A narrative review: an update on primary adrenal insufficiency (PAI) in pediatric population※※Special series on Clinical Pearls in Pediatric Endocrinology and Metabolism.
Review Article

A narrative review: an update on primary adrenal insufficiency (PAI) in pediatric population

Berrin Ergun-Longmire, Dustin Rowland, John Dewey, Patricia Vining-Maravolo

Department of Pediatric & Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Berrin Ergun-Longmire, MD. Associate Professor, Pediatric Endocrinology, Department of Pediatric & Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, MI 49008-1284, USA. Email: berrin.ergun-longmire@med.wmich.edu.

Background and Objective: This article aims to provide an update on etiology, diagnosis, and management of primary adrenal insufficiency (PAI), also known as Addison’s disease in pediatric population, while focusing on rare monogenic causes. PAI is a rare condition which results in destruction of the adrenal cortex leading to cortisol and aldosterone deficiency. Autoimmunity, infections, malignancy, genetic and metabolic disorders, and iatrogenic causes such as medications that cause adrenal hemorrhage (anticoagulants) or impairs glucocorticoid synthesis and action are known etiologic factors. Although autoimmunity is the most common cause of PAI in adolescents and adults, genetic causes are more common in children. Despite significant advancements made in understanding the pathogeneses of PAI, availability of diagnostic tools and treatment, in many cases, PAI remains unrecognized until the affected individual presents in acute adrenal crisis.

Methods: Literature was searched from PubMed, MedlinePlus, Science Direct, and Google Scholar, using appropriate search terms. We included articles up to November 2021, focusing on new developments since 2019.

Key Content and Findings: There are a large number of genetic and non-genetic causes of PAI, which can occur at a variety of times and with a variety of co-occurring symptoms across the lifespan. Knowledge of the time of onset of various causes of PAI and chronology of later-onset symptoms can aid the clinician in determining the utility of genetic testing in patients with PAI of unknown etiology.

Conclusions: PAI has a number of potential underlying etiologies and can affect a broad spectrum of patients, from pediatrics to geriatrics. Since its first description by Addison, significant advancements have been made in understanding adrenal gland development and function. While PAI remains a relatively rare condition, it has severe and precipitous implications when it arises. Having a high index of suspicion is the key for early diagnosis of PAI and the prevention of life-threatening adrenal crisis. Corticosteroid and mineralocorticoid replacement is necessary to prevent morbidity and life-threatening adrenal crisis.

Keywords: Primary adrenal insufficiency (PAI); Addison’s disease; steroidogenesis; autoimmunity; stress dose


Received: 07 January 2022; Accepted: 11 August 2022; Published online: 09 September 2022.

doi: 10.21037/pm-22-2


Introduction

Primary adrenal insufficiency (PAI), also known as Addison’s disease, is characterized by glucocorticoid (cortisol) and mineralocorticoid (aldosterone) deficiency and can be a potentially life-threatening condition if unrecognized. The association between clinical features of PAI and pathological changes of adrenal glands on 11 patients was first described by Dr. Thomas Addison, a British surgeon, in 1855 (1). PAI was a fatal disease until the discovery of synthetic steroids in 1940s (2).

PAI is a rare disorder, though its prevalence is noticeably increased from 40 to 70 per million in the 1960s to 110–140 per million in the 2000s (3-5). In adults, the incidence of PAI is estimated at 4.4–6.0/million/year (6) and is primarily due to autoimmune destruction of the adrenal cortex. There are no data on the incidence of PAI in children other than congenital adrenal hyperplasia (CAH) due to 21 hydroxylase deficiency, which is the most common cause of PAI in children with the incidence of 1:15,000 to 1:20,000 worldwide (7).

Other causes of PAI include infectious diseases, malignancies, rare genetic disorders, iatrogenic factors, infiltrative, and metabolic conditions (Table 1). Regardless of the etiologic factor, PAI continues to present significant challenges regarding diagnosis in early stages due to its rarity and nonspecific signs and symptoms in either children or adults. Despite advancements in our understanding of pathogenesis of PAI and availability of diagnostic tools and treatment, most patients with PAI seek medical attention from two or three physicians before the correct diagnosis is made (28). In many cases, it remains unrecognized until the affected individual presents in acute adrenal crisis.

Table 1

Causes of primary adrenal insufficiency*

Syndrome Protein Gene Locus
Adrenal gland dysgenesis
   X-linked adrenal hypoplasia Congenita (8) DAX1 NR0B1 Xp21.2
   Steroidogenic factor-1 (SF1) (9) SF-1 NR5A1 9q33.3
   ACTH resistance syndromes
    Familial glucocorticoid deficiency type 1 (10) ACTHR MC2R 18p11.2
    Familial glucocorticoid deficiency type 2 (11) MRAP MRAP 21q22.11
    Triple A (Allgrove) syndrome (12) ALADIN AAAS 12q13.13
   SERKAL syndrome (13) WNT4 WNT4 1p36.12
   IMAGe syndrome (14) CDKN1C CDKN1C 11p15.4
   IMAGe-like syndrome (15) POLE1 POLE1 12q24.33
   MIRAGE syndrome (16) SAMD9 SAMD9 7q21.2
Adrenal gland destruction
   Autoimmune Variable Variable
   Metabolic lipid disorders
    Adrenoleukodystrophy (17) ABCD1 ABCD1 Xq28
    Wolman disease (18) Lysosomal acid lipase LIPA 10q23.31
    Zellweger spectrum syndrome (19) PEX family proteins PEX gene family Multiple
    Sphingolipidosis (20) SGPL1 SGPL1 10q22.1
   Infections
   Bilateral adrenal hemorrhage
   Metastatic tumors
   Other (sarcoidosis, amyloidosis)
Steroidogenetic defects
   Congenital adrenal hyperplasia (CAH)
    21-hydroxylase deficiency (21) CYP21A2 CYP21A2 6p21.33
    11-β-hydroxylase deficiency (22) CYP11B1 CYP11B1 8q24.3
    3-β-hydroxysteroid dehydrogenase deficiency (23) 3β-HSD HSD3B1, HSD3B2 1p12
    17-α-hydroxylase/17,20-lyase deficiency (24) CYP17A1 CYP17A1 10q24.32
    Cholesterol desmolase deficiency (25) CYP11A1 CYP11A1 15q24.1
   Mitochondrial deletion syndromes (26) Variable Variable mtDNA
   Smith-Lemli-Opitz syndrome DHCR7 DHCR7 11q13.4
   Sphingolipidosis SGPL1 SGPL1 10q22.1
   Drugs

*, gene loci confirmed using the HUGO Gene Nomenclature Committee at the European Bioinformatics Institute (27). ACTH, adrenocorticotropic hormone; MIRAGE, major findings of myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, and enteropathy; SERKAL, SEx Reversion, Kidneys, Adrenal and Lung dysgenesis; IMAGe, intrauterine growth restriction, metaphyseal dysplasia, adrenal hypoplasia congenita, genital anomalies; SGPL1, sphingosine-1-phosphate lyase-1; HUGO, Human Genome Organisation.

This narrative review aims to provide an update on etiology, including recently added new genetic causes, diagnosis, and management of PAI in pediatrics. We present this article in accordance with the Narrative Review reporting checklist (available at https://pm.amegroups.com/article/view/10.21037/pm-22-2/rc).


Methods

A literature search was performed using the following scholarly search engines: PubMed, MedlinePlus, Science Direct, and Google Scholar. Appropriate search terms were utilized with a time range up to November 2021. Particular focus was given to articles published since 2019. Search terms focused on pathologic conditions with a significant presentation of PAI and genetic dysfunctions that highlight important molecular mechanisms within the adrenal gland (Table 2).

