.

 

The GHRHR is a 423 amino acid G-coupled protein receptor. It contains seven transmembrane domains encoded for by a 13-exon gene on chromosome 7p15. While human mutations leading to isolated GH deficiency have been found in the GHRHR gene, to date no such mutations have been identified in the gene encoding the ligand, GHRH. The initial link between a GHRHR mutation and impaired growth was in the little mouse, where Lin et al identified an amino acid substitution in codon 60 of the mouse GHRHR (88). The substitution of glycine for aspartic acid (D60G) prevented the binding of GHRH to the mutant receptor. Subsequent to the identification of the mutation in mouse a nonsense mutation (p.E72X) was identified in two patients in a consanguineous family of Indian ethnic origin (89). Since this initial report multiple families have been reported and splice site mutations, missense mutations, nonsense mutations, microdeletions and one mutation in the promoter (90). The clinical phenotype of an individual with a GHRHR mutation is that of autosomal recessive inheritance of IGHD, anterior pituitary hypoplasia (defined as pituitary height more than 2 SD below mean), GH concentrations are either undetectable or very low in response to provocation tests and IGF-I/IGFBP-3 levels are low. In contrast to patients with GH1 mutations midface hypoplasia, neonatal hypoglycemia and microphallus are less common. Intelligence is normal and affected individuals are fertile.

 

Expression of GHRHR is upregulated by the pituitary transcription factor POU1F1 and this results in somatotroph hypertrophy. Because of this effect on somatotrophs anterior pituitary hypoplasia is commonly seen on MR imaging but there have been reports of GHRHR mutations with normal pituitary morphology (91).

 

IGHD TYPE 2

 

IGHD Type 2 is an autosomal dominant disorder caused by mutations in the GH1 gene.  The severity of GH deficiency is highly variable. While the name of the condition suggests only GH is affected, in practice loss of other pituitary hormones has been reported and patient must be followed up to identify these additional hormone deficiencies. Loss of TSH, ACTH, prolactin and gonadotrophins have all been reported (92).

 

IGHD type 2 is most commonly caused by mutations that affect splicing of GH1, particularly splicing of exon 3 (93).  The most frequent mutations are within the first six bp of the exon 3 donor splice site (93) but mutations in the exon 3 splice enhancers and intron splice enhancers have also been reported (90). The exon 3 splice mutations lead to the exclusion of exon 3 and the production of a 17.5kDa isoform of GH lacking amino acids 31-71, responsible for connecting helix 1 and helix 2 of the mature GH molecule. This abnormal 17.5 kDa variant GH is retained within the endoplasmic reticulum, disrupts the Golgi apparatus and reduces the stability of the 22kDa GH isoform (94). In addition to GH trafficking of other hormones including ACTH is disrupted. A mouse model overexpressing the 17.5kDa isoform demonstrated anterior pituitary hypoplasia with invasion by activated macrophages. The loss of additional pituitary hormones is likely to result from the disrupted hormone trafficking as well as the pituitary inflammation and destruction. Children with IGHD type II may display anterior pituitary hypoplasia on MR imaging. Currently there is no specific treatment in man to ameliorate the effects of the 17.5kDa isoform. A small interfering RNA based therapy has been successful in the mouse model of IGHD type 2 (95) but the delivery system used involved inserting the short hairpin RNA as a transgene. Successful implementation of such a therapy in humans will require an alternative mode of delivery capable of crossing the blood-brain barrier. As well as the classical exon 3 splice site mutations IGHD type 2 is also caused by missense mutations. These have been reported to lead to impaired GH release (96) or to alter folding of GH (97).  

 

IGHD TYPE 3

 

IGHD Type 3 is of x-linked recessive inheritance and the males described were both immunoglobulin and GH deficient. A single patient has been reported with a mutation in the BTK gene (resulting in exon skipping) with x-linked agammaglobulinemia and GH deficiency (98).

 

One family has been reported with isolated GHD caused by mutations in RNPC3 (99). The three affected sisters had compound heterozygous mutations in RNPC3 (p.P474T and p.R502X) and presented with classical severe isolated GHD with anterior pituitary hypoplasia on MR imaging. RNPC3 encodes a component of the minor spliceosome responsible for splicing of a small subset (<0.5%) of introns which are present in ~3% of human genes. Given that splicing is an essential basic process present in all tissues it is interesting that the phenotype seen is pituitary specific.  The patients displayed relatively minor perturbations in splicing which is hypothesized to be tolerated in most tissues, but not in the developing pituitary. Response to GH treatment is reported to be excellent (100).

 

Genetic Disorders Leading to Abnormal Pituitary Development and Multiple Pituitary Hormone Deficiency

 

Mutations in an increasing number of genes lead to loss of multiple pituitary hormones including growth hormone (summarized in Table 2).  A brief summary of each is given below – for an extensive review of pituitary development and it’s genetic control see Bancalari et al (101).

 

HESX1

 

The paired homeobox domain protein HESX1 is one of the earliest specific markers of the pituitary primordium and it acts as a transcriptional repressor. Mutations in HESX1 are associated with septo-optic dysplasia (102) and MPHD (103,104) which can be inherited in an autosomal recessive or autosomal dominant pattern. In addition to the MRI appearances associated with septo-optic dysplasia patients with HESX1 mutations can have an ectopic posterior pituitary (104).

 

OTX2

 

The OTX2 homeobox gene is a homologue of the Drosophila orthodenticle protein. It is expressed early in gastrulation and is involved in development of the central nervous system and eye. In humans OTX2 mutations have been identified in patients with anophthalmia or microphthalmia with isolated GHD or MPHD (105). On MR imaging an ectopic posterior pituitary and small anterior pituitary have been associated with OTX2 mutations. 

 

SOX3

 

SOX3 is a single exon gene located on the X chromosome, is expressed widely throughout the ventral diencephalon and is involved in the development of Rathke’s pouch (106). In humans SOX3 duplications (107) or polyalanine expansion (108,109) have been associated with X-linked hypopituitarism with or without mental retardation. The pituitary phenotype is variable from isolated GHD to MPHD. MRI findings may include anterior pituitary hypoplasia, ectopic posterior pituitary, and corpus callosum abnormalities.

 

PITX2

 

PITX2 is a homeodomain transcription factor expressed in the rostral brain and oral ectoderm during development and throughout the anterior pituitary in adult life. Axenfeld-Riegler syndrome is an autosomal dominant disorder characterized by ocular, dental and craniofacial abnormalities in addition to pituitary abnormalities. Mutations in PITX2 have been found in patients with Axenfeld-Riegler syndrome and GH deficiency (110).

 

LHX3 and LHX4

 

LHX3 and LHX4 encode LIM domain proteins expressed in Rathke’s pouch involved in transcriptional regulation. Homozygous loss of function mutations in LHX3 have been associated with hypopituitarism, sensorineural deafness and cervical abnormalities (rigid cervical spine and cervical spina bifida occulta) (111,112). The MRI appearance may be of a small or enlarged pituitary or a hypointense lesion compatible with a microadenoma.  Mutations in LHX4 lead to a range of pituitary dysfunction from GHD to MPHD (113) with a pituitary phenotype including anterior pituitary hypoplasia, ectopic posterior pituitary and in one family there was pointed cerebellar tonsils suggestive of an Arnold Chiari Malformation (114).

 

GLI2

 

GLI2 is a mediator of Sonic Hedgehog signal transduction and is expressed in the oral ectoderm and ventral diencephalon. Heterozygous mutations in GLI2 lead to a variable combination of holoprosencephaly and hypopituitarism (115,116). Other clinical findings may include a cleft lip/palate, postaxial polydactyly and anophthalmia.

 

FGFR1, FGF8 and PROKR2

 

FGFR1, FGF8 and PROKR2 were previously known to be involved in the pathogenesis of Kallmann syndrome (hypogonadotropic hypogonadism with anosmia). Screening of a cohort of 103 patients with hypopituitarism identified mutations in these Kallmann syndrome genes in eight patients (FGFR1 n=3, FGF8 n=1, PROKR2 n=4) (117).  An EPP was identified in one patient with an FGFR1 mutation and a hypoplastic anterior pituitary in one patient with a PROKR2 mutation.

 

PROP1

 

Prophet of Pit-1 (PROP1) is a homeodomain transcription factor with expression limited to the anterior pituitary. It acts as a transcriptional repressor downregulating HESX1 and as an activator of POU1F1. PROP1 mutations are associated with GH, prolactin, TSH and LH/FSH deficiency with rare cases of ACTH deficiency. PROP1 mutations are the commonest genetic cause of hereditary MPHD accounting for ~50% of familial cases (117).  MRI findings include both small and large anterior pituitary glands and even extension of the pituitary to form a large suprasellar mass which waxes and wanes before involuting (118).  Gonadotrophin deficiency in patients with PROP1 mutations is highly variable and can present with micropenis and cryptorchidism to delayed pubertal onset potentially indicating a role of PROP1 in maintenance of gonadotrophin function.

 

POU1F1

 

The first genetic cause of multiple pituitary hormone deficiency, identified in 1992, was mutations in the POU1F1transcription factor (119).  It is essential for the development of somatotrophs, lactotrophs, and thyrotrophs, consequently mutations in POU1F1 lead to deficiency of GH, TSH and prolactin. Anterior pituitary size is most often small but can be normal with normal stalk and normally sited posterior pituitary. The hormone deficiencies can present at any time from birth to adolescence.

 

IGSF1

 

Mutations in IGSF1 (immunoglobulin superfamily member 1) were identified initially as a cause of central hypothyroidism and macro-orchidism (120). IGSF1 is a membrane glycoprotein expressed in Rathke’s pouch. The identified mutations lead to aberrant protein trafficking and protein mislocalisation.  In a small number of subjects mild or transient GHD has been identified (121,122). It is clear that the immunoglobulin superfamily of proteins may have a wider role in controlling pituitary hormone secretion with mutations in immunoglobulin superfamily member 10 associated with constitutional delay in growth and puberty (123).

 

ARNT2

 

A single family with a homozygous frameshift loss of function mutation in ARNT2 has been described. The affected individuals demonstrated multiple pituitary hormone deficiency including diabetes insipidus along with post-natal microcephaly, frontal and temporal lobe hypoplasia, seizures, developmental delay, visual impairment and congenital abnormalities of the urinary tract (124). ARNT2 is a HLH transcription factor which is known to dimerize with SIM1, a known regulator of neuronal differentiation.

 

TCF7L1

 

Transcription factor 7-like 1 is a regulator of WNT/β-catenin signaling and is expressed in the developing forebrain and pituitary. Two patients with heterozygous missense variants have been reported – one diagnosed with GHD and one with low IGF-I concentrations (124). MRI findings are listed in Table 2. In both families there were unaffected family members also carrying the variant. Given functional studies confirmed the deleterious nature of the variant this is likely to represent autosomal dominant inheritance with variable penetrance.

 

RAX

 

RAX encodes a transcription factor involved in eye and forebrain development. A child with a homozygous frameshift truncating mutation in RAX has been identified with a phenotype including anophthalmia, bilateral cleft lip and palate with congenital hypopituitarism (125).

 

LAMB2

 

Laminin b2 is a basement membrane protein with autosomal recessive mutations associated with congenital nephrotic syndrome, ocular abnormalities and developmental delay. One patient has been reported with isolated growth hormone deficiency, optic nerve hypoplasia, and a small anterior pituitary in association with focal segmental glomerulosclerosis with a compound heterozygous missense mutation in LAMB2 (126).

 

TBC1D32

 

TBC1 Domain Family member 32 is thought to be a ciliary protein and a cause of oral facial digital syndrome type IX (127). Two families with biallelic mutations in TBC1D32 and hypopituitarism have been reported (128). For the first family there were two affected siblings and they had panhypopituitarism with an absent anterior pituitary, ectopic posterior pituitary and retinal dystrophy while in a third family the affected proband had anterior pituitary hypoplasia, growth hormone deficiency and developmental delay (128). Facial dysmorphism was present with prominent forehead, low set posteriorly rotated ears, hypertelorism and a flat nasal bridge.  Autosomal recessive mutations in another ciliopathy related gene IFT172 have been reported to cause GHD with an ectopic posterior pituitary (129).

 

MAGEL2 and L1CAM

 

MAGEL2 and L1CAM mutations have been identified in patients with a combination of hypopituitarism and arthrogryposis (130). MAGEL2 mutations cause Schaaf-Yang syndrome which is similar to Prader-Willi Syndrome with hypotonic, obesity, developmental delay, contractures and dysmorphism.  GHD, diabetes insipidus and ACTH deficiency have been reported in 4 patients. In one patient with L1 syndrome due to a L1CAM mutation arthrogryposis was present with GHD.

 

EIF2S3

 

EIF2S3 encodes a protein involved in the initiation of protein synthesis with mutations associated with developmental delay and microcephaly. In three patients’ mutations in EIF2S3 have been associated with GHD and central hypothyroidism (131). Inheritance is X-linked.

 

FOXA2

 

FOXA2 is a transcription factor involved in pituitary and pancreatic B-cell development and de novo heterozygous mutations cause a phenotype of congenital hypopituitarism with congenital hyperinsulinism (132).

 

OTHER MUTATIONS

 

In addition to the above mutations in CDON (133) (nonsense heterozygous), GPR161(134) (homozygous missense) and ROBO1(135) (heterozygous frameshift, nonsense and missense) have been associated with pituitary stalk interruption syndrome.

 

Table 2. Genetic Defects of Pituitary Development and their Phenotype

Gene

Pituitary Deficiencies

MRI phenotype

Inheritance

Other phenotypic features

ARNT2

 

GH, TSH, ACTH, LH, FSH, ADH

Absent PP, ectopic PP, thin stalk, thin corpus callosum, delayed myelination

AR

Hip dysplasia, hydronephrosis, vesico-ureteric reflux, neuropathic bladder, microcephaly, prominent forehead, deep set eyes, retrognathia

CDON

GH, TSH, ACTH

Small anterior pituitary, ectopic posterior pituitary, absent stalk

AD

 

EIF2S3

GH, TSH

Small anterior pituitary, white matter loss,

X-linked recessive

Developmental delay and microcephaly, glucose dysregulation (hyperinsulinemia hypoglycemia and post-prandial hyperglycemia)

GPR161

GH, TSH, ADH

Small anterior pituitary, ectopic posterior pituitary

AR

Congenital ptosis, alopecia, syndactyly, nail hypoplasia

FGFR1

GH, TSH, LH, FSH and ACTH

Normal or small anterior pituitary, corpus callosum agenesis

AD

ASD and VSD, brachydactyly, brachycephaly, preauricular skin tags, ocular abnormalities, seizures

FGF8

GH, TSH, ACTH, ADH

Absent corpus callosum, optic nerve hypoplasia

AD or AR

Holoprosencephaly, Moebius syndrome, craniofacial defects, high arched palate, maxillary hypoplasia, microcephaly, spastic diplegia

FOXA2

GH, TSH, ACTH

Small shallow sella turcica, anterior pituitary hypoplasia, absent stalk

AR

Congenital hyperinsulinism

GLI2

GH, TSH and ACTH with variable gonadotrophin deficiency

Anterior pituitary hypoplasia

AD

Holoprosencephaly, cleft lip and palate, anophthalmia, postaxial polydactyly, imperforate anus, laryngeal cleft, renal agenesis

GLI3

GH, TSH, LH, FSH, ACTH

Anterior pituitary hypoplasia

AD

Pallister-Hall syndrome Postaxial polydactyly, hamartoblastoma

HESX1

Isolated GHD through to panhypopituitarism with TSH, LH, FSH, ACTH, prolactin and ADH deficiency

Optic nerve hypoplasia, absence of the septum pellucidum, ectopic posterior pituitary, anterior pituitary hypoplasia

AR and AD

Developmental delay

IFT172

GHD

Ectopic posterior pituitary, anterior pituitary hypoplasia

AR

Retinopathy, metaphyseal dysplasia, and hypertension with renal failure

IGSF1

 

GH (transient/partial), TSH, prolactin

Normal in the majority of cases.  Frontoparietal hygroma, hypoplasia of the corpus callosum, and small stalk lesion reported.

X-linked recessive

Macro-orchidism, delay in puberty

L1CAM

GHD

Generalized white matter loss and thin corpus callosum

X-linked recessive

Arthrogryposis, hydrocephalus, VSD, developmental delay, scoliosis, astigmatism

LAMB2

GHD

Small anterior pituitary, optic nerve hypoplasia

AR

Congenital nephrotic syndrome, focal segmental glomerulosclerosis, developmental delay

LHX3

GH, TSH, LH, FSH, prolactin

Small, normal or enlarged anterior pituitary

AR

Short neck with limited rotation

LHX4

GH, TSH and ACTH deficiency

Small anterior pituitary, ectopic posterior pituitary, cerebellar abnormalities, corpus callosum hypoplasia

AD

 

MAGEL2

GHD, ACTH, ADH

Small posterior pituitary, thin corpus callosum and optic nerve hypoplasia

Heterozygous mutations on paternal allele

hypotonia, obesity, developmental delay, contractures and dysmorphism

OTX2

GH, TSH, LH, FSH and ACTH

Normal or small AP, pituitary stalk agenesis, ectopic posterior pituitary, Chiari I malformation

AR or AD

Microcephaly, bilateral anophthalmia, developmental delay, cleft palate

POU1F1

GH, TSH, prolactin

Small or normally sized anterior pituitary

AR and AD

 

PROKR2

GH, TSH, ACTH

Hypoplastic corpus callosum, normal or small anterior pituitary

AD

Club foot, syrinx spinal cord, microcephaly, epilepsy

PROP1

GH, TSH, LH, FSH, prolactin, evolving ACTH deficiency

Small, normal or enlarged anterior pituitary – may evolve over time

AR

 

RAX

GH, TSH, LH, FSH, ACTH, ADH

Absent sella turcica and pituitary

AR

Anophthalmia, bilateral cleft lip and palate

ROBO1

GH, TSH

Small or absent anterior pituitary, ectopic or absent posterior pituitary, interrupted or absent stalk

AD

Strabismus, ptosis

SOX3

GH, TSH, LH, FSH, ACTH.  Most commonly isolated GHD

Anterior pituitary and infundibular hypoplasia, ectopic posterior pituitary, corpus callosum abnormalities including cysts

X-linked recessive

Learning difficulties

SOX2

LH, FSH variable GH deficiency

Anterior pituitary hypoplasia, optic nerve hypoplasia, septo-optic dysplasia, hypothalamic hamartoma

AR

Microphthalmia, anophthalmia, micropenis, sensorineural deafness, gastro-intestinal tract defects.

TBC1D32

Isolated GHD to panhypopituitarism

Absent or hypoplastic anterior pituitary, ectopic posterior pituitary

AR

Retinal dystrophy, developmental delay, facial dysmorphism (prominent forehead, low set posteriorly rotated ears, hypertelorism and a flat nasal bridge). 

TCFL7

 

GH

Absent posterior pituitary, anterior pituitary hypoplasia, optic nerve hypoplasia, parital agenesis of corpus callosum, thin anterior commissure

AD

 

 

Bioinactive GH

 

Short stature associated with normal to high levels of growth hormone with low serum IGF-I concentrations “bioinactive GH” was first described in 1978 (136). This disorder is associated with a good clinical response to GH therapy and multiple subsequent cases have been reported in the literature (90). These multiple case reports contained no information on the genetic cause of the disorder.  The first demonstration of the mechanism responsible for bioinactive GH came in 1997 (137) when Takashi and co-workers described a heterozygous glycine to aspartic acid substitution at amino acid 112 of the GH molecule resulting in impaired binding of the mutant GH to GHR. Reported mutations such as the R77C mutation (138,139) have also been found in normally statured relatives and functional work has failed to identify any difference between wild type and R77C GH on GHR binding, activation of the JAK/STAT pathway, secretion studies or ability to induce cell proliferation (140,141). The clinical scenario of normal to high GH concentrations with low IGF-I levels is not uncommon and a diagnosis of bioinactive GH should not be made unless a mutation is identified where there is a demonstration that the function of the variant GH is impaired.

 

A homozygous missense mutation (C53S) in the GH1 gene was reported in a Serbian patient with height SDS of -3.6 at 9 years of age (142). Altered affinity for the GH receptor was demonstrated in functional studies, presumably due to alteration of the disulphide bond between Cys-53 and Cys-65 in the GH molecule.

 

Laron Syndrome

 

Laron syndrome, caused by loss of function mutations in the GHR gene(143), was first described in 1966 (144). Since then more than 250 patients have been described in the literature with over 70 missense, nonsense, indels and splice mutations within the GHR gene (145). The majority of mutations describe are inherited in an autosomal recessive manner but autosomal dominant inheritance has been described in a small number of cases (146).  Patients present with severe short stature having been born with normal birth size. The facial phenotype is similar to severe GH deficiency with frontal bossing and midface hypoplasia. Intellect, development and head circumference are normal. IGF-I, IGFBP-3 and ALS concentrations are low in serum with normal to raised baseline GH levels with raised peak stimulated GH level. Typical adult height is around -5 SD. Measurement of GHBP in serum is useful as, when markedly low, indicates absence of the extracellular component of the GHR. Since mutations can occur in the transmembrane or intracellular domains, the presence of GHBP in serum does not exclude a diagnosis of Laron syndrome.  The standard diagnostic test is an IGF-I generation test. Specificity of this test is around 77-91% and when applied to a population with low prevalence of GH insensitivity the positive predictive value of the test is likely to be low (147). In addition, there is a limited normative data for the IGF-I generation test. Buckway at al reported the results of IGF-I generation tests in normal subjects and subjects with GH deficiency, Laron syndrome and idiopathic short stature (148). Sensitivity of the IGF-I generation test in this population (who all had the same E180 splice mutation in the GHR, was 77% (the cut off for a normal result on this test was an increase in IGF-I to >15ng/mL post-GH stimulation (149)). Diagnosis of Laron syndrome therefore relies upon integration of clinical and biochemical findings and selecting patients for further genetic studies.

 

Recombinant human IGF-I therapy provides limited benefit in improving height. In an observational study containing 28 patients with Laron syndrome the results of treatment with 120 mg/kg/day IGF-I for a mean duration of 5 years increased height SDS from -6.1 SD to -5.1 SD (150). In the first year of treatment there was a marked increase in height velocity from 2.8 to 8.7 cm but height velocity markedly decreased after the first year of treatment. In a separate report of 21 individuals with GH insensitivity – 5 of whom had Laron syndrome there was an increase in height SDS from baseline of +1.9 SD with treatment of 120 mcg/kg/day IGF-I for a mean of 10.5 years (151). The treatment effect is markedly lower than that of GH in children with severe congenital GH deficiency (an example of a growth chart of a child with Laron syndrome treated with IGF-I is given in Figure 6). While GH therapy stimulates both hepatic and local IGF-I production, subcutaneous injections of IGF-I do not simulate this local IGF-I production. In addition, GH therapy normalizes not only IGF-I levels but levels of IGFBP-3 and ALS whereas in GH insensitive subjects treated with IGF-I there is no increase in IGFBP-3 or ALS concentrations. Thus, it would be expected that the injected IGF-I would have a much lower half life than endogenous IGF-I. A combined therapy of IGF-I with IGFBP-3 disappointingly was less effective in improving height (152).

Figure 6. Growth chart of girl with Laron syndrome treated with recombinant human IGF-I (Increlex) from age of 5.8 years when height SDS was -4.2 SD. There is an increase in height velocity over the first year of treatment which is reduced in subsequent years of therapy. Height SDS improves to -2.1 SD by 10.25 years but this has been associated with the onset of puberty at 9 years (treatment with the GnRH analogue Zoladex was introduced at 9.8 years). Current height lies within parental target range. M denotes maternal height and F denotes adjusted paternal height.

STAT5b Mutations

 

The signal transducers and activators of transcription (STAT) family contains seven proteins (STAT1, -2, -3, -4, -5a, -5b and -6). Mutations in STAT1(153) and STAT3 are associated with immune deficiency and a mutation in STAT5b was described in a patient with growth hormone insensitivity and immune deficiency (154).  The initial report was of a homozygous missense mutation in exon 15, encoding the critical SH2 domain leading to aberrant folding and aggregation of the protein. Six other mutations have been described including a nonsense mutation in exon 5 (155), two distinct nucleotide insertions (156,157) in exons 9 and 10 containing the DNA binding domain, a missense variant within the SH2 domain (158), a four nucleotide deletion in exon 5 (159) and a single nucleotide deletion in the Linker domain (160).

 

Until recently all the mutations identified were homozygous and the disorder is predominantly inherited in an autosomal recessive manner but dominant negative mutations have now been reported (161).  There is some evidence of a mild effect of the heterozygous state as height SDS in parents of affected children is consistently below mean height for the population with range from -0.3 SD to -2.8 SD. Birth weight appears to be within normal limits but postnatal height is severely impaired with height SDS range of -3 to -9.9 (158). Growth is comparable to children with Laron syndrome. Bone age and puberty is commonly delayed perhaps reflecting in part the chronic state of ill health. A prominent forehead, depressed nasal bridge and high-pitched voice are seen in some patients. The biochemical findings are compatible with growth hormone insensitivity with normal to high basal growth hormone concentrations and a raised stimulated peak GH level. Of note, 1 subject had a low stimulated peak GH concentration of 6.6 mcg/. Serum IGF-I, IGFBP-3 and ALS concentrations were consistently low in all subjects, remaining low at end of an IGF-I stimulation test.

 

Clinical differentiation of patients with STAT5b mutations form those with Laron syndrome can be made with the immunodeficiency. All but one of the reported cases has presented with chronic pulmonary disease, particularly lymphoid interstitial pneumonia, with the other child having severe hemorrhagic varicella. Two patients have died from their lung disease and a further patient has required lung transplantation. Patients with STAT5b mutations also have raised serum prolactin levels which can also be helpful with diagnosis.

 

Acid Labile Subunit Deficiency

 

The human IGFALS gene is located on chromosome 16p13.3 and ALS deficiency is inherited in an autosomal recessive pattern with homozygous and compound heterozygous mutations identified including missense, nonsense, deletions, duplications and insertions. The mutations are spread throughout the IGFALS gene which contains 2 exons and encodes a protein of 605 amino acids (162). The majority of the mutations are located in the 20 central leucine rich domains.  The clinical phenotype, first described in 2004 (163), is of very low serum concentrations of IGF-I, IGFBP-3 and ALS with a moderate degree of short stature (-2 to -3SD). 

 

Limited data is available on size at birth but weight appears to be within the lower half of the normal range (-0.2 to -1.9 SD) with only one individual reported to be SGA with a birth weight of -2.2 SD. The data on birth length is even more limited but all individuals measured were within normal range at -1.5 to +1.0 SD. Data on height during childhood is more abundant and hemorrhagic it is clear that postnatal growth is affected in the majority of individuals carrying ALS mutations.  Mean prepubertal height in 17 patients was reported as -2.61 SD (range -3.9 to -1.06 SD) with final adult height of -2.15 SD (range -0.5 to -4.2 SD). There is a preponderance of males in the literature (88% reported cases) which may represent the increased likelihood of males with short stature to present to health care providers. In male’s pubertal onset is commonly delayed (6/11 with onset puberty >14 years and 3/11 onset >15 years).  Serum IGF-I and IGFBP-3 standard deviation scores are very low (-3.3 to -11.2 SD for IGF-I and -3.6 to -18.5 for IGFBP-3), with undetectable ALS concentrations in all but one case (164). Levels of GH are increased with a mean peak GH of 46µg/L.

 

The relatively modest growth impairment in ALS deficiency is likely to be due to the preservation of the local production and action of IGF-I with deficiency of hepatic derived IGF-I. The diagnosis should be suggested by the presence of very low concentrations of IGF-I and, in particular, IGFBP-3 in the presence of moderate growth impairment. Although measurement of ALS is not routinely available this would also be a useful diagnostic tool.

 

Response to treatment with GH therapy has been poor and one child treated with recombinant human IGF-I did not improve height after 1 year of treatment.

 

IGF-I Gene Mutations

 

Deletions and mutations within the IGF1 gene are an extremely rare cause of GH insensitivity. The first patient was reported in 1996 (3) and there have been four subsequent affected families reported (165-168). The first patient described had a homozygous deletion of exons 3 and 5 of the IGF1 gene leading to frameshift and generation of a premature termination codon. He had undetectable levels of serum IGF-I with normal concentrations of IGFBP-3 and ALS with raised baseline and spontaneous GH peak levels. He was born small for gestational age at 1.4 kg at term and displayed profound post-natal growth impairment with sensorineural hearing loss, microcephaly and developmental delay. 

 

One subsequent report identified a similar phenotype of growth impairment, developmental delay, microcephaly and hearing impairment with a homozygous missense variant in exon 6 of IGF-1(167). The patient also had low IGF-I concentrations and high GH levels. Subsequent studies have identified this variant in individuals with normal height and there may be an alternative cause for this child’s growth impairment.

 

There have been two cases reported with similar phenotype of growth impairment, microcephaly and hearing impairment in individuals associated with homozygous mutations within the IGF1 gene (166,168). These mutations (V44M and R36Q) reduce the binding affinity of IGF-I for IGF1R. A large family with short stature and a heterozygous IGF1 mutation (c.402+1G>C) inducing splicing out of exon 4 with subsequent frameshift and truncated peptide (165)has also been reported.  This family included 5 short individuals with the heterozygous IGF1 mutation and an additional 5 individuals who are short but do not have the IGF1 variant. The phenotype of the proband was less severe than other IGF1 mutation patients with normal birth size (3.0kg) but significant post-natal growth impairment (presenting height -4.0 SD), normal hearing, normal development except for attention deficit hyperactivity disorder and mildly reduced serum concentrations of IGF-I (-2.2 SD) with normal IGFBP-3 serum levels (-1.25 SD).

 

For all patients reported to date, treatment with GH has been ineffective. Treatment with recombinant human IGF-1 may be more effective but may be complicated by the development of antibodies in those patients with IGF1deletions. It should however be effective for patients with bioinactive IGF-I.

 

Chromosome 15 Abnormalities and Mutations Affecting the IGF-I Receptor

 

The phenotype of patients with mutations in the IGF1R gene is similar, if slightly milder, to patients with IGF1 gene defects. They are born SGA and continue to grow poorly with microcephaly and variable developmental delay. Reported birth weights are from -1.5 to -3.5 SD with head circumference of -2.0 to -3.2 SD. Birth length SDS is highly variable at -1.0 to -5.0 SD while childhood height ranges from -2.1 to -4.8 SD (169). The initial patient described had a compound heterozygous mutation (170) within IGF1R while all other patients to date have heterozygous mutations. These mutations are dispersed throughout the gene (169). Missense (171,172), nonsense (170), small deletions (173)and duplications (174) have already been identified leading to a variety of deleterious effects on the IGF1R including loss via nonsense mediated decay (174), production of a truncated protein (170), altered trafficking(171), reduced ligand binding (175) and altered tyrosine kinase activity (172). Serum IGF-I concentrations can be normal or raised but are generally > +1 SD.

 

Response to treatment with GH therapy is variable – of 5 patients reported no response was seen in two patients, an equivocal response seen in another two patients and only one patient responded well to therapy (169). GH dose ranged from 0.025 to 0.07 mg/kg/day with the best responder treated with the lowest dose of GH. The rationale behind GH therapy is that it increases hepatic and local production of IGF-I to improve growth. Where there is resistance to IGF-I it is not surprising that GH therapy is less effective. For most disorders clinicians the aim of GH therapy is to improve growth without generating IGF-I concentrations above the normal rage. For IGF1R mutations, given the IGF-I resistance, it may not be possible to achieve adequate growth without using high dose GH therapy with subsequent IGF-I concentrations above the normal reference range.  The long-term effects of such therapy in this patient group are unknown and before embarking on such a strategy a careful discussion about the risks and benefits should be undertaken with the child/parents

 

Prior to the identification of mutations within the IGF1R gene there were reports of patients with abnormalities of chromosome 15 including monosomy, ring chromosome and unbalanced translocations. Allelic loss of chromosome 15 was described to result in growth impairment (176) while trisomy of chromosome 15 results in overgrowth (177), given the location of IGF1R at chromosome 15q26 it was hypothesized that the growth alterations were due to a dosage effect on IGF1R. The clinical phenotype is highly variable depending on the chromosomal aberration e.g., 15q26 deletion is associated with congenital diaphragmatic hernia as well as growth impairment (178). Response to GH therapy appears better for patients with chromosome 15 abnormalities with a first-year increase in height SDS of 0.8-1.5 (179).  

 

Pregnancy-Associated Plasma Protein A2 Deficiency

 

Pregnancy-associated plasma protein A2 is a metalloproteinase responsible for the cleavage of IGFBP-3 and IGFBP-5, an essential step in releasing IGF-I from the ternary complex and allowing it to bind to the IGF1R. Two families have been reported with loss of function mutations in PAPPA2 leading to growth impairment with increased concentrations of IGF-I, ALS, IGFBP-3 and IGFBP-5 and a resultant reduction in free IGF-I (180). GH concentrations are raised due to the reduced free IGF-I. Birth size is moderately reduced in some subjects and the degree of postnatal growth impairment is highly variable ranging from -3.8 SD to -1.0 SD. Other clinical features include mild microcephaly, small chins and long thin fingers. Treatment with rhIGF-I in one family demonstrated an increase in height SDS of +0.4 SD over 1 year of treatment (181) while in the second family treatment was discontinued due to headache in one of two siblings (182).