Table 2

The search strategy summary

Items Specification
Date of search Multiple dates
Databases and other sources searched PubMed, MedlinePlus, Science Direct, and Google Scholar
Search terms used Primary adrenal insufficiency, Addison’s disease, steroidogenesis, autoimmunity, pediatric, stress dose
Timeframe Up to November 2021, especially focusing on articles published
last 3 years
Inclusion criteria Clinical trial
Meta-analysis
Randomized controlled trial
Review
Systematic review
Language: English
Exclusion criteria Study was published in a language other than English
Observational study design
Selection process All authors equally conducted the selection, reviewed and agreed with the selected article

Etiology

PAI may present at any age. However, the frequency of etiology varies noticeably in relation to the patient’s age and gender. While tuberculosis was the main cause of PAI until the early 20th century, autoimmune destruction of all three layers of the adrenal cortex became the leading cause in developed countries and is being the most common etiologic factor in adolescents and adults. Autoimmunity may occur isolated or as a part of autoimmune polyglandular syndrome type 1 (APS-1) or autoimmune polyglandular syndrome type 2 (APS-2). Autoimmune PAI is more common in women than men (female to male ratio 1.5–3.5:1) (29,30). In infants and children, however, genetic causes, especially steroidogenic enzyme defects known as CAH, are the most common cause of PAI (19). In addition to CAH, mitochondrial disorders, Wolman disease, and the Smith-Lemli-Opitz syndrome are rare causes of PAI due to impaired steroidogenesis. While mutations in genes responsible for adrenal gland development including DAX-1 [X-linked adrenal hypoplasia congenita (XL-AHC)], steroidogenic factor 1 (SF1), melanocortin 2 receptor (MC2R), MC2R accessory protein (MRAP), CDKN1C, sterile alpha motif domain containing 9 (SAMD9), and polymerase epsilon-1 (POLE1) result in adrenal gland dysgenesis or hypoplasia, APS, infections, amyloidosis, sarcoidosis, adrenal leukodystrophy, adrenal metastases or hemorrhage can cause adrenal gland destruction and lead to PAI (Table 1).

Adrenal gland dysgenesis

XL-AHC—DAX1 (NR0B1)

DAX1 (NR0B1) (dosage sensitive sex reversal, adrenal hypoplasia congenita critical region on the X chromosome) is an orphan protein of the nuclear receptor superfamily that is a key regulator of appropriate adrenal and gonadal development. The protein regulates transcription when bound to its ligand, acting on both promoters and transcription factor targets (31). Mutations have been characterized in the protein-encoding gene that lead to either protein truncation or alterations in ligand binding activity, resulting in partial loss of function (19). DAX1 mutation is a relatively common and likely underdiagnosed cause of PAI, especially XL-AHC; in one study, 12/95 patients with PAI of unknown etiology were found to have DAX1 mutations. The most commonly characterized pathologic gene contained an amino-terminal stop variant, with p.W235 and p.E256 identified in 50% of cases (32). Clinically, these patients present with adrenal insufficiency that often presents in early infancy but may instead appear during childhood after a stressful stimulus is encountered (bimodal distribution) (33). All children with DAX-1 mutation will require mineralocorticoid replacement and any who survive to puberty will develop hypogonadotropic hypogonadism (34). The typical phenotype at puberty is PAI with hypogonadotropic hypogonadism, with DAX1 mutations accounting for two-thirds of all patients with PAI and hypogonadotropic hypogonadism in adolescence (19,34). Due to the variable age of presentation and many atypical phenotypes of subjects with DAX-1 mutations, clinicians should consider genetic testing any children with PAI of unknown etiology who later develop hypogonadotropic hypogonadism (35).

SF1 (NR5A1)

SF1 is a protein of the nuclear receptor superfamily that is implicated in both adrenal and gonadal differentiation Most identified mutations of this gene are heterozygous and arose from de novo mutation during spermatogenesis; in rare cases, female carriers may pass this mutation to their male child in a sex-limited dominant manner (19). Adrenal insufficiency is very rare in patients with SF1 deficiency, with the primary presenting symptoms being more related to disordered sexual differentiation, ovarian failure, or male factor infertility (32). Males will most often display testicular dysfunction but may also manifest hypospadias (5–7%) or rarely 46,XY sex development (DSD). A broad spectrum of 46,XY DSD phenotypes have been described in association with SF1 deficiency; the most common is ambiguous genitalia without Mullerian structures with and normal adrenal function (34). In females, the primary manifestation is 46XX sex reversal; early-onset PAI is still rare but more common than in 46,XY and may present before 14 months with salt-losing adrenal failure and seizures (36). Heterozygous females may also present with hypertension and ambiguous genitalia during early adulthood (37). Streak gonads may be observed in conjunction with phenotypically normal external female genitalia (31). Although homozygotic mutations are not generally identified in humans, mouse models have shown that complete loss of SF1 function results in agenesis of adrenal glands and gonads leading to severe adrenal insufficiency and neonatal death (38).

Adrenocorticotropic hormone (ACTH) resistance syndromes

Familial glucocorticoid deficiency (FGD)

Mutations in the MC2R or the MRAP are the etiology of FGD (39). About 25% of FGD cases are due to mutation of MC2R, which acts as the endogenous receptor for ACTH; these cases are categorized as FGD-1. Affected individuals mostly present in the neonatal period with hypoglycemic seizures, hyperpigmentation, and signs of immunosuppressed status (40).

A more recently identified second gene, MRAP encodes a small single transmembrane domain protein that is essential for proper targeting and transport of the ACTH receptor (MC2R) to the cell membrane (41). Due to this interconnected functionality, mutation or loss of MRAP presents similarly to MC2R deficiency and is responsible for 20% of identified cases of hereditary unresponsiveness to ACTH (42). Clinically, lack of appropriate MRAP activity will manifest in the first months of life as an FGD-like condition with electrolyte imbalances, hypoglycemia, and other signs of PAI (32). Generally, only glucocorticoids are affected in FGD as the renin-angiotensin-aldosterone system is intact, but some volume-depleted patients may have elevation of aldosterone (43). Symptomatically distinguishing defects in MRAP from defects in MC2R is difficult, but patients with MRAP mutations may have onset of symptoms later in life than the classic first 6 months associated with MC2R mutations. This has led to the concept of FGD type 1 and 2, with type 2 being similar to other etiologies of FGD but with later onset and lack of extra-adrenal features such as cryptorchidism and testis adenoma (34,44). However, 56% of patients will present with hypoglycemia leading to convulsions; in severe cases respiratory distress will also be noted. Other etiologies of PAI cause these symptoms significantly less often (32). Despite an average higher age at presentation for MRAP deficiency, patients may still manifest severe salt loss, adrenal insufficiency, and hyperpigmentation within the first months of life (36).

In addition to MC2R and MRAP, FGD has been associated with mutations in MCM4 (the mini chromosome maintenance-deficient 4 homolog gene), NNT (nicotinamide nucleotide transhydrogenase), TXNRD2 (thioredoxin reductase 2), GPX1 (glutathione Peroxidase 1), and PRDX3 (peroxiredoxin 3) (41).

Allgrove syndrome

One form of ACTH resistance syndrome is Allgrove syndrome, also termed “Triple A” syndrome (45). This syndrome is due to autosomal recessive mutation of the AAAS gene on chromosome 12q13, resulting in a truncated protein product (46). AAAS codes for the protein ALADIN, a nucleoporin that is primarily expressed within the pituitary, adrenals, and pancreas (47). The pathogenesis of Allgrove syndrome is multifactorial and has not been clearly elucidated, but likely is related to impaired oxidative stress response within affected cells (48). The age of presentation for various features of Allgrove syndrome is highly variable, with glucocorticoid deficiency appearing at any time from 6 to 19 years of age. Achalasia similarly may present early in infancy or in adulthood (49). A recent genetic analysis of 70 Chinese children with adrenal insufficiency determined that 2/70 of these children had a mutation within the AAAS gene, highlighting Allgrove syndrome as an important component of the PAI differential (50).