 

ACQUIRED GH DEFICIENCY

 

Tumors of the Hypothalamus or Pituitary

 

CRANIOPHARYNGIOMA

 

Craniopharyngiomas are non-glial intracranial tumors derived from malformed embryonal tissue thought to originate from ectodermal remnants of Rathke’s pouch or residual embryonal epithelium of the anterior pituitary (183). More than 70% of adamantinomatous craniopharyngiomas contain a mutation of the β-catenin gene (184). Although rare, craniopharyngiomas are the commonest childhood tumor affecting the hypothalamo-pituitary axis accounting for 55-90% of sellar and parasellar lesions in childhood (185). The incidence is 0.5-2 per million per year (186) with 30-50% of cases diagnosed in childhood. In contrast to adulthood where the commonest histological type of craniopharyngioma is papillary, in excess of 70% of childhood craniopharyngiomas are adamantinomatous and associated with cyst formation. Survival rates with craniopharyngiomas are excellent exceeding 90% at 10 years (187)after diagnosis but morbidity with visual defects, hypothalamic obesity, and pituitary hormone deficiency is high.

 

Presentation is with a combination of symptoms of raised intracranial pressure, visual impairment, and endocrine deficits. Up to 87% of cases present with a least one pituitary hormone deficiency – the commonest being GH deficiency present in up to 75% of cases at diagnosis (188). The prevalence of GH deficiency rises after treatment to >90% of patients – with both surgical intervention and radiotherapy implicated in this increase in GH deficiency.  Additional pituitary hormone deficits are common including diabetes insipidus which is present in 92% of cases (189).

 

Therapy for craniopharyngiomas can include a combination of surgery, radiotherapy and intra-lesional chemotherapy. Surgery can be via the transcranial or transsphenoidal route.  Where it is possible to remove the entire tumor without causing damage to the hypothalamus or optic nerves this is the treatment of choice. For larger tumors involving these structures controversy exists on whether the benefits of a complete resection, namely a reduction in the risk of recurrence/progression, are outweighed by the surgical morbidity particularly hypothalamic obesity, visual impairment, and adipsic diabetes insipidus (190,191).  The alternative strategy is a limited surgical resection followed by adjuvant treatment with either conventional radiotherapy or proton beam therapy.  Recurrence rates for complete resection are 15-46% (192), 70-90% for patients treated with surgical partial resection alone and 21% for patients treated with surgical resection and radiotherapy (193).

 

There is good evidence to suggest that replacement GH therapy does not increase the risk of recurrence in craniopharyngioma (194,195) and that the gain in height is similar to that seen in congenital isolated GH deficiency. In one report the mean time between diagnosis and initiation of GH therapy was 2.3 years (194). A period of time after diagnosis, prior to the introduction of GH therapy, allows the completion of surgery and radiotherapy and a period of observation. Despite the reports on the overall safety of GH in craniopharyngioma rapid regrowth of the tumor after the initiation of GH therapy has been reported (196).

 

PITUITARY ADENOMAS

 

Pituitary adenomas are rare in childhood comprising only 3% of supratentorial tumors of childhood (197). Functioning adenomas are more common than non-functioning adenomas with the commonest being prolactinomas, followed by ACTH secreting adenomas then GH secreting adenomas. In one series of 41 patients with childhood onset adenomas, 29 (70%) were prolactinomas, 5 (12%) were ACTH secreting adenomas, one patient (2%) presented with a GH secreting adenoma and the remaining 6 patients (15%) presented with non-functioning adenomas (198).  GH deficiency was present in four out of the 41 patients during childhood and 13 patients during follow up into adulthood. All patients who developed GH deficiency had a macroadenoma. In approximately 5% of cases pituitary adenomas are familial and this is known to be caused by mutations in the genes encoding MENIN (199) and Aryl Hydrocarbon Receptor Interacting Protein (200).

 

OPTIC PATHWAY GLIOMA

 

Optic pathway gliomas are tumors of the pre-cortical visual pathway which may also involve the hypothalamus. In around 1 in 3 cases they are associated with neurofibromatosis type 1 (201). They commonly present with ophthalmological signs and symptoms with the main endocrine presentation being precocious puberty. In the majority of cases there is limited or no progression of the tumor and only monitoring is required. Surgery not recommended for most cases due to the possibility of post-operative visual impairment. Where required initial treatment is with chemotherapy with radiotherapy reserved for teenagers and younger children who have not responded to chemotherapy. Although effective with a 90% 10-year progression free survival radiotherapy is associated with an increased risk of worsening visual impairment, endocrine deficits, cerebrovascular disease, and neurocognitive deficits.

 

GH deficiency in optic pathway gliomas can be present prior to radiotherapy but is much more common post radiotherapy. In one study of 68 children with optic pathway gliomas 19 developed GH deficiency, 15 of whom had received radiotherapy (202). In another study of 21 patients with optic pathway gliomas treated with radiotherapy only one patient had GH deficiency pre-radiotherapy while all patients had GH deficiency post radiotherapy (203).  GH therapy is highly effective and restores adult height to within normal range (204). Optic pathway gliomas can be associated with GH excess, especially in NF1 syndrome related cases.

 

LANGERHANS CELL HISTIOCYTOSIS

 

Langerhans cell histiocytosis (LCH) is a rare disorder with a prevalence of ~4 per million children (205). It is a condition in which there is proliferation and accumulation of clonal dendritic cells (LCH cells) bearing an immunophenotype very close to that of the normal epidermal Langerhans cells of the skin (205). LCH cells can spread to nearly any site in the body, proliferate and lead to local inflammation and tissue destruction. The commonest pituitary hormone deficit in Langerhans cell histiocytosis is diabetes insipidus which develops in around 25% of childhood patients with LCH while GH deficiency is the second commonest endocrinopathy present in 9-12% of childhood LCH patients.

 

Radiation

 

Neuroendocrine abnormalities of the hypothalamo-pituitary axis evolve with time after radiation induced damage. The first, and sometimes only, hormone deficiency following radiation exposure of the HPA axis is growth hormone deficiency. The risk of GH deficiency is related to the total radiation dose, fraction size and time between fractions. Almost all children exposed to >30 Gy cranial irradiation will develop GH deficiency around 65% of those receiving <30 Gy develop GH deficiency by 5 years post radiotherapy (206). Isolated GH deficiency has also been reported in children exposed to 18-24 Gy as used prophylactically in acute lymphoblastic leukemia (207) and in children exposed to as little as 10 Gy as part of total body irradiation (208).  

 

The hypothalamus is thought to be the site of radiation induced damage to the HPA as when exposed to radiation <50 Gy hormone deficiencies remain common ~90% after 10 years (209)  but in contrast delivery of radiation doses 500-1500 Gy to the pituitary alone result in lower rates of endocrinopathy – 40% 14 years after exposure (210).  Additional evidence of the susceptibility of the hypothalamus to radiation induced damage comes from the high prevalence rates of hypothalamic dysfunction on dynamic endocrine tests observed after radiation exposure (211)  and the presence of impaired GH secretion to stimuli acting through the hypothalamus with normal GH secretion to stimuli acting directly on the pituitary (212). Within the hypothalamic-pituitary axis there is differential sensitivity to radiation induced damage with the somatotrophic axis being the most vulnerable to damage, followed by GnRH-FSH/LH and then the CRH-ACTH and TRH-TSH axes which are the least sensitive to radiation induced damage (213).  This sequence of loss of pituitary hormones in radiation induced damage is seen in both animal models (213,214) and in humans where lower doses of radiation (e.g. 18-30 Gy used in treatment of childhood leukemia and brain tumors) leads to isolated GHD(215) whereas higher doses of radiation >60 Gy, used in the treatment of skull base tumors and nasopharyngeal carcinomas, leads to multiple pituitary hormone deficiency (216,217). Risk of pituitary hormone deficiency increases with time elapsed after radiation exposure in addition to the radiation dose – in one study around 50% of children treated with 27-32 Gy for a brain tumor were GHD after one year of treatment, with 85% GHD by 5 years post treatment and almost all GHD by 9 years post treatment (206).

 

GH NEUROSECRETORY DYSFUNCTION

 

One form of GHD particularly well described following radiation injury to the hypothalamic-pituitary axis is GH neurosecretory dysfunction (218-220). Neurosecretory dysfunction is characterized by normal responses to pharmacological stimuli of GH secretion but reduced spontaneous physiological GH secretion.  GH neurosecretory function in seen most frequently with lower radiation doses of <24 Gy (220) and it appears that doses >27 Gy both spontaneous and pharmacologically stimulated GH responses are reduced (221).  The possibility of GH neurosecretory dysfunction makes the diagnosis of GHD in children exposed to cranial irradiation challenging. The presence of normal IGF-I and IGFBP-3 concentrations in many children with radiation induced GH deficiency (222-225) (proven with multiple pharmacological stimulation tests) compounds these difficulties.

 

For children with brain tumors that can exfoliate cells into the cerebrospinal fluid (e.g., ependymoma or medulloblastoma) radiotherapy is delivered to the spine in addition to cranial irradiation. Spinal irradiation has a profound effect on growth and leads to reduced height and disproportionate growth with decreased upper to lower segment ratio (226).   Brauner et al compared children treated with craniospinal irradiation to those receiving cranial irradiation alone with height SDS being significantly lower in the craniospinal group at 1.46 SD compared to the cranial irradiation only group with a height SDS of -0.15 (221). Final height in adults who received craniospinal irradiation is also significantly lower than adults receiving cranial irradiation alone (-2.37 v -1.14 SD) (227). Lower age at radiation exposure is associated with a lower adult height SDS (227) with height loss from spinal irradiation estimated at 9 cm when exposed at 1 year, 7cm when exposed at 5 years and 5.5 cm when exposed at 10 years.

 

Response to treatment with GH therapy is poorer in children with radiation induced GH deficiency than in children with congenital GHD. For most patients with congenital GHD, GH therapy will lead to significant catch-up growth but in patients with radiation induced GH deficiency catch up growth is rare (228,229). However, while GH treatment does not appear to induce catch up growth it prevents a further decline in height SDS (228,230).  The cause of the poorer response seen in patients with radiation induced GH deficiency are likely to be multifactorial including early puberty, delayed GH therapy, use of lower doses of GH and the direct effect of spinal irradiation. GH therapy in children who have previously received craniospinal radiotherapy does prevent further height loss but does so at the expense of further exaggerating the skeletal disproportion seen in these patients.

 

There is extensive evidence linking the GH-IGF-I system to risk of cancer via several sources:

 

  1. Up-regulating the activity of the GH-IGF-I axis in leads to increased development of tumors in animal models (for review see Yaker et al (231)).
  2. In vitro evidence of expression of GH, GHR and IGF-I/II by tumors and the ability GH and IGF-I to induce cancer cell proliferation and metastases (for review see Clayton et al (232))
  3. Epidemiological evidence has linked higher serum IGF-I concentrations to cancer risk (233-236)
  4. Increased risk colorectal and thyroid cancers in patients with acromegaly (a condition of chronic GH excess) (237-239)

 

This evidence did lead to concerns about the risk of administration of GH therapy to patients with GH deficiency and a history of cancer. The majority of studies examining risk of recurrence in children with cancers treated with GH indicate that there is no increased risk of recurrence (240-244).  One notable exception is the Childhood Cancer Survivor Study based in centers in North America where the standardized incidence ratio of second malignancy was elevated (2.1 at average follow up of 18 years) in the 361 individuals treated with GH (245).  The majority of brain tumor recurrences occur during the first two years after completion of primary treatment and this has led to the recommendation that treatment with GH should be considered after this time point. This strategy prevents the association between early tumor recurrence and GH therapy by families but potentially denies children with tumor or radiation induced GH deficiency treatment for a considerable length of time. Children with brain tumors require monitoring of growth and consideration should be given to testing for GH deficiency in children with growth failure who have completed primary treatment. GH therapy should be carefully discussed with the family and oncologist where it is considered before 2 years post primary treatment.

 

Trauma

 

Traumatic brain injury is relatively common in childhood with ~180 children per 100,000 population sustaining a closed head injury each year. The proposed mechanism for traumatic brain injury induced hypopituitarism is that injury to the hypophyseal vessels which transverse the stalk leads to anterior pituitary ischemia and infarction. Postmortem studies of fatal closed head injuries identified hypothalamic lesions suggestive of infarction and ischemia in 43% of cases and pituitary lesions in 28% of cases (246). Although there have been multiple published case reports of anterior pituitary dysfunction in traumatic brain injury for many years (247) there has been a large increase in the number of systematic studies of pituitary function in survivors of traumatic brain injury since 2000. Several moderately sized studies of adult traumatic brain injury survivors have demonstrated risk of post-injury hypopituitarism. Deficiency of GH and gonadotrophins was more common than TSH or ACTH deficiency with 10-28% of patients being GH deficient and 8-30% of patients being gonadotrophin deficient (248-253). In the majority of these studies there has been no relationship between time post injury or injury severity and risk of pituitary dysfunction.

 

Until 2006 the literature on childhood traumatic brain injury and hypopituitarism was limited to case reports (for review of the case reports see Acerini et al (254)). The first report of pituitary function in children with traumatic brain injury studies a cohort of 55 patients (22 studied retrospectively and 30 studied prospectively) and identified 2 patients with low peak GH concentrations (255). Khadr et al reported a 39% rate of abnormalities of pituitary function tests in 33 childhood traumatic brain injury patients (256). None of these were felt to be clinically significant.  In this study 7 patients had a low peak GH concentration but 6 out of the 7 were thought to have peri-pubertal blunting of the GH response with one borderline post-pubertal GHD patient who declined further examination (256).  Poomthavorn et al (257) described a cohort of 54 patients with childhood brain injury 4 of whom had known multiple pituitary hormone deficiency prior to the start of the study, in the 50 patients screened however, there were no patients identified with GH deficiency.

 

The largest study of childhood traumatic brain injury and pituitary function is by Heather et al (258). It examined the pituitary function of 198 survivors of childhood traumatic brain injury. Importantly they used an integrated assessment of GH stimulation tests (including 2nd test with priming where required), auxology and IGF-I concentrations in order to reach a diagnosis of GHD. While a low peak GH concentration (<5µg/L, used as the cut off for diagnosis of GHD in New Zealand at the time of the study) was identified in 16 patients, height SDS ranged from -0.9 SD to +3.6 SD and IGF-I concentrations were within normal limits for all subjects. For this study population had the diagnosis of GHD been based solely on a GH stimulation test and a cut off of 10µg/L for the diagnosis of GHD, 33% of patients would have been incorrectly diagnosed as GHD.

 

The risk of hypopituitarism in childhood traumatic brain injury appears to be low and currently routine screening of pituitary function in this group is not justified outside the context of on-going research studies.

 

Hypophysitis

 

Hypophysitis is characterized by cellular infiltration and inflammation and can be classified as lymphocytic, xanthomatous, granulomatous, necrotizing, IgG4-related and mixed forms.  Lymphocytic hypophysitis is the commonest type but overall the disease is extremely rare with an estimated incidence of 1 per 9 million population (259). Presentation is often with visual disturbance, headache and vomiting. MRI may identify a homogeneous enhancing sellar mass. In adults’ deficiency of TSH and ACTH are particularly common and diabetes insipidus is said to be rare (260). The limited pediatric case reports include several children with diabetes insipidus and it may be that the pattern of hormone insufficiency is influenced by age at presentation.  Hypophysitis is more common in pregnant women but can also occur in non-pregnant women, men and in children (261,262).  Definitive diagnosis is with histopathology while treatment includes hormone replacement therapy and surgery where the sellar mass compresses the optic chiasm. The medical treatment of choice is high dose glucocorticoid therapy but alternative reported therapies include azathioprine (263), methotrexate (264), cyclosporin A (265) and stereotactic radiation (266).

 

GROWTH HORMONE EXCESS

 

While short stature and GHD are common reasons to consult a pediatric endocrinologist, tall stature is a far less common reason to present to a pediatric endocrinologist. Within the group of patients presenting with tall stature in childhood the majority will have either familial tall stature or a genetic/syndromic cause for their tall stature (e.g., Beckwith Wiedemann syndrome, Sotos syndrome, Marfan Syndrome, Simpson-Golabi-Behmel syndrome).  GH excess is an extremely rare disorder in pediatric practice. Causes of GH excess include GH secreting pituitary micro or macroadenomas, ectopic GHRH production and genetic abnormalities affecting GH secretion (McCune Albright syndrome and Carney complex).

 

The commonest symptom of GH excess in childhood is rapid growth. In a series of 15 childhood patients (6 female) with GH secreting adenomas reported by Takumi eta al (267) all the patients presented with rapid growth although 3 also had visual signs/symptoms, 3 amenorrhea, 2 headaches, 1 with hypogonadism and 1 with precocious puberty. Microadenomas were present in 4/15 patients. Acromegalic features such as soft tissue growth of the hands and feet, mandibular overgrowth with prognathism, forehead protrusion and deepening of voice can also occur. The presence of acromegalic features in likely to be linked to the timing of onset (more common with onset in adolescence) and the presence of hypogonadism.  Additional clinical features include excessive sweating, carpal tunnel syndrome, lethargy, arthropathy, impaired glucose tolerance and hypertension. Although rare in childhood, hypertension and glucose intolerance are seen in approximately 15% of adolescents presenting with GH excess (268).

 

The diagnosis of GH excess is based on the clinical features and auxology in combination with biochemical evidence. Measurement of IGF-I concentration is useful but the reference range used must be specific for the gender, age and pubertal stage of the child. As IGF-I concentrations rise during puberty a precocious puberty will lead to a raised growth velocity with a serum IGF-I concentration which may be raised for age and gender will not be raised for pubertal stage. Due to the variability of GH levels throughout the day assessment of growth hormone levels is either via an oral glucose tolerance test for GH suppression or a GH day curve. The oral glucose tolerance test for GH suppression is essentially identical to a standard oral glucose tolerance test but with measurement of glucose, insulin and GH at 0, 60, 90, 120 and 150 minutes. A normal response is suppression of GH levels to < 0.4 mcg/L (269). Some centers will undertake a GH day curve – measurement of at least 5 separate GH levels over 12 hours, however, given that adolescence is the age at which there is maximal physiological GH secretion and the lack of GH day curve normative data in adolescence interpretation of this test can be challenging.

 

Benign GH secreting adenomas are the most common cause of GH excess. Mutations in the genes encoding GPR101 (causing X-linked acrogigantism), MENIN (270), aryl hydrocarbon receptor interacting protein (200) and p27 (271) are known to predispose to the development of pituitary adenomas. Overall, most GH secreting adenomas are sporadic but the proportion with a genetic basis is likely to be higher in childhood.

 

Transsphenoidal surgery is the treatment of choice for patients with microadenomas, macroadenomas without cavernous sinus or bone extension or where the tumor is causing symptoms from compression (272). Surgical removal is expected to lead to a biochemical cure in 75-95% of patients, with lower probability of cure in patients with macroadenomas. There are three classes of medical treatments for GH excess:

 

  1. Dopamine agonists – cabergoline, bromocriptine
  2. Somatostatin analogues – octreotide, pasireotide, lanreotide
  3. GH receptor antagonists - pegvisomant

 

Medical therapy can be used either where there is failure of surgical therapy, where the tumor is not amenable to surgery or prior to surgery/radiotherapy. Dopamine agonists are the only oral therapy available. Of the dopamine agonists available, only cabergoline has shown efficacy (273) in acromegalic patients and as monotherapy achieves a biochemical cure in a minority of patients (274). Cabergoline is most useful either in tumors which co-secrete prolactin as well as GH or in combination with another therapeutic agent. The somatostatin analogues are effective in both reducing GH and IGF-I levels as well as reducing tumor size. Long acting, once monthly preparations of the somatostatin analogues represent the mainstay of therapy. Somatostatin analogues achieve biochemical resolution in up to 70% of patients (275) and tumor shrinkage (mean size reduction of 50%) in 75% of patients (276). Pegvisomant is the only GH receptor antagonist therapy available and is the most effective therapy at achieving a biochemical cure but in a small proportion (~2%) leads to tumor growth.  Radiotherapy is generally reserved as a third line treatment due to the long-time taken to achieve maximum effect (up to 10 years (277)) and risks of hypopituitarism (up to 50% by 5 years post radiotherapy), visual problems and late effects of cerebrovascular disease and second tumors.  Given the rarity of GH secreting tumors in childhood close liaison with an adult endocrinologist experienced in the management of acromegaly is recommended for a pediatric endocrinologist when faces with such a patient.

 

 

McCune Albright Syndrome

 

McCune Albright syndrome is disorder characterized by the clinical triad of polyostotic fibrous dysplasia, café au lait skin hyperpigmentation and gonadotrophin independent precocious puberty. It is caused by postzygotic activating mutations of GNAS which encodes a stimulatory subunit of G protein, Gsα (278).  GHRH receptor is a G protein coupled receptor and thus McCune Albright syndrome can lead to autonomous GH hypersecretion from the pituitary by activating the signal transduction pathway downstream of this receptor. In a cohort of 58 children and adults with McCune Albright syndrome Akintoye et al (279) identified 12 patients (21%) with GH excess including 6 (4 female, 2 male) who were <16 years. IGF-I concentrations in 10/12 were >2.5 SD above mean but in 2 patients surprisingly they were low at -2.5 and -0.2 SD. This may be due to the cyclical nature of the hormone hypersecretion in McCune Albright syndrome. MR imaging identified microadenomas in 4 patients and no tumor visible in the remaining patients. Clinical diagnosis of GH excess remains difficult as the facial changes can be masked or mistaken for the development of fibrous dysplastic changes in bone and the precocious puberty can mask the GH induced growth excess. The presence of a normal final height in a patient with precocious puberty indicates the potential presence of GH excess (279). Co-secretion of prolactin is common and the majority of patients have hyperprolactinemia. Due to bone thickening and fibrous dysplasia surgery is not usually an option for treatment and radiotherapy is contra-indicated because of the potential for sarcomatous change in fibrous dysplasia. Of the 11 patients with MAS associated acromegaly 6 were treated with cabergoline and then octreotide. Although 5/6 responded to cabergoline treatment with a reduction in IGF-I concentrations none normalized their IGF-I concentration and a combination of cabergoline and octreotide normalized IGF-I concentrations in 4/6 patients.  In a crossover trial of somatostatin analogue therapy and Pegvisomant in McCune Albright induced GH excess pegvisomant was effective in normalizing IGF-I concentrations in 4/5 patients while somatostatin therapy was effective in 3/5 patients (280).

 

Carney Complex  

 

The Carney complex is an autosomal dominant disorder characterized by skin pigmentary abnormalities, myxomas, endocrine tumors or overactivity, and schwannomas. It is known to be caused by loss of function mutations in the PRKAR1A gene which encodes the regulatory subunit of protein kinase A (281). Dissociation of the regulatory subunits from the catalytic subunits of protein kinase A leads to activation of signal transduction. Under normal circumstances this dissociation is triggered by cAMP. Carney complex associated mutations lead to loss of the regulatory subunit and increased activity of protein kinase A associated signal transduction.  GH secreting adenomas are reported in 10% of patients with carney complex but these are rare before puberty (282). Mild abnormalities in GH, IGF-I and prolactin levels are present in up to 79% of patients and there probably a long period of sommatomammotroph cell hypertrophy and mild hypersecretion prior to the development of true GH excess (283). Histology of Carney complex associated GH tumors is distinct and includes the presence of multifocal tumors, somatomammotroph hypertrophy and the secretion of multiple hormones from the tumor (284).

 

CONCLUSIONS

 

Growth disorders are one of the most common reasons for referral to a pediatric endocrinologist. GH deficiency can be effectively treated with recombinant human growth hormone but controversy still exists over the diagnosis of GH deficiency in childhood, particularly in relation to priming of GH stimulation tests. Over the past decade there has been a great expansion in our knowledge of the genetic causes underlying the congenital disorders causing hypopituitarism and GH deficiency but this has not yet led to any new therapies. While extremely rare in pediatric practice GH excess is an important diagnosis to consider in the tall child/adolescent and management should be undertaken in conjunction with an adult endocrinologist.

 

Important Concepts

 

  • GH signal transduction is not induced by GHR dimerization but by a conformational change in the predimerized GHR leading to repositioning of the BOX1 motifs
  • The diagnosis of growth hormone deficiency is made by combining information from auxology, biochemistry, and neuroimaging.
  • In addition to GH deficiency and Laron syndrome there are now additional disorders of the GH-IGF-I axis – Stat5b deficiency, ALS deficiency, haploinsufficiency, and mutations in IGF1R and mutations in the IGF-I gene.
  • There is an expanding number of genes where mutations lead to a disturbance of pituitary gland formation and pituitary hormone deficiency, however in the majority of patients with congenital hypopituitarism the genetic etiology remains unknown. Consider genetic screening in patients where there are multiple affected individuals in the family and in children where they have associated eye abnormalities.
  • Response to growth hormone therapy is generally very good in patients with congenital GH deficiency where a final adult height within parental target range should be expected. In contrast, in patients with radiation induced GH deficiency, GH treatment is less effective and acts mainly to prevent further height loss.
  • Recombinant human IGF-I is available for treating children with GH insensitivity. While first year height velocity often improves significantly the long-term effects on height are less effective than in children with congenital GH deficiency treated with growth hormone.
  • GH excess is an extremely rare disorder in childhood. All childhood patients with a GH secreting adenoma should be screened for mutations in AIP and MEN1 and management should be shared with an adult endocrinologist.

 

REFERENCES

 