SERKAL (SEx Reversion, Kidneys, Adrenal and Lung dysgenesis) syndrome

Wnt family member 4 (WNT4) is an essential WNT ligand for proper development of adrenocortical cells, as well as organogenesis of the lung and kidney. Inadequate WNT4 function results in overactivity of protein kinase A during fetal development; double inactivation of WNT4 (autosomal recessive) will result in fetal nonviability (38). Fetuses with characterized WNT4 double inactivation will undergo spontaneous abortion with autopsy revealing lack or underdevelopment of lungs, kidneys, and adrenal glands, as well as sex reversal in 46,XX fetuses (34). This is termed SERKAL syndrome (female sex reversal and dysgenesis of kidneys, adrenals, and lungs), may also result in Mullerian aplasia or hyperandrogenism, and has only been characterized within one family and two fetuses (36).

IMAGe and IMAGe-like syndromes

IMAGe syndrome is caused by mutations in CDKN1C (cyclin-dependent kinase inhibitor 1C), a key negative cell cycle regulator gene (19). IMAGe syndrome is an acronym for the major findings of intrauterine growth retardation (IUGR), metaphyseal dysplasia, adrenal hypoplasia congenita, and genital anomalies in males (51). This syndrome may be diagnosed at a wide range of ages across the lifespan, from prenatal to teenage years, but is most commonly identified at birth (52).

Logan et al. recently described biallelic loss of function variants in POLE1 (polymerase epsilon-1) in 15 individuals from 12 families; these individuals had similar phenotypic features of IMAGe syndrome, distinctive facial features, and immunodeficiency characterized by lymphocyte depletion (53). POLE1 is a key DNA-leading strand polymerase, and its loss of function disrupts DNA replication (19).

MIRAGE (major findings of myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, and enteropathy) syndrome

MIRAGE syndrome is a recently described complex disorder characterized by myelodysplasia, infections, restriction of growth, adrenal hypoplasia, genital anomalies, and enteropathy (16). Affected individuals also have developmental delay, dysmorphism, chronic lung disease, central nervous system abnormalities, thrombocytopenia/anemia, and high mortality rate (54). Most mortality occurs early in the life span due to infection (55).

MIRAGE syndrome is caused by a gain-of-function variant in the growth repressor, SAMD9 gene located on the long arm of chromosome 7 (7q21.2) (56).

Adrenal gland destruction

Autoimmune PAI (Addison’s disease)

Autoimmune adrenalitis is the most common cause of PAI in older children and adults, accounting for 68–94% of cases (57). More than 80% of patients have circulating serum autoantibodies to the 21-hydroxylase, the key steroidogenic enzyme (29). The presentation of Addison’s disease occurs at a mean age of 30; although autoimmune PAI may occur in isolation, over 50% of cases have other autoimmune disorders as part of APS-1 or APS-2 (58).

Genetic etiologies of PAI and distinguishing signs and symptoms by age of manifestation are shown in Figure 1. Mutation of the autoimmune regulator (AIRE) gene on chromosome 21q22.3 results in the disorder APS-1, also referred as autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). The AIRE gene is responsible for appropriate clonal selection of T-cells within the thymus, and loss of this function results in proliferation of auto-sensitized immune cells. The estimated prevalence of APS-1 is about 1 in 80,000 in general population, equally affecting males and females (59,60) but is higher in certain ethnic groups, such as Iranian Jews (1/9,000) (61), Sardinians (1/14,000) (62), and Finns (1/25,000) (63,64). APS-1 is defined by the presence of at least two of the three major components of the disease: chronic mucocutaneous candidiasis, acquired hypoparathyroidism, and autoimmune Addison’s disease. APS-1 typically presents first with chronic mucocutaneous candidiasis by 5 years of age—with 50% presenting before 2 years of age—hypoparathyroidism by 8 to 10 years of age, and Addison’s disease by 12 years of age (65). However, the clinical presentation of APS-1 is highly variable even within the same family and this phenomenon may result in delayed diagnosis (63). While 80–90% of European patients present with one or more major components of APS-1 at the onset of disease, the majority of American APS-1 patients initially show organ-specific nonendocrine symptoms (urticarial eruption, hepatitis, gastritis) early in life and significant delay in the development of classical components of APS-1 (66). In addition, almost all patients have anti-interferon-alpha and anti-interferon omega autoantibodies before the appearance of clinical symptoms of APS-1. These observations have led to changes in criteria for diagnosis of APS-1 (67):

Figure 1 Genetic etiologies of PAI and distinguishing signs and symptoms by age of manifestation. PAI, primary adrenal insufficiency; IUGR, intrauterine growth retardation; HSM, hepatosplenomegaly; T2DM, diabetes mellitus; OH, hydroxylase; SERKAL, SEx Reversion, Kidneys, Adrenal and Lung dysgenesis; HSD, hydroxysteroid dehydrogenase; CAH, congenital adrenal hyperplasia; STAR, steroidogenic acute regulatory protein; MIRAGE, major findings of myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, and enteropathy; IMAGe, intrauterine growth restriction, metaphyseal dysplasia, adrenal hypoplasia congenita, genital anomalies; FGD, familial glucocorticoid deficiency; SGPL1, sphingosine-1-phosphate lyase; SF1, steroidogenic factor 1; AIRE, autoimmune regulator; CAH, congenital adrenal hyperplasia; ALD, adrenoleukodystrophy; APS1, autoimmune polyglandular syndrome type 1.
One of the three following criteria is required for a definitive diagnosis:
  • Presence of at least two of the three major components: chronic mucocutaneous candidiasis, hypoparathyroidism, autoimmune adrenal insufficiency;
  • One of the major components if a sibling has a confirmed diagnosis of APS-1;
  • APS-1 mutations in both AIRE genes.
Probable diagnostic criteria
  • Presence of one of the three major components: chronic mucocutaneous candidiasis, hypoparathyroidism, autoimmune adrenal insufficiency (before 30 years of age) and at least one of the minor components (chronic diarrhea, keratitis, periodic rash with fever, severe constipation, autoimmune hepatitis, vitiligo, alopecia, enamel hypoplasia);
  • Any component and presence of anti-interferon antibodies;
  • Any component and antibodies against aromatic L-amino acid decarboxylase, tryptophan hydroxylase, tyrosine hydroxylase or NACHT leucine-rich repeat protein.

Approximately 50% of autoimmune PAI is associated with APS-2, also known as Schmidt’s syndrome (68). APS-2 is defined by autoimmune PAI in conjunction with autoimmune thyroid disease and/or type 1 diabetes mellitus (T1DM) (68,69). Other autoimmune conditions such as vitiligo, chronic atrophic gastritis with or without pernicious anemia, chronic autoimmune hepatitis, alopecia, and myasthenia gravis may also present at the time of diagnosis or develop with time in patients with APS-2. The prevalence of disease is estimated as one in 20,000 in the general population (70) with the peak incidence at the 3rd and the 4th decade of life. It is three times more common in females than males (71).

APS-2 is a polygenic disease and does not have a simple inheritance pattern (72). The genetics of APS-2 are controlled by the major histocompatibility (MHC) complex human leukocyte antigen (HLA) haplotypes including DR3-DQ2, DR4-DQ8, DRB1-0301, and DRB1-0404 which confer disease risk to multiple autoimmune disorders. Moreover, genes within the MHC region and some of genes responsible for T-cell signaling including CTLA4, the PTPN22 have been linked to APS-2 and isolated autoimmune PAI (29).

Other causes of adrenal gland destruction

Metabolic lipid disorders [X-linked adrenoleukodystrophy (ALD), Wolman syndrome, Zellweger syndrome], sphingolipidosis, multiple infection agents (mycobacterial, bacterial, viral, and fungal), amyloidosis, sarcoidosis, adrenal metastases (e.g., lung, breast, colon, lymphomas), or hemorrhage can cause adrenal gland destruction and lead to PAI (28).

X-linked ALD

X-linked ALD is characterized by defective peroxisomal beta oxidation of saturated unbranched very long-chain fatty acids (VLCFA) that results accumulation of saturated VLCFA (≥22 carbons) in tissues and body fluids, particularly, in adrenal cortex and central nervous system causing PAI and progressive cerebral inflammatory demyelination, respectively (73). Additionally, the accumulation of VLCFA in Leydig cells causes testicular dysfunction (74), and in hair cells results in thin and sparse hair (75). ALD is a progressive neurodegenerative disease. The responsible gene for ALD is the ABCD1 [ATP-binding cassette (ABC) transporter subfamily D member 1] gene that encodes the peroxisomal ABC half-transporter ABCD1 (formerly adrenoleukodystrophy protein, ALDP), and is mapped to chromosome Xq28 (76). The estimated incidence of ALD is 1:16,800 (77). Although VLCFA levels in plasma are already elevated at birth, there is a wide variation of clinical outcomes from the rapidly progressive and if untreated fatal cerebral demyelination, cerebral ALD, in boys between 5–12 years, to milder forms of adrenomyeloneuropathy in boys and adult men, or isolated adrenal insufficiency (78). In isolated adrenal insufficiency, neurological symptoms of ALD may present years later. Therefore, in any boy with PAI, but no adrenal autoantibodies, VLCFA measurement should be included in the assessment (32).

Adrenal insufficiency affects >80% of ALD patients. The exact mechanism of adrenal insufficiency is poorly understood. It is thought that the chronic accumulation of cholesterol with saturated VLCFA in the adrenal cortex leads to cell dysfunction and cell death (79).

Neonatal ALD is a variant of Zellweger syndrome spectrum, characterized by hypotonia, leukodystrophy, and vision and sensorineural hearing deficiencies. Adrenal insufficiency and renal calcifications can present later in childhood (80).

Women who are heterozygous carriers of ALD may develop signs of myelopathy as they get older (from 18% before 40 years of age to 80% by 60 years of age). They may also develop adrenal insufficiency (<1%) (81).

Wolman disease

Wolman disease is an autosomal recessive disorder due to mutations in the LIPA gene at locus 10q23.31 on the chromosome 10 (18). The LIPA gene codes for lysosomal acid lipase (LAL), which catalyzes the conversion of cholesterol esters and triglycerides to cholesterol, glycerols, and free fatty acids (82). LAL deficiency results in intracellular massive accumulation of cholesterol esters and triglycerides, particularly in liver, spleen, gastrointestinal tract, and adrenal glands. It is very rare with the incidence of less than 1 in 100,000 live births (83). Affected infants present with hepatosplenomegaly, failure to thrive, steatorrhea, and vomiting within the first months of life. Enlarged adrenal glands with calcifications is evident on plain X-rays by 6 months of age (82). If left untreated, Wolman disease is fatal within the first year of life. Cholesterol ester storage disease (CESD) is the moderate form of LAL deficiency, presenting at various times throughout the lifespan with hepatomegaly, hyperlipoproteinemia, and accelerated atherosclerosis. The age of presentation varies with the amount of functional enzyme produced, with the most common presenting symptom being hepatomegaly (84). Adrenal gland calcification is rare in CESD form (85).

Peroxisome biogenesis disorders

Peroxisome biogenesis disorders are a group of autosomal recessive disorders in the Zellweger spectrum (PBD-ZSD) and characterized by impaired peroxisome function due to mutations in any of PEX genes (86). The Zellweger spectrum includes Zellweger syndrome (severe form), neonatal ALD (intermediate form), and infantile Refsum disease (mild form). There is a significant overlap of signs and symptoms. Affected infants present with craniofacial anomalies, failure to thrive, hypotonia, seizures, cataracts, and adrenal insufficiency (87).

Sphingolipidoses

Sphingolipidoses are a broad category of disease that result from a deficiency of any enzyme required for the proper degradation or storage of lipids. Sphingosine-1-phosphate lyase (SGPL1) deficiency is a sphingolipidosis that affects the adrenal glands resulting in PAI within 12 months of birth. Other manifestations include nephrotic syndrome, failure of sexual development, ichthyosis, neurological dysfunction, immunodeficiency, and hypothyroidism (36).

Infectious agents

Infectious agents may cause adrenal insufficiency by either colonizing and destroying the adrenal gland directly or by secreting endotoxins that secondarily damage the adrenal gland. One example is the secretion of endotoxins by N. meningitidis, which may cause hemorrhage within the adrenal glands and shock, referred to as Waterhouse-Friderichsen syndrome (88).

Bilateral adrenal hemorrhage

Bilateral adrenal hemorrhage owing to trauma, especially in a newborn after prolonged labor or traumatic delivery, sepsis, antiphospholipid syndrome, and heparin induced thrombocytopenia may cause the loss of adrenal function (28).

Steroidogenic enzyme defects

Congenital adrenal hyperplasia

CAH is a group of autosomal recessive disorders of steroidogenesis characterized by impaired cortisol synthesis with a compensatory increase in ACTH leading to adrenal hyperplasia. The most common cause of CAH is deficiency of 21-hydroxylase (95% of cases) (89) followed by 11-beta-hydroxylase, and 3-beta-hydroxysteroid dehydrogenase enzymes (90). The less often enzyme deficiencies are 17-alpha-hydroxylase/17,20-lyase and cholesterol desmolase. Each enzyme is encoded by a specific gene and mutations causing CAH have been identified. Depending on the severity of enzyme deficiency, CAH can be divided into classical and non-classical (milder and symptoms start later in life) forms. Classical CAH due to 21 hydroxylase deficiency is further divided into salt-wasting and simple virilizing forms (91). In both 21-hydroxylase and 11-beta hydroxylase deficiencies, excess androgen production causes ambiguous genitalia in newborn females and progressive postnatal virilization in both sexes. In 3-beta-hydroxysteroid dehydrogenase deficiency, mineralocorticoid, glucocorticoid, and sex steroids production is decreased resulting in undervirilized males and some virilization in females. CAH due to 17-alpha-hydroxylase/17,20-lyase is characterized by decreased production of adrenal and gonadal steroids; females have no adrenarche and lack of pubertal development, males have ambiguous genitalia (92).

Congenital lipoid adrenal hyperplasia—deficiency of steroidogenic acute regulatory protein (StAR) is a rare, severe form of CAH and most common in Japan and Korea (89). The STAR gene is affected in this disorder preventing the initial conversion of cholesterol into pregnenolone, entirely disabling the adrenal steroid synthesis pathway. Due to loss of androgen production, both 46,XY, and 46,XX infants will present with a female phenotype; corticosteroid production loss results in severe salt-wasting presenting in the first month of life (93). A non-classic form of lipoid CAH exists, corresponding to only partial deficiency of STAR protein activity which presents at a median of 4 years of age. In addition to virilization, non-classic lipoid CAH will manifest with electrolyte imbalances—due to decreased mineralocorticoid production—and PAI (94).

More detailed information on CAH can be found in numerous review articles that have already been published (95,96).

Mitochondrial DNA deletion syndromes are characterized by mitochondrial respiratory chain deficiency that result in a spectrum of overlapping phenotypes, including Kearns-Sayre syndrome (KSS), Pearson syndrome, and progressive external ophthalmoplegia (PEO) (97). Although neuromuscular symptoms are prominent, endocrine dysfunction, particularly diabetes, is common. Other endocrine dysfunctions include short stature, hypothyroidism, hypogonadism, and adrenal insufficiency (98).

Smith-Lemli-Opitz syndrome

Smith-Lemli-Opitz syndrome is a rare disorder due to a deficiency of the 7-dehydrocholesterol reductase enzyme (DHCR7), which converts 7-dehydrocholesterol into cholesterol. This leads to an accumulation of 7-dehydrocholesterol and a deficiency of all adrenal steroids (99). All patients will present with intellectual disability, as well as electrolyte abnormalities, adrenal insufficiency, hypoparathyroidism, hypothyroidism, and various manifestations of immunodeficiency (100).


Diagnosis and treatment

Clinical presentation of PAI is the result of glucocorticoid and mineralocorticoid deficiency. Signs and symptoms include weakness, fatigue, musculoskeletal pain, weight loss, hypotension, abdominal pain, and hyperpigmentation of the skin and mucous membranes (Table 3). As melanocyte-stimulating hormone (MSH) is made simultaneously with ACTH, overproduction of ACTH by the pituitary in response to PAI also increases circulating levels of MSH (101). The characteristic hyperpigmentation is a result of the increased stimulation of the melanocortin 1 receptor (encoded by MC1R) by MSH (102,103). In children, this can be most noticeable in areas that typically do not get significant sun exposure, such as the axilla, palms, and groin. Patients may present in acute adrenal crisis if their condition is not previously known or poorly managed.