  1. Gluckman PD, Johnson-Barrett JJ, Butler JH, Edgar BW, Gunn TR. Studies of insulin-like growth factor -I and -II by specific radioligand assays in umbilical cord blood. Clinical endocrinology 1983; 19:405-413
  2. Verhaeghe J, Van Bree R, Van Herck E, Laureys J, Bouillon R, Van Assche FA. C-peptide, insulin-like growth factors I and II, and insulin-like growth factor binding protein-1 in umbilical cord serum: correlations with birth weight. American journal of obstetrics and gynecology 1993; 169:89-97
  3. Woods KA, Camacho-Hubner C, Savage MO, Clark AJ. Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. The New England journal of medicine 1996; 335:1363-1367
  4. Binder G, Hettmann S, Weber K, Kohlmuller D, Schweizer R. Analysis of the GH content within archived dried blood spots of newborn screening cards from children diagnosed with growth hormone deficiency after the neonatal period. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society 2011; 21:314-317
  5. Mayer M, Schmitt K, Kapelari K, Frisch H, Kostl G, Voigt M. Spontaneous growth in growth hormone deficiency from birth until 7 years of age: development of disease-specific growth curves. Hormone research in paediatrics 2010; 74:136-144
  6. Tanner JM. Fetus into man:physical growth from conception to maturity. . Cambridge, MA: Harvard University Press.
  7. Tanner JM. Regulation of Growth in Size in Mammals. Nature 1963; 199:845-850
  8. Baron J, Klein KO, Colli MJ, Yanovski JA, Novosad JA, Bacher JD, Cutler GB, Jr. Catch-up growth after glucocorticoid excess: a mechanism intrinsic to the growth plate. Endocrinology 1994; 135:1367-1371
  9. Giustina A, Scalvini T, Tassi C, Desenzani P, Poiesi C, Wehrenberg WB, Rogol AD, Veldhuis JD. Maturation of the regulation of growth hormone secretion in young males with hypogonadotropic hypogonadism pharmacologically exposed to progressive increments in serum testosterone. The Journal of clinical endocrinology and metabolism 1997; 82:1210-1219
  10. Weissberger AJ, Ho KK. Activation of the somatotropic axis by testosterone in adult males: evidence for the role of aromatization. The Journal of clinical endocrinology and metabolism 1993; 76:1407-1412
  11. Veldhuis JD, Metzger DL, Martha PM, Jr., Mauras N, Kerrigan JR, Keenan B, Rogol AD, Pincus SM. Estrogen and testosterone, but not a nonaromatizable androgen, direct network integration of the hypothalamo-somatotrope (growth hormone)-insulin-like growth factor I axis in the human: evidence from pubertal pathophysiology and sex-steroid hormone replacement. The Journal of clinical endocrinology and metabolism 1997; 82:3414-3420
  12. Meinhardt UJ, Ho KK. Modulation of growth hormone action by sex steroids. Clinical endocrinology 2006; 65:413-422
  13. Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. The New England journal of medicine 1994; 331:1056-1061
  14. Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K. Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. The Journal of clinical endocrinology and metabolism 1995; 80:3689-3698
  15. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999; 402:656-660
  16. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, Bhattacharya S, Carpenter R, Grossman AB, Korbonits M. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. The Journal of clinical endocrinology and metabolism 2002; 87:2988
  17. Inui A, Asakawa A, Bowers CY, Mantovani G, Laviano A, Meguid MM, Fujimiya M. Ghrelin, appetite, and gastric motility: the emerging role of the stomach as an endocrine organ. FASEB J 2004; 18:439-456
  18. Date Y, Nakazato M, Hashiguchi S, Dezaki K, Mondal MS, Hosoda H, Kojima M, Kangawa K, Arima T, Matsuo H, Yada T, Matsukura S. Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes 2002; 51:124-129
  19. Takaya K, Ariyasu H, Kanamoto N, Iwakura H, Yoshimoto A, Harada M, Mori K, Komatsu Y, Usui T, Shimatsu A, Ogawa Y, Hosoda K, Akamizu T, Kojima M, Kangawa K, Nakao K. Ghrelin strongly stimulates growth hormone release in humans. The Journal of clinical endocrinology and metabolism 2000; 85:4908-4911
  20. Tannenbaum GS, Epelbaum J, Bowers CY. Interrelationship between the novel peptide ghrelin and somatostatin/growth hormone-releasing hormone in regulation of pulsatile growth hormone secretion. Endocrinology 2003; 144:967-974
  21. Panetta R, Patel YC. Expression of mRNA for all five human somatostatin receptors (hSSTR1-5) in pituitary tumors. Life sciences 1995; 56:333-342
  22. Siler TM, VandenBerg G, Yen SS, Brazeau P, Vale W, Guillemin R. Inhibition of growth hormone release in humans by somatostatin. The Journal of clinical endocrinology and metabolism 1973; 37:632-634
  23. Broglio F, Koetsveld Pv P, Benso A, Gottero C, Prodam F, Papotti M, Muccioli G, Gauna C, Hofland L, Deghenghi R, Arvat E, Van Der Lely AJ, Ghigo E. Ghrelin secretion is inhibited by either somatostatin or cortistatin in humans. The Journal of clinical endocrinology and metabolism 2002; 87:4829-4832
  24. Hindmarsh PC, Matthews DR, Brook CG. Growth hormone secretion in children determined by time series analysis. Clinical endocrinology 1988; 29:35-44
  25. Skinner AM, Price DA, Addison GM, Clayton PE, Mackay RI, Soo A, Mui CY. The influence of age, size, pubertal status and renal factors on urinary growth hormone excretion in normal children and adolescents. Growth Regul 1992; 2:156-160
  26. Hindmarsh PC, Fall CH, Pringle PJ, Osmond C, Brook CG. Peak and trough growth hormone concentrations have different associations with the insulin-like growth factor axis, body composition, and metabolic parameters. The Journal of clinical endocrinology and metabolism 1997; 82:2172-2176
  27. Niall HD. Revised primary structure for human growth hormone. Nature: New biology 1971; 230:90-91
  28. Masuda N, Watahiki M, Tanaka M, Yamakawa M, Shimizu K, Nagai J, Nakashima K. Molecular cloning of cDNA encoding 20 kDa variant human growth hormone and the alternative splicing mechanism. Biochimica et biophysica acta 1988; 949:125-131
  29. Lewis UJ, Dunn JT, Bonewald LF, Seavey BK, Vanderlaan WP. A naturally occurring structural variant of human growth hormone. The Journal of biological chemistry 1978; 253:2679-2687
  30. Zhang Y, Jiang J, Black RA, Baumann G, Frank SJ. Tumor necrosis factor-alpha converting enzyme (TACE) is a growth hormone binding protein (GHBP) sheddase: the metalloprotease TACE/ADAM-17 is critical for (PMA-induced) GH receptor proteolysis and GHBP generation. Endocrinology 2000; 141:4342-4348
  31. Martini JF, Pezet A, Guezennec CY, Edery M, Postel-Vinay MC, Kelly PA. Monkey growth hormone (GH) receptor gene expression. Evidence for two mechanisms for the generation of the GH binding protein. The Journal of biological chemistry 1997; 272:18951-18958
  32. Baumann G. Growth hormone binding protein 2001. Journal of pediatric endocrinology & metabolism : JPEM 2001; 14:355-375
  33. Fairhall KM, Carmignac DF, Robinson IC. Growth hormone (GH) binding protein and GH interactions in vivo in the guinea pig. Endocrinology 1992; 131:1963-1969
  34. Maheshwari H, Lillioja S, Castillo CE, Mercado M, Baumann G. Growth hormone-binding protein in human lymph. The Journal of clinical endocrinology and metabolism 1995; 80:3582-3584
  35. Brown RJ, Adams JJ, Pelekanos RA, Wan Y, McKinstry WJ, Palethorpe K, Seeber RM, Monks TA, Eidne KA, Parker MW, Waters MJ. Model for growth hormone receptor activation based on subunit rotation within a receptor dimer. Nature structural & molecular biology 2005; 12:814-821
  36. Strous GJ, dos Santos CA, Gent J, Govers R, Sachse M, Schantl J, van Kerkhof P. Ubiquitin system-dependent regulation of growth hormone receptor signal transduction. Current topics in microbiology and immunology 2004; 286:81-118
  37. Brooks AJ, Waters MJ. The growth hormone receptor: mechanism of activation and clinical implications. Nature reviews Endocrinology 2010; 6:515-525
  38. Brooks AJ, Dai W, O'Mara ML, Abankwa D, Chhabra Y, Pelekanos RA, Gardon O, Tunny KA, Blucher KM, Morton CJ, Parker MW, Sierecki E, Gambin Y, Gomez GA, Alexandrov K, Wilson IA, Doxastakis M, Mark AE, Waters MJ. Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science 2014; 344:1249783
  39. Lanning NJ, Carter-Su C. Recent advances in growth hormone signaling. Reviews in endocrine & metabolic disorders 2006; 7:225-235
  40. Rowlinson SW, Yoshizato H, Barclay JL, Brooks AJ, Behncken SN, Kerr LM, Millard K, Palethorpe K, Nielsen K, Clyde-Smith J, Hancock JF, Waters MJ. An agonist-induced conformational change in the growth hormone receptor determines the choice of signalling pathway. Nature cell biology 2008; 10:740-747
  41. Saharinen P, Vihinen M, Silvennoinen O. Autoinhibition of Jak2 tyrosine kinase is dependent on specific regions in its pseudokinase domain. Molecular biology of the cell 2003; 14:1448-1459
  42. Boisclair YR, Rhoads RP, Ueki I, Wang J, Ooi GT. The acid-labile subunit (ALS) of the 150 kDa IGF-binding protein complex: an important but forgotten component of the circulating IGF system. The Journal of endocrinology 2001; 170:63-70
  43. Seccareccia E, Brodt P. The role of the insulin-like growth factor-I receptor in malignancy: an update. Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the International IGF Research Society 2012; 22:193-199
  44. Dupont J, Holzenberger M. IGF type 1 receptor: a cell cycle progression factor that regulates aging. Cell cycle 2003; 2:270-272
  45. Baker J, Liu JP, Robertson EJ, Efstratiadis A. Role of insulin-like growth factors in embryonic and postnatal growth. Cell 1993; 75:73-82
  46. Liu JL, Yakar S, LeRoith D. Conditional knockout of mouse insulin-like growth factor-1 gene using the Cre/loxP system. Proc Soc Exp Biol Med 2000; 223:344-351
  47. Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL, Wu Y, Liu JL, Ooi GT, Setser J, Frystyk J, Boisclair YR, LeRoith D. Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest 2002; 110:771-781
  48. Growth Hormone Research S. Consensus guidelines for the diagnosis and treatment of growth hormone (GH) deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society. The Journal of clinical endocrinology and metabolism 2000; 85:3990-3993
  49. Richmond EJ, Rogol AD. Growth hormone deficiency in children. Pituitary 2008; 11:115-120
  50. Eugster EA, Pescovitz OH. Gigantism. The Journal of clinical endocrinology and metabolism 1999; 84:4379-4384
  51. Murray PG, Hague C, Fafoula O, Patel L, Raabe AL, Cusick C, Hall CM, Wright NB, Amin R, Clayton PE. Associations with multiple pituitary hormone deficiency in patients with an ectopic posterior pituitary gland. Clinical endocrinology 2008; 69:597-602
  52. Clemmons DR. Consensus statement on the standardization and evaluation of growth hormone and insulin-like growth factor assays. Clinical chemistry 2011; 57:555-559
  53. Kaplan SL, Abrams CA, Bell JJ, Conte FA, Grumbach MM. Growth and growth hormone. I. Changes in serum level of growth hormone following hypoglycemia in 134 children with growth retardation. Pediatric research 1968; 2:43-63
  54. Shah A, Stanhope R, Matthew D. Hazards of pharmacological tests of growth hormone secretion in childhood. Bmj 1992; 304:173-174
  55. Mitchell ML, Sawin CT. Growth hormone response to glucagon in diabetic and nondiabetic persons. Israel journal of medical sciences 1972; 8:867
  56. Ghigo E, Bellone J, Aimaretti G, Bellone S, Loche S, Cappa M, Bartolotta E, Dammacco F, Camanni F. Reliability of provocative tests to assess growth hormone secretory status. Study in 472 normally growing children. The Journal of clinical endocrinology and metabolism 1996; 81:3323-3327
  57. Zadik Z, Chalew SA, Gilula Z, Kowarski AA. Reproducibility of growth hormone testing procedures: a comparison between 24-hour integrated concentration and pharmacological stimulation. The Journal of clinical endocrinology and metabolism 1990; 71:1127-1130
  58. Muller A, Scholz M, Blankenstein O, Binder G, Pfaffle R, Korner A, Kiess W, Heider A, Bidlingmaier M, Thiery J, Kratzsch J. Harmonization of growth hormone measurements with different immunoassays by data adjustment. Clinical chemistry and laboratory medicine : CCLM / FESCC 2011; 49:1135-1142
  59. Corneli G, Di Somma C, Baldelli R, Rovere S, Gasco V, Croce CG, Grottoli S, Maccario M, Colao A, Lombardi G, Ghigo E, Camanni F, Aimaretti G. The cut-off limits of the GH response to GH-releasing hormone-arginine test related to body mass index. European journal of endocrinology / European Federation of Endocrine Societies 2005; 153:257-264
  60. Tauber M, Moulin P, Pienkowski C, Jouret B, Rochiccioli P. Growth hormone (GH) retesting and auxological data in 131 GH-deficient patients after completion of treatment. The Journal of clinical endocrinology and metabolism 1997; 82:352-356
  61. Marin G, Domene HM, Barnes KM, Blackwell BJ, Cassorla FG, Cutler GB, Jr. The effects of estrogen priming and puberty on the growth hormone response to standardized treadmill exercise and arginine-insulin in normal girls and boys. The Journal of clinical endocrinology and metabolism 1994; 79:537-541
  62. Lazar L, Phillip M. Is sex hormone priming in peripubertal children prior to growth hormone stimulation tests still appropriate? Hormone research in paediatrics 2010; 73:299-302
  63. Saini S, Hindmarsh PC, Matthews DR, Pringle PJ, Jones J, Preece MA, Brook CG. Reproducibility of 24-hour serum growth hormone profiles in man. Clinical endocrinology 1991; 34:455-462
  64. Spiliotis BE, August GP, Hung W, Sonis W, Mendelson W, Bercu BB. Growth hormone neurosecretory dysfunction. A treatable cause of short stature. Jama 1984; 251:2223-2230
  65. Juul A, Skakkebaek NE. Prediction of the outcome of growth hormone provocative testing in short children by measurement of serum levels of insulin-like growth factor I and insulin-like growth factor binding protein 3. The Journal of pediatrics 1997; 130:197-204
  66. Cianfarani S, Tondinelli T, Spadoni GL, Scire G, Boemi S, Boscherini B. Height velocity and IGF-I assessment in the diagnosis of childhood onset GH insufficiency: do we still need a second GH stimulation test? Clinical endocrinology 2002; 57:161-167
  67. Triulzi F, Scotti G, di Natale B, Pellini C, Lukezic M, Scognamiglio M, Chiumello G. Evidence of a congenital midline brain anomaly in pituitary dwarfs: a magnetic resonance imaging study in 101 patients. Pediatrics 1994; 93:409-416
  68. Genovese E, Maghnie M, Beluffi G, Villa A, Sammarchi L, Severi F, Campani R. Hypothalamic-pituitary vascularization in pituitary stalk transection syndrome: is the pituitary stalk really transected? The role of gadolinium-DTPA with spin-echo T1 imaging and turbo-FLASH technique. Pediatric radiology 1997; 27:48-53
  69. Murray PG, Hague C, Fafoula O, Gleeson H, Patel L, Banerjee I, Raabe AL, Hall CM, Wright NB, Amin R, Clayton PE. Likelihood of persistent GH deficiency into late adolescence: relationship to the presence of an ectopic or normally sited posterior pituitary gland. Clinical endocrinology 2009; 71:215-219
  70. Collett-Solberg PF, Ambler G, Backeljauw PF, Bidlingmaier M, Biller BMK, Boguszewski MCS, Cheung PT, Choong CSY, Cohen LE, Cohen P, Dauber A, Deal CL, Gong C, Hasegawa Y, Hoffman AR, Hofman PL, Horikawa R, Jorge AAL, Juul A, Kamenicky P, Khadilkar V, Kopchick JJ, Kristrom B, Lopes MLA, Luo X, Miller BS, Misra M, Netchine I, Radovick S, Ranke MB, Rogol AD, Rosenfeld RG, Saenger P, Wit JM, Woelfle J. Diagnosis, Genetics, and Therapy of Short Stature in Children: A Growth Hormone Research Society International Perspective. Horm Res Paediatr 2019; 92:1-14
  71. Kristrom B, Aronson AS, Dahlgren J, Gustafsson J, Halldin M, Ivarsson SA, Nilsson NO, Svensson J, Tuvemo T, Albertsson-Wikland K. Growth hormone (GH) dosing during catch-up growth guided by individual responsiveness decreases growth response variability in prepubertal children with GH deficiency or idiopathic short stature. The Journal of clinical endocrinology and metabolism 2009; 94:483-490
  72. Cohen P, Rogol AD, Weng W, Kappelgaard AM, Rosenfeld RG, Germak J, American Norditropin Study G. Efficacy of IGF-based growth hormone (GH) dosing in nonGH-deficient (nonGHD) short stature children with low IGF-I is not related to basal IGF-I levels. Clinical endocrinology 2013; 78:405-414
  73. Bang P, Ahmed SF, Argente J, Backeljauw P, Bettendorf M, Bona G, Coutant R, Rosenfeld RG, Walenkamp MJ, Savage MO. Identification and management of poor response to growth-promoting therapy in children with short stature. Clinical endocrinology 2012; 77:169-181
  74. Lal RA, Hoffman AR. Long-Acting Growth Hormone Preparations in the Treatment of Children. Pediatr Endocrinol Rev 2018; 16:162-167
  75. Johannsson G, Gordon MB, Hojby Rasmussen M, Hakonsson IH, Karges W, Svaerke C, Tahara S, Takano K, Biller BMK. Once-weekly Somapacitan is Effective and Well Tolerated in Adults with GH Deficiency: A Randomized Phase 3 Trial. The Journal of clinical endocrinology and metabolism 2020; 105
  76. Ranke MB, Lindberg A, Chatelain P, Wilton P, Cutfield W, Albertsson-Wikland K, Price DA. Derivation and validation of a mathematical model for predicting the response to exogenous recombinant human growth hormone (GH) in prepubertal children with idiopathic GH deficiency. KIGS International Board. Kabi Pharmacia International Growth Study. The Journal of clinical endocrinology and metabolism 1999; 84:1174-1183
  77. Ranke MB, Lindberg A, Albertsson-Wikland K, Wilton P, Price DA, Reiter EO. Increased response, but lower responsiveness, to growth hormone (GH) in very young children (aged 0-3 years) with idiopathic GH Deficiency: analysis of data from KIGS. The Journal of clinical endocrinology and metabolism 2005; 90:1966-1971
  78. Schonau E, Westermann F, Rauch F, Stabrey A, Wassmer G, Keller E, Bramswig J, Blum WF, German Lilly Growth Response Study G. A new and accurate prediction model for growth response to growth hormone treatment in children with growth hormone deficiency. European journal of endocrinology / European Federation of Endocrine Societies 2001; 144:13-20
  79. Sudfeld H, Kiese K, Heinecke A, Bramswig JH. Prediction of growth response in prepubertal children treated with growth hormone for idiopathic growth hormone deficiency. Acta Paediatr 2000; 89:34-37
  80. Dos Santos C, Essioux L, Teinturier C, Tauber M, Goffin V, Bougneres P. A common polymorphism of the growth hormone receptor is associated with increased responsiveness to growth hormone. Nature genetics 2004; 36:720-724
  81. Renehan AG, Solomon M, Zwahlen M, Morjaria R, Whatmore A, Audi L, Binder G, Blum W, Bougneres P, Santos CD, Carrascosa A, Hokken-Koelega A, Jorge A, Mullis PE, Tauber M, Patel L, Clayton PE. Growth hormone receptor polymorphism and growth hormone therapy response in children: a Bayesian meta-analysis. American journal of epidemiology 2012; 175:867-877
  82. Clayton P, Chatelain P, Tato L, Yoo HW, Ambler GR, Belgorosky A, Quinteiro S, Deal C, Stevens A, Raelson J, Croteau P, Destenaves B, Olivier C. A pharmacogenomic approach to the treatment of children with GH deficiency or Turner syndrome. European journal of endocrinology / European Federation of Endocrine Societies 2013; 169:277-289
  83. Stevens A, Clayton P, Tato L, Yoo HW, Rodriguez-Arnao MD, Skorodok J, Ambler GR, Zignani M, Zieschang J, Della Corte G, Destenaves B, Champigneulle A, Raelson J, Chatelain P. Pharmacogenomics of insulin-like growth factor-I generation during GH treatment in children with GH deficiency or Turner syndrome. The pharmacogenomics journal 2013;
  84. Dauber A, Meng Y, Audi L, Vedantam S, Weaver B, Carrascosa A, Albertsson-Wikland K, Ranke MB, Jorge AAL, Cara J, Wajnrajch MP, Lindberg A, Camacho-Hubner C, Hirschhorn JN. A Genome-Wide Pharmacogenetic Study of Growth Hormone Responsiveness. The Journal of clinical endocrinology and metabolism 2020; 105
  85. Cacciari E, Zucchini S, Carla G, Pirazzoli P, Cicognani A, Mandini M, Busacca M, Trevisan C. Endocrine function and morphological findings in patients with disorders of the hypothalamo-pituitary area: a study with magnetic resonance. Archives of disease in childhood 1990; 65:1199-1202
  86. Maghnie M, Lindberg A, Koltowska-Haggstrom M, Ranke MB. Magnetic resonance imaging of CNS in 15,043 children with GH deficiency in KIGS (Pfizer International Growth Database). European journal of endocrinology / European Federation of Endocrine Societies 2013; 168:211-217
  87. Phillips JA, 3rd, Cogan JD. Genetic basis of endocrine disease. 6. Molecular basis of familial human growth hormone deficiency. The Journal of clinical endocrinology and metabolism 1994; 78:11-16
  88. Lin SC, Lin CR, Gukovsky I, Lusis AJ, Sawchenko PE, Rosenfeld MG. Molecular basis of the little mouse phenotype and implications for cell type-specific growth. Nature 1993; 364:208-213
  89. Wajnrajch MP, Gertner JM, Harbison MD, Chua SC, Jr., Leibel RL. Nonsense mutation in the human growth hormone-releasing hormone receptor causes growth failure analogous to the little (lit) mouse. Nature genetics 1996; 12:88-90
  90. Mullis PE. Genetics of isolated growth hormone deficiency. Journal of clinical research in pediatric endocrinology 2010; 2:52-62
  91. Alba M, Hall CM, Whatmore AJ, Clayton PE, Price DA, Salvatori R. Variability in anterior pituitary size within members of a family with GH deficiency due to a new splice mutation in the GHRH receptor gene. Clinical endocrinology 2004; 60:470-475
  92. Alatzoglou KS, Dattani MT. Phenotype-genotype correlations in congenital isolated growth hormone deficiency (IGHD). Indian journal of pediatrics 2012; 79:99-106
  93. Binder G, Keller E, Mix M, Massa GG, Stokvis-Brantsma WH, Wit JM, Ranke MB. Isolated GH deficiency with dominant inheritance: new mutations, new insights. The Journal of clinical endocrinology and metabolism 2001; 86:3877-3881
  94. Lee MS, Wajnrajch MP, Kim SS, Plotnick LP, Wang J, Gertner JM, Leibel RL, Dannies PS. Autosomal dominant growth hormone (GH) deficiency type II: the Del32-71-GH deletion mutant suppresses secretion of wild-type GH. Endocrinology 2000; 141:883-890
  95. Shariat N, Ryther RC, Phillips JA, 3rd, Robinson IC, Patton JG. Rescue of pituitary function in a mouse model of isolated growth hormone deficiency type II by RNA interference. Endocrinology 2008; 149:580-586
  96. Zhu YL, Conway-Campbell B, Waters MJ, Dannies PS. Prolonged retention after aggregation into secretory granules of human R183H-growth hormone (GH), a mutant that causes autosomal dominant GH deficiency type II. Endocrinology 2002; 143:4243-4248
  97. Salemi S, Yousefi S, Baltensperger K, Robinson IC, Eble A, Simon D, Czernichow P, Binder G, Sonnet E, Mullis PE. Variability of isolated autosomal dominant GH deficiency (IGHD II): impact of the P89L GH mutation on clinical follow-up and GH secretion. European journal of endocrinology / European Federation of Endocrine Societies 2005; 153:791-802
  98. Duriez B, Duquesnoy P, Dastot F, Bougneres P, Amselem S, Goossens M. An exon-skipping mutation in the btk gene of a patient with X-linked agammaglobulinemia and isolated growth hormone deficiency. FEBS Lett 1994; 346:165-170
  99. Argente J, Flores R, Gutierrez-Arumi A, Verma B, Martos-Moreno GA, Cusco I, Oghabian A, Chowen JA, Frilander MJ, Perez-Jurado LA. Defective minor spliceosome mRNA processing results in isolated familial growth hormone deficiency. EMBO molecular medicine 2014; 6:299-306
  100. Martos-Moreno GA, Travieso-Suarez L, Pozo-Roman J, Munoz-Calvo MT, Chowen JA, Frilander MJ, Perez-Jurado LA, Hawkins FG, Argente J. Response to growth hormone in patients with RNPC3 mutations. EMBO Mol Med 2018; 10
  101. Bancalari RE, Gregory LC, McCabe MJ, Dattani MT. Pituitary gland development: an update. Endocrine development 2012; 23:1-15
  102. Dattani MT, Martinez-Barbera JP, Thomas PQ, Brickman JM, Gupta R, Martensson IL, Toresson H, Fox M, Wales JKH, Hindmarsh PC, Krauss S, Beddington RSP, Robinson ICAF. Mutations in the homeobox gene HESX1/Hesx1 associated with septo-optic dysplasia in human and mouse. Nature genetics 1998; 19:125-133
  103. Brickman JM, Clements M, Tyrell R, McNay D, Woods K, Warner J, Stewart A, Beddington RS, Dattani M. Molecular effects of novel mutations in Hesx1/HESX1 associated with human pituitary disorders. Development (Cambridge, England) 2001; 128:5189-5199
  104. Reynaud R, Albarel F, Saveanu A, Kaffel N, Castinetti F, Lecomte P, Brauner R, Simonin G, Gaudart J, Carmona E, Enjalbert A, Barlier A, Brue T. Pituitary stalk interruption syndrome in 83 patients: novel HESX1 mutation and severe hormonal prognosis in malformative forms. European journal of endocrinology / European Federation of Endocrine Societies 2011; 164:457-465
  105. Dateki S, Kosaka K, Hasegawa K, Tanaka H, Azuma N, Yokoya S, Muroya K, Adachi M, Tajima T, Motomura K, Kinoshita E, Moriuchi H, Sato N, Fukami M, Ogata T. Heterozygous orthodenticle homeobox 2 mutations are associated with variable pituitary phenotype. The Journal of clinical endocrinology and metabolism 2010; 95:756-764
  106. Rizzoti K, Lovell-Badge R. Early development of the pituitary gland: induction and shaping of Rathke's pouch. Reviews in endocrine & metabolic disorders 2005; 6:161-172
  107. Solomon NM, Nouri S, Warne GL, Lagerstrom-Fermer M, Forrest SM, Thomas PQ. Increased gene dosage at Xq26-q27 is associated with X-linked hypopituitarism. Genomics 2002; 79:553-559
  108. Laumonnier F, Ronce N, Hamel BC, Thomas P, Lespinasse J, Raynaud M, Paringaux C, Van Bokhoven H, Kalscheuer V, Fryns JP, Chelly J, Moraine C, Briault S. Transcription factor SOX3 is involved in X-linked mental retardation with growth hormone deficiency. American journal of human genetics 2002; 71:1450-1455
  109. Alatzoglou KS, Kelberman D, Cowell CT, Palmer R, Arnhold IJ, Melo ME, Schnabel D, Grueters A, Dattani MT. Increased transactivation associated with SOX3 polyalanine tract deletion in a patient with hypopituitarism. The Journal of clinical endocrinology and metabolism 2011; 96:E685-690
  110. Tumer Z, Bach-Holm D. Axenfeld-Rieger syndrome and spectrum of PITX2 and FOXC1 mutations. Eur J Hum Genet 2009; 17:1527-1539
  111. Bonfig W, Krude H, Schmidt H. A novel mutation of LHX3 is associated with combined pituitary hormone deficiency including ACTH deficiency, sensorineural hearing loss, and short neck-a case report and review of the literature. European journal of pediatrics 2011; 170:1017-1021
  112. Pfaeffle RW, Savage JJ, Hunter CS, Palme C, Ahlmann M, Kumar P, Bellone J, Schoenau E, Korsch E, Bramswig JH, Stobbe HM, Blum WF, Rhodes SJ. Four novel mutations of the LHX3 gene cause combined pituitary hormone deficiencies with or without limited neck rotation. The Journal of clinical endocrinology and metabolism 2007; 92:1909-1919
  113. Pfaeffle RW, Hunter CS, Savage JJ, Duran-Prado M, Mullen RD, Neeb ZP, Eiholzer U, Hesse V, Haddad NG, Stobbe HM, Blum WF, Weigel JF, Rhodes SJ. Three novel missense mutations within the LHX4 gene are associated with variable pituitary hormone deficiencies. The Journal of clinical endocrinology and metabolism 2008; 93:1062-1071
  114. Machinis K, Pantel J, Netchine I, Leger J, Camand OJ, Sobrier ML, Dastot-Le Moal F, Duquesnoy P, Abitbol M, Czernichow P, Amselem S. Syndromic short stature in patients with a germline mutation in the LIM homeobox LHX4. American journal of human genetics 2001; 69:961-968
  115. Franca MM, Jorge AA, Carvalho LR, Costalonga EF, Vasques GA, Leite CC, Mendonca BB, Arnhold IJ. Novel heterozygous nonsense GLI2 mutations in patients with hypopituitarism and ectopic posterior pituitary lobe without holoprosencephaly. The Journal of clinical endocrinology and metabolism 2010; 95:E384-391
  116. Roessler E, Du YZ, Mullor JL, Casas E, Allen WP, Gillessen-Kaesbach G, Roeder ER, Ming JE, Ruiz i Altaba A, Muenke M. Loss-of-function mutations in the human GLI2 gene are associated with pituitary anomalies and holoprosencephaly-like features. Proceedings of the National Academy of Sciences of the United States of America 2003; 100:13424-13429
  117. Raivio T, Avbelj M, McCabe MJ, Romero CJ, Dwyer AA, Tommiska J, Sykiotis GP, Gregory LC, Diaczok D, Tziaferi V, Elting MW, Padidela R, Plummer L, Martin C, Feng B, Zhang C, Zhou QY, Chen H, Mohammadi M, Quinton R, Sidis Y, Radovick S, Dattani MT, Pitteloud N. Genetic overlap in Kallmann syndrome, combined pituitary hormone deficiency, and septo-optic dysplasia. The Journal of clinical endocrinology and metabolism 2012; 97:E694-699
  118. Turton JP, Mehta A, Raza J, Woods KS, Tiulpakov A, Cassar J, Chong K, Thomas PQ, Eunice M, Ammini AC, Bouloux PM, Starzyk J, Hindmarsh PC, Dattani MT. Mutations within the transcription factor PROP1 are rare in a cohort of patients with sporadic combined pituitary hormone deficiency (CPHD). Clinical endocrinology 2005; 63:10-18
  119. Tatsumi K, Miyai K, Notomi T, Kaibe K, Amino N, Mizuno Y, Kohno H. Cretinism with combined hormone deficiency caused by a mutation in the PIT1 gene. Nature genetics 1992; 1:56-58
  120. Sun Y, Bak B, Schoenmakers N, van Trotsenburg AS, Oostdijk W, Voshol P, Cambridge E, White JK, le Tissier P, Gharavy SN, Martinez-Barbera JP, Stokvis-Brantsma WH, Vulsma T, Kempers MJ, Persani L, Campi I, Bonomi M, Beck-Peccoz P, Zhu H, Davis TM, Hokken-Koelega AC, Del Blanco DG, Rangasami JJ, Ruivenkamp CA, Laros JF, Kriek M, Kant SG, Bosch CA, Biermasz NR, Appelman-Dijkstra NM, Corssmit EP, Hovens GC, Pereira AM, den Dunnen JT, Wade MG, Breuning MH, Hennekam RC, Chatterjee K, Dattani MT, Wit JM, Bernard DJ. Loss-of-function mutations in IGSF1 cause an X-linked syndrome of central hypothyroidism and testicular enlargement. Nature genetics 2012; 44:1375-1381
  121. Joustra SD, Schoenmakers N, Persani L, Campi I, Bonomi M, Radetti G, Beck-Peccoz P, Zhu H, Davis TM, Sun Y, Corssmit EP, Appelman-Dijkstra NM, Heinen CA, Pereira AM, Varewijck AJ, Janssen JA, Endert E, Hennekam RC, Lombardi MP, Mannens MM, Bak B, Bernard DJ, Breuning MH, Chatterjee K, Dattani MT, Oostdijk W, Biermasz NR, Wit JM, van Trotsenburg AS. The IGSF1 deficiency syndrome: characteristics of male and female patients. The Journal of clinical endocrinology and metabolism 2013; 98:4942-4952
  122. Joustra SD, Heinen CA, Schoenmakers N, Bonomi M, Ballieux BE, Turgeon MO, Bernard DJ, Fliers E, van Trotsenburg AS, Losekoot M, Persani L, Wit JM, Biermasz NR, Pereira AM, Oostdijk W, Group ICC. IGSF1 Deficiency: Lessons From an Extensive Case Series and Recommendations for Clinical Management. The Journal of clinical endocrinology and metabolism 2016; 101:1627-1636
  123. Howard SR, Guasti L, Ruiz-Babot G, Mancini A, David A, Storr HL, Metherell LA, Sternberg MJ, Cabrera CP, Warren HR, Barnes MR, Quinton R, de Roux N, Young J, Guiochon-Mantel A, Wehkalampi K, Andre V, Gothilf Y, Cariboni A, Dunkel L. IGSF10 mutations dysregulate gonadotropin-releasing hormone neuronal migration resulting in delayed puberty. EMBO molecular medicine 2016; 8:626-642
  124. Webb EA, AlMutair A, Kelberman D, Bacchelli C, Chanudet E, Lescai F, Andoniadou CL, Banyan A, Alsawaid A, Alrifai MT, Alahmesh MA, Balwi M, Mousavy-Gharavy SN, Lukovic B, Burke D, McCabe MJ, Kasia T, Kleta R, Stupka E, Beales PL, Thompson DA, Chong WK, Alkuraya FS, Martinez-Barbera JP, Sowden JC, Dattani MT. ARNT2 mutation causes hypopituitarism, post-natal microcephaly, visual and renal anomalies. Brain : a journal of neurology 2013; 136:3096-3105
  125. Brachet C, Kozhemyakina EA, Boros E, Heinrichs C, Balikova I, Soblet J, Smits G, Vilain C, Mathers PH. Truncating RAX Mutations: Anophthalmia, Hypopituitarism, Diabetes Insipidus, and Cleft Palate in Mice and Men. The Journal of clinical endocrinology and metabolism 2019; 104:2925-2930
  126. Tahoun M, Chandler JC, Ashton E, Haston S, Hannan A, Kim JS, D'Arco F, Bockenhauer D, Anderson G, Lin MH, Marzouk S, Saied MH, Miner JH, Dattani MT, Waters AM. Mutations in LAMB2 Are Associated With Albuminuria and Optic Nerve Hypoplasia With Hypopituitarism. The Journal of clinical endocrinology and metabolism 2020; 105
  127. Adly N, Alhashem A, Ammari A, Alkuraya FS. Ciliary genes TBC1D32/C6orf170 and SCLT1 are mutated in patients with OFD type IX. Hum Mutat 2014; 35:36-40
  128. Hietamaki J, Gregory LC, Ayoub S, Iivonen AP, Vaaralahti K, Liu X, Brandstack N, Buckton AJ, Laine T, Kansakoski J, Hero M, Miettinen PJ, Varjosalo M, Wakeling E, Dattani MT, Raivio T. Loss-of-Function Variants in TBC1D32 Underlie Syndromic Hypopituitarism. The Journal of clinical endocrinology and metabolism 2020; 105
  129. Lucas-Herald AK, Kinning E, Iida A, Wang Z, Miyake N, Ikegawa S, McNeilly J, Ahmed SF. A case of functional growth hormone deficiency and early growth retardation in a child with IFT172 mutations. The Journal of clinical endocrinology and metabolism 2015; 100:1221-1224
  130. Gregory LC, Shah P, Sanner JRF, Arancibia M, Hurst J, Jones WD, Spoudeas H, Le Quesne Stabej P, Williams HJ, Ocaka LA, Loureiro C, Martinez-Aguayo A, Dattani MT. Mutations in MAGEL2 and L1CAM Are Associated With Congenital Hypopituitarism and Arthrogryposis. The Journal of clinical endocrinology and metabolism 2019; 104:5737-5750
  131. Gregory LC, Ferreira CB, Young-Baird SK, Williams HJ, Harakalova M, van Haaften G, Rahman SA, Gaston-Massuet C, Kelberman D, Gosgene, Qasim W, Camper SA, Dever TE, Shah P, Robinson I, Dattani MT. Impaired EIF2S3 function associated with a novel phenotype of X-linked hypopituitarism with glucose dysregulation. EBioMedicine 2019; 42:470-480
  132. Vajravelu ME, Chai J, Krock B, Baker S, Langdon D, Alter C, De Leon DD. Congenital Hyperinsulinism and Hypopituitarism Attributable to a Mutation in FOXA2. The Journal of clinical endocrinology and metabolism 2018; 103:1042-1047
  133. Bashamboo A, Bignon-Topalovic J, Rouba H, McElreavey K, Brauner R. A Nonsense Mutation in the Hedgehog Receptor CDON Associated With Pituitary Stalk Interruption Syndrome. The Journal of clinical endocrinology and metabolism 2016; 101:12-15
  134. Karaca E, Buyukkaya R, Pehlivan D, Charng WL, Yaykasli KO, Bayram Y, Gambin T, Withers M, Atik MM, Arslanoglu I, Bolu S, Erdin S, Buyukkaya A, Yaykasli E, Jhangiani SN, Muzny DM, Gibbs RA, Lupski JR. Whole-exome sequencing identifies homozygous GPR161 mutation in a family with pituitary stalk interruption syndrome. The Journal of clinical endocrinology and metabolism 2015; 100:E140-147
  135. Bashamboo A, Bignon-Topalovic J, Moussi N, McElreavey K, Brauner R. Mutations in the Human ROBO1 Gene in Pituitary Stalk Interruption Syndrome. The Journal of clinical endocrinology and metabolism 2017; 102:2401-2406
  136. Kowarski AA, Schneider J, Ben-Galim E, Weldon VV, Daughaday WH. Growth failure with normal serum RIA-GH and low somatomedin activity: somatomedin restoration and growth acceleration after exogenous GH. The Journal of clinical endocrinology and metabolism 1978; 47:461-464
  137. Takahashi Y, Shirono H, Arisaka O, Takahashi K, Yagi T, Koga J, Kaji H, Okimura Y, Abe H, Tanaka T, Chihara K. Biologically inactive growth hormone caused by an amino acid substitution. J Clin Invest 1997; 100:1159-1165
  138. Takahashi Y, Kaji H, Okimura Y, Goji K, Abe H, Chihara K. Short stature caused by a mutant growth hormone with an antagonistic effect. Endocrine journal 1996; 43 Suppl:S27-32
  139. Takahashi Y, Kaji H, Okimura Y, Goji K, Abe H, Chihara K. Brief report: short stature caused by a mutant growth hormone. The New England journal of medicine 1996; 334:432-436
  140. Petkovic V, Besson A, Thevis M, Lochmatter D, Eble A, Fluck CE, Mullis PE. Evaluation of the biological activity of a growth hormone (GH) mutant (R77C) and its impact on GH responsiveness and stature. The Journal of clinical endocrinology and metabolism 2007; 92:2893-2901