Table 3

Signs and symptoms of adrenal insufficiency and crisis

Adrenal insufficiency
   Signs
    Hyperpigmentation of sun exposed areas, skin creases, mucous membranes, groin, axillae, scars
    Hypotension with worsening postural hypotension
    Dehydration
    Failure to thrive
    Hypoglycemia
    Apnea (newborn)
   Symptoms
    General malaise
    Fatigue
    Headache
    Weakness
    Dizziness with standing
    Weight loss
    Anorexia
    Abdominal pain, nausea, vomiting, diarrhea
    Salt craving
    Icterus (newborn)
Adrenal crisis
   Signs
    Hypotension
    Tachycardia
    Reduced or altered consciousness
    Dehydration (hypovolemia)
    Apnea (newborn)
   Symptoms
    Severe weakness
    Altered behavior, confusion or extreme fatigue
    Abdominal pain, vomiting
    History of febrile illness or stress

Diagnosis of PAI is based on low morning serum cortisol concentrations and confirmed by low stimulated cortisol during a corticotropin stimulation test. PAI is highly likely if serum cortisol is <140 nmol/L (5 µg/dL) and ACTH concentration that is elevated greater than 2 times above the upper reference limit for the specific assay (104). The gold standard for PAI diagnosis is the corticotropin stimulation test (cosyntropin or ACTH test) and should be performed to unless basal results are absolute. Commonly used, the short corticotropin test measures cortisol levels before and 30 and/or 60 minutes after IV or IM administration of cosyntropin. A dose of 250 µg is utilized in children ≥2 years of age; in children less than 2 years of age a dose of 125 µg is used and in infants a weight-adjusted dose of 15 µg/kg is recommended (104). While the cutoff used to make the diagnosis is assay-specific, a peak stimulated cortisol level exceeding 500 nmol/L (18 µg/dL) is widely accepted as evidence of sufficient adrenocortical response.

Children with PAI require glucocorticoid and mineralocorticoid replacement. Hydrocortisone is preferred in children with PA to minimize linear growth retardation and pubertal delay often common in more potent, longer acting glucocorticoids. Dosing of corticosteroids is based on physiological basal secretion of cortisol at 6–9 mg/m2/day (105,106). Recommended starting dose is 8 mg/m2/day in two to four divided doses with maintenance dosing typically between 6–12 mg/m2/day (104,107). Improved adherence and ease of administration comes with fewer daily doses. First morning dose should be higher than subsequent doses with last dose administered 4–6 hours before bedtime to mimic physiological cortisol secretion (108). Longer acting glucocorticoids such as prednisone or prednisolone at 5.0–7.5 or 0.25–0.5 mg/day in divided doses, respectively, are alternative treatment options in older adolescent patients. Mineralocorticoid replacement is accomplished by giving fludrocortisone at 50 to 200 µg/day with typical dose at 100 µg/day. Infants who require mineralocorticoid replacement will need sodium chloride supplementation due to insufficient sodium intake through breast milk or formula at a dose of 17–34 mEq/day in several divided doses with feedings (Table 4).

Table 4

Treatment of primary adrenal insufficiency in children and adolescents (73,75,77)

Medications/condition Dose/comments
Maintenance dosing
   Hydrocortisone 8 mg/m2/day in 3 divided doses
   Fludrocortisone 100 μg daily
   Sodium chloride (for infants less than 6 months of age) 1–2 g/day in divided doses given in feedings
Hydrocortisone stress dosing
   Febrile illness
    Temperature >38 °C Double maintenance dose
    Temperature >39 °C Triple maintenance dose
    If unable to tolerate oral intake Intramuscular or subcutaneous hydrocortisone to be given:
  Infants: 25 mg
  School age children: 50 mg
  Adolescents: 100 mg
   Surgery
    Minor surgery 25–75 mg/day in divided doses for 1–2 days, then returning to maintenance dosing
    Major surgery or trauma 50 mg/m2 IV bolus, followed by 50–100 mg/m2/day in divided doses given every 6 hours. Appropriate IV fluids with dextrose and NaCl should also be given
Return to oral dosing as clinically able
Adrenal crisis
   Hydrocortisone 50–100 mg/m2 bolus; followed by 50–100 mg/m2/day in divided doses given every 6 hours
   0.9% normal saline (for shock/hypovolemia) 20 mL/kg bolus, repeated to max 60 mL/kg in 1 hour
   10% Dextrose (for hypoglycemia) 5–10 mL/kg administered slowly at a rate of 2–3 mL/min

IV, intravenous.

Patients should be closely monitored for signs of inadequate or excessive glucocorticoid and mineralocorticoid replacement. Decreased growth velocity, excessive weight gain and other signs and symptoms of Cushing syndrome may indicate excessive glucocorticoid treatment. Likewise, poor weight gain, fatigue and hyperpigmentation may suggest need for increased glucocorticoid treatment. Inadequate mineralocorticoid replacement may present with salt craving, poor weight gain, dehydration, and hyponatremia with hyperkalemia and elevated renin. Hypertension, edema, and low renin concentration may suggest excessive mineralocorticoid replacement. Infants and young children should be monitored more frequently, monitoring growth and blood pressure. The latest Endocrine Society Clinical Practice Guidelines recommend to use clinical assessment including growth velocity, body weight, blood pressure, and energy levels to adjust glucocorticoid and mineralocorticoid dosing in children (103).

Stress dosing and management of adrenal crisis

Patients with PAI require increased corticosteroid dosing during illness and stress. Recommended hydrocortisone dose is double or triple the daily maintenance for 24–48 hours or until afebrile for 24 hours before returning to maintenance. Children with low grade fever (<38 °C) may not need stress dosing. Patients who are vomiting or have diarrhea, who may be unable to take medication orally, will require intramuscular hydrocortisone (Table 4).

Adrenal crisis may occur in severe illness, physical or physiological stress, inadequate treatment, or abrupt withdrawal of corticosteroids. Patients with suspected crisis should immediately be treated with Solu-Cortef (hydrocortisone sodium succinate) 100 mg/m2 intravenous or intramuscular. High dose hydrocortisone should continue at 100 mg/m2/day intravenously divided every 6 hours. Once patient is able to tolerate oral intake, hydrocortisone should be given at 25–30 mg/m2/day or triple maintenance dose in three divided doses. Once stress is over, hydrocortisone should be gradually weaned back to daily maintenance dose.


Conclusions

PAI has a number of potential underlying etiologies and can affect a wide spectrum of patients, from pediatrics to geriatrics. Since its first description by Addison, great advancements have been made in the understanding of adrenal gland development and function. While PAI remains a relatively rare condition, it has serious and precipitous implications when it arises. Having a high index of suspicion is the key for early diagnosis of PAI and the prevention of life-threatening adrenal crisis. Corticosteroid and mineralocorticoid replacement is necessary to prevent morbidity and life-threatening adrenal crisis. Adrenal crisis is an endocrine emergency that requires high doses of corticosteroids along with intensive medical monitoring.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Pediatric Medicine for the series “Clinical Pearls in Pediatric Endocrinology and Metabolism”. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://pm.amegroups.com/article/view/10.21037/pm-22-2/rc

Peer Review File: Available at https://pm.amegroups.com/article/view/10.21037/pm-22-2/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://pm.amegroups.com/article/view/10.21037/pm-22-2/coif). The series “Clinical Pearls in Pediatric Endocrinology and Metabolism” was commissioned by the editorial office without any funding or sponsorship. B.E.L. served as the unpaid Guest Editor of the series and serves an unpaid editorial board member of Pediatric Medicine from October 2020 to September 2024. P.V.M. served as the unpaid Guest Editor of the series. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.

Special series on Clinical Pearls in Pediatric Endocrinology and Metabolism.


References

  1. Addison T. On the constitutional and local effects of disease of the supra-renal capsules. London, UK: Samuel Highley, 1855.
  2. Thorn GW, Forsham PH, Frawley TF, et al. Clinical experience with the use of cortisone acetate pellets in the treatment of adrenal cortical insufficiency. J Clin Endocrinol Metab 1950;10:833-4.
  3. Willis AC, Vince FP. The prevalence of Addison's disease in Coventry, UK. Postgrad Med J 1997;73:286-8. [Crossref] [PubMed]
  4. Laureti S, Vecchi L, Santeusanio F, et al. Is the prevalence of Addison's disease underestimated? J Clin Endocrinol Metab 1999;84:1762. [Crossref] [PubMed]
  5. Løvås K, Husebye ES. High prevalence and increasing incidence of Addison's disease in western Norway. Clin Endocrinol (Oxf) 2002;56:787-91. [Crossref] [PubMed]
  6. Chabre O, Goichot B, Zenaty D, et al. Group 1. Epidemiology of primary and secondary adrenal insufficiency: Prevalence and incidence, acute adrenal insufficiency, long-term morbidity and mortality. Ann Endocrinol (Paris) 2017;78:490-4. [Crossref] [PubMed]
  7. Burdea L, Mendez MD. 21 Hydroxylase Deficiency. In: StatPearls. Treasure Island (FL): StatPearls Publishing; March 15, 2022.
  8. Walker AP, Chelly J, Love DR, et al. A YAC contig in Xp21 containing the adrenal hypoplasia congenita and glycerol kinase deficiency genes. Hum Mol Genet 1992;1:579-85. [Crossref] [PubMed]
  9. Taketo M, Parker KL, Howard TA, et al. Homologs of Drosophila Fushi-Tarazu factor 1 map to mouse chromosome 2 and human chromosome 9q33. Genomics 1995;25:565-7. [Crossref] [PubMed]
  10. Cone RD, Mountjoy KG, Robbins LS, et al. Cloning and functional characterization of a family of receptors for the melanotropic peptides. Ann N Y Acad Sci 1993;680:342-63. [Crossref] [PubMed]
  11. Gardiner K, Slavov D, Bechtel L, et al. Annotation of human chromosome 21 for relevance to Down syndrome: gene structure and expression analysis. Genomics 2002;79:833-43. [Crossref] [PubMed]
  12. Tullio-Pelet A, Salomon R, Hadj-Rabia S, et al. Mutant WD-repeat protein in triple-A syndrome. Nat Genet 2000;26:332-5. [Crossref] [PubMed]
  13. Huguet EL, McMahon JA, McMahon AP, et al. Differential expression of human Wnt genes 2, 3, 4, and 7B in human breast cell lines and normal and disease states of human breast tissue. Cancer Res 1994;54:2615-21.
  14. Matsuoka S, Edwards MC, Bai C, et al. p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev 1995;9:650-62. [Crossref] [PubMed]
  15. Szpirer J, Pedeutour F, Kesti T, et al. Localization of the gene for DNA polymerase epsilon (POLE) to human chromosome 12q24.3 and rat chromosome 12 by somatic cell hybrid panels and fluorescence in situ hybridization. Genomics 1994;20:223-6. [Crossref] [PubMed]
  16. Narumi S, Amano N, Ishii T, et al. SAMD9 mutations cause a novel multisystem disorder, MIRAGE syndrome, and are associated with loss of chromosome 7. Nat Genet 2016;48:792-7. [Crossref] [PubMed]
  17. Migeon BR, Moser HW, Moser AB, et al. Adrenoleukodystrophy: evidence for X linkage, inactivation, and selection favoring the mutant allele in heterozygous cells. Proc Natl Acad Sci U S A 1981;78:5066-70. [Crossref] [PubMed]
  18. Anderson RA, Rao N, Byrum RS, et al. In situ localization of the genetic locus encoding the lysosomal acid lipase/cholesteryl esterase (LIPA) deficient in Wolman disease to chromosome 10q23.2-q23.3. Genomics 1993;15:245-7. [Crossref] [PubMed]
  19. Buonocore F, Achermann JC. Primary adrenal insufficiency: New genetic causes and their long-term consequences. Clin Endocrinol (Oxf) 2020;92:11-20. [Crossref] [PubMed]
  20. Zhou J, Saba JD. Identification of the first mammalian sphingosine phosphate lyase gene and its functional expression in yeast. Biochem Biophys Res Commun 1998;242:502-7. [Crossref] [PubMed]
  21. Chiou SH, Hu MC, Chung BC. A missense mutation at Ile172----Asn or Arg356----Trp causes steroid 21-hydroxylase deficiency. J Biol Chem 1990;265:3549-52.
  22. Lifton RP, Dluhy RG, Powers M, et al. Hereditary hypertension caused by chimaeric gene duplications and ectopic expression of aldosterone synthase. Nat Genet 1992;2:66-74. [Crossref] [PubMed]
  23. Persson B, Kallberg Y, Bray JE, et al. The SDR (short-chain dehydrogenase/reductase and related enzymes) nomenclature initiative. Chem Biol Interact 2009;178:94-8. [Crossref] [PubMed]
  24. Krohn K, Uibo R, Aavik E, et al. Identification by molecular cloning of an autoantigen associated with Addison's disease as steroid 17 alpha-hydroxylase. Lancet 1992;339:770-3. [Crossref] [PubMed]
  25. Chung BC, Guo IC, Chou SJ. Transcriptional regulation of the CYP11A1 and ferredoxin genes. Steroids 1997;62:37-42. [Crossref] [PubMed]
  26. Zeviani M, Moraes CT, DiMauro S, et al. Deletions of mitochondrial DNA in Kearns-Sayre syndrome. Neurology 1988;38:1339-46. [Crossref] [PubMed]
  27. Tweedie S, Braschi B, Gray K, et al. Genenames.org: the HGNC and VGNC resources in 2021. Nucleic Acids Res 2021;49:D939-46. [Crossref] [PubMed]
  28. Ten S, New M, Maclaren N. Clinical review 130: Addison's disease 2001. J Clin Endocrinol Metab 2001;86:2909-22. [Crossref] [PubMed]
  29. Mitchell AL, Pearce SH. Autoimmune Addison disease: pathophysiology and genetic complexity. Nat Rev Endocrinol 2012;8:306-16. [Crossref] [PubMed]
  30. Erichsen MM, Løvås K, Fougner KJ, et al. Normal overall mortality rate in Addison's disease, but young patients are at risk of premature death. Eur J Endocrinol 2009;160:233-7. [Crossref] [PubMed]
  31. Phelan JK, McCabe ER. Mutations in NR0B1 (DAX1) and NR5A1 (SF1) responsible for adrenal hypoplasia congenita. Hum Mutat 2001;18:472-87. [Crossref] [PubMed]
  32. Guran T, Buonocore F, Saka N, et al. Rare Causes of Primary Adrenal Insufficiency: Genetic and Clinical Characterization of a Large Nationwide Cohort. J Clin Endocrinol Metab 2016;101:284-92. [Crossref] [PubMed]
  33. Achermann JC, Silverman BL, Habiby RL, et al. Presymptomatic diagnosis of X-linked adrenal hypoplasia congenita by analysis of DAX1. J Pediatr 2000;137:878-81. [Crossref] [PubMed]
  34. Yoo HW. Diverse etiologies, diagnostic approach, and management of primary adrenal insufficiency in pediatric age. Ann Pediatr Endocrinol Metab 2021;26:149-57. [Crossref] [PubMed]
  35. Komachali SR, Nouri N, Zaker E, et al. A novel NR0B1 mutation correlated with X-linked adrenal hypoplasia congenital (AHC). Gene Reports 2022;27:101598.