  141. Petkovic V, Thevis M, Lochmatter D, Besson A, Eble A, Fluck CE, Mullis PE. GH mutant (R77C) in a pedigree presenting with the delay of growth and pubertal development: structural analysis of the mutant and evaluation of the biological activity. European journal of endocrinology / European Federation of Endocrine Societies 2007; 157 Suppl 1:S67-74
  142. Besson A, Salemi S, Deladoey J, Vuissoz JM, Eble A, Bidlingmaier M, Burgi S, Honegger U, Fluck C, Mullis PE. Short stature caused by a biologically inactive mutant growth hormone (GH-C53S). The Journal of clinical endocrinology and metabolism 2005; 90:2493-2499
  143. Godowski PJ, Leung DW, Meacham LR, Galgani JP, Hellmiss R, Keret R, Rotwein PS, Parks JS, Laron Z, Wood WI. Characterization of the human growth hormone receptor gene and demonstration of a partial gene deletion in two patients with Laron-type dwarfism. Proceedings of the National Academy of Sciences of the United States of America 1989; 86:8083-8087
  144. Laron Z, Pertzelan A, Mannheimer S. Genetic pituitary dwarfism with high serum concentation of growth hormone--a new inborn error of metabolism? Israel journal of medical sciences 1966; 2:152-155
  145. David A, Hwa V, Metherell LA, Netchine I, Camacho-Hubner C, Clark AJ, Rosenfeld RG, Savage MO. Evidence for a continuum of genetic, phenotypic, and biochemical abnormalities in children with growth hormone insensitivity. Endocrine reviews 2011; 32:472-497
  146. Ayling RM, Ross R, Towner P, Von Laue S, Finidori J, Moutoussamy S, Buchanan CR, Clayton PE, Norman MR. A dominant-negative mutation of the growth hormone receptor causes familial short stature. Nature genetics 1997; 16:13-14
  147. Coutant R, Dorr HG, Gleeson H, Argente J. Diagnosis of endocrine disease: limitations of the IGF1 generation test in children with short stature. European journal of endocrinology / European Federation of Endocrine Societies 2012; 166:351-357
  148. Buckway CK, Guevara-Aguirre J, Pratt KL, Burren CP, Rosenfeld RG. The IGF-I generation test revisited: a marker of GH sensitivity. The Journal of clinical endocrinology and metabolism 2001; 86:5176-5183
  149. Blum WF, Cotterill AM, Postel-Vinay MC, Ranke MB, Savage MO, Wilton P. Improvement of diagnostic criteria in growth hormone insensitivity syndrome: solutions and pitfalls. Pharmacia Study Group on Insulin-like Growth Factor I Treatment in Growth Hormone Insensitivity Syndromes. Acta Paediatr Suppl 1994; 399:117-124
  150. Chernausek SD, Backeljauw PF, Frane J, Kuntze J, Underwood LE, Group GHISC. Long-term treatment with recombinant insulin-like growth factor (IGF)-I in children with severe IGF-I deficiency due to growth hormone insensitivity. The Journal of clinical endocrinology and metabolism 2007; 92:902-910
  151. Backeljauw PF, Kuntze J, Frane J, Calikoglu AS, Chernausek SD. Adult and near-adult height in patients with severe insulin-like growth factor-I deficiency after long-term therapy with recombinant human insulin-like growth factor-I. Hormone research in paediatrics 2013; 80:47-56
  152. Tonella P, Fluck CE, Mullis PE. Insulin-like growth factor-I treatment in primary growth hormone insensitivity: effect of recombinant human IGF-I (rhIGF-I) and rhIGF-I/rhIGF-binding protein-3 complex. Hormone research in paediatrics 2010; 73:140-147
  153. Chapgier A, Wynn RF, Jouanguy E, Filipe-Santos O, Zhang S, Feinberg J, Hawkins K, Casanova JL, Arkwright PD. Human complete Stat-1 deficiency is associated with defective type I and II IFN responses in vitro but immunity to some low virulence viruses in vivo. Journal of immunology 2006; 176:5078-5083
  154. Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J, Pratt KL, Bezrodnik L, Jasper H, Tepper A, Heinrich JJ, Rosenfeld RG. Growth hormone insensitivity associated with a STAT5b mutation. The New England journal of medicine 2003; 349:1139-1147
  155. Bernasconi A, Marino R, Ribas A, Rossi J, Ciaccio M, Oleastro M, Ornani A, Paz R, Rivarola MA, Zelazko M, Belgorosky A. Characterization of immunodeficiency in a patient with growth hormone insensitivity secondary to a novel STAT5b gene mutation. Pediatrics 2006; 118:e1584-1592
  156. Hwa V, Little B, Adiyaman P, Kofoed EM, Pratt KL, Ocal G, Berberoglu M, Rosenfeld RG. Severe growth hormone insensitivity resulting from total absence of signal transducer and activator of transcription 5b. The Journal of clinical endocrinology and metabolism 2005; 90:4260-4266
  157. Vidarsdottir S, Walenkamp MJ, Pereira AM, Karperien M, van Doorn J, van Duyvenvoorde HA, White S, Breuning MH, Roelfsema F, Kruithof MF, van Dissel J, Janssen R, Wit JM, Romijn JA. Clinical and biochemical characteristics of a male patient with a novel homozygous STAT5b mutation. The Journal of clinical endocrinology and metabolism 2006; 91:3482-3485
  158. Hwa V, Nadeau K, Wit JM, Rosenfeld RG. STAT5b deficiency: lessons from STAT5b gene mutations. Best Pract Res Clin Endocrinol Metab 2011; 25:61-75
  159. Pugliese-Pires PN, Tonelli CA, Dora JM, Silva PC, Czepielewski M, Simoni G, Arnhold IJ, Jorge AA. A novel STAT5B mutation causing GH insensitivity syndrome associated with hyperprolactinemia and immune dysfunction in two male siblings. European journal of endocrinology / European Federation of Endocrine Societies 2010; 163:349-355
  160. Hwa V, Camacho-Hubner C, Little BM, David A, Metherell LA, El-Khatib N, Savage MO, Rosenfeld RG. Growth hormone insensitivity and severe short stature in siblings: a novel mutation at the exon 13-intron 13 junction of the STAT5b gene. Horm Res 2007; 68:218-224
  161. Klammt J, Neumann D, Gevers EF, Andrew SF, Schwartz ID, Rockstroh D, Colombo R, Sanchez MA, Vokurkova D, Kowalczyk J, Metherell LA, Rosenfeld RG, Pfaffle R, Dattani MT, Dauber A, Hwa V. Dominant-negative STAT5B mutations cause growth hormone insensitivity with short stature and mild immune dysregulation. Nat Commun 2018; 9:2105
  162. Suwanichkul A, Boisclair YR, Olney RC, Durham SK, Powell DR. Conservation of a growth hormone-responsive promoter element in the human and mouse acid-labile subunit genes. Endocrinology 2000; 141:833-838
  163. Domene HM, Bengolea SV, Martinez AS, Ropelato MG, Pennisi P, Scaglia P, Heinrich JJ, Jasper HG. Deficiency of the circulating insulin-like growth factor system associated with inactivation of the acid-labile subunit gene. The New England journal of medicine 2004; 350:570-577
  164. Domene HM, Hwa V, Argente J, Wit JM, Camacho-Hubner C, Jasper HG, Pozo J, van Duyvenvoorde HA, Yakar S, Fofanova-Gambetti OV, Rosenfeld RG. Human acid-labile subunit deficiency: clinical, endocrine and metabolic consequences. Horm Res 2009; 72:129-141
  165. Fuqua JS, Derr M, Rosenfeld RG, Hwa V. Identification of a novel heterozygous IGF1 splicing mutation in a large kindred with familial short stature. Hormone research in paediatrics 2012; 78:59-66
  166. Walenkamp MJ, Karperien M, Pereira AM, Hilhorst-Hofstee Y, van Doorn J, Chen JW, Mohan S, Denley A, Forbes B, van Duyvenvoorde HA, van Thiel SW, Sluimers CA, Bax JJ, de Laat JA, Breuning MB, Romijn JA, Wit JM. Homozygous and heterozygous expression of a novel insulin-like growth factor-I mutation. The Journal of clinical endocrinology and metabolism 2005; 90:2855-2864
  167. Bonapace G, Concolino D, Formicola S, Strisciuglio P. A novel mutation in a patient with insulin-like growth factor 1 (IGF1) deficiency. J Med Genet 2003; 40:913-917
  168. Netchine I, Azzi S, Houang M, Seurin D, Perin L, Ricort JM, Daubas C, Legay C, Mester J, Herich R, Godeau F, Le Bouc Y. Partial primary deficiency of insulin-like growth factor (IGF)-I activity associated with IGF1 mutation demonstrates its critical role in growth and brain development. The Journal of clinical endocrinology and metabolism 2009; 94:3913-3921
  169. Klammt J, Kiess W, Pfaffle R. IGF1R mutations as cause of SGA. Best Pract Res Clin Endocrinol Metab 2011; 25:191-206
  170. Abuzzahab MJ, Schneider A, Goddard A, Grigorescu F, Lautier C, Keller E, Kiess W, Klammt J, Kratzsch J, Osgood D, Pfaffle R, Raile K, Seidel B, Smith RJ, Chernausek SD. IGF-I receptor mutations resulting in intrauterine and postnatal growth retardation. The New England journal of medicine 2003; 349:2211-2222
  171. Wallborn T, Wuller S, Klammt J, Kruis T, Kratzsch J, Schmidt G, Schlicke M, Muller E, van de Leur HS, Kiess W, Pfaffle R. A heterozygous mutation of the insulin-like growth factor-I receptor causes retention of the nascent protein in the endoplasmic reticulum and results in intrauterine and postnatal growth retardation. The Journal of clinical endocrinology and metabolism 2010; 95:2316-2324
  172. Kruis T, Klammt J, Galli-Tsinopoulou A, Wallborn T, Schlicke M, Muller E, Kratzsch J, Korner A, Odeh R, Kiess W, Pfaffle R. Heterozygous mutation within a kinase-conserved motif of the insulin-like growth factor I receptor causes intrauterine and postnatal growth retardation. The Journal of clinical endocrinology and metabolism 2010; 95:1137-1142
  173. Choi JH, Kang M, Kim GH, Hong M, Jin HY, Lee BH, Park JY, Lee SM, Seo EJ, Yoo HW. Clinical and functional characteristics of a novel heterozygous mutation of the IGF1R gene and IGF1R haploinsufficiency due to terminal 15q26.2->qter deletion in patients with intrauterine growth retardation and postnatal catch-up growth failure. The Journal of clinical endocrinology and metabolism 2011; 96:E130-134
  174. Fang P, Schwartz ID, Johnson BD, Derr MA, Roberts CT, Jr., Hwa V, Rosenfeld RG. Familial short stature caused by haploinsufficiency of the insulin-like growth factor i receptor due to nonsense-mediated messenger ribonucleic acid decay. The Journal of clinical endocrinology and metabolism 2009; 94:1740-1747
  175. Inagaki K, Tiulpakov A, Rubtsov P, Sverdlova P, Peterkova V, Yakar S, Terekhov S, LeRoith D. A familial insulin-like growth factor-I receptor mutant leads to short stature: clinical and biochemical characterization. The Journal of clinical endocrinology and metabolism 2007; 92:1542-1548
  176. Roback EW, Barakat AJ, Dev VG, Mbikay M, Chretien M, Butler MG. An infant with deletion of the distal long arm of chromosome 15 (q26.1----qter) and loss of insulin-like growth factor 1 receptor gene. American journal of medical genetics 1991; 38:74-79
  177. Kant SG, Kriek M, Walenkamp MJ, Hansson KB, van Rhijn A, Clayton-Smith J, Wit JM, Breuning MH. Tall stature and duplication of the insulin-like growth factor I receptor gene. European journal of medical genetics 2007; 50:1-10
  178. Klaassens M, Galjaard RJ, Scott DA, Bruggenwirth HT, van Opstal D, Fox MV, Higgins RR, Cohen-Overbeek TE, Schoonderwaldt EM, Lee B, Tibboel D, de Klein A. Prenatal detection and outcome of congenital diaphragmatic hernia (CDH) associated with deletion of chromosome 15q26: two patients and review of the literature. Am J Med Genet A 2007; 143A:2204-2212
  179. Ester WA, van Duyvenvoorde HA, de Wit CC, Broekman AJ, Ruivenkamp CA, Govaerts LC, Wit JM, Hokken-Koelega AC, Losekoot M. Two short children born small for gestational age with insulin-like growth factor 1 receptor haploinsufficiency illustrate the heterogeneity of its phenotype. The Journal of clinical endocrinology and metabolism 2009; 94:4717-4727
  180. Dauber A, Munoz-Calvo MT, Barrios V, Domene HM, Kloverpris S, Serra-Juhe C, Desikan V, Pozo J, Muzumdar R, Martos-Moreno GA, Hawkins F, Jasper HG, Conover CA, Frystyk J, Yakar S, Hwa V, Chowen JA, Oxvig C, Rosenfeld RG, Perez-Jurado LA, Argente J. Mutations in pregnancy-associated plasma protein A2 cause short stature due to low IGF-I availability. EMBO molecular medicine 2016; 8:363-374
  181. Teresa Munoz-Calvo M, Barrios V, Pozo J, Chowen JA, Martos-Moreno GA, Hawkins F, Dauber A, Domene HM, Yakar S, Rosenfeld RG, Perez-Jurado LA, Oxvig C, Frystyk J, Argente J. Treatment with recombinant human insulin-like growth factor-I improves growth in patients with PAPP-A2 deficiency. The Journal of clinical endocrinology and metabolism 2016:jc20162751
  182. Cabrera Salcedo CH, V.; Tyzinski, L;,Andrew, M.; Wasserman, H.; Backeljauw, P.;  Dauber, A. 2016 PAPP-A2 Gene Mutation Effects on Glucose Metabolism and Bone Mineral Density and Response to Therapy with Recombinant Human IGF-I. ESPE; 2016; Paris.
  183. Garre ML, Cama A. Craniopharyngioma: modern concepts in pathogenesis and treatment. Current opinion in pediatrics 2007; 19:471-479
  184. Holsken A, Buchfelder M, Fahlbusch R, Blumcke I, Buslei R. Tumour cell migration in adamantinomatous craniopharyngiomas is promoted by activated Wnt-signalling. Acta neuropathologica 2010; 119:631-639
  185. Schroeder JW, Vezina LG. Pediatric sellar and suprasellar lesions. Pediatric radiology 2011; 41:287-298; quiz 404-285
  186. Bunin GR, Surawicz TS, Witman PA, Preston-Martin S, Davis F, Bruner JM. The descriptive epidemiology of craniopharyngioma. Neurosurgical focus 1997; 3:e1
  187. Muller HL. Childhood craniopharyngioma--current concepts in diagnosis, therapy and follow-up. Nature reviews Endocrinology 2010; 6:609-618
  188. Muller HL. Childhood craniopharyngioma. Pituitary 2013; 16:56-67
  189. Muller HL. Consequences of craniopharyngioma surgery in children. The Journal of clinical endocrinology and metabolism 2011; 96:1981-1991
  190. Visser J, Hukin J, Sargent M, Steinbok P, Goddard K, Fryer C. Late mortality in pediatric patients with craniopharyngioma. J Neurooncol 2010; 100:105-111
  191. Steno J, Bizik I, Steno A, Matejcik V. Craniopharyngiomas in children: how radical should the surgeon be? Child's nervous system : ChNS : official journal of the International Society for Pediatric Neurosurgery 2011; 27:41-54
  192. Muller HL, Gebhardt U, Schroder S, Pohl F, Kortmann RD, Faldum A, Zwiener I, Warmuth-Metz M, Pietsch T, Calaminus G, Kolb R, Wiegand C, Sorensen N, study committee of K. Analyses of treatment variables for patients with childhood craniopharyngioma--results of the multicenter prospective trial KRANIOPHARYNGEOM 2000 after three years of follow-up. Hormone research in paediatrics 2010; 73:175-180
  193. Becker G, Kortmann RD, Skalej M, Bamberg M. The role of radiotherapy in the treatment of craniopharyngioma--indications, results, side effects. Frontiers of radiation therapy and oncology 1999; 33:100-113
  194. Price DA, Wilton P, Jonsson P, Albertsson-Wikland K, Chatelain P, Cutfield W, Ranke MB. Efficacy and safety of growth hormone treatment in children with prior craniopharyngioma: An analysis of the Pharmacia and Upjohn International Growth Database (KIGS) from 1988 to 1996. Hormone Research 1998; 49:91-97
  195. Olsson DS, Buchfelder M, Wiendieck K, Kremenevskaja N, Bengtsson BA, Jakobsson KE, Jarfelt M, Johannsson G, Nilsson AG. Tumour recurrence and enlargement in patients with craniopharyngioma with and without GH replacement therapy during more than 10 years of follow-up. European journal of endocrinology / European Federation of Endocrine Societies 2012; 166:1061-1068
  196. Taguchi T, Takao T, Iwasaki Y, Pooh K, Okazaki M, Hashimoto K, Terada Y. Rapid recurrence of craniopharyngioma following recombinant human growth hormone replacement. J Neurooncol 2010; 100:321-322
  197. Lafferty AR, Chrousos GP. Pituitary tumors in children and adolescents. The Journal of clinical endocrinology and metabolism 1999; 84:4317-4323
  198. Steele CA, MacFarlane IA, Blair J, Cuthbertson DJ, Didi M, Mallucci C, Javadpour M, Daousi C. Pituitary adenomas in childhood, adolescence and young adulthood: presentation, management, endocrine and metabolic outcomes. European journal of endocrinology / European Federation of Endocrine Societies 2010; 163:515-522
  199. Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, EmmertBuck MR, Debelenko LV, Zhuang ZP, Lubensky IA, Liotta LA, Crabtree JS, Wang YP, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong QH, Spiegel AM, Burns AL, Marx SJ. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 1997; 276:404-407
  200. Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, Gundogdu S, De Menis E, Makinen MJ, Launonen V, Karhu A, Aaltonen LA. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science 2006; 312:1228-1230
  201. Cnossen MH, Stam EN, Cooiman LC, Simonsz HJ, Stroink H, Oranje AP, Halley DJ, de Goede-Bolder A, Niermeijer MF, de Muinck Keizer-Schrama SM. Endocrinologic disorders and optic pathway gliomas in children with neurofibromatosis type 1. Pediatrics 1997; 100:667-670
  202. Collet-Solberg PF, Sernyak H, Satin-Smith M, Katz LL, Sutton L, Molloy P, Moshang T, Jr. Endocrine outcome in long-term survivors of low-grade hypothalamic/chiasmatic glioma. Clinical endocrinology 1997; 47:79-85
  203. Brauner R, Malandry F, Rappaport R, Zucker JM, Kalifa C, Pierre-Kahn A, Bataini P, Dufier JL. Growth and endocrine disorders in optic glioma. European journal of pediatrics 1990; 149:825-828
  204. Kim RJ, Janss A, Shanis D, Homan S, Moshang T, Jr. Adult heights attained by children with hypothalamic/chiasmatic glioma treated with growth hormone. The Journal of clinical endocrinology and metabolism 2004; 89:4999-5002
  205. Minkov M. Multisystem Langerhans cell histiocytosis in children: current treatment and future directions. Paediatric drugs 2011; 13:75-86
  206. Clayton PE, Shalet SM. Dose dependency of time of onset of radiation-induced growth hormone deficiency. The Journal of pediatrics 1991; 118:226-228
  207. Kirk JA, Raghupathy P, Stevens MM, Cowell CT, Menser MA, Bergin M, Tink A, Vines RH, Silink M. Growth failure and growth-hormone deficiency after treatment for acute lymphoblastic leukaemia. Lancet 1987; 1:190-193
  208. Ogilvy-Stuart AL, Clark DJ, Wallace WH, Gibson BE, Stevens RF, Shalet SM, Donaldson MD. Endocrine deficit after fractionated total body irradiation. Archives of disease in childhood 1992; 67:1107-1110
  209. Littley MD, Shalet SM, Beardwell CG, Ahmed SR, Applegate G, Sutton ML. Hypopituitarism following external radiotherapy for pituitary tumours in adults. The Quarterly journal of medicine 1989; 70:145-160
  210. Jadresic A, Jimenez LE, Joplin GF. Long-term effect of 90Y pituitary implantation in acromegaly. Acta endocrinologica 1987; 115:301-306
  211. Darzy KH, Shalet SM. Circadian and stimulated thyrotropin secretion in cranially irradiated adult cancer survivors. J Clin Endocr Metab 2005; 90:6490-6497
  212. Achermann JC, Brook CG, Hindmarsh PC. The GH response to low-dose bolus growth hormone-releasing hormone (GHRH(1-29)NH2) is attenuated in patients with longstanding post-irradiation GH insufficiency. European journal of endocrinology / European Federation of Endocrine Societies 2000; 142:359-364
  213. Robinson IC, Fairhall KM, Hendry JH, Shalet SM. Differential radiosensitivity of hypothalamo-pituitary function in the young adult rat. The Journal of endocrinology 2001; 169:519-526
  214. Hochberg Z, Kuten A, Hertz P, Tatcher M, Kedar A, Benderly A. The effect of single-dose radiation on cell survival and growth hormone secretion by rat anterior pituitary cells. Radiation research 1983; 94:508-512
  215. Duffner PK, Cohen ME, Voorhess ML, MacGillivray MH, Brecher ML, Panahon A, Gilani BB. Long-term effects of cranial irradiation on endocrine function in children with brain tumors. A prospective study. Cancer 1985; 56:2189-2193
  216. Samaan NA, Vieto R, Schultz PN, Maor M, Meoz RT, Sampiere VA, Cangir A, Ried HL, Jesse RH, Jr. Hypothalamic, pituitary and thyroid dysfunction after radiotherapy to the head and neck. International journal of radiation oncology, biology, physics 1982; 8:1857-1867
  217. Chen MS, Lin FJ, Huang MJ, Wang PW, Tang S, Leung WM, Leung W. Prospective hormone study of hypothalamic-pituitary function in patients with nasopharyngeal carcinoma after high dose irradiation. Japanese journal of clinical oncology 1989; 19:265-270
  218. Spoudeas HA, Hindmarsh PC, Matthews DR, Brook CG. Evolution of growth hormone neurosecretory disturbance after cranial irradiation for childhood brain tumours: a prospective study. The Journal of endocrinology 1996; 150:329-342
  219. Chrousos GP, Poplack D, Brown T, O'Neill D, Schwade J, Bercu BB. Effects of cranial radiation on hypothalamic-adenohypophyseal function: abnormal growth hormone secretory dynamics. The Journal of clinical endocrinology and metabolism 1982; 54:1135-1139
  220. Blatt J, Bercu BB, Gillin JC, Mendelson WB, Poplack DG. Reduced pulsatile growth hormone secretion in children after therapy for acute lymphoblastic leukemia. The Journal of pediatrics 1984; 104:182-186
  221. Brauner R, Rappaport R, Prevot C, Czernichow P, Zucker JM, Bataini P, Lemerle J, Sarrazin D, Guyda HJ. A prospective study of the development of growth hormone deficiency in children given cranial irradiation, and its relation to statural growth. The Journal of clinical endocrinology and metabolism 1989; 68:346-351
  222. Tillmann V, Buckler JM, Kibirige MS, Price DA, Shalet SM, Wales JK, Addison MG, Gill MS, Whatmore AJ, Clayton PE. Biochemical tests in the diagnosis of childhood growth hormone deficiency. The Journal of clinical endocrinology and metabolism 1997; 82:531-535
  223. Sklar C, Sarafoglou K, Whittam E. Efficacy of insulin-like growth factor binding protein 3 in predicting the growth hormone response to provocative testing in children treated with cranial irradiation. Acta endocrinologica 1993; 129:511-515
  224. Tillmann V, Shalet SM, Price DA, Wales JK, Pennells L, Soden J, Gill MS, Whatmore AJ, Clayton PE. Serum insulin-like growth factor-I, IGF binding protein-3 and IGFBP-3 protease activity after cranial irradiation. Horm Res 1998; 50:71-77
  225. Achermann JC, Hindmarsh PC, Brook CG. The relationship between the growth hormone and insulin-like growth factor axis in long-term survivors of childhood brain tumours. Clinical endocrinology 1998; 49:639-645
  226. Oberfield SE, Allen JC, Pollack J, New MI, Levine LS. Long-term endocrine sequelae after treatment of medulloblastoma: prospective study of growth and thyroid function. The Journal of pediatrics 1986; 108:219-223
  227. Shalet SM, Gibson B, Swindell R, Pearson D. Effect of spinal irradiation on growth. Archives of disease in childhood 1987; 62:461-464
  228. Clayton PE, Shalet SM, Price DA. Growth response to growth hormone therapy following craniospinal irradiation. European journal of pediatrics 1988; 147:597-601
  229. Clayton PE, Shalet SM, Price DA. Growth response to growth hormone therapy following cranial irradiation. European journal of pediatrics 1988; 147:593-596
  230. Sulmont V, Brauner R, Fontoura M, Rappaport R. Response to growth hormone treatment and final height after cranial or craniospinal irradiation. Acta Paediatr Scand 1990; 79:542-549
  231. Yakar S, Kim H, Zhao H, Toyoshima Y, Pennisi P, Gavrilova O, Leroith D. The growth hormone-insulin like growth factor axis revisited: lessons from IGF-1 and IGF-1 receptor gene targeting. Pediatric nephrology 2005; 20:251-254
  232. Clayton PE, Banerjee I, Murray PG, Renehan AG. Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nature reviews Endocrinology 2011; 7:11-24
  233. Chen B, Liu S, Xu W, Wang X, Zhao W, Wu J. IGF-I and IGFBP-3 and the risk of lung cancer: a meta-analysis based on nested case-control studies. Journal of experimental & clinical cancer research : CR 2009; 28:89
  234. Rinaldi S, Cleveland R, Norat T, Biessy C, Rohrmann S, Linseisen J, Boeing H, Pischon T, Panico S, Agnoli C, Palli D, Tumino R, Vineis P, Peeters PH, van Gils CH, Bueno-de-Mesquita BH, Vrieling A, Allen NE, Roddam A, Bingham S, Khaw KT, Manjer J, Borgquist S, Dumeaux V, Torhild Gram I, Lund E, Trichopoulou A, Makrygiannis G, Benetou V, Molina E, Donate Suarez I, Barricarte Gurrea A, Gonzalez CA, Tormo MJ, Altzibar JM, Olsen A, Tjonneland A, Gronbaek H, Overvad K, Clavel-Chapelon F, Boutron-Ruault MC, Morois S, Slimani N, Boffetta P, Jenab M, Riboli E, Kaaks R. Serum levels of IGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies. Int J Cancer 2010; 126:1702-1715
  235. Roddam AW, Allen NE, Appleby P, Key TJ, Ferrucci L, Carter HB, Metter EJ, Chen C, Weiss NS, Fitzpatrick A, Hsing AW, Lacey JV, Jr., Helzlsouer K, Rinaldi S, Riboli E, Kaaks R, Janssen JA, Wildhagen MF, Schroder FH, Platz EA, Pollak M, Giovannucci E, Schaefer C, Quesenberry CP, Jr., Vogelman JH, Severi G, English DR, Giles GG, Stattin P, Hallmans G, Johansson M, Chan JM, Gann P, Oliver SE, Holly JM, Donovan J, Meyer F, Bairati I, Galan P. Insulin-like growth factors, their binding proteins, and prostate cancer risk: analysis of individual patient data from 12 prospective studies. Annals of internal medicine 2008; 149:461-471, W483-468
  236. Endogenous H, Breast Cancer Collaborative G, Key TJ, Appleby PN, Reeves GK, Roddam AW. Insulin-like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: pooled individual data analysis of 17 prospective studies. The lancet oncology 2010; 11:530-542
  237. Kauppinen-Makelin R, Sane T, Valimaki MJ, Markkanen H, Niskanen L, Ebeling T, Jaatinen P, Juonala M, Finnish Acromegaly Study G, Pukkala E. Increased cancer incidence in acromegaly--a nationwide survey. Clinical endocrinology 2010; 72:278-279
  238. Ron E, Gridley G, Hrubec Z, Page W, Arora S, Fraumeni JF, Jr. Acromegaly and gastrointestinal cancer. Cancer 1991; 68:1673-1677
  239. Orme SM, McNally RJ, Cartwright RA, Belchetz PE. Mortality and cancer incidence in acromegaly: a retrospective cohort study. United Kingdom Acromegaly Study Group. The Journal of clinical endocrinology and metabolism 1998; 83:2730-2734
  240. Sklar CA, Mertens AC, Mitby P, Occhiogrosso G, Qin J, Heller G, Yasui Y, Robison LL. Risk of disease recurrence and second neoplasms in survivors of childhood cancer treated with growth hormone: a report from the Childhood Cancer Survivor Study. The Journal of clinical endocrinology and metabolism 2002; 87:3136-3141
  241. Swerdlow AJ, Reddingius RE, Higgins CD, Spoudeas HA, Phipps K, Qiao Z, Ryder WD, Brada M, Hayward RD, Brook CG, Hindmarsh PC, Shalet SM. Growth hormone treatment of children with brain tumors and risk of tumor recurrence. The Journal of clinical endocrinology and metabolism 2000; 85:4444-4449
  242. Blethen SL, Allen DB, Graves D, August G, Moshang T, Rosenfeld R. Safety of recombinant deoxyribonucleic acid-derived growth hormone: The National Cooperative Growth Study experience. The Journal of clinical endocrinology and metabolism 1996; 81:1704-1710
  243. Maneatis T, Baptista J, Connelly K, Blethen S. Growth hormone safety update from the National Cooperative Growth Study. Journal of pediatric endocrinology & metabolism : JPEM 2000; 13 Suppl 2:1035-1044
  244. Wyatt D. Lessons from the national cooperative growth study. European journal of endocrinology / European Federation of Endocrine Societies 2004; 151 Suppl 1:S55-59
  245. Ergun-Longmire B, Mertens AC, Mitby P, Qin J, Heller G, Shi W, Yasui Y, Robison LL, Sklar CA. Growth hormone treatment and risk of second neoplasms in the childhood cancer survivor. The Journal of clinical endocrinology and metabolism 2006; 91:3494-3498
  246. Crompton MR. Hypothalamic lesions following closed head injury. Brain : a journal of neurology 1971; 94:165-172
  247. Benvenga S, Campenni A, Ruggeri RM, Trimarchi F. Clinical review 113: Hypopituitarism secondary to head trauma. The Journal of clinical endocrinology and metabolism 2000; 85:1353-1361
  248. Kelly DF, Gonzalo IT, Cohan P, Berman N, Swerdloff R, Wang C. Hypopituitarism following traumatic brain injury and aneurysmal subarachnoid hemorrhage: a preliminary report. Journal of neurosurgery 2000; 93:743-752
  249. Lieberman SA, Oberoi AL, Gilkison CR, Masel BE, Urban RJ. Prevalence of neuroendocrine dysfunction in patients recovering from traumatic brain injury. The Journal of clinical endocrinology and metabolism 2001; 86:2752-2756
  250. Bondanelli M, De Marinis L, Ambrosio MR, Monesi M, Valle D, Zatelli MC, Fusco A, Bianchi A, Farneti M, degli Uberti EC. Occurrence of pituitary dysfunction following traumatic brain injury. Journal of neurotrauma 2004; 21:685-696
  251. Leal-Cerro A, Flores JM, Rincon M, Murillo F, Pujol M, Garcia-Pesquera F, Dieguez C, Casanueva FF. Prevalence of hypopituitarism and growth hormone deficiency in adults long-term after severe traumatic brain injury. Clinical endocrinology 2005; 62:525-532
  252. Agha A, Rogers B, Sherlock M, O'Kelly P, Tormey W, Phillips J, Thompson CJ. Anterior pituitary dysfunction in survivors of traumatic brain injury. The Journal of clinical endocrinology and metabolism 2004; 89:4929-4936
  253. Tanriverdi F, Senyurek H, Unluhizarci K, Selcuklu A, Casanueva FF, Kelestimur F. High risk of hypopituitarism after traumatic brain injury: a prospective investigation of anterior pituitary function in the acute phase and 12 months after trauma. The Journal of clinical endocrinology and metabolism 2006; 91:2105-2111
  254. Acerini CL, Tasker RC, Bellone S, Bona G, Thompson CJ, Savage MO. Hypopituitarism in childhood and adolescence following traumatic brain injury: the case for prospective endocrine investigation. European journal of endocrinology / European Federation of Endocrine Societies 2006; 155:663-669
  255. Einaudi S, Matarazzo P, Peretta P, Grossetti R, Giordano F, Altare F, Bondone C, Andreo M, Ivani G, Genitori L, de Sanctis C. Hypothalamo-hypophysial dysfunction after traumatic brain injury in children and adolescents: a preliminary retrospective and prospective study. Journal of pediatric endocrinology & metabolism : JPEM 2006; 19:691-703
  256. Khadr SN, Crofton PM, Jones PA, Wardhaugh B, Roach J, Drake AJ, Minns RA, Kelnar CJ. Evaluation of pituitary function after traumatic brain injury in childhood. Clinical endocrinology 2010; 73:637-643
  257. Poomthavorn P, Maixner W, Zacharin M. Pituitary function in paediatric survivors of severe traumatic brain injury. Archives of disease in childhood 2008; 93:133-137
  258. Heather NL, Jefferies C, Hofman PL, Derraik JG, Brennan C, Kelly P, Hamill JK, Jones RG, Rowe DL, Cutfield WS. Permanent hypopituitarism is rare after structural traumatic brain injury in early childhood. The Journal of clinical endocrinology and metabolism 2012; 97:599-604
  259. Buxton N, Robertson I. Lymphocytic and granulocytic hypophysitis: a single centre experience. British journal of neurosurgery 2001; 15:242-245, discussion 245-246
  260. Carmichael JD. Update on the diagnosis and management of hypophysitis. Current opinion in endocrinology, diabetes, and obesity 2012; 19:314-321
  261. Gellner V, Kurschel S, Scarpatetti M, Mokry M. Lymphocytic hypophysitis in the pediatric population. Child's nervous system : ChNS : official journal of the International Society for Pediatric Neurosurgery 2008; 24:785-792
  262. Cemeroglu AP, Blaivas M, Muraszko KM, Robertson PL, Vazquez DM. Lymphocytic hypophysitis presenting with diabetes insipidus in a 14-year-old girl: case report and review of the literature. European journal of pediatrics 1997; 156:684-688
  263. Papanastasiou L, Pappa T, Tsiavos V, Tseniklidi E, Androulakis I, Kontogeorgos G, Piaditis G. Azathioprine as an alternative treatment in primary hypophysitis. Pituitary 2011; 14:16-22
  264. Tubridy N, Saunders D, Thom M, Asa SL, Powell M, Plant GT, Howard R. Infundibulohypophysitis in a man presenting with diabetes insipidus and cavernous sinus involvement. Journal of neurology, neurosurgery, and psychiatry 2001; 71:798-801
  265. Ward L, Paquette J, Seidman E, Huot C, Alvarez F, Crock P, Delvin E, Kampe O, Deal C. Severe autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy in an adolescent girl with a novel AIRE mutation: response to immunosuppressive therapy. The Journal of clinical endocrinology and metabolism 1999; 84:844-852
  266. Selch MT, DeSalles AA, Kelly DF, Frighetto L, Vinters HV, Cabatan-Awang C, Wallace RE, Solberg TD. Stereotactic radiotherapy for the treatment of lymphocytic hypophysitis. Report of two cases. Journal of neurosurgery 2003; 99:591-596
  267. Abe T, Tara LA, Ludecke DK. Growth hormone-secreting pituitary adenomas in childhood and adolescence: features and results of transnasal surgery. Neurosurgery 1999; 45:1-10
  268. Bhansali A, Upreti V, Dutta P, Mukherjee KK, Nahar U, Santosh R, Das S, Walia R, Pathak A. Adolescent acromegaly: clinical parameters and treatment outcome. Journal of pediatric endocrinology & metabolism : JPEM 2010; 23:1047-1054
  269. Katznelson L, Atkinson JL, Cook DM, Ezzat SZ, Hamrahian AH, Miller KK, American Association of Clinical E. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the diagnosis and treatment of acromegaly--2011 update. Endocrine practice : official journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists 2011; 17 Suppl 4:1-44
  270. Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 1997; 276:404-407
  271. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Hofler H, Fend F, Graw J, Atkinson MJ. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proceedings of the National Academy of Sciences of the United States of America 2006; 103:15558-15563
  272. Melmed S, Colao A, Barkan A, Molitch M, Grossman AB, Kleinberg D, Clemmons D, Chanson P, Laws E, Schlechte J, Vance ML, Ho K, Giustina A, Acromegaly Consensus G. Guidelines for acromegaly management: an update. The Journal of clinical endocrinology and metabolism 2009; 94:1509-1517
  273. Colao A, Ferone D, Marzullo P, Di Sarno A, Cerbone G, Sarnacchiaro F, Cirillo S, Merola B, Lombardi G. Effect of different dopaminergic agents in the treatment of acromegaly. The Journal of clinical endocrinology and metabolism 1997; 82:518-523
  274. Abs R, Verhelst J, Maiter D, Van Acker K, Nobels F, Coolens JL, Mahler C, Beckers A. Cabergoline in the treatment of acromegaly: a study in 64 patients. The Journal of clinical endocrinology and metabolism 1998; 83:374-378
  275. Maiza JC, Vezzosi D, Matta M, Donadille F, Loubes-Lacroix F, Cournot M, Bennet A, Caron P. Long-term (up to 18 years) effects on GH/IGF-1 hypersecretion and tumour size of primary somatostatin analogue (SSTa) therapy in patients with GH-secreting pituitary adenoma responsive to SSTa. Clinical endocrinology 2007; 67:282-289
  276. Bevan JS. Clinical review: The antitumoral effects of somatostatin analog therapy in acromegaly. The Journal of clinical endocrinology and metabolism 2005; 90:1856-1863
  277. Minniti G, Jaffrain-Rea ML, Osti M, Esposito V, Santoro A, Solda F, Gargiulo P, Tamburrano G, Enrici RM. The long-term efficacy of conventional radiotherapy in patients with GH-secreting pituitary adenomas. Clinical endocrinology 2005; 62:210-216
  278. Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. The New England journal of medicine 1991; 325:1688-1695
  279. Akintoye SO, Chebli C, Booher S, Feuillan P, Kushner H, Leroith D, Cherman N, Bianco P, Wientroub S, Robey PG, Collins MT. Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. The Journal of clinical endocrinology and metabolism 2002; 87:5104-5112
  280. Akintoye SO, Kelly MH, Brillante B, Cherman N, Turner S, Butman JA, Robey PG, Collins MT. Pegvisomant for the treatment of gsp-mediated growth hormone excess in patients with McCune-Albright syndrome. The Journal of clinical endocrinology and metabolism 2006; 91:2960-2966
  281. 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. Nature genetics 2000; 26:89-92
  282. Stratakis CA, Kirschner LS, Carney JA. Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. The Journal of clinical endocrinology and metabolism 2001; 86:4041-4046
  283. Horvath A, Stratakis CA. Clinical and molecular genetics of acromegaly: MEN1, Carney complex, McCune-Albright syndrome, familial acromegaly and genetic defects in sporadic tumors. Reviews in endocrine & metabolic disorders 2008; 9:1-11
  284. Pack SD, Kirschner LS, Pak E, Zhuang Z, Carney JA, Stratakis CA. Genetic and histologic studies of somatomammotropic pituitary tumors in patients with the "complex of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas" (Carney complex). The Journal of clinical endocrinology and metabolism 2000; 85:3860-3865

 

 

Thyroid Storm

CLINICAL RECOGNITION


Thyroid (or thyrotoxic) storm is an acute, life-threatening syndrome due to an exacerbation of thyrotoxicosis. It is now an infrequent condition because of earlier diagnosis and treatment of thyrotoxicosis and better pre- and postoperative medical management. In the United States the incidence of thyroid storm ranged between 0.57 and 0.76 cases/100,000 persons per year. Thyroid storm may be precipitated by a number of factors including intercurrent illness, especially infections (Table 1). Pneumonia, upper respiratory tract infection, enteric infections, or any other infection can precipitate thyroid storm. Thyroid storm in the past most frequently occurred after surgery, but this is now unusual. Occasionally it occurs as a manifestation of untreated or partially treated thyrotoxicosis without another apparent precipitating factor. In the Japanese experience approximately 20% of patients developed thyroid storm before they received anti-thyroid drug treatment. Finally, if patients are not compliant with anti-thyroid medications thyroid storm may occur and this is a relatively common cause. Thyroid storm is typically associated with Graves' disease, but it may occur in patients with toxic nodular goiter or any other cause of thyrotoxicosis.

 

Table 1. Factors That May Precipitate Thyroid Storm

Infections

Acute Illness such as acute myocardial infarction, stroke, congestive heart failure, trauma, etc.

Non-thyroid surgery in a hyperthyroid patient

Thyroid surgery in a patient poorly prepared for surgery

Discontinuation of anti-thyroid medications

Radioiodine therapy

Recent use of iodinated contrast

Pregnancy particularly during labor and delivery

 

Classic features of thyroid storm include fever, marked tachycardia, heart failure, tremors, nausea and vomiting, diarrhea, dehydration, restlessness, extreme agitation, delirium or coma (Table 2). Fever is typical and may be higher than 105.8 F (41 C). Patients may present with a true psychosis or a marked deterioration of previously abnormal behavior. Rarely thyroid storm takes a strikingly different form, called apathetic storm, with extreme weakness, emotional apathy, confusion, and absent or low fever.

 

Signs and symptoms of decompensation in organ systems may be present. Delirium is one example. Congestive heart failure may also occur with peripheral edema, congestive hepatomegaly, and respiratory distress. Marked sinus tachycardia or tachyarrhythmia, such as atrial fibrillation, are common. Liver damage and jaundice may result from congestive heart failure or the direct action of thyroid hormone on the liver. Fever and vomiting may produce dehydration and prerenal azotemia. Abdominal pain may be a prominent feature. The clinical picture may be masked by a secondary infection such as pneumonia, a viral infection, or infection of the upper respiratory tract.

 

Table 2. Clinical Manifestations of Thyroid Storm

History of thyroid disease

Goiter/thyroid eye disease

High fever

Marked tachycardia, occasionally atrial fibrillation

Heart Failure

Tremor

Sweating

Nausea and vomiting

Agitation/psychosis

Delirium/coma

Jaundice

Abdominal pain

 

Death from thyroid storm is not as common as in the past if it is promptly recognized and aggressively treated in an intensive care unit, but is still approximately 10-25%. In recent nationwide studies from Japan the mortality rate was >10%. Death may be from cardiac failure, shock, hyperthermia, multiple organ failure, or other complications. Additionally, even when patients survive, some have irreversible damage including brain damage, disuse atrophy, cerebrovascular disease, renal insufficiency, and psychosis. 

 

PATHOPHYSIOLOGY

Thyroid storm classically began a few hours after thyroidectomy performed on a patient prepared for surgery by potassium iodide alone. Many such patients were not euthyroid and would not be considered appropriately prepared for surgery by current standards. Exacerbation of thyrotoxicosis is still seen in patients sent to surgery before adequate preparation, but it is unusual in the anti-thyroid drug-controlled patient. Thyroid storm occasionally occurs in patients operated on for some other illness while severely thyrotoxic. Severe exacerbation of thyrotoxicosis is rarely seen following 131-I therapy for hyperthyroidism; but some of these exacerbations may be defined as thyroid storm.

Thyroid storm appears most commonly following infection, which seems to induce an escape from control of thyrotoxicosis. Pneumonia, upper respiratory tract infections, enteric infections, or any other infection can cause this condition. Interestingly, serum free T4 concentrations were higher in patients with thyroid storm than in those with uncomplicated thyrotoxicosis, while serum total T4 levels did not differ in the two groups, suggesting that events like infections may decrease serum binding of T4 and cause a greater increase in free T4 responsible for storm occurrence. Another common cause of thyroid storm is a hyperthyroid patient suddenly stopping their anti-thyroid drugs.

 

DIAGNOSIS AND DIFFERENTIAL

Diagnosis of thyroid storm is made on clinical grounds and involves the usual diagnostic measures for thyrotoxicosis. A history of hyperthyroidism or physical findings of an enlarged thyroid or hyperthyroid eye findings is helpful in suggesting the diagnosis. The central features are thyrotoxicosis, abnormal CNS function, fever, tachycardia (usually above 130bpm), GI tract symptoms, and evidence of impending or present CHF. There are no distinctive laboratory abnormalities. Free T4 and, if possible, free T3 should be measured. Note that T3 levels may be markedly reduced in relation to the severity of the illness, as part of the associated “non-thyroidal illness syndrome”. As expected, TSH levels are suppressed. Electrolytes, blood urea nitrogen (BUN), blood sugar, liver function tests, and plasma cortisol should be monitored. While the diagnosis of thyroid storm remains largely a matter of clinical judgment, there are two scales for assessing the severity of hyperthyroidism and determining the likelihood of thyroid storm (Figures 1 and 2). Recognize that these scoring systems are just guidelines and clinical judgement is still crucial. Data comparing these two diagnostic systems suggest an overall agreement, but a tendency toward underdiagnosis using the Japanese criteria. Unfortunately, there are no unique laboratory abnormalities that facilitate the diagnosis of thyroid storm.

 

Figure 1. Burch-Wartofsky Point Scale for the Diagnosis of Thyroid Storm. When it is not possible to distinguish the effects of an intercurrent illness from those of severe thyrotoxicosis per se, points are awarded such as to favor the diagnosis of storm and hence, empiric therapy. Endocrinol Metab Clin North Am 22:263–277.

Figure 2. Japanese Thyroid Association Criteria for Thyroid Storm

 

THERAPY

Thyroid storm is a medical emergency that has to be recognized and treated immediately (Table 3). Admission to an intensive care unit is usually required. Besides treatment for thyroid storm, it is essential to treat precipitating factors such as infections. As would be expected given the rare occurrence of thyroid storm there are very few randomized controlled treatment trials and therefore much of what is recommended is based on expert opinion.

 

Table 3. Treatment of Thyroid Storm

Supportive Measures
1. Rest
2. Mild sedation
3. Fluid and electrolyte replacement
4. Nutritional support and vitamins as needed
5. Oxygen therapy
6. Nonspecific therapy as indicated
7. Antibiotics
8. Cardio-support as indicated
9. Cooling, aided by cooling blankets and acetaminophen
Specific therapy
1. Beta-blocking agents. Propranolol (60 to 80 mg orally every 4 hours, or 1 to 3 mg intravenously every 4 to 6 hours), Start with low doses. Esmolol in ICU setting (loading dose of 250 mcg/kg to 500 mcg/kg followed by 50 mcg/kg to 100 mcg/kg/minute).
2. Antithyroid drugs (PTU 500–1000mg load, then 250mg every 4 hours or Methimazole 60-80mg/day), then taper as condition improves
3. Potassium iodide (one hour after first dose of antithyroid drugs):
 250mg orally every 6 hours
4. Hydrocortisone 300mg intravenous load, then 100mg every 8 hours.
Second Line Therapy
1. Plasmapheresis
2. Oral T4 and T3 binding resins- colestipol or cholestyramine
3. Dialysis

4. Lithium in patients who cannot take iodine

5. Thyroid surgery

 

It should be noted that if any possibility is present that orally given drugs will not be appropriately absorbed (e.g., due to stomach distention, vomiting, diarrhea or severe heart failure), the intravenous route should be used. If the thyrotoxic patient is untreated, an antithyroid drug should be given. PTU, 500–1000mg load, then 250mg every 4 hours, should be used if possible, rather than methimazole, since PTU also prevents peripheral conversion of T4 to T3, thus it may more rapidly reduce circulating T3 levels. Methimazole (60–80mg/day) can be given orally, or if necessary, the pure compound can be made up in a 10 mg/ml solution for parenteral administration. Methimazole is also absorbed when given rectally in a suppository. After initial stabilization, one should taper the dose and treat with Methimazole if PTU was started at the beginning as the safety profile of Methimazole is superior. If the thyroid storm is due to thyroiditis neither PTU nor Methimazole will be effective and should not be used.

 

An hour after PTU or Methimazole has been given, iodide should be administered. A dosage of 250 mg every 6 hours is more than sufficient. The iodine is given after PTU or Methimazole because the iodine could stimulate thyroid hormone synthesis. Unless congestive heart failure contraindicates it, propranolol or other beta-blocking agents should be given at once, orally or parenterally, depending on the patient's clinical status. Beta-blocking agents control tachycardia, restlessness, and other symptoms. Additionally, propranolol inhibits type 1 deiodinase decreasing the conversion of T4 to T3. Probably lower doses should be administered initially, since the administration of beta-blockers to patients with severe thyrotoxicosis has been associated with vascular collapse. Esmolol, a short-acting beta blocker, at a loading dose of 250 mcg/kg to 500 mcg/kg followed by 50 mcg/kg to 100 mcg/kg/minute can be used in an ICU setting.  For patients with reactive airway disease, a cardioselective beta blocker like atenolol or metoprolol can be employed.

 

Permanent correction of thyrotoxicosis by either 131-I or thyroidectomy should be deferred until euthyroidism is restored. Other supporting measures should fully be exploited, including sedation, oxygen, treatment for tachycardia or congestive heart failure, rehydration, multivitamins, occasionally supportive transfusions, and cooling the patient to lower body temperature down. Antibiotics may be given on the presumption of infection while results of cultures are awaited.

 

The adrenal gland may be limited in its ability to increase steroid production during thyrotoxicosis. Therefore, hydrocortisone (100-300 mg/day) or dexamethasone (2mg every 6 hours) or its equivalent should be given. The dose can rapidly be reduced when the acute process subsides. Pharmacological doses of glucocorticoids (2 mg dexamethasone every 6 h) acutely depress serum T3 levels by reducing T4 to T3 conversion. This effect of glucocorticoids is beneficial in thyroid storm and supports their routine use in this clinical setting.

 

Usually rehydration, repletion of electrolytes, treatment of concomitant disease, such as infection, and specific agents (antithyroid drugs, iodine, propranolol, and corticosteroids) produce a marked improvement within 24 hours. A variety of additional approaches have been reported and may be used if the response to standard treatments is not sufficient. For example, plasmapheresis can remove circulating thyroid hormone and rapidly decrease thyroid hormone levels. Orally administered bile acid sequestrants (20-30g/day Colestipol-HCl or Cholestyramine) can trap thyroid hormone in the intestine and prevent recirculation. In most cases these therapies are not required but in the occasion patient that does not respond rapidly to initial therapy these modalities can be effective. Finally, in rare situations where medical therapy is ineffective, or the patient develops side effects and contraindications to the available therapies’ thyroid surgery may be necessary.

 

FOLLOW-UP

Antithyroid treatment should be continued until euthyroidism is achieved, when a decision regarding definitive treatment of the hyperthyroidism with antithyroid drugs, surgery, or 131-I therapy can be made. Rarely urgent thyroidectomy is performed with antithyroid drugs, iodide, and beta blocker preparation.

 

Prevention of thyroid storm is key and involves recognizing and actively avoiding common precipitants, educating patients about avoiding abrupt discontinuation of anti-thyroid drugs, and ensuring that patients are euthyroid prior to elective surgery and labor and delivery.

 

GUIDELINES

Ross DS, Burch HB, Cooper DS, Greenlee MC, Laurberg P, Maia AL, Rivkees SA, Samuels M, Sosa JA, Stan MN, Walter MA. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid. 2016 Oct;26(10):1343-1421.

 

Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, Tsuboi K, Kanamoto N, Otani H, Furukawa Y, Teramukai S, Akamizu T. 2016 Guidelines for the management of thyroid storm from The Japan Thyroid Association and Japan Endocrine Society (First edition). Endocr J. 2016 Dec 30;63(12):1025-1064

 

REFERENCES

Bartalena L. Graves’ Disease: Complications. 2018 Feb 20. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrère B, Levy M, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000–

 

Akamizu T1, Satoh T, Isozaki O, Suzuki A, Wakino S, Iburi T, Tsuboi K, Monden T, Kouki T, Otani H, Teramukai S, Uehara R, Nakamura Y, Nagai M, Mori M Diagnostic criteria, clinical features, and incidence of thyroid storm based on nationwide surveys. Thyroid. 2012 Jul;22(7):661-79.

Swee du S, Chng CL, Lim A. Clinical characteristics and outcome of thyroid storm: a case series and review of neuropsychiatric derangements in thyrotoxicosis. Endocr Pract. 2015 Feb;21(2):182-9.

Angell TE, Lechner MG, Nguyen CT, Salvato VL, Nicoloff JT, LoPresti JS. Clinical features and hospital outcomes in thyroid storm: a retrospective cohort study. J. Clin. Endocrinol. Metab. 2015 Feb;100(2):451-9.

 

Chiha M, Samarasinghe S, Kabaker AS. Thyroid storm: an updated review. J Intensive Care Med. 2015 Mar;30(3):131-40

 

Akamizu T. Thyroid Storm: A Japanese Perspective. Thyroid. 2018 Jan;28(1):32-40

 

Galindo RJ, Hurtado CR, Pasquel FJ, García Tome R, Peng L, Umpierrez GE. National Trends in Incidence, Mortality, and Clinical Outcomes of Patients Hospitalized for Thyrotoxicosis With and Without Thyroid Storm in the United States, 2004-2013. Thyroid. 2019 Jan;29(1):36-43.

 

Anaerobic Infections and Endocrinology

ABSTRACT

 

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

 

INTRODUCTION

 

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

Figure 1. Types of anaerobic bacteria

 

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

 

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

 

INTERPLAY BETWEEN ANAEROBIC BACTERIA AND HORMONES

 

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

Figure 2. Types of anaerobic bacteria

 

Estrogen and Vaginal Flora

 

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

 

Adrenal Hormone and Anaerobes

 

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

 

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

 

DIABETES AND ANAEROBIC INFECTIONS

 

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

 

Diabetic Foot Disease

 

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

 

Fournier’s Gangrene

 

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

 

Necrotizing Fasciitis

 

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

 

Periodontitis

 

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

 

ORGAN SPECIFIC ANAEROBIC INFECTIONS

 

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

 

Thyroid Gland

 

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

 

Pituitary Gland

 

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

 

Adrenal Gland

 

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

 

INFERTILITY AND ANAEROBIC INFECTIONS

 

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

 

Female Infertility

 

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

 

Male Infertility

 

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

 

GUT ANAEROBES AND METABOLIC DISORDERS

 

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

 

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

 

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

 

ENDOCRINE ISSUES WITH THE ANTIMICROBIALS USED AGAINST ANAEROBES

 

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

 

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

Drug

Endocrine side-effects

Metronidazole

Altered gut microbiome

Anterior pituitary inhibition

Quinolones

Dysglycemia,

Reduced absorption of levothyroxine

Seizures in thyrotoxicosis patients

Beta-lactams

Fractures

Tigecycline

Hypoglycemia

Chloramphenicol

Inhibition of thyroid hormones production

Clindamycin & Carbapenems

Nil

 

CONCLUSION

 

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

 

REFERENCES

 

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

Adrenal Disorders in the Tropics

ABSTRACT

 

The adrenal gland in conjunction with the pituitary gland is one of the major components of the endocrine system and regulates blood volume, blood pressure, serum electrolytes, and stress responses. Dysfunction of the adrenal glands may be related to diseases of the adrenal glands or pituitary gland. Adrenal disorders may present either due to structural or functional abnormalities. In the tropical countries, adrenal insufficiency is primarily due to adrenal infection by tuberculosis, adrenal mycosis infections, and adrenal hemorrhages. HIV (Human immunodeficiency virus) related adrenal problems are also common. Adrenal dysfunction due to pituitary disorders still occur frequently in tropical region and include Sheehan’s syndrome, vasculotoxic snake bite, and thalassemia. Adrenal hormone excess typically occurs secondary to exogenous glucocorticoid use. Adrenal disorders that occur in the developed world occur with similar frequencies in tropical regions.

INTRODUCTION  

Adrenal glands are one of the major peripheral organs necessary for homeostasis including maintenance of blood volume, blood pressure, and serum electrolytes. Disorders of adrenal glands are common in clinical practice. Adrenal dysfunction in tropical countries often occurs due to specific etiologies that differ from the typical causes of adrenal dysfunctions that commonly occur in other parts of the world (Table 1). 

Table 1. Classification of Adrenal Disease in the Tropics

Adrenal insufficiency:

Primary: 

1)     Adrenal Tuberculosis

2)     Adrenal Mycosis

3)     Adrenal Haemorrhage

Secondary:

1)     Sheehan’s Syndrome

2)     Vasculotoxic Snake Bite

3)     Thalassemia’s

Both Primary and Secondary:

1)    HIV

Adrenal Hormone excess syndromes:

1.    Exogenous Glucocorticoid hormone excess syndromes

2.  Licorice induced syndrome of apparent mineralocorticoid excess

PRIMARY ADRENAL INSUFFICIENCY  

The causes of primary adrenal insufficiency that are more frequent in tropical regions include infection of the adrenal glands by tuberculosis or mycotic infections. In addition, autoimmune Addison’s disease or adrenal failure as a component of polyglandular syndromes are equally prevalent in tropical regions as is in other parts of the world. 

Adrenal Gland Tuberculosis

Adrenal gland tuberculosis or Tuberculous adrenalitis is the result of infection of adrenal gland by mycobacterium tuberculosis. The infection causes a destructive lesion of the adrenal cortex with uncertain chances of recovery and remains one of the most important causes of Addison’s disease in the tropical countries (1). In fact, the adrenal glands are the most common endocrine organs to be involved in tuberculosis (2). Adrenal gland tuberculosis occurs almost always secondarily due to the hematogenous spread of the bacilli to the gland with the primary focus in lung. Adrenal failure or Addison’s disease clinically manifest when at least 90% of the gland has been destroyed (1,2,3). Though classically the adrenal cortex is involved, the medulla also may be involved in many cases of adrenal tuberculosis (3,4).

PATHOPHYSIOLOGY 

It is interesting to know why the adrenal glands are susceptible to infections. In fact, adrenal gland infections are common in response to a distant infection elsewhere in the body and in disseminated infection. Autopsy examination revealed that the prevalence of adrenal tuberculosis is about 6% in patients with active tuberculosis (4). However, subclinical adrenal dysfunction may be present in about 60-70% of patients with active tuberculosis (5). In any of these situations, there is an exaggerated response of the hypothalamo-pituitary-adrenal axis to produce excess cortisol in response to the stress of infection. This stress induced hypercortisolemia shifts the balance in the Th1/Th2 cell ratio towards a Th2 response (6). This T cell dysfunction (which is primarily responsible for cell mediated immunity) and low DHEA levels increases the host susceptibility to infection to mycobacterium tuberculosis and other organisms (6). Low DHEAS levels have been documented in tuberculosis (1,6). In addition, endotoxin released in response to the hyperactive HPA axis can cause pathological changes in the adrenal glands to increase the susceptibility to infection (7). The intrinsically rich vascularity of the adrenal glands promotes all of these pathophysiological events.

Histopathologically, four classic patterns have been described in adrenal tuberculosis (3). These are:  granuloma (caseating or non-caseating), enlargement of the gland with destruction by inflammatory granuloma, mass lesions due to cold abscesses, and adrenal atrophy due to fibrosis related to chronic infection. Caseating granuloma is the commonest one and this is identified in about 70% of cases (4). However, granuloma with typical presence of Langhan’s giant cell are less common and identified in less than 50% of cases (4), probably due to anti-inflammatory effects of local glucocorticoids. Calcification of the gland is a common but it is present in other chronic infections of the adrenal glands (3). In about 25 % cases the infection may be unilateral (1).

PRESENTATION

Typical symptoms of adrenal gland tuberculosis in a patient with diagnosed tuberculosis (whether or not on anti-tubercular chemotherapy) are mucocutaneous pigmentation in association with chronic ill health, vomiting, postural hypotension, and anorexia (3). The features are similar to Addison’s disease due to other conditions. As the features of progressively evolving adrenal hypofunction are mostly nonspecific, a high index of suspicion is necessary in subjects with diagnosed active tuberculosis especially when pigmentation is absent. However clinical manifestations may take months to years to become apparent.

The patient may also present rarely with frank adrenal crisis with hypotension, hyponatremia, hyperkalemia, and low serum cortisol levels. The crisis may even be precipitated after administration of rifampicin which increases the hepatic metabolism of cortisol in the background of subclinical adrenal dysfunction (8). 

Adrenal tuberculosis may also present as an adrenal incidentaloma. Nonspecific abdominal pain, weight loss, dizziness, and vomiting may lead to imaging of the abdomen which may reveal an incidental adrenal mass often with calcification. The differential diagnosis of Addison’s disease with adrenal enlargement includes (apart from tuberculosis) malignancy, fungal infections, hemorrhage, amyloidosis, sarcoidosis, etc. (3).

Subclinical adrenal dysfunction is also very common and should be actively sought in all cases of active tuberculosis (5).

INVESTIGATIONS

Laboratory Studies

Common laboratory findings include anemia, hyponatremia, and hyperkalemia. In the presence of a positive Mantoux test in association with typical clinical manifestations of adrenal hypofunction, adrenal tuberculosis must be ruled out. Adrenal insufficiency should be ruled out by using a standard protocol. Serum cortisol levels <5 µg/dL and a plasma ACTH more than 2-fold the upper limit of the reference range is suggestive of primary adrenal insufficiency (9). The serum cortisol may remain in the low-normal to mid-normal range in many cases.  However, a standard dose (250 µg) intravenous cosyntropin (Synacthen) stimulation test establishes the diagnosis of adrenal insufficiency when the peak level of cortisol remains below 18 µg/d (9). Random cortisol levels, though useful during an acute crisis, is not usually sufficient to rule out adrenal insufficiency (9). Documentation of subclinical adrenal dysfunction may reveal mineralocorticoid deficiency alone (as demonstrated by raised plasma rennin activity) when stimulated cortisol is within the normal range (8).

Imaging of Adrenal Glands

CT scan of the abdomen is the most important non-invasive investigation with a very good spatial resolution to diagnose adrenal tuberculosis. The findings are usually bilateral and vary with the duration of the disease before diagnosis (1, 3). The most common early findings during the initial 2 years include a mass lesion with smooth adrenal contour preserved. The glands may show central or patchy hypodensity corresponding to areas of caseous necrosis (3). On contrast administration there is peripheral rim enhancement. Calcification is not a common feature in early tuberculosis (3).

With chronic infection, the adrenal glands become small and shrunken, often with associated calcifications and the margins become irregular (3). Though prevalence and intensity of calcification increases with the duration of tuberculosis, this is not a specific finding and may be associated with other conditions.

Though MRI is also done in many cases, this imaging modality has limitations to assess calcification. However, T1 weighted image shows hypointense or isointense areas and T2 weighted image shows hyperintense areas because of necrosis (3).

Percutaneous FNA/ TB PCR 

 For confirmation of adrenal tuberculosis tissue diagnosis is required. CT scan guided fine needle aspiration from the adrenal gland is necessary to obtain adequate tissue specimens (3, 10). Pathological and microbiological confirmation is necessary, especially where there is isolated adrenal involvement. However, it should be remembered that PCR and culture of these specimens for tuberculosis bacilli are not consistently positive (3). Hence a combination of histopathology, PCR, and culture may be necessary to confirm the diagnosis (3). However, routine search for pulmonary tuberculosis with necessary investigations is mandatory.

TREATMENT

Treatment of adrenal insufficiency in tuberculosis requires administration of both glucocorticoids and mineralocorticoids. As the medulla is frequently involved, patients may require higher doses for maintenance of blood pressure. At the same time, rifampicin used in the anti-tubercular regimen is a potent hepatic enzyme inducer and accelerates cortisol metabolism. This also may necessitate a higher dose of glucocorticoids for adequate treatment. However, aldosterone is less likely to be involved. Adrenal crisis is also reported to occur following the administration of rifampicin (11).

Therapy is monitored with blood pressure, body weight, well-being, serum electrolytes and blood glucose. Patients should be also be monitored for over treatment with glucocorticoids with weight gain, blood pressure, decreasing bone mineral density, and other manifestations of Cushing’s syndrome. All subjects should carry a ‘steroid card’ and should be advised strictly on how to increase the dose of glucocorticoid in stressful situations such as fever, infection, vomiting, trauma, etc.

PROGNOSIS FOR ADRENAL FUNCTION RECOVERY

Chances of adrenal recovery with anti-tuberculosis therapy are uncertain and unpredictable. When the disease is diagnosed late, the glandular destruction is usually significant and the gland becomes atrophic, and anti-tuberculosis therapy does not lead to a recovery of adrenal function (12, 13). If therapy is started early before the gland is destroyed recovery may occur (14, 15). It is also suggested that if the gland size remains the same on subsequent follow up CT scans, it is prudent to follow up the patient for adrenal function recovery.

Adrenal Mycosis

HISTOPLASMOSIS 

Adrenal Histoplasmosis caused by the dimorphic fungus Histoplasma capsulatum, is a recognized cause of adrenal insufficiency. Though this opportunistic pathogen is known to affect immunocompromised individuals predominantly (16), it can rarely infect immunocompetent individuals (16, 17).  This is the most fungal infection of the adrenal glands (16, 18).

Involvement of the adrenals can occur during disseminated infection or many years after disease resolution (18). Adrenal involvement can vary from an asymptomatic milder form to a very severe form that presents with extensive bilateral granulomatous involvement of the entire adrenal gland with calcified lesions culminating in acute adrenal insufficiency (18, 19). Rarely the involvement can be unilateral (17). The common differential diagnosis includes tuberculosis, other fungal infections, adrenal metastasis, primary adrenal malignancy, and primary adrenal lymphoma (16). In immunocompetent individuals it commonly presents with a unilateral or bilateral adrenal mass with constitutional symptoms.

The hypothesis for why histoplasmosis involves the adrenal glands with increased frequency includes the local high levels of glucocorticoids in association with a relative paucity of reticulo-endothelial cells within the adrenal gland (6). The gland is destroyed by direct infection that leads to local ischemia and infarction due to perivasculitis, and caseation (6).

Diagnosis depends on imaging studies with pathological confirmation. CT scan of the adrenal glands typically reveals symmetric enlargement with central hypodensity and characteristic peripheral rim like enhancement (20). Frequently calcification is also present, particularly during the healing phase (20). Percutaneous ultrasound or CT guided fine-needle aspiration or biopsy is necessary for tissue diagnosis (18). The characteristic cytopathological findings are the presence of numerous small oval yeast like structures inside the cytoplasm of macrophages (16). On a necrotic background, this yeast like structures inside the macrophages is surrounded by a clear ring of space resembling a capsule. However, the gold standard for diagnosis is documentation of the organism in the culture of pathological specimen (16). Bhansali et al reported a high uptake in adrenal glands in FDG-PET scan in patients with adrenal histoplasmosis (17). 

Treatment for adrenal histoplasmosis depends on the severity of the infection and the condition of the patient. For severe infection in critically ill patient’s amphotericin B is used initially followed by long-term therapy with oral itraconazole (16). Parenteral liposomal amphotericin B is given 3mg/kg body weight for 2 weeks (17). The duration of therapy with itraconazole varies from six months to two years depending on the patient’s condition. For mild to-moderate histoplasmosis, the recommended treatment is itraconazole. The recommended dose is 200 mg twice daily given for 12 months (16). When itraconazole is used, liver enzymes should be monitored on a regular basis (18).  Treatment for adrenal insufficiency follows the same principles as described earlier.

Though the remission rate from adrenal histoplasmosis is high with long-term oral itraconazole, adrenal insufficiency rarely resolves and reversal of adrenal dysfunction can be seen only in some patients after prolonged antifungal therapy (21).  However, histoplasma in adrenals is reported to persist even 7 years after antifungal therapy (22). 