  36. Buonocore F, McGlacken-Byrne SM, Del Valle I, et al. Current Insights Into Adrenal Insufficiency in the Newborn and Young Infant. Front Pediatr 2020;8:619041. [Crossref] [PubMed]
  37. Ferraz-de-Souza B, Lin L, Achermann JC. Steroidogenic factor-1 (SF-1, NR5A1) and human disease. Mol Cell Endocrinol 2011;336:198-205. [Crossref] [PubMed]
  38. Pignatti E, Flück CE. Adrenal cortex development and related disorders leading to adrenal insufficiency. Mol Cell Endocrinol 2021;527:111206. [Crossref] [PubMed]
  39. Kyritsi EM, Sertedaki A, Charmandari E, et al. Familial or Sporadic Adrenal Hypoplasia Syndromes 2000.
  40. Chan LF, Clark AJ, Metherell LA. Familial glucocorticoid deficiency: advances in the molecular understanding of ACTH action. Horm Res 2008;69:75-82. [Crossref] [PubMed]
  41. Flück CE. Mechanisms in endocrinology: Update on pathogenesis of primary adrenal insufficiency: beyond steroid enzyme deficiency and autoimmune adrenal destruction. Eur J Endocrinol 2017;177:R99-R111. [Crossref] [PubMed]
  42. Tsai SL, Green J, Metherell LA, et al. Primary Adrenocortical Insufficiency Case Series: Genetic Etiologies More Common than Expected. Horm Res Paediatr 2016;85:35-42. [Crossref] [PubMed]
  43. Novoselova TV, Chan LF, Clark AJL. Pathophysiology of melanocortin receptors and their accessory proteins. Best Pract Res Clin Endocrinol Metab 2018;32:93-106. [Crossref] [PubMed]
  44. Flück CE. Update on genetics of primary adrenal insufficiency. Curr Opin Endocr Metab Res 2019;8:96-103.
  45. Huebner A, Elias LL, Clark AJ. ACTH resistance syndromes. J Pediatr Endocrinol Metab 1999;12:277-93.
  46. Pogliaghi G, Cangiano B, Duminuco P, et al. Triple-A Syndrome (TAS): An In-Depth Overview on Genetic and Phenotype Heterogeneity. Protein Pept Lett 2020;27:1192-203. [Crossref] [PubMed]
  47. Cho AR, Yang KJ, Bae Y, et al. Tissue-specific expression and subcellular localization of ALADIN, the absence of which causes human triple A syndrome. Exp Mol Med 2009;41:381-6. [Crossref] [PubMed]
  48. Hirano M, Furiya Y, Asai H, et al. ALADINI482S causes selective failure of nuclear protein import and hypersensitivity to oxidative stress in triple A syndrome. Proc Natl Acad Sci U S A 2006;103:2298-303. [Crossref] [PubMed]
  49. Aghajanzadeh M, Safarpoor F, Hydayati MH, et al. Allgrove syndrome: reports of cases and literature review. Saudi J Gastroenterol 2006;12:34-5. [Crossref] [PubMed]
  50. Chang Z, Lu W, Zhao Z, et al. Genetic aetiology of primary adrenal insufficiency in Chinese children. BMC Med Genomics 2021;14:172. [Crossref] [PubMed]
  51. Vilain E, Le Merrer M, Lecointre C, et al. IMAGe, a new clinical association of intrauterine growth retardation, metaphyseal dysplasia, adrenal hypoplasia congenita, and genital anomalies. J Clin Endocrinol Metab 1999;84:4335-40. [Crossref] [PubMed]
  52. Schrier Vergano SA, Deardorff MA. IMAGe Syndrome. In: Adam MP, Everman DB, Mirzaa GM, et al., eds. GeneReviews®. Seattle (WA): University of Washington, Seattle; March 13, 2014.
  53. Logan CV, Murray JE, Parry DA, et al. DNA Polymerase Epsilon Deficiency Causes IMAGe Syndrome with Variable Immunodeficiency. Am J Hum Genet 2018;103:1038-44. [Crossref] [PubMed]
  54. Onuma S, Wada T, Araki R, et al. MIRAGE syndrome caused by a novel missense variant (p.Ala1479Ser) in the SAMD9 gene. Hum Genome Var 2020;7:4. [Crossref] [PubMed]
  55. Ishiwa S, Kamei K, Tanase-Nakao K, et al. A girl with MIRAGE syndrome who developed steroid-resistant nephrotic syndrome: a case report. BMC Nephrol 2020;21:340. [Crossref] [PubMed]
  56. Buonocore F, Kühnen P, Suntharalingham JP, et al. Somatic mutations and progressive monosomy modify SAMD9-related phenotypes in humans. J Clin Invest 2017;127:1700-13. [Crossref] [PubMed]
  57. Bancos I, Hahner S, Tomlinson J, et al. Diagnosis and management of adrenal insufficiency. Lancet Diabetes Endocrinol 2015;3:216-26. [Crossref] [PubMed]
  58. Betterle C, Dal Pra C, Mantero F, et al. Autoimmune adrenal insufficiency and autoimmune polyendocrine syndromes: autoantibodies, autoantigens, and their applicability in diagnosis and disease prediction. Endocr Rev 2002;23:327-64. [Crossref] [PubMed]
  59. Ergun-Longmire B, Maclaren NK. Chapter 147 - Autoimmune endocrine disorders. In: Jameson J, De Groot L, de Kretser D, et al. editors. Endocrinology: Adult and Pediatric, vol. 2–2. 7th edition. Philadelphia: W.B. Saunders, 2016:2549-2565.e5.
  60. Husebye ES, Anderson MS, Kämpe O. Autoimmune Polyendocrine Syndromes. N Engl J Med 2018;378:1132-41. [Crossref] [PubMed]
  61. Zlotogora J, Shapiro MS. Polyglandular autoimmune syndrome type I among Iranian Jews. J Med Genet 1992;29:824-6. [Crossref] [PubMed]
  62. Rosatelli MC, Meloni A, Meloni A, et al. A common mutation in Sardinian autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients. Hum Genet 1998;103:428-34. [Crossref] [PubMed]
  63. Ahonen P, Myllärniemi S, Sipilä I, et al. Clinical variation of autoimmune polyendocrinopathy–candidiasis–ectodermal dystrophy (APECED) in a series of 68 patients. N Engl J Med 1990;322:1829-36. [Crossref] [PubMed]
  64. Perheentupa J. Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J Clin Endocrinol Metab 2006;91:2843-50. [Crossref] [PubMed]
  65. Faiyaz-Ul-Haque M, Bin-Abbas B, Al-Abdullatif A, et al. Novel and recurrent mutations in the AIRE gene of autoimmune polyendocrinopathy syndrome type 1 (APS1) patients. Clin Genet 2009;76:431-40. [Crossref] [PubMed]
  66. Ferre EM, Rose SR, Rosenzweig SD, et al. Redefined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI Insight 2016;1:e88782. [Crossref] [PubMed]
  67. Husebye ES, Perheentupa J, Rautemaa R, et al. Clinical manifestations and management of patients with autoimmune polyendocrine syndrome type I. J Intern Med 2009;265:514-29. [Crossref] [PubMed]
  68. Betterle C, Volpato M, Greggio AN, et al. Type 2 polyglandular autoimmune disease (Schmidt’s syndrome). J Pediatr Endocrinol Metab 1996;9:113-23. [Crossref] [PubMed]
  69. Neufeld M, Maclaren NK, Blizzard RM. Two types of autoimmune Addison's disease associated with different polyglandular autoimmune (PGA) syndromes. Medicine (Baltimore) 1981;60:355-62. [Crossref] [PubMed]
  70. Cutolo M. Autoimmune polyendocrine syndromes. Autoimmun Rev 2014;13:85-9. [Crossref] [PubMed]
  71. Kahaly GJ, Frommer L. Polyglandular autoimmune syndromes. J Endocrinol Invest 2018;41:91-8. [Crossref] [PubMed]
  72. Michels AW, Eisenbarth GS. Autoimmune polyendocrine syndrome type 1 (APS-1) as a model for understanding autoimmune polyendocrine syndrome type 2 (APS-2). J Intern Med 2009;265:530-40. [Crossref] [PubMed]