OTHER FUNGAL INFECTIONS

Paracoccidioidomycosis Brasiliensis

Paracoccidioidomycosis brasiliensis is a dimorphic fungus and can cause chronic, progressive, suppurative and granulomatous disease which can lead to adrenal insufficiency (3). The disease is endemic in Latin America. Humans are the accidental host for the organism and females are rarely affected (23). Smoking and alcohol increase the risk. The lungs are the usual portals of entry. Juvenile forms of the disease are also known (23). Apart from frank adrenal crisis, it can present as progressive constitutional symptoms, hyperpigmentation, and low blood pressure with postural drop and bilateral adrenal enlargement in imaging studies with frank adrenal calcification detected by CT scans (24, 25). Histopathology with GMS stain shows multiple budding yeast with steering wheels appearance which is consistent with Paracoccidioides brasiliensis (24). However, confirmation of the organism by culture material is the gold standard for diagnosis. Serology for antibody detection is also useful in the diagnosis. Diagnosis and treatment of adrenal insufficiency is not different than described above for histoplasmosis. P. brasiliensis primarily causes adrenal destruction by embolic infection of small vessels by large fungal cells and granuloma formation (3). Subjects who receive early antifungals with itraconazole over a 1–2-year period may have a full recovery of adrenal function by preventing fungal embolism in adrenal gland vasculature and reducing ischemic necrotic destruction of the gland (3). Hence an early diagnosis is crucial for preventing the progression of adrenal dysfunction. However, persistence of high antibody titer against paracoccidioidomycosis at the end of treatment or during follow-up is a frequent finding in subjects with paracoccidioidomycosis.

Blastomyces Dermatitidis

Blastomyces dermatitidis is also a dimorphic fungus, which has a strong affinity for the adrenal gland for reasons described earlier. Overt adrenal insufficiency is less common and adrenal Blastomyces dermatitidis typically presents as bilateral adrenal incidentaloma during radiological investigations for other reasons (3). The portal of entry is through the lungs and when there is lymphohematogenous dissemination the disease spreads to other organs (26). In situations when it presents as adrenal insufficiency, the presentation, investigations, and management are similar to those described above. Diagnosis is by fine-needle aspiration guided by ultrasound or CT scan followed by cytologic and histologic examinations. However, the gold standard is fungal culture showing thick-walled, broad-based budding yeast cells (27). Treatment is with long term oral itraconazole. In patients with severe manifestations initial treatment with liposomal amphotericin B for 2 weeks could be used.

Cryptocoocus Neoformans

Cryptocoocus neoformans is an encapsulated yeast-like fungus which infects primarily immunodeficient hosts, particularly subjects infected with HIV or lymphohematogenous malignancies (28). In immunocompromised hosts it usually affects the central nervous system and lungs.  However immune-competent individuals may also suffer adrenal cryptococcosis (29). Adrenal dysfunction is uncommon until almost the whole of adrenal gland is infiltrated with C. neoformans and caseating granulomas. Cryptococcosis is diagnosed by fine-needle aspiration biopsy of the adrenal mass. The serum cryptococcal antigen titer is highly elevated. Treatment is with antifungal therapy with fluconazole and amphotericin B. Adrenal enlargement by Cryptococcus may be completely reversible without any abnormality after antifungal treatment (30). Cases not responsive to anti-fungal therapy have been reported to improve after unilateral or bilateral adrenalectomy (28, 29).

Miscellaneous

Pneumocystis jirovecii (previously known P. carinii) occurs in individuals with advanced HIV due to defects in cell mediated immunity. Spread to other organs including the adrenal glands is also possible (3). Adrenal failure associated with coccidioidomycosis and rarely candidiasis has also been reported.

Adrenal Hemorrhage; the Waterhouse Friderichsen Syndrome

This is a condition in which patient presents with acute hypotension and shock due to adrenal insufficiency arising from acute adrenal hemorrhage. The syndrome is typically related to infection with Neisseria meningitides infection (3). However, this is also known to occur in septicemia due to infections with Staphylococcus aureus, Streptococcus spp, Haemophilus influenzae, Corynebacterium diphtheria, etc. (3). Hence this is more common in the tropical region.  The condition is hypothesized to be due to interplay between endotoxemia and elevated ACTH. The adrenal gland is anatomically prone to hemorrhage as it has three separate arterial supplies and does not have proportional venous drainage (3). In endotoxemia, elevated ACTH increases the blood supply several fold in this compromised anatomical setting. At the same time increased adrenaline secretion in relation to stress leads to constriction of adrenal veins, which further increases this imbalance between arterial supply and venous drainage. Management includes immediate fluid replacement and parenteral glucocorticoids apart from the management of the underlying infection.

SECONDARY ADRENAL INSUFFICIENCY 

Adrenal insufficiency secondary to disorders of pituitary gland is also very common in developing countries in tropical regions.  Secondary adrenal insufficiency caused by pituitary tumors and apoplexy, pituitary surgery, radiation therapy, hypophysitis, various genetic disorders, and withdrawal of exogenous steroids are equally common in tropical regions but certain other disorders like Sheehan’s syndrome, thalassemia, and vasculotoxic snake bite induced pituitary failure are more common in tropical regions.

Sheehan’s Syndrome 

Sheehan’s syndrome consists of various degrees of pituitary insufficiency, which develops as a result of ischemic pituitary necrosis due to severe postpartum hemorrhage. The important pathogenetic/predisposing factors include a small sella, increased pituitary volume, vasospasm induced by postpartum hemorrhage, thrombosis, and probable pituitary autoimmunity (31). In developed countries there has been a drastic reduction in the incidence of Sheehan’s syndrome. This is primarily due to the remarkable improvement in obstetric care and availability of rapid blood transfusion. However, this remains as a major cause of hypopituitarism in the other parts of the world.

CLINICAL FEATURES

Most commonly the disorder presents as a lactation failure in the post-partum state and non-resumption of menses following child birth, which was complicated by massive post-partum hemorrhage leading to hypotension and shock. However, it may very rarely occur without massive bleeding or after normal delivery. Patients may present in the emergency with altered sensorium, loss of consciousness, seizure, shock, intractable vomiting, or more commonly with chronic complaints like asthenia and weakness, dizziness, anorexia, weight loss, nausea, and vomiting with a typical history of failure to resume menses and lactation failure following child birth (31). Apart from anterior pituitary hormone deficiency, symptoms like anemia, pancytopenia, osteoporosis, cognitive impairment, and poor quality of life are also present in these patients (31,32).  Very rarely diabetes insipidus may occur. However, the mean age of the participants may be as late as 40 years or more and the mean interval between inciting event to diagnosis may be as high as 10 years or more (33).

Adrenal insufficiency due to ACTH deficiency is reported to occur in up to 100% of cases (in fact deficiency of all anterior pituitary hormones occur in a variable percentage of patients and may be up to 100%) (32). Weakness, fatigue, and postural drop are common manifestations. Hyponatremia is particularly common in Sheehan’s syndrome, which may be due to glucocorticoids deficiency coupled with increased AVP release as a consequence of reduced blood pressure and cardiac output resulting from glucocorticoid deficiency (32).

DIAGNOSIS

The basal pituitary hormonal levels and those after dynamic tests are beyond the purview of this chapter. However adrenal insufficiency is diagnosed with a morning cortisol level of 3 mcg/dl with low or inappropriately normal ACTH or a cosyntropin stimulated cortisol level <18 mcg/dl. Documentation of growth hormone deficiency does not require a dynamic test in presence of other pituitary hormone deficiencies. Only low age specific and assay specific IGF-1 assay may be sufficient to document adult growth hormone deficiency (AGHD) (34).

The preferred radiological imag­ing is an MRI of hypothalamic pituitary area.  CT scan may also be helpful. MRI findings in Sheehan’s syndrome usually vary with the stages of the disease. In earlier stages of the disease there may be an enlarged pituitary gland with central hypodensity (suggestive of infarction). However, an empty sella (complete or partial) is considered to be a characteristic of Sheehan’s syndrome in established cases (32).

TREATMENT

The acute adrenal crisis in Sheehan’s syndrome is treated with intravenous glucocorticoids. In other patients’ glucocorticoids should be started orally with hydrocortisone 15-25 mg daily in 2-3 divided doses with the higher dose in the morning and a lower dose in the evening (35). Mineralocorticoids are not necessary in general (35). Once daily prednisolone may also be used at a dose of 2.5-5 mg once daily in the early morning. As GH deficiency decreases cortisol clearance, it may necessary to increase the dose of glucocorticoid for those who receive GH treatment (35). Therapy is monitored with blood pressure, body weight, well-being, serum electrolytes, and blood glucose. Patients should be also be monitored for an overdose of glucocorticoids with weight gain, blood pressure, decreased bone mineral density, and other symptoms and signs of Cushing’s syndrome. All subjects with Sheehan’s syndrome should carry a ‘steroid card’ and should be advised strictly on how to increase the dose of glucocorticoid in stressful situation such as fever, infection, vomiting, trauma, etc.

Subjects with Sheehan’s syndrome should also be treated with levothyroxine, combined oral contraceptives according to guideline, calcium and vitamin D supplements, and growth hormone therapy (if possible) according to the protocol of adult growth hormone deficiency.

Viscerotropic Snake Bite

Snakebite is a major public health problem in tropical regions and is considered as one of the most neglected tropical diseases. The development of a Sheehan-like syndrome with chronic hypopituitarism following Russell viper envenomation is fairly common. Hypoadrenalism due to ACTH deficiency is the commonest abnormality (36). However acute hypopituitarism with predominant glucocorticoids deficiency has also been reported (37).

The venom of vipers is vasculotoxic in nature and the clinical features of viper venomation include local cellulitis and tissue necrosis, bleeding manifestations, disseminated intravascular coagulation, shock, and acute kidney injury (AKI) (38). Hypopituitarism is particularly common following vasculotoxic snake bite in subjects who develop AKI requiring hemodialysis. Hypopituitarism can develop as early as 7 days following snake bites and should be evaluated for particularly in younger subjects, especially those requiring increasing number of sessions of hemodialysis and in subjects with abnormal 20 min WBCT (whole blood clotting test) at presentation (36,39). On the other hand, the time of onset/presentation of hypopituitarism following snake bite may be as long as up to 24 years (40). Acute hypopituitarism is thought to occur due to acute damage to the pituitary gland at the time of the precipitating event, but a gradual/slower progression of pituitary damage may occur over years due to other unknown mechanisms (36).

Those who survive acute snake bite may later present with altered sensorium, loss of consciousness, seizure, shock, intractable vomiting, or more commonly with chronic complaints like asthenia and weakness, dizziness, anorexia, weight loss, nausea, vomiting and amenorrhea in females (36).

Variable degrees of hypopituitarism may be present. Cortisol deficiency is reported to be the commonest abnormality. Secondary adrenal insufficiency is diagnosed with a morning cortisol level of 3 mcg/dl with low or inappropriately normal ACTH or a co-syntropin stimulated cortisol level <18 mcg/dl (36). Documentation of growth hormone deficiency is done as mentioned in section of Sheehan’s Syndrome (34).

The preferred radiological imag­ing is the MRI of hypothalamic pituitary area which may show partial or complete empty sella or evidences of old hemorrhage. However, these changes are not present in all cases (41).

Treatment of secondary adrenal insufficiency and other hormone deficiencies are similar to described above. All subjects with hypopituitarism on glucocorticoids supplements should carry a ‘steroid card’ and should be advised on how to increase the dose of glucocorticoid in stressful situation such as fever, infection, vomiting, trauma, etc.

Thalassemia Major

Thalassemia’s are inherited autosomal recessive disorders of hemoglobin synthesis. Thalassemia major is the most severe form of beta thalassemia which involves the beta chain of hemoglobin. Organ dysfunction in thalassemia is principally attributed to excessive iron overload and suboptimal chelation. The precise underlying mechanism of iron overload induced organ dysfunction is not very unclear. The current management of thalassemia includes regular transfusion programs and chelation therapy. Pre-marital counselling and assessment with HPLC to assess the asymptomatic carrier has reduced its prevalence significantly in the developed world. However, this is still a major problem in many parts of the world.  Prevalence of adrenal insufficiency is variable and depends on the severity of iron overload. This secondary hemochromatosis can disrupt adrenal function by affecting the hypothalamic-pituitary-adrenal axis at the hypothalamic or pituitary level (42). In more severe cases primary adrenal failure may supervene due to iron deposition in the adrenal glands (42). Additionally, an extramedullary hematopoietic tumor has been reported in HbE thalassemia and beta thalassemia as non-hormone secretory unilateral or bilateral adrenal enlargement resembling adrenal myelolipoma (43). 

Biochemical adrenal insufficiency is reported to occur from   0% to 45% of subjects with thalassemia major (42), but adrenal crisis or clinical adrenal insufficiency is extremely uncommon and mostly they are asymptomatic. However, subclinical cortisol deficiency is not uncommon. In this context it should be remembered that mild symptoms of adrenal insufficiency like asthenia, weight loss, or postural drops are frequently overlooked as these features are common in thalassemia subjects with low levels of hemoglobin (42).                           

The unique finding in subjects with thalassemia is the dissociation between adrenal androgen levels with cortisol and aldosterone levels. This paradox is reflected by frequent documentation of low serum DHEA, DHEA-sulfate, androstenedione, and testosterone levels in the presence of normal serum cortisol and aldosterone levels (44). Absence of adrenarche occurring in most adolescents with thalassemia major is probably explained by this phenomenon (45). 

Diagnosis of adrenal dysfunction in thalassemia is similar to other causes of secondary adrenal insufficiency. If the morning cortisol is not unequivocally low, synacthen stimulation test should be done with either the low dose (1 µg) or the standard high dose (250 µg). A peak cortisol level of >18 µg/dL after 30-60 min of intravenous synacthen excludes adrenal insufficiency. Alternately an insulin tolerance test with a similar cut-off may also be done.

Treatment of clinical adrenal insufficiency is similar to that described above. Subjects with subclinical adrenal insufficiency require only steroid coverage during periods of stress.

HIV AND ADRENAL DYSFUNCTION

Endocrine manifestations of HIV infection may include adrenal dysfunction, hypothyroidism, hypogonadism, insulin resistance and diabetes etc. Changes in the HPA (hypothalamic-pituitary-adrenal) axis are the most frequent abnormality (46). Adrenal dysfunction in HIV infection may be a consequence of concomitant systemic illness, opportunistic infections, and neoplasm (47).

Probably the most frequent adrenal abnormality is a stress induced elevation in serum cortisol and ACTH (46). This may be due to activation of the HPA axis due to HIV infection itself or pro-inflammatory cytokines (e.g., IL-1β, IL-6 and TNF-α) (46). Alternately a peripheral increase in the conversion of cortisone to cortisol due to activation of 11-β HSD type 1 in adipose tissue or decrease in cortisol metabolism may be responsible for increased cortisol with subnormal ACTH (46). Tissue hypersensitivity to glucocorticoids is also reported in subjects with HIV-1 infection, which may result in hippocampal atrophy, altered secretion of cytokine/interleukins, etc. (48).

On the other hand, subclinical or clinical adrenal dysfunction can happen in about 10-20% of subjects with advanced disease and multiple co-morbidities when about 80-90% of the gland is destroyed (46). The involvement and destruction by HIV, opportunistic infections, or malignancies in the adrenal glands or the hypothalamus and/or pituitary area can result in either primary or secondary adrenal sufficiency (47).

The opportunistic infections include cytomegalovirus (CMV), Mycobacterium avium-intracellular and M. tuberculosis, fungal infections (such as Histoplasma, Cryptococcus, and Pneumocystis jirovecii), and Toxoplasma gondii (47). Of these opportunistic infections, CMV infection is known to be the commonest etiology with earlier literature reporting Cytomegalovirus adrenalitis in nearly 80 % of cases of HIV infection (46). However, due to improvements in active management of HIV by HAART (highly active anti- retroviral therapy), the prevalence of adrenal insufficiency has decreased over the last two decades.

Medications used for the treatment of HIV infection and its complication may also result in adrenal dysfunction. For example:  Rifampicin used for mycobacterial infection is a known hepatic Cytochrome P 450 (CYP) enzyme inducer and can lower serum cortisol levels by enhanced cortisol metabolism. Ketoconazole used to treat severe mycotic infections inhibits adrenal steroid synthesis and can lead to glucocorticoid deficiency or even adrenal crisis in patients with impaired adrenal reserve (49). Interestingly, ART-related lipodystrophy (dorsocervical fat pad enlargement and visceral adiposity) may mimic Cushing’s syndrome but it is typically not associated with hypercortisolism (49). On the contrary, some protease inhibitors (e.g., ritonavir) used in ART are reported to decrease metabolism of endogenous and exogenously co-administered glucocorticoids, resulting in an iatrogenic Cushing's syndrome.

Tumors of the adrenal gland in HIV infected patients include Kaposi’s sarcoma and high-grade non-Hodgkin’s lymphoma. Kaposi’s sarcoma is secondary to co-infection with the oncogenic human herpes virus type 8 (HHV8) and non-Hodgkin’s lymphoma could be secondary to Epstein-Barr virus (EBV).

Assessment for symptoms of adrenal involvement requires a high degree of suspicion as constitutional symptoms of HIV may mask the features of adrenal insufficiency.  Morning serum cortisol should be done in all cases suspected for adrenal dysfunction. Stress induced hypercortisolemia does not require any further testing and low serum cortisol <5 μg/dl with an elevated ACTH level requires treatment with glucocorticoids and mineralocorticoids. In other cases, synthacthen stimulated cortisol is used to determine the course of treatment. Stimulated cortisol <18 μg/dl, especially if associated with elevated plasma ACTH, should be treated as adrenal insufficiency. Asymptomatic subjects with stimulated serum cortisol <18 μg/dl should be advised to take stress doses of glucocorticoids only as mentioned before.

Diagnosis and management of adrenal disorders in a patient with HIV infection does not differ from that in immunocompetent persons in general.

ADRENAL HORMONE EXCESS SYNDROMES

Glucocorticoid Excess Syndromes 

The primary cause of Cushing’s syndrome, more common in tropical regions, is exogenous glucocorticoids. The background etiology for exogenous steroid usage includes: nephrotic syndrome, rheumatoid arthritis and other collagen vascular disease, bronchial asthma, Graves’ orbitopathy, etc.  Glucocorticoids used as inhalational agent for bronchial asthma, in creams and ointments for eczematous skin lesions may also be responsible. Endogenous steroid excess (Cushing’s disease, ectopic ACTH syndromes, adrenal tumors) are equally common in tropical regions as in other areas of the world.

Often it is a challenge to suspect exogenous glucocorticoid use based on the patient’s history, especially in situations when glucocorticoids were not being used for a therapeutic purpose. Subjects presenting with features suggestive of Cushing’s syndrome should therefore mandatorily undergo testing for basal morning cortisol (with paired ACTH if possible) to rule out exogenous glucocorticoid use. A suppressed morning cortisol and plasma ACTH strongly suggests the diagnosis (50). One important caveat is that prednisolone may cross react with some cortisol assays giving false positive results in some chemiluminescent assay (51). Additionally, if the patient is receiving hydrocortisone, the result will also be fallacious to interpret. It is not uncommon in tropical regions that some form of glucocorticoids is being used in disguise as an alternative medicine for joint pain, respiratory problems, fever, or even as a weight gain therapy for young lean subjects. Hence a more detailed evaluation of the history with leading questions and scrutiny of all past records of medicine, including that of the alternative medicines, may sometimes reveal the offending agent. 

The clinical features that suggest exogenous Cushing’s syndrome are lack of pigmentation and the absence of hypertension and hirsutism (as exogenous Cushing’s syndrome does not contain mineralocorticoids and androgens as opposed to endogenous Cushing’s syndrome). Patients with exogenous Cushing’s syndrome are prone to develop glaucoma, osteoporosis, psychiatric disturbances, etc. (50).

Once diagnosed, these subjects should be advised to withdraw the offending agents and should be given hydrocortisone in the lowest possible dose for preventing adrenal crisis. The withdrawal of hydrocortisone subsequently after 3 months depends on the morning cortisol, after stopping the previous evening dose and subjecting the patient to short synacthen test to assess the recovery of HPA axis. Those with morning cortisol between 5 -18 µ/dl should be advised stress coverage only. For bone protection, all subjects with exogenous Cushing’s syndrome should receive bisphosphonate therapy unless contraindicated (52). Adequate calcium supplements with cholecalciferol should also be used.

For subjects receiving glucocorticoids for therapeutic purpose, it is essential to maintain bone protection, check for secondary diabetes and hypertension, and prevent gastric ulceration. Withdrawal (if at all possible) should be performed very slowly. When the therapeutic steroid reaches the lowest possible dose to prevent crisis, it is converted to equivalent dose of hydrocortisone and the same principle is used as described before.

Licorice Induced Syndrome Of Apparent Mineralocorticoid Excess 

Licorice root extracts are used as a herbal medicine for several conditions like cough, peptic ulceration, etc. Licorice is also used as a sweetener and mouth freshener particularly in tropical regions (53). Licorice possesses some glucocorticoid activity, antiandrogen effect, estrogenic activity, and mineralocorticoid like activity. Subjects consuming excessive licorice may develop hypertension and hypokalemia (53). Sometimes this is severe enough to cause a cardiac arrhythmia. While screening for primary aldosteronism for subjects presenting with hypertension and hypokalemia, plasma aldosterone and plasma rennin activity are found to be suppressed in patients using licorice (53). 

                                                                                                                                                                           The active ingredient of liquorice is glycyrrhizic acid, which is hydrolyzed into glycyrrhetinic acid in vivo. Glycyrrhetinic acid has a very low affinity for the mineralocorticoid receptor but is a potent competitive inhibitor of the enzyme 11β-HSD type 2 which is preferentially expressed in kidney (54). Hence it may cause acquired 11β-HSD type 2 deficiency. The physiological role of the enzyme 11β-HSD type 2 is to inactivate cortisol to cortisone and thereby preventing access of cortisol to mineralocorticoid receptor. Cortisol and aldosterone have equipotent stimulating activity on mineralocorticoid receptor (54). Hence any situation associated with suppressed 11β-HSD type 2 activities may lead to overstimulation of mineralocorticoid receptors by cortisol, leading to hypertension with hypokalemia and metabolic alkalosis. After correction of hypokalemia, the screening test reveals suppressed aldosterone and plasma rennin activity (54). The hypertension is primarily due to sodium and water retention. A careful history for licorice ingestion clinches the diagnosis.

Treatment consists of avoidance of licorice products. In the interim period patients should be treated with oral potassium and spironolactone after the completion of screening of aldosterone rennin ratio (ARR). Withdrawal of licorice, even after prolonged use or ingestion of large amounts, leads to a complete resolution of the symptoms of acquired apparent mineralocorticoid excess (55).

REFERENCES

  1. Upadhyay J, Sudhindra P, Abraham G, Trivedi N. Tuberculosis of the adrenal gland: a case report and review of the literature of infections of the adrenal gland. Int J Endocrinol. 2014;2014:876037. doi: 10.1155/2014/876037. Epub 2014 Aug 6. PMID: 25165474; PMCID: PMC4138934.
  2. Kelestimur F. The endocrinology of adrenal tuberculosis: the effects of tuberculosis on the hypothalamo-pituitary-adrenal axis and adrenocortical function. J Endocrinol Invest. 2004 Apr;27(4):380-6. doi: 10.1007/BF03351067. PMID: 15233561.
  3. Vinnard C, Blumberg EA. Endocrine and Metabolic Aspects of Tuberculosis. Microbiol Spectr. 2017 Jan;5(1):10.1128/microbiolspec.TNMI7-0035-2016. doi: 10.1128/microbiolspec.TNMI7-0035-2016. PMID: 28233510; PMCID: PMC5785104.
  4. Lam KY, Lo CY. A critical examination of adrenal tuberculosis and a 28-year autopsy experience of active tuberculosis. Clin Endocrinol (Oxf). 2001 May;54(5):633-9. doi: 10.1046/j.1365-2265.2001.01266.x. PMID: 11380494.
  5. Neogi S, Mukhopadhyay P, Sarkar N, Datta PK, Basu M, Ghosh S. Overt and Subclinical Adrenal Insufficiency in Pulmonary Tuberculosis. Endocr Pract. 2020 Dec 14:S1530-891X(20)48391-0. doi: 10.1016/j.eprac.2020.11.012. Epub ahead of print. PMID: 33645514.
  6. Paolo WF Jr, Nosanchuk JD. Adrenal infections. Int J Infect Dis. 2006 Sep;10(5):343-53. doi: 10.1016/j.ijid.2005.08.001. Epub 2006 Feb 17. PMID: 16483815; PMCID: PMC7110804.
  7. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo-pituitary-adrenal (HPA) axis. J Endotoxin Res. 2003;9(1):3-24. doi: 10.1179/096805103125001298. PMID: 12691614.
  8. Denny N, Raghunath S, Bhatia P, Abdelaziz M. Rifampicin-induced adrenal crisis in a patient with tuberculosis: a therapeutic challenge. BMJ Case Rep. 2016 Nov 29;2016:bcr2016216302. doi: 10.1136/bcr-2016-216302. PMID: 27899384; PMCID: PMC5175016.
  9. Stefan R. Bornstein, Bruno Allolio, Wiebke Arlt, Andreas Barthel, Andrew Don-Wauchope, Gary D. Hammer, Eystein S. Husebye, Deborah P. Merke, M. Hassan Murad, Constantine A. Stratakis, David J. Torpy, Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline, The Journal of Clinical Endocrinology & Metabolism, Volume 101, Issue 2, 1 February 2016, Pages 364–389, https://doi.org/10.1210/jc.2015-1710
  10. Liatsikos EN, Kalogeropoulou CP, Papathanassiou Z, Tsota I, Athanasopoulos A, Perimenis P, Barbalias GA, Petsas T. Primary adrenal tuberculosis: role of computed tomography and CT-guided biopsy in diagnosis. Urol Int. 2006;76(3):285-7. doi: 10.1159/000091637. PMID: 16601397.
  11. Kyriazopoulou V, Parparousi O, Vagenakis AG. Rifampicin-induced adrenal crisis in addisonian patients receiving corticosteroid replacement therapy. J Clin Endocrinol Metab. 1984 Dec; 59(6):1204-6. doi: 10.1210/jcem-59-6-1204. PMID: 6490796.
  12. Bhatia E, Jain SK, Gupta RK, Pandey R. Tuberculous Addison's disease: lack of normalization of adrenocortical function after anti-tuberculous chemotherapy. Clin Endocrinol (Oxf). 1998 Mar;48(3):355-9. doi: 10.1046/j.1365-2265.1998.00409.x. PMID: 9578827.
  13. Laway, B.A., Mir, S.A., Ganie, M.A. et al. Nonreversal of adrenal hypofunction after treatment of adrenal tuberculosis. Egypt J Intern Med 27, 42–44 (2015). https://doi.org/10.4103/1110-7782.155860
  14. Penrice J, Nussey SS. Recovery of adrenocortical function following treatment of tuberculous Addison's disease. Postgrad Med J. 1992 Mar; 68(797):204-5. doi: 10.1136/pgmj.68.797.204. PMID: 1589379; PMCID: PMC2399240.
  15. Kelestimur F. Recovery of adrenocortical function following treatment of tuberculous Addison's disease. Postgrad Med J (1993) 69, 832-34
  16. Roxas MCA, Sandoval MAS, Salamat MS, Matias PJ, Cabal NP, Bartolo SS. Bilateral adrenal histoplasmosis presenting as adrenal insufficiency in an immunocompetent host in the Philippines. BMJ Case Rep. 2020 May 12;13(5):e234935. doi: 10.1136/bcr-2020-234935. PMID: 32404324; PMCID: PMC7228487.
  17. Bhansali A, Das S, Dutta P, Walia R, Nahar U, Singh SK, Vellayutham P, Gopal S. Adrenal histoplasmosis: unusual presentations. J Assoc Physicians India. 2012 Oct;60:54-8. PMID: 23777028.
  18. Jayathilake WAPP, Kumarihamy KWMPP, Ralapanawa DMPUK, Jayalath WATA, "A Rare Presentation of Possible Disseminated Histoplasmosis with Adrenal Insufficiency Leading to Adrenal Crisis in an Immunocompetent Adult: A Case Report", Case Reports in Medicine, vol. 2020, Article ID 8506746, 5 pages, 2020. https://doi.org/10.1155/2020/8506746
  19. Vyas S, Kalra N, Das PJ, Lal A, Radhika S, Bhansali A, Khandelwal N. Adrenal histoplasmosis: An unusual cause of adrenomegaly. Indian J Nephrol. 2011 Oct;21(4):283-5. doi: 10.4103/0971-4065.78071. PMID: 22022092; PMCID: PMC3193675.
  20. Mukherjee JJ, Villa ML, Tan L, Lee KO. Bilateral adrenal masses due to histoplasmosis. J Clin Endocrinol Metab. 2005 Dec; 90(12):6725-6. doi: 10.1210/jc.2005-1868. PMID: 16330806.
  21. Robinson LJ, Lu M, Elsayed S, Joy TR. Bilateral adrenal histoplasmosis manifesting as primary adrenal insufficiency. CMAJ. 2019 Nov 4;191(44):E1217-E1221. doi: 10.1503/cmaj.190710. PMID: 31685665; PMCID: PMC6834444.
  22. Kothari D, Chopra S, Bhardwaj M, Ajmani AK, Kulshreshtha B. Persistence of histoplasma in adrenals 7 years after antifungal therapy. Indian J Endocrinol Metab. 2013 May;17(3):529-31. doi: 10.4103/2230-8210.111679. PMID: 23869317; PMCID: PMC3712391.
  23. de Oliveira FM, Fragoso MCBV, Meneses AF, Vilela LAP, Almeida MQ, Palhares RB, de Arruda Mattos TV, Scalissi NM, Viana Lima J. Adrenal insufficiency caused by Paracoccidioidomycosis: three case reports and review. AACE Clin Case Rep. 2019 Mar 13;5(4):e238-e243. doi: 10.4158/ACCR-2018-0632. PMID: 31967043; PMCID: PMC6873835.
  24. Cataño J, Porras J. Adrenal Paracoccidioidomycosis. Am J Trop Med Hyg. 2020 Sep;103(3):935-936. doi: 10.4269/ajtmh.20-0083. PMID: 32896237; PMCID: PMC7470546.
  25. Tobón AM, Agudelo CA, Restrepo CA, Villa CA, Quiceno W, Estrada S, Restrepo A. Adrenal function status in patients with paracoccidioidomycosis after prolonged post-therapy follow-up. Am J Trop Med Hyg. 2010 Jul; 83(1):111-4. doi: 10.4269/ajtmh.2010.09-0634. PMID: 20595488; PMCID: PMC2912586.
  26. Kumar A, Sreehari S, Velayudhan K, Biswas L, Babu R, Ahmed S, Sharma N, Kurupath VP, Jojo A, Dinesh KR, Karim S, Biswas R. Autochthonous blastomycosis of the adrenal: first case report from Asia. Am J Trop Med Hyg. 2014 Apr;90(4):735-9. doi: 10.4269/ajtmh.13-0444. Epub 2014 Feb 3. PMID: 24493676; PMCID: PMC3973522.
  27. Rimondi AP, Bianchini E, Barucchello G, Panzavolta R. Addison's disease caused by adrenal blastomycosis: a case report with fine needle aspiration (FNA) cytology. Cytopathology. 1995 Aug;6(4):277-9. doi: 10.1111/j.1365-2303.1995.tb00480.x. PMID: 8520008.
  28. Matsuda Y, Kawate H, Okishige Y, Abe I, Adachi M, Ohnaka K, Satoh N, Inokuchi J, Tatsugami K, Naito S, Nomura M, Takayanagi R. Successful management of cryptococcosis of the bilateral adrenal glands and liver by unilateral adrenalectomy with antifungal agents: a case report. BMC Infect Dis. 2011 Dec 14; 11:340. doi: 10.1186/1471-2334-11-340. PMID: 22166121; PMCID: PMC3254187.
  29. Ito M, Hinata T, Tamura K, Koga A, Ito T, Fujii H, Hirata F, Sakuta H. Disseminated Cryptococcosis with Adrenal Insufficiency and Meningitis in an Immunocompetent Individual. Intern Med. 2017; 56(10):1259-1264. doi: 10.2169/internalmedicine.56.7356. Epub 2017 May 15. PMID: 28502948; PMCID: PMC5491828.
  30. Muraoka Y, Iwama S, Arima H. Normalization of Bilateral Adrenal Gland Enlargement after Treatment for Cryptococcosis. Case Rep Endocrinol. 2017; 2017:1543149. doi: 10.1155/2017/1543149. Epub 2017 Mar 26. PMID: 28458934; PMCID: PMC5385225.
  31. Keleştimur F. Sheehan's syndrome. Pituitary. 2003;6(4):181-8. doi: 10.1023/b:pitu.0000023425.20854.8e. PMID: 15237929.
  32. Karaca Z, Laway BA, Dokmetas HS, Atmaca H, Kelestimur F. Sheehan syndrome. Nat Rev Dis Primers. 2016 Dec 22; 2:16092. doi: 10.1038/nrdp.2016.92. PMID: 28004764.
  33. Mandal S, Mukhopadhyay P, Banerjee M, Ghosh S. Clinical, endocrine, metabolic profile, and bone health in Sheehan’s syndrome. Indian J Endocr Metab 2020; 24:338-42.
  34. Hartman ML, Crowe BJ, Biller BM, Ho KK, Clemmons DR, Chipman JJ; HyposCCS Advisory Board; U.S. HypoCCS Study Group. Which patients do not require a GH stimulation test for the diagnosis of adult GH deficiency? J Clin Endocrinol Metab. 2002 Feb; 87(2):477-85. doi: 10.1210/jcem.87.2.8216. PMID: 11836272.
  35. Kilicli F, Dokmetas HS, Acibucu F. Sheehan's syndrome. Gynecol Endocrinol. 2013 Apr;29(4):292-5. doi: 10.3109/09513590.2012.752454. Epub 2012 Dec 18. PMID: 23245206.
  36. Bhat S, Mukhopadhyay P, Raychaudhury A, Chowdhury S, Ghosh S. Predictors of hypopituitarism due to vasculotoxic snake bite with acute kidney injury. Pituitary. 2019 Dec; 22(6):594-600. doi: 10.1007/s11102-019-00990-8. PMID: 31556012
  37. Rajagopala S, Thabah MM, Ariga KK, Gopalakrishnan M. Acute hypopituitarism complicating Russell's viper envenomation: case series and systematic review. QJM. 2015 Sep;108(9):719-28. doi: 10.1093/qjmed/hcv011. Epub 2015 Jan 27. PMID: 25630907.
  38. Shivaprasad C, Aiswarya Y, Sridevi A, Anupam B, Amit G, Rakesh B, Annie PA, Anish K. Delayed hypopituitarism following Russell's viper envenomation: a case series and literature review. Pituitary. 2019 Feb; 22(1):4-12. doi: 10.1007/s11102-018-0915-1. PMID: 30317419.
  39. Benjamin JM, Chippaux JP, Sambo BT, Massougbodji A. Delayed double reading of whole blood clotting test (WBCT) results at 20 and 30 minutes enhances diagnosis and treatment of viper envenomation. J Venom Anim Toxins Incl Trop Dis. 2018 May 16; 24:14. doi: 10.1186/s40409-018-0151-1. PMID: 29796013; PMCID: PMC5956810
  40. Tun-Pe, Phillips RE, Warrell DA, Moore RA, Tin-Nu-Swe, Myint-Lwin, Burke CW. Acute and chronic pituitary failure resembling Sheehan's syndrome following bites by Russell's viper in Burma. Lancet. 1987 Oct 3;2(8562):763-7. doi: 10.1016/s0140-6736(87)92500-1. PMID: 2888987.
  41. Naik BN, Bhalla A, Sharma N, Mokta J, Singh S, Gupta P, Rai A, Subbiah S, Bhansali A, Dutta P (2018) Pituitary dysfunction in survivors of Russell’s viper snake bite envenomation: A prospective study. Neurol India 66(5):1351
  42. De Sanctis V, Soliman AT, Elsedfy H, Skordis N, Kattamis C, Angastiniotis M, Karimi M, Yassin MA, El Awwa A, Stoeva I, Raiola G, Galati MC, Bedair EM, Fiscina B, El Kholy M. Growth and endocrine disorders in thalassemia: The international network on endocrine complications in thalassemia (I-CET) position statement and guidelines. Indian J Endocrinol Metab. 2013 Jan;17(1):8-18. doi: 10.4103/2230-8210.107808. PMID: 23776848; PMCID: PMC3659911.
  43. Saraogi RK, Chowdhury S, Mukherjee S, Roy S, Chatterjee P. Adrenal extramedullary haematopoietic tumor in HbE Thalassemia. BMJ 2002; 18(7). South Asia Edition.
  44. Tiosano D, Hochberg Z. Endocrine complications of thalassemia. J Endocrinol Invest. 2001 Oct; 24(9):716-23. doi: 10.1007/BF03343916. PMID: 11716158.
  45. De P, Mistry R, Wright C, Pancham S, Burbridge W, Gangopadhayay K, Pang T, Das D. A Review of Endocrine Disorders in Thalassaemia. OJEMD 2014; 4(2).
  46. Bhatia E. Adrenal disorders in people with HIV: The highs and lows. Indian J Med Res. 2018 Feb; 147(2):125-127. doi: 10.4103/ijmr.IJMR_1087_17. PMID: 29806599; PMCID: PMC5991116.
  47. Eledrisi MS, Verghese AC. Adrenal insufficiency in HIV infection: a review and recommendations. Am J Med Sci. 2001 Feb; 321(2):137-44. doi: 10.1097/00000441-200102000-00005. PMID: 11217816.
  48. Bakari Adamu Girei, Sani-Bello Fatima. Endocrine Manifestations of HIV Infection.2013. http://dx.doi.org/10.5772/52684
  49. Unachukwu CN, Uchenna DI, Young EE. Endocrine and metabolic disorders associated with human immune deficiency virus infection. West Afr J Med. 2009 Jan;28(1):3-9. doi: 10.4314/wajm.v28i1.48415. PMID: 19662737.
  50. Hopkins RL, Leinung MC. Exogenous Cushing's syndrome and glucocorticoid withdrawal. Endocrinol Metab Clin North Am. 2005 Jun;34(2):371-84, ix. doi: 10.1016/j.ecl.2005.01.013. PMID: 15850848.
  51. http://www.meditecno.pt/Upload/Product/Archive/lkco1.pdf . Last accessed on 14.4.2021
  52. Patt H, Bandgar T, Lila A, Shah N. Management issues with exogenous steroid therapy. Indian J Endocrinol Metab. 2013 Dec; 17(Suppl 3):S612-7. doi: 10.4103/2230-8210.123548. PMID: 24910822; PMCID: PMC4046616.
  53. Palermo M, Quinkler M, Stewart PM. Apparent mineralocorticoid excess syndrome: an overview. Arq Bras Endocrinol Metabol. 2004 Oct; 48(5):687-96. doi: 10.1590/s0004-27302004000500015. Epub 2005 Mar 7. PMID: 15761540.
  54. Farese RV Jr, Biglieri EG, Shackleton CH, Irony I, Gomez-Fontes R. Licorice-induced hypermineralocorticoidism. N Engl J Med. 1991 Oct 24;325(17):1223-7. doi: 10.1056/NEJM199110243251706. PMID: 1922210.
  55. Gallacher SD, Tsokolas G, Dimitropoulos I. Liquorice-induced apparent mineralocorticoid excess presenting in the emergency department. Clin Med (Lond). 2017 Feb; 17(1):43-45. doi: 10.7861/clinmedicine.17-1-43. PMID: 28148579; PMCID: PMC6297599.