  73. Berger J, Forss-Petter S, Eichler FS. Pathophysiology of X-linked adrenoleukodystrophy. Biochimie 2014;98:135-42. [Crossref] [PubMed]
  74. Brennemann W, Köhler W, Zierz S, et al. Testicular dysfunction in adrenomyeloneuropathy. Eur J Endocrinol 1997;137:34-9. [Crossref] [PubMed]
  75. Höftberger R, Kunze M, Weinhofer I, et al. Distribution and cellular localization of adrenoleukodystrophy protein in human tissues: implications for X-linked adrenoleukodystrophy. Neurobiol Dis 2007;28:165-74. [Crossref] [PubMed]
  76. Mosser J, Douar AM, Sarde CO, et al. Putative X-linked adrenoleukodystrophy gene shares unexpected homology with ABC transporters. Nature 1993;361:726-30. [Crossref] [PubMed]
  77. Bezman L, Moser AB, Raymond GV, et al. Adrenoleukodystrophy: incidence, new mutation rate, and results of extended family screening. Ann Neurol 2001;49:512-7.
  78. Kemp S, Huffnagel IC, Linthorst GE, et al. Adrenoleukodystrophy - neuroendocrine pathogenesis and redefinition of natural history. Nat Rev Endocrinol 2016;12:606-15. [Crossref] [PubMed]
  79. Powers JM, Schaumburg HH, Johnson AB, et al. A correlative study of the adrenal cortex in adreno-leukodystrophy--evidence for a fatal intoxication with very long chain saturated fatty acids. Invest Cell Pathol 1980;3:353-76.
  80. Steinberg SJ, Raymond GV, Braverman NE, et al. Zellweger Spectrum Disorder. In: Adam MP, Everman DB, Mirzaa GM, et al. editors. GeneReviews®. Seattle (WA): University of Washington, Seattle; December 12, 2003.
  81. Engelen M, Barbier M, Dijkstra IM, et al. X-linked adrenoleukodystrophy in women: a cross-sectional cohort study. Brain 2014;137:693-706. [Crossref] [PubMed]
  82. Potter JE, Petts G, Ghosh A, et al. Enzyme replacement therapy and hematopoietic stem cell transplant: a new paradigm of treatment in Wolman disease. Orphanet J Rare Dis 2021;16:235. [Crossref] [PubMed]
  83. Aguisanda F, Thorne N, Zheng W. Targeting Wolman Disease and Cholesteryl Ester Storage Disease: Disease Pathogenesis and Therapeutic Development. Curr Chem Genom Transl Med 2017;11:1-18. [Crossref] [PubMed]
  84. Bernstein DL, Hülkova H, Bialer MG, et al. Cholesteryl ester storage disease: review of the findings in 135 reported patients with an underdiagnosed disease. J Hepatol 2013;58:1230-43. [Crossref] [PubMed]
  85. Edelstein RA, Filling-Katz MR, Pentchev P, et al. Cholesteryl ester storage disease: a patient with massive splenomegaly and splenic abscess. Am J Gastroenterol 1988;83:687-92.
  86. Klouwer FC, Berendse K, Ferdinandusse S, et al. Zellweger spectrum disorders: clinical overview and management approach. Orphanet J Rare Dis 2015;10:151. [Crossref] [PubMed]
  87. Braverman NE, Raymond GV, Rizzo WB, et al. Peroxisome biogenesis disorders in the Zellweger spectrum: An overview of current diagnosis, clinical manifestations, and treatment guidelines. Mol Genet Metab 2016;117:313-21. [Crossref] [PubMed]
  88. Paolo WF Jr, Nosanchuk JD. Adrenal infections. Int J Infect Dis 2006;10:343-53. [Crossref] [PubMed]
  89. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol 2018;179:R125-41. [Crossref] [PubMed]
  90. New MI. Inborn errors of adrenal steroidogenesis. Mol Cell Endocrinol 2003;211:75-83. [Crossref] [PubMed]
  91. Shetty VB, Bower C, Jones TW, et al. Ethnic and gender differences in rates of congenital adrenal hyperplasia in Western Australia over a 21 year period. J Paediatr Child Health 2012;48:1029-32. [Crossref] [PubMed]
  92. Ergun-Longmire B, Auchus R, Papari-Zareei M, et al. Two novel mutations found in a patient with 17alpha-hydroxylase enzyme deficiency. J Clin Endocrinol Metab 2006;91:4179-82. [Crossref] [PubMed]
  93. Chen H, Zhang Q, Chen R, et al. Lipoid congenital adrenal hyperplasia due to steroid acute regulatory protein (STAR) variants in Three Chinese patients. J Steroid Biochem Mol Biol 2020;200:105635. [Crossref] [PubMed]
  94. Ishii T, Tajima T, Kashimada K, et al. Clinical Features of 57 Patients with Lipoid Congenital Adrenal Hyperplasia: Criteria for Nonclassic Form Revisited. J Clin Endocrinol Metab 2020;105:dgaa557. [Crossref] [PubMed]
  95. New M, Yau M, Lekarev O, et al. Congenital Adrenal Hyperplasia 2000.
  96. Merke DP, Auchus RJ. Congenital Adrenal Hyperplasia Due to 21-Hydroxylase Deficiency. N Engl J Med 2020;383:1248-61. [Crossref] [PubMed]
  97. Goldstein A, Falk MJ, Adam MP, et al. Mitochondrial DNA Deletion Syndromes 1993.
  98. Schaefer AM, Walker M, Turnbull DM, et al. Endocrine disorders in mitochondrial disease. Mol Cell Endocrinol 2013;379:2-11. [Crossref] [PubMed]
  99. Abdel-Khalik J, Hearn T, Dickson AL, et al. Bile acid biosynthesis in Smith-Lemli-Opitz syndrome bypassing cholesterol: Potential importance of pathway intermediates. J Steroid Biochem Mol Biol 2021;206:105794. [Crossref] [PubMed]
  100. Donoghue SE, Pitt JJ, Boneh A, et al. Smith-Lemli-Opitz syndrome: clinical and biochemical correlates. J Pediatr Endocrinol Metab 2018;31:451-9. [Crossref] [PubMed]
  101. Solomon S. POMC-derived peptides and their biological action. Ann N Y Acad Sci 1999;885:22-40. [Crossref] [PubMed]
  102. Shulman DI, Palmert MR, Kemp SF, et al. Adrenal insufficiency: still a cause of morbidity and death in childhood. Pediatrics 2007;119:e484-94. [Crossref] [PubMed]
  103. Robbins LS, Nadeau JH, Johnson KR, et al. Pigmentation phenotypes of variant extension locus alleles result from point mutations that alter MSH receptor function. Cell 1993;72:827-34. [Crossref] [PubMed]
  104. Bornstein SR, Allolio B, Arlt W, et al. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2016;101:364-89. [Crossref] [PubMed]
  105. White P, Sarafoglou K. Adrenal insufficiency. In: Sarafoglou K, Hoffmann GF, Roth KS. editors. Pediatric Endocrinology and Inborn Errors of Metabolism. 2nd edition. New York: McGraw-Hill Education, 2017:513-31.
  106. Bowden SA, Henry R. Pediatric Adrenal Insufficiency: Diagnosis, Management, and New Therapies. Int J Pediatr 2018;2018:1739831. [Crossref] [PubMed]
  107. Ramirez Alcantara J, Halper A. Adrenal insufficiency updates in children. Curr Opin Endocrinol Diabetes Obes 2021;28:75-81. [Crossref] [PubMed]
  108. Kirkgoz T, Guran T. Primary adrenal insufficiency in children: Diagnosis and management. Best Pract Res Clin Endocrinol Metab 2018;32:397-424. [Crossref] [PubMed]
doi: 10.21037/pm-22-2
Cite this article as: Ergun-Longmire B, Rowland D, Dewey J, Vining-Maravolo P. A narrative review: an update on primary adrenal insufficiency (PAI) in pediatric population. Pediatr Med 2023;6:36.

Download Citation