 

Snakebite Envenomation and Endocrine Dysfunction

ABSTRACT

 

Snakebite envenoming (SBE) is a life-threatening medical emergency encountered in tropical parts of Asia, Africa, and Latin America. Toxins in the venom cause local damage and multi-organ dysfunction, predominantly affecting neurological, hematological, and vascular systems. Endocrine anomalies are less frequently reported and often masked by more severe disorders. Anterior pituitary insufficiency is the most common endocrine manifestation and mainly observed after Russell’s viper (Daboia russelii and D. siamensis) bite. SBE-induced hypopituitarism can manifest early or have a delayed presentation. Primary adrenal insufficiency, hyponatremia, hypokalemia, hyperkalemia, and hyperglycemia are also described. These complications are uncommon and under-reported, as SBE occurs in remote areas and medical facilities for endocrine assessment might not be available. Timely identification and management of these problems are critical for optimum medical outcome.

 

INTRODUCTION

 

Snakebite envenoming (SBE) occurs predominantly in rural parts of Asia, Africa, and Latin America (1,2). The World Health Organization (WHO) included SBE in the priority list of neglected tropical diseases in 2018. According to the WHO, 4.5–5.4 million people get bitten by snakes annually, but many cases are not reported as people living in remote areas with limited healthcare access are affected. Clinical illness from SBE develops in 1.8–2.7 million, and the annual mortality is around 81,000 to 138,000 (3).

 

Toxins in snake venom can cause local tissue destruction, neurological damage, hemorrhagic tendency, renal failure, and cardiovascular compromise. Endocrine dysfunctions are uncommon but can have ominous consequences if not recognized. Anterior pituitary insufficiency (API) after Russell’s viper (RV) envenomation (RVE) is the most common endocrine manifestation of SBE. Electrolyte disturbances and hyperglycemia are the other complications described (4). Timely recognition and appropriate management of endocrine derangements like hypocortisolism and electrolyte imbalances can save lives.

 

SPECIES OF SNAKES AND SNAKE VENOM

 

The medically relevant poisonous snakes usually belong to the Elapidae and Viperidae families. Rare cases of envenoming from Atractaspididae and Colubridae families are also described. The common Elapidae snakes include cobras, mambas, kraits, coral snakes, death adders, and sea snakes. The Viperidae snakes of significance are vipers, including RV, adders, asps, and pit vipers. The general notion that Elapidae envenomation results in neuroparalytic manifestation and Viperidae bite induce local reaction and vasculotoxicity does not always hold.

 

Snake venoms contain mixtures of polypeptides, amines, carbohydrates, lipids, phospholipids, nucleosides, and minerals. The principal constituents are proteins belonging to the four families: phospholipase A2, metalloprotease, serine protease, and three-finger peptides. Additional secondary protein families include cysteine-rich secretory proteins, l-amino acid oxidases, kunitz peptides, C-type lectins/snaclecs, disintegrins, and natriuretic peptides (5).

 

Snake venom toxicity can be classified into three main categories: vasculotoxic, neurotoxic, and cytotoxic. Various proteins with enzymatic properties such as phospholipase A2, hyaluronidases, peptidases, and metalloprotease can cause local tissue destruction. Phospholipase A2, metalloproteases, and other protein components can cause neurotoxicity, damage the coagulation cascade, induce muscle necrosis, and sometimes exert cardiotoxic and nephrotoxic effects (6). Cardiac compromise and acute kidney injury (AKI) can also emerge as secondary complications. Endocrine disorders after SBE are uncommon and pathophysiologic mechanisms are incompletely understood.

 

ANTERIOR PITUITARY INSUFFICIENCY

 

Anterior pituitary insufficiency (API) is the most well-recognized endocrine manifestation of SBE. Most cases are from Sri Lanka, India, and Myanmar and occur after RVE (Daboia russelii and D. siamensis).

 

Etiology

 

Wolff first narrated SBE-induced hypopituitarism in 1958 after bite from Bothrops jararacussu (7). The first description of API following RVE in the Indian subcontinent came from Eapen et al. (8). Although RV is found in many south Asian countries, most accounts of RVE-induced API are almost exclusively from Myanmar, India, and Sri Lanka (9–16). It could be related to the geographic variation in venom composition among the same snake species. The incidence of API in a study from northern India was 14.6% (6/41) among patients admitted with vasculotoxic snakebites (presumed RVE) (12).

 

Pathophysiology

 

The pituitary gland is a highly vascular structure enclosed in a bony cavity called the sella tursica. The low-pressure hypothalamic-pituitary portal system originating from the superior hypophyseal artery provides blood supply to the anterior pituitary. The hypophyseal portal system is susceptible to compressive effects from an enlarged or engorged gland and renders the anterior pituitary vulnerable to vascular insults after stimulation from any cause.

 

Sheehan’s syndrome and RVE-induced hypopituitarism share similar pathophysiology (16,17). In Sheehan’s syndrome, hemorrhagic infarction of the pregnancy-induced hyperplastic gland occurs during severe postpartum bleeding-related hypovolemic shock (18). Predisposition to vascular damage in RVE could result from gland engorgement due to a generalized increase in capillary permeability as in capillary leak syndrome (19–21). Additionally, the toxins in RV venom can stimulate pituitary cells as suggested by in-vitro studies, further increasing the susceptibility to damage (22).

 

The vascular supply to an engorged and stimulated gland might be compromised due to microthrombi deposition or hemorrhage from disseminated intravascular coagulation (DIC) (16,23), circulatory or hypovolemic shock (14), thrombotic occlusion of major vessels including cerebral venous thrombosis (24,25), and increased intracranial pressure (14). Autoimmune damage has been postulated to contribute to delayed pituitary injury in Sheehan’s syndrome (26,27). The role of similar immune-mediated damage in the development of delayed hypopituitarism after RVE has not been studied.

 

Clinical Features

 

ACUTE HYPOPITUITARISM

 

Acute onset API has been observed after RVE in several series and can present as early as the first day (11,12,15). In one series of nine patients, API occurred after a median interval of nine days (range 2-14 days) (15). The usual manifestations of RVE include local reaction, coagulopathy, neuromuscular paralysis, and AKI (28,29). Circulatory shock, another feature of RVE, is multifactorial in etiology (14). In the acute phase, adrenal insufficiency (AI) dominates the clinical presentation of API. The symptoms of AI often get masked by other systemic effects of RVE. Clinical clues could be refractory hypotension and the presence of hypoglycemia or hyponatremia.

 

Central hypothyroidism may coexist with secondary AI and is diagnosed if serum thyroid-stimulating hormone (TSH) is low or normal along with decreased serum thyroxine levels. TSH can also get suppressed due to sick euthyroid syndrome and glucocorticoid administration. The diagnosis of central hypothyroidism is difficult to discern in the acute phase, and a follow-up test after recovery is necessary for confirmation. Reassessment of hypothalamic-pituitary-adrenal (HPA) and additional evaluation of the gonadotrophic and growth hormone (GH) secretion should be performed after 4-6 weeks. Acute hypopituitarism is sometimes transient, but the typical outcome is a permanent disease (12,13).

 

DELAYED HYPOPITUITARISM

 

API is often diagnosed years after RVE (30–33). The symptoms depend on the hormone axis involved and the extent of hormone deficiency (34). Delayed hypopituitarism often presents as secondary amenorrhea and infertility in females. In males, hypogonadotropic hypogonadism usually manifests as loss of libido and erectile dysfunction. Loss of secondary sexual characteristics can be present in both genders.

 

Standard features of secondary hypothyroidism include cold intolerance, weight gain, constipation, dry skin, and hoarseness. Secondary AI presents as fatigue, loss of appetite, and orthostatic dizziness (34). Involvement in early childhood can cause stunted growth and delayed or absent puberty (31). In a case series of delayed API, secondary hypothyroidism and hypogonadotropic hypogonadism were present in all the cases (8/8); and GH and secondary AI were present in 75% (6/8) (30). Table 1 depicts the recent case series describing API.

 

Predictors of Hypopituitarism

 

The presence of acute kidney injury (AKI) most consistently correlates with the development of API after RVE (13,30). Bhat et al. found that in patients (n=51) with vasculotoxic snakebite-associated AKI, the risk factors for API were younger age, the number of hemodialysis sessions, and 20-min whole blood clotting time (13). There was a history of AKI in 75% of cases of delayed hypopituitarism in another series (30). In a study describing nine patients with acute API, the predictors were multi-organ dysfunction, lower platelet counts, and more bleeding with a requirement for transfusions (15). However, coagulopathy, AKI, hemodialysis, and clinical severity scores failed to show any association with hypopituitarism in a prospective study (12).

 

Table 1. Case Series of Hypopituitarism After Snakebite Envenoming

Author, year

Region

Snake species

No. of patients

Onset,  Time

Clinical features/ hormone axes involved/ comments

Tun Pe, 1987(10)

Myanmar

Not defined

Snakebite – 220

Acute API – 3

PH (on autopsy) – 4

 

Delayed  API – 11

Acute: 21 hr - 9 d

 

Delayed: 2 wk - 24 yr

Acute - C, GH, PRL

 

Delayed

Symptomatic - 7 Asymptomatic – 4

Proby, 1989 (35)

Myanmar

RV

Acute API (probable) – 20

 

Delayed API – 11/12

Acute, NA

 

Delayed – 8 - 226 wk

C - 10/15

T - 19/20

G -12/17

Golay, 2014 (11)

West Bengal, India

Vasculotoxic snakebite

API - 9/96 cases of snakebite associated AKI

Acute and delayed, 2 wk - 10 yr

C - 6/9

G, GH, T - 9/9

1 empty sella, rest normal

Rajagopala, 2014 (15)

Puducherry, India

Vasculotoxic snakebite

9/989 cases

Acute, 2-14 d

Hypoglycemia (100%), hypotension (67%)

C - 9/9

Partial empty sella in 6/9

Naik, 2018 (12)

India

Vasculotoxic snakebite

9/41 cases

Acute (10%), Mean - 32 hr

Primary AI - 2/6,

C, GH, G - 6/6

PRL - 2/6

 

White, 2019 (36)

Myanmar

RV (85.4%), Rest - cobra, krait, green pit viper, others

20/948 cases

Acute (2%)

Coagulopathy - 68.9%,

 AKI - 72.2%

Gopalkrishnan, 2018 (14)

India

RV, saw-scaled viper

SB - 248

AI – 12

API - 4

Acute

C -19/48

Autopsy - 52.

PH or ischemic necrosis - 46%

Bilateral adrenal hemorrhage - 26%, Adrenal ischemic necrosis – 6%

Shivaprasad, 2018 (30)

Karnataka, India

RV

Delayed API - 8

Delayed, 5-11 yr

C, GH - 6/8

T, G - 8/8

Bhat, 2019 (13)

West Bengal, India

Vasculotoxic snakebite

API - 11/51 at 7 d and 13/33 at 3 mn after snakebite associated AKI

Acute and delayed,

7 d – 3 mn

C – 12/13

PRL – 9/13

G – 9/13

GH – 5/13

T – 4/13

RV - Russell’s viper, C - cortisol, GH - growth hormone, G - gonadotropin, PRL - prolactin, T - thyroid, API – anterior pituitary insufficiency, AI - adrenal insufficiency, PH – pituitary hemorrhage, AKI – acute kidney injury, SB- snake bite

 

Diagnosis

 

ACUTE HYPOPITUITARISM

 

It is challenging to diagnose API during the acute phase. The indicators associated with API are summarized in table 2. The assessment of the HPA axis is required to decide the necessity for glucocorticoid replacement. Hypocortisolism in the acute phase is diagnosed from random cortisol or with the cosyntropin stimulation test. In remote areas, if a delay is anticipated in obtaining the cortisol report, hydrocortisone replacement should be started in suspected cases. The different criteria that have been used to diagnose hypocortisolism in the acute phase are (a) fasting serum cortisol < 3 μg/dL (83 nmol/L) (13), (b) random serum cortisol < 5 μg/dL (138 nmol/L) in suspected pituitary apoplexy (37), (c) random serum cortisol < 10 μg/dL (275 nmol/L) in a critically ill patient (14), (d) post-cosyntropin peak cortisol < 18 μg/dl (500 nmol/L), and (e) post-cosyntropin delta cortisol < 9 μg/dL (250 nmol/L) (38). Note in very ill patients serum cortisol levels can be artifactually low secondary to a decrease in cortisol binding protein.

 

The interpretation of the thyroid function test can be problematic in the acute phase. Low luteinizing hormone (LH), follicle-stimulating hormone (FSH), and prolactin might be indicative but not diagnostic of API. The gonadal axis and sex hormone secretion (testosterone in males and estradiol in females) is usually suppressed during any severe illness. The pituitary function should be reassessed after 4-6 weeks of recovery. The MRI findings reveal a normal gland on imaging in acute cases (11,12,15). Pituitary hemorrhage has been demonstrated in autopsy findings but is seldom observed in imaging studies (15).

 

Table 2. Features Suggestive Of or Associated with Acute Hypopituitarism after Russell’s Viper Bite

Clinical features

Laboratory results

Imaging

Persistent or unexplained hypotension

Hypoglycemia

Pituitary hemorrhage or infarct on MRI

Acute kidney injury

Hyponatremia

Capillary leak syndrome

 

 

Disseminated intravascular coagulation

 

 

 

DELAYED HYPOPITUITARISM

 

Acute hypopituitarism may progress to chronic disease or manifest insidiously years later (11,12,30). It may be prudent to perform periodic surveillance to rule out the development of API following RVE. Hypogonadism has been described in 100% of cases, central hypothyroidism in 96.4%, secondary AI in 82%, and GH deficiency in 77% (30). Central diabetes insipidus (CDI) is very rare and discussed next. The clinical presentation depends on the age, the hormone axes involved, and the mode of onset. The tests recommended for the diagnosis of pituitary hormone deficiency are outlined in table 3. However, facilities for the dynamic tests may not be available in remote areas where SBE is prevalent (34). MRI usually reveals a normal pituitary during the initial year, but partial or complete empty sella may be found later on (4). 

 

Table 3. Investigations for Diagnosis of Chronic Anterior Pituitary Insufficiency

Hormone

Tests

Interpretation/comments

ACTH

Cortisol, ACTH between 8- 9 AM

Serum cortisol values < 3 μg/dL (83 nmol/L) at 8–9 AM on 2 occasions strongly suggest AI in an appropriate clinical setting. Intermediate levels (3-18 μg/dl; 83 – 497 nmol/L) require cosyntropin stimulation test. Concomitant normal or low ACTH levels indicate secondary AI.

 

Cosyntropin (Synacthen) stimulation test

Cosyntropin injection 250 μg (i.v. or i.m.) followed by serum cortisol at 30 min and 60 min. Peak cortisol < 18 μ/dl (500 nmol/L) is suggestive of AI.

 

Insulin tolerance test

Serum cortisol < 20 μg/dL (550 nmol/L) at the time of insulin induced hypoglycemia < 40 mg/dL (2.2 mmol/L). Extreme caution required, not practiced in many centers.

 

 

 

TSH

T4 (total or free), TSH

Low T4 with low or normal TSH suggests the diagnosis of central hypothyroidism

 

 

 

GH

IGF-1

Low IGF-1 suggestive but not diagnostic.

 

GH stimulation test

Adults: Insulin tolerance test, arginine, GHRH stimulation test, Macimorelin stimulation test, glucagon stimulation test.

Children: Clonidine stimulation test in addition to tests used for adults.

 

 

 

Gonadotrophin (Males)

LH, FSH, testosterone (total), SHBG

Low total testosterone (<300 ng/dl (10.41 nmol/L)) between 8–9 AM, preferably on 2 occasions along with low or normal LH, FSH is suggestive of gonadotrophin deficiency. SHBG and free or bioavailable testosterone measurement should be considered in borderline cases.

Gonadotrophin (Females)

LH, FSH, estradiol

Low estradiol in the setting of low or normal LH and FSH in the appropriate clinical setting (amenorrhea/ oligomeorrhea) suggests gonadotrophin deficiency.

 

 

 

Prolactin

Prolactin

Low levels found in hypopituitarism

 ACTH – adrenocorticotrophic hormone, AI – adrenal insufficiency, TSH – thyroid-stimulating hormone, T4 – thyroxine, GH - growth hormone, IGF-1 – insulin-like growth factor 1, LH – luteinizing hormone, FSH – follicle-stimulating hormone, SHBG – sex-hormone binding globulin

 

Management

 

Acute hypopituitarism typically occurs in critically ill patients with severe envenomation from RV. Intravenous hydrocortisone is required if AI is suspected or diagnosed. Thyroxine supplementation for hypothyroidism should be started only after correcting AI. There is a risk of precipitating adrenal crisis because of the accelerated metabolic clearance of cortisol, if thyroxine is administered before treatment of AI (39). Monitoring of electrolytes, and slow correction of hyponatremia when present, in order to prevent central pontine myelinolysis, are important adjuncts.

 

If oral intake is proper and the patient is hemodynamically stable, intravenous hydrocortisone can be substituted by oral glucocorticoids. Hormonal evaluation of the entire pituitary axes should be performed after 4-6 weeks of recovery. Replacement of deficient hormones as per standard practice should be instituted if API persists (34).

 

Post-mortem Findings

 

Pituitary hemorrhage and ischemic necrosis have been described in autopsy studies of 43% (36/84) cases of RVE in Myanmar (40). Areas of ischemic necrosis with hemorrhage at the center were observed in studies from India (15). Deposition of fibrin microthrombi in the pituitary and other organs, including the kidney, suggests a possible role of DIC in the pathogenesis of API (23). 

 

DIABETES INSIPIDUS

 

Involvement of the posterior pituitary gland is exceedingly rare in SBE. There are only a few case reports of central diabetes insipidus (CDI) after RVE (13,31,41,42). The posterior pituitary receives direct arterial supply from the inferior hypophyseal artery and is resistant to vascular damage. On the other hand, the anterior gland is susceptible to vascular compromise as it is supplied by the low-pressure hypophyseal-portal system (43). Moreover, CDI occurs when more than 80% of arginine vasopressin (AVP)-producing hypothalamic magnocellular neurons are lost. The posterior pituitary acts as a storage and secretory organ, and persistent CDI ensues only in the presence of significant damage to the hypothalamus (44). Polyuria, a cardinal feature of CDI, may be obscured due to concomitant hypocortisolism and manifest only after glucocorticoid replacement (45). CDI should be treated with nasal or oral desmopressin.

 

ADRENAL DISORDERS

 

Etiopathogenesis

 

Secondary AI from hypopituitarism is the classically described adrenal disorder resulting from SBE. Primary AI is exceptionally uncommon, though adrenal hemorrhage (AH) has been described in imaging studies and autopsy findings. There are cases of AH occurring after RVE and saw scale viper (Echis carinatus) bite (40,46,47).

 

The pathophysiology of AH is related to DIC and has been postulated to resemble Waterhouse–Friderichsen syndrome (48). The adrenal gland is a highly vascular structure that derives its arterial supply from three arteries but is drained by only one adrenal vein and has a dense internal network of capillaries (49). The causal factors behind predisposition to AH after RVE are summarized in table 4 (14,50,51).

 

Table 4. Factors Predisposing to Hemorrhage after Russell’s Viper Envenomation

Intrinsic predisposition of adrenal vascular structure due to arterial supply by 3 vessels but drainage by one vein

Rich subcapsular plexus with limited drainage by venules forming a “dam”

Disseminated intravascular coagulation and hemorrhagic toxins in the venom increase bleeding tendency

Formation of microthrombi in venules impair venous drainage and cause pooling of blood

Pooling of blood due to capillary leak syndrome

Stress-induced trophic effect of adrenocorticotrophic hormone induces adrenal cortical hyperplasia and increase vascularity

Stress-induced catecholamine secretion causes adrenal venous constriction resulting in pooling of blood in the adrenal gland

 

Diagnosis And Treatment

 

AH has been described in 36% of cases in an autopsy series, though primary AI is rare (40). Refractory hypotension, hypoglycemia, hyponatremia and hyperkalemia should raise suspicion of primary AI. The presence of associated secondary AI can confound the diagnosis. Bilateral AH with transient AI has been described following RVE. The hemorrhage and adrenal function resolved in weeks (47). Cases depicting chronic AI have been published after vasculotoxic SBE (12). Diagnosis and treatment are similar to secondary AI; the primary differentiating point is elevated plasma ACTH. Mineralocorticoid supplementation may be additionally required in primary AI.

 

HYPERGLYCEMIA

 

Etiopathogenesis

 

Hyperglycemia, an infrequent endocrine complication after SBE, has been described following both elapid (Bungarus multicinctus multicinctus) and viper envenomation (Vipera ammodytes ammodytes, European viper spp) (52–54). In rat models, common krait (Bungarus caeruleus) venom produces hyperglycemia (55). Intraperitoneal injection of saw-scaled viper (Echis carinatus) venom in rats suppressed plasma insulin and depleted liver glycogen stores (56). The hyperglycemic effect of Egyptian cobra (Naja haje) was also associated with concomitant depletion of liver and kidney glycogen stores. The mechanism of hyperglycemia is presumed to be triggered by a massive surge of catecholamines, a phenomenon observed after scorpion envenomation and in pheochromocytoma (4,57). Scorpion toxins stimulate sodium and inhibit potassium channels leading to intense and persistent excitation of the autonomic nervous system and release of neurotransmitters from the adrenal medulla, activating parasympathetic (early hours) and sympathetic nerve endings (4–48 hours). Catecholamine-mediated activation of the alpha receptors inhibits insulin secretion and contributes to hyperglycemia (54,58).

 

Clinical Features and Management

 

In a series of 83 children, viper envenomation resulted in hyperglycemia starting 4 hours after the bite, was moderate in severity, and usually transient. Moreover, hyperglycemia at presentation was a marker of high-grade envenomation (54). Severe hyperglycemia up to 480 mg/dl (26.7 mmol/L) occurred in a 45-day baby after two hours of bite from a nose-horned viper (V. a. ammodytes). (53). A retrospective study from Taiwan found hyperglycemia in 15% (7/44) patients of Bungarus multicinctus envenomation. Only one of them had persistent diabetes after recovery (52). Acute pancreatitis can result from the bite of the adder (Vipera berus), but associated hyperglycemia was not observed (59,60). Insulin and other antihyperglycemic drugs should be administered for management of hyperglycemia as and when necessary.

 

ELECTROLYTE DISTURBANCES

 

Hyponatremia

 

ETIOPATHOGENESIS

 

Envenomation by Malayan krait (Bungarus candidus), banded krait (Bungarus fasciatus)), and vipers can result in hyponatremia (61–68). Hyponatremia sometimes occur secondary to anterior pituitary insufficiency (API) after vasculotoxic envenoming but it has also been reported in the absence of API (4). Initial descriptions suggested that the syndrome of inappropriate antidiuretic hormone secretion (SIADH) could be responsible for hyponatremia (64). However, subsequent accounts revealed that urinary salt loss from natriuretic peptides in venom, rather than SIADH, is the pathogenic mechanism. The urinary salt loss is secondary to venom-derived natriuretic peptides, similar to endogenous natriuretic peptides, and acts on the renal tubules to decrease sodium and water reabsorption (69). Cerebral salt wasting has also been postulated to cause hyponatremia (61). An unusually high prevalence of hyponatremia (89%) was observed in a series of 14 patients with berg adder (Bitis atropos) bite in South Africa (67). Many-banded krait (Bungarus multicinctus) envenomation caused hyponatremia in 42% of cases (63).

 

Natriuretic peptides are found in the venom of Elapidae species such as Bungarus candidus,  Bungarus multicinctus, Dendroaspi sangusticeps, Oxyuranus microlepidotus, Pseudonaja textillis, and Pseudechis australis and a few Viperidae species e.g. Hypnale hypnale,  Psudocerastus persicus, and Macrovipera lebetina (66).

 

CLINICAL FEATURES

 

The presentation of hyponatremia depends on the severity and acuteness of onset. The clinical profile ranges from asymptomatic hyponatremia to varying alteration in sensorium to frank coma (61,62). Seizures occur in severe cases (62). Usually there are associated systemic features but isolated hyponatremia, hypovolemia, urinary salt loss, and generalized tonic-clonic seizures, following hump-nosed pit viper bite (Hypnale hypnale) has been described (66). In a case series of 42 patients admitted in Vietnam, 31 people (73.8%) had hyponatremia, the lowest values occurring an average of two days after the bite. Approximately 42–50% of patients who did not receive antivenom developed significant hyponatremia (< 130 mmol/L) 2–3 days post-bite. (70).  Another series of 78 cases of krait bite from Thailand reported hyponatremia in 17.6%, with severe hyponatremia (< 120 mmol/L) developing in four pediatric patients, two of whom developed seizures (71).

 

MANAGEMENT

 

Hyponatremia resulting from natriuretic peptides should be corrected by intravenous saline administration. SIADH is not the cause of hyponatremia, and fluid restriction is not recommended. If chronic hyponatremia is suspected, the correction rate should not exceed 10-12 meq/L in any 24 hours to avoid osmotic demyelination (72). If primary or secondary AI is the cause of hyponatremia, glucocorticoid supplementation is necessary.

 

Hypokalemia

 

ETIOPATHOGENESIS

 

Hypokalemia results from both elapid (73,74) and viper envenomation (75–77). Patients with hypokalemia after RV, common krait (Bungarus caeruleus), and Balkan adder (Vipera berus) bite demonstrated low trans-tubular potassium gradient (TTKG) ruling out renal potassium loss. The intracellular redistribution of potassium has been suggested as the likely pathophysiological mechanism, as gastrointestinal loss was also unlikely. Beta-adrenergic stimulation from toxin-mediated autonomic dysfunction leads to the intracellular shift of potassium and is the likely cause of hypokalemia (74,75). Concomitant hypomagnesemia and high urinary magnesium excretion were also observed in patients with hypokalemia, following Viperidae bite, presumably resulting from the direct toxic action of venom on the renal tubules (77). A high incidence (71%) of hypokalemia (<3.5 mmol/l) was found in a series of 210 patients from Sri Lanka during the first 48 hours. It was accompanied by metabolic acidosis but not respiratory alkalosis (78).

 

CLINICAL FEATURES AND MANAGEMENT

 

Hypokalemia manifests as muscular cramps or weakness, constipation, abdominal bloating, polyuria, and sometimes cause life-threatening complications like arrhythmias, rhabdomyolysis, hypokalemic paralysis, diaphragmatic palsy, and respiratory failure (75). The treatment strategy is similar to that of hypokalemic periodic paralysis. Rebound hyperkalemia is a potential complication during recovery. Potassium should be replaced orally or intravenously, along with appropriate monitoring. Magnesium deficit should be corrected if present (79).

 

Hyperkalemia

 

ETIOPATHOGENESIS

 

Hyperkalemia can complicate envenomation by nose-horned viper (Vipera ammodytes ammodytes), European viper (Vipera berus), and hump-nosed viper (Hypnale hypnale) (53,80–83). Severe envenomation from these snakes causes hyperkalemia secondary to rhabdomyolysis and AKI, and can be fatal (53,80). Hyperkalemia was present in 7% of cases of SBE in 258 patients from Thailand (77).

 

Type 4 renal tubular acidosis (T4RTA) is another possible cause of hyperkalemia. It was described during the recovery phase of bite by hump-nosed viper (81,82). Renal biopsy from these patients showed tubular atrophy and focal segmental glomerulosclerosis pattern (81). The underlying cause could be thrombotic microangiopathy caused by toxins in venom leading to patchy cortical necrosis with delayed or partial recovery of renal functions (83).

 

CLINICAL FEATURES AND MANAGEMENT

 

Hyperkalemia associated with rhabdomyolysis and AKI can cause life-threatening arrhythmias. The presence of hyperkalemia along with hyperchloremic metabolic acidosis and low trans-tubular potassium gradient (TTKG) is suggestive of T4RTA and has been described in victims of hump-nosed viper bites during recovery from AKI. Fludrocortisone has been used successfully in such cases. T4RTA, in most cases, was transient (82). Hyperkalemia associated with rhabdomyolysis and AKI will require potassium lowering therapy and, in severe cases, dialysis.

Table 5. Summary of Snakebite Envenoming Induced Endocrine Dysfunctions

Endocrine manifestation

Pathophysiology

Onset of symptoms

Clinical features

Management

Acute hypopituitarism

Hemorrhagic infarction of the anterior pituitary (pathogenesis similar to Sheehan’s syndrome)

Hours to days

Hypotension not responding to standard therapy, hypoglycemia, hyponatremia

Glucocorticoid +/- thyroxine replacement

Delayed hypopituitarism

Sequalae of vascular insult to pituitary during acute phase

Months to years

Amenorrhea, hypogonadism, hypothyroidism, secondary adrenal insufficiency, growth hormone deficiency

Replacement of deficient hormones

Diabetes Insipidus

Very rare, possible vascular insult during acute phase

Immediate or delayed

Polyuria, polydipsia

Desmopressin

Adrenal insufficiency

Hemorrhage with or without infarction in the adrenals secondary to coagulopathy

Hours to days

Hypotension and circulatory collapse

Glucocorticoid +/- mineralocorticoid replacement

Hyperglycemia

Massive catecholamine surge

First 4-6 hours

Children more than adults

Standard treatment of hyperglycemia

Hyponatremia

Venom derived natriuretic peptides – renal salt wasting

First 2-3 days

Asymptomatic to varying alteration in sensorium to coma, seizures

Intravenous saline

 

 

Pituitary or adrenal insufficiency

Glucocorticoid replacement

Hypokalemia

Intracellular redistribution of potassium secondary to autonomic dysfunction.

Within first 24 hours

Muscle cramps, constipation, abdominal bloating, paralysis, respiratory failure, arrhythmia

Replacement of potassium with precaution to avoid rebound hyperkalemia

Venom-mediated renal tubular damage

Hyperkalemia

Venom mediated thrombotic microangiopathy in the kidneys leading to type 4 renal tubular acidosis

Weeks

 

 

 

 

Arrhythmia

 

 

Fludrocortisone

 

 

 

 

Rhabdomyolysis or kidney injury related

Days

Supportive, dialysis in severe cases

 

CONCLUSION

 

Endocrine dysfunctions associated with SBE are rare. However, missing the diagnosis can have life-threatening consequences. Acute or delayed anterior pituitary insufficiency is the most common manifestation. Establishing the diagnosis of hypocortisolism and timely glucocorticoid initiation in acute hypopituitarism are critical. Reports of adrenal dysfunction are scarce, though adrenal hemorrhage following RVE has been described more often in autopsy series. Electrolyte abnormalities should be anticipated and managed appropriately. Awareness and appropriate treatment of endocrine dysfunctions in resource-limited settings are necessary for optimal outcome.

 

REFERENCES

 

  1. Feasey N, Wansbrough-Jones M, Mabey DCW, Solomon AW. Neglected tropical diseases. Br Med Bull. 2010 Mar 1;93(1):179–200.
  2. Williams DJ, Faiz MA, Abela-Ridder B, Ainsworth S, Bulfone TC, Nickerson AD, et al. Strategy for a globally coordinated response to a priority neglected tropical disease: Snakebite envenoming. PLoS Negl Trop Dis. 2019 Feb 21;13(2):e0007059.
  3. Gutiérrez JM, Calvete JJ, Habib AG, Harrison RA, Williams DJ, Warrell DA. Snakebite envenoming. Nat Rev Dis Primer. 2017 Sep 14;3:17063.
  4. Bhattacharya S, Krishnamurthy A, Gopalakrishnan M, Kalra S, Kantroo V, Aggarwal S, et al. Endocrine and Metabolic Manifestations of Snakebite Envenoming. Am J Trop Med Hyg. 2020 Oct;103(4):1388–96.
  5. Tasoulis T, Isbister GK. A Review and Database of Snake Venom Proteomes. Toxins. 2017 18;9(9).
  6. Ferraz CR, Arrahman A, Xie C, Casewell NR, Lewis RJ, Kool J, et al. Multifunctional Toxins in Snake Venoms and Therapeutic Implications: From Pain to Hemorrhage and Necrosis. Front Ecol Evol. 2019 Jun 19;7:218.
  7. Wolff H. Insuficiência hipofisária anterior por picada de ofídio. Arq Bras Endocrinol Metab. 1958;7:25–47.
  8. Animal, Plant, and Microbial Toxins - Volume 1—Biochemistry | Akira Ohsaka | Springer [Internet]. [cited 2021 May 2]. Available from: https://www.springer.com/gp/book/9781468408881
  9. Myint-Lwin, Warrell DA, Phillips RE, Tin-Nu-Swe, Tun-Pe, Maung-Maung-Lay. Bites by Russell’s viper (Vipera russelli siamensis) in Burma: haemostatic, vascular, and renal disturbances and response to treatment. Lancet Lond Engl. 1985 Dec 7;2(8467):1259–64.
  10. Tun-Pe, Phillips RE, Warrell DA, Moore RA, Tin-Nu-Swe, Myint-Lwin, et al. Acute and chronic pituitary failure resembling Sheehan’s syndrome following bites by Russell’s viper in Burma. Lancet Lond Engl. 1987 Oct 3;2(8562):763–7.
  11. Golay V, Roychowdhary A, Dasgupta S, Pandey R. Hypopituitarism in patients with vasculotoxic snake bite envenomation related acute kidney injury: a prospective study on the prevalence and outcomes of this complication. Pituitary. 2014 Apr;17(2):125–31.
  12. Naik BN, Bhalla A, Sharma N, Mokta J, Singh S, Gupta P, et al. Pituitary dysfunction in survivors of Russell’s viper snake bite envenomation: A prospective study. Neurol India. 2018 Oct;66(5):1351–8.
  13. Bhat S, Mukhopadhyay P, Raychaudhury A, Chowdhury S, Ghosh S. Predictors of hypopituitarism due to vasculotoxic snake bite with acute kidney injury. Pituitary. 2019 Dec;22(6):594–600.
  14. Gopalakrishnan M, Vinod KV, Dutta TK, Shaha KK, Sridhar MG, Saurabh S. Exploring circulatory shock and mortality in viper envenomation: a prospective observational study from India. QJM Mon J Assoc Physicians. 2018 Nov 1;111(11):799–806.
  15. Rajagopala S, Thabah MM, Ariga KK, Gopalakrishnan M. Acute hypopituitarism complicating Russell’s viper envenomation: case series and systematic review. QJM Mon J Assoc Physicians. 2015 Sep;108(9):719–28.
  16. Antonypillai CN, Wass J a. H, Warrell DA, Rajaratnam HN. Hypopituitarism following envenoming by Russell’s vipers (Daboia siamensis and D. russelii) resembling Sheehan’s syndrome: first case report from Sri Lanka, a review of the literature and recommendations for endocrine management. QJM Mon J Assoc Physicians. 2011 Feb;104(2):97–108.
  17. Burke CW. The anterior pituitary, snakebite and Sheehan’s syndrome. Q J Med. 1990 Apr;75(276):331–3.
  18. Diri H, Karaca Z, Tanriverdi F, Unluhizarci K, Kelestimur F. Sheehan’s syndrome: new insights into an old disease. Endocrine. 2016 Jan;51(1):22–31.
  19. Kendre PP, Jose MP, Varghese AM, Menon JC, Joseph JK. Capillary leak syndrome in Daboia russelii bite-a complication associated with poor outcome. Trans R Soc Trop Med Hyg. 2018 Feb 1;112(2):88–93.
  20. Lingam TMC, Tan KY, Tan CH. Capillary leak syndrome induced by the venoms of Russell’s Vipers (Daboia russelii and Daboia siamensis) from eight locales and neutralization of the differential toxicity by three snake antivenoms. Comp Biochem Physiol Toxicol Pharmacol CBP. 2021 Sep 9;250:109186.
  21. Rucavado A, Escalante T, Camacho E, Gutiérrez JM, Fox JW. Systemic vascular leakage induced in mice by Russell’s viper venom from Pakistan. Sci Rep. 2018 Oct 31;8(1):16088.
  22. Hart GR, Proby C, Dedhia G, Yeo TH, Joplin GF, Burrin JM. Burmese Russell’s viper venom causes hormone release from rat pituitary cells in vitro. J Endocrinol. 1989 Aug;122(2):489–94.
  23. Than-Thannull, Francis N, Tin-Nu-Swe  null, Myint-Lwin  null, Tun-Pe  null, Soe-Soe  null, et al. Contribution of focal haemorrhage and microvascular fibrin deposition to fatal envenoming by Russell’s viper (Vipera russelli siamensis) in Burma. Acta Trop. 1989 Jan;46(1):23–38.
  24. Hung D-Z, Wu M-L, Deng J-F, Yang D-Y, Lin-Shiau S-Y. Multiple thrombotic occlusions of vessels after Russell’s viper envenoming. Pharmacol Toxicol. 2002 Sep;91(3):106–10.
  25. Das SK, Khaskil S, Mukhopadhyay S, Chakrabarti S. A patient of Russell’s viper envenomation presenting with cortical venous thrombosis: an extremely uncommon presentation. J Postgrad Med. 2013 Sep;59(3):235–6.
  26. Goswami R, Kochupillai N, Crock PA, Jaleel A, Gupta N. Pituitary autoimmunity in patients with Sheehan’s syndrome. J Clin Endocrinol Metab. 2002 Sep;87(9):4137–41.
  27. De Bellis A, Kelestimur F, Sinisi AA, Ruocco G, Tirelli G, Battaglia M, et al. Anti-hypothalamus and anti-pituitary antibodies may contribute to perpetuate the hypopituitarism in patients with Sheehan’s syndrome. Eur J Endocrinol. 2008 Feb;158(2):147–52.
  28. Kularatne S a. M. Epidemiology and clinical picture of the Russell’s viper (Daboia russelii russelii) bite in Anuradhapura, Sri Lanka: a prospective study of 336 patients. Southeast Asian J Trop Med Public Health. 2003 Dec;34(4):855–62.
  29. Hung D-Z, Wu M-L, Deng J-F, Lin-Shiau S-Y. Russell’s viper snakebite in Taiwan: differences from other Asian countries. Toxicon Off J Int Soc Toxinology. 2002 Sep;40(9):1291–8.
  30. Shivaprasad C, Aiswarya Y, Sridevi A, Anupam B, Amit G, Rakesh B, et al. Delayed hypopituitarism following Russell’s viper envenomation: a case series and literature review. Pituitary. 2019 Feb;22(1):4–12.
  31. Golay V, Roychowdhary A, Pandey R, Pasari A, Praveen M, Arora P, et al. Growth retardation due to panhypopituitarism and central diabetes insipidus following Russell’s viper bite. Southeast Asian J Trop Med Public Health. 2013 Jul 4;44(4):697–702.
  32. Ratnakaran B, Punnoose VP, Das S, Kartha A. Psychosis in Secondary Empty Sella Syndrome following a Russell’s Viper Bite. Indian J Psychol Med. 2016;38(3):254–6.
  33. Yerawar C, Punde D, Pandit A, Deokar P. Russell’s viper bite and the empty sella syndrome. QJM Mon J Assoc Physicians. 2021 Jul 28;114(4):255–7.
  34. Chung T-T, Koch CA, Monson JP. Hypopituitarism. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Oct 17]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK278989/
  35. Proby C, Tha-Aung, Thet-Win, Hla-Mon, Burrin JM, Joplin GF. Immediate and long-term effects on hormone levels following bites by the Burmese Russell’s viper. Q J Med. 1990 Apr;75(276):399–411.
  36. White J, Alfred S, Bates D, Mahmood MA, Warrell D, Cumming R, et al. Twelve month prospective study of snakebite in a major teaching hospital in Mandalay, Myanmar; Myanmar Snakebite Project (MSP). Toxicon X. 2019 Jan;1:100002.
  37. Hannoush ZC, Weiss RE. Pituitary Apoplexy. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000 [cited 2021 Oct 27]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK279125/
  38. Annane D, Pastores SM, Rochwerg B, Arlt W, Balk RA, Beishuizen A, et al. Guidelines for the Diagnosis and Management of Critical Illness-Related Corticosteroid Insufficiency (CIRCI) in Critically Ill Patients (Part I): Society of Critical Care Medicine (SCCM) and European Society of Intensive Care Medicine (ESICM) 2017. Crit Care Med. 2017 Dec;45(12):2078–88.
  39. Higham CE, Johannsson G, Shalet SM. Hypopituitarism. The Lancet. 2016 Nov 12;388(10058):2403–15.
  40. Thein CM, Byard RW. Characteristics and relative numbers of lethal snake bite cases in medicolegal practice in central Myanmar - A five year study. J Forensic Leg Med. 2019 Apr;63:52–5.
  41. Krishnan MN, Kumar S, Ramamoorthy KP. Severe panhypopituitarism and central diabetes insipidus following snake bite: unusual presentation as torsades de pointes. J Assoc Physicians India. 2001 Sep;49:923–4.
  42. Gupta UC, Garg OP, Kataria ML. Cranial diabetes insipidus due to viper bite. J Assoc Physicians India. 1992 Oct;40(10):686–7.
  43. Anderson JR, Antoun N, Burnet N, Chatterjee K, Edwards O, Pickard JD, et al. Neurology of the pituitary gland. J Neurol Neurosurg Psychiatry. 1999 Jun 1;66(6):703–21.
  44. Christ-Crain M, Bichet DG, Fenske WK, Goldman MB, Rittig S, Verbalis JG, et al. Diabetes insipidus. Nat Rev Dis Primer. 2019 Aug 8;5(1):1–20.
  45. Garrahy A, Moran C, Thompson CJ. Diagnosis and management of central diabetes insipidus in adults. Clin Endocrinol (Oxf). 2019 Jan;90(1):23–30.
  46. Lakhotia M, Pahadiya HR, Singh J, Gandhi R, Bhansali S. Adrenal hematoma and right hemothorax after echis carinatus bite: an unusual manifestation. Toxicol Int. 2014 Dec;21(3):325–7.
  47. Senthilkumaran S, Menezes RG, Hussain SA, Luis SA, Thirumalaikolundusubramanian P. Russell’s Viper Envenomation-Associated Addisonian Crisis. Wilderness Environ Med. 2018 Dec;29(4):504–7.
  48. Fox B. Disseminated intravascular coagulation and the Waterhouse-Friderichsen syndrome. Arch Dis Child. 1971 Oct;46(249):680–5.
  49. Gagnon R. The venous drainage of the human adrenal gland. Rev Can Biol. 1956 Feb;14(4):350–9.
  50. Tan GXV, Sutherland T. Adrenal congestion preceding adrenal hemorrhage on CT imaging: a case series. Abdom Radiol N Y. 2016 Feb;41(2):303–10.
  51. Senthilkumaran S, Menezes RG, Hussain SA, Luis SA, Thirumalaikolundusubramanian P. Russell’s Viper Envenomation-Associated Addisonian Crisis. Wilderness Environ Med. 2018 Dec;29(4):504–7.
  52. Mao Y-C, Liu P-Y, Chiang L-C, Liao S-C, Su H-Y, Hsieh S-Y, et al. Bungarus multicinctus multicinctus Snakebite in Taiwan. Am J Trop Med Hyg. 2017 Jun 7;96(6):1497–504.
  53. Lukšić B, Culić V, Stričević L, Brizić I, Poljak NK, Tadić Z. Infant death after nose-horned viper (Vipera ammodytes ammodytes) bite in Croatia: A case report. Toxicon Off J Int Soc Toxinology. 2010 Dec;56(8):1506–9.
  54. Claudet I, Grouteau E, Cordier L, Franchitto N, Bréhin C. Hyperglycemia is a risk factor for high-grade envenomations after European viper bites (Vipera spp.) in children. Clin Toxicol Phila Pa. 2016;54(1):34–9.
  55. Kiran KM, More SS, Gadag JR. Biochemical and clinicopathological changes induced by Bungarus coeruleus venom in a rat model. J Basic Clin Physiol Pharmacol. 2004;15(3–4):277–87.
  56. Al-Saleh SSM. The effect of Echis carinatus crude venom and purified protein fractions on carbohydrate metabolism in rats. Cell Biochem Funct. 2002 Mar;20(1):1–10.
  57. Bouaziz M, Bahloul M, Kallel H, Samet M, Ksibi H, Dammak H, et al. Epidemiological, clinical characteristics and outcome of severe scorpion envenomation in South Tunisia: multivariate analysis of 951 cases. Toxicon Off J Int Soc Toxinology. 2008 Dec 15;52(8):918–26.
  58. Bahloul M, Turki O, Chaari A, Bouaziz M. Incidence, mechanisms and impact outcome of hyperglycaemia in severe scorpion-envenomed patients. Ther Adv Endocrinol Metab. 2018 Jul;9(7):199–208.
  59. Pande R, Khan H. Acute pancreatitis following adder bite in the UK: a case report. Ann R Coll Surg Engl. 2010 Sep;92(6):e25–6.
  60. Kjellström BT. Acute pancreatitis after snake bite. Case report. Acta Chir Scand. 1989 May;155(4–5):291–2.
  61. Kumar Keyal N, Shrestha R, Thapa S, Adhikari P. Krait Snake Bite Presenting as a Cerebral Salt Wasting. Indian J Crit Care Med Peer-Rev Off Publ Indian Soc Crit Care Med. 2019 Jul;23(7):347–8.
  62. Höjer J, Tran Hung H, Warrell D. Life-threatening hyponatremia after krait bite envenoming – A new syndrome. Clin Toxicol. 2010 Nov 1;48(9):956–7.
  63. Hung HT, Höjer J, Du NT. Clinical features of 60 consecutive ICU-treated patients envenomed by Bungarus multicinctus. Southeast Asian J Trop Med Public Health. 2009 May;40(3):518–24.
  64. Trinh KX, Khac QL, Trinh LX, Warrell DA. Hyponatraemia, rhabdomyolysis, alterations in blood pressure and persistent mydriasis in patients envenomed by Malayan kraits (Bungarus candidus) in southern Viet Nam. Toxicon Off J Int Soc Toxinology. 2010 Nov;56(6):1070–5.
  65. Tongpoo A, Sriapha C, Pradoo A, Udomsubpayakul U, Srisuma S, Wananukul W, et al. Krait envenomation in Thailand. Ther Clin Risk Manag. 2018;14:1711–7.
  66. de Silva U, Sarathchandra C, Senanayake H, Pilapitiya S, Siribaddana S, Silva A. Hyponatraemia and seizures in Merrem’s hump-nosed pit viper (Hypnale hypnale) envenoming: a case report. J Med Case Reports. 2018 Aug 2;12(1):213.
  67. van der Walt AJ, Muller GJ. Berg adder (Bitis atropos) envenoming: an analysis of 14 cases. Clin Toxicol Phila Pa. 2019 Feb;57(2):131–6.
  68. Wium CA, Marks CJ, Du Plessis CE, Müller GJ. Berg adder (Bitis atropos): An unusual case of acute poisoning. South Afr Med J Suid-Afr Tydskr Vir Geneeskd. 2017 Nov 27;107(12):1075–7.
  69. Vink S, Jin AH, Poth KJ, Head GA, Alewood PF. Natriuretic peptide drug leads from snake venom. Toxicon Off J Int Soc Toxinology. 2012 Mar 15;59(4):434–45.
  70. Trinh KX, Khac QL, Trinh LX, Warrell DA. Hyponatraemia, rhabdomyolysis, alterations in blood pressure and persistent mydriasis in patients envenomed by Malayan kraits (Bungarus candidus) in southern Viet Nam. Toxicon Off J Int Soc Toxinology. 2010 Nov;56(6):1070–5.
  71. Tongpoo A, Sriapha C, Pradoo A, Udomsubpayakul U, Srisuma S, Wananukul W, et al. Krait envenomation in Thailand. Ther Clin Risk Manag. 2018;14:1711–7.
  72. Spasovski G, Vanholder R, Allolio B, Annane D, Ball S, Bichet D, et al. Clinical practice guideline on diagnosis and treatment of hyponatraemia. Eur J Endocrinol. 2014 Mar;170(3):G1-47.
  73. Gawarammana IB, Mudiyanselage Kularatne SA, Kularatne K, Waduge R, Weerasinghe VS, Bowatta S, et al. Deep coma and hypokalaemia of unknown aetiology following Bungarus caeruleus bites: Exploration of pathophysiological mechanisms with two case studies. J Venom Res. 2010 Dec 14;1:71–5.
  74. Kularatne S a. M. Common krait (Bungarus caeruleus) bite in Anuradhapura, Sri Lanka: a prospective clinical study, 1996-98. Postgrad Med J. 2002 May;78(919):276–80.
  75. Jeevagan V, Katulanda P, Gnanathasan CA, Warrell DA. Acute pituitary insufficiency and hypokalaemia following envenoming by Russell’s viper (Daboia russelii) in Sri Lanka: Exploring the pathophysiological mechanisms. Toxicon Off J Int Soc Toxinology. 2013 Mar 1;63:78–82.
  76. Malina T, Krecsák L, Jelić D, Maretić T, Tóth T, Siško M, et al. First clinical experiences about the neurotoxic envenomings inflicted by lowland populations of the Balkan adder, Vipera berus bosniensis. Neurotoxicology. 2011 Jan;32(1):68–74.
  77. Aye K-P, Thanachartwet V, Soe C, Desakorn V, Thwin K-T, Chamnanchanunt S, et al. Clinical and laboratory parameters associated with acute kidney injury in patients with snakebite envenomation: a prospective observational study from Myanmar. BMC Nephrol. 2017 Mar 16;18(1):92.
  78. Sellahewa K. Lessons from four studies on the management of snake bite in Sri Lanka. Ceylon Med J. 1997 Mar;42(1):8–15.
  79. Kardalas E, Paschou SA, Anagnostis P, Muscogiuri G, Siasos G, Vryonidou A. Hypokalemia: a clinical update. Endocr Connect. 2018 Apr;7(4):R135–46.
  80. Denis D, Lamireau T, Llanas B, Bedry R, Fayon M. Rhabdomyolysis in European viper bite. Acta Paediatr Oslo Nor 1992. 1998 Sep;87(9):1013–5.
  81. Weerakkody RM, Lokuliyana PN, Lanerolle RD. Transient distal renal tubular acidosis following hump nosed viper bite: Two cases from Sri Lanka. Saudi J Kidney Dis Transplant. 2016 Sep 1;27(5):1018.
  82. Karunarathne S, Udayakumara Y, Govindapala D, Fernando H. Type IV renal tubular acidosis following resolution of acute kidney injury and disseminated intravascular coagulation due to hump-nosed viper bite. Indian J Nephrol. 2013;23(4):294–6.
  83. Herath N, Wazil A, Kularatne S, Ratnatunga N, Weerakoon K, Badurdeen S, et al. Thrombotic microangiopathy and acute kidney injury in hump-nosed viper (Hypnale species) envenoming: a descriptive study in Sri Lanka. Toxicon Off J Int Soc Toxinology. 2012 Jul;60(1):61–5.

 

Fungi and Endocrine Dysfunction

ABSTRACT

 

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

INTRODUCTION

 

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

 

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

 

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

 

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

 

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

 

TYPES OF FUNGAL INFECTION

 

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

 

Anatomical Categories

 

MUCOCUTANEOUS INFECTIONS

 

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

 

DEEP ORGAN INFECTIONS

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

 

Epidemiological Categories

 

ENDEMIC MYCOSES

 

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

 

OPPORTUNISTIC MYCOSES

 

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

 

Table 1. Fungi Affecting Specific Endocrine Glands

Type of fungus

Organs affected     

Aspergillosis

Pituitary, Thyroid, Pancreas, Adrenal

Zygomycosis

Thyroid

Candidiasis

Pituitary, Thyroid, Pancreas, Testis

Cryptococcosis

Thyroid, Pancreas, Adrenal, Testis

Histoplasmosis

Thyroid, Adrenal, Ovaries,

Blastomycosis

Testis, Ovaries

Coccidioidomycosis

Thyroid, Adrenal,

Paracoccidioidomycosis

Thyroid, Adrenal

Pneumocystis jirovecii

Pituitary, Thyroid, Parathyroid, Pancreas, Adrenal

 

Based on Morphology

 

YEASTS

 

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

 

MOLDS

 

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

 

DIMORPHIC

 

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

Figure 1. Classification of Fungal Infections

FUNGAL DISEASES OF MAJOR ENDOCRINE GLANDS

 

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

 

Table 2. Conditions Predisposing to Systemic Fungal Infections

A.    Endocrine diseases

1.     Diabetes mellitus

2.     Cushing’s syndrome

3.     Autoimmune polyendocrine syndrome-1

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

B.    Immunosuppressed states

1.     Cancer

2.     Acquired immunodeficiency syndrome

3.     Acute leukemia

4.     Hematopoietic stem cell transplant recipients

5.     Solid-organ transplant recipients

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

 

Pituitary Fungal Infections

 

ETIOLOGY

 

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

 

CLINICAL FEATURES

 

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

 

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

Author, year

Clinical setting

Symptoms

Diagnosis

Management/

outcome

Moore, 2016 (8)

74-year old male,

CAD, CKD, AHA hypertension

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

 

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

Fatal outcome, autopsy findings revealed fungal hyphae in pituitary

Choi, 2021(15)

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

Headache, visual disturbance, hyponatremia

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

Endoscopic debridement of sinuses. Oral voriconazole given, gradual improvement

Saffarian,2018 (16)

60-year old male

DM, hypertension, sphenoid aspergilloma

Headache, progressive visual loss, 4thcranial nerve palsy

MRI findings, endoscopy by nasal approach demonstrated aspergillus in biopsy

Endoscopic drainage, intravenous amphotericin, responded to treatment

Ouyang, 2015 (18)

55-year old female,

no comorbidities

Headache, dizziness, and decreased visual acuity

 

MRI - sellar and sphenoid sinus

mass

Prolactin - 815 ng/mL

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

 

Vijay-vargiya, 2013 (14)

68-year old female,

kidney transplant recipient

Headache, left temporal hemianopsia, ptosis.

MRI – sellar mass

Intraoperative frozen

section showed organisms consistent with aspergillus

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

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

 

DIAGNOSIS

 

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

 

TREATMENT

 

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

 

Thyroid Disorders

 

ETIOLOGY

 

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

 

Table 4. Predisposing Conditions Where Fungus Affects the Thyroid Gland

Type of fungus

Predisposing condition

Aspergillus

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

Pneumocystis

AIDS (25), Thymic alymphoplasia (36)

Candida

ALL (37), AML (38)

Coccidiodes

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

Histoplasmosis

NHL (41)

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

 

CLINICAL FEATURES

 

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

 

Aspergillus

 

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

 

Pneumocystis

 

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

 

Others

 

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

 

DIAGNOSIS

 

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

 

TREATMENT

 

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

 

Disorders of Calcium Metabolism

 

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

 

MONOCYTE 1α HYDROXYLASE MEDIATED HYPERCALCEMIA

 

Etiology and Pathogenesis

 

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

 

Clinical Profile

 

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

 

Laboratory Features

 

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

 

Treatment

 

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

 

PARATHYROID HORMONE REALTED PROTEIN (PTHrP) MEDIATED HYPERCALCEMIA

 

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

 

OTHER DISORDERS OF CALCIUM METABOLISM

 

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

 

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

 

Fungal Infection of the Adrenal Gland

 

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

 

ETIOLOGY AND PATHOGENESIS

 

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

 

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

 

CLINICAL FEATURES

 

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

 

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

 

DIAGNOSIS

 

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

 

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

 

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

 

MANGEMENT AND PROGNOSIS

 

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

 

Fungal Infection of the Pancreas

 

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

 

ETIOLOGY AND PATHOGENESIS

 

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

 

CLINICAL COURSE AND MANAGEMENT

 

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

 

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

 

 

Fungal Infection of the Testis

 

ETIOLOGY AND PATHOGENESIS

 

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

 

CLINICAL COURSE AND MANAGEMENT

 

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

 

Fungal Infection of the Ovary

 

ETIOLOGY AND PATHOGENESIS

 

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

 

CLINICAL COURSE AND MANAGEMENT

 

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

 

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

 

FUNGAL INFECTIONS OCCURING IN ENDOCRINE DISORDERS

 

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

 

Fungal Infections in Patients with Diabetes

 

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

 

CANDIDIASIS

 

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

 

Table 5. Candida Infections in Diabetes

Site

Usual species

Predisposing factors

Clinical features

Diagnosis

Treatment

Oral candidiasis

C. albicans

C. glabrata

C. tropicalis

C. krusei

C. dubliniensis C. parapsilosis

(124)

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

Types of lesions: Pseudo-membranous

Hyperplastic

Erythematous

Atrophic (denture stomatitis)

Angular cheilitis

Median rhomboid glossitis (128)

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

Oral hygiene

Topical: Clotrimazole, miconazole, nystatin, amphotericin B suspension

Oral: Fluconazole, itraconazole

(129)

Vulvo-vaginal candidiasis

C. albicans

C. glabrata (124)

 

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

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

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

Glycemic control

Vaginal: Clotrimazole, miconazole, tioconazole, terconazole, butoconazole

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

 

Balanoposthitis

C. albicans

C. glabrata

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

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

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

Glycemic control

Topical: Clotrimazole, miconazole

Oral: Fluconazole (150 mg single dose), Itraconazole

Esophageal candidiasis

C. albicans,  C. dubliniensis (124)

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

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

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

Initial therapy: Oral fluconazole

Second-line therapy: Itraconazole,

voriconazole

isavuconazole,

echinocandin,

liposomal amphotericin B

Urinary tract candidiasis

C. albicans,

C. glabrata,

C. tropicalis (137)

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

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

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

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

First line: Fluconazole

Second line: Flucytosine, amphotericin B (138)

Onychomycosis

C. albicans,

C. parapsilosis

C. tropicalis (124)

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

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

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

Oral itraconazole treatment of choice.

Terbinafine might also be efficacious (141)

Systemic candidiasis

C. albicans,

C. parapsilosis, C. krusei,

C. tropicalis,

C. glabrata (142)

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

Can vary from minimal fever to a full-blown sepsis

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

Preferred therapy Echinocandin: anidulafungin, capsofungin, micofungin

Alternative: Amphotericin B

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

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

 

MUCORMYCOSIS

 

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

 

Pathogenic Organisms

 

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

 

Pathogenesis

 

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

 

Table 6. Risk Factors for Mucormycosis

Uncontrolled diabetes mellitus especially if associated with ketoacidosis

Underlying malignancy receiving chemotherapy or immunotherapy

Solid organ transplant

Hematopoietic stem cell transplant

Treatment with deferoxamine

Iron overload

Corticosteroid therapy

Trauma or burns

Malnutrition

Coronavirus disease 2019

 

Clinical Features

 

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

 

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

Site

Symptoms

Signs

Paranasal sinuses

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

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

Systemic features

Fever

Fever

Orbit

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

Periorbital swelling, chemosis, proptosis, loss of visual acuity

Cavernous sinus

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

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

Palate

Ulceration, pain, swelling

Ulceration, eschar formation

Central nervous system

Headache, drowsiness, seizures, hemiparesis, obtundation, coma

Focal seizures, hemiparesis, altered sensorium

Vascular invasion

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

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

 

Diagnosis

 

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

 

Treatment and Prognosis

 

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

 

DERMATOPHYTES

 

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

 

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

 

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

 

Fungal Infections in Cushing’s Syndrome

 

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

 

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

Cell/Mediator

Dysfunction

Innate immunity

Neutrophils

Impaired neutrophil adherence to endothelium

Monocytes and macrophages

Decreased circulating monocytes

Decreased degranulation capacity

Decreased phagocyte function

Natural Killer cells

Suppressed cytotoxic activity

Adaptive immunity

T Cells

Lymphopenia due to a reduction in CD4+ subset

 

Influences the Th1/Th2 balance

 

Induces apoptosis in mature T lymphocytes

Cytokines

Cytokines

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

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

 

CANDIDIASIS

 

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

 

ASPERGILLUS

 

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

 

PNEUMOCYSTIS

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

 

CRYPTOCOCCOSIS

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

 

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

 

HISTOPLASMOSIS

 

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

 

OTHER FUNGAL INFECTIONS

 

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

 

Chronic Mucocutaneous Candidiasis in Autoimmune Polyendocrine Syndrome Type 1

 

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

 

PATHOGENESIS

 

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

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

 

CLINICAL SPECTRUM

 

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

 

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

 

TREATMENT

 

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

 

ADVERSE ENDOCRINE EFFECTS OF ANTIFUNGAL AGENTS

 

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

 

Azoles

 

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

 

ADRENAL GLAND

 

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

 

Ketoconazole

 

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

 

Fluconazole and Posaconazole

 

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

 

Itraconazole and Voriconazole

 

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

 

Pseudohyperaldosteronism

 

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

 

GONADS

 

Male Sexual Dysfunction

 

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

 

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

 

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

 

Female Reproductive Dysfunction

 

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

 

HYPONATREMIA

 

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

 

FLUORIDE-INDUCED PERIOSTITIS

 

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

 

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

 

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

 

OTHER ENDOCRINE ABNORMALITIES

 

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

 

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

 

Polyenes

 

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

 

TUBULAR DAMAGE

 

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

 

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

 

Echinocandins

 

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

 

Other Agents

 

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

 

CONCLUSION

 

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

 

REFERENCES

 

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